Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ChemMedChem ; 11(15): 1638-45, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27377671

RESUMO

Iodoglucoazomycin (I-GAZ; N-(2-iodo-3-(6-O-glucosyl)propyl)-2-nitroimidazole), a non-glycosidic nitroimidazole-6-O-glucose adduct, was synthesized, radioiodinated, and evaluated as a substrate of glucose transporter 1 (GLUT1) for radiotheranostic (therapy+diagnostic) management of hypoxic tumors. Nucleophilic iodination of the nosylate synthon of I-GAZ followed by deprotection afforded I-GAZ in 74 % overall yield. I-GAZ was radioiodinated via 'exchange' labeling using [(123/131) I]iodide (50-70 % RCY) and then purified by Sep-Pak™ (>96 % RCP). [(131) I]I-GAZ was stable in 2 % ethanolic solution in sterile water for 14 days when stored at 5 °C. In cell culture, I-GAZ was found to be nontoxic to EMT-6 cells at concentrations <0.5 mm, and weakly radiosensitizing (SER 1.1 at 10 % survival of EMT-6 cells; 1.2 at 0.1 % survival in MCF-7 cells). The hypoxic/normoxic uptake ratio of [(123) I]I-GAZ in EMT-6 cells was 1.46 at 2 h, and under normoxic conditions the uptake of [(123) I]I-GAZ by EMT-6 cells was unaltered in the presence of 5 mm glucose. The biodistribution of [(131) I]I-GAZ in EMT-6 tumor-bearing Balb/c mice demonstrated rapid clearance from blood and extensive renal and hepatic excretion. Tumor/blood and tumor/muscle ratios reached ∼3 and 8, respectively, at 4 h post-injection. Regression analysis of the first order polynomial plots of the blood and tumor radioactivity concentrations supported a perfusion-excretion model with low hypoxia-dependent binding. [(131) I]I-GAZ was found to be stable in vivo, and did not deiodinate.


Assuntos
Glucose/análogos & derivados , Glucosídeos/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia , Nitroimidazóis/uso terapêutico , Compostos Radiofarmacêuticos/uso terapêutico , Animais , Linhagem Celular Tumoral , Glucose/síntese química , Glucose/farmacocinética , Glucose/uso terapêutico , Transportador de Glucose Tipo 1/química , Glucosídeos/síntese química , Glucosídeos/farmacocinética , Xenoenxertos , Humanos , Radioisótopos do Iodo , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias/patologia , Nitroimidazóis/síntese química , Nitroimidazóis/farmacocinética , Radiossensibilizantes/síntese química , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/uso terapêutico , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual , Carga Tumoral , Hipóxia Tumoral
2.
Semin Nucl Med ; 45(2): 122-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25704385

RESUMO

Hypoxia is prevalent in many solid tumors. Hypoxic tumors tend to exhibit rapid growth and aberrant vasculature, which lead to oxygen (O2) depletion and impaired drug delivery. The reductive environment in hypoxic tumors alters cellular metabolism, which can trigger transcriptional responses; induce genetic alterations; promote invasion, metastasis, resistance to radiotherapy and chemotherapy, tumor progression, and recurrence; and leads to poor local control and reduced survival rates. Therefore, exploiting the reductive microenvironment in hypoxic tumors by delivering electron-affinic, O2-mimetic radioactive drugs that bioreductively activate selectively in the hypoxic microenvironment offers a logical approach to molecular imaging of focal hypoxia. Because these agents also radiosensitize hypoxic cells, they provide an innovative approach to the therapy management of such tumors. To date, nuclear imaging of hypoxic tumor has proven to be clinically effective, whereas chemical radiosensitization by these compounds has not been helpful. The current review provides an insight into the chemistry, radiochemistry, and purification strategies for selected nitroaromatics that directly exploit the bioreductive environment in hypoxic cells. Both experimental and calculated single-electron reduction potentials of electron-affinic compounds, nitroimidazoles in particular, correlate with in vitro radiosensitizing properties, making them preferred choices for use as radiopharmaceuticals for diagnostic imaging and as sensitizers to enhance the killing effects of low-energy-transfer x-rays (O2-mimetic radiosensitization). Extensive research and careful drug design have led to the development of several potentially useful hypoxia-targeting drugs, for example, [(18)F]FAZA, [(18)F]FMISO, [(18)F]EF5, and [(123)I]IAZA, that accrue selectively in hypoxic cells. These molecular probes are now globally used in clinical hypoxia imaging, including cancer. Future innovative developments must, however, consider hypoxia-selective molecular processes and the physicochemical properties of the drugs that dictate their biodistribution, hypoxia-selective accumulation, pharmacokinetics, clearance, biochemical behavior, and metabolism. This will facilitate their ultimate transformation to effective molecular theranostics, leading to improved multimodal management of cancer.


Assuntos
Halogenação , Imagem Molecular/métodos , Sondas Moleculares/química , Nitrocompostos/química , Radioquímica/métodos , Hipóxia Celular , Humanos , Radioquímica/instrumentação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...