Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Toxicol Pathol ; 52(1): 21-34, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38379371

RESUMEN

In nonclinical toxicology studies, lab animals are fasted typically overnight, to reduce variability in some clinical pathology parameters. However, fasting adds undue stress, and this is particularly concerning in rodents given their fast metabolic rates. Furthermore, as rodents are nocturnal animals, an overnight fasting may cause a protracted negative metabolic state even when the fasting has technically ended, given their minimal activity and food consumption during the day. Therefore, to evaluate the impacts of different fasting durations (±DietGel supplementation) on rats' welfare, we assessed the traditional and ancillary clinical pathology parameters in Sprague-Dawley rats, along with body weight, organ weight, and histopathology. Although most endpoints were comparable between the different fasting durations (±DietGel supplementation), the long fasting times (≥8 hr) without DietGel supplementation caused significant decreases in body weight, liver weight, liver glycogen content, serum glucose, triglyceride, and creatinine concentrations-all findings suggestive of a negative energy balance that could impact animal welfare and consequently, data quality; while the short fasting time (4 hr) and DietGel supplementation were associated with higher triglycerides variability. Hence, we propose that short fasting time should be adequate for most toxicology studies in rats, and long fasting times should only be accommodated with scientific justification.


Asunto(s)
Bienestar del Animal , Peso Corporal , Ayuno , Ratas Sprague-Dawley , Animales , Ayuno/fisiología , Masculino , Ratas , Tamaño de los Órganos , Hígado/metabolismo , Femenino , Suplementos Dietéticos , Glucemia
2.
Animals (Basel) ; 13(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38136900

RESUMEN

Based on the current state of science, the use of animals remains essential in bringing safe and effective medicines to patients. Respect for laboratory animal welfare and the application of 3Rs principles (the replacement, reduction, and refinement of animal use in research) are a priority throughout the pharmaceutical industry. Given the rapid pace of development, technological progress, and the emergence of new-approach methodologies (NAMs) in the field of biomedical research, maintaining a leading position in scientific advancements with a focus on the principles of replace, reduce, and refine (3Rs) can be quite challenging. To effectively address these challenges and sustain a prominent position in the scientific community, organizations can derive significant advantages from establishing an internal 3Rs advisory group (3Rs AG). The primary objective of a 3Rs AG is to stay at the forefront of the knowledge of best practices related to the 3Rs principles in the industry. This group plays a crucial role in fostering innovation and facilitating the seamless integration and implementation of 3Rs principles into a company's policies and procedures. The thoughtful reduction in and replacement of animal studies and the refinement of study designs and practices, enabled by a 3Rs AG, can minimize animal use as well as guide resources and positively impact study and data quality. This article provides guidance on how to establish a successful and impactful 3Rs AG.

3.
Toxicol Pathol ; 51(5): 264-277, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37702042

RESUMEN

During toxicology studies, fasting animals prior to clinical pathology blood collection is believed to reduce variability in some clinical chemistry analytes. However, fasting adds stress to animals that are already stressed from the administration of potentially toxic doses of the test article. The purpose of this study was to assess the impacts of different fasting durations on cynomolgus monkeys' welfare during toxicology studies. To this end, we assessed the cynomolgus monkeys traditional and ancillary clinical pathology endpoints at different fasting times. We showed that most clinical pathology endpoints were largely comparable between different fasting times suggesting that cynomolgus monkeys could be fasted for as little as 4 hours for toxicology studies, as longer fasting times (up to 20 hours) resulted in stress, dehydration, and significant decreases in blood glucose- changes that impacts animal welfare. Shorter fasting times were associated with higher triglycerides variability among individual animals. Therefore, we propose that shorter fasting time (i.e., 4 hours) should be adequate for most toxicology studies except when: (1) parameters that could be affected by non-fasting conditions are important for safety and pharmacodynamic assessments (i.e., glucose and lipids) and (2) fasting would be needed for the bioavailability of an orally administered test article.


Asunto(s)
Bienestar del Animal , Ayuno , Animales , Macaca fascicularis
4.
Animals (Basel) ; 13(16)2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37627357

RESUMEN

Challenges and issues related to the use of pentobarbital euthanasia and disposal of animal remains within the US have recently been reviewed. Environmental and public health challenges increasingly necessitate consideration of alternative methods such as gunshots, an American Veterinary Medical Association (AVMA) "acceptable with conditions" method, for the humane euthanasia of horses. A recent study reported a correctly aimed gunshot provides a humane option for euthanizing horses. However, although aiming guidelines exist, studies examining bullet trajectories in animals euthanized by gunshot have reported that inadequate disruption of the brain is a serious welfare issue. Here, we report the development and production of a portable, reusable, equine gunshot euthanasia training model. Using 3D printing, an anatomically accurate model of an equine head has been developed, with external aiming landmarks and equipped with integrated laser sensors and LED eyes. The laser sensors are embedded in two specific anatomical tracts (pons and medulla) with aiming paths associated with the aiming landmarks to train correct aiming angle. The LED eyes are linked to the laser sensors to provide instant feedback on aiming accuracy. When a beam from a commercially available blue training gun laser travels along the correct aiming path and strikes the sensor inside the head, the lights in the model's eyes go out and there is an audible signal, providing immediate feedback on the accuracy of the shot. The model facilitates the training of veterinary personnel and first responders in successful gunshot euthanasia, providing instantaneous feedback on the likelihood of a shot causing immediate, humane death in a live animal.

5.
J Pharmacol Toxicol Methods ; 121: 107266, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36963703

RESUMEN

INTRODUCTION: Characterization of the incidence of spontaneous arrhythmias to identify possible drug-related effects is often an important part of the analysis in safety pharmacology studies using telemetry. METHODS: A retrospective analysis in non-clinical species with and without telemetry transmitters was conducted. Electrocardiograms (24 h) from male and female beagle dogs (n = 131), Göttingen minipigs (n = 108) and cynomolgus non-human primates (NHP; n = 78) were analyzed. RESULTS: Ventricular tachycardia (VT) was observed in 3% of the dogs but was absent in minipigs and NHPs. Ventricular fibrillation (VF) was not observed in the 3 species. Ventricular premature beats (VPBs) were more frequent during daytime and atrioventricular blocks (AVBs) were more frequent at night in all species. A limited number of animals exhibited a high arrhythmia frequency and there was no correlation between animals with higher frequency of an arrhythmia type and the frequency of other arrythmias in the same animals. Clinical chemistry or hematology parameters were not different with or without telemetry devices. NHP with a transmural left ventricular pressure (LVP) catheter exhibited a greater incidence of VPBs and PJCs compared to telemetry animals without LVP. DISCUSSION: All species were similar with regards to the frequency of ventricular ectopic beats (26-46%) while the dog seemed to have more frequent junctional complexes and AVB compared to NHP and minipigs. Arrhythmia screening may be considered during pre-study evaluations, to exclude animals with abnormally high arrhythmia incidence.


Asunto(s)
Arritmias Cardíacas , Telemetría , Animales , Perros , Porcinos , Masculino , Femenino , Porcinos Enanos , Incidencia , Estudios Retrospectivos , Electrocardiografía
6.
Regul Toxicol Pharmacol ; 138: 105339, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36649820

RESUMEN

Assessment of reversibility from nonclinical toxicity findings in animals with potential adverse clinical impact is required during pharmaceutical development, but there is flexibility around how and when this is performed and if recovery animals are necessary. For monoclonal antibodies (mAbs) and in accordance with ICH S6(R1) if inclusion of recovery animals is warranted, this need only occur in one study. Data on study designs for first-in-human (FIH)-enabling and later-development toxicity studies were shared from a recent collaboration between the NC3Rs, EPAA, Netherlands Medicines Evaluation Board (MEB) and 14 pharmaceutical companies. This enabled a review of practices on recovery animal use during mAb development and identification of opportunities to reduce research animal use. Recovery animals were included in 68% of FIH-enabling and 69% of later-development studies, often in multiple studies in the same program. Recovery groups were commonly in control plus one test article-dosed group or in all dose groups (45% of studies, each design). Based on the shared data review and conclusions, limiting inclusion of recovery to a single nonclinical toxicology study and species, study design optimisation and use of existing knowledge instead of additional recovery groups provide opportunities to further reduce animal use within mAb development programs.


Asunto(s)
Anticuerpos Monoclonales , Proyectos de Investigación , Animales , Humanos , Anticuerpos Monoclonales/efectos adversos , Evaluación Preclínica de Medicamentos , Desarrollo de Medicamentos , Grupos Control
7.
Regul Toxicol Pharmacol ; 138: 105329, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36592682

RESUMEN

To support registration of monoclonal antibodies (mAbs) for chronic indications, 6-month toxicity studies have historically been conducted. Experience with mAb development has shown a relatively benign and well-understood safety profile for this class, with most toxicity findings anticipated based on pharmacology. We evaluated whether a 6-month toxicity study is necessary to assess the long-term safety of mAbs. Data on First-in-Human (FIH)-enabling and chronic toxicity studies were shared for 142 mAbs submitted by 11 companies. Opportunities to further optimize study designs to reduce animal usage were identified. For 71% of mAbs, no toxicities or no new toxicities were noted in chronic studies compared to FIH-enabling study findings. New toxicities of potential concern for human safety or that changed trial design were identified in 13.5% of cases, with 7% being considered critical and 2% leading to program termination. An iterative, weight-of-evidence model which considers factors that influence the overall risk for a mAb to cause toxicity was developed. This model enables an evidence-based justification, suggesting when 3-month toxicity studies are likely sufficient to support late-stage clinical development and registration for some mAbs.


Asunto(s)
Anticuerpos Monoclonales , Proyectos de Investigación , Animales , Humanos , Anticuerpos Monoclonales/toxicidad
8.
Toxicol Pathol ; 51(6): 390-396, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-38293937

RESUMEN

In the last decade, numerous initiatives have emerged worldwide to reduce the use of animals in drug development, including more recently the introduction of Virtual Control Groups (VCGs) concept for nonclinical toxicity studies. Although replacement of concurrent controls (CCs) by virtual controls (VCs) represents an exciting opportunity, there are associated challenges that will be discussed in this paper with a more specific focus on anatomic pathology. Coordinated efforts will be needed from toxicologists, clinical and anatomic pathologists, and regulators to support approaches that will facilitate a staggered implementation of VCGs in nonclinical toxicity studies. Notably, the authors believe that a validated database for VC animals will need to include histopathology (digital) slides for microscopic assessment. Ultimately, the most important step lies in the validation of the concept by performing VCG and the full control group in parallel for studies of varying duration over a reasonable timespan to confirm there are no differences in outcomes (dual study design). The authors also discuss a hybrid approach, whereby control groups comprised both concurrent and VCs to demonstrate proof-of-concept. Once confidence is established by sponsors and regulators, VCs have the potential to replace some or all CC animals.


Asunto(s)
Desarrollo de Medicamentos , Patología , Animales , Grupos Control , Proyectos de Investigación
9.
Breast Cancer Res Treat ; 191(2): 303-317, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34708303

RESUMEN

PURPOSE: Assessment of non-clinical safety signals relies on understanding species selectivity of antibodies. This is particularly important with antibody-drug conjugates, where it is key to determine target-dependent versus target-independent toxicity. Although it appears to be widely accepted that trastuzumab does not bind mouse or rat HER2/ErbB2/neu, numerous investigators continue to use mouse models to investigate safety signals of trastuzumab and trastuzumab emtansine (T-DM1). We, therefore, conducted a broad array of both binding and biologic studies to demonstrate selectivity of trastuzumab for human HER2 versus mouse/rat neu. METHODS: Binding of anti-neu and anti-HER2 antibodies was assessed by ELISA, FACS, IHC, Scatchard, and immunoblot methods in human, rat, and mouse cell lines. In human hepatocytes, T-DM1 uptake and catabolism were measured by LC-MS/MS; cell viability changes were determined using CellTiter-Glo. RESULTS: Our data demonstrate, using different binding methods, lack of trastuzumab binding to rat or mouse neu. Structural studies show important amino acid differences in the trastuzumab-HER2 binding interface between mouse/rat and human HER2 ECD. Substitution of these rodent amino acid residues into human HER2 abolish binding of trastuzumab. Cell viability changes, uptake, and catabolism of T-DM1 versus a DM1 non-targeted control ADC were comparable, indicating target-independent effects of the DM1-containing ADCs. Moreover, trastuzumab binding to human or mouse hepatocytes was not detected. CONCLUSIONS: These data, in total, demonstrate that trastuzumab, and by extension T-DM1, do not bind rat or mouse neu, underscoring the importance of species selection for safety studies investigating trastuzumab or trastuzumab-based therapeutics.


Asunto(s)
Neoplasias de la Mama , Maitansina , Animales , Anticuerpos Monoclonales Humanizados , Cromatografía Liquida , Femenino , Humanos , Maitansina/efectos adversos , Ratones , Ratas , Receptor ErbB-2/genética , Espectrometría de Masas en Tándem , Trastuzumab/efectos adversos
10.
Mol Cancer Ther ; 20(11): 2177-2188, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34433660

RESUMEN

BRAF-targeted therapies including vemurafenib (Zelboraf) induce dramatic cancer remission; however, drug resistance commonly emerges. The purpose was to characterize a naturally occurring canine cancer model harboring complex features of human cancer, to complement experimental models to improve BRAF-targeted therapy. A phase I/II clinical trial of vemurafenib was performed in pet dogs with naturally occurring invasive urothelial carcinoma (InvUC) harboring the canine homologue of human BRAF V600E The safety, MTD, pharmacokinetics, and antitumor activity were determined. Changes in signaling and immune gene expression were assessed by RNA sequencing and phosphoproteomic analyses of cystoscopic biopsies obtained before and during treatment, and at progression. The vemurafenib MTD was 37.5 mg/kg twice daily. Anorexia was the most common adverse event. At the MTD, partial remission occurred in 9 of 24 dogs (38%), with a median progression-free interval of 181 days (range, 53-608 days). In 18% of the dogs, new cutaneous squamous cell carcinoma and papillomas occurred, a known pharmacodynamic effect of vemurafenib in humans. Upregulation of genes in the classical and alternative MAPK-related pathways occurred in subsets of dogs at cancer progression. The most consistent transcriptomic changes were the increase in patterns of T lymphocyte infiltration during the first month of vemurafenib, and of immune failure accompanying cancer progression. In conclusion, the safety, antitumor activity, and cutaneous pharmacodynamic effects of vemurafenib, and the development of drug resistance in dogs closely mimic those reported in humans. This suggests BRAF-mutated canine InvUC offers an important complementary animal model to improve BRAF-targeted therapies in humans.


Asunto(s)
Carcinoma de Células Transicionales/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/efectos de los fármacos , Vemurafenib/uso terapéutico , Adolescente , Animales , Carcinoma de Células Transicionales/patología , Niño , Modelos Animales de Enfermedad , Perros , Humanos , Mutación , Vemurafenib/farmacología
11.
JCI Insight ; 5(7)2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32271166

RESUMEN

Systemic cytokine release and on-target/off-tumor toxicity to normal tissues are the main adverse effects limiting the clinical utility of T cell-redirecting therapies. This study was designed to determine how binding affinity for CD3 and tumor target HER2 impact the efficacy and nonclinical safety of anti-HER2/CD3 T cell-dependent antibodies (TDBs). Affinity was found to be a major determinant for the overall tolerability. Higher affinity for CD3 associated with rapidly elevated peripheral cytokine concentrations, weight loss in mice, and poor tolerability in cynomolgus monkeys. A TDB with lower CD3 affinity was better tolerated in cynomolgus monkeys compared with a higher CD3-affinity TDB. In contrast to tolerability, T cell binding affinity had only limited impact on in vitro and in vivo antitumor activity. High affinity for HER2 was critical for the tumor-killing activity of anti-HER2/CD3 TDBs, but higher HER2 affinity also associated with a more severe toxicity profile, including cytokine release and damage to HER2-expressing tissues. The tolerability of the anti-HER2/CD3 was improved by implementing a dose-fractionation strategy. Fine-tuning the affinities for both the tumor target and CD3 is likely a valuable strategy for achieving maximal therapeutic index of CD3 bispecific antibodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Afinidad de Anticuerpos , Antineoplásicos Inmunológicos/inmunología , Receptor ErbB-2/inmunología , Animales , Anticuerpos Biespecíficos/química , Antineoplásicos Inmunológicos/química , Complejo CD3/química , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Macaca fascicularis , Receptor ErbB-2/química
12.
Toxicol Pathol ; 48(2): 350-361, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31594487

RESUMEN

As ovarian toxicity is often a safety concern for cancer therapeutics, identification of ovarian pathology is important in early stages of preclinical drug development, particularly when the intended patient population include women of child-bearing potential. Microscopic evaluation by pathologists of hematoxylin and eosin (H&E)-stained tissues is the current gold standard for the assessment of organs in toxicity studies. However, digital pathology and advanced image analysis are being explored with greater frequency and broader applicability to tissue evaluations in toxicologic pathology. Our objective in this work was to develop an automated method that rapidly enumerates rat ovarian corpora lutea on standard H&E-stained slides with comparable accuracy to the gold standard assessment by a pathologist. Herein, we describe an algorithm generated by a deep learning network and tested on 5 rat toxicity studies, which included studies that both had and had not previously been diagnosed with effects on number of ovarian corpora lutea. Our algorithm could not only enumerate corpora lutea accurately in all studies but also revealed distinct trends for studies with and without reproductive toxicity. Our method could be a widely applied tool to aid analysis in general toxicity studies.


Asunto(s)
Cuerpo Lúteo/efectos de los fármacos , Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos , Animales , Femenino , Ratas , Ratas Sprague-Dawley , Estudios Retrospectivos
13.
Toxicol Pathol ; 47(7): 789-798, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31648619

RESUMEN

Pathologists are trained medical professionals with special expertise in diagnostics, research, and pathophysiology. In these roles, pathologists are well qualified and positioned to engage in conversations about animal use replacement, reduction, and refinement (3Rs), thereby championing the guiding principles of the 3Rs. In particular, toxicology or nonclinical safety assessment is an important area where the discipline of toxicologic pathology can have a critical role in adopting 3Rs principles. As such, a working group of the Society of Toxicologic Pathology Scientific and Regulatory Policy Committee was formed to investigate and summarize some of the areas where veterinary pathologists working in the field of toxicology can increase involvement and impact on 3Rs. This "Points to Consider" publication provides an overview of areas within toxicology where the veterinary pathologist's perspective may maximize animal value, including refinement of study design, optimizing sample collection, the development of 3Rs focused regulatory policy, and humane end point determination.[Box: see text].


Asunto(s)
Alternativas al Uso de Animales/legislación & jurisprudencia , Patólogos , Rol del Médico , Toxicología , Bienestar del Animal , Animales , Humanos , Proyectos de Investigación , Manejo de Especímenes
14.
AAPS J ; 20(6): 107, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30298434

RESUMEN

We previously performed a comparative assessment of tissue-level vascular physiological parameters in mice and rats, two of the most commonly utilized species in translational drug development. The present work extends this effort to non-human primates by measuring tissue- and organ-level vascular volumes (Vv), interstitial volumes (Vi), and blood flow rates (Q) in cynomolgus monkeys. These measurements were accomplished by red blood cell labeling, extracellular marker infusion, and rubidium chloride bolus distribution, respectively, the same methods used in previous rodent measurements. In addition, whole-body blood volumes (BV) were determined across species. The results demonstrate that Vv, Vi, and Q, measured using our methods scale approximately by body weight across mouse, rat, and monkey in the tissues considered here, where allometric analysis allowed extrapolation to human parameters. Significant differences were observed between the values determined in this study and those reported in the literature, including Vv in muscle, brain, and skin and Q in muscle, adipose, heart, thymus, and spleen. The impact of these differences for selected tissues was evaluated via sensitivity analysis using a physiologically based pharmacokinetic model. The blood-brain barrier in monkeys was shown to be more impervious to an infused radioactive tracer, indium-111-pentetate, than in mice or rats. The body weight-normalized total BV measured in monkey agreed well with previously measured value in rats but was lower than that in mice. These findings have important implications for the common practice of scaling physiological parameters from rodents to primates in translational pharmacology.


Asunto(s)
Desarrollo de Medicamentos/métodos , Modelos Animales , Investigación Farmacéutica/métodos , Animales , Velocidad del Flujo Sanguíneo/fisiología , Volumen Sanguíneo/fisiología , Barrera Hematoencefálica/metabolismo , Peso Corporal/fisiología , Femenino , Macaca fascicularis/fisiología , Masculino , Ratones/fisiología , Radiofármacos/administración & dosificación , Radiofármacos/farmacocinética , Ratas/fisiología , Especificidad de la Especie , Distribución Tisular
15.
Toxicol Sci ; 165(1): 186-197, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29893934

RESUMEN

Onartuzumab is an engineered single arm, monovalent monoclonal antibody that targets the MET receptor and prevents hepatocyte growth factor (HGF) signaling. Knockout mice have clearly demonstrated that HGF/MET signaling is developmentally critical. A pre- and postnatal development study (enhanced design) was conducted in cynomolgus monkeys to evaluate the potential developmental consequences following onartuzumab administration. Control or onartuzumab, at loading/maintenance doses of 75/50 mg/kg (low) or 100/100 mg/kg (high), was administered intravenously once weekly to 12 confirmed pregnant female cynomolgus monkeys per group from gestation day (GD) 20 through GD 174. Onartuzumab administration resulted in decreased gestation length, decreased birth weight, and increased fetal and perinatal mortality. A GD147 C-section was conducted for a subset of Control and High Dose monkeys, and identified placental infarcts with hemorrhage in the chorionic plate, chorionic villus and/or decidual plate. These findings were limited to placentas from onartuzumab-treated animals. In addition, decreased cellularity of the hepatocytes with dilated hepatic sinusoids was inconsistently observed in the liver of a few fetal or infant monkeys that died in the perinatal period. Surviving offspring had some evidence of developmental delay compared with controls, but no overt teratogenicity. Overall, effects on the perinatal fetuses were consistent with those reported in knockout mice, but not as severe. Onartuzumab concentrations were low or below the level of detection in most offspring, with cord blood concentrations only 1%-2% of maternal levels on GD 147. Malperfusion secondary to onartuzumab-induced placental injury could explain the adverse pregnancy outcomes, fetal growth restriction and relatively low fetal exposures.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Desarrollo Fetal/efectos de los fármacos , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Exposición Materna/efectos adversos , Placenta/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/sangre , Femenino , Retardo del Crecimiento Fetal/tratamiento farmacológico , Macaca fascicularis , Placenta/metabolismo , Embarazo , Resultado del Embarazo , Transducción de Señal
16.
Regul Toxicol Pharmacol ; 92: 382-389, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29278695

RESUMEN

Vismodegib (also known as GDC-0449) is a novel small molecule inhibitor of the Hedgehog (Hh) signaling pathway currently approved for the treatment of metastatic or locally advanced basal cell carcinoma (BCC) in humans. Its tumorigenic potential was assessed in dedicated carcinogenicity studies in rasH2 transgenic (Tg.rasH2) mice and Sprague Dawley (SD) rats. Tumorigenicity potential of vismodegib was identified in rats only and was limited to benign hair follicle tumors, including pilomatricomas and keratoacanthomas at exposures of ≥0.1-fold and ≥0.6-fold, respectively, of the steady-state exposure (AUC0-24h) of the recommended human dose. No malignant tumors were identified in either species. Overall, the totality of pharmacology and nonclinical safety data (lack of genotoxicity, in vitro secondary pharmacological binding, and immunoregulatory effects, and limited effects on the endocrine system) suggests that the development of the benign hair follicle tumors may be related to pharmacologically-mediated disruption of hair follicle morphogenesis, although the exact mechanism of tumorigenesis is unclear. Hair follicle tumors have not been reported in vismodegib-treated patients. The relevance of this finding in rats to patients is uncertain.


Asunto(s)
Anilidas/farmacología , Carcinogénesis/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Carcinogénesis/metabolismo , Femenino , Folículo Piloso/efectos de los fármacos , Folículo Piloso/metabolismo , Humanos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo
17.
Toxicol Pathol ; 45(8): 1055-1066, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233079

RESUMEN

To test the diagnostic approach described in part 1 of this article, 2 exercises were completed by pathologists from multiple companies/agencies. Pathologist's examination of whole slide image (WSI) heart sections from rats using personal diagnostic approaches (exercise #1) corroborated conclusions from study #1. Using the diagnostic approach described in part 1, these pathologists examined the same WSI heart sections (exercise #2) to determine whether that approach increased consistency of diagnosis of rodent progressive cardiomyopathy (PCM) lesions. In exercise #2, there was improved consistency of categorization of small borderline morphologies and mild lesions, but a decrement in consistency of categorizing minimal lesions. Exercises 1 and 2 suggest the described diagnostic approach is representative of that in use by the majority of toxicologic pathologists across companies/agencies and that application by all may improve diagnostic consistency of PCM/like lesions. Additionally, a criterion of approximately 5% heart section involvement is suggested for separating mild from moderate or greater severity. While evidence is not absolute, until further investigation shows otherwise, microscopic changes resembling PCM, but located in the epicardial and subepicardial region of the right ventricle, may be considered as part of the spectrum of PCM.


Asunto(s)
Cardiomiopatías/patología , Diagnóstico por Imagen/métodos , Ventrículos Cardíacos/patología , Ratas Sprague-Dawley , Enfermedades de los Roedores/patología , Pruebas de Toxicidad/métodos , Animales , Cardiomiopatías/veterinaria , Cardiotoxicidad/patología , Cardiotoxicidad/veterinaria , Simulación por Computador , Diagnóstico por Imagen/normas , Diagnóstico por Imagen/veterinaria , Progresión de la Enfermedad , Masculino , Pruebas de Toxicidad/veterinaria
18.
Toxicol Pathol ; 45(8): 1043-1054, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29173114

RESUMEN

Spontaneous rodent progressive cardiomyopathy (PCM) in the Sprague Dawley rat may confound identification and/or interpretation of potential test article (TA)-related cardiotoxicity. Pathologists apply diagnostic term(s) and thresholds for diagnosing and assigning severity grades for PCM and/or PCM-like (PCM/like) lesions consistently within a study, which is necessary to identify and interpret TA-related findings. Due to differences in training and/or experiences, diagnostic terms and thresholds may vary between pathologists. Harmonized terminology and thresholds across studies will generate better historical control data, will likely enhance interpretation of study data, and may further enhance our understanding of the spontaneous change. An assessment of the diagnostic approaches of a group of 37 pathologists identified an approach that is relatively easily applied; and if adopted, it could enhance diagnostic consistency across studies. This approach uses the single "slash" term "necrosis/inflammatory cell infiltrate (NICI)" as the diagnosis for the spectrum of lesions seen in younger rats, uses no threshold for diagnosis (e.g., diagnose all lesions clearly identifiable as PCM/like), and uses aggregate lesion size of approximately ≥45% of the field of view (FOV) using a 10×/22 eyepiece and the 40× objective or approximately ≥100% of the FOV using the 60× objective as the criterion separating minimal from mild severities.


Asunto(s)
Cardiomiopatías/patología , Diagnóstico por Imagen/métodos , Ratas Sprague-Dawley , Enfermedades de los Roedores/patología , Pruebas de Toxicidad/veterinaria , Animales , Cardiomiopatías/veterinaria , Cardiotoxicidad/patología , Cardiotoxicidad/veterinaria , Simulación por Computador , Diagnóstico por Imagen/normas , Diagnóstico por Imagen/veterinaria , Progresión de la Enfermedad , Masculino , Necrosis , Índice de Severidad de la Enfermedad
19.
Cell Death Dis ; 7(8): e2338, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27512959

RESUMEN

Apo2L/TRAIL is a member of the tumor necrosis factor superfamily and an important inducer of apoptosis. Recombinant human (rhu) Apo2L/TRAIL has been attractive as a potential cancer therapeutic because many types of tumor cells are sensitive to its apoptosis-inducing effects. Nonclinical toxicology studies were conducted to evaluate the safety of rhuApo2L/TRAIL for possible use in humans. The cynomolgus monkey was chosen for this safety assessment based on high protein sequence homology between human and cynomolgus Apo2L/TRAIL and comparable expression of their receptors. Although hepatotoxicity was observed in repeat-dose monkey studies with rhuApo2L/TRAIL, all animals that displayed hepatotoxicity had developed antitherapeutic antibodies (ATAs). The cynomolgus ATAs augmented the cytotoxicity of rhuApo2L/TRAIL but not of its cynomolgus counterpart. Of note, human and cynomolgus Apo2L/TRAIL differ by four amino acids, three of which are surface-exposed. In vivo studies comparing human and cynomolgus Apo2L/TRAIL supported the conclusion that these distinct amino acids served as epitopes for cross-species ATAs, capable of crosslinking rhuApo2L/TRAIL and thus triggering hepatocyte apoptosis. We describe a hapten-independent mechanism of immune-mediated, drug-related hepatotoxicity - in this case - associated with the administration of a human recombinant protein in monkeys. The elucidation of this mechanism enabled successful transition of rhuApo2L/TRAIL into human clinical trials.


Asunto(s)
Anticuerpos/toxicidad , Anticuerpos/uso terapéutico , Proteínas Recombinantes/toxicidad , Proteínas Recombinantes/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/toxicidad , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Modelos Animales de Enfermedad , Humanos , Células Jurkat , Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Macaca fascicularis , Especificidad de la Especie
20.
Artículo en Inglés | MEDLINE | ID: mdl-27039257

RESUMEN

INTRODUCTION: The Safety Pharmacology Society (SPS) and National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs) conducted a survey and workshop in 2015 to define current industry practices relating to housing of non-rodents during telemetry recordings in safety pharmacology and toxicology studies. The aim was to share experiences, canvas opinion on the study procedures/designs that could be used and explore the barriers to social housing. METHODS: Thirty-nine sites, either running studies (Sponsors or Contract Research Organisations, CROs) and/or outsourcing work responded to the survey (51% from Europe; 41% from USA). RESULTS: During safety pharmacology studies, 84, 67 and 100% of respondents socially house dogs, minipigs and non-human primates (NHPs) respectively on non-recording days. However, on recording days 20, 20 and 33% of respondents socially house the animals, respectively. The main barriers for social housing were limitations in the recording equipment used, study design and animal temperament/activity. During toxicology studies, 94, 100 and 100% of respondents socially house dogs, minipigs and NHPs respectively on non-recording days. However, on recording days 31, 25 and 50% of respondents socially house the animals, respectively. The main barriers for social housing were risk of damage to and limitations in the recording equipment used, food consumption recording and temperament/activity of the animals. CONCLUSIONS: Although the majority of the industry does not yet socially house animals during telemetry recordings in safety pharmacology and toxicology studies, there is support to implement this refinement. Continued discussions, sharing of best practice and data from companies already socially housing, combined with technology improvements and investments in infrastructure are required to maintain the forward momentum of this refinement across the industry.


Asunto(s)
Hemodinámica/efectos de los fármacos , Vivienda para Animales , Medio Social , Animales , Perros , Evaluación Preclínica de Medicamentos , Farmacología/métodos , Primates , Seguridad , Encuestas y Cuestionarios , Porcinos , Porcinos Enanos , Telemetría , Temperamento , Toxicología/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA