Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Breast Care (Basel) ; 16(5): 523-531, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34720812

ABSTRACT

BACKGROUND: PITX2 DNA methylation has been shown to predict outcomes in high-risk breast cancer patients after anthracycline-based chemotherapy. To determine its prognostic versus predictive value, the impact of PITX2 DNA methylation on outcomes was studied in an untreated cohort vs. an anthracycline-treated triple-negative breast cancer (TNBC) cohort. MATERIAL AND METHODS: The percent DNA methylation ratio (PMR) of paired-like homeodomain transcription factor 2 (PITX2) was determined by a validated methylation-specific real-time PCR test. Patient samples of routinely collected archived formalin-fixed paraffin-embedded (FFPE) tissue and clinical data from 144 TNBC patients of 2 independent cohorts (i.e., 66 untreated patients and 78 patients treated with anthracycline-based chemotherapy) were analyzed. RESULTS: The risk of 5- and 10-year overall survival (OS) increased continuously with rising PITX2 DNA methylation in the anthracycline-treated population, but it increased only slightly during 10-year follow-up time in the untreated patient population. PITX2 DNA methylation with a PMR cutoff of 2 did not show significance for poor vs. good outcomes (OS) in the untreated patient cohort (HR = 1.55; p = 0.259). In contrast, the PITX2 PMR cutoff of 2 identified patients with poor (PMR >2) vs. good (PMR ≤2) outcomes (OS) with statistical significance in the anthracycline-treated cohort (HR = 3.96; p = 0.011). The results in the subgroup of patients who did receive anthracyclines only (no taxanes) confirmed this finding (HR = 5.71; p = 0.014). CONCLUSION: In this hypothesis-generating study PITX2 DNA methylation demonstrated predominantly predictive value in anthracycline treatment in TNBC patients. The risk of poor outcome (OS) correlates with increasing PITX2 DNA methylation.

2.
Cell Biochem Funct ; 39(4): 571-583, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33608886

ABSTRACT

Despite the concerning adverse effects on tumour development, epigenetic drugs are very promising in cancer treatment. The aim of this study was to compare the differential effects of standard chemotherapy regimens (FEC: 5-fluorouracil plus epirubicine plus cyclophosphamide) in combination with epigenetic modulators (decitabine, valproic acid): (a) on gene methylation levels of selected tumour biomarkers (LINE-1, uPA, PAI-1, DAPK); (b) their expression status (uPA and PAI-1); (c) differentiation status (5meC and H3K27me3). Furthermore, cell survival as well as changes concerning the invasion capacity were monitored in cell culture models of breast cancer (MCF-7, MDA-MB-231). A significant overall decrease of cell survival was observed in the FEC-containing combination therapies for both cell lines. Methylation results showed a general tendency towards increased demethylation of the uPA and PAI-1 gene promoters for the MCF-7 cells, as well as the proapoptotic DAPK gene in the treatment regimens for both cell lines. The uPA and PAI-1 antigen levels were mainly increased in the supernatant of FEC-only treated MDA-MB-231 cells. DAC-only treatment induced an increase of secreted uPA protein in MCF-7 cell culture, while most of the VPA-containing regimens also induced uPA and PAI-1 expression in MCF-7 cell fractions. Epigenetically active substances can also induce a re-differentiation in tumour cells, as shown by 5meC, H3K27me3 applying ICC. SIGNIFICANCE OF THE STUDY: Epigenetic modulators especially in the highly undifferentiated and highly malignant MDA-MB-231 tumour cells significantly reduced tumour malignancy thus; further clinical studies applying specific combination therapies with epigenetic modulators may be warranted.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Decitabine/pharmacology , Epigenesis, Genetic/drug effects , Valproic Acid/pharmacology , Antimetabolites, Antineoplastic/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Decitabine/chemistry , Drug Screening Assays, Antitumor , Epigenesis, Genetic/genetics , Female , Humans , Tumor Cells, Cultured , Valproic Acid/chemistry
3.
Sci Rep ; 8(1): 16861, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442983

ABSTRACT

Significant evidence has accumulated that DNA-methylation of the paired-like homeodomain transcription factor 2 (PITX2) gene can serve as a prognostic and predictive biomarker in breast cancer. PITX2 DNA-methylation data have been obtained so far from microarray and polymerase chain reaction (PCR)-based research tests. The availability of an analytically validated in vitro methylation-specific real-time PCR assay format (therascreen PITX2 RGQ PCR assay) intended for the determination of the percent methylation ratio (PMR) in the (PITX2) promoter 2 prompted us to investigate whether the clinical performance of these different assay systems generate comparable clinical outcome data. Mathematically converted microarray data of a previous breast cancer study (n = 204) into PMR values leads to a PITX2 cut-off value at PMR 14.73. Recalculation of the data to experimentally equivalent PMRs with the PCR PITX2 assay leads to a cut-off value at PMR 12 with the highest statistical significance. This cut-off predicts outcome of high-risk breast cancer patients to adjuvant anthracycline-based chemotherapy (n = 204; Hazard Ratio 2.48; p < 0.001) comparable to microarray generated results (n = 204; Hazard ratio 2.32; p < 0.0001). The therascreen PITX2 RGQ PCR assay is an analytically validated test with high reliability and robustness and predicts outcome of high-risk breast cancer patients to anthracycline-based chemotherapy.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation/genetics , Homeodomain Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Transcription Factors/genetics , Biological Assay , Breast Neoplasms/pathology , DNA, Neoplasm/genetics , Female , Genetic Heterogeneity , Humans , Kaplan-Meier Estimate , Middle Aged , Paraffin Embedding , Reproducibility of Results , Tissue Fixation , Homeobox Protein PITX2
4.
J Cancer Res Clin Oncol ; 144(6): 1109-1118, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29546479

ABSTRACT

PURPOSE: Gene expression of a variety of the 15 members of the KLK serine protease family is dysregulated in ovarian cancer. We aimed at determining the clinical relevance of KLK13 and KLK14 mRNA expression in tumor tissues of a homogeneous patient cohort afflicted with advanced high-grade serous ovarian cancer (FIGO stage III/IV). METHODS: mRNA expression levels of KLK13 and KLK14 were assessed by quantitative PCR in tumor tissue of 91 patients and related with clinical factors and patients' outcome. RESULTS: There was no significant association of KLK13 and KLK14 mRNA expression with the clinical factors ascitic fluid volume or residual tumor mass. In univariate Cox regression analysis, elevated KLK13 mRNA levels were significantly linked with shorter progression-free (PFS; hazard ratio [HR] = 1.97, P = 0.020) and overall survival (OS; HR = 1.81, P = 0.041). High KLK14 mRNA levels were significantly associated with prolonged PFS (HR = 0.44, P = 0.017) and showed a trend towards significance for OS (HR = 0.55, P = 0.070). In multivariable analysis, including the factors age, residual tumor mass, ascitic fluid volume, KLK13, and KLK14, both KLKs, apart from residual tumor mass, remained statistically independent predictive markers: patients with high KLK13 mRNA expression levels displayed a more than twofold increase risk for shorter PFS (HR = 2.14, P = 0.020) as well as OS (HR = 2.05, P = 0.028), whereas elevated KLK14 mRNA values were found to be significant for both, prolonged PFS (HR = 0.36, P = 0.007) and OS (HR = 0.46, P = 0.037). CONCLUSION: These results indicate that in advanced high-grade serous ovarian cancer KLK13 may become proficient for tumor-supporting functions, whereas KLK14 may have adopted tumor-suppressing activity.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Kallikreins/genetics , Ovarian Neoplasms/genetics , RNA, Messenger/biosynthesis , Adult , Age Factors , Aged , Aged, 80 and over , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Disease-Free Survival , Female , Humans , Kallikreins/biosynthesis , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Survival Rate
5.
Int J Oncol ; 52(3): 755-767, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29328369

ABSTRACT

Triple-negative breast cancer (TNBC) constitutes a heterogeneous breast cancer subgroup with poor prognosis; survival rates are likely to be lower with TNBC compared to other breast cancer subgroups. For this disease, systemic adjuvant chemotherapy regimens often yield suboptimal clinical results. To improve treatment regimens in TNBC, identification of molecular biomarkers may help to select patients for individualized adjuvant therapy. Evidence has accumulated that determination of the methylation status of the PITX2 gene provides a predictive value in various breast cancer subgroups, either treated with endocrine-based therapy or anthracycline-containing chemotherapy. To further explore the validity of this novel predictive candidate biomarker, in the present exploratory retrospective study, determination of the PITX2 DNA-methylation status was assessed for non-metastatic TNBC patients treated with adjuvant anthracycline-based chemotherapy by molecular analysis of breast cancer tissues. The PITX2 DNA-methylation status was determined in fresh-frozen tumor tissue specimens (n=56) by methylation-specific qRT-PCR (qMSP) and the data related to disease-free and overall survival, applying an optimized DNA-methylation score of 6.35%. For non-metastatic TNBC patients treated with adjuvant systemic anthracycline-based chemotherapy, a low PITX2 DNA-methylation status (<6.35) defines TNBC patients with poor disease-free and overall survival. Univariate and multivariate analyses demonstrate the statistically independent predictive value of PITX2 DNA-methylation. For non-metastatic TNBC patients, selective determination of the PITX2 DNA-methylation status may serve as a cancer biomarker for predicting response to anthracycline-based adjuvant chemotherapy. The assay based on methylation of the PIXT2 gene can be applied to frozen and routinely available formalin-fixed, paraffin-embedded (FFPE) breast cancer tumor tissues that will not only define those TNBC patients who may benefit from anthracycline-based chemotherapy but also those who should be spared the necessity of such potentially toxic treatment. Such patients should be allocated to alternative treatment options.


Subject(s)
Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/therapy , Adult , Aged , Breast/pathology , Breast/surgery , Chemotherapy, Adjuvant/methods , DNA Methylation , Disease-Free Survival , Female , Follow-Up Studies , Humans , Middle Aged , Patient Selection , Pilot Projects , Retrospective Studies , Treatment Outcome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Homeobox Protein PITX2
6.
Dis Markers ; 2017: 4934608, 2017.
Article in English | MEDLINE | ID: mdl-29138528

ABSTRACT

High-risk breast cancer comprises distinct tumor entities such as triple-negative breast cancer (TNBC) which is characterized by lack of estrogen (ER) and progesterone (PR) and the HER2 receptor and breast malignancies which have spread to more than three lymph nodes. For such patients, current (inter)national guidelines recommend anthracycline-based chemotherapy as the standard of care, but not all patients do equally benefit from such a chemotherapy. To further improve therapy decision-making, predictive biomarkers are of high, so far unmet, medical need. In this respect, predictive biomarkers would permit patient selection for a particular kind of chemotherapy and, by this, guide physicians to optimize the treatment plan for each patient individually. Besides DNA mutations, DNA methylation as a patient selection marker has received increasing clinical attention. For instance, significant evidence has accumulated that methylation of the PITX2 (paired-like homeodomain transcription factor 2) gene might serve as a novel predictive and prognostic biomarker, for a variety of cancer diseases. This review highlights the current understanding of treatment modalities of high-risk breast cancer patients with a focus on recommended treatment options, with special attention on the future clinical application of PITX2 as a predictive biomarker to personalize breast cancer management.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , DNA Methylation , Homeodomain Proteins/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents/standards , Female , Humans , Practice Guidelines as Topic , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Homeobox Protein PITX2
7.
Biol Chem ; 398(10): 1151-1164, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28755528

ABSTRACT

Triple-negative breast cancer (TNBC), lacking the steroid hormone receptors ER and PR and the oncoprotein HER2, is characterized by its aggressive pattern and insensitivity to endocrine and HER2-directed therapy. Human kallikrein-related peptidases KLK1-15 provide a rich source of serine protease-type biomarkers associated with tumor growth and cancer progression for a variety of malignant diseases. In this study, recombinant KLK4 protein was generated and affinity-purified KLK4-directed polyclonal antibody pAb587 established to allow localization of KLK4 protein expression in tumor cell lines and archived formalin-fixed, paraffin-embedded TNBC tumor tissue specimens. For this, KLK4 protein expression was assessed by immunohistochemistry in primary tumor tissue sections (tissue microarrays) of 188 TNBC patients, mainly treated with anthracycline- or CMF-based polychemotherapy. KLK4 protein is localized in the cytoplasm of tumor and stroma cells. In this patient cohort, elevated stroma cell KLK4 expression, but not tumor cell KLK4 expression, is predictive for poor disease-free survival by univariate analysis (hazard ratio: 2.26, p=0.001) and multivariable analysis (hazard ratio: 2.12, p<0.01). Likewise, univariate analysis revealed a trend for statistical significance of elevated KLK4 stroma cell expression for overall survival of TNBC patients as well.


Subject(s)
Biomarkers, Tumor/metabolism , Kallikreins/metabolism , Triple Negative Breast Neoplasms/enzymology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/biosynthesis , Humans , Kallikreins/biosynthesis , Multivariate Analysis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
8.
Biol Chem ; 398(7): 765-773, 2017 06 27.
Article in English | MEDLINE | ID: mdl-27935848

ABSTRACT

In serous ovarian cancer, the clinical relevance of tumor cell-expressed plasmin(ogen) (PLG) has not yet been evaluated. Due to its proteolytic activity, plasmin supports tumorigenesis, however, angiostatin(-like) fragments, derived from PLG, can also function as potent anti-tumorigenic factors. In the present study, we assessed PLG protein expression in 103 cases of advanced high-grade serous ovarian cancer (FIGO III/IV) by immunohistochemistry (IHC). In 70/103 cases, positive staining of tumor cells was observed. In univariate Cox regression analysis, PLG staining was positively associated with prolonged overall survival (OS) [hazard ratio (HR)=0.59, p=0.026] of the patients. In multivariable analysis, PLG, together with residual tumor mass, remained a statistically significant independent prognostic marker (HR=0.49, p=0.009). In another small patient cohort (n=29), we assessed mRNA expression levels of PLG by quantitative PCR. Here, elevated PLG mRNA levels were also significantly associated with prolonged OS of patients (Kaplan-Meier analysis; p=0.001). This finding was validated by in silico analysis of a microarray data set (n=398) from The Cancer Genome Atlas (Kaplan-Meier analysis; p=0.031). In summary, these data indicate that elevated PLG expression represents a favorable prognostic biomarker in advanced (FIGO III/IV) high-grade serous ovarian cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Plasminogen/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Plasminogen/genetics , Plasminogen Activator Inhibitor 1/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retrospective Studies , Survival Analysis , Urokinase-Type Plasminogen Activator/genetics , Young Adult
9.
Biol Chem ; 397(12): 1265-1276, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27483364

ABSTRACT

Most members of the kallikrein-related peptidase family have been demonstrated to be dysregulated in ovarian cancer and modulate tumor growth, migration, invasion, and resistance to chemotherapy. In the present study, we assessed the mRNA expression levels of KLK6 and KLK8 by quantitative PCR in 100 patients with advanced serous ovarian cancer FIGO stage III/IV. A pronounced correlation between KLK6 and KLK8 mRNA expression (rs = 0.636, p < 0.001) was observed, indicating coordinate expression of both peptidases. No significant associations of clinical parameters with KLK6, KLK8, and a combined score KLK6+KLK8 were found. In univariate Cox regression analysis, elevated mRNA levels of KLK6 were significantly linked with shortened overall survival (OS) (hazard ratio [HR] = 2.07, p = 0.007). While KLK8 values were not associated with patients' outcome, high KLK6+KLK8 values were significantly associated with shorter progression-free survival (HR = 1.82, p = 0.047) and showed a trend towards significance in the case of OS (HR = 1.82, p = 0.053). Strikingly, in multivariable analysis, elevated KLK6 mRNA values, apart from residual tumor mass, remained an independent predictive marker for poor OS (HR = 2.33, p = 0.005). As KLK6 mRNA and protein levels correlate, KLK6 may represent an attractive therapeutic target for potent and specific inhibitors of its enzymatic activity.


Subject(s)
Gene Expression Regulation, Neoplastic , Kallikreins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Middle Aged , Multivariate Analysis , Ovarian Neoplasms/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
10.
BMC Med Genomics ; 6: 53, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24299561

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC), a leading cause of cancer deaths, represents a heterogeneous group of neoplasms, mostly comprising squamous cell carcinoma (SCC), adenocarcinoma (AC) and large-cell carcinoma (LCC). The objectives of this study were to utilize integrated genomic data including copy-number alteration, mRNA, microRNA expression and candidate-gene full sequencing data to characterize the molecular distinctions between AC and SCC. METHODS: Comparative genomic hybridization followed by mutational analysis, gene expression and miRNA microarray profiling were performed on 123 paired tumor and non-tumor tissue samples from patients with NSCLC. RESULTS: At DNA, mRNA and miRNA levels we could identify molecular markers that discriminated significantly between the various histopathological entities of NSCLC. We identified 34 genomic clusters using aCGH data; several genes exhibited a different profile of aberrations between AC and SCC, including PIK3CA, SOX2, THPO, TP63, PDGFB genes. Gene expression profiling analysis identified SPP1, CTHRC1 and GREM1 as potential biomarkers for early diagnosis of the cancer, and SPINK1 and BMP7 to distinguish between AC and SCC in small biopsies or in blood samples. Using integrated genomics approach we found in recurrently altered regions a list of three potential driver genes, MRPS22, NDRG1 and RNF7, which were consistently over-expressed in amplified regions, had wide-spread correlation with an average of ~800 genes throughout the genome and highly associated with histological types. Using a network enrichment analysis, the targets of these potential drivers were seen to be involved in DNA replication, cell cycle, mismatch repair, p53 signalling pathway and other lung cancer related signalling pathways, and many immunological pathways. Furthermore, we also identified one potential driver miRNA hsa-miR-944. CONCLUSIONS: Integrated molecular characterization of AC and SCC helped identify clinically relevant markers and potential drivers, which are recurrent and stable changes at DNA level that have functional implications at RNA level and have strong association with histological subtypes.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genomics , Lung Neoplasms/genetics , Adult , Aged , Aged, 80 and over , DNA Copy Number Variations/genetics , Female , Gene Expression Profiling , Humans , Male , MicroRNAs/genetics , Middle Aged , RNA, Messenger/genetics
11.
Cell Biochem Funct ; 29(8): 651-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21887697

ABSTRACT

Epigenetic drugs are promising add-ons to cancer treatment; still, adverse effects concerning tumour promotion have been reported occasionally. In this in vitro study, we investigated the effect of combination treatment of decitabine with anthracycline-based chemotherapy [5-fluorouracil plus epirubicine plus cyclophosphamide (FEC)] on viability and metastatic activity of breast cancer cell lines, MDA-MB-231 (estrogen receptor-negative) and MCF-7 (estrogen receptor-positive). The effect of decitabine and its combined treatment with FEC on viability of both cancer cell lines was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide and adenosine triphosphate (ATP) cell survival assays. DNA methylation specific real-time polymerase chain reaction (PCR) (Methylight®) was employed to document the methylation status of the metastasis-relevant urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-I (PAI-1) genes. Additionally, protein expression levels of uPA and PAI-1 were determined using enzyme-linked immunosorbent assays. Invasion capacity of cells was assayed using Matrigel® invasion assay. Decitabine lowered the viability of MCF-7 cells, although MDA-MB-231 cells were not affected. Decitabine did not augment FEC-mediated cytotoxicity in both cell lines. In MCF-7 cells, methylation of the uPA and PAI-1 gene promoter was significantly reduced by decitabine or decitabine plus FEC. Protein levels of uPA and PAI-1 were induced by all treatments. Decitabine significantly induced the invasion capacity of MCF-7 cells, whereas all of the drugs resulted in decreased invasion capacity of MDA-MB-231. Our results suggest differential effects of single-dose decitabine and its combination with FEC on the metastatic capacity and survival of breast cancer cell lines endowed with different metastatic behaviour.


Subject(s)
Anthracyclines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/analogs & derivatives , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Plasminogen Activator Inhibitor 1/genetics , Urokinase-Type Plasminogen Activator/genetics , Azacitidine/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Methylation/drug effects , Decitabine , Female , Humans , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism
12.
Expert Rev Mol Diagn ; 10(8): 1051-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21080821

ABSTRACT

The prognostic and/or predictive value of the cancer biomarkers, urokinase-type plasminogen activator (uPA) and its inhibitor (plasminogen activator inhibitor [PAI]-1), determined by ELISA in tumor-tissue extracts, was demonstrated for several cancer types in numerous clinically relevant retrospective or prospective studies, including a multicenter breast cancer therapy trial (Chemo-N0). Consequently, for the first time ever for any cancer biomarker for breast cancer, uPA and PAI-1 have reached the highest level of evidence, level-of-evidence-1. At present, two other breast cancer therapy trials, NNBC-3 and Plan B, also incorporating uPA and PAI-1 as treatment-assignment tools are in effect. Furthermore, small synthetic molecules targeting uPA are currently in Phase II clinical trials in patients afflicted with advanced cancer of the ovary, breast or pancreas.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasms/diagnosis , Neoplasms/drug therapy , Plasminogen Activator Inhibitor 1/analysis , Urokinase-Type Plasminogen Activator/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Enzyme-Linked Immunosorbent Assay , Female , Humans , Neoplasms/chemistry , Oligonucleotides, Antisense/therapeutic use , Plasminogen Activator Inhibitor 1/genetics , Prognosis , RNA, Small Interfering/therapeutic use , Retrospective Studies , Serine Proteinase Inhibitors/therapeutic use , Urokinase-Type Plasminogen Activator/genetics
13.
Expert Rev Mol Diagn ; 10(7): 947-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20964613

ABSTRACT

In cancer, the serine protease urokinase-type plasminogen activator, its inhibitor (plasminogen activator inhibitor type-1) and the receptor (CD87), among other proteolytic factors, are involved in tumor cell dissemination and turnover of the extracellular matrix. Unsurprisingly, a battery of very uniform data, amassed since the end of the 1990s, has put these members of the plasminogen activation system into the forefront of prognostic/predictive cancer biomarkers relevant to predict the clinical course of cancer patients and their response to cancer therapy. The present review focuses on the molecular characteristics of the disease forecast biomarkers urokinase-type plasminogen activator and plasminogen activator inhibitor type-1, and techniques to quantitatively assess these cancer biomarkers, in the context of potential clinical application and personalized disease management.


Subject(s)
Neoplasm Metastasis/diagnosis , Neoplasms/diagnosis , Plasminogen Activator Inhibitor 1/analysis , Urokinase-Type Plasminogen Activator/analysis , Biomarkers, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Plasminogen Activator Inhibitor 1/therapeutic use , Prognosis , Urokinase-Type Plasminogen Activator/metabolism
14.
Biomark Med ; 4(4): 505-22, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20701440

ABSTRACT

Currently, decision-making for breast cancer treatment in the clinical setting is mainly based on clinical data, histomorphological features of the tumor tissue and a few cancer biomarkers such as steroid hormone receptor status (estrogen and progesterone receptors) and oncoprotein HER2 status. Although various therapeutic options were introduced into the clinic in recent decades, with the objective of improving surgery, radiotherapy, biochemotherapy and chemotherapy, varying response of individual patients to certain types of therapy and therapy resistance is still a challenge in breast cancer care. Therefore, since breast cancer treatment should be based on individual features of the patient and her tumor, tailored therapy should be an option by integrating cancer biomarkers to define patients at risk and to reliably predict their course of the disease and/or response to cancer therapy. Recently, candidate-marker approaches and genome-wide transcriptomic and epigenetic screening of different breast cancer tissues and bodily fluids resulted in new promising biomarker panels, allowing breast cancer prognosis, prediction of therapy response and monitoring of therapy efficacy. These biomarkers are now subject of validation in prospective clinical trials.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Humans
16.
Clin Cancer Res ; 13(17): 5095-102, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17785563

ABSTRACT

PURPOSE: The objective of this study was to analyze the hypermethylation of tumor-related gene promoters for an association with therapy response and clinicopathologic features of neoadjuvant-treated gastric cancer patients. Furthermore, we analyzed the relationship of promoter hypermethylation with microsatellite instability and loss of heterozygosity (LOH) of the tumors. EXPERIMENTAL DESIGN: Pretherapeutic biopsies of 61 patients, subsequently treated with cisplatin and 5-fluorouracil, were studied. Methylation analysis of six gene promoters was done using MethyLight technology. Microsatellite analysis was mainly done in previous studies. RESULTS: The methylation frequencies for the analyzed genes were MGMT, 44%; LOX, 53%; p16, 46%, E-cadherin, 30%; 14-3-3sigma, 69%; and HPP1, 82%. Concordant methylation of more than three genes was found in 46% of the tumors and was inversely correlated with the LOH rate (P = 9 x 10(-5)) and associated with female gender (P = 0.049), nonintestinal type tumors (P = 0.04), and a nonproximal tumor location (P = 0.003). No statistically significant association between the methylation of a single gene or the concordant methylation of multiple genes was found with response or survival. However, patients with none or only one methylated gene showed a trend for an increase in survival (5-year survival rate, 83% versus 35%; P = 0.067). CONCLUSION: The highly significant inverse correlation of promoter methylation and LOH rate reflects major alternative molecular pathways in gastric carcinogenesis. Methylation was not statistically significantly associated with the response to cisplatin/5-fluorouracil-based therapy. However, a concordant methylation of more than three genes defines subgroups of gastric cancer with distinct biological and genetic characteristics.


Subject(s)
DNA Methylation , Stomach Neoplasms/genetics , Adult , Aged , Cisplatin/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Loss of Heterozygosity , Male , Microsatellite Instability , Middle Aged , Neoadjuvant Therapy , Promoter Regions, Genetic , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
17.
Clin Cancer Res ; 11(8): 3025-31, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15837757

ABSTRACT

PURPOSE: We evaluated the expression of seven therapy-related genes to predict the clinical outcome of advanced gastric cancer patients treated with a neoadjuvant chemotherapeutic protocol. EXPERIMENTAL DESIGN: Pretherapeutic, formalin-fixed, and paraffin-embedded biopsies of 61 patients, who received a 5-fluorouracil (5-FU)- and cisplatin-based chemotherapy were studied. The expressions of the 5-FU-related genes TS, DPD, and TP and of the cisplatin-related genes ERCC1, ERCC4, KU80, and GADD45A were analyzed by quantitative real-time PCR. The expression levels of single genes and of various combinations were tested for an association with response and overall survival. RESULTS: High DPD levels were more frequently found in nonresponding patients and were associated with worse survival. GADD45A and TP levels showed weak associations with response, but GADD45A expression correlated with survival. There was no association with response for TS expression, but tumors with a high TS level were associated with worse survival. The combination of GADD45A and TP revealed the strongest predictive effect. High expression values of TP and/or GADD45A were exclusively found in nonresponding patients (P = 0.002) and were associated with a significantly poorer survival (P = 0.04). CONCLUSIONS: Combined gene expression levels of TP and GADD45A represent a new variable to predict the clinical outcome after neoadjuvant chemotherapy in gastric cancer. The association of DPD expression with response and survival underlines a predominant role of DPD to predict 5-FU sensitivity. The association of TS expression levels with survival but not with response suggests an importance of this gene for tumor progression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/genetics , Stomach Neoplasms/drug therapy , Thymidine Phosphorylase/genetics , Adult , Aged , Antigens, Nuclear/genetics , Cisplatin/administration & dosage , DNA-Binding Proteins/genetics , Dihydrouracil Dehydrogenase (NADP)/genetics , Endonucleases/genetics , Female , Fluorouracil/administration & dosage , Humans , Ku Autoantigen , Male , Middle Aged , Neoadjuvant Therapy , Predictive Value of Tests , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Analysis , Thymidylate Synthase/genetics , Treatment Outcome
18.
Biol Chem ; 384(3): 483-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12715899

ABSTRACT

In the human B cell line P493-6 two mitogenic signals, the Epstein-Barr virus nuclear antigen 2 (EBNA2) and myc, can be independently regulated by means of an estrogen receptor fusion construct or an inducible expression vector, respectively. Shut off of EBNA2, either in the presence or absence of myc, leads to a significant increase in enzymatic activity and surface expression of ecto-5'-nucleotidase (CD73) as well as an increased adenosine receptor response in cyclic AMP formation. Shut off of myc expression has a small additional positive effect on CD73 activity. Among the four different subtypes of adenosine receptors, the A2a receptor exclusively is subject to regulation in this system, which is substantiated by pharmacologic data (specific agonists and inhibitors), as well as on the mRNA level. With up-regulated CD73 and A2a, cells also respond to 5'-AMP with increased cyclic AMP formation. Turn on of EBNA2 has the reverse effect of repression of CD73 and A2a expression. The time course of both induction and repression of CD73 and A2a is rather slow.


Subject(s)
5'-Nucleotidase/biosynthesis , B-Lymphocytes/metabolism , Receptors, Adenosine A2/biosynthesis , Cell Line , Cyclic AMP/biosynthesis , Down-Regulation , Epstein-Barr Virus Nuclear Antigens/biosynthesis , Humans , Phenotype , Proto-Oncogene Proteins c-myc/biosynthesis , Up-Regulation , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...