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










Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 19(1): 152, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35705957

RESUMO

BACKGROUND: Inflammation is a potential risk factor of mental disturbance. FKBP5 that encodes FK506-binding protein 51 (FKBP51), a negative cochaperone of glucocorticoid receptor (GR), is a stress-inducible gene and has been linked to psychiatric disorders. Yet, the role of FKBP51 in the inflammatory stress-associated mental disturbance remained unclear. METHODS: Fkbp5-deficient (Fkbp5-KO) mice were used to study inflammatory stress by a single intraperitoneal injection of lipopolysaccharide (LPS). The anxiety-like behaviors, neuroimaging, immunofluorescence staining, immunohistochemistry, protein and mRNA expression analysis of inflammation- and neurotransmission-related mediators were evaluated. A dexamethasone drinking model was also applied to examine the effect of Fkbp5-KO in glucocorticoid-induced stress. RESULTS: LPS administration induced FKBP51 elevation in the liver and hippocampus accompanied with transient sickness. Notably, Fkbp5-KO but not wild-type (WT) mice showed anxiety-like behaviors 7 days after LPS injection (LPS-D7). LPS challenge rapidly increased peripheral and central immune responses and hippocampal microglial activation followed by a delayed GR upregulation on LPS-D7, and these effects were attenuated in Fkbp5-KO mice. Whole-brain [18F]-FEPPA neuroimaging, which target translocator protein (TSPO) to indicate neuroinflammation, showed that Fkbp5-KO reduced LPS-induced neuroinflammation in various brain regions including hippocampus. Interestingly, LPS elevated glutamic acid decarboxylase 65 (GAD65), the membrane-associated GABA-synthesizing enzyme, in the hippocampus of WT but not Fkbp5-KO mice on LPS-D7. This FKBP51-dependent GAD65 upregulation was observed in the ventral hippocampal CA1 accompanied by the reduction of c-Fos-indicated neuronal activity, whereas both GAD65 and neuronal activity were reduced in dorsal CA1 in a FKBP51-independent manner. GC-induced anxiety was also examined, which was attenuated in Fkbp5-KO and hippocampal GAD65 expression was unaffected. CONCLUSIONS: These results suggest that FKBP51/FKBP5 is involved in the systemic inflammation-induced neuroinflammation and hippocampal GR activation, which may contribute to the enhancement of GAD65 expression for GABA synthesis in the ventral hippocampus, thereby facilitating resilience to inflammation-induced anxiety.


Assuntos
Ansiedade/metabolismo , Glutamato Descarboxilase/metabolismo , Lipopolissacarídeos , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Ansiedade/patologia , Glucocorticoides/farmacologia , Glutamato Descarboxilase/genética , Hipocampo/metabolismo , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Receptores de GABA/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Ácido gama-Aminobutírico/metabolismo
2.
Neuropathol Appl Neurobiol ; 41(2): 245-57, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24861260

RESUMO

AIMS: Frontotemporal lobar degeneration (FTLD) is clinically and pathologically heterogeneous. Although associated with variations in MAPT, GRN and C9ORF72, the pathogenesis of these, and of other nongenetic, forms of FTLD, remains unknown. Epigenetic factors such as histone regulation by histone deacetylases (HDAC) may play a role in the dysregulation of transcriptional activity, thought to underpin the neurodegenerative process. METHODS: The distribution and intensity of HDACs 4, 5 and 6 was assessed semi-quantitatively in immunostained sections of temporal cortex with hippocampus, and cerebellum, from 33 pathologically confirmed cases of FTLD and 27 controls. RESULTS: We found a significantly greater intensity of cytoplasmic immunostaining for HDAC4 and HDAC6 in granule cells of the dentate gyrus in cases of FTLD overall compared with controls, and specifically in cases of FTLD tau-Picks compared with FTLD tau-MAPT and controls. No differences were noted between FTLD-TDP subtypes, or between the different genetic and nongenetic forms of FTLD. No changes were seen in HDAC5 in any FTLD or control cases. CONCLUSIONS: Dysregulation of HDAC4 and/or HDAC6 could play a role in the pathogenesis of FTLD-tau associated with Pick bodies, although their lack of immunostaining implies that such changes do not contribute directly to the formation of Pick bodies.


Assuntos
Giro Denteado/patologia , Degeneração Lobar Frontotemporal/enzimologia , Degeneração Lobar Frontotemporal/patologia , Histona Desacetilases/biossíntese , Proteínas Repressoras/biossíntese , Idoso , Feminino , Desacetilase 6 de Histona , Histona Desacetilases/análise , História do Século XVI , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteínas Repressoras/análise
3.
J Nucl Med ; 55(4): 678-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24639460

RESUMO

UNLABELLED: Overexpressed histone deacetylase (HDAC) activity has been linked with tumor initiation and progression that prompt the development of histone deacetylase inhibitors (HDACIs) as anticancer agents. HDACI was reported to be able to activate p21 promoter through the SP1 binding sites in the proximal region of p21(WAF1/CIP1) promoter. In this study, we established a p21(WAF1/CIP1) promoter-driven triple-fused reporter gene system (p21-3H) to evaluate the efficacy of HDACI and the ganciclovir (GCV)-mediated anticancer effect contributed by HDACI-induced and p21-driven truncated herpes simplex virus-1 thymidine kinase sr39 mutant (ttksr39) in vitro and in vivo. METHODS: The p21-3H construct was generated and stably or transiently transfected into H1299 cell lines. These cells were treated with trichostatin A or vorinostat (suberoylanilide hydroxamic acid [SAHA]) to evaluate the activation of p21 promoter-driven reporter gene expression by in vitro confocal fluorescence microscopy, luciferase assay, 2'-fluoro-2'-deoxyarabinofuranosyl-5-ethyluracil ((3)H-FEAU) cellular uptake, in vivo bioluminescence imaging, and 9-(4-(18)F-fluoro-3-hydroxymethylbutyl) guanine ((18)F-FHBG) small-animal PET imaging. The therapeutic efficacy on p21-3H-expressing tumor xenografts was assessed by daily administration with SAHA (100 mg/kg intraperitoneally) or GCV (20 mg/kg) for 9 d, followed by tumor volume measurement. RESULTS: On treatment with trichostatin A or SAHA, H1299 cells carrying p21-3H showed a significant increase of luciferase activity, cellular uptake of (3)H-FEAU (Moravek), and DsRed expression. In vivo tumor xenografts carrying p21-3H also showed increased luciferase activity by luminescent imaging and enhanced accumulation of (18)F-FHBG by small-animal PET imaging. Furthermore, when cells transfected with p21-3H or p21/PstI-3H (which lacks p53-binding sites) were treated, the increase of luciferase activity was similar in both groups, indicating that HDACI-induced p21 promoter activation is independent of p53. Both in vitro and in vivo results showed improved therapeutic effect by combined treatment of GCV and HDACI. CONCLUSION: We have established an HDACI-inducible, p21-driven reporter system that has the potential for evaluating the anticancer effect of HDACIs on cancer cells by multiple molecular imaging modalities. Furthermore, ttksr39 in a p21-3H reporter construct provides a potential combination with thymidine kinase-mediated gene therapy to optimize the therapeutic benefit of HDACI.


Assuntos
Antineoplásicos/uso terapêutico , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidores de Histona Desacetilases/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Transcrição Gênica/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Corantes , Regulação para Baixo/efeitos dos fármacos , Feminino , Fusão Gênica , Humanos , Indicadores e Reagentes , Luciferases/genética , Luminescência , Camundongos , Camundongos Nus , Microscopia Confocal , Imagem Molecular , Plasmídeos/genética , Tomografia por Emissão de Pósitrons , Sais de Tetrazólio , Tiazóis , Transfecção
4.
Brain Res ; 1504: 16-24, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23419892

RESUMO

BACKGROUND AND OBJECTIVE: Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, caused by a CAG/polyglutamine repeat expansion, which is associated with a dysregulation of histone function and an impairment of protein transcription. Histone deacetylase (HDAC) inhibitors, such as vorinostat (SAHA), have shown promise as therapeutic agents. However, there have been few studies on the expression of HDACs and acetylated core histones (AcHs) in either normal animals or humans, or in HD patients or HD animal models. Therefore, we investigated the expression of HDACs and AcHs in HD brain by immunohistochemistry, and have compared findings with elderly control subjects and patients with frontotemporal lobar degeneration (FTLD) to determine whether any observed changes were specific for HD. RESULTS AND CONCLUSION: we show specific and significant losses of AcH2A, AcH2B, AcH3 and AcH4 expression from cells in the caudate nucleus and Purkinje cells of the cerebellum in HD compared to patients with FTLD and control subjects, while the level of HDAC 5 was increased in these cells.


Assuntos
Encéfalo/metabolismo , Histona Desacetilase 2/biossíntese , Histonas/biossíntese , Doença de Huntington/metabolismo , Acetilação , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Histona Desacetilase 2/análise , Histonas/análise , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
5.
Neuroimage ; 64: 630-9, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22995777

RESUMO

Epigenetic modifications mediated by histone deacetylases (HDACs) play important roles in the mechanisms of different neurologic diseases and HDAC inhibitors (HDACIs) have shown promise in therapy. However, pharmacodynamic profiles of many HDACIs in the brain remain largely unknown due to the lack of validated methods for noninvasive imaging of HDAC expression-activity. In this study, dynamic PET/CT imaging was performed in 4 rhesus macaques using [(18)F]FAHA, a novel HDAC substrate, and [(18)F]fluoroacetate, the major radio-metabolite of [(18)F]FAHA, and fused with corresponding MR images of the brain. Quantification of [(18)F]FAHA accumulation in the brain was performed using a customized dual-tracer pharmacokinetic model. Immunohistochemical analyses of brain tissue revealed the heterogeneity of expression of individual HDACs in different brain structures and cell types and confirmed that PET/CT/MRI with [(18)F]FAHA reflects the level of expression-activity of HDAC class IIa enzymes. Furthermore, PET/CT/MRI with [(18)F]FAHA enabled non-invasive, quantitative assessment of pharmacodynamics of HDAC inhibitor SAHA in the brain.


Assuntos
Encéfalo/enzimologia , Encéfalo/metabolismo , Radioisótopos de Flúor/farmacocinética , Histona Desacetilases/metabolismo , Imageamento por Ressonância Magnética/métodos , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Anilidas , Animais , Epigênese Genética/fisiologia , Feminino , Regulação Enzimológica da Expressão Gênica/fisiologia , Macaca mulatta , Masculino , Técnica de Subtração
6.
Mol Imaging Biol ; 15(2): 175-85, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22875335

RESUMO

PURPOSE: The understanding of the role of genetic alterations in Wilms tumor development could be greatly advanced using a genetically engineered mouse models that can replicate the development and progression of this disease in human patients and can be monitored using non-invasive structural and molecular imaging optimized for renal tumors. PROCEDURES: Repetitive dual-contrast computed tomography (CT; intravenous and intraperitoneal contrast), T2-weighted magnetic resonance imaging (MRI), and delayed 2-deoxy-2-[(18)F]fluoro-D-glucose ((18)F-FDG) positron emission tomography (PET) were utilized for characterization of Igf2 biallelic expression/Wt1 knockout mouse model of Wilms tumor. For CT imaging, Ioversol 678 mg/ml in 200 µl was administered i.p. followed by 100 µl injected intravenously at 20 and 15 min prior to imaging, respectively. Static PET imaging studies were acquired at 1, 2, and 3 h after i.v. administration of (18)F-FDG (400 µCi). Coronal and sagittal T1-weighted images (TE/TR 8.5/620 ms) were acquired before and immediately after i.v. injection of 0.4 ml/kg gadopentetate dimeglumine followed by T2-weighted images (TE/TR 60/300 ms). Tumor tissue samples were characterized by histopathology and immunohistochemistry for Glut1, FASN, Ki67, and CD34. In addition, six Wt1-Igf2 mice were treated with a mitogen-activated protein kinase (MEK) inhibitor U0126 (50 µmol/kg i.p.) every 4 days for 6 weeks. (18)F-FDG PET/CT imaging was repeated at different days after initiation of therapy with U0126. The percent change of initial tumor volume and SUV was compared to non-treated historic control animals. RESULTS: Overall, the best tumor-to-adjacent kidney contrast as well as soft tissue contrast for other abdominal organs was achieved using T2-weighted MRI. Delayed (18)F-FDG PET (3-h post (18)F-FDG administration) and dual-contrast CT (intravenous and intraperitoneal contrast) provided a more accurate anatomic and metabolic characterization of Wilms tumors in Wt1-Igf2 mice during early development and progression of renal tumors. Over the 8-month period, 46 Wt1-Igf2 mice and 8 littermate control mice were studied. Renal tumors were identified in 54.3 % of Wt1-Igf2 mice between post-natal 50-100 days. In 35.6 % of Wt1-Igf2 mice, tumors were localized in the right kidney; in 24 %, in the left kidney, while 40.4 % of Wt1-Igf2 mice had bilateral kidney tumors. Metastatic lesions were identified in 15.4 % of Wt1-Igf2 mice. Increased levels of Glut1 and IGF1R expression, high Ki67 labeling index, and a dense network of CD34+ microvessels in renal tumors was consistent with increased (18)F-FDG accumulation. Treatment with a MEK 1/2 inhibitor U0126 did not cause the inhibition of tumor growth as compared to untreated animals. However, after the first three to four doses (~2 weeks of treatment), a decrease in (18)F-FDG SUV was observed, as compared to pre-treatment levels (p < 0.05, paired Student t test), which constitutes a metabolic response. Six weeks later, despite continuing therapy, the (18)F-FDG SUV increased again to previous levels. CONCLUSIONS: The optimized dual contrast PET/CT imaging with early post i.v. and i.p. contrast CT and 3 h delayed PET imaging after (18)F-FDG administration provides a sensitive and reliable method for detecting early tumor lesions in this endogenous mouse model of Wilms tumor and for monitoring their growth in response to targeted therapies. Therapy with MEK inhibitor U0126 produces only a transient inhibition of tumor glycolytic activity but does not inhibit tumor growth, which is due to continuing IGF2-induced signaling from IGF1R through the PI3K-AKT-mTOR pathway.


Assuntos
Butadienos/farmacologia , Fluordesoxiglucose F18 , Imageamento por Ressonância Magnética/métodos , Imagem Multimodal/métodos , Nitrilas/farmacologia , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Tumor de Wilms/diagnóstico , Tumor de Wilms/tratamento farmacológico , Animais , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Tumor de Wilms/diagnóstico por imagem , Tumor de Wilms/patologia
7.
Nucl Med Biol ; 39(4): 593-600, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22226022

RESUMO

INTRODUCTION: The cannabinoid receptor type 2 (CB(2)) is an important target for development of drugs and imaging agents for diseases, such as neuroinflammation, neurodegeneration and cancer. Recently, we reported synthesis and results of in vitro receptor binding of a focused library of fluorinated 2-oxoquinoline derivatives as CB(2) receptor ligands. Some of the compounds demonstrated to be good CB(2)-specific ligands with Ki values in the nanomolar to subnanomolar concentrations; therefore, we pursued the development of their (18)F-labeled analogues that should be useful for positron emission tomography (PET) imaging of CB(2) receptor expression. Here, we report the radiosynthesis of two (18)F-labeled 2-oxoquinoline derivatives and the preliminary in vitro and ex vivo evaluation of one compound as a CB(2)-specific radioligand. METHODS: 4-[(18)F]fluorobenzyl amine [(18)F]-3 was prepared by radiofluorination of 4-cyano-N,N,N-trimethylanilinium triflate salt followed by reduction with LiAlH(4) and then coupled with acid chlorides 11 and 12 to afford [(18)F]-13 and [(18)F]-14. In vitro CB(2) receptor binding assay was performed using U87 cells transduced with CB(2) and CB(1) receptor. Ex vivo autoradiography was performed with [(18)F]-14 on spleen and on CB(2)- and CB(1)-expressing and wild-type U87 subcutaneous tumors grown in mice. RESULTS: The radiochemical yields of [(18)F]-13 and [(18)F]-14 were 10%-15.0% with an average of 12% (n=10); radiochemical purity was >99% with specific activity 1200 mCi/µmol. The dissociation constant Kd for [(18)F]-14 was 3.4 nM. Ex vivo autoradiography showed accumulation of [(18)F]-14 in the CB(2)-expressing tumor. CONCLUSION: Two new [(18)F]-labeled CB(2) ligands have been synthesized. Compound [(18)F]-14 appears to be a potential PET imaging agent for the assessment of CB(2) receptor expression; however, poor solubility restrain its use in vivo.


Assuntos
Técnicas de Química Sintética , Radioisótopos de Flúor , Tomografia por Emissão de Pósitrons/métodos , Quinolonas/síntese química , Receptor CB2 de Canabinoide/metabolismo , Animais , Autorradiografia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Quinolonas/química , Quinolonas/metabolismo , Radioquímica
8.
Nucl Med Biol ; 38(8): 1129-34, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21741850

RESUMO

INTRODUCTION: It is important to identify all circulating metabolites, including free fluoride, for accurate pharmacokinetic modeling of [(18)F]-labeled radiotracers. We sought to determine the most efficient method to detect and quantify low levels of free [(18)F]fluoride in biological samples. METHODS: Low levels of [(18)F]fluoride were analyzed using two methods: (A) an ion-exchange cartridge and gamma counting, and (B) radio-HPLC, to compare the detection limits of these two analytical methods. Twenty microliters of [(18)F]fluoride solution was loaded onto an ion-exchange cartridge, then eluted with 20% MeCN/water (5 ml) and radioactivity trapped in the cartridge counted on a gamma counter. [(18)F]Fluoride was also determined in plasma and urine from mice injected with [(18)F]-labeled thymidine analogues using Method A. RESULTS: The detection sensitivity of Method A was 9.4-fold higher than that of Method B (0.075±0.004 vs. 0.71±0.02 nCi). With Method A, [(18)F]fluoride was determined in plasma for [(18)F]FLT, [(18)F]FMAU, [(18)F]FEAU and N(3)-[(18)F]FPrT as 1.4±0.31% (n=4), 0.17±0.49% (n=3), 4.88±1.62% (n=3) and 12.94±0.48% (n=4), respectively. The amount of [(18)F]fluoride determined in the urine was 11.49±1.60% (n=4) from [(18)F]FLT, 5.36±2.34% (n=3) from [(18)F]FMAU, 13.57±1.96% (n=3) from [(18)F]FEAU and 11.19±1.98% (n=4) from N(3)-[(18)F]FPrT. CONCLUSION: Low levels of [(18)F]fluoride in biological samples can be detected and quantified using an ion-exchange cartridge and gamma counting. This methodology is simple, accurate and superior to the standard use of radio-HPLC on a C(18) column for metabolite analysis, and it should be useful in pharmacokinetic modeling for animal imaging studies using an [(18)F]-labeled radiotracer and PET.


Assuntos
Análise Química do Sangue/métodos , Cromatografia Líquida de Alta Pressão/métodos , Fluoretos/análise , Radioisótopos de Flúor/análise , Urina/química , Animais , Troca Iônica , Camundongos , Sensibilidade e Especificidade
9.
Mol Cancer Res ; 9(3): 259-70, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21364021

RESUMO

PURPOSE: Hypoxia-inducible factor 1 (HIF-1) promotes cancer cell survival and tumor progression. The specific role played by HIF-1 and tumor-stromal interactions toward determining tumor resistance to radiation treatment remains undefined. We applied a multimodality preclinical imaging platform to mechanistically characterize tumor response to radiation, with a focus on HIF-1-dependent resistance pathways. METHODS: C6 glioma and HN5 human squamous carcinoma cells were stably transfected with a dual HIF-1 signaling reporter construct (dxHRE-tk/eGFP-cmvRed2XPRT). Reporter cells were serially interrogated in vitro before and after irradiation as monolayer and multicellular spheroid cultures and as subcutaneous xenografts in nu/nu mice. RESULTS: In vitro, single-dose irradiation of C6 and HN5 reporter cells modestly impacted HIF-1 signaling in normoxic monolayers and inhibited HIF-1 signaling in maturing spheroids. In contrast, irradiation of C6 or HN5 reporter xenografts with 8 Gy in vivo elicited marked upregulation of HIF-1 signaling and downstream proangiogenic signaling at 48 hours which preceded recovery of tumor growth. In situ ultrasound imaging and dynamic contrast-enhanced (DCE) MRI indicated that HIF-1 signaling followed acute disruption of stromal vascular function. High-resolution positron emission tomography and dual-contrast DCE-MRI of immobilized dorsal skin window tumors confirmed postradiotherapy HIF-1 signaling to spatiotemporally coincide with impaired stromal vascular function. Targeted disruption of HIF-1 signaling established this pathway to be a determinant of tumor radioresistance. CONCLUSIONS: Our results illustrate that tumor radioresistance is mediated by a capacity to compensate for stromal vascular disruption through HIF-1-dependent proangiogenic signaling and that clinically relevant vascular imaging techniques can spatially define mechanisms associated with tumor irradiation.


Assuntos
Fator 1 Induzível por Hipóxia/metabolismo , Fator 1 Induzível por Hipóxia/efeitos da radiação , Isquemia/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/radioterapia , Tolerância a Radiação/fisiologia , Fatores de Crescimento do Endotélio Vascular/efeitos da radiação , Adaptação Fisiológica , Animais , Hipóxia Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Humanos , Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Nus , Neoplasias/patologia , Tomografia por Emissão de Pósitrons/mortalidade , Ratos , Esferoides Celulares/efeitos da radiação , Transplante Heterólogo , Carga Tumoral/efeitos da radiação , Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(4): 1603-8, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21220318

RESUMO

The importance of the EGF receptor (EGFR) signaling pathway in the development and progression of nonsmall cell lung carcinomas (NSCLC) is widely recognized. Gene sequencing studies revealed that a majority of tumors responding to EGFR kinase inhibitors harbor activating mutations in the EGFR kinase domain. This underscores the need for novel biomarkers and diagnostic imaging approaches to identify patients who may benefit from particular therapeutic agents and approaches with improved efficacy and safety profiles. To this goal, we developed 4-[(3-iodophenyl)amino]-7-{2-[2-{2-(2-[2-{2-([(18)F]fluoroethoxy)-ethoxy}-ethoxy]-ethoxy)-ethoxy}-ethoxy]-quinazoline-6-yl-acrylamide ([(18)F]F-PEG6-IPQA), a radiotracer with increased selectivity and irreversible binding to the active mutant L858R EGFR kinase. We show that PET with [(18)F]F-PEG6-IPQA in tumor-bearing mice discriminates H3255 NSCLC xenografts expressing L858R mutant EGFR from H441 and PC14 xenografts expressing EGFR or H1975 xenografts with L858R/T790M dual mutation in EGFR kinase domain, which confers resistance to EGFR inhibitors (i.e., gefitinib). The T790M mutation precludes the [(18)F]F-PEG6-IPQA from irreversible binding to EGFR. These results suggest that PET with [(18)F]F-PEG6-IPQA could be used for the selection of NSCLC patients for individualized therapy with small molecular inhibitors of EGFR kinase that are currently used in the clinic and have a similar structure (i.e., iressa, gefitinib, and erlotinib).


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Imagem Molecular/métodos , Proteínas Mutantes/metabolismo , Substituição de Aminoácidos , Animais , Ligação Competitiva , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/genética , Fluordesoxiglucose F18/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Proteínas Mutantes/genética , Mutação , Tomografia por Emissão de Pósitrons/métodos , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Tomografia Computadorizada por Raios X/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Mol Imaging Biol ; 13(3): 536-546, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20593279

RESUMO

INTRODUCTION: Previous studies demonstrated that the lactose-binding protein (hepatocellular carcinoma-intestine-pancreas and pancreatitis-associated proteins (HIP/PAP)) is upregulated >130 times in peritumoral pancreatic tissue as compared to normal pancreatic tissue. Therefore, we developed a new radiolabeled ligand of HIP/PAP, the ethyl-ß-D-galactopyranosyl-(1,4')-2'-deoxy-2'-[¹8F]fluoro-ß-D-glucopyranoside (Et-[¹8F]FDL) for noninvasive imaging of pancreatic carcinoma using positron emission tomography and computerized tomography (PET/CT). METHODS: The novel precursor and radiolabeling methods for synthesis of Et-[¹8F]FDL produced no isomers; the average decay-corrected radiochemical yield was 68%, radiochemical purity >99%, and specific activity >74 GBq/µmol. The radioligand properties of Et-[¹8F]FDL were evaluated using an ex vivo autoradiography and immunohistochemistry in pancreatic tissue sections obtained from mice-bearing orthotopic pancreatic tumor xenografts. RESULTS AND DISCUSSION: Et-[¹8F]FDL binding to peritumoral pancreatic tissue sections strongly correlated with HIP/PAP expression (r = 0.81) and could be completely blocked by treatment with 1 mM lactose. CONCLUSION: These results suggest that Et-[¹8F]FDL is a promising agent which should be evaluated for detection of early pancreatic carcinomas by PET/CT imaging.


Assuntos
Autorradiografia/métodos , Dissacarídeos/síntese química , Detecção Precoce de Câncer , Glucosídeos/síntese química , Lactose/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Animais , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Cromatografia Líquida de Alta Pressão , Dissacarídeos/química , Dissacarídeos/isolamento & purificação , Glucosídeos/química , Glucosídeos/isolamento & purificação , Halogenação , Imuno-Histoquímica , Lectinas Tipo C/metabolismo , Ligantes , Camundongos , Proteínas Associadas a Pancreatite , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química
12.
Nucl Med Biol ; 35(6): 697-705, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18678355

RESUMO

INTRODUCTION: [(18)F]-Labeled analogues of thymidine have demonstrated efficacy for PET imaging of cellular proliferation. We have synthesized two [(18)F]-labeled N(3)-substituted thymidine analogues, N(3)-[(18)F]fluoroethyl thymidine (N(3)-[(18)F]-FET) and N(3)-[(18)F]fluoropropyl thymidine (N(3)-[(18)F]-FPrT), and performed preliminary PET imaging studies in tumor-bearing mice. METHODS: Thymidine was converted to its 3',5'-O-bis-tetrahydropyranyl ether, which was then converted to the N(3)-ethyl and propyl-substituted mesylate precursors. Reactions of these mesylate precursors with n-Bu(4)N[(18)F] or K[(18)F]/kryptofix followed by acid hydrolysis and HPLC purification yielded N(3)-[(18)F]-FET and N(3)-[(18)F]-FPrT, respectively. Subcutaneous (sc) xenografts of H441 human non-small cell lung cancer were established in two groups of mice (each n=6). Micro-PET images of the tumor-bearing animals were acquired after intravenous injection of N(3)-[(18)F]-FET or N(3)-[(18)F]-FPrT (3700 KBq/animal). RESULTS: The radiochemical yields were 2-12% (d.c.) for N(3)-[(18)F]-FET and 30-38% (d.c.) for N(3)-[(18)F]-FPrT. Radiochemical purity was >99% and calculated specific activity was >74 GBq/mumol at the end of synthesis. The accumulation of N(3)-[(18)F]-FET and N(3)-[(18)F]-FPrT in the tumor tissue at 2 h postinjection was 1.81+/-0.78 and 2.95+/-1.14 percent injected dose per gram (%ID/g), respectively; tumor/muscle ratios were 5.57+/-0.82 and 7.69+/-2.18, respectively; the unidirectional influx rates (K(i)) were 0.013 and 0.018 ml/g per minute, respectively. CONCLUSION: Two novel [(18)F]- N(3)-substituted thymidine analogues have been synthesized in good yields, high purity and high specific activity. Preliminary in vivo studies demonstrated the efficacy of these [(18)F]- N(3)-substituted thymidine analogues for PET imaging of tumors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Timidina/análogos & derivados , Tirosina/análogos & derivados , Animais , Linhagem Celular Tumoral , Humanos , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Especificidade de Órgãos , Projetos Piloto , Compostos Radiofarmacêuticos/farmacocinética , Timidina/farmacocinética , Distribuição Tecidual , Tirosina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...