Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Pathol ; 47(4): 444-460, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30898082

RESUMEN

If nonhuman primates represent the only relevant species for nonclinical safety evaluation of biotechnology-derived products, male and female fertility effects can be assessed in repeat dose toxicity studies given that sexually mature monkeys are used. This opinion piece provides recommendations for determining sexual maturity and when/how fertility assessments should be conducted in the cynomolgus monkey. Male sexual maturity should be proven by presence of sperm in a semen sample, female sexual maturity by at least two consecutive menstrual bleedings. As per regulatory guidance, default parameters for an indirect assessment of fertility in both sexes are reproductive organ weight and histopathology. Beyond default parameters, daily vaginal swabs are recommended for females, and for males, it is recommended to include blood collections (for potential analysis of reproductive hormones), testis volume sonography, and collection of frozen testis samples at necropsy. Only if there is a cause for concern, blood collection for potential reproductive hormone analysis should be conducted in females and semen analysis in males. In principle, adverse reproductive effects can be detected within 4 weeks of test article administration, depending on study design and reproductive end point chosen. Therefore, there are options for addressing reproductive toxicity aspects with studies of less than 3 months dosing duration. *This is an opinion article submitted to the Toxicologic Pathology Forum. It represents the views of the authors. It does not constitute an official position of the Society of Toxicologic Pathology, British Society of Toxicological Pathology, or European Society of Toxicologic Pathology, and the views expressed might not reflect the best practices recommended by these Societies. This article should not be construed to represent the policies, positions, or opinions of their respective organizations, employers, or regulatory agencies.


Asunto(s)
Fertilidad/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Pruebas de Toxicidad/métodos , Pruebas de Toxicidad/normas , Animales , Femenino , Guías como Asunto , Macaca fascicularis , Masculino
2.
Trends Genet ; 30(11): 482-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24954183

RESUMEN

The success of personalized medicine rests on understanding the genetic variation between individuals. Thus, as medical practice evolves and variation among individuals becomes a fundamental aspect of clinical medicine, a thorough consideration of the genetic and genomic information concerning the animals used as models in biomedical research also becomes critical. In particular, nonhuman primates (NHPs) offer great promise as models for many aspects of human health and disease. These are outbred species exhibiting substantial levels of genetic variation; however, understanding of the contribution of this variation to phenotypes is lagging behind in NHP species. Thus, there is a pivotal need to address this gap and define strategies for characterizing both genomic content and variability within primate models of human disease. Here, we discuss the current state of genomics of NHP models and offer guidelines for future work to ensure continued improvement and utility of this line of biomedical research.


Asunto(s)
Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Variación Genética , Genómica/métodos , Animales , Investigación Biomédica/tendencias , Estudio de Asociación del Genoma Completo/métodos , Estudio de Asociación del Genoma Completo/tendencias , Genómica/tendencias , Humanos , Medicina de Precisión/métodos , Medicina de Precisión/tendencias , Primates , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de ADN/tendencias
3.
Crit Rev Toxicol ; 47(5): 402-414, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27766926

RESUMEN

A database of embryo-fetal developmental toxicity (EFDT) studies of 379 pharmaceutical compounds in rat and rabbit was analyzed for species differences based on toxicokinetic parameters of area under the curve (AUC) and maximum concentration (Cmax) at the developmental lowest adverse effect level (dLOAEL). For the vast majority of cases (83% based on AUC of n = 283), dLOAELs in rats and rabbits were within the same order of magnitude (less than 10-fold different) when compared based on available data on AUC and Cmax exposures. For 13.5% of the compounds the rabbit was more sensitive and for 3.5% of compounds the rat was more sensitive when compared based on AUC exposures. For 12% of the compounds the rabbit was more sensitive and for 1.3% of compounds the rat was more sensitive based on Cmax exposures. When evaluated based on human equivalent dose (HED) conversion using standard factors, the rat and rabbit were equally sensitive. The relative extent of embryo-fetal toxicity in the presence of maternal toxicity was not different between species. Overall effect severity incidences were distributed similarly in rat and rabbit studies. Individual rat and rabbit strains did not show a different general distribution of systemic exposure LOAELs as compared to all strains combined for each species. There were no apparent species differences in the occurrence of embryo-fetal variations. Based on power of detection and given differences in the nature of developmental effects between rat and rabbit study outcomes for individual compounds, EFDT studies in two species have added value over single studies.


Asunto(s)
Embrión de Mamíferos/fisiología , Desarrollo Embrionario/efectos de los fármacos , Preparaciones Farmacéuticas , Animales , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Embrión de Mamíferos/efectos de los fármacos , Conejos , Ratas
4.
Int J Toxicol ; 36(4): 325-339, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28466670

RESUMEN

In support of a clinical trial in the pediatric population, available nonclinical and clinical data provide input on the study design and safety monitoring considerations. When the existing data are lacking to support the safety of the planned pediatric clinical trial, a juvenile animal toxicity study is likely required. Usually a single relevant species, preferably a rodent, is chosen as the species of choice, while a nonrodent species can be appropriate when scientifically justified. Juvenile toxicology studies, in general, are complicated both conceptually and logistically. Development in young animals is a continuous process with different organs maturing at different rates and time. Structural and functional maturational differences have been shown to affect drug safety. Key points to consider in conducting a juvenile toxicology study include a comparative development of the organ systems, differences in the pharmacokinetics/absorption, distribution, metabolism, excretion (PK/ADME) profiles of the drug between young animal and child, and logistical requirement in the juvenile study design. The purpose of this publication is to note pertinent points to consider when designing and conducting juvenile toxicology studies and to aid in future modifications and enhancements of these studies to enable a superior predictability of safety of medicines in the pediatric population.


Asunto(s)
Animales de Laboratorio/crecimiento & desarrollo , Animales de Laboratorio/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Proyectos de Investigación , Pruebas de Toxicidad/normas , Factores de Edad , Animales , Evaluación Preclínica de Medicamentos/normas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Guías como Asunto , Humanos , Especificidad de la Especie
5.
Crit Rev Toxicol ; 46(10): 900-910, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27848393

RESUMEN

Regulatory non-clinical safety testing of human pharmaceuticals typically requires embryo-fetal developmental toxicity (EFDT) testing in two species (one rodent and one non-rodent). The question has been raised whether under some conditions EFDT testing could be limited to one species, or whether the testing in a second species could be decided on a case-by-case basis. As part of a consortium initiative, we built and queried a database of 379 compounds with EFDT studies (in both rat and rabbit animal models) conducted for marketed and non-marketed pharmaceuticals for their potential for adverse developmental and maternal outcomes, including EFDT incidence and the nature and severity of adverse findings. Manifestation of EFDT in either one or both species was demonstrated for 282 compounds (74%). EFDT was detected in only one species (rat or rabbit) in almost a third (31%, 118 compounds), with 58% (68 compounds) of rat studies and 42% (50 compounds) of rabbit studies identifying an EFDT signal. For 24 compounds (6%), fetal malformations were observed in one species (rat or rabbit) in the absence of any EFDT in the second species. In general, growth retardation, fetal variations, and malformations were more prominent in the rat, whereas embryo-fetal death was observed more often in the rabbit. Discordance across species may be attributed to factors such as maternal toxicity, study design differences, pharmacokinetic differences, and pharmacologic relevance of species. The current analysis suggests that in general both species are equally sensitive on the basis of an overall EFDT LOAEL comparison, but selective EFDT toxicity in one species is not uncommon. Also, there appear to be species differences in the prevalence of various EFDT manifestations (i.e. embryo-fetal death, growth retardation, and dysmorphogenesis) between rat and rabbit, suggesting that the use of both species has a higher probability of detecting developmental toxicants than either one alone.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Sustancias Peligrosas/toxicidad , Modelos Animales , Pruebas de Mutagenicidad/métodos , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos , Animales , Conejos , Ratas
6.
Birth Defects Res B Dev Reprod Toxicol ; 98(1): 91-103, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23349029

RESUMEN

This study was designed to determine the effects of Compound A on spermatogenesis including assessment of inhibin B levels and on fertility in the male rat over a 15 to 19 weeks treatment and a 19 weeks treatment-free period in control and 30, 60, and 180 mg/kg dose groups (n = 22/group). Compound A in a dose-dependent manner induced various degrees of spermatogenic alterations compatible with Sertoli cells being the primary target, for example, inter- and intracellular Sertoli cell vacuolization and altered cellular morphology followed by germ cell degeneration and marked reduction of epididymidal sperm numbers. Blood-testis barrier remained intact (electron microscopy and hyperosmotic fixation test) until germ cells disappeared. Mating behavior and weights of androgen-dependent prostate and seminal vesicles remained unaffected. Inhibin B levels correlated only with moderate to severe spermatogenic alterations. Ten animals with inhibin B levels below detection limit were encountered and five of these animals were fertile in week 19 but following 19 weeks without treatment, another five animals were rendered infertile and inhibin B levels remained undetectable. In the rat, inhibin B only reflects major spermatogenic alterations and markedly reduced inhibin B levels might indicate irreversibility of these alterations and even infertility.


Asunto(s)
Inhibinas/sangre , Células de Sertoli/patología , Espermatogénesis , Animales , Biomarcadores/sangre , Peso Corporal , Epidídimo/patología , Femenino , Masculino , Tamaño de los Órganos , Ratas , Reproducción , Epitelio Seminífero/patología , Células de Sertoli/metabolismo , Células de Sertoli/ultraestructura , Espermatozoides/metabolismo
7.
Birth Defects Res B Dev Reprod Toxicol ; 95(6): 431-43, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23212752

RESUMEN

BACKGROUND: ABT-874 is an anti-IL-12/23 monoclonal antibody that binds to the p40 subunit of human IL-12 and IL-23. As part of its preclinical safety assessment, studies were conducted to assess its potential effects on pre- and postnatal development in cynomolgus monkeys. METHODS: In the embryo-fetal development studies, ABT-874 was administered once weekly subcutaneously to adult female cynomolgus monkeys at doses of 0, 5, 25, or 100 mg/kg during gestation days (GD) 20 to 48. Fetuses were examined for external, visceral, and skeletal development on GD 100 or 150. In the pre- and postnatal study, ABT-874 was administered once weekly subcutaneously to adult female cynomolgus monkeys at doses of 10, 50, or 200 mg/kg from GD 20 through postpartum day 182. Infants were examined from birth up to 9 months of age for morphological and functional development. Potential effects on the infant immune system were evaluated by immunophenotyping of peripheral blood lymphocytes and by T-dependent antibody response to KLH. RESULTS: There was no ABT-874-related maternal toxicity or adverse effects on fetuses or infants. ABT-874 was present in maternal and fetal serum at GD 100 and 150, and in infant serum through day 63 postbirth. ABT-874 was also present at low levels in breast milk through postpartum day 175. CONCLUSIONS: Exposure of cynomolgus monkey fetuses and infants to ABT-874 had no adverse effects on embryo-fetal or postnatal development.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Macaca fascicularis/fisiología , Exposición Materna/efectos adversos , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales Humanizados , Formación de Anticuerpos/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Hemocianinas/farmacología , Sistema Inmunológico/efectos de los fármacos , Inyecciones Subcutáneas , Lactancia/efectos de los fármacos , Linfocitos/efectos de los fármacos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Pruebas de Toxicidad
8.
Birth Defects Res B Dev Reprod Toxicol ; 95(6): 421-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23213064

RESUMEN

BACKGROUND: ABT-874 is an anti-IL-12/23 monoclonal antibody that binds to the p40 subunit of human IL-12 and IL-23. As part of its preclinical safety assessment, studies were conducted to asses its potential effects on the reproductive system in male and female cynomolgus monkeys. METHODS: Sexually mature male cynomolgus monkeys (n = 6/group) were administered once weekly subcutaneous doses of 0, 5, 25, or 100 mg/kg ABT-874 for 13 weeks. Four monkeys/group were necropsied at the end of the 13-week dosing period and two monkeys/group were necropsied following an 8-week recovery period. Endpoints assessed in these males included sperm parameters such as sperm count and morphology, male reproductive hormones, and testes histopathology. Sexually mature female cynomolgus monkeys (n = 6/group) were administered subcutaneous doses of 0, 5, 25, or 100 mg/kg/week ABT-874 for two menstrual cycles, and recovery was subsequently assessed in each of these animals over two menstrual cycles. Endpoints assessed in these females included menses and reproductive hormone levels. RESULTS: In both the male and female fertility studies, there were no unscheduled deaths and there was no evidence of toxicity. In male monkeys, there were no ABT-874-related effects on sperm count or motility, histopathology of the testes or effects on testosterone and inhibin B levels. In addition, menstrual cycle length, progesterone, 17ß-estradiol, and luteinizing hormone levels in female monkeys were comparable among control and ABT-874-treated groups. CONCLUSIONS: These results demonstrate that ABT-874 had no adverse effects on reproductive hormones or fertility parameters in male or female cynomolgus monkeys.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Fertilidad/efectos de los fármacos , Macaca fascicularis , Ovario/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Femenino , Fertilidad/fisiología , Infertilidad/inducido químicamente , Infertilidad/fisiopatología , Inyecciones Subcutáneas , Hormona Luteinizante/sangre , Macaca fascicularis/fisiología , Masculino , Ciclo Menstrual/efectos de los fármacos , Ovario/metabolismo , Progesterona/sangre , Recuento de Espermatozoides , Espermatozoides/efectos de los fármacos , Espermatozoides/fisiología , Testículo/patología , Pruebas de Toxicidad
9.
Birth Defects Res C Embryo Today ; 93(4): 324-33, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22271681

RESUMEN

The enhanced pre- and postnatal (ePPND) study design has been developed in response to new scientific knowledge and subsequent guideline changes, that is, ICH M3(R2) and ICH S6(R1). The design changes were basically driven by the experiences obtained during preclinical development of biopharmaceuticals. The ePPND concept typically does not apply to pharmaceuticals. In essence, the ePPND design is a PPND study in which key elements of an embryofetal development (EFD) study are being investigated in newborns and infants rather than in the fetus. The current relevant nonhuman primate model is the cynomolgus monkey. The ICH S6(R1) has reached step 4 during June 2011 and provides detailed recommendations on various parameters and the conduct of an ePPND study. By the time this article is written, it appears that for monoclonal antibodies, the ePPND study is the preferred approach although ICH S6(R1) also leaves options for modified EFD and PPND study concepts. Our data also demonstrate that social housing is feasible for developmental toxicity studies in the cynomolgus monkey model.


Asunto(s)
Alternativas al Uso de Animales , Anticuerpos Monoclonales/toxicidad , Modelos Animales de Enfermedad , Macaca fascicularis , Proyectos de Investigación/tendencias , Animales , Animales Recién Nacidos , Guías como Asunto , Humanos , Lactante , Recién Nacido , Primates , Pruebas de Toxicidad
10.
Birth Defects Res B Dev Reprod Toxicol ; 92(4): 359-80, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21770023

RESUMEN

Evaluation of pharmaceutical agents in children is now conducted earlier in the drug development process. An important consideration for this pediatric use is how to assess and support its safety. This article is a collaborative effort of industry toxicologists to review strategies, challenges, and current practice regarding preclinical safety evaluations supporting pediatric drug development with biopharmaceuticals. Biopharmaceuticals include a diverse group of molecular, cell-based or gene therapeutics derived from biological sources or complex biotechnological processes. The principles of preclinical support of pediatric drug development for biopharmaceuticals are similar to those for small molecule pharmaceuticals and in general follow the same regulatory guidances outlined by the Food and Drug Administration and European Medicines Agency. However, many biopharmaceuticals are also inherently different, with limited species specificity or immunogenic potential which may impact the approach taken. This article discusses several key areas to aid in the support of pediatric clinical use, study design considerations for juvenile toxicity studies when they are needed, and current practices to support pediatric drug development based on surveys specifically targeting biopharmaceutical development.


Asunto(s)
Animales de Laboratorio/crecimiento & desarrollo , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Drogas en Investigación , Necesidades y Demandas de Servicios de Salud , Pediatría/legislación & jurisprudencia , Pruebas de Toxicidad , Animales , Investigación Biomédica/legislación & jurisprudencia , Niño , Humanos , Industrias , Modelos Animales
11.
Birth Defects Res B Dev Reprod Toxicol ; 89(3): 175-87, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20544806

RESUMEN

BACKGROUND: This work evaluates pregnancy and infant loss in 1,069 vehicle-treated cynomolgus monkeys from 78 embryo-fetal development (EFD) studies and 14 pre-postnatal development (PPND) studies accrued during 1981-2007. METHODS: Losses were analysed by survival function and hazard ratio using logistic regression for influence of year, study type (e.g., dose duration), and test item route of administration (ig, im, iv, sc). RESULTS: Neither study type nor route of dosing affected pregnancy outcome. Losses were higher pre-1990 (104 losses/347 pregnancies) compared to 1990 onwards (94 losses/722 pregnancies). Losses were greatest before gestation day 50 and at parturition. Using post-1989 data, Monte-Carlo simulations of pregnancy outcomes were created. The power associated with the comparison of vehicle survival curves and simulated adverse survival curves was examined. This showed that EFD studies with initial vehicle group sizes of 16 and 20 have an 80% probability of having 13 and 16 ongoing pregnancies at gestational day 100, respectively. For PPND studies with initial vehicle group sizes of 16, 20, or 28, there is an 80% likelihood of having 9, 11, or 16 infants at day 7 post-partum, respectively. A PPND study initiated with group size 20 could detect a threefold increase of test item-related pregnancy or infant loss. CONCLUSIONS: For designing and managing primate developmental toxicity studies, this type of analysis provides an objective tool to facilitate decisions either by supplementing groups with additional pregnant animals or stopping a group because an adverse effect on offspring survival has already been adequately revealed.


Asunto(s)
Pérdida del Embrión/inducido químicamente , Desarrollo Embrionario , Macaca fascicularis/embriología , Modelos Animales , Modelos Estadísticos , Pruebas de Toxicidad , Animales , Animales Recién Nacidos , Simulación por Computador , Femenino , Estimación de Kaplan-Meier , Método de Montecarlo , Periodo Posparto , Embarazo , Resultado del Embarazo , Modelos de Riesgos Proporcionales
12.
Int J Toxicol ; 29(6): 552-68, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926830

RESUMEN

Developmental toxicity studies for pharmaceutical safety testing are designed to evaluate potential adverse effects of drug treatment on pregnancy and on the developing embryo/fetus. Biopharmaceuticals present specific challenges for developmental toxicity testing because the pharmacology of these molecules, which are frequently human-specific proteins, is often restricted to humans and nonhuman primates (NHPs). For those species-restricted molecules, the only option for the evaluation of potential effects on development of the human biopharmaceutical is to use NHPs. This article reviews each of the stages of development in cynomolgus macaques (the most frequently used NHP) and the potential exposure of the embryo, fetus, and infant following administration of a biopharmaceutical during pregnancy and lactation. Because the purpose of the NHP developmental studies is to identify potential human risks, a comparison between macaque and human development and potential exposure has been made when possible. Understanding the potential exposure of the conceptus relative to critical periods in development is essential to designing a scientifically based study that adequately addresses human risks. Some options for NHP study designs, including the option of combining end points into a single study, and the pros and cons of each of the study options have been reviewed. Developmental studies for biopharmaceuticals in NHPs need to be optimally designed on a case-by-case basis taking into consideration the pharmacology of the molecule, the type of molecule (antibody or non-antibody), the potential exposure relative to the development of potential target organs, the clinical use, and the ethical considerations associated with the use of NHPs.


Asunto(s)
Evaluación Preclínica de Medicamentos/tendencias , Efectos Tardíos de la Exposición Prenatal , Proteínas Recombinantes/toxicidad , Teratógenos/toxicidad , Animales , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/prevención & control , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Lactancia , Macaca fascicularis , Masculino , Embarazo , Especificidad de la Especie , Pruebas de Toxicidad/tendencias
13.
Primate Biol ; 7(1): 1-4, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32232119

RESUMEN

Enhanced pre- and postnatal development (ePPND) studies have become the default developmental toxicity test for biopharmaceuticals if nonhuman primates represent the relevant species. Spontaneous pregnancy losses and infant deaths can be significant in macaques such as long-tailed macaques. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guideline S6(R1) states that pregnancy outcome can be judged also by the normogram-based variability of reference data according to a publication by Jarvis et al. (2010) defining a study as valid with six to eight live infants in the control group on postnatal day 7 (PND7). Since the release of ICH S6(R1) (2011), ePPND studies for biologics have replaced the former separate embryo-fetal and PPND study types. This work provides a retrospective analysis of pregnancy outcomes from 21 ePPND studies and group sizes of 14-24 animals per group. All studies reached the goal of at least six to eight infants on PND7, with overall losses ranging between 5 % and 45 %. Consistently, a group size of 14-24 maternal animals yielded more than six to eight infants on PND7. Therefore, it is suggested to reduce ePPND study group sizes from 20 to 14, yielding an animal number reduction of approx. 30 %.

14.
Reprod Toxicol ; 95: 29-36, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32413491

RESUMEN

The incidence of spontaneous pregnancy/infant losses is highly variable in long-tailed macaques (cynomolgus monkey), making it potentially difficult to ascertain test item-related effects in developmental toxicity studies. Therefore, pregnancy normograms had been developed by Jarvis et al. [1] to aid in the distinction of normal (e.g. test facility background) versus non-normal pregnancy outcomes. These normograms were mostly derived from embryo-fetal development studies and from PPND studies with a postnatal phase limited to seven days. However, the enhanced pre- and postnatal developmental (ePPND) study paradigm has essentially replaced these former study types. This work aims at providing enhanced normograms (e-normograms) in the context of regulatory ePPND studies. Survival functions for the prenatal phase (286 control pregnancies) and the postnatal phase (222 live infants) were estimated using the Kaplan-Meier estimator. Normograms were generated from survival curves and pseudo-study simulations. Data were available from two test facilities with comparable EU-compliant animal husbandry. Pregnancy duration/outcome as well as survival functions did not differ significantly between test facilities indicating that this husbandry system yields comparable developmental observations across different test facilities, at least in this NHP species. These novel e-normograms were developed for pregnant long-tailed macaques and provide an extended postnatal period up to three months, a new concept of separate normograms for the prenatal and the postnatal period, specific information on the perinatal phase events, a prediction of expected number of live infants for group size management, and the option to evaluate effects on pregnancy duration through distinction of live births and infant losses.


Asunto(s)
Nomogramas , Resultado del Embarazo , Pruebas de Toxicidad/estadística & datos numéricos , Animales , Desarrollo Embrionario , Femenino , Desarrollo Fetal , Estimación de Kaplan-Meier , Macaca fascicularis , Embarazo
15.
Birth Defects Res ; 112(1): 105-117, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746560

RESUMEN

Antibody-like biopharmaceuticals cross the placenta by utilizing existing transport pathways (e.g., FcRn receptor). There are limited data evaluating this transfer during organogenesis in any species. Understanding placental transfer of antibody-like biopharmaceuticals can help to predict risk of developmental toxicity across species, including humans. To complement previously published placental transfer data in the rat with humanized IgGΔ2 (hIgG2), the timing and magnitude of transfer in the cynomolgus monkey and embryo/fetal biodistribution of maternally administered 125 I-radiolabeled hIgG2 was quantified on gestation days (GD) 35, 40, 50, 70, and 140 using gamma counting and whole body autoradiography 24 hr following intravenous injection. Chorioallantoic placental tissues were collected at all time points for Western Blot analysis with anti-FcRn antibody. Maternally administered 125 I-hIgG2 was found in embryo/fetal tissues at all time points, including organogenesis. Embryo/fetal plasma 125 I-hIgG2 concentration increased during gestation, but only slightly up to GD 70 in embryo/fetal tissues, with hIgG2 tissue concentrations generally similar between GD70 and 140. The embryo/fetal:maternal 125 I-hIgG2 plasma concentration ratio was approximately 2.3 fold higher on GD 140, in comparison to ratios on GD 40. Importantly, placental FcRn protein expression was confirmed at all timepoints. These data demonstrate placental transfer of hIgG2 in a nonhuman primate model, and at levels comparable to the rat model, raising the potential for adverse developmental outcomes by direct antibody binding to biological targets.


Asunto(s)
Inmunoglobulina G/inmunología , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Feto/metabolismo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Radioisótopos de Yodo/metabolismo , Macaca fascicularis/inmunología , Macaca fascicularis/metabolismo , Intercambio Materno-Fetal/fisiología , Organogénesis/inmunología , Organogénesis/fisiología , Embarazo , Distribución Tisular/inmunología
16.
Birth Defects Res B Dev Reprod Toxicol ; 86(6): 446-62, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20025046

RESUMEN

Developmental and reproductive toxicology testing in nonhuman primates (NHPs) has become more common due to the increasing number of biopharmaceuticals in drug development, since NHPs are frequently the only species to express pharmacologic responses similar to humans. NHPs may also be used to help resolve issues associated with small-molecule reproductive toxicology in traditional species (rodents and rabbits). Adequate designs in NHP are presented for developmental toxicity (embryo-fetal development, pre-postnatal development, enhanced pre-postnatal development), reproductive toxicity (male and female), and juvenile toxicity studies. Optional parameters that may be included in these studies are discussed, as are new study designs that consolidate multiple aspects of the reproductive assessment and thereby conserve the limited supply of sexually mature NHPs available for testing. The details described will assist scientists in pharmaceutical, regulatory, and contract research organizations who are involved in conducting these unique studies to optimize their design based on case-by-case considerations.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Haplorrinos/fisiología , Reproducción/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Animales de Laboratorio , Callithrix/fisiología , Femenino , Macaca fascicularis/fisiología , Macaca mulatta/fisiología , Masculino , Preparaciones Farmacéuticas/clasificación , Conejos , Ratas , Reproducción/fisiología , Especificidad de la Especie
17.
Toxicol Pathol ; 36(7S): 7S-23S, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20852722

RESUMEN

Macaques provide excellent models for preclinical testing and safety assessment of female reproductive toxicants. Currently, cynomolgus monkeys are the predominant species for (reproductive) toxicity testing. Marmosets and rhesus monkeys are being used occasionally. The authors provide a brief review on physiology and endocrinology of the cynomolgus monkey ovarian cycle, practical guidance on assessment and monitoring of ovarian cyclicity, and new data on effects of social housing on ovarian cyclicity in toxicological studies. In macaques, cycle monitoring is achieved using daily vaginal smears for menstruation combined with cycle-timed frequent sampling for steroid and peptide hormone analysis. Owing to requirements of frequent and timed blood sampling, it is not recommended to incorporate these special evaluations into a general toxicity study design. Marmosets lack external signs of ovarian cyclicity, and cycle monitoring is done by regular determinations of progesterone. Cynomolgus and marmoset monkeys do not exhibit seasonal variations in ovarian activity, whereas such annual rhythm is pronounced in rhesus monkeys. Studies on pair- and group-housed cynomolgus monkeys revealed transient alterations in the duration and endocrinology of the ovarian cycle followed by return to normal cyclicity after approximately six months. This effect is avoided if the animals had contact with each other prior to mingling. These experiments also demonstrated that synchronization of ovarian cycles did not occur.

18.
Toxicol Pathol ; 36(7): 142s-163s, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21475639

RESUMEN

The authors describe a selection of normal findings and common naturally occurring lesions in the reproductive system of female macaques, including changes in the ovaries, uterus, cervix, vagina, and mammary glands. Normal features of immature ovaries, uteri, and mammary glands are described. Common non-neoplastic lesions in the ovaries include cortical mineralization, polyovular follicles, cysts, ovarian surface epithelial hyperplasia, and ectopic ovarian tissue. Ovarian neoplasms include granulosa cell tumors, teratomas, and ovarian surface epithelial tumors. Common non-neoplastic uterine findings include loss of features of normal cyclicity, abnormal bleeding, adenomyosis, endometriosis, epithelial plaques, and pregnancy-associated vascular remodeling. Hyperplastic and neoplastic lesions of the uterus include endometrial polyps, leiomyomas, and rarely endometrial hyperplasia and endometrial adenocarcinoma. Vaginitis is common. Cervical lesions include endocervical squamous metaplasia, polyps, and papillomavirus-associated lesions. Lesions in the mammary gland are most often proliferative and range from ductal hyperplasia to invasive carcinoma. Challenges to interpretation include the normal or pathologic absence of menstrual cyclicity and the potential misinterpretation of sporadic lesions, such as epithelial plaques or papillomavirus-associated lesions. Interpretation of normal and pathologic findings is best accomplished with knowledge of the life stage, reproductive history, and hormonal status of the animal.

19.
MAbs ; 10(1): 1-17, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28991509

RESUMEN

Monoclonal antibodies (mAbs) are improving the quality of life for patients suffering from serious diseases due to their high specificity for their target and low potential for off-target toxicity. The toxicity of mAbs is primarily driven by their pharmacological activity, and therefore safety testing of these drugs prior to clinical testing is performed in species in which the mAb binds and engages the target to a similar extent to that anticipated in humans. For highly human-specific mAbs, this testing often requires the use of non-human primates (NHPs) as relevant species. It has been argued that the value of these NHP studies is limited because most of the adverse events can be predicted from the knowledge of the target, data from transgenic rodents or target-deficient humans, and other sources. However, many of the mAbs currently in development target novel pathways and may comprise novel scaffolds with multi-functional domains; hence, the pharmacological effects and potential safety risks are less predictable. Here, we present a total of 18 case studies, including some of these novel mAbs, with the aim of interrogating the value of NHP safety studies in human risk assessment. These studies have identified mAb candidate molecules and pharmacological pathways with severe safety risks, leading to candidate or target program termination, as well as highlighting that some pathways with theoretical safety concerns are amenable to safe modulation by mAbs. NHP studies have also informed the rational design of safer drug candidates suitable for human testing and informed human clinical trial design (route, dose and regimen, patient inclusion and exclusion criteria and safety monitoring), further protecting the safety of clinical trial participants.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Primates , Animales , Anticuerpos Monoclonales/efectos adversos , Seguridad de Productos para el Consumidor , Humanos , Modelos Animales , Medición de Riesgo , Factores de Riesgo , Especificidad de la Especie
20.
Reprod Toxicol ; 63: 82-95, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27211603

RESUMEN

An enhanced embryo-fetal development study was conducted in cynomolgus monkeys using pateclizumab, a humanized IgG1 monoclonal antibody (mAb) targeting lymphotoxin-alpha. Pateclizumab administration between gestation days (GD) 20 and 132 did not induce maternal or developmental toxicities. The ratio of fetal-to-maternal serum concentration of pateclizumab was 0.73% on GD 50 and 61% by GD 139. Decreased fetal inguinal lymph node-to-body weight ratio was present in the high-dose group without microscopic abnormalities, a change attributable to inhibition of lymphocyte recruitment, which is a pharmacologic effect of pateclizumab during late lymph node development. The effect was observed in inguinal but not submandibular or mesenteric lymph nodes; this was attributed to differential susceptibility related to sequential lymph node development. Placental transfer of therapeutic IgG1 antibodies; thus, begins during the first trimester in non-human primates. Depending on the potency and dose levels administered, antibody levels in the fetus may be pharmacologically or toxicologically relevant.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/farmacocinética , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Linfotoxina-alfa/inmunología , Intercambio Materno-Fetal , Animales , Anticuerpos Monoclonales Humanizados/sangre , Células Dendríticas Foliculares/efectos de los fármacos , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/farmacología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/crecimiento & desarrollo , Ganglios Linfáticos/inmunología , Linfocitosis/inducido químicamente , Macaca fascicularis , Masculino , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA