Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Chem Res Toxicol ; 37(5): 698-710, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38619497

ABSTRACT

Reactive metabolite formation is a major mechanism of hepatotoxicity. Although reactive electrophiles can be soft or hard in nature, screening strategies have generally focused on the use of glutathione trapping assays to screen for soft electrophiles, with many data sets available to support their use. The use of a similar assay for hard electrophiles using cyanide as the trapping agent is far less common, and there is a lack of studies with sufficient supporting data. Using a set of 260 compounds with a defined hepatotoxicity status by the FDA, a comprehensive literature search yielded cyanide trapping data on an unbalanced set of 20 compounds that were all clinically hepatotoxic. Thus, a further set of 19 compounds was selected to generate cyanide trapping data, resulting in a more balanced data set of 39 compounds. Analysis of the data demonstrated that the cyanide trapping assay had high specificity (92%) and a positive predictive value (83%) such that hepatotoxic compounds would be confidently flagged. Structural analysis of the adducts formed revealed artifactual methylated cyanide adducts to also occur, highlighting the importance of full structural identification to confirm the nature of the adduct formed. The assay was demonstrated to add the most value for compounds containing typical structural alerts for hard electrophile formation: half of the severe hepatotoxins with these structural alerts formed cyanide adducts, while none of the severe hepatotoxins with no relevant structural alerts formed adducts. The assay conditions used included cytosolic enzymes (e.g., aldehyde oxidase) and an optimized cyanide concentration to minimize the inhibition of cytochrome P450 enzymes by cyanide. Based on the demonstrated added value of this assay, it is to be initiated for use at GSK as part of the integrated hepatotoxicity strategy, with its performance being reviewed periodically as more data is generated.


Subject(s)
Chemical and Drug Induced Liver Injury , Cyanides , Cyanides/metabolism , Cyanides/chemistry , Humans , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/etiology , Imines/chemistry , Imines/metabolism , Liver/metabolism , Liver/drug effects , Molecular Structure
2.
Eur J Nucl Med Mol Imaging ; 50(3): 667-678, 2023 02.
Article in English | MEDLINE | ID: mdl-36305907

ABSTRACT

PURPOSE: Sotrovimab (VIR-7831), a human IgG1κ monoclonal antibody (mAb), binds to a conserved epitope on the SARS-CoV-2 spike protein receptor binding domain (RBD). The Fc region of VIR-7831 contains an LS modification to promote neonatal Fc receptor (FcRn)-mediated recycling and extend its serum half-life. Here, we aimed to evaluate the impact of the LS modification on tissue biodistribution, by comparing VIR-7831 to its non-LS-modified equivalent, VIR-7831-WT, in cynomolgus monkeys. METHODS: 89Zr-based PET/CT imaging of VIR-7831 and VIR-7831-WT was performed up to 14 days post injection. All major organs were analyzed for absolute concentration as well as tissue:blood ratios, with the focus on the respiratory tract, and a physiologically based pharmacokinetics (PBPK) model was used to evaluate the tissue biodistribution kinetics. Radiomics features were also extracted from the PET images and SUV values. RESULTS: SUVmean uptake in the pulmonary bronchi for 89Zr-VIR-7831 was statistically higher than for 89Zr-VIR-7831-WT at days 6 (3.43 ± 0.55 and 2.59 ± 0.38, respectively) and 10 (2.66 ± 0.32 and 2.15 ± 0.18, respectively), while the reverse was observed in the liver at days 6 (5.14 ± 0.80 and 8.63 ± 0.89, respectively), 10 (4.52 ± 0.59 and 7.73 ± 0.66, respectively), and 14 (4.95 ± 0.65 and 7.94 ± 0.54, respectively). Though the calculated terminal half-life was 21.3 ± 3.0 days for VIR-7831 and 16.5 ± 1.1 days for VIR-7831-WT, no consistent differences were observed in the tissue:blood ratios between the antibodies except in the liver. While the lung:blood SUVmean uptake ratio for both mAbs was 0.25 on day 3, the PBPK model predicted the total lung tissue and the interstitial space to serum ratio to be 0.31 and 0.55, respectively. Radiomics analysis showed VIR-7831 had mean-centralized PET SUV distribution in the lung and liver, indicating more uniform uptake than VIR-7831-WT. CONCLUSION: The half-life extended VIR-7831 remained in circulation longer than VIR-7831-WT, consistent with enhanced FcRn binding, while the tissue:blood concentration ratios in most tissues for both drugs remained statistically indistinguishable throughout the course of the experiment. In the bronchiolar region, a higher concentration of 89Zr-VIR-7831 was detected. The data also allow unparalleled insight into tissue distribution and elimination kinetics of mAbs that can guide future biologic drug discovery efforts, while the residualizing nature of the 89Zr label sheds light on the sites of antibody catabolism.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Infant, Newborn , Humans , Tissue Distribution , Macaca fascicularis/metabolism , SARS-CoV-2/metabolism , Positron Emission Tomography Computed Tomography , Antibodies, Monoclonal/metabolism , Zirconium
3.
Xenobiotica ; 52(6): 541-554, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36083110

ABSTRACT

The pharmacokinetics, elimination, and metabolism of fostemsavir (FTR), a prodrug of the HIV-1 attachment inhibitor temsavir (TMR), were investigated in healthy volunteers. FTR was administered with and without ritonavir (RTV), a protease inhibitor previously shown to boost TMR exposures. In vitro studies were also used to identify the enzymes responsible for the metabolism of TMR.Total recovery of the administered dose ranged from 78% to 89%. Approximately 44% to 58% of the dose was excreted in urine, 20%-36% in faeces, and 5% in bile, as TMR and metabolites. RTV had no effect on the recovery of radioactivity in any matrix.Compared to FTR alone, pre-treatment of subjects with RTV increased the exposure of TMR by ∼66% and reduced the exposure of plasma total radioactivity by ∼68%.The major route of TMR elimination was through biotransformation. TMR, M28 (N-dealkylation), and M4 (amide hydrolysis) were the major circulating components in plasma. Pre-treatment with RTV increased the amount of TMR present, decreased the amount of circulating M28, and M4 was unchanged.CYP3A4 metabolism accounted for 21% of the dose, forming multiple oxidative metabolites. This pathway was inhibited by coadministration of RTV.


Subject(s)
Anti-HIV Agents , Prodrugs , Administration, Oral , Amides , Anti-HIV Agents/pharmacokinetics , Cytochrome P-450 CYP3A , Feces , Healthy Volunteers , Humans , Male , Organophosphates , Piperazines , Protease Inhibitors , Ritonavir/pharmacokinetics
4.
Clin Pharmacol Drug Dev ; 10(4): 334-345, 2021 04.
Article in English | MEDLINE | ID: mdl-33606922

ABSTRACT

Rearranged during transfection (RET), a neuronal growth factor receptor tyrosine kinase, regulates the development of sympathetic, parasympathetic, motor, and sensory neurons in the enteric nervous system. The intended site of action for GSK3352589 is intestinal tissues. GSK3352589 is an RET kinase inhibitor that was administered in double-blind, randomized, placebo-controlled single-dose (SD) and repeat-dose (RD) studies in healthy subjects to investigate its safety/tolerability and pharmacokinetics. In the SD study (n = 28), GSK3352589 was dosed from 2 to 400 mg, including a food effect arm (25 mg). In the RD study (n = 40), GSK3352589 was dosed for 14 days with food twice daily from 5 to 200 mg. With single (fed and fasted) and repeat (fed) doses, bioavailability was low and less than dose-proportional. There was a food effect with 25 mg once daily but may not be clinically relevant. Elimination half-life was ≥17 hours at SD ≥ 15 mg. Accumulation ratios for Cmax , AUCt , AUCtau , and AUC24 after twice-daily dosing to steady state ranged from 1.2 to 1.8 for all doses except the 200-mg dose, which had ratios between 1.9 and 2.7. Administration of GSK3352589 after SD and RD was well tolerated with no safety concerns in healthy subjects.


Subject(s)
Food-Drug Interactions , Protein Kinase Inhibitors , Adult , Female , Humans , Male , Young Adult , Area Under Curve , Biological Availability , Cohort Studies , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Half-Life , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-ret/antagonists & inhibitors
5.
Drug Metab Rev ; 51(4): 453-497, 2019 11.
Article in English | MEDLINE | ID: mdl-31448961

ABSTRACT

The increase in the application of herbal medicines and dietary products over the last decades has been accompanied with a substantial increase in case reports of herb-induced toxicities. Metabolic activation of relatively inert functional groups to chemically reactive metabolites is often considered to be an obligatory event in the etiology of drug-induced adverse reactions. Circumstantial evidence suggests that several herb-induced toxicities are a result of transformation of herbal constituents to electrophilic reactive metabolites that can covalently bind to vital macromolecules in the body, exemplified by aristolochic acids and pyrrolizidine alkaloids. At physiologically relevant concentrations, bioactivation of furanocoumarins and methylenedioxyphenyl compounds leads to mechanism-based inactivation of drug metabolizing enzymes and clinically manifested herb-drug interactions. Of particular interest is that several organic functional groups embedded in herbal constituents act as a toxicophore as well as a pharmacophore, resembling the electrophilic warheads in the development of targeted covalent inhibitors. The aim of this review is to provide a cataloging of bioactivation mechanisms of herbal substructures, structure-activity relationships, biological targets, and assist in circumventing the structural liability in the development of more effective and safer herb-based NCEs.


Subject(s)
Plant Preparations/pharmacokinetics , Plant Preparations/toxicity , Animals , Biotransformation , Humans , Plant Preparations/pharmacology
6.
Clin Pharmacol Drug Dev ; 8(2): 234-245, 2019 02.
Article in English | MEDLINE | ID: mdl-30277655

ABSTRACT

Rearranged during transfection (RET), a neuronal growth factor receptor tyrosine kinase, regulates the development of the sympathetic, parasympathetic, motor, and sensory neurons in the enteric nervous system. GSK3179106 is a RET kinase inhibitor that was administered in double-blind, randomized, placebo-controlled single-dose and repeat-dose studies in healthy subjects to investigate its pharmacokinetics and safety/tolerability. In the single-dose study (n = 16), GSK3179106 was dosed from 10 mg to 800 mg, including a food effect arm. In the repeat-dose study (n = 46), GSK3179106 was dosed for 14 days with food once daily (QD) from 5 mg to 100 mg and twice daily (BID) at 100 mg and 200 mg. With single fasted doses, bioavailability was low and less than dose proportional. A significant food effect was observed with a 100-mg QD dose. Drug exposure after QD and BID repeat dosing with food showed dose dependency up to 100 mg but was not dose proportional. There were no significant differences in exposure between 100-mg and 200-mg BID doses of GSK3179106. Accumulation was observed with both QD and BID dosing. Single doses up to 800 mg and repeat doses up to 400 mg were well tolerated with no safety concerns in healthy subjects.


Subject(s)
Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Fasting , Female , Healthy Volunteers , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Young Adult
7.
Drug Metab Dispos ; 42(8): 1314-25, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872378

ABSTRACT

GSK1322322 (N-((R)-2-(cyclopentylmethyl)-3-(2-(5-fluoro-6-((S)-hexahydropyrazino[2,1-c][1,4]oxazin-8(1H)-yl)-2-methylpyrimidin-4-yl)hydrazinyl)-3-oxopropyl)-N-hydroxy-formamide) is an antibiotic in development by GlaxoSmithKline. In this study, we investigated the metabolism and disposition of [(14)C]GSK1322322 in healthy humans and demonstrated the utility of the Entero-Test in a human radiolabel study. We successfully collected bile from five men using this easy-to-use device after single i.v. (1000 mg) or oral administration (1200 mg in a solution) of [(14)C]GSK1322322. GSK1322322 had low plasma clearance (23.6 liters/hour) with a terminal elimination half-life of ∼4 hours after i.v. administration. After oral administration, GSK1322322 was readily and almost completely absorbed (time of maximal concentration of 0.5 hour; bioavailability 97%). GSK1322322 predominated in the systemic circulation (>64% of total plasma radioactivity). An O-glucuronide of GSK1322322 (M9) circulated at levels between 10% and 15% of plasma radioactivity and was pharmacologically inactive. Humans eliminated the radioactive dose in urine and feces at equal proportions after both i.v. and oral doses (∼45%-48% each). Urine contained mostly unchanged GSK1322322, accounting for 30% of the dose. Bile contained mostly M9, indicating that glucuronidation was likely a major pathway in humans (up to 30% of total dose). In contrast, M9 was found in low amounts in feces, indicating its instability in the gastrointestinal tract. Therefore, without the Entero-Test bile data, the contribution of glucuronidation would have been notably underestimated. An unusual N-dehydroxylated metabolite (a secondary amide) of GSK1322322 was observed primarily in the feces and was most likely formed by gut microbes.


Subject(s)
Bile/metabolism , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Hydroxamic Acids/metabolism , Protease Inhibitors/metabolism , Administration, Oral , Adult , Biological Availability , Bridged Bicyclo Compounds, Heterocyclic/urine , Cross-Over Studies , Feces/chemistry , Gastrointestinal Tract/metabolism , Half-Life , Humans , Hydroxamic Acids/urine , Male , Metabolic Clearance Rate/physiology , Middle Aged , Peptide Hydrolases
8.
Drug Metab Dispos ; 41(12): 2215-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24097902

ABSTRACT

A phase I study was conducted to assess the metabolism and excretion of [(14)C]dabrafenib (GSK2118436; N-{3-[5-(2-amino-4-pyrimidinyl)-2-(1,1-dimethylethyl)-1,3-thiazol-4-yl]-2-fluorophenyl}-2,6-difluorobenzene sulfonamide, methanesulfonate salt), a BRAF inhibitor, in four patients with BRAF V600 mutation-positive tumors after a single oral dose of 95 mg (80 µCi). Assessments included the following: 1) plasma concentrations of dabrafenib and metabolites using validated ultra-high-performance liquid chromatography--tandem mass spectrometry methods, 2) plasma and blood radioactivity, 3) urinary and fecal radioactivity, and 4) metabolite profiling. Results showed the mean total recovery of radioactivity was 93.8%, with the majority recovered in feces (71.1% of administered dose). Urinary excretion accounted for 22.7% of the dose, with no detection of parent drug in urine. Dabrafenib is metabolized primarily via oxidation of the t-butyl group to form hydroxy-dabrafenib. Hydroxy-dabrafenib undergoes further oxidation to carboxy-dabrafenib, which subsequently converts to desmethyl-dabrafenib via a pH-dependent decarboxylation. The half-lives for carboxy- and desmethyl-dabrafenib were longer than for parent and hydroxy-dabrafenib (18-20 vs. 5-6 hours). Based on area under the plasma concentration-time curve, dabrafenib, hydroxy-, carboxy-, and desmethyl-dabrafenib accounted for 11%, 8%, 54%, and 3% of the plasma radioactivity, respectively. These results demonstrate that the major route of elimination of dabrafenib is via oxidative metabolism (48% of the dose) and biliary excretion. Based on our understanding of the decarboxylation of carboxy-dabrafenib, a low pH-driven, nonenzymatic mechanism involving participation of the aryl nitrogen is proposed to allow prediction of metabolic oxidation and decarboxylation of drugs containing an aryl nitrogen positioned α to an alkyl (ethyl or t-butyl) side chain.


Subject(s)
Carbon/metabolism , Decarboxylation/physiology , Imidazoles/metabolism , Neoplasms/metabolism , Nitrogen/metabolism , Oximes/metabolism , Administration, Oral , Adult , Feces/chemistry , Female , Half-Life , Humans , Male , Middle Aged , Oxidation-Reduction , Young Adult
9.
Xenobiotica ; 43(5): 443-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23548165

ABSTRACT

1. Pazopanib (Votrient) is an oral tyrosine kinase inhibitor that was recently approved for the treatment of renal cell carcinoma and soft tissue sarcoma. 2. In this two-part study, we investigated the metabolism, disposition of [(14)C]pazopanib, and the oral bioavailability of pazopanib tablets in patients with advanced cancer. 3. In part A, three men each received a single oral dose of [(14)C]pazopanib in suspension (400 mg, 70 µCi). Pazopanib was the predominant drug-related component in circulation. Two metabolites derived from hydroxylation and one from N-demethylation were also circulating, but were minor, each accounting for <5% of plasma radioactivity. Faecal elimination predominated, accounting for 82.2% of the administered radio-dose, with negligible renal elimination (2.6% of dose). Pazopanib was primarily excreted as the unchanged drug in faeces (67% of dose). 4. In part B, seven additional patients received a single intravenous administration of 5 mg pazopanib (day 1) followed by oral administration of 800 mg pazopanib tablet once daily for 26 days (days 3 or 5-28). In the three evaluable patients from part B, pazopanib had a slow plasma clearance and a small volume of distribution. The absolute oral bioavailability of the 800 mg pazopanib tablet ranged from 14% to 39%.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Pyrimidines/pharmacokinetics , Sulfonamides/pharmacokinetics , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Carbon Radioisotopes , Humans , Indazoles , Male , Middle Aged , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use
10.
Drug Metab Dispos ; 39(9): 1620-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21628498

ABSTRACT

GSK977779 is a potent HM74a agonist evaluated for the treatment of dyslipidemia. The disposition and metabolism of [(14)C]GSK977779 (67.6 µmol/kg p.o.) was studied in male and female rats. The compound was well absorbed and its primary route of elimination was in the feces. Based on metabolite profiling of plasma extracts and urine and bile samples, it was demonstrated that GSK977779 was extensively metabolized in the rat by N-dealkylation, mono- and dioxygenation, reductive and oxidative cleavage of the 1,2,4-oxadiazole ring, and conjugative pathways. After plasma extraction high amounts of nonextractable radioactivity were observed, which were more pronounced in female rats. Size-exclusion chromatography and SDS gel electrophoresis indicated that the majority of the nonextractable radioactivity was covalently bound to plasma proteins. Solubilization of the plasma protein pellet followed by high-performance liquid chromatography and mass spectrometry suggested that a carboxylic acid metabolite derived from oxadiazole ring cleavage may be responsible for the observed covalent binding of the radioactivity to rat plasma proteins.


Subject(s)
Oxadiazoles/metabolism , Oxadiazoles/pharmacokinetics , Purines/metabolism , Purines/pharmacokinetics , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Animals , Bile/metabolism , Blood Proteins/metabolism , Carbon Radioisotopes/chemistry , Chromatography, High Pressure Liquid/methods , Feces , Female , Liver/metabolism , Male , Mass Spectrometry/methods , Microsomes, Liver/metabolism , Oxadiazoles/chemistry , Plasma/metabolism , Protein Binding , Purines/chemistry , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism
11.
Drug Metab Dispos ; 39(9): 1734-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21646437

ABSTRACT

The metabolism and disposition of eltrombopag, the first-in-class small molecule human thrombopoietin receptor agonist, were studied in six healthy men after a single oral administration of a solution dose of [(14)C]eltrombopag (75 mg, 100 µCi). Eltrombopag was well tolerated. The drug was quickly absorbed and was the predominant circulating component in plasma (accounting for 63% of the total plasma radioactivity). A mono-oxygenation metabolite (M1) and acyl glucuronides (M2) of eltrombopag were minor circulating components. The predominant route of elimination of radioactivity was fecal (58.9%). Feces contained approximately 20% of dose as glutathione-related conjugates (M5, M6, and M7) and another 20% as unchanged eltrombopag. The glutathione conjugates were probably detoxification products of a p-imine methide intermediate formed by metabolism of M1, which arises through cytochrome P450-dependent processes. Low levels of covalently bound drug-related intermediates to plasma proteins, which could result from the reaction of the imine methide or acyl glucuronide conjugates with proteins, were detected. The bound material contributes to the longer plasma elimination half-life of radioactivity. Renal elimination of conjugates of hydrazine cleavage metabolites (mostly as M3 and M4) accounted for 31% of the radiodose, with no unchanged eltrombopag detected in urine.


Subject(s)
Benzoates/pharmacokinetics , Hydrazines/pharmacokinetics , Pyrazoles/pharmacokinetics , Receptors, Thrombopoietin/agonists , Administration, Oral , Adult , Benzoates/blood , Benzoates/metabolism , Benzoates/urine , Biotransformation , Blood Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Feces/chemistry , Glucuronides/blood , Glutathione/metabolism , Half-Life , Humans , Hydrazines/blood , Hydrazines/metabolism , Hydrazines/urine , Male , Middle Aged , Protein Binding , Pyrazoles/blood , Pyrazoles/metabolism , Pyrazoles/urine , Receptors, Thrombopoietin/metabolism
12.
Drug Metab Dispos ; 39(9): 1747-54, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21646438

ABSTRACT

After oral administration to humans, eltrombopag undergoes extensive cleavage of its hydrazine linkage to metabolites, which are exclusively eliminated in urine. In vitro, the cleavage pathway was not detected in systems using cytochrome P450 enzymes, renal or hepatic microsomes, or hepatocytes but was readily evident after anaerobic incubation with rodent cecal contents or human fecal homogenate. Antibiotic treatment in vitro and in vivo inhibited eltrombopag cleavage, further indicating that cleavage is via gut microbes. Antibiotic treatment did not alter the systemic exposure of eltrombopag in mice. Oral and intravenous pharmacokinetic characterization in the mice with one of the cleavage products indicated that it was readily absorbed, conjugated, and eliminated in urine, consistent with its fate after oral administration of eltrombopag. Variation in this microbial pathway, for example by antibiotic cotherapy, is unlikely to be clinically significant.


Subject(s)
Benzoates/metabolism , Hydrazines/metabolism , Pyrazoles/metabolism , Administration, Oral , Animals , Anti-Bacterial Agents/pharmacology , Benzoates/pharmacokinetics , Cecum/drug effects , Cecum/microbiology , Cytochrome P-450 Enzyme System/metabolism , Feces/chemistry , Female , Hepatocytes/metabolism , Humans , Hydrazines/pharmacokinetics , Kidney/metabolism , Liver/metabolism , Male , Mice , Microsomes/enzymology , Microsomes/metabolism , Pyrazoles/pharmacokinetics , Rats, Sprague-Dawley
13.
Bioorg Med Chem Lett ; 17(6): 1713-7, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17236763

ABSTRACT

N,N'-diarylsquaramides were prepared and evaluated as antagonists of CXCR2. The compounds were found to be potent and selective antagonists of CXCR2. Significant differences in SAR was observed relative to the previously described N,N'-diarylurea series. As was the case in the N,N'-diarylurea series, placing sulfonamide substituent adjacent to the acidic phenol significantly reduced the clearance in rat pharmacokinetic studies.


Subject(s)
Cyclobutanes/chemical synthesis , Cyclobutanes/pharmacology , Cyclobutanes/pharmacokinetics , Receptors, Interleukin-8B/antagonists & inhibitors , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/pharmacology , Urea/pharmacokinetics , Animals , CHO Cells , Chemical Phenomena , Chemistry, Physical , Cricetinae , Cricetulus , Indicators and Reagents , Mass Spectrometry , Phenols/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemistry
14.
Bioorg Med Chem Lett ; 14(17): 4375-8, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15357956

ABSTRACT

A series of 3-substituted N,N'-diarylureas was prepared and the structure-activity relationship relative to CXCR2 receptor affinity as well as their pharmacokinetic properties were examined. In vitro microsomal metabolism studies indicated that the lower clearance rates of the 3-sulfonamido-substituted compounds were most likely due to the suppression of glucuronidation.


Subject(s)
Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonylurea Compounds/chemistry , Administration, Oral , Animals , Biological Availability , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8B/metabolism , Sulfonylurea Compounds/metabolism , Sulfonylurea Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...