Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
CA Cancer J Clin ; 60(6): 351-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20959401

RESUMEN

Inflammatory breast cancer (IBC) is a rare and aggressive form of invasive breast cancer accounting for 2.5% of all breast cancer cases. It is characterized by rapid progression, local and distant metastases, younger age of onset, and lower overall survival compared with other breast cancers. Historically, IBC is a lethal disease with less than a 5% survival rate beyond 5 years when treated with surgery or radiation therapy. Because of its rarity, IBC is often misdiagnosed as mastitis or generalized dermatitis. This review examines IBC's unique clinical presentation, pathology, epidemiology, imaging, and biology and details current multidisciplinary management of the disease, which comprises systemic therapy, surgery, and radiation therapy.


Asunto(s)
Neoplasias Inflamatorias de la Mama/diagnóstico , Neoplasias Inflamatorias de la Mama/terapia , Biomarcadores de Tumor/genética , Índice de Masa Corporal , Quimioterapia Adyuvante , Terapia Combinada/métodos , Diagnóstico Diferencial , Progresión de la Enfermedad , Femenino , Humanos , Incidencia , Neoplasias Inflamatorias de la Mama/epidemiología , Neoplasias Inflamatorias de la Mama/genética , Imagen por Resonancia Magnética , Mamografía , Estadificación de Neoplasias , Obesidad/complicaciones , Tomografía de Emisión de Positrones , Pronóstico , Radioterapia Adyuvante , Enfermedades Raras , Medición de Riesgo , Factores de Riesgo , Tasa de Supervivencia , Tomografía Computarizada por Rayos X , Ultrasonografía Mamaria , Estados Unidos/epidemiología
2.
Breast Cancer Res Treat ; 140(1): 23-33, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23784380

RESUMEN

Inflammatory breast cancer (IBC) is the most aggressive type of advanced breast cancer characterized by rapid proliferation, early metastatic development and poor prognosis. Since there are few preclinical models of IBC, there is a general lack of understanding of the complexity of the disease. Recently, we have developed a new model of IBC derived from the pleural effusion of a woman with metastatic secondary IBC. FC-IBC02 cells are triple negative and form clusters (mammospheres) in suspension that are strongly positive for E-cadherin, ß-catenin and TSPAN24, all adhesion molecules that play an important role in cell migration and invasion. FC-IBC02 cells expressed stem cell markers and some, but not all of the characteristics of cells undergoing epithelial mesenchymal transition (EMT). Breast tumor FC-IBC02 xenografts developed quickly in SCID mice with the presence of tumor emboli and the development of lymph node and lung metastases. Remarkably, FC-IBC02 cells were able to produce brain metastasis in mice on intracardiac or intraperitoneal injections. Genomic studies of FC-IBC02 and other IBC cell lines showed that IBC cells had important amplification of 8q24 where MYC, ATAD2 and the focal adhesion kinase FAK1 are located. MYC and ATAD2 showed between 2.5 and 7 copies in IBC cells. FAK1, which plays important roles in anoikis resistance and tumor metastasis, showed 6-4 copies in IBC cells. Also, CD44 was amplified in triple-negative IBC cells (10-3 copies). Additionally, FC-IBC02 showed amplification of ALK and NOTCH3. These results indicate that MYC, ATAD2, CD44, NOTCH3, ALK and/or FAK1 may be used as potential targeted therapies against IBC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/patología , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Quinasa de Linfoma Anaplásico , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Antígenos CD4/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Cromosomas Humanos Par 8 , Proteínas de Unión al ADN/genética , Transición Epitelial-Mesenquimal , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Genes myc , Humanos , Neoplasias Inflamatorias de la Mama/metabolismo , Pérdida de Heterocigocidad , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Terapia Molecular Dirigida , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Notch3 , Receptores Notch/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Exp Ther Oncol ; 10(3): 219-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24416998

RESUMEN

Inflammatory breast cancer (IBC) is the most metastatic variant of locally advanced breast cancer. IBC has distinctive characteristics including invasion of tumor emboli into the skin and rapid disease progression. Given our previous studies suggesting that HDAC inhibitors have promise in targeting IBC, the present study revealed that the class I HDAC inhibitor Romidepsin (FK-288, Istodax; Celgene Corporation, Summit, NJ) potently induced destruction of IBC tumor emboli and lymphatic vascular architecture. associated with inhibition of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1alpha, (HIF1alpha) proteins in the Mary-X pre-clinical model of IBC. Romidepsin treatment induced clinically relevant biomarkers in including induction of acetylated Histone 3 (Ac-H3) proteins, apoptosis, and increased p21WAF1/CIP1. Romidepsin, alone and synergistically when combined with Paclitaxel, effectively eliminated both primary tumors and metastatic lesions at multiple sites formed by the SUM149 IBC cell line. This is the first report of the ability of an HDAC inhibitor to eradicate IBC tumor emboli, to destroy the integrity of lymphatic vessel architecture and to target metastasis. Furthermore, Romidepsin, in combination with a taxane, warrants evaluation as a therapeutic strategy that may effectively target the skin involvement and rapid metastasis that are hallmarks of IBC.


Asunto(s)
Depsipéptidos/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Células Neoplásicas Circulantes/efectos de los fármacos , Paclitaxel/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Depsipéptidos/administración & dosificación , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/patología , Ratones , Metástasis de la Neoplasia/prevención & control , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Breast Cancer Res Treat ; 134(2): 495-510, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22547109

RESUMEN

Basal breast cancer, common among patients presenting with inflammatory breast cancer (IBC), has been shown to be resistant to radiation and enriched in cancer stem cells. The Notch pathway plays an important role in self-renewal of breast cancer stem cells and contributes to inflammatory signaling which promotes the breast cancer stem cell phenotype. Herein, we inhibited Notch signaling using a gamma secretase inhibitor, RO4929097, in an in vitro model that enriches for cancer initiating cells (3D clonogenic assay) and conventional 2D clonogenic assay to compare the effect on radiosensitization of the SUM149 and SUM190 IBC cell lines. RO4929097 downregulated the Notch target genes Hes1, Hey1, and HeyL, and showed a significant reduction in anchorage independent growth in SUM190 and SUM149. However, the putative self-renewal assay mammosphere formation efficiency was increased with the drug. To assess radiosensitization of putative cancer stem cells, cells were exposed to increasing doses of radiation with or without 1 µM RO4929097 in their standard (2D) and self-renewal enriching (3D) culture conditions. In the conventional 2D clonogenic assay, RO4929097 significantly sensitized SUM190 cells to ionizing radiation and has a modest radiosensitization effect in SUM149 cells. In the 3D clonogenic assays, however, a radioprotective effect was seen in both SUM149 and SUM190 cells at higher doses. Both cell lines express IL-6 and IL-8 cytokines known to mediate the efficacy of Notch inhibition and to promote self-renewal of stem cells. We further showed that RO429097 inhibits normal T-cell synthesis of some inflammatory cytokines, including TNF-α, a potential mediator of IL-6 and IL-8 production in the microenvironment. These data suggest that additional targeting agents may be required to selectively target IBC stem cells through Notch inhibition, and that evaluation of microenvironmental influences may shed further light on the potential effects of this inhibitor.


Asunto(s)
Antineoplásicos/farmacología , Benzazepinas/farmacología , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/farmacología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Aldehído Deshidrogenasa/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antígenos CD/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/efectos de la radiación , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Notch/antagonistas & inhibidores , Esferoides Celulares
5.
J Exp Ther Oncol ; 10(2): 155-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23350355

RESUMEN

Signal transducer and activator of transcription 3 (Stat3) is constitutively activated in a number of human cancers and cancer cell lines. Via its Src homology 2 (SH2) domain, Stat3 is recruited to phosphotyrosine residues on intracellular domains of cytokine and growth factor receptors, whereupon it is phosphorylated on Tyr705, dimerizes, translocates to the nucleus and is reported to participate in the expression of genes related to angiogenesis, metastasis, growth and survival. To block this process, we are developing cell-permeable, phosphatase-stable phosphopeptide mimics, targeted to the SH2 domain of Stat3, that inhibit the phosphorylation of Tyr705 of Stat3 in cultured tumor cells (Mandal et al., J. Med. Chem. 54, 3549-5463, 2011). At concentrations that inhibit tyrosine phosphorylation, these materials were not cytotoxic, similar to recent reports on JAK inhibitors. At higher concentrations, cytotoxicity was accompanied by off-target effects. We report that treatment of MDA-MB-468 human breast cancer xenografts in mice with peptidomimetic PM-73G significantly inhibited tumor growth, which was accompanied by reduction in VEGF production and microvessel density. No evidence of apoptosis or changes in the expression of the canonical genes cyclin D1 or survivin were observed. Thus selective inhibition of Stat3 Tyr705 phosphorylation may be a novel anti-angiogenesis strategy for the treatment of cancer.


Asunto(s)
Biomimética , Neoplasias de la Mama/prevención & control , Neovascularización Patológica/prevención & control , Fosfopéptidos/farmacología , Profármacos/farmacología , Factor de Transcripción STAT3/metabolismo , Dominios Homologos src/efectos de los fármacos , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Femenino , Humanos , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem Lett ; 21(20): 6071-3, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21903394

RESUMEN

An efficient synthesis of apricoxib (CS-706), a selective cyclooxygenase inhibitor, was developed using copper catalyzed homoallylic ketone formation from methyl 4-ethoxybenzoate followed by ozonolysis to an aldehyde, and condensation with sulfanilamide. This method provided multi-gram access of aprocoxib in good yield. Apricoxib exhibited potency equal to celecoxib at inhibition of prostaglandin E2 synthesis in two inflammatory breast cancer cell lines.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/antagonistas & inhibidores , Neoplasias Inflamatorias de la Mama/tratamiento farmacológico , Pirroles/síntesis química , Pirroles/farmacología , Sulfonamidas/síntesis química , Sulfonamidas/farmacología , Línea Celular Tumoral , Femenino , Humanos
7.
Future Oncol ; 7(1): 25-30, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21174535

RESUMEN

Inflammatory breast cancer (IBC) is the most aggressive and deadly form of breast cancer. In spite of the comprehensive multidisciplinary approach to the management of this disease, the prognosis remains dismal. Moreover, there have been no major advancements in understanding the etiology and biology of IBC and no significant improvements in the diagnosis of the disease. The International Inflammatory Breast Cancer Conference was established in 2008 with the intention of creating a forum for the discussion, collaboration and development of proposals and working hypotheses. Furthermore, the conference represented an opportunity to raise awareness regarding IBC. The second international conference reported on several new exciting projects based on work from investigators and research teams devoted to making a difference in the fight against this disease.


Asunto(s)
Neoplasias Inflamatorias de la Mama/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/mortalidad
8.
Biomed Microdevices ; 11(1): 49-63, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18663578

RESUMEN

Breast cancer is the field of medicine with the greatest presence of nanotechnological therapeutic agents in the clinic. A pegylated form of liposomally encapsulated doxorubicin is routinely used for treatment against metastatic cancer, and albumin nanoparticulate chaperones of paclitaxel were approved for locally recurrent and metastatic disease in 2005. These drugs have yielded substantial clinical benefit, and are steadily gathering greater beneficial impact. Clinical trials currently employing these drugs in combination with chemo and biological therapeutics exceed 150 worldwide. Despite these advancements, breast cancer morbidity and mortality is unacceptably high. Nanotechnology offers potential solutions to the historical challenge that has rendered breast cancer so difficult to contain and eradicate: the extreme biological diversity of the disease presentation in the patient population and in the evolutionary changes of any individual disease, the multiple pathways that drive disease progression, the onset of 'resistance' to established therapeutic cocktails, and the gravity of the side effects to treatment, which result from generally very poor distribution of the injected therapeutic agents in the body. A fundamental requirement for success in the development of new therapeutic strategies is that breast cancer specialists-in the clinic, the pharmaceutical and the basic biological laboratory-and nanotechnologists-engineers, physicists, chemists and mathematicians-optimize their ability to work in close collaboration. This further requires a mutual openness across cultural and language barriers, academic reward systems, and many other 'environmental' divides. This paper is respectfully submitted to the community to help foster the mutual interactions of the breast cancer world with micro- and nano-technology, and in particular to encourage the latter community to direct ever increasing attention to breast cancer, where an extraordinary beneficial impact may result. The paper initiates with an introductory overview of breast cancer, its current treatment modalities, and the current role of nanotechnology in the clinic. Our perspectives are then presented on what the greatest opportunities for nanotechnology are; this follows from an analysis of the role of biological barriers that adversely determine the biological distribution of intravascularly injected therapeutic agents. Different generations of nanotechnology tools for drug delivery are reviewed, and our current strategy for addressing the sequential bio-barriers is also presented, and is accompanied by an encouragement to the community to develop even more effective ones.


Asunto(s)
Neoplasias de la Mama/terapia , Nanotecnología/métodos , Femenino , Humanos
9.
J Exp Ther Oncol ; 7(4): 299-312, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19227010

RESUMEN

Inflammatory breast cancer (IBC) is the most aggressive form of locally advanced breast cancer (LABC) characterized by rapid growth and aggressive invasion with no selective therapies developed to treat IBC. Cyclooxygenase-2 (Cox-2), which produces prostaglandin E2 (PGE2) is known to be upregulated in primary IBC tumors and metastatic lesions, however the use of selective Cox-2 inhibitors has diminished due to cardiovascular side effects. One alternative approach to targeting Cox-2 enzyme activity is to block binding of the PGE2 ligand to its prostanoid (EP) receptors, which are designated as EP1, EP2, EP3, and EP4 and are members of a subfamily of G protein coupled receptors (GPCRs). While SUM149 IBC tumor cells and MCF-7 non-IBC breast tumor cells produce both EP2 and EP4 receptors, the invasive MDA-MB-231 non-IBC breast tumor cells produced low but detectable levels of these receptors. PGE2 and the EP4 agonist, PGE2 alcohol, stimulated significantly increased (p < 0.05) levels of proliferation and invasion by SUM149 IBC tumor cells, with no effect on proliferation of either of the two non-IBC breast tumor cell lines. In contrast, the EP2 agonist butaprost had no effect on proliferation or invasion of any cell line examined. The selective EP4 antagonist, GW627368X, induced inhibition of proliferation and invasion of human SUM149 IBC tumor cells beginning at 0.1 microM, with inhibition of proliferation and invasion by MDA-MB-231 non-IBC cells at higher concentrations of GW627368X. Molecular knockdown of the EP4 receptor was accomplished by stable transfection of an EP4 short hairpin RNA (shRNA) construct, with a clonally derived cell line designated as SUM149/Clone 1 exhibiting significantly slowed proliferation and diminished invasion compared to SUM149/Vector 5 which contained a scrambled shRNA control vector. This is the first report using both a selective pharmacologic inhibitor and a molecular shRNA knockdown approach to demonstrate that EP4 is directly involved in regulation of proliferation and invasion of IBC cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Receptores de Prostaglandina E/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Isoindoles/farmacología , Modelos Biológicos , Invasividad Neoplásica , Subtipo EP4 de Receptores de Prostaglandina E , Sulfonamidas/farmacología , Factores de Tiempo
10.
Anticancer Res ; 27(2): 719-27, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17465194

RESUMEN

Based on our studies demonstrating first time evidence that the cyclooxygenase-2 (Cox-2) enzyme is abundant within invasive human breast tumors, we developed a clonally derived human breast tumor cell clone designated as MCF-7/Cox-2 Clone 10 by transfection of human Cox-2 cDNA into slow growing, Cox-2 null, non-metastatic MCF-7 human breast tumor cells. The present studies evaluated the biological characteristics of the MCF-7/Cox-2 Clone 10 human breast tumors compared to the characteristics of MCF-7/empty vector control tumors when grown in vivo following injection of 5x10(6) tumor cells into mammary fat pads of ovariectomized female Crl:Nu-Foxn1(nu) mice implanted with slow release 17-beta estradiol pellets. At 60 days after tumor cell injection, MCF-7/Cox-2 Clone 10 human breast tumors were 4-fold greater (p < 0.01) in volume than MCF-7/empty vector control tumors. MCF-7/Cox-2 Clone 10 human breast tumor xenografts were highly angiogenic based on histological observation of large-bore blood vessels, which was confirmed by immunohistochemical staining with anti-CD-31 antibody and quantitation of mean vessel density. MCF-7/Cox-2 Clone 10 human breast tumor cells were present within regional lymph nodes adjacent to mammary fat pads with their local invasion confirmed by Western blotting of Cox-2-protein. This unique Cox-2-dependent breast tumor model rapidly produces large, angiogenic, locally invasive human breast tumor xenografts in mammary fat pads of ovariectomized female Crl:Nu-Foxn1(nu) mice at 42-60 days which recapitulate human breast ductal carcinomas. This unique model may be invaluable as a means to evaluate preclinical safety and efficacy of novel adjuvant therapies for women with metastastic breast cancer including prostanoid receptor antagonists, newly developed anti-angiogenic therapies, as well as other novel approaches for targeting and destruction of human breast tumors and their vasculature.


Asunto(s)
Neoplasias de la Mama/enzimología , Carcinoma Ductal de Mama/enzimología , Ciclooxigenasa 2/metabolismo , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/irrigación sanguínea , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Femenino , Humanos , Inmunohistoquímica , Metástasis Linfática , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transfección , Trasplante Heterólogo
11.
Prostaglandins Other Lipid Mediat ; 81(1-2): 55-70, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16997132

RESUMEN

The present studies evaluated the direct effects of the presence of human cyclooxygenase-2 (Cox-2) on gene expression of specific promoter regions of the P450 Cyp19 enzyme aromatase enzyme and its product, estradiol, in Cox-2 null estrogen-dependent MCF-7 breast tumor cells and in a stable clone of MCF-7 cells containing transfected Cox-2 cDNA, designated as MCF-7/Cox-2 Clone 10. Clone 10 human breast tumor cells have significantly increased gene expression of total mRNA of the P450 Cyp19 enzyme aromatase, with high levels of gene expression of specific aromatase promoter (p) regions pII, pI.3, and p1.7, with no significant change in mRNA levels of p1.4. Clone 10 human breast tumor cells produced significantly increased amounts of both prostaglandin E2 (PGE2) derived from Cox-2 enzyme activity and estradiol derived from aromatase enzyme activity (p<0.01), compared to MCF-7/vector control cells. The greatest inhibition of PGE2 or estradiol production was observed by the combination of the selective Cox-2 inhibitor celecoxib (25 microM) and the aromatase inhibitor, formestane (10nM) (p<0.01). The greatest anti-proliferative effect in Cox-2 null MCF-7/vector control cells was observed with the combination of 25 microM celecoxib and 10nM formestane but not with 10 microM celecoxib, suggesting that there are Cox-2-independent mechanisms involved in the anti-proliferative effect of this agent at doses greater than 10 microM. Celecoxib (25 microM) also significantly inhibited proliferation of MCF-7/Cox-2 Clone 10 human breast tumor cells, with no further anti-proliferative activity with the addition of 10 nM formestane observed at either 24 or 48 h of treatment. These studies demonstrate that Cox-2 directly regulates gene expression of specific aromatase promoter regions and regulates aromatase enzyme activity. Agents that inhibit Cox-2 or block the biological effects of PGE2 may be useful in significantly limiting aromatase activity and proliferation of human breast tumor cells regardless of the presence of Cox-2. In addition, the unique human breast tumor cell model used in these studies may be a useful tool in identifying the spectrum of activities of agents that block the biological effects of PGE2 and estradiol.


Asunto(s)
Aromatasa/genética , Neoplasias de la Mama/genética , Ciclooxigenasa 2/metabolismo , Estradiol/metabolismo , Regulación Enzimológica de la Expresión Génica , Regiones Promotoras Genéticas , Androstenodiona/análogos & derivados , Androstenodiona/metabolismo , Aromatasa/metabolismo , Inhibidores de la Aromatasa/metabolismo , Neoplasias de la Mama/metabolismo , Celecoxib , Línea Celular Tumoral , Proliferación Celular , Inhibidores de la Ciclooxigenasa/metabolismo , Femenino , Humanos , Pirazoles/metabolismo , Sulfonamidas/metabolismo
12.
Anticancer Res ; 26(4B): 2805-20, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16886599

RESUMEN

BACKGROUND: The goal of the present studies was to localize two proteins known to be involved in regulation of cell proliferation and survival in specific cell populations in normal SENCAR mouse skin and during multi-stage skin carcinogenesis. The proteins evaluated included activated Akt, as defined by phosphorylation of Akt at Serine-473 (pAkt) and mammalian target of rapamycin (pmTOR), defined by phosphorylation of mTOR at Serine-2448 (pmTOR). The cell populations examined included mouse keratinocyte stem cells (KSCs) within hair follicles and preneoplastic papilloma cells. MATERIALS AND METHODS: Immunochemical staining analysis as well as triple color immunofluorescence in combination with confocal microscopy were used to evaluate the presence of activated Akt and mammalian target of rapamycin (mTOR) in KSCs within the bulge niche of hair follicles, as identified by expression of the specific markers of mouse KSCs, CD34 and cytokeratin 15 (K15). Western blot analysis was used to examine CD34 and K15 protein levels in dorsal skin isolated from SENCAR mice during multi-stage skin carcinogenesis. RESULTS: CD34+/K15+ KSCs were located only in the outer root sheath (ORS) of a specific niche within hair follicles defined as "the bulge". The location of CD34+/K15+ KSCs remained restricted to the bulge region throughout the 22-week time-period examined during which pre-malignant papillomas developed and rapidly expanded. There was a significant decrease in K15 protein levels at 24 h and 15 weeks in dorsal skin treated with DMBA/TPA compared to CD34 protein levels. CD34+ cells within the numerous hair follicles in hyperplastic skin were found to undergo proliferation during the process of multi-stage skin carcinogenesis based on their staining with antibodies directed against proliferating cell nuclear antigen (PCNA). While pAkt was present within the bulge region of hair follicles, pmTOR was present in cells in the ORS of the bulge region as well as the upper infundibulum of hair follicles in dorsal skin treated with acetone. Within papillomas tissues isolated at 15 weeks following DMBA/TPA treatment, pAkt was localized to suprabasal cells with nominal staining of pAkt in the basal cell layer. There were fewer cells within the basal cell layer that contained pmTOR, in addition to the presence of pmTOR in suprabasal cells within papillomas. CONCLUSION: These results provide first time evidence for pAkt and pmTOR in CD34+/K15+ KSCs localized to the outer root sheath niche of the bulge region of mouse hair follicles. Taken together, the present observations suggest that pAkt and pmTOR may allow this cell population to evade terminal differentiation and to persist for long periods of time in their specific niche. Strategies that target pAkt and pmTOR may deplete both cells within the CD34+/K]5+ KSCs compartment, as well as impacting the survival of nonproliferating suprabasal cells within pre-malignant papillomas.


Asunto(s)
Antígenos CD34/biosíntesis , Queratinocitos/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/metabolismo , Células Madre/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Activación Enzimática , Femenino , Folículo Piloso/citología , Folículo Piloso/metabolismo , Queratinocitos/patología , Ratones , Ratones Endogámicos SENCAR , Papiloma/metabolismo , Papiloma/patología , Fosforilación , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transducción de Señal , Neoplasias Cutáneas/patología , Células Madre/patología , Serina-Treonina Quinasas TOR
13.
DNA Repair (Amst) ; 2(5): 471-82, 2003 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-12713808

RESUMEN

The ability to sensitize cancer cells to radiation would be highly beneficial for successful cancer treatment. One mode of action for ionizing radiation is the induction of cell death through infliction of extensive oxidative damage to cellular DNA, including mitochondrial DNA (mtDNA). The ability of cells to repair mtDNA and otherwise maintain the integrity of their mitochondria is vital for protection of the cells against oxidative damage. Because efficient repair of oxidative damage in mtDNA may play a crucial role in cancer cell resistance, interference with this repair process could be an effective way to achieve a radiation sensitive phenotype in otherwise resistant cancer cells. Successful repair of DNA is achieved through a precise and highly regulated multistep process. Expression of excessive amounts of one of the repair enzymes may cause an imbalance of the whole repair system and lead to the loss of repair efficiency. To study the effects of changing mtDNA repair capacity on overall cell survival following oxidative stress, we expressed a bacterial repair enzyme, Exonuclease III (ExoIII) containing the mitochondrial targeting signal of manganese superoxide dismutase, in a human malignant breast epithelial cell line, MDA-MB-231. Following transfection, specific exonuclease activity was found in mitochondrial extracts. In order to examine the effects on repair of oxidative damage in mtDNA, cells were exposed to the enzyme xanthine oxidase and its substrate hypoxanthine. mtDNA repair was evaluated using quantitative Southern blot analysis. The results revealed that cells expressing ExoIII in mitochondria are deficient in mtDNA repair when compared with control cells that express ExoIII without MTS. This diminished mtDNA repair capacity rendered MDA-MB-231 cells more sensitive to oxidative damage, which resulted in a decrease in their long-term survival following oxidative stress.


Asunto(s)
Neoplasias de la Mama/metabolismo , Reparación del ADN , ADN Mitocondrial/metabolismo , Escherichia coli/enzimología , Exodesoxirribonucleasas/biosíntesis , Mitocondrias/metabolismo , Estrés Oxidativo , Southern Blotting , Western Blotting , Línea Celular Tumoral , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Humanos , Fenotipo , Reacción en Cadena de la Polimerasa , Radiación Ionizante , Especies Reactivas de Oxígeno , Espectrofotometría , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Transfección , Xantina Oxidasa/metabolismo
14.
Prostaglandins Other Lipid Mediat ; 73(3-4): 249-64, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15290792

RESUMEN

To evaluate the direct effect of human cyclooxygenase-2 (hCox-2) on human breast tumor cell proliferation, invasion, and angiogenesis, hCox-2 cDNA was transfected into slow growing, non-metastatic MCF-7 human breast tumor cells that express low levels of Cox-2. Two stable transfectant clones, designated MCF-7/hCox-2 clones 8 and 10, had significantly decreased (P < 0.05) doubling time, with two-fold greater number of cells during exponential growth compared to the MCF-7/vector control. Proliferation of both of the MCF-7/hCox-2 clones was significantly inhibited in a time- and dose-dependent manner by celecoxib. The MCF-7/hCox-2 clones 8 and 10 formed larger and greater numbers of colonies in soft agar than the MCF-7/vector control, with a corresponding increased invasion across an artificial Matrigel basement membrane in response to recombinant human epidermal growth factor (hEGF). The MCF-7/hCox-2 clones 8 and 10 had higher mRNA levels of two splice variants of vascular endothelial growth factor (VEGF), V145 and V165. These results demonstrate that hCox-2 directly increases breast tumor cell proliferation, stimulates invasion across a basement membrane, and induces synthesis of specific heparin binding splice variants of VEGF.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Neoplasias de la Mama/enzimología , Celecoxib , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Colágeno , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Combinación de Medicamentos , Humanos , Laminina , Proteínas de la Membrana , Invasividad Neoplásica , Neovascularización Patológica , Prostaglandina-Endoperóxido Sintasas/genética , Proteoglicanos , Pirazoles/farmacología , Sulfonamidas/farmacología , Transfección , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
15.
Anticancer Res ; 23(2B): 1289-95, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12820385

RESUMEN

Head and neck cancers are significant due to their high morbidity and associated complications. We report, for the first time, that endostatin directly affects epithelial lineage human cells derived from tumors of patients with head and neck squamous cell carcinoma (HNSCC). This study investigated endostatin's effects on several HNSCC cellular functions that are essential for tumor progression. We determined that exposure of HNSCC cells to endostatin activated the transcription-activating factors, NF-xB and AP-1 in a cell-line-dependent fashion. Endostatin also down-regulated the gene expression of several pro-migratory molecules. Migration and invasion assays showed that endostatin significantly inhibited these functions that are essential for tumor progression. Fluorescent labeling studies showed endostatin co-localized to tropomysin-binding HNSCC the microfilaments, suggesting endostatin's suppression of HNSCC cell migration and invasion may reflect perturbation of the microfilament function. Our data imply that endostatin's clinical efficacy extends beyond angiostatic properties to encompass a direct anti-tumorigenic effect against HNSCC cells.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/patología , Colágeno/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Neoplasias de la Lengua/patología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Adulto , Anciano , Antineoplásicos/metabolismo , Movimiento Celular/efectos de los fármacos , Colágeno/metabolismo , Depresión Química , Endostatinas , Factor de Crecimiento Epidérmico/farmacología , Humanos , Integrinas/biosíntesis , Integrinas/genética , Masculino , Metaloproteinasa 10 de la Matriz , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/genética , Persona de Mediana Edad , FN-kappa B/metabolismo , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Fragmentos de Péptidos/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/genética , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Tropomiosina/metabolismo , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos
16.
Anticancer Res ; 24(5A): 2773-81, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15517884

RESUMEN

The goal of the present study was to identify specific populations of cells that contain activated Akt-1, as determined by the presence ofphosphorylated Akt at serine 473 (p Akt), during development of skin tumors using a murine multi-stage carcinogenesis model. Nucleated papillomas cells as well as both epidermal and follicular keratinocytes in hyperplastic skin contained increased pAkt compared to skin treated only with acetone or 7, 12 dimethylbenz[a]anthracene (DMBA). Although the numbers of both mast cells and neutrophils were significantly increased in the stroma of papillomas (p<0.0005; p<0.0001, respectively), only mast cells contained pAkt. The amount of total Akt protein was similar regardless of time or treatment group examined. The present results suggest that activation of Akt-1 may provide specific populations of epidermal keratinocytes that develop into skin tumors with the ability to resist terminal differentiation and have enhanced proliferation during multi-stage skin carcinogenesis. In addition, mast cells which contain activated Akt-1 may persist within the stroma of papillomas during skin tumor development and progression through this signaling pathway, thereby contributing to a pro-oxidant and proangiogenic microenvironment.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias Cutáneas/enzimología , Piel/enzimología , 9,10-Dimetil-1,2-benzantraceno , Acetona , Animales , Western Blotting , Carcinógenos , Activación Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Queratinocitos/enzimología , Queratinocitos/patología , Mastocitos/enzimología , Mastocitos/patología , Ratones , Ratones Endogámicos SENCAR , Neutrófilos/enzimología , Neutrófilos/patología , Papiloma/inducido químicamente , Papiloma/enzimología , Papiloma/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Piel/efectos de los fármacos , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología , Acetato de Tetradecanoilforbol
17.
In Vivo ; 18(5): 525-42, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15523889

RESUMEN

This review focuses on vascular endothelial growth factors (VEGF), a group of structurally-related proteins that serve as key growth factors for tumor-associated angiogenesis. Pathways induced by VEGF proteins that regulate biological functions of key cell types involved in tumor angiogenesis, including vascular endothelial cells, pericytes and tumor cells, are discussed. Strategies that are currently being developed and tested based on the emerging definitions of the roles of the multiple cell types involved in tumor vessel development, their selective production of VEGF-related proteins and other pro-angiogenic growth factors, their expression of associated receptors as well as identification of signal transduction pathways involved in VEGF-induced tumor survival and tumor-associated angiogenesis will be reviewed.


Asunto(s)
Progresión de la Enfermedad , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Humanos , Neoplasias/patología , Neoplasias/fisiopatología , Transducción de Señal
18.
Future Med Chem ; 6(17): 1909-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25495984

RESUMEN

Breast cancer is among the most commonly diagnosed cancer types in women worldwide and is the second leading cause of cancer-related disease in the USA. SH2 domains recruit signaling proteins to phosphotyrosine residues on aberrantly activated growth factor and cytokine receptors and contribute to cancer cell cycling, metastasis, angiogenesis and so on. Herein we review phosphopeptide mimetic and small-molecule approaches targeting the SH2 domains of Grb2, Grb7 and STAT3 that inhibit their targets and reduce proliferation in in vitro breast cancer models. Only STAT3 inhibitors have been evaluated in in vivo models and have led to tumor reduction. Taken together, these studies suggest that targeting SH2 domains is an important approach to the treatment of breast cancer.


Asunto(s)
Proteína Adaptadora GRB2/antagonistas & inhibidores , Proteína Adaptadora GRB7/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Dominios Homologos src , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB7/metabolismo , Humanos , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosfopéptidos/uso terapéutico , Unión Proteica , Factor de Transcripción STAT3/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/uso terapéutico
19.
Oncotarget ; 4(3): 446-62, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23530113

RESUMEN

E-Cadherin is a cell:cell adhesion molecule critical for appropriate embryonic and mammary development. In cancer, E-Cadherin has been primarily viewed as being lost during the process of epithelial-mesenchymal transition (EMT), which occurs with a switch from E-Cadherin expression to a gain of N-Cadherin and other mesenchymal markers. EMT has been shown to play a role in the metastatic process while the reverse process, mesenchymal-epithelial transition (MET), is important for metastatic colonization. Here we report an unexpected role of E-Cadherin in regulating tumorigenicity and hypoxia responses of breast tumors in vivo. Reduced expression of E-Cadherin led to a dramatic reduction of the in vivo growth capability of SUM149, Mary-X and 4T1 tumor cells. Furthermore, over-expression of ZEB1, a known transcriptional repressor of E-Cadherin, led to reduced in vivo growth of SUM149 tumors. Gene set enrichment analysis identified the loss of hypoxia response genes as a major mechanism in mediating the lack of in vivo growth of SUM149 cells that lacked E-Cadherin or over-expressed ZEB1. The in vivo growth defect of SUM149 E-Cadherin knockdown tumors was rescued by the hypoxia-inducible 1α transcription factor (HIF-1α). Given the importance of HIF-1α in cellular metabolism, we observed reduced glycolytic capacity in SUM149 and 4T1 cells that had E-Cadherin knocked down. Our observations shed light on the complex functions of E-Cadherin in retention of an epithelial phenotype and as a mediator of survival of aggressive breast cancer under hypoxic conditions in vivo. Furthermore, we find that patients with basal subtype breast cancer and high E-Cadherin expression in their tumors had a poor clinical outcome. Our data suggests a novel function for E-Cadherin as a bona fide signaling molecule required for the in vivo growth of aggressive breast cancer tumor cells, that retain E-Cadherin expression, in mediating their metabolic function.


Asunto(s)
Neoplasias de la Mama/genética , Cadherinas/genética , Metabolismo Energético/genética , Microambiente Tumoral/genética , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Glucólisis/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células MCF-7 , Metabolómica , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
20.
Clin Cancer Res ; 19(17): 4685-96, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23396049

RESUMEN

BACKGROUND: Inflammatory breast cancer (IBC) is a poorly characterized form of breast cancer. So far, the results of expression profiling in IBC are inconclusive due to various reasons including limited sample size. Here, we present the integration of three Affymetrix expression datasets collected through the World IBC Consortium allowing us to interrogate the molecular profile of IBC using the largest series of IBC samples ever reported. EXPERIMENTAL DESIGN: Affymetrix profiles (HGU133-series) from 137 patients with IBC and 252 patients with non-IBC (nIBC) were analyzed using unsupervised and supervised techniques. Samples were classified according to the molecular subtypes using the PAM50-algorithm. Regression models were used to delineate IBC-specific and molecular subtype-independent changes in gene expression, pathway, and transcription factor activation. RESULTS: Four robust IBC-sample clusters were identified, associated with the different molecular subtypes (P<0.001), all of which were identified in IBC with a similar prevalence as in nIBC, except for the luminal A subtype (19% vs. 42%; P<0.001) and the HER2-enriched subtype (22% vs. 9%; P<0.001). Supervised analysis identified and validated an IBC-specific, molecular subtype-independent 79-gene signature, which held independent prognostic value in a series of 871 nIBCs. Functional analysis revealed attenuated TGF-ß signaling in IBC. CONCLUSION: We show that IBC is transcriptionally heterogeneous and that all molecular subtypes described in nIBC are detectable in IBC, albeit with a different frequency. The molecular profile of IBC, bearing molecular traits of aggressive breast tumor biology, shows attenuation of TGF-ß signaling, potentially explaining the metastatic potential of IBC tumor cells in an unexpected manner.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Inflamatorias de la Mama/genética , Receptor ErbB-2/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA