Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 64(6): 2953-2966, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33682420

RESUMEN

Nav1.7 is an extensively investigated target for pain with a strong genetic link in humans, yet in spite of this effort, it remains challenging to identify efficacious, selective, and safe inhibitors. Here, we disclose the discovery and preclinical profile of GDC-0276 (1) and GDC-0310 (2), selective Nav1.7 inhibitors that have completed Phase 1 trials. Our initial search focused on close-in analogues to early compound 3. This resulted in the discovery of GDC-0276 (1), which possessed improved metabolic stability and an acceptable overall pharmacokinetics profile. To further derisk the predicted human pharmacokinetics and enable QD dosing, additional optimization of the scaffold was conducted, resulting in the discovery of a novel series of N-benzyl piperidine Nav1.7 inhibitors. Improvement of the metabolic stability by blocking the labile benzylic position led to the discovery of GDC-0310 (2), which possesses improved Nav selectivity and pharmacokinetic profile over 1.


Asunto(s)
Azetidinas/farmacología , Benzamidas/farmacología , Descubrimiento de Drogas , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Sulfonamidas/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Azetidinas/química , Azetidinas/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Células Cultivadas , Células HEK293 , Humanos , Piperidinas/química , Piperidinas/farmacocinética , Piperidinas/farmacología , Ratas Sprague-Dawley , Sulfonamidas/química , Sulfonamidas/farmacocinética , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacocinética
2.
J Med Chem ; 62(8): 4091-4109, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30943032

RESUMEN

Using structure- and ligand-based design principles, a novel series of piperidyl chromane arylsulfonamide Nav1.7 inhibitors was discovered. Early optimization focused on improvement of potency through refinement of the low energy ligand conformation and mitigation of high in vivo clearance. An in vitro hepatotoxicity hazard was identified and resolved through optimization of lipophilicity and lipophilic ligand efficiency to arrive at GNE-616 (24), a highly potent, metabolically stable, subtype selective inhibitor of Nav1.7. Compound 24 showed a robust PK/PD response in a Nav1.7-dependent mouse model, and site-directed mutagenesis was used to identify residues critical for the isoform selectivity profile of 24.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Sulfonamidas/química , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Analgésicos/química , Analgésicos/metabolismo , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/patología , Perros , Semivida , Humanos , Ligandos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Canal de Sodio Activado por Voltaje NAV1.7/genética , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Ratas , Relación Estructura-Actividad , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Bloqueadores del Canal de Sodio Activado por Voltaje/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
3.
J Med Chem ; 62(2): 908-927, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30499663

RESUMEN

Herein, we report the discovery and optimization of a series of orally bioavailable acyl sulfonamide NaV1.7 inhibitors that are selective for NaV1.7 over NaV1.5 and highly efficacious in in vivo models of pain and hNaV1.7 target engagement. An analysis of the physicochemical properties of literature NaV1.7 inhibitors suggested that acyl sulfonamides with high fsp3 could overcome some of the pharmacokinetic (PK) and efficacy challenges seen with existing series. Parallel library syntheses lead to the identification of analogue 7, which exhibited moderate potency against NaV1.7 and an acceptable PK profile in rodents, but relatively poor stability in human liver microsomes. Further, design strategy then focused on the optimization of potency against hNaV1.7 and improvement of human metabolic stability, utilizing induced fit docking in our previously disclosed X-ray cocrystal of the NaV1.7 voltage sensing domain. These investigations culminated in the discovery of tool compound 33, one of the most potent and efficacious NaV1.7 inhibitors reported to date.


Asunto(s)
Analgésicos/química , Canal de Sodio Activado por Voltaje NAV1.7/química , Sulfonamidas/química , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Analgésicos/metabolismo , Analgésicos/uso terapéutico , Animales , Sitios de Unión , Diseño de Fármacos , Semivida , Humanos , Masculino , Ratones , Ratones Transgénicos , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Dolor/patología , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Bloqueadores del Canal de Sodio Activado por Voltaje/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
4.
Cell Rep ; 24(12): 3133-3145, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231997

RESUMEN

Selective block of NaV1.7 promises to produce non-narcotic analgesic activity without motor or cognitive impairment. Several NaV1.7-selective blockers have been reported, but efficacy in animal pain models required high multiples of the IC50 for channel block. Here, we report a target engagement assay using transgenic mice that has enabled the development of a second generation of selective Nav1.7 inhibitors that show robust analgesic activity in inflammatory and neuropathic pain models at low multiples of the IC50. Like earlier arylsulfonamides, these newer acylsulfonamides target a binding site on the surface of voltage sensor domain 4 to achieve high selectivity among sodium channel isoforms and steeply state-dependent block. The improved efficacy correlates with very slow dissociation from the target channel. Chronic dosing increases compound potency about 10-fold, possibly due to reversal of sensitization arising during chronic injury, and provides efficacy that persists long after the compound has cleared from plasma.


Asunto(s)
Analgésicos/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Neuralgia/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/uso terapéutico , Sulfonamidas/uso terapéutico , Analgésicos/farmacocinética , Animales , Sitios de Unión , Células Cultivadas , Células HEK293 , Humanos , Concentración 50 Inhibidora , Ratones , Canal de Sodio Activado por Voltaje NAV1.7/química , Unión Proteica , Bloqueadores de los Canales de Sodio/farmacocinética , Sulfonamidas/farmacocinética
5.
Bioorg Med Chem Lett ; 28(19): 3141-3149, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30139550

RESUMEN

The voltage gated sodium channel Nav1.7 plays an essential role in the transmission of pain signals. Strong human genetic validation has motivated extensive efforts to discover potent, selective, and efficacious Nav1.7 inhibitors for the treatment of chronic pain. This digest will introduce the structure and function of Nav1.7 and highlight the wealth of recent developments on a diverse array of Nav1.7 inhibitors, including optimization of their potency, selectivity, and PK/PD relationships.


Asunto(s)
Analgésicos/farmacología , Analgésicos/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Analgésicos/química , Analgésicos/farmacocinética , Humanos , Relación Estructura-Actividad , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacocinética
6.
J Med Chem ; 61(11): 4810-4831, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29737846

RESUMEN

The sodium channel NaV1.7 has emerged as a promising target for the treatment of pain based on strong genetic validation of its role in nociception. In recent years, a number of aryl and acyl sulfonamides have been reported as potent inhibitors of NaV1.7, with high selectivity over the cardiac isoform NaV1.5. Herein, we report on the discovery of a novel series of N-([1,2,4]triazolo[4,3- a]pyridin-3-yl)methanesulfonamides as selective NaV1.7 inhibitors. Starting with the crystal structure of an acyl sulfonamide, we rationalized that cyclization to form a fused heterocycle would improve physicochemical properties, in particular lipophilicity. Our design strategy focused on optimization of potency for block of NaV1.7 and human metabolic stability. Lead compounds 10, 13 (GNE-131), and 25 showed excellent potency, good in vitro metabolic stability, and low in vivo clearance in mouse, rat, and dog. Compound 13 also displayed excellent efficacy in a transgenic mouse model of induced pain.


Asunto(s)
Diseño de Fármacos , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Dolor/tratamiento farmacológico , Sulfonamidas/química , Sulfonamidas/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Secuencia de Aminoácidos , Animales , Perros , Estabilidad de Medicamentos , Humanos , Cinética , Ratones , Conformación Molecular , Dolor/metabolismo , Ratas , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacocinética , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
7.
ACS Med Chem Lett ; 8(9): 936-940, 2017 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-28947940

RESUMEN

A novel selective benzoxazepin inhibitor of PI3Kδ has been discovered. Beginning from compound 3, an αPI3K inhibitor, we utilized structure-based drug design and computational analysis of dihedral torsion angles to optimize for PI3Kδ isoform potency and isoform selectivity. Further medicinal chemistry optimization of the series led to the identification of 24, a highly potent and selective inhibitor of PI3Kδ.

8.
ACS Med Chem Lett ; 7(3): 277-82, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26985315

RESUMEN

We report on a novel series of aryl sulfonamides that act as nanomolar potent, isoform-selective inhibitors of the human sodium channel hNaV1.7. The optimization of these inhibitors is described. We aimed to improve potency against hNaV1.7 while minimizing off-target safety concerns and generated compound 3. This agent displayed significant analgesic effects in rodent models of acute and inflammatory pain and demonstrated that binding to the voltage sensor domain 4 site of NaV1.7 leads to an analgesic effect in vivo. Our findings corroborate the importance of hNaV1.7 as a drug target for the treatment of pain.

9.
J Biol Chem ; 291(11): 5986-5996, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26797127

RESUMEN

FGF21 is a stress-induced hormone with potent anti-obesity, insulin-sensitizing, and hepatoprotective properties. Although proteolytic cleavage of recombinant human FGF21 in preclinical species has been observed previously, the regulation of endogenously produced FGF21 is not well understood. Here we identify fibroblast activation protein (FAP) as the enzyme that cleaves and inactivates human FGF21. A selective chemical inhibitor, immunodepletion, or genetic deletion of Fap stabilized recombinant human FGF21 in serum. In addition, administration of a selective FAP inhibitor acutely increased circulating intact FGF21 levels in cynomolgus monkeys. On the basis of our findings, we propose selective FAP inhibition as a potential therapeutic approach to increase endogenous FGF21 activity for the treatment of obesity, type 2 diabetes, non-alcoholic steatohepatitis, and related metabolic disorders.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Endopeptidasas , Factores de Crecimiento de Fibroblastos/química , Gelatinasas/genética , Eliminación de Gen , Células HEK293 , Humanos , Macaca fascicularis , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/genética
10.
Science ; 350(6267): aac5464, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26680203

RESUMEN

Voltage-gated sodium (Nav) channels propagate action potentials in excitable cells. Accordingly, Nav channels are therapeutic targets for many cardiovascular and neurological disorders. Selective inhibitors have been challenging to design because the nine mammalian Nav channel isoforms share high sequence identity and remain recalcitrant to high-resolution structural studies. Targeting the human Nav1.7 channel involved in pain perception, we present a protein-engineering strategy that has allowed us to determine crystal structures of a novel receptor site in complex with isoform-selective antagonists. GX-936 and related inhibitors bind to the activated state of voltage-sensor domain IV (VSD4), where their anionic aryl sulfonamide warhead engages the fourth arginine gating charge on the S4 helix. By opposing VSD4 deactivation, these compounds inhibit Nav1.7 through a voltage-sensor trapping mechanism, likely by stabilizing inactivated states of the channel. Residues from the S2 and S3 helices are key determinants of isoform selectivity, and bound phospholipids implicate the membrane as a modulator of channel function and pharmacology. Our results help to elucidate the molecular basis of voltage sensing and establish structural blueprints to design selective Nav channel antagonists.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Sulfonamidas/química , Sulfonamidas/farmacología , Tiadiazoles/química , Tiadiazoles/farmacología , Secuencia de Aminoácidos , Membrana Celular/química , Cristalización/métodos , Cristalografía por Rayos X , Análisis Mutacional de ADN , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.7/genética , Percepción del Dolor/efectos de los fármacos , Ingeniería de Proteínas , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
11.
Expert Opin Drug Discov ; 9(8): 969-84, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24857041

RESUMEN

INTRODUCTION: Vismodegib is the first Hedgehog (Hh) pathway inhibitor approved in the US for the treatment of adults with metastatic or locally advanced basal cell carcinoma (BCC). It was approved by the US FDA on 30 January 2012, and by the European Commission on 12 July 2013, for the treatment of adult patients with symptomatic metastatic BCC, or locally advanced BCC inappropriate for surgery or radiotherapy. Vismodegib selectively inhibits the Hh signaling pathway, binding to and inhibiting a critical signal-transducing component of the pathway, Smoothened (SMO). Vismodegib was discovered by Genentech, Inc., under a collaboration agreement with Curis, Inc. AREAS COVERED: This article reviews the development of vismodegib from its discovery, preclinical pharmacology and validation to the clinical pharmacokinetics and validation in Phase I and II clinical investigations. We also provide a survey of other Hh pathway inhibitors in clinical development. EXPERT OPINION: The authors' experience in target-based drug discovery suggests that vismodegib's path to the clinic deserves some reflection to identify key steps that have contributed to its success. Targeting the Hh pathway with vismodegib blocks the abberant signaling caused by mutational inactivation of the negative regulator PTCH1 or mutational activation of SMO. Vismodegib gives physicians a treatment option for patients with locally advanced or metastatic BCC for whom surgery or radiation is not recommended.


Asunto(s)
Anilidas/farmacología , Carcinoma Basocelular/tratamiento farmacológico , Piridinas/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Anilidas/historia , Anilidas/farmacocinética , Animales , Antineoplásicos/historia , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Carcinoma Basocelular/patología , Diseño de Fármacos , Descubrimiento de Drogas/historia , Historia del Siglo XXI , Humanos , Piridinas/historia , Piridinas/farmacocinética , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/patología
12.
Bioorg Med Chem Lett ; 23(17): 4953-9, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23867164

RESUMEN

In an effort to identify potent and isoform selective inhibitors of PI3Kδ, GNE-293 (34) was identified. Inhibitor 2 was found to induce micronuclei formation in both the MNT and HCA in vitro assays. Compounds testing negative for genotoxicity were successfully identified through modifications of the 2-benzimidazole substituent and the methylene moiety to disrupt planarity. A variety of heteroatom linkers were explored to examine their effect on potency and isoform selectivity by restricting torsional angles to favor ligand interactions with PI3Kδ's Trp760. These modifications also resulted in an improved in vivo pharmacokinetic profile.


Asunto(s)
Óxidos S-Cíclicos/química , Óxidos S-Cíclicos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Purinas/química , Purinas/farmacología , Animales , Línea Celular , Perros , Humanos , Simulación del Acoplamiento Molecular , Pruebas de Mutagenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/toxicidad , Ratas , Relación Estructura-Actividad
13.
Toxicol Appl Pharmacol ; 266(1): 86-94, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23142475

RESUMEN

Several toxicities are clearly driven by free drug concentrations in plasma, such as toxicities related to on-target exaggerated pharmacology or off-target pharmacological activity associated with receptors, enzymes or ion channels. However, there are examples in which organ toxicities appear to correlate better with total drug concentrations in the target tissues, rather than with free drug concentrations in plasma. Here we present a case study in which a small molecule Met inhibitor, GEN-203, with significant liver and bone marrow toxicity in preclinical species was modified with the intention of increasing the safety margin. GEN-203 is a lipophilic weak base as demonstrated by its physicochemical and structural properties: high LogD (distribution coefficient) (4.3) and high measured pKa (7.45) due to the basic amine (N-ethyl-3-fluoro-4-aminopiperidine). The physicochemical properties of GEN-203 were hypothesized to drive the high distribution of this compound to tissues as evidenced by a moderately-high volume of distribution (Vd>3l/kg) in mouse and subsequent toxicities of the compound. Specifically, the basicity of GEN-203 was decreased through addition of a second fluorine in the 3-position of the aminopiperidine to yield GEN-890 (N-ethyl-3,3-difluoro-4-aminopiperidine), which decreased the volume of distribution of the compound in mouse (Vd=1.0l/kg), decreased its tissue drug concentrations and led to decreased toxicity in mice. This strategy suggests that when toxicity is driven by tissue drug concentrations, optimization of the physicochemical parameters that drive tissue distribution can result in decreased drug concentrations in tissues, resulting in lower toxicity and improved safety margins.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Femenino , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-met/metabolismo , Distribución Aleatoria , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
14.
J Med Chem ; 55(17): 7686-95, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22877085

RESUMEN

Inhibition of PI3Kδ is considered to be an attractive mechanism for the treatment of inflammatory diseases and leukocyte malignancies. Using a structure-based design approach, we have identified a series of potent and selective benzimidazole-based inhibitors of PI3Kδ. These inhibitors do not occupy the selectivity pocket between Trp760 and Met752 that is induced by other families of PI3Kδ inhibitors. Instead, the selectivity of the compounds for inhibition of PI3Kδ relative to other PI3K isoforms appears to be due primarily to the strong interactions these inhibitors are able to make with Trp760 in the PI3Kδ binding pocket. The pharmacokinetic properties and the ability of compound 5 to inhibit the function of B-cells in vivo are described.


Asunto(s)
Bencimidazoles/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Bencimidazoles/química , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Espectrometría de Masa por Ionización de Electrospray , Espectrofotometría Ultravioleta
15.
Nature ; 487(7408): 505-9, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22763448

RESUMEN

Mutationally activated kinases define a clinically validated class of targets for cancer drug therapy. However, the efficacy of kinase inhibitors in patients whose tumours harbour such alleles is invariably limited by innate or acquired drug resistance. The identification of resistance mechanisms has revealed a recurrent theme­the engagement of survival signals redundant to those transduced by the targeted kinase. Cancer cells typically express multiple receptor tyrosine kinases (RTKs) that mediate signals that converge on common critical downstream cell-survival effectors­most notably, phosphatidylinositol-3-OH kinase (PI(3)K) and mitogen-activated protein kinase (MAPK). Consequently, an increase in RTK-ligand levels, through autocrine tumour-cell production, paracrine contribution from tumour stroma or systemic production, could confer resistance to inhibitors of an oncogenic kinase with a similar signalling output. Here, using a panel of kinase-'addicted' human cancer cell lines, we found that most cells can be rescued from drug sensitivity by simply exposing them to one or more RTK ligands. Among the findings with clinical implications was the observation that hepatocyte growth factor (HGF) confers resistance to the BRAF inhibitor PLX4032 (vemurafenib) in BRAF-mutant melanoma cells. These observations highlight the extensive redundancy of RTK-transduced signalling in cancer cells and the potentially broad role of widely expressed RTK ligands in innate and acquired resistance to drugs targeting oncogenic kinases.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Factor de Crecimiento de Hepatocito/metabolismo , Indoles/farmacología , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Lapatinib , Ligandos , Melanoma/enzimología , Melanoma/genética , Melanoma/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Quinazolinas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Vemurafenib
16.
Bioorg Med Chem Lett ; 22(13): 4296-302, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22672799

RESUMEN

A potent inhibitor of PI3Kδ that is ≥ 200 fold selective for the remaining three Class I PI3K isoforms and additional kinases is described. The hypothesis for selectivity is illustrated through structure activity relationships and crystal structures of compounds bound to a K802T mutant of PI3Kγ. Pharmacokinetic data in rats and mice support the use of 3 as a useful tool compound to use for in vivo studies.


Asunto(s)
Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Triptófano/química , Animales , Sitios de Unión , Simulación por Computador , Femenino , Inyecciones Intravenosas , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
17.
J Med Chem ; 55(12): 5887-900, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22626259

RESUMEN

PI3Kδ is a lipid kinase and a member of a larger family of enzymes, PI3K class IA(α, ß, δ) and IB (γ), which catalyze the phosphorylation of PIP2 to PIP3. PI3Kδ is mainly expressed in leukocytes, where it plays a critical, nonredundant role in B cell receptor mediated signaling and provides an attractive opportunity to treat diseases where B cell activity is essential, e.g., rheumatoid arthritis. We report the discovery of novel, potent, and selective PI3Kδ inhibitors and describe a structural hypothesis for isoform (α, ß, γ) selectivity gained from interactions in the affinity pocket. The critical component of our initial pharmacophore for isoform selectivity was strongly associated with CYP3A4 time-dependent inhibition (TDI). We describe a variety of strategies and methods for monitoring and attenuating TDI. Ultimately, a structure-based design approach was employed to identify a suitable structural replacement for further optimization.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Inhibidores del Citocromo P-450 CYP3A , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Artritis Reumatoide/enzimología , Bencimidazoles/química , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Línea Celular , Citocromo P-450 CYP3A , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Fosfatidilinositol 3-Quinasas/química , Conformación Proteica , Especificidad por Sustrato , Factores de Tiempo
18.
Xenobiotica ; 42(5): 456-65, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22122353

RESUMEN

The R- and S-enantiomer of N-(4-(3-(1-ethyl-3,3-difluoropiperidin-4-ylamino)-1H-pyrazolo[3,4-b]pyridin-4-yloxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide are novel MET kinase inhibitors that have been investigated as potential anticancer agents. The effect of the chirality of these compounds on preclinical in vivo pharmacokinetics and toxicity was studied. The plasma clearance for the S-enantiomer was low in mice and monkeys (23.7 and 7.8 mL min(-1) kg(-1), respectively) and high in rats (79.2 mL min(-1) kg(-1)). The R/S enantiomer clearance ratio was 1.5 except in rats (0.49). After oral single-dose administration at 5 mg kg(-1) the R/S enantiomer ratio of AUC(inf) was 0.95, 1.9 and 0.41 in mice, rats and monkeys, respectively. In an oral single-dose dose-ranging study at 200 and 500 mg kg(-1) and multi-dose toxicity study in mice plasma AUC exposure was approximately 2- to 3-fold higher for the R-enantiomer compared to the S-enantiomer. Greater toxicity of the S-enantiomer was observed which appeared to be due to high plasma C(min) values and tissue concentrations approximately 24 h after the final dose. Both enantiomers showed low to moderate permeability in MDCKI cells with no significant efflux, no preferential distribution into red blood cells and similar plasma protein binding in vitro. Overall, the differences between the enantiomers with respect to low dose pharmacokinetics and in vitro properties were relatively modest. However, toxicity results warrant further development of the R-enantiomer over the S-enantiomer.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Pirazoles/farmacocinética , Piridazinas/farmacocinética , Administración Oral , Animales , Proteínas Sanguíneas/metabolismo , Peso Corporal , Línea Celular , Permeabilidad de la Membrana Celular , Perros , Evaluación Preclínica de Medicamentos , Femenino , Macaca fascicularis , Masculino , Ratones , Unión Proteica , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-met/metabolismo , Pirazoles/administración & dosificación , Pirazoles/sangre , Pirazoles/química , Piridazinas/administración & dosificación , Piridazinas/sangre , Piridazinas/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Factores de Tiempo
19.
J Med Chem ; 54(22): 7815-33, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21985639

RESUMEN

Of the four class I phosphoinositide 3-kinase (PI3K) isoforms, PI3Kα has justly received the most attention for its potential in cancer therapy. Herein we report our successful approaches to achieve PI3Kα vs PI3Kß selectivity for two chemical series. In the thienopyrimidine series of inhibitors, we propose that select ligands achieve selectivity derived from a hydrogen bonding interaction with Arg770 of PI3Kα that is not attained with the corresponding Lys777 of PI3Kß. In the benzoxepin series of inhibitors, the selectivity observed can be rationalized by the difference in electrostatic potential between the two isoforms in a given region rather than any specific interaction.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Modelos Moleculares , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzoxepinas/química , Benzoxepinas/farmacocinética , Benzoxepinas/farmacología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/química , Cristalografía por Rayos X , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Humanos , Enlace de Hidrógeno , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Ratones , Piperazinas/química , Piperazinas/farmacocinética , Piperazinas/farmacología , Conformación Proteica , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Relación Estructura-Actividad
20.
J Med Chem ; 54(21): 7579-87, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21981714

RESUMEN

The discovery of 2 (GDC-0980), a class I PI3K and mTOR kinase inhibitor for oncology indications, is described. mTOR inhibition was added to the class I PI3K inhibitor 1 (GDC-0941) scaffold primarily through the substitution of the indazole in 1 for a 2-aminopyrimidine. This substitution also increased the microsomal stability and the free fraction of compounds as evidenced through a pairwise comparison of molecules that were otherwise identical. Highlighted in detail are analogues of an advanced compound 4 that were designed to improve solubility, resulting in 2. This compound, is potent across PI3K class I isoforms with IC(50)s of 5, 27, 7, and 14 nM for PI3Kα, ß, δ, and γ, respectively, inhibits mTOR with a K(i) of 17 nM yet is highly selective versus a large panel of kinases including others in the PIKK family. On the basis of the cell potency, low clearance in mouse, and high free fraction, 2 demonstrated significant efficacy in mouse xenografts when dosed as low as 1 mg/kg orally and is currently in phase I clinical trials for cancer.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Pirimidinas/síntesis química , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Ratones , Ratones Desnudos , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Trasplante de Neoplasias , Conformación Proteica , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA