Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Reprod Toxicol ; 56: 105-8, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25937597

RESUMEN

Concern has been raised about the number of juvenile animal studies (JAS) being conducted in the European Union (EU) and the number of species required. The question of whether there has been an increase in the number of JAS requested by European Medicines Agency (EMA) and the question of single or multiple species studies are the focus of this manuscript. All source data on Pediatric Investigation Plans (PIP) were obtained from EMA pediatrics website and evaluated by year as obtained and adjusted to the date of the original PIP agreement rather than the newly assigned date of the PIP modification. The analysis of the data originally suggested an increase in the number of juvenile animal studies per year post 2010. However, a thorough examination of the adjusted PIP data from the EMA from December 2007 through 2013 support that the number of JAS being included in PIPs per year is not increasing but is in fact fairly stable at 15-30% per year. The data also, as expected, support that the JAS are mostly proposed with a single species and that species is predominately the rat and that multi-species JAS for an agreed PIP are infrequent.


Asunto(s)
Pruebas de Toxicidad/métodos , Pruebas de Toxicidad/tendencias , Factores de Edad , Animales , Unión Europea , Humanos , Modelos Animales , Evaluación de Programas y Proyectos de Salud , Ratas , Medición de Riesgo , Especificidad de la Especie , Factores de Tiempo
2.
Reprod Toxicol ; 41: 67-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23792114

RESUMEN

To determine the sensitivity of male reproductive toxicity endpoints in NHPs we performed a power analysis of routine and triggered endpoints using control data from sexually mature Asian and Mauritian NHPs. The power to detect a 50% change from control was 13-30% for male reproductive organ weights, ∼30% for testicular volume, 6-66% for seminal analyses and 10-78% for male hormones. Overall, male reproductive endpoints have poor power (less than 80%) to detect a 50% change from control with a group size of 3 monkeys. Confidently identifying adverse male reproductive effects with these endpoints would likely require specialized study designs with larger group sizes. Triggering of non-routine endpoints in cases where there is special concern for male reproductive toxicity is unlikely to increase sensitivity to detect adverse effects.


Asunto(s)
Pruebas de Toxicidad/estadística & datos numéricos , Animales , Interpretación Estadística de Datos , Hormona Folículo Estimulante , Genitales Masculinos , Hormona Luteinizante , Macaca fascicularis , Masculino , Tamaño de los Órganos , Reproducción , Recuento de Espermatozoides , Motilidad Espermática , Testosterona
3.
Drug Chem Toxicol ; 28(4): 409-21, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16298872

RESUMEN

Aspirin (ASA), an irreversible cyclooxygenase (COX) inhibitor, induces ventricular septal defect (VSD) and diaphragmatic hernia (DH) in rat fetuses when administered on gestation days (GDs) 9-10, a critical period for cardiovascular (CV) and midline development. Evaluation of a spectrum of nonsteroidal antiinflammatory drugs (NSAIDs; reversible COX inhibitors) showed that while some NSAIDs induced VSD in rats, none of the NSAIDs evaluated produced DH. In addition to inhibiting COX, ASA also inhibits carbonic anhydrase. The purpose of this study was to determine whether concurrent inhibition of COX and carbonic anhydrase would produce a teratogenic profile that includes both VSD and DH. To inhibit both COX and carbonic anhydrase, ibuprofen (COX inhibitor) and acetazolamide (carbonic anhydrase inhibitor) were coadministered on GDs 9-10. Groups of 20 female Crl:CD(SD)IGS BR rats were given either 300 mg kg(-1) day(-1) ibuprofen, 1000 mg kg(-1) day(-1) acetazolamide, or both (combination of ibuprofen and acetazolamide). Fetuses were evaluated on GD 21 for external and visceral development. Ibuprofen induced VSD in 3.7% of fetuses per litter; no defects in appendicular skeletal development were noted. Acetazolamide induced VSD in 5.9% of the fetuses per litter and appendicular defects in 41% of the fetuses per litter. Coadministration of ibuprofen and acetazolamide produced VSD in 18.7% of the fetuses per litter and appendicular defects in 77% of the fetuses per litter; however, there were no DH. Therefore, while concurrent inhibition of COX and carbonic anhydrase did not produce DH, potentiation was noted for the induction of VSD and appendicular anomalies.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Acetazolamida/toxicidad , Inhibidores de Anhidrasa Carbónica/toxicidad , Inhibidores de la Ciclooxigenasa/toxicidad , Ibuprofeno/metabolismo , Animales , Animales Recién Nacidos , Peso al Nacer/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Sinergismo Farmacológico , Ingestión de Alimentos/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Defectos del Tabique Interventricular/inducido químicamente , Hernia Diafragmática/inducido químicamente , Ibuprofeno/toxicidad , Masculino , Exposición Materna , Embarazo , Ratas , Ratas Sprague-Dawley
4.
Birth Defects Res B Dev Reprod Toxicol ; 74(5): 431-41, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16249996

RESUMEN

BACKGROUND: Feed restriction with its resultant body weight loss impacts the rodent estrous cycle; however, the manifestation of these changes in a regulatory study design has not been documented. This study reports the effects of feed restriction in the context of an FDA regulatory submission. METHODS: Adult female rats (n = 20/group; weighing approximately 200 g each) were provided rodent chow ad lib (control) or at 20, 15, 10, or 7.5 g/rat/day (g/day) during a 2-week pre-mating phase, throughout the mating phase, and up to gestation day (GD) 7. On GD 8, all animals were provided ad lib feed until necropsy on GD 14. Estrous cyclicity, mating, and fertility parameters were evaluated. RESULTS: Ad lib rats consumed approximately 20 and 28 g/day during the pre-mating and gestation phases, respectively. All measured fertility parameters in the 20 g/day group were similar to control values. In the 15 g/day group, body weight was reduced by 16% at 2 weeks, prolonged diestrus occurred, and fertility was compromised due to reductions in corpora lutea. Within 2 weeks, mean body weight in groups receiving < or = 10 g/day was reduced by > or = 29% compared to ad lib values, and overt changes in estrous cyclicity, mating, and fertility occurred. The 7.5 g/day group was not sustainable beyond the pre-mating phase. CONCLUSIONS: For this study type, feed intake at < or = 50% ad lib values (< or = 10 g/day) was inadequate due to the magnitude and rapidity of body weight effects. Estrous parameters appeared slightly more sensitive than functional measures, as body weight changes of approximately 16% appeared near the threshold of changing routinely calculated estrous cycle parameters and were later associated with reduced fertility. In general, body weight differences of 10-15% by themselves were not adverse to normal reproduction (20 g/day).


Asunto(s)
Ciclo Estral/fisiología , Fertilidad/fisiología , Privación de Alimentos/fisiología , Conducta Sexual Animal , Animales , Peso Corporal , Femenino , Masculino , Tamaño de los Órganos , Embarazo , Ratas
5.
Birth Defects Res B Dev Reprod Toxicol ; 74(5): 424-30, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16249998

RESUMEN

BACKGROUND: Appropriate maternal nutrition and body weight gain during pregnancy is well established as a major factor in healthy prenatal development in humans. Given the role of nutrition and body weight gain in normal development, pharmaceuticals intended to reduce appetite and promote weight loss will generate developmental toxicity data that may be challenging to interpret. To aid with this, the effects of feed restriction, and subsequent reduction in maternal body weight gain, on embryo-fetal development was investigated in the rabbit. METHODS: Groups of 15 pregnant New Zealand White rabbits were offered 150 (control), 110, 75, 55, 35, and 15 g feed/day from gestation day (GD) 7-19. Cesarean sections were carried out on GD 29 and fetuses were examined for external, visceral, and skeletal development. RESULTS: Maternal body weights at the end of the feed restriction period (GD 20) were 0.97, 0.98, 0.93, 0.94, and 0.86 x control for the 110, 75, 55, 35, and 15 g feed/day groups, respectively. Only at 15 g feed/day was there a net maternal body weight loss (the GD 20 body weight was 0.93 x the GD 6 body weight) at the end of the feed restriction period. Six does aborted in the 15 g feed/day group; there were no other abortions associated with feed restriction. Fetal body weight was significantly reduced at 75, 55, 35, and 15 g feed/day (0.95, 0.90, 0.86, and 0.84 x control, respectively). There were no external or visceral malformations or variations, and no skeletal malformations associated with feed restriction. The incidence of fetuses with sternebrae 5 or 6 unossified was increased at feed levels < or = 75 g/day. At a feed level of 35 g/day there was an increase in unossified metatarsals and metacarpals, and an increase in the number of fetuses with a reduced number of caudal vertebrae ossified. Although these findings were not increased at a feed level of 15 g/day, the lack of dose response was likely due to increased abortion and subsequent decrease in fetuses available for evaluation at 15 g feed/day. CONCLUSION: These data demonstrate that feed restriction to feed levels that produce substantial reductions in maternal body weight gain can result in developmental toxicity expressed by abortion, reduced fetal weight, and alterations in ossification. Abortion only occurred when feed was restricted to an amount that produced maternal body weight loss (15 g feed/day) whereas reduced fetal weight and increased incidence of fetuses with unossified sternebrae, metatarsals, metacarpals, or caudal vertebrae were noted at feed levels of < or = 75 g/day. There were no fetal malformations associated with feed restriction.


Asunto(s)
Huesos/embriología , Desarrollo Embrionario/fisiología , Desarrollo Fetal/fisiología , Privación de Alimentos , Organogénesis , Alimentación Animal , Animales , Peso Corporal , Embrión de Mamíferos , Femenino , Peso Fetal , Tamaño de los Órganos , Osteogénesis , Embarazo , Conejos
6.
Artículo en Inglés | MEDLINE | ID: mdl-16193501

RESUMEN

BACKGROUND: Given the role of nutrition and body weight gain in normal development, pharmaceuticals intended to reduce appetite and promote weight loss will generate safety data that may be challenging to interpret. To aid with this, the effects of feed restriction and subsequent body weight reductions on embryo-fetal development were investigated in the rat. METHODS: Groups of 20 timed pregnant female Sprague-Dawley rats were offered Certified Rodent Diet 5002 either ad libitum or in restricted amounts of 20, 15, 10, and 7.5 g/day from Gestation Day (GD) 6-17. Clinical signs, body weights, and food consumption were recorded. Cesarean sections were performed on GD 21 and fetuses were sexed, weighed, and examined for external, visceral, and skeletal development. RESULTS: Mean maternal body weights at the end of the feed restriction period, GD 18, were reduced 0.87 x, 0.80 x, 0.69 x, and 0.63 x control mean in the 20, 15, 10, and 7.5 g/day groups, respectively. Mean body weight gains for the restriction period inclusive, GD 6-18, were 0.49 x and 0.24 x control at 10 and 7.5 g/day, respectively, and a mean body weight loss occurred at 10 and 7.5 g/day (0.95 x and 0.85 x mean GD 6 body weight, respectively). Fetal body weights were reduced 0.95 x, 0.93 x, 0.90 x, and 0.76 x control at 20, 15, 10, and 7.5 g/day, respectively. This resulted in a reduction in gravid uterine weight at 10 and 7.5 g/day. There were no external, visceral, or skeletal malformations attributed to feed restriction. There was an increase in the skeletal variation of wavy ribs and a decrease in ossification at 7.5 g/day. CONCLUSIONS: These data demonstrate that feed restriction-induced reductions in maternal gestational body weight gain of approximately 50% compared to ab lib fed rats only caused a reduction in fetal body weight. Even up to a 15% maternal gestational body weight loss had no effect on embryo viability in rats, but retarded fetal growth significantly enough to induce minor changes in skeletal development. There were no external, visceral, or skeletal malformations associated with any of the levels of maternal body weight reduction or loss.


Asunto(s)
Huesos/embriología , Desarrollo Embrionario , Desarrollo Fetal , Privación de Alimentos , Organogénesis , Alimentación Animal , Animales , Peso Corporal , Huesos/fisiología , Embrión de Mamíferos , Femenino , Peso Fetal , Tamaño de los Órganos , Embarazo , Ratas , Ratas Sprague-Dawley
8.
Birth Defects Res B Dev Reprod Toxicol ; 71(3): 150-60, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15282736

RESUMEN

BACKGROUND: Lasofoxifene is a nonsteroidal selective estrogen receptor modulator (SERM). With high affinity to the alpha and beta human estrogen receptors and greater potency than other SERMs, lasofoxifene is potentially a superior treatment for postmenopausal osteoporosis. In light of the known effects of estrogen-modulating compounds on female reproductive indices, two studies were conducted to evaluate the effects of lasofoxifene on female rat cyclicity, reproduction, and parturition. METHODS: One study evaluated effects of lasofoxifene on estrous cyclicity, and the second study assessed effects on implantation and parturition. In the cyclicity study, lasofoxifene was administered to female rats at doses of 0.1, 0.3, and 1.0 mg/kg/day for 14 consecutive days. After treatment, there was a 3-week reversibility phase followed by a mating phase. In the implantation study, lasofoxifene was administered to pregnant female rats at doses of 0.01, 0.03, and 0.1 mg/kg/day for 7 consecutive days (gestation day [GD] 0-6). Some animals were euthanized on GD 21, and the remainder of the group was allowed to deliver the F1 generation. Several developmental indices were evaluated in the F1 pups through post-natal day (PND) 21. RESULTS: In the cyclicity study, all lasofoxifene-treated females were anestrous by Study Day 7 (1.0 mg/kg) or 9 (0.3 and 0.1 mg/kg). The reversibility phase resulted in restoration of normal estrous cycles by the end of 1 (0.1 mg/kg) or 2 weeks (0.3 and 1.0 mg/kg). During the mating phase, no adverse effects occurred in pregnancy success or reproductive parameters. In the implantation study, all doses of lasofoxifene increased pre- and post-implantation losses, increased gestation length, and reduced litter size. None of the developmental parameters measured on the F1 generation was adversely affected. CONCLUSION: Lasofoxifene reversibly altered the estrous cycle and inhibited implantation, consistent with what would be expected from a member of the SERM class.


Asunto(s)
Pirrolidinas/toxicidad , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Tetrahidronaftalenos/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Implantación del Embrión/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Estrógenos/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Fertilidad/efectos de los fármacos , Masculino , Exposición Materna , Osteoporosis/tratamiento farmacológico , Parto/efectos de los fármacos , Embarazo/efectos de los fármacos , Preñez , Pirrolidinas/efectos adversos , Pirrolidinas/sangre , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Tetrahidronaftalenos/efectos adversos , Tetrahidronaftalenos/sangre , Factores de Tiempo
9.
Toxicol Sci ; 75(2): 448-57, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12883086

RESUMEN

The potential developmental toxicity and the in vitro and in vivo genotoxicity of HCC-230fa were assessed. In the developmental toxicity study, groups of 25 mated Crl:CD(R)(SD)BR rats were exposed (whole body) by inhalation to HCC-230fa over days 7-21 of gestation; the day of confirmed mating was designated as gestation day 1 (GD1). Exposures were 6 h per day at concentrations of 0, 0.5, 2.5, or 25 ppm. Body weight, food consumption, and clinical observation data were collected during the study. On day 22 of gestation, the dams were euthanized and examined grossly. The fetuses were removed and subsequently weighed, sexed, and examined for external, visceral, head, and skeletal alterations. Evidence of maternal and developmental toxicity was observed at 25 ppm and was noted as significant, compound-related reductions in mean maternal body weight, weight change, and food consumption. Significant fetal effects also were observed at 25 ppm as compound-related reductions in mean fetal weight and increased fetal malformations (filamentous tail, situs inversus, absent vertebrae) and variations (rudimentary cervical ribs, delayed sternebral ossification). There was no evidence of either maternal or developmental toxicity at 0.5 or 2.5 ppm. The genotoxicity of HCC-230fa was examined in a bacterial reversion assay and in erythrocyte micronucleus studies in two species by different routes of administration. No increases in the number of revertants were observed in the bacterial reversion assay. In one micronucleus study, HCC-230fa was administered by inhalation to rats as part of a 90-day study at doses indicated above. For the second study, ICR mice were given a single ip dose at 0, 166, 330, or 660 mg/kg. In both micronucleus studies, a significant increase in micronucleated erythrocytes was observed. The results of these studies suggest that HCC-230fa affects rapidly dividing cells and may have long-term consequences for occupational exposures.


Asunto(s)
Anomalías Inducidas por Medicamentos , Desarrollo Embrionario y Fetal/efectos de los fármacos , Hidrocarburos Clorados/toxicidad , Mutágenos/toxicidad , Teratógenos/toxicidad , Administración por Inhalación , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Femenino , Hidrocarburos Clorados/administración & dosificación , Exposición Materna , Pruebas de Micronúcleos , Exposición Profesional , Embarazo , Ratas , Ratas Sprague-Dawley , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética
10.
Artículo en Inglés | MEDLINE | ID: mdl-12852482

RESUMEN

BACKGROUND: A review of the nonsteroidal anti-inflammatory drug (NSAID) literature suggested occurrences of low-level incidences of cardiovascular and midline defects in rabbit fetuses exposed in utero. Aspirin (acetylsalicylic acid, ASA) is a widely used NSAID that irreversibly inhibits cyclooxygenases (COXs) 1 and 2. ASA has been studied extensively in rats and has consistently increased low-incidence cardiovascular malformations and defects in midline closure. The objectives of the current study were to comprehensively define the developmental toxicology profile of ASA in rabbits by using a dosing paradigm encompassing the period of organogenesis and to test the hypothesis that maternal gastrointestinal toxicity after repeated dose administrations hampers the detection of low-incidence malformations with ASA in rabbits by limiting ASA administration to sensitive windows for cardiovascular development and midline closure. METHODS: ASA was administered to pregnant New Zealand White rabbits from gestation days (GDs) 7 to 19 at dose levels of 125, 250, and 350 mg/kg per day and as single doses of 500, 750, or 1000 mg/kg on GD 9, 10, or 11. Cesarean sections were performed on GD 29, and the fetuses were examined for external, visceral and skeletal development. RESULTS: In the repeated dose study, maternal toxicity was exhibited in the 250- and 350-mg/kg per day groups by mortality and decreased food consumption and body weight gain. In the single dose studies, maternal toxicity was exhibited at all doses by reductions in body weight gain and food consumption for 3 days after treatment. Fetal body weight was significantly reduced in the repeated dose study at 350 mg/kg per day. Fetal weights were not affected by single doses of ASA on GD 9, 10, or 11. There were no treatment-related external, visceral or skeletal malformations associated with ASA administration throughout organogenesis or with single doses administered during critical developmental windows. CONCLUSION: These findings supported previous work demonstrating that ASA is not teratogenic in rabbits, as opposed to rats, even when large doses are administered on single days during specific windows of development.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Antiinflamatorios no Esteroideos/toxicidad , Aspirina/toxicidad , Organogénesis/efectos de los fármacos , Teratógenos/toxicidad , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Aspirina/administración & dosificación , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Ingestión de Alimentos/efectos de los fármacos , Femenino , Peso Fetal/efectos de los fármacos , Exposición Materna/efectos adversos , Embarazo , Conejos , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología , Factores de Tiempo
11.
Artículo en Inglés | MEDLINE | ID: mdl-12852483

RESUMEN

BACKGROUND: A review of the scientific literature suggested the occurrence of low-level incidences of ventricular septal defect (VSD) and midline defect (MD) in rat fetuses and diaphragmatic hernia (DH), VSD, and MD in rabbit fetuses after maternal exposure to nonsteroidal anti-inflammatory drugs (NSAIDs). Aspirin, an NSAID that irreversibly inhibits cyclooxygenase 1 (COX-1) and COX-2, induces DH, VSD, and MD when administered as one dose during the sensitive periods of development in rats. Unlike aspirin, other NSAIDs, including selective COX-2 inhibitors, reversibly inhibit COX activity. To evaluate whether the dysmorphogenesis observed after maternal NSAID exposure correlates with COX-1 or COX-2 inhibition, a series of compounds with different capacities to inhibit COX-1 and COX-2 were administered to pregnant rats and rabbits during the sensitive period for heart development and midline closure. METHODS: The compounds selected, ranked from the most COX-2 selective to the most COX-1 selective based onCOX inhibition in a human whole blood assay, were CJ-19,209, meloxicam, diclofenac, diflunisal, ibuprofen, and ketorolac. Rat dams were treated on gestation days (GDs) 9 and 10, and rabbit does were treated on GDs 9, 10, and 11. The doses selected for evaluation represented the maximum tolerable dose for the compound, with the exception of CJ-19,209, which was dosed at 1000 mg/kg. Fetuses were collected by cesarean section on GDs 21 and 29 for rats and rabbits, respectively, and all fetuses were examined for external and visceral developmental anomalies. RESULTS: In rabbits, diflunisal induced DH, VSD, and MD (omphalocele) and single incidences of VSD and MD (gastroschisis) were noted in the ibuprofen group; no other developmental findings were associated with treatment. In rats, ibuprofen, diflunisal, and ketorolac induced increases in the incidence of VSD. In general the induction of developmental defects was associated with compounds that selectively inhibit COX-1 or have a high ratio of COX-1 to COX-2 inhibition. CONCLUSIONS: Inhibition of COX-1 may be involved in the disruption of heart development, whereas the selective inhibition of COX-2 (as assessed with CJ-19,209) appears to have no effect on heart development and midline closure in rats and rabbits.


Asunto(s)
Pared Abdominal/anomalías , Anomalías Inducidas por Medicamentos/etiología , Antiinflamatorios no Esteroideos/toxicidad , Inhibidores de la Ciclooxigenasa/toxicidad , Defectos del Tabique Interventricular/inducido químicamente , Teratógenos/toxicidad , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Peso Corporal/efectos de los fármacos , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/administración & dosificación , Esquema de Medicación , Ingestión de Alimentos/efectos de los fármacos , Femenino , Peso Fetal/efectos de los fármacos , Gastrosquisis/inducido químicamente , Gastrosquisis/embriología , Defectos del Tabique Interventricular/embriología , Hernia Umbilical/inducido químicamente , Hernia Umbilical/embriología , Isoenzimas/antagonistas & inhibidores , Proteínas de la Membrana , Embarazo , Prostaglandina-Endoperóxido Sintasas , Ratas , Ratas Sprague-Dawley , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología
12.
Food Chem Toxicol ; 41(5): 611-9, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12659713

RESUMEN

In order to establish the safety of veterinary drug residues in human food, a number of toxicological evaluations are required, including the assessment of potential risks to development. In the light of the use of developmental toxicity testing for risk characterization, we evaluated whether conducting these tests in more than one species was redundant. Review of the published Summary Reports of recommendations of the EU Committee for Veterinary Medicinal Products supplemented with data from the Joint FAO/WHO Expert Committee on Food Additives (JECFA) reports on veterinary drug residues in food identified 120 compounds, of which 105 had teratogenicity information in more than one species. The analysis of these compounds, representing a variety of chemical and pharmacological classes, provided justification for consideration of the use of a tiered approach for developmental toxicity evaluation of veterinary drugs for food-producing animals. The tiered approach begins with developmental toxicity testing in a rodent species, preferably the rat. If teratogenicity is observed, no testing in a second species would be required, except under specific circumstances where the ADI is determined based on the NOEL from this study. If a negative or an equivocal result for teratogenicity were observed in the rodent, then a developmental test in a second species, preferably the rabbit, would be conducted. The tiered approach provides thorough hazard identification, based on the use of a second species for compounds negative for teratogenicity in the rodent, and maintains maximum public protection based on the extremely low potential for human exposure to these compounds, while making a genuine attempt to limit unnecessary animal testing.


Asunto(s)
Animales Domésticos , Contaminación de Alimentos , Guías como Asunto , Teratógenos/toxicidad , Pruebas de Toxicidad/métodos , Drogas Veterinarias/efectos adversos , Anomalías Inducidas por Medicamentos , Animales , Humanos , Conejos , Proyectos de Investigación , Medición de Riesgo , Roedores , Seguridad , Drogas Veterinarias/uso terapéutico
13.
Drug Chem Toxicol ; 25(4): 481-96, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12378954

RESUMEN

Peroxisome proliferators are a class of nongenotoxic rodent hepatocarcinogens that cause peroxisome proliferation and liver tumors when administered to rats and mice; but other species, including guinea pigs, dogs, and primates are less sensitive or refractory to the induction of peroxisome proliferation. Therefore, rodent peroxisome proliferators are not believed to pose a hepatocarcinogenic hazard to humans. Some peroxisome proliferators produce developmental toxicity in rats that is expressed as suppressed postnatal growth. To evaluate the relevance of the rat developmental effect to primates, groups of 4 lactating female Rhesus monkeys and their infants were exposed for 6 h/day, 7 days/week for 3 weeks to air or 1000 ppm HCFC-123. Animals were evaluated for clinical signs, body weights, clinical pathology parameters, and biochemical and pathological evaluations of liver biopsy samples. The effect of HCFC-123 exposure on milk quality (protein and fat concentration) was evaluated. The concentrations of HCFC-123 and the major metabolite, trifluoroacetic acid (TFA), were measured in the blood of the mothers and infants and in the milk. Exposure of monkeys to 1000 ppm HCFC-123 did not result in exposure-related clinical observations, or changes in body weight, appetence and behavior. There were no exposure-related effects on serum triglycerides, cholesterol, or glucose levels. HCFC-123 and TFA were present in milk, although maternal HCFC-123 exposure did not affect milk protein and fat content. In general, HCFC-123 was not detected in maternal or infant blood. TFA was detected in the majority of the mothers and TFA levels in infants ranged from 2 to 6 times higher than levels in the corresponding maternal blood. A pharmacokinetic analysis in a maternal animal indicated a peak concentration of TFA at approximately 1 h post-exposure, with a half-life of approximately 20 h. Liver microsomal P450 and peroxisome oxidase activities showed exposure-related decreases in CYP4A1 and CYP2E1 and acyl-CoA oxidase for animals exposed to HCFC-123. Microscopic evaluation of maternal liver from HCFC-123 exposed animals revealed mild to moderate centrilobular hepatocyte vacuolation, trace to mild centrilobular necrosis, and trace to mild subacute inflammation. The histopathological damage and altered hepatic biochemical activities produced by HCFC-123 in monkeys are not consistent with the HCFC-123 peroxisome proliferation response observed in rat livers. These findings demonstrate that HCFC-123 is not a peroxisome proliferator in adult Rhesus monkeys and postnatal exposure to HCFC-123 does not affect body weight of nursing infant monkeys.


Asunto(s)
Clorofluorocarburos/toxicidad , Lactancia/efectos de los fármacos , Proliferadores de Peroxisomas/toxicidad , Administración por Inhalación , Animales , Animales Recién Nacidos , Animales Lactantes/crecimiento & desarrollo , Peso Corporal/efectos de los fármacos , Clorofluorocarburos/administración & dosificación , Clorofluorocarburos/farmacocinética , Clorofluorocarburos de Etano , Femenino , Semivida , Exposición por Inhalación , Hígado/efectos de los fármacos , Hígado/patología , Macaca mulatta , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Leche/química , Proliferadores de Peroxisomas/administración & dosificación , Proliferadores de Peroxisomas/farmacocinética , Ácido Trifluoroacético/análisis , Ácido Trifluoroacético/metabolismo
14.
Drug Chem Toxicol ; 25(3): 255-66, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12173247

RESUMEN

Some peroxisome proliferators have been reported to reduce body weight gain in suckling rats, possibly through a lactational effect. Decreases in milk production or nutritional quality, either as a result of peroxisome proliferator-induced reductions in lipid content or alterations in the hormonal milieu necessary for milk production, could result in pup growth retardation. Wyeth-14,643 (WY) is hypolipidemic agent and a potent inducer of hepatic peroxisome proliferation in rats and mice. As is commonly seen with rodent hepatic peroxisome proliferators, WY produces minimal or no peroxisome induction in guinea pigs or non-human primates. Goats are an excellent model for studying lactation, however, their sensitivity to peroxisome proliferating chemicals is not known. The present study was performed to assess the sensitivity of goats to the hypolipidemic and peroxisome proliferator properties of WY and to determine the effects of WY on milk quantity and quality. Six lactating adult female goats were assigned to either control or treated groups. Goats in the treated group were administered WY (40 mg/kg/day) for 14 consecutive days. The goats were milked twice daily in order to maintain lactation and the quantity of milk collected was recorded. Milk quality was evaluated by determining the content of total fat, protein, and carbohydrate in milk samples collected following 7 and 14 days of treatment. WY administration had no effects on final body weight, liver weight or, gross and histopathological findings. Milk quantity and quality were unaffected by treatment. Serum cholesterol and triglyceride levels were reduced by 25% compared to controls, although only the difference in cholesterol was statistically significant. Hepatic beta-oxidation (3 x control) and aromatase (1.5 x control) activities were significantly greater in the treatment group; however, there was no treatment-related effect in the total content of hepatic cytochrome P450. There was no difference in aromatase activity in a pooled ovarian microsome sample. Milk estradiol and prolactin concentrations were not affected by treatment. These findings indicate that goats are weak responders to the hepatic peroxisome proliferator effects of WY. Additionally, the slight serum hypolipidemic effect does not impact milk production or nutritional value.


Asunto(s)
Lactancia/efectos de los fármacos , Leche/química , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Peso Corporal , Estradiol/análisis , Femenino , Cabras , Lípidos , Microcuerpos , Ratas
15.
Drug Chem Toxicol ; 24(3): 259-71, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11452398

RESUMEN

The developmental toxicity of tetramethylurea (TMU) was assessed in rats by inhalation exposure of the test material over days 6-20 of gestation. Groups of 25 mated female Crl:CD BR rats were exposed whole-body for 6 hours/day to concentrations of either 0, 2, 20 or 100 ppm TMU. The dams were euthanized on day 21 and the offspring were weighted, sexed, and examined for external, visceral, and skeletal alterations. Maternal toxicity was demonstrated at both 20 and 100 ppm. Maternal body weights, weight changes, and food consumption were statistically significantly reduced at these concentrations; effects were more pronounced at 100 ppm. There was evidence of developmental toxicity only at 100 ppm. The only finding was a decrease in mean fetal weight. No fetal malformations or variations occurred in fetuses derived from rats exposed to all 3 test concentrations (up to 100 ppm). The maternal no-observed-effect-level (NOEL) was 2 ppm, the fetal NOEL was 20 ppm. Thus, TMU was not considered to be uniquely toxic to the rat conceptus.


Asunto(s)
Desarrollo Embrionario y Fetal/efectos de los fármacos , Compuestos de Metilurea/toxicidad , Solventes/toxicidad , Administración por Inhalación , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Peso Fetal/efectos de los fármacos , Exposición por Inhalación , Compuestos de Metilurea/administración & dosificación , Compuestos de Metilurea/análisis , Nivel sin Efectos Adversos Observados , Proyectos Piloto , Embarazo , Ratas , Ratas Sprague-Dawley , Solventes/administración & dosificación
16.
Toxicol Sci ; 62(1): 155-65, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11399803

RESUMEN

Male and female rats were exposed by inhalation (whole body) to HCC-230fa (1,1,1,3,3,3-hexachloropropane) for 6 h/day, 5 days/week over a 15-week period. Concentrations of 0, 0.50, 2.5, and 25 ppm were studied. A total of eight groups/sex were exposed. Four groups of male and four groups of female rats were used to measure clinical signs and growth, clinical pathology, and tissue pathology. The remaining four groups of male rats were used for immunotoxicological and sperm assessment evaluations, and the remaining four groups of female rats were used for immunotoxicological evaluation. Following the exposure period, surviving male rats were kept for a 1- or 3-month recovery period. Male and female rats exposed to 25 ppm had lower mean body weights, mean body weight gains, and food consumption during the exposure period. Male and female rats exposed to 25 ppm and sacrificed immediately after the exposure period had minimally decreased total leukocyte and lymphocyte counts. These changes were considered to be marginally adverse. Pathologic examination revealed hepatocellular hypertrophy in 0-day recovery males and an increased incidence and/or severity in chronic progressive nephropathy in 0-day, 1-month recovery, and 3-month recovery males at 25 ppm. No other pathological changes, including the testis, epididymis, and other accessory sex organs, were noted in rats during the study. Evaluation of sperm parameters at the end of the exposure period showed statistically significant decreases in epididymal sperm number per cauda epididymis, percent motile sperm, and percent normal sperm morphology at 25 ppm. The biological significance of the slight changes observed in the sperm parameters in the absence of histopathological changes is unclear. After a 1-month recovery period, no biologically significant differences in sperm parameters were noted at 25 ppm compared with controls. Exposure to HCC-230fa did not significantly alter the primary humoral immune response to sheep red blood cell (SRBC). Under the conditions of this study, the no-observed-adverse-effect level (NOAEL) was considered to be 2.5 ppm.


Asunto(s)
Hidrocarburos Clorados/toxicidad , Exposición Profesional , Solventes/toxicidad , Administración por Inhalación , Animales , Formación de Anticuerpos/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Recuento de Linfocitos , Linfocitos/efectos de los fármacos , Masculino , Nefrosis/inducido químicamente , Nefrosis/patología , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Solventes/administración & dosificación , Espermatozoides/efectos de los fármacos , Espermatozoides/patología , Aumento de Peso/efectos de los fármacos
17.
Drug Chem Toxicol ; 24(2): 87-101, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11360437

RESUMEN

The potential developmental toxicity of 3-aminopentanenitrile (3-APN) was assessed in rats. Groups of 25 time-mated female Crl:CD(SD)IGS BR rats were orally gavaged at daily dose levels of 0, 5, 30, 100 or 300 mg/kg over days 6-20 of gestation (days 6-20G); the day of copulation plug detection was designated day 0G. The dams were euthanized on day 21G and their abdominal and thoracic viscera were examined grossly. The fetuses were weighed, sexed, and examined for external, visceral, and skeletal alterations. Evidence of maternal and developmental toxicity was seen at 100 and 300 mg/kg. Regarding maternal toxicity, there were compound-related, statistically significant reductions in maternal body weight and food consumption at 100 and 300 mg/kg. The incidence of alopecia was significantly increased at these levels as well. Regarding developmental toxicity, mean fetal weight was slightly but significantly reduced at 100 and 300 mg/kg. In addition, at 300 mg/kg, there were significant increases in several skeletal variations (wavy ribs and skull, rib, and vertebral ossification delays) consistent with developmental delay. There was no evidence of either maternal or developmental toxicity at 5 or 30 mg/kg. Thus, the maternal and developmental no-observed-effect level (NOEL) was 30 mg/kg. Because developmental toxicity was observed only after administration of doses that also produced signs of maternal toxicity, 3-APN is not considered to be a selective developmental toxicant in the rat.


Asunto(s)
Desarrollo Embrionario y Fetal/efectos de los fármacos , Reproducción/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Peso Fetal/efectos de los fármacos , Edad Gestacional , Masculino , Embarazo , Ratas , Salivación/efectos de los fármacos
18.
Toxicol Sci ; 60(1): 44-55, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11222872

RESUMEN

Wyeth-14,643 (WY) and ammonium perfluorooctanoate (C8) belong to a diverse class of compounds which have been shown to produce hepatic peroxisome proliferation in rodents. From previous work, WY, but not C8, has been shown to produce hepatocellular carcinoma in rats, while C8 has been shown to produce Leydig cell adenomas. In addition, based on a review of bioassay data a relationship appears to exist between peroxisome-proliferating compounds and Leydig cell adenoma and pancreatic acinar cell hyperplasia/adenocarcinoma formation. To further investigate the relationship between peroxisome-proliferating compounds and hepatic, Leydig cell, and pancreatic acinar cell tumorigenesis, a 2-year feeding study in male CD rats was initiated to test the hypothesis that peroxisome proliferating compounds induce a tumor triad (liver, Leydig cell, pancreatic acinar cell), and to examine the potential mechanism for the Leydig cell tumors. The study was conducted using 50 ppm WY and 300 ppm C8. The concentration of WY in the diet was decreased to 25 ppm on test day 301 due to increased mortality. In addition to the ad libitum control, a second control was pair-fed to the C8 group. Interim sacrifices were performed at 1- or 3-month intervals. Peroxisome proliferation measured by beta-oxidation activity and cell proliferation were measured in the liver and testis at all time points and in the pancreas beginning at the 9-month time point (cell proliferation only). Serum hormone concentrations (estradiol, testosterone, LH, FSH, and prolactin) were also measured at each time point. Increased relative liver weights and hepatic beta-oxidation activity were observed in both the WY- and C8-treated rats at all time points. In contrast, hepatic cell proliferation was significantly increased only in the WY-treated group. Neither WY nor C8 significantly altered the rate of Leydig cell beta-oxidation or Leydig cell proliferation when compared to the control groups. Moreover, the basal rate of beta-oxidation in Leydig cells was approximately 20 times less than the rate of hepatic beta-oxidation. There were no biologically meaningful differences in serum testosterone, FSH, prolactin, or LH concentrations in the WY- and C8-treated rats when compared to their respective controls. There were, however, significant increases in serum estradiol concentrations in the WY- and C8-treated rats at 1, 3, 6, 9, 15, 18, and 21 months. At 12 months, only the C8-treated rats had elevated serum estradiol concentrations when compared to the pair-fed control. Histopathological evaluation revealed compound-related increases in liver, Leydig cell, and pancreatic acinar cell tumors in both WY- and C8-treated rats. The data support the hypothesis that the peroxisome-proliferating compounds induce the previously described tumor triad. In addition, both C8 and WY produced a sustained increase in serum estradiol concentrations that correlated with the potency of the 2 compounds to induce Leydig cell tumors (i.e., WY caused a more consistent sustained increase in serum estradiol throughout the entire study, and more specifically at the end of the study, than did C8). This study suggests that estradiol may play a role in enhancement of Leydig cell tumors in the rat, and that peroxisome proliferators may induce tumors via a non-LH type mechanism.


Asunto(s)
Caprilatos/toxicidad , Carcinógenos/toxicidad , Fluorocarburos/toxicidad , Neoplasias Experimentales/inducido químicamente , Proliferadores de Peroxisomas/toxicidad , Pirimidinas/toxicidad , Adenocarcinoma/inducido químicamente , Adenocarcinoma/patología , Animales , Pruebas de Carcinogenicidad , División Celular/efectos de los fármacos , Dieta , Estradiol/sangre , Hormona Folículo Estimulante/sangre , Tumor de Células de Leydig/inducido químicamente , Tumor de Células de Leydig/patología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Longevidad/efectos de los fármacos , Hormona Luteinizante/sangre , Neoplasias Experimentales/sangre , Neoplasias Experimentales/patología , Tamaño de los Órganos/efectos de los fármacos , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/patología , Prolactina/sangre , Ratas , Ratas Endogámicas , Testosterona/sangre
19.
Drug Chem Toxicol ; 22(4): 569-82, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10536749

RESUMEN

The developmental toxicity of glycolic acid was assessed in rats by orally administering solutions of the test material in water over days 7-21 of gestation (the day of copulation plug detection was defined as day 1 of gestation). Groups of 25 mated female Crl: CD BR rats were gavaged at daily dose levels of 0, 75, 150, 300 or 600 mg/kg. The dams were euthanized on day 22 and the offspring were weighed, sexed, and examined for external, visceral, and skeletal alterations. Clear evidence of maternal toxicity was demonstrated at 600 mg/kg; adverse clinical observations were statistically significantly increased (wheezing/lung noise, abnormal gait/staggering, lethargy). In addition, maternal body weights, weight changes, and food consumption were statistically significantly reduced at this dose level. Marginal evidence of maternal toxicity was demonstrated at 300 mg/kg; wheezing/lung noise similar to that seen at 600 mg/kg was observed in 2 of 25 dams. This increase approached statistical significance (p = 0.0553). There was marked evidence of developmental toxicity at 600 mg/kg. Mean fetal weight was statistically significantly reduced while the incidences of skeletal (ribs, vertebra, and sternebra) malformations and variations were statistically significantly increased. At 300 mg/kg/day, there was a slight (2 affected fetuses from 2 litters) increase in the incidence of two skeletal malformations: fused ribs and fused vertebra. Although these increases were not statistically significant (p = 0.0555), they were consistent with findings seen at 600 mg/kg/day and thus were considered relevant. There was no other evidence of developmental toxicity at 300 mg/kg/day nor was any developmental toxicity seen at 150 or 75 mg/kg/day. Thus, the maternal and developmental no-observed-effect level (NOEL) was considered 150 mg/kg.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Huesos/anomalías , Desarrollo Embrionario y Fetal/efectos de los fármacos , Glicolatos/toxicidad , Queratolíticos/toxicidad , Anomalías Inducidas por Medicamentos/patología , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Feto/anomalías , Feto/efectos de los fármacos , Glicolatos/administración & dosificación , Queratolíticos/administración & dosificación , Masculino , Nivel sin Efectos Adversos Observados , Embarazo , Ratas , Reproducción/efectos de los fármacos
20.
Food Chem Toxicol ; 37(5): 565-7, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10456686

RESUMEN

Pentane (CAS No. 109-66-0) is a widely-used chemical that finds many industrial applications. As part of a program looking at the inhalation toxicity of pentane, we studied the potential effects of the chemical on the developing embryo. This communication reports the findings of a limited study which was used to determine whether a full-scale developmental toxicity study was necessary. Groups of 7 to 8 pregnant rats were exposed by inhalation to pentane, 6 hr/day from gestation days 6 through 15 at concentrations of 0 (control), 1000, 3000 and 10,000 ppm, Maternal body weights, clinical signs and food consumption were measured; foetuses were weighed and examined for gross external development. Skeletal and internal organ evaluations were not performed. There were no effects in either the maternal or fetal rats (at concentrations up to and including 10,000 ppm) that could be associated with pentane exposure. Based on those findings, we did not conduct a full-spectrum developmental toxicity study in rats, but concluded that it is unlikely that the foetus would be adversely affected by pentane exposure.


Asunto(s)
Pentanos/toxicidad , Administración por Inhalación , Animales , Desarrollo Embrionario y Fetal/efectos de los fármacos , Femenino , Masculino , Intercambio Materno-Fetal , Embarazo , Ratas , Pruebas de Toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...