Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 34(5): 748-55, 2006 May.
Article in English | MEDLINE | ID: mdl-16455802

ABSTRACT

The potential cancer therapeutic agent, 6,7-(dimethoxy-2, 4-dihydroindeno[1,2-c]pyrazol-3-yl)-(3-fluoro-phenyl)-amine (JNJ-10198409), formed three N-glucuronides that were positively identified by liquid chromatography-tandem mass spectrometry and NMR as N-amine-glucuronide (Glu-A), 1-N-pyrazole-glucuronide (Glu-B), and 2-N-pyrazole-glucuronide (Glu-C). All three N-glucuronides were detected in rat liver microsomes, whereas only Glu-A and -B were found in monkey and human liver microsomes. In contrast to common glucuronides, Glu-B was completely resistant to beta-glucuronidase. Kinetic analyses revealed that glucuronidation of JNJ-10198409 in human liver microsomes exhibited atypical kinetics that may be described by a two-site binding model. For the high affinity binding, K(m) values were 1.2 and 5.0 microM, and V(max) values were 2002 and 2,403 nmol min(-1) mg(-1) for Glu-A and Glu-B, respectively. Kinetic constants of low affinity binding were not determined due to low solubility of the drug. Among the human UDP-glucuronosyltransferases (UGTs) tested, UGT1A9, 1A8, 1A7, and 1A4 were the most active isozymes to produce Glu-A; for the formation of Glu-B, UGT1A9 was the most active enzyme, followed by UGT1A3, 1A7, and 1A4. Glucuronidation of JNJ-10198409 by those UGT1A enzymes followed classic Michaelis-Menten kinetics. In contrast, no glucuronides were formed by all UGT2B isozymes tested, including UGT2B4, 2B7, 2B15, and 2B17. Collectively, these results suggested that glucuronidation of JNJ-10198409 in human liver microsomes is catalyzed by multiple UGT1A enzymes. Since UGT1A enzymes are widely expressed in various tissues, it is anticipated that both hepatic and extrahepatic glucuronidation will likely contribute to the elimination of the drug in humans. Additionally, conjugation at the nitrogens of the pyrazole ring represents a new structural moiety for UGT1A-mediated reactions.


Subject(s)
Glucuronosyltransferase/metabolism , Indans/metabolism , Pyrazoles/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Chromatography, High Pressure Liquid , DNA, Complementary/biosynthesis , Glucuronides/metabolism , Glucuronosyltransferase/biosynthesis , Haplorhini , Humans , In Vitro Techniques , Kinetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microsomes, Liver/enzymology , Oxidation-Reduction , Propofol/pharmacology , Rats
2.
Eur J Drug Metab Pharmacokinet ; 30(3): 151-64, 2005.
Article in English | MEDLINE | ID: mdl-16250251

ABSTRACT

The metabolism and excretion of 2,3:4,5-bis-O-(1-methylethylidene)-beta-D-fructopyranose sulfamate (TOPAMAX, topiramate, TPM) have been investigated in animals and humans. Radiolabeled [14C] TPM was orally administered to mice, rats, rabbits, dogs and humans. Plasma, urine and fecal samples were collected and analyzed. TPM and a total of 12 metabolites were isolated and identified in these samples. Metabolites were formed by hydroxylation at the 7- or 8-methyl of an isopropylidene of TPM followed by rearrangement, hydroxylation at the 10-methyl of the other isopropylidene, hydrolysis at the 2,3-O-isopropylidene, hydrolysis at the 4,5-O-isopropylidene, cleavage at the sulfamate group, glucuronide conjugation and sulfate conjugation. A large percentage of unchanged TPM was recovered in animal and human urine. The most dominant metabolite of TPM in mice, male rats, rabbits and dogs appeared to be formed by the hydrolysis of the 2,3-O-isopropylidene group.


Subject(s)
Anticonvulsants/metabolism , Fructose/analogs & derivatives , Migraine Disorders/drug therapy , Adult , Animals , Dogs , Female , Fructose/metabolism , Humans , Male , Mice , Rabbits , Rats , Rats, Sprague-Dawley , Sex Characteristics , Species Specificity , Topiramate
3.
Eur J Drug Metab Pharmacokinet ; 28(3): 223-32, 2003.
Article in English | MEDLINE | ID: mdl-14527096

ABSTRACT

A "dual-activity" microsomal system in which both CYPs and UGTs were active was evaluated for studies of metabolic stability and in-vitro metabolite profiling. In this "dual-activity" system, alamethicin, a pore-forming peptide, was used to activate UGTs in human liver microsomes without affecting CYP activity. Interference studies indicated that CYP cofactors had little effect on UGT surrogate activity as measured by glucuronidation of acetaminophen and trifluoperazine. Further, UGT cofactor, UDPGA (< 2 mM), did not inhibit the marker activity of five major CYPs including 1A2, 2C9, 2C19, 2D6 and 3A4, suggesting that both oxidation and glucuronidation can be co-activated in microsomes. In a comparison study, compounds with significant glucuronidation showed distinct stability profiles in the "dual-activity" system, compared to the conventional microsomal incubation in which only CYPs were active. For compounds with minor or no glucuronidation, the metabolic stability remained similar between the "dual-activity" system and the conventional microsomal incubation. The feasibility of this "dual-activity" system utilized for metabolite profiling was also investigated using tramadol as a model drug. It was found that oxidative metabolites of tramadol generated in the "dual-activity" system matched those detected in the conventional microsomal incubation. However, tramadol glucuronide was observed in the "dual-activity" system but not in the conventional micromosal incubation. Results clearly suggest that the "dual-activity" system is a valuable in vitro model for metabolism studies in drug discovery.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Glucuronosyltransferase/metabolism , Microsomes, Liver/enzymology , Alamethicin/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Microsomes, Liver/drug effects , Rats
4.
Xenobiotica ; 32(11): 949-62, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12487725

ABSTRACT

1. In vitro studies have been carried out to investigate the metabolic pathways and identify the hepatic cytochrome P450 (CYP) enzymes involved in etoperidone (Et) metabolism. 2. Ten in vitro metabolites were profiled, quantified and tentatively identified after incubation with human hepatic S9 fractions. Et was metabolized via three metabolic pathways: (A) alkyl hydroxylation to form OH-ethyl-Et (M1); (B) phenyl hydroxylation to form OH-phenyl-Et (M2); and (C) N-dealkylation to form 1-m-chlorophenylpiperazine (mCPP, M8) and triazole propyl aldehyde (M6). Six additional metabolites were formed by further metabolism of M1, M2, M6 and M8. 3. Kinetic studies revealed that all metabolic pathways were monophasic, and the pathway leading to the formation of OH-ethyl-Et was the most efficient at eliminating the drug. On incubation with microsomes expressing individual recombinant CYPs, formation rates of M1-3 and M8 were 10-100-fold greater for CYP3A4 than that for other CYP forms. The formation of these metabolites was markedly inhibited by the CYP3A4-specific inhibitor ketoconazole, whereas other CYP-specific inhibitors did not show significant effects. In addition, the production of M1-3 and M8 was strongly correlated with CYP3A4-mediated testosterone 6beta-hydroxylase activities in 13 different human liver microsome samples. 4. Dealkylation of the major metabolite M1 to form mCPP (M8) was also investigated using microsomes containing recombinant CYP enzymes. The rate of conversion of M1 to mCPP by CYP3A4 was 503.0 +/- 3.1 pmole nmole(-1) min(-1). Metabolism of M1 to M8 by other CYP enzymes was insignificant. In addition, this metabolism in human liver microsomes was extensively inhibited by the CYP3A4 inhibitor ketoconazole, but not by other CYP-specific inhibitors. In addition, conversion of M1 to M8 was highly correlated with CYP3A4-mediated testosterone 6beta-hydroxylase activity. 5. The results strongly suggest that CYP3A4 is the predominant enzyme-metabolizing Et in humans.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Liver/enzymology , Trazodone/analogs & derivatives , Trazodone/pharmacology , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Kinetics , Liver/drug effects , Mass Spectrometry , Microsomes, Liver/enzymology , Models, Chemical , Protein Isoforms , Recombinant Proteins/metabolism , Steroid Hydroxylases/metabolism , Trazodone/metabolism
5.
J Pharm Biomed Anal ; 28(3-4): 659-71, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12008146

ABSTRACT

The excretion and metabolism of (+/-)-trans-3-(2-bromophenyl)octahydroindolizine hydrochloride (RWJ-22757) have been investigated in male and female CR Wistar rats. Radiolabeled [14C] RWJ-22757 was administered orally to each of the rats as a single 60 mg/kg suspension dose. Plasma (0-48 h), urine (0-168 h) and fecal (0-168 h) samples were collected and analyzed. There were no significant gender differences observed in the data. The estimated elimination half-life of the total radioactivity from plasma was 19 h while the estimated elimination half-life of RWJ-22757 was 15 h. Recoveries of total radioactivity in urine and feces were 58.4+/-5.8 and 42.4+/-6.3%, respectively. RWJ-22757 and a total of 11 metabolites were isolated in rat plasma, urine, and fecal extracts. The structures of four of these metabolites were tentatively identified. Unchanged RWJ-22757 accounted for < 4% of the dose in plasma and urine and 28% in feces; thus, indicating the drug was extensively metabolized and either not absorbed well or biliary excreted. Identified metabolites accounted for > 80% of the total radioactivity contained in the samples. The following pathways were used to describe the formation of the metabolites identified in rats: octahydroindolizine ring oxidation, phenyl hydroxylation, octahydroindolizine ring oxidation followed by ring opening to a carboxylic acid function and octahydroindolizine ring oxidation followed by ring opening and N-methylation.


Subject(s)
Analgesics, Non-Narcotic/pharmacokinetics , Indolizines/pharmacokinetics , Analgesics, Non-Narcotic/analysis , Analgesics, Non-Narcotic/urine , Animals , Area Under Curve , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Feces/chemistry , Female , Indolizines/analysis , Indolizines/urine , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Rats , Rats, Wistar
6.
Eur J Drug Metab Pharmacokinet ; 27(4): 281-7, 2002.
Article in English | MEDLINE | ID: mdl-12587958

ABSTRACT

In this study we have evaluated the reliability of a fluorescence-based method used for rapid identification of irreversible CYP inhibitors (mechanism-based inhibitors). This was accomplished by comparing the time-dependence pattern of IC50 values from fluorometric kinetic measurements. For irreversible CYP inhibitors, IC50 values decreased as incubation proceeded. This was due to progressive inactivation of corresponding enzymes by reactive metabolites generated during the incubation. This change pattern was confirmed using a number of known irreversible CYP inhibitors, including furafylline, midazolam, erythromycin, clarithromycin, oleandomycin, 17alpha-ethynylestradiol and verapamil. The pattern was different in reversible inhibition, depending upon the compounds tested in the fluorometric kinetic assay. For some compounds, such as clotrimazole, IC50 values remained relatively stable, whereas other compounds, such as miconazole, terfenadine and ketoconazole showed a significant increase with incubation time. Monitoring tested compounds by LC-MS/MS during the incubation confirmed that increases of IC50 were probably caused by the loss of inhibitors, resulting from either metabolic degradation, or non-specific binding to microsomal proteins.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Chromatography, Liquid , Cytochrome P-450 CYP3A , Enzyme Inhibitors/pharmacokinetics , Fluorescent Dyes , Kinetics , Mass Spectrometry , Microsomes, Liver/metabolism , Reproducibility of Results , Spectrometry, Fluorescence
7.
Eur J Drug Metab Pharmacokinet ; 26(3): 155-66, 2001.
Article in English | MEDLINE | ID: mdl-11695715

ABSTRACT

The absorption, excretion and metabolism of N-(2, 6-dichlorophenyl)-beta-[[(1-methylcyclohexyl)methoxylmethyl]-N-(phenylmethyl)-1-pyrrolidineethanamine (RWJ-26899; McN-6497) has been investigated in male and female CR Wistar rats and beagle dogs. Radiolabeled [14C] RWJ-26899 was administered to rats as a single 24 mg/kg suspension dose while the dogs received 15 mg/kg capsules. Plasma (0-36 h; rat and 0-48 h; dog), urine (0-192 h; rat and dog) and fecal (0-192 h; rat and dog) samples were collected and analyzed. There were no significant gender differences observed in the data. The terminal half-life of the total radioactivity for rats from plasma was estimated to be 7.7 +/- 0.6 h while for dogs it was 22.9 +/- 4.4 h. Recoveries of total radioactivity in urine and feces for rats were 8.7 +/- 2.9% and 88.3 +/- 10.4% of the dose, respectively. Recoveries of total radioactivity in urine and feces for dogs were 4.1 +/- 1.4% and 90.0 +/- 4.7% of the dose, respectively. RWJ-26899 and a total of nine metabolites were isolated and tentatively identified in rat urine, and fecal extracts. Unchanged RWJ-26899 accounted for approximately 1% of the dose in rat urine and 8% in rat feces. RWJ-26899 and a total of four metabolites were isolated and identified in dog urine, and fecal extracts. Unchanged RWJ-26899 accounted for approximately 1% of the dose in urine and 63% in feces in dog. Five proposed pathways were used to describe the metabolites found in rats: N-oxidation, oxidative N-debenzylation, pyrrolidinyl ring hydroxylation, phenyl hydroxylation and methyl or cyclohexyl hydroxylation. Two biotransformation pathways in dogs are proposed: N-oxidation and methyl or cyclohexyl ring hydroxylation.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Pyrrolidines/pharmacokinetics , Animals , Antihypertensive Agents/urine , Bepridil/pharmacokinetics , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Dogs , Feces/chemistry , Female , Intestinal Absorption , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Pyrrolidines/urine , Rats , Rats, Wistar
8.
Curr Top Med Chem ; 1(5): 353-66, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11899102

ABSTRACT

The attrition rates of new chemical entities (NCEs) in preclinical and clinical development are staggeringly high. NCEs are abandoned due to insufficient efficacy, safety issues, and economic reasons. Uncovering drug defects that produce these failures as early as possible in drug discovery would be highly effective in lowing the cost and time of developing therapeutically useful drugs. Unfortunately, there is no single factor that can account for these NCE failures in preclinical and clinical development since factors, such as solubility, pKa, absorption, metabolism, formulation, pharmacokinetics, toxicity and efficacy, to name a few, are all interrelated. In addition, there are many problems in scaling-up drug candidates from the laboratory bench scale to the pilot plant scale. To address the problem of attrition rates of NCEs in preclinical and clinical development and drug scale-up issues, pharmaceutical companies need to reorganize their preclinical departments from a traditional linear approach to a parallel approach. In this review, a strategy is put forth to integrate certain aspects of drug metabolism/pharmacokinetics, toxicology functions and process chemistry into drug discovery. Compound optimization in early and late phase drug discovery occurs by relating factors such as physicochemical properties, in vitro absorption, in vitro metabolism, in vivo pharmacokinetics and drug scale-up issues to efficacy optimization. This pre-preclinical paradigm will improve the success rate of drug candidates entering development.


Subject(s)
Drug Evaluation, Preclinical/methods , Pharmacology/standards , Animals , Biological Availability , Cells, Cultured , Drug-Related Side Effects and Adverse Reactions , Guinea Pigs , Humans , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Rabbits , Rats
9.
Curr Top Med Chem ; 1(5): 403-25, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11899105

ABSTRACT

To reduce the high attrition rates of NCEs in preclinical and clinical development uncovering pharmacokinetics, toxicokinetics, drug metabolism, and drug-drug interactions early in drug discovery would be highly valuable. There have been many in vitro screens developed for these areas that have higher sample throughput, which is consistent with the iterative cycle of a typical drug discovery research project. We have presented the present status and given detailed descriptions of biotransformation, metabolic stability assays, identification of drug metabolizing P450 enzymes, prediction of pharmacokinetic parameters from in vitro metabolism data, structure elucidation of metabolites, CYP450 inhibition assays and CYP450 induction assays from a drug discovery perspective. Strategies for the proper sequencing of primary and secondary assays employedfor drug metabolism and CYP450 inhibition & induction is discussed.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical/methods , Liver/enzymology , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Cytochrome P-450 Enzyme System/biosynthesis , Drug Interactions , Drug-Related Side Effects and Adverse Reactions , Enzyme Induction/drug effects , Humans , Liver/drug effects , Liver/metabolism
10.
Eur J Drug Metab Pharmacokinet ; 26(4): 263-71, 2001.
Article in English | MEDLINE | ID: mdl-11808869

ABSTRACT

The excretion and metabolism of (+/-) [6-(3,4-dihydro-3-oxo-1,4[2H]-benzoxazine-yl)-2,3,4,5-tetrahydro-5-methylpyridazin-3-one] (bemoradan; RWJ-22867) have been investigated in male Long-Evans rats and female beagle dogs. Radiolabeled [14C] bemoradan was administered to rats as a singkle 1 mg/kg suspension dose while the dogs received 0.1 mg/kg suspension dose. Plasma (0-24 h; rat and dog), urine (0-72 h; rat and dog) and fecal (0-72 h; rat and dog) samples were collected and analyzed. The terminal half-life of the total radioactivity for rats from plasma was estimate to be 4.3 +/- 0.1 h while for dogs it was 7.5 +/- 1.3 h. Recoveries of total radioactivity in urine and feces for rats were 49.1 +/- 2.4% and 51.1 +/- 4.9% of th dose, respectively. Recoveries of total radioactivity in urine and feces for dogs were 56.2 +/- 12.0% and 42.7 V 9.9% of the dose, respectively. Bemoradan and a total of nine metabolites were isolated and tentatively identified in rat and dog plasma, urine, and fecal extracts. Unchanged bemoradan accounted for approimately < 2% of the dose in rat urine and 20% in rat feces. Unchanged bemoradan accounted for approximately 5% of the dose in urine and 16% in feces in dog. Six proposed pathways were used to describe the metabolites found in rats and dogs: pyridazinyl oxidations, methyl hydroxylation, hydration, N-oxidation, dehydration and phase II conjugations.


Subject(s)
Cardiotonic Agents/pharmacokinetics , Oxazines/pharmacokinetics , Pyridazines/pharmacokinetics , Animals , Benzoxazines , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Dogs , Drug Evaluation, Preclinical , Female , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Models, Chemical , Rats , Rats, Long-Evans
11.
Xenobiotica ; 31(11): 823-39, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11765144

ABSTRACT

1. The absorption, excretion and metabolism of 2-[3-[4-(3-chlorophenyl)-1-piperazinyl]propyl]-4,5-diethyl-2,4-dihydro-3H-1,2,4- triazole-3-one hydrochloride (etoperidone HCl) was investigated in six healthy men. Subjects were tasted overnight before receiving a single oral dose of a 100 mg solution [14C] etoperidone HCl. 2. Plasma (0-48 h), urine (0-120 h) and faecal (0-120 h) samples were collected. The terminal half-life of the total radioactivity from plasma was 21.7 +/- 2.8h with an apparent clearance of 1.01 +/- 0.08 ml min(-1). Recoveries of total radioactivity in urine and faeces were 78.8 +/- 3.6% and 9.6 +/- 4.1% of the dose, respectively. 3. Etoperidone and 21 metabolites were isolated and identified in the plasma, urine and faecal extracts. Unchanged etoperidone accounted for <0.01% of the dose in all excreta samples. Nine metabolites were identified in the plasma extracts and 21 urinary metabolites were identified. Seven faecal metabolites were identified. 4. Five proposed pathways were used to describe the formation of the metabolites: alkyl oxidation, piperazinyl oxidation, N-dealkylation, phenyl hydroxylation and conjugation. Alkyl oxidation of etoperidone resulted in the formation of 2-[3-[4-(3-chlorophenyl)-1-piperazinyl]propyl]-4-ethyl-2,4-dihydro-5- (1-hydroxyethyl)-3H-1,2,4-triazole-3-one. Piperazinyl oxidation of this metabolite leads to the formation of its N-oxide. N-dealkylation of the piperazinyl group led to the formation of 1-(3-chlorophenyl) piperazine and triazole propionic acid. Phenyl hydroxylation led to three important metabolites in the urine and faeces.


Subject(s)
Antidepressive Agents/pharmacokinetics , Trazodone/analogs & derivatives , Trazodone/pharmacokinetics , Administration, Oral , Adult , Antidepressive Agents/metabolism , Area Under Curve , Carbon Radioisotopes/metabolism , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Feces , Humans , Hydroxylation , Magnetic Resonance Spectroscopy , Male , Molecular Structure , Trazodone/metabolism
13.
Curr Opin Drug Discov Devel ; 3(1): 30-41, 2000 Jan.
Article in English | MEDLINE | ID: mdl-19649835

ABSTRACT

New chemical entities (NCEs) are abandoned in development primarily because of insufficient efficacy, safety issues and for economic reasons. Since efficacy and safety deficiencies are related in part to pharmacokinetics (PK), uncovering PK defects as early in drug discovery as possible would be highly valuable in reducing NCE failures in preclinical and clinical development. In this review, a strategy is put forth to integrate drug metabolism/pharmacokinetics and toxicology functions into drug discovery. Compound optimization in early- and late-phase drug discovery is covered, emphasizing physicochemical properties, in vitro absorption, metabolism and in vivo animal PK methodologies, primarily from the period 1998 to 1999. The present study also illustrates the idea of sorting oral bioavailability data into high/intermediate/low categories based on combining high/low rank-ordered information from physicochemical properties and in vitro absorption, metabolism and serum binding assays. It is shown that by combining the results from solubility, stability, absorption and metabolism assays, the high/intermediate/low human oral bioavailability for a series of beta- blockers can be approximately predicted. This method has a high sample throughput and should be useful in rank-ordering the predicted oral bioavailability of large collections of compounds at the lead optimization step of drug discovery. These results are useful for selecting compounds for future in vitro/in vivo correlation modeling or in vivo animal testing. This type of approach will improve the decision making process of compound selection in drug discovery.

14.
J Biochem Biophys Methods ; 40(1-2): 17-25, 1999 Jul 28.
Article in English | MEDLINE | ID: mdl-10481948

ABSTRACT

We have shown that affinity capillary electrophoresis (ACE) can be utilized to screen peptides that bind to the extracellular binding domain of the erythropoietin receptor (EBP). The comparison of the cyclic peptides GGTYSCHFGPLTWVCKPQGG (EMP1) GGTYSCHFGPLTAVCKPQGG (EMP13), and LGRKYSCHFGPLTWVCQPAKKD (EMP37) with the linear peptides HFGPLTWV (EMP26) and FMRF as ACE buffer additives were investigated. When EMP1 and EMP37 were the buffer additives, an abrupt change in the electrophoretic mobility of EBP was observed in the electropherogram. When EMP13, EMP26, and FMRF were examined under identical ACE conditions as EMP1 and EMP37, no significant change in the electrophoretic mobility of EBP was observed. These results correlate well with previously reported IC50 competitive binding data; that is, EMP1 and EMP37 bind to EBP while EMP13 and EMP26 bind very weakly. These observations strongly infer that peptide.EBP dimerization were induced by EMP1, and EMP37 but not by EMP13, EMP26 or FMRF. This ACE method provides a rapid tool for the detection of small peptides or drugs that bind to EBP.


Subject(s)
Electrophoresis, Capillary/methods , Peptides, Cyclic/chemistry , Receptors, Erythropoietin/chemistry , Amino Acid Sequence , Ligands , Molecular Sequence Data , Protein Binding
15.
Biochem Biophys Res Commun ; 262(3): 793-800, 1999 Sep 07.
Article in English | MEDLINE | ID: mdl-10471404

ABSTRACT

Our previous studies have shown that the His tag cleaved from fusion proteins contained two distinct components P1 and P2. P1 has been identified to be a His-tagged peptide of G-H-H-H-H-H-H-H-H-H-H-S-S-G-H-I-E-G-R resulted from initiator methionine deletion, and P2 contains an unknown moiety at the second residue glycine of the tag (x-G-H-H-H-H-H-H-H-H-H-H-S-S-G-H-I-E-G-R, x = 178.0 Da). This study aimed to determine the structure of the modification by using a combination of protein isotope labeling and mass spectrometry. His-tagged FKBP was expressed in (15)N and (13)C labeling growth media respectively. Isotopic labeled His-tagged proteins ((15)N-His-FKBP and (13)C-His-FKBP) were isolated by affinity chromatography and subjected to Xa digestions to release the labeled His tag. Subsequent analyses of the released His tag by MALDI-TOF-MS indicated a mass difference of 178.0 +/- 0.2 Da, between the two (15)N-labeled peptides P1 and P2, suggesting that the modification moiety contained no nitrogen. A mass difference of 184.0 +/- 0.2 Da was observed on MALDI between (13)C-labeled peptide P1 and P2, indicating six carbons in the modification group. Also, comparing the mass shift on MALDI spectra of P1 and P2 after hydrogen/deuterium exchange revealed that the modification moiety had five hydroxyl groups. It was concluded that the modification was a gluconic acid derivative attached to the N-terminus of His-tagged proteins expressed in bacteria. The proposed structure was further confirmed by MALDI analysis of periodate oxidation products of His-tagged peptides.


Subject(s)
Gluconates , Immunophilins/biosynthesis , Amino Acid Sequence , Carbon Isotopes , Chromatography, Affinity , Cloning, Molecular , Histidine , Immunophilins/chemistry , Immunophilins/isolation & purification , Molecular Sequence Data , Nitrogen Isotopes , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tacrolimus Binding Proteins
16.
Comb Chem High Throughput Screen ; 2(1): 39-51, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10420975

ABSTRACT

We have assessed the metabolic activity of freshly isolated and commercially preserved rat, monkey, and human primary hepatocytes in a 96 well plate format utilizing eight beta-adrenolytic drugs as model compounds. Sample introduction from 96 well plates, HPLC solvent delivery, mass spectrometric (MS) detection, and/or UV detection were fully integrated and operated unattended. After drugs were incubated with hepatocytes for three or six hr, LC-MS analyses were carried out to determine the amount of drug which was not metabolized. Two LC-MS methods were used which had a sample throughput of 4 samples/hr and 12 samples/hr. Under optimal conditions, this hepatic assay could screen 300 samples/week or 1200 samples/month. Although freshly isolated hepatocytes were more active, commercially available rat, monkey, and human primary isolated hepatocytes metabolized drug substrates in similar relative rank orders. This drug-hepatocyte assay provides useful information for prioritizing pharmaceutical leads in relative rank orders or in a high/low manner according to their resistance toward liver metabolism.


Subject(s)
Chromatography, Liquid/methods , Liver/metabolism , Mass Spectrometry/methods , Animals , Cells, Cultured , Drug-Related Side Effects and Adverse Reactions , Haplorhini , Humans , Liver/cytology , Liver/drug effects , Rats , Spectrophotometry, Ultraviolet
17.
Biochem Biophys Res Commun ; 259(2): 271-82, 1999 Jun 07.
Article in English | MEDLINE | ID: mdl-10362498

ABSTRACT

Two proteins, FKBP, and Spo0F, were expressed in bacteria as histidine-tagged fusion proteins and isolated under native conditions. MALDI-TOF-MS analysis revealed that each protein preparation contained two components, neither of which corresponded to the molecular weights predicted from DNA sequences. The difference in molecular weight between the two FKBP components and two Spo0F components was approximately 178 +/- 14 Da. Site-specific proteolytic cleavage resulted in the release of histidine-tagged peptide from the recombinant proteins. MALDI mass spectra of the cleaved proteins showed a single molecular ion peak for each species with the predicted molecular weights. The histidine-tagged peptide released from both fusion proteins displayed two distinct peaks by MALDI-FT-MS corresponding to monoisotopic molecular weights of 2269. 027 Da and 2447.087 Da, respectively, which were both inconsistent with the predicted peptide sequence M-G-H-H-H-H-H-H-H-H-H-H-S-S-G-H-I-E-G-R of 2400.055 Da. The peptide at 2269.027 Da was sequenced by ESI-MS-MS and found to be a truncated histidine tag resulting from an initiator methionine deletion. ESI-MS-MS analysis of the peptide at 2447.087 Da indicated a moiety of 178.0 Da attached to the second residue glycine of the histidine tag. This alteration of the N-terminus does not fit any known modifications. A synthetic peptide with the identical sequence of the isolated his-tag M-G-H-H-H-H-H-H-H-H-H-H remained unmodified during the protein purification process, suggesting that modification of the initiator methionine was carried out in vivo, rather than the result of a chemical reaction from the isolation procedure.


Subject(s)
Bacterial Proteins/chemistry , Histidine/chemistry , Immunophilins/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Cells, Cultured , Escherichia coli , Factor Xa , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Protein Processing, Post-Translational , Sequence Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tacrolimus Binding Proteins
18.
Xenobiotica ; 28(10): 957-72, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9849643

ABSTRACT

1. The excretion and metabolism of N-[2-(3,4-dimethoxyphenyl)ethyl]-5-methoxy-N,alpha-dimethyl-2-(phenyl ethynyl) benzenepropanamine (RWJ-26240) in the Wistar rat has been investigated after a single oral dose of 14C-RWJ-26240 (50 mg/kg free base). 2. Plasma samples were obtained for 24 h after dosing and urine and faecal samples were collected over 8 days, and they accounted for 0.9 and 96% of the dose, respectively. 3. Representative samples of plasma, urine and faecal samples were purified for metabolite isolation and identification using HPLC, tlc, mass spectra (CI and EI), 1H-NMR and derivatization. 4. Unchanged RWJ-26240 plus 11 metabolites were identified and accounted for > 80% of the sample radioactivity. 5. Four metabolic pathways for RWJ-26240 are proposed; namely (1) N-demethylation, (2) O-demethylation, (3) phenyl hydroxylation and (4) N-dealkylation. Pathways 1-3 appeared to be quantitatively more important.


Subject(s)
Antihypertensive Agents/metabolism , Antihypertensive Agents/pharmacokinetics , Propylamines/metabolism , Propylamines/pharmacology , Animals , Antihypertensive Agents/chemistry , Biotransformation , Chromatography, High Pressure Liquid , Feces/chemistry , Female , Male , Propylamines/chemistry , Rats , Rats, Wistar , Urine/chemistry
19.
J Chromatogr A ; 810(1-2): 95-103, 1998 Jun 12.
Article in English | MEDLINE | ID: mdl-9691294

ABSTRACT

We have demonstrated that significant differences exist between the retention of eight beta-blockers analyzed with immobilized artificial membrane (IAM) and lysophospholipid micellar electrokinetic capillary (LMEKC) chromatographic methods. The general retention trends are maintained with highly hydrophilic compounds such as atenolol eluting first and more hydrophobic compounds such as propranolol eluting last. The retention order, however, is different and would result in major ranking differences. LMEKC demonstrates a better correlation with liposomal partitioning (R2 = 0.95) than does IAM chromatography (R2 = 0.60). LMEKC, with its higher efficiency, can allow a more specific evaluation of lipophilicity than IAM chromatography and is useful in the analysis of pharmaceutical candidates, particularly for ranking purposes.


Subject(s)
Adrenergic beta-Antagonists/chemistry , Lysophospholipids/chemistry , Algorithms , Calibration , Chromatography , Electrochemistry , Indicators and Reagents , Membranes, Artificial , Micelles
20.
J Mass Spectrom ; 33(7): 607-14, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9692244

ABSTRACT

It is demonstrated that the apparent permeability (P(app)) coefficients of beta-adrenoceptor antagonist drugs can easily be determined for Caco-2 cell culture intestinal models utilizing liquid chromatography/mass spectrometry (LC/MS). The LC/MS method with electrospray ionization in the single ion monitoring mode showed an increased sensitivity of 1000-fold compared with LC/UV detection and enhanced selectivity with respect to both LC/UV and radioactivity assays. The P(app) coefficients of beta-adrenoceptor antagonists determined by LC/MS have the same ranking order as those determined by LC/UV and radioactivity assays. However, the P(app) coefficients determined in this study showed significant discrepancies from those determined in other laboratories. There are several experimental factors that directly affect the absolute value of the P(app) coefficients, including pH gradients, additional diffusion barriers (i.e. unstirred water layer and type of filter support), analyte concentration, detection method and possibly cell culture variations. These parameters should be controlled when generating Caco-2 P(app) coefficients for different compounds.


Subject(s)
Adrenergic beta-Antagonists/metabolism , Chromatography, Liquid/methods , Intestinal Mucosa/metabolism , Mass Spectrometry/methods , Acebutolol/metabolism , Adrenergic beta-Antagonists/analysis , Adrenergic beta-Antagonists/pharmacokinetics , Alprenolol/metabolism , Atenolol/metabolism , Biological Availability , Caco-2 Cells , Cell Membrane Permeability , Epithelial Cells/metabolism , Humans , Intestinal Absorption , Intestinal Mucosa/cytology , Metoprolol/metabolism , Molecular Structure , Oxprenolol/metabolism , Pindolol/metabolism , Propranolol/metabolism , Research Design , Timolol/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...