Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Regul Toxicol Pharmacol ; 107: 104413, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31229519

RESUMO

The draft ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high dose in developmental and reproductive toxicity (DART) studies. In 2016, IQ DruSafe conducted an anonymous survey to identify industry practices and experiences related to pharmacokinetic assessments in DART studies in order to facilitate a pragmatic data-driven approach to development of an acceptable multiple of the clinical exposure to be proposed for dose selection in the guideline. Questions in the survey were designed to explore pharmacokinetic differences in pregnant versus non-pregnant animals, and to assess exposure levels attained in the absence of maternal toxicity as well as DART outcomes in animal studies associated with those exposures. Small molecule and therapeutic proteins were analyzed separately. The key findings for small molecules were: a) differences in exposures between pregnant and non-pregnant animals were generally ≤3-fold, b) Cmax or AUC exposures ≥25-fold the clinical exposure were achieved in the absence of maternal toxicity for 31% and 23% of rat and rabbit developmental toxicity studies, respectively, and c) only 3.3% (5/153) and 1.6% (2/128) of the developmental toxicity studies were positive for malformations or embryofetal lethality in rats and rabbits, respectively, that were not observed until exposure margins were ≥25-fold.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Teratogênicos/farmacocinética , Teratogênicos/toxicidade , Testes de Toxicidade , Animais , Indústria Farmacêutica , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Haplorrinos , Gravidez , Coelhos , Ratos , Reprodução/efeitos dos fármacos , Inquéritos e Questionários
2.
Regul Toxicol Pharmacol ; 105: 62-68, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30981719

RESUMO

The draft Step 2 ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high-dose in reproductive and developmental toxicity studies. To help determine an appropriate exposure margin for embryofetal developmental toxicity testing, a retrospective analysis was undertaken to determine what threshold would have been sufficient to detect hazards to embryofetal development in rats and rabbits for 18 known and 4 presumed human teratogens. The analysis showed that using a high dose that provided at least a 6-fold exposure margin in the developmental toxicity studies would have been sufficient to detect the teratogenic hazard with relevance for humans for all these therapeutics. With regards to human risk assessment practices for developmental toxicity, the analysis showed that, after excluding lenalidomide and pomalidomide data in rats, all available AUC margins at the NOAEL for the induction of malformations or embryofetal lethality were <4-fold of the exposure at the MRHD for all 22 therapeutics. These data support the proposed general approach of increased level of concern for human risk when exposure margins of the NOAEL to the MRHD are <10-fold, reduced concern when the exposure margins are 10- to 25-fold, and minimal concern when the exposure margin is > 25-fold.


Assuntos
Embrião de Mamíferos/efeitos dos fármacos , Medição de Risco/métodos , Teratogênicos/toxicidade , Testes de Toxicidade/métodos , Animais , Área Sob a Curva , Relação Dose-Resposta a Droga , Feminino , Humanos , Nível de Efeito Adverso não Observado , Gravidez , Coelhos , Ratos , Estudos Retrospectivos , Especificidade da Espécie
3.
Regul Toxicol Pharmacol ; 96: 167-177, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29763632

RESUMO

Toxicity studies in juvenile animals (JAS) are sometimes performed to support clinical trials in pediatric oncology patients, and there are differing conclusions on the value of JAS for pediatric drug development. This manuscript provides a review of the pediatric clinical data for 25 molecularly-targeted and 4 biologic anticancer therapeutics. Other publications that evaluated the value of JAS in pediatric drug development focus on differences in toxicity between juvenile animals and adult animals. The present paper examines pediatric-specific clinical findings to focus on dose setting in pediatric oncology patients and safety monitoring in terms of the potential value of JAS. Our assessment demonstrates that pediatric starting doses were safe for all 29 therapeutics examined in that no life-threatening toxicities occurred in the first cohort, and overall the ratio of the pediatric maximum tolerated dose (MTD) to the recommended adult dose was close to 1. In addition, the 4 serious adverse events (SAEs) that weren't detectable with standard monitoring plans for pediatric oncology trials would not have been detectable in a standard JAS. This review demonstrates that safe starting doses in pediatric oncology patients for these therapeutics could have been solely based on adult doses without any knowledge of findings in JAS.


Assuntos
Antineoplásicos/toxicidade , Ensaios Clínicos Fase I como Assunto/métodos , Ensaios Clínicos Fase I como Assunto/normas , Testes de Toxicidade , Animais , Humanos
5.
Regul Toxicol Pharmacol ; 67(3): 382-91, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24012707

RESUMO

Antibody drug conjugates (ADCs) include monoclonal antibodies that are linked to cytotoxic small molecules. A number of these agents are currently being developed as anti-cancer agents designed to improve the therapeutic index of the cytotoxin (i.e., cytotoxic small molecule or cytotoxic agent) by specifically delivering it to tumor cells. This paper presents primary considerations for the nonclinical safety evaluation of ADCs and includes strategies for the evaluation of the entire ADC or the various individual components (i.e., antibody, linker or the cytotoxin). Considerations are presented on how to design a nonclinical safety assessment program to identify the on- and off-target toxicities to enable first-in-human (FIH) studies. Specific discussions are also included that provide details as to the need and how to conduct the studies for evaluating ADCs in genetic toxicology, tissue cross-reactivity, safety pharmacology, carcinogenicity, developmental and reproductive toxicology, biotransformation, toxicokinetic monitoring, bioanalytical assays, immunogenicity testing, test article stability and the selection of the FIH dose. Given the complexity of these molecules and our evolving understanding of their properties, there is no single all-encompassing nonclinical strategy. Instead, each ADC should be evaluated on a case-by-case scientifically-based approach that is consistent with ICH and animal research guidelines.


Assuntos
Anticorpos Monoclonais/toxicidade , Antineoplásicos/toxicidade , Imunoconjugados/toxicidade , Testes de Toxicidade , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Antineoplásicos/química , Antineoplásicos/imunologia , Antineoplásicos/farmacocinética , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Guias como Assunto , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Projetos de Pesquisa , Testes de Toxicidade/métodos , Testes de Toxicidade/normas
6.
Regul Toxicol Pharmacol ; 58(2): 243-51, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20558225

RESUMO

Recent advances in our understanding of disease biology, biomarkers, new therapeutic targets, and innovative modalities have each fueled a dramatic expansion in the development of novel human therapeutics. Many are biotechnology-derived biologics possessing high selectivity and affinity for their intended target; as such they often pose challenges in the development path to approval. One challenge is the selection of the first-in-human (FIH) dose. This process has come under increased scrutiny as a result of a FIH trial with a super-agonist monoclonal antibody (TGN1412), which resulted in significant injury to healthy volunteers. Regulatory agencies have responded with supplemental guidance for the development of novel therapeutics. The intent of this paper is to provide experience-based insight, with relevant examples, for those planning the first administration of novel biopharmaceuticals in humans.


Assuntos
Produtos Biológicos/administração & dosagem , Ensaios Clínicos como Assunto/métodos , Sistemas de Liberação de Medicamentos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Produtos Biológicos/efeitos adversos , Biofarmácia/métodos , Biotecnologia/métodos , Relação Dose-Resposta a Droga , Desenho de Fármacos , Humanos
7.
Int J Toxicol ; 29(1): 3-19, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19903873

RESUMO

Vorinostat (SAHA, Zolinza), a histone deacetylase inhibitor, is assessed in nonclinical studies to support its approval for cutaneous T-cell lymphoma. Vorinostat is weakly mutagenic in the Ames assay; is clastogenic in rodent (ie, CHO) cells but not in normal human lymphocytes; and is weakly positive in an in vivo mouse micronucleus assay. No effects are observed on potassium ion currents in the hERG assay up to 300 microM (safety margin approximately 300-fold the approximately 1 microM serum concentration associated with the 400 mg/d maximum recommended human dose. No rat respiratory or central nervous system effects are found at 150 mg/kg (>2-fold maximum recommended human dose). No cardiovascular effects, including effects on QTc interval, are observed after a single oral dose (150 mg/kg) in dogs. Vorinostat is orally dosed daily in rats (controls, 20, 50, or 150 mg/kg/d) and dogs (controls, 60, 80, or 100/125/160 mg/kg/d) for 26 weeks with a 4-week recovery. Rat vorinostat-related adverse findings are decreased food consumption, weight loss, and hematologic changes; a no observed adverse effects level is not established. In dogs, adverse effects are primarily gastrointestinal; the no observed adverse effects level is 60 mg/kg/d (approximately 6-fold maximum recommended human dose). Toxicities are reversible and can be monitored in the clinic.


Assuntos
Inibidores Enzimáticos/toxicidade , Histona Desacetilases , Ácidos Hidroxâmicos/toxicidade , Animais , Células Sanguíneas/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Células CHO , Cricetinae , Cricetulus , DNA/efeitos dos fármacos , Cães , Avaliação Pré-Clínica de Medicamentos , Ingestão de Alimentos/efeitos dos fármacos , Inibidores Enzimáticos/farmacocinética , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Humanos , Ácidos Hidroxâmicos/farmacocinética , Camundongos , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Testes para Micronúcleos , Ratos , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/patologia , Testes de Toxicidade , Vorinostat , Redução de Peso/efeitos dos fármacos
8.
Drug Metab Lett ; 1(2): 153-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19356036

RESUMO

The disposition of vorinostat, an anticancer agent, was investigated in rats and dogs. Vorinostat possessed high serum clearance, a short elimination half-life and low oral bioavailability in both species. The renal route played an important role in the elimination of drug-related material and vorinostat was eliminated primarily by metabolic biotransformation.


Assuntos
Antineoplásicos/farmacocinética , Inibidores Enzimáticos/farmacocinética , Ácidos Hidroxâmicos/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Cães , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Inibidores de Histona Desacetilases , Masculino , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Distribuição Tecidual , Vorinostat
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA