Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
J Toxicol Sci ; 46(7): 311-317, 2021.
Article in English | MEDLINE | ID: mdl-34193768

ABSTRACT

The approved drug thalidomide is teratogenic in humans, nonhuman primates, and rabbits but not in rodents. The extensive biotransformation of 5'-hydroxythalidomide after oral administration of thalidomide (250 mg/kg) in rats was investigated in detail using liquid chromatography-tandem mass spectrometry. Probable metabolites 5'-hydroxythalidomide sulfate and glucuronide were extensively formed, with approximately tenfold and onefold peak areas, respectively, to the primary 5'-hydroxythalidomide measured using authentic standards. As a minor metabolite, 5-hydroxythalidomide was also detected. The output of simplified physiologically based pharmacokinetic rat models was consistent with the observed in vivo data under a metabolic ratio of 0.05 for the hepatic intrinsic clearance of thalidomide to unconjugated 5'-hydroxythalidomide. The aggregate of unconjugated and sulfate/glucuronide conjugated 5'-hydroxythalidomide forms appear to be the predominant metabolites in rats. Two hours after oral administration of thalidomide (100 mg/kg) to chimeric mice humanized with four different batches of genotyped human hepatocytes, the plasma concentration ratios of 5-hydroxythalidomide to 5'-hydroxythalidomide were correlated with replacement indexes of human liver cells previously transplanted in immunodeficient mice. These results indicate that rodent livers mediate thalidomide primary oxidation, leading to extensive deactivation in vivo to unconjugated/conjugated 5'-hydroxythalidomide and suggest that thalidomide activation might be dependent on the humanized livers in mice transplanted with human hepatocytes.


Subject(s)
Hepatocytes/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Teratogens/pharmacokinetics , Teratogens/toxicity , Thalidomide/pharmacokinetics , Thalidomide/toxicity , Animals , Humans , Male , Metabolic Networks and Pathways , Mice , Models, Animal , Rats , Species Specificity , Teratogens/metabolism , Thalidomide/analogs & derivatives , Thalidomide/metabolism
2.
Toxicol Appl Pharmacol ; 414: 115424, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33524444

ABSTRACT

For the determination of acute toxicity of chemicals in zebrafish (Danio rerio) embryos, the OECD test guideline 236, relative to the Fish Embryo Toxicity Test (FET), stipulates a dose-response analysis of four lethal core endpoints and a quantitative characterization of abnormalities including their time-dependency. Routinely, the data are analyzed at the different observation times separately. However, observations at a given time strongly depend on the previous effects and should be analyzed jointly with them. To solve this problem, we developed multistate models for occurrence of developmental malformations and live events in zebrafish embryos exposed to eight concentrations of valproic acid (VPA) the first five days of life. Observations were recorded daily per embryo. We statistically infer on model structure and parameters using a numerical Bayesian framework. Hatching probability rate changed with time and we compared five forms of its time-dependence; a constant rate, a piecewise constant rate with a fixed hatching time at 48 h post fertilization, a piecewise constant rate with a variable hatching time, as well as a Hill and Gaussian form. A piecewise constant function of time adequately described the hatching data. The other transition rates were conditioned on the embryo body concentration of VPA, obtained using a physiologically-based pharmacokinetic model. VPA impacted mostly the malformation probability rate in hatched and non-hatched embryos. Malformation reversion probability rates were lowered by VPA. Direct mortality was low at the concentrations tested, but increased linearly with internal concentration. The model makes full use of data and gives a finer grain analysis of the teratogenic effects of VPA in zebrafish than the OECD-prescribed approach. We discuss the use of the model for obtaining toxicological reference values suitable for inter-species extrapolation. A general result is that complex multistate models can be efficiently evaluated numerically.


Subject(s)
Abnormalities, Drug-Induced/etiology , Models, Biological , Teratogens/toxicity , Toxicity Tests, Acute , Valproic Acid/toxicity , Abnormalities, Drug-Induced/embryology , Animals , Bayes Theorem , Computer Simulation , Dose-Response Relationship, Drug , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Numerical Analysis, Computer-Assisted , Teratogens/pharmacokinetics , Toxicokinetics , Valproic Acid/pharmacokinetics , Zebrafish/embryology
3.
Toxicol Lett ; 333: 71-79, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32768651

ABSTRACT

All vitamin K antagonist active substances used as rodenticides were reclassified in 2016 by the European authorities as active substances "toxic for reproduction", using a "read-across" alternative method based on warfarin, a human vitamin K antagonist drug. Recent study suggested that all vitamin K antagonist active substances are not all teratogenic. Using a neonatal exposure protocol, warfarin evokes skeletal deformities in rats, while bromadiolone, a widely used second-generation anticoagulant rodenticide, failed to cause such effects. Herein, using a rat model we investigated the mechanisms that may explain teratogenicity differences between warfarin and bromadiolone, despite their similar vitamin K antagonist mechanism of action. This study also included coumatetralyl, a first-generation active substance rodenticide. Pharmacokinetic studies were conducted in rats to evaluate a potential difference in the transfer of vitamin K antagonists from mother to fetus. The data clearly demonstrate that warfarin is highly transferred from the mother to the fetus during gestation or lactation. In contrast, bromadiolone transfer from dam to the fetus is modest (5% compared to warfarin). This difference appears to be associated to almost complete uptake of bromadiolone by mother's liver, resulting in very low exposure in plasma and eventually in other peripheric tissues. This study suggests that the pharmacokinetic properties of vitamin K antagonists are not identical and could challenge the classification of such active substances as "toxic for reproduction".


Subject(s)
4-Hydroxycoumarins/toxicity , Prenatal Exposure Delayed Effects/blood , Rodenticides/toxicity , Teratogenesis/drug effects , Teratogens/toxicity , Vitamin K/antagonists & inhibitors , Warfarin/toxicity , 4-Hydroxycoumarins/pharmacokinetics , Administration, Oral , Animals , Animals, Newborn , Animals, Suckling , Female , Fetal Development/drug effects , Liver/drug effects , Liver/embryology , Liver/metabolism , Male , Maternal Exposure , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rodenticides/pharmacokinetics , Teratogens/pharmacokinetics , Warfarin/pharmacokinetics
4.
Regul Toxicol Pharmacol ; 107: 104413, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31229519

ABSTRACT

The draft ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high dose in developmental and reproductive toxicity (DART) studies. In 2016, IQ DruSafe conducted an anonymous survey to identify industry practices and experiences related to pharmacokinetic assessments in DART studies in order to facilitate a pragmatic data-driven approach to development of an acceptable multiple of the clinical exposure to be proposed for dose selection in the guideline. Questions in the survey were designed to explore pharmacokinetic differences in pregnant versus non-pregnant animals, and to assess exposure levels attained in the absence of maternal toxicity as well as DART outcomes in animal studies associated with those exposures. Small molecule and therapeutic proteins were analyzed separately. The key findings for small molecules were: a) differences in exposures between pregnant and non-pregnant animals were generally ≤3-fold, b) Cmax or AUC exposures ≥25-fold the clinical exposure were achieved in the absence of maternal toxicity for 31% and 23% of rat and rabbit developmental toxicity studies, respectively, and c) only 3.3% (5/153) and 1.6% (2/128) of the developmental toxicity studies were positive for malformations or embryofetal lethality in rats and rabbits, respectively, that were not observed until exposure margins were ≥25-fold.


Subject(s)
Drug Evaluation, Preclinical , Teratogens/pharmacokinetics , Teratogens/toxicity , Toxicity Tests , Animals , Drug Industry , Drug-Related Side Effects and Adverse Reactions , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Haplorhini , Pregnancy , Rabbits , Rats , Reproduction/drug effects , Surveys and Questionnaires
5.
Eur J Med Chem ; 176: 456-475, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31128448

ABSTRACT

H2S donors are substitutes of H2S with various biological activities like inhibiting the inflammatory response and protecting myocardial cells from injury. In order to confirm whether the H2S donors have drug-like properties, two series thiophosphamide H2S donors were evaluated including toxicity, bioactivity and pharmacokinetic properties in vivo and in vitro. The following results were obtained. Firstly, all the compounds released H2S under measuring condition; with the increase of pH value, the H2S release rate of all the compounds decreased and the amount reduced, but pH value had little effect on the maximum release of H2S. Secondly, in the organs and tissues of rats, the compounds released H2S in the same way as in PBS. In plasma, compound 1 reached the Cmax after administration 55 min, and no compound 1 was detected after 12 h; for compound 18, the Cmax reached only after administration 100 min, and after 6 h, compound 18 was not detected; in organs and tissues, the H2S-release rates were different from those in PBS, but the mechanism of H2S release was the same. Thirdly, in the test of toxicity, all the compounds displayed low toxicities to 5 cancer cells and W138 cell lines; compounds 1 and 18 had slight effect on the physiological tissue and function of rat liver at low concentration; the compounds had almost no effect on the hatching rate, survival rate of zebrafish embryos, and the spontaneous movement of zebrafish embryos at below 0.5 µM, but when they were over 1 µM, the compounds displayed inhibitory effect in the manner of concentration dependence. Fourthly, in the course of anti-inflammatory test, all the tested compounds significantly reduced the level of TNF-α and increased the level of IL-10; when they were 100 µM, the levels of IL-10 were three times as high as those in the control group. Among them, compounds 10 and 18 displayed stronger activities than the others. In addition, the compounds protected H9c2 cells from injure and improved myocardial injury through anti-oxidation pathway. In summary, the compounds have druglike properties due to low toxicity, better activity and good pharmacokinetic property. Therefore, they have potential to be as candidates to investigate further.


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Cardiotonic Agents/pharmacokinetics , Hydrogen Sulfide/metabolism , Organothiophosphorus Compounds/pharmacokinetics , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Cardiotonic Agents/chemical synthesis , Cardiotonic Agents/chemistry , Cardiotonic Agents/toxicity , Cell Line, Tumor , Drug Liberation , Female , Humans , Hydrogen Sulfide/blood , Hydrogen Sulfide/chemistry , Hydrogen-Ion Concentration , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Models, Chemical , Myocardium/metabolism , Organothiophosphorus Compounds/chemical synthesis , Organothiophosphorus Compounds/chemistry , Organothiophosphorus Compounds/toxicity , RAW 264.7 Cells , Rats, Wistar , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Temperature , Teratogens/chemical synthesis , Teratogens/chemistry , Teratogens/pharmacokinetics , Teratogens/toxicity , Zebrafish
6.
Curr Pain Headache Rep ; 22(12): 80, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30291521

ABSTRACT

PURPOSE OF REVIEW: Migraine is a disabling and prevalent neurological disease, commonly affecting women during their reproductive years. It is crucial for providers to be able to adequately counsel women who are pregnant, planning pregnancy, or nursing, regarding preventive and abortive treatment options for episodic migraine. This review will discuss (1) the expected course of migraine during pregnancy and the post-partum period, (2) recommended preventive therapies for migraine during pregnancy and lactation, and (3) recommended abortive medications for migraine during pregnancy and lactation. RECENT FINDINGS: Recent research has indicated safety for triptan use during pregnancy and ibuprofen use during the first trimester of pregnancy. Considerations for use of emerging migraine-preventive treatment, such as non-invasive neurostimulators, are discussed. For clinical decision-making and patient counseling, it is important to understand both the limitations in determining teratogenic effects in humans and the principles affecting medication transmission from mother to breast milk.


Subject(s)
Lactation , Migraine Disorders/epidemiology , Pregnancy Complications/epidemiology , Adult , Drug-Related Side Effects and Adverse Reactions/epidemiology , Female , Humans , Infant, Newborn , Migraine Disorders/drug therapy , Pregnancy , Pregnancy Complications/therapy , Prevalence , Teratogens/pharmacokinetics
7.
Reprod Toxicol ; 81: 237-245, 2018 10.
Article in English | MEDLINE | ID: mdl-30149139

ABSTRACT

The potent hERG channel blocking drug ondansetron is used off-label for treatment of nausea and vomiting in early pregnancy. Some human epidemiological studies have associated ondansetron with fetal cardiovascular defects and orofacial clefts. This study investigated the effects of ondanestron on embryonic heart rhythm of gestational day (GD) 13 rat embryos in vitro and then integrated the results with published animal teratology, and animal and human pharmacokinetic studies to perform a risk evaluation. Ondansetron caused concentration dependent bradycardia and arrhythmia. Cardiovascular malformations in rats occurred at exposures slightly higher than those in early human pregnancy. Together the results suggest that ondansetron can have teratogenic potential in rats and humans mediated via hERG block and severe heart rhythm disturbances in the embryo. The risk may be increased in human pregnancy if additional risk factors are present such as hypokalemia.


Subject(s)
Antiemetics/toxicity , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ondansetron/toxicity , Teratogens/toxicity , Abnormalities, Drug-Induced/etiology , Animals , Antiemetics/pharmacokinetics , Cardiovascular Abnormalities/chemically induced , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Female , Heart/drug effects , Humans , Ondansetron/pharmacokinetics , Pregnancy , Rats, Sprague-Dawley , Teratogens/pharmacokinetics
8.
J Toxicol Sci ; 43(4): 267-273, 2018.
Article in English | MEDLINE | ID: mdl-29618715

ABSTRACT

Developmental toxicity is an adverse developmental outcome, i.e., death, malformation, growth retardation, or functional deficiency. Recently, alternative methods of assessing developmental toxicity using zebrafish (Danio rerio) as a preliminary screening have attracted attention because of their low cost and high throughput. However, most toxicity evaluations have been based on a chemical concentration in an aqueous solution, and the chemical concentrations in embryos/larvae and their temporal behavior have in most cases been unclear, regardless of differences of chemical hydrophobicity. In the present study, we selected three teratogens with different hydrophobicities (caffeine, CA, log Kow -0.07; sodium valproate, VA, log Kow 0.26 (pH 7.4); and diethylstilbestrol, DES, log Kow 5.07), and we measured their concentrations in embryos/larvae exposed to these chemicals every 24 hr post-fertilization (hpf) until 144 hpf. Kinetic analysis based on a one-compartment fish model that yields first order kinetics for CA and VA revealed that concentrations of both CA and VA in embryos/larvae increased gradually and became saturated by around 100 hpf. In contrast, DES concentrations in embryos/larvae reached a maximum at 48 or 72 hpf and then decreased gradually. The present study suggests that the temporal pattern of chemical concentrations is a function of the hydrophobicity of the chemicals.


Subject(s)
Caffeine/toxicity , Diethylstilbestrol/toxicity , Teratogens/toxicity , Valproic Acid/toxicity , Zebrafish/embryology , Animals , Caffeine/pharmacokinetics , Diethylstilbestrol/pharmacokinetics , Fertilization , Hydrophobic and Hydrophilic Interactions , Teratogens/pharmacokinetics , Time Factors , Valproic Acid/pharmacokinetics , Zebrafish/metabolism
9.
Toxicol Sci ; 157(2): 365-376, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28498972

ABSTRACT

With our recently developed in vitro physiologically based kinetic (PBK) modelling approach, we could extrapolate in vitro toxicity data to human toxicity values applying PBK-based reverse dosimetry. Ideally information on kinetic differences among human individuals within a population should be considered. In the present study, we demonstrated a modelling approach that integrated in vitro toxicity data, PBK modelling and Monte Carlo simulations to obtain insight in interindividual human kinetic variation and derive chemical specific adjustment factors (CSAFs) for phenol-induced developmental toxicity. The present study revealed that UGT1A6 is the primary enzyme responsible for the glucuronidation of phenol in humans followed by UGT1A9. Monte Carlo simulations were performed taking into account interindividual variation in glucuronidation by these specific UGTs and in the oral absorption coefficient. Linking Monte Carlo simulations with PBK modelling, population variability in the maximum plasma concentration of phenol for the human population could be predicted. This approach provided a CSAF for interindividual variation of 2.0 which covers the 99th percentile of the population, which is lower than the default safety factor of 3.16 for interindividual human kinetic differences. Dividing the dose-response curve data obtained with in vitro PBK-based reverse dosimetry, with the CSAF provided a dose-response curve that reflects the consequences of the interindividual variability in phenol kinetics for the developmental toxicity of phenol. The strength of the presented approach is that it provides insight in the effect of interindividual variation in kinetics for phenol-induced developmental toxicity, based on only in vitro and in silico testing.


Subject(s)
Biological Variation, Individual , Glucuronates/metabolism , Glucuronosyltransferase , Microsomes, Liver/enzymology , Models, Biological , Phenol/toxicity , Teratogens/toxicity , Computer Simulation , Dose-Response Relationship, Drug , Female , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , In Vitro Techniques , Kinetics , Microsomes, Liver/drug effects , Monte Carlo Method , Phenol/pharmacokinetics , Predictive Value of Tests , Teratogens/pharmacokinetics
10.
Environ Int ; 88: 53-59, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26708281

ABSTRACT

While the U.S. EPA has issued a draft report with a 1% TCDD effective dose (ED01) of 87.9pg/kg/day based on continuous integration of key scientific evidence, a detailed and comprehensive uncertainty analysis has not been well documented. In this study, a new estimate for ED01 was derived based on uncertainty analysis by quantitatively assessing the potential bias arising from the selection of kinetic models, dose-response models and cohorts. The cumulative serum lipid concentration (CSLC) and cumulative body burden (CBB) were reconstructed as dose metrics using a concentration- and age-dependent pharmacokinetic model (CADM), physiologically based pharmacokinetic model (PBPK), and age-dependent half-life model (FV), and the reconstructed dose metrics based on CADM and PBPK were generally higher than those based on the FV model. Three dose-response curves (linear, multiplicative and power) were used to link dose metrics and cancer risk to estimate ED01, and the linear model resulted in the lowest ED01, followed by the power model and multiplicative model, for the same cohort. Meanwhile, ED01 based on the CADM model was the highest, followed by those based on the PBPK model and first-order model. Finally, the ED01 was estimated to be 17.03±7.83pg/kg/day by statistically analyzing the distribution of ED01 values based on various kinetic models, cohorts and dose-response models. The study presented here strengthens the scientific basis for understanding the potential health implications of TCDD exposure.


Subject(s)
Environmental Pollutants/blood , Neoplasms , Occupational Exposure/adverse effects , Polychlorinated Dibenzodioxins/blood , Teratogens/toxicity , Body Burden , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Germany/epidemiology , Half-Life , Humans , Linear Models , Male , Models, Theoretical , Neoplasms/chemically induced , Neoplasms/mortality , Polychlorinated Dibenzodioxins/pharmacokinetics , Polychlorinated Dibenzodioxins/toxicity , Teratogens/pharmacokinetics , Uncertainty , United States/epidemiology
11.
Chem Res Toxicol ; 28(11): 2088-90, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26492539

ABSTRACT

Plasma concentrations of thalidomide and primary 5-hydroxylated metabolites including 5,6-dihydroxythalidomide and glutathione (GSH) conjugate(s) were investigated in chimeric mice with highly "humanized" liver cells harboring cytochrome P450 3A5*1. Following oral administration of thalidomide (100 mg/kg), plasma concentrations of GSH conjugate(s) of 5-hydroxythalidomide were higher in humanized mice than in controls. Simulation of human plasma concentrations of thalidomide were achieved with a simplified physiologically based pharmacokinetic model in accordance with reported thalidomide concentrations. The results indicate that the pharmacokinetics in humans of GSH conjugate and/or catechol primary 5-hydroxylated thalidomide contributing in vivo activation can be estimated for the first time.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hepatocytes/metabolism , Thalidomide/analogs & derivatives , Thalidomide/blood , Animals , Glutathione/metabolism , Humans , Hydroxylation , Liver/metabolism , Mice , Teratogens/pharmacokinetics , Thalidomide/pharmacokinetics
12.
Reprod Toxicol ; 55: 73-80, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25462785

ABSTRACT

In the EU collaborative project ChemScreen an alternative, in vitro assay-based test strategy was developed to screen compounds for reproductive toxicity. A toxicokinetic modeling approach was used to allow quantitative comparison between effective concentrations in the in vitro test battery and observations of developmental toxicity in vivo. This modeling strategy is based on (1) the definition of relevant observations of toxicity in vivo, (2) simulation of the corresponding systemic concentrations in vivo by toxicokinetic modeling, and (3) correction for differences in protein binding and lipid partitioning between plasma and in vitro test media. The test results of a feasibility study with a number of known reproductive toxicants has been described previously (Piersma et al. [15]). In the present paper, we take a more detailed look at the toxicokinetics of these compounds, and add the analysis of some compounds from subsequent studies. We discuss how the consideration of toxicokinetics allowed comparison between test systems with differing test medium composition, has helped to interpret the in vitro findings in light of in vivo observations, and to gain confidence in the predictive value of the test battery outcomes. The same toxicokinetic modeling strategy, in reverse order, can now be used for risk assessment purposes to predict toxic doses in vivo from effective concentrations in vitro.


Subject(s)
Models, Biological , Teratogens/pharmacokinetics , Teratogens/toxicity , Toxicity Tests , Animals , Biological Assay , Female , Pregnancy , Rats , Risk Assessment , Toxicokinetics
13.
Reprod Toxicol ; 55: 11-9, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25461900

ABSTRACT

Previously we showed a battery consisting of CALUX transcriptional activation assays, the ReProGlo assay, and the embryonic stem cell test, and zebrafish embryotoxicity assay as 'apical' tests to correctly predict developmental toxicity for 11 out of 12 compounds, and to explain the one false negative [7]. Here we report on applying this battery within the context of grouping and read across, put forward as a potential tool to fill data gaps and avoid animal testing, to distinguish in vivo non- or weak developmental toxicants from potent developmental toxicants within groups of structural analogs. The battery correctly distinguished 2-methylhexanoic acid, monomethyl phthalate, and monobutyltin trichloride as non- or weak developmental toxicants from structurally related developmental toxicants valproic acid, mono-ethylhexyl phthalate, and tributyltin chloride, respectively, and, therefore, holds promise as a biological verification model in grouping and read across approaches. The relevance of toxicokinetic information is indicated.


Subject(s)
Animal Testing Alternatives , Teratogens/toxicity , Toxicity Tests/methods , Animals , Cell Line , Cells, Cultured , Embryo, Nonmammalian/drug effects , Embryonic Stem Cells/drug effects , Genes, Reporter , Humans , Mice , Receptors, Estrogen/metabolism , Reproduction , Teratogens/classification , Teratogens/pharmacokinetics , Toxicokinetics , Zebrafish/embryology
14.
PLoS One ; 9(11): e112522, 2014.
Article in English | MEDLINE | ID: mdl-25383890

ABSTRACT

Polyunsaturated aldehydes (PUAs) are released by several diatom species during predation. Besides other attributed activities, these oxylipins can interfere with the reproduction of copepods, important predators of diatoms. While intensive research has been carried out to document the effects of PUAs on copepod reproduction, little is known about the underlying mechanistic aspects of PUA action. Especially PUA uptake and accumulation in copepods has not been addressed to date. To investigate how PUAs are taken up and interfere with the reproduction in copepods we developed a fluorescent probe containing the α,ß,γ,δ-unsaturated aldehyde structure element that is essential for the activity of PUAs as well as a set of control probes. We developed incubation and monitoring procedures for adult females of the calanoid copepod Acartia tonsa and show that the PUA derived fluorescent molecular probe selectively accumulates in the gonads of this copepod. In contrast, a saturated aldehyde derived probe of an inactive parent molecule was enriched in the lipid sac. This leads to a model for PUAs' teratogenic mode of action involving accumulation and covalent interaction with nucleophilic moieties in the copepod reproductive tissue. The teratogenic effect of PUAs can therefore be explained by a selective targeting of the molecules into the reproductive tissue of the herbivores, while more lipophilic but otherwise strongly related structures end up in lipid bodies.


Subject(s)
Aldehydes/chemistry , Aldehydes/pharmacokinetics , Copepoda/physiology , Gonads/drug effects , Aldehydes/pharmacology , Animals , Copepoda/drug effects , Female , Fluorescent Dyes/pharmacokinetics , Fluorescent Dyes/pharmacology , Organ Specificity , Rhodamines/pharmacokinetics , Teratogens/chemistry , Teratogens/pharmacokinetics , Teratogens/pharmacology
15.
Reprod Toxicol ; 49: 162-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25194688

ABSTRACT

Very late antigen-4 (VLA-4), which is concerned with cell-cell adhesion, plays important roles in development of the heart, and some VLA-4 antagonists cause cardiac anomalies. In this study, we evaluated the teratogenic potential of VLA-4 antagonist derivatives as screening, and investigated the conditions that induce cardiac anomalies. Seventeen compounds were orally administered to pregnant rats throughout the organogenesis period, and fetal examinations were performed. In addition, drug concentrations in the embryos were assayed. As a result, the incidence of ventricular septal defect (VSD) ranged from 0 to 100% depending on the compound. Plasma drug concentrations in the dams were related to increased incidence of VSD; however, these incidences were not increased when the concentration of the compound in the embryos at 24h after dosing was low. It is considered that continuous pharmacological activity in the embryo for more than 24h might disrupt closure of the ventricular septum.


Subject(s)
Heart Septal Defects, Ventricular/chemically induced , Integrin alpha4beta1/antagonists & inhibitors , Teratogens/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Fetal Development/drug effects , Male , Mice, Inbred ICR , Pregnancy , Rats , Structure-Activity Relationship , Teratogens/pharmacokinetics
16.
Reprod Toxicol ; 48: 115-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24859269

ABSTRACT

Studies in pregnant rabbits were conducted to evaluate if there are any differences in the uptake of thalidomide into the intrauterine compartment and developmental toxicity risk following oral and intravaginal administration. Thalidomide concentrations in maternal plasma, yolk sac cavity (YSC) fluid and embryo following intravaginal administration were 2- to 7-fold lower than their respective levels after oral administration. Ratios of thalidomide concentration in YSC fluid to maternal plasma were similar between these two routes, indicating no difference in uptake into the intrauterine compartment. A rabbit embryo-fetal development study using oral and intravaginal thalidomide administration at 2mg/kg/day (a dose >10,000-fold higher than the expected amount of thalidomide in human semen) did not result in any developmental abnormalities. These data demonstrated no preferential transfer mechanism of thalidomide from vagina to conceptus, and no additional embryo-fetal developmental toxicity risks with thalidomide exposure via the vaginal route.


Subject(s)
Embryo, Mammalian/drug effects , Fetus/drug effects , Teratogens/toxicity , Thalidomide/administration & dosage , Thalidomide/toxicity , Abnormalities, Drug-Induced , Administration, Intravaginal , Administration, Oral , Animals , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Female , Fetal Development/drug effects , Fetus/metabolism , Male , Maternal-Fetal Exchange , Pregnancy , Rabbits , Teratogens/pharmacokinetics , Thalidomide/blood , Thalidomide/pharmacokinetics , Uterus/metabolism , Vagina/metabolism , Yolk Sac/metabolism
17.
Med. clín (Ed. impr.) ; 142(8): 360-364, abr. 2014.
Article in Spanish | IBECS | ID: ibc-119697

ABSTRACT

La talidomida es un derivado sintético del ácido glutámico, introducido por primera vez en Alemania en 1956 como un medicamento de venta sin receta. La aprobación fue como sedante seguro, incluso a pequeñas dosis, no adictivo y sin efectos secundarios tales como debilidad motora, pero fue retirada de la circulación por asociarse a graves malformaciones en recién nacidos. Más tarde, laFood and Drug Administration aprobó su uso en el tratamiento del eritema nudoso leproso y también demostró eficacia en otros procesos dermatológicos refractarios tales como el prurigo actínico, la histiocitosis de Langerhans del adulto, la estomatitis aftosa, el síndrome de Behçet, la enfermedad del injerto contra el huésped, la sarcoidosis cutánea, el eritema multiforme, la infiltración cutánea linfocitaria de Jessner-Kanof, el sarcoma de Kaposi, el liquen plano, el lupus eritematoso sistémico, el melanoma, el prurigo nodular, el pioderma gangrenoso y otros. En mayo de 2006 fue aprobada para el tratamiento del mieloma múltiple. Nuevos análogos de la talidomida, como la lenalidomida, han sido desarrollados, aunque con escasa experiencia clínica. Este trabajo es una revisión de la historia, farmacología, mecanismo de acción, usos clínicos y efectos adversos de la talidomida y sus análogos (AU)


Thalidomide is a synthetic glutamic acid derivative first introduced in 1956 in Germany as an over the counter medications. It was thought to be one of the safest sedatives ever produced as it was effective in small doses, was not addictive, and did not have acute side-effects such as motor impairment, but was quickly removed from market after it was linked to cases of severe birth defects. The Food and Drug Administration approved use in the treatment of erythema nodosum leprosum. Further, it was shown its effectiveness in unresponsive dermatological conditions such as actinic prurigo, adult Langerhans cell hystiocytosis, aphthous stomatitis, Behçet syndrome, graft-versus-host disease, cutaneous sarcoidosis, erythema multiforme, Jessner-Kanof lymphocytic infiltration of the skin, Kaposi sarcoma, lichen planus, lupus erythematosus, melanoma, prurigo nodularis, pyoderma gangrenosum and others. In May 2006, it was approved for the treating multiple myeloma. New thalidomide analogues have been developed but lack clinical experience. This paper is a review of the history, pharmacology, mechanism of action, clinical applications and side effects of thalidomide and its analogues (AU)


Subject(s)
Humans , Thalidomide/therapeutic use , Hypnotics and Sedatives/therapeutic use , Thalidomide/adverse effects , Immunosuppressive Agents/pharmacokinetics , Teratogens/pharmacokinetics
18.
Mol Pharm ; 11(3): 1014-21, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24446970

ABSTRACT

This work aims to elucidate the mechanism by which N-methylpyrrolidone (NMP) enhances the skin permeation of a compound by combining experimental data with molecular dynamic (MD) simulations. The addition of 10% NMP significantly increased the propranolol (PR) permeation through the human epidermis (∼ 15 µg/cm(2) vs ∼ 30 µg/cm(2)) while resulting inefficacious on hydrocortisone (HC) diffusion. No significant alterations in the stratum corneum structure were found after the in vitro treatment of human epidermis with NMP dispersed in mineral oil or water by attenuated total reflectance Fourier transform infrared (ATR-FTIR) analyses. MD simulations revealed the formation of a complex by H-bonds and the π-π stacking interactions between the NMP's amido group and the drug's aromatic systems. The size of the depicted NMP/PR clusters was in line with the hydrodynamic radius derived by dynamic light scattering analyses (∼ 2.00 nm). Conversely, no interaction, and consequently cluster formation, between NMP and HC occurred. These results suggest that NMP is effective in enhancing the drug permeation through human epidermis by a cotransport mechanism when NMP/drug interaction occurs.


Subject(s)
Cell Membrane Permeability/drug effects , Drug Delivery Systems , Hydrocortisone/administration & dosage , Propranolol/administration & dosage , Pyrrolidinones/pharmacokinetics , Skin Absorption/drug effects , Skin/metabolism , Administration, Cutaneous , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Diffusion , Humans , Hydrocortisone/pharmacokinetics , Molecular Dynamics Simulation , Propranolol/pharmacokinetics , Pyrrolidinones/administration & dosage , Skin/drug effects , Spectroscopy, Fourier Transform Infrared , Teratogens/pharmacokinetics , Tissue Distribution , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacokinetics
19.
Epilepsia ; 55(2): 353-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24313671

ABSTRACT

OBJECTIVE: Valnoctamide (VCD), a central nervous system (CNS)-active chiral constitutional isomer of valpromide, the corresponding amide of valproic acid (VPA), is currently undergoing phase IIb clinical trials in acute mania. VCD exhibits stereoselective pharmacokinetics (PK) in animals and humans. The current study comparatively evaluated the pharmacodynamics (PD; anticonvulsant activity and teratogenicity) and PK of the four individual stereoisomers of VCD. METHODS: The anticonvulsant activity of VCD individual stereoisomers was evaluated in several rodent anticonvulsant models including maximal electroshock, 6 Hz psychomotor, subcutaneous metrazol, and the pilocarpine-induced and soman-induced status epilepticus (SE). The PK-PD (anticonvulsant activity) relationship of VCD stereoisomers was evaluated following intraperitoneal administration (70 mg/kg) to rats. Induction of neural tube defects (NTDs) by VCD stereoisomers was evaluated in a mouse strain that was highly susceptible to teratogen-induced NTDs. RESULTS: VCD had a stereoselective PK, with (2S,3S)-VCD exhibiting the lowest clearance, and consequently a twice-higher plasma exposure than all other stereoisomers. Nervertheless, there was less stereoselectivity in VCD anticonvulsant activity and each stereoisomer had similar median effective dose (ED)50 values in most models. VCD stereoisomers (258 or 389 mg/kg) did not cause NTDs. These doses are 3-12 times higher than VCD anticonvulsant ED50 values. SIGNIFICANCE: VCD displayed stereoselective PK that did not lead to significant stereoselective activity in various anticonvulsant rodent models. If VCD exerted its broad-spectrum anticonvulsant activity using a single mechanism of action (MOA), it is likely that it would exhibit a stereoselective PD. The fact that there was no significant difference between racemic VCD and its individual stereoisomers suggests that VCD's anticonvulsant activity is due to multiple MOAs.


Subject(s)
Amides/pharmacokinetics , Anticonvulsants/pharmacokinetics , Central Nervous System Stimulants/pharmacokinetics , Teratogens/pharmacokinetics , Valproic Acid/pharmacokinetics , Amides/chemistry , Amides/toxicity , Animals , Anticonvulsants/chemistry , Anticonvulsants/toxicity , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/toxicity , Male , Mice , Neural Tube Defects/chemically induced , Random Allocation , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/prevention & control , Stereoisomerism , Teratogens/chemistry , Teratogens/toxicity , Valproic Acid/chemistry , Valproic Acid/toxicity
20.
Arch Toxicol ; 87(9): 1709-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23943240

ABSTRACT

In vitro assays are often used for the hazard characterisation of compounds, but their application for quantitative risk assessment purposes is limited. This is because in vitro assays cannot provide a complete in vivo dose-response curve from which a point of departure (PoD) for risk assessment can be derived, like the no observed adverse effect level (NOAEL) or the 95 % lower confidence limit of the benchmark dose (BMDL). To overcome this constraint, the present study combined in vitro data with a physiologically based kinetic (PBK) model applying reverse dosimetry. To this end, embryotoxicity of phenol was evaluated in vitro using the embryonic stem cell test (EST), revealing a concentration-dependent inhibition of differentiation into beating cardiomyocytes. In addition, a PBK model was developed on the basis of in vitro and in silico data and data available from the literature only. After evaluating the PBK model performance, effective concentrations (ECx) obtained with the EST served as an input for in vivo plasma concentrations in the PBK model. Applying PBK-based reverse dosimetry provided in vivo external effective dose levels (EDx) from which an in vivo dose-response curve and a PoD for risk assessment were derived. The predicted PoD lies within the variation of the NOAELs obtained from in vivo developmental toxicity data from the literature. In conclusion, the present study showed that it was possible to accurately predict a PoD for the risk assessment of phenol using in vitro toxicity data combined with reverse PBK modelling.


Subject(s)
Animal Use Alternatives , Disinfectants/toxicity , Embryonic Stem Cells/drug effects , Models, Biological , Phenol/toxicity , Teratogens/toxicity , Animals , Biotransformation , Cell Differentiation/drug effects , Cell Line , Cytosol/enzymology , Cytosol/metabolism , Disinfectants/metabolism , Disinfectants/pharmacokinetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Humans , Kinetics , Liver/enzymology , Liver/metabolism , Male , Mice , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Organ Specificity , Phenol/metabolism , Phenol/pharmacokinetics , Rats , Risk Assessment/methods , Species Specificity , Teratogens/metabolism , Teratogens/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL