PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 24417241-6 2014 KEY RESULTS: Pre-exposure of cells to erythromycin resulted in an approximately 14-22-fold rightward shift in the hERG concentration-response curve for thioridazine and terfenadine respectively. Terfenadine 169-180 ETS transcription factor ERG Homo sapiens 114-118 24788058-3 2014 Of these, decursin noncompetitively inhibited CYP2J2-mediated astemizole O-demethylation and terfenadine hydroxylation activities with Ki values of 8.34 and 15.8muM, respectively. Terfenadine 93-104 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 46-52 24048439-0 2014 Terfenadine induces anti-proliferative and apoptotic activities in human hormone-refractory prostate cancer through histamine receptor-independent Mcl-1 cleavage and Bak up-regulation. Terfenadine 0-11 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 147-152 25366487-0 2014 Effect of terfenadine and pentamidine on the HERG channel and its intracellular trafficking: combined analysis with automated voltage clamp and confocal microscopy. Terfenadine 10-21 potassium voltage-gated channel subfamily H member 2 Homo sapiens 45-49 25366487-1 2014 The effects of terfenadine and pentamidine on the human ether-a-go-go related gene (hERG) channel current and its intracellular trafficking were evaluated. Terfenadine 15-26 ETS transcription factor ERG Homo sapiens 84-88 25366487-4 2014 Terfenadine directly blocked the hERG channel current but had no effect on trafficking of hERG channels to the cell membrane after application in culture medium for 2 d. In contrast, pentamidine had no direct effect on the hERG channel current but reduced trafficking of hERG channels. Terfenadine 0-11 ETS transcription factor ERG Homo sapiens 33-37 25366487-5 2014 The two drugs inhibited hERG channel function through different mechanisms: terfenadine through direct channel blockade and pentamidine through inhibition of channel trafficking to the cell membrane. Terfenadine 76-87 ETS transcription factor ERG Homo sapiens 24-28 24048439-0 2014 Terfenadine induces anti-proliferative and apoptotic activities in human hormone-refractory prostate cancer through histamine receptor-independent Mcl-1 cleavage and Bak up-regulation. Terfenadine 0-11 BCL2 antagonist/killer 1 Homo sapiens 166-169 24048439-5 2014 Terfenadine induced the cleavage of Mcl-1 cleavage into a pro-apoptotic 28-kDa fragment and up-regulation of Bak, resulting in the loss of mitochondrial membrane potential (DeltaPsim) and the release of cytochrome c and apoptosis-inducing factor into the cytosol. Terfenadine 0-11 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 36-41 24048439-5 2014 Terfenadine induced the cleavage of Mcl-1 cleavage into a pro-apoptotic 28-kDa fragment and up-regulation of Bak, resulting in the loss of mitochondrial membrane potential (DeltaPsim) and the release of cytochrome c and apoptosis-inducing factor into the cytosol. Terfenadine 0-11 BCL2 antagonist/killer 1 Homo sapiens 109-112 24048439-5 2014 Terfenadine induced the cleavage of Mcl-1 cleavage into a pro-apoptotic 28-kDa fragment and up-regulation of Bak, resulting in the loss of mitochondrial membrane potential (DeltaPsim) and the release of cytochrome c and apoptosis-inducing factor into the cytosol. Terfenadine 0-11 cytochrome c, somatic Homo sapiens 203-215 24048439-8 2014 Terfenadine also induced an indirect-but not direct-DNA damage response through the cleavage and activation of caspase-2, phosphorylation and activation of Chk1 and Chk2 kinases, phosphorylation of RPA32 and acetylation of Histone H3; these processes were highly correlated to severe mitochondrial dysfunction and the activation of caspase cascades. Terfenadine 0-11 caspase 2 Homo sapiens 111-120 24048439-8 2014 Terfenadine also induced an indirect-but not direct-DNA damage response through the cleavage and activation of caspase-2, phosphorylation and activation of Chk1 and Chk2 kinases, phosphorylation of RPA32 and acetylation of Histone H3; these processes were highly correlated to severe mitochondrial dysfunction and the activation of caspase cascades. Terfenadine 0-11 checkpoint kinase 1 Homo sapiens 156-160 24048439-8 2014 Terfenadine also induced an indirect-but not direct-DNA damage response through the cleavage and activation of caspase-2, phosphorylation and activation of Chk1 and Chk2 kinases, phosphorylation of RPA32 and acetylation of Histone H3; these processes were highly correlated to severe mitochondrial dysfunction and the activation of caspase cascades. Terfenadine 0-11 checkpoint kinase 2 Homo sapiens 165-169 24048439-8 2014 Terfenadine also induced an indirect-but not direct-DNA damage response through the cleavage and activation of caspase-2, phosphorylation and activation of Chk1 and Chk2 kinases, phosphorylation of RPA32 and acetylation of Histone H3; these processes were highly correlated to severe mitochondrial dysfunction and the activation of caspase cascades. Terfenadine 0-11 replication protein A2 Homo sapiens 198-203 24048439-10 2014 The exposure of cells to terfenadine caused the up-regulation and activation of Bak and the cleavage of Mcl-1, leading to the loss of DeltaPsim and activation of caspase cascades which further resulted in DNA damage response and cell apoptosis. Terfenadine 25-36 BCL2 antagonist/killer 1 Homo sapiens 80-83 24048439-10 2014 The exposure of cells to terfenadine caused the up-regulation and activation of Bak and the cleavage of Mcl-1, leading to the loss of DeltaPsim and activation of caspase cascades which further resulted in DNA damage response and cell apoptosis. Terfenadine 25-36 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 104-109 24120820-5 2013 Terfenadine also showed a limited solubility (measured concentrations ranging from 0.597 muM to 0.833 muM instead of 1 muM) and a loss of substance through the superfusion tubing from -30.2% to -39.2% with dimethylsulfoxide, ethanol or methanol. Terfenadine 0-11 latexin Homo sapiens 89-92 24021950-8 2013 Compared with recombinant CYP2J2, terfenadine was hydroxylated in cardiomyocytes at a similar K(m) value of 1.5 muM. Terfenadine 34-45 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 26-32 24021950-9 2013 The V(max) of terfenadine hydroxylation in recombinant enzyme was found to be 29.4 pmol/pmol P450 per minute and in the cells 6.0 pmol/pmol P450 per minute. Terfenadine 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 24021950-9 2013 The V(max) of terfenadine hydroxylation in recombinant enzyme was found to be 29.4 pmol/pmol P450 per minute and in the cells 6.0 pmol/pmol P450 per minute. Terfenadine 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-144 24120820-5 2013 Terfenadine also showed a limited solubility (measured concentrations ranging from 0.597 muM to 0.833 muM instead of 1 muM) and a loss of substance through the superfusion tubing from -30.2% to -39.2% with dimethylsulfoxide, ethanol or methanol. Terfenadine 0-11 latexin Homo sapiens 102-105 24120820-5 2013 Terfenadine also showed a limited solubility (measured concentrations ranging from 0.597 muM to 0.833 muM instead of 1 muM) and a loss of substance through the superfusion tubing from -30.2% to -39.2% with dimethylsulfoxide, ethanol or methanol. Terfenadine 0-11 latexin Homo sapiens 102-105 24120820-6 2013 Terfenadine solubility was improved with 2-hydroxypropyl-beta-cyclodextrin, no adsorption was observed, but its capacity to block the hERG channel was decreased. Terfenadine 0-11 ETS transcription factor ERG Homo sapiens 134-138 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 40-44 23518063-9 2013 Three compounds (dofetilide, sertindole, and terfenadine) showed >10-fold discrepancy between hERG potency and inhibitory concentrations in the hESC-CM and IRH assays. Terfenadine 45-56 ETS transcription factor ERG Homo sapiens 97-101 23221912-6 2013 In this study, we show that block of WT HERG by bepridil and terfenadine, two drugs previously shown to be trapped inside the HERG channel after the channel closes, is insensitive to extracellular potassium over the range of 0 mM to 20 mM. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 126-130 22300168-5 2012 KEY RESULTS: Terfenadine concentration- and use-dependently inhibited I(Na) in rabbit myocytes and in human atrial myocytes and also inhibited the hERG. Terfenadine 13-24 ETS transcription factor ERG Homo sapiens 147-151 22498908-5 2012 The treatment of the cells with hERG channel inhibitors such as nifekalant, E-4031, cisapride, terfenadine, and verapamil, recovered the prolonged AP and dose-dependently facilitated cell death upon ES. Terfenadine 95-106 ETS transcription factor ERG Homo sapiens 32-36 24296992-7 2013 Infusion of 1 muM Dofetilide and 10 muM Terfenadine prolonged field potentials 10 fold and 2 fold, respectively. Terfenadine 40-51 latexin Homo sapiens 36-39 23688135-8 2013 Additionally, specific inhibitors of CYP2J2, derived from terfenadine, exhibit strong anti-tumor activity in vitro and in vivo. Terfenadine 58-69 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 37-43 23103595-11 2013 Astemizole, terfenadine and quinidine inhibited hERG currents with IC(50) values of 159nM, 224nM and 2muM, respectively (n=51, 10 and 18). Terfenadine 12-23 ETS transcription factor ERG Homo sapiens 48-52 22418875-11 2012 The histamine H1 receptor antagonist terfenadine prevented scratching and alloknesis evoked by histamine, but not 5-HT, a PAR-4 agonist or an MrgprC11 agonist. Terfenadine 37-48 histamine receptor H1 Mus musculus 4-25 22418875-11 2012 The histamine H1 receptor antagonist terfenadine prevented scratching and alloknesis evoked by histamine, but not 5-HT, a PAR-4 agonist or an MrgprC11 agonist. Terfenadine 37-48 coagulation factor II (thrombin) receptor-like 3 Mus musculus 122-127 22418875-11 2012 The histamine H1 receptor antagonist terfenadine prevented scratching and alloknesis evoked by histamine, but not 5-HT, a PAR-4 agonist or an MrgprC11 agonist. Terfenadine 37-48 MAS-related GPR, member X1 Mus musculus 142-150 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Terfenadine 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Terfenadine 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 20847137-3 2010 In this study, catalytic activities of the two alleles found in East Asians, CYP3A4*16 (T185S) and CYP3A4*18 (L293P), were assessed using the following seven substrates: midazolam, carbamazepine, atorvastatin, paclitaxel, docetaxel, irinotecan, and terfenadine. Terfenadine 249-260 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 21477627-9 2011 Importantly, soluble epoxide hydrolase (sEH) inhibitors are anti-hypertensive and anti-inflammatory and, therefore, protect the heart from damage, whereas the terfenadine-related, specific inhibitors of CYP2J2 exhibit strong anti-tumor activity in vitro and in vivo. Terfenadine 159-170 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 203-209 22328583-6 2012 To identify a specific CYP2J2 inhibitor, 138 drugs were screened using terfenadine and astemizole as probe substrates with recombinant CYP2J2. Terfenadine 71-82 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 23-29 21861192-3 2011 Serum-deprived conditions enhance the cytotoxic effect of terfenadine and caspase-4 and -2 are activated upstream of caspase-9. Terfenadine 58-69 caspase 9 Homo sapiens 117-126 21624711-7 2011 Docking and molecular mechanics simulations for both enantiomers of bupivacaine and terfenadine (a non-stereoselective blocker) were performed inside an open-state model of the hERG channel. Terfenadine 84-95 ETS transcription factor ERG Homo sapiens 177-181 20847137-3 2010 In this study, catalytic activities of the two alleles found in East Asians, CYP3A4*16 (T185S) and CYP3A4*18 (L293P), were assessed using the following seven substrates: midazolam, carbamazepine, atorvastatin, paclitaxel, docetaxel, irinotecan, and terfenadine. Terfenadine 249-260 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-105 20847137-6 2010 On the other hand, K(m) values for CYP3A4.18 were comparable to those for CYP3A4.1 for all substrates except terfenadine; but V(max) values were lower for midazolam, paclitaxel, docetaxel, and irinotecan, resulting in partially reduced intrinsic clearance values (by 34-52%). Terfenadine 109-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 35-41 20847137-6 2010 On the other hand, K(m) values for CYP3A4.18 were comparable to those for CYP3A4.1 for all substrates except terfenadine; but V(max) values were lower for midazolam, paclitaxel, docetaxel, and irinotecan, resulting in partially reduced intrinsic clearance values (by 34-52%). Terfenadine 109-120 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 19923256-1 2010 Several antihistamine drugs including terfenadine, ebastine, and astemizole have been identified as substrates for CYP2J2. Terfenadine 38-49 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 115-121 20153443-7 2010 RESULTS: AP prolongation was observed upon exposure to hERG channel blockers (terfenadine, quinidine, cisapride, sotalol, E-4031 and verapamil), with significantly shorter latencies than in PF assays. Terfenadine 78-89 ETS transcription factor ERG Homo sapiens 55-59 20071423-2 2010 The aim of the present study was to characterize possible interactions between terfenadine, binding to a site located inside the central cavity, and the following substances with various binding sites: dofetilide, fluvoxamine, chlorobutanol, and a hERG-specific toxin isolated from scorpion venom (CnErg1). Terfenadine 79-90 ETS transcription factor ERG Homo sapiens 248-252 20071423-5 2010 Slight subadditive inhibitory effects on hERG peak tail currents were observed when terfenadine and CnErg1 were administered in combination. Terfenadine 84-95 ETS transcription factor ERG Homo sapiens 41-45 20071423-6 2010 Terfenadine and chlorobutanol synergistically inhibit hERG peak tail currents and enhance each other"s inhibitory effect in a concentration-dependent way. Terfenadine 0-11 ETS transcription factor ERG Homo sapiens 54-58 20071423-7 2010 In conclusion, terfenadine interacts with CnErg1 and chlorobutanol, but not with dofetilide or fluvoxamine, at hERG channels. Terfenadine 15-26 ETS transcription factor ERG Homo sapiens 111-115 20071423-8 2010 It is shown that interactions between chlorobutanol and a hERG channel blocker binding inside the central cavity (terfenadine) produce synergistic effects on hERG currents. Terfenadine 114-125 ETS transcription factor ERG Homo sapiens 58-62 20071423-8 2010 It is shown that interactions between chlorobutanol and a hERG channel blocker binding inside the central cavity (terfenadine) produce synergistic effects on hERG currents. Terfenadine 114-125 ETS transcription factor ERG Homo sapiens 158-162 20104563-6 2010 We docked the known hERG inhibitors (+)-cisapride, (S)-terfenadine, and MK-499 into the hERG models and simulation snapshots. Terfenadine 51-66 ETS transcription factor ERG Homo sapiens 20-24 20104563-6 2010 We docked the known hERG inhibitors (+)-cisapride, (S)-terfenadine, and MK-499 into the hERG models and simulation snapshots. Terfenadine 51-66 ETS transcription factor ERG Homo sapiens 88-92 18701618-3 2008 Four high-affinity drugs (cisapride, dofetilide, terfenadine, and astemizole) demonstrated lower affinity for the inactivation-deficient N588K mutant hERG channel compared with N588E and wild-type hERG. Terfenadine 49-60 ETS transcription factor ERG Homo sapiens 150-154 19426697-5 2009 We also evaluated the potency of these analogs on an S6 mutant of hERG (F656A) previously shown to significantly reduce the affinity for MK-499 and other known hERG antagonists (e.g. cisapride, terfenadine). Terfenadine 194-205 ETS transcription factor ERG Homo sapiens 66-70 19775279-1 2010 Following reports of death from cardiac arrhythmias with drugs like terfenadine and cisapride, the International Conference for Harmonization formulated a guidance (E14) document. Terfenadine 68-79 nuclear protein, coactivator of histone transcription Homo sapiens 165-168 19289568-0 2009 Selective inhibitors of CYP2J2 related to terfenadine exhibit strong activity against human cancers in vitro and in vivo. Terfenadine 42-53 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 24-30 19289568-3 2009 In the present study, we tested the hypothesis that specific inhibitors of CYP2J2 related to the drug terfenadine are effective antitumor agents. Terfenadine 102-113 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 75-81 18701618-3 2008 Four high-affinity drugs (cisapride, dofetilide, terfenadine, and astemizole) demonstrated lower affinity for the inactivation-deficient N588K mutant hERG channel compared with N588E and wild-type hERG. Terfenadine 49-60 ETS transcription factor ERG Homo sapiens 197-201 18508907-13 2008 Small (but not large) cholangiocytes proliferate in response to HTMT dimaleate and are blocked by terfenadine (HRH1 antagonist), BAPTA/AM, and W7. Terfenadine 98-109 histamine receptor H1 Mus musculus 111-115 18987434-0 2008 Molecular determinants of hERG channel block by terfenadine and cisapride. Terfenadine 48-59 ETS transcription factor ERG Homo sapiens 26-30 18987434-1 2008 Block of cardiac hERG K+ channels by the antihistamine terfenadine and the prokinetic agent cisapride is associated with prolonged ventricular repolarization and an increased risk of ventricular arrhythmia. Terfenadine 55-66 ETS transcription factor ERG Homo sapiens 17-21 18987434-2 2008 Here, we used a site-directed mutagenesis approach to determine the molecular determinants of hERG block by terfenadine and cisapride. Terfenadine 108-119 ETS transcription factor ERG Homo sapiens 94-98 18560330-0 2008 Identification of terfenadine as an inhibitor of human CD81-receptor HCV-E2 interaction: synthesis and structure optimization. Terfenadine 18-29 CD81 molecule Homo sapiens 55-59 18560330-1 2008 Terfenadine (4-[4-(hydroxydiphenylmethyl)-1-piperidyl]-1-(4-tert-butylphenyl)-butan-1-ol) was identified in a biological screening to be a moderate inhibitor (27% inhibition) of the CD81-LEL-HCV-E2 interaction. Terfenadine 0-11 CD81 molecule Homo sapiens 182-186 18974610-3 2008 Ketoconazole strongly inhibited CYP3A4-mediated terfenadine metabolism in vitro, and the method predicted 6- to 37-fold increase of terfenadine AUC by the concomitant dosing of ketoconazole, which reasonably well agreed with the observed 13- to 59-fold increase of AUC in clinical studies. Terfenadine 48-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 18174239-5 2008 In addition, we have observed that knocking down the H1 histamine receptor (HRH1) by small interference RNA approach protects melanoma cells only slightly from TEF-induced apoptosis. Terfenadine 160-163 histamine receptor H1 Homo sapiens 76-80 18974610-3 2008 Ketoconazole strongly inhibited CYP3A4-mediated terfenadine metabolism in vitro, and the method predicted 6- to 37-fold increase of terfenadine AUC by the concomitant dosing of ketoconazole, which reasonably well agreed with the observed 13- to 59-fold increase of AUC in clinical studies. Terfenadine 132-143 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-38 17767395-1 2007 Fexofenadine, an active metabolite of the second-generation histamine H1 receptor antagonist (antihistamine) terfenadine, does not have the disadvantage of QT prolongation. Terfenadine 109-120 histamine receptor H1 Homo sapiens 60-81 17470359-1 2007 Twenty five derivatives of the drugs terfenadine and ebastine have been designed, synthesized and evaluated as inhibitors of recombinant human CYP2J2. Terfenadine 37-48 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 143-149 17355118-2 2007 G1 PAMAM dendrimer-based prodrugs of the water-insoluble P-gp substrate terfenadine (Ter) were synthesized using succinic acid (suc) or succinyl-diethylene glycol (suc-deg) as a linker/spacer (to yield G1-suc-Ter and G1-suc-deg-Ter, respectively). Terfenadine 72-83 ATP binding cassette subfamily B member 1 Homo sapiens 57-61 17355118-2 2007 G1 PAMAM dendrimer-based prodrugs of the water-insoluble P-gp substrate terfenadine (Ter) were synthesized using succinic acid (suc) or succinyl-diethylene glycol (suc-deg) as a linker/spacer (to yield G1-suc-Ter and G1-suc-deg-Ter, respectively). Terfenadine 85-88 ATP binding cassette subfamily B member 1 Homo sapiens 57-61 17180728-8 2007 The second generation antihistamines terfenadine and loratadine, achieved substantial brain penetration, which was further enhanced by Pgp inhibition by cyclosporin A (CSA). Terfenadine 37-48 PGP Canis lupus familiaris 135-138 15684493-3 2005 This might be because concomitant drugs such as itraconazole inhibit cytochrome P450 3A4 (CYP3A4), the enzyme responsible for the degradation of terfenadine and astemizole, and thus the blood concentrations of the drugs are abnormally increased. Terfenadine 145-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-88 17078945-8 2006 At a concentration of 1 muM, terfenadine significantly increased the spontaneous Na-Ca exchange current (I(NCX)) frequency from 0.48+/-0.25 to 1.93+/-0.67 s(-1). Terfenadine 29-40 T cell leukemia homeobox 2 Homo sapiens 107-110 16569656-6 2006 Treatment of melanoma cells with terfenadine induced DNA damage and caspases 2, 3, 6, 8 and 9 activation. Terfenadine 33-44 caspase 2 Homo sapiens 68-93 16569656-7 2006 Furthermore, the general caspase inhibitor (z-VAD-FMK) and a selective inhibitor of caspase-2 (z-VDVAD-FMK) protected melanoma cells from terfenadine-induced apoptosis. Terfenadine 138-149 caspase 2 Homo sapiens 84-93 16495056-2 2006 This paper reports a first series of high-affinity, selective CYP2J2 inhibitors that are related to terfenadine, with K(i) values as low as 160nM, that should be useful tools to determine the biological roles of CYP2J2. Terfenadine 100-111 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 62-68 16495056-2 2006 This paper reports a first series of high-affinity, selective CYP2J2 inhibitors that are related to terfenadine, with K(i) values as low as 160nM, that should be useful tools to determine the biological roles of CYP2J2. Terfenadine 100-111 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 212-218 15684493-3 2005 This might be because concomitant drugs such as itraconazole inhibit cytochrome P450 3A4 (CYP3A4), the enzyme responsible for the degradation of terfenadine and astemizole, and thus the blood concentrations of the drugs are abnormally increased. Terfenadine 145-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 15762770-11 2005 For example, when aforementioned CYP3A4 inhibitors are coadministered with terfenadine, cisapride or astemizole (all CYP3A4 substrates), torsades de pointes (a life-threatening ventricular arrhythmia associated with QT prolongation) may occur.However, predicting drug-drug interactions involving CYP3A4 inactivation is difficult, since the clinical outcomes depend on a number of factors that are associated with drugs and patients. Terfenadine 75-86 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 33-39 15133245-3 2004 This might be because concomitant drugs such as itraconazole inhibit cytochrome P450 3A4 (CYP3A4), the enzyme responsible for degradation of terfenadine and astemizole, and thus the blood concentrations of the drugs are abnormally increased. Terfenadine 141-152 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-88 15304429-6 2004 This small magnitude in change is in contrast to drugs primarily cleared by cytochrome P450 enzymes, where exposures have been reported to increase as much as 35-fold on coadministration with an inhibitor (e.g., ketoconazole inhibition of CYP3A4-catalyzed terfenadine metabolism). Terfenadine 256-267 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 239-245 15331914-7 2004 Finally, terfenadine had a brain-to-plasma ratio of 2.21 +/- 1.00 even though it underwent Pgp-mediated efflux (in vitro ratio = 2.88). Terfenadine 9-20 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 91-94 15331914-8 2004 Terfenadine"s high passive permeability (285 nm/s) overcame the Pgp-mediated efflux to yield brain-to-plasma ratio >1. Terfenadine 0-11 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 64-67 15272206-5 2004 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. Terfenadine 84-95 potassium voltage-gated channel subfamily H member 2 Homo sapiens 167-171 16050267-7 2005 Our findings suggest that the position of Tyr652 and Phe656 in hERG is optimal for interaction with multiple drugs, Tyr652 is an important determinant of voltage-dependent block, and the hydrophobic surface area of residue 656 and aromaticity of residue 652 are the physicochemical features required for high-affinity block by MK-499, cisapride and terfenadine. Terfenadine 349-360 ETS transcription factor ERG Homo sapiens 63-67 15288570-4 2004 Terfenadine stimulated caspase-8, -9 and -3-like activities in an incubation time-dependent manner in thymocytes. Terfenadine 0-11 caspase 8 Rattus norvegicus 23-43 15288570-5 2004 The active forms of caspase-3 and -9 were detected in the extract from terfenadine-treated cells by immunoblotting analysis using specific antibodies to caspases, but active caspase-8 was not found in this fraction. Terfenadine 71-82 caspase 3 Rattus norvegicus 20-36 15288570-5 2004 The active forms of caspase-3 and -9 were detected in the extract from terfenadine-treated cells by immunoblotting analysis using specific antibodies to caspases, but active caspase-8 was not found in this fraction. Terfenadine 71-82 caspase 8 Rattus norvegicus 153-161 15133245-3 2004 This might be because concomitant drugs such as itraconazole inhibit cytochrome P450 3A4 (CYP3A4), the enzyme responsible for degradation of terfenadine and astemizole, and thus the blood concentrations of the drugs are abnormally increased. Terfenadine 141-152 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 90-96 12925206-0 2003 Terfenadine antagonism against interleukin-4-modulated gene expression of T cell cytokines. Terfenadine 0-11 interleukin 4 Homo sapiens 31-44 14709917-5 2004 Terfenadine 1 microM completely inhibited the human ether a go-go-related gene (HERG) channel current expressed in HEK293 cells in the same experimental solution as in microelectrode experiments. Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 80-84 14570767-8 2003 Mechanism-based inhibition of CYP1A2 may explain why zileuton decreases the oral clearance of antipyrine, propranolol, (R)-warfarin, and theophylline, at doses that have a minimal effect on the pharmacokinetics of (S)-warfarin, phenytoin, and terfenadine. Terfenadine 243-254 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 30-36 12925206-6 2003 Electrophoretic mobility shift assays using [32P]-labeled synthetic oligonucleotides encoding the consensus binding motif of activator protein-1 demonstrated that interleukin-4-induced binding of activator protein-1 composed of JunB was interfered by terfenadine. Terfenadine 251-262 JunB proto-oncogene, AP-1 transcription factor subunit Homo sapiens 125-144 12925206-6 2003 Electrophoretic mobility shift assays using [32P]-labeled synthetic oligonucleotides encoding the consensus binding motif of activator protein-1 demonstrated that interleukin-4-induced binding of activator protein-1 composed of JunB was interfered by terfenadine. Terfenadine 251-262 interleukin 4 Homo sapiens 163-176 12925206-6 2003 Electrophoretic mobility shift assays using [32P]-labeled synthetic oligonucleotides encoding the consensus binding motif of activator protein-1 demonstrated that interleukin-4-induced binding of activator protein-1 composed of JunB was interfered by terfenadine. Terfenadine 251-262 JunB proto-oncogene, AP-1 transcription factor subunit Homo sapiens 196-215 12925206-1 2003 Here, we investigated whether an anti-allergy drug, terfenadine, affects interleukin-4-modulated cytokine expression in peripheral T cells. Terfenadine 52-63 interleukin 4 Homo sapiens 73-86 12925206-6 2003 Electrophoretic mobility shift assays using [32P]-labeled synthetic oligonucleotides encoding the consensus binding motif of activator protein-1 demonstrated that interleukin-4-induced binding of activator protein-1 composed of JunB was interfered by terfenadine. Terfenadine 251-262 JunB proto-oncogene, AP-1 transcription factor subunit Homo sapiens 228-232 12925206-7 2003 This study indicates that terfenadine, at least partially, interferes with interleukin-4-activated signaling, leading to terfenadine antagonism against the modulatory impact of interleukin-4 on T cell cytokines. Terfenadine 26-37 interleukin 4 Homo sapiens 75-88 12925206-4 2003 Interestingly, the interleukin-4-induced expression of all T helper 2 cytokines examined was markedly downregulated by terfenadine. Terfenadine 119-130 interleukin 4 Homo sapiens 19-32 12925206-7 2003 This study indicates that terfenadine, at least partially, interferes with interleukin-4-activated signaling, leading to terfenadine antagonism against the modulatory impact of interleukin-4 on T cell cytokines. Terfenadine 26-37 interleukin 4 Homo sapiens 177-190 12925206-7 2003 This study indicates that terfenadine, at least partially, interferes with interleukin-4-activated signaling, leading to terfenadine antagonism against the modulatory impact of interleukin-4 on T cell cytokines. Terfenadine 121-132 interleukin 4 Homo sapiens 75-88 12860441-0 2003 H1-receptor blocker antihistamine, terfenadine durably influences the glucocorticoid receptor, and lymphocyte histamine content of weanling rats. Terfenadine 35-46 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 70-93 12948015-9 2003 The rank order in P-gp inhibitory potency for terfenadine, verapamil, ritonavir. Terfenadine 46-57 ATP binding cassette subfamily B member 1 Homo sapiens 18-22 15618727-0 2003 Substrate dependent inhibition profiles of fourteen drugs on CYP3A4 activity measured by a high throughput LCMS/MS method with four probe drugs, midazolam, testosterone, nifedipine and terfenadine. Terfenadine 185-196 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-67 12720299-1 2003 Terfenadine (TF), a highly potent histamine H1 receptor antagonist, has been shown to exert no significant central nervous system side effects in clinically effective doses. Terfenadine 0-11 histamine receptor H1 Homo sapiens 34-55 12720299-1 2003 Terfenadine (TF), a highly potent histamine H1 receptor antagonist, has been shown to exert no significant central nervous system side effects in clinically effective doses. Terfenadine 13-15 histamine receptor H1 Homo sapiens 34-55 12388285-5 2003 Astemizole, terfenadine, cisapride, and MK-499 inhibited cERG and human ERG (hERG) currents with IC(50) values of 1.3, 13, 19, and 15 nM and 1.2, 9, 14, and 21 nM, respectively, and competitively displaced [(35)S]MK-499 binding from cERG and hERG with IC(50) values of 0.4, 12, 35, and 0.6 nM and 0.8, 5, 47, and 0.7 nM, respectively. Terfenadine 12-23 potassium voltage-gated channel subfamily H member 2 Canis lupus familiaris 57-61 12388285-5 2003 Astemizole, terfenadine, cisapride, and MK-499 inhibited cERG and human ERG (hERG) currents with IC(50) values of 1.3, 13, 19, and 15 nM and 1.2, 9, 14, and 21 nM, respectively, and competitively displaced [(35)S]MK-499 binding from cERG and hERG with IC(50) values of 0.4, 12, 35, and 0.6 nM and 0.8, 5, 47, and 0.7 nM, respectively. Terfenadine 12-23 ETS transcription factor ERG Homo sapiens 58-61 12388285-5 2003 Astemizole, terfenadine, cisapride, and MK-499 inhibited cERG and human ERG (hERG) currents with IC(50) values of 1.3, 13, 19, and 15 nM and 1.2, 9, 14, and 21 nM, respectively, and competitively displaced [(35)S]MK-499 binding from cERG and hERG with IC(50) values of 0.4, 12, 35, and 0.6 nM and 0.8, 5, 47, and 0.7 nM, respectively. Terfenadine 12-23 ETS transcription factor ERG Homo sapiens 77-81 12388285-5 2003 Astemizole, terfenadine, cisapride, and MK-499 inhibited cERG and human ERG (hERG) currents with IC(50) values of 1.3, 13, 19, and 15 nM and 1.2, 9, 14, and 21 nM, respectively, and competitively displaced [(35)S]MK-499 binding from cERG and hERG with IC(50) values of 0.4, 12, 35, and 0.6 nM and 0.8, 5, 47, and 0.7 nM, respectively. Terfenadine 12-23 potassium voltage-gated channel subfamily H member 2 Canis lupus familiaris 233-237 12388285-5 2003 Astemizole, terfenadine, cisapride, and MK-499 inhibited cERG and human ERG (hERG) currents with IC(50) values of 1.3, 13, 19, and 15 nM and 1.2, 9, 14, and 21 nM, respectively, and competitively displaced [(35)S]MK-499 binding from cERG and hERG with IC(50) values of 0.4, 12, 35, and 0.6 nM and 0.8, 5, 47, and 0.7 nM, respectively. Terfenadine 12-23 ETS transcription factor ERG Homo sapiens 242-246 11878205-6 2002 Azelastine and terfenadine inhibited TNF alpha release with IC50 values of 6.2 mumol/l and 4.3 mumol/l, respectively. Terfenadine 15-26 tumor necrosis factor Homo sapiens 37-46 12365197-0 2002 Effects of terfenadine and diphenhydramine on the CYP2D6 activity in healthy volunteers. Terfenadine 11-22 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 50-56 12365197-1 2002 The aim of this study was to investigate the effects of two antihistaminic drugs, terfenadine and diphenhydramine on CYP2D6 activity by using debrisoquine as a model substrate. Terfenadine 82-93 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 117-123 15618707-2 2002 The effect of addition of rat liver cytosol to an in vitro system using human liver microsomes was examined by measuring the catalytic activities of CYP2C9 (tolbutamide and diclofenac) and CYP3A4 (terfenadine). Terfenadine 197-208 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 189-195 15618707-4 2002 After addition of rat liver cytosol, the unbound Km value (Km,u) for terfenadine metabolism by CYP3A4, and the unbound Ki value of miconazole (Ki,u) for CYP2C9 were smaller than for the controls. Terfenadine 69-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 95-101 15618707-5 2002 Addition of HSA resulted in smaller Km,u values for diclofenac and terfenadine metabolism by CYP2C9 and CYP3A4, respectively, and the Ki,u value for ketoconazole inhibition of CYP3A4 was also reduced. Terfenadine 67-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15618695-7 2002 In contrast, the inhibitory effects of phenacetin, diclofenac, S-mephenytoin, dextromethorphan, bufuralol and terfenadine, typical substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, respectively, on each recombinant CYP activity decreased after preincubation. Terfenadine 110-121 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 146-152 15618707-5 2002 Addition of HSA resulted in smaller Km,u values for diclofenac and terfenadine metabolism by CYP2C9 and CYP3A4, respectively, and the Ki,u value for ketoconazole inhibition of CYP3A4 was also reduced. Terfenadine 67-78 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-110 15618695-7 2002 In contrast, the inhibitory effects of phenacetin, diclofenac, S-mephenytoin, dextromethorphan, bufuralol and terfenadine, typical substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, respectively, on each recombinant CYP activity decreased after preincubation. Terfenadine 110-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 12244568-0 2002 Ketoconazole potentiates terfenadine-induced apoptosis in human Hep G2 cells through inhibition of cytochrome p450 3A4 activity. Terfenadine 25-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 99-118 12244568-1 2002 Terfenadine (TF) is a highly potent histamine H1 receptor antagonist that in clinically effective doses is free of significant central nervous system side effects. Terfenadine 0-11 histamine receptor H1 Homo sapiens 36-57 12244568-1 2002 Terfenadine (TF) is a highly potent histamine H1 receptor antagonist that in clinically effective doses is free of significant central nervous system side effects. Terfenadine 13-15 histamine receptor H1 Homo sapiens 36-57 10730552-1 2000 UNLABELLED: Fexofenadine, the active metabolite of terfenadine, is a selective histamine H1 receptor antagonist that does not cross the blood brain barrier and appears to display some anti-inflammatory properties. Terfenadine 51-62 histamine receptor H1 Homo sapiens 79-100 11240972-1 2001 BACKGROUND AND OBJECTIVE: Nefazodone inhibits CYP3A; therefore coadministration with CYP3A substrates such as terfenadine or loratadine may result in increased exposure to these drugs. Terfenadine 110-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 46-51 11240972-1 2001 BACKGROUND AND OBJECTIVE: Nefazodone inhibits CYP3A; therefore coadministration with CYP3A substrates such as terfenadine or loratadine may result in increased exposure to these drugs. Terfenadine 110-121 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 85-90 11259984-9 2001 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. Terfenadine 80-91 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 202-208 18034580-3 2000 The 2 initial second generation less sedating antihistamines, astemizole and terfenadine, were found to prolong the cardiac QT(c) interval, especially when administered with other medications metabolised by the same cytochrome (CYP) P450 isoenzyme, CYP3A4. Terfenadine 77-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 249-255 11032986-1 2000 We previously reported that the histamine H1 receptor antagonist terfenadine enhances the excitotoxic response to N-methyl-D-aspartate (NMDA) receptor agonists in cerebellar neurons. Terfenadine 65-76 histamine receptor H1 Homo sapiens 32-53 10752642-3 2000 During clinical investigations of drug-drug interactions with therapeutics (terfenadine and cyclosporine) known to be metabolized by CYP3A4, pharmacokinetic interactions were noted upon troglitazone multiple-dose treatments. Terfenadine 76-87 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139 11290874-3 2001 Terfenadine, an antihistamine, has been shown to be a P-gp inhibitor. Terfenadine 0-11 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 54-58 11575008-9 2001 On the other hand terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (330 and 480 nmol/L, respectively), whereas loratadine was about 300-fold less potent. Terfenadine 18-29 potassium voltage-gated channel subfamily H member 2 Homo sapiens 65-69 10828461-3 2000 We demonstrate that HERGS631C in its reduced state is fully blocked by 1 microM astemizole, terfenadine and dofetilide, similar to wild-type HERG channels. Terfenadine 92-103 potassium voltage-gated channel subfamily H member 2 Homo sapiens 20-24 10872584-10 2000 The reproducibility and sensitivity of the assay, combined with the selectivity of mass spectrometric detection, should allow an accurate kinetic characterization of terfenadine oxidation mediated by the high affinity CYP3A enzymes in human liver and intestinal microsomes. Terfenadine 166-177 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 218-223 10772630-5 2000 Known Pgp inhibitors, such as cyclosporin A, nicardipine, verapamil, quinidine, terfenadine, tamoxifen, and vinblastine were demonstrated to inhibit the Pgp-mediated efflux of daunorubicin. Terfenadine 80-91 ATP binding cassette subfamily B member 1 Homo sapiens 6-9 10772630-5 2000 Known Pgp inhibitors, such as cyclosporin A, nicardipine, verapamil, quinidine, terfenadine, tamoxifen, and vinblastine were demonstrated to inhibit the Pgp-mediated efflux of daunorubicin. Terfenadine 80-91 ATP binding cassette subfamily B member 1 Homo sapiens 153-156 10556670-1 1999 Terfenadine, a histamine H(1) receptor antagonist, has been associated with clinical ventricular arrhythmias and in vitro excitation-conduction blocks, whereas anti-ischemic and antiarrhythmic effects have been shown with cicletanine, a prostacyclin generation stimulator. Terfenadine 0-11 histamine H1 receptor Cavia porcellus 15-38 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Terfenadine 12-23 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 129-135 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Terfenadine 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-2 1999 The most pronounced effects were observed with terfenadine, astemizole and loratadine which inhibited CYP3A4-mediated testosterone 6beta-hydroxylation (IC50 of 23, 21 and 32 microM, respectively) and CYP2D6-mediated dextromethorphan O-demethylation (IC50 of 18, 36 and 15 microM, respectively). Terfenadine 47-58 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 102-108 10604956-10 2000 However, both drugs potently blocked HERG current amplitude, with a mean IC(50) of 173 nM for loratadine and 204 nM for terfenadine (pacing rate, 0.1 Hz). Terfenadine 120-131 potassium voltage-gated channel subfamily H member 2 Homo sapiens 37-41 10469039-6 1999 RESULTS: Terfenadine inhibited T-cell proliferation and IL-4 and IL-5 production under each costimulatory condition tested, whereas it had no effect on IL-2 and IFN-gamma production. Terfenadine 9-20 interleukin 4 Homo sapiens 56-60 10469039-6 1999 RESULTS: Terfenadine inhibited T-cell proliferation and IL-4 and IL-5 production under each costimulatory condition tested, whereas it had no effect on IL-2 and IFN-gamma production. Terfenadine 9-20 interleukin 5 Homo sapiens 65-69 10422790-1 1999 Terfenadine and astemizole rarely cause cardiac arrhythmias by suppressing the cardiac rapid delayed rectifier K+ channel encoded by the human ether-a-go-go-related gene (HERG). Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 171-175 10496299-12 1999 Troglitazone may enhance the activities of cytochrome P450 (CYP) 3A and/or transporter(s) thereby reducing the plasma concentrations of terfenadine, cyclosporin, atorvastatin and fexofenadine. Terfenadine 136-147 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-67 10444235-4 1999 In fact, both terfenadine and astemizole have been shown to block HERG K+ channels in a concentration range similar to that found in the plasma of subjects with cardiotoxic manifestations. Terfenadine 14-25 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 10422790-4 1999 Terfenadine and astemizole suppressed the HERG current with IC50 of 431 nM and 69 nM, respectively. Terfenadine 0-11 potassium voltage-gated channel subfamily H member 2 Homo sapiens 42-46 10213372-5 1999 RESULTS: Some (terfenadine, erythromycin and lovastatin) but not all (nifedipine and midazolam) CYP3A substrates were found to be P-gp substrates. Terfenadine 15-26 phosphoglycolate phosphatase Homo sapiens 130-134 10350002-8 1999 The relative affinity of CYP3A for terfenadine metabolism in the two cell lines was comparable, but the maximum reaction rate in the TC7 cells was 8-fold higher. Terfenadine 35-46 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 25-30 10350002-11 1999 Ketoconazole increased the AP-BL transport terfenadine dramatically by inhibiting both terfenadine metabolism and Pgp efflux. Terfenadine 43-54 ATP binding cassette subfamily B member 1 Homo sapiens 114-117 10371095-8 1999 RESULTS: Only children treated with terfenadine achieved significant control of symptoms (P<0.05 in 8 out of 12 months) and allergic inflammation, as shown by inflammatory cell infiltrate and ICAM-1 expression at nasal level (P<0.001), and had significantly fewer extra visits and school absences than the placebo group (P<0.03). Terfenadine 36-47 intercellular adhesion molecule 1 Homo sapiens 195-201 10371095-10 1999 CONCLUSIONS: The present study demonstrates that continuous terfenadine treatment (1 mg/kg body weight per day) could decrease respiratory symptoms and allergic inflammation, and it had an additional antiallergic effect in reducing ICAM-1 expression on nasal epithelial cells. Terfenadine 60-71 intercellular adhesion molecule 1 Homo sapiens 232-238 10371099-4 1999 Moreover, the bronchoconstriction induced by IL-8 was significantly inhibited by the antihistamines diphenhydramine and terfenadine, suggesting the involvement of histamine release in the IL-8-induced bronchoconstriction. Terfenadine 120-131 interleukin-8 Cavia porcellus 45-49 10371099-4 1999 Moreover, the bronchoconstriction induced by IL-8 was significantly inhibited by the antihistamines diphenhydramine and terfenadine, suggesting the involvement of histamine release in the IL-8-induced bronchoconstriction. Terfenadine 120-131 interleukin-8 Cavia porcellus 188-192 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Terfenadine 275-286 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 89-95 10078334-1 1999 Exposure of cultured cerebellar neurons to the histamine H1 receptor antagonist terfenadine resulted in neuronal degeneration and death. Terfenadine 80-91 histamine receptor H1 Homo sapiens 47-68 9733666-0 1998 Interaction of terfenadine and its primary metabolites with cytochrome P450 2D6. Terfenadine 15-26 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 60-79 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-123 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 205-211 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 313-319 9733666-2 1998 Based on this substrate structure-activity relationship, computer modeling studies were undertaken to explore the likely interactions of terfenadine with CYP2D6. Terfenadine 137-148 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 154-160 9733666-3 1998 An overlay of terfenadine and dextromethorphan, a known substrate of CYP2D6, showed that it was possible to superimpose the site of hydroxylation (t-butyl group) and the nitrogen atom of terfenadine with similar regions in dextromethorphan. Terfenadine 14-25 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 69-75 9733666-3 1998 An overlay of terfenadine and dextromethorphan, a known substrate of CYP2D6, showed that it was possible to superimpose the site of hydroxylation (t-butyl group) and the nitrogen atom of terfenadine with similar regions in dextromethorphan. Terfenadine 187-198 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 69-75 9733666-4 1998 These observations were substantiated by the ease of docking of terfenadine into a protein model of CYP2D6. Terfenadine 64-75 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 9733666-5 1998 Experimentally, terfenadine inhibited CYP2D6 activity in human liver microsomes with an IC50 of 14-27 microM, depending on the CYP2D6 substrate used. Terfenadine 16-27 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 38-44 9733666-5 1998 Experimentally, terfenadine inhibited CYP2D6 activity in human liver microsomes with an IC50 of 14-27 microM, depending on the CYP2D6 substrate used. Terfenadine 16-27 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 127-133 9733666-6 1998 The inhibition of CYP2D6 was further defined by determining the Ki for terfenadine against bufuralol 1"-hydroxylase activity in four human livers. Terfenadine 71-82 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 18-24 9733666-11 1998 These data indicate that, as predicted from modeling studies, terfenadine has the structural features necessary for interaction with CYP2D6. Terfenadine 62-73 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 133-139 10597864-1 1999 The use of terfenadine and astemizole, two long-acting nonsedating histamine H1 receptor antagonists, has been associated with prolongation of the QT interval, development of ventricular arrhythmias, particularly torsade de pointes, and sudden cardiac death. Terfenadine 11-22 histamine receptor H1 Homo sapiens 67-88 10597865-4 1999 Acquired LQTS can be induced by a variety of drugs, including some nonsedative histamine H1 receptor antagonists (astemizole, terfenadine). Terfenadine 126-137 histamine receptor H1 Homo sapiens 79-100 10597866-1 1999 Since 1990 it has repeatedly been reported that some histamine H1 receptor antagonists (e.g. terfenadine and astemizole) are able to produce ventricular arrhythmias (e.g. torsade de pointes) when they are given at dosages above the therapeutic range and/or administered together with cytochrome P-450 3A4 inhibitors, such as ketoconazole or erythromycin. Terfenadine 93-104 histamine H1 receptor Cavia porcellus 53-74 9892033-0 1998 Terfenadine and fexofenadine reduce in vitro ICAM-1 expression on human continuous cell lines. Terfenadine 0-11 intercellular adhesion molecule 1 Homo sapiens 45-51 9892033-3 1998 OBJECTIVE: The aim of the study was to evaluate the effect exerted by terfenadine and fexofenadine on adhesion molecules expression (CD54/ICAM-1 and CD29) of a human continuously cultured conjunctival epithelial cell line (WK) and a fibroblast cell line (HEL). Terfenadine 70-81 intercellular adhesion molecule 1 Homo sapiens 133-137 9892033-3 1998 OBJECTIVE: The aim of the study was to evaluate the effect exerted by terfenadine and fexofenadine on adhesion molecules expression (CD54/ICAM-1 and CD29) of a human continuously cultured conjunctival epithelial cell line (WK) and a fibroblast cell line (HEL). Terfenadine 70-81 intercellular adhesion molecule 1 Homo sapiens 138-144 9892033-6 1998 RESULTS: Terfenadine and fexofenadine significantly reduced ICAM-1 basal expression on WK cells at the concentration of 1 microg/mL and 50 microg/mL, respectively. Terfenadine 9-20 intercellular adhesion molecule 1 Homo sapiens 60-66 9892033-7 1998 In addition, both terfenadine and fexofenadine were able to decrease soluble ICAM-1 levels in IFN gamma-stimulated WK cells. Terfenadine 18-29 intercellular adhesion molecule 1 Homo sapiens 77-83 9892033-7 1998 In addition, both terfenadine and fexofenadine were able to decrease soluble ICAM-1 levels in IFN gamma-stimulated WK cells. Terfenadine 18-29 interferon gamma Homo sapiens 94-103 9892033-10 1998 CONCLUSION: This study shows that terfenadine and fexofenadine exert a biologic effect directly on epithelial cells and fibroblasts reducing ICAM-1 expression and partially reducing soluble ICAM-1 release. Terfenadine 34-45 intercellular adhesion molecule 1 Homo sapiens 141-147 9892033-10 1998 CONCLUSION: This study shows that terfenadine and fexofenadine exert a biologic effect directly on epithelial cells and fibroblasts reducing ICAM-1 expression and partially reducing soluble ICAM-1 release. Terfenadine 34-45 intercellular adhesion molecule 1 Homo sapiens 190-196 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Terfenadine 292-303 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 9694927-2 1998 Both compounds use the same cytochrome-P450 metabolic pathway, resulting in an increase in plasma concentration of terfenadine. Terfenadine 115-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 9694927-3 1998 We previously showed that terfenadine blocked HERG (Human Ether-a-Gogo Related Gene), an important component of the repolarizing cardiac delayed rectifier IK with concentration needed to obtain 50% of the block (IC50) in the therapeutic range (300 nM). Terfenadine 26-37 potassium voltage-gated channel subfamily H member 2 Homo sapiens 46-50 9694927-11 1998 We conclude that ketoconazole may potentiate the effects of terfenadine first by an indirect pharmacokinetic action to elevate plasma levels and second by a direct pharmacodynamic action on HERG currents. Terfenadine 60-71 potassium voltage-gated channel subfamily H member 2 Homo sapiens 190-194 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Terfenadine 292-303 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-268 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Terfenadine 275-286 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9618706-8 1998 Some antihistamines (i.e., levocabastine, terfenadine, loratadine, azelastine and oxatomide) can reduce ICAM-1 expression. Terfenadine 42-53 intercellular adhesion molecule 1 Homo sapiens 104-110 9691225-15 1998 The PPD antihistamines, namely terfenadine and astemizole, inhibited cough (MED 30 and 10 mg/kg p.o.) Terfenadine 31-42 mediator of RNA polymerase II transcription subunit 30 Cavia porcellus 76-82 9658196-1 1998 In the current study, the potential blocking ability of K+ channels encoded by the human ether-a-go-go related gene (HERG) by the piperazine H1 receptor antagonist cetirizine has been examined and compared with that of other second-generation antihistamines (astemizole, terfenadine, and loratadine). Terfenadine 271-282 potassium voltage-gated channel subfamily H member 2 Homo sapiens 117-121 9658196-3 1998 On the other hand, terfenadine and astemizole effectively blocked HERG K+ channels with nanomolar affinities (the estimated IC50 values were 330 and 480 nM, respectively), whereas loratadine was approximately 300-fold less potent (IC50 approximately 100 microM). Terfenadine 19-30 potassium voltage-gated channel subfamily H member 2 Homo sapiens 66-70 9714291-5 1998 In fact, interference with HERG K+ channels seems to be the main mechanism explaining both the therapeutic actions of the class III antiarrhythmics and the potential cardiotoxicity of second-generation H1 receptor antagonists such as terfenadine and astemizole, as well as of psychotropic drugs such as some antidepressants and neuroleptics. Terfenadine 234-245 potassium voltage-gated channel subfamily H member 2 Homo sapiens 27-31 9626585-6 1998 The potentiating effect of smoking on ethanol-induced gastric mucosal lesion and MPO activity was abolished by pretreatment with allopurinol, terfenadine or ranitidine. Terfenadine 142-153 myeloperoxidase Rattus norvegicus 81-84 9650812-5 1998 The beta-2 agonist salmeterol, the H1-receptor antagonist terfenadine and the phosphodiesterase inhibitor theophylline inhibited the release of IL-4 and IL-13 by more than 50% following 4 h of basophil stimulation with anti-IgE. Terfenadine 58-69 interleukin 4 Homo sapiens 144-148 9650812-5 1998 The beta-2 agonist salmeterol, the H1-receptor antagonist terfenadine and the phosphodiesterase inhibitor theophylline inhibited the release of IL-4 and IL-13 by more than 50% following 4 h of basophil stimulation with anti-IgE. Terfenadine 58-69 interleukin 13 Homo sapiens 153-158 9660842-7 1998 In a one-sequence crossover study in 12 healthy volunteers, nelfinavir inhibited the elimination of the CYP3A substrate terfenadine and the carboxylate metabolite of terfenadine. Terfenadine 120-131 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 104-109 9649346-3 1998 Ketoconazole, terfenadine, and astemizole were observed to inhibit amprenavir depletion, consistent with their known specificity for CYP3A4. Terfenadine 14-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139 9562230-6 1998 Although terfenadine is a substrate for CYP3A (cytochrome P-450 3A), while sertindole is a substrate for both CYP2D6 and CYP3A4, the results in this study suggest that sertindole, at a clinical dose, is not an inhibitor of the metabolism of terfenadine. Terfenadine 9-20 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-45 10022752-0 1998 Human cytochrome P450 3A (CYP3A) mediated midazolam metabolism: the effect of assay conditions and regioselective stimulation by alpha-naphthoflavone, terfenadine and testosterone. Terfenadine 151-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-24 10022752-0 1998 Human cytochrome P450 3A (CYP3A) mediated midazolam metabolism: the effect of assay conditions and regioselective stimulation by alpha-naphthoflavone, terfenadine and testosterone. Terfenadine 151-162 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 26-31 10022752-1 1998 The effect of ionic strength, assay constituents, alpha-naphthoflavone (aNF), terfenadine and testosterone on human CYP3A mediated midazolam (MDZ) 1"-hydroxylation (MDZ 1"-OH) and 4-hydroxylation (MDZ 4-OH) in vitro was examined. Terfenadine 78-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-121 10022752-9 1998 Surprisingly, with expressed CYP3A4, terfenadine (20 microM) inhibited MDZ 1"-OH formation. Terfenadine 37-48 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 29-35 9342502-18 1997 The potential for a clinically significant interaction between nefazodone and other drugs cleared by CYP3A4 (e.g. terfenadine) should be considered before the coadministration of these compounds. Terfenadine 114-125 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 101-107 9506246-1 1998 The nonsedating histamine H1 receptor antagonist fexofenadine is the active metabolite of terfenadine. Terfenadine 90-101 histamine receptor H1 Homo sapiens 16-37 9408807-1 1997 Potent CYP3A4 inhibitors such as ketoconazole can cause dangerous increases in plasma concentrations of the H-1 antagonist terfenadine. Terfenadine 123-134 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 7-13 9541729-8 1998 In contrast, at anti-histamine, doses, the histamine H1-receptor antagonists terfenadine (1 to 30 mg/kg p.o. Terfenadine 77-88 histamine receptor H 1 Rattus norvegicus 43-64 9485522-8 1997 CYP3A4 (testosterone 6 beta-hydroxylation) activity was strongly inhibited by terfenadine with a Ki value of 25 microM, whereas epinastine had no effect at up to 100 microM. Terfenadine 78-89 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-6 9270401-2 1997 TDP has been associated with terfenadine use in cases of liver disease, electrolyte abnormalities, concomitant administration of drugs that inhibit cytochrome P-450, or deliberate overdose. Terfenadine 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 9291293-0 1997 Effect of terfenadine on TNF alpha release from peripheral blood mononuclear cells during cow"s milk allergy. Terfenadine 10-21 tumor necrosis factor Bos taurus 25-34 9349334-9 1997 The mean +/- SEM percentage reduction in flare area for 200 pmol/site of substance P in non-atopics and atopics was 53 +/- 10% and 73 +/- 4% with terfenadine, and 74 +/- 7% and 75 +/- 4% with topical doxepin, respectively. Terfenadine 146-157 tachykinin precursor 1 Homo sapiens 73-84 9349334-11 1997 The inhibitory effects of doxepin were as great as those of terfenadine, and doxepin had a significantly greater effect than terfenadine in inhibiting the weal response to histamine and flare response to substance P in normal volunteers (P < 0.05). Terfenadine 125-136 tachykinin precursor 1 Homo sapiens 204-215 9291293-3 1997 OBJECTIVES: To test the potential ability of terfenadine to inhibit TNF alpha secretion by mononuclear cells from infants with cow"s milk allergy. Terfenadine 45-56 tumor necrosis factor Homo sapiens 68-77 9291293-7 1997 There was a dose-dependent decrease in TNF alpha secretion in the presence of terfenadine, with a maximal inhibition of 62% of this secretion at 1 microM. Terfenadine 78-89 tumor necrosis factor Bos taurus 39-48 9291293-9 1997 CONCLUSION: These results indicate that in infants with intestinal dysfunction due to cow"s milk allergy, terfenadine is a potent inhibitor of the TNF alpha secretion induced by sensitizing milk protein antigens. Terfenadine 106-117 tumor necrosis factor Bos taurus 147-156 9196279-0 1997 Comparative effects of nonsedating histamine H1 receptor antagonists, ebastine and terfenadine, on human Kv1.5 channels. Terfenadine 83-94 potassium voltage-gated channel subfamily A member 5 Homo sapiens 105-110 9279883-1 1997 PURPOSE: To compare the activity of the CYP3A enzyme expressed by TC7, a cell culture model of the intestinal epithelial cell, to the activity of human intestinal CYP3A4, using terfenadine as a substrate. Terfenadine 177-188 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 40-45 9279883-1 1997 PURPOSE: To compare the activity of the CYP3A enzyme expressed by TC7, a cell culture model of the intestinal epithelial cell, to the activity of human intestinal CYP3A4, using terfenadine as a substrate. Terfenadine 177-188 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 163-169 9279883-3 1997 The effect of two CYP3A inhibitors, ketoconazole and troleandomycin (TAO), on the metabolism of terfenadine was also examined. Terfenadine 96-107 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 18-23 9196279-2 1997 Upon depolarization to +60 mV, terfenadine, 1 microM and 3 microM, inhibited the hKv1.5 current by 42.4 +/- 6.4% and 69.3 +/- 4.2% (P < 0.01). Terfenadine 31-42 potassium voltage-gated channel subfamily A member 5 Homo sapiens 81-87 8886862-4 1996 Pre-treatment of the subjects with the H1 histamine receptor antgonist cetirizine (10 mg, orally) or terfenadine (60 mg, orally) 3 h before bradykinin administration caused significant reduction of the bradykinin-induced increase in nasal airway resistance in the upper range of bradykinin doses (300-1000 micrograms) but not in the lower range (10-100 micrograms). Terfenadine 101-112 kininogen 1 Homo sapiens 140-150 9129558-2 1997 Itraconazole strongly interacts with some of the substrates of CYP3A4 (e.g., terfenadine, triazolam and lovastatin); hence it is important to uncover the possible interaction of itraconazole with felodipine. Terfenadine 77-88 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 63-69 9232548-5 1997 Terfenadine prolonged the QT interval in an infusion-rate-dependent manner, its EC50 value was 792-1039 ng mL-1. Terfenadine 0-11 L1 cell adhesion molecule Mus musculus 107-111 9232548-6 1997 An obvious QT prolongation was, moreover, observed even at a plasma terfenadine concentration of 100-200 ng mL-1, which is clinically quite high, but might be achieved under a definite condition such as a restrained terfenadine metabolism. Terfenadine 68-79 L1 cell adhesion molecule Mus musculus 108-112 9232548-6 1997 An obvious QT prolongation was, moreover, observed even at a plasma terfenadine concentration of 100-200 ng mL-1, which is clinically quite high, but might be achieved under a definite condition such as a restrained terfenadine metabolism. Terfenadine 216-227 L1 cell adhesion molecule Mus musculus 108-112 9165365-6 1997 HSR-609 was found to display selective displacement of the [3H]mepyramine binding to H1-receptors for lung vs cerebral cortex as found with terfenadine in guinea pigs ex vivo. Terfenadine 140-151 component of Sp100-rs Mus musculus 0-3 9186817-3 1997 In 6 of the 7 women the effect of terfenadine (TRF, 120 mg os at -60 min), another H1-receptor antagonist, on the GH response to GHRH or HEX was also studied. Terfenadine 34-45 growth hormone releasing hormone Homo sapiens 129-133 9186817-3 1997 In 6 of the 7 women the effect of terfenadine (TRF, 120 mg os at -60 min), another H1-receptor antagonist, on the GH response to GHRH or HEX was also studied. Terfenadine 47-50 growth hormone releasing hormone Homo sapiens 129-133 9013373-1 1996 Terfenadine is metabolized by the cytochrome P-450 3A subfamily of enzymes (CYP3A). Terfenadine 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-81 9013373-3 1996 The authors compared the abilities of dirithromycin (a new macrolide antibiotic agent), its major metabolite erythromycylamine, and the known CYP3A substrate terfenadine to inhibit CYP3A in vitro. Terfenadine 158-169 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 142-147 9013373-3 1996 The authors compared the abilities of dirithromycin (a new macrolide antibiotic agent), its major metabolite erythromycylamine, and the known CYP3A substrate terfenadine to inhibit CYP3A in vitro. Terfenadine 158-169 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 181-186 8886862-4 1996 Pre-treatment of the subjects with the H1 histamine receptor antgonist cetirizine (10 mg, orally) or terfenadine (60 mg, orally) 3 h before bradykinin administration caused significant reduction of the bradykinin-induced increase in nasal airway resistance in the upper range of bradykinin doses (300-1000 micrograms) but not in the lower range (10-100 micrograms). Terfenadine 101-112 kininogen 1 Homo sapiens 202-212 8886862-4 1996 Pre-treatment of the subjects with the H1 histamine receptor antgonist cetirizine (10 mg, orally) or terfenadine (60 mg, orally) 3 h before bradykinin administration caused significant reduction of the bradykinin-induced increase in nasal airway resistance in the upper range of bradykinin doses (300-1000 micrograms) but not in the lower range (10-100 micrograms). Terfenadine 101-112 kininogen 1 Homo sapiens 202-212 8887714-1 1996 A sensitive LC-MS-MS method capable of quantifying terfenadine at levels down to 100 pg ml-1 in human plasma is reported. Terfenadine 51-62 interleukin 17F Homo sapiens 88-92 8610817-8 1996 Cytochrome P450 3A4 metabolizes terfenadine, astemizole, carbamazepine, alprazolam, triazolam, and other benzodiazepines. Terfenadine 32-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 0-19 8772706-5 1996 We found that HERG was 10 times more sensitive than Kv1.5 to terfenadine block. Terfenadine 61-72 potassium voltage-gated channel subfamily H member 2 Homo sapiens 14-18 8772706-5 1996 We found that HERG was 10 times more sensitive than Kv1.5 to terfenadine block. Terfenadine 61-72 potassium voltage-gated channel subfamily A member 5 Homo sapiens 52-57 8772706-8 1996 The Kd value for HERG block is relevant to the toxicity of the antihistamine, since the clinical terfenadine concentrations in human plasma may reach the 100 nmol/L range. Terfenadine 97-108 potassium voltage-gated channel subfamily H member 2 Homo sapiens 17-21 8772706-10 1996 We propose that the blocking of HERG by terfenadine explains the acquired long QT syndrome. Terfenadine 40-51 potassium voltage-gated channel subfamily H member 2 Homo sapiens 32-36 8735853-6 1996 Oral terfenadine and cetirizine and intranasal levocabastine and azelastine have also demonstrated a lowering of ICAM-1 expression on epithelial cells. Terfenadine 5-16 intercellular adhesion molecule 1 Homo sapiens 113-119 8641472-0 1996 Blockade of HERG channels expressed in Xenopus oocytes by the histamine receptor antagonists terfenadine and astemizole. Terfenadine 93-104 potassium voltage-gated channel subfamily H member 2 Homo sapiens 12-16 8641472-2 1996 Here, terfenadine and astemizole both inhibited the human ether-a-go-go related gene (HERG) encoded channels expressed in Xenopus oocytes at nanomolar concentrations in a use- and voltage-dependent fashion. Terfenadine 6-17 potassium voltage-gated channel subfamily H member 2 Homo sapiens 86-90 8641472-4 1996 These results suggest that blockade of HERG channels by terfenadine and astemizole might contribute to the cardiac side effects of these compounds. Terfenadine 56-67 potassium voltage-gated channel subfamily H member 2 Homo sapiens 39-43 8880073-1 1996 The effects of chronic treatment with the non-sedative histamine H1 receptor antagonist terfenadine (5 mg/kg, i.p.) Terfenadine 88-99 histamine receptor H 1 Rattus norvegicus 55-76 8880073-7 1996 These results suggest that the histamine H1 receptor antagonist terfenadine may have antidepressant properties and that terfenadine is effective in reversing some of behavioural and immune changes in the olfactory bulbectomized rat model of depression. Terfenadine 64-75 histamine receptor H 1 Rattus norvegicus 31-52 8613951-5 1996 Ritonavir was found to be a potent inhibitor of CYP3A-mediated biotransformations (nifedipine oxidation, IC50) = 0.07 microM; 17alpha-ethynylestradiol 2-hydroxylation, IC50 = 2 microM; terfenadine hydroxylation, IC50 = 0.14 microM). Terfenadine 185-196 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 48-53 8689766-2 1996 The aim of this study was to compare the effects of terfenadine (an H1-antagonist) and cimetidine (an H2-antagonist) on weal and flare responses to PAF and BK in healthy non-atopic human volunteers. Terfenadine 52-63 PCNA clamp associated factor Homo sapiens 148-151 8689766-2 1996 The aim of this study was to compare the effects of terfenadine (an H1-antagonist) and cimetidine (an H2-antagonist) on weal and flare responses to PAF and BK in healthy non-atopic human volunteers. Terfenadine 52-63 kininogen 1 Homo sapiens 156-158 8689766-19 1996 The aim of this study was to compare the effects of terfenadine (an H1-antagonist) and cimetidine (an H2-antagonist) on PAF- and BK-induced weal and flare responses in healthy, non-atopic human volunteers. Terfenadine 52-63 PCNA clamp associated factor Homo sapiens 120-123 8689766-4 1996 Terfenadine significantly reduced weal and flare responses to PAF (mean reduction 53 and 73%, respectively) and flare responses to BK (mean reduction 78%) but had no effect on weal responses to BK. Terfenadine 0-11 PCNA clamp associated factor Homo sapiens 62-65 8689766-19 1996 The aim of this study was to compare the effects of terfenadine (an H1-antagonist) and cimetidine (an H2-antagonist) on PAF- and BK-induced weal and flare responses in healthy, non-atopic human volunteers. Terfenadine 52-63 kininogen 1 Homo sapiens 129-131 8689766-4 1996 Terfenadine significantly reduced weal and flare responses to PAF (mean reduction 53 and 73%, respectively) and flare responses to BK (mean reduction 78%) but had no effect on weal responses to BK. Terfenadine 0-11 kininogen 1 Homo sapiens 131-133 8788208-6 1995 In rat liver, only the N-dealkylation pathway appears to be mediated by CYP3A since anti-rat CYP3A antibody inhibited azacyclonol but not alcohol metabolite formation in incubations of terfenadine with liver microsomes. Terfenadine 185-196 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 72-77 8626361-10 1996 On the basis of the inhibition of cytochrome P450 3A4 isoenzyme by nefazodone in vitro, coadministration of terfenadine or astemizole with nefazodone is contraindicated because nefazodone can increase the plasma levels of these two drugs. Terfenadine 108-119 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 34-53 8748566-11 1995 Nefazodone, an inhibitor of the hepatic P-450 isoenzyme CYP3A4, may increase concentrations of drugs metabolized by this isoenzyme, such as terfenadine, astemizole, triazolam, alprazolam, and midazolam. Terfenadine 140-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 56-62 8788208-6 1995 In rat liver, only the N-dealkylation pathway appears to be mediated by CYP3A since anti-rat CYP3A antibody inhibited azacyclonol but not alcohol metabolite formation in incubations of terfenadine with liver microsomes. Terfenadine 185-196 cytochrome P450, family 3, subfamily a, polypeptide 62 Rattus norvegicus 93-98 8584574-1 1995 Chronic administration of a low dose of the histamine H1 receptor antagonist terfenadine (2 mg/kg/day) induced an increase in gamma-glutamyltransferase serum activity in rats, 360 h after partial hepatectomy while the enzymatic activity appeared not to be affected during liver regeneration following treatment with the histamine H2 receptor antagonist cimetidine (20 mg/kg/day). Terfenadine 77-88 histamine receptor H 1 Rattus norvegicus 44-65 8564726-0 1995 Terfenadine exerts antiallergic activity reducing ICAM-1 expression on nasal epithelial cells in patients with pollen allergy. Terfenadine 0-11 intercellular adhesion molecule 1 Homo sapiens 50-56 8564726-7 1995 RESULTS: As opposed to the placebo group, patients treated with Terfenadine showed a significant improvement of both symptoms (P < 0.022) and signs P < 0.001), a significant reduction of inflammatory cells infiltrate (P < 0.005), of ECP levels (P < 0.002) and ICAM-1 expression on nasal epithelial cells (P < 0.005). Terfenadine 64-75 ribonuclease A family member 3 Homo sapiens 242-245 8564726-7 1995 RESULTS: As opposed to the placebo group, patients treated with Terfenadine showed a significant improvement of both symptoms (P < 0.022) and signs P < 0.001), a significant reduction of inflammatory cells infiltrate (P < 0.005), of ECP levels (P < 0.002) and ICAM-1 expression on nasal epithelial cells (P < 0.005). Terfenadine 64-75 intercellular adhesion molecule 1 Homo sapiens 272-278 8564726-8 1995 CONCLUSIONS: In conclusion, these data demonstrate that Terfenadine exerts antiallergic activity since it is able to reduce inflammatory cell infiltrate and downregulates ICAM-1 expression. Terfenadine 56-67 intercellular adhesion molecule 1 Homo sapiens 171-177 8584574-1 1995 Chronic administration of a low dose of the histamine H1 receptor antagonist terfenadine (2 mg/kg/day) induced an increase in gamma-glutamyltransferase serum activity in rats, 360 h after partial hepatectomy while the enzymatic activity appeared not to be affected during liver regeneration following treatment with the histamine H2 receptor antagonist cimetidine (20 mg/kg/day). Terfenadine 77-88 gamma-glutamyltransferase 1 Rattus norvegicus 126-151 8584574-1 1995 Chronic administration of a low dose of the histamine H1 receptor antagonist terfenadine (2 mg/kg/day) induced an increase in gamma-glutamyltransferase serum activity in rats, 360 h after partial hepatectomy while the enzymatic activity appeared not to be affected during liver regeneration following treatment with the histamine H2 receptor antagonist cimetidine (20 mg/kg/day). Terfenadine 77-88 histamine receptor H 2 Rattus norvegicus 320-341 7654486-9 1995 Mean (95% CI) measurements for terfenadine vs placebo treatment periods were 1.5 vs 1.5 (-0.3, 0.3) l for FEV1, 259 vs 260 (-42, 40) l min-1 for morning PEF and 0.8 vs 0.8 (-0.3, 0.3) for global symptom scores. Terfenadine 31-42 CD59 molecule (CD59 blood group) Homo sapiens 135-140 7587966-0 1995 In vitro metabolism of terfenadine by a purified recombinant fusion protein containing cytochrome P4503A4 and NADPH-P450 reductase. Terfenadine 23-34 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 87-105 7587966-2 1995 The metabolism of terfenadine was studied with a cDNA-expressed/purified recombinant fusion protein containing human liver microsomal cytochrome P4503A4 (CYP3A4) linked to rat NADPH-P450 reductase (rF450[mHum3A4/mRatOR]L1) and was compared with that observed in the presence of human liver microsomes and precision-cut human liver tissue slices. Terfenadine 18-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 134-152 7587966-2 1995 The metabolism of terfenadine was studied with a cDNA-expressed/purified recombinant fusion protein containing human liver microsomal cytochrome P4503A4 (CYP3A4) linked to rat NADPH-P450 reductase (rF450[mHum3A4/mRatOR]L1) and was compared with that observed in the presence of human liver microsomes and precision-cut human liver tissue slices. Terfenadine 18-29 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 154-160 7587966-7 1995 Ketoconazole, a well-documented CYP3A inhibitor, effectively inhibited terfenadine metabolism in all three models. Terfenadine 71-82 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 32-37 7587966-9 1995 It is concluded that cDNA-expressed CYP3A4, in the form of a NADPH-P450 reductase-linked fusion protein, may also serve as a model for studying the metabolism of terfenadine in vitro and many other drugs. Terfenadine 162-173 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 36-42 8001268-0 1995 Cardiac electrophysiological actions of the histamine H1-receptor antagonists astemizole and terfenadine compared with chlorpheniramine and pyrilamine. Terfenadine 93-104 histamine H1 receptor Cavia porcellus 44-65 7670728-8 1995 Upon depolarization, racemic terfenadine and its enantiomers induced a fast decline of hKv1.5 current towards a reduced steady state current level. Terfenadine 29-40 potassium voltage-gated channel subfamily A member 5 Homo sapiens 87-93 7670728-14 1995 The EC50 for hKv1.5 block by racemic terfenadine was 0.88 microM, while the values for R- and S-terfenadine were 1.19 microM and 1.16 microM, respectively. Terfenadine 37-48 potassium voltage-gated channel subfamily A member 5 Homo sapiens 13-19 7697852-6 1995 Both astemizole and terfenadine suppressed the IK1 channel by 17% to 50% in a voltage-dependent manner in rat and guinea pig myocytes. Terfenadine 20-31 potassium calcium-activated channel subfamily N member 4 Rattus norvegicus 47-50 7587944-0 1995 Metabolism of terfenadine associated with CYP3A(4) activity in human hepatic microsomes. Terfenadine 14-25 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-50 7587944-3 1995 Although testosterone 6 beta-hydroxylation [CYP3A(4)] has been shown to be the principal enzyme involved in the first step in terfenadine"s biotransformation (formation of azacyclonol and terfenadine alcohol), the enzymes catalyzing the subsequent metabolic steps in the conversion of terfenadine alcohol to azacyclonol and terfenadine acid have not been identified. Terfenadine 126-137 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-52 7587944-7 1995 The formation of azacyclonol and terfenadine alcohol from terfenadine is confirmed to be catalyzed predominantly by CYP3A(4) isozyme, and the ratio of the rate of terfenadine alcohol formation to that of azacyclonol is 3:1. Terfenadine 33-44 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-124 7587944-8 1995 Involvement of the CYP3A(4) in terfenadine metabolism was further confirmed by the following studies: a) inhibition of terfenadine alcohol formation by ketoconazole and troleandomycin, two specific inhibitors of CYP3A(4), and b) time course of terfenadine alcohol formation by cloned human CYP3A(4). Terfenadine 31-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 19-27 7587944-8 1995 Involvement of the CYP3A(4) in terfenadine metabolism was further confirmed by the following studies: a) inhibition of terfenadine alcohol formation by ketoconazole and troleandomycin, two specific inhibitors of CYP3A(4), and b) time course of terfenadine alcohol formation by cloned human CYP3A(4). Terfenadine 31-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 212-220 7587944-8 1995 Involvement of the CYP3A(4) in terfenadine metabolism was further confirmed by the following studies: a) inhibition of terfenadine alcohol formation by ketoconazole and troleandomycin, two specific inhibitors of CYP3A(4), and b) time course of terfenadine alcohol formation by cloned human CYP3A(4). Terfenadine 31-42 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 212-220 7573821-4 1995 Terfenadine caused significant inhibition of both PGD2 and LTC4/D4 (49 +/- 9 and 29 +/- 19%, respectively) from human lung cells but had a less marked effect on PGD2 release from human colon cells (21 +/- 9% for PGD2 and 18 +/- 9% for LTC4/D4). Terfenadine 0-11 prostaglandin D2 synthase Homo sapiens 212-223 8067599-3 1994 Since eosinophil degranulation is also associated with tissue inflammation, we further examined the effect of terfenadine on the PAF-induced release of eosinophil cationic protein. Terfenadine 110-121 ribonuclease A family member 3 Homo sapiens 152-179 7995001-2 1994 Ketoconazole and itraconazole may seriously interact with some of the substrates of CYP3A4 (e.g., terfenadine); hence their possible interaction with triazolam in humans is important to uncover. Terfenadine 98-109 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 84-90 7895601-7 1994 Other CYP3A substrates (cyclosporin A, naringenin, and midazolam) also demonstrated potent inhibition of terfenadine biotransformation in human liver microsomes (IC50 = 17-24 microM). Terfenadine 105-116 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 6-11 7895601-8 1994 Substrates of other P450 families [sparteine (CYP2D6), caffeine (CYP1A), and diclofenac (CYP2C)] only very weakly inhibited terfenadine metabolism. Terfenadine 124-135 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 8067599-8 1994 Terfenadine, at concentrations of 500 and 1000 ng/mL, significantly inhibited PAF-induced and FMLP-induced eosinophil chemotaxis, whereas 1000 ng/mL of terfenadine was necessary to suppress neutrophil chemotaxis. Terfenadine 0-11 formyl peptide receptor 1 Homo sapiens 94-98 8067599-8 1994 Terfenadine, at concentrations of 500 and 1000 ng/mL, significantly inhibited PAF-induced and FMLP-induced eosinophil chemotaxis, whereas 1000 ng/mL of terfenadine was necessary to suppress neutrophil chemotaxis. Terfenadine 152-163 formyl peptide receptor 1 Homo sapiens 94-98 8119047-4 1993 Macrolides inhibit cytochrome P450IIIA4 (CYP3A4), which appears to be the most common metabolic enzyme in the human liver and is involved in the metabolism of many drugs, including cyclosporin, warfarin and terfenadine. Terfenadine 207-218 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 41-47 7515418-2 1994 During terfenadine treatment, the mean substance P concentrations in nasal secretions from patients allergic to house dust or pollen were significantly decreased to 62 and 39% of the initial values, respectively. Terfenadine 7-18 tachykinin precursor 1 Homo sapiens 39-50 8112815-1 1993 Terfenadine is a selective histamine H1 receptor antagonist which binds preferentially to peripheral receptors in vivo and is devoid of central nervous system depressant activity and thus has an improved adverse effect profile (1). Terfenadine 0-11 histamine receptor H1 Homo sapiens 27-48 8257954-1 1993 Terfenadine was the first non-sedating histamine H1 receptor antagonist and one of the most frequently prescribed H1 antihistamines. Terfenadine 0-11 histamine receptor H1 Homo sapiens 39-60 8257958-4 1993 The increased risk of both H1-antihistamines was associated with exposure to supratherapeutic doses; for terfenadine the risk was also associated with concomitant exposure to the cytochrome P-450 inhibitors ketoconazole, erythromycin and cimetidine. Terfenadine 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 8112439-0 1993 Effect of oral terfenadine on bronchoconstrictor response to inhaled neurokinin A and histamine in asthmatic subjects. Terfenadine 15-26 tachykinin precursor 1 Homo sapiens 69-81 7693504-2 1993 Our aim was to determine the relationship between the contributions of vagal stimulation (determined by the use of the muscarinic receptor antagonist, ipratropium bromide) and histamine release (determined by the use of the histamine H1-receptor antagonist terfenadine) in EIA. Terfenadine 257-268 histamine receptor H1 Homo sapiens 224-245 8388006-0 1993 The effect of oral terfenadine on neurokinin-A-induced bronchoconstriction. Terfenadine 19-30 tachykinin precursor 1 Homo sapiens 34-46 1587633-0 1992 Effect of terfenadine on neutrophil and eosinophil chemotactic activities after inhalation of platelet-activating factor in vivo and on neutrophil chemotaxis in vitro. Terfenadine 10-21 PCNA clamp associated factor Homo sapiens 94-120 8466096-3 1993 The results of the study showed that both cetirizine, 10 mg, QD and terfenadine, 60 mg, BID used for 1 week are safe and effective in the management of allergic rhinitis. Terfenadine 68-79 BH3 interacting domain death agonist Homo sapiens 88-91 7681268-11 1993 An inhibition of eicosanoids was observed at both 1 and 10 mumol terfenadine (median percentage of inhibition of PGD2: 38.00 +/- 15.65 and 56.00 +/- 13.12; median percentage of inhibition of LTC4/D4: 37.5 +/- 19.80 and 52.5 +/- 26.8). Terfenadine 65-76 prostaglandin D2 synthase Homo sapiens 113-117 8094615-1 1993 In our efforts to identify clinically effective drugs for reversing multidrug resistance (MDR) mediated by P-glycoprotein, we tested terfenadine for anti-MDR activity because it appeared to sensitize a patient to doxorubicin and because it met structural requirements defined for this activity. Terfenadine 133-144 ATP binding cassette subfamily B member 1 Homo sapiens 107-121 8094615-6 1993 The mechanism of action of terfenadine appeared to be due to inhibition of the function of P-glycoprotein since it augmented the accumulation of doxorubicin and inhibited the efflux of rhodamine 123 from MDR lines but had no effect on drug accumulation or efflux in sensitive cells. Terfenadine 27-38 ATP binding cassette subfamily B member 1 Homo sapiens 91-105 8094615-7 1993 Terfenadine displaced azidopine from P-glycoprotein, but at concentrations higher than expected based on its overall potency. Terfenadine 0-11 ATP binding cassette subfamily B member 1 Homo sapiens 37-51 1280680-0 1992 Effect of terfenadine on the plasma concentrations of substance P and vasoactive intestinal polypeptide in volunteers. Terfenadine 10-21 tachykinin precursor 1 Homo sapiens 54-65 1280680-0 1992 Effect of terfenadine on the plasma concentrations of substance P and vasoactive intestinal polypeptide in volunteers. Terfenadine 10-21 vasoactive intestinal peptide Homo sapiens 70-103 1280680-1 1992 The effect of terfenadine on the plasma concentrations of substance P and vasoactive intestinal polypeptide (VIP) was studied in 7 healthy subjects and 8 subjects with the common cold. Terfenadine 14-25 tachykinin precursor 1 Homo sapiens 58-69 1280680-1 1992 The effect of terfenadine on the plasma concentrations of substance P and vasoactive intestinal polypeptide (VIP) was studied in 7 healthy subjects and 8 subjects with the common cold. Terfenadine 14-25 vasoactive intestinal peptide Homo sapiens 74-107 1280680-1 1992 The effect of terfenadine on the plasma concentrations of substance P and vasoactive intestinal polypeptide (VIP) was studied in 7 healthy subjects and 8 subjects with the common cold. Terfenadine 14-25 vasoactive intestinal peptide Homo sapiens 109-112 1280680-2 1992 Before terfenadine administration, the mean plasma substance P concentration of the subjects with the common cold was significantly higher than that of the healthy subjects. Terfenadine 7-18 tachykinin precursor 1 Homo sapiens 51-62 1280680-3 1992 The increased mean plasma substance P concentration of the subjects with the common cold was decreased after terfenadine administration. Terfenadine 109-120 tachykinin precursor 1 Homo sapiens 26-37 1280680-5 1992 The mean plasma VIP concentration of the subjects with common cold was slightly higher than that of the healthy subjects before and after terfenadine administration, with no significant difference. Terfenadine 138-149 vasoactive intestinal peptide Homo sapiens 16-19 1418048-1 1992 In a double-blind placebo-controlled randomized two-phase cross-over study comprising 10 (5/5) male subjects the effect of oral therapeutic doses of terfenadine (Teldane, CAS 50679-08-8) on rated performance, systolic and diastolic blood pressure, heart rate and the electroencephalogram of the subjects in an instrument flight (IF) procedure trainer was investigated. Terfenadine 149-160 BCAR1 scaffold protein, Cas family member Homo sapiens 171-174 8100726-2 1993 Oral terfenadine reduces the release of prostaglandin D2 (PGD2) and, to a lesser extent, the release of histamine in patients after nasal challenge with allergen. Terfenadine 5-16 prostaglandin D2 synthase Homo sapiens 40-56 1387041-5 1992 Bradykinin challenge induced mean maximal increases of 57%, 59%, 77% and 72% in NAR on the placebo, terfenadine, ipratropium bromide and terfenadine plus ipratropium bromide pretreatment days respectively. Terfenadine 100-111 kininogen 1 Homo sapiens 0-10 1387041-5 1992 Bradykinin challenge induced mean maximal increases of 57%, 59%, 77% and 72% in NAR on the placebo, terfenadine, ipratropium bromide and terfenadine plus ipratropium bromide pretreatment days respectively. Terfenadine 137-148 kininogen 1 Homo sapiens 0-10 1387041-8 1992 Bradykinin nasal challenge caused a mean maximal increase in albumin levels in recovered nasal lavages of 11.5, 13.0, 12.2 and 12.3 times of baseline levels on the placebo, terfenadine, ipratropium bromide and terfenadine plus ipratroprium bromide pretreatment days respectively. Terfenadine 173-184 kininogen 1 Homo sapiens 0-10 1387041-8 1992 Bradykinin nasal challenge caused a mean maximal increase in albumin levels in recovered nasal lavages of 11.5, 13.0, 12.2 and 12.3 times of baseline levels on the placebo, terfenadine, ipratropium bromide and terfenadine plus ipratroprium bromide pretreatment days respectively. Terfenadine 210-221 kininogen 1 Homo sapiens 0-10 1587633-1 1992 In this double-blind crossover study we evaluated the effect of terfenadine on the rise in neutrophil chemotactic activity (NCA) and eosinophil chemotactic activity (ECA) in serum induced by platelet-activating factor (PAF) inhalation in 8 asthmatics. Terfenadine 64-75 PCNA clamp associated factor Homo sapiens 191-217 1587633-4 1992 An initial elevation of NCA after PAF inhalation was inhibited by terfenadine, but the effect was diminished after subsequent PAF inhalations. Terfenadine 66-77 PCNA clamp associated factor Homo sapiens 34-37 1587633-6 1992 In vitro PAF- and fMLP-induced neutrophil chemotaxis were significantly inhibited by terfenadine. Terfenadine 85-96 PCNA clamp associated factor Homo sapiens 9-12 1587633-6 1992 In vitro PAF- and fMLP-induced neutrophil chemotaxis were significantly inhibited by terfenadine. Terfenadine 85-96 formyl peptide receptor 1 Homo sapiens 18-22 1970226-4 1990 In nine atopic asthmatic subjects, terfenadine 180 mg, when compared to placebo, increased the geometric mean provocation concentration of inhaled agonist required to reduce FEV1 by 20% of baseline (PC20) from 0.7 to greater than 22.9 mg/ml for histamine (P less than 0.01) and 0.3 to 0.5 mg/ml for bradykinin (P less than 0.01). Terfenadine 35-46 kininogen 1 Homo sapiens 299-309 1914619-7 1991 Terfenadine significantly inhibited the wheal and flare response to histamine and the flare response to injected PAF. Terfenadine 0-11 PCNA clamp associated factor Homo sapiens 113-116 1677829-4 1991 Terfenadine, a non-sedating histamine H1-receptor antagonist, caused bronchodilatation in a single dose. Terfenadine 0-11 histamine receptor H1 Homo sapiens 28-49 1707048-6 1991 The H1 antagonist terfenadine (120 mg po) significantly reduced the flare area associated with endothelin 1: flare 5 min after intradermal endothelin (10 pmol, placebo treated), 668 +/- 405 mm2; terfenadine treated, 201 +/- 257 mm2 (P less than 0.05). Terfenadine 18-29 endothelin 1 Homo sapiens 95-107 1976343-2 1990 We have assessed the effect of a specific histamine H1-receptor antagonist, terfenadine, in the treatment of atopic asthmatics during the grass pollen season. Terfenadine 76-87 histamine receptor H1 Homo sapiens 42-63 1777375-5 1991 Terfenadine improved 04.00 h baseline mean PEFR from 242 to 278 l min-1 (P less than 0.05) but a 38 l min-1 diurnal variation in mean PEFR persisted (P less than 0.05). Terfenadine 0-11 CD59 molecule (CD59 blood group) Homo sapiens 66-71 1976665-1 1990 We used a nonsedating, selective histamine H1-receptor antagonist, terfenadine, to investigate the effects of antihistamines on the microcirculatory changes and vascular permeability induced by topical histamine provocation challenge. Terfenadine 67-78 histamine receptor H1 Homo sapiens 33-54 1976343-0 1990 Terfenadine, a potent histamine H1-receptor antagonist in the treatment of grass pollen sensitive asthma. Terfenadine 0-11 histamine receptor H1 Homo sapiens 22-43 1970226-7 1990 Pharmacological intervention with terfenadine and flurbiprofen led to a significant protection of the airways against the constrictor effect of bradykinin but the effect in each case was small. Terfenadine 34-45 kininogen 1 Homo sapiens 144-154 1972050-2 1990 Terfenadine is a selective histamine H1-receptor antagonist which, in pharmacodynamic studies, is devoid of central nervous system depressant activity. Terfenadine 0-11 histamine receptor H1 Homo sapiens 27-48 1972050-4 1990 Terfenadine is superior to placebo, has a more rapid onset of action than astemizole and is as effective as most other histamine H1-receptor antagonists, in relieving rhinitis symptoms. Terfenadine 0-11 histamine receptor H1 Homo sapiens 119-140 33151169-0 2021 Inhibitory effect of terfenadine on Kir2.1 and Kir2.3 channels. Terfenadine 21-32 potassium inwardly rectifying channel subfamily J member 2 Homo sapiens 36-42 34680411-9 2021 Tg(myl7:GA) larvae treated with terfenadine showed bradycardia, 2:1 atrioventricular block, decreased time-averaged ventricular calcium levels but increased calcium transient amplitude, and reduced cardiac output. Terfenadine 32-43 myosin, light chain 7, regulatory Danio rerio 3-7 18585198-4 2008 Beer"s law is obeyed over the concentration ranges of 3-72, 3-96, 12-168 and 24-240 microg mL(-1) terfenadine using TCNQ, TCNE, DDQ and p-CLA, respectively, with correlation coefficients 0.9999, 0.9974, 0.9997 and 0.9979 and detection limits 0.3, 0.4, 2.6 and 12.3 microg mL(-1), for the reagents in the same order. Terfenadine 98-109 selectin P ligand Homo sapiens 138-141 18585198-5 2008 DDQ and p-CLA react spontaneously with terfenadine to give colored complexes that can be applied for the flow injection analysis of terfenadine in the concentration ranges 2.4-120 and 24-240 microg with correlation coefficients 0.9990 and 0.9985 and detection limits 0.8 and 2.7 microg for DDQ and p-CLA, respectively, in addition to the high sampling through output of 40 sample h(-1). Terfenadine 39-50 selectin P ligand Homo sapiens 10-13 18585198-5 2008 DDQ and p-CLA react spontaneously with terfenadine to give colored complexes that can be applied for the flow injection analysis of terfenadine in the concentration ranges 2.4-120 and 24-240 microg with correlation coefficients 0.9990 and 0.9985 and detection limits 0.8 and 2.7 microg for DDQ and p-CLA, respectively, in addition to the high sampling through output of 40 sample h(-1). Terfenadine 39-50 selectin P ligand Homo sapiens 300-303 18585198-5 2008 DDQ and p-CLA react spontaneously with terfenadine to give colored complexes that can be applied for the flow injection analysis of terfenadine in the concentration ranges 2.4-120 and 24-240 microg with correlation coefficients 0.9990 and 0.9985 and detection limits 0.8 and 2.7 microg for DDQ and p-CLA, respectively, in addition to the high sampling through output of 40 sample h(-1). Terfenadine 132-143 selectin P ligand Homo sapiens 10-13 18585198-5 2008 DDQ and p-CLA react spontaneously with terfenadine to give colored complexes that can be applied for the flow injection analysis of terfenadine in the concentration ranges 2.4-120 and 24-240 microg with correlation coefficients 0.9990 and 0.9985 and detection limits 0.8 and 2.7 microg for DDQ and p-CLA, respectively, in addition to the high sampling through output of 40 sample h(-1). Terfenadine 132-143 selectin P ligand Homo sapiens 300-303 33151169-0 2021 Inhibitory effect of terfenadine on Kir2.1 and Kir2.3 channels. Terfenadine 21-32 potassium inwardly rectifying channel subfamily J member 4 Homo sapiens 47-53 33151169-4 2021 Terfenadine inhibited Kir2.3 channels with a strikingly greater potency (IC50 = 1.06 +- 0.11 mumol L-1) compared to Kir2.1 channels (IC50 = 27.8 +- 4.8 mumol L-1). Terfenadine 0-11 potassium inwardly rectifying channel subfamily J member 4 Homo sapiens 22-28 33151169-5 2021 The Kir2.3(I213L) mutant, possessing a larger affinity for phosphatidylinositol 4,5-bisphosphate (PIP2) than the wild-type Kir2.3, was less sensitive to terfenadine inhibition (IC50 = 13.0 +- 2.9 mumol L-1). Terfenadine 153-164 potassium inwardly rectifying channel subfamily J member 4 Homo sapiens 4-10 33151169-5 2021 The Kir2.3(I213L) mutant, possessing a larger affinity for phosphatidylinositol 4,5-bisphosphate (PIP2) than the wild-type Kir2.3, was less sensitive to terfenadine inhibition (IC50 = 13.0 +- 2.9 mumol L-1). Terfenadine 153-164 potassium inwardly rectifying channel subfamily J member 4 Homo sapiens 123-129 33151169-6 2021 Additionally, the PIP2 intracellular application had largely reduced the inhibition of Kir2.1 channels by terfenadine. Terfenadine 106-117 potassium inwardly rectifying channel subfamily J member 2 Homo sapiens 87-93 2892555-9 1987 5 The H1-histamine antagonist terfenadine (60 mg orally) significantly inhibited the wheal and distant flare response to histamine (5 nmol) and NKA, but not that caused by NKB. Terfenadine 30-41 tachykinin precursor 1 Homo sapiens 144-147 2568881-1 1989 We have attempted to use a potent and selective histamine H1-receptor antagonist terfenadine to allow a larger dose of allergen to be administered to previous single early responders to investigate if an increased dose of allergen could induce a late asthmatic response. Terfenadine 81-92 histamine receptor H1 Homo sapiens 48-69 2562852-3 1989 In particular 1-[4-[3-[4-[bis(4-fluorophenyl)hydroxymethyl]-1-piperidinyl] propoxy]-3-methoxyphenyl]ethanone (1, AHR-5333) was more potent than oxatomide and terfenadine in this assay. Terfenadine 158-169 aryl hydrocarbon receptor Homo sapiens 113-116 3147222-7 1988 Two of these non-sedating antihistamines, terfenadine and astemizole, have novel binding characteristics with the histamine H1 receptor, exhibiting irreversible binding at higher concentrations. Terfenadine 42-53 histamine receptor H1 Homo sapiens 114-135 2903817-2 1988 In patients with chronic idiopathic urticaria, terfenadine was found to be as effective as the traditional antihistamine clemastine in reducing the number of wheals and the severity of itch, without causing drowsiness. Terfenadine 47-58 itchy E3 ubiquitin protein ligase Homo sapiens 185-189 2574555-1 1989 Results of a double-blind, randomized, placebo-controlled, parallel study in 37 patients indicate that terfenadine, 60 mg bid, is significantly more effective than placebo and as effective as hydroxyzine, 25 mg qid, in the treatment of chronic idiopathic urticaria without causing the somnolence that was associated with the use of hydroxyzine. Terfenadine 103-114 BH3 interacting domain death agonist Homo sapiens 122-125 2568212-1 1989 The chemistry, pharmacology, pharmacokinetics, clinical efficacy, adverse effects, and dosages of the nonsedating histamine H1-receptor antagonists terfenadine, astemizole, loratadine, and acrivastine are reviewed. Terfenadine 148-159 histamine receptor H1 Homo sapiens 114-135 3125852-2 1987 Terfenadine, a histamine H1-receptor antagonist, was studied in 52 patients with stable mild perennial asthma with an allergic basis. Terfenadine 0-11 histamine receptor H1 Homo sapiens 15-36 2879489-0 1987 Terfenadine (Seldane) is a potent and selective histamine H1 receptor antagonist in asthmatic airways. Terfenadine 0-11 histamine receptor H1 Homo sapiens 48-69 2885357-1 1987 For some patients, terfenadine, in the currently recommended dose of 60 mg twice daily (bid), may be only modestly effective in the treatment of allergic rhinitis. Terfenadine 19-30 BH3 interacting domain death agonist Homo sapiens 88-91 2885357-4 1987 The 300 mg bid terfenadine regimen produced significantly greater skin test suppression (p less than 0.05) than the currently recommended 60 mg bid dose. Terfenadine 15-26 BH3 interacting domain death agonist Homo sapiens 11-14 2879489-0 1987 Terfenadine (Seldane) is a potent and selective histamine H1 receptor antagonist in asthmatic airways. Terfenadine 13-20 histamine receptor H1 Homo sapiens 48-69 6129861-5 1982 The result of this exercise is terfenadine, a preferred antihistamine, potent and selective in antagonizing histamine H1-receptor-mediated responses but lacking the usual side effect profile of known histamine H1-receptor antagonists. Terfenadine 31-42 histamine receptor H1 Homo sapiens 108-129 2857636-8 1985 Thus, terfenadine offers a worthwhile improvement in side effect profile over "traditional" antihistamines, and could well become an "agent of choice" (along with other non-sedating antihistamines) in many patients in whom a histamine H1-receptor antagonist is indicated. Terfenadine 6-17 histamine receptor H1 Homo sapiens 225-246 2875234-7 1986 Terfenadine, a purportedly selective histamine H1-receptor antagonist, blocked both histamine and acetylcholine-induced bronchoconstriction at doses similar to SK&F 93944. Terfenadine 0-11 histamine receptor H1 Canis lupus familiaris 37-58 6129861-5 1982 The result of this exercise is terfenadine, a preferred antihistamine, potent and selective in antagonizing histamine H1-receptor-mediated responses but lacking the usual side effect profile of known histamine H1-receptor antagonists. Terfenadine 31-42 histamine receptor H1 Homo sapiens 200-221 6129862-5 1982 The unsurmountable antagonism of histamine by terfenadine may result from a slow dissociation of terfenadine from the histamine H1-receptor. Terfenadine 46-57 histamine H1 receptor Cavia porcellus 118-139 6129862-7 1982 It is concluded that terfenadine is a potent, selective histamine H1-receptor antagonist; the kinetics of association/dissociation of terfenadine with histamine H1-receptors may account for the long-lasting antihistaminic effect in various animal models. Terfenadine 21-32 histamine H1 receptor Cavia porcellus 56-77 6129874-1 1982 alpha-[4-(1,1-Dimethylethyl)phenyl]-4-(hydroxydiphenylmethyl)-1- piperidinebutanol (terfenadine, RMI 9918, Triludan, Teldane, resp.) is a new histamine H1-receptor antagonist. Terfenadine 0-82 histamine receptor H1 Homo sapiens 142-163 30561737-7 2019 Dofetilide, cisapride, sotalol, terfenadine, and verapamil blocked hERG channels at 37 C with an IC50 of 7 nM, 18 nM, 343 muM, 165 nM, and 214 nM, respectively. Terfenadine 32-43 ETS transcription factor ERG Homo sapiens 67-71 31730936-13 2020 Recalculating the TdP metric and margin values for terfenadine, risperidone and astemizole using the unbound concentration normally associated with treatment and a clinical worst case changes the qNet metric to higher risk values and illustrates the potential benefit to the algorithm of consistently using a clinical high exposure scenario accounting for all "hERG-active species". Terfenadine 51-62 ETS transcription factor ERG Homo sapiens 361-365 6109770-2 1980 The keto analog of terfenadine was converted to its O-carboxymethyloxime derivative, which was conjugated to bovine thyroglobulin by a mixed anhydride technique. Terfenadine 19-30 thyroglobulin Bos taurus 116-129 33335233-7 2020 Molecular docking of a series of compounds including amiodarone, astemizole, danazol, ebastine, ketoconazole, terfenadine, terfenadone, and arachidonic acid to CYP2J2 confirmed the role of those residues in determining substrate binding and specificity of CYP2J2. Terfenadine 110-121 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 160-166 33335233-7 2020 Molecular docking of a series of compounds including amiodarone, astemizole, danazol, ebastine, ketoconazole, terfenadine, terfenadone, and arachidonic acid to CYP2J2 confirmed the role of those residues in determining substrate binding and specificity of CYP2J2. Terfenadine 110-121 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 256-262 32164398-6 2020 Treatment with terfenadine was associated with 10-fold higher expression of atrial natriuretic peptide (p < 0.001 vs. vehicle), increased p53 mRNA expression, and chromatin fragmentation in the TUNEL assay, all of which were significantly reduced by fimasartan treatment. Terfenadine 15-26 tumor protein p53 Danio rerio 138-141 32249832-8 2020 In addition, hiPSC-IECs oxidatively metabolized terfenadine (CYP3A and CYP2J2 substrate) and midazolam (CYP3A substrate). Terfenadine 48-59 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 61-66 32249832-8 2020 In addition, hiPSC-IECs oxidatively metabolized terfenadine (CYP3A and CYP2J2 substrate) and midazolam (CYP3A substrate). Terfenadine 48-59 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 71-77 31664867-5 2019 Our functional data from isolated zkcnh6a channels show that these channels respond to hERG1a channel blockers (dofetilide and terfenadine), and type 1 (RPR260243) and type 2 (NS1643, PD-118057) hERG1a activator compounds, in a similar manner to hKCNH2a channels, with minor differences largely accounted for by subtly different biophysical properties in the two channels. Terfenadine 127-138 potassium voltage-gated channel subfamily H member 2 Homo sapiens 87-92 31183371-8 2019 Conclusions: Our study revealed for the first time that second-generation antihistamines, including cetirizine, fexofenadine, azelastine, and terfenadine, exert suppressive effects on lymphocyte Kv1.3-channels. Terfenadine 142-153 potassium voltage-gated channel, shaker-related subfamily, member 3 Mus musculus 195-200 30483130-5 2018 In vivo arrhythmia model in Cmlc2-GFP transgenic zebrafish was induced by terfenadine, which exhibited obvious reduction of heart rate and occurrence of atrioventricular block. Terfenadine 74-85 myosin, light chain 7, regulatory Danio rerio 28-33 30206647-7 2018 In current clamp, ERG channel blockers, terfenadine (10 microM) and E-4031 (10 microM), were applied in both cell types. Terfenadine 40-51 ETS transcription factor Mus musculus 18-21 28436281-2 2018 Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Terfenadine 81-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-41 29759885-2 2018 In a previous study, we used recombinant CYP2J2 and determined whether danazol, hydroxyebastine, telmisartan, and terfenadone inhibited CYP2J2 by using four representative CYP2J2 substrates, namely albendazole, astemizole, ebastine, and terfenadine. Terfenadine 237-248 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 136-142 29759885-2 2018 In a previous study, we used recombinant CYP2J2 and determined whether danazol, hydroxyebastine, telmisartan, and terfenadone inhibited CYP2J2 by using four representative CYP2J2 substrates, namely albendazole, astemizole, ebastine, and terfenadine. Terfenadine 237-248 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 136-142 29759885-4 2018 Among the four CYP2J2 inhibitors tested, terfenadone was strongest inhibitor of CYP2J2-mediated metabolism of albendazole, astemizole, and terfenadine with IC50 values of 0.31, 0.15, and 2.11 muM, respectively, in human liver microsomes (HLMs). Terfenadine 139-150 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 15-21 29759885-4 2018 Among the four CYP2J2 inhibitors tested, terfenadone was strongest inhibitor of CYP2J2-mediated metabolism of albendazole, astemizole, and terfenadine with IC50 values of 0.31, 0.15, and 2.11 muM, respectively, in human liver microsomes (HLMs). Terfenadine 139-150 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 80-86 29759885-4 2018 Among the four CYP2J2 inhibitors tested, terfenadone was strongest inhibitor of CYP2J2-mediated metabolism of albendazole, astemizole, and terfenadine with IC50 values of 0.31, 0.15, and 2.11 muM, respectively, in human liver microsomes (HLMs). Terfenadine 139-150 latexin Homo sapiens 192-195 29548821-5 2018 Using terfenadine, a selective chemical inhibitor of HRH1, we showed that the inhibition of HRH1 activity in basal BC cells leads to sub-G0 cell accumulation, suppresses proliferation, promotes cell motility and triggers the activation of extracellular signal-regulated kinase (ERK) signaling, initiating the mitochondrial apoptotic pathway. Terfenadine 6-17 histamine receptor H1 Homo sapiens 53-57 29548821-5 2018 Using terfenadine, a selective chemical inhibitor of HRH1, we showed that the inhibition of HRH1 activity in basal BC cells leads to sub-G0 cell accumulation, suppresses proliferation, promotes cell motility and triggers the activation of extracellular signal-regulated kinase (ERK) signaling, initiating the mitochondrial apoptotic pathway. Terfenadine 6-17 histamine receptor H1 Homo sapiens 92-96 29548821-5 2018 Using terfenadine, a selective chemical inhibitor of HRH1, we showed that the inhibition of HRH1 activity in basal BC cells leads to sub-G0 cell accumulation, suppresses proliferation, promotes cell motility and triggers the activation of extracellular signal-regulated kinase (ERK) signaling, initiating the mitochondrial apoptotic pathway. Terfenadine 6-17 mitogen-activated protein kinase 1 Homo sapiens 239-276 29548821-5 2018 Using terfenadine, a selective chemical inhibitor of HRH1, we showed that the inhibition of HRH1 activity in basal BC cells leads to sub-G0 cell accumulation, suppresses proliferation, promotes cell motility and triggers the activation of extracellular signal-regulated kinase (ERK) signaling, initiating the mitochondrial apoptotic pathway. Terfenadine 6-17 mitogen-activated protein kinase 1 Homo sapiens 278-281 29548821-6 2018 Furthermore, HER2-targeted therapy-resistant cells express higher levels of HRH1 and are more sensitive to terfenadine treatment. Terfenadine 107-118 erb-b2 receptor tyrosine kinase 2 Homo sapiens 13-17 28436281-2 2018 Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Terfenadine 147-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-41 29563525-7 2018 Here, we have directly assessed the role of inactivation in cisparide and terfenadine drug binding in mutant (I663P) hERG channels where the activation gate is trapped-open. Terfenadine 74-85 ETS transcription factor ERG Homo sapiens 117-121 29223463-0 2018 Terfenadine metabolism of human cytochrome P450 2J2 containing genetic variations (G312R, P351L and P115L). Terfenadine 0-11 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 32-51 29035434-11 2017 A significant increase of natriuretic peptide B (NPPB) mRNA was found in terfenadine-treated zebrafish. Terfenadine 73-84 natriuretic peptide B Danio rerio 26-47 29175952-4 2018 Three chemically diverse ERG channel blockers (terfenadine, ErgToxin-1, and E-4031) abolished persistent firing and the underlying increase in input resistance in deep pyramidal cells in temporal and prefrontal association neocortex. Terfenadine 47-58 ETS transcription factor ERG Rattus norvegicus 25-28 29035434-11 2017 A significant increase of natriuretic peptide B (NPPB) mRNA was found in terfenadine-treated zebrafish. Terfenadine 73-84 natriuretic peptide B Danio rerio 49-53 26899760-0 2016 Marmoset cytochrome P450 2J2 mainly expressed in small intestines and livers effectively metabolizes human P450 2J2 probe substrates, astemizole and terfenadine. Terfenadine 149-160 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 9-28 27553911-7 2017 Compared to PSS, the IC50 values, conducted at room temperature, of the classic hERG blocking drugs cisapride, moxifloxacin, and terfenadine were shifted to the right by an extent predicted by their known plasma protein binding, but we did not detect any differences in IC50 s between male and female serum. Terfenadine 129-140 ETS transcription factor ERG Homo sapiens 80-84 28237650-4 2017 Additionally, CYP2J2 metabolizes a number of xenobiotics such as astemizole and terfenadine and is potently inhibited by danazol and telmisartan. Terfenadine 80-91 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 14-20 29911373-3 2017 Additionally, CYP2J2 plays an indispensable role in the intestinal metabolism of various drugs, such as astemizole, terfenadine and ebastine. Terfenadine 116-127 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 14-20 27681411-6 2016 Using qRT-PCR analysis, we demonstrated that, among those genes, p21 (Cdc42/Rac)-activated kinase 1 (pak1) mRNA was downregulated by treatment with terfenadine and astemizole. Terfenadine 148-159 KRAS proto-oncogene, GTPase Rattus norvegicus 65-68 27681411-6 2016 Using qRT-PCR analysis, we demonstrated that, among those genes, p21 (Cdc42/Rac)-activated kinase 1 (pak1) mRNA was downregulated by treatment with terfenadine and astemizole. Terfenadine 148-159 cell division cycle 42 Rattus norvegicus 70-99 27681411-6 2016 Using qRT-PCR analysis, we demonstrated that, among those genes, p21 (Cdc42/Rac)-activated kinase 1 (pak1) mRNA was downregulated by treatment with terfenadine and astemizole. Terfenadine 148-159 p21 (RAC1) activated kinase 1 Rattus norvegicus 101-105 27035465-7 2016 RESULTS: A 48 hours exposure of human erythrocytes to Terfenadine (>= 5 microM) significantly increased the percentage of annexin-V-binding cells and triggered hemolysis without significantly modifying the average forward scatter. Terfenadine 54-65 annexin A5 Homo sapiens 125-134 27035465-9 2016 The effect of Terfenadine on annexin-V-binding was significantly blunted but not abolished by removal of extracellular Ca2+. Terfenadine 14-25 annexin A5 Homo sapiens 29-38 27035465-10 2016 Exposure of human erythrocytes to Ca2+ ionophore ionomycin (1 microM, 15 min) triggered annexin-V-binding, an effect augmented by Terfenadine pretreatment (10 microM, 48 hours). Terfenadine 130-141 annexin A5 Homo sapiens 88-97 26358220-6 2015 The alpha-MSH-induced scratching was inhibited by the mu-opioid receptor antagonist naltrexone and the H1 histamine receptor antagonist terfenadine. Terfenadine 136-147 pro-opiomelanocortin-alpha Mus musculus 4-13 26358220-7 2015 In mast cell-deficient mice, alpha-MSH also elicited scratching, which was inhibited by terfenadine. Terfenadine 88-99 pro-opiomelanocortin-alpha Mus musculus 29-38 26048912-4 2015 Of these four CYP2J2 inhibitors, danazol strongly inhibited CYP2J2-mediated albendazole, astemizole, ebastine, and terfenadine metabolism in a substrate-independent manner, with IC50 values of 0.05, 0.07, 0.18, and 0.34 muM, respectively. Terfenadine 115-126 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 60-66 26376488-11 2015 Furthermore, the equine KV11.1 channel was susceptible to pharmacological block with terfenadine. Terfenadine 85-96 potassium voltage-gated channel subfamily H member 2 Homo sapiens 24-30 26048912-1 2015 Cytochrome P450 2J2 (CYP2J2) is an enzyme responsible for the metabolism of endogenous substrates including arachidonic acid, as well as therapeutic drugs such as albendazole, astemizole, ebastine, and terfenadine. Terfenadine 202-213 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 0-19 26048912-1 2015 Cytochrome P450 2J2 (CYP2J2) is an enzyme responsible for the metabolism of endogenous substrates including arachidonic acid, as well as therapeutic drugs such as albendazole, astemizole, ebastine, and terfenadine. Terfenadine 202-213 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 21-27 26048912-6 2015 Terfenadone strongly inhibited CYP2J2-mediated albendazole, astemizole, and terfenadine metabolism (IC50 < 0.21 muM), whereas it showed weak inhibition against CYP2J2-catalyzed ebastine hydroxylase activity (IC50 = 6.04 muM). Terfenadine 76-87 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 31-37 25238555-5 2014 This scaffold suffers from hERG liabilities which were not remedied through this round of optimization; however, given the overall improvement in activity of the set, terfenadine-based analogues provide a novel structural class of antimicrobial compounds with potential for further characterization as part of the continuing process to meet the current need for new antibiotics. Terfenadine 167-178 ETS transcription factor ERG Homo sapiens 27-31 25857771-6 2015 RESULTS: We show that drug-related compounds, debrisoquine and terfenadine derivatives, subtrates of CYP2D6 and CYP2J2, are hydroxylated by recombinant CYP2U1 with regioselectivities different from those reported for CYP2D6 and 2J2. Terfenadine 63-74 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 101-107 25857771-6 2015 RESULTS: We show that drug-related compounds, debrisoquine and terfenadine derivatives, subtrates of CYP2D6 and CYP2J2, are hydroxylated by recombinant CYP2U1 with regioselectivities different from those reported for CYP2D6 and 2J2. Terfenadine 63-74 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 112-118 25857771-6 2015 RESULTS: We show that drug-related compounds, debrisoquine and terfenadine derivatives, subtrates of CYP2D6 and CYP2J2, are hydroxylated by recombinant CYP2U1 with regioselectivities different from those reported for CYP2D6 and 2J2. Terfenadine 63-74 cytochrome P450 family 2 subfamily U member 1 Homo sapiens 152-158 25857771-6 2015 RESULTS: We show that drug-related compounds, debrisoquine and terfenadine derivatives, subtrates of CYP2D6 and CYP2J2, are hydroxylated by recombinant CYP2U1 with regioselectivities different from those reported for CYP2D6 and 2J2. Terfenadine 63-74 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 217-223 24997310-0 2014 A note on CYP2J2-mediated terfenadine hydroxylation in human liver microsomes. Terfenadine 26-37 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 10-16 24998194-0 2014 Response to comment: "A note on CYP2J2-mediated terfenadine hydroxylation in human liver microsomes". Terfenadine 48-59 cytochrome P450 family 2 subfamily J member 2 Homo sapiens 32-38