Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(5)2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36901734

RESUMEN

Targeting the botulinum neurotoxin light chain (LC) metalloprotease using small-molecule metal chelate inhibitors is a promising approach to counter the effects of the lethal toxin. However, to overcome the pitfalls associated with simple reversible metal chelate inhibitors, it is crucial to investigate alternative scaffolds/strategies. In conjunction with Atomwise Inc., in silico and in vitro screenings were conducted, yielding a number of leads, including a novel 9-hydroxy-4H-pyrido [1,2-a]pyrimidin-4-one (PPO) scaffold. From this structure, an additional series of 43 derivatives were synthesized and tested, resulting in a lead candidate with a Ki of 150 nM in a BoNT/A LC enzyme assay and 17 µM in a motor neuron cell-based assay. These data combined with structure-activity relationship (SAR) analysis and docking led to a bifunctional design strategy, which we termed "catch and anchor" for the covalent inhibition of BoNT/A LC. Kinetic evaluation was conducted on structures prepared from this catch and anchor campaign, providing kinact/Ki values, and rationale for inhibition seen. Covalent modification was validated through additional assays, including an FRET endpoint assay, mass spectrometry, and exhaustive enzyme dialysis. The data presented support the PPO scaffold as a novel candidate for targeted covalent inhibition of BoNT/A LC.


Asunto(s)
Clostridium botulinum , Péptido Hidrolasas , Diálisis Renal , Relación Estructura-Actividad , Metaloproteasas , Metales , Inhibidores de Proteasas/farmacología
2.
ACS Med Chem Lett ; 14(2): 199-210, 2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36793435

RESUMEN

B cell lymphoma 6 (BCL6), a highly regulated transcriptional repressor, is deregulated in several forms of non-Hodgkin lymphoma (NHL), most notably in diffuse large B-cell lymphoma (DLBCL). The activities of BCL6 are dependent on protein-protein interactions with transcriptional co-repressors. To find new therapeutic interventions addressing the needs of patients with DLBCL, we initiated a program to identify BCL6 inhibitors that interfere with co-repressor binding. A virtual screen hit with binding activity in the high micromolar range was optimized by structure-guided methods, resulting in a novel and highly potent inhibitor series. Further optimization resulted in the lead candidate 58 (OICR12694/JNJ-65234637), a BCL6 inhibitor with low nanomolar DLBCL cell growth inhibition and an excellent oral pharmacokinetic profile. Based on its overall favorable preclinical profile, OICR12694 is a highly potent, orally bioavailable candidate for testing BCL6 inhibition in DLBCL and other neoplasms, particularly in combination with other therapies.

3.
Adv Sci (Weinh) ; 8(14): e2004846, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34060252

RESUMEN

Chromosomal translocation results in development of an Ewing sarcoma breakpoint region 1-Friend leukemia integration 1 (EWS-FLI1) fusion oncogene in the majority of Ewing sarcoma. The persistent dependence of the tumor for this oncoprotein points to EWS-FLI1 as an ideal drug target. Although EWS-FLI1 transcriptional targets and binding partners are evaluated, the mechanisms regulating EWS-FLI1 protein stability remain elusive. Speckle-type POZ protein (SPOP) and OTU domain-containing protein 7A (OTUD7A) are identified as the bona fide E3 ligase and deubiquitinase, respectively, that control EWS-FLI1 protein turnover in Ewing sarcoma. Casein kinase 1-mediated phosphorylation of the VTSSS degron in the FLI1 domain enhances SPOP activity to degrade EWS-FLI1. Opposing this process, OTUD7A deubiquitinates and stabilizes EWS-FLI1. Depletion of OTUD7A in Ewing sarcoma cell lines reduces EWS-FLI1 protein abundance and impedes Ewing sarcoma growth in vitro and in mice. Performing an artificial-intelligence-based virtual drug screen of a 4-million small molecule library, 7Ai is identified as a potential OTUD7A catalytic inhibitor. 7Ai reduces EWS-FLI1 protein levels and decreases Ewing sarcoma growth in vitro and in a xenograft mouse model. This study supports the therapeutic targeting of OTUD7A as a novel strategy for Ewing sarcoma bearing EWS-FLI1 and related fusions, and may also be applicable to other cancers dependent on aberrant FLI1 expression.


Asunto(s)
Enzimas Desubicuitinizantes/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Proteínas Represoras/genética , Sarcoma de Ewing/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Estabilidad Proteica
4.
J Biol Chem ; 295(39): 13516-13531, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32723867

RESUMEN

Prion disease is a rapidly progressive neurodegenerative disorder caused by misfolding and aggregation of the prion protein (PrP), and there are currently no therapeutic options. PrP ligands could theoretically antagonize prion formation by protecting the native protein from misfolding or by targeting it for degradation, but no validated small-molecule binders have been discovered to date. We deployed a variety of screening methods in an effort to discover binders of PrP, including 19F-observed and saturation transfer difference (STD) NMR spectroscopy, differential scanning fluorimetry (DSF), DNA-encoded library selection, and in silico screening. A single benzimidazole compound was confirmed in concentration-response, but affinity was very weak (Kd > 1 mm), and it could not be advanced further. The exceptionally low hit rate observed here suggests that PrP is a difficult target for small-molecule binders. Whereas orthogonal binder discovery methods could yield high-affinity compounds, non-small-molecule modalities may offer independent paths forward against prion disease.


Asunto(s)
Bencimidazoles/farmacología , Enfermedades por Prión/tratamiento farmacológico , Proteínas Priónicas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Bencimidazoles/química , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Espectroscopía de Resonancia Magnética , Enfermedades por Prión/metabolismo , Proteínas Priónicas/metabolismo , Bibliotecas de Moléculas Pequeñas/química
5.
Cell Metab ; 30(6): 1131-1140.e7, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564441

RESUMEN

The identification of molecular targets and pharmacodynamic markers for Parkinson's disease (PD) will empower more effective clinical management and experimental therapies. Miro1 is localized on the mitochondrial surface and mediates mitochondrial motility. Miro1 is removed from depolarized mitochondria to facilitate their clearance via mitophagy. Here, we explore the clinical utility of Miro1 for detecting PD and for gauging potential treatments. We measure the Miro1 response to mitochondrial depolarization using biochemical assays in skin fibroblasts from a broad spectrum of PD patients and discover that more than 94% of the patients' fibroblast cell lines fail to remove Miro1 following depolarization. We identify a small molecule that can repair this defect of Miro1 in PD fibroblasts. Treating patient-derived neurons and fly models with this compound rescues the locomotor deficits and dopaminergic neurodegeneration. Our results indicate that tracking this Miro1 marker and engaging in Miro1-based therapies could open new avenues to personalized medicine.


Asunto(s)
Antiparkinsonianos/farmacología , Proteínas de Drosophila , Proteínas Mitocondriales , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Enfermedad de Parkinson , Proteínas de Unión al GTP rho , Adulto , Anciano , Animales , Antiparkinsonianos/uso terapéutico , Biomarcadores/metabolismo , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Femenino , Fibroblastos , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neuronas/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
6.
Proteins ; 83(7): 1316-26, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25974248

RESUMEN

Protein families involved in chromatin-templated events are emerging as novel target classes in oncology and other disease areas. The ability to discover selective inhibitors against chromatin factors depends on the presence of structural features that are unique to the targeted sites. To evaluate challenges and opportunities toward the development of selective inhibitors, we calculated all pair wise structural distances between 575 structures from the protein databank representing 163 unique binding pockets found in protein domains that write, read or erase post-translational modifications on histones, DNA, and RNA. We find that the structural similarity of binding sites does not always follow the sequence similarity of protein domains. Our analysis reveals increased risks of activity across target-class for compounds competing with the cofactor of protein arginine methyltransferases, lysine acetyltransferases, and sirtuins, while exploiting the conformational plasticity of a protein target is a path toward selective inhibition. The structural diversity landscape of the epigenetics pocketome can be explored via an open-access graphic user interface at thesgc.org/epigenetics_pocketome.


Asunto(s)
Antineoplásicos/química , Epigénesis Genética , Histonas/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional , Bibliotecas de Moléculas Pequeñas/química , Programas Informáticos , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/química , Acetiltransferasas/genética , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Sitios de Unión , Cromatina/química , Cromatina/efectos de los fármacos , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/química , ADN de Neoplasias/genética , Bases de Datos de Proteínas , Histonas/química , Histonas/genética , Humanos , Internet , Ligandos , Modelos Moleculares , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/química , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Unión Proteica , Estructura Terciaria de Proteína , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , ARN Neoplásico/antagonistas & inhibidores , ARN Neoplásico/química , ARN Neoplásico/genética , Sirtuinas/antagonistas & inhibidores , Sirtuinas/química , Sirtuinas/genética , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/farmacología
7.
J Chem Inf Model ; 53(3): 681-91, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23410263

RESUMEN

Protein methyltransferases (PMTs) are a novel gene family of therapeutic relevance involved in chromatin-mediated signaling and other biological mechanisms. Most PMTs are organized around the structurally conserved SET domain that catalyzes the methylation of a substrate lysine. A few potent chemical inhibitors compete with the protein substrate, and all are anchored in the channel recruiting the methyl-accepting lysine. We propose a novel strategy to design focused chemical libraries targeting the substrate binding site, where a limited number of warheads each occupying the lysine-channel of multiple enzymes would be decorated by different substituents. A variety of sequence and structure-based approaches used to analyze the diversity of the lysine channel of SET domain PMTs support the relevance of this strategy. We show that chemical fragments derived from published inhibitors are valid warheads that can be used in the design of novel focused libraries targeting other PMTs.


Asunto(s)
Proteína Metiltransferasas/metabolismo , Aminoácidos/análisis , Cromatina/fisiología , Cristalografía por Rayos X , Diseño de Fármacos , Enlace de Hidrógeno , Canales Iónicos/metabolismo , Lisina/metabolismo , Modelos Moleculares , Filogenia , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas , Especificidad por Sustrato
8.
Bioorg Med Chem ; 21(7): 1787-1794, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23433670

RESUMEN

Chemical inhibition of proteins involved in chromatin-mediated signaling is an emerging strategy to control chromatin compaction with the aim to reprogram expression networks to alter disease states. Protein methyltransferases constitute one of the protein families that participate in epigenetic control of gene expression, and represent a novel therapeutic target class. Recruitment of the protein lysine methyltransferase DOT1L at aberrant loci is a frequent mechanism driving acute lymphoid and myeloid leukemias, particularly in infants, and pharmacological inhibition of DOT1L extends survival in a mouse model of mixed lineage leukemia. A better understanding of the structural chemistry of DOT1L inhibition would accelerate the development of improved compounds. Here, we report that the addition of a single halogen atom at a critical position in the cofactor product S-adenosylhomocysteine (SAH, an inhibitor of SAM-dependent methyltransferases) results in an 8-fold increase in potency against DOT1L, and reduced activities against other protein and non-protein methyltransferases. We solved the crystal structure of DOT1L in complex with Bromo-deaza-SAH and rationalized the observed effects. This discovery reveals a simple strategy to engineer selectivity and potency towards DOT1L into the adenosine scaffold of the cofactor shared by all methyltransferases, and can be exploited towards the development of clinical candidates against mixed lineage leukemia.


Asunto(s)
Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , S-Adenosilhomocisteína/análogos & derivados , S-Adenosilhomocisteína/farmacología , Cristalografía por Rayos X , Halogenación , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Metiltransferasas/química , Modelos Moleculares
9.
Biochem J ; 449(1): 151-9, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22989411

RESUMEN

WDR5 (WD40 repeat protein 5) is an essential component of the human trithorax-like family of SET1 [Su(var)3-9 enhancer-of-zeste trithorax 1] methyltransferase complexes that carry out trimethylation of histone 3 Lys4 (H3K4me3), play key roles in development and are abnormally expressed in many cancers. In the present study, we show that the interaction between WDR5 and peptides from the catalytic domain of MLL (mixed-lineage leukaemia protein) (KMT2) can be antagonized with a small molecule. Structural and biophysical analysis show that this antagonist binds in the WDR5 peptide-binding pocket with a Kd of 450 nM and inhibits the catalytic activity of the MLL core complex in vitro. The degree of inhibition was enhanced at lower protein concentrations consistent with a role for WDR5 in directly stabilizing the MLL multiprotein complex. Our data demonstrate inhibition of an important protein-protein interaction and form the basis for further development of inhibitors of WDR5-dependent enzymes implicated in MLL-rearranged leukaemias or other cancers.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Dominio Catalítico/fisiología , Cristalografía por Rayos X , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Péptidos y Proteínas de Señalización Intracelular , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología
10.
ACS Med Chem Lett ; 4(3): 353-7, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24900672

RESUMEN

The WD40-repeat protein WDR5 plays a critical role in maintaining the integrity of MLL complexes and fully activating their methyltransferase function. MLL complexes, the trithorax-like family of SET1 methyltransferases, catalyze trimethylation of lysine 4 on histone 3, and they have been widely implicated in various cancers. Antagonism of WDR5 and MLL subunit interaction by small molecules has recently been presented as a practical way to inhibit activity of the MLL1 complex, and N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides were reported as potent and selective antagonists of such an interaction. Here, we describe the protein crystal structure guided optimization of prototypic compound 2 (K dis = 7 µM), leading to identification of more potent antagonist 47 (K dis = 0.3 µM).

11.
J Chem Inf Model ; 51(8): 1817-30, 2011 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-21699246

RESUMEN

Drug discovery research often relies on the use of virtual screening via molecular docking to identify active hits in compound libraries. An area for improvement among many state-of-the-art docking methods is the accuracy of the scoring functions used to differentiate active from nonactive ligands. Many contemporary scoring functions are influenced by the physical properties of the docked molecule. This bias can cause molecules with certain physical properties to incorrectly score better than others. Since variation in physical properties is inevitable in large screening libraries, it is desirable to account for this bias. In this paper, we present a method of normalizing docking scores using virtually generated decoy sets with matched physical properties. First, our method generates a set of property-matched decoys for every molecule in the screening library. Each library molecule and its decoy set are docked using a state-of-the-art method, producing a set of raw docking scores. Next, the raw docking score of each library molecule is normalized against the scores of its decoys. The normalized score represents the probability that the raw docking score was drawn from the background distribution of nonactive property-matched decoys. Assuming that the distribution of scores of active molecules differs from the nonactive score distribution, we expect that the score of an active compound will have a low probability of having been drawn from the nonactive score distribution. In addition to the use of decoys in normalizing docking scores, we suggest that decoy sets may be a useful tool to evaluate, improve, or develop scoring functions. We show that by analyzing docking scores of library molecules with respect to the docking scores of their virtually generated property-matched decoys, one can gain insight into the advantages, limitations, and reliability of scoring functions.


Asunto(s)
Química Farmacéutica/métodos , Descubrimiento de Drogas/métodos , Proteínas/análisis , Algoritmos , Sitios de Unión , Química Farmacéutica/estadística & datos numéricos , Minería de Datos , Bases de Datos Factuales , Descubrimiento de Drogas/estadística & datos numéricos , Ligandos , Modelos Moleculares , Modelos Estadísticos , Posición Específica de Matrices de Puntuación , Unión Proteica , Proteínas/química
12.
J Chem Inf Model ; 51(3): 612-23, 2011 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-21366357

RESUMEN

Histone methyltransferases (HMTs) transfer a methyl group from the cofactor S-adenosyl methionine to lysine or arginine residues on histone tails, thereby regulating chromatin compaction, binding of effector proteins and gene transcription. HMTs constitute an emerging target class in diverse disease areas, and selective chemical probes are necessary for target validation. Potent and selective competitors of the substrate peptide have been reported, but the chemical tractability of the cofactor binding site is poorly understood. Here, a systematic analysis of this site across structures of 14 human HMTs or close homologues was conducted. The druggability, interaction hotspots, and diversity of the cofactor binding pocket were dissected. This analysis strongly suggests that this site is chemically tractable. General principles underlying tight binding and specific guidelines to achieve selective inhibition are presented.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/química , Sitios de Unión , Histona Metiltransferasas , Humanos , Modelos Moleculares , Filogenia , Conformación Proteica , Alineación de Secuencia
13.
J Med Chem ; 53(19): 7236-50, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20812729

RESUMEN

A variety of conformationally constrained aspartate and glutamate analogues inhibit the glutamate transporter 1 (GLT-1, also known as EAAT2). To expand the search for such analogues, a virtual library of aliphatic aspartate and glutamate analogues was generated starting from the chemical universe database GDB-11, which contains 26.4 million possible molecules up to 11 atoms of C, N, O, F, resulting in 101026 aspartate analogues and 151285 glutamate analogues. Virtual screening was realized by high-throughput docking to the glutamate binding site of the glutamate transporter homologue from Pyrococcus horikoshii (PDB code: 1XFH ) using Autodock. Norbornane-type aspartate analogues were selected from the top-scoring virtual hits and synthesized. Testing and optimization led to the identification of (1R*,2R*,3S*,4R*,6R*)-2-amino-6-phenethyl-bicyclo[2.2.1]heptane-2,3-dicarboxylic acid as a new inhibitor of GLT-1 with IC(50) = 1.4 µM against GLT-1 and no inhibition of the related transporter EAAC1. The systematic diversification of known ligands by enumeration with help of GDB followed by virtual screening, synthesis, and testing as exemplified here provides a general strategy for drug discovery.


Asunto(s)
Aminoácidos Cíclicos/síntesis química , Ácido Aspártico/análogos & derivados , Ácido Aspártico/síntesis química , Bases de Datos Factuales , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Glutamatos/síntesis química , Norbornanos/síntesis química , Aminoácidos Cíclicos/química , Aminoácidos Cíclicos/farmacología , Animales , Ácido Aspártico/química , Ácido Aspártico/farmacología , Proteínas Bacterianas/química , Transportador 3 de Aminoácidos Excitadores/antagonistas & inhibidores , Femenino , Glutamatos/química , Glutamatos/farmacología , Ligandos , Modelos Moleculares , Conformación Molecular , Norbornanos/química , Norbornanos/farmacología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Unión Proteica , Pyrococcus horikoshii , Estereoisomerismo , Relación Estructura-Actividad , Xenopus laevis
14.
ACS Med Chem Lett ; 1(8): 422-6, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24900227

RESUMEN

Virtual analogues (1167860 compounds) of the nicotinic α7-receptor (α7 nAChR) ligands PNU-282,987 and SSR180711 were generated from the chemical universe database GDB-11 by extracting all aliphatic diamine analogues of the aminoquinuclidine and 1,4-diazabicyclo[3.2.2]nonane scaffolds of these ligands and converting them to the corresponding aryl amides using five different aromatic acyl groups. The library was ranked by docking to the nicotinic binding site of the acetylcholine binding protein (AChBP, 1UW6.pdb) using Autodock and Glide. Thirty-eight ligands derived from the best docking hits were synthesized and tested for modulation of the acetylcholine signal at the human α7 nAChR receptor expressed in Xenopus oocytes, leading to competitive and noncompetitive antagonists with IC50 = 5-7 µM. These experiments demonstrate the first example of using GDB in a fragment-based approach by diversifying the scaffold of known drugs.

16.
Bioorg Med Chem Lett ; 19(14): 3832-5, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19394821

RESUMEN

Docking of randomly selected compounds from the chemical universe database GDB-11, which contains all organic molecules up to 11 atoms of C, N, O, F possible under consideration of simple chemical stability and synthetic feasibility rules, into the NMDA receptor glycine site (1pb7.pdb) lead to the identification of 3-(aminomethyl)piperazine-2,5-dione 3 and its close analog 5-(aminomethyl)piperazine-2,3-dione 4 as possible new ligands for this drug target, which is implicated in synaptic plasticity, neuronal development, learning and memory. Synthesis of these compounds in 4 and 6 steps, respectively, and testing by radioligand displacement assays and electrophysiological measurements in Xenopus oocytes show that while 4 is inactive, 3 is indeed an inhibitor of glycine, with an estimated K(D) of 50 microM.


Asunto(s)
Dicetopiperazinas/química , Glicina/química , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Sitios de Unión , Simulación por Computador , Bases de Datos Factuales , Dicetopiperazinas/síntesis química , Dicetopiperazinas/farmacología , Oocitos/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Programas Informáticos , Termodinámica , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...