Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(42): E6409-E6417, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27702896

RESUMEN

Genetically engineered mouse models (GEMMs) of cancer are increasingly being used to assess putative driver mutations identified by large-scale sequencing of human cancer genomes. To accurately interpret experiments that introduce additional mutations, an understanding of the somatic genetic profile and evolution of GEMM tumors is necessary. Here, we performed whole-exome sequencing of tumors from three GEMMs of lung adenocarcinoma driven by mutant epidermal growth factor receptor (EGFR), mutant Kirsten rat sarcoma viral oncogene homolog (Kras), or overexpression of MYC proto-oncogene. Tumors from EGFR- and Kras-driven models exhibited, respectively, 0.02 and 0.07 nonsynonymous mutations per megabase, a dramatically lower average mutational frequency than observed in human lung adenocarcinomas. Tumors from models driven by strong cancer drivers (mutant EGFR and Kras) harbored few mutations in known cancer genes, whereas tumors driven by MYC, a weaker initiating oncogene in the murine lung, acquired recurrent clonal oncogenic Kras mutations. In addition, although EGFR- and Kras-driven models both exhibited recurrent whole-chromosome DNA copy number alterations, the specific chromosomes altered by gain or loss were different in each model. These data demonstrate that GEMM tumors exhibit relatively simple somatic genotypes compared with human cancers of a similar type, making these autochthonous model systems useful for additive engineering approaches to assess the potential of novel mutations on tumorigenesis, cancer progression, and drug sensitivity.


Asunto(s)
Adenocarcinoma/genética , Transformación Celular Neoplásica/genética , Receptores ErbB/genética , Genes myc , Genes ras , Neoplasias Pulmonares/genética , Mutación , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Carcinógenos , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Dosificación de Gen , Estudio de Asociación del Genoma Completo , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Mutación Puntual , Proto-Oncogenes Mas , Curva ROC , Secuenciación del Exoma
2.
Oncotarget ; 10(47): 4822-4839, 2019 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-31448050

RESUMEN

Mesenchymal stem-like (MSL) breast cancers are enriched for cells with tumor reconstituting and mesenchymal characteristics. These cancers are often triple-negative and have a poor prognosis. Few effective targeted treatment options exist for patients with these cancers, and even when targeted therapies exist, resistance often arises and tumors recur, due in part to drug-tolerant persisting tumor cells with self-renewal capability. Effective treatment strategies will combine agents that target the bulk-tumor and reconstituting cells. In order to identify such a combination therapy, we conducted an inhibitor screen using 40 targeted agents at three different doses in all pairwise combinations. Checkpoint Kinase 1 (CHK1) inhibitors were identified as potent inhibitors of MSL breast cancers. When combined with a pro-apoptotic agent/B Cell Lymphoma 2 (BCL2) inhibitor, the effectiveness of the combination regimen was super-additive compared to either treatment alone and was selective for MSL cancers. Treatment of MSL breast cancer cells results in DNA damage, cell-cycle defects characterized by a prolonged S-phase, increased apoptosis and decreased colony forming abilities compared to untreated cells. These data suggest that a combination of a CHK1 and BCL2 inhibitor could be an effective treatment for patients with MSL breast cancer. Several other effective drug combinations were also identified.

3.
Cancer Res ; 77(2): 566-578, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27872098

RESUMEN

Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias de la Mama Triple Negativas , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Humanos , Inmunoprecipitación , Análisis de Componente Principal
4.
Mol Cancer Ther ; 16(6): 1041-1053, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28292938

RESUMEN

Pancreatic adenocarcinoma (PDAC) is the fourth most common cause of cancer-related death in the United States. PDAC is difficult to manage effectively, with a five-year survival rate of only 5%. PDAC is largely driven by activating KRAS mutations, and as such, cannot be directly targeted with therapeutic agents that affect the activated protein. Instead, inhibition of downstream signaling and other targets will be necessary to effectively manage PDAC. Here, we describe a tiered single-agent and combination compound screen to identify targeted agents that impair growth of a panel of PDAC cell lines. Several of the combinations identified from the screen were further validated for efficacy and mechanism. Combination of the bromodomain inhibitor JQ1 and the neddylation inhibitor MLN4294 altered the production of reactive oxygen species in PDAC cells, ultimately leading to defects in the DNA damage response. Dual bromodomain/neddylation blockade inhibited in vivo growth of PDAC cell line xenografts. Overall, this work revealed novel combinatorial regimens, including JQ1 plus MLN4294, which show promise for the treatment of RAS-driven PDAC. Mol Cancer Ther; 16(6); 1041-53. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Sinergismo Farmacológico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Superóxidos , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
5.
Pigment Cell Melanoma Res ; 28(4): 417-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25854919

RESUMEN

BRAF inhibitors have revolutionized treatment of mutant BRAF metastatic melanomas. However, resistance develops rapidly following BRAF inhibitor treatment. We have found that BRAF-mutant melanoma cell lines are more sensitive than wild-type BRAF cells to the small molecule tyrosine kinase inhibitor dovitinib. Sensitivity is associated with inhibition of a series of known dovitinib targets. Dovitinib in combination with several agents inhibits growth more effectively than either agent alone. These combinations inhibit BRAF-mutant melanoma and colorectal carcinoma cell lines, including cell lines with intrinsic or selected BRAF inhibitor resistance. Hence, combinations of dovitinib with second agents are potentially effective therapies for BRAF-mutant melanomas, regardless of their sensitivity to BRAF inhibitors.


Asunto(s)
Bencimidazoles/farmacología , Melanoma/genética , Melanoma/patología , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Quinolonas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Indoles/farmacología , Masculino , Melanoma/enzimología , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Neoplasias Cutáneas , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/farmacología , Vemurafenib , Melanoma Cutáneo Maligno
6.
Oncotarget ; 6(35): 37410-25, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26485762

RESUMEN

Targeting anti-apoptotic proteins can sensitize tumor cells to conventional chemotherapies or other targeted agents. Antagonizing the Inhibitor of Apoptosis Proteins (IAPs) with mimetics of the pro-apoptotic protein SMAC is one such approach. We used sensitization compound screening to uncover possible agents with the potential to further sensitize lung adenocarcinoma cells to the SMAC mimetic Debio 1143. Several compounds in combination with Debio 1143, including taxanes, topoisomerase inhibitors, and bromodomain inhibitors, super-additively inhibited growth and clonogenicity of lung adenocarcinoma cells. Co-treatment with Debio 1143 and the bromodomain inhibitor JQ1 suppresses the expression of c-IAP1, c-IAP2, and XIAP. Non-canonical NF-κB signaling is also activated following Debio 1143 treatment, and Debio 1143 induces the formation of the ripoptosome in Debio 1143-sensitive cell lines. Sensitivity to Debio 1143 and JQ1 co-treatment was associated with baseline caspase-8 expression. In vivo treatment of lung adenocarcinoma xenografts with Debio 1143 in combination with JQ1 or docetaxel reduced tumor volume more than either single agent alone. As Debio 1143-containing combinations effectively inhibited both in vitro and in vivo growth of lung adenocarcinoma cells, these data provide a rationale for Debio 1143 combinations currently being evaluated in ongoing clinical trials and suggest potential utility of other combinations identified here.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Azepinas/farmacología , Azocinas/farmacología , Compuestos de Bencidrilo/farmacología , Camptotecina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/farmacología , Taxoides/farmacología , Inhibidores de Topoisomerasa/farmacología , Triazoles/farmacología , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Camptotecina/farmacología , Línea Celular Tumoral , Docetaxel , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Irinotecán , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 75(7): 1399-412, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25712345

RESUMEN

Melanoma development involves members of the AGC kinase family, including AKT, PKC, and, most recently, PDK1, as elucidated recently in studies of Braf::Pten mutant melanomas. Here, we report that PDK1 contributes functionally to skin pigmentation and to the development of melanomas harboring a wild-type PTEN genotype, which occurs in about 70% of human melanomas. The PDK1 substrate SGK3 was determined to be an important mediator of PDK1 activities in melanoma cells. Genetic or pharmacologic inhibition of PDK1 and SGK3 attenuated melanoma growth by inducing G1 phase cell-cycle arrest. In a synthetic lethal screen, pan-PI3K inhibition synergized with PDK1 inhibition to suppress melanoma growth, suggesting that focused blockade of PDK1/PI3K signaling might offer a new therapeutic modality for wild-type PTEN tumors. We also noted that responsiveness to PDK1 inhibition associated with decreased expression of pigmentation genes and increased expression of cytokines and inflammatory genes, suggesting a method to stratify patients with melanoma for PDK1-based therapies. Overall, our work highlights the potential significance of PDK1 as a therapeutic target to improve melanoma treatment.


Asunto(s)
Melanoma/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/enzimología , Animales , Benzoatos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Indazoles/farmacología , Metástasis Linfática , Melanoma/tratamiento farmacológico , Melanoma/secundario , Ratones Noqueados , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/farmacología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Piel/enzimología , Piel/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología
8.
Cancer Cell ; 27(1): 41-56, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25584893

RESUMEN

Braf(V600E) induces benign, growth-arrested melanocytic nevus development, but also drives melanoma formation. Cdkn2a loss in Braf(V600E) melanocytes in mice results in rare progression to melanoma, but only after stable growth arrest as nevi. Immediate progression to melanoma is prevented by upregulation of miR-99/100, which downregulates mTOR and IGF1R signaling. mTORC1 activation through Stk11 (Lkb1) loss abrogates growth arrest of Braf(V600E) melanocytic nevi, but is insufficient for complete progression to melanoma. Cdkn2a loss is associated with mTORC2 and Akt activation in human and murine melanocytic neoplasms. Simultaneous Cdkn2a and Lkb1 inactivation in Braf(V600E) melanocytes results in activation of both mTORC1 and mTORC2/Akt, inducing rapid melanoma formation in mice. In this model, activation of both mTORC1/2 is required for Braf-induced melanomagenesis.


Asunto(s)
Melanoma Experimental/patología , Complejos Multiproteicos/metabolismo , Nevo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Línea Celular Tumoral , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Mutación , Nevo/patología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Neoplasias Cutáneas/patología
9.
Cancer Gene Ther ; 9(10): 813-8, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12224021

RESUMEN

Salmonella enterica servovar Typhimurium is capable of targeting, colonizing, and eliciting growth suppression of tumors in mice. We examined the effects of mutations on this anticancer phenotype in two Salmonella virulence gene clusters. Salmonella pathogenicity island (SPI)-1 genes promote systemic invasion from the intestine, whereas SPI-2 genes support systemic survival within macrophages and other cells. Disabling SPI-1 (prgH(-)) strongly reduced invasion in vitro, but had no effect on tumor growth suppression in vivo. However, disabling SPI-2 (ssaT(-)) ablated tumor growth suppression. In addition to ssaT(-), mutations in SPI-2 genes sseA, sseB, sseC, sscA, and ssrA also eliminated antitumor activity, whereas mutations in sseF or sseG yielded partial loss of function. Impaired tumor amplification was seen in three SPI-2 mutants tested after intravenous or intratumoral injection. A SPI-2(-) strain was unable to suppress tumor growth in CD18-deficient mice with defective macrophages and neutrophils, suggesting that loss of tumor growth suppression in wild-type mice by SPI-2 mutants was not solely a function of increased susceptibility to immune attack. Thus, SPI-2 is essential for the Salmonella antitumor effects, perhaps by aiding bacterial amplification within tumors, and is the first identified genetic system for this Salmonella phenotype.


Asunto(s)
Proteínas Bacterianas/genética , Melanoma Experimental/prevención & control , Proteínas de la Membrana/genética , Salmonella typhimurium/fisiología , Neoplasias Cutáneas/prevención & control , Animales , Antígenos CD18/metabolismo , División Celular , Femenino , Técnicas In Vitro , Inyecciones Intralesiones , Melanoma Experimental/microbiología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Mutación , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias Cutáneas/microbiología , Neoplasias Cutáneas/patología , Tasa de Supervivencia
10.
Mol Cancer Res ; 12(8): 1140-55, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829397

RESUMEN

UNLABELLED: Associations of ErbB4 (ERBB4/HER4), the fourth member of the EGFR family, with cancer are variable, possibly as a result of structural diversity of this receptor. There are multiple structural isoforms of ERBB4 arising by alternative mRNA splicing, and a subset undergo proteolysis that releases membrane-anchored and soluble isoforms that associate with transcription factors and coregulators to modulate transcription. To compare the differential and common signaling activities of full-length (FL) and soluble intracellular isoforms of ERBB4, four JM-a isoforms (FL and soluble intracellular domain (ICD) CYT-1 and CYT-2) were expressed in isogenic MCF10A cells and their biologic activities were analyzed. Both FL and ICD CYT-2 promoted cell proliferation and invasion, and CYT-1 suppressed cell growth. Transcriptional profiling revealed several new and underexplored ERBB4-regulated transcripts, including: proteases/protease inhibitors (MMP3 and SERPINE2), the YAP/Hippo pathway (CTGF, CYR61, and SPARC), the mevalonate/cholesterol pathway (HMGCR, HMGCS1, LDLR, and DHCR7), and cytokines (IL8, CCL20, and CXCL1). Many of these transcripts were subsequently validated in a luminal breast cancer cell line that normally expresses ERBB4. Furthermore, ChIP-seq experiments identified ADAP1, APOE, SPARC, STMN1, and MXD1 as novel molecular targets of ERBB4. These findings clarify the diverse biologic activities of ERBB4 isoforms, and reveal new and divergent functions. IMPLICATIONS: ErbB4 as a regulator of Hippo and mevalonate pathways provides new insight into milk production and anabolic processes in normal mammary epithelia and cancer.


Asunto(s)
Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Isoformas de Proteínas/metabolismo , Receptor ErbB-4/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Células Cultivadas , Femenino , Humanos , Isoformas de Proteínas/genética , Receptor ErbB-4/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
11.
Pigment Cell Melanoma Res ; 26(4): 527-41, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23617806

RESUMEN

The receptor tyrosine kinase AXL regulates melanoma cell proliferation and migration. We now demonstrate that AXL and the related kinase MERTK are alternately expressed in melanoma and are associated with different transcriptional signatures. MERTK-positive melanoma cells are more proliferative and less migratory than AXL-positive melanoma cells and overexpression of AXL increases cell motility relative to MERTK. MERTK is expressed in up to 50% of melanoma cells and shRNA-mediated knockdown of MERTK reduces colony formation and cell migration in a CDC42-dependent fashion. Targeting MERTK also decreases cell survival and proliferation in an AKT-dependent manner. Finally, we identify a novel mutation in the kinase domain of MERTK, MERTK(P) (802S) , that increases the motility of melanoma cells relative to wild-type MERTK. Together, these data demonstrate that MERTK is a possible therapeutic target in melanoma, that AXL and MERTK are associated with differential cell behaviors, and that mutations in MERTK may contribute to melanoma pathogenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Citofotometría , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Tirosina Quinasa c-Mer , Proteína de Unión al GTP cdc42/metabolismo , Tirosina Quinasa del Receptor Axl
12.
Cancer Discov ; 3(1): 52-67, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23239741

RESUMEN

UNLABELLED: Resistance and partial responses to targeted monotherapy are major obstacles in cancer treatment. Systematic approaches to identify efficacious drug combinations for cancer are not well established, especially in the context of genotype. To address this, we have tested pairwise combinations of an array of small-molecule inhibitors on early-passage melanoma cultures using combinatorial drug screening. Results reveal several inhibitor combinations effective for melanomas with activating RAS or BRAF mutations, including mutant BRAF melanomas with intrinsic or acquired resistance to vemurafenib. Inhibition of both EGF receptor and AKT sensitized treatment-resistant BRAF mutant melanoma cultures to vemurafenib. Melanomas with RAS mutations were more resistant to combination therapies relative to BRAF mutants, but were sensitive to combinations of statins and cyclin-dependent kinase inhibitors in vitro and in vivo. These results show the use of combinatorial drug screening for discovering unique treatment regimens that overcome resistance phenotypes of mutant BRAF- and RAS-driven melanomas. SIGNIFICANCE: We have used drug combinatorial screening to identify effective combinations for mutant BRAF melanomas, including those resistant to vemurafenib, and mutant RAS melanomas that are resistant to many therapies. Mechanisms governing the interactions of the drug combinations are proposed, and in vivo xenografts show the enhanced benefit and tolerability of a mutant RAS -selective combination, which is currently lacking in the clinic.


Asunto(s)
Antineoplásicos/administración & dosificación , Melanoma/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas ras/genética , Animales , Línea Celular Tumoral , Interacciones Farmacológicas , Resistencia a Antineoplásicos , Quimioterapia Combinada , Genes ras/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Melanoma/genética , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Cell ; 20(6): 741-54, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22172720

RESUMEN

Malignant melanoma is characterized by frequent metastasis, however, specific changes that regulate this process have not been clearly delineated. Although it is well known that Wnt signaling is frequently dysregulated in melanoma, the functional implications of this observation are unclear. By modulating ß-catenin levels in a mouse model of melanoma that is based on melanocyte-specific Pten loss and Braf(V600E) mutation, we demonstrate that ß-catenin is a central mediator of melanoma metastasis to the lymph nodes and lungs. In addition to altering metastasis, ß-catenin levels control tumor differentiation and regulate both MAPK/Erk and PI3K/Akt signaling. Highly metastatic tumors with ß-catenin stabilization are very similar to a subset of human melanomas. Together these findings establish Wnt signaling as a metastasis regulator in melanoma.


Asunto(s)
Neoplasias Pulmonares/secundario , Melanoma Experimental/secundario , Fosfohidrolasa PTEN/deficiencia , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología , beta Catenina/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Benzamidas , Transformación Celular Neoplásica , Neoplasias Colorrectales/secundario , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Mesilato de Imatinib , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Piperazinas/uso terapéutico , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/uso terapéutico , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias del Bazo/secundario , Transcripción Genética , Células Tumorales Cultivadas , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA