Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Oncol ; 19(7): 2084-90, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11283142

ABSTRACT

PURPOSE: TAS-103 is an inhibitor of both topoisomerase I and II enzymes with broad antitumor activity. It is metabolized to TAS-103-glucuronide (TAS-103-G) predominantly by uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1). We conducted a phase I study to determine the maximum-tolerated dose (MTD) and dose-limiting toxicity (DLT) of TAS-103 when administered on a weekly schedule to patients with advanced cancer. In addition, we evaluated the influence of UGT1A1 genotype on the pharmacokinetics and toxicity of TAS-103. PATIENTS AND METHODS: Thirty-two patients were treated with escalating doses (50 to 200 mg/m(2)) of TAS-103, administered intravenously over 1 hour each week for 3 weeks. Pharmacokinetic analysis was performed at the 130-, 160-, and 200-mg/m(2) dose levels. UGT1A1 genotypes were determined using reverse-transcription polymerase chain reaction techniques. RESULTS: DLT (grade 3 neutropenia) was observed in 5 of 12 patients at 160 mg/m(2) and in 3 of 6 patients at 200 mg/m(2). At 160 mg/m(2), there was a significant correlation between areas under the curve (AUCs) for TAS-103 and TAS-103-G (r = 0.76, P <.05) and an apparent relationship between TAS-103 AUC and D 15 absolute neutrophil count (r = -0.63, P <.05, n = 11, one outlier excluded). UGT1A1 genotype did not influence clearance of TAS-103. CONCLUSION: We recommend a dose of 130 to 160 mg/m(2), or 250 to 300 mg administered using the above weekly schedule for phase II studies. Further studies to characterize the pharmacodynamics and pharmacogenetics of TAS-103 are warranted.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Indenes/pharmacology , Neoplasms/drug therapy , Salvage Therapy/methods , Topoisomerase II Inhibitors , Adult , Aged , Aminoquinolines/administration & dosage , Aminoquinolines/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Female , Genotype , Glucuronosyltransferase/genetics , Humans , Indenes/administration & dosage , Indenes/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Pharmacogenetics
2.
Cancer Chemother Pharmacol ; 47(1): 63-9, 2001.
Article in English | MEDLINE | ID: mdl-11221964

ABSTRACT

PURPOSE: The purpose of this study was to determine the usefulness of various 8-substituted O6-benzylguanine (BG) analogs as modulators of the DNA repair protein. O6-alkylguanine-DNA alkyltransferase (AGT). More specifically, the degree of inactivation of AGT in mouse brain, liver, kidney and tumor by O6-benzyl-8-oxoguanine (8-oxoBG), 8-aza-O6-benzylguanine (8-azaBG), O6-benzyl-8-bromoguanine (8-bromoBG) and O6-benzyl-8-trifluoromethylguanine (8-tfmBG) was compared to inactivation by BG, a modulator in phase II clinical trials. BG is converted rapidly to 8-oxoBG in rodents, monkeys and humans. It was reasoned that 8-substituted analogs of BG would exhibit different pharmacological properties compared to BG which could influence tissue bioavailability and, thus, the extent of AGT inactivation in vivo. We compared the tissue distribution of these agents and AGT activity following administration of the 8-substituted analogs. MATERIALS AND METHODS: At various time points up to 24 h after i.p. administration of the BG analogs, tissues (i.e. brain, liver, kidney), A549 lung tumor xenografts (i.p.) or D456 brain tumor xenografts (i.c.) were harvested from athymic nude mice for AGT analysis. AGT activity was quantified in tissue extracts using a biochemical assay with [3H]methylated DNA as a substrate. In addition, concentrations of BG and 8-oxoBG were determined by HPLC with fluorescence detection in mouse tissues following administration of drug. RESULTS: Each of the 8-substituted analogs of BG demonstrated variable AGT inactivation capabilities that were comparable to or better than those of BG especially in kidney and brain tissues. There was a more pronounced depletion of AGT inactivation in brain and D456 brain tumor xenografts following administration of BG compared to 8-oxoBG that could be explained by a much greater concentration of AGT-inactivating drug (BG plus the metabolite 8-oxoBG for mice treated with BG versus 8-oxoBG for mice treated with 8-oxoBG) present in these tissues. The AUCs for brain, kidney and liver were 3.2, 6.9 and 1 1.8 times greater for BG than for 8-oxoBG. CONCLUSIONS: 8-substituted analogs of BG possess unique AGT-inactivation profiles in vivo that are different from that of BG. The AGT-inhibitory activities of BG and its major metabolite, 8-oxoBG, are related to tissue disposition of both drugs.


Subject(s)
Guanine/analogs & derivatives , Guanine/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Animals , Female , Guanine/metabolism , Mice , Mice, Nude , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Organ Specificity , Structure-Activity Relationship
3.
J Clin Oncol ; 18(20): 3522-8, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11032594

ABSTRACT

PURPOSE: The major mechanism of resistance to alkylnitrosourea therapy involves the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT), which removes chloroethylation or methylation damage from the O(6) position of guanine. O(6)-benzylguanine (O(6)-BG) is an AGT substrate that inhibits AGT by suicide inactivation. We conducted a phase I trial of carmustine (BCNU) plus O(6)-BG to define the toxicity and maximum-tolerated dose (MTD) of BCNU in conjunction with the preadministration of O(6)-BG with recurrent or progressive malignant glioma. PATIENTS AND METHODS: Patients were treated with O(6)-BG at a dose of 100 mg/m(2) followed 1 hour later by BCNU. Cohorts of three to six patients were treated with escalating doses of BCNU, and patients were observed for at least 6 weeks before being considered assessable for toxicity. Plasma samples were collected and analyzed for O(6)-BG, 8-oxo-O(6)-BG, and 8-oxoguanine concentration. RESULTS: Twenty-three patients were treated (22 with glioblastoma multiforme and one with anaplastic astrocytoma). Four dose levels of BCNU (13.5, 27, 40, and 55 mg/m(2)) were evaluated, with the highest dose level being complicated by grade 3 or 4 thrombocytopenia and neutropenia. O(6)-BG rapidly disappeared from plasma (elimination half-life = 0. 54 +/- 0.14 hours) and was converted to a longer-lived metabolite, 8-oxo-O(6)-BG (elimination half-life = 5.6 +/- 2.7 hours) and further to 8-oxoguanine. There was no detectable O(6)-BG 5 hours after the start of the O(6)-BG infusion; however, 8-oxo-O(6)-BG and 8-oxoguanine concentrations were detected 25 hours after O(6)-BG infusion. The mean area under the concentration-time curve (AUC) of 8-oxo-O(6)-BG was 17.5 times greater than the mean AUC for O(6)-BG. CONCLUSION: These results indicate that the MTD of BCNU when given in combination with O(6)-BG at a dose of 100 mg/m(2) is 40 mg/m(2) administered at 6-week intervals. This study provides the foundation for a phase II trial of O(6)-BG plus BCNU in nitrosourea-resistant malignant glioma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Astrocytoma/drug therapy , Central Nervous System Neoplasms/drug therapy , Glioblastoma/drug therapy , Guanine/analogs & derivatives , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/blood , Carmustine/administration & dosage , Carmustine/adverse effects , Carmustine/pharmacokinetics , Central Nervous System Neoplasms/blood , Drug Administration Schedule , Glioblastoma/blood , Guanine/administration & dosage , Guanine/adverse effects , Guanine/blood , Guanine/pharmacokinetics , Humans , Middle Aged , Neoplasm Recurrence, Local/drug therapy
4.
Cancer Chemother Pharmacol ; 46(2): 150-5, 2000.
Article in English | MEDLINE | ID: mdl-10972485

ABSTRACT

PURPOSE: O6-Benzylguanine (BG) is a modulator of the DNA repair protein, O6-alkylguanine-DNA alkyltransferase (AGT). BG is converted in mice, rats and humans to an equally active, yet longer-lived metabolite, O6-benzyl-8-oxoguanine (8-oxo-BG) by CYP1A2, CYP3A4 and aldehyde oxidase. Since intravenous BG is expected to enter phase I development with orally administered anticancer agents such as temozolomide, procarbazine or SarCNU, we determined the bioavailability of orally administered BG, as well as the effect of ketoconazole, a potent intestinal and hepatic CYP3A4 inhibitor, on the disposition of BG. METHODS: Following intravenous or oral administration of BG in PEG-400/saline (40:60) to Sprague-Dawley rats, the pharmacokinetics of BG and 8-oxo-BG were determined. To determine the effect of CYP3A inhibition on disposition, oral BG was coadministered with ketoconazole. RESULTS: The peak plasma concentration (Cmax), time to Cmax (tmax), and bioavailability (F) of oral BG were: 2.3 +/- 0.9 microg/ml, 2.3 +/- 0.6 h, and 65.5% respectively. The AUCs of BG and 8-oxo-BG were 13.1 +/- 4.6 microg x h/ml and 1.7 +/- 0.4 microg x h/ml after oral administration of BG. Coadministration with ketoconazole resulted in an increase in mean absorption time from 2.0 +/- 0.3 h to 6.0 +/- 0.9 h, a shift in tmax to 5 +/- 3.3 h, a decrease in Cmax to 0.96 +/- 0.8 microg/ml, and a decrease in AUC0-inf ratio of 8-oxo-BG:BG from about 0.12 to 0.04 (P < 0.05). The bioavailability of BG was not changed (65.5% vs 56.9%, P= 0.78). CONCLUSIONS: The oral bioavailability of BG is high, warranting consideration of an oral formulation for clinical development. Coadministration of ketoconazole and BG resulted in delayed oral absorption and inhibition of conversion of BG to 8-oxo-BG in the rat model.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Guanine/analogs & derivatives , Ketoconazole/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Area Under Curve , Biological Availability , Cytochrome P-450 CYP1A2/metabolism , Guanine/administration & dosage , Guanine/blood , Guanine/pharmacokinetics , Injections, Intravenous , Ketoconazole/administration & dosage , Male , Mice , Rats , Rats, Sprague-Dawley
5.
Clin Cancer Res ; 6(8): 3025-31, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10955780

ABSTRACT

O6-benzylguanine (BG) is a potent inactivator of the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT) that enhances sensitivity to nitrosoureas in tumor cell lines and tumor-bearing animals. The major objectives of this study were to define the optimal modulatory dose and associated toxicities of benzylguanine administered alone and in combination with carmustine; to define the maximally tolerated dose and associated toxicities of carmustine administered with benzylguanine and to describe the pharmacokinetics of BG in humans and its effects on AGT depletion and recovery in peripheral blood mononuclear cells. Patients with histologically confirmed advanced solid tumors or lymphoma that had failed to respond to standard therapy or for which no standard therapy was available were eligible to participate in this study. Patients initially received BG as a 1-h i.v. infusion without carmustine. After a 14-day washout (ie., without therapy) period, patients received BG as a 1-h i.v. infusion followed, 1 h later, by a 15-min i.v. infusion of carmustine. Cycles of chemotherapy were repeated every 6 weeks. Cohorts of patients received BG doses ranging from 10 to 120 mg/m2 and carmustine doses ranging from 13 to 50 mg/m2. Plasma and urine samples were collected and analyzed for BG, and O6-benzyl-8-oxoguanine concentrations and AGT activity was determined in peripheral blood mononuclear cells. There was no toxicity attributable to BG alone at any dose tested. Bone marrow suppression was the primary and dose-limiting toxicity of BG combined with carmustine and was cumulative in some patients. The neutrophil nadir occurred at a median of day 27, with complete recovery in most patients by day 43. Nonhematological toxicity included fatigue, anorexia, increased bilirubin, and transaminase elevation. Recommended doses for Phase II testing are 120 mg/m2 BG given with carmustine at 40 mg/m2. BG rapidly disappeared from plasma and was converted to a major metabolite, O6-benzyl-8-oxoguanine, which has a 2.4-fold higher maximal concentration and 20-fold higher area under the concentration versus time curve than BG. AGT activity in peripheral blood mononuclear cells was rapidly and completely suppressed at all of the BG doses. The rate of AGT regeneration was more rapid for patients treated with the lowest dose of BG but was similar for BG doses ranging from 20-120 mg/m2. In conclusion, coadministration of BG and carmustine is feasible in cancer patients, but the maximal dose of carmustine that can be safely administered with BG is approximately one-third of the standard clinical dose. Bone marrow suppression, which may be cumulative, is the dose-limiting toxicity of the combination. Prolonged AGT suppression is likely attributable primarily to the effect of O6-benzyl-8-oxoguanine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Guanine/analogs & derivatives , Neoplasms/drug therapy , Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carmustine/administration & dosage , Carmustine/adverse effects , Carmustine/pharmacokinetics , Dose-Response Relationship, Drug , Female , Guanine/administration & dosage , Guanine/adverse effects , Guanine/pharmacokinetics , Humans , Male , Middle Aged , Neoplasms/enzymology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/metabolism
6.
Article in English | MEDLINE | ID: mdl-1350354

ABSTRACT

1. The analgesic effect of quinine and the influence of some dopaminergic agents on it were studied in mice. 2. Quinine (25-130mg/kg, ip) effectively elicited antinociceptive effect in a dose related manner. 3. D-Amphetamine (2.5-4mg/kg, ip), L-dopa (25mg/kg, sc), L-dopa (25mg/kg, sc) plus benserazide (12.5mg/kg, sc), alpha-methyl-p-tyrosine (50mg/kg, ip) plus L-dopa (25mg/kg, sc) and pargyline (50mg/kg, ip) significantly attenuated the antinociceptive effect of quinine (50mg/kg, ip), while DOPS (4mg/kg, ip) did not affect quinine antinociception. 4. Pimozide (4mg/kg, ip), L-sulpiride (40mg/kg, ip), SCH 23390 (0.2mg/kg, sc) and alpha-methyl-p-tyrosine (50mg/kg, ip) effectively potentiated the antinociceptive effects of quinine (50mg/kg, ip). 5. Pimozide (4mg/kg, ip) also antagonised the antagonistic effect of d-amphetamine (4mg/kg, ip) on the antinociceptive effect of quinine (50mg/kg, ip). 6. These data indicate that quinine elicited antinociception dose dependently. Furthermore, the influence of pimozide, L-sulpiride and SCH 23390 on quinine antinociception suggests the involvement of dopaminergic mechanisms.


Subject(s)
Analgesics/pharmacology , Dopamine Agents/pharmacology , Quinine/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Synergism , Male , Mice , Pain Measurement , Quinine/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL