Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Bioorg Med Chem ; 22(1): 623-32, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24280068

RESUMO

We synthesized two series of imatinib mesylate (STI-571) analogs to develop a Bcr-Abl and c-KIT receptor-specific labeling agent for positron emission tomography (PET) imaging to measure Bcr-Abl and c-KIT expression levels in a mouse model. The methods of molecular modeling, synthesis of STI-571 and its analogs, in vitro kinase assays, and radiolabeling are described. Molecular modeling revealed that these analogs bind the same Bcr-Abl and c-KIT binding sites as those bound by STI-571. The analogs potently inhibit the tyrosine kinase activity of Bcr-Abl and c-KIT, similarly to STI-571. [(18)F]-labeled STI-571 was prepared with high specific activity (75 GBq/µmol) by nucleophilic displacement and an average radiochemical yield of 12%. [(131)I]-labeled STI-571 was prepared with high purity (>95%) and an average radiochemical yield of 23%. The uptake rates of [(18)F]-STI-571 in K562 cells expressing Abl and in U87WT cells overexpressing c-KIT were significantly higher than those in the U87 cell and could be inhibited by STI-71 (confirming the specificity of uptake). PET scans of K562 and U87WT tumor-bearing mice with [(18)F]-STI-571 as a contrast agent showed visible tumor uptake and tumor-to-non-target contrast.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Proteínas de Fusão bcr-abl/metabolismo , Piperazinas/uso terapêutico , Tomografia por Emissão de Pósitrons/métodos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirimidinas/uso terapêutico , Animais , Antineoplásicos/química , Benzamidas/química , Modelos Animais de Doenças , Humanos , Mesilato de Imatinib , Camundongos , Modelos Moleculares , Piperazinas/química , Pirimidinas/química
2.
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
3.
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
4.
J Nucl Med ; 49(4): 637-48, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344433

RESUMO

UNLABELLED: Herpes virus type 1 thymidine kinase (HSV1-tk) and the mutant HSV1-sr39tk are the 2 most widely used "reporter genes" for radiotracer-based imaging. Two pyrimidine nucleoside analogs, [18F]FEAU (1-(2'-deoxy-2'-fluoro-beta-d-arabinofuranosyl)-5-ethyluridine) and [18F]FFEAU (1-(2'-deoxy-2'-fluoro-beta-d-arabinofuranosyl)-5-(2-fluoroethyl)uridine), have generated recent interest as potential new probes for imaging HSV1-tk and HSV1-sr39tk gene expression. METHODS: We compared [18F]FEAU and [18F]FFEAU with a series of other pyrimidine nucleoside derivatives (including 1-(2'-deoxy-2'-fluoro-beta-d-arabinofuranosyl)-5-iodouridine [FIAU]) and with acycloguanosine analogs using a stable HSV1-tk transduced cell line (RG2TK+) and wild-type RG2 cells. RESULTS: The in vitro accumulation data and the calculated and normalized clearance constant, nKi, as well as sensitivity and selectivity indices indicated that 2 pyrimidine nucleoside probes, [18F]FEAU and [18F]FFEAU, had the best uptake characteristics. These probes were selected for further dynamic PET studies in nude rats bearing subcutaneous RG2TK+ and RG2 tumors. The 2-h postinjection [18F]FEAU uptake levels were 3.3% +/- 1.0% and 0.28% +/- 0.07% dose/cm3 in subcutaneous RG2TK+ and RG2 tumors, respectively, and 2.3% +/- 0.2% and 0.19% +/- 0.01% dose/cm3, respectively, for [18F]FFEAU. The corresponding RG2TK+/RG2 uptake ratios were 11.5 +/- 1.5 and 12.2 +/- 1.4, respectively. The inherent problem of comparing different radiolabeled pyrimidine nucleoside and guanosine-based probes for imaging HSV1-tk expression using different transduced cell lines and assay systems in the absence of an independent thymidine kinase-enzyme assay is discussed. CONCLUSION: For HSV1-tk reporter systems that require a 1- to 4-h PET paradigm, HSV1-tk-[18F]FEAU is the current top contender.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Radioisótopos de Flúor , Genes Reporter , Herpesvirus Humano 1/enzimologia , Compostos Radiofarmacêuticos , Timidina Quinase/genética , Animais , Linhagem Celular Tumoral , Ratos
5.
J Nucl Med ; 48(5): 819-26, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17468435

RESUMO

UNLABELLED: A human-derived intrinsically nonimmunogenic reporter gene was tested for PET imaging of different molecular-genetic processes for potential clinical use. METHODS: The human mitochondrial thymidine kinase type 2 (hTK2) reporter gene truncated at the N terminus (DeltahTK2), alone or fused with green fluorescent protein (GFP), was used for preclinical evaluation in a mouse model. The levels of enzymatic activity of DeltahTK2 and DeltahTK2 GFP proteins were assessed using radiotracer accumulation and prodrug activation assays in vitro and in subcutaneous tumors grown from the corresponding cell lines in nude mice. Kinetic analyses of (124)I-2'-fluoro-2'-deoxy-1-beta-D-beta-arabinofuranosyl-5-iodouracil (FIAU), (18)F-2'-fluoro-2'-deoxy-1-beta-D-beta-arabinofuranosyl-5-ethyluracil (FEAU), or (18)F-9-(4-(18)F-fluoro-3-hydroxymethylbutyl)guanine (FHBG) uptake in tumors and biodistribution studies were performed. RESULTS: DeltahTK2 was successfully expressed in the cytoplasm of transduced cells. A new anti-hTK2 monoclonal antibody 8G2 was developed. The levels of FIAU and FEAU accumulation in cells expressing DeltahTK2 and DeltahTK2 GFP were at least 10-fold higher than in wild-type cells in vitro and about 6 times higher in vivo. We determined that FEAU is a more specific reporter substrate for DeltahTK2 than FIAU, whereas FHBG is not phosphorylated by this enzyme. In addition, we showed that DeltahTK2 transduced cells can be eliminated by treatment with d-arabinofuranosyl-cytosine. CONCLUSION: We have tested a human-derived reporter gene that is likely to be nonimmunogenic and potentially allows for long-term monitoring of different molecular-genetic processes by nuclear imaging techniques in humans. Using (124)I-FIAU, (18)F-FIAU, or (18)F-FEAU, it should be possible to image DeltahTK2 reporter gene expression with PET in preclinical and clinical studies.


Assuntos
Glioma/diagnóstico por imagem , Glioma/metabolismo , Mitocôndrias/enzimologia , Timidina Quinase/metabolismo , Linhagem Celular Tumoral , Genes Reporter/genética , Glioma/genética , Humanos , Cintilografia , Timidina Quinase/genética
6.
Nat Biotechnol ; 21(4): 405-13, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12652311

RESUMO

New technologies are needed to characterize the migration, survival, and function of antigen-specific T cells in vivo. Here, we demonstrate that Epstein-Barr virus (EBV)--specific T cells transduced with vectors encoding herpes simplex virus-1 thymidine kinase (HSV-TK) selectively accumulate radiolabeled 2'-fluoro-2'-deoxy-1-beta-D-arabinofuranosyl-5-iodouracil (FIAU). After adoptive transfer, HSV-TK+ T cells labeled in vitro or in vivo with [131I]FIAU or [124I]FIAU can be noninvasively tracked in SCID mice bearing human tumor xenografts by serial images obtained by scintigraphy or positron emission tomography (PET), respectively. These T cells selectively accumulate in EBV+ tumors expressing the T cells' restricting HLA allele but not in EBV- or HLA-mismatched tumors. The concentrations of transduced T cells detected in tumors and tissues are closely correlated with the concentrations of label retained at each site. Radiolabeled transduced T cells retain their capacity to eliminate targeted tumors selectively. This technique for imaging the migration of ex vivo-transduced antigen-specific T cells in vivo is informative, nontoxic, and potentially applicable to humans.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/farmacocinética , Radioisótopos do Iodo/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Linfócitos T/diagnóstico por imagem , Linfócitos T/metabolismo , Animais , Apoptose/fisiologia , Biomarcadores Tumorais/metabolismo , Linhagem Celular , Movimento Celular , Sobrevivência Celular/fisiologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Marcação por Isótopo/métodos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Sensibilidade e Especificidade , Simplexvirus/enzimologia , Simplexvirus/genética , Linfócitos T/fisiologia , Timidina Quinase/genética , Timidina Quinase/metabolismo , Distribuição Tecidual , Tomografia Computadorizada de Emissão/métodos , Transdução Genética/métodos
7.
Cancer Res ; 64(17): 6101-8, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15342393

RESUMO

Tumor hypoxia is a spatially and temporally heterogeneous phenomenon, which results from several tumor and host tissue-specific processes. To study the dynamics and spatial heterogeneity of hypoxia-inducible factor-1 (HIF-1)-specific transcriptional activity in tumors, we used repetitive noninvasive positron emission tomography (PET) imaging of hypoxia-induced HIF-1 transcriptional activity in tumors in living mice. This approach uses a novel retroviral vector bearing a HIF-1-inducible "sensor" reporter gene (HSV1-tk/GFP fusion) and a constitutively expressed "beacon" reporter gene (DsRed2/XPRT). C6 glioma cells transduced with this multireporter system revealed dose-dependent patterns in temporal dynamics of HIF-1 transcriptional activity induced by either CoCl2 or decreased atmospheric oxygen concentration. Multicellular spheroids of C6 reporter cells developed a hypoxic core when >350 microm in diameter. 18F-2'-fluoro-2'deoxy-1beta-D-arabionofuranosyl-5-ethyl-uracil (FEAU) PET revealed spatial heterogeneity of HIF-1 transcriptional activity in reporter xenografts in mice as a function of size or ischemia-reperfusion injury. With increasing tumor diameter (>3 mm), a marked increase in HIF-1 transcriptional activity was observed in the core regions of tumors. Even a moderate ischemia-reperfusion injury in small C6 tumors caused a rapid induction of HIF-1 transcriptional activity, which persisted for a long time because of the inability of C6 tumors to rapidly compensate acute changes in tumor microcirculation.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Genes Reporter/genética , Glioma/genética , Proteínas Luminescentes/genética , Proteínas Recombinantes de Fusão/genética , Timidina Quinase/genética , Fatores de Transcrição/genética , Ativação Transcricional/fisiologia , Animais , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Radioisótopos de Flúor , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas Luminescentes/biossíntese , Camundongos , Oxigênio/metabolismo , Compostos Radiofarmacêuticos , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Retroviridae/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Timidina Quinase/biossíntese , Tomografia Computadorizada de Emissão , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Appl Radiat Isot ; 107: 317-322, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26595775

RESUMO

Interest in using (89)Zr is rapidly increasing for immuno-PET applications due to its unique characteristics and increased availability. The focus of this study was to develop an optimized semi-automated methodology for producing (89)Zr-oxalate/(89)Zr-chloride, and evaluate the potential application of (89)Zr-chloride for radiopharmaceutical compounding. The data presented herein will be useful for the production of (89)Zr-labeled radiopharmaceuticals and their compliance with regulatory issues for both preclinical and clinical use.


Assuntos
Marcação por Isótopo/métodos , Radioisótopos/isolamento & purificação , Compostos Radiofarmacêuticos/isolamento & purificação , Zircônio/isolamento & purificação , Química Farmacêutica/instrumentação , Química Farmacêutica/métodos , Cloretos/isolamento & purificação , Ciclotrons , Desferroxamina , Humanos , Imunoglobulina G/isolamento & purificação , Marcação por Isótopo/instrumentação , Oxalatos/isolamento & purificação , Tomografia por Emissão de Pósitrons , Extração em Fase Sólida
9.
Cancer Gene Ther ; 12(1): 101-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15499377

RESUMO

In order to noninvasively detect Salmonella delivery vectors within tumors, we used a genetically modified Salmonella, VNP20009, that expresses the herpes simplex thymidine kinase (HSV1-tk) reporter gene. VNP20009-TK were able to selectively localize within murine tumor models and to effectively sequester a radiolabeled nucleoside analogue, 2'-fluoro-1-beta-D-arabino-furanosyl-5-iodo-uracil (FIAU). A quantitative relationship between the level of radioactivity accumulated and the number of bacteria in tumor and different tissues was demonstrated. The in vivo accumulation of [14C]FIAU measured in tissue sample homogenates and sections were related to Salmonella number and to immunohistochemical bacterial staining, respectively. Quantitative autoradiography (QAR) revealed the relative intensity of [14C]FIAU accumulation in a tumor cross-section, demonstrating that the peripheral region of the tumor was significantly less active than internal regions. [124I]FIAU positron emission tomography (PET) and subsequent tissue radioactivity and bacterial concentration measurements were compared. A log-log relationship was found, and the PET images could identify multiple tumor sites. The ability to noninvasively detect Salmonella vectors by PET imaging has the potential to be conducted in a clinical setting, and could aid in development of these vectors by demonstrating the efficiency and duration of targeting as well as indicating the locations of tumors.


Assuntos
Genes Reporter , Herpesvirus Humano 1/enzimologia , Tomografia por Emissão de Pósitrons , Salmonella/genética , Timidina Quinase/genética , Animais , Autorradiografia , Vacinas Bacterianas , Carcinoma/genética , Carcinoma/microbiologia , Carcinoma/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/microbiologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Imuno-Histoquímica , Melanoma/genética , Melanoma/microbiologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/microbiologia , Neoplasias Cutâneas/patologia , Timidina Quinase/biossíntese , Vacinas Atenuadas
10.
Nucl Med Biol ; 32(8): 811-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16253805

RESUMO

The preparation and radiolabeling of 2'-fluoro-2'-deoxy-1-beta-d-arabinofuranosyl-5-(2-fluoroethyl)-uracil (FFEAU) with 18F and its evaluation as a probe for imaging herpes simplex virus 1 thymidine kinase (HSV1-tk) gene expression are described. 2'-Fluoro-2'-deoxy-3',5'-di-O-benzoyl-1-beta-d-arabinofuranosyl-3-N-benzoyl-5-(2-[18F]fluoroethyl)-uracil 12 was prepared by nucleophilic substitution of the corresponding tosyl 8 or trifluoroethanesulfonyl 9 derivative with n-Bu4N[18F]F. Base hydrolysis was used to remove the benzoyl protecting groups, followed by HPLC purification, to afford [18F]FFEAU 13. The trifluoroethanesulfonyl substrate 9 appears to be the better labeling precursor. Carrier n-Bu4NF was added to the labeling reaction, which resulted in specific activities of 40-70 Ci/mmol (estimated). Radiochemical purity averaged 94+/-4%. Although [18F]FFEAU was obtained in low radiochemical yield with 9 and further optimization of the radiosynthesis will be required, sufficient product was available for a series of in vitro and in vivo studies. [18F]FFEAU was directly compared with [3H]TdR in a series of in vitro accumulation studies involving a HSV1-tk stably transduced cell line, RG2TK+ and a nontransduced, wild-type RG2 cells. The initial in vitro and in vivo imaging studies are promising; FFEAU has in vitro accumulation and sensitivity characteristics similar to that previously reported for FIAU, but greater selectivity than FIAU due to lower uptake and retention in nontransduced cells and tissues. The animal imaging experiment showed low levels of radioactivity in the lungs, with little or no radioactivity seen in the heart, liver, spleen and intestines.


Assuntos
Perfilação da Expressão Gênica/métodos , Glioblastoma/diagnóstico por imagem , Glioblastoma/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transfecção/métodos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/farmacocinética , Linhagem Celular Tumoral , Genes Reporter , Glioblastoma/genética , Taxa de Depuração Metabólica , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Coloração e Rotulagem/métodos , Distribuição Tecidual
11.
Nucl Med Biol ; 32(4): 313-21, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15878500

RESUMO

The pyridopyrimidinones are a potent class of inhibitors of c-Abl kinase and Bcr-Abl kinase, the causative fusion protein in chronic myelogenous leukemia and Src family kinases. A novel method for routine, high-yield no-carrier-added synthesis of [(124)I]-, [(125)I]- and [(131)I]-6-(2,6-dichlorophenyl)-2-(4-iodophenylamino)-8-methyl-8H-pyrido[2,3-d]pyrimidin-7-one has been developed. The 4'-trimethylstannyl- or 4'-tri-n-butylstannyl-pyridopyrimidinone precursors were prepared from the aryl bromide via a palladium-mediated coupling with hexaalkylditin (dioxane/microwave irradiation/10 min at 160 degrees C). The radioiodination of 4'-stannylpyridopyrimidinones was found to optimally occur via an iododestannylation with Na(124)I, Na(125)I or Na(131)I in the presence of an oxidant [30% H(2)O(2)/HOAc (1:3)/10 min] in 79-87% radiochemical yield with >99% radiochemical purity. The total radiosynthesis time was 30 min. The 4-iodophenylpyridopyrimidinone 2 inhibited recombinant Abl kinase activity with an IC(50) of 2.0 nM. Cell proliferation of K562 and A431 cells was inhibited with an IC(50) of 2.0 and 20 nM, respectively. Rapid cellular uptake and equilibrium were observed within 10-15 min using [(131)I]-4-iodophenylpyridopyrimidinone 6c in K562 and A431 cells and demonstrated a 2.8-fold uptake selectivity for the Bcr-Abl-expressing K562 cells at 60 min. These results suggest that pyridopyrimidinone radiotracers may be useful in imaging Abl-, Bcr-Abl- or Src-expressing malignancies.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridonas/farmacocinética , Pirimidinas/farmacocinética , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteínas de Fusão bcr-abl , Humanos , Radioisótopos do Iodo/efeitos adversos , Radioisótopos do Iodo/química , Radioisótopos do Iodo/farmacocinética , Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico por imagem , Taxa de Depuração Metabólica , Piridonas/efeitos adversos , Piridonas/química , Pirimidinas/efeitos adversos , Pirimidinas/química , Cintilografia , Compostos Radiofarmacêuticos/efeitos adversos , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética
12.
Neoplasia ; 5(3): 245-54, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12869307

RESUMO

To optimize the sensitivity of imaging HSV1-tk/GFP reporter gene expression, a series of HSV1-tk/GFP mutants was developed with altered nuclear localization and better cellular enzymatic activity, compared to that of the native HSV1-tk/GFP fusion protein (HSV1-tk/GFP). Several modifications of HSV1-tk/GFP reporter gene were performed, including targeted inactivating mutations in the nuclear localization signal (NLS), the addition of a nuclear export signal (NES), a combination of both mutation types, and a truncation of the first 135 bp of the native hsv1-tk coding sequence containing a "cryptic" testicular promoter and the NLS. A recombinant HSV1-tk/GFP protein and a highly sensitive sandwich enzyme-linked immunosorbent assay for HSV1-tk/GFP were developed to quantitate the amount of reporter gene product in different assays to allow normalization of the data. These different mutations resulted in various degrees of nuclear clearance, predominant cytoplasmic distribution, and increased total cellular enzymatic activity of the HSV1-tk/GFP mutants, compared to native HSV1-tk/GFP when expressed at the same levels. This appears to be the result of improved metabolic bioavailability of cytoplasmically retargeted mutant HSV1-tk/GFP enzymes for reaction with the radiolabeled probe (e.g., FIAU). The analysis of enzymatic properties of different HSV1-tk/GFP mutants using FIAU as a substrate revealed no significant differences from that of the native HSV1-tk/GFP. Improved total cellular enzymatic activity of cytoplasmically retargeted HSV1-tk/GFP mutants observed in vitro was confirmed by noninvasive imaging of transduced subcutaneous tumor xenografts bearing these reporters using [(131)I]FIAU and a gamma-camera.


Assuntos
Diagnóstico por Imagem/métodos , Genes Reporter/genética , Simplexvirus/genética , Timidina Quinase/metabolismo , Animais , Arabinofuranosiluracila/análogos & derivados , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Mutação , Reação em Cadeia da Polimerase , Transporte Proteico , RNA Mensageiro/análise , Proteínas Recombinantes de Fusão/genética , Simplexvirus/enzimologia , Timidina Quinase/genética
13.
J Nucl Med ; 43(8): 1072-83, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163634

RESUMO

UNLABELLED: The efficacy of 3 radiolabeled probes of current interest for imaging herpes simplex virus type 1 thymidine kinase (HSV1-tk) expression in vivo with PET, including (124)I- or (131)I-labeled 2'-fluoro-2'-deoxy-1-beta-D-arabinofuranosyl-5-iodouracil (FIAU), (18)F-labeled 9-[4-fluoro-3-(hydroxymethyl)butyl]guanine (FHBG), and (18)F-labeled 9-[3-fluoro-1-hydroxy-2-propoxymethyl]guanine (FHPG), was compared. METHODS: Two established rat glioma cell lines, stably transduced RG2TK+ and wild-type RG2, were used for paired comparisons of probe accumulation in vitro and for paired comparisons of subcutaneous xenografts produced from these cell lines in athymic rnu/rnu rats. RESULTS: The in vitro paired probe uptake (0-3 h) comparisons in RG2TK+ cells showed that FIAU accumulation was 15-fold greater than that of FHBG and 41-fold greater than that of FHPG. The net accumulation rate values (+/-SD) calculated for RG2TK+ cells were 0.317 +/- 0.066, 0.022 +/- 0.001, and 0.0077 +/- 0.0003 mL/min/g cells for FIAU, FHBG, and FHPG, respectively. These results and similar uptake studies in RG2 wild-type cells suggest a possible cell membrane transport limitation for FHBG and FHPG. The paired 2-h in vivo uptake studies produced similar differences in RG2TK+ xenografts for FIAU and FHBG (1.22 +/- 0.21 vs. 0.074 +/- 0.49 %dose/g) and for FIAU and FHPG (1.27 +/- 0.14 vs. 0.023 +/- 0.008 %dose/g). These differences were clearly visible on the images. FIAU accumulation at 24 h was 1.53 +/- 0.40 %dose/g. Plasma clearance was FHBG > FHPG >> FIAU. The FIAU images showed significant stomach and some intestinal background radioactivities, whereas hepatobiliary and intestinal background activities were very high for the guanosine analogs (FHBG > FHPG). Dynamic imaging showed early ( approximately 10 min) selective localization of FIAU in RG2TK+ xenografts, whereas FHBG and FHPG are being cleared from the HSV1-tk transduced and wild-type xenografts over the initial 2-h imaging period. CONCLUSION: The in vitro and in vivo results (including the PET images) show that FIAU is a substantially more efficient probe than FHBG or FHPG for imaging HSV1-tk expression, with greater sensitivity and contrast as well as lower levels of abdominal background radioactivity at 2 and 24 h.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Herpesvirus Humano 1/enzimologia , Nucleosídeos , Timidina Quinase/genética , Tomografia Computadorizada de Emissão , Animais , Radioisótopos de Carbono , Fluordesoxiglucose F18 , Expressão Gênica , Radioisótopos do Iodo , Compostos Radiofarmacêuticos , Ratos , Ratos Nus , Timidina Quinase/biossíntese
14.
Mol Imaging Biol ; 14(2): 213-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21437735

RESUMO

INTRODUCTION: To facilitate the clinical translation of (18)F-fluoroacetate ((18)F-FACE), the pharmacokinetics, biodistribution, radiolabeled metabolites, radiation dosimetry, and pharmacological safety of diagnostic doses of (18)F-FACE were determined in non-human primates. METHODS: (18)F-FACE was synthesized using a custom-built automated synthesis module. Six rhesus monkeys (three of each sex) were injected intravenously with (18)F-FACE (165.4 ± 28.5 MBq), followed by dynamic positron emission tomography (PET) imaging of the thoracoabdominal area during 0-30 min post-injection and static whole-body PET imaging at 40, 100, and 170 min. Serial blood samples and a urine sample were obtained from each animal to determine the time course of (18)F-FACE and its radiolabeled metabolites. Electrocardiograms and hematology analyses were obtained to evaluate the acute and delayed toxicity of diagnostic dosages of (18)F-FACE. The time-integrated activity coefficients for individual source organs and the whole body after administration of (18)F-FACE were obtained using quantitative analyses of dynamic and static PET images and were extrapolated to humans. RESULTS: The blood clearance of (18)F-FACE exhibited bi-exponential kinetics with half-times of 4 and 250 min for the fast and slow phases, respectively. A rapid accumulation of (18)F-FACE-derived radioactivity was observed in the liver and kidneys, followed by clearance of the radioactivity into the intestine and the urinary bladder. Radio-HPLC analyses of blood and urine samples demonstrated that (18)F-fluoride was the only detectable radiolabeled metabolite at the level of less than 9% of total radioactivity in blood at 180 min after the (18)F-FACE injection. The uptake of free (18)F-fluoride in the bones was insignificant during the course of the imaging studies. No significant changes in ECG, CBC, liver enzymes, or renal function were observed. The estimated effective dose for an adult human is 3.90-7.81 mSv from the administration of 185-370 MBq of (18)F-FACE. CONCLUSIONS: The effective dose and individual organ radiation absorbed doses from administration of a diagnostic dosage of (18)F-FACE are acceptable. From a pharmacologic perspective, diagnostic dosages of (18)F-FACE are non-toxic in primates and, therefore, could be safely administered to human patients for PET imaging.


Assuntos
Radioisótopos de Flúor/metabolismo , Radioisótopos de Flúor/farmacocinética , Fluoracetatos/metabolismo , Fluoracetatos/farmacocinética , Macaca mulatta/metabolismo , Radiometria , Animais , Cromatografia Líquida de Alta Pressão , Eletrocardiografia , Radioisótopos de Flúor/sangue , Radioisótopos de Flúor/toxicidade , Fluoracetatos/química , Fluoracetatos/toxicidade , Humanos , Injeções Intravenosas , Imagem Multimodal , Especificidade de Órgãos/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Fatores de Tempo , Distribuição Tecidual/efeitos dos fármacos , Tomografia Computadorizada por Raios X , Testes de Toxicidade Aguda
15.
Mol Imaging Biol ; 13(5): 853-61, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20859697

RESUMO

INTRODUCTION: Epidermal growth factor receptor (EGFR)-targeted therapies with antibodies and small molecular EGFR kinase inhibitors have shown poor efficacy in unselected populations of patients with advanced non-small cell lung carcinomas (NSCLC). In contrast, patients with overexpression of EGFR and activating mutations in EGFR kinase domain demonstrated improved responses to EGFR kinase inhibitors. Therefore, we have developed a novel radiotracer, [(18)F]F-PEG(6)-IPQA for PET imaging of EGFR expression-activity in NSCLC, and have described its radiosynthesis and in vitro evaluation in two NSCLC cell lines with wild-type and L858R active mutant EGFR. METHODS: A mesylate precursor was synthesized in multiple steps and radiofluorinated using K(18)F/Kryptofix. The fluorinated intermediate compound was reduced to an amino derivative then treated with acryloyl isobutyl carbonate, followed by purification by HPLC to obtain the desired product. RESULTS: Decay-corrected radiochemical yields of [(18)F]F-PEG(6)-IPQA were 3.9-17.6%, with an average of 9.0% (n = 11). Radiochemical purity was >97% with specific activity of 34 GBq/µmol (mean value, n = 10) at the end of synthesis. The accumulation of [(18)F]F-PEG(6)-IPQA in H3255 cells was ten-fold higher than in H441 cells, despite a two-fold lower level of activated phospho-EGFR expression in H3255 cells compared with H441 cells. The accumulation of [(18)F]F-PEG(6)-IPQA in both cell lines was significantly decreased in the presence of a small molecular EGFR kinase inhibitor, Iressa, at 100 µM concentration in culture medium. CONCLUSION: We have synthesized [(18)F]F-PEG(6)-IPQA and demonstrated its highly selective accumulation in active mutant L858R EGFR-expressing NSCLC cells in vitro. Further in vivo studies are warranted to assess the ability of PET imaging with [(18)F]F-PEG(6)-IPQA to discriminate the active mutant L858R EGFR-expressing NSCLC that are sensitive to therapy with EGFR kinase inhibitors vs NSCLC that express wild-type EGFR.


Assuntos
Receptores ErbB/metabolismo , Radioisótopos de Flúor/farmacocinética , Neoplasias Pulmonares/metabolismo , Tomografia por Emissão de Pósitrons , Quinazolinas/farmacocinética , Carcinoma de Pequenas Células do Pulmão/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Humanos , Técnicas In Vitro , Neoplasias Pulmonares/diagnóstico por imagem , Carcinoma de Pequenas Células do Pulmão/diagnóstico por imagem
16.
J Nucl Med ; 52(6): 934-41, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21571803

RESUMO

UNLABELLED: We recently developed the radiotracer 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-PEG(6)-IPQA) for noninvasive detection of active mutant epidermal growth factor receptor kinase-expressing non-small cell lung cancer xenografts in rodents. In this study, we determined the pharmacokinetics, biodistribution, metabolism, and radiation dosimetry of (18)F-PEG(6)-IPQA in nonhuman primates. METHODS: Six rhesus macaques were injected intravenously with 141 ± 59.2 MBq of (18)F-PEG(6)-IPQA, and dynamic PET/CT images covering the thoracoabdominal area were acquired for 30 min, followed by whole-body static images at 60, 90, 120, and 180 min. Blood samples were obtained from each animal at several time points after radiotracer administration. Radiolabeled metabolites in blood and urine were analyzed using high-performance liquid chromatography. The (18)F-PEG(6)-IPQA pharmacokinetic and radiation dosimetry estimates were determined using volume-of-interest analysis of PET/CT image datasets and blood and urine time-activity data. RESULTS: (18)F-PEG(6)-IPQA exhibited rapid redistribution and was excreted via the hepatobiliary and urinary systems. (18)F-PEG(6) was the major radioactive metabolite. The critical organ was the gallbladder, with an average radiation-absorbed dose of 0.394 mSv/MBq. The other key organs with high radiation doses were the kidneys (0.0830 mSv/MBq), upper large intestine wall (0.0267 mSv/MBq), small intestine (0.0816 mSv/MBq), and liver (0.0429 mSv/MBq). Lung tissue exhibited low uptake of (18)F-PEG(6)-IPQA due to the low affinity of this radiotracer to wild-type epidermal growth factor receptor kinase. The effective dose was 0.0165 mSv/MBq. No evidence of acute cardiotoxicity or of acute or delayed systemic toxicity was observed. On the basis of our estimates, diagnostic dosages of (18)F-PEG(6)-IPQA up to 128 MBq (3.47 mCi) per injection should be safe for administration in the initial cohort of human patients in phase I clinical PET studies. CONCLUSION: The whole-body and individual organ radiation dosimetry characteristics and pharmacologic safety of diagnostic dosages of (18)F-PEG(6)-IPQA in nonhuman primates indicate that this radiotracer should be acceptable for PET/CT studies in human patients.


Assuntos
Quinazolinas/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Envelhecimento/fisiologia , Animais , Peso Corporal/fisiologia , Feminino , Vesícula Biliar/diagnóstico por imagem , Vesícula Biliar/metabolismo , Processamento de Imagem Assistida por Computador , Macaca mulatta , Masculino , Tomografia por Emissão de Pósitrons , Quinazolinas/sangue , Quinazolinas/urina , Radiometria , Compostos Radiofarmacêuticos/sangue , Compostos Radiofarmacêuticos/urina , Distribuição Tecidual , Tomografia Computadorizada de Emissão , Contagem Corporal Total
17.
J Nucl Med ; 51(1): 121-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20048131

RESUMO

UNLABELLED: Because of the recent development of an iodopyridopyrimidinone Abl protein kinase inhibitor (PKI), (124)I-SKI-212230 ((124)I-SKI230), we investigated the feasibility of a PET-based molecular imaging method for the direct visualization of Abl kinase expression and PKI treatment. METHODS: In vitro pharmacokinetic properties, including specific and nonspecific binding of (124)I-SKI230 to its Abl kinase target and interaction with other PKIs, were assessed in cell-free medium and chronic myelogenous leukemia (CML) cells overexpressing BCR-Abl (K562), in comparison with BT-474 cells that are low in Abl expression. In a xenograft tumor model, we assessed the in vivo pharmacokinetics of (124)I-SKI230 using PET and postmortem tissue sampling. We also tested a paradigm of (124)I-SKI230 PET after treatment of the animal with a dose of Abl-specific PKI for the monitoring of the tumor response. RESULTS: In vitro studies confirmed that SKI230 binds to Abl kinase with nanomolar affinity, that selective uptake occurs in cell lines known to express Abl kinase, that RNAi knock-down supports specificity of cellular uptake due to Abl kinase, and that imatinib, an archetype Abl PKI, completely displaces SKI230. With SKI230, we obtained successful in vivo PET of Abl-expressing human tumors in a nude rat. We were also able to demonstrate evidence of substrate inhibition of in vivo radiotracer uptake in the xenograft tumor after treatment of the animal as a model of PKI treatment monitoring. CONCLUSION: These results support the hypothesis that molecular imaging using PET will be useful for the study of in vivo pharmacodynamics of Abl PKI molecular therapy in humans.


Assuntos
Inibidores Enzimáticos , Neoplasias/diagnóstico por imagem , Neoplasias/enzimologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Piridonas , Pirimidinas , Compostos Radiofarmacêuticos , Ligação Competitiva/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacocinética , Humanos , Marcação por Isótopo , Células K562 , Tomografia por Emissão de Pósitrons , Ligação Proteica , Piridonas/farmacocinética , Pirimidinas/farmacocinética , RNA Interferente Pequeno , Compostos Radiofarmacêuticos/farmacocinética
18.
Mol Imaging ; 1(1): 36-42, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12920859

RESUMO

Double prodrug activation gene therapy using the Escherichia coli cytosine deaminase (CD)-herpes simplex virus type 1 thymidine kinase (HSV1-tk) fusion gene (CD/TK) with 5-fluorocytosine (5FC), ganciclovir (GCV), and radiotherapy is currently under evaluation for treatment of different tumors. We assessed the efficacy of noninvasive imaging with [124I]FIAU (2'-fluoro-2'-deoxy-1-beta-D-arabinofuranosyl-5-iodo-uracil) and positron emission tomography (PET) for monitoring expression of the CD/TK fusion gene. Walker-256 tumor cells were transduced with a retroviral vector bearing the CD/TK gene (W256CD/TK cells). The activity of HSV1-TK and CD subunits of the CD/TK gene product was assessed in different single cell-derived clones of W256CD/TK cells using the FIAU radiotracer accumulation assay in cells and a CD enzyme assay in cell homogenates, respectively. A linear relationship was observed between the levels of CD and HSV1-tk subunit expression in corresponding clones in vitro over a wide range of CD/TK expression levels. Several clones of W256CD/TK cells with significantly different levels of CD/TK expression were selected and used to produce multiple subcutaneous tumors in rats. PET imaging of HSV1-TK subunit activity with [124I]FIAU was performed on these animals and demonstrated that different levels of CD/TK expression in subcutaneous W256CD/TK tumors can be imaged quantitatively. CD expression in subcutaneous tumor sample homogenates was measured using a CD enzyme assay. A comparison of CD and HSV1-TK subunit enzymatic activity of the CD/TK fusion protein in vivo showed a significant correlation. Knowing this relationship, the parametric images of CD subunit activity were generated. Imaging with [124I]FIAU and PET could provide pre- and posttreatment assessments of CD/TK-based double prodrug activation in clinical gene therapy trials.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/farmacocinética , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/genética , Radioisótopos do Iodo/farmacocinética , Nucleosídeo Desaminases/genética , Timidina Quinase/genética , Citosina Desaminase , Regulação Enzimológica da Expressão Gênica , Vetores Genéticos , Herpesvirus Humano 1/enzimologia , Humanos , Proteínas Recombinantes de Fusão/genética , Tomografia Computadorizada de Emissão , Células Tumorais Cultivadas
19.
Proc Natl Acad Sci U S A ; 99(6): 3400-5, 2002 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11891321

RESUMO

Human cells exposed to antifolates show a rapid increase in the levels of the enzyme dihydrofolate reductase (DHFR). We hypothesized that this adaptive response mechanism can be used to elevate cellular levels of proteins fused to DHFR. In this study, mouse cells transfected to express a green fluorescent protein-DHFR fusion protein and subsequently exposed to the antifolate trimetrexate (TMTX) showed a specific and time-dependent increase in cellular levels of the fusion protein. Next, human HCT-8 and HCT-116 colon cancer cells retrovirally transduced to express a DHFR-herpes simplex virus 1 thymidine kinase (HSV1 TK) fusion protein and treated with the DHFR inhibitor TMTX exhibited increased levels of the DHFR-HSV1 TK fusion protein and an increase in ganciclovir sensitivity by 250-fold. The level of fusion protein in antifolate-treated human tumor cells was increased in response to a 24-h exposure of methotrexate, trimetrexate, as well as dihydrofolate. This effect depended on the antifolate concentration and was independent of the fusion-protein mRNA levels, consistent with this increase occurring at a translational level. In a xenograft model, nude rats bearing DHFR-HSV1 TK-transduced HCT-8 tumors and treated with TMTX showed, after 24 h, a 2- to 4-fold increase of fusion-protein levels in tumor tissue from treated animals compared with controls, as determined by Western blotting. The fusion-protein increase was imaged with positron-emission tomography, where a substantially enhanced signal of the transduced tumor was detected in animals after antifolate administration. Drug-mediated elevation of cellular DHFR-fused proteins is a very useful method to modulate gene expression in vivo for imaging as well as therapeutic purposes.


Assuntos
Antagonistas do Ácido Fólico/farmacologia , Ácido Fólico/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/biossíntese , Tetra-Hidrofolato Desidrogenase/biossíntese , Animais , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/terapia , Relação Dose-Resposta a Droga , Indução Enzimática/efeitos dos fármacos , Ácido Fólico/farmacologia , Ganciclovir/farmacologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Metotrexato/farmacologia , Camundongos , Transplante de Neoplasias , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidina Quinase/biossíntese , Timidina Quinase/genética , Timidina Quinase/metabolismo , Tomografia Computadorizada de Emissão , Transdução Genética , Transplante Heterólogo , Trimetrexato/farmacologia , Células Tumorais Cultivadas
20.
Eur J Nucl Med Mol Imaging ; 30(9): 1281-91, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12664136

RESUMO

Radionuclide imaging has been demonstrated to be feasible to monitor transgene expression in vivo. We hypothesized that a potential application of this technique is to non-invasively detect in deep tissue, such as cancer cells metastatic to the liver, a specific molecular response following systemic drug treatment. Utilizing human colon adenocarcinoma cells derived from a patient's liver lesion we first developed a nude rat xenograft model for colorectal cancer metastatic to the liver. Expression of a dihydrofolate reductase-herpes simplex virus 1 thymidine kinase fusion (DHFR-HSV1 TK) transgene in the hepatic tumors was monitored in individual animals using the tracer [(124)I]2'-fluoro-2'-deoxy-5-iodouracil-beta- d-arabinofuranoside (FIAU) and a small animal micro positron emission tomograph (microPET), while groups of rats were imaged using the tracer [(131)I]FIAU and a clinical gamma camera. Growth of the human metastatic colorectal cancer cells in the rat liver was detected using magnetic resonance imaging and confirmed by surgical inspection. Single as well as multiple lesions of different sizes and sites were observed in the liver of the animals. Next, using a subset of rats bearing hepatic tumors, which were retrovirally bulk transduced to express the DHFR-HSV1 TK transgene, we imaged the fusion protein expression in the hepatic tumor of living rats using the tracer [(124)I]FIAU and a microPET. The observed deep tissue signals were highly specific for the tumors expressing the DHFR-HSV1 TK fusion protein compared with parental untransduced tumors and other tissues as determined by gamma counting of tissue samples. A subsequent study used the tracer [(131)I]FIAU and a gamma camera to monitor two groups of transduced hepatic tumor-bearing rats. Prior to imaging, one group was treated with trimetrexate to exploit DHFR-mediated upregulation of the fusion gene product. Imaging in the living animal as well as subsequent gamma counting of tissue samples showed increased signal and tracer accumulation, respectively, as compared to the group not treated with the antifolate. It is concluded that the two examined nucleotide imaging methods are feasible techniques for monitoring of DHFR-HSV TK fusion protein expression in hepatic colorectal tumor tissue in living animals.


Assuntos
Arabinofuranosiluracila/análogos & derivados , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Arabinofuranosiluracila/farmacocinética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Estudos de Viabilidade , Antagonistas do Ácido Fólico/uso terapêutico , Humanos , Radioisótopos do Iodo/farmacocinética , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Masculino , Transplante de Neoplasias , Especificidade de Órgãos , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Nus , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Tetra-Hidrofolato Desidrogenase/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Distribuição Tecidual , Transplante Heterólogo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Contagem Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA