Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-28197095

RESUMEN

Heterozygous missense mutations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been reported to significantly increase the risk of developing Alzheimer's disease (AD). Since TREM2 is specifically expressed by microglia in the brain, we hypothesized that soluble TREM2 (sTREM2) levels may increase together with in vivo biomarkers of microglial activity and amyloidosis in an AD mouse model as assessed by small animal positron-emission-tomography (µPET). In this cross-sectional study, we examined a strong amyloid mouse model (PS2APP) of four age groups by µPET with [18F]-GE180 (glial activation) and [18F]-florbetaben (amyloidosis), followed by measurement of sTREM2 levels and amyloid levels in the brain. Pathology affected brain regions were compared between tracers (dice similarity coefficients) and pseudo-longitudinally. µPET results of both tracers were correlated with terminal TREM2 levels. The brain sTREM2 levels strongly increased with age of PS2APP mice (5 vs. 16 months: +211%, p < 0.001), and correlated highly with µPET signals of microglial activity (R = 0.89, p < 0.001) and amyloidosis (R = 0.92, p < 0.001). Dual µPET enabled regional mapping of glial activation and amyloidosis in the mouse brain, which progressed concertedly leading to a high overlap in aged PS2APP mice (dice similarity 67%). Together, these results substantiate the use of in vivo µPET measurements in conjunction with post mortem sTREM2 in future anti-inflammatory treatment trials. Taking human data into account sTREM2 may increase during active amyloid deposition.

2.
Front Neurosci ; 10: 260, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27378835

RESUMEN

Noninvasive tumor growth monitoring is of particular interest for the evaluation of experimental glioma therapies. This study investigates the potential of positron emission tomography (PET) using O-(2-(18)F-fluoroethyl)-L-tyrosine ([(18)F]-FET) to determine tumor growth in a murine glioblastoma (GBM) model-including estimation of the biological tumor volume (BTV), which has hitherto not been investigated in the pre-clinical context. Fifteen GBM-bearing mice (GL261) and six control mice (shams) were investigated during 5 weeks by PET followed by autoradiographic and histological assessments. [(18)F]-FET PET was quantitated by calculation of maximum and mean standardized uptake values within a universal volume-of-interest (VOI) corrected for healthy background (SUVmax/BG, SUVmean/BG). A partial volume effect correction (PVEC) was applied in comparison to ex vivo autoradiography. BTVs obtained by predefined thresholds for VOI definition (SUV/BG: ≥1.4; ≥1.6; ≥1.8; ≥2.0) were compared to the histologically assessed tumor volume (n = 8). Finally, individual "optimal" thresholds for BTV definition best reflecting the histology were determined. In GBM mice SUVmax/BG and SUVmean/BG clearly increased with time, however at high inter-animal variability. No relevant [(18)F]-FET uptake was observed in shams. PVEC recovered signal loss of SUVmean/BG assessment in relation to autoradiography. BTV as estimated by predefined thresholds strongly differed from the histology volume. Strikingly, the individual "optimal" thresholds for BTV assessment correlated highly with SUVmax/BG (ρ = 0.97, p < 0.001), allowing SUVmax/BG-based calculation of individual thresholds. The method was verified by a subsequent validation study (n = 15, ρ = 0.88, p < 0.01) leading to extensively higher agreement of BTV estimations when compared to histology in contrast to predefined thresholds. [(18)F]-FET PET with standard SUV measurements is feasible for glioma imaging in the GBM mouse model. PVEC is beneficial to improve accuracy of [(18)F]-FET PET SUV quantification. Although SUVmax/BG and SUVmean/BG increase during the disease course, these parameters do not correlate with the respective tumor size. For the first time, we propose a histology-verified method allowing appropriate individual BTV estimation for volumetric in vivo monitoring of tumor growth with [(18)F]-FET PET and show that standardized thresholds from routine clinical practice seem to be inappropriate for BTV estimation in the GBM mouse model.

3.
Oncotarget ; 7(34): 54795-54810, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27458162

RESUMEN

Adoptively transferred mesenchymal stem cells (MSCs) home to solid tumors. Biologic features within the tumor environment can be used to selectively activate transgenes in engineered MSCs after tumor invasion. One of the characteristic features of solid tumors is hypoxia. We evaluated a hypoxia-based imaging and therapy strategy to target expression of the sodium iodide symporter (NIS) gene to experimental hepatocellular carcinoma (HCC) delivered by MSCs.MSCs engineered to express transgenes driven by a hypoxia-responsive promoter showed robust transgene induction under hypoxia as demonstrated by mCherry expression in tumor cell spheroid models, or radioiodide uptake using NIS. Subcutaneous and orthotopic HCC xenograft mouse models revealed significant levels of perchlorate-sensitive NIS-mediated tumoral radioiodide accumulation by tumor-recruited MSCs using 123I-scintigraphy or 124I-positron emission tomography. Functional NIS expression was further confirmed by ex vivo 123I-biodistribution analysis. Administration of a therapeutic dose of 131I in mice treated with NIS-transfected MSCs resulted in delayed tumor growth and reduced tumor perfusion, as shown by contrast-enhanced sonography, and significantly prolonged survival of mice bearing orthotopic HCC tumors. Interestingly, radioiodide uptake into subcutaneous tumors was not sufficient to induce therapeutic effects. Our results demonstrate the potential of using tumor hypoxia-based approaches to drive radioiodide therapy in non-thyroidal tumors.


Asunto(s)
Radioisótopos de Yodo/uso terapéutico , Neoplasias Hepáticas Experimentales/radioterapia , Neoplasias Hepáticas/radioterapia , Células Madre Mesenquimatosas/metabolismo , Simportadores/metabolismo , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Hipoxia , Radioisótopos de Yodo/farmacocinética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones Desnudos , Simportadores/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Ther ; 24(8): 1395-404, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27157666

RESUMEN

The sodium iodide symporter (NIS) as well-characterized theranostic gene represents an outstanding tool to target different cancer types allowing noninvasive imaging of functional NIS expression and therapeutic radioiodide application. Based on its overexpression on the surface of most cancer types, the cMET/hepatocyte growth factor receptor serves as ideal target for tumor-selective gene delivery. Sequence-defined polymers as nonviral gene delivery vehicles comprising polyethylene glycol (PEG) and cationic (oligoethanoamino) amide cores coupled with a cMET-binding peptide (cMBP2) were complexed with NIS-DNA and tested for receptor-specificity, transduction efficiency, and therapeutic efficacy in hepatocellular cancer cells HuH7. In vitro iodide uptake studies demonstrated high transduction efficiency and cMET-specificity of NIS-encoding polyplexes (cMBP2-PEG-Stp/NIS) compared to polyplexes without targeting ligand (Ala-PEG-Stp/NIS) and without coding DNA (cMBP2-PEG-Stp/Antisense-NIS). Tumor recruitment and vector biodistribution were investigated in vivo in a subcutaneous xenograft mouse model showing high tumor-selective iodide accumulation in cMBP2-PEG-Stp/NIS-treated mice (6.6 ± 1.6% ID/g (123)I, biological half-life 3 hours) by (123)I-scintigraphy. Therapy studies with three cycles of polyplexes and (131)I application resulted in significant delay in tumor growth and prolonged survival. These data demonstrate the enormous potential of cMET-targeted sequence-defined polymers combined with the unique theranostic function of NIS allowing for optimized transfection efficiency while eliminating toxicity.


Asunto(s)
Técnicas de Transferencia de Gen , Polímeros , Proteínas Proto-Oncogénicas c-met/genética , Simportadores/genética , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Xenoinjertos , Humanos , Ratones , Polietilenglicoles/química , Polímeros/química , Proteínas Proto-Oncogénicas c-met/metabolismo , Nanomedicina Teranóstica , Transfección
5.
Front Neurosci ; 10: 45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26973442

RESUMEN

Preclinical PET studies of ß-amyloid (Aß) accumulation are of growing importance, but comparisons between research sites require standardized and optimized methods for quantitation. Therefore, we aimed to evaluate systematically the (1) impact of an automated algorithm for spatial brain normalization, and (2) intensity scaling methods of different reference regions for Aß-PET in a large dataset of transgenic mice. PS2APP mice in a 6 week longitudinal setting (N = 37) and another set of PS2APP mice at a histologically assessed narrow range of Aß burden (N = 40) were investigated by [(18)F]-florbetaben PET. Manual spatial normalization by three readers at different training levels was performed prior to application of an automated brain spatial normalization and inter-reader agreement was assessed by Fleiss Kappa (κ). For this method the impact of templates at different pathology stages was investigated. Four different reference regions on brain uptake normalization were used to calculate frontal cortical standardized uptake value ratios (SUVRCTX∕REF), relative to raw SUVCTX. Results were compared on the basis of longitudinal stability (Cohen's d), and in reference to gold standard histopathological quantitation (Pearson's R). Application of an automated brain spatial normalization resulted in nearly perfect agreement (all κ≥0.99) between different readers, with constant or improved correlation with histology. Templates based on inappropriate pathology stage resulted in up to 2.9% systematic bias for SUVRCTX∕REF. All SUVRCTX∕REF methods performed better than SUVCTX both with regard to longitudinal stability (d≥1.21 vs. d = 0.23) and histological gold standard agreement (R≥0.66 vs. R≥0.31). Voxel-wise analysis suggested a physiologically implausible longitudinal decrease by global mean scaling. The hindbrain white matter reference (R mean = 0.75) was slightly superior to the brainstem (R mean = 0.74) and the cerebellum (R mean = 0.73). Automated brain normalization with reference region templates presents an excellent method to avoid the inter-reader variability in preclinical Aß-PET scans. Intracerebral reference regions lacking Aß pathology serve for precise longitudinal in vivo quantification of [(18)F]-florbetaben PET. Hindbrain white matter reference performed best when considering the composite of quality criteria.

6.
J Nucl Med ; 57(5): 792-8, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26912432

RESUMEN

Abnormal accumulation of tau aggregates in the brain is one of the hallmarks of Alzheimer disease neuropathology. We visualized tau deposition in vivo with the previously developed 2-arylquinoline derivative (18)F-THK5117 using small-animal PET in conjunction with autoradiography and immunohistochemistry gold standard assessment in 2 transgenic mouse models expressing hyperphosphorylated tau. Small-animal PET recordings were obtained in groups of P301S (n = 11) and biGT mice (n = 16) of different ages, with age-matched wild-type (WT) serving as controls. After intravenous administration of 16 ± 2 MBq of (18)F-THK5117, a dynamic 90-min emission recording was initiated for P301S mice and during 20-50 min after injection for biGT mice, followed by a 15-min transmission scan. After coregistration to the MRI atlas and scaling to the cerebellum, we performed volume-of-interest-based analysis (SUV ratio [SUVR]) and statistical parametric mapping. Small-animal PET results were compared with autoradiography ex vivo and in vitro and further validated with AT8 staining for neurofibrillary tangles. SUVRs calculated from static recordings during the interval of 20-50 min after tracer injection correlated highly with estimates of binding potential based on the entire dynamic emission recordings (R = 0.85). SUVR increases were detected in the brain stem of aged P301S mice (+11%; P < 0.001) and in entorhinal/amygdaloidal areas (+15%; P < 0.001) of biGT mice when compared with WT, whereas aged WT mice did not show increased tracer uptake. Immunohistochemical tau loads correlated with small-animal PET SUVR for both P301S (R = 0.8; P < 0.001) and biGT (R = 0.7; P < 0.001) mice, and distribution patterns of AT8-positive neurons matched voxelwise statistical parametric mapping analysis. Saturable binding of the tracer was verified by autoradiographic blocking studies. In the first dedicated small-animal PET study in 2 different transgenic tauopathy mouse models using the tau tracer (18)F-THK5117, the temporal and spatial progression could be visualized in good correlation with gold standard assessments of tau accumulation. The serial small-animal PET method could afford the means for preclinical testing of novel therapeutic approaches by accommodating interanimal variability at baseline, while detection thresholds in young animals have to be considered.


Asunto(s)
Compuestos de Anilina , Tomografía de Emisión de Positrones/métodos , Quinolinas , Proteínas tau/metabolismo , Compuestos de Anilina/química , Animales , Autorradiografía , Inmunohistoquímica , Ratones , Ratones Transgénicos , Fosforilación , Quinolinas/química , Radioquímica , Proteínas tau/genética
7.
Endocr Relat Cancer ; 22(6): 941-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26307023

RESUMEN

To improve our understanding of non-genomic, integrin αvß3-mediated thyroid hormone action in tumour stroma formation, we examined the effects of triiodo-l-thyronine (T3), l-thyroxine (T4) and integrin-specific inhibitor tetrac on differentiation, migration and invasion of mesenchymal stem cells (MSCs) that are an integral part of the tumour's fibrovascular network. Primary human bone marrow-derived MSCs were treated with T3 or T4 in the presence of hepatocellular carcinoma (HCC) cell-conditioned medium (CM), which resulted in stimulation of the expression of genes associated with cancer-associated fibroblast-like differentiation as determined by qPCR and ELISA. In addition, T3 and T4 increased migration of MSCs towards HCC cell-CM and invasion into the centre of three-dimensional HCC cell spheroids. All these effects were tetrac-dependent and therefore integrin αvß3-mediated. In a subcutaneous HCC xenograft model, MSCs showed significantly increased recruitment and invasion into tumours of hyperthyroid mice compared to euthyroid and, in particular, hypothyroid mice, while treatment with tetrac almost completely eliminated MSC recruitment. These studies significantly improve our understanding of the anti-tumour activity of tetrac, as well as the mechanisms that regulate MSC differentiation and recruitment in the context of tumour stroma formation, as an important prerequisite for the utilisation of MSCs as gene delivery vehicles.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Integrina alfaVbeta3/fisiología , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas de Neoplasias/fisiología , Células del Estroma/patología , Tiroxina/análogos & derivados , Tiroxina/farmacología , Triyodotironina/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Linaje de la Célula , Movimiento Celular , Medios de Cultivo Condicionados , Xenoinjertos , Humanos , Hipertiroidismo/inducido químicamente , Hipertiroidismo/complicaciones , Hipotiroidismo/inducido químicamente , Hipotiroidismo/complicaciones , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/complicaciones , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Organismos Libres de Patógenos Específicos , Esferoides Celulares , Tiroxina/uso terapéutico , Tiroxina/toxicidad , Triyodotironina/uso terapéutico , Triyodotironina/toxicidad , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Nucl Med ; 56(4): 600-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25745085

RESUMEN

UNLABELLED: The tumor-homing property of mesenchymal stem cells (MSCs) allows targeted delivery of therapeutic genes into the tumor microenvironment. The application of sodium iodide symporter (NIS) as a theranostic gene allows noninvasive imaging of MSC biodistribution and transgene expression before therapeutic radioiodine application. We have previously shown that linking therapeutic transgene expression to induction of the chemokine CCL5/RANTES allows a more focused expression within primary tumors, as the adoptively transferred MSC develop carcinoma-associated fibroblast-like characteristics. Although RANTES/CCL5-NIS targeting has shown efficacy in the treatment of primary tumors, it was not clear if it would also be effective in controlling the growth of metastatic disease. METHODS: To expand the potential range of tumor targets, we investigated the biodistribution and tumor recruitment of MSCs transfected with NIS under control of the RANTES/CCL5 promoter (RANTES-NIS-MSC) in a colon cancer liver metastasis mouse model established by intrasplenic injection of the human colon cancer cell line LS174t. RANTES-NIS-MSCs were injected intravenously, followed by (123)I scintigraphy, (124)I PET imaging, and (131)I therapy. RESULTS: Results show robust MSC recruitment with RANTES/CCL5-promoter activation within the stroma of liver metastases as evidenced by tumor-selective iodide accumulation, immunohistochemistry, and real-time polymerase chain reaction. Therapeutic application of (131)I in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved overall survival. CONCLUSION: This novel gene therapy approach opens the prospect of NIS-mediated radionuclide therapy of metastatic cancer after MSC-mediated gene delivery.


Asunto(s)
Neoplasias del Colon/radioterapia , Radioisótopos de Yodo/uso terapéutico , Células Madre Mesenquimatosas/citología , Simportadores/química , Simportadores/genética , Animales , Línea Celular Tumoral , Quimiocina CCL5/metabolismo , Femenino , Fibroblastos/metabolismo , Terapia Genética/métodos , Humanos , Neoplasias Hepáticas/radioterapia , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Plásmidos/metabolismo , Tomografía de Emisión de Positrones , Cintigrafía , Transgenes , Microambiente Tumoral
9.
PLoS One ; 10(2): e0115543, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25668193

RESUMEN

OBJECTIVES: To investigate a multimodal, multiparametric perfusion MRI / 18F-fluoro-deoxyglucose-(18F-FDG)-PET imaging protocol for monitoring regorafenib therapy effects on experimental colorectal adenocarcinomas in rats with immunohistochemical validation. MATERIALS AND METHODS: Human colorectal adenocarcinoma xenografts (HT-29) were implanted subcutaneously in n = 17 (n = 10 therapy group; n = 7 control group) female athymic nude rats (Hsd:RH-Foxn1rnu). Animals were imaged at baseline and after a one-week daily treatment protocol with regorafenib (10 mg/kg bodyweight) using a multimodal, multiparametric perfusion MRI/18F-FDG-PET imaging protocol. In perfusion MRI, quantitative parameters of plasma flow (PF, mL/100 mL/min), plasma volume (PV, %) and endothelial permeability-surface area product (PS, mL/100 mL/min) were calculated. In 18F-FDG-PET, tumor-to-background-ratio (TTB) was calculated. Perfusion MRI parameters were correlated with TTB and immunohistochemical assessments of tumor microvascular density (CD-31) and cell proliferation (Ki-67). RESULTS: Regorafenib significantly (p<0.01) suppressed PF (81.1±7.5 to 50.6±16.0 mL/100mL/min), PV (12.1±3.6 to 7.5±1.6%) and PS (13.6±3.2 to 7.9±2.3 mL/100mL/min) as well as TTB (3.4±0.6 to 1.9±1.1) between baseline and day 7. Immunohistochemistry revealed significantly (p<0.03) lower tumor microvascular density (CD-31, 7.0±2.4 vs. 16.1±5.9) and tumor cell proliferation (Ki-67, 434.0 ± 62.9 vs. 663.0 ± 98.3) in the therapy group. Perfusion MRI parameters ΔPF, ΔPV and ΔPS showed strong and significant (r = 0.67-0.78; p<0.01) correlations to the PET parameter ΔTTB and significant correlations (r = 0.57-0.67; p<0.03) to immunohistochemical Ki-67 as well as to CD-31-stainings (r = 0.49-0.55; p<0.05). CONCLUSIONS: A multimodal, multiparametric perfusion MRI/PET imaging protocol allowed for non-invasive monitoring of regorafenib therapy effects on experimental colorectal adenocarcinomas in vivo with significant correlations between perfusion MRI parameters and 18F-FDG-PET validated by immunohistochemistry.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Monitoreo de Drogas/métodos , Angiografía por Resonancia Magnética/métodos , Compuestos de Fenilurea/uso terapéutico , Tomografía de Emisión de Positrones/métodos , Piridinas/uso terapéutico , Animales , Femenino , Fluorodesoxiglucosa F18 , Xenoinjertos/patología , Humanos , Inmunohistoquímica/métodos , Neoplasias Experimentales/tratamiento farmacológico , Ratas , Ratas Desnudas
10.
PLoS One ; 10(2): e0116678, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25706990

RESUMEN

We aimed to compare [18F]-florbetaben PET imaging in four transgenic mouse strains modelling Alzheimer's disease (AD), with the main focus on APPswe/PS2 mice and C57Bl/6 mice serving as controls (WT). A consistent PET protocol (N = 82 PET scans) was used, with cortical standardized uptake value ratio (SUVR) relative to cerebellum as the endpoint. We correlated methoxy-X04 staining of ß-amyloid with PET results, and undertook ex vivo autoradiography for further validation of a partial volume effect correction (PVEC) of PET data. The SUVR in APPswe/PS2 increased from 0.95±0.04 at five months (N = 5) and 1.04±0.03 (p<0.05) at eight months (N = 7) to 1.07±0.04 (p<0.005) at ten months (N = 6), 1.28±0.06 (p<0.001) at 16 months (N = 6) and 1.39±0.09 (p<0.001) at 19 months (N = 6). SUVR was 0.95±0.03 in WT mice of all ages (N = 22). In APPswe/PS1G384A mice, the SUVR was 0.93/0.98 at five months (N = 2) and 1.11 at 16 months (N = 1). In APPswe/PS1dE9 mice, the SUVR declined from 0.96/0.96 at 12 months (N = 2) to 0.91/0.92 at 24 months (N = 2), due to ß-amyloid plaques in cerebellum. PVEC reduced the discrepancy between SUVR-PET and autoradiography from -22% to +2% and increased the differences between young and aged transgenic animals. SUVR and plaque load correlated highly between strains for uncorrected (R = 0.94, p<0.001) and PVE-corrected (R = 0.95, p<0.001) data. We find that APPswe/PS2 mice may be optimal for longitudinal amyloid-PET monitoring in planned interventions studies.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina , Encéfalo/diagnóstico por imagen , Placa Amiloide/diagnóstico por imagen , Estilbenos , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Estudios Transversales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Tomografía de Emisión de Positrones
11.
J Nucl Med ; 55(8): 1342-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25024426

RESUMEN

UNLABELLED: Human mesenchymal stem cells (hMSCs) represent a promising treatment approach for tissue repair and regeneration. However, little is known about the underlying mechanisms and the fate of the transplanted cells. The objective of the presented work was to determine the feasibility of PET imaging and in vivo monitoring after transplantation of dopamine type 2 receptor-expressing cells. METHODS: An hMSC line constitutively expressing a mutant of the dopamine type 2 receptor (D2R80A) was generated by lentiviral gene transfer. D2R80A messenger RNA expression was confirmed by reverse transcriptase-polymerase chain reaction. Localization of the transmembrane protein was analyzed by confocal fluorescence microscopy. The stem cell character of transduced hMSCs was investigated by adipogenic and osteogenic differentiation. Migration capacity was assessed by scratch assays in time-lapse imaging. In vitro specific binding of ligands was tested by fluorescence-activated cell sorting analysis and by radioligand assay using (18)F-fallypride. Imaging of D2R80A overexpressing hMSC transplanted into athymic rats was performed by PET using (18)F-fallypride. RESULTS: hMSCs showed long-term overexpression of D2R80A. As expected, the fluorescence signal suggested the primary localization of the protein in the membrane of the transduced cells. hMSC and D2R80A retained their stem cell character demonstrated by their osteogenic and adipogenic differentiation capacity and their proliferation and migration behavior. For in vitro hMSCs, at least 90% expressed the D2R80A transgene and hMSC-D2R80A showed specific binding of (18)F-fallypride. In vivo, a specific signal was detected at the transplantation site up to 7 d by PET. CONCLUSION: The mutant of the dopamine type 2 receptor (D2R80A) is a potent reporter to detect hMSCs by PET in vivo.


Asunto(s)
Benzamidas , Rastreo Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/diagnóstico por imagen , Tomografía de Emisión de Positrones , Pirrolidinas , Receptores de Dopamina D2/genética , Animales , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Mutación , Ratas , Transgenes/genética
12.
Neuroimage ; 84: 843-53, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24055703

RESUMEN

We previously investigated the progression of ß-amyloid deposition in brain of mice over-expressing amyloid-precursor protein (APP-Swe), a model of Alzheimer's disease (AD), in a longitudinal PET study with the novel ß-amyloid tracer [(18)F]-florbetaben. There were certain discrepancies between PET and autoradiographic findings, which seemed to arise from partial volume effects (PVE). Since this phenomenon can lead to bias, most especially in the quantitation of brain microPET studies of mice, we aimed in the present study to investigate the magnitude of PVE on [(18)F]-florbetaben quantitation in murine brain, and to establish and validate a useful correction method (PVEC). Phantom studies with solutions of known radioactivity concentration were performed to measure the full-width-at-half-maximum (FWHM) resolution of the Siemens Inveon DPET and to validate a volume-of-interest (VOI)-based PVEC algorithm. Several VOI-brain-masks were applied to perform in vivo PVEC on [(18)F]-florbetaben data from C57BL/6(N=6) mice, while uncorrected and PVE-corrected data were cross-validated with gamma counting and autoradiography. Next, PVEC was performed on longitudinal PET data set consisting of 43 PET scans in APP-Swe (13-20months) and age-matched wild-type (WT) mice using the previously defined masks. VOI-based cortex-to-cerebellum ratios (SUVR) were compared for uncorrected and PVE-corrected results. Brains from a subset of transgenic mice were ultimately examined by autoradiography ex vivo and histochemistry in vitro as gold standard assessments, and compared to VOI-based PET results. The phantom study indicated a FWHM of 1.72mm. Applying a VOI-brain-mask including extracerebral regions gave robust PVEC, with increased precision of the SUVR results. Cortical SUVR increased with age in APP-Swe mice compared to baseline measurements (16months: +5.5%, p<0.005; 20months: +15.5%, p<0.05) with uncorrected data, and to a substantially greater extent with PVEC (16months: +12.2% p<0.005; 20months: +36.4% p<0.05). WT animals showed no binding changes, irrespective of PVEC. Relative to autoradiographic results, the error [%] for uncorrected cortical SUVR was 18.9% for native PET data, and declined to 4.8% upon PVEC, in high correlation with histochemistry results. We calculate that PVEC increases by 10% statistical power for detecting altered [(18)F]-florbetaben uptake in aging APP-Swe mice in planned studies of disease modifying treatments on amyloidogenesis.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Compuestos de Anilina , Encéfalo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Radiofármacos , Estilbenos , Algoritmos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Autorradiografía , Encéfalo/patología , Modelos Animales de Enfermedad , Radioisótopos de Flúor , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fantasmas de Imagen , Tomografía de Emisión de Positrones
13.
J Nucl Med ; 54(7): 1127-34, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23729696

RESUMEN

UNLABELLED: The progression of ß-amyloid deposition in the brains of mice overexpressing Swedish mutant ß-amyloid precursor protein (APP-Swe), a model of Alzheimer disease (AD), was investigated in a longitudinal PET study using the novel ß-amyloid tracer (18)F-florbetaben. METHODS: Groups of APP-Swe and age-matched wild-type (WT) mice (age range, 10-20 mo) were investigated. Dynamic emission recordings were acquired with a small-animal PET scanner during 90 min after the administration of (18)F-florbetaben (9 MBq, intravenously). After spatial normalization of individual PET recordings to common coordinates for mouse brain, binding potentials (BPND) and standardized uptake value ratios (SUVRs) were calculated relative to the cerebellum. Voxelwise analyses were performed using statistical parametric mapping (SPM). Histochemical analyses and ex vivo autoradiography were ultimately performed in a subset of animals as a gold standard assessment of ß-amyloid plaque load. RESULTS: SUVRs calculated from static recordings during the interval of 30-60 min after tracer injection correlated highly with estimates of BPND based on the entire dynamic emission recordings. (18)F-florbetaben binding did not significantly differ in APP-Swe mice and WT animals at 10 and 13 mo of age. At 16 mo of age, the APP-Swe mice had a significant 7.9% increase (P < 0.01) in cortical (18)F-florbetaben uptake above baseline and at 20 mo there was a 16.6% increase (P < 0.001), whereas WT mice did not show any temporal changes in tracer uptake during the interval of follow-up. Voxelwise SPM analyses revealed the first signs of increased cortical binding at 13 mo and confirmed progressive binding increases in both the frontal and the temporal cortices (P < 0.001 uncorrected) to 20 mo. The SUVR strongly correlated with percentage plaque load (R = 0.95, P < 0.001). CONCLUSION: In the first longitudinal PET study in an AD mouse model using the novel ß-amyloid tracer (18)F-florbetaben, the temporal and spatial progression of amyloidogenesis in the brain of APP-Swe mice were sensitively monitored. This method should afford the means for preclinical testing of novel therapeutic approaches to the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Compuestos de Anilina/farmacocinética , Encéfalo/metabolismo , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos , Estilbenos/farmacocinética , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/diagnóstico por imagen , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
14.
Neuro Oncol ; 11(6): 861-70, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19401596

RESUMEN

Inhibitors targeting the integrin alpha(v)beta(3) are promising new agents currently tested in clinical trials for supplemental therapy of glioblastoma multiforme (GBM). The aim of our study was to evaluate (18)F-labeled glycosylated Arg-Gly-Asp peptide ([(18)F]Galacto-RGD) PET for noninvasive imaging of alpha(v)beta(3) expression in patients with GBM, suggesting eligibility for this kind of additional treatment. Patients with suspected or recurrent GBM were examined with [(18)F]Galacto-RGD PET. Standardized uptake values (SUVs) of tumor hotspots, galea, and blood pool were derived by region-of-interest analysis. [(18)F]Galacto-RGD PET images were fused with cranial MR images for image-guided surgery. Tumor samples taken from areas with intense tracer accumulation in the [(18)F]Galacto-RGD PET images and were analyzed histologically and immunohistochemically for alpha(v)beta(3) integrin expression. While normal brain tissue did not show significant tracer accumulation (mean SUV, 0.09 +/- 0.04), GBMs demonstrated significant but heterogeneous tracer uptake, with a maximum in the highly proliferating and infiltrating areas of tumors (mean SUV, 1.6 +/- 0.5). Immunohistochemical staining was prominent in tumor microvessels as well as glial tumor cells. In areas of highly proliferating glial tumor cells, tracer uptake (SUVs) in the [(18)F]Galacto-RGD PET images correlated with immunohistochemical alpha(v)beta(3) integrin expression of corresponding tumor samples. These data suggest that [(18)F] Galacto-RGD PET successfully identifies alpha(v)beta(3) expression in patients with GBM and might be a promising tool for planning and monitoring individualized cancer therapies targeting this integrin.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Galactosa/análogos & derivados , Glioma/diagnóstico por imagen , Integrina alfaVbeta3/metabolismo , Péptidos Cíclicos , Tomografía de Emisión de Positrones , Adulto , Anciano , Neoplasias Encefálicas/metabolismo , Femenino , Radioisótopos de Flúor , Glioma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Pronóstico , Radiofármacos , Distribución Tisular
15.
Eur J Nucl Med Mol Imaging ; 35(8): 1507-15, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18369617

RESUMEN

PURPOSE: alphavbeta3 integrins are important cell adhesion receptors involved in angiogenic processes. Recently, we demonstrated using [(18)F]Galacto-RGD that monitoring of alphavbeta3 expression is feasible. Here, we introduce (68)Ga- and (111)In-labelled derivatives and compare them with [(18)F]Galacto-RGD. METHODS: For radiolabelling, cyclo(RGDfK(DOTA)) was synthesised using SPPS. For in vitro characterisation determination of partition coefficients, protein binding, metabolic stability, alphavbeta3 affinity and cell uptake and for in vivo characterization, biodistribution studies and micro positron emission tomography (PET) imaging were carried out. For in vivo and in vitro studies, human melanoma M21 (alphavbeta3 positive) and M21-L (alphavbeta3 negative) cells were used. RESULTS: Both tracers can be synthesised straightforward. The compounds showed hydrophilic properties and high metabolic stability. Up to 23% protein-bound activity for [(68)Ga]DOTA-RGD and only up to 1.4% for [(111)In]DOTA-RGD was found. Cell uptake studies indicate receptor-specific accumulation. This is confirmed by the biodistribution data. One hour p.i. accumulation in alphavbeta3-positive tumours was 2.9 +/- 0.3%ID/g and in alphavbeta3-negative tumours 0.8 +/- 0.1%ID/g for [(68)Ga]DOTA-RGD ([(111)In]DOTA-RGD: 1.9 +/- 0.3%ID/g and 0.5 +/- 0.2%ID/g; [(18)F]Galacto-RGD: 1.6 +/- 0.2%ID/g and 0.4 +/- 0.1%ID/g). Thus, tumour uptake ratios were comparable. Due to approx. 3-fold higher blood pool activities for [(68)Ga]DOTA-RGD, tumour/blood ratios were higher for [(111)In]DOTA-RGD and [(18)F]Galacto-RGD. However, microPET studies demonstrated that visualisation of alphavbeta3-positive tumours using [(68)Ga]DOTA-RGD is possible. CONCLUSIONS: Our data indicate that [(68)Ga]DOTA-RGD allows monitoring of alphavbeta3 expression. Especially, the much easier radiosynthesis compared to [(18)F]Galacto-RGD would make it an attractive alternative. However, due to higher blood pool activity, [(18)F]Galacto-RGD remains superior for imaging alphavbeta3 expression. Introduction of alternative chelator systems may overcome the disadvantages.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Integrina alfaVbeta3/metabolismo , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/metabolismo , Compuestos Organometálicos/farmacocinética , Péptidos Cíclicos/farmacocinética , Animales , Melanoma/irrigación sanguínea , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Técnicas de Sonda Molecular , Especificidad de Órganos , Cintigrafía , Distribución Tisular
16.
Acad Radiol ; 15(3): 342-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18280932

RESUMEN

RATIONALE AND OBJECTIVES: This study was performed to assess the imaging characteristics and pharmacokinetics of 1,3-Bis-[7-(3-amino-2,4,6-triiodophenyl)-heptanoyl]-2-oleoyl glycerol (DHOG, Fenestra LC), a hepatobiliary contrast agent for microCT. MATERIALS AND METHODS: We investigated the abdomen of 18 female C3H mice in a MicroCAT II microCT scanner before contrast agent injection and at multiple time points up to 48 hours after intravenous injection of DHOG (1 g I/kg body weight). The contrast agent effect was determined quantitatively and dynamically by measuring pre- and postcontrast Hounsfield units (HU) of the liver, aorta, spleen, and kidneys. Based on additional phantom measurements, the reproducibility of lesion detection was estimated for different lesion sizes. RESULTS: DHOG caused a marked early postcontrast enhancement of blood in the aorta and a very high enhancement of the spleen, both slowly declined after 90 minutes. The liver parenchyma showed a slow contrast agent accumulation and clearly increased HU data between 3 and 7 hours after injection. No significant renal parenchymal enhancement or excretion was noticed. At early time points after administration, DHOG exhibits characteristics of a macromolecular contrast agent by demonstrating a blood pool effect. At later time points, DHOG provides a prolonged, marked liver enhancement on microCT images due to its specific liver uptake. For a lesion size of 1 mm diameter, the variability in between two scans was 27.7 HU (P < .05) and the variability for different planes of one scan was 19.8 HU (P < .05). CONCLUSIONS: DHOG yields a very good visualization of the liver and delineation of the surrounding structures with a long plateau. It is a very suitable contrast agent for liver imaging in mice for microCT imaging. The presented protocol provides a high reproducibility for lesion detection with a relatively low radiation dose.


Asunto(s)
Colangiografía/métodos , Medios de Contraste , Isótopos de Yodo , Hígado/diagnóstico por imagen , Intensificación de Imagen Radiográfica/métodos , Tomografía Computarizada por Rayos X/métodos , Triglicéridos , Animales , Aortografía , Medios de Contraste/administración & dosificación , Femenino , Imagenología Tridimensional/métodos , Inyecciones Intravenosas , Isótopos de Yodo/administración & dosificación , Riñón/diagnóstico por imagen , Hepatopatías/diagnóstico por imagen , Ratones , Ratones Endogámicos C3H , Fantasmas de Imagen , Dosis de Radiación , Reproducibilidad de los Resultados , Bazo/diagnóstico por imagen , Factores de Tiempo , Tomógrafos Computarizados por Rayos X , Triglicéridos/administración & dosificación
17.
J Nucl Med ; 49(2): 255-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18199623

RESUMEN

UNLABELLED: The integrin alpha(v)beta(3) is a key player in angiogenesis and metastasis. Our aim was to study the uptake patterns of the alpha(v)beta(3)-selective PET tracer (18)F-galacto-RGD in invasive ductal breast cancer. METHODS: Sixteen patients with primary (n = 12) or metastasized breast cancer (n = 4) were examined with (18)F-galacto-RGD PET. Standardized uptake values (SUVs) were derived by region-of-interest analysis, and immunohistochemistry of alpha(v)beta(3) expression was performed (n = 5). RESULTS: (18)F-Galacto-RGD PET identified all invasive carcinomas, with SUVs from 1.4 to 8.7 (mean +/- SD, 3.6 +/- 1.8; tumor-to-blood and tumor-to-muscle ratios, 2.7 +/- 1.6 and 6.2 +/- 2.2, respectively). Lymph-node metastases were detected in 3 of 8 patients (mean SUV, 3.3 +/- 0.8). SUVs in distant metastases were heterogeneous (2.9 +/- 1.4). Immunohistochemistry confirmed alpha(v)beta(3) expression predominantly on microvessels (5/5) and, to a lesser extent, on tumor cells (3/5). CONCLUSION: Our results suggest generally elevated and highly variable alpha(v)beta(3) expression in human breast cancer lesions. Consequently, further imaging studies with (18)F-galacto-RGD PET in breast cancer patients for assessment of angiogenesis or planning of alpha(v)beta(3)-targeted therapies are promising.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Carcinoma Ductal/diagnóstico por imagen , Carcinoma Ductal/metabolismo , Galactosa/análogos & derivados , Integrina alfaVbeta3/metabolismo , Péptidos Cíclicos/farmacocinética , Biomarcadores de Tumor/metabolismo , Femenino , Galactosa/farmacocinética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
18.
Clin Cancer Res ; 13(22 Pt 1): 6610-6, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18006761

RESUMEN

PURPOSE: [(18)F]Galacto-RGD has been developed for positron emission tomography (PET)-imaging of alphavbeta3 expression, a receptor involved in angiogenesis and metastasis. Our aim was to study the feasibility of PET imaging with [(18)F]Galacto-RGD in patients with squamous cell carcinoma of the head and neck (SCCHN). EXPERIMENTAL DESIGN: Eleven patients with primary diagnosis of SCCHN were examined. After injection of 140 to 200 MBq [(18)F]Galacto-RGD, static emission scans 60 min post injection from the head to the abdomen (n = 11) and dynamic scans >60 min covering the tumor region (n = 6) for kinetic modeling were acquired. Standardized uptake values (SUV) were measured in tumors, muscle and oral mucosa. Immunohistochemistry was done using an alphavbeta3-specific antibody (n = 7). Image fusion with magnetic resonance imaging and/or computed tomography (CT) scans (n = 8) and calculation of tumor subvolumes based on SUVs was done using the iPlan software (BrainLAB). RESULTS: [(18)F]Galacto-RGD PET identified 10 of 12 tumors, with SUVs ranging from 2.2 to 5.8 (mean, 3.4 +/- 1.2). Two tumors <5 mm were missed. Tumor/blood and tumor/muscle ratios were 2.8 +/- 1.1 and 5.5 +/- 1.6, respectively. Tumor kinetics was consistent with a two-tissue compartmental model with reversible specific binding. Immunohistochemistry confirmed alphavbeta3 expression in all tumors with alphavbeta3 being located on the microvessels in all specimens and additionally on tumor cells in one specimen. Image fusion of [(18)F]Galacto-RGD PET with magnetic resonance imaging/multislice CT and definition of tumor subvolumes was feasible in all cases. CONCLUSIONS: [(18)F]Galacto-RGD PET allows for specific imaging of alphavbeta3 expression in SCCHN with good contrast. Image fusion and definition of tumor subvolumes is feasible. This technique might be used for the assessment of angiogenesis and for planning and response evaluation of alphavbeta3-targeted therapies.


Asunto(s)
Carcinoma de Células Escamosas/irrigación sanguínea , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Integrina alfaVbeta3/análisis , Neovascularización Patológica/diagnóstico , Tomografía de Emisión de Positrones/métodos , Adulto , Femenino , Galactosa/análogos & derivados , Humanos , Masculino , Persona de Mediana Edad , Péptidos Cíclicos , Radiofármacos
19.
J Nucl Med ; 48(6): 973-80, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17536108

RESUMEN

UNLABELLED: We evaluated the predictive value of PET using the hypoxia tracer (18)F-fluoroazomycin arabinoside ((18)F-FAZA) for success of radiotherapy in combination with tirapazamine, a specific cytotoxin for hypoxic cells. METHODS: Imaging was performed on EMT6 tumor-bearing nude mice before allocating mice into 4 groups: radiochemotherapy (RCT: 8 fractions of 4.5 Gy within 4 d combined with tirapazamine, 14 mg/kg), radiotherapy alone (RT), chemotherapy alone (tirapazamine) (CHT), or control. Treatment success was assessed by several tumor growth assays, including tumor growth time from 70 to 500 microL and absolute growth delay (aGD). The median pretreatment (18)F-FAZA tumor-to-background ratio served as a discriminator between "hypoxic" and "normoxic" tumors. RESULTS: The mean tumor growth was significantly accelerated in hypoxic control tumors (growth time from 70 to 500 microL, 11.0 d) compared with normoxic control tumors (growth time from 70 to 500 microL, 15.6 d). Whereas RT delayed tumor growth regardless of the level of hypoxia, an additive beneficial therapeutic effect of tirapazamine to RT was observed only in hypoxic tumors (aGD, 12.9 d) but not in normoxic tumors (aGD, 6.0 d). CONCLUSION: This study provides compelling evidence that hypoxia imaging using (18)F-FAZA PET is able to predict the success of RCT of tumor-bearing mice using the hypoxia-activated chemotherapeutic agent tirapazamine. Pretreatment (18)F-FAZA PET, therefore, offers a way for the individualization of tumor treatment involving radiation. The data suggest that by reserving hypoxia-directed therapy to tumors with high (18)F-FAZA uptake, improvement of the therapeutic ratio is possible, as the therapeutic effect of tirapazamine seems to be restricted to hypoxic tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Nitroimidazoles , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radiofármacos , Triazinas/uso terapéutico , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Terapia Combinada , Femenino , Radioisótopos de Flúor , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/radioterapia , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Tomografía de Emisión de Positrones/métodos , Tirapazamina , Trasplante Heterólogo , Imagen de Cuerpo Entero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...