Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Histopathology ; 66(2): 270-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25307864

ABSTRACT

AIMS: Epigenetic changes are of crucial importance in cancer development and are potentially reversible; they are therefore targets of interest for anti-cancer therapy. The aim of this study was to investigate the clinical prognostic value of the histone deacetylases SIRT1, HDAC1 and HDAC2 and the histone modifications H4K16Ac and H3K56Ac in colorectal cancer. METHODS AND RESULTS: The epigenetic markers were immunohistochemically stained on tissue microarrays containing colorectal tumours (n = 254) and normal colorectal tissues (n = 50). Nuclear expression was assessed on the semi-automated Ariol system. Multivariate trend survival analyses of the combined markers showed better patient survival and less tumour recurrence when more markers showed high nuclear expression. For the combination of the histone deacetylases and H3K56Ac, the hazard ratio (HR) for overall survival (OS) was 0.82 [95% confidence interval (CI) 0.72-0.94; P = 0.005] and the HR for distant recurrence-free survival (DRFS) was 0.77 (95% CI 0.64-0.92; P = 0.003) per additional marker showing high expression. Similarly, for the combination of histone deactylases and H4K16Ac, HRs of 0.86 (95% CI 0.76-0.97; P = 0.01) for OS and 0.79 (95% CI 0.68-0.93; P = 0.006) for DRFS were observed per additional marker showing high expression. CONCLUSIONS: The studied epigenetic markers showed clinical prognostic value in colorectal cancer, both as individual markers and when combined into multimarker analyses. These results indicate that epigenetic mechanisms play an important role in colorectal carcinogenesis.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Epigenesis, Genetic , Histone Deacetylases/biosynthesis , Histones/metabolism , Aged , Biomarkers, Tumor/analysis , Cell Nucleus/metabolism , Colorectal Neoplasms/mortality , Female , Histone Deacetylases/analysis , Histones/analysis , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Proportional Hazards Models , Tissue Array Analysis
2.
Transl Res ; 165(5): 578-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25488396

ABSTRACT

Aging is one of the prime risk factors for the development of cancer. Expression patterns of epigenetic regulators, including histone modification levels, are altered during aging of normal cells, a phenomenon referred to as epigenetic drift. Furthermore, it is known that epigenetic mechanisms are involved in the development of cancer. We hypothesized that expression of histone modifications, acetylation of histone 3 lysine 9 (H3K9Ac) and trimethylation of histone 3 lysine 27 (H3K27me3), with reported normal age-related expression patterns might show an age-dependent prognostic value in colorectal cancer (CRC). To quantify expression, we performed immunohistochemical staining of these histone modifications on a tissue microarray containing colorectal tissues of the 254 patients with TNM stage I-III CRC. Stratification of patients according to survival status revealed age-related tumor expression patterns of both H3K9Ac and H3K27me3. Decreased expression with advancing age was observed in patients who were alive after follow-up (no-event group), whereas increased expression with advancing age was observed in patients who presented with a recurrence or death in follow-up (event group). These opposite expression patterns translated into an age-dependent prognostic value in CRC for the individual histone modifications and their combination. The prognostic value reverses with advancing age, high nuclear expression associated with good clinical outcome in young adults, and, in contrast, with worse clinical outcome in elderly patients. In conclusion, for the first time, we demonstrated prognostic impact of epigenetic biomarkers that reverses with advancing age. This new insight supports the hypothesis that CRC biology is different in young vs elderly patients and emphasizes the importance of focusing on age-related effects in CRC.


Subject(s)
Aging/genetics , Aging/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Histones/genetics , Histones/metabolism , Acetylation , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cohort Studies , Epigenesis, Genetic , Female , Gene Expression Profiling , Histones/chemistry , Humans , Male , Methylation , Middle Aged , Prognosis , Translational Research, Biomedical
3.
PLoS One ; 9(9): e108265, 2014.
Article in English | MEDLINE | ID: mdl-25243792

ABSTRACT

Numerous changes in epigenetic mechanisms have been described in various types of tumors. In search for new biomarkers, we investigated the expression of Polycomb-group (PcG) proteins EZH2, BMI1 and SUZ12 and associated histone modification H3K27me3 in colorectal cancer. Nuclear expression of PcG proteins and histone modification H3K27me3 were immunohistochemically (IHC) stained on a tissue microarray (TMA), including 247 tumor tissues and 47 normal tissues, and scored using the semi-automated Ariol system. Tumor tissues showed higher expression of EZH2 (p = 0.05) and H3K27me3 (p<0.001) as compared to their normal counterparts. Combined marker trend analyses indicated that an increase in the number of markers showing high expression was associated with better prognosis. High expression of all four markers in the combined marker analyses was correlated with the best patient survival and the longest recurrence-free survival, with overall survival (p = 0.01, HR 0.42(0.21-0.84)), disease-free survival (p = 0.007, HR 0.23(0.08-0.67) and local recurrence-free survival (p = 0.02, HR 0.30(0.11-0.84)). In conclusion, we found that expression of PcG proteins and H3K27me3 showed prognostic value in our study cohort. Better stratification of patients was obtained by combining the expression data of the investigated biomarkers as compared to the individual markers, underlining the importance of investigating multiple markers simultaneously.


Subject(s)
Colorectal Neoplasms/metabolism , Histones/metabolism , Polycomb-Group Proteins/physiology , Aged , Female , Humans , Male , Middle Aged , Prognosis
4.
BMC Cancer ; 14: 604, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25139823

ABSTRACT

BACKGROUND: Breast cancer is a heterogeneous disease with a highly variable clinical outcome in which both genetic and epigenetic changes have critical roles. We investigated tumor expression levels of histone-modifying enzymes LSD1, HDAC2 and SIRT1 in relation with patient survival and tumor relapse in a retrospective cohort of 460 breast cancer patients. Additionally, we correlated expression levels with tumor differentiation and tumor cell proliferation. METHODS: Immunohistochemical staining for LSD1, HDAC2 and SIRT1 was performed on tissue microarrays of tumor and corresponding normal formalin-fixed paraffin-embedded tissues from breast cancer patients. Median nuclear expression levels in tumor tissues were used to divide the patients into low and high expression categories. In combined expression analyses, patients were divided into four subgroups: 1, all enzymes below-median; 2, one enzyme above-median; 3, two enzymes above-median; 4, all three enzymes above-median. The Cox proportional hazard model was used for univariate and multivariate survival analyses. The Pearson Chi-square method was used to assess correlation of combined expression levels with tumor cell proliferation and tumor differentiation. RESULTS: Expression of LSD1 and SIRT1, but not of HDAC2, was significantly increased in tumor tissues compared to their normal counterparts (both p < 0.001). Multivariate survival analyses identified SIRT1 as independent prognostic factor for relapse-free survival (RFS) with a hazard ratio (HR) of 1.34 (95% CI = 1.04-1.74, p = 0.02). For overall survival (OS), no significant differences were found when the individual enzymes were analyzed. Analyses of combined expression levels of the three histone-modifying enzymes correlated with OS (p = 0.03) and RFS (p = 0.006) with a HR of respectively 1.49 (95% CI = 1.07-2.08) and 1.68 (95% CI = 1.16-2.44) in multivariate analyses and were also related to tumor differentiation (p < 0.001) and tumor cell proliferation (p = 0.002). CONCLUSIONS: When the combined expression levels were analyzed, high expression of LSD1, HDAC2 and SIRT1 showed shorter patient survival time and shorter time to tumor relapse and correlated with poor tumor differentiation and a high level of tumor cell proliferation. Expression of these histone-modifying enzymes might therefore be involved in breast cancer pathogenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Histone Deacetylase 2/metabolism , Histone Demethylases/metabolism , Sirtuin 1/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Retrospective Studies , Survival Analysis , Young Adult
5.
BMC Cancer ; 14: 531, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25047223

ABSTRACT

BACKGROUND: Post-translational modification of histone tails by methylation plays an important role in tumorigenesis. In this study, we investigated the nuclear expression of H3K4me3, H3K9me3 and H4K20me3 in early-stage colon cancer in relation to clinical outcome. METHODS: Tumor tissue cores of 254 TNM stage I-III colorectal cancer patients were immunohistochemically stained for H3K4me3, H3K9me3 and H4K20me3 and scored using the semi-automated Ariol system. Cox proportional hazard trend analyses were performed to assess the prognostic value of the combined markers with respect to patient survival and tumor recurrence. RESULTS: The histone methylation markers only showed prognostic value in early-stage (TNM stage I and II) colon cancer. Therefore, only this patient set (n = 121) was used for further statistical analyses. Low nuclear expression of H3K4me3, and high expression of H3K9me3 and H4K20me3 were associated with good prognosis. In combined marker analyses, the patient group showing most favorable expression (low H3K4me3, high H3K9me3 and high H4K20me3) was associated with the best prognosis. Multivariate trend analyses showed significantly increased hazard ratios (HR) for each additional marker showing unfavorable expression, as compared to the "all favorable" reference group. The HR for disease-free survival was 3.81 (1.72-8.45; p = 0.001), for locoregional recurrence-free survival 2.86 (1.59-5.13; p < 0.001) and for distant recurrence-free survival 2.94 (1.66-5.22; p < 0.001). CONCLUSIONS: Combined nuclear expression of histone modifications H3K4me3, H3K9me3 and H4K20me3 is prognostic in early-stage colon cancer. The combination of expression of the three histone modifications provides better stratification of patient groups as compared to the individual markers and provides a good risk assessment for each patient group.


Subject(s)
Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Histones/metabolism , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/pathology , Adult , Aged , Aged, 80 and over , Cell Nucleus/metabolism , Colonic Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Methylation , Middle Aged , Neoplasm Recurrence, Local/metabolism , Prognosis , Proportional Hazards Models , Survival Analysis , Tissue Array Analysis
6.
Cancer Microenviron ; 6(1): 31-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-21732187

ABSTRACT

Immune cells are known to affect clinical outcome in colorectal cancer. Subsets of immune cells can both support and inhibit immunological interaction with tumor cells. We examined the clinical impact of T cells that are supposed to be responsible for the down regulation of a T cell response: regulatory T cells or Tregs. The study population (n = 76) consisted of a random population of colorectal cancer patients who did not receive any (neo-) adjuvant therapy, with a median follow-up time of 7.3 years (range 0.1-23.1 years). Expression of FoxP3 was used as an immunohistochemical marker to identify Tregs. We considered FoxP3+ cells present in tumor stroma and tumor epithelium separately, and related results to clinical outcome and to data on CD8+ immune cell infiltration that we previously obtained in the same patient cohort. All samples showed presence of Foxp3+ cells and in the majority of the patients (85.5%) these cells were also present in the tumor epithelial compartment. A relative high level of Foxp3+ cells in the tumor epithelium was significantly related to down regulation of HLA Class I expression (p-value 0.03). There was a trend, but no significant relation, towards a longer overall survival (p-value; 0.084) and disease-free survival (p-value; 0.073) when high levels of Foxp3+ cells were present in the tumor epithelium. More importantly, the ratio of CD8+/Foxp3+ cells did show a significant correlation with distant-recurrence-free survival. This was the case for both Foxp3+ cells specifically located in the tumor epithelium (p-value 0.024) as well as in the stroma compartment (p-value 0.018). Unfortunately due to the small sample size the ratios did not retain their statistical significance in multivariate analysis. These results provide further evidence that local interactions in the cancer microenvironment between tumor cells and immune cells are not only determined by tumor cell-related factors like HLA expression, but also by interactions among immune cells.

7.
Breast Cancer Res Treat ; 134(3): 1103-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22476853

ABSTRACT

Long interspersed element 1 (LINE-1), a non-coding genomic repeat sequence, methylation status can influence tumor progression. In this study, the clinical significance of LINE-1 methylation status was assessed in primary breast cancer in young versus old breast cancer patients. LINE-1 methylation index (MI) was assessed by absolute quantitative assessment of methylated alleles (AQAMA) PCR assay. Initially, LINE-1 MI was assessed in a preliminary study of 235 tissues representing different stages of ductal breast cancer development. Next, an independent cohort of 379 primary ductal breast cancer patients (median follow-up 18.9 years) was studied. LINE-1 hypomethylation was shown to occur in DCIS and invasive breast cancer. In primary breast cancer it was associated with pathological tumor stage (p = 0.026), lymph node metastasis (p = 0.022), and higher age at diagnosis (>55, p < 0.001). In multivariate analysis, LINE-1 hypomethylation was associated with decreased OS (HR 2.19, 95 % CI 1.17-4.09, log-rank p = 0.014), DFS (HR 2.05, 95 % CI 1.14-3.67, log-rank p = 0.016) and increased DR (HR 2.83, 95 % CI 1.53-5.21, log-rank p = 0.001) in younger (≤55 years), but not older patients (>55 years). LINE-1 analysis of primary breast cancer demonstrated cancer-related age-dependent hypomethylation. In patients ≤55 years, LINE-1 hypomethylation portends a high-risk of DR.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , DNA Methylation , Long Interspersed Nucleotide Elements , Adult , Age Factors , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Transformation, Neoplastic/genetics , Chemotherapy, Adjuvant , Cohort Studies , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Young Adult
8.
Breast Cancer Res Treat ; 131(3): 859-69, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21479925

ABSTRACT

Breast cancer patients with similar clinical stage may experience different disease outcomes. Aberrant DNA methylation of primary breast tumors can have impact on the clinical outcome. This study aimed to assess clinical utility of tumor-specific methylated sequences (MINT17, 31) and tumor-related gene (RARß2) methylation classification in primary breast tumors. Absolute quantitative assessment of methylated alleles (AQAMA) was used to determine the methylation index (MI) of MINT17, MINT31, and RARß2 in 242 primary tumors of early stage breast cancer patients. Patients were classified into three methylation groups: meth-N, with normal methylation levels of all biomarkers; meth-L, with one biomarker hypermethylation; and meth-H, with hypermethylation of >1 biomarker. Disease outcome of methylation groups was compared during follow-up. MI of all biomarkers was successfully obtained in 237 tumors of which 79 (33%) were classified as meth-N, 86 (36%) as meth-L, and 72 (30%) as meth-H. Meth-H status was a risk factor for distant recurrence (DR) (log-rank P = 0.007) and shorter disease-free survival (DFS) (log-rank P = 0.039). Methylation classification had strongest prognostic value for patients with ER-negative tumors. In multivariate analysis (n = 222), ER-negative meth-H patients had a 4.1-fold increased risk of DR (95% CI 1.80-9.59; meth-N HR 1.0, P = 0.001), a 4.2-fold increased risk of overall recurrence (OR) (95% CI 1.88-9.47; meth-N HR 1.0, P = 0.001), and a 3.1-fold shorter DFS (95% CI 1.57-5.98; meth-N HR 1.0, P = 0.003). Methylation classification of primary breast cancer is an independent prognostic factor for disease outcome in patients with ER-negative tumors. The study's findings will have to be confirmed in an independent dataset.


Subject(s)
Breast Neoplasms/classification , DNA Methylation , Receptors, Estrogen/analysis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , CpG Islands , Female , Humans , Middle Aged , Neoplasm Staging , Prognosis , Recurrence
9.
Clin Cancer Res ; 15(5): 1801-7, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223509

ABSTRACT

PURPOSE: The CpG island methylator phenotype (CIMP) may be associated with development of malignancy through coordinated inactivation of tumor suppressor and tumor-related genes (TRG) and methylation of multiple noncoding, methylated-in-tumor (MINT) loci. These epigenetic changes create a distinct CIMP pattern that has been linked to recurrence and survival in gastrointestinal cancers. Because epigenetic inactivation of TRGs also has been shown in malignant melanoma, we hypothesized the existence of a clinically significant CIMP in cutaneous melanoma progression. EXPERIMENTAL DESIGN: The methylation status of the CpG island promoter region of TRGs related to melanoma pathophysiology (WIF1, TFPI2, RASSF1A, RARbeta2, SOCS1, and GATA4) and a panel of MINT loci (MINT1, MINT2, MINT3, MINT12, MINT17, MINT25, and MINT31) in primary and metastatic tumors of different clinical stages (n=122) was assessed. RESULTS: Here, we show an increase in hypermethylation of the TRGs WIF1, TFPI2, RASSF1A, and SOCS1 with advancing clinical tumor stage. Furthermore, we find a significant positive association between the methylation status of MINT17, MINT31, and TRGs. The methylation status of MINT31 is associated with disease outcome in stage III melanoma. CONCLUSIONS: These findings show the significance of a CIMP pattern that is associated with advancing clinical stage of malignant melanoma. Future prospective large-scale studies may determine if CIMP-positive primary melanomas are at high risk of metastasis or recurrence.


Subject(s)
Biomarkers, Tumor/genetics , CpG Islands/genetics , DNA Methylation , Genes, Tumor Suppressor , Melanoma/genetics , Skin Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cells, Cultured , Child , Dermis/cytology , Dermis/metabolism , Disease Progression , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lymphatic Metastasis , Male , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Middle Aged , Neoplasm Staging , Phenotype , Prognosis , Promoter Regions, Genetic , Skin Neoplasms/secondary , Survival Rate , Young Adult
10.
Expert Opin Ther Targets ; 11(12): 1639-48, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18020983

ABSTRACT

The estrogen receptor (ER) belongs to the group of sex hormone receptors and binds the biologically active form of estrogen, 17beta-estradiol. Expression of ER in tumor tissue is a well-established prognostic marker in breast cancer. The role of ER has been extensively studied in several other types of human cancers. This report investigates the potential role of ER as a surrogate marker for predicting melanoma progression, response to therapy, and patient survival. In addition, the authors review what is known, so far, about ER signaling pathways and their potential role in carcinogenesis and progression of cutaneous melanoma. Possibilities and limitations of using ER as a therapeutic target in the treatment of melanoma is also discussed.


Subject(s)
Melanoma/metabolism , Receptors, Estrogen/metabolism , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Humans , Melanoma/drug therapy , Melanoma/genetics , Receptors, Estrogen/genetics
11.
J Clin Oncol ; 25(31): 4887-94, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17971584

ABSTRACT

PURPOSE: Overexpression of cyclooxygenase-2 (COX-2) in gastric cancer has been shown to enhance tumor progression. We investigated whether silencing by promoter region hypermethylation of the COX-2 gene contributes to disease outcome in gastric cancer. MATERIALS AND METHODS: COX-2 methylation status was initially assessed by capillary array electrophoresis methylation-specific polymerase chain reaction (CAE-MSP) and COX-2 protein expression by immunohistochemistry (IHC) in 40 primary gastric cancer tissues in a pilot study. Prognostic end points of correlative studies of COX-2 methylation status were time to recurrence, overall survival, and standard clinicopathologic features. CAE-MSP analysis was then validated in a second independent gastric cancer population (n = 137). RESULTS: COX-2 methylation was detected in 23% and 28% of the pilot and validation patient groups, respectively. COX-2 expression (IHC) in gastric tumors inversely correlated with COX-2 gene methylation status in the pilot study (P = .02). COX-2 methylation in tumors was significantly associated with lower T, N, and TNM stage in the validation patient group (P = .02, P = .006, and P = .008, respectively). Patients with COX-2 methylated tumors had significantly longer time to recurrence and improved overall survival in a multivariate analysis in the validation patient group (hazard ratio[HR], 0.49; 95% CI, 0.24% to 0.99%; HR, 0.62; 95% CI, 0.38% to 0.99%, respectively). CONCLUSION: Hypermethylation of COX-2 gene promoter was identified as an independent prognostic factor in gastric cancer patients. The results suggest promoter hypermethylation to be an important regulatory mechanism of COX-2 expression in gastric cancer and an important prognostic biomarker.


Subject(s)
Cyclooxygenase 2/genetics , Gene Silencing/physiology , Stomach Neoplasms/genetics , DNA Methylation , Humans , Pilot Projects , Prognosis , Retrospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...