Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Bioorg Med Chem Lett ; 39: 127904, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33684441

RESUMEN

Free Energy Perturbation (FEP) calculations can provide high-confidence predictions of the interaction strength between a ligand and its protein target. We sought to explore a series of triazolopyrimidines which bind to the EED subunit of the PRC2 complex as potential anticancer therapeutics, using FEP calculations to inform compound design. Combining FEP predictions with a late-stage functionalisation (LSF) inspired synthetic approach allowed us to rapidly evaluate structural modifications in a previously unexplored region of the EED binding site. This approach generated a series of novel triazolopyrimidine EED ligands with improved physicochemical properties and which inhibit PRC2 methyltransferase activity in a cancer-relevant G401 cell line.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Purinas/farmacología , Termodinámica , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ligandos , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Complejo Represivo Polycomb 2/metabolismo , Purinas/síntesis química , Purinas/química , Teoría Cuántica , Relación Estructura-Actividad
2.
J Med Chem ; 64(23): 17146-17183, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34807608

RESUMEN

Aberrant activity of the histone methyltransferase polycomb repressive complex 2 (PRC2) has been linked to several cancers, with small-molecule inhibitors of the catalytic subunit of the PRC2 enhancer of zeste homologue 2 (EZH2) being recently approved for the treatment of epithelioid sarcoma (ES) and follicular lymphoma (FL). Compounds binding to the EED subunit of PRC2 have recently emerged as allosteric inhibitors of PRC2 methyltransferase activity. In contrast to orthosteric inhibitors that target EZH2, small molecules that bind to EED retain their efficacy in EZH2 inhibitor-resistant cell lines. In this paper we disclose the discovery of potent and orally bioavailable EED ligands with good solubilities. The solubility of the EED ligands was optimized through a variety of design tactics, with the resulting compounds exhibiting in vivo efficacy in EZH2-driven tumors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Regulación Alostérica , Animales , Dominio Catalítico , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2/química , Proteína Potenciadora del Homólogo Zeste 2/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Compuestos Heterocíclicos/química , Humanos , Ligandos , Complejo Represivo Polycomb 2/química , Ratas , Relación Estructura-Actividad
3.
Cell Chem Biol ; 27(1): 41-46.e17, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31786184

RESUMEN

Deregulation of the PRC2 complex, comprised of the core subunits EZH2, SUZ12, and EED, drives aberrant hypermethylation of H3K27 and tumorigenicity of many cancers. Although inhibitors of EZH2 have shown promising clinical activity, preclinical data suggest that resistance can be acquired through secondary mutations in EZH2 that abrogate drug target engagement. To address these limitations, we have designed several hetero-bifunctional PROTACs (proteolysis-targeting chimera) to efficiently target EED for elimination. Our PROTACs bind to EED (pKD ∼ 9.0) and promote ternary complex formation with the E3 ubiquitin ligase. The PROTACs potently inhibit PRC2 enzyme activity (pIC50 ∼ 8.1) and induce rapid degradation of not only EED but also EZH2 and SUZ12 within the PRC2 complex. Furthermore, the PROTACs selectively inhibit proliferation of PRC2-dependent cancer cells (half maximal growth inhibition [GI50] = 49-58 nM). In summary, our data demonstrate a therapeutic modality to target PRC2-dependent cancer through a PROTAC-mediated degradation mechanism.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Proteolisis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Relación Estructura-Actividad
4.
Cancer Res ; 77(17): 4613-4625, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28655788

RESUMEN

Through an shRNA screen, we identified the protein arginine methyltransferase Prmt1 as a vulnerable intervention point in murine p53/Rb-null osteosarcomas, the human counterpart of which lacks effective therapeutic options. Depletion of Prmt1 in p53-deficient cells impaired tumor initiation and maintenance in vitro and in vivo Mechanistic studies reveal that translation-associated pathways were enriched for Prmt1 downstream targets, implicating Prmt1 in translation control. In particular, loss of Prmt1 led to a decrease in arginine methylation of the translation initiation complex, thereby disrupting its assembly and inhibiting translation. p53/Rb-null cells were sensitive to p53-induced translation stress, and analysis of human cancer cell line data from Project Achilles further revealed that Prmt1 and translation-associated pathways converged on the same functional networks. We propose that targeted therapy against Prmt1 and its associated translation-related pathways offer a mechanistic rationale for treatment of osteosarcomas and other cancers that exhibit dependencies on translation stress response. Cancer Res; 77(17); 4613-25. ©2017 AACR.


Asunto(s)
Neoplasias Óseas/patología , Osteosarcoma/patología , Biosíntesis de Proteínas , Proteína-Arginina N-Metiltransferasas/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Noqueados , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteómica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Neuro Oncol ; 17(5): 697-707, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25366336

RESUMEN

BACKGROUND: Glioblastoma (GBM) is poorly responsive to current chemotherapy. The nuclear transporter exportin 1 (XPO1, CRM1) is often highly expressed in GBM, which may portend a poor prognosis. Here, we determine the efficacy of novel selective inhibitors of nuclear export (SINE) specific to XPO1 in preclinical models of GBM. METHODS: Seven patient-derived GBM lines were treated with 3 SINE compounds (KPT-251, KPT-276, and Selinexor) in neurosphere culture conditions. KPT-276 and Selinexor were also evaluated in a murine orthotopic patient-derived xenograft (PDX) model of GBM. Cell cycle effects were assayed by flow cytometry in vitro and immunohistochemistry in vivo. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and caspase 3/7 activity assays. RESULTS: Treatment of GBM neurosphere cultures with KPT-276, Selinexor, and KPT-251 revealed dose-responsive growth inhibition in all 7 GBM lines [range of half-maximal inhibitory concentration (IC50), 6-354 nM]. In an orthotopic PDX model, treatment with KPT-276 and Selinexor demonstrated pharmacodynamic efficacy, significantly suppressed tumor growth, and prolonged animal survival. Cellular proliferation was not altered with SINE treatment. Instead, induction of apoptosis was apparent both in vitro and in vivo with SINE treatment, without overt evidence of neurotoxicity. CONCLUSIONS: SINE compounds show preclinical efficacy utilizing in vitro and in vivo models of GBM, with induction of apoptosis as the mechanism of action. Selinexor is now in early clinical trials in solid and hematological malignancies. Based on these preclinical data and excellent brain penetration, we have initiated clinical trials of Selinexor in patients with relapsed GBM.


Asunto(s)
Acrilamidas/uso terapéutico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Hidrazinas/uso terapéutico , Carioferinas/antagonistas & inhibidores , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Tiazoles/uso terapéutico , Triazoles/farmacología , Triazoles/uso terapéutico , Acrilamidas/farmacocinética , Acrilamidas/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Hidrazinas/farmacocinética , Hidrazinas/farmacología , Carioferinas/metabolismo , Macaca fascicularis , Masculino , Ratones , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo , Tiazoles/farmacocinética , Tiazoles/farmacología , Resultado del Tratamiento , Triazoles/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
6.
PLoS One ; 6(4): e19305, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559395

RESUMEN

Ewing sarcoma family tumors (ESFT) are aggressive bone and soft tissue tumors that express EWS-ETS fusion genes as driver mutations. Although the histogenesis of ESFT is controversial, mesenchymal (MSC) and/or neural crest (NCSC) stem cells have been implicated as cells of origin. For the current study we evaluated the consequences of EWS-FLI1 expression in human embryonic stem cell-derived NCSC (hNCSC). Ectopic expression of EWS-FLI1 in undifferentiated hNCSC and their neuro-mesenchymal stem cell (hNC-MSC) progeny was readily tolerated and led to altered expression of both well established as well as novel EWS-FLI1 target genes. Importantly, whole genome expression profiling studies revealed that the molecular signature of established ESFT is more similar to hNCSC than any other normal tissue, including MSC, indicating that maintenance or reactivation of the NCSC program is a feature of ESFT pathogenesis. Consistent with this hypothesis, EWS-FLI1 induced hNCSC genes as well as the polycomb proteins BMI-1 and EZH2 in hNC-MSC. In addition, up-regulation of BMI-1 was associated with avoidance of cellular senescence and reversible silencing of p16. Together these studies confirm that, unlike terminally differentiated cells but consistent with bone marrow-derived MSC, NCSC tolerate expression of EWS-FLI1 and ectopic expression of the oncogene initiates transition to an ESFT-like state. In addition, to our knowledge this is the first demonstration that EWS-FLI1-mediated induction of BMI-1 and epigenetic silencing of p16 might be critical early initiating events in ESFT tumorigenesis.


Asunto(s)
Células Madre Embrionarias/citología , Cresta Neural/citología , Sarcoma de Ewing/diagnóstico , Western Blotting , Diferenciación Celular , Senescencia Celular , Epigénesis Genética , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Lentivirus/metabolismo , Modelos Genéticos , Células-Madre Neurales/citología , Oncogenes
7.
Clin Cancer Res ; 17(1): 56-66, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21047978

RESUMEN

PURPOSE: Ewing sarcoma family tumors (ESFT) are aggressive tumors of putative stem cell origin for which prognostic biomarkers and novel treatments are needed. In several human cancers, high expression of the polycomb protein BMI-1 is associated with poor outcome. We have assessed the potential clinical significance of BMI-1 expression level in ESFT. EXPERIMENTAL DESIGN: BMI-1 expression was assessed in 130 tumors by immunostaining and associations with clinical features and outcome determined. The molecular signatures of BMI-1-low and BMI-1-high tumors were compared using microarrays and differentially activated canonical pathways identified by gene-specific enrichment analysis. Automated quantitative analysis of phosphoproteins was used to assess relative levels of pathway activation. Sensitivity to IGF1-R inhibition was determined using MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assays. RESULTS: BMI-1 is overexpressed by the vast majority of ESFTs. However, in 20% of cases, BMI-1 levels are low to undetectable. Significantly, although clinical presentation and outcome were similar between BMI-1-high and BMI-1-low tumors, whole genome expression array analysis showed marked differences in their respective gene expression profiles. Gene-specific enrichment analysis identified that several cancer-associated canonical biological pathways, including IGF1, mTOR, and WNT, are significantly downregulated in BMI-1-low compared with BMI-1-high tumors. Consistent with these in vivo data, the response to IGF1-R inhibition in vitro was diminished in BMI-1-low compared with BMI-1-high ESFT cells. CONCLUSION: ESFT that do not overexpress BMI-1 represent a novel subclass with a distinct molecular profile and altered activation of and dependence on cancer-associated biological pathways.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Sarcoma de Ewing/clasificación , Sarcoma de Ewing/genética , Adolescente , Adulto , Línea Celular Tumoral , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Fenotipo , Complejo Represivo Polycomb 1 , Pronóstico , ARN Mensajero/genética , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Ewing/diagnóstico , Adulto Joven
8.
Cancer Res ; 68(16): 6507-15, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701473

RESUMEN

Deregulation of the polycomb group gene BMI-1 is implicated in the pathogenesis of many human cancers. In this study, we have investigated if the Ewing sarcoma family of tumors (ESFT) expresses BMI-1 and whether it functions as an oncogene in this highly aggressive group of bone and soft tissue tumors. Our data show that BMI-1 is highly expressed by ESFT cells and that, although it does not significantly affect proliferation or survival, BMI-1 actively promotes anchorage-independent growth in vitro and tumorigenicity in vivo. Moreover, we find that BMI-1 promotes the tumorigenicity of both p16 wild-type and p16-null cell lines, demonstrating that the mechanism of BMI-1 oncogenic function in ESFT is, at least in part, independent of CDKN2A repression. Expression profiling studies of ESFT cells following BMI-1 knockdown reveal that BMI-1 regulates the expression of hundreds of downstream target genes including, in particular, genes involved in both differentiation and development as well as cell-cell and cell-matrix adhesion. Gain and loss of function assays confirm that BMI-1 represses the expression of the adhesion-associated basement membrane protein nidogen 1. In addition, although BMI-1 promotes ESFT adhesion, nidogen 1 inhibits cellular adhesion in vitro. Together, these data support a pivotal role for BMI-1 ESFT pathogenesis and suggest that its oncogenic function in these tumors is in part mediated through modulation of adhesion pathways.


Asunto(s)
Neoplasias Óseas/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Sarcoma de Ewing/patología , Animales , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Adhesión Celular , Proliferación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares/antagonistas & inhibidores , Análisis de Secuencia por Matrices de Oligonucleótidos , Complejo Represivo Polycomb 1 , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Proteínas Represoras/antagonistas & inhibidores , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA