Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cancer Cell ; 5(3): 221-30, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15050914

RESUMEN

Insulin-like growth factors and their receptor (IGF-1R) have been implicated in cancer pathophysiology. We demonstrate that IGF-1R is universally expressed in various hematologic (multiple myeloma, lymphoma, leukemia) and solid tumor (breast, prostate, lung, colon, thyroid, renal, adrenal cancer, retinoblastoma, and sarcoma) cells. Specific IGF-1R inhibition with neutralizing antibody, antagonistic peptide, or the selective kinase inhibitor NVP-ADW742 has in vitro activity against diverse tumor cell types (particularly multiple myeloma), even those resistant to conventional therapies, and triggers pleiotropic antiproliferative/proapoptotic molecular sequelae, delineated by global transcriptional and proteomic profiling. NVP-ADW742 monotherapy or its combination with cytotoxic chemotherapy had significant antitumor activity in an orthotopic xenograft MM model, providing in vivo proof of principle for therapeutic use of selective IGF-1R inhibitors in cancer.


Asunto(s)
Neoplasias Hematológicas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Antineoplásicos , Médula Ósea/metabolismo , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Perfilación de la Expresión Génica , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Mieloma Múltiple , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Trasplante Heterólogo/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cancer Cell ; 5(3): 231-9, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15050915

RESUMEN

IGF-IR-mediated signaling promotes survival, anchorage-independent growth, and oncogenic transformation, as well as tumor growth and metastasis formation in vivo. NVP-AEW541 is a pyrrolo[2,3-d]pyrimidine derivative small molecular weight kinase inhibitor of the IGF-IR, capable of distinguishing between the IGF-IR (IC50 = 0.086 microM) and the closely related InsR (IC50 = 2.3 microM) in cells. As expected for a specific IGF-IR kinase inhibitor, NVP-AEW541 abrogates IGF-I-mediated survival and colony formation in soft agar at concentrations that are consistent with inhibition of IGF-IR autophosphorylation. In vivo, this orally bioavailable compound inhibits IGF-IR signaling in tumor xenografts and significantly reduces the growth of IGF-IR-driven fibrosarcomas. Thus, NVP-AEW541 represents a class of selective, small molecule IGF-IR kinase inhibitors with proven in vivo antitumor activity and potential therapeutic application.


Asunto(s)
Antineoplásicos/farmacología , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/fisiología , Células 3T3 , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , División Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Fosforilación/efectos de los fármacos , Receptor IGF Tipo 1/efectos de los fármacos , Receptor de Insulina/efectos de los fármacos , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo
3.
Oncoimmunology ; 11(1): 2019466, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154905

RESUMEN

Antibody-mediated cancer immunotherapy targets inhibitory surface molecules, such as PD1, PD-L1, and CTLA-4, aiming to re-invigorate dysfunctional T cells. We purified and characterized tumor-infiltrating lymphocytes (TILs) and their patient-matched non-tumor counterparts from treatment-naïve NSCLC patient biopsies to evaluate the effect of PD1 expression on the functional and molecular profiles of tumor-resident T cells. We show that PD1+ CD8+ TILs have elevated expression of the transcriptional regulator ID3 and that the cytotoxic potential of CD8 T cells can be improved by knocking down ID3, defining it as a potential regulator of T cell effector function. PD1+ CD4+ memory TILs display transcriptional patterns consistent with both helper and regulator function, but can robustly facilitate B cell activation and expansion. Furthermore, we show that expanding ex vivo-prepared TILs in vitro broadly preserves their functionality with respect to tumor cell killing, B cell help, and TCR repertoire. Although purified PD1+ CD8+ TILs generally maintain an exhausted phenotype upon expansion in vitro, transcriptional analysis reveals a downregulation of markers of T-cell dysfunction, including the co-inhibitory molecules PD1 and CTLA-4 and transcription factors ID3, TOX and TOX2, while genes involved in cell cycle and DNA repair are upregulated. We find reduced expression of WNT signaling components to be a hallmark of PD1+ CD8+ exhausted T cells in vivo and in vitro and demonstrate that restoring WNT signaling, by pharmacological blockade of GSK3ß, can improve effector function. These data unveil novel targets for tumor immunotherapy and have promising implications for the development of a personalized TIL-based cell therapy for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Antígeno CTLA-4 , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Receptor de Muerte Celular Programada 1/genética
4.
Proc Natl Acad Sci U S A ; 104(52): 20926-31, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18087038

RESUMEN

Loss of imprinting (LOI) of the insulin-like growth factor-II gene (IGF2), leading to abnormal activation of the normally silent maternal allele, is a common human epigenetic population variant associated with a 5-fold increased frequency of colorectal neoplasia. Here, we show first that LOI leads specifically to increased expression of proliferation-related genes in mouse intestinal crypts. Surprisingly, LOI(+) mice also have enhanced sensitivity to IGF-II signaling, not simply increased IGF-II levels, because in vivo blockade with NVP-AEW541, a specific inhibitor of the IGF-II signaling receptor, showed reduction of proliferation-related gene expression to levels half that seen in LOI(-) mice. Signal transduction assays in microfluidic chips confirmed this enhanced sensitivity with marked augmentation of Akt/PKB signaling in LOI(+) cells at low doses of IGF-II, which was reduced in the presence of the inhibitor to levels below those found in LOI(-) cells, and was associated with increased expression of the IGF1 and insulin receptor genes. We exploited this increased IGF-II sensitivity to develop an in vivo chemopreventive strategy using the azoxymethane (AOM) mutagenesis model. LOI(+) mice treated with AOM showed a 60% increase in premalignant aberrant crypt foci (ACF) formation over LOI(-) mice. In vivo IGF-II blockade with NVP-AEW541 abrogated this effect, reducing ACF to a level 30% lower even than found in exposed LOI(-) mice. Thus, LOI increases cancer risk in a counterintuitive way, by increasing the sensitivity of the IGF-II signaling pathway itself, providing a previously undescribed epigenetic chemoprevention strategy in which cells with LOI are "IGF-II addicted" and undergo reduced tumorigenesis in the colon upon IGF-II pathway blockade.


Asunto(s)
Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Anticarcinógenos/farmacología , Azoximetano/farmacología , Proliferación Celular , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirimidinas/farmacología , Pirroles/farmacología , Transducción de Señal
5.
Life Sci Alliance ; 3(7)2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32467316

RESUMEN

The cohesin subunit STAG2 has emerged as a recurrently inactivated tumor suppressor in human cancers. Using candidate approaches, recent studies have revealed a synthetic lethal interaction between STAG2 and its paralog STAG1 To systematically probe genetic vulnerabilities in the absence of STAG2, we have performed genome-wide CRISPR screens in isogenic cell lines and identified STAG1 as the most prominent and selective dependency of STAG2-deficient cells. Using an inducible degron system, we show that chemical genetic degradation of STAG1 protein results in the loss of sister chromatid cohesion and rapid cell death in STAG2-deficient cells, while sparing STAG2-wild-type cells. Biochemical assays and X-ray crystallography identify STAG1 regions that interact with the RAD21 subunit of the cohesin complex. STAG1 mutations that abrogate this interaction selectively compromise the viability of STAG2-deficient cells. Our work highlights the degradation of STAG1 and inhibition of its interaction with RAD21 as promising therapeutic strategies. These findings lay the groundwork for the development of STAG1-directed small molecules to exploit synthetic lethality in STAG2-mutated tumors.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Neoplasias/genética , Proteínas Nucleares/genética , Mutaciones Letales Sintéticas , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Susceptibilidad a Enfermedades , Silenciador del Gen , Marcación de Gen , Estudio de Asociación del Genoma Completo , Humanos , Modelos Moleculares , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Proteolisis , Relación Estructura-Actividad , Cohesinas
6.
Sci Rep ; 9(1): 11661, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406271

RESUMEN

SMARCA4/BRG1 and SMARCA2/BRM, the two mutually exclusive catalytic subunits of the BAF complex, display a well-established synthetic lethal relationship in SMARCA4-deficient cancers. Using CRISPR-Cas9 screening, we identify SMARCA4 as a novel dependency in SMARCA2-deficient esophageal squamous cell carcinoma (ESCC) models, reciprocal to the known synthetic lethal interaction. Restoration of SMARCA2 expression alleviates the dependency on SMARCA4, while engineered loss of SMARCA2 renders ESCC models vulnerable to concomitant depletion of SMARCA4. Dependency on SMARCA4 is linked to its ATPase activity, but not to bromodomain function. We highlight the relevance of SMARCA4 as a drug target in esophageal cancer using an engineered ESCC cell model harboring a SMARCA4 allele amenable to targeted proteolysis and identify SMARCA4-dependent cell models with low or absent SMARCA2 expression from additional tumor types. These findings expand the concept of SMARCA2/SMARCA4 paralog dependency and suggest that pharmacological inhibition of SMARCA4 represents a novel therapeutic opportunity for SMARCA2-deficient cancers.


Asunto(s)
ADN Helicasas/antagonistas & inhibidores , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/genética , ADN Helicasas/genética , Epigénesis Genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Edición Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Humanos , Mutación con Pérdida de Función , Terapia Molecular Dirigida/métodos , Proteínas Nucleares/genética , ARN Guía de Kinetoplastida/genética , ARN Interferente Pequeño/metabolismo , Mutaciones Letales Sintéticas , Factores de Transcripción/deficiencia
7.
Elife ; 82019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30910006

RESUMEN

Targeted cancer therapy is based on exploiting selective dependencies of tumor cells. By leveraging recent functional screening data of cancer cell lines we identify Werner syndrome helicase (WRN) as a novel specific vulnerability of microsatellite instability-high (MSI-H) cancer cells. MSI, caused by defective mismatch repair (MMR), occurs frequently in colorectal, endometrial and gastric cancers. We demonstrate that WRN inactivation selectively impairs the viability of MSI-H but not microsatellite stable (MSS) colorectal and endometrial cancer cell lines. In MSI-H cells, WRN loss results in severe genome integrity defects. ATP-binding deficient variants of WRN fail to rescue the viability phenotype of WRN-depleted MSI-H cancer cells. Reconstitution and depletion studies indicate that WRN dependence is not attributable to acute loss of MMR gene function but might arise during sustained MMR-deficiency. Our study suggests that pharmacological inhibition of WRN helicase function represents an opportunity to develop a novel targeted therapy for MSI-H cancers.


Asunto(s)
Inestabilidad de Microsatélites , Neoplasias/terapia , Helicasa del Síndrome de Werner/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular , Reparación de la Incompatibilidad de ADN , Humanos , Modelos Teóricos , Helicasa del Síndrome de Werner/genética
9.
J Phys Chem B ; 111(45): 12977-84, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17958412

RESUMEN

Thermal degradation of a filled, cross-linked siloxane material synthesized from poly(dimethylsiloxane) chains of three different average molecular weights and with two different cross-linking species has been studied by (1)H multiple quantum (MQ) NMR methods. Multiple domains of polymer chains were detected by MQ NMR exhibiting residual dipolar coupling () values of 200 and 600 Hz, corresponding to chains with high average molecular weight between cross-links and chains with low average molecular weight between cross-links or near the multifunctional cross-linking sites. Characterization of the values and changes in distributions present in the material were studied as a function of time at 250 degrees C and indicate significant time-dependent degradation. For the domains with low , a broadening in the distribution was observed with aging time. For the domain with high , increases in both the mean and the width in were observed with increasing aging time. Isothermal thermal gravimetric analysis reveals a 3% decrease in weight over 20 h of aging at 250 degrees C. Degraded samples also were analyzed by traditional solid-state (1)H NMR techniques, and off-gassing products were identified by solid-phase microextraction followed by gas chromatography-mass spectrometry. The results, which will be discussed here, suggest that thermal degradation proceeds by complex competition between oxidative chain scissioning and postcuring cross-linking that both contribute to embrittlement.

10.
Elife ; 62017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-28691904

RESUMEN

Recent genome analyses have identified recurrent mutations in the cohesin complex in a wide range of human cancers. Here we demonstrate that the most frequently mutated subunit of the cohesin complex, STAG2, displays a strong synthetic lethal interaction with its paralog STAG1. Mechanistically, STAG1 loss abrogates sister chromatid cohesion in STAG2 mutated but not in wild-type cells leading to mitotic catastrophe, defective cell division and apoptosis. STAG1 inactivation inhibits the proliferation of STAG2 mutated but not wild-type bladder cancer and Ewing sarcoma cell lines. Restoration of STAG2 expression in a mutated bladder cancer model alleviates the dependency on STAG1. Thus, STAG1 and STAG2 support sister chromatid cohesion to redundantly ensure cell survival. STAG1 represents a vulnerability of cancer cells carrying mutations in the major emerging tumor suppressor STAG2 across different cancer contexts. Exploiting synthetic lethal interactions to target recurrent cohesin mutations in cancer, e.g. by inhibiting STAG1, holds the promise for the development of selective therapeutics.


Asunto(s)
Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutaciones Letales Sintéticas , Proteínas de Ciclo Celular , División Celular , Línea Celular Tumoral , Supervivencia Celular , Humanos
11.
Expert Rev Anticancer Ther ; 4(6): 1113-24, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15606337

RESUMEN

The fundamental role of kinases in cancer progression has promoted the development of a plethora of therapeutic inhibitors. Despite the promise of effective treatment with little associated toxicity, the clinical experience with these agents has been mixed. This review will summarize recent advances made in the development of kinase inhibitors to highlight emerging issues and the strategies by which they being addressed.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Neoplasias/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA