Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 114: 104662, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32325112

RESUMEN

Nonclinical testing of human pharmaceuticals is conducted to assess the safety of compounds to be studied in human clinical trials and for marketing of new drugs. Although there is no exact number and type of nonclinical studies required for safety assessments, as there is inherent flexibility for each new compound, the traditional approach is outlined in various FDA and ICH guidance documents and involves a combination of in vitro assays and whole animal testing methods. Recent advances in science have led to the emergence of numerous new approach methodologies (NAMs) for nonclinical testing that are currently being used in various aspects of drug development. Traditional nonclinical testing methods can predict clinical outcomes, although improvements in these methods that can increase predictivity of clinical outcomes are encouraged and needed. This paper discusses FDA/CDER's view on the opportunities and challenges of using NAMs in drug development especially for regulatory purposes, and also includes examples where NAMs are currently being used in nonclinical safety assessments and where they may supplement and/or enhance current testing methods. FDA/CDER also encourages communication with stakeholders regarding NAMs and is committed to exploring the use of NAMs to improve regulatory efficiency and potentially expedite drug development.


Asunto(s)
Preparaciones Farmacéuticas/química , Animales , Desarrollo de Medicamentos , Humanos , Medición de Riesgo , Estados Unidos , United States Food and Drug Administration
3.
Reprod Toxicol ; 82: 111-123, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30316929

RESUMEN

2-hydroxy-4-methoxybenzophenone (HMB) is an ultraviolet light-absorbing compound that is used in sunscreens, cosmetics and plastics. HMB has been reported to have weak estrogenic activity by in vivo and in vitro studies, making it a chemical with potential reproductive concern. To explore if prenatal and lactational HMB exposure alters gene expression profiles of the developing reproductive organs, we performed microarray analysis using the prostate and testis of postnatal day (PND) 30 male Sprague-Dawley rats offspring exposed to 0, 3000, or 30,000 ppm of HMB from gestational day 6 through PND 21. Gene expression profiles of the prostate and testis were differentially affected by HMB dose with significant alterations observed at the 30,000 ppm HMB group. Tissue-specific gene expression was also identified. These genes, whose expression was altered by HMB exposure, may be considered as candidate biomarker(s) for testicular or prostatic toxicity; however, further studies are necessary to explore this potential.


Asunto(s)
Benzofenonas/toxicidad , Cosméticos/toxicidad , Próstata/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Lactancia , Masculino , Intercambio Materno-Fetal , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal , Próstata/metabolismo , Ratas Sprague-Dawley , Testículo/metabolismo
4.
Reprod Toxicol ; 69: 75-83, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28189605

RESUMEN

The mouse embryonic stem cell test (mEST) is a promising in vitro assay for predicting developmental toxicity. In the current study, early differentiation of D3 mouse embryonic stem cells (mESCs) under osteoblast culture conditions and embryotoxicity of cadmium sulfate were examined. D3 mESCs were exposed to cadmium sulfate for 24, 48 or 72h, and whole genome transcriptional profiles were determined. The results indicate a track of differentiation was identified as mESCs differentiate. Biological processes that were associated with differentiation related genes included embryonic development and, specifically, skeletal system development. Cadmium sulfate inhibited mESC differentiation at all three time points. Functional pathway analysis indicated biological pathways affected included those related to skeletal development, renal and reproductive function. In summary, our results suggest that transcriptional profiles are a sensitive indicator of early mESC differentiation. Transcriptomics may improve the predictivity of the mEST by suggesting possible modes of action for tested chemicals.


Asunto(s)
Compuestos de Cadmio/toxicidad , Diferenciación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias de Ratones/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Sulfatos/toxicidad , Animales , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Osteoblastos/citología
5.
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
6.
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
7.
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
8.
Birth Defects Res C Embryo Today ; 105(3): 190-200, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26404176

RESUMEN

Pregnancy is a hypercoagulable state which carries an excess risk of maternal venous thrombosis. Endothelial injury, alterations in blood flow and activation of the coagulation pathway are proposed to contribute to the hypercoagulability. The risk for thrombosis may be accentuated by certain drugs and device implants that directly or indirectly affect the coagulation pathway. To help ensure that these interventions do not result in adverse maternal or fetal outcomes during pregnancy, gravid experimental animals can be exposed to such treatments at various stages of gestation and over a dosage range that would identify hazards and inform risk assessment. Circulating soluble biomarkers can also be evaluated for enhancing the assessment of any increased risk of venous thrombosis during pregnancy. In addition to traditional in vivo animal testing, efforts are under way to incorporate reliable non-animal methods in the assessment of embryofetal toxicity and thrombogenic effects. This review summarizes hemostatic balance during pregnancy in animal species, embryofetal development, biomarkers of venous thrombosis, and alterations caused by drug-induced venous thrombosis.


Asunto(s)
Complicaciones Hematológicas del Embarazo/fisiopatología , Trombosis/fisiopatología , Animales , Biomarcadores/sangre , Coagulación Sanguínea , Desarrollo Embrionario , Femenino , Desarrollo Fetal , Hemostasis , Humanos , Embarazo , Complicaciones Hematológicas del Embarazo/sangre , Trombosis/sangre , Trombosis de la Vena/sangre , Trombosis de la Vena/fisiopatología
9.
Reprod Toxicol ; 53: 131-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25929818

RESUMEN

The mouse Embryonic Stem cell Test (EST) using cardiomyocyte differentiation is a promising in vitro assay for detecting potential embryotoxicity; however, the addition of another differentiation endpoint, such as osteoblasts, may improve the predictive value of the test. A number of variables such as culture conditions and starting cell number were investigated. A 14 day direct plating method of D3 mouse embryonic stem cells (mESCs) was used to test the predictivity of osteoblast differentiation as an endpoint in the EST. Twelve compounds were tested using the prediction model developed in the ECVAM validation study. Eight of the compounds selected from the EST validation study served as model compounds; four additional compounds known to produce skeletal defects were also tested. Our results indicate comparable chemical classification between the validated cardiomyocyte endpoint and the osteoblast endpoint. These results suggest that differentiation to osteoblasts may provide confirmatory information in predicting embryotoxicity.


Asunto(s)
Osteoblastos/efectos de los fármacos , Teratógenos/toxicidad , Fosfatasa Alcalina/metabolismo , Animales , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Hep G2 , Humanos , Ratones , Células Madre Embrionarias de Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Reproducibilidad de los Resultados
10.
Birth Defects Res B Dev Reprod Toxicol ; 104(1): 35-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25707689

RESUMEN

BACKGROUND: 2-Hydroxy-4-methoxybenzophenone (HMB) is an ultraviolet (UV) absorbing compound used in many cosmetic products as a UV-protecting agent and in plastics for preventing UV-induced photodecomposition. HMB has been detected in over 95% of randomly collected human urine samples from adults and from premature infants, and it may have estrogenic potential. METHODS: To determine the effects of maternal and lactational exposure to HMB on development and reproductive organs of offspring, time-mated female Harlan Sprague-Dawley rats were dosed with 0, 1000, 3000, 10,000, 25,000, or 50,000 ppm HMB (seven to eight per group) added to chow from gestation day 6 until weaning on postnatal day (PND) 23. RESULTS AND CONCLUSION: Exposure to HMB was associated with reduced body and organ weights in female and male offspring. No significant differences were observed in the number of implantation sites/litter, mean resorptions/litter, % litters with resorptions, number and weights of live fetuses, or sex ratios between the control and HMB dose groups. Normalized anogenital distance in male pups at PND 23 was decreased in the highest dose group. Spermatocyte development was impaired in testes of male offspring in the highest dose group. In females, follicular development was delayed in the highest dose group. However, by evaluating levels of the compound in rat serum, the doses at which adverse events occurred are much higher than usual human exposure levels. Thus, exposure to less than 10,000 ppm HMB does not appear to be associated with adverse effects on the reproductive system in rats.


Asunto(s)
Benzofenonas/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Lactancia/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/patología , Reproducción/efectos de los fármacos , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Recuento de Células , Femenino , Masculino , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Ratas Sprague-Dawley , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/patología , Espermatocitos/efectos de los fármacos , Espermatocitos/patología , Testosterona/sangre
11.
Toxicol Sci ; 135(2): 277-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897986

RESUMEN

Endocrine-active chemicals can potentially have adverse effects on both humans and wildlife. They can interfere with the body's endocrine system through direct or indirect interactions with many protein targets. Estrogen receptors (ERs) are one of the major targets, and many endocrine disruptors are estrogenic and affect the normal estrogen signaling pathways. However, ERs can also serve as therapeutic targets for various medical conditions, such as menopausal symptoms, osteoporosis, and ER-positive breast cancer. Because of the decades-long interest in the safety and therapeutic utility of estrogenic chemicals, a large number of chemicals have been assayed for estrogenic activity, but these data exist in various sources and different formats that restrict the ability of regulatory and industry scientists to utilize them fully for assessing risk-benefit. To address this issue, we have developed an Estrogenic Activity Database (EADB; http://www.fda.gov/ScienceResearch/BioinformaticsTools/EstrogenicActivityDatabaseEADB/default.htm) and made it freely available to the public. EADB contains 18,114 estrogenic activity data points collected for 8212 chemicals tested in 1284 binding, reporter gene, cell proliferation, and in vivo assays in 11 different species. The chemicals cover a broad chemical structure space and the data span a wide range of activities. A set of tools allow users to access EADB and evaluate potential endocrine activity of chemicals. As a case study, a classification model was developed using EADB for predicting ER binding of chemicals.


Asunto(s)
Bases de Datos de Compuestos Químicos , Disruptores Endocrinos/toxicidad , Glándulas Endocrinas/efectos de los fármacos , Estrógenos/farmacología , Animales , Humanos
12.
Int J Toxicol ; 31(6): 507-28, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23134714

RESUMEN

Over the past 3 decades minipigs have moved from being an obscure alternative to dogs and nonhuman primates to being a standard animal model in regulatory toxicity studies. This article covers the use of minipigs as a model in the context of nonclinical drug safety and provides an overview of the minipig's developmental history and relates minipigs to other animal species commonly used in toxicology; and the minipig's translational power is supported by 43 case studies of marketed drug products covered. Special focus is given to criteria for selecting minipigs in nonclinical programs supporting the development of new medicines; the use of swine in the assessment of food additives, agrochemicals, and pesticides; as well as a regulatory perspective on the use of minipigs in Food and Drug Administration (FDA)-regulated products. This article presents the main points conveyed at a symposium held at the 2010 American College of Toxicology meeting in Baltimore, Maryland.


Asunto(s)
Animales de Laboratorio/fisiología , Evaluación Preclínica de Medicamentos , Porcinos Enanos/fisiología , Pruebas de Toxicidad/métodos , Xenobióticos/toxicidad , Animales , Perros , Aprobación de Drogas/legislación & jurisprudencia , Inocuidad de los Alimentos , Regulación Gubernamental , Maryland , Modelos Animales , Sociedades Científicas , Porcinos , Pruebas de Toxicidad/tendencias , Estados Unidos , United States Food and Drug Administration
13.
Birth Defects Res B Dev Reprod Toxicol ; 92(5): 404-12, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22006510

RESUMEN

In April 2009, the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute's (HESI) Developmental and Reproductive Toxicology Technical Committee held a two-day workshop entitled "Developmental Toxicology-New Directions." The third session of the workshop focused on ways to refine animal studies to improve relevance and predictivity for human risk. The session included five presentations on: (1) considerations for refining developmental toxicology testing and data interpretation; (2) comparative embryology and considerations in study design and interpretation; (3) pharmacokinetic considerations in study design; (4) utility of genetically modified models for understanding mode-of-action; and (5) special considerations in reproductive testing for biologics. The presentations were followed by discussion by the presenters and attendees. Much of the discussion focused on aspects of refining current animal testing strategies, including use of toxicokinetic data, dose selection, tiered/triggered testing strategies, species selection, and use of alternative animal models. Another major area of discussion was use of non-animal-based testing paradigms, including how to define a "signal" or adverse effect, translating in vitro exposures to whole animal and human exposures, validation strategies, the need to bridge the existing gap between classical toxicology testing and risk assessment, and development of new technologies. Although there was general agreement among participants that the current testing strategy is effective, there was also consensus that traditional methods are resource-intensive and improved effectiveness of developmental toxicity testing to assess risks to human health is possible. This article provides a summary of the session's presentations and discussion and describes some key areas that warrant further consideration.


Asunto(s)
Alternativas a las Pruebas en Animales , Modelos Animales , Proyectos de Investigación , Pruebas de Toxicidad/métodos , Animales , Animales Modificados Genéticamente , Desarrollo Fetal/efectos de los fármacos , Humanos , Pruebas de Mutagenicidad/métodos , Riesgo , Medición de Riesgo , Seguridad , Toxicología
14.
Birth Defects Res B Dev Reprod Toxicol ; 92(5): 384-94, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21922641

RESUMEN

A review is presented of the use of developmental toxicity testing in the United States and international regulatory assessment of human health risks associated with exposures to pharmaceuticals (human and veterinary), chemicals (agricultural, industrial, and environmental), food additives, cosmetics, and consumer products. Developmental toxicology data are used for prioritization and screening of pharmaceuticals and chemicals, for evaluating and labeling of pharmaceuticals, and for characterizing hazards and risk of exposures to industrial and environmental chemicals. The in vivo study designs utilized in hazard characterization and dose-response assessment for developmental outcomes have not changed substantially over the past 30 years and have served the process well. Now there are opportunities to incorporate new technologies and approaches to testing into the existing assessment paradigm, or to apply innovative approaches to various aspects of risk assessment. Developmental toxicology testing can be enhanced by the refinement or replacement of traditional in vivo protocols, including through the use of in vitro assays, studies conducted in alternative nonmammalian species, the application of new technologies, and the use of in silico models. Potential benefits to the current regulatory process include the ability to screen large numbers of chemicals quickly, with the commitment of fewer resources than traditional toxicology studies, and to refine the risk assessment process through an enhanced understanding of the mechanisms of developmental toxicity and their relevance to potential human risk. As the testing paradigm evolves, the ability to use developmental toxicology data to meet diverse critical regulatory needs must be retained.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Pruebas de Toxicidad , Toxicología , Animales , Cosméticos/efectos adversos , Salud Ambiental , Contaminantes Ambientales/toxicidad , Aditivos Alimentarios/toxicidad , Ensayos Analíticos de Alto Rendimiento , Humanos , Medición de Riesgo , Seguridad , Pruebas de Toxicidad/métodos , Toxicología/legislación & jurisprudencia , Toxicología/métodos , Toxicología/normas
15.
Crit Rev Toxicol ; 41(6): 507-44, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21591905

RESUMEN

Quantitative methods for estimation of cancer risk have been developed for daily, lifetime human exposures. There are a variety of studies or methodologies available to address less-than-lifetime exposures. However, a common framework for evaluating risk from less-than-lifetime exposures (including short-term and/or intermittent exposures) does not exist, which could result in inconsistencies in risk assessment practice. To address this risk assessment need, a committee of the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute conducted a multisector workshop in late 2009 to discuss available literature, different methodologies, and a proposed framework. The proposed framework provides a decision tree and guidance for cancer risk assessments for less-than-lifetime exposures based on current knowledge of mode of action and dose-response. Available data from rodent studies and epidemiological studies involving less-than-lifetime exposures are considered, in addition to statistical approaches described in the literature for evaluating the impact of changing the dose rate and exposure duration for exposure to carcinogens. The decision tree also provides for scenarios in which an assumption of potential carcinogenicity is appropriate (e.g., based on structural alerts or genotoxicity data), but bioassay or other data are lacking from which a chemical-specific cancer potency can be determined. This paper presents an overview of the rationale for the workshop, reviews historical background, describes the proposed framework for assessing less-than-lifetime exposures to potential human carcinogens, and suggests next steps.


Asunto(s)
Carcinógenos/toxicidad , Exposición a Riesgos Ambientales/normas , Mutágenos/toxicidad , Bioensayo/métodos , Carcinógenos/administración & dosificación , Bases de Datos Factuales , Árboles de Decisión , Relación Dosis-Respuesta a Droga , Determinación de Punto Final , Contaminación de Alimentos/análisis , Guías como Asunto , Productos Domésticos/efectos adversos , Humanos , Mutágenos/administración & dosificación , National Institute of Environmental Health Sciences (U.S.) , Neoplasias/inducido químicamente , Plaguicidas/efectos adversos , Medición de Riesgo , Factores de Tiempo , Estados Unidos , United States Environmental Protection Agency , United States Food and Drug Administration
16.
Artículo en Inglés | MEDLINE | ID: mdl-21312321

RESUMEN

Workshops on maternal toxicity were held at the annual Society of Toxicology, Teratology Society, and European Teratology Society meetings in 2009. Speakers presented background information prior to a general discussion on this topic. The following recommendations/options are based on the outcome of the discussions at the workshops: 1. A comprehensive evaluation of all available data from general toxicity studies, range-finding Developmental and Reproductive Toxicology (DART) studies, class effects, structure-activity relationships, exposure studies, etc. is essential for appropriate dose selection for definitive DART studies. The intent is to avoid marked maternal toxicity leading to mortality or decreased body weight gains of greater than 20% for prolonged periods. (a) Evaluate alternative endpoints for dose selection and data interpretation (e.g., target tissue effects and pharmacology) for biotherapeutics. (B) Evaluate additional maternal parameters based on effects and/or target organs observed in short-term (e.g., 2- or 4-week) general toxicity studies. 2. Evaluate all available data to determine a cause-effect relationship for developmental toxicity. (a) Conduct a pair-feeding/pair-watering study as a follow-up. (b) Evaluate individual data demonstrating maternal toxicity in the mother with adverse embryo-fetal outcomes in the litter associated with the affected mother. (c) Conduct single-dose studies at increasing doses as a complement to conventional embryo-fetal toxicity studies for certain classes of compounds that affect the hERG channel. 3. Support statements that embryo-fetal effects are caused by maternal toxicity and/or exaggerated pharmacology, especially for malformations. (a) Provide mechanistic or other supporting data. (b) Establish the relevance of the DART findings in animals for human exposures. Birth Defects Res (Part B) 92:36-51, 2010. © 2011 Wiley-Liss, Inc.


Asunto(s)
Exposición Materna , Proyectos de Investigación , Estadística como Asunto , Pruebas de Toxicidad/métodos , Animales , Peso Corporal , Desarrollo Embrionario , Femenino , Feto/patología , Humanos , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Conejos , Ratas
17.
Artículo en Inglés | MEDLINE | ID: mdl-20859822

RESUMEN

Triclosan has broad-spectrum anti-microbial activity against most gram-negative and gram-positive bacteria. It is widely used in personal care products, household items, medical devices, and clinical settings. Due to its extensive use, there is potential for humans in all age groups to receive life-time exposures to triclosan, and, indeed, triclosan has been detected in human tissues and the environment. Data gaps exist regarding the chronic dermal toxicity and carcinogenicity of triclosan, which is needed for the risk assessment of triclosan. The US Food and Drug Administration (FDA) nominated triclosan to the National Toxicology Program (NTP) for toxicological evaluations. Currently, the NTP is conducting several dermal toxicological studies to determine the carcinogenic potential of triclosan, evaluate its endocrine and developmental-reproductive effects, and investigate the potential UV-induced dermal formation of chlorinated phenols and dioxins of triclosan. This paper reviews data on the human exposure, environmental fate, efficacy of anti-microbial activity, absorption, distribution, metabolism and elimination, endocrine disrupting effects, and toxicity of triclosan.


Asunto(s)
Antiinfecciosos , Triclosán , Antiinfecciosos/metabolismo , Antiinfecciosos/toxicidad , Humanos , Triclosán/metabolismo , Triclosán/toxicidad
18.
Pharmacogenomics ; 10(12): 1979-86, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19958096

RESUMEN

Pharmacogenomic biomarkers are becoming increasingly common in medicine and drug development. However, there is a genuine concern that the healthcare workforce will be ill-equipped to translate this information to clinical practice. As a result, a major effort is underway to educate future healthcare professionals on pharmacogenomics. This paper describes the development of a year-long course that aims to instill the fundamental concepts of this rapidly growing field into the minds of undergraduate students. This course offers the advantage of exposing students to the concepts of pharmacogenomics prior to their enrollment in PhD, PharmD or MD/DO graduate programs.


Asunto(s)
Educación de Pregrado en Medicina/métodos , Educación en Farmacia/métodos , Farmacogenética/educación , Enseñanza/métodos , Curriculum/tendencias
19.
Environ Health Perspect ; 114(4): 573-8, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16581548

RESUMEN

Over the past few years, both the U.S. Food and Drug Administration (FDA) and the pharmaceutical industry have recognized the potential importance of pharmacogenomics and toxicogenomics to drug development. To resolve the uncertainties surrounding the use of microarray technology and the presentation of genomics data for regulatory purposes, several pharmaceutical companies and genomics technology providers have provided the FDA with reports of genomics studies that included supporting toxicology data (e.g., serum chemistry, histopathology). These studies were not associated with any active drug application and were exploratory or hypothesis generating in nature. For training purposes, these reports were reviewed by the Nonclinical Pharmacogenomics Subcommittee consisting of the Center for Drug Evaluation and Research pharmacology and toxicology researchers and reviewers. In this article, we describe some of these submissions and report on our assessment of data content, format, and quality control metrics that were useful for evaluating these nonclinical genomics submissions, specifically in relation to the proposed MIAME/MINTox (minimum information about a microarray experiment/minimum information needed for a toxicology experiment) recommendations. These genomics submissions allowed both researchers and regulators to gain experience in the process of reviewing and analyzing toxicogenomics data. The experience will allow development of recommendations for the submission and review of these data as the state of the science evolves.


Asunto(s)
Genómica , Farmacogenética , Toxicología , Animales , Sistemas de Administración de Bases de Datos , Educación , Análisis de Secuencia por Matrices de Oligonucleótidos
20.
Birth Defects Res B Dev Reprod Toxicol ; 74(3): 268-76, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15954087

RESUMEN

BACKGROUND: The individual effects of boric acid (BA) and hyperthermia on the development of the axial skeleton have been reported previously. Both cause an increased incidence of axial skeletal defects including a decrease in the total number of ribs and vertebrae. Because of the similarity in the effects of the two agents, we examined their interaction when given in combination to pregnant rats on gestational day (GD) 10. METHODS: Dams were treated on GD 10 with BA (0, 250, or 500 mg/kg) and hyperthermia (37, 41, or 42 degrees C) and allowed to deliver their pups. Doses of BA were based on results from a dose-finding study. Litters were evaluated on postnatal days (PND) 1 and 3 for number, gender, and weight of pups. On PND3, pups were examined externally and viscerally, and double-stained for skeletal evaluation. RESULTS: A dose-dependent, statistically significant increase in fetal skeletal defects was seen on PND 3 with BA or hyperthermia alone with even greater effects when given in combination. Defects included rib and vertebral fusions, split vertebral centra in the thoracic and lumbar areas, and a decrease in the total number of ribs and vertebrae. CONCLUSIONS: The increased incidence of skeletal defects resulting from combined exposure to hyperthermia and BA was additive for segmentation defects and synergistic for the reduction in numbers of vertebrae.


Asunto(s)
Anomalías Inducidas por Medicamentos , Ácidos Bóricos/toxicidad , Embrión de Mamíferos/efectos de los fármacos , Calor , Insecticidas/toxicidad , Columna Vertebral/anomalías , Animales , Huesos/anomalías , Huesos/embriología , Femenino , Feto/anomalías , Masculino , Ratas , Ratas Sprague-Dawley , Columna Vertebral/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...