Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sabine Maier is active.

Publication


Featured researches published by Sabine Maier.


Journal of Cellular Biochemistry | 2003

Oligonucleotide‐based microarray for DNA methylation analysis: Principles and applications

Huidong Shi; Sabine Maier; Inko Nimmrich; Pearlly S. Yan; Charles W. Caldwell; Alexander Olek; Tim Hui Ming Huang

Gene silencing via promoter CpG island hypermethylation offers tumor cells growth advantages. This epigenetic event is pharmacologically reversible, and uncovering a unique set of methylation‐silenced genes in tumor cells can bring a new avenue to cancer treatment. However, high‐throughput tools capable of surveying the methylation status of multiple gene promoters are needed for this discovery process. Herein we describe an oligonucleotide‐based microarray technique that is both versatile and sensitive in revealing hypermethylation in defined regions of the genome. DNA samples are bisulfite‐treated and PCR‐amplified to distinguish CpG dinucleotides that are methylated from those that are not. Fluorescently labeled PCR products are hybridized to arrayed oligonucleotides that can discriminate between methylated and unmethylated alleles in regions of interest. Using this technique, two clinical subtypes of non‐Hodgkins lymphomas, mantle cell lymphoma, and grades I/II follicular lymphoma, were further separated based on the differential methylation profiles of several gene promoters. Work is underway in our laboratory to extend the interrogation power of this microarray system in multiple candidate genes. This novel tool, therefore, holds promise to monitor the outcome of various epigenetic therapies on cancer patients.


Cancer Research | 2005

Association of DNA Methylation of Phosphoserine Aminotransferase with Response to Endocrine Therapy in Patients with Recurrent Breast Cancer

John W.M. Martens; Inko Nimmrich; Thomas Koenig; Maxime P. Look; Nadia Harbeck; Fabian Model; Antje Kluth; Joan Bolt-de Vries; Anieta M. Sieuwerts; Henk Portengen; Marion E. Meijer-van Gelder; Christian Piepenbrock; Alexander Olek; Heinz Höfler; Marion Kiechle; J.G.M. Klijn; Manfred Schmitt; Sabine Maier; John A. Foekens

To understand the biological basis of resistance to endocrine therapy is of utmost importance in patients with steroid hormone receptor-positive breast cancer. Not only will this allow us prediction of therapy success, it may also lead to novel therapies for patients resistant to current endocrine therapy. DNA methylation in the promoter regions of genes is a prominent epigenetic gene silencing mechanism that contributes to breast cancer biology. In the current study, we investigated whether promoter DNA methylation could be associated with resistance to endocrine therapy in patients with recurrent breast cancer. Using a microarray-based technology, the promoter DNA methylation status of 117 candidate genes was studied in a cohort of 200 steroid hormone receptor-positive tumors of patients who received the antiestrogen tamoxifen as first-line treatment for recurrent breast cancer. Of the genes analyzed, the promoter DNA methylation status of 10 genes was significantly associated with clinical outcome of tamoxifen therapy. The association of the promoter hypermethylation of the strongest marker, phosphoserine aminotransferase (PSAT1) with favorable clinical outcome was confirmed by an independent quantitative DNA methylation detection method. Furthermore, the extent of DNA methylation of PSAT1 was inversely associated with its expression at the mRNA level. Finally, also at the mRNA level, PSAT1 was a predictor of tamoxifen therapy response. Concluding, our work indicates that promoter hypermethylation and mRNA expression of PSAT1 are indicators of response to tamoxifen-based endocrine therapy in steroid hormone receptor-positive patients with recurrent breast cancer.


Clinical Cancer Research | 2009

DNA methylation markers predict outcome in node-positive, Estrogen receptor-positive breast cancer with adjuvant anthracycline-based chemotherapy

Hartmann O; Spyratos F; Nadia Harbeck; Dimo Dietrich; Fassbender A; Manfred Schmitt; Serenella Eppenberger-Castori; Vuaroqueaux; Lerebours F; Welzel K; Sabine Maier; Achim Plum; Niemann S; John A. Foekens; Lesche R; John W. M. Martens

Purpose: We have shown that DNA methylation of the PITX2 gene predicts risk of distant recurrence in steroid hormone receptor-positive, node-negative breast cancer. Here, we present results from a multicenter study investigating whether PITX2 and other candidate DNA methylation markers predict outcome in node-positive, estrogen receptor-positive, HER-2-negative breast cancer patients who received adjuvant anthracycline-based chemotherapy. Experimental Design: Using a microarray platform, we analyzed DNA methylation in regulatory regions of PITX2 and 60 additional candidate genes in 241 breast cancer specimens. Using Cox regression analysis, we assessed the predictive power of the individual marker/marker panel candidates. Clinical endpoints were time to distant metastasis, disease-free survival, and overall survival. A nested bootstrap/cross-validation strategy was applied to identify and validate marker panels. Results: DNA methylation of PITX2 and 14 other genes was correlated with clinical outcome. In multivariate models, each methylation marker added significant information to established clinical factors. A four-marker panel including PITX2, BMP4, FGF4, and C20orf55 was identified that resulted in improvement of outcome prediction compared with PITX2 alone. Conclusions: This study provides further evidence for the PITX2 biomarker, which has now been successfully confirmed to predict outcome among different breast cancer patient populations. We further identify new DNA methylation biomarkers, three of which can be combined into a panel with PITX2 to increase the outcome prediction performance in our anthracycline-treated primary breast cancer population. Our results show that a well-defined panel of DNA methylation markers enables outcome prediction in lymph node-positive, HER-2-negative breast cancer patients treated with anthracycline-based chemotherapy.


Journal of Clinical Oncology | 2008

Multicenter Study Using Paraffin-Embedded Tumor Tissue Testing PITX2 DNA Methylation As a Marker for Outcome Prediction in Tamoxifen-Treated, Node-Negative Breast Cancer Patients

Nadia Harbeck; Inko Nimmrich; Arndt Hartmann; Jeffrey S. Ross; Tanja Cufer; Robert Grützmann; Glen Kristiansen; Angelo Paradiso; Oliver Hartmann; Astrid Margossian; John W.M. Martens; Ina Schwope; Antje Lukas; Volkmar Müller; Karin Milde-Langosch; Jörg Nährig; John A. Foekens; Sabine Maier; Manfred Schmitt; Ralf Lesche

PURPOSE We recently reported DNA methylation of the paired-like homeodomain transcription factor 2 (PITX2) gene to be strongly correlated with increased risk of recurrence in node-negative, hormone receptor-positive, tamoxifen-treated breast cancer patients using fresh frozen specimens. Aims of the present study were to establish determination of PITX2 methylation for routine analysis in formalin-fixed paraffin-embedded (FFPE) breast cancer tissue and to test PITX2 DNA methylation as a biomarker for outcome prediction in an independent patient cohort. PATIENTS AND METHODS Real-time polymerase chain reaction (PCR) technology was validated for FFPE tissue by comparing methylation measurements in FFPE specimens with those in fresh frozen specimens from the same tumor. The impact of PITX2 methylation on time to distant metastasis was then evaluated in FFPE specimens from hormone receptor-positive, node-negative breast cancer patients (n = 399, adjuvant tamoxifen monotherapy). RESULTS Reproducibility of the PCR assay in replicate measurements (r(s) > or = 0.95; n = 150) and concordant measurements between fresh frozen and FFPE tissues (r(s) = 0.81; n = 89) were demonstrated. In a multivariate model, PITX2 methylation added significant information (hazard ratio = 2.35; 95% CI, 1.20 to 4.60) to established prognostic factors (tumor size, grade, and age). CONCLUSION PITX2 methylation can be reliably assessed by real-time PCR technology in FFPE tissue. Together with our earlier studies, we have accumulated substantial evidence that PITX2 methylation analysis holds promise as a practical assay for routine clinical use to predict outcome in node-negative, tamoxifen-treated breast cancer, which might allow, based on future validation studies, the identification of low-risk patients who may be treated by tamoxifen alone.


American Journal of Pharmacogenomics | 2005

Identifying DNA Methylation Biomarkers of Cancer Drug Response

Sabine Maier; Christina Dahlstroem; Carolina Haefliger; Achim Plum; Christian Piepenbrock

In the last few years, DNA methylation has become one of the most studied gene regulation mechanisms in carcinogenesis as a result of the cumulative evidence produced by the scientific community. Moreover, advances in the technologies that allow detection of DNA methylation in a variety of analytes have opened the possibility of developing methylation-based tests. A number of studies have provided evidence that specific methylation changes can alter the response to different therapeutic agents in cancer and, therefore, be useful biomarkers. For example, the association of the methylation status of DNA repair genes such as MGMT and MLH1 illustrate the two main mechanisms of response to DNA damaging agents. Loss of methylation of MGMT, and the subsequent increase in gene expression, leads to a reduction in response to alkylating agents as a result of enhanced repair of drug-induced DNA damage. Conversely, the increase in methylation of MLH1 and its resulting loss of expression has been consistently observed in drug-resistant tumor cells. MLH1 encodes a mismatch repair enzyme activated in response to DNA damage; activation of MLH1 also induces apoptosis of tumor cells, and thus loss of its expression leads to resistance to DNA-damaging agents. Other methylation-regulated genes that could serve as biomarkers in cancer therapy include drug transporters, genes involved in microtubule formation and stability, and genes related to hormonal therapy response. These methylation markers have potential applications for disease prognosis, treatment response prediction, and the development of novel treatment strategies.


Annals of Hematology | 2005

Distinction of acute lymphoblastic leukemia from acute myeloid leukemia through microarray-based DNA methylation analysis.

Christian W. Scholz; Inko Nimmrich; Matthias Burger; Evelyne Becker; Bernd Dörken; Wolf-Dieter Ludwig; Sabine Maier

Patterns of DNA methylation are substantially altered in malignancies compared to normal tissue, with both genome-wide hypomethylation and regional increase of cytosine methylation at dinucleotides of cytosine and guanine, i.e., CpG dinucleotides. While genome-wide hypomethylation renders chromosomes instable, hypermethylation of CpGs in promoter regions is generally associated with transcriptional silencing, e.g., of tumor suppressor genes. To investigate whether disease-specific methylation profiles exist for different entities of acute leukemia, a microarray-based DNA methylation analysis simultaneously assessing 249 CpG dinucleotides originating from 57 genes was employed. Hereby, samples from precursor B-cell acute lymphoblastic leukemia (ALL) could be distinguished from cases of acute myeloid leukemia by virtue of N33, EGR4, CDC2, CCND2, or MOS hypermethylation in ALL.


American Journal of Clinical Pathology | 2005

Differential DNA methylation of gene promoters in small B-cell lymphomas

Juyuan Guo; Matthias Burger; Inko Nimmrich; Sabine Maier; Evelyne Becker; Buelent Genc; Dieter Duff; Farahnaz Rahmatpanah; Rebecca Chitma-Matsiga; Huidong Shi; Kurt Berlin; Tim H M Huang; Charles W. Caldwell

Improved care of patients with small B-cell lymphomas (SBCLs) is likely to result from the ongoing discovery of molecular markers that better define these malignant neoplasms. We identified multiple gene loci whose DNA methylation patterns differed between 3 types of SBCL: B-cell chronic lymphocytic leukemia/small lymphocytic lymphoma, mantle cell lymphoma, and grades I and II follicular lymphoma. This analysis was performed using an oligonucleotide microarray that allowed determination of the DNA methylation status of 156 loci in 38 genes. Combined bisulfite restriction analysis and methylation-specific polymerase chain reaction were used to validate the differential methylation of 6 of these genes. By using non-Hodgkin lymphoma cell lines as models, these genes were examined further for methylation and gene expression relationships. This study illustrates nonrandom epigenetic alterations in SBCLs that seem to preferentially involve lymphomas of germinal center derivation.


Personalized Medicine | 2005

DNA methylation markers – an opportunity to further individualize therapy in breast cancer?

Sabine Maier; Ralf Lesche; Inko Nimmrich; Florian Eckhardt; Christina Dahlstroem; Achim Plum

Over the last few decades, a wealth of treatment options have become available for breast cancer. To specifically direct those therapies to patients with the highest need who will receive the greatest benefit, biomarkers are urgently needed. Two specific needs seem to be most pressing: first is the need for prognostic markers, which would determine which group of patients may recover without adjuvant chemotherapy. Second, predictive markers for specific treatments, such as different endocrine treatments, chemotherapies or targeted drugs, are expected to play a major role in the near future. Ideally, such markers should be strong single markers, or low-complexity marker panels containing only a few markers, to allow for easier assay development and improved reproducibility. The possibility to measure the marker(s) in formalin-fixed specimens would greatly facilitate integration into routine clinical practice. A common and early event in breast cancer is aberrant DNA methylation within gene regulatory regions, affecting a variety of genes with different functions. Data from recently published studies indicate that altered DNA methylation carries prognostic as well as predictive information in breast cancer. Together with the technical advantages of a DNA-based marker, DNA methylation may well constitute the ideal biomarker to further individualize breast cancer treatment. Here the recent literature is reviewed and the most interesting markers, which have the potential to significantly change breast cancer treatment and, therefore, warrant further systematic clinical validation, are highlighted.


Archive | 2005

Methylation Analysis in Cancer

Carolina Haefliger; Sabine Maier; Alexander Olek

Aberrant DNA methylation is an early and common event in human cancers. Methylation acts as an epigenetic regulator of gene expression and is involved in cancer development as well as resistance to drug treatments. Specific methylation patterns have been shown for different cancer types and there is evidence that methylation can be used as a diagnostic tool. Several methods have been developed to study methylation on a genome wide basis. However they are labor intensive and can assess only a limited number of tissues at a time preventing the assessment of these genes in larger populations. Methylation microarrays now offer the possibility to validate these candidate genes statistically filling the gap between genome wide discovery methods and single gene assays which could be adjusted to routine clinical use. Here we show how all these methods can be combined to broaden our knowledge regarding DNA methylation and transform some of this information into powerful diagnostic tests.


Lung Cancer | 2003

E-26. Molecular markers in early detection

John K. Field; Jim Heighway; Shirley L. Smith; Triantafyllos Liloglou; Timothy H. Payne; Stephen W. Duffy; Daniel C. Betticher; Sabine Maier

‘Roy Castle ~nte~ational Centre for Lung Cancer Research, The University of Liverpool, 200 London Road, Liverpool, L3 9TA, UK; ‘Cancer Researcla UK Department of Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventive Medicine, Charterhouse Squam, London ECIM 6BQ; ‘institute of Medical Uncology. Inselspital, 3010 Bern, Switzerfand; 4Epigenomics AG, Berlin, Germany; SHoy Castle International Centre for Lung Cancer Research, The University of Liverpool, 200 London Road,

Collaboration


Dive into the Sabine Maier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John A. Foekens

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Adorjan

Technical University of Berlin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John W. M. Martens

Erasmus University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge