Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 48(3): 235-40, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11592346

ABSTRACT

PURPOSE: Intrathecal methotrexate (MTX) achieves high concentrations in the cerebrospinal fluid (CSF) following intralumbar administration. However, peak ventricular CSF MTX concentrations are highly variable and are < 10% of those achieved with intraventricular dosing. The objectives of this study were to evaluate the effect of intralumbar and intravenous probenecid on ventricular CSF MTX concentrations after intralumbar administration of MTX, and to compare the pharmacokinetics of MTX after intralumbar and intraventricular administration. METHODS: Nonhuman primates (Macaca mulatta) with permanently implanted catheters in the lateral and fourth ventricles received 0.5 mg intraventricular (lateral ventricle) MTX, or 0.5 mg intralumbar MTX with and without intralumbar or intravenous probenecid. Animals were kept prone for 1 h after MTX administration, and ventricular CSF was sampled up to 48 h from a fourth ventricular Ommaya reservoir. MTX concentrations were measured using the dihydrofolate reductase enzyme inhibition assay. Area under the ventricular CSF MTX concentration-time curve (AUC) was used as a measure of MTX exposure. RESULTS: Peak ventricular CSF MTX concentrations and AUCs were highly variable after intralumbar MTX administration. Ventricular CSF MTX AUCs increased by a mean of 3.2-fold after the addition of intralumbar probenecid. Intravenous administration of probenecid did not result in an increase in ventricular CSF MTX AUCs. Asymptomatic pleocytosis was observed in all animals after intralumbar probenecid administration. Ventricular CSF MTX concentrations and AUCs were less variable after intraventricular administration of MTX. CONCLUSION: The administration of intralumbar but not intravenous probenecid increases the ventricular CSF MTX exposure after intralumbar MTX administration.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Cerebrospinal Fluid/metabolism , Methotrexate/pharmacokinetics , Probenecid/pharmacology , Uricosuric Agents/pharmacology , Animals , Antimetabolites, Antineoplastic/cerebrospinal fluid , Area Under Curve , Cerebral Ventricles/metabolism , Drug Administration Routes , Infusions, Intravenous , Injections, Intraventricular , Injections, Spinal , Lumbosacral Region , Macaca mulatta , Methotrexate/cerebrospinal fluid , Toxicity Tests
2.
Cancer Chemother Pharmacol ; 47(1): 41-4, 2001.
Article in English | MEDLINE | ID: mdl-11221960

ABSTRACT

UNLABELLED: Hypericin, a polycyclic aromatic dianthroquinone, is a natural pigment derived from the plant Hypericum perforatum (St John's Wort). The compound has been synthesized and shown to inhibit the growth of malignant glioma cell lines in vitro via inhibition of protein kinase C. Oral hypericin has entered clinical trials in adults with recurrent malignant glioma. PURPOSE: The present study was performed to characterize the plasma pharmacokinetics (PK) and cerebrospinal fluid (CSF) penetration of hypericin in nonhuman primates. METHODS: Hypericin was administered as an intravenous bolus dose of 2 mg/kg (n = 3) or 5 mg/kg (n = 1). Plasma and CSF (ventricular or lumbar) were sampled prior to administration and at frequent intervals for up to 50 h after administration of the drug. Hypericin concentrations in plasma and CSF were determined using a specific reverse-phase HPLC assay. RESULTS: Mean peak plasma concentration of hypericin following the 2 mg/kg dose was 142 +/- 45 microM. Elimination of hypericin from plasma was biexponential, with an average alpha half-life of 2.8 +/- 0.3 h and average terminal half-life of 26 +/- 14 h. CONCLUSIONS: The 2 mg/kg dose in the nonhuman primate was sufficient to maintain plasma concentrations above 10 microM (the in vitro concentration required for growth inhibition of human glioma cell lines) for up to 12 h. No hypericin was detected in the CSF of any animal (lower limit of detection 0.1 microM); the CSF penetration is therefore less than 1%. A severe dose-limiting photosensitivity skin rash was seen at the 5 mg/kg dose level.


Subject(s)
Antineoplastic Agents/blood , Antineoplastic Agents/cerebrospinal fluid , Perylene/analogs & derivatives , Perylene/blood , Perylene/cerebrospinal fluid , Animals , Anthracenes , Antineoplastic Agents/adverse effects , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Half-Life , Injections, Intravenous , Macaca mulatta , Male , Perylene/adverse effects , Time Factors
3.
Clin Cancer Res ; 6(9): 3662-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10999759

ABSTRACT

O6-Benzylguanine (BG) is a potent, specific inactivator of the DNA repair protein, O6-alkylguanine-DNA alkyltransferase, that enhances the sensitivity of tumor cell lines and tumor xenografts to chloroethylnitrosoureas. To search for BG analogues with greater penetration into the cerebrospinal fluid (CSF), we evaluated plasma and CSF pharmacokinetics of BG, 8-aza-O6-benzylguanine (8-azaBG), O6-benzyl-8-bromoguanine (8-BrBG), O6-benzyl-8-oxoguanine (8-oxoBG), O6-benzyl-8-trifluoromethylguanine (8-tfmBG), and O6-benzyl-2'-deoxyguanosine (B2dG) after i.v. administration of 200 mg/m2 of drug through an indwelling Ommaya reservoir in a nonhuman primate model. BG and its analogues were quantified in plasma and CSF using reverse-phase high-performance liquid chromatography assays. The plasma clearances of the four 8-substituted BG analogues were similar (0.04-0.06 l/h/kg), but half-lives ranged from <2 to >24 h. BG was converted to 8-oxoBG, an equally potent O6-alkylguanine-DNA alkyltransferase inactivator, and the elimination of 8-oxoBG was much slower than that of BG. As a result, the plasma area under the curve of 8-oxoBG was 3.5-fold greater than that of BG. B2dG was metabolized to BG and 8-oxoBG, but this pathway accounted for only 20% of B2dG elimination. The CSF penetration percentages (based on the ratio of AUC(CSF): AUCplasma) for BG, 8-azaBG, 8-oxoBG, 8-tfmBG, 8-BrBG, and B2dG were 3.2, 0.18, 4.1, 1.4, <0.3, and 2.0%, respectively. The CSF penetration of BG and its active metabolite 8-oxoBG is greater than the penetration of 8-azaBG, 8-BrBG, 8-tfmBG, and B2dG.


Subject(s)
Guanine/analogs & derivatives , Guanine/pharmacokinetics , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/cerebrospinal fluid , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Enzyme Inhibitors/cerebrospinal fluid , Enzyme Inhibitors/pharmacokinetics , Guanine/blood , Guanine/cerebrospinal fluid , Humans , Macaca mulatta , Male , Microsomes, Liver/metabolism , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Structure-Activity Relationship
4.
J Pharmacol Exp Ther ; 294(3): 894-901, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10945838

ABSTRACT

The novel methotrexate (MTX) rescue agent carboxypeptidase-G(2) (CPDG(2)) converts >98% of plasma MTX to 2, 4-diamino-N(10)-methylpteroic acid (DAMPA) and glutamate in patients with MTX-induced renal failure and delayed MTX excretion. DAMPA is eliminated more rapidly than MTX in these patients, suggesting nonrenal elimination. The pharmacokinetics and metabolism of DAMPA were studied in four nonhuman primates with reverse-phase HPLC with UV, photodiode array detection, and mass spectroscopy. The mean peak plasma DAMPA concentration was 51 microM and the plasma disposition was described by a three-compartment open model with first order elimination. The mean clearance of DAMPA was 1.9 l/kg/h and the mean terminal half-life was 51 min. Forty-six percent of the dose was excreted in the urine as parent compound. Three DAMPA metabolites, hydroxy-DAMPA, DAMPA-glucuronide, and hydroxy-DAMPA-glucuronide, were identified in plasma and urine. These metabolites also were identified in plasma from patients who received CPDG(2) as an MTX rescue agent. The cytotoxicity of DAMPA and its effect on MTX cytotoxicity were assessed in the Molt-4 human leukemic cell line. DAMPA was not cytotoxic and did not significantly alter the cytotoxicity of MTX. In nonhuman primates metabolism of DAMPA is a major route of DAMPA elimination, and metabolism underlies the more rapid elimination of DAMPA versus MTX in patients with MTX-induced renal dysfunction after administration of CPDG(2).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Methotrexate/analogs & derivatives , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Chromatography, High Pressure Liquid , Humans , Injections, Intravenous , Macaca mulatta , Male , Mass Spectrometry , Methotrexate/metabolism , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Tumor Cells, Cultured
5.
Antimicrob Agents Chemother ; 44(4): 1100-1, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10722523

ABSTRACT

Antifungal prophylaxis in cancer patients who are undergoing chemotherapy is associated with prolonged neutropenia. We measured the effect of fluconazole on doxorubicin pharmacokinetics in nonhuman primates to determine if neutropenia is related to a pharmacokinetic interaction that delays the clearance of the chemotherapeutic agent. Fluconazole pretreatment had no effect on doxorubicin pharmacokinetics.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Antifungal Agents/pharmacology , Doxorubicin/pharmacokinetics , Fluconazole/pharmacology , Animals , Area Under Curve , Chromatography, High Pressure Liquid , Drug Interactions , Half-Life , Injections, Intravenous , Macaca mulatta , Male , Spectrometry, Fluorescence
6.
Cancer Chemother Pharmacol ; 45(3): 207-12, 2000.
Article in English | MEDLINE | ID: mdl-10663638

ABSTRACT

PURPOSE: P-glycoprotein (Pgp) is a transmembrane drug efflux pump that is expressed in multidrug-resistant cancer cells and in a variety of normal tissues, including brain capillary endothelial cells which comprise the blood-brain barrier. We studied the effects of the Pgp inhibitor, cyclosporin A (CsA), on the cerebrospinal fluid (CSF) penetration of the Pgp substrate, doxorubicin, in non-human primates. METHODS: The animals received doxorubicin alone (2.0 mg/kg i.v. over 60 min) or doxorubicin (1 mg/kg i.v. over 60 min) and CsA (loading dose 4.0 mg/kg i.v. over 2 h, followed by continuous infusion of 12 mg/kg per day over 48 h). Plasma and CSF were collected over 48 h and the doxorubicin concentration was measured by reverse-phase high-pressure liquid chromatography (HPLC) with fluorescence detection (detection limit 5 nM). A two-compartment model was fitted to the plasma concentration-time data. RESULTS: Pgp was demonstrated to be present in the epithelium of the choroid plexus by immunohistochemical methods, indicating that CSF drug penetration could be used as a surrogate for blood-brain barrier penetration. Steady state whole blood CsA concentrations, which were measured with a fluorescence-polarization immunoassay (TDX) that detects both CsA and its metabolites, ranged from 551-1315 microg/l at 24 h. The clearance of doxorubicin in four animals was reduced by 34%, 38%, 45% and 49% when given with CsA. The doxorubicin concentration in the CSF was <5 nM in all animals, both after doxorubicin alone and doxorubicin with CsA. CONCLUSIONS: The Pgp inhibitor, CsA, at a concentration that alters systemic clearance of doxorubicin, does not appear to significantly increase the CSF penetration of doxorubicin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Antineoplastic Agents/cerebrospinal fluid , Cyclosporine/pharmacology , Doxorubicin/cerebrospinal fluid , ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Blood Vessels/chemistry , Brain/blood supply , Choroid Plexus/chemistry , Choroid Plexus/cytology , Dose-Response Relationship, Drug , Doxorubicin/blood , Doxorubicin/pharmacokinetics , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Epithelial Cells/chemistry , Immunohistochemistry , Infusions, Intravenous , Macaca mulatta , Metabolic Clearance Rate
7.
Cancer Chemother Pharmacol ; 45(3): 259-64, 2000.
Article in English | MEDLINE | ID: mdl-10663645

ABSTRACT

PURPOSE: Intrathecal methotrexate achieves high concentrations in cerebrospinal fluid (CSF), but drug distribution throughout the subarachnoid space after an intralumbar dose is limited. The objective of this study was to quantify methotrexate distribution in CSF after intraventricular and intravenous administration and to identify factors that influence CSF distribution. METHODS: Nonhuman primates (Macaca mulatta) with permanently implanted catheters in the lateral and fourth ventricles received methotrexate by bolus injection (0.5 mg) and infusion (0.05 to 0.5 mg/day over 24 to 168 h) into the lateral ventricle, as well as intravenous infusions. CSF was sampled from the lumbar space, fourth ventricle and the subarachnoid space at the vertex. Methotrexate in CSF and plasma was measured with the dihydrofolate reductase inhibition assay. RESULTS: After bolus intraventricular injection, methotrexate exposure in lumbar CSF ranged from 11% to 69% of that achieved in the fourth ventricle. During continuous intraventricular infusions, methotrexate steady-state concentrations (C(ss)) in lumbar CSF and CSF from the vertex were only 20% to 25% of the ventricular CSF C(ss). The dose, duration of infusion, and infusate volume did not influence drug distribution to the lumbar CSF, but probenicid increased the lumbar to ventricular C(ss) ratio, suggesting the involvement of a probenicid-sensitive transport pump in the efflux of MTX from the CSF. During the intravenous infusions, the ventricular methotrexate C(ss) was lower than the lumbar C(ss) and the C(ss) in CSF from the vertex. CONCLUSION: Methotrexate CSF distribution after intraventricular injection was uneven, and at steady-state CSF methotrexate concentrations were lower at sites that were more distant from the injection site.


Subject(s)
Antimetabolites, Antineoplastic/cerebrospinal fluid , Methotrexate/cerebrospinal fluid , Subarachnoid Space/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacokinetics , Brain/drug effects , Brain/metabolism , Cerebral Ventricles/drug effects , Dose-Response Relationship, Drug , Infusions, Intravenous , Infusions, Parenteral , Injections, Intraventricular , Macaca mulatta , Male , Metabolic Clearance Rate , Methotrexate/pharmacokinetics , Probenecid/pharmacology , Uricosuric Agents/pharmacology
8.
Cancer Chemother Pharmacol ; 44(6): 439-43, 1999.
Article in English | MEDLINE | ID: mdl-10550563

ABSTRACT

PURPOSE: Raltitrexed (Tomudex), ZD1694) is a novel quinazoline folate analog that selectively inhibits thymidylate synthase. Intracellularly, raltitrexed is polyglutamated to its active form which can be retained in cells for prolonged periods. The pharmacokinetics of raltitrexed in plasma and cerebrospinal fluid (CSF) were studied in a nonhuman primate model. METHODS: Animals received 3 mg/m(2) (n = 1), 6 mg/m(2) (n = 3), or 10 mg/m(2) (n = 3) i.v. over 15 min, and frequent plasma samples were obtained over 48 h. CSF samples were drawn from an indwelling 4th ventricular Ommaya reservoir over 48 h. Plasma and CSF raltitrexed concentrations were measured with a novel, sensitive enzyme inhibition assay with a lower limit of quantification of 0.005 microM. A three-compartment pharmacokinetic model was fitted to the raltitrexed plasma concentration-time data. RESULTS: The plasma concentration-time profile of raltitrexed was triexponential with a rapid initial decline and a prolonged terminal elimination phase (t(1/2) > 24 h), which was related to retention of raltitrexed in a deep tissue compartment. At the peak approximately 30% of the administered dose was in the deep tissue compartment, and 24 h after the dosing >20% of the administered dose remained in the body with >99% in the deep tissue compartment. The mean peak (end of infusion) plasma concentrations after the 3, 6, and 10 mg/m(2) doses were 1.5, 2.4 and 4.8 microM, respectively. The clearance of raltitrexed ranged from 110 to 165 ml/min per m(2), and the steady-state volume of distribution exceeded 200 l/m(2). The CSF penetration of raltitrexed was limited (0.6 to 2.0%) and drug could only be detected in the CSF following a 10 mg/m(2 )dose. CONCLUSIONS: The elimination of raltitrexed is triexponential with a prolonged terminal elimination phase. The pharmacokinetic profile is consistent with extensive polyglutamation and intracellular retention of ralitrexed. The three-compartment model presented here may be useful for the analysis of the pharmacokinetics of raltitrexed in humans.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Quinazolines/pharmacokinetics , Thiophenes/pharmacokinetics , Thymidylate Synthase/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/cerebrospinal fluid , Half-Life , Infusions, Intravenous , Macaca mulatta , Male , Metabolic Clearance Rate , Models, Biological , Quinazolines/blood , Quinazolines/cerebrospinal fluid , Sensitivity and Specificity , Thiophenes/blood , Thiophenes/cerebrospinal fluid
9.
Drug Metab Dispos ; 27(10): 1128-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10497138

ABSTRACT

2'-beta-Fluoro-2',3'-dideoxyadenosine (F-ddA, lodenosine) is a nucleoside analog that was rationally designed as a more chemically and enzymatically stable anti-AIDS drug than its parent compound 2', 3'-dideoxyadenosine or didanosine. Plasma and cerebrospinal fluid (CSF) pharmacokinetics of this compound and its major metabolite, 2'-beta-fluoro-2',3'-dideoxyinosine (F-ddI), were studied in three rhesus monkeys after a single 20 mg/kg dose administered as an i.v. push. F-ddA exhibited a mean residence time of 0.17 h in plasma and its plasma concentration time profile appeared to be biexponential. The majority of plasma exposure was from F-ddI, with a mean parent drug area under the curve (AUC) to metabolite AUC ratio of 0.16. CSF levels were low, with a mean CSF AUC to plasma AUC ratio of 0.068, with approximately one-quarter of this exposure in CSF due to unchanged drug. Urinary excretion accounted for half of the drug administered with the majority recovered as the metabolite, F-ddI. In a separate experiment, one monkey received a 20 mg/kg i.v. dose of F-ddI. The total dideoxynucleoside plasma exposure was greater than it was after administration of F-ddA; however, the CSF AUC to plasma AUC ratio was a factor of 4 lower (0.017). Thus, F-ddA central nervous system penetration is at least comparable to that of didanosine, indicating that this experimental drug has potential as an addition to currently approved AIDS therapies.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dideoxyadenosine/analogs & derivatives , Animals , Dideoxyadenosine/blood , Dideoxyadenosine/cerebrospinal fluid , Dideoxyadenosine/pharmacokinetics , Macaca mulatta , Male
10.
Clin Cancer Res ; 4(11): 2891-4, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829757

ABSTRACT

O6-Benzylguanine (O6BG) irreversibly inactivates the single-turnover DNA repair protein alkylguanine-alkyltransferase. Thus, O6BG increases tumor-cell sensitivity to alkylating agents such as carmustine, lomustine, procarbazine, and temozolomide. We investigated the pharmacokinetic behavior of O6BG and O6-benzyl-8-oxoguanine (8-oxo-O6BG) in cerebrospinal fluid (CSF) and plasma after intraventricular administration of O6BG in a nonhuman primate model. In our study, three animals received a single 1-mg dose of O6BG into the lateral ventricle. CSF from the 4th ventricle and plasma samples were collected after administration, and O6BG and 8-oxo-O6BG concentrations were measured by high-performance liquid chromatography. Four additional animals received 1 mg of O6BG via the intralumbar route weekly for 6 weeks to assess the feasibility and toxicity of this route of administration. The peak O6BG CSF concentration was 412+/-86 microM, the t1/2 was 0.52+/-0.02 h, the clearance was 0.22+/-0.01 ml/min, and the area under the concentration-time curve was 319+/-15 microM x h in 4th ventricular CSF. The peak CSF concentration of 8-oxo-O6BG in CSF was 1.9+/-0.4 microM, the t1/2 was 0.76+/-0.03 h, and the area under the concentration-time curve was 5.0+/-1.1 microM x h. Both O6BG and 8-oxo-O6BG were detected in the plasma 0.5-3 h after intraventricular O6BG administration. The plasma peak concentration of O6BG was 0.4 microM at 30 min, and the concentration was <0.1 microM by 3 h. The plasma concentration of 8-oxo-O6BG was 0.2 microM at 30 min and 0.6 microM at 3 h. The animals tolerated the single intraventricular dose and 6 weekly intralumbar doses of O6BG without toxicity. We concluded that intrathecal administration of O6BG is well tolerated in the nonhuman primate and seems to have a substantial pharmacokinetic advantage over systemic administration for meningeal tumors.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Guanine/analogs & derivatives , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Guanine/metabolism , Guanine/pharmacokinetics , Guanine/toxicity , Injections, Spinal , Macaca mulatta , Male
11.
Antimicrob Agents Chemother ; 42(7): 1815-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9661027

ABSTRACT

KNI-272 is a human immunodeficiency virus (HIV) protease inhibitor with potent activity in vitro. We studied the pharmacokinetics of KNI-272 in the plasma and cerebrospinal fluid (CSF) of a nonhuman primate model and after intravenous and oral administration to children with HIV infection. Plasma and CSF were sampled over 24 h after the administration of an intravenous dose of 50 mg of KNI-272 per kg of body weight (approximately 1,000 mg/m2) to three nonhuman primates. The pharmacokinetics of KNI-272 were also studied in 18 children (9 males and 9 females; median age, 9.4 years) enrolled in a phase I trial of four dose levels of KNI-272 (100, 200, 330, and 500 mg/m2 per dose given four times daily). The plasma concentration-time profile of KNI-272 in the nonhuman primate model was characterized by considerable interanimal variability and rapid elimination (clearance, 2.5 liters/h/kg; terminal half-life, 0.54 h). The level of drug exposure achieved in CSF, as measured by the area under the KNI-272 concentration-time curve, was only 1% of that achieved in plasma. The pharmacokinetics of KNI-272 in children were characterized by rapid elimination (clearance, 276 ml/min/m2; terminal half-life, 0.44 h), limited (12%) and apparently saturable bioavailability, and limited distribution (volume of distribution at steady state, 0.11 liter/kg). The concentrations in plasma were maintained above a concentration that is active in vitro for less than half of the 6-h dosing interval. There was no significant increase in CD4 cell counts or decrease in p24 antigen or HIV RNA levels. The pharmacokinetic profile of KNI-272 may limit the drug's efficacy in vivo. It appears that KNI-272 will play a limited role in the treatment of HIV-infected children.


Subject(s)
HIV Infections/metabolism , HIV Protease Inhibitors/pharmacokinetics , Oligopeptides/pharmacokinetics , Administration, Oral , Adolescent , Animals , Child , Child, Preschool , Disease Models, Animal , Female , HIV Infections/drug therapy , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/cerebrospinal fluid , Humans , Infusions, Intravenous , Macaca mulatta , Male , Oligopeptides/administration & dosage , Oligopeptides/blood , Oligopeptides/cerebrospinal fluid
12.
Cancer Chemother Pharmacol ; 41(6): 464-8, 1998.
Article in English | MEDLINE | ID: mdl-9554590

ABSTRACT

PURPOSE: The plasma and cerebrospinal fluid (CSF) pharmacokinetics of the camptothecin analogs, 9-aminocamptothecin (9-AC) and irinotecan, were studied in a nonhuman primate model to determine their CSF penetration. METHODS: 9-AC, 0.2 mg/kg (4 mg/m2) or 0.5 mg/kg (10 mg/m2), was infused intravenously over 15 min and irinotecan, 4.8 mg/kg (96 mg/m2) or 11.6 mg/kg (225 mg/m2), was infused over 30 min. Plasma and CSF samples were obtained at frequent intervals over 24 h. Lactone and total drug forms of 9-AC, irinotecan, and the active metabolite of irinotecan, SN-38, were quantified by reverse-phase HPLC. RESULTS: 9-AC lactone had a clearance (CL) of 2.1 +/- 0.9 l/kg per h, a volume of distribution at steady state (Vd[ss]) of 1.6 +/- 0.7 l/kg and a half-life (t1/2) of 3.2 +/- 0.8 h. The lactone form of 9-AC accounted for 26 +/- 7% of the total drug in plasma. The CSF penetration of 9-AC lactone was limited. CSF 9-AC lactone concentration peaked 30 to 45 min after the dose at 11 to 21 nM (0.5 mg/kg dose), and the ratio of the areas under the CSF and plasma concentration-time curves (AUC(CSF):AUC[P]) was only 3.5 +/- 2.1%. For irinotecan, the CL was 3.4 +/- 0.4 l/kg per h, the Vd(ss) was 7.1 +/- 1.3 l/kg, and the t1/2 was 4.9 +/- 2.2 h. Plasma AUCs of the lactone form of SN-38 were only 2.0% to 2.4% of the AUCs of irinotecan lactone. The lactone form of irinotecan accounted for 26 +/- 5% of the total drug in plasma, and the lactone form of SN-38 accounted for 55 +/- 6% of the total SN-38 in plasma. The AUC(CSF):AUC(P) ratio for irinotecan lactone was 14 +/- 3%. SN-38 lactone and carboxylate could not be measured (< 1.0 nM) in CSF. The AUC(CSF):AUC(P) ratio for SN-38 lactone was estimated to be < or = 8%. CONCLUSION: Despite their structural similarity, the CSF penetration of 9-AC and SN-38 is substantially less than that of topotecan which we previously found to have an AUC(CSF):AUC(P) ratio of 32%.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Macaca mulatta/metabolism , Animals , Antineoplastic Agents, Phytogenic/toxicity , Area Under Curve , Camptothecin/pharmacokinetics , Camptothecin/toxicity , Irinotecan , Male , Nausea/chemically induced , Vomiting/chemically induced
13.
Antimicrob Agents Chemother ; 39(6): 1259-64, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7574512

ABSTRACT

The pharmacokinetics of 2',3'-dideoxyadenosine (ddA), didanosine, 2',3'-dideoxyguanosine (ddG), and 6-halogenated prodrugs of ddG, 6-chloro-ddG and 6-iodo-ddG, in plasma and cerebrospinal fluid (CSF) were studied in a non-human primate model. ddA was rapidly and completely deaminated to didanosine, such that didanosine concentration profiles in plasma and CSF were identical following administration of ddA and didanosine. The mean clearance of didanosine was 0.50 liters/h/kg, the terminal half-life was 1.8 h, and the CSF-to-plasma ratio was 4.8%. The disposition of ddG was similar, with a clearance of 0.70 liters/h/kg and a half-life of 1.7 h. The adenosine deaminase-mediated conversion of the 6-halogenated-ddG prodrugs to ddG was rapid but incomplete (48% for 6-chloro-ddG and 29% for 6-iodo-ddG). The CSF-to-plasma ratios of ddG with equimolar doses of ddG, 6-chloro-ddG, and 6-iodo-ddG were 8.5, 24, and 17%, respectively, but the actual ddG exposures in CSF (area under the CSF concentration-time curve) were comparable for ddG (12.1 microM.h) and the 6-halogenated-ddG prodrugs (18.8 microM.h for 6-chloro-ddG, 9.3 microM.h for 6-iodo-ddG).6-Chloro-ddG was not detectable in plasma or CSF, and the CSF-to-plasma ratio of 6-iodo-ddG was 9.4%, so the higher CSF-to-plasma ratios of ddG with the administration of the 6-halogenated-ddG prodrugs does not appear to be the result of enhanced penetration of the prodrug and subsequent dehalogenation to ddG. The penetration of ddG into CSF exceeds that of didanosine and is enhanced by administration of the 6-halogenated prodrugs, although the mechanism of this enhanced penetration is unclear.


Subject(s)
Dideoxynucleosides/pharmacokinetics , Animals , Antiviral Agents/blood , Antiviral Agents/cerebrospinal fluid , Central Nervous System/metabolism , Didanosine/blood , Didanosine/cerebrospinal fluid , Didanosine/pharmacokinetics , Dideoxyadenosine/blood , Dideoxyadenosine/cerebrospinal fluid , Dideoxyadenosine/pharmacokinetics , Dideoxynucleosides/blood , Dideoxynucleosides/cerebrospinal fluid , Macaca mulatta , Male , Metabolic Clearance Rate , Models, Biological
15.
J Clin Oncol ; 13(1): 177-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7799017

ABSTRACT

PURPOSE: Intralumbar methotrexate is one of the primary therapeutic modalities for the prevention and treatment of meningeal leukemia. However, methotrexate distribution to the ventricles is limited and highly variable following intralumbar dosing, and cytotoxic concentrations of methotrexate are not always achieved or sustained in the ventricular CSF. We used a nonhuman primate model to determine the effect of body position on the caudal distribution of an intralumbar dose of methotrexate. METHODS: Methotrexate (1.0 mg) was administered by intralumbar injection to four animals, which were then immediately placed either in an upright sitting position or in a prone position for 1 hour, then upright. Each animal served as its own control and was studied in each position on at least one occasion. RESULTS: The mean peak ventricular methotrexate concentration was 0.12 mumol/L (range, 0.091 to 0.20) in animals that were immediately placed upright, compared with 2.81 mumol/L (range, 0.21 to 8.9) in animals that remained prone for 1 hour. The mean area under the concentration-versus-time curves (AUC) was 0.51 mumol/L.h (range, 0.26 to 1.1) in the upright animals and 12.0 mumol/L.h (range, 0.9 to 35.4) in the prone animals. CONCLUSION: Maintaining a prone position for 1 hour after an intralumbar dose increased the peak methotrexate concentration and drug exposure in ventricular CSF. CSF drug distribution following intralumbar therapy can be influenced by body position after the injection.


Subject(s)
Methotrexate/administration & dosage , Methotrexate/cerebrospinal fluid , Posture , Animals , Female , Injections, Spinal , Macaca mulatta , Male , Time Factors
16.
Lab Anim Sci ; 44(2): 148-52, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8028278

ABSTRACT

A previously described rhesus monkey model with two intraventricular catheter systems was expanded to provide a means of studying drug concentrations of chemotherapeutic agents such as methotrexate (MTX) in the cerebral subarachnoid cerebrospinal fluid (CSF) following intrathecal drug injection. A continuous intraventricular infusion of MTX was started through the lateral ventricular catheter 44 h before surgical placement of the cerebral subarachnoid catheter to allow for steady-state concentrations to be attained throughout the subarachnoid space. Monkeys were anesthetized intramuscularly with ketamine hydrochloride (7 mg/kg) and xylazine (6 mg/kg) to allow placement of a temporary lumbar catheter for sampling of lumbar CSF Sodium thiopental (2.5%) was given intravenously if needed for intubation and anesthesia was maintained with isoflurane (0.5 to 1.5%) and oxygen during the surgical placement and sampling of the cerebral subarachnoid catheter. A midline incision was made over the frontal bone and a 3/8-inch trephine was used to expose the dura adjacent to the lateral midline. Following puncture of the dura with an 18-gauge spinal needle, the cerebral subarachnoid catheter (0.025-inch [i.d.] x 0.047-inch [o.d.] Silastic tube) was passed into the subarachnoid space for approximately 10 to 17 mm. The CSF from the cerebral subarachnoid catheter was collected by gravity flow Ventricular, lumbar subarachnoid, and cerebral subarachnoid CSF were collected concurrently at 44, 46, and 48 h after the start of MTX infusion. Mean ventricular, lumbar subarachnoid, and cerebral subarachnoid CSF methotrexate concentrations at steady state were 5.8, 1.0, and 1.5 mumol/L, respectively.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Macaca mulatta , Methotrexate/cerebrospinal fluid , Methotrexate/pharmacokinetics , Subarachnoid Space , Animals , Catheterization/methods , Cerebral Ventricles , Infusions, Parenteral , Injections, Intraventricular , Injections, Spinal , Male , Meningeal Neoplasms/drug therapy , Methotrexate/administration & dosage
18.
Invest New Drugs ; 11(2-3): 135-40, 1993.
Article in English | MEDLINE | ID: mdl-7505267

ABSTRACT

Arabinosyl-5-azacytosine (AAC), a new nucleoside antimetabolite, is broadly active in preclinical tumor screening evaluations. To assess the potential for intrathecal use of this drug, we studied the toxicity and pharmacokinetics of intrathecal and intraventricular administration in nonhuman primates. Four adult male rhesus monkeys were given single 10 mg intrathecal (n = 1) or intraventricular (n = 3) doses of AAC to determine its acute toxicity and pharmacokinetic parameters. An additional 3 animals were given four weekly 10 mg intrathecal doses to assess the systemic and neurologic toxicity associated with chronic administration. Disappearance from the cerebrospinal fluid (CSF) was biexponential, and CSF clearance was 0.2 ml/min, which exceeds the rate of CSF bulk flow by 5-fold. The peak CSF concentration and area under the concentration x time curve achieved with the intraventricular administration of 10 mg were one hundred, and fifty fold greater, respectively, than those achieved after an intravenous dose of 200 mg/kg (1500-2400 mg) in prior experiments. No clinically evident neurotoxicity was observed in either the single or the weekly x 4 dose groups. A slight, transient CSF pleocytosis and increased CSF protein was observed. Systemic toxicity was limited to one animal in the weekly x 4 dose group who demonstrated a mild and transient decrease in his peripheral leukocyte count unassociated with a change in his hematocrit or platelet count. These studies in nonhuman primates demonstrate a clear pharmacokinetic advantage for intrathecal vs systemic administration of AAC. This is demonstrated by a 50-fold greater CSF drug exposure with an intrathecal or intraventricular dose 1/200th of that which can be given systemically.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/cerebrospinal fluid , Antineoplastic Agents/toxicity , Azacitidine/cerebrospinal fluid , Azacitidine/toxicity , Animals , Antineoplastic Agents/pharmacokinetics , Azacitidine/pharmacokinetics , Injections, Intraventricular , Injections, Spinal , Macaca mulatta , Male
19.
Cancer Res ; 53(7): 1596-8, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-8453629

ABSTRACT

Prolonged maintenance of a therapeutic drug concentration in the cerebrospinal fluid is required for optimal treatment of leptomeningeal leukemia or carcinomatosis with cell cycle-specific antimetabolites. The pharmacokinetics of 1-beta-D-arabinofuranosylcytosine (ara-C) encapsulated into DepoFoam (Depo/Ara-C) was studied in six rhesus monkeys after intrathecal injection into the lumbar sac. Following a single 2-mg dose, the Depo/Ara-C concentration decreased biexponentially with initial and terminal half-lives of 14.6 and 156 h, respectively. The free drug concentration remained above the reported minimal cytotoxic level of 0.1 micrograms/ml (0.4 microM) for more than 672 h (28 days). In contrast, the half-life of ara-C following an intralumbar bolus dose of unencapsulated drug in a single animal was 0.74 h. A single intrathecal injection of Depo/Ara-C can maintain a therapeutic drug concentration in the cerebrospinal fluid for a very prolonged period.


Subject(s)
Cytarabine/cerebrospinal fluid , Animals , Cytarabine/administration & dosage , Drug Carriers , Drug Compounding , Half-Life , Injections, Spinal , Lipid Bilayers , Macaca mulatta , Male
20.
Cancer Chemother Pharmacol ; 32(4): 310-4, 1993.
Article in English | MEDLINE | ID: mdl-8324873

ABSTRACT

The pharmacokinetics of the polyethylene glycol-conjugated form of the enzyme L-asparaginase and the depletion of L-asparagine from the plasma and cerebrospinal fluid (CSF) following an i.m. dose of 2500 IU/m2 PEG-L-asparaginase was studied in rhesus monkeys. PEG-L-asparaginase activity in plasma was detectable by 1 h after injection and maintained a plateau of approximately 4 IU/ml for more than 5 days. Subsequent elimination from plasma was monoexponential with a half-life of 6 +/- 1 days. Plasma L-asparagine concentrations fell from pretreatment levels of 14-47 microM to < 2 microM by 24 h after injection in all animals and remained undetectable for the duration of the 25-day observation period in four of six animals. In two animals, plasma L-asparagine became detectable when the PEG-L-asparaginase plasma concentration dropped below 0.1 IU/ml. Pretreatment CSF L-asparagine levels ranged from 4.7 to 13.6 microM and fell to < 0.25 microM by 48 h in five of six animals. CSF L-asparagine concentrations remained below 0.25 microM for 10-14 days in four animals. One animal had detectable CSF L-asparagine concentrations within 24 h and another had detectable concentrations within 1 week of drug administration despite a plasma PEG-L-asparaginase activity profile that did not differ from that of the other animals. These observations may be useful in the design of clinical trials with PEG-L-asparaginase in which correlations among PEG-L-asparaginase pharmacokinetics, depletion of L-asparagine, and clinical outcome should be sought.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Asparaginase/pharmacokinetics , Asparagine/blood , Asparagine/cerebrospinal fluid , Polyethylene Glycols/pharmacokinetics , Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Asparaginase/administration & dosage , Chromatography, High Pressure Liquid , Enzymes, Immobilized , Injections, Intramuscular , Isoenzymes , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Polyethylene Glycols/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL