Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Comput Biol ; 20(2): e1011252, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38363799

RESUMEN

Tumour angiogenesis leads to the formation of blood vessels that are structurally and spatially heterogeneous. Poor blood perfusion, in conjunction with increased hypoxia and oxygen heterogeneity, impairs a tumour's response to radiotherapy. The optimal strategy for enhancing tumour perfusion remains unclear, preventing its regular deployment in combination therapies. In this work, we first identify vascular architectural features that correlate with enhanced perfusion following radiotherapy, using in vivo imaging data from vascular tumours. Then, we present a novel computational model to determine the relationship between these architectural features and blood perfusion in silico. If perfusion is defined to be the proportion of vessels that support blood flow, we find that vascular networks with small mean diameters and large numbers of angiogenic sprouts show the largest increases in perfusion post-irradiation for both biological and synthetic tumours. We also identify cases where perfusion increases due to the pruning of hypoperfused vessels, rather than blood being rerouted. These results indicate the importance of considering network composition when determining the optimal irradiation strategy. In the future, we aim to use our findings to identify tumours that are good candidates for perfusion enhancement and to improve the efficacy of combination therapies.


Asunto(s)
Hipoxia , Neoplasias , Humanos , Perfusión , Terapia Combinada , Oxígeno , Neoplasias/radioterapia
2.
EMBO Rep ; 23(9): e53221, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-35848459

RESUMEN

The effect of radiation therapy on tumor vasculature has long been a subject of debate. Increased oxygenation and perfusion have been documented during radiation therapy. Conversely, apoptosis of endothelial cells in irradiated tumors has been proposed as a major contributor to tumor control. To examine these contradictions, we use multiphoton microscopy in two murine tumor models: MC38, a highly vascularized, and B16F10, a moderately vascularized model, grown in transgenic mice with tdTomato-labeled endothelium before and after a single (15 Gy) or fractionated (5 × 3 Gy) dose of radiation. Unexpectedly, even these high doses lead to little structural change of the perfused vasculature. Conversely, non-perfused vessels and blind ends are substantially impaired after radiation accompanied by apoptosis and reduced proliferation of their endothelium. RNAseq analysis of tumor endothelial cells confirms the modification of gene expression in apoptotic and cell cycle regulation pathways after irradiation. Therefore, we conclude that apoptosis of tumor endothelial cells after radiation does not impair vascular structure.


Asunto(s)
Células Endoteliales , Neoplasias , Animales , Apoptosis , Células Endoteliales/metabolismo , Endotelio/metabolismo , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Radiación Ionizante
3.
Proc Natl Acad Sci U S A ; 117(45): 27811-27819, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33109723

RESUMEN

Oxygen heterogeneity in solid tumors is recognized as a limiting factor for therapeutic efficacy. This heterogeneity arises from the abnormal vascular structure of the tumor, but the precise mechanisms linking abnormal structure and compromised oxygen transport are only partially understood. In this paper, we investigate the role that red blood cell (RBC) transport plays in establishing oxygen heterogeneity in tumor tissue. We focus on heterogeneity driven by network effects, which are challenging to observe experimentally due to the reduced fields of view typically considered. Motivated by our findings of abnormal vascular patterns linked to deviations from current RBC transport theory, we calculated average vessel lengths [Formula: see text] and diameters [Formula: see text] from tumor allografts of three cancer cell lines and observed a substantial reduction in the ratio [Formula: see text] compared to physiological conditions. Mathematical modeling reveals that small values of the ratio λ (i.e., [Formula: see text]) can bias hematocrit distribution in tumor vascular networks and drive heterogeneous oxygenation of tumor tissue. Finally, we show an increase in the value of λ in tumor vascular networks following treatment with the antiangiogenic cancer agent DC101. Based on our findings, we propose λ as an effective way of monitoring the efficacy of antiangiogenic agents and as a proxy measure of perfusion and oxygenation in tumor tissue undergoing antiangiogenic treatment.


Asunto(s)
Circulación Sanguínea/fisiología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Biomarcadores de Tumor/fisiología , Línea Celular Tumoral , Eritrocitos/metabolismo , Heterogeneidad Genética , Hematócrito , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Teóricos , Neoplasias/tratamiento farmacológico , Oxígeno/metabolismo , Perfusión
4.
Semin Cancer Biol ; 71: 143-156, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32526354

RESUMEN

Liver metastases from gastrointestinal and non-gastrointestinal malignancies remain a major cause of cancer-related mortality and a major clinical challenge. The liver has unique properties that facilitate metastatic expansion, including a complex immune system that evolved to dampen immunity to neoantigens entering the liver from the gut, through the portal circulation. In this review, we describe the unique microenvironment encountered by cancer cells in the liver, focusing on elements of the innate and adaptive immune response that can act as a double-edge sword, contributing to the elimination of cancer cells on the one hand and promoting their survival and growth, on the other. We discuss this microenvironment in a clinical context, particularly for colorectal carcinoma, and highlight how a better understanding of the role of the microenvironment has spurred an intense effort to develop novel and innovative strategies for targeting liver metastatic disease, some of which are currently being tested in the clinic.


Asunto(s)
Neoplasias Colorrectales/patología , Inmunidad , Neoplasias Hepáticas/secundario , Microambiente Tumoral , Animales , Neoplasias Colorrectales/inmunología , Humanos , Neoplasias Hepáticas/inmunología
5.
Br J Cancer ; 125(4): 534-546, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34155340

RESUMEN

BACKGROUND: There is a need to improve the treatment of prostate cancer (PCa) and reduce treatment side effects. Vascular-targeted photodynamic therapy (VTP) is a focal therapy for low-risk low-volume localised PCa, which rapidly disrupts targeted tumour vessels. There is interest in expanding the use of VTP to higher-risk disease. Tumour vasculature is characterised by vessel immaturity, increased permeability, aberrant branching and inefficient flow. FRT alters the tumour microenvironment and promotes transient 'vascular normalisation'. We hypothesised that multimodality therapy combining fractionated radiotherapy (FRT) and VTP could improve PCa tumour control compared against monotherapy with FRT or VTP. METHODS: We investigated whether sequential delivery of FRT followed by VTP 7 days later improves flank TRAMP-C1 PCa tumour allograft control compared to monotherapy with FRT or VTP. RESULTS: FRT induced 'vascular normalisation' changes in PCa flank tumour allografts, improving vascular function as demonstrated using dynamic contrast-enhanced magnetic resonance imaging. FRT followed by VTP significantly delayed tumour growth in flank PCa allograft pre-clinical models, compared with monotherapy with FRT or VTP, and improved overall survival. CONCLUSION: Combining FRT and VTP may be a promising multimodal approach in PCa therapy. This provides proof-of-concept for this multimodality treatment to inform early phase clinical trials.


Asunto(s)
Neovascularización Patológica/terapia , Fotoquimioterapia/métodos , Neoplasias de la Próstata/terapia , Animales , Línea Celular Tumoral , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Neoplasias de la Próstata/irrigación sanguínea , Análisis de Supervivencia , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Br J Cancer ; 123(7): 1089-1100, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32641865

RESUMEN

BACKGROUND: Radiotherapy enhances innate and adaptive anti-tumour immunity. It is unclear whether this effect may be harnessed by combining immunotherapy with radiotherapy fractions used to treat prostate cancer. We investigated tumour immune microenvironment responses of pre-clinical prostate cancer models to radiotherapy. Having defined this landscape, we tested whether radiotherapy-induced tumour growth delay could be enhanced with anti-PD-L1. METHODS: Hypofractionated radiotherapy was delivered to TRAMP-C1 and MyC-CaP flank allografts. Tumour growth delay, tumour immune microenvironment flow-cytometry, and immune gene expression were analysed. TRAMP-C1 allografts were then treated with 3 × 5 Gy ± anti-PD-L1. RESULTS: 3 × 5 Gy caused tumour growth delay in TRAMP-C1 and MyC-CaP. Tumour immune microenvironment changes in TRAMP-C1 at 7 days post-radiotherapy included increased tumour-associated macrophages and dendritic cells and upregulation of PD-1/PD-L1, CD8+ T-cell, dendritic cell, and regulatory T-cell genes. At tumour regrowth post-3 × 5 Gy the tumour immune microenvironment flow-cytometry was similar to control tumours, however CD8+, natural killer and dendritic cell gene transcripts were reduced. PD-L1 inhibition plus 3 × 5 Gy in TRAMP-C1 did not enhance tumour growth delay versus monotherapy. CONCLUSION: 3 × 5 Gy hypofractionated radiotherapy can result in tumour growth delay and immune cell changes in allograft prostate cancer models. Adjuncts beyond immunomodulation may be necessary to improve the radiotherapy-induced anti-tumour response.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias de la Próstata/terapia , Hipofraccionamiento de la Dosis de Radiación , Microambiente Tumoral , Animales , Antígeno B7-H1/análisis , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase I/análisis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
7.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G625-G639, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31545917

RESUMEN

Dissemination of primary tumors to distant anatomical sites has a substantial negative impact on patient prognosis. The liver is a common site for metastases from colorectal cancer, and patients with hepatic metastases have generally much shorter survival, raising a need to develop and implement novel strategies for targeting metastatic disease. The extracellular matrix (ECM) is a meshwork of highly crosslinked, insoluble high-molecular-mass proteins maintaining tissue integrity and establishing cell-cell interactions. Emerging evidence identifies the importance of the ECM in cancer cell migration, invasion, intravasation, and metastasis. Here, we isolated the ECM from MC38 mouse liver metastases using our optimized method of mild detergent solubilization followed by biochemical enrichment. The matrices were subjected to label-free quantitative mass spectrometry analysis, revealing proteins highly abundant in the metastatic matrisome. The resulting list of proteins upregulated in the ECM significantly predicted survival in patients with colorectal cancer but not other cancers with strong involvement of the ECM component. One of the proteins upregulated in liver metastatic ECM, annexin A1, was not previously studied in the context of cancer-associated matrisome. Here, we show that annexin A1 was markedly upregulated in colon cancer cell lines compared with cancer cells of other origin and also over-represented in human primary colorectal lesions, as well as hepatic metastases, compared with their adjacent healthy tissue counterparts. In conclusion, our study provides a comprehensive ECM characterization of MC38 experimental liver metastases and proposes annexin A1 as a putative target for this disease.NEW & NOTEWORTHY Here, the authors provide an extensive proteomics characterization of murine colorectal cancer liver metastasis matrisome (the ensemble of all extracellular matrix molecules). The findings presented in this study may enable identification of therapeutic targets or biomarkers of hepatic metastases.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Hepáticas/genética , Proteoma/metabolismo , Animales , Anexina A1/genética , Anexina A1/metabolismo , Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas de la Matriz Extracelular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Ratones Endogámicos C57BL , Proteoma/genética , Regulación hacia Arriba
8.
Am J Pathol ; 188(3): 795-804, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29339090

RESUMEN

There is an unmet clinical need for adequate biomarkers to aid risk stratification and management of prostate cancer (PCa) patients. Even within the high-risk PCa category, not all patients will invariably have a poor prognosis, and improved stratification of this heterogeneous group is needed. In this context, components of the hedgehog (Hh) pathway may have promise as biomarkers, because the available evidence suggests increased Hh pathway activity may confer a poorer outcome in advanced and castrate-resistant PCa. In this study, potential associations between Hh pathway protein expression and clinicopathological factors, including time to biochemical recurrence (BCR), were investigated using a tissue microarray constructed from benign and malignant prostate samples from 75 predominantly high-risk PCa patients who underwent radical prostatectomy. Hh signaling activity was found to differ between benign and malignant prostate tissue, with a greater amount of active Hh signaling present in malignant than benign prostate epithelium. High expression of Patched 1 in malignant prostate epithelium was found to be an independent predictor of BCR in high-risk PCa patients. Glioma-associated oncogene 1 may potentially represent a clinically useful biomarker of an aggressive tumor phenotype. Evaluation of Hh signaling activity in PCa patients may be useful for risk stratification, and epithelial Patched 1 expression, in particular, may be a prognostic marker for BCR in high-risk PCa patients.


Asunto(s)
Adenocarcinoma/metabolismo , Receptor Patched-1/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/patología , Anciano , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Próstata/patología , Neoplasias de la Próstata/patología , Recurrencia
9.
Mol Cell ; 41(2): 210-20, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21195000

RESUMEN

Expression of BRCA1 is commonly decreased in sporadic breast tumors, and this correlates with poor prognosis of breast cancer patients. Here we show that BRCA1 transcripts are selectively enriched in the Argonaute/miR-182 complex and miR-182 downregulates BRCA1 expression. Antagonizing miR-182 enhances BRCA1 protein levels and protects them from IR-induced cell death, while overexpressing miR-182 reduces BRCA1 protein, impairs homologous recombination-mediated repair, and render cells hypersensitive to IR. The impaired DNA repair phenotype induced by miR-182 overexpression can be fully rescued by overexpressing miR-182-insensitive BRCA1. Consistent with a BRCA1-deficiency phenotype, miR-182-overexpressing breast tumor cells are hypersensitive to inhibitors of poly (ADP-ribose) polymerase 1 (PARP1). Conversely, antagonizing miR-182 enhances BRCA1 levels and induces resistance to PARP1 inhibitor. Finally, a clinical-grade PARP1 inhibitor impacts outgrowth of miR-182-expressing tumors in animal models. Together these results suggest that miR-182-mediated downregulation of BRCA1 impedes DNA repair and may impact breast cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/genética , Reparación del ADN/efectos de los fármacos , MicroARNs/fisiología , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Diferenciación Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de la radiación , Regulación hacia Abajo , Humanos , Células K562 , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Poli(ADP-Ribosa) Polimerasa-1
10.
Br J Cancer ; 118(3): 435-440, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29360819

RESUMEN

BACKGROUND: Accumulating evidence implicates the tumour stroma as an important determinant of cancer progression but the protein constituents relevant for this effect are unknown. Here we utilised a bioinformatics approach to identify an extracellular matrix (ECM) gene signature overexpressed in multiple cancer types and strongly predictive of adverse outcome. METHODS: Gene expression levels in cancers were determined using Oncomine. Geneset enrichment analysis was performed using the Broad Institute desktop application. Survival analysis was performed using KM plotter. Survival data were generated from publically available genesets. RESULTS: We analysed ECM genes significantly upregulated across a large cohort of patients with ovarian, lung, gastric and colon cancers and defined a signature of nine commonly upregulated genes. Each of these nine genes was considerably overexpressed in all the cancers studied, and cumulatively, their expression was associated with poor prognosis across all data sets. Further, the gene signature expression was associated with enrichment of genes governing processes linked to poor prognosis, such as EMT, angiogenesis, hypoxia, and inflammation. CONCLUSIONS: Here we identify a nine-gene ECM signature, which strongly predicts outcome across multiple cancer types and can be used for prognostication after validation in prospective cancer cohorts.


Asunto(s)
Adenocarcinoma/genética , Matriz Extracelular/genética , Expresión Génica , Neoplasias/genética , Neoplasias de la Mama/genética , Hipoxia de la Célula/genética , Neoplasias del Colon/genética , Biología Computacional , Transición Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Femenino , Humanos , Inflamación/genética , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neovascularización Patológica/genética , Neoplasias Ováricas/genética , Pronóstico , Neoplasias Gástricas/genética , Tasa de Supervivencia , Regulación hacia Arriba
11.
Proc Natl Acad Sci U S A ; 112(31): 9710-5, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195776

RESUMEN

Current strategies for early detection of breast and other cancers are limited in part because some lesions identified as potentially malignant do not develop into aggressive tumors. Acid pH has been suggested as a key characteristic of aggressive tumors that might distinguish aggressive lesions from more indolent pathology. We therefore investigated the novel class of molecules, pH low insertion peptides (pHLIPs), as markers of low pH in tumor allografts and of malignant lesions in a mouse model of spontaneous breast cancer, BALB/neu-T. pHLIP Variant 3 (Var3) conjugated with fluorescent Alexa546 was shown to insert into tumor spheroids in a sequence-specific manner. Its signal reflected pH in murine tumors. It was induced by carbonic anhydrase IX (CAIX) overexpression and inhibited by acetazolamide (AZA) administration. By using (31)P magnetic resonance spectroscopy (MRS), we demonstrated that pHLIP Var3 was retained in tumors of pH equal to or less than 6.7 but not in tissues of higher pH. In BALB/neu-T mice at different stages of the disease, the fluorescent signal from pHLIP Var3 marked cancerous lesions with a very low false-positive rate. However, only ∼60% of the smallest lesions retained a pHLIP Var3 signal, suggesting heterogeneity in pH. Taken together, these results show that pHLIP can identify regions of lower pH, allowing for its development as a theranostic tool for clinical applications.


Asunto(s)
Ácidos/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mutantes/metabolismo , Neoplasias/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Proteínas de la Membrana/química , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Invasividad Neoplásica , Neoplasias/patología , Curva ROC , Sensibilidad y Especificidad , Esferoides Celulares/metabolismo
12.
Biophys J ; 112(9): 1767-1772, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28494948

RESUMEN

Spatial models of vascularized tissues are widely used in computational physiology. We introduce a software library for composing multiscale, multiphysics models for applications including tumor growth, angiogenesis, osteogenesis, coronary perfusion, and oxygen delivery. Composition of such models is time consuming, with many researchers writing custom software. Recent advances in imaging have produced detailed three-dimensional (3D) datasets of vascularized tissues at the scale of individual cells. To fully exploit such data there is an increasing need for software that allows user-friendly composition of efficient, 3D models of vascularized tissues, and comparison of predictions with in vivo or in vitro experiments and alternative computational formulations. Microvessel Chaste can be used to build simulations of vessel growth and adaptation in response to mechanical and chemical stimuli; intra- and extravascular transport of nutrients, growth factors and drugs; and cell proliferation in complex 3D geometries. In addition, it can be used to develop custom software for integrating modeling with experimental data processing workflows, facilitated by a comprehensive Python interface to solvers implemented in C++. This article links to two reproducible example problems, showing how the library can be used to build simulations of tumor growth and angiogenesis with realistic vessel networks.


Asunto(s)
Simulación por Computador , Microvasos , Modelos Biológicos , Programas Informáticos , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Algoritmos , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Neoplasias del Colon/fisiopatología , Córnea/irrigación sanguínea , Córnea/fisiología , Imagenología Tridimensional , Internet , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
BMC Cancer ; 17(1): 634, 2017 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-28877722

RESUMEN

BACKGROUND: Prostate cancer (PCa) is a heterogeneous disease with a variable natural history, genetics, and treatment outcome. The Hedgehog (Hh) signaling pathway is increasingly recognized as being potentially important for the development and progression of PCa. In this retrospective study, we compared the activation status of the Hh signaling pathway between benign and tumor tissue, and evaluated the clinical significance of Hh signaling in PCa. METHODS: In this tissue microarray (TMA) study, the protein expression of several Hh signaling components and Hh target proteins, along with microvessel density, were compared between benign (n = 64) and malignant (n = 170) prostate tissue, and correlated with PCa clinicopathological characteristics and biochemical recurrence (BCR). RESULTS: The Hh signaling pathway appeared to be more active in PCa than in benign prostate tissue, as demonstrated by lower expression of the negative regulators PTCH1 and GLI3 in the tumor tissue compared to benign. In addition, high epithelial GLI2 expression correlated with higher pathological Gleason score. Overall, higher epithelial GLI3 expression in the tumor was shown to be an independent marker of a favorable prognosis. CONCLUSION: Hh signaling activation might reflect aggressive tumoral behavior, since high epithelial GLI2 expression positively correlates with a higher pathological Gleason score. Moreover, higher epithelial GLI3 expression is an independent marker of a more favorable prognosis.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/mortalidad , Transducción de Señal , Anciano , Progresión de la Enfermedad , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Modelos Biológicos , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Neoplasias de la Próstata/patología , Recurrencia , Análisis de Supervivencia , Análisis de Matrices Tisulares
14.
Int J Mol Sci ; 18(2)2017 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-28208838

RESUMEN

The anti-diabetes drug metformin has been shown to have anti-neoplastic effects in several tumor models through its effects on energy metabolism and protein synthesis. Recent studies show that metformin also targets Hedgehog (Hh) signaling, a developmental pathway re-activated in several tumor types, including prostate cancer (PCa). Furthermore, we and others have shown that Hh signaling is an important target for radiosensitization. Here, we evaluated the combination of metformin and the Hh inhibitor GANT61 (GLI-ANTagonist 61) with or without ionizing radiation in three PCa cell lines (PC3, DU145, 22Rv1). The effect on proliferation, radiosensitivity, apoptosis, cell cycle distribution, reactive oxygen species production, DNA repair, gene and protein expression was investigated. Furthermore, this treatment combination was also assessed in vivo. Metformin was shown to interact with Hh signaling by inhibiting the effector protein glioma-associated oncogene homolog 1 (GLI1) in PCa cells both in vitro and in vivo. The combination of metformin and GANT61 significantly inhibited PCa cell growth in vitro and enhanced the radiation response of 22Rv1 cells compared to either single agent. Nevertheless, neither the growth inhibitory effect nor the radiosensitization effect of the combination treatment observed in vitro was seen in vivo. Although the interaction between metformin and Hh signaling seems to be promising from a therapeutic point of view in vitro, more research is needed when implementing this combination strategy in vivo.


Asunto(s)
Metformina/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/radioterapia , Radiación Ionizante , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biochim Biophys Acta ; 1855(1): 61-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25489989

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death. Despite improvements in the clinical management, the prognosis of PDAC remains dismal. In the present comprehensive review, we will examine the knowledge of PDAC genetics and the new insights into human genome sequencing and clonal evolution. Additionally, the biology and the role of the stroma in tumour progression and response to treatment will be presented. Furthermore, we will describe the evidence on tumour chemoresistance and radioresistance and will provide an overview on the recent advances in PDAC metabolism and circulating tumour cells. Next, we will explore the characteristics and merits of the different mouse models of PDAC. The inflammatory milieu and the immunosuppressive microenvironment mediate tumour initiation and treatment failure. Hence, we will also review the inflammatory and immune escaping mechanisms and the new immunotherapies tested in PDAC. A better understanding of the different mechanisms of tumour formation and progression will help us to identify the best targets for testing in future clinical studies of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Inmunoterapia , Neoplasias Pancreáticas , Tolerancia a Radiación , Animales , Vacunas contra el Cáncer/uso terapéutico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/terapia , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Humanos , Inmunoterapia/métodos , Inflamación/patología , Ratones , Células Neoplásicas Circulantes/inmunología , Células Neoplásicas Circulantes/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Tolerancia a Radiación/genética , Neoplasias Pancreáticas
16.
Hepatology ; 62(2): 521-33, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25854806

RESUMEN

UNLABELLED: Myeloid cells are known to mediate metastatic progression. Here, we attempted to elucidate the mechanisms underlying these effects by identifying gene expression alterations in cancer cells forming hepatic metastases after myeloid cell depletion. Hepatic metastases are heavily infiltrated by CD11b(+) myeloid cells. We established hepatic metastases in transgenic CD11b-diphtheria toxin receptor mice by intrasplenic injection of MC38 colon and Lewis lung carcinoma cells before depleting myeloid cells with diphtheria toxin. Myeloid cell depletion inhibited metastatic growth with a marked diminishment of tumor vasculature. Expression of ANGPTL7 (angiopoietin-like 7), a protein not previously linked to metastasis, was highly up-regulated in cancer cells after myeloid cell depletion. This effect was duplicated in tissue culture, where coculture of cancer cells with tumor-conditioned myeloid cells from liver metastases or myeloid cell conditioned media down-regulated ANGPTL7 expression. Analogous to myeloid cell depletion, overexpression of ANGPTL7 in cancer cells significantly reduced hepatic metastasis formation and angiogenesis. We found that ANGPTL7 itself has strong antiangiogenic effects in vitro. Furthermore, analysis of The Cancer Genome Atlas colorectal and breast cancer data sets revealed striking ANGPTL7 underexpression in cancerous compared to normal tissues. Also, ANGPTL7 was down-regulated in metastatic liver colonies of colorectal cancer patients compared to their adjacent liver tissue. CONCLUSION: Myeloid cells promote liver metastasis by down-regulating ANGPTL7 expression in cancer cells; our findings implicate ANGPTL7 as a mediator of metastatic progression and a potential target for interference with liver metastases.


Asunto(s)
Angiopoyetinas/genética , Antígeno CD11b/genética , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Células Mieloides/patología , Neovascularización Patológica/patología , Proteína 7 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Movimiento Celular , Medios de Cultivo Condicionados , Regulación hacia Abajo , Femenino , Humanos , Neoplasias Hepáticas/fisiopatología , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/fisiología , Células Tumorales Cultivadas
17.
Blood ; 121(16): 3289-97, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23407548

RESUMEN

Pulmonary metastasis is a frequent cause of poor outcome in cancer patients. The formation of pulmonary metastasis is greatly facilitated by recruitment of myeloid cells, which are crucial for tumor cell survival and extravasation. During inflammation, homing of myeloid cells is mediated by endothelial activation, raising the question of a potential role for endothelial activation in myeloid cell recruitment during pulmonary metastasis. Here, we show that metastatic tumor cell attachment causes the induction of the endothelial activation markers vascular cell adhesion molecule-1 (VCAM-1) and vascular adhesion protein-1 (VAP-1). Induction of VCAM-1 is dependent on tumor cell-clot formation, decreasing upon induction of tissue factor pathway inhibitor or treatment with hirudin. Furthermore, inhibition of endothelial activation with a VCAM-1 blocking antibody or a VAP-1 small molecule inhibitor leads to reduced myeloid cell recruitment and diminished tumor cell survival and metastasis without affecting tumor cell adhesion. Simultaneous inhibition of VCAM-1 and VAP-1 does not result in further reduction in myeloid cell recruitment and tumor cell survival, suggesting that both act through closely related mechanisms. These results establish VCAM-1 and VAP-1 as mediators of myeloid cell recruitment in metastasis and identify VAP-1 as a potential target for therapeutic intervention to combat early metastasis.


Asunto(s)
Amina Oxidasa (conteniendo Cobre)/inmunología , Moléculas de Adhesión Celular/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Pulmón/patología , Células Mieloides/patología , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Coagulación Sanguínea , Adhesión Celular , Línea Celular Tumoral , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Pulmón/inmunología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Células Mieloides/inmunología
18.
Proc Natl Acad Sci U S A ; 109(17): 6674-9, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22451897

RESUMEN

Metastasis to the brain is a leading cause of cancer mortality. The current diagnostic method of gadolinium-enhanced MRI is sensitive only to larger tumors, when therapeutic options are limited. Earlier detection of brain metastases is critical for improved treatment. We have developed a targeted MRI contrast agent based on microparticles of iron oxide that enables imaging of endothelial vascular cell adhesion molecule-1 (VCAM-1). Our objectives here were to determine whether VCAM-1 is up-regulated on vessels associated with brain metastases, and if so, whether VCAM-1-targeted MRI enables early detection of these tumors. Early up-regulation of cerebrovascular VCAM-1 expression was evident on tumor-associated vessels in two separate murine models of brain metastasis. Metastases were detectable in vivo using VCAM-1-targeted MRI 5 d after induction (<1,000 cells). At clinical imaging resolutions, this finding is likely to translate to detection at tumor volumes two to three orders of magnitude smaller (0.3-3 × 10(5) cells) than those volumes detectable clinically (10(7)-10(8) cells). VCAM-1 expression detected by MRI increased significantly (P < 0.0001) with tumor progression, and tumors showed no gadolinium enhancement. Importantly, expression of VCAM-1 was shown in human brain tissue containing both established metastases and micrometastases. Translation of this approach to the clinic could increase therapeutic options and change clinical management in a substantial number of cancer patients.


Asunto(s)
Neoplasias Encefálicas/secundario , Imagen por Resonancia Magnética/métodos , Animales , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Progresión de la Enfermedad , Humanos , Inmunohistoquímica , Ratones , Sensibilidad y Especificidad , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
Am J Pathol ; 183(1): 26-34, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23665347

RESUMEN

Brain metastasis is a frequent occurrence in patients with cancer, with devastating consequences. The current animal models for brain metastasis are highly variable, leading to a need for improved in vivo models that recapitulate the clinical disease. Herein, we describe an experimental brain metastasis model that uses ultrasonographic guidance to perform intracardiac injections. This method is easy to perform, giving consistent and quantitative results. Demonstrating the utility of this method, we have assessed a variety of metastatic cell lines for their ability to develop into brain metastases. Those cell lines that were competent at brain colonization could be detected in the brain vasculature 4 hours after intracardiac injection, and a few adherent cells persisted until colonization occurred. In contrast, those cell lines that were deficient in brain colonization were infrequently found 4 hours after introduction into the arterial circulation and were not detected at later time points. All of these cells were capable of brain colonization after intraparenchymal injection. We propose that adherence to the brain vasculature may be the key limiting step that determines the ability of a cancer cell to form brain metastases successfully. Identifying brain endothelium-specific adhesion molecules may enable development of screening modalities to detect brain-colonizing cancer cells and therapies to prevent these metastatic cells from seeding the brain.


Asunto(s)
Neoplasias Encefálicas/secundario , Micrometástasis de Neoplasia , Ultrasonografía Intervencional , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Ventrículos Cardíacos , Humanos , Inyecciones , Mediciones Luminiscentes , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID
20.
Hepatology ; 57(2): 829-39, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23081697

RESUMEN

UNLABELLED: Liver metastasis from colorectal cancer is a leading cause of cancer mortality. Myeloid cells play pivotal roles in the metastatic process, but their prometastatic functions in liver metastasis remain incompletely understood. To investigate their role, we simulated liver metastasis in C57BL/6 mice through intrasplenic inoculation of MC38 colon carcinoma cells. Among the heterogeneous myeloid infiltrate, we identified a distinct population of CD11b/Gr1(mid) cells different from other myeloid populations previously associated with liver metastasis. These cells increased in number dramatically during establishment of liver metastases and were recruited from bone marrow by tumor-derived CCL2. Liver metastasis of Lewis lung carcinoma cells followed this pattern but this mechanism is not universal as liver colonization by B16F1 melanoma cells did not recruit similar subsets. Inhibition of CCL2 signaling and absence of its cognate receptor CCR2 reduced CD11b/Gr1(mid) recruitment and decreased tumor burden. Depletion of the CD11b/Gr1(mid) subset in a transgenic CD11b-diphtheria toxin receptor mouse model markedly reduced tumor cell proliferation. There was no evidence for involvement of an adaptive immune response in the prometastatic effects of CD11b/Gr1(mid) cells. Additionally, an analogous myeloid subset was found in liver metastases of some colorectal cancer patients. CONCLUSION: Collectively, our findings highlight the importance of myeloid cells--in this case a selective CD11b/Gr1(mid) subset--in sustaining development of colorectal cancer liver metastasis and identify a potential target for antimetastatic therapy.


Asunto(s)
Quimiocina CCL2/fisiología , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Células Mieloides/inmunología , Receptores CCR2/fisiología , Animales , Antígeno CD11b/inmunología , Neoplasias Colorrectales/inmunología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Modelos Animales , Células Mieloides/patología , Células Mieloides/trasplante , Trasplante de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA