1
|
Taheri H, Ahmed E, Hu P, Sparreboom A, Hu S. Determination and Disposition of the Aromatase Inhibitor Exemestane in CYP3A-Deficient Mice. Molecules 2025; 30:1440. [PMID: 40286062 PMCID: PMC11990089 DOI: 10.3390/molecules30071440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Exemestane, a steroidal aromatase inhibitor prescribed for post-menopausal women with estrogen receptor-positive breast cancer, is associated with debilitating musculoskeletal side effects and exhibits considerable interindividual variability in systemic exposure. Although exemestane is metabolized mainly by cytochrome P-450 3A4, the contribution of this metabolic pathway to the elimination of exemestane and its drug-drug interaction liabilities remains uncertain. Here, we developed a novel quantification method for exemestane and applied it to evaluate the role of CYP3A-mediated metabolism in the pharmacokinetics of exemestane using wild-type and Cyp3a-deficient mice. Liquid chromatography-mass spectrometry was used to quantify exemestane in selective reaction monitoring (SRM) mode, in which precursor ion and fragment ion data were obtained simultaneously. Validation results demonstrated that the developed method was accurate and precise, and sufficiently sensitive to be applied to murine pharmacokinetic studies involving serial blood sampling strategies. Although in vitro studies indicate that exemestane undergoes extensive metabolism in the liver to inactive metabolites by CYP3A4, complete Cyp3a deficiency in mice did not influence the systemic exposure to exemestane. This unequivocal evidence from genetic approaches using preclinical mouse models confirms that the potential for such DDI liabilities is very low. Our newly developed method provides a robust platform for further pharmacokinetic studies with exemestane in mice to delineate DDI liabilities and define the mechanisms of elimination.
Collapse
Affiliation(s)
| | | | | | | | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (H.T.); (E.A.); (P.H.); (A.S.)
| |
Collapse
|
2
|
Hoseinpour Kouhestany R, Tamaddon A, Ahmad Panahi H, Afshar Ebrahimi A, Amiri R. Hyper-branched nanodendrimer as a novel solid-phase extraction sorbent followed by three phase hollow fiber microextraction for trace separation of exemestane. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.121142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
3
|
Ishii T, Nojiri N, Mano Y. A simple UPLC-MS/MS assay with a core-shell column for the determination of exemestane in human plasma for clinical application. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2022; 28:94-103. [PMID: 36128912 DOI: 10.1177/14690667221126276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Exemestane is one of the aromatase inhibitors and has been used to treat breast cancer by lowering estrogen levels. Accurate quantification of exemestane is important to set an optimal dose, and thus a simple assay for exemestane is developed by ultra-performance liquid chromatography with tandem mass spectrometer. Exemestane was extracted from human plasma samples (100 μL) by simple protein precipitation with acetonitrile/methanol (1/1, v/v). Interference peaks observed close to the elution of exemestane led us to use a core shell column for higher selectivity instead of totally porous columns. The extracts were chromatographed on CORTECS UPLC C18, under a gradient elution at a flow rate of 0.25 mL/min and detected in the selected reaction monitoring. Validation parameters were assessed in accordance with the bioanalytical guidelines using quality control samples. Exemestane in human plasma was quantifiable from 0.5 to 50 ng/mL with high extraction recovery and minimal matrix effects. Hemolyzed or hyperlipemic plasma did not impact the exemestane assay. Exemestane was stable in human plasma for 392 days at -15°C or below. The developed assay was robust and successfully applied to quantifying exemestane concentrations in human plasma to support a clinical trial.
Collapse
Affiliation(s)
- Takuho Ishii
- DMPK & Bioanalysis Unit, Tsukuba R&D Supporting Division, 204948Sunplanet Co., Ltd, Tsukuba-shi, Japan
| | - Nana Nojiri
- DMPK & Bioanalysis Unit, Tsukuba R&D Supporting Division, 204948Sunplanet Co., Ltd, Tsukuba-shi, Japan
| | - Yuji Mano
- Global Drug Metabolism and Pharmacokinetics, 8030Eisai Co., Ltd, Tsukuba-shi, Japan
- Laboratory of Genomics-based Drug Discovery, Faculty of Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
4
|
Garcia AP, Hatvany JB, Murphy MA, Atchley DH, Gurley BJ, Kamdem LK. Effect of Aromatase Inhibition (Exemestane) on Urine Concentration of Osteoprotegerin in Healthy Postmenopausal Women. J Clin Pharmacol 2019; 60:209-217. [PMID: 31535401 DOI: 10.1002/jcph.1519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/12/2019] [Indexed: 11/06/2022]
Abstract
This pilot study examined how exemestane (an aromatase inhibitor [AI]) affected osteoprotegerin (OPG) urine concentrations in postmenopausal women. Exemestane (25 mg, single dose) was given to 14 disease-free women past menopause in this nonrandomized, open-label study. Before dosing, urine specimens were gathered. Three days later, these women returned to provide urine specimens for pharmacokinetic (measurement of major parent drug and enzymatic product) and pharmacodynamic (profiling of OPG) analysis. Urine concentrations of the major parent drug (exemestane) and enzymatic product (17-hydroexemestane) were quantified using liquid chromatography-tandem mass spectrometry. An analyst software package was used for data processing. Following the manufacturer's guidelines, OPG urine concentrations were quantified using a human osteoprotegerin TNFRSF11b ELISA kit from Sigma-Aldrich. A microplate reader helped to carry out OPG data analysis and processing. Our results highlight that OPG urine concentrations were decreased 3 days after drug dosage (mean predosage OPG concentration, 61.4 ± 24.1 pg/mL; vs mean postdosage OPG concentration, 45.7 ± 22.1 pg/mL; P = .02, Wilcoxon rank test). Among the 14 volunteers enrolled in the study, 4 subjects had an increase of less than 1-fold, and the rest showed an average of a 2-fold decrease in OPG concentration (range, 1.1-5.4; standard deviation, 1.3) after exemestane administration. There was no association between fold decrease in OPG urine concentration and the pharmacokinetics of the major parent drug (exemestane) and its enzymatic product (17-hydroexemestane). We concluded that one of the off-target pharmacological effects of AIs (eg ,exemestane) may result in the reduction of osteoprotegerin.
Collapse
Affiliation(s)
| | - Jacob B Hatvany
- Harding University College of Pharmacy, Searcy, Arkansas, USA
| | - Michael A Murphy
- Harding University Physician Assistant Program, Searcy, Arkansas, USA
| | - Daniel H Atchley
- University of Pikeville-Kentucky College of Osteopathic Medicine, KYCOM, Pikeville, Kentucky, USA
| | - Bill J Gurley
- University of Arkansas for Medical Sciences, College of Pharmacy, Little Rock, Arkansas, USA
| | - Landry K Kamdem
- Harding University College of Pharmacy, Searcy, Arkansas, USA
| |
Collapse
|
5
|
van Nuland M, Venekamp N, de Vries N, de Jong KAM, Rosing H, Beijnen JH. Development and validation of an UPLC-MS/MS method for the therapeutic drug monitoring of oral anti-hormonal drugs in oncology. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1106-1107:26-34. [PMID: 30639947 DOI: 10.1016/j.jchromb.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/31/2018] [Accepted: 01/02/2019] [Indexed: 10/27/2022]
Abstract
A liquid chromatography-mass spectrometry assay was developed and validated for simultaneous quantification of anti-hormonal compounds abiraterone, anastrozole, bicalutamide, Δ(4)-abiraterone (D4A), N-desmethyl enzalutamide, enzalutamide, Z-endoxifen, exemestane and letrozole for the purpose of therapeutic drug monitoring (TDM). Plasma samples were prepared with protein precipitation. Analyses were performed with a triple quadrupole mass spectrometer operating in the positive and negative ion-mode. The validated assay ranges from 2 to 200 ng/mL for abiraterone, 0.2-20 ng/mL for D4A, 10-200 ng/mL for anastrozole and letrozole, 1-20 ng/mL for Z-endoxifen, 1.88-37.5 ng/mL for exemestane and 1500-30,000 ng/mL for enzalutamide, N-desmethyl enzalutamide and bicalutamide. Due to low sensitivity for exemestane, the final extract of exemestane patient samples should be concentrated prior to injection and a larger sample volume should be prepared for exemestane patient samples and QC samples to obtain adequate sensitivity. Furthermore, we observed a batch-dependent stability for abiraterone in plasma at room temperature and therefore samples should be shipped on ice. This newly validated method has been successfully applied for routine TDM of anti-hormonal drugs in cancer patients.
Collapse
Affiliation(s)
- M van Nuland
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands; Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - N Venekamp
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - N de Vries
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - K A M de Jong
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands; Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - H Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, the Netherlands; Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
6
|
Role of the UGT2B17 deletion in exemestane pharmacogenetics. THE PHARMACOGENOMICS JOURNAL 2017; 18:295-300. [PMID: 28534527 PMCID: PMC5700874 DOI: 10.1038/tpj.2017.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/11/2016] [Accepted: 01/09/2017] [Indexed: 12/11/2022]
Abstract
Exemestane (EXE) is an aromatase inhibitor used for the prevention and treatment of breast cancer. The major metabolic pathway for EXE is reduction to form the active 17β-dihydro-EXE (17β-DHE) and subsequent glucuronidation to 17β-hydroxy-EXE-17-O-β-D-glucuronide (17β-DHE-Gluc) by UGT2B17. The aim of the present study was to determine the effects of UGT2B17 copy number variation on the levels of urinary and plasma 17β-DHE-Gluc and 17β-DHE in patients taking EXE. Ninety-six post-menopausal Caucasian breast cancer patients with ER+ breast tumors taking 25 mg EXE daily were recruited into this study. UGT2B17 copy number was determined by a real-time PCR copy number variant assay and the levels of EXE, 17β-DHE and 17β-DHE-Gluc were quantified by UPLC/MS in patients’ urine and plasma. A 39-fold decrease (P<0.0001) in the levels of creatinine-adjusted urinary 17β-DHE-Gluc was observed among UGT2B17 (*2/*2) subjects vs. subjects with the UGT2B17 (*1/*1) genotype. The plasma levels of 17β-DHE-Gluc was decreased 29-fold (P<0.0001) in subjects with the UGT2B17 (*2/*2) genotype vs. subjects with UGT2B17 (*1/*1) genotype. The levels of plasma EXE-adjusted 17β-DHE was 28% higher (P=0.04) in subjects with the UGT2B17 (*2/*2) genotype vs. subjects with the UGT2B17 (*1/*1) genotype. These data indicate that UGT2B17 is the major enzyme responsible for 17β-DHE-Gluc formation in vivo and that the UGT2B17 copy number variant may play a role in inter-individual variability in 17β-DHE levels in vivo.
Collapse
|
7
|
Peterson A, Xia Z, Chen G, Lazarus P. In vitro metabolism of exemestane by hepatic cytochrome P450s: impact of nonsynonymous polymorphisms on formation of the active metabolite 17 β-dihydroexemestane. Pharmacol Res Perspect 2017; 5:e00314. [PMID: 28603633 PMCID: PMC5464343 DOI: 10.1002/prp2.314] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 11/29/2022] Open
Abstract
Exemestane (EXE) is an endocrine therapy commonly used by postmenopausal women with hormone‐responsive breast cancer due to its potency in inhibiting aromatase‐catalyzed estrogen synthesis. Preliminary in vitro studies sought to identify phase I EXE metabolites and hepatic cytochrome P450s (CYP450s) that participate in EXE biotransformation. Phase I metabolites were identified by incubating EXE with HEK293‐overexpressed CYP450s. CYP450s 1A2, 2C8, 2C9, 2C19, 2D6, 3A4, and 3A5 produce 17β‐dihydroexemestane (17β‐DHE), an active major metabolite, as well as two inactive metabolites. 17β‐DHE formation in pooled human liver microsomes subjected to isoform‐specific CYP450 inhibition was also monitored using tandem mass spectrometry. 17β‐DHE production in human liver microsomes was unaffected by isoform‐specific inhibition of CYP450s 2A6, 2B6, and 2E1 but decreased 12–39% following inhibition of drug‐metabolizing enzymes from CYP450 subfamilies 1A, 2C, 2D, and 3A. These results suggest that redundancy exists in the EXE metabolic pathway with multiple hepatic CYP450s catalyzing 17β‐DHE formation in vitro. To further expand the knowledge of phase I EXE metabolism, the impact of CYP450 genetic variation on 17β‐DHE formation was assessed via enzyme kinetic parameters. Affinity for EXE substrate and enzyme catalytic velocity were calculated for hepatic wild‐type CYP450s and their common nonsynonymous variants by monitoring the reduction of EXE to 17β‐DHE. Several functional polymorphisms in xenobiotic‐metabolizing CYP450s 1A2, 2C8, 2C9, and 2D6 resulted in deviant enzymatic activity relative to wild‐type enzyme. Thus, it is possible that functional polymorphisms in EXE‐metabolizing CYP450s contribute to inter‐individual variability in patient outcomes by mediating overall exposure to the drug and its active metabolite, 17β‐DHE.
Collapse
Affiliation(s)
- Amity Peterson
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Zuping Xia
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Gang Chen
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| | - Philip Lazarus
- Department of Pharmaceutical Sciences Washington State University Spokane Washington
| |
Collapse
|
8
|
Impact of nonsynonymous single nucleotide polymorphisms on in-vitro metabolism of exemestane by hepatic cytosolic reductases. Pharmacogenet Genomics 2017; 26:370-80. [PMID: 27111237 DOI: 10.1097/fpc.0000000000000226] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Exemestane (EXE) is a potent third-generation aromatase inhibitor used as endocrine therapy in breast cancer treatment and prevention. Characterization of its metabolic pathway is incomplete, with ambiguity existing in the identity of enzymes driving the production of its key metabolite, 17β-dihydroexemestane (17β-DHE). The impact of genetic variation on EXE metabolism is also unknown. This study aims to describe cytosolic reductase involvement in hepatic EXE metabolism and to assess the impact of functional polymorphisms on metabolite production. MATERIALS AND METHODS Phase I metabolites were identified in incubations of EXE with pooled human liver cytosol or recombinant protein for AKR1Cs and CBR1. Kinetic parameters characterizing EXE reduction were measured for purified wild-type enzymes, and nonsynonymous variants occurring at greater than 1% minor allele frequency using UPLC/MS/MS. RESULTS Human liver cytosol, CBR1, AKR1C1, AKR1C2, AKR1C3, and AKR1C4 reduce EXE to active primary metabolite 17β-DHE. The formation of a novel metabolite, 17α-DHE, was catalyzed by recombinant AKR1C4 and CBR1 in addition to hepatic cytosol. Variants AKR1C3 Arg258Cys and AKR1C4 Gly135Glu had significantly decreased affinity for EXE relative to their respective wild types. Five common AKR1C3 polymorphisms were associated with decreased rates of catalysis, whereas AKR1C4 Gly135Glu increased the velocity of EXE reduction. CONCLUSION AKR1Cs and CBR1 catalyze EXE reduction in vitro. These results imply that cytosolic ketosteroid reductases may participate in the EXE metabolic pathway in vivo. In addition, several common variants were associated with altered enzymatic activity, suggesting that functional polymorphisms could play an important role in overall EXE metabolism and activity by altering the extent and duration of 17β-DHE exposure.
Collapse
|
9
|
Schwartzberg LS, Yardley DA, Elias AD, Patel M, LoRusso P, Burris HA, Gucalp A, Peterson AC, Blaney ME, Steinberg JL, Gibbons JA, Traina TA. A Phase I/Ib Study of Enzalutamide Alone and in Combination with Endocrine Therapies in Women with Advanced Breast Cancer. Clin Cancer Res 2017; 23:4046-4054. [PMID: 28280092 DOI: 10.1158/1078-0432.ccr-16-2339] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/17/2016] [Accepted: 03/02/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Several lines of evidence support targeting the androgen signaling pathway in breast cancer. Enzalutamide is a potent inhibitor of androgen receptor signaling. Preclinical data in estrogen-expressing breast cancer models demonstrated activity of enzalutamide monotherapy and enhanced activity when combined with various endocrine therapies (ET). Enzalutamide is a strong cytochrome P450 3A4 (CYP3A4) inducer, and ETs are commonly metabolized by CYP3A4. The pharmacokinetic (PK) interactions, safety, and tolerability of enzalutamide monotherapy and in combination with ETs were assessed in this phase I/Ib study.Experimental Design: Enzalutamide monotherapy was assessed in dose-escalation and dose-expansion cohorts of patients with advanced breast cancer. Additional cohorts examined effects of enzalutamide on anastrozole, exemestane, and fulvestrant PK in patients with estrogen receptor-positive/progesterone receptor-positive (ER+/PgR+) breast cancer.Results: Enzalutamide monotherapy (n = 29) or in combination with ETs (n = 70) was generally well tolerated. Enzalutamide PK in women was similar to prior data on PK in men with prostate cancer. Enzalutamide decreased plasma exposure to anastrozole by approximately 90% and exemestane by approximately 50%. Enzalutamide did not significantly affect fulvestrant PK. Exposure of exemestane 50 mg/day given with enzalutamide was similar to exemestane 25 mg/day alone.Conclusions: These results support a 160 mg/day enzalutamide dose in women with breast cancer. Enzalutamide can be given in combination with fulvestrant without dose modifications. Exemestane should be doubled from 25 mg/day to 50 mg/day when given in combination with enzalutamide; this combination is being investigated in a randomized phase II study in patients with ER+/PgR+ breast cancer. Clin Cancer Res; 23(15); 4046-54. ©2017 AACR.
Collapse
Affiliation(s)
| | - Denise A Yardley
- Sarah Cannon Research Institute, Nashville, Tennessee.,Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Anthony D Elias
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Manish Patel
- Sarah Cannon Research Institute, Nashville, Tennessee.,Florida Cancer Specialists, Sarah Cannon Research Institute, Sarasota, Florida
| | - Patricia LoRusso
- Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Howard A Burris
- Sarah Cannon Research Institute, Nashville, Tennessee.,Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Ayca Gucalp
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Amy C Peterson
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | - Martha E Blaney
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | | | - Jacqueline A Gibbons
- Medivation, Inc. (Medivation, Inc., was acquired by Pfizer, Inc., in September 2016), San Francisco, California
| | - Tiffany A Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| |
Collapse
|
10
|
Chen SM, Atchley DH, Murphy MA, Gurley BJ, Kamdem LK. Impact of UGT2B17 Gene Deletion on the Pharmacokinetics of 17-Hydroexemestane in Healthy Volunteers. J Clin Pharmacol 2015; 56:875-84. [PMID: 26608382 DOI: 10.1002/jcph.673] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/04/2015] [Indexed: 11/06/2022]
Abstract
Exemestane is an aromatase inhibitor drug used for the treatment of hormone-dependent breast cancer. 17-Hydroexemestane, the major and biologically active metabolite of exemestane in humans, is eliminated via glucuronidation by the polymorphic UGT2B17 phase II drug-metabolizing enzyme. Previous microsomal studies have shown that UGT2B17 gene deletion affects the intrinsic hepatic clearances of 17-hydroexemestane in vitro. In this open-label study we set out to assess the effect of UGT2B17 gene deletion on the pharmacokinetics of 17-hydroexemestane in healthy female volunteers with and without UGT2B17. To achieve this goal, 14 healthy postmenopausal women (8 carriers of the homozygous UGT2B17 wild-type allele and 6 carriers of the homozygous UGT2B17 gene-deletion allele) were enrolled and invited to receive a single 25-mg oral dose of exemestane. Pharmacokinetics was assessed over 72 hours postdosing. Our results showed that there were statistically significant differences in plasma 17-hydroexemestane AUC0-∞ (P = .0007) and urine 17-hydroexemestane C24h (P = .001) between UGT2B17 genotype groups. Our data suggest that UGT2B17 gene deletion influences 17-hydroexemestane pharmacokinetics in humans.
Collapse
Affiliation(s)
- Shanly M Chen
- Harding University College of Pharmacy, Searcy, AR, USA
| | - Daniel H Atchley
- University of Pikeville, Kentucky College of Osteopathic Medicine, Pikeville, KY, USA
| | | | - Bill J Gurley
- University of Arkansas for Medical Sciences, College of Pharmacy, Little Rock, AR, USA
| | | |
Collapse
|
11
|
Wang LZ, Goh SH, Wong ALA, Thuya WL, Lau JYA, Wan SC, Lee SC, Ho PC, Goh BC. Validation of a rapid and sensitive LC-MS/MS method for determination of exemestane and its metabolites, 17β-hydroxyexemestane and 17β-hydroxyexemestane-17-O-β-D-glucuronide: application to human pharmacokinetics study. PLoS One 2015; 10:e0118553. [PMID: 25793887 PMCID: PMC4368747 DOI: 10.1371/journal.pone.0118553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/20/2015] [Indexed: 11/24/2022] Open
Abstract
A novel, rapid and sensitive liquid chromatography-tandem mass spectrometric (LC-MS/MS) method was developed and validated for the evaluation of exemestane pharmacokinetics and its metabolites, 17β-dihydroexemestane (active metabolite) and 17β-dihydroexemestane-17-O-β-D-glucuronide (inactive metabolite) in human plasma. Their respective D3 isotopes were used as internal standards. Chromatographic separation of analytes was achieved using Thermo Fisher BDS Hypersil C18 analytic HPLC column (100 × 2.1 mm, 5 μm). The mobile phase was delivered at a rate of 0.5 mL/min by gradient elution with 0.1 % aqueous formic acid and acetonitrile. The column effluents were detected by API 4000 triple quadrupole mass spectrometer using electrospray ionisation (ESI) and monitored by multiple reaction monitoring (MRM) in positive mode. Mass transitions 297 > 121 m/z, 300 > 121 m/z, 299 > 135 m/z, 302 > 135 m/z, 475 > 281 m/z, and 478 > 284 m/z were monitored for exemestane, exemestane-d3, 17β-dihydroexemestane, 17β-dihydroexemestane-d3, 17β-dihydroexemestane-17-O-β-D-glucuronide, and 17β-dihydroexemestane-17-O-β-D-glucuronide-d3 respectively. The assay demonstrated linear ranges of 0.4 – 40.0 ng/mL, for exemestane; and 0.2 – 15.0 ng/mL, for 17β-dihydroexemestane and 17β-dihydroexemestane-17-O-β-D-glucuronide, with coefficient of determination (r2) of > 0.998. The precision (coefficient of variation) were ≤10.7%, 7.7% and 9.5% and the accuracies ranged from 88.8 to 103.1% for exemestane, 98.5 to 106.1% for 17β-dihydroexemestane and 92.0 to 103.2% for 17β-dihydroexemestane-17-O-β-D-glucuronide. The method was successfully applied to a pharmacokinetics/dynamics study in breast cancer patients receiving exemestane 25mg daily orally. For a representative patient, 20.7% of exemestane in plasma was converted into 17β-dihydroexemestane and 29.0% of 17β-dihydroexemestane was inactivated as 17β-dihydroexemestane-17-O-β-D-glucuronide 24 hours after ingestion of exemestane, suggesting that altered 17-dihydroexemestane glucuronidation may play an important role in determining effect of exemestane against breast cancer cells.
Collapse
Affiliation(s)
- Ling-Zhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117599, Singapore
- * E-mail: (LZW); (BCG)
| | - Sok-Hwei Goh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- Department of Haematology & Oncology, National University Health System, Singapore 119074, Singapore
| | - Win-Lwin Thuya
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Jie-Ying Amelia Lau
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Seow-Ching Wan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Soo-Chin Lee
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- Department of Haematology & Oncology, National University Health System, Singapore 119074, Singapore
| | - Paul C. Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117599, Singapore
- Department of Haematology & Oncology, National University Health System, Singapore 119074, Singapore
- * E-mail: (LZW); (BCG)
| |
Collapse
|
12
|
Esteva FJ, Moulder SL, Gonzalez-Angulo AM, Ensor J, Murray JL, Green MC, Koenig KB, Lee MH, Hortobagyi GN, Yeung SC. Phase I trial of exemestane in combination with metformin and rosiglitazone in nondiabetic obese postmenopausal women with hormone receptor-positive metastatic breast cancer. Cancer Chemother Pharmacol 2012; 71:63-72. [PMID: 23053261 DOI: 10.1007/s00280-012-1977-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 09/13/2012] [Indexed: 12/21/2022]
Abstract
PURPOSE Obese women with breast cancer have worse prognosis than women with normal body mass index. Endocrine therapy resistance is in part mediated by insulin resistance in obese women with breast cancer. We investigated the tolerability and pharmacokinetics of exemestane in combination with metformin and rosiglitazone in nondiabetic overweight and obese postmenopausal women with hormone receptor-positive metastatic breast cancer. METHODS Patients had previously received chemotherapy and endocrine therapy for breast cancer. Exemestane was given as 25 mg orally per day. Metformin (M) and rosiglitazone (R) were given twice daily. Dose level 1 consisted of M 1,500 mg/day and R 6 mg/day. Dose level 2 consisted of M 2,000 mg/day and R 8 mg/day. Plasma concentrations of exemestane were measured on days 1, 8, and 15. RESULTS Twenty patients were enrolled. Fourteen patients received exemestane, metformin, and rosiglitazone. Six patients received exemestane with metformin only (2,000 mg/day). Both regimens were well tolerated at the highest doses tested, and there were no notable changes in plasma exemestane levels. Six patients (30%) had stable disease for 6 months or longer. CONCLUSIONS Oral daily administration of exemestane (25 mg) and metformin (2,000 mg) with and without rosiglitazone (8 mg) daily was well tolerated. Exemestane pharmacokinetics were not altered by metformin and rosiglitazone.
Collapse
Affiliation(s)
- Francisco J Esteva
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Singh A, Chaurasiya A, Warsi MH, Chaurasiya M, Jain GK, Asati D, Khar RK, Mukherjee R. ORAL PHARMACOKINETIC STUDY OF EXEMESTANE SMEDDS AND SUSPENSION IN RAT PLASMA BY LIQUID CHROMATOGRAPHY-MASS SPECTROMETRIC ANALYSIS. J LIQ CHROMATOGR R T 2012. [DOI: 10.1080/10826076.2011.629388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Ajeet Singh
- a Department of Pharmaceutics, Faculty of Pharmacy , Hamdard University , New Delhi , India
- b Matrix Laboratories Limited , Hyderabad , India
| | - Akash Chaurasiya
- a Department of Pharmaceutics, Faculty of Pharmacy , Hamdard University , New Delhi , India
| | - Musarrat H. Warsi
- a Department of Pharmaceutics, Faculty of Pharmacy , Hamdard University , New Delhi , India
| | - Manika Chaurasiya
- c Department of Pharmaceutical Sciences , Dr. H. S. Gour University , Sagar , India
| | - Gaurav K. Jain
- a Department of Pharmaceutics, Faculty of Pharmacy , Hamdard University , New Delhi , India
| | | | - Roop K. Khar
- a Department of Pharmaceutics, Faculty of Pharmacy , Hamdard University , New Delhi , India
| | - Rama Mukherjee
- e ARA Healthcare Pvt. Ltd., Electronic City , Haryana , India
| |
Collapse
|
14
|
Ksycińska H, Buś-Kwaśnik K, Szlagowska A, Rudzki PJ. Development and validation of a sensitive liquid chromatography/tandem mass spectrometry method for the determination of exemestane in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879:1905-10. [DOI: 10.1016/j.jchromb.2011.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 05/06/2011] [Accepted: 05/08/2011] [Indexed: 10/18/2022]
|
15
|
Kamdem LK, Flockhart DA, Desta Z. In vitro cytochrome P450-mediated metabolism of exemestane. Drug Metab Dispos 2011; 39:98-105. [PMID: 20876785 PMCID: PMC3014267 DOI: 10.1124/dmd.110.032276] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Accepted: 09/28/2010] [Indexed: 11/22/2022] Open
Abstract
Exemestane is a potent and irreversible steroidal aromatase inhibitor drug used for the treatment of estrogen receptor-positive breast cancer. Our aim was to identify and assess the contribution of the specific cytochromes P450 (P450s) responsible for exemestane primary in vitro metabolism. With the use of high-performance liquid chromatography and liquid chromatography-tandem mass spectrometry analytical techniques, 17-hydroexemestane (MI) formation and 6-hydroxymethylexemestane (MII) formation were found to be the predominant exemestane metabolic pathways. In a bank of 15 well characterized human liver microsomes with known P450 isoform-specific activities, the MI formation rate correlated significantly with CYP1A2 (Spearman r = 0.60, p = 0.02) and CYP4A11 (Spearman r = 0.67, p = 0.01) isoform-specific activities, whereas the MII production rate significantly correlated with CYP2B6 (Spearman r = 0.57, p = 0.03) and CYP3A (Spearman r = 0.76, p = 0.005) isoform-specific activities. Recombinant CYP1A1 metabolized exemestane to MI with a catalytic efficiency (Cl(int)) of 150 nl/pmol P450 × min that was at least 3.5-fold higher than those of other P450s investigated. Recombinant CYP3A4 catalyzed MII formation from exemestane with a catalytic efficiency of 840 nl/pmol P450 × min that was at least 4-fold higher than those of other P450s investigated. Among a panel of 10 chemical inhibitors tested, only ketoconazole and troleandomycin (CYP3A-specific chemical inhibitors) significantly inhibited the formation of MII by 45 and 95%, respectively. None of them markedly inhibited the formation of MI. In summary, exemestane seems to be metabolized to MI by multiple P450s that include CYP4A11 and CYP1A1/2, whereas its oxidation to MII is primarily mediated by CYP3A.
Collapse
Affiliation(s)
- Landry K Kamdem
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | | | | |
Collapse
|
16
|
Sun D, Chen G, Dellinger RW, Sharma AK, Lazarus P. Characterization of 17-dihydroexemestane glucuronidation: potential role of the UGT2B17 deletion in exemestane pharmacogenetics. Pharmacogenet Genomics 2010; 20:575-85. [PMID: 20697310 PMCID: PMC3076703 DOI: 10.1097/fpc.0b013e32833b04af] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Exemestane is a third-generation aromatase inhibitor used in the treatment of breast cancer in postmenopausal women. Reduction to form 17-dihydroexemestane and subsequent glucuronidation to exemestane-17-O-glucuronide is a major pathway for exemestane metabolism. The goal of this study was to analyze 17-dihydroexemestane anti-aromatase activity, characterize the 17-dihydroexemestane glucuronidation pathway, and determine whether the functional polymorphisms in active UGTs could play a role in altered 17-dihydroexemestane glucuronidation. METHODS Homogenates from a HEK293 aromatase-overexpressing cell line (HEK293-aro) were used to examine exemestane versus 17-dihydroexemestane anti-aromatase activities. UGT-overexpressing cell lines and a panel (n=110) of human liver microsome (HLM) were screened for glucuronidation activity against 17-dihydroexemestane. UGT2B17 genotyping and liver mRNA expression were performed by real-time PCR. RESULTS The inhibition of estrone formation from androst-4-ene-3,17-dione in HEK293-aro cell homogenates was similar for 17-dihydroexemestane (IC(50)=2.3±0.83 μmol/l) and exemestane (IC(50)=1.4±0.42 μmol/l). UGTs 2B17 and 1A4 were high-expression hepatic UGTs that exhibited activity against 17-dihydroexemestane, with UGT2B17 exhibiting a 17-fold higher V(max)/K(M) than UGT1A4. The rate of exemestane-17-O-glucuronide formation was shown to be significantly (P<0.001) decreased (14-fold) in HLMs exhibiting the UGT2B17(*2/*2) deletion genotype versus wild-type UGT2B17(*1/*1) HLMs; a 36-fold lower V(max)/K(M) (P=0.023) was observed in UGT2B17(*2/*2) versus UGT2B17(*1/*1) HLMs. A significant (P<0.0001, R(2)=0.72) correlation was observed between HLM exemestane-17-O-glucuronide formation and liver UGT2B17 expression. CONCLUSION These data suggest that 17-dihydroexemestane is an active metabolite of exemestane and that the UGT2B17 deletion polymorphism could play an important role in determining levels of excretion of 17-dihydroexemestane and overall exemestane metabolism.
Collapse
Affiliation(s)
- Dongxiao Sun
- Molecular Epidemiology and Cancer Control, Penn State Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Gang Chen
- Molecular Epidemiology and Cancer Control, Penn State Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ryan W. Dellinger
- Molecular Epidemiology and Cancer Control, Penn State Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Arun K. Sharma
- Chemical Carcinogenesis and Chemoprevention Programs, Penn State Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Philip Lazarus
- Molecular Epidemiology and Cancer Control, Penn State Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, USA
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
17
|
Thomas A, Guddat S, Kohler M, Krug O, Schänzer W, Petrou M, Thevis M. Comprehensive plasma-screening for known and unknown substances in doping controls. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2010; 24:1124-1132. [PMID: 20301105 DOI: 10.1002/rcm.4492] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Occasionally, doping analysis has been recognized as a competitive challenge between cheating sportsmen and the analytical capabilities of testing laboratories. Both have made immense progress during the last decades, but obviously the athletes have the questionable benefit of frequently being able to switch to new, unknown and untested compounds to enhance their performance. Thus, as analytical counteraction and for effective drug testing, a complementary approach to classical targeted methods is required in order to implement a comprehensive screening procedure for known and unknown xenobiotics. The present study provides a new analytical strategy to circumvent the targeted character of classical doping controls without losing the required sensitivity and specificity. Using 50 microL of plasma only, the method potentially identifies illicit drugs in low ng/mL concentrations. Plasma provides the biological fluid with the circulating, unmodified xenobiotics; thus the identification of unknown compounds is facilitated. After a simple protein precipitation, liquid chromatographic separation and subsequent detection by means of high resolution/high accuracy orbitrap mass spectrometry, the procedure enables the determination of numerous compounds from different classes prohibited by the World Anti-Doping Agency (WADA). A new hyphenated mass spectrometry technology was employed without precursor ion selection for higher collision energy dissociation (HCD) fragmentation experiments. Thus the mass spectra contained all the desired information to identify unknown substances retrospectively. The method was validated for 32 selected model compounds for qualitative purposes considering the parameters specificity, selectivity, limit of detection (<0.1-10 ng/mL), precision (9-28%), robustness, linearity, ion suppression and recovery (80-112%). In addition to the identification of unknown compounds, the plasma samples were simultaneously screened for known prohibited targets.
Collapse
Affiliation(s)
- Andreas Thomas
- Institute of Biochemistry, Center for Preventive Doping Research, German Sport University Cologne, Am Sportpark Müngersdorf, 50933 Cologne, Germany.
| | | | | | | | | | | | | |
Collapse
|
18
|
Shou WZ, Zhang J. Recent development in high-throughput bioanalytical support forin vitroADMET profiling. Expert Opin Drug Metab Toxicol 2010; 6:321-36. [DOI: 10.1517/17425250903547829] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Corona G, Elia C, Casetta B, Da Ponte A, Del Pup L, Ottavian E, Toffoli G. Liquid chromatography tandem mass spectrometry assay for fast and sensitive quantification of estrone-sulfate. Clin Chim Acta 2010; 411:574-80. [PMID: 20096278 DOI: 10.1016/j.cca.2010.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND The circulating pool of estrone-sulfate is considered as a "reservoir" of slowly-metabolized estrogen that can be exploited for assessing overall individual estrogenicity. The aim of this study was to develop a rapid and sensitive liquid chromatography-tandem mass spectrometry assay for the determination of estrone-sulfate, suitable for routine clinical investigations. METHODS The proposed assay is based on a simple protein precipitation procedure and on a fast measurement with a triple-quadrupole mass spectrometer operating in negative ion mode and in multiple reaction monitoring. The method was assessed for intra- and inter-day precision, accuracy, recovery, and clinical suitability. A comparison with available radioimmunoassay was also performed. RESULTS The LC-MS/MS method is able to detect estrone-sulfate concentrations < or =1pg/mL and has a low limit of quantification of 7.8pg/mL. Intra- and inter-day precision and accuracy were less than 10.5% and 5.0% respectively. The recovery was in the range of 93%-110%. When compared with radioimmunoassay the method resulted more accurate and therefore more suitable for quantifying the estrone-sulfate in different clinical settings, including patients treated with aromatase inhibitors. CONCLUSIONS The proposed LC-MS/MS method represents a convincing alternative to the immunoassay for a fast, cost-effective and reliable measurement of estrone-sulfate in routine clinical investigations and in large epidemiological studies. It may contribute in shedding a new light on the diagnostic value of estrone-sulfate in normal and pathological conditions.
Collapse
Affiliation(s)
- Giuseppe Corona
- National Cancer Institute and Center for Molecular Biomedicine, Aviano, PN, Italy.
| | | | | | | | | | | | | |
Collapse
|