Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cancer Med ; 5(11): 3176-3185, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27734608

RESUMO

Regorafenib is an orally administered inhibitor of protein kinases involved in tumor angiogenesis, oncogenesis, and maintenance of the tumor microenvironment. Phase III studies showed that regorafenib has efficacy in patients with advanced gastrointestinal stromal tumors or treatment-refractory metastatic colorectal cancer. In clinical studies, steady-state exposure to the M-2 and M-5 metabolites of regorafenib was similar to that of the parent drug; however, the contribution of these metabolites to the overall observed clinical activity of regorafenib cannot be investigated in clinical trials. Therefore, we assessed the pharmacokinetics and pharmacodynamics of regorafenib, M-2, and M-5 in vitro and in murine xenograft models. M-2 and M-5 showed similar kinase inhibition profiles and comparable potency to regorafenib in a competitive binding assay. Inhibition of key target kinases by all three compounds was confirmed in cell-based assays. In murine xenograft models, oral regorafenib, M-2, and M-5 significantly inhibited tumor growth versus controls. Total peak plasma drug concentrations and exposure to M-2 and M-5 in mice after repeated oral dosing with regorafenib 10 mg/kg/day were comparable to those in humans. In vitro studies showed high binding of regorafenib, M-2, and M-5 to plasma proteins, with unbound fractions of ~0.6%, ~0.9%, and ~0.4%, respectively, in murine plasma and ~0.5%, ~0.2%, and ~0.05%, respectively, in human plasma. Estimated free plasma concentrations of regorafenib and M-2, but not M-5, exceeded the IC50 at human and murine VEGFR2, suggesting that regorafenib and M-2 are the primary contributors to the pharmacologic activity of regorafenib in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Inibidores da Angiogênese/farmacocinética , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Metaboloma , Metabolômica/métodos , Camundongos , Compostos de Fenilureia/farmacocinética , Ligação Proteica , Mapeamento de Interação de Proteínas , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Quinases/metabolismo , Piridinas/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Eur J Cancer ; 48(13): 2054-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21937219

RESUMO

Two new water-soluble folate receptor-targeted drug conjugates that contain the highly active doxorubicin derivative N-(5,5-diacetoxybut-1-yl)doxorubicin were designed and evaluated for their biological activity against folate receptor positive tumours. The prodrugs were designed to contain an acid-sensitive hydrazone bond KO019 or in addition a disulphide bond KO013 in order to elucidate the importance of the pre-determined breaking point for their in vitro and in vivo properties. Fluorescence microscopy studies confirmed higher uptake of the prodrugs in folate receptor positive KB cells than in the folate receptor negative A549 lung cancer cells. In subsequent in vivo studies in the folate receptor positive KB xenograft model, KO019 was as active as the free drug but significantly less toxic when dosed at twice the dose of the free drug whereas KO013 showed no anticancer efficacy. As an explanation, we could show by HPLC that the prodrug KO013 that additionally contains a disulphide bond undergoes rapid disulphide exchange in murine plasma in the order of 40% after 5h at 37°C in contrast to KO019 which was essentially stable after a 5h incubation.


Assuntos
Doxorrubicina/análogos & derivados , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/análogos & derivados , Pró-Fármacos/farmacologia , Pterinas/farmacologia , Alcaloides de Vinca/farmacologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Receptores de Folato com Âncoras de GPI/antagonistas & inibidores , Ácido Fólico/farmacologia , Camundongos , Camundongos Nus , Pró-Fármacos/química , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Biotechnol ; 46(2): 197-205, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20512535

RESUMO

The cytomegalovirus-immediate early (CMV-IE) promoter is widely used as a strong and constitutively active promoter. Although the CMV-IE promoter does not harbor heat-responsive sequences, we determined its heat inducibility. We analyzed in vitro and in vivo heat responsiveness and possible mechanisms of heat induction of the CMV-IE promoter. We used transfected SW480 human colon carcinoma cells (SW480/CMVCD), expressing CMV-IE promoter-driven bacterial cytosine deaminase (CD) gene. These cells were heated at 42 degrees C. The SW480/CMVCD cells were also used for in vivo studies, in which tumor-bearing animals were treated with hyperthermia at 41.5 degrees C. As controls, SW480 (SW480/HSPCD) cells were used, in which CD expression is driven by the HSP70-promoter. In vitro, we observed a biphasic, up to 25-fold heat induction of CMV-IE-driven CD expression after hyperthermia in SW480/CMVCD cells. In vivo, we found a 2.5-fold induction of CD expression after hyperthermia in SW480/CMVCD tumor-bearing animals. The analysis of the CMV-IE promoter sequence revealed several transcription factor-binding sites, which mediate stress responsiveness. YB-1 and C/EBP-beta might mediate heat responsiveness of the CMV-IE promoter. These data point to limitations in heat-induction gene therapy studies, in which the CMV-IE promoter is used as control system. In addition, the CMV-IE promoter itself could well be used for construction of heat-inducible vectors.


Assuntos
Citosina Desaminase/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Terapia Genética/métodos , Hipertermia Induzida , Regiões Promotoras Genéticas/genética , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/terapia , Citosina Desaminase/genética , Modelos Animais de Doenças , Indução Enzimática/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Fatores de Transcrição/metabolismo
4.
Clin Cancer Res ; 16(5): 1452-65, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20179216

RESUMO

PURPOSE: Characterization of new anticancer drugs in a few xenograft models derived from established human cancer cell lines frequently results in the discrepancy between preclinical and clinical results. To take the heterogeneity of tumors into consideration more thoroughly, we describe here a preclinical approach that may allow a more rational clinical development of new anticancer drugs. EXPERIMENTAL DESIGN: We tested Sagopilone, an optimized fully synthetic epothilone, in 22 well-characterized patient-derived non-small cell lung cancer models and correlated results with mutational and genome-wide gene expression analysis. RESULTS: Response analysis according to clinical trial criteria revealed that Sagopilone induced overall responses in 64% of the xenograft models (14 of 22), with 3 models showing stable disease and 11 models showing partial response. A comparison with response rates for established drugs showed the strong efficacy of Sagopilone in non-small cell lung cancer. In gene expression analyses, Sagopilone induced tubulin isoforms in all tumor samples, but genes related to mitotic arrest only in responder models. Moreover, tumors with high expression of genes involved in cell adhesion/angiogenesis as well as of wild-type TP53 were more likely to be resistant to Sagopilone therapy. As suggested by these findings, Sagopilone was combined with Bevacizumab and Sorafenib, drugs targeting vascular endothelial growth factor signaling, in Sagopilone-resistant models and, indeed, antitumor activity could be restored. CONCLUSION: Analyses provided here show how preclinical studies can provide hypotheses for the identification of patients who more likely will benefit from new drugs as well as a rationale for combination therapies to be tested in clinical trials.


Assuntos
Antineoplásicos/uso terapêutico , Benzotiazóis/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Epotilonas/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Benzenossulfonatos/uso terapêutico , Bevacizumab , Carcinoma Pulmonar de Células não Pequenas/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Camundongos , Niacinamida/análogos & derivados , Compostos de Fenilureia , Piridinas/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sorafenibe
5.
Am J Chin Med ; 37(6): 1153-65, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19938223

RESUMO

The aim of the present work was to explore the anti-hepatoma effects of icariin both in vitro and in vivo and to elucidate its potential mechanism of action. The MTT assay was applied to test the anti-proliferative effects of icariin in vitro. HepG2 bearing NMRI nu/nu mice were used to test the anticancer effects of icariin in vivo. Immunohistochemical assay and flow cytometry assay (FACS) were applied to detect the possible mechanisms of action of icariin. MTT assay illustrated that icariin inhibited the proliferation of HepG2 cells in a concentration dependent manner; meanwhile, icariin inhibited the tumor growth in HepG2 bearing NMRI nu/nu mice. The tumor weight was inhibited by 55.6% and tumor volume was inhibited by 47.2%. Icariin did not influence the spleen and body weights or blood parameters. Immunohistochemical analysis indicated that the expressions of both CD31 and Ki67 in the icariin treated group were significantly lower than those in the control group (p < 0.01). FACS assay showed that icariin dramatically decreased the percentage of CD4+ and CD8+ cells in bone marrow and CD19+ cells in blood on day 8. On day 17, the percentage of CD8+ cells in blood was lower than those in the control group. CD4/CD8 ratio in icariin group was significantly elevated in bone marrow on day 17. Icariin showed anticancer efficacy both in vitro and in vivo. The possible mechanism of action could be related to its anti-angiogenesis and anti-proliferative effects in tumors.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Epimedium/química , Flavonoides/farmacologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Antígenos CD19/sangue , Antineoplásicos Fitogênicos/uso terapêutico , Medula Óssea/imunologia , Relação CD4-CD8 , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Flavonoides/uso terapêutico , Citometria de Fluxo , Células Hep G2 , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Nus , Fitoterapia , Extratos Vegetais/uso terapêutico , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Eur J Cardiothorac Surg ; 36(3): 454-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19502076

RESUMO

OBJECTIVE: Recent trials have evaluated adjuvant chemotherapy in patients with non-small-cell lung cancer (NSCLC). For stage IB to IIIA, a significant improvement of treatment results for platin-based chemotherapy was shown, but only one of the 20 patients treated has a benefit of disease-free and overall 5-year survival. In future the implementation of biomarkers, novel agents and individual selection may contribute to better treatment results in adjuvant therapy. Pre-clinical models are one way to study treatment innovations. MATERIALS AND METHODS: We have developed a lung cancer xenograft model. Fresh tumour material of patients with NSCLC was subcutaneously transplanted to immunodeficient mice shortly after surgical resection. In total, 102 samples have been transplanted from which 25 passagable models could be generated. Of the established xenograft lines, 48% were derived from squamous cell carcinomas and 24% from adenocarcinomas. All but one originated from long-term smokers. RESULTS: It could be shown that the early murine passages (maximum 10) were similar to the original tumour with regard to histology and the expression of the surface proteins as E-cadherin, EpCAM or the cell proliferation marker Ki-67. The growth rate of the established xenografts was a unique feature of the different models and not related to patient characteristics or to the histology type. All xenograft models showed a wide variability in response to both classical chemotherapy and targeted anti-epidermal growth factor receptor agents. Response rates were in good accordance with the results of recent clinical studies. DISCUSSION: In summary, we have developed a panel of patient-derived NSCLC xenografts. These xenograft models could be used for pre-clinical studies to evaluate chemotherapy, novel targeted therapies and expression of potential biomarkers.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Modelos Animais de Doenças , Neoplasias Pulmonares/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adulto , Idoso , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Cetuximab , Quimioterapia Adjuvante , Avaliação Pré-Clínica de Medicamentos/métodos , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Quinazolinas/uso terapêutico , Resultado do Tratamento
7.
Onkologie ; 31 Suppl 2: 34-8, 2008.
Artigo em Alemão | MEDLINE | ID: mdl-18487867

RESUMO

The preclinical development of novel anticancer compounds mainly resembles the same principle as valid for other drug classes. New target structures on tumors are identified within academic or industrial fundamental research groups enabling a selective attack of cancerostatics. After the proof of principle in molecular or cellular systems, substance candidates are selected which undergo a stepwise preclinical testing. This procedure consists of cell culture models (in vitro) and animal experiments (in vivo) using suitable tumor models. With selected lead compounds the pharmaceutical formulation is prepared, and the legally required preclinical toxicological investigations are performed before initiation of clinical phase I studies. There is one special feature distinguishing cytostatics from all other drugs - their narrow therapeutic index. This constraint is to be considered already in early phases of anticancer drug development, and a special focus should be given to the revealing of potential side effects. This contribution especially gives attention to this aspect of preclinical anticancer drug development. Further on, focus is put on target-oriented compounds, whose preclinical development generates special challenges.


Assuntos
Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto/normas , Avaliação Pré-Clínica de Medicamentos/normas , Fidelidade a Diretrizes/organização & administração , Neoplasias/tratamento farmacológico , Guias de Prática Clínica como Assunto , Pesquisa/normas , Animais , Composição de Medicamentos/normas , União Europeia , Alemanha , Humanos
8.
Mol Cancer Ther ; 6(1): 236-43, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17237282

RESUMO

The promoter of the human multidrug resistance gene (mdr1) harbors defined heat-responsive elements, which could be exploited for construction of heat-inducible expression vectors. To analyze the hyperthermia inducibility of the mdr1 promoter in vitro and in vivo, we used the pcDNA3-mdrp-hTNF vector construct for heat-induced tumor necrosis factor alpha (TNF-alpha) expression in transfected HCT116 human colon carcinoma cells at mRNA level by quantitative real-time reverse transcription-PCR and at protein level by TNF-alpha ELISA. For the in vitro studies, the pcDNA3-mdrp-hTNF-transfected tumor cells were treated with hyperthermia at 43 degrees C for 2 h. In the animal studies, stably transfected or in vivo jet-injected tumor-bearing Ncr:nu/nu mice were treated for 60 min at 42 degrees C to induce TNF-alpha expression. Both the in vitro and in vivo experiments show that hyperthermia activates the mdr1 promoter in a temperature- and time-dependent manner, leading to an up to 4-fold increase in mdr1 promoter-driven TNF-alpha expression at mRNA and an up to 3-fold increase at protein level. The in vivo heat-induced TNF-alpha expression combined with Adriamycin (8 mg/kg) treatment leads to the inhibition of tumor growth in the animals. These experiments support the idea that heat-induced mdr1 promoter-driven expression of therapeutic genes is efficient and feasible for combined cancer gene therapy approaches.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Neoplasias do Colo/genética , Neoplasias do Colo/terapia , Terapia Genética/métodos , Hipertermia Induzida , Regiões Promotoras Genéticas/genética , Fator de Necrose Tumoral alfa/genética , Animais , Sequência de Bases , Neoplasias do Colo/patologia , Terapia Combinada , Doxorrubicina/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Injeções a Jato , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
10.
Mol Cancer Ther ; 1(4): 239-46, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12467219

RESUMO

Novel 2-(4-aminophenyl)benzothiazoles (e.g., compounds 1 and 2) possess highly selective, potent antitumor properties in vitro and in vivo. Elucidation of the mechanism of action of this structurally simple class of compounds has occurred in parallel with selection of a candidate clinical agent. Antitumor benzothiazoles induce and are biotransformed by cytochrome P 450 1A1 to putative active, as well as inactive metabolites. Metabolic inactivation of the molecule has been thwarted by isosteric replacement of hydrogen with fluorine atoms at positions around the benzothiazole nucleus. Amino acid conjugation to the exocyclic primary amine function of 2-(4-aminophenyl)benzothiazoles has been used to overcome limitations posed by drug lipophilicity. Water soluble, chemically stable prodrugs rapidly and quantitatively revert to their parent amine in mice, rats, and dogs in vivo. Plasma concentrations of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (2) regenerated from the lysylamide prodrug (2b), sufficient to elicit cytocidal activity against ZR-75-1 and T47D human mammary carcinoma cell lines persist > 6 h. The growth of breast (MCF-7) and ovarian (IGROV-1) xenograft tumors is significantly retarded by 2b. Manageable toxic side effects are reported from preclinically efficacious doses of 2b. Cytochrome P 450 1A1 protein expression, selectively induced in sensitive carcinoma cells, was detected in MCF-7 and IGROV-1 tumors 24 h after treatment of mice with 2b (20 mg/kg). The lysyl amide prodrug of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole is potentially suitable for clinical evaluation.


Assuntos
Aminoácidos/metabolismo , Compostos de Anilina/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Tiazóis/uso terapêutico , Compostos de Anilina/farmacocinética , Animais , Antineoplásicos/farmacocinética , Benzotiazóis , Western Blotting , Divisão Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/metabolismo , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Masculino , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Camundongos Nus , Neoplasias Ovarianas/enzimologia , Pró-Fármacos , Ratos , Ratos Sprague-Dawley , Tiazóis/farmacocinética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA