Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Drug Metab Pharmacokinet ; 34(5): 325-333, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31564409

ABSTRACT

By using the Relative Factor (RF) method-a method that can simply assess cytochrome P450 (CYP) induction risk based on a maximum induction effect model-we evaluated the risk of CYP2C9 induction and examined its relationship with risk of CYP3A4 induction. In cryopreserved human hepatocytes, the magnitude of CYP2C9 induction by eight drugs known to induce CYP3A4 was lower than the magnitude of CYP3A4 induction, but the magnitudes of induction of both were correlated. The RF values determined for CYP2C9 had a one-to-one linear relationship with values determined for CYP3A4, supporting reports that the induction mechanism of both enzymes is the same. Furthermore, clinical CYP2C9 induction data of compounds reported to induce CYP2C9 clinically were shown to be lower than those of CYP3A4. The thresholds for CYP2C9 induction risk assessment by the RF approach were determined to be at higher steady-state plasma concentrations than those for CYP3A4. Based on these results, induction of CYP2C9 was correlated with that of CYP3A4, and induction risk could be evaluated by the RF method using hepatocytes. The CYP2C9 induction risk of a compound was confirmed to be lower than its CYP3A4 induction risk.


Subject(s)
Cytochrome P-450 CYP2C9 Inducers/pharmacology , Cytochrome P-450 CYP2C9/metabolism , Hepatocytes/drug effects , Cells, Cultured , Cytochrome P-450 CYP2C9 Inducers/analysis , Dose-Response Relationship, Drug , Hepatocytes/enzymology , Humans , Risk Factors , Structure-Activity Relationship
2.
Cancer Chemother Pharmacol ; 84(2): 393-404, 2019 08.
Article in English | MEDLINE | ID: mdl-31227862

ABSTRACT

PURPOSE: Ipatasertib is a selective inhibitor of Akt, a frequently activated protein kinase in human cancers. The current study assessed the safety, tolerability, and pharmacokinetics of ipatasertib in Japanese patients with solid tumors. METHODS: This was a phase I, open-label, 3 + 3 dose-escalation study conducted in two stages. In stage I, Japanese patients with solid tumors were administered ipatasertib 200, 400, or 600 mg/day for 21 days of a 28-day cycle. In stage II, Japanese patients with castration-resistant prostate cancer were administered ipatasertib 200 or 400 mg/day in combination with abiraterone and prednisolone in 28-day cycles. Dose-limiting toxicity (DLT) was assessed at each dose before enrolling patients at a higher dose; DLT was used to determine the maximum tolerated dose (MTD) and maximum administered dose (MAD). Pharmacokinetic parameters were assessed after a single dose and at steady state. RESULTS: Fifteen patients were enrolled in Stage I and six in Stage II. The ipatasertib MTD was 600 mg as monotherapy and MAD was 400 mg in combination with abiraterone and prednisolone. Ipatasertib plasma exposure was dose proportional across the dose range, and was not markedly affected by concurrent administration of abiraterone plus prednisolone. Stable disease (SD) was observed in eight patients treated with ipatasertib monotherapy (53.3%); four patients had SD and one had complete response with ipatasertib plus abiraterone and prednisolone. CONCLUSIONS: Ipatasertib, at the monotherapy MTD of 600 mg/day and MAD of 400 mg/day in combination with abiraterone and prednisolone, was safe and tolerable in Japanese patients with solid tumors.


Subject(s)
Androstenes/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Prednisolone/therapeutic use , Adult , Androstenes/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Female , Humans , Japan , Male , Middle Aged , Prednisolone/pharmacology
3.
AAPS J ; 21(3): 42, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30887238

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model was used to simulate the impact of elevated levels of interleukin (IL)-6 on the exposure of several orally administered cytochrome P450 (CYP) probe substrates (caffeine, S-warfarin, omeprazole, dextromethorphan, midazolam, and simvastatin). The changes in exposure of these substrates in subjects with rheumatoid arthritis (and hence elevated IL-6 levels) compared with healthy subjects were predicted with a reasonable degree of accuracy. The PBPK model was then used to simulate the change in oral exposure of the probe substrates in North European Caucasian, Chinese, and Japanese population of patients with neuromyelitis optica (NMO) or NMO spectrum disorder with elevated plasma IL-6 levels (up to 100 pg/mL). Moderate interactions [mean AUC fold change, ≤ 2.08 (midazolam) or 2.36 (simvastatin)] was predicted for CYP3A4 probe substrates and weak interactions (mean AUC fold change, ≤ 1.29-1.97) were predicted for CYP2C19, CYP2C9, and CYP2D6 substrates. No notable interaction was predicted with CYP1A2. Although ethnic differences led to differences in simulated exposure for some of the probe substrates, there were no marked differences in the predicted magnitude of the change in exposure following IL-6-mediated suppression of CYPs. Decreased levels of serum albumin (as reported in NMO patients) had little impact on the magnitude of the simulated IL-6-mediated drug interactions. This PBPK modeling approach allowed us to leverage knowledge from different disease and ethnic populations to make predictions of cytokine-related DDIs in a rare disease population where actual clinical studies would otherwise be difficult to conduct.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Interleukin-6/metabolism , Models, Biological , Neuromyelitis Optica/drug therapy , Rare Diseases/drug therapy , Administration, Oral , Adult , Caffeine/administration & dosage , Caffeine/pharmacokinetics , Clinical Trials as Topic , Computer Simulation , Dextromethorphan/administration & dosage , Dextromethorphan/pharmacokinetics , Down-Regulation , Drug Development , Drug Interactions , Female , Humans , Interleukin-6/blood , Male , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Middle Aged , Neuromyelitis Optica/blood , Neuromyelitis Optica/ethnology , Neuromyelitis Optica/metabolism , Omeprazole/administration & dosage , Omeprazole/pharmacokinetics , Rare Diseases/blood , Rare Diseases/ethnology , Rare Diseases/metabolism , Serum Albumin, Human/analysis , Simvastatin/administration & dosage , Simvastatin/pharmacokinetics , Warfarin/administration & dosage , Warfarin/pharmacokinetics
4.
Biol Pharm Bull ; 41(1): 47-56, 2018.
Article in English | MEDLINE | ID: mdl-29311482

ABSTRACT

Determinants of interindividual variability in erlotinib pharmacokinetics (PK) and adverse events remain to be elucidated. This study with 50 Japanese non-small-cell lung cancer patients treated with oral erlotinib at a standard dose of 150 mg aimed to investigate whether genetic polymorphisms affect erlotinib PK and adverse events. Single nucleotide polymorphisms (SNPs) in genes encoding metabolizing enzymes (CYP1A1, CYP1A2, CYP2D6, CYP3A4, CYP3A5, UGT1A1, UGT2B7, GSTM1, and GSTT1) or efflux transporters (ABCB1, and ABCG2) were analyzed as covariates in a population PK model. The ABCB1 1236C>T (rs1128503) polymorphism, not ABCB1*2 haplotype (1236TT-2677TT-3455TT, rs1128503 TT-rs2032582 TT-rs1045642 TT), was a significant covariate for the apparent clearance (CL/F), with the TT genotype showing a 29.4% decrease in CL/F as compared with the CC and the CT genotypes. A marginally higher incidence of adverse events (mainly skin rash) was observed in the TT genotype group; however, patients with high plasma erlotinib exposure did not always experience skin rash. None of the other SNPs affected PK or adverse events. The ABCB1 genotype is a potential predictor for erlotinib adverse events. Erlotinib might be used with careful monitoring of adverse events in patients with ABCB1 polymorphic variants.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Drug-Related Side Effects and Adverse Reactions/etiology , Erlotinib Hydrochloride/pharmacokinetics , Lung Neoplasms/metabolism , Models, Biological , Polymorphism, Single Nucleotide , ATP-Binding Cassette Transporters/genetics , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/epidemiology , Erlotinib Hydrochloride/adverse effects , Erlotinib Hydrochloride/therapeutic use , Female , Glucuronosyltransferase/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...