Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Psychiatry ; 28(8): 3384-3390, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532797

ABSTRACT

In humans, the negative effects of alcohol are linked to immune dysfunction in both the periphery and the brain. Yet acute effects of alcohol on the neuroimmune system and its relationships with peripheral immune function are not fully understood. To address this gap, immune response to an alcohol challenge was measured with positron emission tomography (PET) using the radiotracer [11C]PBR28, which targets the 18-kDa translocator protein, a marker sensitive to immune challenges. Participants (n = 12; 5 F; 25-45 years) who reported consuming binge levels of alcohol (>3 drinks for females; >4 drinks for males) 1-3 months before scan day were enrolled. Imaging featured a baseline [11C]PBR28 scan followed by an oral laboratory alcohol challenge over 90 min. An hour later, a second [11C]PBR28 scan was acquired. Dynamic PET data were acquired for at least 90 min with arterial blood sampling to measure the metabolite-corrected input function. [11C]PBR28 volume of distributions (VT) was estimated in the brain using multilinear analysis 1. Subjective effects, blood alcohol levels (BAL), and plasma cytokines were measured during the paradigm. Full completion of the alcohol challenge and data acquisition occurred for n = 8 (2 F) participants. Mean peak BAL was 101 ± 15 mg/dL. Alcohol significantly increased brain [11C]PBR28 VT (n = 8; F(1,49) = 34.72, p > 0.0001; Cohen's d'=0.8-1.7) throughout brain by 9-16%. Alcohol significantly altered plasma cytokines TNF-α (F(2,22) = 17.49, p < 0.0001), IL-6 (F(2,22) = 18.00, p > 0.0001), and MCP-1 (F(2,22) = 7.02, p = 0.004). Exploratory analyses identified a negative association between the subjective degree of alcohol intoxication and changes in [11C]PBR28 VT. These findings provide, to our knowledge, the first in vivo human evidence for an acute brain immune response to alcohol.


Subject(s)
Brain , Positron-Emission Tomography , Male , Female , Humans , Positron-Emission Tomography/methods , Brain/metabolism , Radionuclide Imaging , Blood Alcohol Content , Receptors, GABA/metabolism , Immunity , Cytokines/metabolism
2.
Molecules ; 25(3)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32012954

ABSTRACT

Glioblastoma multiforme (GBM) is the deadliest type of brain tumor, affecting approximately three in 100,000 adults annually. Positron emission tomography (PET) imaging provides an important non-invasive method of measuring biochemically specific targets at GBM lesions. These powerful data can characterize tumors, predict treatment effectiveness, and monitor treatment. This review will discuss the PET imaging agents that have already been evaluated in GBM patients so far, and new imaging targets with promise for future use. Previously used PET imaging agents include the tracers for markers of proliferation ([11C]methionine; [18F]fluoro-ethyl-L-tyrosine, [18F]Fluorodopa,[18F]fluoro-thymidine, and [18F]clofarabine), hypoxia sensing ([18F]FMISO, [18F]FET-NIM, [18F]EF5, [18F]HX4, and [64Cu]ATSM), and ligands for inflammation. As cancer therapeutics evolve toward personalized medicine and therapies centered on tumor biomarkers, the development of complimentary selective PET agents can dramatically enhance these efforts. Newer biomarkers for GBM PET imaging are discussed, with some already in use for PET imaging other cancers and neurological disorders. These targets include Sigma 1, Sigma 2, programmed death ligand 1, poly-ADP-ribose polymerase, and isocitrate dehydrogenase. For GBM, these imaging agents come with additional considerations such as blood-brain barrier penetration, quantitative modeling approaches, and nonspecific binding.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , B7-H1 Antigen/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Positron-Emission Tomography , Precision Medicine , Receptors, sigma/metabolism , Sigma-1 Receptor
3.
J Nucl Med ; 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360052

ABSTRACT

PET imaging of synaptic vesicle glycoprotein 2A allows for noninvasive quantification of synapses. This first-in-human study aimed to evaluate the kinetics, test-retest reproducibility, and extent of specific binding of a recently developed synaptic vesicle glycoprotein 2A PET ligand, (R)-4-(3-(18F-fluoro)phenyl)-1-((3-methylpyridin-4-yl)methyl)pyrrolidine-2-one (18F-SynVesT-2), with fast brain kinetics. Methods: Nine healthy volunteers participated in this study and were scanned on a High Resolution Research Tomograph scanner with 18F-SynVesT-2. Five volunteers were scanned twice on 2 different days. Five volunteers were rescanned with preinjected levetiracetam (20 mg/kg, intravenously). Arterial blood was collected to calculate the plasma free fraction and generate the arterial input function. Individual MR images were coregistered to a brain atlas to define regions of interest for generating time-activity curves, which were fitted with 1- and 2-tissue-compartment (1TC and 2TC) models to derive the regional distribution volume (V T). The regional nondisplaceable binding potential (BP ND) was calculated from 1TC V T, using the centrum semiovale (CS) as the reference region. Results: 18F-SynVesT-2 was synthesized with high molar activity (187 ± 69 MBq/nmol, n = 19). The parent fraction of 18F-SynVesT-2 in plasma was 28% ± 8% at 30 min after injection, and the plasma free fraction was high (0.29 ± 0.04). 18F-SynVesT-2 entered the brain quickly, with an SUVpeak of 8 within 10 min after injection. Regional time-activity curves fitted well with both the 1TC and the 2TC models; however, V T was estimated more reliably using the 1TC model. The 1TC V T ranged from 1.9 ± 0.2 mL/cm3 in CS to 7.6 ± 0.8 mL/cm3 in the putamen, with low absolute test-retest variability (6.0% ± 3.6%). Regional BP ND ranged from 1.76 ± 0.21 in the hippocampus to 3.06 ± 0.29 in the putamen. A 20-min scan was sufficient to provide reliable V T and BP ND Conclusion: 18F-SynVesT-2 has fast kinetics, high specific uptake, and low nonspecific uptake in the brain. Consistent with the nonhuman primate results, the kinetics of 18F-SynVesT-2 is faster than the kinetics of 11C-UCB-J and 18F-SynVesT-1 in the human brain and enables a shorter dynamic scan to derive physiologic information on cerebral blood flow and synapse density.

4.
J Pharm Anal ; 10(5): 452-465, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33133729

ABSTRACT

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

5.
ACS Chem Neurosci ; 10(8): 3839-3846, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31339297

ABSTRACT

[18F]AV-1451 is one of the most widely used radiotracers for positron emission tomography (PET) imaging of tau protein aggregates in neurodegenerative disorders. While the radiotracer binds with high affinity to tau neurofibrillary tangles, extensive clinical studies have simultaneously revealed off-target tracer accumulation in areas of low tau burden such as the basal ganglia and choroid plexus. Though there are a number of possible reasons for this accumulation, it is often attributed to off-target binding to monoamine oxidase (MAO). In this paper, we investigate the association between [18F]AV-1451 and MAO through (i) enzyme inhibition assays, (ii) autoradiography with postmortem tissue samples, and (iii) nonhuman primate PET imaging. We confirm that [18F]AV-1451 is a weak inhibitor of MAO-A and -B and that MAO inhibitors can alter binding of [18F]AV-1451 in autoradiography and in vivo PET imaging.


Subject(s)
Basal Ganglia/drug effects , Carbolines/pharmacology , Choroid Plexus/drug effects , Monoamine Oxidase Inhibitors/pharmacology , Substantia Nigra/drug effects , Aged , Animals , Autoradiography , Basal Ganglia/diagnostic imaging , Basal Ganglia/metabolism , Choroid Plexus/diagnostic imaging , Choroid Plexus/metabolism , Humans , Monoamine Oxidase/metabolism , Positron-Emission Tomography , Primates , Radiopharmaceuticals , Substantia Nigra/diagnostic imaging , Substantia Nigra/metabolism
6.
ACS Chem Neurosci ; 10(4): 1867-1871, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30789704

ABSTRACT

In this Viewpoint, we highlight the history of positron emission tomography (PET) radiotracer development to quantify changes in monoamine oxidase (MAO)-A and -B enzyme expression or activity. MAO-A and MAO-B are critical for understanding monoaminergic pathways in psychiatric addiction disorders, and more recently in neurodegenerative disorders with MAO-B expression in astrogliosis. Unique radiochemical innovations have been shown for neuroimaging of MAOs including the clinical translation of irreversible propargylamine-based suicide inhibitors, application of deuterium-substitution to slow down metabolism, development of trapped metabolite imaging agents, and unique 11C-carbonylation chemistry toward novel high-affinity reversibly binding inhibitors.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Drug Development/trends , Monoamine Oxidase/metabolism , Positron-Emission Tomography/trends , Radiopharmaceuticals/metabolism , Drug Development/methods , Humans , Monoamine Oxidase/analysis , Neuroimaging/methods , Neuroimaging/trends , Positron-Emission Tomography/methods , Radiopharmaceuticals/analysis
7.
Int J Nanomedicine ; 14: 3069-3086, 2019.
Article in English | MEDLINE | ID: mdl-31118623

ABSTRACT

Background: Synthetic HDLs (sHDLs), small nanodiscs of apolipoprotein mimetic peptides surrounding lipid bilayers, were developed clinically for atheroma regression in cardiovascular patients. Formation of HDL involves interaction of apolipoprotein A-I (ApoA-I) with phospholipid bilayers and assembly into lipid-protein nanodiscs. Purpose: The objective of this study is to improve understanding of physico-chemical aspects of HDL biogenesis such as the thermodynamics of ApoA-I-peptide membrane insertion, lipid binding, and HDL self-assembly to improve our ability to form homogeneous sHDL nanodiscs that are suitable for clinical administration. Methods: The ApoA-I-mimetic peptide, 22A, was combined with either egg sphingomyelin (eSM) or 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) phospholipid vesicles to form sHDL. The sHDL assembly process was investigated through lipid vehicle solubilization assays and characterization of purity, size, and morphology of resulting nanoparticles via gel permeation chromatography (GPC), dynamic light scattering (DLS), and transmission electron microscopy (TEM). Peptide-lipid interactions involved were further probed by sum frequency generation (SFG) vibrational spectroscopy and attenuated total reflection-Fourier transform infrared spectroscopy (ATR-FTIR). The pharmacokinetics of eSM-sHDL and POPC-sHDL nanodiscs were investigated in Sprague Dawley rats. Results: sHDL formation was temperature-dependent, with spontaneous formation of sHDL nanoparticles occurring only at temperatures exceeding lipid transition temperatures as evidenced by DLS, GPC, and TEM characterization. SFG and ATR-FTIR spectroscopy findings support a change in peptide-lipid bilayer interactions at temperatures above the lipid transition temperature. Lipid-22A interactions were stronger with eSM than with POPC, which resulted in the formation of more homogeneous sHDL nanoparticles with longer in vivo circulation time as evidenced the PK study. Conclusion: Physico-chemical characteristics of sHDL are in part determined by phospholipid composition. Optimization of phospholipid composition may be utilized to improve the stability and homogeneity of sHDL.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Nanoparticles/chemistry , Peptides/metabolism , Phospholipids/metabolism , Amino Acid Sequence , Animals , Apolipoprotein A-I/chemistry , Dynamic Light Scattering , Kinetics , Lipid Bilayers/chemistry , Lipoproteins, HDL/chemistry , Male , Nanoparticles/ultrastructure , Peptides/chemistry , Peptides/pharmacokinetics , Phosphatidylcholines/administration & dosage , Rats, Sprague-Dawley , Solubility , Spectroscopy, Fourier Transform Infrared , Sphingomyelins/administration & dosage , Thermodynamics , Vibration
8.
Theranostics ; 8(22): 6352-6354, 2018.
Article in English | MEDLINE | ID: mdl-30613302

ABSTRACT

The receptor for advanced glycation end-products (RAGE) is implicated in multiple disease states such as cancer, diabetes and neurodegenerative disorders, and RAGE inhibitors are being explored as potential new therapies in such cases. Despite the known role RAGE plays in these conditions, there remains an urgent need for a molecular imaging agent that can accurately quantify RAGE levels in vivo, aid in validation of RAGE as a biomarker and/or therapeutic target, and support development of new RAGE inhibitors. This editorial highlights a multimodal nanoparticle-based imaging agent targeted at RAGE that was recently developed by Konopka and colleagues (Theranostics 2018; 8(18):5012-5024. doi:10.7150/thno.24791).


Subject(s)
Diabetes Mellitus , Nanoparticles , Glycation End Products, Advanced , Humans , Multimodal Imaging , Receptor for Advanced Glycation End Products/antagonists & inhibitors
9.
ACS Chem Neurosci ; 9(10): 2358-2372, 2018 10 17.
Article in English | MEDLINE | ID: mdl-29630337

ABSTRACT

In this Review, we consider the story of cocaine from its humble origins in South America to its status as one of the most abused substances in 21st century society. The synthesis and biosynthesis of cocaine are discussed, as well as its pharmacokinetics, metabolism, pharmacology, and importance in modern neuroscience and molecular imaging.


Subject(s)
Cocaine/chemistry , Cocaine/history , Cocaine/pharmacology , Dopamine Uptake Inhibitors/chemistry , Dopamine Uptake Inhibitors/history , Dopamine Uptake Inhibitors/pharmacology , Crack Cocaine/chemistry , Crack Cocaine/history , Crack Cocaine/pharmacology , History, 19th Century , History, 20th Century , History, 21st Century , Humans
10.
ACS Med Chem Lett ; 9(11): 1140-1143, 2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30429959

ABSTRACT

The development of a positron emission tomography (PET)/magnetic resonance spectroscopy (MRS) hybrid imaging agent allows for functional imaging by both methods with a single imaging agent. Enzyme substrates that are cleaved to form two metabolites present an interesting opportunity, as the unique metabolites generated might each be detected by a different modality. To be successful, such enzyme substrates would require administration of doses that (a) reach the in vivo target tissue at concentrations necessary for MRS imaging, (b) do not show substrate inhibition of tissue uptake or enzymatic activity, and (c) provide PET images that still reflect the action of the enzyme. We report in vitro and in vivo proof-of-concept studies of a carbon-11 small molecule substrate for brain monoamine oxidases that, upon enzyme-mediated cleavage, produces two metabolites, one detectable by PET and the other by MRS.

11.
ACS Chem Neurosci ; 9(12): 3024-3027, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30074755

ABSTRACT

Visualizing the in vivo activity of monoamine oxidase B (MAO-B) is a valuable tool in the ongoing investigation of astrogliosis in neurodegeneration. Existing strategies for imaging changes in MAO enzyme expression or activity have utilized the irreversible suicide inhibitors or high-affinity reversibly binding inhibitors as positron emission tomography (PET) ligands. As an alternative approach, we developed 4-methyl-7-[(1-[11C]methyl-1,2,3,6-tetrahydropyridin-4-yl)oxy]-2 H-chromen-2-one ([11C]Cou) as a metabolic trapping agent for MAO-B. Trapping of [11C]Cou in rhesus monkey brain demonstrated MAO-B selectivity. In this work, we have attempted to improve on the in vivo pharmacokinetics of [11C]Cou by using the deuterium kinetic isotope effect (KIE) to slow the MAO-B-mediated oxidation step and thus reduce the rate of trapping in brain tissues. However, in vitro assays of enzyme kinetics and in vivo PET imaging of pharmacokinetics in primate brain showed no effects of deuterium substitution on the tetrahydropyridine ring of [11C]Cou. The results are possibly due to masking of the KIE by a second step in the overall metabolism of the new imaging agent.


Subject(s)
Benzopyrans/pharmacokinetics , Brain/metabolism , Monoamine Oxidase/metabolism , Pyrrolidines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Animals , Brain/diagnostic imaging , Carbon Radioisotopes , Deuterium , Kinetics , Macaca mulatta , Positron-Emission Tomography
12.
ACS Chem Neurosci ; 7(3): 391-8, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26771209

ABSTRACT

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


Subject(s)
Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptor for Advanced Glycation End Products/analysis , Alzheimer Disease/metabolism , Animals , Autoradiography , Brain/metabolism , Chromatography, High Pressure Liquid , Fluorine Radioisotopes/pharmacokinetics , Humans , Immunohistochemistry , Macaca mulatta , Molecular Docking Simulation , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley
13.
Pharm Pat Anal ; 5(1): 17-47, 2016.
Article in English | MEDLINE | ID: mdl-26670619

ABSTRACT

The most commonly utilized PET radionuclide is fluorine-18 ((18)F) because of its convenient half-life and excellent imaging properties. In this review, we present the first analysis of patents issued for radiotracers labeled with fluorine-18 (between 2009 and 2015), and provide perspective on current trends and future directions in PET radiotracer development.


Subject(s)
Fluorine Radioisotopes , Radiopharmaceuticals , Animals , Heart Diseases/diagnostic imaging , Humans , Neoplasms/diagnostic imaging , Nervous System Diseases/diagnostic imaging , Neuroimaging , Patents as Topic , Radionuclide Imaging
SELECTION OF CITATIONS
SEARCH DETAIL