Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biomed Opt ; 23(12): 1-6, 2018 12.
Article in English | MEDLINE | ID: mdl-30520275

ABSTRACT

Premature cervical remodeling is a critical precursor of spontaneous preterm birth, and the remodeling process is characterized by an increase in tissue hydration. Nevertheless, current clinical measurements of cervical remodeling are subjective and detect only late events, such as cervical effacement and dilation. Here, we present a photoacoustic endoscope that can quantify tissue hydration by measuring near-infrared cervical spectra. We quantify the water contents of tissue-mimicking hydrogel phantoms as an analog of cervical connective tissue. Applying this method to pregnant women in vivo, we observed an increase in the water content of the cervix throughout pregnancy. The application of this technique in maternal healthcare may advance our understanding of cervical remodeling and provide a sensitive method for predicting preterm birth.


Subject(s)
Cervix Uteri/diagnostic imaging , Connective Tissue/diagnostic imaging , Image Processing, Computer-Assisted/methods , Photoacoustic Techniques/methods , Spectroscopy, Near-Infrared/methods , Adult , Equipment Design , Female , Humans , Phantoms, Imaging , Photoacoustic Techniques/instrumentation , Pregnancy , Spectroscopy, Near-Infrared/instrumentation
2.
J Biomed Opt ; 23(12): 1-4, 2018 12.
Article in English | MEDLINE | ID: mdl-30520276

ABSTRACT

Photoacoustic endoscopy offers in vivo examination of the visceral tissue using endogenous contrast, but its typical B-scan rate is ∼10 Hz, restricted by the speed of the scanning unit and the laser pulse repetition rate. Here, we present a transvaginal fast-scanning optical-resolution photoacoustic endoscope with a 250-Hz B-scan rate over a 3-mm scanning range. Using this modality, we not only illustrated the morphological differences of vasculatures among the human ectocervix, uterine body, and sublingual mucosa but also showed the longitudinal and cross-sectional differences of cervical vasculatures in pregnant women. This technology is promising for screening the visceral pathological changes associated with angiogenesis.


Subject(s)
Endoscopy/instrumentation , Photoacoustic Techniques , Adult , Cervix Uteri/diagnostic imaging , Equipment Design , Female , Humans , Photoacoustic Techniques/instrumentation , Photoacoustic Techniques/methods , Pregnancy , Young Adult
3.
ACS Omega ; 3(3): 3228-3234, 2018 Mar 31.
Article in English | MEDLINE | ID: mdl-31458580

ABSTRACT

The ability of capturing metals and/or radionuclides from large amounts of water is important for radioisotope waste treatment and environmental cleanup. We have developed an approach that rapidly optimizes the capturing of radioisotopes in large-volume aqueous environments. The approach was scaled up to capture beryllium-7 from 300 gallons of cooling water associated with a linear accelerator. Solid supports with the functional groups sulfonic acid, iminodiacetate, pyridine amine, pyridine amine acid, or quaternary amine were incubated in the cooling water for 1 week. One sulfonic acid solid support was able to capture 2.1 mCi of Be-7. Subsequent studies with the sulfonic acid solid support focused on the uptake over time of Be-7, scale-up of capturing Be-7, and subsequent purification of Be-7. The uptake over time of Be-7 was found to be linear in the first 24 h, with an equation of Y = 4.11X (% uptake/time (h)) (R 2 = 0.998). The uptake of Be-7 reached the maximum at 24 h and was identical to the uptake at 168 h. To purify Be-7, the optimal purification approach was to release the Be-7 from the solid support with 10 M HCl, which could be immediately passed through an AG1 resin to remove radioimpurities. The radiopurity of the purified Be-7 was greater than 99%, and this method was used to purify 65 mCi of the isotope.

4.
Neuropsychopharmacology ; 40(3): 650-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25249059

ABSTRACT

Selegiline (L-deprenyl) is a selective, irreversible inhibitor of monoamine oxidase B (MAO-B) at the conventional dose (10 mg/day oral) that is used in the treatment of Parkinson's disease. However, controlled studies have demonstrated antidepressant activity for high doses of oral selegiline and for transdermal selegiline suggesting that when plasma levels of selegiline are elevated, brain MAO-A might also be inhibited. Zydis selegiline (Zelapar) is an orally disintegrating formulation of selegiline, which is absorbed through the buccal mucosa producing higher plasma levels of selegiline and reduced amphetamine metabolites compared with equal doses of conventional selegiline. Although there is indirect evidence that Zydis selegiline at high doses loses its selectivity for MAO-B, there is no direct evidence that it also inhibits brain MAO-A in humans. We measured brain MAO-A in 18 healthy men after a 28-day treatment with Zydis selegiline (2.5, 5.0, or 10 mg/day) and in 3 subjects receiving the selegiline transdermal system (Emsam patch, 6 mg/day) using positron emission tomography and the MAO-A radiotracer [(11)C]clorgyline. We also measured dopamine transporter (DAT) availability in three subjects from the 10 mg group. The 10 mg Zydis selegiline dose significantly inhibited MAO-A (36.9±19.7%, range 11-70%, p<0.007)) but not DAT; and while Emsam also inhibited MAO-A (33.2±28.9 (range 9-68%) the difference did not reach significance (p=0.10)) presumably because of the small sample size. Our results provide the first direct evidence of brain MAO-A inhibition in humans by formulations of selegiline, which are currently postulated but not verified to target brain MAO-A in addition to MAO-B.


Subject(s)
Brain/drug effects , Brain/enzymology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Selegiline/pharmacology , Administration, Cutaneous , Administration, Oral , Adolescent , Adult , Brain/metabolism , Carbon Radioisotopes/metabolism , Clorgyline/metabolism , Cocaine/metabolism , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Functional Neuroimaging , Humans , Male , Monoamine Oxidase Inhibitors/administration & dosage , Positron-Emission Tomography , Selegiline/administration & dosage , Young Adult
5.
ACS Chem Neurosci ; 5(7): 588-96, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-24780082

ABSTRACT

Recent studies have revealed that several histone deacetylase (HDAC) inhibitors, which are used to study/treat brain diseases, show low blood-brain barrier (BBB) penetration. In addition to low HDAC potency and selectivity observed, poor brain penetrance may account for the high doses needed to achieve therapeutic efficacy. Here we report the development and evaluation of highly potent and blood-brain barrier permeable HDAC inhibitors for CNS applications based on an image-guided approach involving the parallel synthesis and radiolabeling of a series of compounds based on the benzamide HDAC inhibitor, MS-275 as a template. BBB penetration was optimized by rapid carbon-11 labeling and PET imaging in the baboon model and using the imaging derived data on BBB penetration from each compound to feed back into the design process. A total of 17 compounds were evaluated, revealing molecules with both high binding affinity and BBB permeability. A key element conferring BBB penetration in this benzamide series was a basic benzylic amine. These derivatives exhibited 1-100 nM inhibitory activity against recombinant human HDAC1 and HDAC2. Three of the carbon-11 labeled aminomethyl benzamide derivatives showed high BBB penetration (∼0.015%ID/cc) and regional binding heterogeneity in the brain (high in thalamus and cerebellum). Taken together this approach has afforded a strategy and a predictive model for developing highly potent and BBB permeable HDAC inhibitors for CNS applications and for the discovery of novel candidate molecules for small molecule probes and drugs.


Subject(s)
Benzamides/chemical synthesis , Benzamides/pharmacokinetics , Blood-Brain Barrier/metabolism , Capillary Permeability , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Animals , Benzamides/chemistry , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Drug Evaluation/methods , Female , Histone Deacetylase 1/chemistry , Histone Deacetylase 2/chemistry , Histone Deacetylase Inhibitors/chemistry , Humans , Papio anubis , Positron-Emission Tomography , Radiopharmaceuticals , Recombinant Proteins/chemistry
6.
Int J Neuropsychopharmacol ; 17(8): 1257-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24679922

ABSTRACT

Dopamine D3 receptor (D3R) antagonists may be effective medications for multiple substance use disorders (SUDs). However, no selective D3R antagonists are currently available for clinical testing. Buspirone, originally characterized as a 5-HT1A partial agonist and used as an anxiolytic, also binds to D3R and D4R with high affinity, with lower affinity to D2R, and interferes with cocaine reward. Here we used PET with [11C]PHNO (D3R-preferring radioligand), [11C]raclopride (D2R/D3R radioligand) and [11C]NNC-112 (D1R radioligand) to measure occupancy of oral and parenteral buspirone in the primate brain. Intramuscular buspirone (0.19 and 0.5 mg/kg) blocked both [11C]PHNO and [11C]raclopride binding to striatum, exhibiting high occupancy (50-85%) at 15 min and rapid wash-out over 2-6 h. In contrast, oral buspirone (3 mg/kg) significantly blocked [11C]PHNO binding in D3-rich regions (globus pallidum and midbrain) at 3 h, but had minimal effects on [11C]raclopride binding (28-37% at 1 h and 10% at 3 h). Buspirone did not block [11C]NNC-112. Our findings provide evidence that i.m. buspirone blocks D3R and D2R, whereas oral buspirone is more selective towards D3R blockade in vivo, consistent with extensive first pass metabolism and supporting the hypothesis that its metabolites (5- and 6'-hydroxybuspirone) merit evaluation for treating SUDs. They also indicate that for oral buspirone to achieve greater than 80% sustained D3R occupancy, as might be needed to treat addiction, higher doses (at least three-fold) than those used to treat anxiety (maximal 60 mg) will be required. Nonetheless, based on previous clinical studies, these doses would be safe and well tolerated.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Buspirone/administration & dosage , Buspirone/pharmacology , Corpus Striatum/drug effects , Globus Pallidus/drug effects , Mesencephalon/drug effects , Receptors, Dopamine D3/antagonists & inhibitors , Administration, Oral , Animals , Benzazepines , Benzofurans , Corpus Striatum/diagnostic imaging , Dopamine D2 Receptor Antagonists/administration & dosage , Dopamine D2 Receptor Antagonists/pharmacology , Dose-Response Relationship, Drug , Female , Functional Neuroimaging , Globus Pallidus/diagnostic imaging , Injections, Intramuscular , Mesencephalon/diagnostic imaging , Oxazines , Papio anubis , Positron-Emission Tomography , Raclopride , Radioligand Assay
7.
Bioorg Med Chem Lett ; 23(24): 6700-5, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24210501

ABSTRACT

Histone deacetylases (HDACs) regulate gene expression by inducing conformational changes in chromatin. Ever since the discovery of a naturally occurring HDAC inhibitor, trichostatin A (TSA) stimulated the recent development of suberoylanilide (SAHA, Zolinza®), HDAC has become an important molecular target for drug development. This has created the need to develop specific in vivo radioligands to study epigenetic regulation and HDAC engagement for drug development for diseases including cancer and psychiatric disorders. 6-([(18)F]Fluoroacetamido)-1-hexanoicanilide ([(18)F]FAHA) was recently developed as a HDAC substrate and shows moderate blood-brain barrier (BBB) permeability and specific signal (by metabolic trapping/or deacetylation) but rapid metabolism. Here, we report the radiosynthesis of two carbon-11 labeled candidate radiotracers (substrate- and inhibitor-based radioligand) for HDAC and their evaluation in non-human primate brain. PET studies showed very low brain uptake and rapid metabolism of both labeled compounds but revealed a surprising enhancement of brain penetration by F for H substitution when comparing one of these to [(18)F]FAHA. Further structural refinement is needed for the development of brain-penetrant, metabolically stable HDAC radiotracers and to understand the role of fluorine substitution on brain penetration.


Subject(s)
Brain/diagnostic imaging , Brain/enzymology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Radiopharmaceuticals/chemistry , Anilides/chemistry , Animals , Blood-Brain Barrier/metabolism , Carbon Radioisotopes/chemistry , Fluorine Radioisotopes/chemistry , Fluoroacetates/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylases/metabolism , Permeability/drug effects , Positron-Emission Tomography , Primates , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology
8.
Nucl Med Biol ; 40(7): 912-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23906667

ABSTRACT

The fatty acids, n-butyric acid (BA), 4-phenylbutyric acid (PBA) and valproic acid (VPA, 2-propylpentanoic acid) have been used for many years in the treatment of a variety of CNS and peripheral organ diseases including cancer. New information that these drugs alter epigenetic processes through their inhibition of histone deacetylases (HDACs) has renewed interest in their biodistribution and pharmacokinetics and the relationship of these properties to their therapeutic and side effect profiles. In order to determine the pharmacokinetics and biodistribution of these drugs in primates, we synthesized their carbon-11 labeled analogues and performed dynamic positron emission tomography (PET) in six female baboons over 90 min. The carbon-11 labeled carboxylic acids were prepared by using (11)CO2 and the appropriate Grignard reagents. [(11)C]BA was metabolized rapidly (only 20% of the total carbon-11 in plasma was parent compound at 5 min post injection) whereas for VPA and PBA 98% and 85% of the radioactivity were the unmetabolized compound at 30 min after their administration respectively. The brain uptake of all three carboxylic acids was very low (<0.006%ID/cc, BA>VPA>PBA), which is consistent with the need for very high doses for therapeutic efficacy. Most of the radioactivity was excreted through the kidneys and accumulated in the bladder. However, the organ biodistribution between the drugs differed. [(11)C]BA showed relatively high uptake in spleen and pancreas whereas [(11)C]PBA showed high uptake in liver and heart. Notably, [(11)C]VPA showed exceptionally high heart uptake possibly due to its involvement in lipid metabolism. The unique biodistribution of each of these drugs may be of relevance in understanding their therapeutic and side effect profile including their teratogenic effects.


Subject(s)
Histone Deacetylase Inhibitors/pharmacokinetics , Positron-Emission Tomography , Animals , Blood Proteins/metabolism , Brain/diagnostic imaging , Brain/metabolism , Butyric Acid/blood , Butyric Acid/metabolism , Butyric Acid/pharmacokinetics , Carbon Radioisotopes , Female , Histone Deacetylase Inhibitors/blood , Histone Deacetylase Inhibitors/metabolism , Isotope Labeling , Papio , Phenylbutyrates/blood , Phenylbutyrates/metabolism , Phenylbutyrates/pharmacokinetics , Radiochemistry , Tissue Distribution , Valproic Acid/blood , Valproic Acid/metabolism , Valproic Acid/pharmacokinetics
9.
Neuroimage ; 64: 277-83, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22947541

ABSTRACT

Alcohol intoxication results in marked reductions in brain glucose metabolism, which we hypothesized reflect not just its GABAergic enhancing effects but also the metabolism of acetate as an alternative brain energy source. To test this hypothesis we separately assessed the effects of alcohol intoxication on brain glucose and acetate metabolism using Positron Emission Tomography (PET). We found that alcohol intoxication significantly decreased whole brain glucose metabolism (measured with FDG) with the largest decrements in cerebellum and occipital cortex and the smallest in the thalamus. In contrast, alcohol intoxication caused a significant increase in [1-(11)C]acetate brain uptake (measured as standard uptake value, SUV), with the largest increases occurring in the cerebellum and the smallest in the thalamus. In heavy alcohol drinkers [1-(11)C]acetate brain uptake during alcohol challenge tended to be higher than in occasional drinkers (p<0.06) and the increases in [1-(11)C]acetate uptake in cerebellum with alcohol were positively associated with the reported amount of alcohol consumed (r=0.66, p<0.01). Our findings corroborate a reduction of brain glucose metabolism during intoxication and document an increase in brain acetate uptake. The opposite changes observed between regional brain metabolic decrements and regional increases in [1-(11)C]acetate uptake support the hypothesis that during alcohol intoxication the brain may rely on acetate as an alternative brain energy source and provides preliminary evidence that heavy alcohol exposures may facilitate the use of acetate as an energy substrate. These findings raise the question of the potential therapeutic benefits that increasing plasma acetate concentration (i.e. ketogenic diets) may have in alcoholics undergoing alcohol detoxification.


Subject(s)
Acetates/pharmacokinetics , Alcoholic Intoxication/etiology , Alcoholic Intoxication/metabolism , Brain/metabolism , Carbon/pharmacokinetics , Ethanol/poisoning , Fluorodeoxyglucose F18/pharmacokinetics , Glucose/metabolism , Adolescent , Adult , Brain/diagnostic imaging , Child , Female , Humans , Male , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Young Adult
10.
Mol Imaging ; 12(8)2013.
Article in English | MEDLINE | ID: mdl-24447618

ABSTRACT

The aim of this work was to quantify the brain distribution of the enzyme aromatase in the female baboon with positron emission tomography and the tracer [11C]vorozole using three different quantification methods for estimating the total distribution volume (V(T)): a graphical method, compartment modeling, and a tissue to plasma ratio. The graphical model and the compartment modeling gave similar estimates to the data and similar values (correlation R  =  .988; p  =  .0001). [11C]Vorozole shows a rapid uptake by the brain followed by a relatively constant accumulation, suggesting the possibility of using the tissue to plasma ratio as an estimate of V(T). The highest uptake of [11C]vorozole in the baboon brain was measured in the amygdala, followed by the preoptic area and hypothalamus, basal ganglia, and cortical areas. Pretreatment studies with vorozole or letrozole showed a generalized decrease in brain accumulation and V(T). The results suggested that the physiologic changes in gonadal hormone levels accompanying the menstrual cycle had a significant effect on brain aromatase V(T).


Subject(s)
Aromatase/metabolism , Brain/diagnostic imaging , Brain/enzymology , Menstrual Cycle , Nitriles/pharmacokinetics , Triazoles/pharmacokinetics , Animals , Carbon Radioisotopes , Female , Letrozole , Papio , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics
11.
Psychopharmacology (Berl) ; 223(4): 447-55, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22614669

ABSTRACT

RATIONALE: The preclinical characterization of a series of aryloxypyridine amides has identified JNJ-39220675 ((4-cyclobutyl-1,4-diazepan-1-yl)(6-(4-fluorophenoxy)pyridin-3-yl)methanone) as a high-affinity histamine H(3) receptor antagonist and a candidate for further drug development particularly in the treatment of alcohol-related behaviors. OBJECTIVE: This study measured brain histamine H(3) receptor blockade by JNJ-39220675 (1 mg/kg) in the female baboon. METHODS: Positron emission tomography imaging and [(11)C]GSK189254, a reversible high-affinity radiotracer with specificity for the histamine H(3) receptor, was used to measure histamine H(3) receptor availability at baseline and after i.v. and oral administration of JNJ-39220675 (1 mg/kg) in the anesthetized baboon. Histamine H(3) receptor availability was estimated as the total distribution volume (V (T)) in brain regions. The sensitivity of [(11)C]GSK189254 binding to injected mass and carryover effects was determined. RESULTS: JNJ-39220675 produces robust (ca. 90 %) blockade of [(11)C]GSK189254 binding after i.v. and oral administration. After oral administration of JNJ-39220675 (1 mg/kg), the fractional receptor occupancy was >0.9 at 90 min with a slight increase from 90 to 240 min. Similar to prior studies in humans, V (T) was highly sensitive to the mass of GSK189254 with ED(50) estimated to be 0.16 µg/kg. CONCLUSIONS: The robust blockade of binding of [(11)C]GSK189254 by JNJ-39220675 demonstrates that this compound readily penetrates the blood-brain barrier and occupies the histamine H(3) receptor after oral administration at low plasma concentrations (∼1 ng/cc) supporting further drug development for alcohol addiction and other disorders. This study corroborates prior reports of the high sensitivity of [(11)C]GSK189254 to injected mass at doses >0.1 µg/kg.


Subject(s)
Azepines/pharmacology , Benzazepines/pharmacology , Histamine H3 Antagonists/pharmacology , Niacinamide/analogs & derivatives , Pyridines/pharmacology , Receptors, Histamine H3/drug effects , Administration, Oral , Animals , Azepines/administration & dosage , Azepines/pharmacokinetics , Benzazepines/administration & dosage , Benzazepines/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Dose-Response Relationship, Drug , Female , Histamine H3 Antagonists/administration & dosage , Histamine H3 Antagonists/pharmacokinetics , Injections, Intravenous , Niacinamide/administration & dosage , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Papio , Positron-Emission Tomography , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Receptors, Histamine H3/metabolism , Sensitivity and Specificity , Time Factors , Tissue Distribution
12.
Synapse ; 64(11): 801-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20842717

ABSTRACT

Aromatase catalyzes the last step in estrogen biosynthesis. Brain aromatase is involved in diverse neurophysiological and behavioral functions including sexual behavior, aggression, cognition, and neuroprotection. Using positron emission tomography (PET) with the radiolabeled aromatase inhibitor [N-methyl-(11)C]vorozole, we characterized the tracer distribution and kinetics in the living human brain. Six young, healthy subjects, three men and three women, were administered the radiotracer alone on two separate occasions. Women were scanned in distinct phases of the menstrual cycle. Specificity was confirmed by pretreatment with a pharmacological (2.5 mg) dose of the aromatase inhibitor letrozole. PET data were acquired over a 90-min period and regions of interest placed over selected brain regions. Brain and plasma time activity curves, corrected for metabolites, were used to derive kinetic parameters. Distribution volume (V(T)) values in both men and women followed the following rank order: thalamus > amygdala = preoptic area > medulla (inferior olive) > accumbens, pons, occipital and temporal cortex, putamen, cerebellum, and white matter. Pretreatment with letrozole reduced V(T) in all regions, though the size of the reduction was region-dependent, ranging from ∼70% blocking in thalamus andpreoptic area to ∼10% in cerebellum. The high levels of aromatase in thalamus and medulla (inferior olive) appear to be unique to humans. These studies set the stage for the noninvasive assessment of aromatase involvement in various physiological and pathological processes affecting the human brain.


Subject(s)
Aromatase Inhibitors/pharmacokinetics , Aromatase/metabolism , Brain/diagnostic imaging , Brain/enzymology , Positron-Emission Tomography , Triazoles/pharmacokinetics , Adult , Brain/drug effects , Brain Mapping , Female , Humans , Male , Protein Binding/drug effects , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Young Adult
13.
Biol Psychiatry ; 67(8): 774-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20188349

ABSTRACT

BACKGROUND: Cigarette smoking and nicotine have complex effects on human physiology and behavior, including some effects similar to those elicited by inhibition of aromatase, the last enzyme in estrogen biosynthesis. We report the first in vivo primate study to determine whether there is a direct effect of nicotine administration on brain aromatase. METHODS: Brain aromatase availability was examined with positron emission tomography and the selective aromatase inhibitor [(11)C]vorozole in six baboons before and after exposure to IV nicotine at .015 and .03 mg/kg. RESULTS: Nicotine administration produced significant, dose-dependent reductions in [(11)C]vorozole binding. The amygdala and preoptic area showed the largest reductions. Plasma levels of nicotine and its major metabolite cotinine were similar to those found in cigarette smokers. CONCLUSIONS: Nicotine interacts in vivo with primate brain aromatase in regions involved in mood, aggression, and sexual behavior.


Subject(s)
Aromatase Inhibitors , Brain/enzymology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Papio/metabolism , Animals , Aromatase Inhibitors/pharmacokinetics , Brain/diagnostic imaging , Brain/drug effects , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Female , Injections, Intravenous , Magnetic Resonance Imaging , Positron-Emission Tomography , Triazoles/pharmacokinetics
14.
Neuropsychopharmacology ; 35(3): 623-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19890267

ABSTRACT

Reversible inhibitors of monoamine oxidase-A (RIMA) inhibit the breakdown of three major neurotransmitters, serotonin, norepinephrine and dopamine, offering a multi-neurotransmitter strategy for the treatment of depression. CX157 (3-fluoro-7-(2,2,2-trifluoroethoxy)phenoxathiin-10,10-dioxide) is a RIMA, which is currently in development for the treatment of major depressive disorder. We examined the degree and reversibility of the inhibition of brain monoamine oxidase-A (MAO-A) and plasma CX157 levels at different times after oral dosing to establish a dosing paradigm for future clinical efficacy studies, and to determine whether plasma CX157 levels reflect the degree of brain MAO-A inhibition. Brain MAO-A levels were measured with positron emission tomography (PET) imaging and [(11)C]clorgyline in 15 normal men after oral dosing of CX157 (20-80 mg). PET imaging was conducted after single and repeated doses of CX157 over a 24-h time course. We found that 60 and 80 mg doses of CX157 produced a robust dose-related inhibition (47-72%) of [(11)C]clorgyline binding to brain MAO-A at 2 h after administration and that brain MAO-A recovered completely by 24 h post drug. Plasma CX157 concentration was highly correlated with the inhibition of brain MAO-A (EC(50): 19.3 ng/ml). Thus, CX157 is the first agent in the RIMA class with documented reversible inhibition of human brain MAO-A, supporting its classification as a RIMA, and the first RIMA with observed plasma levels that can serve as a biomarker for the degree of brain MAO-A inhibition. These data were used to establish the dosing regimen for a current clinical efficacy trial with CX157.


Subject(s)
Brain/drug effects , Brain/enzymology , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Monoamine Oxidase Inhibitors/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Adult , Clorgyline/metabolism , Heterocyclic Compounds/chemistry , Humans , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Positron-Emission Tomography/methods , Protein Binding/drug effects , Protein Binding/physiology , Young Adult
15.
JAMA ; 301(11): 1148-54, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19293415

ABSTRACT

CONTEXT: Modafinil, a wake-promoting drug used to treat narcolepsy, is increasingly being used as a cognitive enhancer. Although initially launched as distinct from stimulants that increase extracellular dopamine by targeting dopamine transporters, recent preclinical studies suggest otherwise. OBJECTIVE: To measure the acute effects of modafinil at doses used therapeutically (200 mg and 400 mg given orally) on extracellular dopamine and on dopamine transporters in the male human brain. DESIGN, SETTING, AND PARTICIPANTS: Positron emission tomography with [(11)C]raclopride (D(2)/D(3) radioligand sensitive to changes in endogenous dopamine) and [(11)C]cocaine (dopamine transporter radioligand) was used to measure the effects of modafinil on extracellular dopamine and on dopamine transporters in 10 healthy male participants. The study took place over an 8-month period (2007-2008) at Brookhaven National Laboratory. MAIN OUTCOME MEASURES: Primary outcomes were changes in dopamine D(2)/D(3) receptor and dopamine transporter availability (measured by changes in binding potential) after modafinil when compared with after placebo. RESULTS: Modafinil decreased mean (SD) [(11)C]raclopride binding potential in caudate (6.1% [6.5%]; 95% confidence interval [CI], 1.5% to 10.8%; P = .02), putamen (6.7% [4.9%]; 95% CI, 3.2% to 10.3%; P = .002), and nucleus accumbens (19.4% [20%]; 95% CI, 5% to 35%; P = .02), reflecting increases in extracellular dopamine. Modafinil also decreased [(11)C]cocaine binding potential in caudate (53.8% [13.8%]; 95% CI, 43.9% to 63.6%; P < .001), putamen (47.2% [11.4%]; 95% CI, 39.1% to 55.4%; P < .001), and nucleus accumbens (39.3% [10%]; 95% CI, 30% to 49%; P = .001), reflecting occupancy of dopamine transporters. CONCLUSIONS: In this pilot study, modafinil blocked dopamine transporters and increased dopamine in the human brain (including the nucleus accumbens). Because drugs that increase dopamine in the nucleus accumbens have the potential for abuse, and considering the increasing use of modafinil, these results highlight the need for heightened awareness for potential abuse of and dependence on modafinil in vulnerable populations.


Subject(s)
Benzhydryl Compounds/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine/metabolism , Adult , Benzhydryl Compounds/administration & dosage , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Caudate Nucleus/metabolism , Central Nervous System Stimulants/administration & dosage , Cocaine , Humans , Male , Middle Aged , Modafinil , Nucleus Accumbens/metabolism , Pilot Projects , Positron-Emission Tomography , Putamen/metabolism , Raclopride , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Substance-Related Disorders , Young Adult
16.
Nucl Med Biol ; 36(3): 323-34, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19324278

ABSTRACT

INTRODUCTION: We reinvestigated the synthesis of [N-methyl-(11)C]vorozole, a radiotracer for aromatase, and discovered the presence of an N-methyl isomer which was not removed in the original purification method. Herein we report the preparation and positron emission tomography (PET) studies of pure [N-methyl-(11)C]vorozole. METHODS: Norvorozole was alkylated with [(11)C]methyl iodide as previously described and also with unlabeled methyl iodide. A high-performance liquid chromatography (HPLC) method was developed to separate the regioisomers. Nuclear magnetic resonance (NMR) spectroscopy ((13)C and 2D-nuclear Overhauser effect spectroscopy NMR) was used to identify and assign structures to the N-methylated products. Pure [N-methyl-(11)C]vorozole and the contaminating isomer were compared by PET imaging in the baboon. RESULTS: Methylation of norvorozole resulted in a mixture of isomers (1:1:1 ratio) based on new HPLC analysis using a pentafluorophenylpropyl bonded silica column, in which vorozole coeluted one of its isomers under the original HPLC conditions. Baseline separation of the three labeled isomers was achieved. The N-3 isomer was the contaminant of vorozole, thus correcting the original assignment of isomers. PET studies of pure [N-methyl-(11)C]vorozole with and without the contaminating N-3 isomer revealed that only [N-methyl-(11)C]vorozole binds to aromatase. [N-methyl-(11)C]Vorozole accumulated in all brain regions with highest accumulation in the aromatase-rich amygdala and preoptic area. Accumulation was blocked with vorozole and letrozole consistent with reports of some level of aromatase in many brain regions. CONCLUSIONS: The discovery of a contaminating labeled isomer and the development of a method for isolating pure [N-methyl-(11)C]vorozole combine to provide a new scientific tool for PET studies of the biology of aromatase and for drug research and development.


Subject(s)
Aromatase/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , Alkylation , Animals , Aromatase/analysis , Brain/diagnostic imaging , Brain/metabolism , Chromatography, High Pressure Liquid , Female , Hydrocarbons, Iodinated/chemistry , Magnetic Resonance Spectroscopy , Papio , Positron-Emission Tomography , Radioactive Tracers , Stereoisomerism , Time Factors , Triazoles/chemistry
17.
Neuroimage ; 43(4): 756-63, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18708148

ABSTRACT

Methamphetamine is one of the most addictive and neurotoxic drugs of abuse. It produces large elevations in extracellular dopamine in the striatum through vesicular release and inhibition of the dopamine transporter. In the U.S. abuse prevalence varies by ethnicity with very low abuse among African Americans relative to Caucasians, differentiating it from cocaine where abuse rates are similar for the two groups. Here we report the first comparison of methamphetamine and cocaine pharmacokinetics in brain between Caucasians and African Americans along with the measurement of dopamine transporter availability in striatum. Methamphetamine's uptake in brain was fast (peak uptake at 9 min) with accumulation in cortical and subcortical brain regions and in white matter. Its clearance from brain was slow (except for white matter which did not clear over the 90 min) and there was no difference in pharmacokinetics between Caucasians and African Americans. In contrast cocaine's brain uptake and clearance were both fast, distribution was predominantly in striatum and uptake was higher in African Americans. Among individuals, those with the highest striatal (but not cerebellar) methamphetamine accumulation also had the highest dopamine transporter availability suggesting a relationship between METH exposure and DAT availability. Methamphetamine's fast brain uptake is consistent with its highly reinforcing effects, its slow clearance with its long-lasting behavioral effects and its widespread distribution with its neurotoxic effects that affect not only striatal but also cortical and white matter regions. The absence of significant differences between Caucasians and African Americans suggests that variables other than methamphetamine pharmacokinetics and bioavailability account for the lower abuse prevalence in African Americans.


Subject(s)
Black or African American , Brain/metabolism , Cocaine/pharmacokinetics , Methamphetamine/pharmacokinetics , Positron-Emission Tomography/methods , White People , Adult , Brain/diagnostic imaging , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/pharmacokinetics , Cocaine/administration & dosage , Humans , Male , Metabolic Clearance Rate , Methamphetamine/administration & dosage , Middle Aged , Tissue Distribution , Young Adult
18.
J Nucl Med ; 48(10): 1724-32, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873134

ABSTRACT

UNLABELLED: The methamphetamine molecule has a chiral center and exists as 2 enantiomers, d-methamphetamine (the more active enantiomer) and l-methamphetamine (the less active enantiomer). d-Methamphetamine is associated with more intense stimulant effects and higher abuse liability. The objective of this study was to measure the pharmacokinetics of d-methamphetamine for comparison with both l-methamphetamine and (-)-cocaine in the baboon brain and peripheral organs and to assess the saturability and pharmacologic specificity of binding. METHODS: d- and l-methamphetamine and (-)-cocaine were labeled with (11)C via alkylation of the norprecursors with (11)C-methyl iodide using literature methods. Six different baboons were studied in 11 PET sessions at which 2 radiotracer injections were administered 2-3 h apart to determine the distribution and kinetics of (11)C-d-methamphetamine in brain and peripheral organs. Saturability and pharmacologic specificity were assessed using pretreatment with d-methamphetamine, methylphenidate, and tetrabenazine. (11)C-d-Methamphetamine pharmacokinetics were compared with (11)C-l-methamphetamine and (11)C-(-)-cocaine in both brain and peripheral organs in the same animal. RESULTS: (11)C-d- and l-methamphetamine both showed high uptake and widespread distribution in the brain. Pharmacokinetics did not differ between enantiomers, and the cerebellum peaked earlier and cleared more quickly than the striatum for both. (11)C-d-Methamphetamine distribution volume ratio was not substantially affected by pretreatment with methamphetamine, methylphenidate, or tetrabenazine. Both enantiomers showed rapid, high uptake and clearance in the heart and lungs and slower uptake and clearance in the liver and kidneys. A comparison of (11)C-d-methamphetamine and (11)C-(-)-cocaine showed that (11)C-d-methamphetamine peaked later in the brain than did (11)C-(-)-cocaine and cleared more slowly. The 2 drugs showed similar behavior in all peripheral organs examined except the kidneys and pancreas, which showed higher uptake for (11)C-d-methamphetamine. CONCLUSION: Brain pharmacokinetics did not differ between d-and l-methamphetamine and thus cannot account for the more intense stimulant effects of d-methamphetamine. Lack of pharmacologic blockade by methamphetamine indicates that the PET image represents nonspecific binding, though the fact that methamphetamine is both a transporter substrate and an inhibitor may also play a role. A comparison of (11)C-d-methamphetamine and (11)C-(-)-cocaine in the same animal showed that the slower clearance of methamphetamine is likely to contribute to its previously reported longer-lasting stimulant effects relative to those of (-)-cocaine. High kidney uptake of d-methamphetamine or its labeled metabolites may account for the reported renal toxicity of d-methamphetamine in humans.


Subject(s)
Brain/metabolism , Cocaine/pharmacokinetics , Methamphetamine/pharmacokinetics , Papio/metabolism , Animals , Brain/diagnostic imaging , Metabolic Clearance Rate , Positron-Emission Tomography/methods , Tissue Distribution
19.
Nucl Med Biol ; 34(6): 667-79, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17707807

ABSTRACT

UNLABELLED: Results from human studies with the PET radiotracer (S,S)-[(11)C]O-methyl reboxetine ([(11)C](S,S)-MRB), a ligand targeting the norepinephrine transporter (NET), are reported. Quantification methods were determined from test/retest studies, and sensitivity to pharmacological blockade was tested with different doses of atomoxetine (ATX), a drug that binds to the NET with high affinity (K(i)=2-5 nM). METHODS: Twenty-four male subjects were divided into different groups for serial 90-min PET studies with [(11)C](S,S)-MRB to assess reproducibility and the effect of blocking with different doses of ATX (25, 50 and 100 mg, po). Region-of-interest uptake data and arterial plasma input were analyzed for the distribution volume (DV). Images were normalized to a template, and average parametric images for each group were formed. RESULTS: [(11)C](S,S)-MRB uptake was highest in the thalamus (THL) and the midbrain (MBR) [containing the locus coeruleus (LC)] and lowest for the caudate nucleus (CDT). The CDT, a region with low NET, showed the smallest change on ATX treatment and was used as a reference region for the DV ratio (DVR). The baseline average DVR was 1.48 for both the THL and MBR with lower values for other regions [cerebellum (CB), 1.09; cingulate gyrus (CNG) 1.07]. However, more accurate information about relative densities came from the blocking studies. MBR exhibited greater blocking than THL, indicating a transporter density approximately 40% greater than THL. No relationship was found between DVR change and plasma ATX level. Although the higher dose tended to induce a greater decrease than the lower dose for MBR (average decrease for 25 mg=24+/-7%; 100 mg=31+/-11%), these differences were not significant. The different blocking between MBR (average decrease=28+/-10%) and THL (average decrease=17+/-10%) given the same baseline DVR indicates that the CDT is not a good measure for non-NET binding in both regions. Threshold analysis of the difference between the average baseline DV image and the average blocked image showed the expected NET distribution with the MBR (LC) and hypothalamus>THL>CNG and CB, as well as a significant change in the supplementary motor area. DVR reproducibility for the different brain regions was approximately 10%, but intersubject variability was large. CONCLUSIONS: The highest density of NETs was found in the MBR where the LC is located, followed by THL, whereas the lowest density was found in basal ganglia (lowest in CDT), consistent with the regional localization of NETs in the nonhuman primate brain. While all three doses of ATX were found to block most regions, no significant differences between doses were found for any region, although the average percent change across subjects of the MBR did correlate with ATX dose. The lack of a dose effect could reflect a low signal-to-noise ratio coupled with the possibility that a sufficient number of transporters were blocked at the lowest dose and further differences could not be detected. However, since the lowest (25 mg) dose is less than the therapeutic doses used in children for the treatment of attention-deficit/hyperactivity disorder ( approximately 1.0 mg/kg/day), this would suggest that there may be additional targets for ATX's therapeutic actions.


Subject(s)
Morpholines , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Radiopharmaceuticals , Adrenergic Uptake Inhibitors/pharmacokinetics , Adrenergic Uptake Inhibitors/pharmacology , Adult , Algorithms , Atomoxetine Hydrochloride , Brain/diagnostic imaging , Carbon Radioisotopes , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Humans , Image Processing, Computer-Assisted , Male , Morpholines/blood , Morpholines/pharmacokinetics , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Positron-Emission Tomography , Propylamines/pharmacokinetics , Propylamines/pharmacology , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacokinetics , Reboxetine , Reproducibility of Results
20.
Nucl Med Biol ; 34(5): 541-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591554

ABSTRACT

INTRODUCTION: (3E)-3-[(2,4-dimethoxyphenyl)methylene]-3,4,5,6-tetrahydro-2,3'-bipyridine (GTS-21), a partial alpha7 nicotinic acetylcholine receptor agonist drug, has recently been shown to improve cognition in schizophrenia and Alzheimer's disease. One of its two major demethylated metabolites, 4-OH-GTS-21, has been suggested to contribute to its therapeutic effects. METHODS: We labeled GTS-21 in two different positions with carbon-11 ([2-methoxy-(11)C]GTS-21 and [4-(11)C]GTS-21) along with two corresponding demethylated metabolites ([2-methoxy-(11)C]4-OH-GTS-21 and [4-methoxy-(11)C]2-OH-GTS-21) for pharmacokinetic studies in baboons and mice with positron emission tomography (PET). RESULTS: Both [2-(11)C]GTS-21 and [4-methoxy-(11)C]GTS-21 showed similar initial high rapid uptake in baboon brain, peaking from 1 to 3.5 min (0.027-0.038%ID/cc) followed by rapid clearance (t(1/2)<15 min), resulting in low brain retention by 30 min. However, after 30 min, [2-methoxy-(11)C]GTS-21 continued to clear while [4-methoxy-(11)C]GTS-21 plateaued, suggesting the entry of a labeled metabolite into the brain. Comparison of the pharmacokinetics of the two labeled metabolites confirmed expected higher brain uptake and retention of [4-methoxy-(11)C]2-OH-GTS-21 (the labeled metabolite of [4-methoxy-(11)C]GTS-21) relative to [2-methoxy-(11)C]4-OH-GTS-21 (the labeled metabolite of [2-methoxy-(11)C]GTS-21), which had negligible brain uptake. Ex vivo studies in mice showed that GTS-21 is the major chemical form in the mouse brain. Whole-body dynamic PET imaging in baboon and mouse showed that the major route of excretion of C-11 is through the gallbladder. CONCLUSIONS: The major findings are as follows: (a) extremely rapid uptake and clearance of [2-methoxy-(11)C]GTS-21 from the brain, which may need to be considered in developing optimal dosing of GTS-21 for patients, and (b) significant brain uptake of 2-OH-GTS-21, suggesting that it might contribute to the therapeutic effects of GTS-21. This study illustrates the value of comparing different label positions and labeled metabolites to gain insight on the behavior of a central nervous system drug and its metabolites in the brain, providing an important perspective on drug pharmacokinetics.


Subject(s)
Benzylidene Compounds/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Positron-Emission Tomography/methods , Pyridines/pharmacokinetics , Receptors, Nicotinic/drug effects , Animals , Benzylidene Compounds/chemistry , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Female , Isotope Labeling/methods , Metabolic Clearance Rate , Nicotinic Agonists/chemical synthesis , Nicotinic Agonists/pharmacokinetics , Organ Specificity , Papio anubis , Pyridines/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...