Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
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
2.
Toxicol Appl Pharmacol ; 473: 116582, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37295732

ABSTRACT

A high incidence of thymic lymphoma has been noted in mice deficient of retinoid-related orphan receptor γ2 (RORγ2), which is required for differentiation of naïve CD4+ T cells into TH17 cells. Using a RORγ homozygous knockout (KO) mouse model of thymic lymphoma, we characterized this tumor progression and investigated the utility of 5-hydroxymethylcytosine (5hmC) signatures as a non-invasive circulating biomarker for early prediction of malignancy. No evidence for malignancy was noted in the wild-type mice, while primary thymic lymphoma with multi-organ metastasis was observed microscopically in 97% of the homozygous RORγ KO mice. The severity of thymic lymphoma was not age-dependent in the KO mice of 2 to 4 months old. Differential enrichment of 5hmC in thymic DNA and plasma cell-free DNA (cfDNA) was compared across different stages of tumor progression. Random forest modeling of plasma cfDNA achieved good predictivity (AUC = 0.74) in distinguishing early non-metastatic thymic lymphoma compared to cancer-free controls, while perfect predictivity was achieved with advanced multi-organ metastatic disease (AUC = 1.00). Lymphoid-specific genes involved in thymocyte selection during T cell development (Themis, Tox) were differentially enriched in both plasma and thymic tissue. This could help in differentiating thymic lymphoma from other tumors commonly detected in rodent carcinogenicity studies used in pharmaceutical drug development to inform human malignancy risk. Overall, these results provide a proof-of-concept for using circulating cfDNA profiles in rodent carcinogenicity studies for early risk assessment of novel pharmaceutical targets.


Subject(s)
Cell-Free Nucleic Acids , Neoplasms , Animals , Humans , Infant , Mice , Cell-Free Nucleic Acids/genetics , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3
3.
Intest Res ; 21(3): 295-305, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37248173

ABSTRACT

Inflammatory bowel disease (IBD), comprising Crohn's disease and ulcerative colitis, is a lifelong disease that manifests with chronic intestinal inflammation, sequential fibrosis, and an increased risk of colitis-associated colon cancer (CAC). The combined effects of genetic, immunological, environmental, and microbial factors render it difficult to determine the specific mechanism underlying the induction and perpetuation of IBD. Various animal models of IBD have contributed enormously to the understanding of IBD pathogenesis in terms of genomics, transcriptomics, proteomics, microbiome, and drug development of novel therapeutics. Although comprehensive research on IBD has been enabled by advanced technologies, such as genetically engineered models, there is a great need to develop relevant in vivo models of colitis and fibrosis. Here, we review 4 categories of animal models of acute and chronic intestinal inflammation, fibrosis, and CAC: chemically induced, genetically engineered, T cell transfer, and spontaneous gene mutation models.

4.
Toxicol Appl Pharmacol ; 466: 116471, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36934859

ABSTRACT

Thymoma, a tumor of thymic lymphocytes or thymic epithelial cells (TECs), is a common spontaneous tumor in Wistar Han rats, especially in females with up to 18% incidence in controls. In addition to sex, there are rat strain differences in background incidence of thymomas such as Sprague Dawley versus Wistar Han rats. Human thymomas are very rare and without clear differences in incidence between males and females. Immunomodulatory and anti-inflammatory pharmaceutical drug classes, including Janus kinase inhibitors, increase the incidence of benign thymoma in two-year rat carcinogenicity studies. Potential non-genotoxic mechanisms that might contribute to the pathogenesis of thymoma development in one sex (female) Wistar Han rats include: (1) hormonal differences, (2) high proliferation rate of TECs, (3) delayed physiologic thymic involution, and/or (4) significant level of immunosuppression at high doses of a pharmaceutical drug. Factors to consider in the human cancer risk assessment of pharmaceutical-induced thymoma are: the genotoxicity of the test article, sex and strain of rats, exposure safety margins, and pathophysiologic differences and similarities of thymoma between rats and humans. Totality of weight of evidence approach and available data suggest thymomas observed in carcinogenicity studies of pharmaceutical drugs are not relevant for human risk at clinically relevant therapeutic doses.


Subject(s)
Anti-Inflammatory Agents , Immunomodulating Agents , Janus Kinase Inhibitors , Thymoma , Thymus Neoplasms , Animals , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Risk Assessment , Thymoma/chemically induced , Thymoma/pathology , Thymus Neoplasms/chemically induced , Thymus Neoplasms/pathology , Anti-Inflammatory Agents/adverse effects , Janus Kinase Inhibitors/adverse effects , Immunomodulating Agents/adverse effects
5.
Toxicol Pathol ; 48(6): 712-717, 2020 08.
Article in English | MEDLINE | ID: mdl-32815472

ABSTRACT

Immune tolerance is defined by an active state of immune system unresponsiveness to foreign and self-antigens. Loss of immune tolerance to self-antigens and the resulting overexpression of autoantibodies can lead to tissue injury and development of various autoimmune diseases. In drug development, the goal of newly emerging immune tolerance therapies is to treat autoimmune disorders by restoring the immunoregulatory capacity of the immune system. Development of immune tolerance targets is initiated with the establishment of pharmacological efficacy in relevant disease animal models, followed by their stepwise translation to humans. This review discusses the major challenges to developing tolerance inducing pharmaceutical drugs, including the selection of appropriate disease models to establish efficacy, adequate, and acceptable in vitro and in vivo safety assessments, relevant biomarkers of human safety and efficacy, and finally, some regulatory guidelines to successfully develop immune tolerance therapeutics. [Box: see text].


Subject(s)
Autoimmune Diseases , Immune Tolerance , Animals , Autoantibodies , Autoimmune Diseases/drug therapy , Humans
6.
Toxicol Pathol ; 48(6): 721-724, 2020 08.
Article in English | MEDLINE | ID: mdl-32602415

ABSTRACT

Influx and efflux kidney tubular transporters are major determinants of the disposition of xenobiotics, including pharmaceutical drugs. On the basolateral membrane of proximal tubular cells, there are influx transporters, such as organic cation transporters. On the apical membrane of proximal tubular cells, there are efflux transporters, such as multidrug and toxin extrusion proteins. The secretion process across the apical membrane into the lumen occurs via efflux transporters which plays an important role in serum creatinine (sCr) elimination in urine. The interference of a pharmaceutical drug with transporters can lead to changes in sCr with no alterations in biomarkers or light microscopic evidence indicative of renal injury. Identification of transporters that influence drug disposition, toxicity, and overall nonclinical safety assessment is important in drug discovery and development programs. This mini review describes some key aspects of kidney tubular transporters and drug-induced renal toxicities in safety risk assessment and drug development.


Subject(s)
Kidney , Xenobiotics , Biological Transport , Creatinine , Drug Development , Kidney/metabolism , Xenobiotics/toxicity
7.
J Toxicol Sci ; 44(6): 373-391, 2019.
Article in English | MEDLINE | ID: mdl-31168026

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely used therapeutic class in clinical medicine. These are sub-divided based on their selectivity for inhibition of cyclooxygenase (COX) isoforms (COX-1 and COX-2) into: (1) non-selective (ns-NSAIDs), and (2) selective NSAIDs (s-NSAIDs) with preferential inhibition of COX-2 isozyme. The safety and pathophysiology of NSAIDs on the renal and cardiovascular systems have continued to evolve over the years following short- and long-term treatment in both preclinical models and humans. This review summarizes major learnings on cardiac and renal complications associated with pharmaceutical inhibition of COX-1 and COX-2 with focus on preclinical to clinical translatability of cardio-renal data.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Heart/drug effects , Kidney/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Humans , Kidney/metabolism , Myocardium/metabolism
8.
Int J Toxicol ; 38(3): 215-227, 2019.
Article in English | MEDLINE | ID: mdl-30845865

ABSTRACT

Anatomically, the kidneys are paired, bean-shaped (in most mammals), excretory organs that lie in the retroperitoneum. High blood flow to the kidneys, together with high oxygen consumption, makes them more vulnerable to exposure, via the circulation, and subsequent injury related to high concentrations of xenobiotics and chemicals. In preclinical drug development and safety assessment of new investigational drugs, changes in kidney structure and/or function following drug administration in experimental laboratory animals need to be put in context with interspecies differences in kidney functional anatomy, physiology, spontaneous pathologies, and toxicopathological responses to injury. In addition, translation to human relevance to avoid premature drug termination from development is vital. Thus, detection and characterization of kidney toxicity in preclinical species and human relevance will depend on the preclinical safety testing strategy and collective weight-of-evidence approach including new investigational drug mechanism of action (MOA), preclinical and clinical interspecies differences, and MOA relevance to humans. This review describes kidney macroscopic and microscopic functional anatomy, physiology, pathophysiology, toxicology, and drug-induced kidney toxicities in safety risk assessment and drug development.


Subject(s)
Acute Kidney Injury/chemically induced , Drug-Related Side Effects and Adverse Reactions , Kidney/drug effects , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Drug Development , Humans , Kidney/anatomy & histology , Kidney/physiology
9.
Toxicol Pathol ; 46(8): 930-943, 2018 12.
Article in English | MEDLINE | ID: mdl-30282524

ABSTRACT

Pathophysiologically, the classification of acute kidney injury (AKI) can be divided into three categories: (1) prerenal, (2) intrinsic, and (3) postrenal. Emerging evidence supports the involvement of renal tubular epithelial cells and the innate and adaptive arms of the immune system in the pathogenesis of intrinsic AKI. Pro-inflammatory damage-associated molecular patterns, pathogen-associated molecular patterns, hypoxia inducible factors, toll-like receptors, complement system, oxidative stress, adhesion molecules, cell death, resident renal dendritic cells, neutrophils, T and B lymphocytes, macrophages, natural killer T cells, cytokines, and secreted chemokines contribute to the immunopathogenesis of AKI. However, other immune cells and pathways such as M2 macrophages, regulatory T cells, progranulin, and autophagy exhibit anti-inflammatory properties and facilitate kidney tissue repair after AKI. Thus, therapies for AKI include agents such as anti-inflammatory (e.g., recombinant alkaline phosphatase), antioxidants (iron chelators), and apoptosis inhibitors. In preclinical toxicity studies, drug-induced kidney injury can be seen after exposure to a nephrotoxicant test article due to immune mechanisms and dysregulation of innate, and/or adaptive cellular immunity. The focus of this review will be on intrinsic AKI, as it relates to the immune and renal systems cross talks focusing on the cellular and pathophysiologic mechanisms of AKI.


Subject(s)
Acute Kidney Injury/immunology , Acute Kidney Injury/physiopathology , Animals , Humans
10.
Curr Protoc Toxicol ; 76(1): e51, 2018 05.
Article in English | MEDLINE | ID: mdl-30040239

ABSTRACT

Cellular development and homeostasis are regulated via programmed cell death (PCD; apoptosis), which is a genetically regulated cellular process. Accidental cell death (ACD; necrosis) can be triggered by chemical, physical, or mechanical stress. Necrosis is the presence of dead tissues or cells in a living organism regardless of the initiating process and can be observed in infectious and non-infectious diseases and toxicities. This article describes tissue-based immunohistotechnical protocols used for assessing PCD and necrosis in formalin-fixed tissues obtained from preclinical species used in investigative and toxicologic pathology. Two commonly employed protocols for the identification of PCD and necrosis are described in this article: immunohistochemistry (IHC) for cleaved caspase 3, and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL). TUNEL has been used to detect DNA fragmentation by labeling the terminal ends of nucleic acids in necrotic and apoptotic cells. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Apoptosis , Cell Death , Necrosis/pathology , Pathology/methods , Toxicology/methods , Humans , Immunohistochemistry/methods , Necrosis/chemically induced
11.
Int J Toxicol ; 37(3): 223-233, 2018.
Article in English | MEDLINE | ID: mdl-29554821

ABSTRACT

Glomerulopathy and body weight gain were noted after chronic oral administration of a novel nonsteroidal dissociated agonist of the glucocorticoid receptor compound, fosdagrocorat, to beagle dogs fed an ad libitum diet. To further investigate the role of diet and treatment with either fosdagrocorat or the glucocorticoid comparator, prednisone, on renal safety, a 13-week investigative study was conducted in beagle dogs. Renal histopathology, clinical chemistry, urinalysis, glomerular filtration rate (GFR), body weight, heart rate, blood pressure (BP), and hematology were investigated in restricted- and ad libitum-fed dogs administered prednisone (2.2 mg/kg/d), fosdagrocorat (5 mg/kg/d), or vehicle for 13 weeks. Glomerulopathy was primarily observed in fosdagrocorat- and prednisone-treated ad libitum but not in feed-restricted or ad libitum vehicle-treated dogs. Kidneys in dogs from the prednisone-treated ad libitum had the greatest incidence and severity of tubular degenerative changes. Increased urine volume and decreased urine-specific gravity were present in prednisone- and fosdagrocorat-treated dogs, regardless of diet. These changes were not associated with consistent changes in GFR. Fosdagrocorat or prednisone treatment ad libitum dogs had the greatest increase in body weight gain. Sporadic changes in systolic and diastolic BP were noted in fosdagrocorat- and prednisone-treated groups. Significant reductions in serum cortisol and absolute eosinophils were noted in both ad libitum- and restriction-fed prednisone- and fosdagrocorat-treated dogs. In conclusion, prednisone-treated dogs fed ad libitum had greater glucocorticoid-induced renal effects than those dosed with fosdagrocorat.


Subject(s)
Kidney/drug effects , Organophosphates/pharmacology , Phenanthrenes/pharmacology , Prednisone/pharmacology , Receptors, Glucocorticoid/agonists , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Dogs , Eating/drug effects , Female , Food Deprivation , Glomerular Filtration Rate/drug effects , Heart Rate/drug effects , Kidney/pathology , Organophosphates/administration & dosage , Organophosphates/adverse effects , Phenanthrenes/administration & dosage , Phenanthrenes/adverse effects , Prednisone/adverse effects , Receptors, Glucocorticoid/drug effects
12.
Regul Toxicol Pharmacol ; 94: 306-322, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29454012

ABSTRACT

Tofacitinib, an oral Janus kinase (JAK) inhibitor for treatment of rheumatoid arthritis, targets JAK1, JAK3, and to a lesser extent JAK2 and TYK2. JAK1/3 inhibition impairs gamma common chain cytokine receptor signaling, important in lymphocyte development, homeostasis and function. Adult and juvenile cynomolgus monkey and rat studies were conducted and the impact of tofacitinib on immune parameters (lymphoid tissues and lymphocyte subsets) and function (T-dependent antibody response (TDAR), mitogen-induced T cell proliferation) assessed. Tofacitinib administration decreased circulating T cells and NK cells in juvenile and adult animals of both species. B cell decreases were observed only in rats. These changes and decreased lymphoid tissue cellularity are consistent with the expected pharmacology of tofacitinib. No differences were observed between juvenile and adult animals, either in terms of doses at which effects were observed or differential effects on immune endpoints. Lymphomas were observed in three adult monkeys. Tofacitinib impaired the primary TDAR in juvenile monkeys, although a recall response was generated. Complete or partial reversal of the effects on the immune system was observed.


Subject(s)
Aging/immunology , Janus Kinase Inhibitors/toxicity , Piperidines/toxicity , Pyrimidines/toxicity , Pyrroles/toxicity , Administration, Oral , Animals , Antigens/immunology , Erythrocyte Count , Female , Hematocrit , Hemocyanins/immunology , Hemoglobins/analysis , Janus Kinase Inhibitors/pharmacokinetics , Leukocyte Count , Leukocytes/drug effects , Leukocytes/immunology , Lymphoma, B-Cell/chemically induced , Macaca fascicularis , Male , Organ Size/drug effects , Piperidines/pharmacokinetics , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/pathology , Thymus Gland/drug effects , Thymus Gland/pathology , Toxicity Tests, Chronic
13.
Regul Toxicol Pharmacol ; 91: 93-102, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29074274

ABSTRACT

Tofacitinib is an oral JAK inhibitor for the treatment of rheumatoid arthritis. In the 2-year carcinogenicity study with tofacitinib, increased incidence of hibernoma (a neoplasm of brown adipose tissue [BAT]) was noted in female rats at ≥30 mg/kg/day (≥41x human exposure multiples). Thus, signaling pathways within BAT were investigated by measuring BAT: weight, cell proliferation biomarkers, content of basal and prolactin-induced phosphorylated Signal Transducer and Activator of Transcription (STAT), and uncoupling protein 1 (UCP-1). The relationship between cardiovascular hemodynamics and plasma norepinephrine (NE) levels was also investigated. Tofacitinib administered to female rats at doses of 10, 30, or 75 mg/kg/day for 14 days increased BAT weight at 75 mg/kg/day and cell proliferation at ≥30 mg/kg/day. JAK inhibition, observed as lower pSTAT3 and pSTAT5 in BAT, was noted at ≥10 mg/kg/day, while lower activity of BAT was observed as lower UCP-1 protein at ≥30 mg/kg/day. In cultured brown adipocytes, prolactin-induced increase in pSTAT5 and pSTAT3 were inhibited by tofacitinib in a concentration-dependent manner. Tofacitinib lowered blood pressure, increased heart rate, and resulted in dose-dependent increases in circulating NE. Thus, JAK/STAT inhibition in BAT and sympathetic stimulation are two factors which might contribute to the genesis of hibernomas by tofacitinib in rats.


Subject(s)
Lipoma/chemically induced , Piperidines/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyrimidines/adverse effects , Pyrroles/adverse effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Blood Pressure/drug effects , Cell Proliferation/drug effects , Female , Heart Rate/drug effects , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/pharmacology , Lipoma/metabolism , Male , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects , Trans-Activators/adverse effects , Trans-Activators/pharmacology
14.
Toxicol Sci ; 155(1): 148-156, 2017 01.
Article in English | MEDLINE | ID: mdl-27708194

ABSTRACT

Tofacitinib is an oral Janus kinase (JAK) inhibitor for the treatment of rheumatoid arthritis. Tofacitinib preferentially inhibits receptor signaling through JAK3 and JAK1, relative to JAK2. In the 2-year rat carcinogenicity study, there were tofacitinib, dose-related increases in the incidences of testicular Leydig cell hyperplasia and benign adenomas in male rats, and decreased incidences of mammary tumors and duct dilatation/galactocele in female rats. Such findings in rats are typical of agents, such as dopamine agonists, which decrease prolactin (PRL) activity. Since prolactin signals through the JAK2 pathway, we hypothesized that these findings were off-target effects due to inhibition of PRL signaling via JAK2. The studies reported here were designed to investigate the interruption of PRL signaling pathways in Leydig cells. In isolated primary rat Leydig cells, PRL increased phosphorylated Signal Transducer and Activator of Transcription-5 protein, and mRNA levels for luteinizing hormone receptor. Tofacitinib, at concentrations observed in the rat carcinogenicity study, dose-dependently inhibited these effects. These observations illustrate a novel mechanism, the inhibition of prolactin signaling by which modulation of JAK activity can modulate PRL signaling pathways to induce Leydig cell tumors in rats. Since human Leydig cells lack this PRL dependence for normal function, these rodent tumors do not indicate a health risk to human patients.


Subject(s)
Adenoma/pathology , Hyperplasia/chemically induced , Leydig Cells/drug effects , Piperidines/pharmacology , Prolactin/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Testis/drug effects , Animals , Leydig Cells/metabolism , Male , Rats , Testis/pathology
15.
Exp Toxicol Pathol ; 66(5-6): 263-5, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24795276

ABSTRACT

An incidental, asymptomatic, focal inflammatory lesion was detected in brain cerebrum of an approximately 6-year-old, female cynomolgus macaque from a chronic toxicology study. No gross lesions were noted at necropsy. Microscopically, the lesion contained a cross-section of larvae approximately 70-80 µm in diameter, a centrally located intestine flanked on either side by large triangular excretory columns, and prominent single lateral cuticular alae. Mixed inflammatory cells of eosinophils, macrophages, and lymphocytes admixed with abundant connective tissue stroma and necrosis surrounded the larvae. Histochemical stains for trichrome revealed significant amount of fibrous connective tissue. The morphology of the larvae was compatible with Baylisascaris spp. Based on the microscopic and histochemical examination, a diagnosis of neural Baylisascaris spp. larva migrans was made.


Subject(s)
Ascaridida Infections/veterinary , Ascaridoidea/isolation & purification , Brain/parasitology , Helminthiasis, Animal/parasitology , Larva Migrans/veterinary , Monkey Diseases/parasitology , Animals , Ascaridida Infections/parasitology , Ascaridida Infections/pathology , Brain/pathology , Female , Helminthiasis, Animal/pathology , Larva Migrans/parasitology , Larva Migrans/pathology , Macaca fascicularis , Monkey Diseases/pathology , Toxicity Tests/veterinary
16.
Comp Med ; 64(1): 68-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24512964

ABSTRACT

An incidental, asymptomatic, well-circumscribed, solitary, submucosal nodular mass was detected on the mucosal surface of the inner lower lip in a female cynomolgus macaque (age, approximately 2.4 y) during a juvenile chronic toxicology study. Grossly, the nodule was soft with brown to tan discoloration and measured approximately 4 mm in diameter. Microscopically, the nodule was covered by normal stratified squamous epithelium and composed of well-circumscribed irregular lobules containing hyperplastic and normal-appearing mucinous salivary gland acini and ducts, which were separated by thick connective tissue septae. In light of the gross pathology and results of microscopic examination, salivary gland hamartoma was diagnosed. This lesion resembles adenomatoid hyperplasia of mucous salivary glands in humans, which is a rare nonneoplastic swelling. To our knowledge, this case description is the first report of a cynomolgus macaque with the rare entity of lip salivary gland hamartoma, which likely represents adenomatous hyperplasia in humans.


Subject(s)
Hamartoma/veterinary , Lip Neoplasms/veterinary , Macaca fascicularis , Monkey Diseases/pathology , Salivary Gland Neoplasms/veterinary , Animals , Autopsy/veterinary , Biopsy/veterinary , Female , Hamartoma/pathology , Incidental Findings , Lip Neoplasms/pathology , Salivary Gland Neoplasms/pathology , Tumor Burden
17.
Toxicol Pathol ; 42(1): 118-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24178572

ABSTRACT

Sphingosine-1-phosphate (S1P) is a major bioactive phospholipid, which binds to and activates a family of five G-protein-coupled receptors designated as S1P 1 (S1P1) through S1P5. The S1P1 receptor subtype, expressed primarily on lymphocytes, is known to play a critical role in the regulation of lymphocyte trafficking. S1P1 inhibitors result in the inhibition of lymphoid cell trafficking and are of interest to treat various inflammatory conditions. In this study, we describe a gastric finding associated with oral gavage administration of a small molecule S1P1 inhibitor to Sprague-Dawley rats. Rats were administered an S1P1 inhibitor once daily for 4 weeks and necropsies were conducted at the end of the dosing phase, and clinical pathology and histopathologic examination were performed. Lymphopenia and changes in lymphoid tissues were noted and were consistent with the pharmacodynamic effects for S1P1 inhibitory action. Histopathologic examination of the stomach revealed atrophy and depletion of gastric parietal cells in the glandular portion of the stomach. There are no literature data to suggest that this gastric effect is related to S1P1 pharmacology. Therefore, the mechanism of the observed gastric lesion is likely chemotype mediated.


Subject(s)
Lysophospholipids/antagonists & inhibitors , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/pathology , Sphingosine/analogs & derivatives , Administration, Oral , Animals , Atrophy , Female , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphoid Tissue/pathology , Male , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/antagonists & inhibitors , Stomach/pathology
18.
Toxicol Pathol ; 41(1): 32-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22700649

ABSTRACT

Renal tubular inclusion bodies are rarely associated with drug administration. The authors describe the finding of renal cortical tubular intranuclear and intracytoplasmic inclusion bodies associated with the oral administration of a norepinephrine/serotonin reuptake inhibitor (NSRI) test article in Sprague-Dawley (SD) rats. Rats were given an NSRI daily for 4 weeks, and kidney histopathologic, ultrastructural pathology, and immunohistochemical examinations were performed. Round eosinophilic intranuclear inclusion bodies were observed histologically in the tubular epithelial cells of the renal cortex in male and female SD rats given the NSRI compound. No evidence of degeneration or necrosis was noted in the inclusion-containing renal cells. By ultrastructural pathology, inclusion bodies consisted of finely granular, amorphous, and uniformly stained nonmembrane-bound material. By immunohistochemistry, inclusion bodies stained positive for d-amino acid oxidase (DAO) protein. In addition, similar inclusion bodies were noted in the cytoplasmic tubular epithelial compartment by ultrastructural and immunohistochemical examination.  This is the first description of these renal inclusion bodies after an NSRI test article administration in SD rats. Such drug-induced renal inclusion bodies are rat-specific, do not represent an expression of nephrotoxicity, represent altered metabolism of d-amino acids, and are not relevant to human safety risk assessment.


Subject(s)
Inclusion Bodies/pathology , Kidney Cortex/physiopathology , Kidney Diseases/physiopathology , Kidney Tubules/physiopathology , Animals , D-Amino-Acid Oxidase/chemistry , D-Amino-Acid Oxidase/metabolism , Female , Immunohistochemistry , Inclusion Bodies/chemistry , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Kidney/chemistry , Kidney/cytology , Kidney/pathology , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Necrosis/pathology , Rats , Rats, Sprague-Dawley
19.
Regul Toxicol Pharmacol ; 64(3): 435-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23088954

ABSTRACT

The occurrence of mesenteric lymph node angiomas (benign vascular neoplasms including lymphangioma and hemangioma) in untreated control rats in 2-year carcinogenicity studies can range from rare to common depending on the strain used. This lesion is most common in male rats. Factors and conditions that may contribute to the etiopathogenesis of lymph node angiomas in rats include: (1) genetic drift, (2) congenital/developmental malformation, (3) sinus vascular transformation/venous obstruction of outflow, (4) "inflammatory" pseudo-tumors, and/or (5) defects of endothelial lymphatic vascular secretion/permeability. Lymph node angiomas in humans are extremely rare, not reported in mesenteric lymph nodes, and more common in females than males. The evaluation of increased mesenteric lymph node angiomas in rats for overall human safety risk assessment of novel pharmaceutical therapeutics should consider: genotoxicity of the test article, occurrence of vascular neoplasms in other locations in rats and in mice, occurrence of proliferative vascular lesions in nonclinical toxicology studies in non-rodent species, dose/exposure response, and pathophysiologic/morphologic differences and similarities of lymph node angiomas between rats and humans. Angiomas are independent lesions from angiosarcomas and are not precursors for angiosarcomas in either humans or animals. Mesenteric lymph node angiomas in rats are unlikely to be relevant for human risk assessment of pharmaceutical agents.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Hemangioma/etiology , Lymphangioma/etiology , Animals , Drug Design , Female , Hemangioma/epidemiology , Humans , Lymph Nodes/pathology , Lymphangioma/epidemiology , Male , Mesentery/pathology , Mice , Rats , Risk Assessment/methods , Sex Factors , Species Specificity
20.
Curr Protoc Toxicol ; Chapter 23: Unit 23.3.1-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22896010

ABSTRACT

This unit describes in detail the in vitro methods for measuring the interaction of new chemical entities (NCEs) with human renal transporters (hOAT1, hOAT2, and hOCT2) as both a substrate and inhibitor. Renal transporter substrate assays help in the identification of renal secretion mechanisms and assessment of the potential renal drug-drug interactions (DDIs) for NCE as a target, as well as to predict its renal clearance in humans. Human renal transporter (hOAT1, hOAT2, and hOCT2) inhibition assays characterize the inhibition potency of NCE and predict the potential for renal DDIs as a perpetrator with xenobiotics and drugs that are mainly renally cleared. In addition, such inhibition assays enable a better assessment of the potential for renal transporter-mediated nephrotoxicity and pathology. Therefore, renal transporter substrate and inhibition assays are pivotal in drug discovery and development for renally cleared drugs and those that are co-administered with marketed compounds mainly eliminated via the kidney.


Subject(s)
Biological Transport/physiology , Carrier Proteins/metabolism , Kidney/metabolism , Pharmaceutical Preparations/metabolism , Toxicity Tests/methods , Biological Assay/methods , Biological Transport/drug effects , Carrier Proteins/drug effects , Drug Evaluation, Preclinical/methods , Drug Interactions , Humans , Kidney/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...