Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Clin Pharmacol Ther ; 60(9): 408-416, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35818823

ABSTRACT

OBJECTIVES: To evaluate the pharmacokinetics, pharmacodynamics (PD), safety, and tolerability of epidural SP-102 (10 mg dexamethasone sodium phosphate injectable gel) compared to an intravenous injection of 10 mg dexamethasone sodium phosphate, USP (IV USP). MATERIALS AND METHODS: Subjects with lumbosacral radiculopathy received a single dose of epidural SP-102, followed by a single dose of IV USP 4 weeks later. Dexamethasone plasma levels, cortisol levels, white blood cells (WBC), and blood glucose levels were assessed. RESULTS: Twelve subjects entered and completed the study. The mean total dexamethasone exposure (AUClast and AUCinf) following SP-102 by epidural injection was equivalent to the total exposure following IV USP. A lower mean plasma Cmax (~ 50% lower) was observed following epidural administration compared to IV injection. PD parameters were similar between treatments. Adverse events (AEs) were mild, with no serious AEs or study discontinuations due to AEs. CONCLUSION: In this small study, epidural SP-102 injection was well tolerated, was not associated with greater systemic dexamethasone exposure than IV USP, and both treatments had similar PD effects on cortisol suppression, blood glucose, and WBC levels.


Subject(s)
Blood Glucose , Hydrocortisone , Dexamethasone/adverse effects , Dexamethasone/analogs & derivatives , Humans , Pain
2.
J Urol ; 190(2): 799-806, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23321580

ABSTRACT

PURPOSE: We evaluated the effects of the highly selective phosphodiesterase type 5 inhibitor avanafil on electroretinogram and hemodynamics in dogs, and compared the effects with those of sildenafil. MATERIALS AND METHODS: Three experiments were performed in anesthetized dogs, including determination of the 1) influence on electroretinogram induced by a light adapted 30 Hz flicker stimulation, 2) direct hemodynamic changes and 3) potentiation of nitroglycerin induced hypotension. Avanafil was administered at doses that were pharmacologically equipotent to or higher than those of sildenafil for penile tumescence. RESULTS: 1) Intraduodenal doses of avanafil did not influence the electroretinogram waveform. In contrast, sildenafil changed the waveform shape and significantly delayed time to the peak of the electroretinogram positive waveform (vs vehicle p <0.05). 2) Intravenous infusion of avanafil or sildenafil (1 to 300 µg/kg per minute) significantly decreased systemic blood pressure, total peripheral resistance and pulmonary arterial pressure (vs vehicle p <0.05). Administration of sildenafil but not avanafil significantly decreased the resistance of common carotid and vertebral arteries (vs vehicle p <0.05). 3) Intraduodenal doses of avanafil or sildenafil (0.1 and 1 mg/kg) potentiated the AUC of nitroglycerin induced hypotension. However, the potentiating effect of avanafil at 1 mg/kg was significantly weaker than that of sildenafil (p <0.05). CONCLUSIONS: Data suggest that avanafil has a favorable safety profile for erectile dysfunction, which is attributable to its high inhibitory selectivity for phosphodiesterase type 5 against type 6 (retina) and 1 (vessels, etc), respectively, and its short acting pharmacodynamic property.


Subject(s)
Hemodynamics/drug effects , Hypotension/chemically induced , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrimidines/pharmacology , Retina/drug effects , Analysis of Variance , Animals , Area Under Curve , Dogs , Electroretinography , Erectile Dysfunction/drug therapy , Male , Piperazines/pharmacology , Purines/pharmacology , Signal Transduction , Sildenafil Citrate , Sulfones/pharmacology
3.
J Sex Med ; 9(8): 2122-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22759639

ABSTRACT

INTRODUCTION: Phosphodiesterase type 5 (PDE5) inhibitors are indicated for the treatment of erectile dysfunction (ED); however, they can also inhibit other PDE isozymes, affecting their target tissues (e.g., PDE1: heart; PDE6: retina; and PDE11: skeletal muscle), which in some cases can cause unwanted side effects and therapy discontinuation. Data from in vitro studies showed that avanafil, a PDE5 inhibitor for the treatment of ED, exhibited strong selectivity toward PDE5 and against all other PDE isozymes. AIM: To review the inhibitory effects of avanafil for PDE isozymes compared with those of sildenafil, tadalafil, and vardenafil and to discuss these results within the context of clinical trial safety observations. METHODS: Review of in vitro selectivity data for avanafil (published primary data from a peer-reviewed journal and scientific congress abstracts); PubMed search for pertinent publications on PDE5 inhibitor safety data; and review of published articles and abstracts from avanafil phase 1, 2, and 3 clinical trials. MAIN OUTCOME MEASURES: A low incidence of some PDE-related adverse events may be reflected by the high selectivity of avanafil against non-PDE5 isozymes. RESULTS: Avanafil is highly selective toward PDE5 and against all other PDE isozymes tested. Lower selectivity against PDE1, PDE6, and PDE11 is consistent with results from randomized, placebo-controlled, phase 3 trials in which musculoskeletal and hemodynamic adverse events were reported in <2% of patients and no color vision-related abnormalities were reported with avanafil doses up to 200 mg once daily. CONCLUSIONS: Data suggest that avanafil may confer a safety benefit, in terms of a lower incidence of specific adverse events, by virtue of its high specificity to PDE5 and its overall selectivity against other PDE isozymes.


Subject(s)
Erectile Dysfunction/drug therapy , Phosphodiesterase 5 Inhibitors/adverse effects , Phosphodiesterase 5 Inhibitors/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Carbolines/adverse effects , Carbolines/therapeutic use , Humans , Imidazoles/adverse effects , Imidazoles/therapeutic use , Male , Penile Erection/drug effects , Piperazines/adverse effects , Piperazines/therapeutic use , Purines/adverse effects , Purines/therapeutic use , Randomized Controlled Trials as Topic , Sildenafil Citrate , Sulfones/adverse effects , Sulfones/therapeutic use , Tadalafil , Triazines/adverse effects , Triazines/therapeutic use , Vardenafil Dihydrochloride
4.
J Urol ; 188(2): 668-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22704456

ABSTRACT

PURPOSE: We investigated the in vitro inhibitory effects of avanafil, a novel, potent inhibitor of phosphodiesterase-5, on 11 phosphodiesterases. We also studied its potentiation of penile tumescence in dogs. MATERIALS AND METHODS: Phosphodiesterase assay was done with the 4 phosphodiesterase-5 inhibitors avanafil, sildenafil, vardenafil and tadalafil using 11 phosphodiesterase isozymes. In anesthetized dogs the pelvic nerve was repeatedly stimulated to evoke tumescence. Intracavernous pressure was measured after avanafil or sildenafil administration. RESULTS: Avanafil specifically inhibited phosphodiesterase-5 activity at a 50% inhibitory concentration of 5.2 nM. Avanafil showed higher selectivity (121-fold) against phosphodiesterase-6 than sildenafil and vardenafil (16 to 21-fold) and showed excellent selectivity (greater than 10,000-fold) against phosphodiesterase-1 compared with sildenafil (375-fold). Avanafil also had higher selectivity against phosphodiesterase-11 than tadalafil (greater than 19,000 vs 25-fold). Avanafil also showed excellent selectivity against all other phosphodiesterases. After intravenous administration in anesthetized dogs the 200% effective dose of avanafil and sildenafil on the penile tumescence was 37.5 and 34.6 µg/kg, respectively. After intraduodenal administration the 200% effective dose of avanafil and sildenafil on tumescence was 151.7 and 79.0 µg/kg at the peak time, respectively. Time to peak response with avanafil and sildenafil was 10 and 30 minutes, respectively, indicating a more rapid onset of avanafil. CONCLUSIONS: Avanafil has a favorable phosphodiesterase-5 selectivity profile compared to that of marketed phosphodiesterase-5 inhibitors. Avanafil shows excellent in vitro and in vivo potency, and fast onset of action for penile erection. Cumulative data suggest that avanafil has a promising pharmacological profile for erectile dysfunction.


Subject(s)
Erectile Dysfunction/physiopathology , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Carbolines/pharmacology , Dogs , Dose-Response Relationship, Drug , Humans , Imidazoles/pharmacology , Infusions, Intravenous , Male , Penile Erection/drug effects , Piperazines/pharmacology , Purines/pharmacology , Sildenafil Citrate , Sulfones/pharmacology , Tadalafil , Triazines/pharmacology , Vardenafil Dihydrochloride
5.
J Clin Pharmacol ; 49(3): 360-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19246733

ABSTRACT

Intestinal peptide transporters, including hPEPT1, facilitate the absorption of cephalosporins and angiotensin-converting enzyme inhibitors, and have been investigated as a means to improve oral drug absorption. Renal peptide transporters including hPEPT2, may also facilitate renal reabsorption of such compounds. In vitro and animal studies suggest that co-administration of peptidomimetic compounds may alter oral pharmacokinetics, although this has not been well studied in humans. The purpose of this study was to determine whether co-administration of the hPEPT substrates captopril and cephradine alters the oral pharmacokinetics of either agent. Nine healthy male volunteers received a single oral 25-mg dose of captopril, a single oral 500-mg dose of cephradine, or concurrent ingestion of captopril and cephradine in a cross-over manner. Venous blood samples were taken and captopril and cephradine pharmacokinetics were determined using noncompartmental analyses. No significant differences were observed in captopril or cephradine pharmacokinetics when administered together as compared to each agent alone (a marginal decrease in C(max) was observed for both captopril and cephradine during co-administration [5-15%]; however, differences were not statistically significant). The results of our study suggest that hPEPT1 and hPEPT2 are unlikely to contribute to clinically important drug interactions in humans.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology , Captopril/pharmacology , Cephradine/pharmacology , Intestinal Absorption/drug effects , Symporters/physiology , Adult , Angiotensin-Converting Enzyme Inhibitors/metabolism , Anti-Bacterial Agents/metabolism , Biological Transport , Captopril/metabolism , Cephradine/metabolism , Cross-Over Studies , Drug Interactions , Humans , Kidney/metabolism , Male , Peptide Transporter 1
6.
J Clin Invest ; 118(6): 2337-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18483622

ABSTRACT

Retinal and choroidal vascular diseases, with their associated abnormalities in vascular permeability, account for the majority of patients with vision loss in industrialized nations. VEGF is upregulated in ischemic retinopathies such as diabetes and is known to dramatically alter vascular permeability in a number of nonocular tissues via Src kinase-regulated signaling pathways. VEGF antagonists are currently in clinical use for treating the new blood vessels and retinal edema associated with neovascular eye diseases, but such therapies require repeated intraocular injections. We have found that vascular leakage following intravitreal administration of VEGF in mice was abolished by systemic or topical delivery of what we believe is a novel VEGFR2/Src kinase inhibitor; this was confirmed in rabbits. The relevance of Src inhibition to VEGF-associated alterations in vascular permeability was further substantiated by genetic studies in which VEGF injection or laser-induced vascular permeability failed to augment retinal vascular permeability in Src-/- and Yes-/- mice (Src and Yes are ubiquitously expressed Src kinase family members; Src-/- and Yes-/- mice lacking expression of these kinases show no vascular leak in response to VEGF). These findings establish a role for Src kinase in VEGF-mediated retinal vascular permeability and establish a potentially safe and painless topically applied therapeutic option for treating vision loss due to neovascular-associated retinal edema.


Subject(s)
Capillary Permeability , Enzyme Inhibitors/pharmacology , Retina/pathology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Biological , Permeability , Rabbits , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/metabolism
7.
J Med Chem ; 51(6): 1546-59, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18311895

ABSTRACT

Age-related macular degeneration (AMD) is one of the leading causes of loss of vision in the industrialized world. Attenuating the VEGF signal in the eye to treat AMD has been validated clinically. A large body of evidence suggests that inhibitors targeting the VEGFr pathway may be effective for the treatment of AMD. Recent studies using Src/YES knockout mice suggest that along with VEGF, Src and YES play a crucial role in vascular leak and might be useful in treating edema associated with AMD. Therefore, we have developed several potent benzotriazine inhibitors designed to target VEGFr2, Src, and YES. One of the most potent compounds is 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-1-yl-ethoxy)phenylamino]benzo[1,2,4]triazin-7-yl}phenol ( 5), a dual inhibitor of both VEGFr2 and the Src family (Src and YES) kinases. Several ester analogues of 5 were prepared as prodrugs to improve the concentration of 5 at the back of the eye after topical administration. The thermal stability of these esters was studied, and it was found that benzoyl and substituted benzoyl esters of 5 showed good thermal stability. The hydrolysis rates of these prodrugs were studied to analyze their ability to undergo conversion to 5 in vivo so that appropriate concentrations of 5 are available in the back-of-the-eye tissues. From these studies, we identified 4-chloro-3-(5-methyl-3-{[4-(2-pyrrolidin-1-ylethoxy)phenyl]amino}-1,2,4-benzotriazin-7-yl)phenyl benzoate ( 12), a topically administered prodrug delivered as an eye drop that is readily converted to the active compound 5 in the eye. This topically delivered compound exhibited excellent ocular pharmacokinetics and poor systemic circulation and showed good efficacy in the laser induced choroidal neovascularization model. On the basis of its superior profile, compound 12 was advanced. It is currently in a clinical trial as a first in class, VEGFr2 targeting, topically applied compound for the treatment of AMD.


Subject(s)
Macular Degeneration/drug therapy , Ophthalmic Solutions/therapeutic use , Phenols/therapeutic use , Prodrugs/therapeutic use , Triazines/therapeutic use , Administration, Topical , Animals , Choroidal Neovascularization/drug therapy , Clinical Trials as Topic , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Eye/drug effects , Eye/radiation effects , Lasers , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacokinetics , Phenols/chemistry , Phenols/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacokinetics , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors
8.
Drug Metab Dispos ; 35(12): 2242-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881660

ABSTRACT

[7-(2,6-Dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-amine (TG100435) is a novel multi-targeted Src family kinase inhibitor with demonstrated anticancer activity in preclinical species. Potent kinase inhibition is associated with TG100435 and its major N-oxide metabolite [7-(2,6-dichlorophenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-{4-[2-(1-oxy-pyrrolidin-1-yl)-ethoxy]-phenyl}-amine (TG100855). The objectives of the current study were to identify the hepatic enzyme(s) responsible for 1) the total metabolic flux of TG100435, 2) the formation of TG100855, and 3) the subsequent metabolism of TG100855. Flavin-containing monooxygenases (FMO) and cytochrome P450 monooxygenases (P450s) contribute to TG100435 total metabolic flux. TG100435 metabolic flux was completely inhibited by methimazole and ketoconazole, suggesting only FMO- and CYP3A4-mediated metabolism. TG100855 formation was markedly inhibited (~90%) by methimazole or heat inactivation (>99%). FMO3 was the primary enzyme responsible for TG100855 formation. In addition, an enzyme mediated retroreduction of TG100855 back to TG100435 was observed. The N-oxidation reaction was approximately 15 times faster than the retroreduction reaction. Interestingly, the retroreduction of TG100855 to TG100435 in recombinant P450 or liver microsomes lacked inhibition by the P450 inhibitors. TG100435 formation in the human liver microsomes or recombinant P450 increased as a function of cytochrome P450 reductase activity, suggesting potential involvement of cytochrome P450 reductase. The results of this in vitro study demonstrate the potential of TG100435 and TG100855 to be interconverted metabolically. FMO seem to be the major N-oxidizing enzymes, whereas cytochrome P450 reductase seems to be responsible for the retroreduction reaction.


Subject(s)
Microsomes, Liver/enzymology , NADPH-Ferrihemoprotein Reductase/metabolism , Oxygenases/metabolism , Protein Kinase Inhibitors/metabolism , Pyrrolidines/metabolism , Triazines/metabolism , src-Family Kinases/antagonists & inhibitors , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Stability , Humans , In Vitro Techniques , Ketoconazole/pharmacology , Kinetics , Methimazole/pharmacology , Microsomes, Liver/drug effects , Oxidation-Reduction , Oxygenases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrrolidines/pharmacology , Recombinant Proteins/metabolism , Temperature , Triazines/pharmacology , src-Family Kinases/metabolism
9.
J Med Chem ; 50(18): 4279-94, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17685602

ABSTRACT

In studies aimed toward identifying effective and safe inhibitors of kinase signaling cascades that underlie ischemia/reperfusion (I/R) injury, we synthesized a series of pteridines and pyridopyrazines. The design strategy was inspired by the examination of naturally occurring PI3K inhibitors such as wortmannin and quercetin, and building a pharmacophore-based model used for optimization. Structural modifications led to hybrid molecules which incorporated aminopyrimidine and aminopyridine moieties with ATP mimetic characteristics into the pharmacophore motifs to modulate kinase affinity and selectivity. Elaborations involving substitutions of the 2 and 4 positions of the pyrimidine or pyridine ring and the 6 and 7 positions of the central pyrazine ring resulted in in vivo activity profiles which identified potent inhibitors of vascular endothelial growth factor (VEGF) induced vascular leakage. Pathway analysis identified a diaminopteridine-diphenol as a potent and selective phosphatidylinositol-3-kinase (PI3K) inhibitor. The structure-activity relationship studies of various analogues of diaminopteridine-diphenol-based on biochemical assays resulted in potent inhibitors of PI3K.


Subject(s)
Myocardial Infarction/complications , Myocardial Reperfusion Injury/drug therapy , Phenols/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Pteridines/chemical synthesis , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Cell Proliferation , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Male , Models, Molecular , Myocardial Reperfusion Injury/etiology , Phenols/pharmacokinetics , Phenols/pharmacology , Phosphorylation , Pteridines/pharmacokinetics , Pteridines/pharmacology , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology
10.
Drug Metab Dispos ; 35(6): 929-36, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17371799

ABSTRACT

TG100435 ([7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-amine) is a novel multitargeted, orally active protein tyrosine kinase inhibitor. The inhibition constants (K(i)) of TG100435 against Src, Lyn, Abl, Yes, Lck, and EphB4 range from 13 to 64 nM. TG100435 has systemic clearance values of 20.1, 12.7, and 14.5 ml/min/kg and oral bioavailability of 74%, 23%, and 11% in mouse, rat, and dog, respectively. Four oxidation metabolites of TG100435 have been found in human, dog, and rat in vitro and in vivo. The ethylpyrrolidine N-oxide of TG100435 is the predominant metabolite (TG100855; [7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-{4-[2-(1-oxy-pyrrolidin-1-yl)-ethoxy]-phenyl}-amine) in human, dog, and rat. TG100855 is 2 to 9 times more potent than the parent compound. Flavin-containing monooxygenases are the primary enzymes mediating the biotransformation. Significant conversion of TG100435 to TG100855 has been observed in rat and dog after oral administration. Systemic exposure of TG100855 is 1.1- and 2.1-fold greater than that of TG100435 in rat and dog after oral dosing of TG100435. Since TG100435 is predominantly converted to the more potent N-oxide metabolite across species in vivo and in vitro, the overall tyrosine kinase inhibition in animal models may be substantially increased after oral administration of TG100435.


Subject(s)
Protein Kinase Inhibitors/pharmacokinetics , Pyrrolidines/blood , Pyrrolidines/pharmacokinetics , Triazines/blood , Triazines/pharmacokinetics , Animals , Biotransformation , Dogs , Female , Male , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Protein Kinase Inhibitors/blood , Rats , Rats, Sprague-Dawley , src-Family Kinases/antagonists & inhibitors
11.
Bioorg Med Chem Lett ; 17(3): 602-8, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17113292
12.
Drug Metab Dispos ; 33(11): 1688-99, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16081672

ABSTRACT

The absorption, metabolism, and excretion of N-[3-fluoro-4-[2-(propylamino)ethoxy]phenyl]-4,5,6,7-tetrahydro-4-oxo-1H-indole-3-carboxamide monomethanesulfonate (1), a GABAA receptor partial agonist potentially useful in treating generalized anxiety disorder, have been evaluated in both Sprague-Dawley rats and cynomolgus monkeys using [14C]1. In both species, mass balance was achieved within 48 h postdose, with the majority of drug-related material excreted within the feces; the clearance of 1 in each species had both metabolic and renal components. In addition to the metabolites produced by aliphatic hydroxylation and/or N-dealkylation of 1, two unique metabolites were detected: a putative carbamic acid (M7) in rat plasma and monkey bile, and an N-carbamoyl glucuronide (M8) in both rat and monkey bile. Metabolite M8 was structurally deciphered by liquid chromatographytandem mass spectrometry and NMR, and was readily generated in vitro upon incubation of [14C]1 with rat liver microsomes fortified with uridine 5'-diphosphoglucuronic acid trisodium salt and alamethicin under a CO2 atmosphere. Treatment of M8 with beta-glucuronidase afforded 1 directly. The presence of M8 in bile and its notable absence from other matrices suggests the enterohepatic cycling of 1 via M8. Although the structure of M7 was not elucidated unequivocally due to its inability to be formed in vitro and its minimal absolute quantities in limited biological matrices, data herein clearly support its structural rationalization. Furthermore, since M7 is the precursor of M8, detection of M8 is indirect evidence of its existence. It is proposed that M7 arises from an equilibrium between 1 and dissolved CO2-equivalents both in vivo and in vitro, similar to carbamino bonds observed in hemoglobin and certain amino acids, respectively.


Subject(s)
GABA Agonists/pharmacokinetics , Indoles/pharmacokinetics , Animals , Area Under Curve , Bile/metabolism , Carbamates/blood , Carbamates/metabolism , Carbamates/urine , Carbon Radioisotopes , Chromatography, Liquid , Feces/chemistry , Female , GABA Agonists/blood , GABA Agonists/urine , GABA-A Receptor Agonists , Glucuronides/blood , Glucuronides/metabolism , Glucuronides/urine , Indoles/blood , Indoles/urine , Macaca fascicularis , Magnetic Resonance Spectroscopy , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...