Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 867457, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36120380

RESUMEN

Disrupted tau proteostasis and transneuronal spread is a pathological hallmark of Alzheimer's disease. Neurodegenerative diseases remain an unmet medical need and novel disease modifying therapeutics are paramount. Our objective was to develop a mechanistic mathematical model to enhance our understanding of tau antibody pharmacokinetics and pharmacodynamics in animals and humans. A physiologically-based pharmacokinetic-pharmacodynamic (PBPK-PD) modeling approach was employed to support the preclinical development and clinical translation of therapeutic antibodies targeting tau for the treatment of Alzheimer's disease. The pharmacokinetics of a tau antibody was evaluated in rat and non-human primate microdialysis studies. Model validation for humans was performed using publicly available clinical data for gosuranemab. In-silico analyses were performed to predict tau engagement in human brain for a range of tau antibody affinities and various dosing regimens. PBPK-PD modeling enabled a quantitative understanding for the relationship between dose, affinity, and target engagement, which supported lead candidate optimization and predictions of clinically efficacious dosing regimens.

2.
Mol Imaging Biol ; 23(2): 250-259, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33104972

RESUMEN

PURPOSE: Programmed cell death-1 receptor (PD-1) and its ligand (PD-L1) are the targets for immunotherapy in many cancer types. Although PD-1 blockade has therapeutic effects, the efficacy differs between patients. Factors contributing to this variability are PD-L1 expression levels and immune cells present in tumors. However, it is not well understood how PD-1 expression in the tumor microenvironment impacts immunotherapy response. Thus, imaging of PD-1-expressing immune cells is of interest. This study aims to evaluate the biodistribution of Zirconium-89 (89Zr)-labeled pembrolizumab, a humanized IgG4 kappa monoclonal antibody targeting PD-1, in healthy cynomolgus monkeys as a translational model of tracking PD-1-positive immune cells. PROCEDURES: Pembrolizumab was conjugated with the tetrafluorophenol-N-succinyl desferal-Fe(III) ester (TFP-N-sucDf) and subsequently radiolabeled with 89Zr. Four cynomolgus monkeys with no previous exposure to humanized monoclonal antibodies received tracer only or tracer co-injected with pembrolizumab intravenously over 5 min. Thereafter, a static whole-body positron emission tomography (PET) scan was acquired with 10 min per bed position on days 0, 2, 5, and 7. Image-derived standardized uptake values (SUVmean) were quantified by region of interest (ROI) analysis. RESULTS: 89Zr-N-sucDf-pembrolizumab was synthesized with high radiochemical purity (> 99 %) and acceptable molar activity (> 7 MBq/nmol). In animals dosed with tracer only, 89Zr-N-sucDf-pembrolizumab distribution in lymphoid tissues such as mesenteric lymph nodes, spleen, and tonsils increased over time. Except for the liver, low radiotracer distribution was observed in all non-lymphoid tissue including the lung, muscle, brain, heart, and kidney. When a large excess of pembrolizumab was co-administered with a radiotracer, accumulation in the lymph nodes, spleen, and tonsils was reduced, suggestive of target-mediated accumulation. CONCLUSIONS: 89Zr-N-sucDf-pembrolizumab shows preferential uptake in the lymphoid tissues including the lymph nodes, spleen, and tonsils. 89Zr-N-sucDf-pembrolizumab may be useful in tracking the distribution of a subset of immune cells in non-human primates and humans. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02760225.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Imagen Molecular/métodos , Neoplasias/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/farmacocinética , Femenino , Inmunoterapia/métodos , Macaca fascicularis , Masculino , Modelos Animales , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Radioisótopos , Distribución Tisular , Circonio
3.
AAPS J ; 21(3): 39, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30868312

RESUMEN

Assessment of the factors that regulate antibody exposure-response relationships in the relevant animal models is critical for the design of successful translational strategies from discovery to the clinic. Depending on the specific clinical indication, preclinical development paradigms may require that the efficacy or dosing-related attributes for the existing antibody be assessed in various species when cross-reactivity of the lead antibody to the intended species is justified. Additionally, with the success of monoclonal antibodies for management of various human conditions, a parallel interest in therapeutic use of these novel modalities in various veterinary species has followed. The protective role of neonatal Fc receptor (FcRn) in regulation of IgG homeostasis and clearance is now well recognized and the "nonspecific clearance" of antibodies through bone marrow-derived phagocytic and vascular endothelial cells (via lysosomal processes) is modulated by interactions with FcRn receptors. In this study, we have attempted to examine the PK properties of human IgG antibodies in dog and monkey. These studies establish a translational framework for evaluation of IgG antibody PK properties across species.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Inmunoglobulina G/farmacología , Administración Intravenosa , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Perros , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Macaca fascicularis , Macaca mulatta , Ratones , Modelos Animales , Virus Sincitiales Respiratorios/inmunología , Especificidad de la Especie
4.
Clin Chem ; 62(1): 227-35, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26430077

RESUMEN

BACKGROUND: Proglucagon-derived peptides (PGDPs), which include glucagon-like peptide (GLP)-1, glucagon, and oxyntomodulin, are key regulators of glucose homeostasis and satiety. These peptide hormones are typically measured with immuno-based assays (e.g., ELISA, RIA), which often suffer from issues of selectivity. METHODS: We developed a multiplexed assay for measuring PGDPs including GLP-1 (7-36) amide, GLP-1 (9-36) amide, glucagon, and oxyntomodulin by mass spectrometry and used this assay to examine the effect of a meal tolerance test on circulating concentrations of these hormones. Participants fasted overnight and were either given a meal (n = 8) or continued to fast (n = 4), with multiple blood collections over the course of 3 h. Plasma samples were analyzed by microflow immunoaffinity (IA)-LC-MS/MS with an isotope dilution strategy. RESULTS: Assay performance characteristics were examined and established during analytical validation for all peptides. Intra- and interassay imprecision were found to be 2.2%-10.7% and 6.8%-22.5%, respectively. Spike recovery was >76%, and dilution linearity was established up to a 16-fold dilution. Immediately after the meal tolerance test, GLP-1 and oxyntomodulin concentrations increased and had an almost identical temporal relationship, and glucagon concentrations increased with a slight delay. CONCLUSIONS: IA-LC-MS/MS was used for the simultaneous and selective measurement of PGDPs. This work includes the first indication of the physiological concentrations and modulation of oxyntomodulin after a meal.


Asunto(s)
Ayuno , Péptido 1 Similar al Glucagón/sangre , Glucagón/sangre , Inmunoensayo , Oxintomodulina/sangre , Espectrometría de Masas en Tándem , Animales , Cromatografía Líquida de Alta Presión , Glucagón/inmunología , Péptido 1 Similar al Glucagón/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Oxintomodulina/inmunología
5.
Immunity ; 43(4): 727-38, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26431948

RESUMEN

Whether interleukin-17A (IL-17A) has pathogenic and/or protective roles in the gut mucosa is controversial and few studies have analyzed specific cell populations for protective functions within the inflamed colonic tissue. Here we have provided evidence for IL-17A-dependent regulation of the tight junction protein occludin during epithelial injury that limits excessive permeability and maintains barrier integrity. Analysis of epithelial cells showed that in the absence of signaling via the IL-17 receptor adaptor protein Act-1, the protective effect of IL-17A was abrogated and inflammation was enhanced. We have demonstrated that after acute intestinal injury, IL-23R(+) γδ T cells in the colonic lamina propria were the primary producers of early, gut-protective IL-17A, and this production of IL-17A was IL-23 independent, leaving protective IL-17 intact in the absence of IL-23. These results suggest that IL-17-producing γδ T cells are important for the maintenance and protection of epithelial barriers in the intestinal mucosa.


Asunto(s)
Colitis/fisiopatología , Interleucina-17/fisiología , Interleucina-23/fisiología , Mucosa Intestinal/fisiopatología , Enfermedad Aguda , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Línea Celular Tumoral , Polaridad Celular , Colitis/inducido químicamente , Neoplasias del Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Epitelio/fisiopatología , Proteínas de Homeodominio/fisiología , Humanos , Interleucina-17/deficiencia , Interleucina-17/farmacología , Subgrupos Linfocitarios/metabolismo , Ratones , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Ocludina/metabolismo , Permeabilidad , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Proteínas Recombinantes/farmacología , Uniones Estrechas/fisiología , Factor de Necrosis Tumoral alfa/farmacología
6.
Cancer Cell ; 20(6): 781-96, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22172723

RESUMEN

Tumor immune surveillance and cancer immunotherapies are thought to depend on the intratumoral infiltration of activated CD8(+) T cells. Intratumoral CD8(+) T cells are rare and lack activity. IL-10 is thought to contribute to the underlying immune suppressive microenvironment. Defying those expectations we demonstrate that IL-10 induces several essential mechanisms for effective antitumor immune surveillance: infiltration and activation of intratumoral tumor-specific cytotoxic CD8(+) T cells, expression of the Th1 cytokine interferon-γ (IFNγ) and granzymes in CD8(+) T cells, and intratumoral antigen presentation molecules. Consequently, tumor immune surveillance is weakened in mice deficient for IL-10 whereas transgenic overexpression of IL-10 protects mice from carcinogenesis. Treatment with pegylated IL-10 restores tumor-specific intratumoral CD8(+) T cell function and controls tumor growth.


Asunto(s)
Interferón gamma/metabolismo , Interleucina-10/metabolismo , Neoplasias Experimentales/inmunología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Granzimas/metabolismo , Humanos , Interferón gamma/genética , Interleucina-10/genética , Interleucina-10/inmunología , Subunidad alfa del Receptor de Interleucina-10/genética , Subunidad alfa del Receptor de Interleucina-10/metabolismo , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Perforina/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Bazo/metabolismo , Trasplante Heterólogo , Carga Tumoral , Escape del Tumor
7.
Autoimmunity ; 43(8): 642-53, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20380588

RESUMEN

Bone erosion is a clinical endpoint for various diseases including rheumatoid arthritis. In this paper, we used rodent arthritis models with severe bone erosion to examine the structural, cellular, and molecular aspects of the inflammation-driven bone resorption process. Our data show that bone loss is observed only in chronically, severely inflamed joints. The most severely affected anatomic sites were the metatarsal phalangeal joint and tarsal bones of the paw. The magnitude of the inflammation-driven bone erosion was dependent on both the duration of inflammatory response and the severity of the joint swelling response. The application of micro-computed tomography well demonstrated the therapeutic benefit of anti-IL-17A in protection of bones from erosion. Alterations in the cellular profile of the joint occurred prior to any major structural deterioration of the bone. Receptor activator for nuclear factor κB ligand, a potent inducer of osteoclast differentiation and bone resorption, was elevated in animals coincident with severe arthritis initiation. The experimental approaches and concepts outlined in this paper provide a valuable process to evaluate and quantify therapies that modulate rodent arthritis-associated bone-erosion models.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Resorción Ósea/inmunología , Interleucina-17/farmacología , Animales , Artritis Experimental/fisiopatología , Artritis Reumatoide/fisiopatología , Biomarcadores/sangre , Resorción Ósea/fisiopatología , Pie/fisiopatología , Histocitoquímica , Interleucina-17/uso terapéutico , Masculino , Ratones , Ligando RANK/sangre , Ratas , Tomografía Computarizada por Rayos X
8.
J Biotechnol ; 119(3): 219-44, 2005 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16005536

RESUMEN

Successful drug discovery requires accurate decision making in order to advance the best candidates from initial lead identification to final approval. Chemogenomics, the use of genomic tools in pharmacology and toxicology, offers a promising enhancement to traditional methods of target identification/validation, lead identification, efficacy evaluation, and toxicity assessment. To realize the value of chemogenomics information, a contextual database is needed to relate the physiological outcomes induced by diverse compounds to the gene expression patterns measured in the same animals. Massively parallel gene expression characterization coupled with traditional assessments of drug candidates provides additional, important mechanistic information, and therefore a means to increase the accuracy of critical decisions. A large-scale chemogenomics database developed from in vivo treated rats provides the context and supporting data to enhance and accelerate accurate interpretation of mechanisms of toxicity and pharmacology of chemicals and drugs. To date, approximately 600 different compounds, including more than 400 FDA approved drugs, 60 drugs approved in Europe and Japan, 25 withdrawn drugs, and 100 toxicants, have been profiled in up to 7 different tissues of rats (representing over 3200 different drug-dose-time-tissue combinations). Accomplishing this task required evaluating and improving a number of in vivo and microarray protocols, including over 80 rigorous quality control steps. The utility of pairing clinical pathology assessments with gene expression data is illustrated using three anti-neoplastic drugs: carmustine, methotrexate, and thioguanine, which had similar effects on the blood compartment, but diverse effects on hepatotoxicity. We will demonstrate that gene expression events monitored in the liver can be used to predict pathological events occurring in that tissue as well as in hematopoietic tissues.


Asunto(s)
Biotecnología/métodos , Diseño de Fármacos , Industria Farmacéutica/métodos , 5-Aminolevulinato Sintetasa/biosíntesis , Animales , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Automatización , Conductos Biliares/patología , Carmustina/toxicidad , Biología Computacional , Bases de Datos como Asunto , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Expresión Génica , Humanos , Hiperplasia/etiología , Hígado/efectos de los fármacos , Masculino , Metotrexato/toxicidad , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de los Órganos , Farmacología/métodos , ARN/química , ARN Complementario/metabolismo , Ratas , Ratas Sprague-Dawley , Reticulocitos/citología , Reticulocitos/metabolismo , Tioguanina/toxicidad , Factores de Tiempo , Distribución Tisular , Toxicología/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA