Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Publication year range
1.
J Cancer ; 7(8): 947-56, 2016.
Article in English | MEDLINE | ID: mdl-27313785

ABSTRACT

Among patients with hormone receptor (HR)-positive breast cancer, those with residual disease after neoadjuvant chemotherapy have a higher risk of relapse and poorer survival than those with a complete response. Previous studies have revealed a correlation between activation of cell cycle-regulating pathways in HR-positive breast cancer, particularly cyclin-dependent kinase (CDK) 4 and 6/cyclin D1 signaling, and resistance to standard therapies. Although CDK4/6 inhibition by palbociclib in combination with endocrine therapy has shown potent antiproliferative effects in HR-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer, the potential role of palbociclib in re-sensitizing chemotherapy-resistant HR-positive breast cancer is not well defined. We hypothesized that CDK4/6 inhibition by palbociclib re-sensitizes HR-positive/HER2-negative residual breast cancer to taxane-based adjuvant therapy. Using cell counting, flow cytometry, and western blotting, we evaluated the efficacy of palbociclib alone and in concurrent or sequential combination with paclitaxel in parental and paclitaxel-resistant T47D HR-positive/HER2-negative breast cancer cells. The CDK4/6 pathway was constitutively active in both parental and paclitaxel-resistant T47D cells; thus, both cell types were highly sensitive to the inhibitory effects of single-agent palbociclib on cell growth and cell cycle progression. However, palbociclib did not re-sensitize resistant cells to paclitaxel-induced G2/M arrest and cell death in any of the combinations tested. Our results suggest that CDK4/6 inhibition by palbociclib does not re-sensitize HR-positive/HER2-negative residual breast cancer to chemotherapy. Nevertheless, the fact that CDK4/6 activation remained intact in paclitaxel-resistant cells indicates that patients who have HR-positive/HER2-negative residual disease after taxane-based neoadjuvant chemotherapy may still benefit from palbociclib in combination with other regimens, such as endocrine therapies, for adjuvant therapy.

2.
Cancer Genomics Proteomics ; 9(5): 297-310, 2012.
Article in English | MEDLINE | ID: mdl-22990109

ABSTRACT

Breast cancer (BC) remains the most common cancer type diagnosed in women. Although targeted therapies have improved patient survival for advanced BC, these tumors frequently relapse due to drug resistance mechanisms. A systems biology approach integrates DNA, RNA and protein alterations generated from multidimensional platforms to better understand the mechanisms that regulate the metastatic process. Downstream functional analyses in pre-clinical studies might integrate the role of these aberrations into the cell, leading to discovery of new therapeutic targets. In the present report, we review relevant findings associated with genomic, transcriptomic and proteomic analyses and the contribution of the systems biology concept to the interpretation of these data in the metastatic context. Also, we highlight the importance of re-designing clinical trials towards metastasis prevention for improvement of personalized care.


Subject(s)
Breast Neoplasms/drug therapy , Genes, Neoplasm , Neoplasm Metastasis/drug therapy , Precision Medicine/methods , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Clinical Trials as Topic , DNA Copy Number Variations , DNA Methylation , DNA, Neoplasm/genetics , Female , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Proteome/analysis , Systems Biology/methods , Transcriptome , Tumor Microenvironment
3.
Mol Cancer Ther ; 11(2): 464-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22084168

ABSTRACT

HER-2-positive breast cancers frequently sustain elevated AKT/mTOR signaling, which has been associated with resistance to doxorubicin treatment. Here, we investigated whether rapamycin, an mTOR inhibitor, increased the sensitivity to doxorubicin therapy in two HER-2-overexpressing cell lines: C5.2, which was derived from the parental HB4a by transfection with HER-2 and SKBR3, which exhibits HER-2 amplification. The epithelial mammary cell line HB4a was also analyzed. The combined treatment using 20 nmol/L of rapamycin and 30 nmol/L of doxorubicin arrested HB4a and C5.2 cells in S to G(2)-M, whereas SKBR3 cells showed an increase in the G(0)-G(1) phase. Rapamycin increased the sensitivity to doxorubicin in HER-2-overexpressing cells by approximately 2-fold, suggesting that the combination displayed a more effective antiproliferative action. Gene expression profiling showed that these results might reflect alterations in genes involved in canonical pathways related to purine metabolism, oxidative phosphorylation, protein ubiquitination, and mitochondrial dysfunction. A set of 122 genes modulated by the combined treatment and specifically related to HER-2 overexpression was determined by finding genes commonly regulated in both C5.2 and SKBR3 that were not affected in HB4a cells. Network analysis of this particular set showed a smaller subgroup of genes in which coexpression pattern in HB4a cells was disrupted in C5.2 and SKBR3. Altogether, our data showed a subset of genes that might be more robust than individual markers in predicting the response of HER-2-overexpressing breast cancers to doxorubicin and rapamycin combination.


Subject(s)
Doxorubicin/pharmacology , Gene Expression Profiling , Receptor, ErbB-2/genetics , Sirolimus/pharmacology , Antibiotics, Antineoplastic/pharmacology , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Regulatory Networks/drug effects , Humans , Models, Genetic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
PLoS One ; 6(6): e21022, 2011.
Article in English | MEDLINE | ID: mdl-21731642

ABSTRACT

We report the first quantitative and qualitative analysis of the poly (A)⁺ transcriptome of two human mammary cell lines, differentially expressing (human epidermal growth factor receptor) an oncogene over-expressed in approximately 25% of human breast tumors. Full-length cDNA populations from the two cell lines were digested enzymatically, individually tagged according to a customized method for library construction, and simultaneously sequenced by the use of the Titanium 454-Roche-platform. Comprehensive bioinformatics analysis followed by experimental validation confirmed novel genes, splicing variants, single nucleotide polymorphisms, and gene fusions indicated by RNA-seq data from both samples. Moreover, comparative analysis showed enrichment in alternative events, especially in the exon usage category, in ERBB2 over-expressing cells, data indicating regulation of alternative splicing mediated by the oncogene. Alterations in expression levels of genes, such as LOX, ATP5L, GALNT3, and MME revealed by large-scale sequencing were confirmed between cell lines as well as in tumor specimens with different ERBB2 backgrounds. This approach was shown to be suitable for structural, quantitative, and qualitative assessment of complex transcriptomes and revealed new events mediated by ERBB2 overexpression, in addition to potential molecular targets for breast cancer that are driven by this oncogene.


Subject(s)
Breast/cytology , Breast/metabolism , Gene Expression Profiling , Poly A/metabolism , Receptor, ErbB-2/metabolism , Alternative Splicing/genetics , Base Sequence , Cell Line , Computational Biology , Female , Gene Fusion/genetics , Gene Library , Genome, Human/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Receptor, ErbB-2/genetics , Reproducibility of Results
5.
São Paulo; s.n; 2011. 125 p. tab, ilus.
Thesis in Portuguese | LILACS | ID: lil-609342

ABSTRACT

A resistência à quimioterapia, a qual se constitui em um dos maiores desafios a serem superados durante o tratamento do cancer de mama, pode ser modulada por várias cascatas de sinalização cellular que levam ao aumento da sobrevivência celular. A via PI3K/Akt/mTOR é uma importante reguladora desse processo, visto que ela pode favorecer o crescimento tumoral após sua ativação por diversos receptores de fatores de crescimento, incluindo os receptores HER-2. Assim, nossa proposta foi investigar se a inibição dessa via poderia aumentar a sensibilidade de linhagens epiteliais mamárias humanas ao agente quimioterápico doxorrubicina. Também decidimos analisar o perfil de expressão gênica gerado por essa abordagem com o objetivo de compreender os mecanismos moleculares associados à resposta biológica. A rapamicina (um inibidor de mTOR) e a doxorrubicina foram utilizadas individualmente ou em combinação no tratamento de três linhagens epiteliais mamárias humanas: HB4a, C5.2 e SKBR3. A linhagem HB4a, uma linhagem derivada de epitélio mamário normal, expressa níveis basais do receptor HER-2. As outras duas linhagens expressam altos níveis desse receptor: a linhagem SKBR3 (derivada de adenocarcinoma mamário), expressa constitutivamente esse receptor e a linhagem C5.2 foi derivada da linhagem HB4a pela transfecção com o cDNA full-lenght do gene HER-2. A células foram tratadas com 20 nM de rapamicina associada ou não a doses de 30 nM a 1000 nM de doxorrubicina por 24, 48 e 72 horas. Após 24 horas, os resultados de citometria de fluxo mostraram que, enquanto as linhagens SKBR3 e C5.2 apresentaram indução de parada na fase G0/G1 do ciclo celular, a linhagem HB4a não demonstrou alterações significativas na distribuição do ciclo celular pelo tratamento com rapamicina. Esses resultados sugeriram que as linhagens SKBR3 e C5.2 foram mais sensíveis à rapamicina quando comparadas à linhagem HB4a. A doxorrubicina, por sua vez, induziu parada na fase S-G2/M do ciclo celular...


Chemotherapy resistance, which is the major challenge to be overcome during breast cancer treatment, can be modulated by several signaling cascades that lead to increased cell survival. The PI3K/Akt/mTOR pathway is an important regulator of this process, as it can favour the tumor growth after its activation by several growth factor receptors, including HER-2 receptors. Thus, we proposed to investigate whether the inhibition of this pathway could improve the sensitivity of human HER-2-overexpressing breast epithelial cell lines to the chemotherapeutic agent doxorubicin. We also decided to analyze the gene expression profile generated by this approach to better understand the molecular mechanisms associated with the biological response. Rapamycin (a mTOR inhibitor) and doxorubicin were used individually or in combination on three human breast epithelial cell lines: HB4a, C5.2 e SKBR3. The HB4a cell, a normal mammary epithelial cell line, expresses basal levels of HER-2. The two other cell lines express high levels of this receptor: the SKBR3 cell (derived from a mammary adenocarcinoma) expresses constitutively this receptor.and the C5.2 cell was derived from HB4a cells transfected with the cDNA-full length of HER-2. The cells were treated with 20 M of rapamycin with or without 30nM to 1000nM of doxorubicin for 24, 48 and 72 hours. After 24 hours, the flow citometry results showed that while SKBR3 and C5.2 were arrested in G0/G1 phase, HB4a demonstrated no alterations in the cell cycle distribution after rapamycin treatment. These results showed that SKBR3 and C5.2 cells were more sensitive to rapamycin compared to HB4a. Doxorubicin, in turn, led the cells to a S-G2/M arrest which was greater in HB4a cells compared to SKBR3 and C5.2. Similarly to doxorubicin alone, the combined treatment also caused an increase of S-G2/M arrest in HB4a and C5.2 cells. So, doxorubicin effects were predominant to the rapamycin effects on the cell cycle distribution of the...


Subject(s)
Breast Neoplasms , Sirolimus
SELECTION OF CITATIONS
SEARCH DETAIL
...