Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Pharmaceuticals (Basel) ; 14(7)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34358096

ABSTRACT

Among the well-established alterations contributing to prostate cancer (PCa) pathogenesis, epigenetics is an important player in its development and aggressive disease state. Moreover, since no curative therapies are available for advanced stage disease, there is an urgent need for novel therapeutic strategies targeting this subset of patients. Thus, we aimed to evaluate the combined antineoplastic effects of DNA methylation inhibitor hydralazine and histone deacetylase inhibitors panobinostat and valproic acid in several prostate cell lines. The effect of these drugs was assessed in four PCa (LNCaP, 22Rv1, DU145 and PC-3) cell lines, as well as in non-malignant epithelial (RWPE-1) and stromal (WPMY-1) cell lines, using several assays to evaluate cell viability, apoptosis, proliferation, DNA damage and clonogenic potential. We found that exposure to each epidrug separately reduced viability of all PCa cells in a dose-dependent manner and that combined treatments led to synergic growth inhibitory effects, impacting also on colony formation, invasion, apoptotic and proliferation rates. Interestingly, antitumoral effects of combined treatment were particularly expressive in DU145 cells. We concluded that hydralazine and panobinostat attenuate malignant properties of PCa cells, constituting a potential therapeutic tool to counteract PCa progression.

2.
Biomedicines ; 9(8)2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34440180

ABSTRACT

Advanced prostate cancers frequently develop resistance to androgen-deprivation therapy with serious implications for patient survival. Considering their importance in this type of neoplasia, epigenetic modifications have drawn attention as alternative treatment strategies. The aim of this study was to assess the antitumoral effects of the combination of hydralazine, a DNA methylation inhibitor, with enzalutamide, an antagonist of the androgen receptor, in prostate cancer cell lines. Several biological parameters, such as cell viability, proliferation, DNA damage, and apoptosis, as well as clonogenic and invasive potential, were evaluated. The individual treatments with hydralazine and enzalutamide exerted growth-inhibitory effects in prostate cancer cells and their combined treatment displayed synergistic effects. The combination of these two drugs was very effective in decreasing malignant features of prostate cancer and may become an alternative therapeutic option for prostate cancer patient management.

3.
Biomark Res ; 8(1): 68, 2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33292587

ABSTRACT

BACKGROUND: Esophageal cancer (ECa) is the 7th most incident cancer and the 6th leading cause of cancer-related death. Most patients are diagnosed with locally advanced or metastatic disease, enduring poor survival. Biomarkers enabling early cancer detection may improve patient management, treatment effectiveness, and survival, are urgently needed. In this context, epigenetic-based biomarkers such as DNA methylation are potential candidates. METHODS: Herein, we sought to identify and validate DNA methylation-based biomarkers for early detection and prediction of response to therapy in ECa patients. Promoter methylation levels were assessed in a series of treatment-naïve ECa, post-neoadjuvant treatment ECa, and normal esophagus tissues, using quantitative methylation-specific PCR for COL14A1, GPX3, and ZNF569. RESULTS: ZNF569 methylation (ZNF569me) levels significantly differed between ECa and normal samples (p < 0.001). Moreover, COL14A1 methylation (COL14A1me) and GPX3 methylation (GPX3me) levels discriminated adenocarcinomas and squamous cell carcinomas, respectively, from normal samples (p = 0.002 and p = 0.009, respectively). COL14A1me & ZNF569me accurately identified adenocarcinomas (82.29%) whereas GPX3me & ZNF569me identified squamous cell carcinomas with 81.73% accuracy. Furthermore, ZNF569me and GPX3me levels significantly differed between normal and pre-treated ECa. CONCLUSION: The biomarker potential of a specific panel of methylated genes for ECa was confirmed. These might prove useful for early detection and might allow for the identification of minimal residual disease after adjuvant therapy.

4.
Int J Mol Sci ; 21(10)2020 May 15.
Article in English | MEDLINE | ID: mdl-32429269

ABSTRACT

Oesophageal cancer is a life-threatening disease, accounting for high mortality rates. The poor prognosis of this malignancy is mostly due to late diagnosis and lack of effective therapies for advanced disease. Epigenetic alterations may constitute novel and attractive therapeutic targets, owing to their ubiquity in cancer and their reversible nature. Herein, we offer an overview of the most important studies which compared differences in expression of enzymes that mediate epigenetic alterations between oesophageal cancer and normal mucosa, as well as in vitro data addressing the role of these genes/proteins in oesophageal cancer. Furthermore, The Cancer Genome Atlas database was interrogated for the correlation between expression of these epigenetic markers and standard clinicopathological features. We concluded that most epigenetic players studied thus far are overexpressed in tumours compared to normal tissue. Furthermore, functional assays suggest an oncogenic role for most of those enzymes, supporting their potential as therapeutic targets in oesophageal cancer.


Subject(s)
Epigenesis, Genetic , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Publications
5.
Gastric Cancer ; 23(5): 811-823, 2020 09.
Article in English | MEDLINE | ID: mdl-32215766

ABSTRACT

BACKGROUND: Gastric cancer is the fifth most common cancer and the third cause of global cancer mortality. CDX2 is an intestinal differentiation marker with prognostic value in gastric cancer and transcriptionally regulates the expression of glycoprotein A33 (GPA33) and liver intestine cadherin (LI-cadherin). METHODS: This study evaluated the clinical significance of the combined expression of CDX2 and its targets GPA33 and LI-cadherin in gastric cancer by fluorescence-based multiplex immunohistochemistry together with digital image analysis and chromogenic immunohistochemistry in 329 gastric cancer samples arranged in tissue microarrays. Additionally, publicly available RNA-seq expression data from 354 gastric cancer samples from the TCGA database were used to validate the immunohistochemistry results. RESULTS: Expression of the three markers (CDX2, GPA33, and LI-cadherin) was strongly correlated, defining an intestinal differentiation panel. Low or negative protein expression of the intestinal differentiation panel identified patients with particularly poor overall survival, irrespective of the methodology used, and was validated in the independent series at the RNA-seq level. CONCLUSIONS: Expression of the intestinal differentiation panel (CDX2, GPA33, and LI-cadherin) defines a set of biomarkers with a strong biological rationale and favourable impact for prognostication of gastric cancer patients.


Subject(s)
Antigens, Differentiation/metabolism , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor/metabolism , Cadherins/metabolism , Intestines/cytology , Membrane Glycoproteins/metabolism , Stomach Neoplasms/pathology , Aged , Cell Differentiation , Female , Follow-Up Studies , Humans , Male , Prognosis , Retrospective Studies , Stomach Neoplasms/metabolism , Stomach Neoplasms/surgery , Survival Rate
6.
Lab Invest ; 100(1): 120-134, 2020 01.
Article in English | MEDLINE | ID: mdl-31641225

ABSTRACT

Flourescence-based multiplex immunohistochemistry (mIHC) combined with multispectral imaging and digital image analysis (DIA) is a quantitative high-resolution method for determination of protein expression in tissue. We applied this method for five biomarkers (CDX2, SOX2, SOX9, E-cadherin, and ß-catenin) using tissue microarrays of a Norwegian unselected series of primary colorectal cancer. The data were compared with previously obtained chromogenic IHC data of the same tissue cores, visually assessed by the Allred method. We found comparable results between the methods, although confirmed that DIA offered improved resolution to differentiate cases with high and low protein expression. However, we experienced inherent challenges with digital image analysis of membrane staining, which was better assessed visually. DIA and mIHC enabled quantitative analysis of biomarker coexpression on the same tissue section at the single-cell level, revealing a strong negative correlation between the differentiation markers CDX2 and SOX2. Both methods confirmed known prognostic associations for CDX2, but DIA improved data visualization and detection of clinicopathological and biological associations. In summary, mIHC combined with DIA is an efficient and reliable method to evaluate protein expression in tissue, here shown to recapitulate and improve detection of known clinicopathological and survival associations for the emerging biomarker CDX2, and is therefore a candidate approach to standardize CDX2 detection in pathology laboratories.


Subject(s)
Biomarkers, Tumor/analysis , Fluorescent Antibody Technique , Image Interpretation, Computer-Assisted , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor/metabolism , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Humans , SOXB1 Transcription Factors/metabolism , Tissue Array Analysis
7.
Dis Markers ; 2019: 8267021, 2019.
Article in English | MEDLINE | ID: mdl-31275454

ABSTRACT

Gastric cancer is one of the most frequent tumours and the third leading cause of cancer-related death worldwide. The investigation of new biomarkers that can predict patient outcome more accurately and allow better treatment and follow-up decisions is of crucial importance. SOX9 (sex-determining region Y (SRY)-box 9) is a regulator of cell fate decisions in embryogenesis and adulthood. Here, we sought to ascertain the relevance of SOX9 transcription factor as a prognostic marker in gastric cancer. SOX9 expression was analyzed by immunohistochemistry in 333 gastric adenocarcinoma cases, and its association with clinicopathological and follow-up data was evaluated. SOX9 nuclear expression was absent in 17% of gastric cancer cases and predicted worse disease-free survival (P = 0.03). SOX9 expression was associated with lower risk of relapse in Cox univariable analysis (HR = 0.58; 95% CI = 0.35-0.97; P = 0.04). The prognostic value of SOX9 was more pronounced in tumours with expansive growth (P = 0.01) or with venous invasion (P = 0.02). Two validation cohorts from the Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG) confirmed that low SOX9 expression was significantly associated with poor patient outcome. In conclusion, we have identified SOX9 as a biomarker of disease relapse in gastric cancer patients. Further experiments are needed to elucidate its biological relevance at the cellular level.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , SOX9 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Female , Humans , Male , Middle Aged , SOX9 Transcription Factor/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
8.
Eur J Immunol ; 46(6): 1490-503, 2016 06.
Article in English | MEDLINE | ID: mdl-27005442

ABSTRACT

T lymphocytes stimulated through their antigen receptor (TCR) preferentially express mRNA isoforms with shorter 3´ untranslated regions (3´-UTRs) derived from alternative pre-mRNA cleavage and polyadenylation (APA). However, the physiological relevance of APA programs remains poorly understood. CD5 is a T-cell surface glycoprotein that negatively regulates TCR signaling from the onset of T-cell activation. CD5 plays a pivotal role in mediating outcomes of cell survival or apoptosis, and may prevent both autoimmunity and cancer. In human primary T lymphocytes and Jurkat cells we found three distinct mRNA isoforms encoding CD5, each derived from distinct poly(A) signals (PASs). Upon T-cell activation, there is an overall increase in CD5 mRNAs with a specific increase in the relative expression of the shorter isoforms. 3´-UTRs derived from these shorter isoforms confer higher reporter expression in activated T cells relative to the longer isoform. We further show that polypyrimidine tract binding protein (PTB/PTBP1) directly binds to the proximal PAS and PTB siRNA depletion causes a decrease in mRNA derived from this PAS, suggesting an effect on stability or poly(A) site selection to circumvent targeting of the longer CD5 mRNA isoform by miR-204. These mechanisms fine-tune CD5 expression levels and thus ultimately T-cell responses.


Subject(s)
CD5 Antigens/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , MicroRNAs/genetics , Polyadenylation , Polypyrimidine Tract-Binding Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , 3' Untranslated Regions , Base Sequence , CD5 Antigens/metabolism , Gene Expression Regulation , Humans , Jurkat Cells , Models, Biological , Poly A , RNA Interference , RNA Isoforms , RNA, Messenger/genetics
9.
BMC Cancer ; 14: 734, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25269858

ABSTRACT

BACKGROUND: Cancer stem cells are hypoxia-resistant and present a preponderant glycolytic metabolism. These characteristics are also found in basal-like breast carcinomas (BLBC), which show increased expression of cancer stem cell markers.Recently, we demonstrated that P-cadherin, a biomarker of BLBC and a poor prognostic factor in this disease, mediates stem-like properties and resistance to radiation therapy. Thus, the aim of the present study was to evaluate if P-cadherin expression was associated to breast cancer cell populations with an adapted phenotype to hypoxia. METHODS: Immunohistochemistry was performed to address the expression of P-cadherin, hypoxic, glycolytic and acid-resistance biomarkers in primary human breast carcinomas. In vitro studies were performed using basal-like breast cancer cell lines. qRT-PCR, FACS analysis, western blotting and confocal microscopy were used to assess the expression of P-cadherin after HIF-1α stabilization, achieved by CoCl2 treatment. siRNA-mediated knockdown was used to silence the expression of several targets and qRT-PCR was employed to evaluate the effects of P-cadherin on HIF-1α signaling. P-cadherin high and low breast cancer cell populations were sorted by FACS and levels of GLUT1 and CAIX were assessed by FACS and western blotting. Mammosphere forming efficiency was used to determine the stem cell activity after specific siRNA-mediated knockdown, further confirmed by western blotting. RESULTS: We demonstrated that P-cadherin overexpression was significantly associated with the expression of HIF-1α, GLUT1, CAIX, MCT1 and CD147 in human breast carcinomas. In vitro, we showed that HIF-1α stabilization was accompanied by increased membrane expression of P-cadherin and that P-cadherin silencing led to a decrease of the mRNA levels of GLUT1 and CAIX. We also found that the cell fractions harboring high levels of P-cadherin were the same exhibiting more GLUT1 and CAIX expression. Finally, we showed that P-cadherin silencing significantly decreases the mammosphere forming efficiency in the same range as the silencing of HIF-1α, CAIX or GLUT1, validating that all these markers are being expressed by the same breast cancer stem cell population. CONCLUSIONS: Our results establish a link between aberrant P-cadherin expression and hypoxic, glycolytic and acid-resistant breast cancer cells, suggesting a possible role for this marker in cancer cell metabolism.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Carcinoma, Ductal, Breast/metabolism , Glycolysis , Acid-Base Imbalance/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Female , Gene Expression , Glucose Transporter Type 1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phenotype , Spheroids, Cellular/metabolism , Tissue Array Analysis
10.
Breast Cancer Res ; 14(3): 211, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22676419

ABSTRACT

Breast cancer is a heterogeneous disease associated with diverse biological behaviours and clinical outcome. Although some molecular subgroups of breast cancer have a targeted therapy, the most aggressive tumours still lack a molecular target. Despite vitamin D being classically associated with the physiological role of calcium regulation and phosphate transport in bone metabolism, several studies have demonstrated a wide range of functions for this hormone, which are particularly important in the field of cancer. The mechanisms underlying the protective actions of vitamin D in cancer development are only sparsely understood, but evidence shows that vitamin D participates in cell growth regulation, apoptosis and cell differentiation. In addition, it has been implicated in the suppression of cancer cell invasion, angiogenesis and metastasis. Most of vitamin D biological actions are mediated by the vitamin D receptor and the synthesis and catabolism of this hormone are regulated by the enzymes CYP27B1 and CYP24A1. In the present review we highlight research data concerning the function of this hormone in the mammary gland, with a special focus on breast carcinogenesis. Hence, and although the available data are controversial, we consider not only updated information on the epidemiology of vitamin D in breast cancer and its potential value as a therapeutic agent or prophylactic (with an emphasis on molecular mechanisms and effectors of vitamin D action), but include data on its role in other stages of breast cancer progression as well. Accordingly, we review data on the influence of vitamin D in the development of normal breast and the expression of vitamin D-related proteins (VDR, CYP27B1 and CYP24A21) in benign mammary lesions and ductal carcinomas in situ.


Subject(s)
Breast Neoplasms , Cell Transformation, Neoplastic , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Receptors, Calcitriol/metabolism , Vitamin D/physiology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Apoptosis , Breast Neoplasms/epidemiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Cycle , Cell Differentiation , Cell Proliferation , Female , Humans , Mammary Glands, Human/pathology , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
11.
Anticancer Res ; 32(1): 249-57, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22213313

ABSTRACT

BACKGROUND: The triple-negative subgroup of breast cancer includes a cluster of tumors exhibiting low E-cadherin expression (metaplastic carcinomas). In several cancer models, 1 alpha,25-dihydroxyvitamin D(3) (1α,25(OH)(2)D(3)) induces differentiation by increasing E-cadherin expression. The Vitamin D receptor (VDR) was evaluated as a possible therapeutic target for metaplastic carcinomas and 1α,25(OH)(2)D(3) effects as a differentiating agent in triple-negative breast cancer cells were assessed. MATERIALS AND METHODS: Metaplastic carcinomas were assessed for VDR expression by immunohistochemistry; differences in E-cadherin expression in triple-negative breast cancer cells were evaluated by real-time PCR, western blotting and Cadherin 1 (CDH1) methylation status. RESULTS: Most of the metaplastic carcinomas were positive for VDR expression. Furthermore, 1α,25(OH)(2)D(3) promoted differentiation of MDA-MB-231 cells by inducing de novo E-cadherin expression, an effect that was time- and dose-dependent. Also, E-cadherin expression was due to promoter demethylation. CONCLUSION: Metaplastic carcinomas may respond to 1α,25(OH)(2)D(3), since they express VDR and 1α,25(OH)(2)D(3) induces de novo E-cadherin expression in breast cancer cells by promoter demethylation.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cadherins/genetics , DNA Methylation/drug effects , Receptors, Calcitriol/metabolism , Vitamin D/analogs & derivatives , Antigens, CD , Blotting, Western , Breast Neoplasms/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/metabolism , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tumor Cells, Cultured , Vitamin D/pharmacology
12.
Hum Pathol ; 42(10): 1438-46, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21450330

ABSTRACT

The current system of pathologic classification of human breast cancers does not take into account the biologic determinants of prognosis, nor is there a consensus regarding the progression from in situ to invasive carcinoma. The present study compared the molecular phenotypes of in situ and invasive components of breast cancer in the same sample. We built a series of 189 in situ and invasive carcinomas using tissue microarrays and classified them according to their immunoprofiles regarding estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, epidermal growth factor receptor, cytokeratin 5, P-cadherin, and the antigen Ki-67 into luminal A and B, human epidermal growth factor receptor 2 overexpressing, and basal-like carcinomas. We also correlated the subgroups of carcinomas with some of the classical prognostic factors such as histologic grade, tumor size, and lymph node metastasis, as well as with the age of the patient at diagnosis. The overall concordance on the molecular phenotypes between in situ and invasive components was 94%. For the in situ component, 63% of the cases were luminal A; 15%, luminal B; 12%, human epidermal growth factor receptor 2 overexpressing; and 7%, basal-like. Regarding the invasive component, 61% of the cases were luminal A; 16%, luminal B; 12%, human epidermal growth factor receptor 2 overexpressing; and 8%, basal-like. The present study allowed the identification of different immunoprofiles of in situ and invasive breast carcinomas using a specific panel of biomarkers and showed that in most cases, there is a concordance between in situ and invasive component profiles, supporting the theory of parallel disease in breast tumorigenesis.


Subject(s)
Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Biomarkers, Tumor/metabolism , Breast Neoplasms/classification , Breast Neoplasms/metabolism , Carcinoma in Situ/classification , Carcinoma in Situ/metabolism , Carcinoma, Ductal, Breast/classification , Carcinoma, Ductal, Breast/metabolism , Female , Humans , Immunohistochemistry , Neoplasm Invasiveness , Tissue Array Analysis
13.
BMC Cancer ; 10: 483, 2010 Sep 11.
Article in English | MEDLINE | ID: mdl-20831823

ABSTRACT

BACKGROUND: Breast cancer is a heterogeneous disease associated with different patient prognosis and responses to therapy. Vitamin D has been emerging as a potential treatment for cancer, as it has been demonstrated that it modulates proliferation, apoptosis, invasion and metastasis, among others. It acts mostly through the Vitamin D receptor (VDR) and the synthesis and degradation of this hormone are regulated by the enzymes CYP27B1 and CYP24A1, respectively. We aimed to study the expression of these three proteins by immunohistochemistry in a series of breast lesions. METHODS: We have used a cohort comprising normal breast, benign mammary lesions, carcinomas in situ and invasive carcinomas and assessed the expression of the VDR, CYP27B1 and CYP24A1 by immunohistochemistry. RESULTS: The results that we have obtained show that all proteins are expressed in the various breast tissues, although at different amounts. The VDR was frequently expressed in benign lesions (93.5%) and its levels of expression were diminished in invasive tumours (56.2%). Additionally, the VDR was strongly associated with the oestrogen receptor positivity in breast carcinomas. CYP27B1 expression is slightly lower in invasive carcinomas (44.6%) than in benign lesions (55.8%). In contrast, CYP24A1 expression was augmented in carcinomas (56.0% in in situ and 53.7% in invasive carcinomas) when compared with that in benign lesions (19.0%). CONCLUSIONS: From this study, we conclude that there is a deregulation of the Vitamin D signalling and metabolic pathways in breast cancer, favouring tumour progression. Thus, during mammary malignant transformation, tumour cells lose their ability to synthesize the active form of Vitamin D and respond to VDR-mediated Vitamin D effects, while increasing their ability to degrade this hormone.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Breast Neoplasms/metabolism , Breast/metabolism , Receptors, Calcitriol/metabolism , Steroid Hydroxylases/metabolism , Vitamin D/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast/pathology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Cohort Studies , Disease Progression , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Metabolic Networks and Pathways , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vitamin D3 24-Hydroxylase
14.
Histol Histopathol ; 25(8): 963-74, 2010 08.
Article in English | MEDLINE | ID: mdl-20552547

ABSTRACT

INTRODUCTION: The most suitable immunohistochemical criterion to identify basal-like breast carcinomas (BLBC), a molecular subgroup of breast cancer associated with poor prognosis, is the triple negative phenotype along with CK5 and/or EGFR immunoreactivity. However, several putative basal markers have been suggested as alternatives to identify BLBC with more accuracy. EXPERIMENTAL DESIGN: The expression of CK5, EGFR, P-cadherin, CK14, Vimentin and p63 were evaluated in 462 invasive breast carcinomas to determine their sensitivity and specificity for BLBC identification. RESULTS: P-cadherin and CK5 showed higher sensitivity values, while EGFR, Vimentin and CK14 were the most specific markers. The combination of CK5 with P-cadherin, Vimentin or CK14 proved to be a reliable option for distinguishing the basal phenotype, compared to the "gold standard" pair CK5/EGFR. Furthermore, P-cadherin was still able to recognize a large number of putative BLBC among the "unclassified" group (ER-/PR-/HER2-/CK5-/EGFR-). CONCLUSIONS: P-cadherin, Vimentin and CK14 can recognize BLBC already identified in triple negative/ CK5 and/or EGFR+ tumors, and due to P-cadherin sensitivity for BLBC identification this marker can reliably recruit a large number of breast carcinomas with basal phenotype among immunohistochemistry triple negative/ CK5 and/or EGFR - pool of tumors. Although they need GEP validation, our results can introduce the idea of these markers as additional options in the daily workup of breast pathology laboratories to identify BLBC.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma/genetics , Female , Humans , Immunohistochemistry , Phenotype , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Vimentin/genetics
15.
Histopathology ; 55(5): 594-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19912365

ABSTRACT

AIMS: To evaluate the relationship between microvessel density assessed by endoglin expression and the molecular subtypes of human invasive breast carcinomas and whether there is evidence to indicate that angiogenesis could be a putative target for therapy in specific subsets of breast cancer. METHODS AND RESULTS: We studied a series of 161 breast carcinomas, but information was available on only 142 tumours. We correlated endoglin expression with distinct breast carcinoma subgroups classified according to immunohistochemical profiling. Additionally, we compared it with other biomarkers for the aggressive basal-like subset and with available histopathological data. Although the basal-like subtype has higher microvessel density, there are no significant differences with the other molecular subtypes of breast cancer. CONCLUSIONS: This study found no significant differences in tumour vascularity in different molecular subtypes of breast cancer.


Subject(s)
Antigens, CD/biosynthesis , Biomarkers, Tumor/analysis , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Neovascularization, Pathologic/pathology , Receptors, Cell Surface/biosynthesis , Antigens, CD/genetics , Breast Neoplasms/pathology , Endoglin , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Receptors, Cell Surface/genetics
16.
Breast Cancer Res ; 11(3): R40, 2009.
Article in English | MEDLINE | ID: mdl-19549328

ABSTRACT

INTRODUCTION: The expression of additional genes, other than oestrogen receptor (ER), may be important to the hormone-responsive phenotype of breast cancer. Microarray analyses have revealed that forkhead box A1 (FOXA1) and GATA binding protein 3 (GATA-3) are expressed in close association with ERalpha, both encoding for transcription factors with a potential involvement in the ERalpha-mediated action in breast cancer. The purpose of this study was to explore if the expression of FOXA1 and GATA-3 may provide an opportunity to stratify subsets of patients that could have better outcome, among the ERalpha-negative/poor prognosis breast cancer group. METHODS: We evaluate FOXA1 and GATA-3 expression in 249 breast carcinomas by immunohistochemistry, associating it with breast cancer molecular markers, clinicopathological features and patient's survival. The clinicopathological features and immunohistochemical markers of the tumours were compared using the chi-square test and ANOVA. Disease-free survival was analysed through Kaplan-Meier survival curves and Cox regression. RESULTS: FOXA1 expression was demonstrated in 42% of invasive carcinomas, while GATA-3 was detected in 48% of the cases. FOXA1 expression was inversely associated with tumour size, Nottingham Prognostic Index, histological grade, lymph vascular invasion, lymph node stage and human epidermal growth factor receptor-2 (HER-2) overexpression, while GATA-3 expression showed inverse association with histological grade and HER-2. Both FOXA1 and GATA-3 were directly associated with ERalpha and progesterone receptor. Among FOXA1-positive tumours, 83.1% are comprised in the luminal A subtype, similar to GATA-3 where 87.7% of positive tumours were classified within this molecular subtype. In the subset of ERalpha-negative patients, those who were FOXA1-negative had a 3.61-fold increased risk of breast cancer recurrence when compared with the FOXA1-positive. CONCLUSIONS: FOXA1 was a significant predictor of good outcome in breast cancer, whereas GATA-3 was an important luminal marker. The expression of FOXA1 may be used for risk stratification among ERalpha-negative patients.


Subject(s)
GATA3 Transcription Factor/biosynthesis , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/biosynthesis , Receptors, Estrogen/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Immunohistochemistry/methods , Middle Aged , Prognosis , Proportional Hazards Models , Treatment Outcome
17.
Virchows Arch ; 450(1): 73-80, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17123107

ABSTRACT

Gene expression profiles of invasive breast carcinomas have identified a subgroup of tumours with worse prognosis, which have been called "basal-like". These are characterized by a specific pattern of expression, being estrogen receptor (ER) and HER2 negative, and frequently expressing at least one basal marker such as basal cytokeratins or epidermal growth factor receptor (EGFR). Previously, our group characterized basal-like tumours in a series of invasive breast carcinomas using P-cadherin (P-CD), p63 and cytokeratin 5 (CK5). Based on that study, we hypothesized that those high-grade basal-like invasive carcinomas might have a pre-invasive counterpart, which could be identified using the same approach. A series of 79 ductal carcinomas in situ (DCIS) were classified into distinct subgroups according to their ER, HER2 and basal markers expression. Luminal DCIS expressed ER and constituted 64.6% of the series; the HER2 overexpressing tumours did not express ER and represented 25.3% of the cases, whereas 10.1% lack the expression of ER and HER2 and expressed at least one basal marker (P-CD, CK5, CK14, p63, vimentin and/or EGFR). These basal-like DCIS were mostly high-grade, with comedo-type necrosis, and consistently showed expression of P-CD and CK5. In conclusion, DCIS with a basal-like phenotype represent a small percentage in our series, being P-CD and CK5, the most useful adjunct markers to distinguish this subset of carcinomas in situ of the breast.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Cadherins/analysis , Carcinoma in Situ/chemistry , Carcinoma, Ductal, Breast/chemistry , Keratin-5/analysis , Breast Neoplasms/diagnosis , Carcinoma in Situ/diagnosis , Carcinoma, Ductal, Breast/diagnosis , Female , Humans , Immunohistochemistry , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...