Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Sci Rep ; 10(1): 22155, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335114

RESUMEN

Arginine methylation has been recognized as a post-translational modification with pleiotropic effects that span from regulation of transcription to metabolic processes that contribute to aberrant cell proliferation and tumorigenesis. This has brought significant attention to the development of therapeutic strategies aimed at blocking the activity of protein arginine methyltransferases (PRMTs), which catalyze the formation of various methylated arginine products on a wide variety of cellular substrates. GSK3368715 is a small molecule inhibitor of type I PRMTs currently in clinical development. Here, we evaluate the effect of type I PRMT inhibition on arginine methylation in normal human peripheral blood mononuclear cells and utilize a broad proteomic approach to identify type I PRMT substrates. This work identified heterogenous nuclear ribonucleoprotein A1 (hnRNP-A1) as a pharmacodynamic biomarker of type I PRMT inhibition. Utilizing targeted mass spectrometry (MS), methods were developed to detect and quantitate changes in methylation of specific arginine residues on hnRNP-A1. This resulted in the development and validation of novel MS and immune assays useful for the assessment of GSK3368715 induced pharmacodynamic effects in blood and tumors that can be applied to GSK3368715 clinical trials.


Asunto(s)
Antineoplásicos/farmacocinética , Biomarcadores , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Arginina/metabolismo , Células Cultivadas , Cromatografía Liquida , Monitoreo de Drogas , Activación Enzimática , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ribonucleoproteína Nuclear Heterogénea A1/sangre , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Espectrometría de Masas , Metilación , Ratones , Terapia Molecular Dirigida , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Especificidad por Sustrato
2.
ACS Med Chem Lett ; 11(2): 133-140, 2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32071679

RESUMEN

We report herein the discovery of isoxazole amides as potent and selective SET and MYND Domain-Containing Protein 3 (SMYD3) inhibitors. Elucidation of the structure-activity relationship of the high-throughput screening (HTS) lead compound 1 provided potent and selective SMYD3 inhibitors. The SAR optimization, cocrystal structures of small molecules with SMYD3, and mode of inhibition (MOI) characterization of compounds are described. The synthesis and biological and pharmacokinetic profiles of compounds are also presented.

3.
Nat Genet ; 52(2): 177-186, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32015526

RESUMEN

Lung cancer is the world's leading cause of cancer death and shows strong ancestry disparities. By sequencing and assembling a large genomic and transcriptomic dataset of lung adenocarcinoma (LUAD) in individuals of East Asian ancestry (EAS; n = 305), we found that East Asian LUADs had more stable genomes characterized by fewer mutations and fewer copy number alterations than LUADs from individuals of European ancestry. This difference is much stronger in smokers as compared to nonsmokers. Transcriptomic clustering identified a new EAS-specific LUAD subgroup with a less complex genomic profile and upregulated immune-related genes, allowing the possibility of immunotherapy-based approaches. Integrative analysis across clinical and molecular features showed the importance of molecular phenotypes in patient prognostic stratification. EAS LUADs had better prediction accuracy than those of European ancestry, potentially due to their less complex genomic architecture. This study elucidated a comprehensive genomic landscape of EAS LUADs and highlighted important ancestry differences between the two cohorts.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Neoplasias Pulmonares/genética , Mutación , Adenocarcinoma del Pulmón/etiología , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/terapia , Anciano , Pueblo Asiatico/genética , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Receptores ErbB/genética , Exoma , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas p21(ras)/genética , Singapur , Proteína p53 Supresora de Tumor/genética
4.
Clin Cancer Res ; 25(24): 7331-7339, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31471312

RESUMEN

PURPOSE: Enhancer of zeste homolog 2 (EZH2) activity is dysregulated in many cancers. PATIENTS AND METHODS: This phase I study determined the safety, maximum-tolerated dose (MTD), pharmacokinetics, and pharmacodynamics of the intravenously administered, highly selective EZH2 inhibitor, GSK2816126, (NCT02082977). Doses of GSK2816126 ranged from 50 to 3,000 mg twice weekly, and GSK2816126 was given 3-weeks-on/1-week-off in 28-day cycles. Eligible patients had solid tumors or B-cell lymphomas with no available standard treatment regimen. RESULTS: Forty-one patients (21 solid tumors, 20 lymphoma) received treatment. All patients experienced ≥1 adverse event (AE). Fatigue [22 of 41 (53.7%)] and nausea [20 of 41 (48.8%)] were the most common toxicity. Twelve (32%) patients experienced a serious AE. Dose-limiting elevated liver transaminases occurred in 2 of 7 patients receiving 3,000 mg of GSK2816126; 2,400 mg was therefore established as the MTD. Following intravenous administration of 50 to 3,000 mg twice weekly, plasma GSK2816126 levels decreased biexponentially, with a mean terminal elimination half-life of approximately 27 hours. GSK2816126 exposure (maximum observed plasma concentration and area under the plasma-time curve) increased in a dose-proportional manner. No change from baseline in H3K27me3 was seen in peripheral blood mononuclear cells. Fourteen of 41 (34%) patients had radiological best response of stable disease, 1 patient with lymphoma achieved a partial response, 21 of 41 (51%) patients had progressive disease, and 5 patients were unevaluable for antitumor response. CONCLUSIONS: The MTD of GSK2816126 was established at 2,400 mg, but the dosing method and relatively short half-life limited effective exposure, and modest anticancer activity was observed at tolerable doses.


Asunto(s)
Antineoplásicos/administración & dosificación , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Indoles/administración & dosificación , Linfoma de Células B/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Piridonas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Humanos , Indoles/efectos adversos , Indoles/farmacocinética , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Seguridad del Paciente , Pronóstico , Piridonas/efectos adversos , Piridonas/farmacocinética , Distribución Tisular , Adulto Joven
5.
Cancer Discov ; 9(7): 872-889, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31076479

RESUMEN

Disruption of epigenetic regulation is a hallmark of acute myeloid leukemia (AML), but epigenetic therapy is complicated by the complexity of the epigenome. Herein, we developed a long-term primary AML ex vivo platform to determine whether targeting different epigenetic layers with 5-azacytidine and LSD1 inhibitors would yield improved efficacy. This combination was most effective in TET2 mut AML, where it extinguished leukemia stem cells and particularly induced genes with both LSD1-bound enhancers and cytosine-methylated promoters. Functional studies indicated that derepression of genes such as GATA2 contributes to drug efficacy. Mechanistically, combination therapy increased enhancer-promoter looping and chromatin-activating marks at the GATA2 locus. CRISPRi of the LSD1-bound enhancer in patient-derived TET2 mut AML was associated with dampening of therapeutic GATA2 induction. TET2 knockdown in human hematopoietic stem/progenitor cells induced loss of enhancer 5-hydroxymethylation and facilitated LSD1-mediated enhancer inactivation. Our data provide a basis for rational targeting of cooperating aberrant promoter and enhancer epigenetic marks driven by mutant epigenetic modifiers. SIGNIFICANCE: Somatic mutations of genes encoding epigenetic modifiers are a hallmark of AML and potentially disrupt many components of the epigenome. Our study targets two different epigenetic layers at promoters and enhancers that cooperate to aberrant gene silencing, downstream of the actions of a mutant epigenetic regulator.This article is highlighted in the In This Issue feature, p. 813.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Animales , Azacitidina/farmacología , ADN (Citosina-5-)-Metiltransferasa 1/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Dioxigenasas , Elementos de Facilitación Genéticos , Epigenoma , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Genes Supresores de Tumor , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Distribución Aleatoria , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nat Med ; 25(3): 403-418, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30842676

RESUMEN

Epigenetic dysregulation is a common feature of most cancers, often occurring directly through alteration of epigenetic machinery. Over the last several years, a new generation of drugs directed at epigenetic modulators have entered clinical development, and results from these trials are now being disclosed. Unlike first-generation epigenetic therapies, these new agents are selective, and many are targeted to proteins which are mutated or translocated in cancer. This review will provide a summary of the epigenetic modulatory agents currently in clinical development and discuss the opportunities and challenges in their development. As these drugs advance in the clinic, drug discovery has continued with a focus on both novel and existing epigenetic targets. We will provide an overview of these efforts and the strategies being employed.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética , Neoplasias/tratamiento farmacológico , Proteína-Arginina N-Metiltransferasas/uso terapéutico , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Descubrimiento de Drogas , Histona Acetiltransferasas/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Terapia Molecular Dirigida , Neoplasias/genética
7.
SLAS Discov ; 23(1): 34-46, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28957646

RESUMEN

A persistent problem in early small-molecule drug discovery is the frequent lack of rank-order correlation between biochemical potencies derived from initial screens using purified proteins and the diminished potency and efficacy observed in subsequent disease-relevant cellular phenotypic assays. The introduction of the cellular thermal shift assay (CETSA) has bridged this gap by enabling assessment of drug target engagement directly in live cells based on ligand-induced changes in protein thermal stability. Initial success in applying CETSA across multiple drug target classes motivated our investigation into replacing the low-throughput, manually intensive Western blot readout with a quantitative, automated higher-throughput assay that would provide sufficient capacity to use CETSA as a primary hit qualification strategy. We introduce a high-throughput dose-response cellular thermal shift assay (HTDR-CETSA), a single-pot homogenous assay adapted for high-density microtiter plate format. The assay features titratable BacMam expression of full-length target proteins fused to the DiscoverX 42 amino acid ePL tag in HeLa suspension cells, facilitating enzyme fragment complementation-based chemiluminescent quantification of ligand-stabilized soluble protein. This simplified format can accommodate determination of full-dose CETSA curves for hundreds of individual compounds/analyst/day in replicates. HTDR-CETSA data generated for substrate site and alternate binding mode inhibitors of the histone-lysine N-methyltransferase SMYD3 in HeLa suspension cells demonstrate excellent correlation with rank-order potencies observed in cellular mechanistic assays and direct translation to target engagement of endogenous Smyd3 in cancer-relevant cell lines. We envision this workflow to be generically applicable to HTDR-CETSA screening spanning a wide variety of soluble intracellular protein target classes.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Bibliotecas de Moléculas Pequeñas , Flujo de Trabajo
8.
Cancer Sci ; 109(1): 215-224, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29121415

RESUMEN

Gemcitabine-based therapy remains the mainstay of treatment for patients with biliary tract cancers (BTCs) with no second-line treatment(s) established yet. Aberrant activation of the MAPK pathway in patients with BTC indicates its importance in BTC. Trametinib is a potent, highly selective, allosteric non-competitive inhibitor of MEK1/MEK2. In this phase IIa open-label, single-arm study, we investigated the efficacy and safety of trametinib in Japanese patients with advanced BTC refractory to gemcitabine-based therapy. All patients received oral trametinib 2 mg once daily until progressive disease (PD), death, or unacceptable toxicity. The primary objective was to determine the 12-week non-PD rate. Secondary assessments included safety, progression-free survival (PFS), overall survival, and overall response rate. Targeted exome sequencing was used to identify biomarkers for sensitivity or resistance to trametinib monotherapy. Twenty patients (median age, 61.5 years) with carcinoma of gall bladder (40%), intrahepatic (25%) or extrahepatic (30%) bile duct, and ampulla of Vater (5%) were enrolled. The non-PD rate at week 12 was 10% (95% confidence interval, 1.2-31.7); it did not reach the threshold rate of 25%. Median PFS was 10.6 weeks (95% confidence interval, 4.6-12.1) and 1-year overall survival was 20.0%. Stable disease and PD were observed in 13 (65%) and seven (35%) patients, respectively. No new safety signals were reported. Although the primary end-point was not met, prolonged PFS was observed in one patient having six somatic variants including synonymous NF1 exon 12 splice variant and a loss-of-function variant in ARID1A. Efforts to understand responsive mutations and sensitivity to targeted therapies are warranted. This trial was registered with ClinicalTrials.gov: NCT01943864.


Asunto(s)
Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias del Sistema Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Administración Oral , Anciano , Anciano de 80 o más Años , Ampolla Hepatopancreática/patología , Proteínas de Unión al ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neurofibromina 1/genética , Proteínas Nucleares/genética , Inhibidores de Proteínas Quinasas/efectos adversos , Piridonas/efectos adversos , Pirimidinonas/efectos adversos , Análisis de Supervivencia , Factores de Transcripción/genética , Resultado del Tratamiento , Secuenciación del Exoma
10.
Sci Transl Med ; 9(378)2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28228601

RESUMEN

Trithorax-like group complex containing KDM6A acts antagonistically to Polycomb-repressive complex 2 (PRC2) containing EZH2 in maintaining the dynamics of the repression and activation of gene expression through H3K27 methylation. In urothelial bladder carcinoma, KDM6A (a H3K27 demethylase) is frequently mutated, but its functional consequences and therapeutic targetability remain unknown. About 70% of KDM6A mutations resulted in a total loss of expression and a consequent loss of demethylase function in this cancer type. Further transcriptome analysis found multiple deregulated pathways, especially PRC2/EZH2, in KDM6A-mutated urothelial bladder carcinoma. Chromatin immunoprecipitation sequencing analysis revealed enrichment of H3K27me3 at specific loci in KDM6A-null cells, including PRC2/EZH2 and their downstream targets. Consequently, we targeted EZH2 (an H3K27 methylase) and demonstrated that KDM6A-null urothelial bladder carcinoma cell lines were sensitive to EZH2 inhibition. Loss- and gain-of-function assays confirmed that cells with loss of KDM6A are vulnerable to EZH2. IGFBP3, a direct KDM6A/EZH2/H3K27me3 target, was up-regulated by EZH2 inhibition and contributed to the observed EZH2-dependent growth suppression in KDM6A-null cell lines. EZH2 inhibition delayed tumor onset in KDM6A-null cells and caused regression of KDM6A-null bladder tumors in both patient-derived and cell line xenograft models. In summary, our study demonstrates that inactivating mutations of KDM6A, which are common in urothelial bladder carcinoma, are potentially targetable by inhibiting EZH2.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Transcripción Genética , Neoplasias de la Vejiga Urinaria/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Ratones Desnudos , Modelos Biológicos , Invasividad Neoplásica , Neoplasias de la Vejiga Urinaria/patología , Urotelio/patología
11.
Nat Commun ; 6: 6051, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25609585

RESUMEN

Increased activity of the epigenetic modifier EZH2 has been associated with different cancers. However, evidence for a functional role of EZH2 in tumorigenesis in vivo remains poor, in particular in metastasizing solid cancers. Here we reveal central roles of EZH2 in promoting growth and metastasis of cutaneous melanoma. In a melanoma mouse model, conditional Ezh2 ablation as much as treatment with the preclinical EZH2 inhibitor GSK503 stabilizes the disease through inhibition of growth and virtually abolishes metastases formation without affecting normal melanocyte biology. Comparably, in human melanoma cells, EZH2 inactivation impairs proliferation and invasiveness, accompanied by re-expression of tumour suppressors connected to increased patient survival. These EZH2 target genes suppress either melanoma growth or metastasis in vivo, revealing the dual function of EZH2 in promoting tumour progression. Thus, EZH2-mediated epigenetic repression is highly relevant especially during advanced melanoma progression, which makes EZH2 a promising target for novel melanoma therapies.


Asunto(s)
Silenciador del Gen , Melanoma/metabolismo , Complejo Represivo Polycomb 2/fisiología , Neoplasias Cutáneas/metabolismo , Adenosilmetionina Descarboxilasa/metabolismo , Animales , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Transición Epitelial-Mesenquimal , Femenino , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genotipo , Homeostasis , Humanos , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Complejo Represivo Polycomb 2/genética , Resultado del Tratamiento , Melanoma Cutáneo Maligno
12.
Mol Cancer Ther ; 13(12): 3062-73, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25253781

RESUMEN

The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin lymphoma. Although the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates dimethylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a monomethylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants that dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell-derived cancer cell lines identified an acute lymphoblastic leukemia cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641- or A677-mutant lines. Treatment of A687V EZH2-mutant cells with GSK126, a selective EZH2 inhibitor, was associated with a global decrease in H3K27me3, robust gene activation, caspase activation, and decreased proliferation. Structural modeling of the A687V EZH2 active site suggests that the increased catalytic activity with H3K27me1 may be due to a weakened interaction with an active site water molecule that must be displaced for dimethylation to occur. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival.


Asunto(s)
Histonas/metabolismo , Mutación , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Análisis por Conglomerados , Proteína Potenciadora del Homólogo Zeste 2 , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Heterocigoto , Humanos , Lisina/metabolismo , Metilación , Modelos Moleculares , Datos de Secuencia Molecular , Complejo Represivo Polycomb 2/química , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Conformación Proteica , Alineación de Secuencia , Especificidad por Sustrato , Activación Transcripcional
13.
Epigenomics ; 6(3): 341-51, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25111487

RESUMEN

Over the last several years, dysregulation of epigenetic mechanisms including DNA and histone methylation has been recognized as a hallmark of cancer. Alterations of epigenetic regulators themselves, including the histone lysine methyltransferase EZH2, have been reported in numerous cancer types. With the discovery of small molecule inhibitors of EZH2, we can now begin to evaluate EZH2 as a therapeutic target in cancer. This article will provide an overview of the dysregulation of EZH2 in cancer, possible mechanisms for inhibition of EZH2 activity, and the preclinical activity of currently available EZH2 inhibitors.


Asunto(s)
Neoplasias/metabolismo , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Humanos , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/genética
14.
ACS Chem Biol ; 9(3): 622-9, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24304166

RESUMEN

EZH2/PRC2 catalyzes transcriptionally repressive methylation at lysine 27 of histone H3 and has been associated with numerous cancer types. Point mutations in EZH2 at Tyr641 and Ala677 identified in non-Hodgkin lymphomas alter substrate specificity and result in increased trimethylation at histone H3K27. Interestingly, EZH2/PRC2 is activated by binding H3K27me3 marks on histones, and this activation is proposed as a mechanism for self-propagation of gene silencing. Recent work has identified GSK126 as a potent, selective, SAM-competitive inhibitor of EZH2 capable of globally decreasing H3K27 trimethylation in cells. Here we show that activation of PRC2 by an H3 peptide trimethylated at K27 is primarily an effect on the rate-limiting step (kcat) with no effect on substrate binding (Km). Additionally, GSK126 is shown to have a significantly longer residence time of inhibition on the activated form of EZH2/PRC2 as compared to unactivated EZH2/PRC2. Overall inhibition constant (Ki*) values for GSK126 were determined to be as low as 93 pM and appear to be driven by slow dissociation of inhibitor from the activated enzyme. The data suggest that activation of EZH2 allows the enzyme to adopt a conformation that possesses greater affinity for GSK126. The long residence time of GSK126 may be beneficial in vivo and may result in durable target inhibition after drug systemic clearance.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Piridonas/farmacología , Regulación Alostérica , Sitio Alostérico , Unión Competitiva , Relación Dosis-Respuesta a Droga , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/química , Células HeLa , Humanos , Indoles/química , Metilación , Nucleosomas/efectos de los fármacos , Nucleosomas/enzimología , Mutación Puntual , Complejo Represivo Polycomb 2/genética , Unión Proteica , Piridonas/química , Relación Estructura-Actividad , Especificidad por Sustrato , Factores de Tiempo
15.
Oncotarget ; 4(12): 2419-29, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24293458

RESUMEN

BET (bromodomain and extra-terminal) proteins regulate gene expression through their ability to bind to acetylated chromatin and subsequently activate RNA PolII-driven transcriptional elongation. Small molecule BET inhibitors prevent binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate cell growth and survival in several hematologic cancer models, partially through the down-regulation of the critical oncogene, MYC. We hypothesized that BET inhibitors will regulate MYC expression in solid tumors that frequently over-express MYC. Here we describe the effects of the highly specific BET inhibitor, I-BET762, on MYC expression in prostate cancer models. I-BET762 potently reduced MYC expression in prostate cancer cell lines and a patient-derived tumor model with subsequent inhibition of cell growth and reduction of tumor burden in vivo. Our data suggests that I-BET762 effects are partially driven by MYC down-regulation and underlines the critical importance of additional mechanisms of I-BET762 induced phenotypes.


Asunto(s)
Benzodiazepinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones SCID , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/patología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Cell ; 23(5): 677-92, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23680150

RESUMEN

The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Neoplásica/genética , Centro Germinal/metabolismo , Mutación , Complejo Represivo Polycomb 2/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Centro Germinal/efectos de los fármacos , Histonas/metabolismo , Metilación , Ratones , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/fisiología
17.
J Biol Chem ; 288(12): 8762-8771, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23386615

RESUMEN

The serine/threonine protein kinases Mst1 and Mst2 can be activated by cellular stressors including hydrogen peroxide. Using two independent protein interaction screens, we show that these kinases associate, in an oxidation-dependent manner, with Prdx1, an enzyme that regulates the cellular redox state by reducing hydrogen peroxide to water and oxygen. Mst1 inactivates Prdx1 by phosphorylating it at Thr-90 and Thr-183, leading to accumulation of hydrogen peroxide in cells. These results suggest that hydrogen peroxide-stimulated Mst1 activates a positive feedback loop to sustain an oxidizing cellular state.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Peroxirredoxinas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Daño del ADN , Células HEK293 , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Datos de Secuencia Molecular , Oxidación-Reducción , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Serina-Treonina Quinasa 3 , Técnicas del Sistema de Dos Híbridos
18.
Nature ; 492(7427): 108-12, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-23051747

RESUMEN

In eukaryotes, post-translational modification of histones is critical for regulation of chromatin structure and gene expression. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) and is involved in repressing gene expression through methylation of histone H3 on lysine 27 (H3K27). EZH2 overexpression is implicated in tumorigenesis and correlates with poor prognosis in several tumour types. Additionally, somatic heterozygous mutations of Y641 and A677 residues within the catalytic SET domain of EZH2 occur in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. The Y641 residue is the most frequently mutated residue, with up to 22% of germinal centre B-cell DLBCL and follicular lymphoma harbouring mutations at this site. These lymphomas have increased H3K27 tri-methylation (H3K27me3) owing to altered substrate preferences of the mutant enzymes. However, it is unknown whether specific, direct inhibition of EZH2 methyltransferase activity will be effective in treating EZH2 mutant lymphomas. Here we demonstrate that GSK126, a potent, highly selective, S-adenosyl-methionine-competitive, small-molecule inhibitor of EZH2 methyltransferase activity, decreases global H3K27me3 levels and reactivates silenced PRC2 target genes. GSK126 effectively inhibits the proliferation of EZH2 mutant DLBCL cell lines and markedly inhibits the growth of EZH2 mutant DLBCL xenografts in mice. Together, these data demonstrate that pharmacological inhibition of EZH2 activity may provide a promising treatment for EZH2 mutant lymphoma.


Asunto(s)
Indoles/farmacología , Indoles/uso terapéutico , Linfoma Folicular/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Mutación/genética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Piridonas/farmacología , Piridonas/uso terapéutico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Linfoma Folicular/enzimología , Linfoma Folicular/genética , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Metilación/efectos de los fármacos , Ratones , Trasplante de Neoplasias , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Activación Transcripcional/efectos de los fármacos , Trasplante Heterólogo
19.
J Biomol Screen ; 17(10): 1279-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22904200

RESUMEN

Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [(3)H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Nucleosomas/metabolismo , Péptidos/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Indicadores y Reactivos , Cinética , Péptidos/antagonistas & inhibidores , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/química , Reproducibilidad de los Resultados
20.
Proc Natl Acad Sci U S A ; 109(8): 2989-94, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22323599

RESUMEN

Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive posttranslational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the polycomb repressive complex 2 and is overexpressed in many cancers. In B-cell lymphomas, its substrate preference is frequently altered through somatic mutation of the EZH2 Y641 residue. Herein, we identify mutation of EZH2 A677 to a glycine (A677G) among lymphoma cell lines and primary tumor specimens. Similar to Y641 mutant cell lines, an A677G mutant cell line revealed aberrantly elevated H3K27me3 and decreased monomethylated H3K27 (H3K27me1) and dimethylated H3K27 (H3K27me2). A677G EZH2 possessed catalytic activity with a substrate specificity that was distinct from those of both WT EZH2 and Y641 mutants. Whereas WT EZH2 displayed a preference for substrates with less methylation [unmethylated H3K27 (H3K27me0):me1:me2 k(cat)/K(m) ratio = 9:6:1] and Y641 mutants preferred substrates with greater methylation (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1:2:13), the A677G EZH2 demonstrated nearly equal efficiency for all three substrates (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1.1:0.6:1). When transiently expressed in cells, A677G EZH2, but not WT EZH2, increased global H3K27me3 and decreased H3K27me2. Structural modeling of WT and mutant EZH2 suggested that the A677G mutation acquires the ability to methylate H3K27me2 through enlargement of the lysine tunnel while preserving activity with H3K27me0/me1 substrates through retention of the Y641 residue that is crucial for orientation of these smaller substrates. This mutation highlights the interplay between Y641 and A677 residues in the substrate specificity of EZH2 and identifies another lymphoma patient population that harbors an activating mutation of EZH2.


Asunto(s)
Alanina/genética , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Lisina/metabolismo , Mutación/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Análisis Mutacional de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica , Glicina/genética , Heterocigoto , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Complejo Represivo Polycomb 2 , Especificidad por Sustrato , Factores de Transcripción/química , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...