Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 588(7838): 509-514, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32927473

RESUMEN

The MAPK/ERK kinase MEK is a shared effector of the frequent cancer drivers KRAS and BRAF that has long been pursued as a drug target in oncology1, and more recently in immunotherapy2,3 and ageing4. However, many MEK inhibitors are limited owing to on-target toxicities5-7 and drug resistance8-10. Accordingly, a molecular understanding of the structure and function of MEK within physiological complexes could provide a template for the design of safer and more effective therapies. Here we report X-ray crystal structures of MEK bound to the scaffold KSR (kinase suppressor of RAS) with various MEK inhibitors, including the clinical drug trametinib. The structures reveal an unexpected mode of binding in which trametinib directly engages KSR at the MEK interface. In the bound complex, KSR remodels the prototypical allosteric pocket of the MEK inhibitor, thereby affecting binding and kinetics, including the drug-residence time. Moreover, trametinib binds KSR-MEK but disrupts the related RAF-MEK complex through a mechanism that exploits evolutionarily conserved interface residues that distinguish these sub-complexes. On the basis of these insights, we created trametiglue, which limits adaptive resistance to MEK inhibition by enhancing interfacial binding. Our results reveal the plasticity of an interface pocket within MEK sub-complexes and have implications for the design of next-generation drugs that target the RAS pathway.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Piridonas/química , Piridonas/farmacología , Pirimidinonas/química , Pirimidinonas/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión/efectos de los fármacos , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Especificidad por Sustrato , Quinasas raf/química , Quinasas raf/metabolismo
2.
Nature ; 537(7618): 112-116, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27556948

RESUMEN

Deregulation of the Ras-mitogen activated protein kinase (MAPK) pathway is an early event in many different cancers and a key driver of resistance to targeted therapies. Sustained signalling through this pathway is caused most often by mutations in K-Ras, which biochemically favours the stabilization of active RAF signalling complexes. Kinase suppressor of Ras (KSR) is a MAPK scaffold that is subject to allosteric regulation through dimerization with RAF. Direct targeting of KSR could have important therapeutic implications for cancer; however, testing this hypothesis has been difficult owing to a lack of small-molecule antagonists of KSR function. Guided by KSR mutations that selectively suppress oncogenic, but not wild-type, Ras signalling, we developed a class of compounds that stabilize a previously unrecognized inactive state of KSR. These compounds, exemplified by APS-2-79, modulate KSR-dependent MAPK signalling by antagonizing RAF heterodimerization as well as the conformational changes required for phosphorylation and activation of KSR-bound MEK (mitogen-activated protein kinase kinase). Furthermore, APS-2-79 increased the potency of several MEK inhibitors specifically within Ras-mutant cell lines by antagonizing release of negative feedback signalling, demonstrating the potential of targeting KSR to improve the efficacy of current MAPK inhibitors. These results reveal conformational switching in KSR as a druggable regulator of oncogenic Ras, and further suggest co-targeting of enzymatic and scaffolding activities within Ras-MAPK signalling complexes as a therapeutic strategy for overcoming Ras-driven cancers.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oncogenes/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Quinazolinas/farmacología , Proteínas ras/antagonistas & inhibidores , Alelos , Regulación Alostérica/efectos de los fármacos , Línea Celular , Estabilidad de Enzimas/efectos de los fármacos , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Moleculares , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Conformación Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Piridonas/farmacología , Pirimidinonas/farmacología , Quinasas raf/química , Quinasas raf/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
3.
Biochemistry ; 60(4): 289-302, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33440120

RESUMEN

Pseudokinases play important roles in signal transduction and cellular processes similar to those of catalytically competent kinases. However, pseudokinase pharmacological tractability and conformational space accessibility are poorly understood. Pseudokinases have only recently been suggested to adopt "inactive" conformations or interact with conformation-specific kinase inhibitors (e.g., type II compounds). In this work, the heavily substituted pseudokinase STRADα, which possesses a DFG → GLR substitution in the catalytic site that permits nucleotide binding while impairing divalent cation coordination, is used as a test case to demonstrate the potential applicability of conformation-specific, type II compounds to pseudokinase pharmacology. Integrated structural modeling is employed to generate a "GLR-out" conformational ensemble. Likely interacting type II compounds are identified through virtual screening against this ensemble model. Biophysical validation of compound binding is demonstrated through protein thermal stabilization and ATP competition. Localization of a top-performing compound through surface methylation strongly suggests that STRADα can adopt the "GLR-out" conformation and interact with compounds that comply with the standard type II pharmacophore. These results suggest that, despite a loss of catalytic function, some pseudokinases, including STRADα, may retain the conformational switching properties of conventional protein kinases.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/química , Adenosina Trifosfato/química , Humanos , Dominios Proteicos , Estabilidad Proteica
4.
Nat Chem Biol ; 14(3): 291-298, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29355849

RESUMEN

Synthetic tailoring of approved drugs for new indications is often difficult, as the most appropriate targets may not be readily apparent, and therefore few roadmaps exist to guide chemistry. Here, we report a multidisciplinary approach for accessing novel target and chemical space starting from an FDA-approved kinase inhibitor. By combining chemical and genetic modifier screening with computational modeling, we identify distinct kinases that strongly enhance ('pro-targets') or limit ('anti-targets') whole-animal activity of the clinical kinase inhibitor sorafenib in a Drosophila medullary thyroid carcinoma (MTC) model. We demonstrate that RAF-the original intended sorafenib target-and MKNK kinases function as pharmacological liabilities because of inhibitor-induced transactivation and negative feedback, respectively. Through progressive synthetic refinement, we report a new class of 'tumor calibrated inhibitors' with unique polypharmacology and strongly improved therapeutic index in fly and human MTC xenograft models. This platform provides a rational approach to creating new high-efficacy and low-toxicity drugs.


Asunto(s)
Carcinoma Neuroendocrino/metabolismo , Carcinoma/metabolismo , Drosophila/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Tiroides/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Diseño de Fármacos , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Simulación del Acoplamiento Molecular , Trasplante de Neoplasias , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal , Sorafenib/farmacología
5.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31026276

RESUMEN

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Neuroendocrino , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas , Neoplasias de la Tiroides , Animales , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/metabolismo , Biología Computacional/métodos , Drosophila , Modelos Biológicos , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/metabolismo
6.
Mol Pharmacol ; 85(1): 83-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24113749

RESUMEN

The κ-opioid receptor (KOR)-dynorphin system has been implicated in the control of affect, cognition, and motivation, and is thought to be dysregulated in mood and psychotic disorders, as well as in various phases of opioid dependence. KOR agonists exhibit analgesic effects, although the adverse effects produced by some KOR agonists, including sedation, dysphoria, and hallucinations, have limited their clinical use. Interestingly, KOR-mediated dysphoria, assessed in rodents as aversion, has recently been attributed to the activation of the p38 mitogen-activated protein kinase pathway following arrestin recruitment to the activated KOR. Therefore, KOR-selective G protein-biased agonists, which do not recruit arrestin, have been proposed to be more effective analgesics, without the adverse effects triggered by the arrestin pathway. As an initial step toward identifying novel biased KOR agonists, we applied a multifaceted screening strategy utilizing both in silico and parallel screening approaches. We identified several KOR-selective ligand scaffolds with a range of signaling bias in vitro. The arylacetamide-based scaffold includes both G protein- and ß-arrestin-biased ligands, while the endogenous peptides and the diterpene scaffolds are G protein biased. Interestingly, we found scaffold screening to be more successful than library screening in identifying biased ligands. Many of the identified functionally selective ligands are potent selective KOR agonists that are reported to be active in the central nervous system. They therefore represent excellent candidates for in vivo studies aiming at determining the behavioral effects mediated by specific KOR-mediated signaling cascades.


Asunto(s)
Analgésicos Opioides/química , Receptores Opioides kappa/agonistas , Acetamidas/química , Acetamidas/farmacología , Analgésicos Opioides/farmacología , Arrestinas/metabolismo , Simulación por Computador , Bases de Datos de Compuestos Químicos , Diterpenos/química , Diterpenos/farmacología , Dinorfinas/química , Dinorfinas/farmacología , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Transporte de Proteínas , Receptores Opioides kappa/química , Receptores Opioides kappa/metabolismo , Transducción de Señal , Relación Estructura-Actividad , beta-Arrestinas
7.
Nat Cancer ; 4(8): 1157-1175, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37537299

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide. ß-Catenin (CTNNB1)-mutated HCC represents 30% of cases of the disease with no precision therapeutics available. Using chemical libraries derived from clinical multi-kinase inhibitor (KI) scaffolds, we screened HCC organoids to identify WNTinib, a KI with exquisite selectivity in CTNNB1-mutated human and murine models, including patient samples. Multiomic and target engagement analyses, combined with rescue experiments and in vitro and in vivo efficacy studies, revealed that WNTinib is superior to clinical KIs and inhibits KIT/mitogen-activated protein kinase (MAPK) signaling at multiple nodes. Moreover, we demonstrate that reduced engagement on BRAF and p38α kinases by WNTinib relative to several multi-KIs is necessary to avoid compensatory feedback signaling-providing a durable and selective transcriptional repression of mutant ß-catenin/Wnt targets through nuclear translocation of the EZH2 transcriptional repressor. Our studies uncover a previously unknown mechanism to harness the KIT/MAPK/EZH2 pathway to potently and selectively antagonize CTNNB1-mutant HCC with an unprecedented wide therapeutic index.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ratones , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , beta Catenina/genética , beta Catenina/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Factores de Transcripción/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
8.
Org Lett ; 12(8): 1836-8, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20337419

RESUMEN

Simple enantioselective routes to the two key intermediates shown above (at center) for the synthesis of laurenditerpenol have been developed using a Diels-Alder step and the same catalyst system for each.


Asunto(s)
Diterpenos/síntesis química , Diterpenos/química , Furanos/química , Modelos Moleculares , Conformación Molecular , Naftalenos/química , Estereoisomerismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA