Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Lab Invest ; 100(1): 16-26, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31292541

RESUMEN

Gastroesophageal junction (GEJ) cancer remains a clinically significant disease in Western countries due to its increasing incidence, which mirrors that of esophageal cancer, and poor prognosis. To develop novel and effective approaches for prevention, early detection, and treatment of patients with GEJ cancer, a better understanding of the mechanisms driving pathogenesis and malignant progression of this disease is required. These efforts have been limited by the small number of available cell lines and appropriate preclinical animal models for in vitro and in vivo studies. We have established and characterized a novel GEJ cancer cell line, GEAMP, derived from the malignant pleural effusion of a previously treated GEJ cancer patient. Comprehensive genetic analyses confirmed a clonal relationship between GEAMP cells and the primary tumor. Targeted next-generation sequencing identified 56 nonsynonymous alterations in 51 genes including TP53 and APC, which are commonly altered in GEJ cancer. In addition, multiple copy-number alterations were found including EGFR and K-RAS gene amplifications and loss of CDKN2A and CDKN2B. Histological examination of subcutaneous flank xenografts in nude and NOD-SCID mice showed a carcinoma with mixed squamous and glandular differentiation, suggesting GEAMP cells contain a subpopulation with multipotent potential. Finally, pharmacologic inhibition of the EGFR signaling pathway led to downregulation of key downstream kinases and inhibition of cell proliferation in vitro. Thus, GEAMP represents a valuable addition to the limited number of bona fide GEJ cancer cell lines.


Asunto(s)
Adenocarcinoma/patología , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Unión Esofagogástrica/patología , Derrame Pleural Maligno/patología , Adenocarcinoma/terapia , Animales , Receptores ErbB/antagonistas & inhibidores , Neoplasias Esofágicas/terapia , Resultado Fatal , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Derrame Pleural Maligno/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Exp Med ; 204(10): 2321-33, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17785507

RESUMEN

The host responds to lymphopenic environments by acute homeostatic proliferation, which is a cytokine- and endogenous peptide-driven expansion of lymphocytes that restores the numbers and diversity of T cells. It is unknown how these homeostatically proliferating (HP) cells are ultimately controlled. Using a system where lymphocytic choriomeningitis virus-immune C57BL/6 splenocytes were transferred into lymphopenic T cell-deficient hosts and allowed to reconstitute the environment, we defined the following three populations of T cells: slowly dividing Ly6C+ cells, which contained bona fide virus-specific memory cells, and more rapidly dividing Ly6C- cells segregating into programmed death (PD)-1+ and PD-1- fractions. The PD-1+ HP cell population, which peaked in frequency at day 21, was dysfunctional in that it failed to produce interferon gamma or tumor necrosis factor alpha on T cell receptor (TCR) stimulation, had down-regulated expression of interleukin (IL)-7Ralpha, IL-15Rbeta, and Bcl-2, and reacted with Annexin V, which is indicative of a preapoptotic state. The PD-1+ HP cells, in contrast to other HP cell fractions, displayed highly skewed TCR repertoires, which is indicative of oligoclonal expansion; these skewed repertoires and the PD-1+ population disappeared by day 70 from the host, presumably because of apoptosis. These results suggest that PD-1 may play a negative regulatory role to control rapidly proliferating and potentially pathogenic autoreactive CD8+ T cells during homeostatic reconstitution of lymphopenic environments.


Asunto(s)
Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Homeostasis/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Anexina A5/metabolismo , Antígenos Ly/metabolismo , Proliferación Celular , Citocinas/biosíntesis , Memoria Inmunológica/inmunología , Activación de Linfocitos , Masculino , Ratones , Receptor de Muerte Celular Programada 1 , Linfocitos T/citología , Factores de Tiempo
3.
Respirology ; 18(5): 757-64, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23586700

RESUMEN

The cancer stem cell (CSC) model is based on a myriad of experimental and clinical observations suggesting that the malignant phenotype is sustained by a subset of cells characterized by the capacity for self-renewal, differentiation and innate resistance to chemotherapy and radiation. CSC may be responsible for disease recurrence after definitive therapy and may therefore be functionally synonymous with minimal residual disease. Similar to other solid tumours, several putative surface markers for lung CSC have been identified, including CD133 and CD44. In addition, expression and/or activity of the cytoplasmic enzyme aldehyde dehydrogenase ALDH and capacity of cells to exclude membrane permeable dyes (known as the 'side population') correlate with stem-like function in vitro and in vivo. Embryonic stem cell pathways such as Hedgehog, Notch and WNT may also be active in lung cancers stem cells and therefore may be therapeutically targetable for maintenance therapy in patients achieving a complete response to surgery, radiotherapy or chemotherapy. This paper will review the evidence regarding the existence and function of lung CSC in the context of the experimental and clinical evidence and discuss some ongoing controversies regarding this model.


Asunto(s)
Neoplasias Pulmonares/fisiopatología , Modelos Biológicos , Recurrencia Local de Neoplasia/fisiopatología , Células Madre Neoplásicas/fisiología , Antígeno AC133 , Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Glicoproteínas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Péptidos/metabolismo , Transducción de Señal/fisiología
4.
Nat Commun ; 14(1): 7209, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37938582

RESUMEN

The metastasis-invasion cascade describes the series of steps required for a cancer cell to successfully spread from its primary tumor and ultimately grow within a secondary organ. Despite metastasis being a dynamic, multistep process, most omics studies to date have focused on comparing primary tumors to the metastatic deposits that define end-stage disease. This static approach means we lack information about the genomic and epigenomic changes that occur during the majority of tumor progression. One particularly understudied phase of tumor progression is metastatic colonization, during which cells must adapt to the new microenvironment of the secondary organ. Through temporal profiling of chromatin accessibility and gene expression in vivo, we identify dynamic changes in the epigenome that occur as osteosarcoma tumors form and grow within the lung microenvironment. Furthermore, we show through paired in vivo and in vitro CRISPR drop-out screens and pharmacological validation that the upstream transcription factors represent a class of metastasis-specific dependency genes. While current models depict lung colonization as a discrete step within the metastatic cascade, our study shows it is a defined trajectory through multiple epigenetic states, revealing new therapeutic opportunities undetectable with standard approaches.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Cromatina/genética , Osteosarcoma/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Epigenoma , Neoplasias Óseas/genética , Microambiente Tumoral
5.
Oncogene ; 42(47): 3529-3541, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37845394

RESUMEN

TP53 and RB1 loss-of-function mutations are common in osteosarcoma. During development, combined loss of TP53 and RB1 function leads to downregulation of autophagy and the aberrant formation of primary cilia, cellular organelles essential for the transmission of canonical Hedgehog (Hh) signaling. Excess cilia formation then leads to hypersensitivity to Hedgehog (Hh) ligand signaling. In mouse and human models, we now show that osteosarcomas with mutations in TP53 and RB1 exhibit enhanced ligand-dependent Hh pathway activation through Smoothened (SMO), a transmembrane signaling molecule required for activation of the canonical Hh pathway. This dependence is mediated by hypersensitivity to Hh ligand and is accompanied by impaired autophagy and increased primary cilia formation and expression of Hh ligand in vivo. Using a conditional genetic mouse model of Trp53 and Rb1 inactivation in osteoblast progenitors, we further show that deletion of Smo converts the highly malignant osteosarcoma phenotype to benign, well differentiated bone tumors. Conversely, conditional overexpression of SHH ligand, or a gain-of-function SMO mutant in committed osteoblast progenitors during development blocks terminal bone differentiation. Finally, we demonstrate that the SMO antagonist sonidegib (LDE225) induces growth arrest and terminal differentiation in vivo in osteosarcomas that express primary cilia and Hh ligand combined with mutations in TP53. These results provide a mechanistic framework for aberrant Hh signaling in osteosarcoma based on defining mutations in the tumor suppressor, TP53.


Asunto(s)
Antineoplásicos , Osteosarcoma , Humanos , Animales , Ratones , Proteínas Hedgehog/metabolismo , Ligandos , Transducción de Señal , Antineoplásicos/farmacología , Osteosarcoma/genética , Osteosarcoma/metabolismo , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Cilios/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
6.
Sci Adv ; 8(50): eabp8674, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36516249

RESUMEN

Studies to date have not resolved how diverse transcriptional programs contribute to the intratumoral heterogeneity of small cell lung carcinoma (SCLC), an aggressive tumor associated with a dismal prognosis. Here, we identify distinct and commutable transcriptional states that confer discrete functional attributes in individual SCLC tumors. We combine an integrative approach comprising the transcriptomes of 52,975 single cells, high-resolution measurement of cell state dynamics at the single-cell level, and functional and correlative studies using treatment naïve xenografts with associated clinical outcomes. We show that individual SCLC tumors contain distinctive proportions of stable cellular states that are governed by bidirectional cell state transitions. Using drugs that target the epigenome, we reconfigure tumor state composition in part by altering individual state transition rates. Our results reveal new insights into how single-cell transition behaviors promote cell state equilibrium in SCLC and suggest that facile plasticity underlies its resistance to therapy and lethality.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Pronóstico
7.
Nat Commun ; 10(1): 5143, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31723142

RESUMEN

Molecular determinants governing the evolution of tumor subclones toward phylogenetic branches or fixation remain unknown. Using sequencing data, we model the propagation and selection of clones expressing distinct categories of BRAF mutations to estimate their evolutionary trajectories. We show that strongly activating BRAF mutations demonstrate hard sweep dynamics, whereas mutations with less pronounced activation of the BRAF signaling pathway confer soft sweeps or are subclonal. We use clonal reconstructions to estimate the strength of "driver" selection in individual tumors. Using tumors cells and human-derived murine xenografts, we show that tumor sweep dynamics can significantly affect responses to targeted inhibitors of BRAF/MEK or DNA damaging agents. Our study uncovers patterns of distinct BRAF clonal evolutionary dynamics and nominates therapeutic strategies based on the identity of the BRAF mutation and its clonal composition.


Asunto(s)
Evolución Clonal/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas B-raf/genética , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Clonales , Daño del ADN , Dosificación de Gen , Sitios Genéticos , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación/genética , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología
8.
NPJ Precis Oncol ; 2: 14, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30202792

RESUMEN

There has been little progress in the use of patient-derived xenografts (PDX) to guide individual therapeutic strategies. In part, this can be attributed to the operational challenges of effecting successful engraftment and testing multiple candidate drugs in a clinically workable timeframe. It also remains unclear whether the ancestral tumor will evolve along similar evolutionary trajectories in its human and rodent hosts in response to similar selective pressures (i.e., drugs). Herein, we combine a metastatic clear cell adenocarcinoma PDX with a timely 3 mouse x 1 drug experimental design, followed by a co-clinical trial to longitudinally guide a patient's care. Using this approach, we accurately predict response to first- and second-line therapies in so far as tumor response in mice correlated with the patient's clinical response to first-line therapy (gemcitabine/nivolumab), development of resistance and response to second-line therapy (paclitaxel/neratinib) before these events were observed in the patient. Treatment resistance to first-line therapy in the PDX is coincident with biologically relevant changes in gene and gene set expression, including upregulation of phase I/II drug metabolism (CYP2C18, UGT2A, and ATP2A1) and DNA interstrand cross-link repair (i.e., XPA, FANCE, FANCG, and FANCL) genes. A total of 5.3% of our engrafted PDX collection is established within 2 weeks of implantation, suggesting our experimental designs can be broadened to other cancers. These findings could have significant implications for PDX-based avatars of aggressive human cancers.

9.
J Thorac Oncol ; 12(3): 510-519, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28017592

RESUMEN

PURPOSE: Stereotactic body radiation therapy (SBRT) is the standard of care for medically inoperable patients with early-stage NSCLC. However, NSCLC is composed of several histological subtypes and the impact of this heterogeneity on SBRT treatments has yet to be established. METHODS: We analyzed 740 patients with early-stage NSCLC treated definitively with SBRT from 2003 through 2015. We calculated cumulative incidence curves using the competing risk method and identified predictors of local failure using Fine and Gray regression. RESULTS: Overall, 72 patients had a local failure, with a cumulative incidence of local failure at 3 years of 11.8%. On univariate analysis, squamous histological subtype, younger age, fewer medical comorbidities, higher body mass index, higher positron emission tomography standardized uptake value, central tumors, and lower radiation dose were associated with an increased risk for local failure. On multivariable analysis, squamous histological subtype (hazard ratio = 2.4 p = 0.008) was the strongest predictor of local failure. Patients with squamous cancers fail SBRT at a significantly higher rate than do those with adenocarcinomas or NSCLC not otherwise specified, with 3-year cumulative rates of local failure of 18.9% (95% confidence interval [CI]: 12.7-25.1), 8.7% (95% CI: 4.6-12.8), and 4.1% (95% CI: 0-9.6), respectively. CONCLUSION: Our results demonstrate an increased rate of local failure in patients with squamous cell carcinoma. Standard approaches for radiotherapy that demonstrate efficacy for a population may not achieve optimal results for individual patients. Establishing the differential dose effect of SBRT across histological groups is likely to improve efficacy and inform ongoing and future studies that aim to expand indications for SBRT.


Asunto(s)
Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/patología , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/patología , Radiocirugia , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/cirugía , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Carcinoma de Células Escamosas/diagnóstico por imagen , Carcinoma de Células Escamosas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Procesamiento de Imagen Asistido por Computador , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/cirugía , Masculino , Recurrencia Local de Neoplasia/diagnóstico por imagen , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Tomografía de Emisión de Positrones , Pronóstico , Tasa de Supervivencia , Tomografía Computarizada por Rayos X
10.
Nat Commun ; 7: 11428, 2016 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-27109210

RESUMEN

Radiotherapy is not currently informed by the genetic composition of an individual patient's tumour. To identify genetic features regulating survival after DNA damage, here we conduct large-scale profiling of cellular survival after exposure to radiation in a diverse collection of 533 genetically annotated human tumour cell lines. We show that sensitivity to radiation is characterized by significant variation across and within lineages. We combine results from our platform with genomic features to identify parameters that predict radiation sensitivity. We identify somatic copy number alterations, gene mutations and the basal expression of individual genes and gene sets that correlate with the radiation survival, revealing new insights into the genetic basis of tumour cellular response to DNA damage. These results demonstrate the diversity of tumour cellular response to ionizing radiation and establish multiple lines of evidence that new genetic features regulating cellular response after DNA damage can be identified.


Asunto(s)
Daño del ADN , Neoplasias/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Variaciones en el Número de Copia de ADN/efectos de la radiación , Daño del ADN/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias/metabolismo , Neoplasias/fisiopatología , Neoplasias/radioterapia , Tolerancia a Radiación , Radiación Ionizante
11.
Sci Transl Med ; 7(291): 291ra96, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26062848

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations resulting in constitutive kinase activity are common in acute myeloid leukemia (AML) and carry a poor prognosis. Several agents targeting FLT3 have been developed, but their limited clinical activity suggests that the inhibition of other factors contributing to the malignant phenotype is required. We examined gene expression data sets as well as primary specimens and found that the expression of GLI2, a major effector of the Hedgehog (Hh) signaling pathway, was increased in FLT3-ITD compared to wild-type FLT3 AML. To examine the functional role of the Hh pathway, we studied mice in which Flt3-ITD expression results in an indolent myeloproliferative state and found that constitutive Hh signaling accelerated the development of AML by enhancing signal transducer and activator of transcription 5 (STAT5) signaling and the proliferation of bone marrow myeloid progenitors. Furthermore, combined FLT3 and Hh pathway inhibition limited leukemic growth in vitro and in vivo, and this approach may serve as a therapeutic strategy for FLT3-ITD AML.


Asunto(s)
Proteínas Hedgehog/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Mutantes/metabolismo , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Compartimento Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Progresión de la Enfermedad , Sinergismo Farmacológico , Duplicación de Gen/efectos de los fármacos , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Trastornos Mieloproliferativos/patología , Niacinamida/análogos & derivados , Niacinamida/farmacología , Proteínas Nucleares/metabolismo , Compuestos de Fenilurea/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Smoothened , Sorafenib , Células Madre/citología , Alcaloides de Veratrum/farmacología , Proteína Gli2 con Dedos de Zinc
12.
Microbes Infect ; 4(15): 1481-90, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12505520

RESUMEN

Viral infections induce first a loss and then an increase in natural killer (NK) and CD8(+) T cells. NK cells expressing Ly49G2 were selectively expanded by several viruses and poly I:C. CD8(+) T cells expressing Ly49G2 were selectively expanded by poly I:C and participated in the antigen-specific response to lymphocytic choriomeningitis virus.


Asunto(s)
Antígenos Ly/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Virosis/inmunología , Animales , Regulación de la Expresión Génica , Lectinas Tipo C , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores Similares a Lectina de Células NK
13.
PLoS One ; 9(9): e106862, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25191746

RESUMEN

Patient-derived xenograft (PDX) models generated from surgical specimens are gaining popularity as preclinical models of cancer. However, establishment of PDX lines from small cell lung cancer (SCLC) patients is difficult due to very limited amount of available biopsy material. We asked whether SCLC cells obtained from endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) could generate PDX lines that maintained the phenotypic and genetic characteristics of the primary tumor. Following successful EBUS-TBNA sampling for diagnostic purposes, we obtained an extra sample for cytologic analysis and implantation into the flanks of immunodeficient mice. Animals were monitored for engraftment for up to 6 months. Histopathologic and immunohistochemical analysis, and targeted next-generation re-sequencing, were then performed in both the primary sample and the derivative PDX line. A total of 12 patients were enrolled in the study. EBUS-TBNA aspirates yielded large numbers of viable tumor cells sufficient to inject between 18,750 and 1,487,000 cells per flank, and to yield microgram quantities of high-quality DNA. Of these, samples from 10 patients generated xenografts (engraftment rate 83%) with a mean latency of 104 days (range 63-188). All but one maintained a typical SCLC phenotype that closely matched the original sample. Identical mutations that are characteristic of SCLC were identified in both the primary sample and xenograft line. EBUS-TBNA has the potential to be a powerful tool in the development of new targeting strategies for SCLC patients by providing large numbers of viable tumor cells suitable for both xenografting and complex genomic analysis.


Asunto(s)
Broncoscopía/métodos , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico/métodos , Endosonografía/métodos , Genómica/métodos , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Anciano , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Neoplasias Pulmonares/genética , Masculino , Ratones , Persona de Mediana Edad , Trasplante de Neoplasias , Análisis de Secuencia de ADN/métodos , Carcinoma Pulmonar de Células Pequeñas/genética , Células Tumorales Cultivadas
14.
J Natl Cancer Inst ; 105(14): 1059-65, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23739064

RESUMEN

We assessed the efficacy of Seneca Valley virus (SVV-001), a neuroendocrine cancer-selective oncolytic picornavirus, in primary heterotransplant mouse models of small cell lung cancer (SCLC), including three lines each of classic and variant SCLC. Half-maximal effective concentrations for cell lines derived from three variant heterotransplants ranged from 1.6×10(-3) (95% confidence interval [CI] = 1×10(-3) to 2.5×10(-3)) to 3.9×10(-3) (95% CI = 2.8×10(-3) to 5.5×10(-3)). Sustained tumor growth inhibition in vivo was only observed in variant lines (two-sided Student t test, P < .005 for each). Doses of 10(14) vp/kg were able to completely and durably eradicate tumors in a variant SCLC heterotransplant model in two of six mice. Gene expression profiling revealed that permissive lines are typified by lower expression of the early neurogenic transcription factor ASCL1 and, conversely, by higher expression of the late neurogenic transcription factor NEUROD1. This classifier demonstrates a sensitivity of .89, specificity of .92, and accuracy of .91. The NEUROD1 to ASCL1 ratio may serve as a predictive biomarker of SVV-001 efficacy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Carcinoma de Células Pequeñas/terapia , Neoplasias Pulmonares/terapia , Virus Oncolíticos , Picornaviridae , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células Pequeñas/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Neoplasias Pulmonares/química , Ratones , Ratones Endogámicos , Ratones SCID , Valor Predictivo de las Pruebas , Análisis por Matrices de Proteínas , Trasplante Heterólogo , Tropismo
15.
PLoS One ; 8(9): e74432, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086345

RESUMEN

Next-generation sequencing (NGS) studies in cancer are limited by the amount, quality and purity of tissue samples. In this situation, primary xenografts have proven useful preclinical models. However, the presence of mouse-derived stromal cells represents a technical challenge to their use in NGS studies. We examined this problem in an established primary xenograft model of small cell lung cancer (SCLC), a malignancy often diagnosed from small biopsy or needle aspirate samples. Using an in silico strategy that assign reads according to species-of-origin, we prospectively compared NGS data from primary xenograft models with matched cell lines and with published datasets. We show here that low-coverage whole-genome analysis demonstrated remarkable concordance between published genome data and internal controls, despite the presence of mouse genomic DNA. Exome capture sequencing revealed that this enrichment procedure was highly species-specific, with less than 4% of reads aligning to the mouse genome. Human-specific expression profiling with RNA-Seq replicated array-based gene expression experiments, whereas mouse-specific transcript profiles correlated with published datasets from human cancer stroma. We conclude that primary xenografts represent a useful platform for complex NGS analysis in cancer research for tumours with limited sample resources, or those with prominent stromal cell populations.


Asunto(s)
Modelos Animales de Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN/genética , Exoma/genética , Perfilación de la Expresión Génica , Genoma Humano/genética , Humanos , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de la Especie
16.
Cancer Biol Ther ; 12(7): 617-28, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21750403

RESUMEN

PURPOSE: To investigate the frequency of xenotropic murine leukemia virus (MLV) presence in human cell lines established from mouse xenografts and to search for the evidence of horizontal viral spread to other cell lines. RESULTS: Six of 23 (26%) mouse DNA free xenograft cultures were strongly positive for MLV and their sequences had greater than 99% homology to known MLV strains. Four of five available supernatant fluids from these viral positive cultures were strongly positive for RT activity. Three of these supernatant fluids were studied to confirm the infectivity of the released virions for other human culture cells. Of the 78 non-xenograft derived cell lines maintained in the xenograft culture-containing facilities, 13 (17%) were positive for MLV, including XMRV, a virus strain first identified in human tissues. By contrast, all 50 cultures maintained in a xenograft culture-free facility were negative for viral sequences. METHODOLOGY: We examined xenograft tumor cell lines from seven independent laboratories and 128 non-xenografted tumor cell lines. Cell line DNA was examined for mouse DNA contamination, and by 3 Taqman qPCR assays targeting the gag, env or pol regions of MLV. Sequencing was used for viral strain identification. Supernatant fluids were tested for reverse transcriptase (RT) activity. CONCLUSIONS: Human cultures derived after mouse xenografting frequently contain and release highly infectious xenotropic MLV viruses. Laboratories working with xenograft-derived human cultures should be aware of the risk of contamination with potentially biohazardous human-tropic mouse viruses and their horizontal spread to other cultures.


Asunto(s)
Línea Celular Tumoral/virología , Virus de la Leucemia Murina/aislamiento & purificación , Animales , ADN/análisis , Transmisión de Enfermedad Infecciosa , Genes env , Genes gag , Humanos , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Ratones/genética , Infecciones por Retroviridae/transmisión , Trasplante Heterólogo , Virión
17.
Nat Med ; 17(11): 1504-8, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983857

RESUMEN

Small-cell lung cancer (SCLC) is an aggressive neuroendocrine subtype of lung cancer for which there is no effective treatment. Using a mouse model in which deletion of Rb1 and Trp53 in the lung epithelium of adult mice induces SCLC, we found that the Hedgehog signaling pathway is activated in SCLC cells independently of the lung microenvironment. Constitutive activation of the Hedgehog signaling molecule Smoothened (Smo) promoted the clonogenicity of human SCLC in vitro and the initiation and progression of mouse SCLC in vivo. Reciprocally, deletion of Smo in Rb1 and Trp53-mutant lung epithelial cells strongly suppressed SCLC initiation and progression in mice. Furthermore, pharmacological blockade of Hedgehog signaling inhibited the growth of mouse and human SCLC, most notably following chemotherapy. These findings show a crucial cell-intrinsic role for Hedgehog signaling in the development and maintenance of SCLC and identify Hedgehog pathway inhibition as a therapeutic strategy to slow the progression of disease and delay cancer recurrence in individuals with SCLC.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neoplasias Pulmonares/metabolismo , Transducción de Señal/fisiología , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Animales , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Células Epiteliales/citología , Células Epiteliales/fisiología , Proteínas Hedgehog/genética , Humanos , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
Cancer Prev Res (Phila) ; 3(10): 1213-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20858762

RESUMEN

This perspective places the report by Villani et al. that appears in this issue of the journal (beginning on page 1222) in the context of recent work showing an intersection between two important developmental pathways implicated in oncogenesis: the hedgehog and insulin-like growth factor (IGF) pathways. Villani et al. define a key role for the IGF regulatory protein Igfbp2 in a genetic model of basal cell carcinogenesis driven by targeted constitutive activation of hedgehog signaling. Placed in the framework of other recently published work, the observations of Villani et al. both raise questions about the cell of origin for basal cell cancers and define additional putative therapeutic and preventive targets for this disease.


Asunto(s)
Carcinoma Basocelular/genética , Transformación Celular Neoplásica/genética , Proteínas Hedgehog/genética , Factor I del Crecimiento Similar a la Insulina/genética , Transducción de Señal/fisiología , Neoplasias Cutáneas/genética , Animales , Carcinoma Basocelular/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Cutáneas/metabolismo
20.
Cancer Res ; 69(8): 3364-73, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351829

RESUMEN

Traditional approaches to the preclinical investigation of cancer therapies rely on the use of established cell lines maintained in serum-based growth media. This is particularly true of small-cell lung cancer (SCLC), where surgically resected tissue is rarely available. Recent attention has focused on the need for better models that preserve the integrity of cancer stem cell populations, as well as three-dimensional tumor-stromal interactions. Here we describe a primary xenograft model of SCLC in which endobronchial tumor specimens obtained from chemo-naive patients are serially propagated in vivo in immunodeficient mice. In parallel, cell lines grown in conventional tissue culture conditions were derived from each xenograft line, passaged for 6 months, and then reimplanted to generate secondary xenografts. Using the Affymetrix platform, we analyzed gene expression in primary xenograft, xenograft-derived cell line, and secondary xenograft, and compared these data to similar analyses of unrelated primary SCLC samples and laboratory models. When compared with normal lung, primary tumors, xenografts, and cell lines displayed a gene expression signature specific for SCLC. Comparison of gene expression within the xenograft model identified a group of tumor-specific genes expressed in primary SCLC and xenografts that was lost during the transition to tissue culture and that was not regained when the tumors were reestablished as secondary xenografts. Such changes in gene expression may be a common feature of many cancer cell culture systems, with functional implications for the use of such models for preclinical drug development.


Asunto(s)
Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Animales , Carcinoma de Células Pequeñas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Reproducibilidad de los Resultados , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA