Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(11): e0206279, 2018.
Article in English | MEDLINE | ID: mdl-30427871

ABSTRACT

Two chemotypes were examined in vitro with CYPs 3A4 and 2C19 by molecular docking, metabolic profiles, and intrinsic clearance deuterium isotope effects with specifically deuterated form to assess the potential for enhancement of pharmacokinetic parameters. The results show the complexity of deuteration as an approach for pharmacokinetic enhancement when CYP enzymes are involved in metabolic clearance. With CYP3A4 the rate limiting step was chemotype-dependent. With one chemotype no intrinsic clearance deuterium isotope effect was observed with any deuterated form, whereas with the other chemotype the rate limiting step was isotopically sensitive, and the magnitude of the intrinsic clearance isotope effect was dependent on the position(s) and extent of deuteration. Molecular docking and metabolic profiles aided in identifying sites for deuteration and predicted the possibility for metabolic switching. However, the potential for an isotope effect on the intrinsic clearance cannot be predicted and must be established by examining select deuterated versions of the chemotypes. The results show how in a deuteration strategy molecular docking, in-vitro metabolic profiles, and intrinsic clearance assessments with select deuterated versions of new chemical entities can be applied to determine the potential for pharmacokinetic enhancement in a discovery setting. They also help explain the substantial failures reported in the literature of deuterated versions of drugs to elicit a systemic enhancement on pharmacokinetic parameters.


Subject(s)
Cytochrome P-450 CYP2C19/chemistry , Cytochrome P-450 CYP3A/chemistry , Deuterium/chemistry , Pharmacokinetics , Cytochrome P-450 CYP2C19/radiation effects , Cytochrome P-450 CYP3A/radiation effects , Deuterium/pharmacology , Heme/chemistry , Heme/radiation effects , Humans , Inactivation, Metabolic , Kinetics , Microsomes/radiation effects , Molecular Docking Simulation , Oxidation-Reduction/radiation effects , Substrate Specificity
2.
Rapid Commun Mass Spectrom ; 29(22): 2175-83, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26467230

ABSTRACT

RATIONALE: The covalent modification of proteins by toxicants, new chemical entities or drug molecules, either by metabolic activation or the presence of inherently reactive functional groups, is commonly implicated in organ toxicity and idiosyncratic reactions. In efforts to better prosecute protein modifications, we investigated a tag-free technique capable of detecting protein-small molecule adducts based solely on the collision-induced dissociation (CID) of the protein-small molecule complex. Detection of proteins using unique CID small molecule (SM) product ions would mitigate common issues associated with tagging technologies (e.g., altered reactivity/affinity of the protein-SM complex). METHODS: A Waters SYNAPT G2 mass spectrometer (MS) was operated in MS(e) mode with appropriate collision energy conditions during the MS(2) acquisition for fragmentation of protein-small molecule adducts to generate characteristic small molecule product ions. RESULTS: Ibrutinib, an acrylamide-containing small molecule drug, was shown to form adducts with rat serum albumin in ex vivo experiments and these adducts were detected by relying solely on the CID product ions generated from ibrutinib. Additionally, ibrutinib produced three CID product ions, one of which was a selective protein-ibrutinib fragment ion not produced by the compound alone. CONCLUSIONS: Herein we describe a tag-free mass spectral detection technique for protein-small molecule conjugates that relies on the unique product ion fragmentation profile of the small molecule. This technique allows the detection of macromolecular ions containing the adducted small molecule from complex protein matrices through mass range selection for the unique product ions in the CID spectra.


Subject(s)
Ions/chemistry , Mass Spectrometry/methods , Pharmaceutical Preparations/chemistry , Proteins/chemistry , Adenine/analogs & derivatives , Animals , Chromatography, High Pressure Liquid , Humans , Ions/analysis , Ions/metabolism , Models, Chemical , Pharmaceutical Preparations/metabolism , Piperidines , Proteins/analysis , Proteins/metabolism , Pyrazoles , Pyrimidines , Rats
3.
J Med Chem ; 57(4): 1616-20, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24472070

ABSTRACT

The bioavailability of aromatic azaheterocyclic drugs can be affected by the activity of aldehyde oxidase (AO). Susceptibility to AO metabolism is difficult to predict computationally and can be complicated in vivo by differences between species. Here we report the use of bis(((difluoromethyl)sulfinyl)oxy)zinc (DFMS) as a source of CF2H radical for a rapid and inexpensive chemical "litmus test" for the early identification of heteroaromatic drug candidates that have a high probability of metabolism by AO.


Subject(s)
Aldehyde Oxidase/metabolism , Hydrocarbons/metabolism
4.
PLoS One ; 7(4): e34822, 2012.
Article in English | MEDLINE | ID: mdl-22514671

ABSTRACT

BACKGROUND: Atherosclerosis starts by lipid accumulation in the arterial intima and progresses into a chronic vascular inflammatory disease. A major atherogenic process is the formation of lipid-loaded macrophages in which a breakdown of the endolysomal pathway results in irreversible accumulation of cargo in the late endocytic compartments with a phenotype similar to several forms of lipidosis. Macrophages exposed to oxidized LDL exihibit this phenomenon in vitro and manifest an impaired degradation of internalized lipids and enhanced inflammatory stimulation. Identification of the specific chemical component(s) causing this phenotype has been elusive because of the chemical complexity of oxidized LDL. METHODOLOGY/PRINCIPAL FINDINGS: Lipid "core aldehydes" are formed in oxidized LDL and exist in atherosclerotic plaques. These aldehydes are slowly oxidized in situ and (much faster) by intracellular aldehyde oxidizing systems to cholesteryl hemiesters. We show that a single cholesteryl hemiester incorporated into native, non-oxidized LDL induces a lipidosis phenotype with subsequent cell death in macrophages. Internalization of the cholesteryl hemiester via the native LDL vehicle induced lipid accumulation in a time- and concentration-dependent manner in "frozen" endolysosomes. Quantitative shotgun lipidomics analysis showed that internalized lipid in cholesteryl hemiester-intoxicated cells remained largely unprocessed in those lipid-rich organelles. CONCLUSIONS/SIGNIFICANCE: The principle elucidated with the present cholesteryl hemiester-containing native-LDL model, extended to other molecular components of oxidized LDL, will help in defining the molecular etiology and etiological hierarchy of atherogenic agents.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Lipidoses/chemically induced , Macrophages/drug effects , Macrophages/metabolism , Animals , Cell Line , Cell Survival/drug effects , Electrophoresis, Agar Gel , Fluorescent Antibody Technique , Lipoproteins, LDL/pharmacology , Mice , Microscopy, Confocal
5.
Drug Metab Dispos ; 40(3): 625-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190693

ABSTRACT

The pharmacokinetic properties of drugs may be altered by kinetic deuterium isotope effects. With specifically deuterated model substrates and drugs metabolized by aldehyde oxidase, we demonstrate how knowledge of the enzyme's reaction mechanism, species differences in the role played by other enzymes in a drug's metabolic clearance, and differences in systemic clearance mechanisms are critically important for the pharmacokinetic application of deuterium isotope effects. Ex vivo methods to project the in vivo outcome using deuterated carbazeran and zoniporide with hepatic systems demonstrate the importance of establishing the extent to which other metabolic enzymes contribute to the metabolic clearance mechanism. Differences in pharmacokinetic outcomes in guinea pig and rat, with the same metabolic clearance mechanism, show how species differences in the systemic clearance mechanism can affect the in vivo outcome. Overall, to gain from the application of deuteration as a strategy to alter drug pharmacokinetics, these studies demonstrate the importance of understanding the systemic clearance mechanism and knowing the identity of the metabolic enzymes involved, the extent to which they contribute to metabolic clearance, and the extent to which metabolism contributes to the systemic clearance.


Subject(s)
Aldehyde Oxidase/metabolism , Carbamates/pharmacokinetics , Deuterium/metabolism , Guanidines/pharmacokinetics , Pyrazoles/pharmacokinetics , Animals , Carbamates/metabolism , Cytosol/metabolism , Guanidines/metabolism , Guinea Pigs , Hepatocytes/metabolism , Humans , Kinetics , Liver/metabolism , Male , Pyrazoles/metabolism , Rats , Rats, Sprague-Dawley
6.
Rapid Commun Mass Spectrom ; 24(14): 2151-61, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20552706

ABSTRACT

Oxidation of N-alkyl-substituted amides is a common transformation observed in metabolism studies of drugs and other chemicals. Metabolism at the alpha carbon atom can produce stable carbinolamide compounds, which may be abundant enough to require complete confidence in structural assignments. In a drug discovery setting, rapid structural elucidation of test compounds is critical to inform the compound selection process. Traditional approaches to the analysis of carbinolamides have relied upon the time-consuming synthesis of authentic standards or purification of large enough quantities for characterization by nuclear magnetic resonance (NMR). We describe a simple technique used in conjunction with liquid chromatography/tandem mass spectrometry (LC/MS/MS) which demonstrates the chemical identity of a carbinolamide by its distinctive ability to reversibly exchange [(18)O]water through an imine intermediate. A key advantage of the technique is that the chromatographic retention times of metabolites are preserved, allowing direct comparisons of mass chromatograms from non-treated and [(18)O]water-treated samples. Metabolites susceptible to the treatment are clearly indicated by the addition of 2 mass units to their original mass. An additional test which can be used in conjunction with (18)O-exchange is base-catalyzed N-dealkylation of N-(alpha-hydroxy)alkyl compounds. The use of the technique is described for carbinolamide metabolites of dirlotapide, loperamide, and a proprietary compound.


Subject(s)
Carbamates/chemistry , Carbamates/metabolism , Chromatography, Liquid/methods , Indoles/chemistry , Indoles/metabolism , Oxygen Isotopes/analysis , Tandem Mass Spectrometry/methods , Animals , Carbamates/administration & dosage , Catalysis , Dealkylation , Female , Indoles/administration & dosage , Male , Molecular Structure , Rats , Rats, Sprague-Dawley
7.
Rapid Commun Mass Spectrom ; 24(14): 2109-21, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20552710

ABSTRACT

A simple procedure is described to identify acyl-glucuronides by coupled liquid chromatography/mass spectrometry after derivatization to a hydroxamic acid with hydroxylamine. The reaction specificity obviates the need for isolation of the acyl-glucuronide from an extract. Glucuronides derived from carbamic acids, and alkyl- and aromatic amines, are inert to the derivatization reaction conditions, making the hydroxamic acid derivative a fingerprint for acyl-glucuronides.


Subject(s)
Glucuronides/chemistry , Mass Spectrometry/methods , Carbamates/chemistry , Hydroxamic Acids/chemistry , Hydroxylamine/chemistry
8.
J Med Chem ; 52(23): 7446-57, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19775168

ABSTRACT

Respiratory tract bacterial strains are becoming increasingly resistant to currently marketed macrolide antibiotics. The current alternative telithromycin (1) from the newer ketolide class of macrolides addresses resistance but is hampered by serious safety concerns, hepatotoxicity in particular. We have discovered a novel series of azetidinyl ketolides that focus on mitigation of hepatotoxicity by minimizing hepatic turnover and time-dependent inactivation of CYP3A isoforms in the liver without compromising the potency and efficacy of 1.


Subject(s)
Azetidines/chemistry , Drug Resistance, Multiple/drug effects , Ketolides/chemistry , Ketolides/pharmacology , Respiratory Tract Infections/drug therapy , Animals , Bacteria/drug effects , Community-Acquired Infections/drug therapy , Disease Susceptibility , Drug Discovery , Drug-Related Side Effects and Adverse Reactions , Humans , Ketolides/adverse effects , Ketolides/chemical synthesis , Ketolides/therapeutic use , Mice , Microbial Sensitivity Tests
9.
Chem Biol Interact ; 155(1-2): 10-20, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-15978881

ABSTRACT

Therapy with the antidepressant trazodone has been associated with several cases of idiosyncratic hepatotoxicity. While the mechanism of hepatotoxicity remains unknown, it is possible that reactive metabolites of trazodone play a causative role. Studies were initiated to determine whether trazodone undergoes bioactivation in human liver microsomes to electrophilic intermediates. LC/MS/MS analysis of incubations containing trazodone and NADPH-supplemented microsomes or recombinant P4503A4 in the presence of glutathione revealed the formation of conjugates derived from the addition of the sulfydryl nucleophile to mono-hydroxylated- and hydrated-trazodone metabolites. Product ion spectra suggested that mono-hydroxylation and sulfydryl conjugation occurred on the 3-chlorophenyl-ring, whereas hydration and subsequent sulfydryl conjugation had occurred on the triazolopyridinone ring system. These findings are consistent with bioactivation sequences involving: (1) aromatic hydroxylation of the 3-chlorophenyl-ring in trazodone followed by the two-electron oxidation of this metabolite to a reactive quinone-imine intermediate, which reacts with glutathione in a 1,4-Michael fashion and (2) oxidation of the pyridinone ring to an electrophilic epoxide, ring opening of which, by glutathione or water generates the corresponding hydrated-trazodone-thiol conjugate or the stable diol metabolite, respectively. The pathway involving trazodone bioactivation to the quinone-imine has also been observed with many para-hydroxyanilines including the structurally related antidepressant nefazodone. It is proposed that the quinone-imine and/or the epoxide intermediate(s) may represent a rate-limiting step in the initiation of trazodone-mediated hepatotoxicity.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacokinetics , Benzoquinones/metabolism , Cytochrome P-450 Enzyme System/metabolism , Epoxy Compounds/metabolism , Microsomes, Liver/metabolism , Trazodone/pharmacokinetics , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Humans , Imines/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Microsomes, Liver/drug effects , Recombinant Proteins , Spectrometry, Mass, Electrospray Ionization
10.
Biochim Biophys Acta ; 1707(2-3): 157-69, 2005.
Article in English | MEDLINE | ID: mdl-15863094

ABSTRACT

We report the molecular cloning, expression and partial characterization of MT FdR, an FAD-associated flavoprotein, from Mycobacterium tuberculosis similar to the oxygenase-coupled NADH-dependent ferredoxin reductases (ONFR). We establish, through kinetic and spectral analysis, that MT FdR preferentially uses NADH as cofactor. Furthermore, MT FdR forms a complex with mycobacterial ferredoxin (MT Fdx) and MT CYP51, a cytochrome P450 (CYP) from M. tuberculosis that is similar to lanosterol 14alpha-demethylase isozymes. This reconstituted system transfers electrons from the cofactor to the heme iron of MT CYP51 and effects the demethylation of lanosterol.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/enzymology , Oxidoreductases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cloning, Molecular , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Electron Transport , Ferredoxins/metabolism , Kinetics , Lanosterol/metabolism , Multiprotein Complexes , NAD , Oxidoreductases/chemistry , Oxidoreductases/genetics , Protein Binding , Spectrum Analysis
11.
Drug Metab Dispos ; 33(2): 243-53, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15523046

ABSTRACT

The therapeutic benefits of the antidepressant nefazodone have been hampered by several cases of acute hepatotoxicity/liver failure. Although the mechanism of hepatotoxicity remains unknown, it is possible that reactive metabolites of nefazodone play a causative role. Studies were initiated to determine whether nefazodone undergoes bioactivation in human liver microsomes to electrophilic intermediates. Following incubation of nefazodone with microsomes or recombinant P4503A4 in the presence of sulfydryl nucleophiles, conjugates derived from the addition of thiol to a monohydroxylated nefazodone metabolite were observed. Product ion spectra suggested that hydroxylation and sulfydryl conjugation occurred on the 3-chlorophenylpiperazine-ring, consistent with a bioactivation pathway involving initial formation of p-hydroxynefazodone, followed by its two-electron oxidation to the reactive quinone-imine intermediate. The formation of novel N-dearylated nefazodone metabolites was also discernible in these incubations, and 2-chloro-1,4-benzoquinone, a by-product of N-dearylation, was trapped with glutathione to afford the corresponding hydroquinone-sulfydryl adduct. Nefazodone also displayed NADPH-, time-, and concentration-dependent inactivation of P4503A4 activity, suggesting that reactive metabolites derived from nefazodone bioactivation are capable of covalently modifying P4503A4. A causative role for 2-chloro-1,4-benzoquinone and/or the quinone-imine intermediate(s) in nefazodone hepatotoxicity is speculated. Although the antianxiety agent buspirone, which contains a pyrimidine ring in place of the 3-chlorophenyl-ring, also generated p-hydroxybuspirone in liver microsomes, no sulfydryl conjugates of this metabolite were observed. This finding is consistent with the proposal that two-electron oxidation of p-hydroxybuspirone to the corresponding quinone-imine is less favorable due to differences in the protonation state at physiological pH and due to weaker resonance stabilization of the oxidation products as predicted from ab initio measurements.


Subject(s)
Antidepressive Agents, Second-Generation/metabolism , Benzoquinones/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Triazoles/metabolism , Antidepressive Agents, Second-Generation/analysis , Antidepressive Agents, Second-Generation/chemistry , Benzoquinones/analysis , Benzoquinones/chemistry , Biotransformation , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Humans , Imines/analysis , Imines/chemistry , Imines/metabolism , Piperazines , Triazoles/analysis , Triazoles/chemistry
12.
Drug Metab Dispos ; 31(10): 1240-50, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12975333

ABSTRACT

The 3-unsubstituted isoxazole ring in the anti-inflammatory drug leflunomide undergoes a unique N-O bond cleavage to the active alpha-cyanoenol metabolite A771726, which resides in the same oxidation state as the parent. In vitro studies were conducted to characterize drug-metabolizing enzyme(s) responsible for ring opening and to gain insight into the mechanism of ring opening. Under physiological conditions, leflunomide was converted to A771726 in rat and human plasma (rat plasma,t1/2 = 36 min; human plasma, t1/2 = 12 min) and whole blood (rat blood, t1/2 = 59 min; human blood, t1/2 = 43 min). Human serum albumin also catalyzed A771726 formation, albeit at a much slower rate (t1/2 = 110 min). Rat and human liver microsomes also demonstrated NADPH-dependent A771726 formation (human liver microsomes, Vmax = 1797 pmol/min/mg and Km = 274 microM). Leflunomide metabolism in microsomes was sensitive to furafylline treatment, suggesting p4501A2 involvement. 3-Methylleflunomide, which contained a 3-methyl substituent on the isoxazole ring, was resistant to ring opening in base, plasma, blood, and liver microsomes. In microsomes, two monohydroxylated metabolites were formed, and metabolite identification studies established the 3- and the 5-methyl groups on the isoxazole ring as sites of hydroxylation. These results indicate that the C3-H in leflunomide is essential for ring opening. Although A771726 formation in human liver microsomes or recombinant p4501A2 required NADPH, its formation was greatly reduced by oxygen or carbon monoxide, suggesting that the isoxazole ring opening was catalyzed by the p450Fe(II) form of the enzyme. A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond.


Subject(s)
Aniline Compounds/metabolism , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytochrome P-450 Enzyme System/metabolism , Hydroxybutyrates/metabolism , Isoxazoles/metabolism , Oximes/metabolism , Aniline Compounds/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Crotonates , Desiccation/methods , Humans , Hydroxybutyrates/chemistry , Isoxazoles/chemistry , Leflunomide , Male , Microsomes, Liver/metabolism , Nitriles , Oximes/chemistry , Rats , Rats, Sprague-Dawley , Toluidines
13.
Drug Metab Dispos ; 31(8): 1005-15, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12867489

ABSTRACT

Clinically observed drug interactions with cytochrome p450 (p450) enzymes have increased the need to assess drug interactions of new chemical entities early in the discovery process. To meet this need, fluorogenic substrates have been commercialized. However, only limited evaluations of their utility and comparisons to drug probes have been reported. This study examines the correlation between IC50 values obtained with fluorogenic and conventional drug probes for structurally diverse inhibitors of the five major human p450 isoforms. In general, correlations are weak, with significant numbers of compounds being missed as inhibitors by either probe. For p450s 1A2, 2C9, and 2C19, correlation coefficients were above 0.6 with slopes that ranged from 1.5 to 4.2. However, for p450s 1A2 and 2C9, about 20% of compounds were not included because an IC50 value could not be determined with one of the two probes. CYP 2C19 had the highest correlation (correlation coefficient 0.84), with a slope of 2.0 and less than 5% of compounds excluded. CYP 2D6 showed a good correlation for IC50 values less than 10 microM. However, at higher IC50 values, a high degree of scatter was observed. CYP 3A4 had the weakest correlation, and a large number of compounds were excluded with the fluorogenic probe. Overall, the study shows the care needed when selecting fluorogenic probes and the caution needed when results with fluorogenic probes are used to drive structure-activity relationships with respect to drug interactions.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Chemistry, Pharmaceutical , Cytochrome P-450 Enzyme Inhibitors , Drug Evaluation, Preclinical , Drug Industry , Drug Interactions , Fluorescent Dyes/economics , Fluorescent Dyes/metabolism , Guidelines as Topic , Inhibitory Concentration 50 , Research Design , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...