17 research outputs found

    Decreased defluorination using the novel beta-cell imaging agent [18F]FE-DTBZ-d4 in pigs examined by PET

    Get PDF
    The aim of the thesis was twofold. The first aim was to radiolabel small molecules by using carbon-11 and fluorine-18 for visualising beta cell mass (BCM) in the pancreas by PET. Diabetes Mellitus (DM) is a chronic metabolic disorder that results from an absolute or relative lack of BCM of endocrine pancreas. The lack of an adequate non-invasive imaging PET probe prevents detailed examination of beta cell loss during onset and progression of DM as well as development of novel treatments and islets transplantation progress. The second aim of the thesis was to radiolabel peptide molecules with fluorine-18 to visualise beta amyloid in Alzheimer’s disease (AD) brain. AD is a chronic, progressive neurodegenerative disorder. Brain penetration study of a labelled peptide, specific for beta amyloid that can cross blood-brain-barrier (BBB), is important to gain knowledge about the fate of the molecule as a diagnostic probe. A series of three novel radioligands for BCM imaging has been developed in this thesis. In paper I, a vesicular monoamine transporter type 2 (VMAT2) specific radioligand [18F]FE-DTBZ-d4 was synthesised in two steps. First step is the nucleophilic [18F]fluorination to produce deuterated-[18F]fluoroethylbromide followed by the O- alkylation of desmethyl-DTBZ precursor to produce [18F]FE-DTBZ-d4. The in vivo pharmacokinetics (PK) studies in pigs by PET/CT demonstrated reduced in vivo defluorination; therefore, it may be an improved potential candidate for imaging VMAT2 dense tissue i.e. islets transplantation in proximity to cortical bone structure. In Paper II, a glucokinase (GK) specific radioligand, [11C]AZ12504948, was synthesised in one step via alkylation of O-desmethyl precursor using [11C]methyl iodide. Both in vitro and in vivo (pig and monkey) studies with [11C]AZ12504948 for imaging GK in the pancreas and liver indicated low specificity. Increased target specificity is required for further progress in GK imaging using PET radioligands. In Paper III, a radioligand for G-protein coupled receptor 44 (GPR44), [11C/3H]AZ Compound X, was synthesised via S-methylation of sodium sulfinate salt in one step using [11C/3H]methyl iodide. In vitro binding of the radioligand, evaluated by autoradiography (ARG) on human and rat pancreatic tissues, confirmed higher specific binding in islets of human pancreatic tissue and no measurable binding in rat pancreas, which is devoid of GPR44. These studies indicate that the radioligand has suitable properties for beta cell imaging with high potential for further preclinical and clinical evaluation. Three novel D-peptides were radiolabelled with fluorine-18 ([18F]ACI-87, [18F]ACI- 88, [18F]ACI-89) by using prosthetic group N-succinimidyl-4-[18F]fluorobenzoate, [18F]SFB, with epsilon (Δ)-amino groups of lysine residues of peptide precursors in two steps. First step is the synthesis of [18F]SFB followed by the addition of [18F]SFB via acylation to the peptide molecule. Trimethylammonium salt [N(CH3)3+] precursor for synthesising [18F]SFB as well as the reference standard SFB were synthesised with good yields. Three 19F-peptide reference standards were also synthesised by using SFB. Preliminary ARG measurements were performed in AD and control human brains. ARG demonstrated higher radioligand uptake in the AD brain compared to age-matched control brain, which makes them potential for further use in in vivo testing by PET. However, preliminary PET (in vivo) studies in cynomolgus monkey brain, using these 18F-D-peptides, confirmed too low BBB penetration, making them unsuitable for further use as in vivo PET probes

    Glia Imaging Differentiates Multiple System Atrophy from Parkinson's Disease: A Positron Emission Tomography Study with [C-11]PBR28 and Machine Learning Analysis

    Get PDF
    Background The clinical diagnosis of multiple system atrophy (MSA) is challenged by overlapping features with Parkinson's disease (PD) and late-onset ataxias. Additional biomarkers are needed to confirm MSA and to advance the understanding of pathophysiology. Positron emission tomography (PET) imaging of the translocator protein (TSPO), expressed by glia cells, has shown elevations in MSA. Objective In this multicenter PET study, we assess the performance of TSPO imaging as a diagnostic marker for MSA.Methods We analyzed [C-11]PBR28 binding to TSPO using imaging data of 66 patients with MSA and 24 patients with PD. Group comparisons were based on regional analysis of parametric images. The diagnostic readout included visual reading of PET images against clinical diagnosis and machine learning analyses. Sensitivity, specificity, and receiver operating curves were used to discriminate MSA from PD and cerebellar from parkinsonian variant MSA. Results We observed a conspicuous pattern of elevated regional [C-11]PBR28 binding to TSPO in MSA as compared with PD, with "hotspots" in the lentiform nucleus and cerebellar white matter. Visual reading discriminated MSA from PD with 100% specificity and 83% sensitivity. The machine learning approach improved sensitivity to 96%. We identified MSA subtype-specific TSPO binding patterns. Conclusions We found a pattern of significantly increased regional glial TSPO binding in patients with MSA. Intriguingly, our data are in line with severe neuroinflammation in MSA. Glia imaging may have potential to support clinical MSA diagnosis and patient stratification in clinical trials on novel drug therapies for an alpha-synucleinopathy that remains strikingly incurable. </p

    Toward molecular imaging of the free fatty acid receptor 1

    No full text
    Molecular imaging of the free fatty acid receptor 1 (FFAR1) would be a valuable tool for drug development by enabling in vivo target engagement studies in human. It has also been suggested as a putative target for beta cell imaging, but the inherent lipophilicity of most FFAR1 binders produces high off-target binding, which has hampered progress in this area. The aim of this study was to generate a suitable lead compound for further PET labeling. In order to identify a lead compound for future PET labeling for quantitative imaging of FFAR1 in human, we evaluated tritiated small molecule FFAR1 binding probes ([H-3]AZ1, [H-3]AZ2 and [H-3]TAK-875) for their off-target binding, receptor density and affinity in human pancreatic tissue (islets and exocrine) and rodent insulinoma. [H-3]AZ1 showed improved specificity to FFAR1, with decreased off-target binding compared to [H-3]AZ2 and [H-3]TAK-875, while retaining high affinity in the nanomolar range. FFAR1 density in human islets was approximately 50% higher than in exocrine tissue. AZ1 is a suitable lead compound for PET labeling for molecular imaging of FFAR1 in humans, due to high affinity and reduced off-target binding

    Positron emission tomography using (18)F-labelled endothelin-1 reveals prevention of binding to cardiac receptors owing to tissue-specific clearance by ET(B) receptors in vivo

    No full text
    1. Our aim was to synthesise an (18)F analogue of endothelin-1 (ET-1), to dynamically image ET receptors in vivo by positron emission tomography (PET) and to elucidate the function of the ET(B) subtype as a clearing receptor in organs expressing high densities including kidney and lung. 2. [(18)F]-ET-1 was characterised in vitro and bound with a single subnanomolar affinity (K(D)=0.43±0.05 nM, B(max)=27.8±2.1 fmol mg(−1) protein) to human left ventricle (n=4). 3. The in vivo distribution of [(18)F]-ET-1 in anaesthetised rats was measured using a dedicated small animal PET scanner (microPET) and ex vivo analysis. 4. Dynamic PET data demonstrated that high levels of radioligand accumulated rapidly in the lung, kidney and liver, consistent with receptor binding. The in vivo distribution correlated with the anatomical localisation of receptors detected in vitro using [(125)I]-ET-1. However, the receptor density visualised in the heart was unexpectedly low compared with that predicted from the in vitro measurements. 5. [(18)F]-ET-1 binding in lungs could not be displaced by the ET(B) selective antagonist BQ788, in agreement with the proposed internalisation of ET-1 by ET(B) receptors. In contrast, infusion of BQ788 prior to injecting [(18)F]-ET-1 significantly reduce the amount of radioligand visualised in the ET(B) rich lung and kidney by 85% (P<0.05, n=3) and 55% (P<0.05, n=3), respectively. 6. Under conditions of ET(B) receptor blockade, the heart could be visualised by microPET imaging. 7. These results suggest that clearance by ET(B) receptors in the lung and kidney prevents binding of ET-1 to receptors in the heart

    In vivo visualization of ÎČ-cells by targeting of GPR44

    No full text
    GPR44 expression has recently been described as highly ÎČ-cell selective in the human pancreas and constitutes a tentative surrogate imaging biomarker in diabetes. A radiolabeled small-molecule GPR44 antagonist, [11C]AZ12204657, was evaluated for visualization of ÎČ-cells in pigs and nonhuman primates by positron emission tomography as well as in immunodeficient mice transplanted with human islets under the kidney capsule. In vitro autoradiography of human and animal pancreatic sections from subjects without and with diabetes, in combination with insulin staining, was performed to assess ÎČ-cell selectivity of the radiotracer. Proof of principle of in vivo targeting of human islets by [11C]AZ12204657 was shown in the immunodeficient mouse transplantation model. Furthermore, [11C]AZ12204657 bound by a GPR44-mediated mechanism in pancreatic sections from humans and pigs without diabetes, but not those with diabetes. In vivo [11C]AZ12204657 bound specifically to GPR44 in pancreas and spleen and could be competed away dose-dependently in nondiabetic pigs and nonhuman primates. [11C]AZ12204657 is a first-in-class surrogate imaging biomarker for pancreatic ÎČ-cells by targeting the protein GPR44

    Imaging atherosclerotic plaque inflammation with [18F]-fluorodeoxyglucose positron emission tomography

    No full text
    This article is not available through ChesterRep. It can be accessed at http://circ.ahajournals.org/cgi/content/full/105/23/2708This study demonstrates that atherosclerotic plaque inflammation can be imaged with 18FDG-PET, and that symptomatic, unstable plaques accumulate more 18FDG than asymptomatic lesions.This article was submitted to the RAE2008 for the University of Chester - Allied Health Professions and Studies

    The development of a GPR44 targeting radioligand [11C]AZ12204657 for in vivo assessment of beta cell mass.

    No full text
    BACKGROUND: The G-protein-coupled receptor 44 (GPR44) is a beta cell-restricted target that may serve as a marker for beta cell mass (BCM) given the development of a suitable PET ligand. METHODS: The binding characteristics of the selected candidate, AZ12204657, at human GPR44 were determined using in vitro ligand binding assays. AZ12204657 was radiolabeled using 11C- or 3H-labeled methyl iodide ([11C/3H]CH3I) in one step, and the conversion of [11C/3H]CH3I to the radiolabeled product [11C/3H]AZ12204657 was quantitative. The specificity of radioligand binding to GPR44 and the selectivity for beta cells were evaluated by in vitro binding studies on pancreatic sections from human and non-human primates as well as on homogenates from endocrine and exocrine pancreatic compartments. RESULTS: The radiochemical purity of the resulting radioligand [11C]AZ12204657 was &gt; 98%, with high molar activity (MA), 1351 ± 575 GBq/ÎŒmol (n = 18). The radiochemical purity of [3H]AZ12204657 was &gt; 99% with MA of 2 GBq/ÎŒmol. Pancreatic binding of [11C/3H]AZ12204657 was co-localized with insulin-positive islets of Langerhans in non-diabetic individuals and individuals with type 2 diabetes (T2D). The binding of [11C]AZ12204657 to GPR44 was &gt; 10 times higher in islet homogenates compared to exocrine homogenates. In human islets of Langerhans GPR44 was co-expressed with insulin, but not glucagon as assessed by co-staining and confocal microscopy. CONCLUSION: We radiolabeled [11C]AZ12204657, a potential PET radioligand for the beta cell-restricted protein GPR44. In vitro evaluation demonstrated that [3H]AZ12204657 and [11C]AZ12204657 selectively target pancreatic beta cells. [11C]AZ12204657 has promising properties as a marker for human BCM

    Low background and high contrast PET imaging of amyloid-ÎČ with [11C]AZD2995 and [11C]AZD2184 in Alzheimer’s disease patients

    Get PDF
    PURPOSE: The aim of this study was to evaluate AZD2995 side by side with AZD2184 as novel PET radioligands for imaging of amyloid-ÎČ in Alzheimer’s disease (AD). METHODS: In vitro binding of tritium-labelled AZD2995 and AZD2184 was studied and compared with that of the established amyloid-ÎČ PET radioligand PIB. Subsequently, a first-in-human in vivo PET study was performed using [(11)C]AZD2995 and [(11)C]AZD2184 in three healthy control subjects and seven AD patients. RESULTS: AZD2995, AZD2184 and PIB were found to share the same binding site to amyloid-ÎČ. [(3)H]AZD2995 had the highest signal-to-background ratio in brain tissue from patients with AD as well as in transgenic mice. However, [(11)C]AZD2184 had superior imaging properties in PET, as shown by larger effect sizes comparing binding potential values in cortical regions of AD patients and healthy controls. Nevertheless, probably due to a lower amount of nonspecific binding, the group separation of the distribution volume ratio values of [(11)C]AZD2995 was greater in areas with lower amyloid-ÎČ load, e.g. the hippocampus. CONCLUSION: Both AZD2995 and AZD2184 detect amyloid-ÎČ with high affinity and specificity and also display a lower degree of nonspecific binding than that reported for PIB. Overall [(11)C]AZD2184 seems to be an amyloid-ÎČ radioligand with higher uptake and better group separation when compared to [(11)C]AZD2995. However, the very low nonspecific binding of [(11)C]AZD2995 makes this radioligand potentially interesting as a tool to study minute levels of amyloid-ÎČ. This sensitivity may be important in investigating, for example, early prodromal stages of AD or in the longitudinal study of a disease modifying therapy. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s00259-012-2322-6) contains supplementary material, which is available to authorized users
    corecore