Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Cancer ; 125(1): 28-37, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33941878

RESUMEN

BACKGROUND: This Phase 1 study assessed the safety and efficacy of the Porcupine inhibitor, WNT974, in patients with advanced solid tumours. METHODS: Patients (n = 94) received oral WNT974 at doses of 5-30 mg once-daily, plus additional dosing schedules. RESULTS: The maximum tolerated dose was not established; the recommended dose for expansion was 10 mg once-daily. Dysgeusia was the most common adverse event (50% of patients), likely resulting from on-target Wnt pathway inhibition. No responses were seen by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1; 16% of patients had stable disease (median duration 19.9 weeks). AXIN2 expression by RT-PCR was reduced in 94% of paired skin biopsies (n = 52) and 74% of paired tumour biopsies (n = 35), confirming inhibition of the Wnt pathway. In an exploratory analysis, an inverse association was observed between AXIN2 change and immune signature change in paired tumour samples (n = 8). CONCLUSIONS: Single-agent WNT974 treatment was generally well tolerated. Biomarker analyses suggest that WNT974 may influence immune cell recruitment to tumours, and may enhance checkpoint inhibitor activity. CLINICAL TRIAL REGISTRATION: NCT01351103.


Asunto(s)
Proteína Axina/genética , Inhibidores Enzimáticos/administración & dosificación , Neoplasias/tratamiento farmacológico , Pirazinas/administración & dosificación , Piridinas/administración & dosificación , Administración Oral , Adulto , Anciano , Inhibidores Enzimáticos/farmacocinética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/genética , Pirazinas/farmacocinética , Piridinas/farmacocinética , Resultado del Tratamiento , Vía de Señalización Wnt/efectos de los fármacos
2.
Mol Cancer Ther ; 20(7): 1270-1282, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33879555

RESUMEN

The cell surface glycoprotein P-cadherin is highly expressed in a number of malignancies, including those arising in the epithelium of the bladder, breast, esophagus, lung, and upper aerodigestive system. PCA062 is a P-cadherin specific antibody-drug conjugate that utilizes the clinically validated SMCC-DM1 linker payload to mediate potent cytotoxicity in cell lines expressing high levels of P-cadherin in vitro, while displaying no specific activity in P-cadherin-negative cell lines. High cell surface P-cadherin is necessary, but not sufficient, to mediate PCA062 cytotoxicity. In vivo, PCA062 demonstrated high serum stability and a potent ability to induce mitotic arrest. In addition, PCA062 was efficacious in clinically relevant models of P-cadherin-expressing cancers, including breast, esophageal, and head and neck. Preclinical non-human primate toxicology studies demonstrated a favorable safety profile that supports clinical development. Genome-wide CRISPR screens reveal that expression of the multidrug-resistant gene ABCC1 and the lysosomal transporter SLC46A3 differentially impact tumor cell sensitivity to PCA062. The preclinical data presented here suggest that PCA062 may have clinical value for treating patients with multiple cancer types including basal-like breast cancer.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Biomarcadores de Tumor , Cadherinas/genética , Inmunoconjugados/farmacología , Neoplasias/genética , Secuencia de Aminoácidos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacocinética , Sitios de Unión , Cadherinas/química , Cadherinas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Expresión Génica , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Inmunohistoquímica , Macaca fascicularis , Ratones , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Unión Proteica , Transporte de Proteínas , Ratas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Res ; 80(19): 4278-4287, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32747364

RESUMEN

Advanced ovarian cancers are a leading cause of cancer-related death in women and are currently treated with surgery and chemotherapy. This standard of care is often temporarily successful but exhibits a high rate of relapse, after which, treatment options are few. Here we investigate whether biomarker-guided use of multiple targeted therapies, including small molecules and antibody-drug conjugates, is a viable alternative. A panel of patient-derived ovarian cancer xenografts (PDX), similar in genetics and chemotherapy responsiveness to human tumors, was exposed to 21 monotherapies and combination therapies. Three monotherapies and one combination were found to be active in different subsets of PDX. Analysis of gene expression data identified biomarkers associated with responsiveness to each of the three targeted therapies, none of which directly inhibits an oncogenic driver. While no single treatment had as high a response rate as chemotherapy, nearly 90% of PDXs were eligible for and responded to at least one biomarker-guided treatment, including tumors resistant to standard chemotherapy. The distribution of biomarker positivity in The Cancer Genome Atlas data suggests the potential for a similar precision approach in human patients. SIGNIFICANCE: This study exploits a panel of patient-derived xenografts to demonstrate that most ovarian tumors can be matched to effective biomarker-guided treatments.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/genética , Neoplasias Ováricas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Antineoplásicos/farmacología , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/mortalidad , Carcinoma Epitelial de Ovario/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Terapia Molecular Dirigida/métodos , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Medicina de Precisión , Prueba de Estudio Conceptual
4.
Blood ; 129(8): 1008-1020, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28011678

RESUMEN

Tyrosine kinase inhibitor (TKI) treatment of chronic myeloid leukemia (CML) has limited efficacy against leukemia stem cells (LSC) responsible for disease propagation, and most CML patients require continued TKI treatment to maintain remission. LSC maintenance is related, at least in part, to signals from the bone marrow microenvironment (BMM). Our previous studies have shown that Wnt signaling from the BMM contributes to preservation of CML LSC following TKI treatment. Secretion of Wnt ligands requires their modification by the O-acyl transferase Porcupine (PORCN). Here we investigated the activity of a potent and selective PORCN inhibitor, WNT974, against CML stem and progenitor cells. WNT974 efficiently antagonized Wnt signaling in human CML CD34+ cells, and in combination with the TKI nilotinib (NIL) significantly enhanced inhibition of proliferation and colony-forming potential of CML stem and progenitor cells and reduced their growth in immunodeficient mice in vivo, in comparison with NIL alone. Treatment of transgenic CML mice in vivo with NIL in combination with WNT974 significantly reduced leukemic stem and progenitor cell numbers, reduced regeneration of leukemic long-term hematopoietic stem cells in secondary transplant recipients, and enhanced survival of mice after discontinuation of treatment, in comparison with NIL alone. CML progenitors demonstrated enhanced sensitivity to Wnt stimulation, associated with increased expression of the FZD4 receptor. FZD4 knockdown inhibited CML progenitor growth. These results support further investigation of PORCN targeting to inhibit Wnt secretion and signaling and enhance targeting of CML stem cells while sparing their normal counterparts.


Asunto(s)
Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteínas de la Membrana/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Aciltransferasas , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Células Tumorales Cultivadas , Vía de Señalización Wnt/efectos de los fármacos
5.
Nat Med ; 21(11): 1318-25, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26479923

RESUMEN

Profiling candidate therapeutics with limited cancer models during preclinical development hinders predictions of clinical efficacy and identifying factors that underlie heterogeneous patient responses for patient-selection strategies. We established ∼1,000 patient-derived tumor xenograft models (PDXs) with a diverse set of driver mutations. With these PDXs, we performed in vivo compound screens using a 1 × 1 × 1 experimental design (PDX clinical trial or PCT) to assess the population responses to 62 treatments across six indications. We demonstrate both the reproducibility and the clinical translatability of this approach by identifying associations between a genotype and drug response, and established mechanisms of resistance. In addition, our results suggest that PCTs may represent a more accurate approach than cell line models for assessing the clinical potential of some therapeutic modalities. We therefore propose that this experimental paradigm could potentially improve preclinical evaluation of treatment modalities and enhance our ability to predict clinical trial responses.


Asunto(s)
Antineoplásicos/uso terapéutico , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Ratones , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Reproducibilidad de los Resultados , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico
6.
Mol Cancer Ther ; 13(6): 1567-77, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24825858

RESUMEN

B-cell activating factor receptor (BAFF-R) is expressed on precursor B acute lymphoblastic leukemia (pre-B ALL) cells, but not on their pre-B normal counterparts. Thus, selective killing of ALL cells is possible by targeting this receptor. Here, we have further examined therapeutic targeting of pre-B ALL based on the presence of the BAFF-R. Mouse pre-B ALL cells lacking BAFF-R function had comparable viability and proliferation to wild-type cells, but were more sensitive to drug treatment in vitro. Viability of human pre-B ALL cells was further reduced when antibodies to the BAFF-R were combined with other drugs, even in the presence of stromal protection. This indicates that inhibition of BAFF-R function reduces fitness of stressed pre-B ALL cells. We tested a novel humanized anti-BAFF-R monoclonal antibody optimalized for FcRγIII-mediated, antibody-dependent cell killing by effector cells. Antibody binding to human ALL cells was inhibitable, in a dose-dependent manner, by recombinant human BAFF. There was no evidence for internalization of the antibodies. The antibodies significantly stimulated natural killer cell-mediated killing of different human patient-derived ALL cells. Moreover, incubation of such ALL cells with these antibodies stimulated phagocytosis by macrophages. When this was tested in an immunodeficient transplant model, mice that were treated with the antibody had a significantly decreased leukemia burden in bone marrow and spleen. In view of the restricted expression of the BAFF-R on normal cells and the multiple anti-pre-B ALL activities stimulated by this antibody, a further examination of its use for treatment of pre-B ALL is warranted.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Receptor del Factor Activador de Células B/inmunología , Inmunoterapia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Animales , Receptor del Factor Activador de Células B/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/inmunología , Ratones , Terapia Molecular Dirigida , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología
7.
Proc Natl Acad Sci U S A ; 111(8): 3128-33, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24520176

RESUMEN

Defects in epigenetic regulation play a fundamental role in the development of cancer, and epigenetic regulators have recently emerged as promising therapeutic candidates. We therefore set out to systematically interrogate epigenetic cancer dependencies by screening an epigenome-focused deep-coverage design shRNA (DECODER) library across 58 cancer cell lines. This screen identified BRM/SMARCA2, a DNA-dependent ATPase of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex, as being essential for the growth of tumor cells that harbor loss of function mutations in BRG1/SMARCA4. Depletion of BRM in BRG1-deficient cancer cells leads to a cell cycle arrest, induction of senescence, and increased levels of global H3K9me3. We further demonstrate the selective dependency of BRG1-mutant tumors on BRM in vivo. Genetic alterations of the mSWI/SNF chromatin remodeling complexes are the most frequent among chromatin regulators in cancers, with BRG1/SMARCA4 mutations occurring in ∼10-15% of lung adenocarcinomas. Our findings position BRM as an attractive therapeutic target for BRG1 mutated cancers. Because BRG1 and BRM function as mutually exclusive catalytic subunits of the mSWI/SNF complex, we propose that such synthetic lethality may be explained by paralog insufficiency, in which loss of one family member unveils critical dependence on paralogous subunits. This concept of "cancer-selective paralog dependency" may provide a more general strategy for targeting other tumor suppressor lesions/complexes with paralogous subunits.


Asunto(s)
ADN Helicasas/deficiencia , Epigénesis Genética/fisiología , Complejos Multiproteicos/genética , Neoplasias/genética , Proteínas Nucleares/deficiencia , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Western Blotting , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Senescencia Celular/genética , Técnicas de Silenciamiento del Gen , Biblioteca de Genes , Histonas/metabolismo , Humanos , Inmunoprecipitación , Complejos Multiproteicos/metabolismo , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo
8.
Proc Natl Acad Sci U S A ; 110(50): 20224-9, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24277854

RESUMEN

Wnt signaling is one of the key oncogenic pathways in multiple cancers, and targeting this pathway is an attractive therapeutic approach. However, therapeutic success has been limited because of the lack of therapeutic agents for targets in the Wnt pathway and the lack of a defined patient population that would be sensitive to a Wnt inhibitor. We developed a screen for small molecules that block Wnt secretion. This effort led to the discovery of LGK974, a potent and specific small-molecule Porcupine (PORCN) inhibitor. PORCN is a membrane-bound O-acyltransferase that is required for and dedicated to palmitoylation of Wnt ligands, a necessary step in the processing of Wnt ligand secretion. We show that LGK974 potently inhibits Wnt signaling in vitro and in vivo, including reduction of the Wnt-dependent LRP6 phosphorylation and the expression of Wnt target genes, such as AXIN2. LGK974 is potent and efficacious in multiple tumor models at well-tolerated doses in vivo, including murine and rat mechanistic breast cancer models driven by MMTV-Wnt1 and a human head and neck squamous cell carcinoma model (HN30). We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide both a strategy and tools for targeting Wnt-driven cancers through the inhibition of PORCN.


Asunto(s)
Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Pirazinas/farmacología , Piridinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Aciltransferasas , Animales , Proteína Axina/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Clonación Molecular , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Mutagénesis , Fosforilación/efectos de los fármacos , Pirazinas/uso terapéutico , Piridinas/uso terapéutico , Ensayo de Unión Radioligante , Ratas , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Proc Natl Acad Sci U S A ; 110(31): 12649-54, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23847203

RESUMEN

A growing number of agents targeting ligand-induced Wnt/ß-catenin signaling are being developed for cancer therapy. However, clinical development of these molecules is challenging because of the lack of a genetic strategy to identify human tumors dependent on ligand-induced Wnt/ß-catenin signaling. Ubiquitin E3 ligase ring finger 43 (RNF43) has been suggested as a negative regulator of Wnt signaling, and mutations of RNF43 have been identified in various tumors, including cystic pancreatic tumors. However, loss of function study of RNF43 in cell culture has not been conducted, and the functional significance of RNF43 mutations in cancer is unknown. Here, we show that RNF43 inhibits Wnt/ß-catenin signaling by reducing the membrane level of Frizzled in pancreatic cancer cells, serving as a negative feedback mechanism. Inhibition of endogenous Wnt/ß-catenin signaling increased the cell surface level of Frizzled. A panel of 39 pancreatic cancer cell lines was tested for Wnt dependency using LGK974, a selective Porcupine inhibitor being examined in a phase 1 clinical trial. Strikingly, all LGK974-sensitive lines carried inactivating mutations of RNF43. Inhibition of Wnt secretion, depletion of ß-catenin, or expression of wild-type RNF43 blocked proliferation of RNF43 mutant but not RNF43-wild-type pancreatic cancer cells. LGK974 inhibited proliferation and induced differentiation of RNF43-mutant pancreatic adenocarcinoma xenograft models. Our data suggest that mutational inactivation of RNF43 in pancreatic adenocarcinoma confers Wnt dependency, and the presence of RNF43 mutations could be used as a predictive biomarker for patient selection supporting the clinical development of Wnt inhibitors in subtypes of cancer.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas de Unión al ADN/metabolismo , Mutación , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina , Aciltransferasas , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , Proteínas de Unión al ADN/genética , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Ubiquitina-Proteína Ligasas , Proteínas Wnt/genética , Vía de Señalización Wnt
10.
J Med Chem ; 56(13): 5464-72, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23738526

RESUMEN

Glaucoma is a leading cause of vision loss and blindness, with increased intraocular pressure (IOP) a prominent risk factor. IOP can be efficaciously reduced by administration of topical agents. However, the repertoire of approved IOP-lowering drug classes is limited, and effective new alternatives are needed. Agonism of the cannabinoid receptors CB1/2 significantly reduces IOP clinically and experimentally. However, development of CB1/2 agonists has been complicated by the need to avoid cardiovascular and psychotropic side effects. 1 is a potent CB1/2 agonist that is highly excluded from the brain. In a phase I study, compound 1 eyedrops were well tolerated and generated an IOP-lowering trend but were limited in dose and exposure due to poor solubility and ocular absorption. Here we present an innovative strategy to rapidly identify compound 1 prodrugs that are efficiently metabolized to the parent compound for improved solubility and ocular permeability while maintaining low systemic exposures.


Asunto(s)
Soluciones Oftálmicas/farmacología , Profármacos/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB2/agonistas , Animales , Área Bajo la Curva , Ojo/metabolismo , Ojo/fisiopatología , Glaucoma/metabolismo , Glaucoma/fisiopatología , Glaucoma/prevención & control , Humanos , Presión Intraocular/efectos de los fármacos , Masculino , Tasa de Depuración Metabólica , Modelos Químicos , Estructura Molecular , Soluciones Oftálmicas/síntesis química , Soluciones Oftálmicas/farmacocinética , Permeabilidad , Profármacos/síntesis química , Profármacos/farmacocinética , Ratas , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo
11.
Oncotarget ; 4(6): 816-29, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23615731

RESUMEN

The molecular chaperone heat shock protein 90 (HSP90) facilitates the appropriate folding of various oncogenic proteins and is necessary for the survival of some cancer cells. HSP90 is therefore an attractive drug target, but the efficacy of HSP90 inhibitor may be limited by HSP90 inhibition induced feedback mechanisms. Through pooled RNA interference screens, we identified that heat shock factor 1(HSF1) is a sensitizer of HSP90 inhibitor. A striking combinational effect was observed when HSF1 knockdown plus with HSP90 inhibitors treatment in various cancer cell lines and tumor mouse models. Interestingly, HSF1 is highly expressed in hepatocellular carcinoma (HCC) patient samples and HCC is sensitive to combinational treatment, indicating a potential indication for the combinational treatment. To understand the mechanism of the combinational effect, we identified that a HSF1-target gene DEDD2 is involved in attenuating the effect of HSP90 inhibitors. Thus, the transcriptional activities of HSF1 induced by HSP90 inhibitors provide a feedback mechanism of limiting the HSP90 inhibitor's activity, and targeting HSF1 may provide a new avenue to enhance HSP90 inhibitors activity in human cancers.


Asunto(s)
Carcinoma Hepatocelular/terapia , Proteínas de Unión al ADN/genética , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Hepáticas/terapia , Factores de Transcripción/genética , Animales , Apoptosis/fisiología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Doxiciclina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Células HCT116 , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Ratones , Terapia Molecular Dirigida , Proteínas Nucleares/genética , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Proc Natl Acad Sci U S A ; 108(41): 17135-40, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949247

RESUMEN

Persistent expression of certain oncogenes is required for tumor maintenance. This phenotype is referred to as oncogene addiction and has been clinically validated by anticancer therapies that specifically inhibit oncoproteins such as BCR-ABL, c-Kit, HER2, PDGFR, and EGFR. Identifying additional genes that are required for tumor maintenance may lead to new targets for anticancer drugs. Although the role of aberrant Wnt pathway activation in the initiation of colorectal cancer has been clearly established, it remains unclear whether sustained Wnt pathway activation is required for colorectal tumor maintenance. To address this question, we used inducible ß-catenin shRNAs to temporally control Wnt pathway activation in vivo. Here, we show that active Wnt/ß-catenin signaling is required for maintenance of colorectal tumor xenografts harboring APC mutations. Reduced tumor growth upon ß-catenin inhibition was due to cell cycle arrest and differentiation. Upon reactivation of the Wnt/ß-catenin pathway colorectal cancer cells resumed proliferation and reacquired a crypt progenitor phenotype. In human colonic adenocarcinomas, high levels of nuclear ß-catenin correlated with crypt progenitor but not differentiation markers, suggesting that the Wnt/ß-catenin pathway may also control colorectal tumor cell fate during the maintenance phase of tumors in patients. These results support efforts to treat human colorectal cancer by pharmacological inhibition of the Wnt/ß-catenin pathway.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Genes APC , Mutación , Vía de Señalización Wnt , beta Catenina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Interferente Pequeño/genética , Transducción de Señal , Trasplante Heterólogo , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
14.
Cell ; 145(4): 571-83, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565614

RESUMEN

The molecular basis for p53-mediated tumor suppression remains unclear. Here, to elucidate mechanisms of p53 tumor suppression, we use knockin mice expressing an allelic series of p53 transcriptional activation mutants. Microarray analysis reveals that one mutant, p53(25,26), is severely compromised for transactivation of most p53 target genes, and, moreover, p53(25,26) cannot induce G(1)-arrest or apoptosis in response to acute DNA damage. Surprisingly, p53(25,26) retains robust activity in senescence and tumor suppression, indicating that efficient transactivation of the majority of known p53 targets is dispensable for these pathways. In contrast, the transactivation-dead p53(25,26,53,54) mutant cannot induce senescence or inhibit tumorigenesis, like p53 nullizygosity. Thus, p53 transactivation is essential for tumor suppression but, intriguingly, in association with a small set of novel p53 target genes. Together, our studies distinguish the p53 transcriptional programs involved in acute DNA-damage responses and tumor suppression-a critical goal for designing therapeutics that block p53-dependent side effects of chemotherapy without compromising p53 tumor suppression.


Asunto(s)
Reparación del ADN , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Ciclo Celular , Senescencia Celular , Daño del ADN , Técnicas de Sustitución del Gen , Humanos , Ratones , Mutación , Neoplasias/metabolismo , Estructura Terciaria de Proteína , Activación Transcripcional , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética
15.
Arch Pathol Lab Med ; 134(12): 1778-84, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21128775

RESUMEN

CONTEXT: Intraocular melanoma of the ciliary body and choroid is the most common primary ocular malignant tumor in adults and the most common noncutaneous melanoma. OBJECTIVE: To describe the most salient clinical features, histopathologic findings, and treatment modalities of intraocular melanoma, as well as the novel therapies currently being tested. DATA SOURCES: Clinically, it is important to determine which lesions carry a worse prognosis so as to offer patients the best treatment modalities available. Tumor location, size, histopathology, cytogenetic abnormalities, and tumor profiling are all used in determining the risk of death from metastatic disease of uveal melanocytic lesions. Despite successful local tumor control, up to 50% of patients have metastatic disease within 15 years of diagnosis; there is no effective treatment for metastatic disease. CONCLUSIONS: Pathologists should be aware of the importance of tumor gross description, cellular histopathology classification, the use of fine-needle aspiration biopsy coupled with cytogenetics, and the new classification of uveal malignant melanomas that is based on chromosome 3 status.


Asunto(s)
Melanoma/patología , Neoplasias de la Úvea/patología , Anciano , Biopsia con Aguja Fina , Aberraciones Cromosómicas , Femenino , Humanos , Melanoma/genética , Melanoma/terapia , Metástasis de la Neoplasia , Pronóstico , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/terapia
16.
Cell Stem Cell ; 3(4): 416-28, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18940733

RESUMEN

Individual members of the retinoblastoma (Rb) tumor suppressor gene family serve critical roles in the control of cellular proliferation and differentiation, but the extent of their contributions is masked by redundant and compensatory mechanisms. Here we employed a conditional knockout strategy to simultaneously inactivate all three members, Rb, p107, and p130, in adult hematopoietic stem cells (HSCs). Rb family triple knockout (TKO) mice develop a cell-intrinsic myeloproliferation that originates from hyperproliferative early hematopoietic progenitors and is accompanied by increased apoptosis in lymphoid progenitor populations. Loss of quiescence in the TKO HSC pool is associated with an expansion of these mutant stem cells but also with an enhanced mobilization and an impaired reconstitution potential upon transplantation. The presence of a single p107 allele is sufficient to largely rescue these defects. Thus, Rb family members collectively maintain HSC quiescence and the balance between lymphoid and myeloid cell fates in the hematopoietic system.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Madre Hematopoyéticas/metabolismo , Homeostasis/genética , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Animales , Apoptosis/genética , Proliferación Celular , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Trastornos Mieloproliferativos/etiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/genética
17.
Arch Ophthalmol ; 125(3): 389-94, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17353411

RESUMEN

OBJECTIVE: To gain insight into the pathogenesis of neurofibromatosis type 2 (NF2) by investigating the ocular manifestations of this disease. METHODS: Using standard histologic techniques, immunohistochemistry, and electron microscopy, we described the ocular pathologic findings of a 34-year-old woman who died from complications of NF2. RESULTS: We identified 3 types of NF2-associated lesions: juvenile posterior subcapsular cataracts, epiretinal membranes, and an intrascleral schwannoma. CONCLUSIONS: Our analysis indicated that dysplastic lens cells accumulate just anterior to the posterior lens capsule in juvenile posterior subcapsular cataracts and that dysplastic Müller cells may be a major component of NF2-associated epiretinal membranes. Clinical Relevance Our findings suggest that a subset of glial cells with epithelial features (Schwann cells, ependymal cells, and Müller cells) may be particularly sensitive to loss of the NF2 gene. Understanding the molecular basis for this sensitivity may lead to novel strategies for treating NF2.


Asunto(s)
Catarata/patología , Membrana Epirretinal/patología , Neoplasias del Ojo/patología , Neurilemoma/patología , Neurofibromatosis 2/patología , Enfermedades de la Esclerótica/patología , Adulto , Resultado Fatal , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Técnicas para Inmunoenzimas , Queratinas/metabolismo , Mucina-1/metabolismo , Proteínas S100/metabolismo
19.
Proc Natl Acad Sci U S A ; 104(9): 3261-6, 2007 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-17360635

RESUMEN

Tissue fusion, the morphogenic process by which epithelial sheets are drawn together and sealed, has been extensively studied in Drosophila. However, there are unique features of mammalian tissue fusion that remain poorly understood. Notably, detachment and apoptosis occur at the leading front in mammals but not in invertebrates. We found that in the mouse embryo, expression of the Nf2 tumor suppressor, merlin, is dynamically regulated during tissue fusion: Nf2 expression is low at the leading front before fusion and high across the fused tissue bridge. Mosaic Nf2 mutants exhibit a global defect in tissue fusion characterized by ectopic detachment and increased detachment-induced apoptosis (anoikis). By contrast with core components of the junctional complex, we find that merlin is required specifically for the assembly but not the maintenance of the junctional complex. Our work reveals that regulation of Nf2 expression is a previously unrecognized means of controlling adhesion at the leading front, thereby ensuring successful tissue fusion.


Asunto(s)
Adhesión Celular/fisiología , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/fisiología , Neurofibromina 2/metabolismo , Animales , Apoptosis/fisiología , Cartilla de ADN , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/ultraestructura , Epitelio/embriología , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente
20.
Nature ; 445(7128): 661-5, 2007 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-17251932

RESUMEN

Tumorigenesis is a multi-step process that requires activation of oncogenes and inactivation of tumour suppressor genes. Mouse models of human cancers have recently demonstrated that continuous expression of a dominantly acting oncogene (for example, Hras, Kras and Myc) is often required for tumour maintenance; this phenotype is referred to as oncogene addiction. This concept has received clinical validation by the development of active anticancer drugs that specifically inhibit the function of oncoproteins such as BCR-ABL, c-KIT and EGFR. Identifying additional gene mutations that are required for tumour maintenance may therefore yield clinically useful targets for new cancer therapies. Although loss of p53 function is a common feature of human cancers, it is not known whether sustained inactivation of this or other tumour suppressor pathways is required for tumour maintenance. To explore this issue, we developed a Cre-loxP-based strategy to temporally control tumour suppressor gene expression in vivo. Here we show that restoring endogenous p53 expression leads to regression of autochthonous lymphomas and sarcomas in mice without affecting normal tissues. The mechanism responsible for tumour regression is dependent on the tumour type, with the main consequence of p53 restoration being apoptosis in lymphomas and suppression of cell growth with features of cellular senescence in sarcomas. These results support efforts to treat human cancers by way of pharmacological reactivation of p53.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Apoptosis , División Celular , Senescencia Celular , Modelos Animales de Enfermedad , Humanos , Linfoma/metabolismo , Linfoma/patología , Ratones , Neoplasias/tratamiento farmacológico , Especificidad de Órganos , Sarcoma/metabolismo , Sarcoma/patología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...