Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Med Chem ; 67(4): 3112-3126, 2024 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-38325398

RESUMEN

CDK2 is a critical regulator of the cell cycle. For a variety of human cancers, the dysregulation of CDK2/cyclin E1 can lead to tumor growth and proliferation. Historically, early efforts to develop CDK2 inhibitors with clinical applications proved unsuccessful due to challenges in achieving selectivity over off-target CDK isoforms with associated toxicity. In this report, we describe the discovery of (4-pyrazolyl)-2-aminopyrimidines as a potent class of CDK2 inhibitors that display selectivity over CDKs 1, 4, 6, 7, and 9. SAR studies led to the identification of compound 17, a kinase selective and highly potent CDK2 inhibitor (IC50 = 0.29 nM). The evaluation of 17 in CCNE1-amplified mouse models shows the pharmacodynamic inhibition of CDK2, measured by reduced Rb phosphorylation, and antitumor activity.


Asunto(s)
Quinasas Ciclina-Dependientes , Neoplasias , Animales , Humanos , Ratones , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina/metabolismo , Fosforilación , Pirimidinas/farmacología , Pirazoles/química , Pirazoles/metabolismo , Pirazoles/farmacología
2.
ACS Med Chem Lett ; 13(7): 1159-1164, 2022 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-35859885

RESUMEN

Activin receptor-like kinase 2 (ALK2) is a transmembrane kinase receptor that mediates the signaling of the members of the TGF-ß superfamily. The aberrant activation of ALK2 has been linked to the rare genetic disorder fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) that are associated with severely reduced life expectancy in pediatric patients. ALK2 has also been shown to play an essential role in iron metabolism by regulating hepcidin levels and affecting anemia of chronic disease. Thus, selective inhibition of ALK2 has emerged as a promising strategy for the treatment of multiple disorders. Herein, we report the discovery of a novel pyrazolopyrimidines series as highly potent, selective, and orally bioavailable inhibitors of ALK2. Structure-based drug design and systematic structure-activity relationship studies were employed to identify potent inhibitors displaying high selectivity against other ALK subtypes with good pharmacokinetic profiles.

3.
Cancer Discov ; 12(6): 1482-1499, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35254416

RESUMEN

Blocking the activity of the programmed cell death protein 1 (PD-1) inhibitory receptor with therapeutic antibodies against either the ligand (PD-L1) or PD-1 itself has proven to be an effective treatment modality for multiple cancers. Contrasting with antibodies, small molecules could demonstrate increased tissue penetration, distinct pharmacology, and potentially enhanced antitumor activity. Here, we describe the identification and characterization of INCB086550, a novel, oral, small-molecule PD-L1 inhibitor. In vitro, INCB086550 selectively and potently blocked the PD-L1/PD-1 interaction, induced PD-L1 dimerization and internalization, and induced stimulation-dependent cytokine production in primary human immune cells. In vivo, INCB086550 reduced tumor growth in CD34+ humanized mice and induced T-cell activation gene signatures, consistent with PD-L1/PD-1 pathway blockade. Preliminary data from an ongoing phase I study confirmed PD-L1/PD-1 blockade in peripheral blood cells, with increased immune activation and tumor growth control. These data support continued clinical evaluation of INCB086550 as an alternative to antibody-based therapies. SIGNIFICANCE: We have identified a potent small-molecule inhibitor of PD-L1, INCB086550, which has biological properties similar to PD-L1/PD-1 monoclonal antibodies and may represent an alternative to antibody therapy. Preliminary clinical data in patients demonstrated increased immune activation and tumor growth control, which support continued clinical evaluation of this approach. See related commentary by Capparelli and Aplin, p. 1413. This article is highlighted in the In This Issue feature, p. 1397.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1
4.
Bioorg Med Chem Lett ; 55: 128452, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34780900

RESUMEN

Activin receptor-like kinase 2 (ALK2) has been implicated as a key target in multiple rare diseases. Herein, we describe the design of a novel bicyclic lactam series of potent and selective ALK2 inhibitors. This manuscript details an improvement in potency of two orders of magnitude from the initial bicyclic structure as well as a two-fold improvement in cellular potency from the original monocyclic inhibitor. Furthermore, we provide a detailed strategy for progressing this project in the future.


Asunto(s)
Receptores de Activinas Tipo I/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , beta-Lactamas/farmacología , Receptores de Activinas Tipo I/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , beta-Lactamas/síntesis química , beta-Lactamas/química
5.
J Am Chem Soc ; 141(20): 8064-8067, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31034218

RESUMEN

Transition state stabilization is essential for rate acceleration of enzymatic reactions. Despite extensive studies on various transition state structures of enzymes, an intriguing puzzle is whether an enzyme can accommodate multiple transition states (TSs) to catalyze a chemical reaction. It is experimentally challenging to study this proposition in terms of the choices of suitable enzymes and the feasibility to distinguish multiple TSs. As a paradigm with the protein lysine methyltransferase (PKMT) SET7/9 paired with its physiological substrates H3 and p53, their TSs were solved with experimental kinetic isotope effects as computational constraints. Remarkably, SET7/9 adopts two structurally distinct TSs, a nearly symmetric SN2 and an extremely early SN2, for H3K4 and p53K372 methylation, respectively. The two TSs are also different from those previously revealed for other PKMTs. The setting of multiple TSs is expected to be essential for SET7/9 and likely other PKMTs to act on broad substrates with high efficiency.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , S-Adenosilmetionina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Catálisis , N-Metiltransferasa de Histona-Lisina/química , Histonas/química , Humanos , Cinética , Lisina/química , Lisina/metabolismo , Metilación , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional , S-Adenosilmetionina/química , Proteína p53 Supresora de Tumor/química
6.
Proc Natl Acad Sci U S A ; 113(52): E8369-E8378, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27940912

RESUMEN

Protein lysine methyltransferases (PKMTs) catalyze the methylation of protein substrates, and their dysregulation has been linked to many diseases, including cancer. Accumulated evidence suggests that the reaction path of PKMT-catalyzed methylation consists of the formation of a cofactor(cosubstrate)-PKMT-substrate complex, lysine deprotonation through dynamic water channels, and a nucleophilic substitution (SN2) transition state for transmethylation. However, the molecular characters of the proposed process remain to be elucidated experimentally. Here we developed a matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) method and corresponding mathematic matrix to determine precisely the ratios of isotopically methylated peptides. This approach may be generally applicable for examining the kinetic isotope effects (KIEs) of posttranslational modifying enzymes. Protein lysine methyltransferase SET8 is the sole PKMT to monomethylate histone 4 lysine 20 (H4K20) and its function has been implicated in normal cell cycle progression and cancer metastasis. We therefore implemented the MS-based method to measure KIEs and binding isotope effects (BIEs) of the cofactor S-adenosyl-l-methionine (SAM) for SET8-catalyzed H4K20 monomethylation. A primary intrinsic 13C KIE of 1.04, an inverse intrinsic α-secondary CD3 KIE of 0.90, and a small but statistically significant inverse CD3 BIE of 0.96, in combination with computational modeling, revealed that SET8-catalyzed methylation proceeds through an early, asymmetrical SN2 transition state with the C-N and C-S distances of 2.35-2.40 Å and 2.00-2.05 Å, respectively. This transition state is further supported by the KIEs, BIEs, and steady-state kinetics with the SAM analog Se-adenosyl-l-selenomethionine (SeAM) as a cofactor surrogate. The distinct transition states between protein methyltransferases present the opportunity to design selective transition-state analog inhibitors.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/química , Isótopos/química , Unión Competitiva , Catálisis , Ciclo Celular , Simulación por Computador , Histonas/química , Humanos , Cinética , Lisina/química , Metilación , Modelos Moleculares , Modelos Teóricos , Metástasis de la Neoplasia , Péptidos/química , Estructura Secundaria de Proteína , S-Adenosilmetionina/química , Programas Informáticos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato
7.
Chem Sci ; 7(9): 5945-5954, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27547299

RESUMEN

Drug-target kinetics enable time-dependent changes in target engagement to be quantified as a function of drug concentration. When coupled to drug pharmacokinetics (PK), drug-target kinetics can thus be used to predict in vivo pharmacodynamics (PD). Previously we described a mechanistic PK/PD model that successfully predicted the antibacterial activity of an LpxC inhibitor in a model of Pseudomonas aeruginosa infection. In the present work we demonstrate that the same approach can be used to predict the in vivo activity of an enoyl-ACP reductase (FabI) inhibitor in a model of methicillin-resistant Staphylococcus aureus (MRSA) infection. This is significant because the LpxC inhibitors are cidal, whereas the FabI inhibitors are static. In addition P. aeruginosa is a Gram-negative organism whereas MRSA is Gram-positive. Thus this study supports the general applicability of our modeling approach across antibacterial space.

8.
Proc Natl Acad Sci U S A ; 113(31): E4523-30, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27432954

RESUMEN

The rising incidence of antimicrobial resistance (AMR) makes it imperative to understand the underlying mechanisms. Mycobacterium tuberculosis (Mtb) is the single leading cause of death from a bacterial pathogen and estimated to be the leading cause of death from AMR. A pyrido-benzimidazole, 14, was reported to have potent bactericidal activity against Mtb. Here, we isolated multiple Mtb clones resistant to 14. Each had mutations in the putative DNA-binding and dimerization domains of rv2887, a gene encoding a transcriptional repressor of the MarR family. The mutations in Rv2887 led to markedly increased expression of rv0560c. We characterized Rv0560c as an S-adenosyl-L-methionine-dependent methyltransferase that N-methylates 14, abolishing its mycobactericidal activity. An Mtb strain lacking rv0560c became resistant to 14 by mutating decaprenylphosphoryl-ß-d-ribose 2-oxidase (DprE1), an essential enzyme in arabinogalactan synthesis; 14 proved to be a nanomolar inhibitor of DprE1, and methylation of 14 by Rv0560c abrogated this activity. Thus, 14 joins a growing list of DprE1 inhibitors that are potently mycobactericidal. Bacterial methylation of an antibacterial agent, 14, catalyzed by Rv0560c of Mtb, is a previously unreported mechanism of AMR.


Asunto(s)
Antituberculosos/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Mycobacterium tuberculosis/metabolismo , Antituberculosos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bencimidazoles/química , Bencimidazoles/metabolismo , Regulación Bacteriana de la Expresión Génica , Metilación , Metiltransferasas/química , Metiltransferasas/genética , Metiltransferasas/metabolismo , Modelos Moleculares , Estructura Molecular , Mutación , Mycobacterium tuberculosis/genética , Dominios Proteicos , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , S-Adenosilmetionina/metabolismo
9.
J Med Chem ; 59(11): 5377-90, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27187871

RESUMEN

ß-Ketoacyl-ACP synthases (KAS) are key enzymes involved in the type II bacterial fatty acid biosynthesis (FASII) pathway and are putative targets for antibacterial discovery. Several natural product KAS inhibitors have previously been reported, including thiolactomycin (TLM), which is produced by Nocardia spp. Here we describe the synthesis and characterization of optically pure 5R-thiolactomycin (TLM) analogues that show improved whole cell activity against bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA) and priority pathogens such as Francisella tularensis and Burkholderia pseudomallei. In addition, we identify TLM analogues with in vivo efficacy against MRSA and Klebsiella pneumoniae in animal models of infection.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/antagonistas & inhibidores , Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Animales , Antibacterianos/síntesis química , Antibacterianos/química , Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/enzimología , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Francisella tularensis/efectos de los fármacos , Francisella tularensis/enzimología , Humanos , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/enzimología , Ratones , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/química , Tiofenos/farmacología , Yersinia pestis/efectos de los fármacos , Yersinia pestis/enzimología
10.
J Enzyme Inhib Med Chem ; 31(5): 695-703, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26118420

RESUMEN

Resveratrol is a natural polyphenol with plethora of biological activities. Resveratrol has previously shown to decrease DNA-methyltransferase (DNMT) enzymes expression and to reactivate silenced tumor suppressor genes. Currently, it seems that no resveratrol analogs have been developed as DNMT inhibitors. Recently, we reported the synthesis of resveratrol-salicylate derivatives and by examining the chemical structure of these analogs, we proposed that these compounds could exhibit DNMT inhibition especially that they resembled NSC 14778, a compound we previously identified as a DNMT inhibitor by virtual screening. Indeed, using in vitro DNMT inhibition assay, some of the resveratrol-salicylate analogs we screened in this work that showed selective inhibition against DNMT3 enzymes which were greater than resveratrol. A molecular docking study revealed key binding interactions with DNMT3A and DNMT3B enzymes. In addition, the most active analog, 10 showed considerable cytotoxicity against three human cancer cells; HT-29, HepG2 and SK-BR-3, which was greater than resveratrol. Further studies are needed to understand the anticancer mechanisms of these derivatives.


Asunto(s)
Antineoplásicos/farmacología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Salicilatos/farmacología , Estilbenos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Activación Enzimática/efectos de los fármacos , Células HT29 , Células Hep G2 , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Unión Proteica/efectos de los fármacos , Resveratrol , Salicilatos/química , Estilbenos/química , ADN Metiltransferasa 3B
11.
J Leukoc Biol ; 99(2): 387-98, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26497246

RESUMEN

Salmonellae are pathogenic bacteria that induce immunosuppression by mechanisms that remain largely unknown. Previously, we showed that a putative type II l-asparaginase produced by Salmonella Typhimurium inhibits T cell responses and mediates virulence in a murine model of infection. Here, we report that this putative L-asparaginase exhibits L-asparagine hydrolase activity required for Salmonella Typhimurium to inhibit T cells. We show that L-asparagine is a nutrient important for T cell activation and that L-asparagine deprivation, such as that mediated by the Salmonella Typhimurium L-asparaginase, causes suppression of activation-induced mammalian target of rapamycin signaling, autophagy, Myc expression, and L-lactate secretion. We also show that L-asparagine deprivation mediated by the Salmonella Typhimurium L-asparaginase causes suppression of cellular processes and pathways involved in protein synthesis, metabolism, and immune response. Our results advance knowledge of a mechanism used by Salmonella Typhimurium to inhibit T cell responses and mediate virulence, and provide new insights into the prerequisites of T cell activation. We propose a model in which l-asparagine deprivation inhibits T cell exit from quiescence by causing suppression of activation-induced metabolic reprogramming.


Asunto(s)
Asparaginasa/fisiología , Asparagina/fisiología , Proteínas Bacterianas/fisiología , Evasión Inmune/fisiología , Salmonella typhimurium/enzimología , Subgrupos de Linfocitos T/metabolismo , Secuencia de Aminoácidos , Animales , Asparaginasa/genética , Asparaginasa/farmacología , Asparagina/deficiencia , Asparagina/farmacología , Autofagia/efectos de los fármacos , Proteínas Bacterianas/genética , Células Cultivadas , Femenino , Genes myc , Evasión Inmune/genética , Interleucina-2/biosíntesis , Interleucina-2/genética , Ácido Láctico/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Virulencia
12.
J Biol Chem ; 288(47): 34190-34204, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24108128

RESUMEN

The survival of Mycobacterium tuberculosis depends on mycolic acids, very long α-alkyl-ß-hydroxy fatty acids comprising 60-90 carbon atoms. However, despite considerable efforts, little is known about how enzymes involved in mycolic acid biosynthesis recognize and bind their hydrophobic fatty acyl substrates. The condensing enzyme KasA is pivotal for the synthesis of very long (C38-42) fatty acids, the precursors of mycolic acids. To probe the mechanism of substrate and inhibitor recognition by KasA, we determined the structure of this protein in complex with a mycobacterial phospholipid and with several thiolactomycin derivatives that were designed as substrate analogs. Our structures provide consecutive snapshots along the reaction coordinate for the enzyme-catalyzed reaction and support an induced fit mechanism in which a wide cavity is established through the concerted opening of three gatekeeping residues and several α-helices. The stepwise characterization of the binding process provides mechanistic insights into the induced fit recognition in this system and serves as an excellent foundation for the development of high affinity KasA inhibitors.


Asunto(s)
Antituberculosos/química , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/química , Ácido Graso Sintasas/química , Mycobacterium tuberculosis/enzimología , Ácidos Micólicos/química , Tuberculosis/enzimología , Antituberculosos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Ácido Graso Sintasas/antagonistas & inhibidores , Ácido Graso Sintasas/metabolismo , Ácidos Micólicos/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tuberculosis/tratamiento farmacológico
13.
J Biol Chem ; 288(9): 6045-52, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23306195

RESUMEN

Thiolactomycin (TLM) is a natural product inhibitor of KasA, the ß-ketoacyl synthase A from Mycobacterium tuberculosis. To improve the affinity of TLM for KasA, a series of TLM analogs have been synthesized based on interligand NOEs between TLM and a pantetheine analog when both are bound simultaneously to the enzyme. Kinetic binding data reveal that position 3 of the thiolactone ring is a suitable position for elaboration of the TLM scaffold, and the structure-activity relationship studies provide information on the molecular features that govern time-dependent inhibition in this enzyme system. These experiments also exemplify the utility of transient one-dimensional NOE spectroscopy for obtaining interligand NOEs compared with traditional steady state two-dimensional NOESY spectroscopy.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Mycobacterium tuberculosis/enzimología , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/genética , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Inhibidores Enzimáticos/síntesis química , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Unión Proteica , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/química
14.
J Biol Chem ; 285(9): 6161-9, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20018879

RESUMEN

Thiolactomycin (TLM), a natural product thiolactone antibiotic produced by species of Nocardia and Streptomyces, is an inhibitor of the beta-ketoacyl-acyl carrier protein synthase (KAS) enzymes in the bacterial fatty acid synthase pathway. Using enzyme kinetics and direct binding studies, TLM has been shown to bind preferentially to the acyl-enzyme intermediates of the KASI and KASII enzymes from Mycobacterium tuberculosis and Escherichia coli. These studies, which utilized acyl-enzyme mimics in which the active site cysteine was replaced by a glutamine, also revealed that TLM is a slow onset inhibitor of the KASI enzymes KasA and ecFabB but not of the KASII enzymes KasB and ecFabF. The differential affinity of TLM for the acyl-KAS enzymes is proposed to result from structural change involving the movement of helices alpha5 and alpha6 that prepare the enzyme to bind malonyl-AcpM or TLM and that is initiated by formation of hydrogen bonds between the acyl-enzyme thioester and the oxyanion hole. The finding that TLM is a slow onset inhibitor of ecFabB supports the proposal that the long residence time of TLM on the ecFabB homologues in Serratia marcescens and Klebsiella pneumonia is an important factor for the in vivo antibacterial activity of TLM against these two organisms despite the fact that the in vitro MIC values are only 100-200 microg/ml. The mechanistic data on the interaction of TLM with KasA will provide an important foundation for the rational development of high affinity KasA inhibitors based on the thiolactone skeleton.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/antagonistas & inhibidores , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Dominio Catalítico/genética , Inhibidores Enzimáticos/farmacología , Escherichia coli/enzimología , Cinética , Mycobacterium tuberculosis/enzimología , Unión Proteica , Conformación Proteica , Especificidad por Sustrato , Tiofenos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA