Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 49(10): 870-881, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34321251

RESUMEN

Point mutations in isocitrate dehydrogenase 1 (IDH1) result in conversion of α-ketoglutarate to the oncometabolite, d-2-hydroxyglutarate (2-HG). Ivosidenib is a once daily (QD), orally available, potent, mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor approved for the treatment of patients with relapsed or refractory acute myeloid leukemia (AML) and intensive chemotherapy-ineligible newly diagnosed AML, with a susceptible IDH1 mutation. We characterized the protein binding, metabolism, metabolites, cell permeability, and drug-drug interaction potential of ivosidenib in humans, monkeys, dogs, rats, and/or mice in in vitro experiments. In vivo pharmacokinetic (PK) profiling and assessment of drug distribution and excretion was undertaken in rats, dogs, and monkeys administered single-dose ivosidenib. The PK/pharmacodynamic (PD) relationship between ivosidenib and 2-HG was analyzed in an mIDH1 xenograft mouse model. Ivosidenib was well absorbed, showed low clearance, and moderate to long terminal half-life (5.3-18.5 hours) in rats, dogs, and monkeys. Brain to plasma exposure ratio was low (2.3%), plasma protein binding was high, and oxidative metabolism was the major elimination pathway. Ivosidenib had high cell permeability and was identified as a substrate for P-glycoprotein. There was moderate induction of cytochrome P450 (P450) enzymes CYP3A4 and CYP2B6 but minimal P450 inhibition or autoinduction. Tumor 2-HG reduction appeared to be dose- and drug-exposure-dependent. Ivosidenib showed a favorable PK profile in several animal species, along with a clear PK/PD relationship demonstrating 2-HG inhibition that translated well to patients with AML. SIGNIFICANCE STATEMENT: Ivosidenib is a mutant IDH1 (mIDH1) inhibitor approved for the treatment of certain patients with mIDH1 acute myeloid leukemia. In Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys, ivosidenib demonstrated a favorable pharmacokinetic profile, and in female BALB/c mice showed clear dose- and exposure-dependent inhibition of the oncometabolite, d-2-hydroxyglutarate, which is present at abnormal levels in mIDH1 tumors. These findings led to the further development of ivosidenib and are consistent with data from patients with mIDH1 cancers and healthy participants.


Asunto(s)
Glicina/análogos & derivados , Isocitrato Deshidrogenasa/metabolismo , Leucemia Mieloide Aguda , Piridinas/farmacocinética , Animales , Antineoplásicos/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Vías de Eliminación de Fármacos , Interacciones Farmacológicas , Glutaratos/metabolismo , Glicina/farmacocinética , Haplorrinos , Humanos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Ácidos Cetoglutáricos/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Tasa de Depuración Metabólica , Ratones , Mutación Puntual , Unión Proteica , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
Toxicol Appl Pharmacol ; 401: 115103, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32522582

RESUMEN

Small cell lung cancer (SCLC) is a particularly aggressive subset of lung cancer, and identification of new therapeutic options is of significant interest. We recently reported that SCLC cell lines display a specific vulnerability to inhibition of squalene epoxidase (SQLE), an enzyme in the cholesterol biosynthetic pathway that catalyzes the conversion of squalene to 2,3-oxidosqualene. Since it has been reported that SQLE inhibition can result in dermatitis in dogs, we conducted a series of experiments to determine if SQLE inhibitors would be tolerated at exposures predicted to drive maximal efficacy in SCLC tumors. Detailed profiling of the SQLE inhibitor NB-598 showed that dogs did not tolerate predicted efficacious exposures, with dose-limiting toxicity due to gastrointestinal clinical observations, although skin toxicities were also observed. To extend these studies, two SQLE inhibitors, NB-598 and Cmpd-4″, and their structurally inactive analogs, NB-598.ia and Cmpd-4″.ia, were profiled in monkeys. While both active SQLE inhibitors resulted in dose-limiting gastrointestinal toxicity, the structurally similar inactive analogs did not. Collectively, our data demonstrate that significant toxicities arise at exposures well below the predicted levels needed for anti-tumor activity. The on-target nature of the toxicities identified is likely to limit the potential therapeutic utility of SQLE inhibition for the treatment of SCLC.


Asunto(s)
Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/toxicidad , Escualeno-Monooxigenasa/antagonistas & inhibidores , Escualeno-Monooxigenasa/sangre , Animales , Perros , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Macaca fascicularis , Masculino , Piel/efectos de los fármacos , Piel/enzimología , Piel/patología
3.
ACS Med Chem Lett ; 11(2): 101-107, 2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32071674

RESUMEN

Inhibitors of mutant isocitrate dehydrogenase (mIDH) 1 and 2 cancer-associated enzymes prevent the accumulation of the oncometabolite d-2-hydroxyglutarate (2-HG) and are under clinical investigation for the treatment of several cancers harboring an IDH mutation. Herein, we describe the discovery of vorasidenib (AG-881), a potent, oral, brain-penetrant dual inhibitor of both mIDH1 and mIDH2. X-ray cocrystal structures allowed us to characterize the compound binding site, leading to an understanding of the dual mutant inhibition. Furthermore, vorasidenib penetrates the brain of several preclinical species and inhibits 2-HG production in glioma tissue by >97% in an orthotopic glioma mouse model. Vorasidenib represents a novel dual mIDH1/2 inhibitor and is currently in clinical development for the treatment of low-grade mIDH glioma.

4.
Nat Commun ; 10(1): 97, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626872

RESUMEN

Squalene epoxidase (SQLE), also known as squalene monooxygenase, catalyzes the stereospecific conversion of squalene to 2,3(S)-oxidosqualene, a key step in cholesterol biosynthesis. SQLE inhibition is targeted for the treatment of hypercholesteremia, cancer, and fungal infections. However, lack of structure-function understanding has hindered further progression of its inhibitors. We have determined the first three-dimensional high-resolution crystal structures of human SQLE catalytic domain with small molecule inhibitors (2.3 Å and 2.5 Å). Comparison with its unliganded state (3.0 Å) reveals conformational rearrangements upon inhibitor binding, thus allowing deeper interpretation of known structure-activity relationships. We use the human SQLE structure to further understand the specificity of terbinafine, an approved agent targeting fungal SQLE, and to provide the structural insights into terbinafine-resistant mutants encountered in the clinic. Collectively, these findings elucidate the structural basis for the specificity of the epoxidation reaction catalyzed by SQLE and enable further rational development of next-generation inhibitors.


Asunto(s)
Escualeno-Monooxigenasa/química , Escualeno-Monooxigenasa/metabolismo , Animales , Dominio Catalítico , Línea Celular , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Insectos , Conformación Proteica , Dominios Proteicos , Escualeno/metabolismo , Escualeno-Monooxigenasa/antagonistas & inhibidores
5.
Nat Commun ; 10(1): 96, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626880

RESUMEN

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Escualeno-Monooxigenasa/antagonistas & inhibidores , Escualeno-Monooxigenasa/metabolismo , Antineoplásicos/química , Línea Celular Tumoral , Colesterol/biosíntesis , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
6.
ACS Med Chem Lett ; 9(4): 300-305, 2018 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-29670690

RESUMEN

Somatic point mutations at a key arginine residue (R132) within the active site of the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) confer a novel gain of function in cancer cells, resulting in the production of d-2-hydroxyglutarate (2-HG), an oncometabolite. Elevated 2-HG levels are implicated in epigenetic alterations and impaired cellular differentiation. IDH1 mutations have been described in an array of hematologic malignancies and solid tumors. Here, we report the discovery of AG-120 (ivosidenib), an inhibitor of the IDH1 mutant enzyme that exhibits profound 2-HG lowering in tumor models and the ability to effect differentiation of primary patient AML samples ex vivo. Preliminary data from phase 1 clinical trials enrolling patients with cancers harboring an IDH1 mutation indicate that AG-120 has an acceptable safety profile and clinical activity.

7.
Blood ; 130(11): 1347-1356, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28760888

RESUMEN

Pyruvate kinase (PK) deficiency is a rare genetic disease that causes chronic hemolytic anemia. There are currently no targeted therapies for PK deficiency. Here, we describe the identification and characterization of AG-348, an allosteric activator of PK that is currently in clinical trials for the treatment of PK deficiency. We demonstrate that AG-348 can increase the activity of wild-type and mutant PK enzymes in biochemical assays and in patient red blood cells treated ex vivo. These data illustrate the potential for AG-348 to restore the glycolytic pathway activity in patients with PK deficiency and ultimately lead to clinical benefit.


Asunto(s)
Activadores de Enzimas/farmacología , Activadores de Enzimas/uso terapéutico , Eritrocitos/enzimología , Piruvato Quinasa/deficiencia , Piruvato Quinasa/metabolismo , Quinolinas/farmacología , Quinolinas/uso terapéutico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Anemia Hemolítica Congénita no Esferocítica , Animales , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/química , Eritrocitos/efectos de los fármacos , Humanos , Cinética , Ratones , Piperazinas , Piruvato Quinasa/efectos de los fármacos , Errores Innatos del Metabolismo del Piruvato , Quinolinas/química , Proteínas Recombinantes/metabolismo , Sulfonamidas/química , Donantes de Tejidos
10.
Nature ; 513(7516): 110-4, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25043045

RESUMEN

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are among the most common genetic alterations in intrahepatic cholangiocarcinoma (IHCC), a deadly liver cancer. Mutant IDH proteins in IHCC and other malignancies acquire an abnormal enzymatic activity allowing them to convert α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), which inhibits the activity of multiple αKG-dependent dioxygenases, and results in alterations in cell differentiation, survival, and extracellular matrix maturation. However, the molecular pathways by which IDH mutations lead to tumour formation remain unclear. Here we show that mutant IDH blocks liver progenitor cells from undergoing hepatocyte differentiation through the production of 2HG and suppression of HNF-4α, a master regulator of hepatocyte identity and quiescence. Correspondingly, genetically engineered mouse models expressing mutant IDH in the adult liver show an aberrant response to hepatic injury, characterized by HNF-4α silencing, impaired hepatocyte differentiation, and markedly elevated levels of cell proliferation. Moreover, IDH and Kras mutations, genetic alterations that co-exist in a subset of human IHCCs, cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic IHCC. These studies provide a functional link between IDH mutations, hepatic cell fate, and IHCC pathogenesis, and present a novel genetically engineered mouse model of IDH-driven malignancy.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Diferenciación Celular/genética , Colangiocarcinoma/patología , Factor Nuclear 4 del Hepatocito/antagonistas & inhibidores , Hepatocitos/patología , Isocitrato Deshidrogenasa/genética , Proteínas Mutantes/metabolismo , Animales , Neoplasias de los Conductos Biliares/enzimología , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/enzimología , Conductos Biliares Intrahepáticos/patología , División Celular/genética , Linaje de la Célula/genética , Colangiocarcinoma/enzimología , Colangiocarcinoma/genética , Modelos Animales de Enfermedad , Femenino , Glutaratos/metabolismo , Factor Nuclear 4 del Hepatocito/biosíntesis , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Isocitrato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Mutantes/genética , Mutación/genética , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Células Madre/patología , Proteínas ras/genética , Proteínas ras/metabolismo
11.
Bioorg Med Chem ; 22(7): 2303-10, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24588962

RESUMEN

The ribonucleotide reductase (RNR) enzyme is a heteromer of RRM1 and RRM2 subunits. The active enzyme catalyzes de novo reduction of ribonucleotides to generate deoxyribonucleotides (dNTPs), which are required for DNA replication and DNA repair processes. Complexity in the generation of physiologically relevant, active RRM1/RRM2 heterodimers was perceived as limiting to the identification of selective RRM1 inhibitors by high-throughput screening of compound libraries and led us to seek alternative methods to identify lead series. In short, we found that gemcitabine, as its diphosphate metabolite, represents one of the few described active site inhibitors of RRM1. We herein describe the identification of novel 5'-amino gemcitabine analogs as potent RRM1 inhibitors through in-cell phenotypic screening.


Asunto(s)
Desoxicitidina/análogos & derivados , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Línea Celular Tumoral , Desoxicitidina/química , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Humanos , Espectroscopía de Resonancia Magnética , Estructura Molecular , Ribonucleósido Difosfato Reductasa , Relación Estructura-Actividad , Gemcitabina
12.
J Biol Chem ; 289(20): 13717-25, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24668804

RESUMEN

Two mutant forms (R132H and R132C) of isocitrate dehydrogenase 1 (IDH1) have been associated with a number of cancers including glioblastoma and acute myeloid leukemia. These mutations confer a neomorphic activity of 2-hydroxyglutarate (2-HG) production, and 2-HG has previously been implicated as an oncometabolite. Inhibitors of mutant IDH1 can potentially be used to treat these diseases. In this study, we investigated the mechanism of action of a newly discovered inhibitor, ML309, using biochemical, cellular, and biophysical approaches. Substrate binding and product inhibition studies helped to further elucidate the IDH1 R132H catalytic cycle. This rapidly equilibrating inhibitor is active in both biochemical and cellular assays. The (+) isomer is active (IC50 = 68 nm), whereas the (-) isomer is over 400-fold less active (IC50 = 29 µm) for IDH1 R132H inhibition. IDH1 R132C was similarly inhibited by (+)-ML309. WT IDH1 was largely unaffected by (+)-ML309 (IC50 >36 µm). Kinetic analyses combined with microscale thermophoresis and surface plasmon resonance indicate that this reversible inhibitor binds to IDH1 R132H competitively with respect to α-ketoglutarate and uncompetitively with respect to NADPH. A reaction scheme for IDH1 R132H inhibition by ML309 is proposed in which ML309 binds to IDH1 R132H after formation of the IDH1 R132H NADPH complex. ML309 was also able to inhibit 2-HG production in a glioblastoma cell line (IC50 = 250 nm) and had minimal cytotoxicity. In the presence of racemic ML309, 2-HG levels drop rapidly. This drop was sustained until 48 h, at which point the compound was washed out and 2-HG levels recovered.


Asunto(s)
Acetamidas/farmacología , Bencimidazoles/farmacología , Fenómenos Biofísicos , Inhibidores Enzimáticos/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación , Acetamidas/metabolismo , Acetamidas/farmacocinética , Animales , Bencimidazoles/metabolismo , Bencimidazoles/farmacocinética , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Humanos , Isocitrato Deshidrogenasa/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/farmacología
13.
Bioorg Med Chem Lett ; 24(1): 199-203, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24332088

RESUMEN

A new class of quinoline-based kinase inhibitors has been discovered that both disrupt cyclin dependent 2 (CDK2) interaction with its cyclin A subunit and act as ATP competitive inhibitors. The key strategy for discovering this class of protein-protein disrupter compounds was to screen the monomer CDK2 in an affinity-selection/mass spectrometry-based technique and to perform secondary assays that identified compounds that bound only to the inactive CDK2 monomer and not the active CDK2/cyclin A heterodimer. Through a series of chemical modifications the affinity (Kd) of the original hit improved from 1 to 0.005µM.


Asunto(s)
Ciclina A/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Cristalografía por Rayos X , Ciclina A/química , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Quinolinas/química , Relación Estructura-Actividad
14.
Science ; 340(6132): 626-30, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23558169

RESUMEN

The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant--but not IDH1-wild-type--glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


Asunto(s)
Bencenoacetamidas/farmacología , Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Glioma/enzimología , Glioma/patología , Imidazoles/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Animales , Bencenoacetamidas/administración & dosificación , Bencenoacetamidas/toxicidad , Diferenciación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Inhibidores Enzimáticos/toxicidad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/genética , Glutaratos/metabolismo , Histonas/metabolismo , Imidazoles/administración & dosificación , Imidazoles/toxicidad , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/metabolismo , Metilación , Ratones , Ratones SCID , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Multimerización de Proteína , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Science ; 340(6132): 622-6, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23558173

RESUMEN

A number of human cancers harbor somatic point mutations in the genes encoding isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2). These mutations alter residues in the enzyme active sites and confer a gain-of-function in cancer cells, resulting in the accumulation and secretion of the oncometabolite (R)-2-hydroxyglutarate (2HG). We developed a small molecule, AGI-6780, that potently and selectively inhibits the tumor-associated mutant IDH2/R140Q. A crystal structure of AGI-6780 complexed with IDH2/R140Q revealed that the inhibitor binds in an allosteric manner at the dimer interface. The results of steady-state enzymology analysis were consistent with allostery and slow-tight binding by AGI-6780. Treatment with AGI-6780 induced differentiation of TF-1 erythroleukemia and primary human acute myelogenous leukemia cells in vitro. These data provide proof-of-concept that inhibitors targeting mutant IDH2/R140Q could have potential applications as a differentiation therapy for cancer.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hematopoyesis/efectos de los fármacos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/enzimología , Compuestos de Fenilurea/farmacología , Sulfonamidas/farmacología , Sitio Alostérico , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Eritropoyesis/efectos de los fármacos , Regulación Leucémica de la Expresión Génica , Glutaratos/metabolismo , Humanos , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/metabolismo , Leucemia Eritroblástica Aguda , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Compuestos de Fenilurea/química , Compuestos de Fenilurea/metabolismo , Mutación Puntual , Multimerización de Proteína , Estructura Secundaria de Proteína , Bibliotecas de Moléculas Pequeñas , Sulfonamidas/química , Sulfonamidas/metabolismo
16.
ACS Med Chem Lett ; 3(10): 850-5, 2012 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24900389

RESUMEN

Optimization of a series of R132H IDH1 inhibitors from a high throughput screen led to the first potent molecules that show robust tumor 2-HG inhibition in a xenograft model. Compound 35 shows good potency in the U87 R132H cell based assay and ∼90% tumor 2-HG inhibition in the corresponding mouse xenograft model following BID dosing. The magnitude and duration of tumor 2-HG inhibition correlates with free plasma concentration.

17.
Bioorg Med Chem Lett ; 21(10): 3172-6, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21458257

RESUMEN

TNF-α converting enzyme (TACE) inhibitors are promising agents to treat inflammatory disorders and cancer. We have investigated novel tartrate diamide TACE inhibitors where the tartrate core binds to zinc in a unique tridentate fashion. Incorporating (R)-2-(2-N-alkylaminothiazol-4-yl)pyrrolidines into the left hand side amide of the tartrate scaffold led to the discovery of potent and selective TACE inhibitors, some of which exhibited good rat oral bioavailability.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Amidas/farmacología , Inhibidores Enzimáticos/farmacología , Pirrolidinas/química , Tartratos/química , Proteína ADAM17 , Amidas/síntesis química , Amidas/química , Animales , Disponibilidad Biológica , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Estructura Molecular , Ratas
18.
Bioorg Med Chem Lett ; 20(24): 7283-7, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21106451

RESUMEN

Our research on hydantoin based TNF-α converting enzyme (TACE) inhibitors has led to an acetylene containing series that demonstrates sub-nanomolar potency (K(i)) as well as excellent activity in human whole blood. These studies led to the discovery of highly potent TACE inhibitors with good DMPK profiles.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Antiinflamatorios/química , Artritis Reumatoide/tratamiento farmacológico , Inhibidores de Proteasas/química , Proteínas ADAM/metabolismo , Proteína ADAM17 , Acetileno/análogos & derivados , Acetileno/farmacocinética , Acetileno/uso terapéutico , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Perros , Haplorrinos , Humanos , Inhibidores de Proteasas/farmacocinética , Inhibidores de Proteasas/uso terapéutico , Ratas
20.
Bioorg Med Chem Lett ; 20(6): 1877-80, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20172725

RESUMEN

We disclose inhibitors of TNF-alpha converting enzyme (TACE) designed around a hydantoin zinc binding moiety. Crystal structures of inhibitors bound to TACE revealed monodentate coordination of the hydantoin to the zinc. SAR, X-ray, and modeling designs are described. To our knowledge, these are the first reported X-ray structures of TACE with a hydantoin zinc ligand.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Hidantoínas/farmacología , Proteína ADAM17 , Inhibidores Enzimáticos/química , Hidantoínas/química , Enlace de Hidrógeno , Modelos Moleculares , Relación Estructura-Actividad , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...