Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Nucleic Acid Ther ; 30(5): 265-275, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32833564

RESUMEN

Inotersen (TEGSEDI™) is a 2'-O-(2-methoxyethyl)-modified antisense oligonucleotide, intended for treating hereditary transthyretin (TTR) amyloidosis with polyneuropathy. The potential immunogenicity (IM) response to inotersen was evaluated in chronic nonclinical safety studies and the pivotal phase 2/3 clinical study. The evaluation was designed to assess the characteristics of antidrug antibodies (ADAs) and their effects on the pharmacokinetics, pharmacodynamics, clinical efficacy, and safety in animals and humans. No immunogenic response was observed after long-term treatment with inotersen in mice. In monkeys, the incidence rate of IM to inotersen appeared to be dose dependent, with 28.6%-50.0% of animals developing ADAs after 36 weeks of treatment. This was characterized as late onset (median onset of 185 days) with low titers (median titer of 8, or 400 if minimum required dilution of 50 is included). The overall incidence rate of patients who developed ADAs was 30% after 65 weeks of treatment with median onset of 203 days and median peak titer of 300. IM had minimal effect on plasma peak (Cmax) and total exposure (i.e. area under curve, AUC) of inotersen, but showed elevated plasma trough levels in both IM-positive animals and humans. However, ADAs had no effect on tissue exposure, TTR messenger RNA, or plasma TTR levels in the long-term monkey study. Similarly, IM showed no effect on plasma TTR levels in clinical studies. Thus, ADAs antibodies were binding antibodies, but not neutralizing antibodies. Finally, no association was observed between IM and toxicity findings (eg, platelet, complement activation, and histopathology findings) in the inotersen 9-month monkey study. In humans, no difference was observed in hematology, including platelets, kidney function tests, or incidence of adverse events between IM-positive and -negative patients. Overall, IM showed no effect on toxicity or safety of inotersen evaluated in both monkeys and humans. ClinicalTrials.gov Identifier: NCT01737398.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/tratamiento farmacológico , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos/administración & dosificación , Oligorribonucleótidos/administración & dosificación , Prealbúmina/genética , Animales , Anticuerpos Antiidiotipos/sangre , Anticuerpos Antiidiotipos/inmunología , Plaquetas/inmunología , Enfermedad de Charcot-Marie-Tooth/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Haplorrinos , Humanos , Inmunogenicidad Vacunal/genética , Inmunogenicidad Vacunal/inmunología , Pruebas de Función Renal , Masculino , Ratones , Oligonucleótidos/efectos adversos , Oligonucleótidos Antisentido/efectos adversos , Oligonucleótidos Antisentido/sangre , Oligonucleótidos Antisentido/farmacocinética , Oligorribonucleótidos/efectos adversos , Oligorribonucleótidos/sangre , Oligorribonucleótidos/farmacocinética , Prealbúmina/antagonistas & inhibidores , Prealbúmina/inmunología
2.
Nucleic Acid Ther ; 29(5): 266-277, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31368839

RESUMEN

Although antisense oligonucleotides (ASOs) are well tolerated preclinically and in the clinic, some sequences of ASOs can trigger an inflammatory response leading to B cell and macrophage activation in rodents. This prompted our investigation into the contribution of genetic architecture to the ASO-mediated inflammatory response. Genome-wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the acute and delayed inflammatory response to a single 75 mg/kg dose of an inflammatory 2'-O-methoxyethyl (2'MOE) modified ASO. The acute response was measured 6 h after ASO administration, via evaluation for increased plasma production of interleukin 6 (IL6), IL10, monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein-1ß (MIP-1ß). Delayed inflammation was evaluated by spleen weight increases after 96 h. We identified single nucleotide polymorphisms (SNPs) on chromosomes 16 and 17 associated with plasma MIP-1ß, IL6, and MCP-1 levels, and one on chromosome 8 associated with increases in spleen weight. Systems genetic analysis utilizing transcriptomic data from HMDP strain macrophages determined that the acute inflammatory SNPs were expression quantitative trait locis (eQTLs) for CCAAT/enhancer-binding protein beta (Cebpb) and salt inducible kinase 1 (Sik1). The delayed inflammatory SNP was an eQTL for Rho guanine nucleotide exchange factor 10 (Arhgef10). In vitro assays in mouse primary cells and human cell lines have confirmed the HMDP finding that lower Sik1 expression increases the acute inflammatory response. Our results demonstrate the utility of using mouse GWA study (GWAS) and the HMDP for detecting genes modulating the inflammatory response to pro-inflammatory ASOs in a pharmacological setting.


Asunto(s)
Predisposición Genética a la Enfermedad , Inflamación/terapia , Oligonucleótidos Antisentido/farmacología , Transcriptoma/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL4/genética , Perfilación de la Expresión Génica/métodos , Estudio de Asociación del Genoma Completo/métodos , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética
3.
Curr Protoc Toxicol ; 80(1): e74, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30982234

RESUMEN

Macrophages are innate immune cells that play important roles in various physiological and pathological processes. Evaluation of pro-inflammatory effects of drugs on macrophages has become commonplace in preclinical drug development prior to human clinical trials. Despite their body-wide distribution, tissue macrophages are often difficult to collect from large animals and humans in a noninvasive manner. Therefore, in vitro-differentiated macrophages are important tools to facilitate cross-species analysis of macrophage function. Although cynomolgus monkeys are an essential non-rodent species for preclinical research, in vitro differentiation of cynomolgus-monkey macrophages has been poorly characterized. In the present unit, we describe a protocol to differentiate cynomolgus-monkey macrophages from isolated bone marrow mononuclear cells (BMMCs). In contrast to monocytes, cynomolgus-monkey BMMCs show robust expansion in the presence of macrophage colony-stimulating factor in vitro, which allows expansion of many cells from a single animal donor. Macrophages differentiated from BMMCs retain many of the macrophage phenotypes and functions, including phagocytosis and cytokine release, and therefore can be used as a surrogate to assess effects of drugs on cynomolgus-monkey macrophages. © 2019 by John Wiley & Sons, Inc.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Macrófagos/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Evaluación de Medicamentos , Inmunofenotipificación , Macaca fascicularis , Macrófagos/inmunología
4.
Curr Protoc Toxicol ; 79(1): e68, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30673165

RESUMEN

Phagocytosis of platelets by monocytes and macrophages is a primary mechanism of platelet clearance in vivo and has been increasingly implicated in playing an important role in thrombocytopenia mediated by monoclonal antibodies intended for therapeutic purposes. In the present article, we describe an in vitro flow cytometry assay to assess the effect of antibody-mediated platelet phagocytosis by monocytes. Freshly isolated platelets were labeled with a fluorescent probe, 5-chloromethylfluorescein diacetate (CMFDA) and then co-cultured with isolated peripheral blood mononuclear cells (PBMCs) from the same donor in the presence of increasing concentrations of a monoclonal antibody drug. After incubation, an increase in CMFDA fluorescence intensity of CD14 positive monocytes was evaluated by flow cytometry as an assessment for drug-mediated platelet phagocytosis by monocytes. The assay has been evaluated using both human and cynomolgus monkey cells for the prediction of drug-induced thrombocytopenia. © 2019 by John Wiley & Sons, Inc.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Bioensayo/métodos , Plaquetas/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Trombocitopenia/inducido químicamente , Animales , Plaquetas/inmunología , Técnicas de Cocultivo , Citometría de Flujo , Fluoresceínas/química , Colorantes Fluorescentes/química , Humanos , Leucocitos Mononucleares/inmunología , Macaca fascicularis , Fagocitosis/inmunología , Valor Predictivo de las Pruebas , Trombocitopenia/inmunología
5.
PLoS One ; 12(1): e0169976, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28081568

RESUMEN

Systemic inflammation co-activates coagulation, which unchecked culminates in a lethal syndrome of multi-organ microvascular thrombosis known as disseminated intravascular coagulation (DIC). We studied an endotoxin-induced inflammatory state in rats to identify biomarkers of hemostatic imbalance favoring hypercoagulability. Intraperitoneal injection of LPS at 15 mg/kg body weight resulted in peripheral leukopenia and widespread neutrophilic sequestration characteristic of an acute systemic inflammatory response. Early indicators of hemostatic pathway activation developed within 4 hours, including increased circulating concentrations of procoagulant extracellular vesicles (EVs), EVs expressing endothelial cell and platelet membrane markers, and high concentration of soluble intercellular adhesion molecule-1 (sICAM-1), plasminogen activator inhibitor-1 (PAI-1), and D-dimers. Inflammation persisted throughout the 48-hour observation period; however, increases were found in a subset of serum microRNA (miRNA) that coincided with gradual resolution of hemostatic protein abnormalities and reduction in EV counts. Dose-adjusted LPS treatment in rats provides a time-course model to develop biomarker profiles reflecting procoagulant imbalance and rebalance under inflammatory conditions.


Asunto(s)
Lipopolisacáridos , Trombofilia/inducido químicamente , Trombofilia/fisiopatología , Enfermedad Aguda , Animales , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucopenia/inducido químicamente , Masculino , MicroARNs/sangre , Neutrófilos/metabolismo , Neutrófilos/patología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas , Ratas Wistar , Trombofilia/inmunología , Factores de Tiempo
6.
J Biol Chem ; 292(5): 1876-1883, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27994063

RESUMEN

The stable effector functionLess (SEFL) antibody was designed as an IgG1 antibody with a constant region that lacks the ability to interact with Fcγ receptors. The engineering and stability and pharmacokinetic assessments of the SEFL scaffold is described in the accompanying article (Jacobsen, F. W., Stevenson, R., Li, C., Salimi-Moosavi, H., Liu, L., Wen, J., Luo, Q., Daris, K., Buck, L., Miller, S., Ho, S-Y., Wang, W., Chen, Q., Walker, K., Wypych, J., Narhi, L., and Gunasekaran, K. (2017) J. Biol. Chem 292). The biological properties of these SEFL antibodies were assessed in a variety of human and cynomolgus monkey in vitro assays. Binding of parent molecules and their SEFL variants to human and cynomolgus monkey FcγRs were evaluated using flow cytometry-based binding assays. The SEFL variants tested showed decreased binding affinity to human and cynomolgus FcγRs compared with the wild-type IgG1 antibody. In addition, SEFL variants demonstrated no antibody-dependent cell-mediated cytotoxicity in vitro against Daudi cells with cynomolgus monkey peripheral blood mononuclear cells, and had minimal complement-dependent cytotoxicity activity similar to that of the negative control IgG2 in a CD20+ human Raji lymphoma cell line. SEFL mutations eliminated off-target antibody-dependent monocyte phagocytosis of cynomolgus monkey platelets, and cynomolgus platelet activation in vitro These experiments demonstrate that the SEFL modifications successfully eliminated Fc-associated effector binding and functions.


Asunto(s)
Anticuerpos Monoclonales , Plaquetas/inmunología , Inmunoglobulina G , Monocitos/inmunología , Fagocitosis/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Receptores de IgG , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Macaca fascicularis , Ratones , Fagocitosis/inmunología , Activación Plaquetaria/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología
7.
Cytometry A ; 89(2): 196-206, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26484737

RESUMEN

Extracellular vesicles (EVs) are attracting attention as vehicles for inter-cellular signaling that may have value as diagnostic or therapeutic targets. EVs are released by many cell types and by different mechanisms, resulting in phenotypic heterogeneity that makes them a challenge to study. Flow cytometry is a popular tool for characterizing heterogeneous mixtures of particles such as cell types within blood, but the small size of EVs makes them difficult to measure using conventional flow cytometry. To address this limitation, a high sensitivity flow cytometer was constructed and EV measurement approaches that allowed them to enumerate and estimate the size of individual EVs, as well as measure the presence of surface markers to identify phenotypic subsets of EVs. Several fluorescent membrane probes were evaluated and it was found that the voltage sensing dye di-8-ANEPPS could produce vesicle fluorescence in proportion to vesicle surface area, allowing for accurate measurements of EV number and size. Fluorescence-labeled annexin V and anti-CD61 antibody was used to measure the abundance of these surface markers on EVs in rat plasma. It was shown that treatment of platelet rich plasma with calcium ionophore resulted in an increase in the fraction of annexin V and CD61-positive EVs. Vesicle flow cytometry using fluorescence-based detection of EVs has the potential to realize the potential of cell-derived membrane vesicles as functional biomarkers for a variety of applications.


Asunto(s)
Vesículas Extracelulares/fisiología , Citometría de Flujo/métodos , Animales , Calibración , Vesículas Extracelulares/química , Femenino , Citometría de Flujo/normas , Límite de Detección , Plasma Rico en Plaquetas/química , Ratas Sprague-Dawley , Coloración y Etiquetado
8.
Curr Protoc Cytom ; 73: 9.48.1-9.48.9, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26132178

RESUMEN

Mitochondrial dysfunction has been increasingly implicated as an important mechanism for chemical-induced toxicity. In the present unit, we describe a multi-parametric flow cytometry assay to assess the effects of drug or chemical-induced mitochondrial dysfunction in cells. Cells are cultured in a glucose-supplemented medium and exposed to increasing concentrations of various chemicals. Several key mitochondrial/cellular parameters known to be directly impacted by mitochondrial dysfunction, such as mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (ROS) production, intracellular reduced glutathione (GSH) level, and cell viability, are simultaneously measured by flow cytometry.


Asunto(s)
Citometría de Flujo/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Mitocondrias/metabolismo , Colorantes Fluorescentes/metabolismo , Células HL-60 , Humanos , Coloración y Etiquetado
9.
Toxicol Sci ; 142(1): 261-73, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25163676

RESUMEN

Mitochondrial perturbation has been recognized as a contributing factor to various drug-induced organ toxicities. To address this issue, we developed a high-throughput flow cytometry-based mitochondrial signaling assay to systematically investigate mitochondrial/cellular parameters known to be directly impacted by mitochondrial dysfunction: mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (ROS), intracellular reduced glutathione (GSH) level, and cell viability. Modulation of these parameters by a training set of compounds, comprised of established mitochondrial poisons and 60 marketed drugs (30 nM to 1mM), was tested in HL-60 cells (a human pro-myelocytic leukemia cell line) cultured in either glucose-supplemented (GSM) or glucose-free (containing galactose/glutamine; GFM) RPMI-1640 media. Post-hoc bio-informatic analyses of IC50 or EC50 values for all parameters tested revealed that MMP depolarization in HL-60 cells cultured in GSM was the most reliable parameter for determining mitochondrial dysfunction in these cells. Disruptors of mitochondrial function depolarized MMP at concentrations lower than those that caused loss of cell viability, especially in cells cultured in GSM; cellular GSH levels correlated more closely to loss of viability in vitro. Some mitochondrial respiratory chain inhibitors increased mitochondrial ROS generation; however, measuring an increase in ROS alone was not sufficient to identify mitochondrial disruptors. Furthermore, hierarchical cluster analysis of all measured parameters provided confirmation that MMP depletion, without loss of cell viability, was the key signature for identifying mitochondrial disruptors. Subsequent classification of compounds based on ratios of IC50s of cell viability:MMP determined that this parameter is the most critical indicator of mitochondrial health in cells and provides a powerful tool to predict whether novel small molecule entities possess this liability.


Asunto(s)
Glutatión/metabolismo , Sustancias Peligrosas/toxicidad , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Células HL-60 , Sustancias Peligrosas/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción
10.
Toxicol Pathol ; 42(1): 286-92, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24190913

RESUMEN

To assess relative sensitivity for detection of cytokines and chemokines in cynomolgus serum samples, we tested three commercially available multiplex array kits using the Luminex® platform with sera from animals exposed by intravenous injection to 150 µg/kg staphylococcal enterotoxin B (SEB) or 20 µg/kg lipopolysaccharide (LPS). Each of these kits detected similar patterns of changes in circulating cytokines/chemokines in response to SEB or LPS stimulation, especially the induction of high amounts of interleukin (IL)-2 and interferon-gamma (IFN-γ) in response to SEB but not LPS. However, there were clear differences in sensitivity for particular analytes, especially for IL-10. Additional experiments that focused on one multiplex array kit demonstrated very low or undetectable levels of cytokines in naive cynomolgus macaques, except for highly variable background levels of IL-8, monocyte chemoattractant protein-1, and macrophage inflammatory protein-1ß. Therefore, multiplex array analysis of circulating cytokine/chemokine patterns was capable of detection of systemic activation of diverse immune cell subsets.


Asunto(s)
Quimiocina CCL2/sangre , Interferón gamma/sangre , Interleucina-10/sangre , Interleucina-2/sangre , Interleucina-8/sangre , Análisis por Matrices de Proteínas/métodos , Administración Intravenosa , Animales , Quimiocina CCL4/sangre , Enterotoxinas/administración & dosificación , Enterotoxinas/efectos adversos , Femenino , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/efectos adversos , Macaca fascicularis/inmunología , Masculino , Juego de Reactivos para Diagnóstico
11.
J Immunol ; 191(11): 5551-8, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24184554

RESUMEN

IL-15 is a proinflammatory cytokine that plays an important role in the development and activation of NK cells and is a potential target for inflammatory disease therapy. Studies conducted in IL-15- and IL-15R knockout mice identified IL-15 as an important cytokine for NK cell homeostasis. Consistent with this information derived from genetically modified mice, we demonstrated that neutralizing IL-15 with a mouse anti-mouse IL-15 mAb (M96) depletes C57BL/6 mouse NK cells. An mAb directed against macaque IL-15 (Hu714MuXHu) was manufactured and demonstrated to block IL-15-induced activation of nonhuman primate (NHP) NK cells in vitro. Neutralization of macaque IL-15 by parenteral administration of Hu714MuXHu reduces (>95%) circulating NK cell counts in NHPs. A blocking mAb directed against human IL-15 (huIL-15; AMG 714) was manufactured. Unexpectedly, when human subjects were treated with the blocking anti-IL-15 Ab AMG 714 in clinical trials, no reductions in circulating NK cell counts were observed despite achieving significantly higher exposures than the levels of Hu714MuXHu needed to cause NK cell count reductions in NHPs in vivo. Both AMG 714 and Hu714MuXHu are able to block huIL-15 activity in a human T cell blast proliferation and IFN-γ production assay. Both Abs block huIL-15-mediated Stat5 activation and CD69 expression in human NK cells. Collectively, these results demonstrate that NK cell homeostasis is obligatorily dependent upon IL-15 in both mice and NHPs, but that IL-15 is dispensable for maintenance of circulating human NK cells.


Asunto(s)
Homeostasis , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayos Clínicos como Asunto , Homeostasis/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Interleucina-15/genética , Interleucina-15/inmunología , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Macaca , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT5/metabolismo , Activación Transcripcional/efectos de los fármacos
12.
J Immunol Methods ; 392(1-2): 29-37, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23541488

RESUMEN

Immunoglobulin E (IgE) is the least abundant immunoglobulin in serum. However, development of an IgE immune response can induce IgE receptor-expressing cells to carry out potent effector functions. A reliable antigen-specific IgE biomarker method for use in non-human primate studies would facilitate (i) confirmation of Type-I hypersensitivity reactions during safety toxicology testing, and (ii) a better understanding of non-human primate models of allergic disease. We cloned and expressed a recombinant cynomolgus monkey IgE molecule in order to screen a panel of commercially available detection reagents raised against human IgE for cross-reactivity. The reagent most reactive to cynomolgus IgE was confirmed to be specific for IgE and did not bind recombinant cynomolgus monkey IgG1-4. A drug-specific IgE assay was developed on the MSD electrochemiluminescent (ECL) platform. The assay is capable of detecting 10 ng/mL drug-specific IgE. Importantly, the assay is able to detect IgE in the presence of excess IgG, the scenario likely to be present in a safety toxicology study. Using our ECL assay, we were able to confirm that serum from cynomolgus monkeys that had experienced clinical symptoms consistent with hypersensitivity responses contained IgE specific for a candidate therapeutic antibody. In addition, a bioassay for mast cell activation was developed using CD34(+)-derived cynomolgus monkey mast cells. This assay confirmed that plasma from animals identified as positive in the drug-specific IgE immunoassay contained biologically active IgE (i.e. could sensitize cultured mast cells), resulting in histamine release after exposure to the therapeutic antibody. These sensitive assays for Type-I hypersensitivity in the NHP can confirm that secondary events are downstream of immunogenicity.


Asunto(s)
Hipersensibilidad Inmediata/inmunología , Inmunoensayo/métodos , Inmunoglobulina E/química , Inmunoglobulina E/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD34/inmunología , Biomarcadores/química , Reacciones Cruzadas , Epítopos , Humanos , Hipersensibilidad/inmunología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Macaca fascicularis , Mastocitos/inmunología , Datos de Secuencia Molecular , Receptores de IgE/inmunología
13.
Toxicol Pathol ; 41(7): 951-69, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475561

RESUMEN

Cynomolgus monkeys dosed with a therapeutic monoclonal antibody (mAbY.1) at ≥ 50 mg/kg had unexpected acute thrombocytopenia (nadir ~3,000 platelets/µl), sometimes with decreases in red cell mass. Increased activated macrophages, mitotic figures, and erythrophagocytosis were observed in the spleen. Binding of mAbY.1 to cynomolgus peripheral blood cells could not be detected in vitro. mAbY.1 induced phagocytosis of platelets by peripheral blood monocytes from cynomolgus monkeys, but not from humans. mAbs sharing the same constant domain (Fc) sequences, but differing from mAbY.1 in their variable domains, bound competitively to and had similar biological activity against the intended target. None of these antibodies had hematologic liabilities in vitro or in vivo. Neither the F(ab')2 portion of mAbY.1 nor the F(ab')2 portion on an aglycosylated Fc (IgG1) framework caused phagocytosis of platelets in vitro. These data suggest that the hematologic effects of mAbY.1 in cynomolgus monkeys likely occurred through an off-target mechanism, shown to be driven by 1 to 3 amino acid differences in the light chain. The hematologic effects made mAbY.1 an unsuitable candidate for further development as a therapeutic agent. This example demonstrates that nonclinical safety studies may be essential for understanding off-target effects of mAbs prior to clinical trials.


Asunto(s)
Anemia/inducido químicamente , Anticuerpos Monoclonales/toxicidad , Trombocitopenia/inducido químicamente , Anemia/sangre , Animales , Anticuerpos Monoclonales/administración & dosificación , Plaquetas/patología , Femenino , Humanos , Macaca fascicularis , Activación de Macrófagos , Masculino , Fagocitosis , Reticulocitos/patología , Bazo/efectos de los fármacos , Bazo/patología , Trombocitopenia/sangre
14.
Toxicol Appl Pharmacol ; 268(2): 113-22, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23416206

RESUMEN

Itraconazole (ITZ) is an approved antifungal agent that carries a "black box warning" in its label regarding a risk of negative cardiac inotropy based on clinical findings. Since the mechanism of the negative inotropic effect is unknown, we performed a variety of preclinical and mechanistic studies to explore the pharmacological profile of ITZ and understand the negative inotropic mechanism. ITZ was evaluated in: (1) an isolated rabbit heart (IRH) preparation using Langendorff retrograde perfusion; (2) ion channel studies; (3) a rat heart mitochondrial function profiling screen; (4) a mitochondrial membrane potential (MMP) assay; (5) in vitro pharmacology profiling assays (148 receptors, ion channels, transporters, and enzymes); and (6) a kinase selectivity panel (451 kinases). In the IRH, ITZ decreased cardiac contractility (>30%) at 0.3µM, with increasing effect at higher concentrations, which indicated a direct negative inotropic effect upon the heart. It also decreased heart rate and coronary flow (≥1µM) and prolonged PR/QRS intervals (3µM). In mechanistic studies, ITZ inhibited the cardiac NaV channel (IC50: 4.2µM) and was devoid of any functional inhibitory effect at the remaining pharmacological targets. Lastly, ITZ did not affect MMP, nor interfere with mitochondrial enzymes or processes involved with fuel substrate utilization or energy formation. Overall, the cardiovascular and mechanistic data suggest that ITZ-induced negative inotropy is a direct effect on the heart, in addition, the potential involvement of mitochondria function and L-type Ca(2+) channels are eliminated. The exact mechanism underlying the negative inotropy is uncertain, and requires further study.


Asunto(s)
Antifúngicos/farmacología , Itraconazol/farmacología , Contracción Miocárdica/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Animales , Depresión Química , Femenino , Técnicas In Vitro , Canales Iónicos/efectos de los fármacos , MAP Quinasa Quinasa 5/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Conejos , Ratas
15.
J Lab Autom ; 18(1): 85-98, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22968419

RESUMEN

We have developed an automated system for drug screening using a single-cell-multiple functional response technology. The approach uses a semiautomated preparatory system, high-speed sample collection, and a unique analytical tool that provides instantaneous results for compound dilutions using 384-well plates. The combination of automation and rapid robotic sampling increases quality control and robustness. High-speed flow cytometry is used to collect single-cell results together with a newly defined analytical tool for extraction of IC(50) curves for multiple assays per cell. The principal advantage is the extreme speed of sample collection, with results from a 384-well plate being completed for both collection and data processing in less than 10 min. Using this approach, it is possible to extract detailed drug response information in a highly controlled fashion. The data are based on single-cell results, not populations. With simultaneous assays for different functions, it is possible to gain a more detailed understanding of each drug/compound interaction. Combined with integrated advanced data processing directly from raw data files, the process from sampling to analytical results is highly intuitive. Direct PubMed links allow review of drug structure and comparisons with similar compounds.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Análisis de la Célula Individual/métodos , Automatización , Citometría de Flujo , Células HL-60 , Humanos , Concentración 50 Inhibidora , Mitocondrias/metabolismo , Factores de Tiempo
16.
PLoS One ; 7(10): e45226, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077490

RESUMEN

Early evaluation of new drug entities for their potential to cause mitochondrial dysfunction is becoming an important task for drug development. Multi-parametric high-content screening (mp-HCS) of mitochondrial toxicity holds promise as a lead in-vitro strategy for drug testing and safety evaluations. In this study, we have developed a mp-HCS and multi-parametric data analysis scheme for assessing cell responses to induced mitochondrial perturbation. The mp-HCS measurements are shown to be robust enough to allow for quantitative comparison of biological systems with different metabolic pathways simulated by alteration of growth media. Substitution of medium glucose for galactose sensitized cells to drug action and revealed novel response parameters. Each compound was quantitatively characterized according to induced phenotypic changes of cell morphology and functionality measured by fluorescent biomarkers for mitochondrial activity, plasma membrane permeability, and nuclear morphology. Descriptors of drug effects were established by generation of a SCRIT (Specialized-Cell-Response-to-Induced-Toxicity) vector, consisting of normalized statistical measures of each parameter at each dose and growth condition. The dimensionality of SCRIT vectors depends on the number of parameters chosen, which in turn depends on the hypothesis being tested. Specifically, incorporation of three parameters of response into SCRIT vectors enabled clustering of 84 training compounds with known pharmacological and toxicological activities according to the degree of toxicity and mitochondrial involvement. Inclusion of 6 parameters enabled the resolution of more subtle differences between compounds within a common therapeutic class; scoring enabled a ranking of statins in direct agreement with clinical outcomes. Comparison of drug-induced changes required variations in glucose for separation of mitochondrial dysfunction from other types of cytotoxicity. These results also demonstrate that the number of drugs in a training set, the choice of parameters used in analysis, and statistical measures are fundamental for specific hypothesis testing and assessment of quantitative phenotypic differences.


Asunto(s)
Mitocondrias/efectos de los fármacos , Pruebas de Toxicidad , Automatización , Análisis por Conglomerados , Medios de Cultivo , Mitocondrias/fisiología , Análisis Multivariante
17.
Toxicol Pathol ; 40(6): 899-917, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22552394

RESUMEN

AMG X, a human neutralizing monoclonal antibody (mAb) against a soluble human protein, caused thrombocytopenia, platelet activation, reduced mean arterial pressure, and transient loss of consciousness in cynomolgus monkeys after first intravenous administration. In vitro, AMG X induced activation in platelets from macaque species but not from humans or baboons. Other similar mAbs against the same pharmacological target failed to induce these in vivo and in vitro effects. In addition, the target protein was known to not be expressed on platelets, suggesting that platelet activation occurred through an off-target mechanism. AMG X bound directly to cynomolgus platelets and required both the Fab and Fc portion of the mAb for platelet activation. Binding to platelets was inhibited by preincubation of AMG X with its pharmacological target or with anti-human Fc antibodies or by preincubation of platelets with AMG X F(ab')(2) or human immunoglobulin (IVIG). AMG X F(ab')(2) did not activate platelets. Thus, platelet activation required both recognition/binding of a platelet ligand with the Fab domain and interaction of platelet Fc receptors (i.e., FcγRIIa) with the Fc domain. These findings reflect the complexity of the mechanism of action of mAbs and the increasing awareness of potential for unintended effects in preclinical species.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Plaquetas/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Administración Intravenosa , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacocinética , Plaquetas/metabolismo , Humanos , Hipotensión/sangre , Hipotensión/inducido químicamente , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Macaca fascicularis , Masculino , Papio , Agregación Plaquetaria/efectos de los fármacos , Unión Proteica , Serotonina/metabolismo , Síncope/sangre , Síncope/inducido químicamente , Trombocitopenia/sangre , Trombocitopenia/inducido químicamente , Tromboxano B2/metabolismo
18.
Toxicol Pathol ; 40(1): 107-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22033502

RESUMEN

Regulatory T cells (Tregs) are a rare subset of lymphocytes that inhibit the activation and effector functions of T cells and are important regulators of immune responses. Although Tregs are well characterized in humans and rodents, little is known about their immunophenotyping (IP) profile in cynomolgus macaques (Macaca fascicularis), which is an important species for pharmacological and toxicological evaluation of potential immune modulators because of their similar physiologic, genetic, and metabolic response patterns to humans. The authors have developed an immunophenotyping panel using a high-throughput 96-well microtiter plate-based assay to detect circulating Tregs (CD3(+)CD4(+)CD25(hi)FoxP3(+)) and have determined the normal range for the number of Tregs in naive healthy cynomolgus macaques to be 56.4 to 179.7 cells/µL (mean ± SEM = 113.6 ± 5.1 cells/µL; n = 25). Furthermore, the authors compared the resulting FoxP3(+) Treg profiles with a CD127(lo) cell-surface panel (CD3(+)CD4(+)CD25(hi) CD127(lo)) and found a close correlation between the absolute numbers of CD3(+)CD4(+)CD25(hi)FoxP3(+) and CD3(+)CD4(+)CD25(hi)CD127(lo) cells (mean ± SD = 120 ± 8.0 cells/µL). Quantification of circulating Tregs in cynomolgus macaques in this high-throughput assay may help to identify drug candidates that affect this rare, but critical, immunoregulatory cell population.


Asunto(s)
Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Macaca fascicularis/sangre , Linfocitos T Reguladores/citología , Animales , Antígenos CD/sangre , Femenino , Factores de Transcripción Forkhead/sangre , Ensayos Analíticos de Alto Rendimiento , Macaca fascicularis/inmunología , Masculino , Modelos Animales , Linfocitos T Reguladores/química
19.
J Immunotoxicol ; 9(1): 34-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22074378

RESUMEN

Human peripheral blood mononuclear cells (PBMC) are routinely used in vitro to detect cytokine secretion as part of preclinical screens to delineate agonistic and antagonistic action of therapeutic monoclonal antibodies (mAbs). Preclinical value of standard human PBMC assays to detect cytokine release syndrome (CRS) has been questioned, as they did not predict the "cytokine storm" that occurred when healthy human volunteers were given a CD28-specific super-agonist mAb, TGN1412. In this article, we describe a three-dimensional biomimetic vascular test-bed that can be used as a more physiologically relevant assay for testing therapeutic Abs. For developing such a system, we used TGN1412 as a model mAb. We tested soluble TGN1412 on various combinations of human blood components in a module containing endothelial cells grown on a collagen scaffold and measured cytokine release using multiplex array. Our system, consisting of whole leukocytes, endothelial cells, and 100% autologous platelet-poor plasma (PPP) consistently produced proinflammatory cytokines in response to soluble TGN1412. In addition, other mAb therapeutics known to induce CRS or first infusion reactions, such as OKT3, Campath-1H, or Herceptin, generated cytokine profiles in our model system consistent with their in vivo responses. As a negative control we tested the non-CRS mAbs Avastin and Remicade and found little difference between these mAbs and the placebo control. Our data indicate that this novel assay may have preclinical value for predicting the potential of CRS for mAb therapeutics.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Biomimética , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Granulocitos/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Pruebas de Toxicidad , Células Cultivadas , Técnicas de Cocultivo , Colágeno , Granulocitos/inmunología , Granulocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Plasma , Medición de Riesgo , Factores de Tiempo , Andamios del Tejido
20.
J Appl Toxicol ; 30(5): 387-96, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20589744

RESUMEN

Biopharmaceuticals represent significant advances in therapeutic approaches for unmet medical needs, and increasingly, traditional pharmaceutical firms have been incorporating biotechnology capabilities into their product portfolios. There are some differences in the overall safety testing paradigms for small molecules and biopharmaceuticals, this safety testing including both quality and toxicology aspects. These differences are associated with both the manufacturing processes involved and the molecules themselves. For example, for biopharmaceuticals, living cells represent the factories for synthesizing complex molecular entities. As a result of this, safety testing for this class of drugs includes adventitious agent testing (e.g. viral, mycoplasma, transmissible spongiform encephalopathy agents) not normally needed for small molecules. Also, strategies for nonclinical toxicology testing of biopharmaceuticals differ from the paradigms used for small molecules and often need to be defined on a case-by-case basis, primarily taking into consideration species cross-reactivity attributes of the molecule of interest. Certain studies required for small molecules are not applicable to most biopharmaceuticals (i.e. genotoxicity testing, testing for interactions with the hERG channel). This manuscript provides an overview of both the quality and nonclinical toxicology testing for these mammalian-cell-derived products, two elements pivotal to the overall nonclinical assessment of the safety of these biopharmaceutical products.


Asunto(s)
Biofarmacia/métodos , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Proteínas Recombinantes/efectos adversos , Pruebas de Toxicidad/métodos , Animales , Células Cultivadas , Contaminación de Medicamentos/prevención & control , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Preparaciones Farmacéuticas/normas , Control de Calidad , Proteínas Recombinantes/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...