Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Leuk Res ; 109: 106649, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34271301

RESUMEN

The E26 transformation sequence-related gene ERG encodes a transcription factor involved in normal hematopoiesis, and its expression is abnormal in leukemia. Especially in a type of acute lymphoblastic leukemia (ALL) that is refractory and easy to relapse, the expression of ERG protein is abnormally increased. Chemotherapy can alleviate the condition of ALL, but the location and survival mechanism of the remaining ALL cells after chemotherapy are still not fully understood. It is becoming increasingly clear that the interaction between leukemia cells and their microenvironment plays an important role in the acquisition of drug resistance mutations and disease recurrence. We selected an acute lymphocytic leukemia cell line with high ERG expression, and studied the synergistic effect of chemotherapeutics and small molecule peptides through cell proliferation, apoptosis, and cell cycle experiments; At the same time, we inoculated acute lymphocytic leukemia cells with high ERG expression into mice with severe immunodeficiency to simulate human ALL and investigated (i) the effects of co-administration on the nesting and invasion of leukemia cells and (ii) the effects of the small molecule peptide drug EIP, which targets ERG, on the sensitivity of ALL chemotherapy and the underlying mechanisms.Ara-c and EIP synergistically reduces viability of ALL cells with high ERG expression may be achieved by promoting their apoptosis and inhibiting their nesting.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Citarabina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores de Tumor/genética , Ciclo Celular , Movimiento Celular , Proliferación Celular , Quimioterapia Combinada , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Enzyme Inhib Med Chem ; 36(1): 831-846, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33752569

RESUMEN

Small molecule modulators of mitochondrial function have been attracted much attention in recent years due to their potential therapeutic applications for neurodegenerative diseases. The mitochondrial translocator protein (TSPO) is a promising target for such compounds, given its involvement in the formation of the mitochondrial permeability transition pore in response to mitochondrial stress. In this study, we performed a ligand-based pharmacophore design and virtual screening, and identified a potent hit compound, 7 (VH34) as a TSPO ligand. After validating its biological activity against amyloid-ß (Aß) induced mitochondrial dysfunction and in acute and transgenic Alzheimer's disease (AD) model mice, we developed a library of analogs, and we found two most active compounds, 31 and 44, which restored the mitochondrial membrane potential, ATP production, and cell viability under Aß-induced mitochondrial toxicity. These compounds recovered learning and memory function in acute AD model mice with improved pharmacokinetic properties.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Ligandos , Ratones , Mitocondrias/metabolismo , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo
3.
Bioorg Med Chem ; 28(22): 115776, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33032189

RESUMEN

The global increase in multidrug-resistant pathogens has caused severe problems in the treatment of infections. To overcome these difficulties, the advent of a new chemical class of antibacterial drug is eagerly desired. We aimed at creating novel antibacterial agents against bacterial type II topoisomerases, which are well-validated targets. TP0480066 (compound 32) has been identified by using structure-based optimization originated from lead compound 1, which was obtained as a result of our previous lead identification studies. The MIC90 values of TP0480066 against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococci (VRE), and genotype penicillin-resistant Streptococcus pneumoniae (gPRSP) were 0.25, 0.015, and 0.06 µg/mL, respectively. Hence, TP0480066 can be regarded as a promising antibacterial drug candidate of this chemical class.


Asunto(s)
Antibacterianos/farmacología , ADN-Topoisomerasas de Tipo II/metabolismo , Quinolinas/farmacología , Inhibidores de Topoisomerasa II/farmacología , Antibacterianos/síntesis química , Antibacterianos/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Bacteriana/efectos de los fármacos , Células Hep G2 , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/enzimología , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/química , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/enzimología , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/síntesis química , Inhibidores de Topoisomerasa II/química , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/enzimología
4.
Bioorg Chem ; 105: 104377, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33091668

RESUMEN

Bruton's tyrosine kinase (BTK), a non-receptor tyrosine kinase, is a member of the Tec kinases family and is essential for B cell receptor (BCR) mediated signaling. BTK inhibitors such as ibrutinib hold a prominent role in the treatment of B cell malignancies. Here we disclose a potent, selective, and covalent BTK inhibitor, HZ-A-005, currently in preclinical development. HZ-A-005 demonstrated dose-dependent activity in two xenograft models of lymphoma in vivo. It showed highly favourable safety, pharmacokinetic (PK), and pharmacodynamic (PD) profiles in preclinical studies. On the basis of its potency, selectivity, and covalent mode of inhibition, HZ-A-005 reveals the potential to be a promising BTK inhibitor for a wide range of cancer indications.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Antineoplásicos/farmacología , Desarrollo de Medicamentos , Inhibidores de Proteínas Quinasas/farmacología , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo
5.
PLoS One ; 15(9): e0238999, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32915889

RESUMEN

Aberrant expression of the transcription factor ERG is a key driving event in approximately one-half of all of prostate cancers. Lacking an enzymatic pocket and mainly disordered, the structure of ERG is difficult to exploit for therapeutic design. We recently identified EWS as a specific interacting partner of ERG that is required for oncogenic function. In this study, we aimed to target this specific protein-protein interaction with small molecules. A high-throughput screening (HTS) strategy was implemented to identify potential protein-protein interaction inhibitors. Secondary assays verified the function of several hit compounds, and one lead compound inhibited ERG-mediated phenotypes in prostate cells. This is the first study aimed at targeting the ERG-EWS protein-protein interaction for the development of a small molecule-based prostate cancer therapy.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Estudios de Factibilidad , Humanos , Masculino , Neoplasias de la Próstata/genética , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
6.
Br J Cancer ; 123(6): 1024-1032, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32581342

RESUMEN

BACKGROUND: The ERG oncogene, a member of the ETS family of transcription factor encoding genes, is a genetic driver of prostate cancer. It is activated through a fusion with the androgen-responsive TMPRSS2 promoter in 50% of cases. There is therefore significant interest in developing novel therapeutic agents that target ERG. We have taken an antisense approach and designed morpholino-based oligonucleotides that target ERG by inducing skipping of its constitutive exon 4. METHODS: We designed antisense morpholino oligonucleotides (splice-switching oligonucleotides, SSOs) that target both the 5' and 3' splice sites of ERG's exon 4. We tested their efficacy in terms of inducing exon 4 skipping in two ERG-positive cell lines, VCaP prostate cancer cells and MG63 osteosarcoma cells. We measured their effect on cell proliferation, migration and apoptosis. We also tested their effect on xenograft tumour growth in mice and on ERG protein expression in a human prostate cancer radical prostatectomy sample ex vivo. RESULTS: In VCaP cells, both SSOs were effective at inducing exon 4 skipping, which resulted in a reduction of overall ERG protein levels up to 96 h following a single transfection. SSO-induced ERG reduction decreased cell proliferation, cell migration and significantly increased apoptosis. We observed a concomitant reduction in protein levels for cyclin D1, c-Myc and the Wnt signalling pathway member ß-catenin as well as a marker of activated Wnt signalling, p-LRP6. We tested the 3' splice site SSO in MG63 xenografts in mice and observed a reduction in tumour growth. We also demonstrated that the 3' splice site SSO caused a reduction in ERG expression in a patient-derived prostate tumour tissue cultured ex vivo. CONCLUSIONS: We have successfully designed and tested morpholino-based SSOs that cause a marked reduction in ERG expression, resulting in decreased cell proliferation, a reduced migratory phenotype and increased apoptosis. Our initial tests on mouse xenografts and a human prostate cancer radical prostatectomy specimen indicate that SSOs can be effective for oncogene targeting in vivo. As such, this study encourages further in vivo therapeutic studies using SSOs targeting the ERG oncogene.


Asunto(s)
Oligonucleótidos Antisentido/uso terapéutico , Oncogenes , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Línea Celular Tumoral , Exones , Masculino , Ratones , Neoplasias de la Próstata/patología , Serina Endopeptidasas/genética , Regulador Transcripcional ERG/análisis , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Vía de Señalización Wnt , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Mol Sci ; 20(14)2019 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-31295848

RESUMEN

Human ether a-go-go related gene (hERG) or KV11.1 potassium channels mediate the rapid delayed rectifier current (IKr) in cardiac myocytes. Drug-induced inhibition of hERG channels has been implicated in the development of acquired long QT syndrome type (aLQTS) and fatal arrhythmias. Several marketed drugs have been withdrawn for this reason. Therefore, there is considerable interest in developing better tests for predicting drugs which can block the hERG channel. The drug-binding pocket in hERG channels, which lies below the selectivity filter, normally contains K+ ions and water molecules. In this study, we test the hypothesis that these water molecules impact drug binding to hERG. We developed 3D QSAR models based on alignment independent descriptors (GRIND) using docked ligands in open and closed conformations of hERG in the presence (solvated) and absence (non-solvated) of water molecules. The ligand-protein interaction fingerprints (PLIF) scheme was used to summarize and compare the interactions. All models delineated similar 3D hERG binding features, however, small deviations of about ~0.4 Å were observed between important hotspots of molecular interaction fields (MIFs) between solvated and non-solvated hERG models. These small changes in conformations do not affect the performance and predictive power of the model to any significant extent. The model that exhibits the best statistical values was attained with a cryo_EM structure of the hERG channel in open state without water. This model also showed the best R2 of 0.58 and 0.51 for the internal and external validation test sets respectively. Our results suggest that the inclusion of water molecules during the docking process has little effect on conformations and this conformational change does not impact the predictive ability of the 3D QSAR models.


Asunto(s)
Antineoplásicos/química , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Agua/química , Antineoplásicos/farmacología , Humanos , Unión Proteica , Relación Estructura-Actividad Cuantitativa , Solubilidad , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/química , Flujo de Trabajo
8.
J Mol Cell Cardiol ; 135: 22-30, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31362019

RESUMEN

Rett Syndrome (RTT) is an X-linked neurodevelopmental disorder associated with respiratory abnormalities and, in up to ~40% of patients, with prolongation of the cardiac QTc interval. QTc prolongation calls for cautious use of drugs with a propensity to inhibit hERG channels. The STARS trial has been undertaken to investigate the efficacy of sarizotan, a 5-HT1A receptor agonist, at correcting RTT respiratory abnormalities. The present study investigated whether sarizotan inhibits hERG potassium channels and prolongs ventricular repolarization. Whole-cell patch-clamp measurements were made at 37 °C from hERG-expressing HEK293 cells. Docking analysis was conducted using a recent cryo-EM structure of hERG. Sarizotan was a potent inhibitor of hERG current (IhERG; IC50 of 183 nM) and of native ventricular IKr from guinea-pig ventricular myocytes. 100 nM and 1 µM sarizotan prolonged ventricular action potential (AP) duration (APD90) by 14.1 ±â€¯3.3% (n = 6) and 29.8 ±â€¯3.1% (n = 5) respectively and promoted AP triangulation. High affinity IhERG inhibition by sarizotan was contingent upon channel gating and intact inactivation. Mutagenesis experiments and docking analysis implicated F557, S624 and Y652 residues in sarizotan binding, with weaker contribution from F656. In conclusion, sarizotan inhibits IKr/IhERG, accessing key binding residues on channel gating. This action and consequent ventricular AP prolongation occur at concentrations relevant to those proposed to treat breathing dysrhythmia in RTT. Sarizotan should only be used in RTT patients with careful evaluation of risk factors for QTc prolongation.


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Síndrome de Rett/tratamiento farmacológico , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Potasio Éter-A-Go-Go , Regulación de la Expresión Génica/efectos de los fármacos , Cobayas , Células HEK293 , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Masculino , Miocitos Cardíacos/metabolismo , Compuestos Orgánicos/farmacología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/patología , Síndrome de Rett/genética , Síndrome de Rett/patología , Factores de Riesgo , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética
9.
Mol Pharmacol ; 95(4): 386-397, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30665971

RESUMEN

The human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel (IKr). Drug-mediated or medical condition-mediated disruption of hERG function is the primary cause of acquired long-QT syndrome, which predisposes affected individuals to ventricular arrhythmias and sudden death. Fentanyl abuse poses a serious health concern, with abuse and death rates rising over recent years. As fentanyl has a propensity to cause sudden death, we investigated its effects on the hERG channel. The effects of norfentanyl, the main metabolite, and naloxone, an antidote used in fentanyl overdose, were also examined. Currents of hERG channels stably expressed in HEK293 cells were recorded using the whole-cell voltage-clamp method. When hERG tail currents were analyzed upon -50 mV repolarization after a 50 mV depolarization, fentanyl and naloxone blocked hERG current (IhERG) with IC50 values of 0.9 and 74.3 µM, respectively, whereas norfentanyl did not block. However, fentanyl-mediated block of IhERG was voltage dependent. When a voltage protocol that mimics a human ventricular action potential (AP) was used, fentanyl blocked IhERG with an IC50 of 0.3 µM. Furthermore, fentanyl (0.5 µM) prolonged AP duration and blocked IKr in ventricular myocytes isolated from neonatal rats. The concentrations of fentanyl used in this study were higher than seen with clinical use but overlap with postmortem overdose concentrations. Although mechanisms of fentanyl-related sudden death need further investigation, blockade of hERG channels may contribute to the death of individuals with high-concentration overdose or compromised cardiac repolarization.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/metabolismo , Femenino , Fentanilo , Células HEK293 , Humanos , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Regulador Transcripcional ERG/antagonistas & inhibidores
10.
PLoS Genet ; 14(11): e1007826, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30500808

RESUMEN

Endothelial cell (EC) plasticity in pathological settings has recently been recognized as a driver of disease progression. Endothelial-to-mesenchymal transition (EndMT), in which ECs acquire mesenchymal properties, has been described for a wide range of pathologies, including cancer. However, the mechanism regulating EndMT in the tumor microenvironment and the contribution of EndMT in tumor progression are not fully understood. Here, we found that combined knockdown of two ETS family transcription factors, ERG and FLI1, induces EndMT coupled with dynamic epigenetic changes in ECs. Genome-wide analyses revealed that ERG and FLI1 are critical transcriptional activators for EC-specific genes, among which microRNA-126 partially contributes to blocking the induction of EndMT. Moreover, we demonstrated that ERG and FLI1 expression is downregulated in ECs within tumors by soluble factors enriched in the tumor microenvironment. These data provide new insight into the mechanism of EndMT, functions of ERG and FLI1 in ECs, and EC behavior in pathological conditions.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Proteína Proto-Oncogénica c-fli-1/genética , Animales , Regulación hacia Abajo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Epigénesis Genética , Transición Epitelial-Mesenquimal/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/metabolismo , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo , Microambiente Tumoral/genética
11.
Cancer Res ; 78(13): 3659-3671, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29712692

RESUMEN

Oncogenic activation of the ETS-related gene (ERG) by recurrent gene fusions (predominantly TMPRSS2-ERG) is one of the most validated and prevalent genomic alterations present in early stages of prostate cancer. In this study, we screened small-molecule libraries for inhibition of ERG protein in TMPRSS2-ERG harboring VCaP prostate cancer cells using an In-Cell Western Assay with the highly specific ERG-MAb (9FY). Among a subset of promising candidates, 1-[2-Thiazolylazo]-2-naphthol (NSC139021, hereafter ERGi-USU) was identified and further characterized. ERGi-USU selectively inhibited growth of ERG-positive cancer cell lines with minimal effect on normal prostate or endothelial cells or ERG-negative tumor cell lines. Combination of ERGi-USU with enzalutamide showed additive effects in inhibiting growth of VCaP cells. A screen of kinases revealed that ERGi-USU directly bound the ribosomal biogenesis regulator atypical kinase RIOK2 and induced ribosomal stress signature. In vivo, ERGi-USU treatment inhibited growth of ERG-positive VCaP tumor xenografts with no apparent toxicity. Structure-activity-based derivatives of ERGi-USU recapitulated the ERG-selective activity of the parental compound. Taken together, ERGi-USU acts as a highly selective inhibitor for the growth of ERG-positive cancer cells and has potential for further development of ERG-targeted therapy of prostate cancer and other malignancies.Significance: A highly selective small-molecule inhibitor of ERG, a critical driver of early stages of prostate cancer, will be imperative for prostate cancer therapy. Cancer Res; 78(13); 3659-71. ©2018 AACR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Compuestos Azo/farmacología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Compuestos Azo/uso terapéutico , Benzamidas , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Desnudos , Nitrilos , Proteínas de Fusión Oncogénica/genética , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Bibliotecas de Moléculas Pequeñas , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Pharm ; 15(3): 705-720, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-28853901

RESUMEN

In this study, we catalog structure activity relationships (SAR) of several short chain fatty acid (SCFA)-modified hexosamine analogues used in metabolic glycoengineering (MGE) by comparing in silico and experimental measurements of physiochemical properties important in drug design. We then describe the impact of these compounds on selected biological parameters that influence the pharmacological properties and safety of drug candidates by monitoring P-glycoprotein (Pgp) efflux, inhibition of cytochrome P450 3A4 (CYP3A4), hERG channel inhibition, and cardiomyocyte cytotoxicity. These parameters are influenced by length of the SCFAs (e.g., acetate vs n-butyrate), which are added to MGE analogues to increase the efficiency of cellular uptake, the regioisomeric arrangement of the SCFAs on the core sugar, the structure of the core sugar itself, and by the type of N-acyl modification (e.g., N-acetyl vs N-azido). By cataloging the influence of these SAR on pharmacological properties of MGE analogues, this study outlines design considerations for tuning the pharmacological, physiochemical, and the toxicological parameters of this emerging class of small molecule drug candidates.


Asunto(s)
Inhibidores del Citocromo P-450 CYP3A/farmacología , Diseño de Fármacos , Ácidos Grasos Volátiles/farmacología , Hexosaminas/farmacología , Ingeniería Metabólica/métodos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/química , Evaluación Preclínica de Medicamentos , Ácidos Grasos Volátiles/química , Hexosaminas/química , Estructura Molecular , Miocitos Cardíacos/efectos de los fármacos , Cultivo Primario de Células , Ratas , Relación Estructura-Actividad , Pruebas de Toxicidad/métodos , Regulador Transcripcional ERG/antagonistas & inhibidores
13.
Bioorg Med Chem ; 25(24): 6332-6344, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29042223

RESUMEN

Astemizole is a H1-antagonist endowed with antimalarial activity, but has hERG liabilities. Systematic structural modifications of astemizole led to the discovery of analogues that display very potent activity as inhibitors of the growth of the Plasmodium parasite, but show a decreased hERG inhibition, when compared to astemizole. These compounds can be used as starting point for the development of a new class of antimalarials.


Asunto(s)
Antimaláricos/farmacología , Astemizol/farmacología , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/síntesis química , Antimaláricos/química , Astemizol/síntesis química , Astemizol/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/crecimiento & desarrollo , Relación Estructura-Actividad , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo
14.
Anticancer Res ; 37(7): 3385-3396, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28668826

RESUMEN

BACKGROUND/AIM: The aim of the current study was to determine the effects of the ERG small-molecule inhibitor YK-4-279 on ERG+ prostate cancer patient-derived xenografts (PDX). MATERIALS AND METHODS: ERG activity was blocked using YK-4-279 in three subcutaneously-implanted ERG+ (LuCaP 23.1, 86.2 and 35) and one ERG- (LuCaP 96) PDX. Treated animals tumor volume (TV), body weight (BW) and serum prostate-specific antigen (PSA) were compared to vehicle-treated control animals. Gene expression, proliferation, apoptosis, microvessel density and ERG expression were also assessed. RESULTS: Administration of YK-4-279 decreased TV (p=0.026), proliferation (p=0.0038) and PSA (p=0.022) in Severe Combined Immunodeficiency (SCID) mice bearing LuCaP 23.1 tumors. LuCaP 86.2, LuCaP 35 and LuCaP 96 showed no significant changes in TV, or PSA. Mineralocorticoid receptor (MR) and MR-direct target genes were up-regulated in treatment-resistant LuCaP 86.2 and LuCaP 35 PDX. CONCLUSION: YK-4-279 decreased ERG+ LuCaP 23.1 tumor growth, but not LuCaP 86.2 and LuCaP 35 ERG+ tumor growth.


Asunto(s)
Xenoinjertos/efectos de los fármacos , Indoles/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Xenoinjertos/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores de Mineralocorticoides/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Regulador Transcripcional ERG/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
15.
Bioorg Med Chem Lett ; 27(16): 3939-3943, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28720503

RESUMEN

8-Amino-imidazo[1,5-a]pyrazine-based Bruton's tyrosine kinase (BTK) inhibitors, such as 6, exhibited potent inhibition of BTK but required improvements in both kinase and hERG selectivity (Liu et al., 2016; Gao et al., 2017). In an effort to maintain the inhibitory activity of these analogs and improve their selectivity profiles, we carried out SAR exploration of groups at the 3-position of pyrazine compound 6. This effort led to the discovery of the morpholine group as an optimized pharmacophore. Compounds 13, 23 and 38 displayed excellent BTK potencies, kinase and hERG selectivities, and pharmacokinetic profiles.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Descubrimiento de Drogas , Imidazoles/farmacología , Morfolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Artritis Reumatoide/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/síntesis química , Imidazoles/química , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Modelos Moleculares , Estructura Molecular , Morfolinas/síntesis química , Morfolinas/química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Relación Estructura-Actividad , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo
16.
Cancer Cell ; 31(4): 532-548.e7, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28344039

RESUMEN

Transcription factors play a key role in the development of diverse cancers, and therapeutically targeting them has remained a challenge. In prostate cancer, the gene encoding the transcription factor ERG is recurrently rearranged and plays a critical role in prostate oncogenesis. Here, we identified a series of peptides that interact specifically with the DNA binding domain of ERG. ERG inhibitory peptides (EIPs) and derived peptidomimetics bound ERG with high affinity and specificity, leading to proteolytic degradation of the ERG protein. The EIPs attenuated ERG-mediated transcription, chromatin recruitment, protein-protein interactions, cell invasion and proliferation, and tumor growth. Thus, peptidomimetic targeting of transcription factor fusion products may provide a promising therapeutic strategy for prostate cancer as well as other malignancies.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Peptidomiméticos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/química , Línea Celular Tumoral , Embrión de Pollo , ADN/metabolismo , Humanos , Masculino , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Proteínas de Fusión Oncogénica/genética , Biblioteca de Péptidos , Peptidomiméticos/química , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Dominios Proteicos , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Bioorg Med Chem Lett ; 26(23): 5695-5702, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27839686

RESUMEN

Following the discovery of small molecule acyl piperazine ROMK inhibitors, the acyl octahydropyrazino[2,1-c][1,4]oxazine series was identified. This series displays improved ROMK/hERG selectivity, and as a consequence, the resulting ROMK inhibitors do not evoke QTc prolongation in an in vivo cardiovascular dog model. Further efforts in this series led to the discovery of analogs with improved pharmacokinetic profiles. This new series also retained comparable ROMK potency compared to earlier leads.


Asunto(s)
Oxazinas/química , Oxazinas/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Animales , Diuresis/efectos de los fármacos , Perros , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Hipertensión/tratamiento farmacológico , Macaca mulatta , Oxazinas/farmacocinética , Canales de Potasio de Rectificación Interna/metabolismo , Ratas Sprague-Dawley , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/metabolismo
18.
Nat Struct Mol Biol ; 23(7): 663-72, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27273514

RESUMEN

Control of mRNA levels, a fundamental aspect in the regulation of gene expression, is achieved through a balance between mRNA synthesis and decay. E26-related gene (Erg) proteins are canonical transcription factors whose previously described functions are confined to the control of mRNA synthesis. Here, we report that ERG also regulates gene expression by affecting mRNA stability and identify the molecular mechanisms underlying this function in human cells. ERG is recruited to mRNAs via interaction with the RNA-binding protein RBPMS, and it promotes mRNA decay by binding CNOT2, a component of the CCR4-NOT deadenylation complex. Transcriptome-wide mRNA stability analysis revealed that ERG controls the degradation of a subset of mRNAs highly connected to Aurora signaling, whose decay during S phase is necessary for mitotic progression. Our data indicate that control of gene expression by mammalian transcription factors may follow a more complex scheme than previously anticipated, integrating mRNA synthesis and degradation.


Asunto(s)
Mitosis , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Aurora Quinasas/genética , Aurora Quinasas/metabolismo , Línea Celular Tumoral , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Transducción de Señal , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...