Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Med Res Rev ; 43(2): 343-398, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36262046

RESUMEN

Over the past decade, Clk1 has been identified as a promising target for the treatment of various diseases, in which deregulated alternative splicing plays a role. First small molecules targeting Clk1 are in clinical trials for the treatment of solid cancer, where variants of oncogenic proteins derived from alternative splicing promote tumor progression. Since many infectious pathogens hi-jack the host cell's splicing machinery to ensure efficient replication, further indications in this area are under investigation, such as Influenza A, HIV-1 virus, and Trypanosoma infections, and more will likely be discovered in the future. In addition, Clk1 was found to contribute to the progression of Alzheimer's disease through causing an imbalance of tau splicing products. Interestingly, homozygous Clk1 knockout mice showed a rather mild phenotype, opposed to what might be expected in view of the profound role of Clk1 in alternative splicing. A major drawback of most Clk1 inhibitors is their insufficient selectivity; in particular, Dyrk kinases and haspin were frequently identified as off-targets, besides the other Clk isoforms. Only few inhibitors were shown to be selective over Dyrk1A and haspin, whereas no Clk1 inhibitor so far achieved selectivity over the Clk4 isoform. In this review, we carefully compiled all Clk1 inhibitors from the scientific literature and summarized their structure-activity relationships (SAR). In addition, we critically discuss the available selectivity data and describe the inhibitor's efficacy in cellular models, if reported. Thus, we provide a comprehensive overview on the current state of Clk1 drug discovery and highlight the most promising chemotypes.


Asunto(s)
Enfermedad de Alzheimer , Descubrimiento de Drogas , Animales , Ratones , Relación Estructura-Actividad , Empalme Alternativo , Inhibidores de Proteínas Quinasas/farmacología
2.
Chemistry ; 22(4): 1313-21, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26560738

RESUMEN

Determination of the targets of a compound remains an essential aspect in drug discovery. A complete understanding of all binding interactions is critical to recognize in advance both therapeutic effects and undesired consequences. However, the complete polypharmacology of many drugs currently in clinical development is still unknown, especially in the case of G protein-coupled receptor (GPCR) ligands. In this work we have developed a chemoproteomic platform based on the use of chemical probes to explore the target profile of a compound in biological systems. As proof of concept, this methodology has been applied to selected ligands of the therapeutically relevant serotonin 5-HT1A and 5-HT6 receptors, and we have identified and validated some of their off-targets. This approach could be extended to other drugs of interest to study the targeted proteome in disease-relevant systems.


Asunto(s)
Receptor de Serotonina 5-HT1A/química , Receptores Acoplados a Proteínas G/química , Receptores de Serotonina/química , Diseño de Fármacos , Descubrimiento de Drogas , Humanos , Ligandos , Receptor de Serotonina 5-HT1A/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/metabolismo
3.
Proteomics ; 15(17): 3066-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25959371

RESUMEN

The Janus Kinase (JAK) signaling pathway plays a key role for many cellular processes and has recently been correlated with neuronal disorders. In order to understand new links of JAK family members with other signaling pathways, chemical proteomics tools with broad kinase coverage are desirable. A probe that shows outstanding kinase selectivity and allows for the enrichment of up to 133 kinases including many mitogen activated kinase (MAPK) members and JAK kinases has been developed. Furthermore, this probe was applied to establish the selectivity profile of the JAK1/2 inhibitor momelotinib that is currently evaluated in clinical phase 3 studies. These results render this probe a valuable tool for the investigation of JAK and JAK related signaling pathways and the selectivity profiling of kinase inhibitors.


Asunto(s)
Benzamidas/química , Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Sondas Moleculares/química , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Benzamidas/farmacología , Línea Celular/efectos de los fármacos , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Sondas Moleculares/síntesis química , Sondas Moleculares/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/análisis , Proteínas/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , TYK2 Quinasa/metabolismo
4.
ChemMedChem ; 17(15): e202200097, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35760756

RESUMEN

Vildagliptin is a marketed DPP4 inhibitor, used in the management of type 2 diabetes. The molecule also has notable DPP8/9 affinity, with some preference for DPP9. Therefore, we aimed to use vildagliptin as a starting point for selective DPP8/9 inhibitors, and to engineer out the parent compound's DPP4-affinity. In addition, we wanted to identify substructures in the obtained molecules that allow their further optimization into inhibitors with maximal DPP9 selectivity. Various 2S-cyanopyrrolidines and isoindoline were investigated as P1 residues of vildagliptin analogs. The obtained set was expanded with derivatives bearing O-substituted, N-(3-hydroxyadamantyl)glycine moieties at the P2 position. In this way, representatives were discovered with DPP8/9 potencies comparable to the parent molecule, but with overall selectivity towards DPP4, DPP2, FAP, and PREP. Furthermore, the most promising molecules in this series have a 4- to 7-fold preference for DPP9 over DPP8. Finally, a molecular dynamics study was carried out to maximize our insight into experimental selectivity data.


Asunto(s)
Diabetes Mellitus Tipo 2 , Dipeptidasas , Inhibidores de la Dipeptidil-Peptidasa IV , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4 , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Humanos , Vildagliptina
5.
Methods Mol Biol ; 2228: 237-252, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33950495

RESUMEN

In order to understand the full mechanism of action of candidate drug molecules, it is critical to thoroughly characterize their interactions with endogenously expressed pharmacological targets and potentially undesired off-targets. Here we describe a chemoproteomics approach that is based on functionalized analogs of the compound of interest to affinity enrich target proteins from cell or tissue extracts. Experiments are designed as competition binding assays where free parental compound is spiked at a range of concentrations into the extracts to compete specific binders off the immobilized compound matrix. Quantification of matrix-bound proteins enables generation of dose-response curves and half-binding concentrations. In addition, the influence of the affinity matrix on the equilibrium is determined in rebinding experiments. TMT10 isobaric mass tags enable analyzing repeat binding and dose-dependent competition samples in a single mass spectrometry analysis run, thus enabling the efficient identification of targets, apparent dissociation constants, and selectivity of small molecules in a single experiment. The workflow is exemplified with the kinase inhibitor sunitinib.


Asunto(s)
Inhibidores de Proteínas Quinasas/metabolismo , Proteínas/análisis , Proteómica , Sunitinib/metabolismo , Espectrometría de Masas en Tándem , Animales , Unión Competitiva , Femenino , Humanos , Placenta/metabolismo , Embarazo , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proyectos de Investigación , Sunitinib/farmacología
6.
ChemMedChem ; 12(12): 999-1011, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28544567

RESUMEN

The receptor tyrosine kinase EPHA2 has gained attention as a therapeutic drug target for cancer and infectious diseases. However, EPHA2 research and EPHA2-based therapies have been hampered by the lack of selective small-molecule inhibitors. Herein we report the synthesis and evaluation of dedicated EPHA2 inhibitors based on the clinical BCR-ABL/SRC inhibitor dasatinib as a lead structure. We designed hybrid structures of dasatinib and the previously known EPHA2 binders CHEMBL249097, PD-173955, and a known EPHB4 inhibitor in order to exploit both the ATP pocket entrance as well as the ribose pocket as binding epitopes in the kinase EPHA2. Medicinal chemistry and inhibitor design were guided by a chemical proteomics approach, allowing early selectivity profiling of the newly synthesized inhibitor candidates. Concomitant protein crystallography of 17 inhibitor co-crystals delivered detailed insight into the atomic interactions that underlie the structure-affinity relationship. Finally, the anti-proliferative effect of the inhibitor candidates was confirmed in the glioblastoma cell line SF-268. In this work, we thus discovered a novel EPHA2 inhibitor candidate that features an improved selectivity profile while maintaining potency against EPHA2 and anticancer activity in SF-268 cells.


Asunto(s)
Química Farmacéutica , Descubrimiento de Drogas , Inhibidores de Proteínas Quinasas/farmacología , Proteómica , Receptor EphA2/antagonistas & inhibidores , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Receptor EphA2/metabolismo , Relación Estructura-Actividad
7.
Curr Cancer Drug Targets ; 17(2): 177-190, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27585695

RESUMEN

BACKGROUND: ATP-binding cassette (ABC) transporters, P-glycoprotein (P-gp, ABCB1) and breast cancer resistance protein (BCRP/ABCG2) are major determinants of pharmacokinetic, safety and efficacy profiles of drugs thereby effluxing a broad range of endogenous substances across the plasma membrane. Overexpression of these transporters in various tumors is also implicated in the development of multidrug resistance (MDR) and thus, hampers the success of cancer chemotherapy. Modulators of these efflux transporters in combination with chemotherapeutics could be a promising concept to increase the effective intracellular concentration of anticancer drugs. However, broad and overlapped specificity for substrates and modulators of ABCB1 and ABCG2, merely induce toxicity and unwanted drug-drug interactions and thus, lead to late-stage failure of drugs. OBJECTIVE: In present investigation, we aim to identify specific 3D structural requirements for selective inhibition of ABCB1 and ABCG2 transport function. METHOD: GRID Independent Molecular Descriptor (GRIND) models of selective inhibitors of both transporters have been developed, using their most probable binding conformations obtained from molecular docking protocol. RESULTS: Our results demonstrated a dominant role of molecular shape and different H-bonding patterns in drug-ABCB1/ABCG2 selective interactions. Moreover, distinct distances of different pharmacophoric features from steric hot spots of the molecules provided a strong basis of selectivity for both transporters. Additionally, our results suggested the presence of two H-bond donors at a distance of 8.4-8.8 Å in selective modulators of ABCG2. CONCLUSION: Our findings concluded that molecular shape along with three dimensional pattern of Hbonding in MDR modulators play a critical role in determining the selectivity between the two targets.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Antineoplásicos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Relación Estructura-Actividad , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Antineoplásicos/química , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias/química , Propafenona/química , Propafenona/farmacología , Quinolinas/química , Quinolinas/farmacología , Reproducibilidad de los Resultados , Homología Estructural de Proteína
8.
Biochem Pharmacol ; 105: 34-41, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26930564

RESUMEN

Classical evaluation of target selectivity is usually undertaken by measuring the binding affinity of lead compounds against a number of potential targets under equilibrium conditions, without considering the kinetics of the ligand-receptor interaction. In the present study we propose a combined strategy including both equilibrium- and kinetics-based selectivity profiling. The adenosine receptor (AR) was chosen as a prototypical drug target. Six in-house AR antagonists were evaluated in a radioligand displacement assay for their affinity and in a competition association assay for their binding kinetics on three AR subtypes. One of the compounds with a promising kinetic selectivity profile was also examined in a [(35)S]-GTPγS binding assay for functional activity. We found that XAC and LUF5964 were kinetically more selective for the A1R and A3R, respectively, although they are non-selective in terms of their affinity. In comparison, LUF5967 displayed a strong equilibrium-based selectivity for the A1R over the A2AR, yet its kinetic selectivity thereon was less pronounced. In a GTPγS assay, LUF5964 exhibited insurmountable antagonism on the A3R while having a surmountable effect on the A1R, consistent with its kinetic selectivity profile. This study provides evidence that equilibrium and kinetic selectivity profiling can both be important in the early phases of the drug discovery process. Our proposed combinational strategy could be considered for future medicinal chemistry efforts and aid the design and discovery of different or even better leads for clinical applications.


Asunto(s)
Antagonistas de Receptores Purinérgicos P1/farmacocinética , Receptores Purinérgicos P1/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Humanos , Cinética , Unión Proteica/fisiología
9.
Methods Mol Biol ; 1439: 143-57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27316993

RESUMEN

Biochemical selectivity profiling is an integral part of early drug development. Typically compounds from optimization phase are regularly tested for off-target activities within or across target families. This article presents workflow and critical aspects of biochemical protein kinase profiling based on microfluidic mobility shift assays.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Técnicas Analíticas Microfluídicas/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Animales , Evaluación Preclínica de Medicamentos/instrumentación , Ensayo de Cambio de Movilidad Electroforética/instrumentación , Diseño de Equipo , Humanos , Técnicas Analíticas Microfluídicas/instrumentación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda