Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Article in English | MEDLINE | ID: mdl-38636136

ABSTRACT

A liquid chromatography - electrospray ionization-mass spectrometry (LC-ESI-MS) method was developed for the quantification of letrozole, a third-generation aromatase inhibitor, and its main carbinol metabolite (CM) in support of murine pharmacokinetic studies. Using polarity switching, simultaneous ESI-MS measurement of letrozole and CM was achieved in positive and negative mode, respectively. The assay procedure involved a one-step protein precipitation and extraction of all analytes from mouse plasma requiring only 5 µL of sample. Separation was optimized on an Accucore aQ column with gradient elution at a flow rate of 0.4 mL/min in 5 min. Two calibration curves per day over four consecutive measurement days showed satisfactory linear responses (r2 > 0.99) over concentration ranges of 5-1000 ng/mL and 20-2000 ng/mL for letrozole and CM, respectively. No matrix effect was found, and the mean extraction recoveries were 103-108 % for letrozole and 99.8-107 % for CM. Precision and accuracy within a single run and over four consecutive measurement days were verified to be within acceptable limits. Application of the developed method to preclinical pharmacokinetic studies in mice receiving oral letrozole at a dose 1 or 10 mg/kg revealed that the systemic exposure to letrozole was dose-, formulation-, and strain-dependent. These findings may inform the future design of preclinical studies aimed at refining the pharmacological profile of this clinically important drug.


Subject(s)
Aromatase Inhibitors , Letrozole , Nitriles , Tandem Mass Spectrometry , Triazoles , Animals , Letrozole/blood , Letrozole/pharmacokinetics , Letrozole/chemistry , Mice , Tandem Mass Spectrometry/methods , Aromatase Inhibitors/blood , Aromatase Inhibitors/pharmacokinetics , Aromatase Inhibitors/chemistry , Chromatography, High Pressure Liquid/methods , Nitriles/blood , Nitriles/pharmacokinetics , Triazoles/blood , Triazoles/pharmacokinetics , Triazoles/chemistry , Reproducibility of Results , Linear Models , Limit of Detection , Female , Male
2.
Drug Metab Dispos ; 49(12): 1047-1055, 2021 12.
Article in English | MEDLINE | ID: mdl-34593616

ABSTRACT

Exemestane (EXE) is a hormonal therapy used to treat estrogen receptor-positive breast cancer by inhibiting the final step of estrogen biosynthesis catalyzed by the enzyme aromatase. Cysteine conjugates of EXE and its active metabolite 17ß-dihydro-EXE (DHE) are the major metabolites found in both the urine and plasma of patients taking EXE. The initial step in cysteine conjugate formation is glutathione conjugation catalyzed by the glutathione S-transferase (GST) family of enzymes. The goal of the present study was to identify cytosolic hepatic GSTs active in the GST-mediated metabolism of EXE and 17ß-DHE. Twelve recombinant cytosolic hepatic GSTs were screened for their activity against EXE and 17ß-DHE, and glutathionylated EXE and 17ß-DHE conjugates were detected by ultra-performance liquid chromatography tandem mass spectrometry. GST α (GSTA) isoform 1, GST µ (GSTM) isoform 3 and isoform 1 were active against EXE, whereas only GSTA1 exhibited activity against 17ß-DHE. GSTM1 exhibited the highest affinity against EXE with a Michaelis-Menten constant (KM) value that was 3.8- and 7.1-fold lower than that observed for GSTA1 and GSTM3, respectively. Of the three GSTs, GSTM3 exhibited the highest intrinsic clearance against EXE (intrinsic clearance = 0.14 nl·min-1·mg-1). The KM values observed for human liver cytosol against EXE (46 µM) and 17ß-DHE (77 µM) were similar to those observed for recombinant GSTA1 (53 and 30 µM, respectively). Western blot analysis revealed that GSTA1 and GSTM1 composed 4.3% and 0.57%, respectively, of total protein in human liver cytosol; GSTM3 was not detected. These data suggest that GSTA1 is the major hepatic cytosolic enzyme involved in the clearance of EXE and its major active metabolite, 17ß-DHE. SIGNIFICANCE STATEMENT: Most previous studies related to the metabolism of the aromatase inhibitor exemestane (EXE) have focused mainly on phase I metabolic pathways and the glucuronidation phase II metabolic pathway. However, recent studies have indicated that glutathionylation is the major metabolic pathway for EXE. The present study is the first to characterize hepatic glutathione S-transferase (GST) activity against EXE and 17ß-dihydro-EXE and to identify GST α 1 and GST µ 1 as the major cytosolic GSTs involved in the hepatic metabolism of EXE.


Subject(s)
Androstadienes/pharmacokinetics , Breast Neoplasms , Glutathione Transferase/metabolism , Inactivation, Metabolic/physiology , Liver/enzymology , Antineoplastic Agents, Hormonal/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Chromatography, Liquid , Cysteine/metabolism , Cytosol/metabolism , Estrogens/biosynthesis , Glutathione Transferase/chemistry , Hepatobiliary Elimination/physiology , Humans , Protein Isoforms , Receptors, Estrogen
3.
PLoS One ; 16(4): e0248579, 2021.
Article in English | MEDLINE | ID: mdl-33798227

ABSTRACT

Based on the discovery that the estrogen synthase aromatase (CYP19A1) is abundantly expressed in high- grade gliomas, the aromatase inhibitor, letrozole is being investigated in pre-clinical models as a novel agent against this malignancy. Here, we investigated the systemic and brain pharmacokinetics of letrozole following single and steady state dosing in both male and female Sprague-Dawley rats. Furthermore, we employed physiologically-based pharmacokinetic (PBPK) modeling to gain quantitative insights into the blood-brain barrier penetration of this drug. Letrozole (4 mg/kg) was administered intraperitoneally daily for 5 days (for males) and 11 days (for females) and intracerebral microdialysis was performed for brain extracellular fluid (ECF) collection simultaneously with venous blood sampling. Drug levels were measured using HPLC and non-compartmental analysis was conducted employing WinNonlin®. Simcyp animal simulator was used for conducting bottom-up PBPK approach incorporating the specified multi-compartment brain model. Overall, marked gender-specific differences in the systemic and brain pharmacokinetics of letrozole were observed. Letrozole clearance was much slower in female rats resulting in markedly higher plasma and brain drug concentrations. At steady state, the plasma AUC 0-24 was 103.0 and 24.8 µg*h/ml and brain ECF AUC 0-12 was 24.0 and 4.8 µg*h/ml in female and male rats, respectively. The PBPK model simulated brain concentration profiles were in close agreement with the observed profiles. While gender-specific differences in letrozole PK are not observed in the clinical setting, these findings will guide the dose optimization during pre-clinical investigations of this compound. The PBPK model will serve as an important clinical translational tool.


Subject(s)
Aromatase Inhibitors/pharmacokinetics , Brain/metabolism , Letrozole/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Aromatase Inhibitors/blood , Female , Letrozole/blood , Male , Models, Biological , Rats, Sprague-Dawley , Sex Characteristics , Sex Factors
4.
Cancer Sci ; 112(6): 2381-2392, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33686753

ABSTRACT

This post hoc analysis of MONARCH 2 and MONARCH 3 assesses the efficacy, safety, and pharmacokinetics (PK) of abemaciclib in combination with endocrine therapy (ET) in East Asian patients with hormone receptor positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer. MONARCH 2 and MONARCH 3 are global, randomized, double-blind, phase 3 studies of abemaciclib/placebo + fulvestrant and abemaciclib/placebo + nonsteroidal aromatase inhibitor (NSAI, anastrozole or letrozole), respectively. The East Asian population comprised 212 (31.7%) of the 669 intent-to-treat (ITT) population in the MONARCH 2 trial and 144 (29.2%) of the 493 ITT patients in the MONARCH 3 trial. In the East Asian population, median progression-free survival (PFS) was significantly prolonged in the abemaciclib arm compared with placebo in both MONARCH 2 (hazard ratio [HR], 0.520; 95% confidence interval [CI], 0.362 to 0.747; P < .001; median: 21.2 vs 11.6 months) and MONARCH 3 (HR, 0.326; 95% CI, 0.200 to 0.531, P < .001; median: not reached vs 12.82 months). Diarrhea (MONARCH 2: 90%; MONARCH 3: 88%) and neutropenia (MONARCH 2: 68%; MONARCH 3: 58%) were the most frequent adverse events observed in the East Asian populations. Abemaciclib exposures and PK were similar in East Asians and the non-East Asian populations of both trials. Abemaciclib in combination with ET in the East Asian populations of MONARCH 2 and MONARCH 3 provided consistent results with the ITT populations, demonstrating improvements in efficacy with generally tolerable safety profiles for patients with HR+, HER2- advanced breast cancer.


Subject(s)
Aminopyridines/administration & dosage , Aromatase Inhibitors/administration & dosage , Benzimidazoles/administration & dosage , Breast Neoplasms/drug therapy , Fulvestrant/administration & dosage , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Anastrozole/administration & dosage , Anastrozole/adverse effects , Anastrozole/pharmacokinetics , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Breast Neoplasms/blood , Breast Neoplasms/genetics , Diarrhea/chemically induced , Diarrhea/epidemiology , Double-Blind Method , Drug Therapy, Combination , Female , Fulvestrant/adverse effects , Fulvestrant/pharmacokinetics , Humans , Letrozole/administration & dosage , Letrozole/adverse effects , Letrozole/pharmacokinetics , Neutropenia/chemically induced , Neutropenia/epidemiology , Receptor, ErbB-2/genetics , Treatment Outcome
5.
Curr Drug Deliv ; 17(9): 728-735, 2020.
Article in English | MEDLINE | ID: mdl-32713341

ABSTRACT

The conventional treatment regimen for cancer with a single chemotherapeutic agent is far behind the clinical expectations due to the complexity of cancer biology and is also associated with poor Quality of Life (QOL) due to off-site toxicity and multidrug resistance. In recent years, nanopotentiated combination therapy has shown significant improvement in cancer treatment via a synergistic approach. However, being synthetic in nature, nanocarriers have been associated with the activation of the Complement (C) activation system resulting in serious hypersensitivity reactions known as CActivation Related Pseudoallergy (CARPA) effect once given via intravenous injection. On the other hand, nanopotentiated oral drug delivery offers several advantages for the effective and safe delivery of the drug to the target site. This hypothesis aims to put forward wherein Exemestane (chemotherapeutic agent) and lycopene (herbal bioactive) co-laden into PEGylated liposomes and delivered to the breast cancer via the oral route. PEGylation of the liposomes would prevent both molecules from the harsh microenvironment of the Gastrointestinal Tract (GIT) and would eventually promote their intestinal absorption via the lymphatic pathway to the systemic circulation. Lycopene being a potent antioxidant and anti-cancer herbal bioactive would promote the therapeutic efficacy of the Exemestane via a synergistic approach. This nanopotentiated oral combination therapy would pave the path for the safe and effective treatment of cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Breast Neoplasms/drug therapy , Nanoparticles/chemistry , Administration, Oral , Androstadienes/administration & dosage , Androstadienes/adverse effects , Androstadienes/pharmacokinetics , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/pharmacokinetics , Biological Availability , Breast Neoplasms/immunology , Cell Line, Tumor , Complement Activation/drug effects , Drug Synergism , Female , Gastrointestinal Absorption , Humans , Liposomes , Lycopene/administration & dosage , Lycopene/adverse effects , Lycopene/pharmacokinetics , Mice , Polyethylene Glycols/chemistry , Proof of Concept Study , Tissue Distribution , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
6.
JCI Insight ; 5(16)2020 08 20.
Article in English | MEDLINE | ID: mdl-32701512

ABSTRACT

Aromatase inhibitors (AIs) reduce breast cancer recurrence and prolong survival, but up to 30% of patients exhibit recurrence. Using a genome-wide association study of patients entered on MA.27, a phase III randomized trial of anastrozole versus exemestane, we identified a single nucleotide polymorphism (SNP) in CUB And Sushi multiple domains 1 (CSMD1) associated with breast cancer-free interval, with the variant allele associated with fewer distant recurrences. Mechanistically, CSMD1 regulates CYP19 expression in an SNP- and drug-dependent fashion, and this regulation is different among 3 AIs: anastrozole, exemestane, and letrozole. Overexpression of CSMD1 sensitized AI-resistant cells to anastrozole but not to the other 2 AIs. The SNP in CSMD1 that was associated with increased CSMD1 and CYP19 expression levels increased anastrozole sensitivity, but not letrozole or exemestane sensitivity. Anastrozole degrades estrogen receptor α (ERα), especially in the presence of estradiol (E2). ER+ breast cancer organoids and AI- or fulvestrant-resistant breast cancer cells were more sensitive to anastrozole plus E2 than to AI alone. Our findings suggest that the CSMD1 SNP might help to predict AI response, and anastrozole plus E2 serves as a potential new therapeutic strategy for patients with AI- or fulvestrant-resistant breast cancers.


Subject(s)
Anastrozole/pharmacology , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/drug therapy , Membrane Proteins/genetics , Polymorphism, Single Nucleotide , Tumor Suppressor Proteins/genetics , Anastrozole/administration & dosage , Anastrozole/pharmacokinetics , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aromatase/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Genome-Wide Association Study , Humans , Pharmacogenetics , Postmenopause
7.
J Clin Pharmacol ; 60(2): 209-217, 2020 02.
Article in English | MEDLINE | ID: mdl-31535401

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.


Subject(s)
Androstadienes/pharmacology , Androstadienes/pharmacokinetics , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/pharmacokinetics , Osteoprotegerin/urine , Aged , Androstadienes/administration & dosage , Androstadienes/urine , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/urine , Female , Healthy Volunteers , Humans , Middle Aged , Pilot Projects , Postmenopause , Retrospective Studies
8.
Mater Sci Eng C Mater Biol Appl ; 105: 110099, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31546395

ABSTRACT

Multi-modality strategies of albumin-mediated drug accumulation in tumor, boronate-based active tumor targeting and synergistic cancer therapy were combined together for effective treatment of breast cancer. Herein we report the development of albumin-shell oily-core nanocapsules (NCs), loaded with novel combination of hydrophobic drugs, exemestane (EXE) and hesperetin (HES), for targeted breast cancer therapy. This protein-lipid nanohybrid carrier was successfully fabricated using a simple protein-coating method based on the electrostatic adsorption of negatively charged albumin shell onto the oily core containing cationic surfactant. While EXE was directly encapsulated into the oily core, HES was pre-formulated in the form of phospholipid complex before solubilization in oily phase. In addition to albumin-mediated binding to albondin and SPARC, phenylboronic acid was chemically coupled to the albumin shell to confer additional tumor targeting. The targeted nanocarrier (TNC) demonstrated enhanced internalization into MCF-7 breast cancer cells resulting in synergistic cytotoxic activity with a combination index (CI) of 0.662 and dose reduction index (DRI) of 8.22 and 1.84 for EXE and HES, respectively. In vivo, TNC displayed superior anti-cancer activity in tumor-bearing mice compared to their non-targeted counterparts and the free drug combination. A significant reduction of both tumor volume (7-folds) and Ki67 expression (3-folds) was obtained by the targeted nanocarriers compared to positive control. Overall, the boronic-targeted albumin NCs offer a promising platform for hydrophobic drug combination against cancer therapy.


Subject(s)
Androstadienes , Antineoplastic Agents, Phytogenic , Aromatase Inhibitors , Breast Neoplasms , Hesperidin , Nanocapsules , Albumins/chemistry , Albumins/pharmacokinetics , Albumins/pharmacology , Androstadienes/chemistry , Androstadienes/pharmacokinetics , Androstadienes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacokinetics , Aromatase Inhibitors/pharmacology , Boron/chemistry , Boron/pharmacokinetics , Boron/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Hesperidin/chemistry , Hesperidin/pharmacokinetics , Hesperidin/pharmacology , Humans , MCF-7 Cells , Nanocapsules/chemistry , Nanocapsules/therapeutic use , Phospholipids/chemistry , Phospholipids/pharmacokinetics , Phospholipids/pharmacology
9.
Breast Cancer Res ; 21(1): 69, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31126332

ABSTRACT

BACKGROUND: The combined use of a FGFR1 blocker and aromatase inhibitors is appealing for treating breast cancer patients with FGFR1 amplification. However, no pharmacodynamic studies have addressed the effects of this combined target modulation. We conducted a phase 0/I clinical trial in an adjuvant setting, with the goal of obtaining pharmacodynamic proof of the effects of combined aromatase and FGFR1 inhibition and to establish the RP2D for nintedanib combined with letrozole. PATIENTS AND METHODS: Women with early-stage luminal breast cancer were eligible for enrollment in the study. Dose level 1 was nintedanib (150 mg/bid) plus letrozole (2.5 mg/day) administered for a single 28-day cycle (DLT assessment period), followed by a classic 3 + 3 schedule. FGF23 and 17-B-estradiol levels were determined on days 0 and 15; pharmacokinetic parameters were assessed on days 1 and 28. Patients were allowed to continue treatment for 6 cycles. The primary study endpoint was a demonstration of FGFR1 modulation (defined as a 25% increase in the plasma FGF23 level). RESULTS: A total of 19 patients were enrolled in the study (10 in the expansion cohort following dose escalation). At the RP2D (nintedanib 200 mg/bid plus letrozole 2.5 mg/day), we observed a 55% mean increase in the plasma FGF23 level, and 81.2% of the patients had no detectable level of 17-B-estradiol in their plasma (87.5% of the patients treated with letrozole alone). Nintedanib and letrozole displayed a pharmacokinetic interaction that led to three- and twofold increases in their respective plasma concentrations. Most G3 toxic events (5 out of 6: 2 diarrhea and 3 hypertransaminasemia) occurred subsequent to the DLT assessment period. CONCLUSION: Combined treatment with nintedanib (200 mg/bid) plus letrozole (2.5 mg/day) effectively suppressed FGFR1 and aromatase activity, and these respective doses can be used as starting doses in any subsequent trials. However, drug-drug interactions may produce tolerability issues when these drugs are co-administered for an extended time period (e.g., 6 months). Patients enrolled in future trials with these drugs should be carefully monitored for their FGF23 levels and signs of toxicity, and those findings should guide individualized treatment decisions. TRIAL REGISTRATION: This trial was registered at www.clinicaltrials.gov under reg. # NCT02619162, on December 2, 2015.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/pharmacokinetics , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Monitoring , Female , Fibroblast Growth Factor-23 , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Letrozole/administration & dosage , Letrozole/pharmacokinetics , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Treatment Outcome
10.
Clin Pharmacol Ther ; 106(1): 219-227, 2019 07.
Article in English | MEDLINE | ID: mdl-30648747

ABSTRACT

Anastrozole is a widely prescribed aromatase inhibitor for the therapy of estrogen receptor positive (ER+) breast cancer. We performed a genome-wide association study (GWAS) for plasma anastrozole concentrations in 687 postmenopausal women with ER+ breast cancer. The top single-nucleotide polymorphism (SNP) signal mapped across SLC38A7 (rs11648166, P = 2.3E-08), which we showed to encode an anastrozole influx transporter. The second most significant signal (rs28845026, P = 5.4E-08) mapped near ALPPL2 and displayed epistasis with the SLC38A7 signal. Both of these SNPs were cis expression quantitative trait loci (eQTL)s for these genes, and patients homozygous for variant genotypes for both SNPs had the highest drug concentrations, the highest SLC38A7 expression, and the lowest ALPPL2 expression. In summary, our GWAS identified a novel gene encoding an anastrozole transporter, SLC38A7, as well as epistatic interaction between SNPs in that gene and SNPs near ALPPL2 that influenced both the expression of the transporter and anastrozole plasma concentrations.


Subject(s)
Alkaline Phosphatase/genetics , Anastrozole/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Epistasis, Genetic/genetics , Anastrozole/blood , Anastrozole/therapeutic use , Aromatase Inhibitors/blood , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 2/genetics , Female , GPI-Linked Proteins/genetics , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide , Postmenopause , Receptors, Estrogen/biosynthesis
11.
Clin Pharmacol Ther ; 103(2): 243-252, 2018 02.
Article in English | MEDLINE | ID: mdl-29052219

ABSTRACT

Biomedical research is undergoing rapid change, with the development of a series of analytical omics techniques that are capable of generating Biomedical Big Data. These developments provide an unprecedented opportunity to gain novel insight into disease pathophysiology and mechanisms of drug action and response-but they also present significant challenges. Pharmacogenomics is a discipline within Clinical Pharmacology that has been at the forefront in defining, taking advantage of, and dealing with the opportunities and challenges of this aspect of the Post-Genome Project world. This overview will describe the evolution of germline pharmacogenomic research strategies as we have moved from an era of candidate genes to agnostic genome-wide association studies (GWAS) coupled with the functional and mechanistic pursuit of GWAS signals. Germline pharmacogenomic studies of breast cancer endocrine therapy will be used to illustrate research strategies that are being applied broadly to omics studies of drug response phenotypes.


Subject(s)
Antineoplastic Agents/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Drug Development/methods , Drug Discovery/methods , Evidence-Based Medicine/methods , Pharmacogenetics/methods , Translational Research, Biomedical/methods , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Data Mining , Databases, Genetic , Drug Resistance, Neoplasm , Female , Genome-Wide Association Study , Humans , Models, Animal , Models, Theoretical , Patient Safety , Pharmacogenomic Variants , Risk Assessment
12.
Drug Metab Pharmacokinet ; 32(6): 293-300, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29137842

ABSTRACT

The aim of the present study is to investigate the pharmacokinetics of our newly developed aromatase inhibitors (cetrozole and TMD-322) in healthy subjects by a cassette microdose strategy. A cocktail of cetrozole and TMD-322 was administered intravenously or orally (1.98 µg for each drug) to six healthy volunteers in a crossover fashion. Anastrozole (1.98 µg) was also included in the oral cocktail. Total body clearance and bioavailability were 12.1 ± 7.1 mL/min/kg and 34.9 ± 32.3% for cetrozole, and 16.8 ± 3.5 mL/min/kg and 18.4 ± 12.2% for TMD-322, respectively. The area under the plasma concentration-time curves of cetrozole and TMD-322 after oral administration was markedly lower than that of anastrozole because of their high hepatic clearance. Two subjects out of six exhibited 4- and 17-fold larger exposure of cetrozole than the others following intravenous and oral administration, respectively. Such variation was not observed for TMD-322 and anastrozole. Extensive metabolism of cetrozole and TMD-322 was observed in the CYP2C19 expression system among the test CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). We report the first clinical investigation of our aromatase inhibitors by a cassette microdose strategy in healthy Japanese subjects. This strategy offers an optional approach for candidate selection as a phase zero study in drug development.


Subject(s)
Aniline Compounds/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Aromatase/metabolism , Drug Discovery , Nitriles/pharmacokinetics , Triazoles/pharmacokinetics , Administration, Intravenous , Administration, Oral , Aged , Anastrozole , Aniline Compounds/administration & dosage , Aniline Compounds/chemistry , Animals , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/chemistry , Cross-Over Studies , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Japan , Male , Molecular Structure , Nitriles/administration & dosage , Nitriles/chemistry , Rats , Structure-Activity Relationship , Triazoles/administration & dosage , Triazoles/chemistry
13.
Arch Endocrinol Metab ; 61(4): 391-397, 2017.
Article in English | MEDLINE | ID: mdl-28977209

ABSTRACT

Aromatase is a cytochrome P450 enzyme (CYP19A1 isoform) able to catalyze the conversion of androgens to estrogens. The aromatase gene mutations highlighted the action of estrogen as one of the main regulators of bone maturation and closure of bone plate. The use of aromatase inhibitors (AI) in boys with short stature has showed its capability to improve the predicted final height. Anastrozole (ANZ) and letrozole (LTZ) are nonsteroidal inhibitors able to bind reversibly to the heme group of cytochrome P450. In this review, we describe the pharmacokinetic profile of both drugs, discussing possible drug interactions between ANZ and LTZ with other drugs. AIs are triazolic compounds that can induce or suppress cytochrome P450 enzymes, interfering with metabolism of other compounds. Hydroxilation, N-dealkylation and glucoronidation are involved in the metabolism of AIs. Drug interactions can occur with azole antifungals, such as ketoconazole, by inhibiting CYP3A4 and by reducing the clearance of AIs. Antiepileptic drugs (lamotrigine, phenobarbital, and phenytoin) also inhibit aromatase. Concomitant use of phenobarbital or valproate has a synergistic effect on aromatase inhibition. Therefore, it is important to understand the pharmacokinetics of AIs, recognizing and avoiding possible drug interactions and offering a safer prescription profile of this class of aromatase inhibitors. Arch Endocrinol Metab. 2017;61(3):391-7.


Subject(s)
Aromatase Inhibitors/pharmacokinetics , Body Height/drug effects , Nitriles/pharmacokinetics , Triazoles/pharmacokinetics , Anastrozole , Aromatase Inhibitors/therapeutic use , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Female , Humans , Letrozole , Male
14.
J Clin Pharm Ther ; 42(5): 547-553, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28868654

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: OATP1B1 mediates the transport of a diverse range of amphiphilic organic compounds that include bile acids, steroid conjugates and hormones. This retrospective pharmacogenetic study was conducted to assess the impact of the OATP1B1 c.521T>C single nucleotide polymorphism (SNP) on the pharmacokinetics of the steroidal aromatase inhibitor drug exemestane in healthy volunteers. METHODS: Exemestane (25 mg) was administered orally to 14 healthy post-menopausal women. All of the 14 subjects were sampled for pharmacokinetic (PK) analyses and retrospectively genotyped for OATP1B1 c.521T>C (rs 4149056). RESULTS AND DISCUSSION: Of the 14 subjects enrolled in the study, five were carriers of the minor C allele (OATP1B1 c.521TC+CC) and the remaining nine were carriers of the OATP1B1 c.521TT genotype. PK was assessed over 8 hours post-dosing. Our results showed statistically significant differences (P=.04) in the plasma exemestane AUC0-8 between the OATP1B1 genotype groups. Our data also showed statistically significant differences (P=.04) in the plasma AUC0-8 of 17-hydroexemestane (the major biologically active metabolite) between the OATP1B1 genotype groups. WHAT IS NEW AND CONCLUSION: Our data suggest that the OAPTP1B1 c.521T>C SNP may influence exemestane pharmacokinetics in humans.


Subject(s)
Androstadienes/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/genetics , Postmenopause , Administration, Oral , Adult , Androstadienes/administration & dosage , Area Under Curve , Aromatase Inhibitors/administration & dosage , Female , Genotype , Humans , Liver-Specific Organic Anion Transporter 1/metabolism , Middle Aged , Pharmacogenetics , Polymorphism, Single Nucleotide , Retrospective Studies
15.
Arch. endocrinol. metab. (Online) ; 61(4): 391-397, July-Aug. 2017. tab, graf
Article in English | LILACS | ID: biblio-887576

ABSTRACT

ABSTRACT Aromatase is a cytochrome P450 enzyme (CYP19A1 isoform) able to catalyze the conversion of androgens to estrogens. The aromatase gene mutations highlighted the action of estrogen as one of the main regulators of bone maturation and closure of bone plate. The use of aromatase inhibitors (AI) in boys with short stature has showed its capability to improve the predicted final height. Anastrozole (ANZ) and letrozole (LTZ) are nonsteroidal inhibitors able to bind reversibly to the heme group of cytochrome P450. In this review, we describe the pharmacokinetic profile of both drugs, discussing possible drug interactions between ANZ and LTZ with other drugs. AIs are triazolic compounds that can induce or suppress cytochrome P450 enzymes, interfering with metabolism of other compounds. Hydroxilation, N-dealkylation and glucoronidation are involved in the metabolism of AIs. Drug interactions can occur with azole antifungals, such as ketoconazole, by inhibiting CYP3A4 and by reducing the clearance of AIs. Antiepileptic drugs (lamotrigine, phenobarbital, and phenytoin) also inhibit aromatase. Concomitant use of phenobarbital or valproate has a synergistic effect on aromatase inhibition. Therefore, it is important to understand the pharmacokinetics of AIs, recognizing and avoiding possible drug interactions and offering a safer prescription profile of this class of aromatase inhibitors. Arch Endocrinol Metab. 2017;61(3):391-7.


Subject(s)
Humans , Male , Female , Triazoles/pharmacokinetics , Body Height/drug effects , Aromatase Inhibitors/pharmacokinetics , Nitriles/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Aromatase Inhibitors/therapeutic use , Drug Interactions , Letrozole , Anastrozole
16.
Rev Assoc Med Bras (1992) ; 63(4): 371-378, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28614542

ABSTRACT

Aromatase inhibitors have emerged as an alternative endocrine therapy for the treatment of hormone sensitive breast cancer in postmenopausal women. The use of third-generation inhibitors represented by exemestane, letrozol and anastrozole is currently indicated. Anastrozole is a nonsteroidal compound and a potent selective inhibitor of the aromatase enzyme. Although a few studies have shown that its pharmacodynamic and pharmacokinetic properties may be affected by interindividual variability, this drug has been recently used in all configurations of breast cancer treatment. In metastatic disease, it is currently considered the first-line treatment for postmenopausal women with estrogen receptor-positive breast tumors. Anastrozole has shown promising results in the adjuvant treatment of early-stage breast cancer in postmenopausal women. It has also achieved interesting results in the chemoprevention of the disease. Therefore, due to the importance of anastrozole both for endocrine treatment and chemoprevention of hormone-sensitive breast cancer in postmenopausal women, we proposed the current literature review in the SciELO and PubMed database of articles published in the last 10 years.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Chemoprevention/methods , Nitriles/therapeutic use , Triazoles/therapeutic use , Anastrozole , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/pharmacokinetics , Chemotherapy, Adjuvant , Female , Humans , Nitriles/pharmacokinetics , Postmenopause , Reproducibility of Results , Tamoxifen/pharmacokinetics , Tamoxifen/therapeutic use , Treatment Outcome , Triazoles/pharmacokinetics
17.
Breast Cancer Res Treat ; 165(3): 659-668, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28643023

ABSTRACT

PURPOSE: The aromatase inhibitors (AI) exemestane (EXE), letrozole (LET), and anastrozole suppress estrogen biosynthesis, and are effective treatments for estrogen receptor (ER)-positive breast cancer. Prior work suggests that anastrozole blood concentrations are associated with the magnitude of estrogen suppression. The objective of this study was to determine whether the magnitude of estrogen suppression, as determined by plasma estradiol (E2) concentrations, in EXE or LET treated patients is associated with plasma AI concentrations. METHODS: Five hundred post-menopausal women with ER-positive breast cancer were enrolled in the prospective Exemestane and Letrozole Pharmacogenetic (ELPh) Study conducted by the COnsortium on BReast cancer phArmacogomics (COBRA) and randomly assigned to either drug. Estrogen concentrations were measured at baseline and after 3 months of AI treatment and drug concentrations were measured after 1 or 3 months. EXE or LET concentrations were compared with 3-month E2 concentration or the change from baseline to 3 months using several complementary statistical procedures. RESULTS: Four-hundred patients with on-treatment E2 and AI concentrations were evaluable (EXE n = 200, LET n = 200). Thirty (7.6%) patients (EXE n = 13, LET n = 17) had 3-month E2 concentrations above the lower limit of quantification (LLOQ) (median: 4.75; range: 1.42-63.8 pg/mL). EXE and LET concentrations were not associated with on-treatment E2 concentrations or changes in E2 concentrations from baseline (all p > 0.05). CONCLUSIONS: Steady-state plasma AI concentrations do not explain variability in E2 suppression in post-menopausal women receiving EXE or LET therapy, in contrast with prior evidence in anastrozole treated patients.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Estrogens/blood , Postmenopause , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Biomarkers , Drug Monitoring , Female , Humans , Middle Aged , Time Factors
18.
Breast Cancer Res Treat ; 164(2): 411-419, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28451964

ABSTRACT

PURPOSE: Aromatase inhibitors (AI), which decrease circulating estradiol concentrations in post-menopausal women, are associated with toxicities that limit adherence. Approximately one-third of patients will tolerate a different AI after not tolerating the first. We report the effect of crossover from exemestane to letrozole or vice versa on patient-reported outcomes (PROs) and whether the success of crossover is due to lack of estrogen suppression. METHODS: Post-menopausal women enrolled on a prospective trial initiating AI therapy for early-stage breast cancer were randomized to exemestane or letrozole. Those that discontinued for intolerance were offered protocol-directed crossover to the other AI after a washout period. Changes in PROs, including pain [Visual Analog Scale (VAS)] and functional status [Health Assessment Questionnaire (HAQ)], were compared after 3 months on the first versus the second AI. Estradiol and drug concentrations were measured. RESULTS: Eighty-three patients participated in the crossover protocol, of whom 91.3% reported improvement in symptoms prior to starting the second AI. Functional status worsened less after 3 months with the second AI (HAQ mean change AI #1: 0.2 [SD 0.41] vs. AI #2: -0.05 [SD 0.36]; p = 0.001); change in pain scores was similar between the first and second AI (VAS mean change AI #1: 0.8 [SD 2.7] vs. AI #2: -0.2 [SD 2.8]; p = 0.19). No statistical differences in estradiol or drug concentrations were found between those that continued or discontinued AI after crossover. CONCLUSIONS: Although all AIs act via the same mechanism, a subset of patients intolerant to one AI report improved PROs with a different one. The mechanism of this tolerance remains unknown, but does not appear to be due to non-adherence to, or insufficient estrogen suppression by, the second AI.


Subject(s)
Androstadienes/administration & dosage , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/drug therapy , Estradiol/blood , Nitriles/administration & dosage , Triazoles/administration & dosage , Adult , Aged , Aged, 80 and over , Androstadienes/adverse effects , Androstadienes/pharmacokinetics , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/pharmacokinetics , Chemotherapy, Adjuvant/adverse effects , Cross-Over Studies , Female , Humans , Letrozole , Middle Aged , Nitriles/adverse effects , Nitriles/pharmacokinetics , Patient Reported Outcome Measures , Prospective Studies , Random Allocation , Treatment Outcome , Triazoles/adverse effects , Triazoles/pharmacokinetics
19.
Bioorg Med Chem Lett ; 27(11): 2384-2388, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28416132

ABSTRACT

We report the discovery and hit-to-lead optimization of a structurally novel indazole series of CYP11B2 inhibitors. Benchmark compound 34 from this series displays potent inhibition of CYP11B2, high selectivity versus related steroidal and hepatic CYP targets, and lead-like physical and pharmacokinetic properties. On the basis of these and other data, the indazole series was progressed to lead optimization for further refinement.


Subject(s)
Antihypertensive Agents/pharmacology , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Hypertension/drug therapy , Indazoles/pharmacology , Animals , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/pharmacokinetics , Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/pharmacokinetics , Aromatase Inhibitors/pharmacology , Cell Line , Cricetulus , Cytochrome P-450 CYP2D6 Inhibitors/chemical synthesis , Cytochrome P-450 CYP2D6 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Humans , Indazoles/chemical synthesis , Indazoles/pharmacokinetics , Macaca mulatta , Male , Rats, Sprague-Dawley , Stereoisomerism , Steroid 11-beta-Hydroxylase/antagonists & inhibitors
20.
Rev. Assoc. Med. Bras. (1992) ; 63(4): 371-378, Apr. 2017. graf
Article in English | LILACS | ID: biblio-842545

ABSTRACT

Summary Aromatase inhibitors have emerged as an alternative endocrine therapy for the treatment of hormone sensitive breast cancer in postmenopausal women. The use of third-generation inhibitors represented by exemestane, letrozol and anastrozole is currently indicated. Anastrozole is a nonsteroidal compound and a potent selective inhibitor of the aromatase enzyme. Although a few studies have shown that its pharmacodynamic and pharmacokinetic properties may be affected by interindividual variability, this drug has been recently used in all configurations of breast cancer treatment. In metastatic disease, it is currently considered the first-line treatment for postmenopausal women with estrogen receptor-positive breast tumors. Anastrozole has shown promising results in the adjuvant treatment of early-stage breast cancer in postmenopausal women. It has also achieved interesting results in the chemoprevention of the disease. Therefore, due to the importance of anastrozole both for endocrine treatment and chemoprevention of hormone-sensitive breast cancer in postmenopausal women, we proposed the current literature review in the SciELO and PubMed database of articles published in the last 10 years.


Resumo Os inibidores de aromatase têm emergido como uma endocrinoterapia alternativa para o tratamento de câncer de mama sensível a hormônios em mulheres pós-menopáusicas. A utilização de inibidores de terceira geração, representados por exemestano, letrozol e anastrozol, é atualmente indicada. Anastrozol é um composto não esteroide e um inibidor potente e seletivo da enzima aromatase. Embora alguns estudos tenham demonstrado que as suas propriedades farmacodinâmicas e farmacocinéticas podem ser afetadas pela variabilidade interindividual, esse fármaco tem sido recentemente utilizado em todas as configurações de tratamento do câncer de mama. Na doença metastática, é atualmente considerado o tratamento de primeira linha em mulheres pós-menopáusicas com tumores de mama e receptor de estrogênio positivo. O anastrozol tem mostrado resultados promissores no tratamento adjuvante do câncer de mama em estágio inicial em mulheres na pós-menopausa. Ele também conseguiu resultados interessantes na quimioprevenção da doença. Portanto, em virtude da importância do anastrozol tanto no tratamento endócrino quanto na quimioprevenção do câncer de mama hormoniossensível em mulheres na pós-menopausa, propusemos a atual revisão da literatura na base de dados SciELO e PubMed de artigos publicados nos últimos 10 anos.


Subject(s)
Humans , Female , Triazoles/therapeutic use , Breast Neoplasms/prevention & control , Breast Neoplasms/drug therapy , Chemoprevention/methods , Aromatase Inhibitors/therapeutic use , Nitriles/therapeutic use , Tamoxifen/therapeutic use , Tamoxifen/pharmacokinetics , Triazoles/pharmacokinetics , Reproducibility of Results , Treatment Outcome , Chemotherapy, Adjuvant , Postmenopause , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Anastrozole , Nitriles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...