Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Immunother Cancer ; 11(7)2023 07.
Article in English | MEDLINE | ID: mdl-37419511

ABSTRACT

BACKGROUND: Most immunotherapies approved for clinical use rely on the use of recombinant proteins and cell-based approaches, rendering their manufacturing expensive and logistics onerous. The identification of novel small molecule immunotherapeutic agents might overcome such limitations. METHOD: For immunopharmacological screening campaigns, we built an artificial miniature immune system in which dendritic cells (DCs) derived from immature precursors present MHC (major histocompatibility complex) class I-restricted antigen to a T-cell hybridoma that then secretes interleukin-2 (IL-2). RESULTS: The screening of three drug libraries relevant to known signaling pathways, FDA (Food and Drug Administration)-approved drugs and neuroendocrine factors yielded two major hits, astemizole and ikarugamycin. Mechanistically, ikarugamycin turned out to act on DCs to inhibit hexokinase 2, hence stimulating their antigen presenting potential. In contrast, astemizole acts as a histamine H1 receptor (H1R1) antagonist to activate T cells in a non-specific, DC-independent fashion. Astemizole induced the production of IL-2 and interferon-γ (IFN-γ) by CD4+ and CD8+ T cells both in vitro and in vivo. Both ikarugamycin and astemizole improved the anticancer activity of the immunogenic chemotherapeutic agent oxaliplatin in a T cell-dependent fashion. Of note, astemizole enhanced the CD8+/Foxp3+ ratio in the tumor immune infiltrate as well as IFN-γ production by local CD8+ T lymphocytes. In patients with cancer, high H1R1 expression correlated with low infiltration by TH1 cells, as well as with signs of T-cell exhaustion. The combination of astemizole and oxaliplatin was able to cure the majority of mice bearing orthotopic non-small cell lung cancers (NSCLC), then inducing a state of protective long-term immune memory. The NSCLC-eradicating effect of astemizole plus oxaliplatin was lost on depletion of either CD4+ or CD8+ T cells, as well as on neutralization of IFN-γ. CONCLUSIONS: These findings underscore the potential utility of this screening system for the identification of immunostimulatory drugs with anticancer effects.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-2 , United States , Mice , Animals , Interleukin-2/metabolism , Astemizole/pharmacology , Astemizole/therapeutic use , Astemizole/metabolism , Oxaliplatin , Immunity, Cellular , Histocompatibility Antigens Class I , Interferon-gamma/metabolism
2.
Phytomedicine ; 42: 199-206, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29655687

ABSTRACT

BACKGROUND: Broussonetia papyrifera (L.) Ventenat, a traditional medicinal herb, has been applied as a folk medicine to treat various diseases. Broussochalcone A (BCA), a chalcone compound isolated from the cortex of Broussonetia papyrifera (L.) Ventenat, exhibits several biological activities including potent anti-oxidant, antiplatelet, and cytotoxic effects. PURPOSE: The purpose of this study is to elucidate the inhibitory effect of BCA against CYP2J2 enzyme which is predominantly expressed in human tumor tissues and carcinoma cell lines. STUDY DESIGN: The inhibitory effect of BCA on the activities of CYP2J2-mediated metabolism were investigated using human liver microsomes (HLMs), and its anti-cancer effect against human hepatoma HepG2 cells was also evaluated. METHODS: Two representative CYP2J2-specific probe substrates, astemizole and ebastine, were incubated in HLMs with BCA. After incubation, the samples were analyzed using liquid chromatography-tandem mass spectrometry. To investigate the binding model between BCA and CYP2J2, we carried out structure-based docking simulations by using software and scripts written in-house. RESULTS: BCA inhibited CYP2J2-mediated astemizole O-demethylation and ebastine hydroxylase activities in a concentration dependent manner with Ki values of 2.3 and 3.7 µM, respectively. It also showed cytotoxic effects against human hepatoma HepG2 cells in a dose-dependent manner with activation of apoptosis related proteins. CONCLUSION: Overall, this was the first report of the inhibitory effects of BCA on CYP2J2 in HLMs. The present data suggest that BCA is a potential candidate for further evaluation for its CYP2J2 targeting anti-cancer activities.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Chalcones/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Forkhead Box Protein O3/metabolism , Resorcinols/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Astemizole/metabolism , Butyrophenones/metabolism , Cell Proliferation/drug effects , Chalcones/administration & dosage , Chalcones/chemistry , Chromatography, Liquid , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Cytochrome P-450 Enzyme System/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Piperidines/metabolism , Resorcinols/administration & dosage , Resorcinols/chemistry , Tandem Mass Spectrometry
3.
Xenobiotica ; 46(11): 977-85, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26899760

ABSTRACT

1. Common marmoset (Callithrix jacchus), a New World Monkey, has potential to be a useful animal model in preclinical studies. However, drug metabolizing properties have not been fully understood due to insufficient information on cytochrome P450 (P450), major drug metabolizing enzymes. 2. Marmoset P450 2J2 cDNA was isolated from marmoset livers. The deduced amino acid sequence showed a high-sequence identity (91%) with cynomolgus monkey and human P450 2J2 enzymes. A phylogenetic tree revealed that marmoset P450 2J2 was evolutionarily closer to cynomolgus monkey and human P450 2J2 enzymes, than P450 2J forms in pigs, rabbits, rats or mice. 3. Marmoset P450 2J2 mRNA was abundantly expressed in the small intestine and liver, and to a lesser extent in the brain, lung and kidney. Immunoblot analysis also showed expression of marmoset P450 2J2 protein in the small intestine and liver. 4. Enzyme assays using marmoset P450 2J2 protein heterologously expressed in Escherichia coli indicated that marmoset P450 2J2 effectively catalyzed astemizole O-demethylation and terfenadine t-butyl hydroxylation, similar to human and cynomolgus monkey P450 2J2 enzymes. 5. These results suggest the functional characteristics of P450 2J2 enzymes are similar among marmosets, cynomolgus monkeys and humans.


Subject(s)
Astemizole/metabolism , Cytochrome P-450 Enzyme System/metabolism , Histamine H1 Antagonists, Non-Sedating/metabolism , Macaca fascicularis/metabolism , Terfenadine/metabolism , Animals , Cytochrome P-450 CYP2J2 , Humans , Intestine, Small/metabolism , Liver/metabolism , Mice , Rats
4.
Eur J Pharmacol ; 764: 480-488, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26209360

ABSTRACT

Cytochrome P450 2J2 (CYP2J2) is highly expressed in human tumors and carcinoma cell lines, and has been implicated in the pathogenesis of human cancers. The aim of this study was to identify a compound that could inhibit the activity of CYP2J2, and to examine its anticancer activity. To identify CYP2J2 inhibitors, 10 terpenoids obtained from plants were screened using astemizole as a CYP2J2 probe substrate in human liver microsomes (HLMs). Of these, tanshinone IIA dose-dependently and non-competitively inhibited CYP2J2-mediated astemizole O-demethylation activity. Tanshinone IIA significantly decreased viability of human hepatoma HepG2 cells and SiHa cervical cancer cells; however, it was not cytotoxic against mouse hepatocytes. Furthermore, treatment of cells with tanshinone IIA significantly increased apoptotic cell death rate, as shown by the increase in Annexin V-stained cell populations, Bcl-2 associated X protein (Bax)/B-cell lymphoma 2 (Bcl-2) ratio, and poly (ADP-ribose) polymerase 1 (PARP-1) cleavage in HepG2 cells. Furthermore, the results of this study showed that tanshinone IIA significantly decreased HepG2 cell-based tumor growth in nude mice in a dose-dependent manner. On the other hand, the tanshinone IIA-induced apoptotic cell death rate was significantly attenuated by enhanced up-regulation of CYP2J2 expression. Thus, our data strongly suggest that tanshinone IIA exerts its anticancer effect by inhibiting CYP2J2 activity.


Subject(s)
Abietanes/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Liver Neoplasms/drug therapy , Animals , Apoptosis Regulatory Proteins/metabolism , Astemizole/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/genetics , Dealkylation , Dose-Response Relationship, Drug , Female , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Species Specificity , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Microb Cell Fact ; 14: 15, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25656388

ABSTRACT

The hERG potassium channel is essential for repolarization of the cardiac action potential. Due to this vital function, absence of unintended and potentially life-threatening interactions with hERG is required for approval of new drugs. The structure of hERG is therefore one of the most sought-after. To provide purified hERG for structural studies and new hERG biomimetic platforms for detection of undesirable interactions, we have developed a hERG expression platform generating unprecedented amounts of purified and functional hERG channels. Full-length hERG, with or without a C-terminally fused green fluorescent protein (GFP) His 8-tag was produced from a codon-optimized hERG cDNA in Saccharomyces cerevisiae. Both constructs complemented the high potassium requirement of a knock-out Saccharomyces cerevisiae strain, indicating correct tetramer assembly in vivo. Functionality was further demonstrated by Astemizole binding to membrane embedded hERG-GFP-His 8 with a stoichiometry corresponding to tetramer assembly. The 156 kDa hERG-GFP protein accumulated to a membrane density of 1.6%. Fluorescence size exclusion chromatography of hERG-GFP-His 8 solubilized in Fos-Choline-12 supplemented with cholesteryl-hemisuccinate and Astemizole resulted in a monodisperse elution profile demonstrating a high quality of the hERG channels. hERG-GFP-His 8 purified by Ni-affinity chromatography maintained the ability to bind Astemizole with the correct stoichiometry indicating that the native, tetrameric structure was preserved. To our knowledge this is the first reported high-yield production and purification of full length, tetrameric and functional hERG. This significant breakthrough will be paramount in obtaining hERG crystal structures, and in establishment of new high-throughput hERG drug safety screening assays.


Subject(s)
Astemizole/metabolism , Ether-A-Go-Go Potassium Channels/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Biomass , Cell Membrane/metabolism , Chromatography, Affinity/methods , DNA, Complementary/genetics , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/isolation & purification , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histamine H1 Antagonists, Non-Sedating/metabolism , Humans , Microscopy, Fluorescence , Protein Binding , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Saccharomyces cerevisiae/genetics , Temperature , Time Factors
6.
Drug Metab Dispos ; 42(10): 1698-707, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25074871

ABSTRACT

Cytochrome P450 4F12 is a drug-metabolizing enzyme that is primarily expressed in the liver, kidney, colon, small intestine, and heart. The properties of CYP4F12 that may impart an increased catalytic selectivity (decreased promiscuity) were explored through in vitro metabolite elucidation, kinetic isotope effect experiments, and computational modeling of the CYP4F12 active site. By using astemizole as a probe substrate for CYP4F12 and CYP3A4, it was observed that although CYP4F12 favored astemizole O-demethylation as the primary route of metabolism, CYP3A4 was capable of metabolizing astemizole at multiple sites on the molecule. Deuteration of astemizole at the site of O-demethylation resulted in an isotope effect of 7.1 as well as an 8.3-fold decrease in the rate of clearance for astemizole by CYP4F12. Conversely, although an isotope effect of 3.8 was observed for the formation of the O-desmethyl metabolite when deuterated astemizole was metabolized by CYP3A4, there was no decrease in the clearance of astemizole. Development of a homology model of CYP4F12 based on the crystal structure of cytochrome P450 BM3 predicted an active site volume for CYP4F12 that was approximately 76% of the active site volume of CYP3A4. As predicted, multiple favorable binding orientations were available for astemizole docked into the active site of CYP3A4, but only a single binding orientation with the site of O-demethylation oriented toward the heme was identified for CYP4F12. Overall, it appears that although CYP4F12 may be capable of binding similar ligands to other cytochrome P450 enzymes such as CYP3A4, the ability to achieve catalytically favorable orientations may be inherently more difficult because of the increased steric constraints of the CYP4F12 active site.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Aryl Hydrocarbon Hydroxylases/metabolism , Astemizole/metabolism , Biotransformation , Catalytic Domain , Cytochrome P-450 CYP3A/metabolism , Humans , In Vitro Techniques , Models, Molecular , Substrate Specificity
7.
Drug Metab Dispos ; 40(5): 943-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22328583

ABSTRACT

CYP2J2, an arachidonic acid epoxygenase, is recognized for its role in the first-pass metabolism of astemizole and ebastine. To fully assess the role of CYP2J2 in drug metabolism, a selective substrate and potent specific chemical inhibitor are essential. In this study, we report amiodarone 4-hydoxylation as a specific CYP2J2-catalyzed reaction with no CYP3A4, or other drug-metabolizing enzyme, involvement. Amiodarone 4-hydroxylation enabled the determination of liver relative activity factor and intersystem extrapolation factor for CYP2J2. Amiodarone 4-hydroxylation correlated with astemizole O-demethylation but not with CYP2J2 protein content in a sample of human liver microsomes. To identify a specific CYP2J2 inhibitor, 138 drugs were screened using terfenadine and astemizole as probe substrates with recombinant CYP2J2. Forty-two drugs inhibited CYP2J2 activity by ≥50% at 30 µM, but inhibition was substrate-dependent. Of these, danazol was a potent inhibitor of both hydroxylation of terfenadine (IC(50) = 77 nM) and O-demethylation of astemizole (K(i) = 20 nM), and inhibition was mostly competitive. Danazol inhibited CYP2C9, CYP2C8, and CYP2D6 with IC(50) values of 1.44, 1.95, and 2.74 µM, respectively. Amiodarone or astemizole were included in a seven-probe cocktail for cytochrome P450 (P450) drug-interaction screening potential, and astemizole demonstrated a better profile because it did not appreciably interact with other P450 probes. Thus, danazol, amiodarone, and astemizole will facilitate the ability to determine the metabolic role of CYP2J2 in hepatic and extrahepatic tissues.


Subject(s)
Amiodarone/metabolism , Astemizole/metabolism , Cytochrome P-450 Enzyme System/metabolism , Danazol , Enzyme Inhibitors , Microsomes, Liver/enzymology , Terfenadine/metabolism , Amiodarone/chemistry , Astemizole/chemistry , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2J2 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Danazol/chemistry , Danazol/metabolism , Danazol/pharmacology , Drug Discovery , Drug Interactions , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydroxylation , In Vitro Techniques , Methylation , Microsomes, Liver/metabolism , Models, Biological , Molecular Structure , Substrate Specificity , Tandem Mass Spectrometry , Terfenadine/chemistry
8.
Drug Metab Pharmacokinet ; 26(3): 300-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21383522

ABSTRACT

The oral bioavailability of some drugs is markedly lower in cynomolgus monkeys than in humans. One of the reasons for the low bioavailability in cynomolgus monkeys may be the higher metabolic activity of intestinal CYP3A; however, the species differences in intestinal metabolic activities of other CYP isoforms between cynomolgus monkeys and humans are not well known. In the present study, we investigated the intrinsic clearance (CL(int)) values in pooled intestinal microsomes from cynomolgus monkeys and humans using 25 substrates of human CYP1A2, CYP2J2, CYP2C, and CYP2D6. As in humans, intestinal CL(int) values of human CYP1A2 and CYP2D6 substrates in cynomolgus monkeys were low. On the other hand, intestinal CL(int) values of human CYP2J2 and CYP2C substrates in cynomolgus monkeys were greatly higher than those in humans. Using immunoinhibitory antibodies and chemical inhibitors, we showed that the higher intestinal CL(int) values of the human CYP2J2 and CYP2C substrates in cynomolgus monkeys might be caused by monkey CYP4F and CYP2C subfamily members, respectively. In conclusion, there is a possibility that the greatly higher metabolic activity of CYP2C and CYP4F in cynomolgus monkey intestine is one of the causes of the species difference of intestinal first-pass metabolism between cynomolgus monkeys and humans.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Intestines/enzymology , Macaca fascicularis/metabolism , Pharmaceutical Preparations/metabolism , 2-Pyridinylmethylsulfinylbenzimidazoles/metabolism , Amodiaquine/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Astemizole/metabolism , Biocatalysis/drug effects , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2J2 , Cytochrome P-450 CYP3A/immunology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/immunology , Fatty Acids, Unsaturated/pharmacology , Humans , Isoenzymes/metabolism , Lansoprazole , Microsomes/drug effects , Microsomes/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Nicardipine/metabolism , Nimodipine/metabolism , Paroxetine/metabolism , Species Specificity , Terfenadine/metabolism
9.
Methods Mol Biol ; 656: 147-58, 2010.
Article in English | MEDLINE | ID: mdl-20680589

ABSTRACT

Matrix-assisted laser desorption/ionization-tandem mass spectrometric method (MALDI-MS/MS) has proven to be a reliable tool for direct measurement of the disposition of small molecules in animal tissue sections. As example, MALDI-MS/MS imaging system was employed for visualizing the spatial distribution of astemizole and its primary metabolite in rat brain tissues. Astemizole is a second-generation antihistamine, a block peripheral H1 receptor, which was introduced to provide comparable therapeutic benefit but was withdrawn in most countries due to toxicity risks. Astemizole was observed to be heterogeneously distributed to most parts of brain tissue slices including cortex, hippocampus, hypothalamic, thalamus, and ventricle regions while its major metabolite, desmethylastemizole, was only found around ventricle sites. We have shown that astemizole alone is likely to be responsible for the central nervous system (CNS) side effects when its exposures became elevated.


Subject(s)
Diagnostic Imaging/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Astemizole/metabolism , Astemizole/pharmacokinetics , Brain/metabolism , Molecular Structure , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
10.
J Cardiovasc Pharmacol ; 55(4): 368-76, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20125032

ABSTRACT

In vitro hERG blocking potency is measured in drug discovery as part of an integrated cardiovascular risk assessment. Typically, the concentrations producing 50% inhibition are measured in protein-free saline solutions and compared with calculated free therapeutic in vivo Cmax values to estimate a hERG safety multiple. The free/unbound fraction is believed responsible for activity. We tested the validity of this approach with 12 compounds by determining potencies in voltage clamp studies conducted in the absence and presence of 100% dialyzed fetal bovine serum (FBS). Bath drug concentrations in saline solutions were measured to account for loss of compounds due to solubility, stability, and/or adsorption. Protein binding in dialyzed FBS was measured to enable predictions of serum IC50s based on the unbound fraction and the saline IC50. For 11 of 12 compounds, the measured potency in the presence of dialyzed FBS was within 2-fold of the predicted potency. The predicted IC50 in dialyzed FBS for one highly bound compound, amiodarone, was 9-fold higher than the measured serum IC50. These data suggest that for highly bound compounds, direct measurement of IC50s in the presence of 100% serum may provide a more accurate estimate of in vivo potencies than the approach based on calculated serum shifts.


Subject(s)
Blood Proteins/metabolism , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ion Channel Gating/drug effects , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Amiodarone/metabolism , Amiodarone/pharmacology , Animals , Astemizole/metabolism , Astemizole/pharmacology , Cattle , Cell Line , Cisapride/metabolism , Cisapride/pharmacology , Dialysis , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/physiology , Fluvoxamine/metabolism , Fluvoxamine/pharmacology , Humans , Ion Channel Gating/physiology , Mice , Patch-Clamp Techniques , Protein Binding/physiology , Serum/metabolism , Sodium Chloride , Thioridazine/metabolism , Thioridazine/pharmacology , Transfection
11.
Chemistry ; 15(43): 11723-9, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19774562

ABSTRACT

Herein we demonstrate that a small panel of variants of cytochrome P450 BM3 from Bacillus megaterium covers the breadth of reactivity of human P450s by producing 12 of 13 mammalian metabolites for two marketed drugs, verapamil and astemizole, and one research compound. The most active enzymes support preparation of individual metabolites for preclinical bioactivity and toxicology evaluations. Underscoring their potential utility in drug lead diversification, engineered P450 BM3 variants also produce novel metabolites by catalyzing reactions at carbon centers beyond those targeted by animal and human P450s. Production of a specific metabolite can be improved by directed evolution of the enzyme catalyst. Some variants are more active on the more hydrophobic parent drug than on its metabolites, which limits production of multiply-hydroxylated species, a preference that appears to depend on the evolutionary history of the P450 variant.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Pharmaceutical Preparations/metabolism , Astemizole/chemistry , Astemizole/metabolism , Bacillus megaterium/enzymology , Biocatalysis , Catalytic Domain , Chromones/chemistry , Chromones/metabolism , Cytochrome P-450 Enzyme System/genetics , Evolution, Molecular , Humans , Morpholines/chemistry , Morpholines/metabolism , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Verapamil/chemistry , Verapamil/metabolism
12.
Bioanalysis ; 1(2): 299-307, 2009 May.
Article in English | MEDLINE | ID: mdl-21083169

ABSTRACT

BACKGROUND: Matrix-assisted laser desorption/ionization (MALDI)-tandem mass spectrometry (MS)/MS is a proven reliable tool for visualizing the spatial distribution of dosed drugs and their primary metabolites in animal tissue sections. MATERIALS & METHODS: The rat brain tissue sections coated with dihydroxybenzoic acid as matrix, were analyzed by MALDI-MS/MS imaging experiments. The potential metabolites of astemizole in rat brain homogenate selected for MALDI-MS/MS imaging experiments were first identified by high-performance liquid chromatography coupled to an electrospray ionization source and a hybrid-quadrupole-linear-ion-trap mass spectrometer. RESULTS: Astemizole was observed to be heterogeneously distributed to most parts of the brain tissue slices including the cortex, hippocampus, hypothalamic, thalamus and ventricle regions, while its major metabolite, desmethylastemizole, was only found around ventricle sites. CONCLUSION: The results indicated that the dosed compound alone might be responsible for the CNS side-effects when drug exposures became elevated.


Subject(s)
Astemizole/analogs & derivatives , Astemizole/analysis , Brain Chemistry , Histamine H1 Antagonists, Non-Sedating/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Astemizole/metabolism , Cerebral Ventricles/chemistry , Chromatography, High Pressure Liquid , Diagnostic Imaging , Histamine H1 Antagonists, Non-Sedating/analysis , Histamine H1 Antagonists, Non-Sedating/metabolism , Rats , Spectrometry, Mass, Electrospray Ionization
13.
Methods Find Exp Clin Pharmacol ; 29(7): 457-65, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17982510

ABSTRACT

Blockade of human ether-a-go-go-related gene (hERG) potassium channels is an undesirable activity of many drugs because it may be the primary cause for the prolongation of the QT interval, which appears to be associated with the induction of potentially life-threatening ventricular arrhythmias. Despite the fact that several antipsychotic drugs exert hERG-blocking activity that may underlie their propensity to cause electrocardiogram (ECG) abnormalities, including QT interval prolongation and associated ventricular arrhythmias, a considerable number of available typical and atypical antipsychotics have not been characterized for hERG-blocking activity. This study has evaluated the hERG-blocking activity of several of these antipsychotic drugs in human embryonic kidney cells (HEK293) stably transfected with the human recombinant hERG potassium channel and using a high throughput whole-cell patch-clamp technique to determine their respective IC(50) values. The ability of antipsychotics to competitively displace [(3)H]-astemizole binding from hERG-transfected HEK293 cells was also examined to properly establish the concentration range for testing in the subsequent patch clamp assays. The results showed that most of the tested antipsychotics effectively blocked the hERG channel in a dose-dependent manner, with IC(50) values ranging from 3.37 nM for the butyrophenone benperidol to >100 microM for the substituted benzamide sulpiride. Markedly, differential potencies were obtained depending on the chemical class, so that butyrophenones and diphenylbutylpiperidines emerged as the most potent hERG-blocking antipsychotics, while dibenzoxazepines and, particularly, substituted benzamides, exhibited the lowest activity. Because the hERG-blocking properties of drugs appear to be associated with their ability to prolong the QT interval, the present results will enable the establishment of the potential arrhythmogenic risk for each antipsychotic evaluated. Thus, their cardiotoxic risk will be discussed on the basis of their hERG-blocking properties and previous clinical data when available.


Subject(s)
Antipsychotic Agents/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Astemizole/metabolism , Binding, Competitive , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Electrocardiography/drug effects , Humans
14.
Nat Chem Biol ; 2(8): 415-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16816845

ABSTRACT

The high cost and protracted time line of new drug discovery are major roadblocks to creating therapies for neglected diseases. To accelerate drug discovery we created a library of 2,687 existing drugs and screened for inhibitors of the human malaria parasite Plasmodium falciparum. The antihistamine astemizole and its principal human metabolite are promising new inhibitors of chloroquine-sensitive and multidrug-resistant parasites, and they show efficacy in two mouse models of malaria.


Subject(s)
Antimalarials/pharmacology , Astemizole/analogs & derivatives , Astemizole/pharmacology , Plasmodium falciparum/drug effects , Animals , Antimalarials/adverse effects , Antimalarials/metabolism , Astemizole/adverse effects , Astemizole/metabolism , Chloroquine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance , Drug Resistance, Multiple , Humans , Mice , Plasmodium yoelii/drug effects
15.
Pharmacogenet Genomics ; 15(2): 105-13, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15861034

ABSTRACT

CYP2J2 plays important roles in the metabolism of therapeutic drugs, such as astemizole and ebastine, as well as endogenous fatty acids. This study aimed to identify CYP2J2 genetic variants in Koreans and to characterize their functional consequences. From direct sequencing of the CYP2J2 gene, 12 genetic variations, including the two novel nonsynonymous mutations G312R and P351L, were identified from 93 Korean subjects. The two novel CYP2J2 variants were co-expressed with NADPH-cytochrome P450 reductase in Sf9 cells and their catalytic activities were quantified. The recombinant CYP2J2 G312R variant showed almost complete loss of enzymatic activity, as determined by CYP2J2-catalysed astemizole O-demethylation and ebastine hydroxylation. The CYP2J2 P351L variant showed enzymatic activities that were comparable with the wild-type CYP2J2. The reduced CO spectra of the recombinant CYP2J2 proteins suggested no CO binding to the heme in CYP2J2 G312R. In addition, molecular modelling of the three-dimensional structure consistently predicted that there might be spatial hindrance between heme and the bulky side chain of the R312 residue in CYP2J2 G312R variant. The CYP2J2 G312R variant was not found in 192 Chinese, 99 African-Americans, 100 Caucasians and 159 Vietnamese subjects. Two of the 192 Chinese subjects (0.52%) were heterozygous for CYP2J2 P351L. Twelve CYP2J2 variants, including two novel nonsynonymous variants, were identified in a Korean population. The G312R variant is the first nonfunctional CYP2J2 allele to be identified, and is expected to influence the disposition of its substrate therapeutics, as well as endogenous compounds.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Mutation , Oxygenases/genetics , Alleles , Animals , Astemizole/metabolism , Astemizole/pharmacology , Butyrophenones/metabolism , Catalysis , Cell Line , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/physiology , DNA Primers/chemistry , Dose-Response Relationship, Drug , Gene Frequency , Genetic Variation , Heterozygote , Histamine H1 Antagonists/pharmacology , Humans , Insecta , Kinetics , Korea , Methylation , Models, Molecular , Oxygenases/chemistry , Oxygenases/physiology , Piperidines/metabolism , Polymerase Chain Reaction , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Recombinant Proteins/chemistry , Recombination, Genetic , Sequence Analysis, DNA , Time Factors
16.
J Pharmacol Sci ; 95(3): 311-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15272206

ABSTRACT

A radioligand binding assay for the HERG (human ether-a-go-go-related gene) K(+) channel was developed to identify compounds which may have inhibitory activity and potential cardiotoxicity. Pharmacological characterization of the [(3)H]astemizole binding assay for HERG K(+) channels was performed using HERG-expressing HEK293 cells. The assay conditions employed yielded 90% specific binding using 10 microg/well of membrane protein with 1.5 nM of [(3)H]astemizole at 25 degrees C. The K(d) and B(max) values were 5.91 +/- 0.81 nM and 6.36 +/- 0.26 pmol/mg, respectively. The intraassay and interassay variations were 11.4% and 14.9%, respectively. Binding affinities for 32 reference compounds (including dofetilide, cisapride, and terfenadine) with diverse structures demonstrated a similar potency rank order for HERG inhibition to that reported in the literature. Moreover, the [(3)H]astemizole binding data demonstrated a rank order of affinity that was highly correlated to that of inhibitory potency in the electrophysiological studies for HERG in HEK293 (r(SP) = 0.91, P<0.05). In conclusion, the [(3)H]astemizole binding assay is rapid and capable of detecting HERG inhibitors.


Subject(s)
Astemizole/pharmacology , Calcium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Astemizole/metabolism , Binding Sites , Binding, Competitive , Calcium Channel Blockers/metabolism , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Humans , Ligands , Patch-Clamp Techniques , Radioligand Assay , Time Factors , Transfection , Tritium
17.
Xenobiotica ; 33(6): 615-23, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12851038

ABSTRACT

1. The effects of chemical agents on the metabolism of the antihistamine drug astemizole were investigated to evaluate drug-drug interactions. 2. Chemical inhibitors of astemizole O-demethylation were screened using the small intestinal and liver microsomes from rabbit as an animal model for the first-pass metabolism of humans. In the rabbit small intestine, astemizole O-demethylation was clearly inhibited by ebastine, arachidonic acid, alpha-naphthoflavone, ketoconazole, tranylcypromine, troglitazone and terfenadine. 3. In humans, these inhibitors also reduced microsomal astemizole O-demethylation in both the small intestine and liver. However, the inhibition rate of almost all these chemicals were clearly greater in the small intestine than in the liver. Thus, a different contribution of cytochrome p450 in each tissue is suggested. 4. All the chemicals inhibited astemizole O-demethylation in recombinant CYP2J2 microsomes. The results suggest that CYP2J2 is involved in astemizole O-demethylation in both the human small intestine and liver; however, the contribution in the liver is lower than in the small intestine. The effects of the CYP2J2 inhibitors during first-pass metabolism may be more important in the small intestine than in the liver. Since all the inhibition profiles of astemizole O-demethylation were different in the liver and small intestine, involvement of another p450 in astemizole O-demethylation in human liver may be speculated. 5. In the rabbit microsomal systems, the same metabolites found in humans were qualitatively detected and the inhibition profiles of the chemical agents in the microsomes resembled that of humans.


Subject(s)
Astemizole/metabolism , Cytochrome P-450 Enzyme System/metabolism , Histamine H1 Antagonists/metabolism , Intestine, Small/metabolism , Liver/enzymology , Microsomes, Liver/enzymology , Oxygenases/metabolism , Animals , Cytochrome P-450 CYP2J2 , Dealkylation , Humans , In Vitro Techniques , Intestine, Small/drug effects , Intestine, Small/enzymology , Liver/drug effects , Mass Spectrometry , Microsomes, Liver/drug effects , Oxidation-Reduction , Recombinant Proteins/metabolism
18.
Br J Clin Pharmacol ; 51(2): 133-42, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11259984

ABSTRACT

AIMS: The aims of the present study were to investigate the metabolism of astemizole in human liver microsomes, to assess possible pharmacokinetic drug-interactions with astemizole and to compare its metabolism with terfenadine, a typical H1 receptor antagonist known to be metabolized predominantly by CYP3A4. METHODS: Astemizole or terfenadine were incubated with human liver microsomes or recombinant cytochromes P450 in the absence or presence of chemical inhibitors and antibodies. RESULTS: Troleandomycin, a CYP3A4 inhibitor, markedly reduced the oxidation of terfenadine (26% of controls) in human liver microsomes, but showed only a marginal inhibition on the oxidation of astemizole (81% of controls). Three metabolites of astemizole were detected in a liver microsomal system, i.e. desmethylastemizole (DES-AST), 6-hydroxyastemizole (6OH-AST) and norastemizole (NOR-AST) at the ratio of 7.4 : 2.8 : 1. Experiments with recombinant P450s and antibodies indicate a negligible role for CYP3A4 on the main metabolic route of astemizole, i.e. formation of DES-AST, although CYP3A4 may mediate the relatively minor metabolic routes to 6OH-AST and NOR-AST. Recombinant CYP2D6 catalysed the formation of 6OH-AST and DES-AST. Studies with human liver microsomes, however, suggest a major role for a mono P450 in DES-AST formation. CONCLUSIONS: In contrast to terfenadine, a minor role for CYP3A4 and involvement of multiple P450 isozymes are suggested in the metabolism of astemizole. These differences in P450 isozymes involved in the metabolism of astemizole and terfenadine may associate with distinct pharmacokinetic influences observed with coadministration of drugs metabolized by CYP3A4.


Subject(s)
Anti-Allergic Agents/metabolism , Astemizole/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism , Mixed Function Oxygenases/metabolism , Terfenadine/metabolism , Anti-Bacterial Agents/pharmacology , Antibodies/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/immunology , Drug Interactions , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/immunology , Recombinant Proteins/metabolism , Statistics as Topic , Substrate Specificity , Troleandomycin/pharmacology
19.
Biol Pharm Bull ; 23(1): 112-5, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10706423

ABSTRACT

This study demonstrates that astemizole, a non-sedating anti-histaminergic drug with low toxicity in vivo, greatly potentiates the growth-inhibitory activity of doxorubicin in doxorubicin-resistant human leukemia cells (K562/DXR). Astemizole synergistically potentiated the cytotoxicity of doxorubicin for K562/DXR cells at a concentration of 0.1-3 microM in a dose-dependent manner, whereas they showed hardly any synthergistic effect in the parental cell line (K562) at the same concentration. Since doxorubicin resistance in these cells is associated with the expression of high levels of P-glycoprotein, we evaluated the effect of astemizole on P-glycoprotein activity in cytofluorographic efflux experiments with doxorubicin. Our results indicate that astemizole inhibits the P-glycoprotein pump-efflux activity in a dose-related manner. Moreover, it also inhibits the photolabeling of P-glycoprotein by [3H]azidopine in a dose-dependent manner. These findings provide a biological basis for the potential therapeutic application of astemizole as an anticancer drug either alone or in combination with doxorubicin to multidrug-resistant leukemic cells.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Astemizole/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple , K562 Cells/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Astemizole/administration & dosage , Astemizole/metabolism , Azides/antagonists & inhibitors , Azides/metabolism , Colorimetry , Dihydropyridines/antagonists & inhibitors , Dihydropyridines/metabolism , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Humans , K562 Cells/metabolism , Photoaffinity Labels/metabolism
20.
Clin Ther ; 21(2): 281-95, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10211532

ABSTRACT

Some second-generation antihistamines, notably terfenadine and astemizole, have been associated with prolongation of the QT interval and the development of torsades de pointes, a potentially fatal ventricular arrhythmia. This rare adverse event has been associated with greatly elevated blood levels of these agents, resulting from drug overdose, hepatic insufficiency (dysfunction), or interactions with other drugs that inhibit their metabolism. This paper reviews the data concerning the effects of selected second-generation antihistamines on cardiac conduction, particularly the QT interval, to evaluate whether ventricular arrhythmias are a class effect of these agents. Electrocardiographic studies indicate that terfenadine and astemizole, but not loratadine or cetirizine, prolong the QT interval in laboratory animals. In vitro studies demonstrate that terfenadine and astemizole block the cardiac K+ channels, leading to delayed ventricular repolarization and QT-interval prolongation; in contrast, neither loratadine nor its metabolite, desloratadine, significantly inhibits cardiac K+ channels at clinically achievable blood levels. Studies in human volunteers confirm the absence of electrocardiographic effects of azelastine, cetirizine, fexofenadine, and loratadine administered at several times the recommended dose or concomitantly with agents that inhibit their metabolism and elimination. In conclusion, the data indicate that the potential to cause ventricular arrhythmias is not a class effect of second-generation antihistamines and that loratadine, cetirizine, azelastine, and fexofenadine are not associated with torsades de pointes or other ventricular arrhythmias.


Subject(s)
Heart/drug effects , Histamine H1 Antagonists/adverse effects , Tachycardia, Ventricular/chemically induced , Animals , Astemizole/adverse effects , Astemizole/metabolism , Astemizole/pharmacokinetics , Cytochrome P-450 Enzyme System , Drug Interactions , Electrocardiography , Histamine H1 Antagonists/metabolism , Histamine H1 Antagonists/pharmacokinetics , Humans , Myocardium/metabolism , Potassium Channels , Risk , Terfenadine/adverse effects , Terfenadine/metabolism , Terfenadine/pharmacokinetics , Torsades de Pointes/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...