Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 19(2): 469-79, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23087409

ABSTRACT

PURPOSE: To investigate the feasibility of adaptive dosing and the impact of pharmacogenetic variation on 13-cis-retinoic acid (13-cisRA) disposition in high-risk patients with neuroblastoma. EXPERIMENTAL DESIGN: 13-cisRA (160 mg/m(2) or 5.33 mg/kg/d) was administered to 103 patients ages 21 years or less and plasma concentrations of 13-cisRA and 4-oxo-13-cisRA quantitated on day 14 of treatment. Seventy-one patients were recruited to a dose adjustment group, targeting a 13-cisRA C(max) of 2 µmol/L, with dose increases of 25% to 50% implemented for patients with C(max) values less than 2 µmol/L. A population pharmacokinetic model was applied and polymorphisms in relevant cytochrome P450 genes analyzed. RESULTS: 13-cisRA C(max) values ranged from 0.42 to 11.2 µmol/L, with 34 of 103 (33%) patients failing to achieve a C(max) more than 2 µmol/L. Dose increases carried out in 20 patients in the dose adjustment study group led to concentrations more than 2 µmol/L in 18 patients (90%). Eight of 11 (73%) patients less than 12 kg, receiving a dose of 5.33 mg/kg, failed to achieve a C(max) of 2 µmol/L or more. Significantly, lower C(max) values were observed for patients treated with 5.33 mg/kg versus 160 mg/m(2) (1.9 ± 1.2 vs. 3.1 ± 2.0 µmol/L; mean ± SD; P = 0.023). C(max) was higher in patients who swallowed 13-cisRA capsules as compared with receiving the drug extracted from capsules (4.0 ± 2.2 vs. 2.6 ± 1.8 µmol/L; P = 0.0012). The target C(max) was achieved by 93% (25/27) versus 55% (42/76) of patients in these 2 groups, respectively. No clear relationships were found between genetic variants and 13-cisRA pharmacokinetic parameters. CONCLUSIONS: Dosing regimen and method of administration have a marked influence on 13-cisRA plasma concentrations. Body weight-based dosing should not be implemented for children less than 12 kg and pharmacologic data support higher doses for children unable to swallow 13-cisRA capsules.


Subject(s)
Antineoplastic Agents/administration & dosage , Isotretinoin/administration & dosage , Neuroblastoma/drug therapy , Adolescent , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Body Weight/drug effects , Child , Child, Preschool , Cytochrome P-450 Enzyme System/genetics , Female , Genotype , Humans , Infant , Infant, Newborn , Isotretinoin/adverse effects , Isotretinoin/pharmacokinetics , Male , Neuroblastoma/blood , Neuroblastoma/genetics , Treatment Outcome
2.
Mol Cancer Ther ; 12(1): 27-37, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23033491

ABSTRACT

We identify cytochrome P450 1A1 (CYP1A1) as a target for tumor-selective drug development in bladder cancer and describe the characterization of ICT2700, designed to be metabolized from a prodrug to a potent cytotoxin selectively by CYP1A1. Elevated CYP1A1 expression was shown in human bladder cancer relative to normal human tissues. RT112 bladder cancer cells, endogenously expressing CYP1A1, were selectively chemosensitive to ICT2700, whereas EJ138 bladder cells that do not express CYP1A1 were significantly less responsive. Introduction of CYP1A1 into EJ138 cells resulted in 75-fold increased chemosensitivity to ICT2700 relative to wild-type EJ138. Negligible chemosensitivity was observed in ICT2700 in EJ138 cells expressing CYP1A2 or with exposure of EJ138 cells to CYP1B1- or CYP3A4-generated metabolites of ICT2700. Chemosensitivity to ICT2700 was also negated in EJ138-CYP1A1 cells by the CYP1 inhibitor α-naphthoflavone. Furthermore, ICT2700 did not induce expression of the AhR-regulated CYP1 family, indicating that constitutive CYP1A1 expression is sufficient for activation of ICT2700. Consistent with the selective activity by CYP1A1 was a time and concentration-dependent increase in γ-H2AX protein expression, indicative of DNA damage, associated with the activation of ICT2700 in RT112 but not EJ138 cells. In mice-bearing CYP1A1-positive and negative isogenic tumors, ICT2700 administration resulted in an antitumor response only in the CYP1A1-expressing tumor model. This antitumor response was associated with detection of the CYP1A1-activated metabolite in tumors but not in the liver. Our findings support the further development of ICT2700 as a tumor-selective treatment for human bladder cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Transitional Cell/drug therapy , Cytochrome P-450 CYP1A1/metabolism , Indoles/pharmacology , Pyrroles/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Biotransformation , CHO Cells , Carcinoma, Transitional Cell/enzymology , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Cytochrome P-450 CYP1A1/genetics , Female , Gene Expression , Humans , Indoles/metabolism , Indoles/pharmacokinetics , Liver/metabolism , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Tumor Burden/drug effects , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 20(20): 6080-8, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22989911

ABSTRACT

The role of all-trans-retinoic acid (ATRA) in the development and maintenance of many epithelial and neural tissues has raised great interest in the potential of ATRA and related compounds (retinoids) as pharmacological agents, particularly for the treatment of cancer, skin, neurodegenerative and autoimmune diseases. The use of ATRA or prodrugs as pharmacological agents is limited by a short half-life in vivo resulting from the activity of specific ATRA hydroxylases, CYP26 enzymes, induced by ATRA in liver and target tissues. For this reason retinoic acid metabolism blocking agents (RAMBAs) have been developed for treating cancer and a wide range of other diseases. The synthesis, CYP26A1 inhibitory activity and molecular modeling studies of novel methyl 3-[4-(arylamino)phenyl]-3-(azole)-2,2-dimethylpropanoates are presented. From this series of compounds clear SAR can be derived for 4-substitution of the phenyl ring with electron-donating groups more favourable for inhibitory activity. Both the methylenedioxyphenyl imidazole (17, IC(50) = 8 nM) and triazole (18, IC(50) = 6.7 nM) derivatives were potent inhibitors with additional binding interactions between the methylenedioxy moiety and the CYP26 active site likely to be the main factor. The 6-bromo-3-pyridine imidazole 15 (IC(50) = 5.7 nM) was the most active from this series compared with the standards liarozole (IC(50) = 540 nM) and R116010 (IC(50) = 10 nM).


Subject(s)
Aminopyridines/chemical synthesis , Azoles/chemistry , Cytochrome P-450 Enzyme Inhibitors , Phenylpropionates/chemical synthesis , Propionates/chemistry , Aminopyridines/chemistry , Aminopyridines/pharmacology , Binding Sites , Catalytic Domain , Cell Survival/drug effects , Cytochrome P-450 Enzyme System/metabolism , Humans , Imidazoles , MCF-7 Cells , Microsomes/metabolism , Molecular Docking Simulation , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Propionates/chemical synthesis , Propionates/pharmacology , Retinoic Acid 4-Hydroxylase , Structure-Activity Relationship , Tretinoin/pharmacology , Triazoles/chemistry
4.
Bioorg Med Chem ; 20(14): 4201-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22727372

ABSTRACT

Retinoic acid (RA), the biologically active metabolite of vitamin A, is used medicinally for the treatment of hyperproliferative diseases including dermatological conditions and cancer. The antiproliferative effects of RA have been well documented as well as the limitations owing to toxicity and the development of resistance to RA therapy. RA metabolism inhibitors (RAMBAs or CYP26 inhibitors) are attracting increasing interest as an alternative method for enhancing endogenous levels of retinoic acid in the treatment of hyperproliferative disease. Here the synthesis and inhibitory activity of novel 3-(1H-imidazol- and triazol-1-yl)-2,2-dimethyl-3-(4-(phenylamino)phenyl)propyl derivatives in a MCF-7 CYP26A1 microsomal assay are described. The most promising inhibitor methyl 2,2-dimethyl-3-(4-(phenylamino)phenyl)-3-(1H-1,2,4-triazol-1-yl)propanoate (6) exhibited an IC(50) of 13 nM (compared with standards Liarozole IC(50) 540 nM and R116010 IC(50) 10 nM) and was further evaluated for CYP selectivity using a panel of CYP with >100-fold selectivity for CYP26 compared with CYP1A2, 2C9 and 2D6 observed and 15-fold selectivity compared with CYP3A4. The results demonstrate the potential for further development of these potent inhibitors.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Propionates/chemistry , Triazoles/chemistry , Cell Line, Tumor , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemical synthesis , Esters , Humans , Propionates/chemical synthesis , Retinoic Acid 4-Hydroxylase , Structure-Activity Relationship , Triazoles/chemical synthesis
5.
Oncol Rep ; 27(1): 293-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21964808

ABSTRACT

Despite the successful introduction of 13-cis retinoic acid (13cisRA) therapy for the treatment of neuroblastoma, approximately 50% patients do not respond or experience relapse. A retinoid analogue, fenretinide [N-(4-hydroxyphenyl) retinamide; 4-HPR] can induce apoptosis in neuroblastoma cell lines and could have clinical use after therapy with 13cisRA. However, there are important questions concerning potential retinoid drug interactions which need to be addressed. The aim of this study was to investigate the influence of retinoic acid pre-treatment on fenretinide-induced apoptosis and fenretinide metabolism in neuroblastoma cell lines. Apoptosis was measured by flow cytometry of propidium iodide-stained neuroblastoma cells and a live-cell imaging assay. Intracellular fenretinide metabolism was determined by HPLC analysis. Pre-treatment of neuroblastoma cell lines with retinoic acid (RA) resulted in a significant decrease in the apoptotic response to fenretinide in three of the four lines tested. Comparison between responsive and non-responsive cell lines suggested that RA sensitivity was required to promote fenretinide resistance, and that this was mediated by up-regulation of Bcl-2 and the inhibition of pro-apoptotic fenretinide signalling pathways. Induction of the oxidative metabolism of fenretinide after RA pre-treatment did not significantly impact on intracellular parent drug levels and is unlikely to explain the decreased apoptotic response observed. The interaction between RA and fenretinide could have important implications for the scheduling of fenretinide in therapeutic protocols for neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fenretinide/pharmacology , Neuroblastoma/metabolism , Tretinoin/pharmacology , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Interactions , Fenretinide/metabolism , Flow Cytometry , Humans , Neuroblastoma/pathology
6.
Nanomedicine ; 8(6): 880-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22094120

ABSTRACT

This study reports on the preparation and evaluation of amphiphilic macromolecules based on branched polyethylene glycol covalently linked with alkyl hydrocarbon chains. These macromolecules easily dissolved in an aqueous environment, with formation of micellar nanoaggregates endowed with hydrophobic inner cores capable of hosting fenretinide by complexation. The complexes increased fenretinide aqueous solubility, while hindering its release as a free drug in an aqueous environment. Particle size analysis indicated dimensional suitability of the complexes for intravenous administration. Neuroblastoma cell lines (SH-SY5Y and NGP) exhibited increased sensitivity to fenretinide in complex as compared to free drug, associated with higher intracellular concentrations of fenretinide observed after treatment with the complex. Transmission electronic microscopy images revealed endocytosis of the micellar complex. Moreover, fenretinide conversion to its metabolite 4-oxo-fenretinide was delayed in cells treated with the complex, further supporting the hypothesis that fenretinide may be absorbed by micellar transport and exposed to the cytoplasm for conversion to its metabolite only after micelle destabilization.


Subject(s)
Fenretinide/administration & dosage , Fenretinide/chemistry , Nanocapsules/chemistry , Neuroblastoma/drug therapy , Neuroblastoma/physiopathology , Polyethylene Glycols/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Crystallization/methods , Drug Design , Humans , Micelles , Neuroblastoma/pathology
7.
Drug Metab Dispos ; 38(7): 1211-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20308471

ABSTRACT

13-cis Retinoic acid (13cisRA, isotretinoin) is an important drug in both dermatology, and the treatment of high-risk neuroblastoma. 13cisRA is known to undergo cytochrome P450-mediated oxidation, mainly by CYP2C8, but phase II metabolic pathways have not been characterized. In the present study, the glucuronidation activities of human liver (HLM) and intestinal microsomes (HIM), as well as a panel of human UDP-glucuronosyltransferases (UGTs) toward both 13cisRA and the 4-oxo metabolite, 4-oxo 13cisRA, were compared using high-performance liquid chromatography. Both HLM and, to a greater extent, HIM catalyzed the glucuronidation of 13cisRA and 4-oxo 13cisRA. Based on the structures of 13cisRA and 4-oxo 13cisRA, the glucuronides formed are conjugated at the terminal carboxylic acid. Further analysis revealed that UGT1A1, UGT1A3, UGT1A7, UGT1A8, and UGT1A9 were the major isoforms responsible for the glucuronidation of both substrates. For 13cisRA, a pronounced substrate inhibition was observed with individual UGTs and with HIM. UGT1A3 exhibited the highest rate of activity toward both substrates, and a high rate of activity toward 13cisRA glucuronidation was also observed with UGT1A7. However, for both substrates, K(m) values were above concentrations reported in clinical studies. Therefore, UGT1A9 is likely to be the most important enzyme in the glucuronidation of both substrates as this enzyme had the lowest K(m) and is expressed in both the intestine and at high levels in the liver.


Subject(s)
Glucuronosyltransferase/metabolism , Isoenzymes/metabolism , Isotretinoin/pharmacokinetics , Tretinoin/analogs & derivatives , Glucuronides/pharmacokinetics , Glucuronosyltransferase/antagonists & inhibitors , Humans , In Vitro Techniques , Intestinal Mucosa/metabolism , Kinetics , Microsomes/enzymology , Microsomes, Liver/enzymology , Substrate Specificity , Tretinoin/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...