Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Reprod Toxicol ; 121: 108468, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37666285

RESUMEN

RTH258 (brolucizumab) is a humanized single chain antibody fragment, the smallest functional unit of an antibody designed to target vascular endothelial growth factor in angiogenic retinal disease. To further understand the safe use of RTH258, this study assessed the potential impact of intravitreal RTH258 on pre- and postnatal development in the offspring of cynomolgus monkeys following administration to the mother. Three groups of 16 pregnant females were included: a low dose group (RTH258 3 mg/50 µl [60 mg/ml]), a high dose group (RTH258 6 mg/50 µl [120 mg/ml]), and a control group. Maternal animals were administered a single injection of 50 µl in the right eye once every four weeks. Animals were observed daily and detailed observations were collected before and after the first dose, and then weekly thereafter. Following parturition, observations of infants included external, morphological, and ophthalmic examinations; neurobehavioral test battery; grip strength; and skeletal development. Blood samples for hematology, coagulation, and clinical chemistry were collected from non-fasted maternal and infant animals. No RTH258-related deaths occurred in maternal dams or infants. No RTH258-related clinical observations were noted in maternal animals or in surviving infants - there were no changes in gestation length; pregnancy loss; deaths; body weight/weight change; infant grip strength; infant external, morphological, or skeletal evaluations; ophthalmoscopy or neurobehavioral observations; or clinical pathology parameters. RTH258 had no impact on pregnancy or parturition; embryo-fetal development; or survival, growth, or postnatal development of offspring when administered via repeated intravitreal administration.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Factor A de Crecimiento Endotelial Vascular , Humanos , Embarazo , Animales , Femenino , Macaca fascicularis , Anticuerpos Monoclonales Humanizados/farmacología , Parto
2.
Reprod Toxicol ; 91: 142-146, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31698003

RESUMEN

The aim of this study was to directly test and measure in vivo, if placental transfer of monoclonal antibodies takes place in pregnant Göttingen Minipigs to assess their suitability for reproductive assessment of therapeutic monoclonal antibodies. Simulect®, an approved anti CD25 (anti IL-2 receptor alpha) chimeric monoclonal IgG1 antibody, was used as a model monoclonal antibody. Maternal systemic exposure and potential placental transfer of Simulect® to fetuses were investigated following 4 weekly bolus intravenous administration of 5.0 mg/kg from gestation day (GD) 79 or 80 (e.g GD 79, 86, 93 and 100) and with terminal Caesarean section on GD 108 or GD 109 respectively. Results clearly showed exposure in maternal animals, detectable compound in the amniotic fluid from one out of 9 maternal animals, but no exposure in fetuses confirming absence of placental transfer of the selected model antibody Simulect® in minipigs. The absence of Simulect® in the fetuses further supports that the presence of Simulect® in the amniotic fluid in one maternal animal was likely due to contamination with maternal blood during sampling. The demonstrated absence of fetal exposure clearly indicates that, the minipig is not a suitable species for conduct of reproductive toxicity studies with monoclonal antibodies.


Asunto(s)
Basiliximab/farmacocinética , Inmunosupresores/farmacocinética , Intercambio Materno-Fetal , Líquido Amniótico/química , Animales , Basiliximab/sangre , Femenino , Sangre Fetal/química , Inmunosupresores/sangre , Embarazo , Porcinos , Porcinos Enanos
3.
Birth Defects Res B Dev Reprod Toxicol ; 107(6): 243-257, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28032463

RESUMEN

The combination of artemether plus lumefantrine is a type of artemisinin-based combination therapy (ACT) recommended by the World Health Organization for uncomplicated falciparum malaria except in the first trimester of pregnancy. The first trimester restriction was based on the marked embryotoxicity in animals (including embryo death and cardiac and skeletal malformations) of artemisinins such as artesunate, dihydroartemisinin, and artemether. Before recommending ACTs for use in the first trimester, the World Health Organization has requested that all information relevant to the assessment of risk of ACTs to the embryo be made available to the public. This report describes the results of embryo-fetal development studies of artemether alone, lumefantrine alone, and the combination in rats and rabbits as well as toxicokinetic studies of lumefantrine in pregnant rabbits. The developmental no-effect levels for lumefantrine were 300 mg/kg/day in rats (based on a 25% decrease in litter size at 1000 mg/kg/day) and 1000 mg/kg/day in rabbits. The calculated safety margins based on human equivalent dose and plasma Cmax and AUC values were in the range of 2.5- to 17-fold. The developmental no-effect levels for artemether were 3 mg/kg/day in rats and 25 mg/kg/day in rabbits. Lumefantrine caused no teratogenicity and was not a potent embryotoxin in rats and rabbits. Expected artemisinin-like findings were seen with artemether alone and with artemether/lumefantrine combined except that no malformations were observed. There were no findings in pregnant rats and rabbits that would cause increased concern for the use of artemether-lumefantrine in the first trimester compared to other ACTs.


Asunto(s)
Antimaláricos/toxicidad , Artemisininas/toxicidad , Etanolaminas/toxicidad , Fluorenos/toxicidad , Pruebas de Toxicidad , Animales , Arteméter , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Lumefantrina , Malaria Falciparum/tratamiento farmacológico , Masculino , Exposición Materna/efectos adversos , Embarazo , Conejos , Ratas , Ratas Sprague-Dawley , Toxicocinética
4.
Regul Toxicol Pharmacol ; 77: 100-8, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26930635

RESUMEN

During the past two decades the use and refinements of imaging modalities have markedly increased making it possible to image embryos and fetuses used in pivotal nonclinical studies submitted to regulatory agencies. Implementing these technologies into the Good Laboratory Practice environment requires rigorous testing, validation, and documentation to ensure the reproducibility of data. A workshop on current practices and regulatory requirements was held with the goal of defining minimal criteria for the proper implementation of these technologies and subsequent submission to regulatory agencies. Micro-computed tomography (micro-CT) is especially well suited for high-throughput evaluations, and is gaining popularity to evaluate fetal skeletons to assess the potential developmental toxicity of test agents. This workshop was convened to help scientists in the developmental toxicology field understand and apply micro-CT technology to nonclinical toxicology studies and facilitate the regulatory acceptance of imaging data. Presentations and workshop discussions covered: (1) principles of micro-CT fetal imaging; (2) concordance of findings with conventional skeletal evaluations; and (3) regulatory requirements for validating the system. Establishing these requirements for micro-CT examination can provide a path forward for laboratories considering implementing this technology and provide regulatory agencies with a basis to consider the acceptability of data generated via this technology.


Asunto(s)
Anomalías Inducidas por Medicamentos/diagnóstico por imagen , Huesos/diagnóstico por imagen , Biología Evolutiva/métodos , Feto/diagnóstico por imagen , Pruebas de Toxicidad/métodos , Microtomografía por Rayos X , Animales , Huesos/anomalías , Huesos/efectos de los fármacos , Consenso , Biología Evolutiva/normas , Feto/anomalías , Feto/efectos de los fármacos , Guías como Asunto , Humanos , Variaciones Dependientes del Observador , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Pruebas de Toxicidad/normas , Microtomografía por Rayos X/normas
5.
Reprod Toxicol ; 59: 22-30, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26854737

RESUMEN

Small molecule pharmaceutical products are assumed to reach concentrations in semen similar to those in blood plasma. Exposure modeling for these small-molecule products in humans assumes a daily dose of 5mL of semen and 100% absorption from the vagina with distribution to the conceptus through the maternal systemic circulation. Monoclonal antibody drugs are present in semen at concentrations about 2% or less of those in blood, and the modeling used for small molecules will over-estimate the possibility of conceptus exposure to immunoglobulins. It is not known whether peptide products reach semen, but in general peptide medications are destroyed by vaginal peptidases, and conceptus exposure is predicted to be minimal. Theoretical exposure routes to pharmaceuticals that might result in exposure of the conceptus greater than that of maternal systemic exposures include direct access through the cervical canal, adsorption to sperm for carriage into the oocyte, and direct delivery from the vaginal veins or lymphatics to the uterine artery. There is some evidence for direct access to the uterus for progesterone, terbutaline, and danazol, but the evidence does not involve exposures during pregnancy in most instances. Studies in mice, rats, rabbits, and monkeys do not suggest that exposure to small molecule pharmaceuticals in semen imposes risks to the conceptus beyond those that can be predicted using modeling of systemic maternal exposure. Monoclonal antibody and peptide exposure in semen does not pose a significant risk to the conceptus.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Embrión de Mamíferos/metabolismo , Feto/metabolismo , Péptidos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Semen/metabolismo , Vagina/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/toxicidad , Transporte Biológico , Embrión de Mamíferos/efectos de los fármacos , Femenino , Feto/efectos de los fármacos , Haplorrinos , Humanos , Masculino , Exposición Materna , Ratones , Modelos Animales , Modelos Biológicos , Exposición Paterna , Péptidos/química , Péptidos/toxicidad , Permeabilidad , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Conejos , Medición de Riesgo , Absorción Vaginal
6.
Reprod Toxicol ; 58: 213-21, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26545974

RESUMEN

Small molecule pharmaceutical products are assumed to reach concentrations in semen similar to those in blood plasma. Exposure modeling for these small-molecule products in humans assumes a daily dose of 5mL of semen and 100% absorption from the vagina with distribution to the conceptus through the maternal systemic circulation. Monoclonal antibody drugs are present in semen at concentrations about 2% or less of those in blood, and the modeling used for small molecules will over-estimate the possibility of conceptus exposure to immunoglobulins. It is not known whether peptide products reach semen, but in general peptide medications are destroyed by vaginal peptidases, and conceptus exposure is predicted to be minimal. Theoretical exposure routes to pharmaceuticals that might result in exposure of the conceptus greater than that of maternal systemic exposures include direct access through the cervical canal, adsorption to sperm for carriage into the oocyte, and direct delivery from the vaginal veins or lymphatics to the uterine artery. There is some evidence for direct access to the uterus for progesterone, terbutaline, and danazol, but the evidence does not involve exposures during pregnancy in most instances. Studies in mice, rats, rabbits, and monkeys do not suggest that exposure to small molecule pharmaceuticals in semen imposes risks to the conceptus beyond those that can be predicted using modeling of systemic maternal exposure. Monoclonal antibody and peptide exposure in semen does not pose a significant risk to the conceptus.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Cuello del Útero/metabolismo , Embrión de Mamíferos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Proteínas/metabolismo , Semen/metabolismo , Vagina/metabolismo , Animales , Anticuerpos Monoclonales/efectos adversos , Transporte Biológico , Cuello del Útero/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Femenino , Humanos , Masculino , Modelos Biológicos , Embarazo , Proteínas/efectos adversos , Medición de Riesgo , Factores de Riesgo , Especificidad de la Especie , Vagina/efectos de los fármacos
7.
Reprod Toxicol ; 48: 138-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24887033

RESUMEN

A ß-actin-luc transgenic mouse model was used to evaluate whether embryo-fetal exposure could occur after intravaginal administration of a compound. A bioluminescent substrate, d-luciferin, was delivered intravaginally to mimic compound exposure to the female reproductive track and the embryo-fetus. Bioluminescence was observed throughout the reproductive tract during diestrus, but not during estrus, 2-5min after intravaginal d-luciferin administration to female ß-actin-luc mice. Intravaginal administration of d-luciferin to wild-type females mated with male ß-actin-luc mice indicated that the substrate reached the developing embryo-fetus, with bioluminescence corresponding to transgene expression in the embryo-fetus. d-Luciferin substrate rapidly reached the embryo-fetus regardless of the administration route (intravaginal, intraperitoneal, subcutaneous, or intravenous). Vaginal ligation appeared to block at least some direct exposure to the embryo-fetus, but did not prevent d-luciferin from eventually reaching the embryo-fetus. Additional work will be necessary to form the basis for a reliable assessment of the human risk for male-mediated teratogenicity.


Asunto(s)
Benzotiazoles/administración & dosificación , Benzotiazoles/farmacocinética , Embrión de Mamíferos/metabolismo , Feto/metabolismo , Administración Intravaginal , Animales , Ciclo Estral , Femenino , Luciferasas/genética , Luminiscencia , Masculino , Ratones Transgénicos , Imagen Óptica , Embarazo
9.
J Cell Biochem ; 87(3): 342-59, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12397616

RESUMEN

Cartilage formation in the embryonic limb is presaged by a cellular condensation phase that is mediated by both cell-cell and cell-matrix interactions. N-Cadherin, a Ca(2+)-dependent cell-cell adhesion molecule, is expressed at higher levels in the condensing mesenchyme, followed by down-regulation upon chondrogenic differentiation, strongly suggesting a functional role in the cellular condensation process. To further examine the role of N-cadherin, we have generated expression constructs of wild type and two deletion mutants (extracellular and intracellular) of N-cadherin in the avian replication-competent, RCAS retrovirus, and transfected primary chick limb mesenchymal cell cultures with these constructs. The effects of altered, sustained expression of N-cadherin and its mutant forms on cellular condensation, on the basis of peanut agglutinin (DNA) staining, and chondrogenesis, based on expression of chondrocyte phenotypic markers, were characterized. Cellular condensation was relatively unchanged in cultures overexpressing wild type N-cadherin, compared to controls on all days in culture. However, expression of either of the deletion mutant forms of N-cadherin resulted in decreased condensation, with the extracellular deletion mutant demonstrating the most severe inhibition, suggesting a requirement for N-cadherin mediated cell-cell adhesion and signaling in cellular condensation. Subsequent chondrogenic differentiation was also affected in all cultures overexpressing the N-cadherin constructs, on the basis of metabolic sulfate incorporation, the presence of the cartilage matrix proteins collagen type II and cartilage proteoglycan link protein, and alcian blue staining of the matrix. The characteristics of the cultures suggest that the N-cadherin mutants disrupt proper cellular condensation and subsequent chondrogenesis, while the cultures overexpressing wild type N-cadherin appear to condense normally, but are unable to proceed toward differentiation, possibly due to the prolonged maintenance of increased cell-cell adhesiveness. Thus, spatiotemporally regulated N-cadherin expression and function, at the level of both homotypic binding and linkage to the cytoskeleton, is required for chondrogenesis of limb mesenchymal cells.


Asunto(s)
Cadherinas/fisiología , Agregación Celular/fisiología , Condrogénesis/fisiología , Esbozos de los Miembros/metabolismo , Mesodermo/metabolismo , Animales , Cadherinas/biosíntesis , Cartílago/metabolismo , Embrión de Pollo , Colágeno Tipo II/metabolismo , Técnicas de Cultivo , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Esbozos de los Miembros/citología , Mesodermo/citología , Proteoglicanos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Factores de Tiempo , Transactivadores/metabolismo , Transfección , beta Catenina
10.
Dev Dyn ; 225(2): 195-204, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12242719

RESUMEN

During embryonic limb development, cartilage formation is presaged by a crucial mesenchymal cell condensation phase. N-Cadherin, a Ca2+ -dependent cell-cell adhesion molecule, is expressed in embryonic chick limb buds in a spatiotemporal pattern suggestive of its involvement during cellular condensation; functional blocking of N-cadherin homotypic binding, by using a neutralizing monoclonal antibody, results in perturbed chondrogenesis in vitro and in vivo. In high-density micromass cultures of embryonic limb mesenchymal cells, N-cadherin expression level is high during days 1 and 2, coincident with active cellular condensation, and decreases upon overt chondrogenic differentiation from day 3 on. In this study, we have used a transfection approach to evaluate the effects of gain- and loss-of-function expression of N-cadherin constructs on mesenchymal condensation and chondrogenesis in vitro. Chick limb mesenchymal cells were transfected by electroporation with recombinant expression plasmids encoding wild-type or two mutant extracellular/cytoplasmic deletion forms of N-cadherin. Expression of the transfected N-cadherin forms showed a transient profile, being high on days 1-2 of culture, and decreasing by day 3, fortuitously coincident with the temporal profile of endogenous N-cadherin gene expression. Examined by means of peanut agglutinin (PNA) staining for condensing precartilage mesenchymal cells, cultures overexpressing wild-type N-cadherin showed enhanced cellular condensation on culture days 2 and 3, whereas expression of the deletion mutant forms (extracellular/cytoplasmic) of N-cadherin resulted in a decrease in PNA staining, suggesting that a complete N-cadherin protein is required for normal cellular condensation to occur. Subsequent chondrogenesis was also affected. Cultures overexpressing the wild-type N-cadherin protein showed enhanced chondrogenesis, indicated by increased production of cartilage matrix (sulfated proteoglycans, collagen type II, and cartilage proteoglycan link protein), as well as increased cartilage nodule number and size of individual nodules, compared with control cultures and cultures transfected with either of the two mutant N-cadherin constructs. These results demonstrate that complete N-cadherin function, at the levels of both extracellular homotypic binding and cytoplasmic linkage to the cytoskeleton by means of the catenin complex, is required for chondrogenesis by mediating functional mesenchymal cell condensation.


Asunto(s)
Cadherinas/fisiología , Condrocitos/metabolismo , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Azul Alcián/farmacología , Animales , Anticuerpos Monoclonales/metabolismo , Adhesión Celular , Supervivencia Celular , Embrión de Pollo , Citoplasma/metabolismo , Citoesqueleto/metabolismo , Hibridomas/metabolismo , Immunoblotting , Inmunohistoquímica , Plásmidos/metabolismo , Ratas , Sulfatos/metabolismo , Factores de Tiempo , Transfección
11.
Exp Cell Res ; 273(2): 197-203, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11822875

RESUMEN

Wnt signaling has been implicated in the regulation of limb mesenchymal chondrogenesis. In this study, we have analyzed the molecular mechanism of Wnt-7a inhibition of chondrogenic differentiation by examining the involvement of mitogen-activated protein kinase (MAPK) pathways, i.e., Erk and p38. The combination of Wnt-7a misexpression and Erk inhibition partially recovers Wnt-7a inhibition of chondrogenic differentiation, whereas the combination of Wnt-7a misexpression and p38 inhibition acts in a synergistic chondro-inhibitory fashion. Although Wnt-7a misexpression has no direct effect on Erk signaling, it increases the activity of one of the ultimate targets of the MAPK pathway, c-jun, a major component of the activator protein-1 (AP-1) transcription factor complex. In addition, Wnt-7a misexpression enhances the activity of an AP-1 promoter-luciferase reporter construct by approximately 2.3-fold in vitro. Interestingly, misexpression of wild-type N-cadherin in these micromass cultures suppresses the activity of the same AP-1 promoter by approximately 40%, whereas misexpression of an extracellular 390-amino-acid N-terminal deletion mutant of N-cadherin has a stimulatory effect on the AP-1 promoter activity by approximately 2.6-fold. Thus, our results suggest that at least a part of the chondro-inhibitory effect of Wnt-7a misexpression may involve AP-1 transcription factor stimulation. Furthermore, a very tightly regulated level of AP-1 activity is necessary for the process of limb mesenchymal chondrogenesis, and signals from Wnt-ligands (e.g., Wnt-7a), cell adhesion molecules (e.g., N-cadherin), and MAPK pathways (e.g., Erk and p38) are interactively involved in this regulation.


Asunto(s)
Proteínas Aviares , Cadherinas/metabolismo , Condrogénesis/fisiología , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción AP-1/genética , Animales , Cadherinas/genética , Adhesión Celular , Embrión de Pollo , Activación Enzimática , Mesodermo/citología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Wnt , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...