We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
ReviewOpen Accesscc iconby iconnc iconnd icon

BCOR involvement in cancer

    Annalisa Astolfi

    ‘Giorgio Prodi’ Cancer Research Center, University of Bologna, 40138 Bologna, Italy

    ,
    Michele Fiore

    Pediatric Oncology & Hematology Unit ‘Lalla Seràgnoli’, S Orsola-Malpighi Hospital, 40138 Bologna, Italy

    ,
    Fraia Melchionda

    *Author for correspondence:

    E-mail Address: fraia.melchionda@aosp.bo.it

    Pediatric Oncology & Hematology Unit ‘Lalla Seràgnoli’, S Orsola-Malpighi Hospital, 40138 Bologna, Italy

    ,
    Valentina Indio

    ‘Giorgio Prodi’ Cancer Research Center, University of Bologna, 40138 Bologna, Italy

    ,
    Salvatore N Bertuccio

    Pediatric Oncology & Hematology Unit ‘Lalla Seràgnoli’, S Orsola-Malpighi Hospital, 40138 Bologna, Italy

    &
    Andrea Pession

    Pediatric Oncology & Hematology Unit ‘Lalla Seràgnoli’, S Orsola-Malpighi Hospital, 40138 Bologna, Italy

    Department of Medical & Surgical Sciences, University of Bologna, S Orsola-Malpighi Hospital, 40138 Bologna, Italy

    Published Online:https://doi.org/10.2217/epi-2018-0195

    Abstract

    BCOR is a gene that encodes for an epigenetic regulator involved in the specification of cell differentiation and body structure development and takes part in the noncanonical polycomb repressive complex 1. This review provides a comprehensive summary of BCOR’s involvement in oncology, illustrating that various BCOR aberrations, such as the internal tandem duplications of the PCGF Ub-like fold discriminator domain and different gene fusions (mainly BCOR–CCNB3, BCOR–MAML3 and ZC3H7B–BCOR), represent driver elements of various sarcomas such as clear cell sarcoma of the kidney, primitive mesenchymal myxoid tumor of infancy, small round blue cell sarcoma, endometrial stromal sarcoma and histologically heterogeneous CNS neoplasms group with similar genomic methylation patterns known as CNS-HGNET-BCOR. Furthermore, other BCOR alterations (often loss of function mutations) recur in a large variety of mesenchymal, epithelial, neural and hematological tumors, suggesting a central role in cancer evolution.

    BCOR is an epigenetic regulator

    It is known that tumorigenesis depends on the sequential acquisition of proto-oncogene-activating mutations and/or oncosuppressor loss of function. In this oversimplified perspective, epigenetic alterations are less easily framed, even if the resulting inappropriate expression of oncogenes and oncosuppressors can play an equally crucial role in cancer development and progression. Therefore, epigenetic regulators are potential proto-oncogenes or oncosuppressors, depending on the function of their target genes. A gene implicated in transcriptional regulation, BCOR is increasingly reported as mutated in different human cancers, with a key role in neoplastic transformation or in tumor progression.

    BCOR is located on chromosome X, in the Xp11.4 locus, and derives its name from its function as an interacting corepressor of BCL-6 that enhances BCL-6-mediated transcriptional repression [1]. The gene has 16 alternative exons coding several protein isoforms, with the principal isoform, encoded by 14 exons, that gives rise to a protein of 1755 amino acids. BCOR shows virtually ubiquitous expression, but only a few isoforms retain known protein interactions, depending on the domains preserved by alternative splicing.

    The function of BCOR is primarily mediated by two domains: the BCL-6-binding domain that interacts with the transcriptional repressor BCL-6 and PUFD, a common domain recurrent in different proteins implicated in histone regulation that binds the RAWUL domain of PCGF proteins (Figure 1).

    Figure 1. Structure and functional domains of BCOR, including BCL-6- and MLLT3-binding domains, ANK repeats and the PUFD domain.

    A schematic representation of the exon structure is also shown below the protein domains.

    BCOR takes part into one specific type of polycomb repressive complex that mediates transcriptional repression through epigenetic modifications of histones. Polycomb repressive complexes (PRCs) are molecular complexes involved in histone modification processes that are usually classified into two types: PRC1, which adds an ubiquitin moiety to histone H2A at Lys119 (H2AK119ub1), and PRC2, which catalyzes the addition of one to three methyl groups to histone H3 at Lys27, leading to H3K27me1, H3K27me2 or H3K27me3 [2]. PRCs silent a wide range of genes including HOX group genes, the first identified PRC targets [2,3]. The core protein subunits of PRC2 complex are Suz12 (with a Zinc finger domain), Eed (with a WD repeat domain that recognizes trimethylated peptides) and Ezh2 (the SET-domain containing catalytic subunit) [4]. These core components associate with different proteins that work to regulate PRC2 enzymatic activity or its recruitment to specific genomic loci.

    Conversely, all PRC1 complexes contain a core consisting of a RING1 protein (either RING1A or RING1B), which is an E3-ubiquitin ligase that adds an ubiquitin group to histone H2A at Lysine 119, and a PCGF protein (PCGF 1–6). Different PRC1 complexes are defined by the different PCGF protein that associates to the complex and directs interaction with distinct auxiliary proteins, giving rise to PRC1 complexes with differential target specificity and chromatin recruitment patterns [5]. Moreover, based on the cofactor that binds to the RING1 subunit, PRC1 complexes are often divided into ‘canonical’, ‘noncanonical’ or ‘variant’. Canonical PRC1 complexes (cPRC1) are assembled around PCGF2/4 and contain one chromobox protein (CBX2, 4, 6–8) that is able to recognize and bind the H3K27me3 histone mark implemented by the PRC2 complex [6]. Conversely, noncanonical PRC1 complexes (ncPRC1) bind to the RYBP or its paralog YAF2 and associate with PCGF1, PCGF3/5 or PCGF6 to give rise to PRC1.1, PRC1.3/1.5 or PRC1.6, respectively [4,6]. RYBP and YAF2 play a fundamental role in stimulating E3 ligase activity of PRC1 and enhancing H2AK119ub deposition [7]. Importantly, RYBP and YAF2 lack the capacity to bind to H3K27me3 and can be recruited to chromatin in cells lacking functional PRC2.

    BCOR participates in the constitution of one of the six currently described noncanonical variants of PRC1, the PRC1.1 (Table 1) [8–17]. PRC1.1’s core components include RING1A/B and PCGF1 that heterodimerize via their N-terminal RING domains (Figure 2). The RING1/PCGF1 heterodimer forms a scaffold for PRC1 assembly. PRC1.1 contains RYBP or its homolog YAF2, altogether with BCOR and other proteins (Figure 2). BCOR associates with the complex by binding to PCGF1 through its PUFD domain located at the C-terminus. Since RYBP and YAF2 are not able to bind H3K27me3, recruitment to chromatin depends on the activity of KDM2B, a H3K36 me2 histone demethylase (Figure 2) that harbors a zinc finger CxxC-binding domain involved in DNA binding that recognizes and drives targeting of PRC1.1 to nonmethylated CpG islands as observed in murine embryonic stem (ES) cells [5,18].

    Table 1. PRC 1.1 complex components.
     Principal interactionsFunctional notes
    PRC1.1 subunits  
    BCORPCGF1, KDM2B, BCL6, MLLT3, IRF8PRC1.1 core protein. It seems able to bind AF9, a protein associated with SEC complex, but the relevance of this interaction has not been determined
    BCORL1PCGF1, KDM2B, CTBP1, HDACsPRC1.1 core protein, alternatively to BCoR. It does not bind BCL-6, but the transcriptional corepressor CTBP1. Interacts with different Class II HDACs
    PCGF1BCOR, KDM2B, RYBP, YAF2PRC1.1 core protein
    KDM2BBCOR, PCGF1, SKP1, unmethylated CpG islandsPRC1.1 core protein. It demethylates H3K36me3/me2. It can participate in the formation of the SCF complex
    RING1a/RING1bPCGF1, RYBP, YAF2, CBX8Canonical and noncanonical PRC1s core protein. It ubiquitinates H2AK119
    UBP7BCOR, PCGF1, CBX8, RING1a/b, PCGF2/MEL18, PCGF4/BMI1, PTEN, MDM2, P53, DMNTDe-ubiquitinating action. It can be associated with other noncanonical PRC1 as well as the classic PRC1.
    SKP1KDM2B, other SCF proteinsUbiquitinating action. It binds KDM2B. It participates in the formation of the SCF complex
    RYBP/YAF2PGGF1, RING1b, YY1They may link PRCs to YY1
    Associated proteins  
    YY1RYBP, YAF2, DNA, INO80 complex, P53, HDACsTranscription factor. The real relevance in the in vivo PRCs recruitment is not yet determined
    CBX8RING1a/b, H3K27me3, PCGF1/2/3/5/6, PHC1/2, MLLT1It recruits the PRCs that contain it at H3K27me3 histones, establishing a functional cross-link with the PRC2s. Not present in PRC1.1
    BCL6BCOR, NCOR1, SMRT, HDACs, NuRD, MET3, DNATranscription factor. It recruits PRC1.1 by link with BCOR. It also has PRC independent activity: it is able to bind other proteins involved in gene silencing

    PRC: Polycomb repressive complex.

    Figure 2. Schematic representation of the polycomb repressive complex 1.1 model.

    The core complex is composed of the catalytic enzyme RING1A/B that forms a dimer with PCGF1 through the RING finger domains, and that deposits an ubiquitin moiety to histone H2A at Lys119 (H2AK119ub). BCOR binds to PCGF1 by means of its PUFD domain, while RYBP is bound to the RAWUL domain of RING1A/B. Recruitment to chromatin is due to KDM2B that recognizes nonmethylated CpG islands by its CXXC-binding domain. Other members of the complex are SKP1, that associates with KDM2B, and USP7, acting as a deubiquitinating enzyme.

    PUFD: PCGF Ub-like fold discriminator.

    In mammals, PRCs are recruited to target sites by multiple mechanisms, including the binding of hypomethylated CpG islands, the specific interaction with transcription factors and long ncRNAs, and the recognition of chromatin marks deposited by other histone-modifying complexes [6]. This last mechanism is at least partly responsible for the coordinated activity of PRC1 and PRC2, since it is known that the PRC2-dependent H3K27me3 mark increases affinity and recruits CBX-containing cPRC1 [19], while KDM2B-mediated recruitment to nonmethylated CpG islands of ncPRC1 drives H2AK119ub that conversely promotes binding of PRC2 [5]. However, it is also known that in some contexts PRC1.1 can act independently from PRC2; for example, in leukemic stem cells PRC1.1 was found to bind a unique set of genes independently of PRC2 and of the H3K27me3 histone mark [8].

    Not surprisingly, the genes encoding for polycomb proteins (PcG) were initially identified, along with trithorax (TrxG), as important elements for homeostasis of body development in Drosophila melanogaster. Indeed, even in mammals and humans, PcG gene germinal mutations often cause severe morphological defects and complex syndromes [20–22]. Specifically, a number of BCOR germinal loss of function mutations (>40 were previously identified) [23] induce oculo-facio-cardio-dental syndrome (OFCD) [24], inherited in an X-linked dominant mode and lethal in males [25]. The typical phenotype of OFCD syndrome is characterized by ocular defects, such as congenital cataracts and microphthalmia, craniofacial dysmorphisms, such as bifid nose tip and palatoschisis, cardiac abnormalities, such as atrial and ventricular septal defects, as well as dental abnormalities, such as radiculomegaly and oligodontia [26]. An attenuated variant of OFCD syndrome, known as Lenz’s microphthalmia, has also been described, holding a classical X-linked recessive inheritance and therefore asymptomatic in females [24].

    Nevertheless, the normal evolution of body structure during embryo development is not the only biological process that depends on proper activity of the PcG group proteins. These are also involved in the maintenance of cell identity and are necessary for correct cell differentiation [27]. Studies performed on murine ES showed that BCOR is required for the formation of primitive erythrocytes, plays a role in B- and T-cell development and is necessary for the timely expression of genes regulating ES cell pluripotency and ectodermal and mesodermal development [28]. Moreover, in ES cells, BCOR is highly expressed and required for the maintenance of the pluripotent state, since its depletion induces a robust differentiative phenotype, further supporting the idea that BCOR is a core regulator of the primed pluripotent state [29].

    Due to its central role in pluripotency maintenance, differentiation induction and cell fate determination, it is not surprising that mutations in BCOR play a central role in cancer development. This review provides an in-depth description of the different tumor types for which the pathogenetic role of BCOR seems to be particularly relevant.

    Sarcomas

    Clear cell sarcoma of the kidney

    Studies of clear cell sarcoma of the kidney (CCSK) have revealed a lot about the oncogenic role of BCOR somatic alteration on the diagnosis and biological understanding of this disease. CCSK is the second most common renal malignancy in childhood, representing 4–5% of all renal tumors [30] and accounting for approximately 20 new cases per year in the USA [31]. Although CCSK is recognized as an independent entity from Wilms’ tumor, there is still much to learn about this sarcoma.

    Microscopically, the classical pattern of CCSK presentation (91% of all cases) [31] is characterized by an architecture consisting of nests or cords of round cells with clear cytoplasm, separated by regular fibrovascular septa [32]. The extracellular matrix, rich in mucopolysaccharides, contributes to confer the ‘clear’ appearance of the tumor. However, classical architecture always accompanies at least one other pattern among the many described, albeit to a lesser extent [31,33].

    From a molecular point of view, CCSK appears as a genetically stable tumor, and over the years, the genetic alterations found were few and not recurring. A decisive step forward was made between 2015 and 2016, when five independent studies described and confirmed the recurrence of different internal tandem duplications (ITD) in the last exon of the BCOR gene, involving the PUFD domain (Figure 3A) [34–38]. These events were found in less than 75% of analyzed cases (Table 2), and many different types of ITD were identified in CCSK, all residing in exon 15 of the PUFD domain, and all preserving the protein frame. In particular, the different ITDs involved the region encompassing amino acids 1700–1755, with a minimal identified duplication of 22 amino acids up to a maximum of 38, and a minimal overlapping duplicated region covering protein positions 1725–1737 [38]. The involved region is almost always duplicated, while only in very rare cases was a partial triplication found [38]. These achievements, confirmed by subsequent works [39,40], laid the basis for the investigation of CCSK pathobiology. In fact, the features of duplication, that is strictly in-frame and associated with overexpression of the protein encoded by BCOR, suggested important speculations about the origins of CCSK.

    Figure 3. Schematic representation of different BCOR alterations, including internal tandem duplications and chimeric fusion transcripts.

    The numbers inside the boxes represent the exons, while open boxes indicate UTR regions. (A) ITD–BCOR, (B) BCOR–CCNB3, (C) BCOR–MAML3, (D) ZC3H7B–BCOR and (E) BCOR–RARA.

    ITD: Internal tandem duplication.

    Table 2. BCOR mutations in different tumor hystotypes.
    Tumor familyTumor hystotypeExamined subgroupBCORMolecular methodsRef.
       Genetic alteration %Genetic alteration type  
    SarcomasClear cell sarcoma of the kidney91% (10/11)ITD-exon 15RNA-seq/targeted RT-PCR[35]
       85% (23/27)ITD-exon 15WES/RNA-seq[32]
       9.1% (1/11)BCOR–CCNB3 fusionRNA-seq/targeted RT-PCR[35]
       83% (132/159)ITD-exon 15PCR/targeted RT-PCR[34]
       91% (20/22)ITD-exon 15RNA-seq[33]
       100% (8/8)ITD-exon 15RNA-seq[31]
       75% (3/4)ITD-exon 15RNA-seq/targeted RT-PCR/FISH[36]
       Two case reportsBCOR–CCNB3 fusionFISH[41]
       Five case reportsITD-exon 15Targeted PCR[42]
       100% (20/20)ITD-exon 15Targeted PCR[30]
     Primitive myxoid mesenchymal tumor of infancy100% (5/5)ITD-exon 15Targeted PCR[43]
       86% (6/7)ITD-exon 15RNA-seq/targeted RT-PCR/FISH[36]
       One case reportITD-exon 15Targeted PCR[42]
       One case reportITD-exon 15Targeted PCR[44]
     Undifferentiated/unclassified sarcomas: various hystologies1.5% (11/753)BCOR–CCNB3 fusionTargeted RT-PCR[45]
       14% (6/43)BCOR–CCNB3 fusionRNA-seq[46]
     Undifferentiated/unclassified sarcomas: (Ewing-like) small blue round cell or spindle cell URCS4% (24/594)BCOR–CCNB3 fusionRNA-seq[47]
       6.5% (12/184)Five BCOR–CCNB3 fusions, one BCOR–MAML3 fusion, one ZC3H7B–BCOR fusion, five BCOR-ITDRNA-seq[48]
       13% (11/87)BCOR–CCNB3 fusionRNA-seq/targeted RT-PCR/FISH[49]
       12% (2/17)BCOR–CCNB3 fusionTargeted RT-PCR/FISH[50]
       2.5% (5/200)BCOR–CCNB3 fusionTargeted RT-PCR[51]
       Ten casesBCOR–CCNB3 fusionTargeted RT-PCR[52]
       5% (2/41)BCOR–CCNB3 fusionTargeted RT-PCR/FISH[53]
       4.3% (7/164)BCOR–CCNB3 fusionTargeted RT-PCR/FISH[54]
       One case reportKMT2D–BCOR fusionRNA-seq/targeted RT-PCR[55]
      Soft tissue URCSsFour case reportsBCOR–CCNB3 fusionTargeted RT-PCR[56]
       41% (9/22)ITD-exon 15RNA-seq/targeted RT-PCR/FISH[36]
      SBRCSs lacking EWSR1, FUS, SYT and CIC gene rearrangements22% (19/86)11 BCOR–CCNB3 fusion, two BCOR–MAML3 fusions, two ZC3H7B–BCOR fusions, four other BCOR rearrangmentsFISH[57]
      BCOR–CCNB3 fusion positive sarcomas36 case reportsBCOR–CCNB3 fusionRNA-seq/targeted RT-PCR[55]
     Various hystologiesRound cell sarcoma of bone4% (24/594)BCOR–CCNB3 fusionn.a.[58]
      Soft tissue tumors1.5% (2/133)BCOR–CCNB3 fusionRNA-microarray[59]
     Endometrial stromal sarcoma29% (9/31)Five ZC3H7B–BCOR fusion, three BCOR–ZC3H7B fusion, one ITD exon 15Various[60]
       9% (3/27)Two ZC3H7B–BCOR fusion, one ZC3H7B–BCOR fusion + BCOR–ZC3H7B fusionVarious[61]
       Two case reportsOne ZC3H7B–BCOR fusion, one ZC3H7B–BCOR fusion + BCOR–ZC3H7B fusionRNA-seq/targeted RT-PCR[62]
       Three case reportsZC3H7B–BCOR fusionsTargeted RT-PCR/FISH[63]
       17 case reportsZC3H7B–BCOR fusionsTargeted RNA NGS/FISH[64]
       Three case reportsITD-exon 15Targeted PCR[65]
     Rhabdomyosarcoma8.3% (5/60)Frameshift insert/deletion, nonsenseWES/RNA-seq[66]
       7% (10/147)Seven frameshift insert/deletion, one nonsense, two focal homozygous deletionWGS/WES/RNA-seq[67]
      Anaplastic RMSOne case reportFrameshift insertionTargeted PCR[68]
      PAX-fusion negative RMS9.5% (9/94)Six frameshift insert/deletion, one nonsense, two focal homozygous deletionWGS/WES/RNA-seq[67]
      PAX-fusion positive RMS1.9% (1/53)Frameshift insert/deletionWGS/WES/RNA-seq[67]
     Ossifying fibromyxoid tumor2.5% (1/39)ZC3H7B–BCOR fusionRNA-seq[69]
    CNS neoplasm:CNS neoplasms‘CNS-HG-NET-BCOR altereted’ cluster of methylation tumor79% (15/19)ITD-exon 15DNA and RNA NGS/targeted PCR[70]
       Five case reportsITD-exon 15Targeted PCR[42]
       One case reportITD-exon 15Targeted PCR[71]
     Medulloblastoma2% (4/189)One nonsense, three frameshift insert/deletionVarious[72]
       2.2% (1/46)NonsenseWGS/WES[73]
       1.6% (2/125)Frameshift insert/deletionWGS/WES[74]
       3% (3/92)Nonsense, frameshift insert/deletionWES[75]
      Sonic-hedgehog-driven MB7% (4/58)One nonsense, three frameshift insert/deletionVarious[72]
       5.2% (7/133)Missense, nonsense, frameshift insert/deletionWGS/WES[76]
      Sonic-hedgehog-driven MB in infants (age <3)8% (4/50)n.a.WGS/WES[76]
      Sonic-hedgehog-driven MB in children (age 3 ≤ × <18)3% (1/33)n.a.WGS/WES[76]
      Sonic-hedgehog-driven MB in adults (age ≥18)4% (2/50)n.a.WGS/WES[76]
     Retinoblastoma10% (7/71)One missense, two nonsense, two frameshift insert/deletions, two large deletionsWES[77]
       13% (6/46)Frameshift insert/deletion, large deletionWGS/SNP array[78]
       4.2% (4/94)Focal deletionWGS/SNP array[79]
     Gliomas, various hystologiesRecurrent high-grade astroblastoma25% (1/4)Frameshift insert/deletionTargeted DNA NGS/array CGH[80]
      pGBM and DIPG4,3% (14/326)Nonsense, frameshift insert/deletionWGS/WES/RNA-seq[81]
      Diffuse glioma14% (1/7)MissenseWES[82]
    Hemolymphopoietic system tumorsMyeloid neoplasmMDS5.3% (6/114)Nonsense, frameshift insert/deletion, splice siteWES[83]
       2.8% (2/71)Deletion (Xp11.4)Targeted DNA NGS/array CGH[84]
       4.2% (40/944)VariousTargeted DNA NGS/array CGH[85]
       7% (2/29)Nonsense, frameshift insert/deletionWES[86]
       4.2% (15/354)Eight frameshift insert/deletion, five nonsense and two splice siteTargeted PCR[87]
      MDS with multilineage dysplasia8% (2/25)Frameshift insertion, splice siteWES[83]
      MDS with excess blasts5.4% (2/37)Frameshift deletion, nonsenseWES[83]
      AML with chromosome 11 trisomy4% (1/23)n.a.Targeted DNA NGS[88]
      AML with chromosome 13 trisomy25% of 34n.a.WES/targeted DNA NGS[89]
      Cohesin-altered myeloid neoplasm11.4% (14/123)n.a.WES/targeted DNA NGS[90]
      Cohesin-WT myeloid neoplasm5% (47/937)n.a.WES/targeted DNA NGS[90]
      Unselected AML8% (72/494 adult, vs 3/179 in children)n.a.DNA and RNA NGS[91]
       5% of 143n.a.WES/targeted DNA NGS[92]
       8.7% (58/664)n.a.Targeted DNA NGS[93]
      Primary AML in adults3% (6/197)n.a.Targeted DNA NGS[94]
       1.6% (4/247)n.a.WES / Targeted DNA NGS[92]
       5% (83/1603)n.a.Targeted DNA NGS[95]
       5% (19/377)Missense, nonsense, frameshift insert/deletion, splice siteTargeted DNA NGS[96]
      Aged ≤65 with intermediate cytogenetic prognosis15.2% (7/46)n.a.Targeted DNA NGS[96]
      AML with balanced chromosomic rearrangements8% (18/224, but 38% in cases with inv(3)(q21q26.2/t(3;3)(q21q26.2))n.a.Targeted DNA NGS[95]
      AML with unbalanced chromosomic rearrangements9% (31/349)n.a.Targeted DNA NGS[95]
      Secondary AML7.2% (16/221)n.a.WES/targeted DNA NGS[92]
      RUNX1-mutated18% (29/163)n.a.Targeted DNA NGS[97]
       10.8% (13/1381)n.a.Targeted DNA NGS[98]
      Normal karyotype AML2.5% (1/40)Focal deletionTargeted DNA NGS[99]
       4.2% (10/262)n.a.Targeted PCR[100]
       4% (27/716)n.a.Targeted DNA NGS[95]
      Normal karyotype17.1% (14/82)Frameshift insert/deletion, nonsense, splice siteTargeted PCR[100]
      Pediatric normal karyotype AML6.3% (3/48)MissenseTargeted PCR[101]
      Pediatric AML1.1% (2/182)One frameshift insert/deletion, one splice siteWES/targeted DNA NGS[102]
       1.1% (4/369)n.a.Targeted PCR[103]
       5% (2/40)Frameshift insert/deletion, nonsenseTargeted DNA NGS/SNP array[104]
       4.8% (4/83)MissenseTargeted PCR[101]
      Acute promyelocytic leukemiaOne case reportBCOR–RARα5′-RACE-PCR[105]
       One case reportBCOR–RARαTargeted PCR[106]
      CMML10% (15/150)n.a.WES/targeted DNA NGS[107]
       7.4% (3/54)One frameshift insert/deletion, three nonsenseTargeted PCR[87]
       7.7% (2/26)Frameshift insertion, splice siteWES[83]
     Lymphoid neoplasmExtranodal NK/T-cell lymphoma nasal type17% ([5/30] or 20.6% [7/34] including cell lines)Two missense, two frameshift insertion/deletion, three nonsenseWES/targeted DNA NGS/RNA-seq[108]
       32% (8/25)Frameshift insert/deletion, large deletion, nonsense, splice siteTargeted DNA NGS[109]
       21% (24/113 including cell lines)n.a.Targeted DNA NGS[109]
      T-cell prolymphocytic leukemia9% (2/23)MissenseTargeted DNA NGS[110]
       8% (4/51)Deletion (Xp11.4, 50%)Targeted DNA NGS/array CGH[111]
      Chronic lymphocytic leukemia3% (4/149)Three frameshift insert/deletion, one nonsenseWES/SNP array[112]
       1.6% 10/643n.a.Targeted DNA NGS[113]
       6.3% (3/48)Missense, frameshift insert/deletionTargeted DNA NGS[114]
       1.2% (5/428)Nonsense, frameshift insert/deletionWGS/WES/RNA-seq/array CGH[115]
      Splenic diffuse red pulp lymphoma24% (10/42)Three frameshift insert/deletion, two nonsense, one splice site, four large deletionsWES/targeted DNA NGS/array CGH[116]
     OtherMixed-phenotype acute leukemia8.3% (1/12)n.a.Targeted DNA NGS[117]
    CarcinomasSalivary glands cancer, various hystologiesRecurrent and metastatic SGC8% (4/50)MissenseTargeted DNA NGS[118]
     Adenoid cystic carcinomaMetastatic ACCs60% (3/5)Frameshift insertion, nonsenseWGS/RNA-seq[119]
     Endometrial carcinomaPOLE-negative nonultramutated endometrioid endometrial carcinoma13% (10/76)Missense, splice siteWES/targeted DNA NGS[120]
     Gynecologic carcinosarcoma n.a.n.a (missense N1459S 2 time recurrent)WES[121]
     Thymoma and thymic carcinomaB3 thymoma50% (3/6)Missense, frameshift insert/deletionWES[122]
       One case reportFrameshift insert/deletionWGS/RNA-seq/array CGH[123]

    Only the techniques used in the individual studies to establish BCOR mutational status have been reported.

    5′-RACE PCR: 5′-Rapid amplification of cDNA ends polymerase chain reaction; ACC: Adenoid cystic carcinoma; AML: Acute myeloid leukemia; Array CGH: Array comparative genome hybridization; CMML: Chronic myelo-monocytic leukemia; DIPG: Diffuse intrinsic pontine glioma; FISH: Fluorescent in situ hybridization; ITD: Internal tandem duplication; MB: Medulloblastoma; MDS: Myelodysplastic syndrome; n.a.: Not available; NK: Natural killer; NGS: Next-generation sequencing; pGBM: Pediatric glioblastoma; RNA-seq: RNA sequencing; RT-PCR: Real-time PCR; SGC: Salivary glands cancer; URCS: Undifferentiated round-cell sarcoma; WES: Whole exome sequencing; WGS: Whole genome sequencing; WT: Wild type.

    There have been several attempts to attribute biological significance in terms of gain or loss of function to the ITD–BCOR; however, until now, no predicted structural model of the BCOR protein carrying the ITD has been analyzed and released, even though protein–protein interaction studies suggested that ITD–BCOR might represent an hypomorphic allele with altered PCGF1-binding affinity [13]. Currently, we are still far from having conclusive results on the question, and even though there is evidence supporting a loss-of-function effect of ITD-BCOR, (overexpression of Cyclin D1 [43,44], of PRC2 targets and of several classes of HOX genes [36]), the lack of any frameshift or nonsense mutation in this setting is instead in favor of a gain-of-function event.

    Primitive mesenchymal myxoid tumor of infancy

    Primitive mesenchymal myxoid tumor of infancy (PMMTI) is a newly introduced myofibroblastic pediatric tumor [124]. It is an intermediate grade mesenchymal neoplasm with a high local recurrence rate, but with low metastatic potential [47]. Histologically, it is composed of cells that are fused in a myxoid matrix. Morphologically, differential diagnosis is warranted with respect to other stromal tumors, such as congenital infant fibrosarcoma, fibromyxoid sarcoma, myofibrosarcoma and dermatofibrosarcoma protuberans [47], from which it clearly diverges from the immunophenotypical pattern and specific molecular features. Actually, few studies that investigated PMMTI at a molecular level have found the presence of ITD–BCOR (Figure 3A) associated with the overexpression of BCOR in almost all the cases analyzed [40,52,125], similarly to CCSK. Recently, Kao et al. demonstrated that these two tumors also show a very similar transcriptional signature that, together with epidemiological, morphological and genetic overlap, suggests the possibility that PMMTI is the equivalent of CCSK arising in soft tissue [40].

    Small round blue cell sarcomas

    The typical pediatric tumors of Ewing sarcoma family can be molecularly distinguished by the presence of translocations involving the EWSR1 gene and one of ETS/FLI family genes. These tumors are morphologically distinct per their microscopic appearance consisting of a bed of round monomorphic cells with regular nuclei and dispersed chromatin, with or without the presence of rosettes [45]. However, progressive diffusion of molecular profiling and high-throughput sequencing technologies has allowed the identification of an increasing number of molecular alterations in undifferentiated sarcomas that are morphologically similar or indistinguishable from Ewing’s sarcoma, but lacking the aforementioned molecular features. The need to set up a new classification scheme for these tumors led to the use of the term ‘Ewing-like small round-blue-cell sarcoma’ (SRBCS), although the specific molecular entities belonging to this subgroup often present atypical, undifferentiated or with nonspecific histology (undifferentiated round cell sarcomas [URCS]). Among the soft tissue URCS, Kao et al. have shown that nearly half of them carry ITD–BCOR [40], whereas Pierron et al. identified a group characterized by the presence of the BCOR–CCNB3 fusion gene (Figure 3B) and overexpression of the resulting chimeric protein [46]. These tumors showed a transcriptional signature different from pediatric tumors of Ewing sarcoma family (they were negative for BCOR immunostaining [54]) and a broader morphological spectrum [46,56], which is why the classification of Ewing-like sarcomas appears reductive. Other works have frequently found cases of BCOR–CCNB3 positive sarcomas, in variable percentages depending on the group of tumors analyzed, but in any case definitely consistent (Table 2) [40,48–51,53,55–59,126–129]. CCNB3 encodes for cyclin B3, a protein with promeiotic function in which its expression is normally limited to the testis. CCNB3 activity in the chimeric protein (evaluated through cellular models of engineered fibroblasts) suggested a possible oncogenic driver role due to the observed ectopic expression of CCNB3 in this type of tumors, although further evaluations are needed to assess the role of BCOR in the chimeric protein, as well as any potential new activity acquired from the fusion of the two proteins [46].

    Regarding ‘Ewing-like’ SRBCS, Specht et al. [55] described, in addition to the BCOR–CCNB3 fusion gene, the presence of distinct rearrangements of BCOR in eight cases: two cases carrying BCOR–MAML3 fusion (Figure 3C), two carrying ZC3H7B–BCOR (Figure 3D) and four with internal rearrangements of BCOR. In the same study, the increased expression of HOX genes was reported in one of the two cases with BCOR–MAML3 fusion, suggestive of a loss of function of BCOR and PRC activity. MAML3 encodes a member of the mastermind-like (MAML) family of transcriptional coactivators that constitutes a component of the Notch signaling pathway, with roles in biological processes such as cell proliferation, differentiation and survival. The BCOR–MAML3 fusion transcript contains exons 2-5 of MAML3, thus retaining the transactivation domain, but losing the Notch-binding site of MAML3. ZC3H7B encodes a nuclear protein containing a domain able to interact with the rotavirus NSP3 36 protein [130], as well as several other domains involved in multiple protein interactions and nucleic acid/protein interactions. Little can be said at present about the ways in which these two genes exert their oncogenic roles in SRBCS, since their activity in the context of chimeric proteins containing BCOR requires further study. However, BCOR is involved in its entirety in the BCOR–MAML3 fusion and in its terminal 3′ portion only in the ZC3H7B–BCOR chimeric transcript. The recurrence of these two fusion genes in SRBCS was also confirmed in the most recent study by Watson et al. [126], which highlighted the existence of SRBCS characterized by the presence of ITD–BCOR (Table 2). An extremely interesting result of this last study is that all SRBCS-carrying BCOR alterations generate a common cluster at the transcriptional level, thus reinforcing a view in which BCOR alterations, either ITD or rearrangements, exert a common pathogenic pathway leading to abnormal activity of the PRC1.1 complex. In fact, as in CCSK, a significant enrichment of gene sets regulating morphogenesis, differentiation of neurons and tyrosine kinase receptors was observed, and HOX genes are overexpressed. In the same study, the authors noticed an overexpression of SMARCA2, a component of the chromatin modifier SWI/SNF complex, and its activity was opposite to that of the PRC1 and PRC2 complexes [67], with observed strict association and functional linkage [68]. This insight may suggest that PRCs loss-of-function caused by BCOR alterations can lead to increased SWI/SNF complex activity. This hypothesis is reinforced via in vitro work in which the codeletion of EZH2 reverts the oncogenic phenotype caused by the loss of function of the SMARCB1 gene [66], similarly to SMARCA2, which encodes for a subunit of the SWI/SNF complex. Recently, Kao et al. [51] also identified a single case of URCS harboring the KMT2D–BCOR fusion gene.

    Rhabdomyosarcoma

    BCOR’s involvement appears to also be relevant in rhabdomyosarcoma (RMS), another typical blue round cell malignant childhood tumor. Histologically, the two major RMS subtypes are alveolar RMS and embryonal RMS, both of which have distinct molecular and clinical hallmarks [131]. Alveolar RMS results in poor prognosis and is genetically defined by the presence of a fusion involving PAX3 or PAX7 genes, two transcription factors. The embryonal RMS subtype typically affects younger children and is generally comprised of PAX-negative tumors. A rare subtype of RMS is anaplastic, where a single case was recently reported to carry a BCOR alteration, together with mutation in ARID1A, a member of the SWI/SNF family, and of SETD2, a histone methyl-transferase [61]. Two studies [131,132] confirmed that BCOR mutations in RMS recur in about 7–8% of total cases, with an apparent major involvement in PAX-negative RMS, for which the percentage nears 10% of cases (Table 2).

    Endometrial stromal sarcoma

    Endometrial stromal sarcoma (ESS) is a gynecological sarcoma composed of cells that resemble those of the endometrial stroma in the proliferative phase. The WHO classification recognizes four distinct entities [133,134]: benign stromal endometrial nodules; low-grade ESS genetically characterized by various translocations among which the most frequent involves JAZF1 and/or PHF1; high-grade ESS generally carrying YWHAE–NUTM2 fusion gene [135]; and undifferentiated endometrial sarcoma with a complex karyotype.

    In particular, the involvement of the YWHAE–NUTM2 fusion gene is noteworthy since this was the first genetic alteration associated with CCSK [136], as well as the second in order of frequency (with a percentage between 0 and 12% of cases in various studies) [34–38,63,137,138]. YWHAE genes (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein) encode different variants of the 14-3-3 protein family, which includes seven highly conserved and ubiquitously expressed proteins that seem to play a role in modulation of cytoskeletal organization, metabolism, differentiation and proliferation [62]. NUTM2 genes of unknown function belong instead to the FAM22 family, which was more recently renamed NUTM2 because of sequence homology to the NUT gene locus that is involved in the oncogenesis of malignant tumor of midline [64].

    Several patients with ZC3H7B–BCOR and/or BCOR–ZC3H7B [60,65,134,139] fusion genes were also described and initially included among low-grade ESS; however, it was recently proposed to identify ZC3H7B–BCOR ESS as an independent subgroup of high-grade ESS (Table 2) due to new prognostic evidence and myxoid leiomyosarcoma-like histologic appearance [65]. Recently, on subsets of 31 various grade ESS, Mariño-Enriquez et al. identified three cases carrying ITD–BCOR, one defined as high-grade ESS and two as undifferentiated ESS, all three cases holding typical immunophenotypic features of ITD–BCOR-positive tumors, such as the overexpression of cyclin D1 and BCOR [70].

    This description limited to the mesenchymal tumor histotypes carrying BCOR-related abnormalities clearly underlined that the evident morphological and molecular overlaps between CCSK and PMMTI, as well as the common recurrence of ITD–BCOR, are not the only similarities found among these tumor histotypes. In fact, the YWHAE–NUTM2 fusion gene, typical of some CCSKs, seems to be the driver element of high-grade ESS subgroup. Conversely, low-grade ESS is often characterized by the involvement of BCOR as a fusion gene with ZC3H7B. The absence of substantial histological differences between ITD–BCOR-positive CCSK and those carrying YWHAE–NUTM2, together with the simultaneous presence of BCOR molecular anomalies and YWHAE–NUTM2 fusion in the ESS, as well as the evidence that various BCOR aberrations (such as ITD, ZC3H7B–BCOR and BCOR–CCNB3) lead to the overexpression of BCOR and of cyclin D1 [34–36,40,41,43,44,52,69,70,128,140] are all elements that reinforce the hypothesis that these alterations activate a common pathogenetic pathway, or at least a partially shared one.

    It is also interesting that the link between CCSK and SRBCS due to the common involvement of BCOR was also strengthened by the findings of YWHAE–NUTM2B/E and ITD–BCOR in a group of URCS of soft tissue, the last previously identified also in SRBCS [126], as well as by the description of three cases of CCSK carrying the BCOR–CCNB3 fusion gene [77].

    Another example of biological intersection between these tumors is the finding of ZC3H7B–BCOR fusion typical of low-grade ESS in some cases of SRBCS [55] and in one case of ossifying fibromyxoid tumor [78], a rare soft tissue tumor.

    CNS neoplasms

    BCOR is hypothesized to have an active role in neuronal development. In fact, BCOR expression patterns in several tissues during the various phases of embryonic development, conducted by Wamstad and Bardwell on mouse models, revealed intense BCOR expression in the neural tube and retina during development [79].

    Therefore, it is not surprising that several CNS tumors carry BCOR alterations (Table 2). Mutations with supposed loss of function (e.g., nonsense, frameshift, splice sites and deletions) have been identified in up to 13% of retinoblastomas [80–82]; between 1 and 14% of various glial tumors, particularly in high-grade tumors [72–74] and also in pediatric patients [73,140], with a 25% peak found in recurrent high-grade astroblastoma [74]. Moreover, BCOR genetic alterations also appear in 2–8% of medulloblastomas [42,75,76,141,142], and in particular, such aberrations seem to be more represented in SHH-driven cases, especially in infant patients [42].

    CNS-HGNET-BCOR

    Primary CNS neuroectodermal tumors (CNS-PNET) are highly malignant neoplasms that predominantly affect children but may also occur in adolescents and adults. Histologically, CNS-PNETs are characterized by poorly differentiated embryonic cells, with a propensity both for glial and neuronal differentiation [143]. The WHO classification of 2016 [71] has removed CNS-PNET as an independent group of tumors, grouping them in a class of ‘embryonic tumors’ with medulloblastoma, as an example. In the attempt to establish new classification criteria to better define the molecular nature of CNS-PNET, Sturm et al. [140] proposed to evaluate a large series of CNS tumors via genomic methylation patterns. From this, new molecular entities emerged, clustering in well-defined methylation subgroups, but often lacking histological homogeneity. One of these new histologically nonhomogeneous entities was shown to carry in the vast majority of cases ITD–BCOR alteration. This group of tumors, defined CNS-HGNET-BCOR (high-grade primitive neuroectodermal tumors of the CNS with alterations of BCOR), showed a remarkable overexpression of BCOR similar to what is found in other tumors-carrying ITD-BCOR (Table 2). Curiously, as reported by Santiago et al. [52], the triad ‘CCSK/PMMTI/CNS-HGNET-BCOR’, after the group ‘rhabdoid tumor of the kidney/extra-renal rhabdoid tumor of soft tissues/atypical teratoid-rhabdoid tumor of the CNS’, would constitute the second trio of tumors joined by molecular anomalies and kidney, soft tissues, and brain districts. It is however necessary to highlight that Yoshida et al. recently reported important immunophenotypical differences between CNS-HGNET-BCOR and CSSK/PMMTI [144].

    In addition to the recurrence of ITD–BCOR, CNS tumors and mesenchymal tumors with BCOR abnormalities also share common characteristics regarding transcriptional regulation. In fact, WNT and in particular SHH pathways, wherein up-regulation had already been reported as a typical feature of CCSK [145], were also found up-regulated in a CNS-HGNET-BCOR patient [146], as well as in many cases of retinoblastoma [147] and even represented the driver element of two subgroups of medulloblastoma [148]. The recurrence of the deregulation of these signaling pathways in at least two entities carrying ITD–BCOR supports the hypothesis that such deregulation may actually affect all ITD–BCOR-positive tumors, and it can be envisaged as a potential therapeutic target [146].

    It is also interesting to note that, like mesenchymal tumors, also among CNS tumors with BCOR involvement, there are numerous cases arising in the pediatric setting. This evidence can indicate that, coherently with the functions of BCOR at the embryonic level, mutations in this gene lead to a premature disruption of the differentiation pathway in progenitor stem cells. However, further studies are necessary to support this hypothesis.

    Hemolymphopoietic system neoplasms

    There is evidence regarding the importance of BCOR for physiological hematopoiesis. Mutations of this gene in knockout organisms result in hematological abnormalities, consisting in functional deficiency of primitive erythrocytes and lymphocyte depletion, and confirming the relevance of BCOR on the activity of BCL-6, a known mitogenic agent for lymphoid cells [28]. Conversely, the loss-of-function of BCOR in murine bone marrow cells produces significantly enhanced proliferation and myeloid differentiation rates with upregulated expression of HOX genes [84]. Even if the introduction of next-generation sequencing (NGS) techniques for the molecular screening of large cohorts of patients affected by hemolymphopoietic system neoplasms highlighted the clear involvement of BCOR alterations in these diseases, at present it is still challenging to clarify the biological role of BCOR in myeloid and lymphoid precursors and to translate this molecular knowledge for clinical applications. Actually, there are still few reports aimed at clarifying the role of BCOR in lymphoid and myeloid oncogenesis [85,86]. Tanaka et al. generated transgenic mice harboring exon 4 deletion leading to loss of the BCL-6-binding domain, which developed lethal acute T-cell lymphoblastic leukemia in a Notch1-dependent manner and demonstrated myc upregulation [85]. Another report by Lefebure et al. [86] showed that BCOR is a tumor suppressor in the Eμ-myc lymphoma murine model and its loss-of-function mutations act as myc-cooperating events in this setting.

    Myeloid neoplasms

    The involvement of BCOR alterations in clonal disorders of myeloid lineage (Table 2) has been assessed primarily for myelodysplasias [83,87,91–93] and acute myeloid leukemia (AML) [94,102–104], with cases ranging from less than 1–10% in unselected cohorts. By analyzing these and other cohort studies, it was possible to identify clinical contexts where the presence of BCOR alterations is most frequent. Particularly in AML, it is clear that the frequency of BCOR mutations is significantly higher in older patients than in pediatric ones [90,94,96,101], with pediatric AMLs with normal karyotype as the exception [88] and higher in secondary AML with respect to primary AML [102]. NGS techniques allow for the identification of new molecular subgroups potentially related to specific clinical characteristics, in addition to those already identifiable by cytogenetic studies. In the context of these specific entities, association with BCOR comutations (Table 2) is particularly relevant in the following instances: intermediate cytogenetic prognosis and FLT3–ITD-negative AML in adult patients under 65 [89], nonselected myeloid neoplasms with mutations in cohesin-coding genes [95], AML with changes associated with myelodysplasia [92], AML with trisomy 11 or 13 [99,100], AML with balanced or unbalanced chromosomal rearrangements [97], AML with normal karyotype [97,98,107] (in particular, pediatric AML) [88] or if co-occurring mutations in NPM1, CEBPA, FLT3-ITD, MLL-PTD or RUNX1 are present [105,107,117].

    The high percentage of BCOR involvement in patients with chronic myelo-monocytic leukemia [92,93,106] or with acute leukemia with mixed phenotype is also noteworthy [149]. Lastly, two cases of a variant of acute promyelocytic leukemia have been described for which BCOR represented the partner of RARα (the transcription factor typically involved in APL) in the chimeric gene (Figure 3E) [150,151].

    BCOR alterations found in myeloid clonal diseases are apparently clustered around exon 4 and for the most part are mutations with presumed loss-of-function. This consideration derived both from the analysis of the type of alterations found and the evaluated expression of HOX genes, the target of PRC activity when these genes are strongly overexpressed. The prognostic significance of the presence of BCOR mutations in myeloid clonal diseases was also evaluated. Damm et al. identified the presence of mutated BCOR as an independent negative prognostic factor in MDS [92]. Regarding adult AML, Terada et al. identified BCOR mutations as unfavorable prognostic factors in 5-year overall survival (8.7% BCOR mutated vs 34.0% BCOR WT) [89].

    Finally, BCOR alterations have been identified and/or described in several studies conducted on acquired aplastic anemia patients, in percentages similar to those described for nonselected MDS and AML [110–112,152,153]. This suggested the possibility that these mutations may contribute to clonal selection during the onset of myeloid malignancies on the aplastic anemia background.

    Lymphoid neoplasms

    In the lymphoid line, there is evidence of a significant presence of BCOR genetic alterations, seemingly all causing loss-of-function of the corresponding protein. BCOR alterations have been identified in up to 9% of prolymphocytic T leukemias [114,115], chronic lymphocytic leukemias [108,109,113,116] and, respectively, in 24% and up to 32% of the evaluated cases of diffuse splenic lymphoma of the red pulp [122] and of extranodal NK/T-cells lymphoma, nasal type [118,123]. The latter one is an aggressive lymphoma, strongly associated with EBV infection, which arises primarily, but not exclusively, in nasal and paranasal areas. Considering the high recurrence of mutation and inferences deriving from murine models, it is clear that a wider evaluation of BCOR involvement in these tumors is warranted.

    Carcinomas

    The functional studies conducted on BCOR showed that the activity of this gene primarily affects cells committed to the mesenchymal and neuroectodermal lineages. In fact, knockout organisms have shown alterations, especially in tissues involved in OFCD syndrome, as well as important hematological abnormalities [28]. Analysis of BCOR expression in various tissues during murine embryogenesis also confirmed strong expression in the eye, limb buds and branchial arches, neural tube, adnexal and nervous system tissues, and craniofacial structures [79]. These studies are partly strengthened by the analysis of tumor histotypes associated with BCOR mutations. The most represented tumors are stromal tumors, CNS neoplasms and hemolymphopoietic system tumors, sometimes concentrated in the head and neck or upper chest districts in organs such as the thymus [119] and salivary glands [120]. In the context of thymic tumors, it is particularly interesting that BCOR mutations recur in 50% of cases of aggressive B3-thymoma [121]. Considering the high percentage of recurrence, an active role of BCOR in the pathogenesis of this thymoma subtype, as well as the potential use of this gene as a classifying marker, is hypothesized. As for salivary glands tumors, the occurrence of BCOR mutations seems to characterize more aggressive diseases, regardless of the histological type. This is particularly true of adenoid cystic carcinoma, which is a rare type of cancer that most frequently occurs in salivary glands, but which can also be found in breast, lacrimal gland, lung, Bartholin’s gland, trachea and paranasal sinuses [154]. While definitive publications on the recurrence and role of BCOR mutations in various epithelial malignancies are still lacking, it is evident from comprehensive sequencing efforts (https://portal.gdc.cancer.govwww.cbioportal.org) that BCOR abnormalities are recurrent in uterine corpus endometrial carcinoma, colon and stomach adenocarcinoma, and lung tumors regardless of histology. In the context of The Cancer Genome Atlas project, in the EBV + gastric carcinomas subgroup, the percentage of mutations involving BCOR reached 23%. All mutations were nonsense mutations, therefore suggesting a complete loss-of-gene function. The high involvement of BCOR mutations in EBV + gastric carcinomas is emphasized by the consideration that BCOR also affects another EBV-related tumor: extranodal NK/T-cell lymphoma, nasal type.

    A special case is that of the uterus because in this organ, BCOR alterations are recurrently involved both in epithelial tumor histotypes (in this case, endometrioid endometrial adenocarcinoma) [155] and in uterine carcinosarcoma (involving BCOR in 23% of cases) [156], in addition to the previously described ESS.

    Conclusion

    Somatic alterations of BCOR are found in many tumor subtypes, with fusion genes discovered in rare subclasses of sarcomas, internal tandem duplications (ITD) in almost all pediatric clear cell sarcomas of the kidney, in a subgroup of undifferentiated pediatric sarcomas (undifferentiated round cell sarcoma of infancy, primitive myxoid mesenchymal tumor of infancy), in endometrial stromal sarcoma and in the subset of high-grade neuroepithelial tumor of the central nervous system with BCOR gene alteration (CNS HGNET-BCOR). Additionally, loss-of-function mutations were also found in myelodysplasias and acute myeloid leukemias and in selected subtypes of lymphoid neoplasms. A deeper knowledge of the functional role of different BCOR alterations in tumors of the mesenchymal, hematopoietic and neuroectodermal lineages will allow the identification of actionable pathways in these malignancies.

    Future perspective

    Deeper molecular knowledge of oncology enables better identification of diagnostic or prognostic markers and/or therapeutic targets. The recent introduction of NGS techniques has exponentially amplified the potential to obtain such data and to compare and find similarities among the molecular landscapes of histologically distant tumor types. This review has shown that BCOR represents one of the possible genes and a viable factor to focus our efforts. In fact, the recognition of recurrent alterations of this gene in tumors such as CCSK, PMMT1, SRBCS and CNS-HGNET-BCOR already plays a key role in the diagnosis of these malignancies and there are many examples of new entities wherein BCOR plays a relevant clinical role. Further work will be necessary to dissect the activity of different BCOR mutations in specific cell contexts, and mechanistic and functional studies are warranted to understand the overall effects of these alterations on PRC1.1 function and target recruitment.

    We believe that a better knowledge of PRC activity and the epigenetic regulators in which BCOR is an important component can lead to the advancement of new therapies.

    Executive summary

    BCOR is an epigenetic regulator

    • BCOR is a transcriptional corepressor, wherein the gene is located on the Xp11.4 locus.

    • It binds BCL-6 and takes part in the polycomb repressive complex (PRC) 1.1.

    • In the PRC1.1 complex, BCOR binds to PCGF1 through its PCGF Ub-like fold discriminator domain.

    • PRC1.1 silences genes through ubiquitination of Lys119 in histone H2A (H2AK119).

    • BCOR germinal loss-of-function mutations determine oculo-facio-cardio-dental syndrome.

    • BCOR is necessary for regulation of embryonic stem cell pluripotency and ectodermal and mesodermal development.

    Involvement of BCOR in sarcomas

    • BCOR internal tandem duplications (ITD) are present in more than 75% of CCSK, a pediatric renal sarcoma.

    • ITD-BCOR are characteristic of almost all primitive mesenchymal myxoid tumor of infancy.

    • Soft tissue undifferentiated round cell sarcomas are characterized by ITD-BCOR or BCOR–CCNB3 fusion gene.

    • Other fusions found in Ewing-like small blue–round-cell sarcoma involve BCOR–MAML3 or ZC3H7B–BCOR.

    • BCOR mutations recur in 10% of PAX-negative rhabdomyosarcomas.

    • ZC3H7B–BCOR or ITD-BCOR characterize a subgroup of endometrial stromal sarcomas.

    BCOR in CNS tumors

    • Loss-of-function BCOR mutations are found in retinoblastomas, high-grade glial tumors and medulloblastomas.

    • A subgroup of central nervvous system high-grade primitive neuroectodermal tumors with alterations of BCOR (CNS-PNET) is characterized by ITD-BCOR.

    • Soft tissue undifferentiated round cell sarcomas are characterized by BCOR-ITD or BCOR–CCNB3 fusion gene.

    • Other fusions found in Ewing-like small blue round cell sarcoma involve BCOR–MAML3 or ZC3H7B–BCOR.

    BCOR alterations in hemolymphopoietic system tumors

    • Loss-of-function mutations of BCOR are present in around 10% of unselected acute myeloid leukemia and myelodysplastic syndrome.

    • BCOR mutations are unfavorable prognostic factors in myelodysplastic syndrome and adult acute myeloid leukemia.

    • Prolymphocytic T leukemias and chronic lymphocytic leukemias carry BCOR mutations in less than 10% of cases.

    • Up to 20–30% of diffuse splenic lymphoma of the red pulp and extra-nodal NK/T-cells lymphoma, nasal type carry BCOR alterations.

    BCOR involvement in carcinomas

    • BCOR mutations are found in aggressive B3-thymomas, adenoid cystic carcinoma, uterine corpus endometrial carcinoma, EBV + gastric carcinomas, colon and stomach adenocarcinoma, and lung tumors, regardless of histology.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0

    Author contributions

    A Pession conceived the study and revised the paper; M Fiore and A Astolfi drafted the manuscript and collected and interpreted the data; F Melchionda, V Indio and SN Bertuccio participated in planning and revising the manuscript. All the authors read and approved the final manuscript.

    Acknowledgments

    The authors thank Quaglia A, Paltrinieri S, Nanni S and Serafini S for their contribution in data collection.

    Financial & competing interests disclosure

    This work was supported by fund donation in memory of Maestro Claudio Abbado and by Associazione Margherita Onlus. The authors have no other relevant affiliation or financial involvement with an organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

    Funded writing assistance was utilized in the production of this article from Enago. No other writing assistance was utilized.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Huynh KD, Fischle W, Verdin E, Bardwell VJ. BCOR, a novel corepressor involved in BCL-6 repression. Genes Dev. 14(14), 1810–1823 (2000).
    • 2. Blackledge NP, Rose NR, Klose RJ. Targeting polycomb systems to regulate gene expression: modifications to a complex story. Nat. Rev. Mol. Cell Biol. 16(11), 643–649 (2015).
    • 3. Junco SE, Wang R, Gaipa JC et al. Structure of the polycomb group protein PCGF1 in complex with BCOR reveals basis for binding selectivity of PCGF homologs. Structure 21(4), 665–671 (2013).
    • 4. Chittock EC, Latwiel S, Miller TCR, Müller CW. Molecular architecture of polycomb repressive complexes. Biochem. Soc. Trans. 45(1), 193–205 (2017).
    • 5. Blackledge NP, Farcas AM, Kondo T et al. Variant PRC1 complex-dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell 157(6), 1445–1459 (2014).
    • 6. Schuettengruber B, Bourbon H-M, Di Croce L, Cavalli G. Genome regulation by polycomb and trithorax: 70 years and counting. Cell 171(1), 34–57 (2017).
    • 7. Rose NR, King HW, Blackledge NP et al. RYBP stimulates PRC1 to shape chromatin-based communication between polycomb repressive complexes. Elife 5, e18591 (2016).,
    • 8. van den Boom V, Maat H, Geugien M et al. Non-canonical PRC1.1 targets active genes independent of H3K27me3 and is essential for leukemogenesis. Cell Rep. 14(2), 332–346 (2016).
    • 9. Béguelin W, Teater M, Gearhart MD et al. EZH2 and BCL6 cooperate to assemble CBX8–BCOR complex to repress bivalent promoters, mediate germinal center formation and lymphomagenesis. Cancer Cell. 30(2), 197–213 (2016).
    • 10. Koh DI, Choi WI, Jeon BN, Lee CE, Yun CO, Hur MW. A novel POK family transcription factor, ZBTB5, represses transcription of p21CIP1 gene. J. Biol. Chem. 284(30), 19856–19866 (2009).
    • 11. Lanzuolo C, Orlando V. Memories from the polycomb group proteins. Annu. Rev. Genet. 46(1), 561–589 (2012).
    • 12. Gearhart MD, Corcoran CM, Wamstad JA, Bardwell VJ. Polycomb group and SCF ubiquitin ligases are found in a novel BCOR complex that is recruited to BCL6 targets. Mol. Cell. Biol. 26(18), 6880–6889 (2006).
    • 13. Wong SJ, Gearhart MD, Taylor AB et al. KDM2B recruitment of the polycomb group complex, PRC1.1, requires cooperation between PCGF1 and BCORL1. Structure 24(10), 1795–1801 (2016).
    • 14. Gil J, O’Loghlen A. PRC1 complex diversity: where is it taking us? Trends Cell Biol. 24(11), 632–641 (2014).
    • 15. Schwartz YB, Pirrotta V. A new world of polycombs: unexpected partnerships and emerging functions. Nat. Rev. Genet. 14(12), 853–864 (2013).
    • 16. Gao Z, Zhang J, Bonasio R et al. PCGF homologs, CBX proteins, and RYBP define functionally distinct PRC1 family complexes. Mol. Cell. 45(3), 344–356 (2012).
    • 17. Di Croce L, Helin K. Transcriptional regulation by polycomb group proteins. Nat. Struct. Mol. Biol. 20(10), 1147–1155 (2013).
    • 18. Farcas AM, Blackledge NP, Sudbery I et al. KDM2B links the polycomb repressive complex 1 (PRC1) to recognition of CpG islands. Elife 1, e00205 (2012).
    • 19. Wang L, Brown JL, Cao R, Zhang Y, Kassis JA, Jones RS. Hierarchical recruitment of polycomb group silencing complexes. Mol. Cell. 14(5), 637–646 (2004).
    • 20. Van der Lugt NM, Domen J, Linders K et al. Posterior transformation, neurological abnormalities, and severe hematopoietic defects in mice with a targeted deletion of the BMI-1 proto-oncogene. Genes Dev. 8(7), 757–769 (1994).
    • 21. Akasaka T, Kanno M, Balling R, Mieza MA, Taniguchi M, Koseki H. A role for mel-18, a polycomb group-related vertebrate gene, during theanteroposterior specification of the axial skeleton. Development 122(5), 1513–1522 (1996).
    • 22. Imagawa E, Higashimoto K, Sakai Y et al. Mutations in genes encoding polycomb repressive complex 2 subunits cause Weaver syndrome. Hum. Mutat. 38(6), 637–648 (2017).
    • 23. Feberwee HE, Feenstra I, Oberoi S et al. Novel BCOR mutations in patients with oculofaciocardiodental (OFCD) syndrome. Clin. Genet. 85(2), 194–197 (2014).
    • 24. Ng D, Thakker N, Corcoran CM et al. Oculofaciocardiodental and Lenz microphthalmia syndromes result from distinct classes of mutations in BCOR. Nat. Genet. 36(4), 411–416 (2004).
    • 25. Hedera P, Gorski JL. Oculo-facio-cardio-dental syndrome: skewed X chromosome inactivation in mother and daughter suggest X-linked dominant inheritance. Am. J. Med. Genet. A 123A(3), 261–266 (2003).
    • 26. O’Byrne JJ, Laffan E, Murray DJ, Reardon W. Oculo-facio-cardio-dental syndrome with craniosynostosis, temporal hypertrichosis, and deafness. Am. J. Med. Genet. Part A 173(5), 1374–1377 (2017).
    • 27. Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat. Rev. Cancer. 6(11), 846–856 (2006).
    • 28. Wamstad JA, Corcoran CM, Keating AM, Bardwell VJ. Role of the transcriptional corepressor BCOR in embryonic stem cell differentiation and early embryonic development. PLoS ONE 3(7), e2814 (2008). •• Demonstrates for the first time the role of BCOR in the differentiation of embryonic stem cells into ectoderm, mesoderm and hematopoietic lineages.
    • 29. Wang Z, Gearhart MD, Lee Y-W et al. A non-canonical BCOR–PRC1.1 complex represses differentiation programs in human ESCs. Cell Stem Cell. 22(2), 235–251.e9 (2018). •• Demonstrates that BCOR is necessary for maintaining primed pluripotency in human embryonic stem cells and to inhibit differentiation toward mesoderm and endoderm.
    • 30. Gooskens SLM, Furtwängler R, Vujanic GM, Dome JS, Graf N, van den Heuvel-Eibrink MM. Clear cell sarcoma of the kidney: a review. Eur. J. Cancer. 48(14), 2219–2226 (2012).
    • 31. Argani P, Perlman EJ, Breslow NE et al. Clear cell sarcoma of the kidney: a review of 351 cases from the National Wilms Tumor Study Group Pathology Center. Am. J. Surg. Pathol. 24(1), 4–18 (2000).
    • 32. Boo Y-J, Fisher JC, Haley MJ, Cowles RA, Kandel JJ, Yamashiro DJ. Vascular characterization of clear cell sarcoma of the kidney in a child: a case report and review. J. Pediatr. Surg. 44(10), 2031–2036 (2009).
    • 33. Balarezo FS, Joshi VV. Clear cell sarcoma of the pediatric kidney: detailed description and analysis of variant histologic patterns of a tumor with many faces. Adv. Anat. Pathol. 8(2), 98–108 (2001).
    • 34. Ueno-Yokohata H, Okita H, Nakasato K et al. Consistent in-frame internal tandem duplications of BCOR characterize clear cell sarcoma of the kidney. Nat. Genet. 47(8), 861–863 (2015). •• The first identification of internal tandem duplications (ITD)-BCOR as the pathogenetic event in clear cell sarcoma of the kidney.
    • 35. Astolfi A, Melchionda F, Perotti D et al. Whole transcriptome sequencing identifies BCOR internal tandem duplication as a common feature of clear cell sarcoma of the kidney. Oncotarget 6(38), 40934–40939 (2015).
    • 36. Roy A, Kumar V, Zorman B et al. Recurrent internal tandem duplications of BCOR in clear cell sarcoma of the kidney. Nat. Commun. 6(1), 8891 (2015).
    • 37. Karlsson J, Valind A, Gisselsson D. BCOR internal tandem duplication and YWHAE–NUTM2B/E fusion are mutually exclusive events in clear cell sarcoma of the kidney. Genes Chromosomes Cancer 55(2), 120–123 (2016).
    • 38. Kenny C, Bausenwein S, Lazaro A et al. Mutually exclusive BCOR internal tandem duplications and YWHAE–NUTM2 fusions in clear cell sarcoma of kidney: not the full story. J. Pathol. 238(5), 617–620 (2016).
    • 39. Wong MK, Ng CCY, Kuick CH et al. Clear cell sarcomas of the kidney are characterised by BCOR gene abnormalities, including exon 15 internal tandem duplications and BCOR–CCNB3 gene fusion. Histopathology 72(2), 320–329 (2018).
    • 40. Kao Y-C, Sung Y-S, Zhang L et al. Recurrent BCOR internal tandem duplication and YWHAE–NUTM2B fusions in soft tissue undifferentiated round cell sarcoma of infancy. Am. J. Surg. Pathol. 40(8), 1009–1020 (2016).
    • 41. Chiang S, Lee C-H, Stewart CJR et al. BCOR is a robust diagnostic immunohistochemical marker of genetically diverse high-grade endometrial stromal sarcoma, including tumors exhibiting variant morphology. Mod. Pathol. 30(9), 1251–1261 (2017).
    • 42. Kool M, Jones DTW, Jäger N et al. Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition. Cancer Cell. 25(3), 393–405 (2014).
    • 43. Mirkovic J, Calicchio M, Fletcher CD, Perez-Atayde AR. Diffuse and strong cyclin D1 immunoreactivity in clear cell sarcoma of the kidney. Histopathology 67(3), 306–312 (2015).
    • 44. Jet Aw S, Hong Kuick C, Hwee Yong M et al. Novel karyotypes and cyclin D1 immunoreactivity in clear cell sarcoma of the kidney. Pediatr. Dev. Pathol. 18(4), 297–304 (2015).
    • 45. Antonescu C. Round cell sarcomas beyond Ewing: emerging entities. Histopathology 64(1), 26–37 (2014).
    • 46. Pierron G, Tirode F, Lucchesi C et al. A new subtype of bone sarcoma defined by BCOR–CCNB3 gene fusion. Nat. Genet. 44(4), 461–466 (2012). •• The first identification of BCOR–CCNB3 gene fusion.
    • 47. Cuthbertson DW, Caceres K, Hicks J, Friedman EM. A cooperative approach to diagnosis of rare diseases: primitive myxoid mesenchymal tumor of infancy. Ann. Clin. Lab. Sci. 44(3), 310–316 (2014).
    • 48. Wei S, Siegal GP. Round cell tumors of bone. Adv. Anat. Pathol. 21(5), 359–372 (2014).
    • 49. Chang KTE, Goytain A, Tucker T et al. Development and evaluation of a pan-sarcoma fusion gene detection assay using the NanoString nCounter platform. J. Mol. Diagn. 20(1), 63–77 (2018).
    • 50. Machado I, Yoshida A, Morales MGN et al. Review with novel markers facilitates precise categorization of 41 cases of diagnostically challenging, “undifferentiated small round cell tumors”. A clinicopathologic, immunophenotypic and molecular analysis. Ann. Diagn. Pathol. 34, 1–12 (2018).
    • 51. Kao Y-C, Owosho AA, Sung Y-S et al. BCOR–CCNB3 fusion positive sarcomas: a clinicopathologic and molecular analysis of 36 cases with comparison to morphologic spectrum and clinical behavior of other round cell sarcomas. Am. J. Surg. Pathol. 42(5), 604–615 (2018).
    • 52. Santiago T, Clay MR, Allen SJ, Orr BA. Recurrent BCOR internal tandem duplication and BCOR or BCL6 expression distinguish primitive myxoid mesenchymal tumor of infancy from congenital infantile fibrosarcoma. Mod. Pathol. 30(6), 884–891 (2017).
    • 53. Yamada Y, Kuda M, Kohashi K et al. Histological and immunohistochemical characteristics of undifferentiated small round cell sarcomas associated with CIC–DUX4 and BCOR–CCNB3 fusion genes. Virchows Arch. 470(4), 373–380 (2017).
    • 54. Machado I, Yoshida A, López-Guerrero JA et al. Immunohistochemical analysis of NKX2.2, ETV4, and BCOR in a large series of genetically confirmed Ewing sarcoma family of tumors. Pathol. Res. Pract. 213(9), 1048–1053 (2017).
    • 55. Specht K, Zhang L, Sung Y-S et al. Novel BCOR–MAML3 and ZC3H7B–BCOR gene fusions in undifferentiated small blue round cell sarcomas. Am. J. Surg. Pathol. 40(4), 433–442 (2016).
    • 56. Puls F, Niblett A, Marland G et al. BCOR–CCNB3 (Ewing-like) sarcoma: a clinicopathologic analysis of 10 cases, in comparison with conventional Ewing sarcoma. Am. J. Surg. Pathol. 38(10), 1307–1318 (2014).
    • 57. Ludwig K, Alaggio R, Zin A et al. BCOR–CCNB3 undifferentiated sarcoma – does immunohistochemistry help in the identification? Pediatr. Dev. Pathol. 20(4), 321–329 (2017).
    • 58. Peters TL, Kumar V, Polikepahad S et al. BCOR–CCNB3 fusions are frequent in undifferentiated sarcomas of male children. Mod. Pathol. 28(4), 575–586 (2015).
    • 59. Li W-S, Liao I-C, Wen M-C, Lan HH-C, Yu S-C, Huang H-Y. BCOR–CCNB3-positive soft tissue sarcoma with round-cell and spindle-cell histology: a series of four cases highlighting the pitfall of mimicking poorly differentiated synovial sarcoma. Histopathology 69(5), 792–801 (2016).
    • 60. Panagopoulos I, Thorsen J, Gorunova L et al. Fusion of the ZC3H7B and BCOR genes in endometrial stromal sarcomas carrying an X;22-translocation. Genes Chromosomes Cancer 52(7), 610–618 (2013).
    • 61. Cramer SL, Miller AL, Pressey JG et al. Pediatric anaplastic embryonal rhabdomyosarcoma: targeted therapy guided by genetic analysis and a patient-derived xenograft study. Front. Oncol. 7, 327 (2017).
    • 62. Mackintosh C. Dynamic interactions between 14-3-3 proteins and phosphoproteins regulate diverse cellular processes. Biochem. J. 381(2), 329–342 (2004).
    • 63. Rakheja D, Weinberg AG, Tomlinson GE, Partridge K, Schneider NR. Translocation (10;17)(q22;p13): a recurring translocation in clear cell sarcoma of kidney. Cancer Genet. Cytogenet. 154(2), 175–179 (2004).
    • 64. French CA, Miyoshi I, Kubonishi I, Grier HE, Perez-Atayde AR, Fletcher JA. BRD4-NUT fusion oncogene: a novel mechanism in aggressive carcinoma. Cancer Res. 63(2), 304–307 (2003).
    • 65. Hoang LN, Aneja A, Conlon N et al. Novel high-grade endometrial stromal sarcoma. Am. J. Surg. Pathol. 41(1), 12–24 (2017).
    • 66. Knutson SK, Warholic NM, Wigle TJ et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc. Natl Acad. Sci. USA 110(19), 7922–7927 (2013).
    • 67. Bantignies F, Cavalli G. Polycomb group proteins: repression in 3D. Trends Genet. 27(11), 454–464 (2011).
    • 68. Kalb R, Latwiel S, Baymaz HI et al. Histone H2A monoubiquitination promotes histone H3 methylation in polycomb repression. Nat. Struct. Mol. Biol. 21(6), 569–571 (2014).
    • 69. Kao Y-C, Sung Y-S, Zhang L et al. BCOR overexpression is a highly sensitive marker in round cell sarcomas with BCOR genetic abnormalities. Am. J. Surg. Pathol. 40(12), 1670–1678 (2016).
    • 70. Mariño-Enriquez A, Lauria A, Przybyl J et al. BCOR internal tandem duplication in high-grade uterine sarcomas. Am. J. Surg. Pathol. 42(3), 335–341 (2018).
    • 71. Louis DN, Perry A, Reifenberger G et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 131(6), 803–820 (2016).
    • 72. Hoffman LM, DeWire M, Ryall S et al. Spatial genomic heterogeneity in diffuse intrinsic pontine and midline high-grade glioma: implications for diagnostic biopsy and targeted therapeutics. Acta Neuropathol. Commun. 4(1), 1 (2016).
    • 73. Mackay A, Burford A, Carvalho D et al. Integrated molecular meta-analysis of 1,000 pediatric high-grade and diffuse intrinsic pontine glioma. Cancer Cell 32(4), 520–537.e5 (2017).
    • 74. Bale TA, Abedalthagafi M, Bi WL et al. Genomic characterization of recurrent high-grade astroblastoma. Cancer Genet. 209(7–8), 321–330 (2016).
    • 75. Northcott PA, Jones DTW, Kool M et al. Medulloblastomics: the end of the beginning. Nat. Rev. Cancer. 12(12), 818–834 (2012).
    • 76. Morrissy AS, Garzia L, Shih DJH et al. Divergent clonal selection dominates medulloblastoma at recurrence. Nature 529(7586), 351–357 (2016).
    • 77. Argani P, Kao Y-C, Zhang L et al. Primary renal sarcomas with BCOR–CCNB3 gene fusion: a report of 2 cases showing histologic overlap with clear cell sarcoma of kidney, suggesting further link between BCOR-related sarcomas of the kidney and soft tissues. Am. J. Surg. Pathol. 41(12), 1702–1712 (2017).
    • 78. Antonescu CR, Sung Y-S, Chen C-L et al. Novel ZC3H7B–BCOR, MEAF6–PHF1, and EPC1–PHF1 fusions in ossifying fibromyxoid tumors-molecular characterization shows genetic overlap with endometrial stromal sarcoma. Genes Chromosomes Cancer 53(2), 183–193 (2014).
    • 79. Wamstad JA, Bardwell VJ. Characterization of BCOR expression in mouse development. Gene Expr. Patterns 7(5), 550–557 (2007).
    • 80. McEvoy J, Nagahawatte P, Finkelstein D et al. RB1 gene inactivation by chromothripsis in human retinoblastoma. Oncotarget 5(2), 438–450 (2014).
    • 81. Zhang J, Benavente CA, McEvoy J et al. A novel retinoblastoma therapy from genomic and epigenetic analyses. Nature 481(7381), 329–334 (2012).
    • 82. Kooi IE, Mol BM, Massink MPG et al. Somatic genomic alterations in retinoblastoma beyond RB1 are rare and limited to copy number changes. Sci. Rep. 6(1), 25264 (2016).
    • 83. Haferlach T, Nagata Y, Grossmann V et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28(2), 241–247 (2014).
    • 84. Cao Q, Gearhart MD, Gery S et al. BCOR regulates myeloid cell proliferation and differentiation. Leukemia 30(5), 1155–1165 (2016). • Defines for the first time the role of BCOR in myeloid cell differentiation.
    • 85. Tanaka T, Nakajima-Takagi Y, Aoyama K et al. Internal deletion of BCOR reveals a tumor suppressor function for BCOR in T lymphocyte malignancies. J. Exp. Med. 214(10), 2901–2913 (2017).
    • 86. Lefebure M, Tothill RW, Kruse E et al. Genomic characterisation of Eμ-Myc mouse lymphomas identifies BCOR as a Myc co-operative tumour-suppressor gene. Nat. Commun. 8, 14581 (2017).
    • 87. Abáigar M, Robledo C, Benito R et al. Chromothripsis is a recurrent genomic abnormality in high-risk myelodysplastic syndromes. PLoS ONE 11(10), e0164370 (2016).
    • 88. de Rooij JDE, van den Heuvel-Eibrink MM, Hermkens MCH et al. BCOR and BCORL1 mutations in pediatric acute myeloid leukemia. Haematologica 100(5), e194–e195 (2015).
    • 89. Terada K, Yamaguchi H, Ueki T et al. Usefulness of BCOR gene mutation as a prognostic factor in acute myeloid leukemia with intermediate cytogenetic prognosis. Genes Chromosomes Cancer 57(8), 401–408 (2018).
    • 90. Olsson L, Zettermark S, Biloglav A et al. The genetic landscape of paediatric de novo acute myeloid leukaemia as defined by single nucleotide polymorphism array and exon sequencing of 100 candidate genes. Br. J. Haematol. 174(2), 292–301 (2016).
    • 91. Yoshida K, Sanada M, Shiraishi Y et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478(7367), 64–69 (2011).
    • 92. Damm F, Chesnais V, Nagata Y et al. BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders. Blood 122(18), 3169–3177 (2013).
    • 93. Montalban-Bravo G, Takahashi K, Patel K et al. Impact of the number of mutations in survival and response outcomes to hypomethylating agents in patients with myelodysplastic syndromes or myelodysplastic/myeloproliferative neoplasms. Oncotarget 9(11), 9714–9727 (2018).
    • 94. Tarlock K, Zhong S, He Y et al. Distinct age-associated molecular profiles in acute myeloid leukemia defined by comprehensive clinical genomic profiling. Oncotarget 9(41), 26417–26430 (2018).
    • 95. Thota S, Viny AD, Makishima H et al. Genetic alterations of the cohesin complex genes in myeloid malignancies. Blood 124(11), 1790–1798 (2014).
    • 96. Yamato G, Shiba N, Yoshida K et al. ASXL2 mutations are frequently found in pediatric AML patients with t(8;21)/ RUNX1–RUNX1T1 and associated with a better prognosis. Genes Chromosomes Cancer 56(5), 382–393 (2017).
    • 97. Eisfeld A-K, Mrózek K, Kohlschmidt J et al. The mutational oncoprint of recurrent cytogenetic abnormalities in adult patients with de novo acute myeloid leukemia. Leukemia 31(10), 2211–2218 (2017).
    • 98. Bolli N, Manes N, McKerrell T et al. Characterization of gene mutations and copy number changes in acute myeloid leukemia using a rapid target enrichment protocol. Haematologica 100(2), 214–222 (2015).
    • 99. Eisfeld A-K, Kohlschmidt J, Mrózek K et al. Adult acute myeloid leukemia with trisomy 11 as the sole abnormality is characterized by the presence of five distinct gene mutations: MLL–PTD, DNMT3A, U2AF1, FLT3–ITD and IDH2. Leukemia 30(11), 2254–2258 (2016).
    • 100. Herold T, Metzeler KH, Vosberg S et al. Isolated trisomy 13 defines a homogeneous AML subgroup with high frequency of mutations in spliceosome genes and poor prognosis. Blood 124(8), 1304–1311 (2014).
    • 101. Shiba N, Yoshida K, Shiraishi Y et al. Whole-exome sequencing reveals the spectrum of gene mutations and the clonal evolution patterns in paediatric acute myeloid leukaemia. Br. J. Haematol. 175(3), 476–489 (2016).
    • 102. Nazha A, Zarzour A, Al-Issa K et al. The complexity of interpreting genomic data in patients with acute myeloid leukemia. Blood Cancer J. 6(12), e510(2016).
    • 103. Metzeler KH, Herold T, Rothenberg-Thurley M et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 128(5), 686–698 (2016).
    • 104. Kihara R, Nagata Y, Kiyoi H et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia 28(8), 1586–1595 (2014).
    • 105. Gaidzik VI, Teleanu V, Papaemmanuil E et al. RUNX1 mutations in acute myeloid leukemia are associated with distinct clinico-pathologic and genetic features. Leukemia 30(11), 2160–2168 (2016).
    • 106. Patel BJ, Przychodzen B, Thota S et al. Genomic determinants of chronic myelomonocytic leukemia. Leukemia 31(12), 2815–2823 (2017).
    • 107. Grossmann V, Tiacci E, Holmes AB et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood 118(23), 6153–6163 (2011). • The first identification of somatic BCOR mutations in natural killer cell-acute myeloid leukemia.
    • 108. Puente XS, Beà S, Valdés-Mas R et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature 526(7574), 519–524 (2015).
    • 109. Leeksma AC, Taylor J, Wu B et al. Clonal diversity predicts adverse outcome in chronic lymphocytic leukemia. Leukemia 33(2), 390–402 (2019).
    • 110. Ogawa S. Clonal hematopoiesis in acquired aplastic anemia. Blood 128(3), 337–347 (2016).
    • 111. Marsh JCW, Mufti GJ. Clinical significance of acquired somatic mutations in aplastic anaemia. Int. J. Hematol. 104(2), 159–167 (2016).
    • 112. Kulasekararaj AG, Jiang J, Smith AE et al. Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood 124(17), 2698–2704 (2014).
    • 113. Landau DA, Carter SL, Stojanov P et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell 152(4), 714–726 (2013).
    • 114. López C, Bergmann AK, Paul U et al. Genes encoding members of the JAK–STAT pathway or epigenetic regulators are recurrently mutated in T-cell prolymphocytic leukaemia. Br. J. Haematol. 173(2), 265–273 (2016).
    • 115. Stengel A, Kern W, Zenger M et al. Genetic characterization of T-PLL reveals two major biologic subgroups and JAK3 mutations as prognostic marker. Genes Chromosomes Cancer 55(1), 82–94 (2016).
    • 116. Kim J-A, Hwang B, Park SN et al. Genomic profile of chronic lymphocytic leukemia in Korea identified by targeted sequencing. PLoS ONE 11(12), e0167641 (2016).
    • 117. Stengel A, Kern W, Meggendorfer M et al. Number of RUNX1 mutations, wild-type allele loss and additional mutations impact on prognosis in adult RUNX1-mutated AML. Leukemia 32(2), 295–302 (2018).
    • 118. Lee S, Park HY, Kang SY et al. Genetic alterations of JAK/STAT cascade and histone modification in extranodal NK/T-cell lymphoma nasal type. Oncotarget 6(19), 17764–17776 (2015).
    • 119. Moreira AL, Won HH, McMillan R et al. Massively parallel sequencing identifies recurrent mutations in TP53 in thymic carcinoma associated with poor prognosis. J. Thorac. Oncol. 10(2), 373–380 (2015).
    • 120. Morris LGT, Chandramohan R, West L et al. The molecular landscape of recurrent and metastatic head and neck cancers. JAMA Oncol. 3(2), 244 (2017).
    • 121. Petrini I, Rajan A, Pham T et al. Whole genome and transcriptome sequencing of a B3 thymoma. PLoS ONE 8(4), e60572 (2013).
    • 122. Jallades L, Baseggio L, Sujobert P et al. Exome sequencing identifies recurrent BCOR alterations and the absence of KLF2, TNFAIP3 and MYD88 mutations in splenic diffuse red pulp small B-cell lymphoma. Haematologica 102(10), 1758–1766 (2017).
    • 123. Dobashi A, Tsuyama N, Asaka R et al. Frequent BCOR aberrations in extranodal NK/T-Cell lymphoma, nasal type. Genes Chromosomes Cancer 55(5), 460–471 (2016).
    • 124. Alaggio R, Ninfo V, Rosolen A, Coffin CM. Primitive myxoid mesenchymal tumor of infancy. Am. J. Surg. Pathol. 30(3), 388–394 (2006).
    • 125. Cramer SL, Li R, Ali S, Bradley JA, Kim HK, Pressey JG. Successful treatment of recurrent primitive myxoid mesenchymal tumor of infancy with BCOR internal tandem duplication. J. Natl. Compr. Cancer Netw. 15(7), 868–871 (2017).
    • 126. Watson S, Perrin V, Guillemot D et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J. Pathol. 245(1), 29–40 (2018).
    • 127. Baldauf MC, Orth MF, Dallmayer M et al. Robust diagnosis of Ewing sarcoma by immunohistochemical detection of super-enhancer-driven EWSR1–ETS targets. Oncotarget 9(2), 1587–1601 (2018).
    • 128. Matsuyama A, Shiba E, Umekita Y et al. Clinicopathologic diversity of undifferentiated sarcoma with BCOR–CCNB3 fusion: analysis of 11 cases with a reappraisal of the utility of immunohistochemistry for BCOR and CCNB3. Am. J. Surg. Pathol. 41(12), 1713–1721 (2017).
    • 129. Machado I, Navarro L, Pellin A et al. Defining Ewing and Ewing-like small round cell tumors (SRCT): the need for molecular techniques in their categorization and differential diagnosis. A study of 200 cases. Ann. Diagn. Pathol. 22, 25–32 (2016).
    • 130. Vitour D, Lindenbaum P, Vende P, Becker MM, Poncet D. RoXaN, a novel cellular protein containing TPR, LD, and zinc finger motifs, forms a ternary complex with eukaryotic initiation factor 4G and rotavirus NSP3. J. Virol. 78(8), 3851–3862 (2004).
    • 131. Shern JF, Chen L, Chmielecki J et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov. 4(2), 216–231 (2014).
    • 132. Seki M, Nishimura R, Yoshida K et al. Integrated genetic and epigenetic analysis defines novel molecular subgroups in rhabdomyosarcoma. Nat. Commun. 6(1), 7557 (2015).
    • 133. Lee C-H, Nucci MR. Endometrial stromal sarcoma – the new genetic paradigm. Histopathology 67(1), 1–19 (2015).
    • 134. Micci F, Gorunova L, Agostini A et al. Cytogenetic and molecular profile of endometrial stromal sarcoma. Genes Chromosomes Cancer 55(11), 834–846 (2016).
    • 135. Lee C-H, Ou W-B, Marino-Enriquez A et al. 14-3-3 fusion oncogenes in high-grade endometrial stromal sarcoma. Proc. Natl Acad. Sci. USA 109(3), 929–934 (2012).
    • 136. Punnett HH, Halligan GE, Zaeri N, Karmazin N. Translocation 10;17 in clear cell sarcoma of the kidney. A first report. Cancer Genet. Cytogenet. 41(1), 123–128 (1989).
    • 137. O’Meara E, Stack D, Lee C-H et al. Characterization of the chromosomal translocation t(10;17)(q22;p13) in clear cell sarcoma of kidney. J. Pathol. 227(1), 72–80 (2012).
    • 138. Brownlee NA, Perkins LA, Stewart W et al. Recurring translocation (10;17) and deletion (14q) in clear cell sarcoma of the kidney. Arch. Pathol. Lab. Med. 131(3), 446–451 (2007).
    • 139. Lewis N, Soslow RA, Delair DF et al. ZC3H7B–BCOR high-grade endometrial stromal sarcomas: a report of 17 cases of a newly defined entity. Mod. Pathol. 31(4), 674–684 (2018).
    • 140. Sturm D, Orr BA, Toprak UH et al. New brain tumor entities emerge from molecular classification of CNS-PNETs. Cell 164(5), 1060–1072 (2016). •• The first identification of the CNS-PNET group ‘CNS high-grade neuroepithelial tumor with BCOR alteration’ (CNS HGNET-BCOR).
    • 141. Jones DTW, Jäger N, Kool M et al. Dissecting the genomic complexity underlying medulloblastoma. Nature 488(7409), 100–105 (2012).
    • 142. Pugh TJ, Weeraratne SD, Archer TC et al. Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations. Nature 488(7409), 106–110 (2012).
    • 143. Louis DN, Ohgaki H, Wiestler OD et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114(2), 97–109 (2007).
    • 144. Yoshida Y, Nobusawa S, Nakata S et al. CNS high-grade neuroepithelial tumor with BCOR internal tandem duplication: a comparison with its counterparts in the kidney and soft tissue. Brain Pathol. 28(5), 710–720 (2018).
    • 145. Cutcliffe C, Kersey D, Huang C-C et al. Clear cell sarcoma of the kidney: up-regulation of neural markers with activation of the sonic hedgehog and Akt pathways. Clin. Cancer Res. 11(22), 7986–7994 (2005).
    • 146. Paret C, Theruvath J, Russo A et al. Activation of the basal cell carcinoma pathway in a patient with CNS HGNET-BCOR diagnosis: consequences for personalized targeted therapy. Oncotarget 7(50), 83378–83391 (2016).
    • 147. Choe J-Y, Yun JY, Jeon YK et al. Sonic hedgehog signalling proteins are frequently expressed in retinoblastoma and are associated with aggressive clinicopathological features. J. Clin. Pathol. 68(1), 6–11 (2015).
    • 148. Kool M, Korshunov A, Remke M et al. Molecular subgroups of medulloblastoma: an international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, Group 3, and Group 4 medulloblastomas. Acta Neuropathol. 123(4), 473–484 (2012).
    • 149. Yan L, Ping N, Zhu M et al. Clinical, immunophenotypic, cytogenetic, and molecular genetic features in 117 adult patients with mixed-phenotype acute leukemia defined by WHO-2008 classification. Haematologica 97(11), 1708–1712 (2012).
    • 150. Yamamoto Y, Tsuzuki S, Tsuzuki M, Handa K, Inaguma Y, Emi N. BCOR as a novel fusion partner of retinoic acid receptor alpha in a t(X;17)(p11;q12) variant of acute promyelocytic leukemia. Blood 116(20), 4274–4283 (2010).
    • 151. Ichikawa S, Ichikawa S, Ishikawa I, Takahashi T, Fujiwara T, Harigae H. Successful treatment of acute promyelocytic leukemia with a t(X;17)(p11.4;q21) and BCOR–RARA fusion gene. Cancer Genet. 208(4), 162–163 (2015).
    • 152. Yoshizato T, Dumitriu B, Hosokawa K et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N. Engl. J. Med. 373(1), 35–47 (2015).
    • 153. Park HS, Park SN, Im K et al. Telomere length and somatic mutations in correlation with response to immunosuppressive treatment in aplastic anaemia. Br. J. Haematol. 178(4), 603–615 (2017).
    • 154. Chahal M, Pleasance E, Grewal J et al. Personalized oncogenomic analysis of metastatic adenoid cystic carcinoma: using whole-genome sequencing to inform clinical decision-making. Mol. Case Stud. 4(2), a002626 (2018).
    • 155. García-Sanz P, Triviño JC, Mota A et al. Chromatin remodelling and DNA repair genes are frequently mutated in endometrioid endometrial carcinoma. Int. J. Cancer. 140(7), 1551–1563 (2017).
    • 156. Zhao S, Bellone S, Lopez S et al. Mutational landscape of uterine and ovarian carcinosarcomas implicates histone genes in epithelial–mesenchymal transition. Proc. Natl Acad. Sci. USA 113(43), 12238–12243 (2016).