Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Publication year range
1.
Drug Metab Dispos ; 51(6): 782-791, 2023 06.
Article in English | MEDLINE | ID: mdl-36921993

ABSTRACT

The anti-hypertensive agent hydralazine is a time-dependent inhibitor of the cytosolic drug-metabolizing enzyme aldehyde oxidase (AO). Glutathione (GSH) was found to suppress the inhibition of AO by hydralazine in multiple enzyme sources (human liver and kidney cytosol, human liver S9, rat liver S9, and recombinant human AO) and with different AO substrates (zoniporide, O6 -benzylguanine, and dantrolene). Hydralazine-induced AO inactivation was unaffected when GSH was added to the incubation mixture after pre-incubation of hydralazine with AO (rather than during the pre-incubation), suggesting that GSH traps a hydralazine reactive intermediate prior to enzyme inactivation. Consistent with previous reports of 1-phthalazylmercapturic acid formation when hydralazine was incubated with N-acetylcysteine, we detected a metabolite producing an MS/MS spectrum consistent with a 1-phthalazyl-GSH conjugate. O6 -Benzylguanine, an AO substrate, did not protect against hydralazine-induced AO inactivation, implying that hydralazine does not compete with O6 -benzylguanine for binding to the AO active site. Catalase also failed to protect AO from hydralazine-induced inactivation, suggesting that hydrogen peroxide is not involved. However, an allosteric AO inhibitor (thioridazine) offered some protection, indicating a catalytic role for AO in the bioactivation of hydralazine. AO inhibition by phthalazine (a substrate and inhibitor of AO and a metabolite of hydralazine) was unaffected by the presence of GSH. GSH also prevented hydralazine from inhibiting the nitro-reduction of dantrolene by AO. Furthermore, the GSH-hydralazine combination stimulated dantrolene reduction. Phthalazine inhibited only oxidation reactions, not reduction of dantrolene. Together, these results support the hypothesis that hydralazine is converted to a reactive intermediate that inactivates AO. SIGNIFICANCE STATEMENT: These studies suggest that a reactive intermediate of hydralazine plays a primary role in the mechanism of aldehyde oxidase (AO) inactivation. Inactivation was attenuated by glutathione and unaffected by catalase. Phthalazine (hydralazine metabolite) inhibited AO regardless of the presence of glutathione; however, phthalazine inhibited only oxidation reactions, while hydralazine inhibited both oxidation and reduction reactions. This report advances our mechanistic understanding of hydralazine as an AO inhibitor and provides information to facilitate appropriate use of hydralazine when probing AO metabolism.


Subject(s)
Aldehyde Oxidase , Tandem Mass Spectrometry , Rats , Animals , Humans , Aldehyde Oxidase/metabolism , Catalase , Dantrolene , Hydralazine/pharmacology , Phthalazines/metabolism , Glutathione
2.
Molecules ; 26(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807659

ABSTRACT

G-quadruplexes (G4) are the most actively studied non-canonical secondary structures formed by contiguous repeats of guanines in DNA or RNA strands. Small molecule mediated targeting of G-quadruplexes has emerged as an attractive tool for visualization and stabilization of these structures inside the cell. Limited number of DNA and RNA G4-selective assays have been reported for primary ligand screening. A combination of fluorescence spectroscopy, AFM, CD, PAGE, and confocal microscopy have been used to assess a dimeric carbocyanine dye B6,5 for screening G4-binding ligands in vitro and in cellulo. The dye B6,5 interacts with physiologically relevant DNA and RNA G4 structures, resulting in fluorescence enhancement of the molecule as an in vitro readout for G4 selectivity. Interaction of the dye with G4 is accompanied by quadruplex stabilization that extends its use in primary screening of G4 specific ligands. The molecule is cell permeable and enables visualization of quadruplex dominated cellular regions of nucleoli using confocal microscopy. The dye is displaced by quarfloxin in live cells. The dye B6,5 shows remarkable duplex to quadruplex selectivity in vitro along with ligand-like stabilization of DNA G4 structures. Cell permeability and response to RNA G4 structures project the dye with interesting theranostic potential. Our results validate that B6,5 can serve the dual purpose of visualization of DNA and RNA G4 structures and screening of G4 specific ligands, and adds to the limited number of probes with such potential.


Subject(s)
Carbocyanines/chemistry , Carbocyanines/metabolism , G-Quadruplexes , Molecular Imaging/methods , DNA/chemistry , DNA/metabolism , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Ligands , Nucleic Acid Conformation , Phthalazines/chemistry , Phthalazines/metabolism , Piperazines/chemistry , Piperazines/metabolism , Porphyrins/chemistry , Porphyrins/metabolism , RNA/chemistry , RNA/metabolism , Taq Polymerase/chemistry , Taq Polymerase/metabolism
3.
J Biol Chem ; 294(52): 19923-19933, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31732559

ABSTRACT

Plant development is regulated by both synergistic and antagonistic interactions of different phytohormones, including a complex crosstalk between ethylene and auxin. For instance, auxin and ethylene synergistically control primary root elongation and root hair formation. However, a lack of chemical agents that specifically modulate ethylene or auxin production has precluded precise delineation of the contribution of each hormone to root development. Here, we performed a chemical genetic screen based on the recovery of root growth in ethylene-related Arabidopsis mutants with constitutive "short root" phenotypes (eto1-2 and ctr1-1). We found that ponalrestat exposure recovers root elongation in these mutants in an ethylene signal-independent manner. Genetic and pharmacological investigations revealed that ponalrestat inhibits the enzymatic activity of the flavin-containing monooxygenase YUCCA, which catalyzes the rate-limiting step of the indole-3-pyruvic acid branch of the auxin biosynthesis pathway. In summary, our findings have identified a YUCCA inhibitor that may be useful as a chemical tool to dissect the distinct steps in auxin biosynthesis and in the regulation of root development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Indoleacetic Acids/metabolism , Oxygenases/metabolism , Phthalazines/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ethylenes/metabolism , Indoles/chemistry , Indoles/metabolism , Molecular Docking Simulation , Mutagenesis , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Phenotype , Phthalazines/metabolism , Phthalazines/pharmacology , Plant Roots/drug effects , Plant Roots/growth & development , Plant Roots/metabolism , Protein Structure, Tertiary , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Rapid Commun Mass Spectrom ; 34(3): e8575, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31499579

ABSTRACT

RATIONALES: Olaparib is a Poly (ADP-ribose) Polymerase (PARP) inhibitor which has been developed as an anti-cancer agent. The purpose of this study was to characterize the metabolites of olaparib from liver microsomes and to reveal the interspecies differences between animals and humans. METHODS: Olaparib (20 µM) was incubated with different species of liver microsomes at 37°C for 1 h in the presence of NADPH. The incubation samples were analyzed by liquid chromatography/electrospray ionization high-resolution mass spectrometry (LC/ESI-HRMS) operated in positive ion mode. The metabolites were characterized by accurate masses, MS2 spectra and retention times. RESULTS: A total of 12 metabolites were detected and the structures of the metabolites were characterized based on their accurate masses, fragment ions and retention times. Four metabolites, i.e., M1, M10, M11 and M12, were unambiguously identified by using reference standards. The metabolic pathways of olaparib included hydroxylation, bis-hydroxylation, hydrolysis, dealkylation, dehydrogenation, and alcohol oxidation. CONCLUSIONS: Compared with animal species, no human-specific metabolite was found in HLM. Dog also had a closer metabolic profile to humans. This study will be helpful for a better understanding of the species difference in pharmacokinetics/pharmacodynamics.


Subject(s)
Chromatography, Liquid/methods , Microsomes, Liver/metabolism , Phthalazines/analysis , Phthalazines/metabolism , Piperazines/analysis , Piperazines/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Biotransformation , Dogs , Humans , Macaca fascicularis , Mice , Rats , Rats, Sprague-Dawley
5.
Bioorg Med Chem Lett ; 30(23): 127556, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32941989

ABSTRACT

A series of novel 4-substituted phthalazinones as Aurora B kinase inhibitors was synthesized and evaluated the anti-proliferative activities against A549, HCT116, MCF-7 and HepG2 cells. 1-(4-(2-((4-Oxo-3,4-dihydrophthalazin-1-yl)amino)ethyl) phenyl)-3-(3-(trifluoromethyl)phenyl)urea (17b) exhibited the most potent anti-proliferative activity against HCT116 cells with IC50 value of 4.35 ± 1.21 µM, as well as the moderate Aurora B inhibitory activity with the IC50 value of 142 nM. Furthermore, 17b inhibited the phosphorylation of Aurora B on Thr232, leading to cell cycle arrest in the G2/M phase by down-regulating the expression of CyclinB1 and Cdc2 proteins, and apoptosis by up-regulating the expression of BAD and Bax proteins in HCT116 cells. In addition, a docking study revealed that 17b could form key hydrogen bonds with Ala173, Glu171 and Glu177 in Aurora B. All the results reveal that 17b is worthy of further development as an Aurora B kinase inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase B/antagonists & inhibitors , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Aurora Kinase B/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Assays , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , Phosphorylation/drug effects , Phthalazines/chemical synthesis , Phthalazines/metabolism , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Structure-Activity Relationship
6.
Bioorg Chem ; 103: 104233, 2020 10.
Article in English | MEDLINE | ID: mdl-32882440

ABSTRACT

Herein we report the design and synthesis of a new series of phthalazine derivatives as Topo II inhibitors and DNA intercalators. The synthesized compounds were in vitro evaluated for their cytotoxic activities against HepG-2, MCF-7 and HCT-116 cell lines. Additionally, Topo II inhibitory activity and DNA intercalating affinity were investigated for the most active compounds as a potential mechanism for the anticancer activity. Compounds 15h, 23c, 32a, 32b, and 33 exhibited the highest activities against Topo II with IC50 ranging from 5.44 to 8.90 µM, while compounds 27 and 32a were found to be the most potent DNA binders at IC50 values of 36.02 and 48.30 µM, respectively. Moreover, compound 32a induced apoptosis in HepG-2 cells and arrested the cell cycle at the G2/M phase. Besides, compound 32a showed Topo II poisoning effect at concentrations of 2.5 and 5 µM, and Topo II catalytic inhibitory effect at a concentration of10 µM. In addition, compound 32b showed in vivo a significant tumor growth inhibition effect. Furthermore, molecular docking studies were carried out against DNA-Topo II complex and DNA to investigate the binding patterns of the designed compounds.


Subject(s)
Antineoplastic Agents/therapeutic use , Intercalating Agents/therapeutic use , Neoplasms/drug therapy , Phthalazines/therapeutic use , Topoisomerase II Inhibitors/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , DNA/metabolism , DNA Topoisomerases, Type II/metabolism , Drug Design , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Intercalating Agents/chemical synthesis , Intercalating Agents/metabolism , Molecular Docking Simulation , Molecular Structure , Phthalazines/chemical synthesis , Phthalazines/metabolism , Protein Binding , Rats , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism
7.
J Pharmacol Exp Ther ; 368(1): 79-87, 2019 01.
Article in English | MEDLINE | ID: mdl-30396915

ABSTRACT

Endogenous canine ATP binding cassette B1 (cABCB1) is expressed abundantly in Madin-Darby canine kidney type II (MDCKII) cells, and its presence often complicates phenotyping of the transport process. Errors in estimating the corrected efflux ratio (cER), as a result of the variable expression of cABCB1, were examined for the dual substrates of ABCB1 and ABCG2 in MDCKII cells expressing human ABCG2 (hABCG2). cABCB1 mRNA and protein expression was 60% and 55% lower, respectively, in MDCKII cells expressing hABCG2 compared with the wild type, suggesting that the expression of endogenous cABCB1 became variable after the expression of hABCG2. To minimize the contribution of endogenous efflux, cABCB1 was suppressed kinetically (using verapamil as a selective inhibitor) or biochemically (transfecting short-hairpin RNA against cABCB1). Under these suppression conditions, cER values for irinotecan and topotecan (dual substrates of ABCB1 and ABCG2) were elevated by more than 4-fold and 2-fold, respectively, compared with cER values without the suppression. The cER of olaparib was similarly increased to 3- and 5-fold in MDCKII cells under the kinetic and biochemical suppression of cABCB1, respectively, suggesting that hABCG2-mediated efflux cannot be ruled out for olaparib. Since the substrate selectivity for ABCB1 and ABCG2 overlapped considerably, the possibility of an inaccurate estimation of cER must be considered for dual substrates in the case of the variable expression of cABCB1 in MDCKII cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Neoplasm Proteins/metabolism , Phthalazines/metabolism , Piperazines/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Dogs , Dose-Response Relationship, Drug , Humans , LLC-PK1 Cells , Madin Darby Canine Kidney Cells , Phthalazines/pharmacology , Piperazines/pharmacology , Swine
8.
Drug Metab Dispos ; 47(5): 473-483, 2019 05.
Article in English | MEDLINE | ID: mdl-30787100

ABSTRACT

Many promising drug candidates metabolized by aldehyde oxidase (AOX) fail during clinical trial owing to underestimation of their clearance. AOX is species-specific, which makes traditional allometric studies a poor choice for estimating human clearance. Other studies have suggested using half-life calculated by measuring substrate depletion to measure clearance. In this study, we proposed using numerical fitting to enzymatic pathways other than Michaelis-Menten (MM) to avoid missing the initial high turnover rate of product formation. Here, product formation over a 240-minute time course of six AOX substrates-O6-benzylguanine, N-(2-dimethylamino)ethyl)acridine-4-carboxamide, zaleplon, phthalazine, BIBX1382 [N8-(3-Chloro-4-fluorophenyl)-N2-(1-methyl-4-piperidinyl)-pyrimido[5,4-d]pyrimidine-2,8-diamine dihydrochloride], and zoniporide-have been provided to illustrate enzyme deactivation over time to help better understand why MM kinetics sometimes leads to underestimation of rate constants. Based on the data provided in this article, the total velocity for substrates becomes slower than the initial velocity by 3.1-, 6.5-, 2.9-, 32.2-, 2.7-, and 0.2-fold, respectively, in human expressed purified enzyme, whereas the K m remains constant. Also, our studies on the role of reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, show that ROS did not significantly alter the change in enzyme activity over time. Providing a new electron acceptor, 5-nitroquinoline, did, however, alter the change in rate over time for mumerous compounds. The data also illustrate the difficulties in using substrate disappearance to estimate intrinsic clearance.


Subject(s)
Aldehyde Oxidase/metabolism , Acetamides/metabolism , Acridines/metabolism , Guanidines/metabolism , Humans , Hydralazine/metabolism , Kinetics , Liver/metabolism , Nitroquinolines/metabolism , Phthalazines/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Reactive Oxygen Species/metabolism
9.
Bioorg Chem ; 85: 293-307, 2019 04.
Article in English | MEDLINE | ID: mdl-30654221

ABSTRACT

Searching for new leads in the battle of cancer will never ends, we herein disclose the design and synthesis of new phthalazine derivatives and their in vitro and in vivo testing for their antiproliferative activity. Phthalazine was selected as a privilege moiety that is incorporated in a big number of anticancer drugs in clinical use or that are still under clinical or preclinical studies. We utilized the drug extension strategy to tailor the designed compounds to fit the EGFR hydrophobic sub pocket and cleft region. The designed phthalazine derivatives was synthesized by linking phthalazine moiety with 1,3,4-oxadiazole-thione and 1,2,4-triazole-thione. Alkylation and glycosylation of the new heterocyclic systems were successfully performed to be used in the drug extension. Coupling of some phthalazine derivatives with different amino acids was also performed to improve the drug selectivity. The synthesized compounds were tested for their antiproliferative activity against cancer cells both in vivo and in vitro. The in vitro activity against hepatocellular carcinoma (HepG2 cell line) ranged from 5.7 µg/mL to 43.4 µg/mL. Compounds 31a and 16 were the most active with an IC50 5.7 µg/mL and 7.09 µg/mL, respectively compared to the standard compound doxorubicin (4.0 µg/mL). In vivo, compounds 10 and 16 showed IC50 values 7.25 µg/mL and 7.5 µg/mL, respectively compared to the standard compound cisplatin (IC50 9.0 µg/mL). In silico, testing of the phthalazine derivatives showed that they are good inhibitors for EGFR. The docking studies substantiated compounds 4, 10, 16 and 31a as new lead compounds and identified Arg841 as a key residue in the cleft region for binding stronger inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Molecular Docking Simulation , Phthalazines/chemical synthesis , Phthalazines/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism
10.
Nature ; 497(7449): 338-43, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23636324

ABSTRACT

The smoothened (SMO) receptor, a key signal transducer in the hedgehog signalling pathway, is responsible for the maintenance of normal embryonic development and is implicated in carcinogenesis. It is classified as a class frizzled (class F) G-protein-coupled receptor (GPCR), although the canonical hedgehog signalling pathway involves the GLI transcription factors and the sequence similarity with class A GPCRs is less than 10%. Here we report the crystal structure of the transmembrane domain of the human SMO receptor bound to the small-molecule antagonist LY2940680 at 2.5 Å resolution. Although the SMO receptor shares the seven-transmembrane helical fold, most of the conserved motifs for class A GPCRs are absent, and the structure reveals an unusually complex arrangement of long extracellular loops stabilized by four disulphide bonds. The ligand binds at the extracellular end of the seven-transmembrane-helix bundle and forms extensive contacts with the loops.


Subject(s)
Antineoplastic Agents/chemistry , Phthalazines/chemistry , Receptors, G-Protein-Coupled/chemistry , Amino Acid Motifs , Amino Acid Sequence , Antineoplastic Agents/metabolism , Binding Sites , Crystallography, X-Ray , Disulfides/chemistry , Frizzled Receptors/chemistry , Frizzled Receptors/classification , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Phthalazines/metabolism , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , Structural Homology, Protein
11.
Bioorg Med Chem ; 26(12): 3217-3226, 2018 07 23.
Article in English | MEDLINE | ID: mdl-29705376

ABSTRACT

A series of 2,4-disubstituted phthalazinones were synthesized and their biological activities, including antiproliferation, inhibition against Aurora kinases and cell cycle effects were evaluated. Among them, N-cyclohexyl-4-((4-(1-methyl-1H-pyrazol-4-yl)-1-oxophthalazin-2(1H)-yl) methyl) benzamide (12c) exhibited the most potent antiproliferation against five carcinoma cell lines (HeLa, A549, HepG2, LoVo and HCT116 cells) with IC50 values in range of 2.2-4.6 µM, while the IC50 value of reference compound VX-680 was 8.5-15.3 µM. Moreover, Aurora kinase assays exhibited that compound 12c was potent inhibitor of AurA and AurB kinase with the IC50 values were 118 ±â€¯8.1 and 80 ±â€¯4.2 nM, respectively. Molecular docking studies indicated that compound 12c forms better interaction with both AurA and AurB. Furthermore, compound 12c induced G2/M cell cycle arrest in HeLa cells by regulating protein levels of cyclinB1 and cdc2. These results suggested that 12c is a promising pan-Aurora kinase inhibitor for the potential treatment of cancer.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Phthalazines/chemistry , Protein Kinase Inhibitors/chemical synthesis , Aurora Kinases/metabolism , Binding Sites , CDC2 Protein Kinase/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B1/metabolism , Down-Regulation/drug effects , Drug Design , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , HeLa Cells , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Phosphorylation/drug effects , Phthalazines/metabolism , Phthalazines/pharmacology , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
12.
Biomed Chromatogr ; 32(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-28677821

ABSTRACT

A sensitive and selective RP-HPLC method has been developed and validated for the quantification of a highly potent poly ADP ribose polymerase inhibitor talazoparib (TZP) in rat plasma. Chromatographic separation was performed with isocratic elution method. Absorbance for TZP was measured with a UV detector (SPD-20A UV-vis) at a λmax of 227 nm. Protein precipitation was used to extract the drug from plasma samples using methanol-acetonitrile (65:35) as the precipitating solvent. The method proved to be sensitive and reproducible over a 100-2000 ng/mL linearity range with a lower limit of quantification (LLQC) of 100 ng/mL. TZP recovery was found to be >85%. Following analytical method development and validation, it was successfully employed to determine the plasma protein binding of TZP. TZP has a high level of protein binding in rat plasma (95.76 ± 0.38%) as determined by dialysis method.


Subject(s)
Chromatography, High Pressure Liquid/methods , Phthalazines/blood , Animals , Blood Proteins/analysis , Blood Proteins/chemistry , Blood Proteins/metabolism , Chromatography, Reverse-Phase , Drug Stability , Linear Models , Phthalazines/chemistry , Phthalazines/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
13.
Xenobiotica ; 47(10): 903-915, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27684210

ABSTRACT

1. In vitro assessments were conducted to examine interactions between olaparib (a potent oral inhibitor of poly[ADP-ribose] polymerase) and drug transporters. 2. Olaparib showed inhibition of the hepatic drug uptake transporters OATP1B1 (IC50 values of 20.3 µM and 27.1 µM) and OCT1 (IC50 37.9 µM), but limited inhibition of OATP1B3 (25% at 100 µM); inhibition of the renal uptake transporters OCT2 (IC50 19.9 µM) and OAT3 (IC50 18.4 µM), but limited inhibition of OAT1 (13.5% at 100 µM); inhibition of the renal efflux transporters MATE1 and MATE2K (IC50s 5.50 µM and 47.1 µM, respectively); inhibition of the efflux transporter MDR1 (IC50 76.0 µM), but limited inhibition of BCRP (47% at 100 µM) and no inhibition of MRP2. At clinically relevant exposures, olaparib has the potential to cause pharmacokinetic interactions via inhibition of OCT1, OCT2, OATP1B1, OAT3, MATE1 and MATE2K in the liver and kidney, as well as MDR1 in the liver and GI tract. Olaparib was found to be a substrate of MDR1 but not of several other transporters. 3. Our assessments indicate that olaparib is a substrate of MDR1 and may cause clinically meaningful inhibition of MDR1, OCT1, OCT2, OATP1B1, OAT3, MATE1 and MATE2K.


Subject(s)
Antineoplastic Agents/metabolism , Drug Interactions , Phthalazines/metabolism , Piperazines/metabolism , Animals , Biological Transport , Humans , Liver-Specific Organic Anion Transporter 1/metabolism
14.
Postepy Hig Med Dosw (Online) ; 70: 280-94, 2016 Apr 13.
Article in Polish | MEDLINE | ID: mdl-27117104

ABSTRACT

Current cancer therapies are based mainly on the use of compounds that cause DNA damage. Unfortunately, even the combination therapies do not give rewarding effects, due to the high efficiency of DNA damage repair mechanisms in tumor cells. Therefore, the present studies should be focused on proteins that are involved in DNA repair systems. Poly(ADP-ribose) polymerase-1 is an example of a protein commonly known as an enzyme that plays a role in the detection of DNA damage and repair. Activation of PARP1 in response to DNA damage leads to poly-ADP-ribosylation of proteins contributing to DNA repair systems, therefore facilitating the maintenance of genome stability. On the other hand, inhibition of PARP1 enzyme results in the accumulation of DNA damage, which in turn contributes to cell death. Studies on inhibitors of PARP1 are still ongoing, and some of them are currently in the third phase of clinical trials. To date, only one representative of the PARP1 inhibitors, called olaparib, has been approved for anti-cancer therapy in the EU and the USA. Moreover, a growing body of evidence indicates a role of this protein in various intracellular processes such as bioenergetics, proliferation, regulation of gene expression, cell death as well as immunoregulation. A number of different intracellular processes regulated by PARP1 give rise to potential wider use of PARP1 inhibitors in treatment of other diseases, including immune or autoimmune disorders.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation , Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Antineoplastic Agents/metabolism , Cell Death , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA Replication/genetics , Humans , Neoplasms/enzymology , Phthalazines/metabolism , Piperazines/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerases/genetics
15.
BMC Bioinformatics ; 16: 308, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26403354

ABSTRACT

BACKGROUND: Searching for two-dimensional (2D) structural similarities is a useful tool to identify new active compounds in drug-discovery programs. However, as 2D similarity measures neglect important structural and functional features, similarity by 2D might be underestimated. In the present study, we used combined 2D and three-dimensional (3D) similarity comparisons to reveal possible new functions and/or side-effects of known bioactive compounds. RESULTS: We utilised more than 10,000 compounds from the SuperTarget database with known inhibition values for twelve different anti-cancer targets. We performed all-against-all comparisons resulting in 2D similarity landscapes. Among the regions with low 2D similarity scores are inhibitors of vascular endothelial growth factor receptor (VEGFR) and inhibitors of poly ADP-ribose polymerase (PARP). To demonstrate that 3D landscape comparison can identify similarities, which are untraceable in 2D similarity comparisons, we analysed this region in more detail. This 3D analysis showed the unexpected structural similarity between inhibitors of VEGFR and inhibitors of PARP. Among the VEGFR inhibitors that show similarities to PARP inhibitors was Vatalanib, an oral "multi-targeted" small molecule protein kinase inhibitor being studied in phase-III clinical trials in cancer therapy. An in silico docking simulation and an in vitro HT universal colorimetric PARP assay confirmed that the VEGFR inhibitor Vatalanib exhibits off-target activity as a PARP inhibitor, broadening its mode of action. CONCLUSION: In contrast to the 2D-similarity search, the 3D-similarity landscape comparison identifies new functions and side effects of the known VEGFR inhibitor Vatalanib.


Subject(s)
Phthalazines/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Pyridines/chemistry , Binding Sites , Colorimetry , Computational Biology , Drug Discovery , Humans , MCF-7 Cells , Microscopy, Fluorescence , Molecular Docking Simulation , Phthalazines/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Structure, Tertiary , Pyridines/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
16.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 10): 2740-53, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286857

ABSTRACT

The poly(ADP-ribose) polymerase (PARP) family represents a new class of therapeutic targets with diverse potential disease indications. PARP1 and PARP2 inhibitors have been developed for breast and ovarian tumors manifesting double-stranded DNA-repair defects, whereas tankyrase 1 and 2 (TNKS1 and TNKS2, also known as PARP5a and PARP5b, respectively) inhibitors have been developed for tumors with elevated ß-catenin activity. As the clinical relevance of PARP inhibitors continues to be actively explored, there is heightened interest in the design of selective inhibitors based on the detailed structural features of how small-molecule inhibitors bind to each of the PARP family members. Here, the high-resolution crystal structures of the human TNKS2 PARP domain in complex with 16 various PARP inhibitors are reported, including the compounds BSI-201, AZD-2281 and ABT-888, which are currently in Phase 2 or 3 clinical trials. These structures provide insight into the inhibitor-binding modes for the tankyrase PARP domain and valuable information to guide the rational design of future tankyrase-specific inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Tankyrases/antagonists & inhibitors , Tankyrases/chemistry , Benzamides/chemistry , Benzamides/metabolism , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Phthalazines/chemistry , Phthalazines/metabolism , Piperazines/chemistry , Piperazines/metabolism , Protein Conformation , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Quinazolines/chemistry , Quinazolines/metabolism , Tankyrases/genetics , Tankyrases/metabolism
17.
Xenobiotica ; 44(8): 722-33, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24533630

ABSTRACT

1. Aldehyde oxidase (AOX) is a cytosolic molybdoflavoprotein enzyme widely distributed across many tissues. In this study, we report the effect of commonly used organic solvents such as dimethyl sulfoxide (DMSO), acetonitrile (ACN), methanol and ethanol on AOX activity in human, rat and mouse liver S9 fractions using vanillin, phthalazine and methotrexate as probe substrates. 2. Methanol was found to be the most potent solvent in inhibiting vanillic acid and 1-phthalazinone formation in comparison to DMSO, ACN and ethanol across the species tested, except 7-hydroxy methotrexate. 3. Treatment with these solvents at approximate IC50 (% v/v) concentrations showed significant reduction in Clint and Vmax of the probe substrates and also resulted in different effects on Km across the species. 4. Marked differences in the activity and affinity towards AOX were observed with different probe substrates with methotrexate showing least activity and affinity as compared to vanillin and phthalazine. 5. Overall, AOX activity seemed to be more resilient to the presence of organic solvents at higher concentrations in human and rodent species. These results suggest that low concentrations of organic solvents are acceptable for in vitro incubations involving AOX-mediated metabolism.


Subject(s)
Aldehyde Oxidase/metabolism , Benzaldehydes/metabolism , Liver/enzymology , Methotrexate/metabolism , Organic Chemicals/pharmacology , Phthalazines/metabolism , Solvents/pharmacology , Animals , Benzaldehydes/chemistry , Chromatography, Liquid , Female , Humans , Inhibitory Concentration 50 , Kinetics , Liver/drug effects , Male , Methotrexate/chemistry , Mice , Oxidation-Reduction/drug effects , Phthalazines/chemistry , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors
18.
Xenobiotica ; 44(3): 197-204, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24156774

ABSTRACT

1. Aldehyde oxidase (AO) is a liver cytosolic molybdoflavoprotein enzyme whose importance in drug metabolism is gaining in the recent. The objective of this work is to find a potent and selective inhibitor for AO activity using phthalazine oxidation as a marker reaction. 2. Among organic solvents tested, it was identified that methanol was not a suitable choice for AO activity even at concentrations less than 0.2% v/v. Acetonitrile and DMSO did not show any effect till 0.5% v/v but thereafter activites tend to decrease. 3. For selectivity, 23 compounds were selected and evaluated for their effects on AO and nine CYP450 enzymes. Among the tested compounds chlorpromazine, estradiol, hydralazine, quetiapine and raloxifene were selected based on their potency of inhibition towards AO activity. 4. Raloxifene was found to be a non-specific inhibitor of all major tested CYP450 enzymes and was excluded as a selective inhibitor for AO. Quetiapine also showed a degree of inhibition towards the major CYP450 tested. Hydralazine used as a specific inhibitor during the past for AO activity demonstrated a stimulation of AO activity at high and low concentrations respectively and the inhibition noted to be time dependent while inhibiting other enzymes like monoamine oxidase. 5. Estradiol showed no inhibition towards the tested CYP450 enzymes and thus proved to be a selective and specific inhibitor for AO activity with an uncompetitive mode of inhibition.


Subject(s)
Aldehyde Oxidase/antagonists & inhibitors , Inactivation, Metabolic/physiology , Liver/metabolism , Solvents/pharmacology , Aldehyde Oxidase/metabolism , Biomarkers/metabolism , Chromatography, Liquid , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dibenzothiazepines/pharmacology , Estradiol/pharmacology , Humans , Liver/physiology , Microsomes, Liver/metabolism , Oxidation-Reduction , Phthalazines/metabolism , Quetiapine Fumarate , Raloxifene Hydrochloride/pharmacology , Tandem Mass Spectrometry
19.
J Mol Recognit ; 26(7): 297-307, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23657985

ABSTRACT

This paper reports the previously unknown interactions between eight low molecular weight commercially available drugs (130-800 Da) and DNA repair protein photolyase using computational docking simulations and surface plasmon resonance (SPR) experiments. Theoretical dissociation constants, K(d), obtained from molecular docking simulations were compared with the values found from SPR experiments. Among the eight drugs analyzed, computational and experimental values showed similar binding affinities between selected drug and protein pairs. We found no significant differences in binding interactions between pure and commercial forms of the drug lornoxicam and DNA photolyase. Among the eight drugs studied, prednisone, desloratadine, and azelastine exhibited the highest binding affinity (K(d) = 1.65, 2.05, and 8.47 µM, respectively) toward DNA photolyase. Results obtained in this study are promising for use in the prediction of unknown interactions of common drugs with specific proteins such as human clock protein cryptochrome.


Subject(s)
Deoxyribodipyrimidine Photo-Lyase/metabolism , Bacterial Proteins/metabolism , Kinetics , Loratadine/analogs & derivatives , Loratadine/metabolism , Molecular Weight , Phthalazines/metabolism , Piroxicam/analogs & derivatives , Piroxicam/metabolism , Prednisone/metabolism , Protein Binding , Surface Plasmon Resonance , Vibrio cholerae/enzymology
20.
Drug Metab Dispos ; 41(10): 1852-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23918666

ABSTRACT

Aldehyde oxidase (AOX) is a cytosolic enzyme expressed across a wide range of species, including guinea pig and rhesus monkey. These species are believed to be the best preclinical models for studying human AOX-mediated metabolism. We compared AOX activity in rhesus monkeys, guinea pigs, and humans using phthalazine and N-[2-(dimethylamino)ethyl]acridone-4-carboxamide (DACA) as substrates and raloxifene as an inhibitor. Michaelis-Menten kinetics was observed for phthalazine oxidation in rhesus monkey, guinea pig, and human liver cytosol, whereas substrate inhibition was seen with DACA oxidase activity in all three livers. Raloxifene inhibited phthalazine and DACA oxidase activity uncompetitively in guinea pig, whereas mixed-mode inhibition was seen in rhesus monkey. Our analysis of the primary sequence alignment of rhesus monkey, guinea pig, and human aldehyde oxidase isoform 1 (AOX1) along with homology modeling has led to the identification of several amino acid residue differences within the active site and substrate entrance channel of AOX1. We speculate that some of these residues might be responsible for the differences observed in activity. Overall, our data indicate that rhesus monkeys and guinea pigs would overestimate intrinsic clearance in humans and would be unsuitable to use as animal models. Our study also showed that AOX metabolism in species is substrate-dependent and no single animal model can be reliably used to predict every drug response in humans.


Subject(s)
Aldehyde Oxidase/metabolism , Cytosol/enzymology , Cytosol/metabolism , Liver/enzymology , Liver/metabolism , Animals , Guinea Pigs , Humans , Kinetics , Macaca mulatta/metabolism , Male , Oxidation-Reduction , Phthalazines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL