Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Nucl Med ; 58(7): 1124-1131, 2017 07.
Article in English | MEDLINE | ID: mdl-27908967

ABSTRACT

18F-AV-1451 is currently the most widely used of several experimental tau PET tracers. The objective of this study was to evaluate 18F-AV-1451 binding with full kinetic analysis using a metabolite-corrected arterial input function and to compare parameters derived from kinetic analysis with SUV ratio (SUVR) calculated over different imaging time intervals. Methods:18F-AV-1451 PET brain imaging was completed in 16 subjects: 4 young healthy volunteers (YHV), 4 aged healthy volunteers (AHV), and 8 Alzheimer disease (AD) subjects. Subjects were imaged for 3.5 h, with arterial blood samples obtained throughout. PET data were analyzed using plasma and reference tissue-based methods to estimate the distribution volume, binding potential (BPND), and SUVR. BPND and SUVR were calculated using the cerebellar cortex as a reference region and were compared across the different methods and across the 3 groups (YHV, AHV, and AD). Results: AD demonstrated increased 18F-AV-1451 retention compared with YHV and AHV based on both invasive and noninvasive analyses in cortical regions in which paired helical filament tau accumulation is expected in AD. A correlation of R2 > 0.93 was found between BPND (130 min) and SUVR-1 at all time intervals. Cortical SUVR curves reached a relative plateau around 1.0-1.2 for YHV and AHV by approximately 50 min, but increased in AD by up to approximately 20% at 110-130 min and approximately 30% at 160-180 min relative to 80-100 min. Distribution volume (130 min) was lower by 30%-35% in the YHV than AHV. Conclusion: Our data suggest that although 18F-AV-1451 SUVR curves do not reach a plateau and are still increasing in AD, an SUVR calculated over an imaging window of 80-100 min (as currently used in clinical studies) provides estimates of paired helical filament tau burden in good correlation with BPND, whereas SUVR sensitivity to regional cerebral blood changes needs further investigation.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Carbolines/pharmacokinetics , Models, Biological , Positron-Emission Tomography , tau Proteins/metabolism , Aged , Alzheimer Disease/diagnostic imaging , Biomarkers/metabolism , Brain/diagnostic imaging , Computer Simulation , Female , Humans , Image Interpretation, Computer-Assisted , Kinetics , Male , Metabolic Clearance Rate , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reference Values , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
2.
Eur J Nucl Med Mol Imaging ; 44(3): 382-391, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27633250

ABSTRACT

PURPOSE: In Alzheimer's disease (AD), increased metabolism of monoamines by monoamine oxidase type B (MAO-B) leads to the production of toxic reactive oxygen species (ROS), which are thought to contribute to disease pathogenesis. Inhibition of the MAO-B enzyme may restore brain levels of monoaminergic neurotransmitters, reduce the formation of toxic ROS and reduce neuroinflammation (reactive astrocytosis), potentially leading to neuroprotection. Sembragiline (also referred as RO4602522, RG1577 and EVT 302 in previous communications) is a potent, selective and reversible inhibitor of MAO-B developed as a potential treatment for AD. METHODS: This study assessed the relationship between plasma concentration of sembragiline and brain MAO-B inhibition in patients with AD and in healthy elderly control (EC) subjects. Positron emission tomography (PET) scans using [11C]-L-deprenyl-D2 radiotracer were performed in ten patients with AD and six EC subjects, who received sembragiline each day for 6-15 days. RESULTS: At steady state, the relationship between sembragiline plasma concentration and MAO-B inhibition resulted in an Emax of ∼80-90 % across brain regions of interest and in an EC50 of 1-2 ng/mL. Data in patients with AD and EC subjects showed that near-maximal inhibition of brain MAO-B was achieved with 1 mg sembragiline daily, regardless of the population, whereas lower doses resulted in lower and variable brain MAO-B inhibition. CONCLUSIONS: This PET study confirmed that daily treatment of at least 1 mg sembragiline resulted in near-maximal inhibition of brain MAO-B enzyme in patients with AD.


Subject(s)
Acetamides/therapeutic use , Alzheimer Disease/diagnostic imaging , Monoamine Oxidase Inhibitors/pharmacokinetics , Positron-Emission Tomography , Pyrrolidinones/therapeutic use , Acetamides/blood , Acetamides/pharmacokinetics , Administration, Oral , Aged , Alzheimer Disease/drug therapy , Case-Control Studies , Female , Humans , Male , Middle Aged , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/administration & dosage , Monoamine Oxidase Inhibitors/therapeutic use , Protein Binding , Pyrrolidinones/blood , Pyrrolidinones/pharmacokinetics
3.
Eur J Nucl Med Mol Imaging ; 42(7): 1081-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833352

ABSTRACT

PURPOSE: Activated microglia play a key role in inflammatory demyelinating injury in multiple sclerosis (MS). Microglial activation can be measured in vivo using a positron emission tomography (PET) ligand (11)C-PBR28. We evaluated the test-retest variability (TRV) and lesion detectability of (11)C-PBR28 binding in MS subjects and healthy controls (HCs) with high-resolution PET. METHODS: Four clinically and radiologically stable relapsing-remitting MS subjects (age 41 ± 7 years, two men/two women) and four HCs (age 42 ± 8 years, 2 two men/two women), matched for translocator protein genotype [two high- and two medium-affinity binders according to DNA polymorphism (rs6971) in each group], were studied for TRV. Another MS subject (age 41 years, male) with clinical and radiological activity was studied for lesion detectability. Dynamic data were acquired over 120 min after injection of 634 ± 101 MBq (11)C-PBR28. For the TRV study, subjects were scanned twice, on average 1.4 weeks apart. Volume of distribution (V T) derived from multilinear analysis (MA1) modeling (t* = 30 min, using arterial input data) was the main outcome measure. RESULTS: Mean test V T values (ml cm(-3)) were 3.9 ± 1.4 in the whole brain gray matter (GM), 3.6 ± 1.2 in the whole brain white matter (WM) or normal-appearing white matter (NAWM), and 3.3 ± 0.6 in MS WM lesions; mean retest V T values were 3.7 ± 1.0 in GM, 3.3 ± 0.9 in WM/NAWM, and 3.3 ± 0.7 in MS lesions. Test-retest results showed a mean absolute TRV ranging from 7 to 9 % across GM, WM/NAWM, and MS lesions. High-affinity binders demonstrated 30 % higher V T than medium-affinity binders in GM. Focal (11)C-PBR28 uptake was detected in two enhancing lesions of the active MS patient. CONCLUSION: High-resolution (11)C-PBR28 PET can visualize focal areas where microglial activation is known to be present and has good test-retest reproducibility in the human brain. (11)C-PBR28 PET is likely to be valuable for monitoring both MS disease evolution and response to therapeutic strategies that target microglial activation.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Positron-Emission Tomography , Pyrimidines , Radiopharmaceuticals , White Matter/diagnostic imaging , Adult , Case-Control Studies , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/genetics , Receptors, GABA/genetics , Reproducibility of Results , White Matter/pathology
4.
Ann Clin Transl Neurol ; 1(9): 659-69, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25493280

ABSTRACT

OBJECTIVE: The mechanism of action of anti-B cell therapy in multiple sclerosis (MS) is not fully understood. Here, we compared the effect of anti-CD20 therapy on microglial activation in two distinct focal rat models of MS. METHODS: The effect of anti-CD20 therapy on lesion formation and extralesional microglial activation was evaluated in the fDTH-EAE (experimental allergic encephalomyelitis) model, which is a focal demyelinating type-IV delayed-type hypersensitivity lesion. For comparison, effects were also assessed in the focal humoral MOG model induced by intracerebral injection of cytokine in myelin oligodendrocyte glycoprotein immunized rats. Microglial activation was assessed in situ and in vivo using the TSPO SPECT ligand [(125)I]DPA-713, and by immunostaining for MHCII. The effect of treatment on demyelination and lymphocyte recruitment to the brain were evaluated. RESULTS: Anti-CD20 therapy reduced microglial activation, and lesion formation in the humoral model, but it was most effective in the antibody-independent fDTH-EAE. Immunohistochemistry for MHCII also demonstrated a reduced volume of microglial activation in the brains of anti-CD20-treated fDTH-EAE animals, which was accompanied by a reduction in T-cell recruitment and demyelination. The effect anti-CD20 therapy in the latter model was similarly strong as compared to the T-cell targeting MS compound FTY720. INTERPRETATION: The suppression of lesion development by anti-CD20 treatment in an antibody-independent model suggests that B-cells play an important role in lesion development, independent of auto-antibody production. Thus, CD20-positive B-cell depletion has the potential to be effective in a wider population of individuals with MS than might have been predicted from our knowledge of the underlying histopathology.

5.
Nucl Med Biol ; 40(4): 547-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23541825

ABSTRACT

INTRODUCTION: The aims of the present positron emission tomography (PET) study were to set up a system for (11)C-cyanation labeling of the selective mGluR5-antagonist [(11)C]AZD9272 and to perform the first in vivo characterization of [(11)C]AZD9272 binding in cynomolgus monkeys. METHODS: [(11)C]AZD9272 was labeled using palladium mediated (11)C-cyanation. Altogether seven PET measurements were performed in three cynomolgus monkeys including baseline and co-injection experiments with unlabelled AZD9272 (0.04 and 0.4 mg/kg). Radiometabolites in plasma were measured using HPLC. RESULTS: [(11)C]AZD9272 was prepared in over 50% incorporation yield from hydrogen [(11)C]cyanide in a total synthesis time of 45-50 min. The radiochemical purity of the radioligand in its final formulation was high (>99%) and the mean specific radioactivity was 47 GBq/ µmol (1278 Ci/mmol, n=7) calculated at end of bombardment (EOB). In the baseline measurements 10% of the total injected radioactivity was present in monkey brain at five minutes after i.v. injection. The radioactivity concentration was high in the caudate, cingulate gyrus and thalamus whereas it was moderate in the temporal cortex and lower for the cerebellum. After co-injection with cold AZD9272 the binding of [(11)C]AZD9272 was reduced in a dose-dependent fashion. Analysis of radiometabolites showed relatively slow metabolism and resulted only in hydrophilic radiometabolites. CONCLUSION: A fast and efficient method was developed to label AZD9272 with (11)C. PET-examination in Cynomolgus monkeys showed that [(11)C]AZD9272 entered the brain to a high extent, that binding was saturable and that the regional radioactivity pattern was in accordance with the known distribution of mGluR5. The results support further examination of [(11)C]AZD9272 binding in human subjects.


Subject(s)
Brain/metabolism , Nitriles/chemistry , Oxadiazoles/chemistry , Palladium/chemistry , Pyridines/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Biological Transport , Brain/diagnostic imaging , Carbon Radioisotopes , Catalysis , Chromatography, High Pressure Liquid , Feasibility Studies , Injections , Ligands , Macaca fascicularis , Oxadiazoles/metabolism , Oxadiazoles/pharmacology , Positron-Emission Tomography , Pyridines/metabolism , Pyridines/pharmacology , Radiochemistry , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
6.
Eur J Nucl Med Mol Imaging ; 40(3): 403-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23229747

ABSTRACT

PURPOSE: This study evaluated the potential of functional imaging to monitor disease activity and response to treatment with disease-modifying antirheumatic drugs (DMARD) in DMARD-naive patients with early rheumatoid arthritis (RA). METHODS: The study involved 17 patients with active RA in whom combination therapy was initiated with methotrexate, sulfasalazine, hydroxychloroquine, and low-dose oral prednisolone. Clinical disease activity was assessed at screening, at baseline and after 2, 4, 8 and 12 weeks of therapy. (18)F-FDG PET/CT of all joints was performed at baseline and after 2 and 4 weeks of therapy. RESULTS: (18)F-FDG maximum standardized uptake values showed a reduction of 22 ± 13 % in 76 % of patients from baseline to week 2 and a reduction of 29 ± 13 % in 81 % of patients from baseline to week 4. The percentage decrease in (18)F-FDG uptake from baseline to week 2 correlated with clinical outcome, as measured by the disease activity score (DAS-28) at week 12. In addition, changes in C-reactive protein levels and erythrocyte sedimentation rate were positively associated with changes shown by PET. CONCLUSION: (18)F-FDG PET/CT findings after 2 and 4 weeks of triple combination oral DMARD therapy correlated with treatment efficacy and clinical outcome in patients with early RA. (18)F-FDG PET/CT may help predict the therapeutic response to novel drug treatments.


Subject(s)
Antirheumatic Agents/administration & dosage , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/drug therapy , Fluorodeoxyglucose F18 , Multimodal Imaging , Positron-Emission Tomography , Tomography, X-Ray Computed , Administration, Oral , Biomarkers/metabolism , Drug Combinations , Female , Humans , Male , Middle Aged , Time Factors , Treatment Outcome
7.
Psychopharmacology (Berl) ; 218(3): 579-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21625907

ABSTRACT

RATIONALE: Cariprazine is a novel antipsychotic drug candidate that exhibits high selectivity and affinity to dopamine D(3) and D(2) receptors and moderate affinity to serotonin 5-HT(1A) receptors. Targeting receptors other than D(2) may provide a therapeutic benefit for both positive and negative symptoms associated with schizophrenia. Positron emission tomography (PET) can be used as a tool in drug development to assess the in vivo distribution and pharmacological properties of a drug. OBJECTIVES: The objective of this study was to determine dopamine D(2)/D(3) and serotonin 5-HT(1A) receptor occupancy in monkey brain after the administration of cariprazine. METHODS: We examined three monkeys using the following PET radioligands: [(11)C]MNPA (an agonist at D(2) and D(3) receptors), [(11)C]raclopride (an antagonist at D(2) and D(3) receptors), and [(11)C]WAY-100635 (an antagonist at 5-HT(1A) receptors). During each experimental day, the first PET measurement was a baseline study, the second after a low dose of cariprazine, and the third after the administration of a high dose. RESULTS: We found that cariprazine occupied D(2)/D(3) receptors in a dose-dependent and saturable manner, with the lowest dose occupying ~5% of receptors and the highest dose showing more than 90% occupancy. 5-HT(1A) receptor occupancy was considerably lower compared with D(2)/D(3) occupancy at the same doses, with a maximal value of ~30% for the raphe nuclei. CONCLUSIONS: We conclude that cariprazine binds preferentially to dopamine D(2)/D(3) rather than to serotonin 5-HT(1A) receptors in monkey brain. These findings can be used to guide the selection of cariprazine dosing in humans.


Subject(s)
Antipsychotic Agents/metabolism , Piperazines/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacology , Brain/metabolism , Dose-Response Relationship, Drug , Macaca fascicularis , Piperazines/administration & dosage , Piperazines/pharmacology , Positron-Emission Tomography , Protein Binding , Receptor, Serotonin, 5-HT1A/metabolism
8.
J Nucl Med ; 51(4): 559-66, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20237038

ABSTRACT

UNLABELLED: Permeability-glycoprotein (P-gp), an efflux transporter in several organs, acts at the blood-brain barrier to protect the brain from exogenous toxins. P-gp almost completely blocks brain entry of the PET radiotracer (11)C-N-desmethyl-loperamide ((11)C-dLop). We examined the ability of (11)C-dLop to quantify P-gp function in humans after increasing doses of tariquidar, an inhibitor of P-gp. METHODS: Seventeen healthy volunteers had a total of 23 PET scans with (11)C-dLop at baseline and after increasing doses of tariquidar (2, 4, and 6 mg/kg intravenously). A subset of subjects received PET with (15)O-H(2)O to measure cerebral blood flow. Brain uptake of (11)C-dLop was quantified in 2 ways. Without blood data, uptake was measured as area under the time-activity curve in the brain from 10 to 30 min (AUC(10-30)). With arterial blood data, brain uptake was quantified with compartmental modeling to estimate the rates of entry into (K(1)) and efflux from (k(2)) the brain. RESULTS: Brain uptake of radioactivity was negligible at baseline and increased only slightly (approximately 30%) after 2 mg of tariquidar per kilogram. In contrast, 4 and 6 mg of tariquidar per kilogram increased brain uptake 2- and 4-fold, respectively. Greater brain uptake reflected greater brain entry (K(1)), because efflux (k(2)) and cerebral blood flow did not differ between tariquidar-treated and untreated subjects. In the subjects who received the highest dose of tariquidar (and had the highest brain uptake), regional values of K(1) correlated linearly with absolute cerebral blood flow, consistent with high single-pass extraction of (11)C-dLop. AUC(10-30) correlated linearly with K(1). CONCLUSION: P-gp function at the blood-brain barrier in humans can be quantified using PET and (11)C-dLop. A simple measure of brain uptake (AUC(10-30)) may be used as a surrogate of the fully quantified rate constant for brain entry (K(1)) and thereby avoid arterial sampling. However, to dissect the function of P-gp itself, both brain uptake and the influx rate constant must be corrected for radiotracer delivery (blood flow).


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Blood-Brain Barrier/metabolism , Loperamide/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Adult , Biological Transport/drug effects , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Cerebrovascular Circulation/drug effects , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Kinetics , Loperamide/metabolism , Magnetic Resonance Imaging , Male , Positron-Emission Tomography , Quinolines/pharmacology , Radioactive Tracers
9.
Nucl Med Biol ; 37(3): 335-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20346873

ABSTRACT

INTRODUCTION: [(11)C]Loperamide and [(11)C]N-desmethyl-loperamide ([(11)C]dLop) have been proposed as radiotracers for imaging brain P-glycoprotein (P-gp) function. A major route of [(11)C]loperamide metabolism is N-demethylation to [(11)C]dLop. We aimed to test whether inhibition of CYP3A4 with ketoconazole might reduce the metabolism of [(11)C]loperamide and [(11)C]dLop in mice, and thereby improve the quality of these radiotracers. METHODS: Studies were performed in wild-type and P-gp knockout (mdr-1a/b -/-) mice. During each of seven study sessions, one pair of mice, comprising one wild-type and one knockout mouse, was pretreated with ketoconazole (50 mg/kg, ip), while another such pair was left untreated. Mice were sacrificed at 30 min after injection of [(11)C]loperamide or [(11)C]dLop. Whole brain and plasma samples were measured for radioactivity and analyzed with radio-high-performance liquid chromatography. RESULTS: Ketoconazole increased the plasma concentrations of [(11)C]loperamide and its main radiometabolite, [(11)C]dLop, by about twofold in both wild-type and knockout mice, whereas the most polar radiometabolite was decreased threefold. Furthermore, ketoconazole increased the brain concentrations of [(11)C]loperamide and the radiometabolite [(11)C]dLop by about twofold in knockout mice, and decreased the brain concentrations of the major and most polar radiometabolite in wild-type and knockout mice by 82% and 49%, respectively. In contrast, ketoconazole had no effect on plasma and brain distribution of administered [(11)C]dLop and its radiometabolites in either wild-type or knockout mice, except to increase the low plasma [(11)C]dLop concentration. The least polar radiometabolite of [(11)C]dLop was identified with LC-MS(n) as the N-hydroxymethyl analog of [(11)C]dLop and this also behaved as a P-gp substrate. CONCLUSION: In this study, ketoconazole (50 mg/kg, ip) proved partially effective for inhibiting the N-demethylation of [(11)C]loperamide in mouse in vivo but had relatively smaller or no effect on [(11)C]dLop.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/diagnostic imaging , Brain/metabolism , Ketoconazole/administration & dosage , Loperamide/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Brain/drug effects , Carbon Radioisotopes/pharmacokinetics , Loperamide/analogs & derivatives , Metabolic Clearance Rate/drug effects , Mice , Mice, Knockout , Organ Specificity/drug effects , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution/drug effects
10.
Eur J Nucl Med Mol Imaging ; 37(8): 1499-506, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20333514

ABSTRACT

PURPOSE: Cannabinoid subtype 1 (CB(1)) receptors are found in nearly every organ in the body, may be involved in several neuropsychiatric and metabolic disorders, and are therefore an active target for pharmacotherapy and biomarker development. We recently reported brain imaging of CB(1) receptors with two PET radioligands: (11)C-MePPEP and (18)F-FMPEP-d (2). Here we describe the biodistribution and dosimetry estimates for these two radioligands. METHODS: Seven healthy subjects (four men and three women) underwent whole-body PET scans for 120 min after injection with (11)C-MePPEP. Another seven healthy subjects (two men and five women) underwent whole-body PET scans for 300 min after injection with (18)F-FMPEP-d (2). Residence times were acquired from regions of interest drawn on tomographic images of visually identifiable organs for both radioligands and from radioactivity excreted in urine for (18)F-FMPEP-d (2). RESULTS: The effective doses of (11)C-MePPEP and (18)F-FMPEP-d (2) are 4.6 and 19.7 microSv/MBq, respectively. Both radioligands demonstrated high uptake of radioactivity in liver, lung, and brain shortly after injection and accumulated radioactivity in bone marrow towards the end of the scan. After injection of (11)C-MePPEP, radioactivity apparently underwent hepatobiliary excretion only, while radioactivity from (18)F-FMPEP-d (2) showed both hepatobiliary and urinary excretion. CONCLUSION: (11)C-MePPEP and (18)F-FMPEP-d (2) yield an effective dose similar to other PET radioligands labeled with either (11)C or (18)F. The high uptake in brain confirms the utility of these two radioligands to image CB(1) receptors in brain, and both may also be useful to image CB(1) receptors in the periphery.


Subject(s)
Drug Inverse Agonism , Positron-Emission Tomography/methods , Pyrrolidinones/pharmacology , Pyrrolidinones/pharmacokinetics , Receptor, Cannabinoid, CB1/metabolism , Adult , Brain/diagnostic imaging , Brain/metabolism , Female , Humans , Male , Radiometry
11.
Synapse ; 64(9): 699-703, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20336622

ABSTRACT

In vitro, D(2) dopamine receptors (DAR) can exist in low- and high-affinity states for agonists and increases of D(2) receptors in high-affinity state have been proposed to underlie DA receptor supersensitivity in vivo. Deletion of the gene for dopamine beta-hydroxylase (DBH) causes mice to become hypersensitive to the effects of psychostimulants, and in vitro radioligand binding results suggest an increased percentage of D(2) receptors in a high-affinity state. To determine whether DBH knockout mice display an increase of high-affinity state D(2) receptors in vivo, we scanned DBH knockout and control mice with the agonist PET radioligand [(11)C]MNPA, which is thought to bind preferentially to the high-affinity state of the D(2) receptor. In addition, we performed in vitro binding experiments on striatal homogenates with [(3)H]methylspiperone to measure B(max) values and the percentages of high- and low-affinity states of the D(2) receptor. We found that the in vivo striatal binding of [(11)C]MNPA was similar in DBH knockout mice and heterozygous controls and the in vitro B(max) values and percentages of D(2) receptors in the high-affinity state, were not significantly different between these two groups. In summary, our results suggest that DBH knockout mice have normal levels of D(2) receptors in the high-affinity state and that additional mechanisms contribute to their behavioral sensitivity to psychostimulants.


Subject(s)
Dopamine beta-Hydroxylase/deficiency , Receptors, Dopamine D2/metabolism , Animals , Apomorphine/analogs & derivatives , Binding, Competitive/drug effects , Cerebellum/diagnostic imaging , Cerebellum/drug effects , Cerebellum/metabolism , Dopamine Agonists , Dopamine beta-Hydroxylase/genetics , Female , Kinetics , Male , Mice , Mice, Knockout , Neostriatum/diagnostic imaging , Neostriatum/drug effects , Neostriatum/metabolism , Positron-Emission Tomography , Radioligand Assay , Radiopharmaceuticals , Spiperone/analogs & derivatives
12.
Neuroimage ; 50(4): 1402-7, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20097293

ABSTRACT

Dopamine released by amphetamine decreases the in vivo binding of PET radioligands to the dopamine D(2) receptor. Although concentrations of extracellular dopamine largely return to baseline within 1 to 2 h after amphetamine treatment, radioligand binding remains decreased for several hours. The purpose of this study was to determine whether the prolonged decrease of radioligand binding after amphetamine administration is caused by receptor internalization. To distinguish dopamine displacement from receptor internalization, we used wild-type and arrestin3 (arr3) knockout mice, which are incapable of internalizing D(2) receptors. In addition, we used both the D(2) selective agonist [(11)C]MNPA (which is thought to bind to the high affinity state of the receptor) and the D(2) selective antagonist [(18)F]fallypride (which does not differentiate between high and low affinity state). After an initial baseline scan, animals were divided in three groups for a second scan: either 30 min or 4 h after amphetamine administration (3 mg/kg, i.p.) or as retest. At 30 min, [(11)C]MNPA showed greater displacement than [(18)F]fallypride, but each radioligand gave similar displacement in knockout and wild-type mice. At 4 h, the binding of both radioligands returned to baseline in arr3 knockout mice, but remained decreased in wild-type mice. Radioligand binding was unaltered on retest scanning. Our results suggest that the prolonged decrease of radioligand binding after amphetamine is mainly due to internalization of the D(2) receptor rather than dopamine displacement. In addition, this study demonstrates the utility of small animal PET to study receptor trafficking in vivo in genetically modified mice.


Subject(s)
Amphetamine/pharmacology , Brain/drug effects , Cell Membrane/drug effects , Dopamine Agents/pharmacology , Radiopharmaceuticals/metabolism , Receptors, Dopamine D2/metabolism , Animals , Arrestins/deficiency , Arrestins/genetics , Arrestins/metabolism , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes/metabolism , Cell Membrane/diagnostic imaging , Cell Membrane/metabolism , Cerebellum/diagnostic imaging , Cerebellum/drug effects , Cerebellum/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Female , Fluorine Radioisotopes/metabolism , Male , Mice , Mice, Knockout , Naproxen/pharmacokinetics , Positron-Emission Tomography , Time Factors
13.
J Nucl Med ; 50(5): 807-13, 2009 May.
Article in English | MEDLINE | ID: mdl-19372478

ABSTRACT

UNLABELLED: P-glycoprotein (P-gp) is a membrane-bound efflux pump that limits the distribution of drugs to several organs of the body. At the blood-brain barrier, P-gp blocks the entry of both loperamide and its metabolite, N-desmethyl-loperamide (N-dLop), and thereby prevents central opiate effects. Animal studies have shown that (11)C-dLop, compared with (11)C-loperamide, is an especially promising radiotracer because it generates negligible radiometabolites that enter the brain. The purposes of this study were to determine whether (11)C-dLop is a substrate for P-gp at the blood-brain barrier in humans and to measure the distribution of radioactivity in the entire body to estimate radiation exposure. METHODS: Brain PET scans were acquired in 4 healthy subjects for 90 min and included concurrent measurements of the plasma concentration of unchanged radiotracer. Time-activity data from the whole brain were quantified using a 1-tissue-compartment model to estimate the rate of entry (K(1)) of radiotracer into the brain. Whole-body PET scans were acquired in 8 healthy subjects for 120 min. RESULTS: For brain imaging, after the injection of (11)C-dLop the concentration of radioactivity in the brain was low (standardized uptake value, approximately 15%) and stable after approximately 20 min. In contrast, uptake of radioactivity in the pituitary was about 50-fold higher than that in the brain. The plasma concentration of (11)C-dLop declined rapidly, but the percentage composition of plasma was unusually stable, with the parent radiotracer constituting 85% of total radioactivity after approximately 5 min. The rate of brain entry was low (K(1) = 0.009 +/- 0.002 mL.cm(-3).min(-1); n = 4). For whole-body imaging, as a measure of radiation exposure to the entire body the effective dose of (11)C-dLop was 7.8 +/- 0.6 muSv/MBq (n = 8). CONCLUSION: The low brain uptake of radioactivity is consistent with (11)C-dLop being a substrate for P-gp in humans and confirms that this radiotracer generates negligible quantities of brain-penetrant radiometabolites. In addition, the low rate of K(1) is consistent with P-gp rapidly effluxing substrates while they transit through the lipid bilayer. The radiation exposure of (11)C-dLop is similar to that of many other (11)C-radiotracers. Thus, (11)C-dLop is a promising radiotracer to study the function of P-gp at the blood-brain barrier, at which impaired function would allow increased uptake into the brain.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Body Burden , Brain/diagnostic imaging , Brain/metabolism , Loperamide/analogs & derivatives , Positron-Emission Tomography/methods , Adult , Female , Humans , Loperamide/pharmacokinetics , Male , Metabolic Clearance Rate , Organ Specificity , Radiation Dosage , Radiometry , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
14.
J Neurosci ; 29(6): 1887-96, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19211895

ABSTRACT

Aberrant neurotransmissions via glutamate and dopamine receptors have been the focus of biomedical research on the molecular basis of psychiatric disorders, but the mode of their interaction is yet to be uncovered. In this study, we demonstrated the pharmacological reversal of methamphetamine-stimulated dopaminergic overflow by suppression of group I metabotropic glutamate (mGlu) receptor in living primates and rodents. In vivo positron emission tomography (PET) was conducted on cynomolgus monkeys and rats using a full agonistic tracer for dopamine D(2/3) receptor, [(11)C]MNPA [(R)-2-(11)CH(3)O-N-n-propylnorapomorphine], and fluctuation of kinetic data resulting from anesthesia was avoided by scanning awake subjects. Excessive release of dopamine induced by methamphetamine and abolishment of this alteration by treatment with an antagonist of group I mGlu receptors, 2-methyl-6-(phenylethynyl)pyridine (MPEP), were measured in both species as decreased binding potential because of increased dopamine and its recovery to baseline levels, respectively. Counteraction of MPEP to the methamphetamine-induced dopamine spillover was also supported neurochemically by microdialysis of unanesthetized rat striatum. Moreover, patch-clamp electrophysiological assays using acute brain slices prepared from rats indicated that direct targets of MPEP mechanistically involved in the effects of methamphetamine are present locally within the striatum. Because MPEP alone did not markedly alter the baseline dopaminergic neurotransmission according to our PET and electrophysiological data, the present findings collectively extend the insights on dopamine-glutamate cross talk from extrastriatal localization of responsible mGlu receptors to intrastriatal synergy and support therapeutic interventions in case of disordered striatal dopaminergic status using group I mGlu receptor antagonists assessable by in vivo imaging techniques.


Subject(s)
Corpus Striatum/diagnostic imaging , Corpus Striatum/physiology , Dopamine/physiology , Glutamic Acid/physiology , Positron-Emission Tomography , Synaptic Transmission/physiology , Animals , Macaca , Male , Positron-Emission Tomography/methods , Rats , Rats, Sprague-Dawley
15.
J Nucl Med ; 50(1): 108-15, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19091890

ABSTRACT

UNLABELLED: 11C-Loperamide is an avid substrate for P-glycoprotein (P-gp), but it is rapidly metabolized to 11C-N-desmethyl-loperamide (11C-dLop), which is also a substrate for P-gp and thereby contaminates the radioactive signal in the brain. Should further demethylation of 11C-dLop occur, radiometabolites with low entry into the brain are generated. Therefore, we evaluated the ability of 11C-dLop to quantify the function of P-gp at the blood-brain barrier in monkeys. METHODS: Six monkeys underwent 12 PET scans of the brain, 5 at baseline and 7 after pharmacologic blockade of P-gp. A subset of monkeys also underwent PET scans with 15O-water to measure cerebral blood flow. To determine whether P-gp blockade affected peripheral distribution of 11C-dLop, we measured whole-body biodistribution in 4 monkeys at baseline and after P-gp blockade. RESULTS: The concentration of 11C-dLop in the brain was low under baseline conditions and increased 5-fold after P-gp blockade. This increase was primarily caused by an increased rate of entry into the brain rather than a decreased rate of removal from the brain. With P-gp blockade, uptake of radioactivity among brain regions correlated linearly with blood flow, suggesting a high single-pass extraction. After correction for cerebral blood flow, the uptake of 11C-dLop was fairly uniform among brain regions, suggesting that the function of P-gp is fairly uniformly distributed in the brain. On whole-body imaging, P-gp blockade significantly affected distribution of radioactivity only to the brain and not to other visually identified source organs. The effective dose estimated for humans was approximately 9 microSv/MBq. CONCLUSION: PET with 11C-dLop can quantify P-gp function at the blood-brain barrier in monkeys. The single-pass extraction of 11C-dLop is high and requires correction for blood flow to accurately measure the function of this efflux transporter. The low uptake at baseline and markedly increased uptake after P-gp blockade suggest that 11C-dLop will be useful to measure a wide range of P-gp functions at the blood-brain barrier in humans.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Loperamide/analogs & derivatives , Macaca mulatta/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Blood-Brain Barrier/physiology , Humans , Loperamide/metabolism , Loperamide/pharmacokinetics , Macaca mulatta/physiology , Male , Positron-Emission Tomography , Radioactivity , Regional Blood Flow , Tissue Distribution
16.
J Med Chem ; 51(19): 6034-43, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18783208

ABSTRACT

[(11)C]Loperamide has been proposed for imaging P-glycoprotein (P-gp) function with positron emission tomography (PET), but its metabolism to [N-methyl-(11)C] N-desmethyl-loperamide ([(11)C]dLop; [(11)C]3) precludes quantification. We considered that [(11)C]3 might itself be a superior radiotracer for imaging brain P-gp function and therefore aimed to prepare [(11)C]3 and characterize its efficacy. An amide precursor (2) was synthesized and methylated with [(11)C]iodomethane to give [(11)C]3. After administration of [(11)C]3 to wild-type mice, brain radioactivity uptake was very low. In P-gp (mdr-1a(-/-)) knockout mice, brain uptake of radioactivity at 30 min increased about 3.5-fold by PET measures, and over 7-fold by ex vivo measures. In knockout mice, brain radioactivity was predominantly (90%) unchanged radiotracer. In monkey PET experiments, brain radioactivity uptake was also very low but after P-gp blockade increased more than 7-fold. [(11)C]3 is an effective new radiotracer for imaging brain P-gp function and, in favor of future successful quantification, appears free of extensive brain-penetrant radiometabolites.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Brain/metabolism , Loperamide/analogs & derivatives , Loperamide/chemical synthesis , Loperamide/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Brain/diagnostic imaging , Carbon Radioisotopes , Injections, Intravenous , Loperamide/chemistry , Macaca mulatta , Male , Mice , Mice, Knockout , Molecular Structure , Positron-Emission Tomography/methods , Radiopharmaceuticals/chemistry , Stereoisomerism , Time Factors
17.
Synapse ; 62(10): 756-63, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18651641

ABSTRACT

Estimates of dopamine D(2/3) receptor occupancy by endogenous dopamine using positron emission tomography (PET) in animals have varied almost threefold. This variability may have been caused by incomplete depletion of dopamine or by the use of antagonist radioligands, which appear less sensitive than agonist radioligands to changes in endogenous dopamine. PET scans were performed in rats with the agonist PET radioligand [(11)C]MNPA ([O-methyl-(11)C]2-methoxy-N-propylnorapomorphine). [(11)C]MNPA was injected as a bolus plus constant infusion to achieve steady-state concentration in the body and equilibrium receptor binding in the brain. Radioligand binding was compared at baseline and after treatment with reserpine plus alpha-methyl-para-tyrosine, which cause approximately 95% depletion of endogenous dopamine. Depletion of dopamine increased radioligand binding in striatum but had little effect in cerebellum. Striatal [(11)C]MNPA binding potential was 0.93 +/- 0.12 at baseline and increased to 1.99 +/- 0.25 after dopamine depletion. Occupancy of D(2/3) receptors by endogenous dopamine at baseline was calculated to be approximately 53%. Striatal binding was displaceable with raclopride, but not with BP 897 (a selective D(3) compound), thus confirming the D(2) receptor specificity of [(11)C]MNPA binding. Radioactivity extracted from rat brain contained only 8-10% radiometabolites and was insignificantly altered by administration of reserpine plus alpha-methyl-para-tyrosine. Hence, dopamine depletion did not increase the PET measurements via an effect on radiotracer metabolism. Our in vivo estimate of dopamine's occupancy of D(2/3) receptors at baseline is higher than that previously reported using antagonist radioligands and PET, but is similar to that reported using agonist radioligands and ex vivo measurements.


Subject(s)
Apomorphine/analogs & derivatives , Brain/diagnostic imaging , Brain/metabolism , Dopamine Agonists/metabolism , Dopamine/metabolism , Positron-Emission Tomography/methods , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Animals , Apomorphine/metabolism , Brain/drug effects , Carbon Radioisotopes/metabolism , Dopamine/physiology , Dopamine Agonists/pharmacology , Male , Protein Binding , Radiopharmaceuticals/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/agonists
18.
Synapse ; 62(9): 700-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18566975

ABSTRACT

With a view to future extension of the use of the agonist radioligand [(11)C]MNPA ([O-methyl-(11)C]2-methoxy-N-propylnorapomorphine) from animals to humans, we performed two positron emission tomography (PET) studies in monkeys. First, we assessed the ability to quantify the brain uptake of [(11)C]MNPA with compartmental modeling. Second, we estimated the radiation exposure of [(11)C]MNPA to human subjects based on whole-body imaging in monkeys. Brain PET scans were acquired for 90 min and included concurrent measurements of the plasma concentration of unchanged radioligand. Time-activity data from striatum and cerebellum were quantified with two methods, a reference tissue model and distribution volume. Whole-body PET scans were acquired for 120 min using four bed positions from head to mid thigh. Regions of interest were drawn on compressed planar whole-body images to identify organs with the highest radiation exposures. After injection of [(11)C]MNPA, the highest concentration of radioactivity in brain was in striatum, with lowest levels in cerebellum. Distribution volume was well identified with a two-tissue compartmental model and was quite stable from 60 to 90 min. Whole-body PET scans showed the organ with the highest radiation burden (muSv/MBq) was the urinary bladder wall (26.0), followed by lungs (22.5), gallbladder wall (21.9), and heart wall (16.1). With a 2.4-h voiding interval, the effective dose was 6.4 muSv/MBq (23.5 mrem/mCi). In conclusion, brain uptake of [(11)C]MNPA reflected the density of D(2/3) receptors, quantified relative to serial arterial measurements, and caused moderate to low radiation exposure.


Subject(s)
Brain/diagnostic imaging , Naproxen , Positron-Emission Tomography/methods , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Animals , Brain/metabolism , Carbon Radioisotopes/blood , Carbon Radioisotopes/pharmacokinetics , Macaca mulatta , Magnetic Resonance Imaging , Male , Naproxen/blood , Naproxen/pharmacokinetics , Radiometry , Tissue Distribution , Whole Body Imaging
19.
Neuroimage ; 41(3): 1075-85, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18434202

ABSTRACT

The 5-HT1B receptor has been implicated in several psychiatric disorders and is a potential pharmacological target in the treatment of depression. Here we report the synthesis of a novel PET radioligand, [11C]AZ10419369 (5-methyl-8-(4-methyl-piperazin-1-yl)-4-oxo-4H-chromene-2-carboxylic acid (4-morpholin-4-yl-phenyl)-amide), for in vivo visualization of 5-HT1B receptors in the brains of macaques and humans subjects. [11C]AZ10419369 was prepared by N-methylation of (8-(1-piperazinyl)-5-methylchrom-2-en-4-one-2-(4-morpholinophenyl) carboxamide, using carbon-11 methyl triflate. Regional brain uptake patterns of [11C]AZ10419369 were characterized by PET measurements in two macaques and a preliminary study in two human subjects. In addition, AZ10419369 was prepared in tritium labeled form for in vitro autoradiography studies in macaque brain tissue sections. The radiochemical purity of [11C]AZ10419369 was >99% and specific radioactivity was >3600 Ci/mmol. After iv injection of [11C]AZ10419369, 3-4% was in brain after 7.5 min. The regional brain distribution of radioactivity was similar in humans and macaques showing the highest uptake of radioactivity in the occipital cortex and the basal ganglia, in accord with autoradiographic studies performed using [3H]AZ10419369. Uptake was moderate in the temporal and frontal cortical regions, lower in the thalamus and lowest in the cerebellum. In macaques pre-treated with the selective 5-HT1B receptor antagonist, AR-A000002, binding was reduced in a dose-dependent manner, consistent with specific binding to 5-HT1B receptors. These data support [11C]AZ10419369 as a suitable radioligand for labeling 5-HT1B receptors in the primate brain. This radioligand may be useful in future studies evaluating drug-induced receptor occupancy and measurement of brain 5-HT1B receptor levels in patients with psychiatric disorders.


Subject(s)
Benzopyrans/pharmacokinetics , Brain/metabolism , Morpholines/pharmacokinetics , Piperazines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptor, Serotonin, 5-HT1B/metabolism , Animals , Autoradiography , Benzopyrans/chemical synthesis , Carbon Radioisotopes/pharmacokinetics , Female , Humans , Macaca , Morpholines/chemical synthesis , Piperazines/chemical synthesis , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis
20.
J Med Chem ; 51(1): 148-58, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18078311

ABSTRACT

6-Thiolato-substituted 2-(4'- N,N-dimethylamino)phenylimidazo[1,2- a]pyridines ( RS-IMPYs; 1- 4) were synthesized as candidates for labeling with carbon-11 ( t 1/2 = 20.4 min) and imaging of A beta plaques in living human brain using positron emission tomography (PET). K i values for binding of these ligands to Alzheimer's disease brain homogenates were measured in vitro against tritium-labeled 6 (Pittsburgh compound B). MeS-IMPY ( 3, K i = 7.93 nM) was labeled with carbon-11 at its S- or N-methyl position to give [ (11)C] 7 or [ (11)C] 8, respectively. After injection into rats, [ (11)C] 7 or [ (11)C] 8 gave moderately high brain uptakes of radioactivity followed by rapid washout to low levels. The ratio of radioactivity at maximal uptake to that at 60 min reached 18.7 for [ (11)C] 7. [ (11)C] 7 behaved similarly in mouse and monkey. [ (11)C] 7 also bound selectively to A beta plaques in post mortem human Alzheimer's disease brain. Although rapidly metabolized in rat by N-demethylation, [ (11)C] 7 was stable in rat brain homogenates. The ex vivo brain radiometabolites observed in rats have a peripheral origin. Overall, [ (11)C] 7 merits further evaluation in human subjects.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Imidazoles/chemical synthesis , Pyridines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Alzheimer Disease/diagnostic imaging , Animals , Autoradiography , Brain/diagnostic imaging , Carbon Radioisotopes , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Macaca mulatta , Male , Mice , Positron-Emission Tomography , Pyridines/chemistry , Pyridines/pharmacokinetics , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...