Skip to main content

BCOR::CREBBP fusion in malignant neuroepithelial tumor of CNS expands the spectrum of methylation class CNS tumor with BCOR/BCOR(L1)-fusion

Abstract

Methylation class “CNS tumor with BCOR/BCOR(L1)-fusion” was recently defined based on methylation profiling and tSNE analysis of a series of 21 neuroepithelial tumors with predominant presence of a BCOR fusion and/or characteristic CNV breakpoints at chromosome 22q12.31 and chromosome Xp11.4. Clear diagnostic criteria are still missing for this tumor type, specially that BCOR/BCOR(L1)-fusion is not a consistent finding in these tumors despite being frequent and that none of the Heidelberger classifier versions is able to clearly identify these cases, in particular tumors with alternative fusions other than those involving BCOR, BCORL1, EP300 and CREBBP. In this study, we introduce a BCOR::CREBBP fusion in an adult patient with a right temporomediobasal tumor, for the first time in association with methylation class “CNS tumor with BCOR/BCOR(L1)-fusion” in addition to 35 cases of CNS neuroepithelial tumors with molecular and histopathological characteristics compatible with “CNS tumor with BCOR/BCOR(L1)-fusion” based on a comprehensive literature review and data mining in the repository of 23 published studies on neuroepithelial brain Tumors including 7207 samples of 6761 patients. Based on our index case and the 35 cases found in the literature, we suggest the archetypical histological and molecular features of “CNS tumor with BCOR/BCOR(L1)-fusion”. We also present four adult diffuse glioma cases including GBM, IDH-Wildtype and Astrocytoma, IDH-Mutant with CREBBP fusions and describe the necessity of complementary molecular analysis in “CNS tumor with BCOR/BCOR(L1)-alterations for securing a final diagnosis.

Introduction

Methylation class “CNS tumor with BCOR/BCOR(L1)-fusion” was recently defined based on methylation profiling of a series of 21 neuroepithelial tumors with predominant presence of a BCOR fusion and/or characteristic CNV breakpoints at chromosome 22q12.31 and chromosome Xp11.4 [32]. Clear diagnostic criteria are still missing for this tumor type, specially that BCOR/BCOR(L1)-fusion is not a consistent finding in these tumors despite being frequent and that none of the Heidelberger classifier versions is able to clearly identify these cases, in particular tumors with alternative fusions other than those involving BCOR, BCORL1, EP300 and CREBBP. In this study, we introduce a BCOR::CREBBP fusion in an adult patient with a right temporomediobasal tumor and methylation class “CNS tumor with BCOR/BCOR(L1)-fusion”. This study presents a comprehensive literature review for additional inputs to this methylation class and explores the genetic alterations of BCOR, BCORL1 and their typical fusion partners EP300, TRAP1, CREBBP, and L3MBTL2 in the repository of 23 published studies on neuroepithelial brain Tumors.

Case presentation

Our case was a 14 years old girl with a large inhomogeneous right temporomediobasal mass with slight contrast enhancement and consecutive shift of midline structures in MRI (Fig. 1). After gross total resection, histological exam showed a hypercellular neuroepithelial tumor with ependymoma-like histological pattern including perivascular pseudorosettes, focal calcifications, and a high proliferation activity (Fig. 2). IDH1/2 pyrosequencing and pTERT Sanger sequencing showed wildtype sequences. Methylation profiling showed a significant calibrated score of 0.99 in v12.5 and v12.8 versions of Heidelberger brain tumor classifier and lower calibrated score of 0.79 in v11.b4 for methylation family class “CNS tumor with BCOR alteration”. While the versions 12.5 showed a calibrated score of 0.65 for the subtype “CNS tumor with BCOR/BCOR(L1)-fusion”, the versions 12.8 showed a discrepant result with calibrated score of 0.91 for the subtype HGNET-BCOR-ITD; however, PCR analysis showed no BCOR-ITD. RNA-based NGS analyses revealed a BCOR::CREBBP fusion (Fig. 2). The BCOR::CREBBP fusion was predicted to be out-of-frame, creating a premature stop codon at codon 2337 within the CREBBP segment. It was a persistent finding in 850 K-methylation analysis with corresponding copy number alterations at chromosomal regions 16p13.3 and Xp11.4 representing the CREBBP and BCOR gene locations (Fig. 2). The patient underwent proton radiation and adjuvant temozolomide therapy. Thirty months later the patient had a tumor recurrence and underwent a partial resection (Fig. 1). At the last follow up 32 months after the first diagnosis, the patient was alive with no evidence of recurrence or progress.

Fig. 1
figure 1

Radiological presentation of primary and recurrent tumor. The primary tumor was an inhomogeneous right temporomediobasal mass with slight contrast enhancement (T1-C) and consecutive shift of midline structure (a-c). Thirty months later, the PET/MRI and MR spectroscopy findings were clearly indicative of tumor recurrence. 18 F-fluoroethyl-tyrosine PET showed increased tracer uptake in the contrast-enhanced areas of the right basal ganglia and thalamus (d-f), and MR spectroscopy showed a corresponding tumor profile with increased choline and decreased N-acetylaspartate peaks (g-h)

Fig. 2
figure 2

Histological, immunohistochemical and genetic/epigenetic features of the tumor. (a) Histological exam revealed a hypercellular neuroepithelial tumor with ependymoma-like histological pattern including perivascular pseudorosettes, focal calcifications (a, b), and a high proliferation activity (e) with frequent mitotic figures (b, f). Olig2 was expressed in tumor cells and BCOR was negative (c, d). NF staining confirmed a diffuse growth pattern (g). GFAP was negative (h). (j) RNA-based next generation sequencing revealed a BCOR::CREBBP fusion with breakpoints in intron 6 of BCOR and exon 31 of CREBBP. (i) The copy number profile showed focal alterations on chromosomal regions 16p13.3 and Xp11.4 representing the CREBBP and BCOR gene locations

Discussion

Pathogenic alterations of BCOR have been reported in various tumor types including HGNET-BCOR-ITD, CNS tumor with BCOR/BCOR(L1)-fusion, endometrial stromal and soft tissue sarcomas, primitive myxoid mesenchymal tumors of infancy and undifferentiated round cell sarcoma. Regarding CNS tumor with BCOR/BCOR(L1)-fusion, beside the original publication very few cases have been reported in the literature [12, 25, 26, 29, 33, 34]. The core characteristic of “CNS tumor with BCOR/BCOR(L1)-fusion” is defined as predominant presence of a BCOR fusion with often EP300 as the fusion partner and/or characteristic CNV as mentioned before. In the primary study, occasional cases presented a BCOR stop mutation or MEAF6::CXXC5 fusion instead of the typical BCOR fusions [32].

In a comparative systematic review of the literature, we found 35 published cases of neuroepithelial CNS tumors that either carried a BCOR/BCOR(L1)- or other related fusions, or had a methylation profile compatible with CNS tumors with BCOR/BCOR(L1)-fusion (Supplementary Table 1). The majority of reported cases are supratentorial, but cerebellar localization occurs occasionally. Most of the patients are of pediatric or young adult age. However, this tumor occurs in a wide age range (5–72) with 55% of the cases under 40 and 85% under 50 years of age. The tumor is typically T1-hypointense, T2-hyperintense and can be well circumscribed or diffusely infiltrating both in radiological and in histological exam. Contrast enhancement might be present at the first manifestation. The archetypical histomorphology includes an oligodendroglial or ependymoma-like histological pattern with a delicate ‘chicken-wire’ vasculature and perivascular pseudorosettes. Focal calcifications are frequent and occasionally microcystic/ myxoid changes may be present. The tumor cells show round to oval nuclei with finely speckled chromatin and mild to moderate atypia as well as prominent glial processes. Malignancy features such as necrosis, frequent mitoses and microvascular proliferations might be present at the first manifestation. With the exception of Olig2, which is frequently positive in these tumors, other glial markers are variably expressed. Nuclear expression of BCOR is also variable and cannot be used as a surrogate marker. Although, within the reported cases, the majority did not show a positive nuclear expression of BCOR (15 of 25), 10 cases still showed a nuclear expression of BCOR (Supplementary Table 1). Our case was negative for BCOR. Similarly, the BCOR staining cannot differentiate between HGNET-BCOR-ITD and CNS tumor with BCOR/BCOR(L1)-fusion, since a majority of HGNET-BCOR-ITD also show a nuclear BCOR expression [11]. BCOR-ITD is not present in any of the CNS tumors with BCOR/BCOR(L1)-fusion and this helps differentiating from HGNET-BCOR-ITD. EP300::BCOR is the typical fusion found in these tumors with the common breakpoints exon 31 for EP300 and exon 4–7 for BCOR. Alternative fusions have been reported such as MEAF6::CXXC5, CREBBP::BCORL1, EP300::BCORL1, and BCOR::L3MBTL2 among tSNE-defined cases [2, 32].

We found a reported BCOR::CREBBP fusion in a neuroepithelial tumor with breakpoints in exon 4 of BCOR and exon 31 of CREBBP in the literature [22]. However, this study did not perform a methylation profiling at the time and the tumor did not show archetypical histopathological features of CNS tumors with BCOR/BCOR(L1)-fusion.

Besides, five additional inputs were found after searching for alterations in BCOR, BCORL1, EP300, TRAP1, CREBBP, and L3MBTL2 in the repository of 23 published studies on neuroepithelial brain Tumors including 7207 samples of 6761 patients (Supplementary Fig. 1) [3,4,5,6,7,8,9,10, 13,14,15,16,17, 19,20,21, 23, 24, 27, 28, 30, 31, 35]. These cases showed further related fusions including EP300::BCOR; BCOR::L3MBTL2; BCOR::EP300; BCOR::TRAP1; CREBBP::GOLGA6L2; CREBBP::ENPP7P14; CREBBP::LCMT1 (Supplementary Tables 1 and 2). Two of the cases with CREBBP::GOLGA6L2 and CREBBP::LCMT1 fusions fulfilled the molecular criteria for GBM, IDH wildtype including + 7/-10 signature and homozygous deletion of CDKN2A/B as well as IDH wildtype status. One case with CREBBP::ENPP7P14 fusion fulfilled the molecular criteria for astrocytoma, IDH-mutant including IDHR132H-mut, TP53-mut, ATRX-mut, no 1p/19q. One case with three fusions (EP300::BCOR; BCOR::L3MBTL2; BCOR::EP300) was described as a neuroepithelial tumor with small spindle cell pattern and spongioblastoma polar-like or ependymoma-like areas, calcifications, chicken-wire vasculature, necrosis, high mitotic activity and seemed to fit the archetypical histological pattern of CNS tumors with BCOR/BCOR(L1)-fusion. However, no methylation-based classification was available for this tumor. One case with BCOR::TRAP1 fusion was diagnosed as diffuse glioma grade 4 and showed mutations on SETD2 C495Lfs*6, PTPN11 A72V, PTPN11 E76G, TP53 E258K and a CDKN2A/B homozygous deletion but could not be further classified (Supplementary Table 1).

The BCOR::CREBBP fusion in our case was predicted to be out-of-frame, resulting in no chimeric transcript and potentially leading to a loss of function of BCOR. BCOR plays a role in pluripotency maintenance, differentiation induction and cell fate determination via regulation of histone methylation through its PCGF Ub-like fold discriminator (PUFD) domain. PUFD is an essential domain for tumor suppressor function of BCOR [18]. Loss of BCOR function is associated with tumorigenesis [1]. On the other hand, the BCOR gene is located on the X-chromosome, available as only one allele, so that the fusion in the index case would potentially lead to a complete loss of putative tumor-suppressor activity mediated by the PUFD. Out-of-frame BCOR::CREBBP fusion has already been reported in malignant diffuse glioma of pediatric age, potentially a supporting evidence against a stochastic event [22].

Conclusion

CNS tumor with BCOR/BCOR(L1)-fusion seems to be an independently existing tumor type with archetypical histological and molecular features, however, consistent diagnostic criteria are still missing. These tumors frequently show oligodendroglial or ependymoma-like morphology with a delicate ‘chicken-wire’ vasculature and perivascular pseudorosettes, focal calcifications, occasionally microcystic/myxoid changes and malignancy features such as necrosis, frequent mitoses and microvascular proliferations, as well as frequent Olig2 expression. None of the Heidelberger classifier versions is able to clearly identify the cases, especially tumors with alternative fusions other than those involving BCOR, BCORL1, EP300 and CREBBP. Nuclear expression of BCOR is variable and cannot be used as a surrogate marker. Nevertheless, lack of BCOR-ITD helps differentiating them from HGNET-BCOR-ITD. Knowing that adult diffuse gliomas such as GBM, IDH-Wildtype and Astrocytoma, IDH-Mutant may present CREBBP fusions, it is of utmost importance to take additional molecular findings and the histomorphology into account while interpreting the results. Interpretation of survival data is recommended to be postponed to larger cohorts. Published cases in the literature can be used as a guide for individual decision making for patients.

Data availability

Data are available from the corresponding author on reasonable request.

References

  1. Astolfi A, Fiore M, Melchionda F, Indio V, Bertuccio SN, Pession A (2019) BCOR involvement in cancer. Epigenomics 11:835–855. https://doi.org/10.2217/epi-2018-0195

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Barresi V, Cardoni A, Miele E, Pedace L, Masotto B, Nardini C, Barresi S, Rossi S (2024) CNS tumor with CREBBP::BCORL1 Fusion and pathogenic mutations in BCOR and CREBBP: expanding the spectrum of BCOR-altered tumors. Acta Neuropathol Commun 12:8. https://doi.org/10.1186/s40478-024-01726-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Barthel FP, Johnson KC, Varn FS, Moskalik AD, Tanner G, Kocakavuk E, Anderson KJ, Abiola O, Aldape K, Alfaro KD al (2019) Longitudinal molecular trajectories of diffuse glioma in adults. Nature 576:112–120. https://doi.org/10.1038/s41586-019-1775-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Bhandari V, Hoey C, Liu LY, Lalonde E, Ray J, Livingstone J, Lesurf R, Shiah YJ, Vujcic T, Huang X al (2019) Molecular landmarks of tumor hypoxia across cancer types. Nat Genet 51:308–318. https://doi.org/10.1038/s41588-018-0318-2

    Article  CAS  PubMed  Google Scholar 

  5. Bonneville R, Krook MA, Kautto EA, Miya J, Wing MR, Chen HZ, Reeser JW, Yu L, Roychowdhury S (2017) Landscape of microsatellite instability across 39 Cancer types. JCO Precis Oncol 2017. https://doi.org/10.1200/PO.17.00073

  6. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, Zheng S, Chakravarty D, Sanborn JZ, Berman SHet al et al (2013) The somatic genomic landscape of glioblastoma. Cell 155:462–477. https://doi.org/10.1016/j.cell.2013.09.034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cancer Genome Atlas Research N (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455:1061–1068. https://doi.org/10.1038/nature07385

    Article  CAS  Google Scholar 

  8. Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray BA, Morozova O, Newton Y, Radenbaugh A, Pagnotta SM al (2016) Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164:550–563. https://doi.org/10.1016/j.cell.2015.12.028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ding L, Bailey MH, Porta-Pardo E, Thorsson V, Colaprico A, Bertrand D, Gibbs DL, Weerasinghe A, Huang KL, Tokheim C et al (2018) Perspective on Oncogenic Processes at the End of the Beginning of Cancer Genomics. Cell 173: 305–320 e310 https://doi.org/10.1016/j.cell.2018.03.033

  10. Ellrott K, Bailey MH, Saksena G, Covington KR, Kandoth C, Stewart C, Hess J, Ma S, Chiotti KE, McLellan M et al (2018) Scalable Open Science Approach for Mutation Calling of Tumor Exomes Using Multiple Genomic Pipelines. Cell Syst 6: 271–281 e277 https://doi.org/10.1016/j.cels.2018.03.002

  11. Ferris SP, Velazquez Vega J, Aboian M, Lee JC, Van Ziffle J, Onodera C, Grenert JP, Saunders T, Chen YY, Banerjee A et al (2020) High-grade neuroepithelial tumor with BCOR exon 15 internal tandem duplication-a comprehensive clinical, radiographic, pathologic, and genomic analysis. Brain Pathol 30: 46–62 https://doi.org/10.1111/bpa.12747

  12. Fukuoka K, Kanemura Y, Shofuda T, Fukushima S, Yamashita S, Narushima D, Kato M, Honda-Kitahara M, Ichikawa H, Kohno T et al (2018) Significance of molecular classification of ependymomas: C11orf95-RELA fusion-negative supratentorial ependymomas are a heterogeneous group of tumors. Acta Neuropathol Commun 6: 134 https://doi.org/10.1186/s40478-018-0630-1

  13. Gao Q, Liang WW, Foltz SM, Mutharasu G, Jayasinghe RG, Cao S, Liao WW, Reynolds SM, Wyczalkowski MA, Yao L et al (2018) Driver Fusions and Their Implications in the Development and Treatment of Human Cancers. Cell Rep 23: 227–238 e223 https://doi.org/10.1016/j.celrep.2018.03.050

  14. Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, Shen R, Taylor AM, Cherniack AD, Thorsson V (2018) al Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer. Cell 173: 291–304 e296 https://doi.org/10.1016/j.cell.2018.03.022

  15. Johnson BE, Mazor T, Hong C, Barnes M, Aihara K, McLean CY, Fouse SD, Yamamoto S, Ueda H, Tatsuno Ket al et al (2014) Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343:189–193. https://doi.org/10.1126/science.1239947

    Article  CAS  PubMed  Google Scholar 

  16. Jones DT, Hutter B, Jager N, Korshunov A, Kool M, Warnatz HJ, Zichner T, Lambert SR, Ryzhova M, Quang DA al (2013) Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet 45:927–932. https://doi.org/10.1038/ng.2682

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jonsson P, Lin AL, Young RJ, DiStefano NM, Hyman DM, Li BT, Berger MF, Zehir A, Ladanyi M Solit DB (2019) genomic correlates of Disease Progression and Treatment Response in prospectively characterized gliomas. Clin Cancer Res 25: 5537–5547 Doi 10.1158/1078– 0432.CCR-19-0032

  18. Junco SE, Wang R, Gaipa JC, Taylor AB, Schirf V, Gearhart MD, Bardwell VJ, Demeler B, Hart PJ, Kim CA (2013) Structure of the polycomb group protein PCGF1 in complex with BCOR reveals basis for binding selectivity of PCGF homologs. Structure 21:665–671. https://doi.org/10.1016/j.str.2013.02.013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, Kovatich AJ, Benz CC, Levine DA, Lee AV et al (2018) An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell 173: 400–416 e411 https://doi.org/10.1016/j.cell.2018.02.052

  20. Miller AM, Shah RH, Pentsova EI, Pourmaleki M, Briggs S, Distefano N, Zheng Y, Skakodub A, Mehta SA, Campos Cet al et al (2019) Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature 565:654–658. https://doi.org/10.1038/s41586-019-0882-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Petralia F, Tignor N, Reva B, Koptyra M, Chowdhury S, Rykunov D, Krek A, Ma W, Zhu Y, Ji Jet al et al (2020) Integrated Proteogenomic Characterization across Major Histological Types of Pediatric Brain Cancer. Cell 183: 1962–1985 e1931 https://doi.org/10.1016/j.cell.2020.10.044

  22. Pisapia DJ, Ohara K, Bareja R, Wilkes DC, Hissong E, Croyle JA, Kim JH, Saab J, MacDonald TY, Beg Set al et al (2020) Fusions involving BCOR and CREBBP are rare events in infiltrating glioma. Acta Neuropathol Commun 8:80. https://doi.org/10.1186/s40478-020-00951-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, Kosciolek T, Janssen S, Metcalf J, Song SJ al (2020) Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature 579:567–574. https://doi.org/10.1038/s41586-020-2095-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia Set al et al (2018) Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 173: 321–337 e310 https://doi.org/10.1016/j.cell.2018.03.035

  25. Tauziede-Espariat A, Pierron G, Siegfried A, Guillemot D, Uro-Coste E, Nicaise Y, Castel D, Catalaa I, Larrieu-Ciron D, Chaynes Pet al et al (2020) The EP300:BCOR fusion extends the genetic alteration spectrum defining the new tumoral entity of CNS tumors with BCOR internal tandem duplication. Acta Neuropathol Commun 8:178. https://doi.org/10.1186/s40478-020-01064-8

    Article  PubMed  PubMed Central  Google Scholar 

  26. Tauziede-Espariat A, Uro-Coste E, Sievers P, Nicaise Y, Mariet C, Siegfried A, Pierron G, Guillemot D, Benzakoun J Pallud J (2023) CNS tumor with EP300::BCOR fusion: discussing its prevalence in adult population. Acta Neuropathol Commun 11: 26 https://doi.org/10.1186/s40478-023-01523-y

  27. Taylor AM, Shih J, Ha G, Gao GF, Zhang X, Berger AC, Schumacher SE, Wang C, Hu H, Liu J et al (2018) Genomic and Functional Approaches to Understanding Cancer Aneuploidy. Cancer Cell 33: 676–689 e673 https://doi.org/10.1016/j.ccell.2018.03.007

  28. Thomas AA, Abrey LE, Terziev R, Raizer J, Martinez NL, Forsyth P, Paleologos N, Matasar M, Sauter CS, Moskowitz C al (2017) Multicenter phase II study of temozolomide and myeloablative chemotherapy with autologous stem cell transplant for newly diagnosed anaplastic oligodendroglioma. Neuro Oncol 19:1380–1390. https://doi.org/10.1093/neuonc/nox086

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Torre M, Meredith DM, Dubuc A, Solomon DA, Perry A, Vasudevaraja V, Serrano J, Snuderl M, Ligon KL, Alexandrescu S (2019) Recurrent EP300-BCOR fusions in Pediatric Gliomas with distinct clinicopathologic features. J Neuropathol Exp Neurol 78:305–314. https://doi.org/10.1093/jnen/nlz011

    Article  CAS  PubMed  Google Scholar 

  30. Vaubel RA, Tian S, Remonde D, Schroeder MA, Mladek AC, Kitange GJ, Caron A, Kollmeyer TM, Grove R, Peng S et al (2020) Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma. Clin Cancer Res 26: 1094–1104 https://doi.org/10.1158/1078-0432.CCR-19-0909

  31. Wang LB, Karpova A, Gritsenko MA, Kyle JE, Cao S, Li Y, Rykunov D, Colaprico A, Rothstein JH, Hong Ret al et al (2021) Proteogenomic and metabolomic characterization of human glioblastoma. Cancer Cell 39: 509–528 e520 https://doi.org/10.1016/j.ccell.2021.01.006

  32. Wu Z, Rajan S, Chung HJ, Raffeld M, Panneer Selvam P, Schweizer L, Perry A, Samuel D, Giannini C, Ragunathan Aet al et al (2022) Molecular and clinicopathologic characteristics of gliomas with EP300::BCOR fusions. Acta Neuropathol 144:1175–1178. https://doi.org/10.1007/s00401-022-02508-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xu Y, Hou Y, Gao X, Li J, Jiang D, Li Z, Hu P, Wang Y, Wen Y, Yao Xet al et al (2023) Report two adult cases of high-grade neuroepithelial neoplasm harbouring EP300::BCOR fusions with comprehensive molecular detection. Brain Pathol 33:e13177. https://doi.org/10.1111/bpa.13177

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yamazaki A, Arai Y, Fukuoka K, Nakano Y, Hama N, Nakata S, Makino K, Kuroda JI, Shinojima N, Mukasa Aet al et al (2022) Diffusely infiltrating glioma with CREBBP-BCORL1 fusion showing overexpression of not only BCORL1 but BCOR: a case report. Brain Tumor Pathol 39:171–178. https://doi.org/10.1007/s10014-022-00435-4

    Article  CAS  PubMed  Google Scholar 

  35. Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, Bordbar D, Shan D, Samanamud J Mahajan A (2019) Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med 25: 462–469 https://doi.org/10.1038/s41591-019-0349-y

Download references

Funding

Not applicable.

Open Access funding enabled and organized by Projekt DEAL.

Author information

Authors and Affiliations

Authors

Contributions

AE: conception and design of the study, acquisition, analysis, and interpretation of data, writing the manuscript; AW, JS, GG, AW: acquisition, analysis, and interpretation of data; MUS, TP: revision of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Azadeh Ebrahimi.

Ethics declarations

Ethics approval and consent to participate

The study of the present case report has been performed in accordance with the Declaration of Helsinki and the local standards.

Consent for publication

The patients signed informed consent for using surgical material and anonymous health information for scientific purposes and publication. All authors have approved the publication of the study.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Supplementary Material 3

Rights and permissions

Open Access This 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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ebrahimi, A., Waha, A., Schittenhelm, J. et al. BCOR::CREBBP fusion in malignant neuroepithelial tumor of CNS expands the spectrum of methylation class CNS tumor with BCOR/BCOR(L1)-fusion. acta neuropathol commun 12, 60 (2024). https://doi.org/10.1186/s40478-024-01780-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40478-024-01780-5

Keywords