PMID-sentid Pub_year Sent_text comp_official_name comp_offsetprotein_name organism prot_offset 34169526-6 2021 Ruxolitinib PK was adequately described with a 2-compartment model with first-order absorption and elimination with distribution volumes normalized to mean body weight (91.5 kg) and a separate typical CL for participants administered efavirenz (a known CYP3A4 inducer). ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 253-259 34505930-12 2021 CONCLUSION: Ruxolitinib exposure is increased in GvHD patients in comparison to myelofibrosis patients due to reduced clearance and comedication with CYP3A4 or CYP2C9 inhibitors. ruxolitinib 12-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 150-156 30069628-10 2018 The metabolization of ruxolitinib through CYP3A4 needs to be considered particularly if co-administered with potent CYP3A4 inhibitors. ruxolitinib 22-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 30069628-10 2018 The metabolization of ruxolitinib through CYP3A4 needs to be considered particularly if co-administered with potent CYP3A4 inhibitors. ruxolitinib 22-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 30645821-14 2016 Ruxolitinib is mainly metabolised by cytochrome P450 isoenzymes CYP3A4 and CYP2C9, creating a risk of multiple drug interactions. ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 64-70 25670523-2 2015 A ruxolitinib physiologically based pharmacokinetic model was constructed using all baseline PK data in healthy subjects, and verified by retrospective predictions of observed drug-drug interactions with rifampin (a potent CYP3A4 inducer), ketoconazole (a potent CYP3A4 reversible inhibitor) and erythromycin (a moderate time-dependent inhibitor of CYP3A4). ruxolitinib 2-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 223-229 25670523-2 2015 A ruxolitinib physiologically based pharmacokinetic model was constructed using all baseline PK data in healthy subjects, and verified by retrospective predictions of observed drug-drug interactions with rifampin (a potent CYP3A4 inducer), ketoconazole (a potent CYP3A4 reversible inhibitor) and erythromycin (a moderate time-dependent inhibitor of CYP3A4). ruxolitinib 2-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 263-269 25670523-2 2015 A ruxolitinib physiologically based pharmacokinetic model was constructed using all baseline PK data in healthy subjects, and verified by retrospective predictions of observed drug-drug interactions with rifampin (a potent CYP3A4 inducer), ketoconazole (a potent CYP3A4 reversible inhibitor) and erythromycin (a moderate time-dependent inhibitor of CYP3A4). ruxolitinib 2-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 263-269 25670523-3 2015 The model prospectively predicts that 100-200 mg daily dose of fluconazole, a dual inhibitor of CYP3A4 and 2C9, would increase ruxolitinib plasma concentration area under the curve by ~two-fold, and that as a perpetrator, ruxolitinib is highly unlikely to have any discernible effect on digoxin, a sensitive P-glycoprotein substrate. ruxolitinib 127-138 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 96-110 24756798-6 2014 The metabolization of ruxolitinib through CYP3A4 needs to be considered particularly if co-administered with potent CYP3A4 inhibitors. ruxolitinib 22-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 24756798-6 2014 The metabolization of ruxolitinib through CYP3A4 needs to be considered particularly if co-administered with potent CYP3A4 inhibitors. ruxolitinib 22-33 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 116-122 21602517-1 2012 Ruxolitinib, a selective Janus kinase (JAK) 1&2 inhibitor in development for the treatment of myeloproliferative neoplasms, is primarily metabolized by CYP3A4. ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 156-162 21602517-2 2012 The effects of inhibition or induction of CYP3A4 on single oral dose ruxolitinib pharmacokinetics (PK) and pharmacodynamics (PD) were evaluated in healthy volunteers. ruxolitinib 69-80 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 42-48 21602517-3 2012 Coadministration of ketoconazole (a potent CYP3A4 inhibitor) and erythromycin (a moderate CYP3A4 inhibitor) increased total ruxolitinib plasma exposure (AUC(0- )) by 91% and 27%, respectively, and ruxolitinib PD, as measured by the inhibition of interleukin (IL)-6-stimulated STAT3 phosphorylation in whole blood, was generally consistent with the PK observed. ruxolitinib 124-135 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 21602517-4 2012 Pretreatment with rifampin, a potent CYP3A4 inducer, decreased ruxolitinib AUC(0- ) by 71% while resulting in only a 10% decrease in the overall PD activity. ruxolitinib 63-74 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 37-43 21602517-6 2012 The collective PK/PD data suggest that starting doses of ruxolitinib should be reduced by 50% if coadministered with a potent CYP3A4 inhibitor, whereas adjustments in ruxolitinib starting doses may not be needed when coadministered with inducers or mild/moderate inhibitors of CYP3A4. ruxolitinib 57-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 126-132 32234672-6 2020 IL-22 was next found to activate the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) 3 pathway, whose inhibition by the JAK inhibitor ruxolitinib fully prevented the IL-22-mediated CYP3A4, CYP2B6 and NTCP repression in HepaRG cells. ruxolitinib 161-172 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 208-214 31941838-9 2020 Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. ruxolitinib 134-145 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 105-111 31145690-0 2019 Drug-drug interaction (DDI) assessments of ruxolitinib, a dual substrate of CYP3A4 and CYP2C9, using a verified physiologically based pharmacokinetic (PBPK) model to support regulatory submissions. ruxolitinib 43-54 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 31145690-1 2019 Ruxolitinib is mainly metabolized by cytochrome P450 (CYP) enzymes CYP3A4 and CYP2C9 followed by minor contributions of other hepatic CYP enzymes in vitro. ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 67-73 31145690-2 2019 A physiologically based pharmacokinetic (PBPK) model was established to evaluate the changes in the ruxolitinib systemic exposures with co-administration of CYP3A4 and CYP2C9 perpetrators. ruxolitinib 100-111 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 157-163 31145690-3 2019 The fractions metabolized in the liver via oxidation by CYP enzymes (fm,CYP3A4 = 0.75, fm,CYP2C9 = 0.19, and fm,CYPothers = 0.06) for an initial ruxolitinib model based on in vitro data were optimized (0.43, 0.56, and 0.01, respectively) using the observed exposure changes of ruxolitinib (10 mg) with co-administered ketoconazole (200 mg). ruxolitinib 145-156 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 72-78 31145690-7 2019 The estimated AUC ratios of ruxolitinib by co-administration of the moderate CYP3A4 inhibitor erythromycin (500 mg) and the strong CYP3A4 inducer rifampicin (600 mg) were within a 20% error compared with the clinically observed values. ruxolitinib 28-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 77-83 31145690-7 2019 The estimated AUC ratios of ruxolitinib by co-administration of the moderate CYP3A4 inhibitor erythromycin (500 mg) and the strong CYP3A4 inducer rifampicin (600 mg) were within a 20% error compared with the clinically observed values. ruxolitinib 28-39 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 131-137 31145690-9 2019 Furthermore, an AUC increase of ruxolitinib in the absence or presence of the dual CYP3A4 and CYP2C9 inhibitor fluconazole (100-400 mg) was prospectively estimated to be 1.94- to 4.31-fold. ruxolitinib 32-43 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 83-89 31145690-11 2019 A ruxolitinib PBPK model with optimized fm,CYP3A4 and fm,CYP2C9 was established to evaluate victim DDI risks. ruxolitinib 2-13 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 43-49 24589536-5 2014 Ruxolitinib is primarily metabolized by the cytochrome P-450 (CYP) 3A4 isoenzyme system; therefore, if concomitant use with a strong CYP3A4 inhibitor is unavoidable, an initial dosage reduction is warranted. ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 44-70 24589536-5 2014 Ruxolitinib is primarily metabolized by the cytochrome P-450 (CYP) 3A4 isoenzyme system; therefore, if concomitant use with a strong CYP3A4 inhibitor is unavoidable, an initial dosage reduction is warranted. ruxolitinib 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 133-139