Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
Proc Natl Acad Sci U S A ; 105(44): 17079-84, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18971340

RESUMO

Whereas target-specific drugs are available for treating ERBB2-overexpressing and hormone receptor-positive breast cancers, no tailored therapy exists for hormone receptor- and ERBB2-negative ("triple-negative") mammary carcinomas. Triple-negative tumors account for 15% of all breast cancers and frequently harbor defects in DNA double-strand break repair through homologous recombination (HR), such as BRCA1 dysfunction. The DNA-repair defects characteristic of BRCA1-deficient cells confer sensitivity to poly(ADP-ribose) polymerase 1 (PARP1) inhibition, which could be relevant to treatment of triple-negative tumors. To evaluate PARP1 inhibition in a realistic in vivo setting, we tested the PARP inhibitor AZD2281 in a genetically engineered mouse model (GEMM) for BRCA1-associated breast cancer. Treatment of tumor-bearing mice with AZD2281 inhibited tumor growth without signs of toxicity, resulting in strongly increased survival. Long-term treatment with AZD2281 in this model did result in the development of drug resistance, caused by up-regulation of Abcb1a/b genes encoding P-glycoprotein efflux pumps. This resistance to AZD2281 could be reversed by coadministration of the P-glycoprotein inhibitor tariquidar. Combination of AZD2281 with cisplatin or carboplatin increased the recurrence-free and overall survival, suggesting that AZD2281 potentiates the effect of these DNA-damaging agents. Our results demonstrate in vivo efficacy of AZD2281 against BRCA1-deficient breast cancer and illustrate how GEMMs of cancer can be used for preclinical evaluation of novel therapeutics and for testing ways to overcome or circumvent therapy resistance.


Assuntos
Antineoplásicos/uso terapêutico , Proteína BRCA1/genética , Neoplasias Mamárias Animais/tratamento farmacológico , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Compostos de Platina/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Inibidores da Aromatase/uso terapêutico , Cisplatino/uso terapêutico , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Transplante de Neoplasias , Poli(ADP-Ribose) Polimerases/genética
3.
Clin Cancer Res ; 14(12): 3916-25, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559613

RESUMO

PURPOSE: To assess efficacy of the novel, selective poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor AZD2281 against newly established BRCA2-deficient mouse mammary tumor cell lines and to determine potential synergy between AZD2281 and cisplatin. EXPERIMENTAL DESIGN: We established and thoroughly characterized a panel of clonal cell lines from independent BRCA2-deficient mouse mammary tumors and BRCA2-proficient control tumors. Subsequently, we assessed sensitivity of these lines to conventional cytotoxic drugs and the novel PARP inhibitor AZD2281. Finally, in vitro combination studies were done to investigate interaction between AZD2281 and cisplatin. RESULTS: Genetic, transcriptional, and functional analyses confirmed the successful isolation of BRCA2-deficient and BRCA2-proficient mouse mammary tumor cell lines. Treatment of these cell lines with 11 different anticancer drugs or with gamma-irradiation showed that AZD2281, a novel and specific PARP inhibitor, caused the strongest differential growth inhibition of BRCA2-deficient versus BRCA2-proficient mammary tumor cells. Finally, drug combination studies showed synergistic cytotoxicity of AZD2281 and cisplatin against BRCA2-deficient cells but not against BRCA2-proficient control cells. CONCLUSION: We have successfully established the first set of BRCA2-deficient mammary tumor cell lines, which form an important addition to the existing preclinical models for BRCA-mutated breast cancer. The exquisite sensitivity of these cells to the PARP inhibitor AZD2281, alone or in combination with cisplatin, provides strong support for AZD2281 as a novel targeted therapeutic against BRCA-deficient cancers.


Assuntos
Proteína BRCA2/genética , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Neoplasias Mamárias Animais/tratamento farmacológico , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia , Feminino , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos da radiação , Inibidores de Poli(ADP-Ribose) Polimerases , Rad51 Recombinase/genética
4.
Int J Biochem Cell Biol ; 39(3): 565-75, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17098463

RESUMO

Thymidine phosphorylase (TP) and uridine phosphorylase (UP) are often upregulated in solid tumors and catalyze the phosphorolysis of natural (deoxy)nucleosides and a wide variety of fluorinated pyrimidine nucleosides. Because the relative contribution of each of the two enzymes to these reactions is still largely unknown, we investigated the substrate specificity of TP and UP in colon cancer cells for the (fluoro)pyrimidine nucleosides thymidine (TdR), uridine (Urd), 5'-deoxy-5-fluorouridine (5'DFUR), and 5FU. Specific inhibitors of TP (TPI) and UP (BAU) were used to determine the contribution of each enzyme in relation to their cytotoxic effect. The high TP expressing Colo320TP1 cells were most sensitive to 5'DFUR and 5FU, with IC50 values of 1.4 and 0.2 microM, respectively, while SW948 and SW1398 were insensitive to 5'DFUR (IC50>150 microM for 5'DFUR). TPI and BAU only moderately affected sensitivity of Colo320, SW948, and SW1398, whereas TPI significantly increased IC(50) for 5'DFUR (50-fold) and 5FU (11-fold) in Colo320TP1 and BAU that in C26A (9-fold for 5'DFUR; p<0.01). In the epithelial skin cell line HaCaT both inhibitors were able to decrease sensitivity to 5'DFUR and 5FU separately. HaCaT might be a model for 5'DFUR toxicity. In the colon cancer cells 5'DFUR degradation varied from 0.4 to 50 nmol 5FU/h/10(6)cells, that of TdR from 0.3 to 103 nmol thymine/h/10(6)cells, that of Urd from 0.8 to 79 nmol uracil/h/10(6)cells, while conversion of 5FU to FUrd was from 0.3 to 46 nmol/h/10(6)cells. SW948 and SW1398 were about equally sensitive to 5'DFUR and 5FU, but SW1398 had higher phosphorylase activity (>65-fold) compared to SW948. In SW948 and HaCaT TPI and BAU inhibited TdR and Urd phosphorolysis (>80%), respectively. Both TP and UP contributed to the phosphorolysis of 5'DFUR and 5FU. In the presence of both inhibitors, still phosphorolysis of 5FU (>40%) was detected in the tumor and HaCaT cell lines, and remarkably, that of all four substrates in SW1398 cells. 5'DFUR phosphorolysis was also measured in situ, where Colo320TP1, SW1398, and HaCaT cells produced significant amounts 5FU from 5'DFUR (>10 nmol/24h/10(6)cells). In Colo320TP1 and in HaCaT cells TPI completely prevented 5FU production, but not in SW1398 cells, where BAU decreased this by 67% (p<0.01). High uracil and dUrd levels were detected in the medium. Uracil accumulation was heavily reduced in the presence of TPI for Colo320TP1 and HaCaT cells, whereas 5FU-induced dUrd production by these cell lines increased (p<0.01). In contrast, for SW1398 cells only BAU was able to reduce uracil levels, and dUrd production remained unchanged. In conclusion, overlapping substrate specificity was found for TP and UP in the cell lines, in which both enzymes were responsible for converting TdR and Urd, and 5'DFUR. 5'DFUR and 5FU were converted to their products in both the colon cancer cells and keratinocytes.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Pirimidinas/farmacologia , Timidina Fosforilase/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Floxuridina/metabolismo , Floxuridina/farmacologia , Humanos , Cinética , Pirimidinas/metabolismo , Especificidade por Substrato , Timidina Fosforilase/antagonistas & inibidores , Uridina/análogos & derivados , Uridina/metabolismo , Uridina/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Uridina Fosforilase/metabolismo
6.
Eur J Pharmacol ; 491(2-3): 93-9, 2004 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15140625

RESUMO

Thymidine phosphorylase (platelet-derived-endothelial-cell-growth-factor) catalyzes the reversible phosphorolysis of thymidine to thymine and 2-deoxyribose-1-phosphate, activates 5'-deoxy-5-fluorouridine (5'DFUR) and inactivates trifluorothymidine (TFT). The effect of 5'DFUR and TFT with or without a specific thymidine phosphorylase inhibitor (TPI) on thymidine phosphorylase mRNA, protein expression and activity was studied, in three human colon cancer cell lines, WiDR, HT29 and Lovo exposed for 72 h at IC50 concentrations. In Lovo cells TFT plus TPI only increased thymidine phosphorylase-protein expression 1.7-fold; 5'DFUR and TFT treatment increased thymidine phosphorylase mRNA levels 5- and 1.4-fold, respectively. In WiDR cells, 5'DFUR plus TPI significantly decreased thymidine phosphorylase-protein. TFT and TFT plus TPI increased thymidine phosphorylase-protein 2- and 3-fold, respectively. TPI and 5'DFUR decreased thymidine phosphorylase-mRNA levels significantly. In HT29 cells, 5'DFUR and 5'DFUR plus TPI decreased both thymidine phosphorylase-protein and thymidine phosphorylase-mRNA. In all cell lines 5'DFUR and TFT did not affect thymidine phosphorylase activity, but treatment with TPI (alone or in combination) eliminated thymidine phosphorylase activity. This demonstrated that regulation is drug and cell line dependent.


Assuntos
Inibidores Enzimáticos/farmacologia , Flúor/farmacologia , Pirimidinas/farmacologia , Timidina Fosforilase/antagonistas & inibidores , Timidina Fosforilase/biossíntese , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos
7.
Cancer Res ; 69(6): 2244-51, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19258500

RESUMO

Fibroblast growth factor receptor 1 (FGFR1) is frequently amplified and highly expressed in lobular carcinomas of the breast. In this report, we evaluated the biological activity of FGFR1 in a wide range of in vitro assays. Conditional activation of FGFR1 in the nontransformed MCF10A human mammary cell line, MCF10A, resulted in cellular transformation marked by epidermal growth factor-independent cell growth, anchorage-independent cell proliferation and survival, loss of cell polarity, and epithelial-to-mesenchymal transition. Interestingly, small-molecule or small interfering RNA inhibition of ribosomal S6 kinase (RSK) activity induced death of the FGFR1-transformed cells, but not of the parental MCF10A cell line. The dependence of FGFR1-transformed cells on RSK activity was further confirmed in cell lines derived from mouse and human lobular carcinomas that possess high FGFR1 activity. Taken together, these results show the transforming activity of FGFR1 in mammary epithelial cells and identify RSK as a critical component of FGFR1 signaling in lobular carcinomas, thus implicating RSK as a candidate therapeutic target in FGFR1-expressing tumors.


Assuntos
Neoplasias da Mama/metabolismo , Transformação Celular Neoplásica/metabolismo , Glândulas Mamárias Humanas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Lobular/enzimologia , Carcinoma Lobular/genética , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Glândulas Mamárias Humanas/enzimologia , Glândulas Mamárias Humanas/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Transdução de Sinais
8.
Cancer Sci ; 98(6): 779-89, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17441963

RESUMO

Current treatment modalities for cancer combine cytotoxic drugs against DNA and novel targeted drugs affecting signal transduction pathways, which are required for growth progression and metastasizing tumors. Classical chemotherapeutic regimens for gastro-intestinal tumors include antimetabolites based on 5-fluorouracil (5FU), the platinum analog oxaliplatin and the topoisomerase inhibitor irinotecan. The thymidine analog trifluorothymidine (TFT) has been shown to bypass resistance pathways for 5FU derivatives (S-1, UFT, Xeloda) in model systems, while concurrent application with a thymidine phosphorylase inhibitor (TPI) increases the bioavailability of TFT, thereby potentiating the in vivo efficacy of TFT. The formulation TAS-102 is given orally in a 1.0:0.5 molar ratio (TFT:TPI). The formulation is dual-targeted due to the cytotoxic effect of TFT, which is enhanced by TPI, while TPI also exerts antiangiogenic effects by inhibiting thymidine phosphorylase (TP), also known as platelet-derived endothelial cell growth factor. Evidence is accumulating from in vitro and in vivo preclinical studies that these properties favor further combinations with other cytotoxic agents currently being used in the treatment of gastro-intestinal tumors. Also treatment with targeted agents will synergistically down-regulate signal transduction pathways responsible for growth and progression of tumors. In this review, we summarize the available information on (clinical) pharmacology, mechanisms of action, pharmacodynamic and pharmacokinetic properties, early clinical trials and future directions of the new potent combination drug TAS-102.


Assuntos
Neoplasias Gastrointestinais/tratamento farmacológico , Trifluridina/uso terapêutico , Uracila/análogos & derivados , Administração Oral , Antimetabólitos Antineoplásicos/uso terapêutico , Disponibilidade Biológica , Ensaios Clínicos como Assunto , Combinação de Medicamentos , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Previsões , Humanos , Pirimidinas , Pirrolidinas , Timidina/análogos & derivados , Timidina/farmacologia , Timina , Trifluridina/farmacocinética , Trifluridina/farmacologia , Uracila/farmacocinética , Uracila/farmacologia , Uracila/uso terapêutico
9.
Anticancer Drugs ; 16(3): 285-92, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15711180

RESUMO

Trifluorothymidine (TFT) is a fluorinated thymidine analog that after conversion to its monophosphate derivative can inhibit thymidylate synthase (TS) and be incorporated into DNA. TFT is a good substrate for thymidine phosphorylase (TP), and the combination of TFT and a TP inhibitor (TPI), called TAS-102, has been developed to enhance the bioavailability of TFT in vivo, and is currently being studied in a phase I study. We aimed to determine the limiting factor(s) in the cytotoxicity of TFT with or without TPI to cancer cells. Colon cancer and lung cancer cell lines with either an overexpression or deficiency of one of the enzymes involved in TFT metabolism were used to study the effect of TPI on TFT sensitivity and the role of TS inhibition. The synthesis of radioactive TFT metabolites was studied using thin-layer chromatography together with the incorporation of TFT into DNA. We found that despite a high rate of TFT phosphorolysis, cells with high TP expression are not more resistant to TFT, while TPI did not increase TFT sensitivity. High TS-expressing cells were shown to be cross-resistant to a 72-h exposure to TFT compared to 5-fluorouracil (5-FU), although this was more pronounced at a 4-h exposure (3.4-fold or more for TFT and 1.4-fold or more for 5-FU). Despite a moderate inhibition of TS activity in cells expressing high TS, these cells were more sensitive to TFT than 5-FU (3.8-fold or more). Only in Colo320TP1 cells expressing high TP, inhibition of TFT phosphorolysis by TPI increased formation of active TFT metabolites 1.8-fold, although this was not related to an increase in TFT incorporation into DNA. These studies show that uptake of TFT and subsequent phosphorylation of TFT by cancer cells is very rapid. Despite a high rate of degradation, the activation pathways are still saturated and sufficient to inhibit TS and enable incorporation into DNA, although the contribution of each effect is exposure time dependent.


Assuntos
Antimetabólitos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Fluoruracila/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Timidina Fosforilase/metabolismo , Timidilato Sintase/antagonistas & inibidores , Trifluridina/farmacologia , Antimetabólitos/metabolismo , Antimetabólitos/farmacocinética , Antimetabólitos Antineoplásicos , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , DNA/metabolismo , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/metabolismo , Timidilato Sintase/metabolismo , Trifluridina/metabolismo , Trifluridina/farmacocinética , Células Tumorais Cultivadas
10.
Mol Pharmacol ; 66(4): 1054-60, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15269287

RESUMO

Platelet-derived endothelial cell growth factor/thymidine phosphorylase (PD-ECGF/TP) and interleukin-8 (IL-8) are angiogenic factors produced by tumor infiltrating macrophages. Here, we show that prolonged exposure of human monocytic/macrophage THP1 and U937 cells to sulfasalazine, an anti-inflammatory drug and inhibitor of nuclear factor-kappaB (NF-kappaB), resulted in down-regulation of PD-ECGF/TP and IL-8 (mRNA, protein and activity) along with elimination of their induction by tumor necrosis factor-alpha and interferon-gamma. Concomitantly, sulfasalazine-exposed cells were markedly resistant to 5'-deoxyfluorouridine, the last intermediate of capecitabine requiring activation by PD-ECGF/TP. This is the first report suggesting that disruption of NF-kappaB-dependent signaling pathways can provoke a marked and sustained down-regulation of macrophage-related angiogenic factors. However, this may also negatively affect capecitabine efficacy.


Assuntos
Expressão Gênica/efeitos dos fármacos , Interleucina-8/metabolismo , Macrófagos/efeitos dos fármacos , Sulfassalazina/farmacologia , Timidina Fosforilase/metabolismo , Indutores da Angiogênese/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Western Blotting , Humanos , Interleucina-8/genética , Macrófagos/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B , RNA Mensageiro/metabolismo , Receptores de Interferon/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Timidina Fosforilase/genética , Fator de Transcrição RelA , Fatores de Transcrição/metabolismo , Células U937 , Receptor de Interferon gama
11.
J Gene Med ; 6(3): 317-27, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15026993

RESUMO

BACKGROUND: To improve the prognosis of patients with gastric cancer it is important to develop novel treatment modalities targeting the malignant behavior of tumor cells. Concerning this, NK4, which acts as HGF-antagonist and angiogenesis inhibitor, might be a potential therapeutic agent for gastric cancer. The HGF-c-MET pathway plays a pivotal role in gastric tumor growth, invasion, metastasis and angiogenesis. Therefore, the current study investigates whether adenoviral vector-mediated NK4 gene therapy has therapeutic potential for gastric cancer. METHODS: Expression of HGF and c-MET in normal and (pre-)malignant gastric tissue was studied by immunohistochemistry. The effects of adenoviral vector-mediated expression of NK4 on the biological behavior of gastric cancer cells were studied in vitro and in vivo. RESULTS: The majority of gastric cancers, i.e. 76%, express c-MET and in all carcinomas HGF is expressed in either tumor or stromal cells. Normal gastric epithelial cells do not express either of these proteins. Transduction of gastric cancer cells with the replication-deficient adenoviral vector AdCMV.NK4 resulted in efficient production and secretion of NK4. Consequently, proliferation, migration and invasion of gastric cancer cells were significantly inhibited. In addition, significantly reduced proliferation of vascular endothelial cells and efficient inhibition of angiogenesis were achieved. Finally, treatment of established human gastric tumor xenografts with AdCMV.NK4 resulted in significant tumor growth delay and significant reduction of intratumoral microvessel density. CONCLUSIONS: The present study shows that adenoviral vector-mediated expression of NK4 is a promising strategy to treat human gastric cancer by simultaneous interfering with primary tumor growth, metastasis and angiogenesis.


Assuntos
Adenoviridae/genética , Fator de Crescimento de Hepatócito/genética , Mitógenos/genética , Neovascularização Patológica/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/terapia , Animais , Aorta/metabolismo , Movimento Celular , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos , Imunoquímica , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-met/metabolismo , Ratos , Ratos Wistar , Esferoides Celulares , Neoplasias Gástricas/irrigação sanguínea , Neoplasias Gástricas/patologia , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA