Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Bioorg Med Chem Lett ; 29(16): 2428-2436, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31133531

RESUMEN

Treatment of hepatitis C virus (HCV) infection has been historically challenging due the high viral genetic complexity wherein there are eight distinct genotypes and at least 86 viral subtypes. While HCV NS3/4A protease inhibitors are an established treatment option for genotype 1 infection, limited coverage of genotypes 2 and/or 3 combined with serum alanine transaminase (ALT) elevations for some compounds has limited the broad utility of this therapeutic class. Our discovery efforts were focused on identifying an NS3/4A protease inhibitor with pan-genotypic antiviral activity, improved coverage of resistance associated substitutions, and a decreased risk of hepatotoxicity. Towards this goal, distinct interactions with the conserved catalytic triad of the NS3/4A protease were identified that improved genotype 3 antiviral activity. We further discovered that protein adduct formation strongly correlated with clinical ALT elevation for this therapeutic class. Improving metabolic stability and decreasing protein adduct formation through structural modifications ultimately resulted in voxilaprevir. Voxilaprevir, in combination with sofosbuvir and velpatasvir, has demonstrated pan-genotypic antiviral clinical activity. Furthermore, hepatotoxicity was not observed in Phase 3 clinical trials with voxilaprevir, consistent with our design strategy. Vosevi® (sofosbuvir, velpatasvir, and voxilaprevir) is now an approved pan-genotypic treatment option for the most difficult-to-cure individuals who have previously failed direct acting antiviral therapy.


Asunto(s)
Antivirales/farmacología , Carbamatos/química , Descubrimiento de Drogas , Hepacivirus/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Inhibidores de Proteasas/farmacología , Sofosbuvir/química , Sulfonamidas/química , Sulfonamidas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Ácidos Aminoisobutíricos , Antivirales/síntesis química , Antivirales/química , Ciclopropanos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Hepacivirus/genética , Humanos , Lactamas Macrocíclicas , Leucina/análogos & derivados , Compuestos Macrocíclicos/síntesis química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Prolina/análogos & derivados , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Quinoxalinas , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
2.
Bioorg Med Chem ; 27(3): 457-469, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30606676

RESUMEN

The bromodomain and extra-terminal (BET) family of proteins, consisting of the bromodomains containing protein 2 (BRD2), BRD3, BRD4, and the testis-specific BRDT, are key epigenetic regulators of gene transcription and has emerged as an attractive target for anticancer therapy. Herein, we describe the discovery of a novel potent BET bromodomain inhibitor, using a systematic structure-based approach focused on improving potency, metabolic stability, and permeability. The optimized dimethylisoxazole aryl-benzimidazole inhibitor exhibited high potency towards BRD4 and related BET proteins in biochemical and cell-based assays and inhibited tumor growth in two proof-of-concept preclinical animal models.


Asunto(s)
Bencimidazoles/farmacología , Descubrimiento de Drogas , Isoxazoles/farmacología , Mieloma Múltiple/tratamiento farmacológico , Factores de Transcripción/antagonistas & inhibidores , Administración Oral , Animales , Bencimidazoles/química , Bencimidazoles/metabolismo , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Isoxazoles/administración & dosificación , Isoxazoles/química , Isoxazoles/metabolismo , Ratones , Estructura Molecular , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Dominios Proteicos/efectos de los fármacos , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
3.
Bioorg Med Chem Lett ; 28(3): 541-546, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29254643

RESUMEN

We hereby disclose the discovery of inhibitors of CaMKII (7h and 7i) that are highly potent in rat ventricular myocytes, selective against hERG and other off-target kinases, while possessing good CaMKII tissue isoform selectivity (cardiac γ/δ vs. neuronal α/ß). In vitro and in vivo ADME/PK studies demonstrated the suitability of these CaMKII inhibitors for PO (7h rat F = 73%) and IV pharmacological studies.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores de Proteínas Quinasas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 26(24): 5926-5930, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27876318

RESUMEN

Herein, we describe the synthesis of Pyk2 inhibitors via macrocyclization of FAK and dual Pyk2-FAK inhibitors. We identified macrocycle 25a as a highly potent Pyk2 inhibitor (IC50=0.7nM), with ∼175-fold improvement in Pyk2 potency as compared to its acyclic counterpart. In many cases, macrocyclization improved Pyk2 potency while weakening FAK potency, thereby improving the Pyk2/FAK selectivity ratio for this structural class of inhibitors. Various macrocyclic linkers were studied in an attempt to optimize Pyk2 selectivity. We observed macrocyclic atropisomerism during the synthesis of 19-membered macrocycles 10a-d, and successfully obtained crystallographic evidence of one atropisomer (10a-AtropB) preferentially bound to Pyk2.


Asunto(s)
Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Proteínas Tirosina Quinasas/farmacología , Animales , Ciclización , Perros , Relación Dosis-Respuesta a Droga , Quinasa 2 de Adhesión Focal/metabolismo , Humanos , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Sustancias Macromoleculares/farmacología , Ratones , Modelos Moleculares , Estructura Molecular , Proteínas Tirosina Quinasas/síntesis química , Proteínas Tirosina Quinasas/química , Relación Estructura-Actividad
5.
Bioorg Med Chem Lett ; 26(13): 3207-3211, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27038498

RESUMEN

Previously we disclosed the discovery of potent Late INa current inhibitor 2 (GS-458967, IC50 of 333nM) that has a good separation of late versus peak Nav1.5 current, but did not have a favorable CNS safety window due to high brain penetration (3-fold higher partitioning into brain vs plasma) coupled with potent inhibition of brain sodium channel isoforms (Nav1.1, 1.2, 1.3). We increased the polar surface area from 50 to 84Å(2) by adding a carbonyl to the core and an oxadiazole ring resulting in 3 GS-462808 that had lower brain penetration and serendipitously lower activity at the brain isoforms. Compound 3 has an improved CNS window (>20 rat and dog) relative to 2, and improved anti-ischemic potency relative to ranolazine. The development of 3 was not pursued due to liver lesions in 7day rat toxicology studies.


Asunto(s)
Azoles/farmacología , Descubrimiento de Drogas , Corazón/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Piridinas/farmacología , Ranolazina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Animales , Azoles/síntesis química , Azoles/química , Perros , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Haplorrinos , Humanos , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Conejos , Ranolazina/síntesis química , Ranolazina/química , Ratas , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad
6.
Bioorg Med Chem Lett ; 26(13): 3202-3206, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27080178

RESUMEN

We started with a medium throughput screen of heterocyclic compounds without basic amine groups to avoid hERG and ß-blocker activity and identified [1,2,4]triazolo[4,3-a]pyridine as an early lead. Optimization of substituents for Late INa current inhibition and lack of Peak INa inhibition led to the discovery of 4h (GS-458967) with improved anti-arrhythmic activity relative to ranolazine. Unfortunately, 4h demonstrated use dependent block across the sodium isoforms including the central and peripheral nervous system isoforms that is consistent with its low therapeutic index (approximately 5-fold in rat, 3-fold in dog). Compound 4h represents our initial foray into a 2nd generation Late INa inhibitor program and is an important proof-of-concept compound. We will provide additional reports on addressing the CNS challenge in a follow-up communication.


Asunto(s)
Descubrimiento de Drogas , Corazón/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Piridinas/farmacología , Ranolazina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Triazoles/farmacología , Animales , Células CACO-2 , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Macaca fascicularis , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Conejos , Ranolazina/síntesis química , Ranolazina/química , Ratas , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/química , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química
7.
Proc Natl Acad Sci U S A ; 109(44): 18102-7, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23077250

RESUMEN

Human nociceptive voltage-gated sodium channel (Na(v)1.7), a target of significant interest for the development of antinociceptive agents, is blocked by low nanomolar concentrations of (-)-tetrodotoxin(TTX) but not (+)-saxitoxin (STX) and (+)-gonyautoxin-III (GTX-III). These findings question the long-accepted view that the 1.7 isoform is both tetrodotoxin- and saxitoxin-sensitive and identify the outer pore region of the channel as a possible target for the design of Na(v)1.7-selective inhibitors. Single- and double-point amino acid mutagenesis studies along with whole-cell electrophysiology recordings establish two domain III residues (T1398 and I1399), which occur as methionine and aspartate in other Na(v) isoforms, as critical determinants of STX and gonyautoxin-III binding affinity. An advanced homology model of the Na(v) pore region is used to provide a structural rationalization for these surprising results.


Asunto(s)
Activación del Canal Iónico , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Saxitoxina/toxicidad , Tetrodotoxina/toxicidad , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Humanos , Metionina/química , Metionina/metabolismo , Mutagénesis , Canal de Sodio Activado por Voltaje NAV1.7/química , Canal de Sodio Activado por Voltaje NAV1.7/fisiología
8.
J Pharmacol Exp Ther ; 344(1): 23-32, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23010360

RESUMEN

Inhibition of cardiac late sodium current (late I(Na)) is a strategy to suppress arrhythmias and sodium-dependent calcium overload associated with myocardial ischemia and heart failure. Current inhibitors of late I(Na) are unselective and can be proarrhythmic. This study introduces GS967 (6-[4-(trifluoromethoxy)phenyl]-3-(trifluoromethyl)-[1,2,4]triazolo[4,3-a]pyridine), a potent and selective inhibitor of late I(Na), and demonstrates its effectiveness to suppress ventricular arrhythmias. The effects of GS967 on rabbit ventricular myocyte ion channel currents and action potentials were determined. Anti-arrhythmic actions of GS967 were characterized in ex vivo and in vivo rabbit models of reduced repolarization reserve and ischemia. GS967 inhibited Anemonia sulcata toxin II (ATX-II)-induced late I(Na) in ventricular myocytes and isolated hearts with IC(50) values of 0.13 and 0.21 µM, respectively. Reduction of peak I(Na) by GS967 was minimal at a holding potential of -120 mV but increased at -80 mV. GS967 did not prolong action potential duration or the QRS interval. GS967 prevented and reversed proarrhythmic effects (afterdepolarizations and torsades de pointes) of the late I(Na) enhancer ATX-II and the I(Kr) inhibitor E-4031 in isolated ventricular myocytes and hearts. GS967 significantly attenuated the proarrhythmic effects of methoxamine+clofilium and suppressed ischemia-induced arrhythmias. GS967 was more potent and effective to reduce late I(Na) and arrhythmias than either flecainide or ranolazine. Results of all studies and assays of binding and activity of GS967 at numerous receptors, transporters, and enzymes indicated that GS967 selectively inhibited late I(Na). In summary, GS967 selectively suppressed late I(Na) and prevented and/or reduced the incidence of experimentally induced arrhythmias in rabbit myocytes and hearts.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Cardiotónicos/farmacología , Piridinas/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Triazoles/farmacología , Acetanilidas/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Venenos de Cnidarios/farmacología , Femenino , Flecainida/farmacología , Sistema de Conducción Cardíaco/efectos de los fármacos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Mutación/fisiología , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Piperazinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Compuestos de Amonio Cuaternario/farmacología , Conejos , Ranolazina
9.
J Med Chem ; 63(18): 10188-10203, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32407112

RESUMEN

Toll-like receptor 8 (TLR8) recognizes pathogen-derived single-stranded RNA fragments to trigger innate and adaptive immune responses. Chronic hepatitis B (CHB) is associated with a dysfunctional immune response, and therefore a selective TLR8 agonist may be an effective treatment option. Structure-based optimization of a dual TLR7/8 agonist led to the identification of the selective TLR8 clinical candidate (R)-2-((2-amino-7-fluoropyrido[3,2-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol (GS-9688, (R)-7). Potent TLR8 agonism (IL-12p40 EC50 = 220 nM) and >100-fold TLR7 selectivity (IFN-α EC50 > 50 µM) was observed in human peripheral blood mononuclear cells (PBMCs). The TLR8-ectodomain:(R)-7 complex confirmed TLR8 binding and a direct ligand interaction with TLR8 residue Asp545. Oral (R)-7 had good absorption and high first pass clearance in preclinical species. A reduction in viral markers was observed in HBV-infected primary human hepatocytes treated with media from PBMCs stimulated with (R)-7, supporting the clinical development of (R)-7 for the treatment of CHB.


Asunto(s)
Antivirales/farmacología , Hepatitis B Crónica/tratamiento farmacológico , Hexanoles/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Receptor Toll-Like 8/agonistas , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/síntesis química , Antivirales/metabolismo , Cristalografía por Rayos X , Perros , Descubrimiento de Drogas , Virus de la Hepatitis B/efectos de los fármacos , Hexanoles/administración & dosificación , Hexanoles/síntesis química , Hexanoles/metabolismo , Humanos , Macaca fascicularis , Estructura Molecular , Dominios Proteicos , Piridinas/administración & dosificación , Piridinas/síntesis química , Piridinas/metabolismo , Pirimidinas/administración & dosificación , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Ratas , Relación Estructura-Actividad , Receptor Toll-Like 8/metabolismo
10.
Alcohol Clin Exp Res ; 33(11): 1935-44, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19673742

RESUMEN

BACKGROUND: Inherited human aldehyde dehydrogenase 2 (ALDH-2) deficiency reduces the risk for alcoholism. Kudzu plants and extracts have been used for 1,000 years in traditional Chinese medicine to treat alcoholism. Kudzu contains daidzin, which inhibits ALDH-2 and suppresses heavy drinking in rodents. Decreased drinking due to ALDH-2 inhibition is attributed to aversive properties of acetaldehyde accumulated during alcohol consumption. However, daidzin can reduce drinking in some rodents without necessarily increasing acetaldehyde. Therefore, a selective ALDH-2 inhibitor might affect other metabolic factors involved in regulating drinking. METHODS: Aldehyde dehydrogenase 2 inhibitors were synthesized based on the co-crystal structure of ALDH-2 and daidzin. We tested the efficacy of a highly selective reversible ALDH-2 inhibitor, CVT-10216, in models of moderate and high alcohol drinking rats. We studied 2-bottle choice and deprivation-induced drinking paradigms in Fawn Hooded (FH) rats, operant self-administration in Long Evans (LE), FH, and inbred P (iP) rats and in cue-induced reinstatement in iP rats. We also assayed blood acetaldehyde levels as well as dopamine (DA) release in the nucleus accumbens (NAc) and tested possible rewarding/aversive effects of the inhibitor in a conditioned place preference (CPP) paradigm. RESULTS: CVT-10216 increases acetaldehyde after alcohol gavage and inhibits 2-bottle choice alcohol intake in heavy drinking rodents, including deprivation-induced drinking. Moreover, CVT-10216 also prevents operant self-administration and eliminates cue-induced reinstatement of alcohol seeking even when alcohol is not available (i.e., no acetaldehyde). Alcohol stimulates DA release in the NAc, which is thought to contribute to increased drinking and relapse in alcoholism. CVT-10216 prevents alcohol-induced increases in NAc DA without changing basal levels. CVT-10216 does not show rewarding or aversive properties in the CPP paradigm at therapeutic doses. CONCLUSION: Our findings suggest that selective reversible ALDH-2 inhibitors may have therapeutic potential to reduce excessive drinking and to suppress relapse in abstinent alcoholics.


Asunto(s)
Disuasivos de Alcohol , Consumo de Bebidas Alcohólicas/psicología , Aldehído Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Isoflavonas/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Acetaldehído/sangre , Aldehído Deshidrogenasa Mitocondrial , Animales , Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Dopamina/fisiología , Extinción Psicológica/efectos de los fármacos , Masculino , Microdiálisis , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Pueraria/química , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Recurrencia , Autoadministración
11.
Bioorg Med Chem Lett ; 19(7): 2048-52, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19249203

RESUMEN

We identified a series of structurally novel SCD (Delta9 desaturase) inhibitors via high-throughput screening and follow-up SAR studies. Modification of the central bicyclic scaffold has proven key to our potency optimization effort. The most potent analog (8g) had IC(50) value of 50 pM in a HEPG2 SCD assay and has been shown to be metabolically stable and selective against Delta5 and Delta6 desaturases.


Asunto(s)
Inhibidores Enzimáticos/química , Pteridinas/química , Quinoxalinas/química , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Microsomas/metabolismo , Pteridinas/metabolismo , Pteridinas/farmacología , Quinoxalinas/farmacología , Ratas , Estearoil-CoA Desaturasa/metabolismo , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 19(15): 4070-4, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19577469

RESUMEN

Two structurally distinct series of SCD (Delta9 desaturase) inhibitors (1 and 2) have been previously reported by our group. In the present work, we merged the structural features of the two series. This led to the discovery of compound 5b (CVT-12,012) which is highly potent in a human cell-based (HEPG2) SCD assay (IC(50)=6nM). This compound has 78% oral bioavailability in rats and is preferentially distributed into liver (76 times vs plasma) with relatively low brain penetration. In a five-day study (sucrose fed rats) compound 5b significantly reduced SCD activity in a dose-dependent manner as determined by GC analysis of fatty acid composition in plasma and liver, and significantly reduced liver triglycerides versus the control group ( approximately 50%).


Asunto(s)
Química Farmacéutica/métodos , Hígado/enzimología , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Acetamidas/química , Administración Oral , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ácidos Grasos/química , Humanos , Concentración 50 Inhibidora , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Microsomas/metabolismo , Ratas , Ratas Sprague-Dawley , Estearoil-CoA Desaturasa/química , Sacarosa/química , Triglicéridos/química
13.
Bioorg Med Chem Lett ; 19(11): 3050-3, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19394219

RESUMEN

We discovered a structurally novel SCD (Delta9 desaturase) inhibitor 4a (CVT-11,563) that has 119 nM potency in a human cell-based (HEPG2) SCD assay and selectivity against Delta5 and Delta6 desaturases. This compound has 90% oral bioavailability (rat) and excellent plasma exposure (dAUC 935 ng h/mL). Additionally, 4a shows moderately selective liver distribution (three times vs plasma and adipose tissue) and relatively low brain penetration. In a five-day study (high sucrose diet, rat) compound 4a significantly reduced SCD activity as determined by GC analysis of fatty acid composition in plasma and liver. We describe the discovery of 4a from HTS hit 1 followed by scaffold replacement and SAR studies focused on DMPK properties.


Asunto(s)
Compuestos de Bencilo/química , Inhibidores Enzimáticos/química , Pirimidinonas/química , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Administración Oral , Animales , Compuestos de Bencilo/síntesis química , Compuestos de Bencilo/farmacocinética , Línea Celular Tumoral , Carbohidratos de la Dieta/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Microsomas Hepáticos/metabolismo , Pirimidinonas/síntesis química , Pirimidinonas/farmacocinética , Ratas , Ratas Sprague-Dawley , Estearoil-CoA Desaturasa/metabolismo , Distribución Tisular
14.
Handb Exp Pharmacol ; (193): 25-58, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19639278

RESUMEN

Intense efforts of many pharmaceutical companies and academicians in the A(1) adenosine receptor (AR) field have led to the discovery of clinical candidates that are antagonists, agonists, and allosteric enhancers. The A(1)AR antagonists currently in clinical development are KW3902, BG9928, and SLV320. All three have high affinity for the human (h) A(1)AR subtype (hA(1) K (i) < 10 nM), > 200-fold selectivity over the hA(2A) subtype, and demonstrate renal protective effects in multiple animal models of disease and pharmacologic effects in human subjects. In the A(1)AR agonist area, clinical candidates have been discovered for the following conditions: atrial arrhythmias (tecadenoson, selodenoson and PJ-875); Type II diabetes and insulin sensitizing agents (GR79236, ARA, RPR-749, and CVT-3619); and angina (BAY 68-4986). The challenges associated with the development of any A(1)AR agonist are to obtain tissue-specific effects but avoid off-target tissue side effects and A(1)AR desensitization leading to tachyphylaxis. For the IV antiarrhythmic agents that act as ventricular rate control agents, a selective response can be accomplished by careful IV dosing paradigms. The treatment of type II diabetes using A(1)AR agonists in the clinic has met with limited success due to cardiovascular side effects and a well-defined desensitization of full agonists in human trials (GR79236, ARA, and RPR 749). However, new partial A(1)AR agonists are in development, including CVT-3619 hA(1) AR K(i) = 55nM, hA(2A:hA2B:hA(3))1,000:20, CV Therapeutics), which have the potential to provide enhanced insulin sensitivity without cardiovascular side effects and tachyphylaxis. The nonnucleosidic A(1)AR agonist BAY 68-4986 (capadenoson) represents a novel approach to angina wherein both animal studies and early human studies are promising. T-62 is an A(1)AR allosteric enhancer that is currently being evaluated in clinical trials as a potential treatment for neuropathic pain. The challenges associated with developing A(1)AR antagonists, agonists, or allosteric enhancers for therapeutic intervention are now well defined in humans. Significant progress has been made in identifying A(1)AR antagonists for the treatment of edema associated with congestive heart failure (CHF), A(1)AR agonists for the treatment of atrial arrhythmias, type II diabetes and angina, and A(1)AR allosteric enhancers for the treatment of neuropathic pain.


Asunto(s)
Receptor de Adenosina A1/efectos de los fármacos , Agonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A1 , Regulación Alostérica , Animales , Antiarrítmicos/farmacología , Ciclohexanos/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Lipólisis/efectos de los fármacos , Xantinas/farmacología
15.
Handb Exp Pharmacol ; (193): 99-122, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19639280

RESUMEN

A selective, high-affinity A(2B) adenosine receptor (AR) antagonist will be useful as a pharmacological tool to help determine the role of the A(2B)AR in inflammatory diseases and angiogenic diseases. Based on early A(2B)AR-selective ligands with nonoptimal pharmaceutical properties, such as 15 (MRS 1754: K(i)(hA(2B)) = 2 nM; K(i)(hA(1)) = 403 nM; K(i)(hA(2A)) = 503 NM, and K(i)(hA(3)) = 570 nM), several groups have discovered second-generation A(2B)AR ligands that are suitable for development. Scientists at CV Therapeutics have discovered the selective, high-affinity A(2B)AR antagonist 22, a 8-(4-pyrazolyl)-xanthine derivative, (CVT-6883, K(i)(hA(2B)) = 22 nM; K(i)(hA(1)) = 1,940 nM; K(i)(hA(2A)) = 3,280; and K(i)(hA(3)) = 1,070 nM). Compound 22 has demonstrated favorable pharmacokinetic (PK) properties (T(1/2) = 4 h and F > 35% rat), and it is a functional antagonist at the A(2B)AR(K (B) = 6 nM). In a mouse model of asthma, compound 22 demonstrated a dose-dependent efficacy supporting the role of the A(2B)AR in asthma. In two Phase I clinical trails, 22 (CVT-6883) was found to be safe, well tolerated, and suitable for once-daily dosing. Baraldi et al. have independently discovered a selective, high-affinity A(2B)AR antagonist, 30 (MRE2029F20), 8-(5-pyrazolyl)-xanthine (K(i)(hA(2B)) = 5.5 nM; K(i)(hA(1)) = 200 nM; K(i)(hA(2A), A(3)) > 1,000, that has been selected for development in conjunction with King Pharmaceuticals. Compound 30 has been demonstrated to be a functional antagonist of the A(2B)AR, and it has been radiolabeled for use in pharmacological studies. A third compound, 58 (LAS-38096), is a 2-aminopyrimidine derivative (discovered by the Almirall group) that has high A(2B)AR affinity and selectivity (K(i)(hA(2B)) = 17 nM; K(i)(hA(1)) > 1,000 nM; K(i)(hA(2A)) > 2,500; and K(i)(hA(3)) > 1,000 nM), and 58 has been moved into preclinical safety testing. A fourth selective, high-affinity A(2B)AR antagonist, 54 (OSIP339391 K(i))(hA(2B)) = 0.5 nM; K(i))(hA(1)) = 37 nM; K(i))(hA(2A)) = 328; and K(i))(hA(3)) = 450 nm) was discovered by the OSI group. The three highly selective, high-affinity A(2B)AR antagonists that have been selected for development should prove useful in subsequent clinical trials that will establish the role of the A(2B)ARs in various disease states.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Adenina/farmacología , Aminopiridinas/farmacología , Animales , Asma/etiología , Descubrimiento de Drogas , Humanos , Pirazinas/farmacología , Pirimidinas/farmacología , Receptor de Adenosina A2B/fisiología , Xantinas/farmacología
16.
J Med Chem ; 51(7): 2267-78, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18321039

RESUMEN

Recently, we have reported a series of new 1,3-symmetrically (R 1 = R 3) substituted xanthines ( 3 and 4) which have high affinity and selectivity for the human adenosine A 2B receptors (hA(2B)-AdoR). Unfortunately, this class of compounds had poor pharmacokinetic properties. This prompted us to investigate the effect of differential alkyl substitution at the N-1 and N-3 positions ( N 1-R not equal to N 3-R) on A(2B)-AdoR affinity and selectivity; we had the dual objectives of enhancing affinity and selectivity for the A(2B)-AdoR, as well as improving oral bioavailability. This effort has led to the discovery of compound 62, that displayed high affinity and selectivity for the hA(2B)-AdoR (K(i) = 22 nM). In addition, compound 62 showed high functional potency in inhibiting the accumulation of cyclic adenosine monophosphate induced by 5'- N-ethylcarboxamidoadenosine in HEK-A(2B)-AdoR and NIH3T3 cells with K(B) values of 6 and 2 nM, respectively. In a single ascending-dose phase I clinical study, compound 62 had no serious adverse events and was well tolerated.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Xantinas/síntesis química , Xantinas/farmacología , Animales , Sitios de Unión , Línea Celular , AMP Cíclico/análisis , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Ratones , Estructura Molecular , Células 3T3 NIH , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Xantinas/química
17.
Bioorg Med Chem Lett ; 18(4): 1397-401, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18226896

RESUMEN

A series of N-1 monosubstituted 8-pyrazolyl xanthines have been synthesized and evaluated for their affinity for the adenosine receptors (AdoRs). We have discovered two compounds 18 (CVT-7124) and 28 (CVT-6694) that display good affinity for the A(2B) AdoR (K(i)=6 nM and 7 nM, respectively) and greater selectivity for the human A(1), A(2A), and A(3) AdoRs (>1000-, >830-, and >1500-fold; >850-, >700-, and >1280-fold, respectively). CVT-6694 has been shown to block the release of interleukin-6 and monocyte chemotactic protein-1 from bronchial smooth muscle cells (BSMC), a process believed to be promoted by activation of A(2B) AdoR.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Pirazoles/farmacología , Xantinas/farmacología , Animales , Unión Competitiva , Células CHO , Cricetinae , Cricetulus , Humanos , Cinética , Pirazoles/síntesis química , Pirazoles/química , Receptor de Adenosina A2B/metabolismo , Especificidad por Sustrato , Uracilo/análogos & derivados , Uracilo/química , Xantinas/síntesis química , Xantinas/química
18.
Naunyn Schmiedebergs Arch Pharmacol ; 375(2): 133-44, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17310264

RESUMEN

Antagonists of adenosine A2A receptors (A2A -antagonists) with different chemical structures have been developed by several pharmaceutical companies for the potential treatment of Parkinson's disease. Pharmacological characterization of these antagonists was incomplete, and different assay conditions were used in different labs. Therefore, we characterized the potencies, selectivities, and pharmacokinetic profiles of six prototypical A2A -antagonists. Displacements of [3H]MSX-2 and of [3H]CGS21680 binding to the human cloned and rat A2A receptors were performed. The rank order of potency of antagonists to displace [(3)H]MSX-2 binding to the human A2A was SCH58261 > or = Biogen-34 > or = Ver-6623 > or = MSX-2 > KW-6002 > > DMPX. For the rat striatal A2A, the order of potency was Biogen-34 > or = SCH58261 > or = Ver-6623 > or = MSX-2 > or = KW-6002 > > DMPX. SCH58261 was the most potent antagonist of the human A2A with a K(i) value of 4 nM, whereas Biogen-34 was the most potent antagonist of the rat A2A with a K(i) value of 1.2 nM. Similar results were obtained from cAMP assays. Selectivities of A2A-antagonists were determined using radioligands [3H]DPCPX, [3H]ZM241385, and [125I]-AB-MECA for A1, A2B, and A3 receptors, respectively. KW-6002 and Biogen-34 exhibited the highest selectivity for A2A vs A1 (human and rat), respectively. The pharmacokinetic profiles of antagonists were evaluated in vivo in rats. DMPX and KW-6002 had the greatest oral bioavailability. In contrast, SCH58261, MSX-2, and Ver-6623 had low or poor oral bioavailability. In summary, SCH58261, Biogen-34, MSX-2, and Ver-6623 had high affinities for both human and rat A2A receptors, with reasonable selectivity for A2A over A1 and A2B receptors. They are suitable as A2A -antagonists for in vitro pharmacological studies. Among the six A2A-antagonists, KW-6002 is the best for use in in vivo animal studies, particularly for a CNS target, based on its bioavailability, half life, and brain penetration.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Adenosina/análogos & derivados , Adenosina/química , Adenosina/farmacocinética , Adenosina/farmacología , Antagonistas del Receptor de Adenosina A3 , Animales , Unión Competitiva/efectos de los fármacos , Disponibilidad Biológica , Línea Celular , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Tasa de Depuración Metabólica , Estructura Molecular , Células PC12 , Fenetilaminas/química , Fenetilaminas/farmacocinética , Fenetilaminas/farmacología , Purinas/química , Purinas/farmacocinética , Purinas/farmacología , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A2A/genética , Receptor de Adenosina A3/genética , Receptor de Adenosina A3/metabolismo , Teobromina/análogos & derivados , Teobromina/química , Teobromina/farmacocinética , Teobromina/farmacología , Triazinas/química , Triazinas/farmacocinética , Triazinas/farmacología , Triazoles/química , Triazoles/farmacocinética , Triazoles/farmacología , Xantinas/química , Xantinas/farmacocinética , Xantinas/farmacología
19.
J Med Chem ; 49(12): 3682-92, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16759111

RESUMEN

Adenosine has been suggested to induce bronchial hyperresponsiveness in asthmatics, which is believed to be an A(2B) adenosine receptor (AdoR) mediated pathway. We hypothesize that a selective, high-affinity A(2B) AdoR antagonist may provide therapeutic benefit in the treatment of asthma. In an attempt to identify a high-affinity, selective antagonist for the A(2B) AdoR, we synthesized 8-(C-4-pyrazolyl) xanthines. Compound 22, 8-(1H-pyrazol-4-yl)-1,3-dipropyl xanthine, is a N-1 unsubstituted pyrazole derivative that has favorable binding affinity (K(i) = 9 nM) for the A(2B) AdoR, but it is only 2-fold selective versus the A(1) AdoR. Introduction of a benzyl group at the N-1-pyrazole position of 22 resulted in 19, which had moderate selectivity. The initial focus of the SAR study was on the preparation of substituted benzyl derivatives of 19 because the corresponding phenyl, phenethyl, and phenpropyl derivatives showed a decrease in A(2B) AdoR affinity and selectivity relative to 19. The preferred substitution on the phenyl ring of 19 contains an electron-withdrawing group, specifically F or CF(3) at the m-position, as in 33 and 36 respectively, increases the selectivity while retaining the affinity for the A(2B) AdoR. Exploring disubstitutions on the phenyl ring of derivatives 33 and36 led to the 2-chloro-5-trifluoromethylphenyl derivative 50, which retained the A(2B) AdoR affinity but enhanced the selectivity relative to 36. After optimization of the substitution on the 8-pyrazole xanthine, 1,3-disubstitution of the xanthine core was explored with methyl, ethyl, butyl, and isobutyl groups. In comparison to the corresponding dipropyl analogues, the smaller 1,3-dialkyl groups (methyl and ethyl) increased the A(2B) AdoR binding selectivity of the xanthine derivatives while retaining the affinity. However, the larger 1,3-dialkyl groups (isobutyl and butyl) resulted in a decrease in both A(2B) AdoR affinity and selectivity. This final SAR optimization led to the discovery of 1,3-dimethyl derivative 60, 8-(1-(3-(trifluoromethyl) benzyl)-1H-pyrazol-4-yl)-1,3-dimethyl xanthine, a high-affinity (K(i) = 1 nM) A(2B) AdoR antagonist with high selectivity (990-, 690-, and 1,000-) for the human A(1), A(2A,) and A(3) AdoRs.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Pirazoles/síntesis química , Teofilina/análogos & derivados , Xantinas/síntesis química , Animales , Línea Celular , Cricetinae , Cricetulus , Humanos , Pirazoles/química , Pirazoles/farmacología , Ensayo de Unión Radioligante , Teofilina/síntesis química , Teofilina/química , Teofilina/farmacología , Xantinas/química , Xantinas/farmacología
20.
Br J Pharmacol ; 173(21): 3088-3098, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27449698

RESUMEN

BACKGROUND AND PURPOSE: Enhanced late Na+ current (late INa ) in the myocardium is pro-arrhythmic. Inhibition of this current is a promising strategy to stabilize ventricular repolarization and suppress arrhythmias. Here, we describe GS-6615, a selective inhibitor of late INa , already in clinical development for the treatment of long QT syndrome 3 (LQT3). EXPERIMENTAL APPROACH: The effects of GS-6615 to inhibit late INa , versus other ion currents to shorten the ventricular action potential duration (APD), monophasic APD (MAPD) and QT interval, and decrease to the incidence of ventricular arrhythmias was determined in rabbit cardiac preparations. To mimic the electrical phenotype of LQT3, late INa was increased using the sea anemone toxin (ATX-II). KEY RESULTS: GS-6615 inhibited ATX-II enhanced late INa in ventricular myocytes (IC50  = 0.7 µM), shortened the ATX-II induced prolongation of APD, MAPD, QT interval, and decreased spatiotemporal dispersion of repolarization and ventricular arrhythmias. Inhibition by GS-6615 of ATX-II enhanced late INa was strongly correlated with shortening of myocyte APD and isolated heart MAPD (R2  = 0.94 and 0.98 respectively). In contrast to flecainide, GS-6615 had the minimal effects on peak INa . GS-6615 did not decrease the maximal upstroke velocity of the action potential (Vmax) nor widen QRS intervals. CONCLUSIONS AND IMPLICATIONS: GS-6615 was a selective inhibitor of late INa , stabilizes the ventricular repolarization and suppresses arrhythmias in a model of LQT3. The concentrations at which the electrophysiological effects of GS-6615 were observed are comparable to plasma levels associated with QTc shortening in patients with LQT3, indicating that these effects are clinically relevant.


Asunto(s)
Antiarrítmicos/farmacología , Corazón/efectos de los fármacos , Oxazepinas/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Animales , Antiarrítmicos/química , Síndrome de QT Prolongado/tratamiento farmacológico , Estructura Molecular , Oxazepinas/química , Conejos , Bloqueadores de los Canales de Sodio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA