Network


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

Hotspot


Dive into the research topics where Matthias Meinhardt is active.

Publication


Featured researches published by Matthias Meinhardt.


International Journal of Cancer | 2009

Gene signatures of pulmonary metastases of renal cell carcinoma reflect the disease‐free interval and the number of metastases per patient

Daniela Wuttig; Barbara Baier; Susanne Fuessel; Matthias Meinhardt; Alexander Herr; Christian Hoefling; Marieta Toma; Marc-Oliver Grimm; Axel Meye; Axel Rolle; Manfred P. Wirth

Our understanding of metastatic spread is limited and molecular mechanisms causing particular characteristics of metastasis are largely unknown. Herein, transcriptome‐wide expression profiles of a unique cohort of 20 laser‐resected pulmonary metastases (Mets) of 18 patients with clear‐cell renal cell carcinoma (RCC) were analyzed to identify expression patterns associated with two important prognostic factors in RCC: the disease‐free interval (DFI) after nephrectomy and the number of Mets per patient. Differentially expressed genes were identified by comparing early (DFI ≤ 9 months) and late (DFI ≥ 5 years) Mets, and Mets derived from patients with few (≤8) and multiple (≥16) Mets. Early and late Mets could be separated by the expression of genes involved in metastasis‐associated processes, such as angiogenesis, cell migration and adhesion (e.g., PECAM1, KDR). Samples from patients with multiple Mets showed an elevated expression of genes associated with cell division and cell cycle (e.g., PBK, BIRC5, PTTG1) which indicates that a high number of Mets might result from an increased growth potential. Minimal sets of genes for the prediction of the DFI and the number of Mets per patient were identified. Microarray results were confirmed by quantitative PCR by including nine further pulmonary Mets of RCC. In summary, we showed that subgroups of Mets are distinguishable based on their expression profiles, which reflect the DFI and the number of Mets of a patient. To what extent the identified molecular factors contribute to the development of these characteristics of metastatic spread needs to be analyzed in further studies.


Embo Molecular Medicine | 2016

Endothelial cell‐derived angiopoietin‐2 is a therapeutic target in treatment‐naive and bevacizumab‐resistant glioblastoma

Alexander Scholz; Patrick N. Harter; Sebastian Cremer; Burak Hasan Yalcin; Stefanie Gurnik; Maiko Yamaji; Mariangela Di Tacchio; Kathleen Sommer; Peter Baumgarten; Oliver Bähr; Joachim P. Steinbach; Jörg Trojan; Martin Glas; Ulrich Herrlinger; Dietmar Krex; Matthias Meinhardt; Astrid Weyerbrock; Marco Timmer; Roland Goldbrunner; Martina Deckert; Christian Braun; Jens Schittenhelm; Jochen T. Frueh; Evelyn Ullrich; Michel Mittelbronn; Karl H. Plate; Yvonne Reiss

Glioblastoma multiforme (GBM) is treated by surgical resection followed by radiochemotherapy. Bevacizumab is commonly deployed for anti‐angiogenic therapy of recurrent GBM; however, innate immune cells have been identified as instigators of resistance to bevacizumab treatment. We identified angiopoietin‐2 (Ang‐2) as a potential target in both naive and bevacizumab‐treated glioblastoma. Ang‐2 expression was absent in normal human brain endothelium, while the highest Ang‐2 levels were observed in bevacizumab‐treated GBM. In a murine GBM model, VEGF blockade resulted in endothelial upregulation of Ang‐2, whereas the combined inhibition of VEGF and Ang‐2 leads to extended survival, decreased vascular permeability, depletion of tumor‐associated macrophages, improved pericyte coverage, and increased numbers of intratumoral T lymphocytes. CD206+ (M2‐like) macrophages were identified as potential novel targets following anti‐angiogenic therapy. Our findings imply a novel role for endothelial cells in therapy resistance and identify endothelial cell/myeloid cell crosstalk mediated by Ang‐2 as a potential resistance mechanism. Therefore, combining VEGF blockade with inhibition of Ang‐2 may potentially overcome resistance to bevacizumab therapy.


PLOS ONE | 2013

Analyses of potential predictive markers and survival data for a response to sunitinib in patients with metastatic renal cell carcinoma.

Juana Dornbusch; Aristeidis Zacharis; Matthias Meinhardt; Kati Erdmann; Ingmar Wolff; Michael Froehner; Manfred P. Wirth; Stefan Zastrow; Susanne Fuessel

Background Patients with metastatic clear cell renal cell carcinoma (ccRCC) are frequently treated with tyrosine kinase inhibitors (TKI) such as sunitinib. It inhibits angiogenic pathways by mainly targeting the receptors of VEGF and PDGF. In ccRCC, angiogenesis is characterized by the inactivation of the von Hippel-Lindau gene (VHL) which in turn leads to the induction of HIF1α target genes such as CA9 and VEGF. Furthermore, the angiogenic phenotype of ccRCC is also reflected by endothelial markers (CD31, CD34) or other tumor-promoting factors like Ki67 or survivin. Methods Tissue microarrays from primary tumor specimens of 42 patients with metastatic ccRCC under sunitinib therapy were immunohistochemically stained for selected markers related to angiogenesis. The prognostic and predictive potential of theses markers was assessed on the basis of the objective response rate which was evaluated according to the RECIST criteria after 3, 6, 9 months and after last report (12–54 months) of sunitinib treatment. Additionally, VHL copy number and mutation analyses were performed on DNA from cryo-preserved tumor tissues of 20 ccRCC patients. Results Immunostaining of HIF-1α, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRα and -β was significantly associated with the sunitinib response after 6 and 9 months as well as last report under therapy. Furthermore, HIF-1α, CA9, CD34, VEGFR1 and -3 and PDGRFα showed significant associations with progression-free survival (PFS) and overall survival (OS). In multivariate Cox proportional hazards regression analyses high CA9 membrane staining and a response after 9 months were independent prognostic factors for longer OS. Frequently observed copy number loss and mutation of VHL gene lead to altered expression of VHL, HIF-1α, CA9, and VEGF. Conclusions Immunoexpression of HIF-1α, CA9, Ki67, CD31, pVEGFR1, VEGFR1 and -2, pPDGFRα and -β in the primary tumors of metastatic ccRCC patients might support the prediction of a good response to sunitinib treatment.


Oncotarget | 2015

Distribution and prognostic relevance of tumor-infiltrating lymphocytes (TILs) and PD-1/PD-L1 immune checkpoints in human brain metastases

Patrick N. Harter; Simon Bernatz; Alexander Scholz; Pia Zeiner; Jenny Zinke; Makoto Kiyose; Stella Blasel; Rudi Beschorner; Christian Senft; Benjamin Bender; Michael W. Ronellenfitsch; Harriet Wikman; Markus Glatzel; Matthias Meinhardt; Tareq A. Juratli; Joachim P. Steinbach; Karl H. Plate; Jörg Wischhusen; Benjamin Weide; Michel Mittelbronn

The activation of immune cells by targeting checkpoint inhibitors showed promising results with increased patient survival in distinct primary cancers. Since only limited data exist for human brain metastases, we aimed at characterizing tumor infiltrating lymphocytes (TILs) and expression of immune checkpoints in the respective tumors. Two brain metastases cohorts, a mixed entity cohort (n = 252) and a breast carcinoma validation cohort (n = 96) were analyzed for CD3+, CD8+, FOXP3+, PD-1+ lymphocytes and PD-L1+ tumor cells by immunohistochemistry. Analyses for association with clinico-epidemiological and neuroradiological parameters such as patient survival or tumor size were performed. TILs infiltrated brain metastases in three different patterns (stromal, peritumoral, diffuse). While carcinomas often show a strong stromal infiltration, TILs in melanomas often diffusely infiltrate the tumors. Highest levels of CD3+ and CD8+ lymphocytes were seen in renal cell carcinomas (RCC) and strongest PD-1 levels on RCCs and melanomas. High amounts of TILs, high ratios of PD-1+/CD8+ cells and high levels of PD-L1 were negatively correlated with brain metastases size, indicating that in smaller brain metastases CD8+ immune response might get blocked. PD-L1 expression strongly correlated with TILs and FOXP3 expression. No significant association of patient survival with TILs was observed, while high levels of PD-L1 showed a strong trend towards better survival in melanoma brain metastases (Log-Rank p = 0.0537). In summary, melanomas and RCCs seem to be the most immunogenic entities. Differences in immunotherapeutic response between tumor entities regarding brain metastases might be attributable to this finding and need further investigation in larger patient cohorts.


International Journal of Cancer | 2012

CD31, EDNRB and TSPAN7 are promising prognostic markers in clear-cell renal cell carcinoma revealed by genome-wide expression analyses of primary tumors and metastases†

Daniela Wuttig; Stefan Zastrow; Susanne Füssel; Marieta Toma; Matthias Meinhardt; Kristin Kalman; Kerstin Junker; Jimsgene Sanjmyatav; Kerstin Boll; Jörg Hackermüller; Axel Rolle; Marc-Oliver Grimm; Manfred P. Wirth

Currently used clinicopathological parameters are insufficient for a reliable prediction of metastatic risk and disease‐free survival (DFS) of patients with clear‐cell renal cell carcinoma (ccRCC). To identify prognostic genes, the expression profiles of primary ccRCC obtained from patients with different DFS — eight synchronously, nine metachronously and seven not metastasized tumors — were determined by genome‐wide expression analyses. Synchronously and metachronously metastasized primary ccRCC differed in the expression of 167 genes. Thirty‐six of these genes were also differentially expressed in synchronously vs. metachronously developed pulmonary metastases analyzed in a previous study. Because of their DFS‐associated deregulation that is concordant in metastases and primary ccRCC, these genes are potentially functionally involved in metastatic tumor growth and are also prognostically useful. A prognostic impact was confirmed for the genes CD31, EDNRB and TSPAN7 at the mRNA level (n = 86), and for TSPAN7 at the protein level (n = 106). Patients with a higher gene expression of EDNRB or TSPAN7, or with TSPAN7‐positive vessels in both cores investigated on tissue microarrays had a significantly longer DFS and tumor‐specific survival (TSS). Patients with a higher CD31 gene expression showed a significantly longer TSS. EDNRB was an independent prognostic marker for the DFS. CD31, EDNRB and TSPAN7 had an independent impact on the TSS. In summary, comparative analysis of primary tumors and metastases is appropriate to identify independent prognostic markers in ccRCC. Gene expression of CD31 and EDNRB, and endothelial TSPAN7 protein level are potentially useful to improve outcome prediction because of their independent prognostic impact.


The Journal of Clinical Endocrinology and Metabolism | 2016

Epigenetic Mutation of the Succinate Dehydrogenase C Promoter in a Patient With Two Paragangliomas

Susan Richter; Barbara Klink; Brit Nacke; Aguirre A. de Cubas; Anastasios Mangelis; Elena Rapizzi; Matthias Meinhardt; Christina Skondra; Massimo Mannelli; Mercedes Robledo; Mario Menschikowski; Graeme Eisenhofer

CONTEXT Mutational inactivation of the succinate dehydrogenase (SDH) complex is a well-described cause of tumor development in pheochromocytomas/paragangliomas (PPGLs) and gastrointestinal stromal tumors (GISTs). Epigenetic inactivation of the SDHC gene is a more recently discovered phenomenon, which so far has only been described in GISTs and PPGLs from patients with Carney triad syndrome. CASE DESCRIPTION A 33-year-old patient presented with two abdominal paragangliomas (PGLs) and an adrenocortical adenoma. Both PGLs showed high succinate:fumarate ratios indicative of SDHx mutations; however, no mutations in any of the known PPGL susceptibility genes were found in leucocyte or tumor DNA. We identified methylation of the SDHC promoter region in both PGLs, which coincided with decreased SDHC expression at mRNA and protein levels and a hypermethylated epigenomic signature (CpG island methylator phenotype). Low-level SDHC promoter methylation was also observed in the adenoma but not in normal adrenal tissue or blood, suggesting postzygotic somatic mosaicism for SDHC promoter methylation in the patient. CONCLUSIONS This report provides evidence that SDHC promoter methylation can cause PGLs due to SDHC inactivation, emphasizing the importance of considering epigenetic changes and functional readouts in the genetic evaluation of patients not only with GISTs and Carney triad but also with PPGL.


Clinical Cancer Research | 2016

PI3K pathway inhibition achieves potent antitumor activity in melanoma brain metastases in vitro and in vivo

Heike Niessner; Jennifer Schmitz; Ghazaleh Tabatabai; Andreas Schmid; Carsten Calaminus; Tobias Sinnberg; Benjamin Weide; Thomas K. Eigentler; Claus Garbe; Birgit Schittek; Leticia Quintanilla-Fend; Benjamin Bender; Marion Mai; Christian Praetorius; Stefan Beissert; Gabriele Schackert; Michael H. Muders; Matthias Meinhardt; Gustavo Baretton; Reinhard Dummer; Keith T. Flaherty; Bernd J. Pichler; Dagmar Kulms; Dana Westphal; Friedegund Meier

Purpose: Great advances have recently been made in treating patients with metastatic melanoma. However, existing therapies are less effective on cerebral than extracerebral metastases. This highlights the potential role of the brain environment on tumor progression and drug resistance and underlines the need for “brain-specific” therapies. We previously showed that the PI3K-AKT survival pathway is hyperactivated in brain but not extracerebral melanoma metastases and that astrocyte-conditioned medium activates AKT in melanoma cells in vitro. We therefore tested the PI3K inhibitor buparlisib as an antitumor agent for melanoma brain metastases. Experimental Design and Results: Buparlisib inhibited AKT activity, decreased proliferation, and induced apoptosis in metastatic melanoma cell lines and short-term brain melanoma cells, irrespective of their BRAF and NRAS mutation status. In addition, buparlisib inhibited hyperactivated AKT and induced apoptosis in melanoma cells that were stimulated with astrocyte-conditioned medium. The growth of tumors induced by injecting human BRAF- and NRAS-mutant metastatic melanoma cells into the brain of mice was significantly inhibited by buparlisib. Conclusions: These results emphasize the value of targeting the PI3K pathway as a strategy to develop drugs for melanoma brain metastases. Clin Cancer Res; 22(23); 5818–28. ©2016 AACR.


Cancer Investigation | 2013

Immunohistochemical Analysis of Prognostic Protein Markers for Primary Localized Clear Cell Renal Cell Carcinoma

Thomas Weber; Matthias Meinhardt; Stefan Zastrow; Andreas Wienke; Susanne Fuessel; Manfred P. Wirth

Recurrence is not reliably predictable in localized clear cell renal cell carcinoma. Proteinmarkers could improve predictive accuracy. Tissue-microarrays from 132 patients with primary localized ccRCC were immunohistochemically analyzed for VHL, Ki67, p53, p21, survivin, and, for microvessel-density, UEA-1. Nuclear stainings of Ki67, p21, and survivin were significantly associated with disease-specific survival and increased predictive ability from 74% to 76%, 77%, and 78%, respectively in a multivariate model including T-stage and Fuhrman grade. A score-variable, combining Ki67-, p21-, and nS-staining identified a subset of patients with high risk of disease recurrence and increased predictive ability in the multivariate model to 84%.


Cancer Investigation | 2011

Expression of the Forkhead Transcription Factor FOXP1 is Associated with Tumor Grade and Ki67 Expression in Clear Cell Renal Cell Carcinoma

Marieta Toma; Thomas Weber; Matthias Meinhardt; Stefan Zastrow; Marc-Oliver Grimm; Susanne Füssel; Manfred P. Wirth; Gustavo Baretton

Expression of FOXP1 and Ki67 was immunohistochemically assessed on tissue microarrays of 129 cases of clear cell renal cell carcinomas. Overall and disease-specific survival correlated inversely with pT-category, grading and lymph node metastasis in (p < .05). Expression of FOXP1 correlated negatively with tumor grading (p = .02), but neither with pT-category nor with lymph node metastasis. Significant positive correlation was shown for Ki67 expression and tumor stage and lymph node metastasis (p < .05). The overall survival and the disease-specific survival correlated negatively with the Ki67 status (p < .05). FOXP1 expression negatively correlated with Ki67 expression in clear cell renal cell carcinomas (p = .036).


Genes, Chromosomes and Cancer | 2013

PARK2 and PACRG are commonly downregulated in clear‐cell renal cell carcinoma and are associated with aggressive disease and poor clinical outcome

Marieta Toma; Daniela Wuttig; Sandy Kaiser; Alexander Herr; Thomas Weber; Stefan Zastrow; Rainer Koch; Matthias Meinhardt; Gustavo Baretton; Manfred P. Wirth; Susanne Fuessel

PARK2 is an E3 ligase, known to be involved in ubiquitination of several proteins and to play a role in neuronal protection. The gene PARK2 and its potentially co‐regulated gene PACRG have been previously found to be deleted in clear‐cell renal cell carcinomas (ccRCCs). The aim of our study was to evaluate the mRNA and protein expression of PARK2 and PACRG in a large cohort of ccRCC, and to investigate their association with outcome. The expression of both genes was measured by quantitative PCR in 94 primary ccRCCs and autologous nonmalignant kidney tissues. PACRG and PARK2 protein expression was determined immunohistochemically using tissue microarrays comprising 133 ccRCCs. The mRNA and protein expression of PARK2 and PACRG was significantly downregulated in ccRCCs compared with nonmalignant tissues. Low levels of PARK2 mRNA were associated with high‐grade ccRCC and lymph node metastasis. Patients with low PARK2 mRNA levels showed a higher tumor‐specific mortality rate and a shorter overall survival (OS) than those with high PARK2 expression. Patients without PACRG mRNA expression in the tumor had a shorter disease‐free survival and OS than those with tumors expressing PACRG. In multivariate analyses, neither PARK2 nor PACRG expression were independent prognostic factors. The protein expression of PARK2 and PACRG was significantly downregulated in ccRCCs (82.8, and 96.9%, respectively), but no association with clinical outcome was noticed.

Collaboration


Dive into the Matthias Meinhardt's collaboration.

Top Co-Authors

Avatar

Manfred P. Wirth

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Susanne Fuessel

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Stefan Zastrow

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Gustavo Baretton

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Gabriele Schackert

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Marc-Oliver Grimm

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Axel Meye

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Daniela Wuttig

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Kati Erdmann

Dresden University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge