Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Environ Toxicol Pharmacol ; 9(4): 153-160, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11292578

ABSTRACT

Our objective was to construct a physiologically-based pharmacokinetic (PBPK) model describing the kinetic behavior of 2,4-dichlorophenoxyacetic acid (2,4-D) on rats after long-term exposures to low doses. Our study demonstrated the model's ability to simulate uptake of 2,4-D in discrete areas of the rat brain. The model was derived from the generic PBPK model that was first developed for high-dose, single exposures of 2,4-D to rats or rabbits (Kim, C.S., Gargas, M.L., Andersen, M.E., 1994. Pharmacokinetic modeling of 2,4-dichlorophenoxyacetic acid (2,4-D) in rats and rabbits brain following single dose administration. Toxicol. Lett. 74, 189-201; Kim, C.S., Slikker, W., Jr., Binienda, Z., Gargas, M.L., Andersen, M.E., 1995. Development of a physiologically based pharmacokinetic (PBPK) model for 2,4-dichlorophenoxyacetic acid (2,4-D) dosimetry in discrete areas of the brain following a single intraperitoneal or intravenous dose. Neurotox. Teratol. 17, 111-120.), to which a subcutaneous (hypodermal) compartment was incorporated for low-dose, long-term infusion. It consisted of two body compartments, along with compartments for venous and arterial blood, cerebrospinal fluid, brain plasma and six brain regions. Uptake of the toxin was membrane-limited by the blood-brain barrier with clearance from the brain provided by cerebrospinal fluid 'sink' mechanisms. This model predicted profiles of 2,4-D levels in brain and blood over a 28-day period that compared well with concentrations measured in vivo with rats that had been given 2,4-D (1 or 10 mg/kg per day) with [14C]-2,4-D subcutaneously (s.c.) for 7, 14, or 28 days, respectively. This PBPK model should be an effective tool for evaluating the target tissue doses that may produce the neurotoxicity of organic acid toxicants after low-dose, long-term exposures to contaminated foods or the environment.

2.
Hepatology ; 32(3): 640-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10960462

ABSTRACT

A nuclease resistant ribozyme targeting the 5' untranslated region (5' UTR) of hepatitis C virus (HCV) at site 195 has been identified. To investigate the therapeutic utility of this ribozyme, we evaluated the pharmacokinetics and tissue distribution with two labeled forms of this ribozyme. [(32)P]-labeled ribozyme was administered as a single subcutaneous (SC) or intravenous (IV) bolus at a dose of 10 mg/kg or 30 mg/kg in C57Bl/6 mice. Regardless of route of administration, peak liver concentrations achieved were greater than the concentration necessary to inhibit HCV-IRES-luciferase expression in cell culture. The ribozyme was well absorbed after SC administration (89%) and had an elimination half-life of 23 minutes. To show intracellular localization of the ribozyme in target tissue, a tetramethyl rhodamine (TMR)-labeled ribozyme was administered as a single SC or IV bolus at a dose of 30 mg/kg in C57Bl/6 mice. Mice treated SC or IV with TMR-labeled ribozyme had positive fluorescence in the liver from 15 minutes to 48 hours after dosing. Definite positive fluorescence was still present at 72 hours in the mice dosed via the IV route. At early time points (15 and 30 minutes postinjection), nuclear and possibly cytoplasmic fluorescence was present in the hepatocytes, and sinusoidal fluorescence was intense. At the later time points, fluorescence became more punctate. Abundant staining was often present in Kupffer cells. This study confirms the retention of ribozyme in liver cells and supports the potential of an anti-HCV ribozyme as a therapeutic agent for treatment of chronic hepatitis C.


Subject(s)
DNA, Viral/drug effects , Hepacivirus/genetics , RNA, Catalytic/administration & dosage , RNA, Catalytic/pharmacokinetics , Animals , Base Sequence/genetics , Female , Fluorescent Dyes , Injections, Intravenous , Injections, Subcutaneous , Intracellular Membranes/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Osmolar Concentration , Phosphorus Radioisotopes , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , Rhodamines , Tissue Distribution
3.
Teratology ; 62(2): 93-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931506

ABSTRACT

BACKGROUND: The overall goal of human immunodeficiency virus (HIV) therapy during pregnancy is to maintain maternal health and reduce the probability of vertical transmission during gestation and delivery, while keeping toxicity risks low. Azidothymidine (AZT) is currently recommended for pregnant women infected with HIV; however, many pregnant women are unable to tolerate AZT because of toxicity. In the present study, the placental transfer and fetal accumulation of the anti-HIV compound 2',3'-didehydro-3'-deoxythymidine (d4T) and its active (triphosphorylated) and inactive (thymine and beta-aminoisobutyric acid) metabolites were examined at steady state in late-term rhesus macaques. METHODS: On the day of the hysterotomy, the mother was administered an intravenous loading dose of d4T, followed by a 3-hr steady-state intravenous infusion that also included [(3)H]d4T as a tracer. After 3 hr of infusion, the fetus was delivered by cesarean section under halothane/N(2)O anesthesia. Plasma, amniotic fluid, and tissues were analyzed for d4T and its inactive metabolites by HPLC; tissue samples were analyzed for d4T and active (phosphorylated) metabolites by strong anion-exchange HPLC. RESULTS: Maternal steady-state plasma concentrations of d4T were 1-2 microg/ml, with a fetal-to-maternal plasma ratio of 0.85 +/- 0.09. The fetal tissue distribution of radioactivity was highest in the kidney and lowest in the brain. D4T, thymine, and beta-aminoisobutyric acid were detected in all fetal tissues examined. CONCLUSIONS: Our data indicate that d4T readily crosses the placenta and is present in the fetus as parent compound or its inactive metabolites after maternal infusion. Although fetal plasma concentrations of d4T were similar to clinical d4T concentrations, no phosphorylated metabolites were detected. Teratology 62:93-99, 2000. Published 2000 Wiley-Liss, Inc.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Fetus/metabolism , Macaca mulatta/embryology , Placenta/drug effects , Stavudine/pharmacokinetics , Aminoisobutyric Acids/pharmacokinetics , Animals , Anti-HIV Agents/blood , Anti-HIV Agents/metabolism , Chromatography, High Pressure Liquid , Female , Kidney/drug effects , Kidney/embryology , Maternal-Fetal Exchange , Pregnancy , Stavudine/blood , Stavudine/metabolism , Thymine/pharmacokinetics , Time Factors , Tissue Distribution
4.
Antisense Nucleic Acid Drug Dev ; 10(3): 153-62, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10905552

ABSTRACT

The potential acute toxicity of a ribozyme (ANGIOZYME) targeting the flt-1 vascular endothelial growth factor (VEGF) receptor mRNA was evaluated in cynomolgus monkeys following i.v. infusion or s.c. injection. ANGIOZYME was administered as a 4-hour i.v. infusion at doses of 10, 30, or 100 mg/kg or a s.c. bolus at 100 mg/kg. End points included blood pressure, electrocardiogram (ECG), clinical chemistry, hematology, complement factors, coagulation parameters, and ribozyme plasma concentrations. ANGIOZYME was well tolerated, with no drug-associated morbidity or mortality. There was no clear evidence of ANGIOZYME-related adverse effects in this study. Slight increases in spleen weight and lymphoid hyperplasia were observed in several animals. However, these changes were not dose dependent. Steady-state concentrations of ANGIOZYME were achieved during the 4-hour infusion of 10, 30, or 100 mg/kg. Dose-dependent elimination of ANGIOZYME was observed, with faster clearance at the two highest doses. ANGIOZYME was slowly absorbed after s.c. administration, resulting in steady-state concentrations for the 9-hour sampling period. Monkeys in this toxicology study received significant plasma ANGIOZYME exposure by both the s.c. and i.v. routes.


Subject(s)
Gene Targeting , RNA, Catalytic/pharmacokinetics , RNA, Catalytic/toxicity , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/toxicity , Animals , Blood Chemical Analysis , Blood Coagulation Factors/analysis , Chromatography, High Pressure Liquid , Complement System Proteins/analysis , Drug Administration Schedule , Female , Infusions, Intravenous , Injections, Intravenous , Injections, Subcutaneous , Macaca fascicularis , Male , RNA, Catalytic/administration & dosage , RNA, Catalytic/blood , Receptors, Vascular Endothelial Growth Factor
5.
Anal Biochem ; 283(2): 228-40, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10906244

ABSTRACT

Versatile bioanalytical assays to detect chemically stabilized hammerhead ribozyme and putative ribozyme metabolites from plasma are described. The extraction protocols presented are based on serial solid-phase extractions performed on a 96-well plate format and are compatible with either IEX-HPLC or CGE back-end analysis. A validation of both assays confirmed that both the HPLC and the CGE methods possess the required linearity, accuracy, and precision to accurately measure concentrations of hammerhead ribozyme extracted from plasma. These methods should be of general use to detect and quantitate ribozymes from other biological fluids such as serum and urine.


Subject(s)
Chromatography, High Pressure Liquid/methods , Electrophoresis, Capillary/methods , RNA, Catalytic/blood , Animals , Ethanolamines/chemistry , Male , Mice , Oligonucleotides/chemistry , Quality Control , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results
6.
Neurotoxicol Teratol ; 22(3): 429-34, 2000.
Article in English | MEDLINE | ID: mdl-10840187

ABSTRACT

The anti-HIV therapeutic dideoxyinosine (ddI) has been reported to produce a painful, dose-limiting peripheral myelinopathy in HIV-infected patients after chronic administration. We have previously demonstrated ddI-induced myelinopathy in a non-HIV-infected rat model after 20 weeks of dosing, characterized by myelin splitting and intramyelin edema. The present study examined the time course needed to produce the ddI-induced neuropathy. Adult male Sprague-Dawley rats were gavaged with vehicle or 415 mg/kg ddI twice daily for up to 20 weeks. Groups of treated (n = 6-8) and control (n = 3-5) animals were killed after 5, 10, 15, and 20 weeks of dosing and the distal end of the sciatic nerve was removed. The nerve was postfixed by immersion in neutral phosphate-buffered formalin, dehydrated in graded alcohols, and embedded in plastic embedding media. One-micrometer-thick sections were cut and stained with toluidine blue and basic fuchsin. Plasma levels of ddI on the day the animals were killed were greater than 10 microgram/ml within the first hour after dosing and fell rapidly to less than 1 microgram/ml (clinical range 1-2 microgram/ml) within 3 h after dosing. The abnormalities observed in the sciatic nerve were few, if any, after 5 or 10 weeks, but very prominent after 15 weeks of dosing. Four of the six ddI-treated rats exhibited abnormal morphology as evidenced by myelin splitting and ballooned myelin sheaths. Although abnormal morphology was present at 20 weeks of dosing, the effect was not as robust as at 15 weeks. This suggests that the nerve may partially recover from the effects of ddI with time. Published by Elsevier Science Inc.


Subject(s)
Anti-HIV Agents/toxicity , Didanosine/toxicity , Myelin Sheath/pathology , Neurotoxicity Syndromes/pathology , Peripheral Nervous System Diseases/chemically induced , Animals , Anti-HIV Agents/blood , Blood Chemical Analysis , Didanosine/blood , Male , Peripheral Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/pathology
7.
J Clin Pharmacol ; 40(12 Pt 2): 1462-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11185667

ABSTRACT

The pharmacokinetics and tolerability of a chemically stabilized synthetic ribozyme (ANGIOZYME) targeting the Flt-1 VEGF receptor mRNA were evaluated in healthy volunteers. In a placebo-controlled, single-dose escalation study, ribozyme was administered as a 4-hour i.v. infusion of 10 or 30 mg/m2 or as a s.c. bolus of 20 mg/m2. Peak ribozyme plasma concentrations of 1.5 and 3.8 micrograms/mL were observed after the 10 and 30 mg/m2 i.v. infusions, respectively. When normalized to dose, AUC values as well as peak concentrations increased proportionally as the dose was increased from 10 to 30 mg/m2. Peak concentrations of 0.9 microgram/mL were observed approximately 3.25 hours after a 20 mg/m2 s.c. bolus of ribozyme. The dose-normalized AUCs obtained after s.c. dosing were compared to the mean dose-normalized AUC after i.v. dosing to estimate an absolute s.c. bioavailability (f) of approximately 69%. An average elimination half-life of 28 to 40 minutes was observed after i.v. administration, which increased to 209 minutes after s.c. administration. Only 4 of 12 reported adverse events were possibly related to administration of ribozyme (headache and somnolence). Thus, ribozyme administration was well tolerated after a single 4-hour i.v. infusion of up to 30 mg/m2 or a single s.c. bolus of 20 mg/m2.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , RNA, Catalytic/pharmacokinetics , Adult , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/blood , Double-Blind Method , Female , Humans , Male , Metabolic Clearance Rate , RNA, Catalytic/adverse effects , RNA, Catalytic/blood
8.
Curr Issues Mol Biol ; 2(4): 113-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11471755

ABSTRACT

A significant amount of research has been devoted to the chemical stabilization of synthetic ribozymes, in part, so that applications to systemic disease can be explored. A nuclease-stabilized synthetic hammerhead ribozyme, ANGIOZYME, has been developed which targets the mRNA encoding a vascular endothelial growth factor receptor, Flt-1. Because the stimulation of this receptor may contribute to tumor neovascularization and subsequent tumor growth and metastasis, we have explored the systemic use of ANGIOZYME to down regulate this receptor in a syngeneic model of metastatic cancer. We describe here the application of pharmacokinetic analysis to the selection of a dosing regimen for pharmacodynamic screening in this murine cancer model. These studies demonstrate that the appropriate application of pharmacokinetic analysis is necessary for the optimization of systemic pharmacodynamic studies using synthetic ribozymes.


Subject(s)
RNA, Catalytic/pharmacology , RNA, Catalytic/pharmacokinetics , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Base Sequence , Extracellular Matrix Proteins/genetics , Female , Mice , Mice, Inbred C57BL , Myosin Heavy Chains , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Nonmuscle Myosin Type IIB , RNA Stability , RNA, Catalytic/administration & dosage , RNA, Catalytic/genetics , Vascular Endothelial Growth Factor Receptor-1
9.
Antisense Nucleic Acid Drug Dev ; 9(3): 271-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10435752

ABSTRACT

Vascular endothelial growth factor (VEGF) is a growth factor that contributes to the angiogenesis of developing tumors. To interfere with the action of VEGF, a nuclease-stabilized ribozyme, ANGIOZYME, has been developed against VEGF receptor subtype Flt-1 mRNA. To determine which routes of administration would be useful for systemic delivery of this ribozyme, a dose of 30 mg/kg [32P]ANGIOZYME was administered as an i.v., i.p., or s.c. bolus. Concentrations of ANGIOZYME in plasma, femur, kidney, liver, and lung were examined. ANGIOZYME was well absorbed after i.p. (90%) or s.c. administration (77%), with peak plasma concentrations occurring 30 minutes after dosing. Total body clearance after a single dose of 30 mg/kg ANGIOZYME was 20 ml/min/kg, and the elimination half-life was 33 minutes. The apparent volume of distribution at steady-state ranged from 0.5 to 1.3 L/kg. ANGIOZYME was detected in the four tissues examined through the 3 hour sampling period after i.v. or i.p. administration. After s.c. administration, ANGIOZYME was detected in femur, kidney, and lung but not in the liver. The highest concentrations of ANGIOZYME were found in kidney and femur with all three routes. Because of the rapid and extensive absorption after extravascular injections, either i.p. or s.c. administration could be considered for use in pharmacodynamic studies examining the effects of ANGIOZYME or other ribozymes with similar chemical modifications.


Subject(s)
Neovascularization, Pathologic , RNA, Catalytic/pharmacokinetics , Animals , Female , Half-Life , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/prevention & control , RNA, Catalytic/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor , Tissue Distribution
10.
Environ Toxicol Pharmacol ; 6(1): 49-58, 1998 Aug 04.
Article in English | MEDLINE | ID: mdl-21781881

ABSTRACT

The purpose of this study was to construct a physiologically based pharmacokinetic model and demonstrate its ability to predict low-dose uptake of domoic acid, a seafood contaminant, in discrete areas of the rat brain. The model we used was derived from the generic PBPK model of our previous studies with 2,4-dichlorophenoxyacetic acid (Kim et al., 1994. Pharmacokinetic modeling of 2,4-dichlorophenoxyacetic acid (2,4-D) in rats and in rabbits brain following single dose administration. Toxicol. Lett. 74, 189; Kim et al., 1995. Development of a physiologically based pharmacokinetic model for 2,4-dichlorophenoxyacetic acid dosimetry in discrete areas of the rabbit brain. Neurotoxicol. Teratol. 17, 111), to which physiological- and chemical-specific parameters for domoic acid were applied. It incorporates two body compartments along with compartments for venous and arterial blood, cerebrospinal fluid, brain plasma and seven brain regions. Uptake of the blood-borne toxin is membrane-limited by the blood-brain barrier with clearance from the brain provided by cerebrospinal fluid `sink' mechanisms. This model generated predicted profiles of toxin level in brain and blood over a 1-h period that compared reasonably well with concentrations calculated from in vivo data of rats that had been given [(3)H]domoic acid intravenously (Preston and Hynie, 1991. Transfer constants for blood-brain barrier permeation of the neuroexcitatory shellfish toxin, domoic acid. Can. J. Neurol. Sci. 18, 39). This PBPK model should be an effective tool for evaluating the target doses that produce the potential neurotoxicity of domoic acid found in foods.

11.
Drug Metab Dispos ; 25(4): 453-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9107545

ABSTRACT

3'-Azido-3'-deoxythymidine (AZT) is currently prescribed to pregnant women infected with human immunodeficiency virus to reduce the risk of vertical transmission of the virus to the fetus. Consequently, more information is needed concerning the placental transfer and tissue distribution of AZT and its metabolites. In the present study, the placental transfer and fetal accumulation of AZT, its glucuronide metabolite [3'-azido-3'-deoxythymidine-beta-D-glucuronide (AZTG)], and phosphorylated metabolites were examined at steady-state in near-term rhesus macaques. One to 2 weeks before a chronic infusion, an intravenous bolus of 8 mg/kg AZT was administered to pregnant animals to determine the dose of AZT needed to reach steady-state plasma concentrations. On the day of hysterotomy, the mother was administered an intravenous loading dose of AZT, followed by a 3-hr steady-state intravenous infusion that also included a trace of [3H]AZT. After 3 hr of infusion, the mother was anesthetized, and the fetus was delivered. Plasma and amniotic fluid were analyzed for AZT and AZTG by HPLC, and tissue samples were analyzed for AZT, AZTG, and phosphorylated metabolites by strong anion exchange HPLC. Maternal steady-state plasma concentrations were 1.3-2.2 micrograms/ml for AZT and 2.3-8.0 micrograms/ml for AZTG. Fetal AZT and AZTG plasma concentrations were both lower (0.98-2.3 micrograms/ml and 1.3-5.4 micrograms/ml, respectively) than maternal concentrations, with fetal-to-maternal plasma ratios of 0.63-1.0 for AZT. Fetal tissue distribution of tritium was highest in the kidney and lowest in the brain. Although the active triphosphorylated metabolite was not detected in the fetus, the AZT-monophosphate was detected in almost all fetal tissues examined. Our data indicate that AZT is rapidly converted to the glucuronide and monophosphate metabolites in the fetus after maternal infusion.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Fetus/metabolism , Maternal-Fetal Exchange , Pregnancy, Animal/metabolism , Zidovudine/pharmacokinetics , Animals , Anti-HIV Agents/administration & dosage , Female , Macaca mulatta , Pregnancy , Tissue Distribution , Zidovudine/administration & dosage , Zidovudine/analogs & derivatives
12.
Neurotoxicol Teratol ; 19(2): 151-5, 1997.
Article in English | MEDLINE | ID: mdl-9136132

ABSTRACT

Fumonisin B1 (FB1), a toxic metabolite of the fungus Fusarium moniliforme found in contaminated corn, is considered an etiologic agent of equine leukoencephalomalacia. Because FB1 exposure is associated with alteration of sphingolipid metabolism, the purpose of this study was to elucidate whether blood sphinganine (Sa) levels affect brain Sa levels. Sa and sphingosine (So) levels in brain tissue and plasma were analyzed by HPLC. Area under the curve (AUC0-20h) ratios of brain Sa to plasma Sa levels were about 40 after a single 0.8 or 8 mg/kg SC dose of FB1. The AUC0-12h ratio of brain FB1 to plasma FB1 was 0.02. The fact that FB1 alters brain Sa levels and Sa/So ratios indicates that sphingolipid metabolism in the central nervous system of developing rats is vulnerable to FB1 exposure. These data support our hypothesis that alterations of the brain Sa levels are related to the direct action of FB1 on the brain rather than transport of peripheral Sa to the brain.


Subject(s)
Blood-Brain Barrier/drug effects , Carboxylic Acids/toxicity , Carcinogens, Environmental/toxicity , Fumonisins , Sphingosine/analogs & derivatives , Animals , Area Under Curve , Blood-Brain Barrier/physiology , Carboxylic Acids/blood , Carcinogens, Environmental/analysis , Chromatography, High Pressure Liquid , Female , Male , Pregnancy , Prosencephalon/chemistry , Rats , Rats, Sprague-Dawley , Sphingosine/blood , Sphingosine/pharmacokinetics
13.
J Toxicol Environ Health ; 49(5): 497-509, 1996 Dec 06.
Article in English | MEDLINE | ID: mdl-8968410

ABSTRACT

The distribution of 2,4-dichlorophenoxyacetic acid (2,4-D) was examined in maternal and fetal rabbits. Pregnant New Zealand rabbits (28-30 d gestational age) were anesthetized with ketamine/xylazine and the femoral vein and artery were catheterized for compound administration and sampling. Dams received iv [14C]2,4-D (12.5 microCi/kg) with unlabeled sodium 2,4-D (1, 10, or 40 mg/kg) in saline. Blood and tissue were collected up to 2 h after dosing. Fetal to maternal plasma AUC ratios were 0.09, 0.07, and 0.16 after the 1, 10, or 40 mg/kg dose, respectively. Extraplasma AUCs were greatest in maternal kidney and uterus and lowest in maternal and fetal brain. A greater than fourfold elevation in fetal AUC was found when the dose was increased from 10 to 40 mg/kg, suggesting saturation of maternal plasma binding of 2,4-D. Although the in vitro fetal brain tissue to incubation media ratio was unity (1.03 +/- 0.1, mean +/- SD), fetal brain AUCs were 10% or less of the fetal plasma AUCs, indicating the brain barrier system to 2,4-D is functioning in the late-gestation fetal rabbit. However, its development may not be complete due to the higher brain tissue to plasma ratios in the fetus compared to the dam.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/pharmacokinetics , Fetus/metabolism , 2,4-Dichlorophenoxyacetic Acid/administration & dosage , 2,4-Dichlorophenoxyacetic Acid/blood , Animals , Area Under Curve , Brain/metabolism , Brain Chemistry , Female , Fetus/chemistry , In Vitro Techniques , Injections, Intravenous , Kidney/chemistry , Kidney/metabolism , Pregnancy , Rabbits , Tissue Distribution , Uterus/chemistry , Uterus/metabolism
15.
Neurotoxicol Teratol ; 18(5): 555-63, 1996.
Article in English | MEDLINE | ID: mdl-8888020

ABSTRACT

The antiviral adenosine analog, 2',3'-dideoxyinosine (ddI), and the antitubercular nicotinic acid analogue, isoniazid, have recently received widespread clinical application in the treatment of acquired immunodeficiency syndrome (AIDS). Clinical studies indicate that the primary dose limiting side effect of both drugs is neurological in nature. Most clinical studies are confounded by the fact that the observed neuropathy must be evaluated in the presence of the ongoing disease process associated with human immunodeficiency virus (HIV) infection. The purpose of this study was to develop and validate a rat model of ddI-and isoniazid-induced neuropathy in the absence of any disease-induced pathology. Myelin splitting and intramyelin edema were the most frequent abnormalities observed in the sciatic nerves of ddI-dosed animals, whereas whorls, extracellular debris, macrophages, and reduced myelinated axon number were seen following chronic isoniazid administration. Isoniazid also resulted in myelinopathy of the CNS. Thus, contrary to previous reports, the rodent is a suitable model for ddI- and isoniazid-induced neuropathies.


Subject(s)
Brain/pathology , Didanosine/toxicity , Isoniazid/toxicity , Neural Conduction/drug effects , Neurotoxins/toxicity , Sciatic Nerve/pathology , Animals , Anti-HIV Agents/toxicity , Antitubercular Agents/toxicity , Axons/pathology , Brain/drug effects , Didanosine/adverse effects , Edema , HIV Infections/drug therapy , Humans , Male , Mast Cells/pathology , Myelin Sheath/pathology , Rats , Rats, Sprague-Dawley , Reference Values , Sciatic Nerve/drug effects , Sciatic Nerve/physiology
16.
Toxicol Appl Pharmacol ; 136(2): 250-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8619233

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model that describes the kinetics of organic anions by using 2,4-dichlorophenoxyacetic (2,4-D) as a representative compound was constructed for the developing rabbit brain at near-term pregnancy (Gestation Day 30). The model consisted of brain, body, and venous and arterial compartments for the mother which were linked to the fetus by a placenta. Maternal brain compartments in the model were brain plasma, cerebrospinal fluid (CSF), and brain tissue including hypothalamus, caudate nucleus, hippocampus, forebrain, brainstem, and cerebellum. The fetus consisted of brain, body, amniotic fluid, and venous and arterial compartments. the maternal body had both a central and a deep compartment; the fetal body had only one compartment. Maternal blood flow to the fetus was modeled as blood flowing to the placenta, where it was equilibrated before it reached the fetus. The brain uptake was membrane-limited by the blood-brain barrier, with saturable clearance from the CSF into the venous blood by the choroid plexus in both fetus and mother. The model was used to compare concentrations of 2,4-D in maternal and fetal brain, maternal and fetal plasma, and amniotic fluid over time with experimental data from pregnant rabbits given 2,4-D intravenously (1, 10, or 40 mg/kg). The model adequately simulated the 2-hr time course of 2,4-D concentrations in both mother and fetus. With continued development, this generic PBPK model should be a useful tool for evaluating the safety of organic acid neurotoxicants in the developing brain.


Subject(s)
2,4-Dichlorophenoxyacetic Acid/pharmacokinetics , Brain/embryology , Brain/metabolism , Fetus/metabolism , Herbicides/pharmacokinetics , 2,4-Dichlorophenoxyacetic Acid/administration & dosage , 2,4-Dichlorophenoxyacetic Acid/blood , 2,4-Dichlorophenoxyacetic Acid/toxicity , Amniotic Fluid/metabolism , Animals , Brain/drug effects , Female , Fetus/drug effects , Herbicides/toxicity , Maternal Exposure , Maternal-Fetal Exchange , Models, Biological , Placenta/metabolism , Pregnancy , Rabbits , Tissue Distribution
17.
FASEB J ; 9(12): 1157-63, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7672508

ABSTRACT

As the incidence of human immunodeficiency virus (HIV) infection has increased in women over the past decade, the need for safe, effective therapy during pregnancy has increased concomitantly. Although dideoxynucleosides such as 3'-deoxy-3'-azidothymidine (AZT), 2',3'-dideoxyinosine (ddI), 2',3'-dideoxycytidine (ddC), and 2',3'-didehydro-3'-deoxythymidine have been approved for use in the general population, the administration, efficacy, and toxicity of these compounds during pregnancy and development are now being investigated. Initial human studies suggest that maternal use of AZT during pregnancy is well tolerated by both mother and child and provides a promising degree of protection from vertical HIV transmission to the infant. In vitro and animal models have greatly increased our understanding of the distribution and toxicity resulting from fetal dideoxynucleoside exposure. AZT, ddI, and ddC rapidly cross the placenta by simple diffusion but with different rates of transfer. In vivo data confirm the differential transfer of these compounds with AZT fetal exposure approximately twice that of ddI or ddC. Active phosphorylated metabolites have been detected in placental tissue after in vitro perfusion with AZT. The active triphosphate has not been detected in placental perfusion studies or in the fetal rhesus monkey 3 h after maternal exposure to ddI or ddC. Although in vitro and in vivo laboratory animal studies suggest the potential for toxicity with preimplantation exposure, the risk for teratogenic events after postimplantational exposure appears to be low at therapeutically effective concentrations of these dideoxynucleosides.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Antiviral Agents/toxicity , Dideoxynucleosides/toxicity , Pregnancy Complications/drug therapy , Prenatal Exposure Delayed Effects , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/transmission , Animals , Antiviral Agents/adverse effects , Dideoxynucleosides/adverse effects , Embryonic and Fetal Development , Female , Humans , Maternal-Fetal Exchange , Pregnancy
19.
Neurotoxicology ; 16(1): 169-77, 1995.
Article in English | MEDLINE | ID: mdl-7603638

ABSTRACT

The pharmacokinetics and metabolism of cocaine (COC) were determined in late gestation maternal and fetal guinea pigs. After a single i.v. dose of 2-12 mg/kg, the average +/- SD total body clearance of COC was 59 +/- 16 ml/min/kg and was not dose dependent. However, volume of distribution was 2.1 and 3.9 l/kg, mean resident time (MRT) was 42 and 57 min, and elimination half-life was 34 and 49 min at the 2 and 4 mg/kg dose of COC, respectively. With the exception of an increased MRT, the pharmacokinetics were similar after s.c. COC administration. Benzoylecgonine (BE) and benzoylnorecgonine (BN) were major and persistent metabolites. Norcocaine (NOR) concentrations were low and transient. After chronic maternal administration of 6 mg/kg COC s.c., there was no difference between maternal and fetal plasma COC concentrations one hour after the last injection, but COC and BN accumulated in amniotic fluid. Examination of in vitro metabolism of COC in fetal and maternal guinea pig hepatic microsomes demonstrated minimal fetal N-demethylation and induction of maternal N-demethylation by chronic COC exposure. The minimal fetal N-demethylation suggest BN seen previously in vivo after chronic maternal COC administration resulted from maternal formation of NOR and subsequent maternal and/or fetal hydrolysis to BN.


Subject(s)
Cocaine/pharmacokinetics , Maternal-Fetal Exchange , Animals , Animals, Newborn , Biotransformation , Cocaine/analogs & derivatives , Cocaine/metabolism , Female , Fetus/metabolism , Guinea Pigs , Injections, Intravenous , Kinetics , Microsomes/metabolism , Placenta/metabolism , Pregnancy , Sodium Fluoride/pharmacology , Time Factors , Tissue Distribution
20.
Drug Metab Dispos ; 22(1): 154-60, 1994.
Article in English | MEDLINE | ID: mdl-8149876

ABSTRACT

The intravenous pharmacokinetics of 13-cis-, all-trans-, 13-cis-4-oxo, and all-trans-4-oxo retinoic acid (RA) were determined in nonpregnant female cynomolgus monkeys. All-trans- and 13-cis-RA were injected at two doses (0.25 or 0.0125 mg/kg) and all-trans-4-oxo RA and 13-cis-4-oxo RA at 0.25 mg/kg. Total body clearance, volume of distribution, and volume of distribution at steady state of all-trans-RA were dose-dependent with greater values at the lower dose. Elimination half-life was longer for the cis-compounds and not dose-dependent (N = 1 for 13-cis-4-oxo RA, N = 3 for other compounds, harmonic mean +/- pseudostandard deviation, min): 13-cis-4-oxo RA (837) > or = 13-cis-RA (301 +/- 204) > all-trans-RA (38 +/- 3) > all-trans-4-oxo RA (11 +/- 2). Secondary plasma peaks were noted only after administration of 13-cis-4-oxo RA. The low area under the time concentration curves for observable metabolites after intravenous injection of the oxidated compounds suggests further metabolism plays a minimal role in the elimination of these compounds from the monkey. Plasma-time concentration curves were fitted to multicompartmental models and suggested < 30% of each compound was available in the central compartment for elimination in the postdistribution phase. A comparison of the kinetics of the isomers indicated oxidation of all-trans-RA to all-trans-4-oxo RA increased mean total body clearance values 4-fold.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Tretinoin/pharmacokinetics , Animals , Female , Injections, Intravenous , Macaca fascicularis , Time Factors , Tretinoin/administration & dosage , Tretinoin/analogs & derivatives , Tretinoin/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...