Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Sci Transl Med ; 15(681): eabq5068, 2023 02.
Article in English | MEDLINE | ID: mdl-36724241

ABSTRACT

Immunogenicity against intravitreally administered brolucizumab has been previously described and associated with cases of severe intraocular inflammation, including retinal vasculitis/retinal vascular occlusion (RV/RO). The presence of antidrug antibodies (ADAs) in these patients led to the initial hypothesis that immune complexes could be key mediators. Although the formation of ADAs and immune complexes may be a prerequisite, other factors likely contribute to some patients having RV/RO, whereas the vast majority do not. To identify and characterize the mechanistic drivers underlying the immunogenicity of brolucizumab and the consequence of subsequent ADA-induced immune complex formation, a translational approach was performed to bridge physicochemical characterization, structural modeling, sequence analysis, immunological assays, and a quantitative systems pharmacology model that mimics physiological conditions within the eye. This approach revealed that multiple factors contributed to the increased immunogenic potential of brolucizumab, including a linear epitope shared with bacteria, non-natural surfaces due to the single-chain variable fragment format, and non-native drug species that may form over prolonged time in the eye. Consideration of intraocular drug pharmacology and disease state in a quantitative systems pharmacology model suggested that immune complexes could form at immunologically relevant concentrations modulated by dose intensity. Assays using circulating immune cells from treated patients or treatment-naïve healthy volunteers revealed the capacity of immune complexes to trigger cellular responses such as enhanced antigen presentation, platelet aggregation, endothelial cell activation, and cytokine release. Together, these studies informed a mechanistic understanding of the clinically observed immunogenicity of brolucizumab and associated cases of RV/RO.


Subject(s)
Antigen-Antibody Complex , Root Cause Analysis , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Inflammation , Angiogenesis Inhibitors , Intravitreal Injections
2.
Front Immunol ; 11: 745, 2020.
Article in English | MEDLINE | ID: mdl-32425939

ABSTRACT

Genetic disruption or short-term pharmacological inhibition of MALT1 protease is effective in several preclinical models of autoimmunity and B cell malignancies. Despite these protective effects, the severe reduction in regulatory T cells (Tregs) and the associated IPEX-like pathology occurring upon congenital disruption of the MALT1 protease in mice has raised concerns about the long-term safety of MALT1 inhibition. Here we describe the results of a series of toxicology studies in rat and dog species using MLT-943, a novel potent and selective MALT1 protease inhibitor. While MLT-943 effectively prevented T cell-dependent B cell immune responses and reduced joint inflammation in the collagen-induced arthritis rat pharmacology model, in both preclinical species, pharmacological inhibition of MALT1 was associated with a rapid and dose-dependent reduction in Tregs and resulted in the progressive appearance of immune abnormalities and clinical signs of an IPEX-like pathology. At the 13-week time point, rats displayed severe intestinal inflammation associated with mast cell activation, high serum IgE levels, systemic T cell activation and mononuclear cell infiltration in multiple tissues. Importantly, using thymectomized rats we demonstrated that MALT1 protease inhibition affects peripheral Treg frequency independently of effects on thymic Treg output and development. Our data confirm the therapeutic potential of MALT1 protease inhibitors but highlight the safety risks and challenges to consider before potential application of such inhibitors into the clinic.


Subject(s)
Diabetes Mellitus, Type 1/congenital , Diarrhea/etiology , Genetic Diseases, X-Linked/etiology , Immune System Diseases/congenital , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Diabetes Mellitus, Type 1/etiology , Dogs , Female , Humans , Immune System Diseases/etiology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Rats, Wistar , T-Lymphocytes, Regulatory/immunology
3.
Arch Toxicol ; 92(5): 1877-1891, 2018 05.
Article in English | MEDLINE | ID: mdl-29556671

ABSTRACT

A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans.


Subject(s)
Azetidines/adverse effects , Benzyl Compounds/adverse effects , Endothelial Cells/drug effects , Hemangiosarcoma/chemically induced , Toxicity Tests, Chronic/methods , Administration, Oral , Animals , Azetidines/administration & dosage , Benzyl Compounds/administration & dosage , Cells, Cultured , Endothelium, Vascular/cytology , Hemangiosarcoma/genetics , Humans , Male , Mice, Inbred Strains , Placenta Growth Factor/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Species Specificity , Toxicokinetics , Transcriptome/drug effects
4.
Exp Toxicol Pathol ; 69(5): 275-283, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-28190562

ABSTRACT

Early detection of drug-induced alterations of hemostasis is challenging. Drugs can affect different components of the Virchow's triad and measurement of plasmatic coagulation times lacks sensitivity. New techniques for a more global assessment of the hemostasis are now available: the impedance platelet aggregometry, the thromboelastography and the thrombin generation measurement. The aim of this study was to evaluate three techniques (i.e.: Multiplate®, TEG® and CAT) for the in vitro detection of the effect of a drug known to induce hemostatic alterations in a preclinical safety environment. Cyclosporine A was chosen and tested at 4 concentrations after solubilization in DMSO in Wistar rats and Beagle dogs. The results obtained were comparable between both species except for the thrombin generation in platelet rich plasma. Enhanced platelet aggregability was observed after ADP stimulation and alterations of the thromboelastograms consisted in decreased maximum amplitude and increased LY30. A dual effect on thrombin generation was observed and suggested that CsA may interact with platelets in rat platelet rich plasma and speed up thrombin generation. The results of this study indicate that using a combined approach on hemostasis testing in preclinical safety it is possible to detect in vitro drug-induced alterations of hemostasis.


Subject(s)
Blood Coagulation Tests/methods , Cyclosporine/toxicity , Drug Evaluation, Preclinical/methods , Hemostasis/drug effects , Immunosuppressive Agents/toxicity , Animals , Dogs , Female , Male , Rats , Rats, Wistar
5.
Toxicol Sci ; 155(1): 283-297, 2017 01.
Article in English | MEDLINE | ID: mdl-27742868

ABSTRACT

The aim of this study was to determine the relative safety of 4 antiviral drugs (telbivudine, tenofovir, adefovir, and entecavir) against hepatitis B virus with respect to kidney function and toxicity in male Sprague Dawley rats. The antiviral drugs were administered once daily for 4 weeks by oral gavage at ∼10 and 25-40 times the human equivalent dose. Main assessments included markers of renal toxicity in urine, magnetic resonance imaging (MRI) of kidney function, histopathology, and electron microscopic examination. Administration of adefovir at 11 and 28 mg/kg for 4 weeks caused functional and morphological kidney alterations in a time- and dose-dependent manner, affecting mainly the proximal tubules and suggesting a mechanism of toxicity related to mitochondrial degeneration/depletion. Of note, the observed adefovir-induced reduction of kidney function was not detected by the standard method of glomerular filtration rate (GFR) measurements (clearance rate of the endogenous marker, creatinine), thereby emphasizing the superiority of MRI in terms of sensitive detection of GFR in rats. For the low dose of 300 mg/kg of tenofovir, minor kidney effects such as nuclear enlargement in the tubular epithelium, and hyaline droplets accumulation were detected, which was also observed for the low dose (11 mg/kg) of adefovir. No assessments could be done at the higher dose of 600/1000 mg/kg tenofovir due to gastrointestinal tract toxicity which prevented treatment of the animals for longer than 1 week. Entecavir at 1 and 3 mg/kg and telbivudine at 600 and 1600 mg/kg caused no toxicologically relevant effects on the kidney.


Subject(s)
Antiviral Agents/adverse effects , Hepatitis B/drug therapy , Kidney/drug effects , Animals , Antiviral Agents/therapeutic use , Kidney/diagnostic imaging , Magnetic Resonance Imaging , Male , Rats , Rats, Sprague-Dawley
6.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26129975

ABSTRACT

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Subject(s)
Blood Platelets/enzymology , Lysophospholipids/blood , Phosphotransferases (Alcohol Group Acceptor)/blood , Platelet Aggregation , Sphingosine/analogs & derivatives , Animals , Arachidonic Acid/blood , Blood Coagulation , Blood Coagulation Tests , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Disease Models, Animal , Erythrocytes/enzymology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Platelet Adhesiveness , Platelet Function Tests , Receptors, Lysosphingolipid/blood , Signal Transduction , Sphingosine/blood , Sphingosine-1-Phosphate Receptors , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/prevention & control , Thromboxane A2/blood , Vascular System Injuries/blood , Vascular System Injuries/enzymology
7.
Exp Toxicol Pathol ; 67(4): 287-96, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25773704

ABSTRACT

BACKGROUND: An experimental rat model served for evaluation of bone- and energy metabolism in early and late stages of osteoporosis. For the early stage, we hypothesized that bilateral ovariectomy (OVX)+multi-deficiency diet (OVXD; depletion of vitamin D, calcium, vitamin K, phosphorus) would induce increased bone turnover while the late stage would be characterized by enhanced bone catabolism. Obesity, insulin resistance and hyperleptinemia would be seen during the whole course of disease. Healthy female Sprague Dawley rats (n=41) aged 10 weeks were randomly assigned to sham and treatment groups and sacrificed at 3, 12, and 14 months after the study began. RESULTS: In the early phase, OVXD was associated with an increase in body weight, but not, however, in later stages. There was a decrease in bone mineral density and relative bone volume (BV/TV) as assessed by Dual Energy X-ray Absorptiometry and micro computed tomography that was most severe in the later stages of disease, indicating bone catabolism. Osteocalcin limiting bone formation was increased initially, whereas later stages (14 months) were characterized by elevated osteopontin, suggesting bone remodeling. Severe hyperparathyroidism was present during all stages of disease. Only the early phases of disease were characterized by hyperinsulinemia and increased adrenocorticotrophic stimulating hormone, whereas in the late stage hypoleptinemia rather than hyperleptinemia was seen. CONCLUSION: Markers of bone and energy metabolism reflected both an increased bone turn over and ongoing bone remodeling associated with initial hyperinsulinemia. Osteopontin and osteocalcin can be used to differentiate early and late stages of osteoporosis.


Subject(s)
Bone Remodeling/physiology , Energy Metabolism/physiology , Osteoporosis/metabolism , Animals , Bone Density , Disease Models, Animal , Female , Osteoporosis/etiology , Ovariectomy , Rats , Rats, Sprague-Dawley , X-Ray Microtomography
8.
J Pharmacol Toxicol Methods ; 75: 101-10, 2015.
Article in English | MEDLINE | ID: mdl-25637943

ABSTRACT

INTRODUCTION: With the recent development of more sensitive biomarkers to assess kidney injury preclinically, a survey was designed i) to investigate what strategies are used to investigate renal toxicity in both ICH S7A compliant Safety Pharmacology (SP) studies after a single dose of a compound and within repeat-dose toxicity studies by large pharmaceutical companies today; ii) to understand whether renal SP studies have impact or utility in drug development and/or if it may be more appropriate to assess renal effects after multiple doses of compounds; iii) to ascertain how much mechanistic work is performed by the top 15 largest pharmaceutical companies (as determined by R&D revenue size); iv) to gain an insight into the impact of the validation of DIKI biomarkers and their introduction in the safety evaluation paradigm; and v) to understand the impact of renal/urinary safety study data on progression of projects. METHODS: Two short anonymous surveys were submitted to SP leaders of the top 15 pharmaceutical companies, as defined by 2012 R&D portfolio size. Fourteen multiple choice questions were designed to explore the strategies used to investigate renal effects in both ICH S7A compliant SP studies and within toxicology studies. RESULTS: A 67% and 60% response rate was obtained in the first and second surveys, respectively. Nine out of ten respondent companies conduct renal excretory measurements (eg. urine analysis) in toxicology studies whereas only five out of ten conduct specific renal SP studies; and all of those 5 also conduct the renal excretory measurements in toxicology studies. These companies measure and/or calculate a variety of parameters as part of these studies, and also on a case by case basis include regulatory qualified and non-qualified DIKI biomarkers. Finally, only one company has used renal/urinary functional data alone to stop a project, whereas the majority of respondents combine renal data with other target organ assessments to form an integrated decision-making set. CONCLUSION: These short surveys highlighted areas of similarity: a) urinary measurements are most commonly taken on repeat-dose toxicity studies, and b) renal SP studies are less often utilised. The two major differences are a) lack of consistent use of DIKI biomarkers in urinary safety studies and b) the way large pharmaceutical companies assess renal function. Finally, suggestions were made to improve the safety assessment methods for determining the safety of compounds with potential renal liability.


Subject(s)
Drug Evaluation, Preclinical/methods , Drug Industry/methods , Kidney Diseases/chemically induced , Animals , Biomarkers/metabolism , Drug Design , Drug Industry/statistics & numerical data , Drug-Related Side Effects and Adverse Reactions , Humans , Surveys and Questionnaires , Toxicity Tests/methods
9.
Toxicol Pathol ; 43(5): 694-703, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25630683

ABSTRACT

Sphingosine-1-phosphate (S1P) lyase is considered as a drug target in autoimmune diseases based on the protective effect of reducing activity of the enzyme in animal models of inflammation. Since S1P lyase deficiency in mice causes a severe, lethal phenotype, it was of interest to investigate any pathological alterations associated with only partially reduced activity of S1P lyase as may be encountered upon pharmacological inhibition. Both genetic reduction of S1P lyase activity in mice and inhibition of S1P lyase with a low-molecular-weight compound in rats consistently resulted in podocyte-based kidney toxicity, which is the most severe finding. In addition, skin irritation and platelet activation were observed in both instances. The similarity of the findings in both the genetic model and the pharmacological study supports the value of analyzing inducible partially target-deficient mice for safety assessment. If the findings described in rodents translate to humans, target-related toxicity, particularly podocyte dysfunction, may limit chronic systemic treatment of autoimmune diseases with S1P lyase inhibitors. Furthermore, partial deficiency or inhibition of S1P lyase appears to provide an in vivo rodent model to enable studies on the mechanism of podocyte dysfunction.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Aldehyde-Lyases/metabolism , Platelet Activation/physiology , Podocytes/enzymology , Proteinuria/enzymology , Aldehyde-Lyases/genetics , Animals , Female , Kidney/enzymology , Kidney/pathology , Male , Mice , Proteinuria/blood , Rats , Skin/enzymology , Skin/pathology , Tamoxifen/pharmacology
10.
Exp Toxicol Pathol ; 65(5): 637-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22884258

ABSTRACT

This study was performed to optimize and standardize the use of the Multiplate(®) whole blood impedance aggregometer in the Beagle dog and Wistar rat for use in a research laboratory environment. The anticoagulants citrate, heparin and hirudin were compared and platelet aggregation responses to ADP, collagen, arachidonic acid and Par-4 agonist were evaluated to determine their half maximal effective concentrations (EC(50)) in blood containing low concentrations of a drug solvent (0.1% DMSO). The results indicate that citrate anticoagulation is not suitable for Multiplate(®) whole blood aggregometry because of the presence of spontaneous aggregation. ADP and collagen were found to be appropriate agonists for both species, whereas in the Beagle dog Par-4 agonist failed to induce aggregation and arachidonic acid induced platelet aggregation showed a high interindividual variability. The agonists EC(50) calculated in hirudin blood were 2.70 µM ADP, 0.85 µg/ml collagen, 0.03 mM arachidonic acid and 165.7 µM Par-4 agonist in the Wistar rat, and 0.95 µM ADP and 0.23 µg/ml collagen in the Beagle dog.


Subject(s)
Blood Platelets/drug effects , Drug-Related Side Effects and Adverse Reactions/blood , Platelet Aggregation/drug effects , Platelet Function Tests/methods , Toxicity Tests/methods , Animals , Dogs , Platelet Aggregation Inhibitors/pharmacology , Platelet Function Tests/standards , Rats , Rats, Wistar , Species Specificity , Toxicity Tests/standards
11.
Leuk Res ; 35(5): 631-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21129774

ABSTRACT

In vitro, concentrations ≥ 10 µM of nilotinib were needed to induce markers of cytotoxicity, apoptosis, and endoplasmic reticulum stress in both neonatal rat ventricular myocytes, a putative target tissue, and non-target heart fibroblasts, indicating a lack of cardiomyocyte-specific nilotinib toxicity in vitro. In rats, oral nilotinib treatment at 80 mg/kg for 4 weeks induced increased heart weight; however, this was not associated with relevant histopathological changes or effects on heart function. Thus, nilotinib at and above clinically relevant concentrations (4.27 µM) did not induce overt cardiovascular pathologies or heart failure in vitro or in vivo under study conditions.


Subject(s)
Cardiotoxins , Heart/drug effects , Pyrimidines/adverse effects , Animals , Animals, Newborn , Cardiotoxins/adverse effects , Cardiotoxins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Heart/physiology , Heart Ventricles/anatomy & histology , Heart Ventricles/drug effects , Heart Ventricles/ultrastructure , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pyrimidines/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Rats, Wistar , Ventricular Function/drug effects
12.
Exp Toxicol Pathol ; 63(1-2): 187-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20724123

ABSTRACT

The International Federation of Societies of Toxicologic Pathologists (IFSTP) proposes a common global framework for training future toxicologic pathologists who will support regulatory-type - nonclinical toxicology studies. Trainees optimally should undertake a scientific curriculum of at least 5 years at an accredited institution leading to a clinical degree (veterinary medicine or medicine). Trainees should then obtain 4 or more years of intensive pathology practice during a residency and/or on-the-job "apprenticeship," at least 2 years of which must be focused on regulatory-type toxicologic pathology topics. Possession of a recognized pathology qualification (i.e., certification) is highly recommended. A nonclinical pathway (e.g., a graduate degree in medical biology or pathology) may be possible if medically trained pathologists are scarce, but this option is not optimal. Regular, lifelong continuing education (peer review of nonclinical studies, professional meetings, reading, short courses) will be necessary to maintain and enhance one's understanding of current toxicologic pathology knowledge, skills, and tools. This framework should provide a rigorous yet flexible way to reliably train future toxicologic pathologists to generate, interpret, integrate, and communicate data in regulatory-type, nonclinical toxicology studies.


Subject(s)
Education, Professional/methods , Pathology/education , Professional Competence/standards , Toxicity Tests/standards , Toxicology/education , Guidelines as Topic , International Cooperation , Pathology/standards , Toxicity Tests/methods , Toxicology/standards
13.
Lab Anim ; 44(4): 352-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696790

ABSTRACT

In a large number of animal experiments, blood collection is crucial for achieving the study aim. Requirements on sampling techniques used include their practicability, their effectiveness in terms of obtaining the desired blood volume, sample quality and low impact on animal's wellbeing. Numerous methods for blood collection from mice have been published. For large blood volumes, submandibular and sublingual bleeding was developed as alternatives to the retrobulbar bleeding method, which is considered controversial as it results in severe tissue damage. Only a few studies report the use of submandibular and sublingual techniques in mice. In particular, the degree of tissue damage or influence on animal's wellbeing due to submandibular bleeding in conscious mice has not yet been clearly assessed. This gap in the knowledge base has been filled by our exploratory study that revealed clear differences between both techniques. Defence movements during submandibular blood collection in conscious mice resulted in more complications and revealed insufficient blood sample quality due to the prolonged duration of blood collection. In addition, it is likely that these movements may have caused lesions to be more pronounced. Changes in red blood cell parameters (red blood cell count, haemoglobin and haematocrit), in glucose and in total protein concentrations observed in anaesthetized animals were most likely related to anaesthesia. Sublingually punctured mice gained significantly more body weight than submandibularly punctured mice, likely due to less severe tissue lesions and improved healing processes. Based on these results, we recommend the sublingual blood collection technique to be used in mice. However, if the submandibular bleeding technique is used, it should be performed in anaesthetized mice only.


Subject(s)
Blood Specimen Collection/veterinary , Body Weight/physiology , Mandible/blood supply , Stress, Physiological/physiology , Tongue/blood supply , Animals , Blood Specimen Collection/adverse effects , Blood Specimen Collection/methods , Eating , Female , Laboratory Animal Science/methods , Mandibular Injuries/etiology , Mice , Mice, Inbred Strains , Tongue/injuries
14.
Toxicol Pathol ; 38(6): 984-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20716784

ABSTRACT

The International Federation of Societies of Toxicologic Pathologists (IFSTP) proposes a common global framework for training future toxicologic pathologists who will support regulatory-type, nonclinical toxicology studies. Optimally, trainees should undertake a scientific curriculum of at least five years at an accredited institution leading to a clinical degree (veterinary medicine or medicine). Trainees should then obtain four or more years of intensive pathology practice during a residency and/or on-the-job "apprenticeship," at least two years of which must be focused on regulatory-type toxicologic pathology topics. Possession of a recognized pathology qualification (i.e., certification) is highly recommended. A nonclinical pathway (e.g., a graduate degree in medical biology or pathology) may be possible if medically trained pathologists are scarce, but this option is not optimal. Regular, lifelong continuing education (peer review of nonclinical studies, professional meetings, reading, short courses) will be necessary to maintain and enhance one's understanding of current toxicologic pathology knowledge, skills, and tools. This framework should provide a rigorous yet flexible way to reliably train future toxicologic pathologists to generate, interpret, integrate, and communicate data in regulatory-type, nonclinical toxicology studies.


Subject(s)
Education , International Cooperation , Pathology/education , Professional Competence , Toxicology/education , Animals , Animals, Laboratory , Certification , Health Planning Guidelines , Pathology/standards , Toxicology/standards
15.
Mol Cancer Ther ; 9(7): 1945-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20587663

ABSTRACT

The recent discovery of an acquired activating point mutation in JAK2, substituting valine at amino acid position 617 for phenylalanine, has greatly improved our understanding of the molecular mechanism underlying chronic myeloproliferative neoplasms. Strikingly, the JAK2(V617F) mutation is found in nearly all patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia and primary myelofibrosis. Thus, JAK2 represents a promising target for the treatment of myeloproliferative neoplasms and considerable efforts are ongoing to discover and develop inhibitors of the kinase. Here, we report potent inhibition of JAK2(V617F) and JAK2 wild-type enzymes by a novel substituted quinoxaline, NVP-BSK805, which acts in an ATP-competitive manner. Within the JAK family, NVP-BSK805 displays more than 20-fold selectivity towards JAK2 in vitro, as well as excellent selectivity in broader kinase profiling. The compound blunts constitutive STAT5 phosphorylation in JAK2(V617F)-bearing cells, with concomitant suppression of cell proliferation and induction of apoptosis. In vivo, NVP-BSK805 exhibited good oral bioavailability and a long half-life. The inhibitor was efficacious in suppressing leukemic cell spreading and splenomegaly in a Ba/F3 JAK2(V617F) cell-driven mouse mechanistic model. Furthermore, NVP-BSK805 potently suppressed recombinant human erythropoietin-induced polycythemia and extramedullary erythropoiesis in mice and rats.


Subject(s)
Cell Proliferation/drug effects , Janus Kinase 2/antagonists & inhibitors , Polycythemia/prevention & control , Quinoxalines/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Erythropoiesis/drug effects , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , K562 Cells , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Molecular , Molecular Structure , Mutation , Phosphorylation/drug effects , Polycythemia/metabolism , Polycythemia/pathology , Protein Structure, Tertiary , Quinoxalines/chemistry , Rats , STAT5 Transcription Factor/metabolism , Splenomegaly/metabolism , Splenomegaly/pathology , Splenomegaly/prevention & control
16.
Toxicol Pathol ; 38(5): 810-28, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20585144

ABSTRACT

Hepatic drug-metabolizing enzyme (DME) induction is an adaptive response associated with changes in preclinical species; this response can include increases in liver weight, hepatocellular hyperplasia and hypertrophy, and upregulated tissue expression of DMEs. Effects of DME induction on clinical pathology markers of hepatobiliary injury and function in animals as well as humans are not well established. This component of a multipart review of the comparative pathology of xenobiotically mediated induction of hepatic metabolizing enzymes reviews pertinent data from retrospective and prospective preclinical and clinical studies. Particular attention is given to studies with confirmation of DME induction and concurrent evaluation of liver and/or serum hepatobiliary marker enzyme activities and histopathology. These results collectively indicate that in the rat, when histologic findings are limited to hepatocellular hypertrophy, DME induction is not expected to be associated with consistent or substantive changes in serum or plasma activity of hepatobiliary marker enzymes such as alanine aminotransferase, alkaline phosphatase, and gamma glutamyltransferase. In the dog and the monkey, published studies also do not demonstrate a consistent relationship across DME-inducing agents and changes in these clinical pathology parameters. However, increased liver alkaline phosphatase or gamma glutamyltransferase activity in dogs treated with phenobarbital or corticosteroids suggests that direct or indirect induction of select hepatobiliary injury markers can occur both in the absence of liver injury and independently of induction of DME activity. Although correlations between tissue and serum levels of these hepatobiliary markers are limited and inconsistent, increases in serum/plasma activities that are substantial or involve changes in other markers generally reflect hepatobiliary insult rather than DME induction. Extrahepatic effects, including disruption of the hypothalamic-pituitary-thyroid axis, can also occur as a direct outcome of hepatic DME induction in humans and animals. Importantly, hepatic DME induction and associated changes in preclinical species are not necessarily predictive of the occurrence, magnitude, or enzyme induction profile in humans.


Subject(s)
Enzyme Induction/physiology , Liver/enzymology , Liver/pathology , Xenobiotics/metabolism , Animals , Humans , Liver/drug effects
17.
Leuk Res ; 34(9): 1180-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20122731

ABSTRACT

Cytotoxic concentrations of imatinib mesylate (10-50 microM) were required to trigger markers of apoptosis and endoplasmic reticulum stress response in neonatal rat ventricular myocytes and fibroblasts, with no significant differences observed between c-Abl silenced and nonsilenced cells. In mice, oral or intraperitoneal imatinib treatment did not induce cardiovascular pathology or heart failure. In rats, high doses of oral imatinib did result in some cardiac hypertrophy. Multi-organ toxicities may have increased the cardiac workload and contributed to the cardiac hypertrophy observed in rats only. These data suggest that imatinib is not cardiotoxic at clinically relevant concentrations (5 microM).


Subject(s)
Antineoplastic Agents/adverse effects , Heart/drug effects , Piperazines/adverse effects , Pyrimidines/adverse effects , Animals , Base Sequence , Benzamides , DNA Primers , Heart/physiology , Imatinib Mesylate , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Rats , Rats, Wistar
18.
J Toxicol Pathol ; 23(3): 171-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-22272030

ABSTRACT

The International Federation of Societies of Toxicologic Pathologists (IFSTP) proposes a common global framework for training future toxicologic pathologists who will support regulatory-type nonclinical toxicology studies. Trainees optimally should undertake a scientific curriculum of at least 5 years at an accredited institution leading to a clinical degree (veterinary medicine or medicine). Trainees should then obtain 4 or more years of intensive pathology practice during a residency and/or on-the-job "apprenticeship," at least 2 years of which must be focused on regulatory-type toxicologic pathology topics. Possession of a recognized pathology qualification (i.e., certification) is highly recommended. A non-clinical pathway (e.g., a graduate degree in medical biology or pathology) may be possible if medically trained pathologists are scarce, but this option is not optimal. Regular, lifelong continuing education (peer review of nonclinical studies, professional meetings, reading, short courses) will be necessary to maintain and enhance one's understanding of current toxicologic pathology knowledge, skills, and tools. This framework should provide a rigorous yet flexible way to reliably train future toxicologic pathologists to generate, interpret, integrate, and communicate data in regulatory-type, nonclinical toxicology studies.

19.
Exp Toxicol Pathol ; 60(6): 443-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18603417

ABSTRACT

As opposed to erythropoiesis, which is regularly assessed in the peripheral blood of animals by reticulocyte count, thrombopoiesis is rarely assessed in assays that detect immature platelets in the peripheral blood. An assessment of recent thrombopoiesis is feasible with the analysis of reticulated platelets in the peripheral blood via flow cytometry, but rarely performed. The aim of this study was to establish an assay for the detection of reticulated platelets in whole blood of rats via flow cytometry, using a two-color staining method with a platelet-specific antibody (CD61-PE) and thiazole orange to detect RNA-containing platelets. Platelets were detected in K3EDTA-anticoagulated, paraformaldehyde-fixed samples, using a CD61-PE antibody as well as a gate specific for the light scatter properties of platelets. The intra-assay coefficient of variation varied between 3.6% and 8.3% (n=6 animals). The stability of the assay was determined by storing blood prior to staining, storing stained samples for up to 2h at room temperature, and by diluting the blood prior to analysis with autologous plasma to create samples with artificial anemia and thrombocytopenia. Only samples stored at room temperature prior to analysis showed a significantly lower percentage of reticulated platelets. Percentage of reticulated platelets in the reference population (n=41 rats) was 10.0+/-1.3% reticulated platelets (mean+/-SD; min=6.2%; max=12.5%). These data show that the detection of reticulated platelets in whole blood of rats using a platelet-specific antibody is feasible. This test presents a minimal-invasive method to assess thrombopoiesis in rats that can be used for example in preclinical toxicological studies.


Subject(s)
Blood Platelets/cytology , Flow Cytometry/methods , Platelet Count/methods , Thrombopoiesis/physiology , Toxicity Tests/methods , Animals , Blood Platelets/immunology , Rats , Rats, Wistar , Reproducibility of Results , Temperature , Time Factors
20.
Toxicol Appl Pharmacol ; 218(1): 52-63, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17157341

ABSTRACT

Vascular injury is a relatively common finding during the pre-clinical toxicity testing of drugs. The mechanisms of the injury are poorly understood and in turn, sensitive and specific biomarkers for pre-clinical and clinical monitoring do not exist. The present study was undertaken to investigate the molecular mechanisms of drug-induced vascular injury in mesenteric tissue of rats treated with the selective phosphodiesterase 4 (PDE4) inhibitor CI-1044. In a time-course study, male Sprague Dawley rats were given daily doses of 40 or 80 mg/kg for 1, 2 or 3 successive days and were euthanized the following day. Gene expression profiles in mesenteric tissue were determined using Affymetrix RG_U34A microarrays and fibrinogen and cytokine measurements were performed in blood samples. Hierarchical clustering analysis produced a clear pattern separation of the animals with inflammation, animal with inflammation and necrosis and animals without any lesion. Genes associated with inflammation, procoagulation, extracellular matrix remodeling were up-regulated. An altered expression of genes involved in vascular tone regulation, lipid and glucose metabolism was also observed. Selected genes expression changes were confirmed by TaqMan real-time RT-PCR. The inflammatory process was also detected in the bloodstream at the protein level since fibrinogen, IL6 and IL1beta concentrations were increased in treated animals. Overall, the present study reveals several molecular changes supporting the hypothesis by which PDE4 inhibitor-induced vascular lesions in rats are triggered by an inflammatory mechanism and/or a vascular tone dysregulation.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Azepines/toxicity , Gene Expression Regulation/drug effects , Mesenteric Arteries/drug effects , Mesentery/drug effects , Niacinamide/analogs & derivatives , Phosphodiesterase Inhibitors/toxicity , Animals , Cluster Analysis , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dose-Response Relationship, Drug , Fibrinogen/metabolism , Gene Expression Profiling , Inflammation/metabolism , Interleukins/blood , Male , Mesenteric Arteries/pathology , Mesentery/metabolism , Niacinamide/toxicity , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...