Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
AAPS J ; 22(2): 42, 2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32020345

RESUMEN

The first author's name was published incorrectly as "Gorovits Boris". The correct name is "Boris Gorovits".

2.
AAPS J ; 22(2): 24, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31907680

RESUMEN

Viral vector-based gene therapies (GTx) have received significant attention in the recent years and the number of ongoing GTx clinical trials is increasing. A platform of choice for many of these studies is adeno-associated virus (AAV). All humans may be exposed to natural AAV infections and could mount an immune response against the virus. Consequently, there can be a high prevalence of pre-existing anti-AAV immunity. This presents a potential limitation for AAV-based GTx due to the potential for AAV-specific antibodies to reduce the efficacy of the GTx. Therefore, appropriate assessment of potential subjects enrolled in these studies should include evaluation for the presence and degree of anti-AAV immunity, including anti-AAV neutralizing antibodies (NAb). Recommendations for the development and validation of cell-based anti-AAV NAb detection methods, including considerations related to selection of appropriate cell line, surrogate vector/reporter gene, assay matrix and controls, and methodologies for calculating assay cut-point are discussed herein. General recommendations for the key assay validation parameters are provided as well as considerations for the development of NAb diagnostic tests. This manuscript is produced by a group of scientists involved in GTx therapeutic development representing various companies. It is our intent to provide recommendations and guidance to industrial and academic laboratories working on viral vector based GTx modalities with the goal of achieving a more consistent approach to anti-AAV NAb assessment.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bioensayo , Dependovirus/inmunología , Terapia Genética , Vectores Genéticos/inmunología , Pruebas Inmunológicas , Línea Celular , Dependovirus/genética , Genes Reporteros , Vectores Genéticos/genética , Humanos , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
3.
BioDrugs ; 33(3): 275-284, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31069709

RESUMEN

Chimeric antigen receptor (CAR) T-cell immunotherapy has gained significant attention in the past decade due to its considerable potential in the treatment of various types of malignancies, particularly hematological. While success has been achieved in a number of studies, and two CAR-T-cell products were recently approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) (YESCARTA®, KYMRIAH®), this treatment modality continues to present challenges for clinical development. One major potential side effect is the ability of CAR-T products to induce host immune responses. Immunogenicity induction risk factors have been shown to be associated with the presence of non-human or partially human sequences in the CAR construct, suicide domain, or other components of the CAR-T, and also with the presence of residual viral proteins or other non-human origin proteins utilized as part of the gene editing step of CAR-T production. Both humoral (antibody-based) and cellular-type responses have been described, leading to various degrees of impact on CAR-T expansion and persistence, and therefore the overall safety and clinically meaningful response of the treatment. In this article we discuss various types of immune responses specific to CAR-T therapy, their impact on treatment outcome, and methodologies used to detect them.


Asunto(s)
Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Edición Génica/métodos , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
4.
Cardiovasc Revasc Med ; 15(5): 269-77, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24972512

RESUMEN

PURPOSE: The purpose of this study was to investigate the effects of everolimus on foam cell (FC) viability, mRNA levels, and inflammatory cytokine production to better understand its potential inhibitory effects on atheroma progression. METHODS AND MATERIALS: Human THP1 macrophage-derived FC were formed using acetylated LDL (acLDL, 100 µg/mL) for 72 hours, followed by everolimus treatment (10(-5)-10(-11) M) for 24 hours. FC viability was quantified using fluorescent calcein AM/DAPI staining. FC lysates and media supernatants were analyzed for apoptosis and necrosis using a Cell Death ELISA(PLUS) assay. FC lysates and media supernatants were also analyzed for inflammatory cytokine (IL1ß, IL8, MCP1, TNFα) mRNA levels and protein expression using quantitative reverse transcription real-time polymerase chain reaction (QPCR) and a Procarta® immunoassay, respectively. mRNA levels of autophagy (MAP1LC3), apoptosis (survivin, clusterin), and matrix degradation (MMP1, MMP9) markers were evaluated by Quantigene® Plex assay and verified with QPCR. Additionally, hypercholesterolemic rabbits received everolimus-eluting stents (EES) for 28 or 60 days. RAM-11 immunohistochemical staining was performed to compare %RAM-11 positive area between stented sections and unstented proximal sections. Statistical significance was calculated using one-way ANOVA (p≤0.05). RESULTS: Calcein AM/DAPI staining showed that FC exposed to everolimus (10(-5) M) had significantly decreased viability compared to control. FC apoptosis was significantly increased at a high dose of everolimus (10(-5)M), with no necrotic effects at any dose tested. Everolimus did not affect endothelial (HUVEC) and smooth muscle (HCASMC) cell apoptosis or necrosis. Everolimus (10(-5)M) significantly increased MAP1LC3, caused an increased trend in clusterin (p=0.10), and significantly decreased survivin and MMP1 mRNA levels in FC. MCP1 cytokine mRNA levels and secreted protein expression was significantly decreased by everolimus (10(-5) M) in FC. Percentage of RAM-11 positive area exhibited a reduction trend within sections stented with EES compared to unstented proximal sections at 60 days (p=0.09). CONCLUSION: Everolimus, a potent anti-proliferative agent used in drug-eluting stents and bioresorbable vascular scaffolds, may inhibit atheroma progression and/or promote atheroma stabilization through diminished viability of FC, decreased matrix degradation, and reduced pro-inflammatory cytokine secretion. EXECUTIVE SUMMARY: We explored the effects of everolimus on the behavior of human THP1 macrophage-derived foam cells in culture, including cell viability, mRNA levels, and pro-inflammatory cytokine production. We conclude that everolimus, a potent anti-proliferative agent used in drug-eluting stents/bioresorbable vascular scaffolds, may potentially inhibit atheroma progression and/or promote atheroma stabilization through diminished viability of foam cells, decreased matrix degradation, and reduced pro-inflammatory cytokine secretion.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Stents Liberadores de Fármacos , Células Espumosas/efectos de los fármacos , Macrófagos/citología , Sirolimus/análogos & derivados , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Everolimus , Células Espumosas/citología , Humanos , Necrosis/inducido químicamente , Sirolimus/farmacología
5.
Mol Genet Metab ; 104(3): 325-37, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21784683

RESUMEN

Late infantile neuronal ceroid lipofuscinosis (LINCL) is caused by mutations in the gene encoding tripeptidyl-peptidase 1 (TPP1). LINCL patients accumulate lysosomal storage materials in the CNS accompanied by neurodegeneration, blindness, and functional decline. Dachshunds homozygous for a null mutation in the TPP1 gene recapitulate many symptoms of the human disease. The objectives of this study were to determine whether intrathecal (IT) TPP1 treatment attenuates storage accumulation and functional decline in TPP1-/- Dachshunds and to characterize the CNS distribution of TPP1 activity. TPP1 was administered to one TPP1-/- and one homozygous wild-type (WT) dog. An additional TPP1-/- and WT dog received vehicle. Four IT administrations of 32 mg TPP1 formulated in 2.3 mL of artificial cerebrospinal fluid (aCSF) or vehicle were administered monthly via the cerebellomedullary cistern from four to seven months of age. Functional decline was assessed by physical and neurological examinations, electrophysiology, and T-maze performance. Neural tissues were collected 48 h after the fourth administration and analyzed for TPP1 activity and autofluorescent storage material. TPP1 was distributed at greater than WT levels in many areas of the CNS of the TPP1-/- dog administered TPP1. The amount of autofluorescent storage was decreased in this dog relative to the vehicle-treated affected control. No improvement in overall function was observed in this dog compared to the vehicle-treated TPP1-/- littermate control. These results demonstrate for the first time in a large animal model of LINCL widespread delivery of biochemically active TPP1 to the brain after IT administration along with a decrease in lysosomal storage material. Further studies with this model will be necessary to optimize the dosing route and regimen to attenuate functional decline.


Asunto(s)
Aminopeptidasas/farmacología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/farmacología , Lisosomas/metabolismo , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/metabolismo , Serina Proteasas/farmacología , Aminopeptidasas/administración & dosificación , Aminopeptidasas/sangre , Aminopeptidasas/genética , Aminopeptidasas/uso terapéutico , Animales , Células CHO , Sistema Nervioso Central/metabolismo , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Cricetinae , Cricetulus , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/administración & dosificación , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/sangre , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/uso terapéutico , Perros , Electrofisiología , Fluorescencia , Técnicas de Inactivación de Genes , Humanos , Inmunoensayo , Inmunoglobulina E/sangre , Inyecciones Espinales , Imagen por Resonancia Magnética , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Recombinantes/farmacología , Serina Proteasas/administración & dosificación , Serina Proteasas/sangre , Serina Proteasas/genética , Serina Proteasas/uso terapéutico , Tripeptidil Peptidasa 1
6.
J Pharm Biomed Anal ; 48(5): 1267-81, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18993008

RESUMEN

Most biological drug products elicit some level of anti-drug antibody (ADA) response. This antibody response can, in some cases, lead to potentially serious side effects and/or loss of efficacy. In humans, ADA often causes no detectable clinical effects, but in the instances of some therapeutic proteins these antibodies have been shown to cause a variety of clinical consequences ranging from relatively mild to serious adverse events. In nonclinical (preclinical) studies, ADA can affect drug exposure, complicating the interpretation of the toxicity, pharmacokinetic (PK) and pharmacodynamic (PD) data. Therefore, the immunogenicity of therapeutic proteins is a concern for clinicians, manufacturers and regulatory agencies. In order to assess the immunogenic potential of biological drug molecules, and be able to correlate laboratory results with clinical events, it is important to develop reliable laboratory test methods that provide valid assessments of antibody responses in both nonclinical and clinical studies. For this, method validation is considered important, and is a necessary bioanalytical component of drug marketing authorization applications. Existing regulatory guidance documents dealing with the validation of methods address immunoassays in a limited manner, and in particular lack information on the validation of immunogenicity methods. Hence this article provides scientific recommendations for the validation of ADA immunoassays. Unique validation performance characteristics are addressed in addition to those provided in existing regulatory documents pertaining to bioanalyses. The authors recommend experimental and statistical approaches for the validation of immunoassay performance characteristics; these recommendations should be considered as examples of best practice and are intended to foster a more unified approach to antibody testing across the biopharmaceutical industry.


Asunto(s)
Anticuerpos/análisis , Productos Biológicos/inmunología , Biotecnología , Inmunoensayo/normas , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Directrices para la Planificación en Salud , Humanos , Reproducibilidad de los Resultados
7.
J Immunol Methods ; 333(1-2): 1-9, 2008 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-18275969

RESUMEN

The appropriate evaluation of the immunogenicity of biopharmaceuticals is of major importance for their successful development and licensure. Antibodies elicited by these products in many cases cause no detectable clinical effects in humans. However, antibodies to some therapeutic proteins have been shown to cause a variety of clinical consequences ranging from relatively mild to serious adverse events. In addition, antibodies can affect drug efficacy. In non-clinical studies, anti-drug antibodies (ADA) can complicate interpretation of the toxicity, pharmacokinetic (PK) and pharmacodynamic (PD) data. Therefore, it is important to develop testing strategies that provide valid assessments of antibody responses in both non-clinical and clinical studies. This document provides recommendations for antibody testing strategies stemming from the experience of contributing authors. The recommendations are intended to foster a more unified approach to antibody testing across the biopharmaceutical industry. The strategies proposed are also expected to contribute to better understanding of antibody responses and to further advance immunogenicity evaluation.


Asunto(s)
Anticuerpos/análisis , Productos Biológicos/inmunología , Biotecnología/métodos , Animales , Humanos , Medición de Riesgo
8.
J Immunol Methods ; 308(1-2): 101-8, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16356511

RESUMEN

Neutralizing antibodies against therapeutic proteins can be potentially harmful if the antibody blocks not only the therapeutic activities of the therapeutic protein but also the normal functions of the endogenous counterpart. Detection of the neutralizing anti-therapeutic protein antibodies generally relies on bioassays measuring changes in the biologic activity of the therapeutic protein triggered by the presence of the antibody. Most of the bioassays, particularly the cell-based in vitro assays, fail to detect neutralizing anti-therapeutic protein antibodies when the remaining therapeutic protein level in the assay samples is high. The remaining therapeutic protein, either a free molecule or an immune complex with anti-therapeutic protein antibodies, can inhibit the neutralizing activity of the antibody and prevent detection. We describe the development of a procedure that uses acid dissociation and affinity adsorption to remove therapeutic protein from assay samples. With this procedure, we can detect the presence of neutralizing anti-therapeutic protein antibodies from samples containing high levels of therapeutic protein.


Asunto(s)
Anticuerpos/sangre , Pruebas de Neutralización/métodos , Proteínas Recombinantes/sangre , Proteínas Recombinantes/inmunología , Anticuerpos Monoclonales/sangre , Complejo Antígeno-Anticuerpo/sangre , Complejo Antígeno-Anticuerpo/aislamiento & purificación , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Técnicas de Inmunoadsorción , Técnicas In Vitro , Interleucina-8/biosíntesis , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/inmunología , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico
10.
J Immunol Methods ; 304(1-2): 189-95, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16107258

RESUMEN

Bridging Enzyme-Linked Immunosorbent Assay (ELISA) is commonly used in detection of antibodies directed against therapeutic proteins. Advantages of the bridging ELISA include the capability to detect antibodies regardless of their isotype or the species of origin. However, detection of antibodies can be difficult, if not impossible, in the presence of high levels of the antigen in the sample matrix. This protocol describes a bridging ELISA that uses a covalently coupled high density antigen surface combined with an acid dissociation step to allow for antibody detection in the presence of antigen in human serum.


Asunto(s)
Anticuerpos/análisis , Antígenos/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas/inmunología , Proteínas/uso terapéutico , Calibración , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...