Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Eur J Med Chem ; 239: 114522, 2022 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-35749987

RESUMEN

Lysine-specific demethylase 1 (LSD1) is an enzyme that demethylates methylated histone H3 lysine 4 (H3K4). Inhibition of LSD1 enzyme activity could increase H3K4 methylation levels and treat diseases associated with epigenetic dysregulation. However, known LSD1 inhibitors disrupt the interaction between LSD1 and cofactors such as GFI1B, causing the risk of hematological toxicity, including thrombocytopenia. Starting from a known LSD1 inhibitor (±)1 as a lead compound, a novel series of LSD1 inhibitors that do not induce the expression of GFI1 mRNA, an in vitro surrogate marker of LSD1-GFI1B dissociation, has been designed and synthesized. Initial structure-activity relationship (SAR) studies revealed the structural features key to avoiding GFI1 mRNA induction. Such SAR information enables optimization of LSD1 inhibitors with lowered risk of hematological side effects; TAK-418 ((1R,2R)-2n), the clinical candidate compound found through this optimization, has a hematological safety profile in rodents and humans. We further confirmed that oral administration of TAK-418 at 0.3 and 1 mg/kg for 2 weeks ameliorated memory deficits in mice with NMDA receptor hypofunction, suggesting potential of efficacy in neurodevelopmental disorders. TAK-418 warrants further investigation as a novel class of LSD1 inhibitors with a superior safety profile for the treatment of CNS disorders.


Asunto(s)
Histona Demetilasas , Lisina , Animales , Inhibidores Enzimáticos/química , Lisina/metabolismo , Ratones , ARN Mensajero , Relación Estructura-Actividad
2.
J Med Chem ; 65(12): 8456-8477, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35686954

RESUMEN

To discover a novel series of potent inhibitors of enteropeptidase, a membrane-bound serine protease localized to the duodenal brush border, 4-guanidinobenzoate derivatives were evaluated with minimal systemic exposure. The 1c docking model enabled the installation of an additional carboxylic acid moiety to obtain an extra interaction with enteropeptidase, yielding 2a. The oral administration of 2a significantly elevated the fecal protein output, a pharmacodynamic marker, in diet-induced obese (DIO) mice, whereas subcutaneous administration did not change this parameter. Thus, systemic exposure of 2a was not required for its pharmacological effects. Further optimization focusing on the in vitro IC50 value and T1/2, an indicator of dissociation time, followed by enhanced in vivo pharmacological activity based on the ester stability of the compounds, revealed two series of potent enteropeptidase inhibitors, a dihydrobenzofuran analogue ((S)-5b, SCO-792) and phenylisoxazoline (6b), which exhibited potent anti-obesity effects despite their low systemic exposure following their oral administration to DIO rats.


Asunto(s)
Enteropeptidasa , Obesidad , Animales , Benzoatos , Enteropeptidasa/metabolismo , Guanidinas/farmacología , Guanidinas/uso terapéutico , Ratones , Ratones Obesos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratas
3.
J Med Chem ; 65(5): 4270-4290, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35188773

RESUMEN

Inhibition of glucosylceramide synthase (GCS) is a major therapeutic strategy for Gaucher's disease and has been suggested as a potential target for treating Parkinson's disease. Herein, we report the discovery of novel brain-penetrant GCS inhibitors. Assessment of the structure-activity relationship revealed a unique pharmacophore in this series. The lipophilic ortho-substituent of aromatic ring A and the appropriate directionality of aromatic ring B were key for potency. Optimization of the absorption, distribution, metabolism, elimination, toxicity (ADMETox) profile resulted in the discovery of T-036, a potent GCS inhibitor in vivo. Pharmacophore-based scaffold hopping was performed to mitigate safety concerns associated with T-036. The ring opening of T-036 resulted in another potent GCS inhibitor with a lower toxicological risk, T-690, which reduced glucosylceramide in a dose-dependent manner in the plasma and cortex of mice. Finally, we discuss the structural aspects of the compounds that impart a unique inhibition mode and lower the cardiovascular risk.


Asunto(s)
Enfermedad de Gaucher , Glucosiltransferasas , Animales , Encéfalo/metabolismo , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/metabolismo , Glucosilceramidas/metabolismo , Glucosilceramidas/uso terapéutico , Glucosiltransferasas/metabolismo , Glucosiltransferasas/uso terapéutico , Ratones
4.
J Neurochem ; 159(3): 543-553, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34398463

RESUMEN

Gaucher disease (GD), the most common lysosomal storage disorders, is caused by GBA gene mutations resulting in glycosphingolipids accumulations in various tissues, such as the brain. While suppressing glycosphingolipid accumulation is the central strategy for treating peripheral symptoms of GD, there is no effective treatment for the central nervous system symptoms. As glycosphingolipid biosynthesis starts from ceramide glycosylation by glucosylceramide synthase (GCS), inhibiting GCS in the brain is a promising strategy for neurological GD. Herein, we discovered T-036, a potent and brain-penetrant GCS inhibitor with a unique chemical structure and binding property. T-036 does not harbor an aliphatic amine moiety and has a noncompetitive inhibition mode to the substrates, unlike other known inhibitors. T-036 exhibited sufficient exposure and a significant reduction of glucosylsphingolipids in the plasma and brain of the GD mouse model. Therefore, T-036 could be a promising lead molecule for treating central nervous system symptoms of GD.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Gaucher/tratamiento farmacológico , Glucosiltransferasas/antagonistas & inhibidores , Animales , Corteza Cerebral/metabolismo , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Glucosilceramidasa , Glicoesfingolípidos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad por Sustrato
5.
Sci Adv ; 7(11)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33712455

RESUMEN

Persistent epigenetic dysregulation may underlie the pathophysiology of neurodevelopmental disorders, such as autism spectrum disorder (ASD). Here, we show that the inhibition of lysine-specific demethylase 1 (LSD1) enzyme activity normalizes aberrant epigenetic control of gene expression in neurodevelopmental disorders. Maternal exposure to valproate or poly I:C caused sustained dysregulation of gene expression in the brain and ASD-like social and cognitive deficits after birth in rodents. Unexpectedly, a specific inhibitor of LSD1 enzyme activity, 5-((1R,2R)-2-((cyclopropylmethyl)amino)cyclopropyl)-N-(tetrahydro-2H-pyran-4-yl)thiophene-3-carboxamide hydrochloride (TAK-418), almost completely normalized the dysregulated gene expression in the brain and ameliorated some ASD-like behaviors in these models. The genes modulated by TAK-418 were almost completely different across the models and their ages. These results suggest that LSD1 enzyme activity may stabilize the aberrant epigenetic machinery in neurodevelopmental disorders, and the inhibition of LSD1 enzyme activity may be the master key to recover gene expression homeostasis. TAK-418 may benefit patients with neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Histona Demetilasas/metabolismo , Humanos
6.
Structure ; 29(3): 203-212.e4, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33450182

RESUMEN

The hERG channel is a voltage-gated potassium channel involved in cardiac repolarization. Off-target hERG inhibition by drugs has become a critical issue in the pharmaceutical industry. The three-dimensional structure of the hERG channel was recently reported at 3.8-Å resolution using cryogenic electron microscopy (cryo-EM). However, the drug inhibition mechanism remains unclear because of the scarce structural information regarding the drug- and potassium-bound hERG channels. In this study, we obtained the cryo-EM density map of potassium-bound hERG channel complexed with astemizole, a well-known hERG inhibitor that increases risk of potentially fatal arrhythmia, at 3.5-Å resolution. The structure suggested that astemizole inhibits potassium conduction by binding directly below the selectivity filter. Furthermore, we propose a possible binding model of astemizole to the hERG channel and provide insights into the unusual sensitivity of hERG to several drugs.


Asunto(s)
Astemizol/química , Canal de Potasio ERG1/química , Bloqueadores de los Canales de Potasio/química , Astemizol/farmacología , Sitios de Unión , Microscopía por Crioelectrón , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Bloqueadores de los Canales de Potasio/farmacología , Unión Proteica
7.
Neuropsychopharmacology ; 44(8): 1505-1512, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30580376

RESUMEN

Dysregulation of histone H3 lysine 4 (H3K4) methylation has been implicated in the pathogenesis of several neurodevelopmental disorders. Targeting lysine-specific demethylase 1 (LSD1), an H3K4 demethylase, is therefore a promising approach to treat these disorders. However, LSD1 forms complexes with cofactors including growth factor independent 1B (GFI1B), a critical regulator of hematopoietic differentiation. Known tranylcypromine-based irreversible LSD1 inhibitors bind to coenzyme flavin adenine dinucleotide (FAD) and disrupt the LSD1-GFI1B complex, which is associated with hematotoxicity such as thrombocytopenia, representing a major hurdle in the development of LSD1 inhibitors as therapeutic agents. To discover LSD1 inhibitors with potent epigenetic modulation and lower risk of hematotoxicity, we screened small molecules that enhance H3K4 methylation by the inhibition of LSD1 enzyme activity in primary cultured rat neurons but have little impact on LSD1-GFI1B complex in human TF-1a erythroblasts. Here we report the discovery of a specific inhibitor of LSD1 enzyme activity, T-448 (3-((1S,2R)-2-(cyclobutylamino)cyclopropyl)-N-(5-methyl-1,3,4-thiadiazol-2-yl)benzamide fumarate). T-448 has minimal impact on the LSD1-GFI1B complex and a superior hematological safety profile in mice via the generation of a compact formyl-FAD adduct. T-448 increased brain H3K4 methylation and partially restored learning function in mice with NMDA receptor hypofunction. T-448-type LSD1 inhibitors with improved safety profiles may provide unique therapeutic approaches for central nervous system disorders associated with epigenetic dysregulation.


Asunto(s)
Benzamidas/farmacología , Histona Demetilasas/antagonistas & inhibidores , Aprendizaje por Laberinto/efectos de los fármacos , Trombocitopenia/inducido químicamente , Animales , Benzamidas/efectos adversos , Encéfalo/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Masculino , Metilación/efectos de los fármacos , Ratones , Neuronas/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Proteínas Represoras/metabolismo
8.
Chem Pharm Bull (Tokyo) ; 65(11): 1058-1077, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29093293

RESUMEN

It has been hypothesized that selective inhibition of phosphodiesterase (PDE) 2A could potentially be a novel approach to treat cognitive impairment in neuropsychiatric and neurodegenerative disorders through augmentation of cyclic nucleotide signaling pathways in brain regions associated with learning and memory. Following our earlier work, this article describes a drug design strategy for a new series of lead compounds structurally distinct from our clinical candidate 2 (TAK-915), and subsequent medicinal chemistry efforts to optimize potency, selectivity over other PDE families, and other preclinical properties including in vitro phototoxicity and in vivo rat plasma clearance. These efforts resulted in the discovery of N-((1S)-2-hydroxy-2-methyl-1-(4-(trifluoromethoxy)phenyl)propyl)-6-methyl-5-(3-methyl-1H-1,2,4-triazol-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (20), which robustly increased 3',5'-cyclic guanosine monophosphate (cGMP) levels in the rat brain following an oral dose, and moreover, attenuated MK-801-induced episodic memory deficits in a passive avoidance task in rats. These data provide further support to the potential therapeutic utility of PDE2A inhibitors in enhancing cognitive performance.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores de Fosfodiesterasa/farmacología , Pirazinas/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Células 3T3 , Administración Oral , Animales , Células COS , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Trastornos del Conocimiento/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Estructura Molecular , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/química , Difracción de Polvo , Pirazinas/química , Pirazoles/química , Piridinas/química , Pirimidinas/química , Ratas , Ratas Long-Evans , Solubilidad , Relación Estructura-Actividad , Termodinámica
9.
J Med Chem ; 60(18): 7658-7676, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28759228

RESUMEN

Herein, we describe the discovery of a potent, selective, brain-penetrating, in vivo active phosphodiesterase (PDE) 2A inhibitor lead series. To identify high-quality leads suitable for optimization and enable validation of the physiological function of PDE2A in vivo, structural modifications of the high-throughput screening hit 18 were performed. Our lead generation efforts revealed three key potency-enhancing functionalities with minimal increases in molecular weight (MW) and no change in topological polar surface area (TPSA). Combining these structural elements led to the identification of 6-methyl-N-((1R)-1-(4-(trifluoromethoxy)phenyl)propyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (38a), a molecule with the desired balance of preclinical properties. Further characterization by cocrystal structure analysis of 38a bound to PDE2A uncovered a unique binding mode and provided insights into its observed potency and PDE selectivity. Compound 38a significantly elevated 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brain following oral administration, thus validating this compound as a useful pharmacological tool and an attractive lead for future optimization.


Asunto(s)
Encéfalo/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/farmacocinética , Pirimidinas/farmacología , Pirimidinas/farmacocinética , Administración Oral , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Descubrimiento de Drogas , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/química , Pirimidinas/administración & dosificación , Pirimidinas/química , Ratas
10.
J Med Chem ; 60(18): 7677-7702, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28796496

RESUMEN

Phosphodiesterase (PDE) 2A inhibitors have emerged as a novel mechanism with potential therapeutic option to ameliorate cognitive dysfunction in schizophrenia or Alzheimer's disease through upregulation of cyclic nucleotides in the brain and thereby achieve potentiation of cyclic nucleotide signaling pathways. This article details the expedited optimization of our recently disclosed pyrazolo[1,5-a]pyrimidine lead compound 4b, leading to the discovery of clinical candidate 36 (TAK-915), which demonstrates an appropriate combination of potency, PDE selectivity, and favorable pharmacokinetic (PK) properties, including brain penetration. Successful identification of 36 was realized through application of structure-based drug design (SBDD) to further improve potency and PDE selectivity, coupled with prospective design focused on physicochemical properties to deliver brain penetration. Oral administration of 36 demonstrated significant elevation of 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brains and improved cognitive performance in a novel object recognition task in rats. Consequently, compound 36 was advanced into human clinical trials.


Asunto(s)
Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/farmacocinética , Pirazinas/farmacología , Pirazinas/farmacocinética , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Diseño de Fármacos , Halogenación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Inhibidores de Fosfodiesterasa/química , Pirazinas/química , Pirazoles/química , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley
11.
Peptides ; 94: 56-63, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28676225

RESUMEN

Cytidine triphosphate synthase 1 (CTPS1) is an enzyme expressed in activated lymphocytes that catalyzes the conversion of uridine triphosphate (UTP) to cytidine triphosphate (CTP) with ATP-dependent amination, using either L-glutamine or ammonia as the nitrogen source. Since CTP plays an important role in DNA/RNA synthesis, phospholipid synthesis, and protein sialyation, CTPS1-inhibition is expected to control lymphocyte proliferation and size expansion in inflammatory diseases. In contrast, CTPS2, an isozyme of CTPS1 possessing 74% amino acid sequence homology, is expressed in normal lymphocytes. Thus, CTPS1-selective inhibition is important to avoid undesirable side effects. Here, we report the discovery of CTpep-3: Ac-FRLGLLKAFRRLF-OH from random peptide libraries displayed on T7 phage, which exhibited CTPS1-selective binding with a KD value of 210nM in SPR analysis and CTPS1-selective inhibition with an IC50 value of 110nM in the enzyme assay. Furthermore, two fundamentally different approaches, enzyme inhibition assay and HDX-MS, provided the same conclusion that CTpep-3 acts by binding to the amidoligase (ALase) domain on CTPS1. To our knowledge, CTpep-3 is the first CTPS1-selective inhibitor.


Asunto(s)
Bacteriófago T7/metabolismo , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Linfocitos/enzimología , Péptidos/farmacología , Humanos , Linfocitos/efectos de los fármacos , Biblioteca de Péptidos
12.
J Med Chem ; 60(10): 4358-4368, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28471657

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcriptional factor that expresses in lymphocytes and regulates the differentiation and proliferation of lymphocytes. Therefore, BCL6 is a therapeutic target for autoimmune diseases and cancer treatment. This report presents the discovery of BCL6-corepressor interaction inhibitors by using a biophysics-driven fragment-based approach. Using the surface plasmon resonance (SPR)-based fragment screening, we successfully identified fragment 1 (SPR KD = 1200 µM, ligand efficiency (LE) = 0.28), a competitive binder to the natural ligand BCoR peptide. Moreover, we elaborated 1 into the more potent compound 7 (SPR KD = 0.078 µM, LE = 0.37, cell-free protein-protein interaction (PPI) IC50 = 0.48 µM (ELISA), cellular PPI IC50 = 8.6 µM (M2H)) by a structure-based design and structural integration with a second high-throughput screening hit.


Asunto(s)
Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Descubrimiento de Drogas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Proto-Oncogénicas c-bcl-6/química , Resonancia por Plasmón de Superficie
13.
ACS Chem Biol ; 12(7): 1760-1768, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28440616

RESUMEN

Eukaryotic initiation factor 4A-3 (eIF4A3) is an Asp-Glu-Ala-Asp (DEAD) box-family adenosine triphosphate (ATP)-dependent RNA helicase. Subtypes eIF4A1 and eIF4A2 are required for translation initiation, but eIF4A3 participates in the exon junction complex (EJC) and functions in RNA metabolism including nonsense-mediated RNA decay (NMD). No small molecules for NMD inhibition via selective inhibition of eIF4A3 have been discovered. Here, we identified allosteric eIF4A3 inhibitors from a high-throughput screening campaign. Chemical optimization of the lead compounds based on ATPase activity yielded compound 2, which exhibited noncompetitive inhibition with ATP or RNA and high selectivity for eIF4A3 over other helicases. The optimized compounds suppressed the helicase activity of eIF4A3 in an ATPase-dependent manner. Hydrogen/deuterium exchange mass spectrometry demonstrated that the deuterium-incorporation pattern of compound 2 overlapped with that of an allosteric pan-eIF4A inhibitor, hippuristanol, suggesting that compound 2 binds to an allosteric region on eIF4A3. We examined NMD activity using a luciferase-based cellular reporter system and a quantitative real-time polymerase chain-reaction-based cellular system to monitor levels of endogenous NMD substrates. NMD suppression by the compounds correlated positively with their ATPase-inhibitory activity. In conclusion, we developed a novel eIF4A3 inhibitor that targets the EJC. The optimized chemical probes represent useful tools for understanding the functions of eIF4A3 in RNA homeostasis.


Asunto(s)
ADN Helicasas/química , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Degradación de ARNm Mediada por Codón sin Sentido/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Regulación Alostérica , Secuencia de Aminoácidos , Unión Competitiva , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Concentración 50 Inhibidora , Alineación de Secuencia , Bibliotecas de Moléculas Pequeñas/farmacología , Esteroles/química , Esteroles/farmacología
14.
J Med Chem ; 60(2): 608-626, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27966948

RESUMEN

On the basis of a superposition study of X-ray crystal structures of complexes of quinazoline derivative 1 and triazole derivative 2 with matrix metalloproteinase (MMP)-13 catalytic domain, a novel series of fused pyrimidine compounds which possess a 1,2,4-triazol-3-yl group as a zinc binding group (ZBG) was designed. Among the herein described and evaluated compounds, 31f exhibited excellent potency for MMP-13 (IC50 = 0.036 nM) and selectivities (greater than 1,500-fold) over other MMPs (MMP-1, -2, -3, -7, -8, -9, -10, and -14) and tumor necrosis factor-α converting enzyme (TACE). Furthermore, the inhibitor was shown to protect bovine nasal cartilage explants against degradation induced by interleukin-1 and oncostatin M. In this article, we report the discovery of extremely potent, highly selective, and orally bioavailable fused pyrimidine derivatives that possess a 1,2,4-triazol-3-yl group as a novel ZBG for selective MMP-13 inhibition.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Pirimidinas/farmacología , Pirimidinonas/farmacología , Tiofenos/farmacología , Triazoles/farmacología , Zinc/química , Animales , Cartílago/metabolismo , Bovinos , Quelantes/síntesis química , Quelantes/farmacología , Colágeno/metabolismo , Diseño de Fármacos , Inhibidores de la Metaloproteinasa de la Matriz/síntesis química , Pirimidinas/síntesis química , Pirimidinonas/síntesis química , Quinazolinas/síntesis química , Quinazolinas/farmacología , Tiofenos/síntesis química , Triazoles/síntesis química
15.
Bioorg Med Chem ; 24(23): 6149-6165, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27825552

RESUMEN

Matrix metalloproteinase-13 (MMP-13), a member of the collagenase family of enzymes, has been implicated to play a key role in the pathology of osteoarthritis. Recently, we have reported the discovery of a series of quinazoline-2-carboxamide based non-zinc-binding MMP-13 selective inhibitors, as exemplified by compound 1. We then continued our research of a novel class of zinc-binding inhibitors to obtain follow-up compounds with different physicochemical, pharmacokinetic, and biological activity profiles. In order to design selective MMP-13 inhibitors, we adopted a strategy of connecting a zinc-binding group with the quinazoline-2-carboxamide system, a unique S1' binder, by an appropriate linker. Among synthesized compounds, a triazolone inhibitor 35 exhibited excellent potency (IC50=0.071nM) and selectivity (greater than 170-fold) over other MMPs (MMP-1, 2, 3, 7, 8, 9, 10, 12, and 14) and tumor necrosis factor-α converting enzyme (TACE). In this article, the design, synthesis, and biological activity of novel zinc-binding MMP-13 inhibitors are described.


Asunto(s)
Amidas/farmacología , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Pirimidinas/farmacología , Quinazolinonas/farmacología , Triazoles/farmacología , Zinc/química , Proteína ADAM17/antagonistas & inhibidores , Amidas/síntesis química , Amidas/farmacocinética , Animales , Diseño de Fármacos , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/síntesis química , Inhibidores de la Metaloproteinasa de la Matriz/farmacocinética , Microsomas Hepáticos/metabolismo , Pirimidinas/síntesis química , Pirimidinas/farmacocinética , Quinazolinonas/síntesis química , Quinazolinonas/farmacocinética , Ratas , Triazoles/síntesis química , Triazoles/farmacocinética
16.
Bioorg Med Chem ; 24(22): 6066-6074, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27720325

RESUMEN

A novel approach was conducted for fragment-based lead discovery and applied to renin inhibitors. The biochemical screening of a fragment library against renin provided the hit fragment which showed a characteristic interaction pattern with the target protein. The hit fragment bound only to the S1, S3, and S3SP (S3 subpocket) sites without any interactions with the catalytic aspartate residues (Asp32 and Asp215 (pepsin numbering)). Prior to making chemical modifications to the hit fragment, we first identified its essential binding sites by utilizing the hit fragment's substructures. Second, we created a new and smaller scaffold, which better occupied the identified essential S3 and S3SP sites, by utilizing library synthesis with high-throughput chemistry. We then revisited the S1 site and efficiently explored a good building block attaching to the scaffold with library synthesis. In the library syntheses, the binding modes of each pivotal compound were determined and confirmed by X-ray crystallography and the library was strategically designed by structure-based computational approach not only to obtain a more active compound but also to obtain informative Structure Activity Relationship (SAR). As a result, we obtained a lead compound offering synthetic accessibility as well as the improved in vitro ADMET profiles. The fragments and compounds possessing a characteristic interaction pattern provided new structural insights into renin's active site and the potential to create a new generation of renin inhibitors. In addition, we demonstrated our FBDD strategy integrating highly sensitive biochemical assay, X-ray crystallography, and high-throughput synthesis and in silico library design aimed at fragment morphing at the initial stage was effective to elucidate a pocket profile and a promising lead compound.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de Proteasas/farmacología , Renina/antagonistas & inhibidores , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetulus , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Renina/metabolismo , Relación Estructura-Actividad
17.
Bioorg Med Chem ; 24(16): 3447-55, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27301679

RESUMEN

Utilizing structure-based drug design techniques, we designed and synthesized phosphodiesterase 10A (PDE10A) inhibitors based on pyridazin-4(1H)-one. These compounds can interact with Tyr683 in the PDE10A selectivity pocket. Pyridazin-4(1H)-one derivative 1 was linked with a benzimidazole group through an alkyl spacer to interact with the OH of Tyr683 and fill the PDE10A selectivity pocket. After optimizing the linker length, we identified 1-(cyclopropylmethyl)-5-[3-(1-methyl-1H-benzimidazol-2-yl)propoxy]-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one (16f) as having highly potent PDE10A inhibitory activity (IC50=0.76nM) and perfect selectivity against other PDEs (>13,000-fold, IC50=>10,000nM). The crystal structure of 16f bound to PDE10A revealed that the benzimidazole moiety was located deep within the PDE10A selectivity pocket and interacted with Tyr683. Additionally, a bidentate interaction existed between the 5-alkoxypyridazin-4(1H)-one moiety and the conserved Gln716 present in all PDEs.


Asunto(s)
Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Cristalografía por Rayos X , Estructura Molecular , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad
18.
Bioorg Med Chem ; 23(22): 7138-49, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26494583

RESUMEN

Highly potent and brain-penetrant phosphodiesterase 10A (PDE10A) inhibitors based on the 2-oxindole scaffold were designed and synthesized. (2-Oxo-1,3-oxazolidin-3-yl)phenyl derivative 1 showed the high P-glycoprotein (P-gp) efflux (efflux ratio (ER)=6.2) despite the potent PDE10A inhibitory activity (IC50=0.94 nM). We performed an optimization study to improve both the P-gp efflux ratio and PDE10A inhibitory activity by utilizing structure-based drug design (SBDD) techniques based on the X-ray crystal structure with PDE10A. Finally, 1-(cyclopropylmethyl)-4-fluoro-5-[5-methoxy-4-oxo-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-1(4H)-yl]-3,3-dimethyl-1,3-dihydro-2H-indol-2-one (19e) was identified with improved P-gp efflux (ER=1.4) and an excellent PDE10A inhibitory activity (IC50=0.080 nM). Compound 19e also exhibited satisfactory brain penetration, and suppressed PCP-induced hyperlocomotion with a minimum effective dose of 0.3mg/kg by oral administration in mice.


Asunto(s)
Encéfalo/metabolismo , Diseño de Fármacos , Indoles/química , Indoles/síntesis química , Indoles/farmacología , Hidrolasas Diéster Fosfóricas/química , Piridazinas/síntesis química , Piridazinas/farmacología , Administración Oral , Animales , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Semivida , Indoles/farmacocinética , Ratones , Conformación Molecular , Simulación del Acoplamiento Molecular , Actividad Motora/efectos de los fármacos , Oxindoles , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacocinética , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Piridazinas/química , Piridazinas/farmacocinética
19.
J Med Chem ; 57(22): 9627-43, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25384088

RESUMEN

A novel series of pyridazinone-based phosphodiesterase 10A (PDE10A) inhibitors were synthesized. Our optimization efforts using structure-based drug design (SBDD) techniques on the basis of the X-ray crystal structure of PDE10A in complex with hit compound 1 (IC50 = 23 nM; 110-fold selectivity over other PDEs) led to the identification of 1-[2-fluoro-4-(1H-pyrazol-1-yl)phenyl]-5-methoxy-3-(1-phenyl-1H-pyrazol-5-yl)pyridazin-4(1H)-one (27h). Compound 27h has potent inhibitory activity (IC50 = 0.30 nM), excellent selectivity (>15000-fold selectivity over other PDEs), and favorable pharmacokinetics, including high brain penetration, in mice. Oral administration of compound 27h to mice elevated striatal 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) levels at 0.3 mg/kg and showed potent suppression of phencyclidine (PCP)-induced hyperlocomotion at a minimum effective dose (MED) of 0.3 mg/kg. Compound 27h (TAK-063) is currently being evaluated in clinical trials for the treatment of schizophrenia.


Asunto(s)
Inhibidores de Fosfodiesterasa/química , Hidrolasas Diéster Fosfóricas/química , Pirazoles/química , Piridazinas/química , Administración Oral , Animales , Encéfalo/efectos de los fármacos , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Microsomas Hepáticos/efectos de los fármacos , Movimiento/efectos de los fármacos , Fenciclidina/química , Conformación Proteica
20.
Bioorg Med Chem ; 22(19): 5487-505, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25192810

RESUMEN

On the basis of X-ray co-crystal structures of matrix metalloproteinase-13 (MMP-13) in complex with its inhibitors, our structure-based drug design (SBDD) strategy was directed to achieving high affinity through optimal protein-ligand interaction with the unique S1″ hydrophobic specificity pocket. This report details the optimization of lead compound 44 to highly potent and selective MMP-13 inhibitors based on fused pyrimidine scaffolds represented by the thienopyrimidin-4-one 26c. Furthermore, we have examined the release of collagen fragments from bovine nasal cartilage in response to a combination of IL-1 and oncostatin M.


Asunto(s)
Derivados del Benceno/química , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Pirimidinas/farmacología , Administración Oral , Derivados del Benceno/administración & dosificación , Derivados del Benceno/farmacología , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/química , Modelos Moleculares , Estructura Molecular , Pirimidinas/administración & dosificación , Pirimidinas/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA