Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Front Psychiatry ; 12: 680177, 2021.
Article in English | MEDLINE | ID: mdl-34483982

ABSTRACT

Background: Prenatal exposure to serotonin reuptake inhibitor (SRI) antidepressants increases risk for adverse neurodevelopmental outcomes, yet little is known about whether effects are present before birth. In relation to maternal SRI pharmacokinetics, this study investigated chronic and acute effects of prenatal SRI exposure on third-trimester fetal heart rate variability (HRV), while evaluating confounding effects of maternal depressed mood. Methods: At 36-weeks' gestation, cardiotocograph measures of fetal HR and HRV were obtained from 148 pregnant women [four groups: SRI-Depressed (n = 31), SRI-Non-Depressed (n = 18), Depressed (unmedicated; n = 42), and Control (n = 57)] before, and ~5-h after, typical SRI dose. Maternal plasma drug concentrations were quantified at baseline (pre-dose) and four time-points post-dose. Mixed effects modeling investigated group differences between baseline/pre-dose and post-dose fetal HR outcomes. Post hoc analyses investigated sex differences and dose-dependent SRI effects. Results: Maternal SRI plasma concentrations were lowest during the baseline/pre-dose fetal assessment (trough) and increased to a peak at the post-dose assessment; concentration-time curves varied widely between individuals. No group differences in fetal HR or HRV were observed at baseline/pre-dose; however, following maternal SRI dose, short-term HRV decreased in both SRI-exposed fetal groups. In the SRI-Depressed group, these post-dose decreases were displayed by male fetuses, but not females. Further, episodes of high HRV decreased post-dose relative to baseline, but only among SRI-Non-Depressed group fetuses. Higher maternal SRI doses also predicted a greater number of fetal HR decelerations. Fetuses exposed to unmedicated maternal depressed mood did not differ from Controls. Conclusions: Prenatal SRI exposure had acute post-dose effects on fetal HRV in late gestation, which differed depending on maternal mood response to SRI pharmacotherapy. Importantly, fetal SRI effects were sex-specific among mothers with persistent depressive symptoms, as only male fetuses displayed acute HRV decreases. At trough (pre-dose), chronic fetal SRI effects were not identified; however, concurrent changes in maternal SRI plasma levels suggest that fetal drug exposure is inconsistent. Acute SRI-related changes in fetal HRV may reflect a pharmacologic mechanism, a transient impairment in autonomic functioning, or an early adaption to altered serotonergic signaling, which may differ between males and females. Replication is needed to determine significance with postnatal development.

2.
Xenobiotica ; 49(11): 1360-1372, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30587071

ABSTRACT

1. The objective was to determine the ontogeny of stereoselective fluoxetine (FX) disposition in postnatal sheep from newborn to adulthood. 2. Catheters were implanted in a carotid artery and jugular vein. FX was administered intravenously, followed by serial arterial blood and cumulative urine collection. The concentrations of R,S-FX and R,S-norfluoxetine (R,S-NFX) in samples were measured using a validated enantioselective LC/MS/MS analytical method. 3. The metabolism of FX at 4.2 ± 0.4 days was limited compared to adults, but had developed compared to the fetus. Total body clearance (ClTB) did not significantly increase up to 33.6 ± 0.9 days, but significantly increased at 98.5 ± 2.0 days, with no further changes up to 397.3 ± 8.5 days. Up to 13.4 ± 0.8 days, the disposition of FX included Phase I metabolism to NFX and trifluoromethylphenol (TFMP), and renal elimination. At 32.9 ± 0.9 days, metabolism included Phase II conjugates of FX and NFX. Renal elimination of these compounds was low. 4. The elimination of FX increased in a non-linear manner during the first year in sheep. The metabolism and disposition of FX and NFX in plasma and urine were stereoselective and this appeared due to both stereoselective protein binding and metabolism.


Subject(s)
Fluoxetine/pharmacokinetics , Animals , Animals, Newborn , Blood Proteins/metabolism , Female , Fluoxetine/analogs & derivatives , Fluoxetine/blood , Fluoxetine/chemistry , Fluoxetine/metabolism , Inactivation, Metabolic , Injections, Intravenous , Isomerism , Male , Metabolic Clearance Rate , Sheep
3.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(5-6): 349-58, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242112

ABSTRACT

A liquid chromatography-tandem mass spectrometry (LC/MS/MS) method was developed and validated for the quantitation of (R)-, (S)-fluoxetine, and (R)-, (S)-norfluoxetine in ovine plasma. The analytes were extracted from ovine plasma at a basic pH using a single-step liquid-liquid extraction with methyl-tert-butyl ether. Chromatographic separation of all enantiomers was achieved using an AGP-chiral column with a run time of 10 min. (R)-, (S)-fluoxetine, and (R)-, (S)-norfluoxetine were quantitated at the total ion current (TIC) of multiple reaction monitoring (MRM) transitions of m/z 310.2→44.1, m/z 310.2→147.7 for (R)-, (S)-fluoxetine, and m/z 296.2→30.3, m/z 296.2→133.9 for (R)-, (S)-norfluoxetine. This method was validated for accuracy, precision, linearity, range, limit of quantitation (LOQ), selectivity, recovery, dilution integrity, matrix effect, and evaluation of carry-over. Observed accuracy ranges were as follows: (R)-fluoxetine -8.82 to 3.75%; (S)-fluoxetine -10.8 to 1.46%; (R)-norfluoxetine -7.50 to 0.37% and (S)-norfluoxetine -8.77% to -1.33%. Observed precision ranges were as follows: (R)-fluoxetine 5.29-11.5%; (S)-fluoxetine 3.91-11.1%; (R)-norfluoxetine 4.32-7.67% and (S)-norfluoxetine -8.77% to -1.33%. The calibration curves were weighted (1/X(2), n=4) and observed to be linear for all analytes with the following r(2) values: (R)-fluoxetine ≥ 0.997; (S)-fluoxetine ≥ 0.996; (R)-norfluoxetine ≥ 0.989 and (S)-norfluoxetine ≥ 0.994. The analytical range of the method was 1-500 ng/ml with an LOQ of 1 ng/ml for all analytes, using a sample volume of 300 µL.


Subject(s)
Chromatography, Liquid/methods , Fluoxetine/analogs & derivatives , Fluoxetine/blood , Tandem Mass Spectrometry/methods , Animals , Fluoxetine/chemistry , Fluoxetine/pharmacokinetics , Least-Squares Analysis , Reproducibility of Results , Sensitivity and Specificity , Sheep , Stereoisomerism
4.
Reprod Fertil Dev ; 22(2): 459-67, 2010.
Article in English | MEDLINE | ID: mdl-20047731

ABSTRACT

The objective was to compare gestation length in chronically instrumented (laboratory) pregnant sheep (n = 131) and in the breeding flock (n = 476) that provided the experimental sheep. In the breeding flock, gestation length was normally distributed and varied between 141 and 151 days (mean = 147 +/- 0.1 days). In the laboratory sheep, gestation length varied between 128 and 151 days (mean = 142 +/- 1 day), and was bimodal, with 35.9% delivering preterm (<141 days). To examine potential factors that contributed to the preterm birth, a severity score was used, which comprised surgery characteristics, number of experiments and maternal or fetal complications. There was a significant inverse linear relationship (P < 0.001) between the total severity score and gestation length. The median values for the surgical (15 v. 12), overall complication (6 v. 2), maternal complication (2 v. 0) and fetal complication (2 v. 2) components were significantly greater in the preterm compared with the term groups. There was no relationship between fetal number and gestation length in either group. It is concluded that in chronic pregnant sheep preparations, there is a significant incidence of preterm birth and that this is associated with the severity of the surgical intervention and with several maternal and fetal complications.


Subject(s)
Pregnancy, Animal/physiology , Sheep/embryology , Animals , Animals, Domestic , Animals, Laboratory , Animals, Newborn , Birth Weight , Female , Fetal Blood/metabolism , Gestational Age , Pregnancy , Pregnancy Complications/veterinary , Premature Birth/etiology , Premature Birth/veterinary , Stress, Physiological
5.
Drug Metab Dispos ; 35(8): 1380-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17496206

ABSTRACT

Sigmoidal or autoactivation kinetics has been observed in vitro for both cytochrome P450- and UDP-glucuronosyltransferase-catalyzed enzymatic reactions. However, the in vivo relevance of sigmoidal kinetics has never been clearly demonstrated. In the current study we investigate the kinetics of valproic acid glucuronide (VPAG) formation both in vivo in adult sheep and in vitro in sheep liver microsomes (pool of 10). After a 100 mg/kg i.v. bolus dose of valproic acid (VPA) to adult sheep (n = 5), the majority of the dose was recovered in urine as VPAG (approximately 79%). Eadie-Hofstee plots of the VPAG formation rate (calculated from urinary excretion rate data for VPAG) were characteristic of autoactivation kinetics and provided estimates of the apparent maximum velocity of an enzymatic reaction (V(max)(app)), the substrate concentration resulting in 50% of V(max)(app) (S(50)(app)), and Hill coefficient (n) of 2.10 +/- 0.75 micromol/min/kg, 117 +/- 56 microM, and 1.34 +/- 0.14, respectively. Comparable estimates of V(max)(app) (2.63 +/- 0.33 micromol/min/kg), S(50)(app) (118 +/- 53 microM), and n (2.06 +/- 0.47) describing overall VPA elimination from plasma were obtained by fitting VPA unbound plasma concentration-time data to a two-compartment model with elimination described by the Hill equation. Consistent with our in vivo observations, Eadie-Hofstee plots of VPAG formation in sheep liver microsomes were characteristic of autoactivation kinetics. To our knowledge, these data provide the first clear demonstration that autoactivation kinetics observed in vitro in liver preparations can translate to the in vivo situation at least under certain experimental conditions and confirm its relevance.


Subject(s)
Valproic Acid/analogs & derivatives , Valproic Acid/metabolism , Animals , Catalysis , Enzyme Activation , Female , Kinetics , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Sheep , Valproic Acid/pharmacokinetics , Valproic Acid/urine
6.
Drug Metab Dispos ; 35(8): 1285-91, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17485495

ABSTRACT

The central nervous system (CNS) pharmacokinetics of the H(1) receptor antagonist diphenhydramine (DPHM) were studied in 100- and 120-day-old fetuses, 10- and 30-day-old newborn lambs, and adult sheep using in vivo microdialysis. DPHM was administered i.v. at five infusion rates, with each step lasting 7 h. In all ages, cerebrospinal fluid (CSF) and extracellular fluid (ECF) concentrations were very similar to each other, which suggests that DPHM between these two compartments is transferred by passive diffusion. In addition, the brain-to-plasma concentration ratios were >or=3 in all age groups, suggesting the existence of a transport process for DPHM into the brain. Both brain and plasma DPHM concentrations increased in a linear fashion over the dose range studied. However, the ECF/unbound plasma and CSF/unbound plasma DPHM concentration ratios were significantly higher in the fetus and lambs (approximately 5 to 6) than in the adult (approximately 3). The factors f(CSF) and f(ECF), the ratios of DPHM areas under the curves (AUCs) in CSF and ECF to the plasma DPHM AUC, respectively, decreased with age, indicating that DPHM is more efficiently removed from the brain with increasing age. The extent of plasma protein binding of the drug increased with age. This study provides evidence for a transporter-mediated mechanism for the influx of DPHM into the brain and also for an efflux transporter for the drug, whose activity increases with age. Moreover, the higher brain DPHM levels in the fetus and lamb compared with the adult may explain the greater CNS effects of the drug at these ages.


Subject(s)
Central Nervous System/metabolism , Diphenhydramine/pharmacokinetics , Microdialysis/methods , Age Factors , Animals , Animals, Newborn , Area Under Curve , Blood Proteins/metabolism , Cerebrospinal Fluid/metabolism , Diphenhydramine/blood , Diphenhydramine/cerebrospinal fluid , Extracellular Fluid/metabolism , Female , Fetus/metabolism , Histamine H1 Antagonists/administration & dosage , Histamine H1 Antagonists/pharmacokinetics , Infusions, Intravenous , Sheep
7.
Drug Metab Dispos ; 34(6): 955-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16510542

ABSTRACT

The purpose of this study was to examine the disposition of diphenhydramine (DPHM) across the ovine blood-brain barrier (BBB). In six adult sheep, we characterized the central nervous system (CNS) pharmacokinetics of DPHM in brain extracellular fluid (ECF) and cerebrospinal fluid (CSF) using microdialysis in two experiments. In the first experiment, DPHM was administered via a five-step i.v. infusion (1.5, 5.5, 9.5, 13.5, and 17.5 microg/kg/min; 7 h per step). Average steady-state CNS/total plasma concentration ratios (i.e., [CNS]/[total plasma]) for steps 1 to 5 ranged from 0.4 to 0.5. However, average steady-state [CNS]/[free plasma] ratios ranged from 2 to 3, suggesting active transport of DPHM into the CNS. Plasma protein binding averaged 86.1 +/- 2.3% (mean +/- S.D.) and was not altered with increasing drug dose. Plasma, CSF, and ECF demonstrated biexponential pharmacokinetics with terminal elimination half-lives (t1/2beta) of 10.8 +/- 5.4, 3.6 +/- 1.0, and 5.3 +/- 4.2 h, respectively. The bulk flow of CSF and transport-mediated efflux of DPHM may explain the observed higher CNS clearances. In the second experiment, DPHM was coadministered with propranolol (PRN) to examine its effect on blood-brain CSF and blood-brain ECF DPHM relationships. Plasma total DPHM concentration decreased by 12.8 +/- 6.3% during PRN, whereas ECF and CSF concentrations increased (88.1 +/- 45.4 and 91.6 +/- 34.3%, respectively). This increase may be due to the inhibitory effect of PRN on a transporter-mediated efflux mechanism for DPHM brain elimination.


Subject(s)
Blood-Brain Barrier/metabolism , Diphenhydramine/pharmacokinetics , Histamine H1 Antagonists/pharmacokinetics , Animals , Biological Transport, Active/drug effects , Diphenhydramine/administration & dosage , Diphenhydramine/cerebrospinal fluid , Extracellular Fluid/metabolism , Female , Half-Life , Histamine H1 Antagonists/administration & dosage , Histamine H1 Antagonists/cerebrospinal fluid , Infusions, Intravenous , Microdialysis , Propranolol/pharmacology , Sheep
8.
Br J Clin Pharmacol ; 61(2): 155-63, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16433870

ABSTRACT

AIMS: To compare the disposition of fluoxetine and norfluoxetine enantiomers in the mother, foetus and infant. METHODS: Blood from pregnant women taking fluoxetine (n = 9), during pregnancy was sampled in the third trimester and at delivery (maternal and cord venous blood), and from the infants 48 h after delivery. The subset of these women who were breastfeeding, plus additional subjects recruited in the postpartum period, were studied further, and maternal and infant blood, and breast milk was sampled between 6 days and 11 months (n = 23). Drug and metabolite concentrations were measured using gas chromatography/mass spectrometry or liquid chromatography, tandem mass spectrometry. RESULTS: There was a high correlation between maternal and foetal (cord blood) fluoxetine and norfluoxetine enantiomers (r(2)-0.9), the mean foetal/maternal ratios (95% confidence intervals) being 0.91 (0.61, 1.02) and 1.04 (0.93, 1.05), for fluoxetine and norfluoxetine, respectively. In 2 day old infants exposed to the drug in utero, the fluoxetine and norfluoxetine plasma concentrations were the same as in cord blood at delivery. Over the next 2 months, the plasma concentrations in the infants fell progressively. Stereoselective disposition of both the drug and metabolite in the mother, foetus, infant and breast milk was observed. The S : R ratios in the foetus and newborn ( approximately 3) were significantly higher than in the serum ( approximately 2) or breast milk ( approximately 1.9) of the mothers, resulting in greater exposure of the foetus and infants to the biologically active enantiomers, particularly S-norfluoxetine. CONCLUSIONS: Foetal and infant exposure to fluoxetine and norfluoxetine is enhanced by their stereoselective disposition in the mother, foetus, breast milk and infant. Increased exposure may also result from decreased metabolism of the drug in the foetus and neonate.


Subject(s)
Antidepressive Agents, Second-Generation/blood , Breast Feeding , Depressive Disorder, Major/blood , Fluoxetine/blood , Maternal-Fetal Exchange , Pregnancy Complications/blood , Adult , Aging/blood , Antidepressive Agents, Second-Generation/therapeutic use , Depressive Disorder, Major/drug therapy , Female , Fetal Blood/metabolism , Fluoxetine/analogs & derivatives , Fluoxetine/therapeutic use , Humans , Infant, Newborn , Milk, Human/chemistry , Postpartum Period/blood , Pregnancy , Pregnancy Complications/drug therapy , Prospective Studies , Stereoisomerism
9.
Reprod Fertil Dev ; 17(6): 641-50, 2005.
Article in English | MEDLINE | ID: mdl-16263070

ABSTRACT

Women are at greatest risk of suffering from depression during the childbearing years and thus may either become pregnant while taking an antidepressant or may require a prescription for one during pregnancy. The antidepressant fluoxetine (FX) is a selective serotonin reuptake inhibitor (SSRI), which increases serotonin neurotransmission. Serotonin is involved in the regulation of a variety of physiological systems, including the sleep-wake cycle, circadian rhythms and the hypothalamic-pituitary-adrenal axis. Each of these systems also plays an important role in fetal development. Compared with other antidepressant drugs, the SSRIs, such as FX, have fewer side effects. Because of this, they are now frequently prescribed, especially during pregnancy. Clinical studies suggest poor neonatal outcome after exposure to FX in utero. Recent studies in the sheep fetus describe the physiological effects of in utero exposure to FX with an 8 day infusion during late gestation in the sheep. This is a useful model for determining the effects of FX on fetal physiology. The fetus can be studied for weeks in its normal intrauterine environment with serial sampling of blood, thus permitting detailed studies of drug disposition in both mother and fetus combined with monitoring of fetal behavioural state and cardiovascular function. Fluoxetine causes an acute increase in plasma serotonin levels, leading to a transient reduction in uterine blood flow. This, in turn, reduces the delivery of oxygen and nutrients to the fetus, thereby presenting a mechanism for reducing growth and/or eliciting preterm delivery. Moreover, because FX crosses the placenta, the fetus is exposed directly to FX, as well as to the effects of the drug on the mother. Fluoxetine increases high-voltage/non-rapid eye movement behavioural state in the fetus after both acute and chronic exposure and, thus, may interfere with normal fetal neurodevelopment. Fluoxetine also alters hypothalamic function in the adult and increases the magnitude of the prepartum rise in fetal cortisol concentrations in sheep. Fetal FX exposure does not alter fetal circadian rhythms in melatonin or prolactin. Studies of the effects of FX exposure on fetal development in the sheep are important in defining possible physiological mechanisms that explain human clinical studies of birth outcomes after FX exposure. To date, there have been insufficient longer-term follow-up studies in any precocial species of offspring exposed to SSRIs in utero. Thus, further investigation of the long-term consequences of in utero exposure to FX and other SSRIs, as well as the mechanisms involved, are required for a complete understanding of the impact of these agents on development. This should involve studies in both humans and appropriate animal models.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Fetal Development/drug effects , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Animals , Antidepressive Agents, Second-Generation/adverse effects , Antidepressive Agents, Second-Generation/pharmacology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Depression/drug therapy , Disease Models, Animal , Female , Fluoxetine/adverse effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/embryology , Male , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/embryology , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Complications/psychology , Prenatal Exposure Delayed Effects , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sheep , Teratogens/toxicity
10.
J Soc Gynecol Investig ; 12(5): 356-64, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15979548

ABSTRACT

OBJECTIVE: Depression during pregnancy is frequently treated with the selective serotonin reuptake inhibitor (SSRI), fluoxetine (FX), commonly known as Prozac (Eli Lilly & Co, Indianapolis, IN). FX potentiates serotoninergic neurotransmission and serotonin has been implicated in the regulation of circadian rhythms. We have therefore investigated the effect of chronic administration of FX on maternal and fetal circadian rhythms in sheep. METHODS: Following an initial bolus dose of 70 mg FX, an 8-day continuous infusion of FX (n = 11, 98.5 microg/kg x d) was performed. Controls (n = 13) were treated with sterile water vehicle only. Maternal and fetal plasma melatonin and prolactin concentrations were determined every 3 hours for 24 hours and then every 6 hours for 24 hours beginning on the fourth day of infusion. RESULTS: FX treatment did not alter either the basal or circadian rhythms of either maternal or fetal plasma melatonin and prolactin concentrations. Fetal cardiovascular and behavioral state parameters were measured continuously. While the incidence of low-voltage (LV) electrocortical (ECOG) activity was significantly reduced in fetuses in the FX group, there was no effect of FX on the diurnal rhythms in fetal arterial pressure, heart rate, breathing movements, or behavioral state. CONCLUSION: These results show that maternal FX treatment does not result in significant alterations in maternal and fetal hormonal and behavioral circadian rhythms.


Subject(s)
Circadian Rhythm/drug effects , Fluoxetine/pharmacology , Pregnancy, Animal/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Blood Pressure , Electrocardiography , Female , Fluoxetine/administration & dosage , Heart Rate , Infusions, Intravenous , Injections, Intravenous , Maternal-Fetal Exchange , Melatonin/blood , Pregnancy , Prolactin/blood , Respiration , Selective Serotonin Reuptake Inhibitors/administration & dosage , Sheep
11.
Pediatr Res ; 56(1): 40-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15128928

ABSTRACT

Depression during pregnancy is frequently treated with the selective serotonin reuptake inhibitor, fluoxetine (FX). FX increases serotonergic neurotransmission and serotonin plays a role in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis. We have therefore investigated the effect of chronic administration of FX to the pregnant ewe on the maternal and fetal HPA axes. Nineteen late-gestation sheep were surgically prepared for chronic study of the fetus. FX (n = 7, 98.5 microg/kg/d) or sterile water (control, n = 8) was administered to the ewe for 8 d by constant rate i.v. infusion with an initial FX bolus dose of 70 mg. Maternal and fetal plasma ACTH and cortisol concentrations were determined at 0700 h each day. Maternal plasma ACTH concentrations fell on infusion d 2, but no changes were observed in maternal plasma cortisol concentrations. Fetal plasma ACTH concentrations increased on infusion d 7, and fetal plasma cortisol concentrations increased on infusion d 6, 7, and 8 in the FX group. In addition, the regression coefficient for the relationship between fetal ACTH and cortisol levels was significantly greater in the FX group compared with the control group. Thus, maternal FX treatment increased fetal plasma cortisol concentration. These results are of particular interest in the context that exposure of the fetus to excess glucocorticoids at critical windows during development has been shown to increase the risk of poor health outcomes in later life.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluoxetine/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Adrenocorticotropic Hormone/blood , Animals , Antidepressive Agents, Second-Generation/blood , Carbon Dioxide/blood , Female , Fetus/drug effects , Fluoxetine/blood , Gestational Age , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/embryology , Oxygen/blood , Pituitary-Adrenal System/embryology , Pregnancy , Sheep
12.
Drug Metab Dispos ; 32(2): 212-21, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14744943

ABSTRACT

We examined the stereoselective disposition of fluoxetine (FX) and its metabolite norfluoxetine (NFX) in five pregnant sheep. Racemic FX was administered i.v. to the ewe (50 mg) and the fetus (10 mg) on separate occasions. Maternal and fetal blood, maternal urine, and fetal amniotic and tracheal fluid samples were collected for 72 h. FX and NFX isomers were quantified by gas chromatography-mass spectrometry. They rapidly crossed the placenta [maternal to fetal area under the plasma concentration versus time curve (AUC) ratios 0.59 and 0.65, respectively]. There was significant FX stereoselectivity with S/R FX AUC ratios averaging 1.65 +/- 0.33 and 1.73 +/- 0.29 in ewe and fetus, respectively, after maternal dosing. The maternal clearance and volume of distribution were also higher for (R)-fluoxetine than for (S)-fluoxetine. FX, NFX, and their glucuronides were present in maternal urine but accounted for only 3.4% of maternal drug elimination. In contrast, NFX was not detected in the fetus after fetal FX administration, which is consistent with the absence of measurable fetal nonplacental clearance of the drug and the lack of NFX formation in fetal hepatic microsomal incubations. There was also no fetal production of FX and NFX glucuronides in vivo. Both FX and NFX were extensively and stereoselectively bound in maternal and fetal plasma, with the free fraction S/R FX ratio averaging 0.46 +/- 0.06 and 0.58 +/- 0.10 in ewe and fetus, respectively. Thus, FX exhibits extensive stereoselective disposition, which is likely due to differential plasma protein binding of the FX isomers, and there is no detectable fetal formation of NFX, FX, and NFX glucuronides.


Subject(s)
Fetus/metabolism , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacokinetics , Pregnancy, Animal/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Animals , Area Under Curve , Cytochrome P-450 Enzyme System/metabolism , Female , Fluoxetine/blood , Fluoxetine/urine , Half-Life , In Vitro Techniques , Maternal-Fetal Exchange , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Pregnancy , Selective Serotonin Reuptake Inhibitors/blood , Selective Serotonin Reuptake Inhibitors/urine , Sheep , Stereoisomerism , Time Factors
13.
Arch Pharm Res ; 25(5): 709-17, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12433210

ABSTRACT

Hypoxemia is known to induce various physiological changes which can result in alteration in drug pharmacokinetics. To examine the effect of acute moderate hypoxemia on metoclopramide (MCP) pharmacokinetics, a continuous 14-hour infusion of MCP during a normoxemic, hypoxemic and subsequent normoxemic period was conducted in eight adult sheep. Arterial blood and urine samples were collected to examine the effects on the pharmacokinetics of MCP and its deethylated metabolites. MCP and its mono- and di-deethylated metabolites were quantitated using a GC/MS method. Steady-state concentrations of MCP were achieved in each of the three periods. During hypoxemia, MCP plasma steady-state concentration increased significantly from 50.72 +/- 1.06 to 63.62 +/- 1.79 ng/mL, and later decreased to 55.83 +/- 1.15 ng/mL during the post-hypoxemic recovery period. Total body clearance (CL(TB)) of MCP was significantly decreased from 274.2 +/- 48.0 L/h to 205.40 +/- 28.2 L/h during hypoxemia, and later restored to 245.8 +/- 44.2 L/h during the post-hypoxemic period. Plasma mono-deethylated MCP concentration (32.78 +/- 1.73 ng/mL) also increased, compared to the control group (21.20 +/- 1.39 ng/mL), during hypoxemia and subsequent normoxemic period. Renal excretion of MCP and its metabolites was also decreased during hypoxemia, while urine flow was increased with a concomitant decrease in urine osmolality. Thus, the results indicate that acute moderate hypoxemia affects MCP pharmacokinetics.


Subject(s)
Hypoxia/metabolism , Metoclopramide/pharmacokinetics , Sheep/metabolism , Acute Disease , Animals , Female , Metabolic Clearance Rate , Metoclopramide/metabolism
14.
J Pharm Pharmacol ; 54(6): 801-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12078996

ABSTRACT

Indometacin is used in pregnancy for the treatment of premature labour, but there are limited data on the disposition of the drug in the fetus. In order to elucidate fetal indometacin pharmacokinetics at plasma levels and duration comparable with those occurring with use of the drug for tocolysis in humans, indometacin was administered at doses of 1.9 (low dose, LD; n = 5) or 7.5 (high dose, HD; n = 9) microg min(-1) to steady state over a 3-day period in chronically instrumented fetal lambs. Indometacin concentrations in biological fluid samples were analysed by a sensitive capillary gas chromatography-electron capture detection method. The mean steady-state fetal arterial plasma indometacin concentrations were 68.6+/-16.5 ng mL(-1) in the LD infusion and 230.3+/-28.8 ng mL(-1) in the HD infusion. Indometacin concentrations in amniotic fluid were approximately 10% of those in fetal plasma, and below assay detection limits in tracheal fluid. Total body clearance (TBC) in the LD and HD infusions were not different and the overall mean was 11.3+/-1.2 mL min(-1) kg(-1). In the 11 experiments where paired fetal arterial and umbilical venous samples were collected, the extraction of indometacin across the placenta averaged only 5.2+/-1.1%, indicating low placental permeability to the drug in sheep. However, fetal placental clearance (CLpl) of indometacin (10.0+/-2.5 mL min(-1) kg(-1), n = 10) averaged 115.1+/-41.2% of TBC in these animals and the calculated value for fetal non-placental clearance (0.6+/-2.8 mL min(-1) kg(-1)) was not significantly different from zero. Fetal renal clearance of intact indometacin (3.8+/-1.1 microL min(-1) kg(-1); n = 12) was also very low. However, treatment of fetal urine with glucuronidase indicated the presence of glucuronide conjugates and these comprised 69.9+/-8.2% of the total drug concentration (i.e. intact+conjugated) in urine. Thus, the fetal lamb appears to be able to glucuronidate indometacin, but the contribution of this and other non-placental routes to overall fetal elimination of the drug appear minimal. CLpl of the drug is also low owing to the physicochemical properties of indometacin (high polarity) and the permeability characteristics of the sheep placenta.


Subject(s)
Fetus/metabolism , Indomethacin/pharmacokinetics , Placenta/metabolism , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Female , Indomethacin/administration & dosage , Indomethacin/urine , Infusions, Intravenous , Maternal-Fetal Exchange , Metabolic Clearance Rate , Pregnancy , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...