1
|
Zaki MK, Abed MN, Alassaf FA. Antidiabetic Agents and Bone Quality: A Focus on Glycation End Products and Incretin Pathway Modulations. J Bone Metab 2024; 31:169-181. [PMID: 39307518 PMCID: PMC11416877 DOI: 10.11005/jbm.2024.31.3.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/01/2024] [Accepted: 05/18/2024] [Indexed: 09/26/2024] Open
Abstract
Diabetes mellitus is associated with inadequate bone health and quality and heightened susceptibility to fractures, even in patients with normal or elevated bone mineral density. Elevated advanced glycation end-products (AGEs) and a suppressed incretin pathway are among the mechanisms through which diabetes affects the bone. Accordingly, the present review aimed to investigate the effects of antidiabetic medications on bone quality, primarily through AGEs and the incretin pathway. Google Scholar, Cochrane Library, and PubMed were used to examine related studies until February 2024. Antidiabetic medications influence AGEs and the incretin pathway directly or indirectly. Certain antidiabetic drugs including metformin, glucagon-like peptide-1 receptor agonists (GLP-1RA), dipeptidyl-peptidase-4 (DDP-4) inhibitors, α-glucosidase inhibitors (AGIs), sodium-glucose co-transporter-2 inhibitors, and thiazolidinediones (TZDs), directly affect AGEs through multiple mechanisms. These mechanisms include decreasing the formation of AGEs and the expression of AGEs receptor (RAGE) in tissue and increasing serum soluble RAGE levels, resulting in the reduced action of AGEs. Similarly, metformin, GLP-1RA, DDP-4 inhibitors, AGIs, and TZDs may enhance incretin hormones directly by increasing their production or suppressing their metabolism. Additionally, these medications could influence AGEs and the incretin pathway indirectly by enhancing glycemic control. In contrast, sulfonylureas have not demonstrated any obvious effects on AGEs or the incretin pathway. Considering their favorable effects on AGEs and the incretin pathway, a suitable selection of antidiabetic drugs may facilitate more protective effects on the bone in diabetic patients.
Collapse
Affiliation(s)
- Muthanna K. Zaki
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Mosul, Mosul,
Iraq
| | - Mohammed N. Abed
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul,
Iraq
| | - Fawaz A. Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Mosul, Mosul,
Iraq
| |
Collapse
|
2
|
Amengual-Cladera E, Morla-Barcelo PM, Morán-Costoya A, Sastre-Serra J, Pons DG, Valle A, Roca P, Nadal-Serrano M. Metformin: From Diabetes to Cancer-Unveiling Molecular Mechanisms and Therapeutic Strategies. BIOLOGY 2024; 13:302. [PMID: 38785784 PMCID: PMC11117706 DOI: 10.3390/biology13050302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/06/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
Metformin, a widely used anti-diabetic drug, has garnered attention for its potential in cancer management, particularly in breast and colorectal cancer. It is established that metformin reduces mitochondrial respiration, but its specific molecular targets within mitochondria vary. Proposed mechanisms include inhibiting mitochondrial respiratory chain Complex I and/or Complex IV, and mitochondrial glycerophosphate dehydrogenase, among others. These actions lead to cellular energy deficits, redox state changes, and several molecular changes that reduce hyperglycemia in type 2 diabetic patients. Clinical evidence supports metformin's role in cancer prevention in type 2 diabetes mellitus patients. Moreover, in these patients with breast and colorectal cancer, metformin consumption leads to an improvement in survival outcomes and prognosis. The synergistic effects of metformin with chemotherapy and immunotherapy highlights its potential as an adjunctive therapy for breast and colorectal cancer. However, nuanced findings underscore the need for further research and stratification by molecular subtype, particularly for breast cancer. This comprehensive review integrates metformin-related findings from epidemiological, clinical, and preclinical studies in breast and colorectal cancer. Here, we discuss current research addressed to define metformin's bioavailability and efficacy, exploring novel metformin-based compounds and drug delivery systems, including derivatives targeting mitochondria, combination therapies, and novel nanoformulations, showing enhanced anticancer effects.
Collapse
Affiliation(s)
- Emilia Amengual-Cladera
- Grupo Metabolismo Energético y Nutrición, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain; (E.A.-C.); (A.M.-C.); (A.V.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
| | - Pere Miquel Morla-Barcelo
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
| | - Andrea Morán-Costoya
- Grupo Metabolismo Energético y Nutrición, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain; (E.A.-C.); (A.M.-C.); (A.V.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
| | - Jorge Sastre-Serra
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Gabriel Pons
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
| | - Adamo Valle
- Grupo Metabolismo Energético y Nutrición, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain; (E.A.-C.); (A.M.-C.); (A.V.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pilar Roca
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn, CB06/03/0043), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercedes Nadal-Serrano
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma, Illes Balears, Spain; (P.M.M.-B.); (J.S.-S.); (D.G.P.); (M.N.-S.)
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universitat de les Illes Balears, Ctra. de Valldemossa, km 7.5, 07122 Palma, Illes Balears, Spain
| |
Collapse
|
3
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [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: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
4
|
Cheng M, Ren L, Jia X, Wang J, Cong B. Understanding the action mechanisms of metformin in the gastrointestinal tract. Front Pharmacol 2024; 15:1347047. [PMID: 38617792 PMCID: PMC11010946 DOI: 10.3389/fphar.2024.1347047] [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: 12/06/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Metformin is the initial medication recommended for the treatment of type 2 diabetes mellitus (T2DM). In addition to diabetes treatment, the function of metformin also can be anti-aging, antiviral, and anti-inflammatory. Nevertheless, further exploration is required to fully understand its mode of operation. Historically, the liver has been acknowledged as the main location where metformin reduces glucose levels, however, there is increasing evidence suggesting that the gastrointestinal tract also plays a significant role in its action. In the gastrointestinal tract, metformin effects glucose uptake and absorption, increases glucagon-like peptide-1 (GLP-1) secretion, alters the composition and structure of the gut microbiota, and modulates the immune response. However, the side effects of it cannot be ignored such as gastrointestinal distress in patients. This review outlines the impact of metformin on the digestive system and explores potential explanations for variations in metformin effectiveness and adverse effects like gastrointestinal discomfort.
Collapse
Affiliation(s)
- Meihui Cheng
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lili Ren
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianxian Jia
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Pathogen Biology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jianwei Wang
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Cong
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
5
|
Huber H, Schieren A, Holst JJ, Simon MC. Dietary impact on fasting and stimulated GLP-1 secretion in different metabolic conditions - a narrative review. Am J Clin Nutr 2024; 119:599-627. [PMID: 38218319 PMCID: PMC10972717 DOI: 10.1016/j.ajcnut.2024.01.007] [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: 09/07/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1), a gastrointestinal peptide and central mediator of glucose metabolism, is secreted by L cells in the intestine in response to food intake. Postprandial secretion of GLP-1 is triggered by nutrient-sensing via transporters and G-protein-coupled receptors (GPCRs). GLP-1 secretion may be lower in adults with obesity/overweight (OW) or type 2 diabetes mellitus (T2DM) than in those with normal glucose tolerance (NGT), but these findings are inconsistent. Because of the actions of GLP-1 on stimulating insulin secretion and promoting weight loss, GLP-1 and its analogs are used in pharmacologic preparations for the treatment of T2DM. However, physiologically stimulated GLP-1 secretion through the diet might be a preventive or synergistic method for improving glucose metabolism in individuals who are OW, or have impaired glucose tolerance (IGT) or T2DM. This narrative review focuses on fasting and postprandial GLP-1 secretion in individuals with different metabolic conditions and degrees of glucose intolerance. Further, the influence of relevant diet-related factors (e.g., specific diets, meal composition, and size, phytochemical content, and gut microbiome) that could affect fasting and postprandial GLP-1 secretion are discussed. Some studies showed diminished glucose- or meal-stimulated GLP-1 response in participants with T2DM, IGT, or OW compared with those with NGT, whereas other studies have reported an elevated or unchanged GLP-1 response in T2DM or IGT. Meal composition, especially the relationship between macronutrients and interventions targeting the microbiome can impact postprandial GLP-1 secretion, although it is not clear which macronutrients are strong stimulants of GLP-1. Moreover, glucose tolerance, antidiabetic treatment, grade of overweight/obesity, and sex were important factors influencing GLP-1 secretion. The results presented in this review highlight the potential of nutritional and physiologic stimulation of GLP-1 secretion. Further research on fasting and postprandial GLP-1 concentrations and the resulting metabolic consequences under different metabolic conditions is needed.
Collapse
Affiliation(s)
- Hanna Huber
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology, Mölndal, Sweden; Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Alina Schieren
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Marie-Christine Simon
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany.
| |
Collapse
|
6
|
Ji L, Lu J, Gao L, Yan X, Li J, Cheng Z, Zhang L, Tian J, Li P, Bai J, Xie D, Zhao J, Ding J, Yu Q, Wang T. A randomized, double-blind, placebo controlled, phase 3 trial to evaluate the efficacy and safety of cetagliptin added to ongoing metformin therapy in patients with uncontrolled type 2 diabetes with metformin monotherapy. Diabetes Obes Metab 2023; 25:3788-3797. [PMID: 37724698 DOI: 10.1111/dom.15274] [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: 07/18/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023]
Abstract
AIM This trial was designed to assess the efficacy and safety of cetagliptin added to metformin in Chinese patients with type 2 diabetes who had inadequate glycaemic control with metformin monotherapy. METHODS In total, 446 patients with type 2 diabetes on metformin monotherapy were randomized to receive the addition of once-daily cetagliptin 100 mg, cetagliptin 50 mg and placebo in a 2:2:1 ratio for 24-week double-blind treatment. At week 24, patients initially randomized to cetagliptin 50 mg and placebo were switched to cetagliptin 100 mg for 28 weeks open-label treatment. The primary endpoint was the change in haemoglobin A1c (HbA1c) from baseline, and the efficacy analyses were based on an all-patients-treated population using an analysis of co-variance. RESULTS After 24 weeks, both add-on therapies led to greater glycaemic control. Reductions in HbA1c from baseline were -1.17 ± 0.794%, -1.23 ± 0.896% in cetagliptin 100 mg and 50 mg plus metformin group, respectively. No difference was observed between the cetagliptin 100 mg and 50 mg plus metformin group. Patients with higher baseline HbA1c levels (≥8.5%) experienced greater reductions in HbA1c. A significantly greater proportion of patients achieved an HbA1c <7.0% with cetagliptin 100 mg (49.4%) and cetagliptin 50 mg (51.1%) plus metformin than metformin monotherapy (14.4%). Both combination therapies also improved the homeostasis model assessment β-function index and decreased systolic blood pressure. There was no increased risk of adverse effects with combination therapy, and both combination therapies were generally well tolerated. CONCLUSIONS The addition of cetagliptin once daily to metformin was more efficacious and well tolerated than metformin monotherapy in Chinese patients with type 2 diabetes who had inadequate glycaemic control with metformin monotherapy.
Collapse
Affiliation(s)
- Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Jinmiao Lu
- CGeneTech (Suzhou, China) Co., Ltd, Suzhou, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Leili Gao
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | | | - Jifang Li
- Puyang Oilfield general Hospital, Puyang, China
| | - Zhifeng Cheng
- The Fourth Hospital of Medical University, Harbin, China
| | - Lili Zhang
- Overview of Shijiazhuang Second Hospital, Shijiazhuang, China
| | | | - Ping Li
- Yuncheng Central Hospital, Yuncheng, China
| | - Jie Bai
- Liaocheng People's Hospital, Liaocheng, China
| | - Daosheng Xie
- Beijing noahpharm Medical Technology Co., Ltd, Beijing, China
| | - Jiahong Zhao
- CGeneTech (Suzhou, China) Co., Ltd, Suzhou, China
| | - Juping Ding
- CGeneTech (Suzhou, China) Co., Ltd, Suzhou, China
| | - Qiang Yu
- CGeneTech (Suzhou, China) Co., Ltd, Suzhou, China
| | - Tong Wang
- CGeneTech (Suzhou, China) Co., Ltd, Suzhou, China
| |
Collapse
|
7
|
Smits MM, Holst JJ. Endogenous glucagon-like peptide (GLP)-1 as alternative for GLP-1 receptor agonists: Could this work and how? Diabetes Metab Res Rev 2023; 39:e3699. [PMID: 37485788 DOI: 10.1002/dmrr.3699] [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: 10/26/2022] [Revised: 03/21/2023] [Accepted: 06/18/2023] [Indexed: 07/25/2023]
Abstract
In recent years, we have witnessed the many beneficial effects of glucagon-like peptide (GLP)-1 receptor agonists, including the reduction in cardiovascular risk in patients with type 2 diabetes, and the reduction of body weight in those with obesity. Increasing evidence suggests that these agents differ considerably from endogenous GLP-1 when it comes to their routes of action, although their clinical effects appear to be the same. Given the limitations of the GLP-1 receptor agonists, could it be useful to develop agents which stimulate GLP-1 release? Here we will discuss the differences and similarities between GLP-1 receptor agonists and endogenous GLP-1, and will detail how endogenous GLP-1-when stimulated appropriately-could have clinically relevant effects.
Collapse
Affiliation(s)
- Mark M Smits
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Mizoguchi M, Takemori H, Furukawa S, Ito M, Asai M, Morino H, Miura T, Yabe D, Shibata T. Increased expression of glucagon-like peptide-1 and cystic fibrosis transmembrane conductance regulator in the ileum and colon in mouse treated with metformin. Endocr J 2023; 70:149-159. [PMID: 36198615 DOI: 10.1507/endocrj.ej22-0260] [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] [Indexed: 11/23/2022] Open
Abstract
Metformin, an oral medication, is prescribed to patients with type 2 diabetes mellitus. Although the efficacy, safety, and low economic burden of metformin on patients have long been recognized, approximately 5% of the patients treated with this drug develop severe diarrhea and discontinue the treatment. We previously reported that 1,000 mg·kg-1·day-1 of metformin induced diarrhea in diabetic obese (db/db) mice and wood creosote (traditional medication for diarrhea) ameliorated the symptoms. In this study, we attempted to elucidate the molecular mechanisms by which metformin induces diarrhea. Cystic fibrosis transmembrane conductance regulator (CFTR) is a key ion (chloride) channel in cyclic adenosine monophosphate (cAMP)-induced diarrhea. Metformin treatment increased bile flow (bile acids and bilirubin) in the ileum of mice. In addition, the treatment was accompanied by an increase in mRNA and protein levels of CFTR in the mucosa of the ileum and colon in both wild-type (C57BL/6J) and db/db mice. Glucagon-like peptide-1 (GLP-1), as well as cholic acid, induces CFTR mRNA expression in human colon carcinoma Caco-2 cells through cAMP signaling. Although wood creosote (10 mg/kg) ameliorated diarrhea symptoms, it did not alter the mRNA levels of Glp-1 or Cftr. Similar to overeating, metformin upregulated GLP-1 and CFTR expression, which may have contributed to diarrhea symptoms in mice. Although we could not identify db/db mouse-specific factors associated with metformin-induced diarrhea, these factors may modulate colon function. Wood creosote may not interact with these factors but ameliorates diarrhea symptoms.
Collapse
Affiliation(s)
- Momoka Mizoguchi
- Department of Life Science and Chemistry, Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan
| | - Hiroshi Takemori
- Department of Life Science and Chemistry, Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Saho Furukawa
- Department of Life Science and Chemistry, Graduate School of Natural Science and Technology, Gifu University, Gifu 501-1193, Japan
| | - Masafumi Ito
- Taiko Pharmaceutical Co., Ltd., Osaka 550-0005, Japan
| | - Mutsumi Asai
- Taiko Pharmaceutical Co., Ltd., Osaka 550-0005, Japan
| | | | | | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism/Department of Rheumatology and Clinical Nutrition, Gifu University Graduate School of Medicine, Gifu 501-1193, Japan
| | - Takashi Shibata
- Taiko Pharmaceutical Co., Ltd., Osaka 550-0005, Japan
- Strategic Global Partnership Cross-Innovation Initiative, Graduate School of Medicine, Osaka University Hospital, Osaka 565-0871, Japan
| |
Collapse
|
9
|
Harada M, Kondo Y, Sugiyama M, Ohira A, Ichikawa M, Akiyama T, Orime K, Takai T, Yamakawa T, Terauchi Y. The METRO study: a retrospective analysis of the efficacy of metformin for type 2 diabetes in Japan. Endocr J 2023; 70:121-128. [PMID: 36261368 DOI: 10.1507/endocrj.ej22-0330] [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] [Indexed: 02/01/2023] Open
Abstract
Metformin monotherapy as first-line treatment for patients with type 2 diabetes (T2D) has been shown to effectively improve blood glucose levels and motivation to undergo treatment and prevent complications. However, no studies have reported its effect when combined with other drugs or compared the effect based on administration time. This study aimed to investigate the effect of metformin administration in Japanese patients with T2D, examine how the introduction line impacts the effect of metformin, and examine the characteristics of patients demonstrating improved blood glucose levels. Data on characteristics of patients who were newly prescribed metformin with no shifting of hypoglycemic agents in the subsequent 24-week observation period, and their age [mean, 56.8 years], body mass index [mean, 27.5 kg/m2], glycated hemoglobin [HbA1c] [mean, 8.1%], and duration of diabetes [mean, 3.0 years] were obtained from the medical records of 201 patients. The changes in HbA1c by introduction line after 24 weeks were -1.59%, -0.91%, -0.89%, and -0.65% in the first, second, third, and fourth induction lines, respectively; earlier introduction more significantly improved blood glucose. The factors significantly associated with HbA1c changes were early introduction, high baseline HbA1c, high estimated glomerular filtration rate, decreased insulin secretion, short estimated duration of diabetes, and increased metformin dose. Furthermore, factors contributing to the largest HbA1c improvement by metformin were high baseline HbA1c and early administration. Metformin is expected to lower blood glucose levels in Japanese patients with T2D, even in those with decreased insulin secretion, due to its early introduction as a first-line drug.
Collapse
Affiliation(s)
- Marina Harada
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Yoshinobu Kondo
- Tsunashima East Internal Medicine and Diabetes Clinic, Yokohama 223-0052, Japan
| | - Mai Sugiyama
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Akeo Ohira
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Masahiro Ichikawa
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Tomoaki Akiyama
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Kazuki Orime
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Takanori Takai
- Urafune Kanazawa Internal Medicine Clinic, Yokohama 232-0024, Japan
| | - Tadashi Yamakawa
- Department of Endocrinology and Diabetes, Yokohama City University Medical Center, Yokohama 232-0024, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
10
|
Di Magno L, Di Pastena F, Bordone R, Coni S, Canettieri G. The Mechanism of Action of Biguanides: New Answers to a Complex Question. Cancers (Basel) 2022; 14:cancers14133220. [PMID: 35804992 PMCID: PMC9265089 DOI: 10.3390/cancers14133220] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Biguanides are a family of antidiabetic drugs with documented anticancer properties in preclinical and clinical settings. Despite intensive investigation, how they exert their therapeutic effects is still debated. Many studies support the hypothesis that biguanides inhibit mitochondrial complex I, inducing energy stress and activating compensatory responses mediated by energy sensors. However, a major concern related to this “complex” model is that the therapeutic concentrations of biguanides found in the blood and tissues are much lower than the doses required to inhibit complex I, suggesting the involvement of additional mechanisms. This comprehensive review illustrates the current knowledge of pharmacokinetics, receptors, sensors, intracellular alterations, and the mechanism of action of biguanides in diabetes and cancer. The conditions of usage and variables affecting the response to these drugs, the effect on the immune system and microbiota, as well as the results from the most relevant clinical trials in cancer are also discussed.
Collapse
Affiliation(s)
- Laura Di Magno
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Fiorella Di Pastena
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Rosa Bordone
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Sonia Coni
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy; (L.D.M.); (F.D.P.); (R.B.); (S.C.)
- Istituto Pasteur—Fondazione Cenci—Bolognetti, 00161 Rome, Italy
- Correspondence:
| |
Collapse
|
11
|
Metabolic Action of Metformin. Pharmaceuticals (Basel) 2022; 15:ph15070810. [PMID: 35890109 PMCID: PMC9317619 DOI: 10.3390/ph15070810] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/14/2022] [Accepted: 06/26/2022] [Indexed: 12/12/2022] Open
Abstract
Metformin, a cheap and safe biguanide derivative, due to its ability to influence metabolism, is widely used as a first-line drug for type 2 diabetes (T2DM) treatment. Therefore, the aim of this review was to present the updated biochemical and molecular effects exerted by the drug. It has been well explored that metformin suppresses hepatic glucose production in both AMPK-independent and AMPK-dependent manners. Substantial scientific evidence also revealed that its action is related to decreased secretion of lipids from intestinal epithelial cells, as well as strengthened oxidation of fatty acids in adipose tissue and muscles. It was recognized that metformin’s supra-therapeutic doses suppress mitochondrial respiration in intestinal epithelial cells, whereas its therapeutic doses elevate cellular respiration in the liver. The drug is also suggested to improve systemic insulin sensitivity as a result of alteration in gut microbiota composition, maintenance of intestinal barrier integrity, and alleviation of low-grade inflammation.
Collapse
|
12
|
Vieira IH, Barros LM, Baptista CF, Rodrigues DM, Paiva IM. Recommendations for Practical Use of Metformin, a Central Pharmacological Therapy in Type 2 Diabetes. Clin Diabetes 2022; 40:97-107. [PMID: 35221479 PMCID: PMC8865803 DOI: 10.2337/cd21-0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Inês H. Vieira
- Coimbra Hospital and University Centre, Coimbra, Portugal
- Corresponding author: Inês H. Vieira,
| | | | | | - Dírcea M. Rodrigues
- Coimbra Hospital and University Centre, Coimbra, Portugal
- University of Coimbra, Coimbra, Portugal
| | | |
Collapse
|
13
|
|
14
|
The Function of Gastrointestinal Hormones in Obesity-Implications for the Regulation of Energy Intake. Nutrients 2021; 13:nu13061839. [PMID: 34072172 PMCID: PMC8226753 DOI: 10.3390/nu13061839] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity and the challenges of prevention prompted researchers to investigate the mechanisms that control food intake. Food ingestion triggers several physiological responses in the digestive system, including the release of gastrointestinal hormones from enteroendocrine cells that are involved in appetite signalling. Disturbed regulation of gut hormone release may affect energy homeostasis and contribute to obesity. In this review, we summarize the changes that occur in the gut hormone balance during the pre- and postprandial state in obesity and the alterations in the diurnal dynamics of their plasma levels. We further discuss how obesity may affect nutrient sensors on enteroendocrine cells that sense the luminal content and provoke alterations in their secretory profile. Gastric bypass surgery elicits one of the most favorable metabolic outcomes in obese patients. We summarize the effect of different strategies to induce weight loss on gut enteroendocrine function. Although the mechanisms underlying obesity are not fully understood, restoring the gut hormone balance in obesity by targeting nutrient sensors or by combination therapy with gut peptide mimetics represents a novel strategy to ameliorate obesity.
Collapse
|
15
|
El-Yazbi AF. Green methods for the simultaneous analysis of pharmaceutical mixtures present in disparate concentration ranges: Application to antidiabetic mixtures. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
16
|
LaMoia TE, Shulman GI. Cellular and Molecular Mechanisms of Metformin Action. Endocr Rev 2021; 42:77-96. [PMID: 32897388 PMCID: PMC7846086 DOI: 10.1210/endrev/bnaa023] [Citation(s) in RCA: 390] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
Metformin is a first-line therapy for the treatment of type 2 diabetes, due to its robust glucose-lowering effects, well-established safety profile, and relatively low cost. While metformin has been shown to have pleotropic effects on glucose metabolism, there is a general consensus that the major glucose-lowering effect in patients with type 2 diabetes is mostly mediated through inhibition of hepatic gluconeogenesis. However, despite decades of research, the mechanism by which metformin inhibits this process is still highly debated. A key reason for these discrepant effects is likely due to the inconsistency in dosage of metformin across studies. Widely studied mechanisms of action, such as complex I inhibition leading to AMPK activation, have only been observed in the context of supra-pharmacological (>1 mM) metformin concentrations, which do not occur in the clinical setting. Thus, these mechanisms have been challenged in recent years and new mechanisms have been proposed. Based on the observation that metformin alters cellular redox balance, a redox-dependent mechanism of action has been described by several groups. Recent studies have shown that clinically relevant (50-100 μM) concentrations of metformin inhibit hepatic gluconeogenesis in a substrate-selective manner both in vitro and in vivo, supporting a redox-dependent mechanism of metformin action. Here, we review the current literature regarding metformin's cellular and molecular mechanisms of action.
Collapse
Affiliation(s)
- Traci E LaMoia
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
17
|
Ji L, Li L, Ma J, Li X, Li D, Meng B, Lu W, Sun J, Liu Y, Takayanagi G, Wang Y. Efficacy and safety of teneligliptin added to metformin in Chinese patients with type 2 diabetes mellitus inadequately controlled with metformin: A phase 3, randomized, double-blind, placebo-controlled study. ENDOCRINOLOGY DIABETES & METABOLISM 2021; 4:e00222. [PMID: 33855222 PMCID: PMC8029565 DOI: 10.1002/edm2.222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/09/2020] [Accepted: 12/19/2020] [Indexed: 11/11/2022]
Abstract
Introduction We evaluated the efficacy and safety of teneligliptin compared with placebo when added to metformin therapy in Chinese patients with type 2 diabetes inadequately controlled with metformin monotherapy. Methods This multicentre, randomized, double-blind, placebo-controlled, parallel-group study enrolled type 2 diabetes patients with glycosylated haemoglobin (HbA1c) 7.0%-<10.0% and fasting plasma glucose (FPG) <270 mg/dl, receiving a stable metformin dose ≥1000 mg/day. Teneligliptin 20 mg or placebo was administered orally once daily (qd) before breakfast for 24 weeks. The primary efficacy end-point was change in HbA1c from baseline to Week 24. Safety end-points included the incidence of adverse events (AEs). Results The least square mean (LSM) change from baseline (standard error [SE]) was -0.72 (0.07) (95% confidence intervals [CI], -0.87, -0.58) for teneligliptin and -0.01 (0.07) (95% CI, -0.16, 0.13) for placebo. The differences (LSM ± SE) between the placebo and teneligliptin groups in HbA1c and FPG were -0.71% ± 0.11% (p < .0001) and -16.5 ± 4.7 mg/dl (p = .0005), respectively. Teneligliptin yielded significant changes in HbA1c (-0.81%; p < .0001) and FPG (-22.2 mg/dl; p < .0001) at Week 12. At Week 24, more patients achieved HbA1c <7.0% with teneligliptin (41.7%) compared with placebo (16.1%; p < .0001). Treatment-emergent AE incidence was similar with teneligliptin (58.9%) and placebo (68.3%); upper respiratory tract infection, hyperuricaemia and hyperlipidaemia were the most common AEs. Conclusions Teneligliptin 20 mg qd for 24 weeks added to ongoing metformin treatment significantly decreased HbA1c and FPG levels compared with placebo in Chinese type 2 diabetes patients. The combination was safe and tolerable.
Collapse
Affiliation(s)
- Linong Ji
- Peking University People's Hospital Beijing China
| | - Ling Li
- Shengjing Hospital of China Medical University Liaoning China
| | - Jianhua Ma
- Nanjing First Hospital Nanjing Jiangsu China
| | | | - Dongmei Li
- Inner Mongolia People's Hospital Inner Mongolia China
| | - Bangzhu Meng
- The Affiliated Hospital of Inner Mongolia University for Nationalities Inner Mongolia China
| | - Weiping Lu
- Huai'an First People's Hospital Nanjing Medical University Nanjing Jiangsu China
| | - Jiao Sun
- Huadong Hospital Affiliated to Fudan University Shanghai China
| | - Yanmei Liu
- Yancheng City No.1 People's Hospital Jiangsu China
| | - Gen Takayanagi
- Mitsubishi Tanabe Pharma Development America, Inc. Jersey City NJ USA
| | - Yi Wang
- Mitsubishi Tanabe Pharma Development (Beijing) Co., Ltd. Beijing China
| |
Collapse
|
18
|
Moon JH, Lim S. Pharmacotherapy for patients with diabetes mellitus. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2020. [DOI: 10.5124/jkma.2020.63.12.766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Diabetes mellitus (DM) is a complex, chronic illness requiring continuous medical care with multifactorial riskreduction strategies besides glycemic control. The pathophysiology of type 2 DM is characterized by a combination of insulin resistance in peripheral organs, including the liver, adipose tissues, and muscle, and inadequate insulin secretion from the pancreatic β-cells to compensate for insulin resistance, which eventually leads to β-cell failure. DM is accompanied by micro- and macro-vascular complications, including cardiovascular events and renal complications, resulting in high mortality rates. After insulin was first discovered in 1922, many antidiabetic agents including metformin, sulfonylureas, thiazolidinediones, and α-glucosidase inhibitors have been developed. Among them, metformin is the preferred pharmacologic agent for the initial treatment of DM. Recently, novel antidiabetic agents, such as dipeptidyl peptidase-4 inhibitors, sodium-glucose cotransporter-2 inhibitors, and glucagon-like peptide-1 receptor agonists, were introduced and are currently available for clinical practice. Studies with dipeptidyl peptidase-4 inhibitors showed non-inferiority compared with placebo, in terms of cardiovascular safety. Some glucagon-like peptide-1 receptor agonists (liraglutide, semaglutide, albiglutide, and dulaglutide) showed favorable results in both cardiovascular and renal outcomes. Sodium-glucose cotransporter-2 inhibitors (empagliflozin, canagliflozin, and dapagliflozin) also showed beneficial effects on cardiovascular and renal outcomes. It is important for clinicians to study novel DM medications and prescribe them accordingly to improve patients’ clinical outcomes.
Collapse
|
19
|
Deacon CF. Dipeptidyl peptidase 4 inhibitors in the treatment of type 2 diabetes mellitus. Nat Rev Endocrinol 2020; 16:642-653. [PMID: 32929230 DOI: 10.1038/s41574-020-0399-8] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/17/2022]
Abstract
Dipeptidyl peptidase 4 inhibitors (DPP4i) have been available for treating type 2 diabetes mellitus since 2006. Although they are a diverse group, DPP4i are all small, orally available molecules that interact with the catalytic site of DPP4 without disturbing any of its other known functions, including its effects on the immune system. DPP4i have no intrinsic glucose-lowering activity, so their efficacy as anti-diabetic agents is related directly to their ability to inhibit DPP4 activity and is mediated through the effects of the substrates they protect. Of these, the incretin hormone, glucagon-like peptide 1, is probably the most important. As the effects of glucagon-like peptide 1 are glucose-dependent, the risk of hypoglycaemia with DPP4i is low. Class effects, which are directly related to the mechanism of action, are common to all DPP4i; these include their overall good safety profile and tolerability, as well as their efficacy in improving glycaemic control, but also, potentially, a small increased risk of acute pancreatitis. Compound-specific effects are those related to their differing chemistries and/or pharmacokinetic profiles. These compound-specific effects could affect the way in which individual DPP4i are used therapeutically and potentially explain off-target adverse effects, such as hospitalization for heart failure, which is seen only with one DPP4i. Overall, DPP4i have a favourable therapeutic profile and are safe and effective in the majority of patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Carolyn F Deacon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
20
|
Plasma levels of DPP4 activity and sDPP4 are dissociated from inflammation in mice and humans. Nat Commun 2020; 11:3766. [PMID: 32724076 PMCID: PMC7387453 DOI: 10.1038/s41467-020-17556-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP4) modulates inflammation by enzymatic cleavage of immunoregulatory peptides and through its soluble form (sDPP4) that directly engages immune cells. Here we examine whether reduction of DPP4 activity alters inflammation. Prolonged DPP4 inhibition increases plasma levels of sDPP4, and induces sDPP4 expression in lymphocyte-enriched organs in mice. Bone marrow transplantation experiments identify hematopoietic cells as the predominant source of plasma sDPP4 following catalytic DPP4 inhibition. Surprisingly, systemic DPP4 inhibition increases plasma levels of inflammatory markers in regular chow-fed but not in high fat-fed mice. Plasma levels of sDPP4 and biomarkers of inflammation are lower in metformin-treated subjects with type 2 diabetes (T2D) and cardiovascular disease, yet exhibit considerable inter-individual variation. Sitagliptin therapy for 12 months reduces DPP4 activity yet does not increase markers of inflammation or levels of sDPP4. Collectively our findings dissociate levels of DPP4 enzyme activity, sDPP4 and biomarkers of inflammation in mice and humans.
Collapse
|
21
|
Panaro BL, Yusta B, Matthews D, Koehler JA, Song Y, Sandoval DA, Drucker DJ. Intestine-selective reduction of Gcg expression reveals the importance of the distal gut for GLP-1 secretion. Mol Metab 2020; 37:100990. [PMID: 32278655 PMCID: PMC7200938 DOI: 10.1016/j.molmet.2020.100990] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Glucagon-like peptide-1 is a nutrient-sensitive hormone secreted from enteroendocrine L cells within the small and large bowel. Although GLP-1 levels rise rapidly in response to food ingestion, the greatest density of L cells is localized to the distal small bowel and colon. Here, we assessed the importance of the distal gut in the acute L cell response to diverse secretagogues. METHODS Circulating levels of glucose and plasma GLP-1 were measured in response to the administration of L cell secretagogues in wild-type mice and in mice with (1) genetic reduction of Gcg expression throughout the small bowel and large bowel (GcgGut-/-) and (2) selective reduction of Gcg expression in the distal gut (GcgDistalGut-/-). RESULTS The acute GLP-1 response to olive oil or arginine administration was markedly diminished in GcgGut-/- but preserved in GcgDistalGut-/- mice. In contrast, the increase in plasma GLP-1 levels following the administration of the GPR119 agonist AR231453, or the melanocortin-4 receptor (MC4R) agonist LY2112688, was markedly diminished in the GcgDistalGut-/- mice. The GLP-1 response to LPS was also markedly attenuated in the GcgGut-/- mice and remained submaximal in the GcgDistalGut-/- mice. Doses of metformin sufficient to lower glucose and increase GLP-1 levels in the GcgGut+/+ mice retained their glucoregulatory activity, yet they failed to increase GLP-1 levels in the GcgGut-/- mice. Surprisingly, the actions of metformin to increase plasma GLP-1 levels were substantially attenuated in the GcgDistalGut-/- mice. CONCLUSION These findings further establish the importance of the proximal gut for the acute response to nutrient-related GLP-1 secretagogues. In contrast, we identify essential contributions of the distal gut to (i) the rapid induction of circulating GLP-1 levels in response to pharmacological selective agonism of G-protein-coupled receptors, (ii) the increased GLP-1 levels following the activation of Toll-Like Receptors with LPS, and iii) the acute GLP-1 response to metformin. Collectively, these results reveal that distal gut Gcg + endocrine cells are rapid responders to structurally and functionally diverse GLP-1 secretagogues.
Collapse
Affiliation(s)
- Brandon L Panaro
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Bernardo Yusta
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Dianne Matthews
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jacqueline A Koehler
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Youngmi Song
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Daniel J Drucker
- Department of Medicine and the Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Gao Y, Zhang S, Zhang Y, Qian J. Identification of MicroRNA-Target Gene-Transcription Factor Regulatory Networks in Colorectal Adenoma Using Microarray Expression Data. Front Genet 2020; 11:463. [PMID: 32508878 PMCID: PMC7248367 DOI: 10.3389/fgene.2020.00463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/15/2020] [Indexed: 12/18/2022] Open
Abstract
Objective The aim of the study was to find the key genes, microRNAs (miRNAs) and transcription factors (TFs) and construct miRNA-target gene-TF regulatory networks to investigate the underlying molecular mechanism in colorectal adenoma (CRA). Methods Four mRNA expression datasets and one miRNA expression dataset were downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed miRNAs (DEMs) and differentially expressed genes (DEGs) were identified between CRA and normal samples. Moreover, functional enrichment analysis for DEGs was carried out utilizing the Cytoscape-plugin, known as ClueGO. These DEGs were mapped to STRING database to construct a protein-protein interaction (PPI) network. Then, a miRNA-target gene regulatory network was established to screen key DEMs. In addition, similar workflow of the analyses were also performed comparing the CRC samples with CRA ones to screen key DEMs. Finally, miRNA-target gene-TF regulatory networks were constructed for these key DEMs using iRegulon plug-in in Cytoscape. Results We identified 514 DEGs and 167 DEMs in CRA samples compared to healthy samples. Functional enrichment analysis revealed that these DEGs were significantly enriched in several terms and pathways, such as regulation of cell migration and bile secretion pathway. A PPI network was constructed including 325 nodes as well as 890 edges. A total of 59 DEGs and 65 DEMs were identified in CRC samples compared to CRA ones. In addition, Two key DEMs in CRA samples compared to healthy samples were identified, such as hsa-miR-34a and hsa-miR-96. One key DEM, hsa-miR-29c, which was identified when we compared the differentially expressed molecules found in the comparison CRA versus normal samples to the ones obtained in the comparison CRC versus CRA, was also identified in CRC samples compared to CRA ones. The miRNA-target gene-TF regulatory networks for these key miRNAs included two TFs, one TF and five TFs, respectively. Conclusion These identified key genes, miRNA, TFs and miRNA-target gene-TF regulatory networks associated with CRA, to a certain degree, may provide some hints to enable us to better understand the underlying pathogenesis of CRA.
Collapse
Affiliation(s)
- Yadong Gao
- Department of Gastroenterology, The Second Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, The First People's Hospital of Nantong, Nantong, China
| | - Shenglai Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, The First People's Hospital of Nantong, Nantong, China
| | - Yan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, The First People's Hospital of Nantong, Nantong, China
| | - Junbo Qian
- Department of Gastroenterology, The Second Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, The First People's Hospital of Nantong, Nantong, China
| |
Collapse
|
23
|
Sansome DJ, Xie C, Veedfald S, Horowitz M, Rayner CK, Wu T. Mechanism of glucose-lowering by metformin in type 2 diabetes: Role of bile acids. Diabetes Obes Metab 2020; 22:141-148. [PMID: 31468642 DOI: 10.1111/dom.13869] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/07/2019] [Accepted: 08/28/2019] [Indexed: 02/05/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is an increasingly prevalent chronic condition, characterized by abnormally elevated blood glucose concentrations and, as a consequence, increased risk of micro- and macrovascular complications. Metformin is usually the first-line glucose-lowering medication in T2DM; however, despite being used for more than 60 years, the mechanism underlying the glucose-lowering action of metformin remains incompletely understood. Although metformin reduces hepatic glucose production, there is persuasive evidence that the gastrointestinal tract is crucial in mediating this effect, particularly via secretion of the incretin hormone glucagon-like peptide 1 (GLP-1). It is now well recognized that bile acids, in addition to their established function in fat digestion and absorption, are important regulators of glucose metabolism. Exposure of the small and large intestine to bile acids induces GLP-1 secretion, modulates the composition of the gut microbiota, and reduces postprandial blood glucose excursions in humans with and without T2DM. Metformin reduces intestinal bile acid resorption substantially, such that intraluminal bile acids may, at least in part, account for its glucose-lowering effect. The present review focuses on the conceptual shift in our understanding as to how metformin lowers blood glucose in T2DM, with a particular emphasis on the role of intestinal bile acids.
Collapse
Affiliation(s)
- Daniel J Sansome
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Cong Xie
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Simon Veedfald
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Christopher K Rayner
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
24
|
Shawky LM, Morsi AA, El Bana E, Hanafy SM. The Biological Impacts of Sitagliptin on the Pancreas of a Rat Model of Type 2 Diabetes Mellitus: Drug Interactions with Metformin. BIOLOGY 2019; 9:E6. [PMID: 31881657 PMCID: PMC7167819 DOI: 10.3390/biology9010006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
Sitagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, is a beneficial class of antidiabetic drugs. However, a major debate about the risk of developing pancreatitis is still existing. The aim of the work was to study the histological and immunohistochemical effects of sitagliptin on both endocrine and exocrine pancreases in a rat model of type 2 diabetes mellitus and to correlate these effects with the biochemical findings. Moreover, a possible synergistic effect of sitagliptin, in combination with metformin, was also evaluated. Fifty adult male rats were used and assigned into five equal groups. Group 1 served as control. Group 2 comprised of untreated diabetic rats. Group 3 diabetic rats received sitagliptin. Group 4 diabetic rats received metformin. Group 5 diabetic rats received both combined. Treatments were given for 4 weeks after the induction of diabetes. Blood samples were collected for biochemical assay before the sacrification of rats. Pancreases were removed, weighed, and were processed for histological and immunohistochemical examination. In the untreated diabetic group, the islets appeared shrunken with disturbed architecture and abnormal immunohistochemical reactions for insulin, caspase-3, and inducible nitric oxide synthase (iNOS). The biochemical findings were also disturbed. Morphometrically, there was a significant decrease in the islet size and islet number. Treatment with sitagliptin, metformin, and their combination showed an improvement, with the best response in the combined approach. No evidence of pancreatic injury was identified in the sitagliptin-treated groups. In conclusion, sitagliptin had a cytoprotective effect on beta-cell damage. Furthermore, the data didn't indicate any detrimental effects of sitagliptin on the exocrine pancreas.
Collapse
Affiliation(s)
- Lamiaa M. Shawky
- Department of Histology and Cell Biology, Benha Faculty of Medicine, Benha University, Benha 13511, Egypt;
| | - Ahmed A. Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Eman El Bana
- Department of Anatomy, Benha Faculty of Medicine, Benha University, Benha 13511, Egypt;
| | - Safaa Masoud Hanafy
- Department of Anatomy, Faculty of Medicine for Girls, Al-Azhar University, Cairo 11865, Egypt;
| |
Collapse
|
25
|
Zilov AV, Abdelaziz SI, AlShammary A, Al Zahrani A, Amir A, Assaad Khalil SH, Brand K, Elkafrawy N, Hassoun AA, Jahed A, Jarrah N, Mrabeti S, Paruk I. Mechanisms of action of metformin with special reference to cardiovascular protection. Diabetes Metab Res Rev 2019; 35:e3173. [PMID: 31021474 PMCID: PMC6851752 DOI: 10.1002/dmrr.3173] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Management guidelines continue to identify metformin as initial pharmacologic antidiabetic therapy of choice for people with type 2 diabetes without contraindications, despite recent randomized trials that have demonstrated significant improvements in cardiovascular outcomes with newer classes of antidiabetic therapies. The purpose of this review is to summarize the current state of knowledge of metformin's therapeutic actions on blood glucose and cardiovascular clinical evidence and to consider the mechanisms that underlie them. The effects of metformin on glycaemia occur mainly in the liver, but metformin-stimulated glucose disposal by the gut has emerged as an increasingly import site of action of metformin. Additionally, metformin induces increased secretion of GLP-1 from intestinal L-cells. Clinical cardiovascular protection with metformin is supported by three randomized outcomes trials (in newly diagnosed and late stage insulin-treated type 2 diabetes patients) and a wealth of observational data. Initial evidence suggests that cotreatment with metformin may enhance the impact of newer incretin-based therapies on cardiovascular outcomes, an important observation as metformin can be combined with any other antidiabetic agent. Multiple potential mechanisms support the concept of cardiovascular protection with metformin beyond those provided by reduced blood glucose, including weight loss, improvements in haemostatic function, reduced inflammation, and oxidative stress, and inhibition of key steps in the process of atherosclerosis. Accordingly, metformin remains well placed to support improvements in cardiovascular outcomes, from diagnosis and throughout the course of type 2 diabetes, even in this new age of improved outcomes in type 2 diabetes.
Collapse
Affiliation(s)
- Alexey V. Zilov
- Department of EndocrinologySechenov's First Moscow State Medical UniversityMoscowRussia
| | | | - Afaf AlShammary
- Diabetes Center, Department of MedicineKing Abdulaziz Medical CityRiyadhKingdom of Saudi Arabia
| | - Ali Al Zahrani
- Department of Medicine, Molecular Endocrinology Section, Department of Molecular Oncology, Research CenterKing Faisal Specialist Hospital & Research CentreRiyadhKingdom of Saudi Arabia
| | - Ashraf Amir
- Department of Family MedicineInternational Medical CenterJeddahKingdom of Saudi Arabia
| | - Samir Helmy Assaad Khalil
- Department of Internal Medicine, Unit of Diabetology, Lipidology & Metabolism, Alexandria Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Kerstin Brand
- Global Medical AffairsMerck Healthcare KGaADarmstadtGermany
| | - Nabil Elkafrawy
- Diabetes and Endocrinology UnitMenoufia UniversityAl MinufyaEgypt
| | | | - Adel Jahed
- Gabric Diabetes Education Association, Tehran, Iran and Consultant EndocrinologistTehran General HospitalTehranIran
| | - Nadim Jarrah
- Internal Medicine DepartmentThe Specialty HospitalAmmanJordan
| | - Sanaa Mrabeti
- General Medicine and EndocrinologyMedical Affairs EMEA Merck Serono Middle East FZ‐LLCDubaiUnited Arab Emirates
| | - Imran Paruk
- Department of Diabetes and Endocrinology, Nelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
26
|
Mo SY, Lai OM, Chew BH, Ismail R, Bakar SA, Jabbar NA, Teng KT. Interesterified palm olein lowers postprandial glucose-dependent insulinotropic polypeptide response in type 2 diabetes. Eur J Nutr 2019; 58:1873-1885. [PMID: 29872922 DOI: 10.1007/s00394-018-1738-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/29/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE We aim to investigate the postprandial effects of palm olein (PO) and chemically interesterified palm olein (IPO) with different proportions of palmitic acid at the sn-2 position using high oleic sunflower oil (HOS) as control fat on concentrations of gut hormones, glucose homeostasis, satiety, lipid and inflammatory parameters in type 2 diabetic (T2D) subjects. METHODS Using a randomised double-blind crossover design, 21 (men = 6, women = 15) T2D subjects consumed test meals (3.65 MJ) consisting of a high fat muffin (containing 50 g test fats provided as PO, IPO or HOS) and a milkshake. Postprandial changes in gut hormones, glucose homeostasis, satiety, lipid and inflammatory parameters after meals were analysed. Some of the solid fractions of the IPO were removed and thus the fatty acid composition of the PO and IPO was not entirely equal (PO vs IPO: palmitate 39.8 vs 38.7; oleate 43.6 vs 45.1). PO, IPO and HOS contained 9.7, 38.9 and 0.2 g/100 g total fatty acids of palmitic acid at the sn-2 position, respectively. At 37 °C, IPO contained 4.2% SFC whereas PO and HOS were completely melted. RESULTS Our novel observation shows that the incremental area under curve (iAUC) 0-6 h of plasma GIP concentration was on average 16% lower following IPO meal compared with PO and HOS (P < 0.05) meals. Serum C-peptide concentrations exhibited a significant meal × gender interaction (P = 0.009). No differences between test meals were noted for other measurements. CONCLUSIONS This study shows no adverse effect of interesterification on hormones associated with glucose homeostasis notably GLP-1 in T2D subjects. TRIAL REGISTRATION ClinicalTrials.gov NCT01906359. https://clinicaltrials.gov/ct2/show/NCT01906359.
Collapse
Affiliation(s)
- Shuen-Yeing Mo
- Product Development and Advisory Services, Malaysian Palm Oil Board, Kajang, Selangor, Malaysia
- Department of Bioprocess Engineering, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Serdang, Selangor, Malaysia
| | - Oi-Ming Lai
- Department of Bioprocess Engineering, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang, Serdang, Selangor, Malaysia
- Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Serdang, Selangor, Malaysia
| | - Boon-How Chew
- Department of Family Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Serdang, Selangor, Malaysia
| | - Ruhaini Ismail
- Sepang District Health Office, Selangor State Health Department, Sepang, Selangor, Malaysia
| | - Sallehudin Abu Bakar
- Hulu Langat District Health Office, Selangor State Health Department, Kajang, Selangor, Malaysia
| | - Norli Abdul Jabbar
- Non-communicable Diseases Unit, Selangor State Health Department, Shah Alam, Selangor, Malaysia
| | - Kim-Tiu Teng
- Product Development and Advisory Services, Malaysian Palm Oil Board, Kajang, Selangor, Malaysia.
| |
Collapse
|
27
|
Takahashi H, Nishimura R, Tsujino D, Utsunomiya K. Which is better, high-dose metformin monotherapy or low-dose metformin/linagliptin combination therapy, in improving glycemic variability in type 2 diabetes patients with insufficient glycemic control despite low-dose metformin monotherapy? A randomized, cross-over, continuous glucose monitoring-based pilot study. J Diabetes Investig 2019; 10:714-722. [PMID: 30171747 PMCID: PMC6497608 DOI: 10.1111/jdi.12922] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/06/2018] [Accepted: 08/19/2018] [Indexed: 01/06/2023] Open
Abstract
AIMS/INTRODUCTION The present study investigated the effect of high-dose metformin or low-dose metformin/linagliptin combination therapy on glycemic variability (GV) in type 2 diabetes patients with insufficient glycemic control despite low-dose metformin monotherapy in a cross-over study using continuous glucose monitoring. MATERIALS AND METHODS The present study was carried out with 11 type 2 diabetes outpatients (7% < glycated hemoglobin < 10%) receiving low-dose metformin monotherapy (500-1,000 mg). All patients were assigned to either metformin 1,500 mg monotherapy (HMET) or combination therapy of low-dose (750 mg) metformin and linagliptin 5 mg (LMET + dipeptidyl peptidase-4 [DPP4]). GV was evaluated by continuous glucose monitoring after >4 weeks of the initial treatment and again after cross-over to the other treatment. GV metrics were compared between the treatments using the Wilcoxon signed-rank test. RESULTS Of the continuous glucose monitoring-derived GV metrics for the HMET versus LMET + DPP4, mean glucose levels, standard deviations and mean amplitude of glucose excursions were not significantly different. Although the pre-breakfast glucose levels were not significantly different among the treatments (P = 0.248), the 3-h postprandial glucose area under the curve (>160 mg/dL) after breakfast was significantly larger with HMET versus LMET + DPP4 (9,550 [2,075-11,395] vs 4,065 [1,950-8,895]; P = 0.041). CONCLUSIONS A comparison of GV with HMET versus LMET + DPP4 suggested that LMET + DPP4 might reduce post-breakfast GV to a greater degree than HMET in type 2 diabetes patients receiving low-dose metformin monotherapy.
Collapse
Affiliation(s)
- Hiroshi Takahashi
- Division of Diabetes, Metabolism and EndocrinologyDepartment of Internal MedicineJikei University School of MedicineTokyoJapan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism and EndocrinologyDepartment of Internal MedicineJikei University School of MedicineTokyoJapan
| | - Daisuke Tsujino
- Division of Diabetes, Metabolism and EndocrinologyDepartment of Internal MedicineJikei University School of MedicineTokyoJapan
| | - Kazunori Utsunomiya
- Division of Diabetes, Metabolism and EndocrinologyDepartment of Internal MedicineJikei University School of MedicineTokyoJapan
| |
Collapse
|
28
|
Maher HM, Abdelrahman AE, Alzoman NZ, Aljohar HI. Stability-indicating capillary electrophoresis method for the simultaneous determination of metformin hydrochloride, saxagliptin hydrochloride, and dapagliflozin in pharmaceutical tablets. J LIQ CHROMATOGR R T 2019. [DOI: 10.1080/10826076.2019.1590208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Hadir M. Maher
- College of Pharmacy, Department of Pharmaceutical Chemistry, King Saud University, Riyadh, Saudi Arabia
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, University of Alexandria, Alexandria, Egypt
| | - Afnan E. Abdelrahman
- College of Pharmacy, Department of Pharmaceutical Chemistry, King Saud University, Riyadh, Saudi Arabia
| | - Nourah Z. Alzoman
- College of Pharmacy, Department of Pharmaceutical Chemistry, King Saud University, Riyadh, Saudi Arabia
| | - Haya I. Aljohar
- College of Pharmacy, Department of Pharmaceutical Chemistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Rehman K, Ali MB, Akash MSH. Genistein enhances the secretion of glucagon-like peptide-1 (GLP-1) via downregulation of inflammatory responses. Biomed Pharmacother 2019; 112:108670. [PMID: 30784939 DOI: 10.1016/j.biopha.2019.108670] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) an incretin hormone, is known to regulate the glucose-mediated insulin secretion. However, reduction in the level of GLP-1 is considered to be a major cause for the reduction of GLP-1-dependent insulin secretory response. Genistein an isoflavone, is an important polyphenol and has wide range of therapeutic potentials, but its therapeutic effects alone and/or in combination with metformin on GLP-1 secretion have not been investigated yet. Hence, we aimed to investigate the stimulatory action of genistein in combination with metformin on GLP-1 via downregulation of inflammatory mediators, hyperlipidemia and hyperglycemia in alloxan-induced diabetic rats. Diabetes was induced in experimental rats by single administration of alloxan intraperitoneally. Metformin (50 mg/kg/day), genistein (20 mg/kg/day) and combination of genistein and metformin was administered in alloxan-induced diabetic rats. We found that genistein alone and/or in combination with metformin significantly increased the serum level (P < 0.01) and tissue content (P < 0.05) of GLP-1 in intestine when compared with that of metformin-treated animals. Similarly, genistein alone and/or in combination with metformin also resulted in normoglycemia (P < 0.001), glucose tolerance (P < 0.01), insulin sensitivity (P < 0.0001), hyperlipidemia (P < 0.01), liver and kidney function biomarkers (P < 0.01) as compared to that of metformin-treated experimental animals. Moreover, genistein alone and/or in combination with metformin also downregulated the inflammatory responses by decreasing the levels of interleuin-6, tumor necrosis factor-α and C-reactive protein in serum (P < 0.05) and intestine (P < 0.001) more efficiently as compared to that of metformin-treated experimental animals. The downregulation of inflammatory responses in intestine, was positively associated with increased secretion of GLP-1 from intestine. Histopathology of pancreas and intestine also showed that genistein significantly improved the deleterious effects of alloxan on pancreas and intestine. Hence, our work provides new insights on the synergistic effects of genistein and metformin on GLP-1 secretion. This may significantly improve the perception for proposing new GLP-1-based synergistic approaches for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Mehwish Bagh Ali
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
30
|
Bahne E, Sun EWL, Young RL, Hansen M, Sonne DP, Hansen JS, Rohde U, Liou AP, Jackson ML, de Fontgalland D, Rabbitt P, Hollington P, Sposato L, Due S, Wattchow DA, Rehfeld JF, Holst JJ, Keating DJ, Vilsbøll T, Knop FK. Metformin-induced glucagon-like peptide-1 secretion contributes to the actions of metformin in type 2 diabetes. JCI Insight 2018; 3:93936. [PMID: 30518693 DOI: 10.1172/jci.insight.93936] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Metformin reduces plasma glucose and has been shown to increase glucagon-like peptide 1 (GLP-1) secretion. Whether this is a direct action of metformin on GLP-1 release, and whether some of the glucose-lowering effect of metformin occurs due to GLP-1 release, is unknown. The current study investigated metformin-induced GLP-1 secretion and its contribution to the overall glucose-lowering effect of metformin and underlying mechanisms in patients with type 2 diabetes. METHODS Twelve patients with type 2 diabetes were included in this placebo-controlled, double-blinded study. On 4 separate days, the patients received metformin (1,500 mg) or placebo suspended in a liquid meal, with subsequent i.v. infusion of the GLP-1 receptor antagonist exendin9-39 (Ex9-39) or saline. During 240 minutes, blood was sampled. The direct effect of metformin on GLP-1 secretion was tested ex vivo in human ileal and colonic tissue with and without dorsomorphin-induced inhibiting of the AMPK activity. RESULTS Metformin increased postprandial GLP-1 secretion compared with placebo (P = 0.014), and the postprandial glucose excursions were significantly smaller after metformin + saline compared with metformin + Ex9-39 (P = 0.004). Ex vivo metformin acutely increased GLP-1 secretion (colonic tissue, P < 0.01; ileal tissue, P < 0.05), but the effect was abolished by inhibition of AMPK activity. CONCLUSIONS Metformin has a direct and AMPK-dependent effect on GLP-1-secreting L cells and increases postprandial GLP-1 secretion, which seems to contribute to metformin's glucose-lowering effect and mode of action. TRIAL REGISTRATION NCT02050074 (https://clinicaltrials.gov/ct2/show/NCT02050074). FUNDING This study received grants from the A.P. Møller Foundation, the Novo Nordisk Foundation, the Danish Medical Association research grant, the Australian Research Council, the National Health and Medical Research Council, and Pfizer Inc.
Collapse
Affiliation(s)
- Emilie Bahne
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Emily W L Sun
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
| | - Richard L Young
- Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Morten Hansen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark
| | - David P Sonne
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Pharmacology, Frederiksberg and Bispebjerg Hospital, University of Copenhagen, Denmark
| | - Jakob S Hansen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Ulrich Rohde
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark
| | - Alice P Liou
- Cardiovascular and Metabolic Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, USA
| | - Margaret L Jackson
- Cardiovascular and Metabolic Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, USA
| | - Dayan de Fontgalland
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - Philippa Rabbitt
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - Paul Hollington
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - Luigi Sposato
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - Steven Due
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- Discipline of Surgery, Flinders University, Adelaide, South Australia, Australia
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark
| | - Damien J Keating
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia.,Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Tina Vilsbøll
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Cai X, Gao X, Yang W, Ji L. Disparities in the Efficacy of Metformin in Combination with Dipeptidyl Peptidase-4 Inhibitor as Initial Treatment Stratified by Dosage and Ethnicity: A Meta-Analysis. Diabetes Technol Ther 2018; 20:704-714. [PMID: 30095971 DOI: 10.1089/dia.2018.0124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND As initial combination therapy of metformin and dipeptidyl peptidase-4 (DPP-4) inhibitor, the efficacy and safety for the use of high dose of metformin or low dose of metformin and the efficacy and safety for the combination use for Asian and Caucasian patients were not clear. METHODS Double-blind randomized controlled trials comparing the efficacy of initial combination therapy of metformin and DPP-4 inhibitors with metformin monotherapy were included. The primary outcome was a result of comparisons between high-dose combination therapy and low-dose combination therapy in terms of efficacy and safety. RESULTS A total of 11 studies were included. The results indicated that the high-dose combination therapy showed significant decreases in hemoglobin A1c (HbA1c) (-0.32%, P < 0.05), fasting plasma glucose (FPG) (-0.63 mmol/L, P < 0.05), and postprandial glucose (PPG) (-0.99 mmol/L, P < 0.05), but less increase in body weight (-0.54 kg, P < 0.05) when compared with low-dose combination therapy, corrected by metformin monotherapy. Moreover, the high-dose combination therapy exhibited significant decreases in HbA1c (-0.24%, P < 0.05), FPG (-0.54 mmol/L, P < 0.05), and PPG (-0.94 mmol/L, P < 0.05) in the Caucasian population than in the Asian population, corrected by metformin monotherapy. CONCLUSION As an initial treatment, the high dose of metformin in combination with DPP-4 inhibitors not only provided better glycemic control but also had less effect on weight gain compared with the low-dose combination therapy through the correction of metformin monotherapy. Moreover, initial combination therapy in the Caucasian population showed better glycemic control and less increase in body weight compared with the Asian population.
Collapse
Affiliation(s)
- Xiaoling Cai
- Department of Endocrine and Metabolism, Peking University People's Hospital , Beijing, China
| | - Xueying Gao
- Department of Endocrine and Metabolism, Peking University People's Hospital , Beijing, China
| | - Wenjia Yang
- Department of Endocrine and Metabolism, Peking University People's Hospital , Beijing, China
| | - Linong Ji
- Department of Endocrine and Metabolism, Peking University People's Hospital , Beijing, China
| |
Collapse
|
32
|
Corbin KD, Driscoll KA, Pratley RE, Smith SR, Maahs DM, Mayer-Davis EJ. Obesity in Type 1 Diabetes: Pathophysiology, Clinical Impact, and Mechanisms. Endocr Rev 2018; 39:629-663. [PMID: 30060120 DOI: 10.1210/er.2017-00191] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 06/21/2018] [Indexed: 02/07/2023]
Abstract
There has been an alarming increase in the prevalence of obesity in people with type 1 diabetes in recent years. Although obesity has long been recognized as a major risk factor for the development of type 2 diabetes and a catalyst for complications, much less is known about the role of obesity in the initiation and pathogenesis of type 1 diabetes. Emerging evidence suggests that obesity contributes to insulin resistance, dyslipidemia, and cardiometabolic complications in type 1 diabetes. Unique therapeutic strategies may be required to address these comorbidities within the context of intensive insulin therapy, which promotes weight gain. There is an urgent need for clinical guidelines for the prevention and management of obesity in type 1 diabetes. The development of these recommendations will require a transdisciplinary research strategy addressing metabolism, molecular mechanisms, lifestyle, neuropsychology, and novel therapeutics. In this review, the prevalence, clinical impact, energy balance physiology, and potential mechanisms of obesity in type 1 diabetes are described, with a special focus on the substantial gaps in knowledge in this field. Our goal is to provide a framework for the evidence base needed to develop type 1 diabetes-specific weight management recommendations that account for the competing outcomes of glycemic control and weight management.
Collapse
Affiliation(s)
- Karen D Corbin
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Kimberly A Driscoll
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, Colorado.,Barbara Davis Center for Diabetes, Aurora, Colorado
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida
| | - David M Maahs
- Division of Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
33
|
Bae JH, Kim LK, Min SH, Ahn CH, Cho YM. Postprandial glucose-lowering effect of premeal consumption of protein-enriched, dietary fiber-fortified bar in individuals with type 2 diabetes mellitus or normal glucose tolerance. J Diabetes Investig 2018; 9:1110-1118. [PMID: 29502350 PMCID: PMC6123026 DOI: 10.1111/jdi.12831] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/21/2017] [Accepted: 02/26/2018] [Indexed: 12/24/2022] Open
Abstract
AIMS/INTRODUCTION Protein preload improves postprandial glycemia by stimulating secretion of insulin and incretin hormones. However, it requires a large dose of protein to produce a significant effect. The present study was carried out to investigate the postprandial glucose-lowering effect of a premeal protein-enriched, dietary fiber-fortified bar (PFB), which contains moderate amounts of protein, in individuals with type 2 diabetes mellitus or normal glucose tolerance (NGT). MATERIALS AND METHODS The participants (15 type 2 diabetes mellitus and 15 NGT) were randomly assigned to either a premeal or postmeal PFB group and underwent two mixed meal tolerance tests, 1 week apart in reverse order. Plasma levels of glucose, insulin, glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide were measured. RESULTS During the mixed meal tolerance tests, the incremental area under the curve from 0 to 180 min of plasma glucose levels was lower with premeal PFB than with postmeal PFB in the type 2 diabetes mellitus (14,723 ± 1,310 mg min/dL vs 19,642 ± 1,367 mg min/dL; P = 0.0002) and NGT participants (3,943 ± 416 mg min/dL vs 4,827 ± 520 mg min/dL, P = 0.0296). In the type 2 diabetes mellitus participants, insulinogenic index and the incremental area under the curve from 0 to 180 min of plasma total glucagon-like peptide-1 levels were higher with premeal PFB than with postmeal PFB, but not in the NGT participants. There was no difference in postprandial glucose-dependent insulinotropic polypeptide levels between premeal and postmeal PFB in both groups. CONCLUSIONS Acute administration of premeal PFB decreased postprandial glucose excursion in both type 2 diabetes mellitus and NGT participants. In the type 2 diabetes mellitus participants, premeal PFB augmented the early-phase insulin secretion, possibly through enhancing glucagon-like peptide-1 secretion.
Collapse
Affiliation(s)
- Jae Hyun Bae
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| | - Lee Kyung Kim
- Department of Internal MedicineCheju Halla General HospitalJejuKorea
| | - Se Hee Min
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| | - Chang Ho Ahn
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| | - Young Min Cho
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| |
Collapse
|
34
|
Ruetten H, Gebauer M, Raymond RH, Calle RA, Cobelli C, Ghosh A, Robertson RP, Shankar SS, Staten MA, Stefanovski D, Vella A, Wright K, Fryburg DA. Mixed Meal and Intravenous L-Arginine Tests Both Stimulate Incretin Release Across Glucose Tolerance in Man: Lack of Correlation with β Cell Function. Metab Syndr Relat Disord 2018; 16:406-415. [PMID: 30117761 DOI: 10.1089/met.2018.0022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The aims of this study were to 1. define the responses of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), glucagon, and peptide YY (PYY) to an oral meal and to intravenous L-arginine; and 2. examine correlation of enteroendocrine hormones with insulin secretion. We hypothesized a relationship between circulating incretin concentrations and insulin secretion. METHODS Subjects with normal glucose tolerance (NGT, n = 23), prediabetes (PDM, n = 17), or with type 2 diabetes (T2DM, n = 22) were studied twice, following a mixed test meal (470 kCal) (mixed meal tolerance test [MMTT]) or intravenous L-arginine (arginine maximal stimulation test [AST], 5 g). GLP-1 (total and active), PYY, GIP, glucagon, and β cell function were measured before and following each stimulus. RESULTS Baseline enteroendocrine hormones differed across the glucose tolerance (GT) spectrum, T2DM generally >NGT and PDM. In response to MMTT, total and active GLP-1, GIP, glucagon, and PYY increased in all populations. The incremental area-under-the-curve (0-120 min) of analytes like total GLP-1 were often higher in T2DM compared with NGT and PDM (35-51%; P < 0.05). At baseline glucose, L-arginine increased total and active GLP-1 and glucagon concentrations in all GT populations (all P < 0.05). As expected, the MMTT and AST provoked differential glucose, insulin, and C-peptide responses across GT populations. Baseline or stimulated enteroendocrine hormone concentrations did not consistently correlate with either measure of β cell function. CONCLUSIONS/INTERPRETATION Both MMTT and AST resulted in insulin and enteroendocrine hormone responses across GT populations without consistent correlation between release of incretins and insulin, which is in line with other published research. If a defect is in the enteroendocrine/β cell axis, it is probably reduced response to rather than diminished secretion of enteroendocrine hormones.
Collapse
Affiliation(s)
| | | | | | | | - Claudio Cobelli
- 4 Department of Information Engineering, University of Padova , Padova, Italy
| | - Atalanta Ghosh
- 5 Janssen Research and Development , Raritan, New Jersey
| | - R Paul Robertson
- 6 Pacific Northwest Diabetes Institute, University of Washington , Seattle, Washington
| | - Sudha S Shankar
- 7 Lilly Research Laboratories, Lilly Corporate Center , Indianapolis, Indiana
| | | | - Darko Stefanovski
- 9 School of Veterinary Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Adrian Vella
- 10 Division of Endocrinology, Mayo Clinic and Foundation , Rochester, Minnesota
| | - Kathryn Wright
- 11 Wright Biomarker Consulting , Gales Ferry, Connecticut
| | | | | |
Collapse
|
35
|
Farngren J, Persson M, Ahrén B. Effects on the glucagon response to hypoglycaemia during DPP-4 inhibition in elderly subjects with type 2 diabetes: A randomized, placebo-controlled study. Diabetes Obes Metab 2018; 20:1911-1920. [PMID: 29645341 DOI: 10.1111/dom.13316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/28/2018] [Accepted: 04/01/2018] [Indexed: 12/19/2022]
Abstract
AIMS Maintainance of glucagon response to hypoglycaemia is important as a safeguard against hypoglycaemia during glucose-lowering therapy in type 2 diabetes. During recent years, DPP-4 (dipeptidyl peptidase-4) inhibition has become more commonly used in elderly patients. However, whether DPP-4 inhibition affects the glucagon response to hypoglycaemia in the elderly is not known and was the aim of this study. METHODS In a single-centre, double-blind, randomized, placebo-controlled crossover study, 28 subjects with metformin-treated type 2 diabetes (17 male, 11 female; mean age, 74 years [range 65-86]; mean HbA1c, 51.5 mmol/mol [6.9%]) received sitagliptin (100 mg once daily) as add-on therapy or placebo for 4 weeks with a 4-week washout period in between. After each treatment period, the subjects underwent a standard breakfast test, followed by a 2-step hyperinsulinaemic hypoglycaemic clamp (target 3.5 and 3.0 mmol/L), followed by lunch. RESULTS Glucagon levels after breakfast and lunch, and the glucagon response at 3.5 mmol/L, were lower after sitagliptin than after placebo. However, the glucagon response to hypoglycaemia at 3.1 mmol/L did not differ significantly between the two. Similarly, the noradrenaline, adrenaline and cortisol responses were lower with sitagliptin than with placebo at 3.5 mmol/L, but not at 3.1 mmol/L glucose. Responses in pancreatic polypeptide did not differ between the two. CONCLUSIONS Elderly subjects with metformin-treated type 2 diabetes have lower glucagon levels at 3.5 mmol/L glucose, but maintain the glucagon response to hypoglycaemia at 3.1 mmol/L during DPP-4 inhibition, which safeguards against hypoglycaemia and may contribute to decreasing the risk of hypoglycaemia by DPP-4 inhibition in this age group.
Collapse
Affiliation(s)
- Johan Farngren
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Bo Ahrén
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Abstract
Glucagon-like peptide-1 (GLP-1) released from gut enteroendocrine cells controls meal-related glycemic excursions through augmentation of insulin and inhibition of glucagon secretion. GLP-1 also inhibits gastric emptying and food intake, actions maximizing nutrient absorption while limiting weight gain. Here I review the circuits engaged by endogenous versus pharmacological GLP-1 action, highlighting key GLP-1 receptor (GLP-1R)-positive cell types and pathways transducing metabolic and non-glycemic GLP-1 signals. The role(s) of GLP-1 in the benefits and side effects associated with bariatric surgery are discussed and actions of GLP-1 controlling islet function, appetite, inflammation, and cardiovascular pathophysiology are highlighted. Refinement of the risk-versus-benefit profile of GLP-1-based therapies for the treatment of diabetes and obesity has stimulated development of orally bioavailable agonists, allosteric modulators, and unimolecular multi-agonists, all targeting the GLP-1R. This review highlights established and emerging concepts, unanswered questions, and future challenges for development and optimization of GLP-1R agonists in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, 600 University Avenue, Mailbox 39, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
37
|
Mechanistic study for the simultaneous determination of metformin and teneligliptin in human plasma using hydrophilic interaction liquid chromatography–MS/MS. Bioanalysis 2018; 10:475-488. [DOI: 10.4155/bio-2018-0007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: A simple, selective and sensitive hydrophilic interaction liquid chromatography-MS/MS method is developed for the simultaneous determination of metformin (MET) and teneligliptin (TEN) in human plasma using deuterated internal standards. The mechanism of retention of analytes was studied by varying the proportion of organic diluent, buffer strength, pH of the mobile phase and temperature. Results: The results showed a mixed-mode mechanism comprising of hydrophilic (partition) and electrostatic interaction (ion exchange) for MET and essentially hydrophilic for TEN. The linear calibration curves were established in the concentration range of 1.0–1000 ng/ml for MET and 0.50–750 ng/ml for TEN. Conclusion: The method was applied to determine plasma concentration of MET and TEN in healthy subjects.
Collapse
|
38
|
Peng Y, Chang Q, Yang N, Gu S, Zhou Y, Yin L, Aa J, Wang G, Sun J. Quantitative determination of metformin, saxagliptin and 5-hydroxy saxagliptin simultaneously by hydrophilic interaction liquid chromatography - electrospray ionization mass spectrometry and its application to a bioequivalence study with a single-pill combination in human. J Chromatogr B Analyt Technol Biomed Life Sci 2018. [DOI: 10.1016/j.jchromb.2018.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Alsalim W, Göransson O, Carr RD, Bizzotto R, Tura A, Pacini G, Mari A, Ahrén B. Effect of single-dose DPP-4 inhibitor sitagliptin on β-cell function and incretin hormone secretion after meal ingestion in healthy volunteers and drug-naïve, well-controlled type 2 diabetes subjects. Diabetes Obes Metab 2018; 20:1080-1085. [PMID: 29227575 DOI: 10.1111/dom.13192] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/30/2017] [Accepted: 12/07/2017] [Indexed: 12/17/2022]
Abstract
To explore the effects of a single dose of the DPP-4 inhibitor sitagliptin on glucose-standardized insulin secretion and β-cell glucose sensitivity after meal ingestion, 12 healthy and 12 drug-naïve, well-controlled type 2 diabetes (T2D) subjects (mean HbA1c 43 mmol/mol, 6.2%) received sitagliptin (100 mg) or placebo before a meal (525 kcal). β-cell function was measured as the insulin secretory rate at a standardized glucose concentration and the β-cell glucose sensitivity (the slope between glucose and insulin secretory rate). Incretin levels were also monitored. Sitagliptin increased standardized insulin secretion, in both healthy and T2D subjects, compared to placebo, but without increasing β-cell glucose sensitivity. Sitagliptin also increased active glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) and reduced total (reflecting the secretion) GIP, but not total GLP-1 levels. We conclude that a single dose of DPP-4 inhibition induces dissociated effects on different aspects of β-cell function and incretin hormones after meal ingestion in both healthy and well-controlled T2D subjects.
Collapse
Affiliation(s)
- Wathik Alsalim
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Olga Göransson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Richard D Carr
- MSD, Copenhagen, Denmark and University College London, London, UK
| | - Roberto Bizzotto
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Andrea Tura
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Giovanni Pacini
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Bo Ahrén
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
40
|
McKillop AM, Stevenson CL, Moran BM, Abdel-Wahab YHA, Flatt PR. Tissue expression of DPP-IV in obesity-diabetes and modulatory effects on peptide regulation of insulin secretion. Peptides 2018; 100:165-172. [PMID: 29412816 DOI: 10.1016/j.peptides.2017.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 02/01/2023]
Abstract
Dipeptidyl peptidase type 4 (DPP-4) inhibitors represent an important class of glucose-lowering drug for type 2 diabetes. DPP-4 enzyme activity has been observed to be significantly altered in type 2 diabetes. Here, the role of DPP-4 was examined in a high fat fed (HFF) mouse model of insulin resistance. HFF mice had an increased bodyweight (p < .01), were hyperglycaemic (p < .01) and hyperinsulinaemic (p < .05). Compared to normal diet, HFF mice exhibited increased plasma DPP-4 activity (p < .01). Tissue distribution patterns in lean and HFF mice demonstrated highest levels of DPP-4 activity in lung (20-26 μmol/min/mg protein) and small intestine (13-14 μmol/min/mg protein), and lowest activity in the spleen (3.8 μmol/min/mg protein). Modulation of DPP-4 activity by high fat feeding was observed in several tissues with increases in the lung (p < .05), liver (p < .05), kidney (p < .05) and pancreas (p < .05). With a high fat diet, DPP-4 gene expression was upregulated in the liver (p < .001) and downregulated in the pancreas (p < 0.001) and small intestine (p < .001). Immunohistochemical analysis revealed increased DPP-4 immunostaining localised primarily in the pancreatic islets of HFF mice (p < .01) with no change in islet GLP-1 expression. Treatment of HFF mice with metformin for 21-days resulted in inhibition of circulating DPP-4 activity (p < .05), decreased blood glucose (p < .05) and increased GLP-1 gene expression (p < .001). These data indicate that DPP-4 is modulated in a tissue specific manner and is dependent on physiological conditions such as hyperglycaemia and insulin resistance, suggesting a significant role in disorders such as diabetes.
Collapse
Affiliation(s)
- Aine M McKillop
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, United Kingdom.
| | - Claire L Stevenson
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, United Kingdom
| | - Brian M Moran
- Department of Biopharmaceutical and Medical Science, School of Science and Computing, Galway-Mayo Institute of Technology, Galway, Ireland
| | - Yasser H A Abdel-Wahab
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, United Kingdom
| | - Peter R Flatt
- SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, United Kingdom
| |
Collapse
|
41
|
Affiliation(s)
- André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège B-4000, Belgium; and the Division of Clinical Pharmacology, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège B-4000, Belgium
| |
Collapse
|
42
|
Andersen ES, Deacon CF, Holst JJ. Do we know the true mechanism of action of the DPP-4 inhibitors? Diabetes Obes Metab 2018; 20:34-41. [PMID: 28544214 DOI: 10.1111/dom.13018] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/18/2017] [Accepted: 05/21/2017] [Indexed: 12/19/2022]
Abstract
The prevalence of type 2 diabetes is increasing, which is alarming because of its serious complications. Anti-diabetic treatment aims to control glucose homeostasis as tightly as possible in order to reduce these complications. Dipeptidyl peptidase-4 (DPP-4) inhibitors are a recent addition to the anti-diabetic treatment modalities, and have become widely accepted because of their good efficacy, their benign side-effect profile and their low hypoglycaemia risk. The actions of DPP-4 inhibitors are not direct, but rather are mediated indirectly through preservation of the substrates they protect from degradation. The two incretin hormones, glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide, are known substrates, but other incretin-independent mechanisms may also be involved. It seems likely therefore that the mechanisms of action of DPP-4 inhibitors are more complex than originally thought, and may involve several substrates and encompass local paracrine, systemic endocrine and neural pathways, which are discussed here.
Collapse
Affiliation(s)
- Emilie S Andersen
- Department of Internal Medicine F, Hospital Gentofte, Copenhagen University, Copenhagen, Denmark
- Department of Biomedical Sciences, NNF Center of Basic Metabolic Research, The Panum Institute, Copenhagen University, Copenhagen, Denmark
| | - Carolyn F Deacon
- Department of Biomedical Sciences, NNF Center of Basic Metabolic Research, The Panum Institute, Copenhagen University, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, NNF Center of Basic Metabolic Research, The Panum Institute, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
43
|
Trifluorophenyl-based inhibitors of dipeptidyl peptidase-IV as antidiabetic agents: 3D-QSAR COMFA, CoMSIA methodologies. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s13721-017-0163-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Brønden A, Albér A, Rohde U, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. Single-Dose Metformin Enhances Bile Acid-Induced Glucagon-Like Peptide-1 Secretion in Patients With Type 2 Diabetes. J Clin Endocrinol Metab 2017; 102:4153-4162. [PMID: 28938439 DOI: 10.1210/jc.2017-01091] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022]
Abstract
CONTEXT Despite a position as the first-line pharmacotherapy in type 2 diabetes, the glucose-lowering mechanisms of metformin remain to be fully clarified. Gut-derived modes of action, including suppression of bile acid reabsorption and a resulting increase in glucagon-like peptide-1 (GLP-1) secretion, have been proposed. OBJECTIVE The aim of this study was to assess the GLP-1 secretory and glucometabolic effects of endogenously released bile, with and without concomitant single-dose administration of metformin in patients with type 2 diabetes. DESIGN Randomized, placebo-controlled, and double-blinded crossover study. SETTING This study was conducted at Center for Diabetes Research, Gentofte Hospital, Denmark. PATIENTS Fifteen metformin-treated patients with type 2 diabetes; all participants completed the study. INTERVENTIONS Four experimental study days in randomized order with administration of either 1500 mg metformin or placebo in combination with intravenous infusion of cholecystokinin (0.4 pmol × kg-1 × min-1) or saline. MAIN OUTCOME MEASURE Plasma GLP-1 excursions as measured by baseline-subtracted area under the curve. RESULTS Single-dose metformin further enhanced bile acid-mediated induction of GLP-1 secretion (P = 0.02), whereas metformin alone did not increase plasma GLP-1 concentrations compared with placebo (P = 0.17). Metformin, both with (P = 0.02) and without (P = 0.02) concomitant cholecystokinin-induced gallbladder emptying, elicited reduced plasma glucose excursions compared with placebo. No GLP-1-mediated induction of insulin secretion or suppression of glucagon was observed. CONCLUSIONS Metformin elicited an enhancement of the GLP-1 response to cholecystokinin-induced gallbladder emptying in patients with type 2 diabetes, whereas no derived effects on insulin or glucagon secretion were evident in this acute setting.
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Anders Albér
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Ulrich Rohde
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, 2820 Gentofte, Denmark
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
45
|
Liao WL, Lee WJ, Chen CC, Lu CH, Chen CH, Chou YC, Lee IT, Sheu WHH, Wu JY, Yang CF, Wang CH, Tsai FJ. Pharmacogenetics of dipeptidyl peptidase 4 inhibitors in a Taiwanese population with type 2 diabetes. Oncotarget 2017; 8:18050-18058. [PMID: 28160554 PMCID: PMC5392306 DOI: 10.18632/oncotarget.14951] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are oral anti-hyperglycemic drugs enabling effective glycemic control in type 2 diabetes (T2D). Despite DPP-4 inhibitors' advantages, the patients' therapeutic response varies. In this retrospective cohort study, 171 Taiwanese patients with T2D were classified as sensitive or resistant to treatment based on the mean change in HbA1c levels. Using an assumption-free genome-wide association study, 45 single nucleotide polymorphisms (SNPs) involved in the therapeutic response to DPP-4 inhibitors (P < 1 × 10-4) were identified at or near PRKD1, CNTN3, ASK, and LOC10537792. A SNP located within the fourth intron of PRKD1 (rs57803087) was strongly associated with DPP-4 inhibitor response (P = 3.2 × 10-6). This is the first pharmacogenomics study on DPP-4 inhibitor treatment for diabetes in a Taiwanese population. Our data suggest that genes associated with β-cell function and apoptosis are involved in the therapeutic effect of DPP-4 inhibitors, even in the presence of additional oral anti-diabetic drugs. Our findings provide information on how genetic variants influence drug response and may benefit the development of personalized medicine.
Collapse
Affiliation(s)
- Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.,Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chieh Hsiang Lu
- Department of Internal Medicine, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi City, Taiwan.,Department of Nursing, College of Nursing and Health Sciences, DAYEH University, Taiwan.,Department of Business management, College of Management, National Chung Cheng University, Taiwan
| | - Chien-Hsiun Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Chou
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans Hospital, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Defense Medical Center, Taipei, Taiwan.,Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | - Jer-Yuarn Wu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Fan Yang
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chung-Hsing Wang
- Department of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
46
|
Sonoda J, Chen MZ, Baruch A. FGF21-receptor agonists: an emerging therapeutic class for obesity-related diseases. Horm Mol Biol Clin Investig 2017; 30:/j/hmbci.ahead-of-print/hmbci-2017-0002/hmbci-2017-0002.xml. [PMID: 28525362 DOI: 10.1515/hmbci-2017-0002] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 02/13/2017] [Indexed: 01/10/2023]
Abstract
Fibroblast growth factor 21 (FGF21) analogs and FGF21 receptor agonists (FGF21RAs) that mimic FGF21 ligand activity constitute the new "FGF21-class" of anti-obesity and anti-diabetic molecules that improve insulin sensitivity, ameliorate hepatosteatosis and promote weight loss. The metabolic actions of FGF21-class proteins in obese mice are attributed to stimulation of brown fat thermogenesis and increased secretion of adiponectin. The therapeutic utility of this class of molecules is being actively investigated in clinical trials for the treatment of type 2 diabetes and non-alcoholic steatohepatitis (NASH). This review is focused on various FGF21-class molecules, their molecular designs and the preclinical and clinical activities. These molecules include modified FGF21 as well as agonistic antibodies against the receptor for FGF21, namely the complex of FGF receptor 1 (FGFR1) and the obligatory coreceptor βKlotho (KLB). In addition, a novel approach to increase endogenous FGF21 activity by inhibiting the FGF21-degrading protease fibroblast activation protein (FAP) is discussed.
Collapse
|
47
|
Pan C, Han P, Ji Q, Li C, Lu J, Yang J, Li W, Zeng J, Hsieh AT, Chan J. Efficacy and safety of alogliptin in patients with type 2 diabetes mellitus: A multicentre randomized double-blind placebo-controlled Phase 3 study in mainland China, Taiwan, and Hong Kong. J Diabetes 2017; 9:386-395. [PMID: 27171508 DOI: 10.1111/1753-0407.12425] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/18/2016] [Accepted: 05/06/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND This study determined the efficacy and safety of once-daily oral alogliptin in patients from mainland China, Taiwan, and Hong Kong with type 2 diabetes mellitus. METHODS In this Phase 3 multicenter double-blind placebo-controlled 16-week trial, 506 patients were randomized to receive once-daily alogliptin 25 mg or placebo: 185 in the monotherapy group, 197 in the add-on to metformin group, and 124 in the add-on to pioglitazone group. The primary efficacy variable was the change from baseline (CFB) in HbA1c at Week 16; other efficacy measures included CFB to Week 16 in fasting plasma glucose (FPG), incidence of marked hyperglycemia (FPG ≥11.1 mmol/L), and the incidence of clinical HbA1c ≤6.5 % (48 mmol/mol) and ≤7.0 % (53 mmol/mol) at Week 16. Safety was assessed throughout the trial. RESULTS Alogliptin monotherapy provided a significantly greater decrease in HbA1c from baseline to Week 16 compared with placebo (-0.58 %; 95 % confidence interval [CI] -0.78 %, -0.37 %; P < 0.001). As an add-on to metformin or pioglitazone, alogliptin also significantly decreased HbA1c compared with placebo (-0.69 % [95 % CI -0.87 %, -0.51 %; P < 0.001] and -0.52 % [95 % CI -0.75 %, -0.28 %; P < 0.001], respectively). In any treatment group versus placebo, alogliptin led to greater decreases in FPG (P ≤ 0.004) and a higher percentage of patients who achieved an HbA1c target of ≤6.5 % and ≤7.0 % (P ≤ 0.003). No weight gain was observed in any treatment group. A similar percentage of patients experienced drug-related, treatment-emergent adverse events in the alogliptin and placebo arms. Four and two patients in the alogliptin and placebo arms, respectively, experienced mild or moderate hypoglycemia. CONCLUSIONS Alogliptin 25 mg once daily reduced HbA1c and FPG and enhanced clinical response compared with placebo when used as monotherapy or as an add-on to metformin or pioglitazone. Therapy with alogliptin was well tolerated.
Collapse
Affiliation(s)
- Changyu Pan
- The General Hospital of the People's Liberation Army, Beijing, China
| | - Ping Han
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiuhe Ji
- The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Chengjiang Li
- First Affiliated Hospital Zhejiang University College of Medicine, Hangzhou, China
| | - Juming Lu
- The General Hospital of the People's Liberation Army, Beijing, China
| | | | - Wenhui Li
- Peking Union Medical College Hospital, Beijing, China
| | - Jiaoe Zeng
- Affiliated Jingzhou Hospital Tongji Medical School, Wuhan, China
| | - An-Tsz Hsieh
- Taipei Medical University Shuang Ho Hospital, Taipei, Taiwan
| | - Juliana Chan
- The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, SAR, China
| |
Collapse
|
48
|
Preiss D, Dawed A, Welsh P, Heggie A, Jones AG, Dekker J, Koivula R, Hansen TH, Stewart C, Holman RR, Franks PW, Walker M, Pearson ER, Sattar N. Sustained influence of metformin therapy on circulating glucagon-like peptide-1 levels in individuals with and without type 2 diabetes. Diabetes Obes Metab 2017; 19:356-363. [PMID: 27862873 PMCID: PMC5330429 DOI: 10.1111/dom.12826] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/31/2016] [Accepted: 11/08/2016] [Indexed: 12/20/2022]
Abstract
AIMS To investigate, in the Carotid Atherosclerosis: Metformin for Insulin Resistance (CAMERA) trial (NCT00723307), whether the influence of metformin on the glucagon-like peptide (GLP)-1 axis in individuals with and without type 2 diabetes (T2DM) is sustained and related to changes in glycaemia or weight, and to investigate basal and post-meal GLP-1 levels in patients with T2DM in the cross-sectional Diabetes Research on Patient Stratification (DIRECT) study. MATERIALS AND METHODS CAMERA was a double-blind randomized placebo-controlled trial of metformin in 173 participants without diabetes. Using 6-monthly fasted total GLP-1 levels over 18 months, we evaluated metformin's effect on total GLP-1 with repeated-measures analysis and analysis of covariance. In the DIRECT study, we examined active and total fasting and 60-minute post-meal GLP-1 levels in 775 people recently diagnosed with T2DM treated with metformin or diet, using Student's t-tests and linear regression. RESULTS In CAMERA, metformin increased total GLP-1 at 6 (+20.7%, 95% confidence interval [CI] 4.7-39.0), 12 (+26.7%, 95% CI 10.3-45.6) and 18 months (+18.7%, 95% CI 3.8-35.7), an overall increase of 23.4% (95% CI 11.2-36.9; P < .0001) vs placebo. Adjustment for changes in glycaemia and adiposity, individually or combined, did not attenuate this effect. In the DIRECT study, metformin was associated with higher fasting active (39.1%, 95% CI 21.3-56.4) and total GLP-1 (14.1%, 95% CI 1.2-25.9) but not post-meal incremental GLP-1. These changes were independent of potential confounders including age, sex, adiposity and glycated haemoglobin. CONCLUSIONS In people without diabetes, metformin increases total GLP-1 in a sustained manner and independently of changes in weight or glycaemia. Metformin-treated patients with T2DM also have higher fasted GLP-1 levels, independently of weight and glycaemia.
Collapse
Affiliation(s)
- David Preiss
- MRC Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit, University of Oxford, Oxford, UK
| | - Adem Dawed
- Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Alison Heggie
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Angus G Jones
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, UK
| | - Jacqueline Dekker
- VU Medical Center, Dept Epidemiology and Biostatistics, Amsterdam, Netherlands
| | - Robert Koivula
- Department of Clinical Sciences, Lund University, Genetic and Molecular Epidemiology, Malmö, Sweden
| | - Tue H Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | - Caitlin Stewart
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Rury R Holman
- Diabetes Trials Unit, University of Oxford, Oxford, UK
| | - Paul W Franks
- Department of Clinical Sciences, Lund University, Genetic and Molecular Epidemiology, Malmö, Sweden
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ewan R Pearson
- Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
49
|
Cahn A, Cernea S, Raz I. An update on DPP-4 inhibitors in the management of type 2 diabetes. Expert Opin Emerg Drugs 2016; 21:409-419. [PMID: 27809608 DOI: 10.1080/14728214.2016.1257608] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION DPP-4 inhibitors are a class of compounds used for the treatment of type 2 diabetes. The drugs inhibit the degradation of GLP-1, thus amplifying the incretin effect. They have moderate glycemic efficacy, a low propensity of causing hypoglycaemia and are weight neutral. The drugs are often used as second line therapy after metformin. Areas covered: This review summarizes the available compounds in the market and discusses the novel compounds that are currently under development. Several large cardiovascular outcome trials with some of the compounds have been completed, and their results and implications are considered. Fixed dose combination pills are currently the main focus of research and the contribution of these to the care of patients with diabetes is further discussed. Expert opinion: The DPP-4 inhibitors have been a successful class in drug development for diabetes. Taken orally and available as fixed dose combinations with metformin or with SGLT-2 inhibitors, they have reached a large market share of over 7 billion dollars. Other than retagliptin, it does not appear that any additional compound will be launched soon. Currently, the main focus is on the development of additional fixed dose combinations with SGLT-2 inhibitors, but the success of these combinations remains to be seen.
Collapse
Affiliation(s)
- Avivit Cahn
- a The Diabetes Research Unit, Internal Medicine Section , Hadassah Hebrew University Hospital , Jerusalem , Israel.,b Endocrinology and Metabolism Unit, Internal Medicine Section , Hadassah Hebrew University Hospital , Jerusalem , Israel
| | - Simona Cernea
- c Department M3/Internal Medicine IV , University of Medicine and Pharmacy , Târgu Mureş , Romania.,d Diabetes, Nutrition and Metabolic Diseases Unit , Emergency County Clinical Hospital , Târgu Mureş , Romania
| | - Itamar Raz
- a The Diabetes Research Unit, Internal Medicine Section , Hadassah Hebrew University Hospital , Jerusalem , Israel
| |
Collapse
|
50
|
Baranov O, Kahle M, Deacon CF, Holst JJ, Nauck MA. Feedback suppression of meal-induced glucagon-like peptide-1 (GLP-1) secretion mediated through elevations in intact GLP-1 caused by dipeptidyl peptidase-4 inhibition: a randomized, prospective comparison of sitagliptin and vildagliptin treatment. Diabetes Obes Metab 2016; 18:1100-1109. [PMID: 27300579 DOI: 10.1111/dom.12706] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
AIM To compare directly the clinical effects of vildagliptin and sitagliptin in patients with type 2 diabetes, with a special emphasis on incretin hormones and L-cell feedback inhibition induced by dipeptidyl peptidase (DPP-4) inhibition. METHODS A total of 24 patients (12 on a diet/exercise regimen, 12 on metformin) were treated, in randomized order, for 7-9 days, with either vildagliptin (50 mg twice daily = 100 mg/d), sitagliptin (100 mg once daily in those on diet, 50 mg twice daily in those on metformin treatment = 100 mg/d) or placebo (twice daily). A mixed-meal test was performed. RESULTS Intact glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide concentrations were doubled by both DPP-4 inhibitors. Meal-related total GLP-1 responses were reduced by vildagliptin and sitagliptin treatment alike in the majority of patients (vildagliptin: p = 0.0005; sitagliptin: p = 0.019), but with substantial inter-individual variation. L-cell feedback appeared to be more pronounced in those whose intact GLP-1 relative to total GLP-1 increased more, and who had greater reductions in fasting plasma glucose after DPP-4 inhibition. K-cell feedback inhibition overall was not significant. There were no differences in any of the clinical variables (glycaemia, insulin and glucagon secretory responses) between vildagliptin and sitagliptin treatment. CONCLUSIONS Vildagliptin and sitagliptin affected incretin hormones, glucose concentrations, insulin and glucagon secretion in a similar manner. Inter-individual variations in L-cell feedback inhibition may indicate heterogeneity in the clinical response to DPP-4 inhibition.
Collapse
Affiliation(s)
- Oleg Baranov
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany
| | - Melanie Kahle
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany
- Division of Diabetology, Medical Department I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Carolyn F Deacon
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michael A Nauck
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany.
- Division of Diabetology, Medical Department I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|