Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Bioorg Med Chem Lett ; 30(12): 127186, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32312583

RESUMEN

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.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Sulfonas/farmacología , Animales , Radioisótopos de Carbono , Relación Dosis-Respuesta a Droga , Furanos , Compuestos Heterocíclicos de 4 o más Anillos/síntesis química , Compuestos Heterocíclicos de 4 o más Anillos/química , Indenos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Sulfonamidas , Sulfonas/síntesis química , Sulfonas/química
2.
Molecules ; 25(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192089

RESUMEN

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.


Asunto(s)
Fentanilo/análogos & derivados , Naloxona/administración & dosificación , Naloxona/farmacología , Receptores Opioides mu/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Fentanilo/farmacología , Inyecciones Intramusculares , Macaca mulatta , Naloxona/sangre , Tomografía de Emisión de Positrones
3.
J Pharmacol Exp Ther ; 359(2): 366-373, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27625351

RESUMEN

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.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Nocicepción/efectos de los fármacos , Tomografía de Emisión de Positrones , Administración Intranasal , Animales , Conducta Animal/efectos de los fármacos , Fentanilo/administración & dosificación , Fentanilo/farmacología , Inyecciones Intramusculares , Macaca mulatta , Masculino , Naloxona/administración & dosificación , Naloxona/farmacología
4.
Bioconjug Chem ; 27(5): 1382-9, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-27043721

RESUMEN

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.


Asunto(s)
Dióxido de Carbono/química , Dióxido de Carbono/síntesis química , Radioisótopos de Carbono , Tomografía de Emisión de Positrones , Animales , Técnicas de Química Sintética , Ciclización , Ratones , Trazadores Radiactivos , Ratas
5.
Bioorg Med Chem Lett ; 23(6): 1612-6, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23416009

RESUMEN

A new cardiac sympathetic nerve imaging agent, [(18)F]4-fluoro-m-hydroxyphenethylguanidine ([(18)F]4F-MHPG), was synthesized and evaluated. The radiosynthetic intermediate [(18)F]4-fluoro-m-tyramine ([(18)F]4F-MTA) was prepared and then sequentially reacted with cyanogen bromide and NH4Br/NH4OH to afford [(18)F]4F-MHPG. Initial bioevaluations of [(18)F]4F-MHPG (biodistribution studies in rats and kinetic studies in the isolated rat heart) were similar to results previously reported for the carbon-11 labeled analog [(11)C]4F-MHPG. The neuronal uptake rate of [(18)F]4F-MHPG into the isolated rat heart was 0.68ml/min/g wet and its retention time in sympathetic neurons was very long (T1/2 >13h). A PET imaging study in a nonhuman primate with [(18)F]4F-MHPG provided high quality images of the heart, with heart-to-blood ratios at 80-90min after injection of 5-to-1. These initial kinetic and imaging studies of [(18)F]4F-MHPG suggest that this radiotracer may allow for more accurate quantification of regional cardiac sympathetic nerve density than is currently possible with existing neuronal imaging agents.


Asunto(s)
Medios de Contraste/síntesis química , Guanidinas/síntesis química , Metoxihidroxifenilglicol/química , Fenetilaminas/síntesis química , Animales , Medios de Contraste/farmacocinética , Radioisótopos de Flúor/química , Guanidinas/farmacocinética , Semivida , Corazón/diagnóstico por imagen , Macaca mulatta , Metoxihidroxifenilglicol/farmacocinética , Miocardio/metabolismo , Fenetilaminas/farmacocinética , Tomografía de Emisión de Positrones , Ratas , Distribución Tisular
6.
J Labelled Comp Radiopharm ; 56(12): 595-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24285235

RESUMEN

(-)-[(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.


Asunto(s)
Benzamidas/farmacocinética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Radioisótopos de Flúor/farmacocinética , Marcaje Isotópico/métodos , Radiofármacos/farmacocinética , Animales , Automatización de Laboratorios , Benzamidas/efectos adversos , Benzamidas/síntesis química , Encéfalo/diagnóstico por imagen , Compuestos Bicíclicos Heterocíclicos con Puentes/efectos adversos , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Evaluación Preclínica de Medicamentos , Femenino , Radioisótopos de Flúor/efectos adversos , Radioisótopos de Flúor/química , Macaca mulatta , Tomografía de Emisión de Positrones/métodos , Radiofármacos/efectos adversos , Radiofármacos/síntesis química
7.
ACS Med Chem Lett ; 11(6): 1299-1304, 2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32551015

RESUMEN

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.

8.
J Pharm Anal ; 10(5): 452-465, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33133729

RESUMEN

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.

9.
J Med Chem ; 62(21): 9600-9617, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31535859

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Descubrimiento de Drogas , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagen , Dominio Catalítico , Glucógeno Sintasa Quinasa 3 beta/química , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Neuroimagen , Oxazoles/química , Oxazoles/metabolismo , Oxazoles/farmacología , Inhibidores de Proteínas Quinasas/metabolismo , Triazoles/química , Triazoles/metabolismo , Triazoles/farmacología
10.
Pharmaceuticals (Basel) ; 11(4)2018 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-30551596

RESUMEN

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.

11.
J Med Chem ; 61(4): 1737-1743, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29257860

RESUMEN

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.


Asunto(s)
Radioisótopos de Flúor , Tomografía de Emisión de Positrones/métodos , Receptor trkA/antagonistas & inhibidores , Diseño de Fármacos , Humanos , Neoplasias/diagnóstico por imagen , Enfermedades Neurodegenerativas/diagnóstico por imagen , Inhibidores de Proteínas Quinasas
12.
J Med Chem ; 50(9): 2078-88, 2007 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-17419605

RESUMEN

The norepinephrine transporter (NET) substrates [123I]-m-iodobenzylguanidine (MIBG) and [11C]-m-hydroxyephedrine (HED) are used as markers of cardiac sympathetic neurons and adrenergic tumors (pheochromocytoma, neuroblastoma). However, their rapid NET transport rates limit their ability to provide accurate measurements of cardiac nerve density. [11C]Phenethylguanidine ([11C]1a) and 12 analogues ([11C]1b-m) were synthesized and evaluated as radiotracers with improved kinetics for quantifying cardiac nerve density. In isolated rat hearts, neuronal uptake rates of [11C]1a-m ranged from 0.24 to 1.96 mL min-1 (g wet wt)-1, and six compounds had extremely long neuronal retention times (clearance T1/2 > 20 h) due to efficient vesicular storage. Positron emission tomography (PET) studies in nonhuman primates with [11C]1e, N-[11C]guanyl-m-octopamine, which has a slow NET transport rate, showed improved myocardial kinetics compared to HED. Compound [11C]1c, [11C]-p-hydroxyphenethylguanidine, which has a rapid NET transport rate, avidly accumulated into rat pheochromocytoma xenograft tumors in mice. These encouraging findings demonstrate that radiolabeled phenethylguanidines deserve further investigation as radiotracers of cardiac sympathetic innervation and adrenergic tumors.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/diagnóstico por imagen , Guanidinas/síntesis química , Guanina/análogos & derivados , Corazón/inervación , Neuronas/metabolismo , Octopamina/análogos & derivados , Radiofármacos/síntesis química , Sistema Nervioso Simpático/metabolismo , Animales , Radioisótopos de Carbono , Guanidinas/química , Guanidinas/farmacocinética , Guanidinas/farmacología , Guanina/síntesis química , Guanina/química , Guanina/farmacocinética , Corazón/diagnóstico por imagen , Técnicas In Vitro , Macaca mulatta , Masculino , Ratones , Ratones Desnudos , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Octopamina/síntesis química , Octopamina/química , Octopamina/farmacocinética , Feocromocitoma , Tomografía de Emisión de Positrones , Radiofármacos/química , Radiofármacos/farmacología , Ratas , Relación Estructura-Actividad , Sistema Nervioso Simpático/citología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nucl Med Biol ; 34(5): 479-82, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591547

RESUMEN

The in vivo binding of N-[18F]fluoroethyl-piperidinyl benzilate ([18F]FEPB) to the muscarinic cholinergic receptor was measured in awake and anesthetized rats. Studies were done using an equilibrium infusion technique to provide estimates of specific binding as distribution volume ratios. Anesthesia with either isoflurane or sodium pentobarbital produced a significant (65-90%) increase of radiotracer binding in receptor-rich brain regions (striatum, cortex, hippocampus) relative to awake controls. Pretreatment of anesthetized animals with the acetylcholinesterase inhibitor phenserine produced no further increases in radioligand binding, in contrast to the large (>70%) increases previously observed in awake animals following drug treatment. These studies demonstrate that anesthesia can produce significant changes in baseline biochemical measures that can obscure even very large effects of pharmacological challenges.


Asunto(s)
Bencilatos/farmacocinética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Isoflurano/administración & dosificación , Pentobarbital/administración & dosificación , Piperidinas/farmacocinética , Anestésicos por Inhalación/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Radioisótopos de Flúor/farmacocinética , Hipnóticos y Sedantes/administración & dosificación , Masculino , Tasa de Depuración Metabólica/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Distribución Tisular/efectos de los fármacos
14.
ACS Chem Neurosci ; 8(7): 1530-1542, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28322043

RESUMEN

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.


Asunto(s)
Guanidinas , Corazón/inervación , Tomografía de Emisión de Positrones , Radiofármacos , Sistema Nervioso Simpático/diagnóstico por imagen , Animales , Evaluación Preclínica de Medicamentos , Guanidinas/sangre , Guanidinas/síntesis química , Guanidinas/química , Corazón/diagnóstico por imagen , Técnicas In Vitro , Isomerismo , Cinética , Macaca mulatta , Masculino , Estructura Molecular , Radiofármacos/sangre , Radiofármacos/síntesis química , Radiofármacos/química , Ratas Sprague-Dawley
15.
J Med Chem ; 60(16): 6897-6910, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28696690

RESUMEN

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.


Asunto(s)
Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridazinas/farmacología , Radiofármacos/farmacología , Receptor trkB/antagonistas & inhibidores , Receptor trkC/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Enfermedad de Alzheimer/diagnóstico por imagen , Animales , Benzamidas/farmacología , Barrera Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Carbono , Perros , Humanos , Imidazoles/síntesis química , Imidazoles/farmacocinética , Indazoles/farmacología , Macaca mulatta , Células de Riñón Canino Madin Darby , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Neuroimagen , Tomografía de Emisión de Positrones , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Piridazinas/síntesis química , Piridazinas/farmacocinética , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Ratas Sprague-Dawley , Receptor trkA/antagonistas & inhibidores , Estereoisomerismo , Relación Estructura-Actividad
16.
J Nucl Med ; 47(9): 1490-6, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16954558

RESUMEN

UNLABELLED: The norepinephrine analog (11)C-meta-hydroxyephedrine (HED) is used with PET to map the regional distribution of cardiac sympathetic neurons. HED is rapidly transported into sympathetic neurons by the norepinephrine transporter (NET) and stored in vesicles. Although much is known about the neuronal mechanisms of HED uptake and retention, there is little information about the functional relationship between HED retention and cardiac sympathetic nerve density. The goal of this study was to characterize the dependence of HED retention on nerve density in rats with graded levels of cardiac denervation induced chemically with the neurotoxin 6-hydroxydopamine (6-OHDA). METHODS: Thirty male Sprague-Dawley rats were divided into 6 groups, and each group was administered a different dose of 6-OHDA: 0 (controls), 7, 11, 15, 22, and 100 mg/kg intraperitoneally. One day after 6-OHDA injection, HED (3.7-8.3 MBq) was injected intravenously into each animal and HED concentrations in heart and blood at 30 min after injection were determined. Heart tissues were frozen and later processed by tissue homogenization and differential centrifugation into a membrane preparation for in vitro measurement of cardiac NET density. A saturation binding assay using (3)H-mazindol as the radioligand was used to measure NET density (maximum number of binding sites [B(max)], fmol/mg protein) for each heart. RESULTS: In control animals, NET B(max) was 388 +/- 23 fmol/mg protein and HED heart uptake (HU) at 30 min was 2.89% +/- 0.35 %ID/g (%ID/g is percentage injected dose per gram tissue). The highest 6-OHDA dose of 100 mg/kg caused severe cardiac denervation, decreasing both NET B(max) and HED HU to 8% of their control values. Comparing values for all doses of 6-OHDA, HED retention had a strong linear correlation with NET density: HU = 0.0077B(max) -0.028, r(2) = 0.95. CONCLUSION: HED retention is linearly dependent on NET density in rat hearts that have been chemically denervated with 6-OHDA, suggesting that HED retention is a good surrogate measure of NET density in the rat heart. This finding is discussed in relation to clinical observations of the dependence of HED retention on cardiac nerve density in human subjects using PET.


Asunto(s)
Efedrina/análogos & derivados , Corazón/diagnóstico por imagen , Miocardio/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Sistema Nervioso Simpático/diagnóstico por imagen , Sistema Nervioso Simpático/metabolismo , Animales , Radioisótopos de Carbono , Efedrina/farmacocinética , Corazón/inervación , Masculino , Tasa de Depuración Metabólica , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
17.
ACS Chem Neurosci ; 7(3): 391-8, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26771209

RESUMEN

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.


Asunto(s)
Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Receptor para Productos Finales de Glicación Avanzada/análisis , Enfermedad de Alzheimer/metabolismo , Animales , Autorradiografía , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Radioisótopos de Flúor/farmacocinética , Humanos , Inmunohistoquímica , Macaca mulatta , Simulación del Acoplamiento Molecular , Tomografía de Emisión de Positrones , Ratas , Ratas Sprague-Dawley
18.
ACS Med Chem Lett ; 7(8): 746-50, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27563397

RESUMEN

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.

19.
ACS Chem Neurosci ; 6(12): 1965-71, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26393369

RESUMEN

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.


Asunto(s)
Monoaminooxidasa/metabolismo , Radioquímica , Animales , Isótopos de Carbono/farmacología , Diagnóstico por Imagen , Humanos , Monoaminooxidasa/química , Inhibidores de la Monoaminooxidasa/farmacología
20.
Medchemcomm ; 6(6): 1065-1068, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26101580

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA