Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Pediatr Radiol ; 45(7): 1039-45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25524437

RESUMEN

BACKGROUND: Thyroid cancer is the most common endocrine malignancy with relatively good prognosis in children. However, unlike adults, children usually present with more advanced disease and have a higher local recurrence and distant metastases. Thus surveillance for recurrence is a major goal of long-term follow-up. OBJECTIVE: This retrospective study evaluates the diagnostic value of ultrasound (US) imaging in the post-therapy surveillance of children with differentiated thyroid cancer. MATERIALS AND METHODS: We reviewed the charts of 54 children (40 girls; mean age 14.3 ± 3.6 years) with differentiated thyroid cancer treated with total or near-total thyroidectomy. Forty children (29 girls and 11 boys) who had routine follow-up US examinations (112 studies) were included for the evaluation of US accuracy in the follow-up of pediatric differentiated thyroid cancer. Histopathology, stimulated thyroglobulin determination, post-therapy whole-body iodine scan and clinical follow-up were used as the standards of reference. RESULTS: Mean period of follow-up was 34 months. The frequency of recurrence was 42% (17/40). Seventeen percent of the children had lung metastases either at presentation or on follow-up. In all cases of lung metastases, stimulated thyroglobulin level was greater than 10 ng/ml. The sensitivity was 85.7%, specificity 89.4%, negative predictive value 94.4% and positive predictive value 75% for US in detecting loco-regional recurrence in follow-up studies of pediatric differentiated thyroid cancer. In 17.3% (18/104) of studies, the results of stimulated thyroglobulin and US were discordant. CONCLUSION: US showed very good sensitivity and specificity and a high negative predictive value for evaluation of loco-regional involvement in follow-up of pediatric differentiated thyroid cancer. Diagnostic whole-body iodine scan is indicated when serum anti-thyroglobulin Ab is high, or in cases of discordant findings between US and stimulated thyroglobulin levels, or when stimulated thyroglobulin levels are >10 ng/ml (to evaluate for lung metastasis).


Asunto(s)
Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/secundario , Recurrencia Local de Neoplasia/diagnóstico por imagen , Neoplasias Primarias Secundarias/diagnóstico por imagen , Complicaciones Posoperatorias/diagnóstico por imagen , Neoplasias de la Tiroides/diagnóstico por imagen , Adolescente , Femenino , Estudios de Seguimiento , Humanos , Pulmón/diagnóstico por imagen , Masculino , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Estudios Retrospectivos , Sensibilidad y Especificidad , Glándula Tiroides/diagnóstico por imagen , Glándula Tiroides/cirugía , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/cirugía , Ultrasonografía
2.
AJR Am J Roentgenol ; 200(2): 408-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23345365

RESUMEN

OBJECTIVE: The utility of dual-time-point (18)F-FDG PET/CT in differentiating benign from malignant processes in pediatric patients was assessed. SUBJECTS AND METHODS: Twenty-one patients (13 girls and eight boys; age range, 1-17 years) with suspected malignancy underwent dual-time-point FDG PET/CT. Scan 1 was performed at approximately 60 minutes after i.v. injection of 5.18 MBq/kg of FDG, and scan 2 was performed at 121 ± 43 minutes after the first scan. Regions of interest were overlaid onto each non-attenuated-corrected image, and semiquantitative analysis was performed using the standardized uptake value (SUV) obtained from early and delayed images. A retention index was calculated according to the following equation: [(delayed SUV - early SUV) / early SUV] × 100. Results were compared prospectively in relation to pathologic examination or other conventional radiologic imaging or clinical follow-up. A retention index of 10% or higher was chosen as a cutoff for differentiating malignant from benign entities. RESULTS: For patients with malignant disease, the average SUV increased from 7.3 ± 1.2 to 10.9 ± 2.7 between the two time points, whereas the SUV changed from 4.5 ± 0.8 to 4.2 ± 1.0 for patients with benign lesions. The average retention index was 37.1% ± 10.8% for patients with malignant lesions versus -9.9% ± 7.1% for benign lesions (p < 0.01). With a cutoff value of 10% or higher for the retention index, the sensitivity and specificity of dual-time-point FDG PET/CT were 77% and 80%, respectively. CONCLUSION: These data show that dual-time-point FDG PET/CT is useful in distinguishing malignant from benign processes in pediatric patients.


Asunto(s)
Fluorodesoxiglucosa F18 , Imagen Multimodal/métodos , Neoplasias/diagnóstico por imagen , Tomografía de Emisión de Positrones , Radiofármacos , Tomografía Computarizada por Rayos X , Adolescente , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Masculino , Estudios Prospectivos , Sensibilidad y Especificidad
3.
J Nucl Med ; 50(8): 1340-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19617342

RESUMEN

UNLABELLED: Pertuzumab is a HER2 dimerization inhibitor that binds to an epitope unique from that of trastuzumab. Our objective was to determine whether SPECT with (111)In-diethylenetriaminepentaacetic acid-pertuzumab ((111)In-DTPA-pertuzumab) could sensitively detect an early molecular response to trastuzumab manifested by HER2 downregulation and a later tumor response revealed by a decreased number of HER2-positive viable tumor cells. METHODS: Changes in HER2 density in SKBr-3 and MDA-MB-361 BC cells exposed to trastuzumab (14 microg/mL) in vitro were measured by saturation binding assays using (111)In-DTPA-pertuzumab and by confocal immunofluorescence microscopy and flow cytometry with fluorescein isothiocyanate-labeled HER2/neu antibodies. Imaging of HER2 downregulation was studied in vivo in athymic mice with subcutaneous MDA-MB-361 tumors treated for 3 d with trastuzumab (4 mg/kg) or nonspecific human IgG (hIgG) or phosphate-buffered saline (PBS). Imaging of tumor response to trastuzumab was studied in mice bearing subcutaneous MDA-MB-361 xenografts treated with trastuzumab (4 mg/kg), followed by weekly doses of nonspecific hIgG or rituximab or PBS (2 mg/kg). Mice were imaged on a micro-SPECT/CT system at 72 h after injection of (111)In-DTPA-pertuzumab. Tumor and normal-tissue biodistribution was determined. RESULTS: (111)In-DTPA-pertuzumab saturation binding to SKBr-3 and MDA-MB-361 cells was significantly decreased at 72 h after exposure in vitro to trastuzumab (14 microg/mL), compared with untreated controls (62% +/- 2%, P < 0.0001; 32% +/- 9%, P < 0.0002, respectively). After 3 d of trastuzumab, in vivo tumor uptake of (111)In-DTPA-pertuzumab decreased 2-fold in trastuzumab- versus PBS-treated mice (13.5 +/- 2.6 percentage injected dose per gram [%ID/g] vs. 28.5 +/- 9.1 %ID/g, respectively; P < 0.05). There was also a 2-fold decreased tumor uptake in trastuzumab- versus PBS-treated mice by image volume-of-interest analysis (P = 0.05), suggesting trastuzumab-mediated HER2 downregulation. After 3 wk of trastuzumab, tumor uptake of (111)In-DTPA-pertuzumab decreased 4.5-fold, compared with PBS-treated mice (7.6 +/- 0.4 vs. 34.6 +/- 9.9 %ID/g, respectively; P < 0.001); this decrease was associated with an almost-completed eradication of HER2-positive tumor cells determined immunohistochemically. CONCLUSION: (111)In-DTPA-pertuzumab sensitively imaged HER2 downregulation after 3 d of treatment with trastuzumab and detected a reduction in viable HER2-positive tumor cells after 3 wk of therapy in MDA-MB-361 human breast cancer xenografts.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Neoplasias de la Mama , Receptor ErbB-2/metabolismo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada por Rayos X/métodos , Animales , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Radioisótopos de Indio/farmacocinética , Ratones , Ratones Desnudos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Tomografía Computarizada de Emisión de Fotón Único/veterinaria , Tomografía Computarizada por Rayos X/veterinaria , Trastuzumab , Resultado del Tratamiento
4.
J Nucl Med ; 49(9): 1498-505, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18703606

RESUMEN

UNLABELLED: Our goals in this study were to determine whether (111)In-trastuzumab coupled to peptides harboring nuclear localizing sequences (NLSs) could kill trastuzumab-resistant breast cancer cell lines through the emission of Auger electrons and whether the combination of radiosensitization with methotrexate (MTX) would augment the cytotoxicity of this radiopharmaceutical. METHODS: Trastuzumab was derivatized with sulfosuccinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate for reaction with NLS peptides and then conjugated with diethylenetriaminepentaacetic acid for labeling with (111)In. HER2 expression was determined by Western blot and by radioligand binding assay using (111)In-trastuzumab in a panel of breast cancer cell lines, including SK-BR-3, MDA-MB-231 and its HER2-transfected subclone (231-H2N), and 2 trastuzumab-resistant variants (TrR1 and TrR2). Nuclear importation of (111)In-NLS-trastuzumab and (111)In-trastuzumab in breast cancer cells was measured by subcellular fractionation, and the clonogenic survival of these cells was determined after incubation with (111)In-NLS-trastuzumab, (111)In-trastuzumab, or trastuzumab (combined with or without MTX). Survival curves were analyzed according to the dose-response model, and the radiation-enhancement ratio was calculated from the survival curve parameters. RESULTS: The expression of HER2 was highest in SK-BR-3 cells (12.6 x 10(5) receptors/cell), compared with 231-H2N and TrR1 cells (6.1 x 10(5) and 5.1 x 10(5) receptors/cell, respectively), and lowest in MDA-MB-231 and TrR2 cells (0.4 x 10(5) and 0.6 x 10(5) receptors/cell, respectively). NLS peptides increased the nuclear uptake of (111)In-trastuzumab in MDA-MB-231, 231-H2N, TrR1, and TrR2 cells from 0.1%+/-0.01%, 2.5%+/-0.2%, 2.8%+/-0.7%, and 0.5%+/-0.1% to 0.5%+/-0.1%, 4.6%+/-0.1%, 5.2%+/-0.6%, and 1.5%+/-0.2%, respectively. The cytotoxicity of (111)In-NLS-trastuzumab on breast cancer cells was directly correlated with the HER2 expression densities of the cells. On a molar concentration basis, the effective concentration required to kill 50% of 231-H2N and TrR1 cells for (111)In-NLS-trastuzumab was 9- to 12-fold lower than for (111)In-trastuzumab and 16- to 77-fold lower than for trastuzumab. MDA-MB-231 and TrR2 cells were less sensitive to (111)In-NLS-trastuzumab or (111)In-trastuzumab, and both cell lines were completely insensitive to trastuzumab. The radiation-enhancement ratio induced by MTX for 231-H2N and TrR1 cells after exposure to (111)In-NLS-trastuzumab was 1.42 and 1.68, respectively. CONCLUSION: Targeted Auger electron radioimmunotherapy with (111)In-NLS-trastuzumab can overcome resistance to trastuzumab, and MTX can potently enhance the sensitivity of HER2-overexpressing breast cancer cells to the lethal Auger electrons emitted by this radiopharmaceutical.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Metotrexato/administración & dosificación , Señales de Localización Nuclear/administración & dosificación , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de la radiación , Humanos , Radioisótopos de Indio/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Radiofármacos/administración & dosificación , Trastuzumab
5.
Cancer Biother Radiopharm ; 23(1): 3-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18298325

RESUMEN

Intracellular compartments, in particular the cytoplasm or nucleus, have generally been poorly accessible or inaccessible to radiolabeled biomolecules (e.g., monoclonal antibodies [mAbs], peptides, or oligonucleotides [ODNs]). However, recently cell-penetrating peptides (CPPs) and nuclear localizing peptide sequences (NLSs) have been shown to have the capability of inserting biomolecules into cells and transporting them to the cell nucleus. This discovery now presents intriguing new opportunities to design radiopharmaceuticals that could potentially probe, through imaging, the expression of key intracellular or intranuclear regulatory proteins that define the tumor phenotype, predict outcome, or act as sensitive reporters of response or resistance to treatment. CPPs could also more efficiently internalize radiolabeled antisense ODNs or peptide nucleic acids (PNAs) into tumor cells to enhance the sensitivity of imaging gene expression at the mRNA level. Perhaps one of the most exciting new developments to emerge is the use of NLS to route mAbs and peptides conjugated to nanometer-micrometer range Auger-electron-emitting radionuclides (e.g., (111)In) to the nucleus of cancer cells following their receptor-mediated internalization. In the nucleus, these electrons are highly potent in causing lethal DNA strand breaks. In some cases, NLSs are present naturally in peptide growth factors or their receptors, where they function to deliver internalized ligands to the nucleus, or alternatively, they can be introduced synthetically. This update reviews the properties of CPPs and NLS and focuses on their use for inserting radiolabeled biomolecules into cancer cells for imaging or targeted Auger electron radiotherapy of malignancies.


Asunto(s)
Secuencias de Aminoácidos , Núcleo Celular/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia , Radiofármacos/metabolismo , Transporte Activo de Núcleo Celular , Humanos , Señales de Localización Nuclear , Transporte de Proteínas , Cintigrafía
6.
J Nucl Med ; 48(8): 1357-68, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17631548

RESUMEN

UNLABELLED: The cytotoxicity and tumor-targeting properties of the anti-HER2/neu monoclonal antibody trastuzumab modified with peptides (CGYGPKKKRKVGG) harboring the nuclear localization sequence ([NLS] italicized) of simian virus 40 large T-antigen and radiolabeled with (111)In were evaluated. METHODS: Trastuzumab was derivatized with sulfosuccinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (sulfo-SMCC) for reaction with NLS-peptides and labeled with (111)In using diethylenetriaminepentaacetic acid (DTPA). The immunoreactivity of (111)In-NLS-trastuzumab was determined by its ability to displace the binding of trastuzumab to SK-BR-3 human breast cancer (BC) cells. Cellular uptake and nuclear localization were evaluated in SK-BR-3, MDA-MB-361, and MDA-MB-231 BC cells, expressing high, intermediate, or very low levels of HER2/neu, respectively, by cell fractionation and confocal microscopy. Biodistribution and nuclear uptake were compared in athymic mice bearing MDA-MB-361 xenografts. The cytotoxicity of (111)In-trastuzumab and (111)In-NLS-trastuzumab was studied by clonogenic assays, and DNA damage was assessed by probing for phosphorylated histone H2AX (gammaH2AX) foci. RESULTS: The dissociation constant for binding of (111)In-NLS-trastuzumab to SK-BR-3 cells was reduced <3-fold compared with that of (111)In-trastuzumab, demonstrating relatively preserved receptor-binding affinity. The receptor-mediated internalization of (111)In-trastuzumab in SK-BR-3, MDA-MB-361, and MDA-MB-231 cells increased significantly from 7.2% +/- 0.9%, 1.3% +/- 0.1%, and 0.2% +/- 0.05% to 14.4% +/- 1.8%, 6.3% +/- 0.2%, and 0.9% +/- 0.2% for (111)In-NLS-trastuzumab harboring 6 NLS-peptides, respectively. NLS-trastuzumab localized in the nuclei of BC cells, whereas unmodified trastuzumab remained surface-bound. Conjugation of (111)In-trastuzumab to NLS-peptides did not affect its tissue biodistribution but promoted specific nuclear uptake in MDA-MB-361 xenografts (2.4-2.9 %ID/g [percentage injected dose per gram] for (111)In-NLS-trastuzumab and 1.1 %ID/g for (111)In-trastuzumab). (111)In-NLS-trastuzumab was 5- and 2-fold more potent at killing SK-BR-3 and MDA-MB-361 cells than (111)In-trastuzumab, respectively, whereas toxicity toward MDA-MB-231 cells was minimal. (111)In-NLS-trastuzumab was 6-fold more effective at killing SK-BR-3 cells than unlabeled trastuzumab. Formation of gammaH2AX foci occurred in a greater proportion of BC cells after incubation with (111)In-NLS-trastuzumab compared with (111)In-trastuzumab or unlabeled trastuzumab. CONCLUSION: NLS-peptides routed (111)In-trastuzumab to the nucleus of HER2/neu-positive human BC cells, rendering the radiopharmaceutical lethal to the cells through the emission of nanometer-micrometer range Auger electrons. The greater cytotoxic potency of (111)In-NLS-trastuzumab compared with unlabeled trastuzumab in vitro and its favorable tumor-targeting properties in vivo suggest that it could be an effective targeted radiotherapeutic agent for HER2/neu-amplified BC in humans.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/radioterapia , Radioisótopos de Indio/uso terapéutico , Señales de Localización Nuclear , Radioinmunoterapia , Receptor ErbB-2/análisis , Transporte Activo de Núcleo Celular , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Células Cultivadas , Daño del ADN , Electrones , Femenino , Humanos , Receptor ErbB-2/antagonistas & inhibidores , Distribución Tisular , Trastuzumab
7.
J Nucl Med ; 48(9): 1562-70, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17704253

RESUMEN

UNLABELLED: (111)In-DTPA-human epidermal growth factor ((111)In-DTPA-hEGF [DTPA is diethylenetriaminepentaacetic acid]) is an Auger electron-emitting radiopharmaceutical that targets EGF receptor (EGFR)-positive cancer. The purpose of this study was to determine the effect of EGFR inhibition by gefitinib on the internalization, nuclear translocation, and cytotoxicity of (111)In-DTPA-hEGF in EGFR-overexpressing MDA-MB-468 human breast cancer cells. METHODS: Western blot analysis was used to determine the optimum concentration of gefitinib to abolish EGFR activation. Internalization and nuclear translocation of fluorescein isothiocyanate-labeled hEGF were evaluated by confocal microscopy in MDA-MB-468 cells (1.3 x 10(6) EGFRs/cell) in the presence or absence of 1 microM gefitinib. The proportion of radioactivity partitioning into the cytoplasm and nucleus of MDA-MB-468 cells after incubation with (111)In-DTPA-hEGF for 24 h at 37 degrees C in the presence or absence of 1 microM gefitinib was measured by cell fractionation. DNA double-strand breaks caused by (111)In were quantified using the gamma-H2AX assay, and radiation-absorbed doses were estimated. Clonogenic survival assays were used to measure the cytotoxicity of (111)In-DTPA-hEGF alone or in combination with gefitinib. RESULTS: Gefitinib (1 microM) completely abolished EGFR phosphorylation in MDA-MB-468 cells. Internalization and nuclear translocation of fluorescein isothiocyanate-labeled EGF were not diminished in gefitinib-treated cells compared with controls. The proportion of internalized (111)In that localized in the nucleus was statistically significantly greater when (111)In-DTPA-hEGF was combined with gefitinib compared with (111)In-DTPA-hEGF alone (mean +/- SD: 26.0% +/- 5.5% vs. 14.6% +/- 4.0%, respectively; P < 0.05). Induction of gamma-H2AX foci was greater in MDA-MB-468 cells that were treated with (111)In-DTPA-hEGF (250 ng/mL, 1.5 MBq/mL) plus gefitinib (1 microM ) compared with those treated with (111)In-DTPA-hEGF alone (mean +/- SD: 35 +/- 4 vs. 24 +/- 5 foci per nucleus, respectively). In clonogenic assays, a significant reduction in the surviving fraction was observed when (111)In-DTPA-hEGF (5 ng/mL, 6 MBq/microg) was combined with gefitinib (1 microM ) compared with (111)In-DTPA-hEGF alone (42.9% +/- 5.7% vs. 22.9% +/- 3.6%, respectively; P < 0.01). CONCLUSION: The efficacy of (111)In-DTPA-hEGF depends on internalization and nuclear uptake of the radionuclide. Nuclear uptake, DNA damage, and cytotoxicity are enhanced when (111)In-DTPA-hEGF is combined with gefitinib. These results suggest a potential therapeutic role for peptide receptor radionuclide therapy in combination with tyrosine kinase inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Núcleo Celular/metabolismo , Electrones , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Ácido Pentético/análogos & derivados , Radiofármacos/farmacología , Antineoplásicos/farmacocinética , Compartimento Celular , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacocinética , Receptores ErbB/metabolismo , Fluoresceína-5-Isotiocianato , Colorantes Fluorescentes , Gefitinib , Humanos , Radioisótopos de Indio , Ácido Pentético/farmacocinética , Ácido Pentético/farmacología , Fosforilación , Transporte de Proteínas , Quinazolinas/farmacología , Radiofármacos/farmacocinética , Transducción de Señal
8.
Int J Mol Imaging ; 2016: 6045894, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27313888

RESUMEN

We performed an observational pilot study of 18F-FLT PET/CT in pediatric lymphoma. Eight patients with equivocal 18F-FDG PET/CT underwent imaging with 18F-FLT PET/CT. No immediate adverse reactions to 18F-FLT were observed. Compared to 18F-FDG, 18F-FLT uptake was significantly higher in bone marrow and liver (18F-FLT SUV 8.6 ± 0.6 and 5.0 ± 0.3, versus 18F-FDG SUV 1.9 ± 0.1 and 3.4 ± 0.7, resp., p < 0.05). In total, 15 lesions were evaluated with average 18F-FDG and 18F-FLT SUVs of 2.6 ± 0.1 and 2.0 ± 0.4, respectively. Nonspecific uptake in reactive lymph nodes and thymus was observed. Future studies to assess the clinical utility of 18F-FLT PET/CT in pediatric lymphoma are planned.

9.
J Nucl Med ; 51(3): 477-83, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20150272

RESUMEN

UNLABELLED: Our goal in this study was to elucidate the mechanisms by which methotrexate radiosensitizes HER2-positive human breast cancer cells to the Auger electron emitter (111)In-trastuzumab modified with nuclear-localization sequence peptides ((111)In-NLS-trastuzumab) and to compare these mechanisms with the potential sensitizing effects of paclitaxel and doxorubicin when combined with this radiopharmaceutical. METHODS: Experiments were performed in MDA-MB-231 human breast cancer cells, their HER2-transfected subclones (231-H2N), and 2 trastuzumab-resistant variants (trastuzumab-resistant-1 and -2 [TrR1 and TrR2]). Effects of coexposure of these cells to (111)In-NLS-trastuzumab and low-dose, radiosensitizing methotrexate, paclitaxel, or doxorubicin were assessed by clonogenic cell-survival assay. Quantification of residual DNA damage was measured by the gammaH2AX-immunofluorescence assay, and cell cycle distribution was measured by fluorescence-activated cell sorting analysis. The radiation-enhancement ratio was calculated as the ratio of the surviving fraction (SF) of cells treated with (111)In-NLS-trastuzumab alone to that of cells treated concurrently with (111)In-NLS-trastuzumab and methotrexate, paclitaxel, or doxorubicin. RESULTS: A reduction in the SF in HER2-positive 231-H2N (55.7% +/- 1.3%) and TrR1 (62.6% +/- 6.5%) cells was demonstrated after exposure to (111)In-NLS-trastuzumab (approximately 0.2 MBq/microg, 100 nmol/L) but not in MDA-MB-231 or TrR2 cells expressing low levels of HER2 (SF > 90%, P > 0.05). Coadministration of methotrexate, paclitaxel, or doxorubicin enhanced the cytotoxicity of (111)In-NLS-trastuzumab toward 231-H2N and TrR1 cells but not toward MDA-MB-231 or TrR2 cells. The radiation-enhancement ratios for methotrexate, paclitaxel, and doxorubicin for 231-H2N or TrR1 cells were 2.0-2.2, 1.6-1.8, and 2.7-2.8, respectively. Methotrexate or doxorubicin combined with (111)In-NLS-trastuzumab, compared to treatment with (111)In-NLS-trastuzumab alone, significantly increased residual gammaH2AX foci in 231-H2N and TrR1 cells but not in MDA-MB-231 or TrR2 cells or in any cell line treated concurrently with paclitaxel and (111)In-NLS-trastuzumab. Cells exposed to low-dose methotrexate accumulated in the G(1)/S phase of the cell cycle, whereas low-dose paclitaxel or doxorubicin caused cells to arrest in the G(2)/M phase. CONCLUSION: Low-dose methotrexate, paclitaxel, or doxorubicin potently sensitized HER2-overexpressing human breast cancer cells, with and without acquired trastuzumab-resistance, to the Auger electron emissions from (111)In-NLS-trastuzumab through cell cycle distribution changes and in part through the inhibitory effects of these agents on DNA damage repair.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/patología , Electrones , Radioisótopos de Indio/química , Señales de Localización Nuclear/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Receptor ErbB-2/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Citometría de Flujo , Humanos , Metotrexato/administración & dosificación , Metotrexato/farmacología , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Trastuzumab
10.
J Nucl Med ; 51(7): 1084-91, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20554744

RESUMEN

UNLABELLED: (111)In-nuclear localization sequence-trastuzumab is a radioimmunotherapeutic agent consisting of trastuzumab modified with NLS peptides (CGYGPKKKRKVGG) and labeled with the Auger electron emitter (111)In. Our objectives were to evaluate the tumor growth-inhibitory properties and normal-tissue toxicity of (111)In-NLS-trastuzumab in mice after intraperitoneal administration. METHODS: The pharmacokinetics of (111)In-NLS-trastuzumab after intravenous (tail vein) or intraperitoneal injection in BALB/c mice were compared. Normal-tissue toxicity was determined in BALB/c mice at 2 wk after intraperitoneal injection of 3.7-18.5 MBq (4 mg/kg) of (111)In-NLS-trastuzumab by monitoring body weight, histopathologic examination of tissues, and hematology (white blood cell, platelet, red blood cell, and hemoglobin) or clinical biochemistry (alanine transaminase and creatinine) parameters. A no-observable-adverse-effect-level (NOAEL) dose was defined. Athymic mice bearing subcutaneous MDA-MB-361 or MDA-MB-231 human breast cancer xenografts (5.0 x 10(5) or 0.5 x 10(5) HER2/cell, respectively) were treated with a single NOAEL dose or 2 doses administered intraperitoneally and separated by 2 wk. Control groups were administered (111)In-trastuzumab, trastuzumab, nonspecific (111)In-NLS-human IgG (hIgG), or normal saline. RESULTS: The bioavailability of (111)In-NLS-trastuzumab after intraperitoneal injection was 0.7. The NOAEL dose was 9.25 MBq (4 mg/kg); doses greater than or equal to 18.5 MBq decreased white blood cell or platelet counts, and doses of 27.7 MBq decreased red blood cell counts. There was no increase in alanine transaminase or creatinine at any doses tested. There were no morphologic changes to the liver, kidneys, heart, or spleen or loss of body weight. A single dose of (111)In-NLS-trastuzumab (9.25 MBq)-compared with mice receiving (111)In-trastuzumab, trastuzumab, (111)In-NLS-hIgG, or normal saline-significantly slowed the rate of growth of MDA-MB-361 tumors over 60 d (0.014 d(-1) vs. 0.033 d(-1), 0.046 d(-1), 0.030 d(-1), and 0.061 d(-1), respectively; P < 0.05). (111)In-NLS-trastuzumab had no effect on the growth of MDA-MB-231 tumors. Two doses of (111)In-NLS-trastuzumab (9.25 MBq; 4 mg/kg) separated by 2 wk increased the survival of mice with MDA-MB-361 tumors, compared with mice treated with trastuzumab or normal saline (>140 d vs. 96 and 84 d, respectively; P < 0.001 or 0.027, respectively). CONCLUSION: (111)In-NLS-trastuzumab is a promising radioimmunotherapeutic agent that could be effective for treatment of HER2-overexpressing breast cancer in humans.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/toxicidad , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Neoplasias de la Mama/radioterapia , Señales de Localización Nuclear/farmacología , Señales de Localización Nuclear/toxicidad , Radiofármacos/farmacocinética , Radiofármacos/toxicidad , Receptor ErbB-2/metabolismo , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacocinética , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Inmunoglobulina G/química , Radioisótopos de Indio/farmacocinética , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Marcaje Isotópico , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Señales de Localización Nuclear/farmacocinética , Radioinmunoterapia , Análisis de Supervivencia , Trastuzumab
11.
J Nucl Med ; 50(11): 1848-56, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19837760

RESUMEN

UNLABELLED: Breast cancers (BCs) with high human epidermal growth factor receptor type 2 (HER2) expression are most likely to respond to trastuzumab; however, the mechanisms of action of trastuzumab are complex and there are no established biomarkers to accurately monitor treatment outcome in individual patients. Therefore, our aim was to determine, in human BC xenografts in athymic mice treated with trastuzumab, whether there were any changes in (18)F-FDG uptake that were associated with response to the drug and that could have utility in monitoring response in patients. METHODS: Baseline tumor uptake of (18)F-FDG was measured in mice with MDA-MB-361 HER2-overexpressing xenografts and MDA-MB-231 xenografts with low HER2 expression by small-animal PET imaging on day 0. Mice were treated with phosphate-buffered saline (PBS) or trastuzumab (4 mg/kg), and small-animal PET was repeated 2 d after treatment. Maintenance doses of trastuzumab (2 mg/kg) or PBS were administered on days 7 and 14, and mice were imaged again on days 9 and 16. Tumor uptake was measured as percentage injected dose per gram (%ID/g) by volume-of-interest analysis on days 0 (baseline), 2, 9, and 16, followed by biodistribution studies on day 16. Tumor growth was measured, and a tumor growth index was calculated. RESULTS: The treatment of mice with trastuzumab, compared with control mice treated with PBS, resulted in a significant decrease in tumor uptake of (18)F-FDG in HER2-overexpressing MDA-MB-361 xenografts after 16 d of treatment (2.6 +/- 0.8 %ID/g vs. 4.6 +/- 1.8 %ID/g, respectively; P < 0.03) but not after 2 or 9 d of treatment (P = 0.28-0.32). In contrast, there was no significant change in the tumor uptake of MDA-MB-231 xenografts with low HER2 expression during the entire course of therapy (4.4 +/- 1.7 %ID/g vs. 3.6 +/- 1.1 %ID/g, respectively; P = 0.31). Trastuzumab treatment, compared with PBS treatment of controls, resulted in significant growth inhibition of MDA-MB-361 xenografts as early as 10 d from the initiation of treatment (tumor growth index, 0.7 +/- 0.2 vs. 1.7 +/- 0.3, respectively; P < 0.0005), whereas no tumor growth inhibition was observed for MDA-MB-231 xenografts (5.3 +/- 2.7 and 5.2 +/- 3.0; P = 0.95). CONCLUSION: Changes in the tumor uptake of (18)F-FDG after therapy accurately identified responding and nonresponding human BC xenografts in athymic mice treated with trastuzumab; however, diminished glucose utilization did not precede changes in tumor volume.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Fluorodesoxiglucosa F18 , Animales , Anticuerpos Monoclonales Humanizados , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Regulación Neoplásica de la Expresión Génica , Genes erbB-2/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Tomografía de Emisión de Positrones , Sensibilidad y Especificidad , Distribución Tisular , Tomografía Computarizada por Rayos X , Trastuzumab , Resultado del Tratamiento
12.
Cell ; 123(2): 347-58, 2005 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-16239150

RESUMEN

Rhythmic cardiac contractions depend on the organized propagation of depolarizing and repolarizing wavefronts. Repolarization is spatially heterogeneous and depends largely on gradients of potassium currents. Gradient disruption in heart disease may underlie susceptibility to fatal arrhythmias, but it is not known how this gradient is established. We show that, in mice lacking the homeodomain transcription factor Irx5, the cardiac repolarization gradient is abolished due to increased Kv4.2 potassium-channel expression in endocardial myocardium, resulting in a selective increase of the major cardiac repolarization current, I(to,f), and increased susceptibility to arrhythmias. Myocardial Irx5 is expressed in a gradient opposite that of Kv4.2, and Irx5 represses Kv4.2 expression by recruiting mBop, a cardiac transcriptional repressor. Thus, an Irx5 repressor gradient negatively regulates potassium-channel-gene expression in the heart, forming an inverse I(to,f) gradient that ensures coordinated cardiac repolarization while also preventing arrhythmias.


Asunto(s)
Potenciales de Acción/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Función Ventricular Izquierda/fisiología , Función Ventricular , Potenciales de Acción/fisiología , Animales , Western Blotting , Cruzamientos Genéticos , Perros , Electrocardiografía , Electrofisiología , Endocardio/citología , Endocardio/fisiología , Genes Reporteros , Ventrículos Cardíacos/citología , Heterocigoto , Homocigoto , Inmunohistoquímica , Luciferasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Pericardio/citología , Pericardio/fisiología , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio con Entrada de Voltaje/fisiología , Pruebas de Precipitina , Proteínas/análisis , ARN Mensajero/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA