Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Bioorg Med Chem Lett ; 30(6): 126998, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32014383

ABSTRACT

[Thiocarbonyl-11C]disulfiram ([11C]DSF) was synthesized via iodine oxidation of [11C]diethylcarbamodithioic acid ([11C]DETC), which was prepared from [11C]carbon disulfide and diethylamine. The decay-corrected isolated radiochemical yield (RCY) of [11C]DSF was greatly affected by the addition of unlabeled carbon disulfide. In the presence of carbon disulfide, the RCY was increased up to 22% with low molar activity (Am, 0.27 GBq/µmol). On the other hand, [11C]DSF was obtained in 0.4% RCY with a high Am value (95 GBq/µmol) in the absence of carbon disulfide. The radiochemical purity of [11C]DSF was always >98%. The first PET study on [11C]DSF was performed in mice. A high uptake of radioactivity was observed in the liver, kidneys, and gallbladder. The uptake level and distribution pattern in mice were not significantly affected by the Am value of the [11C]DSF sample used. In vivo metabolite analysis showed the rapid decomposition of [11C]DSF in mouse plasma.


Subject(s)
Carbon Radioisotopes/chemistry , Disulfiram/chemical synthesis , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemical synthesis , Animals , Carbon Disulfide/chemistry , Coordination Complexes/chemistry , Diethylamines/chemistry , Disulfiram/metabolism , Ditiocarb/chemistry , Gallbladder/metabolism , Iodine/chemistry , Kidney/metabolism , Ligands , Liver/metabolism , Mice , Oxidation-Reduction , Radiopharmaceuticals/metabolism , Tissue Distribution
2.
Bioorg Med Chem Lett ; 27(19): 4521-4524, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28888821

ABSTRACT

The purpose of this study was to synthesize a new positron emission tomography radiotracer, N-(4-tertiarybutylphenyl)-4-(3-chloropyridin-2-yl)tetrahydropyrazine-1(2H)-[11C]carboxamide ([11C]BCTC, [11C]1), and assess its in vivo binding to the transient receptor potential vanilloid subfamily member 1 (TRPV1) receptor in mice. [11C]BCTC was synthesized by reacting the hydrochloride of 4-tertiarybutylaniline (2·HCl) with [11C]phosgene ([11C]COCl2) to give isocyanate [11C]4, followed by reaction with 4-(3-chloropyridin-2-yl)tetrahydropyrazine (3). [11C]BCTC was obtained at a 16-20% radiochemical yield, based on the cyclotron-produced [11C]CO2 (decay-corrected to the end of bombardment), with >98% radiochemical purity and 65-110GBq/µmol specific activity at the end of synthesis. An ex vivo biodistribution study in mice confirmed the specific binding of [11C]BCTC to TRPV1 in the trigeminal nerve, which is a tissue with high TRPV1 expression.


Subject(s)
Pyrazines/pharmacokinetics , Pyridines/pharmacokinetics , TRPV Cation Channels/chemistry , Trigeminal Nerve/chemistry , Animals , Binding Sites/drug effects , Carbon Isotopes , Dose-Response Relationship, Drug , Mice , Mice, Inbred Strains , Molecular Structure , Positron-Emission Tomography , Pyrazines/chemical synthesis , Pyrazines/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Radioactive Tracers , Structure-Activity Relationship , TRPV Cation Channels/biosynthesis , Tissue Distribution
3.
Bioorg Med Chem ; 25(3): 1014-1021, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28049619

ABSTRACT

Brain metabotropic glutamate receptor 2 (mGluR2) has been proposed as a therapeutic target for the treatment of schizophrenia-like symptoms arising from increased glutamate transmission in the forebrain. However, there does not exist a reliable tool for the study of mGluR2 in human neuroimaging. The purpose of this study was to radiosynthesize 1-(cyclopropylmethyl)-4-(4-[11C]methoxyphenyl)piperidin-1-yl-2-oxo-1,2-dihydropyridine-3-carbonitrile ([11C]CMDC) and evaluate its potential as a positron emission tomography (PET) radiotracer for imaging mGluR2 in the rat brain. CMDC, a positive allosteric modulator of mGluR2, showed potent functional activity (EC50: 98nM) for human mGluR2 in vitro. [11C]CMDC was synthesized by O-[11C]methylation of 1-(cyclopropylmethyl)-4-(4-hydroxyphenyl)piperidin-1-yl-2-oxo-1,2-dihydropyridine-3-carbonitrile (1) with [11C]methyl iodide. [11C]CMDC (2.2±0.9GBq; n=20) was obtained from [11C]CO2 of 14.0-17.8GBq with >98% radiochemical purity and 86-150GBq/µmol specific activity at the end of synthesis. In vitro autoradiography indicated that [11C]CMDC binding was expressed (>50% of total binding) in mGluR2-rich brain regions including the cerebral cortex, striatum and hippocampus. However, small-animal PET showed low in vivo specific binding of [11C]CMDC in the rat brain. While [11C]CMDC has limited potential as a PET tracer for brain mGluR2, it can be used to develop new radiotracers with improved behaviors.


Subject(s)
Dihydropyridines/chemistry , Piperidines/chemistry , Positron-Emission Tomography , Receptors, Metabotropic Glutamate/analysis , Animals , Dihydropyridines/chemical synthesis , Dihydropyridines/pharmacokinetics , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Molecular Structure , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Radioactive Tracers , Rats , Rats, Sprague-Dawley , Tissue Distribution
4.
Nucl Med Biol ; 45: 1-7, 2017 02.
Article in English | MEDLINE | ID: mdl-27835825

ABSTRACT

INTRODUCTION: A positron emission tomography (PET) probe with ultra-high specific radioactivity (SA) enables measuring high receptor specific binding in brain regions by avoiding mass effect of the PET probe itself. It has been reported that PET probe with ultra-high SA can detect small change caused by endogenous or exogenous ligand. Recently, Kealey et al. developed [11C]BU99008, a more potent PET probe for I2-imidazoline receptors (I2Rs) imaging, with a conventional SA (mean 76GBq/µmol) showed higher specific binding in the brain. Here, to detect small change of specific binding for I2Rs caused by endogenous or exogenous ligand in an extremely small region, such as hypothalamus in the brain, we synthesized and evaluated [11C]BU99008 with ultra-high SA as a useful PET probe for small-animal PET imaging of I2Rs. METHODS: [11C]BU99008 was prepared by [11C]methylation of N-desmethyl precursor with [11C]methyl iodide. Biodistribution, metabolite analysis, and brain PET studies were conducted in rats. RESULTS: [11C]BU99008 with ultra-high SA in the range of 5400-16,600GBq/µmol were successfully synthesized (n=7), and had appropriate radioactivity for in vivo study. In the biodistribution study, the mean radioactivity levels in all investigated tissues except for the kidney did not show significant difference between [11C]BU99008 with ultra-high SA and that with conventional SA. In the metabolite analysis, the percentage of unchanged [11C]BU99008 at 30min after the injection of probes with ultra-high and conventional SA was similar in rat brain and plasma. In the PET study of rats' brain, radioactivity level (AUC30-60 min) in the hypothalamus of rats injected with [11C]BU99008 with ultra-high SA (64 [SUV ∙ min]) was significantly higher than that observed for that with conventional SA (50 [SUV ∙ min]). The specific binding of [11C]BU99008 with ultra-high SA (86% of total binding) for I2R was higher than that of conventional SA (76% of total binding). CONCLUSION: A PET study using [11C]BU99008 with ultra-high SA would thus contribute to the detection of small changes in or small regions with I2R expression and hence may be useful in elucidating new functions of I2R.


Subject(s)
Carbon Radioisotopes , Hypothalamus/diagnostic imaging , Hypothalamus/metabolism , Imidazoles , Imidazoline Receptors/metabolism , Indoles , Positron-Emission Tomography/methods , Animals , Imidazoles/blood , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Indoles/blood , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacokinetics , Radiochemistry , Rats , Tissue Distribution
5.
Theranostics ; 6(8): 1145-59, 2016.
Article in English | MEDLINE | ID: mdl-27279908

ABSTRACT

Monoacylglycerol lipase (MAGL) is a 33 kDa member of the serine hydrolase superfamily that preferentially degrades 2-arachidonoylglycerol (2-AG) to arachidonic acid in the endocannabinoid system. Inhibition of MAGL is not only of interest for probing the cannabinoid pathway but also as a therapeutic and diagnostic target for neuroinflammation. Limited attempts have been made to image MAGL in vivo and a suitable PET ligand for this target has yet to be identified and is urgently sought to guide small molecule drug development in this pathway. Herein we synthesized and evaluated the physiochemical properties of an array of eleven sulfonamido-based carbamates and ureas with a series of terminal aryl moieties, linkers and leaving groups. The most potent compounds were a novel MAGL inhibitor, N-((1-(1H-1,2,4-triazole-1-carbonyl)piperidin-4-yl) methyl)-4-chlorobenzenesulfonamide (TZPU; IC50 = 35.9 nM), and the known inhibitor 1,1,1,3,3,3-hexafluoropropan-2-yl 4-(((4-chlorophenyl)sulfonamido) methyl)piperidine-1-carboxylate (SAR127303; IC50 = 39.3 nM), which were also shown to be selective for MAGL over fatty acid amide hydrolase (FAAH), and cannabinoid receptors (CB1 & CB2). Both of these compounds were radiolabeled with carbon-11 via [(11)C]COCl2, followed by comprehensive ex vivo biodistribution and in vivo PET imaging studies in normal rats to determine their brain permeability, specificity, clearance and metabolism. Whereas TZPU did not show adequate specificity to warrant further evaluation, [(11)C]SAR127303 was advanced for preliminary PET neuroimaging studies in nonhuman primate. The tracer showed good brain permeability (ca. 1 SUV) and heterogeneous regional brain distribution which is consistent with the distribution of MAGL.


Subject(s)
Carbamates/metabolism , Carbon Radioisotopes/metabolism , Monoacylglycerol Lipases/analysis , Neuroimaging/methods , Positron-Emission Tomography/methods , Sulfonamides/metabolism , Urea/metabolism , Animals , Carbamates/chemical synthesis , Carbamates/pharmacokinetics , Carbon Radioisotopes/pharmacokinetics , Rats , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Urea/chemical synthesis , Urea/pharmacokinetics
6.
J Med Chem ; 59(12): 5847-56, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27232368

ABSTRACT

Multidrug resistance-associated protein 4 (MRP4) and organic anion transporter 3 (OAT3) mediate the efflux of organic anions from the brain and heart. In this study, we have developed a probe for estimating the activity of these transporters in these tissues using positron emission tomography. Several (11)C-labeled hippuric acid ester derivatives were screened with the expectation that they would be hydrolyzed in situ to form the corresponding (11)C-labeled organic acids in target tissues. Among the compounds screened, benzyl [(11)C]hippurate showed favorable hydrolysis rates and uptake properties in the target tissues of mice. Subsequent evaluation using transporter knockout mice revealed that radioactivity was retained in the brain and heart of Oat3(-/-) and Mrp4(-/-) mice, respectively, compared with that of control mice after the intravenous administration of benzyl [(11)C]hippurate. Benzyl [(11)C]hippurate could therefore be used as a probe for estimating the activities of OAT3 and MRP4 in mouse brain and heart, respectively.


Subject(s)
Brain/metabolism , Hippurates/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Administration, Intravenous , Animals , Carbon Radioisotopes , Heart , Hippurates/administration & dosage , Hippurates/chemical synthesis , Hippurates/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Proteins/deficiency , Organic Anion Transporters, Sodium-Independent/deficiency , Positron-Emission Tomography , Tissue Distribution
7.
Nucl Med Biol ; 43(7): 445-53, 2016 07.
Article in English | MEDLINE | ID: mdl-27183465

ABSTRACT

INTRODUCTION: [(18)F]FEDAC ([(18)F]1) has potent binding affinity and selectivity for translocator protein (18kDa, TSPO), and has been used to noninvasively visualize neuroinflammation, lung inflammation, acute liver damage, nonalcoholic fatty liver disease, and liver fibrosis. We had previously synthesized [(18)F]1 in two steps: (i) preparation of [(18)F]fluoroethyl bromide and (ii) coupling of [(18)F]fluoroethyl bromide with the appropriate precursor (2) for labeling. In this study, to clinically utilize [(18)F]1 as a PET radiopharmaceutical and to transfer the production technique of [(18)F]1 to other PET centers, we simplified its preparation by using a direct, one-step, tosyloxy-for-fluorine substitution. We also performed an acute toxicity study as a major non-clinical safety test, and determined radiometabolites using human liver microsomes. METHODS: [(18)F]1 was prepared via direct (18)F-fluorination by heating the corresponding tosylated derivative (3) with [(18)F]fluoride as its Kryptofix 222 complex in dimethyl sulfoxide at 110°C for 15min, following by HPLC purification. Non-clinical safety tests were performed for the extended single-dose toxicity study in rats, and for the in vitro metabolite analysis with human liver microsomal incubation. RESULTS: High quality batches of [(18)F]1, compatible with clinical applications, were obtained. At the end of irradiation, the decay-corrected radiochemical yield of [(18)F]1 using 1 and 5mg of precursor based on [(18)F]fluoride was 18.5±7.9% (n=10) and 52.0±5.8% (n=3), respectively. A single-dose of [(18)F]1 did not show toxicological effects for 14 days after the injection in male and female rats. In human liver microsomal incubations, [(18)F]1 was easily metabolized to [(18)F]desbenzyl-FEDAC ([(18)F]10) by CYPs (4.2% of parent compound left 60min after incubation). CONCLUSION: We successfully synthesized clinical grade batches of [(18)F]1 and verified the absence of innocuity of this radiotracer. [(18)F]1 will be used to first-in-human studies in our facility.


Subject(s)
Acetamides/metabolism , Acetamides/toxicity , Carrier Proteins/metabolism , Purines/metabolism , Purines/toxicity , Receptors, GABA-A/metabolism , Safety , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Humans , Microsomes, Liver/metabolism , Positron-Emission Tomography , Purines/chemical synthesis , Purines/chemistry , Radiochemistry , Rats , Rats, Sprague-Dawley
8.
J Med Chem ; 59(8): 3980-90, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27015128

ABSTRACT

We found out 3-[5-(pyridin-2-yl)-2H-tetrazol-2-yl]benzonitrile analogues as the candidate for positron emission tomography (PET) imaging agents of metabotropic glutamate receptor subtype 5 (mGluR5). Among these compounds, 3-methyl-5-(5-(pyridin-2-yl)-2H-tetrazol-2-yl)benzonitrile (10) exhibited high binding affinity (Ki = 9.4 nM) and moderate lipophilicity (cLogD, 2.4). Subsequently, [(11)C]10 was radiosynthesized at 25 ± 14% radiochemical yield (n = 11) via C-[(11)C]methylation of the arylstannyl precursor 15 with [(11)C]methyl iodide. In vitro autoradiography and PET assessments using [(11)C]10 showed high specific binding in the striatum and hippocampus, two brain regions enriched with mGluR5. Moreover, test-retest PET studies with [(11)C]10 indicated high reliability to quantify mGluR5 density, such as the intraclass correlation coefficient (0.90) and Pearson r (0.91) in the striatum of rat brain. We demonstrated that [(11)C]10 is a useful PET ligand for imaging and quantitative analysis of mGluR5. Furthermore, [(11)C]10 might be modified using its skeleton as a lead compound.


Subject(s)
Pyridines/chemical synthesis , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Tetrazoles/chemical synthesis , Tetrazoles/pharmacology , Animals , Autoradiography , Male , Mice , Positron-Emission Tomography , Radioligand Assay , Rats
9.
Bioorg Med Chem ; 24(4): 627-34, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26740152

ABSTRACT

To visualize fatty acid amide hydrolase (FAAH) in brain in vivo, we developed a novel positron emission tomography (PET) ligand N-(3,4-dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[(11)C]methylphenyl)thiazol-2-yl]-1-carboxamide ([(11)C]DFMC, [(11)C]1). DFMC (1) was shown to have high binding affinity (IC50: 6.1nM) for FAAH. [(11)C]1 was synthesized by C-(11)C coupling reaction of arylboronic ester 2 with [(11)C]methyl iodide in the presence of Pd catalyst. At the end of synthesis, [(11)C]1 was obtained with a radiochemical yield of 20±10% (based on [(11)C]CO2, decay-corrected, n=5) and specific activity of 48-166GBq/µmol. After the injection of [(11)C]1 in mice, high uptake of radioactivity (>2% ID/g) was distributed in the lung, liver, kidney, and brain, organs with high FAAH expression. PET images of rat brains for [(11)C]1 revealed high uptakes in the cerebellar nucleus (SUV=2.4) and frontal cortex (SUV=2.0), two known brain regions with high FAAH expression. Pretreatment with the FAAH-selective inhibitor URB597 reduced the brain uptake. Higher than 90% of the total radioactivity in the rat brain was irreversible at 30min after the radioligand injection. The present results indicate that [(11)C]1 is a promising PET ligand for imaging of FAAH in living brain.


Subject(s)
Amidohydrolases/metabolism , Piperazines/pharmacology , Positron-Emission Tomography/methods , Radiopharmaceuticals/analysis , Thiazoles/pharmacology , Amidohydrolases/antagonists & inhibitors , Animals , Benzamides/pharmacology , Brain/drug effects , Brain/metabolism , Carbamates/pharmacology , Carbon Radioisotopes , Humans , Ligands , Mice , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Rats , Thiazoles/chemical synthesis , Thiazoles/chemistry , Tissue Distribution
10.
J Neurosci ; 36(2): 375-84, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758830

ABSTRACT

Parkinson's disease (PD) is a prevalent degenerative disorder affecting the CNS that is primarily characterized by resting tremor and movement deficits. Group I metabotropic glutamate receptor subtypes 1 and 5 (mGluR1 and mGluR5, respectively) are important targets for investigation in several CNS disorders. In the present study, we investigated the in vivo roles of mGluR1 and mGluR5 in chronic PD pathology by performing longitudinal positron emission tomography (PET) imaging in A53T transgenic (A53T-Tg) rats expressing an abnormal human α-synuclein (ASN) gene. A53T-Tg rats showed a dramatic decline in general motor activities with age, along with abnormal ASN aggregation and striatal neuron degeneration. In longitudinal PET imaging, striatal nondisplaceable binding potential (BPND) values for [(11)C]ITDM (N-[4-[6-(isopropylamino) pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methyl-4-[(11)C]methylbenzamide), a selective PET ligand for mGluR1, temporarily increased before PD symptom onset and dramatically decreased afterward with age. However, striatal BPND values for (E)-[(11)C]ABP688 [3-(6-methylpyridin-2-ylethynyl)-cyclohex-2-enone-(E)-O-[(11)C]methyloxime], a specific PET ligand for mGluR5, remained constant during experimental terms. The dynamic changes in striatal mGluR1 BPND values also showed a high correlation in pathological decreases in general motor activities. Furthermore, declines in mGluR1 BPND values were correlated with decreases in BPND values for [(18)F]FE-PE2I [(E)-N-(3-iodoprop-2E-enyl)-2ß-carbo-[(18)F]fluoroethoxy-3ß-(4-methylphenyl) nortropane], a specific PET ligand for the dopamine transporter, a biomarker for dopaminergic neurons. In conclusion, our results have demonstrated for the first time that dynamic changes occur in mGluR1, but not mGluR5, that accompany pathological progression in a PD animal model. SIGNIFICANCE STATEMENT: Synaptic signaling by glutamate, the principal excitatory neurotransmitter in the brain, is modulated by group I metabotropic glutamate receptors, including the mGluR1 and mGluR5 subtypes. In the brain, mGluR1 and mGluR5 have distinct functional roles and regional distributions. Their roles in brain pathology, however, are not well characterized. Using longitudinal PET imaging in a chronic rat model of PD, we demonstrated that expression of mGluR1, but not mGluR5, dynamically changed in the striatum accompanying pathological PD progression. These findings imply that monitoring mGluR1 in vivo may provide beneficial information to further understand central nervous system disorders.


Subject(s)
Corpus Striatum/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptors, Metabotropic Glutamate/metabolism , alpha-Synuclein/genetics , Alanine/genetics , Animals , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Disease Models, Animal , Disease Progression , Excitatory Amino Acid Agents/pharmacokinetics , Exploratory Behavior/physiology , Female , Humans , Motor Activity/genetics , Oximes , Protein Binding/drug effects , Pyridines , Radioisotopes/pharmacokinetics , Radioisotopes/pharmacology , Radionuclide Imaging , Rats , Rats, Transgenic , Threonine/genetics , Time Factors
11.
Bioorg Med Chem Lett ; 26(2): 370-374, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26707390

ABSTRACT

ADX88178 (1) has been recently developed as a potent positive allosteric modulator for metabotropic glutamate receptor 4 (mGluR4). The aim of this study was to develop [(11)C]1 as a novel positron emission tomography ligand and to evaluate its binding ability for mGluR4. Using stannyl precursor 3, [(11)C]1 was efficiently synthesized by introducing an [(11)C]methyl group into a pyrimidine ring via C-(11)C coupling and deprotection reactions, in 16±6% radiochemical yield (n=10). At the end of synthesis, 0.54-1.10GBq of [(11)C]1 was acquired with >98% radiochemical purity and 90-120GBq/µmol of specific activity. In vitro autoradiography and ex vivo biodistribution study in rat brains showed specific binding of [(11)C]1 in the cerebellum, striatum, thalamus, cerebral cortex, and medulla oblongata, which showed dose-dependent decreases by administration with multi-dose of unlabeled 1.


Subject(s)
Brain/metabolism , Positron-Emission Tomography/methods , Pyrimidines/metabolism , Radiopharmaceuticals/metabolism , Receptors, Metabotropic Glutamate/metabolism , Thiazoles/metabolism , Animals , Autoradiography , Brain/diagnostic imaging , Male , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/analysis , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Tissue Distribution
12.
ACS Chem Neurosci ; 7(1): 109-18, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26505525

ABSTRACT

Fatty acid amide hydrolase (FAAH) is one of the principle enzymes for metabolizing endogenous cannabinoid neurotransmitters such as anandamide, and thus regulates endocannabinoid (eCB) signaling. Selective pharmacological blockade of FAAH has emerged as a potential therapy to discern the endogenous functions of anandamide-mediated eCB pathways in anxiety, pain, and addiction. Quantification of FAAH in the living brain by positron emission tomography (PET) would help our understanding of the endocannabinoid system in these conditions. While most FAAH radiotracers operate by an irreversible ("suicide") binding mechanism, a FAAH tracer with reversibility would facilitate quantitative analysis. We have identified and radiolabeled a reversible FAAH inhibitor, 7-(2-[(11)C]methoxyphenyl)-1-(5-(pyridin-2-yl)oxazol-2-yl)heptan-1-one ([(11)C]MPPO) in 13% radiochemical yield (nondecay corrected) with >99% radiochemical purity and 2 Ci/µmol (74 GBq/µmol) specific activity. The tracer showed moderate brain uptake (0.8 SUV) with heterogeneous brain distribution. However, blocking studies with a potent FAAH inhibitor URB597 demonstrated a low to modest specificity to the target. Measurement of lipophilicity, metabolite, and efflux pathway analysis were also performed to study the pharmacokinetic profile of [(11)C]MPPO. In all, we reported an efficient radiolabeling and preliminary evaluation of the first-in-class FAAH inhibitor [(11)C]MPPO with α-ketoheterocyclic scaffold.


Subject(s)
Amidohydrolases/pharmacokinetics , Carbon Radioisotopes/pharmacokinetics , Positron-Emission Tomography , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Amidohydrolases/chemical synthesis , Amidohydrolases/chemistry , Animals , Benzamides/pharmacology , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Transgenic , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley , Tissue Distribution/drug effects
13.
Sci Rep ; 5: 17327, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26612465

ABSTRACT

Hepatic fibrosis is the wound healing response to chronic hepatic injury caused by various factors. In this study, we aimed to evaluate the utility of translocator protein (18 kDa) (TSPO) as a molecular imaging biomarker for monitoring the progression of hepatic fibrosis to cirrhosis. Model rats were induced by carbon tetrachloride (CCl4), and liver fibrosis was assessed. Positron emission tomography (PET) with N-benzyl-N-methyl-2-[7,8-dihydro-7-(2-[(18)F]fluoroethyl)-8-oxo-2-phenyl-9H-purin-9-yl]-acetamide ([(18)F]FEDAC), a radioprobe specific for TSPO, was used for noninvasive visualisation in vivo. PET scanning, immunohistochemical staining, ex vivo autoradiography, and quantitative reverse-transcription polymerase chain reaction were performed to elucidate the relationships among radioactivity uptake, TSPO levels, and cellular sources enriching TSPO expression in damaged livers. PET showed that uptake of radioactivity in livers increased significantly after 2, 4, 6, and 8 weeks of CCl4 treatment. Immunohistochemistry demonstrated that TSPO was mainly expressed in macrophages and hepatic stellate cells (HSCs). TSPO-expressing macrophages and HSCs increased with the progression of liver fibrosis. Interestingly, the distribution of radioactivity from [(18)F]FEDAC was well correlated with TSPO expression, and TSPO mRNA levels increased with the severity of liver damage. TSPO was a useful molecular imaging biomarker and could be used to track the progression of hepatic fibrosis to cirrhosis with PET.


Subject(s)
Carrier Proteins/metabolism , Liver Cirrhosis/diagnosis , Liver Cirrhosis/genetics , Molecular Imaging/methods , Positron-Emission Tomography/methods , RNA, Messenger/genetics , Receptors, GABA-A/metabolism , Acetamides/metabolism , Acetamides/pharmacokinetics , Animals , Autoradiography , Biomarkers/metabolism , Carbon Tetrachloride , Carrier Proteins/genetics , Disease Progression , Gene Expression , Immunohistochemistry , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Molecular Weight , Purines/metabolism , Purines/pharmacokinetics , RNA, Messenger/metabolism , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Severity of Illness Index , Tissue Distribution
14.
Sci Rep ; 5: 16417, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26552594

ABSTRACT

1-Methyl-tryptophan (1MTrp) is known as a specific inhibitor targeting the immune-checkpoint protein indoleamine-2,3-dioxygenase, in two stereoisomers of levorotary (L) and dextrorotary (D). A long-standing debate exists in immunology and oncology: which stereoisomer has the potential of antitumor immunotherapy. Herein, we developed two novel radioprobes, 1-N-(11)C-methyl-L- and -D-tryptophan ((11)C-L-1MTrp and (11)C-D-1MTrp), without modifying the chemical structures of the two isomers, and investigated their utility for pharmacokinetic imaging of the whole body. (11)C-L-1MTrp and (11)C-D-1MTrp were synthesized rapidly with radiochemical yields of 47 ± 6.3% (decay-corrected, based on (11)C-CO2), a radiochemical purity of >98%, specific activity of 47-130 GBq/µmol, and high enantiomeric purity. PET/CT imaging in rats revealed that for (11)C-L-1MTrp, the highest distribution of radioactivity was observed in the pancreas, while for (11)C-D-1MTrp, it was observed in the kidney. Ex vivo biodistribution confirmed the PET/CT results, indicating the differences in pharmacokinetics between the two isomers. Both (11)C-L-1MTrp and (11)C-D-1MTrp are therefore useful PET probes for delineating the distribution and action of the checkpoint inhibitor 1MTrp in vivo. This study represents the first step toward using whole-body and real-time insight to disentangle the antitumor potential of the two stereoisomers of 1MTrp, and it can facilitate the development of 1MTrp immunotherapy.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Tryptophan/analogs & derivatives , Animals , Antineoplastic Agents/pharmacokinetics , Carbon Radioisotopes/chemistry , Drug Stability , Immunologic Factors/pharmacokinetics , Male , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Rats , Tissue Distribution , Tryptophan/chemistry , Tryptophan/pharmacokinetics , X-Ray Microtomography
15.
Nucl Med Biol ; 42(12): 905-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26420569

ABSTRACT

INTRODUCTION: [(11)C]PBB3 is a clinically used positron emission tomography (PET) probe for in vivo imaging of tau pathology in the brain. Our previous study showed that [(11)C]PBB3 was rapidly decomposed to a polar radiometabolite in the plasma of mice. For the pharmacokinetic evaluation of [(11)C]PBB3 it is important to elucidate the characteristics of radiometabolites. In this study, we identified the chemical structure of a major radiometabolite of [(11)C]PBB3 and proposed the metabolic pathway of [(11)C]PBB3. METHODS: Carrier-added [(11)C]PBB3 was injected into a mouse for in vivo metabolite analysis. The chemical structure of a major radiometabolite was identified using LC-MS. Mouse and human liver microsomes and liver S9 samples were incubated with [(11)C]PBB3 in vitro. In silico prediction software was used to assist in the determination of the metabolite and metabolic pathway of [(11)C]PBB3. RESULTS: In vivo analysis showed that the molecular weight of a major radiometabolite of [(11)C]PBB3, which was called as [(11)C]M2, was m/z 390 [M+H(+)]. In vitro analysis assisted by in silico prediction showed that [(11)C]M2, which was not generated by cytochrome P450 enzymes (CYPs), was generated by sulfated conjugation mediated by a sulfotransferase. CONCLUSION: The major radiometabolite, [(11)C]M2, was identified as a sulfated conjugate of [(11)C]PBB3. [(11)C]PBB3 was metabolized mainly by a sulfotransferase and subsidiarily by CYPs.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/metabolism , Benzothiazoles/chemistry , Benzothiazoles/metabolism , Chromatography, Liquid/methods , Mass Spectrometry/methods , Microsomes, Liver/metabolism , Radiopharmaceuticals/metabolism , Animals , Computer Simulation , Cytochrome P-450 Enzyme System/metabolism , Humans , Metabolomics , Mice , Radiochemistry
16.
Biol Pharm Bull ; 38(11): 1722-31, 2015.
Article in English | MEDLINE | ID: mdl-26277991

ABSTRACT

α5ß1 Integrin, a fibronectin receptor, is becoming a pertinent therapeutic target and a promising prognostic biomarker for cancer patients. The aim of this study was to functionalize an α5ß1-specific fibronectin-mimetic peptide sequence KSSPHSRN(SG)5RGDSP (called PR_b) as a positron emission tomography (PET) probe. PR_b was modified by addition of a ß-alanine residue, conjugated with 2-S-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid (p-SCN-Bn-NOTA), and radiolabeled with (18)F based on the chelation of (18)F-aluminum fluoride. A control probe was produced by glycine to alanine substitution in the RGD motif of PR_b. Cell binding and blocking assays, autoradiographic evaluation of tissue binding and blocking, dynamic PET scans, and a biodistribution study were conducted using cell lines and murine tumor models with determined expression levels of α5ß1 and other related integrins. (18)F-PR_b was produced with a labeling yield of 22.3±1.9% based on (18)F-F(-), a radiochemical purity of >99%, and a specific activity of 30-70 GBq/µmol; it exhibited α5ß1-binding activity and specificity in vitro, ex vivo, and in vivo, and had a rapid blood clearance and a predominant renal excretion pathway. In vivo α5ß1-positive tumors could be clearly visualized by (18)F-PR_b PET imaging. Both imaging and biodistribution studies suggested higher uptake of (18)F-PR_b in α5ß1-positive tumors than in α5ß1-negative tumors and higher α5ß1-positive tumor uptake of (18)F-PR_b than the control probe. In contrast, there was no significant difference seen in the contralateral muscle uptake. A PET radioprobe, (18)F-PR_b, was developed de novo and potentially can be used for noninvasive detection of α5ß1 expression in tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Fibronectins/metabolism , Integrin alpha5beta1/metabolism , Molecular Imaging/methods , Molecular Probes , Neoplasms/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Humans , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Probes/chemistry , Neoplasms/diagnosis , Positron-Emission Tomography/methods , Rats
17.
Theranostics ; 5(9): 961-9, 2015.
Article in English | MEDLINE | ID: mdl-26155312

ABSTRACT

We evaluated the efficacy of 2-[5-(4-[(18)F]fluoroethoxy-2-oxo-1,3-benzoxazol-3(2H)-yl)-N-methyl-N-phenylacetamide] ([(18)F]FEBMP) for positron emission tomography (PET) imaging of translocator protein (18 kDa, TSPO). Dissection was used to determine the distribution of [(18)F]FEBMP in mice, while small-animal PET and metabolite analysis were used for a rat model of focal cerebral ischemia. [(18)F]FEBMP showed high radioactivity uptake in mouse peripheral organs enriched with TSPO, and relatively high initial brain uptake (2.67 ± 0.12% ID/g). PET imaging revealed an increased accumulation of radioactivity in the infarcted striatum, with a maximum ratio of 3.20 ± 0.12, compared to non-injured striatum. Displacement with specific TSPO ligands lowered the accumulation levels in infarcts to those on the contralateral side. This suggests that the increased accumulation reflected TPSO-specific binding of [(18)F]FEBMP in vivo. Using a simplified reference tissue model, the binding potential on the infarcted area was 2.72 ± 0.27. Metabolite analysis in brain tissues showed that 83.2 ± 7.4% and 76.4 ± 2.1% of radioactivity was from intact [(18)F]FEBMP at 30 and 60 min, respectively, and that this ratio was higher than in plasma (8.6 ± 1.9% and 3.9 ± 1.1%, respectively). In vitro autoradiography on postmortem human brains showed that TSPO rs6971 polymorphism did not affect binding sites for [(18)F]FEBMP. These findings suggest that [(18)F]FEBMP is a promising new tool for visualization of neuroinflammation.


Subject(s)
Brain Ischemia/diagnosis , Carrier Proteins/analysis , Fluorine Radioisotopes/administration & dosage , Inflammation/diagnosis , Neuroimaging/methods , Positron-Emission Tomography/methods , Receptors, GABA-A/analysis , Acetanilides/administration & dosage , Acetanilides/pharmacokinetics , Animals , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacokinetics , Brain/diagnostic imaging , Brain/pathology , Brain Ischemia/pathology , Disease Models, Animal , Fluorine Radioisotopes/pharmacokinetics , Humans , Inflammation/pathology , Male , Mice , Radiography , Rats, Sprague-Dawley , Receptors, GABA/analysis
18.
Bioorg Med Chem Lett ; 25(16): 3230-3, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26067173

ABSTRACT

Three compounds 1-3 containing methyl-sufanyl, sufinyl, or sulfonyl groups are strong inhibitors of glycogen synthase kinase 3ß (GSK-3ß), an enzyme associated with Alzheimer's disease. We labeled 1-3 with (11)C for a positron emission tomography (PET) brain imaging study. A novel thiophenol precursor 4 for radiosynthesis was prepared by reacting sulfoxide 2 with trifluoroacetic anhydride. [(11)C]1 was synthesized by reacting 4 with [(11)C]methyl iodide in 52 ± 5% radiochemical yield (n = 5, based on [(11)C]CO2, corrected for decay). Oxidation of [(11)C]1 with Oxone® produced [(11)C]2 and [(11)C]3, respectively. PET with [(11)C]1 and [(11)C]3 showed 2 fold higher brain uptake of radioactivity in a mouse model of cold water stress in which GSK-3ß expression was increased, than in the controls.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Glycogen Synthase Kinase 3/antagonists & inhibitors , Radiopharmaceuticals/chemical synthesis , Animals , Area Under Curve , Brain/diagnostic imaging , Carbon Radioisotopes , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta , Indicators and Reagents , Isotope Labeling , Mice , Oxidation-Reduction , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Stress, Psychological/diagnostic imaging , Stress, Psychological/enzymology , Sulfuric Acids/chemistry
19.
PLoS One ; 10(6): e0130006, 2015.
Article in English | MEDLINE | ID: mdl-26076143

ABSTRACT

Metabotropic glutamate receptor subtype 1 (mGluR1) is a crucial target in the development of new medications to treat central nervous system (CNS) disorders. Recently, we developed N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-4-[11C]methoxy-N-methyl-benzamide ([11C]ITMM) as a useful positron emission tomography (PET) probe for mGluR1 in clinical studies. Here, we aimed to improve visualization and threshold of specific binding for mGluR1 using [11C]ITMM with ultra-high specific activity (SA) of > 3,500 GBq/µmol in rat brains. A two-tissue compartment model indicated large differences between the two SAs in the constants k3 and k4, representing binding ability for mGluR1, while constants K1 and k2 showed no differences. The total distribution volume (VT) values of conventional and ultra-high SA were 9.1 and 11.2 in the thalamus, 7.7 and 9.7 in the striatum, and 6.4 and 8.5 mL/cm3 in the substantia nigra, respectively. The specific binding of [11C]ITMM with ultra-high SA was significantly higher than the conventional SA, especially in the basal ganglia. Parametric PET images scaled with VT of the ultra-high SA clearly identified regional differences in the rat brain. In conclusion, PET studies using [11C]ITMM with ultra-high SA could sufficiently improve visualization and specific binding for mGluR1, which could help further understanding for mGluR1 functions in CNS disorders.


Subject(s)
Benzamides/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, Metabotropic Glutamate/metabolism , Thiazoles/pharmacokinetics , Animals , Image Processing, Computer-Assisted , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
20.
J Med Chem ; 58(3): 1513-23, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25602363

ABSTRACT

Metabotropic glutamate 1 (mGlu1) receptor is found not only in the brain but also in melanomas and breast cancers. mGlu1 is a promising target for molecular imaging-based diagnosis and treatment of melanoma because its overexpression induces melanocyte carcinogenesis. Here we developed three PET tracers: 4-halogeno-N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol- 2-yl]-N-[(11)C]methylbenzamide ([(11)C]4-6), which exhibited high uptake in target tumor and decreased uptake in nontarget brain tissues. In vitro binding assay indicated high to moderate binding affinities of 4-6 (Ki, 22-143 nM) for mGlu1 receptor. In vivo biodistribution studies in mice implanted with B16F10 melanoma cells confirmed high radioactive uptake in tumor and low uptake in blood, skin, and muscles. Inhibition of mGlu1 receptor using an mGlu1-selective ligand led to reduced radioactive uptake in the tumor. [(11)C]6 displayed the highest ratio of uptake between tumor and nontarget tissue and may prove useful as a PET tracer for mGlu1 imaging in melanoma.


Subject(s)
Benzamides , Melanoma/diagnosis , Melanoma/metabolism , Molecular Imaging , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Receptors, Metabotropic Glutamate/analysis , Thiazoles , Animals , Benzamides/chemical synthesis , Benzamides/chemistry , Dose-Response Relationship, Drug , Humans , Male , Melanoma/drug therapy , Mice , Mice, Inbred C57BL , Molecular Structure , Neoplasms, Experimental/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL