PT2385 is a first-in-class, selective small-molecule inhibitor of hypoxia-inducible factor-2alpha (HIF-2alpha) developed for the treatment of advanced clear cell renal cell carcinoma. Preclinical results demonstrated that PT2385 has potent antitumor efficacy in mouse xenograft models of kidney cancer. Recent studies revealed that PT2385 inhibits in the intestine and has efficacy in the treatment of diet induced obesity, insulin resistance and fatty liver disease. It also has activity toward metabolic disease in a mouse model. However, no metabolism data are currently publicly available. It is of great importance to characterize the metabolism of PT2385 and identify its effect on systemic homeostasis in mice. High-resolution mass spectrometry-based metabolomics was performed to profile the biotransformation of PT2385 and PT2385-induced changes in endogenous metabolites. Liver microsomes and recombinant drug-metabolizing enzymes were used to determine the mechanism of PT2385 metabolism. Real-time polymerase chain reaction analysis was employed to investigate the reason for the PT2385-induced bile acid dysregulation. A total of 12 metabolites of PT2385 was characterized, generated from hydroxylation (M1, M2), dihydroxylation and desaturation (M3, M4), oxidative-defluorination (M7), glucuronidation (M8), N-acetylcysteine conjugation (M9), and secondary methylation (M5, M6) and glucuronidation (M10, M11, and M12). CYP2C19 was the major contributor to the formation of M1, M2, and M7, UGT2B17 to M8, and UGT1A1/3 to M10-M12. The bile acid metabolites taurocholic acid and tauro-beta-muricholic acid were elevated in serum and liver of mice after PT2385 treatment. Gene expression analysis further revealed that intestinal HIF-2alpha inhibition by PT2385 treatment upregulated the hepatic expression of CYP7A1, the rate-limiting enzyme in bile acid synthesis. This study provides metabolic data and an important reference basis for the safety evaluation and rational clinical application of PT2385. Rutaecarpine (RUT), evodiamine (EOD) and dehydroevodiamine (DHED), are the three main bioactive indoloquinazoline alkaloids isolated from Euodia ruticarpa, a widely prescribed traditional Chinese medicine. These compounds have a variety of intriguing biological properties such as antitumor, antithrombotic, anti-inflammatory, analgesic, anti-obesity, anti-cholinesterase and anti-amnesic activities. The structure-activity relationships of these analogues for aryl hydrocarbon receptor (AHR) activation were explored by use of Ahr-deficient (Ahr-/-) mice, primary hepatocyte cultures, luciferase reporter gene assays, in silico ligand docking studies and metabolomics. In vitro, both mRNA analysis of AHR target genes in mouse primary hepatocytes and luciferase reporter assays in hepatocarcinoma cell lines demonstrated that RUT, EOD and DHED significantly activated AHR, with an efficacy order of RUT DHED EOD. Ligand-docking analysis predicted that the methyl substitute at the N-14 atom was a key factor affecting AHR activation. In vivo, EOD was poorly orally absorbed and failed to activate AHR, while RUT and DHED markedly upregulated the hepatic AHR battery gene expression in wild-type mice, but not in Ahr-/- mice. Furthermore, RUT, EOD and DHED were not hepatoxic at the doses employed. However, RUT and DHED disrupted bile acid homeostasis in an AHR-dependent manner. These findings revealed that the methyl group at the N-14 atom of these analogues and their pharmacokinetic behaviors were the main determinants for AHR activation. These data suggest that attention should be given to monitoring bile acid metabolism in the clinical use of Euodia ruticarpa. Triptolide, a major active constitute of Tripterygium wilfordii Hook. F, is prescribed for the treatment of autoimmune diseases in China. One of its most severe adverse effects observed in the clinical use is hepatotoxicity, but the mechanism is still unknown. Therefore, the present study applied an LC/MS-based metabolomic analysis to characterize the metabolomic changed in serum and liver induced by triptolide in mice. Mice were administered triptolide by gavage to establish the acute liver injury model, and serum biochemical and liver histological analysis were applied to assess the degree of toxicity. Multivariate data analyses were performed to investigate the metabolic alterations. Potential metabolites were identified using variable importance in the projection values and student's t-test. A total of 30 metabolites were observed that were significantly changed by triptolide treatment and the abundance of 29 metabolites were correlated with the severity of toxicity. Pathway analysis indicated that the mechanism of triptolide-induced hepatotoxicity was related to alterations in multiple metabolic pathways, including glutathione metabolism, tricarboxylic acid cycle, purine metabolism, glycerophospholipid metabolism, taurine and hypotaurine metabolism, pantothenate and CoA biosynthesis, pyrimidine metabolism and amino acids metabolism. The current study provides new mechanistic insights into the metabolic alterations that lead to triptolide-induced hepatotoxicity.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC005708-27
Application #
9779559
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
27
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Wang, Yongtao; Chen, Yixin; Guan, Lihuan et al. (2018) Carnitine palmitoyltransferase 1C regulates cancer cell senescence through mitochondria-associated metabolic reprograming. Cell Death Differ 25:733-746
Zhao, Jie; Xie, Cen; Mu, Xiyan et al. (2018) Metabolic alterations in triptolide-induced acute hepatotoxicity. Biomed Chromatogr 32:e4299
Qin, Zifei; Li, Shishi; Yao, Zhihong et al. (2018) Metabolic profiling of corylin in vivo and in vitro. J Pharm Biomed Anal 155:157-168
Qin, Zifei; Li, Shishi; Yao, Zhihong et al. (2018) Chemical inhibition and stable knock-down of efflux transporters leads to reduced glucuronidation of wushanicaritin in UGT1A1-overexpressing HeLa cells: the role of breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRPs Food Funct 9:1410-1423
Nair, Sneha G; Patel, Daxesh P; Gonzalez, Frank J et al. (2018) Simultaneous determination of etonogestrel and ethinyl estradiol in human plasma by UPLC-MS/MS and its pharmacokinetic study. Biomed Chromatogr 32:e4165
Hong, Xiaodan; Zheng, Yuanru; Qin, Zifei et al. (2017) In Vitro Glucuronidation of Wushanicaritin by Liver Microsomes, Intestine Microsomes and Expressed Human UDP-Glucuronosyltransferase Enzymes. Int J Mol Sci 18:
Zhao, Jin; Sun, Tao; Wu, Jing-Jing et al. (2017) Inhibition of human CYP3A4 and CYP3A5 enzymes by gomisin C and gomisin G, two lignan analogs derived from Schisandra chinensis. Fitoterapia 119:26-31
Yao, Pei-Li; Chen, Liping; Dobrza?ski, Tomasz P et al. (2017) Peroxisome proliferator-activated receptor-?/? inhibits human neuroblastoma cell tumorigenesis by inducing p53- and SOX2-mediated cell differentiation. Mol Carcinog 56:1472-1483
Pingili, Ajeeth K; Davidge, Karen N; Thirunavukkarasu, Shyamala et al. (2017) 2-Methoxyestradiol Reduces Angiotensin II-Induced Hypertension and Renal Dysfunction in Ovariectomized Female and Intact Male Mice. Hypertension 69:1104-1112
Zhao, Qi; Li, Xin-Mei; Liu, Hong-Ning et al. (2017) Metabolic map of osthole and its effect on lipids. Xenobiotica :1-15

Showing the most recent 10 out of 215 publications