Network


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

Hotspot


Dive into the research topics where Jochen Meyer is active.

Publication


Featured researches published by Jochen Meyer.


Acta Neuropathologica | 2008

Analysis of the IDH1 codon 132 mutation in brain tumors

Jörg Balss; Jochen Meyer; Wolf Mueller; Andrey Korshunov; Christian Hartmann; Andreas von Deimling

A recent study reported on mutations in the active site of the isocitrate dehydrogenase (IDH1) gene in 12% of glioblastomas. All mutations detected resulted in an amino acid exchange in position 132. We analyzed the genomic region spanning wild type R132 of IDH1 by direct sequencing in 685 brain tumors including 41 pilocytic astrocytomas, 12 subependymal giant cell astrocytomas, 7 pleomorphic xanthoastrocytomas, 93 diffuse astrocytomas, 120 adult glioblastomas, 14 pediatric glioblastomas, 105 oligodendrogliomas, 83 oligoastrocytomas, 31 ependymomas, 58 medulloblastomas, 9 supratentorial primitive neuroectodermal tumors, 17 schwannomas, 72 meningiomas and 23 pituitary adenomas. A total of 221 somatic IDH1 mutations were detected and the highest frequencies occurred in diffuse astrocytomas (68%), oligodendrogliomas (69%), oligoastrocytomas (78%) and secondary glioblastomas (88%). Primary glioblastomas and other entities were characterized by a low frequency or absence of mutations in amino acid position 132 of IDH1. The very high frequency of IDH1 mutations in WHO grade II astrocytic and oligodendroglial gliomas suggests a role in early tumor development.


Acta Neuropathologica | 2009

Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas

Christian Hartmann; Jochen Meyer; Jörg Balss; David Capper; Wolf Mueller; Arne Christians; Jörg Felsberg; Marietta Wolter; Christian Mawrin; Wolfgang Wick; Michael Weller; Christel Herold-Mende; Andreas Unterberg; Judith W. M. Jeuken; Peter Wesseling; Guido Reifenberger; Andreas von Deimling

Somatic mutations in the IDH1 gene encoding cytosolic NADP+-dependent isocitrate dehydrogenase have been shown in the majority of astrocytomas, oligodendrogliomas and oligoastrocytomas of WHO grades II and III. IDH2 encoding mitochondrial NADP+-dependent isocitrate dehydrogenase is also mutated in these tumors, albeit at much lower frequencies. Preliminary data suggest an importance of IDH1 mutation for prognosis showing that patients with anaplastic astrocytomas, oligodendrogliomas and oligoastrocytomas harboring IDH1 mutations seem to fare much better than patients without this mutation in their tumors. To determine mutation types and their frequencies, we examined 1,010 diffuse gliomas. We detected 716 IDH1 mutations and 31 IDH2 mutations. We found 165 IDH1 (72.7%) and 2 IDH2 mutations (0.9%) in 227 diffuse astrocytomas WHO grade II, 146 IDH1 (64.0%) and 2 IDH2 mutations (0.9%) in 228 anaplastic astrocytomas WHO grade III, 105 IDH1 (82.0%) and 6 IDH2 mutations (4.7%) in 128 oligodendrogliomas WHO grade II, 121 IDH1 (69.5%) and 9 IDH2 mutations (5.2%) in 174 anaplastic oligodendrogliomas WHO grade III, 62 IDH1 (81.6%) and 1 IDH2 mutations (1.3%) in 76 oligoastrocytomas WHO grade II and 117 IDH1 (66.1%) and 11 IDH2 mutations (6.2%) in 177 anaplastic oligoastrocytomas WHO grade III. We report on an inverse association of IDH1 and IDH2 mutations in these gliomas and a non-random distribution of the mutation types within the tumor entities. IDH1 mutations of the R132C type are strongly associated with astrocytoma, while IDH2 mutations predominantly occur in oligodendroglial tumors. In addition, patients with anaplastic glioma harboring IDH1 mutations were on average 6 years younger than those without these alterations.


Brain Pathology | 2010

Characterization of r132h mutation-specific idh1 antibody binding in brain tumors

David Capper; Susanne Weißert; Jörg Balss; Antje Habel; Jochen Meyer; Diana Jäger; Ulrike Ackermann; Claudia Tessmer; Andrey Korshunov; Hanswalter Zentgraf; Christian Hartmann; Andreas von Deimling

Heterozygous point mutations of isocitrate dehydrogenase (IDH)1 codon 132 are frequent in grade II and III gliomas. Recently, we reported an antibody specific for the IDH1R132H mutation. Here we investigate the capability of this antibody to differentiate wild type and mutated IDH1 protein in central nervous system (CNS) tumors by Western blot and immunohistochemistry. Results of protein analysis are correlated to sequencing data. In Western blot, anti‐IDH1R132H mouse monoclonal antibody mIDH1R132H detected a specific band only in mutated tumors. Immunohistochemistry of 345 primary brain tumors demonstrated a strong cytoplasmic and weaker nuclear staining in 122 cases. Correlation with direct sequencing of 186 cases resulted in consensus of 177 cases. Genetic retesting of cases with conflicting findings resulted in a match of 186/186 cases, with all discrepancies resolving in favor of immunohistochemistry. Intriguing is the ability of mIDH1R132H to detect single infiltrating tumor cells. The very high frequency and the distribution of this mutation among specific brain tumor entities allow the highly sensitive and specific discrimination of various tumors by immunohistochemistry, such as anaplastic astrocytoma from primary glioblastoma or diffuse astrocytoma World Health Organization (WHO) grade II from pilocytic astrocytoma or ependymoma. Noteworthy is the discrimination of the infiltrating edge of tumors with IDH1 mutation from reactive gliosis.


Cancer Cell | 2013

Reduced H3K27me3 and DNA Hypomethylation Are Major Drivers of Gene Expression in K27M Mutant Pediatric High-Grade Gliomas

Sebastian Bender; Yujie Tang; Anders M. Lindroth; Volker Hovestadt; David T. W. Jones; Marcel Kool; Marc Zapatka; Paul A. Northcott; Dominik Sturm; Wei Wang; Bernhard Radlwimmer; Jonas W. Højfeldt; Nathalene Truffaux; David Castel; Simone Schubert; Marina Ryzhova; Huriye Şeker-Cin; Jan Gronych; Pascal-David Johann; Sebastian Stark; Jochen Meyer; Till Milde; Martin U. Schuhmann; Martin Ebinger; Camelia Maria Monoranu; Anitha Ponnuswami; Spenser Chen; Chris Jones; Olaf Witt; V. Peter Collins

Two recurrent mutations, K27M and G34R/V, within histone variant H3.3 were recently identified in ∼50% of pHGGs. Both mutations define clinically and biologically distinct subgroups of pHGGs. Here, we provide further insight about the dominant-negative effect of K27M mutant H3.3, leading to a global reduction of the repressive histone mark H3K27me3. We demonstrate that this is caused by aberrant recruitment of the PRC2 complex to K27M mutant H3.3 and enzymatic inhibition of the H3K27me3-establishing methyltransferase EZH2. By performing chromatin immunoprecipitation followed by next-generation sequencing and whole-genome bisulfite sequencing in primary pHGGs, we show that reduced H3K27me3 levels and DNA hypomethylation act in concert to activate gene expression in K27M mutant pHGGs.


Acta Neuropathologica | 2009

Combined molecular analysis of BRAF and IDH1 distinguishes pilocytic astrocytoma from diffuse astrocytoma

Andrey Korshunov; Jochen Meyer; David Capper; Arne Christians; Marc Remke; Hendrik Witt; Stefan M. Pfister; Andreas von Deimling; Christian Hartmann

Separation of pilocytic astrocytoma from diffuse astrocytomas frequently poses problems mostly related to small sample size. Precise classification and grading are essential due to different therapeutic strategies prompted by diagnoses of pilocytic astrocytoma WHO grade I, diffuse astrocytomas WHO grade II or anaplastic astrocytoma WHO grade III. Recently, genomic aberrations with a high specificity for distinct glioma entities have been described. Pilocytic astrocytomas carry a duplication at chromosome band 7q34 containing a BRAF–KIAA1549 gene fusion in the majority of cases. IDH1 mutations are observed very frequently in adult astrocytomas and IDH2 mutations have been reported in some astrocytomas. We examined a series of 120 astrocytomas including 70 pilocytic astrocytomas WHO grade I and 50 diffuse astrocytomas WHO grade II for both, BRAF–KIAA1549 fusion with a newly developed FISH assay and mutations in IDH1 and IDH2 by direct sequencing. Pilocytic astrocytomas contained the BRAF fusion in 49 cases (70%) but neither IDH1 nor IDH2 mutations. Astrocytomas WHO grade II exhibited IDH1 mutations in 38 cases (76%) but neither IDH2 mutations nor BRAF fusions. Thus, combined molecular analysis of BRAF and IDH1 is a sensitive and highly specific approach to separate pilocytic astrocytoma from diffuse astrocytoma.


Acta Neuropathologica | 2012

Immunohistochemical testing of BRAF V600E status in 1,120 tumor tissue samples of patients with brain metastases

David Capper; Anna Sophie Berghoff; Manuel Magerle; Aysegul Ilhan; Adelheid Wöhrer; Monika Hackl; Josef Pichler; Stefan Pusch; Jochen Meyer; Antje Habel; Peter Petzelbauer; Peter Birner; Andreas von Deimling; Matthias Preusser

Brain metastases (BM) are frequent and carry a dismal prognosis. BRAF V600E mutations are found in a broad range of tumor types and specific inhibitors targeting BRAF V600E protein exist. We analyzed tumoral BRAF V600E-mutant protein expression using the novel mutation-specific antibody VE1 in a series of 1,120 tumor specimens (885 BM, 157 primary tumors, 78 extra-cranial metastases) of 874 BM patients. In 85 cases, we performed validation of immunohistochemical results by BRAF exon 15 gene sequencing. BRAF V600E protein was expressed in BM of 42/76 (55.3%) melanomas, 1/15 (6.7%) ovarian cancers, 4/72 (5.5%) colorectal cancers, 1/355 (0.3%) lung cancers, 2/6 thyroid cancers and 1/2 choriocarcinomas. BRAF V600E expression showed high intra-tumoral homogeneity and was similar in different tumor manifestations of individual patients. VE1 immunohistochemistry and BRAF exon 15 sequencing were congruent in 68/70 (97.1%) cases, but VE1 immunostaining identified small BRAF V600E expressing tumor cell aggregates in 10 cases with inconclusive genetic results. Melanoma patients with BRAF V600E mutant protein expressing tumors were significantly younger at diagnosis of the primary tumor and at operation of BM than patients with non-mutated tumors. In conclusion, expression of BRAF V600E mutant protein occurs in approximately 6% of BM and is consistent in different tumor manifestations of the same patient. Thus, BRAF V600E inhibiting therapies seem feasible in selected BM patients. Immunohistochemical visualization of V600E-mutant BRAF protein is a promising tool for patient stratification. An integrated approach combining both, VE1 immunohistochemistry and genetic analysis may increase the diagnostic accuracy of BRAF mutation analysis.


Acta Neuropathologica | 2013

Meningeal hemangiopericytoma and solitary fibrous tumors carry the NAB2-STAT6 fusion and can be diagnosed by nuclear expression of STAT6 protein

Leonille Schweizer; Christian Koelsche; Felix Sahm; Rosario M. Piro; David Capper; David E. Reuss; Stefan Pusch; Antje Habel; Jochen Meyer; Tanja Göck; David T. W. Jones; Christian Mawrin; Jens Schittenhelm; Albert J. Becker; Stephanie Heim; Matthias Simon; Christel Herold-Mende; Gunhild Mechtersheimer; Werner Paulus; Rainer König; Otmar D. Wiestler; Stefan M. Pfister; Andreas von Deimling

Non-central nervous system hemangiopericytoma (HPC) and solitary fibrous tumor (SFT) are considered by pathologists as two variants of a single tumor entity now subsumed under the entity SFT. Recent detection of frequent NAB2-STAT6 fusions in both, HPC and SFT, provided additional support for this view. On the other hand, current neuropathological practice still distinguishes between HPC and SFT. The present study set out to identify genes involved in the formation of meningeal HPC. We performed exome sequencing and detected the NAB2-STAT6 fusion in DNA of 8/10 meningeal HPC thereby providing evidence of close relationship of these tumors with peripheral SFT. Due to the considerable effort required for exome sequencing, we sought to explore surrogate markers for the NAB2-STAT6 fusion protein. We adopted the Duolink proximity ligation assay and demonstrated the presence of NAB2-STAT6 fusion protein in 17/17 HPC and the absence in 15/15 meningiomas. More practical, presence of the NAB2-STAT6 fusion protein resulted in a strong nuclear signal in STAT6 immunohistochemistry. The nuclear reallocation of STAT6 was detected in 35/37 meningeal HPC and 25/25 meningeal SFT but not in 87 meningiomas representing the most important differential diagnosis. Tissues not harboring the NAB2-STAT6 fusion protein presented with nuclear expression of NAB2 and cytoplasmic expression of STAT6 proteins. In conclusion, we provide strong evidence for meningeal HPC and SFT to constitute variants of a single entity which is defined by NAB2-STAT6 fusion. In addition, we demonstrate that this fusion can be rapidly detected by STAT6 immunohistochemistry which shows a consistent nuclear reallocation. This immunohistochemical assay may prove valuable for the differentiation of HPC and SFT from other mesenchymal neoplasms.


PLOS ONE | 2012

Prognostic Value of Three Different Methods of MGMT Promoter Methylation Analysis in a Prospective Trial on Newly Diagnosed Glioblastoma

Arne Christians; Christian Hartmann; Axel Benner; Jochen Meyer; Andreas von Deimling; Michael Weller; Wolfgang Wick; Markus Weiler

Hypermethylation in the promoter region of the MGMT gene encoding the DNA repair protein O6-methylguanine-DNA methyltransferase is among the most important prognostic factors for patients with glioblastoma and predicts response to treatment with alkylating agents like temozolomide. Hence, the MGMT status is widely determined in most clinical trials and frequently requested in routine diagnostics of glioblastoma. Since various different techniques are available for MGMT promoter methylation analysis, a generally accepted consensus as to the most suitable diagnostic method remains an unmet need. Here, we assessed methylation-specific polymerase chain reaction (MSP) as a qualitative and semi-quantitative method, pyrosequencing (PSQ) as a quantitative method, and methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) as a semi-quantitative method in a series of 35 formalin-fixed, paraffin-embedded glioblastoma tissues derived from patients treated in a prospective clinical phase II trial that tested up-front chemoradiotherapy with dose-intensified temozolomide (UKT-05). Our goal was to determine which of these three diagnostic methods provides the most accurate prediction of progression-free survival (PFS). The MGMT promoter methylation status was assessable by each method in almost all cases (n = 33/35 for MSP; n = 35/35 for PSQ; n = 34/35 for MS-MLPA). We were able to calculate significant cut-points for the continuous methylation signals at each CpG site analysed by PSQ (range, 11.5 to 44.9%) and at one CpG site assessed by MS-MLPA (3.6%) indicating that a dichotomisation of continuous methylation data as a prerequisite for comparative survival analyses is feasible. Our results show that, unlike MS-MLPA, MSP and PSQ provide a significant improvement of predicting PFS compared with established clinical prognostic factors alone (likelihood ratio tests: p<0.001). Conclusively, taking into consideration prognostic value, cost effectiveness and ease of use, we recommend pyrosequencing for analyses of MGMT promoter methylation in high-throughput settings and MSP for clinical routine diagnostics with low sample numbers.


Brain Pathology | 2010

PCR- and Restriction Endonuclease-Based Detection of IDH1 Mutations

Jochen Meyer; Stefan Pusch; Jörg Balss; David Capper; Wolf Mueller; Arne Christians; Christian Hartmann; Andreas von Deimling

Hotspot mutations in codon 132 of the gene encoding isocitrate dehydrogenase 1 (IDH1) have emerged as the most frequent DNA alteration in astrocytomas, oligodendrogliomas and oligoastrocytomas. These mutations have been shown to be of significant diagnostic and prognostic value. So far, assessment of IDH1 mutation relied on DNA sequencing techniques.


European Journal of Haematology | 2010

Increased levels of 2-hydroxyglutarate in AML patients with IDH1-R132H and IDH2-R140Q mutations

Leopold Sellner; David Capper; Jochen Meyer; Claus Dieter Langhans; Christine Maria Hartog; Heike Pfeifer; Hubert Serve; Anthony D. Ho; Jürgen G. Okun; Alwin Krämer; Andreas von Deimling

To the Editor: Acute myeloid leukemia (AML) is a clonal malignancy of myeloid blood progenitor cells. Karyotyping provides the most powerful prognostic information. Especially for AML cases with normal karyotype, recurring mutations with prognostic significance in genes such as FLT3, NPM1, CEBPA, WT1 and MLL have been identified (1). Recently, isocitrate dehydrogenase (IDH) 1 mutations, which are well characterized in gliomas, were also reported for patients with AML (2). In astrocytomas, oligodendrogliomas and mixed oligoastrocytomas, it was found that IDH1-R132H mutations are the most frequent genetic alterations (3). Notably, IDH1 mutations are emerging as a major diagnostic and prognostic marker in gliomas. Other malignancies only rarely exhibit IDH mutations. However, recently, it has been shown that up to 10% of patients with AML carry mutations in IDH1, with approximately half of the patients harboring R132H and R132C substitutions, respectively (2, 4–7). Additionally, mutations in IDH2 can be found in up to 20% of patients with AML, with the R140Q mutation being detected more often compared to R172K (7). Of interest, the cytogenetic profile of patients with IDH1 mutations is strongly associated with a normal karyotype, intermediate cytogenetic risk and the prevalence of NPM1 mutations (2, 8). The impact of IDH mutations on prognosis

Collaboration


Dive into the Jochen Meyer's collaboration.

Top Co-Authors

Avatar

Andreas von Deimling

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David Capper

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrey Korshunov

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Mawrin

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

David E. Reuss

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge