Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
CA Cancer J Clin ; 60(6): 351-75, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20959401

RESUMO

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.


Assuntos
Neoplasias Inflamatórias Mamárias/diagnóstico , Neoplasias Inflamatórias Mamárias/terapia , Biomarcadores Tumorais/genética , Índice de Massa Corporal , Quimioterapia Adjuvante , Terapia Combinada/métodos , Diagnóstico Diferencial , Progressão da Doença , Feminino , Humanos , Incidência , Neoplasias Inflamatórias Mamárias/epidemiologia , Neoplasias Inflamatórias Mamárias/genética , Imageamento por Ressonância Magnética , Mamografia , Estadiamento de Neoplasias , Obesidade/complicações , Tomografia por Emissão de Pósitrons , Prognóstico , Radioterapia Adjuvante , Doenças Raras , Medição de Risco , Fatores de Risco , Taxa de Sobrevida , Tomografia Computadorizada por Raios X , Ultrassonografia Mamária , Estados Unidos/epidemiologia
2.
J Proteome Res ; 12(6): 2805-17, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23647160

RESUMO

In this study we selected three breast cancer cell lines (SKBR3, SUM149 and SUM190) with different oncogene expression levels involved in ERBB2 and EGFR signaling pathways as a model system for the evaluation of selective integration of subsets of transcriptomic and proteomic data. We assessed the oncogene status with reads per kilobase per million mapped reads (RPKM) values for ERBB2 (14.4, 400, and 300 for SUM149, SUM190, and SKBR3, respectively) and for EGFR (60.1, not detected, and 1.4 for the same 3 cell lines). We then used RNA-Seq data to identify those oncogenes with significant transcript levels in these cell lines (total 31) and interrogated the corresponding proteomics data sets for proteins with significant interaction values with these oncogenes. The number of observed interactors for each oncogene showed a significant range, e.g., 4.2% (JAK1) to 27.3% (MYC). The percentage is measured as a fraction of the total protein interactions in a given data set vs total interactors for that oncogene in STRING (Search Tool for the Retrieval of Interacting Genes/Proteins, version 9.0) and I2D (Interologous Interaction Database, version 1.95). This approach allowed us to focus on 4 main oncogenes, ERBB2, EGFR, MYC, and GRB2, for pathway analysis. We used bioinformatics sites GeneGo, PathwayCommons and NCI receptor signaling networks to identify pathways that contained the four main oncogenes and had good coverage in the transcriptomic and proteomic data sets as well as a significant number of oncogene interactors. The four pathways identified were ERBB signaling, EGFR1 signaling, integrin outside-in signaling, and validated targets of C-MYC transcriptional activation. The greater dynamic range of the RNA-Seq values allowed the use of transcript ratios to correlate observed protein values with the relative levels of the ERBB2 and EGFR transcripts in each of the four pathways. This provided us with potential proteomic signatures for the SUM149 and 190 cell lines, growth factor receptor-bound protein 7 (GRB7), Crk-like protein (CRKL) and Catenin delta-1 (CTNND1) for ERBB signaling; caveolin 1 (CAV1), plectin (PLEC) for EGFR signaling; filamin A (FLNA) and actinin alpha1 (ACTN1) (associated with high levels of EGFR transcript) for integrin signalings; branched chain amino-acid transaminase 1 (BCAT1), carbamoyl-phosphate synthetase (CAD), nucleolin (NCL) (high levels of EGFR transcript); transferrin receptor (TFRC), metadherin (MTDH) (high levels of ERBB2 transcript) for MYC signaling; S100-A2 protein (S100A2), caveolin 1 (CAV1), Serpin B5 (SERPINB5), stratifin (SFN), PYD and CARD domain containing (PYCARD), and EPH receptor A2 (EPHA2) for PI3K signaling, p53 subpathway. Future studies of inflammatory breast cancer (IBC), from which the cell lines were derived, will be used to explore the significance of these observations.


Assuntos
Neoplasias da Mama/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/genética , RNA Mensageiro/genética , Receptor ErbB-2/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Inflamação , Anotação de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Proteômica , RNA Mensageiro/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais
3.
Breast Cancer Res Treat ; 140(1): 23-33, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23784380

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/patologia , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Quinase do Linfoma Anaplásico , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Antígenos CD4/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos Par 8 , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal , Feminino , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Genes myc , Humanos , Neoplasias Inflamatórias Mamárias/metabolismo , Perda de Heterozigosidade , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores Proteína Tirosina Quinases/genética , Receptor Notch3 , Receptores Notch/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Exp Ther Oncol ; 10(3): 219-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24416998

RESUMO

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.


Assuntos
Depsipeptídeos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , Células Neoplásicas Circulantes/efeitos dos fármacos , Paclitaxel/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Depsipeptídeos/administração & dosagem , Sinergismo Farmacológico , Feminino , Humanos , Neoplasias Inflamatórias Mamárias/patologia , Camundongos , Metástase Neoplásica/prevenção & controle , Esferoides Celulares , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Breast Cancer Res Treat ; 134(2): 495-510, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22547109

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Benzazepinas/farmacologia , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , Radiossensibilizantes/farmacologia , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Aldeído Desidrogenase/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Antígenos CD/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Notch/antagonistas & inibidores , Esferoides Celulares
6.
J Exp Ther Oncol ; 10(2): 155-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23350355

RESUMO

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.


Assuntos
Biomimética , Neoplasias da Mama/prevenção & controle , Neovascularização Patológica/prevenção & controle , Fosfopeptídeos/farmacologia , Pró-Fármacos/farmacologia , Fator de Transcrição STAT3/metabolismo , Domínios de Homologia de src/efeitos dos fármacos , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
NPJ Breast Cancer ; 8(1): 12, 2022 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-35042871

RESUMO

Inflammatory breast cancer (IBC) is an aggressive disease for which the spectrum of preclinical models was rather limited in the past. More recently, novel cell lines and xenografts have been developed. This study evaluates the transcriptome of an extended series of IBC preclinical models and performed a comparative analysis with patient samples to determine the extent to which the current models recapitulate the molecular characteristics of IBC observed clinically. We demonstrate that the IBC preclinical models are exclusively estrogen receptor (ER)-negative and of the basal-like subtype, which reflects to some extent the predominance of these subtypes in patient samples. The IBC-specific 79-signature we previously reported was retrained and discriminated between IBC and non-IBC preclinical models, but with a relatively high rate of false positive predictions. Further analyses of gene expression profiles revealed important roles for cell proliferation, MYC transcriptional activity, and TNFɑ/NFκB in the biology of IBC. Patterns of MYC expression and transcriptional activity were further explored in patient samples, which revealed interactions with ESR1 expression that are contrasting in IBC and nIBC and notable given the comparatively poor outcomes of ER+ IBC. Our analyses also suggest important roles for NMYC, MXD3, MAX, and MLX in shaping MYC signaling in IBC. Overall, we demonstrate that the IBC preclinical models can be used to unravel cancer cell intrinsic molecular features, and thus constitute valuable research tools. Nevertheless, the current lack of ER-positive IBC models remains a major hurdle, particularly since interactions with the ER pathway appear to be relevant for IBC.

8.
Breast Cancer Res Treat ; 125(3): 785-95, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21153052

RESUMO

The goal of this study was to determine whether gene expression differences exist between inflammatory breast cancers (IBC) and T stage-matched non-IBC patients stratified by hormone receptor and HER2 status. We used Affymetrix GeneChips to analyze 82 tumor samples (25 T4d patients, and 57 T4a-c patients) of newly diagnosed breast cancers. Genes that were differentially expressed between the IBC and non-IBC specimens were identified using the t test, and differential expression of gene sets was assessed using gene set analysis. Three distinct clinical subtypes of IBC and non-IBC were compared: ER-positive/HER2-normal, HER2-amplified, and ER-negative/HER2-normal. When we compared expression data from all IBC with all non-IBC, we found no significant differences after adjusting for multiple testing. When IBC and non-IBC tumors were compared by clinical subtype, however, significant differences emerged. Complement and immune system-related pathways were overexpressed in ER-positive/HER2-normal IBC. Protein translation and mTOR signaling were overexpressed in HER2-amplified IBC. Apoptosis-, neural-, and lipid metabolism-related pathways were overexpressed in ER-negative/HER2-normal IBC compared with non-IBC of the same receptor phenotype. In this T stage-matched case-control study, the survival curves of patients with IBC and non-IBC were similar for all three clinical subtypes. IBC tumors can be divided into molecular and clinical subtypes similar to those of non-IBC. Clinical subtypes of IBC show molecular differences compared with similar subtypes of non-IBC.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Inflamatórias Mamárias/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Estudos de Coortes , Feminino , Humanos , Neoplasias Inflamatórias Mamárias/diagnóstico , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
9.
Bioorg Med Chem Lett ; 21(20): 6071-3, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21903394

RESUMO

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.


Assuntos
Inibidores de Ciclo-Oxigenase 2/síntese química , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/antagonistas & inibidores , Neoplasias Inflamatórias Mamárias/tratamento farmacológico , Pirróis/síntese química , Pirróis/farmacologia , Sulfonamidas/síntese química , Sulfonamidas/farmacologia , Linhagem Celular Tumoral , Feminino , Humanos
10.
Future Oncol ; 7(1): 25-30, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21174535

RESUMO

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.


Assuntos
Neoplasias Inflamatórias Mamárias/terapia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/mortalidade
11.
Mol Cancer ; 9: 180, 2010 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-20615238

RESUMO

BACKGROUND: Since the first suggestion of prospectively identifiable cancer stem cells in solid tumors, efforts have been made to characterize reported cancer stem cell surrogates in existing cancer cell lines, and cell lines rich with these surrogates have been used to screen for cancer stem cell targeted agents. Although 293T cells were derived from human embryonic kidney, transplantation of these cells into the mammary fat pad yields aggressive tumors that self-renew as evidenced by serial xenograft passages through transplantation. Herein we fully characterize cancer stem cell-like features in 293T human embryonic kidney cells. RESULTS: 293T cells can be readily cultured and passaged as spheres in serum-free stem cell promoting culture conditions. Cells cultured in vitro as three-dimensional spheres (3D) were shown to contain higher ALDH1 and CD44+/CD24- population compared to monolayer cells. These cells were also resistant to radiation and upregulate stem cell survival signaling including beta-catenin, Notch1 and Survivin in response to radiation. Moreover, 3D spheres generated from the 293T cells have increased expression of mesenchymal genes including vimentin, n-cadherin, zeb1, snail and slug as well as pro-metastatic genes RhoC, Tenascin C and MTA1. In addition, microRNAs implicated in self-renewal and metastases were markedly reduced in 3D spheres. CONCLUSIONS: 293T cells exhibit a cancer stem cell-like phenotype when cultured as 3D spheres and represent an important research tool for studying the molecular and biological mechanisms of cancer stem cells and for testing and developing novel targets for cancer therapy.


Assuntos
Rim/embriologia , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Linhagem Celular , Meios de Cultura Livres de Soro , Humanos , Rim/citologia , MicroRNAs/genética , Metástase Neoplásica/patologia , Estudos Prospectivos , Regulação para Cima
12.
Biomed Microdevices ; 11(1): 49-63, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18663578

RESUMO

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.


Assuntos
Neoplasias da Mama/terapia , Nanotecnologia/métodos , Feminino , Humanos
13.
J Exp Ther Oncol ; 7(4): 299-312, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19227010

RESUMO

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.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Receptores de Prostaglandina E/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Isoindóis/farmacologia , Modelos Biológicos , Invasividade Neoplásica , Receptores de Prostaglandina E Subtipo EP4 , Sulfonamidas/farmacologia , Fatores de Tempo
14.
Med Clin North Am ; 91(5): 899-927, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17826110

RESUMO

Nanotechnology may hold the key to controlling many devastating diseases. In the fight against the pain, suffering, and death due to cancer, nanotechnology will allow earlier diagnosis and even prevention of malignancy at premalignant stages, in addition to providing multimodality treatment not possible with current conventional techniques. This review discusses nanotechnology already used in diagnostic and therapeutic applications for cancer. Also addressed are theoretic and evolving uses of nanotechnology, including multifunctional nanoparticles for imaging and therapy, nanochannel implants for controlled release of drugs, nanoscale devices for evaluation of proteomics and genomics, and diagnostic techniques that take advantage of physical changes in diseased tissue.


Assuntos
Antineoplásicos/uso terapêutico , Nanomedicina/métodos , Nanoestruturas/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Animais , Humanos , Prognóstico
15.
Anticancer Res ; 27(2): 719-27, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17465194

RESUMO

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.


Assuntos
Neoplasias da Mama/enzimologia , Carcinoma Ductal de Mama/enzimologia , Ciclo-Oxigenase 2/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/irrigação sanguínea , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/enzimologia , Neovascularização Patológica/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Transfecção , Transplante Heterólogo
16.
Prostaglandins Other Lipid Mediat ; 81(1-2): 55-70, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16997132

RESUMO

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.


Assuntos
Aromatase/genética , Neoplasias da Mama/genética , Ciclo-Oxigenase 2/metabolismo , Estradiol/metabolismo , Regulação Enzimológica da Expressão Gênica , Regiões Promotoras Genéticas , Androstenodiona/análogos & derivados , Androstenodiona/metabolismo , Aromatase/metabolismo , Inibidores da Aromatase/metabolismo , Neoplasias da Mama/metabolismo , Celecoxib , Linhagem Celular Tumoral , Proliferação de Células , Inibidores de Ciclo-Oxigenase/metabolismo , Feminino , Humanos , Pirazóis/metabolismo , Sulfonamidas/metabolismo
17.
Anticancer Res ; 26(4B): 2805-20, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16886599

RESUMO

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.


Assuntos
Antígenos CD34/biossíntese , Queratinócitos/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/metabolismo , Células-Tronco/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Ativação Enzimática , Feminino , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Endogâmicos SENCAR , Papiloma/metabolismo , Papiloma/patologia , Fosforilação , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/patologia , Células-Tronco/patologia , Serina-Treonina Quinases TOR
18.
DNA Repair (Amst) ; 2(5): 471-82, 2003 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12713808

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Reparo do DNA , DNA Mitocondrial/metabolismo , Escherichia coli/enzimologia , Exodesoxirribonucleases/biossíntese , Mitocôndrias/metabolismo , Estresse Oxidativo , Southern Blotting , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Humanos , Fenótipo , Reação em Cadeia da Polimerase , Radiação Ionizante , Espécies Reativas de Oxigênio , Espectrofotometria , Frações Subcelulares/metabolismo , Fatores de Tempo , Transfecção , Xantina Oxidase/metabolismo
19.
Prostaglandins Other Lipid Mediat ; 73(3-4): 249-64, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15290792

RESUMO

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.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Neoplasias da Mama/enzimologia , Celecoxib , Linhagem Celular Tumoral , Proliferação de Células , Células Clonais , Colágeno , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Combinação de Medicamentos , Humanos , Laminina , Proteínas de Membrana , Invasividade Neoplásica , Neovascularização Patológica , Prostaglandina-Endoperóxido Sintases/genética , Proteoglicanas , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Transfecção , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
20.
Anticancer Res ; 23(2B): 1289-95, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12820385

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Colágeno/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Neoplasias da Língua/patologia , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Adulto , Idoso , Antineoplásicos/metabolismo , Movimento Celular/efeitos dos fármacos , Colágeno/metabolismo , Depressão Química , Endostatinas , Fator de Crescimento Epidérmico/farmacologia , Humanos , Integrinas/biossíntese , Integrinas/genética , Masculino , Metaloproteinase 10 da Matriz , Metaloendopeptidases/biossíntese , Metaloendopeptidases/genética , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Invasividade Neoplásica , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/metabolismo , Inibidor Tecidual de Metaloproteinase-2/biossíntese , Inibidor Tecidual de Metaloproteinase-2/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Tropomiosina/metabolismo , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA