1
|
Deng W, Wang X, Niu X, Zhang X, Hou Y, Qin M. Inverse Agonists of Peroxisome Proliferator-Activated Receptor Gamma: Advances and Prospects in Cancer Treatment. J Med Chem 2025. [PMID: 40300091 DOI: 10.1021/acs.jmedchem.5c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-dependent nuclear transcription factor that modulates metabolic homeostasis and cell proliferation. Inverse agonism of PPARγ is an emerging anticancer strategy, particularly for the treatment of bladder cancer. The first-in-class PPARγ inverse agonist, FX-909, is currently being studied in clinical trials for cancer treatment. However, PPARγ inverse agonists are still in the early stages of development. The discovery of compounds with novel chemical structures, potent efficacy, and favorable pharmacokinetic properties is urgently needed. In this perspective, the biological functions of PPARγ and its role in cancer pathology are introduced, and currently available PPARγ inverse agonists and their preliminary structure-activity relationships (SARs) are discussed from a medicinal chemistry viewpoint. These findings inform the development of anticancer agents that act as PPARγ inverse agonists. Furthermore, our discussion of the complex biological functions of PPARγ provides insights into the exploration of its role in various diseases.
Collapse
Affiliation(s)
- Wensong Deng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xuejian Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinyu Niu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiangjie Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yunlei Hou
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingze Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
2
|
Orsi D, Pook E, Bräuer N, Friberg A, Lienau P, Lemke CT, Stellfeld T, Brüggemeier U, Pütter V, Meyer H, Baco M, Tang S, Cherniack AD, Westlake L, Bender SA, Kocak M, Strathdee CA, Meyerson M, Eis K, Goldstein JT. Discovery and Structure-Based Design of Potent Covalent PPARγ Inverse-Agonists BAY-4931 and BAY-0069. J Med Chem 2022; 65:14843-14863. [PMID: 36270630 PMCID: PMC9662185 DOI: 10.1021/acs.jmedchem.2c01379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Indexed: 11/29/2022]
Abstract
The ligand-activated nuclear receptor peroxisome-proliferator-activated receptor-γ (PPARG or PPARγ) represents a potential target for a new generation of cancer therapeutics, especially in muscle-invasive luminal bladder cancer where PPARγ is a critical lineage driver. Here we disclose the discovery of a series of chloro-nitro-arene covalent inverse-agonists of PPARγ that exploit a benzoxazole core to improve interactions with corepressors NCOR1 and NCOR2. In vitro treatment of sensitive cell lines with these compounds results in the robust regulation of PPARγ target genes and antiproliferative effects. Despite their imperfect physicochemical properties, the compounds showed modest pharmacodynamic target regulation in vivo. Improvements to the in vitro potency and efficacy of BAY-4931 and BAY-0069 compared to those of previously described PPARγ inverse-agonists show that these compounds are novel tools for probing the in vitro biology of PPARγ inverse-agonism.
Collapse
Affiliation(s)
- Douglas
L. Orsi
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Elisabeth Pook
- Research
and Development, Pharmaceuticals, Bayer
AG, 13353 Berlin, Germany
| | | | | | - Philip Lienau
- Research
and Development, Pharmaceuticals, Bayer
AG, 13353 Berlin, Germany
| | - Christopher T. Lemke
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | - Ulf Brüggemeier
- Research
and Development, Pharmaceuticals, Bayer
AG, 13353 Berlin, Germany
| | | | | | - Maria Baco
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Stephanie Tang
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andrew D. Cherniack
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02215, United States
| | - Lindsay Westlake
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Samantha A. Bender
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Mustafa Kocak
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Craig A. Strathdee
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Matthew Meyerson
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02215, United States
- Center for
Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department
of Genetics and Medicine, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Knut Eis
- Research
and Development, Pharmaceuticals, Bayer
AG, 13353 Berlin, Germany
| | - Jonathan T. Goldstein
- Cancer
Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
3
|
Peng T, Wang G, Cheng S, Xiong Y, Cao R, Qian K, Ju L, Wang X, Xiao Y. The role and function of PPARγ in bladder cancer. J Cancer 2020; 11:3965-3975. [PMID: 32328200 PMCID: PMC7171493 DOI: 10.7150/jca.42663] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/08/2020] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily, participates in multiple physiological and pathological processes. Extensive studies have revealed the relationship between PPARγ and various tumors. However, the expression and function of PPARγ in bladder cancer seem to be controversial. It has been demonstrated that PPARγ affects the occurrence and progression of bladder cancer by regulating proliferation, apoptosis, metastasis, and reactive oxygen species (ROS) and lipid metabolism, probably through PPARγ-SIRT1 feedback loops, the PI3K-Akt signaling pathway, and the WNT/β-catenin signaling pathway. Considering the frequent relapses after chemotherapy, some researchers have focused on the relationship between PPARγ and chemotherapy sensitivity in bladder cancer. Moreover, the feasibility of PPARγ ligands as potential therapeutic targets for bladder cancer has been uncovered. Taken together, this review summarizes the relevant literature and our findings to explore the complicated role and function of PPARγ in bladder cancer.
Collapse
Affiliation(s)
- Tianchen Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Songtao Cheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Yaoyi Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Rui Cao
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Kaiyu Qian
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| |
Collapse
|
4
|
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: 1.0] [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
|
5
|
Cheng S, Qian K, Wang Y, Wang G, Liu X, Xiao Y, Wang X. PPARγ inhibition regulates the cell cycle, proliferation and motility of bladder cancer cells. J Cell Mol Med 2019; 23:3724-3736. [PMID: 30912275 PMCID: PMC6484405 DOI: 10.1111/jcmm.14280] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear receptor family of ligand-activated transcription factors and plays an important role in regulating cell proliferation, inflammation and lipid and glucose homeostasis. Our results revealed that PPARγ was up-regulated in human bladder cancer (BCa) tissues both at transcriptional and translational levels. Moreover, down-regulation of PPARγ mRNA or inhibition of PPARγ function (using GW9662, antagonist of PPARγ) could significantly suppress the proliferation of BCa cells. Furthermore, the cell cycle arrested in G0/G1 phase was also induced by the down-regulated PPARγ possibly through AKT-mediated up-regulation of p21/p27, whereas no significant transformation of apoptosis was observed. In addition, knockdown or inhibition of PPARγ might reduce the invasion and migration of BCa cells by affecting epithelial-mesenchymal transition-related proteins through AKT/GSK3β signalling pathway. Additionally, in vivo studies showed that BCa cell proliferation was significantly suppressed by GW9662. In conclusion, our results indicated that PPARγ might be crucial for BCa tumorigenesis by interfering with the motility and viability of BCa cells.
Collapse
Affiliation(s)
- Songtao Cheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaiyu Qian
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yejinpeng Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, District of Columbia
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Lv S, Wang W, Wang H, Zhu Y, Lei C. PPARγ activation serves as therapeutic strategy against bladder cancer via inhibiting PI3K-Akt signaling pathway. BMC Cancer 2019; 19:204. [PMID: 30845932 PMCID: PMC6407222 DOI: 10.1186/s12885-019-5426-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/01/2019] [Indexed: 01/07/2023] Open
Abstract
Background Heterogeneity in bladder cancer results in variable clinical outcomes, posing challenges for clinical management of this malignancy. Recent studies suggest both tumor suppressive and oncogenic role of PPARγ in bladder cancer. The fuction of PPARγ signaling pathway in modulating carcinogenesis is controversial. Methods The expression of PPARγ and association with overall survival were analyzed in patients from two cohorts. The effect of PPARγ activation on cell proliferation, cell cycle, and cell apoptosis were determined with the agonists (rosiglitazone and pioglitazone), the inverse agonist (T0070907), and the antagonist (GW9662) in Umuc-3 and 5637 bladder cancer cells. The correlation of PPARγ activation with PI3K-Akt pathway was evaluated with RNA sequencing data from the TCGA cases and 30 human bladder cancer cell lines. The effect of PPARγ activation on tumor growth was validated with subcutaneous tumor models in vivo. The effect of PPARγ activation on PI3K-Akt signaling transduction was determined with multiple assays including immunohistochemistry, flow cytometry, proteomic array, and western blotting. Results We showed that PPARγ was a favorable prognostic factor in patients with bladder cancer. PPARγ activation by rosiglitazone and pioglitazone markedly induced cell cycle G2 arrest and apoptosis in bladder cancer cells, which resulted in inhibition of cell proliferation in vitro and suppression of tumor growth in vivo. The underlying mechanism involved marked inhibition of PI3K-Akt pathway. Conclusions This study reported the tumor-suppressive effect of PPARγ agonists in bladder cancer, suggesting that transactivation of PPARγ could be served as a potential strategy for the chemoprevention and therapeutic treatment of bladder cancer. Electronic supplementary material The online version of this article (10.1186/s12885-019-5426-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shidong Lv
- Department of Urology, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, China
| | - Wei Wang
- Department of Pathology, General Hospital of Southern Theater Command, PLA, Guangzhou, 510010, China
| | - Hongyi Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, China
| | - Yongtong Zhu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengyong Lei
- Department of Urology, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Kim SH, Hong SH, Park YJ, Sung JH, Suh W, Lee KW, Jung K, Lim C, Kim JH, Kim H, Park KS, Park SG. MD001, a Novel Peroxisome Proliferator-activated Receptor α/γ Agonist, Improves Glucose and Lipid Metabolism. Sci Rep 2019; 9:1656. [PMID: 30733541 PMCID: PMC6367362 DOI: 10.1038/s41598-018-38281-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/20/2018] [Indexed: 01/14/2023] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have been developed to treat metabolic diseases; however, most of them exhibit side effects such as body weight gain and oedema. Therefore, we developed a novel PPARα/γ dual agonist that modulates glucose and lipid metabolism without adverse effects. We synthesised novel compounds composed of coumarine and chalcone, determined their crystal structures, and then examined their binding affinity toward PPARα/γ. We investigated the expression of PPARα and PPARγ target genes by chemicals in HepG2, differentiated 3T3-L1, and C2C12 cells. We examined the effect of chemicals on glucose and lipid metabolism in db/db mice. Only MD001 functions as a PPARα/γ dual agonist in vitro. MD001 increased the transcriptional activity of PPARα and PPARγ, resulting in enhanced expression of genes related to β-oxidation and fatty acid and glucose uptake. MD001 significantly improved blood metabolic parameters, including triglycerides, free fatty acids, and glucose, in db/db mice. In addition, MD001 ameliorated hepatic steatosis by stimulating β-oxidation in vitro and in vivo. Our results demonstrated the beneficial effects of the novel compound MD001 on glucose and lipid metabolism as a PPARα/γ dual agonist. Consequently, MD001 may show potential as a novel drug candidate for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Seok-Ho Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Shin Hee Hong
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Young-Joon Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Songdo, Incheon, 405-750, Korea
| | - Wonhee Suh
- College of Pharmacy, Chung-Ang University, Seoul, 156-756, Korea
| | - Kyeong Won Lee
- Marine Biotechnology Research Center, Korea Institute of Ocean Science & Technology 787 Haeanlo, Ansan, Gyeonggi-do, 426-744, Korea
| | - Kiwon Jung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Changjin Lim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Korea
| | - Jin-Hee Kim
- College of Pharmacy, Yonsei University, Songdo, Incheon, 405-750, Korea
| | - Hyoungsu Kim
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea
| | - Kyong Soo Park
- The Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, 16499, Korea.
| |
Collapse
|
8
|
Goldstein JT, Berger AC, Shih J, Duke FF, Furst L, Kwiatkowski DJ, Cherniack AD, Meyerson M, Strathdee CA. Genomic Activation of PPARG Reveals a Candidate Therapeutic Axis in Bladder Cancer. Cancer Res 2017; 77:6987-6998. [PMID: 28923856 DOI: 10.1158/0008-5472.can-17-1701] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/02/2017] [Accepted: 09/13/2017] [Indexed: 01/16/2023]
Abstract
The PPARG gene encoding the nuclear receptor PPARγ is activated in bladder cancer, either directly by gene amplification or mutation, or indirectly by mutation of the RXRA gene, which encodes the heterodimeric partner of PPARγ. Here, we show that activating alterations of PPARG or RXRA lead to a specific gene expression signature in bladder cancers. Reducing PPARG activity, whether by pharmacologic inhibition or genetic ablation, inhibited proliferation of PPARG-activated bladder cancer cells. Our results offer a preclinical proof of concept for PPARG as a candidate therapeutic target in bladder cancer. Cancer Res; 77(24); 6987-98. ©2017 AACR.
Collapse
Affiliation(s)
| | - Ashton C Berger
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Juliann Shih
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Fujiko F Duke
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Laura Furst
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Andrew D Cherniack
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Matthew Meyerson
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts. .,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
9
|
Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 2017; 48:52-108. [PMID: 28816105 DOI: 10.1080/10408444.2017.1351420] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thiazolidinedione (TZD) drugs used in the treatment of type 2 diabetes mellitus (T2DM) have proven effective in improving insulin sensitivity, hyperglycemia, and lipid metabolism. Though well tolerated by some patients, their mechanism of action as ligands of peroxisome proliferator-activated receptors (PPARs) results in the activation of several pathways in addition to those responsible for glycemic control and lipid homeostasis. These pathways, which include those related to inflammation, bone formation, and cell proliferation, may lead to adverse health outcomes. As treatment with TZDs has been associated with adverse hepatic, cardiovascular, osteological, and carcinogenic events in some studies, the role of TZDs in the treatment of T2DM continues to be debated. At the same time, new therapeutic roles for TZDs are being investigated, with new forms and isoforms currently in the pre-clinical phase for use in the prevention and treatment of some cancers, inflammatory diseases, and other conditions. The aims of this review are to provide an overview of the mechanism(s) of action of TZDs, a review of their safety for use in the treatment of T2DM, and a perspective on their current and future therapeutic roles.
Collapse
Affiliation(s)
- Melissa A Davidson
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada
| | - Donald R Mattison
- b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada
| | - Laurent Azoulay
- d Center for Clinical Epidemiology , Lady Davis Research Institute, Jewish General Hospital , Montreal , Canada.,e Department of Oncology , McGill University , Montreal , Canada
| | - Daniel Krewski
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada.,f Faculty of Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
10
|
Bugge A, Holst D. PPAR agonists, - Could tissue targeting pave the way? Biochimie 2016; 136:100-104. [PMID: 27916640 DOI: 10.1016/j.biochi.2016.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/28/2016] [Accepted: 10/28/2016] [Indexed: 01/20/2023]
Abstract
Over the last couple of decades, the PPAR family of transcription factors has received much attention from the pharmaceutical industry due to their profound ability to improve glucose and lipid metabolism upon agonist activation. However, more recently the interest in these nuclear receptors has faded because several clinical trials have shown that it is difficult to develop a ligand that significantly ameliorates glucose and lipid metabolism disorders without concomitantly inducing unacceptable side-effects. Nevertheless, the data also suggests that tissue specific targeting could pave the way to renewed interest and clinical use of PPAR ligands. In this review we summarize the results and learnings from the clinical trials on PPAR agonism and discuss the possibilities for tissue targeting of PPAR ligands by using state of the art technology to fuse them to peptides homing selectively to tissues expressing the cognate surface receptor.
Collapse
Affiliation(s)
- Anne Bugge
- Department of Obesity Biology, Global Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Måløv, Denmark
| | - Dorte Holst
- Department of Obesity Biology, Global Research, Novo Nordisk A/S, Novo Nordisk Park, DK-2760, Måløv, Denmark.
| |
Collapse
|
11
|
Ge J, Miao JJ, Sun XY, Yu JY. Huangkui capsule, an extract from Abelmoschus manihot (L.) medic, improves diabetic nephropathy via activating peroxisome proliferator-activated receptor (PPAR)-α/γ and attenuating endoplasmic reticulum stress in rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 189:238-49. [PMID: 27224243 DOI: 10.1016/j.jep.2016.05.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 04/29/2016] [Accepted: 05/14/2016] [Indexed: 05/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Abelmoschus manihot (L.) medic (AM) is a natural medicinal plant used for the treatment of chronic kidney disease (CKD) in China. Huangkui capsule (HKC), an extract from AM, has been proved clinically effective in improving renal inflammation and glomerular injury in CKD. However, the mechanisms of HKC are still not fully understood. AIM OF THE STUDY Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have the potential to be used as therapeutic agents for the treatment of type 2 diabetes and diabetic nephropathy (DN). This study evaluated the function of Huangkui capsule (HKC), an extract from Abelmoschus manihot (L.) medic (AM), as a dual agonist for PPARα/γ and investigated its anti-DN effects in a DN rat model. MATERIALS AND METHODS ChIP and reporter gene assays were performed and the expression of PPARα/γ target genes was monitored to examine the ability of HKC to activate PPARα/γ. DN was induced in male Sprague-Dawley rats via unilateral nephrectomy and intraperitoneal injection of streptozotocin. HKC was administered to the diabetic nephropathy rats at three different doses: high dose HKC (300mg/kg/d); middle dose HKC (175mg/kg/d); and low dose HKC (75mg/kg/d). Irbesartan (4mg/kg/d body weight) was used as a positive control. Following 12 weeks' treatment, we measured general status, renal morphological appearance, proteinuria, blood biochemical parameters, and glomerular morphological changes. The expression of collagen IV, TGFβ, TNFα and IL-6 in renal tissue was evaluated. Endoplasmic reticulum (ER) stress in renal tissue was also analyzed. RESULTS HKC enhanced the transcriptional activity of PPARα and PPARγ in cultured cells, livers and kidneys of DN rats, and it reduced serum triglyceride and cholesterol levels and fat in livers of DN rats. Furthermore, HKC reduced the expressions of inflammatory genes in kidneys of DN rats. Strikingly, HKC reduced ER stress and c-Jun NH2-terminal kinase activation in the liver and kidney of DN rats and subsequently improved renal injury. CONCLUSIONS Our results show that HKC improved lipid metabolic disorders by activating PPARα/γ and attenuating ER stress. HKC could dose-dependently ameliorate renal inflammation and glomerular injury in DN rats. These results suggest that HKC has potential as an anti-DN agent for the treatment of DN in humans.
Collapse
Affiliation(s)
- Jing Ge
- Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing Hanzhong Road, Nanjing, China
| | - Jun-Jun Miao
- Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing Hanzhong Road, Nanjing, China
| | - Xin-Yi Sun
- Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing Hanzhong Road, Nanjing, China
| | - Jiang-Yi Yu
- Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing Hanzhong Road, Nanjing, China.
| |
Collapse
|
12
|
Wright MB, Bortolini M, Tadayyon M, Bopst M. Minireview: Challenges and opportunities in development of PPAR agonists. Mol Endocrinol 2014; 28:1756-68. [PMID: 25148456 PMCID: PMC5414793 DOI: 10.1210/me.2013-1427] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 08/08/2014] [Indexed: 01/06/2023] Open
Abstract
The clinical impact of the fibrate and thiazolidinedione drugs on dyslipidemia and diabetes is driven mainly through activation of two transcription factors, peroxisome proliferator-activated receptors (PPAR)-α and PPAR-γ. However, substantial differences exist in the therapeutic and side-effect profiles of specific drugs. This has been attributed primarily to the complexity of drug-target complexes that involve many coregulatory proteins in the context of specific target gene promoters. Recent data have revealed that some PPAR ligands interact with other non-PPAR targets. Here we review concepts used to develop new agents that preferentially modulate transcriptional complex assembly, target more than one PPAR receptor simultaneously, or act as partial agonists. We highlight newly described on-target mechanisms of PPAR regulation including phosphorylation and nongenomic regulation. We briefly describe the recently discovered non-PPAR protein targets of thiazolidinediones, mitoNEET, and mTOT. Finally, we summarize the contributions of on- and off-target actions to select therapeutic and side effects of PPAR ligands including insulin sensitivity, cardiovascular actions, inflammation, and carcinogenicity.
Collapse
Affiliation(s)
- Matthew B Wright
- F. Hoffmann-La Roche Pharmaceuticals (M.B.W., M.Bor., M.Bop.), CH-4070 Basel, Switzerland; and MediTech Media (M.T.), London EC1V 9AZ, United Kingdom
| | | | | | | |
Collapse
|
13
|
Ninomiya I, Yamazaki K, Oyama K, Hayashi H, Tajima H, Kitagawa H, Fushida S, Fujimura T, Ohta T. Pioglitazone inhibits the proliferation and metastasis of human pancreatic cancer cells. Oncol Lett 2014; 8:2709-2714. [PMID: 25364454 PMCID: PMC4214501 DOI: 10.3892/ol.2014.2553] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 09/08/2014] [Indexed: 01/03/2023] Open
Abstract
Proliferator-activated receptor-γ (PPAR-γ) is a nuclear receptor that acts as a transcription factor in several types of tissue. PPAR-γ ligands are known to inhibit numerous cancer cell processes, including pancreatic cancer cell proliferation through terminal differentiation. Previous studies concerning the inhibitory effect of PPAR-γ ligands derived from thiazolidinediones (TZDs) on the metastatic potential of cancer cells have been reported. The present study aimed to investigate whether pioglitazone, a prescription TZD class drug and a ligand of PPAR-γ, inhibits the proliferation and metastasis of pancreatic cancer cells. The inhibitory effect of pioglitazone on the proliferation of the Capan-1, Aspc-1, BxPC-3, PANC-1 and MIApaCa-2 pancreatic cancer cell lines was analyzed. Alterations in carcinoembryonic antigen (CEA), interleukin-8 (IL-8) and cyclooxygenase-2 (COX-2) mRNA expression levels subsequent to pioglitazone treatment were examined in BxPC-3 cells by quantitative reverse transcription polymerase chain reaction. In addition, whether the oral administration of pioglitazone prevents tumorigenesis and spontaneous BxPC-3 cell lymph node and lung metastases was investigated using a rectal xenograft model. Pioglitazone treatment resulted in the inhibition of proliferation in all five pancreatic cancer cell lines in vitro. Pioglitazone induced CEA mRNA expression, suppressed IL-8 and COX-2 mRNA expression in vitro, and inhibited BxPC-3 xenograft growth. Pioglitazone also reduced BxPC-3 cell lymph node and lung metastasis in the rectal xenograft model. These results suggest that pioglitazone treatment inhibited the proliferation and metastasis of pancreatic cancer cells through the induction of differentiation and the inhibition of angiogenesis-associated protein expression.
Collapse
Affiliation(s)
- Itasu Ninomiya
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Keisuke Yamazaki
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Katsunobu Oyama
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hironori Hayashi
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hidehiro Tajima
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hirohisa Kitagawa
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Sachio Fushida
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takashi Fujimura
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Tetsuo Ohta
- Gastroenterologic Surgery, Department of Oncology, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
14
|
Lee HS, Chang M, Lee JE, Kim W, Hwang IC, Kim DH, Park HK, Choi HJ, Jo W, Cha SW, Son WC. Carcinogenicity study of CKD-501, a novel dual peroxisome proliferator-activated receptors α and γ agonist, following oral administration to Sprague Dawley rats for 94-101 weeks. Regul Toxicol Pharmacol 2014; 69:207-16. [PMID: 24747398 DOI: 10.1016/j.yrtph.2014.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 11/26/2022]
Abstract
CKD-501 is a peroxisome proliferator-activated receptor (PPAR) agonist. The current study was conducted in Sprague Dawley (SD) rats for 94-101 weeks to investigate the carcinogenic potential of CKD-501. 60 males received 0, 0.03, 0.12, or 1.0mg/kg/day, which was changed after 66 weeks to 0.24 mg/kg/day due to increased mortality, while 60 females received 0, 0.03, 0.06, or 0.12 mg/kg/day throughout the study period. After switching the dosage, no significant changes in the survival rates were observed. Non-neoplastic lesions such as bladder transitional cell hyperplasia and a diminished corpus luteum were observed in females administered 0.12 mg/kg/day and the right chamber dilation and left ventricular hypertrophy were increased dose dependently in both males and females. Non-neoplastic lesions such as bone marrow hypoplasia and fat cell proliferation and neoplastic lesions such as lipomas and liposarcomas observed in males and/or females were considered expected pharmacological effects for this compound. Compared to rosiglitazone, CKD-501 had a 4.4-fold higher margin of safety for tumor induction and did not cause bladder carcinoma as was observed with pioglitazone.
Collapse
Affiliation(s)
- Hee Su Lee
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Minsun Chang
- Department of Medical and Pharmaceutical Science, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ji-Eun Lee
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea; College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Woojin Kim
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - In-Chang Hwang
- Chong Kun Dang Research Institute, CKD Pharmaceuticals Inc., Kyunggi-do, Republic of Korea
| | - Dal-Hyun Kim
- Chong Kun Dang Research Institute, CKD Pharmaceuticals Inc., Kyunggi-do, Republic of Korea
| | - Hyun-Kyu Park
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyun-Ji Choi
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Woori Jo
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Shin-Woo Cha
- Division of Non-clinical Studies, Korea Institute of Toxicology (KIT), Daejeon, Republic of Korea
| | - Woo-Chan Son
- Department of Pathology, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Moon KS, Lee JE, Lee HS, Hwang IC, Kim DH, Park HK, Choi HJ, Jo W, Son WC, Yun HI. CKD-501, a novel selective PPARγ agonist, shows no carcinogenic potential in ICR mice following oral administration for 104 weeks. J Appl Toxicol 2013; 34:1271-84. [PMID: 24026970 DOI: 10.1002/jat.2918] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/11/2013] [Accepted: 07/11/2013] [Indexed: 12/31/2022]
Abstract
CKD-501 is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist that is effective for the treatment of diabetes. However, its carcinogenic potential remains controversial. The current carcinogenicity study was conducted over a period of 104 weeks in ICR mice. Three groups, each consisting of 60 male and 60 female mice, received oral CKD-501 dosages of 0.2, 1.0 or 6.0 mg kg(-1) day(-1). The mortality rates of the male control, 0.2, 1.0 and 6.0 mg kg(-1) day(-1) treated groups were 60%, 68%, 58% and 67%, respectively and 57%, 68% and 67% in the female control, 0.2 and 1.0 mg kg(-1) day(-1) treated groups. It was 67% in the female 6.0 mg kg(-1) day(-1) treated group, which was terminated at week 98 due to its increased mortality rate. No significant treatment-related effects were observed on the survival rates, with the exception of females in the 6.0 mg kg(-1) day(-1) group. Body weights increased in females receiving 1.0 and 6.0 mg kg(-1) day(-1) due to the class effects of the PPARγ agonist. Differences were not found in hematology parameters between the CKD-501-treated groups and their corresponding controls, but the histopathological evidence did not reveal any findings attributed to CKD-501. Treated animals exhibited non-neoplastic findings (adipocyte proliferation, bone marrow hypoplasia cardiomyopathy), but all of these were expected changes for this class of compound. There were no treatment-related neoplastic changes in this study. The results of this study therefore demonstrate a lack of carcinogenicity following oral administration of CKD-501 to ICR mice for 104 weeks.
Collapse
Affiliation(s)
- Kyoung-Sik Moon
- Division of Non-clinical Studies, Korea Institute of Toxicology (KIT), Daejeon, Korea; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kim SY. Letter: the risk of bladder cancer in korean diabetic subjects treated with pioglitazone (diabetes metab j 2012;36:371-8). Diabetes Metab J 2012; 36:460-1. [PMID: 23275940 PMCID: PMC3530717 DOI: 10.4093/dmj.2012.36.6.460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Sang-Yong Kim
- Division of Endocrinology and Metabolism, Chosun University School of Medicine, Gwangju, Korea
| |
Collapse
|
17
|
Bortolini M, Wright MB, Bopst M, Balas B. Examining the safety of PPAR agonists - current trends and future prospects. Expert Opin Drug Saf 2012; 12:65-79. [PMID: 23134541 DOI: 10.1517/14740338.2013.741585] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The peroxisome proliferator-activated receptor (PPAR)-α and -γ agonists, fibrates and glitazones, are effective treatments for dyslipidemia and type 2 diabetes mellitus, respectively, but exhibit class-related, as well as compound-specific safety characteristics. AREAS COVERED This article reviews the profiles of PPAR-α, PPAR-γ, and dual PPAR-α/γ agonists with regard to class-related and compound-specific efficacy and adverse effects. We explore how learnings from first-generation drugs are being applied to develop safer PPAR-targeted therapies. EXPERT OPINION The finding that rosiglitazone may increase risk for cardiovascular events has led to regulatory guidelines requiring demonstration of cardiovascular safety in appropriate outcome trials for new type 2 diabetes mellitus drugs. The emerging data on the possibly increased risk of bladder cancer with pioglitazone may prompt the need for post-approval safety studies for new drugs. Since PPAR-α and -γ affect key cardiometabolic risk factors (diabetic dyslipidemia, insulin resistance, hyperglycemia, and inflammation) in a complementary fashion, combining their benefits has emerged as a particularly attractive option. New PPAR-targeted therapies that balance the relative potency and/or activity toward PPAR-α and -γ have shown promise in retaining efficacy while reducing potential side effects.
Collapse
Affiliation(s)
- Michele Bortolini
- Cardiometabolic Franchise Safety Science Leader, F. Hoffmann-La Roche, Ltd, PDS-Safety Risk Management, 663/2028, CH4070 Basel, Switzerland.
| | | | | | | |
Collapse
|
18
|
Zhu Z, Shen Z, Lu Y, Zhong S, Xu C. Increased risk of bladder cancer with pioglitazone therapy in patients with diabetes: a meta-analysis. Diabetes Res Clin Pract 2012; 98:159-63. [PMID: 22705039 DOI: 10.1016/j.diabres.2012.05.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/20/2012] [Accepted: 05/03/2012] [Indexed: 11/24/2022]
Abstract
AIMS Emerging studies suggest a possible increased risk of bladder cancer with pioglitazone therapy. We therefore pooled data available to examine the association between pioglitazone therapy and bladder cancer in patients with diabetes. METHODS We searched Medline and Embase to identify studies that reported the effect of pioglitazone on bladder cancer among diabetic patients. Summary effect estimates were derived using a fixed-effects meta-analysis model. RESULTS Five studies included 2,350,908 diabetic patients. Pioglitazone was associated with a significantly higher risk of bladder cancer (relative risk [RR] 1.17, 95% confidence interval (CI) 1.03-1.32, P=0.013). No relation between pioglitazone and bladder cancer was found for duration of therapy <12 months and cumulative dose <28,000 mg. The RR for bladder cancer in subjects with 12-24 months of pioglitazone use was 1.34 (95% CI 1.08-1.66, P=0.008). The effect was even stronger for cumulative treatment duration >24 months (RR 1.38, 95% CI 1.12-1.70, P=0.003). There was a significant risk for patients with cumulative dose >28,000 mg (RR 1.58, 95% CI 1.12-2.06, P=0.001). CONCLUSIONS Pioglitazone treatment appears to be associated with a significantly increased risk of bladder cancer in patients with diabetes.
Collapse
Affiliation(s)
- Zhaowei Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
19
|
Sandes EO, Lodillinsky C, Langle Y, Belgorosky D, Marino L, Gimenez L, Casabé AR, Eiján AM. Inducible nitric oxide synthase and PPARγ are involved in bladder cancer progression. J Urol 2012; 188:967-73. [PMID: 22819108 DOI: 10.1016/j.juro.2012.04.099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Indexed: 10/28/2022]
Abstract
PURPOSE We evaluated the role of inducible nitric oxide synthase and PPARγ as prognostic factors for bladder cancer. MATERIALS AND METHODS Inducible nitric oxide synthase and PPARγ were evaluated by Western blot and immunohistochemistry in a mouse bladder cancer model of nonmuscle invasive and invasive MB49-I tumor cells, and in human bladder cancer samples. RESULTS Inducible nitric oxide synthase expression was negative in mouse normal urothelium and higher in invasive than in noninvasive MB49 tumors. In vitro inducible nitric oxide synthase activity, determined as nitrite, was higher in MB49-I than in MB49 cells (p <0.001). In human samples expression was also associated with tumor invasion. Nuclear PPARγ expression was negative in normal mouse urothelium but higher in nonmuscle invasive MB49 than in MB49-I tumors. Similarly in human tumors low PPARγ was associated with poor prognosis factors, such as high histological grade (p = 0.0160) and invasion status (p = 0.0352). A positive correlation was noted between inducible nitric oxide synthase and PPARγ in low histological grade and nonmuscle invasive tumors (Pearson correlation index 0.6368, p = 0.0351, 0.4438 and 0.0168, respectively). As determined by gene reporter assay, PPARγ activity was induced by nitric oxide only in nonmuscle invasive MB49 cells (p <0.001). CONCLUSIONS Results suggest that increased PPARγ controls inducible nitric oxide synthase expression at early tumor stages. However, regulation is lost at advanced tumor stages, when the increase in inducible nitric oxide synthase and the decrease in PPARγ seem to be associated with bladder cancer progression.
Collapse
Affiliation(s)
- Eduardo Omar Sandes
- Research Area, Institute of Oncology Angel H. Roffo, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Pruimboom-Brees IM, Francone O, Pettersen JC, Kerlin RL, Will Y, Amacher DE, Boucher GG, Morton D. The development of subcutaneous sarcomas in rodents exposed to peroxisome proliferators agonists: hypothetical mechanisms of action and de-risking attitude. Toxicol Pathol 2012; 40:810-8. [PMID: 22504321 DOI: 10.1177/0192623312441406] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) represent therapeutic targets for the management of type 2 diabetes mellitus and dyslipidemia. Rodent carcinogenicity studies have revealed a link between γ and dual γ/α PPAR agonist treatment and the increased incidence of subcutaneous (SC) liposarcomas/fibrosarcomas or hemangiosarcomas, but very little has been reported for potent and selective PPARα agonists. We present a mode of action framework for the development of SC mesenchymal tumors in rodents given PPAR agonists. (1) Tumor promotion results from pharmacologically mediated recruitment (proliferation and differentiation), thermogenesis and adipogenesis of stromovascular cells, and subsequent generation of oxidative free radicals. (2) Tumor initiation consists of chemotype-driven mitochondrial dysfunction causing uncontrolled oxidative stress and permanent DNA damage. Promotion is characterized by enhanced adipogenesis in the SC adipose tissue, where the baseline PPARγ expression and responsiveness to PPARγ ligands is the highest, and by thermogenesis through expression of the uncoupling protein 1 (UCP-1) and the PPARγ co-activator 1 α (PGC-1α), two factors more highly expressed in brown versus white adipose tissue. Initiation is supported by the demonstration of mitochondrial uncoupling and OXPHOS Complexes dysfunction (Complexes III, IV and V) by compounds associated with increased incidences of sarcomas (muraglitazar and troglitazone), but not others lacking malignant tumor effects (pioglitazone, rosiglitazone).
Collapse
|
21
|
Tseng CH, Tseng FH. Peroxisome proliferator-activated receptor agonists and bladder cancer: lessons from animal studies. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2012; 30:368-402. [PMID: 23167631 DOI: 10.1080/10590501.2012.735519] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This article reviews available animal studies on the possible link between the use of peroxisome proliferator-activated receptor (PPAR) agonists and bladder cancer, with further discussion on the possible implications to humans. Carcinogenicity studies suggest that the PPARγ agonist pioglitazone and dual PPARα/γ agonists such as ragaglitazar, muraglitazar, and naveglitazar may increase the risk of bladder cancer in a dose-responsive pattern in rats. It is interesting that bladder cancer related to PPAR agonists shows remarkable species- and sex-specificity and has a predilection to occur in the ventral dome of bladder in rodents. While male rats treated with pioglitazone or muraglitazar have a higher propensity to develop bladder cancer than female rats, mice of both sexes do not develop bladder cancer even when exposed to very high doses. Direct genotoxicity or cytotoxicity of PPAR agonists is unlikely to be the mode of action because most of the parent compounds or their metabolites of the PPAR agonists are neither mutagenic nor genotoxic, and they are rarely excreted in the urine; but a receptor-mediated PPAR effect cannot be excluded. Some suggest a "urolithiasis hypothesis" referring to the formation of urinary solids and calculi, which subsequently causes bladder necrosis, regenerative proliferation, hypertrophy, and cancer. However, whether these animal findings could have human relevance is not yet fully understood. Some argue that the urolithiasis-induced bladder cancer might be rat-specific and would probably not be applicable to humans. An effect of increased urinary growth factors induced by PPAR agonists has also been proposed, but this requires more investigations. Before fully clarified, a balance between the risks and benefits of the use of pioglitazone, an approved oral antidiabetic agent that has recently been linked to an increased but not yet confirmed risk of bladder cancer in humans, should be justified for individual use.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | | |
Collapse
|
22
|
van Tongeren S, Fagerland JA, Conner MW, Diegel K, Donnelly K, Grubor B, Lopez-Martinez A, Bolliger AP, Sharma A, Tannehill-Gregg S, Turner PV, Wancket LM. The Role of the Toxicologic Pathologist in the Biopharmaceutical Industry. Int J Toxicol 2011; 30:568-82. [DOI: 10.1177/1091581811413304] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Toxicologic pathologists contribute significantly to the development of new biopharmaceuticals, yet there is often a lack of awareness of this specialized role. As the members of multidisciplinary teams, toxicologic pathologists participate in all aspects of the drug development process. This review is part of an initiative by the Society of Toxicologic Pathology to educate scientists about toxicologic pathology and to attract junior scientists, veterinary students, and veterinarians into the field. We describe the role of toxicologic pathologists in identifying candidate agents, elucidating bioactive pathways, and evaluating efficacy and toxicity in preclinical animal models. Educational and specialized training requirements and the challenges of working in a global environment are discussed. The biopharmaceutical industry provides diverse, challenging, and rewarding career opportunities in toxicologic pathology. We hope that this review promotes understanding of the important role the toxicologic pathologist plays in drug development and encourages exploration of an important career option.
Collapse
Affiliation(s)
- Susan van Tongeren
- Nonclinical Drug Safety US, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | | | - Michael W. Conner
- Preclinical Safety Assessment, Theravance, Inc., South San Francisco, CA, USA
| | | | - Kevin Donnelly
- Discovery and Translational Services, Covance Laboratories, Greenfield, IN, USA
| | - Branka Grubor
- PCS Discovery Pathology, NIBRI-Novartis, Cambridge, MA, USA
| | | | | | - Alok Sharma
- Department of Pathology, Covance Laboratories Inc., Madison, WI, USA
| | | | - Patricia V. Turner
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Lyn M. Wancket
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
23
|
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ) exerts multiple functions in determination of cell fate, tissue metabolism, and host immunity. Two synthetic PPARγ ligands (rosiglitazone and pioglitazone) were approved for the therapy of type-2 diabetes mellitus and are expected to serve as novel cures for inflammatory diseases and cancer. However, PPARγ and its ligands exhibit a janus-face behaviour as tumor modulators in various systems, resulting in either tumor suppression or tumor promotion. This may be in part due to signaling crosstalk to the mitogen-activated protein kinase (MAPK) cascades. The genomic activity of PPARγ is modulated, in addition to ligand binding, by phosphorylation of a serine residue by MAPKs, such as extracellular signal-regulated protein kinases-1/2 (ERK-1/2), or by nucleocytoplasmic compartmentalization through the ERK activators MAPK kinases-1/2 (MEK-1/2). PPARγ ligands themselves activate the ERK cascade through nongenomic and often PPARγ-independent signaling. In the current review, we discuss the molecular mechanisms and physiological implications of the crosstalk of PPARγ with MEK-ERK signaling and its potential as a novel drug target for cancer therapy in patients.
Collapse
|
24
|
Rogue A, Renaud MP, Claude N, Guillouzo A, Spire C. Comparative gene expression profiles induced by PPARγ and PPARα/γ agonists in rat hepatocytes. Toxicol Appl Pharmacol 2011; 254:18-31. [PMID: 21515302 DOI: 10.1016/j.taap.2011.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/21/2011] [Accepted: 04/07/2011] [Indexed: 02/09/2023]
Abstract
Species-differential toxic effects have been described with PPARα and PPARγ agonists between rodent and human liver. PPARα agonists (fibrates) are potent hypocholesterolemic agents in humans while they induce peroxisome proliferation and tumors in rodent liver. By contrast, PPARγ agonists (glitazones) and even dual PPARα/γ agonists (glitazars) have caused idiosyncratic hepatic and nonhepatic toxicities in human without evidence of any damage in rodent during preclinical studies. The mechanisms involved in such differences remain largely unknown. Several studies have identified the major target genes of PPARα agonists in rodent liver while no comprehensive analysis has been performed on gene expression changes induced by PPARγ and dual PPARα/γ agonists. Here, we investigated transcriptomes of rat hepatocytes after 24h treatment with two PPARγ (troglitazone and rosiglitazone) and two PPARα/γ (muraglitazar and tesaglitazar) agonists. Although, hierarchical clustering revealed a gene expression profile characteristic of each PPAR agonist class, only a limited number of genes was specifically deregulated by glitazars. Functional analyses showed that many genes known as PPARα targets were also modulated by both PPARγ and PPARα/γ agonists and quantitative differences in gene expression profiles were observed between these two classes. Moreover, most major genes modulated in rat hepatocytes were also found to be deregulated in rat liver after tesaglitazar treatment. Taken altogether, these results support the conclusion that differential toxic effects of PPARα and PPARγ agonists in rodent liver do not result from transcriptional deregulation of major PPAR target genes but rather from qualitative and/or quantitative differential responses of a small subset of genes.
Collapse
Affiliation(s)
- Alexandra Rogue
- UMR INSERM U991, Faculté des Sciences Pharmaceutiques et Biologiques, Rennes, France
| | | | | | | | | |
Collapse
|
25
|
Jeong HW, Lee JW, Kim WS, Choe SS, Kim KH, Park HS, Shin HJ, Lee GY, Shin D, Lee H, Lee JH, Choi EB, Lee HK, Chung H, Park SB, Park KS, Kim HS, Ro S, Kim JB. A newly identified CG301269 improves lipid and glucose metabolism without body weight gain through activation of peroxisome proliferator-activated receptor alpha and gamma. Diabetes 2011; 60:496-506. [PMID: 21270261 PMCID: PMC3028349 DOI: 10.2337/db09-1145] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor (PPAR)-α/γ dual agonists have been developed to alleviate metabolic disorders. However, several PPARα/γ dual agonists are accompanied with unwanted side effects, including body weight gain, edema, and tissue failure. This study investigated the effects of a novel PPARα/γ dual agonist, CG301269, on metabolic disorders both in vitro and in vivo. RESEARCH DESIGN AND METHODS Function of CG301269 as a PPARα/γ dual agonist was assessed in vitro by luciferase reporter assay, mammalian one-hybrid assay, and analyses of PPAR target genes. In vitro profiles on fatty acid oxidation and inflammatory responses were acquired by fatty acid oxidation assay and quantitative (q)RT-PCR of proinflammatory genes. In vivo effect of CG301269 was examined in db/db mice. Total body weight and various tissue weights were measured, and hepatic lipid profiles were analyzed. Systemic glucose and insulin tolerance were measured, and the in vivo effect of CG301269 on metabolic genes and proinflammatory genes was examined by qRT-PCR. RESULTS CG301269 selectively stimulated the transcriptional activities of PPARα and PPARγ. CG301269 enhanced fatty acid oxidation in vitro and ameliorated insulin resistance and hyperlipidemia in vivo. In db/db mice, CG301269 reduced inflammatory responses and fatty liver, without body weight gain. CONCLUSIONS We demonstrate that CG301269 exhibits beneficial effects on glucose and lipid metabolism by simultaneous activation of both PPARα and PPARγ. Our data suggest that CG301269 would be a potential lead compound against obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Hyun Woo Jeong
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Joo-Won Lee
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Woo Sik Kim
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Sung Sik Choe
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Kyung-Hee Kim
- Division of Cardiology, Seoul National University College of Medicine, Seoul, Korea
| | - Ho Seon Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jung Shin
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Gha Young Lee
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
| | - Dongkyu Shin
- Crystal Genomics, Seoul National University, Seoul, Korea
| | - Hanjae Lee
- Department of Chemistry, Seoul National University, Seoul, Korea
| | - Jun Hee Lee
- Crystal Genomics, Seoul National University, Seoul, Korea
| | - Eun Bok Choi
- Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Hyeon Kyu Lee
- Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Heekyoung Chung
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
| | - Seung Bum Park
- Department of Chemistry, Seoul National University, Seoul, Korea
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea
| | - Kyong Soo Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo-Soo Kim
- Division of Cardiology, Seoul National University College of Medicine, Seoul, Korea
| | - Seonggu Ro
- Crystal Genomics, Seoul National University, Seoul, Korea
| | - Jae Bum Kim
- School of Biological Sciences, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, Korea
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea
- Corresponding author: Jae Bum Kim,
| |
Collapse
|
26
|
Glinghammar B, Berg AL, Bjurström S, Stockling K, Blomgren B, Westerberg R, Skånberg I, Hellmold H, Andersson U. Proliferative and molecular effects of the dual PPARalpha/gamma agonist tesaglitazar in rat adipose tissues: relevance for induction of fibrosarcoma. Toxicol Pathol 2011; 39:325-36. [PMID: 21270424 DOI: 10.1177/0192623310394210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The dual peroxisome-proliferator-activated receptor (PPAR) α/γ agonist tesaglitazar has been shown to produce fibrosarcomas in rats. Here, the authors studied morphology, proliferation, differentiation, and inflammation markers in adipose tissue from rats exposed to 1, 3, or 10 µmol/kg tesaglitazar for 2 or 12 weeks, including recovery groups (12 weeks treatment followed by 12 weeks recovery), and 3 or 10 µmol/kg tesaglitazar for 24 weeks. Subcutaneous white and brown fat revealed reversible dose-related histopathological alterations and after 12 and 24 weeks developed areas of thickened skin (fatty lumps). There was a dose-dependent increase in proliferation of interstitial cells in white and brown fat as shown by increased mitotic index in all dose groups after 2 weeks. This was limited to the high dose after 12 and 24 weeks in white fat. Gene expression analyses showed that while tesaglitazar induced differentiation of adipose tissue characterized with a switch in cyclin D1 and D3 mRNA by 12 weeks, longer exposure at high doses reversed this differentiation concurrent with a reappearance of early adipocyte and inflammatory markers. These data suggest that sustained increased turnover of mesenchymal cells in adipose tissues, concomitant with onset of inflammation and fibrosis, drives development of fibrosarcomas in rats treated with tesaglitazar.
Collapse
Affiliation(s)
- Björn Glinghammar
- Safety Assessment, Molecular Toxicology, AstraZeneca R&D, Södertälje, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ettlin RA, Kuroda J, Plassmann S, Hayashi M, Prentice DE. Successful drug development despite adverse preclinical findings part 2: examples. J Toxicol Pathol 2010; 23:213-34. [PMID: 22272032 PMCID: PMC3234630 DOI: 10.1293/tox.23.213] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 09/06/2010] [Indexed: 12/14/2022] Open
Abstract
To illustrate the process of addressing adverse preclinical findings (APFs) as
outlined in the first part of this review, a number of cases with unexpected APF
in toxicity studies with drug candidates is discussed in this second part. The
emphasis is on risk characterization, especially regarding the mode of action
(MoA), and risk evaluation regarding relevance for man. While severe APFs such
as retinal toxicity may turn out to be of little human relevance, minor findings
particularly in early toxicity studies, such as vasculitis, may later pose a
real problem. Rodents are imperfect models for endocrine APFs, non-rodents for
human cardiac effects. Liver and kidney toxicities are frequent, but they can
often be monitored in man and do not necessarily result in early termination of
drug candidates. Novel findings such as the unusual lesions in the
gastrointestinal tract and the bones presented in this review can be difficult
to explain. It will be shown that well known issues such as phospholipidosis and
carcinogenicity by agonists of peroxisome proliferator-activated receptors
(PPAR) need to be evaluated on a case-by-case basis. The latter is of particular
interest because the new PPAR α and dual α/γ agonists resulted in a change of
the safety paradigm established with the older PPAR α agonists. General
toxicologists and pathologists need some understanding of the principles of
genotoxicity and reproductive toxicity testing. Both types of preclinical
toxicities are major APF and clinical monitoring is difficult, generally leading
to permanent use restrictions.
Collapse
Affiliation(s)
- Robert A. Ettlin
- Ettlin Consulting Ltd., 14 Mittelweg, 4142 Muenchenstein,
Switzerland
| | - Junji Kuroda
- KISSEI Pharmaceutical Co., Ltd., 2320-1 Maki, Hotaka, Azumino,
Nagano 399-8305, Japan
| | - Stephanie Plassmann
- PreClinical Safety (PCS) Consultants Ltd., 7 Gartenstrasse, 4132
Muttenz, Switzerland
| | - Makoto Hayashi
- Biosafety Research Center, Foods, Drugs, and Pesticides (BSRC),
582-2 Shioshinden, Iwata, Shizuoka 437-1213, Japan
| | - David E. Prentice
- PreClinical Safety (PCS) Consultants Ltd., 7 Gartenstrasse, 4132
Muttenz, Switzerland
| |
Collapse
|
28
|
Jeong HW, Lee JW, Kim WS, Choe SS, Shin HJ, Lee GY, Shin D, Lee JH, Choi EB, Lee HK, Yon GH, Cho B, Kim HR, Choi SH, Chung YS, Park SB, Chung H, Ro S, Kim JB. A nonthiazolidinedione peroxisome proliferator-activated receptor α/γ dual agonist CG301360 alleviates insulin resistance and lipid dysregulation in db/db mice. Mol Pharmacol 2010; 78:877-85. [PMID: 20724462 DOI: 10.1124/mol.110.065748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Activation of peroxisome proliferator-activated receptors (PPARs) have been implicated in the treatment of metabolic disorders with different mechanisms; PPARα agonists promote fatty acid oxidation and reduce hyperlipidemia, whereas PPARγ agonists regulate lipid redistribution from visceral fat to subcutaneous fat and enhance insulin sensitivity. To achieve combined benefits from activated PPARs on lipid metabolism and insulin sensitivity, a number of PPARα/γ dual agonists have been developed. However, several adverse effects such as weight gain and organ failure of PPARα/γ dual agonists have been reported. By use of virtual ligand screening, we identified and characterized a novel PPARα/γ dual agonist, (R)-1-(4-(2-(5-methyl-2-p-tolyloxazol-4-yl)ethoxy)benzyl)piperidine-2-carboxylic acid (CG301360), exhibiting the improvement in insulin sensitivity and lipid metabolism. CG301360 selectively stimulated transcriptional activities of PPARα and PPARγ and induced expression of their target genes in a PPARα- and PPARγ-dependent manner. In cultured cells, CG301360 enhanced fatty acid oxidation and glucose uptake and it reduced pro-inflammatory gene expression. In db/db mice, CG301360 also restored insulin sensitivity and lipid homeostasis. Collectively, these data suggest that CG301360 would be a novel PPARα/γ agonist, which might be a potential lead compound to develop against insulin resistance and hyperlipidemia.
Collapse
Affiliation(s)
- Hyun Woo Jeong
- School of Biological Sciences, Seoul National University, Kwanak-Gu, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Long GG. Commentary on “Evaluation of Possible Carcinogenic Risk to Humans Based on Liver Tumors in Rodent Assays. Toxicol Pathol 2010; 38:502-5. [DOI: 10.1177/0192623309359795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Gerald G. Long
- Eli Lilly and Company, Lilly Research Laboratories, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Suzuki S, Arnold LL, Pennington KL, Kakiuchi-Kiyota S, Wei M, Wanibuchi H, Cohen SM. Effects of Pioglitazone, a Peroxisome Proliferator–Activated Receptor Gamma Agonist, on the Urine and Urothelium of the Rat. Toxicol Sci 2009; 113:349-57. [DOI: 10.1093/toxsci/kfp256] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Long GG, Reynolds VL, Dochterman LW, Ryan TE. Neoplastic and Non-neoplastic Changes in F-344 Rats Treated with Naveglitazar, a γ-Dominant PPAR α/γ Agonist. Toxicol Pathol 2009; 37:741-53. [DOI: 10.1177/0192623309343775] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The carcinogenic potential of naveglitazar, a γ-dominant peroxisome proliferator-activated receptor (PPAR) α/γ dual agonist, was evaluated in a two-year study in F344 rats (0, 0.3, 1.0, or 3.0 mg/kg, males; 0, 0.1, 0.3, or 1.0 mg/kg, females). Increased mortality in male rats of the high-dose group was related to cardiac-associated lesions, neoplasms, and undetermined causes. Degeneration and hypertrophy of the myocardium occurred with dose-responsive increased incidence and severity. Neoplasms with increased incidence included sarcomas in male rats and urinary bladder neoplasms in female rats. Most sarcomas in male rats occurred in the adipose tissue of the subcutis and were diagnosed as fibrosarcomas, with fewer liposarcomas and other histologic types. Non-neoplastic changes in adipose tissue included expansion of adipose tissue in multiple sites, alterations in cytoplasmic vesicular pattern in brown and white fat, increases in stroma and mesenchymal cells, and fibrosis. The severity of chronic progressive nephropathy was decreased in a dose-responsive manner in males, and hyperplasia and neoplasia of the mammary gland were decreased in incidence in females. The adverse effects of cardiotoxicity and increased incidence of neoplasms occurred with dose-responsive incidence and/or severity, and a no-effect level for these effects was not achieved in this study.
Collapse
Affiliation(s)
- Gerald G. Long
- Lilly Research Laboratories, Indianapolis, IN 46225, USA
| | | | | | | |
Collapse
|
32
|
Cohen SM, Storer RD, Criswell KA, Doerrer NG, Dellarco VL, Pegg DG, Wojcinski ZW, Malarkey DE, Jacobs AC, Klaunig JE, Swenberg JA, Cook JC. Hemangiosarcoma in rodents: mode-of-action evaluation and human relevance. Toxicol Sci 2009; 111:4-18. [PMID: 19525443 DOI: 10.1093/toxsci/kfp131] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although rarely occurring in humans, hemangiosarcomas (HS) have become important in evaluating the potential human risk of several chemicals, including industrial, agricultural, and pharmaceutical agents. Spontaneous HS arise frequently in mice, less commonly in rats, and frequently in numerous breeds of dogs. This review explores knowledge gaps and uncertainties related to the mode of action (MOA) for the induction of HS in rodents, and evaluates the potential relevance for human risk. For genotoxic chemicals (vinyl chloride and thorotrast), significant information is available concerning the MOA. In contrast, numerous chemicals produce HS in rodents by nongenotoxic, proliferative mechanisms. An overall framework is presented, including direct and indirect actions on endothelial cells, paracrine effects in local tissues, activation of bone marrow endothelial precursor cells, and tissue hypoxia. Numerous obstacles are identified in investigations into the MOA for mouse HS and the relevance of the mouse tumors to humans, including lack of identifiable precursor lesions, usually late occurrence of the tumors, and complexities of endothelial biology. This review proposes a working MOA for HS induced by nongenotoxic compounds that can guide future research in this area. Importantly, a common MOA appears to exist for the nongenotoxic induction of HS, where there appears to be a convergence of multiple initiating events (e.g., hemolysis, decreased respiration, adipocyte growth) leading to either dysregulated angiogenesis and/or erythropoiesis that results from hypoxia and macrophage activation. These later events lead to the release of angiogenic growth factors and cytokines that stimulate endothelial cell proliferation, which, if sustained, provide the milieu that can lead to HS formation.
Collapse
Affiliation(s)
- Samuel M Cohen
- University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kim MK, Chae YN, Kim HS, Choi SH, Son MH, Kim SH, Kim JK, Moon HS, Park SK, Shin YA, Kim JG, Lee CH, Lim JI, Shin CY. PAR-1622 is a selective peroxisome proliferator-activated receptor gamma partial activator with preserved antidiabetic efficacy and broader safety profile for fluid retention. Arch Pharm Res 2009; 32:721-7. [PMID: 19471887 DOI: 10.1007/s12272-009-1511-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 02/26/2009] [Accepted: 03/09/2009] [Indexed: 01/27/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) gamma is known to be a key regulator of insulin resistance. PAR-1622 is a novel small molecule compound synthesized in Dong-A research center. In this study, we characterized the pharmacological profiles of PAR-1622, a selective partial activator of PPARgamma. In transient transactivation assays, PAR-1622 [(S)-2-ethoxy-3(4-(5-(4-(5-(methoxymethyl)isoxazol-3-yl)phenyl)-3-methylthiophen-2-yl)methoxy)phenyl)propanoic acid] showed a partial activator against human PPARgamma with an EC(50) of 41 nM and a maximal response of 37% relative to the full agonist rosiglitazone without activating human PPARdelta. PAR-1622 was 56 folds more selective for human PPARgamma than for human PPARalpha (EC(50), 2304 nM), which means that it is a selective partial activator of PPARgamma. PAR-1622 also showed a partial activator against mouse PPARgamma with an EC(50) of 427 nM and a maximal response was 57% of that of rosiglitazone. INT-131, a selective PPARgamma partial agonist in clinical stage, also was a partial activator against human PPARgamma with an EC(50) of 83 nM and a maximal response achieved by INT-131 was 49% of that observed with full agonist rosiglitazone. In functional assays using human mesenchymal stem cells, PAR-1622 induced adipocyte differentiation, which was 3-fold more potent with a comparable maximum response compared to INT-131. Furthermore, PAR-1622 significantly improved hyperglycemia in db/db when orally administered at a dose of 1 mg/kg/day for 5 days. In hemodilution assays with Evans Blue, rosiglitazone significantly increased the plasma volume in ICR mice that were orally administered 30 mg/kg/day for 9 days; however, PAR-1622 showed no significant effects on plasma volume, similar to INT-131. These results suggest that PAR-1622 is a selective partial activator of PPARgamma and has excellent antihyperglycemic activities and a broad safety profile for fluid retention.
Collapse
Affiliation(s)
- Mi-Kyung Kim
- Dong-A Research Center, 47-5 Sanggal-dong, Giheung-gu, Yongin-si, Gyeonggi-do, 446-905, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tannehill-Gregg SH, Dominick MA, Reisinger AJ, Moehlenkamp JD, Waites CR, Stock DA, Sanderson TP, Cohen SM, Arnold LL, Schilling BE. Strain-related Differences in Urine Composition of Male Rats of Potential Relevance to Urolithiasis. Toxicol Pathol 2009; 37:293-305. [DOI: 10.1177/0192623309332990] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In carcinogenicity studies with PPAR γ and α/γ agonists, urinary bladder tumors have been reported in Harlan Sprague-Dawley (HSD) and Charles River Sprague-Dawley (SD) but not Wistar (WI) rats, with urolithiasis purported to be the inciting event. In two 3-month studies, the authors investigated strain-related differences in urine composition by sampling urine multiple times daily. Urine pH, electrolytes, creatinine, protein, citrate and oxalate levels, and serum citrate were assessed; urine sediment was analyzed by scanning electron microscopy and energy dispersive x-ray spectroscopy. HSD rats had significantly higher urine calcium than SD or WI rats, primarily as calcium phosphate-containing precipitate. When compared to SD rats, HSD rats had lower urine volume, higher urine protein, and a comparable (week 4) to lower (week 13) burden of MgNH4PO4 aggregates. Relative to WI rats, HSD rats had higher urine protein and magnesium and lower serum and urine citrate. Overall, the susceptibility to urolithiasis in male rats was HSD > SD > WI; this was likely due to strain-related differences in the amount of urine protein (a nidus for crystal formation), lithogenic ions, citrate (an inhibitor of lithogenesis), and/or volume. Strain-related differences in urine composition need to be considered when interpreting the outcome of studies with compounds that alter urine composition.
Collapse
Affiliation(s)
- Sarah H. Tannehill-Gregg
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| | - Mark A. Dominick
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| | - Amy J. Reisinger
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| | - Jeffrey D. Moehlenkamp
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| | - C. Robbie Waites
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
- SABIC Innovative Plastics, Mount Vernon, Indiana, USA
| | - David A. Stock
- Bristol-Myers Squibb Research & Development, Department of Global Biometric Sciences, Wallingford, Connecticut, USA
| | - Thomas P. Sanderson
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| | - Samuel M. Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Lora L. Arnold
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Beth E. Schilling
- Bristol-Myers Squibb Research & Development, Department of Drug Safety Evaluation, Mount Vernon, Indiana, USA
| |
Collapse
|
35
|
Chopra B, Hinley J, Oleksiewicz MB, Southgate J. Trans-Species Comparison of PPAR and RXR Expression by Rat and Human Urothelial Tissues. Toxicol Pathol 2008; 36:485-95. [DOI: 10.1177/0192623308315672] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Because some investigational peroxisome proliferator-activated receptors (PPAR) agonists cause tumors in the lower urinary tract of rats, we compared normal human and rat urothelium in terms of PPAR and retinoid X receptor (RXR) expression and proliferation-associated phenotypes. In situ, few human but most rat urothelial cells were Ki67 positive, indicating fundamental differences in cell cycle control. Rat and human urothelia expressed all 3 PPAR and the RXRα and RXRβ isoforms in a predominantly nuclear localization, indicating that they may be biologically active. However, immunolocalization differences were observed between species. First, whereas PPARα and PPARβ/δ were expressed throughout the human bladder or ureteric urothelium, in the rat urothelium PPARα was primarily, and PPARβ/δ exclusively, restricted to superficial cells. Second, RXRβ was restricted to intermediate and superficial layers of the human urothelium but tended to be absent from the rat superficial cells. Third, PPARγ expression was present throughout the urothelia of both species but was most intense in the superficial human urothelium. Species differences were also observed in the expression of PPAR and RXR isoforms between cultured rat and human urothelial cells and in the smooth muscle. Our findings highlight the unique coexpression of multiple PPAR and RXR isoforms by urothelium and suggest that species differences in PPAR function between rat and human urothelia may be explored in an in vitro setting.
Collapse
Affiliation(s)
- Bikramjit Chopra
- Jack Birch Unit of Molecular Carcinogenesis, Department of Biology, University of York, York, Y010 5YW, UK
| | - Jennifer Hinley
- Jack Birch Unit of Molecular Carcinogenesis, Department of Biology, University of York, York, Y010 5YW, UK
| | | | - Jennifer Southgate
- Jack Birch Unit of Molecular Carcinogenesis, Department of Biology, University of York, York, Y010 5YW, UK
| |
Collapse
|
36
|
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]
|
37
|
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: 2.1] [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
|
38
|
Aoki T. Current Status of Carcinogenicity Assessment of Peroxisome Proliferator-Activated Receptor Agonists by the US FDA and a Mode-of-Action Approach to the Carcinogenic Potential. J Toxicol Pathol 2007. [DOI: 10.1293/tox.20.197] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|