Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 20(2): 357-366, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33298591

RESUMO

Targeted T-cell redirection is a promising field in cancer immunotherapy. T-cell bispecific antibodies (TCB) are novel antibody constructs capable of binding simultaneously to T cells and tumor cells, allowing cross-linking and the formation of immunologic synapses. This in turn results in T-cell activation, expansion, and tumor killing. TCB activity depends on system-related properties such as tumor target antigen expression as well as antibody properties such as binding affinities to target and T cells. Here, we developed a systems model integrating in vitro data to elucidate further the mechanism of action and to quantify the cytotoxic effects as the relationship between targeted antigen expression and corresponding TCB activity. In the proposed model, we capture relevant processes, linking immune synapse formation to T-cell activation, expansion, and tumor killing for TCBs in vitro to differentiate the effect between tumor cells expressing high or low levels of the tumor antigen. We used cibisatamab, a TCB binding to carcinoembryonic antigen (CEA), to target different tumor cell lines with high and low CEA expression in vitro We developed a model to capture and predict our observations, as a learn-and-confirm cycle. Although full tumor killing and substantial T-cell activation was observed in high expressing tumor cells, the model correctly predicted partial tumor killing and minimal T-cell activation in low expressing tumor cells when exposed to cibisatamab. Furthermore, the model successfully predicted cytotoxicity across a wide range of tumor cell lines, spanning from very low to high CEA expression.


Assuntos
Anticorpos Biespecíficos/metabolismo , Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , Humanos
2.
J Pharmacokinet Pharmacodyn ; 44(6): 617-630, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29090407

RESUMO

Non-small cell lung cancer (NSCLC) patients greatly benefit from the treatment with tyrosine kinase inhibitors (TKIs) targeting the epidermal growth factor receptor (EGFR). However, emergence of acquired resistance inevitable occurs after long-term treatment in most patients and limits clinical improvement. In the present study, resistance to drug treatment in patient-derived NSCLC xenograft mice was assessed and modeling and simulation was applied to understand the dynamics of drug resistance as a basis to explore more beneficial drug regimen. Two semi-mechanistic models were fitted to tumor growth inhibition profiles during and after treatment with erlotinib or gefitinib. The base model proposes that as a result of drug treatment, tumor cells stop proliferating and undergo several stages of damage before they eventually die. The acquired resistance model adds a resistance term to the base model which assumes that resistant cells are emerging from the pool of damaged tumor cells. As a result, tumor cells sensitive to drug treatment will either die or be converted to a drug resistant cell population which is proliferating at a slower growth rate as compared to the sensitive cells. The observed tumor growth profiles were better described by the resistance model and emergence of resistance was concluded. In simulation studies, the selection of resistant cells was explored as well as the time-variant fraction of resistant over sensitive cells. The proposed model provides insight into the dynamic processes of emerging resistance. It predicts tumor regrowth during treatment driven by the selection of resistant cells and explains why faster tumor regrowth may occur after discontinuation of TKI treatment. Finally, it is shown how the semi-mechanistic model can be used to explore different scenarios and to identify optimal treatment schedules in clinical trials.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Modelos Biológicos , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Ensaios Clínicos como Assunto/métodos , Relação Dose-Resposta a Droga , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Inibidores de Proteínas Quinases/farmacocinética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Mol Cancer Ther ; 15(12): 3110-3119, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27638857

RESUMO

We quantitatively compare the efficacy of two approved EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, based on in vivo and in vitro data and show how a modeling approach can be used to scale from animal to humans. Gefitinib shows a higher tumor uptake in cancer patients, and we explored the potential impact on pharmacologic and antitumor activity in in vitro and in xenograft mice. Tumor growth inhibition was monitored, and the pharmacokinetics (PK) in plasma and tumor, as well as temporal changes of phospho-Erk (pErk) signals were examined in patient-derived tumor xenograft mice. These data were integrated in a translational PKPD model, allowing us to project an efficacious human dose, which we retrospectively compared with prescribed doses for cancer patients. In vitro experiments showed that cell-cycle arrest was similar for erlotinib and gefitinib. Similar pERK biomarker responses were obtained despite a 6.6-fold higher total tumor exposure for gefitinib. The PKPD model revealed a 3.7-fold higher in vivo potency for gefitinib, which did not translate into a lower anticipated efficacious dose in humans. The model-based dose prediction matched the recommended clinical doses well. These results suggest that despite having lower total tumor-to-plasma ratios, active drug exposure at target site is higher for erlotinib. Considering the PK properties, this translates in a 50% lower recommended daily dose of erlotinib in cancer patients. In summary, total exposure at target site is not suitable to rank compounds, and an integrated modeling and experimental approach can assess efficacy more accurately. Mol Cancer Ther; 15(12); 3110-9. ©2016 AACR.


Assuntos
Antineoplásicos/farmacocinética , Cloridrato de Erlotinib/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/farmacocinética , Algoritmos , Animais , Biomarcadores , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Gefitinibe , Humanos , Camundongos , Modelos Biológicos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA