RESUMEN
Poly(ADP-ribose)polymerase-1 inhibitor (PARPi) AZD2461 was designed to be a weak P-glycoprotein (P-gp) analogue of FDA approved olaparib. With this chemical property in mind, we utilized the AZD2461 ligand architecture to develop a CNS penetrant and PARP-1 selective imaging probe, in order to investigate PARP-1 mediated neuroinflammation and neurodegenerative diseases, such as Alzheimer's and Parkinson's. Our work led to the identification of several high-affinity PARPi, including AZD2461 congener 9e (PARP-1 IC50â¯=â¯3.9⯱â¯1.2â¯nM), which was further evaluated as a potential 18F-PET brain imaging probe. However, despite the similar molecular scaffolds of 9e and AZD2461, our studies revealed non-appreciable brain-uptake of [18F]9e in non-human primates, suggesting AZD2461 to be non-CNS penetrant.
Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Ftalazinas/farmacología , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/agonistas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Radioisótopos de Flúor/química , Humanos , Macaca mulatta , Masculino , Ratones Endogámicos BALB C , Ftalazinas/síntesis química , Piperidinas/síntesis químicaRESUMEN
BACKGROUND: Altered myocardial energy metabolism has been linked to worsening of RV function in pulmonary arterial hypertension (PAH). The aim of this study was to evaluate RV glucose and fatty acid metabolism in vivo in a rat model of PAH using positron emission tomography (PET) and investigate the effects of Macitentan on RV substrate utilization. METHODS: PAH was induced in male Sprague-Dawley rats by a single subcutaneous injection of Sugen 5416 (20 mg/kg) followed by 3 weeks of hypoxia (10% oxygen). At week 5 post-injection, the PAH rats were randomized to Macitentan (30 mg/kg daily) treatment or no treatment. Substrate utilization was serially assessed 5 and 8 weeks post-injection with 2-[18F]fluoro-2-deoxyglucose (FDG) and 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid (FTHA) PET for glucose and fatty acid metabolism respectively and correlated with in vivo functional measurements. RESULTS: PAH induction resulted in a 2.5-fold increase in RV FDG uptake (standardized uptake value (SUV) of normal control: 1.6 ± 0.4, week 5: 4.1 ± 1.9, week 8: 4.0 ± 1.6, P < 0.05 for all groups vs. control). RV FTHA showed twofold increased uptake at week 5 (SUV control: 1.50 ± 0.39, week 5: 3.06 ± 1.10, P = 0.03). Macitentan significantly decreased RV FDG uptake at 8 weeks (SUV: 2.5 ± 0.9, P = 0.04), associated with improved RV ejection fraction and reduced RV systolic pressure, while FTHA uptake was maintained. CONCLUSION: PAH is associated with metabolic changes in the RV, characterized by a marked increase in FDG and FTHA uptake. Macitentan treatment reduced PAH severity and was associated with a decrease in RV FDG uptake and improved RV function.
Asunto(s)
Antagonistas de los Receptores de Endotelina/farmacología , Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Pirimidinas/farmacología , Sulfonamidas/farmacología , Función Ventricular Derecha/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Hipertensión Pulmonar/fisiopatología , Hipoxia , Masculino , Ratas , Ratas Sprague-DawleyRESUMEN
BACKGROUND: Sphingosine-1-phosphate receptor 1 (S1PR1) is highly expressed in vascular smooth muscle cells from intimal lesions. PET imaging using S1PR1 as a biomarker would increase our understanding of its role in vascular pathologies including in-stent restenosis. METHODS: The S1PR1 compound TZ3321 was synthesized for in vitro characterization and labeled with Carbon-11 for in vivo studies. The biodistribution of [11C]TZ3321 was evaluated in normal mice; microPET and immunohistochemistry (IHC) studies were performed using a murine femoral artery wire-injury model of restenosis. RESULTS: The high potency of TZ3321 for S1PR1 (IC 50 = 2.13 ± 1.63 nM), and high selectivity (>1000 nM) for S1PR1 over S1PR2 and S1PR3 were confirmed. Biodistribution data revealed prolonged retention of [11C]TZ3321 in S1PR1-enriched tissues. MicroPET imaging of [11C]TZ3321 showed higher uptake in the wire-injured arteries of ApoE-/- mice than in injured arteries of wild-type mice (SUV 0.40 ± 0.06 vs 0.28 ± 0.04, n = 6, P < .001); FDG-PET showed no difference (SUV 0.98 ± 0.04 vs 0.94 ± 0.01, n = 6, P > .05). Post-PET autoradiography showed >4-fold higher [11C]TZ3321 retention in the injured artery of ApoE-/- mice than in wild-type mice. Subsequent IHC staining confirmed higher expression of S1PR1 in the neointima of the injured artery of ApoE-/- mice than in wild-type mice. CONCLUSIONS: This preliminary study supports the potential use of PET for quantification of the S1PR1 expression as a biomarker of neointimal hyperplasia.
Asunto(s)
Radioisótopos de Carbono/farmacocinética , Oclusión de Injerto Vascular/diagnóstico por imagen , Oclusión de Injerto Vascular/metabolismo , Aumento de la Imagen/métodos , Tomografía de Emisión de Positrones/métodos , Receptores de Lisoesfingolípidos/metabolismo , Animales , Estudios de Factibilidad , Femenino , Humanos , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Diagnóstico Molecular/métodos , Especificidad de Órganos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Receptores de Esfingosina-1-Fosfato , Distribución TisularRESUMEN
BACKGROUND: Inflammation and angiogenesis play an important role in atherosclerotic plaque rupture. Therefore, molecular imaging of these processes could be used for determination of rupture-prone atherosclerotic plaques. αvß3 integrin is involved in the process of angiogenesis. Targeted imaging of αvß3 integrin has been shown to be possible in previous studies on tumor models, using radiolabeled arginine-glycine-aspartate (RGD). Our aim was to investigate feasibility of ex vivo detection of αvß3 integrin in carotid endarterectomy (CEA) specimens. METHODS AND RESULTS: Nineteen CEA specimens were incubated in 5 MBq [18F]-RGD-K5 for 1 hour followed by 1 hour emission microPET scan. The results were quantified in 4 mm wide segments as percent incubation dose per gram (%Inc/g). Segmental-to-total ratio was calculated and presence of αvß3 integrin and endothelial cells in each segment was confirmed by immunohistochemical staining for CD31 and αvß3 integrin, respectively. [18F]-RGD-K5 uptake was heterogeneously distributed across CEA specimens and was localized within the vessel wall. Significant correlations were observed between segmental-to-total ratio with αvß3 integrin staining score (r = 0.58, P = .038) and CD31 staining score (ρ = 0.67, P < .002). CONCLUSION: This study showed the feasibility of integrin imaging by determination of αvß3 integrin expression in human atherosclerotic plaques.
Asunto(s)
Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/metabolismo , Fluorodesoxiglucosa F18/farmacocinética , Integrina alfaVbeta3/metabolismo , Imagen Molecular/métodos , Oligopéptidos/farmacocinética , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular/métodos , Cintigrafía , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y EspecificidadRESUMEN
Background: Acute liver injury (ALI) that progresses into acute liver failure (ALF) is a life-threatening condition with an increasing incidence and associated costs. Acetaminophen (N-acetyl-p-aminophenol, APAP) overdosing is among the leading causes of ALI and ALF in the Northern Hemisphere. Brain dysfunction defined as hepatic encephalopathy is one of the main diagnostic criteria for ALF. While neuroinflammation and brain metabolic alterations significantly contribute to hepatic encephalopathy, their evaluation at early stages of ALI remained challenging. To provide insights, we utilized post-mortem analysis and non-invasive brain micro positron emission tomography (microPET) imaging of mice with APAP-induced ALI. Methods: Male C57BL/6 mice were treated with vehicle or APAP (600 mg/kg, i.p.). Serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), liver damage (using H&E staining), hepatic and serum IL-6 levels, and hippocampal IBA1 (using immunolabeling) were evaluated at 24h and 48h. Vehicle and APAP treated animals also underwent microPET imaging utilizing a dual tracer approach, including [11C]-peripheral benzodiazepine receptor ([11C]PBR28) to assess microglia/astrocyte activation and [18F]-fluoro-2-deoxy-2-D-glucose ([18F]FDG) to assess energy metabolism. Brain images were pre-processed and evaluated using conjunction and individual tracer uptake analysis. Results: APAP-induced ALI and hepatic and systemic inflammation were detected at 24h and 48h by significantly elevated serum ALT and AST levels, hepatocellular damage, and increased hepatic and serum IL-6 levels. In parallel, increased microglial numbers, indicative for neuroinflammation were observed in the hippocampus of APAP-treated mice. MicroPET imaging revealed overlapping increases in [11C]PBR28 and [18F]FDG uptake in the hippocampus, thalamus, and habenular nucleus indicating microglial/astroglial activation and increased energy metabolism in APAP-treated mice (vs. vehicle-treated mice) at 24h. Similar significant increases were also found in the hypothalamus, thalamus, and cerebellum at 48h. The individual tracer uptake analyses (APAP vs vehicle) at 24h and 48h confirmed increases in these brain areas and indicated additional tracer- and region-specific effects including hippocampal alterations. Conclusion: Peripheral manifestations of APAP-induced ALI in mice are associated with brain neuroinflammatory and metabolic alterations at relatively early stages of disease progression, which can be non-invasively evaluated using microPET imaging and conjunction analysis. These findings support further PET-based investigations of brain function in ALI/ALF that may inform timely therapeutic interventions.
RESUMEN
Despite an apparently silent imaging, some patients with mild traumatic brain injury (TBI) experience cognitive dysfunctions, which may persist chronically. Brain changes responsible for these dysfunctions are unclear and commonly overlooked. It is thus crucial to increase our understanding of the mechanisms linking the initial event to the functional deficits, and to provide objective evidence of brain tissue alterations underpinning these deficits. We first set up a murine model of closed-head controlled cortical impact, which provoked persistent cognitive and sensorimotor deficits, despite no evidence of brain contusion or bleeding on MRI, thus recapitulating features of mild TBI. Molecular MRI for P-selectin, a key adhesion molecule, detected no sign of cerebrovascular inflammation after mild TBI, as confirmed by immunostainings. By contrast, in vivo PET imaging with the TSPO ligand [18F]DPA-714 demonstrated persisting signs of neuroinflammation in the ipsilateral cortex and hippocampus after mild TBI. Interestingly, immunohistochemical analyses confirmed these spatio-temporal profiles, showing a robust parenchymal astrogliosis and microgliosis, at least up to 3 weeks post-injury in both the cortex and hippocampus. In conclusion, we show that even one single mild TBI induces long-term behavioural deficits, associated with a persistent neuro-inflammatory status that can be detected by PET imaging.
Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Animales , Humanos , Ratones , Encéfalo , Conmoción Encefálica/complicaciones , Conmoción Encefálica/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Enfermedades Neuroinflamatorias , Tomografía de Emisión de Positrones/métodos , Receptores de GABARESUMEN
BACKGROUND: Brain metabolic alterations and neuroinflammation have been reported in several peripheral inflammatory conditions and present significant potential for targeting with new diagnostic approaches and treatments. However, non-invasive evaluation of these alterations remains a challenge. METHODS: Here, we studied the utility of a micro positron emission tomography (microPET) dual tracer ([11C]PBR28 - for microglial activation and [18F]FDG for energy metabolism) approach to assess brain dysfunction, including neuroinflammation in murine endotoxemia. MicroPET imaging data were subjected to advanced conjunction and individual analyses, followed by post-hoc analysis. RESULTS: There were significant increases in [11C]PBR28 and [18F]FDG uptake in the hippocampus of C57BL/6 J mice 6 h following LPS (2 mg/kg) intraperitoneal (i.p.) administration compared with saline administration. These results confirmed previous postmortem observations. In addition, patterns of significant simultaneous activation were demonstrated in the hippocampus, the thalamus, and the hypothalamus in parallel with other tracer-specific and region-specific alterations. These changes were observed in the presence of robust systemic inflammatory responses manifested by significantly increased serum cytokine levels. CONCLUSIONS: Together, these findings demonstrate the applicability of [11C]PBR28 - [18F]FDG dual tracer microPET imaging for assessing neuroinflammation and brain metabolic alterations in conditions "classically" characterized by peripheral inflammatory and metabolic pathogenesis.
RESUMEN
PURPOSE: The dopamine transporter (DAT) is a marker of the occurrence and development of Parkinson's disease (PD) and other diseases with nigrostriatal degeneration. 2ß-Carbomethoxy-3ß-(4-chlorophenyl)-8-(2-[18F]-fluoroethyl)nortropane ([18F]FECNT), an 18F-labelled tropane derivative, was reported to be a useful positron-emitting probe for DAT. However, the rapid formation of brain-penetrating radioactive metabolites is an impediment to the proper quantitation of DAT in PET studies with [18F]FECNT. Deuterium-substituted analogues have presented better in vivo stability to reduce metabolites. This study aimed to synthesize a deuterium-substituted DAT radiotracer, [18F]FECNT-d4, and to make a preliminary investigation of its properties as a DAT tracer in vivo. PROCEDURES: The ligand [18F]FECNT-d4 was obtained by one-step radiolabelling reaction. The lipophilicity was measured by the shake-flask method. Binding properties of [18F]FECNT-d4 were estimated by in vitro binding assay, biodistribution, and microPET imaging in rats. In vivo stability of [18F]FECNT-d4 was estimated by radio-HPLC. RESULTS: [18F]FECNT-d4 was synthesized at an average activity yield of 46 ± 17 % (n = 15) and the molar activity was 67 ± 12 GBq/µmol. The deuterated tracer showed suitable lipophilicity and the ability to penetrate the blood-brain barrier (brain uptake of 1.72 % ID at 5 min). [18F]FECNT-d4 displayed a high binding affinity for DAT comparable to that of [18F]FECNT in rat striatum homogenates. Biodistribution results in normal rats showed that [18F]FECNT-d4 exhibited a higher ratio of the target to non-target (striatum/cerebellum) at 15 min post administration (5.00 ± 0.44 vs 3.84 ± 0.24 for [18F]FECNT-d4 vs [18F]FECNT). MicroPET imaging studies of [18F]FECNT-d4 in normal rats showed that the ligand selectively localized to DAT-rich striatal regions and the accumulation could be blocked with DAT inhibitor. Furthermore, in the unilateral PD model rat, a significant reduction of the signal was found in the lesioned side relative to the unlesioned side. Striatal standardized uptake value of [18F]FECNT-d4 remained ~4.02 in the striatum between 5 and 20 min, whereas that of [18F]FECNT fell rapidly from 4.11 to 2.95. Radio-HPLC analysis of the plasma demonstrated better in vivo stability of [18F]FECNT-d4 than [18F]FECNT. CONCLUSION: The deuterated compound [18F]FECNT-d4 may serve as a promising PET imaging agent to assess DAT-related disorders.
Asunto(s)
Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Radioisótopos de Flúor , Nortropanos , Tomografía de Emisión de Positrones/métodos , Animales , Deuterio , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/análisis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Masculino , Nortropanos/química , Nortropanos/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución TisularRESUMEN
Introduction: Previous studies have shown that peptides containing the asparagine-glycine-arginine (NGR) sequence can specifically bind to CD13 (aminopeptidase N) receptor, a tumor neovascular biomarker that is over-expressed on the surface of angiogenic blood vessels and various tumor cells, and it plays an important role in angiogenesis and tumor progression. In the present study, we aimed to evaluate the efficacy of a gallium-68 (68Ga)-labeled dimeric cyclic NGR (cNGR) peptide as a new molecular probe that binds to CD13 in vitro and in vivo. Materials and Methods: A dimeric cNGR peptide conjugated with 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) was synthesized and labeled with 68Ga. In vitro uptake and binding analyses of the 68Ga- DOTA-c(NGR)2 were performed in two ovarian tumor cell lines, ES2 and SKOV3, which had different CD13 expression patterns. An in vivo biodistribution study was performed in normal mice, and micro positron emission tomography (PET) imaging was conducted in nude mice bearing ES2 and SKOV3 tumors. Results: 68Ga-DOTA-c(NGR)2 was prepared with high radiochemical purity (>95%), and it was stable both in saline at room temperature and in bovine serum at 37°C for 3 h. In vitro studies showed that the uptake of 68Ga-DOTA-c(NGR)2 in ES2 cells was higher compared with SKOV3 cells, and such uptake could be blocked by the cold DOTA-c(NGR)2. Biodistribution studies demonstrated that 68Ga-DOTA-c(NGR)2 was rapidly cleared from blood and mainly excreted from the kidney. MicroPET imaging of ES2 tumor xenografts showed the focal uptake of 68Ga-DOTA-c(NGR)2 in tumors from 1 to 1.5 h post-injection. The high-contrast tumor visualization occurred at 1 h, corresponding to the highest tumor/background ratio of 10.30±0.26. The CD13-specific tumor targeting of the 68Ga-DOTA-c(NGR)2 was further supported by the reduced uptake of the probe in ES2 tumors by co-injection of the unlabeled cold peptide. In SKOV3 tumor models, the tumor was not obviously visible under the same imaging conditions. Conclusions: 68Ga-DOTA-c(NGR)2 was easily synthesized, and it showed favorable CD13-specific targeting ability by in vitro data and microPET imaging with ovarian cancer xenografts. Collectively, 68Ga-DOTA-c(NGR)2 might be a potential PET imaging probe for non-invasive evaluation of the CD13 receptor expression in tumors.
RESUMEN
Organically modified silica (ORMOSIL) nanoparticles have found many biomedical applications and emerged as biocompatible and efficient carriers of diagnostic and therapeutic agents, such as fluorophores, drugs, and DNA. Herein, we describe two major in vivo studies exemplifying the use of these nanoparticles as carriers of active agents. The first part of this report details a systemic administration and biodistribution of radiolabeled and fluorophore-incorporated ORMOSIL nanoparticles in mice. The second part of this report focuses on the use of ORMOSIL nanoparticles as carriers of plasmid DNA for nonviral gene delivery to the mouse brain. We provide detailed protocols describing preparation and characterization of ORMOSIL nanoparticles, methods used for loading the particles with active agents (e.g., radioimaging agents, plasmid DNA), and in vivo administration of the particles.
Asunto(s)
Encéfalo/metabolismo , Plásmidos/administración & dosificación , Siloxanos/química , Animales , Disponibilidad Biológica , Sistemas de Liberación de Medicamentos , Femenino , Colorantes Fluorescentes/química , Radioisótopos de Yodo/química , Ratones , Nanopartículas , Plásmidos/genética , Plásmidos/farmacocinética , Siloxanos/farmacocinéticaRESUMEN
PURPOSE: A novel radiolabeled probe 1(17[18F]fluoro3,6,9,12,15pentaoxaheptadecyl1H1,2,3triazole testosterone ([18F]FPTT) was synthesized and evaluated for PET imaging of progesterone receptor (PR)-positive breast cancer. METHODS: The ethinyl group of ethisterone, a PR targeting pharmacophore, was coupled with azide modified PEG-OTs by click chemistry to obtain the labeling precursor. The final [18F]FPTT was synthesized by a one-step nucleophilic substitution reaction with 18F. The in vitro stabilities of [18F]FPTT in saline or rat serum were determined after 2â¯h incubation. Then the in vitro cell binding, ex vivo biodistribution and in vivo imaging of [18F]FPTT were further investigated to evaluate the PR targeting ability and feasibility for the diagnosis of PR-positive breast cancer with PET imaging. RESULTS: [18F]FPTT was obtained in high decay-corrected radiochemical yield (78⯱â¯9%) at the end of synthesis. It had high radiochemical purity (>98%) after HPLC purification and good in vitro stability. The molar activity of [18F]FPTT was calculated as 17â¯GBq/µmol. The microPET imaging of [18F]FPTT in tumor-bearing mice showed much higher tumor uptake in PR-positive MCF-7 tumor (3.9⯱â¯0.20%ID/g) than that of PR-negative MDA-MB-231 tumor (1.3⯱â¯0.08%ID/g). The high MCF-7 tumor uptake could be specifically inhibited by blocking with ethisterone (1.3⯱â¯0.11%ID/g) or [19F]FPTT (2.20⯱â¯0.17%ID/g), respectively. The biodistribution in estrogen-primed female SD rats of [18F]FPTT showed high uterus and ovary uptakes (8.31⯱â¯1.74%ID/g and 3.79⯱â¯0.82%ID/g at 1â¯h post-injection). The specific uptakes of uterus and ovary in normal rats were 3.52⯱â¯0.29%ID/g and 3.22⯱â¯0.50%ID/g respectively and could be inhibited by co-injecting of ethisterone. CONCLUSION: A novel [18F]FPTT probe based on ethisterone modification could be a potential diagnostic agent for PR-positive breast cancer.
Asunto(s)
Neoplasias de la Mama/patología , Etisterona/química , Radioisótopos de Flúor/farmacocinética , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Receptores de Progesterona/metabolismo , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Radioquímica , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de XenoinjertoRESUMEN
Postoperative Cognitive Dysfunction (POCD) is a complication that has been observed in a subset of adult and elderly individuals after general anesthesia and surgery. Although the pathogenesis of POCD is largely unknown, a growing body of preclinical research suggests that POCD may be caused by general anesthesia. A significant amount of research has examined the effects of general anesthesia on neurocognitive function in rodents, yet no studies have assessed the adverse effects of general anesthesia on brain function in adult nonhuman primates. Thus, this study sought to determine the effects of an extended exposure to sevoflurane anesthesia on cognitive function and neural inflammation in adult rhesus macaques. Five adult rhesus macaques (16-17 years of age) were exposed to sevoflurane anesthesia for 8 h and, and micro-positron emission tomography (PET)/computed tomography (CT) imaging and a battery of operant tasks were used to assess the effects of anesthesia exposure on 18F-labeled fluoroethoxybenzyl-N-(4-phenoxypyridin-3-yl) acetamide ([18F]-FEPPA) uptake, a biomarker of microglia activation, and aspects of complex cognitive function. Exposure to sevoflurane anesthesia for 8 h did not increase [18F]-FEPPA uptake in the adult monkey brain. Sevoflurane anesthesia significantly decreased accuracy (mean difference = 22.79) on a learning acquisition task 6 days after exposure [t(3) = 6.92, p = 0.006], but this effect did not persist when measured 1 week and 2 weeks after additional exposures. Further, sevoflurane anesthesia had no impact on performance in 4 additional cognitive tasks. These data suggest that exposure to anesthesia alone may not be sufficient to cause persistent POCD in adult populations.
Asunto(s)
Anestésicos por Inhalación/toxicidad , Encéfalo/efectos de los fármacos , Encefalitis/inducido químicamente , Aprendizaje/efectos de los fármacos , Microglía/efectos de los fármacos , Sevoflurano/toxicidad , Animales , Encéfalo/metabolismo , Encéfalo/patología , Condicionamiento Operante/efectos de los fármacos , Femenino , Macaca mulatta , Masculino , Microglía/metabolismoRESUMEN
BACKGROUND: With the advances in radiopharmaceutical research, the development of image-guided therapy has become a major interest. While the development of theranostic nanotherapeutics is frequently associated with cancer chemotherapy, phototherapy and radiotherapy, there is little information available on the in vivo monitoring of gene delivery systems and the application of image-guided approach in gene therapy. The goal of this work was to determine the in vivo behavior of DNA delivery nanosystems - based on cationic gemini surfactants - designed for image-guided gene therapy. We tested the feasibility of monitoring tumor accumulation of gene delivery nanoparticles by positron emission tomography. METHODS: To be able to conjugate radiotracers to the nanoparticles, a deferoxamine-modified gemini surfactant was synthesized, DNA-containing lipoplex nanoparticles were formulated, and radiolabeled with Zirconium-89 (89Zr). The pharmacokinetics and biodistribution of 89Zr labeled surfactant and 89Zr labeled nanoparticles were monitored in mice by microPET/CT imaging and ex vivo gamma counting. RESULTS: Modification of the nanoparticles with deferoxamine did not alter their physicochemical properties. The radiolabeled nanoparticles (labeling efficiency of 95±3%) were stable in PBS and serum. The biological half-life of the 89Zr labeled nanoparticles was significantly higher compared to 89Zr labeled surfactant. As expected, the nanoparticles had significantly higher liver accumulation than the radiolabeled surfactant alone and lower kidney accumulation. Tumor uptake was detected at 2 hours post injection and decreased throughout the 3-day monitoring. CONCLUSION: We propose that radiolabeling DNA delivery lipoplex nanosystems is a promising approach for the design and optimization of image-guided nanomedicines, especially in the context of cancer gene therapy.
Asunto(s)
Técnicas de Transferencia de Gen , Imagenología Tridimensional , Lípidos/química , Nanopartículas/química , Radioisótopos/química , Circonio/química , Animales , Supervivencia Celular , Deferoxamina/química , Regulación de la Expresión Génica , Terapia Genética , Ratones Desnudos , Nanopartículas/ultraestructura , Neoplasias/metabolismo , Neoplasias/patología , Tomografía de Emisión de Positrones , Radioisótopos/farmacocinética , Radiofármacos/farmacocinética , Tensoactivos/química , Distribución Tisular , Circonio/farmacocinéticaRESUMEN
We report the synthesis and preclinical evaluation of a 11C-labeled probe to target melanoma using PET. Methods: The target compound 4-11C-methoxy N-(2-diethylaminoethyl) benzamide (4-11C-MBZA) was prepared via the 11C-methylation of 4-hydroxy N-(2-diethylaminoethyl) benzamide (4-HBZA). The in vitro binding was performed using B16F1 (melanoma cells), MCF-10A (breast epithelial cells), and MDA-MB 231 (breast cancer cells). The internalization studies were conducted using B16F1 cells. In vivo biodistribution and small-animal PET imaging were performed in mice bearing B16F1 melanoma tumor xenografts. Results: The target compound 4-11C-MBZA was prepared in 46% ± 7% radiochemical yields by reacting 11C-methyltriflate with 4-HBZA followed by high-performance liquid chromatography purification. The specific activity of this compound was 853 ± 29.6 GBq/µmol (23 ± 0.8 Ci/µmol). The binding of 4-11C-MBZA to B16F1, MCF-10A, and MDA-MB-231 cells was 6.41% ± 1.28%, 1.51% ± 0.17%, and 0.30% ± 0.17%, respectively. Internalization studies using B16F1 melanoma cells show 60.7% of the cell-bound activity was internalized. Results from biodistribution studies show a rapid and high uptake of radioactivity in the tumor, with uptake levels reaching 5.85 ± 0.79 and 8.13 ± 1.46 percentage injected dose per gram at 10 and 60 min, respectively. Low uptake in normal tissues in conjunction with high tumor uptake resulted in high tumor-to-tissue ratios. On small-animal PET images, the tumor was clearly delineated soon after 4-11C-MBZA injection and tumor uptake reached 4.2 percentage injected dose per gram by 20 min. These preclinical evaluations show a high propensity of 4-11C-MBZA toward melanoma tumor. Conclusion: We successfully developed 4-11C-MBZA as a PET imaging probe, displaying properties advantageous over those for its 18F analogs. These preclinical evaluation results demonstrate the clinical potential of this probe to selectively target melanoma.
Asunto(s)
Benzamidas/síntesis química , Benzamidas/farmacocinética , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Técnicas de Sonda Molecular , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Marcaje Isotópico , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Radiofármacos/farmacocinética , Distribución TisularRESUMEN
BACKGROUND: αVß3, αVß5 and α5ß1 integrins are known to be involved in carcinogenesis and are overexpressed in many types of tumours compared to healthy tissues; thereby they have been selected as promising therapeutic targets. Positron emission tomography (PET) is providing a unique non-invasive screening assay to discriminate which patient is more prone to benefit from antiangiogenic therapies, and extensive research has been carried out to develop a clinical radiopharmaceutical that binds specifically to integrin receptors. We recently reported the synthesis of a new 18F-labelled RGD peptide prepared by 2-cyanobenzothiazole (CBT)/1,2-aminothiol conjugation. This study aims at characterising the preclinical biologic properties of this new tumour-targeting ligand, named [18F]FPyPEGCBT-c(RGDfK).The in vitro binding properties of [18F]FPyPEGCBT-c(RGDfK) were analysed by standard binding assay in U-87 MG and SKOV-3 cancer models and its selectivity towards integrins by siRNA depletions. Its preclinical potential was studied in mice bearing subcutaneous tumours by ex vivo biodistribution studies and in vivo microPET/CT imaging. RESULTS: In vitro, FPyPEGCBT-c(RGDfK) efficiently bound RGD-recognising integrins as compared to a control c(RGDfV) peptide (IC50 = 30.8 × 10-7 M vs. 6.0 × 10-7 M). [18F]FPyPEGCBT-c(RGDfK) cell uptake was mediated by an active transport through binding to αV, ß3 and ß5 but not to ß1 subunits. In vivo, this new tracer demonstrated specific tumour uptake with %ID/g of 2.9 and 2.4 in U-87 MG and SKOV-3 tumours 1 h post injection. Tumour-to-muscle ratios of 4 were obtained 1 h after intravenous administration of the tracer allowing good visualisation of the tumours. However, unfavourable background accumulation and high hepatobiliary excretion were observed. CONCLUSION: [18F]FPyPEGCBT-c(RGDfK) specifically detects tumours expressing RGD-recognising integrin receptors in preclinical studies. Further optimisation of this radioligand may yield candidates with improved imaging properties and would warrant the further use of this efficient labelling technique for alternative targeting vectors.
RESUMEN
BACKGROUND: The purpose of this study was to develop a positron emission tomography (PET) imaging probe that is easy to synthesize and selectively targets melanoma in vivo. Herein, we report the synthesis and preclinical evaluation of N-(2-diethylaminoethyl) 4-[18F]Fluoroethoxy benzamide (4-[18F]FEBZA). A one-step synthesis was developed to prepare 4-[18F]FEBZA in high radiochemical yields and specific activity. The binding affinity, the in vitro binding, and internalization studies were performed using B16F1 melanoma cell line. The biodistribution studies were performed in C57BL/6 normal mice, C57BL/6 mice bearing B16F1 melanoma tumor xenografts, and nu/nu athymic mice bearing HT-29 human adenocarcinoma tumor and C-32 amelanotic melanoma tumor xenografts. MicroPET studies were performed in mice bearing B16F1 and HT-29 tumor xenografts. RESULTS: 4-[18F]FEBZA was prepared in 53 ± 14% radiochemical yields and a specific activity of 8.7 ± 1.1 Ci/µmol. The overall synthesis time for 4-[18F]FEBZA was 54 ± 7 min. The in vitro binding to B16F1 cells was 60.03 ± 0.48% after 1 h incubation at 37 °C. The in vivo biodistribution studies show a rapid and high uptake of F-18 in B16F1 tumor with 8.66 ± 1.02%IA/g in this tumor at 1 h. In contrast, the uptake at 1 h in HT-29 colorectal adenocarcinoma and C-32 amelanotic melanoma tumors was significantly lower with 3.68 ± 0.47%IA/g and 3.91 ± 0.23%IA/g in HT-29 and C-32 tumors, respectively. On microPET images, the melanoma tumor was clearly visible by 10 min post-injection and the intensity in the tumor continued to increase with time. In contrast, the HT-29 tumor was not visible on the microPET scans. CONCLUSIONS: A rapid and facile synthesis of 4-[18F]FEBZA is developed. This method offers a reliable production of 4-[18F]FEBZA in high radiochemical yields and specific activity. A high binding affinity to melanoma cells and high uptake in tumor was noted. The microPET scan clearly delineates the melanoma tumor by 10 min post-injection. The results from these preclinical studies support the potential of 4-[18F]FEBZA as an effective probe to image melanoma.