Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS One ; 15(3): e0229672, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32214362

RESUMEN

More than 170 types of human papilloma viruses (HPV) exist with many causing proliferative diseases linked to malignancy in indications such as cervical cancer and head and neck squamous cell carcinoma. Characterization of antibody levels toward HPV serology is challenging due to complex biology of oncoproteins, pre-existing titers to multiple HPV types, cross-reactivity, and low affinity, polyclonal responses. Using multiplex technology from MSD, we have developed an assay that simultaneously characterizes antibodies against E6 and E7 oncoproteins of HPV16 and 18, the primary drivers of HPV-associated oncogenesis. We fusion tagged our E6 and E7 proteins with MBP via two-step purification, spot-printed an optimized concentration of protein into wells of MSD 96-well plates, and assayed various cynomolgus monkey, human and HPV+ cervical cancer patient serum to validate the assay. The dynamic range of the assay covered 4-orders of magnitude and antibodies were detected in serum at a dilution up to 100,000-fold. The assay was very precise (n = 5 assay runs) with median CV of human serum samples ~ 5.3% and inter-run variability of 11.4%. The multiplex serology method has strong cross-reactivity between E6 oncoproteins from human serum samples as HPV18 E6 antigens neutralized 5 of 6 serum samples as strongly as HPV16 E6. Moderate concordance (Spearman's Rank = 0.775) was found between antibody responses against HPV16 E7 in the multiplex assay compared to standard ELISA serology methods. These results demonstrate the development of a high-throughput, multi-plex assay that requires lower sample quantity input with greater dynamic range to detect type-specific anti-HPV concentrations to E6 and E7 oncoproteins of HPV16 and 18.


Asunto(s)
Anticuerpos Antivirales/sangre , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 18/inmunología , Inmunoensayo/métodos , Inmunoglobulina G/sangre , Animales , Especificidad de Anticuerpos , Reacciones Cruzadas , Proteínas de Unión al ADN/inmunología , Técnicas Electroquímicas , Ensayo de Inmunoadsorción Enzimática , Femenino , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/estadística & datos numéricos , Humanos , Inmunoensayo/estadística & datos numéricos , Límite de Detección , Mediciones Luminiscentes/métodos , Mediciones Luminiscentes/estadística & datos numéricos , Macaca fascicularis , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/inmunología , Proteínas Represoras/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología
2.
Mol Pharm ; 17(2): 507-516, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841002

RESUMEN

Targeted strategies to deliver and retain drugs to kidneys are needed to improve drug accumulation and efficacy in a myriad of kidney diseases. These drug delivery systems show potential for improving the therapeutic windows of drugs acting in the kidney. Biodistribution of antibody-based therapeutics in vivo is governed by several factors including binding affinity, size, and valency. Investigations of how the biophysical and biochemical properties of biologics enable them to overcome biological barriers and reach kidneys are therefore of interest. Although renal accumulation of antibody fragments in cancer diagnostics and treatment has been observed, reports on effective delivery of antibody fragments to the kidneys remain scarce. Previously, we demonstrated that targeting plasmalemma vesicle-associated protein (PV1), a caveolae-associated protein, can promote accumulation of antibodies in both the lungs and the kidneys. Here, by fine-tuning the binding affinity of an antibody toward PV1, we observe that the anti-PV1 antibody with reduced binding affinity lost the capability for kidney targeting while retaining the lung targeting activity, suggesting that binding affinity is a critical factor for kidney targeting of the anti-PV1 antibody. We next use the antibody fragment F(ab')2 targeting PV1 to assess the dual effects of rapid kidney filtration and PV1 targeting on kidney-selective targeting. Ex vivo fluorescence imaging results demonstrated that after rapidly accumulating in kidneys at 4 h, PV1-targeted F(ab')2 was continually retained in the kidney at 24 h, whereas the isotype control F(ab')2 underwent urinary elimination with significantly reduced signaling in the kidney. Confocal imaging studies confirmed the localization of PV1-targeted F(ab')2 in the kidney. In addition, the monovalent antibody fragment (Fab-C4) lost the capability for kidney homing, indicating that the binding avidity of anti-PV1 F(ab')2 is important for kidney targeting. Our findings suggest that PV1-targeted F(ab')2 might be useful as a drug carrier for renal targeting and highlight the importance of affinity optimization for tissue targeting antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Caveolas/metabolismo , Portadores de Fármacos/farmacocinética , Fragmentos Fab de Inmunoglobulinas/inmunología , Riñón/efectos de los fármacos , Proteínas de la Membrana/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Afinidad de Anticuerpos , Portadores de Fármacos/administración & dosificación , Femenino , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Riñón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
3.
Bioconjug Chem ; 30(4): 1232-1243, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30912649

RESUMEN

Despite some clinical success with antibody-drug conjugates (ADCs) in patients with solid tumors and hematological malignancies, improvements in ADC design are still desirable due to the narrow therapeutic window of these compounds. Tumor-targeting antibody fragments have distinct advantages over monoclonal antibodies, including more rapid tumor accumulation and enhanced penetration, but are subject to rapid clearance. Half-life extension technologies such as PEGylation and albumin-binding domains (ABDs) have been widely used to improve the pharmacokinetics of many different types of biologics. PEGylation improves pharmacokinetics by increasing hydrodynamic size to reduce renal clearance, whereas ABDs extend half-life via FcRn-mediated recycling. In this study, we used an anti-oncofetal antigen 5T4 diabody conjugated with a highly potent cytotoxic pyrrolobenzodiazepine (PBD) warhead to assess and compare the effects of PEGylation and albumin binding on the in vivo efficacy of antibody fragment drug conjugates. Conjugation of 2× PEG20K to a diabody improved half-life from 40 min to 33 h, and an ABD-diabody fusion protein exhibited a half-life of 45 h in mice. In a xenograft model of breast cancer MDA-MB-436, the ABD-diabody-PBD showed greater tumor growth suppression and better tolerability than either PEG-diabody-PBD or diabody-PBD. These results suggest that the mechanism of half-life extension is an important consideration for designing cytotoxic antitumor agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Inmunoadsorción Enzimática , Femenino , Semivida , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Ratones , Ratones Desnudos , Polietilenglicoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Commun Biol ; 2: 92, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30854484

RESUMEN

Systemic administration of bio-therapeutics can result in only a fraction of drug reaching targeted tissues, with the majority of drug being distributed to tissues irrelevant to the drug's site of action. Targeted delivery to specific organs may allow for greater accumulation, better efficacy, and improved safety. We investigated how targeting plasmalemma vesicle-associated protein (PV1), a protein found in the endothelial caveolae of lungs and kidneys, can promote accumulation in these organs. Using ex vivo fluorescence imaging, we show that intravenously administered αPV1 antibodies localize to mouse lungs and kidneys. In a bleomycin-induced idiopathic pulmonary fibrosis (IPF) mouse model, αPV1 conjugated to Prostaglandin E2 (PGE2), a known anti-fibrotic agent, significantly reduced collagen content and fibrosis whereas a non-targeted PGE2 antibody conjugate failed to slow fibrosis progression. Our results demonstrate that PV1 targeting can be utilized to deliver therapeutics to lungs and this approach is potentially applicable for various lung diseases.


Asunto(s)
Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Animales , Biomarcadores , Bleomicina/efectos adversos , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones
5.
J Control Release ; 279: 126-135, 2018 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-29653224

RESUMEN

The accumulation, dissemination and clearance of monoclonal antibody-based therapeutics or imaging reagents targeting tumor associated antigens is governed by several factors including affinity, size, charge, and valency. Tumor targeting antibody fragments have distinct advantages over intact monoclonal antibodies such as enhanced penetration within the tumor and rapid accumulation but are subject to rapid clearance. Polyethylene glycol (PEG)-modified antibody fragments can provide a way to balance tumor penetration and accumulation with improved serum persistence. In this study, we use a diabody, the dimeric antibody fragment, targeting the 5T4 antigen to assess the impact of PEGs of distinct size and shape on tumor accumulation and pharmacokinetics (PK). We show that PEG-modified diabodies improved the PK of the parental diabody from a half-life of 40 min to over 40 h for the higher molecular weight PEG conjugated diabodies. This improvement correlates with the increasing hydrodynamic size of pegylated diabodies, and can serve as a better predictor of the PK behavior of pegylated molecules than molecular weight alone. Tumor uptake profiles determined by quantitative PET imaging differed significantly based on PEG size and shape with diabody-PEG5K showing peak accumulation early on, but with the larger diabody-PEG20K showing better sustained tumor uptake at later time points. In addition, we demonstrate that a diabody-PEG20K-B with a hydrodynamic radius (Rh) of 6 nm had superior tumor uptake than the larger diabody-PEG40K-B with Rh of 12 nm, indicating that beyond 6 nm, larger pegylated diabodies have a slower tumor uptake rate while having comparable clearance kinetics. Our data demonstrate that pegylated diabodies with Rh of ~6 nm have an optimal size and PK profile for tumor uptake. Understanding the impact of pegylation on PK and tumor uptake could facilitate the development of pegylated diabodies as therapeutics.


Asunto(s)
Sistemas de Liberación de Medicamentos , Fragmentos de Inmunoglobulinas/administración & dosificación , Neoplasias/metabolismo , Polietilenglicoles/química , Animales , Transporte Biológico , Línea Celular Tumoral , Femenino , Semivida , Humanos , Hidrodinámica , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/metabolismo , Ratones , Ratones Desnudos , Peso Molecular , Tomografía de Emisión de Positrones , Distribución Tisular
6.
J Biomol Screen ; 21(6): 608-19, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26969322

RESUMEN

The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection-mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.


Asunto(s)
Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento/métodos , FN-kappa B/antagonistas & inhibidores , Relación Estructura-Actividad , Ligandos , Espectrometría de Masas/métodos , FN-kappa B/química , Unión Proteica , Transducción de Señal/efectos de los fármacos , Factor 5 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 5 Asociado a Receptor de TNF/química , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/química
7.
MAbs ; 8(3): 501-12, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26852694

RESUMEN

The enormous diversity created by gene recombination and somatic hypermutation makes de novo protein sequencing of monoclonal antibodies a uniquely challenging problem. Modern mass spectrometry-based sequencing will rarely, if ever, provide a single unambiguous sequence for the variable domains. A more likely outcome is computation of an ensemble of highly similar sequences that can satisfy the experimental data. This outcome can result in the need for empirical testing of many candidate sequences, sometimes iteratively, to identity one which can replicate the activity of the parental antibody. Here we describe an improved approach to antibody protein sequencing by using phage display technology to generate a combinatorial library of sequences that satisfy the mass spectrometry data, and selecting for functional candidates that bind antigen. This approach was used to reverse engineer 2 commercially-obtained monoclonal antibodies against murine CD137. Proteomic data enabled us to assign the majority of the variable domain sequences, with the exception of 3-5% of the sequence located within or adjacent to complementarity-determining regions. To efficiently resolve the sequence in these regions, small phage-displayed libraries were generated and subjected to antigen binding selection. Following enrichment of antigen-binding clones, 2 clones were selected for each antibody and recombinantly expressed as antigen-binding fragments (Fabs). In both cases, the reverse-engineered Fabs exhibited identical antigen binding affinity, within error, as Fabs produced from the commercial IgGs. This combination of proteomic and protein engineering techniques provides a useful approach to simplifying the technically challenging process of reverse engineering monoclonal antibodies from protein material.


Asunto(s)
Biblioteca de Péptidos , Ingeniería de Proteínas/métodos , Análisis de Secuencia de Proteína , Anticuerpos de Cadena Única , Animales , Ratones , Ratas , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química
8.
J Med Chem ; 56(6): 2294-310, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23379595

RESUMEN

This report documents the first example of a specific inhibitor of protein kinases with preferential binding to the activated kinase conformation: 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one 11r (MK-8033), a dual c-Met/Ron inhibitor under investigation as a treatment for cancer. The design of 11r was based on the desire to reduce time-dependent inhibition of CYP3A4 (TDI) by members of this structural class. A novel two-step protocol for the synthesis of benzylic sulfonamides was developed to access 11r and analogues. We provide a rationale for the observed selectivity based on X-ray crystallographic evidence and discuss selectivity trends with additional examples. Importantly, 11r provides full inhibition of tumor growth in a c-Met amplified (GTL-16) subcutaneous tumor xenograft model and may have an advantage over inactive form kinase inhibitors due to equal potency against a panel of oncogenic activating mutations of c-Met in contrast to c-Met inhibitors without preferential binding to the active kinase conformation.


Asunto(s)
Benzocicloheptenos/metabolismo , Benzocicloheptenos/farmacología , Descubrimiento de Drogas , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Animales , Benzocicloheptenos/química , Línea Celular Tumoral , Perros , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/química , Ratas , Especificidad por Sustrato , Sulfonamidas/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Med Chem ; 54(12): 4092-108, 2011 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-21608528

RESUMEN

c-Met is a transmembrane tyrosine kinase that mediates activation of several signaling pathways implicated in aggressive cancer phenotypes. In recent years, research into this area has highlighted c-Met as an attractive cancer drug target, triggering a number of approaches to disrupt aberrant c-Met signaling. Screening efforts identified a unique class of 5H-benzo[4,5]cyclohepta[1,2-b]pyridin-5-one kinase inhibitors, exemplified by 1. Subsequent SAR studies led to the development of 81 (MK-2461), a potent inhibitor of c-Met that was efficacious in preclinical animal models of tumor suppression. In addition, biochemical studies and X-ray analysis have revealed that this unique class of kinase inhibitors binds preferentially to the activated (phosphorylated) form of the kinase. This report details the development of 81 and provides a description of its unique biochemical properties.


Asunto(s)
Antineoplásicos/síntesis química , Benzocicloheptenos/síntesis química , Piridinas/síntesis química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzocicloheptenos/farmacocinética , Benzocicloheptenos/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Perros , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Haplorrinos , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Mutación , Trasplante de Neoplasias , Fosforilación , Unión Proteica , Pirazoles/síntesis química , Pirazoles/farmacocinética , Pirazoles/farmacología , Piridinas/farmacocinética , Piridinas/farmacología , Ratas , Proteínas Tirosina Quinasas Receptoras/genética , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Trasplante Heterólogo
10.
J Biol Chem ; 286(13): 11218-25, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21247903

RESUMEN

The receptor tyrosine kinase c-Met is implicated in oncogenesis and is the target for several small molecule and biologic agents in clinical trials for the treatment of cancer. Binding of the hepatocyte growth factor to the cell surface receptor of c-Met induces activation via autophosphorylation of the kinase domain. Here we describe the structural basis of c-Met activation upon autophosphorylation and the selective small molecule inhibiton of autophosphorylated c-Met. MK-2461 is a potent c-Met inhibitor that is selective for the phosphorylated state of the enzyme. Compound 1 is an MK-2461 analog with a 20-fold enthalpy-driven preference for the autophosphorylated over unphosphorylated c-Met kinase domain. The crystal structure of the unbound kinase domain phosphorylated at Tyr-1234 and Tyr-1235 shows that activation loop phosphorylation leads to the ejection and disorder of the activation loop and rearrangement of helix αC and the G loop to generate a viable active site. Helix αC adopts a orientation different from that seen in activation loop mutants. The crystal structure of the complex formed by the autophosphorylated c-Met kinase domain and compound 1 reveals a significant induced fit conformational change of the G loop and ordering of the activation loop, explaining the selectivity of compound 1 for the autophosphorylated state. The results highlight the role of structural plasticity within the kinase domain in imparting the specificity of ligand binding and provide the framework for structure-guided design of activated c-Met inhibitors.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Animales , Línea Celular , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Spodoptera , Relación Estructura-Actividad , Tirosina Quinasa c-Mer
11.
Arch Biochem Biophys ; 469(2): 220-31, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17999913

RESUMEN

The kinesin spindle protein (KSP, also known as Eg5) is essential for the proper separation of spindle poles during mitosis, and inhibition results in mitotic arrest and the formation of characteristic monoaster spindles. Several distinct classes of KSP inhibitors have been described previously in the public and patent literature. However, most appear to share a common induced-fit allosteric binding site, suggesting a common mechanism of inhibition. In a high-throughput screen for inhibitors of KSP, a novel class of thiazole-containing inhibitors was identified. Unlike the previously described allosteric KSP inhibitors, the thiazoles described here show ATP competitive kinetic behavior, consistent with binding within the nucleotide binding pocket. Although they bind to a pocket that is highly conserved across kinesins, these molecules exhibit significant selectivity for KSP over other kinesins and other ATP-utilizing enzymes. Several of these compounds are active in cells and produce a phenotype similar to that observed with previously published allosteric inhibitors of KSP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Bioquímica/métodos , Cinesinas/antagonistas & inhibidores , Cinesinas/química , Mitosis , Adenosina Trifosfato/química , Sitio Alostérico , Unión Competitiva , Diseño de Fármacos , Humanos , Cinesinas/metabolismo , Modelos Biológicos , Modelos Químicos , Nucleótidos/química , Fenotipo , Unión Proteica , Tiazoles/farmacología
12.
J Med Chem ; 47(25): 6363-72, 2004 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-15566305

RESUMEN

A series of N-(1,3-thiazol-2-yl)pyridin-2-amine KDR kinase inhibitors have been developed that possess optimal properties. Compounds have been discovered that exhibit excellent in vivo potency. The particular challenges of overcoming hERG binding activity and QTc increases in vivo in addition to achieving good pharmacokinetics have been acomplished by discovering a unique class of amine substituents. These compounds have a favorable kinase selectivity profile that can be accentuated with appropriate substitution.


Asunto(s)
Aminopiridinas/síntesis química , Canales de Potasio con Entrada de Voltaje/metabolismo , Piridinas/síntesis química , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Tiazoles/síntesis química , Administración Oral , Aminopiridinas/farmacocinética , Aminopiridinas/farmacología , Animales , Disponibilidad Biológica , Línea Celular , Perros , Canal de Potasio ERG1 , Electrocardiografía/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go , Técnicas In Vitro , Pulmón/enzimología , Macaca mulatta , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Fosforilación , Piridinas/farmacocinética , Piridinas/farmacología , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Tiazoles/farmacocinética , Tiazoles/farmacología , Distribución Tisular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA