Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Pharm Anal ; 10(5): 452-465, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33133729

ABSTRACT

The implication of the receptor for advanced glycation end-products (RAGE) in numerous diseases and neurodegenerative disorders makes it interesting both as a therapeutic target and as an inflammatory biomarker. In the context of investigating RAGE as a biomarker, there is interest in developing radiotracers that will enable quantification of RAGE using positron emission tomography (PET) imaging. We have synthesized potential small molecule radiotracers for both the intracellular ([18F]InRAGER) and extracellular ([18F]RAGER) domains of RAGE. Herein we report preclinical evaluation of both using in vitro (lead panel screens) and in vivo (rodent and nonhuman primate PET imaging) methods. Both radiotracers have high affinity for RAGE and show good brain uptake, but suffer from off-target binding. The source of the off-target PET signal is not attributable to binding to melatonin receptors, but remains unexplained. We have also investigated use of lipopolysaccharide (LPS)-treated mice as a possible animal model with upregulated RAGE for evaluation of new imaging agents. Immunoreactivity of the mouse brain sections revealed increases in RAGE in the male cohorts, but no difference in the female groups. However, it proves challenging to quantify the changes in RAGE due to off-target binding of the radiotracers. Nevertheless, they are appropriate lead scaffolds for future development of 2nd generation RAGE PET radiotracers because of their high affinity for the receptor and good CNS penetration.

2.
ACS Med Chem Lett ; 11(6): 1299-1304, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32551015

ABSTRACT

PD-132301, an inhibitor of sterol O-acyltransferase 1 (SOAT1; also known as acyl-coenzyme A:cholesterol acyltransferase-1, ACAT1), is under clinical investigation for numerous adrenal disorders. Radiolabeled SOAT1 inhibitors could support drug discovery and help diagnose SOAT1-related disorders, such as atherosclerosis. We synthesized two radiolabeled SOAT1 inhibitors, [11C]PD-132301 and fluorine analogue [18F]1. Rat biodistribution studies were conducted with both agents and, as the most selective tracer, [11C]PD-132301 was advanced to preclinical positron emission tomography studies in (atherosclerotic) ApoE-/- mice. The uptake of [11C]PD-132301 in SOAT1-rich tissue warrants further investigation into the compound as an atherosclerosis and adrenal imaging agent.

3.
Bioorg Med Chem Lett ; 30(12): 127186, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32312583

ABSTRACT

The diaryl sulfonylurea MCC950/CRID3 is a potent NLRP3 inhibitor (IC50 = 8 nM) and, in animal models, MCC950 protects against numerous NLRP3-related neurodegenerative disorders. To evaluate the brain uptake and investigate target engagement of MCC950, we synthesised [11C-urea]MCC950 via carrier added [11C]CO2 fixation chemistry (activity yield = 237 MBq; radiochemical purity >99%; molar activity = 7 GBq/µmol; radiochemical yield (decay-corrected from [11C]CO2) = 1.1%; synthesis time from end-of-bombardment = 31 min; radiochemically stable for >1 h). Despite preclinical efficacy in neurodegeneration studies, preclinical positron emission tomography (PET) imaging studies in mouse, rat and rhesus monkey revealed poor brain uptake of low molar activity [11C]MCC950 and rapid washout. In silico prediction tools suggest efflux transporter liabilities for MCC950 at microdoses, and this information should be taken into account when developing next generation NLRP3 inhibitors and/or PET radiotracers.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/pharmacology , Animals , Carbon Radioisotopes , Dose-Response Relationship, Drug , Furans , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , Indenes , Macaca mulatta , Mice , Mice, Inbred C57BL , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides , Sulfones/chemical synthesis , Sulfones/chemistry
4.
Molecules ; 25(6)2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32192089

ABSTRACT

Naloxone (NLX) is a mu receptor antagonist used to treat acute opioid overdoses. Currently approved doses of naloxone to treat opioid overdoses are 4 mg intranasal (IN) and 2 mg intramuscular (IM). However, higher mu receptor occupancy (RO) may be required to treat overdoses due to more potent synthetic opioids such as fentanyl and carfentanil that have entered the illicit drug market recently. To address this need, a higher dose of NLX has been investigated in a 5 mg IM formulation called ZIMHI but, while the effects of intravenous (IV) and IN administration of NLX on the opioid mu receptor occupancy (RO) have been studied, comparatively little is known about RO for IM administration of NLX. The goal of this study was to examine the effect of IM dosing of NLX on mu RO in rhesus macaques using [11C]carfentanil positron emission tomography (PET) imaging. The lowest dose of NLX (0.06 mg/kg) approximated 51% RO. Higher doses of NLX (0.14 mg/kg, 0.28 mg/kg) resulted in higher mu RO of 70% and 75%, respectively. Plasma levels were 4.6 ng/mL, 16.8 ng/mL, and 43.4 ng/mL for the three IM doses, and a significant correlation between percent RO and plasma NLX level was observed (r = 0.80). These results suggest that higher doses of IM NLX result in higher mu RO and could be useful in combating overdoses resulting from potent synthetic opioids.


Subject(s)
Fentanyl/analogs & derivatives , Naloxone/administration & dosage , Naloxone/pharmacology , Receptors, Opioid, mu/metabolism , Animals , Brain/diagnostic imaging , Brain/drug effects , Dose-Response Relationship, Drug , Female , Fentanyl/pharmacology , Injections, Intramuscular , Macaca mulatta , Naloxone/blood , Positron-Emission Tomography
5.
J Med Chem ; 62(21): 9600-9617, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31535859

ABSTRACT

Using structure-guided design, several cell based assays, and microdosed positron emission tomography (PET) imaging, we identified a series of highly potent, selective, and brain-penetrant oxazole-4-carboxamide-based inhibitors of glycogen synthase kinase-3 (GSK-3). An isotopologue of our first-generation lead, [3H]PF-367, demonstrates selective and specific target engagement in vitro, irrespective of the activation state. We discovered substantial ubiquitous GSK-3-specific radioligand binding in Tg2576 Alzheimer's disease (AD), suggesting application for these compounds in AD diagnosis and identified [11C]OCM-44 as our lead GSK-3 radiotracer, with optimized brain uptake by PET imaging in nonhuman primates. GSK-3ß-isozyme selectivity was assessed to reveal OCM-51, the most potent (IC50 = 0.030 nM) and selective (>10-fold GSK-3ß/GSK-3α) GSK-3ß inhibitor known to date. Inhibition of CRMP2T514 and tau phosphorylation, as well as favorable therapeutic window against WNT/ß-catenin signaling activation, was observed in cells.


Subject(s)
Brain/metabolism , Drug Discovery , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Catalytic Domain , Glycogen Synthase Kinase 3 beta/chemistry , HEK293 Cells , Humans , Mice , Models, Molecular , Neuroimaging , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Protein Kinase Inhibitors/metabolism , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
6.
Pharmaceuticals (Basel) ; 11(4)2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30551596

ABSTRACT

Positron emission tomography (PET) imaging of Colony Stimulating Factor 1 Receptor (CSF1R) is a new strategy for quantifying both neuroinflammation and inflammation in the periphery since CSF1R is expressed on microglia and macrophages. AZ683 has high affinity for CSF1R (Ki = 8 nM; IC50 = 6 nM) and >250-fold selectivity over 95 other kinases. In this paper, we report the radiosynthesis of [11C]AZ683 and initial evaluation of its use in CSF1R PET. [11C]AZ683 was synthesized by 11C-methylation of the desmethyl precursor with [11C]MeOTf in 3.0% non-corrected activity yield (based upon [11C]MeOTf), >99% radiochemical purity and high molar activity. Preliminary PET imaging with [11C]AZ683 revealed low brain uptake in rodents and nonhuman primates, suggesting that imaging neuroinflammation could be challenging but that the radiopharmaceutical could still be useful for peripheral imaging of inflammation.

7.
J Med Chem ; 61(4): 1737-1743, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29257860

ABSTRACT

Changes in expression and dysfunctional signaling of TrkA/B/C receptors and oncogenic Trk fusion proteins are found in neurological diseases and cancers. Here, we describe the development of a first 18F-labeled optimized lead suitable for in vivo imaging of Trk, [18F]TRACK, which is radiosynthesized with ease from a nonactivated aryl precursor concurrently combining largely reduced P-gp liability and improved brain kinetics compared to previous leads while displaying high on-target affinity and human kinome selectivity.


Subject(s)
Fluorine Radioisotopes , Positron-Emission Tomography/methods , Receptor, trkA/antagonists & inhibitors , Drug Design , Humans , Neoplasms/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , Protein Kinase Inhibitors
8.
J Med Chem ; 60(16): 6897-6910, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28696690

ABSTRACT

The proto-oncogenes NTRK1/2/3 encode the tropomyosin receptor kinases TrkA/B/C which play pivotal roles in neurobiology and cancer. We describe herein the discovery of [11C]-(R)-3 ([11C]-(R)-IPMICF16), a first-in-class positron emission tomography (PET) TrkB/C-targeting radiolabeled kinase inhibitor lead. Relying on extensive human kinome vetting, we show that (R)-3 is the most potent and most selective TrkB/C inhibitor characterized to date. It is demonstrated that [11C]-(R)-3 readily crosses the blood-brain barrier (BBB) in rodents and selectively binds to TrkB/C receptors in vivo, as evidenced by entrectinib blocking studies. Substantial TrkB/C-specific binding in human brain tissue is observed in vitro, with specific reduction in the hippocampus of Alzheimer's disease (AD) versus healthy brains. We additionally provide preliminary translational data regarding the brain disposition of [11C]-(R)-3 in primates including first-in-human assessment. These results illustrate for the first time the use of a kinome-wide selective radioactive chemical probe for endogenous kinase PET neuroimaging in human.


Subject(s)
Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Radiopharmaceuticals/pharmacology , Receptor, trkB/antagonists & inhibitors , Receptor, trkC/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Alzheimer Disease/diagnostic imaging , Animals , Benzamides/pharmacology , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Dogs , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacokinetics , Indazoles/pharmacology , Macaca mulatta , Madin Darby Canine Kidney Cells , Membrane Glycoproteins/antagonists & inhibitors , Mice , Neuroimaging , Positron-Emission Tomography , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyridazines/chemical synthesis , Pyridazines/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Rats, Sprague-Dawley , Receptor, trkA/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
9.
ACS Chem Neurosci ; 8(7): 1530-1542, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28322043

ABSTRACT

Fluorine-18 labeled phenethylguanidines are currently under development in our laboratory as radiotracers for quantifying regional cardiac sympathetic nerve density using PET imaging techniques. In this study, we report an efficient synthesis of 18F-hydroxyphenethylguanidines consisting of nucleophilic aromatic [18F]fluorination of a protected diaryliodonium salt precursor followed by a single deprotection step to afford the desired radiolabeled compound. This approach has been shown to reliably produce 4-[18F]fluoro-m-hydroxyphenethylguanidine ([18F]4F-MHPG, [18F]1) and its structural isomer 3-[18F]fluoro-p-hydroxyphenethylguanidine ([18F]3F-PHPG, [18F]2) with good radiochemical yields. Preclinical evaluations of [18F]2 in nonhuman primates were performed to compare its imaging properties, metabolism, and myocardial kinetics with those obtained previously with [18F]1. The results of these studies have demonstrated that [18F]2 exhibits imaging properties comparable to those of [18F]1. Myocardial tracer kinetic analysis of each tracer provides quantitative metrics of cardiac sympathetic nerve density. Based on these findings, first-in-human PET studies with [18F]1 and [18F]2 are currently in progress to assess their ability to accurately measure regional cardiac sympathetic denervation in patients with heart disease, with the ultimate goal of selecting a lead compound for further clinical development.


Subject(s)
Guanidines , Heart/innervation , Positron-Emission Tomography , Radiopharmaceuticals , Sympathetic Nervous System/diagnostic imaging , Animals , Drug Evaluation, Preclinical , Guanidines/blood , Guanidines/chemical synthesis , Guanidines/chemistry , Heart/diagnostic imaging , In Vitro Techniques , Isomerism , Kinetics , Macaca mulatta , Male , Molecular Structure , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 359(2): 366-373, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27625351

ABSTRACT

The goal of this study was to evaluate the effects of intranasally administered opioids in rhesus monkeys using the tail-withdrawal assay, and to correlate these effects with measures of receptor occupancy using positron emission tomography (PET) imaging. Initial experiments characterized the antinociceptive effects of intranasal (IN) fentanyl and buprenorphine relative to intramuscular (IM) injection. Fentanyl (0.010-0.032 mg/kg) and buprenorphine (0.1-1.0 mg/kg) produced dose-dependent increases in tail-withdrawal latency that did not differ between routes of delivery. The second experiment compared the ability of IN and intravenous (IV) naloxone (NLX) to block the antinociceptive effects IV fentanyl, and to measure receptor occupancy at equipotent doses of NLX using PET imaging. IN and IV NLX (0.0032-0.032 mg/kg) produced dose-dependent decreases in fentanyl-induced antinociception. Again, there was no difference observed in overall potency between routes. PET imaging showed that IV and IN NLX produced similar decreases in receptor occupancy as measured by [11C]carfentanil blocking, although there was a trend for IV NLX to produce marginally greater occupancy changes. This study validated the first procedures to evaluate the IN effects of opioids in rhesus monkeys.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Nociception/drug effects , Positron-Emission Tomography , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Fentanyl/administration & dosage , Fentanyl/pharmacology , Injections, Intramuscular , Macaca mulatta , Male , Naloxone/administration & dosage , Naloxone/pharmacology
11.
ACS Med Chem Lett ; 7(8): 746-50, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27563397

ABSTRACT

Clarithromycin is a potential treatment for hypersomnia acting through proposed negative allosteric modulation of GABAA receptors. We were interested whether this therapeutic benefit might extend to Parkinson's disease (PD) patients because GABAergic neurotransmission is implicated in postural control. Prior to initiating clinical studies in PD patients, we wished to better understand clarithromycin's mechanism of action. In this work we investigated whether the proposed activity of clarithromycin at the GABAA receptor is associated with the benzodiazepine binding site using in vivo [(11)C]flumazenil positron emission tomography (PET) in primates and ex vivo [(3)H]flumazenil autoradiography in rat brain. While the studies demonstrate that clarithromycin does not change the K d of FMZ, nor does it competitively displace FMZ, there is preliminary evidence from the primate PET imaging studies that clarithromycin delays dissociation and washout of flumazenil from the primate brain in a dose-dependent fashion. These findings would be consistent with the proposed GABAA allosteric modulator function of clarithromycin. While the results are only preliminary, further investigation of the interaction of clarithromycin with GABA receptors and/or GABAergic medications is warranted, and therapeutic applications of clarithromycin alone or in combination with flumazenil, to treat hyper-GABAergic status in PD at minimally effective doses, should also be pursued.

12.
Bioconjug Chem ; 27(5): 1382-9, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27043721

ABSTRACT

Three new positron emission tomography (PET) radiotracers of interest to our functional neuroimaging and translational oncology programs have been prepared through new developments in [(11)C]CO2 fixation chemistry. [(11)C]QZ (glutaminyl cyclase) was prepared via a tandem trapping of [(11)C]CO2/intramolecular cyclization; [(11)C]tideglusib (glycogen synthase kinase-3) was synthesized through a tandem trapping of [(11)C]CO2 followed by an intermolecular cycloaddition between a [(11)C]isocyanate and an isothiocyanate to form the 1,2,4-thiadiazolidine-3,5-dione core; [(11)C]ibrutinib (Bruton's tyrosine kinase) was synthesized through a HATU peptide coupling of an amino precursor with [(11)C]acrylic acid (generated from [(11)C]CO2 fixation with vinylmagnesium bromide). All radiochemical syntheses are fully automated on commercial radiochemical synthesis modules and provide radiotracers in 1-5% radiochemical yield (noncorrected, based upon [(11)C]CO2). All three radiotracers have advanced to rodent imaging studies and preliminary PET imaging results are also reported.


Subject(s)
Carbon Dioxide/chemistry , Carbon Dioxide/chemical synthesis , Carbon Radioisotopes , Positron-Emission Tomography , Animals , Chemistry Techniques, Synthetic , Cyclization , Mice , Radioactive Tracers , Rats
13.
ACS Chem Neurosci ; 7(3): 391-8, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26771209

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) is a 35 kDa transmembrane receptor that belongs to the immunoglobulin superfamily of cell surface molecules. Its role in Alzheimer's disease (AD) is complex, but it is thought to mediate influx of circulating amyloid-ß into the brain as well as amplify Aß-induced pathogenic responses. RAGE is therefore of considerable interest as both a diagnostic and a therapeutic target in AD. Herein we report the synthesis and preliminary preclinical evaluation of [(18)F]RAGER, the first small molecule PET radiotracer for RAGE (Kd = 15 nM). Docking studies proposed a likely binding interaction between RAGE and RAGER, [(18)F]RAGER autoradiography showed colocalization with RAGE identified by immunohistochemistry in AD brain samples, and [(18)F]RAGER microPET confirmed CNS penetration and increased uptake in areas of the brain known to express RAGE. This first generation radiotracer represents initial proof-of-concept and a promising first step toward quantifying CNS RAGE activity using PET. However, there were high levels of nonspecific [(18)F]RAGER binding in vitro, likely due to its high log P (experimental log P = 3.5), and rapid metabolism of [(18)F]RAGER in rat liver microsome studies. Therefore, development of second generation ligands with improved imaging properties would be advantageous prior to anticipated translation into clinical PET imaging studies.


Subject(s)
Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptor for Advanced Glycation End Products/analysis , Alzheimer Disease/metabolism , Animals , Autoradiography , Brain/metabolism , Chromatography, High Pressure Liquid , Fluorine Radioisotopes/pharmacokinetics , Humans , Immunohistochemistry , Macaca mulatta , Molecular Docking Simulation , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley
14.
ACS Chem Neurosci ; 6(12): 1965-71, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26393369

ABSTRACT

The isozymes of monoamine oxidase (MAO-A and MAO-B) are important enzymes involved in the metabolism of numerous biogenic amines, including the neurotransmitters serotonin, dopamine, and norepinephrine. Recently, changes in concentrations of MAO-B have been proposed to be an in vivo marker of neuroinflammation associated with Alzheimer's disease. Previous developments of in vivo radiotracers for imaging changes in MAO enzyme expression or activity have utilized the irreversible propargylamine-based suicide inhibitors or high-affinity reversibly binding inhibitors. As an alternative approach, we have investigated 1-[(11)C]methyl-4-aryloxy-1,2,3,6-tetrahydropyridines as metabolic trapping agents for the monoamine oxidases. MAO-mediated oxidation and spontaneous hydrolysis yield 1-[(11)C]methyl-2,3-dihydro-4-pyridinone as a hydrophilic metabolite that is trapped within brain tissues. Radiotracers with phenyl, biphenyl, and 7-coumarinyl ethers were evaluated using microPET imaging in rat and primate brains. No isozyme selectivity for radiotracer trapping was observed in the rat brain for any compound, but in the monkey brain, the phenyl ether demonstrated MAO-A selectivity and the coumarinyl ether showed MAO-B selectivity. These are lead compounds for further development of 1-[(11)C]methyl-4-aryloxy-1,2,3,6-tetrahydropyridines with optimized brain pharmacokinetics and isozyme selectivity.


Subject(s)
Monoamine Oxidase/metabolism , Radiochemistry , Animals , Carbon Isotopes/pharmacology , Diagnostic Imaging , Humans , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/pharmacology
15.
Medchemcomm ; 6(6): 1065-1068, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26101580

ABSTRACT

The phenol of 1-(3-(1H-imidazol-1-yl)propyl)-3-(4-hydroxy-3-methoxyphenyl)thiourea was selectively carbon-11 labelled to generate [11C]PBD150 in 7.3% yield from [11C]methyl triflate (non-decay corrected; radiochemical purity ≥95%, specific activity = 5.7 Ci/µmol, n=5). Evaluation of [11C]PBD150 by small animal PET imaging (mouse and rat) determined it does not permeate the blood brain barrier, indicating previously described therapeutic effect in transgenic mice was likely not the result of inhibiting central nervous system glutaminyl cyclase.

16.
ACS Med Chem Lett ; 6(5): 548-52, 2015 May 14.
Article in English | MEDLINE | ID: mdl-26005531

ABSTRACT

Quantifying glycogen synthase kinase-3 (GSK-3) activity in vivo using positron emission tomography (PET) imaging is of interest because dysregulation of GSK-3 is implicated in numerous diseases and neurological disorders for which GSK-3 inhibitors are being considered as therapeutic strategies. Previous PET radiotracers for GSK-3 have been reported, but none of the published examples cross the blood-brain barrier. Therefore, we have an ongoing interest in developing a brain penetrating radiotracer for GSK-3. To this end, we were interested in synthesis and preclinical evaluation of [(11)C]SB-216763, a high-affinity inhibitor of GSK-3 (K i = 9 nM; IC50 = 34 nM). Initial radiosyntheses of [(11)C]SB-216763 proved ineffective in our hands because of competing [3 + 3] sigmatropic shifts. Therefore, we have developed a novel one-pot two-step synthesis of [(11)C]SB-216763 from a 2,4-dimethoxybenzyl-protected maleimide precursor, which provided high specific activity [(11)C]SB-216763 in 1% noncorrected radiochemical yield (based upon [(11)C]CH3I) and 97-100% radiochemical purity (n = 7). Initial preclinical evaluation in rodent and nonhuman primate PET imaging studies revealed high initial brain uptake (peak rodent SUV = 2.5 @ 3 min postinjection; peak nonhuman primate SUV = 1.9 @ 5 min postinjection) followed by washout. Brain uptake was highest in thalamus, striatum, cortex, and cerebellum, areas known to be rich in GSK-3. These results make the arylindolemaleimide skeleton our lead scaffold for developing a PET radiotracer for quantification of GSK-3 density in vivo and ultimately translating it into clinical use.

17.
ACS Med Chem Lett ; 6(2): 112-116, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25705326

ABSTRACT

Interest in quantifying metal-Aß species in vivo led to the synthesis and evaluation of [11C]L2-b and [18F]FL2-b as radiopharmaceuticals for studying the metallobiology of Alzheimer's disease (AD) using positron emission tomography (PET) imaging. [11C]L2-b was synthesized in 3.6% radiochemical yield (nondecay corrected, n = 3), >95% radiochemical purity, from the corresponding desmethyl precursor. [18F]FL2-b was synthesized in 1.0% radiochemical yield (nondecay corrected, n = 3), >99% radiochemical purity, from a 6-chloro pyridine precursor. Autoradiography experiments with AD positive and healthy control brain samples were used to determine the specificity of binding for the radioligands compared to [11C]PiB, a known imaging agent for ß-amyloid (Aß) aggregates. The Kd for [11C]L2-b and [18F]FL2-b were found to be 3.5 and 9.4 nM, respectively, from those tissue studies. Displacement studies of [11C]L2-b and [18F]FL2-b with PiB and AV-45 determined that L2-b binds to Aß aggregates differently from known radiopharmaceuticals. Finally, brain uptake of [11C]L2-b was examined through microPET imaging in healthy rhesus macaque, which revealed a maximum uptake at 2.5 min (peak SUV = 2.0) followed by rapid egress (n = 2).

18.
ACS Chem Neurosci ; 5(8): 718-30, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-24896980

ABSTRACT

Abnormally aggregated tau is the hallmark pathology of tauopathy neurodegenerative disorders and is a target for development of both diagnostic tools and therapeutic strategies across the tauopathy disease spectrum. Development of carbon-11- or fluorine-18-labeled radiotracers with appropriate affinity and specificity for tau would allow noninvasive quantification of tau burden using positron emission tomography (PET) imaging. We have synthesized [(18)F]lansoprazole, [(11)C]N-methyl lansoprazole, and [(18)F]N-methyl lansoprazole and identified them as high affinity radiotracers for tau with low to subnanomolar binding affinities. Herein, we report radiosyntheses and extensive preclinical evaluation with the aim of selecting a lead radiotracer for translation into human PET imaging trials. We demonstrate that [(18)F]N-methyl lansoprazole, on account of the favorable half-life of fluorine-18 and its rapid brain entry in nonhuman primates, favorable kinetics, low white matter binding, and selectivity for binding to tau over amyloid, is the lead compound for progression into clinical trials.


Subject(s)
Alzheimer Disease/diagnostic imaging , Lansoprazole , Positron-Emission Tomography , Radiopharmaceuticals , Supranuclear Palsy, Progressive/diagnostic imaging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Autoradiography , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Drug Evaluation, Preclinical , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacokinetics , Humans , Lansoprazole/chemistry , Lansoprazole/pharmacokinetics , Mice , Peptide Fragments/metabolism , Positron-Emission Tomography/methods , Primates , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Supranuclear Palsy, Progressive/metabolism , tau Proteins/metabolism
19.
Nucl Med Biol ; 41(6): 507-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768148

ABSTRACT

INTRODUCTION: The dysfunction of glycogen synthase kinase-3ß (GSK-3ß) has been implicated in a number of diseases, including Alzheimer's disease. The ability to non-invasively quantify GSK-3ß activity in vivo is therefore of critical importance, and this work is focused upon development of inhibitors of GSK-3ß radiolabeled with carbon-11 to examine quantification of the enzyme using positron emission tomography (PET) imaging. METHODS: (11)C PyrATP-1 was prepared from the corresponding desmethyl-piperazine precursor in an automated synthesis module. In vivo rodent and primate imaging studies were conducted on a Concorde MicroPET P4 scanner to evaluate imaging properties and in vitro autoradiography studies with rat brain samples were carried out to examine specific binding. RESULTS: 2035±518MBq (55±14mCi) of [(11)C]PyrATP-1 was obtained (1%-2% non-corrected radiochemical yield at end-of-synthesis based upon [(11)C]CO2) with high chemical (>95%) and radiochemical (>99%) purities, and good specific activities (143±52GBq/µmol (3874±1424Ci/mmol)), n=5. In vivo microPET imaging studies revealed poor brain uptake in rodents and non-human primates. Pretreatment of rodents with cyclosporin A resulted in moderately increased brain uptake suggesting Pgp transporter involvement. Autoradiography demonstrated high levels of specific binding in areas of the rodent brain known to be rich in GSK-3ß. CONCLUSION: (11)C PyrATP-1 is readily synthesized using standard carbon-11 radiochemistry. However the poor brain uptake in rodents and non-human primates indicates that the radiotracer is not suitable for the purposes of quantifying GSK-3ß in neurological and psychiatric disorders.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/chemical synthesis , Pyrazinamide/analogs & derivatives , Pyrazines/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Brain/diagnostic imaging , Carbon Radioisotopes , Chemistry Techniques, Synthetic , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazinamide/chemical synthesis , Pyrazinamide/chemistry , Pyrazinamide/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Radioactive Tracers , Radiochemistry , Rats , Sulfonamides/chemistry , Sulfonamides/pharmacology
20.
J Labelled Comp Radiopharm ; 56(12): 595-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24285235

ABSTRACT

(-)-[(18) F]Flubatine was selected for clinical imaging of α4 ß2 nicotinic acetylcholine receptors because of its high affinity and appropriate kinetic profile. A fully automated synthesis of (-)-[(18) F]flubatine as a sterile isotonic solution suitable for clinical use is reported, as well as the first evaluation in nonhuman primates (rhesus macaques). (-)-[(18) F]Flubatine was prepared by fluorination of the Boc-protected trimethylammonium iodide precursor with [(18) F]fluoride in an automated synthesis module. Subsequent deprotection of the Boc group with 1-M HCl yielded (-)-[(18) F]flubatine, which was purified by semi-preparative HPLC. (-)-[(18) F]Flubatine was prepared in 25% radiochemical yield (formulated for clinical use at end of synthesis, n = 3), >95% radiochemical purity, and specific activity = 4647 Ci/mmol (171.9 GBq/µmol). Doses met all quality control criteria confirming their suitability for clinical use. Evaluation of (-)-[(18) F]flubatine in rhesus macaques was performed with a Concorde MicroPET P4 scanner (Concorde MicroSystems, Knoxville, TN). The brain was imaged for 90 min, and data were reconstructed using the 3-D maximum a posteriori algorithm. Image analysis revealed higher uptake and slower washout in the thalamus than those in other areas of the brain and peak uptake at 45 min. Injection of 2.5 µg/kg of nifene at 60 min initiated a slow washout of [(18) F]flubatine, with about 25% clearance from the thalamus by the end of imaging at 90 min.


Subject(s)
Benzamides/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Fluorine Radioisotopes/pharmacokinetics , Isotope Labeling/methods , Radiopharmaceuticals/pharmacokinetics , Animals , Automation, Laboratory , Benzamides/adverse effects , Benzamides/chemical synthesis , Brain/diagnostic imaging , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Drug Evaluation, Preclinical , Female , Fluorine Radioisotopes/adverse effects , Fluorine Radioisotopes/chemistry , Macaca mulatta , Positron-Emission Tomography/methods , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...