Skip to main content
Erschienen in: Brain Tumor Pathology 2/2022

Open Access 11.01.2022 | Case Report

A patient with two gliomas with independent oligodendroglioma and glioblastoma biology proved by DNA-methylation profiling: a case report and review of the literature

verfasst von: Theo F. J. Kraus, Christoph Schwartz, Lukas Machegger, Barbara Zellinger, Dorothee Hölzl, Hans U. Schlicker, Johannes Pöppe, Barbara Ladisich, Mathias Spendel, Michael Kral, Karl Sotlar

Erschienen in: Brain Tumor Pathology | Ausgabe 2/2022

Abstract

Here, we report on a patient presenting with two histopathologically distinct gliomas. At the age of 42, the patient underwent initial resection of a right temporal oligodendroglioma IDH mutated 1p/19q co-deleted WHO Grade II followed by adjuvant radiochemotherapy with temozolomide. 15 months after initial diagnosis, the patient showed right hemispheric tumor progression and an additional new left frontal contrast enhancement in the subsequent imaging. A re-resection of the right-sided tumor and resection of the left frontal tumor were conducted. Neuropathological work-up showed recurrence of the right-sided oligodendroglioma with features of an anaplastic oligodendroglioma WHO Grade III, but a glioblastoma WHO grade IV for the left frontal lesion. In depth molecular profiling revealed two independent brain tumors with distinct molecular profiles of anaplastic oligodendroglioma IDH mutated 1p/19q co-deleted WHO Grade III and glioblastoma IDH wildtype WHO grade IV. This unique and rare case of a patient with two independent brain tumors revealed by in-depth molecular work-up and epigenomic profiling emphasizes the importance of integrated work-up of brain tumors including methylome profiling for advanced patient care.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The 2016 World Health Organization (WHO) classification of tumors of the central nervous system (CNS) integrates, both, histology and molecular pathology as integrated aspects of brain tumor classification [8]. Thereby, DNA-methylation analysis is a promising novel technology for accurate brain tumor classification since previous studies revealed that distinct methylation profiles define distinct brain tumor entities with high accuracy [2, 3, 5, 9, 10, 12]. One of the most prominent examples is the inclusion of isocitrate dehydrogenase (IDH) 1 and 2 status, and loss of chromosomes 1p and 19q as integrated parts of the classification of glioma: Since 2016 the diagnosis of astrocytomas requires the analysis of IDH mutation status, and the diagnosis of oligodendrogliomas requires the assessment of both IDH mutations, as well as combined 1p/19q losses. [8] Thereby, oligodendrogliomas IDH mutated 1p/19q co-deleted show significantly better overall survival compared to astrocytomas IDH mutated and glioblastomas IDH wildtype [8].
Gliomas show a typical diffusely infiltrating growth pattern into surrounding brain tissue and recurrences after initial resection/treatment. Importantly, it has been established that the molecular features of gliomas, i.e. IDH-, 1p/19q- and TERT-Status, do not change during tumor recurrence and/or progression [8]. The distinct molecular background of astrocytomas WHO grade II and III as well as secondary glioblastomas WHO grade IV can be proven by revealing IDH1 mutations in codon 132 and IDH2 mutations in codon 172 [8]. The molecular background of oligodendrogliomas WHO grades II and III can be confirmed by demonstrating combined IDH1/2 mutations and chromosomal losses on 1p and 19q [8]. In contrast to the aforementioned gliomas, primary glioblastomas show IDH1/2 wildtype status [8].
Here, we report on a 42 years old patient with two brain tumors that showed distinct molecular patterns in integrated work-up and epigenomic profiling proving independent tumor origins.

Clinical summary

A 42 year-old male Caucasian patient was diagnosed with two intracranial lesions due to headache and nausea. The larger lesion, located in the right temporomesial lobe, showed signs of intratumoral hemorrhage as well as contrast enhancement with associated perifocal edema and midline shift (Fig. 1a). The other tumor was a cystoid mass located in the trigonal area (Fig. 1b). Upon decision in the interdisciplinary neuro-oncological tumorboard and receival of written informed consent, the patient underwent resection of the temporomesial tumor via a transtemporal approach. Postoperative magnetic resonance imaging (MRI) revealed a subtotal resection with minimal residual ventral contrast-enhancement (Fig. 1c). Histopathological evaluation revealed an oligodendroglioma, IDH1 mutated, 1p/19q-co-deleted WHO II, and a concomitant and adjuvant radiochemotherapy (50 Gy) with temozolomide (6 cycles) was initiated [11]. Initial follow-up imaging showed a stable temporomesial tumor and a decreased trigonal lesion. However, fifteen months after initial diagnosis a right-sided peritrigonal tumor progression was seen on MRI, and confirmed by [18F]fluoroethyltyrosine (FET)-PET CT (Fig. 1d). Due to only little mass effects of the progression, a wait-and-scan procedure was performed. However, in the subsequent MRIs, a new irregular circularly contrast enhancing lesion in the left frontal lobe was detected (Fig. 1e) sowing rapid tumor pregression (Fig. 1f). Re-resection of the right-sided tumor as well as the contralateral lesion was performed. The right frontotemporal lesion was now graded as an anaplastic oligodendroglioma, IDH mutated, 1p/19q co-deleted WHO III; and the left frontal tumor was classified as a glioblastoma IDH wildtype WHO IV. Subsequently the patient underwent re-irradiation with adjuvant bevacizumab therapy.

Pathological findings

The first manifestation showed in H&E staining a pleomorphic glial tumor with round tumor cells and perinuclear halos and only sparse mitoses (Fig. 2a). Immunohistochemistry performed on a Ventana Benchmark Ultra System with standard protocols showed that glial tumor cells were positive for GFAP (glial fibrillary acidic protein) with only short processes (Fig. 2b). Nuclear expression of ATRX (nuclear immunopositivity for α-thalassemia/mental-retardation-syndrome-X-linked) was retained (Fig. 2c), and there was expression of IDH1 (isocitrate dehydrogenase 1) R132H mutant protein (Fig. 2d). There were only sparse PHH3 (phosphorylated histone H3, H3S10p) positive cells (Fig. 2e) and proliferation was increased with 5% Ki67 positive cells (Fig. 2f). Analysis of the 1p and 19q status was performed by fluorescence in situ hybridization (FISH) using standard protocols, revealing a combined loss of 1p (Fig. 2g) and 19q (Fig. 2h). Thus, the tumor was classified as oligodendroglioma, IDH mutated, 1p/19q co-deleted, WHO grade II.
Recurrence of the right temporomesial lesion showed a similar picture as the first manifestation in H&E staining with round tumor cells and perinuclear halos but there was increased pleomorphy and brisk mitotic activity (Fig. 2i). Immunohistochemistry showed GFAP positive tumor cells with only short processes (Fig. 2j). Nuclear expression of ATRX was retained (Fig. 2k) and there was expression of IDH1 R132H mutant protein (Fig. 2l). There were increased PHH3 positive cells (Fig. 2m) and proliferation was increased with 25% Ki67 positive cells (Fig. 2n). Analysis of the 1p and 19q status performed by FISH revealed a combined loss of 1p (Fig. 2o) and 19q (Fig. 2p). Thus, this tumor was classified as recurrence of the previously described oligidendroglioma, then with features of anaplastic oligodendroglioma IDH mutated 1p/19q co-deleted WHO grade III.
Analysis of the left frontal lesion showed in H&E staining a highly pleomorphic glial tumor with long tumor processes, high mitotic activity and microvascular proliferation (Fig. 2q). Immunohistochemistry showed GFAP positive tumor cells with long processes (Fig. 2r). Nuclear expression of ATRX was retained (Fig. 2s). There was no expression of IDH1 R132H mutant protein (Fig. 2t). Reactions with antibodies against PHH3 showed increased mitoses (Fig. 2u). Proliferation was increased with 20% Ki67 positive cells (Fig. 2v). Analysis of the 1p and 19q status performed by FISH revealed no combined loss of 1p (Fig. 2w) and 19q (Fig. 2x). Thus, this tumor showed all the key hallmarks of a glioblastoma IDH wildtype WHO grade IV.

Molecular genetic profiling

Molecular genetic analysis was performed by extracting DNA from FFPE material using the Maxwell system (Promega) according to the manufacturer’s protocol and subsequent application of the Illumina Focus Panel (Illumina) on an Illumina MiniSeq device (Illumina) according to the manufacturer’s protocols enabling us to analyze 41 genes in parallel, including IDH1 and IDH2 hot spot regions (the complete gene list can be found in Table 1). Hot spot loci of TERT promoter were analyzed by Sanger sequencing [4, 7]. DNA-methylation profiling was performed using Illumina EPIC bead chips that were scanned on an Illumina NextSeq 550DX device. Data analysis was performed using the Molecular Neuropathology Pipeline of the German Cancer Research Center (DKFZ) [1].
Table 1
AmpliSeq for ilumina focus panel gene list
AKT1
EGFR
GNA11
KRAS
PIK3CA
ALK
ERBB2
GNAQ
MAP2K1
RAF1
AR
ERBB3
HRAS
MAP2K2
RET
BRAF
ERBB4
IDH1
MET
ROS1
CCND1
ESR1
IDH2
MTOR
SMO
CDK4
FGFR1
JAK1
MYC
 
CDK6
FGFR2
JAK2
MYCN
 
CTNNB1
FGFR3
JAK3
NRAS
 
DDR2
FGFR4
KIT
PDGFRA
 
An overview of all 41 genes coveres using the AmpliSeq for illumina focus panel
Integrated work-up of the first tumor manifestation showed an IDH1 R132H mutation (Fig. 3a) with IDH2 wildtype (Fig. 3b) and TERT C250T promoter mutation (Fig. 3c). DNA Methylation profiling showed methylated MGMT promoter (Fig. 3d), 1p and 19q losses in copy number profiling (Fig. 3e) and allocated the tumor to the methylation class of oligodendroglioma IDH mutated 1p/19q co-deleted (Fig. 3f).
Analysis of the recurrence revealed an analogous molecular profile: The tumor showed an IDH1 R132H mutation (Fig. 3g) with IDH2 wildtype (Fig. 3h) and TERT C250T promoter mutation (Fig. 3i). DNA Methylation profiling showed methylated MGMT promoter (Fig. 3j), 1p and 19q losses in copy number profiling (Fig. 3k) and allocated the tumor to the methylation class of oligodendroglioma IDH mutated 1p/19q co-deleted (Fig. 3l).
Interestingly, profiling of the left-sided tumor manifestation revealed a fundamentally different profile: This tumor showed IDH1 (Fig. 3m) and IDH2 wildtype (Fig. 3n) and TERT C228T promoter mutation (Fig. 3o).
DNA Methylation profiling showed unmethylated MGMT promoter (Fig. 3p); there was no 1p and 19q loss in copy number profiling (Fig. 3q) and allocated the tumor to the methylation class of glioblastoma IDH wildtype, subclass RTK I (Fig. 3r).
All other 40 genes covered by the AmpliSeq for Illumina Gene Panel showed an identical gene alteration profile in all three tumors (Table 2).
Table 2
Detected gen alterations
First tumor
Recurrence
Second tumor
Prediction
Gene
Allele frequency (reads) [forward/reverse]
c. HGVS
p. HGVS
Gene
Allele frequency (reads) [forward/reverse]
c. HGVS
p. HGVS
Gene
Allele frequency (reads) [forward/reverse]
c. HGVS
p. HGVS
Mutational effect
ALK
100% (8978) [100% (4363)/100% (4615)]
c.4381A > G
p.Ile1461Val
ALK
100% (8530) [100% (4109)/100% (4421)]
c.4381A > G
p.Ile1461Val
ALK
100% (6466) [100% (3128)/100% (3338)]
c.4381A > G
p.Ile1461Val
Class 1 (benign)
DDR2
31% (1079) [31% (542)/31% (537)]
c.278C > T
p.Thr93Ile
DDR2
26% (862) [26% (430)/27% (432)]
c.278C > T
p.Thr93Ile
DDR2
19% (579) [19% (294)/19% (285)]
c.278C > T
p.Thr93Ile
 
EGFR
29% (1418) [31% (627)/28% (791)]
c.89-10986delT
EGFR
26% (1358) [29% (591)/24% (767)]
c.89-10986delT
EGFR
26% (1579) [29% (696)/25% (883)]
c.89-10986delT
 
EGFR
52% (2986) [52% (1465)/51% (1521)]
c.1498 + 22A > T
 
EGFR
53% (2637) [54% (1297)/52% (1340)]
c.1498 + 22A > T
 
EGFR
34% (1794) [35% (884)/33% (910)]
c.1498 + 22A > T
 
Class 1 (benign)
ERBB3
100% (4558) [100% (2337)/100% (2221)]
c.234 + 8A > T
 
ERBB3
100% (5482) [100% (2817)/100% (2665)]
c.234 + 8A > T
 
ERBB3
100% (5267) [100% (2687)/100% (2580)]
c.234 + 8A > T
  
FGFR3
100% (4370) [100% (2228)/100% (2142)]
c.1953G > A
p.Thr651= 
FGFR3
100% (3469) [100% (1794)/100% (1675)]
c.1956G > A
p.Thr652= 
FGFR3
99% (2811) [100% (1457)/99% (1354)]
c.1956G > A
p.Thr652= 
Class 1 (benign)
FGFR4
48% (2725) [49% (1420)/47% (1305)]
c.92-65 T > C
 
FGFR4
51% (3086) [52% (1577)/51% (1509)]
c.92-65 T > C
 
FGFR4
53% (2405) [54% (1231)/53% (1174)]
c.92-65 T > C
  
FGFR4
49% (1362) [49% (697)/48% (665)]
c.407C > T
p.Pro136Leu
FGFR4
50% (1956) [50% (987)/50% (969)]
c.407C > T
p.Pro136Leu
FGFR4
48% (1747) [48% (895)/48% (852)]
c.407C > T
p.Pro136Leu
Class 1 (benign)
FGFR4
54% (833) [54% (432)/53% (401)]
c.483A > G
p.Ala161= 
FGFR4
54% (1041) [54% (537)/54% (504)]
c.483A > G
p.Ala16= 
FGFR4
55% (1115) [55% (573)/56% (542)]
c.483A > G
p.Ala161= 
 
FGFR4
46% (265) [48% (141)/45% (124)]
c.2016-43C > A
 
FGFR4
45% (365) [47% (190)/44% (175)]
c.1896-43C > A
 
FGFR4
50% (462) [52% (237)/49% (225)]
c.1896-43C > A
  
FGFR4
48% (272) [50% (137)/47% (135)]
c.2016-8A > G
 
FGFR4
48% (374) [52% (185)/45% (189)]
c.1896-8A > G
 
FGFR4
53% (475) [56% (233)/50% (242)]
c.1896-8A > G
  
IDH1
46% (5687) [46% (2808)/46% (2879)]
c.395G > A
p.Arg132His
IDH1
48% (5691) [48% (2838)/48% (2853)]
c.395G > A
p.Arg132His
IDH1
Class 5 (pathogenic)
KIT
48% (4257) [47% (2140)/49% (2117)]
c.67 + 4913A > G
 
KIT
48% (3192) [47% (1615)/48% (1577)]
c.67 + 4913A > G
 
KIT
46% (2393) [45% (1213)/46% (1180)]
c.67 + 4913A > G
  
KIT
23% (2198) [23% (1076)/23% (1122)]
c.67 + 4923delA
 
KIT
21% (1505) [21% (742)/21% (763)]
c.67 + 4923delA
 
KIT
22% (1249) [23% (615)/22% (634)]
c.67 + 4923delA
  
KIT
43% (4173) [37% (1745)/49% (2428)]
c.67 + 4953dupA
KIT
43% (3102) [37% (1306)/49% (1796)]
c.67 + 4953dupA
KIT
42% (2340) [36% (995)/47% (1345)]
c.67 + 4953dupA
 
KIT
100% (9294) [100% (4544)/100% (4750)]
c.756 + 334G > A
 
KIT
100% (7169) [100% (3547)/100% (3622)]
c.756 + 334G > A
 
KIT
100% (5445) [100% (2678)/100% (2767)]
c.756 + 334G > A
  
KIT
50% (4812) [50% (2414)/50% (2398)]
c.2362-333A > T
 
KIT
50% (4707) [50% (2358)/50% (2349)]
c.2362-333A > T
 
KIT
50% (4102) [50% (2048)/50% (2054)]
c.2362-333A > T
  
KRAS
51% (1012) [51% (496)/52% (516)]
c.-11-1877C > A
 
KRAS
44% (859) [43% (418)/44% (441)]
c.-11-1877C > A
 
KRAS
50% (1081) [50% (539)/50% (542)]
c.-11-1877C > A
  
KRAS
50% (6037) [50% (3022)/51% (3015)]
c.111 + 6969C > G
KRAS
46% (4123) [46% (2061)/46% (2062)]
c.112-3079C > G
 
KRAS
49% (3521) [49% (1762)/49% (1759)]
c.112-3079C > G
  
PDGFRA
100% (6940) [100% (3405)/100% (3535)]
c.1701A > G
p.Pro567= 
PDGFRA
100% (7371) [100% (3629)/100% (3742)]
c.1701A > G
p.Pro567= 
PDGFRA
100% (6090) [100% (3020)/100% (3070)]
c.1701A > G
p.Pro567= 
Class 1 (benign)
PIK3CA
12% (2249) [12% (1141)/12% (1108)]
c.2119G > A
p.Glu707Lys
PIK3CA
12% (1979) [12% (1004)/12% (975)]
c.2119G > A
p.Glu707Lys
PIK3CA
12% (1470) [12% (751)/11% (719)]
c.2119G > A
p.Glu707Lys
Class 3 (uv)
PIK3CA
23% (4396) [23% (2182)/23% (2214)]
c.2155C > G
p.Leu719Val
PIK3CA
22% (3717) [22% (1846)/23% (1871)]
c.2155C > G
p.Leu719Val
PIK3CA
22% (2822) [22% (1395)/23% (1427)]
c.2155C > G
p.Leu719Val
Class 2 (likely benign)
PIK3CA
24% (4435) [24% (2215)/23% (2220)]
c.2187 + 1G > T
 
PIK3CA
23% (3688) [22% (1804)/23% (1884)]
c.2187 + 1G > T
 
PIK3CA
23% (2813) [23% (1380)/23% (1433)]
c.2187 + 1G > T
 
Class 3 (uv)
RET
50% (3715) [51% (1930)/48% (1785)]
c.2307G > T
p.Leu769= 
RET
50% (3281) [51% (1692)/49% (1589)]
c.2307G > T
p.Leu769= 
RET
13% (406) [13% (209)/13% (197)]
c.2307G > T
p.Leu769= 
Class 1 (benign)
An overview of all detected gene alterations using the AmpliSeq for Illumina Focus Panel. All three tumors showed the same gene alterations except for the IDH1 R132H mutation (indicated in red color) that was not detected in the second tumor. Note that TERT mutations are not indicated here since TERT hot spot loci were analyzed by Sanger sequencing

Discussion

Here we report on an unique case of a patient that developed two molecularly independent gliomas: oligodendroglioma and glioblastoma.
To our knowledge, this is the first reported case of a patient with two independent gliomas of oligodendroglioma and glioblastoma biology that were confirmed by integrated in-depth molecular profiling including epigenomic DNA-methylation analysis.
A literature search revealed only one other published case of a cerebellar glioblastoma and a supratentorial oligodendroglioma [6]. Junaid et al. reported on a 44-years old patient, who suffered from a cerebellar glioma with typical histological features of glioblastoma, i.e. microvascular proliferation and necrosis, and a supratentorial glioma with histological hallmarks of an oligodendroglioma, i.e. small round cells with perinuclear halos [6]. However, only a conventional histological work-up of the specimens had been performed, and no immunohistochemical and molecular profiling to prove different biological background of the two reported gliomas had been provided [6].
In the case presented here, it is astonishing, that the completely removed WHO Grade II oligodendroglioma recurred after only 15 weeks after radiochemotherapy. This might be due to hypermutations occurred by temozolomide chemotherapy. Of the 41 genes covered by the AmpliSeq for Illumina Focus Panel (Table 1), we did not find any changes in the gene alteration profile in the first tumor and the recurrence (Table 2), however, this panel may be too small to answer the question of hypermutations occurring after temozolomide chemotherapy and a larger gene panel may be appropriate. Furthermore, the question rises if there were any germline mutations in the patients. Germline mutations may be an important co-factor in this unique case showing recurrence and progression of a WHO Grade II oligodendroglioma after only 15 weeks and a molecularly independent WHO Grade IV glioblastoma. Unfortunately, we did not have the chance to check for germline mutations in the presented case.
In summary, our presented case is an unique example of a patient with two different gliomas proved by in-depth molecular work-up. Besides different histology of oligodendroglioma and glioblastoma, the two brain tumors showed different molecular profiles of oligodendroglioma (i.e. IDH1 R132H mutation, combined 1p/19q loss, TERT C250T mutation) and glioblastoma (i.e. IDH1 wildtype, retained 1p/19q, TERT C228T mutation), respectively. Additionally, epigenomic DNA-methylation profiling clustered the tumors to the classes of oligodendroglioma IDH mutant 1p/19q co-deleted and glioblastoma IDH wildtype subclass RTK I.
Thus, this unique case emphasizes the need for integrated molecular work-up and demonstrates the power of in-depth profiling including DNA-methylation profiling in better understanding tumor biology and revealing tumor heterogeneity.

Acknowledgements

We would like to thank the members of the histology, immunohistochemistry and molecular pathology facilities at the Institute of Pathology for their support in conducting this study.

Declarations

Conflict of interest

None declared.

Ethical approval

The patient gave written informed consent for publication. All procedures performed in studies were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Capper D, Jones DTW, Sill M et al (2018) DNA methylation-based classification of central nervous system tumours. Nature 555:469–474CrossRef Capper D, Jones DTW, Sill M et al (2018) DNA methylation-based classification of central nervous system tumours. Nature 555:469–474CrossRef
2.
Zurück zum Zitat Capper D, Stichel D, Sahm F et al (2018) Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: the Heidelberg experience. Acta Neuropathol 136:181–210CrossRef Capper D, Stichel D, Sahm F et al (2018) Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: the Heidelberg experience. Acta Neuropathol 136:181–210CrossRef
3.
Zurück zum Zitat Halliday GC, Junckerstorff RC, Bentel JM et al (2018) The case for DNA methylation based molecular profiling to improve diagnostic accuracy for central nervous system embryonal tumors (not otherwise specified) in adults. J Clin Neurosci 47:163–167CrossRef Halliday GC, Junckerstorff RC, Bentel JM et al (2018) The case for DNA methylation based molecular profiling to improve diagnostic accuracy for central nervous system embryonal tumors (not otherwise specified) in adults. J Clin Neurosci 47:163–167CrossRef
4.
Zurück zum Zitat Holzl D, Hutarew G, Zellinger B et al (2021) Integrated analysis of programmed cell death ligand 1 expression reveals increased levels in high-grade glioma. J Cancer Res Clin Oncol 147:2271–2280CrossRef Holzl D, Hutarew G, Zellinger B et al (2021) Integrated analysis of programmed cell death ligand 1 expression reveals increased levels in high-grade glioma. J Cancer Res Clin Oncol 147:2271–2280CrossRef
5.
Zurück zum Zitat Jaunmuktane Z, Capper D, Jones DTW et al (2019) Methylation array profiling of adult brain tumours: diagnostic outcomes in a large, single centre. Acta Neuropathol Commun 7:24CrossRef Jaunmuktane Z, Capper D, Jones DTW et al (2019) Methylation array profiling of adult brain tumours: diagnostic outcomes in a large, single centre. Acta Neuropathol Commun 7:24CrossRef
6.
Zurück zum Zitat Junaid M, Bukhari SS, Sarfraz T (2014) A rare case of cerebellar glioblastoma multiforme and supratentorial oligodendroglioma presenting as synchronous primary brain tumors. J Pioneer Med Sci 4:129–131 Junaid M, Bukhari SS, Sarfraz T (2014) A rare case of cerebellar glioblastoma multiforme and supratentorial oligodendroglioma presenting as synchronous primary brain tumors. J Pioneer Med Sci 4:129–131
7.
Zurück zum Zitat Kraus TFJ, Machegger L, Poppe J et al (2020) Diffuse midline glioma of the cervical spinal cord with H3 K27M genotype phenotypically mimicking anaplastic ganglioglioma: a case report and review of the literature. Brain Tumor Pathol 37:89–94CrossRef Kraus TFJ, Machegger L, Poppe J et al (2020) Diffuse midline glioma of the cervical spinal cord with H3 K27M genotype phenotypically mimicking anaplastic ganglioglioma: a case report and review of the literature. Brain Tumor Pathol 37:89–94CrossRef
8.
Zurück zum Zitat Louis DN, Perry A, Reifenberger G et al (2016) The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820CrossRef Louis DN, Perry A, Reifenberger G et al (2016) The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol 131:803–820CrossRef
9.
Zurück zum Zitat Pajtler KW, Witt H, Sill M et al (2015) Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell 27:728–743CrossRef Pajtler KW, Witt H, Sill M et al (2015) Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell 27:728–743CrossRef
10.
Zurück zum Zitat Sahm F, Schrimpf D, Stichel D et al (2017) DNA methylation-based classification and grading system for meningioma: a multicentre, retrospective analysis. Lancet Oncol 18:682–694CrossRef Sahm F, Schrimpf D, Stichel D et al (2017) DNA methylation-based classification and grading system for meningioma: a multicentre, retrospective analysis. Lancet Oncol 18:682–694CrossRef
11.
Zurück zum Zitat Stupp R, Mason WP, Van Den Bent MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996CrossRef Stupp R, Mason WP, Van Den Bent MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996CrossRef
12.
Zurück zum Zitat Sturm D, Witt H, Hovestadt V et al (2012) Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22:425–437CrossRef Sturm D, Witt H, Hovestadt V et al (2012) Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 22:425–437CrossRef
Metadaten
Titel
A patient with two gliomas with independent oligodendroglioma and glioblastoma biology proved by DNA-methylation profiling: a case report and review of the literature
verfasst von
Theo F. J. Kraus
Christoph Schwartz
Lukas Machegger
Barbara Zellinger
Dorothee Hölzl
Hans U. Schlicker
Johannes Pöppe
Barbara Ladisich
Mathias Spendel
Michael Kral
Karl Sotlar
Publikationsdatum
11.01.2022
Verlag
Springer Nature Singapore
Erschienen in
Brain Tumor Pathology / Ausgabe 2/2022
Print ISSN: 1433-7398
Elektronische ISSN: 1861-387X
DOI
https://doi.org/10.1007/s10014-021-00423-0

Weitere Artikel der Ausgabe 2/2022

Brain Tumor Pathology 2/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Bluttest erkennt Parkinson schon zehn Jahre vor der Diagnose

10.05.2024 Parkinson-Krankheit Nachrichten

Ein Bluttest kann abnorm aggregiertes Alpha-Synuclein bei einigen Menschen schon zehn Jahre vor Beginn der motorischen Parkinsonsymptome nachweisen. Mit einem solchen Test lassen sich möglicherweise Prodromalstadien erfassen und die Betroffenen früher behandeln.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Wartezeit nicht kürzer, aber Arbeit flexibler

Psychotherapie Medizin aktuell

Fünf Jahren nach der Neugestaltung der Psychotherapie-Richtlinie wurden jetzt die Effekte der vorgenommenen Änderungen ausgewertet. Das Hauptziel der Novellierung war eine kürzere Wartezeit auf Therapieplätze. Dieses Ziel wurde nicht erreicht, es gab jedoch positive Auswirkungen auf andere Bereiche.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.