Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Transl Sci ; 16(9): 1713-1724, 2023 09.
Article in English | MEDLINE | ID: mdl-37553804

ABSTRACT

Futibatinib is a covalently binding FGFR1-4 inhibitor that received US Food and Drug Administration approval for the treatment of patients with previously treated, advanced intrahepatic cholangiocarcinoma harboring FGFR2 gene fusions/rearrangements. This phase I trial evaluated the pharmacokinetics (PKs), safety, and tolerability of futibatinib in subjects with impaired hepatic function and matched healthy volunteers. Twenty-two subjects with hepatic impairment (8 mild [Child-Pugh 5-6], 8 moderate [7-9], and 6 severe [10-15]) and 16 matched healthy control subjects received a single oral dose of futibatinib 20 mg. Futibatinib PKs were compared between subjects with mild/moderate/severe hepatic impairment and each corresponding control cohort and the overall control cohort. Relationships between futibatinib PKs and Child-Pugh scores and liver function tests were examined via scatter/regression plots. Compared with matched controls, the area under the plasma concentration-time curve from time zero to infinity increased by 21%/20%/18% and the maximum plasma concentration (Cmax ) increased by 43%/15%/10% in subjects with mild/moderate/severe hepatic impairment, respectively. Changes were not considered clinically relevant: geometric mean ratios were within 80%-125%, except for Cmax in subjects with mild hepatic impairment (143%). No obvious trends were observed among futibatinib PK parameters versus Child-Pugh scores, bilirubin, albumin, international normalized ratio, and aspartate aminotransferase (all p > 0.05). Futibatinib was well-tolerated, with only four grade 1 treatment-emergent adverse events (mild hepatic impairment = 2 and control = 2). The results demonstrate that futibatinib dose adjustments due to mild/moderate/severe hepatic impairment are not necessary in patients receiving futibatinib 20 mg daily.


Subject(s)
Liver Diseases , Humans , Area Under Curve , Liver Diseases/diagnosis , Liver Diseases/drug therapy , Pyrroles/therapeutic use , Pyrimidines/adverse effects
2.
Clin Pharmacol Drug Dev ; 12(9): 927-939, 2023 09.
Article in English | MEDLINE | ID: mdl-37300358

ABSTRACT

Futibatinib, a selective, irreversible fibroblast growth factor receptor 1-4 inhibitor, was recently approved for FGFR2 rearrangement-positive cholangiocarcinoma. This Phase I study evaluated the mass balance and metabolic profile of 14 C-futibatinib single oral 20-mg dose in healthy participants (n = 6). Futibatinib was rapidly absorbed; median time to peak drug concentration was 1.0 hours. The mean elimination half-life in plasma was 2.3 hours for futibatinib, and 11.9 hours for total radioactivity. Mean recovery of total radioactivity was 70% of the dose, with 64% recovered in feces and 6% in urine. The major excretion route was fecal; negligible levels were excreted as parent futibatinib. Futibatinib was the most abundant plasma component, comprising 59% of circulating radioactivity (CRA). The most abundant metabolites were cysteinylglycine-conjugated futibatinib in plasma (13% CRA) and reduction of desmethyl futibatinib in feces (17% of dose). In human hepatocytes, 14 C-futibatinib metabolites included glucuronide and sulfate of desmethyl futibatinib, whose formation was inhibited by 1-aminobenzotriazole (a pan-cytochrome P450 inhibitor), and glutathione- and cysteine-conjugated futibatinib. These data indicate the primary metabolic pathways of futibatinib are O-desmethylation and glutathione conjugation, with cytochrome P450 enzyme-mediated desmethylation as the main oxidation pathway. 14 C-futibatinib was well tolerated in this Phase 1 study.


Subject(s)
Protein Kinase Inhibitors , Pyrazoles , Humans , Healthy Volunteers , Pyrazoles/adverse effects , Cytochrome P-450 Enzyme System , Metabolome
3.
Clin Pharmacol Drug Dev ; 12(10): 966-978, 2023 10.
Article in English | MEDLINE | ID: mdl-37132707

ABSTRACT

Futibatinib, a selective, irreversible fibroblast growth factor receptor 1-4 inhibitor, is being investigated for tumors harboring FGFR aberrations and was recently approved for the treatment of FGFR2 fusion/rearrangement-positive intrahepatic cholangiocarcinoma. In vitro studies identified cytochrome P450 (CYP) 3A as the major CYP isoform in futibatinib metabolism and indicated that futibatinib is likely a P-glycoprotein (P-gp) substrate and inhibitor. Futibatinib also showed time-dependent inhibition of CYP3A in vitro. Phase I studies investigated the drug-drug interactions of futibatinib with itraconazole (a dual P-gp and strong CYP3A inhibitor), rifampin (a dual P-gp and strong CYP3A inducer), or midazolam (a sensitive CYP3A substrate) in healthy adult participants. Compared with futibatinib alone, coadministration of futibatinib with itraconazole increased futibatinib mean peak plasma concentration and area under the plasma concentration-time curve by 51% and 41%, respectively, and coadministration of futibatinib with rifampin lowered futibatinib mean peak plasma concentration and area under the plasma concentration-time curve by 53% and 64%, respectively. Coadministration of midazolam with futibatinib had no effect on midazolam pharmacokinetics compared with midazolam administered alone. These findings suggest that concomitant use of dual P-gp and strong CYP3A inhibitors/inducers with futibatinib should be avoided, but futibatinib can be concomitantly administered with other drugs metabolized by CYP3A. Drug-drug interaction studies with P-gp-specific substrates and inhibitors are planned.


Subject(s)
Cytochrome P-450 CYP3A , Rifampin , Adult , Humans , Cytochrome P-450 CYP3A/metabolism , Rifampin/pharmacokinetics , Itraconazole/pharmacology , Midazolam/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cytochrome P-450 CYP3A Inducers/pharmacology , Drug Interactions
4.
ACS Med Chem Lett ; 14(4): 396-404, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37077386

ABSTRACT

Deregulating fibroblast growth factor receptor (FGFR) signaling is a promising strategy for cancer therapy. Herein, we report the discovery of compound 5 (TAS-120, futibatinib), a potent and selective covalent inhibitor of FGFR1-4, starting from a unique dual inhibitor of mutant epidermal growth factor receptor and FGFR (compound 1). Compound 5 inhibited all four families of FGFRs in the single-digit nanomolar range and showed high selectivity for over 387 kinases. Binding site analysis revealed that compound 5 covalently bound to the cysteine 491 highly flexible glycine-rich loop region of the FGFR2 adenosine triphosphate pocket. Futibatinib is currently in Phase I-III trials for patients with oncogenically driven FGFR genomic aberrations. In September 2022, the U.S. Food & Drug Administration granted accelerated approval for futibatinib in the treatment of previously treated, unresectable, locally advanced, or metastatic intrahepatic cholangiocarcinoma harboring an FGFR2 gene fusion or other rearrangement.

5.
Clin Pharmacol Drug Dev ; 12(3): 304-313, 2023 03.
Article in English | MEDLINE | ID: mdl-36404525

ABSTRACT

Futibatinib, a fibroblast growth factor receptor (FGFR) 1-4 inhibitor, is being investigated for FGFR-aberrant tumors. A 4-period, crossover, phase 1 thorough QT/QTc study compared effects on Fridericia heart rate-corrected QT (QTcF) interval of single doses of futibatinib 20 and 80 mg (therapeutic and supratherapeutic doses, respectively), placebo, and moxifloxacin (positive control) in healthy subjects. The study objective was to assess the time-matched difference in change from baseline in QTcF (ddQTcF) between futibatinib and placebo. In addition, changes from baseline in QTcF and other electrocardiogram (ECG) parameters, pharmacokinetics, ECG morphology, and safety were assessed. Forty-eight subjects were randomized. ddQTcF upper limits of 2-sided 90%CIs remained <10 milliseconds (clinical threshold) for both futibatinib doses at all time points (range, 2.0-4.5 milliseconds). Assay sensitivity was demonstrated by lower limits of 2-sided 97.5%CIs of the dQTcF difference between moxifloxacin and placebo of >5 milliseconds. Futibatinib exposure increased in a dose-dependent manner, and no significant relationship was detected between plasma futibatinib concentration and ddQTcF. There were no significant effects on heart rate, other ECG parameters, or ECG morphology. No serious adverse events occurred. Futibatinib did not prolong QTcF or affect other cardiac measures at therapeutic or supratherapeutic doses.


Subject(s)
Fluoroquinolones , Heart , Humans , Moxifloxacin/adverse effects , Fluoroquinolones/adverse effects , Healthy Volunteers
6.
Clin Pharmacol Drug Dev ; 12(3): 294-303, 2023 03.
Article in English | MEDLINE | ID: mdl-36382853

ABSTRACT

Futibatinib, an oral, irreversible fibroblast growth factor receptor (FGFR) 1-4 inhibitor, is being evaluated for FGFR-aberrant tumors. Two open-label phase 1 studies evaluated the effects of high-fat, high-calorie food and concomitant proton pump inhibitors (PPIs; lansoprazole) on single-dose futibatinib (20 mg) pharmacokinetics and safety in healthy adults. In the food effect study (N  =  17), subjects received futibatinib under fed and fasted conditions, separated by a 7-day washout. In the PPI study (N  =  20), subjects received futibatinib alone, underwent a 2-day washout, and then received lansoprazole 60 mg once daily for 5 days, with futibatinib also administered on day 5. Under fed versus fasted conditions, futibatinib bioavailability was 11.2% lower (area under the plasma concentration-time curve from time 0 to infinity geometric mean ratio 88.8%; 90% confidence interval, 79.8%-98.9%), and median time to maximum plasma concentration was significantly delayed (4.0 vs 1.5 hours; P < .0001). There were no significant differences in futibatinib exposure between futibatinib plus lansoprazole and futibatinib alone. No serious adverse events occurred in either study. These findings suggest that food and PPIs are unlikely to have clinically meaningful impacts on futibatinib bioavailability. Thus, futibatinib may be used with or without food and concomitantly with acid-reducing agents.


Subject(s)
Food-Drug Interactions , Proton Pump Inhibitors , Adult , Humans , Healthy Volunteers , Lansoprazole/adverse effects
7.
Future Oncol ; 18(30): 3377-3387, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36039910

ABSTRACT

PTEN acts as a potent tumor suppressor within the PI3K/AKT/mTOR pathway. Germline mutations in the PTEN gene are a hallmark of PTEN hamartoma tumor syndrome, which includes Cowden syndrome, where they appear to elevate lifetime risk of cancer. Targeted AKT directed therapy has been proposed as an effective approach in cancer patients having germline PTEN mutations. The mechanism of action, safety and dosing regimen for the novel allosteric AKT inhibitor TAS-117 have been explored in a phase I study in Japan in which activity was observed against certain tumor types. Here we describe the study protocol of an international, two-part phase II study evaluating the safety, tolerability, pharmacokinetics, pharmacodynamics and antitumor activity of TAS-117 in patients with advanced solid tumors harboring germline PTEN-inactivating mutations.


Signaling paths control growth and activities inside cells. Overactivity in these paths can encourage many types of cancers to develop. Tumor suppressor proteins can inhibit cell signals that promote cancer. This protection can be lost if there are errors in any gene coding for a tumor suppressor protein. We are carrying out a clinical study to test TAS-117, a potential new oral medicine, in people who have solid tumors and whose cells have changes in their genes that inactivate a tumor suppressor protein called PTEN. TAS-117 targets part of a signaling path that may be overactive due to loss of PTEN activity. In early research, TAS-117 has shown promising activity against certain tumor types. Our trial will explore if TAS-117 can provide a new treatment for rare forms of cancer where genetic changes have led to a loss of PTEN activity. Clinical Trial Registration: NCT04770246 (ClinicalTrials.gov).


Subject(s)
Hamartoma Syndrome, Multiple , Neoplasms , Humans , Clinical Trials, Phase II as Topic , Germ Cells/metabolism , Germ-Line Mutation , Hamartoma Syndrome, Multiple/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , PTEN Phosphohydrolase/genetics
8.
Commun Biol ; 5(1): 571, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35681099

ABSTRACT

Ribonucleotide reductase (RNR) is composed of two non-identical subunits, R1 and R2, and plays a crucial role in balancing the cellular dNTP pool, establishing it as an attractive cancer target. Herein, we report the discovery of a highly potent and selective small-molecule inhibitor, TAS1553, targeting protein-protein interaction between R1 and R2. TAS1553 is also expected to demonstrate superior selectivity because it does not directly target free radical or a substrate binding site. TAS1553 has shown antiproliferative activity in human cancer cell lines, dramatically reducing the intracellular dATP pool and causing DNA replication stress. Furthermore, we identified SLFN11 as a biomarker that predicts the cytotoxic effect of TAS1553. Oral administration of TAS1553 demonstrated robust antitumor efficacy against both hematological and solid cancer xenograft tumors and also provided a significant survival benefit in an acute myelogenous leukemia model. Our findings strongly support the evaluation of TAS1553 in clinical trials.


Subject(s)
Antineoplastic Agents , Enzyme Inhibitors , Ribonucleotide Reductases , Animals , Antineoplastic Agents/pharmacology , DNA Replication , Enzyme Inhibitors/pharmacology , Humans , Nuclear Proteins/metabolism , Ribonucleotide Reductases/antagonists & inhibitors
9.
Cancer Chemother Pharmacol ; 88(3): 485-497, 2021 09.
Article in English | MEDLINE | ID: mdl-34097100

ABSTRACT

PURPOSE: Trifluridine/tipiracil (FTD/TPI) is approved for advanced colorectal and gastric/gastroesophageal cancer; however, data in patients with renal impairment (RI) are limited. This phase I study evaluated FTD/TPI in patients with advanced solid tumors and varying degrees of RI to develop dosing guidance. METHODS: Patients were enrolled into normal renal function (CrCl ≥ 90 mL/min), mild RI (CrCl 60-89 mL/min), or moderate RI (CrCl 30-59 mL/min) cohorts and administered the recommended FTD/TPI dose (35 mg/m2 twice daily, days 1-5 and 8-12; 28-day cycle). Based on interim pharmacokinetics/safety data, patients with severe RI (CrCl 15-29 mL/min) were enrolled and received FTD/TPI 20 mg/m2 twice daily. RESULTS: Forty-three patients (normal renal function [n = 12]; mild RI [n = 12]; moderate RI [n = 11]; severe RI [n = 8]) were enrolled and treated. At steady state, compared to values in patients with normal renal function, FTD area under the curve (AUC) was not significantly different in patients with RI, but TPI AUC was significantly higher and increased with RI severity. FTD/TPI safety profile was consistent with prior experience, but grade ≥ 3 adverse events (AEs) were more frequent in the RI cohorts (83.3% [mild], 90.9% [moderate], 75.0% [severe], and normal [50.0%]). Hematologic AEs (anemia and neutropenia) were more frequent with RI. Overall, seven patients discontinued because of unrelated, nonhematologic AEs. CONCLUSION: FTD/TPI is safe and tolerable at the recommended 35 mg/m2 dose in patients with mild/moderate RI and at the reduced 20 mg/m2 dose in patients with severe RI. TRIAL REGISTRATION: NCT02301117, registration date: November 21, 2014.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Kidney Diseases/physiopathology , Neoplasms/drug therapy , Pyrrolidines/administration & dosage , Thymine/administration & dosage , Trifluridine/administration & dosage , Adult , Aged , Aged, 80 and over , Anemia/chemically induced , Anemia/epidemiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Cohort Studies , Drug Combinations , Female , Humans , Male , Middle Aged , Neutropenia/chemically induced , Neutropenia/epidemiology , Pyrrolidines/adverse effects , Pyrrolidines/pharmacokinetics , Severity of Illness Index , Thymine/adverse effects , Thymine/pharmacokinetics , Trifluridine/adverse effects , Trifluridine/pharmacokinetics
10.
Invest New Drugs ; 39(5): 1324-1334, 2021 10.
Article in English | MEDLINE | ID: mdl-33774767

ABSTRACT

TAS0728 is an oral covalent binding inhibitor of human epidermal growth factor receptor 2 (HER2). A first-in-human open-label, dose-escalation, phase I study (NCT03410927) was initiated to investigate the safety and dose-limiting toxicity (DLT) and to determine the maximum tolerated dose (MTD) and/or recommended phase II dose of TAS0728 in adults with advanced solid tumors with HER2 or HER3 overexpression, amplification or mutation. In total, 19 patients received TAS0728 at escalating doses from 50 to 200 mg BID for 21-day cycles. Following escalation of the dose to 200 mg BID, a total of two DLTs were observed, both cases of Grade 3 diarrhea (lasting >48 h and not responsive to aggressive antidiarrheal treatment). Following de-escalation of the dose to 150 mg BID, another DLT of Grade 3 diarrhea was observed in one patient. Additionally, at 150 mg BID, one patient had a fatal cardiac arrest after receiving 1 cycle (21 days) of TAS0728. The etiology of the cardiac arrest event was not clear, however causal relationship to TAS0728 could not be excluded due to the temporal association observed. Partial responses were observed in 2 of 14 patients evaluable for TAS0728 treatment response. The study was stopped due to unacceptable toxicity during the dose-escalation as the overall risk-benefit ratio no longer favored the dose level being tested, therefore the MTD was not determined. ClinicalTrials.gov registration number: https://clinicaltrials.gov/ct2/show/NCT03410927 ; registered on January 25, 2018.


Subject(s)
Neoplasms/drug therapy , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Adult , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/genetics , Neoplasms/pathology , Piperidines/administration & dosage , Piperidines/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Purines/administration & dosage , Purines/adverse effects , Receptor, ErbB-2/genetics , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/genetics
11.
Brain Dev ; 40(10): 918-925, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30006121

ABSTRACT

BACKGROUND: Patients with Duchenne muscular dystrophy (DMD) exhibit increased prostaglandin D2 (PGD2) expression in necrotic muscle and increased PGD2 metabolites in their urine. In mouse models, inhibiting PGD2 production suppresses muscle necrosis, suggesting a possible intervention through PGD2-mediated activities. OBJECTIVE: We investigated the involvement of PGD2 and its potential use as a marker of pathological progression in DMD. METHODS: Sixty-one male children with DMD and thirty-five age-matched controls were enrolled in the study. DMD patients were divided into "ambulant" and "non-ambulant" groups, which were further subdivided into "steroid" and "non-steroid" therapy groups. Levels of the PGD2 metabolite tetranor-PGDM (t-PGDM) and creatinine were measured in both spot and 24-hour urine samples, with comparisons between groups made according to geometric mean values. RESULTS: DMD patients had significantly higher levels of creatinine-corrected t-PGDM in spot urine samples as compared with the control group. Additionally, both ambulant and non-ambulant DMD groups had significantly higher levels of t-PGDM as compared with controls, with no significant difference in t-PGDM levels observed between steroid and non-steroid groups. Moreover, total creatinine excretion in 24-hour urine samples was significantly lower in DMD patients as compared with controls, and although DMD patients had lower muscle mass than controls, their overall levels of t-PGDM did not differ significantly from those in the non-ambulant and control groups. CONCLUSION: PGD2 might help explain the progression and symptomatic presentations (e.g., ambulatory difficulty) associated with DMD, suggesting it as a useful pathological marker and use of a selective PGD2 inhibitor as a potential treatment modality.


Subject(s)
Creatinine/urine , Disease Progression , Muscular Dystrophy, Duchenne/urine , Prostaglandin D2/urine , Adolescent , Biomarkers/urine , Child , Child, Preschool , Humans , Male
12.
J Pharm Pharmacol ; 66(12): 1686-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25117829

ABSTRACT

OBJECTIVES: Tegafur (FT), a pro-drug of 5-fluorouracil (5-FU), is a racemate consisting of two enantiomers, R and S-FT. The aim of this study was to clarify interspecies variation in the enantioselective metabolism of FT. METHODS: Plasma concentrations of FT enantiomers were determined in rats, dogs and monkeys following intravenous and oral dosing of the racemate (5 mg/kg). In addition, the enzymatic conversion of FT enantiomers to 5-FU was assayed using hepatic preparations. KEY FINDINGS: Metabolic clearance of R-FT was higher than that of S-FT in rats and monkeys, but S-FT was the preferential substrate for dogs. An inhibition study revealed that cytochrome P450 is primarily responsible for the enantioselective metabolism of FT in rats and dogs. In contrast, in monkeys, thymidine phosphorylase was a determinant of the enantioselectivity in FT metabolism. Although oral bioavailability was not enantioselective, in-vitro and in-vivo kinetic studies suggested that the enantioselectivity in the hepatic intrinsic clearance of FT directly influences the body clearance in all animal species examined. CONCLUSIONS: The interspecies variations were observed in the enantioselective pharmacokinetics of FT, and the in-vivo enantioselectivity could be extrapolated from the in-vitro metabolic activities.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Prodrugs/metabolism , Tegafur/metabolism , Administration, Oral , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/chemistry , Biological Availability , Cytochrome P-450 Enzyme System/metabolism , Dogs , Fluorouracil/blood , Fluorouracil/chemistry , Fluorouracil/metabolism , Injections, Intravenous , Macaca fascicularis , Male , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats, Sprague-Dawley , Species Specificity , Stereoisomerism , Tegafur/blood , Tegafur/chemistry
13.
Drug Metab Dispos ; 42(9): 1485-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25002745

ABSTRACT

Tegafur (FT), a prodrug of 5-fluorouracil, is a chiral molecule, a racemate of R- and S-isomers, and CYP2A6 plays an important role in the enantioselective metabolism of FT in human liver microsomes (R-FT >> S-FT). This study examined the enantioselective metabolism of FT by microsomes prepared from Sf9 cells expressing wild-type CYP2A6 and its variants (CYP2A6*7, *8, *10, and *11) that are highly prevalent in the Asian population. We also investigated the metabolism of coumarin and nicotine, both CYP2A6 probe drugs, in these variants. Enzyme kinetic analyses showed that CYP2A6.7 (I471T) and CYP2A6.10 (I471T and R485L) had markedly lower Vmax values for both enantiomers than wild-type enzyme (CYP2A6.1) and other variant enzymes, whereas Km values were higher in most of the variant enzymes for both enantiomers than CYP2A6.1. The ratios of Vmax and Km values for R-FT to corresponding values for S-FT (R/S ratio) were similar among enzymes, indicating little difference in enantioselectivity among the wild-type and variant enzymes. Similarly, both CYP2A6.7 and CYP2A6.10 had markedly lower Vmax values for coumarin 7-hydroxylase and nicotine C-oxidase activities than CYP2A6.1 and other variant enzymes, whereas Km values were higher in most of the variant enzymes for both activities than CYP2A6.1. In conclusion, the amino acid substitutions in CYP2A6 variants generally resulted in lower affinity for substrates, while Vmax values were selectively reduced in CYP2A6.7 and CYP2A6.10. Consistent R/S ratios among CYP2A6.1 and variant enzymes indicated that the amino acid substitutions had little effect on enantioselectivity in the metabolism of FT.


Subject(s)
Cytochrome P-450 CYP2A6/genetics , Cytochrome P-450 CYP2A6/metabolism , Fluorouracil/metabolism , Polymorphism, Genetic/genetics , Tegafur/metabolism , Cell Line , Coumarins/metabolism , DNA, Complementary/genetics , Escherichia coli/metabolism , Humans , Microsomes, Liver/metabolism , Nicotine/metabolism , Sf9 Cells/metabolism
14.
Pharmacol Res Perspect ; 1(1): e00009, 2013 Oct.
Article in English | MEDLINE | ID: mdl-25505563

ABSTRACT

Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. FT has a chiral center and is a racemate consisting of the enantiomers, R- and S-FT. In the present study, we clarified the enantioselectivity in the conversion of FT to 5-FU and identified human CYP isoforms involved in the metabolism of its enantiomers using human hepatic preparations and recombinant CYP isoforms. Although 5-FU was generated from both FT enantiomers, R-FT was a preferred substrate than S-FT, because of the considerably higher intrinsic clearance for 5-FU formation from R-FT in liver. Eadie-Hofstee plots in microsomes showed that the conversions of R- and S-FT to 5-FU followed biphasic and monophasic kinetics, respectively. Based on the evaluation using cDNA-expressed enzymes, CYP2A6 showed the highest activity for 5-FU formation from R-FT with the K m value similar to that of the high-affinity component in microsomes. Also, CYP2A6 was the most effective catalyst for S-FT. Inhibition studies using CYP-selective inhibitors and anti-CYP antibodies demonstrated that CYP2A6 mainly contributed to the enantioselective metabolism of FT, and were almost in accordance with the relative percentage contribution of each CYP isoform to the metabolism of FT estimated using relative activity factor methods. These results suggest that the enantioselectivity in the bioactivation of FT to 5-FU in humans is mainly due to the large difference of the catalytic activity of CYP2A6 between R- and S-FT.

15.
Drug Metab Dispos ; 38(8): 1267-76, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20463005

ABSTRACT

Tegafur (FT) is a 5-fluorouracil (5-FU) prodrug that has been clinically used for various cancer chemotherapies. The following metabolites of FT were identified in patients: 5-FU, fluoro-beta-alanine, and gamma-butyrolactone (GBL) and its acidic form, gamma-hydroxybutyrate (GHB). GBL/GHB, which is probably generated from the furan ring of FT, inhibits tumor cell angiogenesis, contributing to the antitumor effect of FT-based therapies. In the present study, we identified the metabolites formed from the furan ring of FT by CYP2A6 and thymidine phosphorylase (TPase) using 2,4-dinitrophenylhydrazine derivatization procedures and clarified the metabolic pathway of FT to GBL/GHB. Succinaldehyde (SA) and 4-hydroxybutanal (4-OH-BTL) were produced as the metabolites because of the cleavage of the furan ring of FT during its conversion to 5-FU in cDNA-expressed CYP2A6 and purified TPase, respectively; however, GBL/GHB was hardly detected in cDNA-expressed CYP2A6 and purified TPase. GBL/GHB was formed after human hepatic microsomes or cDNA-expressed CYP2A6 mixed with cytosol were incubated with FT. Furthermore, 4-OH-BTL was converted to GBL/GHB in the microsomes and cytosol. These results suggest that GBL/GHB is generated from FT through the formation of SA and 4-OH-BTL but not directly from FT. Furthermore, the amount of 5-FU and GBL/GHB formed in the hepatic S9 was markedly decreased in the presence of a CYP2A6 inhibitor, suggesting that GBL/GHB may be mainly generated through the CYP2A6-mediated formation of SA.


Subject(s)
4-Butyrolactone/metabolism , Fluorouracil/metabolism , Furans/metabolism , Tegafur/metabolism , Aldehydes/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Butanols/metabolism , Cytochrome P-450 CYP2A6 , Cytosol/metabolism , DNA, Complementary/metabolism , Humans , Liver/enzymology , Microsomes, Liver/enzymology , Phenylhydrazines/metabolism , Sodium Oxybate/metabolism , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/metabolism
16.
Free Radic Biol Med ; 40(11): 2040-6, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16716904

ABSTRACT

Diethylnitrosamine (DEN) is a well-known carcinogenic substance that requires microsomal activation before it can react with DNA to cause mutations and cancer. The aim of this study was to use in vivo spin trapping and spin probe techniques to investigate whether free radicals are generated in rat liver tissue during DEN activation. We used alpha-phenyl-n-tert-butylnitrone (PBN) as the spin trapping agent, which was delivered through an intraperitoneal injection before DEN administration. One hour after DEN administration, multicomponent PBN adducts in the bile were detected, and the intensities were diminished by the cytochrome P450 inhibitor SKF-525A. A computer simulation of the ESR signals revealed the presence of a lipid-derived radical. Using the in vivo spin probe/ESR technique, the signal decay rate of methoxycarbonyl-PROXYL was significantly increased in the DEN-treated group compared with the rate in the vehicle group. The enhanced signal decay rate was restored with PBN and/or SKF-525A pretreatment. These results suggested that lipid-derived free radicals were generated in the liver within 1 h after DEN administration.


Subject(s)
Diethylnitrosamine/pharmacology , Free Radicals , Microsomes, Liver/drug effects , Animals , Bile/metabolism , Electron Spin Resonance Spectroscopy , Lipid Peroxidation , Male , Microsomes, Liver/metabolism , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...