Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Genes Environ ; 44(1): 10, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35313995

ABSTRACT

BACKGROUND: Aromatic amines, often used as intermediates for pharmaceutical synthesis, may be mutagenic and therefore pose a challenge as metabolites or impurities in drug development. However, predicting the mutagenicity of aromatic amines using commercially available, quantitative structure-activity relationship (QSAR) tools is difficult and often requires expert review. In this study, we developed a shareable QSAR tool based on nitrenium ion stability. RESULTS: The evaluation using in-house aromatic amine intermediates revealed that our model has prediction accuracy of aromatic amine mutagenicity comparable to that of commercial QSAR tools. The effect of changing the number and position of substituents on the mutagenicity of aromatic amines was successfully explained by the change in the nitrenium ion stability. Furthermore, case studies showed that our QSAR tool can support the expert review with quantitative indicators. CONCLUSIONS: This local QSAR tool will be useful as a quantitative support tool to explain the substituent effects on the mutagenicity of primary aromatic amines. By further refinement through method sharing and standardization, our tool can support the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) M7 expert review with quantitative indicators.

2.
Article in English | MEDLINE | ID: mdl-28163191

ABSTRACT

INTRODUCTION: The use of multi-electrode arrays (MEA) in combination with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provides a promising method to predict comprehensive cardiotoxicity, including drug-induced QT prolongation and arrhythmia. We previously demonstrated that MEA in combination with hiPSC-CMs could provide a generalizable platform by using 7 reference drugs at 10 testing facilities. Using this approach, we evaluated responses to reference drugs that modulate a range of cardiac ion currents and have a range of arrhythmogenic effects. METHODS: We used the MEA system (MED64) and commercially available hiPSC-CMs (iCell cardiomyocytes) to evaluate drug effects on the beat rate, field potential duration (FPD), FPD corrected by Fridericia's formula (FPDc), and the incidence of arrhythmia-like waveforms. RESULTS: This assay detected the repolarization effects of Bay K8644, mibefradil, NS1643, levcromakalim, and ouabain; and the chronotropic effects of isoproterenol, ZD7288, and BaCl2. Chronotropy was also affected by K+ and Ca2+ current modulation. This system detected repolarization delays and the arrhythmogenic effects of quinidine, cisapride, thioridazine, astemizole, bepridil, and pimozide more sensitively than the established guinea pig papillary muscle action potential assay. It also predicted clinical QT prolongation by drugs with multiple ion channel effects (fluoxetine, amiodarone, tolterodine, vanoxerine, alfuzosin, and ranolazine). DISCUSSION: MEA in combination with hiPSC-CMs may provide a powerful method to detect various cardiac electrophysiological effects, QT prolongation, and arrhythmia during drug discovery. However, the data require careful interpretation to predict chronotropic effects and arrhythmogenic effects of candidate drugs with multiple ion channel effects.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiotoxins/pharmacology , Heart Rate/drug effects , Induced Pluripotent Stem Cells/drug effects , Ion Channels , Myocytes, Cardiac/drug effects , Arrhythmias, Cardiac/physiopathology , Cardiotonic Agents/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Heart Rate/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Myocytes, Cardiac/physiology
3.
Br J Pharmacol ; 174(1): 15-27, 2017 01.
Article in English | MEDLINE | ID: mdl-27714763

ABSTRACT

BACKGROUND AND PURPOSE: We conducted preclinical and clinical studies to examine the pharmacological, particularly cardiac, effects of amiselimod (MT-1303), a second-generation sphingosine 1-phosphate (S1P) receptor modulator, designed to reduce the bradycardia associated with fingolimod and other S1P receptor modulators. EXPERIMENTAL APPROACH: The selectivity of the active metabolite amiselimod phosphate (amiselimod-P) for human S1P receptors and activation of G-protein-coupled inwardly rectifying K+ (GIRK) channels in human atrial myocytes were assessed. Its cardiac distribution was determined in rats, and cardiovascular telemetry was assessed in monkeys. We also examined the pharmacokinetics, pharmacodynamics and safety of amiselimod in healthy humans. KEY RESULTS: Amiselimod-P showed potent selectivity for S1P1 and high selectivity for S1P5 receptors, with minimal agonist activity for S1P4 and no distinct agonist activity for S1P2 or S1P3 receptors and approximately five-fold weaker GIRK activation than fingolimod-P. After oral administration of amiselimod or fingolimod at 1 mg·kg-1 , the concentration of amiselimod-P in rat heart tissue was lower than that of fingolimod-P, potentially contributing to the minimal cardiac effects of amiselimod. A telemetry study in monkeys confirmed that amiselimod did not affect heart rate or ECG parameters. In healthy human subjects, peripheral blood lymphocyte counts gradually reduced over the 21 day dosing period, with similar lymphocyte count profiles with the highest doses by day 21, and no clinically significant bradycardia observed on day 1 or during the study. CONCLUSIONS AND IMPLICATIONS: Amiselimod exhibited potent therapeutic efficacy with minimal cardiac effects at the anticipated clinical dose and is unlikely to require dose titration.


Subject(s)
Autoimmune Diseases/drug therapy , Bradycardia/drug therapy , Organophosphates/pharmacology , Propanolamines/pharmacology , Receptors, Lysosphingolipid/agonists , Animals , Autoimmune Diseases/metabolism , Bradycardia/metabolism , Cell Line , Dose-Response Relationship, Drug , Humans , Macaca fascicularis , Male , Molecular Structure , Organophosphates/administration & dosage , Organophosphates/chemistry , Propanolamines/administration & dosage , Propanolamines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , Structure-Activity Relationship
4.
J Pharmacol Toxicol Methods ; 78: 93-102, 2016.
Article in English | MEDLINE | ID: mdl-26657830

ABSTRACT

INTRODUCTION: Drug-induced QT prolongation is a major safety issue during drug development because it may lead to lethal ventricular arrhythmias. In this study, we evaluated the utility of multi-electrode arrays (MEA) with human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) to predict drug-induced QT prolongation and arrhythmia. METHODS: Ten facilities evaluated the effects of 7 reference drugs (E-4031, moxifloxacin, flecainide, terfenadine, chromanol 293B, verapamil, and aspirin) using a MED64 MEA system with commercially available hiPS-CMs. Field potential duration (FPD), beat rate, FPD corrected by Fridericia's formula (FPDc), concentration inducing FPDc prolongation by 10% (FPDc10), and incidence of arrhythmia-like waveform were evaluated. RESULTS: The inter-facility variability of absolute values before drug application was similar to the intra-facility variability for FPD, beat rate, and FPDc. The inter-facility variability of FPDc10 for 5 reference drugs ranged from 1.8- to 5.8-fold. At all 10 facilities, E-4031, moxifloxacin, and flecainide prolonged FPDc and induced arrhythmia-like waveforms at concentrations 1.8- to 6.1-fold higher than their FPDc10. Terfenadine prolonged FPDc and induced beating arrest at 8.0 times the FPDc10. The average FPDc10 values for E-4031, moxifloxacin, and terfenadine were comparable to reported plasma concentrations that caused QT prolongation or Torsade de Pointes in humans. Chromanol 293B, a IKs blocker, also prolonged FPDc but did not induce arrhythmia-like waveforms, even at 7.4 times the FPDc10. In contrast, verapamil shortened FPDc and aspirin did not affect FPDc or FP waveforms. DISCUSSION: MEA with hiPS-CMs can be a generalizable method for accurately predicting both QT prolongation and arrhythmogenic liability in humans.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cell Culture Techniques/methods , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells/drug effects , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Arrhythmias, Cardiac/diagnosis , Congresses as Topic , Cryopreservation/methods , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Induced Pluripotent Stem Cells/physiology , Long QT Syndrome/diagnosis , Myocytes, Cardiac/physiology , Pharmaceutical Preparations/administration & dosage , Predictive Value of Tests
5.
J Pharmacol Sci ; 121(1): 1-8, 2013.
Article in English | MEDLINE | ID: mdl-23238536

ABSTRACT

In short QT syndrome, inherited gain-of-function mutations in the human ether a-gogo-related gene (hERG) K(+) channel have been associated with development of fatal arrhythmias. This implies that drugs that activate hERG as a side effect may likewise pose significant arrhythmia risk. hERG activators have been found to have diverse mechanisms of activation, which may reflect their distinct binding sites. Recently, the new hERG activator ICA-105574 was introduced, which disables inactivation of the hERG channel with very high potency. We explored characteristics of this new drug in several experimental models. Patch clamp experiments were used to verify activation of hERG channels by ICA-105574 in human embryonic kidney cells stably-expressing hERG channels. ICA-105574 significantly shortened QT and QTc intervals and monophasic action potential duration (MAP(90)) in Langendorff-perfused guinea-pig hearts. We also administered ICA-105574 to anesthetized dogs while recording ECG and drug plasma concentrations. ICA-105574 (10 mg/kg) significantly shortened QT and QTc intervals, with a free plasma concentration of approximately 1.7 µM at the point of maximal effect. Our data showed that unbound ICA-105574 caused QT shortening in dogs at concentrations comparable to the half maximal effective concentration (EC(50), 0.42 µM) of hERG activation in the patch clamp studies.


Subject(s)
Benzamides/pharmacology , Electrocardiography/drug effects , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Heart/drug effects , Animals , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Guinea Pigs , Humans , In Vitro Techniques , Male , Patch-Clamp Techniques
6.
J Med Chem ; 53(8): 3154-68, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20337461

ABSTRACT

A series of 2-substituted 2-aminopropane-1,3-diols having a biphenyl moiety and their phosphate esters were synthesized to obtain sphingosine 1-phosphate receptor-1 (S1P(1)) receptor agonists with potent immunomodulatory activity accompanied by little or no effect on heart rate. Many of the synthesized compounds sufficiently decreased the number of peripheral blood lymphocytes. Some of the phosphates had potent agonism at S1P(1) but no agonism at S1P(3), which had been reported to be a receptor responsible for heart rate reduction. Although high S1P(1)/S1P(3) selectivity was considered to be favorable to reduce the effect on heart rate, almost all the phosphates showed a remarkable heart rate lowering effect in vivo. The results suggest that other factors in addition to S1P(3) agonism should be responsible for the heart rate reduction caused by S1P(1) agonists. Only 2-amino-2-[2-[2'-fluoro-4'-(4-methylphenylthio)biphenyl-4-yl]ethyl]propane-1,3-diol (6d) was identified as a desired S1P(1) receptor agonist having both the immunomodulatory activity and an attenuated effect on heart rate by a unique screening flow using in vivo evaluating systems primarily.


Subject(s)
Biphenyl Compounds/chemical synthesis , Heart Rate/drug effects , Immunologic Factors/chemical synthesis , Propanolamines/chemical synthesis , Propylene Glycols/chemical synthesis , Receptors, Lysosphingolipid/agonists , Animals , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacology , Cell Line , Cricetinae , Cricetulus , Immunologic Factors/adverse effects , Immunologic Factors/pharmacology , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Propanolamines/adverse effects , Propanolamines/pharmacology , Propylene Glycols/adverse effects , Propylene Glycols/pharmacology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
J Pharmacol Sci ; 109(1): 152-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19129680

ABSTRACT

We recently reported that physiological concentrations of 17beta-estradiol partially down-regulate cardiac rapidly-activating delayed rectifier K(+) currents (hERG currents) independently of estrogen-receptor signaling. To determine if other estrogens have the same effect as that of 17beta-estradiol, we investigated receptor-independent effects of estrone, estrone 3-sulfate, and estriol on hERG currents in patch-clamped estrogen-negative HEK293 cells. Only estrone 3-sulfate partially suppressed hERG currents in a receptor-independent manner by modifying the gating. The concentration-dependence of estrone 3-sulfate revealed that physiological levels of circulating estrone 3-sulfate can modulate hERG currents to the maximal extent in both women and men at any age.


Subject(s)
Estrone/analogs & derivatives , Ether-A-Go-Go Potassium Channels/physiology , Receptors, Estrogen/physiology , Cell Line , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens/chemistry , Estrogens/pharmacology , Estrone/chemistry , Estrone/pharmacology , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Molecular Structure , Patch-Clamp Techniques , Receptors, Estrogen/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...