Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Biochem Sci ; 49(3): 195-198, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38195289

RESUMEN

Targeting translational factor proteins (TFPs) presents significant promise for the development of innovative antitubercular drugs. Previous insights from antibiotic binding mechanisms and recently solved 3D crystal structures of Mycobacterium tuberculosis (Mtb) elongation factor thermo unstable-GDP (EF-Tu-GDP), elongation factor thermo stable-EF-Tu (EF-Ts-EF-Tu), and elongation factor G-GDP (EF-G-GDP) have opened up new avenues for the design and development of potent antituberculosis (anti-TB) therapies.


Asunto(s)
Antituberculosos , Factor Tu de Elongación Peptídica , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Factor Tu de Elongación Peptídica/química , Factor Tu de Elongación Peptídica/metabolismo , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Factores de Elongación de Péptidos/química , Factores de Elongación de Péptidos/metabolismo , Proteínas/metabolismo
2.
Med Res Rev ; 44(3): 1055-1120, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38142308

RESUMEN

Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.


Asunto(s)
Leishmania , Leishmaniasis Cutánea , Leishmaniasis Visceral , Humanos , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/complicaciones , Leishmaniasis Visceral/epidemiología , Leishmaniasis Cutánea/tratamiento farmacológico , Leishmaniasis Cutánea/complicaciones , Leishmaniasis Cutánea/epidemiología , Ensayos Analíticos de Alto Rendimiento
3.
Int J Mol Sci ; 24(11)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37298573

RESUMEN

The platelet-derived growth factor receptor (PDGFR) is a membrane tyrosine kinase receptor involved in several metabolic pathways, not only physiological but also pathological, as in tumor progression, immune-mediated diseases, and viral diseases. Considering this macromolecule as a druggable target for modulation/inhibition of these conditions, the aim of this work was to find new ligands or new information to design novel effective drugs. We performed an initial interaction screening with the human intracellular PDGFRα of about 7200 drugs and natural compounds contained in 5 independent databases/libraries implemented in the MTiOpenScreen web server. After the selection of 27 compounds, a structural analysis of the obtained complexes was performed. Three-dimensional quantitative structure-activity relationship (3D-QSAR) and absorption, distribution, metabolism, excretion, and toxicity (ADMET) analyses were also performed to understand the physicochemical properties of identified compounds to increase affinity and selectivity for PDGFRα. Among these 27 compounds, the drugs Bafetinib, Radotinib, Flumatinib, and Imatinib showed higher affinity for this tyrosine kinase receptor, lying in the nanomolar order, while the natural products included in this group, such as curcumin, luteolin, and epigallocatechin gallate (EGCG), showed sub-micromolar affinities. Although experimental studies are mandatory to fully understand the mechanisms behind PDGFRα inhibitors, the structural information obtained through this study could provide useful insight into the future development of more effective and targeted treatments for PDGFRα-related diseases, such as cancer and fibrosis.


Asunto(s)
Neoplasias , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Humanos , Simulación del Acoplamiento Molecular , Modelos Moleculares , Mesilato de Imatinib/farmacología , Relación Estructura-Actividad Cuantitativa
4.
Biochem J ; 478(15): 2953-2975, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34375386

RESUMEN

The Unfolded Protein response is an adaptive pathway triggered upon alteration of endoplasmic reticulum (ER) homeostasis. It is transduced by three major ER stress sensors, among which the Inositol Requiring Enzyme 1 (IRE1) is the most evolutionarily conserved. IRE1 is an ER-resident type I transmembrane protein exhibiting an ER luminal domain that senses the protein folding status and a catalytic kinase and RNase cytosolic domain. In recent years, IRE1 has emerged as a relevant therapeutic target in various diseases including degenerative, inflammatory and metabolic pathologies and cancer. As such several drugs altering IRE1 activity were developed that target either catalytic activity and showed some efficacy in preclinical pathological mouse models. In this review, we describe the different drugs identified to target IRE1 activity as well as their mode of action from a structural perspective, thereby identifying common and different modes of action. Based on this information we discuss on how new IRE1-targeting drugs could be developed that outperform the currently available molecules.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Endorribonucleasas/metabolismo , Homeostasis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Homeostasis/efectos de los fármacos , Humanos , Pliegue de Proteína/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Respuesta de Proteína Desplegada/efectos de los fármacos
5.
Prog Med Chem ; 60: 273-343, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34147204

RESUMEN

Molecular docking has become an important component of the drug discovery process. Since first being developed in the 1980s, advancements in the power of computer hardware and the increasing number of and ease of access to small molecule and protein structures have contributed to the development of improved methods, making docking more popular in both industrial and academic settings. Over the years, the modalities by which docking is used to assist the different tasks of drug discovery have changed. Although initially developed and used as a standalone method, docking is now mostly employed in combination with other computational approaches within integrated workflows. Despite its invaluable contribution to the drug discovery process, molecular docking is still far from perfect. In this chapter we will provide an introduction to molecular docking and to the different docking procedures with a focus on several considerations and protocols, including protonation states, active site waters and consensus, that can greatly improve the docking results.


Asunto(s)
Descubrimiento de Drogas/métodos , Simulación del Acoplamiento Molecular , Proteínas/química , Proteínas/metabolismo , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
6.
Bioorg Med Chem ; 52: 116514, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34808405

RESUMEN

Pyruvate dehydrogenase kinases (PDHKs) are fascinating drug targets for numerous diseases, including diabetes and cancers. In this report, we describe the result of our structure-based drug design from tricyclic lead compounds that led to the discovery of highly potent PDHK2 and PDHK4 dual inhibitors in enzymatic assay. The C3-position of the tricyclic core was explored, and the PDHK2 X-ray structure with a representative compound revealed a novel ATP lid conformation in which the phenyl ring of Phe326 mediated the interaction of the Arg258 sidechain and the compound. Compounds with amide linkers were designed to release the ATP lid by forming an intramolecular pi-pi interaction, and these compounds showed single-digit nM IC50 values in an enzymatic assay. We also explored the C4-position of the tricyclic core to reproduce the interaction observed with the C3-position substitution, and the pyrrolidine compound showed the same level of IC50 values. By optimizing an interaction with the Asn255 sidechain through a docking simulation, compounds with 2-carboxy pyrrole moiety also showed single-digit nM IC50 values without having a cation-pi interaction with the Arg258 sidechain.


Asunto(s)
Adenosina Trifosfato/farmacología , Amidas/farmacología , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/química , Amidas/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Relación Estructura-Actividad
7.
J Biomol NMR ; 74(10-11): 595-611, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32761504

RESUMEN

The presence of suitable cavities or pockets on protein structures is a general criterion for a therapeutic target protein to be classified as 'druggable'. Many disease-related proteins that function solely through protein-protein interactions lack such pockets, making development of inhibitors by traditional small-molecule structure-based design methods much more challenging. The 22 kDa bacterial thiol oxidoreductase enzyme, DsbA, from the gram-negative bacterium Burkholderia pseudomallei (BpsDsbA) is an example of one such target. The crystal structure of oxidized BpsDsbA lacks well-defined surface pockets. BpsDsbA is required for the correct folding of numerous virulence factors in B. pseudomallei, and genetic deletion of dsbA significantly attenuates B. pseudomallei virulence in murine infection models. Therefore, BpsDsbA is potentially an attractive drug target. Herein we report the identification of a small molecule binding site adjacent to the catalytic site of oxidized BpsDsbA. 1HN CPMG relaxation dispersion NMR measurements suggest that the binding site is formed transiently through protein dynamics. Using fragment-based screening, we identified a small molecule that binds at this site with an estimated affinity of KD ~ 500 µM. This fragment inhibits BpsDsbA enzymatic activity in vitro. The binding mode of this molecule has been characterized by NMR data-driven docking using HADDOCK. These data provide a starting point towards the design of more potent small molecule inhibitors of BpsDsbA.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular/métodos , Proteína Disulfuro Reductasa (Glutatión)/química , Animales , Sitios de Unión , Burkholderia pseudomallei/enzimología , Burkholderia pseudomallei/patogenicidad , Dominio Catalítico , Ligandos , Ratones , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Proteína Disulfuro Reductasa (Glutatión)/genética , Relación Estructura-Actividad Cuantitativa , Proteínas Recombinantes , Bibliotecas de Moléculas Pequeñas/química , Solubilidad , Tiazoles/química
8.
Bioorg Med Chem Lett ; 30(17): 127405, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32738982

RESUMEN

Apoptosis Signal-Regulating Kinase-1 (ASK1) is a known member of the Mitogen-Activated Protein Kinase Kinase Kinase (MAP3K) family and upon stimulation will activate the p38- and JNK-pathways leading to cardiac apoptosis, fibrosis, and hypertrophy. Using Structure-Based Drug Design (SBDD) in parallel with deconstruction of a published compound, a novel series of ASK1 inhibitors was optimized, which incorporated a saturated heterocycle proximal to the hinge-binding motif. This yielded a unique chemical series with excellent selectivity across the broader kinome, and desirable drug-like properties. The lead compound (10) is highly soluble and permeable, and exhibits a cellular EC50 = 24 nM and Kd < 1 nM. Of the 350 kinases tested, 10 has an IC50 ≤ 500 nM for only eight of them. This paper will describe the design hypotheses behind this series, key data points during the optimization phase, as well as a possible structural rationale for the kinome selectivity. Based on crystallographic data, the presence of an aliphatic cycle adjacent to the hinge-binder in the active site of the protein kinase showed up in <1% of the >5000 structures in the Protein Data Bank, potentially conferring the selectivity seen in this series.


Asunto(s)
MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Animales , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/uso terapéutico , Concentración 50 Inhibidora , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Simulación de Dinámica Molecular , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico
9.
Bioorg Med Chem Lett ; 27(13): 2825-2837, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28479196

RESUMEN

In structure-based drug design, the basic goal is to design molecules that fit complementarily to a given binding pocket. Since such computationally modeled molecules may not adopt the intended bound conformation outside the binding pocket, one challenge is to ensure that the designed ligands adopt similar low energy conformations both inside and outside of the binding pocket. Computational chemistry methods and conformational preferences of small molecules from PDB and Cambridge Structural Database (CSD) can be used to predict the bound structures of the designed molecules. Herein, we review applications of conformational control in structure-based drug design using selected examples from the recent medicinal chemistry literature. The main purpose is to highlight some intriguing conformational features that can be applied to other drug discovery programs.


Asunto(s)
Amidas/química , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Amidas/síntesis química , Amidas/farmacología , Química Farmacéutica , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Ligandos , Modelos Moleculares , Conformación Molecular , Relación Estructura-Actividad
10.
Bioorg Med Chem Lett ; 27(8): 1709-1713, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28291695

RESUMEN

Structure-based drug design is an iterative process that is an established means to accelerate lead optimization, and is most powerful when integrated with information from different sources. Herein is described the use of such methods in conjunction with deconstruction and re-optimization of a diverse series of ASK1 chemotypes along with high-throughput screening that lead to the identification of a novel series of efficient ASK1 inhibitors displaying robust MAP3K pathway inhibition.


Asunto(s)
Diseño de Fármacos , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , MAP Quinasa Quinasa Quinasa 5/química , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Simulación del Acoplamiento Molecular
11.
Bioorg Med Chem ; 25(17): 4876-4886, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28760529

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcriptional repressor that can form complexes with corepressors via protein-protein interactions (PPIs). The complexes of BCL6 and corepressors play an important role in the formation of germinal centers (GCs), and differentiation and proliferation of lymphocytes. Therefore, BCL6-corepressor interaction inhibitors would be drug candidates for managing autoimmune diseases and cancer. Starting from high-throughput screening hits 1a and 2a, we identified a novel BCL6-corepressor interaction inhibitor 8c (cell-free enzyme-linked immunosorbent assay [ELISA] IC50=0.10µM, cell-based mammalian two-hybrid [M2H] assay IC50=0.72µM) by utilizing structure-based drug design (SBDD) based on an X-ray crystal structure of 1a bound to BCL6. Compound 8c also showed a good pharmacokinetic profile, which was acceptable for both in vitro and in vivo studies.


Asunto(s)
Diseño de Fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Aminas/química , Aminas/metabolismo , Aminas/farmacocinética , Sitios de Unión , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Semivida , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Simulación de Dinámica Molecular , Unión Proteica , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Técnicas del Sistema de Dos Híbridos
12.
Bioorg Med Chem ; 25(7): 2105-2132, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28259528

RESUMEN

A predictive structure-based 3D QSAR (COMBINEr 2.0) model of the Schistosoma mansoni lysine deacetylase 8 enzyme (SmKDAC8) was developed, validated and used to perform virtual screening (VS) of the NCI Diversity Set V database (1593 compounds). Three external datasets (with congeneric structures to those experimentally resolved in complexes by X-ray and previously reported as SmKDAC8 inhibitors) were employed to compose and validate the most predictive model. Two series characterized by 104 benzodiazepine derivatives (BZDs) and 60 simplified largazole analogs (SLAs), recently reported by our group as human KDAC inhibitors, were tested for their inhibition potency against SmKDAC8 to probe the predictive capability of the quantitative models against compounds with diverse structures. The SmKDAC8 biochemical results confirmed: (1) the benzodiazepine moiety as a valuable scaffold to further investigate when pursuing SmKDAC8 inhibition; (2) the predictive capability of the COMBINEr 2.0 model towards non-congeneric series of compounds, highlighting the most influencing ligand-protein interactions and refining the structure-activity relationships. From the VS investigations, the first 40 top-ranked compounds were obtained and biologically tested for their inhibition potency against SmKDAC8 and hKDACs 1, 3, 6 and 8. Among them, a non-hydroxamic acid benzothiadiazine dioxide derivative (code NSC163639), showed interesting activity and selectivity against SmKDAC8. To further elucidate the structure-activity relationships of NSC163639, two analogs (herein reported as compounds 3 and 4) were synthesized and biologically evaluated. Results suggest the benzothiadiazine dioxide moiety as a promising scaffold to be used in a next step to derive selective SmKDAC8 inhibitors.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Schistosoma mansoni/efectos de los fármacos , Animales , Inhibidores de Histona Desacetilasas/química , Técnicas In Vitro , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad Cuantitativa , Schistosoma mansoni/enzimología , Schistosoma mansoni/genética
13.
Bioorg Med Chem ; 25(24): 6563-6580, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29133033

RESUMEN

A series of A-ring-modified lamellarin N analogues were designed, synthesized, and evaluated as potential noncovalent inhibitors of the EGFR T790M/L858R mutant, a causal factor in the drug-resistant non-small cell lung cancer. Several water-soluble ammonium- or guanidinium-tethered analogues exhibited good kinase inhibitory activities. The most promising analogue, 14f, displayed an excellent inhibitory profile against the T790M/L858R mutant [IC50 (WT) = 31.8 nM; IC50 (T790M/L858R) = 8.9 nM]. The effects of A-ring-substituents on activity were rationalized by docking studies.


Asunto(s)
Diseño de Fármacos , Receptores ErbB/antagonistas & inhibidores , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Relación Dosis-Respuesta a Droga , Receptores ErbB/genética , Receptores ErbB/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/síntesis química , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Mutación , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
14.
Molecules ; 22(11)2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-29125553

RESUMEN

The four receptors that signal for adenosine, A1, A2A, A2B and A3 ARs, belong to the superfamily of G protein-coupled receptors (GPCRs). They mediate a number of (patho)physiological functions and have attracted the interest of the biopharmaceutical sector for decades as potential drug targets. The many crystal structures of the A2A, and lately the A1 ARs, allow for the use of advanced computational, structure-based ligand design methodologies. Over the last decade, we have assessed the efficient synthesis of novel ligands specifically addressed to each of the four ARs. We herein review and update the results of this program with particular focus on molecular dynamics (MD) and free energy perturbation (FEP) protocols. The first in silico mutagenesis on the A1AR here reported allows understanding the specificity and high affinity of the xanthine-antagonist 8-Cyclopentyl-1,3-dipropylxanthine (DPCPX). On the A2AAR, we demonstrate how FEP simulations can distinguish the conformational selectivity of a recent series of partial agonists. These novel results are complemented with the revision of the first series of enantiospecific antagonists on the A2BAR, and the use of FEP as a tool for bioisosteric design on the A3AR.


Asunto(s)
Receptores Purinérgicos P1/química , Ligandos , Simulación de Dinámica Molecular , Mutación/genética , Antagonistas de Receptores Purinérgicos P1/química , Estereoisomerismo , Termodinámica , Xantinas/química
15.
Bioorg Med Chem ; 24(22): 5771-5780, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27687967

RESUMEN

The action of the aspartyl protease renin is the rate-limiting initial step of the renin-angiotensin-aldosterone system. Therefore, renin is a particularly promising target for blood pressure as well as onset and progression of cardiovascular and renal diseases. New pyrimidine derivatives 5-14 were designed in an attempt to enhance the renin inhibitory activity of compound 3 identified by our previous fragment-based drug design approach. Introduction of a basic amine essential for interaction with the two aspartic acids in the catalytic site and optimization of the S1/S3 binding elements including an induced-fit structural change of Leu114 ('Leu-in' to 'Leu-out') by a rational structure-based drug design approach led to the discovery of N-(piperidin-3-yl)pyrimidine-5-carboxamide 14, a 65,000-fold more potent renin inhibitor than compound 3. Surprisingly, this remarkable enhancement in the inhibitory activity of compound 14 has been achieved by the overall addition of only seven heavy atoms to compound 3. Compound 14 demonstrated excellent selectivity over other aspartyl proteases and moderate oral bioavailability in rats.


Asunto(s)
Diseño de Fármacos , Piperidinas/farmacología , Inhibidores de Proteasas/farmacología , Pirimidinas/farmacología , Renina/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Ratas Sprague-Dawley , Renina/sangre , Relación Estructura-Actividad
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): 3849-55, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27345756

RESUMEN

Chagas disease, also called American trypanosomiasis, is a parasitic disease caused by Trypanosoma cruzi (T. cruzi). Recent findings have underscored the abundance of the causative organism, (T. cruzi), especially in the southern tier states of the US and the risk burden for the rural farming communities there. Due to a lack of safe and effective drugs, there is an urgent need for novel therapeutic options for treating Chagas disease. We report here our first scientific effort to pursue a novel drug design for treating Chagas disease via the targeting of T. cruzi tubulin. First, the anti T. cruzi tubulin activities of five naphthoquinone derivatives were determined and correlated to their anti-trypanosomal activities. The correlation between the ligand activities against the T. cruzi organism and their tubulin inhibitory activities was very strong with a Pearson's r value of 0.88 (P value <0.05), indicating that this class of compounds could inhibit the activity of the trypanosome organism via T. cruzi tubulin polymerization inhibition. Subsequent molecular modeling studies were carried out to understand the mechanisms of the anti-tubulin activities, wherein, the homology model of T. cruzi tubulin dimer was generated and the putative binding site of naphthoquinone derivatives was predicted. The correlation coefficient for ligand anti-tubulin activities and their binding energies at the putative pocket was found to be r=0.79, a high correlation efficiency that was not replicated in contiguous candidate pockets. The homology model of T. cruzi tubulin and the identification of its putative binding site lay a solid ground for further structure based drug design, including molecular docking and pharmacophore analysis. This study presents a new opportunity for designing potent and selective drugs for Chagas disease.


Asunto(s)
Enfermedad de Chagas/tratamiento farmacológico , Tripanocidas/química , Tripanocidas/farmacología , Trypanosoma cruzi/efectos de los fármacos , Tubulina (Proteína)/efectos de los fármacos , Secuencia de Aminoácidos , Diseño de Fármacos , Humanos , Polimerizacion , Homología de Secuencia de Aminoácido , Tripanocidas/uso terapéutico , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
18.
Molecules ; 20(11): 20551-68, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26610437

RESUMEN

Hypoxia-inducible factor (HIF) prolyl hydroxylases (PHDs) are members of the 2-oxoglutarate dependent non-heme iron dioxygenases. Due to their physiological roles in regulation of HIF-1α stability, many efforts have been focused on searching for selective PHD inhibitors to control HIF-1α levels for therapeutic applications. In this review, we first describe the structure of PHD2 as a molecular basis for structure-based drug design (SBDD) and various experimental methods developed for measuring PHD activity. We further discuss the current status of the development of PHD inhibitors enabled by combining SBDD approaches with high-throughput screening. Finally, we highlight the clinical implications of small molecule PHD inhibitors.


Asunto(s)
Descubrimiento de Drogas , Prolil Hidroxilasas/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacología , Anemia/tratamiento farmacológico , Anemia/metabolismo , Animales , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Conformación Molecular , Prolil Hidroxilasas/química , Inhibidores de Prolil-Hidroxilasa/química , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
19.
Bioorg Med Chem Lett ; 24(19): 4714-4723, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25193232

RESUMEN

Use of the tools of SBDD including crystallography led to the discovery of novel and potent 6,5 heterobicyclic MEKi's [J. Med. Chem.2012, 55, 4594]. The core change from a 5,6 heterobicycle to a 6,5 heterobicycle was driven by the desire for increased structural diversity and aided by the co-crystal structure of G-925 [J. Med. Chem.2012, 55, 4594]. The key design feature was the shift of the attachment of the five-membered heterocyclic ring towards the B ring while maintaining the key hydroxamate and anilino pharamcophoric elements in a remarkably similar position as in G-925. From modelling, changing the connection point of the five membered ring heterocycle placed the H-bond accepting nitrogen within a good distance and angle to the Ser212 [J. Med. Chem.2012, 55, 4594]. The resulting novel 6,5 benzoisothiazole MEKi G-155 exhibited improved potency versus aza-benzofurans G-925 and G-963 but was a potent inhibitor of cytochrome P450's 2C9 and 2C19. Lowering the logD by switching to the more polar imidazo[1,5-a] pyridine core significantly diminished 2C9/2C19 inhibition while retaining potency. The imidazo[1,5-a] pyridine G-868 exhibited increased potency versus the starting point for this work (aza-benzofuran G-925) leading to deprioritization of the azabenzofurans. The 6,5-imidazo[1,5-a] pyridine scaffold was further diversified by incorporating a nitrogen at the 7 position to give the imidazo[1,5-a] pyrazine scaffold. The introduction of the C7 nitrogen was driven by the desire to improve metabolic stability by blocking metabolism at the C7 and C8 positions (particularly the HLM stability). It was found that improving on G-868 (later renamed GDC-0623) required combining C7 nitrogen with a diol hydroxamate to give G-479. G-479 with polarity distributed throughout the molecule was improved over G-868 in many aspects.


Asunto(s)
Descubrimiento de Drogas , Compuestos Heterocíclicos/farmacología , Imidazoles/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HCT116 , Compuestos Heterocíclicos/síntesis química , Compuestos Heterocíclicos/química , Humanos , Imidazoles/síntesis química , Imidazoles/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazinas/síntesis química , Pirazinas/química , Relación Estructura-Actividad
20.
Eur J Med Chem ; 268: 116262, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38387334

RESUMEN

Peptides can bind challenging disease targets with high affinity and specificity, offering enormous opportunities for addressing unmet medical needs. However, peptides' unique features, including smaller size, increased structural flexibility, and limited data availability, pose additional challenges to the design process compared to proteins. This review explores the dynamic field of peptide therapeutics, leveraging deep learning to enhance structure prediction and design. Our exploration encompasses various facets of peptide research, ranging from dataset curation handling to model development. As deep learning technologies become more refined, we channel our efforts into peptide structure prediction and design, aligning with the fundamental principles of structure-activity relationships in drug development. To guide researchers in harnessing the potential of deep learning to advance peptide drug development, our insights comprehensively explore current challenges and future directions of peptide therapeutics.


Asunto(s)
Aprendizaje Profundo , Péptidos/farmacología , Desarrollo de Medicamentos , Relación Estructura-Actividad , Tecnología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA