Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
1.
Am J Physiol Cell Physiol ; 321(4): C654-C670, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34348486

ABSTRACT

Although recurrent cancers can become life threatening, little is known about the intracellular events required for cancer recurrences. Due to this lack of mechanistic information, there is no test to predict cancer recurrences or nonrecurrences during early stages of disease. In this retrospective study, we use ductal carcinoma in situ of the breast as a framework to better understand the mechanism of cancer recurrences using patient outcomes as the physiological observable. Conventional pathology slides were labeled with anti-phosphofructokinase type L (PFKL) and anti-phosphofructokinase/fructose-2,6-bisphosphatase type 4 (PFKFB4) reagents. PFKL and PFKFB4 were found in ductal epithelial cell nucleoli from DCIS samples of women who did not experience a cancer recurrence. In contrast, PFKL and PFKFB4 may be found near the plasma membrane in samples from patients who will develop recurrent cancer. With the use of machine learning to predict patient outcomes, holdout studies of individual patient micrographs for the three biomarkers PFKL, PFKFB4, and phosphorylated glucose transporter 1 (GLUT1) demonstrated 38.6% true negatives, 49.5% true positives, 11.9% false positives, and 0% false negatives (n = 101). A subpopulation of recurrent samples demonstrated PFKL, PFKFB4, and phosphorylated GLUT1 accumulation at the apical surface of epithelial cells, suggesting that carbohydrates can be harvested from the ducts' luminal spaces as an energy source. We suggest that PFK isotype patterns are metabolic switches representing key mechanistic steps of recurrences. Furthermore, PFK enzyme patterns within epithelial cells contribute to an accurate diagnostic test to classify DCIS patients as high or low recurrence risk.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Epithelial Cells/enzymology , Phosphofructokinase-1, Liver Type/metabolism , Phosphofructokinase-2/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/secondary , Carcinoma, Intraductal, Noninfiltrating/therapy , Energy Metabolism , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Glucose Transporter Type 1/metabolism , Humans , Image Interpretation, Computer-Assisted , Machine Learning , Microscopy, Fluorescence , Middle Aged , Neoplasm Recurrence, Local , Pattern Recognition, Automated , Phosphorylation , Predictive Value of Tests , Protein Transport , Risk Factors , Treatment Outcome
2.
Virchows Arch ; 478(5): 933-941, 2021 May.
Article in English | MEDLINE | ID: mdl-33237469

ABSTRACT

Salivary gland carcinomas represent a heterogeneous group of poorly characterized head and neck tumors. The purpose of this study was to evaluate ALK gene and protein aberrations in a large, well-characterized cohort of these tumors. A total of 182 salivary gland carcinomas were tested for anaplastic lymphoma kinase (ALK) positivity by immunohistochemistry (IHC) using the cut-off of 10% positive cells. ALK positive tumors were subjected to FISH analysis and followed by hybrid capture-based next generation sequencing (NGS). Of the 182 tumors, 8 were ALK positive by IHC. Further analysis using hybrid capture NGS analysis revealed a novel MYO18A (Exon1-40)-ALK (exon 20-29) gene fusion in one case of intraductal carcinoma. Additional genomic analyses resulted in the detection of inactivating mutations in BRAF and TP53, as well as amplifications of ERBB2 and ALK. ALK rearrangements are a rare entity in salivary gland carcinomas. We identified a potentially targetable novel ALK fusion in an intraductal carcinoma of minor salivary glands.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Biomarkers, Tumor/genetics , Carcinoma/genetics , Salivary Gland Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma/enzymology , Carcinoma/pathology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Child , Female , Gene Amplification , Gene Fusion , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Salivary Gland Neoplasms/enzymology , Salivary Gland Neoplasms/pathology , Young Adult
4.
J Clin Pathol ; 73(2): 76-82, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31444238

ABSTRACT

AIMS: Cathepsin V (CTSV/CTSL2) is a lysosomal cysteine proteinase and plays a role in extracellular matrix degradation. It is associated with poor prognosis in invasive breast cancer (IBC), but its role in breast ductal carcinoma in situ (DCIS) remains unclear. In this study, we aimed to evaluate the prognostic significance of CTSV in DCIS. METHODS: CTSV protein expression was immunohistochemically assessed in a well-characterised and annotated cohort of DCIS comprising pure DCIS (n=776) and DCIS coexisting with IBC (n=239). CTSV expression was analysed in tumour cells and surrounding stroma, including its association with clinicopathological parameters and outcome. RESULTS: In pure DCIS, high CTSV expression was observed in 29% of epithelial tumour cells and 20% of surrounding stroma. High expression in both components was associated with features of poor prognosis including higher nuclear grade, hormone receptor negativity and HER2 positivity. In addition, stromal CTSV expression was associated with larger DCIS size, comedo-type necrosis and high proliferation index. DCIS associated with IBC showed higher CTSV expression than pure DCIS either within the epithelial tumour cells or surrounding stroma (p<0.0001 and p=0.001, respectively). In DCIS/IBC, CTSV expression was higher in the invasive component than DCIS component either in tumour cells or surrounding stroma (both p<0.0001). CTSV stromal expression was associated with invasive recurrence independent of other prognostic factors in patients treated with breast conserving surgery (HR=3.0, p=0.005). CONCLUSION: High expression of CTSV is associated with poor outcome in DCIS and is a potential marker to predict DCIS progression to invasive disease.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Cathepsins/analysis , Cysteine Endopeptidases/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/pathology , Cathepsins/genetics , Cysteine Endopeptidases/genetics , Disease Progression , Female , Humans , Immunohistochemistry , Middle Aged , Prognosis , Up-Regulation
5.
Pathol Oncol Res ; 25(2): 759-768, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30689164

ABSTRACT

Mammary carcinoma is the most common malignant tumor in women, and it is the leading cause of mortality. In tumor context, glycosylation promotes post translational modifications necessary for cell progression, emerging as a relevant tumor hallmarker. This study aimed to analyze the association between polypeptide N-acetylgalactosaminyltransferase-6 (ppGalNAc-T6), -T8, N-acetylglucosaminyltransferase III (GnT-III) expression, Phaseolus vulgaris-leucoagglutinin (PHA-L), wheat germ agglutinin (WGA) and peanut agglutinin (PNA) staining with clinic-histopathological factors from patients with pure ductal carcinoma in situ (DCIS) and DCIS with invasive ductal carcinoma (DCIS-IDC) of breast. Formalin-fixed and paraffin-embedded samples (n = 109) were analyzed. In pure DCIS samples GnT-III was over-expressed in comedo lesions (p = 0.007). In DCIS-IDC, GnT-III expression was associated with high nuclear grade tumors (p = 0.039) while the presence of PHA-L and WGA were inversely related to HER-2 expression (p = 0.001; p = 0.036, respectively). These findings pointed to possible involvement of GnT-III, ppGalNAc-T8, L-PHA and WGA as probes in prognostic evaluation of DCIS.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , N-Acetylglucosaminyltransferases/metabolism , Adult , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Female , Humans , Middle Aged , N-Acetylglucosaminyltransferases/analysis
6.
Mod Pathol ; 32(5): 639-649, 2019 05.
Article in English | MEDLINE | ID: mdl-30429518

ABSTRACT

Legumain is a proteolytic enzyme that plays a role in the regulation of cell proliferation in invasive breast cancer. Studies evaluating its role in ductal carcinoma in situ (DCIS) are lacking. Here, we aimed to characterize legumain protein expression in DCIS and evaluate its prognostic significance. Legumain was assessed immunohistochemically in a tissue microarray of a well-characterized cohort of DCIS (n = 776 pure DCIS and n = 239 DCIS associated with invasive breast cancer (DCIS-mixed)). Legumain immunoreactivity was scored in tumor cells and surrounding stroma and related to clinicopathological parameters and patient outcome. High legumain expression was observed in 23% of pure DCIS and was associated with features of high-risk DCIS including higher nuclear grade, comedo necrosis, hormone receptor negativity, HER2 positivity, and higher proliferation index. Legumain expression was higher in DCIS associated with invasive breast cancer than in pure DCIS (p < 0.0001). In the DCIS-mixed cohort, the invasive component showed higher legumain expression than the DCIS component (p < 0.0001). Legumain was an independent predictor of shorter local recurrencefree interval for all recurrences (p = 0.0003) and for invasive recurrences (p = 0.002). When incorporated with other risk factors, legumain provided better patient risk stratification. High legumain expression is associated with poor prognosis in DCIS and could be a potential marker to predict DCIS progression to invasive disease.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Cysteine Endopeptidases/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/therapy , Cysteine Endopeptidases/genetics , Disease Progression , Female , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Progression-Free Survival , Risk Assessment , Risk Factors , Time Factors , Up-Regulation
7.
Br J Cancer ; 119(12): 1518-1526, 2018 12.
Article in English | MEDLINE | ID: mdl-30410060

ABSTRACT

BACKGROUND: Extracellular matrix (ECM) plays a crucial role in tumour behaviour. Prolyl-4-hydroxlase-A2 (P4HA2) is a key enzyme in ECM remodelling. This study aims to evaluate the prognostic significance of P4HA2 in breast ductal carcinoma in situ (DCIS). METHODS: P4HA2 expression was assessed immunohistochemically in malignant cells and surrounding stroma of a large DCIS cohort comprising 481 pure DCIS and 196 mixed DCIS and invasive carcinomas. Outcome analysis was evaluated using local recurrence free interval (LRFI). RESULTS: High P4HA2 expression was detected in malignant cells of half of pure DCIS whereas its expression in stroma was seen in 25% of cases. Higher P4HA2 expression was observed in mixed DCIS cases compared to pure DCIS both in tumour cells and in stroma. High P4HA2 was associated with features of high risk DCIS including younger age, higher grade, comedo necrosis, triple negative and HER2-positive phenotypes. Interaction between P4HA2 and radiotherapy was also observed regarding the outcome. High P4HA2 expression was an independent prognostic factor in predicting shorter LRFI. CONCLUSION: P4HA2 plays a role in DCIS progression and can potentially be used to predict DCIS outcome. Incorporation of P4HA2 with other clinicopathological parameters could refine DCIS risk stratification that can potentially guide management decisions.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Prolyl Hydroxylases/physiology , Adult , Aged , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Recurrence, Local , Prognosis , Prolyl Hydroxylases/analysis
8.
Zhonghua Zhong Liu Za Zhi ; 38(10): 743-749, 2016 Oct 23.
Article in Chinese | MEDLINE | ID: mdl-27784457

ABSTRACT

Objective: To study the clinical significance of PI3Kp110α expression in breast cancer. Methods: The expressions of PI3Kp110α, HER2, PTEN, p-Akt and Ki-67 in invasive ductal carcinoma (IDC), adjacent ductal carcinoma in situ (DCIS) and normal breast tissue were detected by immunohistochemistry in 102 cases of breast cancer. The expression of PI3Kp110α in IDC was compared with those in DCIS and normal breast tissues. Correlations between expression of PI3Kp110α and expression of HER2, PTEN, p-Akt and Ki-67 index in IDC were analyzed. Correlation between expression of PI3Kp110α and stage of IDC was studied as well. Results: In the normal breast tissues, there were 97 cases (95.1%) with low level and 5 cases (4.9%) with high level expression of PI3Kp110α. In the DCIS tissues, there were 67 cases (65.7%) with low level and 35 cases (34.3%) with high level expression of PI3Kp110α. In the IDC tissues, there were 14 cases (13.7%) with low level and 88 cases (86.3%) with high level expression of PI3Kp110α. The difference between expression of PI3Kp110α in normal breast tissue, DCIS and IDC was significant (P<0.001). In the IDC tissues, expression of PI3Kp110α was negatively correlated with expression of HER2 (rs=-0.213, P=0.032) and PTEN (rs=-0.197, P=0.047), while was not significantly correlated with expression of p-Akt (P=0.119) and Ki-67 index (P=0.636). In contrast, expressions of HER-2 and p-Akt were positively correlated with Ki-67 index (P=0.001, P=0.035), while expression of HER2 was not correlated with p-Akt (P=0.177). Expression of PI3Kp110α was negatively correlated with T stage and TNM stage (P=0.003, P=0.016), while not correlated with N stage and M stage(both P>0.05). Expression of HER-2 was positively correlated with T stage (P=0.037), while not correlated with N stage, M stage and TNM stage (P>0.05 for all). Neither Ki-67 index nor expression of PTEN and p-Akt (P=0.194) were correlated with T stage, N stage, M stage and TNM stage. Conclusions: Expression of PI3Kp110α plays an important role in oncogenesis and development of breast cancer. Based on the fact that expression of PI3Kp110α is negatively correlated with expression of HER2 and PTEN and with T stage and TNM stage, we may conclude that increased expression of PI3Kp110α is another independent pathway in breast cancer development, and breast cancer patients with high expression of PI3Kp110α may have a better prognosis.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Breast/enzymology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Immunohistochemistry , PTEN Phosphohydrolase/metabolism , Prognosis , Receptor, ErbB-2/metabolism
9.
Tumour Biol ; 37(4): 4743-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26515337

ABSTRACT

Absence of therapeutic targets poses a critical hurdle in improving prognosis for patients with triple negative breast cancer (TNBC). We evaluated interaction between SIRT1 and epithelial mesenchymal transition (EMT)-associated proteins as well as the role of combined protein expression as a predictor of lymph node metastasis and clinical outcome in TNBC through in vivo and vitro studies. Three hundred nineteen patients diagnosed with TNBC were chosen, immunohistochemical staining for SIRT1 and EMT-related markers' expression was performed on tissue microarrays, and in vitro experiments with each of the three human TNBC cell lines were carried out. The cohort was reclassified according to the use of adjuvant chemotherapy, tumor size, and AJCC stage to analyze the prognostic role of SIRT1 and EMT-related proteins' expression considering different therapeutic modalities and AJCC stages. Combination of four proteins including SIRT1 and three EMT-related proteins was revealed to be a statistically significant independent predictor of lymph node metastasis in the tumor size cohort as well as in the total patient population. Upon Cox regression analysis, increased expression level of the combined proteins correlated with decreased disease-free survival in the total patients as well as those who received adjuvant chemotherapy and those who had early stage breast cancer. In additional in vitro experiments, inhibition of SIRT1 expression with small interfering RNA (siRNA) suppressed tumor invasion in three different TNBC cell lines, and altered expression levels of EMT-related proteins following SIRT1 gene inhibition were identified on western blotting and fluorescence activated cell sorting (FACS) analysis; on the other hand, no change in expression levels of the cell cycle-related factors was observed. Our analysis showed the potential role of SIRT1 in association with EMT-related factors on tumor invasion, metastasis, and disease-free survival in TNBC, SIRT1, and associated EMT-related markers may offer a new prognostic indicator as well as a novel therapeutic candidate.


Subject(s)
Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Epithelial-Mesenchymal Transition , Sirtuin 1/physiology , Triple Negative Breast Neoplasms/enzymology , Antigens, CD , Cadherins/metabolism , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Carcinoma, Ductal, Breast/therapy , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/secondary , Carcinoma, Intraductal, Noninfiltrating/therapy , Cell Line, Tumor , Cell Proliferation , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Gene Expression , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Multivariate Analysis , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , Snail Family Transcription Factors/metabolism , Treatment Outcome , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Vimentin/metabolism
10.
Diagn Pathol ; 10: 66, 2015 Jun 13.
Article in English | MEDLINE | ID: mdl-26070788

ABSTRACT

BACKGROUND: Aldehyde dehydrogenase 1 (ALDH1) is an important marker of tumor-initiating cells. We aimed to investigate ALDH1 expression in benign breast disease and human breast cancer of different histologic stages. METHODS: Immunohistochemical staining of ALDH1 was applied to 21 cases with benign breast diseases, 47 ductal carcinoma in situ (DCIS) cases, 62 cases diagnosed with invasive cancer with extensive intraductal component (EIC), and 58 cases diagnosed with invasive cancer without EIC. RESULTS: ALDH1 was expressed in tumor cells in 61.0% of 164 breast cancer cases, which was higher than that in benign breast disease (3/21) (P < 0.001). Of these 167 breast cancer cases, a significantly higher rate (54/58) of intratumoral ALDH1 expression was observed in invasive cancer without EIC cases than that in DCIS cases (19/46, one case not available) and invasive cancer with EIC cases (27/60, two cases not available) (P < 0.001). Interestingly, a significantly higher rate (22/48) of intratumoral ALDH1 expression in invasive component was observed than that in in situ component (7/48) in the same tumor (P = 0.001). In 47 DCIS cases, no significant association was observed between ALDH1 positivity and any clinicopathological parameter (all P > 0.05). However, ALDH1 positive invasive breast cancers were significantly more likely to be with large tumor size (P = 0.001), high grade (P < 0.001), and high Ki67 expression (P = 0.009). CONCLUSIONS: ALDH1 may play an important role in the invasion of breast cancer, and may be associated with aggressive phenotypes of breast cancer. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1608671725154947 .


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Isoenzymes/analysis , Retinal Dehydrogenase/analysis , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Phenotype , Predictive Value of Tests , Tumor Burden , Young Adult
11.
Mol Oncol ; 9(1): 58-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25131496

ABSTRACT

Acid ceramidase (ASAH1) a key enzyme of sphingolipid metabolism converting pro-apoptotic ceramide to sphingosine has been shown to be overexpressed in various cancers. We previously demonstrated higher expression of ASAH1 in ER positive compared to ER negative breast cancer. In the current study we performed subtype specific analyses of ASAH1 gene expression in invasive and non invasive breast cancer. We show that expression of ASAH1 is mainly associated with luminal A - like cancers which are known to have the best prognosis of all breast cancer subtypes. Moreover tumors with high ASAH1 expression among the other subtypes are also characterized by an improved prognosis. The good prognosis of tumors with high ASAH1 is independent of the type of adjuvant treatment in breast cancer and is also detected in non small cell lung cancer patients. Moreover, even in pre-invasive DCIS of the breast ASAH1 is associated with a luminal phenotype and a reduced frequency of recurrences. Thus, high ASAH1 expression is generally associated with an improved prognosis in invasive breast cancer independent of adjuvant treatment and could also be valuable as prognostic factor for pre-invasive DCIS.


Subject(s)
Acid Ceramidase/biosynthesis , Breast Neoplasms/enzymology , Breast Neoplasms/mortality , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/mortality , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Breast Neoplasms/drug therapy , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Invasiveness , Survival Rate
12.
Int J Clin Exp Pathol ; 7(5): 2337-49, 2014.
Article in English | MEDLINE | ID: mdl-24966944

ABSTRACT

Increased sialylation and ß1,6-branched oligosaccharides has been associated with a variety of structural changes in cell surface carbohydrates, most notably in tumorigenesis. Lectins are defined as proteins that preferentially recognize and bind carbohydrate complexes protruding from glycolipids and glycoproteins. This interaction with carbohydrates can be as specific as the interaction between antigen and antibody. Due to this type of interaction lectins have been used as experimental auxiliary tools in histopathological diagnosis of cancer. This study was designed to evaluate the differential expression of sialic acids and ß1,6-N-acetylglucosaminyltransferase V (MGAT5) in invasive (IDC) and in situ (DCIS) ductal carcinoma of the breast and its possible application as prognostic biomarkers. A possible transition between pre-malign and malign lesions was evaluated using DCIS samples. Biopsies were analyzed regarding the expression of MUC1, p53, Ki-67, estrogen receptor, progesterone receptor, HER-2 and MGAT5. α2,6-linked sialic acids residues recognized by SNA lectin was overexpressed in 33.3% of IDC samples and it was related with Ki-67 (p=0.042), PR (p=0.029), lymphnodes status (p=0.017) and death (p=0.011). Regarding survival analysis SNA was the only lectin able to correlate with specific-disease survival and disease-free survival (p=0.024 and p=0.041, respectively), besides, it presents itself as an independent variable by Cox Regression analysis (p= 0.004). Comparing IDC and DCIS cases, only SNA showed different staining pattern (p=0.034). The presence of sialic acids on tumor cell surface can be an indicative of poor prognosis and our study provides further evidence that SNA lectin can be used as a prognostic probe in IDC and DCIS patients.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Immunohistochemistry , N-Acetylglucosaminyltransferases/analysis , Plant Lectins , Ribosome Inactivating Proteins , Adult , Aged , Aged, 80 and over , Biopsy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Chi-Square Distribution , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Predictive Value of Tests , Proportional Hazards Models , Risk Factors , Time Factors , Tissue Array Analysis
13.
Br J Cancer ; 111(1): 46-54, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24874483

ABSTRACT

BACKGROUND: Stratification of patients for treatment of ductal carcinoma in situ (DCIS) is suboptimal, with high systemic overtreatment rates. METHODS: A training set of 95 tumours from women with pure DCIS were immunostained for proteins involved in cell survival, hypoxia, growth factor and hormone signalling. A generalised linear regression with regularisation and variable selection was applied to a multiple covariate Cox survival analysis with recurrence-free survival 10-fold cross-validation and leave-one-out iterative approach were used to build and test the model that was validated using an independent cohort of 58 patients with pure DCIS. The clinical role of a COX-2-targeting agent was then tested in a proof-of-concept neoadjuvant randomised trial in ER-positive DCIS treated with exemestane 25 mg day(-1)± celecoxib 800 mg day(-1). RESULTS: The COX-2 expression was an independent prognostic factor for early relapse in the training (HR 37.47 (95% CI: 5.56-252.74) P=0.0001) and independent validation cohort (HR 3.9 (95% CI: 1.8-8.3) P=0.002). There was no significant interaction with other clinicopathological variables. A statistically significant reduction of Ki-67 expression after treatment with exemestane ± celecoxib was observed (P<0.02) with greater reduction in the combination arm (P<0.004). Concomitant reduction in COX-2 expression was statistically significant in the exemestane and celecoxib arm (P<0.03) only. CONCLUSIONS: In patients with DCIS, COX-2 may predict recurrence, aiding clinical decision making. A combination of an aromatase inhibitor and celecoxib has significant biological effect and may be integrated into treatment of COX2-positive DCIS at high risk of recurrence.


Subject(s)
Androstadienes/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/enzymology , Cyclooxygenase 2/biosynthesis , Androstadienes/administration & dosage , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Celecoxib , Cohort Studies , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Recurrence, Local/enzymology , Neoplasm Recurrence, Local/pathology , Prognosis , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Survival Analysis
14.
Ann Surg Oncol ; 21(9): 2911-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24777857

ABSTRACT

BACKGROUND: Patients with ductal carcinoma-in-situ (DCIS) are currently not prescribed adjuvant systemic treatment after surgery and radiotherapy. Prediction of DCIS patients who would benefit from radiotherapy is warranted. Statins have been suggested to exert radio-sensitizing effects. The target for cholesterol-lowering statins is HMG-CoA reductase (HMGCR), the rate-limiting enzyme in the mevalonate pathway. The aim of this study was to examine HMGCR expression in DCIS and study its treatment predictive value. METHODS: A population-based cohort including 458 women diagnosed with primary DCIS between 1986 and 2004 were followed until November 2011 to study long-term survival. Tumor tissue microarrays were constructed, and immunohistochemical analyses were performed to detect cytoplasmic protein expression of HMGCR. The association between DCIS HMGCR expression and invasive breast cancer recurrence-free survival (RFSinv) and overall survival (OS) was analyzed by Kaplan-Meier curves, log rank test, and Cox proportional hazard analysis. RESULTS: HMGCR was strongly expressed in 24 % of the assessed DCIS samples, moderately expressed in 46 %, and weakly expressed in 23 %; no expression was detected in 7 % of the samples. During the follow-up time (median 13.8 years), 61 patients were diagnosed with an invasive breast cancer recurrence, and 80 patients died. A crude analysis showed no survival benefit from radiotherapy. However, patients with strong HMGCR expression showed an improved RFSinv (log rank, p = 0.03) and OS (log rank, p = 0.04) after radiotherapy. No statistically significant interaction was observed for HMGCR and radiotherapy (RFSinv p = 0.69 and OS p = 0.29). CONCLUSIONS: This study demonstrates HMGCR expression in DCIS and suggests HMGCR as a predictive marker of response to postoperative radiotherapy in DCIS, although the test for interaction was nonsignificant. Future DCIS studies addressing the potential of statin treatment targeting HMGCR are warranted.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Hydroxymethylglutaryl CoA Reductases/chemistry , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mastectomy/mortality , Neoplasm Recurrence, Local/enzymology , Postoperative Complications/enzymology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/therapy , Cohort Studies , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Postoperative Complications/mortality , Postoperative Complications/pathology , Postoperative Complications/therapy , Prognosis , Radiotherapy Dosage , Survival Rate
15.
Am J Pathol ; 184(4): 1219-1229, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24518566

ABSTRACT

Cyclooxygenase-2 (COX-2) overexpression is implicated in increased risk and poorer outcomes in breast cancer in young women. We investigated COX-2 regulation in normal premenopausal breast tissue and its relationship to malignancy in young women. Quantitative COX-2 immunohistochemistry was performed on adjacent normal and breast cancer tissues from 96 premenopausal women with known clinical reproductive histories, and on rat mammary glands with distinct ovarian hormone exposures. COX-2 expression in the normal breast epithelium varied more than 40-fold between women and was associated with COX-2 expression levels in ductal carcinoma in situ and invasive cancer. Normal breast COX-2 expression was independent of known breast cancer prognostic indicators, including tumor stage and clinical subtype, indicating that factors regulating physiological COX-2 expression may be the primary drivers of COX-2 expression in breast cancer. Ovarian hormones, particularly at pregnancy levels, were identified as modulators of COX-2 in normal mammary epithelium. However, serial breast biopsy analysis in nonpregnant premenopausal women suggested relatively stable baseline levels of COX-2 expression, which persisted independent of menstrual cycling. These data provide impetus to investigate how baseline COX-2 expression is regulated in premenopausal breast tissue because COX-2 levels in normal breast epithelium may prove to be an indicator of breast cancer risk in young women, and predict the chemopreventive and therapeutic efficacy of COX-2 inhibitors in this population.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Cyclooxygenase 2/biosynthesis , Adult , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Humans , Immunoblotting , Immunohistochemistry , Middle Aged , Rats , Rats, Sprague-Dawley , Young Adult
16.
BMC Cancer ; 14: 57, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24485087

ABSTRACT

BACKGROUND: The HECT family ubiquitin ligase Smurf2 regulates cell polarity, migration, division, differentiation and death, by targeting diverse substrates that are critical for receptor signaling, cytoskeleton, chromatin remodeling and transcription. Recent studies suggest that Smurf2 functions as a tumor suppressor in mice. However, no inactivating mutation of SMURF2 has been reported in human, and information about Smurf2 expression in human cancer remains limited or complicated. Here we demonstrate that Smurf2 expression is downregulated in human breast cancer tissues, especially of the triple-negative subtype, and address the mechanism of Smurf2 downregulation in triple-negative breast cancer cells. METHODS: Human breast cancer tissues (47 samples expressing estrogen receptor (ER) and 43 samples with triple-negative status) were examined by immunohistochemistry for the expression of Smurf2. Ten widely-studied human breast cancer cell lines were examined for the expression of Smurf2. Furthermore, microRNA-mediated regulation of Smurf2 was investigated in triple-negative cancer cell lines. RESULTS: Immunohistochemical analysis showed that benign mammary epithelial cells expressed high levels of Smurf2, so did cells in ductal carcinomas in situ. In contrast, invasive ductal carcinomas showed focal or diffuse decrease in Smurf2 expression, which was observed more frequently in triple-negative tumors than in ER-positive tumors. Consistently, human triple-negative breast cancer cell lines such as BT549, MDA-MB-436, DU-4475 and MDA-MB-468 cells showed significantly lower expression of Smurf2 protein, compared to ER + or HER2+ cell lines. Studies using quantitative PCR and specific microRNA inhibitors indicated that increased expression of miR-15a, miR-15b, miR-16 and miR-128 was involved in Smurf2 downregulation in those triple-negative cancer cell lines, which have mutations in the retinoblastoma (RB) gene. Forced expression of RB increased levels of Smurf2 protein with concomitant decreases in the expression of the microRNAs. CONCLUSIONS: This study provides evidence of posttranscriptional downregulation of Smurf2 in triple-negative breast cancers, and demonstrates that the loss of RB function is involved in microRNA-mediated interference with Smurf2 translation. The new link from RB inactivation to Smurf2 downregulation is likely to play a role in malignant phenotypes of triple-negative breast cancer cells.


Subject(s)
Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , MicroRNAs/metabolism , Retinoblastoma Protein/metabolism , Triple Negative Breast Neoplasms/enzymology , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Middle Aged , Mutation , Phenotype , RNA Interference , Retinoblastoma Protein/genetics , Signal Transduction , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics
17.
Diagn Pathol ; 9: 22, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24456584

ABSTRACT

BACKGROUND AND OBJECTIVES: Breast cancer is one of the most common causes of cancer-related deaths in women worldwide. Studies on glucosylceramide synthase (GCS) activity suggest that this enzyme has a role in the development of multidrug resistance in many cancer cells. However, few studies have shown the expression of GCS in invasive ductal breast cancer and breast intraductal proliferative lesions. METHODS: In total, 196 samples from patients with invasive ductal breast cancer and 61 samples of breast intraductal proliferative lesions were collected. Immunohistochemical analyses were conducted to determine the expression of GCS and other related proteins. RESULTS: Expression of GCS was high in estrogen receptor (ER)-positive and HER-2 negative samples. In contrast, the expression of GCS in invasive ductal cancer was significantly lower than that in intraductal proliferative lesions. CONCLUSION: Our data demonstrates a correlation between the expression of the GCS protein and ER-positive/HER-2 negative breast cancer. Furthermore, in contrast to previous reports, the expression of GCS protein was shown to be much higher in ductal carcinoma in-situ than that in invasive ductal cancer. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1559854430111589.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Ductal, Breast/enzymology , Glucosyltransferases/biosynthesis , Adult , Aged , Biomarkers, Tumor/analysis , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/mortality , Carcinoma, Intraductal, Noninfiltrating/enzymology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/mortality , Female , Glucosyltransferases/analysis , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Middle Aged , Prognosis , Receptor, ErbB-2/analysis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptors, Estrogen/analysis , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
18.
Asian Pac J Cancer Prev ; 15(24): 10773-7, 2014.
Article in English | MEDLINE | ID: mdl-25605174

ABSTRACT

BACKGROUND: Treatment for breast cancer is mainly performed by surgical resection of primary tumors and chemotherapy. However, after tumor invasion and metastases, breast cancer is hard to control. Clarification of the pathogenic mechanisms would be helpful to the prognosis or therapy for the breast cancer. The aim of this study is to investigate the clinical and prognostic implications of legumain protein Materials and Methods: In this study, we examined mastectomy specimens from 114 breast cancer and matching, 26 adjacent non-cancerous tissues using immunohistochemistry. RESULTS: The results indicated that positive expression of legumain protein in breast cancer was 51.8 % (59/114) and the positive expression of legumain protein in adjacent non-cancerous tissue was 11.5% (3/26). It appeared to be related with lymph node metastasis of breast cancer (p=0.02) and correlation analysis indicated that legumain expression was correlated positively with the estrogen receptor (ER) and mutant-type p53 expression (both p<0.05). Positive legumain expression was significantly associated with shorter overall survival time in breast cancer patients (log-rank p<0.01). Multivariate survival analysis suggested that the positive legumain expression was an independent predictor of poorer overall survival in patients with breast cancer (HR=0.24; 95%CI 0.11-0.65, p=0.03). CONCLUSIONS: Legumain might be a new potential biomarker for breast cancer, which may reflect the prognosis and overall survival.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Cysteine Endopeptidases/metabolism , Adult , Aged , Asia , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/secondary , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Rate
19.
Oncology ; 85(4): 248-56, 2013.
Article in English | MEDLINE | ID: mdl-24192633

ABSTRACT

OBJECTIVE: The aim of this study was to elucidate the clinicopathological characteristics of breast ductal carcinomas in situ (DCIS) with aldehyde dehydrogenase 1 (ALDH1)-positive cancer stem cells (CSCs). METHODS: DCIS (n = 194) were subjected to immunohistochemical staining and results were examined for associations with various clinicopathological parameters. The ALDEFLUOR assay for breast cancer cell lines was also performed to determine the proportion of CSCs according to intrinsic subtype. RESULTS: DCIS with ALDH1-positive CSCs were significantly (p < 0.05) more likely to be estrogen receptor-α (ER)-negative, progesterone receptor (PgR)-negative, Ki67-positive and HER2-positive tumors. Luminal A subtype (8.6%) showed a significantly (p < 0.001) lower ALDH1 positivity than the other subtypes [luminal B (50.0%), luminal HER2 (36.8%), HER2 (35.3%) and triple-negative subtype (26.7%)]. Double immunostaining revealed that ALDH1-positive CSCs did not overlap with ER-, PgR- or Ki67-positive tumor cells but did overlap with HER2-positive tumor cells. The percentage of ALDEFLUOR-positive CSCs was lower in the luminal A cell lines (0.02% for T-47D and 0.4% for MCF7) than in the luminal HER2 (9.1% for BT-474 and 9.5% for MDA-MB-361), HER2 (13.9% for AU565 and 33.2% for SK-BR-3) and triple-negative cell lines (28.4% for MDA-MB-231 and 30.7% for MDA-MB-468). CONCLUSIONS: Our results suggest that most CSCs in DCIS are in the G0 phase (Ki67 negative) and do not express ER or PgR, but they do express HER2 in HER2-positive tumors.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Isoenzymes/metabolism , Neoplastic Stem Cells/enzymology , Retinal Dehydrogenase/metabolism , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Middle Aged , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Young Adult
20.
Cell Cycle ; 12(13): 2042-50, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23759596

ABSTRACT

The diagnosis of ductal carcinoma in situ (DCIS) is an increasingly common event due to widespread use of screening mammography. However, appropriate clinical management of DCIS is a major challenge in the absence of prognostic markers. Tumor-initiating cells may be particularly relevant for disease pathogenesis; therefore, two markers associated with such cells, EZH2 and ALDH1, were evaluated. A cohort of 248 DCIS patients was used to determine the association of EZH2 and ALDH1 with ipsilateral breast event, DCIS recurrence and progression to invasive breast cancer (IBC). In this cohort, high EZH2 expression was associated with the risk of an ipsilateral breast event and DCIS recurrence but not invasive progression. ALDH1 expression was observed in both the tumor and stromal compartment; however, in neither compartment were ALDH1 levels independently associated with evaluated study endpoints. Interestingly, the combination of high EZH2 with high epithelial ALDH1 was associated with disease progression. Therefore, ALDH1 within the DCIS lesion can add to the prognostic significance of EZH2, particularly in the context of risk of development of invasive disease.


Subject(s)
Breast Neoplasms/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Isoenzymes/metabolism , Neoplasm Recurrence, Local/enzymology , Polycomb Repressive Complex 2/metabolism , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cohort Studies , Disease Progression , Disease-Free Survival , Enhancer of Zeste Homolog 2 Protein , Female , Humans , Isoenzymes/genetics , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Polycomb Repressive Complex 2/genetics , Retinal Dehydrogenase/genetics , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...