Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Bioorg Med Chem Lett ; 27(3): 590-596, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011221

RESUMEN

The design and synthesis of a series of tripeptide acylsulfonamides as potent inhibitors of the HCV NS3/4A serine protease is described. These analogues house a C4 aryl, C4 hydroxy-proline at the S2 position of the tripeptide scaffold. Information relating to structure-activity relationships as well as the pharmacokinetic and cardiovascular profiles of these analogues is provided.


Asunto(s)
Antivirales/química , Hepacivirus/enzimología , Oligopéptidos/química , Inhibidores de Proteasas/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Semivida , Corazón/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Humanos , Técnicas In Vitro , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Prolina/química , Inhibidores de Proteasas/farmacocinética , Inhibidores de Proteasas/farmacología , Conejos , Ratas , Relación Estructura-Actividad , Sulfonamidas/química , Proteínas no Estructurales Virales/metabolismo
2.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 134-43, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24419386

RESUMEN

The process of iterative structure-based drug design involves the X-ray crystal structure determination of upwards of 100 ligands with the same general scaffold (i.e. chemotype) complexed with very similar, if not identical, protein targets. In conjunction with insights from computational models and assays, this collection of crystal structures is analyzed to improve potency, to achieve better selectivity and to reduce liabilities such as absorption, distribution, metabolism, excretion and toxicology. Current methods for modeling ligands into electron-density maps typically do not utilize information on how similar ligands bound in related structures. Even if the electron density is of sufficient quality and resolution to allow de novo placement, the process can take considerable time as the size, complexity and torsional degrees of freedom of the ligands increase. A new module, Guided Ligand Replacement (GLR), was developed in Phenix to increase the ease and success rate of ligand placement when prior protein-ligand complexes are available. At the heart of GLR is an algorithm based on graph theory that associates atoms in the target ligand with analogous atoms in the reference ligand. Based on this correspondence, a set of coordinates is generated for the target ligand. GLR is especially useful in two situations: (i) modeling a series of large, flexible, complicated or macrocyclic ligands in successive structures and (ii) modeling ligands as part of a refinement pipeline that can automatically select a reference structure. Even in those cases for which no reference structure is available, if there are multiple copies of the bound ligand per asymmetric unit GLR offers an efficient way to complete the model after the first ligand has been placed. In all of these applications, GLR leverages prior knowledge from earlier structures to facilitate ligand placement in the current structure.


Asunto(s)
Cristalografía por Rayos X/métodos , Diseño de Fármacos , Proteasa del VIH/química , Proteasa del VIH/metabolismo , VIH-1/enzimología , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 144-54, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24419387

RESUMEN

High-throughput drug-discovery and mechanistic studies often require the determination of multiple related crystal structures that only differ in the bound ligands, point mutations in the protein sequence and minor conformational changes. If performed manually, solution and refinement requires extensive repetition of the same tasks for each structure. To accelerate this process and minimize manual effort, a pipeline encompassing all stages of ligand building and refinement, starting from integrated and scaled diffraction intensities, has been implemented in Phenix. The resulting system is able to successfully solve and refine large collections of structures in parallel without extensive user intervention prior to the final stages of model completion and validation.


Asunto(s)
Cristalografía por Rayos X/métodos , Proteínas/química , Animales , Diseño de Fármacos , Factor Xa/química , Factor Xa/metabolismo , Proteasa del VIH/química , Proteasa del VIH/metabolismo , VIH-1/enzimología , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Proteínas/metabolismo , Trombina/química , Trombina/metabolismo
4.
Bioorg Med Chem Lett ; 23(14): 4107-11, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23747226

RESUMEN

The design, synthesis and characterization of a phosphonate inhibitor of N-acetylneuraminate-9-phosphate phosphatase (HDHD4) is described. Compound 3, where the substrate C-9 oxygen was replaced with a nonlabile CH2 group, inhibits HDHD4 with a binding affinity (IC50 11µM) in the range of the native substrate Neu5Ac-9-P (compound 1, Km 47µM). Combined SAR, modeling and NMR studies are consistent with the phosphonate group in inhibitor 3 forming a stable complex with native Mg(2+). In addition to this key interaction, the C-1 carboxylate of the sugar interacts with a cluster of basic residues, K141, R104 and R72. Comparative NMR studies of compounds 3 and 1 with Ca(2+) and Mg(2+) are indicative of a highly dynamic process in the active site for the HDHD4/Mg(2+)/3 complex. Possible explanations for this observation are discussed.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Ácidos Siálicos/síntesis química , Fosfatos de Azúcar/síntesis química , Animales , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Cinética , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Monoéster Fosfórico Hidrolasas/metabolismo , Estructura Terciaria de Proteína , Ratas , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Fosfatos de Azúcar/química , Fosfatos de Azúcar/metabolismo
5.
Biochem J ; 436(2): 331-9, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21410432

RESUMEN

CARM1 (co-activator-associated arginine methyltransferase 1) is a PRMT (protein arginine N-methyltransferase) family member that catalyses the transfer of methyl groups from SAM (S-adenosylmethionine) to the side chain of specific arginine residues of substrate proteins. This post-translational modification of proteins regulates a variety of transcriptional events and other cellular processes. Moreover, CARM1 is a potential oncological target due to its multiple roles in transcription activation by nuclear hormone receptors and other transcription factors such as p53. Here, we present crystal structures of the CARM1 catalytic domain in complex with cofactors [SAH (S-adenosyl-L-homocysteine) or SNF (sinefungin)] and indole or pyazole inhibitors. Analysis of the structures reveals that the inhibitors bind in the arginine-binding cavity and the surrounding pocket that exists at the interface between the N- and C-terminal domains. In addition, we show using ITC (isothermal titration calorimetry) that the inhibitors bind to the CARM1 catalytic domain only in the presence of the cofactor SAH. Furthermore, sequence differences for select residues that interact with the inhibitors may be responsible for the CARM1 selectivity against PRMT1 and PRMT3. Together, the structural and biophysical information should aid in the design of both potent and specific inhibitors of CARM1.


Asunto(s)
Indoles/antagonistas & inhibidores , Indoles/química , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Pirazoles/antagonistas & inhibidores , Pirazoles/química , Secuencia de Aminoácidos , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/metabolismo , Datos de Secuencia Molecular , Unión Proteica/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/metabolismo , Pirazoles/metabolismo
6.
Bioorg Med Chem Lett ; 21(22): 6646-51, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21996520

RESUMEN

Design, synthesis, and SAR of 7-oxopyrrolopyridine-derived DPP4 inhibitors are described. The preferred stereochemistry of these atropisomeric biaryl analogs has been identified as Sa. Compound (+)-3t, with a K(i) against DPP4, DPP8, and DPP9 of 0.37 nM, 2.2, and 5.7 µM, respectively, showed a significant improvement in insulin response after single doses of 3 and 10 µmol/kg in ob/ob mice.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Canales de Potasio Éter-A-Go-Go/metabolismo , Piridinas/química , Piridinas/farmacología , Animales , Dominio Catalítico , Diabetes Mellitus/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Humanos , Insulina/sangre , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Piridinas/farmacocinética , Pirroles/química , Pirroles/farmacocinética , Pirroles/farmacología , Ratas , Estereoisomerismo
7.
Bioorg Med Chem Lett ; 21(24): 7516-21, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22041058

RESUMEN

The design, synthesis and SAR of a novel class of valerolactam-based arylsulfonamides as potent and selective FXa inhibitors is reported. The arylsulfonamide-valerolactam scaffold was derived based on the proposed bioisosterism to the arylcyanoguanidine-caprolactam core in known FXa inhibitors. The SAR study led to compound 46 as the most potent FXa inhibitor in this series, with an IC(50) of 7 nM and EC(2×PT) of 1.7 µM. The X-ray structure of compound 40 bound to FXa shows that the sulfonamide-valerolactam scaffold anchors the aryl group in the S1 and the novel acylcytisine pharmacophore in the S4 pockets.


Asunto(s)
Anticoagulantes/química , Inhibidores del Factor Xa , Piperidonas/química , Inhibidores de Serina Proteinasa/química , Anticoagulantes/síntesis química , Anticoagulantes/farmacología , Sitios de Unión , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Factor Xa/metabolismo , Humanos , Lactamas/química , Conformación Molecular , Piperidonas/síntesis química , Piperidonas/farmacología , Estructura Terciaria de Proteína , Inhibidores de Serina Proteinasa/síntesis química , Inhibidores de Serina Proteinasa/farmacología , Relación Estructura-Actividad
8.
Bioorg Med Chem Lett ; 19(15): 4034-41, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19541481

RESUMEN

The N,N'-disubstituted cyanoguanidine is an excellent bioisostere of the thiourea and ketene aminal functional groups. We report the design and synthesis of a novel class of cyanoguanidine-based lactam derivatives as potent and orally active FXa inhibitors. The SAR studies led to the discovery of compound 4 (BMS-269223, K(i)=6.5nM, EC(2xPT)=32muM) as a selective, orally bioavailable FXa inhibitor with an excellent in vitro liability profile, favorable pharmacokinetics and pharmacodynamics in animal models. The X-ray crystal structure of 4 bound in FXa is presented and key ligand-protein interactions are discussed.


Asunto(s)
Antitrombina III/farmacología , Benzofuranos/farmacología , Guanidinas/química , Lactamas/química , Administración Oral , Animales , Antitrombina III/química , Benzofuranos/química , Química Farmacéutica/métodos , Cristalografía por Rayos X/métodos , Perros , Haplorrinos , Humanos , Concentración 50 Inhibidora , Cinética , Lactamas/farmacología , Ligandos , Modelos Químicos , Ratas , Relación Estructura-Actividad , Tiourea/química
9.
Bioorg Med Chem Lett ; 19(24): 6882-9, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19896847

RESUMEN

We report the design and synthesis of a novel class of N,N'-disubstituted aroylguanidine-based lactam derivatives as potent and orally active FXa inhibitors. The structure-activity relationships (SAR) investigation led to the discovery of the nicotinoyl guanidine 22 as a potent FXa inhibitor (FXa IC(50)=4 nM, EC(2xPT)=7 microM). However, the potent CYP3A4 inhibition activity (IC(50)=0.3 microM) of 22 precluded its further development. Detailed analysis of the X-ray crystal structure of compound 22 bound to FXa indicated that the substituent at the 6-position of the nicotinoyl group of 22 would be solvent-exposed, suggesting that efforts to attenuate the unwanted CYP activity could focus at this position without affecting FXa potency significantly. Further SAR studies on the 6-substituted nicotinoyl guanidines resulted in the discovery of 6-(dimethylcarbamoyl) nicotinoyl guanidine 36 (BMS-344577, IC(50)=9 nM, EC(2xPT)=2.5 microM), which was found to be a selective, orally efficacious FXa inhibitor with an excellent in vitro liability profile, favorable pharmacokinetics and pharmacodynamics in animal models.


Asunto(s)
Anticoagulantes/química , Inhibidores del Factor Xa , Guanidinas/química , Inhibidores de Serina Proteinasa/química , Anticoagulantes/farmacología , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Descubrimiento de Drogas , Guanidinas/farmacología , Humanos , Concentración 50 Inhibidora , Inhibidores de Serina Proteinasa/farmacología , Relación Estructura-Actividad
10.
Protein Sci ; 17(2): 240-50, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18227430

RESUMEN

The inhibition of DPP-IV by saxagliptin has been proposed to occur through formation of a covalent but reversible complex. To evaluate further the mechanism of inhibition, we determined the X-ray crystal structure of the DPP-IV:saxagliptin complex. This structure reveals covalent attachment between S630 and the inhibitor nitrile carbon (C-O distance <1.3 A). To investigate whether this serine addition is assisted by the catalytic His-Asp dyad, we generated two mutants of DPP-IV, S630A and H740Q, and assayed them for ability to bind inhibitor. DPP-IV H740Q bound saxagliptin with an approximately 1000-fold reduction in affinity relative to DPP-IV WT, while DPP-IV S630A showed no evidence for binding inhibitor. An analog of saxagliptin lacking the nitrile group showed unchanged binding properties to the both mutant proteins, highlighting the essential role S630 and H740 play in covalent bond formation between S630 and saxagliptin. Further supporting mechanism-based inhibition by saxagliptin, NMR spectra of enzyme-saxagliptin complexes revealed the presence of three downfield resonances with low fractionation factors characteristic of short and strong hydrogen bonds (SSHB). Comparison of the NMR spectra of various wild-type and mutant DPP-IV:ligand complexes enabled assignment of a resonance at approximately 14 ppm to H740. Two additional DPP-IV mutants, Y547F and Y547Q, generated to probe potential stabilization of the enzyme-inhibitor complex by this residue, did not show any differences in inhibitor binding either by ITC or NMR. Together with the previously published enzymatic data, the structural and binding data presented here strongly support a histidine-assisted covalent bond formation between S630 hydroxyl oxygen and the nitrile group of saxagliptin.


Asunto(s)
Adamantano/análogos & derivados , Dipéptidos/química , Dipeptidil Peptidasa 4/química , Adamantano/química , Adamantano/metabolismo , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Dipéptidos/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Enlace de Hidrógeno , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Resonancia Magnética Nuclear Biomolecular , Estructura Cuaternaria de Proteína
11.
Cancer Res ; 66(11): 5790-7, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740718

RESUMEN

Chronic myeloid leukemia (CML) is caused by the constitutively activated tyrosine kinase breakpoint cluster (BCR)-ABL. Current frontline therapy for CML is imatinib, an inhibitor of BCR-ABL. Although imatinib has a high rate of clinical success in early phase CML, treatment resistance is problematic, particularly in later stages of the disease, and is frequently mediated by mutations in BCR-ABL. Dasatinib (BMS-354825) is a multitargeted tyrosine kinase inhibitor that targets oncogenic pathways and is a more potent inhibitor than imatinib against wild-type BCR-ABL. It has also shown preclinical activity against all but one of the imatinib-resistant BCR-ABL mutants tested to date. Analysis of the crystal structure of dasatinib-bound ABL kinase suggests that the increased binding affinity of dasatinib over imatinib is at least partially due to its ability to recognize multiple states of BCR-ABL. The structure also provides an explanation for the activity of dasatinib against imatinib-resistant BCR-ABL mutants.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-abl/química , Pirimidinas/química , Tiazoles/química , Animales , Benzamidas , Cristalografía por Rayos X , Dasatinib , Resistencia a Antineoplásicos , Activación Enzimática , Humanos , Mesilato de Imatinib , Modelos Moleculares , Piperazinas/química , Piperazinas/metabolismo , Piperazinas/farmacología , Conformación Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Relación Estructura-Actividad , Tiazoles/metabolismo , Tiazoles/farmacología
12.
Protein Sci ; 27(1): 182-194, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28901593

RESUMEN

Often similar structures need to be compared to reveal local differences throughout the entire model or between related copies within the model. Therefore, a program to compare multiple structures and enable correction any differences not supported by the density map was written within the Phenix framework (Adams et al., Acta Cryst 2010; D66:213-221). This program, called Structure Comparison, can also be used for structures with multiple copies of the same protein chain in the asymmetric unit, that is, as a result of non-crystallographic symmetry (NCS). Structure Comparison was designed to interface with Coot(Emsley et al., Acta Cryst 2010; D66:486-501) and PyMOL(DeLano, PyMOL 0.99; 2002) to facilitate comparison of large numbers of related structures. Structure Comparison analyzes collections of protein structures using several metrics, such as the rotamer conformation of equivalent residues, displays the results in tabular form and allows superimposed protein chains and density maps to be quickly inspected and edited (via the tools in Coot) for consistency, completeness and correctness.


Asunto(s)
Modelos Moleculares , Proteínas/química , Interfaz Usuario-Computador , Proteínas/genética
13.
ACS Med Chem Lett ; 9(2): 143-148, 2018 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-29456803

RESUMEN

The design and synthesis of potent, tripeptidic acylsulfonamide inhibitors of HCV NS3 protease that contain a difluoromethyl cyclopropyl amino acid at P1 are described. A cocrystal structure of 18 with a NS3/4A protease complex suggests the presence of a H-bond between the polarized C-H of the CHF2 moiety and the backbone carbonyl of Leu135 of the enzyme. Structure-activity relationship studies indicate that this H-bond enhances enzyme inhibitory potency by 13- and 17-fold compared to the CH3 and CF3 analogues, respectively, providing insight into the deployment of this unique amino acid.

14.
J Med Chem ; 59(17): 8042-60, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27564532

RESUMEN

The discovery of a back-up to the hepatitis C virus NS3 protease inhibitor asunaprevir (2) is described. The objective of this work was the identification of a drug with antiviral properties and toxicology parameters similar to 2, but with a preclinical pharmacokinetic (PK) profile that was predictive of once-daily dosing. Critical to this discovery process was the employment of an ex vivo cardiovascular (CV) model which served to identify compounds that, like 2, were free of the CV liabilities that resulted in the discontinuation of BMS-605339 (1) from clinical trials. Structure-activity relationships (SARs) at each of the structural subsites in 2 were explored with substantial improvement in PK through modifications at the P1 site, while potency gains were found with small, but rationally designed structural changes to P4. Additional modifications at P3 were required to optimize the CV profile, and these combined SARs led to the discovery of BMS-890068 (29).


Asunto(s)
Antivirales/química , Hepacivirus/efectos de los fármacos , Isoquinolinas/uso terapéutico , Oligopéptidos/química , Sulfonamidas/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/administración & dosificación , Antivirales/farmacocinética , Antivirales/farmacología , Perros , Esquema de Medicación , Farmacorresistencia Viral , Hepacivirus/genética , Macaca fascicularis , Masculino , Modelos Moleculares , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Conejos , Ratas Sprague-Dawley , Replicón , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
15.
J Med Chem ; 57(5): 1730-52, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24564672

RESUMEN

The discovery of asunaprevir (BMS-650032, 24) is described. This tripeptidic acylsulfonamide inhibitor of the NS3/4A enzyme is currently in phase III clinical trials for the treatment of hepatitis C virus infection. The discovery of 24 was enabled by employing an isolated rabbit heart model to screen for the cardiovascular (CV) liabilities (changes to HR and SNRT) that were responsible for the discontinuation of an earlier lead from this chemical series, BMS-605339 (1), from clinical trials. The structure-activity relationships (SARs) developed with respect to CV effects established that small structural changes to the P2* subsite of the molecule had a significant impact on the CV profile of a given compound. The antiviral activity, preclincial PK profile, and toxicology studies in rat and dog supported clinical development of BMS-650032 (24).


Asunto(s)
Antivirales/uso terapéutico , Hepatitis C/tratamiento farmacológico , Isoquinolinas/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Sulfonamidas/uso terapéutico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/sangre , Antivirales/química , Perros , Humanos , Isoquinolinas/sangre , Isoquinolinas/química , Modelos Moleculares , Inhibidores de Proteasas/sangre , Inhibidores de Proteasas/química , Conejos , Ratas , Sulfonamidas/sangre , Sulfonamidas/química
16.
J Med Chem ; 57(5): 1708-29, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24555570

RESUMEN

The discovery of BMS-605339 (35), a tripeptidic inhibitor of the NS3/4A enzyme, is described. This compound incorporates a cyclopropylacylsulfonamide moiety that was designed to improve the potency of carboxylic acid prototypes through the introduction of favorable nonbonding interactions within the S1' site of the protease. The identification of 35 was enabled through the optimization and balance of critical properties including potency and pharmacokinetics (PK). This was achieved through modulation of the P2* subsite of the inhibitor which identified the isoquinoline ring system as a key template for improving PK properties with further optimization achieved through functionalization. A methoxy moiety at the C6 position of this isoquinoline ring system proved to be optimal with respect to potency and PK, thus providing the clinical compound 35 which demonstrated antiviral activity in HCV-infected patients.


Asunto(s)
Antivirales/uso terapéutico , Descubrimiento de Drogas , Hepatitis C/tratamiento farmacológico , Isoquinolinas/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Sulfonamidas/uso terapéutico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Perros , Evaluación Preclínica de Medicamentos , Humanos , Isoquinolinas/química , Modelos Moleculares , Inhibidores de Proteasas/química , Sulfonamidas/química
17.
PLoS One ; 8(11): e79687, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260279

RESUMEN

Human ß-glucuronidase (GUS) cleaves ß-D-glucuronic acid residues from the non-reducing termini of glycosaminoglycan and its deficiency leads to mucopolysaccharidosis type VII (MPSVII). Here we report a high resolution crystal structure of human GUS at 1.7 Å resolution and present an extensive analysis of the structural features, unifying recent findings in the field of lysosome targeting and glycosyl hydrolases. The structure revealed several new details including a new glycan chain at Asn272, in addition to that previously observed at Asn173, and coordination of the glycan chain at Asn173 with Lys197 of the lysosomal targeting motif which is essential for phosphotransferase recognition. Analysis of the high resolution structure not only provided new insights into the structural basis for lysosomal targeting but showed significant differences between human GUS, which is medically important in its own right, and E. coli GUS, which can be selectively inhibited in the human gut to prevent prodrug activation and is also widely used as a reporter gene by plant biologists. Despite these differences, both human and E. coli GUS share a high structure homology in all three domains with most of the glycosyl hydrolases, suggesting that they all evolved from a common ancestral gene.


Asunto(s)
Glucuronidasa/química , Glucuronidasa/metabolismo , Lisosomas/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Glicosaminoglicanos/metabolismo , Humanos , Datos de Secuencia Molecular
18.
J Med Chem ; 56(18): 7343-57, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23964740

RESUMEN

Optimization of a 5-oxopyrrolopyridine series based upon structure-activity relationships (SARs) developed from our previous efforts on a number of related bicyclic series yielded compound 2s (BMS-767778) with an overall activity, selectivity, efficacy, PK, and developability profile suitable for progression into the clinic. SAR in the series and characterization of 2s are described.


Asunto(s)
Acetamidas/química , Acetamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Diseño de Fármacos , Pirroles/química , Pirroles/farmacología , Acetamidas/síntesis química , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Dominio Catalítico , Dipeptidil Peptidasa 4/química , Inhibidores de la Dipeptidil-Peptidasa IV/síntesis química , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Ratones , Modelos Moleculares , Pirroles/síntesis química , Especificidad por Sustrato
20.
J Med Chem ; 53(15): 5620-8, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20684603

RESUMEN

Continued structure-activity relationship (SAR) exploration within our previously disclosed azolopyrimidine containing dipeptidyl peptidase-4 (DPP4) inhibitors led us to focus on an imidazolopyrimidine series in particular. Further study revealed that by replacing the aryl substitution on the imidazole ring with a more polar carboxylic ester or amide, these compounds displayed not only increased DPP4 binding activity but also significantly reduced human ether-a-go-go related gene (hERG) and sodium channel inhibitory activities. Additional incremental adjustment of polarity led to permeable molecules which exhibited favorable pharmacokinetic (PK) profiles in preclinical animal species. The active site binding mode of these compounds was determined by X-ray crystallography as exemplified by amide 24c. A subsequent lead molecule from this series, (+)-6-(aminomethyl)-5-(2,4-dichlorophenyl)-N-(1-ethyl-1H-pyrazol-5-yl)-7-methylimidazo[1,2-a]pyrimidine-2-carboxamide (24s), emerged as a potent, selective DPP4 inhibitor that displayed excellent PK profiles and in vivo efficacy in ob/ob mice.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV , Hipoglucemiantes/síntesis química , Imidazoles/síntesis química , Pirimidinas/síntesis química , Animales , Dominio Catalítico , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4/química , Perros , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Humanos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Imidazoles/farmacocinética , Imidazoles/farmacología , Masculino , Ratones , Ratones Obesos , Modelos Moleculares , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Bloqueadores de los Canales de Sodio/farmacología , Estereoisomerismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA