Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(28): 16292-16301, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601208

RESUMEN

Notch pathway signaling is implicated in several human cancers. Aberrant activation and mutations of Notch signaling components are linked to tumor initiation, maintenance, and resistance to cancer therapy. Several strategies, such as monoclonal antibodies against Notch ligands and receptors, as well as small-molecule γ-secretase inhibitors (GSIs), have been developed to interfere with Notch receptor activation at proximal points in the pathway. However, the use of drug-like small molecules to target the downstream mediators of Notch signaling, the Notch transcription activation complex, remains largely unexplored. Here, we report the discovery of an orally active small-molecule inhibitor (termed CB-103) of the Notch transcription activation complex. We show that CB-103 inhibits Notch signaling in primary human T cell acute lymphoblastic leukemia and other Notch-dependent human tumor cell lines, and concomitantly induces cell cycle arrest and apoptosis, thereby impairing proliferation, including in GSI-resistant human tumor cell lines with chromosomal translocations and rearrangements in Notch genes. CB-103 produces Notch loss-of-function phenotypes in flies and mice and inhibits the growth of human breast cancer and leukemia xenografts, notably without causing the dose-limiting intestinal toxicity associated with other Notch inhibitors. Thus, we describe a pharmacological strategy that interferes with Notch signaling by disrupting the Notch transcription complex and shows therapeutic potential for treating Notch-driven cancers.


Asunto(s)
Receptores Notch/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Activación Transcripcional/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Drosophila , Resistencia a Antineoplásicos/efectos de los fármacos , Células HeLa , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/química , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Ratones , Mutación , Fenotipo , Multimerización de Proteína , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico
2.
Methods Mol Biol ; 2650: 227-233, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37310635

RESUMEN

The intestinal epithelium is a rapid self-renewing tissue. Stem cells at the bottom of the crypts first give rise to a proliferative progeny that finally differentiates to a variety of cell types. These terminally differentiated intestinal cells are mostly present in the villi of the intestinal wall and serve as functional units to sustain the main purpose of the organ: food absorption. But for a balance homeostasis, the intestine is composed not only by absorptive enterocytes but also by other cell types such as goblet cells that secrete mucus to lubricate the intestinal lumen, Paneth cells that secrete antimicrobial peptides to control microbiome, and others. Many relevant conditions affecting the intestine including chronic inflammation, Crohn's disease, or cancer can alter the composition of these different functional cell types. As a consequence, they can lose their specialized activity as functional units and further contribute to disease progression and malignancy. Measuring the amount of these different cell populations in the intestine is essential to understand the bases of these diseases and their specific contribution to their malignancy. Interestingly, patient-derived xenograft (PDX) models faithfully recapitulate patients' tumors including the proportion of the different cell lineages present in the original tumor. Here we expose some protocols for evaluating the differentiation of intestinal cells in colorectal tumors.


Asunto(s)
Neoplasias Colorrectales , Mucosa Intestinal , Humanos , Animales , Diferenciación Celular , Linaje de la Célula , Péptidos Antimicrobianos , Modelos Animales de Enfermedad
3.
J Vis Exp ; (192)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36847362

RESUMEN

Over the last decade, more sophisticated preclinical colorectal cancer (CRC) models have been established using patient-derived cancer cells and 3D tumoroids. Since patient derived tumor organoids can retain the characteristics of the original tumor, these reliable preclinical models enable cancer drug screening and the study of drug resistance mechanisms. However, CRC related death in patients is mostly associated with the presence of metastatic disease. It is therefore essential to evaluate the efficacy of anti-cancer therapies in relevant in vivo models that truly recapitulate the key molecular features of human cancer metastasis. We have established an orthotopic model based on the injection of CRC patient-derived cancer cells directly into the cecum wall of mice. These tumor cells develop primary tumors in the cecum that metastasize to the liver and lungs, which is frequently observed in patients with advanced CRC. This CRC mouse model can be used to evaluate drug responses monitored by microcomputed tomography (µCT), a clinically relevant small-scale imaging method that can easily identify primary tumors or metastases in patients. Here, we describe the surgical procedure and the required methodology to implant patient-derived cancer cells in the cecum wall of immunodeficient mice.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Humanos , Ratones , Animales , Microtomografía por Rayos X , Neoplasias Colorrectales/patología , Ciego/patología , Implantación del Embrión , Modelos Animales de Enfermedad
4.
Cell Rep ; 42(8): 112927, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37537841

RESUMEN

Tumor relapse is linked to rapid chemoresistance and represents a bottleneck for cancer therapy success. Engagement of a reduced proliferation state is a non-mutational mechanism exploited by cancer cells to bypass therapy-induced cell death. Through combining functional pulse-chase experiments in engineered cells and transcriptomic analyses, we identify DPPA3 as a master regulator of slow-cycling and chemoresistant phenotype in colorectal cancer (CRC). We find a vicious DPPA3-HIF1α feedback loop that downregulates FOXM1 expression via DNA methylation, thereby delaying cell-cycle progression. Moreover, downregulation of HIF1α partially restores a chemosensitive proliferative phenotype in DPPA3-overexpressing cancer cells. In cohorts of CRC patient samples, DPPA3 overexpression acts as a predictive biomarker of chemotherapeutic resistance that subsequently requires reduction in its expression to allow metastatic outgrowth. Our work demonstrates that slow-cycling cancer cells exploit a DPPA3/HIF1α axis to support tumor persistence under therapeutic stress and provides insights on the molecular regulation of disease progression.

5.
Methods Mol Biol ; 2535: 85-92, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35867224

RESUMEN

Dormant or slow-cycling tumor cells can form a residual chemoresistant reservoir responsible for relapse in patients, years after curative surgery and adjuvant therapy. Slow-cycling cancer cells (SCCC) represent a cellular status rather than a cell population present in a minor proportion, even in growing tumors. We have adapted the pulse-chase expression of histone H2B fused to enhanced GFP (H2BeGFP) for labelling and isolating SCCC. SCCC show cancer-initiation potential and enhanced chemoresistance, and present a distinctive nongenetic and cell-autonomous gene expression profile shared across different tumor types. The use of our H2BeGFP pulse-chase method opens the possibility to study live SCCC in any growing tissue either cancerous or normal.


Asunto(s)
Histonas , Neoplasias , Proteínas Fluorescentes Verdes/metabolismo , Histonas/genética , Humanos , Neoplasias/genética
6.
Cell Rep ; 41(3): 111430, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36261015

RESUMEN

Despite the revolution of immunotherapy in cancer treatment, patients eventually progress due to the emergence of resistance. In this scenario, the selection of the tumor antigen can be decisive in the success of the clinical response. T cell bispecific antibodies (TCBs) are engineered molecules that include binding sites to the T cell receptor and to a tumor antigen. Using gastric CEA+/HER2+ MKN45 cells and TCBs directed against CEA or HER2, we show that the mechanism of resistance to a TCB is dependent on the tumor antigen. Acquired resistant models to a high-affinity-CEA-targeted TCB exhibit a reduction of CEA levels due to transcriptional silencing, which is reversible upon 5-AZA treatment. In contrast, a HER2-TCB resistant model maintains HER2 levels and exhibit a disruption of the interferon-gamma signaling. These results will help in the design of combinatorial strategies to increase the efficacy of cancer immunotherapies and to anticipate and overcome resistances.


Asunto(s)
Anticuerpos Biespecíficos , Humanos , Anticuerpos Biespecíficos/uso terapéutico , Antígeno Carcinoembrionario , Interferón gamma/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T , Línea Celular Tumoral
7.
Nat Cancer ; 3(4): 418-436, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35469014

RESUMEN

Patient-derived organoids (PDOs) recapitulate tumor architecture, contain cancer stem cells and have predictive value supporting personalized medicine. Here we describe a large-scale functional screen of dual-targeting bispecific antibodies (bAbs) on a heterogeneous colorectal cancer PDO biobank and paired healthy colonic mucosa samples. More than 500 therapeutic bAbs generated against Wingless-related integration site (WNT) and receptor tyrosine kinase (RTK) targets were functionally evaluated by high-content imaging to capture the complexity of PDO responses. Our drug discovery strategy resulted in the generation of MCLA-158, a bAb that specifically triggers epidermal growth factor receptor degradation in leucine-rich repeat-containing G-protein-coupled receptor 5-positive (LGR5+) cancer stem cells but shows minimal toxicity toward healthy LGR5+ colon stem cells. MCLA-158 exhibits therapeutic properties such as growth inhibition of KRAS-mutant colorectal cancers, blockade of metastasis initiation and suppression of tumor outgrowth in preclinical models for several epithelial cancer types.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias Glandulares y Epiteliales , Anticuerpos Biespecíficos/farmacología , Receptores ErbB/metabolismo , Humanos , Imidazoles , Neoplasias Glandulares y Epiteliales/metabolismo , Células Madre Neoplásicas/metabolismo , Organoides , Pirazinas , Receptores Acoplados a Proteínas G/metabolismo
8.
Nat Med ; 10(9): 917-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322538

RESUMEN

Several non-hypercalcemic analogs of 1alpha,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) show antitumor activity in a subset of cancer patients. High vitamin D receptor (VDR) expression, which is associated with good prognosis but is lost during tumor progression. We show that the SNAIL transcription factor represses VDR gene expression in human colon cancer cells and blocks the antitumor action of EB1089, a 1,25(OH)(2)D(3) analog, in xenografted mice. In human colon cancers, elevated SNAIL expression correlates with downregulation of VDR.


Asunto(s)
Calcitriol/análogos & derivados , Calcitriol/antagonistas & inhibidores , Neoplasias del Colon/metabolismo , Proteínas de Unión al ADN/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores de Calcitriol/metabolismo , Factores de Transcripción/farmacología , Animales , Antineoplásicos/antagonistas & inhibidores , Cadherinas/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Ratones , Regiones Promotoras Genéticas/genética , Receptores de Calcitriol/genética , Factores de Transcripción de la Familia Snail
9.
Cancer Res ; 81(2): 464-475, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33203702

RESUMEN

Aberrant Wnt signaling drives a number of cancers through regulation of diverse downstream pathways. Wnt/ß-catenin signaling achieves this in part by increasing the expression of proto-oncogenes such as MYC and cyclins. However, global assessment of the Wnt-regulated transcriptome in vivo in genetically distinct cancers demonstrates that Wnt signaling suppresses the expression of as many genes as it activates. In this study, we examined the set of genes that are upregulated upon inhibition of Wnt signaling in Wnt-addicted pancreatic and colorectal cancer models. Decreasing Wnt signaling led to a marked increase in gene expression by activating ERK and JNK; these changes in gene expression could be mitigated in part by concurrent inhibition of MEK. These findings demonstrate that increased Wnt signaling in cancer represses MAPK activity, preventing RAS-mediated senescence while allowing cancer cells to proliferate. These results shift the paradigm from Wnt/ß-catenin primarily as an activator of transcription to a more nuanced view where Wnt/ß-catenin signaling drives both widespread gene repression and activation. SIGNIFICANCE: These findings show that Wnt/ß-catenin signaling causes widespread gene repression via inhibition of MAPK signaling, thus fine tuning the RAS-MAPK pathway to optimize proliferation in cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Pancreáticas/patología , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Proliferación Celular , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Quinasas Activadas por Mitógenos/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Células Tumorales Cultivadas , Proteína Wnt1/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
10.
Methods Mol Biol ; 2171: 321-329, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32705653

RESUMEN

In the recent years has being a great expansion of new preclinical models of colorectal cancer (CRC) based on patient-derived cells, from ex vivo 2D cell lines, toward 3D tumoroids or animal xenografts. These new technologies have been key to overcome historical limitations in CRC research such as precision medicine, pharmacogenomic screenings, or investigating mechanism of drug resistance. Here we describe a method to generate metastatic CRC in mice with patient-derived cells and the evaluation of drug response with computerized tomography. CRC at this advanced stage is the most frequent situation in patients enrolled in therapies with novel drugs that in some cases are designed to target metastatic cells. Therefore, these orthotopic models could be considered the best to recapitulate advance CRC and are therefore becoming instrumental to investigate the biology behind drug-response in metastatic disease.


Asunto(s)
Neoplasias Colorrectales/patología , Investigación con Células Madre , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cancer Ther ; 19(1): 312-317, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31540966

RESUMEN

Several biomarkers have been suggested to have prognostic value in differentiated thyroid carcinomas (DTC) with no validation in the refractory setting, including all tumor subtypes. We aim to correlate RNA expression profiles with survival based on patients included in the DECISION trial. We obtained 247 samples from the 417 patients included in the DECISION study and performed RNAseq analysis (77 million paired-end reads for each sample on HiSeq2000). After quality control, 125 samples were included in the secondary analysis and mapped against the human reference genome (GRCh38) with STAR (v2.5.1b) using ENCODE parameter. Survival analysis was calculated using the Kaplan-Meier method and log-rank test was used for statistical comparison. In this post hoc analysis, we identified three groups of tumors based on their gene expression profile: BRAF-like, RAS-like, and non-BRAF-non-RAS-like (NoBRaL). No significant correlation with sorafenib responders was observed. However, we identified a statistically significant correlation between the RNA-expression profiles and progression-free survival. The BRAF-like profile had a significantly better outcome compared with RAS-like and NoBRaL (11.8, 6.2, and 5.5 months, respectively) [HR: 0.31, 95% confidence interval (CI), 0.17-0.60; P < 0.001 and HR: 0.36 (95% CI, 0.21-0.63); P < 0.001] and HR: 0.36 (95% CI, 0.21-0.63; P < 0.001) and maintained significance as an independent prognostic factor for overall survival in the multivariate analysis for papillary thyroid cancers. To our knowledge, this is the first comprehensive RNA-seq analysis of all histologic subtypes of DTC. The RNA expression profiles identified may suggest a new prognostic parameter to be considered before recommendation of systemic therapies or the design of stratification factors for future clinical trials.


Asunto(s)
Neoplasias de la Tiroides/radioterapia , Línea Celular Tumoral , Femenino , Humanos , Radioisótopos de Yodo/uso terapéutico , Masculino , Pronóstico , Transcriptoma , Resultado del Tratamiento
12.
Clin Cancer Res ; 26(15): 4107-4119, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32299813

RESUMEN

PURPOSE: Molecular tumor heterogeneity may have important implications for the efficacy of targeted therapies in metastatic cancers. Inter-metastatic heterogeneity of sensitivity to anticancer agents has not been well explored in colorectal cancer. EXPERIMENTAL DESIGN: We established a platform for ex vivo pharmacogenomic profiling of patient-derived organoids (PDO) from resected colorectal cancer liver metastases. Drug sensitivity testing (n = 40 clinically relevant agents) and gene expression profiling were performed on 39 metastases from 22 patients. RESULTS: Three drug-response clusters were identified among the colorectal cancer metastases, based primarily on sensitivities to EGFR and/or MDM2 inhibition, and corresponding with RAS mutations and TP53 activity. Potentially effective therapies, including off-label use of drugs approved for other cancer types, could be nominated for eighteen patients (82%). Antimetabolites and targeted agents lacking a decisive genomic marker had stronger differential activity than most approved chemotherapies. We found limited intra-patient drug sensitivity heterogeneity between PDOs from multiple (2-5) liver metastases from each of ten patients. This was recapitulated at the gene expression level, with a highly proportional degree of transcriptomic and pharmacological variation. One PDO with a multi-drug resistance profile, including resistance to EGFR inhibition in a RAS-mutant background, showed sensitivity to MEK plus mTOR/AKT inhibition, corresponding with low-level PTEN expression. CONCLUSIONS: Intra-patient inter-metastatic pharmacological heterogeneity was not pronounced and ex vivo drug screening may identify novel treatment options for metastatic colorectal cancer. Variation in drug sensitivities was reflected at the transcriptomic level, suggesting potential to develop gene expression-based predictive signatures to guide experimental therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/terapia , Resistencia a Antineoplásicos/genética , Neoplasias Hepáticas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Variación Biológica Individual , Quimioterapia Adyuvante , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Hepatectomía , Humanos , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Organoides , Variantes Farmacogenómicas , Medicina de Precisión/métodos , Cultivo Primario de Células/métodos , Células Tumorales Cultivadas
13.
Clin Cancer Res ; 26(4): 902-909, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31672771

RESUMEN

PURPOSE: The limited knowledge of the molecular alterations that characterize poorly differentiated neuroendocrine carcinomas has limited the clinical development of targeted agents directed to driver mutations. Here we aim to identify new molecular targets in colon neuroendocrine carcinomas (co-NEC) and proof the efficacy of matching drugs. EXPERIMENTAL DESIGN: We performed a multi-omic analysis of co-NEC to identify genetic or epigenetic alterations that could be exploited as effective drug targets. We compared co-NEC samples with colorectal carcinomas (CRC) to identify neuroendocrine-specific traits. Patients with co-NEC and patient-derived xenografts were treated with a BRAFV600E-blocking drug to demonstrate sensitivity. RESULTS: co-NEC and CRC are similar in their mutational repertoire, although co-NECs are particularly enriched in BRAFV600E mutations. We report for the first time that V600EBRAF-mutant co-NECs may benefit from BRAF inhibition in monotherapy and how EGFR status is essential to predict innate sensitivity and acquired resistance by a differential methylation of its gene regulatory regions. CONCLUSIONS: The identification of V600E BRAF mutations in high-grade co-NECs has allowed the description of radiological responses to combination therapy of BRAF and MEK inhibitors in basket clinical trials. However, the molecular rationale for this treatment combination was based on the presence of the BRAF mutation and the efficacy observed in other cancer types such as melanoma. Future drug development in this setting should test BRAF inhibitors upfront and the addition of anti-EGFR antibodies instead of MEK inhibitors for an efficient blockade of acquired resistance.


Asunto(s)
Neoplasias del Colon/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Carbamatos/farmacología , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/patología , Cetuximab/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Resistencia a Antineoplásicos , Epigénesis Genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/farmacología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Oncol ; 13(8): 1684-1692, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30972907

RESUMEN

Solid pseudopapillary neoplasms (SPNs) are rare and relatively indolent tumors of the pancreas. While primary SPNs can be surgically resected, there are currently no therapies available for patients with advanced stage disease. Given that these tumors frequently carry CTNNB1 hotspot (recurrently mutated loci in a gene) mutations resulting in ß-catenin nuclear accumulation, it has been speculated that the Wnt pathway may be a driver in this disease. Here, we present a comprehensive "multi-omics" study where the genome, transcriptome, and methylome of SPNs were analyzed. We found that SPNs are characterized by a low-complexity genome where somatic mutations in CTNNB1, present in 100% of the cases, are the only actionable genomic lesions. Compared to more common subtypes of pancreatic tumors (adenocarcinomas and pancreatic neuroendocrine tumors), SPNs show high expression levels of genes belonging to the Wnt pathway. Their methylome was consistent with an epithelial cell origin and a general upregulation of Wnt pathway genes. Clinical studies to evaluate the exquisite sensitivity of SPNs to inhibitors of the Wnt pathway are warranted.


Asunto(s)
Carcinoma Papilar/metabolismo , Neoplasias Pancreáticas/metabolismo , Vía de Señalización Wnt , Adulto , Anciano , Carcinoma Papilar/genética , Metilación de ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Neoplasias Pancreáticas/genética , Vía de Señalización Wnt/genética , Adulto Joven
15.
Clin Cancer Res ; 24(4): 794-806, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29242316

RESUMEN

Purpose: Response to standard oncologic treatment is limited in colorectal cancer. The gene expression-based consensus molecular subtypes (CMS) provide a new paradigm for stratified treatment and drug repurposing; however, drug discovery is currently limited by the lack of translation of CMS to preclinical models.Experimental Design: We analyzed CMS in primary colorectal cancers, cell lines, and patient-derived xenografts (PDX). For classification of preclinical models, we developed an optimized classifier enriched for cancer cell-intrinsic gene expression signals, and performed high-throughput in vitro drug screening (n = 459 drugs) to analyze subtype-specific drug sensitivities.Results: The distinct molecular and clinicopathologic characteristics of each CMS group were validated in a single-hospital series of 409 primary colorectal cancers. The new, cancer cell-adapted classifier was found to perform well in primary tumors, and applied to a panel of 148 cell lines and 32 PDXs, these colorectal cancer models were shown to recapitulate the biology of the CMS groups. Drug screening of 33 cell lines demonstrated subtype-dependent response profiles, confirming strong response to EGFR and HER2 inhibitors in the CMS2 epithelial/canonical group, and revealing strong sensitivity to HSP90 inhibitors in cells with the CMS1 microsatellite instability/immune and CMS4 mesenchymal phenotypes. This association was validated in vitro in additional CMS-predicted cell lines. Combination treatment with 5-fluorouracil and luminespib showed potential to alleviate chemoresistance in a CMS4 PDX model, an effect not seen in a chemosensitive CMS2 PDX model.Conclusions: We provide translation of CMS classification to preclinical models and uncover a potential for targeted treatment repurposing in the chemoresistant CMS4 group. Clin Cancer Res; 24(4); 794-806. ©2017 AACR.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Neoplasias Colorrectales/clasificación , Neoplasias Colorrectales/tratamiento farmacológico , Consenso , Fluorouracilo/administración & dosificación , Perfilación de la Expresión Génica/métodos , Humanos , Isoxazoles/administración & dosificación , Ratones Desnudos , Ratones SCID , Resorcinoles/administración & dosificación
16.
J Clin Invest ; 128(9): 3887-3905, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-29944140

RESUMEN

Dormant or slow-cycling tumor cells can form a residual chemoresistant reservoir responsible for relapse in patients, years after curative surgery and adjuvant therapy. We have adapted the pulse-chase expression of H2BeGFP for labeling and isolating slow-cycling cancer cells (SCCCs). SCCCs showed cancer initiation potential and enhanced chemoresistance. Cells at this slow-cycling status presented a distinctive nongenetic and cell-autonomous gene expression profile shared across different tumor types. We identified TET2 epigenetic enzyme as a key factor controlling SCCC numbers, survival, and tumor recurrence. 5-Hydroxymethylcytosine (5hmC), generated by TET2 enzymatic activity, labeled the SCCC genome in carcinomas and was a predictive biomarker of relapse and survival in cancer patients. We have shown the enhanced chemoresistance of SCCCs and revealed 5hmC as a biomarker for their clinical identification and TET2 as a potential drug target for SCCC elimination that could extend patients' survival.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neoplasias/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Recurrencia , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Endocr Relat Cancer ; 14(1): 141-51, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17395983

RESUMEN

The Wnt/beta-catenin signalling pathway is activated in 90% of human colon cancers by nuclear accumulation of beta-catenin protein due to its own mutation or to that of adenomatous polyposis coli. In the nucleus, beta-catenin regulates gene expression promoting cell proliferation, migration and invasiveness. 1alpha,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) inhibits beta-catenin signalling by inducing its binding to vitamin D receptor (VDR) and by promoting beta-catenin nuclear export. The transcription factor Snail1 represses VDR expression and we demonstrate here that Snail1 also abolishes the nuclear export of beta-catenin induced by 1,25(OH)(2)D(3) in SW480-ADH cells. Accordingly, Snail1 relieves the inhibition exerted by 1,25(OH)(2)D(3) on genes whose expression is driven by beta-catenin, such as c-MYC, ectodermal-neural cortex-1 (ENC-1) or ephrin receptor B2 (EPHB2). In addition, Snail1 abrogates the inhibitory effect of 1,25(OH)(2)D(3) on cell proliferation and migration. In xenografted mice, Snail1 impedes the nuclear export of beta-catenin and the inhibition of ENC-1 expression induced by EB1089, a 1,25(OH)(2)D(3) analogue. The elevation of endogenous SNAIL1 protein levels reproduces the effect of an ectopic Snail1 gene. Remarkably, the expression of exogenous VDR in cells with high levels of Snail1 normalizes the transcriptional responses to 1,25(OH)(2)D(3). However, this exogenous VDR failed to fully restore the blockage of the Wnt/beta-catenin pathway by 1,25(OH)(2)D(3). This suggests that the effects of Snail1 on this pathway are not merely due to the repression of VDR gene. We conclude that Snail1 is a positive regulator of the Wnt/beta-catenin signalling pathway in part through the abrogation of the inhibitory action of 1,25(OH)(2)D(3).


Asunto(s)
Factores de Transcripción/metabolismo , Vitamina D/análogos & derivados , beta Catenina/metabolismo , Animales , Calcitriol/análogos & derivados , Calcitriol/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Femenino , Humanos , Ratones , Ratones SCID , Proteínas de Microfilamentos/metabolismo , Trasplante de Neoplasias , Neuropéptidos/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Calcitriol/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Vitamina D/farmacología , Proteínas Wnt/metabolismo , beta Catenina/genética
18.
Mol Cell Biol ; 23(7): 2287-97, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12640114

RESUMEN

beta-Catenin has a key role in the formation of adherens junction through its interactions with E-cadherin and alpha-catenin. We show here that interaction of beta-catenin with alpha-catenin is regulated by the phosphorylation of beta-catenin Tyr-142. This residue can be phosphorylated in vitro by Fer or Fyn tyrosine kinases. Transfection of these kinases to epithelial cells disrupted the association between both catenins. We have also examined whether these kinases are involved in the regulation of this interaction by K-ras. Stable transfectants of the K-ras oncogene in intestinal epithelial IEC18 cells were generated which show little alpha-catenin-beta-catenin association with respect to control clones; this effect is accompanied by increased Tyr-142 phosphorylation and activation of Fer and Fyn kinases. As reported for Fer, Fyn kinase is constitutively bound to p120 catenin; expression of K-ras induces the phosphorylation of p120 catenin on tyrosine residues increasing its affinity for E-cadherin and, consequently, promotes the association of Fyn with the adherens junction complex. Yes tyrosine kinase also binds to p120 catenin but only upon activation, and stimulates Fer and Fyn tyrosine kinases. These results indicate that p120 catenin acts as a docking protein facilitating the activation of Fer/Fyn tyrosine kinases by Yes and demonstrate the role of these p120 catenin-associated kinases in the regulation of beta-catenin-alpha-catenin interaction.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Familia-src Quinasas , Animales , Cateninas , Línea Celular , Células Clonales , Células Epiteliales/citología , Genes ras/fisiología , Humanos , Fosforilación , Pruebas de Precipitina , Unión Proteica/fisiología , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-fyn , Proteínas Proto-Oncogénicas c-yes , Ratas , Transfección , alfa Catenina , beta Catenina , Catenina delta
19.
Mol Oncol ; 11(9): 1263-1272, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28618197

RESUMEN

Sequencing of tumors is now routine and guides personalized cancer therapy. Mutant allele fractions (MAFs, or the 'mutation dose') of a driver gene may reveal the genomic structure of tumors and influence response to targeted therapies. We performed a comprehensive analysis of MAFs of driver alterations in unpaired primary and metastatic colorectal cancer (CRC) at our institution from 2010 to 2015 and studied their potential clinical relevance. Of 763 CRC samples, 622 had detailed annotation on overall survival in the metastatic setting (OSmet) and 89 received targeted agents matched to KRAS (MEK inhibitors), BRAF (BRAF inhibitors), or PIK3CA mutations (PI3K pathway inhibitors). MAFs of each variant were normalized for tumor purity in the sample (adjMAFs). We found lower adjMAFs for BRAFV600E and PIK3CA than for KRAS, NRAS, and BRAF non-V600 variants. TP53 and BRAFV600E adjMAFs were higher in metastases as compared to primary tumors, and high KRAS adjMAFs were found in CRC metastases of patients with KRAS wild-type primary tumors previously exposed to EGFR antibodies. Patients with RAS- or BRAFV600E -mutated tumors, irrespective of adjMAFs, had worse OSmet. There was no significant association between adjMAFs and time to progression on targeted therapies matched to KRAS, BRAF, or PIK3CA mutations, potentially related to the limited antitumor activity of the employed drugs (overall response rate of 4.5%). In conclusion, the lower BRAFV600E and PIK3CA adjMAFs in subsets of primary CRC tumors indicate subclonality of these driver genes. Differences in adjMAFs between metastases and primary tumors suggest that approved therapies may result in selection of BRAFV600E - and KRAS-resistant clones and an increase in genomic heterogeneity with acquired TP53 alterations. Despite significant differences in prognosis according to mutations in driver oncogenes, adjMAFs levels did not impact on survival and did not help predict benefit with matched targeted agents in the metastatic setting.


Asunto(s)
Alelos , Neoplasias Colorrectales/genética , Genes Relacionados con las Neoplasias , Mutación/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Modelos de Riesgos Proporcionales , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA