Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 30(10): 1191-1210.e20, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37557181

RESUMEN

KAT6A, and its paralog KAT6B, are histone lysine acetyltransferases (HAT) that acetylate histone H3K23 and exert an oncogenic role in several tumor types including breast cancer where KAT6A is frequently amplified/overexpressed. However, pharmacologic targeting of KAT6A to achieve therapeutic benefit has been a challenge. Here we describe identification of a highly potent, selective, and orally bioavailable KAT6A/KAT6B inhibitor CTx-648 (PF-9363), derived from a benzisoxazole series, which demonstrates anti-tumor activity in correlation with H3K23Ac inhibition in KAT6A over-expressing breast cancer. Transcriptional and epigenetic profiling studies show reduced RNA Pol II binding and downregulation of genes involved in estrogen signaling, cell cycle, Myc and stem cell pathways associated with CTx-648 anti-tumor activity in ER-positive (ER+) breast cancer. CTx-648 treatment leads to potent tumor growth inhibition in ER+ breast cancer in vivo models, including models refractory to endocrine therapy, highlighting the potential for targeting KAT6A in ER+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Histonas/metabolismo , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Transducción de Señal , Línea Celular Tumoral
2.
Mol Cancer Ther ; 21(1): 3-15, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34737197

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) overexpression in hematologic and solid tumors methylates arginine residues on cellular proteins involved in important cancer functions including cell-cycle regulation, mRNA splicing, cell differentiation, cell signaling, and apoptosis. PRMT5 methyltransferase function has been linked with high rates of tumor cell proliferation and decreased overall survival, and PRMT5 inhibitors are currently being explored as an approach for targeting cancer-specific dependencies due to PRMT5 catalytic function. Here, we describe the discovery of potent and selective S-adenosylmethionine (SAM) competitive PRMT5 inhibitors, with in vitro and in vivo characterization of clinical candidate PF-06939999. Acquired resistance mechanisms were explored through the development of drug resistant cell lines. Our data highlight compound-specific resistance mutations in the PRMT5 enzyme that demonstrate structural constraints in the cofactor binding site that prevent emergence of complete resistance to SAM site inhibitors. PRMT5 inhibition by PF-06939999 treatment reduced proliferation of non-small cell lung cancer (NSCLC) cells, with dose-dependent decreases in symmetric dimethyl arginine (SDMA) levels and changes in alternative splicing of numerous pre-mRNAs. Drug sensitivity to PF-06939999 in NSCLC cells associates with cancer pathways including MYC, cell cycle and spliceosome, and with mutations in splicing factors such as RBM10. Translation of efficacy in mouse tumor xenograft models with splicing mutations provides rationale for therapeutic use of PF-06939999 in the treatment of splicing dysregulated NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , S-Adenosilmetionina/metabolismo , Animales , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Medicamentos , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones
3.
ACS Med Chem Lett ; 11(6): 1175-1184, 2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32550998

RESUMEN

Two novel compounds were identified as Naa50 binders/inhibitors using DNA-encoded technology screening. Biophysical and biochemical data as well as cocrystal structures were obtained for both compounds (3a and 4a) to understand their mechanism of action. These data were also used to rationalize the binding affinity differences observed between the two compounds and a MLGP peptide-containing substrate. Cellular target engagement experiments further confirm the Naa50 binding of 4a and demonstrate its selectivity toward related enzymes (Naa10 and Naa60). Additional analogs of inhibitor 4a were also evaluated to study the binding mode observed in the cocrystal structures.

4.
Mol Cancer Ther ; 17(12): 2530-2542, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30232146

RESUMEN

Tumors use indoleamine 2,3-dioxygenase-1 (IDO1) as a major mechanism to induce an immunosuppressive microenvironment. IDO1 expression is upregulated in many cancers and considered to be a resistance mechanism to immune checkpoint therapies. IDO1 is induced in response to inflammatory stimuli such as IFNγ and promotes immune tolerance by depleting tryptophan and producing tryptophan catabolites, including kynurenine, in the tumor microenvironment. This leads to effector T-cell anergy and enhanced Treg function through upregulation of FoxP3. As a nexus for the induction of key immunosuppressive mechanisms, IDO1 represents an important immunotherapeutic target in oncology. Here, we report the identification and characterization of the novel selective, orally bioavailable IDO1 inhibitor EOS200271/PF-06840003. It reversed IDO1-induced T-cell anergy in vitro In mice carrying syngeneic tumor grafts, PF-06840003 reduced intratumoral kynurenine levels by over 80% and inhibited tumor growth both in monotherapy and, with an increased efficacy, in combination with antibodies blocking the immune checkpoint ligand PD-L1. We demonstrate that anti-PD-L1 therapy results in increased IDO1 metabolic activity thereby providing additional mechanistic rationale for combining PD-(L)1 blockade with IDO1 inhibition in cancer immunotherapies. Supported by these preclinical data and favorable predicted human pharmacokinetic properties of PF-06840003, a phase I open-label, multicenter clinical study (NCT02764151) has been initiated.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Biocatálisis , Inhibidores Enzimáticos/farmacología , Inmunoterapia , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indoles/farmacología , Succinimidas/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Quinurenina/sangre , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Estereoisomerismo , Especificidad por Sustrato/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
5.
J Med Chem ; 61(3): 650-665, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29211475

RESUMEN

A new series of lactam-derived EZH2 inhibitors was designed via ligand-based and physicochemical-property-based strategies to address metabolic stability and thermodynamic solubility issues associated with previous lead compound 1. The new inhibitors incorporated an sp3 hybridized carbon atom at the 7-position of the lactam moiety present in lead compound 1 as a replacement for a dimethylisoxazole group. This transformation enabled optimization of the physicochemical properties and potency compared to compound 1. Analysis of relationships between calculated log D (clogD) values and in vitro metabolic stability and permeability parameters identified a clogD range that afforded an increased probability of achieving favorable ADME data in a single molecule. Compound 23a exhibited the best overlap of potency and pharmaceutical properties as well as robust tumor growth inhibition in vivo and was therefore advanced as a development candidate (PF-06821497). A crystal structure of 23a in complex with the three-protein PRC2 complex enabled understanding of the key structural features required for optimal binding.


Asunto(s)
Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Isoquinolinas/farmacología , Isoquinolinas/farmacocinética , Administración Oral , Disponibilidad Biológica , Línea Celular Tumoral , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Modelos Moleculares , Conformación Molecular
6.
J Med Chem ; 60(23): 9617-9629, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29111717

RESUMEN

Tumors use tryptophan-catabolizing enzymes such as indoleamine 2,3-dioxygenase (IDO-1) to induce an immunosuppressive environment. IDO-1 is induced in response to inflammatory stimuli and promotes immune tolerance through effector T-cell anergy and enhanced Treg function. As such, IDO-1 is a nexus for the induction of a key immunosuppressive mechanism and represents an important immunotherapeutic target in oncology. Starting from HTS hit 5, IDO-1 inhibitor 6 (EOS200271/PF-06840003) has been developed. The structure-activity relationship around 6 is described and rationalized using the X-ray crystal structure of 6 bound to human IDO-1, which shows that 6, differently from most of the IDO-1 inhibitors described so far, does not bind to the heme iron atom and has a novel binding mode. Clinical candidate 6 shows good potency in an IDO-1 human whole blood assay and also shows a very favorable ADME profile leading to favorable predicted human pharmacokinetic properties, including a predicted half-life of 16-19 h.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indoles/farmacología , Succinimidas/farmacología , Animales , Línea Celular , Cristalografía por Rayos X , Perros , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indoles/química , Indoles/farmacocinética , Macaca fascicularis , Masculino , Ratones , Simulación del Acoplamiento Molecular , Ratas , Relación Estructura-Actividad , Succinimidas/química , Succinimidas/farmacocinética
7.
Elife ; 62017 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-29185984

RESUMEN

Polycomb repressive complex 2 (PRC2) is a key chromatin modifier responsible for methylation of lysine 27 in histone H3. PRC2 has been shown to interact with thousands of RNA species in vivo, but understanding the physiological function of RNA binding has been hampered by the lack of separation-of-function mutants. Here, we use comprehensive mutagenesis and hydrogen deuterium exchange mass spectrometry (HDX-MS) to identify critical residues for RNA interaction in PRC2 core complexes from Homo sapiens and Chaetomium thermophilum, for which crystal structures are known. Preferential binding of G-quadruplex RNA is conserved, surprisingly using different protein elements. Key RNA-binding residues are spread out along the surface of EZH2, with other subunits including EED also contributing, and missense mutations of some of these residues have been found in cancer patients. The unusual nature of this protein-RNA interaction provides a paradigm for other epigenetic modifiers that bind RNA without canonical RNA-binding motifs.


Asunto(s)
Aminoácidos/genética , Aminoácidos/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , G-Cuádruplex , ARN/metabolismo , Chaetomium/enzimología , Análisis Mutacional de ADN , Proteína Potenciadora del Homólogo Zeste 2/química , Humanos , Espectrometría de Masas , Unión Proteica
8.
Methods Mol Biol ; 1608: 271-297, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28695516

RESUMEN

This chapter describes the approaches taken in the development of the first PARP inhibitor to enter clinical trial, rucaparib (now called Rubraca), in 2003. We describe the general principles of crystal-based drug design, the purification and crystallization of the PARP-1 catalytic domain and how this was used to develop highly potent PARP inhibitors, based on the nicotinamide pharmacophore. Several methods have been used to determine the inhibitory potency in cell-free and whole cell assays, each described with reference to its advantages and disadvantages.


Asunto(s)
Inhibidores Enzimáticos/química , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Animales , Dominio Catalítico , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/química , Indoles/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
10.
J Med Chem ; 59(18): 8306-25, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27512831

RESUMEN

A new enhancer of zeste homolog 2 (EZH2) inhibitor series comprising a substituted phenyl ring joined to a dimethylpyridone moiety via an amide linkage has been designed. A preferential amide torsion that improved the binding properties of the compounds was identified for this series via computational analysis. Cyclization of the amide linker resulted in a six-membered lactam analogue, compound 18. This transformation significantly improved the ligand efficiency/potency of the cyclized compound relative to its acyclic analogue. Additional optimization of the lactam-containing EZH2 inhibitors focused on lipophilic efficiency (LipE) improvement, which provided compound 31. Compound 31 displayed improved LipE and on-target potency in both biochemical and cellular readouts relative to compound 18. Inhibitor 31 also displayed robust in vivo antitumor growth activity and dose-dependent de-repression of EZH2 target genes.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Piridonas/química , Piridonas/farmacología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Ciclización , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Humanos , Isoquinolinas/química , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Lactamas/química , Lactamas/farmacología , Ratones , Ratones SCID , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Piridonas/uso terapéutico
11.
Nat Commun ; 7: 11384, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-27122193

RESUMEN

Polycomb repressive complex 2 (PRC2) mediates gene silencing through chromatin reorganization by methylation of histone H3 lysine 27 (H3K27). Overexpression of the complex and point mutations in the individual subunits of PRC2 have been shown to contribute to tumorigenesis. Several inhibitors of the PRC2 activity have shown efficacy in EZH2-mutated lymphomas and are currently in clinical development, although the molecular basis of inhibitor recognition remains unknown. Here we report the crystal structures of the inhibitor-bound wild-type and Y641N PRC2. The structures illuminate an important role played by a stretch of 17 residues in the N-terminal region of EZH2, we call the activation loop, in the stimulation of the enzyme activity, inhibitor recognition and the potential development of the mutation-mediated drug resistance. The work presented here provides new avenues for the design and development of next-generation PRC2 inhibitors through establishment of a structure-based drug design platform.


Asunto(s)
Antineoplásicos/química , Inhibidores Enzimáticos/química , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/química , Resistencia a Antineoplásicos , Proteína Potenciadora del Homólogo Zeste 2/química , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Modelos Moleculares , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
12.
Assay Drug Dev Technol ; 13(4): 235-40, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26065559

RESUMEN

Conformational remodeling of chromatin in cells is known to alter gene expression. The histone code hypothesis postulates that multiple modifications present on histone tails can regulate gene expression both through direct effects on chromatin compaction as well as through recruitment of unique complexes that signal specific downstream functions. Histone methylation is an important component of the histone code, and the dysregulation of histone methylation in disease makes methyltransferases and demethylases viable targets for drug discovery. We developed a biochemical assay platform, which takes advantage of the fact that protein methyltransferases (PMTs) all utilize the cofactor S-Adenosyl-L-methionine (SAM) as the methyl donor. The platform utilizes the High-throughput Mass Spectrometry (MS) technology to measure SAM and the S-Adenosyl-L-homocysteine product in a label-free manner. The platform has all the advantages of a label-free system coupled with the benefit of substrate agnostic measurements making it an ideal setup for PMT biochemical studies and drug discovery. In addition, MS is ideally suited for detecting multiple modification events within the same substrate. The ability to adjust the detection to monitor the methyl acceptor product allows for real-time measurements of multiple product species simultaneously, a distinct advantage over other commonly used assay formats.


Asunto(s)
Pruebas de Enzimas/métodos , Ensayos Analíticos de Alto Rendimiento , Espectrometría de Masas , Proteína-Arginina N-Metiltransferasas/análisis , Radiometría/métodos , Sitios de Unión , Humanos , Proteína-Arginina N-Metiltransferasas/metabolismo
13.
Bioorg Med Chem Lett ; 25(7): 1532-7, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25746813

RESUMEN

A series of novel enhancer of zeste homolog 2 (EZH2) inhibitors was designed based on the chemical structure of the histone methyltransferase (HMT) inhibitor SAH (S-adenosyl-l-homocysteine). These nucleoside-based EZH2 inhibitors blocked the methylation of nucleosomes at H3K27 in biochemical assays employing both WT PRC2 complex as well as a Y641N mutant PRC2 complex. The most potent compound, 27, displayed IC50's against both complexes of 270 nM and 70 nM, respectively. To our knowledge, compound 27 is the most potent SAH-derived inhibitor of the EZH2 PRC2 complex yet identified. This compound also displayed improved potency, lipophilic efficiency (LipE), and selectivity profile against other lysine methyltransferases compared with SAH.


Asunto(s)
Complejo Represivo Polycomb 2/antagonistas & inhibidores , S-Adenosilhomocisteína/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Modelos Moleculares , Estructura Molecular , S-Adenosilhomocisteína/síntesis química , S-Adenosilhomocisteína/química , Relación Estructura-Actividad
15.
Bioorg Med Chem Lett ; 23(11): 3358-63, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23622982

RESUMEN

The M2 isoform of pyruvate kinase is an emerging target for antitumor therapy. In this letter, we describe the discovery of 2-((1H-benzo[d]imidazol-1-yl)methyl)-4H-pyrido[1,2-a]pyrimidin-4-ones as potent and selective PKM2 activators which were found to have a novel binding mode. The original lead identified from high throughput screening was optimized into an efficient series via computer-aided structure-based drug design. Both a representative compound from this series and an activator described in the literature were used as molecular tools to probe the biological effects of PKM2 activation on cancer cells. Our results suggested that PKM2 activation alone is not sufficient to alter cancer cell metabolism.


Asunto(s)
Bencimidazoles/química , Proteínas Portadoras/agonistas , Proteínas de la Membrana/agonistas , Pirimidinonas/química , Hormonas Tiroideas/agonistas , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Diseño Asistido por Computadora , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Proteínas de la Membrana/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Estructura Terciaria de Proteína , Pirimidinonas/síntesis química , Pirimidinonas/metabolismo , Relación Estructura-Actividad , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
16.
PLoS One ; 8(1): e53994, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23342057

RESUMEN

The non-receptor tyrosine kinase Ack1 belongs to a unique multi-domain protein kinase family, Ack. Ack is the only family of SH3 domain containing kinases to have an SH3 domain following the kinase domain; others have their SH3 domains preceding the kinase domain. Previous reports have suggested that Ack1 does not require phosphorylation for activation and the enzyme activity of the isolated kinase domain is low relative to other kinases. It has been shown to dimerize in the cellular environment, which augments its enzyme activity. The molecular mechanism of activation, however, remains unknown. Here we present structural and biochemical data on Ack1 kinase domain, and kinase domain+SH3 domain that suggest that Ack1 in its monomeric state is autoinhibited, like EGFR and CDK. The activation of the kinase domain may require N-lobe mediated symmetric dimerization, which may be facilitated by the N-terminal SAM domain. Results presented here show that SH3 domain, unlike in Src family tyrosine kinases, does not directly control the activation state of the enzyme. Instead we speculate that the SH3 domain may play a regulatory role by facilitating binding of the MIG6 homologous region to the kinase domain. We postulate that features of Ack1 activation and regulation parallel those of receptor tyrosine kinase EGFR with some interesting differences.


Asunto(s)
Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Regulación Alostérica , Animales , Cristalografía por Rayos X , Activación Enzimática , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Multimerización de Proteína , Células Sf9 , Spodoptera
17.
Methods Mol Biol ; 780: 463-89, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21870278

RESUMEN

This chapter describes the approaches taken in the development of the first PARP inhibitor to enter into clinical trial, AG-014699. We describe the general principles of crystal-based drug design, the purification, and crystallization of the PARP-1 catalytic domain, and how this approach was used to develop highly potent PARP inhibitors based on the nicotinamide pharmacophore. Several methods have been used to determine the inhibitory potency of designed inhibitors in cell-free and whole cell assays; each is described with reference to its advantages and disadvantages.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Cristalografía , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos
18.
Antiviral Res ; 91(2): 102-11, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21620899

RESUMEN

Several potent inhibitors of hepatitis C virus (HCV) NS3/4A protease have been identified that show great clinical potential against genotype 1. Due to the tremendous genetic diversity that exists among HCV isolates, development of broad spectrum inhibitors is challenging. With a limited number of lab strains available for preclinical testing, new tools are required for assessing protease inhibitor activity. We developed a chimeric replicon system for evaluating NS3 protease inhibitor activity against naturally occurring isolates. NS3/4A genes were cloned from the plasma of HCV-infected individuals and inserted into lab strain replicons, replacing the native sequences. The chimeric reporter replicons were transfected into Huh 7.5 cells, their replication monitored by luciferase assays, and their susceptibilities to inhibitors determined. Viable chimeras expressing heterologous genotypes 1, 2, 3, and 4 protease domains were identified that exhibited varying susceptibilities to inhibitors. Protease inhibitor spectrums observed against the chimeric replicon panel strongly correlated with published enzymatic and clinical results. This cell-based chimeric replicon system can be used to characterize the activities of protease inhibitors against diverse natural isolates and may improve the ability to predict dose and clinical efficacy.


Asunto(s)
Antivirales/farmacología , Hepacivirus/genética , Inhibidores de Proteasas/química , Replicón , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/química , Secuencia de Bases , Sitios de Unión , Carbamatos/química , Carbamatos/farmacología , Línea Celular , Clonación Molecular , Transferencia Resonante de Energía de Fluorescencia/métodos , Variación Genética , Vectores Genéticos/genética , Genotipo , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Humanos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Pruebas de Sensibilidad Microbiana , Oligopéptidos/química , Oligopéptidos/farmacología , Filogenia , Inhibidores de Proteasas/farmacología , Quinolinas/química , Quinolinas/farmacología , Análisis de Secuencia de Proteína/métodos , Tiazoles/química , Tiazoles/farmacología , Proteínas no Estructurales Virales/química , Replicación Viral
19.
Bioorg Med Chem Lett ; 21(12): 3557-62, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21612924

RESUMEN

A series of novel and potent small molecule Hsp90 inhibitors was optimized using X-ray crystal structures. These compounds bind in a deep pocket of the Hsp90 enzyme that is partially comprised by residues Asn51 and Ser52. Displacement of several water molecules observed crystallographically in this pocket using rule-based strategies led to significant improvements in inhibitor potency. An optimized inhibitor (compound 17) exhibited potent Hsp90 inhibition in ITC, biochemical, and cell-based assays (K(d)=1.3 nM, K(i)=15 nM, and cellular IC(50)=0.5 µM).


Asunto(s)
Diseño de Fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Pirimidinas/farmacología , Pirroles/síntesis química , Pirroles/química , Pirroles/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología
20.
J Med Chem ; 54(9): 3368-85, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21438541

RESUMEN

A novel class of heat shock protein 90 (Hsp90) inhibitors was discovered by high-throughput screening and was subsequently optimized using a combination of structure-based design, parallel synthesis, and the application of medicinal chemistry principles. Through this process, the biochemical and cell-based potency of the original HTS lead were substantially improved along with the corresponding metabolic stability properties. These efforts culminated with the identification of a development candidate (compound 42) which displayed desired PK/PD relationships, significant efficacy in a melanoma A2058 xenograft tumor model, and attractive DMPK profiles.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Pirazoles/síntesis química , Pirimidinas/síntesis química , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Unión Competitiva , Disponibilidad Biológica , Proteínas Sanguíneas/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Trasplante de Neoplasias , Unión Proteica , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Relación Estructura-Actividad , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA