Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Exp Metastasis ; 33(3): 211-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26585891

RESUMEN

Clinical trials have shown that adjuvant Zoledronic acid (ZOL) reduces the development of bone metastases irrespective of ER status. However, post-menopausal patients show anti-tumour benefit with ZOL whereas pre-menopausal patients do not. Here we have developed in vivo models of spontaneous ER+ve breast cancer metastasis to bone and investigated the effects of ZOL and oestrogen on tumour cell dissemination and growth. ER+ve (MCF7, T47D) or ER-ve (MDA-MB-231) cells were administered by inter-mammary or inter-cardiac injection into female nude mice ± estradiol. Mice were administered saline or 100 µg/kg ZOL weekly. Tumour growth, dissemination of tumour cells in blood, bone and bone turnover were monitored by luciferase imaging, histology, flow cytometry, two-photon microscopy, micro-CT and TRAP/P1NP ELISA. Estradiol induced metastasis of ER+ve cells to bone in 80-100 % of animals whereas bone metastases from ER-ve cells were unaffected. Administration of ZOL had no effect on tumour growth in the fat pad but significantly inhibited dissemination of ER+ve tumour cells to bone and frequency of bone metastasis. Estradiol and ZOL increased bone volume via different mechanisms: Estradiol increased activity of bone forming osteoblasts whereas administration of ZOL to estradiol supplemented mice decreased osteoclast activity and returned osteoblast activity to levels comparable to that of saline treated mice. ER-ve cells require increased osteoclast activity to grow in bone whereas ER+ve cells do not. Zol does not affect ER+ve tumour growth in soft tissue, however, inhibition of bone turnover by ZOL reduced dissemination and growth of ER+ve breast cancer cells in bone.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Neoplasias Óseas/secundario , Remodelación Ósea/efectos de los fármacos , Neoplasias de la Mama/secundario , Difosfonatos/farmacología , Imidazoles/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Estradiol/farmacología , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/patología , Receptores de Estrógenos/metabolismo , Microtomografía por Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico
2.
Cell Oncol (Dordr) ; 36(6): 505-14, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24177992

RESUMEN

PURPOSE: Multiple cell types of the tumour microenvironment, including macrophages, contribute to the response to cancer therapy. The anti-resorptive agent zoledronic acid (ZOL) has anti-tumour effects in vitro and in vivo, but it is not known to what extent macrophages are affected by this agent. We have therefore investigated the effects of ZOL on macrophages using a combination of in vitro and in vivo models. METHODS: J774 macrophages were treated with ZOL in vitro, alone and in combination with doxorubicin (DOX), and the levels of apoptosis and necrosis determined. Uptake of zoledronic acid was assessed by detection of unprenylated Rap1a in J774 macrophages in vitro, in peritoneal macrophages and in macrophage populations isolated from subcutaneously implanted breast cancer xenografts following ZOL treatment in vivo. RESULTS: Exposure of J774 macrophages to 5 µM ZOL for 24 h caused a significant increase in the levels of uRap1A, and higher doses/longer exposure induced apoptotic cell death. DOX (10 nM/24 h) and ZOL (10 µM/4 h) given in sequence induced significantly increased levels of apoptotic cell death compared to single agents. Peritoneal macrophages and macrophage populations isolated from breast tumour xenografts had detectable levels of uRap1A 24 h following a single, clinically achievable dose of 100 µg/kg ZOL in vivo. CONCLUSION: We demonstrate that macrophages are sensitive to sequential administration of DOX and ZOL, and that both peritoneal and breast tumour associated macrophages rapidly take up ZOL in vivo. Our data support that macrophages may contribute to the anti-tumour effect of ZOL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Difosfonatos/farmacología , Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Difosfonatos/administración & dosificación , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Imidazoles/administración & dosificación , Macrófagos/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Prenilación de Proteína/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico , Proteínas de Unión al GTP rap1/metabolismo
3.
Clin Exp Metastasis ; 29(8): 927-38, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22562502

RESUMEN

Bone metastasis is a common incurable complication of breast cancer affecting around 70% of patients with advanced disease. In order to improve outcomes for these patients, the cellular and molecular mechanisms underlying bone metastasis need to be established. The majority of studies to date have focused on end-stage disease and little is known about the events taking place following initial tumour cell colonisation of bone. Here we report the results of a longitudinal study that provides detailed analysis of the spatial and temporal relationship between bone and cancer cells during progression of bone metastasis. Tumour growth in bone was initiated by intra-cardiac inoculation of MDA-MB-231-GFP breast cancer cells in immunocompromised mice. Differentiating between areas of bone in direct contact with the tumour and areas distal to the cancer cells but within the tumour bearing bone, we performed comprehensive analyses of the number and distribution of osteoclasts and osteoblasts. Tumour colonies were detectable in bone from day 10, while reduced trabecular bone volume was apparent from day 19 onwards. Cancer-induced changes in osteoblast and osteoclast numbers differed substantially depending on whether or not the cells were in direct contact with the tumour. Compared to naïve controls, areas of bone in direct contact with the tumour had significantly reduced osteoblast but increased osteoclast numbers, whereas the reverse was found in distal areas. Our data demonstrate that tumour cells induce substantial changes in the bone microenvironment prior to the appearance of bone lesions, suggesting that early therapeutic intervention may be required to oppose the tumour-induced changes to the microenvironment und thus tumour progression.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Comunicación Celular , Osteoblastos/patología , Osteoclastos/patología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/sangre , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Huesos/patología , Recuento de Células , Diferenciación Celular , Línea Celular Tumoral , Tamaño de la Célula , Femenino , Marcadores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ligando RANK/sangre , Microambiente Tumoral
4.
J Bone Oncol ; 1(2): 47-56, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26909255

RESUMEN

We have previously shown that repeated sequential administration of doxorubicin, followed 24 h later by zoledronic acid, inhibits tumour growth in models of established breast cancer bone metastasis. As breast cancer patients only receive zoledronic acid every 3-4 weeks, the aim of the current study was to establish the anti-tumour and bone effects of a single administration of doxorubicin/zoledronic acid combination therapy in a bone metastasis model. MDA-MB-231-GFP cells were injected i.c. in 6-week-old nude mice. On day 2, animals received PBS, doxorubicin (2 mg/kg i.v.), zoledronic acid (100 µg/kg s.c.) or doxorubicin followed 24 h later by zoledronic acid. Anti-tumour effects were assessed on days 15/23 by quantification of apoptotic and proliferating cells and changes in expression of genes implicated in apoptosis, proliferation and bone turnover. Bone effects were assessed by µCT analysis, bone histomorphometry and measurement of serum markers. A tumour-free control group was included. Combination treatment reduced bone tumour burden compared to single agent or PBS control and increased levels of tumour cell apoptosis on day 15, but this was no longer detectable on day 23. Animals receiving zoledronic acid had increased bone density, without evidence of tumour-induced lesions. Bone histomorphometry showed that zoledronic acid caused a decrease in osteoblast and osteoclast numbers and an increase in osteoclast size, in both tumour-free and tumour-bearing animals. Our data show that although zoledronic acid modifies the bone microenvironment through effects on both osteoblasts and osteoclasts, this does not result in a significant anti-tumour effect in the absence of doxorubicin.

5.
Curr Pharm Des ; 16(27): 2988-97, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20722617

RESUMEN

Bisphosphonates are standard treatment for cancer-induced bone disease, a common feature of many advanced malignancies. Traditionally used to inhibit bone turnover and reduce the risk of skeletal-related events, there is now increasing pre-clinical evidence that these agents may also affect tumour burden and disease progression. In particular, combining bisphosphonates with chemotherapeutic agents has been demonstrated to cause substantially increased anti-tumour effects compared to giving the single agents. Clinical studies are in progress to determine whether adding bisphosphonates to standard anti-cancer therapy results in improved outcome for patients. Here we give an overview of the key pre-clinical studies of anti-tumour effects of bisphosphonates, alone and in combination with other agents, and introduce some of the ongoing clinical trials that aim to determine the clinical relevance of bisphosphonates in combination therapy.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Difosfonatos/farmacología , Difosfonatos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/prevención & control , Neoplasias de la Mama/tratamiento farmacológico , Difosfonatos/administración & dosificación , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico
6.
Breast Cancer Res Treat ; 96(2): 101-13, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16319986

RESUMEN

Numerous mouse models of mammary cancer have been developed that mimic selective aspects of human disease. The use of these models has enabled preclinical chemotherapeutic, chemoprevention, and genetic therapy studies in vivo, the testing of gene delivery systems, and the identification of tumour and metastasis suppressor and inducer genes. This review has discussed the most abundantly used murine models of mammary cancer including: spontaneous tumours, chemically induced tumours, orthotopic and syngeneic tumour transplantation, injected tumours, and genetically engineered mice with a predisposition to neoplasia. Each model has been discussed with regards to its merits and limitations for investigating the genetic and phenotypic alterations involved in the human disease as well as its potential usefulness for the development of new treatment strategies. To date no single mouse model is available with the ability to replicate the entire disease process, however, existing models continue to provide invaluable insights into breast cancer induction and progression that would be impossible to obtain using in vitro models alone.


Asunto(s)
Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Transformación Celular Neoplásica , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia
7.
Am J Physiol Gastrointest Liver Physiol ; 288(3): G541-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15486344

RESUMEN

Transgenic mice (hGAS) that overexpress human progastrin are more susceptible than wild-type mice (FVB/N) to the induction of colonic aberrant crypt foci (ACF) and adenomas by the chemical carcinogen azoxymethane. We have previously shown significantly increased levels of colonic mitosis in hGAS compared with FVB/N mice after gamma-radiation. To investigate whether the effects of progastrin observed in hGAS colon require the presence of other forms of circulating gastrin, we have crossed hGAS (hg(+/+)) with gastrin knockout (G(-/-)) mice to generate mice that express progastrin and no murine gastrin (G(-/-)hg(+/+)). After azoxymethane, G(-/-)hg(+/+) mice developed significantly more ACF than control G(-/-)hg(-/-) mice (which do not express any forms of gastrin). G(-/-)hg(+/+) mice also exhibited significantly increased colonic mitosis both before and after exposure to 8 Gray Gy gamma-radiation or 50 mg/kg azoxymethane compared with G(-/-)hg(-/-). Treatment of G(-/-)hg(-/-) mice with synthetic progastrin (residues 21-101 of human preprogastrin) or G17 extended at its COOH terminus corresponding to the COOH-terminal 26-amino-acid residues of human preprogastrin (residues 76-101, G17-CFP) resulted in continued colonic epithelial mitosis after gamma-radiation, whereas glycine-extended gastrin-17 and the COOH-terminal tryptic fragment of progastrin [human preprogastrin-(96-101)] had no effect. Immunoneutralization with an antibody against G17-CFP before gamma-radiation significantly decreased colonic mitosis in G(-/-)hg(+/+) mice to levels similar to G(-/-)hg(-/-). We conclude that progastrin does not require the presence of other forms of gastrin to exert proliferative effects on colonic epithelia and that the portion of the peptide responsible for these effects is contained within amino acid residues 76-101 of human preprogastrin.


Asunto(s)
Colon/citología , Células Epiteliales/efectos de los fármacos , Gastrinas/farmacología , Mitógenos , Mitosis/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/farmacología , Secuencia de Aminoácidos , Animales , Antimetabolitos , Apoptosis/efectos de los fármacos , Azoximetano/farmacología , Bromodesoxiuridina , Carcinógenos/farmacología , Colon/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Rayos gamma , Gastrinas/química , Gastrinas/genética , Genotipo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/química , Precursores de Proteínas/química
8.
J Pathol ; 195(3): 285-92, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11673824

RESUMEN

The aim of this study was to characterize the activation of caspase-3 along the crypt/villus axis in the normal and irradiated intestine and to compare active caspase-3 expression with existing apoptosis detection techniques. Small and large intestine were removed from mice at various time points after exposure to 8 Gy gamma-radiation. Positive apoptotic cells stained with an antibody against active caspase-3, haematoxylin and eosin (H&E) or TUNEL were scored in histological sections of small and large intestinal crypts and villi. In the control intestine, active caspase-3 expression was rarely observed; however, expression was markedly increased following exposure to radiation and was predominantly confined to apoptotic bodies. Measurement of apoptosis in intestinal crypts using active caspase-3 expression gave similar results to apoptosis detected from H&E-stained sections. In the normal villus, active caspase-3 expression was observed infrequently and did not significantly increase following radiation, consistent with a lack of apoptotic body formation from H&E sections. This study indicates that caspase-3 is activated in intestinal crypts but not in villi following gamma-radiation. Active caspase-3 detection compared favourably with existing immunological techniques, suggesting that it is a suitable alternative method for apoptosis quantification.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Intestinos/enzimología , Animales , Anticuerpos Monoclonales , Caspasa 3 , Caspasas/inmunología , Colon/enzimología , Colon/efectos de la radiación , Activación Enzimática , Eosina Amarillenta-(YS) , Epitelio/enzimología , Epitelio/efectos de la radiación , Hematoxilina , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ/métodos , Intestino Delgado/enzimología , Intestino Delgado/efectos de la radiación , Intestinos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos , Coloración y Etiquetado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...