Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 150: 105630, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38642729

RESUMEN

Immunotoxicology/immunosafety science is rapidly evolving, with novel modalities and immuno-oncology among the primary drivers of new tools and technologies. The Immunosafety Working Group of IQ/DruSafe sought to better understand some of the key challenges in immunosafety evaluation, gaps in the science, and current limitations in methods and data interpretation. A survey was developed to provide a baseline understanding of the needs and challenges faced in immunosafety assessments, the tools currently being applied across the industry, and the impact of feedback received from regulatory agencies. This survey also focused on current practices and challenges in conducting the T-cell-dependent antibody response (TDAR) and the cytokine release assay (CRA). Respondents indicated that ICH S8 guidance was insufficient for the current needs of the industry portfolio of immunomodulators and novel modalities and should be updated. Other challenges/gaps identified included translation of nonclinical immunosafety assessments to the clinic, and lack of relevant nonclinical species and models in some cases. Key areas of emerging science that will add future value to immunotoxicity assessments include development of additional in vitro and microphysiological system models, as well as application of humanized mouse models. Efforts are ongoing in individual companies and consortia to address some of these gaps and emerging science.


Asunto(s)
Factores Inmunológicos , Humanos , Animales , Encuestas y Cuestionarios , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/toxicidad , Citocinas/inmunología , Medición de Riesgo , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Toxicidad/métodos
2.
Mod Rheumatol ; 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38801704

RESUMEN

OBJECTIVES: We characterised changes in absolute lymphocyte counts (ALCs) and lymphocyte subset counts (LSCs), and their relationship to incidence of serious infection events (SIEs) and herpes zoster (HZ) events in Japanese patients with moderate to severe rheumatoid arthritis enrolled in the tofacitinib clinical programme. METHODS: Data included 765 patients receiving tofacitinib in Phase 2, Phase 3, and long-term extension studies. ALCs/LSCs and incidence rates (patients with events/100 patient-years) of SIEs and HZ were analysed over 75 months. RESULTS: Median ALCs were generally stable over 75 months of treatment. Transient numerical increases from baseline in median LSCs were observed at Month 3; LSCs were generally lower than baseline for Months 36-75. SIE/HZ incidence rates were higher in patients with ALC <0.5 × 103 cells/mm3 versus those with ALC ≥0.5 × 103 cells/mm3 during tofacitinib treatment. Baseline LSCs were similar in patients with/without SIEs or HZ events. CONCLUSIONS: SIE/HZ risk was highest in patients with ALC <0.5 × 103 cells/mm3, supporting this threshold as clinically relevant for defining increased SIE/HZ risk in Japanese patients with rheumatoid arthritis receiving tofacitinib. However, SIEs and HZ events did not necessarily occur simultaneously with confirmed lymphopenia, preventing conclusions on possible causal relationships being drawn.

3.
J Pharmacol Exp Ther ; 386(1): 80-92, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37142443

RESUMEN

Blocking chemokine receptor C-C chemoattractant cytokine (chemokine) receptor (CCR) 6-dependent T cell migration has therapeutic promise in inflammatory diseases. PF-07054894 is a novel CCR6 antagonist that blocked only CCR6, CCR7, and C-X-C chemoattractant cytokine (chemokine) receptor (CXCR) 2 in a ß-arrestin assay panel of 168 G protein-coupled receptors. Inhibition of CCR6-mediated human T cell chemotaxis by (R)-4-((2-(((1,4-Dimethyl-1H-pyrazol-3-yl)(1-methylcyclopentyl)methyl)amino)-3,4-dioxocyclobut-1-en-1-yl)amino)-3-hydroxy-N,N-dimethylpicolinamide (PF-07054894) was insurmountable by CCR6 ligand, C-C motif ligand (CCL) 20. In contrast, blockade of CCR7-dependent chemotaxis in human T cells and CXCR2-dependent chemotaxis in human neutrophils by PF-07054894 were surmountable by CCL19 and C-X-C motif ligand 1, respectively. [3H]-PF-07054894 showed a slower dissociation rate for CCR6 than for CCR7 and CXCR2 suggesting that differences in chemotaxis patterns of inhibition could be attributable to offset kinetics. Consistent with this notion, an analog of PF-07054894 with fast dissociation rate showed surmountable inhibition of CCL20/CCR6 chemotaxis. Furthermore, pre-equilibration of T cells with PF-07054894 increased its inhibitory potency in CCL20/CCR6 chemotaxis by 10-fold. The functional selectivity of PF-07054894 for inhibition of CCR6 relative to CCR7 and CXCR2 is estimated to be at least 50- and 150-fold, respectively. When administered orally to naïve cynomolgus monkeys, PF-07054894 increased the frequency of CCR6+ peripheral blood T cells, suggesting that blockade of CCR6 inhibited homeostatic migration of T cells from blood to tissues. PF-07054894 inhibited interleukin-23-induced mouse skin ear swelling to a similar extent as genetic ablation of CCR6. PF-07054894 caused an increase in cell surface CCR6 in mouse and monkey B cells, which was recapitulated in mouse splenocytes in vitro. In conclusion, PF-07054894 is a potent and functionally selective CCR6 antagonist that blocks CCR6-mediated chemotaxis in vitro and in vivo. SIGNIFICANCE STATEMENT: The chemokine receptor, C-C chemoattractant cytokine (chemokine) receptor 6 (CCR6) plays a key role in the migration of pathogenic lymphocytes and dendritic cells into sites of inflammation. (R)-4-((2-(((1,4-Dimethyl-1H-pyrazol-3-yl)(1-methylcyclopentyl)methyl)amino)-3,4-dioxocyclobut-1-en-1-yl)amino)-3-hydroxy-N,N-dimethylpicolinamide (PF-07054894) is a novel CCR6 small molecule antagonist that illustrates the importance of binding kinetics in achieving pharmacological potency and selectivity. Orally administered PF-07054894 blocks homeostatic and pathogenic functions of CCR6, suggesting that it is a promising therapeutic agent for the treatment of a variety of autoimmune and inflammatory diseases.


Asunto(s)
Quimiocinas CC , Interleucina-23 , Humanos , Animales , Ratones , Quimiocinas CC/genética , Receptores CCR7 , Ligandos , Linfocitos T , Inflamación , Receptores CCR6
4.
Clin Immunol ; 191: 10-20, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29518577

RESUMEN

This study evaluated the short-term effects of tofacitinib treatment on peripheral blood leukocyte phenotype and function, and the reversibility of any such effects following treatment withdrawal in healthy volunteers. Cytomegalovirus (CMV)-seropositive subjects received oral tofacitinib 10 mg twice daily for 4 weeks and were followed for 4 weeks after drug withdrawal. There were slight increases in total lymphocyte and total T-cell counts during tofacitinib treatment, and B-cell counts increased by up to 26%. There were no significant changes in granulocyte or monocyte counts, or granulocyte function. Naïve and central memory T-cell counts increased during treatment, while all subsets of activated T cells were decreased by up to 69%. T-cell subsets other than effector memory cluster of differentiation (CD)4+, activated naïve CD4+ and effector CD8+ T-cell counts and B-cell counts, normalized 4 weeks after withdrawal. Following ex vivo activation, measures of CMV-specific T-cell responses, and antigen non-specific T-cell-mediated cytotoxicity and interferon (IFN)-γ production, decreased slightly. These T-cell functional changes were most pronounced at Day 15, partially normalized while still on tofacitinib and returned to baseline after drug withdrawal. Total natural killer (NK)-cell counts decreased by 33%, returning towards baseline after drug withdrawal. NK-cell function decreased during tofacitinib treatment, but without a consistent time course across measured parameters. However, markers of NK-cell-mediated cytotoxicity, antibody-dependent cellular cytotoxicity and IFN-γ production were decreased up to 42% 1 month after drug withdrawal. CMV DNA was not detectable in whole blood, and there were no cases of herpes zoster reactivation. No new safety concerns arose. In conclusion, the effect of short-term tofacitinib treatment on leukocyte composition and function in healthy CMV+ volunteers is modest and largely reversible 4 weeks after withdrawal.


Asunto(s)
Inhibidores de las Cinasas Janus/farmacología , Leucocitos/efectos de los fármacos , Piperidinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Adulto , Anciano , Artritis Reumatoide/tratamiento farmacológico , Femenino , Voluntarios Sanos , Humanos , Leucocitos/inmunología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Fenotipo , Piperidinas/efectos adversos , Pirimidinas/efectos adversos , Pirroles/efectos adversos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
5.
Regul Toxicol Pharmacol ; 94: 306-322, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29454012

RESUMEN

Tofacitinib, an oral Janus kinase (JAK) inhibitor for treatment of rheumatoid arthritis, targets JAK1, JAK3, and to a lesser extent JAK2 and TYK2. JAK1/3 inhibition impairs gamma common chain cytokine receptor signaling, important in lymphocyte development, homeostasis and function. Adult and juvenile cynomolgus monkey and rat studies were conducted and the impact of tofacitinib on immune parameters (lymphoid tissues and lymphocyte subsets) and function (T-dependent antibody response (TDAR), mitogen-induced T cell proliferation) assessed. Tofacitinib administration decreased circulating T cells and NK cells in juvenile and adult animals of both species. B cell decreases were observed only in rats. These changes and decreased lymphoid tissue cellularity are consistent with the expected pharmacology of tofacitinib. No differences were observed between juvenile and adult animals, either in terms of doses at which effects were observed or differential effects on immune endpoints. Lymphomas were observed in three adult monkeys. Tofacitinib impaired the primary TDAR in juvenile monkeys, although a recall response was generated. Complete or partial reversal of the effects on the immune system was observed.


Asunto(s)
Envejecimiento/inmunología , Inhibidores de las Cinasas Janus/toxicidad , Piperidinas/toxicidad , Pirimidinas/toxicidad , Pirroles/toxicidad , Administración Oral , Animales , Antígenos/inmunología , Recuento de Eritrocitos , Femenino , Hematócrito , Hemocianinas/inmunología , Hemoglobinas/análisis , Inhibidores de las Cinasas Janus/farmacocinética , Recuento de Leucocitos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Linfoma de Células B/inducido químicamente , Macaca fascicularis , Masculino , Tamaño de los Órganos/efectos de los fármacos , Piperidinas/farmacocinética , Pirimidinas/farmacocinética , Pirroles/farmacocinética , Ratas Sprague-Dawley , Bazo/efectos de los fármacos , Bazo/patología , Timo/efectos de los fármacos , Timo/patología , Pruebas de Toxicidad Crónica
6.
Toxicol Pathol ; 42(7): 1069-81, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24604381

RESUMEN

Comparative nonclinical studies were conducted with the proposed biosimilar PF-05280586 and rituximab-EU (MabThera®). In side-by-side analyses, peptide maps and complement-dependent cytotoxicity assay results were similar. Sexually-mature cynomolgus monkeys were administered PF-05280586 or rituximab-EU as a single dose of 0, 2, 10, or 20 mg/kg on day 1 and observed for 92 days (single-dose study) or as 5 weekly injections of 0 or 20 mg/kg and necropsied on day 30, the day after the 5th dose, or on day 121 (repeat-dose study). The pharmacokinetic and pharmacodynamic profiles for both molecules were similar. Marked depletion of peripheral blood B cells 4 days after dosing was followed by near or complete repletion (single-dose study) or partial repletion (repeat-dose study). In the single-dose study, anti-drug antibodies (ADA) were detected by day 29 in all animals administered PF-05280586 or rituximab-EU and persisted through day 85, the last day tested. In the repeat-dose study, ADA were detected on day 121 in 50% of animals administered PF-05280586 or rituximab-EU. Both molecules were well tolerated at all doses. In all endpoints evaluated, PF-05280586 exhibited similarity to rituximab-EU.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/farmacocinética , Animales , Antígenos CD20/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Biosimilares Farmacéuticos/administración & dosificación , Biosimilares Farmacéuticos/farmacología , Relación Dosis-Respuesta a Droga , Determinación de Punto Final , Femenino , Macaca fascicularis , Masculino , Reproducibilidad de los Resultados , Rituximab
7.
Regul Toxicol Pharmacol ; 69(1): 7-21, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24566336

RESUMEN

The T-cell-dependent antibody response (TDAR) assay is a measure of immune function that is dependent upon the effectiveness of multiple immune processes, including antigen uptake and presentation, T cell help, B cell activation, and antibody production. It is used for risk and safety assessments, in conjunction with other toxicologic assessments, by the chemical and pharmaceutical industries, and research and regulatory agencies. It is also employed to evaluate investigational drug efficacy in animal pharmacology studies, provide evidence of biological impact in clinical trials, and evaluate immune function in patients with primary or secondary immunodeficiency diseases. Various immunization schemes, analytical methods, approaches to data analysis, and data interpretations are in use. This manuscript summarizes some recommended practices for the conduct and interpretation of the assay in animal studies.


Asunto(s)
Formación de Anticuerpos/inmunología , Bioensayo/métodos , Medición de Riesgo/métodos , Linfocitos T/inmunología , Animales , Ensayos Clínicos como Asunto , Industria Farmacéutica/métodos , Humanos , Proyectos de Investigación
8.
Am J Pathol ; 180(4): 1751-60, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322302

RESUMEN

HuR is a member of the Drosophila Elav protein family that binds mRNA degradation sequences and prevents RNase-mediated degradation. Such HuR-mediated mRNA stabilization, which is stimulated by integrin engagement and is controlled at the level of HuR nuclear export, is critically involved in T-cell cytokine production. However, HuR's role in macrophage soluble factor production, in particular in response to angiogenic stimuli, has not yet been established. We show that the labile transcripts that encode vascular endothelial growth factor and matrix metalloproteinase-9 are stabilized when murine macrophages adhere to the ß(2) integrin ligand intercellular adhesion molecule-1. This mRNA stabilization response was absent in bone marrow-derived macrophages obtained from conditional macrophage-specific HuR knockout mice. The microvascular angiogenic response to an inflammatory stimulus (ie, subcutaneous polyvinyl alcohol sponge implantation) was markedly diminished in these macrophage HuR knockout mice despite the equal levels of macrophage localization to those observed in littermate wild-type controls. Furthermore, blood flow recovery and ischemic muscle neovascularization after femoral artery ligation were impaired in the conditional macrophage-specific HuR knockout mice. These results demonstrate that dynamic effects on mRNA, mediated by the RNA-binding and RNA-stabilizing protein HuR, are required for macrophage production of angiogenic factors, which play critical roles in the neovascular responses to a variety of stimuli, including tissue ischemia.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Antígenos CD18/fisiología , Proteínas ELAV/fisiología , Macrófagos/metabolismo , Neovascularización Patológica/metabolismo , Animales , Adhesión Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas ELAV/deficiencia , Proteínas ELAV/genética , Regulación de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes , Miembro Posterior/irrigación sanguínea , Inflamación/complicaciones , Isquemia/genética , Isquemia/fisiopatología , Ratones , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Neovascularización Patológica/etiología , Neovascularización Patológica/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
9.
J Immunotoxicol ; 20(1): 2176952, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36788724

RESUMEN

Cytotoxic T-lymphocytes (CTL) are a subset of T-cells that play a critical role in protecting against intracellular infections and cancer, and have the ability to identify and kill infected or transformed cells expressing non-self peptides associated with major histocompatibility (MHC) Class I molecules. Conversely, aberrant CTL activity can contribute to immune-related pathology under conditions of overwhelming infection or autoimmunity. Disease-modifying therapeutics can have unintended effects on CTL, and a growing number of therapeutics are intended to either suppress or enhance CTL or their functions. The susceptibility of CTL to unintended effects from common therapeutic modalities underscores the need for a better understanding of the impact that such therapies have on CTL function and the associated safety implications. While there are reliable ways of quantifying CTL, notably via flow cytometric analysis of specific CTL markers, it has been a greater challenge to implement fit-for-purpose methods measuring CTL function in the context of safety studies of therapeutics. This review focuses on methods for measuring CTL responses in the context of drug safety and pharmacology testing, with the goals of informing the reader about current approaches, evaluating their pros and cons, and providing perspectives on the utility of these approaches for safety evaluation.


Asunto(s)
Neoplasias , Linfocitos T Citotóxicos , Animales , Primates , Neoplasias/terapia , Citotoxicidad Inmunológica
10.
Birth Defects Res B Dev Reprod Toxicol ; 95(5): 363-75, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22969064

RESUMEN

BACKGROUND: There is concern about embryo-fetal exposure to antibody-based biopharmaceuticals based on the increase of such therapies being prescribed to women of childbearing potential. Therefore, there is a desire to better characterize embryo-fetal exposure of these molecules. The pregnant rat is a standard model for evaluating the potential consequences of exposure but placental transfer of antibody-based biopharmaceuticals is not well understood in this model. METHODS: The relative embryo-fetal distribution of an antibody-based biopharmaceutical was evaluated in the rat. Bevacizumab (Avastin) was chosen as a tool antibody since it does not have significant target binding in the rat that might influence embryo-fetal biodistribution. Avastin was labeled with a fluorescent dye, characterized, and injected into pregnant rats at different gestation ages. Labeled Avastin in fetal tissues was visualized ex vivo using an IVIS 200 (Caliper, A PerkinElmer Company, Alameda, CA). RESULTS: Avastin localized to the fetus as early as 24-hr post intravenous injection of the dam, and was taken up by the fetus in a dose-dependent manner. Avastin was detectable in the developing embryo as early as gestation day 13 and continued to be transferred until the end of gestation. Fetal transfer of Avastins mutated in the portion of the antibody that binds the neonatal Fc receptor (FcRn) was tested in late gestation and was found to correlate with affinities of the mutant Avastin antibody to FcRn. CONCLUSIONS: The novel application of this imaging technology was used to characterize the onset and duration of Avastin maternal-fetal transfer in rats and the importance of FcRn binding.


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Embrión de Mamíferos/metabolismo , Feto/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Intercambio Materno-Fetal , Receptores Fc/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab , Relación Dosis-Respuesta a Droga , Membranas Extraembrionarias/metabolismo , Femenino , Embarazo , Unión Proteica , Ratas , Ratas Sprague-Dawley
11.
ALTEX ; 40(2): 314­336, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-36044561

RESUMEN

Immune responses are heavily involved in the regulation and pathogenesis of human diseases, including infectious diseases, inflammatory and autoimmune conditions, cancer, neurological disorders, and cardiometabolic syndromes. The immune system is considered a double-edged sword serving as a powerful host defense mechanism against infection and cancerous cells and causing detrimental tissue damage when the immune response is exaggerated or uncontrollable. One of the challenges in studying the efficacy and toxicity of drugs that target or modulate the immune system is the lack of suitable preclinical human models that are predictive of human response. Recent advancements in human microphysiological systems (MPS) have provided a promising in vitro platform to evaluate the response of immune organs ex vivo, to investigate the interaction of immune cells with non-lymphoid tissue cells, and to reduce the reliance on animals in preclinical studies. The development, regulation, trafficking, and responses of immune cells have been extensively studied in preclinical animal models and clinically, providing a wealth of knowledge by which to evaluate new in vitro models. Therefore, the application of immunocompetent MPS in drug discovery and development should first verify that the immune response in an MPS model recapitulates the complexity of the human immune physiology. This manuscript reviews biological functions of immune organ systems and tissue-resident immune cells and discusses contexts-of-use for commonly used immunocompetent and immune organ MPS models. Current perspective and recommendations are provided to guide the continued development of immune organ and immunocompetent MPS models and their application in drug discovery and development.

12.
J Toxicol Sci ; 46(3): 99-114, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33642521

RESUMEN

Microphysiological systems (MPS) are making advances to provide more standardized and predictive physiologically relevant responses to test articles in living tissues and organ systems. The excitement surrounding the potential of MPS to better predict human responses to medicines and improving clinical translation is overshadowed by their relatively slow adoption by the pharmaceutical industry and regulators. Collaboration between multiorganizational consortia and regulators is necessary to build an understanding of the strengths and limitations of MPS models and closing the current gaps. Here, we review some of the advances in MPS research, focusing on liver, intestine, vascular system, kidney and lung and present examples highlighting the context of use for these systems. For MPS to gain a foothold in drug development, they must have added value over existing approaches. Ideally, the application of MPS will augment in vivo studies and reduce the use of animals via tiered screening with less reliance on exploratory toxicology studies to screen compounds. Because MPS support multiple cell types (e.g. primary or stem-cell derived cells) and organ systems, identifying when MPS are more appropriate than simple 2D in vitro models for understanding physiological responses to test articles is necessary. Once identified, MPS models require qualification for that specific context of use and must be reproducible to allow future validation. Ultimately, the challenges of balancing complexity with reproducibility will inform the promise of advancing the MPS field and are critical for realization of the goal to reduce, refine and replace (3Rs) the use of animals in nonclinical research.


Asunto(s)
Desarrollo de Medicamentos/métodos , Desarrollo de Medicamentos/tendencias , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Animales , Productos Biológicos , Industria Farmacéutica , Predicción , Humanos , Dispositivos Laboratorio en un Chip
13.
Toxicol Pathol ; 38(7): 1111-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20962107

RESUMEN

Developmental immunotoxicity (DIT) has gained attention with the recognition that environmental chemicals can potentially affect the developing immune system and the incidence of childhood allergic diseases. Preclinical safety assessment of pharmaceuticals for men and women of childbearing potential as well as for pediatric and juvenile indications may require DIT assessments. Draft documents from environmental and chemical regulatory agencies propose strategies that use the rat as a test species and incorporate histopathology and functional testing as endpoints. While there are no guidelines for DIT assessment of pharmaceuticals, current discussions suggest that combining immunotoxicity and developmental and reproductive toxicology studies may serve this purpose. Knowledge of the principles and applications of DIT will facilitate participation in strategy development and effective conduct of relevant studies.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Desarrollo Fetal/efectos de los fármacos , Enfermedades del Sistema Inmune/inducido químicamente , Sistema Inmunológico/efectos de los fármacos , Xenobióticos/efectos adversos , Animales , Anticuerpos Monoclonales/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Femenino , Desarrollo Fetal/inmunología , Sistema Inmunológico/embriología , Sistema Inmunológico/crecimiento & desarrollo , Enfermedades del Sistema Inmune/fisiopatología , Inmunidad/efectos de los fármacos , Inmunidad/fisiología , Masculino , Ratones , Ratas , Medición de Riesgo , Pruebas de Toxicidad/métodos
14.
J Immunotoxicol ; 17(1): 51-58, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32124652

RESUMEN

Nonclinical immunotoxicity evaluation is an important component of safety assessment for pharmaceuticals. One in vitro assay that can be applied in a weight of evidence assessment is the human lymphocyte activation (HuLA) assay, an antigen recall assay, similar in many respects to the in vivo T-cell-dependent antibody response (TDAR) in that cooperation of multiple immune cell types are needed to produce responses. This assay uses human cells and is more amenable than the TDAR to compound ranking and mechanistic studies. The HuLA assay requires less time and drug than TDAR assays, uses a relevant antigen (influenza), reflects a human immune response, and applies principles of the 3Rs to non-clinical safety assessment. Peripheral blood mononuclear cells (PBMC) from flu-immunized donors are re-stimulated with flu-vaccine in the presence of test articles, and proliferation is measured. Published data demonstrate the applicability of the HuLA assay, but it has not been evaluated for reproducibility across testing sites. To evaluate assay reproducibility, scientists from a consortium of institutions conducted the assay in parallel, using a common pool of donor PBMC, influenza vaccine, and known immunosuppressant compounds (cyclosporine A and mycophenolic acid). The HuLA assay was highly reproducible in identification of inhibition of antigen-specific responses, and there was significant agreement across testing sites in the half maximal inhibitory concentration (IC50) values. Intra-site variability was the largest contributor to the variability observed within the assay. The HuLA assay was demonstrated to be ideally suited to comparing multiple compounds (i.e. compound ranking or benchmarking) within the same assay. Overall, the data reported herein support the HuLA assay as a useful tool in mechanistic evaluations of antigen-specific immune responses.


Asunto(s)
Bioensayo/instrumentación , Pruebas Inmunológicas de Citotoxicidad/métodos , Evaluación Preclínica de Medicamentos/métodos , Activación de Linfocitos/efectos de los fármacos , Células Cultivadas , Ciclosporina/farmacología , Voluntarios Sanos , Humanos , Inmunosupresores/farmacología , Vacunas contra la Influenza/inmunología , Concentración 50 Inhibidora , Leucocitos Mononucleares , Activación de Linfocitos/inmunología , Ácido Micofenólico/farmacología , Reproducibilidad de los Resultados
15.
PLoS One ; 13(7): e0201103, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30048492

RESUMEN

Activation of the ß2 integrin lymphocyte function-associated antigen-1 (LFA-1) in T cells induces stabilization of proinflammatory AU-rich element (ARE)-bearing mRNAs, by triggering the nuclear-to-cytoplasmic translocation of the mRNA-binding and -stabilizing protein HuR. However, the mechanism by which LFA-1 engagement controls HuR localization is not known. Here, we identify and characterize four key regulators of LFA-1-induced changes in HuR activity: the p38 pathway kinase MK2 and the constitutive nuclear proteins hnRNPs C, H1 and K. LFA-1 engagement results in rapid, sequential activation of p38 and MK2. Post-LFA-1 activation, MK2 inducibly associates with both hnRNPC and HuR, resulting in the dissociation of HuR from hnRNPs C, H1 and K. Freed from the three hnRNPs, HuR translocates from the nucleus to the cytoplasm, and mediates the stabilization of labile cytokine transcripts. Our results suggest that the modulation of T cell cytokine mRNA half-life is an intricate process that is negatively regulated by hnRNPs C, H1 and K and requires MK2 as a critical activator.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad del ARN/fisiología , Linfocitos T/metabolismo , Animales , Técnicas de Cultivo de Célula , Citoplasma/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células Jurkat , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteoma , ARN Mensajero/metabolismo , Transducción de Señal , Linfocitos T/citología
16.
J Immunotoxicol ; 12(4): 350-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25412622

RESUMEN

Delayed-type hypersensitivity (DTH) is a T-cell-mediated immune response that may be used for immunotoxicity testing in non-clinical species. However, in some cases DTH assays using T-dependent antigens may be confounded by the production of antibodies to the antigen. The authors have previously modified a DTH assay, initially validated in the mouse, for use in juvenile rats to assess the effect of immunosuppressive drugs on the developing rat immune system. The assay measures footpad swelling induced by subcutaneous footpad injection of Candida albicans (C. albicans) derived-chitosan in rats previously sensitized with C. albicans. Antibodies to chitosan are not produced in this model. However, considerable inter-animal variability inherent in the footpad swelling assay can make it difficult to precisely quantify the magnitude of the immune response and inhibition by immunosuppressants, particularly if complete suppression is not observed. This report describes the development of an ex vivo assay to assess DTH in rats using interferon (IFN)-γ production by splenocytes, obtained from rats sensitized with C. albicans, as the quantifiable measure of the DTH response. Adult and neonatal rats administered dexamethasone (DEX), a known immunosuppressant, exhibited immunosuppression as evidenced by a reduction in ex vivo IFNγ production from splenocytes challenged with C. albicans-derived chitosan. Current data indicate that the ex vivo based DTH assay is more sensitive than the conventional footpad swelling assay due to a lower background response and the ability to detect a response as early as post-natal day (PND) 12. The ex vivo based rat DTH assay offers a highly sensitive and quantitative alternative to the footpad swelling assay for the assessment of the immunotoxic potential of drugs. The increased sensitivity of the ex vivo DTH assay may be useful for identifying smaller changes in response to immunotoxic drugs, as well as detecting responses earlier in animal development.


Asunto(s)
Candida albicans/inmunología , Candidiasis/diagnóstico , Candidiasis/inmunología , Quitosano/inmunología , Hipersensibilidad Tardía/inmunología , Animales , Quitosano/química , Femenino , Hipersensibilidad Tardía/inducido químicamente , Ratones , Ratas , Ratas Sprague-Dawley
17.
Adv Drug Deliv Rev ; 71: 15-33, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24530633

RESUMEN

Alveolar macrophage (AM) responses are commonly induced in inhalation toxicology studies, typically being observed as an increase in number or a vacuolated 'foamy' morphology. Discriminating between adaptive AM responses and adverse events during nonclinical and clinical development is a major scientific challenge. When measuring and interpreting induced AM responses, an understanding of macrophage biology is essential; this includes 'sub-types' of AMs with different roles in health and disease and mechanisms of induction/resolution of AM responses to inhalation of pharmaceutical aerosols. In this context, emerging assay techniques, the utility of toxicokinetics and the requirement for new biomarkers are considered. Risk assessment for nonclinical toxicology findings and their translation to effects in humans is discussed from a scientific and regulatory perspective. At present, when apparently adaptive macrophage-only responses to inhaled investigational products are observed in nonclinical studies, this poses a challenge for risk assessment and an improved understanding of induced AM responses to inhaled pharmaceuticals is required.


Asunto(s)
Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Macrófagos Alveolares/metabolismo , Administración por Inhalación , Aerosoles , Animales , Biomarcadores/metabolismo , Descubrimiento de Drogas/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Medición de Riesgo/métodos , Pruebas de Toxicidad/métodos
18.
J Immunotoxicol ; 10(4): 341-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23282408

RESUMEN

Establishing an in vivo cell-mediated immunity (CMI) assay, such as the delayed-type hypersensitivity (DTH) assay, has been identified as an important gap and recommended to receive highest priority for new model development in several workshops on developmental immunotoxicity. A Candida albicans DTH model has recently been developed that has the advantage over other DTH models, which use alternative sensitizing antigens, in that antigen-specific antibodies, which may interfere with the assay, are not produced. In addition, the in vivo C. albicans DTH model was demonstrated to be more sensitive in detecting immunosuppression than DTH models using keyhole limpet hemocyanin (KLH) or sheep red blood cells as antigens, as well as some ex vivo CMI assays. While KLH and sheep red blood cells are non-physiological immunogens, C. albicans is an important human pathogen. The present studies were conducted in order to optimize and validate the C. albicans DTH model for use in developmental immunotoxicity studies using juvenile rats. Three known immunosuppressive compounds with different mechanisms of action were tested in this model, cyclosprorin A (CsA), cyclophosphamide (CPS), and dexamethasone (DEX). Animals were sensitized with formalin-fixed C. albicans on postnatal day (PND) 28 and challenged with chitosan on PND 38. Drug was administered beginning on PND 23 and continued until PND 37. Exposure to each of the three immunotoxicants resulted in statistically significant decreases in the DTH response to C. albicans-derived chitosan. Decreases in footpad swelling were observed at ≥10 mg CsA/kg/day, ≥5 mg CPS/kg/day, and ≥0.03 mg DEX/kg/day. These results demonstrate that the C. albicans DTH model, optimized for use in juvenile rats, can be used to identify immunotoxic compounds, and fills the need for a sensitive in vivo CMI model for assessments of developmental immunotoxicity. Abbreviations Ab, antibody APC, antigen presenting cell BSA, bovine serum albumin C. albicans, Candida albicans CI, challenge interval CMI, cell-mediated immunity CO, challenge only CPS, cyclophosphamide CsA, cyclosporin A CTL, cytotoxic T lymphocyte DEX, dexamethasone DIT, developmental immunotoxicity DTH, delayed-type hypersensitivity ip, intraperitoneal KLH, keyhole limpet hemocyanin MLR, mixed lymphocyte reaction OVA, ovalbumin PBS, phosphate-buffered saline PND, postnatal day sc, subcutaneous SEM, standard error of the mean SRBC, sheep red blood cells.


Asunto(s)
Candida albicans/inmunología , Modelos Animales de Enfermedad , Hipersensibilidad Tardía/tratamiento farmacológico , Animales , Antígenos Fúngicos/inmunología , Bovinos , Quitosano/inmunología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Ciclosporinas/administración & dosificación , Ciclosporinas/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Femenino , Humanos , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/microbiología , Inmunidad Celular/efectos de los fármacos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Ovinos
19.
J Clin Invest ; 123(12): 5082-97, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24177422

RESUMEN

Aberrant blood vessel formation contributes to a wide variety of pathologies, and factors that regulate angiogenesis are attractive therapeutic targets. Endothelial and smooth muscle cell-derived neuropilin-like protein (ESDN) is a neuropilin-related transmembrane protein expressed in ECs; however, its potential effect on VEGF responses remains undefined. Here, we generated global and EC-specific Esdn knockout mice and demonstrated that ESDN promotes VEGF-induced human and murine EC proliferation and migration. Deletion of Esdn in the mouse interfered with adult and developmental angiogenesis, and knockdown of the Esdn homolog (dcbld2) in zebrafish impaired normal vascular development. Loss of ESDN in ECs blunted VEGF responses in vivo and attenuated VEGF-induced VEGFR-2 signaling without altering VEGF receptor or neuropilin expression. Finally, we found that ESDN associates with VEGFR-2 and regulates its complex formation with negative regulators of VEGF signaling, protein tyrosine phosphatases PTP1B and TC-PTP, and VE-cadherin. These findings establish ESDN as a regulator of VEGF responses in ECs that acts through a mechanism distinct from neuropilins. As such, ESDN may serve as a therapeutic target for angiogenesis regulation.


Asunto(s)
Endotelio Vascular/fisiología , Proteínas de la Membrana/fisiología , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Antígenos CD/fisiología , Vasos Sanguíneos/embriología , Cadherinas/fisiología , Células Cultivadas , Oído Externo/irrigación sanguínea , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/fisiopatología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropilinas/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Vasos Retinianos/crecimiento & desarrollo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
20.
J Immunotoxicol ; 9(2): 210-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22428536

RESUMEN

The development and regulatory approval of immunomodulatory pharmaceuticals to treat many human diseases has increased significantly over the last two decades. As discussed by FDA and ICH guidelines, all human pharmaceuticals in development should be evaluated for potential adverse effects on the immune system. Developmental immunotoxicology (DIT) focuses on the concern that early-life (during pre-/post-natal development) exposure to agents which target the immune system may result in enhanced susceptibility to immune-related disease (e.g., infection, autoimmunity, and cancer, particularly leukemia) compared to adults, unique effects not observed in adults, or more persistent effects in comparison to those following adult exposure. This article provides a substantive review of the literature and presents detailed considerations for DIT testing strategies with a specific focus on pharmaceuticals and biopharmaceuticals. In this regard, differences between small molecule and large molecule therapeutics will be considered, along with recommendations for best practices in the assessment of DIT during drug development. In addition, gaps in the DIT knowledge base and current testing strategies are identified. Finally, a summary of an ILSI-HESI-ITC sponsored Workshop conducted in 2010, entitled 'Developmental Immunotoxicity Testing of Pharmaceuticals' will be presented. This Workshop consisted of participants from the pharmaceutical, biotechnology, academic, and regulatory sectors, where many of the issues relating to DIT outlined in this review were discussed, key points of consensus reached, and current gaps in the science identified.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Factores Inmunológicos/toxicidad , Exposición Materna/efectos adversos , Animales , Educación , Femenino , Directrices para la Planificación en Salud , Sistema Inmunológico/embriología , Factores Inmunológicos/clasificación , Preparaciones Farmacéuticas/clasificación , Pruebas de Toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA