Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cytometry A ; 93(12): 1260-1266, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30551261

RESUMEN

Circulating tumor cells (CTCs) are rare cells shed into the bloodstream by invasive tumors and their analysis offers a promising noninvasive tool to predict and monitor therapeutic responses. CTCs can be isolated from patient blood and their characterization at single-cell level can inform on the genomic landscape of a tumor. All CTC enrichment methods bear a burden of contaminating normal cells, which mandate a further step of purification to enable reliable downstream genetic analysis. Here, we describe the DEPArray™ technology, a microchip-based digital sorter, which combines precise microfluidic and microelectronic enabling precise, image-based isolation of single CTCs, which can then be analyzed by Next Generation Sequencing (NGS) methods. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Asunto(s)
Microfluídica/métodos , Células Neoplásicas Circulantes/patología , Análisis de la Célula Individual/métodos , Recuento de Células/métodos , Humanos
2.
J Immunol ; 191(7): 3545-52, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24006464

RESUMEN

Guiding the interaction of single cells acting as partners in heterotypic interactions (e.g., effectors and targets of immune lysis) and monitoring the outcome of these interactions are regarded as crucial biomedical achievements. In this study, taking advantage of a dielectrophoresis (DEP)-based Laboratory-on-a-chip platform (the DEPArray), we show that it is possible to generate closed DEP cages entrapping CTLs and NK cells as either single cells or clusters; reversibly immobilize a single virus-presenting or tumor cell within the chip at a selected position; move cages and their content to predetermined spatial coordinates by software-guided routing; force a cytotoxic effector to physically interact with a putative target within a secluded area by merging their respective cages; generate cages containing effector and target cells at predetermined E:T ratios; accurately assess cytotoxicity by real-time quantitation of the release kinetics of the fluorescent dye calcein from target cells (>50 lytic events may be tested simultaneously); estimate end points of calcein release within 16 min of initial E:T cell contact; simultaneously deliver Ab-based phenotyping and on-chip lysis assessment; and identify lytic and nonlytic E:T combinations and discriminate nonlytic effector phenotypes from target refractoriness to immune lysis. The proof of principle is provided that DEPArray technology, previously used to levitate and move single cells, can be used to identify highly lytic antiviral CTLs and tumor cells that are particularly refractory to NK cell lysis. These findings are of primary interest in targeted immunotherapy.


Asunto(s)
Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Análisis de la Célula Individual/métodos , Linfocitos T Citotóxicos/inmunología , Comunicación Celular/inmunología , Línea Celular Transformada , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Humanos , Células Asesinas Naturales/metabolismo , Linfocitos T Citotóxicos/metabolismo
3.
Anal Chem ; 85(17): 8219-24, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23968491

RESUMEN

Manipulating single biological objects is a major unmet challenge of biomedicine. Herein, we describe a lab-on-a-chip platform based on dielectrophoresis (DEP). The DEParray is a prototypal version consisting of 320 × 320 arrayed electrodes generating >10,000 spherical DEP cages. It allows the capture and software-guided movement to predetermined spatial coordinates of single biological objects. With the DEParray we demonstrate (a) forced interaction between a single, preselected target cell and a programmable number of either microspheres or natural killer (NK) cells, (b) on-chip immunophenotypic discrimination of individual cells based on differential rosetting with microspheres functionalized with monoclonal antibodies to an inhibitory NK cell ligand (HLA-G), (c) on-chip, real-time (few minutes) assessment of immune lysis by either visual inspection or semiautomated, time-lapse reading of a fluorescent dye released from NK cell-sensitive targets, and (d) manipulation and immunophenotyping with limiting amounts (about 500) cells. To our knowledge, this is the first report describing a DEP-based lab-on-a-chip platform for the quick, arrayed, software-guided binding of individually moved biological objects, the targeting of single cells with microspheres, and the real-time characterization of immunophenotypes. The DEParray candidates as a discovery tool for novel cell:cell interactions with no prior (immuno)phenotypic knowledge.


Asunto(s)
Electroforesis por Microchip/métodos , Células Asesinas Naturales/metabolismo , Microesferas , Electroforesis por Microchip/instrumentación , Humanos , Células K562 , Unión Proteica/fisiología
4.
NPJ Precis Oncol ; 5(1): 67, 2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272470

RESUMEN

Gatekeeper mutations are identified in only 50% of the cases at resistance to Anaplastic Lymphoma Kinase (ALK)-tyrosine kinase inhibitors (TKIs). Circulating tumor cells (CTCs) are relevant tools to identify additional resistance mechanisms and can be sequenced at the single-cell level. Here, we provide in-depth investigation of copy number alteration (CNA) heterogeneity in phenotypically characterized CTCs at resistance to ALK-TKIs in ALK-positive non-small cell lung cancer. Single CTC isolation and phenotyping were performed by DEPArray or fluorescence-activated cell sorting following enrichment and immunofluorescence staining (ALK/cytokeratins/CD45/Hoechst). CNA heterogeneity was evaluated in six ALK-rearranged patients harboring ≥ 10 CTCs/20 mL blood at resistance to 1st and 3rd ALK-TKIs and one presented gatekeeper mutations. Out of 82 CTCs isolated by FACS, 30 (37%) were ALK+/cytokeratins-, 46 (56%) ALK-/cytokeratins+ and 4 (5%) ALK+/cytokeratins+. Sequencing of 43 CTCs showed highly altered CNA profiles and high levels of chromosomal instability (CIN). Half of CTCs displayed a ploidy >2n and 32% experienced whole-genome doubling. Hierarchical clustering showed significant intra-patient and wide inter-patient CTC diversity. Classification of 121 oncogenic drivers revealed the predominant activation of cell cycle and DNA repair pathways and of RTK/RAS and PI3K to a lower frequency. CTCs showed wide CNA heterogeneity and elevated CIN at resistance to ALK-TKIs. The emergence of epithelial ALK-negative CTCs may drive resistance through activation of bypass signaling pathways, while ALK-rearranged CTCs showed epithelial-to-mesenchymal transition characteristics potentially contributing to ALK-TKI resistance. Comprehensive analysis of CTCs could be of great help to clinicians for precision medicine and resistance to ALK-targeted therapies.

5.
Front Cell Dev Biol ; 9: 666156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34178989

RESUMEN

Cancers of unknown primary (CUPs) comprise a heterogeneous group of rare metastatic tumors whose primary site cannot be identified after extensive clinical-pathological investigations. CUP patients are generally treated with empirical chemotherapy and have dismal prognosis. As recently reported, CUP genome presents potentially druggable alterations for which targeted therapies could be proposed. The paucity of tumor tissue, as well as the difficult DNA testing and the lack of dedicated panels for target gene sequencing are further relevant limitations. Here, we propose that circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) could be used to identify actionable mutations in CUP patients. Blood was longitudinally collected from two CUP patients. CTCs were isolated with CELLSEARCH® and DEPArrayTM NxT and Parsortix systems, immunophenotypically characterized and used for single-cell genomic characterization with Ampli1TM kits. Circulating cell-free DNA (ccfDNA), purified from plasma at different time points, was tested for tumor mutations with a CUP-dedicated, 92-gene custom panel using SureSelect Target Enrichment technology. In parallel, FFPE tumor tissue was analyzed with three different assays: FoundationOne CDx assay, DEPArray LibPrep and OncoSeek Panel, and the SureSelect custom panel. These approaches identified the same mutations, when the gene was covered by the panel, with the exception of an insertion in APC gene. which was detected by OncoSeek and SureSelect panels but not FoundationOne. FGFR2 and CCNE1 gene amplifications were detected in single CTCs, tumor tissue, and ccfDNAs in one patient. A somatic variant in ARID1A gene (p.R1276∗) was detected in the tumor tissue and ccfDNAs. The alterations were validated by Droplet Digital PCR in all ccfDNA samples collected during tumor evolution. CTCs from a second patient presented a pattern of recurrent amplifications in ASPM and SEPT9 genes and loss of FANCC. The 92-gene custom panel identified 16 non-synonymous somatic alterations in ccfDNA, including a deletion (I1485Rfs∗19) and a somatic mutation (p. A1487V) in ARID1A gene and a point mutation in FGFR2 gene (p.G384R). Our results support the feasibility of non-invasive liquid biopsy testing in CUP cases, either using ctDNA or CTCs, to identify CUP genetic alterations with broad NGS panels covering the most frequently mutated genes.

6.
Nat Commun ; 11(1): 1884, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313004

RESUMEN

Transformation of castration-resistant prostate cancer (CRPC) into an aggressive neuroendocrine disease (CRPC-NE) represents a major clinical challenge and experimental models are lacking. A CTC-derived eXplant (CDX) and a CDX-derived cell line are established using circulating tumor cells (CTCs) obtained by diagnostic leukapheresis from a CRPC patient resistant to enzalutamide. The CDX and the derived-cell line conserve 16% of primary tumor (PT) and 56% of CTC mutations, as well as 83% of PT copy-number aberrations including clonal TMPRSS2-ERG fusion and NKX3.1 loss. Both harbor an androgen receptor-null neuroendocrine phenotype, TP53, PTEN and RB1 loss. While PTEN and RB1 loss are acquired in CTCs, evolutionary analysis suggest that a PT subclone harboring TP53 loss is the driver of the metastatic event leading to the CDX. This CDX model provides insights on the sequential acquisition of key drivers of neuroendocrine transdifferentiation and offers a unique tool for effective drug screening in CRPC-NE management.


Asunto(s)
Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/metabolismo , Transdiferenciación Celular/genética , Células Neoplásicas Circulantes/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Animales , Benzamidas , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Células Neoplásicas Circulantes/efectos de los fármacos , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Filogenia , Próstata/patología , Receptores Androgénicos/genética , Alineación de Secuencia , Serina Endopeptidasas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Proteína p53 Supresora de Tumor/genética
7.
PLoS One ; 13(3): e0193689, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29494651

RESUMEN

Chromosomal instability and associated chromosomal aberrations are hallmarks of cancer and play a critical role in disease progression and development of resistance to drugs. Single-cell genome analysis has gained interest in latest years as a source of biomarkers for targeted-therapy selection and drug resistance, and several methods have been developed to amplify the genomic DNA and to produce libraries suitable for Whole Genome Sequencing (WGS). However, most protocols require several enzymatic and cleanup steps, thus increasing the complexity and length of protocols, while robustness and speed are key factors for clinical applications. To tackle this issue, we developed a single-tube, single-step, streamlined protocol, exploiting ligation mediated PCR (LM-PCR) Whole Genome Amplification (WGA) method, for low-pass genome sequencing with the Ion Torrent™ platform and copy number alterations (CNAs) calling from single cells. The method was evaluated on single cells isolated from 6 aberrant cell lines of the NCI-H series. In addition, to demonstrate the feasibility of the workflow on clinical samples, we analyzed single circulating tumor cells (CTCs) and white blood cells (WBCs) isolated from the blood of patients affected by prostate cancer or lung adenocarcinoma. The results obtained show that the developed workflow generates data accurately representing whole genome absolute copy number profiles of single cell and allows alterations calling at resolutions down to 100 Kbp with as few as 200,000 reads. The presented data demonstrate the feasibility of the Ampli1™ WGA-based low-pass workflow for detection of CNAs in single tumor cells which would be of particular interest for genome-driven targeted therapy selection and for monitoring of disease progression.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias/genética , Análisis de la Célula Individual/métodos , Secuenciación Completa del Genoma/métodos , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Humanos , Neoplasias Pulmonares/genética , Masculino , Células Neoplásicas Circulantes/patología , Reacción en Cadena de la Polimerasa/instrumentación , Neoplasias de la Próstata/genética , Análisis de la Célula Individual/instrumentación , Secuenciación Completa del Genoma/instrumentación , Flujo de Trabajo
8.
Lab Chip ; 6(1): 121-6, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16372078

RESUMEN

Sorting and recovering specific live cells from samples containing less than a few thousand cells have become major hurdles in rare cell exploration such as stem cell research, cell therapy and cell based diagnostics. We describe here a new technology based on a microelectronic chip integrating an array of over 100,000 independent electrodes and sensors which allow individual and parallel single cell manipulation of up to 10,000 cells while maintaining viability and proliferation capabilities. Manipulation is carried out using dynamic dielectrophoretic traps controlled by an electronic interface. We also demonstrate the capabilities of the chip by sorting and recovering individual live fluorescent cells from an unlabeled population.


Asunto(s)
Separación Celular/instrumentación , Separación Celular/métodos , Electroforesis por Microchip/métodos , Proliferación Celular , Supervivencia Celular , Tamaño de la Muestra
9.
Sci Rep ; 6: 20944, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26864208

RESUMEN

Precision medicine in oncology requires an accurate characterization of a tumor molecular profile for patient stratification. Though targeted deep sequencing is an effective tool to detect the presence of somatic sequence variants, a significant number of patient specimens do not meet the requirements needed for routine clinical application. Analysis is hindered by contamination of normal cells and inherent tumor heterogeneity, compounded with challenges of dealing with minute amounts of tissue and DNA damages common in formalin-fixed paraffin-embedded (FFPE) specimens. Here we present an innovative workflow using DEPArray™ system, a microchip-based digital sorter to achieve 100%-pure, homogenous subpopulations of cells from FFPE samples. Cells are distinguished by fluorescently labeled antibodies and DNA content. The ability to address tumor heterogeneity enables unambiguous determination of true-positive sequence variants, loss-of-heterozygosity as well as copy number variants. The proposed strategy overcomes the inherent trade-offs made between sensitivity and specificity in detecting genetic variants from a mixed population, thus rescuing for analysis even the smaller clinical samples with low tumor cellularity.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis por Micromatrices/métodos , Neoplasias/diagnóstico , Separación Celular/instrumentación , Variaciones en el Número de Copia de ADN , Fijadores , Citometría de Flujo/instrumentación , Formaldehído , Variación Genética , Humanos , Análisis por Micromatrices/instrumentación , Mutación , Neoplasias/genética , Neoplasias/patología , Adhesión en Parafina , Sensibilidad y Especificidad , Análisis de Secuencia de ADN , Fijación del Tejido
10.
Int J Oncol ; 27(6): 1559-66, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16273212

RESUMEN

There is a general agreement on the fact that the Laboratory on chip (Lab-on-a-chip) technology will enable laboratory testing to move from laboratories employing complex equipments into non-laboratory settings. In this respect, dielectrophoresis (DEP) is a very valuable approach to design and produce Lab-on-a-chip devices able to manipulate microparticles and cells. In this study, we report the application of DEP-based devices for facilitating programmable interactions between microspheres and target tumor cells. We used two Lab-on-a-chip devices, one (the SmartSlide) carrying 193 parallel electrodes and generating up to 50 cylinder-shaped DEP cages, the other (the DEP array) carrying 102,400 arrayed electrodes and generating more than 10,000 spherical DEP cages. We determined whether these devices can be used to levitate and move microspheres and cells in order to obtain a forced interaction between microspheres and target cells. The first major point of this manuscript is that the DEP-based SmartSlide can be used for transfection experiments in which microspheres and target cells are forced to share the same DEP cage, leading to efficient binding of the microspheres to target cells. The data obtained using the DEP array show that this system allows the sequential, software-controlled binding of individually and independently moved single microspheres to a single target tumor cell. To our knowledge, this is the first report on the possible use of a DEP-based Lab-on-a-chip device for guided multiple binding of singularly moved microspheres to a single tumor cell. This approach can be of interest in the field of drug discovery, delivery and diagnosis.


Asunto(s)
Técnicas de Laboratorio Clínico/instrumentación , Electroforesis/instrumentación , Microesferas , Algoritmos , Unión Competitiva , Separación Celular/instrumentación , Simulación por Computador , Computadores , Diseño de Equipo/métodos , Humanos , Células K562 , Modelos Biológicos , Reproducibilidad de los Resultados
11.
Int J Mol Med ; 15(6): 913-20, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15870893

RESUMEN

The 'Lab-on-a-chip technology' involves miniaturization of complex analytical procedures and is expected to enable laboratory testing to move from the central laboratory employing complex equipment into non-laboratory settings. We report the application of a printed circuit board (PCB)-based chip, generating dielectrophoretic (DEP)-based cylinder-shaped cages for separation and recovery of white blood cells from erythrocytes. This possibility is of interest to develop low-cost Lab-on-a-chip devices for diagnostic purposes. Accordingly, we demonstrate that white blood cells recovered from this Lab-on-a-chip device are suitable for PCR-based molecular diagnosis procedures employing DNA sequencing or biospecific interaction analysis using surface plasmon resonance and biosensor technology.


Asunto(s)
Separación Celular/instrumentación , Técnicas de Laboratorio Clínico/instrumentación , Electroforesis/instrumentación , Eritrocitos/citología , Leucocitos/citología , Técnicas Biosensibles , Recuento de Células , Separación Celular/métodos , Simulación por Computador , Electroforesis/métodos , Diseño de Equipo , Humanos , Células K562 , Técnicas de Diagnóstico Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reacción en Cadena de la Polimerasa , Resonancia por Plasmón de Superficie
12.
Technol Cancer Res Treat ; 2(1): 31-40, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12625752

RESUMEN

The recent development of advanced analytical and bioseparation methodologies based on microarrays and biosensors is one of the strategic objectives of the so-called post-genomic. In this field, the development of microfabricated devices could bring new opportunities in several application fields, such as predictive oncology, diagnostics and anti-tumor drug research. The so called "Laboratory-on-a-chip technology", involving miniaturisation of analytical procedures, is expected to enable highly complex laboratory testing to move from the central laboratory into non-laboratory settings. The main advantages of Lab-on-a-chip devices are integration of multiple steps of different analytical procedures, large variety of applications, sub-microliter consumption of reagents and samples, and portability. One of the requirement for new generation Lab-on-a-chip devices is the possibility to be independent from additional preparative/analytical instruments. Ideally, Lab-on-a-chip devices should be able to perform with high efficiency and reproducibility both actuating and sensing procedures. In this review, we discuss applications of dielectrophoretic(DEP)-based Lab-on-a-chip devices to cancer research. The theory of dielectrophoresis as well as the description of several devices, based on spiral-shaped, parallel and arrayed electrodes are here presented. In addition, in this review we describe manipulation of cancer cells using advanced DEP-based Lab-on-a-chip devices in the absence of fluid flow and with the integration of both actuating and sensing procedures.


Asunto(s)
Técnicas de Laboratorio Clínico/instrumentación , Técnicas de Laboratorio Clínico/tendencias , Electroforesis/métodos , Electroforesis/tendencias , Neoplasias/diagnóstico , Biotecnología/instrumentación , Biotecnología/tendencias , Separación Celular/instrumentación , Separación Celular/métodos , Diseño de Equipo/tendencias , Perfilación de la Expresión Génica/instrumentación , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/tendencias , Humanos , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/tendencias , Células Tumorales Cultivadas
14.
EMBO Mol Med ; 6(11): 1371-86, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25358515

RESUMEN

Several hundred clinical trials currently explore the role of circulating tumor cell (CTC) analysis for therapy decisions, but assays are lacking for comprehensive molecular characterization of CTCs with diagnostic precision. We therefore combined a workflow for enrichment and isolation of pure CTCs with a non-random whole genome amplification method for single cells and applied it to 510 single CTCs and 189 leukocytes of 66 CTC-positive breast cancer patients. We defined a genome integrity index (GII) to identify single cells suited for molecular characterization by different molecular assays, such as diagnostic profiling of point mutations, gene amplifications and whole genomes of single cells. The reliability of > 90% for successful molecular analysis of high-quality clinical samples selected by the GII enabled assessing the molecular heterogeneity of single CTCs of metastatic breast cancer patients. We readily identified genomic disparity of potentially high relevance between primary tumors and CTCs. Microheterogeneity analysis among individual CTCs uncovered pre-existing cells resistant to ERBB2-targeted therapies suggesting ongoing microevolution at late-stage disease whose exploration may provide essential information for personalized treatment decisions and shed light into mechanisms of acquired drug resistance.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Genómica/métodos , Células Neoplásicas Circulantes/patología , Patología Molecular/métodos , Análisis de la Célula Individual/métodos , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA