Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Nucl Med ; 58(2): 339-345, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27660146

RESUMEN

64Cu emits positrons as well as ß- particles and Auger and internal conversion electrons useful for radiotherapy. Our objective was to model the cellular dosimetry of 64Cu under different geometries commonly used to study the cytotoxic effects of 64Cu. METHODS: Monte Carlo N-Particle (MCNP) was used to simulate the transport of all particles emitted by 64Cu from the cell surface (CS), cytoplasm (Cy), or nucleus (N) of a single cell; monolayer in a well (radius = 0.32-1.74 cm); or a sphere (radius = 50-6,000 µm) of cells to calculate S values. The radius of the cell and N ranged from 5 to 12 µm and 2 to 11 µm, respectively. S values were obtained by MIRDcell for comparison. MCF7/HER2-18 cells were exposed in vitro to 64Cu-labeled trastuzumab. The subcellular distribution of 64Cu was measured by cell fractionation. The surviving fraction was determined in a clonogenic assay. RESULTS: The relative differences of MCNP versus MIRDcell self-dose S values (Sself) for 64Cu ranged from -0.2% to 3.6% for N to N (SN←N), 2.3% to 8.6% for Cy to N (SN←Cy), and -12.0% to 7.3% for CS to N (SN←CS). The relative differences of MCNP versus MIRDcell cross-dose S values were 25.8%-30.6% for a monolayer and 30%-34% for a sphere, respectively. The ratios of SN←N versus SN←Cy and SN←Cy versus SN←CS decreased with increasing ratio of the N of the cell versus radius of the cell and the size of the monolayer or sphere. The surviving fraction of MCF7 /: HER2-18 cells treated with 64Cu-labeled trastuzumab (0.016-0.368 MBq/µg, 67 nM) for 18 h versus the absorbed dose followed a linear survival curve with α = 0.51 ± 0.05 Gy-1 and R2 = 0.8838. This is significantly different from the linear quadratic survival curve of MCF7 /: HER2-18 cells exposed to γ-rays. CONCLUSION: MCNP- and MIRDcell-calculated S values agreed well. 64Cu in the N increases the dose to the N in isolated single cells but has less effect in a cell monolayer or small cluster of cells simulating a micrometastasis, and little effect in a sphere analogous to a tumor xenograft compared with 64Cu in the Cy or on the CS. The dose deposited by 64Cu is less effective for cell killing than γ-rays.


Asunto(s)
Fenómenos Fisiológicos Celulares/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Radioisótopos de Cobre/administración & dosificación , Radioisótopos de Cobre/análisis , Método de Montecarlo , Radiometría/métodos , Absorción de Radiación , Simulación por Computador , Relación Dosis-Respuesta en la Radiación , Humanos , Células MCF-7 , Modelos Biológicos , Modelos Estadísticos , Dosis de Radiación , Dispersión de Radiación
2.
Nucl Med Biol ; 43(12): 818-826, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27788375

RESUMEN

INTRODUCTION: Gold nanoparticles (AuNP; 30nm) were modified with polyethylene glycol (PEG) chains linked to trastuzumab for binding to HER2-positive breast cancer (BC) cells and diethylenetriaminepentaacetic acid (DTPA) for complexing the Auger electron-emitter, 111In (trastuzumab-AuNP-111In). Our objective was to determine the cytotoxicity of trastuzumab-AuNP-111In on HER2-positive BC cells in vitro and evaluate its tumor growth inhibition properties and normal tissue toxicity in vivo following intratumoral (i.t.) injection in mice with s.c. HER2-overexpressing BC xenografts. METHODS: Binding and internalization of trastuzumab-AuNP-111In or non-targeted AuNP-111In in SK-BR-3 (1-2×106 HER2/cell) and MDA-MB-361 (5×105 HER2/cell) human BC cells were studied. The surviving fraction (SF) of SK-BR-3 or MDA-MB-361 cells exposed to trastuzumab-AuNP-111In or AuNP-111In was determined. DNA double-strand breaks (DSBs) were assayed by probing for γ-H2AX. Tumor growth was monitored over 70days in CD1 athymic mice with s.c. MDA-MB-361 xenografts after i.t. injection of 10MBq (0.7mg; 2.6×1012 AuNP) of trastuzumab-AuNP-111In and normal tissue toxicity was assessed by monitoring body weight, complete blood cell (CBC) counts and serum alanine aminotransferase (ALT) and creatinine (Cr). RESULTS: Trastuzumab-AuNP-111In was specifically bound by SK-BR-3 and MDA-MB-361 cells. Trastuzumab-AuNP-111In was more efficiently internalized than AuNP-111In and localized to a peri-nuclear region. The SF fraction of SK-BR-3 cells was reduced by 1.8-fold by treatment with 3nM (7MBq/mL) of trastuzumab-AuNP-111In. The SF of MDA-MB-361 cells was reduced by 3.7-fold at 14.4nM (33.6MBq/mL). In comparison, non-targeted AuNP-111In at these concentrations reduced the SF of SK-BR-3 or MDA-MB-361 cells by 1.2-fold (P=0.03) and 1.7-fold (P<0.0001), respectively. DNA DSBs were greater in SK-BR-3 and MDA-MB-361 cells exposed to trastuzumab-AuNP-111In compared to AuNP-111In, but unlabeled trastuzumab-AuNP did not increase DNA DSBs. Local i.t. injection of trastuzumab-AuNP-111In in CD1 athymic mice with s.c. MDA-MB-361 tumors arrested tumor growth for 70days. There was no apparent normal tissue toxicity. The radiation absorbed dose deposited in the tumor by trastuzumab-AuNP-111In was 60.5Gy, while normal organs received <0.9Gy. CONCLUSION: These results are promising for further development of trastuzumab-AuNP-111In as a novel Auger electron-emitting radiation nanomedicine for local treatment of HER2-positive BC. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: A local radiation treatment for HER2-positive BC based on AuNP modified with trastuzumab and labeled with the Auger electron-emitter, 111In was developed and shown to arrest tumor growth with no normal tissue toxicity.


Asunto(s)
Neoplasias de la Mama/patología , Receptores ErbB/metabolismo , Oro/química , Radioisótopos de Indio/uso terapéutico , Nanopartículas del Metal/química , Trastuzumab/química , Trastuzumab/uso terapéutico , Animales , Transporte Biológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Receptores ErbB/inmunología , Humanos , Inyecciones Intralesiones , Ratones Desnudos , Dosis de Radiación , Trastuzumab/inmunología , Trastuzumab/metabolismo
3.
Med Phys ; 40(11): 114101, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24320476

RESUMEN

PURPOSE: The authors' aims were to model how various factors influence radiation dose enhancement by gold nanoparticles (AuNPs) and to propose a new modeling approach to the dose enhancement factor (DEF). METHODS: The authors used Monte Carlo N-particle (MCNP 5) computer code to simulate photon and electron transport in cells. The authors modeled human breast cancer cells as a single cell, a monolayer, or a cluster of cells. Different numbers of 5, 30, or 50 nm AuNPs were placed in the extracellular space, on the cell surface, in the cytoplasm, or in the nucleus. Photon sources examined in the simulation included nine monoenergetic x-rays (10-100 keV), an x-ray beam (100 kVp), and (125)I and (103)Pd brachytherapy seeds. Both nuclear and cellular dose enhancement factors (NDEFs, CDEFs) were calculated. The ability of these metrics to predict the experimental DEF based on the clonogenic survival of MDA-MB-361 human breast cancer cells exposed to AuNPs and x-rays were compared. RESULTS: NDEFs show a strong dependence on photon energies with peaks at 15, 30/40, and 90 keV. Cell model and subcellular location of AuNPs influence the peak position and value of NDEF. NDEFs decrease in the order of AuNPs in the nucleus, cytoplasm, cell membrane, and extracellular space. NDEFs also decrease in the order of AuNPs in a cell cluster, monolayer, and single cell if the photon energy is larger than 20 keV. NDEFs depend linearly on the number of AuNPs per cell. Similar trends were observed for CDEFs. NDEFs using the monolayer cell model were more predictive than either single cell or cluster cell models of the DEFs experimentally derived from the clonogenic survival of cells cultured as a monolayer. The amount of AuNPs required to double the prescribed dose in terms of mg Au/g tissue decreases as the size of AuNPs increases, especially when AuNPs are in the nucleus and the cytoplasm. For 40 keV x-rays and a cluster of cells, to double the prescribed x-ray dose (NDEF = 2) using 30 nm AuNPs, would require 5.1 ± 0.2, 9 ± 1, 10 ± 1, 10 ± 1 mg Au/g tissue in the nucleus, in the cytoplasm, on the cell surface, or in the extracellular space, respectively. Using 50 nm AuNPs, the required amount decreases to 3.1 ± 0.3, 8 ± 1, 9 ± 1, 9 ± 1 mg Au/g tissue, respectively. CONCLUSIONS: NDEF is a new metric that can predict the radiation enhancement of AuNPs for various experimental conditions. Cell model, the subcellular location and size of AuNPs, and the number of AuNPs per cell, as well as the x-ray photon energy all have effects on NDEFs. Larger AuNPs in the nucleus of cluster cells exposed to x-rays of 15 or 40 keV maximize NDEFs.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Oro/química , Nanopartículas del Metal/química , Braquiterapia/métodos , Núcleo Celular/diagnóstico por imagen , Citoplasma/diagnóstico por imagen , Electrones , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Radioisótopos de Yodo/química , Método de Montecarlo , Paladio/química , Fotones , Dosis de Radiación , Radioisótopos/química , Cintigrafía , Rayos X
4.
Breast Cancer Res Treat ; 137(1): 81-91, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23160926

RESUMEN

The purpose of this study was to evaluate the effect of molecularly targeted gold nanoparticles (AuNPs) on tumor radiosensitization both in vitro and in vivo. Human Epidermal Growth Factor Receptor-2 (HER-2)-targeted AuNPs (Au-T) were synthesized by conjugating trastuzumab (Herceptin) to 30 nm AuNPs. In vitro, the cytotoxicity of Au-T or non-targeted AuNPs (Au-P) was assessed by γ-H2AX immunofluorescence microscopy for DNA damage and clonogenic survival assays. In vivo, athymic mice bearing subcutaneous MDA-MB-361 xenografts were treated with a single dose of 11 Gy of 100 kVp X-rays 24 h after intratumoral injection of Au-T (~0.8 mg of Au) or no X-radiation. Normal tissue toxicity was determined by hematology or biochemistry parameters. The combination of Au-P or Au-T with X-ray exposure increased the formation of γ-H2AX foci by 1.7 (P = 0.054) and 3.3 (P = 0.024) fold in comparison to X-radiation alone, respectively. The clonogenic survival of cells exposed to Au-T and X-rays was significantly lower from that of cells exposed to X-radiation alone, which translated to a dose enhancement factor of 1.6. In contrast, survival of cells exposed to Au-P and X-rays versus X-radiation alone were not significantly different. In vivo, the combination of Au-T and X-radiation resulted in regression of MDA-MB-361 tumors by 46 % as compared to treatment with X-radiation (16.0 % increase in tumor volume). No significant normal tissue toxicity was observed. Radiosensitization of breast cancer to X-radiation with AuNPs was successfully achieved with an optimized therapeutic strategy of molecular targeting of HER-2 and intratumoral administration.


Asunto(s)
Neoplasias de la Mama/radioterapia , Oro/química , Nanopartículas del Metal/química , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena , Femenino , Histonas/metabolismo , Humanos , Inyecciones Intralesiones , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/toxicidad , Dosificación Radioterapéutica , Receptor ErbB-2/metabolismo , Carga Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...