Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Affiliation country
Publication year range
1.
Arch Toxicol ; 97(4): 947-961, 2023 04.
Article in English | MEDLINE | ID: mdl-36795116

ABSTRACT

Drug-induced testicular injury (DITI) is one of the often-observed and challenging safety issues seen during drug development. Semen analysis and circulating hormones currently utilized have significant gaps in their ability to detect testicular damage accurately. In addition, no biomarkers enable a mechanistic understanding of the damage to the different regions of the testis, such as seminiferous tubules, Sertoli, and Leydig cells. MicroRNAs (miRNAs) are a class of non-coding RNAs that modulate gene expression post-transcriptionally and have been indicated to regulate a wide range of biological pathways. Circulating miRNAs can be measured in the body fluids due to tissue-specific cell injury/damage or toxicant exposure. Therefore, these circulating miRNAs have become attractive and promising non-invasive biomarkers for assessing drug-induced testicular injury, with several reports on their use as safety biomarkers for monitoring testicular damage in preclinical species. Leveraging emerging tools such as 'organs-on-chips' that can emulate the human organ's physiological environment and function is starting to enable biomarker discovery, validation, and clinical translation for regulatory qualification and implementation in drug development.


Subject(s)
Circulating MicroRNA , MicroRNAs , Male , Humans , Testis/metabolism , Circulating MicroRNA/metabolism , MicroRNAs/genetics , Biomarkers/metabolism , Leydig Cells/metabolism
2.
Drug Metab Dispos ; 43(6): 829-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25788542

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, representing a spectrum of liver pathologies that include simple hepatic steatosis and the more advanced nonalcoholic steatohepatitis (NASH). The current study was conducted to determine whether pediatric NASH also results in altered disposition of acetaminophen (APAP) and its two primary metabolites, APAP-sulfate and APAP-glucuronide. Pediatric patients with hepatic steatosis (n = 9) or NASH (n = 3) and healthy patients (n = 12) were recruited in a small pilot study design. All patients received a single 1000-mg dose of APAP. Blood and urine samples were collected at 1, 2, and 4 hours postdose, and APAP and APAP metabolites were determined by high-performance liquid chromatography. Moreover, human liver tissues from patients diagnosed with various stages of NAFLD were acquired from the Liver Tissue Cell Distribution System to investigate the regulation of the membrane transporters, multidrug resistance-associated protein 2 and 3 (MRP2 and MRP3, respectively). Patients with the more severe disease (i.e., NASH) had increased serum and urinary levels of APAP-glucuronide along with decreased serum levels of APAP-sulfate. Moreover, an induction of hepatic MRP3 and altered canalicular localization of the biliary efflux transporter, MRP2, describes the likely mechanism for the observed increase in plasma retention of APAP-glucuronide, whereas altered regulation of sulfur activation genes may explain decreased sulfonation activity in NASH. APAP-glucuronide and APAP-sulfate disposition is altered in NASH and is likely due to hepatic membrane transporter dysregulation as well as altered intracellular sulfur activation.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Liver/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Up-Regulation , Acetaminophen/analogs & derivatives , Acetaminophen/blood , Acetaminophen/urine , Adolescent , Analgesics, Non-Narcotic/blood , Analgesics, Non-Narcotic/urine , Bile Canaliculi/metabolism , Bile Canaliculi/pathology , Biotransformation , Child , Cohort Studies , Fatty Liver/blood , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/urine , Female , Humans , Liver/pathology , Male , Multidrug Resistance-Associated Protein 2 , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/urine , Pilot Projects , Protein Transport
3.
Regul Toxicol Pharmacol ; 73(2): 562-70, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26382609

ABSTRACT

Bococizumab is a humanized monoclonal IgG2Δa antibody against proprotein convertase subtilisin/kexin type 9 (PCSK9) for the treatment of hyperlipidemia. The evaluation of potential effects on embryo-fetal development was conducted in the rat. In a pharmacokinetic/pharmacodynamic study bococizumab was administered intravenously to pregnant Sprague-Dawley (SD) rats (n = 8/group) at 0, 10, 30, and 100 mg/kg during organogenesis. Maternal and fetal bococizumab, total cholesterol and HDL concentrations were determined. Bococizumab was well tolerated and there were no effects on ovarian or uterine parameters. Maternal and fetal bococizumab exposure increased with increasing dose, with a corresponding dose-dependent decrease in fetal cholesterol levels. Maternal cholesterol levels were decreased significantly, with reductions that were of a similar magnitude regardless of dose. In the definitive embryo-fetal development study bococizumab was administered to pregnant SD rats (n = 20/group) at 0, 10, 30, and 100 mg/kg and no adverse maternal or developmental effects were observed up to 100 mg/kg. These studies have provided an appropriate and relevant safety assessment of bococizumab in pregnant rats to inform human risk assessment, demonstrating no adverse effects on embryo-fetal development at magnitudes greater than anticipated clinical exposure and in the presence of maximal reductions in maternal cholesterol and dose-dependent reductions in fetal cholesterol.


Subject(s)
Antibodies, Anti-Idiotypic/blood , Antibodies, Monoclonal, Humanized/administration & dosage , Cholesterol/blood , Fetal Development/physiology , Maternal-Fetal Exchange/physiology , Serine Endopeptidases/blood , Animals , Antibodies, Monoclonal, Humanized/toxicity , Dose-Response Relationship, Drug , Female , Fetal Development/drug effects , Maternal-Fetal Exchange/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/chemically induced , Proprotein Convertase 9 , Rats , Rats, Sprague-Dawley
4.
Toxicol Appl Pharmacol ; 274(1): 156-67, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24126418

ABSTRACT

Pretreatment of mice with a low hepatotoxic dose of acetaminophen (APAP) results in resistance to a subsequent, higher dose of APAP. This mouse model, termed APAP autoprotection was used here to identify differentially expressed genes and cellular pathways that could contribute to this development of resistance to hepatotoxicity. Male C57BL/6J mice were pretreated with APAP (400mg/kg) and then challenged 48h later with 600mg APAP/kg. Livers were obtained 4 or 24h later and total hepatic RNA was isolated and hybridized to Affymetrix Mouse Genome MU430_2 GeneChip. Statistically significant genes were determined and gene expression changes were also interrogated using the Causal Reasoning Engine (CRE). Extensive literature review narrowed our focus to methionine adenosyl transferase-1 alpha (MAT1A), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), flavin-containing monooxygenase 3 (Fmo3) and galectin-3 (Lgals3). Down-regulation of MAT1A could lead to decreases in S-adenosylmethionine (SAMe), which is known to protect against APAP toxicity. Nrf2 activation is expected to play a role in protective adaptation. Up-regulation of Lgals3, one of the genes supporting the Nrf2 hypothesis, can lead to suppression of apoptosis and reduced mitochondrial dysfunction. Fmo3 induction suggests the involvement of an enzyme not known to metabolize APAP in the development of tolerance to APAP toxicity. Subsequent quantitative RT-PCR and immunochemical analysis confirmed the differential expression of some of these genes in the APAP autoprotection model. In conclusion, our genomics strategy identified cellular pathways that might further explain the molecular basis for APAP autoprotection.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Gene Expression Regulation/physiology , Models, Animal , Animals , Chemical and Drug Induced Liver Injury/genetics , Gene Expression Regulation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL
5.
Birth Defects Res B Dev Reprod Toxicol ; 101(4): 325-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25044418

ABSTRACT

Treatment-induced epididymal inflammation and granuloma formation is only an occasional problem in preclinical drug development, but it can effectively terminate the development of that candidate molecule. Screening for backup molecules without that toxicity must be performed in animals (generally rats) that requires at least 2 to 3 weeks of in vivo exposure, a great deal of specially synthesized candidate compound, and histologic examination of the target tissues. We instead hypothesized that these treatments induced proinflammatory gene expression, and so used mixed-cell cultures from the rat epididymal tubule to monitor the induction of proinflammatory cytokines. Cells were exposed for 24 hr and then cytotoxicity was evaluated with the MTS assay and mRNA levels of Interleukin-6 (IL-6) and growth-related oncogene (GRO) were measured. We found that compounds that were more toxic in vivo stimulated a greater induction of IL-6 and GRO mRNA levels in vitro. By relating effective concentrations in vitro with the predicted C(eff), we could rank compounds by their propensity to induce inflammation in rats in vivo. This method allowed the identification of several compounds with very low inflammatory induction in vitro. When tested in rats, the compounds produced small degrees of inflammation at an acceptable margin (approximately 20×), and have progressed into further development.


Subject(s)
Epididymis/drug effects , Epididymis/pathology , Epididymitis/chemically induced , Epididymitis/pathology , Animals , Cells, Cultured , Chemokine CXCL1/genetics , Epididymis/immunology , Epididymitis/immunology , Granuloma/chemically induced , Granuloma/pathology , Interleukin-6/genetics , Male , Mitochondria/metabolism , Primary Cell Culture , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
6.
Birth Defects Res ; 116(5): e2345, 2024 May.
Article in English | MEDLINE | ID: mdl-38716582

ABSTRACT

BACKGROUND: Abrocitinib is a Janus kinase (JAK) 1 selective inhibitor approved for the treatment of atopic dermatitis. Female reproductive tissues were unaffected in general toxicity studies, but an initial female rat fertility study resulted in adverse effects at all doses evaluated. A second rat fertility study was conducted to evaluate lower doses and potential for recovery. METHODS: This second study had 4 groups of 20 females each administered abrocitinib (0, 3, 10, or 70 mg/kg/day) 2 weeks prior to cohabitation through gestation day (GD) 7. In addition, 2 groups of 20 rats (0 or 70 mg/kg/day) were dosed for 3 weeks followed by a 4-week recovery period before mating. All mated females were evaluated on GD 14. RESULTS: No effects were observed at ≤10 mg/kg/day. At 70 mg/kg/day (29x human exposure), decreased pregnancy rate, implantation sites, and viable embryos were observed. All these effects reversed 4 weeks after the last dose. CONCLUSIONS: Based on these data and literature on the potential role of JAK signaling in implantation, we hypothesize that these effects may be related to JAK1 inhibition and, generally, that peri-implantation effects such as these, in the absence of cycling or microscopic changes in nonpregnant female reproductive tissues, are anticipated to be reversible.


Subject(s)
Fertility , Janus Kinase 1 , Pyrimidines , Sulfonamides , Female , Animals , Pregnancy , Rats , Fertility/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Rats, Sprague-Dawley , Embryo Implantation/drug effects , Janus Kinase Inhibitors/pharmacology , Pregnancy Rate
7.
Birth Defects Res ; 115(3): 348-356, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36367445

ABSTRACT

Achondroplasia is an autosomal disorder caused by point mutation in the gene encoding fibroblast growth factor receptor 3 (FGFR3) and resulting in gain of function. Recifercept is a potential disease modifying treatment for achondroplasia and functions as a decoy protein that competes for ligands of the mutated FGFR3. Recifercept is intended to restore normal bone growth by preventing the mutated FGFR3 from negative inhibitory signaling in pediatric patients with achondroplasia. Here we evaluated the potential effects of twice weekly administration of recifercept to juvenile cynomolgus monkeys (approximately 3-months of age at the initiation of dosing) for 6-months. No adverse effects were noted in this study, identifying the high dose as the no-observed-adverse-effect-level and supporting the use of recifercept in pediatric patients from birth. Considering that juvenile toxicity studies in nonhuman primates are not frequently conducted, and when they are conducted they typically utilize animals ≥9 months of age, this study demonstrates the feasibility of executing a juvenile toxicity study in very young monkeys prior to weaning.


Subject(s)
Achondroplasia , Receptor, Fibroblast Growth Factor, Type 3 , Animals , Humans , Child , Infant , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 3/pharmacology , Macaca fascicularis/metabolism , Achondroplasia/drug therapy , Achondroplasia/genetics , Achondroplasia/metabolism , Bone Development , Bone and Bones/metabolism
8.
J Med Chem ; 66(5): 3195-3211, 2023 03 09.
Article in English | MEDLINE | ID: mdl-36802610

ABSTRACT

The melanocortin-4 receptor (MC4R) is a centrally expressed, class A GPCR that plays a key role in the regulation of appetite and food intake. Deficiencies in MC4R signaling result in hyperphagia and increased body mass in humans. Antagonism of MC4R signaling has the potential to mitigate decreased appetite and body weight loss in the setting of anorexia or cachexia due to underlying disease. Herein, we report on the identification of a series of orally bioavailable, small-molecule MC4R antagonists using a focused hit identification effort and the optimization of these antagonists to provide clinical candidate 23. Introduction of a spirocyclic conformational constraint allowed for simultaneous optimization of MC4R potency and ADME attributes while avoiding the production of hERG active metabolites observed in early series leads. Compound 23 is a potent and selective MC4R antagonist with robust efficacy in an aged rat model of cachexia and has progressed into clinical trials.


Subject(s)
Appetite , Receptor, Melanocortin, Type 4 , Rats , Humans , Animals , Cachexia/drug therapy , Anorexia/drug therapy , Molecular Conformation
9.
Birth Defects Res B Dev Reprod Toxicol ; 95(3): 225-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22447726

ABSTRACT

Lersivirine is a second-generation nonnucleoside reverse transcriptase inhibitor undergoing clinical development for the treatment of HIV-1. An embryo-fetal developmental toxicity study was performed to evaluate the maternal and developmental toxicity of lersivirine in pregnant mice. Mated Crl:CD1(ICR) mice were administered 0, 150, 350, and 500 mg/kg lersivirine once daily by oral gavage on gestation days 6 to 17, followed by cesarean section on gestation day 18. The first 2 days of dosing for the high-dose group were done at 250 mg/kg to allow induction of hepatic metabolizing enzymes, after which the dose was increased to 500 mg/kg/day. This dosing paradigm allowed for maintenance of exposure in the high-dose group despite the considerable autoinduction that occurs in rodents following lersivirine treatment. Lersivirine did not cause an increase in external, visceral, or skeletal malformations. Intrauterine growth retardation, demonstrated by reduced fetal body weights and increased variations associated with delayed skeletal ossification, was noted at 350 and 500 mg/kg/day. The results of these studies indicate that lersivirine is not teratogenic in mice.


Subject(s)
Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Nitriles/toxicity , Pyrazoles/toxicity , Toxicity Tests , Animals , Body Weight/drug effects , Bone and Bones/abnormalities , Bone and Bones/drug effects , Bone and Bones/embryology , Bone and Bones/physiopathology , Cesarean Section , Embryo, Mammalian/embryology , Feeding Behavior/drug effects , Female , Fetus/abnormalities , Fetus/drug effects , Fetus/pathology , Maternal Exposure , Mice , Osteogenesis/drug effects , Pregnancy
10.
Birth Defects Res B Dev Reprod Toxicol ; 95(3): 238-49, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495808

ABSTRACT

The critical periods of axial skeletal development in rats and mice have been well characterized, however the timing of skeletal development in rabbits is not as well known. It is important to have a more precise understanding of this timing of axial skeletal development in rabbits due to the common use of this species in standard nonclinical studies to assess embryo-fetal developmental toxicity. Hydroxyurea, a teratogen known to induce a variety of fetal skeletal malformations, was administered to New Zealand White rabbits as a single dose (500 mg/kg) on individual days during gestation (gestation day, GD 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 19) and fetal external, visceral, and skeletal morphology was examined following cesarean sections on GD 29. A wide range of fetal skeletal effects was observed following hydroxyurea treatment, with a progression of malformations from anterior to posterior structures over time, as well as from proximal to distal structures over time. The sensitive window of axial skeletal development was determined to be GD 8 to 13, while disruption of appendicular and cranio-facial skeletal development occurred primarily from GD 11 to 16 and GD 11 to 12, respectively. The results of this study provide a better understanding of the critical developmental window for different segments of the rabbit skeleton, which will aid in the design of window studies to investigate teratogenicity in rabbits.


Subject(s)
Bone Development/drug effects , Hydroxyurea/toxicity , Maternal Exposure , Animals , Bone and Bones/abnormalities , Bone and Bones/drug effects , Bone and Bones/embryology , Cesarean Section , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/pathology , Female , Fetus/abnormalities , Fetus/drug effects , Mice , Phenotype , Pregnancy , Rabbits , Rats , Time Factors , Viscera/abnormalities , Viscera/drug effects , Viscera/embryology
11.
Birth Defects Res B Dev Reprod Toxicol ; 95(3): 250-61, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495820

ABSTRACT

BACKGROUND: Lersivirine is a second-generation nonnucleoside reverse transcriptase inhibitor undergoing clinical development for the treatment of human immunodeficiency virus-1. An embryo-fetal development study was performed to evaluate the potential for maternal and developmental toxicity of lersivirine. METHODS: Pregnant New Zealand White rabbits were administered 0, 100, 250, and 500 mg/kg lersivirine by oral gavage once daily on gestation days (GDs) 7 to 19, followed by cesarean section on GD 29 and fetal evaluation. RESULTS: Maternal toxicity was noted at all dose levels (decreased food consumption and body weight gain), with fetal toxicity at 500 mg/kg (decreased fetal weights, increased postimplantation loss). Equivocal findings for axial skeletal malformations were observed in three fetuses at 500 mg/kg. To better understand if these malformations were related to treatment with lersivirine, a follow-up rabbit embryo-fetal development study was performed with 1000 mg/kg/day lersivirine (500 mg/kg BID, 12-hr interdose interval) for two different 3-day windows, GDs 8 to 10 or GDs 11 to 13, which represent the sensitive windows of axial skeletal development in rabbits. Control rabbits were administered vehicle following the same dosing regimen from GDs 8 to 13. Cesarean sections were performed on GD 29, and fetal skeletons were examined for the potential of lersivirine to cause skeletal malformations in rabbits. At maternal exposure levels higher than the initial study, lersivirine did not induce fetal skeletal malformations when administered in the sensitive windows of axial skeletal development. CONCLUSION: The results of these studies indicate that lersivirine did not exhibit any evidence of teratogenicity in rabbits.


Subject(s)
Bone Development/drug effects , Embryonic Development/drug effects , Nitriles/administration & dosage , Nitriles/toxicity , Organogenesis/drug effects , Pyrazoles/administration & dosage , Pyrazoles/toxicity , Toxicity Tests , Animals , Body Weight/drug effects , Bone and Bones/abnormalities , Bone and Bones/drug effects , Bone and Bones/embryology , Bone and Bones/pathology , Cesarean Section , Dose-Response Relationship, Drug , Feeding Behavior/drug effects , Female , Fetus/abnormalities , Fetus/drug effects , Fetus/pathology , Humans , Maternal Exposure , Nitriles/blood , Nitriles/pharmacokinetics , Pregnancy , Pyrazoles/blood , Pyrazoles/pharmacokinetics , Rabbits , Survival Analysis , Viscera/abnormalities , Viscera/drug effects , Viscera/embryology
12.
Drug Chem Toxicol ; 35(1): 20-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21774737

ABSTRACT

Animal and care use practices are constantly evolving. These can have unexpected consequences on the data collected from such procedures. One example is the recent change in our animal facility, based on recommendations from the Newcastle Consensus Meeting on Carbon Dioxide Euthanasia of Laboratory Animals, from CO(2) to isoflurane for anesthesia. The current study was conducted to determine the effects of isoflurane on sperm motility, as compared to two different CO(2) euthanasia procedures. Sperm motility was evaluated after euthanasia by a standard 5-minute CO(2) euthanasia procedure, an extended 10-minute CO(2) euthanasia procedure, or by isoflurane anesthesia followed by exsanguination (iso/exsanguination). The 5-minute CO(2) procedure produced sperm motility of 94.3 ± 1.7% motile sperm with 65.6 ± 16.8 sperm/field. By comparison, iso/exsanguination reduced that count to 3.3 ± 2.3 sperm/field and only 60.7 ± 32.0% motile sperm. The reduction in sperm motility after iso/exsanguination appeared to have been due primarily to the reduction in the number of sperm expelled from the vas deferens (3.3), compared to that after 5-minute CO(2) (65.6). This reduction in number of sperm available for evaluation, in the presence of a constant level of background debris, which was counted by the computer optics system as nonmotile sperm, resulted in an apparent reduction in motility. Using the extended 10-minute CO(2) procedure produced sperm data in between the other two extremes: 77.6 ± 36.1% motile sperm with 34.6 ± 28.3 sperm/field. The results of this study support the hypothesis that isoflurane inhibits contraction of the smooth muscle of the vas deferens, resulting in a decreased number of expelled sperm. Given these findings, it is important that careful consideration be taken to select an appropriate anesthesia/euthanasia method.


Subject(s)
Anesthetics, Inhalation/toxicity , Isoflurane/toxicity , Sperm Motility/drug effects , Vas Deferens/drug effects , Animal Welfare , Animals , Carbon Dioxide/toxicity , Euthanasia, Animal/methods , Image Processing, Computer-Assisted , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Vas Deferens/metabolism
13.
Toxicol Sci ; 189(2): 225-236, 2022 09 24.
Article in English | MEDLINE | ID: mdl-35866640

ABSTRACT

Ervogastat (PF-06865571) is a small molecule diacylglycerol acyltransferase 2 (DGAT2) inhibitor being developed for the oral treatment of nonalcoholic steatohepatitis (NASH) with liver fibrosis. DGAT2 is a key enzyme in triglyceride synthesis in tissues and in regulating energy metabolism. Fertility and developmental toxicity studies with ervogastat were conducted in female rats and rabbits. There were no effects on female rat fertility or rabbit embryo-fetal development. Administration of ervogastat to pregnant rats during organogenesis reduced fetal weight and caused higher incidences of bent bones in fetuses that were shown to resolve by postnatal day 28 and were therefore considered to be transient variations secondary to developmental delay. Extended dosing in rats through the end of gestation and lactation (pre- and post-natal development study) caused impaired skin development, reduced offspring viability, and growth retardation. The spectrum of developmental effects in rats is consistent with the intended pharmacology (altered triglyceride metabolism) and the transient nature of the skeletal findings, along with the late gestational window of sensitivity for the effects on skin barrier development, reduce the concern for potential adverse developmental effects following unintended early gestational exposure to ervogastat in humans where treatment can be discontinued once pregnancy is determined.


Subject(s)
Diacylglycerol O-Acyltransferase , Reproduction , Animals , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/pharmacology , Female , Fertility , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Triglycerides
14.
Dis Model Mech ; 15(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35466995

ABSTRACT

Increased research to improve preclinical models to inform the development of therapeutics for neonatal diseases is an area of great need. This article reviews five common neonatal diseases - bronchopulmonary dysplasia, retinopathy of prematurity, necrotizing enterocolitis, perinatal hypoxic-ischemic encephalopathy and neonatal sepsis - and the available in vivo, in vitro and in silico preclinical models for studying these diseases. Better understanding of the strengths and weaknesses of specialized neonatal disease models will help to improve their utility, may add to the understanding of the mode of action and efficacy of a therapeutic, and/or may improve the understanding of the disease pathology to aid in identification of new therapeutic targets. Although the diseases covered in this article are diverse and require specific approaches, several high-level, overarching key lessons can be learned by evaluating the strengths, weaknesses and gaps in the available models. This Review is intended to help guide current and future researchers toward successful development of therapeutics in these areas of high unmet medical need.


Subject(s)
Bronchopulmonary Dysplasia , Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Bronchopulmonary Dysplasia/drug therapy , Drug Development , Enterocolitis, Necrotizing/drug therapy , Humans , Infant, Newborn , Infant, Newborn, Diseases/drug therapy
15.
Birth Defects Res B Dev Reprod Toxicol ; 92(4): 323-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21594972

ABSTRACT

BACKGROUND: CP-601927 is a selective α(4) ß(2) nicotinic acetylcholine receptor (nAChR) partial agonist. The objective of this study was to assess the potential effects persisting into adulthood when CP-601,927 was administered to neonatal/juvenile rats. Since the juvenile toxicity study was being performed early in the development program and this study would represent the longest dosing period yet evaluated, the study design incorporated standard endpoints typically evaluated in a general toxicity screening study. METHODS: CP-601,927 was administered to Sprague-Dawley rats from postnatal day (PND) 7-70 by oral gavage at doses of 0.3, 1, or 3 mg/kg. During treatment animals were evaluated for growth, development, and sexual maturation. At the end of the treatment period general toxicity screening endpoints were collected (e.g., organ weights, histology, clinical chemistry). Following a 2-week latency period, animals were evaluated for CNS function in a comprehensive behavioral training battery consisting of a functional observational battery, motor activity, acoustic startle response, and learning and memory evaluations. Reproductive competency was evaluated by mating treated rats and allowing pregnant dams to deliver and rear their litters until PND 10. RESULTS AND CONCLUSIONS: Treatment-related findings included the death of 2 males receiving 3 mg/kg CP-601,927, and transient reductions in body weight for both males and females during the third week of dosing which quickly recovered to control levels. The only treatment-related alteration in behavior was decreased motor activity, which occurred only in females at the highest dose tested. CP-601,927 had no effect on acoustic startle response, learning and memory, sexual maturation, reproductive capacity, or general toxicity endpoints.


Subject(s)
Abnormalities, Drug-Induced/etiology , Behavior, Animal/drug effects , Brain/drug effects , Nicotinic Agonists/toxicity , Receptors, Nicotinic/chemistry , Reproduction/drug effects , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Drug Partial Agonism , Female , Learning/drug effects , Male , Memory/drug effects , Organ Size/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism
16.
Reprod Toxicol ; 99: 138-143, 2021 01.
Article in English | MEDLINE | ID: mdl-33065206

ABSTRACT

Traditionally, understanding potential developmental toxicity from pharmaceutical exposures has been based on the results of ICH guideline studies in two species. However, support is growing for the use of weight of evidence approaches when communicating the risk of developmental toxicity, where the intended pharmacologic mode of action affects fundamental pathways in developmental biology or phenotypic data from genetically modified animals may increasingly be included in the overall assessment. Since some concern surrounds the use of data from knockout (KO) mice to accurately predict the risk for pharmaceutical modulation of a target, a deeper understanding of the relevance and predictivity of adverse developmental effects in KO mice for pharmacological target modulation is needed. To this end, we compared the results of embryo-fetal development (EFD) studies for 86 drugs approved by the FDA from 2017 to 2019 that also had KO mouse data available in the public domain. These comparisons demonstrate that data from KO mouse models are overall highly predictive of malformations or embryo-fetal lethality (MEFL) from EFD studies, but less so of a negative outcome in EFD studies. This information supports the use of embryo-fetal toxicity data in KO models as part of weight of evidence approaches in the communication of developmental toxicity risk of pharmaceutical compounds.


Subject(s)
Abnormalities, Drug-Induced , Drug Evaluation, Preclinical/methods , Models, Animal , Teratogens/toxicity , Animals , Embryo, Mammalian/drug effects , Fetal Death/etiology , Mice, Knockout
17.
Reprod Toxicol ; 103: 28-35, 2021 08.
Article in English | MEDLINE | ID: mdl-34058573

ABSTRACT

BNT162b2 is a vaccine developed to prevent coronavirus disease 2019 (COVID-19). BNT162b2 is a lipid nanoparticle formulated nucleoside-modified messenger RNA (mRNA) encoding the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein locked in its prefusion conformation. A developmental and reproductive toxicity study was conducted in rats according to international regulatory guidelines. The full human BNT162b2 dose of 30 µg mRNA/dose (>300 times the human dose on a mg/kg basis) was administered intramuscularly to 44 female rats 21 and 14 days prior to mating and on gestation days 9 and 20. Half of the rats were subject to cesarean section and full fetal examination at the end of gestation, and the other half were allowed to deliver and were monitored to the end of lactation. A robust neutralizing antibody response was confirmed prior to mating and at the end of gestation and lactation. The presence of neutralizing antibodies was also confirmed in fetuses and offspring. Nonadverse effects, related to the local injection site reaction, were noted in dams as expected from other animal studies and consistent with observations in humans. There were no effects of BNT162b2 on female mating performance, fertility, or any ovarian or uterine parameters nor on embryo-fetal or postnatal survival, growth, physical development or neurofunctional development in the offspring through the end of lactation. Together with the safety profile in nonpregnant people, this ICH-compliant nonclinical safety data supports study of BNT162b2 in women of childbearing potential and pregnant and lactating women.


Subject(s)
COVID-19 Vaccines/toxicity , Fertility , Fetal Development , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , BNT162 Vaccine , COVID-19 Vaccines/pharmacology , Cesarean Section , Female , Lactation , Pregnancy , Rats , Rats, Wistar
18.
Toxicol Sci ; 179(2): 183-194, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33247737

ABSTRACT

Acetyl-CoA carboxylase (ACC) is an enzyme within the de novo lipogenesis (DNL) pathway and plays a role in regulating lipid metabolism. Pharmacologic ACC inhibition has been an area of interest for multiple potential indications including oncology, acne vulgaris, metabolic diseases such as type 2 diabetes mellitus, and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. A critical role for ACC in de novo synthesis of long-chain fatty acids during fetal development has been demonstrated in studies in mice lacking Acc1, where the absence of Acc1 results in early embryonic lethality. Following positive predictions of developmental toxicity in the alternative in vitro assays (positive in murine embryonic stem cell [mESC] assay and rat whole embryo culture, but negative in zebrafish), developmental toxicity (growth retardation and dysmorphogenesis associated with disrupted midline fusion) was observed with the oral administration of the dual ACC1 and 2 inhibitors, PF-05175157, in Sprague Dawley rats and New Zealand White rabbits. The results of these studies are presented here to make comparisons across the assays, as well as mechanistic insights from the mESC assay demonstrating high ACC expression in the mESC and that ACC-induced developmental toxicity can be rescued with palmitic acid providing supportive evidence for DNL pathway inhibition as the underlying mechanism. Ultimately, while the battery of alternative approaches and weight-of-evidence case were useful for hazard identification, the embryo-fetal development studies were necessary to inform the risk assessment on the adverse fetal response, as malformations and/or embryo-fetal lethality were limited to doses that caused near-complete inhibition of DNL.


Subject(s)
Acetyl-CoA Carboxylase , Diabetes Mellitus, Type 2 , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Lipogenesis , Mice , Rabbits , Rats , Rats, Sprague-Dawley , Zebrafish/metabolism
19.
Reprod Toxicol ; 96: 11-16, 2020 09.
Article in English | MEDLINE | ID: mdl-32522587

ABSTRACT

Traditionally, understanding potential developmental toxicity from pharmaceutical exposures has been based on the results of ICH guideline studies in two species. However, support is growing for the use of weight of evidence approaches when communicating the risk of developmental toxicity, where the intended pharmacologic mode of action affects fundamental pathways in developmental biology or phenotypic data from genetically modified animals may increasingly be included in the overall assessment. Since some concern surrounds the use of data from knockout (KO) mice to accurately predict the risk for pharmaceutical modulation of a target, a deeper understanding of the relevance and predictivity of adverse developmental effects in KO mice for pharmacological target modulation is needed. To this end, we compared the results of embryo-fetal development (EFD) studies for 86 drugs approved by the FDA from 2017 to 2019 that also had KO mouse data available in the public domain. These comparisons demonstrate that data from KO mouse models are overall highly predictive of malformations or embryo-fetal lethality (MEFL) from EFD studies, but less so of a negative outcome in EFD studies. This information supports the use of embryo-fetal toxicity data in KO models as part of weight of evidence approaches in the communication of developmental toxicity risk of pharmaceutical compounds.


Subject(s)
Abnormalities, Drug-Induced , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions , Embryo Loss , Fetal Death , Models, Animal , Animals , Embryonic Development/drug effects , Fetal Development/drug effects , Mice, Knockout
20.
Toxicol Appl Pharmacol ; 236(1): 49-58, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19371622

ABSTRACT

During APAP toxicity, activation of Kupffer cells is critical for protection from hepatotoxicity and up-regulation of multidrug resistance-associated protein 4 (Mrp4) in centrilobular hepatocytes. The present study was performed to determine the expression profile of uptake and efflux transporters in mouse liver following treatment with allyl alcohol (AlOH), a periportal hepatotoxicant. This study also investigated the role of Kupffer cells in AlOH hepatotoxicity, and whether changes in transport protein expression by AlOH are dependent on the presence of Kupffer cells. C57BL/6J mice received 0.1 ml clodronate liposomes to deplete Kupffer cells or empty liposomes 48 h prior to dosing with 60 mg/kg AlOH, i.p. Hepatotoxicity was assessed by plasma ALT and histopathology. Hepatic transporter mRNA and protein expression were determined by branched DNA signal amplification assay and Western blotting, respectively. Depletion of Kupffer cells by liposomal clodronate treatment resulted in heightened susceptibility to AlOH toxicity. Exposure to AlOH increased mRNA levels of several Mrp genes, while decreasing organic anion transporting polypeptides (Oatps) mRNA expression. Protein analysis mirrored many of these mRNA changes. The presence of Kupffer cells was not required for the observed changes in uptake and efflux transporters induced by AlOH. Immunofluorescent analysis revealed enhanced Mrp4 staining exclusively in centrilobular hepatocytes of AlOH treated mice. These findings demonstrate that Kupffer cells are protective from AlOH toxicity and that induction of Mrp4 occurs in liver regions away from areas of AlOH damage independent of Kupffer cell function. These results suggest that Kupffer cell mediators do not play a role in mediating centrilobular Mrp4 induction in response to periportal damage by AlOH.


Subject(s)
Kupffer Cells/drug effects , Liver/drug effects , Multidrug Resistance-Associated Proteins/drug effects , Organic Anion Transporters/drug effects , Propanols/toxicity , Alanine Transaminase/blood , Animals , Blotting, Western , Clodronic Acid/administration & dosage , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Injections, Intraperitoneal , Injections, Intravenous , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liposomes , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Propanols/administration & dosage , RNA, Messenger/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL