Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 527(7576): 100-104, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26479035

RESUMEN

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Exosomas/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , MicroARNs/genética , Fosfohidrolasa PTEN/deficiencia , Microambiente Tumoral , Adaptación Fisiológica/genética , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/metabolismo , Proteínas de Unión al Calcio , Proliferación Celular/genética , Quimiocina CCL2/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Evolución Molecular , Exosomas/metabolismo , Femenino , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Proteínas de Microfilamentos , Fosfohidrolasa PTEN/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Microambiente Tumoral/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
2.
Am J Pathol ; 182(6): 2368-79, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583652

RESUMEN

Brain metastases occur in more than one-third of metastatic breast cancer patients whose tumors overexpress HER2 or are triple negative. Brain colonization of cancer cells occurs in a unique environment, containing microglia, oligodendrocytes, astrocytes, and neurons. Although a neuroinflammatory response has been documented in brain metastasis, its contribution to cancer progression and therapy remains poorly understood. Using an experimental brain metastasis model, we characterized the brain metastatic microenvironment of brain tropic, HER2-transfected MDA-MB-231 human breast carcinoma cells (231-BR-HER2). A previously unidentified subpopulation of metastasis-associated astrocytes expressing phosphorylated platelet-derived growth factor receptor ß (at tyrosine 751; p751-PDGFRß) was identified around perivascular brain micrometastases. p751-PDGFRß(+) astrocytes were also identified in human brain metastases from eight craniotomy specimens and in primary cultures of astrocyte-enriched glial cells. Previously, we reported that pazopanib, a multispecific tyrosine kinase inhibitor, prevented the outgrowth of 231-BR-HER2 large brain metastases by 73%. Here, we evaluated the effect of pazopanib on the brain neuroinflammatory microenvironment. Pazopanib treatment resulted in 70% (P = 0.023) decrease of the p751-PDGFRß(+) astrocyte population, at the lowest dose of 30 mg/kg, twice daily. Collectively, the data identify a subpopulation of activated astrocytes in the subclinical perivascular stage of brain metastases and show that they are inhibitable by pazopanib, suggesting its potential to prevent the development of brain micrometastases in breast cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Astrocitos/efectos de los fármacos , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sulfonamidas/farmacología , Animales , Antineoplásicos/uso terapéutico , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Neoplasias de la Mama/patología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Indazoles , Ratones , Micrometástasis de Neoplasia/patología , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Fosforilación/efectos de los fármacos , Pirimidinas/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Células Tumorales Cultivadas , Microambiente Tumoral
3.
BMC Cancer ; 12: 583, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23217186

RESUMEN

BACKGROUND: Brain metastasis is an increasingly common complication for breast cancer patients; approximately 15- 30% of breast cancer patients develop brain metastasis. However, relatively little is known about how these metastases form, and what phenotypes are characteristic of cells with brain metastasizing potential. In this study, we show that the targeted knockdown of MMP-1 in breast cancer cells with enhanced brain metastatic ability not only reduced primary tumor growth, but also significantly inhibited brain metastasis. METHODS: Two variants of the MDA-MB-231 human breast cancer cell line selected for enhanced ability to form brain metastases in nude mice (231-BR and 231-BR3 cells) were found to express high levels of matrix metalloproteinase-1 (MMP-1). Short hairpin RNA-mediated stable knockdown of MMP-1 in 231-BR and 231-BR3 cells were established to analyze tumorigenic ability and metastatic ability. RESULTS: Short hairpin RNA-mediated stable knockdown of MMP-1 inhibited the invasive ability of MDA-MB 231 variant cells in vitro, and inhibited breast cancer growth when the cells were injected into the mammary fat pad of nude mice. Reduction of MMP-1 expression significantly attenuated brain metastasis and lung metastasis formation following injection of cells into the left ventricle of the heart and tail vein, respectively. There were significantly fewer proliferating cells in brain metastases of cells with reduced MMP-1 expression. Furthermore, reduced MMP-1 expression was associated with decreased TGFα release and phospho-EGFR expression in 231-BR and BR3 cells. CONCLUSIONS: Our results show that elevated expression of MMP-1 can promote the local growth and the formation of brain metastases by breast cancer cells.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Metaloproteinasa 1 de la Matriz/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Metaloproteinasa 1 de la Matriz/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo , Trasplante Heterólogo
4.
Pharm Res ; 29(3): 770-81, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22011930

RESUMEN

PURPOSE: Lapatinib, a small molecule EGFR/HER2 inhibitor, partially inhibits the outgrowth of HER2+ brain metastases in preclinical models and in a subset of CNS lesions in clinical trials of HER2+ breast cancer. We investigated the ability of lapatinib to reach therapeutic concentrations in the CNS following (14)C-lapatinib administration (100 mg/kg p.o. or 10 mg/kg, i.v.) to mice with MDA-MD-231-BR-HER2 brain metastases of breast cancer. METHODS: Drug concentrations were determined at differing times after administration by quantitative autoradiography and chromatography. RESULTS: (14)C-Lapatinib concentration varied among brain metastases and correlated with altered blood-tumor barrier permeability. On average, brain metastasis concentration was 7-9-fold greater than surrounding brain tissue at 2 and 12 h after oral administration. However, average lapatinib concentration in brain metastases was still only 10-20% of those in peripheral metastases. Only in a subset of brain lesions (17%) did lapatinib concentration approach that of systemic metastases. No evidence was found of lapatinib resistance in tumor cells cultured ex vivo from treated brains. CONCLUSIONS: Results show that lapatinib distribution to brain metastases of breast cancer is partially restricted and blood-tumor barrier permeability is a key component of lapatinib therapeutic efficacy which varies between tumors.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Encéfalo/patología , Neoplasias de la Mama/patología , Quinazolinas/farmacocinética , Receptor ErbB-2/genética , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Inyecciones Intravenosas , Lapatinib , Ratones , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Regulación hacia Arriba
5.
J Transl Med ; 7: 88, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19840404

RESUMEN

BACKGROUND: Establishing a large rodent model of brain metastasis that can be monitored using clinically relevant magnetic resonance imaging (MRI) techniques is challenging. Non-invasive imaging of brain metastasis in mice usually requires high field strength MR units and long imaging acquisition times. Using the brain seeking MDA-MB-231BR transfected with luciferase gene, a metastatic breast cancer brain tumor model was investigated in the nude rat. Serial MRI and bioluminescence imaging (BLI) was performed and findings were correlated with histology. Results demonstrated the utility of multimodality imaging in identifying unexpected sights of metastasis and monitoring the progression of disease in the nude rat. METHODS: Brain seeking breast cancer cells MDA-MB-231BR transfected with firefly luciferase (231BRL) were labeled with ferumoxides-protamine sulfate (FEPro) and 1-3 x 106 cells were intracardiac (IC) injected. MRI and BLI were performed up to 4 weeks to monitor the early breast cancer cell infiltration into the brain and formation of metastases. Rats were euthanized at different time points and the imaging findings were correlated with histological analysis to validate the presence of metastases in tissues. RESULTS: Early metastasis of the FEPro labeled 231BRL were demonstrated on T2*-weighted MRI and BLI within 1 week post IC injection of cells. Micro-metastatic tumors were detected in the brain on T2-weighted MRI as early as 2 weeks post-injection in greater than 85% of rats. Unexpected skeletal metastases from the 231BRL cells were demonstrated and validated by multimodal imaging. Brain metastases were clearly visible on T2 weighted MRI by 3-4 weeks post infusion of 231BRL cells, however BLI did not demonstrate photon flux activity originating from the brain in all animals due to scattering of the photons from tumors. CONCLUSION: A model of metastatic breast cancer in the nude rat was successfully developed and evaluated using multimodal imaging including MRI and BLI providing the ability to study the temporal and spatial distribution of metastases in the brain and skeleton.


Asunto(s)
Neoplasias de la Mama/patología , Mediciones Luminiscentes/métodos , Imagen por Resonancia Magnética/métodos , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia , Animales , Neoplasias Óseas/secundario , Encéfalo/anatomía & histología , Encéfalo/patología , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Dextranos , Femenino , Óxido Ferrosoférrico/química , Óxido Ferrosoférrico/metabolismo , Humanos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Nanopartículas de Magnetita , Ratones , Protaminas/química , Protaminas/metabolismo , Ratas
6.
Pharm Res ; 26(11): 2486-94, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19774344

RESUMEN

PURPOSE: We evaluated the uptake of angiopep-2 paclitaxel conjugate, ANG1005, into brain and brain metastases of breast cancer in rodents. Most anticancer drugs show poor delivery to brain tumors due to limited transport across the blood-brain barrier (BBB). To overcome this, a 19-amino acid peptide (angiopep-2) was developed that binds to low density lipoprotein receptor-related protein (LRP) receptors at the BBB and has the potential to deliver drugs to brain by receptor-mediated transport. METHODS: The transfer coefficient (K(in)) for brain influx was measured by in situ rat brain perfusion. Drug distribution was determined at 30 min after i.v. injection in mice bearing intracerebral MDA-MB-231BR metastases of breast cancer. RESULTS: The BBB K(in) for (125)I-ANG1005 uptake (7.3 +/- 0.2 x 10(-3) mL/s/g) exceeded that for (3)H-paclitaxel (8.5 +/- 0.5 x 10(-5)) by 86-fold. Over 70% of (125)I-ANG1005 tracer stayed in brain after capillary depletion or vascular washout. Brain (125)I-ANG1005 uptake was reduced by unlabeled angiopep-2 vector and by LRP ligands, consistent with receptor transport. In vivo uptake of (125)I-ANG1005 into vascularly corrected brain and brain metastases exceeded that of (14)C-paclitaxel by 4-54-fold. CONCLUSIONS: The results demonstrate that ANG1005 shows significantly improved delivery to brain and brain metastases of breast cancer compared to free paclitaxel.


Asunto(s)
Antineoplásicos Fitogénicos/farmacocinética , Barrera Hematoencefálica/metabolismo , Paclitaxel/análogos & derivados , Animales , Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Cromatografía Líquida de Alta Presión , Femenino , Ratones , Ratones Desnudos , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Péptidos , Ratas
7.
Cancer Res ; 67(15): 7238-46, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671192

RESUMEN

Exogenous overexpression of the metastasis suppressor gene Nm23-H1 reduces the metastatic potential of multiple types of cancer cells and suppresses in vitro tumor cell motility and invasion. Mutational analysis of Nm23-H1 revealed that substitution mutants P96S and S120G did not inhibit motility and invasion. To elucidate the molecular mechanism of Nm23-H1 motility suppression, expression microarray analysis of an MDA-MB-435 cancer cell line overexpressing wild-type Nm23-H1 was done and cross-compared with expression profiles from lines expressing the P96S and S120G mutants. Nine genes, MET, PTN, SMO, FZD1, L1CAM, MMP2, NETO2, CTGF, and EDG2, were down-regulated by wild-type but not by mutant Nm23-H1 expression. Reduced expression of these genes coincident with elevated Nm23-H1 expression was observed in human breast tumor cohorts, a panel of breast carcinoma cell lines, and hepatocellular carcinomas from control versus Nm23-M1 knockout mice. The functional significance of the down-regulated genes was assessed by transfection and in vitro motility assays. Only EDG2 overexpression significantly restored motility to Nm23-H1-suppressed cancer cells, enhancing motility by 60-fold in these cells. In addition, silencing EDG2 expression with small interfering RNA reduced the motile phenotype of metastatic breast cancer cells. These data suggest that Nm23-H1 suppresses metastasis, at least in part, through down-regulation of EDG2 expression.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Nucleósido-Difosfato Quinasa/fisiología , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Movimiento Celular , Estudios de Cohortes , Colágeno/metabolismo , Regulación hacia Abajo , Combinación de Medicamentos , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Laminina/metabolismo , Ratones , Ratones Noqueados , Nucleósido Difosfato Quinasas NM23 , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoglicanos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
8.
Cancer Res ; 67(9): 4190-8, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483330

RESUMEN

Retrospective studies of breast cancer patients suggest that primary tumor Her-2 overexpression or trastuzumab therapy is associated with a devastating complication: the development of central nervous system (brain) metastases. Herein, we present Her-2 expression trends from resected human brain metastases and data from an experimental brain metastasis assay, both indicative of a functional contribution of Her-2 to brain metastatic colonization. Of 124 archival resected brain metastases from breast cancer patients, 36.2% overexpressed Her-2, indicating an enrichment in the frequency of tumor Her-2 overexpression at this metastatic site. Using quantitative real-time PCR of laser capture microdissected epithelial cells, Her-2 and epidermal growth factor receptor (EGFR) mRNA levels in a cohort of 12 frozen brain metastases were increased up to 5- and 9-fold, respectively, over those of Her-2-amplified primary tumors. Co-overexpression of Her-2 and EGFR was also observed in a subset of brain metastases. We then tested the hypothesis that overexpression of Her-2 increases the colonization of breast cancer cells in the brain in vivo. A subclone of MDA-MB-231 human breast carcinoma cells that selectively metastasizes to brain (231-BR) overexpressed EGFR; 231-BR cells were transfected with low (4- to 8-fold) or high (22- to 28-fold) levels of Her-2. In vivo, in a model of brain metastasis, low or high Her-2-overexpressing 231-BR clones produced comparable numbers of micrometastases in the brain as control transfectants; however, the Her-2 transfectants yielded 3-fold greater large metastases (>50 microm(2); P < 0.001). Our data indicate that Her-2 overexpression increases the outgrowth of metastatic tumor cells in the brain in this model system.


Asunto(s)
Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Receptor ErbB-2/biosíntesis , Animales , Neoplasias Encefálicas/genética , Neoplasias de la Mama/genética , Adhesión Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor ErbB-2/genética , Transfección , Trasplante Heterólogo
9.
Clin Exp Metastasis ; 25(7): 799-810, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18649117

RESUMEN

Interactions between tumor cells and the microenvironment are crucial to tumor formation and metastasis. The central nervous system serves as a "sanctuary" site for metastasis, resulting in poor prognosis in diagnosed patients. The incidence of brain metastasis is increasing; however, little is known about interactions between the brain and metastatic cells. Brain pathology was examined in an experimental model system of brain metastasis, using a subline of MDA-MB-231 human breast cancer cells. The results were compared with an analysis of sixteen resected human brain metastases of breast cancer. Experimental metastases formed preferentially in specific brain regions, with a distribution similar to clinical cases. In both the 231-BR model, and in human specimens, Ki67 expression indicated that metastases were highly proliferative (approximately 50%). Little apoptosis was observed in either set of tumors. In the model system, metastases elicited a brain inflammatory response, with extensive reactive gliosis surrounding metastases. Similarly, large numbers of glial cells were found within the inner tumor mass of human brain metastases. In vitro co-cultures demonstrated that glia induced a approximately 5-fold increase in metastatic cell proliferation (P<0.001), suggesting that brain tissue secretes factors conducive to tumor cell growth. Molecules used to signal between tumor cells and the surrounding glia could provide a new avenue of therapeutic targets for brain metastases.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Neuroglía/fisiología , Adulto , Anciano , Animales , Apoptosis , Antígeno CD11b/análisis , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Antígenos Comunes de Leucocito/análisis , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad
10.
Mol Cell Biol ; 25(4): 1379-88, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684389

RESUMEN

Metastatic disease is a significant contributor to cancer patient mortality. We previously reported that the Kinase Suppressor of Ras1 (KSR1) scaffold protein for the Erk mitogen-activated protein kinase pathway coimmunoprecipitated the metastasis suppressor protein Nm23-H1. We now hypothesize that altered expression levels of Nm23-H1 influence the binding properties, stability, and function of the KSR1 scaffold. Increased coimmunoprecipitation of Hsp90 with KSR1 was observed in either stable or transient transfectants of nm23-H1 in MDA-MB-435 human breast carcinoma cells. Similar trends were also observed in the cytoplasmic and nuclear fractions of cells. Cells expressing high levels of Nm23-H1 exhibited increased KSR1 degradation in the presence of either cycloheximide or an Hsp90-directed drug currently in clinical trial, 17-allylamino-17-demethoxygeldanamycin (17-AAG). In agreement with KSR1 degradation data, high-Nm23-H1-expression cells were preferentially inhibited in anchorage-independent colonization assays by 17-AAG. KSR1 scaffold binding patterns are dynamic in both the cytoplasmic and nuclear compartments, modulated by metastasis suppressor expression. Metastasis suppressor expression levels can impact traditional signaling pathways, such as the Erk pathway, resulting in altered tumor cell sensitivity to cancer therapeutics.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Nucleósido-Difosfato Quinasa/metabolismo , Proteínas Quinasas/metabolismo , Rifabutina/análogos & derivados , Transducción de Señal/fisiología , Benzoquinonas , Neoplasias de la Mama/metabolismo , Fraccionamiento Celular , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cicloheximida/farmacología , Citoplasma/metabolismo , Femenino , Genes Supresores de Tumor , Humanos , Lactamas Macrocíclicas , Nucleósido Difosfato Quinasas NM23 , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Desnaturalización Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Rifabutina/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
11.
Clin Cancer Res ; 13(6): 1656-62, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363518

RESUMEN

Metastasis to the brain is prevalent in solid tumors and lymphomas, and is associated with shortened survival. The brain is regarded as a sanctuary site for metastatic tumor cells where they exist partially protected from drugs by the blood-tumor barrier. Model systems for brain metastasis have been developed and are now yielding mechanistic insights into the roles of angiogenesis, energy metabolism, the Her-2 and Stat3 signaling pathways, and dormancy. Specific, new approaches to combat brain metastatic disease are needed.


Asunto(s)
Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/secundario , Animales , Encéfalo/irrigación sanguínea , Neoplasias Encefálicas/irrigación sanguínea , Permeabilidad de la Membrana Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo Energético/fisiología , Humanos , Melanoma Experimental/patología , Modelos Biológicos , Neovascularización Patológica/patología , Receptor ErbB-2/fisiología , Factor de Transcripción STAT3/fisiología
12.
Breast Dis ; 26: 139-47, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17473372

RESUMEN

Central nervous system or brain metastases traditionally occur in 10-16% of metastatic breast cancer patients and are associated with a dismal prognosis. The development of brain metastases has been associated with young age, and tumors that are estrogen receptor negative, Her-2+ or of the basal phenotype. Treatment typically includes whole brain irradiation, or either stereotactic radiosurgery or surgery with whole brain radiation, resulting in an approximately 20% one year survival. The blood-brain barrier is a formidable obstacle to the delivery of chemotherapeutics to the brain. Mouse experimental metastasis model systems have been developed for brain metastasis using selected sublines of human MDA-MB-231 breast carcinoma cells. Using micron sized iron particles and MRI imaging, the fate of MDA-MB-231BR cells has been mapped: Approximately 2% of injected cells form larger macroscopic metastases, while 5% of cells remain as dormant cells in the brain. New therapies with permeability for the blood-brain barrier are needed to counteract both types of tumor cells.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Animales , Antineoplásicos , Barrera Hematoencefálica , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/fisiopatología , Diseño de Fármacos , Receptores ErbB/antagonistas & inhibidores , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Receptor ErbB-2/antagonistas & inhibidores , Factores de Riesgo
13.
Clin Cancer Res ; 22(21): 5287-5299, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27245829

RESUMEN

PURPOSE: The blood-brain barrier (BBB) is modified to a blood-tumor barrier (BTB) as a brain metastasis develops from breast or other cancers. We (i) quantified the permeability of experimental brain metastases, (ii) determined the composition of the BTB, and (iii) identified which elements of the BTB distinguished metastases of lower permeability from those with higher permeability. EXPERIMENTAL DESIGN: A SUM190-BR3 experimental inflammatory breast cancer brain metastasis subline was established. Experimental brain metastases from this model system and two previously reported models (triple-negative MDA-231-BR6, HER2+ JIMT-1-BR3) were serially sectioned; low- and high-permeability lesions were identified with systemic 3-kDa Texas Red dextran dye. Adjoining sections were used for quantitative immunofluorescence to known BBB and neuroinflammatory components. One-sample comparisons against a hypothesized value of one were performed with the Wilcoxon signed-rank test. RESULTS: When uninvolved brain was compared with any brain metastasis, alterations in endothelial, pericytic, astrocytic, and microglial components were observed. When metastases with relatively low and high permeability were compared, increased expression of a desmin+ subpopulation of pericytes was associated with higher permeability (231-BR6 P = 0.0002; JIMT-1-BR3 P = 0.004; SUM190-BR3 P = 0.008); desmin+ pericytes were also identified in human craniotomy specimens. Trends of reduced CD13+ pericytes (231-BR6 P = 0.014; JIMT-1-BR3 P = 0.002, SUM190-BR3, NS) and laminin α2 (231-BR6 P = 0.001; JIMT-1-BR3 P = 0.049; SUM190-BR3 P = 0.023) were also observed with increased permeability. CONCLUSIONS: We provide the first account of the composition of the BTB in experimental brain metastasis. Desmin+ pericytes and laminin α2 are potential targets for the development of novel approaches to increase chemotherapeutic efficacy. Clin Cancer Res; 22(21); 5287-99. ©2016 AACR.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Neoplasias Encefálicas/patología , Encéfalo/patología , Neoplasias de la Mama/patología , Pericitos/metabolismo , Pericitos/patología , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Permeabilidad
14.
Clin Exp Metastasis ; 32(7): 717-27, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26319493

RESUMEN

Most cancer patients with brain metastases are treated with radiation therapy, yet this modality has not yet been meaningfully incorporated into preclinical experimental brain metastasis models. We applied two forms of whole brain radiation therapy (WBRT) to the brain-tropic 231-BR experimental brain metastasis model of triple-negative breast cancer. When compared to sham controls, WBRT as 3 Gy × 10 fractions (3 × 10) reduced the number of micrometastases and large metastases by 87.7 and 54.5 %, respectively (both p < 0.01); whereas a single radiation dose of 15 Gy × 1 (15 × 1) was less effective, reducing metastases by 58.4 % (p < 0.01) and 47.1 % (p = 0.41), respectively. Neuroinflammation in the adjacent brain parenchyma was due solely to a reaction from metastases, and not radiotherapy, while adult neurogenesis in brains was adversely affected following both radiation regimens. The nature of radiation resistance was investigated by ex vivo culture of tumor cells that survived initial WBRT ("Surviving" cultures). The Surviving cultures surprisingly demonstrated increased radiosensitivity ex vivo. In contrast, re-injection of Surviving cultures and re-treatment with a 3 × 10 WBRT regimen significantly reduced the number of large and micrometastases that developed in vivo, suggesting a role for the microenvironment. Micrometastases derived from tumor cells surviving initial 3 × 10 WBRT demonstrated a trend toward radioresistance upon repeat treatment (p = 0.09). The data confirm the potency of a fractionated 3 × 10 WBRT regimen and identify the brain microenvironment as a potential determinant of radiation efficacy. The data also nominate the Surviving cultures as a potential new translational model for radiotherapy.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundario , Irradiación Craneana/métodos , Neoplasias de la Mama Triple Negativas/radioterapia , Neoplasias de la Mama Triple Negativas/secundario , Animales , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Desnudos , Tolerancia a Radiación , Dosificación Radioterapéutica , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Clin Exp Metastasis ; 20(1): 3-10, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12650601

RESUMEN

The first metastasis suppressor gene identified was nm23. Transfection of nm23 into metastatic cell lines resulted in the inhibition of metastasis, but not primary tumor size in vivo. Using in vitro assays, nm23 overexpression resulted in reduced anchorage-independent colonization in response to TGF-beta, reduced invasion and motility in response to multiple factors, and increased differentiation. We hypothesize that the mechanism of action of Nm23 in metastasis suppression involves diminished signal transduction downstream of a particular receptor. Candidate biochemical mechanisms are identified and discussed herein.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Unión al GTP Monoméricas/genética , Metástasis de la Neoplasia/genética , Nucleósido-Difosfato Quinasa , Transducción de Señal/genética , Factores de Transcripción/genética , Animales , Diferenciación Celular , Histidina Quinasa , Humanos , Modelos Biológicos , Nucleósido Difosfato Quinasas NM23 , Proteínas Quinasas/metabolismo , Relación Estructura-Actividad , Transfección
16.
Cancer Lett ; 190(1): 1-12, 2003 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-12536071

RESUMEN

Intensive investigation of protein tyrosine, serine and threonine phosphorylation has lead to advances in signal transduction research and cancer treatment. This feature summarizes research on mammalian proteins exhibiting histidine phosphorylation. Histidine kinases are well known in prokaryotic and lower eukaryotic systems where they form the 'two-component' signal transduction system. The relative invisibility of histidine phosphorylation in mammalian cells may result from technical obstacles such as its acid lability, which precludes detection in electrophoretic systems, amino acid sequencing, etc. Emerging data have identified mammalian histidine kinases for the kinase suppressor of ras, a scaffold molecule for the Map kinase pathway, as well as histone H4, aldolase C and the beta-subunit of heterotrimeric G proteins. Additional mammalian proteins of interest to signal transduction and cancer research exhibit histidine phosphorylation, including P-selectin, annexin I and the 20S proteasome. Other candidate histidine phosphorylated proteins are identified. These data suggest the existence of another series of phosphorylation patterns in signal transduction.


Asunto(s)
Histidina/metabolismo , Neoplasias/metabolismo , Nucleósido-Difosfato Quinasa , Proteínas Quinasas/fisiología , Transducción de Señal , Animales , Anexina A1/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Histidina Quinasa , Humanos , Modelos Biológicos , Modelos Químicos , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multienzimáticos/metabolismo , Nucleósido Difosfato Quinasas NM23 , Metástasis de la Neoplasia , Selectina-P/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal , Factores de Transcripción/metabolismo
17.
Clin Breast Cancer ; 4(1): 51-62, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12744759

RESUMEN

Metastatic disease remains a significant contributor to morbidity and mortality in patients with breast cancer. An improved molecular and biochemical understanding of the metastatic process is expected to fuel the development of new therapeutic approaches. The suppression of tumor metastasis, despite tumor cell expression of oncogenes and metastasis-promoting events, has become a diverse and fruitful field of investigation. Although many genetic events promote metastasis, several genes show relatively reduced expression levels in metastatic tumor cells in mouse model systems and in aggressive human tumors. Re-expression of a metastasis-suppressor gene in a metastatic tumor cell line results in a significant reduction in metastatic behavior in vivo with no effect on tumorigenicity. The known metastasis-suppressor gene products nm23, KAI1, mitogen-activated protein kinase kinase 4, breast cancer metastasis suppressor-1, KiSS1, RHOGDI2, CRSP3, and vitamin D3-upregulated protein/thioredoxin interacting protein exhibit unexpected biochemical functions that have shed new light on signaling events that are important in metastasis. Most metastasis suppressors function at the translationally important stage of outgrowth of micrometastatic tumor cells at a distant site. We hypothesize that elevation of metastasis suppressor gene expression in micrometastatic tumor cells in the adjuvant high-risk population of patients with breast cancer will halt metastatic colonization and have a clinical benefit. DNA methylation inhibitors have shown limited promise in increasing metastasis-suppressor gene expression, and ligands of the nuclear hormone receptor family are currently under investigation in vitro and in vivo. Clinical testing of agents that increase metastasis-suppressor gene expression is expected to require tailored trial designs.


Asunto(s)
Neoplasias de la Mama/genética , Genes Supresores de Tumor , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , División Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia/genética
18.
Clin Cancer Res ; 20(10): 2727-39, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634373

RESUMEN

PURPOSE: Brain metastases of breast cancer cause neurocognitive damage and are incurable. We evaluated a role for temozolomide in the prevention of brain metastases of breast cancer in experimental brain metastasis models. EXPERIMENTAL DESIGN: Temozolomide was administered in mice following earlier injection of brain-tropic HER2-positive JIMT-1-BR3 and triple-negative 231-BR-EGFP sublines, the latter with and without expression of O(6)-methylguanine-DNA methyltransferase (MGMT). In addition, the percentage of MGMT-positive tumor cells in 62 patient-matched sets of breast cancer primary tumors and resected brain metastases was determined immunohistochemically. RESULTS: Temozolomide, when dosed at 50, 25, 10, or 5 mg/kg, 5 days per week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cells. At a 1 mg/kg dose, temozolomide prevented 68% of large brain metastases, and was ineffective at a dose of 0.5 mg/kg. When the 50 mg/kg dose was administered beginning on days 18 or 24, temozolomide efficacy was reduced or absent. Temozolomide was ineffective at preventing brain metastases in MGMT-transduced 231-BR-EGFP and MGMT-expressing JIMT-1-BR3 sublines. In 62 patient-matched sets of primary breast tumors and resected brain metastases, 43.5% of the specimens had concordant low MGMT expression, whereas in another 14.5% of sets high MGMT staining in the primary tumor corresponded with low staining in the brain metastasis. CONCLUSIONS: Temozolomide profoundly prevented the outgrowth of experimental brain metastases of breast cancer in an MGMT-dependent manner. These data provide compelling rationale for investigating the preventive efficacy of temozolomide in a clinical setting.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Neoplasias de la Mama/tratamiento farmacológico , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/análogos & derivados , Proteínas Supresoras de Tumor/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Células MCF-7 , Ratones , Ratones Desnudos , Interferencia de ARN , Temozolomida , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Natl Cancer Inst ; 106(7)2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24948741

RESUMEN

BACKGROUND: Breast cancer frequently metastasizes to the brain, colonizing a neuro-inflammatory microenvironment. The molecular pathways facilitating this colonization remain poorly understood. METHODS: Expression profiling of 23 matched sets of human resected brain metastases and primary breast tumors by two-sided paired t test was performed to identify brain metastasis-specific genes. The implicated DNA repair genes BARD1 and RAD51 were modulated in human (MDA-MB-231-BR) and murine (4T1-BR) brain-tropic breast cancer cell lines by lentiviral transduction of cDNA or short hairpin RNA (shRNA) coding sequences. Their functional contribution to brain metastasis development was evaluated in mouse xenograft models (n = 10 mice per group). RESULTS: Human brain metastases overexpressed BARD1 and RAD51 compared with either matched primary tumors (1.74-fold, P < .001; 1.46-fold, P < .001, respectively) or unlinked systemic metastases (1.49-fold, P = .01; 1.44-fold, P = .008, respectively). Overexpression of either gene in MDA-MB-231-BR cells increased brain metastases by threefold to fourfold after intracardiac injections, but not lung metastases upon tail-vein injections. In 4T1-BR cells, shRNA-mediated RAD51 knockdown reduced brain metastases by 2.5-fold without affecting lung metastasis development. In vitro, BARD1- and RAD51-overexpressing cells showed reduced genomic instability but only exhibited growth and colonization phenotypes upon DNA damage induction. Reactive oxygen species were present in tumor cells and elevated in the metastatic neuro-inflammatory microenvironment and could provide an endogenous source of genotoxic stress. Tempol, a brain-permeable oxygen radical scavenger suppressed brain metastasis promotion induced by BARD1 and RAD51 overexpression. CONCLUSIONS: BARD1 and RAD51 are frequently overexpressed in brain metastases from breast cancer and may constitute a mechanism to overcome reactive oxygen species-mediated genotoxic stress in the metastatic brain.


Asunto(s)
Antioxidantes/farmacología , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Óxidos N-Cíclicos/farmacología , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Estrés Oxidativo , Recombinasa Rad51/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/prevención & control , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Fármacos Neuroprotectores/farmacología , Marcadores de Spin , Regulación hacia Arriba
20.
Clin Cancer Res ; 19(23): 6404-18, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24298071

RESUMEN

Despite major therapeutic advances in the management of patients with breast cancer, central nervous system (CNS) metastases remain an intractable problem, particularly in patients with metastatic HER2-positive and triple-negative breast cancer. As systemic therapies to treat extracranial disease improve, some patients are surviving longer, and the frequency of CNS involvement seems to be increasing. Furthermore, in the early-stage setting, the CNS remains a potential sanctuary site for relapse. This review highlights advances in the development of biologically relevant preclinical models, including the development of brain-tropic cell lines for testing of agents to prevent and treat brain metastases, and summarizes our current understanding of the biology of CNS relapse. From a clinical perspective, a variety of therapeutic approaches are discussed, including methods to improve drug delivery, novel cytotoxic agents, and targeted therapies. Challenges in current trial design and endpoints are reviewed. Finally, we discuss promising new directions, including novel trial designs, correlative imaging techniques, and enhanced translational opportunities.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias del Sistema Nervioso Central/secundario , Recurrencia Local de Neoplasia/secundario , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Neoplasias del Sistema Nervioso Central/diagnóstico por imagen , Neoplasias del Sistema Nervioso Central/terapia , Ensayos Clínicos como Asunto , Femenino , Humanos , Recurrencia Local de Neoplasia/terapia , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Cintigrafía , Radiofármacos , Trastuzumab
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA