Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(9)2023 May 08.
Article in English | MEDLINE | ID: mdl-37176129

ABSTRACT

Sensitivity to opioids varies widely among individuals. To identify potential candidate single-nucleotide polymorphisms (SNPs) that may significantly contribute to individual differences in the minimum effective concentration (MEC) of an opioid, fentanyl, we conducted a three-stage genome-wide association study (GWAS) using whole-genome genotyping arrays in 350 patients who underwent laparoscopic-assisted colectomy. To estimate the MEC of fentanyl, plasma and effect-site concentrations of fentanyl over the 24 h postoperative period were estimated with a pharmacokinetic simulation model based on initial bolus doses and subsequent patient-controlled analgesia doses of fentanyl. Plasma and effect-site MECs of fentanyl were indicated by fentanyl concentrations, estimated immediately before each patient-controlled analgesia dose. The GWAS revealed that an intergenic SNP, rs966775, that mapped to 5p13 had significant associations with the plasma MEC averaged over the 6 h postoperative period and the effect-site MEC averaged over the 12 h postoperative period. The minor G allele of rs966775 was associated with increases in these MECs of fentanyl. The nearest protein-coding gene around this SNP was DRD1, encoding the dopamine D1 receptor. In the gene-based analysis, the association was significant for the SERP2 gene in the dominant model. Our findings provide valuable information for personalized pain treatment after laparoscopic-assisted colectomy.


Subject(s)
Fentanyl , Laparoscopy , Humans , Genome-Wide Association Study , Pain, Postoperative/etiology , Pain, Postoperative/genetics , Analgesics, Opioid/therapeutic use , Polymorphism, Single Nucleotide , Colectomy
2.
Clin Transl Sci ; 15(6): 1519-1531, 2022 06.
Article in English | MEDLINE | ID: mdl-35421902

ABSTRACT

The accurate prediction of OATP1B-mediated drug-drug interactions (DDIs) is challenging for drug development. Here, we report a physiologically-based pharmacokinetic (PBPK) model analysis for clinical DDI data generated in heathy subjects who received oral doses of cyclosporin A (CysA; 20 and 75 mg) as an OATP1B inhibitor, and the probe drugs (pitavastatin, rosuvastatin, and valsartan). PBPK models of CysA and probe compounds were combined assuming inhibition of hepatic uptake of endogenous coproporphyrin I (CP-I) by CysA. In vivo Ki of unbound CysA for OATP1B (Ki,OATP1B ), and the overall intrinsic hepatic clearance per body weight of CP-I (CLint,all,unit ) were optimized to account for the CP-I data (Ki,OATP1B , 0.536 ± 0.041 nM; CLint,all,unit , 41.9 ± 4.3 L/h/kg). DDI simulation using Ki,OATP1B reproduced the dose-dependent effect of CysA (20 and 75 mg) and the dosing interval (1 and 3 h) on the time profiles of blood concentrations of pitavastatin and rosuvastatin, but DDI simulation using in vitro Ki,OATP1B failed. The Cluster Gauss-Newton method was used to conduct parameter optimization using 1000 initial parameter sets for the seven pharmacokinetic parameters of CP-I (ß, CLint, all , Fa Fg , Rdif , fbile , fsyn , and vsyn ), and Ki,OATP1B and Ki,MRP2 of CysA. Based on the accepted 546 parameter sets, the range of CLint, all and Ki,OATP1B was narrowed, with coefficients of variation of 12.4% and 11.5%, respectively, indicating that these parameters were practically identifiable. These results suggest that PBPK model analysis of CP-I is a promising translational approach to predict OATP1B-mediated DDIs in drug development.


Subject(s)
Coproporphyrins , Models, Biological , Drug Interactions , Humans , Liver-Specific Organic Anion Transporter 1 , Rosuvastatin Calcium
3.
Clin Pharmacol Ther ; 111(6): 1315-1323, 2022 06.
Article in English | MEDLINE | ID: mdl-35292967

ABSTRACT

This study was designed to assess the quantitative performance of endogenous biomarkers for organic anion transporting polypeptide (OATP) 1B1/1B3-mediated drug-drug interactions (DDIs). Ten healthy volunteers orally received OATP1B1/1B3 probe cocktail (0.2 mg pitavastatin, 1 mg rosuvastatin, and 2 mg valsartan) and an oral dose of cyclosporin A (CysA, 20 mg and 75 mg) separated by a 1-hour interval (20 mg (-1 hour), and 75 mg (-1 hour)). CysA 75 mg was also given with a 3-hour interval (75 mg (-3 hours)) to examine the persistence of OATP1B1/1B3 inhibition. The area under the plasma concentration-time curve ratios (AUCRs) were 1.63, 3.46, and 2.38 (pitavastatin), 1.39, 2.16, and 1.81 (rosuvastatin), and 1.42, 1.77, and 1.85 (valsartan), at 20 mg, 75 mg (-1 hour) and 75 mg (-3 hours) of CysA, respectively. CysA effect on OATP1B1/1B3 was unlikely to persist at the dose examined. Among 26 putative OATP1B1/1B3 biomarkers evaluated, AUCR and maximum concentration ratio (Cmax R) of CP-I showed the highest Pearson's correlation coefficient with CysA AUC (0.94 and 0.93, respectively). Correlation between AUCR of pitavastatin, and Cmax R or AUCR of CP-I were consistent between this study and our previous study using rifampicin as an OATP1B1/1B3 inhibitor. Nonlinear regression analysis of AUCR-1 of pitavastatin and CP-I against CysA Cmax yielded Ki,OATP1B1/1B3,app (109 ± 35 and 176 ± 42 nM, respectively), similar to the Ki ,OATP1B1/1B3 estimated by our physiologically-based pharmacokinetic model analysis described previously (107 nM). The endogenous OATP1B1/1B3 biomarkers, particularly Cmax R and AUCR of CP-I, corroborates OATP1B1/1B3 inhibition and yields valuable information that improve accurate DDI predictions in drug development, and enhance our understanding of interindividual variability in the magnitude of DDIs.


Subject(s)
Cyclosporine , Organic Anion Transporters , Cyclosporine/pharmacology , Drug Interactions , Humans , Liver-Specific Organic Anion Transporter 1 , Rosuvastatin Calcium/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3 , Valsartan
4.
Sci Rep ; 8(1): 16841, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442903

ABSTRACT

Aromatase, an enzyme that converts androgens to estrogens, has been reported to be involved in several brain functions, including synaptic plasticity, neurogenesis, neuroprotection, and regulation of sexual and emotional behaviours in rodents, pathophysiology of Alzheimer's disease and autism spectrum disorders in humans. Aromatase has been reported to be involved in aggressive behaviours in genetically modified mice and in personality traits by genotyping studies on humans. However, no study has investigated the relationship between aromatase in living brains and personality traits including aggression. We performed a positron emission tomography (PET) study in 21 healthy subjects using 11C-cetrozole, which has high selectivity and affinity for aromatase. Before performing PET scans, subjects answered the Buss-Perry Aggression Questionnaire and Temperament and Character Inventory to measure their aggression and personality traits, respectively. A strong accumulation of 11C-cetrozole was detected in the thalamus, hypothalamus, amygdala, and medulla. Females showed associations between aromatase levels in subcortical regions, such as the amygdala and supraoptic nucleus of the hypothalamus, and personality traits such as aggression, novelty seeking, and self-transcendence. In contrast, males exhibited associations between aromatase levels in the cortices and harm avoidance, persistence, and self-transcendence. The association of aromatase levels in the thalamus with cooperativeness was common to both sexes. The present study suggests that there might exist associations between aromatase in the brain and personality traits. Some of these associations may differ between sexes, while others are likely common to both.


Subject(s)
Aromatase/metabolism , Brain/enzymology , Personality , Adult , Aniline Compounds , Carbon Radioisotopes , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Triazoles
5.
Drug Metab Pharmacokinet ; 32(6): 293-300, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29137842

ABSTRACT

The aim of the present study is to investigate the pharmacokinetics of our newly developed aromatase inhibitors (cetrozole and TMD-322) in healthy subjects by a cassette microdose strategy. A cocktail of cetrozole and TMD-322 was administered intravenously or orally (1.98 µg for each drug) to six healthy volunteers in a crossover fashion. Anastrozole (1.98 µg) was also included in the oral cocktail. Total body clearance and bioavailability were 12.1 ± 7.1 mL/min/kg and 34.9 ± 32.3% for cetrozole, and 16.8 ± 3.5 mL/min/kg and 18.4 ± 12.2% for TMD-322, respectively. The area under the plasma concentration-time curves of cetrozole and TMD-322 after oral administration was markedly lower than that of anastrozole because of their high hepatic clearance. Two subjects out of six exhibited 4- and 17-fold larger exposure of cetrozole than the others following intravenous and oral administration, respectively. Such variation was not observed for TMD-322 and anastrozole. Extensive metabolism of cetrozole and TMD-322 was observed in the CYP2C19 expression system among the test CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). We report the first clinical investigation of our aromatase inhibitors by a cassette microdose strategy in healthy Japanese subjects. This strategy offers an optional approach for candidate selection as a phase zero study in drug development.


Subject(s)
Aniline Compounds/pharmacokinetics , Aromatase Inhibitors/pharmacokinetics , Aromatase/metabolism , Drug Discovery , Nitriles/pharmacokinetics , Triazoles/pharmacokinetics , Administration, Intravenous , Administration, Oral , Aged , Anastrozole , Aniline Compounds/administration & dosage , Aniline Compounds/chemistry , Animals , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/chemistry , Cross-Over Studies , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Japan , Male , Molecular Structure , Nitriles/administration & dosage , Nitriles/chemistry , Rats , Structure-Activity Relationship , Triazoles/administration & dosage , Triazoles/chemistry
6.
Pharm Res ; 33(5): 1235-48, 2016 May.
Article in English | MEDLINE | ID: mdl-26869173

ABSTRACT

PURPOSE: To select appropriate antiemetics relieving teriparatide-induced nausea and vomiting during osteoporosis treatment using PET molecular imaging and pharmacokinetic analysis. METHODS: Rats were pretreated with subcutaneous teriparatide, followed by oral administration of antiemetics with different pharmacological effects. The pharmacokinetics of antiemetics were assessed by oral administration of 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG) under free moving conditions in vivo. The effect of teriparatide on the permeability of Caco-2 cell membranes to [(18)F]FDG was assessed in vitro. The effects of antiemetics on teriparatide-induced suppression of gastrointestinal motility in vivo was assayed by positron emission tomography (PET) using orally administered [(18)F]FDG. RESULTS: Teriparatide delayed the time-radioactivity profile of [(18)F]FDG in blood and significantly reduced its absorption rate constant (k a ), determined from non-compartmental analysis, to 60% of control. In contrast, co-administration of granisetron or mosapride restored the time-radioactivity profile and k a of [(18)F]FDG to control levels. Teriparatide had no effect on Caco-2 membrane permeability to [(18)F]FDG. Pharmacokinetic PET imaging data analysis quantitatively showed the pharmacological effects of teriparatide-induced suppression of upper gastrointestinal motility and its restoration by granisetron and mosapride. CONCLUSIONS: Teriparatide-induced abdominal discomfort might be attributed to GI motility, and PET imaging analysis is a useful tool to for the selection of appropriate antiemetics.


Subject(s)
Antiemetics/therapeutic use , Benzamides/therapeutic use , Bone Density Conservation Agents/adverse effects , Granisetron/therapeutic use , Morpholines/therapeutic use , Nausea/drug therapy , Teriparatide/adverse effects , Vomiting/drug therapy , Animals , Caco-2 Cells , Gastrointestinal Absorption/drug effects , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/metabolism , Humans , Male , Nausea/chemically induced , Nausea/physiopathology , Osteoporosis/drug therapy , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Vomiting/chemically induced , Vomiting/physiopathology
7.
Pharm Res ; 32(8): 2538-47, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25715695

ABSTRACT

PURPOSE: To evaluate the function of multidrug and toxin extrusion proteins (MATEs) using (11)C-labeled metformin ([(11)C]metformin) by positron emission tomography (PET). METHODS: PET was performed by intravenous bolus injection of [(11)C]metformin. Pyrimethamine at 0.5 and 5 mg/kg was intravenously administered to mice 30 min prior to the scan. Integration plot analysis was conducted for calculating liver (CLuptake,liver), kidney (CLuptake,kidney) tissue uptake, intrinsic biliary (CLint,bile) and urinary (CLint,urine) excretion clearances of [(11)C]metformin. RESULTS: Visualization by PET showed that pyrimethamine increased concentrations of [(11)C]metformin in the liver and kidneys, and decreased the concentrations in the urinary bladder without changing the blood profiles. Pyrimethamine had no effect on the CLuptake,liver and CLuptake,kidney, which were similar to the blood-flow rate. CLint,bile with regard to the liver concentration was unable to be determined, but administration of 0.5 and 5 mg/kg of pyrimethamine increased the liver-to-blood ratio to 1.6 and 2.3-fold, respectively, indicating that pyrimethamine inhibited the efflux of [(11)C]metformin from the liver. CLint,urine with regard to the corticomedullary region concentrations was decreased 37 and 68% of the control by administration of 0.5 and 5 mg/kg of pyrimethamine, respectively (P < 0.05). CONCLUSIONS: Tissue concentration based investigations using [(11)C]metformin by PET enables the functional analysis of MATEs in the liver and kidneys.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Animals , Biliary Tract/metabolism , Drug Interactions , Hypoglycemic Agents/blood , Hypoglycemic Agents/urine , Kidney/metabolism , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Liver/metabolism , Male , Metformin/blood , Metformin/urine , Mice , Positron-Emission Tomography , Pyrimethamine/pharmacology , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/urine
8.
Nucl Med Biol ; 41(7): 594-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24853403

ABSTRACT

INTRODUCTION: Neuroinflammatory processes play an important role in the pathogenesis of Alzheimer's disease and other brain disorders, and nonsteroidal anti-inflammatory drugs (NSAIDs) are considered therapeutic candidates. As a biomarker of neuroinflammatory processes, (11)C-labeled ketoprofen methyl ester ([(11)C]KTP-Me) was designed to allow cerebral penetration of ketoprofen (KTP), an active form of a selective cyclooxygenase-1 inhibitor that acts as an NSAID. Rat neuroinflammation models indicate that [(11)C]KTP-Me enters the brain and is retained in inflammatory lesions, accumulating in activated microglia. [(11)C]KTP-Me is washed out from normal tissues, leading to the present first-in-human exploratory study. METHODS: [(11)C]KTP-Me was synthesized by rapid C-[(11)C]methylation of [(11)C]CH3I and the corresponding arylacetate precursor, purified with high-performance liquid chromatography, and prepared as an injectable solution including PEG400, providing radiochemical purity of >99% and specific activity of >25GBq/µmol at injection. Six young healthy male humans were injected with [(11)C]KTP-Me and scanned with PET camera to determine the early-phase brain time course followed by three whole-body scans starting 8, 20, and 40 min post-injection, together with sequential blood sampling and labeled metabolite analysis. RESULTS: No adverse effects were observed during PET scanning after [(11)C]KTP-Me injection. [(11)C]KTP-Me was rapidly metabolized to (11)C-labeled ketoprofen ([(11)C]KTP) within 2-3 min and was gradually cleared from blood. The radioactivity entered the brain with an average peak cortical SUV of 1.5 at 2 min. The cortical activity was gradually washed out. Whole-body images indicated that the urinary bladder was the major excretory pathway. The organ with the highest radiation dose was the urinary bladder (average dose of 41µGy/MBq, respectively). The mean effective dose was 4.7µSv/MBq, which was comparable to other (11)C-labeled radiopharmaceuticals. CONCLUSION: [(11)C]KTP-Me demonstrated a favorable dosimetry, biodistribution, and safety profile. [(11)C]KTP-Me entered the human brain, and the radioactivity was washed out from cerebral tissue. These data warrant further exploratory studies on patients with neuroinflammation.


Subject(s)
Brain/diagnostic imaging , Ketoprofen/analogs & derivatives , Positron-Emission Tomography/methods , Adult , Animals , Biological Transport , Humans , Inflammation/diagnostic imaging , Ketoprofen/adverse effects , Ketoprofen/metabolism , Ketoprofen/pharmacokinetics , Male , Radioactive Tracers , Radiometry , Rats , Safety , Tissue Distribution
9.
J Nucl Med ; 55(5): 852-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24676756

ABSTRACT

UNLABELLED: Aromatase (an enzyme that converts androgens to estrogens) in the brain is involved in neuroprotection, synaptic plasticity, and regulation of sexual and emotional behaviors. To investigate the physiologic and pathologic importance of aromatase in the brain, including in humans, we here report the development of a novel PET probe for aromatase, (11)C-cetrozole, which allows noninvasive quantification of aromatase expression. METHODS: (11)C-cetrozole was synthesized by the C-(11)C-methylation method developed by our group. In vitro autoradiography of frozen sections and a binding study with rat brain homogenates were conducted to demonstrate the specific binding and the dissociation constant. PET studies with anesthetized rhesus monkeys were performed to analyze the dynamics in the brain. RESULTS: In vitro and in vivo studies using (11)C-cetrozole showed its superiority in brain aromatase imaging in terms of specificity and selectivity, compared with previously developed (11)C-vorozole. PET studies showed that (11)C-cetrozole had a higher signal-to-noise ratio, providing a sharper image than (11)C-vorozole, because the radioactive metabolite of (11)C-vorozole was taken up into the brain. High specific binding of (11)C-cetrozole was observed in the amygdala and hypothalamus, and we also noted binding in the nucleus accumbens of rhesus monkeys for the first time. CONCLUSION: These results suggest that PET imaging with newly developed (11)C-cetrozole is suitable for quantifying the expression of brain aromatase in vivo, possibly providing critical information regarding the functional roles of aromatase in human neurologic and emotional disorders.


Subject(s)
Aniline Compounds/chemical synthesis , Aromatase/chemistry , Brain/diagnostic imaging , Carbon Radioisotopes , Positron-Emission Tomography , Triazoles/chemical synthesis , Animals , Aromatase Inhibitors/chemistry , Autoradiography , Brain/metabolism , Female , Inhibitory Concentration 50 , Macaca mulatta , Male , Methylation , Protein Binding , Rats , Rats, Sprague-Dawley , Tissue Distribution , Triazoles/chemistry
10.
Bioorg Med Chem ; 21(24): 7584-90, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24238901

ABSTRACT

In order to develop a new positron emission tomography (PET) probe to study hepatobiliary transport mediated by the multi-drug and toxin extrusion transporter 1 (MATE1), (11)C-labelled metformin was synthesized and then evaluated as a PET probe. [(11)C]Metformin ([(11)C]4) was synthesized in three steps, from [(11)C]methyl iodide. Evaluation by small animal PET of [(11)C]4 showed that there was increased concentrations of [(11)C]4 in the livers of mice pre-treated with pyrimethamine, a potential inhibitor of MATEs, inhibiting the hepatobiliary excretion of metformin. Radiometabolite analysis showed that [(11)C]4 was not degraded in vivo during the PET scan. Biodistribution studies were undertaken and the organ distributions were extrapolated into a standard human model. In conclusion, [(11)C]4 may be useful as a PET probe to non-invasively study the in vivo function of hepatobiliary transport and drug-drug interactions, mediated by MATE1 in future clinical investigations.


Subject(s)
Liver/metabolism , Metformin/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Positron-Emission Tomography , Animals , Biological Transport , Carbon Isotopes , Male , Metformin/chemical synthesis , Metformin/chemistry , Mice , Tissue Distribution
11.
J Pharmacol Exp Ther ; 347(1): 193-202, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926287

ABSTRACT

We developed a pravastatin derivative, sodium (3R,5R)-3,5-dihydroxy-7-((1S,2S,6S,8S)-6-hydroxy-2-methyl-8-((1-[(11)C]-(E)-2-methyl-but-2-enoyl)oxy)-1,2,6,7,8,8a-hexahydronaphthalen-1-yl)heptanoate ([(11)C]DPV), as a positron emission tomography (PET) probe for noninvasive measurement of hepatobiliary transport, and conducted pharmacokinetic analysis in rats as a feasibility study for future clinical study. Transport activities of DPV in freshly isolated rat hepatocytes and rodent multidrug resistance-associated protein 2 (rMrp2; human, MRP2)-expressing membrane vesicles were similar to those of pravastatin. Rifampicin diminished the uptake of DPV and pravastatin by the hepatocytes, with similar inhibition potency. [(11)C]DPV underwent biotransformation to produce at least two metabolites in rat, but metabolism of [(11)C]DPV occurred negligibly in human hepatocytes during a 90-minute incubation. After intravenous injection, [(11)C]DPV was mainly distributed to the liver and kidneys, where the tissue uptake clearances (CLuptake,liver and CLuptake,kidney) were blood-flow-limited (73.6 ± 4.8 and 24.6 ± 0.6 ml/min per kilogram, respectively). Systemic elimination of [(11)C]DPV was delayed in rifampicin-treated rat and an Mrp2-deficient mutant rat, Eisai hyperbilirubinemic mutant rat (EHBR). Rifampicin treatment decreased both CLuptake,liver and CLuptake,kidney of [(11)C]DPV by 30% (P < 0.05), whereas these parameters were unchanged in EHBR. Meanwhile, the canalicular efflux clearance (CLint,bile) of [(11)C]DPV, which was 12.2 ± 1.5 ml/min per kilogram in the control rat, decreased by 60% and 89% in rifampicin-treated rat and EHBR (P < 0.05), respectively. These results indicate that [(11)C]DPV is taken up into the liver by organic anion-transporting polypeptides (rodent, Oatps; human, OATP) and excreted into bile by Mrp2 in rat, and that rifampicin may inhibit Mrp2 as well as Oatps, and consequently increase systemic exposure of [(11)C]DPV. PET using [(11)C]DPV is feasible for studies prior to the future clinical investigation of OATP and MRP2 functionality, especially for personalized medicine.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Biliary Tract/metabolism , Hepatocytes/metabolism , Organic Anion Transporters/metabolism , Positron-Emission Tomography , Pravastatin/metabolism , ATP-Binding Cassette Transporters/physiology , Animals , Biliary Tract/diagnostic imaging , Carbon Radioisotopes/metabolism , Coculture Techniques , Drug Evaluation, Preclinical/methods , Feasibility Studies , Hepatocytes/diagnostic imaging , Humans , Male , Organic Anion Transporters/physiology , Positron-Emission Tomography/methods , Pravastatin/chemistry , Rats , Rats, Sprague-Dawley
12.
J Pharm Sci ; 102(9): 3309-19, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712697

ABSTRACT

The brain distribution of nonsteroidal aromatase inhibitors was investigated in mice to understand their interactions with brain aromatase. The brain-to-plasma ratio (Kp,brain , mL/g brain) of anastrozole was 0.0299 ± 0.0068, which was lower than that of letrozole (0.383 ± 0.048) and vorozole (0.185 ± 0.031) despite their similar physicochemical properties. The brain-to-plasma unbound concentration ratio of anastrozole, measured using microdialysis, was 0.118 ± 0.037 mL/g brain. In situ mouse brain perfusion also demonstrated that the uptake clearance [mL/(min·g brain)] of anastrozole by the brain (0.108 ± 0.018) was lower than that for letrozole and vorozole (0.422 ± 0.068 and 0.910 ± 0.152, respectively). Anastrozole and vorozole were transported by P-glycoprotein (P-gp) in vitro, whereas none of the compounds were transported by breast cancer resistance protein (BCRP). The Kp,brain of anastrozole and vorozole were increased by 12- and 3.3-fold, respectively, in Mdr1a/b/Bcrp(-/-) mice. IC50 (nM) of anastrozole and letrozole against human aromatase was 12.9 ± 0.7 and 3.59 ± 0.75, respectively. Taken together, these results suggest that active efflux mediated by P-gp at the blood-brain barrier limits the effect of anastrozole in the central nervous system, whereas vorozole and letrozole easily traverse the barrier.


Subject(s)
Aromatase Inhibitors/pharmacokinetics , Blood-Brain Barrier/metabolism , Nitriles/metabolism , Nitriles/pharmacokinetics , Triazoles/metabolism , Triazoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Anastrozole , Animals , Aromatase/metabolism , Aromatase Inhibitors/blood , Aromatase Inhibitors/metabolism , Aromatase Inhibitors/pharmacology , Brain/metabolism , Cell Line , Humans , Letrozole , Male , Mice , Nitriles/blood , Nitriles/pharmacology , Triazoles/blood , Triazoles/pharmacology
13.
Mol Pharm ; 10(6): 2261-9, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23600944

ABSTRACT

To develop potent drugs for oral use, information on their pharmacokinetic (PK) properties after oral administration is of great importance. We have recently reported the utility of positron emission tomography (PET) for the analysis of gastrointestinal (GI) absorption of radiolabeled compounds. In this study, PET image analysis was performed in rats using a novel PET probe, [(18)F]deoxyfluoropoly(ethylene glycol)s, with an average molecular weight of 2 kDa ([(18)F]FPEG), as a nonabsorbable marker to elaborate the GI physiology in more detail, such as segmental transition of the administered water, and fluid volume and distribution in the intestine. After oral administration of [(18)F]FPEG solution to rats, a 90 min PET scan with continuous blood sampling was performed, and then the disposition of radioactivity in each part of GI tract was investigated. From blood PK analysis, it was confirmed that the bioavailability of [(18)F]FPEG was quite low in rats. PET image analysis showed that the radioactivity after oral administration of [(18)F]FPEG solution rapidly passed through the stomach, spread into the proximal small intestine, and then transited toward the distal region of the small intestine without decreasing the radioactivity during GI transition. Radiometabolite analysis revealed that the radioactivity in intestinal mucosal tissues, blood, and urine was mainly derived from unchanged [(18)F]FPEG. It was also found that the volume of interest (VOI) after oral administration of the radiotracer enables an understanding of the time-dependent manner of effective fluid volume changes in the stomach and the small intestine. In addition, the rate constant of the intestinal transition of radioactivity in each intestinal segment was calculated by kinetic model analysis, which revealed that PET analysis enables us to determine the GI transit from the same individuals and that it is applicable to determine site-specific intestinal absorption. In conclusion, we demonstrated the high potency of PET imaging technique to elucidate the distribution of orally administered solution in the GI tract in vivo.


Subject(s)
Ethylene Glycol/chemistry , Gastrointestinal Tract/metabolism , Positron-Emission Tomography/methods , Administration, Oral , Animals , Male , Rats , Rats, Sprague-Dawley
14.
J Nucl Med ; 54(2): 267-76, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23287578

ABSTRACT

UNLABELLED: A quantitative PET imaging method was used to assess the in vivo kinetics of hepatobiliary and renal excretion of the breast cancer resistance protein (Bcrp) substrate (11)C-SC-62807 in mice. METHODS: Serial abdominal PET scans were collected in wild-type and Bcrp knockout (Bcrp(-/-)) mice after intravenous injection of (11)C-SC-62807. Venous blood samples and PET images were obtained at frequent intervals up to 30 min after radiotracer administration. Dynamic PET data were analyzed to determine the canalicular and brush-border efflux clearances in the liver and kidney (CL(int,bile,liver) and CL(int,urine,kidney), respectively). RESULTS: SC-62807 is an in vitro substrate of mouse Bcrp and human BCRP. Radioactivity associated with (11)C-SC-62807 was predominantly found in the blood, liver, bile, and urine 30 min after administration. Both biliary and urinary excretion of radioactivity was markedly lower in Bcrp(-/-) mice than in wild-type mice, suggesting greater systemic exposure in Bcrp(-/-) mice. Both the CL(int,bile,liver) and the CL(int,urine,kidney) were significantly lower in Bcrp(-/-) mice (74% ± 10% and 99% ± 1% lower than controls, respectively). We also found that (11)C-SC-62807 is a substrate of the organic anion-transporting polypeptides OATP1B1 and OATP1B3 in vitro. CONCLUSION: The present study demonstrated that Bcrp plays a significant role in the efflux of (11)C-SC-62807 in mouse liver and kidney. We also demonstrated the feasibility of PET using (11)C-SC-62807 to study the activity of BCRP in humans.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Biliary Tract/metabolism , Carbon Radioisotopes/pharmacology , Gene Expression Regulation , Kidney/metabolism , Liver/metabolism , Neoplasm Proteins/metabolism , Positron-Emission Tomography/methods , Pyrazoles/pharmacology , Sulfonamides/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Area Under Curve , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kinetics , Male , Mice , Models, Chemical , Time Factors
15.
Pharm Res ; 29(9): 2419-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22618800

ABSTRACT

PURPOSE: To dynamically analyze the processes of oral absorption and hepatobiliary distribution of telmisartan using positron emission tomography (PET). METHODS: (11)C-labeled telmisartan ([(11)C]TEL) was orally administered to rats with or without non-radiolabeled telmisartan (0.5, and 10 mg/kg). PET scanning of abdominal region and whole body was performed under conscious condition. In situ intestinal closed loop study in rats and in vitro permeation study in MDR1-MDCK II cell monolayers were also conducted. RESULTS: After oral administration of [(11)C]TEL, systemic bioavailability and hepatic distribution of radioactivity increased non-linearly with dose. In the intestinal lumen, both telmisartan and its glucuronide were detected and the ratio of telmisartan decreased dramatically at high dose of telmisartan. In situ closed loop study showed most of telmisartan-glucuronide detected in the intestinal lumen was derived from the bile excretion. In addition, in vitro permeation study revealed that telmisartan is a substrate of P-glycoprotein. CONCLUSION: PET imaging analysis successfully demonstrated the processes of intestinal absorption and hepatic distribution of telmisartan. PET study combined with appropriate in situ and in vitro experiments is highly expected to be a potent tool for better understanding of GI absorption and subsequent tissue distribution of various drugs and drug candidates.


Subject(s)
Benzimidazoles/pharmacokinetics , Benzoates/pharmacokinetics , Gastrointestinal Tract/metabolism , Liver/metabolism , Positron-Emission Tomography/methods , Animals , Biological Availability , Intestinal Absorption , Male , Rats , Rats, Sprague-Dawley , Telmisartan
16.
Bioorg Med Chem ; 20(12): 3703-9, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22607882

ABSTRACT

Drug transporters mediate the uptake and elimination of drugs in various organs; therefore, having knowledge of how a transporter functions in the body would play a key role in ensuring drug efficacy in in vivo systems. In this context, we designed and synthesized [(11)C]dehydropravastatin, a novel PET probe that would be potentially useful for evaluation of the functions of the OATP1B1 and MRP2 transporters, based on the use of palladium(0)-mediated rapid C-[(11)C]methylation (viz., the rapid cross-coupling between [(11)C]methyl iodide and a boron intermediate).


Subject(s)
ATP-Binding Cassette Transporters/analysis , Liver/chemistry , Organic Anion Transporters/analysis , Positron-Emission Tomography , Pravastatin/analogs & derivatives , Radiopharmaceuticals/chemical synthesis , ATP-Binding Cassette Transporters/metabolism , Animals , Liver/metabolism , Liver-Specific Organic Anion Transporter 1 , Male , Organic Anion Transporters/metabolism , Pravastatin/chemical synthesis , Pravastatin/chemistry , Radiopharmaceuticals/chemistry , Rats , Rats, Sprague-Dawley
17.
J Nucl Med ; 53(5): 741-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22499612

ABSTRACT

UNLABELLED: It is well accepted that drug transporters play a pivotal role in hepatobiliary excretion of anionic drugs, in which drug-drug interactions and genetic polymorphisms are known to cause variations. However, PET probes for in vivo functional characterization of these transporters have not been established yet. We used PET to investigate hepatic uptake and subsequent canalicular efflux of (11)C-labeled (15R)-16-m-tolyl-17,18,19,20-tetranorisocarbacyclin methyl ester [(15R)-(11)C-TIC-Me)] in healthy subjects. METHODS: Serial PET scans of the abdominal region in healthy male subjects were obtained with or without the organic anion-transporting polypeptide (OATP) inhibitor rifampicin after intravenous injection of (15R)-(11)C-TIC-Me as a radiotracer. Venous blood samples and PET images were obtained at frequent intervals up to 30 min after administration of the PET tracer. Dynamic imaging data were evaluated by integration plots of data collected for 2-10 min and for 10-30 min after tracer administration for the determination of tissue uptake clearance and biliary efflux clearance, respectively. RESULTS: After rapid hydrolysis in blood, the acid form-(11)C-labeled (15R)-16-m-tolyl-17,18,19,20-tetranorisocarbacyclin [(15R)-(11)C-TIC]-accumulated in the liver (37% of the dose by 17 min), and the radioactivity was then excreted into the bile (6.2% by 30 min). Rifampicin (600 mg by mouth), a potent OATP inhibitor, significantly reduced the radioactivity excreted into the bile (by 44%) by inhibiting both uptake (by 45%) and subsequent canalicular efflux (by 62%). (15R)-(11)C-TIC is an in vitro substrate of OATP1B1 and OATP1B3, and clinically relevant concentrations of rifampicin inhibited uptake by OATP1B1 and OATP1B3. These results demonstrated that in humans, (15R)-(11)C-TIC-associated radioactivity is excreted into the bile by organic anion transport systems. CONCLUSION: We demonstrated that PET image analysis with (15R)-(11)C-TIC-Me is useful for investigating variations in OATP function in the human hepatobiliary transport system.


Subject(s)
Biliary Tract/metabolism , Bridged Bicyclo Compounds/pharmacokinetics , Liver/metabolism , Pentanoic Acids/pharmacokinetics , Positron-Emission Tomography , Abdomen , Adult , Bile Canaliculi/drug effects , Bile Canaliculi/metabolism , Biliary Tract/drug effects , Biological Transport/drug effects , Bridged Bicyclo Compounds/blood , Cells, Cultured , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/drug effects , Male , Metabolic Clearance Rate/drug effects , Organic Anion Transporters/metabolism , Pentanoic Acids/blood , Rifampin/pharmacology , Time Factors
18.
Nucl Med Biol ; 39(6): 847-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22421430

ABSTRACT

INTRODUCTION: Telmisartan, a nonpeptide angiotensin II AT1 receptor antagonist used as an antihypertensive drug, is specifically taken up by the liver through the OATP1B3. PET imaging with [(11)C]telmisartan is expected to provide information about the whole body pharmacokinetics of telmisartan as well as its transport property by OATP1B3. The purpose of the study was to determine the biodistribution and radiation dosimetry of [(11)C]telmisartan in humans. METHODS: Biodistribution of [(11)C]telmisartan was measured in three rats and six healthy male human volunteers. In the rat study, a dynamic emission scan was performed for 90 min. In the human study, dynamic whole-body PET images were acquired after intravenous injection of [(11)C]telmisartan. ROIs were defined for source organs on the PET images to measure time-course of [(11)C]telmisartan uptake as percentage injected dose and the number of disintegration for each organ. Radiation dosimetry was calculated with OLINDA/EXM. RESULTS: In the rat study, most radioactivity was rapidly taken up by the liver and part of it was excreted into the biliary tract and intestine. Extrapolating from the rat data, the effective dose for the adult human being was estimated to be 3.65±0.01 microSv/MBq (n=3). In the human study, most of the tracer was taken up by the liver as well, although not as rapidly as in the rat. The activity in the gall bladder and intestine increased gradually. The effective dose for the adult human being was 4.24±0.09 microSv/MBq (n=6). CONCLUSIONS: [(11)C]Telmisartan is a safe PET tracer with a dosimetry profile comparable to other common (11)C PET tracers.


Subject(s)
Benzimidazoles/pharmacokinetics , Benzoates/pharmacokinetics , Liver/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Adult , Animals , Benzimidazoles/adverse effects , Benzimidazoles/metabolism , Benzoates/adverse effects , Benzoates/metabolism , Biomarkers/metabolism , Carbon Radioisotopes , Female , Humans , Liver/diagnostic imaging , Male , Positron-Emission Tomography , Radiometry , Rats , Safety , Solute Carrier Organic Anion Transporter Family Member 1B3 , Species Specificity , Telmisartan , Young Adult
19.
Bioorg Med Chem Lett ; 22(1): 115-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22153940

ABSTRACT

The synthesis and in vivo evaluation of (11)C -labeled uric acid ([(11)C]1), a potential imaging agent for the diagnosis of urate-related life-style diseases, was performed using positron emission tomography (PET) image analysis. First, the synthesis of [(11)C]1 was achieved by reacting 5,6-diaminouracil (2) with (11)C-labeled phosgene ([(11)C]COCl(2)). The radiochemical yield of [(11)C]1 was 37±7% (decay-corrected based on [(11)C]COCl(2)) with specific radioactivities of 96-152GBq/µmol at the end of synthesis (n=6). The average time of radiosynthesis from the end of bombardment, including formulation, was about 30min with >98% radiochemical purity. Second, the synthetic approach to [(11)C]1 was optimized using 5,6-diaminouracil sulfate (3) with [(11)C]COCl(2) in the presence of 1,8-bis(dimethylamino)naphthalene. [(11)C]1 was synthesized in 36±6% radiochemical yield, 89-142GBq/µmol of specific radioactivities, and 98% radiochemical purity by this method (n=5). This allowed the synthesis of [(11)C]1 to be carried out repeatedly and the radiochemical yield, specific radioactivities, average time of synthesis, and radiochemical purity of [(11)C]1 were similar to those obtained using 2. PET studies in rats showed large differences in the accumulation of radioligand in the limbs under normal and hyperuricemic conditions. Thus, an efficient and convenient automated synthesis of [(11)C]1 has been developed, and preliminary PET evaluation of [(11)C]1 confirmed the increased accumulation of radioactivity in the limbs of a rat model of hyperuricemia.


Subject(s)
Biomarkers/metabolism , Carbon Isotopes/analysis , Gout/diagnosis , Gout/pathology , Phosgene , Positron-Emission Tomography/methods , Uric Acid , Animals , Automation , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Hyperuricemia/diagnosis , Image Processing, Computer-Assisted , Models, Chemical , Rats , Temperature
20.
J Nucl Med ; 52(12): 1964-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22072705

ABSTRACT

UNLABELLED: Aromatase is a rate-limiting enzyme for estrogen biosynthesis and has been implicated in pathophysiological states of various diseases via estrogen production. This enzyme is known to be widely distributed in extragonadal and gonadal tissues including the stomach. In contrast to circulating estrogen, the functional role of gastric aromatase/estrogen has not been elucidated in detail, because there is no efficient methodology to investigate spatiotemporal changes of gastric aromatase/estrogen in vivo. Recently, (S)-(11)C-6-[(4-chlorophenyl)(1H-1,2,4-triazole-1-yl)methyl]-1-methyl-1H-benzotriazole ((11)C-labeled vorozole), based on a potent nonsteroidal aromatase inhibitor, has been developed as a tracer to investigate aromatase distribution in living animals and humans using a noninvasive PET technique. In the present study, we investigated gastric aromatase expression by means of PET with (11)C-vorozole. METHODS: After bolus injection of (11)C-vorozole into the tail vein, emission scans were obtained for 90 min on male and female rats under isoflurane anesthesia. Displacement studies with unlabeled vorozole and autoradiographic analysis were conducted for demonstration of specific binding. Immunohistochemistry was performed to confirm aromatase expression. RESULTS: PET scans revealed that (11)C-vorozole highly accumulated in the stomach and adrenal glands. Displacement studies and autoradiography demonstrated that aromatase was expressed in the stomach but that the accumulation of (11)C-vorozole in the adrenal glands might be through nonspecific binding. Immunohistochemical analysis revealed that aromatase is expressed in gastric parietal cells but not in adrenal glands. Moreover, the accumulation of (11)C-vorozole in the stomach was significantly increased in fatigued rats. CONCLUSION: These results suggest that the (11)C-vorozole PET technique is a useful tool for evaluation of gastric aromatase dynamics in vivo, which may provide important information for understanding the molecular mechanisms of gastric aromatase/estrogen-related pathophysiological processes and for the development of new drugs.


Subject(s)
Aromatase/metabolism , Parietal Cells, Gastric/diagnostic imaging , Parietal Cells, Gastric/enzymology , Positron-Emission Tomography/methods , Triazoles , Animals , Autoradiography , Carbon Radioisotopes , Fatigue/diagnostic imaging , Fatigue/metabolism , Female , Immunohistochemistry , Male , Parietal Cells, Gastric/metabolism , Rats , Rats, Sprague-Dawley , Whole Body Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...