Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Regul Toxicol Pharmacol ; 67(3): 382-91, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24012707

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Antineoplásicos/toxicidad , Inmunoconjugados/toxicidad , Pruebas de Toxicidad , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/química , Antineoplásicos/inmunología , Antineoplásicos/farmacocinética , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Guías como Asunto , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Proyectos de Investigación , Pruebas de Toxicidad/métodos , Pruebas de Toxicidad/normas
2.
Regul Toxicol Pharmacol ; 58(3): 369-81, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20801182

RESUMEN

In order to determine a threshold for nongenotoxic carcinogens, the traditional risk assessment approach has been to identify a mode of action (MOA) with a nonlinear dose-response. The dose-response for one or more key event(s) linked to the MOA for carcinogenicity allows a point of departure (POD) to be selected from the most sensitive effect dose or no-effect dose. However, this can be challenging because multiple MOAs and key events may exist for carcinogenicity and oftentimes extensive research is required to elucidate the MOA. In the present study, a microarray analysis was conducted to determine if a POD could be identified following short-term oral rat exposure with two nongenotoxic rodent carcinogens, fenofibrate and methapyrilene, using a benchmark dose analysis of genes aggregated in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) biological processes, which likely encompass key event(s) for carcinogenicity. The gene expression response for fenofibrate given to rats for 2days was consistent with its MOA and known key events linked to PPARα activation. The temporal response from daily dosing with methapyrilene demonstrated biological complexity with waves of pathways/biological processes occurring over 1, 3, and 7days; nonetheless, the benchmark dose values were consistent over time. When comparing the dose-response of toxicogenomic data to tumorigenesis or precursor events, the toxicogenomics POD was slightly below any effect level. Our results suggest that toxicogenomic analysis using short-term studies can be used to identify a threshold for nongenotoxic carcinogens based on evaluation of potential key event(s) which then can be used within a risk assessment framework.


Asunto(s)
Carcinógenos/toxicidad , Fenofibrato/toxicidad , Metapirileno/análisis , Metapirileno/toxicidad , Neoplasias/inducido químicamente , Toxicogenética/métodos , Animales , Carcinógenos/administración & dosificación , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Relación Dosis-Respuesta a Droga , Femenino , Fenofibrato/administración & dosificación , Expresión Génica , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Metapirileno/administración & dosificación , Neoplasias/genética , Nivel sin Efectos Adversos Observados , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Medición de Riesgo
3.
Nat Biotechnol ; 30(2): 184-9, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22267010

RESUMEN

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Asunto(s)
Anticuerpos/sangre , Anticuerpos/inmunología , Sitios de Unión de Anticuerpos/inmunología , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoglobulina G/química , Ingeniería de Proteínas , Aminobenzoatos/química , Aminobenzoatos/inmunología , Animales , Anticuerpos/química , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Línea Celular Tumoral , Supervivencia Celular , Cisteína/química , Humanos , Inmunoconjugados/administración & dosificación , Inmunoglobulina G/inmunología , Macaca fascicularis , Maleimidas/química , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/inmunología , Maitansina/química , Maitansina/inmunología , Ratones , Ratones Desnudos , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/inmunología , Conformación Proteica , Ratas , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Trastuzumab
4.
Clin Cancer Res ; 16(19): 4769-78, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20805300

RESUMEN

PURPOSE: Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examined the in vitro potency as well as in vivo preclinical efficacy and safety profiles of a heterogeneous preparation of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. EXPERIMENTAL DESIGN AND RESULTS: To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) molecule was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 molecules per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti-cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equivalent doses (µg/m(2)) and retained efficacy at equivalent antibody doses (mg/kg). An improved safety profile of >2-fold was observed in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6,000 µg DM1/m(2)) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6,000 µg DM1/m(2)). CONCLUSIONS: The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clinical development of HER2-positive targeted breast cancer therapies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Maitansina/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Compuestos de Sulfhidrilo/farmacología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales Humanizados , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Humanos , Macaca fascicularis , Maitansina/química , Ratones , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2/química , Compuestos de Sulfhidrilo/química , Trastuzumab
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA