Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Z Med Phys ; 33(2): 135-145, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35688672

RESUMO

Monte Carlo (MC) simulations of X-ray image devices require splitting the simulation into two parts (i.e. the generation of x-rays and the actual imaging). The X-ray production remains unchanged for repeated imaging and can thus be stored in phase space (PhS) files and used for subsequent MC simulations. Especially for medical images these dedicated PhS files require a large amount of data storage, which is partly why Generative Adversarial Networks (GANs) were recently introduced. We enhanced the approach by a conditional GAN to model multiple energies using one network. This study compares the use of PhSs, GANs, and conditional GANs as photon source with measurements. An X-ray -based imaging system (i.e. ImagingRing) was modelled in this study. half-value layers (HVLs), focal spot, and Heel effect were measured for subsequent comparison. MC simulations were performed with GATE-RTion v1.0 considering the geometry and materials of the imaging system with vendor specific schematics. A traditional GAN model as well as the favourable conditional GAN was implemented for PhS generation. Results of the MC simulation were in agreement with the measurements regarding HVL, focal spot, and Heel effect. The conditional GAN performed best with a non-saturated loss function with R1 regularisation and gave similarly results as the traditional GAN approach. GANs proved to be superior to the PhS approach in terms of data storage and calculation overhead. Moreover, a conditional GAN enabled an energy interpolation to separate the network training process from the final required X-ray energies.


Assuntos
Fótons , Raios X , Radiografia , Simulação por Computador , Método de Monte Carlo
2.
Ann N Y Acad Sci ; 1494(1): 70-86, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33502798

RESUMO

Although extensive research on brown adipose tissue (BAT) has stimulated optimism in the battle against obesity and diabetes, BAT physiology and organ crosstalk are not fully understood. Besides BAT, melanin-concentrating hormone (MCH) and its receptor (MCHR1) play an important role in energy homeostasis. Because of the link between hypothalamic MCH neurons and sympathetic BAT activation via ß-adrenoceptors, we investigated the expression and physiological role of the MCHR1 in BAT. MCHR1 was detected in rodent and human BAT with RT-qPCR and western blot analyses. In vivo imaging in rats used the glucose analog [18 F]FDG and the MCHR1-tracer [11 C]SNAP-7941. We found that the ß3-adrenoceptor (ADRB3) agonist CL316,243 increased [11 C]SNAP-7941 uptake in BAT. Additionally, a pharmacological concentration of SNAP-7941-a low-affinity ADRB3 ligand-stimulated [18 F]FDG uptake, reflecting BAT activation. In cultured human adipocytes, CL316,243 induced MCHR1 expression, further supporting a direct interaction between MCHR1 and ADRB3. These findings characterized MCHR1 expression in rodent and human BAT for the first time, including in vitro and in vivo data demonstrating a link between MCHR1 and the ß3-adrenergic system. The presence of MCHR1 in BAT emphasizes the role of BAT in energy homeostasis and may help uncover treatment approaches for obesity.


Assuntos
Tecido Adiposo Marrom/metabolismo , Receptores do Hormônio Hipofisário/metabolismo , Animais , Fluordesoxiglucose F18/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley
3.
J Bone Miner Res ; 34(9): 1632-1645, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31063606

RESUMO

In rheumatoid arthritis (RA), chronic joint inflammation leading to bone and cartilage damage is the major cause of functional impairment. Whereas reduction of synovitis and blockade of joint damage can be successfully achieved by disease modifying antirheumatic therapies, bone repair upon therapeutic interventions has only been rarely reported. The aim of this study was to use fluorodeoxyglucose ([18 F]FDG) and [18 F]fluoride µPET/CT imaging to monitor systemic inflammatory and destructive bone remodeling processes as well as potential bone repair in an established mouse model of chronic inflammatory, erosive polyarthritis. Therefore, human tumor necrosis factor transgenic (hTNFtg) mice were treated with infliximab, an anti-TNF antibody, for 4 weeks. Before and after treatment period, mice received either [18 F]FDG, for detecting inflammatory processes, or [18 F]fluoride, for monitoring bone remodeling processes, for PET scans followed by CT scans. Standardized uptake values (SUVmean ) were analyzed in various joints and histopathological signs of arthritis, joint damage, and repair were assessed. Longitudinal PET/CT scans revealed a significant decrease in [18 F]FDG SUVs in affected joints demonstrating complete remission of inflammatory processes due to TNF blockade. In contrast, [18 F]fluoride SUVs could not discriminate between different severities of bone damage in hTNFtg mice. Repeated in vivo CT images proved a structural reversal of preexisting bone erosions after anti-TNF therapy. Accordingly, histological analysis showed complete resolution of synovial inflammation and healing of bone at sites of former bone erosion. We conclude that in vivo multimodal [18 F]FDG µPET/CT imaging allows to quantify and monitor inflammation-mediated bone damage and reveals not only reversal of synovitis but also bone repair upon TNF blockade in experimental arthritis. © 2019 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.


Assuntos
Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Fluordesoxiglucose F18/química , Inflamação/patologia , Imagem Multimodal , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Fator de Necrose Tumoral alfa/metabolismo , Animais , Regeneração Óssea , Remodelação Óssea , Cartilagem/diagnóstico por imagem , Cartilagem/patologia , Humanos , Articulações/diagnóstico por imagem , Articulações/patologia , Estudos Longitudinais , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoclastos/patologia , Membrana Sinovial/diagnóstico por imagem , Membrana Sinovial/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
4.
Mol Imaging Biol ; 21(2): 257-268, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29948643

RESUMO

PURPOSE: The melanin-concentrating hormone receptor 1 (MCHR1) has become an important pharmacological target, since it may be involved in various diseases, such as diabetes, insulin resistance, and obesity. Hence, a suitable positron emission tomography radiotracer for the in vivo assessment of the MCHR1 pharmacology is imperative. The current paper contrasts the extensive in vitro, in vivo, and ex vivo assessments of the radiotracers [18F]FE@SNAP and [11C]SNAP-7941 and provides comprehensive information about their biological and physicochemical properties. Furthermore, it examines their suitability for first-in-man imaging studies. PROCEDURES: Kinetic real-time cell-binding studies with [18F]FE@SNAP and [11C]SNAP-7941 were conducted on adherent Chines hamster ovary (CHO-K1) cells stably expressing the human MCHR1 and MCHR2. Small animal imaging studies on mice and rats were performed under displacement and baseline conditions, as well as after pretreatment with the P-glycoprotein/breast cancer resistant protein inhibitor tariquidar. After the imaging studies, detailed analyses of the ex vivo biodistribution were performed. Ex vivo metabolism was determined in rat blood and brain and analyzed at various time points using a quantitative radio-HPLC assay. RESULTS: [11C]SNAP-7941 demonstrates high uptake on CHO-K1-hMCHR1 cells, whereas no uptake was detected for the CHO-K1-hMCHR2 cells. In contrast, [18F]FE@SNAP evinced binding to CHO-K1-hMCHR1 and CHO-K1-hMCHR2 cells. Imaging studies with [18F]FE@SNAP and [11C]SNAP-7941 showed an increased brain uptake after tariquidar pretreatment in mice, as well as in rats, and exhibited a significant difference between the time-activity curves of the baseline and blocking groups. Biodistribution of both tracers demonstrated a decreased uptake after displacement. [11C]SNAP-7941 revealed a high metabolic stability in rats, whereas [18F]FE@SNAP was rapidly metabolized. CONCLUSIONS: Both radiotracers demonstrate appropriate imaging properties for the MCHR1. However, the pronounced metabolic stability as well as superior selectivity and affinity of [11C]SNAP-7941 underlines the decisive superiority over [18F]FE@SNAP.


Assuntos
Radioisótopos de Carbono/química , Radioisótopos de Flúor/química , Piperidinas/química , Tomografia por Emissão de Pósitrons , Pirimidinas/química , Receptores de Somatostatina/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Células CHO , Cromatografia de Afinidade , Cricetinae , Cricetulus , Humanos , Cinética , Metaboloma , Camundongos , Ligação Proteica , Ratos , Distribuição Tecidual
5.
Nucl Med Biol ; 58: 20-32, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29309919

RESUMO

INTRODUCTION: Due to the high candidate exclusion rate during a drug development process, an early prediction of the pharmacokinetic behavior would be needed. Accordingly, high performance bioaffinity chromatography (HPBAC) approaches are growing in popularity, however, there is a lack of knowledge and no consensus about the relation between HPBAC measurements, in vivo distribution and blood brain barrier (BBB) penetration behavior. With respect to radiotracers, there is almost no reference data available for plasma protein binding (PPB), permeability (Pm) and the membrane coefficient (KIAM). Thus, this study was aimed at exploring the relevance of measuring PPB, Pm and KIAM for the prediction of BBB penetration. METHODS: Measurements of %PPB, Pm and KIAM were performed using HPBAC. In total, 113 compounds were tested, 43 with brain uptake, 30 not showing brain uptake and 40 with known interactions with efflux transporters. Additionally, ClogP and HPLC logPowpH7.4 data were collected. RESULTS: %PPB, KIAM, Pm and ClogP values were in the same range for each of the three groups. A significant difference was observed for the HPLC logPowpH7.4 between CNS penetrating drug group (CNSpos) and the non-penetrating drug group (CNSneg), as well as for the CNSneg towards the drug group interacting with efflux transporters (DRUGefflux). However, as the other experimental data, also the HPLC logPowpH7.4 showed a broad overlapping of the single values between the groupings. CONCLUSION: Experimental reference values (logP, Pm, KIAM & PPB) of commonly used PET tracers and drugs showing different BBB penetration behavior are provided. The influence of the logP on brain uptake depends strongly on the selected method. However, using a single parameter (experimental or calculated) to predict BBB penetration or for the classification of drug groups is inexpedient.


Assuntos
Distribuição Tecidual , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Cromatografia Líquida de Alta Pressão , Permeabilidade
6.
Sci Rep ; 7(1): 8054, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808288

RESUMO

The MCHR1 is involved in the regulation of energy homeostasis and changes of the expression are linked to a variety of associated diseases, such as diabetes and adiposity. The study aimed at the in vitro and in vivo evaluation of [11C]SNAP-7941 and [18F]FE@SNAP as potential PET-tracers for the MCHR1. Competitive binding studies with non-radioactive derivatives and small-animal PET/CT and MRI brain studies were performed under baseline conditions and tracer displacement with the unlabelled MCHR1 antagonist (±)-SNAP-7941. Binding studies evinced high binding affinity of the non-radioactive derivatives. Small-animal imaging of [11C]SNAP-7941 and [18F]FE@SNAP evinced high tracer uptake in MCHR1-rich regions of the ventricular system. Quantitative analysis depicted a significant tracer reduction after displacement with (±)-SNAP-7941. Due to the high binding affinity of the non-labelled derivatives and the high specific tracer uptake of [11C]SNAP-7941 and [18F]FE@SNAP, there is strong evidence that both radiotracers may serve as highly suitable agents for specific MCHR1 imaging.


Assuntos
Radioisótopos de Carbono/metabolismo , Ventrículos Cerebrais/metabolismo , Radioisótopos de Flúor/metabolismo , Piperidinas/metabolismo , Pirimidinas/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Estudos de Avaliação como Assunto , Imageamento por Ressonância Magnética/métodos , Masculino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Ratos , Ratos Sprague-Dawley
7.
EJNMMI Res ; 7(1): 22, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28271461

RESUMO

BACKGROUND: Resolving the kinetic mechanisms of biomolecular interactions have become increasingly important in early-phase drug development. Since traditional in vitro methods belong to dose-dependent assessments, binding kinetics is usually overlooked. The present study aimed at the establishment of two novel experimental approaches for the assessment of binding affinity of both, radiolabelled and non-labelled compounds targeting the A3R, based on high-resolution real-time data acquisition of radioligand-receptor binding kinetics. A novel time-resolved competition assay was developed and applied to determine the Ki of eight different A3R antagonists, using CHO-K1 cells stably expressing the hA3R. In addition, a new kinetic real-time cell-binding approach was established to quantify the rate constants k on and k off, as well as the dedicated K d of the A3R agonist [125I]-AB-MECA. Furthermore, lipophilicity measurements were conducted to control influences due to physicochemical properties of the used compounds. RESULTS: Two novel real-time cell-binding approaches were successfully developed and established. Both experimental procedures were found to visualize the kinetic binding characteristics with high spatial and temporal resolution, resulting in reliable affinity values, which are in good agreement with values previously reported with traditional methods. Taking into account the lipophilicity of the A3R antagonists, no influences on the experimental performance and the resulting affinity were investigated. CONCLUSIONS: Both kinetic binding approaches comprise tracer administration and subsequent binding to living cells, expressing the dedicated target protein. Therefore, the experiments resemble better the true in vivo physiological conditions and provide important markers of cellular feedback and biological response.

8.
EJNMMI Res ; 6(1): 31, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27033361

RESUMO

BACKGROUND: The melanin-concentrating hormone receptor 1 (MCHR1), which is highly expressed in the lateral hypothalamus, plays a key role in energy homeostasis, obesity and other endocrine diseases. Hence, there is a major interest in in vivo imaging of this receptor. A PET tracer would allow non-invasive in vivo visualization and quantification of the MCHR1. The aim of the study was the ex vivo evaluation of the MCHR1 ligand [(18)F]FE@SNAP as a potential PET tracer for the MCHR1. METHODS: [(18)F]FE@SNAP was injected directly into the jugular vein of awake naïve rats for ex vivo brain autoradiography, biodistribution and additional blood metabolite analysis. Blocking experiments were conducted using the unlabeled MCHR1 ligand SNAP-7941. RESULTS: A high uptake of [(18)F]FE@SNAP was observed in the lateral hypothalamus and the ventricular system. Both regions were significantly blocked by SNAP-7941. Biodistribution evinced the highest uptake in the kidneys, adrenals, lung and duodenum. Specific blocking with SNAP-7941 led to a significant tracer reduction in the heart and adrenals. In plasma samples, 47.73 ± 6.1 % of a hydrophilic radioactive metabolite was found 45 min after tracer injection. CONCLUSIONS: Since [(18)F]FE@SNAP uptake was significantly blocked in the lateral hypothalamus, there is strong evidence that [(18)F]FE@SNAP is a highly suitable agent for specific MCHR1 imaging in the central nervous system. Additionally, this finding is supported by the specific blocking in the ventricular system, where the MCHR1 is expressed in the ependymal cells. These findings suggest that [(18)F]FE@SNAP could serve as a useful imaging and therapy monitoring tool for MCHR1-related pathologies.

9.
Eur J Nucl Med Mol Imaging ; 42(5): 741-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25601336

RESUMO

PURPOSE: The adenosine A3 receptor (A3R) is involved in cardiovascular, neurological and tumour-related pathologies and serves as an exceptional pharmaceutical target in the clinical setting. A3R antagonists are considered antiinflammatory, antiallergic and anticancer agents, and to have potential for the treatment of asthma, COPD, glaucoma and stroke. Hence, an appropriate A3R PET tracer would be highly beneficial for the diagnosis and therapy monitoring of these diseases. Therefore, in this preclinical in vivo study we evaluated the potential as a PET tracer of the A3R antagonist [(18)F]FE@SUPPY. METHODS: Rats were injected with [(18)F]FE@SUPPY for baseline scans and blocking scans (A3R with MRS1523 or FE@SUPPY, P-gp with tariquidar; three animals each). Additionally, metabolism was studied in plasma and brain. In a preliminary experiment in a mouse xenograft model (mice injected with cells expressing the human A3R; three animals), the animals received [(18)F]FE@SUPPY and [(18)F]FDG. Dynamic PET imaging was performed (60 min in rats, 90 min in xenografted mice). In vitro stability of [(18)F]FE@SUPPY in human and rat plasma was also evaluated. RESULTS: [(18)F]FE@SUPPY showed high uptake in fat-rich regions and low uptake in the brain. Pretreatment with MRS1523 led to a decrease in [(18)F]FE@SUPPY uptake (p = 0.03), and pretreatment with the P-gp inhibitor tariquidar led to a 1.24-fold increase in [(18)F]FE@SUPPY uptake (p = 0.09) in rat brain. There was no significant difference in metabolites in plasma and brain in the treatment groups. However, plasma concentrations of [(18)F]FE@SUPPY were reduced to levels similar to those in rat brain after blocking. In contrast to [(18)F]FDG uptake (p = 0.12), the xenograft model showed significantly increased uptake of [(18)F]FE@SUPPY in the tissue masses from CHO cells expressing the human A3R (p = 0.03). [(18)F]FE@SUPPY was stable in human plasma. CONCLUSION: Selective and significant tracer uptake of [(18)F]FE@SUPPY was found in xenografted mice injected with cells expressing human A3R. This finding supports the strategy of evaluating [(18)F]FE@SUPPY in "humanized animal models". In conclusion, preclinical evaluation points to the suitability of [(18)F]FE@SUPPY as an A3R PET tracer in humans.


Assuntos
Ácidos Nicotínicos/farmacocinética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacocinética , Receptor A3 de Adenosina/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Masculino , Camundongos , Neoplasias Experimentais/diagnóstico por imagem , Ligação Proteica , Compostos Radiofarmacêuticos/síntese química , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
10.
Sci Pharm ; 81(3): 625-39, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24106662

RESUMO

The melanin-concentrating hormone (MCH) system is a new target for the treatment of human disorders. Since the knowledge of the MCH system's involvement in a variety of pathologies (obesity, diabetes, and deregulation of metabolic feedback mechanism) is based on in vitro or preclinical studies, a suitable positron emission tomography (PET) tracer needs to be developed. We herein present the preparation and first preclinical evaluation of [(18)F]FE@SNAP - a new PET tracer for MCH receptor-1 (MCHR1). The synthesis was performed using a microfluidic device. Preclinical evaluation included binding affinity, plasma stability, plasma free fraction, stability against the cytochrome P-450 (CYP450) system using liver microsomes, stability against carboxyl-esterase, and methods to assess the penetration of the blood-brain barrier (BBB) such as logD analysis and immobilized artificial membrane (IAM) chromatography. Levels at 374 ± 202 MBq [(18)F]FE@SNAP were obtained after purification. The obtained K d value of [(18)F]FE@SNAP was 2.9 nM. [(18)F]FE@SNAP evinced high stability against carboxylesterase, CYP450 enzymes, and in human plasma. LogD (3.83) and IAM chromatography results (Pm=0.51) were in the same range as for known BBB-penetrating compounds. The synthesis of [(18)F]FE@SNAP was reliable and successful. Due to high binding affinity and stability, [(18)F]FE@SNAP is a promising tracer for MCHR1.

11.
Nucl Med Biol ; 40(7): 919-25, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23829932

RESUMO

INTRODUCTION: Due to its involvement in a variety of pathologies (obesity, diabetes, gut inflammation and depression), the melanin concentrating hormone receptor 1 (MCHR1) is a new target for the treatment of these lifestyle diseases. We previously presented the radiosynthesis of [(11)C]SNAP-7941, the first potential PET tracer for the MCHR1. METHODS: We herein present its in vitro and in vivo evaluation, including binding affinity, plasma stability, stability against liver mircrosomes and carboxylesterase, lipohilicity, biodistribution, in vivo metabolism and small-animal PET. RESULTS: [(11)C]SNAP-7941 evinced high stability against liver microsomes, carboxylesterase and in human plasma. The first small-animal PET experiments revealed a 5 fold increased brain uptake after Pgp/BCRP inhibition. Therefore, it can be assumed that [(11)C]SNAP-7941 is a Pgp/BCRP substrate. No metabolites were found in brain. CONCLUSION: On the basis of these experiments with healthy rats, the suitability of [(11)C]SNAP-7941 for the visualisation of central and peripheral MCHR1 remains speculative.


Assuntos
Piperidinas , Tomografia por Emissão de Pósitrons/métodos , Pirimidinas , Receptores de Somatostatina/metabolismo , Animais , Radioisótopos de Carbono , Hidrolases de Éster Carboxílico/metabolismo , Estabilidade de Medicamentos , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Microssomos Hepáticos/metabolismo , Piperidinas/química , Piperidinas/metabolismo , Piperidinas/farmacocinética , Pirimidinas/química , Pirimidinas/metabolismo , Pirimidinas/farmacocinética , Ratos , Ratos Sprague-Dawley
12.
Bioorg Med Chem ; 20(19): 5936-40, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22921745

RESUMO

Changes in the expression of the melanin concentrating hormone receptor 1 (MCHR1) are involved in a variety of pathologies, especially obesity and anxiety disorders. To monitor these pathologies in-vivo positron emission tomography (PET) is a suitable method. After the successful radiosynthesis of [(11)C]SNAP-7941-the first PET-Tracer for the MCHR1, we aimed to synthesize its [(18)F]fluoroethylated analogue: [(18)F]FE@SNAP. Therefore, microfluidic and vessel-based approaches were tested. [(18)F]fluoroethylation was conducted via various [(18)F]fluoroalkylated synthons and direct [(18)F]fluorination. Only the direct [(18)F]fluorination of a tosylated precursor using a flow-through microreactor was successful, affording [(18)F]FE@SNAP in 44.3 ± 2.6%.


Assuntos
Radioisótopos de Flúor/química , Microfluídica , Piperidinas/química , Tomografia por Emissão de Pósitrons , Pirimidinas/química , Receptores de Somatostatina/análise , Humanos , Microfluídica/métodos , Tomografia por Emissão de Pósitrons/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...