Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 27(3)2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35163938

RESUMO

A large number of applications for fibroblast activation protein inhibitors (FAPI)-based PET agents have been evaluated in conditions ranging from cancer to non-malignant diseases such as myocardial infarction. In particular, 68Ga-FAPI-46 was reported to have a high specificity and affinity for FAP-expressing cells, a fast and high accumulation in tumor lesions/injuries together with a fast body clearance when investigated in vivo. Due to the increasing interest in the use of the agent both preclinically and clinically, we developed an automated synthesis for the production of 68Ga-FAPI-46 on a Trasis AiO platform. The new synthetic procedure, which included the processing of the generator eluate using a strong cation exchange resin and a final purification step through an HLB followed by a QMA cartridge, yielded 68Ga-FAPI-46 with high radiochemical purity (>98%) and apparent molar activity (271.1 ± 105.6 MBq/nmol). Additionally, the in vitro and in vivo properties of the product were assessed on glioblastoma cells and mouse model. Although developed for the preparation of 68Ga-FAPI-46 for preclinical use, our method can be adapted for clinical production as a reliable alternative to the manual (i.e., cold kit) or modular systems preparations already described in the literature.


Assuntos
Glioblastoma/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Quinolinas/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Animais , Apoptose , Proliferação de Células , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Nus , Radioquímica , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Synapse ; 64(4): 301-12, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19957364

RESUMO

[(11)C]-(+)-PHNO is a new dopamine D(2/3) receptor agonist radiotracer which has been successfully used to measure D(2/3) receptor availability in experimental animals and man. Here we report in vivo evaluation in the rat of the biodistribution, metabolism, specificity, selectivity, and dopamine sensitivity of carbon11-labeled PHNO ([(11)C]-3-PHNO) produced by an alternative radiochemical synthesis method. [(11)C]-3-PHNO showed rapid metabolism and clearance from most peripheral organs and tissues. [(11)C]-3-PHNO, but not its polar metabolite, readily crossed the blood-brain barrier and showed high levels of uptake in the D(2/3)-rich striatum. Pretreatment with unlabeled PHNO and the D(2/3) receptor antagonist raclopride indicated that binding in the striatum was specific and selective to D(2/3) receptors. PET studies in anesthetized rats revealed significant reductions in [(11)C]-3-PHNO binding in the striatum following amphetamine administration, indicating sensitivity to increases in endogenous dopamine concentrations. D(2/3) antagonist pretreatment additionally indicated moderate levels of [(11)C]-3-PHNO specific binding in several extrastriatal brain areas-most notably the olfactory bulbs and tubercles, thalamus, and hypothalamus. Of particular interest, approximately 30% of [(11)C]-3-PHNO signal in the cerebellum-a region often used as a "low-binding" reference region for PET quantification-was attributable to specific signal. These data demonstrate that [(11)C]-3-PHNO shows similar tracer characteristics to [(11)C]-(+)-PHNO, but additionally indicate that radiolabeled PHNO may be used to estimate D(2/3) receptor availability in select extrastriatal brain regions with PET.


Assuntos
Radioisótopos de Carbono/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/diagnóstico por imagem , Agonistas de Dopamina/metabolismo , Oxazinas/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Receptores de Dopamina D2/agonistas , Animais , Ligação Competitiva , Cromatografia Líquida de Alta Pressão/métodos , Masculino , Oxazinas/metabolismo , Ligação Proteica , Racloprida/metabolismo , Racloprida/farmacocinética , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Distribuição Tecidual
3.
Sci Rep ; 10(1): 20918, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262374

RESUMO

Targeted radiotherapy with 131I-mIBG, a substrate of the human norepinephrine transporter (NET-1), shows promising responses in heavily pre-treated neuroblastoma (NB) patients. Combinatorial approaches that enhance 131I-mIBG tumour uptake are of substantial clinical interest but biomarkers of response are needed. Here, we investigate the potential of 18F-mFBG, a positron emission tomography (PET) analogue of the 123I-mIBG radiotracer, to quantify NET-1 expression levels in mouse models of NB following treatment with AZD2014, a dual mTOR inhibitor. The response to AZD2014 treatment was evaluated in MYCN amplified NB cell lines (Kelly and SK-N-BE(2)C) by Western blot (WB) and immunohistochemistry. PET quantification of 18F-mFBG uptake post-treatment in vivo was performed, and data correlated with NET-1 protein levels measured ex vivo. Following 72 h AZD2014 treatment, in vitro WB analysis indicated decreased mTOR signalling and enhanced NET-1 expression in both cell lines, and 18F-mFBG revealed a concentration-dependent increase in NET-1 function. AZD2014 treatment failed however to inhibit mTOR signalling in vivo and did not significantly modulate intratumoural NET-1 activity. Image analysis of 18F-mFBG PET data showed correlation to tumour NET-1 protein expression, while further studies are needed to elucidate whether NET-1 upregulation induced by blocking mTOR might be a useful adjunct to 131I-mIBG therapy.


Assuntos
Fluorbenzenos/química , Guanidinas/química , Neuroblastoma/tratamento farmacológico , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , 3-Iodobenzilguanidina/química , Animais , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Linhagem Celular Tumoral , Endocitose/efeitos dos fármacos , Feminino , Humanos , Camundongos Nus , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Neuroblastoma/patologia , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Compostos Radiofarmacêuticos/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Distribuição Tecidual/efeitos dos fármacos
4.
Amino Acids ; 37(4): 717-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19011732

RESUMO

Three strategies for chemoselective labeling of RGD peptides with (18)F have been compared. Aminooxy [(18)F]fluorobenzaldehyde conjugation provided 40 +/- 12% decay-corrected radiochemical yield using a fully automated method. An one-pot protocol for 'click labeling' of the RGD scaffold with 2-[(18)F]fluoroethylazide afforded 47 +/- 8% decay-corrected radiochemical yield. Attempted conjugation with 3-[(18)F]fluoropropanethiol led to extensive decomposition and was therefore found unsuitable for labeling of the RGD peptide investigated. The results suggest that 'click labeling' of RGD peptides provides an attractive alternative to aminooxy aldehyde condensation, however, 2-[(18)F]-fluoroethylazide may be too small to allow separation of large (18)F-labeled RGD peptides from their precursors.


Assuntos
Azidas/química , Radioisótopos de Flúor/química , Marcação por Isótopo/métodos , Oligopeptídeos/química , Oligopeptídeos/síntese química
5.
Nucl Med Biol ; 42(5): 494-498, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25725983

RESUMO

INTRODUCTION: [(18)F]HX4 is a 2-nitroimidazole based investigational radiotracer for imaging hypoxia. METHODS: A two-step, one-pot synthetic procedure was developed on a GE Tracerlab MX-FDG with a disposable cassette. Nucleophilic substitution of a nosylate group with [(18)F]fluoride was followed by solvent evaporation and acidic removal of the acetyl protecting group. HPLC purification in a bio-compatible solvent mixture was developed. RESULTS AND CONCLUSIONS: Using starting activities of 80-110 GBq [(18)F]fluoride, GMP compliant [(18)F]HX4 was produced in non-decay corrected radiochemical yields of 12 ± 3% (n = 9) in 55 min including HPLC purification. No reformulation steps were required. The mean specific activity of the final product was 2450 GBq/µmol. Modifications to the process and final formulation were included to prevent decomposition of the product, and these changes resulted in an improved stability of the formulated [(18)F]HX4, with a shelf-life of at least 8h post-synthesis. The product consistently passed all required quality control tests to determine that the [(18)F]HX4 was suitable for clinical use. Using a 90 minute target bombardment, and 80-110 GBq starting [(18)F]fluoride, the method produced multiple patient doses.


Assuntos
Hipóxia/diagnóstico por imagem , Nitroimidazóis/síntese química , Tomografia por Emissão de Pósitrons , Radioquímica/métodos , Triazóis/síntese química , Automação , Nitroimidazóis/química , Controle de Qualidade , Triazóis/química
6.
Appl Radiat Isot ; 73: 79-83, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23291563

RESUMO

We report here a radiosynthesis for the D(2/3) agonist (+)-4-([3-(11)C]propyl)-3,4,4a,5,6,10b-hexahydro-2H-naphtho[1,2-b][1,4]oxazin-9-ol (3-[(11)C]-(+)-PHNO) labelled at the terminal carbon of the N-propyl chain. The protocol is based on (11)C-methylation of an N-acetyl precursor. This initial step is followed by a reduction with LiAlH(4) to give ([3-(11)C]-(+)-PHNO). We first applied the method for the synthesis of a model compound, N-3-([(11)C]propyl)-1,2,3,4-tetrahydroisoquinoline, which we obtained in 77-97% analytical radiochemical yield (n=6) in 20 min. Similarly, we prepared ([3-(11)C]-(+)-PHNO) in 55-60% analytical radiochemical yield (n=5) using a one-pot procedure. We have also been able to implement the complete process on a semi-automated module. This platform delivered purified and formulated [3-(11)C]PHNO with an average radiochemical yield of 9% (n=13, range 2-30%, non-decay corrected), a radiochemical purity >95%, and a specific radioactivity of 26.8-81.1 GBq/µmol in a total time of 63-65 min.


Assuntos
Radioisótopos de Carbono/química , Hidrocarbonetos Iodados/química , Marcação por Isótopo/métodos , Oxazinas/síntese química , Compostos Radiofarmacêuticos/síntese química , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D3/agonistas , Cromatografia Líquida de Alta Pressão , Oxazinas/química , Compostos Radiofarmacêuticos/química
7.
Clin Cancer Res ; 18(4): 1063-72, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22235095

RESUMO

PURPOSE: (11)C-Choline-positron emission tomography (PET) has been exploited to detect the aberrant choline metabolism in tumors. Radiolabeled choline uptake within the imaging time is primarily a function of transport, phosphorylation, and oxidation. Rapid choline oxidation, however, complicates interpretation of PET data. In this study, we investigated the biologic basis of the oxidation of deuterated choline analogs and assessed their specificity in human tumor xenografts. EXPERIMENTAL DESIGN: (11)C-Choline, (11)C-methyl-[1,2-(2)H(4)]-choline ((11)C-D4-choline), and (18)F-D4-choline were synthesized to permit comparison. Biodistribution, metabolism, small-animal PET studies, and kinetic analysis of tracer uptake were carried out in human colon HCT116 xenograft-bearing mice. RESULTS: Oxidation of choline analogs to betaine was highest with (11)C-choline, with reduced oxidation observed with (11)C-D4-choline and substantially reduced with (18)F-D4-choline, suggesting that both fluorination and deuteration were important for tracer metabolism. Although all tracers were converted intracellularly to labeled phosphocholine (specific signal), the higher rate constants for intracellular retention (K(i) and k(3)) of (11)C-choline and (11)C-D4-choline, compared with (18)F-D4-choline, were explained by the rapid conversion of the nonfluorinated tracers to betaine within HCT116 tumors. Imaging studies showed that the uptake of (18)F-D4-choline in three tumors with similar radiotracer delivery (K(1)) and choline kinase α expression-HCT116, A375, and PC3-M-were the same, suggesting that (18)F-D4-choline has utility for cancer detection irrespective of histologic type. CONCLUSION: We have shown here that both deuteration and fluorination combine to provide protection against choline oxidation in vivo. (18)F-D4-choline showed the highest selectivity for phosphorylation and warrants clinical evaluation.


Assuntos
Radioisótopos de Carbono , Colina , Deutério , Fluordesoxiglucose F18 , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Animais , Linhagem Celular Tumoral , Colina/análogos & derivados , Colina/metabolismo , Modelos Animais de Doenças , Humanos , Rim/metabolismo , Cinética , Masculino , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Oxirredução , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo , Traçadores Radioativos
8.
Eur J Nucl Med Mol Imaging ; 35(9): 1624-35, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18481065

RESUMO

INTRODUCTION: An increase in human cardiac alpha(1)-adrenoceptor (alpha(1)-AR) density is associated with various diseases such as myocardial ischemia, congestive heart failure, hypertrophic cardiomyopathy and hypertension. Positron emission tomography (PET) with an appropriate radioligand offers the possibility of imaging receptor function in the normal and diseased heart. [(11)C]GB67, an analogue of prazosin, has been shown in rats to have potential as a PET ligand with high selectivity to alpha(1)-AR. However, alpha(1)-AR density is up to ten times higher in rat heart compared to that in man. The aim of the present preclinical study was to extend the previous evaluation to a large mammal heart, where the alpha(1)-AR density is comparable to man, and to validate a method for quantification before PET studies in man. METHODS: Seven [(11)C]GB67 PET studies, with weight-adjusted target dose of either 5.29 MBq kg(-1) (pilot, test-retest and baseline-predose studies) or 8.22 MBq kg(-1) (baseline-displacement studies), were performed in four anaesthetised pigs (39.5 +/- 3.9 kg). Total myocardial volume of distribution (V (T)) was estimated under different pharmacological conditions using compartmental analysis with a radiolabelled metabolite-corrected arterial plasma input function. A maximum possible blocking dose of 0.12 mumol kg(-1) of unlabeled GB67 was given 20 min before [(11)C]GB67 administration in the predose study and 45 min after administration of [(11)C]GB67 in the displacement study. In addition, [(15)O]CO (3,000 MBq) and [(15)O]H(2)O, with weight adjusted target dose of 10.57 MBq kg(-1), were also administered for estimation of blood volume recovery (RC) of the left ventricular cavity and myocardial perfusion (MBF), respectively. RESULTS: [(11)C]GB67 V (T) values (in ml cm(-3)) were estimated to be 24.2 +/- 5.5 (range, 17.3-31.3), 10.1 (predose) and 11.6 (displacement). MBF did not differ within each pig, including between baseline and predose conditions. Predose and displacement studies showed that specific binding of [(11)C]GB67 to myocardial alpha(1)-ARs accounts for approximately 50% of V (T). CONCLUSION: The present study offers a methodology for using [(11)C]GB67 as a radioligand to quantify human myocardial alpha(1)-ARs in clinical PET studies.


Assuntos
Miocárdio/metabolismo , Tomografia por Emissão de Pósitrons , Prazosina/análogos & derivados , Prazosina/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Suínos/metabolismo , Animais , Artérias/metabolismo , Monóxido de Carbono/metabolismo , Radioisótopos de Carbono , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Cinética , Miocárdio/citologia , Prazosina/sangue , Ligação Proteica , Água/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa