Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cancer ; 22(1): 129, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563568

RESUMEN

BACKGROUND: This Phase 1 study evaluates the intra- and peritumoral administration by convection enhanced delivery (CED) of human recombinant Bone Morphogenetic Protein 4 (hrBMP4) - an inhibitory regulator of cancer stem cells (CSCs) - in recurrent glioblastoma. METHODS: In a 3 + 3 dose escalation design, over four to six days, fifteen recurrent glioblastoma patients received, by CED, one of five doses of hrBMP4 ranging from 0·5 to 18 mg. Patients were followed by periodic physical, neurological, blood testing, magnetic resonance imaging (MRI) and quality of life evaluations. The primary objective of this first-in-human study was to determine the safety, dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) of hrBMP4. Secondary objectives were to assess potential efficacy and systemic exposure to hrBMP4 upon intracerebral infusion. RESULTS: Intra- and peritumoral infusion of hrBMP4 was safe and well-tolerated. We observed no serious adverse events related to this drug. Neither MTD nor DLT were reached. Three patients had increased hrBMP4 serum levels at the end of infusion, which normalized within 4 weeks, without sign of toxicity. One patient showed partial response and two patients a complete (local) tumor response, which was maintained until the most recent follow-up, 57 and 30 months post-hrBMP4. Tumor growth was inhibited in areas permeated by hrBMP4. CONCLUSION: Local delivery of hrBMP4 in and around recurring glioblastoma is safe and well-tolerated. Three patients responded to the treatment. A complete response and long-term survival occurred in two of them. This warrants further clinical studies on this novel treatment targeting glioblastoma CSCs. TRIAL REGISTRATION: ClinicaTrials.gov identifier: NCT02869243.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Calidad de Vida , Proteína Morfogenética Ósea 4/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Encefálicas/patología , Dosis Máxima Tolerada
2.
Mol Metab ; 32: 15-26, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32029225

RESUMEN

OBJECTIVE: Bone morphogenetic protein 4 (BMP4) adeno-associated viral vectors of serotype 8 (AAV8) gene therapy targeting the liver prevents the development of obesity in initially lean mice by browning the large subcutaneous white adipose tissue (WAT) and enhancing energy expenditure. Here, we examine whether this approach could also reduce established obesity. METHODS: Dietary-induced obese C57BL6/N mice received AAV8 BMP4 gene therapy at 17-18 weeks of age. They were kept on a high-fat diet and phenotypically characterized for an additional 10-12 weeks. Following termination, the mice underwent additional characterization in vitro. RESULTS: Surprisingly, we observed no effect on body weight, browning of WAT, or energy expenditure in these obese mice, but whole-body insulin sensitivity and glucose tolerance were robustly improved. Insulin signaling and insulin-stimulated glucose uptake were increased in both adipose cells and skeletal muscle. BMP4 also decreased hepatic glucose production and reduced gluconeogenic enzymes in the liver, but not in the kidney, in addition to enhancing insulin action in the liver. CONCLUSIONS: Our findings show that BMP4 prevents, but does not reverse, established obesity in adult mice, while it improves insulin sensitivity independent of weight reduction. The BMP antagonist Noggin was increased in WAT in obesity, which may account for the lack of browning.


Asunto(s)
Tejido Adiposo Pardo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/uso terapéutico , Terapia Genética , Insulina/metabolismo , Obesidad/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/inducido químicamente , Transducción de Señal
3.
Cancer Res ; 80(6): 1304-1315, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941699

RESUMEN

Metastasis is the major cause of death in patients with cancer; with no therapeutic cure, treatments remain largely palliative. As such, new targets and therapeutic strategies are urgently required. Here, we show that bone morphogenetic protein-4 (BMP4) blocks metastasis in animal models of breast cancer and predicts improved survival in patients. In preclinical models of spontaneous metastasis, BMP4 acted as an autocrine mediator to modulate a range of known metastasis-regulating genes, including Smad7, via activation of canonical BMP-SMAD signaling. Restored BMP4 expression or therapeutically administered BMP4 protein, blocked metastasis and increased survival by sensitizing cancer cells to anoikis, thereby reducing the number of circulating tumor cells. Gene silencing of Bmp4 or its downstream mediator Smad7, reversed this phenotype. Administration of recombinant BMP4 markedly reduced spontaneous metastasis to lung and bone. Elevated levels of BMP4 and SMAD7 were prognostic for improved recurrence-free survival and overall survival in patients with breast cancer, indicating the importance of canonical BMP4 signaling in the suppression of metastasis and highlighting new avenues for therapy against metastatic disease. SIGNIFICANCE: Targeting the BMP4-SMAD7 signaling axis presents a novel therapeutic strategy to combat metastatic breast cancer, a disease that has had no reduction in patient mortality over 20 years. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/6/1304/F1.large.jpg.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Neoplasias Óseas/genética , Neoplasias de la Mama/patología , Proteína smad7/metabolismo , Animales , Comunicación Autocrina , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/uso terapéutico , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Línea Celular Tumoral/trasplante , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Regulación hacia Abajo , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Animales/patología , Mastectomía , Persona de Mediana Edad , Clasificación del Tumor , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Pronóstico , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/genética , Proteína Smad4/metabolismo , Proteína smad7/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Diabetes ; 67(3): 437-447, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29246974

RESUMEN

Polymorphism in TCF7L2, a component of the canonical Wnt signaling pathway, has a strong association with ß-cell dysfunction and type 2 diabetes through a mechanism that has yet to be defined. ß-Cells rely on cells in their microenvironment, including pericytes, for their proper function. Here, we show that Tcf7l2 activity in pancreatic pericytes is required for ß-cell function. Transgenic mice in which Tcf7l2 was selectively inactivated in their pancreatic pericytes exhibited impaired glucose tolerance due to compromised ß-cell function and glucose-stimulated insulin secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of genes required for ß-cell function and maturity in isolated islets. In addition, we identified Tcf7l2-dependent pericytic expression of secreted factors shown to promote ß-cell function, including bone morphogenetic protein 4 (BMP4). Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucose-stimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity affects ß-cells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2-dependent manner to support ß-cell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes and implicates abnormalities in the islet microenvironment in this disease.


Asunto(s)
Comunicación Celular , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Pericitos/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/uso terapéutico , Diferenciación Celular , Microambiente Celular , Glucosa/metabolismo , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Intolerancia a la Glucosa/fisiopatología , Secreción de Insulina , Células Secretoras de Insulina/patología , Ligandos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Transgénicos , Mutación , Pericitos/patología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Técnicas de Cultivo de Tejidos , Proteína 2 Similar al Factor de Transcripción 7/química , Proteína 2 Similar al Factor de Transcripción 7/genética
5.
Int J Nanomedicine ; 11: 4643-4656, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27695323

RESUMEN

Angiogenesis plays a critical role in the growth and metastasis of cancer, and growth factors released from cancer promote blood-vessel formation in the tumor microenvironment. The angiogenesis is accelerated via interactions of growth factors with the high-affinity receptors on cancer cells. In particular, heparan sulfate proteoglycans (HSPGs) on the surface of cancer cells have been shown to be important in many aspects of determining a tumor's phenotype and development. Specifically, the regulation of the interactions between HSPGs and growth factors results in changes in tumor progression. A peptide with heparin-binding (HBP) activity has been developed and synthesized to inhibit tumor growth via the prevention of angiogenesis. We hypothesized that HBP could inhibit the interaction of growth factors and HSPGs on the surface of cancer cells, decrease paracrine signaling in endothelial cells (ECs), and finally decrease angiogenesis in the tumor microenvironment. In this study, we found that HBP had antiangiogenic effects in vitro and in vivo. The conditioned media obtained from a breast cancer cell line treated with HBP were used to culture human umbilical vein ECs (HUVECs) to evaluate the antiangiogenic effect of HBP on ECs. HBP effectively inhibited the migration, invasion, and tube formation of HUVECs in vitro. In addition, the expressions of angiogenesis-mediating factors, including ERK, FAK, and Akt, were considerably decreased. HBP also decreased the levels of invasive factors, including MMP2 and MMP9, secreted by the HUVECs. We demonstrated significant suppression of tumor growth in a breast cancer xenograft model and enhanced distribution of HBP at the site of tumors. Taken together, our results show that HBP has antiangiogenic effects on ECs, and suggest that it may serve as a potential antitumor agent through control of the tumor microenvironment.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Proteína Morfogenética Ósea 4/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Péptidos/uso terapéutico , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/farmacología , Animales , Proteína Morfogenética Ósea 4/química , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Heparina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/metabolismo , Péptidos/química , Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Sus scrofa , Distribución Tisular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Transl Med ; 11: 155, 2013 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-23800258

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is one of the most aggressive forms of cancer with a high rate of recurrence. We propose a novel oncolytic vaccinia virus (VACV)-based therapy using expression of the bone morphogenetic protein (BMP)-4 for treating GBM and preventing recurrence. METHODS: We have utilized clinically relevant, orthotopic xenograft models of GBM based on tumor-biopsy derived, primary cancer stem cell (CSC) lines. One of the cell lines, after being transduced with a cDNA encoding firefly luciferase, could be used for real time tumor imaging. A VACV that expresses BMP-4 was constructed and utilized for infecting several primary glioma cultures besides conventional serum-grown glioma cell lines. This virus was also delivered intracranially upon implantation of the GBM CSCs in mice to determine effects on tumor growth. RESULTS: We found that the VACV that overexpresses BMP-4 demonstrated heightened replication and cytotoxic activity in GBM CSC cultures with a broad spectrum of activity across several different patient-biopsy cultures. Intracranial inoculation of mice with this virus resulted in a tumor size equal to or below that at the time of injection. This resulted in survival of 100% of the treated mice up to 84 days post inoculation, significantly superior to that of a VACV lacking BMP-4 expression. When mice with a higher tumor burden were injected with the VACV lacking BMP-4, 80% of the mice showed tumor recurrence. In contrast, no recurrence was seen when mice were injected with the VACV expressing BMP-4, possibly due to induction of differentiation in the CSC population and subsequently serving as a better host for VACV infection and oncolysis. This lack of recurrence resulted in superior survival in the BMP-4 VACV treated group. CONCLUSIONS: Based on these findings we propose a novel VACV therapy for treating GBM, which would allow tumor specific production of drugs in the future in combination with BMPs which would simultaneously control tumor maintenance and facilitate CSC differentiation, respectively, thereby causing sustained tumor regression without recurrence.


Asunto(s)
Proteína Morfogenética Ósea 4/uso terapéutico , Glioblastoma/tratamiento farmacológico , Virus Vaccinia/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proteína Morfogenética Ósea 4/farmacología , Efecto Espectador/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Glioblastoma/patología , Humanos , Huésped Inmunocomprometido , Masculino , Ratones Desnudos , Invasividad Neoplásica , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Inducción de Remisión , Análisis de Supervivencia , Factores de Tiempo , Replicación Viral/efectos de los fármacos
7.
SADJ ; 67(7): 354-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23951792

RESUMEN

INTRODUCTION: Demineralised bone matrix (DBM) is an allograft material widely used as a bone filler and bone graft substitute. DBM contains bone morphogenetic proteins (BMPs), which induce and regulate bone formation during embryogenesis and in postnatal life. AIMS AND OBJECTIVES: To investigate the osteoinductivity of DBM augmented with xenogeneic BMP-complex at different doses. MATERIALS AND METHODS: Rat DBM was augmented with BMP-complex purified from porcine diaphyseal bone. RESULTS: Dorsal subcutaneous implantation of 25 mg rat allogeneic DBM augmented with 0, 3, 6 and 12 mg BMP-complex per gram of DBM resulted in dose dependant up-regulation of bone formation on day 21, as scored histologically and biochemically. CONCLUSIONS: Allogeneic DBM can be augmented with xenogeneically sourced BMP-complex to improve DBM performance in vivo. This work demonstrates the potential of BMP-complex augmented DBM to induce new bone formation with improved parameters of bone formation.


Asunto(s)
Matriz Ósea/trasplante , Proteínas Morfogenéticas Óseas/uso terapéutico , Osteogénesis/efectos de los fármacos , Fosfatasa Alcalina/análisis , Animales , Proteína Morfogenética Ósea 2/administración & dosificación , Proteína Morfogenética Ósea 2/uso terapéutico , Proteína Morfogenética Ósea 4/administración & dosificación , Proteína Morfogenética Ósea 4/uso terapéutico , Proteína Morfogenética Ósea 7/administración & dosificación , Proteína Morfogenética Ósea 7/uso terapéutico , Proteínas Morfogenéticas Óseas/administración & dosificación , Relación Dosis-Respuesta a Droga , Fascia/patología , Fasciotomía , Masculino , Osificación Heterotópica/patología , Ratas , Ratas Wistar , Tejido Subcutáneo/patología , Tejido Subcutáneo/cirugía , Porcinos , Factor de Crecimiento Transformador beta1/administración & dosificación , Factor de Crecimiento Transformador beta1/uso terapéutico , Trasplante Heterólogo , Regulación hacia Arriba/efectos de los fármacos
8.
Cancer Res ; 71(20): 6547-57, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21868759

RESUMEN

Lymph node metastasis, an early and prognostically important event in the progression of many human cancers, is associated with expression of VEGF-D. Changes to lymph node vasculature that occur during malignant progression may create a metastatic niche capable of attracting and supporting tumor cells. In this study, we sought to characterize molecules expressed in lymph node endothelium that could represent therapeutic or prognostic targets. Differential mRNA expression profiling of endothelial cells from lymph nodes that drained metastatic or nonmetastatic primary tumors revealed genes associated with tumor progression, in particular bone morphogenetic protein-4 (BMP-4). Metastasis driven by VEGF-D was associated with reduced BMP-4 expression in high endothelial venules, where BMP-4 loss could remodel the typical high-walled phenotype to thin-walled vessels. VEGF-D expression was sufficient to suppress proliferation of the more typical BMP-4-expressing high endothelial venules in favor of remodeled vessels, and mechanistic studies indicated that VEGF receptor-2 contributed to high endothelial venule proliferation and remodeling. BMP-4 could regulate high endothelial venule phenotype and cellular function, thereby determining morphology and proliferation responses. Notably, therapeutic administration of BMP-4 suppressed primary tumor growth, acting both at the level of tumor cells and tumor stromal cells. Together, our results show that VEGF-D-driven metastasis induces vascular remodeling in lymph nodes. Furthermore, they implicate BMP-4 as a negative regulator of this process, suggesting its potential utility as a prognostic marker or antitumor agent.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/patología , Neovascularización Patológica/metabolismo , Factor D de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteína Morfogenética Ósea 4/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ganglios Linfáticos/efectos de los fármacos , Metástasis Linfática , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Proc Natl Acad Sci U S A ; 108(19): E99-107, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21518886

RESUMEN

Axon growth potential is highest in young neurons but diminishes with age, thus becoming a significant obstacle to axonal regeneration after injury in maturity. The mechanism for the decline is incompletely understood, and no effective clinical treatment is available to rekindle innate growth capability. Here, we show that Smad1-dependent bone morphogenetic protein (BMP) signaling is developmentally regulated and governs axonal growth in dorsal root ganglion (DRG) neurons. Down-regulation of the pathway contributes to the age-related decline of the axon growth potential. Reactivating Smad1 selectively in adult DRG neurons results in sensory axon regeneration in a mouse model of spinal cord injury (SCI). Smad1 signaling can be effectively manipulated by an adeno-associated virus (AAV) vector encoding BMP4 delivered by a clinically applicable and minimally invasive technique, an approach devoid of unwanted abnormalities in mechanosensation or pain perception. Importantly, transected axons are able to regenerate even when the AAV treatment is delivered after SCI, thus mimicking a clinically relevant scenario. Together, our results identify a therapeutic target to promote axonal regeneration after SCI.


Asunto(s)
Axones/fisiología , Proteína Morfogenética Ósea 4/fisiología , Regeneración Nerviosa/fisiología , Proteína Smad1/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Animales , Axones/patología , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/uso terapéutico , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Neurológicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Transducción de Señal , Proteína Smad1/deficiencia , Proteína Smad1/genética , Traumatismos de la Médula Espinal/patología
10.
Gastroenterology ; 140(1): 297-309, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20951698

RESUMEN

BACKGROUND & AIMS: The limited clinical response observed in many patients with colorectal cancer may be related to the presence of chemoresistant colorectal cancer stem cells (CRC-SCs). Bone morphogenetic protein 4 (BMP4) promotes the differentiation of normal colonic stem cells. We investigated whether BMP4 might be used to induce differentiation of CRC-SCs and for therapeutic purposes. METHODS: CRC-SCs were isolated from 25 tumor samples based on expression of CD133 or using a selection culture medium. BMP4 expression and activity on CRC-SCs were evaluated in vitro; progeny of the stem cells were evaluated by immunofluorescence, immunoblot, and flow cytometry analyses. The potential therapeutic effect of BMP4 was assessed in immunocompromised mice after injection of CRC-SCs that responded to chemotherapy (n = 4) or that did not (n = 2). RESULTS: CRC-SCs did not express BMP4 whereas differentiated cells did. Recombinant BMP4 promoted differentiation and apoptosis of CRC-SCs in 12 of 15 independent experiments; this effect did not depend on Small Mothers against decapentaplegic (Smad)4 expression level or microsatellite stability. BMP4 activated the canonical and noncanonical BMP signaling pathways, including phosphoInositide 3-kinase (PI3K) and PKB (protein kinase B)/AKT. Mutations in PI3K or loss of Phosphatase and Tensin homolog (PTEN) in Smad4-defective tumors made CRC-SCs unresponsive to BMP4. Administration of BMP4 to immunocompromised mice with tumors that arose from CRC-SCs increased the antitumor effects of 5-fluorouracil and oxaliplatin. CONCLUSIONS: BMP4 promotes terminal differentiation, apoptosis, and chemosensitization of CRC-SCs in tumors that do not have simultaneous mutations in Smad4 and constitutive activation of PI3K. BMP4 might be developed as a therapeutic agent against cancer stem cells in advanced colorectal tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteína Morfogenética Ósea 4/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Fluorouracilo/uso terapéutico , Células Madre Neoplásicas/efectos de los fármacos , Compuestos Organoplatinos/uso terapéutico , Antígeno AC133 , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Anciano , Anciano de 80 o más Años , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular , Células Cultivadas , Neoplasias Colorrectales/patología , Femenino , Glicoproteínas/metabolismo , Humanos , Masculino , Ratones , Inestabilidad de Microsatélites , Persona de Mediana Edad , Mutación , Oxaliplatino , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Péptidos/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Smad4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA