Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Pharm Bull ; 37(4): 688-93, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24694617

RESUMO

Endothelial progenitor cells (EPC) can differentiate into both endothelial cells and contractile smooth muscle cells (SMC). Previously we reported that TR-BME2 cells, a model for EPC, developed contractile SMC-like characteristics in culture medium deprived of endothelial cell growth factors (ECGF). The aim of the present study was to clarify the effect of one of these factors, basic fibroblast growth factor (bFGF) on differentiation of EPC. First it was confirmed that bFGF receptor (FGFR-1) mRNA is expressed in TR-BME2 cultured in both ECGF-rich and ECGF-deprived medium. When TR-BME2 cells were cultured in ECGF-deprived medium, they differentiated into contractile SMC. Expression of an undifferentiated state marker, CD133, and proliferation of TR-BME2 were both reduced by ECGF deprivation, but these changes were diminished in the presence of bFGF. mRNA expression of smooth muscle α-actin (SMA) and smooth muscle protein 22 (SM22), which are contractile SMC markers, was induced by deprivation of ECGF and the induction was suppressed by bFGF. In vascular endothelial cell growth factor (VEGF)-induced tube formation assay, TR-BME2 cells formed tube structures in the presence of bFGF, but not in its absence. Our results indicate that bFGF is essential for the maintenance of EPC phenotype, serving to suppress differentiation to contractile SMC.


Assuntos
Actinas/biossíntese , Diferenciação Celular/fisiologia , Células Progenitoras Endoteliais/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Fenótipo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Proliferação de Células , Meios de Cultura/farmacologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Ratos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/biossíntese , Fator A de Crescimento do Endotélio Vascular/fisiologia
2.
Expert Opin Ther Targets ; 16 Suppl 1: S31-5, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22313367

RESUMO

INTRODUCTION: Carcinogenesis is considered to be a multistep process that may involve cumulative genomic alterations. Loss of chromosomal material would inactivate tumor suppressor genes and gain of chromosomal material has the potential to activate tumor-promoting genes. AREAS COVERED: Recent intensive studies by array comparative genomic hybridization (aCGH) have demonstrated frequent alterations in multiple regions of the genome. This suggests that these regions contain a variety of oncogenes and tumor suppressor genes associated with breast cancer development. The patterns of copy number variations (CNVs) have been suggested to be associated with breast cancer subtypes, indicating the importance of genomic instability in the development of breast cancer. EXPERT OPINION: To further clarify the complexity of gene alterations, one approach is to employ a CNV-targeted platform that harbors a large number of direct CNV markers located in the repeat-rich unstable regions of the human genome. Next generation sequencing is another approach to overcome the limitations of aCGH such as the repeat-rich regions. Genomic analysis should be combined with expression analysis to elucidate individual genes relevant to breast cancer development and progression. The elucidation of the functions of the affected genes would lead to identification of new molecular targets for breast cancer eradication.


Assuntos
Neoplasias da Mama/genética , Variações do Número de Cópias de DNA , Aberrações Cromossômicas , Cromossomos Humanos Par 6 , Hibridização Genômica Comparativa , Feminino , Estudo de Associação Genômica Ampla , Humanos
3.
Oncology ; 77(3-4): 182-91, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19729975

RESUMO

OBJECTIVE: Bone marrow (BM)-derived endothelial progenitor cells (EPC) play an important role in neovascularization and tumor growth. It has been reported that docetaxel and paclitaxel inhibit angiogenesis, but the effect of docetaxel and paclitaxel on EPC-induced neovascularization has not been examined. We aimed to clarify the cytotoxic and inhibitory effects of these drugs on EPC. METHODS: The effects of drugs on growth, tube formation, and migration of EPC were analyzed in vitro using a rat BM-derived EPC cell line (TR-BME). Fluorescence-labeled TR-BME cells were injected into tumor-bearing rats and accumulation at the tumor site was analyzed by fluorescence-activated cell sorting (FACS). RESULTS: In in vitro cytotoxicity assays of these drugs in TR-BME, rat endothelial cell line TR-BBB and rat tumor cell line Walker 256, the IC(50) values for TR-BME were higher than those for TR-BBB or Walker 256. Both drugs inhibited tube formation and migration of TR-BME at lower concentrations than the cytotoxic IC(50). In vivo studies showed that a low dose of both drugs inhibited EPC accumulation at the tumor site in tumor-bearing rats, as determined by FACS, and caused a decrease in microvessel density. CONCLUSION: Docetaxel and paclitaxel directly inhibited EPC-initiated vasculogenesis at low (non-cytotoxic) concentrations, causing suppression of tumor growth.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Células Endoteliais/efeitos dos fármacos , Paclitaxel/farmacologia , Células-Tronco/efeitos dos fármacos , Taxoides/farmacologia , Animais , Carcinoma 256 de Walker/tratamento farmacológico , Linhagem Celular , Inibição de Migração Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Docetaxel , Células Endoteliais/citologia , Feminino , Humanos , Neovascularização Patológica/tratamento farmacológico , Ratos , Ratos Wistar
4.
Drug Metab Pharmacokinet ; 22(4): 255-66, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17827780

RESUMO

The basement membrane at the blood-brain barrier (BBB) plays important roles in maintaining the structure and function of capillary vessels. The BBB is constructed from endothelial cells, astrocytes and pericytes, but their interactions in the formation or maintenance of basement membrane have not been established. Transforming growth factor-beta1 (TGF-beta1) is known to increase fibronectin in brain capillary basement membrane with deposition of beta-amyloid. We previously reported that the mRNA level of alpha-smooth muscle actin in a brain capillary pericyte cell line TR-PCT1 was increased by treatment with TGF-beta1. In this study, expression of mRNAs encoding basement membrane-related molecules in TR-PCT1, a rat endothelial cell line TR-BBB13, and a type 2 astrocyte cell line TR-AST4 was evaluated by RT-PCR. The effects of TGF-beta1 on expression of basement membrane-related genes in these cell lines were also examined. Fibronectin, MMP-9, tPA, TIMP-1, and PAI-l in TR-PCT1 were higher than in TR-BBB13 and TR-AST4. In TR-PCT1 treated with TGF-beta1, collagen type IV, PAI-1, and MMP-9 were increased, and TIMP-2 was reduced. The change in PAI-1 mRNA was faster than those in MMP-9, TIMP-2, collagen type IV mRNAs. These results suggest that pericytes may be key cells in the maintenance of the basement membrane at the BBB.


Assuntos
Membrana Basal/metabolismo , Química Encefálica/efeitos dos fármacos , Encéfalo/citologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/fisiologia , Membrana Basal/efeitos dos fármacos , Membrana Basal/enzimologia , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Expressão Gênica/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Pericitos/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Pancreatology ; 7(2-3): 167-73, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17592230

RESUMO

BACKGROUND/AIM: Local treatment often fails in patients with resectable pancreatic cancer due to the postoperative development of distant metastases, especially liver metastases. We determined the prognostic factors for postoperative liver metastases in pancreatic cancer patients following surgical resection with combined radiotherapy. METHODS: Sixty-four patients with nonmetastatic, resectable pancreatic cancer were entered into this study. All of these patients had pancreatic resection surgery combined with radiotherapy. The development of postoperative liver metastases was carefully followed, and the survival ratio was evaluated using the Kaplan-Meier method. The prognostic importance of clinicopathological factors and molecular characteristics was analyzed by the Cox proportional hazards model. The correlation study was performed using Fisher's exact test. RESULTS: Tumor size, curability, and histological type of differentiation were statistically significant independent prognostic factors. On multivariate analysis, curability and histological type of differentiation were statistically significant. Only tumor size (> or = 3 cm) was significantly correlated with postoperative liver metastases, as well as cyclooxygenase-2 expression. CONCLUSIONS: There were three significant prognostic factors in patients with resectable pancreatic cancer who had local therapy. Patients who have a large tumor require particularly careful follow-up for postoperative liver metastases.


Assuntos
Carcinoma Ductal Pancreático/secundário , Ciclo-Oxigenase 2/metabolismo , Neoplasias Hepáticas/secundário , Proteínas de Membrana/metabolismo , Neoplasias Pancreáticas/patologia , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/enzimologia , Carcinoma Ductal Pancreático/mortalidade , Carcinoma Ductal Pancreático/cirurgia , Feminino , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/cirurgia , Prognóstico , Taxa de Sobrevida
6.
Clin Cancer Res ; 13(6): 1875-82, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363544

RESUMO

PURPOSE: Removal of fucose residues from the oligosaccharides of human antibody is a powerful approach to enhance antibody-dependent cellular cytotoxicity (ADCC), a potential important antitumor mechanism of therapeutic antibodies. To provide clinically relevant evidence of this mechanism, we investigated ADCC of a fucose-negative version of trastuzumab [anti-human epidermal growth factor receptor 2 (HER2) humanized antibody] using peripheral blood mononuclear cells (PBMC) from breast cancer patients as effector cells. EXPERIMENTAL DESIGN: Thirty volunteers, including 20 breast cancer patients and 10 normal healthy control donors, were recruited randomly, and aliquots of peripheral blood were collected. ADCC of commercial trastuzumab (fucosylated) and its fucose-negative version were measured using PBMCs drawn from the volunteers as effector cells and two breast cancer cell lines with different HER2 expression levels as target cells. Relationships between cytotoxicity and characteristics of the patients, such as content of natural killer cells in PBMCs, type of therapy, FCGR3A genotypes, etc. were also analyzed. RESULTS: ADCC was significantly enhanced with the fucose-negative antibody compared with the fucose-positive antibody using PBMCs from either normal donors or breast cancer patients. Enhancement of ADCC was observed irrespective of the various clinical backgrounds of the patients, even in the chemotherapy cohort that presented with a reduced number of natural killer cells and weaker ADCC. CONCLUSIONS: This preliminary study suggests that the use of fucose-negative antibodies may improve the therapeutic effects of anti-HER2 therapy for patients independent of clinical backgrounds.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Neoplasias da Mama/imunologia , Fucose/química , Receptor ErbB-2/imunologia , Adulto , Idoso , Animais , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Humanos , Imunoterapia , Leucócitos/efeitos dos fármacos , Pessoa de Meia-Idade , Receptores de IgG/genética , Trastuzumab
7.
Clin Cancer Res ; 12(1): 264-72, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397051

RESUMO

Cyclooxygenase (COX)-2 inhibition results in tumor regression; however, little is known about the mechanism. In the present study, using a Walker256 tumor model and a rat bone marrow-derived endothelial cell line TR-BME-2, we analyzed the effects of a new selective COX-2 inhibitor, 5,5-dimethyl-3-(3-fluorophenyl)-4-(4-methylsulphonyl)phenyl-2-(5H)-furanone (DFU), on the production of chemokines and growth factors and on the neovascularization. The oral administration of DFU (5 mg/kg/d) significantly suppressed the tumor growth with decreasing microvessel density in vivo, although it showed no direct inhibition of Walker256 cell proliferation in vitro. It was newly found that the recruitment of systemically injected TR-BME-2 cells into the tumor site was significantly inhibited by DFU treatment. In addition, we found that DFU significantly reduced the production of monocyte chemoattractant protein-1 (MCP-1) both in tumor tissues and in the systemic circulation (P < 0.001 and P < 0.001, respectively). Such reduction was not observed in other chemotactic factors, vascular endothelial growth factor and stromal cell-derived factor-1. The induced chemotaxis of TR-BME-2 by serum of tumor-bearing rats was significantly reduced in DFU-treated rat serum, although DFU showed no direct inhibition for TR-BME-2 cells, either cell growth or chemotaxis. Treatment with neutralizing antibodies to soluble mediators, including MCP-1, significantly suppressed the chemotaxis. Regarding the down-regulation machinery of MCP-1 production in vivo, tumor-associated macrophages seem to play crucial roles, because DFU eliminated MCP-1 production in the activated macrophages remarkably but not in Walker256 tumor cells in vitro. In conclusion, COX-2 inhibitor DFU exerts tumor regression activity in a Walker256 tumor model by suppressing MCP-1 production in tumor tissues and in the circulation.


Assuntos
Quimiocina CCL2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Furanos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL2/efeitos dos fármacos , Quimiocinas/metabolismo , Quimiotaxia/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Neoplasias Experimentais/irrigação sanguínea , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Int J Oncol ; 27(5): 1247-55, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16211219

RESUMO

The effects of the nuclear factor (NF)-kappaB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), combined with tumor necrosis factor (TNF)-alpha were evaluated in PK-8 pancreatic cancer cells. NF-kappaB was activated by TNF-alpha; however, the administration of DHMEQ abrogated its transcriptional activity. The addition of DHMEQ to TNF-alpha markedly induced apoptosis in PK-8 cells with down-regulation of anti-apoptotic c-FLIP and survivin. Combined treatment significantly suppressed cell viability in vitro, and the anti-tumor effect of DHMEQ was also significant in vivo. We investigated the apoptosis signaling pathway involved in these cell killing effects. Truncated Bid was produced by activated caspase-8, and the subsequent depolarization of the mitochondrial membrane potential (Delta Psi m) peaked at 6 h. Then, the activity of caspase-3 was up-regulated 8-fold. Z-VAD-fmk (a pan-caspase inhibitor) perfectly inhibited the up-regulation of caspase-3 but failed to reverse the cell viability. The above findings indicated that the growth inhibitory effect of combined treatment largely depended on mitochondria-associated caspase-independent apoptosis. The intracellular behavior of apoptosis-inducing factor (AIF) following depolarization of Delta Psi m suggested that AIF executed such a caspase-independent apoptosis. Interestingly, caspase-dependent apoptosis appeared within 6 h, whereas the caspase-independent apoptosis lagged. Thus, the addition of DHMEQ to TNF-alpha was capable of inducing caspase-independent apoptosis in pancreatic cancer cells. Once caspase-independent apoptosis was induced, the apoptosis demonstrated powerful cytotoxicity. Therefore, DHMEQ in combination with TNF-alpha may be a promising treatment for pancreatic cancer.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Caspases/metabolismo , Cicloexanonas/farmacologia , Neoplasias Pancreáticas/patologia , Fator de Necrose Tumoral alfa/fisiologia , Caspase 3 , Caspases/biossíntese , Sobrevivência Celular , Interações Medicamentosas , Humanos , Cinética , Mitocôndrias/enzimologia , Células Tumorais Cultivadas , Regulação para Cima
9.
Clin Cancer Res ; 11(3): 1287-93, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15709200

RESUMO

We previously designed and synthesized the new nuclear factor kappaB (NF-kappaB) inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) derived from the structure of the antibiotic epoxyquinomicin C. We looked into the effect of DHMEQ on cellular phenotypes and tumor growth in mice injected with human breast carcinoma cell line MDA-MB-231 or MCF-7. In estrogen-independent breast adenocarcinoma cell line MDA-MB-231, NF-kappaB is constitutively activated. The addition of DHMEQ (10 microg/mL) completely inhibited the activated NF-kappaB for at least 8 hours. On the other hand, NF-kappaB is not activated in estrogen-dependent MCF-7 cells. In this cell line, DHMEQ completely inhibited the tumor necrosis factor-alpha-induced activation of NF-kappaB. DHMEQ did not inhibit the degradation of IkappaB but inhibited the nuclear translocation of NF-kappaB by both p65/p50 and RelB/p52 pathways. MDA-MB-231 cells secrete interleukin (IL)-6 and IL-8 without stimulation, and DHMEQ decreased the secretion levels of both cytokines. When MDA-MB-231 or MCF-7 cells were stimulated by tumor necrosis factor-alpha, the inhibitory effects of DHMEQ were still maintained. I.p. administration of DHMEQ (thrice a week) significantly inhibited the tumor growth of MDA-MB-231 (12 mg/kg) or MCF-7 (4 mg/kg) in severe combined immunodeficiency mice. No toxicity was observed during the experiment, including the loss of body weight. An immunohistological study on resected MCF-7 tumors showed that DHMEQ inhibited angiogenesis and promoted apoptosis. Furthermore, in Adriamycin-resistant MCF-7 cells highly expressing multidrug resistance gene-1, DHMEQ also exhibited the above capability, including down-regulation of IL-8. Thus, DHMEQ might be a potent drug for the treatment of various breast carcinomas by inhibiting the NF-kappaB activity.


Assuntos
Benzamidas/farmacologia , Neoplasias da Mama/prevenção & controle , Cicloexanonas/farmacologia , NF-kappa B/metabolismo , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Benzamidas/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cicloexanonas/uso terapêutico , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Feminino , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Linfotoxina-alfa/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , NF-kappa B/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncol Rep ; 12(2): 281-5, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254689

RESUMO

Cyclooxygenase-2 (COX-2) inhibitors are regarded as potentially important in strategies for cancer treatment. however, the precise mechanisms of these anti-inflammatory drugs as anti-cancer therapy are still unknown. In this study, we examined the effect of DFU both in vitro on MCF-7 cell growth, as well as in vivo on tumor growth produced by MCF-7 cell injection in mice. DFU has growth inhibitory effects on tumor growth in mice compared to the control group. We examined the tumor tissues for apoptosis and angiogenesis by immunostaining. Apoptosis was detected only in the treatment group. DFU treatment also resulted in the inhibition of angiogenesis, as well as decreased COX-2 expression. Results of this study suggest that inhibitory effects of DFU might be COX-2 dependent.


Assuntos
Inibidores Enzimáticos/farmacologia , Furanos/farmacologia , Isoenzimas/antagonistas & inibidores , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Ciclo-Oxigenase 2 , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Prostaglandina-Endoperóxido Sintases , Fatores de Tempo
11.
Oncol Rep ; 10(6): 1765-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14534693

RESUMO

To test the hypothesis that genetically modified bone marrow-derived endothelial progenitor cells (EPCs) can be effective carriers of therapeutic agents to tumor sites, we utilized our conditionally immortalized endothelial progenitor cell line, TR-BME-2. In the syngenic rat, systemically injected TR-BME-2 cells were immediately distributed to the organs (lung, bone marrow, peripheral blood, liver, spleen). Trapped cells were cleared within 4 days, but selective accumulation in the Walker256 tumor was maintained for over 4 days. The tumor growth was enhanced by administration of TR-BME-2 cells. It is suggested that accumulated TR-BME-2 differentiated to tumor vasculature, increased the tumor blood supply, and thereby increased the tumor volume. We conducted IL-12 gene transfection of TR-BME-2 cells with a virus vector in vitro, and used the resultant IL-12-secreting TR-BME-2 to deliver IL-12, which strongly activates cytotoxic lymphocytes and natural killer cells, to the tumor site in vivo. However, the tumor-progressive character of TR-BME-2 offset the anti-tumor effect of IL-12. Nevertheless, our results suggest that gene-transfected EPCs could be useful as a tumor-specific drug delivery system, especially if the tumor vasculature-promoting effect of EPCs can be blocked.


Assuntos
Endotélio Vascular/metabolismo , Terapia Genética/métodos , Interleucina-12/genética , Neoplasias/terapia , Células-Tronco/metabolismo , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Progressão da Doença , Corantes Fluorescentes/farmacologia , Interleucina-12/metabolismo , Microscopia de Fluorescência , Transplante de Neoplasias , Compostos Orgânicos/farmacologia , Ratos , Fatores de Tempo , Distribuição Tecidual , Transfecção
12.
Oncol Rep ; 10(5): 1201-6, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12883681

RESUMO

Pancreatic cancer is a solid malignancy with the poor prognosis largely due to frequent and lethal liver metastases. The combination of immunotherapy and anti-angiogenesis therapy might be a hopeful strategy for the treatment of distant metastases. The benefits of the combination therapy by an immune stimulator alpha-galactosylceramide (KRN7000) and an angiogenesis inhibitor AGM-1470 (TNP470) were evaluated on the hamster highly aggressive liver metastasis model using the syngeneic pancreatic cancer cell line HPD-NR. KRN7000 immediately activated hepatic mono-nuclear cells to produce IFN-gamma in vitro. Intraportal injection of KRN7000 exhibited a dense accumulation of CD4-CD8- natural killer T cells, around the liver metastases in vivo. KRN7000 treatment significantly inhibited the growth of liver metastases, and importantly, significant survival prolongation was confirmed when TNP470 treatment was added to it. Furthermore, cytotoxic T lymphocytes were induced at the sites of a few residual metastases in the liver of a long-term survivor. Thus, the combination of KRN7000 and TNP470 showed a high effectiveness for the treatment of liver metastases of pancreatic cancer.


Assuntos
Galactosilceramidas/farmacologia , Neoplasias Hepáticas/patologia , Neoplasias Pancreáticas/patologia , Sesquiterpenos/farmacologia , Adjuvantes Imunológicos/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Divisão Celular , Cricetinae , Cicloexanos , Modelos Animais de Doenças , Imunoterapia/métodos , Interferon gama/metabolismo , Fígado/metabolismo , Neoplasias Hepáticas/secundário , Masculino , Mesocricetus , Metástase Neoplásica , O-(Cloroacetilcarbamoil)fumagilol , Neoplasias Pancreáticas/complicações , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
13.
Oncol Rep ; 10(5): 1213-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12883683

RESUMO

Peripheral stem cells released from bone marrow (BM) are known to be incorporated into foci of neovascularization and to contribute to solid tumor development. In the present rat Walker256 tumor model, BM suppression induced by total body irradiation resulted in poor growth of the tumor with apparently poor vascularization, and BM transplantation restored tumor growth. Endothelial progenitor cells are considered to be crucial for vasculogenesis, but they are not yet well defined, and methodology for their purification has not been established. As a model to examine the significance of endothelial progenitor cells in tumor-specific vasculogenesis, we utilized an immortalized rat BM-derived endothelial cell line named TR-BME-2. Fluorescence-labeled TR-BME-2 cells injected systemically into rats were accumulated at the tumor site 4 days later. Another rat BM-derived cell line named C2-11, which does not have an endothelial profile, did not show tumor-specific accumulation. The tumor volume in rats treated with TR-BME-2 was significantly larger than that in rats treated with C2-11. Thus, our results suggest the importance of neovascularization by bone marrow-derived endothelial cells for the promotion of tumor growth.


Assuntos
Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Neoplasias Experimentais/patologia , Animais , Animais Geneticamente Modificados , Transplante de Medula Óssea , Divisão Celular , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Separação Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
14.
Oncol Rep ; 10(4): 791-6, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12792725

RESUMO

We examined, by immunohistochemical analysis, the expression of aromatase and estrone sulfatase (E1-STS) which are the two major enzymes involved in in situ estrogen synthesis in breast cancer tissue. In the 83 cases examined, E1-STS, which hydrolyses estrone sulfate (E1S) to estrone (E1), was dominantly detected in tumor cells in 43 (59.0%) cases. Aromatase, which converts androgens to estrogens, was dominantly detected in stromal cells of the tumor. It was detected in 39 (47.0%) of the 83 cases. There was no significant correlation between the expression of these two enzymes and clinicopathological factors. We found a tendency for a correlation between aromatase expression and E1-STS expression in breast tumor (p=0.075). In terms of the possible use of these enzymes as prognostic indicators, patients who had aromatase in their tumor showed longer relapse-free survival than those lacking aromatase (p=0.045). Significant correlations between the expression of aromatase and the angiogenesis regulators, vascular endothelial growth factor and thymidine phosphorylase, were found (p=0.047 and p=0.046, respectively), though the presence of aromatase did not correlate with intratumoral microvessel density. This may indicate that aromatase is involved in vascular permeability and recruitment of endothelial cells rather than neovascularization. It may be useful to study the expression of aromatase and E1-STS using immunocytochemical analysis for understanding the tumor characteristics in breast cancer.


Assuntos
Aromatase/metabolismo , Biomarcadores Tumorais/análise , Neoplasias da Mama/enzimologia , Carcinoma Ductal/enzimologia , Proteínas Nucleares , Sulfatases/metabolismo , Adulto , Neoplasias da Mama/patologia , Carcinoma Ductal/patologia , Feminino , Humanos , Menopausa , Microcirculação , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Prognóstico , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Receptores de Estrogênio/análise , Células Estromais/patologia , Taxa de Sobrevida , Timidina Fosforilase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Jpn J Cancer Res ; 93(4): 389-96, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11985788

RESUMO

Macrophage migration inhibitory factor (MIF) is known to exert pleiotropic functions including inhibition of macrophage migration, anchoring, and counteraction of the anti-inflammatory and immunosuppressive activity of glucocorticoids. Ninety-three primary breast cancer tissues and 64 sera of primary breast cancer patients were analyzed for the expression of MIF. The clinico-pathological significance of MIF expression was evaluated. It was found that MIF was frequently over-expressed in primary breast cancer tissues. RT-PCR and western blotting analysis confirmed that wild-type MIF is expressed, and immunohistochemical analysis showed that MIF expression was localized at tumor cells as well as stromal cells, including tumor-associated macrophages. Intratumoral MIF protein concentrations detected by enzyme-linked immunosorbent assay (ELISA) varied with a median value of 1821 ng/mg protein (range: 8 - 8126 ng/mg protein), and correlated inversely with nodal involvement (P = 0.039). No significant correlation was observed with other clinico-pathological factors including tumor size, menopausal status and hormone receptors. The circulating level of MIF protein ranged up to 105.7 ng/ml (median: 17.3 ng/ml), and it was also found to correlate inversely with the number of involved nodes (P = 0.02). A comparative study with other soluble inflammatory mediators showed that intratumoral levels of MIF were significantly associated with those of interleukin-1 beta, suggesting that interactions between tumor cells and tumor-associated macrophages play an important role in the up-regulation of MIF. The multifunctional inflammatory/immune mediator MIF was frequently expressed in primary breast cancer, and its expression level was inversely associated with nodal spread. Thus, MIF seems to play a role in tumor-stroma interactions of primary breast cancers, particularly those with a phenotype of node-negative or minimal nodal spread.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Metástase Linfática , Fatores Inibidores da Migração de Macrófagos/biossíntese , Fatores Inibidores da Migração de Macrófagos/genética , Neovascularização Patológica , Adulto , Idoso , Western Blotting , Neoplasias da Mama/genética , Células Cultivadas , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Interleucina-1/metabolismo , Macrófagos/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Fenótipo , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
16.
Int J Cancer ; 98(1): 14-8, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11857378

RESUMO

Angiogenesis, the formation of new blood vessels, is controlled by a balance between positive and negative endothelial regulatory factors. Soluble vascular endothelial growth factor receptor-1 (sVEGFR1), a naturally occurring soluble form of VEGFR1, is a negative counterpart of the vascular endothelial growth factor (VEGF) signaling pathway, which has been characterized as one of the most important endothelial regulators in human tumor angiogenesis. In our study, we examined the expression of sVEGFR1 in 110 primary breast carcinomas, and assessed its clinical significance. Ninety-four of 110 tumors showed > or = 0.1 ng/mg protein of sVEGFR1 (range:0. 1-6.9 ng/mg protein; median: 1.03 ng/mg protein) as determined by a specific enzyme-linked immunosorbent assay (ELISA). Immunoblot analysis confirmed the presence of sVEGFR1 in breast tumor tissues. The levels of sVEGFR1 were correlated significantly with the levels of VEGF. There was no significant correlation between the levels of sVEGFR1 and any clinico-pathological factors including age, menopause, nodal involvement and hormone receptor status. A univariate prognosis analysis showed that the intratumoral VEGF status, as determined by ELISA, was a significant prognostic indicator, but sVEGFR1 status was not. In the combined analysis, however, the ratio of sVEGFR1 and VEGF levels provided more statistically significant prognostic value than VEGF status alone. Tumors in which the sVEGFR1 levels exceeded VEGF levels 10-fold had a markedly favorable prognosis. Multivariate analysis also demonstrated that the ratio of sVEGFR1 and VEGF was an independent prognostic indicator after nodal status. In conclusion, sVEGFR1, an intrinsic inhibitor of VEGF, frequently co-expressed with VEGF in primary breast cancer tissues. The intratumoral balance between sVEGFR1 and VEGF levels might be crucial for the progression of breast cancer.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Fatores de Crescimento Endotelial/análise , Linfocinas/análise , Proteínas Proto-Oncogênicas/análise , Receptores Proteína Tirosina Quinases/análise , Inibidores da Angiogênese/uso terapêutico , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Immunoblotting , Prognóstico , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...