Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(19): 7922-7, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23620515

RESUMEN

Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.


Asunto(s)
Apoptosis , Neoplasias/terapia , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Tumor Rabdoide/enzimología , Tumor Rabdoide/genética , Animales , Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Línea Celular Tumoral , Proliferación Celular , Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Perfilación de la Expresión Génica , Células HEK293 , Histonas/metabolismo , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Piridinas/farmacología
2.
Blood ; 121(13): 2533-41, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23361907

RESUMEN

The t(6;11)(q27;q23) is a recurrent chromosomal rearrangement that encodes the MLLAF6 fusion oncoprotein and is observed in patients with diverse hematologic malignancies. The presence of the t(6;11)(q27;q23) has been linked to poor overall survival in patients with AML. In this study, we demonstrate that MLL-AF6 requires continued activity of the histone-methyltransferase DOT1L to maintain expression of the MLL-AF6-driven oncogenic gene-expression program. Using gene-expression analysis and genome-wide chromatin immunoprecipitation studies followed by next generation sequencing, we found that MLL-fusion target genes display markedly high levels of histone 3 at lysine 79 (H3K79) dimethylation in murine MLL-AF6 leukemias as well as in ML2, a human myelomonocytic leukemia cell line bearing the t(6;11)(q27;q23) translocation. Targeted disruption of Dot1l using a conditional knockout mouse model inhibited leukemogenesis mediated by the MLL-AF6 fusion oncogene. Moreover, both murine MLL-AF6-transformed cells as well as the human MLL-AF6-positive ML2 leukemia cell line displayed specific sensitivity to EPZ0004777, a recently described, selective, small-molecule inhibitor of Dot1l. Dot1l inhibition resulted in significantly decreased proliferation, decreased expression of MLL-AF6 target genes, and cell cycle arrest of MLL-AF6-transformed cells. These results indicate that patients bearing the t(6;11)(q27;q23) translocation may benefit from therapeutic agents targeting aberrant H3K79 methylation.


Asunto(s)
Transformación Celular Neoplásica/genética , N-Metiltransferasa de Histona-Lisina/genética , Cinesinas/genética , Metiltransferasas/fisiología , Proteína de la Leucemia Mieloide-Linfoide/genética , Miosinas/genética , Proteínas de Fusión Oncogénica/genética , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Lisina/metabolismo , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Compuestos de Fenilurea/farmacología
3.
Blood ; 122(6): 1017-25, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23801631

RESUMEN

Rearrangements of the MLL gene define a genetically distinct subset of acute leukemias with poor prognosis. Current treatment options are of limited effectiveness; thus, there is a pressing need for new therapies for this disease. Genetic and small molecule inhibitor studies have demonstrated that the histone methyltransferase DOT1L is required for the development and maintenance of MLL-rearranged leukemia in model systems. Here we describe the characterization of EPZ-5676, a potent and selective aminonucleoside inhibitor of DOT1L histone methyltransferase activity. The compound has an inhibition constant value of 80 pM, and demonstrates 37 000-fold selectivity over all other methyltransferases tested. In cellular studies, EPZ-5676 inhibited H3K79 methylation and MLL-fusion target gene expression and demonstrated potent cell killing that was selective for acute leukemia lines bearing MLL translocations. Continuous IV infusion of EPZ-5676 in a rat xenograft model of MLL-rearranged leukemia caused complete tumor regressions that were sustained well beyond the compound infusion period with no significant weight loss or signs of toxicity. EPZ-5676 is therefore a potential treatment of MLL-rearranged leukemia and is under clinical investigation.


Asunto(s)
Antineoplásicos/farmacología , Bencimidazoles/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucemia/genética , Leucemia/terapia , Metiltransferasas/antagonistas & inhibidores , Proteína de la Leucemia Mieloide-Linfoide/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Relación Dosis-Respuesta a Droga , Femenino , Histona Metiltransferasas , Histonas/metabolismo , Humanos , Trasplante de Neoplasias , Conformación Proteica , Ratas , Ratas Desnudas
4.
Nat Chem Biol ; 8(11): 890-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023262

RESUMEN

EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Histonas/metabolismo , Indazoles/farmacología , Linfoma/tratamiento farmacológico , Linfoma/patología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Piridonas/farmacología , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/química , Histonas/química , Humanos , Indazoles/química , Linfoma/enzimología , Linfoma/genética , Lisina/metabolismo , Metilación/efectos de los fármacos , Estructura Molecular , Mutación Puntual , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Piridonas/química , Relación Estructura-Actividad
5.
Biopharm Drug Dispos ; 35(4): 237-52, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24415392

RESUMEN

(2R,3R,4S,5R)-2-(6-Amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol (EPZ-5676) is a novel DOT1L histone methyltransferase inhibitor currently in clinical development for the treatment of MLL-rearranged leukemias. This report describes the preclinical pharmacokinetics and metabolism of EPZ-5676, an aminonucleoside analog with exquisite target potency and selectivity that has shown robust and durable tumor growth inhibition in preclinical models. The in vivo pharmacokinetics in mouse, rat and dog were characterized following i.v. and p.o. administration; EPZ-5676 had moderate to high clearance, low oral bioavailability with a steady-state volume of distribution 2-3 fold higher than total body water. EPZ-5676 showed biexponential kinetics following i.v. administration, giving rise to a terminal elimination half-life (t1/2 ) of 1.1, 3.7 and 13.6 h in mouse, rat and dog, respectively. The corresponding in vitro ADME parameters were also studied and utilized for in vitro-in vivo extrapolation purposes. There was good agreement between the microsomal clearance and the in vivo clearance implicating hepatic oxidative metabolism as the predominant elimination route in preclinical species. Furthermore, low renal clearance was observed in mouse, approximating to fu -corrected glomerular filtration rate (GFR) and thus passive glomerular filtration. The metabolic pathways across species were studied in liver microsomes in which EPZ-5676 was metabolized to three monohydroxylated metabolites (M1, M3 and M5), one N-dealkylated product (M4) as well as an N-oxide (M6).


Asunto(s)
Antineoplásicos/farmacocinética , Bencimidazoles/farmacocinética , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Metiltransferasas/antagonistas & inhibidores , Animales , Antineoplásicos/sangre , Bencimidazoles/sangre , Proteínas Sanguíneas/metabolismo , Perros , Hepatocitos/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Permeabilidad , Ratas Sprague-Dawley
6.
Proc Natl Acad Sci U S A ; 107(49): 20980-5, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21078963

RESUMEN

EZH2, the catalytic subunit of the PRC2 complex, catalyzes the mono- through trimethylation of lysine 27 on histone H3 (H3K27). Histone H3K27 trimethylation is a mechanism for suppressing transcription of specific genes that are proximal to the site of histone modification. Point mutations of the EZH2 gene (Tyr641) have been reported to be linked to subsets of human B-cell lymphoma. The mutant allele is always found associated with a wild-type allele (heterozygous) in disease cells, and the mutations were reported to ablate the enzymatic activity of the PRC2 complex for methylating an unmodified peptide substrate. Here we demonstrate that the WT enzyme displays greatest catalytic efficiency (k(cat)/K) for the zero to monomethylation reaction of H3K27 and diminished efficiency for subsequent (mono- to di- and di- to trimethylation) reactions. In stark contrast, the disease-associated Y641 mutations display very limited ability to perform the first methylation reaction, but have enhanced catalytic efficiency for the subsequent reactions, relative to the WT enzyme. These results imply that the malignant phenotype of disease requires the combined activities of a H3K27 monomethylating enzyme (PRC2 containing WT EZH2 or EZH1) together with the mutant PRC2s for augmented conversion of H3K27 to the trimethylated form. To our knowledge, this is the first example of a human disease that is dependent on the coordinated activities of normal and disease-associated mutant enzymatic function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Linfoma de Células B/genética , Lisina/metabolismo , Mutación Puntual , Factores de Transcripción/metabolismo , Catálisis , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Cinética , Linfoma de Células B/enzimología , Linfoma de Células B/etiología , Metilación , Complejo Represivo Polycomb 2 , Factores de Transcripción/genética
7.
Cell Chem Biol ; 28(8): 1158-1168.e13, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-33705687

RESUMEN

PARP14 has been implicated by genetic knockout studies to promote protumor macrophage polarization and suppress the antitumor inflammatory response due to its role in modulating interleukin-4 (IL-4) and interferon-γ signaling pathways. Here, we describe structure-based design efforts leading to the discovery of a potent and highly selective PARP14 chemical probe. RBN012759 inhibits PARP14 with a biochemical half-maximal inhibitory concentration of 0.003 µM, exhibits >300-fold selectivity over all PARP family members, and its profile enables further study of PARP14 biology and disease association both in vitro and in vivo. Inhibition of PARP14 with RBN012759 reverses IL-4-driven protumor gene expression in macrophages and induces an inflammatory mRNA signature similar to that induced by immune checkpoint inhibitor therapy in primary human tumor explants. These data support an immune suppressive role of PARP14 in tumors and suggest potential utility of PARP14 inhibitors in the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Inflamación/tratamiento farmacológico , Interleucina-4/antagonistas & inhibidores , Neoplasias Renales/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Interleucina-4/genética , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Molecular , Poli(ADP-Ribosa) Polimerasas/genética , Células RAW 264.7 , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Cancer Cell ; 39(9): 1214-1226.e10, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34375612

RESUMEN

PARP7 is a monoPARP that catalyzes the transfer of single units of ADP-ribose onto substrates to change their function. Here, we identify PARP7 as a negative regulator of nucleic acid sensing in tumor cells. Inhibition of PARP7 restores type I interferon (IFN) signaling responses to nucleic acids in tumor models. Restored signaling can directly inhibit cell proliferation and activate the immune system, both of which contribute to tumor regression. Oral dosing of the PARP7 small-molecule inhibitor, RBN-2397, results in complete tumor regression in a lung cancer xenograft and induces tumor-specific adaptive immune memory in an immunocompetent mouse cancer model, dependent on inducing type I IFN signaling in tumor cells. PARP7 is a therapeutic target whose inhibition induces both cancer cell-autonomous and immune stimulatory effects via enhanced IFN signaling. These data support the targeting of a monoPARP in cancer and introduce a potent and selective PARP7 inhibitor to enter clinical development.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Interferón Tipo I/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas de Transporte de Nucleósidos/genética , Proteínas de Transporte de Nucleósidos/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Inmunidad Adaptativa/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Escape del Tumor/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Int J Toxicol ; 29(1): 3-19, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19903873

RESUMEN

Vorinostat (SAHA, Zolinza), a histone deacetylase inhibitor, is assessed in nonclinical studies to support its approval for cutaneous T-cell lymphoma. Vorinostat is weakly mutagenic in the Ames assay; is clastogenic in rodent (ie, CHO) cells but not in normal human lymphocytes; and is weakly positive in an in vivo mouse micronucleus assay. No effects are observed on potassium ion currents in the hERG assay up to 300 microM (safety margin approximately 300-fold the approximately 1 microM serum concentration associated with the 400 mg/d maximum recommended human dose. No rat respiratory or central nervous system effects are found at 150 mg/kg (>2-fold maximum recommended human dose). No cardiovascular effects, including effects on QTc interval, are observed after a single oral dose (150 mg/kg) in dogs. Vorinostat is orally dosed daily in rats (controls, 20, 50, or 150 mg/kg/d) and dogs (controls, 60, 80, or 100/125/160 mg/kg/d) for 26 weeks with a 4-week recovery. Rat vorinostat-related adverse findings are decreased food consumption, weight loss, and hematologic changes; a no observed adverse effects level is not established. In dogs, adverse effects are primarily gastrointestinal; the no observed adverse effects level is 60 mg/kg/d (approximately 6-fold maximum recommended human dose). Toxicities are reversible and can be monitored in the clinic.


Asunto(s)
Inhibidores Enzimáticos/toxicidad , Histona Desacetilasas , Ácidos Hidroxámicos/toxicidad , Animales , Células Sanguíneas/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , ADN/efectos de los fármacos , Perros , Evaluación Preclínica de Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Inhibidores Enzimáticos/farmacocinética , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Humanos , Ácidos Hidroxámicos/farmacocinética , Ratones , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Pruebas de Micronúcleos , Ratas , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología , Pruebas de Toxicidad , Vorinostat , Pérdida de Peso/efectos de los fármacos
10.
Clin Cancer Res ; 14(18): 5735-42, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18794082

RESUMEN

PURPOSE: For many tumor cells, de novo lipogenesis is a requirement for growth and survival. A considerable body of work suggests that inhibition of this pathway may be a powerful approach to antineoplastic therapy. It has recently been shown that inhibition of various steps in the lipogenic pathway individually can induce apoptosis or loss of viability in tumor cells. However, it is not clear whether quantitative differences exist in the ability of lipogenic enzymes to control tumor cell survival. We present a systematic approach that allows for a direct comparison of the control of lipogenic pathway enzymes over tumor cell growth and apoptosis using different cancer cells. EXPERIMENTAL DESIGN: RNA interference-mediated, graded down-regulation of fatty acid synthase (FAS) pathway enzymes was employed in combination with measurements of lipogenesis, apoptosis, and cell growth. RESULTS: In applying RNA interference titrations to two lipogenic enzymes, acetyl-CoA carboxylase 1 (ACC1) and FAS, we show that ACC1 and FAS both significantly control cell growth and apoptosis in HCT-116 cells. These results also extend to PC-3 and A2780 cancer cells. CONCLUSIONS: Control of tumor cell survival by different steps in de novo lipogenesis can be quantified. Because ACC1 and FAS both significantly control tumor cell growth and apoptosis, we propose that pharmacologic inhibitors of either enzyme might be useful agents in targeting cancer cells that critically rely on fatty acid synthesis. The experimental approach described here may be extended to other targets or disease-relevant pathways to identify steps suitable for therapeutic intervention.


Asunto(s)
Acetiltransferasas/metabolismo , Neoplasias del Colon/enzimología , Ácido Graso Sintasas/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular , Inhibidores Enzimáticos/farmacología , Células HCT116 , Humanos , Lipogénesis , Transducción de Señal , Transfección
11.
Clin Cancer Res ; 13(24): 7237-42, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18094401

RESUMEN

Histone deacetylase inhibitors (HDI) are a promising new approach to the treatment of cancer. HDIs have been shown to induce differentiation, cell cycle arrest, and apoptosis in a variety of transformed cell lines; inhibit tumor growth in animal models; and show antitumor activity in clinical trials. Vorinostat, which has shown clinical responses in approximately 30% of patients with advanced cutaneous T-cell lymphoma, is the first HDI approved for the treatment of cancer, and it is currently being evaluated in other indications. A better understanding of the molecular determinants of resistance to HDIs may provide the basis for therapeutic combinations with improved clinical efficacy. Poor response to treatment could be linked to systemic factors like pharmacokinetics or to tumor-specific factors both at the level of the malignant cells (tumor intrinsic) or the tumor microenvironment. This review focuses on the tumor intrinsic mechanisms of drug resistance (excluding mechanism of acquired resistance due to chronic exposure). In particular, attention is given to selected mechanisms that are relevant across chemical classes of HDIs and that can aid in the design of rational combination strategies.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Neoplasias/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Humanos , Neoplasias/enzimología
12.
Mol Cancer Ther ; 6(1): 51-60, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17218635

RESUMEN

Growth of prostate cancer cells is initially dependent on androgens, and androgen ablation therapy is used to control tumor growth. Unfortunately, resistance to androgen ablation therapy inevitably occurs, and there is an urgent need for better treatments for advanced prostate cancer. Histone deacetylase inhibitors, such as suberoylanilide hydroxamic acid (SAHA; vorinostat), are promising agents for the treatment of a range of malignancies, including prostate cancer. SAHA inhibited growth of the androgen-responsive LNCaP prostate cancer cell line at low micromolar concentrations and induced caspase-dependent apoptosis associated with chromatin condensation, DNA fragmentation, and mitochondrial membrane depolarization at higher concentrations (>/=5 mumol/L). Gene profiling and immunoblot analyses showed a decrease in androgen receptor (AR) mRNA and protein in LNCaP cells cultured with SAHA compared with control cells, with a corresponding decrease in levels of the AR-regulated gene, prostate-specific antigen. Culture of LNCaP cells in steroid-free medium markedly sensitized the cells to SAHA. Moreover, a combination of low, subeffective doses of SAHA and the AR antagonist bicalutamide resulted in a synergistic reduction in cell proliferation and increase in caspase-dependent cell death. Addition of exogenous androgen prevented the induction of cell death, indicating that suppression of androgen signaling was required for synergy. At the subeffective concentrations, these agents had no effect, alone or in combination, on proliferation or death of AR-negative PC-3 prostate cancer cells. Our findings indicate that SAHA is effective in targeting the AR signaling axis and that androgen deprivation sensitizes prostate cancer cells to SAHA. Consequently, combinatorial treatments that target different components of the AR pathway may afford a more effective strategy to control the growth of prostate cancer cells.


Asunto(s)
Antagonistas de Receptores Androgénicos , Anilidas/farmacología , Anticarcinógenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo , Sinergismo Farmacológico , Perfilación de la Expresión Génica , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Nitrilos , Neoplasias de la Próstata/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Esteroides/farmacología , Compuestos de Tosilo , Células Tumorales Cultivadas , Vorinostat
13.
Mol Cancer Ther ; 5(8): 1967-74, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16928817

RESUMEN

Vorinostat (suberoylanilide hydroxamic acid) is the prototype of a family of hybrid polar compounds that can induce growth arrest in transformed cells and shows promise for the treatment of cancer. Vorinostat specifically binds to and inhibits the activity of histone deacetylases resulting in acetylation of nucleosomal histones and an activation of gene transcription. Because histone deacetylases modulate chromatin structure and gene expression, both of which can influence radioresponse, this study was designed to examine the capacity of Vorinostat to influence radiation response in human tumor cells and investigate the mechanism underlying these interactions. Vorinostat induced hyperacetylation of histone H4 in a dose-dependent manner. We tested its ability to radiosensitize three human tumor cell lines (A375, MeWo, and A549) using clonogenic cell survival assays. Clonogenic cell survival assay showed that Vorinostat significantly radiosensitized all three tumor cell lines, substantially reducing the surviving fraction at 2 Gy. We examined potential mechanisms that may contribute to the enhanced radiation response induced by Vorinostat. Vorinostat and radiation alone did not induce apoptosis in the melanoma cell line. However, enhanced apoptosis was observed when cells were exposed to both Vorinostat and radiation, suggesting that Vorinostat renders tumor cells more susceptible to radiation-induced apoptosis. Results from DNA damage repair analysis in cultured A375 cells showed that Vorinostat had a strong inhibitory effect on the nonhomologous end joining pathway after radiation. A detailed examination of the involvement of the DNA repair pathway following Vorinostat treatment showed that Vorinostat reduced the expression of the repair-related genes Ku70, Ku80, and Rad50 in A375 cells as detected by Western blot analysis. We also examined gamma-H2AX phosphorylation as a predictive marker of radiotherapy response to Vorinostat and observed that the combination of Vorinostat and radiation caused a prolongation of expression of DNA repair proteins such as gamma-H2AX. Overall, we conclude that Vorinostat enhances tumor radioresponse by multiple mechanisms that may involve antiproliferative growth inhibition and effects on DNA repair after exposure to radiation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Histonas/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Acetilación , Ácido Anhídrido Hidrolasas , Antígenos Nucleares/efectos de los fármacos , Antígenos Nucleares/genética , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Enzimas Reparadoras del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Histonas/efectos de la radiación , Humanos , Autoantígeno Ku , Melanoma/tratamiento farmacológico , Melanoma/radioterapia , Radiación Ionizante , Células Tumorales Cultivadas , Vorinostat
14.
J Clin Oncol ; 23(17): 3923-31, 2005 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-15897550

RESUMEN

PURPOSE: To determine the safety, dosing schedules, pharmacokinetic profile, and biologic effect of orally administered histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) in patients with advanced cancer. PATIENTS AND METHODS: Patients with solid and hematologic malignancies were treated with oral SAHA administered once or twice a day on a continuous basis or twice daily for 3 consecutive days per week. Pharmacokinetic profile and bioavailibity of oral SAHA were determined. Western blots and enzyme-linked immunosorbent assays of histones isolated from peripheral-blood mononuclear cells (PBMNCs) pre and post-therapy were performed to evaluate target inhibition. RESULTS: Seventy-three patients were treated with oral SAHA and major dose-limiting toxicities were anorexia, dehydration, diarrhea, and fatigue. The maximum tolerated dose was 400 mg qd and 200 mg bid for continuous daily dosing and 300 mg bid for 3 consecutive days per week dosing. Oral SAHA had linear pharmacokinetics from 200 to 600 mg, with an apparent half-life ranging from 91 to 127 minutes and 43% oral bioavailability. Histones isolated from PBMNCs showed consistent accumulation of acetylated histones post-therapy, and enzyme-linked immunosorbent assay demonstrated a trend towards a dose-dependent accumulation of acetylated histones from 200 to 600 mg of oral SAHA. There was one complete response, three partial responses, two unconfirmed partial responses, and 22 (30%) patients remained on study for 4 to 37+ months. CONCLUSIONS: Oral SAHA has linear pharmacokinetics and good bioavailability, inhibits histone deacetylase activity in PBMNCs, can be safely administered chronically, and has a broad range of antitumor activity.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/administración & dosificación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Disponibilidad Biológica , Esquema de Medicación , Inhibidores Enzimáticos/farmacocinética , Femenino , Neoplasias Hematológicas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacocinética , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/metabolismo , Vorinostat
15.
Clin Cancer Res ; 11(10): 3958-65, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15897598

RESUMEN

Histone deacetylases (HDAC) and histone acetyltransferases exert opposing enzymatic activities that modulate the degree of acetylation of histones and other intracellular molecular targets, thereby regulating gene expression, cellular differentiation, and survival. HDAC inhibition results in accumulation of acetylated histones and induces differentiation and/or apoptosis in transformed cells. In this study, we characterized the effect of two HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and m-carboxycinnamic acid bis-hydroxamide, on thyroid carcinoma cell lines, including lines originating from anaplastic and medullary carcinomas. In these models, both SAHA and m-carboxycinnamic acid bis-hydroxamide induced growth arrest and caspase-mediated apoptosis and increased p21 protein levels, retinoblastoma hypophosphorylation, BH3-interacting domain death agonist cleavage, Bax up-regulation, down-regulation of Bcl-2, A1, and Bcl-x(L) expression, and cleavage of poly(ADP-ribose) polymerase and caspase-8, -9, -3, -7, and -2. Transfection of Bcl-2 cDNA partially suppressed SAHA-induced cell death. SAHA down-regulated the expression of the apoptosis inhibitors FLIP and cIAP-2 and sensitized tumor cells to cytotoxic chemotherapy and death receptor activation. Our studies provide insight into the tumor type-specific mechanisms of antitumor effects of HDAC inhibitors and a framework for future clinical applications of HDAC inhibitors in patients with thyroid cancer, including histologic subtypes (e.g., anaplastic and medullary thyroid carcinomas) for which limited, if any, therapeutic options are available.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma/patología , Cinamatos/farmacología , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Neoplasias de la Tiroides/patología , Apoptosis , Muerte Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células Tumorales Cultivadas , Vorinostat
16.
J Med Chem ; 59(4): 1556-64, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26769278

RESUMEN

Posttranslational methylation of histones plays a critical role in gene regulation. Misregulation of histone methylation can lead to oncogenic transformation. Enhancer of Zeste homologue 2 (EZH2) methylates histone 3 at lysine 27 (H3K27) and abnormal methylation of this site is found in many cancers. Tazemetostat, an EHZ2 inhibitor in clinical development, has shown activity in both preclinical models of cancer as well as in patients with lymphoma or INI1-deficient solid tumors. Herein we report the structure-activity relationships from identification of an initial hit in a high-throughput screen through selection of tazemetostat for clinical development. The importance of several methyl groups to the potency of the inhibitors is highlighted as well as the importance of balancing pharmacokinetic properties with potency.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Histonas/metabolismo , Metilación/efectos de los fármacos , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Descubrimiento de Drogas , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/farmacocinética , Humanos , Ratones , Complejo Represivo Polycomb 2/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacocinética , Relación Estructura-Actividad
17.
Adv Cancer Res ; 91: 137-68, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15327890

RESUMEN

The base sequence of DNA provides the genetic code for proteins. The regulation of expression or suppression of gene transcription is largely determined by the structure of the chromatin--referred to as epigenetic gene regulation (Agalioti et al., 2002; Jenuwein and Allis, 2001; Richards and Elgin, 2002; Spotswood and Turner, 2002; Zhang and Reinberg, 2001). Posttranslational modifications of the histones of chromatin play an important role in regulating gene expression. Some of the most extensively studied epigenetic modifications involve acetylation/deacetylation of lysines in the tails of the core histones, which is controlled by the action of histone deacetylases (HDACs) and histone acetyltransferases (HATs). A controlled balance between histone acetylation and deacetylation appears to be essential for normal cell growth (Waterborg, 2002). Alterations in the structure or expression of HATs and HDACs occur in many cancers (Jones and Baylin, 2002; Marks et al., 2001, 2003; Timmermann et al., 2001; Wang et al., 2001). A structurally diverse group of molecules has been developed that can inhibit HDACs (HDACi) (Arts et al., 2003; Bouchain and Delorme, 2003; Curtin and Glaser, 2003; Johnstone and Licht, 2003; Marks et al., 2003; Remiszewski, 2003; Richon et al., 1998; Yoshida et al., 2003). These inhibitors induce growth arrest, differentiation, and?or apoptosis of cancer cells in vitro and in in vivo tumor-bearing animal models. Clinical trials with several of these agents have shown that certain HDACi have antitumor activity against various cancers at doses that are well tolerated by patients (Gottlicher et al., 2001; Kelly et al., 2002a,b; Piekarz et al., 2001; Wozniak et al., 1999).


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Proteínas de Neoplasias/antagonistas & inhibidores , Acetilación/efectos de los fármacos , Acetiltransferasas/fisiología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Acetiltransferasas , Histona Desacetilasas/fisiología , Humanos , Masculino , Ratones , Modelos Moleculares , Proteínas de Neoplasias/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos
18.
Clin Cancer Res ; 9(15): 5749-55, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14654560

RESUMEN

PURPOSE: A group of histone deacetylase inhibitors has been shown to be effective in suppressing the growth of a variety of transformed cell lines in vitro and in vivo. The effects of two of these agents, suberoylanilide hydroxamic acid (SAHA) and suberoyl-3-aminopyridineamide hydroxamic acid (pyroxamide), were investigated for their growth-suppressive effects on rhabdomyosarcoma (RMS) cells. EXPERIMENTAL DESIGN AND RESULTS: Dose-response experiments of two RMS cell lines, RD (embryonal) and RH30B (alveolar), were performed with SAHA (0.25-3.0 micro M) and pyroxamide (1.25-20.0 micro M). Both agents caused a dose-dependent decrease in viable cell number and an increase in percentage of dead cells over time. Exposure of the RMS cells to SAHA and pyroxamide resulted in an accumulation of acetylated histones with increasing doses by Western blot analysis. Additionally, there was an induction of p21/WAF1 at 15 and 24 h when the cells were cultured with SAHA (2.0 micro M) or pyroxamide (20.0 micro M), concentrations that were tested because they successfully induced inhibition of cell growth and initiated cell death in both RMS cell lines. An increase in nuclei with hypodiploid or sub-G(1) fraction was found by flow cytometry with increasing doses of both SAHA (0.25-3.0 micro M) and pyroxamide (1.25-20.0 micro M) over time. This finding is consistent with DNA fragmentation and cell death by apoptosis. CONCLUSIONS: SAHA and pyroxamide induce growth suppression and cell death in human RMS in vitro. Accumulation of acetylated histones and induction of p21/WAF1 expression are observed in cells exposed to either agent.


Asunto(s)
Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Inhibidores de Histona Desacetilasas , Rabdomiosarcoma/patología , Acetilación , Línea Celular Tumoral , Citometría de Flujo , Histonas/metabolismo , Humanos , Cinética
19.
Clin Cancer Res ; 9(10 Pt 1): 3578-88, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14506144

RESUMEN

PURPOSE: To evaluate the safety, pharmacokinetics, and biological activity of suberoylanilide hydroxamic acid (SAHA) administered by 2-h i.v. infusion in patients with advanced cancer. EXPERIMENTAL DESIGN: SAHA was administered for 3 days every 21 days in part A and 5 days for 1-3 weeks in part B. Dose escalation proceeded independently in patients with solid tumor and hematological malignancies (part B only). Pharmacokinetic studies were performed along with assessment of acetylated histones in peripheral blood mononuclear cells and tumor tissues. RESULTS: No dose-limiting toxicities were observed in 8 patients enrolled in part A (75, 150, 300, 600, and 900 mg/m(2)/day). Among 12 hematological and 17 solid tumor patients enrolled in part B (300, 600, and 900 mg/m(2)/day), therapy was delayed > or = 1 week for grade 3/4 leukopenia and/or thrombocytopenia in 2 of 5 hematological patients at 600 mg/m(2)/day x 5 days for 3 weeks. The maximal-tolerated dose was 300 mg/m(2)/day x 5 days for 3 weeks for hematological patients. One solid patient on 900 mg/m(2)/day x 5 days for 3 weeks developed acute respiratory distress and grade 3 hypotension. The cohort was expanded to 6 patients, and no additional dose-limiting toxicities were observed. Mean terminal half-life ranged from 21 to 58 min, and there was dose-proportional increase in area under the curve. An accumulation of acetylated histones in peripheral blood mononuclear cells up to 4 h postinfusion was observed at higher dose levels. Posttherapy tumor biopsies showed an accumulation of acetylated histones by immunohistochemistry. Four (2 lymphoma and 2 bladder) patients had objective tumor regression with clinical improvement in tumor related symptoms. CONCLUSIONS: Daily i.v. SAHA is well tolerated, inhibits the biological target in vivo, and has antitumor activity in solid and hematological tumors.


Asunto(s)
Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/administración & dosificación , Acetilación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Área Bajo la Curva , Biopsia , Relación Dosis-Respuesta a Droga , Femenino , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacocinética , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Piel/patología , Factores de Tiempo , Resultado del Tratamiento , Vorinostat
20.
Curr Opin Pharmacol ; 3(4): 344-51, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12901942

RESUMEN

Post-translational modification of the histones of chromatin has a fundamental role in regulating gene expression. Enzymes involved in these epigenetic events include histone deacetylases (class I and class II), which can be inhibited by a structurally diverse group of small molecules. These histone deacetylase inhibitors induce growth arrest, differentiation and/or apoptosis of cancer cells in vitro and in vivo. Results of clinical trials with several of these agents have indicated that they are well tolerated at doses that have anti-tumour activity.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Histona Desacetilasas , Animales , Antineoplásicos/farmacología , Ensayos Clínicos como Asunto , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda