Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 353
Filtrar
1.
Breast Cancer Res Treat ; 176(2): 271-289, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31006104

RESUMO

PURPOSE: Primary breast and prostate cancers can be cured, but metastatic disease cannot. Identifying cell factors that predict metastatic potential could guide both prognosis and treatment. METHODS: We used Cell-SELEX to screen an RNA aptamer library for differential binding to prostate cancer cell lines with high vs. low metastatic potential. Mass spectroscopy, immunoblot, and immunohistochemistry were used to identify and validate aptamer targets. Aptamer properties were tested in vitro, in xenograft models, and in clinical biopsies. Gene expression datasets were queried for target associations in cancer. RESULTS: We identified a novel aptamer (Apt63) that binds to the beta subunit of F1Fo ATP synthase (ATP5B), present on the plasma membrane of certain normal and cancer cells. Apt63 bound to plasma membranes of multiple aggressive breast and prostate cell lines, but not to normal breast and prostate epithelial cells, and weakly or not at all to non-metastasizing cancer cells; binding led to rapid cell death. A single intravenous injection of Apt63 induced rapid, tumor cell-selective binding and cytotoxicity in MDA-MB-231 xenograft tumors, associated with endonuclease G nuclear translocation and DNA fragmentation. Apt63 was not toxic to non-transformed epithelial cells in vitro or adjacent normal tissue in vivo. In breast cancer tissue arrays, plasma membrane staining with Apt63 correlated with tumor stage (p < 0.0001, n = 416) and was independent of other cancer markers. Across multiple datasets, ATP5B expression was significantly increased relative to normal tissue, and negatively correlated with metastasis-free (p = 0.0063, 0.00039, respectively) and overall (p = 0.050, 0.0198) survival. CONCLUSION: Ecto-ATP5B binding by Apt63 may disrupt an essential survival mechanism in a subset of tumors with high metastatic potential, and defines a novel category of cancers with potential vulnerability to ATP5B-targeted therapy. Apt63 is a unique tool for elucidating the function of surface ATP synthase, and potentially for predicting and treating metastatic breast and prostate cancer.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Neoplasias da Mama/patologia , Membrana Celular/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Neoplasias da Próstata/patologia , Administração Intravenosa , Animais , Aptâmeros de Nucleotídeos/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Detecção Precoce de Câncer , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Masculino , Camundongos , ATPases Mitocondriais Próton-Translocadoras/antagonistas & inibidores , Estadiamento de Neoplasias , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Técnica de Seleção de Aptâmeros , Resultado do Tratamento , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Breast Cancer Res Treat ; 166(1): 85-94, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28717852

RESUMO

PURPOSE: Elevated S100A8 expression has been observed in cancers of the bladder, esophagus, colon, ovary, and breast. S100A8 is expressed by breast cancer cells as well as by infiltrating immune and myeloid cells. Here we investigate the association of elevated S100A8 protein expression in breast cancer cells and in breast tumor stroma with survival outcomes in a cohort of breast cancer patients. PATIENTS AND METHODS: Tissue microarrays (TMA) were constructed from breast cancer specimens from 417 patients with stage I-III breast cancer treated at the University of Michigan Comprehensive Cancer Center between 2004 and 2006. Representative regions of non-necrotic tumor and distant normal tissue from each patient were used to construct the TMA. Automated quantitative immunofluorescence (AQUA) was used to measure S100A8 protein expression, and samples were scored for breast cancer cell and stromal S100A8 expression. S100A8 staining intensity was assessed as a continuous value and by exploratory dichotomous cutoffs. Associations between breast cancer cell and stromal S100A8 expression with disease-free survival and overall survival were determined using the Kaplan-Meier method and Cox proportional hazard models. RESULTS: High breast cancer cell S100A8 protein expression (as indicated by AQUA scores), as a continuous measure, was a significant prognostic factor for OS [univariable hazard ratio (HR) 1.24, 95% confidence interval (CI) 1.00-1.55, p = 0.05] in this patient cohort. Exploratory analyses identified optimal S100A8 AQUA score cutoffs within the breast cancer cell and stromal compartments that significantly separated survival curves for the complete cohort. Elevated breast cancer cell and stromal S100A8 expression, indicated by higher S100A8 AQUA scores, significantly associates with poorer breast cancer outcomes, regardless of estrogen receptor status. CONCLUSIONS: Elevated breast cancer cell and stromal S1008 protein expression are significant indicators of poorer outcomes in early stage breast cancer patients. Evaluation of S100A8 protein expression may provide additional prognostic information beyond traditional breast cancer prognostic biomarkers.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Calgranulina A/metabolismo , Células Estromais/metabolismo , Biomarcadores Tumorais , Neoplasias da Mama/mortalidade , Calgranulina A/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Feminino , Imunofluorescência , Humanos , Estimativa de Kaplan-Meier , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Modelos de Riscos Proporcionais , Receptores de Estrogênio/metabolismo , Células Estromais/patologia , Análise Serial de Tecidos , Microambiente Tumoral
3.
Breast Cancer Res Treat ; 143(1): 11-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24276281

RESUMO

CD44 is a transmembrane glycoprotein involved in numerous cellular functions, including cell adhesion and extracellular matrix interactions. It is known to be functionally diverse, with alternative splice variants increasingly implicated as a marker for tumor-initiating stem cells associated with poor prognosis. Here, we evaluate CD44 as a potential marker of long-term breast cancer outcomes. Tissue specimens from patients treated on the National Cancer Institute 79-C-0111 randomized trial of breast conservation versus mastectomy between 1979 and 1987 were collected, and immunohistochemistry was performed using the standard isoform of CD44. Specimens were correlated with patient characteristics and outcomes. Survival analysis was performed using the log rank test. Fifty-one patients had evaluable tumor sections and available long-term clinical follow up data at a median follow up of 25.7 years. Significant predictors of OS were tumor size (median OFS 25.4 years for ≤2 cm vs. 7.5 years for >2 cm, p = 0.001), nodal status (median OS 17.2 years for node-negative patients vs. 6.7 years for node positive patients, p = 0.017), and CD44 expression (median OS 18.9 years for CD44 positive patients vs. 8.6 years for CD44 negative patients, p = 0.049). There was a trend toward increased PFS for patients with CD44 positive tumors (median PFS 17.9 vs. 4.3 years, p = 0.17), but this did not reach statistical significance. These findings illustrate the potential utility of CD44 as a prognostic marker for early stage breast cancer. Subgroup analysis in patients with lymph node involvement revealed CD44 positivity to be most strongly associated with increased survival, suggesting a potential role of CD44 in decision making for axillary management. As there is increasing interest in CD44 as a therapeutic target in ongoing clinical trials, the results of this study suggest additional investigation regarding the role CD44 in breast cancer is warranted.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Receptores de Hialuronatos/metabolismo , Adulto , Idoso , Neoplasias da Mama/patologia , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Gradação de Tumores , Metástase Neoplásica , Prognóstico , Fatores de Risco , Carga Tumoral
4.
Breast Cancer Res Treat ; 122(2): 371-80, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19842031

RESUMO

Studies of gene regulated by estrogen in breast cancer 1 (GREB1) have focused on mRNA levels with limited evidence about GREB1 protein expression in normal and breast cancer cells. A monoclonal antibody that recognizes GREB1 protein in breast tissues could be applied to correlate protein expression with established mRNA expression data. A hybridoma expressing a murine monoclonal antibody targeting a 119 amino acid peptide specific to human GREB1 was generated. The novel monoclonal GREB1 antibody (GREB1ab) was validated for use in Western blotting as well as immunohistochemical (IHC) applications. GREB1ab detects a 216 kDa protein corresponding to GREB1 in estrogen receptor alpha (ERalpha+) breast cancer cells as well as ERalpha- breast cancer cells transduced with a GREB1 expression vector. GREB1ab specificity was verified using an ERalpha antagonist to prevent GREB1 induction as well as a silencing siRNA targeting GREB1 mRNA. GREB1ab was further validated for detection of GREB1 by IHC in breast cancer cell lines and breast tissue microarrays (TMA). ERalpha+ cell lines were observed to express GREB1 while ERalpha- cell lines did not express detectable levels of the protein. Using breast cancer tissue whole sections, IHC with the GREB1ab identified protein expression in ERalpha+ breast cancer tissue as well as normal breast tissue, with little GREB1 expression in ERalpha- breast cancer tissue. Furthermore, these data indicate that GREB1 mRNA expression correlates well with protein expression. The novel monoclonal GREB1ab is specific for GREB1 protein. This antibody will serve as a tool for investigations focused on the expression, distribution, and function of GREB1 in normal breast and breast cancer tissues.


Assuntos
Anticorpos Monoclonais/biossíntese , Especificidade de Anticorpos , Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , RNA Mensageiro/metabolismo , Animais , Anticorpos Monoclonais/genética , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Linhagem Celular Tumoral , Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hibridomas , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Interferência de RNA , Receptor ErbB-2/metabolismo , Reprodutibilidade dos Testes , Análise Serial de Tecidos
5.
Science ; 232(4757): 1540-3, 1986 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-3715461

RESUMO

The hormone 17 beta-estradiol acts through its receptor system to induce MCF-7 human breast cancer cells to form tumors in athymic mice. In vitro studies have identified the production of estrogen-induced growth factors from MCF-7 cells that may have a role in growth control. These induced growth factors were sufficient to stimulate MCF-7 tumor growth in ovariectomized athymic mice, thus partially replacing estradiol. Growth factors may act as estrogen-induced "second messengers" in estrogen-responsive growth of human breast cancer.


Assuntos
Neoplasias da Mama/patologia , Estradiol/fisiologia , Receptores de Estradiol/fisiologia , Receptores de Estrogênio/fisiologia , Animais , Divisão Celular , Linhagem Celular , Meios de Cultura , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ovariectomia , Transplante Heterólogo
6.
Science ; 228(4700): 725-8, 1985 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-4039465

RESUMO

The natural history of estrogen-responsive breast cancers often involves a phenotypic change to an estrogen-unresponsive, more aggressive tumor. The human breast cancer cell line, MCF-7, which requires estradiol for tumor formation in vivo and shows growth stimulation in response to estradiol in vitro, is a model for hormone-responsive tumors. The v-rasH onc gene was transfected into MCF-7 cells. The cloned MCF-7ras transfectants, which expressed the v-rasH messenger RNA and v-rasH p21 protein (21,000 daltons), were characterized. In contrast to the parental cell line, MCF-7ras cells no longer responded to exogenous estrogen in culture and their growth was minimally inhibited by exogenous antiestrogens. When tested in the nude mouse, the MCF-7ras cells were fully tumorigenic in the absence of estrogen supplementation. Thus, cells acquiring an activated onc gene can bypass the hormonal regulatory signals that trigger the neoplastic growth of a human breast cancer cell line.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/induzido quimicamente , Estrogênios/farmacologia , Oncogenes , Transfecção , Animais , Neoplasias da Mama/induzido quimicamente , Linhagem Celular , DNA de Neoplasias/genética , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Pirrolidinas/farmacologia , Sequências Repetitivas de Ácido Nucleico , Tiofenos/farmacologia
7.
Science ; 225(4667): 1162-5, 1984 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-6474170

RESUMO

Nuclear estrogen receptor from MCF-7 cells undergoes a time-dependent, hormone-inducible transformation to a form that is less extractable from nuclei and less exchangeable with ligand. This receptor-modifying, intranuclear event is independent of receptor loss (processing) and appears associated with hormone responsiveness (progesterone-receptor induction) in these cells. The magnitude of receptor loss, however, is variable and apparently not a prerequisite for hormone action to induce progesterone receptor.


Assuntos
Neoplasias da Mama/metabolismo , Receptores de Estrogênio/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Feminino , Humanos , Receptores de Estradiol , Receptores de Progesterona/biossíntese , Fatores de Tempo
8.
Science ; 249(4976): 1552-5, 1990 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-2218496

RESUMO

The erbB2 oncogene encodes a 185-kilodalton transmembrane protein whose sequence is similar to the epidermal growth factor receptor (EGFR). A 30-kilodalton factor (gp30) secreted from MDA-MB-231 human breast cancer cells was shown to be a ligand for p185erbB2. An antibody to EGFR abolished the tyrosine phosphorylation induced by EGF and transforming growth factor-alpha (TGF-alpha) but only partially blocked that produced by gp30 in SK-BR-3 breast cancer cells. In two cell lines that overexpress erbB2 but do not expresss EGFR (MDA-MB-453 breast cancer cells and a Chinese hamster ovary cell line that had been transfected with erbB2), phosphorylation of p185erbB2 was induced only by gp30. The gp30 specifically inhibited the growth of cells that overexpressed p185erbB2. An antibody to EGFR had no effect on the inhibition of SK-BR-3 cell colony formation obtained with gp30. Thus, it appeared that gp30 interacted directly with the EGFR and erbB2. Direct binding of gp30 to p185erbB2 was confirmed by binding competition experiments, where gp30 was found to displace the p185erbB2 binding of a specific antibody to p185erbB2. The evidence described here suggests that gp30 is a ligand for p185erbB2.


Assuntos
Receptores ErbB/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Anticorpos Monoclonais , Ligação Competitiva , Neoplasias da Mama , Linhagem Celular , Cromatografia de Afinidade , Receptores ErbB/isolamento & purificação , Feminino , Humanos , Cinética , Ligantes , Peso Molecular , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/imunologia , Proto-Oncogenes , Transfecção
9.
J Clin Invest ; 65(3): 586-91, 1980 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7188772

RESUMO

Vitamin A and its analogues (retinoids) regulate the differentiation of epithelial tissues. Retinoids inhibit the induction of rat mammary cancers by carcinogens in vivo, and cellular binding proteins for retinoids have been demonstrated in some human breast cancer samples. In this study, we examined the model system of human breast cancer cell lines in long-term tissue culture for effects of retinoids on growth and for the presence of cellular retinoid binding proteins. Retinoic acid and retinol inhibit the growth of of MCF-7, Hs578T, and ZR-75-B cell lines. Retinoic acid is more potent than retinol in this regard: 50% growth inhibition is achieved by 6 nM retinoic acid in ZR-75-B and by 700 nM in MCF-7 and Hs578T, whereas 5-8 muM retinol is required in all three cell lines. The time to onset of growth inhibition varies markedly between cell lines and is not related to cell density or doubling time. Retinoic acid increases the doubling time of MCF-7 and ZR-75-B by two- to threefold, but causes cell death in Hs578T. The growth inhibition is reversible in every cell line by removal of retinoic acid. Specific and distinct binding of [(3)H]retinoic acid and [(3)H]retinol is present in cytosols of MCF-7 and Hs578T cells as assessed by sucrose density gradient centrifugation. In ZR-75-B, [(3)H]retinoic acid binding was present, but no binding of [(3)H]retinol was detectable. This study reveals that retinoids may play an important role in the regulation and treatment of human breast cancer and that human breast cancer cell lines represent a useful model to study this role.


Assuntos
Neoplasias da Mama/metabolismo , Tretinoína/metabolismo , Vitamina A/metabolismo , Neoplasias da Mama/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Citosol/metabolismo , Feminino , Humanos , Proteínas de Ligação ao Retinol/metabolismo , Tretinoína/farmacologia , Vitamina A/farmacologia
10.
J Clin Invest ; 52(7): 1715-25, 1973 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-4352461

RESUMO

The first known step in steroid hormone action is the association of the steroid with specific cytoplasmic steroid-binding proteins (SBP). Using a competitive binding assay, we detected, quantified, and partially characterized such a SBP in cytosol from glucocorticoid-sensitive human lymphoblastic leukemic blasts. The affinity of steroids for the SBP was directly related to their known killing potency. For example, steroids without glucocorticoid effect such as androstenedione, etiocholanolone, and tetrahydrocortisol were unable to displace radiolabeled dexamethasone from the SBP in the binding reaction. The dose-response curve for in vitro inhibition of [(3)H]thymidine uptake in leukemic blasts correlated closely with the binding affinity of glucocorticoids to the SBP, providing additional support for an essential physiologic role for SBP in steroid action. SBP activity was either greatly diminished or absent in glucocorticoid-resistant cells. Six patients who intially had SBP in their blasts and were responsive to combinations of drugs including glucocorticoids no longer had SBP activity detectable at a time when they no longer responded to combinations of drugs including glucocorticoids. In vitro [(3)H]thymidine uptake was not inhibited by steroids in leukemic blast cells lacking SBP activity. Other patients who had received some antileukemic therapy including glucocorticoids and who still had SBP in their leukemic blasts, were still responsive to drug combinations that included glucocorticoids. This appears to be the first study demonstrating glucocorticoid receptors in a human tissue.


Assuntos
Leucemia Linfoide/metabolismo , Linfócitos/metabolismo , Ligação Proteica , Androstenodiona/metabolismo , Ligação Competitiva , Citosol , Dexametasona/metabolismo , Relação Dose-Resposta a Droga , Etiocolanolona/metabolismo , Humanos , Hidrocortisona/metabolismo , Técnicas In Vitro , Cinética , Linfócitos/efeitos dos fármacos , Receptores de Superfície Celular , Tetra-Hidrocortisol/metabolismo , Timidina/metabolismo , Trítio
11.
J Clin Invest ; 102(6): 1142-51, 1998 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9739048

RESUMO

LIGHT is a new member of tumor necrosis factor (TNF) cytokine family derived from an activated T cell cDNA library. LIGHT mRNA is highly expressed in splenocytes, activated PBL, CD8(+) tumor infiltrating lymphocytes, granulocytes, and monocytes but not in the thymus and the tumor cells examined. Introduction of LIGHT cDNA into MDA-MB-231 human breast carcinoma caused complete tumor suppression in vivo. Histological examination showed marked neutrophil infiltration and necrosis in LIGHT expressing but not in the parental or the Neo-transfected MDA-MB-231 tumors. Interferon gamma (IFNgamma) dramatically enhances LIGHT-mediated apoptosis. LIGHT protein triggers apoptosis of various tumor cells expressing both lymphotoxin beta receptor (LTbetaR) and TR2/HVEM receptors, and its cytotoxicity can be blocked specifically by addition of a LTbetaR-Fc or a TR2/HVEM-Fc fusion protein. However, LIGHT was not cytolytic to the tumor cells that express only the LTbetaR or the TR2/HVEM or hematopoietic cells examined that express only the TR2/HVEM, such as PBL, Jurkat cells, or CD8(+) TIL cells. In contrast, treatment of the activated PBL with LIGHT resulted in release of IFNgamma. Our data suggest that LIGHT triggers distinct biological responses based on the expression patterns of its receptors on the target cells. Thus, LIGHT may play a role in the immune modulation and have a potential value in cancer therapy.


Assuntos
Apoptose , Genes Supressores de Tumor , Proteínas de Membrana/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Virais/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Mapeamento Cromossômico , Meios de Cultura Livres de Soro , Feminino , Técnicas de Transferência de Genes , Humanos , Hibridização in Situ Fluorescente , Interferon gama/metabolismo , Ligantes , Ativação Linfocitária , Linfócitos do Interstício Tumoral , Receptor beta de Linfotoxina , Masculino , Proteínas de Membrana/genética , Membro 14 de Receptores do Fator de Necrose Tumoral , Células Tumorais Cultivadas , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/genética
12.
Oncogene ; 36(11): 1559-1572, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27669433

RESUMO

The receptor for advanced glycation end products (RAGE) is highly expressed in various cancers and is correlated with poorer outcome in breast and other cancers. Here we tested the role of targeting RAGE by multiple approaches in the tumor and tumor microenvironment, to inhibit the metastatic process. We first tested how RAGE impacts tumor cell-intrinsic mechanisms using either RAGE overexpression or knockdown with short hairpin RNAs (shRNAs). RAGE ectopic overexpression in breast cancer cells increased MEK-EMT (MEK-epithelial-to-mesenchymal transition) signaling, transwell invasion and soft agar colony formation, and in vivo promoted lung metastasis independent of tumor growth. RAGE knockdown with multiple independent shRNAs in breast cancer cells led to decreased transwell invasion and soft agar colony formation, without affecting proliferation. In vivo, targeting RAGE shRNA knockdown in human and mouse breast cancer cells, decreased orthotopic tumor growth, reduced tumor angiogenesis and recruitment of inflammatory cells, and markedly decreased metastasis to the lung and liver in multiple xenograft and syngeneic mouse models. To test the non-tumor cell microenvironment role of RAGE, we performed syngeneic studies with orthotopically injected breast cancer cells in wild-type and RAGE-knockout C57BL6 mice. RAGE-knockout mice displayed striking impairment of tumor cell growth compared with wild-type mice, along with decreased mitogen-activated protein kinase signaling, tumor angiogenesis and inflammatory cell recruitment. To test the combined inhibition of RAGE in both tumor cell-intrinsic and non-tumor cells of the microenvironment, we performed in vivo treatment of xenografted tumors with FPS-ZM1 (1 mg/kg, two times per week). Compared with vehicle, FPS-ZM1 inhibited primary tumor growth, inhibited tumor angiogenesis and inflammatory cell recruitment and, most importantly, prevented metastasis to the lung and liver. These data demonstrate that RAGE drives tumor progression and metastasis through distinct tumor cell-intrinsic and -extrinsic mechanisms, and may represent a novel and therapeutically viable approach for treating metastatic cancers.


Assuntos
Neoplasias da Mama/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Modelos Animais de Doenças , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Ligantes , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Interferente Pequeno , Receptor para Produtos Finais de Glicação Avançada/genética , Carga Tumoral
13.
J Natl Cancer Inst ; 84(22): 1716-24, 1992 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-1279186

RESUMO

BACKGROUND: In a proliferating tumor, locally secreted polypeptide growth factors, which have autocrine and paracrine functions, induce vascularization essential for tumor growth and metastasis. These growth factors may serve as targets for tumor therapy. We have shown that the heparinoid pentosan polysulfate (PPS) can block growth of subcutaneous human tumor xenografts in nude mice and angiogenesis induced by the heparin-binding, Kaposi's sarcoma-derived fibroblast growth factor (K-FGF). PURPOSE: The purpose of this study was to determine whether PPS might also interfere with stimulation of endothelial cells by other growth factors released from tumor cells and whether the promising antitumor effects of PPS extend to other human tumor cell lines. We studied the effects of PPS on stimulation by heparin-binding growth factors released from seven human tumor cell lines in vitro and on tumors growing from these cell lines in athymic nude mice. METHODS: Seven human cell lines established from breast, prostate, epidermoid, and lung carcinomas and rhabdomyosarcoma were used in in vivo as well as in vitro studies of the effects of PPS. We also studied in vitro the effects of PPS on growth factor-induced colony formation of normal rat kidney fibroblasts and human adrenal carcinoma cells. RESULTS: The tumor cell lines released growth factors into their media that stimulated growth of endothelial and epithelial cells as well as fibroblasts. Heparin-affinity chromatography showed that heparin-binding growth factors contributed substantially to this paracrine activity and that PPS inhibited this stimulus. Six of the seven tumor cell lines were resistant to PPS in soft-agar cloning assays and hence did not appear to depend on autocrine stimulation by the heparin-binding growth factors. In contrast to this in vitro resistance, subcutaneous growth of tumors from all cell lines in athymic nude mice was inhibited in a dose-dependent fashion by daily intraperitoneal injections of PPS. CONCLUSIONS: We conclude that heparin-binding growth factors contribute substantially to tumor growth in vivo and that PPS acts by blocking the paracrine effects of heparin-binding growth factors released from the tumor cells. IMPLICATION: PPS could become a novel treatment tool targeting tumor growth factors.


Assuntos
Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Poliéster Sulfúrico de Pentosana/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Animais , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Fator 4 de Crescimento de Fibroblastos , Heparina/metabolismo , Humanos , Fatores Imunológicos , Técnicas In Vitro , Camundongos , Camundongos Nus , Neoplasias Experimentais/patologia , Receptores de Superfície Celular/efeitos dos fármacos , Células Tumorais Cultivadas
14.
J Natl Cancer Inst ; 58(6): 1591-3, 1977 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-559098

RESUMO

Four human breast cancer cell lines were tested for their response to hormones with respect to the lactational function, fatty acid synthesis. Physiologic concentrations of insulin enhanced the incorporation of [14C]acetate into fatty acids in two of four cell lines tested. All cell lines had specific, high-affinity insulin receptors; therefore, the failure of two lines to respond could not be attributed to the absence of receptor. The effect of insulin involved an increase in the maximun velocity of incorporation rather than a decrease in the Michaelis constant.


Assuntos
Neoplasias da Mama/metabolismo , Ácidos Graxos/biossíntese , Insulina/farmacologia , Acetatos/metabolismo , Linhagem Celular , Feminino , Humanos , Cinética , Lactação , Gravidez , Receptor de Insulina
15.
J Natl Cancer Inst ; 85(21): 1758-64, 1993 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-8411260

RESUMO

BACKGROUND: Expression of matrix metalloproteinase-2 (MMP-2), the 72-kd type IV collagenase/gelatinase, by cancer cells has been implicated in metastasis through cancer cell invasion of basement membranes mediated by degradation of collagen IV. However, the abundance of this latent proenzyme in normal tissues and fluids suggests that MMP-2 proenzyme utilization is limited by its physiological activation rather than expression alone. We previously reported activation of this proenzyme by normal and malignant fibroblastoid cells cultured on collagen I (vitrogen) gels. PURPOSE: Our purposes in this study were 1) to determine whether MMP-2 activation is restricted to the more invasive human breast cancer cell lines and 2) to localize the activating mechanism. METHODS: Zymography was used to monitor MMP-2 activation through detection of latent MMP-2 (72 kd) and mature species of smaller molecular weight (59 or 62 kd). Human breast cancer cell lines cultured on plastic, vitrogen, and other matrices were thus screened for MMP-2 activation. Collagen I-cultured cells were exposed to cycloheximide, a protein synthesis inhibitor, or to protease inhibitors to determine the nature of the MMP-2-activating mechanism. Triton X-114 (TX-114) detergent extracts from cells cultured on collagen I or plastic were incubated with latent MMP-2 and analyzed by zymography to localize the MMP-2 activator. RESULTS: MMP-2 activation was only induced by collagen I culture in the more aggressive, highly invasive estrogen receptor-negative, vimentin-positive human breast cancer cell lines (Hs578T, MDA-MB-436, BT549, MDA-MB-231, MDA-MB-435, MCF-7 ADR) and was independent of MMP-2 production. MMP-2 activation was detected in cells cultured on collagen I gels but not in those cultured on gelatin gels, Matrigel, or thin layers of collagen I or IV, gelatin, or fibronectin. Collagen-induced activation was specific for the enzyme species MMP-2, since MMP-9, the 92-kd type IV collagenase/gelatinase, was not activatable under similar conditions. MMP-2 activation was inhibited by cycloheximide and was sensitive to a metalloproteinase inhibitor but not to aspartyl, serine, or cysteinyl protease inhibitors. MMP-2 activation was detected in the hydrophobic, plasma membrane-enriched, TX-114 extracts from invasive collagen I-cultured cells. CONCLUSION: Collagen I-induced MMP-2 activation is restricted to highly invasive estrogen receptor-negative, vimentin-positive human breast cancer cell lines, is independent of MMP-2 production, and is associated with metastatic potential. Our findings are consistent with plasma membrane localization of the activator. IMPLICATIONS: The MMP-2 activation mechanism may represent a new target for diagnosis, prognosis, and treatment of human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Gelatinases/metabolismo , Metaloendopeptidases/metabolismo , Neoplasias da Mama/patologia , Colágeno/metabolismo , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática/efeitos dos fármacos , Feminino , Imunofluorescência , Gelatinases/biossíntese , Gelatinases/isolamento & purificação , Humanos , Metaloproteinase 2 da Matriz , Metaloendopeptidases/biossíntese , Metaloendopeptidases/isolamento & purificação , Metástase Neoplásica , Inibidores de Proteases/farmacologia , Sensibilidade e Especificidade , Células Tumorais Cultivadas
16.
J Natl Cancer Inst ; 83(10): 716-20, 1991 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-1708834

RESUMO

A neoangiogenic response is critical for the unrestricted growth of solid tumors beyond a few millimeters in diameter. Release of adequate growth-stimulating activity from tumor cells is obviously required for the stimulation of blood vessel growth, and blockade of such stimulatory activity should repress tumor growth at the microscopic level. To test this hypothesis and to study appropriate inhibitors, we used a human adrenal cancer cell line (SW-13/K-fgf) engineered to secrete Kaposi's sarcoma-derived fibroblast growth factor (K-FGF), which we previously showed to induce growth of highly vascularized subcutaneous tumors in animals by autocrine and paracrine stimuli. In the present study, we tested different polysulfates for their selective inhibition of proliferation induced by K-FGF versus proliferation independent of K-FGF. Suramin and dextran sulfate showed slight selective inhibition of K-FGF-induced proliferation, ie, inhibition three- and five-fold greater, respectively, than the inhibition of proliferation independent of K-FGF. In contrast, heparin was inactive. The heparin analogue pentosan polysulfate (PPS), however, showed selective inhibition that was more than 2000-fold greater. The inhibitory effects of PPS on growth of SW-13/K-fgf cells, as well as endothelial cells, were fully reversible by an excess of added FGF. Daily intraperitoneal injections of PPS were tolerated well by athymic nude mice and prevented growth of subcutaneous SW-13/K-fgf tumor xenografts. PPS will be a useful tool to elucidate the effects of FGFs in vitro and in vivo and appears to be a prototype for the development of tumoricidal therapy based on targeting of growth factors.


Assuntos
Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos , Poliéster Sulfúrico de Pentosana/farmacologia , Proteínas Proto-Oncogênicas/farmacologia , Neoplasias das Glândulas Suprarrenais/irrigação sanguínea , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Endotélio Vascular/citologia , Feminino , Fator 4 de Crescimento de Fibroblastos , Heparina/farmacologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neovascularização Patológica , Poliéster Sulfúrico de Pentosana/toxicidade , Proteínas Proto-Oncogênicas/genética , Suramina/farmacologia , Transfecção , Células Tumorais Cultivadas
17.
J Natl Cancer Inst ; 85(13): 1068-73, 1993 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-7685831

RESUMO

BACKGROUND: Previous studies indicate that the heparinoid pentosan polysulfate (PPS) can inhibit heparin-binding growth factors (HBGFs) released from tumor cells and thus block tumor growth in animal models. However, because of its heparin-like activity, the major toxic effect expected for PPS is its inhibition of coagulation. PURPOSE: Our purpose was to determine if anti-HBGF activity could be achieved in patients without causing complications from anticoagulation. METHODS: We initiated a phase I trial in cancer patients and developed a cell proliferation assay to detect PPS in human serum based on its antigrowth factor activity. Blood samples from six healthy volunteers were collected in tubes containing different concentrations of PPS (FIBREZYM; concentration range, 0-10 micrograms/mL). Additional samples were obtained from four patients in the phase I trial before and after subcutaneous treatment with 15 mg/m2 of PPS. The activated partial thromboplastin time (aPTT), which is associated with coagulation, was measured in all blood samples. Serum prepared from the blood samples was heat inactivated and then incubated for 4-5 days with proliferating SW-13 cells, allowing determination of antigrowth factor activity. RESULTS: PPS added to blood samples increased aPTT only at concentrations above 1 microgram/mL, whereas HBGF-dependent proliferation was inhibited at less than 0.1 microgram/mL. Sera obtained from patients up to 4 hours after PPS treatment specifically inhibited HBGF-dependent cell proliferation by more than 65% even at a 1:10 dilution. At the same time, the aPTT was not altered in these patients, indicating no significant effect on coagulation by this dose of the heparinoid. CONCLUSIONS: HBGF-inhibitory concentrations of PPS can be achieved in patients' sera without significant effects on coagulation. IMPLICATION: The assay presented here could be useful to determine doses and scheduling of treatment in studies evaluating PPS as an antitumor agent.


Assuntos
Substâncias de Crescimento/sangue , Poliéster Sulfúrico de Pentosana/farmacologia , Bioensaio , Coagulação Sanguínea/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Humanos , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Tempo de Tromboplastina Parcial , Poliéster Sulfúrico de Pentosana/uso terapêutico , Valores de Referência , Células Tumorais Cultivadas
18.
J Natl Cancer Inst ; 88(24): 1821-7, 1996 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-8961971

RESUMO

BACKGROUND: Women who took the synthetic estrogen diethylstilbestrol during pregnancy exhibit an elevated risk of breast cancer, whereas those who suffered from preeclampsia, which is associated with low circulating pregnancy estrogens, exhibit a reduced risk. Since a high-fat diet may increase circulating estrogen levels and possibly breast cancer risk, dietary factors during pregnancy could influence the risk of developing this disease. PURPOSE: We tested the hypothesis that consumption of a high-fat diet during pregnancy increases carcinogen-induced mammary tumor incidence in rats. METHODS: Pregnant or virgin female Sprague-Dawley rats that had been previously treated with 10 mg 7, 12-dimethylbenz[a]anthracene (DMBA) by oral gavage when 55 days old were assigned to one of two isocaloric diets containing either 16% calories from fat (low-fat) or 43% calories from fat (high-fat) for the length of pregnancy or for the equivalent time of approximately 21 days. There were 20 pregnant and 10 nonpregnant DMBA-treated rats per group. Ten additional pregnant animals (not previously treated with DMBA) per group were used for hormone analysis. The fat source used was corn oil, which is high in n-6 polyunsaturated fatty acids, primarily linoleic acid. The animals were checked for tumors at least once per week by palpation. The tumor size, number, and latency to appearance after carcinogen exposure were recorded. The statistical significance of observed differences was tested by use of appropriate two-sided tests. RESULTS: Female rats on different diets had virtually identical food intakes and weight gains during pregnancy. On gestation day 19, serum estradiol levels were approximately twofold higher in rats fed a high-fat diet than in rats fed a low-fat diet (P < .02). The serum insulin levels and insulin/glucose ratios (an index of insulin resistance) in rats fed the high-fat diet were approximately twofold lower than in rats fed the low-fat diet, but the differences did not reach statistical significance (P < .09 and P < .09, respectively). On week 18 following DMBA administration, the number of rats developing mammary tumors was significantly higher in the group exposed to a high-fat diet (40% of animals) than in the group exposed to a low-fat diet (10% of animals) during pregnancy (P < .05). Tumor multiplicity, latency to tumor appearance, and size of tumors upon first detection were similar among the dietary groups. No intergroup differences in the mammary tumor incidence were noted in virgin animals that were exposed to the high- or low-fat diets for an equivalent period of time. CONCLUSIONS: Our findings indicate that consumption of a diet high in fat (primarily in the form of n-6 polyunsaturated fatty acids) during pregnancy increases the risk of developing carcinogen-induced mammary tumors, possibly by increasing the pregnancy levels of circulating estrogens. IMPLICATIONS: If further studies find that the results from animal model studies are applicable to humans, some human breast cancers may be preventable by dietary manipulations during pregnancy.


Assuntos
Gorduras na Dieta/efeitos adversos , Ácidos Graxos Insaturados/efeitos adversos , Neoplasias Mamárias Experimentais/induzido quimicamente , Complicações Neoplásicas na Gravidez/induzido quimicamente , 9,10-Dimetil-1,2-benzantraceno , Animais , Glicemia/metabolismo , Gorduras na Dieta/administração & dosagem , Ingestão de Energia , Estrogênios/sangue , Ácidos Graxos Insaturados/administração & dosagem , Feminino , Humanos , Insulina/sangue , Neoplasias Mamárias Experimentais/sangue , Gravidez , Complicações Neoplásicas na Gravidez/sangue , Ratos , Ratos Sprague-Dawley , Risco
19.
J Natl Cancer Inst ; 74(2): 341-7, 1985 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3856048

RESUMO

Epithelial cells were isolated and cultured from a number of human mammary specimens of both cancerous and noncancerous origin. Doxorubicin (Dx) sensitivity was measured at second passage with the use of a highly efficient clonogenic assay. For 23 different tumor specimens derived from patients without previous chemotherapy, the drug concentrations required to kill 50% of the cells varied approximately 35-fold. In contrast, for 11 tumor specimens from patients who relapsed after regimens containing Dx, the drug concentration for 50% survival varied only fivefold and the dose-response curves for these specimens clustered at the more resistant end of the spectrum. A wide range of sensitivities was also observed among 13 noncancerous mammary specimens; however, tumor tissue and noncancerous tissue from the same donor were similar. When cultures were subjected to drug incubation periods of 1 and 4 hours, dose-response curves were superimposable when plotted as a function of drug concentration multiplied by time.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Mama/efeitos dos fármacos , Carcinoma/patologia , Ensaio de Unidades Formadoras de Colônias , Doxorrubicina/farmacologia , Ensaio Tumoral de Célula-Tronco , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Epitélio/efeitos dos fármacos , Feminino , Congelamento , Humanos , Preservação de Tecido
20.
J Natl Cancer Inst ; 68(2): 267-70, 1982 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-6801367

RESUMO

WRK-1, a cell line in long-term culture derived from a 7, 12-dimethylbenz[a]anthracene-induced rat mammary tumor, responds to physiologic concentrations of vasopressin with increased precursor incorporation into phospholipids and with increased protein accumulation. Because vasopressin has been reported to be a potent mitogen for Hela cells and 3T3 cells, a study was conducted to determine whether it could act as a mitogen for WRK-1 cells. Under no conditions was a clear-cut mitogen response to vasopressin demonstrated.


Assuntos
9,10-Dimetil-1,2-benzantraceno , Benzo(a)Antracenos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Vasopressinas/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Mitógenos/farmacologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA