Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
2.
Drug Metab Dispos ; 48(10): 917-924, 2020 10.
Article in English | MEDLINE | ID: mdl-32739890

ABSTRACT

This drug-drug interaction study determined the effect of cyclosporine, an inhibitor of organic anion transporting polypeptide (OATP) 1B3 and P-gp, on the pharmacokinetics (PK) of fevipiprant, an oral, highly selective, competitive antagonist of the prostaglandin D2 receptor 2 and a substrate of the two transporters. The concomitant administration of an intravenous microdose of stable isotope-labeled fevipiprant provided the absolute bioavailability of fevipiprant as well as mechanistic insights into its PK and sensitivity to drug interactions. Liquid chromatography-mass spectrometry/mass spectrometry was used to measure plasma and urine concentrations. Geometric mean ratios [90% confidence interval (CI)] for oral fevipiprant with or without cyclosporine were 3.02 (2.38, 3.82) for C max, 2.50 (2.17, 2.88) for AUClast, and 2.35 (1.99, 2.77) for AUCinf The geometric mean ratios (90% CI) for fevipiprant intravenous microdose with or without cyclosporine were 1.04 (0.86, 1.25) for C max, 2.04 (1.83, 2.28) for AUClast, and 1.95 (1.76, 2.16) for AUCinf The absolute bioavailability for fevipiprant was approximately 0.3 to 0.4 in the absence and 0.5 in the presence of cyclosporine. The intravenous microdose allowed differentiation between systemic and presystemic effects of cyclosporine on fevipiprant, demonstrating a small (approximately 1.2-fold) presystemic effect of cyclosporine and a larger (approximately twofold) effect on systemic elimination of fevipiprant. Uptake by OATP1B3 appears to be the rate-limiting step in the hepatic elimination of fevipiprant, whereas P-gp does not have a relevant effect on oral absorption. SIGNIFICANCE STATEMENT: The drug interaction investigated here with cyclosporine, an inhibitor of several drug transporters, provides a refined quantitative understanding of the role of active transport processes in liver and intestine for the absorption and elimination of fevipiprant as well as the basis to assess the need for dose adjustment in the presence of transporter inhibitors. The applied intravenous microdose approach presents a strategy to maximize learnings from a trial, limit the number and duration of clinical trials, and enhance mechanistic drug-drug interaction understanding.


Subject(s)
Cyclosporine/pharmacokinetics , Indoleacetic Acids/pharmacokinetics , Pyridines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Area Under Curve , Biological Availability , Cross-Over Studies , Cyclosporine/administration & dosage , Drug Interactions , Female , Healthy Volunteers , Humans , Indoleacetic Acids/administration & dosage , Male , Middle Aged , Pyridines/administration & dosage , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Young Adult
3.
Drug Metab Dispos ; 45(7): 817-825, 2017 07.
Article in English | MEDLINE | ID: mdl-28442499

ABSTRACT

Fevipiprant is a novel oral prostaglandin D2 receptor 2 (DP2; also known as CRTh2) antagonist, which is currently in development for the treatment of severe asthma and atopic dermatitis. We investigated the absorption, distribution, metabolism, and excretion properties of fevipiprant in healthy subjects after a single 200-mg oral dose of [14C]-radiolabeled fevipiprant. Fevipiprant and metabolites were analyzed by liquid chromatography coupled to tandem mass spectrometry and radioactivity measurements, and mechanistic in vitro studies were performed to investigate clearance pathways and covalent plasma protein binding. Biotransformation of fevipiprant involved predominantly an inactive acyl glucuronide (AG) metabolite, which was detected in plasma and excreta, representing 28% of excreted drug-related material. The AG metabolite was found to covalently bind to human plasma proteins, likely albumin; however, in vitro covalent binding to liver protein was negligible. Excretion was predominantly as unchanged fevipiprant in urine and feces, indicating clearance by renal and possibly biliary excretion. Fevipiprant was found to be a substrate of transporters organic anion transporter 3 (OAT3; renal uptake), multidrug resistance gene 1 (MDR1; possible biliary excretion), and organic anion-transporting polypeptide 1B3 (OATP1B3; hepatic uptake). Elimination of fevipiprant occurs via glucuronidation by several uridine 5'-diphospho glucuronosyltransferase (UGT) enzymes as well as direct excretion. These parallel elimination pathways result in a low risk of major drug-drug interactions or pharmacogenetic/ethnic variability for this compound.


Subject(s)
Hepatocytes/metabolism , Indoleacetic Acids/pharmacokinetics , Microsomes, Liver/metabolism , Pyridines/pharmacokinetics , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Biotransformation , Feces/chemistry , Healthy Volunteers , Humans , In Vitro Techniques , Indoleacetic Acids/blood , Indoleacetic Acids/urine , Male , Metabolic Clearance Rate , Metabolome , Middle Aged , Protein Binding , Pyridines/blood , Pyridines/urine , Renal Elimination , Tissue Distribution , Young Adult
4.
Pulm Pharmacol Ther ; 39: 54-63, 2016 08.
Article in English | MEDLINE | ID: mdl-27354118

ABSTRACT

BACKGROUND: There is an unmet medical need for allergic asthma patients who are uncontrolled on conventional therapies. The aim of this study was to collect efficacy and safety data for QAW039, an oral chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTh2) receptor antagonist, for the treatment of asthma. METHODS: This was an exploratory phase II, double-blind, randomized, placebo-controlled multi-center study. Patients with mild-to-moderate uncontrolled allergic asthma (N = 170) were either without or weaned off inhaled corticosteroids (ICS) and long-acting ß-agonists (LABA) and randomized (1:1) to QAW039 (500 mg once daily) or to placebo for 28 days. RESULTS: Overall, 157 patients completed the study. There were no significant differences between QAW039 and placebo for trough forced expiratory volume in 1 s (FEV1) or Asthma control questionnaire (ACQ) in the total population. Subgroup analyses demonstrated that patients with a FEV1 <70% of predicted at baseline treated with QAW039 had significant improvement compared with placebo in trough FEV1 (QAW039- Placebo [Δ] = 207 mL; 90% confidence interval [CI]: 96, 319; P = 0.002) and ACQ7 (Δ = -0.41; 90%CI: -0.69, -0.13; P = 0.009). QAW039 reached a mean maximum concentration (Cmax) of 3440 ng/mL on day 28 at a median Tmax of 1 h (range 0.5-4 h). Most adverse events (AEs) were mild/moderate and balanced between both groups, with no serious AEs. CONCLUSIONS: In the general study population, no improvement in lung function was observed with QAW039. However, a subgroup analysis revealed that patients with greater severity of airflow limitation (FEV1 < 70%) had improved lung function and asthma control when treated with QAW039. QAW039 also demonstrated a favorable safety profile. TRIALS REGISTRATION: ClinicalTrials.govNCT01253603.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Indoleacetic Acids/therapeutic use , Pyridines/therapeutic use , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Adult , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacokinetics , Asthma/physiopathology , Double-Blind Method , Female , Forced Expiratory Volume , Humans , Indoleacetic Acids/adverse effects , Indoleacetic Acids/pharmacokinetics , Male , Middle Aged , Pyridines/adverse effects , Pyridines/pharmacokinetics , Treatment Outcome
5.
Clin Pharmacol Drug Dev ; 5(4): 306-13, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27310331

ABSTRACT

We evaluated the pharmacokinetics (PK), safety, and tolerability of a novel oral CRTh2 antagonist, fevipiprant (QAW039), in healthy subjects. Peak concentrations of fevipiprant in plasma were observed 1-3 hours postdosing. Concentrations declined in a multiexponential manner, followed by an apparent terminal phase (t1/2 , ∼20 hours). Steady state was achieved in 4 days with <2-fold accumulation. Elimination was partly by renal excretion (≤30% of the dose) and glucuronidation. Food had minimal impact on the PK of fevipiprant, and it was well tolerated at single and multiple oral doses up to 500 mg/day. No dose-dependent adverse events were observed, and all the events were mild or moderate in severity. Systemic concentrations were sufficiently high to achieve relevant target occupancy, considering in vitro pharmacology data. In summary, the data support further development as a once-daily oral therapy for allergic diseases.


Subject(s)
Food-Drug Interactions , Indoleacetic Acids/administration & dosage , Pyridines/administration & dosage , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Indoleacetic Acids/adverse effects , Indoleacetic Acids/pharmacokinetics , Male , Middle Aged , Pyridines/adverse effects , Pyridines/pharmacokinetics
6.
PLoS One ; 11(5): e0156453, 2016.
Article in English | MEDLINE | ID: mdl-27228127

ABSTRACT

Auxin has been shown to modulate the fruit ripening process. However, the molecular mechanisms underlying auxin regulation of fruit ripening are still not clear. Illumina RNA sequencing was performed on mature green cherry tomato fruit 1 and 7 days after auxin treatment, with untreated fruit as a control. The results showed that exogenous auxin maintained system 1 ethylene synthesis and delayed the onset of system 2 ethylene synthesis and the ripening process. At the molecular level, genes associated with stress resistance were significantly up-regulated, but genes related to carotenoid metabolism, cell degradation and energy metabolism were strongly down-regulated by exogenous auxin. Furthermore, genes encoding DNA demethylases were inhibited by auxin, whereas genes encoding cytosine-5 DNA methyltransferases were induced, which contributed to the maintenance of high methylation levels in the nucleus and thus inhibited the ripening process. Additionally, exogenous auxin altered the expression patterns of ethylene and auxin signaling-related genes that were induced or repressed in the normal ripening process, suggesting significant crosstalk between these two hormones during tomato ripening. The present work is the first comprehensive transcriptome analysis of auxin-treated tomato fruit during ripening. Our results provide comprehensive insights into the effects of auxin on the tomato ripening process and the mechanism of crosstalk between auxin and ethylene.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Plant/drug effects , Indoleacetic Acids/pharmacokinetics , Plant Proteins/biosynthesis , Solanum lycopersicum/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Ethylenes/metabolism , High-Throughput Nucleotide Sequencing , Solanum lycopersicum/genetics , Plant Proteins/genetics
7.
Biotechnol Lett ; 37(1): 81-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25257586

ABSTRACT

Among various enzyme-based therapies, enzyme-prodrug therapy (EPT) promises minimized side effects in that it activates non-toxic prodrugs locally where the enzymes are placed. The success of such an approach requires high enzyme stability against both structural denaturation and potential immunogenicity. This work examines the efficiency of nanoparticles for enzyme protection in EPT applications. Specifically, horseradish peroxidase (HRP)-encapsulated chitosan nanoparticles (HRP-CSNP) were constructed and examined with respect to stability enhancement. HRP-CSNP retained enzyme activity and had improved stability at 37 °C in the presence of a denaturant, urea. The nanoparticles effectively bound to the surface of human breast cancer cell Bcap37 and led to over 80 % cell death when applied with a prodrug indole-3-acetic acid.


Subject(s)
Antineoplastic Agents/chemistry , Chitosan/chemistry , Horseradish Peroxidase/chemistry , Nanoparticles/chemistry , Prodrugs/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Stability , Humans , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/pharmacology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prohibitins
8.
J Med Chem ; 55(11): 5088-109, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22651823

ABSTRACT

New classes of CRTH2 antagonists, the pyridazine linker containing indole acetic acids, are described. The initial hit 1 had good potency but poor permeability, metabolic stability, and PK. Initial optimization led to compounds of type 2 with low oxidative metabolism but poor oral bioavailability. Poor permeability was identified as a liability for these compounds. Addition of a linker between the indole and diazine moieties afforded a series with good potency, low rates of metabolism, moderate permeability, and good oral bioavailability in rodents. 32 was identified as the development track candidate. It was potent in cell based, binding, and whole blood assays and exhibited good PK profile. It was efficacious in mouse models of contact hypersensitivity (1 mg/kg b.i.d.) and house dust (20 mg/kg q.d.) when dosed orally. In sheep asthma, administration at 1 mg/kg iv completely blocked the LAR and AHR and attenuated the EAR phase.


Subject(s)
Hypersensitivity/drug therapy , Indoleacetic Acids/chemical synthesis , Pyridazines/chemical synthesis , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Bronchoconstriction/drug effects , Caco-2 Cells , Cell Shape/drug effects , Chemotaxis, Leukocyte/drug effects , Dermatitis, Contact/drug therapy , Dermatitis, Contact/immunology , Eosinophils/cytology , Eosinophils/drug effects , Female , High-Throughput Screening Assays , Humans , Hypersensitivity/immunology , Immunoassay , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Permeability , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyroglyphidae/immunology , Rats , Sheep , Structure-Activity Relationship
9.
Cancer Gene Ther ; 19(2): 77-83, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21959967

ABSTRACT

Clinical efficacy of current therapies for hepatocellular carcinoma (HCC) treatment is limited. Indole-3-acetic acid (IAA) is non-toxic for mammalian cells. Oxidative decarboxylation of IAA by horseradish peroxidase (HRP) leads to toxic effects of IAA. The purpose of this study was to investigate the effects of a novel gene-targeted enzyme prodrug therapy with IAA on hepatoma growth in vitro and in vivo mouse hepatoma models. We generated a plasmid using adenovirus to express HRP isoenzyme C (HRPC) with the HCC marker, alpha-fetoprotein (AFP), as the promoter (pAdv-AFP-HRPC). Hepatocellular cells were infected with pAdv-AFP-HRPC and treated with IAA. Cell death was detected using MTT assay. Hepatoma xenografts were developed in mice by injection of mouse hepatoma cells. The size and weight of tumors and organs were evaluated. Cell death in tumors was assessed using hematoxylin and eosin-stained tissue sections. HRPC expression in tissues was detected using Reverse Transcriptase-Polymerase Chain Reaction. IAA stimulated death of hepatocellular cells infected with pAdv-AFP-HRPC, in a dose- and time-dependent manner, but not in control cells. Growth of hepatoma xenografts, including the size and weight, was inhibited in mice treated with pAdv-AFP-HRPC and IAA, compared with that in control group. pAdv-AFP-HRPC/IAA treatment induced cell death in hepatoma xenografts in mice. HRPC gene expressed only in hepatoma, but not in other normal organs of mice. pAdv-AFP-HRPC/IAA treatment did not cause any side effects on normal organs. These findings suggest that pAdv-AFP-HRPC/IAA enzyme/prodrug system may serve as a strategy for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , Horseradish Peroxidase/genetics , Indoleacetic Acids/pharmacology , Liver Neoplasms/therapy , alpha-Fetoproteins/genetics , Adenoviridae/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Disease Models, Animal , Genetic Vectors , Hep G2 Cells , Horseradish Peroxidase/biosynthesis , Horseradish Peroxidase/metabolism , Humans , Indoleacetic Acids/pharmacokinetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Plasmids/genetics , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Xenograft Model Antitumor Assays , alpha-Fetoproteins/biosynthesis
10.
J Pharmacol Exp Ther ; 340(2): 473-82, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22106101

ABSTRACT

D prostanoid receptor 2 (DP2) [also known as chemoattractant receptor-homologous molecule expressed on T helper 2 (Th2) cells (CRTH2)] is selectively expressed by Th2 lymphocytes, eosinophils, and basophils and mediates recruitment and activation of these cell types in response to prostaglandin D2 (PGD2). (5-Fluoro-2-methyl-3-quinolin-2-ylmethylindo-1-yl)-acetic acid (OC000459) is an indole-acetic acid derivative that potently displaces [³H]PGD2 from human recombinant DP2 (K(i) = 0.013 µM), rat recombinant DP2 (K(i) = 0.003 µM), and human native DP2 (Th2 cell membranes; K(i) = 0.004 µM) but does not interfere with the ligand binding properties or functional activities of other prostanoid receptors (prostaglandin E1₋4 receptors, D prostanoid receptor 1, thromboxane receptor, prostacyclin receptor, and prostaglandin F receptor). OC000459 inhibited chemotaxis (IC50 = 0.028 µM) of human Th2 lymphocytes and cytokine production (IC50 = 0.019 µM) by human Th2 lymphocytes. OC000459 competitively antagonized eosinophil shape change responses induced by PGD2 in both isolated human leukocytes (pK(B) = 7.9) and human whole blood (pK(B) = 7.5) but did not inhibit responses to eotaxin, 5-oxo-eicosatetraenoic acid, or complement component C5a. OC000459 also inhibited the activation of Th2 cells and eosinophils in response to supernatants from IgE/anti-IgE-activated human mast cells. OC000459 had no significant inhibitory activity on a battery of 69 receptors and 19 enzymes including cyclooxygenase 1 (COX1) and COX2. OC000459 was found to be orally bioavailable in rats and effective in inhibiting blood eosinophilia induced by 13,14-dihydro-15-keto-PGD2 (DK-PGD2) in this species (ED50 = 0.04 mg/kg p.o.) and airway eosinophilia in response to an aerosol of DK-PGD2 in guinea pigs (ED50 = 0.01 mg/kg p.o.). These data indicate that OC000459 is a potent, selective, and orally active DP2 antagonist that retains activity in human whole blood and inhibits mast cell-dependent activation of both human Th2 lymphocytes and eosinophils.


Subject(s)
Eosinophils/drug effects , Indoleacetic Acids/pharmacology , Lymphocyte Activation/drug effects , Mast Cells/immunology , Prostaglandin Antagonists/pharmacology , Quinolines/pharmacology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Th2 Cells/drug effects , Animals , Apoptosis/drug effects , Arachidonic Acids/pharmacology , Binding, Competitive , CHO Cells , Calcium Signaling/drug effects , Cell Membrane/metabolism , Cell Shape/drug effects , Cell Shape/immunology , Chemokine CCL11/pharmacology , Chemotaxis/drug effects , Chemotaxis/immunology , Complement C5a/pharmacology , Cricetinae , Culture Media, Conditioned/pharmacology , Eosinophilia/chemically induced , Eosinophilia/prevention & control , Eosinophils/cytology , Eosinophils/immunology , Guinea Pigs , Humans , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/therapeutic use , Interleukin-13/metabolism , Interleukin-5/pharmacology , Leukotriene B4/pharmacology , Lymphocyte Activation/immunology , Mast Cells/metabolism , Prostaglandin Antagonists/pharmacokinetics , Prostaglandin Antagonists/therapeutic use , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/prevention & control , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Recombinant Proteins/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Transfection
11.
Cancer Gene Ther ; 17(6): 420-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20075982

ABSTRACT

Gene-directed enzyme prodrug therapy is a form of targeted cancer therapy, in which an enzyme is used to convert a non-toxic prodrug to a cytotoxin within the tumor. Horseradish peroxidase (HRP) is able to convert the indole prodrugs indole-3-acetic acid (IAA) and the halogenated derivative 5-bromo-IAA (5Br-IAA) to toxic agents able to induce cell kill in vitro. This study characterized HRP-directed gene therapy in vivo. Human nasopharyngeal squamous cell carcinoma cells, FaDu, stably expressing HRP were grown as xenografts in SCID mice. Pharmacokinetic analysis of IAA and 5Br-IAA showed satisfactory drug profiles, and millimolar concentrations could be achieved in tumor tissue at non-toxic doses. HRP-expressing tumors showed a modest growth delay when treated with IAA compared with drug-vehicle controls. Treatment response could not be improved using different drug scheduling or drug vehicle, nor by combining HRP-directed gene therapy with fractionated radiotherapy.


Subject(s)
Genetic Therapy/methods , Horseradish Peroxidase/metabolism , Indoleacetic Acids/metabolism , Neoplasms/therapy , Animals , Area Under Curve , Bromine/chemistry , Cell Line, Tumor , Combined Modality Therapy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Horseradish Peroxidase/genetics , Humans , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacokinetics , Mice , Mice, SCID , Neoplasms/genetics , Prodrugs/administration & dosage , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Radiotherapy/methods , Transgenes/genetics , Xenograft Model Antitumor Assays
12.
Int J Clin Pharmacol Ther ; 46(11): 545-55, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19000552

ABSTRACT

PURPOSE: Pactimibe is a novel ACAT inhibitor. The pharmacokinetics of pactimibe and its pharmacologically inactive plasma metabolite, R-125528, of which the main clearance pathway is CYP2D6, was affected by coadministration of quinidine. The aim of this study was to investigate the influence of CYP2D6 polymorphism on pharmacokinetics of pactimibe and R-125528. In addition, exposure was examined after multiple doses of pactimibe sulfate in CYP2D6 poor metabolizer (PMs). METHODS: 24 healthy male Caucasian volunteers, genotyped as extensive, intermediate, and poor metabolizers, were received single dose of 25 mg pactimibe. In a multiple-dose study, six CYP2D6 PMs received 100 mg pactimibe for 21 days and exposure of pactimibe and R-125528 was examined. RESULTS: In contrast to the mild 1.7-fold increase in AUC0-inf of pactimibe, a marked 3.1-fold increase in AUC0-tz of R-125528 was observed in CYP2D6 PMs. After multiple doses of 100 mg pactimibe to CYP2D6 PMs, the accumulation ratio of R-125528 reached 8.8-fold, however, the exposure of R-125528 in CYP2D6 PMs was covered by the exposure in additional metabolite safety testing. CONCLUSIONS: Although CYP2D6 polymorphism greatly affected the pharmacokinetics of R-125528 rather than pactimibe, the exposure in CYP2D6 PMs after a multiple dose of 100 mg pactimibe sulfate was covered by additional non-clinical metabolite safety testing. The finding is clinically informative with respect to the safety testing of drug metabolite present at disproportionately high levels in a special population with specific genetic back ground.


Subject(s)
Alkanes/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/pharmacology , Indoleacetic Acids/pharmacokinetics , Indoles/pharmacokinetics , Adolescent , Adult , Alkanes/blood , Area Under Curve , Cytochrome P-450 CYP2D6/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Genotype , Half-Life , Humans , Indoleacetic Acids/metabolism , Indoleacetic Acids/pharmacology , Indoles/blood , Linear Models , Male , Polymorphism, Genetic , Sterol O-Acyltransferase/antagonists & inhibitors , Young Adult
13.
J Thromb Haemost ; 6(9): 1558-64, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18624980

ABSTRACT

OBJECTIVE: To assess the antithrombotic and profibrinolytic effects of tiplaxtinin (PAI-039), an orally bioavailable antagonist of PAI-1, in rat models of thrombosis. METHODS AND RESULTS: Carotid artery and vena cava vascular injury was produced by application of FeCl3 and blood flow was monitored using ultrasonic technology. To assess efficacy in a thrombosis prevention paradigm, PAI-039 was administered orally 90 min before injury (1-30 mg kg(-1)). To assess efficacy in a thrombosis treatment paradigm, vascular injury and stable thrombus formation were followed 4 h later by recovery and PAI-039 administration. PAI-039 prevented carotid artery occlusion in 20, 68 and 60% of animals pretreated with 0.3, 1.0 and 3.0 mg kg(-1), respectively. Time to occlusive thrombosis was increased from 18.2 +/- 4.6 min in controls to 32.5 +/- 8.7 (P = ns), 46.1 +/- 7.0 (P < 0.05), and 41.6 +/- 11.3 min (P < 0.05) in the respective PAI-039 treatment groups. In the vena cava protocol, PAI-039 pretreatment significantly reduced thrombus weight at PAI-039 doses of 3, 10 and 30 mg kg(-1). When PAI-039 was dosed in a treatment paradigm 4 h after stable arterial and venous thrombosis, a significant reduction in thrombus weight was observed 24 h later at PAI-039 doses of 3, 10 and 30 mg kg(-1). PAI-039 (10, 30 and 100 mg kg(-1)) had no effect on platelet aggregation in response to ADP or collagen and was not associated with increased bleeding or prolonged prothrombin time. In animals bearing no vascular injury, PAI-039 had no effect on circulating, low-levels of PAI-1 activity. In contrast, circulating PAI-1 activity increased 5-fold following the induction of vascular injury, which was completely neutralized by PAI-039. CONCLUSIONS: PAI-039 exerts antithrombotic efficacy in rat models of arterial and venous vascular injury without effecting platelet aggregation.


Subject(s)
Indoleacetic Acids/pharmacology , Plasminogen Activator Inhibitor 1 , Thrombosis/prevention & control , Administration, Oral , Animals , Biological Availability , Disease Models, Animal , Indoleacetic Acids/administration & dosage , Indoleacetic Acids/pharmacokinetics , Male , Platelet Aggregation/drug effects , Prothrombin Time , Rats , Rats, Sprague-Dawley
14.
Drug Metab Dispos ; 36(8): 1505-11, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18448569

ABSTRACT

Pactimibe sulfate is a novel acyl coenzyme A:cholesterol acyltransferase inhibitor developed for the treatment of hypercholesterolemia and atherosclerotic diseases. Pactimibe has two equally dominant clearance pathways forming R-125528 by CYP3A4 and M-1 by CYP2D6 in vitro. R-125528 is a plasma metabolite and is cleared solely by CYP2D6 despite its acidity. To evaluate contributions of the cytochrome P450 enzymes on the pharmacokinetics of pactimibe and R-125528 in humans, drug-drug interaction studies using ketoconazole and quinidine were conducted. Eighteen healthy male subjects were given a single dose of pactimibe sulfate without and with 400 mg of ketoconazole (q.d.). With the concomitant treatment, the area under the plasma concentration-time curve (AUC(0-inf)) of pactimibe modestly increased 1.7-fold and AUC(0-tz) of R-125528 decreased by 55%. In addition, 17 healthy male subjects were given a single dose of pactimibe sulfate without and with 600 mg of quinidine (b.i.d.). With the concomitant treatment, the AUC(0-inf) for pactimibe modestly increased 1.7-fold. On the other hand, the AUC(0-tz) of R-125528 was markedly elevated 5.0-fold, although the AUC(0-inf) could not be adequately defined because the terminal elimination phase of R-125528 was not obtained in the study period up to 72 h. As the f(m CYP3A4) and f(m CYP2D6) values of pactimibe estimated from in vitro studies were 0.40 and 0.33, respectively, AUC increase ratios of pactimibe were estimated to be 1.7 with ketoconazole and 1.5 with quinidine. These values were well in accordance with the values observed in this study. Moreover, the f(m CYP2D6) of R-125528 estimated to be almost 1 would well explain the accumulation of R-125528 observed with the quinidine treatment.


Subject(s)
Alkanes/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Indoleacetic Acids/pharmacokinetics , Indoles/pharmacokinetics , Ketoconazole/pharmacology , Quinidine/pharmacology , Adult , Alkanes/blood , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Interactions , Enzyme Inhibitors/blood , Humans , Indoleacetic Acids/blood , Indoles/blood , Male , Placebos , Sensitivity and Specificity
15.
Drug Metab Dispos ; 36(3): 529-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18056254

ABSTRACT

Pactimibe sulfate is a novel acyl coenzyme A:cholesterol acyltransferase inhibitor. We conducted metabolic studies of pactimibe and its plasma metabolite, R-125528. Pactimibe had multiple metabolic pathways including indolin oxidation to form R-125528, omega-1 oxidation, N-dealkylation, and glucuronidation. Among them, the indolin oxidation and the omega-1 oxidation were dominant and were mainly catalyzed by CYP3A4 and CYP2D6, respectively. The intrinsic clearance (CL(int)) values for these pathways in human hepatic microsomes were 0.63 and 0.76 microl/min/mg-protein, respectively. On the other hand, the metabolic reaction for R-125528 was restricted. It was demonstrated that omega-1 oxidation was the only pathway that could eliminate R-125528 from the systemic circulation. To our surprise, only CYP2D6-expressing microsomes could catalyze the reaction, and omega-1 oxidation was strongly correlated with the CYP2D6 marker reaction, dextromethorphan O-demethylation (r(2) = 0.90), in human hepatic microsomes. Although R-125528 is an atypical substrate for CYP2D6 because of its acidity, the K(m) value was 1.8 microM for the reaction in human hepatic microsomes and the CL(int) value was as high as 75.0 microl/min/mg-protein. These results suggested that the systemic clearance of R-125528 was highly dependent on CYP2D6 activity and that several studies with CYP2D6 including drug-drug interaction and polymorphism sensitivity should be performed during development from the viewpoint of metabolite safety assessment. The finding that R-125528, an acidic compound devoid of basic nitrogen, was a good substrate for CYP2D6 raised a question about previously reported CYP2D6 models based on a critical electrostatic interaction with Asp(301) and/or Glu(216).


Subject(s)
Cytochrome P-450 CYP2D6/metabolism , Indoleacetic Acids/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Indoleacetic Acids/pharmacokinetics , Kinetics , Lymphocytes/enzymology , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism
16.
Eur J Pharmacol ; 578(1): 97-9, 2008 Jan 06.
Article in English | MEDLINE | ID: mdl-17716650

ABSTRACT

The purpose of this study is to clarify involvement ratios between central and peripheral cyclooxygenase (COX)-2 in rat inflammatory pain models, by evaluating celecoxib and [6-chloro-2-(4-chlorobenzoyl)-1H-indol-3-yl]acetic acid (CIAA) on carrageenan-induced mechanical and thermal hyperalgesia. Celecoxib and CIAA exhibited ID(30) values with 1.5 and 7.7 mg/kg on mechanical hyperalgesia, respectively, and ID(25) values with 0.54 and 36 mg/kg on thermal hyperalgesia, respectively. By solving quadratic functional analysis with prostaglandin E(2) (PGE(2)) inhibitory activities, it was calculated that involvement ratios between central and peripheral COX-2 involvement were 0.47 and 0.53 on mechanical hyperalgesia, and 0.97 and 0.03 on thermal hyperalgesia, respectively. These data suggest that central and peripheral COX-2 are equally involved in mechanical hyperalgesia, while central COX-2 is predominantly involved in thermal hyperalgesia.


Subject(s)
Chlorobenzoates/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Indoleacetic Acids/pharmacology , Pain/physiopathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Carrageenan , Celecoxib , Chlorobenzoates/administration & dosage , Chlorobenzoates/pharmacokinetics , Cyclooxygenase 2/drug effects , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Dinoprostone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Hot Temperature , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Indoleacetic Acids/administration & dosage , Indoleacetic Acids/pharmacokinetics , Inflammation/drug therapy , Inflammation/physiopathology , Male , Models, Biological , Pain/drug therapy , Pain Measurement , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Tissue Distribution
17.
J Med Chem ; 48(20): 6174-7, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16190744

ABSTRACT

Prostaglandin D2 (PGD2) acting at the CRTH2 receptor (chemoattractant receptor-homologous molecule expressed on Th2 cells) has been linked with a variety of allergic and other inflammatory diseases. We describe a family of indole-1-sulfonyl-3-acetic acids that are potent and selective CRTH2 antagonists that possess good oral bioavailability. The compounds may serve as novel starting points for the development of treatments of inflammatory disease such as asthma, allergic rhinitis, and atopic dermatitis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Indoleacetic Acids/chemical synthesis , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Availability , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Humans , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/pharmacology , Prostaglandin D2/pharmacology , Rats , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/pharmacokinetics , Sulfones/pharmacology
18.
Eur J Med Chem ; 40(5): 505-19, 2005 May.
Article in English | MEDLINE | ID: mdl-15893024

ABSTRACT

A series of N-(p-alkoxy)benzoyl-5-methoxy-2-methylindole-3-acetic acids and N-(p-butoxy)benzoyl-2-methylindole-4-acetic acid were discovered as new chemical leads for a prostaglandin D2 (PGD2) receptor antagonist. Most of them exhibited PGD2 receptor binding and blocked cyclic adenosine 3',5'-monophosphate (cAMP) formation in vitro. In particular, 2-methylindole-4-acetic acid analog 1 showed markedly increased receptor affinity and cAMP antagonist activity. Chemistry and structure activity relationship (SAR) data are also presented.


Subject(s)
Indoleacetic Acids/chemical synthesis , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Area Under Curve , Binding, Competitive/physiology , CHO Cells , Cricetinae , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Half-Life , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacokinetics , Indomethacin/analogs & derivatives , Indomethacin/chemistry , Indomethacin/pharmacokinetics , Magnetic Resonance Spectroscopy , Male , Mice , Rats , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship
19.
J Med Chem ; 48(9): 3141-52, 2005 May 05.
Article in English | MEDLINE | ID: mdl-15857120

ABSTRACT

Recent efforts to identify treatments for chronic diabetic complications have resulted in the discovery of a novel series of highly potent and selective 3-[(benzothiazol-2-yl)methyl]indole-N-alkanoic acid aldose reductase inhibitors. The lead candidate, 3-[(4,5,7-trifluorobenzothiazol-2-yl)methyl]indole-N-acetic acid (lidorestat, 9) inhibits aldose reductase with an IC(50) of 5 nM, while being 5400 times less active against aldehyde reductase, a related enzyme involved in the detoxification of reactive aldehydes. It lowers nerve and lens sorbitol levels with ED(50)'s of 1.9 and 4.5 mg/kg/d po, respectively, in the 5-day STZ-induced diabetic rat model. In a 3-month diabetic intervention model (1 month of diabetes followed by 2 months of drug treatment at 5 mg/kg/d po), it normalizes polyols and reduces the motor nerve conduction velocity deficit by 59% relative to diabetic controls. It has a favorable pharmacokinetic profile (F, 82%; t(1/2), 5.6 h; Vd, 0.694 L/kg) with good drug penetration in target tissues (C(max) in sciatic nerve and eye are 2.36 and 1.45 mug equiv/g, respectively, when dosed with [(14)C]lidorestat at 10 mg/kg po).


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Complications/drug therapy , Indoleacetic Acids/chemical synthesis , Thiazoles/chemical synthesis , Aldehyde Reductase/chemistry , Aldehyde Reductase/genetics , Animals , Cataract/drug therapy , Chronic Disease , Crystallography, X-Ray , Diabetes Complications/metabolism , Diabetes Complications/physiopathology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Humans , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/pharmacology , Lens, Crystalline/metabolism , Male , Models, Molecular , Rats , Rats, Sprague-Dawley , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Structure-Activity Relationship , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Tissue Distribution
20.
Pharm Res ; 22(4): 619-27, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15846470

ABSTRACT

PURPOSE: Evidence suggests that uremic toxins such as hippurate (HA), indoleacetate (IA), indoxyl sulfate (IS), and 3-carboxy-4-methyl-5-propyl-2-furanpropionate (CMPF) promote the progression of renal failure by damaging tubular cells via rat organic anion transporter 1 (rOat1) and rOat3 on the basolateral membrane of the proximal tubules. The purpose of the current study is to evaluate the in vivo transport mechanism responsible for their renal uptake. METHODS: We investigated the uremic toxins transport mechanism using the abdominal aorta injection technique [i.e., kidney uptake index (KUI) method], assuming minimal mixing of the bolus with serum protein from circulating serum. RESULTS: Maximum mixing was estimated to be 5.8% of rat serum by measuring estrone sulfate extraction after addition of 0-90% rat serum to the arterial injection solution. Saturable renal uptake of p-aminohippurate (PAH, K(m) = 408 microM) and benzylpenicillin (PCG, K(m) = 346 microM) was observed, respectively. The uptake of PAH and PCG was inhibited in a dose-dependent manner by unlabeled PCG (IC(50) = 47.3 mM) and PAH (IC(50) = 512 microM), respectively, suggesting that different transporters are responsible for their uptake. A number of uremic toxins inhibited the renal uptake of PAH and PCG. Excess PAH, which could inhibit rOat1 and rOat3, completely inhibited the saturable uptake of IA, IS, and CMPF by the kidney, and by 85% for HA uptake. PCG inhibited the total saturable uptake of HA, IA, IS, and CMPF by 10%, 10%, 45%, and 65%, respectively, at the concentration selective for rOat3. CONCLUSIONS: rOat1 could be the primary mediator of the renal uptake of HA and IA, accounting for approximately 75% and 90% of their transport, respectively. rOat1 and rOat3 contributed equally to the renal uptake of IS. rOat3 could account for about 65% of the uptake of CMPF under in vivo physiologic conditions. These results suggest that rOat1 and rOat3 play an important role in the renal uptake of uremic toxins and the induction of their nephrotoxicity.


Subject(s)
Kidney/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Cation Transport Proteins/metabolism , Penicillin G/pharmacology , p-Aminohippuric Acid/pharmacology , Animals , Biological Transport/drug effects , Carbon Radioisotopes , Estrone/analogs & derivatives , Estrone/metabolism , Furans/pharmacokinetics , Furans/pharmacology , Furans/toxicity , Hippurates/pharmacokinetics , Hippurates/pharmacology , Hippurates/toxicity , Indican/pharmacokinetics , Indican/pharmacology , Indican/toxicity , Indoleacetic Acids/pharmacokinetics , Indoleacetic Acids/pharmacology , Indoleacetic Acids/toxicity , Kidney/drug effects , Male , Metabolic Clearance Rate , Organic Anion Transport Protein 1/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Penicillin G/pharmacokinetics , Propionates/pharmacokinetics , Propionates/pharmacology , Propionates/toxicity , Rats , Rats, Wistar , Serum , Tritium , p-Aminohippuric Acid/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...