Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Breast Cancer Res ; 21(1): 98, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462307

RESUMEN

BACKGROUND: Circulating tumor cells (CTCs) represent a temporal "snapshot" of a patient's cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer. METHODS: We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice. RESULTS: Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency. CONCLUSION: PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Células Neoplásicas Circulantes/patología , Neoplasias de la Mama Triple Negativas/patología , Animales , Biomarcadores de Tumor/metabolismo , Encéfalo/patología , Recuento de Células , Línea Celular Tumoral , Femenino , Humanos , Queratinas/metabolismo , Hígado/patología , Pulmón/patología , Ratones , Ratones Mutantes , Metástasis de la Neoplasia , Trasplante de Neoplasias , Células Neoplásicas Circulantes/metabolismo , Vimentina/metabolismo
2.
Nat Commun ; 8: 14622, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28332571

RESUMEN

Circulating tumour cells (CTCs) are rare tumour cells found in the circulatory system of certain cancer patients. The clinical and functional significance of CTCs is still under investigation. Protein profiling of CTCs would complement the recent advances in enumeration, transcriptomic and genomic characterization of these rare cells and help define their characteristics. Here we describe a microfluidic western blot for an eight-plex protein panel for individual CTCs derived from estrogen receptor-positive (ER+) breast cancer patients. The precision handling and analysis reveals a capacity to assay sparingly available patient-derived CTCs, a biophysical CTC phenotype more lysis-resistant than breast cancer cell lines, a capacity to report protein expression on a per CTC basis and two statistically distinct GAPDH subpopulations within the patient-derived CTCs. Targeted single-CTC proteomics with the capacity for archivable, multiplexed protein analysis offers a unique, complementary taxonomy for understanding CTC biology and ascertaining clinical impact.


Asunto(s)
Western Blotting/métodos , Neoplasias de la Mama/patología , Perfilación de la Expresión Génica/métodos , Microfluídica/métodos , Células Neoplásicas Circulantes/metabolismo , Adulto , Anciano , Western Blotting/instrumentación , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/análisis , Humanos , Microfluídica/instrumentación , Persona de Mediana Edad , Proyectos Piloto , Proteómica/métodos , Receptores de Estrógenos/metabolismo , Reproducibilidad de los Resultados , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos
3.
Oncotarget ; 7(51): 85349-85364, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27863403

RESUMEN

Treatment of advanced colorectal cancer (CRC) requires multimodal therapeutic approaches and need for monitoring tumor plasticity. Liquid biopsy biomarkers, including CTCs and ctDNA, hold promise for evaluating treatment response in real-time and guiding therapeutic modifications. From 15 patients with advanced CRC undergoing liver metastasectomy with curative intent, we collected 41 blood samples at different time points before and after surgery for CTC isolation and quantification using label-free Vortex technology. For mutational profiling, KRAS, BRAF, and PIK3CA hotspot mutations were analyzed in CTCs and ctDNA from 23 samples, nine matched liver metastases and three primary tumor samples. Mutational patterns were compared. 80% of patient blood samples were positive for CTCs, using a healthy baseline value as threshold (0.4 CTCs/mL), and 81.4% of captured cells were EpCAM+ CTCs. At least one mutation was detected in 78% of our blood samples. Among 23 matched CTC and ctDNA samples, we found a concordance of 78.2% for KRAS, 73.9% for BRAF and 91.3% for PIK3CA mutations. In several cases, CTCs exhibited a mutation that was not detected in ctDNA, and vice versa. Complementary assessment of both CTCs and ctDNA appears advantageous to assess dynamic tumor profiles.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/métodos , Mutación , Células Neoplásicas Circulantes/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Biomarcadores de Tumor/sangre , ADN Tumoral Circulante/sangre , Fosfatidilinositol 3-Quinasa Clase I/sangre , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN/instrumentación , Predisposición Genética a la Enfermedad , Células HCT116 , Humanos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Células Neoplásicas Circulantes/patología , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Proteínas Proto-Oncogénicas B-raf/sangre , Proteínas Proto-Oncogénicas p21(ras)/sangre , Reproducibilidad de los Resultados , Factores de Tiempo
4.
Sci Rep ; 6: 35474, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27739521

RESUMEN

Circulating tumor cells (CTCs) have a great potential as indicators of metastatic disease that may help physicians improve cancer prognostication, treatment and patient outcomes. Heterogeneous marker expression as well as the complexity of current antibody-based isolation and analysis systems highlights the need for alternative methods. In this work, we use a microfluidic Vortex device that can selectively isolate potential tumor cells from blood independent of cell surface expression. This system was adapted to interface with three protein-marker-free analysis techniques: (i) an in-flow automated image processing system to enumerate cells released, (ii) cytological analysis using Papanicolaou (Pap) staining and (iii) fluorescence in situ hybridization (FISH) targeting the ALK rearrangement. In-flow counting enables a rapid assessment of the cancer-associated large circulating cells in a sample within minutes to determine whether standard downstream assays such as cytological and cytogenetic analyses that are more time consuming and costly are warranted. Using our platform integrated with these workflows, we analyzed 32 non-small cell lung cancer (NSCLC) and 22 breast cancer patient samples, yielding 60 to 100% of the cancer patients with a cell count over the healthy threshold, depending on the detection method used: respectively 77.8% for automated, 60-100% for cytology, and 80% for immunostaining based enumeration.


Asunto(s)
Neoplasias de la Mama/sangre , Carcinoma de Pulmón de Células no Pequeñas/sangre , Separación Celular/métodos , Neoplasias Pulmonares/sangre , Microfluídica/métodos , Células Neoplásicas Circulantes/metabolismo , Quinasa de Linfoma Anaplásico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Separación Celular/instrumentación , Femenino , Humanos , Hibridación Fluorescente in Situ/métodos , Células MCF-7 , Masculino , Microfluídica/instrumentación , Células Neoplásicas Circulantes/patología , Prueba de Papanicolaou/métodos , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo
5.
Oncotarget ; 7(11): 12748-60, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26863573

RESUMEN

Circulating tumor cells (CTCs) are emerging as rare but clinically significant non-invasive cellular biomarkers for cancer patient prognosis, treatment selection, and treatment monitoring. Current CTC isolation approaches, such as immunoaffinity, filtration, or size-based techniques, are often limited by throughput, purity, large output volumes, or inability to obtain viable cells for downstream analysis. For all technologies, traditional immunofluorescent staining alone has been employed to distinguish and confirm the presence of isolated CTCs among contaminating blood cells, although cells isolated by size may express vastly different phenotypes. Consequently, CTC definitions have been non-trivial, researcher-dependent, and evolving. Here we describe a complete set of objective criteria, leveraging well-established cytomorphological features of malignancy, by which we identify large CTCs. We apply the criteria to CTCs enriched from stage IV lung and breast cancer patient blood samples using the High Throughput Vortex Chip (Vortex HT), an improved microfluidic technology for the label-free, size-based enrichment and concentration of rare cells. We achieve improved capture efficiency (up to 83%), high speed of processing (8 mL/min of 10x diluted blood, or 800 µL/min of whole blood), and high purity (avg. background of 28.8±23.6 white blood cells per mL of whole blood). We show markedly improved performance of CTC capture (84% positive test rate) in comparison to previous Vortex designs and the current FDA-approved gold standard CellSearch assay. The results demonstrate the ability to quickly collect viable and pure populations of abnormal large circulating cells unbiased by molecular characteristics, which helps uncover further heterogeneity in these cells.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Técnicas Analíticas Microfluídicas/métodos , Células Neoplásicas Circulantes/clasificación , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Células Neoplásicas Circulantes/patología
6.
Oncotarget ; 6(4): 2549-61, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25575824

RESUMEN

BACKGROUND: Circulating tumor DNA (ctDNA) holds promise as a non-invasive means for tumor monitoring in solid malignancies. Assays with high sensitivity and multiplexed analysis of mutations are needed to enable broad application. METHODS: We developed a new assay based on sequence-specific synchronous coefficient of drag alteration (SCODA) technology, which enriches for mutant DNA to achieve high sensitivity and specificity. This assay was applied to plasma and tumor tissue from non-metastatic and metastatic colorectal cancer (CRC) patients, including patients undergoing surgical resection for CRC liver metastases. RESULTS: Across multiple characterization experiments, the assay demonstrated a limit of detection of 0.001% (1 molecule in 100,000) for the majority of the 46 mutations in the panel. In CRC patient samples (n=38), detected mutations were concordant in tissue and plasma for 93% of metastatic patients versus 54% of non-metastatic patients. For three patients, ctDNA identified additional mutations not detected in tumor tissue. In patients undergoing liver metastatectomy, ctDNA anticipated tumor recurrence earlier than carcinoembryonic antigen (CEA) value or imaging. CONCLUSIONS: The multiplexed SCODA mutation enrichment and detection method can be applied to mutation profiling and quantitation of ctDNA, and is likely to have particular utility in the metastatic setting, including patients undergoing metastatectomy.


Asunto(s)
Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/métodos , ADN de Neoplasias/genética , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Fosfatidilinositol 3-Quinasa Clase I , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/cirugía , ADN de Neoplasias/sangre , Receptores ErbB/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/genética , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras) , Reproducibilidad de los Resultados , Proteínas ras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...