Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 136: 1169-1175, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31170489

RESUMO

Gemcitabine (2'-deoxy-2',2'-difluorocytidine, dFdC) is one of the most effective chemotherapy drugs commonly used for treatment of various tumors. Despite its significant anticancer effects, some adverse effects create obstacles to treatment. The main toxicity of gemcitabine is myelosuppression, which not only reduces patient quality of life, but also hinders further anticancer treatment. In this respect, immunotherapy can address these drawbacks because of its ability to enhance the patient's immune system. To improve immune system function, yeast-derived ß-glucans, which are well-known biologic response modifiers, were administered to gemcitabine-treated mice. The in vivo experiment revealed that orally administered yeast (1 → 3)-(1 → 6)-ß-d-glucan effectively alleviated myelosuppression associated with gemcitabine-induced pancytopenia. Moreover, analysis of myelopoiesis-related cytokine expression through real-time PCR demonstrated that ß-glucan treatment significantly upregulated hematopoietic responses in gemcitabine-treated mice. Furthermore, orally administered ß-glucan significantly induced the expression of IFN-γ and IL-2 in splenocytes of gemcitabine-treated mice. It also restored the cytotoxicity of splenocytes against YAC-1 in gemcitabine-treated mice and displayed a positive effect on gemcitabine-damaged bone marrow tissue. In conclusion, yeast ß-glucans have the potential to be used as adjuvants for alleviating chemotherapy-induced immunosuppression in patients.


Assuntos
Desoxicitidina/análogos & derivados , Terapia de Imunossupressão/efeitos adversos , Leveduras/química , beta-Glucanas/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Desoxicitidina/efeitos adversos , Desoxicitidina/antagonistas & inibidores , Hematopoese/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pancitopenia/induzido quimicamente , Pancitopenia/tratamento farmacológico , Pancitopenia/imunologia , Baço/efeitos dos fármacos , Baço/imunologia , beta-Glucanas/uso terapêutico , Gencitabina
2.
Eur J Cancer ; 49(5): 1152-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23146955

RESUMO

AIM OF STUDY: Smoking is an established risk factor for pancreatic cancer and nicotine replacement therapy (NRT) often accompanies chemotherapy. The current study has tested the hypothesis that chronic exposure to low dose nicotine reduces the responsiveness of pancreatic cancer to the leading therapeutic for this cancer, gemcitabine. METHODS: The effects of chronic nicotine (1 µm/L) on two pancreatic cancer cell lines in vitro and in a xenograft model were assessed by immunoassays, Western blots and cell proliferation assays. RESULTS: Exposure in vitro to nicotine for 7 days inhibited the gemcitabine-induced reduction in viable cells, gemcitabine-induced apoptosis as indicated by reduced expression of cleaved caspase-3 while inducing the phosphorylation of signalling proteins extracellular signal-regulated kinase (ERK), v-akt thymoma viral oncogene homolog (protein kinase B, AKT) and Src. Nicotine (1 µm/L) in the drinking water for 4 weeks significantly reduced the therapeutic response of mouse xenografts to gemcitabine while reducing the induction of cleaved caspase-3 and the inhibition of phosphorylated forms of multiple signalling proteins by gemcitabine in xenograft tissues. CONCLUSIONS: Our experimental data suggest that continued moderate smoking and NRT may negatively impact therapeutic outcomes of gemcitabine on pancreatic cancer and that clinical studies in cancer patients are now warranted.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Desoxicitidina/análogos & derivados , Nicotina/efeitos adversos , Neoplasias Pancreáticas/tratamento farmacológico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Esquema de Medicação , Antagonismo de Drogas , Avaliação Pré-Clínica de Medicamentos , Humanos , Masculino , Camundongos , Camundongos Nus , Nicotina/administração & dosagem , Neoplasias Pancreáticas/patologia , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
3.
Biol Pharm Bull ; 33(2): 216-22, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20118543

RESUMO

Z-360 is a novel cholecystokinin (CCK)-2/gastrin receptor antagonist that is being developed for the treatment of pancreatic adenocarcinoma in combination with gemcitabine. A previous study shows that the co-administration of Z-360 with gemcitabine significantly prolonged the survival of mice with orthotopically implanted human pancreatic adenocarcinoma cell lines. To clarify the therapeutic effects of Z-360 in combined with gemcitabine, we analyzed gene expression. When gemcitabine was administered, CCK-2/gastrin receptor expression was induced in an orthotropic xenograft model; the result indicating that Z-360 could act on gemcitabine-sensitive cells. Both in vitro and in vivo studies showed that gemcitabine increased the expression of vascular endothelial growth factor A (VEGFA), a prognostic factor for survival in pancreatic cancer, while Z-360 suppressed this induction of VEGFA gene expression. These results help to explain how Z-360 prolongs survival when used in combination with gemcitabine.


Assuntos
Benzodiazepinonas/farmacologia , Desoxicitidina/análogos & derivados , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Receptor de Colecistocinina B/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Benzodiazepinonas/uso terapêutico , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Feminino , Humanos , Camundongos , Neoplasias Pancreáticas/genética , Receptor de Colecistocinina B/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Gencitabina
4.
Mol Cancer Ther ; 7(8): 2394-404, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18723486

RESUMO

Checkpoints are present in all phases of the cell cycle and are regarded as the gatekeepers maintaining the integrity of the genome. Many conventional agents used to treat cancer impart damage to the genome and activate cell cycle checkpoints. Many tumors are defective in the tumor suppressor p53 and therefore lack a functional G(1) checkpoint. In these tumors, however, the S-G(2) checkpoints remain intact and, in response to DNA damage, arrest cell cycle progression allowing time for DNA repair. Checkpoint kinase 1 (Chk1) is a key element in the DNA damage response pathway and plays a crucial role in the S-G(2)-phase checkpoints. Inhibiting Chk1 represents a therapeutic strategy for creating a "synthetic lethal" response by overriding the last checkpoint defense of tumor cells against the lethal damage induced by DNA-directed chemotherapeutic agents. Chk1 inhibition is consistent with emerging targeted therapies aiming to exploit molecular differences between normal and cancer cells. Adding a Chk1 inhibitor to DNA-damaging cytotoxic therapy selectively targets tumors with intrinsic checkpoint defects while minimizing toxicity in checkpoint-competent normal cells. PF-00477736 was identified as a potent, selective ATP-competitive small-molecule inhibitor that inhibits Chk1 with a K(i) of 0.49 nM. PF-00477736 abrogates cell cycle arrest induced by DNA damage and enhances cytotoxicity of clinically important chemotherapeutic agents, including gemcitabine and carboplatin. In xenografts, PF-00477736 enhanced the antitumor activity of gemcitabine in a dose-dependent manner. PF-00477736 combinations were well tolerated with no exacerbation of side effects commonly associated with cytotoxic agents.


Assuntos
Benzodiazepinonas/farmacologia , Dano ao DNA , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos , Pirazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Quinase 1 do Ponto de Checagem , Cromatografia Líquida , Desoxicitidina/análogos & derivados , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Histonas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Fosforilação , Ratos , Ratos Sprague-Dawley , Fase S/efeitos dos fármacos , Espectrometria de Massas em Tandem , Gencitabina
5.
Int J Oncol ; 32(1): 113-20, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18097549

RESUMO

The marked clinical anticancer activity of the paclitaxel (PTX) and gemcitabine (GEM) combination has suggested that the two drugs may interact more than additively. We have analyzed the in vitro growth and molecular interactions of the two chemotherapy drugs in a panel of human breast cancer cells. We evaluated cell viability in four breast cancer cell lines (i.e., MCF-7, MDA-MB-231, MDA-MB-468, and SKBR3) that were treated with PTX and GEM combined either simultaneously (PTX + GEM) or sequentially (PTX --> GEM; GEM --> PTX). PTX-GEM interactions at the cellular level were assessed mathematically employing both the isobologram analysis (Berenbaum) and the combination index (Chou-Talalay) method. PTX-GEM molecular interactions on the apoptotic markers PARP, Bcl-2 and Bax were analyzed by immunoblotting procedures. Apoptosis was detected using a DNA ladder assay. We observed significant synergistic growth inhibitory interactions when PTX was administered before GEM. Additive interactions were observed when both the simultaneous regimen and the GEM followed by PTX regimen were used. DNA ladder and Western blotting results in the PTX followed by GEM sequence revealed a significant increase in the apoptotic cell death of breast cancer cells related to the Bax/Bcl-2 apoptotic pathway. In summary, the occurrence of clinically relevant synergism between PTX and GEM suggests a sequence-dependent nature in human breast cancer cells. This synergistic interaction on the PTXright curved arrow GEM schedule appears to be related to an increase in the Bcl-2-related mitochondrial apoptotic pathway. The synergism that we have observed may explain the favorable clinical responses that have been achieved in clinical studies, in which patients are administered PTX first, and then GEM.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Desoxicitidina/análogos & derivados , Paclitaxel/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Esquema de Medicação , Sinergismo Farmacológico , Feminino , Humanos , Paclitaxel/antagonistas & inibidores , Poli(ADP-Ribose) Polimerases/metabolismo , Gencitabina
6.
Cancer Biol Ther ; 5(10): 1294-303, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16929163

RESUMO

Although the recently-developed Gemcitabine (GEM) has renewed interest in clinical research in pancreatic carcinoma, it offers modest improvement of tumor-related symptoms and marginal survival advantage, even when combined with other currently-available chemotherapeutic agents such as 5-Fluorouracil (5-FU). We hypothesized that this disappointing result could be due to an interaction between the two drugs affecting cytotoxic activity. We measured in-vitro growth inhibition, cell cycle distribution, gene and protein expression of apoptosis regulators bcl-2, bcl-x and survivin, NFkappaB and telomerase activities of human pancreatic carcinoma cell line Capan-2 following exposure to GEM and 5-FU singly or combined, by MTT assay and median effect analysis, flow cytometry, real-time RT-PCR, Western blotting, electrophoretic mobility shift assay (EMSA) and telomeric repeat amplification protocol (TRAP) assay, respectively. We found cell growth to be inhibited by both drugs, decreasing the percentage of cells in S and G2/M phases and inducing apoptosis, dependent on the levels of bcl-2, bcl-xL and survivin expression in the case of 5-FU, but not for GEM. Moreover, while telomerase activity was reduced equally by both drugs, 5-FU but not GEM effectively downregulated NFkappaB binding activity. Intriguingly, a substantial antagonistic effect was noticed when GEM was combined with 5-FU in the concentration range tested, with the exception of the TRAP assay. These indications of an antagonistic interaction between GEM and 5-FU in some pancreatic cancer context urge further investigation of both genetic and non-genetic differences to identify the variables most relevant for optimal selection and dosing of treatment for the individual patient.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Fluoruracila/farmacologia , Caspase 2/metabolismo , Inibidores de Caspase , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Fluoruracila/antagonistas & inibidores , Humanos , Cinética , Neoplasias Pancreáticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gencitabina
7.
Br J Cancer ; 95(3): 289-97, 2006 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-16868547

RESUMO

The present study was performed to investigate the capability of gemcitabine and pemetrexed to synergistically interact with respect to cytotoxicity and apoptosis in T24 and J82 bladder cancer cells, and to establish a correlation between drug activity and gene expression of selected genes in tumour samples. The interaction between gemcitabine and pemetrexed was synergistic; indeed, pemetrexed favoured gemcitabine cytotoxicity by increasing cellular population in S-phase, reducing Akt phosphorylation as well as by inducing the expression of a major gemcitabine uptake system, the human equilibrative nucleoside transporter-1 (hENT1), and the key activating enzyme deoxycytidine kinase (dCK) in both cell lines. Bladder tumour specimens showed an heterogeneous gene expression pattern and patients with higher levels of dCK and hENT1 had better response. Moreover, human nucleoside concentrative transporter-1 was detectable only in 3/12 patients, two of whom presented a complete response to gemcitabine. These data provide evidence that the chemotherapeutic activity of the combination of gemcitabine and pemetrexed is synergistic against bladder cancer cells in vitro and that the assessment of the expression of genes involved in gemcitabine uptake and activation might be a possible determinant of bladder cancer response and may represent a new tool for treatment optimization.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desoxicitidina/análogos & derivados , Glutamatos/administração & dosagem , Guanina/análogos & derivados , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo , Administração Intravesical , Adulto , Idoso , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Desoxicitidina Quinase/antagonistas & inibidores , Desoxicitidina Quinase/genética , Dipiridamol/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Transportador Equilibrativo 1 de Nucleosídeo/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/genética , Antagonistas de Aminoácidos Excitatórios , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutamatos/farmacologia , Guanina/administração & dosagem , Guanina/antagonistas & inibidores , Guanina/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pemetrexede , Fosforribosilglicinamido Formiltransferase/antagonistas & inibidores , Fosforribosilglicinamido Formiltransferase/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fase S/efeitos dos fármacos , Tioinosina/análogos & derivados , Tioinosina/farmacologia , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/genética , Resultado do Tratamento , Gencitabina
8.
Proc Natl Acad Sci U S A ; 103(16): 6332-7, 2006 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-16601104

RESUMO

Non-small cell lung cancer (NSCLC) demonstrates a strong etiologic association with smoking. Although nicotine is not carcinogenic, it can induce cell proliferation and angiogenesis and suppress apoptosis induced by certain agents. Here we show that nicotine inhibits apoptosis induced by the drugs gemcitabine, cisplatin, and taxol, which are used to treat NSCLCs. This protection correlated with the induction of XIAP and survivin by nicotine in a panel of human NSCLC cell lines, and depletion of XIAP and survivin ablated the protective effects of nicotine. The antiapoptotic effects of nicotine were mediated by dihydro beta-erythroidine-sensitive alpha3-containing nicotinic acetylcholine receptors and required the Akt pathway. Chromatin immunoprecipitation assays demonstrated that nicotine stimulation caused an increased recruitment of E2F1 and concomitant dissociation of retinoblastoma tumor suppressor protein (Rb) from survivin promoter in A549 cells. Moreover, ablation of E2F1 levels caused abrogation of the protective effects of nicotine against cisplatin-induced apoptosis in A549 cells whereas ablation of signal transducer and activator of transcription 3 levels had no effect. These studies suggest that exposure to nicotine might negatively impact the apoptotic potential of chemotherapeutic drugs and that survivin and XIAP play a key role in the antiapoptotic activity of nicotine.


Assuntos
Antineoplásicos/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Nicotina/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Cisplatino/antagonistas & inibidores , Cisplatino/uso terapêutico , Desoxicitidina/análogos & derivados , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/uso terapêutico , Di-Hidro-beta-Eritroidina/farmacologia , Humanos , Proteínas Inibidoras de Apoptose , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/genética , Paclitaxel/antagonistas & inibidores , Paclitaxel/uso terapêutico , Regiões Promotoras Genéticas , Receptores Nicotínicos/efeitos dos fármacos , Receptores Nicotínicos/metabolismo , Proteína do Retinoblastoma/metabolismo , Survivina , Regulação para Cima , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Gencitabina
9.
Anticancer Res ; 25(5): 3347-53, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16101149

RESUMO

BACKGROUND: Gemcitabine is a pyrimidine nucleoside analog that is clinically active against pancreatic cancer. We have recently demonstrated that p38 MAPK is specifically activated by gemcitabine and that pharmacological blockade of p38 MAPK signaling prevented gemcitabine-induced apoptosis in human pancreatic cancer cells. In this study, we further investigated the implication of p38 MAPK in the cytotoxic action of gemcitabine. MATERIALS AND METHODS: Cells expressing a dominant-negative mutant of p38 MAPK were generated. Clonogenic assays were used to assess the long-term effect on cancer cell viability in the human pancreatic cancer cells, PK1 and PCI43. The p38 MAPK activation level was assessed using an antibody specific to the phosphorylated form. RESULTS: Gemcitabine increased the activation level of p38 MAPK in a dose-dependent manner and induced apoptosis in the two tested human pancreatic cancer cell lines. The selective p38 MAPK inhibitors, SB203580 and SB202190, reduced gemcitabine-induced activation of p38 MAPK, prevented the gemcitabine-induced apoptosis and increased long-term clonogenic survival. Overexpression of a dominant-negative p38 mutant in cells resulted in the reduction of gemcitabine-induced p38 MAPK activation and apoptosis, and increases in clonogenic survival. CONCLUSION: These results strongly suggest that the activation of p38 MAPK signaling is necessary for gemcitabine-induced cell death in human pancreatic cancer cells. Based upon these results, we suggest that molecules of p38 MAPK signaling pathways should be listed as novel targets for gemcitabine-based therapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/enzimologia , Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Gencitabina
10.
Biochem Biophys Res Commun ; 316(1): 71-7, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-15003513

RESUMO

In this study, we investigated the involvement of Akt and members of the mitogen-activated protein kinase (MAPK) superfamily, including ERK, JNK, and p38 MAPK, in gemcitabine-induced cytotoxicity in human pancreatic cancer cells. We found that gemcitabine induces apoptosis in PK-1 and PCI-43 human pancreatic cancer cell lines. Gemcitabine specifically activated p38 MAPK in a dose- and time-dependent manner. A selective p38 MAPK inhibitor, SB203580, significantly inhibited gemcitabine-induced apoptosis in both cell lines, suggesting that phosphorylation of p38 MAPK may play a key role in gemcitabine-induced apoptosis in pancreatic cancer cells. A selective JNK inhibitor, SP600125, failed to inhibit gemcitabine-induced apoptosis in both cell lines. MKK3/6, an upstream activator of p38 MAPK, was phosphorylated by gemcitabine, indicating that the MKK3/6-p38 MAPK signaling pathway is indeed involved in gemcitabine-induced apoptosis. Furthermore, gemcitabine-induced cleavage of the caspase substrate poly(ADP-ribose) polymerase was inhibited by pretreatment with SB203580, suggesting that activation of p38 MAPK by gemcitabine induces apoptosis through caspase signaling. These results together suggest that gemcitabine-induced apoptosis in human pancreatic cancer cells is mediated by the MKK3/6-p38 MAPK-caspase signaling pathway. Further, these results lead us to suggest that p38 MAPK should be investigated as a novel molecular target for human pancreatic cancer therapies.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Apoptose , Desoxicitidina/análogos & derivados , Desoxicitidina/toxicidade , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/uso terapêutico , Humanos , Imidazóis/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno , Gencitabina
11.
Cancer Chemother Pharmacol ; 53(6): 459-67, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14752578

RESUMO

PURPOSE: Hematoprotective strategies may offer new approaches to prevent chemotherapy-induced hematotoxicity. The present study was undertaken to investigate the chemoprotective effects of dexamethasone and its optimal dose and the underlying mechanisms. METHODS: Lethal toxicity and hematotoxicity of carboplatin were compared in CD-1 mice with or without dexamethasone pretreatment. Plasma and tissue pharmacokinetics of carboplatin were determined in CD-1 mice. Carboplatin was quantified by HPLC. Gemcitabine was analyzed by radioactivity counting. RESULTS: Pretreatment with dexamethasone prevented lethal toxicity of carboplatin in a dose- and schedule-dependent manner. The best protective effects of dexamethasone pretreatment as measured by survival were observed at the dose level of 0.1 mg/mouse per day for 5 days (80% vs 10% in controls). In contrast, posttreatment with dexamethasone had no protective effects. Pretreatment with dexamethasone significantly prevented the decrease in granulocyte counts. To elucidate the mechanisms by which dexamethasone pretreatment reduces hematotoxicity, we examined the effects of dexamethasone pretreatment on the pharmacokinetics of carboplatin and gemcitabine in CD-1 mice. No significant differences in plasma pharmacokinetics of carboplatin or gemcitabine were observed between control and mice pretreated with dexamethasone. However, dexamethasone pretreatment significantly decreased carboplatin and gemcitabine uptake in spleen and bone marrow with significant decreases in AUC, T(1/2), and C(max), and an increase in CL. CONCLUSIONS: To our knowledge, this is the first time that dexamethasone has been shown to significantly decrease host tissue uptake of chemotherapeutic agents, suggesting a mechanism responsible for the chemoprotective effects of dexamethasone. This study provides a basis for future study to evaluate dexamethasone as a chemoprotectant in cancer patients.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Medula Óssea/efeitos dos fármacos , Carboplatina/farmacocinética , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Animais , Área Sob a Curva , Medula Óssea/patologia , Carboplatina/antagonistas & inibidores , Desoxicitidina/administração & dosagem , Desoxicitidina/antagonistas & inibidores , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Glucocorticoides/administração & dosagem , Injeções Intravenosas , Injeções Subcutâneas , Longevidade/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Gencitabina
12.
Stem Cells ; 21(3): 266-71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12743321

RESUMO

In phase I and II clinical trials, the deoxycytidine analogue 2',2' difluorodeoxycytidine (dFdC, gemcitabine) has shown promising antitumor activity in leukemia as well as in solid tumors. Preclinical and clinical studies of gemcitabine suggested that myelosuppression was the dose-limiting toxicity. The present investigations were designed to test the effect of continuously administered gemcitabine on the in vitro clonal growth of normal CD34(+) cells isolated from peripheral blood and the promyelocytic cell line, HL-60. For this purpose, CD34(+) and HL-60 cells were cultured in methylcellulose in the continuous presence of 0.1-16 nM of gemcitabine. The results show a dose-dependent inhibition of colony growth of normal as well as leukemic cells. However, HL-60 cells were up to 12-fold more sensitive towards gemcitabine than normal progenitors. For rescue experiments, the natural pyrimidine deoxycytidine (dCyd) was added to CD34(+) and HL-60 cells simultaneously or with delay. Coadministration of 1mM dCyd to separate cultures resulted in complete restoration of colony formation capacity of CD34(+) and HL-60 cells. Delayed addition of 1 mM dCyd after 48 and 72 hours recovered up to 90% and 40%, respectively, of stem cell proliferation, whereas HL-60 cells remained substantially inhibited (4.5% +/- 3.5% versus 0%). Delayed addition after 48 and 72 hours protected about 80% and 50%, respectively, of myelopoietic and erythropoetic colony formation, whereas colony formation obtained from HL-60 cells remained significantly inhibited (9.6% +/- 4.17% versus 0%). These in vitro data suggest that there is a marked difference in the susceptibility of leukemic and normal CD34(+) cells to gemcitabine and that delayed administration of dCyd may further reduce the bone marrow cytotoxicity of gemcitabine without impairing its antitumor effect.


Assuntos
Antígenos CD34/biossíntese , Antineoplásicos/antagonistas & inibidores , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/toxicidade , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Antígenos CD34/imunologia , Antineoplásicos/toxicidade , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Técnicas de Cocultura , Citotoxinas/antagonistas & inibidores , Citotoxinas/farmacologia , Desoxicitidina/antagonistas & inibidores , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/imunologia , Quimioterapia Combinada , Células HL-60 , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunossupressores/antagonistas & inibidores , Imunossupressores/toxicidade , Leucemia/imunologia , Leucemia/fisiopatologia , Linfócitos T/imunologia , Gencitabina
13.
Oncology ; 61 Suppl 2: 50-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11694788

RESUMO

Preclinical data indicate that trastuzumab (Herceptin) has the potential for synergistic or additive effects in combination with therapies including chemotherapy and hormonal agents, providing the rationale for a number of clinical trials in women with HER2-positive metastatic breast cancer. A recently reported phase II trial has demonstrated that trastuzumab plus vinorelbine is both effective (overall response rate 75%) and well tolerated, with the major side effects being typical of single-agent vinorelbine. Other combinations of trastuzumab with a variety of other chemotherapeutic and hormonal agents are also being assessed. In an effort to overcome the cardiotoxicity observed with trastuzumab plus doxorubicin in the pivotal phase III trial, combination regimens involving potentially less toxic anthracyclines such as epirubicin and liposomal formulations of doxorubicin are ongoing. In addition, trials are investigating whether trastuzumab can reverse the resistance to hormonal therapy that develops in most women with metastatic breast cancer. These and other studies will identify the regimens that produce the best outcomes with the fewest possible side effects in women with HER2-positive breast cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Desoxicitidina/análogos & derivados , Vimblastina/análogos & derivados , Anastrozol , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais Humanizados , Antineoplásicos Hormonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Ciclofosfamida/administração & dosagem , Ciclofosfamida/farmacologia , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Interações Medicamentosas , Sinergismo Farmacológico , Epirubicina/administração & dosagem , Epirubicina/farmacologia , Feminino , Fluoruracila/antagonistas & inibidores , Fluoruracila/farmacologia , Cardiopatias/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Humanos , Metotrexato/administração & dosagem , Metotrexato/antagonistas & inibidores , Metástase Neoplásica , Nitrilas/administração & dosagem , Compostos Organoplatínicos/administração & dosagem , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Tamoxifeno/administração & dosagem , Tiotepa/administração & dosagem , Tiotepa/farmacologia , Trastuzumab , Resultado do Tratamento , Triazóis/administração & dosagem , Vimblastina/administração & dosagem , Vimblastina/farmacologia , Vinorelbina , Gencitabina
14.
Cancer Res ; 37(6): 1636-9, 1977 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-66984

RESUMO

The in vitro effect of 5-aza-2'-deoxycytidine (5-aza-CdR) on cytotoxicity and macromolecular synthesis in A(T1)C1-3 hamster fibrosarcoma cells was investigated. The in vitro concentrations that produce 50% cell kill for 5-aza-CdR were about 1.0 and 0.01 microng/ml for a 2- and 24-hr exposure, respectively. 5-aza-CdR inhibited the growth of the fibrosarcoma cells by 40% at a concentration of 0.05 microng/ml. Deoxycytidine, but not cytidine, was a potent antagonist of the cytotoxicity produced by 5-aza-CdR. At cytotoxic concentrations 5-aza-CdR did not appear to inhibit DNA, RNA, or protein synthesis during a 1-hr incubation as measured by the incorporation of radioactive thymidine, uridine,, or leucine into acid-insoluble material. At a concentration of 10 microng/ml, 5-aza-CdR stimulated the incorporation of radioactive thymidine into DNA by more than 50%. These results indicate that 5-aza-CdR is a very potent cytotoxic agent to tumor cells in vitro at concentrations that do not inhibit macromolecular synthesis.


Assuntos
Azacitidina/análogos & derivados , Desoxicitidina/análogos & derivados , Fibrossarcoma/tratamento farmacológico , Azacitidina/antagonistas & inibidores , Azacitidina/farmacologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citidina/farmacologia , DNA de Neoplasias/biossíntese , Desoxicitidina/antagonistas & inibidores , Desoxicitidina/farmacologia , Fibrossarcoma/metabolismo , Proteínas de Neoplasias/biossíntese , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , RNA Neoplásico/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...