1
|
Ji L, Song W, Fang H, Li W, Geng J, Wang Y, Guo L, Cai H, Yang T, Li H, Yang G, Li Q, Liu K, Li S, Liu Y, Shi F, Li X, Gao X, Tian H, Ji Q, Su Q, Zhou Z, Wang W, Zhou Z, Li X, Xu Y, Ning Z, Cao H, Pan D, Yao H, Lu X, Jia W. Efficacy and safety of chiglitazar, a novel peroxisome proliferator-activated receptor pan-agonist, in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, phase 3 trial (CMAP). Sci Bull (Beijing) 2021; 66:1571-1580. [PMID: 36654286 DOI: 10.1016/j.scib.2021.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/21/2020] [Accepted: 03/09/2021] [Indexed: 02/03/2023]
Abstract
Chiglitazar (Carfloglitazar) is a novel non-thiazolidinedione (TZD) structured peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes in previous clinical studies. This randomized phase 3 trial aimed to compare the efficacy and safety of chiglitazar with placebo in patients with type 2 diabetes with insufficient glycemic control by strict diet and exercise alone. Eligible patients were randomly assigned to receive chiglitazar 32 mg (n = 167), chiglitazar 48 mg (n = 166), or placebo (n = 202) once daily. The primary endpoint was the change in glycosylated hemoglobin A1c (HbA1c) at week 24 with superiority of chiglitazar over placebo. The results showed that both chiglitazar 32 and 48 mg resulted in significant and clinically meaningful reductions in HbA1c, and placebo-adjusted estimated treatment differences at week 24 for chiglitazar 32 and 48 mg were -0.87% (95% confidential interval (CI): -1.10 to -0.65; P < 0.0001) and -1.05% (95% CI: -1.29 to -0.81; P < 0.0001), respectively. Secondary efficacy parameters including glycemic control, insulin sensitivity and triglyceride reduction were also significantly improved in the chiglitazar groups. The overall frequency of adverse events and study discontinuation attributable to adverse events were similar among the groups. Low incidences of mild edema and body weight gain were reported in the chiglitazar dose groups. The results from this phase 3 trial demonstrated that the PPAR pan-agonist chiglitazar possesses an overall good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.
Collapse
Affiliation(s)
- Linong Ji
- Peking University People's Hospital, Beijing 100044, China.
| | - Weihong Song
- Chenzhou No.1 People's Hospital, Chenzhou 423000, China
| | - Hui Fang
- Tangshan Gongren Hospital, Tangshan 063000, China
| | - Wei Li
- The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Jianlin Geng
- Harrison International Peace Hospital, Hengshui 053000, China
| | - Yangang Wang
- The Affiliate Hospital of Qingdao University, Qingdao 266003, China
| | - Lian Guo
- Chongqing Three Gorges Central Hospital, Chongqing 404000, China
| | - Hanqing Cai
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Tao Yang
- Jiangsu Province Hospital, Nanjing 210029, China
| | - Hongmei Li
- China Meitan General Hospital, Beijing 100028, China
| | - Gangyi Yang
- The Second Affiliate Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qifu Li
- The First Affiliate Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kuanzhi Liu
- The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Shuying Li
- Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanjun Liu
- The 306th Hospital of PLA, Beijing 100101, China
| | - Fuyan Shi
- Baogang Hospital of Inner Mongolia, Baotou 014010, China
| | - Xinsheng Li
- Cangzhou's Central Hospital, Cangzhou 031706, China
| | - Xin Gao
- Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Haoming Tian
- Huaxi Hopsital of Sichuan University, Chengdu 610041, China
| | - Qiuhe Ji
- The First Affiliated Hospital of The 4th Military Medical University, Xi'an 710000, China
| | - Qing Su
- Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhiguang Zhou
- The Second Xiangya Hospital of Central South University, Changsha 410008, China
| | - Wenbo Wang
- Peking University Shougang Hospital, Beijing 100144, China
| | - Zunhai Zhou
- The Central Hospital of Yangpu District of Shanghai, Shanghai 200090, China
| | - Xuejun Li
- The First Affiliate Hospital of Xiamen University, Xiamen 361003, China
| | - Yancheng Xu
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhiqiang Ning
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Haixiang Cao
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - He Yao
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Xianping Lu
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen 518057, China
| | - Weiping Jia
- Shanghai 6th People's Hospital, Shanghai 200233, China.
| |
Collapse
|
2
|
Jia W, Ma J, Miao H, Wang C, Wang X, Li Q, Lu W, Yang J, Zhang L, Yang J, Wang G, Zhang X, Zhang M, Sun L, Yu X, Du J, Shi B, Xiao C, Zhu D, Liu H, Zhong L, Xu C, Xu Q, Liang G, Zhang Y, Li G, Gu M, Liu J, Yuan G, Yan Z, Yan D, Ye S, Zhang F, Ning Z, Cao H, Pan D, Yao H, Lu X, Ji L. Chiglitazar monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomized, double-blind, phase 3 trial (CMAS). Sci Bull (Beijing) 2021; 66:1581-1590. [PMID: 36654287 DOI: 10.1016/j.scib.2021.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 09/13/2020] [Accepted: 02/05/2021] [Indexed: 02/03/2023]
Abstract
Chiglitazar (Carfloglitazar) is a novel peroxisome proliferator-activated receptor (PPAR) pan-agonist that has shown promising effects on glycemic control and lipid regulation in patients with type 2 diabetes. In this randomized phase 3 trial, we compared the efficacy and safety of chiglitazar with sitagliptin in patients with type 2 diabetes who had insufficient glycemic control despite a strict diet and exercise regimen. Eligible patients were randomized (1:1:1) to receive chiglitazar 32 mg (n = 245), chiglitazar 48 mg (n = 246), or sitagliptin 100 mg (n = 248) once daily for 24 weeks. The primary endpoint was the change in glycosylated hemoglobin A1C (HbA1c) from baseline at week 24 with the non-inferiority of chiglitazar over sitagliptin. Both chiglitazar and sitagliptin significantly reduced HbA1c at week 24 with values of -1.40%, -1.47%, and -1.39% for chiglitazar 32 mg, chiglitazar 48 mg, and sitagliptin 100 mg, respectively. Chiglitazar 32 and 48 mg were both non-inferior to sitagliptin 100 mg, with mean differences of -0.04% (95% confidential interval (CI) -0.22 to 0.15) and -0.08% (95% CI -0.27 to 0.10), respectively. Compared with sitagliptin, greater reduction in fasting and 2-h postprandial plasma glucose and fasting insulin was observed with chiglitazar. Overall adverse event rates were similar between the groups. A small increase in mild edema in the chiglitazar 48 mg group and slight weight gain in both chiglitazar groups were reported. The overall results demonstrated that chiglitazar possesses good efficacy and safety profile in patients with type 2 diabetes inadequately controlled with lifestyle interventions, thereby providing adequate supporting evidence for using this PPAR pan-agonist as a treatment option for type 2 diabetes.
Collapse
Affiliation(s)
- Weiping Jia
- Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China.
| | - Jianhua Ma
- Nanjing First Hospital, Nanjing 210029, China
| | - Heng Miao
- The Second Hospital Affiliated to Nanjing Medical University, Nanjing 210011, China
| | - Changjiang Wang
- The First Hospital Affiliated to Anhui Medical University, Hefei 230031, China
| | - Xiaoyue Wang
- The First People's Hospital of Yueyang, Yueyang 414000, China
| | - Quanmin Li
- PLA Rocket Force Characteristic Medical Center, Beijing 100085, China
| | - Weiping Lu
- Huai'an First People's Hospital, Huai'an 223300, China
| | - Jialin Yang
- The Central Hospital of Minhang District of Shanghai, Shanghai 201100, China
| | - Lihui Zhang
- The Second Hospital of Heibei Medical University, Shijiazhuang 050000, China
| | - Jinkui Yang
- Beijing Tongren Hospital Affiliated to Capital Medical University, Beijing 100730, China
| | - Guixia Wang
- The First Hospital of Jilin University, Changchun 130021, China
| | - Xiuzhen Zhang
- Tongji Hospital of Tongji University, Shanghai 200092, China
| | - Min Zhang
- The Qingpu Branch of Zhongshan Hospital Affiliate to Fudan University, Shanghai 201700, China
| | - Li Sun
- Siping Central People's Hospital, Siping 136000, China
| | - Xuefeng Yu
- Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jianling Du
- The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Bingyin Shi
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Changqing Xiao
- The First Affiliated Hospital of Guangxi Medical University (The Western Hospital), Nanning 530021, China
| | - Dalong Zhu
- Gulou Hospital Affiliated to Nanjing Medical University, Nanjing 210008, China
| | - Hong Liu
- The First Affiliated Hospital of Guangxi Medical University (The Eastern Hospital), Nanning 530021, China
| | - Liyong Zhong
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Chun Xu
- The General Hospital of the Chinese People's Armed Police Forces, Beijing 100022, China
| | - Qi Xu
- The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | | | - Ying Zhang
- The Third Hospital Affiliated to Guangzhou Medical College, Guangzhou 510150, China
| | | | - Mingyu Gu
- Shanghai First People's Hospital, Shanghai 200080, China
| | - Jun Liu
- Shanghai 5th People's Hospital, Shanghai 200040, China
| | - Guoyue Yuan
- The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Zhaoli Yan
- The Affiliated Hospital of Inner Mongolia, Hohhot 000306, China
| | - Dewen Yan
- Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Shandong Ye
- Anhui Provincial Hospital, Hefei 518035, China
| | - Fan Zhang
- Beijing University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhiqiang Ning
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Haixiang Cao
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - He Yao
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Xianping Lu
- Shenzhen Chipscreen Biosciences, Ltd., Shenzhen 518057, China
| | - Linong Ji
- Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
3
|
To Probe Full and Partial Activation of Human Peroxisome Proliferator-Activated Receptors by Pan-Agonist Chiglitazar Using Molecular Dynamics Simulations. PPAR Res 2020; 2020:5314187. [PMID: 32308671 PMCID: PMC7152983 DOI: 10.1155/2020/5314187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023] Open
Abstract
Chiglitazar is a promising new-generation insulin sensitizer with low reverse effects for the treatment of type II diabetes mellitus (T2DM) and has shown activity as a nonselective pan-agonist to the human peroxisome proliferator-activated receptors (PPARs) (i.e., full activation of PPARγ and a partial activation of PPARα and PPARβ/δ). Yet, it has no high-resolution complex structure with PPARs and its detailed interactions and activation mechanism remain unclear. In this study, we docked chiglitazar into three experimentally resolved crystal structures of hPPAR subtypes, PPARα, PPARβ/δ, and PPARγ, followed by 3 μs molecular dynamics simulations for each system. Our MM-GBSA binding energy calculation revealed that chiglitazar most favorably bound to hPPARγ (-144.6 kcal/mol), followed by hPPARα (-138.0 kcal/mol) and hPPARβ (-135.9 kcal/mol), and the order is consistent with the experimental data. Through the decomposition of the MM-GBSA binding energy by residue and the use of two-dimensional interaction diagrams, key residues involved in the binding of chiglitazar were identified and characterized for each complex system. Additionally, our detailed dynamics analyses support that the conformation and dynamics of helix 12 play a critical role in determining the activities of the different types of ligands (e.g., full agonist vs. partial agonist). Rather than being bent fully in the direction of the agonist versus antagonist conformation, a partial agonist can adopt a more linear conformation and have a lower degree of flexibility. Our finding may aid in further development of this new generation of medication.
Collapse
|
4
|
Balakumar P, Mahadevan N, Sambathkumar R. A Contemporary Overview of PPARα/γ Dual Agonists for the Management of Diabetic Dyslipidemia. Curr Mol Pharmacol 2020; 12:195-201. [PMID: 30636619 PMCID: PMC6875865 DOI: 10.2174/1874467212666190111165015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 01/11/2023]
Abstract
Background: Diabetes mellitus and concomitant dyslipidemia, being referred to as ‘diabetic dyslipidemia’, are the foremost detrimental factors documented to play a pivotal role in cardiovascular illness. Diabetic dyslipidemia is associated with insulin resistance, high plasma triglyceride levels, low HDL-cholesterol concentration and elevated small dense LDL-cholesterol particles. Maintaining an optimal glucose and lipid levels in patients afflicted with diabetic dyslipidemia could be a major task that might require a well-planned diet-management system and regular physical activity, or otherwise an intake of combined antidiabetic and antihyperlipidemic medications. Synchronized treatment which efficiently controls insulin resistance-associated diabetes mellitus and co-existing dyslipidemia could indeed be a fascinating therapeutic option in the management of diabetic dyslipidemia. Peroxisome proliferator-activated receptors α/γ (PPARα/γ) dual agonists are such kind of drugs which possess therapeutic potentials to treat diabetic dyslipidemia. Nevertheless, PPARα/γ dual agonists like muraglitazar, naveglitazar, tesaglitazar, ragaglitazar and aleglitazar have been reported to have undesirable adverse effects, and their developments have been halted at various stages. On the other hand, a recently introduced PPARα/γ dual agonist, saroglitazar is an emerging therapeutic agent of glitazar class approved in India for the management of diabetic dyslipidemia, and its treatment has been reported to be generally safe and well tolerated. Conclusion: Some additional and new compounds, at initial and preclinical stages, have been recently reported to possess PPARα/γ dual agonistic potentials with considerable therapeutic efficacy and reduced adverse profile. This review sheds light on the current status of various PPARα/γ dual agonists for the management of diabetic dyslipidemia.
Collapse
Affiliation(s)
| | - Nanjaian Mahadevan
- College of Pharmacy, King Khalid University, Guraiger, Abha 62529, Saudi Arabia
| | | |
Collapse
|
5
|
Bell DA, Watts GF. Contemporary and Novel Therapeutic Options for Hypertriglyceridemia. Clin Ther 2015; 37:2732-50. [DOI: 10.1016/j.clinthera.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/16/2022]
|
6
|
Peroxisome Proliferator-Activated Receptors and the Heart: Lessons from the Past and Future Directions. PPAR Res 2015; 2015:271983. [PMID: 26587015 PMCID: PMC4637490 DOI: 10.1155/2015/271983] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear family of ligand activated transcriptional factors and comprise three different isoforms, PPAR-α, PPAR-β/δ, and PPAR-γ. The main role of PPARs is to regulate the expression of genes involved in lipid and glucose metabolism. Several studies have demonstrated that PPAR agonists improve dyslipidemia and glucose control in animals, supporting their potential as a promising therapeutic option to treat diabetes and dyslipidemia. However, substantial differences exist in the therapeutic or adverse effects of specific drug candidates, and clinical studies have yielded inconsistent data on their cardioprotective effects. This review summarizes the current knowledge regarding the molecular function of PPARs and the mechanisms of the PPAR regulation by posttranslational modification in the heart. We also describe the results and lessons learned from important clinical trials on PPAR agonists and discuss the potential future directions for this class of drugs.
Collapse
|
7
|
Della-Morte D, Palmirotta R, Rehni AK, Pastore D, Capuani B, Pacifici F, De Marchis ML, Dave KR, Bellia A, Fogliame G, Ferroni P, Donadel G, Cacciatore F, Abete P, Dong C, Pileggi A, Roselli M, Ricordi C, Sbraccia P, Guadagni F, Rundek T, Lauro D. Pharmacogenomics and pharmacogenetics of thiazolidinediones: role in diabetes and cardiovascular risk factors. Pharmacogenomics 2015; 15:2063-82. [PMID: 25521362 DOI: 10.2217/pgs.14.162] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The most important goal in the treatment of patients with diabetes is to prevent the risk of cardiovascular disease (CVD), the first cause of mortality in these subjects. Thiazolidinediones (TZDs), a class of antidiabetic drugs, act as insulin sensitizers increasing insulin-dependent glucose disposal and reducing hepatic glucose output. TZDs including pioglitazone, rosiglitazone and troglitazone, by activating PPAR-γ have shown pleiotropic effects in reducing vascular risk factors and atherosclerosis. However, troglitazone was removed from the market due to its hepatoxicity, and rosiglitazone and pioglitazone both have particular warnings due to being associated with heart diseases. Specific genetic variations in genes involved in the pathways regulated by TDZs have demonstrated to modify the variability in treatment with these drugs, especially in their side effects. Therefore, pharmacogenomics and pharmacogenetics are an important tool in further understand intersubject variability per se but also to assess the therapeutic potential of such variability in drug individualization and therapeutic optimization.
Collapse
Affiliation(s)
- David Della-Morte
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Altaf QA, Barnett AH, Tahrani AA. Novel therapeutics for type 2 diabetes: insulin resistance. Diabetes Obes Metab 2015; 17:319-34. [PMID: 25308775 DOI: 10.1111/dom.12400] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/23/2014] [Accepted: 10/04/2014] [Indexed: 12/19/2022]
Abstract
Insulin resistance (IR) plays an important role in the pathogenesis of type 2 diabetes (T2D) and cardiovascular disease. Hence improving IR is a major target of treatment in patients with T2D. Obesity and lack of exercise are major causes of IR. However, recent evidence implicates sleep disorders and disorders of the circadian rhythm in the pathogenesis of IR. Weight loss and lifestyle changes are the cornerstone and most effective treatments of IR, but adherence and patient's acceptability are poor. Bariatric surgery results in significant and sustainable long-term weight loss associated with beneficial impact on IR and glucose metabolism, making this an attractive treatment option for patients with T2D. Currently available pharmacological options targeting IR (such as metformin and thiazolidinediones) do not maintain glycaemic measures within targets long term and can be associated with significant side effects. Over the last two decades, many pharmacological agents targeting different aspects of the insulin signalling pathway were developed to improve IR, but only a minority reached clinical trials. Such treatments need to be specific and reversible as many of the components of the insulin signalling pathway are involved in other cellular functions such as apoptosis. Recent evidence highlighted the role of circadian rhythm and sleep-related disorders in the pathogenesis of IR. In this article, we review the latest developments in the pharmacological and non-pharmacological interventions targeting IR including bariatric surgery. We will also review the role of circadian rhythm and sleep-related disorders in the development and treatment of IR.
Collapse
Affiliation(s)
- Q-A Altaf
- Department of Diabetes and Endocrinology, Heart of England NHS Foundation Trust, Birmingham, UK; Centre of Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
9
|
Mansour M. The Roles of Peroxisome Proliferator-Activated Receptors in the Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:217-66. [DOI: 10.1016/b978-0-12-800101-1.00007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
10
|
Paukkeri EL, Leppänen T, Lindholm M, Yam MF, Asmawi MZ, Kolmonen A, Aulaskari PH, Moilanen E. Anti-inflammatory properties of a dual PPARgamma/alpha agonist muraglitazar in in vitro and in vivo models. Arthritis Res Ther 2013; 15:R51. [PMID: 23594962 PMCID: PMC4060226 DOI: 10.1186/ar4211] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 04/09/2013] [Indexed: 12/12/2022] Open
Abstract
Introduction Peroxisome proliferator-activated receptor (PPAR) agonists are widely used drugs in the treatment of diabetes and dyslipidemia. In addition to their metabolic effects, PPAR isoforms PPARα and PPARγ are also involved in the regulation of immune responses and inflammation. In the present study, we investigated the effects of a dual PPARγ/α agonist muraglitazar on inflammatory gene expression in activated macrophages and on carrageenan-induced inflammation in the mouse. Methods J774 murine macrophages were activated by lipopolysaccharide (LPS) and treated with dual PPARγ/α agonist muraglitazar, PPARγ agonist GW1929 or PPARα agonist fenofibrate. The effects of PPAR agonists on cytokine production and the activation of inducible nitric oxide synthase (iNOS) pathway were investigated by ELISA, Griess method, Western blotting and quantitative RT-PCR. Nuclear translocation, DNA-binding activity and reporter gene assays were used to assess the activity of nuclear factor kappa B (NF-kB) transcription factor. Carrageenan-induced paw oedema was used as an in vivo model of acute inflammation. Results Muraglitazar as well as PPARγ agonist GW1929 and PPARα agonist fenofibrate inhibited LPS-induced iNOS expression and NO production in activated macrophages in a dose-dependent manner. Inhibition of iNOS expression by muraglitazar included both transcriptional and post-transcriptional components; the former being shared by GW1929 and the latter by fenofibrate. All tested PPAR agonists also inhibited IL-6 production, while TNFα production was reduced by muraglitazar and GW1929, but not by fenofibrate. Interestingly, the anti-inflammatory properties of muraglitazar were also translated in vivo. This was evidenced by the finding that muraglitazar inhibited carrageenan-induced paw inflammation in a dose-dependent manner in mice as did iNOS inhibitor L-NIL and anti-inflammatory steroid dexamethasone. Conclusions These results show that muraglitazar has anti-inflammatory properties both in vitro and in vivo and these effects reflect the agonistic action through both PPARα and PPARγ.
Collapse
|
11
|
|
12
|
Abstract
Diabetes is a complex disease defined by hyperglycaemia; however, strong associations with abdominal obesity, hypertension and dyslipidaemia contribute to the high risk of cardiovascular disease. Although aggressive glycaemic control reduces microvascular complications, the evidence for macrovascular complications is less certain. The theoretical benefits of the mode of action of peroxisome proliferator-activated receptor (PPAR) agonists are clear. In clinical practice, PPAR-α agonists such as fibrates improve dyslipidaemia, while PPAR-γ agonists such as thiazolidinediones improve insulin resistance and diabetes control. However, although these agents are traditionally classed according to their target, they have different and sometimes conflicting clinical benefit and adverse event profiles. It is speculated that this is because of differing properties and specificities for the PPAR receptors (each of which targets specific genes). This is most obvious in the impact on cardiovascular outcomes--in clinical trials pioglitazone appeared to reduce cardiovascular events, whereas rosiglitazone potentially increased the risk of myocardial infarction. The development of a dual PPAR-α/γ agonist may prove beneficial in effectively managing glycaemic control and improving dyslipidaemia in patients with type 2 diabetes. Yet, development of agents such as muraglitazar and tesaglitazar has been hindered by various serious adverse events. Aleglitazar, a balanced dual PPAR-α/γ agonist, is currently the most advanced in clinical development and has shown promising results in phase II clinical trials with beneficial effects on glucose and lipid variables. A phase III study, ALECARDIO, is ongoing and will establish whether improvements in laboratory test profiles translate into an improvement in cardiovascular outcomes.
Collapse
Affiliation(s)
- J P H Wilding
- Department of Obesity & Endocrinology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
13
|
Scaffold-based pan-agonist design for the PPARα, PPARβ and PPARγ receptors. PLoS One 2012; 7:e48453. [PMID: 23119024 PMCID: PMC3485212 DOI: 10.1371/journal.pone.0048453] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 09/26/2012] [Indexed: 12/25/2022] Open
Abstract
As important members of nuclear receptor superfamily, Peroxisome proliferator-activated receptors (PPAR) play essential roles in regulating cellular differentiation, development, metabolism, and tumorigenesis of higher organisms. The PPAR receptors have 3 identified subtypes: PPARα, PPARβ and PPARγ, all of which have been treated as attractive targets for developing drugs to treat type 2 diabetes. Due to the undesirable side-effects, many PPAR agonists including PPARα/γ and PPARβ/γ dual agonists are stopped by US FDA in the clinical trials. An alternative strategy is to design novel pan-agonist that can simultaneously activate PPARα, PPARβ and PPARγ. Under such an idea, in the current study we adopted the core hopping algorithm and glide docking procedure to generate 7 novel compounds based on a typical PPAR pan-agonist LY465608. It was observed by the docking procedures and molecular dynamics simulations that the compounds generated by the core hopping and glide docking not only possessed the similar functions as the original LY465608 compound to activate PPARα, PPARβ and PPARγ receptors, but also had more favorable conformation for binding to the PPAR receptors. The additional absorption, distribution, metabolism and excretion (ADME) predictions showed that the 7 compounds (especially Cpd#1) hold high potential to be novel lead compounds for the PPAR pan-agonist. Our findings can provide a new strategy or useful insights for designing the effective pan-agonists against the type 2 diabetes.
Collapse
|
14
|
Kalanuria AA, Nyquist P, Ling G. The prevention and regression of atherosclerotic plaques: emerging treatments. Vasc Health Risk Manag 2012; 8:549-61. [PMID: 23049260 PMCID: PMC3459726 DOI: 10.2147/vhrm.s27764] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Indexed: 01/21/2023] Open
Abstract
Occlusive vascular diseases, such as sudden coronary syndromes, stroke, and peripheral arterial disease, are a huge burden on the health care systems of developed and developing countries. Tremendous advances have been made over the last few decades in the diagnosis and treatment of atherosclerotic diseases. Intravascular ultrasound has been able to provide detailed information of plaque anatomy and has been used in several studies to assess outcomes. The presence of atherosclerosis disrupts the normal protective mechanism provided by the endothelium and this mechanism has been implicated in the pathophysiology of coronary artery disease and stroke. Efforts are being put into the prevention of atherosclerosis, which has been shown to begin in childhood. This paper reviews the pathophysiology of atherosclerosis and discusses the current options available for the prevention and reversal of plaque formation.
Collapse
Affiliation(s)
- Atul Ashok Kalanuria
- Division of Neuro Critical Care, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, USA
| | | | | |
Collapse
|
15
|
Whayne TF. Coronary atherosclerosis, low-density lipoproteins and markers of thrombosis, inflammation and endothelial dysfunction. Int J Angiol 2012; 16:12-6. [PMID: 22477242 DOI: 10.1055/s-0031-1278237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Available information regarding the relation among atherosclerosis, low-density lipoproteins, markers of thrombosis, inflammation and endothelial dysfunction has accumulated, but is still very limited, making only minimal contributions to clinical decision-making. Many more clinical trials are needed, but unless there is a relationship between atherosclerosis prevention, specific markers and a pharmaceutical product, financial support for such trials will be difficult to obtain. The anti-inflammatory effect of statins is well established. Angiotensin-converting enzyme inhibitors are generally not thought of as having anti-inflammatory effects, but the European Trial on Reduction of Cardiac Events with Perindopril in Stable Coronary Artery Disease (EUROPA) study observed extensive RR reduction with perindopril. It was explained not simply by control of hypertension, but by reduced activity of multiple factors, supported by specific substudies. The 'cardiovascular continuum' is an excellent unifying term to explain atherosclerosis mechanisms, relate mechanisms to clinical understanding, and assist the clinician in selecting the appropriate prevention and control therapies. This so-called continuum actually describes a relationship among different biochemical, enzymatic and hormonal factors that affect the cardiovascular system. It can be seen in the downregulation of the angiotensin II receptor type 1 by statins, which contributes to hypertension control while lowering low-density lipoproteins. Peroxisome proliferator activator receptor-gamma also demonstrates the cardiovascular continuum with activation of the receptor by glitazones. The glitazones increase insulin sensitivity for diabetes control. Activation of the peroxisome proliferator activator receptor-gamma inhibits inflammation, which is possibly related to atherosclerosis, normalization of endothelial function, suppression of metalloproteinases and a decrease in smooth muscle cell migration. All of these effects may decrease atherosclerosis production while improving control of diabetes mellitus, a key disease in the cardiovascular continuum for development of atherosclerosis. Consideration of such interrelationships is just scratching the surface. Nevertheless, it can be seen that the complicated process of atherosclerosis development has a multifaceted explanation that has been minimally defined, but holds the key to prevention and control of this major medical problem faced in modern society.
Collapse
Affiliation(s)
- Thomas F Whayne
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
16
|
Pirat C, Farce A, Lebègue N, Renault N, Furman C, Millet R, Yous S, Speca S, Berthelot P, Desreumaux P, Chavatte P. Targeting Peroxisome Proliferator-Activated Receptors (PPARs): Development of Modulators. J Med Chem 2012; 55:4027-61. [DOI: 10.1021/jm101360s] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Céline Pirat
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Amaury Farce
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Nicolas Lebègue
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Nicolas Renault
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Christophe Furman
- Institut de Chimie Pharmaceutique
Albert Lespagnol, Université Lille-Nord de France, EA 4481, 3 Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex,
France
| | - Régis Millet
- Institut de Chimie Pharmaceutique
Albert Lespagnol, Université Lille-Nord de France, EA 4481, 3 Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex,
France
| | - Saı̈d Yous
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Silvia Speca
- Faculté de
Médecine, Amphis J et K, Université Lille-Nord de France, INSERM U995, Boulevard du Professeur Jules
Leclerc, 59045 Lille Cedex, France
| | - Pascal Berthelot
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
| | - Pierre Desreumaux
- Faculté de
Médecine, Amphis J et K, Université Lille-Nord de France, INSERM U995, Boulevard du Professeur Jules
Leclerc, 59045 Lille Cedex, France
| | - Philippe Chavatte
- Laboratoire de Chimie Thérapeutique,
Faculté des Sciences Pharmaceutiques et Biologiques, Université Lille-Nord de France, EA 4481, 3
Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex, France
- Institut de Chimie Pharmaceutique
Albert Lespagnol, Université Lille-Nord de France, EA 4481, 3 Rue du Professeur Laguesse, BP 83, 59006 Lille Cedex,
France
| |
Collapse
|
17
|
Kwon HS. New therapeutic agents for glycemic control in diabetes mellitus. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2012. [DOI: 10.5124/jkma.2012.55.3.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Hyuk Sang Kwon
- Department of Endocrinology, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Abstract
There is a rising worldwide prevalence of diabetes, especially type 2 diabetes mellitus (T2DM), which is one of the most challenging health problems in the 21st century. The associated complications of diabetes, such as cardiovascular disease, peripheral vascular disease, stroke, diabetic neuropathy, amputations, renal failure, and blindness result in increasing disability, reduced life expectancy, and enormous health costs. T2DM is a polygenic disease characterized by multiple defects in insulin action in tissues and defects in pancreatic insulin secretion, which eventually leads to loss of pancreatic insulin-secreting cells. The treatment goals for T2DM patients are effective control of blood glucose, blood pressure, and lipids (if elevated) and, ultimately, to avert the serious complications associated with sustained tissue exposure to excessively high glucose concentrations. Prevention and control of diabetes with diet, weight control, and physical activity has been difficult. Treatment of T2DM has centered on increasing insulin levels, either by direct insulin administration or oral agents that promote insulin secretion, improving sensitivity to insulin in tissues, or reducing the rate of carbohydrate absorption from the gastrointestinal tract. This review presents comprehensive and up-to-date information on the mechanism(s) of action, efficacy, pharmacokinetics, pleiotropic effects, drug interactions, and adverse effects of the newer antidiabetic drugs, including (1) peroxisome proliferator-activated-receptor-γ agonists (thiazolidinediones, pioglitazone, and rosiglitazone); (2) the incretin, glucagon-like peptide-) receptor agonists (incretin-mimetics, exenatide. and liraglutide), (3) inhibitors of dipeptidyl-peptidase-4 (incretin enhancers, sitagliptin, and vildagliptin), (4) short-acting, nonsulfonylurea secretagogue, meglitinides (repaglinide and nateglinide), (5) amylin anlog-pramlintide, (6) α-glucosidase inhibitors (miglitol and voglibose), and (7) colesevelam (a bile acid sequestrant). In addition, information is presented on drug candidates in clinical trials, experimental compounds, and some plants used in the traditional treatment of diabetes based on experimental evidence. In the opinion of this reviewer, therapy based on orally active incretins and incretin mimetics with long duration of action that will be efficacious, preserve the β-cell number/function, and block the progression of diabetes will be highly desirable. However, major changes in lifestyle factors such as diet and, especially, exercise will also be needed if the growing burden of diabetes is to be contained.
Collapse
|
19
|
Papanas N, Katsiki N, Hatzitolios AI, Maltezos E. Pioglitazone: a valuable component of combination therapy for type 2 diabetes mellitus. Expert Opin Pharmacother 2011; 12:1457-61. [DOI: 10.1517/14656566.2011.568477] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Ahmed TAN, Karalis I, Jukema JW. Emerging drugs for coronary artery disease. From past achievements and current needs to clinical promises. Expert Opin Emerg Drugs 2011; 16:203-33. [DOI: 10.1517/14728214.2011.549606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Cavender MA, Lincoff AM. Therapeutic potential of aleglitazar, a new dual PPAR-α/γ agonist: implications for cardiovascular disease in patients with diabetes mellitus. Am J Cardiovasc Drugs 2010; 10:209-16. [PMID: 20653327 DOI: 10.2165/11539500-000000000-00000] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Preventing morbidity and mortality from diabetes mellitus is of paramount importance as the incidence of this disease is increasing across the world. While microvascular complications of diabetes such as nephropathy, retinopathy, and neuropathy are reduced with intensive glycemic control, treatment of hyperglycemia has not been consistently shown to have effects on the macrovascular complications of diabetes such as coronary artery, cerebrovascular, and peripheral vascular disease. Preventive efforts have accordingly shifted toward the modification of other cardiovascular risk factors in diabetic patients. Agonism of the peroxisome proliferator-activated receptors (PPARs) has long been an attractive target for antidiabetic therapy due to the role of PPARs in glycemic control and lipid metabolism. PPAR-α agonists such as rosiglitazone and pioglitazone are used in clinical practice for the treatment of diabetes, and there is some evidence that pioglitazone may have positive effects on cardiovascular complications by virtue of its favorable effects on lipid profiles. However, they have not been shown to reduce macrovascular events. PPAR-α agonism is the mechanism of action in the fibrate class of medications; these agents have been shown to increase high-density lipoprotein cholesterol (HDL-C) levels, reduce triglyceride levels, and improve cardiovascular outcomes. Given the prevalence of lipid abnormalities in patients with diabetes, dual PPAR-α/γ agonists (glitazars) could potentially benefit patients with diabetes. A phase II trial examining a novel dual PPAR agonist, aleglitazar, showed that therapy with this agent reduced hyperglycemia and favorably modified levels of HDL-C and triglycerides with an acceptable safety profile. Aleglitazar is currently being studied in large-scale clinical trials to assess whether it will reduce the risk of major cardiovascular endpoints (death, myocardial infarction, or stroke) among patients with diabetes and coronary artery disease. If ongoing studies confirm the theoretical benefit and safety of dual PPAR-α/γ agonism, aleglitazar may become the first therapy demonstrated to reduce macrovascular complications in patients with diabetes.
Collapse
Affiliation(s)
- Matthew A Cavender
- Department of Cardiovascular Medicine, The Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
22
|
Cavender MA, Nicholls SJ, Lincoff AM. Strategies for the development of new PPAR agonists in diabetes. ACTA ACUST UNITED AC 2010; 17 Suppl 1:S32-7. [PMID: 20489419 DOI: 10.1097/01.hjr.0000368196.17109.5e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Diabetes is associated with a range of metabolic abnormalities including insulin resistance, atherogenic dyslipidemia, hypertension, and inflammation. These factors are likely to underlie the adverse cardiovascular outcome typically observed in diabetic cohorts. The family of peroxisome proliferator-activated receptors (PPARs) have been implicated in the regulation of a number of physiologic and metabolic pathways. Pharmacologic agonists directed against various PPARs have been shown to have a beneficial impact on these metabolic risk factors. The potential impact on cardiovascular risk with the use of PPAR agonists will be reviewed.
Collapse
Affiliation(s)
- Matthew A Cavender
- Department of Cardiovascular Medicine, The Cleveland Clinic, Cleveland, Ohio 44195, USA.
| | | | | |
Collapse
|
23
|
Mukherjee S, Mani S. Orphan nuclear receptors as targets for drug development. Pharm Res 2010; 27:1439-68. [PMID: 20372994 PMCID: PMC3518931 DOI: 10.1007/s11095-010-0117-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 03/04/2010] [Indexed: 12/31/2022]
Abstract
Orphan nuclear receptors regulate diverse biological processes. These important molecules are ligand-activated transcription factors that act as natural sensors for a wide range of steroid hormones and xenobiotic ligands. Because of their importance in regulating various novel signaling pathways, recent research has focused on identifying xenobiotics targeting these receptors for the treatment of multiple human diseases. In this review, we will highlight these receptors in several physiologic and pathophysiologic actions and demonstrate how their functions can be exploited for the successful development of newer drugs.
Collapse
Affiliation(s)
- Subhajit Mukherjee
- Departments of Medicine, Genetics and Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Chanin 302-D1, Bronx, New York 10461, USA
| | - Sridhar Mani
- Departments of Medicine, Genetics and Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Chanin 302-D1, Bronx, New York 10461, USA
| |
Collapse
|
24
|
Bailey CJ, Gross JL, Pieters A, Bastien A, List JF. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet 2010; 375:2223-33. [PMID: 20609968 DOI: 10.1016/s0140-6736(10)60407-2] [Citation(s) in RCA: 624] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Correction of hyperglycaemia and prevention of glucotoxicity are important objectives in the management of type 2 diabetes. Dapagliflozin, a selective sodium-glucose cotransporter-2 inhibitor, reduces renal glucose reabsorption in an insulin-independent manner. We assessed the efficacy and safety of dapagliflozin in patients who have inadequate glycaemic control with metformin. METHODS In this phase 3, multicentre, double-blind, parallel-group, placebo-controlled trial, 546 adults with type 2 diabetes who were receiving daily metformin (>/=1500 mg per day) and had inadequate glycaemic control were randomly assigned to receive one of three doses of dapagliflozin (2.5 mg, n=137; 5 mg, n=137; or 10 mg, n=135) or placebo (n=137) orally once daily. Randomisation was computer generated and stratified by site, implemented with a central, telephone-based interactive voice response system. Patients continued to receive their pre-study metformin dosing. The primary outcome was change from baseline in haemoglobin A(1c)(HbA(1c)) at 24 weeks. All randomised patients who received at least one dose of double-blind study medication and who had both a baseline and at least one post-baseline measurement (last observation carried forward) were included in the analysis. Data were analysed by use of ANCOVA models. This trial is registered with ClinicalTrials.gov, number NCT00528879. FINDINGS 534 patients were included in analysis of the primary endpoint (dapagliflozin 2.5 mg, n=135; dapagliflozin 5 mg, n=133; dapagliflozin 10 mg, n=132; placebo, n=134). At week 24, mean HbA(1c) had decreased by -0.30% (95% CI -0.44 to -0.16) in the placebo group, compared with -0.67% (-0.81 to -0.53, p=0.0002) in the dapagliflozin 2.5 mg group, -0.70% (-0.85 to -0.56, p<0.0001) in the dapagliflozin 5 mg group, and -0.84% (-0.98 to -0.70, p<0.0001) in the dapagliflozin 10 mg group. Symptoms of hypoglycaemia occurred in similar proportions of patients in the dapagliflozin (2-4%) and placebo groups (3%). Signs, symptoms, and other reports suggestive of genital infections were more frequent in the dapagliflozin groups (2.5 mg, 11 patients [8%]; 5 mg, 18 [13%]; 10 mg, 12 [9%]) than in the placebo group (seven [5%]). 17 patients had serious adverse events (four in each of the dapagliflozin groups and five in the placebo group). INTERPRETATION Addition of dapagliflozin to metformin provides a new therapeutic option for treatment of type 2 diabetes in patients who have inadequate glycaemic control with metformin alone. FUNDING Bristol-Myers Squibb and AstraZeneca.
Collapse
|
25
|
Natarajan P, Ray KK, Cannon CP. High-Density Lipoprotein and Coronary Heart Disease. J Am Coll Cardiol 2010; 55:1283-99. [PMID: 20338488 DOI: 10.1016/j.jacc.2010.01.008] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 01/04/2010] [Accepted: 01/04/2010] [Indexed: 12/29/2022]
Affiliation(s)
- Pradeep Natarajan
- Department of Medicine, Cardiovascular Division, Brigham & Women's Hospital/Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
26
|
Hosagrahara VP, Chandrasena G, Chang SY, Koplowitz B, Hariharan N, Cheng PTW, Humphreys WG. Pharmacokinetics of muraglitazar (BMS-298585), a dual peroxisome proliferator-activated receptors (PPAR) α and γ activator, in mice, rats, dogs, and monkeys. Xenobiotica 2009; 36:1227-38. [PMID: 17162469 DOI: 10.1080/00498250600829378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The pharmacokinetic parameters of muraglitazar, a novel dual-activator of the peroxisome proliferator-activated receptors (PPAR) alpha and gamma, were determined in mice, rats, dogs, and monkeys after intravenous and oral administration. In the mouse, rat, and monkey the absolute oral bioavailability of muraglitazar ranged from 64 to 88%, and in the dog oral bioavailability was approximately 18%. The systemic clearance values of muraglitazar in the mouse, rat, dog, and cynomolgus monkey were 1.2, 3.0, 12.3 and 1.2 ml min-1 kg-1, respectively. The terminal elimination half-life was 2.4 h in dogs and 7.3 h in rats. The terminal elimination half-life could not be determined in the mouse and monkey because the sampling interval did not adequately cover the terminal elimination phase. Muraglitazar appears to be distributed outside of the vasculature, with the steady-state volume of distribution being approximately twofold that of the vascular volume in rats and dogs, and approximately twofold that of the total body water in mice. The systemic plasma clearance of muraglitazar in humans was predicted to be approximately 12-14 ml min-1 kg-1 based on allometry or by scaling of in vitro clearance parameters. Overall, the pharmacokinetic parameters of muraglitazar in preclinical species were acceptable for the advancement of the compound as a clinical candidate.
Collapse
Affiliation(s)
- V P Hosagrahara
- Department of Metabolism, Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, NJ 08543, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Rubin CJ, Viraswami-Appanna K, Fiedorek FT. Efficacy and safety of muraglitazar: a double-blind, 24-week, dose-ranging study in patients with type 2 diabetes. Diab Vasc Dis Res 2009; 6:205-15. [PMID: 20368213 DOI: 10.1177/1479164109336048] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Muraglitazar is a dual (alpha/gamma) PPAR activator. Dual receptor activation may improve glycaemic and lipid profiles in patients with type 2 diabetes mellitus.This randomised double-blind trial in 1,477 drug-naive patients with type 2 diabetes compared the efficacy and safety of muraglitazar (0.5, 1.5, 5, 10, and 20 mg) with pioglitazone (15 mg). Endpoints included changes in HbA(1C) and plasma lipids, last observation carried forward over 24 weeks. At week 24, mean changes from baseline in HbA(1C) ranged from -0.25% to -1.76% with muraglitazar (p<or=0.0008, 0.5 mg versus each higher muraglitazar dose), compared with -0.57% with pioglitazone. At week 12, tri-glycerides decreased 4-41% with muraglitazar and 9% with pioglitazone. High-density lipoprotein cholesterol increased 6-23% with muraglitazar and 10% with pioglitazone. Oedema-related events occurred with muraglitazar in a dose-dependent incidence (range 9-40%), and at 14% with pioglitazone. Overall, muraglitazar produced simultaneous dose-dependent improvements in glycaemic and lipid parameters in drug-naive patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Cindy J Rubin
- 1Pharmaceutical Research Institute, Bristol-Myers Squibb, Princeton, New Jersey 08543, USA.
| | | | | |
Collapse
|
28
|
Rubin CJ, De Pril V, Fiedorek FT. Coadministration of muraglitazar plus glyburide: improvement of glycaemic and lipid profiles in patients with type 2 diabetes. Diab Vasc Dis Res 2009; 6:120-32. [PMID: 20368202 DOI: 10.1177/1479164109336049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In this trial we evaluated the efficacy and safety of muraglitazar, a dual (alpha/gamma) peroxisome proliferator-activated receptor activator, plus glyburide in patients with type 2 diabetes not controlled with sulphonylurea monotherapy. After 2 weeks of open-label glyburide (15 mg/day), 583 patients were randomised to additionally receive muraglitazar 2.5 mg, 5 mg, or placebo. End points included changes in HbA(1C) and fasting plasma glucose (FPG) at weeks 24 and 102, and changes in lipid parameters at weeks 11/12 and 102.At week 24, mean changes from baseline in HbA(1C) and FPG were significantly greater with glyburide plus muraglitazar 2.5 mg or 5 mg than with glyburide plus placebo (p<0.0001). At week 11/12, triglyceride levels were significantly reduced with muraglitazar (p<0.0001). During the extension phase, muraglitazar demonstrated long-term glycaemic and lipid effects through week 102. Although generally well tolerated, muraglitazar was associated with higher rates of congestive heart failure, cardiovascular events, weight gain and oedema.
Collapse
Affiliation(s)
- Cindy J Rubin
- Pharmaceutical Research Institute, Bristol-Myers Squibb, Princeton, New Jersey 08543, USA.
| | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Peroxisome proliferator activated receptors (PPARs) are ligand-dependent transcription factors that mediate a range of important metabolic functions by transactivation, transrepression or corepression of various gene targets. PPAR agonists also have direct antiatherosclerotic effects, independent of their metabolic effects on glucose and lipid homeostasis. The purpose of this review is to evaluate the currently available evidence for a direct vasculoprotective effect of PPAR agonists. RECENT FINDINGS Current studies have emphasized PPAR-mediated effects on inflammatory and immune responses, oxidative stress, the renin-angiotensin system and modulation of plaque composition. Furthermore, it has become evident that the relative activation of the different PPAR isoforms and the contribution of transactivation of target genes against transrepression of transcription factors need to be considered when assessing the vasculoprotective effects of PPAR agonists. SUMMARY It is anticipated that the antiatherosclerotic effects of PPAR agonists observed in experimental studies will translate into reduced cardiovascular events. This promise is yet to be realized in short-to-medium term studies. Given the central role of the PPAR in gene regulation, particularly in metabolic states, it is possible that more targeted modulation of PPAR signalling may hold many rewards for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Karin A M Jandeleit-Dahm
- Diabetes Division, Albert Einstein JDRF Centre for Diabetes Complications, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
30
|
Rubin CJ, Ledeine JM, Fiedorek FT. Improvement of glycaemic and lipid profiles with muraglitazar plus metformin in patients with type 2 diabetes: an active-control trial with glimepiride. Diab Vasc Dis Res 2008; 5:168-76. [PMID: 18777489 DOI: 10.3132/dvdr.2008.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The efficacy and safety of muraglitazar versus glimepiride were evaluated in patients with type 2 diabetes. After open-label metformin monotherapy, 1,805 patients received randomised therapy with muraglitazar 2.5 mg or 5 mg or with glimepiride 1 mg in a double-blind 52-week study. The primary end point was change in glycosylated haemoglobin (HbA1C); secondary end points were changes in fasting lipid levels and glycaemic indices. At week 52, the reduction in HbA1C with muraglitazar 5 mg plus metformin was superior (p<0.0001) and with muraglitazar 2.5 mg it was non-inferior in comparison with glimepiride. At week 12, muraglitazar significantly decreased triglyceride levels (p<0.0001) and increased levels of high-density lipoprotein cholesterol (HDL-C) (p<0.0001). Oedema, weight gain and heart failure were more evident with muraglitazar. Muraglitazar 5 mg plus metformin significantly improved HbA1C, triglyceride and HDL-C levels in patients with type 2 diabetes. Cardiovascular events were similar among groups (~2%), but there was an imbalance of total mortality in favour of glimepiride.
Collapse
Affiliation(s)
- Cindy J Rubin
- Pharmaceutical Research Institute, Bristol-Myers Squibb, Princeton, New Jersey 08543, USA.
| | | | | |
Collapse
|
31
|
Peroxisome proliferator-activated receptors and the vascular system: beyond their metabolic effects. ACTA ACUST UNITED AC 2008; 2:227-38. [DOI: 10.1016/j.jash.2007.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 12/28/2007] [Accepted: 12/28/2007] [Indexed: 12/19/2022]
|
32
|
Abstract
The dramatic failure of clinical trials evaluating the cholesterol ester transfer protein inhibitor torcetrapib has led to considerable doubt about the value of raising high-density lipoprotein cholesterol (HDL-C) as a treatment for cardiovascular disease. These results have underscored the intricacy of HDL metabolism, with functional quality perhaps being a more important consideration than the circulating quantity of HDL. As a result, HDL-based therapeutics that maintain or enhance HDL functionality warrant closer investigation. In this article, we review the complexity of HDL metabolism, discuss clinical-trial data for HDL-raising agents, including possible reasons for the failure of torcetrapib, and consider the potential for future HDL-based therapies.
Collapse
|
33
|
Rau O, Zettl H, Popescu L, Steinhilber D, Schubert-Zsilavecz M. The Treatment of Dyslipidemia—What's Left in the Pipeline? ChemMedChem 2008; 3:206-21. [DOI: 10.1002/cmdc.200700165] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
34
|
Wang ZJ, Liu Q, Li PP, Zou CH, Shen ZF. Effect of GCP-02, a PPARalpha/gamma dual activator, on glucose and lipid metabolism in insulin-resistant mice. Eur J Pharmacol 2008; 580:277-83. [DOI: 10.1016/j.ejphar.2007.10.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 10/11/2007] [Accepted: 10/16/2007] [Indexed: 12/29/2022]
|
35
|
Calkin AC, Thomas MC. PPAR Agonists and Cardiovascular Disease in Diabetes. PPAR Res 2008; 2008:245410. [PMID: 18288280 PMCID: PMC2233765 DOI: 10.1155/2008/245410] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 10/04/2007] [Indexed: 01/23/2023] Open
Abstract
Peroxisome proliferators activated receptors (PPARs) are ligand-activated nuclear transcription factors that play important roles in lipid and glucose homeostasis. To the extent that PPAR agonists improve diabetic dyslipidaemia and insulin resistance, these agents have been considered to reduce cardiovascular risk. However, data from murine models suggests that PPAR agonists also have independent anti-atherosclerotic actions, including the suppression of vascular inflammation, oxidative stress, and activation of the renin angiotensin system. Many of these potentially anti-atherosclerotic effects are thought to be mediated by transrepression of nuclear factor-kB, STAT, and activator protein-1 dependent pathways. In recent clinical trials, PPARalpha agonists have been shown to be effective in the primary prevention of cardiovascular events, while their cardiovascular benefit in patients with established cardiovascular disease remains equivocal. However, the use of PPARgamma agonists, and more recently dual PPARalpha/gamma coagonists, has been associated with an excess in cardiovascular events, possibly reflecting unrecognised fluid retention with potent agonists of the PPARgamma receptor. Newer pan agonists, which retain their anti-atherosclerotic activity without weight gain, may provide one solution to this problem. However, the complex biologic effects of the PPARs may mean that only vascular targeted agents or pure transrepressors will realise the goal of preventing atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Anna C. Calkin
- JDRF Center for Diabetes Complications,
Baker Heart Research Institute,
Melbourne, VIC 3004,
Australia
| | - Merlin C. Thomas
- JDRF Center for Diabetes Complications,
Baker Heart Research Institute,
Melbourne, VIC 3004,
Australia
| |
Collapse
|
36
|
Abstract
Patients with type-2 diabetes mellitus (T2DM) are considered to be at particularly high risk for cardiovascular disease. Over the last decade, the members of the peroxisome proliferator-activated receptor (PPAR) subfamily of nuclear receptors have emerged as valuable pharmacological targets whose activation can normalize metabolic dysfunctions and reduce some cardiovascular risk factors associated with T2DM. PPARalpha agonists, such as the fibrates, can correct dyslipidemia. PPARgamma agonists, such as the thiazolidinediones, act as insulin sensitizers and improve insulin resistance in patients with T2DM. Because of restricted potency and certain side-effects of PPAR agonists, as well as the increasingly epidemic incidence of T2DM, there is a real need for the development of selective PPAR agonists with improved clinical efficacy. This chapter focuses on the PPAR agonists currently used in the clinic, as well as on the discovery and development of the next generation of PPAR agonists.
Collapse
Affiliation(s)
- Barbara Gross
- Institut Pasteur de Lille, 1 rue du Prof Calmette, Lille, F-59019, France
| | | |
Collapse
|
37
|
Abstract
The goal of pharmacogenetics is to define the genetic determinants of individual drug responsiveness, and thereby provide personalized treatment to each individual. The peroxisome proliferator-activated receptors (PPARs) are polypeptide products of a set of related genes functioning to regulate several cellular processes that are central to cardiovascular health and disease. Given their pleiotropic roles in lipid and glucose homeostasis, cardiac energy balance and regulation of adipocyte release of circulating inflammatory factors, it is not surprising that PPARs represent an attractive target for clinical investigation and intervention in disease states, such as diabetes, obesity, atherosclerosis, cardiomyopathy, cardiac hypertrophy and heart failure. Research into the manipulation of PPAR function by pharmacologic agents has already resulted in important advances in the treatment of diabetes mellitus and cardiovascular disease. It follows that PPAR pharmacogenetics promises important advances in the personalized treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Sharon Cresci
- Washington University School of Medicine, Department of Medicine, Saint Louis, Missouri, 660 South Euclid Avenue, Campus Box 8086 Saint Louis, MO 63110-1093, USA
| |
Collapse
|
38
|
Colca JR. Discontinued drugs in 2006: renal, endocrine and metabolic drugs. Expert Opin Investig Drugs 2007; 16:1517-23. [DOI: 10.1517/13543784.16.10.1517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
39
|
Batsis JA, Nieto-Martinez RE, Lopez-Jimenez F. Metabolic syndrome: from global epidemiology to individualized medicine. Clin Pharmacol Ther 2007; 82:509-24. [PMID: 17851562 DOI: 10.1038/sj.clpt.6100355] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The metabolic syndrome (MetS) encompasses a constellation of metabolic abnormalities that are thought to place patients at higher risk for the development of diabetes and cardiovascular (CV) disease. The underlying pathophysiology is still a point of contention among various professional organizations leading to inconsistencies in the manner in which MetS is defined. Each definition has its advantages and disadvantages. Nonetheless, there is an agreement that insulin resistance and obesity are likely the central contributing factors. Because the prevalence of obesity has been increasing at a frightening rate in the past few decades, MetS represents a major public health problem that should be identified clinically in individual patients. This review describes the changing epidemiology of obesity and of MetS and discusses its importance in CV disease. We outline the existing controversies that surround MetS and discuss the role of lifestyle, pharmacological, surgical, and novel approaches in its management.
Collapse
Affiliation(s)
- J A Batsis
- Division of Primary Care Internal Medicine, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | |
Collapse
|
40
|
Waites CR, Dominick MA, Sanderson TP, Schilling BE. Nonclinical Safety Evaluation of Muraglitazar, a Novel PPARα/γ Agonist. Toxicol Sci 2007; 100:248-58. [PMID: 17675651 DOI: 10.1093/toxsci/kfm193] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The toxicity of muraglitazar, an oxybenzylglycine, nonthiazolidinedione peroxisome proliferator-activated receptor (PPAR) alpha/gamma agonist, was evaluated in a comprehensive nonclinical toxicology program that included single-dose oral toxicity studies in mice, rats, and monkeys; repeat-dose toxicity studies in rats, dogs, and monkeys; a battery of in vitro and in vivo genetic toxicity studies; carcinogenicity studies in mice and rats; reproductive and developmental toxicity studies in rats and rabbits; and studies to investigate species-specific findings. Pharmacologically mediated changes, similar to those observed with other PPARgamma agonists, were observed following chronic administration and included subcutaneous edema, hematologic/hematopoietic and serum chemistry alterations, and morphologic findings in the heart and adipose tissue in rats and monkeys. In dogs, a species highly sensitive to PPARgamma agonists, muraglitazar caused pronounced species-specific clinical toxicity and degenerative changes in the brain, spinal cord, and testes at high doses and exposures. Muraglitazar was nongenotoxic in the standard battery of genotoxicity studies. Gallbladder adenomas in male mice and adipocyte neoplasms in male and female rats were seen at suprapharmacologic exposures, whereas urinary bladder tumors occurred in male rats at lower exposures. Subsequent investigative studies established that the urinary bladder carcinogenic effect was mediated by urolithiasis rather than a direct pharmacologic effect on urothelium. Muraglitazar had no effects on reproductive function in male and female rats at high systemic exposures, was not teratogenic in rats or rabbits, and demonstrated no selective developmental toxicity. Overall, there were no nonclinical findings that precluded the safe administration of muraglitazar to humans.
Collapse
Affiliation(s)
- Crystal R Waites
- Bristol-Myers Squibb Research and Development, Drug Safety Evaluation, Mount Vernon, Indiana 47620, USA
| | | | | | | |
Collapse
|
41
|
Bajaj M, Suraamornkul S, Hardies LJ, Glass L, Musi N, DeFronzo RA. Effects of peroxisome proliferator-activated receptor (PPAR)-alpha and PPAR-gamma agonists on glucose and lipid metabolism in patients with type 2 diabetes mellitus. Diabetologia 2007; 50:1723-31. [PMID: 17520238 DOI: 10.1007/s00125-007-0698-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 03/05/2007] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The aim of the study was to examine the effects of pioglitazone (PIO), a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, and fenofibrate (FENO), a PPAR-alpha agonist, as monotherapy and in combination on glucose and lipid metabolism. SUBJECTS AND METHODS Fifteen type 2 diabetic patients received FENO (n = 8) or PIO (n = 7) for 3 months, followed by the addition of the other agent for 3 months in an open-label study. Subjects received a 4 h hyperinsulinaemic-euglycaemic clamp and a hepatic fat content measurement at 0, 3 and 6 months. RESULTS Following PIO, fasting plasma glucose (FPG) (p < 0.05) and HbA(1c) (p < 0.01) decreased, while plasma adiponectin (AD) (5.5 +/- 0.9 to 13.8 +/- 3.5 microg/ml [SEM], p < 0.03) and the rate of insulin-stimulated total-body glucose disposal (R (d)) (23.8 +/- 3.8 to 40.5 +/- 4.4 micromol kg(-1) min(-1), p < 0.005) increased. After FENO, FPG, HbA(1c), AD and R (d) did not change. PIO reduced fasting NEFA (784 +/- 53 to 546 +/- 43 micromol/l, p < 0.05), triacylglycerol (2.12 +/- 0.28 to 1.61 +/- 0.22 mmol/l, p < 0.05) and hepatic fat content (20.4 +/- 4.8 to 10.2 +/- 2.5%, p < 0.02). Following FENO, fasting NEFA and hepatic fat content did not change, while triacylglycerol decreased (2.20 +/- 0.14 to 1.59 +/- 0.13 mmol/l, p < 0.01). Addition of FENO to PIO had no effect on R (d), FPG, HbA(1c), NEFA, hepatic fat content or AD, but triacylglycerol decreased (1.61 +/- 0.22 to 1.00 +/- 0.15 mmol/l, p < 0.05). Addition of PIO to FENO increased R (d) (24.9 +/- 4.4 to 36.1 +/- 2.2 micromol kg(-1) min(-1), p < 0.005) and AD (4.1 +/- 0.8 to 13.1 +/- 2.5 microg/ml, p < 0.005) and reduced FPG (p < 0.05), HbA(1c) (p < 0.05), NEFA (p < 0.01), hepatic fat content (18.3 +/- 3.1 to 13.5 +/- 2.1%, p < 0.03) and triacylglycerol (1.59 +/- 0.13 to 0.96 +/- 0.9 mmol/l, p < 0.01). Muscle adenosine 5'-monophosphate-activated protein kinase (AMPK) activity did not change following FENO; following the addition of PIO, muscle AMPK activity increased significantly (phosphorylated AMPK:total AMPK ratio 1.2 +/- 0.2 to 2.2 +/- 0.3, p < 0.01). CONCLUSIONS/INTERPRETATION We conclude that PPAR-alpha therapy has no effect on NEFA or glucose metabolism and that addition of a PPAR-alpha agonist to a PPAR-gamma agent causes a further decrease in plasma triacylglycerol, but has no effect on NEFA or glucose metabolism.
Collapse
Affiliation(s)
- M Bajaj
- Diabetes Division, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Devasthale PV, Chen S, Jeon Y, Qu F, Ryono DE, Wang W, Zhang H, Cheng L, Farrelly D, Golla R, Grover G, Ma Z, Moore L, Seethala R, Sun W, Doweyko AM, Chandrasena G, Sleph P, Hariharan N, Cheng PTW. Discovery of tertiary aminoacids as dual PPARα/γ agonists-I. Bioorg Med Chem Lett 2007; 17:2312-6. [PMID: 17292606 DOI: 10.1016/j.bmcl.2007.01.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 01/17/2007] [Accepted: 01/18/2007] [Indexed: 10/23/2022]
Abstract
A novel series of potent dual agonists of PPARalpha and PPARgamma, the alkoxybenzylglycines, was identified and explored using a solution-phase library approach. The synthesis and structure-activity relationships of this series of dual PPARalpha/gamma agonists are described.
Collapse
Affiliation(s)
- Pratik V Devasthale
- Discovery Chemistry, Bristol-Myers Squibb Pharmaceutical Research Institute, PO Box 5400, Princeton, NJ 08543-5400, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rosenson RS. Effects of peroxisome proliferator-activated receptors on lipoprotein metabolism and glucose control in type 2 diabetes mellitus. Am J Cardiol 2007; 99:96B-104B. [PMID: 17307062 DOI: 10.1016/j.amjcard.2006.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are central regulators of lipoprotein metabolism and glucose homeostasis that are considered particularly useful for improving glycemic control and comorbidities in patients with type 2 diabetes mellitus. Clinical trials of PPAR-alpha agonists have demonstrated efficacy in reducing cardiovascular events; however, these benefits have been confined to subgroups of patients with low levels of high-density lipoprotein cholesterol or high levels of triglycerides. While activators of PPAR-gamma reduce early atherosclerotic lesions and reduce cardiovascular events, these agents have the effect of increasing fluid retention in patients, which results in more hospitalizations for congestive heart failure. Future studies of PPAR-gamma agonists or dual PPAR-alpha/gamma agonists will require further delineation of the risk profile to avoid adverse outcomes in susceptible patients.
Collapse
Affiliation(s)
- Robert S Rosenson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48106-0363, USA.
| |
Collapse
|
44
|
Affiliation(s)
- Philip R Liebson
- Section of Cardiology, Rush Medical College, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
45
|
A rapid, homogeneous, fluorescence polarization binding assay for peroxisome proliferator-activated receptors alpha and gamma using a fluorescein-tagged dual PPARalpha/gamma activator. Anal Biochem 2007; 363:263-74. [PMID: 17335769 DOI: 10.1016/j.ab.2007.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 01/16/2007] [Accepted: 01/17/2007] [Indexed: 10/23/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) and other members of the nuclear hormone receptor family are important drug targets for the treatment of metabolic diseases. PPARalpha and PPARgamma play crucial roles in lipid and glucose metabolism, respectively. Therefore, screening methods that help to rapidly identify activators of these receptors should be of considerable value. A homogeneous fluorescence polarization (FP) ligand binding assay capable of rapidly identifying ligands that bind to both PPARalpha and PPARgamma has been developed using purified PPARalpha or PPARgamma ligand binding domains and a fluorescein-labeled analog (FLA) of a potent dual PPARalpha/gamma activator. FLA activator showed good binding affinity toward both PPARalpha (K(i)=0.7microM) and PPARgamma (K(i)=0.4microM). The binding of FLA activator was rapid and reached a plateau within 10 min. The resulting FP signal was stable for at least 18h. The FP binding assay performed robustly in a 384-well format, and the average Z' value was 0.77. There was a good correlation between the binding potency (IC(50) values) and rank order of binding potency for a panel of standard PPAR ligands obtained in FP binding assay and scintillation proximity assay or gel filtration binding assays using (3)H-labeled PPARalpha (r(2)=0.99) and PPARgamma (r(2)=0.99) ligands. There was also a good correlation of IC(50) values obtained by FP binding assay and scintillation proximity assay for the clinically used PPAR activators. Thus, the FP binding assay with a single fluorescein-labeled PPARalpha/gamma dual activator offers a homogeneous nonradioactive, sensitive, robust, and less expensive high-throughput assay for detecting compounds that bind to both PPARgamma and PPARalpha. Using this FP binding assay, we have identified a large number of PPARalpha/gamma dual activators. A similar assay platform may be easily adapted to other members of the nuclear hormone receptor family.
Collapse
|
46
|
Chira EC, McMillen TS, Wang S, Haw A, O'Brien KD, Wight TN, Chait A. Tesaglitazar, a dual peroxisome proliferator-activated receptor alpha/gamma agonist, reduces atherosclerosis in female low density lipoprotein receptor deficient mice. Atherosclerosis 2007; 195:100-9. [PMID: 17214992 PMCID: PMC2702263 DOI: 10.1016/j.atherosclerosis.2006.12.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Revised: 11/22/2006] [Accepted: 12/11/2006] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The transcription factors, peroxisome proliferator-activated receptors (PPAR) alpha (alpha) and gamma (gamma), which are involved in lipid and glucose homeostasis, also exert modulatory actions on vascular cells where they exhibit anti-inflammatory and anti-proliferative properties. Hence, PPAR agonists potentially can affect atherogenesis both via metabolic effects and direct effects on the vessel wall. We tested whether the dual PPAR-alpha/gamma agonist, tesaglitazar (TZ), would reduce atherosclerosis in a non-diabetic, atherosclerosis-prone mouse model, independent of effects on plasma lipids. METHODS AND RESULTS Low-density lipoprotein receptor deficient (LDLr-/-) mice were fed a Western type diet consisting of 21% butterfat and 0.15% cholesterol, with or without TZ 0.5 micromol/kg of diet, for 12 weeks. TZ reduced atherosclerosis in the female, but not male, LDLr-/- mice without affecting cholesterol and triglyceride levels, HDL binding to biglycan, or the inflammatory markers serum amyloid A (SAA) and serum amyloid P (SAP). TZ also decreased adiposity in both genders. CONCLUSIONS TZ reduced atherosclerosis in the female LDLr-/- mice via lipid-independent mechanisms, probably at least in part by direct actions on the vessels. The body weight changes in these mice are different from the effects of dual PPAR agonists seen in humans.
Collapse
Affiliation(s)
- Ebele C Chira
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Ahmed I, Furlong K, Flood J, Treat VP, Goldstein BJ. Dual PPAR α/γ Agonists: Promises and Pitfalls in Type 2 Diabetes. Am J Ther 2007; 14:49-62. [PMID: 17303976 DOI: 10.1097/01.mjt.0000212890.82339.8d] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Type 2 diabetes mellitus is a disease of complex pathogenesis and pleiotropic clinical manifestations. The greatest clinical challenge in this disease is the prevention of the long-term complications, many of which involve cardiovascular outcomes. The peroxisome proliferator-activated receptor (PPAR) alpha and gamma isoforms of the family of nuclear transcription factors are pharmaceutical targets for therapeutic intervention because they can potentially ameliorate not only the hyperglycemia of diabetes, but also the dyslipidemia that is characteristic of this disorder (low high-density lipoprotein cholesterol, high triglycerides, small, dense low-density lipoprotein particles). Novel drugs with dual PPAR alpha and gamma activity have been under clinical development for type 2 diabetes, and they have shown promise in early studies with regard to glucose lowering and improved lipid profile when compared with the PPAR-gamma-specific thiazolidinediones. Unfortunately, the dual PPARs available to date have some of the PPAR-gamma-associated side effect profile, including fluid retention and weight gain, which have limited the further clinical development of higher doses that show improved efficacy. This review will briefly summarize our understanding of the pathogenesis of type 2 diabetes, the role of the PPAR family of receptors, and the potential for clinical use of this novel emerging class of agents that serve as dual activators of both PPAR-alpha and PPAR-gamma.
Collapse
Affiliation(s)
- Intekhab Ahmed
- Division of Endocrinology, Diabetes and Metabolic Diseases, Department of Medicine, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
48
|
Goldstein BJ, Rosenstock J, Anzalone D, Tou C, Ohman KP. Effect of tesaglitazar, a dual PPAR alpha/gamma agonist, on glucose and lipid abnormalities in patients with type 2 diabetes: a 12-week dose-ranging trial. Curr Med Res Opin 2006; 22:2575-90. [PMID: 17166340 DOI: 10.1185/030079906x154169] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The Glucose and Lipid Assessment in Diabetes (GLAD) trial examined the dose-response relationship of the dual peroxisome proliferator-activated receptor (PPAR) alpha/gamma agonist tesaglitazar in type 2 diabetic patients. STUDY DESIGN GLAD was a 12-week, multicenter, international, randomized, parallel-group trial. Five-hundred men and women aged 30-80 years with type 2 diabetes (fasting plasma glucose [FPG] > or = 126 mg/dL [> or = 7.0 mmol/L]) received once-daily, double-blind placebo or tesaglitazar (0.1 mg, 0.5 mg, 1.0 mg, 2.0 mg, or 3.0 mg) or open-label pioglitazone (45 mg), included as a therapeutic benchmark. MAIN OUTCOME MEASURES Placebo-corrected changes from baseline in FPG (primary end point), plasma lipids, and insulin-resistance measures. RESULTS At baseline, the mean patient age was 56.1 years, 57.5 years, and 58.9 years for placebo, across tesaglitazar groups, and for pioglitazone, respectively. For the corresponding groups, mean body mass index was 30.6 kg/m2, 30.9 kg/m2, and 29.7 kg/m2, and mean HbA1c was 7.0%, 7.2%, and 7.0%, respectively. At 12 weeks, tesaglitazar 0.5 mg, 1.0 mg, 2.0 mg, and 3.0 mg produced statistically significant reductions in FPG (-30.3 mg/dL, -41.1 mg/dL, -55.0 mg/dL, -60.9 mg/dL; p < 0.0001), triglycerides (-17.2%, -32.9%, -41.0%, -40.9%; p < 0.01), and apolipoprotein B (-15.0%, -15.7%, -21.0%, -22.3%, respectively; p < 0.0001). Tesaglitazar at doses > or = 1.0 mg significantly increased high-density lipoprotein-cholesterol (HDL-C) (15.0%, 13.0%, 12.9%; p < 0.001), and reduced non-HDL-C (-13.2%, -22.2%, -25.0%; p < 0.0001), very-low-density lipoprotein-cholesterol (VLDL-C) (-36.9%, -49.8%, -52.5%; p < 0.0001), and total cholesterol (-6.8%, -14.1%, -15.5%, respectively; p < 0.01). Tesaglitazar > or = 0.5 mg improved insulin-resistance measures. Although no formal statistical analyses were performed between active treatments, improvements in efficacy measures with tesaglitazar 1.0 mg were numerically similar to or greater than those with pioglitazone. Similar numbers of adverse events occurred in the tesaglitazar < or = 1.0 mg, placebo, and pioglitazone arms, but there was an increasing frequency of discontinuations due to pre-specified hematologic and clinical-chemistry criteria with tesaglitazar doses > or = 1.0 mg. CONCLUSIONS In type 2 diabetic patients, tesaglitazar dose-dependently reduced FPG levels at doses > or = 0.5 mg. Other markers of glycemic control, atherogenic dyslipidemia, and measures associated with insulin resistance were improved at doses > or = 0.5 mg or > or = 1.0 mg. Study limitations included that the majority of patients were white, patients had good glycemic control at baseline, and the increased number of early withdrawals in the tesaglitazar 2.0 mg and 3.0 mg doses limits conclusions about the efficacy of these doses. The 0.5 mg and 1.0 mg tesaglitazar doses were identified for further investigation.
Collapse
Affiliation(s)
- Barry J Goldstein
- Department of Medicine, Jefferson Medical College of Thomas Jefferson University, 349 Jefferson Alumni Hall, 1020 Locust Street, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
49
|
Stefanski DA, Majkowska L. Existing and potential therapeutic approaches targeting peroxisome proliferator-activated receptors in the management of Type 2 diabetes. Expert Opin Ther Pat 2006. [DOI: 10.1517/13543776.16.12.1713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
50
|
Zadelaar ASM, Boesten LSM, Jukema JW, van Vlijmen BJM, Kooistra T, Emeis JJ, Lundholm E, Camejo G, Havekes LM. Dual PPARα/γ Agonist Tesaglitazar Reduces Atherosclerosis in Insulin-Resistant and Hypercholesterolemic ApoE*3Leiden Mice. Arterioscler Thromb Vasc Biol 2006; 26:2560-6. [PMID: 16931788 DOI: 10.1161/01.atv.0000242904.34700.66] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We investigated whether the dual PPARalpha/gamma agonist tesaglitazar has anti-atherogenic effects in ApoE*3Leiden mice with reduced insulin sensitivity. METHODS AND RESULTS ApoE*3Leiden transgenic mice were fed a high-fat (HF) insulin-resistance-inducing diet. One group received a high-cholesterol (HC) supplement (1% wt/wt; HC group). A second group received the same HC supplement along with tesaglitazar (T) 0.5 micromol/kg diet (T group). A third (control) group received a low-cholesterol (LC) supplement (0.1% wt/wt; LC group). Tesaglitazar decreased plasma cholesterol by 20% compared with the HC group; cholesterol levels were similar in the T and LC groups. Compared with the HC group, tesaglitazar caused a 92% reduction in atherosclerosis, whereas a 56% reduction was seen in the cholesterol-matched LC group. Furthermore, tesaglitazar treatment significantly reduced lesion number beyond that expected from cholesterol lowering and induced a shift to less severe lesions. Concomitantly, tesaglitazar reduced macrophage-rich and collagen areas. In addition, tesaglitazar reduced inflammatory markers, including plasma SAA levels, the number of adhering monocytes, and nuclear factor kappaB-activity in the vessel wall. CONCLUSIONS Tesaglitazar has anti-atherosclerotic effects in the mouse model that go beyond plasma cholesterol lowering, possibly caused by a combination of altered lipoprotein profiles and anti-inflammatory vascular effects.
Collapse
Affiliation(s)
- A Susanne M Zadelaar
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|