Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurooncol ; 126(2): 225-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26518542

ABSTRACT

Chemotherapies active in preclinical studies frequently fail in the clinic due to lack of efficacy, which limits progress for rare cancers since only small numbers of patients are available for clinical trials. Thus, a preclinical drug development pipeline was developed to prioritize potentially active regimens for pediatric brain tumors spanning from in vitro drug screening, through intracranial and intra-tumoral pharmacokinetics to in vivo efficacy studies. Here, as an example of the pipeline, data are presented for the combination of 5-fluoro-2'-deoxycytidine and tetrahydrouridine in three pediatric brain tumor models. The in vitro activity of nine novel therapies was tested against tumor spheres derived from faithful mouse models of Group 3 medulloblastoma, ependymoma, and choroid plexus carcinoma. Agents with the greatest in vitro potency were then subjected to a comprehensive series of in vivo pharmacokinetic (PK) and pharmacodynamic (PD) studies culminating in preclinical efficacy trials in mice harboring brain tumors. The nucleoside analog 5-fluoro-2'-deoxycytidine (FdCyd) markedly reduced the proliferation in vitro of all three brain tumor cell types at nanomolar concentrations. Detailed intracranial PK studies confirmed that systemically administered FdCyd exceeded concentrations in brain tumors necessary to inhibit tumor cell proliferation, but no tumor displayed a significant in vivo therapeutic response. Despite promising in vitro activity and in vivo PK properties, FdCyd is unlikely to be an effective treatment of pediatric brain tumors, and therefore was deprioritized for the clinic. Our comprehensive and integrated preclinical drug development pipeline should reduce the attrition of drugs in clinical trials.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain/drug effects , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Tetrahydrouridine/administration & dosage , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Mice , Mice, Nude , Tetrahydrouridine/blood , Tetrahydrouridine/pharmacokinetics , Tetrahydrouridine/therapeutic use
2.
Cancer Chemother Pharmacol ; 76(4): 803-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26321472

ABSTRACT

INTRODUCTION: 5-Fluoro-2'-deoxycytidine (FdCyd; NSC48006), a fluoropyrimidine nucleoside inhibitor of DNA methylation, is degraded by cytidine deaminase (CD). Pharmacokinetic evaluation was carried out in cynomolgus monkeys in support of an ongoing phase I study of the PO combination of FdCyd and the CD inhibitor tetrahydrouridine (THU; NSC112907). METHODS: Animals were dosed intravenously (IV) or per os (PO). Plasma samples were analyzed by LC-MS/MS for FdCyd, metabolites, and THU. Clinical chemistry and hematology were performed at various times after dosing. A pilot pharmacokinetic study was performed in humans to assess FdCyd bioavailability. RESULTS: After IV FdCyd and THU administration, FdCyd C(max) and AUC increased with dose. FdCyd half-life ranged between 22 and 56 min, and clearance was approximately 15 mL/min/kg. FdCyd PO bioavailability after THU ranged between 9 and 25 % and increased with increasing THU dose. PO bioavailability of THU was less than 5 %, but did result in plasma concentrations associated with inhibition of its target CD. Human pilot studies showed comparable bioavailability for FdCyd (10 %) and THU (4.1 %). CONCLUSION: Administration of THU with FdCyd increased the exposure to FdCyd and improved PO FdCyd bioavailability from <1 to 24 %. Concentrations of THU and FdCyd achieved after PO administration are associated with CD inhibition and hypomethylation, respectively. The schedule currently studied in phase I studies of PO FdCyd and THU is daily times three at the beginning of the first and second weeks of a 28-day cycle.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Cytidine Deaminase/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Enzyme Inhibitors/pharmacokinetics , Tetrahydrouridine/pharmacokinetics , Administration, Oral , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Biological Availability , Biotransformation , Cohort Studies , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Combinations , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/blood , Female , Half-Life , Humans , Infusions, Intravenous , Macaca fascicularis , Male , Metabolic Clearance Rate , Pilot Projects , Tetrahydrouridine/administration & dosage , Tetrahydrouridine/blood
3.
Cancer Chemother Pharmacol ; 67(2): 421-30, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20443002

ABSTRACT

PURPOSE: Cytidine drugs, such as gemcitabine, undergo rapid catabolism and inactivation by cytidine deaminase (CD). 3,4,5,6-tetrahydrouridine (THU), a potent CD inhibitor, has been applied preclinically and clinically as a modulator of cytidine analogue metabolism. However, THU is only 20% orally bioavailable, which limits its preclinical evaluation and clinical use. Therefore, we characterized THU pharmacokinetics after the administration to mice of the more lipophilic pro-drug triacetyl-THU (taTHU). METHODS: Mice were dosed with 150 mg/kg taTHU i.v. or p.o. Plasma and urine THU concentrations were quantitated with a validated LC-MS/MS assay. Plasma and urine pharmacokinetic parameters were calculated non-compartmentally and compartmentally. RESULTS: taTHU did not inhibit CD. THU, after 150 mg/kg taTHU i.v., had a 235-min terminal half-life and produced plasma THU concentrations >1 µg/mL, the concentration shown to inhibit CD, for 10 h. Renal excretion accounted for 40-55% of the i.v. taTHU dose, 6-12% of the p.o. taTHU dose. A two-compartment model of taTHU generating THU fitted the i.v. taTHU data best. taTHU, at 150 mg/kg p.o., produced a concentration versus time profile with a plateau of approximately 10 µg/mL from 0.5-2 h, followed by a decline with a 122-min half-life. Approximately 68% of i.v. taTHU is converted to THU. Approximately 30% of p.o. taTHU reaches the systemic circulation as THU. CONCLUSIONS: The availability of THU after p.o. taTHU is 30%, when compared to the 20% achieved with p.o. THU. These data will support the clinical studies of taTHU.


Subject(s)
Prodrugs/pharmacokinetics , Tetrahydrouridine/analogs & derivatives , Tetrahydrouridine/pharmacokinetics , Administration, Oral , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/urine , Area Under Curve , Biocatalysis/drug effects , Biological Availability , Blood/metabolism , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Humans , Injections, Intravenous , Male , Mice , Mice, Inbred Strains , Models, Biological , Prodrugs/metabolism , Prodrugs/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Tetrahydrouridine/blood , Tetrahydrouridine/metabolism , Tetrahydrouridine/pharmacology , Tetrahydrouridine/urine , Urine/chemistry , Gemcitabine
4.
Article in English | MEDLINE | ID: mdl-18562256

ABSTRACT

The 5FU prodrug capecitabine undergoes a 3-step enzymatic conversion, including the conversion of 5'DFRC into 5'DFUR by cytidine deaminase (CDA). The presence of CDA activity in blood led us to analyze the possible ex vivo conversion of 5'DFCR into 5'DFUR in blood samples. We thus examined the impact of the addition of a CDA inhibitor (tetrahydrouridine (THU) 1 microM final) in blood. Blood samples from 3 healthy volunteers were taken on tubes containing or not THU. Blood was spiked with 5'DFCR (20 microM final) (T0) and was maintained at room temperature for 2 h. Plasma concentrations of 5'DFRC and 5'DFUR were analyzed with an optimized HPLC assay. In the absence of THU, 5'DFUR was detectable as early as T0. The percent of 5'DFUR produced relative to 5'DFCR increased over time, up to 7.7 % at 2h. In contrast, the presence of THU totally prevents the formation of 5'DFUR. The impact of THU for preventing the conversion of 5'DFCR was confirmed by the analysis of blood samples from 2 capecitabine-treated patients. Addition of THU in the sampling-tube before the introduction of blood is thus strongly recommended in order to guarantee accurate conditions for reliable measurement of capecitabine metabolites in plasma, and thus faithful pharmacokinetic data.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Fluorouracil/analogs & derivatives , Tetrahydrouridine/pharmacology , Capecitabine , Chromatography, High Pressure Liquid/methods , Colorectal Neoplasms/drug therapy , Cytidine Deaminase/blood , Cytidine Deaminase/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Enzyme Inhibitors/blood , Fluorouracil/administration & dosage , Fluorouracil/blood , Fluorouracil/metabolism , Fluorouracil/pharmacokinetics , Humans , Metabolic Networks and Pathways/drug effects , Prodrugs/administration & dosage , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Tetrahydrouridine/blood
5.
Cancer Chemother Pharmacol ; 62(3): 457-64, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18008070

ABSTRACT

Cytidine analogues such as cytosine arabinoside, gemcitabine, decitabine, 5-azacytidine, 5-fluoro-2'-deoxycytidine and 5-chloro-2'-deoxycytidine undergo rapid catabolism by cytidine deaminase (CD). 3,4,5,6-tetrahydrouridine (THU) is a potent CD inhibitor that has been applied preclinically and clinically as a modulator of cytidine analogue metabolism. However, THU pharmacokinetics has not been fully characterized, which has impaired the optimal preclinical evaluation and clinical use of THU. Therefore, we characterized the THU pharmacokinetics and bioavailability in mice. Mice were dosed with THU iv (100 mg/kg) or po (30, 100, or 300 mg/kg). Plasma and urine THU concentrations were quantitated with a validated LC-MS/MS assay. Plasma pharmacokinetic parameters were calculated compartmentally and non-compartmentally. THU, at 100 mg/kg iv had a 73 min terminal half-life and produced plasma THU concentrations >1 microg/ml, the concentration shown to effectively block deamination, for 4 h. Clearance was 9.1 ml/min/kg, and the distribution volume was 0.95 l/kg. Renal excretion accounted for 36-55% of the THU dose. A three-compartment model fit the iv THU data best. THU, at 100 mg/kg po, produced a concentration versus time profile with a plateau of approximately 10 mug/ml from 0.5-3 h, followed by a decline with an 85 min half-life. The oral bioavailability of THU was approximately 20%. The 20% oral bioavailability of THU is sufficient to produce and sustain, for several hours, plasma concentrations that inhibit CD. This suggests the feasibility of using THU to decrease elimination and first-pass metabolism of cytidine analogues by CD. THU pharmacokinetics are now being evaluated in humans.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Enzyme Inhibitors , Tetrahydrouridine , Administration, Oral , Animals , Biological Availability , Chromatography, Liquid , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Half-Life , Injections, Intravenous , Male , Mice , Mice, Inbred Strains , Protein Binding , Tandem Mass Spectrometry , Tetrahydrouridine/blood , Tetrahydrouridine/pharmacokinetics , Tetrahydrouridine/pharmacology
6.
Cancer Chemother Pharmacol ; 62(2): 363-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17899082

ABSTRACT

PURPOSE: Although the DNA methyltransferase inhibitor 5-fluoro-2'-deoxycytidine (FdCyd), is being evaluated clinically, it must be combined with the cytidine deaminase inhibitor tetrahydrouridine (THU) to prevent rapid metabolism of FdCyd to the pharmacologically active, yet unwanted, metabolites 5-fluoro-2'-deoxyuridine (FdUrd), 5-fluorouracil (FU), and 5-fluorouridine (FUrd). We assessed plasma concentrations of FdCyd and metabolites in patients receiving FdCyd and THU. METHODS: We validated an LC-MS/MS assay, developed for a preclinical study, to quantitate FdCyd and metabolites in human plasma. Patients were treated with five daily, 3-h infusions of FdCyd at doses of 5-80 mg/m(2) with 350 mg/m(2) THU. Plasma was obtained during, and before the end of infusions on days 1 and 5. RESULTS: The lower limits of quantitation for FU, FdUrd, FUrd, FC and FdCyd were 1, 1.5, 10, 3, and 10 ng/ml, respectively. Plasma FdCyd increased with dose, from 19-96 ng/ml at 5 mg/m(2) to 1,600-1,728 ng/ml at 80 mg/m(2). FdUrd was undetectable in patients treated with FdCyd doses <20 mg/m(2), and increased from 2.3 ng/ml at 20 mg/m(2) to 3.5-5.7 ng/ml at 80 mg/m(2). FU increased from 1.2-5.5 ng/ml at 5 mg/m(2) to 6.0-12 ng/ml at 80 mg/m(2). CONCLUSIONS: By co-administering FdCyd with THU, FdCyd plasma concentrations were achieved that are known to inhibit DNA methylation in vitro. The accompanying plasma FU and FdUrd concentrations are <10% those observed after therapeutic infusions of FU or FdUrd, while FdCyd levels are well above those required to inhibit methylation in vitro. Therefore, inhibition of DNA methylation with FdCyd and THU appears feasible.


Subject(s)
Antineoplastic Agents , DNA Modification Methylases/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Tetrahydrouridine , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Chromatography, High Pressure Liquid , Deoxycytidine/administration & dosage , Deoxycytidine/blood , Deoxycytidine/metabolism , Dose-Response Relationship, Drug , Floxuridine/blood , Fluorouracil/blood , Humans , Infusions, Intravenous , Reproducibility of Results , Tandem Mass Spectrometry , Tetrahydrouridine/administration & dosage , Tetrahydrouridine/blood , Tetrahydrouridine/metabolism , Uridine/analogs & derivatives , Uridine/blood
7.
Rapid Commun Mass Spectrom ; 21(13): 1991-7, 2007.
Article in English | MEDLINE | ID: mdl-17526067

ABSTRACT

A number of anticancer drugs are cytidine analogues that undergo metabolic deactivation catalyzed by cytidine deaminase (CD). 3,4,5,6-Tetrahydrouridine (THU) is a potent inhibitor of CD, by acting as a transition-state analogue of its natural substrate cytidine. However, to date its pharmacokinetic properties have not been fully characterized, which has impaired its optimal preclinical evaluation and clinical use. We report a liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay for the sensitive, accurate and precise quantitation of THU in mouse plasma. Validation was performed according to FDA guidelines. The assay employed deuterated THU as the internal standard and an acetonitrile protein precipitation step. Separation, based on hydrophilic interaction chromatography, was achieved with an amino column and an isocratic mobile phase of 0.1% formic acid in acetonitrile and water followed by a wash. Chromatographic separation was followed by positive-mode electrospray ionization MS/MS detection in the multiple reaction monitoring (MRM) mode. The assay was accurate (92.5-109.9%) and precise (2.1-9.0%) in the concentration range of 0.2-50 microg/mL. Recovery from plasma was near-complete (92.9-119.3%) and ion suppression was negligible (-17.5 to -0.2%). Plasma freeze/thaw stability (93.1-102.1%), stability for 3 months at -80 degrees C (99.5-110.9%), and stability for 4 h at room temperature (92.1-102.4%) were all in order. This assay is currently being used to quantitate THU in ongoing pharmacokinetic studies. In addition, the assay is expected to be a useful tool in any future studies involving co-administration of THU with cytidine analogues.


Subject(s)
Chromatography, Liquid/methods , Cytidine Deaminase/antagonists & inhibitors , Enzyme Inhibitors/blood , Tandem Mass Spectrometry/methods , Tetrahydrouridine/blood , Animals , Drug Stability , Enzyme Inhibitors/chemistry , Freezing , Male , Mice , Mice, Inbred Strains , Molecular Structure , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Specific Pathogen-Free Organisms , Temperature , Tetrahydrouridine/chemistry , Time Factors
8.
Cancer Invest ; 5(4): 293-9, 1987.
Article in English | MEDLINE | ID: mdl-3664332

ABSTRACT

Deamination of cytosine arabinoside (ara-C) by cytidine deaminase (Cyt DA) is the main mode of the inactivation of this drug in vivo. Tetrahydrouridine (THU) and the deamination product, uracil arabinoside (ara-U) are potent inhibitors of Cyt DA. We investigated whether ara-U or THU pretreatments can protect ara-C from excessive deamination in tumor- (L1210) bearing mice. In order to determine this, plasma concentrations of ara-C, ara-U, and the intracellular levels of ara-CTP, the active anabolite of ara-C, were assayed. The control peak plasma levels of ara-C and ara-U were 3.3 and 0.78 mM and they were eliminated with a half life (t 1/2) of 1.26 and 1.43 hours, respectively. One hour pretreatment with a nontoxic dose of ara-U (single dose of 300 mg/kg intraperitoneally), resulted in increased ara-C levels by 5.9-fold, while ara-U increased 14.3 fold in comparison with controls. A 24-hour (every 8 hours) pretreatment with ara-U increased ara-C plasma levels by 3.0-fold and it was eliminated with a t 1/2 of 1.21 hours. One hour pretreatment with THU (single dose 25 mg/kg intraperitoneally) enhanced ara-C plasma levels by 5.3-fold. In control L1210/0 acid extracts, ara-CTP peaked at 2 hours and reached 2030 +/- 85 microM; ara-CTP was eliminated with a t 1/2 1.47 hours. The ara-CTP cellular concentrations after 1- and 24-hour pretreatments were 1875 +/- 534 and 2624 +/- 429 microM at 4 hours; the t 1/2 were 2.20 and 1.44 hours, respectively. The THU pretreatment resulted in a peak concentration of ara-CTP of 2208 +/- 366 microM at 2 hours and was eliminated with a t 1/2 of 2.54 hours. We concluded that all pretreatments increased both the peak plasma ara-C concentrations and the area under the plasma concentration-time curve (AUC). One hour ara-U pretreatment did not enhance the peak ara-CTP cellular concentration, but did extend the t 1/2. The 24-hour ara-U and the 1-hour THU pretreatments increased, to some extent, the cellular ara-CTP concentrations, but these differences were not statistically significant. THU pretreatment increased the time the peak occurred and the t 1/2 of ara-CTP. The area under the cellular ara-CTP concentration-time curve (AUC) in L1210 cells was either the same or increased by a small amount after the pretreatments.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Arabinofuranosyluracil/pharmacology , Cytarabine/metabolism , Leukemia L1210/metabolism , Tetrahydrouridine/pharmacology , Uridine/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arabinofuranosyluracil/administration & dosage , Arabinofuranosyluracil/blood , Cytarabine/administration & dosage , Deamination , Leukemia L1210/blood , Leukemia L1210/drug therapy , Male , Mice , Tetrahydrouridine/administration & dosage , Tetrahydrouridine/blood , Time Factors
9.
Cancer Treat Rep ; 60(11): 1627-31, 1976 Nov.
Article in English | MEDLINE | ID: mdl-828522

ABSTRACT

2-14C-tetrahydrouridine was prepared and used to determine the serum levels and excretion of tetrahydrouridine by varous experimental animals. In mice injected ip with the agent (50 mg/kg), the serum level of tetrahydrouridine was maximum (76 mug/ml) at 15 minutes. For rats injected with the same dose, the tetrahydrouridine content of serum was greatest (58 mug/ml) at 30 minutes. Within 3 hours, the serum content of the drug in both mice and rats fell to less than 10% of the maximum. The kidneys of mice selectively accumulated tetrahydrouridine; the concentration rose to 275 mug/g at 1 hour after injection. Nearly all of the dose was excreted unchanged in the urine of mice and rats in 24 hours. For a dog and a monkey given an iv dose (50 mg/kg) of tetrahydrouridine, serum levels of the agent were 210 and 200 mug/ml, respectively, at 5 minutes. The apparent half-lives for the initial phase of disappearance were 18 and 20 minutes and those for the later phase were 65 and 70 minutes, respectively. In 24 hours the dog and monkey excreted most of the dose as unchanged tetrahydrouridine. No metabolites were detected in the biologic samples from either species.


Subject(s)
Tetrahydrouridine/metabolism , Uridine/analogs & derivatives , Animals , Dogs , Female , Haplorhini , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Male , Mice , Rats , Spleen/metabolism , Tetrahydrouridine/blood , Tetrahydrouridine/urine , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...