105 results on '"Heidi Fiegl"'
Search Results
2. A retrospective validation of CanAssist Breast in European early-stage breast cancer patient cohort
- Author
-
Aparna Gunda, Chetana Basavaraj, Chandra Prakash Serkad V, Manjula Adinarayan, Ramu Kolli, Mallikarjuna Siraganahalli Eshwaraiah, Cristina Saura, Fiorella Ruiz, Patricia Gomez, Vicente Peg, Jose Jimenez, Susanne Sprung, Heidi Fiegl, Christine Brunner, Daniel Egle, GS Bhattacharyya, and Manjiri M Bakre
- Subjects
Hormone-receptor positive ,Prognostication ,early-stage breast cancer ,recurrence ,chemotherapy ,European ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
CanAssist Breast (CAB), a prognostic test uses immunohistochemistry (IHC) approach coupled with artificial intelligence-based machine learning algorithm for prognosis of early-stage hormone-receptor positive, HER2/neu negative breast cancer patients. It was developed and validated in an Indian cohort. Here we report the first blinded validation of CAB in a multi-country European patient cohort. FFPE tumor samples from 864 patients were obtained from-Spain, Italy, Austria, and Germany. IHC was performed on these samples, followed by recurrence risk score prediction. The outcomes were obtained from medical records. The performance of CAB was analyzed by hazard ratios (HR) and Kaplan Meier curves. CAB stratified European cohort (n = 864) into distinct low- and high-risk groups for recurrence (P 50 years (HR: 2.93 (1.44–5.96), P = 0.0002). CAB had an HR of 2.57 (1.26–5.26), P = 0.01) in women with N1 disease. CAB stratified significantly higher proportions (77%) as low-risk over IHC4 (55%) (P
- Published
- 2022
- Full Text
- View/download PDF
3. A comparison of four technologies for detecting p53 aggregates in ovarian cancer
- Author
-
Nicole Heinzl, Katarzyna Koziel, Elisabeth Maritschnegg, Astrid Berger, Elisabeth Pechriggl, Heidi Fiegl, Alain G. Zeimet, Christian Marth, Robert Zeillinger, and Nicole Concin
- Subjects
p53 ,protein aggregation ,ovarian cancer ,immunofluorescence staining ,immunoprecipitation ,ELISA ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The tumor suppressor protein p53 is mutated in half of all cancers and has been described to form amyloid-like structures, commonly known from key proteins in neurodegenerative diseases. Still, the clinical relevance of p53 aggregates remains largely unknown, which may be due to the lack of sensitive and specific detection methods. The aim of the present study was to compare the suitability of four different methodologies to specifically detect p53 aggregates: co-immunofluorescence (co-IF), proximity ligation assay (PLA), co-immunoprecipitation (co-IP), and the p53-Seprion-ELISA in cancer cell lines and epithelial ovarian cancer tissue samples. In 7 out of 10 (70%) cell lines, all applied techniques showed concordance. For the analysis of the tissue samples co-IF, co-IP, and p53-Seprion-ELISA were compared, resulting in 100% concordance in 23 out of 30 (76.7%) tissue samples. However, Co-IF lacked specificity as there were samples, which did not show p53 staining but abundant staining of amyloid proteins, highlighting that this method demonstrates that proteins share the same subcellular space, but does not specifically detect p53 aggregates. Overall, the PLA and the p53-Seprion-ELISA are the only two methods that allow the quantitative measurement of p53 aggregates. On the one hand, the PLA represents the ideal method for p53 aggregate detection in FFPE tissue, which is the gold-standard preservation method of clinical samples. On the other hand, when fresh-frozen tissue is available the p53-Seprion-ELISA should be preferred because of the shorter turnaround time and the possibility for high-throughput analysis. These methods may add to the understanding of amyloid-like p53 in cancer and could help stratify patients in future clinical trials targeting p53 aggregation.
- Published
- 2022
- Full Text
- View/download PDF
4. Clinical impact of EZH2 and its antagonist SMARCA4 in ovarian cancer
- Author
-
Katharina Leitner, Irina Tsibulak, Verena Wieser, Katharina Knoll, Daniel Reimer, Christian Marth, Heidi Fiegl, and Alain G. Zeimet
- Subjects
Medicine ,Science - Abstract
Abstract SMARCA4 and EZH2 are two functional key players of their respective antagonizing chromatin remodeling complexes SWI/SNF and PRC2. EZH2 inhibitory drugs may abrogate pro-oncogenic features of PRC2 and turn the balance to cell differentiation via SWI/SNF activity in cancers. SMARCA4 and EZH2 expression was assessed by RT-PCR in 238 epithelial ovarian cancers (OCs) and put in relation to clinico-pathological parameters and patients’ outcome. Optimal thresholds for high and low expression of both variables were calculated by the Youden’s index based on receiver operating characteristic (ROC) curves. High SMARCA4 mRNA expression was independently associated with favorable progression-free survival (PFS) (P = 0.03) and overall survival (OS) (P = 0.018). As Youden’s threshold determination for EZH2 yielded a S-shaped ROC-curve, two cut-off points (29th and 94th percentile) predicting opposite features were defined. Whereas EZH2 mRNA levels beyond the 29th percentile independently predicted poor PFS (P = 0.034), Cox-regression in EZH2 transcripts above the 94th percentile revealed a conversion from unfavorable to favorable PFS and OS (P = 0.009 and P = 0.032, respectively). High SMARCA4 expression associates with improved survival, whereas moderate/high EZH2 expression predicts poor outcome, which converts to favorable survival in ultra-high expressing OCs. This small OC subgroup could be characterized by REV7-abrogated platinum hypersensitivity but concomitant PARP-inhibitor resistance.
- Published
- 2020
- Full Text
- View/download PDF
5. Methylome analysis of extreme chemoresponsive patients identifies novel markers of platinum sensitivity in high-grade serous ovarian cancer
- Author
-
Tushar Tomar, Nicolette G. Alkema, Leroy Schreuder, Gert Jan Meersma, Tim de Meyer, Wim van Criekinge, Harry G. Klip, Heidi Fiegl, Els van Nieuwenhuysen, Ignace Vergote, Martin Widschwendter, Ed Schuuring, Ate G. J. van der Zee, Steven de Jong, and G. Bea A. Wisman
- Subjects
DNA methylation ,Integrated methylome analysis ,Ovarian cancer ,Platinum-based chemotherapy ,Extreme chemoresponders ,Medicine - Abstract
Abstract Background Despite an early response to platinum-based chemotherapy in advanced stage high-grade serous ovarian cancer (HGSOC), the majority of patients will relapse with drug-resistant disease. Aberrant epigenetic alterations like DNA methylation are common in HGSOC. Differences in DNA methylation are associated with chemoresponse in these patients. The objective of this study was to identify and validate novel epigenetic markers of chemoresponse using genome-wide analysis of DNA methylation in extreme chemoresponsive HGSOC patients. Methods Genome-wide next-generation sequencing was performed on methylation-enriched tumor DNA of two HGSOC patient groups with residual disease, extreme responders (≥18 months progression-free survival (PFS), n = 8) and non-responders (≤6 months PFS, n = 10) to platinum-based chemotherapy. DNA methylation and expression data of the same patients were integrated to create a gene list. Genes were validated on an independent cohort of extreme responders (n = 21) and non-responders (n = 31) using pyrosequencing and qRT-PCR. In silico validation was performed using publicly available DNA methylation (n = 91) and expression (n = 208) datasets of unselected advanced stage HGSOC patients. Functional validation of FZD10 on chemosensitivity was carried out in ovarian cancer cell lines using siRNA-mediated silencing. Results Integrated genome-wide methylome and expression analysis identified 45 significantly differentially methylated and expressed genes between two chemoresponse groups. Four genes FZD10, FAM83A, MYO18B, and MKX were successfully validated in an external set of extreme chemoresponsive HGSOC patients. High FZD10 and MKX methylation were related with extreme responders and high FAM83A and MYO18B methylation with non-responders. In publicly available advanced stage HGSOC datasets, FZD10 and MKX methylation levels were associated with PFS. High FZD10 methylation was strongly associated with improved PFS in univariate analysis (hazard ratio (HR) = 0.43; 95% CI, 0.27–0.71; P = 0.001) and multivariate analysis (HR = 0.39; 95% CI, 0.23–0.65; P = 0.003). Consistently, low FZD10 expression was associated with improved PFS (HR = 1.36; 95% CI, 0.99–1.88; P = 0.058). FZD10 silencing caused significant sensitization towards cisplatin treatment in survival assays and apoptosis assays. Conclusions By applying genome-wide integrated methylome analysis on extreme chemoresponsive HGSOC patients, we identified novel clinically relevant, epigenetically-regulated markers of platinum-sensitivity in HGSOC patients. The clinical potential of these markers in predictive and therapeutic approaches has to be further validated in prospective studies.
- Published
- 2017
- Full Text
- View/download PDF
6. Correction to: Epigenetic regulation of L1CAM in endometrial carcinoma: comparison to cancer–testis (CT-X) antigens
- Author
-
Uwe Schirmer, Heidi Fiegl, Marco Pfeifer, Alain G. Zeimet, Elisabeth Müller-Holzner, Peter K. Bode, Verena Tischler, and Peter Altevogt
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Following publication of the original article [1], we have been alerted to errors in Figs. 2 and 8. In Fig. 2B, the GAPDH loading control for Hec1A cells is shown twice in error (in Fig. 2B and Fig. 2C). In Fig. 8, in testis case 1 (first column) the MAGE-A4 staining panel was repeated and also appears as the NY-ESO-1 staining panel in error. The corrected versions of Fig. 2 and Fig. 8 are shown below. We apologize for this inconvenience.
- Published
- 2018
- Full Text
- View/download PDF
7. Glucocorticoid Receptor-Deficient Foxp3+ Regulatory T Cells Fail to Control Experimental Inflammatory Bowel Disease
- Author
-
Lourdes Rocamora-Reverte, Selma Tuzlak, Laura von Raffay, Marcel Tisch, Heidi Fiegl, Mathias Drach, Holger M. Reichardt, Andreas Villunger, Denise Tischner, and G. Jan Wiegers
- Subjects
glucocorticoid ,glucocorticoid receptor ,Foxp3 ,regulatory T cell ,transfer colitis ,suppression ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Activation of the immune system increases systemic adrenal-derived glucocorticoid (GC) levels which downregulate the immune response as part of a negative feedback loop. While CD4+ T cells are essential target cells affected by GC, it is not known whether these hormones exert their major effects on CD4+ helper T cells, CD4+Foxp3+ regulatory T cells (Treg cells), or both. Here, we generated mice with a specific deletion of the glucocorticoid receptor (GR) in Foxp3+ Treg cells. Remarkably, while basal Treg cell characteristics and in vitro suppression capacity were unchanged, Treg cells lacking the GR did not prevent the induction of inflammatory bowel disease in an in vivo mouse model. Under inflammatory conditions, GR-deficient Treg cells acquired Th1-like characteristics and expressed IFN-gamma, but not IL-17, and failed to inhibit pro-inflammatory CD4+ T cell expansion in situ. These findings reveal that the GR is critical for Foxp3+ Treg cell function and suggest that endogenous GC prevent Treg cell plasticity toward a Th1-like Treg cell phenotype in experimental colitis. When equally active in humans, a rationale is provided to develop GC-mimicking therapeutic strategies which specifically target Foxp3+ Treg cells for the treatment of inflammatory bowel disease.
- Published
- 2019
- Full Text
- View/download PDF
8. NADPH oxidase 4 expression in the normal endometrium and in endometrial cancer
- Author
-
Christine Degasper, Andrea Brunner, Natalie Sampson, Irina Tsibulak, Verena Wieser, Hannah Welponer, Christian Marth, Heidi Fiegl, and Alain Gustave Zeimet
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The aim of this study was to explore the role of NOX4 in the biology of the normal endometrium and endometrial cancer. NOX4 plays a key role in other adenocarcinomas and has been implicated in the pathogenesis of diabetes and obesity, which are important risk factors for endometrial cancer. NOX4 expression was assessed in 239 endometrial cancer and 25 normal endometrium samples by quantitative real-time polymerase chain reaction, in situ hybridization, and immunohistochemistry. DNA methylation of the NOX4 promoter was determined by means of MethyLight PCR. Data were correlated with clinicopathological parameters and analyzed in the context of diabetes and body mass index. In the normal endometrium, NOX4 microRNA expression was significantly higher in the secretory transformed compared with proliferative endometrium ( p = 0.008). In endometrial cancer specimens, NOX4 expression did not differ between diabetic and non-diabetic patients, but was the highest in patients with a body mass index ≤ 26 ( p = 0.037). The lowest NOX4 expression was found in carcinosarcomas ( p = 0.007). High NOX4 expression predicted poorer clinical outcome with regard to overall survival, especially in non-diabetic patients and those with a body mass index > 20. Independent prognostic significance of NOX4 transcripts was retained in type I endometrial cancer and was the most meaningful in patients with a body mass index > 20. No prognostic impact was shown for NOX4 promoter methylation in endometrial cancer. For the first time, we demonstrate that NOX4 plays a considerable role in the cycle-dependent changes in the normal endometrium and in the biology of endometrial cancer.
- Published
- 2019
- Full Text
- View/download PDF
9. Role of DNA methylation and epigenetic silencing of HAND2 in endometrial cancer development.
- Author
-
Allison Jones, Andrew E Teschendorff, Quanxi Li, Jane D Hayward, Athilakshmi Kannan, Tim Mould, James West, Michal Zikan, David Cibula, Heidi Fiegl, Shih-Han Lee, Elisabeth Wik, Richard Hadwin, Rupali Arora, Charlotte Lemech, Henna Turunen, Päivi Pakarinen, Ian J Jacobs, Helga B Salvesen, Milan K Bagchi, Indrani C Bagchi, and Martin Widschwendter
- Subjects
Medicine - Abstract
BackgroundEndometrial cancer incidence is continuing to rise in the wake of the current ageing and obesity epidemics. Much of the risk for endometrial cancer development is influenced by the environment and lifestyle. Accumulating evidence suggests that the epigenome serves as the interface between the genome and the environment and that hypermethylation of stem cell polycomb group target genes is an epigenetic hallmark of cancer. The objective of this study was to determine the functional role of epigenetic factors in endometrial cancer development.Methods and findingsEpigenome-wide methylation analysis of >27,000 CpG sites in endometrial cancer tissue samples (n = 64) and control samples (n = 23) revealed that HAND2 (a gene encoding a transcription factor expressed in the endometrial stroma) is one of the most commonly hypermethylated and silenced genes in endometrial cancer. A novel integrative epigenome-transcriptome-interactome analysis further revealed that HAND2 is the hub of the most highly ranked differential methylation hotspot in endometrial cancer. These findings were validated using candidate gene methylation analysis in multiple clinical sample sets of tissue samples from a total of 272 additional women. Increased HAND2 methylation was a feature of premalignant endometrial lesions and was seen to parallel a decrease in RNA and protein levels. Furthermore, women with high endometrial HAND2 methylation in their premalignant lesions were less likely to respond to progesterone treatment. HAND2 methylation analysis of endometrial secretions collected using high vaginal swabs taken from women with postmenopausal bleeding specifically identified those patients with early stage endometrial cancer with both high sensitivity and high specificity (receiver operating characteristics area under the curve = 0.91 for stage 1A and 0.97 for higher than stage 1A). Finally, mice harbouring a Hand2 knock-out specifically in their endometrium were shown to develop precancerous endometrial lesions with increasing age, and these lesions also demonstrated a lack of PTEN expression.ConclusionsHAND2 methylation is a common and crucial molecular alteration in endometrial cancer that could potentially be employed as a biomarker for early detection of endometrial cancer and as a predictor of treatment response. The true clinical utility of HAND2 DNA methylation, however, requires further validation in prospective studies. Please see later in the article for the Editors' Summary.
- Published
- 2013
- Full Text
- View/download PDF
10. Correction: The Dynamics and Prognostic Potential of DNA Methylation Changes at Stem Cell Gene Loci in Women's Cancer.
- Author
-
Joanna Zhuang, Allison Jones, Shih-Han Lee, Esther Ng, Heidi Fiegl, Michal Zikan, David Cibula, Alexandra Sargent, Helga B. Salvesen, Ian J. Jacobs, Henry C. Kitchener, Andrew E. Teschendorff, and Martin Widschwendter
- Subjects
Genetics ,QH426-470 - Published
- 2012
- Full Text
- View/download PDF
11. The dynamics and prognostic potential of DNA methylation changes at stem cell gene loci in women's cancer.
- Author
-
Joanna Zhuang, Allison Jones, Shih-Han Lee, Esther Ng, Heidi Fiegl, Michal Zikan, David Cibula, Alexandra Sargent, Helga B Salvesen, Ian J Jacobs, Henry C Kitchener, Andrew E Teschendorff, and Martin Widschwendter
- Subjects
Genetics ,QH426-470 - Abstract
Aberrant DNA methylation is an important cancer hallmark, yet the dynamics of DNA methylation changes in human carcinogenesis remain largely unexplored. Moreover, the role of DNA methylation for prediction of clinical outcome is still uncertain and confined to specific cancers. Here we perform the most comprehensive study of DNA methylation changes throughout human carcinogenesis, analysing 27,578 CpGs in each of 1,475 samples, ranging from normal cells in advance of non-invasive neoplastic transformation to non-invasive and invasive cancers and metastatic tissue. We demonstrate that hypermethylation at stem cell PolyComb Group Target genes (PCGTs) occurs in cytologically normal cells three years in advance of the first morphological neoplastic changes, while hypomethylation occurs preferentially at CpGs which are heavily Methylated in Embryonic Stem Cells (MESCs) and increases significantly with cancer invasion in both the epithelial and stromal tumour compartments. In contrast to PCGT hypermethylation, MESC hypomethylation progresses significantly from primary to metastatic cancer and defines a poor prognostic signature in four different gynaecological cancers. Finally, we associate expression of TET enzymes, which are involved in active DNA demethylation, to MESC hypomethylation in cancer. These findings have major implications for cancer and embryonic stem cell biology and establish the importance of systemic DNA hypomethylation for predicting prognosis in a wide range of different cancers.
- Published
- 2012
- Full Text
- View/download PDF
12. Tamoxifen-induced epigenetic silencing of oestrogen-regulated genes in anti-hormone resistant breast cancer.
- Author
-
Andrew Stone, Fatima Valdés-Mora, Julia M W Gee, Lynne Farrow, Richard A McClelland, Heidi Fiegl, Carol Dutkowski, Rachael A McCloy, Robert L Sutherland, Elizabeth A Musgrove, and Robert I Nicholson
- Subjects
Medicine ,Science - Abstract
In the present study, we have taken the novel approach of using an in vitro model representative of tamoxifen-withdrawal subsequent to clinical relapse to achieve a greater understanding of the mechanisms that serve to maintain the resistant-cell phenotype, independent of any agonistic impact of tamoxifen, to identify potential novel therapeutic approaches for this disease state. Following tamoxifen withdrawal, tamoxifen-resistant MCF-7 cells conserved both drug resistance and an increased basal rate of proliferation in an oestrogen deprived environment, despite reduced epidermal growth-factor receptor expression and reduced sensitivity to gefitinib challenge. Although tamoxifen-withdrawn cells retained ER expression, a sub-set of ER-responsive genes, including pS2 and progesterone receptor (PgR), were down-regulated by promoter DNA methylation, as confirmed by clonal bisulphite sequencing experiments. Following promoter demethylation with 5-Azacytidine (5-Aza), the co-addition of oestradiol (E2) restored gene expression in these cells. In addition, 5-Aza/E2 co-treatment induced a significant anti-proliferative effect in the tamoxifen-withdrawn cells, in-contrast to either agent used alone. Microarray analysis was undertaken to identify genes specifically up regulated by this co-treatment. Several anti-proliferative gene candidates were identified and their promoters were confirmed as more heavily methylated in the tamoxifen resistant vs sensitive cells. One such gene candidate, growth differentiation factor 15 (GDF15), was carried forward for functional analysis. The addition of 5-Aza/E2 was sufficient to de-methylate and activate GDF15 expression in the tamoxifen resistant cell-lines, whilst in parallel, treatment with recombinant GDF15 protein decreased cell survival. These data provide evidence to support a novel concept that long-term tamoxifen exposure induces epigenetic silencing of a cohort of oestrogen-responsive genes whose function is associated with negative proliferation control. Furthermore, reactivation of such genes using epigenetic drugs could provide a potential therapeutic avenue for the management of tamoxifen-resistant breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
13. Epigenotyping in peripheral blood cell DNA and breast cancer risk: a proof of principle study.
- Author
-
Martin Widschwendter, Sophia Apostolidou, Elke Raum, Dietrich Rothenbacher, Heidi Fiegl, Usha Menon, Christa Stegmaier, Ian J Jacobs, and Hermann Brenner
- Subjects
Medicine ,Science - Abstract
BackgroundEpigenetic changes are emerging as one of the most important events in carcinogenesis. Two alterations in the pattern of DNA methylation in breast cancer (BC) have been previously reported; active estrogen receptor-alpha (ER-alpha) is associated with decreased methylation of ER-alpha target (ERT) genes, and polycomb group target (PCGT) genes are more likely than other genes to have promoter DNA hypermethylation in cancer. However, whether DNA methylation in normal unrelated cells is associated with BC risk and whether these imprints can be related to factors which can be modified by the environment, is unclear.Methodology/principal findingsUsing quantitative methylation analysis in a case-control study (n = 1,083) we found that DNA methylation of peripheral blood cell DNA provides good prediction of BC risk. We also report that invasive ductal and invasive lobular BC is characterized by two different sets of genes, the latter particular by genes involved in the differentiation of the mesenchyme (PITX2, TITF1, GDNF and MYOD1). Finally we demonstrate that only ERT genes predict ER positive BC; lack of peripheral blood cell DNA methylation of ZNF217 predicted BC independent of age and family history (odds ratio 1.49; 95% confidence interval 1.12-1.97; P = 0.006) and was associated with ER-alpha bioactivity in the corresponding serum.Conclusion/significanceThis first large-scale epigenotyping study demonstrates that DNA methylation may serve as a link between the environment and the genome. Factors that can be modulated by the environment (like estrogens) leave an imprint in the DNA of cells that are unrelated to the target organ and indicate the predisposition to develop a cancer. Further research will need to demonstrate whether DNA methylation profiles will be able to serve as a new tool to predict the risk of developing chronic diseases with sufficient accuracy to guide preventive measures.
- Published
- 2008
- Full Text
- View/download PDF
14. Data from DNA Methylation in Serum and Tumors of Cervical Cancer Patients
- Author
-
Martin Widschwendter, Christian Marth, Georg Goebel, Elisabeth Müller-Holzner, Annemarie Wiedemair, Lennart Ivarsson, Heidi Fiegl, Hannes M. Müller, and Andreas Widschwendter
- Abstract
Purpose: Promoter hypermethylation has been recognized to play an important role in carcinogenesis. Numerous studies have demonstrated tumor-specific alterations, such as aberrant promoter hypermethylation, in DNA recovered from plasma or serum of patients with various malignancies. The aim of this study was to investigate the methylation status of various genes in cervical cancer patients and their association with clinicopathological characteristics and outcome of the disease.Experimental Design: The methylation status of CALCA, hTERT, MYOD1, PGR (progesterone receptor), and TIMP3 was investigated in serum samples from 93 cervical cancer patients and 19 corresponding tissue samples using the MethyLight technique.Results: Aberrant promoter hypermethylation was detected in any of these genes in 87% (81 of 93) of the serum samples studied. Methylation of MYOD1 was detected more frequently in advanced stage. All of the genes found to be methylated in serum samples were also methylated in the corresponding tissue sample, except in one patient. Patients with unmethylated MYOD1 serum DNA had significantly better disease-free (P = 0.04) and overall survival (P = 0.02) in comparison with patients with methylated MYOD1.Conclusions: To the best of our knowledge, this is, thus far, the largest study investigating aberrant promoter hypermethylation in serum samples from cancer patients and the first study investigating methylation patterns in sera of cervical cancer patients. Our results suggest that serological detection of MYOD1 promoter hypermethylation may be of potential use as a prognostic marker for discriminating cervical cancer patients at high risk for lymph node metastasis or relapse. Additional studies, including a panel of additional genes, are necessary to elucidate the role of aberrant methylation in serum as a tool for surveillance of cervical cancer.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure from DNA Methylation in Serum and Tumors of Cervical Cancer Patients
- Author
-
Martin Widschwendter, Christian Marth, Georg Goebel, Elisabeth Müller-Holzner, Annemarie Wiedemair, Lennart Ivarsson, Heidi Fiegl, Hannes M. Müller, and Andreas Widschwendter
- Abstract
Supplementary Figure from DNA Methylation in Serum and Tumors of Cervical Cancer Patients
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 1 from FBXW7/hCDC4 Is a General Tumor Suppressor in Human Cancer
- Author
-
Charles Spruck, Olle Sangfelt, Martin Widschwendter, Steven I. Reed, Dan Grander, Hans Nordgren, Per Sangfelt, Fredrik Petersson, Susanne Egyhazi, Johan Hansson, Mohammad Reza Zali, Babak Noori Nayer, Sepideh Zabihi Nejad, Markus Dagnell, Martin Corcoran, Elisabeth Mueller-Holzner, Christian Marth, Dimitra Dofou, Heidi Fiegl, Diana Cepeda, Alena Maljukova, Kathleen Klotz, Sophia Apostolidou, Natalie von der Lehr, Dahui Sun, and Shahab Akhoondi
- Abstract
Supplementary Table 1 from FBXW7/hCDC4 Is a General Tumor Suppressor in Human Cancer
- Published
- 2023
- Full Text
- View/download PDF
17. ESR1 Upstream from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
ESR1 Upstream from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
We have generated DNA methylation profiles of 148 human breast tumors and found significant differences in hormone receptor (HR) status between clusters of DNA methylation profiles. Of 35 DNA methylation markers analyzed, the ESR1 gene, encoding estrogen receptor α, proved to be the best predictor of progesterone receptor status, whereas methylation of the PGR gene, encoding progesterone receptor, was the best predictor of estrogen receptor status. ESR1 methylation outperformed HR status as a predictor of clinical response in patients treated with the antiestrogen tamoxifen, whereas promoter methylation of the CYP1B1 gene, encoding a tamoxifen- and estradiol-metabolizing cytochrome P450, predicted response differentially in tamoxifen-treated and nontamoxifen-treated patients. High levels of promoter methylation of the ARHI gene, encoding a RAS-related small G-protein, were strongly predictive of good survival in patients who had not received tamoxifen therapy. Our results reveal an as yet unrecognized degree of interaction between DNA methylation and HR biology in breast cancer cells and suggest potentially clinically useful novel DNA methylation predictors of response to hormonal and non-hormonal breast cancer therapy.
- Published
- 2023
- Full Text
- View/download PDF
19. Clinical Tamoxifen Data from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
Clinical Tamoxifen Data from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Published
- 2023
- Full Text
- View/download PDF
20. RT-PCR Primers from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
RT-PCR Primers from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Published
- 2023
- Full Text
- View/download PDF
21. Data from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Author
-
Alain G. Zeimet, Peter Altevogt, Karl Illmensee, Christian Marth, Elisabeth Müller-Holzner, Heidi Fiegl, Nicole Concin, Martin Erdel, Sergej Skvortsov, Svenja Riedle, Michael Hubalek, and Daniel Reimer
- Abstract
We describe for the first time a new integral molecular pathway, linking transcription factor E2F3a to epidermal growth factor receptor (EGFR) activation in ovarian cancer cells. Investigations on the role of E2F family members in EGFR-mediated mitogenic signaling revealed that E2F3a was selectively upregulated following EGFR activation, whereas all other E2F family members remained unaffected. In contrast, EGF treatment of healthy ovarian surface epithelial and mesothelial cells yielded a selective upregulation of proliferation-promoting E2F1 and E2F2 without influencing E2F3a expression. In ovarian cancer cell lines, the extent of EGF-induced proliferative stimulus was closely related to the magnitude of E2F3a increase, and proliferation inhibition by E2F3a knockdown was not overcome by EGF exposure. Furthermore, the EGFR-E2F3a axis was found to be signal transducer and activator of transcription 1/3 dependent and the ratio of IFN-regulatory factor (IRF)-1 to IRF-2 was shown to be determinative for E2F3a control. In a pilot study on 32 primary ovarian cancer specimens, a highly significant correlation between activated EGFR and E2F3a expression was disclosed. This new integral pathway in the EGFR-driven mitogenic cell response, which through its key player E2F3a was found to be essential in triggering proliferation in ovarian cancer cells, provides new insights into EGFR signaling and could represent the basis for appealing new therapeutic approaches in ovarian cancer. Cancer Res; 70(11); 4613–23. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Data from FBXW7/hCDC4 Is a General Tumor Suppressor in Human Cancer
- Author
-
Charles Spruck, Olle Sangfelt, Martin Widschwendter, Steven I. Reed, Dan Grander, Hans Nordgren, Per Sangfelt, Fredrik Petersson, Susanne Egyhazi, Johan Hansson, Mohammad Reza Zali, Babak Noori Nayer, Sepideh Zabihi Nejad, Markus Dagnell, Martin Corcoran, Elisabeth Mueller-Holzner, Christian Marth, Dimitra Dofou, Heidi Fiegl, Diana Cepeda, Alena Maljukova, Kathleen Klotz, Sophia Apostolidou, Natalie von der Lehr, Dahui Sun, and Shahab Akhoondi
- Abstract
The ubiquitin-proteasome system is a major regulatory pathway of protein degradation and plays an important role in cellular division. Fbxw7 (or hCdc4), a member of the F-box family of proteins, which are substrate recognition components of the multisubunit ubiquitin ligase SCF (Skp1-Cdc53/Cullin-F-box-protein), has been shown to mediate the ubiquitin-dependent proteolysis of several oncoproteins including cyclin E1, c-Myc, c-Jun, and Notch. The oncogenic potential of Fbxw7 substrates, frequent allelic loss in human cancers, and demonstration that mutation of FBXW7 cooperates with p53 in mouse tumorigenesis have suggested that Fbxw7 could function as a tumor suppressor in human cancer. Here, we carry out an extensive genetic screen of primary tumors to evaluate the role of FBXW7 as a tumor suppressor in human tumorigenesis. Our results indicate that FBXW7 is inactivated by mutation in diverse human cancer types with an overall mutation frequency of ∼6%. The highest mutation frequencies were found in tumors of the bile duct (cholangiocarcinomas, 35%), blood (T-cell acute lymphocytic leukemia, 31%), endometrium (9%), colon (9%), and stomach (6%). Approximately 43% of all mutations occur at two mutational “hotspots,” which alter Arg residues (Arg465 and Arg479) that are critical for substrate recognition. Furthermore, we show that Fbxw7Arg465 hotspot mutant can abrogate wild-type Fbxw7 function through a dominant negative mechanism. Our study is the first comprehensive screen of FBXW7 mutations in various human malignancies and shows that FBXW7 is a general tumor suppressor in human cancer. [Cancer Res 2007;67(19):9006–12]
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 1 from DNA Hypomethylation and Ovarian Cancer Biology
- Author
-
Melanie Ehrlich, Peter W. Laird, Alain G. Zeimet, Elisabeth Müller-Holzner, Christian Marth, Georg Goebel, Heidi Fiegl, Hannes M. Müller, Christian Woods, Guanchao Jiang, and Martin Widschwendter
- Abstract
Supplementary Figure 1 from DNA Hypomethylation and Ovarian Cancer Biology
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure 2 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Author
-
Alain G. Zeimet, Peter Altevogt, Karl Illmensee, Christian Marth, Elisabeth Müller-Holzner, Heidi Fiegl, Nicole Concin, Martin Erdel, Sergej Skvortsov, Svenja Riedle, Michael Hubalek, and Daniel Reimer
- Abstract
Supplementary Figure 2 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Published
- 2023
- Full Text
- View/download PDF
25. MethyLight Primer and Probe Sequences from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
MethyLight Primer and Probe Sequences from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Published
- 2023
- Full Text
- View/download PDF
26. Data from Circulating Tumor-Specific DNA: A Marker for Monitoring Efficacy of Adjuvant Therapy in Cancer Patients
- Author
-
Martin Widschwendter, Georg Goebel, Helmut Klocker, Andreas Berger, Christian Ensinger, Christian Marth, Elisabeth Mueller-Holzner, Simone Millinger, and Heidi Fiegl
- Abstract
Adjuvant systemic therapy (a strategy that targets potential disseminated tumor cells after complete removal of the tumor) has clearly improved survival of patients with cancer. To date, no tool is available to monitor efficacy of these therapies, unless distant metastases arise, a situation that unavoidably leads to death. We analyzed RASSF1A DNA methylation in pretherapeutic sera and serum samples collected 1 year after surgery from 148 patients with breast cancer who were receiving adjuvant tamoxifen; 19.6% and 22.3% of patients with breast cancer showed RASSF1A DNA methylation in their pretherapeutic and 1-year-after serum samples, respectively. RASSF1A methylation 1 year after primary surgery (and during adjuvant tamoxifen therapy) was an independent predictor of poor outcome, with a relative risk (95% confidence interval) for relapse of 5.1 (1.3-19.8) and for death of 6.9 (1.9-25.9). Measurement of serum DNA methylation allows adjuvant systemic treatment to be monitored for efficacy: disappearance of RASSF1A DNA methylation in serum throughout treatment with tamoxifen indicates a response, whereas persistence or new appearance means resistance to adjuvant tamoxifen treatment. It remains to be seen whether modifications made in adjuvant therapeutic strategies based on detection of circulating nucleic acids will improve survival as well as quality of life.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Tables 1-3 from Circulating Tumor-Specific DNA: A Marker for Monitoring Efficacy of Adjuvant Therapy in Cancer Patients
- Author
-
Martin Widschwendter, Georg Goebel, Helmut Klocker, Andreas Berger, Christian Ensinger, Christian Marth, Elisabeth Mueller-Holzner, Simone Millinger, and Heidi Fiegl
- Abstract
Supplementary Tables 1-3 from Circulating Tumor-Specific DNA: A Marker for Monitoring Efficacy of Adjuvant Therapy in Cancer Patients
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Table 1 from DNA Hypomethylation and Ovarian Cancer Biology
- Author
-
Melanie Ehrlich, Peter W. Laird, Alain G. Zeimet, Elisabeth Müller-Holzner, Christian Marth, Georg Goebel, Heidi Fiegl, Hannes M. Müller, Christian Woods, Guanchao Jiang, and Martin Widschwendter
- Abstract
Supplementary Table 1 from DNA Hypomethylation and Ovarian Cancer Biology
- Published
- 2023
- Full Text
- View/download PDF
29. Data from DNA Hypomethylation and Ovarian Cancer Biology
- Author
-
Melanie Ehrlich, Peter W. Laird, Alain G. Zeimet, Elisabeth Müller-Holzner, Christian Marth, Georg Goebel, Heidi Fiegl, Hannes M. Müller, Christian Woods, Guanchao Jiang, and Martin Widschwendter
- Abstract
Hypomethylation of some portions of the genome and hypermethylation of others are very frequent in human cancer. The hypomethylation often involves satellite 2 (Sat2) DNA in the juxtacentromeric (centromere-adjacent) region of chromosome 1. In this study, we analyzed methylation in centromeric and juxtacentromeric satellite DNA in 115 ovarian cancers, 26 non-neoplastic ovarian specimens, and various normal somatic tissue standards. We found that hypomethylation of both types of satellite DNA in ovarian samples increased significantly from non-neoplastic toward cancer tissue. Furthermore, strong hypomethylation was significantly more prevalent in tumors of advanced stage or high grade. Importantly, extensive hypomethylation of Sat2 DNA in chromosome 1 was a highly significant marker of poor prognosis (relative risk for relapse, 4.1, and death, 9.4) and more informative than tumor grade or stage. Also, comparing methylation of satellite DNA and 15 5′ gene regions, which are often hypermethylated in cancer or implicated in ovarian carcinogenesis, we generally found no positive or negative association between methylation changes in satellite DNA and in the gene regions. However, hypermethylation at two loci, CDH13 (at 16q24) and RNR1 (at 13p12), was correlated strongly with lower levels of Sat2 hypomethylation. The CDH13/Sat2 epigenetic correlation was seen also in breast cancers. We conclude that satellite DNA hypomethylation is an important issue in ovarian carcinogenesis as demonstrated by: (a) an increase from non-neoplastic tissue toward ovarian cancer; (b) an increase within the ovarian cancer group toward advanced grade and stage; and (c) the finding that strong hypomethylation was an independent marker of poor prognosis.
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Breast Cancer DNA Methylation Profiles in Cancer Cells and Tumor Stroma: Association with HER-2/neu Status in Primary Breast Cancer
- Author
-
Martin Widschwendter, Peter W. Laird, Christian Marth, Elisabeth Müller-Holzner, Georg Goebel, Simone Millinger, and Heidi Fiegl
- Abstract
The HER-2/neu gene is amplified and overexpressed in 20% to 30% of invasive breast carcinomas and is associated with increased metastatic potential and less tamoxifen sensitivity. We generated the DNA methylation profiles of 143 human breast tumors and found significant differences in HER-2/neu expression and DNA methylation of five genes. For three of these five genes [PGR (coding for the progesterone receptor), HSD17B4 (coding for type 4 17-β-hydroxysteroid dehydrogenase, an enzyme that mainly degrades active 17-β-estradiol into inactive metabolites), and CDH13 (coding for H-cadherin)] a higher prevalence of DNA methylation in HER-2/neu-positive cancers was confirmed in an independent set of microdissected primary breast cancers. DNA methylation was not only present in cancer cells but also in the tumor stroma fraction. Of the isolated fractions in HER-2/neu-positive versus -negative cancers, 27.1% versus 10.5%, respectively, showed DNA methylation of the five genes (P = 0.011, Fisher's exact test). In Her-2++/+++ breast cancers, HSD17B4 mRNA expression was inversely associated with HSD17B4 methylation (P = 0.04). These data support the view that in addition to HER-2/neu-associated signaling, epigenetic changes in cancer as well as in tumor stroma cells might attribute to the specific biological features of HER-2/neu-positive cancers. (Cancer Res 2006; 66(1): 29-33)
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Figure 3 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Author
-
Alain G. Zeimet, Peter Altevogt, Karl Illmensee, Christian Marth, Elisabeth Müller-Holzner, Heidi Fiegl, Nicole Concin, Martin Erdel, Sergej Skvortsov, Svenja Riedle, Michael Hubalek, and Daniel Reimer
- Abstract
Supplementary Figure 3 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figure 1 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Author
-
Alain G. Zeimet, Peter Altevogt, Karl Illmensee, Christian Marth, Elisabeth Müller-Holzner, Heidi Fiegl, Nicole Concin, Martin Erdel, Sergej Skvortsov, Svenja Riedle, Michael Hubalek, and Daniel Reimer
- Abstract
Supplementary Figure 1 from E2F3a Is Critically Involved in Epidermal Growth Factor Receptor–Directed Proliferation in Ovarian Cancer
- Published
- 2023
- Full Text
- View/download PDF
33. PMR Summary from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Author
-
Peter W. Laird, Peter A. Jones, Elisabeth Müller-Holzner, Christian Marth, Heidi Fiegl, Hannes M. Müller, Kimberly D. Siegmund, and Martin Widschwendter
- Abstract
PMR Summary from Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Table 1 from Breast Cancer DNA Methylation Profiles in Cancer Cells and Tumor Stroma: Association with HER-2/neu Status in Primary Breast Cancer
- Author
-
Martin Widschwendter, Peter W. Laird, Christian Marth, Elisabeth Müller-Holzner, Georg Goebel, Simone Millinger, and Heidi Fiegl
- Abstract
Supplementary Table 1 from Breast Cancer DNA Methylation Profiles in Cancer Cells and Tumor Stroma: Association with HER-2/neu Status in Primary Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
35. Influence of Pregnancy-Related Conditions on Human Epididymis Protein 4 Serum Levels in Comparison to CA 125 – a Prospective Cohort Trial
- Author
-
Samira Abdel Azim, Verena Wieser, Heidi Fiegl, A Berger, Bettina Böttcher, Andreas Widschwendter, Irene Mutz-Dehbalaie, and Christian Marth
- Subjects
Percentile ,medicine.medical_specialty ,HE4 ,Tumormarker ,Adnexal mass ,Maternity and Midwifery ,medicine ,Rupture of membranes ,GebFra Science ,Prospective cohort study ,Tumor marker ,Pregnancy ,Predictive marker ,Obstetrics ,business.industry ,adnexal mass ,Adnextumor ,Obstetrics and Gynecology ,medicine.disease ,CA 125 ,Schwangerschaft ,tumor marker ,biomarker ,Biomarker (medicine) ,Original Article ,pregnancy ,business - Abstract
Introduction HE4 and CA 125, two established biomarkers for assessing adnexal masses in non-pregnant women, are hardly investigated in pregnancy, especially in pregnancy-associated conditions. The aim was to evaluate HE4 and CA 125 levels in the course of pregnancy and to assess the impact of pregnancy disorders, contractions and rupture of membranes on HE4 and CA 125 serum levels in order to use these parameters for evaluation of adnexal masses in pregnancy.Patients and Methods Blood samples (n = 238) of 201 women seen at the Medical University of Innsbruck, Austria, were prospectively obtained during pregnancy and postpartum. Serum concentrations of HE4 and CA 125 were analyzed. ROMA index was calculated by the premenopausal formula.Results HE4 serum levels were highest in the third trimester. Contractions (p Conclusions HE4 serum levels are influenced by several pregnancy-related conditions leading to significantly higher levels in these cases. Despite differing medians according to trimester, the 95th percentile cut-offs and almost all maximum values during the entire course of pregnancy were below the established cut-off for premenopausal women. It was also superior to the performance of ROMA index. Therefore, HE4 can be used as a valuable negative predictive marker for the assessment of adnexal masses during pregnancy.
- Published
- 2021
- Full Text
- View/download PDF
36. A retrospective validation of CanAssist Breast in European early-stage breast cancer patient cohort
- Author
-
Aparna Gunda, Chetana Basavaraj, Chandra Prakash Serkad V, Manjula Adinarayan, Ramu Kolli, Mallikarjuna Siraganahalli Eshwaraiah, Cristina Saura, Fiorella Ruiz, Patricia Gomez, Vicente Peg, Jose Jimenez, Susanne Sprung, Heidi Fiegl, Christine Brunner, Daniel Egle, GS Bhattacharyya, Manjiri M Bakre, Institut Català de la Salut, [Gunda A, Basavaraj C, Serkad V CP, Adinarayan M, Kolli R, Siraganahalli Eshwaraiah M] OncoStem Diagnostics, Bengaluru, Karnataka, India. [Saura C, Ruiz F, Gomez P, Peg V, Jimenez J] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Mama - Càncer - Prognosi ,neoplasias::neoplasias por localización::neoplasias de la mama [ENFERMEDADES] ,Receptor, ErbB-2 ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,Otros calificadores::/diagnóstico [Otros calificadores] ,Breast Neoplasms ,General Medicine ,Prognosis ,Diagnòstic immunohistoquímic ,Artificial Intelligence ,Other subheadings::/diagnosis [Other subheadings] ,Biomarkers, Tumor ,Humans ,Surgery ,Female ,Breast ,Neoplasm Recurrence, Local ,Retrospective Studies - Abstract
Hormone-receptor positive; Chemotherapy; Early-stage breast cancer Receptor de hormonas positivo; Quimioterapia; Cáncer de mama en fase inicial Receptor d'hormones positiu; Quimioteràpia; Càncer de mama en fase inicial CanAssist Breast (CAB), a prognostic test uses immunohistochemistry (IHC) approach coupled with artificial intelligence-based machine learning algorithm for prognosis of early-stage hormone-receptor positive, HER2/neu negative breast cancer patients. It was developed and validated in an Indian cohort. Here we report the first blinded validation of CAB in a multi-country European patient cohort. FFPE tumor samples from 864 patients were obtained from-Spain, Italy, Austria, and Germany. IHC was performed on these samples, followed by recurrence risk score prediction. The outcomes were obtained from medical records. The performance of CAB was analyzed by hazard ratios (HR) and Kaplan Meier curves. CAB stratified European cohort (n = 864) into distinct low- and high-risk groups for recurrence (P < 0.0001) with HR of 3.32 (1.85–5.93) like that of mixed (India, USA, and Europe) (n = 1974), 3.43 (2.34–4.93) and Indian cohort (n = 925), 3.09 (1.83–5.21). CAB provided significant prognostic information (P < 0.0001) in women aged ≤ 50 (HR: 4.42 (1.58–12.3), P < 0.0001) and >50 years (HR: 2.93 (1.44–5.96), P = 0.0002). CAB had an HR of 2.57 (1.26–5.26), P = 0.01) in women with N1 disease. CAB stratified significantly higher proportions (77%) as low-risk over IHC4 (55%) (P < 0.0001). Additionally, 82% of IHC4 intermediate-risk patients were stratified as low-risk by CAB. Accurate risk stratification of European patients by CAB coupled with its similar performance inIndian patients shows that CAB is robust and functions independent of ethnic differences. CAB can potentially prevent overtreatment in a greater number of patients compared to IHC4 demonstrating its usefulness for adjuvant systemic therapy planning in European breast cancer patients.
- Published
- 2021
37. High RIG‐I expression in ovarian cancer associates with an immune‐escape signature and poor clinical outcome
- Author
-
Verena Wieser, Heidi Fiegl, Alain G. Zeimet, Susanne Sprung, Dominik Wolf, Sieghart Sopper, Daniel Reimer, Christian Marth, Maximilian Boesch, and Gunther Hartmann
- Subjects
Adult ,Cancer Research ,viruses ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Tumor Immunology and Microenvironment ,Kaplan-Meier Estimate ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,PD-L1 ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Humans ,Receptors, Immunologic ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Aged, 80 and over ,Ovarian Neoplasms ,Tumor microenvironment ,biology ,business.industry ,virus diseases ,FOXP3 ,Immunotherapy ,Middle Aged ,biochemical phenomena, metabolism, and nutrition ,Prognosis ,medicine.disease ,Primary tumor ,Oncology ,Case-Control Studies ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,DEAD Box Protein 58 ,Biomarker (medicine) ,Female ,Tumor Escape ,Neoplasm Grading ,biological phenomena, cell phenomena, and immunity ,business ,Ovarian cancer - Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy, with platinum‐based chemotherapy remaining the mainstay for adjuvant treatment after surgery. The lack of indication for immunotherapy may at least in part result from the lack of suitable biomarkers allowing stratification of potentially responding patients. In this monocentric study of 141 cases with OC, we used real‐time quantitative PCR to assess the expression of retinoic acid‐inducible gene‐I (RIG‐I) in primary tumor and healthy ovarian control tissues. RIG‐I expression was correlated to various clinicopathological characteristics as well as to a set of molecular and immunological markers. The prognostic significance of RIG‐I expression was queried in univariate and multivariate analyses and validated in an independent cohort. RIG‐I was overexpressed in the cancerous ovary and correlated with a higher tumor grade. The more aggressive Type‐II cancers and cancers with inactivating p53 mutations exhibited higher RIG‐I expression. RIG‐I levels were also elevated in cancers that recurred after remission or were platinum‐refractory. Survival analyses disclosed RIG‐I as an independent marker of poor outcome in OC. Continuative analyses revealed the molecular and immunological correlates of RIG‐I expression in the tumor microenvironment, including interferon production and a distinct immune‐regulatory signature involving checkpoint molecules (PD‐L1/PD‐1), the RNA‐editing enzyme ADAR1 and the regulatory T cell‐specific transcription factor FoxP3. We conclude that high RIG‐I expression associates with poor outcome in OC, which is explainable by local immunosuppression in the tumor bed. RIG‐I expression may inform checkpoint blockade and/or RIG‐I agonistic targeting in a subset of high‐risk OC patients., What's new? Previous evidence suggests that the antiviral helicase RIG‐I bears tumor‐suppressive activity. The clinical significance of RIG‐I in gynecological cancer remains unclear, however. This single‐center, retrospective, explorative biomarker study of 141 cases with epithelial ovarian cancer (OC) reveals the negative prognostic impact of RIG‐I. RIG‐I is overexpressed in OC tissue, particularly in the more aggressive type‐II OCs, and is an independent prognostic marker for overall survival. RIG‐I high‐expressing tumors carry an interferon signature, linking RIG‐I activation to a cancer microenvironment fostering tumor immune‐evasion including upregulation of immune checkpoints. RIG‐I expression may indicate high‐risk OC patients who may benefit from immunotherapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
38. Clinical impact of EZH2 and its antagonist SMARCA4 in ovarian cancer
- Author
-
I Tsibulak, Christian Marth, K Leitner, Heidi Fiegl, Daniel Reimer, K Knoll, Verena Wieser, and Alain G. Zeimet
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,Percentile ,Science ,Chromatin remodelling ,macromolecular substances ,Carcinoma, Ovarian Epithelial ,Chromatin remodeling ,Article ,Ovarian cancer ,Internal medicine ,Medicine ,Humans ,Enhancer of Zeste Homolog 2 Protein ,RNA, Messenger ,Fallopian Tubes ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Ovarian Neoplasms ,Multidisciplinary ,Receiver operating characteristic ,business.industry ,EZH2 ,Ovary ,Antagonist ,DNA Helicases ,Nuclear Proteins ,Middle Aged ,medicine.disease ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,ROC Curve ,Concomitant ,Case-Control Studies ,Mad2 Proteins ,SMARCA4 ,Female ,Neoplasm Grading ,Poly(ADP-ribose) Polymerases ,business ,Signal Transduction ,Transcription Factors - Abstract
SMARCA4 and EZH2 are two functional key players of their respective antagonizing chromatin remodeling complexes SWI/SNF and PRC2. EZH2 inhibitory drugs may abrogate pro-oncogenic features of PRC2 and turn the balance to cell differentiation via SWI/SNF activity in cancers. SMARCA4 and EZH2 expression was assessed by RT-PCR in 238 epithelial ovarian cancers (OCs) and put in relation to clinico-pathological parameters and patients’ outcome. Optimal thresholds for high and low expression of both variables were calculated by the Youden’s index based on receiver operating characteristic (ROC) curves. High SMARCA4 mRNA expression was independently associated with favorable progression-free survival (PFS) (P = 0.03) and overall survival (OS) (P = 0.018). As Youden’s threshold determination for EZH2 yielded a S-shaped ROC-curve, two cut-off points (29th and 94th percentile) predicting opposite features were defined. Whereas EZH2 mRNA levels beyond the 29th percentile independently predicted poor PFS (P = 0.034), Cox-regression in EZH2 transcripts above the 94th percentile revealed a conversion from unfavorable to favorable PFS and OS (P = 0.009 and P = 0.032, respectively). High SMARCA4 expression associates with improved survival, whereas moderate/high EZH2 expression predicts poor outcome, which converts to favorable survival in ultra-high expressing OCs. This small OC subgroup could be characterized by REV7-abrogated platinum hypersensitivity but concomitant PARP-inhibitor resistance.
- Published
- 2020
39. Integrated analysis of cervical squamous cell carcinoma cohorts from three continents reveals conserved subtypes of prognostic significance
- Author
-
Ankur Chakravarthy, Ian Reddin, Stephen Henderson, Cindy Dong, Nerissa Kirkwood, Maxmilan Jeyakumar, Daniela Rothschild Rodriguez, Natalia Gonzalez Martinez, Jacqueline McDermott, Xiaoping Su, Nagayasau Egawa, Christina S Fjeldbo, Vilde Eide Skingen, Mari Kyllesø Halle, Camilla Krakstad, Afschin Soleiman, Susanne Sprung, Peter Ellis, Mark Wass, Martin Michaelis, Heidi Lyng, Heidi Fiegl, Helga Salvesen, Gareth Thomas, John Doorbar, Kerry Chester, Andrew Feber, and Tim R Fenton
- Subjects
Cervical cancer ,0303 health sciences ,business.industry ,medicine.medical_treatment ,Disease ,medicine.disease ,3. Good health ,Transcriptome ,Causes of cancer ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunoediting ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,business ,Adjuvant ,030304 developmental biology ,Epigenomics - Abstract
Human papillomavirus (HPV)-associated cervical cancer represents one of the leading causes of cancer death worldwide. Although low-middle income countries are disproportionately affected, our knowledge of the disease predominantly originates from populations in high-income countries. Using the largest multi-omic analysis of cervical squamous cell carcinoma (CSCC) to date, totalling 643 tumours and representing patient populations from the USA, Europe and Sub-Saharan Africa, we identify two CSCC subtypes (C1 and C2) with differing prognosis. C1 tumours are largely HPV16-driven, display increased cytotoxic T-lymphocyte infiltration and frequently harbour PIK3CA and EP300 mutations. C2 tumours are associated with shorter overall survival, are frequently driven by HPVs from the HPV18-containing alpha-7 clade, harbour alterations in the Hippo signalling pathway and increased expression of immune checkpoint genes, B7-H3 (also known as CD276) and NT5E (also known as CD73) and PD-L2 (also known as PDCD1LG2). In conclusion, we identify two novel, therapy-relevant CSCC subtypes that share the same defining characteristics across three geographically diverse cohorts.
- Published
- 2020
- Full Text
- View/download PDF
40. Evaluation of Vav3.1 as prognostic marker in endometrial cancer
- Author
-
Heidi Fiegl, Annemarie Wiedemair, Julia Rössler, Jiri Hatina, Dominik Wolf, Sieghart Sopper, Alain G. Zeimet, Daniel Reimer, Christian Marth, and Maximilian Boesch
- Subjects
Adult ,0301 basic medicine ,Oncology ,VAV3 ,Cancer Research ,medicine.medical_specialty ,Original Article – Clinical Oncology ,Endometrium ,Protein isoform ,03 medical and health sciences ,0302 clinical medicine ,Endometrial cancer ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Protein Isoforms ,Stage (cooking) ,Proto-Oncogene Proteins c-vav ,Cancer ,Aged ,Retrospective Studies ,Aged, 80 and over ,Hematology ,Transcript variant ,business.industry ,General Medicine ,Vav3 ,Middle Aged ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Cystadenocarcinoma, Serous ,Endometrial Neoplasms ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,030220 oncology & carcinogenesis ,Female ,Guanine nucleotide exchange factor ,business ,Ovarian cancer ,Adenocarcinoma, Clear Cell ,Follow-Up Studies - Abstract
Purpose Vav3 is a guanine nucleotide exchange factor that regulates the activity of Rho/Rac family GTPases. In a study on ovarian cancer, we recently demonstrated pronounced prognostic and predictive value of Vav3.1, a specific truncation variant of the parental Vav3 gene. Here, we sought to investigate the role of Vav3.1 in the most prevalent gynecological tumor entity, endometrial cancer. Methods Vav3.1 transcript levels were determined in a large cohort of endometrial cancer patients using variant-specific PCR (n = 239), and non-malignant endometrial tissue served as control (n = 26). Expression levels of Vav3.1 were stratified according to established clinicopathological characteristics and correlated to long-term patient survival (average follow-up of > 7.5 years). Type 1 and type 2 cancers were separately investigated. Results While Vav3.1 was markedly overexpressed in endometrial cancer tissue, we could not detect associations with clinical parameters related to prognosis, such as FIGO stage and tumor grade. Kaplan–Meier estimators of different measures of survival failed to show prognostic significance of Vav3.1 in endometrial cancer. Lack of prognostic value was observed for both type 1 and type 2 cancers. Conclusions Our study shows that Vav3.1 is not suited as a marker of cancer progression and/or treatment response in endometrial cancer. Feasibility and potential benefit of targeting Vav3.1 in endometrial cancer needs to be evaluated in future studies, proceeding from its clear, roughly ten-fold, induction in the malignant endometrium.
- Published
- 2018
- Full Text
- View/download PDF
41. Development of a novel prognostic score for breast cancer patients using mRNA expression of CHAC1
- Author
-
Marjan Arvandi, Beate Jahn, Georg Goebel, Christian Marth, Uwe Siebert, Ursula Rochau, and Heidi Fiegl
- Subjects
Adult ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Multivariate statistics ,Receptor, ErbB-2 ,Breast Neoplasms ,HER2/Neu Status ,Malignancy ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Breast cancer ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Lymph node ,Aged ,Aged, 80 and over ,Proportional hazards model ,business.industry ,Health Policy ,Middle Aged ,Prognosis ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Estrogen ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,business ,gamma-Glutamylcyclotransferase - Abstract
Aim: To develop a prognostic score for primary breast cancer patients integrating conventional predictors and the novel biomarker CHAC1 to aid adjuvant chemotherapy decisions. Patients & methods: A prognostic score for overall survival was developed using: conventional predictors from a dataset of 1777 patients and the weight of CHAC1 mRNA expression from an independent dataset of 106 patients using multivariate Cox regression. Results: The new score includes: CHAC1 mRNA expression, age, tumor size, HER2 neu status, lymph node status and degree of malignancy. Using a cut-off value of 11 score points, 10-year survival was 82% in low-risk (n = 34) and 43% in high-risk patients (n = 72). The addition of CHAC1 resulted in 16% reclassification. Conclusion: Including CHAC1 in prognostic prediction may aid (and change) personalized treatment selection.
- Published
- 2017
- Full Text
- View/download PDF
42. Methylome analysis of extreme chemoresponsive patients identifies novel markers of platinum sensitivity in high-grade serous ovarian cancer
- Author
-
Tim De Meyer, G. Bea A. Wisman, Steven de Jong, Ignace Vergote, Harry G. Klip, Tushar Tomar, Heidi Fiegl, Gert Jan Meersma, Ate G.J. van der Zee, Els Van Nieuwenhuysen, Wim Van Criekinge, Ed Schuuring, Leroy Schreuder, Nicolette G. Alkema, Martin Widschwendter, Damage and Repair in Cancer Development and Cancer Treatment (DARE), Targeted Gynaecologic Oncology (TARGON), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,lcsh:Medicine ,Platinum Compounds ,BETA-CATENIN ,Epigenesis, Genetic ,0302 clinical medicine ,FZD10 ,Medicine and Health Sciences ,Prospective Studies ,EPIGENETIC REGULATION ,Prospective cohort study ,Platinum-based chemotherapy ,Ovarian Neoplasms ,Univariate analysis ,DNA methylation ,Hazard ratio ,DNA, Neoplasm ,General Medicine ,Methylation ,Middle Aged ,030220 oncology & carcinogenesis ,Integrated methylome analysis ,Female ,SIGNALING PATHWAY ,Research Article ,medicine.drug ,HUMAN SYNOVIAL SARCOMA ,BIOMARKERS ,Antineoplastic Agents ,Disease-Free Survival ,03 medical and health sciences ,CPG ISLAND METHYLATION ,Ovarian cancer ,Biomarkers, Tumor ,medicine ,Humans ,Epigenetics ,DNA METHYLATION CHANGES ,Aged ,Retrospective Studies ,Cisplatin ,business.industry ,lcsh:R ,Biology and Life Sciences ,Sequence Analysis, DNA ,medicine.disease ,GENE ,Extreme chemoresponders ,030104 developmental biology ,Cancer research ,Neoplasm Recurrence, Local ,business ,EXPRESSION ANALYSIS - Abstract
Background Despite an early response to platinum-based chemotherapy in advanced stage high-grade serous ovarian cancer (HGSOC), the majority of patients will relapse with drug-resistant disease. Aberrant epigenetic alterations like DNA methylation are common in HGSOC. Differences in DNA methylation are associated with chemoresponse in these patients. The objective of this study was to identify and validate novel epigenetic markers of chemoresponse using genome-wide analysis of DNA methylation in extreme chemoresponsive HGSOC patients. Methods Genome-wide next-generation sequencing was performed on methylation-enriched tumor DNA of two HGSOC patient groups with residual disease, extreme responders (≥18 months progression-free survival (PFS), n = 8) and non-responders (≤6 months PFS, n = 10) to platinum-based chemotherapy. DNA methylation and expression data of the same patients were integrated to create a gene list. Genes were validated on an independent cohort of extreme responders (n = 21) and non-responders (n = 31) using pyrosequencing and qRT-PCR. In silico validation was performed using publicly available DNA methylation (n = 91) and expression (n = 208) datasets of unselected advanced stage HGSOC patients. Functional validation of FZD10 on chemosensitivity was carried out in ovarian cancer cell lines using siRNA-mediated silencing. Results Integrated genome-wide methylome and expression analysis identified 45 significantly differentially methylated and expressed genes between two chemoresponse groups. Four genes FZD10, FAM83A, MYO18B, and MKX were successfully validated in an external set of extreme chemoresponsive HGSOC patients. High FZD10 and MKX methylation were related with extreme responders and high FAM83A and MYO18B methylation with non-responders. In publicly available advanced stage HGSOC datasets, FZD10 and MKX methylation levels were associated with PFS. High FZD10 methylation was strongly associated with improved PFS in univariate analysis (hazard ratio (HR) = 0.43; 95% CI, 0.27–0.71; P = 0.001) and multivariate analysis (HR = 0.39; 95% CI, 0.23–0.65; P = 0.003). Consistently, low FZD10 expression was associated with improved PFS (HR = 1.36; 95% CI, 0.99–1.88; P = 0.058). FZD10 silencing caused significant sensitization towards cisplatin treatment in survival assays and apoptosis assays. Conclusions By applying genome-wide integrated methylome analysis on extreme chemoresponsive HGSOC patients, we identified novel clinically relevant, epigenetically-regulated markers of platinum-sensitivity in HGSOC patients. The clinical potential of these markers in predictive and therapeutic approaches has to be further validated in prospective studies. Electronic supplementary material The online version of this article (doi:10.1186/s12916-017-0870-0) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
43. Correction to: Epigenetic regulation of L1CAM in endometrial carcinoma: comparison to cancer–testis (CT-X) antigens
- Author
-
Alain G. Zeimet, Peter Altevogt, Heidi Fiegl, Marco Pfeifer, Verena Tischler, Elisabeth Müller-Holzner, Uwe Schirmer, Peter K. Bode, University of Zurich, and Altevogt, Peter
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,L1 ,610 Medicine & health ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,1311 Genetics ,10049 Institute of Pathology and Molecular Pathology ,Genetics ,Carcinoma ,medicine ,1306 Cancer Research ,Epigenetics ,Testis cancer ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Staining ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,2730 Oncology ,business - Abstract
Following publication of the original article [1], we have been alerted to errors in Figs. 2 and 8. In Fig. 2B, the GAPDH loading control for Hec1A cells is shown twice in error (in Fig. 2B and Fig. 2C). In Fig. 8, in testis case 1 (first column) the MAGE-A4 staining panel was repeated and also appears as the NY-ESO-1 staining panel in error. The corrected versions of Fig. 2 and Fig. 8 are shown below. We apologize for this inconvenience.
- Published
- 2018
- Full Text
- View/download PDF
44. Evaluating L1CAM expression in human endometrial cancer using qRT-PCR
- Author
-
Daniel Reimer, Heidi Fiegl, Annamarie Wiedermair, Christian Marth, Sara Notaro, Julia Rössler, Peter Altevogt, Michaela Duggan-Peer, and Alain G. Zeimet
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neural Cell Adhesion Molecule L1 ,Real-Time Polymerase Chain Reaction ,Endometrium ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Carcinoma ,Humans ,Promoter Regions, Genetic ,Aged ,Gynecology ,business.industry ,Endometrial cancer ,Case-control study ,Reproducibility of Results ,Cancer ,qRT-PCR ,DNA Methylation ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Endometrial Neoplasms ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Real-time polymerase chain reaction ,Oncology ,Case-Control Studies ,L1CAM ,030220 oncology & carcinogenesis ,endometrial cancer ,DNA methylation ,outcome ,Female ,methylation ,Skin cancer ,business ,Follow-Up Studies ,Research Paper - Abstract
// Sara Notaro 1, 2 , Daniel Reimer 1 , Michaela Duggan-Peer 1 , Heidi Fiegl 1 , Annamarie Wiedermair 1 , Julia Rossler 1 , Peter Altevogt 3, 4 , Christian Marth 1 , Alain Gustave Zeimet 1 1 Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria 2 Department of Gynecology and Obstetrics, University of Brescia, Brescia, Italy 3 Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany 4 Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany Correspondence to: Alain Gustave Zeimet, email: alain.zeimet@uki.at Keywords: L1CAM, endometrial cancer, qRT-PCR, outcome, methylation Received: January 04, 2016 Accepted: May 08, 2016 Published: May 24, 2016 ABSTRACT Background: Management of endometrial carcinoma (EC) still needs improvement of risk assessment. Recently, L1CAM immunohistochemical (IHC) evaluation showed a unique value to predict the outcome of early EC. However IHC results are often conflicting for lack of inter-laboratory standardisation. Methods: Here, as a proof of concept and to increase reproducibility we assayed eighty-two EC and 26 normal endometrium samples for L1CAM expression (L1CAM EXP ) via qRT-PCR. The IHC evaluation was performed in 50 cancer samples. Moreover, we aimed to substantiate the in-vitro findings of L1CAM regulation through its promoter methylation (L1CAM MET ), miR-34a expression and miR-34a promoter methylation. DNA methylation was assessed with MethyLight PCR technique. Results : High overall concordant results between IHC and RT-PCR evaluations were found. L1CAM EXP was detected in 11% of cancer specimens. These positive cancers exhibited a worse DFS (p=0.032) and OS (p=0.016) in a multivariate COX-regression model. L1CAM EXP predicted distant failure (p=0.007) and L1CAM MET predicted risk-reduction of lymph-node involvement (p=0.005). Inverse correlations between L1CAM EXP and L1CAM MET (p=0.004) and between L1CAM EXP and miR-34a expression (p=0.002) were found. Conclusions: In conclusion qRT-PCR analysis is a reliable approach to evaluate L1CAM status in EC and L1CAM EXP was highly predictive for distant failure and poor outcome, confirming the large IHC-based studies. Interestingly, L1CAM MET was able to assess the risk of pelvic lymph-node involvement. Especially the latter finding has to be confirmed in larger prospective series.
- Published
- 2016
- Full Text
- View/download PDF
45. Clinical impact of L1CAM expression measured on the transcriptome level in ovarian cancer
- Author
-
Michaela Duggan-Peer, Daniel Reimer, Samira Abdel Azim, Christian Marth, Afschin Soleiman, Heidi Fiegl, Susanne Sprung, and Alain G. Zeimet
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,RT-PCR ,Neural Cell Adhesion Molecule L1 ,Kaplan-Meier Estimate ,Real-Time Polymerase Chain Reaction ,survival ,Disease-Free Survival ,Targeted therapy ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Obstetrics and gynaecology ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Survival analysis ,Neoplasm Staging ,Retrospective Studies ,Ovarian Neoplasms ,Gynecology ,business.industry ,Cancer ,Middle Aged ,Prognosis ,targeted therapy ,medicine.disease ,Immunohistochemistry ,ovarian cancer ,030104 developmental biology ,Real-time polymerase chain reaction ,Case-Control Studies ,L1CAM ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Neoplasm Grading ,business ,Ovarian cancer ,Research Paper - Abstract
// Samira Abdel Azim 1 , Michaela Duggan-Peer 1 , Susanne Sprung 1 , Daniel Reimer 1 , Heidi Fiegl 1, 2 , Afschin Soleiman 1 , Christian Marth 1 , Alain G. Zeimet 1 1 Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria 2 Department of Obstetrics and Gynecology, Laboratory for Clinical Biochemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria Correspondence to: Alain G. Zeimet, email: alain.zeimet@tirol-kliniken.at , alain.zeimet@i-med.ac.at Keywords: ovarian cancer, L1CAM, targeted therapy, RT-PCR, survival Received: October 21, 2015 Accepted: April 18, 2016 Published: May 11, 2016 ABSTRACT Background: High expression of L1 cell adhesion molecules (L1CAM) has been repeatedly shown to be associated with aggressive disease behavior, which translates in poor clinical outcome in various cancer entities. However, in ovarian cancer results based either on immunohistochemistry or cytosolic protein quantifications remained conflicting regarding clinical behavior. In the present work we assessed L1CAM expression on the transcriptome level with the highly sensitive quantitative real-time PCR (qRT-PCR) to define its relevance in ovarian cancer biology. Results: There was a significant difference in L1CAM high and low mRNA expressing cancers with regard to disease-free (p=0.002) and overall survival (p=0.008). L1CAM proofed to be an independent predictor for disease progression (HR 1.8, p=0.01) and overall survival (HR 1.6, p=0.04). Furthermore, a significant positive correlation between the level of L1CAM and the grade of tumor differentiation (p=0.04), the FIGO stage (p=0.025) as well as the histological subtype (p= 0.002) was found. Methods: This study included fresh frozen tissue samples of 138 patients with FIGO I-IV stage ovarian cancer. L1CAM mRNA expression was determined using qRT-PCR. In the calculations special attention was put on the various histological subtypes. In survival analysis median L1CAM mRNA expression obtained in the entire cohort of ovarian cancer samples was used as a cut-off to distinguish between high and low L1CAM mRNA expression. Conclusion: L1CAM mRNA expression appears to play a substantial role in the pathophysiology of ovarian cancer that is translated into poor clinical outcome. Additionally humanized L1CAM antibodies, which can serve as potential future treatment options are under testing.
- Published
- 2016
- Full Text
- View/download PDF
46. Inborn-like errors of metabolism are determinants of breast cancer risk, clinical response and survival: a study of human biochemical individuality
- Author
-
Sandra Regina Morini da Silva, Carlos Veo, R.B. Tomioka, Celso R Silva, Irmgard Himmel, Edson Guimaraes Loturco, Gustavo Arantes Rosa Maciel, José Salvador Rodrigues de Oliveira, Iara Baldim Rabelo, Heidi Fiegl, Maria Izabel Chiamolera, André Lopes Carvalho, Patricia Eiko Yamakawa, Robert A. Nagourney, Carolina Stella, Ricardo Sobhie Diaz, Celso Francisco Hernandes Granato, Antônio Augusto Ferreira Carioca, Cristovam Scapulatempo Neto, Dirce Maria Lobo Marchioni, Clovis A. Silva, Christina Troi, Paulo D'Amora, Rene da Costa Vieira, Delcio Matos, Bruno Scarpellini, Ismael Dale Cotrim Guerreiro da Silva, Rui M. B. Maciel, Rosa Paula M. Biscolla, Renato Fraietta, Marcelo A. Mori, Felipe C.G. Reis, Marcia Batista Salzgeber, Edmund Chada Baracat, and Daniel Egle
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Early detection ,Hematologic Neoplasms ,survival ,Biochemical phenotype ,Malignant transformation ,03 medical and health sciences ,Breast cancer ,breast cancer ,Internal medicine ,Medicine ,Breast carcinogenesis ,skin and connective tissue diseases ,Chemotherapy ,Squamous cell cancer ,response ,business.industry ,medicine.disease ,030104 developmental biology ,prognosis ,business ,metabolism ,Research Paper - Abstract
Breast cancer remains a leading cause of morbidity and mortality worldwide yet methods for early detection remain elusive. We describe the discovery and validation of biochemical signatures measured by mass spectrometry, performed upon blood samples from patients and controls that accurately identify (>95%) the presence of clinical breast cancer. Targeted quantitative MS/MS conducted upon 1225 individuals, including patients with breast and other cancers, normal controls as well as individuals with a variety of metabolic disorders provide a biochemical phenotype that accurately identifies the presence of breast cancer and predicts response and survival following the administration of neoadjuvant chemotherapy. The metabolic changes identified are consistent with inborn-like errors of metabolism and define a continuum from normal controls to elevated risk to invasive breast cancer. Similar results were observed in other adenocarcinomas but were not found in squamous cell cancers or hematologic neoplasms. The findings describe a new early detection platform for breast cancer and support a role for pre-existing, inborn-like errors of metabolism in the process of breast carcinogenesis that may also extend to other glandular malignancies. Statement of Significance: Findings provide a powerful tool for early detection and the assessment of prognosis in breast cancer and define a novel concept of breast carcinogenesis that characterizes malignant transformation as the clinical manifestation of underlying metabolic insufficiencies.
- Published
- 2018
47. Detection of soluble EpCAM (sEpCAM) in malignant ascites predicts poor overall survival in patients treated with catumaxomab
- Author
-
Jalid Sehouli, Guenther Gastl, Dominic Fong, Laura Botta, Andreas Seeber, Gilbert Spizzo, Alain G. Zeimet, Mani Nassir, Gerold Untergasser, Heidi Fiegl, and Ioana Braicu
- Subjects
Male ,medicine.medical_specialty ,Catumaxomab ,Enzyme-Linked Immunosorbent Assay ,Kaplan-Meier Estimate ,Gastroenterology ,chemistry.chemical_compound ,Antigens, Neoplasm ,Predictive Value of Tests ,Internal medicine ,Antibodies, Bispecific ,Ascites ,Biomarkers, Tumor ,medicine ,Humans ,Prospective cohort study ,soluble EpCAM ,Retrospective Studies ,Ovarian Neoplasms ,Predictive marker ,business.industry ,Cancer ,Epithelial cell adhesion molecule ,Middle Aged ,Epithelial Cell Adhesion Molecule ,Prognosis ,medicine.disease ,Surgery ,ovarian cancer ,Treatment Outcome ,Solubility ,Oncology ,chemistry ,EpCAM ,Predictive value of tests ,catumaxomab ,Female ,medicine.symptom ,business ,Ovarian cancer ,Cell Adhesion Molecules ,Research Paper ,Follow-Up Studies ,medicine.drug - Abstract
EpCAM is an attractive target for cancer therapy and the EpCAM-specific antibody catumaxomab has been used for intraperitoneal treatment of EpCAM-positive cancer patients with malignant ascites. New prognostic markers are necessary to select patients that mostly benefit from catumaxomab. Recent data showed that soluble EpCAM (sEpCAM) is capable to block the effect of catumaxomab in vitro. This exploratory retrospective analysis was performed on archived ascites samples to evaluate the predictive role of sEpCAM in catumaxomab-treated patients. Sixty-six catumaxomab-treated patients with an available archived ascites sample were included in this study and tested for sEpCAM by sandwich ELISA. All probes were sampled before treatment start and all patients received at least one catumaxomab infusion. Overall survival, puncture-free survival and time to next puncture were compared between sEpCAM-positive and -negative patients. We detected sEpCAM in ascites samples of 9 patients (13.6%). These patients showed a significantly shorter overall survival. The prognostic significance of sEpCAM in ascites was particularly strong in patients with ovarian cancer. Puncture-free survival and time to next puncture were not significantly different between sEpCAM-positive and -negative patients. We propose sEpCAM in malignant ascites as a potential predictive marker in cancer patients treated with catumaxomab. Prospective studies with larger patients samples are urgently needed to confirm these findings and studies testing dose-intensified catumaxomab in patients with sEpCAM-positive ascites should be envisaged.
- Published
- 2015
- Full Text
- View/download PDF
48. Epigenetics in the perioperative period
- Author
-
Markus W. Hollmann, Nina C. Weber, Philipp Lirk, and Heidi Fiegl
- Subjects
Pharmacology ,Perioperative medicine ,business.industry ,Chronic pain ,Perioperative ,medicine.disease ,Opioid ,Hyperalgesia ,DNA methylation ,Medicine ,Epigenetics ,medicine.symptom ,business ,Neuroscience ,medicine.drug ,Epigenesis - Abstract
The perioperative period is characterized by profound changes in the body's homoeostatic processes. This review seeks to address whether epigenetic mechanisms may influence an individual's reaction to surgery and anaesthesia. Evidence from animal and human studies suggests that epigenetic mechanisms can explain many facets of susceptibility to acute and chronic pain, making them potential therapeutic targets. Modern pain management is still based upon opiates, and both the developmental expression of opioid receptors and opioid-induced hyperalgesia have been linked to epigenetic mechanisms. In general, opiates seem to increase global DNA methylation levels. This is in contrast to local anaesthetics, which have been ascribed a global demethylating effect. Even though no direct investigations have been carried out, the potential influence of epigenetics on the inflammatory response that follows surgery seems a promising area for research. There is a considerable body of evidence that supports the involvement of epigenetics in the complex process of wound healing. Epigenetics is an important emerging research topic in perioperative medicine, with a huge potential to positively influence patient outcome.
- Published
- 2015
- Full Text
- View/download PDF
49. High prevalence of side population in human cancer cell lines
- Author
-
Julia M. Huber, Maximilian Boesch, Helmut Klocker, Guenther Gastl, Dominik Wolf, Sieghart Sopper, Alain G. Zeimet, Barbara Wolf, and Heidi Fiegl
- Subjects
0301 basic medicine ,Genetics ,Cancer Research ,drug resistance ,High prevalence ,Brief Report ,Biology ,Drug transporter ,Phenotype ,drug transporter ,stem cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Side population ,Cell culture ,030220 oncology & carcinogenesis ,side population ,Cancer cell lines ,Stem cell ,cancer cell line ,Human cancer - Abstract
Cancer cell lines are essential platforms for performing cancer research on human cells. We here demonstrate that, across tumor entities, human cancer cell lines harbor minority populations of putative stem-like cells, molecularly defined by dye extrusion resulting in the side population phenotype. These findings establish a heterogeneous nature of human cancer cell lines and argue for their stem cell origin. This should be considered when interpreting research involving these model systems.
- Published
- 2016
- Full Text
- View/download PDF
50. FOXO3-mediated chemo-protection in high-stage neuroblastoma depends on wild-type TP53 and SESN3
- Author
-
Judith Hagenbuchner, Ursula Kiechl-Kohlendorfer, Martina Rupp, Michael J. Ausserlechner, Bettina Rass, Heidi Fiegl, and Petra Obexer
- Subjects
0301 basic medicine ,Cancer Research ,Cell type ,Programmed cell death ,Apoptosis ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Neuroblastoma ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,neoplasms ,Heat-Shock Proteins ,Neoplasm Staging ,Cell Nucleus ,Mutation ,Bcl-2-Like Protein 11 ,Forkhead Box Protein O3 ,Wild type ,Cell cycle ,3. Good health ,DNA-Binding Proteins ,030104 developmental biology ,BCL2L11 ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,FOXO3 ,Original Article ,Tumor Suppressor Protein p53 ,Protein Binding - Abstract
Forkhead box O class transcription factors are homeostasis regulators that control cell death, longevity and therapy-resistance. In neuroblastoma (NB), nuclear FOXO3 correlates with stage M disease and poor prognosis. To analyze whether FOXO3 contributes to drug-resistance in this childhood cancer, we investigated how different high-stage-derived NB cells respond to the activation of an ectopic FOXO3 allele. We found endogenous FOXO3 mostly localized to the nucleus-upon activation of an ectopic, 4OHT-activated FOXO3(A3)ER fusion protein two of the cell lines underwent apoptosis, whereas in the others FOXO3-activation even increased survival during drug-treatment. In the latter cell type, FOXO3 did not induce the BH3-only protein BCL2L11/BIM due to impaired binding of FOXO3 to the BIM-promoter, but still activated other FOXO3 targets. It was shown before that FOXO3 and TP53 physically interact with each other at two different regions-the TP53-N-terminus binds to the FOXO3-DNA binding domain (DBD) and the FOXO3-C-terminus interacts with the TP53-DBD. Interestingly, cell lines that undergo FOXO3-induced cell death carry homozygous point mutations in the TP53-DBD near the structural hotspot-mutation-site R175H, which abrogated FOXO3-TP53 interaction. In contrast, in FOXO3-death-resistant cells no point mutations in the TP53-DBD were found-in these cells FOXO3-TP53 complexes are formed and FOXO3-binding to the BIM-promoter, but not the induction of the detoxifying protein SESN3, were prevented, which in turn increased chemo-protection in this type of high-stage-derived NB cells. Our combined data suggest that FOXO3 steps in as a death inducer in case of TP53-mutation, whereas functional TP53 alters FOXO3-target-promoter-recognition, which prevents death induction by FOXO3 and instead increases chemo-protection and survival of NB cells. This novel mechanism may explain the low incidence of TP53 mutation in high-stage NB at diagnosis and suggests FOXO3 as a therapeutic target for this childhood malignancy.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.