Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Pharmaceutics ; 10(4)2018 Oct 03.
Article in English | MEDLINE | ID: mdl-30282950

ABSTRACT

The aim of the present study was to assess the regional absorption of fimasartan by an improved in situ absorption method in comparison with the conventional in situ single-pass perfusion method in rats. After each gastrointestinal segment of interest was identified, fimasartan was injected into the starting point of each segment and the unabsorbed fimasartan was discharged from the end point of the segment. Blood samples were collected from the jugular vein to evaluate the systemic absorption of the drug. The relative fraction absorbed (Fabs,relative) values in the specific gastrointestinal region calculated based on the area under the curve (AUC) values obtained after the injection of fimasartan into the gastrointestinal segment were 8.2% ± 3.2%, 23.0% ± 12.1%, 49.7% ± 11.5%, and 19.1% ± 11.9% for the stomach, duodenum, small intestine, and large intestine, respectively, which were comparable with those determined by the conventional in situ single-pass perfusion. By applying the fraction of the dose available at each gastrointestinal segment following the oral administration, the actual fraction absorbed (F'abs) values at each gastrointestinal segment were estimated at 10.9% for the stomach, 27.1% for the duodenum, 40.7% for the small intestine, and 5.4% for the large intestine, which added up to the gastrointestinal bioavailability (FX·FG) of 84.1%. The present method holds great promise to assess the regional absorption of a drug and aid to design new drug formulations.

2.
Food Chem Toxicol ; 115: 375-384, 2018 May.
Article in English | MEDLINE | ID: mdl-29596975

ABSTRACT

Fimasartan (FMS), an angiotensin II receptor antagonist, is metabolized to FMS S-oxide, FMS N-glucuronide, oxidative desulfurized FMS (BR-A-557), and hydroxy-n-butyl FMSs. The purpose of this study was to characterize enzymes involved in NADPH-dependent FMS metabolism using recombinant enzymes such as cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO), as well as selective chemical inhibitors. The results showed that CYP, but not FMO, plays a major role in FMS metabolism. CYP2C9, CYP3A4, and CYP3A5 were involved in the formation of FMS S-oxide, which was further metabolized to BR-A-557 by CYP3A4/5. CYP2C9 played an exclusive role in n-butyl hydroxylation. The specificity constant (kcat/Km) values for S-oxidation by CYP2C9, CYP3A4, and CYP3A5 were 0.21, 0.34, and 0.19 µM-1∙min-1, respectively. The kcat/Km values of hydroxylation at the 1-, 2-/3-, and 4-n-butyl group in CYP2C9 were 0.0076, 0.041, and 0.035 µM-1∙min-1, respectively. The kcat and Km values provide information for the prediction of FMS metabolism in vivo. In addition, simultaneous determination of the FMS metabolites may be used to evaluate CYP2C9 and CYP3A4/5 activity.


Subject(s)
Angiotensin Receptor Antagonists/metabolism , Biphenyl Compounds/metabolism , Cytochrome P-450 Enzyme System/metabolism , Pyrimidines/metabolism , Tetrazoles/metabolism , Chromatography, Liquid , Humans , Hydroxylation , In Vitro Techniques , Kinetics , Oxidation-Reduction , Oxygenases/metabolism , Recombinant Proteins/metabolism , Tandem Mass Spectrometry , Thermodynamics
3.
Biol Pharm Bull ; 40(7): 992-1001, 2017.
Article in English | MEDLINE | ID: mdl-28674263

ABSTRACT

The pharmacological profile of fimasartan, [2-n-butyl-5-dimethylamino-thiocarbonyl-methyl-6-methyl-3-{[2-(1H-tetrazole-5-yl)biphenyl-4-yl]methyl}-pyrimidin-4(3H)-one, a new non-peptide angiotensin type 1 (AT1)-selective angiotensin receptor antagonist, has been investigated in a variety of in vitro and in vivo experimental models. In the present study, fimasartan showed slow dissociation and irreversible binding to AT1 subtype receptors in membrane fractions of HEK-293 cells with a Kd of 0.03 nM and a T1/2 of 63.7 min. The inhibitory effect of fimasartan on angiotensin II (Ang II)-induced contraction persisted longer after washout than that of losartan or candesartan. In conscious rats, a single dose of fimasartan (0.3, 1, or 3 mg/kg; per os (p.o.)) dose-dependently antagonized Ang II-induced pressor responses. Both orally administrated fimasartan and losartan dose-dependently decreased mean arterial pressure in furosemide-treated rats and dogs, and fimasartan administered orally at 1, 3, or 10 mg/kg reduced blood pressure in conscious spontaneously hypertensive rats. Taken together, these findings indicate that fimasartan has potent and sustained binding affinity at the AT1 receptor subtype, and reveal the molecular basis responsible for the marked lowering of blood pressure in various conscious rats and dogs models after its oral administration.


Subject(s)
Angiotensin Receptor Antagonists/pharmacology , Biphenyl Compounds/pharmacology , Pyrimidines/pharmacology , Tetrazoles/pharmacology , Administration, Oral , Angiotensin Receptor Antagonists/administration & dosage , Animals , Biphenyl Compounds/administration & dosage , Blood Pressure/drug effects , Dogs , Furosemide/pharmacology , HEK293 Cells , Humans , Male , Pyrimidines/administration & dosage , Rabbits , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Tetrazoles/administration & dosage
4.
Eur J Pharm Sci ; 107: 32-44, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-28599987

ABSTRACT

Fimasartan is a novel angiotensin II receptor blocker. Our aims were to characterize the time-course of the antihypertensive activity of fimasartan via a new population pharmacokinetic/pharmacodynamic model and to define its optimal dose range. We simultaneously modelled all fimasartan plasma concentrations and 24-h ambulatory blood pressure monitoring (ABPM) data from 39 patients with essential hypertension and 56 healthy volunteers. Patients received placebo, 20, 60, or 180mg fimasartan every 24h for 28days and healthy volunteers received placebo or 20 to 480mg as a single oral dose or as seven doses every 24h. External validation was performed using data on 560 patients from four phase II or III studies. One turnover model each was used to describe diastolic and systolic blood pressure. The input rates into these compartments followed a circadian rhythm and were inhibited by fimasartan. The average predicted (observed) diastolic blood pressure over 24-h in patients decreased by 10.1±7.5 (12.6±9.2; mean±SD)mmHg for 20mg, 14.2±7.0 (15.1±9.3) mmHg for 60mg, and 15.9±6.8 (11.5±9.9)mmHg for 180mg daily relative to placebo. The model explained the saturation of antihypertensive activity by counter-regulation at high fimasartan concentrations. Drug effect was maximal at approximately 23ng/mL fimasartan for diastolic and 12ng/mL for systolic blood pressure. The proposed mechanism-based population model characterized the circadian rhythm of ABPM data and the antihypertensive effect of fimasartan. After internal and external model validation, 30 to 60mg oral fimasartan given once daily was predicted as optimal dose range.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Biphenyl Compounds/pharmacology , Biphenyl Compounds/pharmacokinetics , Blood Pressure/drug effects , Models, Biological , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Tetrazoles/pharmacology , Tetrazoles/pharmacokinetics , Antihypertensive Agents/blood , Antihypertensive Agents/therapeutic use , Biphenyl Compounds/blood , Biphenyl Compounds/therapeutic use , Circadian Rhythm , Essential Hypertension/drug therapy , Essential Hypertension/metabolism , Essential Hypertension/physiopathology , Humans , Pyrimidines/blood , Pyrimidines/therapeutic use , Tetrazoles/blood , Tetrazoles/therapeutic use
5.
BMC Pharmacol Toxicol ; 17(1): 35, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27459959

ABSTRACT

BACKGROUND: Fimasartan (FMS) is a potent angiotensin receptor blocker for the treatment of mild to moderate hypertension. This study aimed to evaluate the transfer of FMS to fetus and breast milk in rats. METHODS: In order to study the transfer to the fetus and nursing pup, pregnant and nursing maternal rats were administered with FMS by a constant intravenous infusion to reach target plasma concentrations of 200 ng/mL and 100 ng/mL. The concentrations of FMS in plasma, placenta, amniotic fluid, fetus, and milk were determined by a validated LC-MS/MS assay. RESULTS: Upon constant intravenous infusion, the plasma FMS concentration reached the target steady state concentrations (Css = 200 ng/mL and 100 ng/mL) in 24 h. The tissue-to-plasma partition coefficients (Kp) for placenta, amniotic fluid, and milk were obtained based on the observed FMS concentrations in the tissues and Css. The Kp values for all tissues were not different between high (Css = 200 ng/mL) and low (Css = 100 ng/mL) dose groups. While the mean Kp of the placenta was 44.6-59.0 %, the mean Kp was 1.3-1.7 % for the amniotic fluid and 14.9-17.0 % for fetus. The mean Kp of milk was 10.4-15.2 %. CONCLUSIONS: Placental transfer and milk excretion of FMS was relatively lower compared to other angiotensin receptor blockers.


Subject(s)
Angiotensin Receptor Antagonists/blood , Biphenyl Compounds/blood , Lactation/blood , Maternal-Fetal Exchange/physiology , Placenta/metabolism , Pyrimidines/blood , Tetrazoles/blood , Angiotensin Receptor Antagonists/administration & dosage , Animals , Animals, Newborn , Biphenyl Compounds/administration & dosage , Female , Infusions, Intravenous , Lactation/drug effects , Lactation/metabolism , Maternal-Fetal Exchange/drug effects , Placenta/drug effects , Pregnancy , Pyrimidines/administration & dosage , Rats , Rats, Sprague-Dawley , Tetrazoles/administration & dosage , Tissue Distribution/drug effects , Tissue Distribution/physiology
6.
J Clin Pharmacol ; 56(5): 576-80, 2016 May.
Article in English | MEDLINE | ID: mdl-26272450

ABSTRACT

The present study was conducted to determine the absolute bioavailability of fimasartan (FMS; Kanarb(®) ) after the single oral administration of a 60-mg tablet or a single 30-mg intravenous (IV) infusion. This investigation was a randomized, single-dose, open-labeled, two-way crossover study of 16 healthy Korean male subjects. The subjects were divided into two groups (n = 8) and each received either the oral or IV formulation followed by one-week washout period. The Cmax (ng/ml) and AUC∞ (h · ng/ml) following oral and IV administration were 62.4 ± 48.6 and 291.1 ± 121.7; and 683.3 ± 104.3 and 782.3 ± 112.7 (mean ± SD), respectively. The Tmax (h) were 3.0 h (range: 0.5-5.0 h) and 1.0 h (range: 0.8-1.0 h) in the test and reference groups, respectively. The terminal elimination half-lives (t1/2 , h) were similar (5.8 and 5.5 h, respectively) indicating that the route of administration did not influence the absorption or elimination of FMS. The systemic clearance (CL, L/h) and the volume of distribution at steady-state (Vdss , L) were 331.3 ± 444.5 L/h and 403.3 ± 710.4 L following oral administration and 39.1 ± 5.3 L/h and 42.4 ± 25.5 L following IV administration. The absolute bioavailability of the FMS tablet was 18.6%.


Subject(s)
Angiotensin Receptor Antagonists/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Pyrimidines/pharmacokinetics , Tetrazoles/pharmacokinetics , Administration, Oral , Adult , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin Receptor Antagonists/blood , Biological Availability , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/blood , Cross-Over Studies , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Pyrimidines/administration & dosage , Pyrimidines/blood , Tetrazoles/administration & dosage , Tetrazoles/blood , Young Adult
7.
Xenobiotica ; 46(1): 40-51, 2016.
Article in English | MEDLINE | ID: mdl-26068523

ABSTRACT

1. The metabolites of fimasartan (FMS), a new angiotensin II receptor antagonist, were characterized in human liver microsomes (HLM) and human subjects. 2. We developed a method for a simultaneous quantitative and qualitative analysis using predictive multiple reaction monitoring information-dependent acquisition-enhanced product ion scanning. To characterize metabolic reactions, FMS metabolites were analyzed using quadrupole-time of flight mass spectrometer in full-scan mode. 3. The structures of metabolites were confirmed by comparison of chromatographic retention times and mass spectra with those of authentic metabolite standards. 4. In the cofactor-dependent microsomal metabolism study, the half-lives of FMS were 56.7, 247.9 and 53.3 min in the presence of NADPH, UDPGA and NADPH + UDPGA, respectively. 5. The main metabolic routes in HLM were S-oxidation, oxidative desulfuration, n-butyl hydroxylation and N-glucuronidation. 6. In humans orally administered with 120 mg FMS daily for 7 days, the prominent metabolites were FMS S-oxide and FMS N-glucuronide in the 0-8-h pooled plasma sample of each subject. 7. This study characterizes, for the first time, the metabolites of FMS in humans to provide information for its safe use in clinical medicine.


Subject(s)
Biphenyl Compounds/blood , Biphenyl Compounds/metabolism , Metabolome , Microsomes, Liver/metabolism , Pyrimidines/blood , Pyrimidines/metabolism , Tetrazoles/blood , Tetrazoles/metabolism , Adult , Biphenyl Compounds/chemistry , Humans , Magnetic Resonance Spectroscopy , Male , NADP/metabolism , Pyrimidines/chemistry , Reference Standards , Spectrometry, Mass, Electrospray Ionization , Tetrazoles/chemistry , Young Adult
8.
AAPS J ; 17(5): 1210-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25990964

ABSTRACT

Enterohepatic recirculation (EHC) can greatly enhance plasma drug exposures and therapeutic effects. This study aimed to develop a population pharmacokinetic model that can simultaneously characterize the extent and time-course of EHC in three species using fimasartan, a novel angiotensin II receptor blocker, as a model drug. All fimasartan plasma concentration profiles in 32 rats (intravenous doses, 0.3-3 mg/kg; oral doses, 1-10 mg/kg), 34 dogs (intravenous doses, 0.3-1 mg/kg; oral doses, 1-10 mg/kg), and 42 healthy volunteers (single or multiple oral doses, 20-480 mg) were determined via liquid chromatography-tandem mass spectrometry (LC-MS/MS) and simultaneously modeled in S-ADAPT. The proposed model quantitatively characterized EHC in three species after oral and intravenous dosing. The median (range) fraction of drug undergoing recirculation was 76.3% (64.9-88.7%) in rats, 33.3% (24.0-45.9%) in dogs, and 65.6% (56.5-72.0%) in humans. In the presence compared with the absence of EHC, the area under the curve in plasma was predicted to be 4.22-fold (2.85-8.85) as high in rats, 1.50-fold (1.32-1.85) in dogs, and 2.91-fold (2.30-3.57) in humans. The modeled oral bioavailability in rats (median (range), 38.7% (20.0-59.8%)) and dogs (median, 7.13% to 15.4%, depending on the formulation) matched the non-compartmental estimates well. In humans, the predicted oral bioavailability was 25.1% (15.1-43.9%) under fasting and 18.2% (12.2-31.0%) under fed conditions. The allometrically scaled area under the curve predicted from rats was 420 ng·h/mL for 60 mg fimasartan compared with 424 ± 63 ng·h/mL observed in humans. The developed population pharmacokinetic model can be utilized to characterize the impact of EHC on plasma drug exposure in animals and humans.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Enterohepatic Circulation , Models, Biological , Pyrimidines/pharmacokinetics , Tetrazoles/pharmacokinetics , Administration, Intravenous , Administration, Oral , Angiotensin Receptor Antagonists/administration & dosage , Angiotensin Receptor Antagonists/pharmacokinetics , Animals , Area Under Curve , Biphenyl Compounds/administration & dosage , Chromatography, Liquid , Dogs , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Pyrimidines/administration & dosage , Rats , Rats, Sprague-Dawley , Species Specificity , Tandem Mass Spectrometry , Tetrazoles/administration & dosage
9.
Xenobiotica ; 45(1): 10-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25034008

ABSTRACT

1. Fimasartan is an angiotensin receptor II antagonist used to treat patients with hypertension. This drug is mainly excreted into bile as either the parent compound or a glucuronide conjugate. In this study, we examined the glucuronidation of fimasartan and characterized the UDP-glucuronosyltransferases (UGTs) responsible for the glucuronidation. 2. Only one type of fimasartan glucuronide was observed after incubation with pooled human liver microsomes (HLMs) and was identified as an N2-glucuronide based on comparison with an authentic standard. 3. Among the 12 UGT isoforms tested, UGT1A1, UGT1A3 and UGT2B7 showed catalytic activity toward fimasartan glucuronidation. The intrinsic clearance (CLint) of UGT1A3 was 68.5- and 21.4-fold higher than that of UGT1A1 and UGT2B7, respectively, and the estimated relative contribution of UGT1A3 in human liver was 94.1%. Both chemical inhibition and correlation studies demonstrated that fimasartan glucuronidation activity in HLMs was significantly related with UGT1A3 activity. Fimasartan glucuronide was identified as a substrate for P-glycoprotein (Pgp) and breast cancer response protein (BCRP). 4. These findings collectively indicate that UGT1A3 is the major UGT isoform responsible for the glucuronidation of fimasartan, and this glucuronide is excreted from hepatocytes via MDR1 and BCRP.


Subject(s)
Angiotensin Receptor Antagonists/metabolism , Biphenyl Compounds/metabolism , Glucuronosyltransferase/metabolism , Microsomes, Liver/enzymology , Pyrimidines/metabolism , Tetrazoles/metabolism , Glucuronosyltransferase/antagonists & inhibitors , Humans , Isoenzymes/metabolism , Kinetics
10.
Xenobiotica ; 44(10): 913-25, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24786684

ABSTRACT

1. The objectives of this study were to evaluate the pharmacokinetics and metabolism of fimasartan in rats. 2. Unlabeled fimasartan or radiolabeled [(14)C]fimasartan was dosed by intravenous injection or oral administration to rats. Concentrations of unlabeled fimasartan in the biological samples were determined by a validated LC/MS/MS assay. Total radioactivity was quantified by liquid scintillation counting and the radioactivity associated with the metabolites was analyzed by using the radiochemical detector. Metabolite identification was conducted by product ion scanning using LC/MS/MS. 3. After oral administration of [(14)C]fimasartan, total radioactivity was found primarily in feces. In bile duct cannulated rats, 58.8 ± 14.4% of the radioactive dose was excreted via bile after oral dosing. Major metabolites of fimasartan including the active metabolite, desulfo-fimasartan, were identified, yet none represented more than 7.2% of the exposure of the parent drug. Fimasartan was rapidly and extensively absorbed and had an oral bioavailability of 32.7-49.6% in rats. Fimasartan plasma concentrations showed a multi-exponential decline after oral administration. Double peaks and extended terminal half-life were observed, which was likely caused by enterohepatic recirculation. 4. These results provide better understanding on the pharmacokinetics of fimasartan and may aid further development of fimasartan analogs.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Biphenyl Compounds/pharmacokinetics , Pyrimidines/pharmacokinetics , Tetrazoles/pharmacokinetics , Administration, Oral , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/metabolism , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/metabolism , Carbon Radioisotopes/analysis , Injections, Intravenous , Pyrimidines/administration & dosage , Pyrimidines/metabolism , Rats, Sprague-Dawley , Tetrazoles/administration & dosage , Tetrazoles/metabolism
11.
Biol Pharm Bull ; 36(7): 1208-15, 2013.
Article in English | MEDLINE | ID: mdl-23811571

ABSTRACT

The pharmacological profile of BR-A-657, 2-n-butyl-5-dimethylamino-thiocarbonyl-methyl-6-methyl-3-{[2-(1H-tetrazole-5-yl)biphenyl-4-yl]methyl}-pyrimidin-4(3H)-one, a new nonpeptide AT1-selective angiotensin receptor antagonist, has been investigated in a variety of in vitro and in vivo experimental models. In the present study, BR-A-657 displaced [(125)I][Sar(1)-Ile(8)]angiotensin II (Ang II) from its specific binding sites to AT1 subtype receptors in membrane fractions of HEK-293 cells with an IC50 of 0.16 nM. In a functional assay using isolated rabbit thoracic aorta, BR-A-657 inhibited the contractile response to Ang II (pD'2: 9.15) with a significant reduction in the maximum. In conscious rats, BR-A-657 (0.01, 0.1, 1 mg/kg; intravenously (i.v.)) dose-dependently antagonized Ang II-induced pressor responses. In addition, BR-A-657 dose-dependently decreased mean arterial pressure in furosemide-treated rats and renal hypertensive rats. Moreover, BR-A-657 given orally at 1 and 3 mg/kg reduced blood pressure in conscious renal hypertensive rats. Taken together, these findings indicate that BR-A-657 is a potent and specific antagonist of Ang II at the AT1 receptor subtype, and reveal the molecular basis responsible for the marked lowering of blood pressure in conscious rats.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Biphenyl Compounds/pharmacology , Pyrimidines/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Binding Sites , Biphenyl Compounds/therapeutic use , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hypertension, Renal/drug therapy , Hypertension, Renal/metabolism , In Vitro Techniques , Male , Molecular Structure , Protein Binding , Pyrimidines/therapeutic use , Rabbits , Rats , Rats, Sprague-Dawley , Tetrazoles/therapeutic use , Vasoconstriction/drug effects
12.
Bioorg Med Chem Lett ; 23(16): 4540-6, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23845221

ABSTRACT

As a development strategy for backups of Fimasartan (1), a comparative molecular similarity indices analysis (CoMSIA) of a set of sixty-five 5-(biphenyl-2-yl)-1H-tetrazole derivatives has been performed to find out the pharmacophore elements for angiotensin II receptor type 1 (AT1) blockade. The most potent compound containing pyrimidin-4(3H)-one ring, Fimasartan (1) was used to align the molecules. As a result, we obtained 3D-QSAR model which provided good predictivity for both the training set (q(2)=0.846, r(2)=0.975) and the external test set (rpred(2)=0.980). This model would guide the design of backups for Fimasartan (1), a launched oral antihypertensive agent.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/chemistry , Biphenyl Compounds/chemistry , Computer Simulation , Models, Chemical , Tetrazoles/chemistry , Inhibitory Concentration 50 , Pyrimidines/chemistry , Quantitative Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 23(7): 2083-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23453071

ABSTRACT

Previously, we have found that BRN-103, a nicotinamide derivative, inhibits vascular endothelial growth factor (VEGF)-mediated angiogenesis signaling in human endothelial cells. During our continuous efforts to identify more potent anti-angiogenic agents, we synthesized various nicotinamide derivatives and evaluated their anti-angiogenic effects. We found that 2-{1-[1-(6-chloro-5-fluoropyrimidin-4-yl)ethyl]piperidin-4-ylamino}-N-(3-chlorophenyl) pyridine-3-carboxamide (BRN-250) significantly inhibited human umbilical vascular endothelial cells (HUVECs) proliferation, migration, tube formation, and microvessel growth in a concentration range of 10-100 nM. Furthermore, BRN-250 inhibited the VEGF-induced phosphorylation and intracellular tyrosine kinase activity of VEGF receptor 2 (VEGFR2) and the activation of its downstream AKT pathway. Taken together, these findings suggest that BRN-250 be considered a potential lead compound for cancer therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Niacinamide/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Molecular Structure , Niacinamide/chemical synthesis , Niacinamide/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Vascular Endothelial Growth Factors/metabolism
14.
Biol Pharm Bull ; 36(3): 467-74, 2013.
Article in English | MEDLINE | ID: mdl-23449332

ABSTRACT

Since inhibition of angiotensin II type 1 (AT1) receptor reduces chronic inflammation associated with hypertension, we evaluated the anti-inflammatory potential and the underlying mechanism of fimasartan, a Korean Food and Drug Administration approved anti-hypertension drug, in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Fimasartan suppressed the expressions of inducible nitric oxide synthase (iNOS) by down-regulating its transcription, and subsequently inhibited the productions of nitric oxide (NO). In addition, fimasartan attenuated LPS-induced transcriptional and DNA-binding activities of nuclear factor-kappa B (NF-κB) and activator protein-1 (AP-1). These reductions were accompanied by parallel reductions in the nuclear translocation of NF-κB and AP-1. Taken together, our data suggest that fimasartan down-regulates the expression of the iNOS in macrophages via NF-κB and AP-1 inactivation.


Subject(s)
Antihypertensive Agents/pharmacology , Biphenyl Compounds/pharmacology , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pyrimidines/pharmacology , Tetrazoles/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Animals , Cells, Cultured , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis
15.
Arch Pharm Res ; 35(7): 1123-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22864732

ABSTRACT

Fimasartan (Kanarb®), an angiotensin II receptor antagonist with selectivity for the AT1 receptor subtype, is a pyrimidinone-related heterocyclic compound that was developed by Boryung Pharm. Co., Ltd. Among numerous synthetic derivatives, fimasartan was chosen as a new drug candidate through in vitro and in vivo screening studies. Pharmadynamic-pharmacokinetic properties and safety profiles were determined in a series of nonclinical and clinical studies. Fimasartan is a new angiotensin receptor blocker, and the first new molecular entity acting on cardiovascular system approved by Korean Food and Drug Administration for the treatment of essential hypertension in September 2010. Further development process for combination therapy and overseas registration is currently ongoing.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Pyrimidines/therapeutic use , Tetrazoles/therapeutic use , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Humans , Hypertension/physiopathology , Patient Safety , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Renin-Angiotensin System/drug effects , Tetrazoles/adverse effects , Tetrazoles/pharmacokinetics
16.
Bioorg Med Chem Lett ; 22(4): 1649-54, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22264484

ABSTRACT

The discovery, in vitro and in vivo studies of the highly potent AT(1) antagonist 12a (BR-A-657, Fimasartan) antagonists are presented. A series of pyrimidin-4(3H)-one derivatives as losartan analogue were synthesized and evaluated for a novel class of AT(1) receptor antagonists. Among them, 12a containing thioamido moiety displayed both high in vitro functional antagonism and binding affinity [IC(50)=0.42 and 0.13 nM, respectively] and inhibited strongly in vivo AngII-induced pressor response in pithed rats with an ED(50) of 0.018 mg/kg. Moreover, in vivo evaluation in furosemide-treated rat and conscious renal hypertensive rat models and the pharmacokinetic study showed that 12a is a highly potent and orally active AT(1) selective antagonist having stronger in vivo potency than losartan.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/pharmacology , Losartan/chemistry , Pyrimidinones/chemical synthesis , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Antihypertensive Agents/chemistry , Inhibitory Concentration 50 , Molecular Structure , Protein Binding/drug effects , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats
17.
Bioorg Med Chem Lett ; 21(21): 6236-41, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21963305

ABSTRACT

Anti-angiogenesis is regarded as an effective strategy for cancer treatment, and vascular endothelial growth factor (VEGF) plays a key role in the regulations of angiogenesis and vasculogenesis. In the present study, the authors synthesized five novel nicotinamide derivatives which structurally mimic the receptor tyrosine kinase inhibitor sunitinib and evaluated their anti-angiogenic effects. Transwell migration assays revealed that 2-(1-benzylpiperidin-4-yl) amino-N-(3-chlorophenyl) nicotinamide (BRN-103), among the five derivatives most potently inhibited VEGF-induced human umbilical vein endothelial cells (HUVECs). In addition, BRN-103 dose-dependently inhibited VEGF-induced migration, proliferation, and capillary-like tube formation of HUVECs and vessel sprouting from mouse aortic rings. To understand the molecular mechanisms responsible for these activities, the authors examined the effect of BRN-103 on VEGF signaling pathways in HUVECs. BRN-103 was found to suppress the VEGF-induced phosphorylation of VEGF receptor 2 (VEGR2) and the activations of AKT and eNOS. Taken together, these results suggest that BRN-103 inhibits VEGF-mediated angiogenesis signaling in human endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Neovascularization, Pathologic/prevention & control , Niacinamide/analogs & derivatives , Piperidines/pharmacology , Umbilical Veins/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Niacinamide/pharmacology , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/physiology
18.
Am J Cardiovasc Drugs ; 11(5): 335-46, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21910510

ABSTRACT

BACKGROUND AND OBJECTIVES: Fimasartan (BR-A-657) is a novel, non-peptide angiotensin II receptor antagonist with a selective type I receptor blockade effect. Two first-in-human studies investigated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of fimasartan. METHODS: Fasted single oral tablet doses of fimasartan 20-480 mg or placebo were administered to 40 healthy male subjects (aged 19-54 years) in a double-blind, randomized, sequential-group design. Subjects receiving fimasartan 240 mg also received the same treatment in the fed state after an interval of 7 days. In another study, oral tablet doses of fimasartan 120 and 360 mg or placebo were given once daily for 7 days to groups of eight fasted healthy male subjects (aged 20-55 years) in a double-blind, randomized, sequential-group design. Safety and tolerability were assessed. The PK and PD of fimasartan were also evaluated and compared for the different doses. RESULTS: Fimasartan was safe and well tolerated, but with an increased incidence of low BP and postural dizziness for the 360 mg dose after repeated administration. Fimasartan produced increases in plasma renin activity, angiotensin I and II, which were not dose dependent. Maximal increases occurred between 6 and 8 hours post-dose, lasting up to 48 hours. Fimasartan was absorbed rapidly after all doses and had a multiphasic distribution. Two peaks in the plasma concentration-time profile were observed in most subjects. Steady state was achieved after three doses, and accumulation was minimal after repeated doses for 7 days (24-30%). The effective half-life ranged from 9.84 to 13.2 hours. The systemic exposure of fimasartan was dose proportional, and no marked food effect was noted after administration of 240 mg in the fed state. Urinary excretion of fimasartan was very low (1.74-2.51%), suggesting non-renal elimination. CONCLUSION: Fimasartan had a good safety profile and was well tolerated after fasted single oral doses of 20-480 mg, a fed single oral dose of 240 mg, and fasted repeated oral doses of 120 and 360 mg in healthy subjects. In addition, the PK and PD of fimasartan in this population were well characterized. Further studies are needed to evaluate the safety, efficacy, and dose-response relationship of fimasartan in patients with hypertension.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Food-Drug Interactions , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Tetrazoles/adverse effects , Tetrazoles/pharmacokinetics , Administration, Oral , Adult , Angiotensin I/blood , Angiotensin II/blood , Angiotensin II Type 1 Receptor Blockers/blood , Angiotensin II Type 1 Receptor Blockers/urine , Antihypertensive Agents/adverse effects , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Biphenyl Compounds/blood , Biphenyl Compounds/urine , Blood Pressure/drug effects , Dizziness/chemically induced , Dose-Response Relationship, Drug , Double-Blind Method , Half-Life , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Middle Aged , Pyrimidines/blood , Pyrimidines/urine , Renin/blood , Tetrazoles/blood , Tetrazoles/urine , Young Adult
19.
Biomed Chromatogr ; 25(11): 1208-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21268050

ABSTRACT

Fimasartan, 2-butyl-5-dimethylaminothiocarbonylmethyl-6-methyl-3-[[2'-(1H tetrazol -5-yl)biphenyl-4-yl]methyl]pyrimidin-4(3H)-one (BR-A-657), is a novel angiotensin II receptor blocker exhibiting potent and selective AT1 receptor blocking activity. This study reports the liquid chromatography-tandem mass spectrometry assay for the simultaneous determination of fimasartan and its active metabolite, BR-A-557, in rat plasma. The assay was validated to demonstrate the specificity, linearity, recovery, lower limit of quantification, accuracy, precision and stability. The multiple reaction monitoring was based on the transition of m/z 502.1 → 207.1 for fimasartan, 486.2 → 207.1 for BR-A-557 and 526.1 → 207.1 for BR-A-563 (internal standard). The assay utilized a simple precipitation procedure with acetonitrile and isocratic elution. The LLOQ was 0.2 ng/mL for fimasartan and BR-A-557 using 50 µL plasma samples. The assay was linear over a concentration range from 0.2 to 500 ng/mL for fimasartan and BR-A-557, with correlation coefficients >0.9995. The intra- and inter-day assay accuracies were 93.6-108.0 and 90.8-101.4% for fimasartan and 102.2-107.1 and 99.6-103.3% for BR-A-557, respectively. The intra- and inter-day precision were 2.4-4.4 and 3.0-13.4% for fimasartan and 3.1-5.2 and 2.8-9.8% for BR-A-557, respectively. The developed assay may be used to study the metabolism and mechanistic pharmacokinetics of fimasartan in future studies.


Subject(s)
Antihypertensive Agents/blood , Biphenyl Compounds/blood , Chromatography, Liquid/methods , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Tetrazoles/blood , Animals , Antihypertensive Agents/chemistry , Biphenyl Compounds/chemistry , Linear Models , Male , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Tetrazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...