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.
Antimicrob Agents Chemother ; 36(2): 353-60, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1318679

ABSTRACT

The metabolism of 6-dimethylaminopurine arabinoside (ara-DMAP), a potent inhibitor of varicella-zoster virus replication in vitro, was studied in rats and cynomolgus monkeys. Rats dosed intraperitoneally or orally with ara-DMAP excreted unchanged ara-DMAP and one major metabolite, 6-methylaminopurine arabinoside (ara-MAP), in the urine. They also excreted allantoin and small amounts (less than 4% of the dose each) of hypoxanthine arabinoside (ara-H) and adenine arabinoside (ara-A). The relative amount of each urinary metabolite excreted remained fairly constant for intraperitoneal ara-DMAP doses of 0.3 to 50 mg/kg of body weight. Rats pretreated with an inhibitor of microsomal N-demethylation, SKF-525-A, excreted more unchanged ara-DMAP and much less ara-MAP than did rats given ara-DMAP alone. Rats pretreated with the adenosine deaminase inhibitor deoxycoformycin excreted more ara-MAP and much less ara-H and allantoin. ara-MAP was shown to be a competitive alternative substrate inhibitor of adenosine deaminase (Ki = 16 microM). Rats given ara-DMAP intravenously rapidly converted it to ara-MAP and purine metabolism end products; however, ara-A generated from ara-DMAP had a half-life that was four times longer than that of ara-A given intravenously. In contrast to rats, cynomolgus monkeys dosed intravenously with ara-DMAP formed ara-H as the major plasma and urinary end metabolite. Rat liver microsomes demethylated ara-DMAP much more rapidly than human liver microsomes did. ara-DMAP is initially N-demethylated by microsomal enzymes to form ara-MAP. This metabolite is further metabolized by either adenosine deaminase, which removes methylamine to form ara-H, or by microsomal enzymes, which remove the second methyl group to form ara-A.


Subject(s)
Antiviral Agents/metabolism , Herpesvirus 3, Human/drug effects , Vidarabine/analogs & derivatives , Adenosine Deaminase Inhibitors , Administration, Oral , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Arabinonucleosides/metabolism , Arabinonucleosides/urine , Chromatography, High Pressure Liquid , Half-Life , Herpesvirus 3, Human/physiology , Humans , In Vitro Techniques , Injections, Intraperitoneal , Injections, Intravenous , Macaca fascicularis , Microsomes, Liver/metabolism , Proadifen/pharmacology , Rats , Rats, Inbred Strains , Species Specificity , Vidarabine/metabolism , Vidarabine/pharmacokinetics , Vidarabine/pharmacology , Vidarabine/urine , Virus Replication/drug effects
2.
Cancer Res ; 43(7): 3451-8, 1983 Jul.
Article in English | MEDLINE | ID: mdl-6601986

ABSTRACT

A patient with refractory T-cell acute lymphoblastic leukemia was treated with eight courses of the adenosine deaminase inhibitor, 2'-deoxycoformycin (dCF), over a 5-month period. After developing resistance to dCF, he responded to treatment with the combination of dCF and 9-beta-D-arabinofuranosyladenine (ara-A). We monitored the levels in plasma and urine of adenosine, 2'-deoxyadenosine, and ara-A as well as the accumulation of their nucleotide derivatives in erythrocytes and circulating lymphoblasts. We also monitored the activities of adenosine deaminase and S-adenosylhomocysteine (AdoHcy) hydrolase and the concentrations of AdoHcy and S-adenosylmethionine in lymphoblasts. Production of 2'-deoxyadenosine was related to both the duration of dCF infusion and the magnitude of cytolysis that occurred during treatment: much more 2'-deoxyadenosine was produced by dCF infusion when disease was active than by the same infusion given during remission. Resistance to dCF was associated with a decrease of greater than 90% in the amount of deoxyadenosine 5'-triphosphate accumulated by circulating lymphoblasts. Infusion of dCF resulted in increases of up to 20-fold in the concentration of AdoHcy in circulating lymphoblasts, causing a decrease in the S-adenosylmethionine:AdoHcy ratio (the "methylation index") from a pretreatment value of greater than 40:1 to less than 4:1. This ratio decreased to 2.5:1 during combined treatment with dCF and ara-A, which caused nearly complete inactivation of lymphoblast AdoHcy hydrolase. Decline in the methylation index was accompanied by inhibition of the methylation of newly synthesized lymphoblast RNA. Impaired ability to catabolize AdoHcy may have contributed to the cytolytic responses to dCF and ara-A, as well as to hepatic and central nervous system toxicity associated with their combined use.


Subject(s)
Coformycin/administration & dosage , Homocysteine/analogs & derivatives , Leukemia, Lymphoid/drug therapy , Ribonucleosides/administration & dosage , S-Adenosylhomocysteine/metabolism , Vidarabine/administration & dosage , Acute Disease , Adenosine/blood , Adenosine/urine , Adult , Coformycin/analogs & derivatives , Deoxyadenosines/blood , Deoxyadenosines/urine , Drug Resistance , Drug Therapy, Combination , Erythrocytes/analysis , Humans , Leukemia, Lymphoid/blood , Leukemia, Lymphoid/urine , Lymphocytes/analysis , Lymphocytes/enzymology , Male , Pentostatin , Time Factors , Vidarabine/blood , Vidarabine/urine
4.
Clin Pharmacol Ther ; 27(5): 690-6, 1980 May.
Article in English | MEDLINE | ID: mdl-7371366

ABSTRACT

Vidarabine (Vira-A) was given intravenously for 5 days to 5 immunosuppressed patients with herpes zoster. The daily dose, 10 mg/kg, was given by slow infusion over 12 hr. Blood samples were taken at 0, 1, 2, 4, 8, and 12 hr on days 1, 3, and 5. Twenty-four-hour urine specimens were collected before treatment and on days 1, 3, and 5. Blood and urine specimens were assayed for vidarabine and its principal metabolite, hypoxanthine arabinoside (ara-Hx), by high pressure liquid chromatography. The results showed that vidarabine is quickly deaminated; virtually all of the drug present in the plasma and urine was in the form of ara-Hx. The highest plasma level, approximately 3 microgram/ml, was at the end of the infusion period. The urinary excretion of ara-Hx accounted for between 40% and 50% of the dose. The renal clearance values varied, but were close to the expected glomerular filtration rate of 125 ml/min. The plasma levels and the excretion levels were much the same on days 1, 3, and 5, indicating that drug did not cumulate. The results of the study were consistent with those observed in single-dose studies. The results indicated that the infusion of vidarabine is clinically appropriate, since therapeutic plasma levels are reached promptly, drug is rapidly excreted, and there is no cumulation.


Subject(s)
Vidarabine/metabolism , Biotransformation , Deamination , Herpes Zoster/drug therapy , Humans , Kinetics , Time Factors , Vidarabine/blood , Vidarabine/urine
5.
J Clin Invest ; 62(6): 1142-53, 1978 Dec.
Article in English | MEDLINE | ID: mdl-219024

ABSTRACT

The minimum inhibitory concentration (MIC) of adenine arabinoside (ara-A) in rabbit kidney microtiter tissue cultures (RK-13) to a prototype strain of herpes simplex virus, type 1 (E115) based upon inhibition of cytopathic effects is 1.5 mug/ml. In this system, the MIC of arabinosylhypoxanthine (ara-Hx), the major in vivo metabolic derivative of ara-A, is 75 mug/ml. Inhibition of cytopathic effects of herpes simplex virus, type 1 (HSV-1) in microtiter wells of RK-13 cells varies directly with the concentrations of ara-A or ara-Hx, and inversely with residual HSV-1. The MIC of ara-A for HSV-1 in RK-13 cells is 5-20 times lower than similar measures with vero renal, mouse embryo, or human foreskin cultures. With RK-13 tissue cultures in microtiter plates, an assay for "ara-A equivalents" in human body fluids was developed which compares in sensitivity with high pressure liquid chromatography and has the advantage of simultaneously measuring combined antiherpesvirus effects of ara-A and its major metabolic derivative, ara-Hx. In vitro checkerboard studies in RK-13 cells confirmed that ara-A and ara-Hx in combination had antiviral effects which are synergistic. The total of the fractional MIC of ara-A plus ara-Hx in combination also varies inversely with residual HSV-1 in microtiter wells. Because virus adsorption is complete at 2 h before specimens to be tested are added in this assay, and because human interferon is not measured in rabbit cells, the antiviral assay is not affected by the presence of type-specific antiherpesvirus antibody or human interferon.Antiviral activity (AVA) was assayed as ara-A equivalents in sera and urines from 10 patients with serious herpesvirus infections who received 2.5-20 mg/kg daily of ara-A by intramuscular or intravenous routes. When a dosage schedule of 10 mg/kg per day or more was used, sustained concentrations of AVA that ranged from 0.8 to 14.4 mug/ml were found. When an inhibitor of adenosine deaminase (covidarabine) was not added to the specimens, mean serum concentrations were congruent with3.0 mug/ml (10 mg/kg per day, i.v.), and 4.1 mug/ml (20 mg/kg per day). However, in a single patient given 20 mg/kg of ara-A daily with covidarabine immediately added to the sera, the mean concentration of AVA was 12.9 mug/ml. Urines contained even higher AVA. Assays of 19 sera were performed both by microbiologic assay for AVA and by high pressure liquid chromatography for ara-A and ara-Hx. AVA was greater by microbiologic assay, and was greater than that which could be accounted for by stoichiometric chromatographic measures of ara-A and ara-Hx. These results with sera of treated patients are consistent both with the in vitro synergy of ara-A and ara-Hx found by checkerboard titrations, and with the beneficial responses to ara-A of patients with herpesvirus infections reported here and elsewhere.


Subject(s)
Arabinonucleosides/pharmacology , Herpesviridae Infections/drug therapy , Hypoxanthines/pharmacology , Simplexvirus/drug effects , Vidarabine/pharmacology , Adult , Aged , Antiviral Agents , Biological Assay/methods , Child , Chromatography, High Pressure Liquid , Culture Techniques , Cytopathogenic Effect, Viral/drug effects , Drug Combinations , Drug Synergism , Female , Herpesviridae Infections/blood , Herpesviridae Infections/urine , Herpesvirus 3, Human/drug effects , Herpesvirus 3, Human/growth & development , Humans , Infant, Newborn , Male , Microbiological Techniques , Middle Aged , Simplexvirus/growth & development , Vidarabine/blood , Vidarabine/therapeutic use , Vidarabine/urine
SELECTION OF CITATIONS
SEARCH DETAIL