Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Toxicol ; 6: 1377990, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38845817

RESUMO

Industry representatives on the ICH S1B(R1) Expert Working Group (EWG) worked closely with colleagues from the Drug Regulatory Authorities to develop an addendum to the ICH S1B guideline on carcinogenicity studies that allows for a weight-of-evidence (WoE) carcinogenicity assessment in some cases, rather than conducting a 2-year rat carcinogenicity study. A subgroup of the EWG composed of regulators have published in this issue a detailed analysis of the Prospective Evaluation Study (PES) conducted under the auspices of the ICH S1B(R1) EWG. Based on the experience gained through the Prospective Evaluation Study (PES) process, industry members of the EWG have prepared the following commentary to aid sponsors in assessing the standard WoE factors, considering how novel investigative approaches may be used to support a WoE assessment, and preparing appropriate documentation of the WoE assessment for presentation to regulatory authorities. The commentary also reviews some of the implementation challenges sponsors must consider in developing a carcinogenicity assessment strategy. Finally, case examples drawn from previously marketed products are provided as a supplement to this commentary to provide additional examples of how WoE criteria may be applied. The information and opinions expressed in this commentary are aimed at increasing the quality of WoE assessments to ensure the successful implementation of this approach.

2.
Toxicol Sci ; 183(1): 93-104, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34240189

RESUMO

BMS-986251 is a retinoid-related orphan receptor γt (RORγt) inverse agonist that was in development for the treatment of autoimmune diseases. RORγt is a nuclear hormone receptor and transcription factor that is involved in the differentiation and function of T helper 17 cells. RORγt-deficient (constitutive or conditional) mice develop thymic lymphomas with >50% mortality at 4 months, whereas heterozygous mice are normal. A 6-month study was conducted in rasH2-Tg hemizygous mice to assess the potential carcinogenicity of BMS-986251. BMS-986251 was administered once daily by oral gavage to groups of 27 mice/sex at doses of 0 (water control), 0 (vehicle control), 5, 25, or 75 mg/kg. The positive control, N-methyl-N-nitrosourea, was administered by a single intraperitoneal injection to 15 mice/sex at a dose of 75 mg/kg. There were no tumors attributed to BMS-986251 except for thymic lymphomas. Thymic lymphoma was observed in 1 male (3.7%) and 3 females (11.1%) at the mid dose, and 6 females (22.2%) at the high dose. No lymphomas were observed in the negative control groups whereas the incidence of lymphomas in the positive control group was 47-60%. The incidence of thymic lymphomas in the BMS-986251-treated groups was higher than published literature and test facility historical control data. Furthermore, increased thymic lymphoid cellularity (lymphoid hyperplasia) was observed at the mid dose in males and at all doses in females. Since lymphoid hyperplasia may represent a preneoplastic change, a no-effect dose for potential tumor induction was not identified in this study. These results led to the discontinuation of BMS-986251 and underscore the challenges in targeting RORγt for drug development.


Assuntos
Linfoma , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Animais , Testes de Carcinogenicidade , Feminino , Hiperplasia , Linfoma/induzido quimicamente , Linfoma/genética , Masculino , Camundongos , Camundongos Transgênicos
3.
Toxicol Appl Pharmacol ; 409: 115285, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33069749

RESUMO

The OX40 receptor plays a crucial co-stimulatory role in T effector cell survival, expansion, cytokine production, and cytotoxicity to tumor cells; therefore, OX40 agonists are being evaluated as anti-cancer immunotherapies, especially in combination with checkpoint inhibitors. To support clinical development of BMS-986178 (an OX40 agonist antibody), two repeat-dose toxicity studies were conducted in cynomolgus monkeys. In the first study, BMS-986178 was administered intravenously (IV) once weekly for one month at doses from 30 to 120 mg/kg. BMS-986178 was well tolerated; surprisingly, immune function was suppressed rather than increased based on pharmacodynamic (PD) and flow cytometry readouts (e.g. T-cell dependent antibody response [TDAR]). To determine whether immune suppression was due to a bi-phasic response, a follow-up study was conducted at lower doses (1 and 10 mg/kg). Although receptor engagement was confirmed, immune function was still suppressed at both doses. In addition, treatment-emergent anti-drug antibodies (ADAs) at 1 mg/kg resulted in hypersensitivity reactions and reduced BMS-986178 exposure after repeated dosing, which precluded a full PD assessment at this dose. In conclusion, BMS-986178 was clinically well-tolerated by monkeys at weekly IV doses from 10 to 120 mg/kg (AUC[0-168] ≤ 712,000 µg●h/mL). However, despite target engagement, PD assays and other immune endpoints demonstrated immune suppression, not stimulation. Due to the inverted immune response at higher doses and the onset of ADAs, additional repeat-dose toxicity studies of BMS-986178 in monkeys (that would typically be required to support Phase 3 clinical trials and registration) would not add value for human safety assessment.


Assuntos
Anticorpos Monoclonais/imunologia , Imunidade/imunologia , Receptores OX40/imunologia , Linfócitos T/imunologia , Animais , Feminino , Seguimentos , Humanos , Imunoterapia/métodos , Macaca fascicularis , Masculino
4.
Toxicol Lett ; 317: 120-129, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31580884

RESUMO

PEGylation is considered a safe mechanism to enhance the pharmacokinetics (PK) and pharmacodynamics (PD) of biotherapeutics. Previous studies using PEGylation as a PK enhancement tool have reported benign PEG-related vacuolation in multiple tissues. This paper establishes a threshold for PEG burden beyond which there are alterations in tissue architecture that could potentially lead to dysfunction. As part of the nonclinical safety assessment of Compound A, a 12 kDa protein conjugated to a 40 kDa branched PEG molecule, monkeys were dosed subcutaneously twice weekly for 3 months at protein doses resulting in weekly PEG doses of 8, 24, 120, or 160 mg/kg. Consistent with previous reports with PEGylated biomolecules, Compound A administration resulted in intracellular vacuoles attributed to the PEG moiety in macrophages in numerous tissues and epithelial cells in the choroid plexus and kidney. Vacuolation occurred at all doses with dose-dependent severity and no evidence of recovery up to 2 months after dosing cessation. The vacuolation was considered nonadverse at PEG doses ≤120 mg/kg/week. However, at 160 mg/kg/week PEG, the vacuolation in choroid plexus, pituitary gland, kidney, and choroid of the eye was considered adverse due to significant alterations of tissue architecture that raised concern for the possibility of compromised tissue function. To our knowledge, this is the first report of potentially adverse cellular consequences of PEG accumulation in tissues other than kidney. Furthermore, the lack of reversibility of vacuolation coupled with the lack of a biomarker for intracellular PEG accumulation highlights a potential risk that should be weighed against the benefits of PK/PD enhancement for long-term administration of PEGylated compounds at high doses.


Assuntos
Células Epiteliais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Polietilenoglicóis/toxicidade , Proteínas/toxicidade , Vacúolos/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Esquema de Medicação , Composição de Medicamentos , Células Epiteliais/patologia , Feminino , Injeções Subcutâneas , Macaca fascicularis , Macrófagos/patologia , Masculino , Polietilenoglicóis/administração & dosagem , Proteínas/administração & dosagem , Medição de Risco , Fatores de Tempo , Vacúolos/patologia
5.
Birth Defects Res ; 111(16): 1178-1191, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31228335

RESUMO

Literature suggests that murine allogeneic pregnancy models are an alternative approach for evaluating the developmental toxicity of immune-stimulating agents. In this study, multiple syngeneic and allogeneic murine pregnancy models were used to assess the potential embryo-fetal effects of four different murine antibodies (IgG1 or IgG2 ) that activate the immune system by binding to T-cell receptors (PD-L1, LAG-3, and GITR). The pregnancy models were generated by within and between matings of five different inbred strains of mice (CBA/CaJ, DBA/2J, BALB/c, C57BL/6, and CBA/J). The antibodies were administered every 2-3 days by intraperitoneal injection (n = 12-29/group) during gestation days 6 to 14. There were no differences in embryo-fetal endpoints between the allogeneic and syngeneic pregnancies. Additionally, treatment with the antibodies had no effect on mean postimplantation loss in either the syngeneic or allogeneic pregnancies despite confirmation of pharmacologically-relevant systemic exposures. These results suggest that allogeneic murine pregnancy models need further validation and testing before they can be reliably used as an alternative approach for assessing the developmental effects of agents that stimulate the immune system.


Assuntos
Imunoglobulina G/imunologia , Linfocinas/imunologia , Camundongos Endogâmicos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/imunologia , Antígeno B7-H1/imunologia , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Modelos Animais , Gravidez , Reprodutibilidade dos Testes , Proteína do Gene 3 de Ativação de Linfócitos
6.
Toxicol Sci ; 171(1): 98-116, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31165171

RESUMO

Avagacestat, a gamma (γ)-secretase inhibitor that was in development for treatment of Alzheimer's disease, produced ovarian granulosa-thecal cell tumors in rats and dogs and a glomerulopathy with profound proteinuria in female rats. This report describes the results of follow-up investigative studies, including the use of ovariectomized (OVX) rats, to further characterize these findings and determine their mechanism(s). Ovarian proliferative changes in rats likely resulted from: (1) inhibition of Notch signaling pathways regulating ovarian follicular differentiation/development, characterized microscopically as altered ovarian cyclicity and/or ovarian follicular degeneration; (2) subsequent disruption of the hypothalamic-pituitary-ovarian axis due to ovarian atrophy with decreases in serum estrogen and progesterone (as low as 0.45× and 0.21× controls, respectively); and (3) chronic gonadotropin stimulation and pituitary hypertrophy/hyperplasia in response to the absence of negative feedback. Gonadotropin stimulation in rats was confirmed by increases in serum follicle-stimulating hormone (up to 7.75× controls) and luteinizing hormone (up to 5.84×). A similar nongenotoxic mechanism was likely responsible for the ovarian findings in dogs although changes in serum hormone levels were not detected. The dose- and time-dependent glomerulopathy with progression to chronic progressive nephropathy in female rats appears to be a direct effect of avagacestat and was not ameliorated with coadministration of 17ß-estradiol or an antihypertensive (enalapril) and was not present in control OVX rats. In contrast, adrenocortical hypertrophy in female rats was considered secondary to ovarian changes based on the absence of this finding in avagacestat-treated OVX rats and no increase in adrenocorticotropic hormone staining in the pituitary.

7.
Int J Toxicol ; 38(2): 110-120, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30760067

RESUMO

Pexacerfont is a corticotropin-releasing factor subtype 1 receptor antagonist that was developed for the treatment of anxiety- and stress-related disorders. This report describes the results of repeat-dose oral toxicity studies in rats (3 and 6 months) and dogs (3 months and 1 year). Pexacerfont was well tolerated in all of these studies at exposures equal to or greater than areas under the curve in humans (clinical dose of 100 mg). Microscopic changes in the liver (hepatocellular hypertrophy), thyroid glands (hypertrophy/hyperplasia and adenomas of follicular cells), and pituitary (hypertrophy/hyperplasia and vacuolation of thyrotrophs) were only observed in rats and were considered adaptive changes in response to hepatic enzyme induction and subsequent alterations in serum thyroid hormone levels. Evidence for hepatic enzyme induction in dogs was limited to increased liver weights and reduced thyroxine (T4) levels. Mammary gland hyperplasia and altered female estrous cycling were only observed in rats, whereas adverse testicular effects (consistent with minimal to moderate degeneration of the germinal epithelium) were only noted following chronic dosing in dogs. The testicular effects were reversible changes with exposure margins of 8× at the no observed adverse effect level. It is not clear whether the changes in mammary gland, estrous cycling, and testes represent secondary hormonal changes due to perturbation of the hypothalamic-pituitary-adrenal axis or are off-target effects. In conclusion, the results of chronic toxicity studies in rats and dogs show that pexacerfont has an acceptable safety profile to support further clinical testing.


Assuntos
Pirazóis/toxicidade , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Triazinas/toxicidade , Administração Oral , Animais , Cães , Feminino , Masculino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Pirazóis/farmacocinética , Ratos Sprague-Dawley , Testículo/efeitos dos fármacos , Testículo/patologia , Testes de Toxicidade Crônica , Testes de Toxicidade Subcrônica , Triazinas/farmacocinética
8.
Regul Toxicol Pharmacol ; 100: 35-44, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30291877

RESUMO

Protein therapeutics represent a rapidly growing proportion of new medicines being developed by the pharmaceutical industry. As with any new drug, an Occupational Exposure Limit (OEL) should be developed to ensure worker safety. Part of the OEL determination addresses bioavailability (BA) after inhalation, which is poorly understood for protein therapeutics. To explore this, male Sprague-Dawley rats were exposed intravenously or by nose-only inhalation to one of five test proteins of varying molecular size (10-150 kDa), including a polyethylene glycol-conjugated protein. Blood, lung tissue and bronchoalveolar lavage (BAL) fluid were collected over various time-points depending on the expected test protein clearance (8 minutes-56 days), and analyzed to determine the pharmacokinetic profiles. Since the BAL half-life of the test proteins was observed to be > 4.5 h after an inhalation exposure, accumulation and direct lung effects should be considered in the hazard assessment for protein therapeutics with lung-specific targets. The key finding was the low systemic bioavailability after inhalation exposure for all test proteins (∼≤1%) which did not appear molecular weight-dependent. Given that this study examined the inhalation of typical protein therapeutics in a manner mimicking worker exposure, a default 1% BA assumption is reasonable to utilize when calculating OELs for protein therapeutics.


Assuntos
Polietilenoglicóis/farmacocinética , Proteínas/farmacocinética , Administração por Inalação , Animais , Disponibilidade Biológica , Líquido da Lavagem Broncoalveolar/química , L-Lactato Desidrogenase/metabolismo , Pulmão/metabolismo , Masculino , Concentração Máxima Permitida , Ratos Sprague-Dawley , Receptores Fc/metabolismo
9.
Toxicol Sci ; 163(2): 525-542, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29529279

RESUMO

The toxicity of avagacestat, a sulfonamide-based gamma (γ)-secretase inhibitor that was in development as a treatment for Alzheimer's disease, was evaluated in a comprehensive nonclinical toxicology program that included 6-month and 1-year repeat-dose toxicity studies in rats and dogs, respectively. There was a spectrum of mechanism-based changes attributed to inhibition of Notch signaling that regulates the differentiation and proliferation of cells throughout development and in adult tissues. In both rats and dogs, ovarian follicular degeneration and atrophy and a low incidence of granulosa cell hyperplasia and benign granulosa-thecal cell tumors were observed. Gastrointestinal (GI) findings, including goblet cell metaplasia, dilatation of intestinal crypts/glands, mucosal epithelial necrosis and regeneration, and villous atrophy, were limited to dogs that had clinical evidence of GI toxicity. Other avagacestat-related findings attributed to interference with Notch signaling included decreases in peripheral lymphocytes (T and/or B cells) and lymphoid depletion in lymph nodes and the spleen in both species, as well as epiphyseal cartilage and trabecular bone changes in rats. Pharmacologically mediated decreases in brain and cerebrospinal fluid levels of ß-amyloid (Aß) peptides Aß40 and Aß42 and decreased expression of white blood cell mRNA levels of the Notch-regulated gene hairy and enhancer of split-1 confirmed target engagement at all doses. Reductions in brain Aß peptide levels (22 to 34%) in dogs after 1 year at exposures up to the no-observed-effect level for GI toxicity of 1.1× the human plasma exposure, and reversible GI changes at a 3.2× multiple, indicated that a sustained pharmacodynamic effect was attained at exposures without dose-limiting toxicity.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Trato Gastrointestinal/efeitos dos fármacos , Oxidiazóis/toxicidade , Receptores Notch/antagonistas & inibidores , Sulfonamidas/toxicidade , Administração Oral , Peptídeos beta-Amiloides/metabolismo , Animais , Área Sob a Curva , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Contagem de Linfócitos , Masculino , Oxidiazóis/farmacocinética , Ratos Sprague-Dawley , Fatores Sexuais , Sulfonamidas/farmacocinética , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Testes de Toxicidade
10.
Toxicol Pathol ; 46(2): 147-157, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29471778

RESUMO

The purpose of this study was to establish a 2-stage model of urinary bladder carcinogenesis in male Sprague-Dawley rats to identify tumor promoters. In phase 1 of the study, rats ( n = 170) were administered 100 mg/kg of the tumor initiator, N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN), twice weekly by oral gavage (po) for a period of 6 weeks. Phase 2 consisted of dividing rats into 4 groups ( n = 40 per group) and administering one of the following for 26 weeks to identify putative tumor promoters: (1) vehicle po, (2) 25 mg/kg/day rosiglitazone po, (3) 5% dietary sodium l-ascorbate, and (4) 3% dietary uracil. Rats were necropsied after 7.5 months, and urinary bladders were evaluated by histopathology. BBN/vehicle treatments induced the development of urothelial hyperplasia (83%) and papillomas (15%) but no transitional cell carcinomas (TCCs). Rosiglitazone increased the incidence and severity of papillomas (93%) and resulted in TCC in 10% of treated rats. Uracil was the most effective tumor promoter in our study and increased the incidence of papillomas (90%) and TCC (74%). Sodium ascorbate decreased the incidence of urothelial hyperplasia (63%) and did not increase the incidence of urothelial papillomas or TCC. These data confirm the capacity of our 2-stage model to identify urinary bladder tumor promoters.


Assuntos
Ácido Ascórbico/toxicidade , Carcinógenos/farmacologia , Carcinoma de Células de Transição/induzido quimicamente , Rosiglitazona/toxicidade , Uracila/toxicidade , Neoplasias da Bexiga Urinária/induzido quimicamente , Animais , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Bexiga Urinária/efeitos dos fármacos
11.
Cancer Chemother Pharmacol ; 79(4): 711-723, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28283735

RESUMO

PURPOSE: Pulmonary arterial hypertension (PAH) results from occlusion or vasoconstriction of pulmonary vessels, leading to progressive right ventricular failure. Dasatinib, a BCR-ABL1 tyrosine kinase inhibitor (TKI) approved for the treatment of chronic myelogenous leukemia, has been associated with PAH. In contrast, the BCR-ABL1 TKI imatinib has demonstrated anti-vasoproliferative properties and has been investigated as a potential treatment for PAH. Here we describe studies evaluating the effects of dasatinib and imatinib on cardiovascular and pulmonary functions to understand the reported differential consequences of the two TKIs in a clinical setting. METHODS: The direct effects of dasatinib and imatinib were explored in vivo to investigate possible mechanisms of dasatinib-induced PAH. In addition, effects of dasatinib and imatinib on PAH-related mediators were evaluated in vitro. RESULTS: In rats, both TKIs increased plasma nitric oxide (NO), did not induce PAH-related structural or molecular changes in PA or lungs, and did not alter hemodynamic lung function compared with positive controls. Similarly, in the pulmonary artery endothelial cells and smooth muscle cells co-culture model, imatinib and dasatinib increased NO and decreased endothelin-1 protein and mRNA. CONCLUSIONS: The results of these studies indicated that dasatinib did not induce physiological changes or molecular signatures consistent with PAH when compared to positive controls. Instead, dasatinib induced changes consistent with imatinib. Both dasatinib and imatinib induced biochemical and structural changes consistent with a protective effect for PAH. These data suggest that other factors of unclear etiology contributed to the development of PAH in patients treated with dasatinib.


Assuntos
Antineoplásicos/toxicidade , Dasatinibe/toxicidade , Hipertensão Pulmonar/induzido quimicamente , Mesilato de Imatinib/toxicidade , Inibidores de Proteínas Quinases/toxicidade , Animais , Antineoplásicos/farmacocinética , Dasatinibe/farmacocinética , Endotelina-1/sangue , Expressão Gênica/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Hipertensão Pulmonar/fisiopatologia , Mesilato de Imatinib/farmacocinética , Pulmão/patologia , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/sangue , Inibidores de Proteínas Quinases/farmacocinética , Artéria Pulmonar/efeitos dos fármacos , Circulação Pulmonar/efeitos dos fármacos , RNA Mensageiro/sangue , Ratos , Ratos Sprague-Dawley
12.
Toxicol Sci ; 153(2): 396-408, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27466212

RESUMO

BMS-986094, the prodrug of a guanosine nucleotide analogue (2'-C-methylguanosine), was withdrawn from clinical trials due to serious safety issues. Nonclinical investigative studies were conducted as a follow up to evaluate the potential for BMS-986094-related mitochondrial-toxicity. In vitro, BMS-986094 was applied to human hepatoma cells (HepG2 and Huh-7) or cardiomyocytes (hiPSCM) up to 19 days to assess mitochondrial DNA content and specific gene expression. There were no mitochondrial DNA changes at concentrations ≤10 µM. Transcriptional effects, such as reductions in Huh-7 MT-ND1 and MT-ND5 mRNA content and hiPSCM MT-ND1, MT-COXII, and POLRMT protein expression levels, occurred only at cytotoxic concentrations (≥10 µM) suggesting these transcriptional effects were a consequence of the observed toxicity. Additionally, BMS-986094 has a selective weak affinity for inhibition of RNA polymerases as opposed to DNA polymerases. In vivo, BMS-986094 was given orally to cynomolgus monkeys for 3 weeks or 1 month at doses of 15 or 30 mg/kg/day. Samples of heart and kidney were collected for assessment of mitochondrial respiration, mitochondrial DNA content, and levels of high energy substrates. Although pronounced cardiac and renal toxicities were observed in some monkeys at 30 mg/kg/day treated for 3-4 weeks, there were no changes in mitochondrial DNA content or ATP/GTP levels. Collectively, these data suggest that BMS-986094 is not a direct mitochondrial toxicant.


Assuntos
DNA Mitocondrial/efeitos dos fármacos , Guanosina Monofosfato/análogos & derivados , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , DNA Mitocondrial/biossíntese , DNA Mitocondrial/fisiologia , Relação Dose-Resposta a Droga , Feminino , Guanosina Monofosfato/metabolismo , Guanosina Monofosfato/toxicidade , Guanosina Trifosfato/metabolismo , Coração/efeitos dos fármacos , Testes de Função Cardíaca , Humanos , Inosina Monofosfato/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Testes de Função Renal , Macaca fascicularis , Masculino
13.
Regul Toxicol Pharmacol ; 69(2): 234-42, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24768934

RESUMO

Different government agencies operating in the European Union regulate different types of chemical products but all require testing for carcinogenicity to support applications for product marketing and commercialisation. A conference was held in Brussels in 2013 where representatives of the pharmaceutical, animal health, chemical and plant protection industries, together with representatives of regulatory agencies, universities and other stakeholders, met under the auspices of The European Partnership for Alternative Approaches to Animal Testing (EPAA) to discuss the varying requirements for carcinogenicity testing, and how these studies might be refined to improve hazard evaluation and risk assessment while implementing principles of the 3Rs (replacement, refinement and reduction in animal studies). While there are some similarities, the regulatory approaches in pharmaceutical, animal health, chemical and plant protection sectors have varying degrees of flexibility in requirements for carcinogenicity testing, to an extent reflecting concerns over the magnitude and duration of human exposure, either directly as in therapeutic exposure to pharmaceuticals, or indirectly through the ingestion of residues of veterinary drugs or plant protection chemicals. The article discusses these differences and other considerations for modified carcinogenicity testing paradigms on the basis of scientific and 3Rs approaches.


Assuntos
Carcinógenos/toxicidade , Indústria Farmacêutica/legislação & jurisprudência , Regulamentação Governamental , Preparações Farmacêuticas , Animais , Testes de Carcinogenicidade , Carcinógenos/administração & dosagem , Carcinógenos/análise , Europa (Continente) , União Europeia , Humanos , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/análise , Medição de Risco
14.
Diabetes Ther ; 5(1): 73-96, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24474422

RESUMO

INTRODUCTION: Dapagliflozin is a selective inhibitor of the sodium-glucose co-transporter 2 (SGLT2) that increases urinary glucose excretion to reduce hyperglycemia in the treatment of type 2 diabetes mellitus. A robust carcinogenicity risk assessment was undertaken to assess the chronic safety of dapagliflozin and SGLT2 inhibition. METHODS: Genotoxicity potential of dapagliflozin and its metabolites was assessed in silico, in vitro, and in vivo. Dapagliflozin was administered daily by oral gavage to mice, rats, and dogs to evaluate carcinogenicity risks, including the potential for tumor promotion. SGLT2(-/-) mice were observed to evaluate the effects of chronic glucosuria. The effects of dapagliflozin and increased glucose levels on a panel of human bladder transitional cell carcinoma (TCC) cell lines were also evaluated in vitro and in an in vivo xenograft model. RESULTS: Dapagliflozin and its metabolites were not genotoxic. In CD-1 mice and Sprague-Dawley rats treated for up to 2 years at ≥100× human clinical exposures, dapagliflozin showed no differences versus controls for tumor incidence, time to onset for background tumors, or urinary bladder proliferative/preneoplastic lesions. No tumors or preneoplastic lesions were observed in dogs over 1 year at >3,000× the clinical exposure of dapagliflozin or in SGLT2(-/-) mice observed over 15 months. Transcription profiling in Zucker diabetic fatty rats showed that 5-week dapagliflozin treatment did not induce tumor promoter-associated or cell proliferation genes. Increasing concentrations of glucose, dapagliflozin, or its primary metabolite, dapagliflozin 3-O-glucuronide, did not affect in vitro TCC proliferation rates and dapagliflozin did not enhance tumor growth in nude mice heterotopically implanted with human bladder TCC cell lines. CONCLUSION: A multitude of assessments of tumorigenicity risk consistently showed no effects, suggesting that selective SGLT2 inhibition and, specifically, dapagliflozin are predicted to not be associated with increased cancer risk.

15.
Diabetes ; 63(4): 1303-14, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24222349

RESUMO

Glucagon-like peptide 1-based therapies, collectively described as incretins, produce glycemic benefits in the treatment of type 2 diabetes. Recent publications raised concern for a potential increased risk of pancreatitis and pancreatic cancer with incretins based in part on findings from a small number of rodents. However, extensive toxicology assessments in a substantial number of animals dosed up to 2 years at high multiples of human exposure do not support these concerns. We hypothesized that the lesions being attributed to incretins are commonly observed background findings and endeavored to characterize the incidence of spontaneous pancreatic lesions in three rat strains (Sprague-Dawley [S-D] rats, Zucker diabetic fatty [ZDF] rats, and rats expressing human islet amyloid polypeptide [HIP]; n = 36/group) on a normal or high-fat diet over 4 months. Pancreatic findings in all groups included focal exocrine degeneration, atrophy, inflammation, ductular cell proliferation, and/or observations in large pancreatic ducts similar to those described in the literature, with an incidence of exocrine atrophy/inflammation seen in S-D (42-72%), HIP (39%), and ZDF (6%) rats. These data indicate that the pancreatic findings attributed to incretins are common background findings, observed without drug treatment and independent of diet or glycemic status, suggesting a need to exercise caution when interpreting the relevance of some recent reports regarding human safety.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Pâncreas/efeitos dos fármacos , Pancreatopatias/etiologia , Animais , Diabetes Mellitus/fisiopatologia , Dieta Hiperlipídica , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Incretinas/efeitos adversos , Pâncreas/patologia , Pancreatite/etiologia , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Aumento de Peso
16.
Toxicol Sci ; 128(1): 9-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22539615

RESUMO

Pregabalin, (S)-3-(aminomethyl)-5-methylhexanoic acid, binds with high affinity to the α(2)δ subunit of voltage-gated calcium channels and exerts analgesic, anxiolytic, and antiseizure activities. Two-year carcinogenicity studies were completed in B6C3F1 and CD-1 mice and two separate studies in Wistar rats. Doses in mice were 200, 1000, and 5000 mg/kg/day, with systemic exposures (AUC(0-24 h)) up to 31 times the mean exposure in humans, given the maximum recommended clinical dose. In rats, doses were 50, 150, and 450 mg/kg/day in males and 100, 300, and 900 mg/kg/day in females; systemic exposures up to 24 times were achieved in clinical trials. In both strains of mice, pregabalin treatment was associated with an increased incidence of hemangiosarcoma primarily in liver, spleen, and bone marrow. The incidence of hemangiosarcoma was higher in B6C3F1 mice than in CD-1 mice, consistent with its spontaneous incidence. Pregabalin did not increase the incidence of any other tumor type in rats and was not genotoxic, based on an extensive battery of in vivo and in vitro tests in bacterial and mammalian systems. Thus, pregabalin is a single-species, single tumor-type, nongenotoxic mouse carcinogen. Hemangiosarcomas occurring in mice treated with pregabalin were genotypically distinct from hemangiosarcomas induced by genotoxic carcinogens in humans with respect to ras and p53 mutation patterns and were similar to spontaneous tumors. Furthermore, there was a strong association between pregabalin treatment and bone marrow changes in these studies in mice, suggesting a possible link between the effects observed in bone marrow and the increase in tumor incidence in pregabalin-treated mice.


Assuntos
Hemangiossarcoma/induzido quimicamente , Mutagênicos/toxicidade , Ácido gama-Aminobutírico/análogos & derivados , Animais , Testes de Carcinogenicidade , Relação Dose-Resposta a Droga , Genes p53 , Genes ras , Hemangiossarcoma/genética , Incidência , Camundongos , Camundongos Endogâmicos ICR , Testes de Mutagenicidade , Pregabalina , Ratos , Ratos Wistar , Ácido gama-Aminobutírico/toxicidade
18.
Toxicol Pathol ; 36(3): 410-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18467688

RESUMO

CI-1033 (canertinib) is an irreversible inhibitor of the erbB family of transmembrane tyrosine kinase receptors, including the epidermal growth factor (EGF) receptor. Various inhibitors of the EGF receptor, including CI-1033, have resulted in cutaneous toxicity in humans as a common adverse event. In a chronic toxicity study in rats, CI-1033 produced cutaneous lesions with morphologic characteristics similar to that reported in man. Here the authors describe in detail the dermal changes observed, along with other noteworthy findings of that study. Male and female Wistar rats (15/sex/group) were administered CI-1033 for 27 weeks at 2.5, 5, or 10 mg/kg (15, 30, or 60 mg/m(2), respectively) by gavage. Control animals (15/sex) received vehicle alone (aqueous 0.5% methylcellulose) in a dose volume of 5 mL/kg. Six animals/sex/dose were included for toxicokinetic evaluations. Skin lesions were the primary drug-related toxicity and occurred at > or = 2.5 mg/kg in a dose-dependent fashion. The major gross lesions were papules that evolved into crusts and scales that were first observed in weeks 1 and 3, respectively. Alopecia developed in conjunction with the papular eruptions. Skin changes were most pronounced in females, possibly due to higher drug levels. In week 13, CI-1033 plasma AUC(0-24) values were 527 to 1980 ng.h/mL in males and 844 to 2920 ng x h/mL in females at 2.5 to 10 mg/kg. Microscopic changes could be described as 3 patterns that affected the tail and body (haired skin). Pattern 1 consisted of epidermal changes that started as a superficial, perivascular spongiotic dermatitis with evolving epidermal hyperplasia, scale-crusts, and areas of ulceration. Areas of hyperplasia on the tail were often associated with the development of new hair follicles. Pattern 2 was characterized by a suppurative to pyogranulomatous infundibular folliculitis. Pattern 3 consisted of abnormally oriented hair follicles with malformed hair shafts that were associated with a deeper (isthmic) folliculitis; this correlated with alopecia. Elevations in bone marrow myeloid counts correlated with a peripheral leukocytosis, consistent with inflammatory changes in the dermis. In addition, hepatic cholestasis and epithelial atrophy in the gastrointestinal tract and vagina occurred at > or = 2.5 mg/kg. In conclusion, CI-1033 produced cutaneous lesions involving the epidermis and hair follicle, and the morphologic characteristics were similar to that reported in clinical studies with various inhibitors of the EGF receptor. These changes are consistent with pharmacologic inhibition of the EGF receptor in these tissues and demonstrate that the rat can serve as an animal model for investigating the mechanisms for this toxicity.


Assuntos
Receptores ErbB/antagonistas & inibidores , Foliculite/induzido quimicamente , Morfolinas/toxicidade , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Dermatopatias Papuloescamosas/induzido quimicamente , Pele/efeitos dos fármacos , Administração Oral , Alopecia/induzido quimicamente , Alopecia/patologia , Animais , Área Sob a Curva , Relação Dose-Resposta a Droga , Feminino , Foliculite/patologia , Longevidade/efeitos dos fármacos , Masculino , Morfolinas/farmacocinética , Ratos , Ratos Wistar , Pele/patologia , Dermatopatias Papuloescamosas/patologia , Testes de Toxicidade
19.
Cancer Chemother Pharmacol ; 59(5): 671-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16944149

RESUMO

The MEK-mitogen-activated protein kinase (MAPK) signal transduction pathway is involved with numerous cellular processes including cell growth and differentiation. Phosphorylation of MAPK (pMAPK) by MEK results in activation of this pathway. In various solid tumors, the MEK-MAPK pathway is constitutively active; therefore inhibition of this pathway may provide a therapeutic strategy for treating cancer. The objective of this study was to determine the extent and duration of inhibition of pMAPK in selected normal tissues in rats following single oral or intravenous (IV) doses of the novel MEK inhibitor, PD0325901. Male Sprague-Dawley rats (9/group) received either single oral (PO) or IV doses of PD0325901 at 10, 30, or 100 mg/kg (60, 180, and 600 mg/m(2), respectively). Controls received vehicle alone which was aqueous 0.5% hydroxypropylmethyl-cellulose/0.2% Tween 80 for PO dosing and 20% beta-cyclodextran sulfobutyl ether in water (w:v) for IV dosing. Animals (3/group/day) were euthanized on Days 2, 3, and 4, at approximately 24, 48, and 72 h after dosing, respectively. The effects on pMAPK in liver and lung were determined by Western blot analysis and compared with plasma PD0325901 levels. Satellite animals (6/dose/route) received single PO or IV doses and serial blood samples were collected for determination of toxicokinetic parameters of PD0325901 and its major metabolite. In general, systemic exposure to PD0325901 was comparable between routes of administration due to high PO bioavailability (56-109%). Plasma area under the concentration-time curve values of the pharmacologically inactive carboxylic acid metabolite ranged from 18 to 40% of PD0325901. Clinical signs of toxicity occurred at 100 mg/kg PO or IV, indicating the maximum-tolerated dose had been achieved. On Day 2, pMAPK was inhibited 57-95% in liver and 86-99% in lung at all doses, irrespective of route of administration. On Day 3, lung pMAPK remained inhibited 75-91% at all IV doses and by 88% after the 100-mg/kg PO dose. Liver pMAPK remained inhibited 79 and 91% on Day 3 after 100 mg/kg by IV and PO doses, respectively. On Day 4, liver pMAPK was still inhibited 66% after the 100-mg/kg PO dose. The EC(50) and EC(90) plasma drug levels for inhibition of lung pMAPK were calculated to be 20 and 99 ng/ml, respectively. Liver pMAPK levels were inhibited at least 50% at plasma PD0325901 concentrations > or =50 ng/ml. In conclusion, single PO or IV doses of PD0325901 resulted in dose-dependent inhibition of pMAPK in liver and lung. Inhibition of pMAPK in liver was comparable between routes of administration at < or =30 mg/kg, whereas inhibition of pMAPK in lung occurred for a longer duration following IV administration. Measurement of pMAPK in normal tissues served as a means for assessing the pharmacologic activity of PD0325901 and should be included in toxicity studies to evaluate toxicity-pharmacology relationships.


Assuntos
Benzamidas/farmacologia , Benzamidas/toxicidade , Difenilamina/análogos & derivados , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Administração Oral , Animais , Área Sob a Curva , Benzamidas/farmacocinética , Difenilamina/farmacocinética , Difenilamina/farmacologia , Difenilamina/toxicidade , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacocinética , Glucuronídeos/metabolismo , Meia-Vida , Injeções Intravenosas , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Oxirredução , Fosforilação , Ratos , Ratos Sprague-Dawley
20.
Toxicol Pathol ; 33(4): 449-55, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16036862

RESUMO

PD176067 is a reversible and selective inhibitor of fibroblast growth factor receptor tyrosine kinase, and was in preclinical development as an angiogenesis inhibitor for the treatment of solid tumors. A 14-day oral toxicity study of PD176067 in young female rats (7 weeks old) was conducted at doses of 2.5, 5, and 10 mg/kg/day (15, 30, and 60 mg/m(2), respectively). Skeletal changes, and vascular and soft tissue mineralization were observed as primary drug-related toxicities. To determine if these changes are specific to young, rapidly growing animals with increased vascular and osseous development, PD176067 was administered to mature (11 months old) rats. Female rats received PD176067 by gavage for 14 days at doses of 2.5, 5, and 10 mg/kg/day and necropsied on day 15. Clinical signs of toxicity were seen at > or =5 mg/kg and one death occurred at 10 mg/kg. Physeal dysplasia (distal femur, proximal tibia, sternum) occurred in all drug-treated animals and was characterized by dose-related increased thickness of the zones of chondrocyte proliferation and hypertrophy, and marked thickening of the zone of ossification. Cartilage hyperplasia was characterized by proliferation of chondrocytes along margins of the synchondrosis and subperiosteum of sternebrae. Serum phosphorus levels increased 47% and 166% at 5 and 10 mg/kg, respectively. Mineralization of cardiac myocytes, aorta, various arteries, renal tubules, and gastric mucosa and muscularis was seen at 10 mg/kg, and consistent with the presence of calcium-phosphorus deposition. Physeal changes occurred at similar plasma PD176067 exposures in young and mature rats (AUC > or = 4.83 microg.hr/mL). PD176067 produced morphologically similar lesions in young and adult rats.


Assuntos
Inibidores da Angiogênese/toxicidade , Inibidores Enzimáticos/toxicidade , Fator 1 de Crescimento de Fibroblastos/antagonistas & inibidores , Compostos Heterocíclicos com 2 Anéis/farmacologia , Osteocondrodisplasias/induzido quimicamente , Piridinas/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Administração Oral , Fatores Etários , Animais , Calcinose/induzido quimicamente , Calcinose/metabolismo , Calcinose/patologia , Proliferação de Células/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Ratos , Ratos Sprague-Dawley , Testes de Toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA