Next Article in Journal
TIMP1 and TIMP2 Downregulate TGFβ Induced Decidual-like Phenotype in Natural Killer Cells
Next Article in Special Issue
Characteristics of Nephroblastoma/Nephroblastomatosis in Children with a Clinically Reported Underlying Malformation or Cancer Predisposition Syndrome
Previous Article in Journal
A Multi-Scale and Multi-Technique Approach for the Characterization of the Effects of Spatially Fractionated X-ray Radiation Therapies in a Preclinical Model
Previous Article in Special Issue
The Psychoneuroimmunology of Stress Regulation in Pediatric Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Stratification of Childhood Ependymomas as a Basis for Personalized Diagnostics and Treatment

1
Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia
2
Research Institute of Medical Cell Technologies, 620026 Yekaterinburg, Russia
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(19), 4954; https://doi.org/10.3390/cancers13194954
Submission received: 7 September 2021 / Revised: 27 September 2021 / Accepted: 29 September 2021 / Published: 1 October 2021
(This article belongs to the Special Issue Pediatric Cancers)

Abstract

:

Simple Summary

The current trend in neuropathology directs to the integrated histo-molecular approach. The traditional concept of histological grade should be complemented by comprehensive diagnostics with the mandatory use of molecular genetic markers. As a consequence, basic types of CNS tumors fall into multiple nosological entities that can be morphologically similar while having fundamentally different pathogenesis and clinical presentation. This trend is particularly evident for ependymal tumors, which harbor molecular markers of decisive importance for the prognosis. This minireview emphasizes recent achievements in ependymoma biology research closely connected with state-of-the-art diagnostics.

Abstract

Ependymomas are among the most enigmatic tumors of the central nervous system, posing enormous challenges for pathologists and clinicians. Despite the efforts made, the treatment options are still limited to surgical resection and radiation therapy, while none of conventional chemotherapies is beneficial. While being histologically similar, ependymomas show considerable clinical and molecular diversity. Their histopathological evaluation alone is not sufficient for reliable diagnostics, prognosis, and choice of treatment strategy. The importance of integrated diagnosis for ependymomas is underscored in the recommendations of Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy. These updated recommendations were adopted and implemented by WHO experts. This minireview highlights recent advances in comprehensive molecular-genetic characterization of ependymomas. Strong emphasis is made on the use of molecular approaches for verification and specification of histological diagnoses, as well as identification of prognostic markers for ependymomas in children.

1. Introduction

Ependymal tumors (ependymomas, EPNs), a common type of malignant neoplasms of the central nervous system (CNS), constitute about 10% of all intracranial tumors and about 20% of spinal cord tumors. EPNs rank third in the prevalence of pediatric CNS tumors (after glial and embryonal tumors) [1]. Despite the use of advanced protocols that include maximal safe surgical resection followed by localized radiotherapy, the mortality remains high due to frequent relapses explained by the strong metastatic potential of EPNs complemented by an efficient spread of metastases with cerebrospinal fluid.
Adverse predictors for EPNs are early age at onset, residual tumor tissue after resection, and metastatic lesions in CNS [2,3,4,5]; however, the detailed prognosis for EPNs is often hampered by (1) clinical and morphological diversity of the tumors and (2) complex relations of histopathological grades with the prognosis [6,7]. In line with the modern trends in neuropathology, the aggressiveness of a tumor and, accordingly, the prognosis is mainly determined by molecular-genetic aberrations, whereas the conventional, histologically defined grade becomes subsidiary [8,9]. For pediatric EPNs, the relevance of molecular stratification is especially obvious.
Over recent years, the diagnostics of CNS malignancies has been significantly reconsidered. The accent has been shifted from pathomorphology to molecular profiling and the search for clinically informative markers that would justify the selection of a particular therapy. Molecular framework-based stratification schemes have been developed and introduced into clinical practice for a number of CNS tumors; examples include IDH1/2 mutations and 1p/19q codeletions for gliomas and oligodendrogliomas [10]; KIAA1549-BRAF fusions, MYB/MYBL rearrangements, recurrent pathogenic mutations in BRAF and H3F3A for pediatric astrocytomas [11,12]; and four molecular groups with the account of MYC/MYCN amplification for medulloblastomas [13].
EPNs of different molecular etiologies occupy distinct anatomical compartments within CNS. Recurrent genetic or epigenetic alterations found in EPNs are invariably linked to tumor localization. Molecular subgrouping of EPNs is superior to histopathological grading based on the WHO criteria [14]. Gene expression signatures and related subgrouping have shown the highest prognostic value among other studied molecular criteria. A tumor retains its affiliation to a particular subgroup indefinitely (it cannot be switched during progression and/or relapse of the disease), which increases its clinical significance [5,14,15]. An advanced EPN classification has been recently proposed by the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) update 7, aimed at connecting localization-dependent molecular groups with tumor progression modes and outcomes [16]. This view has been supported by WHO experts and reflected in the summary of the upcoming fifth edition of the WHO Classification of Tumors of the Central Nervous System (WHO CNS5) [9]. According to the newest CNS tumor nomenclature, ependymomas are subdivided into supratentorial (ST-EPNs), infratentorial (a.k.a. posterior fossa ependymomas, PF-EPNs), and spinal (Sp-EPNs) by localization of the primary tumor; these groups are further stratified by (epi)genetic features.

2. Molecular Profiles of ST-EPNs

ST-EPNs are fairly rare and show considerable genetic heterogeneity. ST-EPNs have been recently stratified into two major groups: supratentorial ependymoma, ZFTA fusion-positive (ST-EPN-ZFTA) and supratentorial ependymoma, YAP1 fusion-positive (ST-EPN-YAP1) [9] consistently with gene expression and/or DNA methylation signatures revealed by transcriptomic methods and/or whole-genome DNA methylation profiling, respectively.

2.1. ST-EPN-ZFTA Group

Gain-of-function rearrangements in ZFTA or YAP1 are specific for ST-EPNs. At that, ST-EPN-ZFTA tumors are prevalent (50–75% and 25% of ST-EPNs in children and adults, respectively [7,14,17,18,19,20]), while ST-EPN-YAP1 tumors are rare (3–10% in different cohorts [7,14,18,19,21,22]). The archetypal chimeric transcript harbored by ZFTA-rearranged ependymomas is ZFTARELA, hence the ST-EPN-RELA is a traditional designation for this group [14]. Alternative ZFTA fusions (non-RELA, e.g., ZFTA-NCOA1, ZFTA-NCOA2, ZFTA-MAML2 [23,24,25,26,27,28], and MN1-ZFTA [28]) are less common.
Recurrent ZFTARELA fusion is a unique molecular hallmark of ZFTA-positive EPNs not found in other CNS tumors. Nine different transcript variants have been described, differing by breakpoints in RELA and its partner gene; the prevalent isoform comprises ZFTA exon 2 spliced to RELA exon 2 [17,22,29]. Formation of the ZFTARELA intrachromosomal gene fusion results from multiple double-strand breaks in 11q13.1 with subsequent random reassociation (typical for chromotrypsis); hence the diversity of fusion points for such transcripts. The oncogenic impact of classical ZFTA–RELA fusions was elucidated in recent studies [30,31].
RELA encodes the RelA (p65) subunit of the dimeric nuclear factor-κB (NF-κB), most known as a master regulator of immune responses and inflammation. NF-κB promotes apoptosis inhibition, cell growth, and pro-angiogenic signaling—the basic components of oncogenesis and tumor progression. Expression of RELA chimeras results in constitutive activation of NF-κB signaling pathway [17] and associated resistance of the tumor to chemo and radiation therapies [32]. ZFTA chimeric proteins accumulate in the nucleus. A zinc finger domain in the truncated ZFTA protein endows the chimeras with extraordinary high affinity to DNA. The oncoprotein interferes with chromatin structure at ST-EPN-associated loci, enabling the RELA transactivation domain to induce their transcription [30]. Moreover, apart from the canonical NF-κB pathway activation, ZFTARELA fusions may trigger other gene expression programs through recruitment of transcriptional co-activators BRD4, EP300, and CBP, which participate in chromatin-related pathways and represent potential druggable targets [31,33].
EPNs with the ZFTA gene fused with a non-RELA partner gene are considerably less common. These tumors have variable histological structures and, apart from the ependymomal component, may additionally involve pleomorphic xanthoastrocytoma-like, astroblastoma-like, malignant teratoma-like, embryonal tumor-like, or sarcoma-like patterns. Despite the heterogeneous morphology, these tumors are (epi)genetically similar and tend to resemble the classic ZFTARELA-fused EPNs, as revealed by methylome assay. A detailed analysis of DNA methylation profiles allows subdivision of these tumors into two clusters, one of them comprising tumors with histological features of astroblastomas and xanthoastrocytomas, harboring ZFTAMAML2 and MN1ZFTA rearrangements; the second cluster comprises tumors histologically resembling small-cell sarcomatoid carcinomas and undifferentiated sarcomas, harboring ZFTANCOA1 and ZFTANCOA2 rearrangements [24,25,26,27,28].
The presence of recurrent ZFTARELA fusions has been repeatedly implicated as an adverse prognostic factor [5,14,18]. Five-year rates of event-free survival (EFS) and overall survival (OS) for ST-EPN-ZFTA tumors never exceed 29% and 75%, respectively [14]. Within the ST-EPN-ZFTA group, additional risks of relapse have been associated with 1q gains [5]. Interestingly, the St Jude Young Children 07 (SJYC07) study (encompassing ependymal tumors diagnosed in <3-year-olds) identified similar 4-year EFS rates for ST-EPN-ZFTA, ST-EPN-YAP1, and PF-EPN group A [7]. Consistently, two other studies conducted independently by the Italian Association of Pediatric Hematology and Oncology AIEOP and the Children’s Oncology Group (trial ACNS0121) revealed no difference in survival rates for ST-EPNs with and without ZFTARELA fusion [4,34].
ZFTA (non-RELA)-fused EPNs have an especially dismal prognosis, with EFS rates significantly lower compared with classical ZFTARELA-fused EPNs, while the corresponding OS rates are comparable [26]. However, these findings are preliminary, given the small number of cases reported so far. In the context of ST-EPN heterogeneity, it might be useful to consider ZFTA fusions with atypical (non-RELA) partners as a distinguishing feature for a separate group, the prognostic and clinical relevance of which is yet to be specified.

2.2. ST-EPN-YAP1 Group

ST-EPN-YAP1 tumors show an aberrant activity of transcription co-activator YAP1 (Yes-associated protein 1) related to its abnormal accumulation in the nucleus. With YAP1 being a direct regulator of TEAD and SMAD transcription factors, its escape from Hippo-dependent sequestration through accumulation in the nucleus results in sustained proliferative signaling via WNT and Hedgehog. More accurately, the nucleus accumulates the oncogenic fusion protein YAP1–MAMLD1 transferred from the cytoplasm to the nucleus independently of its YAP1-Ser127 phosphorylation status that limits the nuclear import of YAP1 in normal cells [35]. Apart from the prevalent YAP1MAMLD1 fusions, ST-EPN-YAP1 may harbor relatively rare structural variants, e.g., YAP1FAM118B [14,36]. In some cases, the formation of YAP1 fusions involves focal copy number alterations mapping to the 3′ portion of the gene (11q22.1–11q21.2) [37]. Unlike ZFTA-positive ependymomas, ST-EPN-YAP1 tumors have balanced genomes with local aberrations in the YAP1 locus and no evidence of chromothripsis.
Andreiuolo et al. (2019) reported a multicenter retrospective study on what is so far the largest cohort of patients with YAP1-positive EPNs (n = 14). Overall survival for these patients (median observation time of 4.8 years within the range of 0.6–16 years) constituted 100%. It is important to note that the boy-to-girl ratio for the studied cohort was 1:6.5, and only three of the patients were over three years old at the time of diagnosis (the median age at diagnosis constituted 8.2 months) [37]. The best survival rates for the ST-EPN-YAP1 group among other EPNs were also reported by other authors [7,14]. Careful de-escalation of conventional EPN treatment protocols specifically for ST-EPN-YAP1 patients is currently under scrutiny. An opportunity to exclude (delay or dismiss) radiation therapy alleviates the risks of severe cognitive dysfunctions, endocrinopathies, and secondary tumors [38].

2.3. Non-ZFTA/Non-YAP1 ST-EPNs

The molecular diversity of ST-EPNs exceeds the currently established ZFTA-YAP stereotype. Tumors with neither ZFTA nor YAP1 alterations are considered as a separate group, and recent findings emphasize the need for a finer specification. A distinct entity is formed by PLAGL1 rearranged EPNs, harboring EWSR1PLAGL1 and less commonly PLAGL1FOXO1 or PLAGL1EP300 fusions [39], which echoes molecular landscapes of soft tissue sarcomas and a group of rare mesenchymal (non-meningothelial) and glioneuronal CNS tumors with EWSR1–non-ETS fusions [40,41]. Nevertheless, for the vast majority of ST-EPNs lacking recurrent chromosomal rearrangements, the oncogenic driver events remain elusive. Several reports reveal the presence of fusion genes MAML2ASCL2, MARK2ADCY3 [19], PTENTAS2R1 [14], PATZ1MN1, MYH9SEC14L2, MTMR3NCOA3 [24], TMEFF2FOXO1, PCGF1CREBBP [20], FOXO1STK24, as well as EP300BCORL1 in such tumors [21]. Olsen et al. (2015) described two cases of hemispheric infantile EPN-like gliomas with ALK fusions (CCDC88AALK and KTN1ALK), both of them morphologically ambiguous: the tumors showed glial phenotypes and resembled glioblastomas [42]. In the summary of the upcoming WHO CNS5, such tumors have been reclassified and renamed as infant-type hemispheric gliomas harboring receptor tyrosine kinase gene rearrangements [9]. Torre et al. (2020) reported in-frame fusions AGKBRAF and MYO5ANTRK3 as potential targets for therapeutic inhibition [20]. In sum, these observations indicate the absence of a single driver mechanism for this group of tumors while underscoring the importance of their distinction from other CNS neoplasms.

3. Molecular Profiles of PF-EPNs

PF-EPNs, more prevalent in children than adults, constitute about 2/3 of intracranial ependymal tumors of childhood. Based on high-throughput molecular techniques, PF-EPNs are subdivided into two molecular groups: PF-EPN group A (PF-EPN-A), and PF-EPN group B (PF-EPN-B) [9].

3.1. PF-EPN-A Group

This highly heterogeneous group comprises 85–90% of infratentorial EPNs. PF-EPN-A tumors are often located laterally within the posterior fossa and occur predominantly in infants and young children, twice more frequently in boys than in girls; the average age at diagnosis constitutes 3.5 years [14]. The patients are at high risk of relapse, even under multimodal therapy and in the absence of extra adverse prognostic markers [5]. Identification with PF-EPN-A represents a strong independent prognostic factor associated with the worst rates of survival. According to Zapotocky et al. (2019), 5-year and 10-year EFS for PF-EPN-A constitute, respectively, 43% and 37% [3]. Relapses are typical for PF-EPN-A but not PF-EPN-B and, consistently, 10-year OS rates for PF-EPN-A are significantly lower than for PF-EPN-B (56–58% vs. 88–100%) [4,14,15]. Additional negative clinical predictors for PF-EPN-A are the presence of residual tumor tissue (incomplete resection) and adjuvant radiation therapy refusal [3]. Retrospective evaluation of outcomes for the patients receiving treatment under HIT-2000 protocol implicated residual tumors, 1q gains, and high mitotic activity of tumor cells (>10 mitotic figures per 10 fields of view) as independent adverse predictors for PF-EPNs in general and PF-EPN-A tumors in particular [2]. Cytogenetic prognostic factors for PF-EPNs include 1q gains and 6q losses [43,44]. These cytogenetic abnormalities are detected in 18.9% and 8.6% of PF-EPNs, respectively. At that, the 1q gained PF-EPNs harbor 6q losses at an increased frequency of 17.7% [43]. Both types of copy number variations have been qualified as adverse predictors. Five-year progression-free survival rates were 50% for EPNs without 1q gain and 6q loss, as opposed to 32% for 1q gain only, 7.3% for 6q loss only, and 0% for both 1q gain/6q loss tumors [43]. The ultra-high risks conferred by the co-occurrence of cytogenetic markers in PF-EPN-A patients should be taken into account for the treatment regimen optimization.
PF-EPN-A tumors reveal characteristic aberrant patterns of DNA methylation, the so-called CpG-island methylator phenotype (CIMP) with extensive hypermethylation of CpG-islands in promoter regions of multiple genes. This effect critically interferes with the function of PRC2 (Polycomb repressive complex 2) [45,46]—a transcription repressor protein complex that facilitates methylation of nucleosome histone H3 at amino acid residues H3K27 and H3K9 thus inhibiting the expression of key regulatory genes responsible for cell fate determination and differentiation. Bayliss et al. (2016) revealed the deficiency or complete loss of H3K27me3 in PF-EPN-A tumors [46]. This finding complements the earlier hypotheses on the central role of epigenetic mechanisms in PF-EPN pathogenesis inferred from the absence of presumably pathogenic mutations in chromatin remodeling genes and enzymes that catalyze post-translational modifications (e.g., methylation) of histones in whole-genome sequencing data [17,45,46]. The recruitment of Polycomb group (PcG) transcription repressors to chromatin requires the presence of non-methylated CpGs; accordingly, the loss of H3K27me3 methylation has been associated with dense hypermethylation of CpG-islands preventing the recruitment of PcG proteins to chromatin by steric hindrance.
An advanced investigation of molecular mechanisms responsible for the observed epigenetic malfunctioning revealed a plausible association of the H3K27me3 deficiency with elevated expression levels of accessory proteins encoded by EZHIP (formerly CXorf67) and EPOP (formerly C17orf96) [47,48,49]. As demonstrated by Hübner et al. (2019), EZHIP is a competitive inhibitor of PRC2. A conservative stretch of amino acids in the C-terminal portion of EZHIP mimics the K27 methylation target in histone H3, albeit with K27M substitution. The binding of methionine M27 (instead of lysine K27) to the active center in the histone-lysine N-methyltransferase subunit of PRC2 blocks its catalytic activity [50]. Somatic missense mutations in EZHIP are detected in a small proportion of PF-EPN-A tumors (<10%) [48]. Jain et al. (2019) demonstrated that such mutations have no influence on H3K27me3 levels thus disproving their functional significance [51]. Noteworthy, no loss-of-function mutations in EZHIP (nonsense substitutions or frameshift indels) have been reported. Elevated expression of EZHIP in tumors may be caused by mutations in cis-regulatory elements; the same effect may be conferred by the formation of fusion genes involving EZHIP locus (for instance, MBTD1EZHIP fusion described for low-grade endometrial stromal sarcoma [52]). However, no fusions comprising EZHIP or PRC2 subunit-encoding genes (e.g., EED, SUZ12) have been described for EPNs.
Related signatures of disrupted epigenetic regulation have been associated with H3 K27M mutations typical for diffuse midline gliomas (DMGs) but rarely found in PF-EPN-A (<5% of the cases). Noteworthy, in EPNs such mutations are harbored by canonical histone-encoding genes HIST1H3C and HIST1H3B, whereas in DMGs they are predominantly found in a replacement histone gene H3F3A (90% of the cases) [48,53,54,55]. Given the mutually agonistic roles of the onco-histone H3 K27M and EZHIP, it would be natural to expect similar patterns of disease progression and therapy outcomes for H3 K27M-mutant DMG and EZHIPhigh PF-EPN-A. Indeed, in DMG, disruption of H3K27me3-mediated epigenetic regulation is associated with an extremely aggressive course of the disease, typically presenting with sustained tumor growth and polychemotherapy resistance [56,57,58]. Similarly, effective chemotherapy regimens for PF-EPN-A are missing [59] and therapeutic options for relapses are extremely limited [5,60,61,62,63].
Despite the uniformity of methylation profiles within PF-EPN-A, tumors of this group show considerable molecular heterogeneity and can be additionally classified into two major subgroups A1 and A2 (and ultimately into nine minor subtypes by using additional markers: gains 1q, deletions 22q, 6q, and 10q, and OTX2 protein expression). PF-EPN-A1 tumors are distinguished by pronounced expression of the homeotic HOX genes (HOXA1/2/3/4, HOXB2/3/4, HOXC4, and HOXD4) which define the segmental (rhombomeric) organization of the hindbrain in early embryogenesis. PF-EPN-A2 tumors hyperexpress EN2, CNPY1, and IRX3—a group of genes involved in the rhombomere differentiation. Expression of A1- and A2-specific genes within the developing hindbrain shows distinct zonality—increased expression of A2 markers is characteristic of the rostral portion at the border with the midbrain, while expression of HOX genes is more pronounced in caudal segments of the brainstem and spinal cord. Differential expression of the spatial patterning genes in A1 and A2 tumors apparently reflects their origin from different hindbrain structures. However, the practical relevance of the advanced A1/A2 subgrouping is questionable. Stratification by clinical factors (gender, age at diagnosis, tumor resection volume, and received therapy) revealed no significant differences between A1 and A2 tumors, except the patterns of relapse (PF-EPN-A1 tumors more often produce local than distant relapses, and vice versa) [48].

3.2. PF-EPN-B Group

In contrast to PF-EPN-A tumors which predominantly affect children, PF-EPN-B tumors are more common in adults. In adolescents (aged 10–17), about 45% of newly diagnosed EPNs fall into this group. The prognosis for PF-EPN-B tumors is favorable: 10-year OS rates for the patients after subtotal and gross total resections reach 66.7% and 96.1%, respectively [3,14,15,64]. Thus, the prognosis for this group strongly depends on the extent of surgical resection. The occurrence of delayed relapses (10 years after the onset) underscores the importance of long-term follow-up [64]. Patients with R0 may benefit from chemo- and radiation-sparing strategies; such possibility is being considered [38]. The observed difference in patterns of recurrence between PF-EPN-A and PF-EPN-B adds to the relevance of comprehensive molecular characterization of a tumor as early as possible.
By now, recurrent mutations or fusion genes in PF-EPN-B tumors are missing, and no clear drivers for this group have been identified. Ciliogenesis and microtubule assembly are deregulated only in PF-EPN-B tumors, while several canonical cancer-associated pathways operate in the PF-EPN-A group (VEGF, PDGF, EGFR, RAS signaling, etc.) [14]. PF-EPN-B tumors harbor major cytogenetic aberrations including gains 1q, monosomies 6, 10, and 17, trisomies 5, 8, and 18, and deletions 22q [64]. The diversity of cytogenetic profiles revealed for PF-EPN-B indicate inherent genomic instability and suggest that these tumors emerge from multiple driving events. Similarly with PF-EPN-A, the PF-EPN-B group shows significant heterogeneity, with distinct molecular subtypes of different demographics, copy number alterations, and gene expression signatures. By contrast with PF-EPN-A, gains 1q pose no extra risks for PF-EPN-B tumors. Losses 13q may represent a more reliable negative prognostic marker than gains 1q; however, this assumption requires further substantiation, particularly as the basis for de-escalation of therapy regimens. However, the extent of resection remains the strongest predictor of poor outcomes for this group. Given the patient data scarcity, advanced stratification within PF-EPN-B remains clinically irrelevant [64].

3.3. ST-EPN-ZFTA-like PF-EPNs

Unique cases of ZFTAMAML2, ZFTARELA, and ZFTANCOA2 fusion in PF-EPN were reported recently. These tumors revealed characteristic ZFTA-mediated gene expression and whole-genome DNA methylation signatures corresponding to the ST-EPN-ZFTA group; accordingly, they were classified as “ST-EPN-ZFTA” despite the infratentorial localization [65].
A summary of the intracranial EPN classification is given in Figure 1.

4. Molecular Profiles of Sp-EPNs

Sp-EPNs constitute a heterogeneous group with a generally favorable prognosis. These tumors mostly occur in adult patients and are rare in children. Clinical outcomes for Sp-EPNs are better than for intracranial EPNs, with 5-year OS rates within the range of 60–90% [14]. Three molecular groups of Sp-EPNs were originally identified, including subependymomas, myxopapillary Sp-EPNs (SP-MPE), and Sp-EPNs per se; the molecular subgrouping shows excellent concordance with corresponding histopathological subtypes [14]. Our knowledge on the molecular pathogenesis of Sp-EPN tumors is limited. The groups reveal characteristic somatic copy number aberrations; most Sp-EPNs harbor 22q deletions involving neurofibromin 2 (NF2) tumor suppressor gene, whereas SP-MPEs show chromosomal instability.
SP-MPEs, the most prevalent type of pediatric spinal cord EPNs, predominantly arise in the conus medullaris, cauda equina, and filum terminale regions [66]. Despite their low mitotic index and slow-growing nature, SP-MPEs generally have far more aggressive behavior than other low-grade CNS tumors. Furthermore, pediatric SP-MPEs are especially aggressive, with much higher rates of local recurrence and secondary seeding to distant craniospinal sites or local spinal sites (64% cf. 32% in adults) [67]. As demonstrated by Ahmad et al. (2021), pediatric SP-MPEs exhibit aberrant activity of the mitochondrial metabolic pathways [68]. The only recurrent focal amplification identified for this group involves HOXB gene cluster mapping to 17q. HOXB13 amplification represents a candidate diagnostic marker for SP-MPEs. The elevated expression of HOXB13 enhances tumor cell proliferation and dissemination, playing a critical role in the development of metastasis [68]. Due to the high propensity for local recurrence and distant neural axis dissemination, the summary of the upcoming WHO CNS5 identifies SP-MPEs with grade 2 (rather than grade 1). SP-MPEs have distinctive histopathological features such as well-organized papillary architecture, with vascular cores and abundant mucinous extracellular matrix. Histological examination of tumor tissue is necessary and sufficient for the diagnosis of SP-MPE, whereas genetic testing is accessory [9].
A rare subtype of Sp-EPN in adult patients has been described recently, presenting highly aggressive clinical behavior with early metastasis, diffuse leptomeningeal spread throughout CNS, and resistance to standard treatment protocols. All of them harbored MYCN amplification and no other recurrent pathogenic events [69,70,71]. Importantly, these tumors formed a distinct methylation cluster of their own, and none of them clustered with any of the previously identified nine EPN groups. Recognizing the importance of clinical and molecular data on such tumors, the summary of the upcoming WHO CNS5 reports a novel nosological entity of MYCN-driven Sp-EPNs with dismal outcomes [9].
Genetic alterations found in particular in EPN groups are summarized in Table 1. Subgroup-specific diagnostic and candidate genes in pediatric EPNs are contained in Table 2.

5. Molecular Profiles of Subependymomas

Subependymomas are exceptionally rare slow-growing benign neoplasms, accounting for only 0.07–0.7% of all intracranial tumors [14,72]. These tumors typically arise in adults (aged 22–76 years) without strict predilection to a particular site in CNS. The most typical locations, the fourth and lateral ventricles, are encountered in up to 85% of the cases, followed by septum pellucidum, brainstem, and spine [73]. DNA methylation profiles of subependymomas differ depending on tumor localization, which provides certain grounds for molecular subgrouping [14]. However, all subependymomas have a favorable prognosis independently of localization. According to the cIMPACT-NOW group recommendations and the summary of the upcoming WHO CNS5, morphological examination provides adequate means for the diagnosis and prognostication; the integrative histo-molecular approach for subependymomas is accessory [9,16]. Recent findings suggest that the process of clonal evolution in subependymomas may give rise to more aggressive tumor clones enriched with pure EPN phenotypes, chromosome 6 losses, and TERT mutations. These markers, associated with increased risks of recurrence, should be considered as an indication for more intensive therapies, especially under conditions of subtotal tumor resection [44].

6. Laboratory Approaches for EPN Diagnostics

From a histological perspective, EPNs show moderate cellularity and variable mitotic activity; they consist of monomorphic rounded or oval cells with scant cytoplasm and vesicular nuclei containing granular (salt-and-pepper) chromatin. Key histological features of EPNs are perivascular (pseudo) rosettes and ependymal rosettes. EPNs are positive for glial fibrillary acidic protein (GFAP), S100, vimentin, rarely cytokeratin, and epithelial membrane antigen (EMA, positive along the luminal surface of ependymal rosettes or as characteristic dot-like or ring-like intracytoplasmic patterns) and negative for most neuronal antigens [6].
No unified standards for molecular diagnostics of EPNs have been introduced so far. The molecular group for each particular case is determined by whole-genome DNA methylation profiling as a golden standard. Comprehensive analysis of DNA methylation signatures is the method of choice for differential diagnosis within a broad spectrum of glial and embryonal tumors including CNS neuroblastoma with FOXR2 activation, CNS Ewing sarcoma family tumor with CIC alteration, CNS high-grade neuroepithelial tumor with MN1 alteration, and CNS high-grade neuroepithelial tumor with BCOR alteration [24,74]. The advantage of using this technique is the suitability of formalin-fixed paraffin-embedded tumor tissue for the analysis (methylated sites in genomic DNA are preserved during fixation, dehydration, etc.). However, the use of DNA methylation assays in routine laboratory practice has serious limitations, as the data processing is complex and the method itself is expensive, sophisticated, and labor-consuming. Alternative diagnostic algorithms may be based on a combination of economically justified methods including histological examination, immunohistochemistry (IHC) testing, reverse transcription-polymerase chain reaction (RT-PCR), Sanger sequencing, fluorescence in situ hybridization (FISH), and probably also the NanoString nCounter® platform (Figure 2).

6.1. Differential Diagnosis of ST-EPNs

In many cases, unambiguous determination of the molecular group of a tumor can be afforded by a certain combination of routine morphological and genetic approaches [75]. For instance, nuclear accumulation of p65 (RelA) and the presence of ZFTARELA fusion are sufficient for the identification of the sample with the core ST-EPN-ZFTA group harboring the classical ZFTARELA transcript. The diversity of fusion transcripts, which results from the variability of breakpoints in RELA and its partner genes, significantly complicates the identification of particular rearrangements and their use as markers. Conventional RT-PCR tests are targeted at the two most frequent fusions ZFTA (exon 2)–RELA (exon 2) and ZFTA (exon 3)–RELA (exon 2) [19,75,76].
A highly efficient way of screening for ZFTA and RELA rearrangements is provided by FISH with break-apart probes for one of the fusion partner genes. This approach allows detecting rearrangement of the gene of interest without accurate determination of the breakpoint [19,21]. Its excellent concordance with other methods including nuclear expression of RelA and DNA methylation profiling should be noted [19].
ZFTA gene rearrangements induce the hyperexpression of L1 cell adhesion molecule (L1CAM) and/or cyclin D1 may be considered as a surrogate marker of the ST-EPN-ZFTA group [18,20,22,23]. However, IHC is not sufficient on its own: the results require confirmation by independent alternative methods since neither of the antibodies (anti-p65, anti-L1CAM, anti-cyclin D1) has enough sensitivity and/or specificity to reliably verify the ST-EPN-ZFTA group [20,22]. The diagnosis of ST-EPN-ZFTA with alternative (non-RELA) fusion gene can be suspected if tumor cells show immunopositivity for L1CAM and negativity for p65 [26].
Identification of a tumor with the ST-EPN-YAP1 group is based on detection of either YAP1 rearrangement by FISH or one of the YAP1-fusions (YAP1MAMLD1, YAP1FAM118B) by RT-PCR, while IHC with anti-YAP1 and/or anti-Claudin-1 is noninformative [19,37].

6.2. Differential Diagnosis of PF-EPNs

Panwalkar et al. (2017) proposed IHC tests for H3K27me3 as a straightforward and affordable method for PF-EPN stratification. The authors demonstrate that PF-EPN-B tumors are completely positive for H3K27me3, whereas the presence of H3K27me3 signal in less than 80% of tumor cell nuclei (against the total positivity of endothelial cells used as an internal control) is indicative of PF-EPN-A. Moreover, groupings based on whole-genome DNA methylation profiling vs. IHC tests for H3K27me3 were 99.1% concordant; the outlier was H3K27me3-negative tumor from a 12-year-old female patient, classified as PF-EPN-B on the basis of DNA methylation analysis [77]. The methodology was further enhanced by Fukuoka et al. (2018) who attempted at finer quantitation of the IHC data. In about 62% of prediagnosed PF-EPN-A tumors examined by the authors, the H3K27me3 reactivity was shown by 5–50% of the tumor cell nuclei; for the rest of PF-EPN-A tumors (about 40%), the proportion was less than 5%. In contrast, the vast majority of PF-EPN-B tumors showed intact expression of H3K27me3, with 90–95% of tumor cell nuclei stained positive, except two cases of reduced positivity (10–60%) classified as PF-EPN-B by DNA methylation profiling. Overall, the cut-off level of 80% H3K27me3-positive tumor cells allowed distinguishing PF-EPN-B from PF-EPN-A with 100% specificity and 86.7% sensitivity [21].
Antin et al. (2020) suggested complementing the routine IHC panels for CNS tumor diagnostics with anti-EZHIP staining. The intensive diffuse nuclear staining with >90% tumor cells immunopositive was obtained for PF-EPN-A (with the exception of rare H3 K27M-mutant tumors), diffuse midline gliomas with wild-type (non-mutant) H3F3A, and germinomas [78]. Nambirajan et al. (2021) evaluated an extended panel of IHC markers (H3K27me3, acetyl-H3K27, H3K27M, ATRX, EZHIP, EPOP, and Tenascin-C) for the use in differential diagnostics of PF-EPNs. The authors demonstrated that a combination of EZHIP- and H3K27me3-specific antibodies is sufficient for reliable verification of PF-EPN-A; this finding illustrates the consistency between genetic and IHC profiles of a tumor [79].
Given the existence of rare EPN subtypes e.g., PF-EPN with ZFTA rearrangement [65] or ST-EPN harboring ZFTA fusions with atypical (non-RELA) partners, correct identification of a tumor with a particular group requires an extended diagnostic algorithm accounting for gene expression signatures. The creation of a universal diagnostic tool for the determination of molecular subtypes of EPN can be based on the analysis of gene expression signatures using the NanoString nCounter® platform. The assay must be carried out with customized panels of markers, corresponding to EPN group-specific signatures of upregulated genes, designed specifically for this purpose. Lastowska et al. (2021) identified group-specific marker genes for RELA-fused EPN (RELA, ELL3, FBP2, PCP4L1, and MYO3A), YAP1-fused EPN (MRAP, IGF1, CAPS, and WWC1), PF-EPN-A (LAMA2, ALDH1L1, SLC6A13, IGSF1, and CXorf67), and PF-EPN-B (NELL2, DNAH1, CEP83, C9orf72, and NXNL2) tumors. The NanoString-based approach clearly separated PF-EPNs into two clusters, corresponding to the defined PF-EPN groups, based on the expression of selected genes. Extended cluster analysis allowed subdivision of PF-EPN-A tumors into PF-EPN-A1 and PF-EPN-A2 subgroups using four marker genes (SKAP2, WIF1, and EN2, CNPY1, respectively). The described gene panel for RELA-positive EPN did not allow differentiation of EPN with ZFTAMAML2 fusion [80]. The potential of the NanoString method for diagnosis and advanced classification of other CNS tumors has been confirmed in several studies [81,82,83]. The approach provides reliable stratification of medulloblastomas into four groups (designated WNT, SHH, group 3, and group 4) based on the evaluation of 22 transcripts expressed differentially among the groups and contributing to characteristic group-specific gene expression signatures [81,82]. Primitive neuroectodermal tumors of the CNS (CNS-PNETs) have been already defined as four new molecularly defined entities (CNS neuroblastoma with FOXR2 activation, CNS Ewing sarcoma family tumor with CIC alteration, CNS high-grade neuroepithelial tumor with MN1 alteration, and CNS high-grade neuroepithelial tumor with BCOR alteration) using a single multi-gene tumor-specific signature [83]. However, differential diagnostics of EPNs in mixed series with other CNS tumors by the NanoString approach is impossible. For correct molecular stratification of ependymal tumors by the NanoString approach, all samples intended for comparison must be morphologically identified as EPN prior to the analysis.

7. Therapeutic Targeting of EPNs

Despite the profound insights into ependymal tumor biology, consensus recommendations for the management of patients with EPNs with regard to molecular diagnosis are missing. As tumors are resistant to conventional chemotherapy, the search for druggable targets is highly relevant. However, no candidate targets have been identified despite the extensive efforts in molecular profiling. Clinical studies on general cohorts showed no significant objective response to ERBB1/2 (lapatinib) and VEGF inhibitors (bevacizumab, sunitinib), despite the evidence on overexpression of ERBB2, ERBB4, or VEGF in EPNs [84,85,86,87].
In the molecular era, cancer-specific somatic aberrations should be taken into account when choosing a treatment strategy. For PF-EPNs, the pathogenesis of which involves epigenetic mechanisms, the possibility of using pharmaceuticals targeted at epigenetic modifications, including abnormalities of DNA methylation and histone modifications, seems most rational. Despite the promising results of pre-clinical research [88,89], histone deacetylase (HDAC) inhibitors showed no therapeutic activity in patients, apparently due to the intricacies of subcellular localization of HDACs and inability of the inhibitors to accumulate inside the brain tissue in concentrations sufficient for therapeutic response [90]. Nevertheless, the search for new HDAC inhibitors with appropriate brain-penetrating capacities and safety profiles may provide a useful treatment strategy [91]. The possibility of using BET-bromodomain inhibitors as anticancer therapeutics is being investigated in pediatric EPN stem cell models [92]. The upregulation of EZHIP could have important implications for therapeutic approaches. As reported by Han et al. (2020), EZHIP has a conservative PALB2-binding domain which enables its functioning as a competitive inhibitor of BRCA2. Elevated levels of EZHIP prevent the formation of BRCA1-PALB2-BRCA2 complexes thus inhibiting the homologous recombination-mediated DNA repair pathway. The results indicate the potential of PARP inhibitors as targeted therapeutics in PF-EPN-A, especially when combined with radiotherapy [61].
NF-kappa B inhibitors are considered potential therapeutic agents for the treatment of ST-EPN-ZFTA with constitutive activation of the NF-κB signaling pathway. A trial currently in phase II is evaluating the effectiveness of marizomib—a second-generation irreversible proteasome inhibitor, enrolling patients with histologically proven spinal or intracranial EPN, including but not limited to ST-EPN-ZFTA (ClinicalTrials.gov Identifier: NCT03727841, accessed on 20.09.2021). At the moment, the enrollment is complete, and the study is in progress. Specific expression of PD-L1 on both tumor and myeloid cells in ST-EPN-RELA has been demonstrated, accompanied by high levels of PD-1 expressed by tumor-infiltrating T cells (both CD4 and CD8) [93,94]. In the context of immunotherapy, ST-EPN-RELA progression may be controlled with PD-1 inhibitors, such as pembroluzimab or nivolumab [95].
Despite the principal shift in the EPN diagnostics and molecular stratification, its immediate impact on the existing treatment regimens is low. The correction would require preclinical and clinical trials for EPNs with due consideration of the molecular subgrouping.

8. Conclusions

As demonstrated by advanced studies of the last decade, ependymomas constitute a heterogeneous group of tumors and differ by molecular etiology. This minireview underscores the importance of comprehensive molecular profiling for ependymal tumors aimed at identifying specific expression signatures and/or (epi)genetic variants. Molecular identification of an ependymal tumor with a particular molecular group should follow its anatomical and histopathological assessment. Advanced stratification of patients into risk groups provides a framework for personalized management, e.g., allows de-escalation of the therapy in patients with low-risk tumors (supratentorial ependymomas group YAP1 and infratentorial ependymomas group B). Detailed understanding of causative molecular abnormalities for particular tumors is pivotal for the development of novel therapeutic options.

Author Contributions

Conceptualization, M.Z.; writing—original draft preparation, M.Z., L.P. and A.D.; writing—review and editing M.Z., L.P., G.N. and A.D.; supervision G.N. and A.D. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by Foundation for support and development in the field of pediatric hematology, oncology and immunology “Science for Children”.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Ostrom, Q.T.; Patil, N.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013–2017. Neuro-Oncol. 2020, 22 (Suppl. 1), iv1–iv96. [Google Scholar] [CrossRef] [PubMed]
  2. Jünger, S.T.; Mynarek, M.; Wohlers, I.; Dörner, E.; Mühlen, A.Z.; Velez-Char, N.; von Hoff, K.; Rutkowski, S.; Warmuth-Metz, M.; Kortmann, R.D.; et al. Improved Risk-Stratification for Posterior Fossa Ependymoma of Childhood Considering Clinical, Histological and Genetic Features—A Retrospective Analysis of the HIT Ependymoma Trial Cohort. Acta Neuropathol. Commun. 2019, 7, 181. [Google Scholar] [CrossRef] [PubMed]
  3. Zapotocky, M.; Beera, K.; Adamski, J.; Laperierre, N.; Guger, S.; Janzen, L.; Lassaletta, A.; Figueiredo Nobre, L.; Bartels, U.; Tabori, U.; et al. Survival and functional outcomes of molecularly defined childhood posterior fossa ependymoma: Cure at a cost. Cancer 2019, 125, 1867–1876. [Google Scholar] [CrossRef] [PubMed]
  4. Massimino, M.; Barretta, F.; Modena, P.; Witt, H.; Minasi, S.; Pfister, S.M.; Pajtler, K.W.; Antonelli, M.; Gandola, L.; Luisa Garrè, M.; et al. Second series by the Italian Association of Pediatric Hematology and Oncology of children and adolescents with intracranial ependymoma: An integrated molecular and clinical characterization with a long-term follow-up. Neuro Oncol. 2021, 23, 848–857. [Google Scholar] [CrossRef]
  5. Ritzmann, T.A.; Rogers, H.A.; Paine, S.M.L.; Storer, L.C.D.; Jacques, T.S.; Chapman, R.J.; Ellison, D.; Donson, A.M.; Foreman, N.K.; Grundy, R.G. A Retrospective Analysis of Recurrent Pediatric Ependymoma Reveals Extremely Poor Survival and Ineffectiveness of Current Treatments across Central Nervous System Locations and Molecular Subgroups. Pediatr. Blood Cancer 2020, 67, e28426. [Google Scholar] [CrossRef]
  6. Ellison, D.W.; Kocak, M.; Figarella-Branger, D.; Felice, G.; Catherine, G.; Pietsch, T.; Frappaz, D.; Massimino, M.; Grill, J.; Boyett, J.M.; et al. Histopathological Grading of Pediatric Ependymoma: Reproducibility and Clinical Relevance in European Trial Cohorts. J. Negat. Results BioMed 2011, 10, 7. [Google Scholar] [CrossRef]
  7. Upadhyaya, S.A.; Robinson, G.W.; Onar-Thomas, A.; Orr, B.A.; Billups, C.A.; Bowers, D.C.; Bendel, A.E.; Hassall, T.; Crawford, J.R.; Partap, S.; et al. Molecular Grouping and Outcomes of Young Children with Newly Diagnosed Ependymoma Treated on the Multi-Institutional SJYC07 Trial. Neuro-Oncology 2019, 21, 1319–1330. [Google Scholar] [CrossRef]
  8. Louis, D.N.; Wesseling, P.; Aldape, K.; Brat, D.J.; Capper, D.; Cree, I.A.; Eberhart, C.; Figarella-Branger, D.; Fouladi, M.; Fuller, G.N.; et al. cIMPACT-NOW update 6: New entity and diagnostic principle recommendations of the cIMPACT-Utrecht meeting on future CNS tumor classification and grading. Brain Pathol. 2020, 30, 844–856. [Google Scholar] [CrossRef]
  9. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  10. Leu, S.; von Felten, S.; Frank, S.; Boulay, J.L.; Mariani, L. IDH Mutation Is Associated with Higher Risk of Malignant Transformation in Low-Grade Glioma. J. Neurooncol. 2016, 127, 363–372. [Google Scholar] [CrossRef]
  11. Jairam, V.; Kann, B.H.; Park, H.S.; Miccio, J.A.; Beckta, J.M.; Yu, J.B.; Prabhu, R.S.; Gao, S.J.; Mehta, M.P.; Curran, W.J.; et al. Defining an Intermediate-Risk Group for Low-Grade Glioma: A National Cancer Database Analysis. Anticancer Res. 2019, 39, 2911–2918. [Google Scholar] [CrossRef] [Green Version]
  12. Yang, R.R.; Aibaidula, A.; Wang, W.W.; Chan, A.K.; Shi, Z.F.; Zhang, Z.Y.; Chan, D.; Poon, W.S.; Liu, X.Z.; Li, W.C.; et al. Pediatric low-grade gliomas can be molecularly stratified for risk. Acta Neuropathol. 2018, 136, 641–655. [Google Scholar] [CrossRef]
  13. Schwalbe, E.C.; Lindsey, J.C.; Nakjang, S.; Crosier, S.; Smith, A.J.; Hicks, D.; Rafiee, G.; Hill, R.M.; Iliasova, A.; Stone, T.; et al. Novel Molecular Subgroups for Clinical Classification and Outcome Prediction in Childhood Medulloblastoma: A Cohort Study. Lancet Oncol. 2017, 18, 958–971. [Google Scholar] [CrossRef] [Green Version]
  14. Pajtler, K.W.; Witt, H.; Sill, M.; Jones, D.T.W.; Hovestadt, V.; Kratochwil, F.; Wani, K.; Tatevossian, R.; Punchihewa, C.; Johann, P.; et al. Molecular Classification of Ependymal Tumors across All CNS Compartments, Histopathological Grades, and Age Groups. Cancer Cell 2015, 27, 728–743. [Google Scholar] [CrossRef] [Green Version]
  15. Ramaswamy, V.; Hielscher, T.; Mack, S.C.; Lassaletta, A.; Lin, T.; Pajtler, K.W.; Jones, D.T.W.; Luu, B.; Cavalli, F.M.G.; Aldape, K.; et al. Therapeutic Impact of Cytoreductive Surgery and Irradiation of Posterior Fossa Ependymoma in the Molecular Era: A Retrospective Multicohort Analysis. J. Clin. Oncol. 2016, 34, 2468–2477. [Google Scholar] [CrossRef]
  16. Ellison, D.W.; Aldape, K.D.; Capper, D.; Fouladi, M.; Gilbert, M.R.; Gilbertson, R.J.; Hawkins, C.; Merchant, T.E.; Pajtler, K.; Venneti, S.; et al. CIMPACT-NOW Update 7: Advancing the Molecular Classification of Ependymal Tumors. Brain Pathol. 2020, 30, 863–866. [Google Scholar]
  17. Parker, M.; Mohankumar, K.M.; Punchihewa, C.; Weinlich, R.; Dalton, J.D.; Li, Y.; Lee, R.; Tatevossian, R.G.; Phoenix, T.N.; Thiruvenkatam, R.; et al. C11orf95–RELA Fusions Drive Oncogenic NF-ΚB Signalling in Ependymoma. Nature 2014, 506, 451–455. [Google Scholar] [CrossRef] [Green Version]
  18. Malgulwar, P.B.; Nambirajan, A.; Pathak, P.; Faruq, M.; Rajeshwari, M.; Singh, M.; Suri, V.; Sarkar, C.; Sharma, M.C. C11orf95-RELA Fusions and Upregulated NF-KB Signalling Characterise a Subset of Aggressive Supratentorial Ependymomas That Express L1CAM and Nestin. J. Neurooncol. 2018, 138, 29–39. [Google Scholar] [CrossRef]
  19. Pagès, M.; Pajtler, K.W.; Puget, S.; Castel, D.; Boddaert, N.; Tauziède-Espariat, A.; Picot, S.; Debily, M.; Kool, M.; Capper, D.; et al. Diagnostics of Pediatric Supratentorial RELA Ependymomas: Integration of Information from Histopathology, Genetics, DNA Methylation and Imaging. Brain Pathol. 2019, 29, 325–335. [Google Scholar] [CrossRef] [Green Version]
  20. Torre, M.; Alexandrescu, S.; Dubuc, A.M.; Ligon, A.H.; Hornick, J.L.; Meredith, D.M. Characterization of Molecular Signatures of Supratentorial Ependymomas. Mod. Pathol. 2020, 33, 47–56. [Google Scholar] [CrossRef]
  21. Fukuoka, K.; Kanemura, Y.; Shofuda, T.; Fukushima, S.; Yamashita, S.; Narushima, D.; Kato, M.; Honda-Kitahara, M.; Ichikawa, H.; Kohno, T.; et al. Significance of Molecular Classification of Ependymomas: C11orf95-RELA Fusion-Negative Supratentorial Ependymomas Are a Heterogeneous Group of Tumors. Acta Neuropathol. Commun. 2018, 6, 134. [Google Scholar] [CrossRef] [Green Version]
  22. Gessi, M.; Giagnacovo, M.; Modena, P.; Elefante, G.; Gianno, F.; Buttarelli, F.R.; Arcella, A.; Donofrio, V.; Diomedi Camassei, F.; Nozza, P.; et al. Role of Immunohistochemistry in the Identification of Supratentorial C11ORF95-RELA Fused Ependymoma in Routine Neuropathology. Am. J. Surg. Pathol. 2019, 43, 56–63. [Google Scholar] [CrossRef]
  23. Pietsch, T.; Wohlers, I.; Goschzik, T.; Dreschmann, V.; Denkhaus, D.; Dörner, E.; Rahmann, S.; Klein-Hitpass, L. Supratentorial Ependymomas of Childhood Carry C11orf95–RELA Fusions Leading to Pathological Activation of the NF-ΚB Signaling Pathway. Acta Neuropathol. 2014, 127, 609–611. [Google Scholar] [CrossRef]
  24. Zschernack, V.; Jünger, S.T.; Mynarek, M.; Rutkowski, S.; Garre, M.L.; Ebinger, M.; Neu, M.; Faber, J.; Erdlenbruch, B.; Claviez, A.; et al. Supratentorial Ependymoma in Childhood: More than Just RELA or YAP. Acta Neuropathol. 2021, 141, 455–466. [Google Scholar] [CrossRef]
  25. Tamai, S.; Nakano, Y.; Kinoshita, M.; Sabit, H.; Nobusawa, S.; Arai, Y.; Hama, N.; Totoki, Y.; Shibata, T.; Ichimura, K.; et al. Ependymoma with C11orf95-MAML2 Fusion: Presenting with Granular Cell and Ganglion Cell Features. Brain Tumor Pathol. 2021, 38, 64–70. [Google Scholar] [CrossRef]
  26. Tauziède-Espariat, A.; Siegfried, A.; Nicaise, Y.; Kergrohen, T.; Sievers, P.; Vasiljevic, A.; Roux, A.; Dezamis, E.; Benevello, C.; Machet, M.C.; et al. Supratentorial non-RELA, ZFTA-fused ependymomas: A comprehensive phenotype genotype correlation highlighting the number of zinc fingers in ZFTA-NCOA1/2 fusions. Acta Neuropathol. Commun. 2021, 9, 135. [Google Scholar] [CrossRef]
  27. Zheng, T.; Ghasemi, D.R.; Okonechnikov, K.; Korshunov, A.; Sill, M.; Maass, K.K.; Goncalves, B.; da Silva, P.; Ryzhova, M.; Gojo, J.; et al. Cross-species genomics reveals oncogenic dependencies in ZFTA/C11orf95 fusion-positive supratentorial ependymomas. Cancer Discov. 2021, 11, 2230–2247. [Google Scholar] [CrossRef]
  28. Tomomasa, R.; Arai, Y.; Kawabata-Iwakawa, R.; Fukuoka, K.; Nakano, Y.; Hama, N.; Nakata, S.; Suzuki, N.; Ishi, Y.; Tanaka, S.; et al. Ependymoma-like tumor with mesenchymal differentiation harboring C11orf95-NCOA1/2 or -RELA fusion: A hitherto unclassified tumor related to ependymoma. Brain Pathol. 2021, 31, e12943. [Google Scholar] [CrossRef]
  29. de Sousa, G.R.; Marie, S.; Oba-Shinjo, S.M.; Ramalho, L.; Tone, L.G.; Valera, E.T. A novel type of C11orf95-LOC-RELA fusion in a grade II supratentorial ependymoma: Report of a case with literature review. Childs Nerv. Syst. 2019, 35, 689–694. [Google Scholar] [CrossRef]
  30. Zhu, J.J.; Jillette, N.; Li, X.N.; Cheng, A.W.; Lau, C.C. C11orf95-RELA reprograms 3D epigenome in supratentorial ependymoma. Acta Neuropathol. 2020, 140, 951–960. [Google Scholar] [CrossRef]
  31. Arabzade, A.; Zhao, Y.; Varadharajan, S.; Chen, H.-C.; Jessa, S.; Rivas, B.; Stuckert, A.J.; Solis, M.; Kardian, A.; Tlais, D.; et al. ZFTA-RELA Dictates Oncogenic Transcriptional Programs to Drive Aggressive Supratentorial Ependymoma. Cancer Discov. 2021, 11, 2200–2215. [Google Scholar] [CrossRef] [PubMed]
  32. Godwin, P.; Baird, A.M.; Heavey, S.; Barr, M.P.; O’Byrne, K.J.; Gately, K. Targeting Nuclear Factor-Kappa B to Overcome Resistance to Chemotherapy. Front. Oncol. 2013, 3, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ozawa, T.; Kaneko, S.; Szulzewsky, F.; Qiao, Z.; Takadera, M.; Narita, Y.; Kondo, T.; Holland, E.C.; Hamamoto, R.; Ichimura, K. C11orf95-RELA Fusion Drives Aberrant Gene Expression through the Unique Epigenetic Regulation for Ependymoma Formation. Acta Neuropathol. Commun. 2021, 9, 36. [Google Scholar] [CrossRef] [PubMed]
  34. Merchant, T.E.; Bendel, A.E.; Sabin, N.D.; Burger, P.C.; Shaw, D.W.; Chang, E.; Wu, S.; Zhou, T.; Eisenstat, D.D.; Foreman, N.K.; et al. Conformal Radiation Therapy for Pediatric Ependymoma, Chemotherapy for Incompletely Resected Ependymoma, and Observation for Completely Resected, Supratentorial Ependymoma. J. Clin. Oncol. 2019, 37, 974–983. [Google Scholar] [CrossRef] [Green Version]
  35. Pajtler, K.W.; Wei, Y.; Okonechnikov, K.; Silva, P.B.G.; Vouri, M.; Zhang, L.; Brabetz, S.; Sieber, L.; Gulley, M.; Mauermann, M.; et al. YAP1 Subgroup Supratentorial Ependymoma Requires TEAD and Nuclear Factor I-Mediated Transcriptional Programmes for Tumorigenesis. Nat. Commun. 2019, 10, 3914. [Google Scholar] [CrossRef] [Green Version]
  36. Wang, J.; Wang, L.; Fu, L.; Li, Q.C.; Qiu, X.S.; Wang, E.H.; Yu, J.H. Supratentorial ependymoma with YAP1:FAM118B fusion: A case report. Neuropathology 2021, 41, 133–138. [Google Scholar] [CrossRef]
  37. Andreiuolo, F.; Varlet, P.; Tauziède-Espariat, A.; Jünger, S.T.; Dörner, E.; Dreschmann, V.; Kuchelmeister, K.; Waha, A.; Haberler, C.; Slavc, I.; et al. Childhood Supratentorial Ependymomas with YAP1-MAMLD1 Fusion: An Entity with Characteristic Clinical, Radiological, Cytogenetic and Histopathological Features: YAP1-MAMLD1 Ependymoma. Brain Pathol. 2019, 29, 205–216. [Google Scholar] [CrossRef] [Green Version]
  38. Pajtler, K.W.; Mack, S.C.; Ramaswamy, V.; Smith, C.A.; Witt, H.; Smith, A.; Hansford, J.R.; von Hoff, K.; Wright, K.D.; Hwang, E.; et al. The Current Consensus on the Clinical Management of Intracranial Ependymoma and Its Distinct Molecular Variants. Acta Neuropathol. 2017, 133, 5–12. [Google Scholar] [CrossRef]
  39. Sievers, P.; Henneken, S.C.; Blume, C.; Sill, M.; Schrimpf, D.; Stichel, D.; Okonechnikov, K.; Reuss, D.E.; Benzel, J.; Maaß, K.K.; et al. Recurrent fusions in PLAGL1 define a distinct subset of pediatric-type supratentorial neuroepithelial tumors. Acta Neuropathol. 2021. [Google Scholar] [CrossRef]
  40. Lopez-Nunez, O.; Cafferata, B.; Santi, M.; Ranganathan, S.; Pearce, T.M.; Kulich, S.M.; Bailey, K.M.; Broniscer, A.; Rossi, S.; Zin, A.; et al. The spectrum of rare central nervous system (CNS) tumors with EWSR1-non-ETS fusions: Experience from three pediatric institutions with review of the literature. Brain Pathol. 2021, 31, 70–83. [Google Scholar] [CrossRef]
  41. Siegfried, A.; Rousseau, A.; Maurage, C.A.; Pericart, S.; Nicaise, Y.; Escudie, F.; Grand, D.; Delrieu, A.; Gomez-Brouchet, A.; Le Guellec, S.; et al. EWSR1-PATZ1 gene fusion may define a new glioneuronal tumor entity. Brain Pathol. 2019, 29, 53–62. [Google Scholar] [CrossRef] [Green Version]
  42. Olsen, T.K.; Panagopoulos, I.; Meling, T.R.; Micci, F.; Gorunova, L.; Thorsen, J.; Due-Tønnessen, B.; Scheie, D.; Lund-Iversen, M.; Krossnes, B.; et al. Fusion Genes with ALK as Recurrent Partner in Ependymoma-like Gliomas: A New Brain Tumor Entity? Neuro Oncol. 2015, 17, 1365–1373. [Google Scholar] [CrossRef] [Green Version]
  43. Baroni, L.V.; Sundaresan, L.; Heled, A.; Coltin, H.; Pajtler, K.W.; Lin, T.; Merchant, T.E.; McLendon, R.; Faria, C.; Buntine, M.; et al. Ultra high-risk PFA ependymoma is characterized by loss of chromosome 6q. Neuro Oncol. 2021, 23, 1360–1370. [Google Scholar] [CrossRef]
  44. Thomas, C.; Thierfelder, F.; Träger, M.; Soschinski, P.; Müther, M.; Edelmann, D.; Förster, A.; Geiler, C.; Kim, H.Y.; Filipski, K.; et al. TERT promoter mutation and chromosome 6 loss define a high-risk subtype of ependymoma evolving from posterior fossa subependymoma. Acta Neuropathol. 2021, 141, 959–970. [Google Scholar] [CrossRef]
  45. Mack, S.C.; Witt, H.; Piro, R.M.; Gu, L.; Zuyderduyn, S.; Stütz, A.M.; Wang, X.; Gallo, M.; Garzia, L.; Zayne, K.; et al. Epigenomic Alterations Define Lethal CIMP-Positive Ependymomas of Infancy. Nature 2014, 506, 445–450. [Google Scholar] [CrossRef]
  46. Bayliss, J.; Mukherjee, P.; Lu, C.; Jain, S.U.; Chung, C.; Martinez, D.; Sabari, B.; Margol, A.S.; Panwalkar, P.; Parolia, A.; et al. Lowered H3K27me3 and DNA Hypomethylation Define Poorly Prognostic Pediatric Posterior Fossa Ependymomas. Sci. Transl. Med. 2016, 8, 366ra161. [Google Scholar] [CrossRef] [Green Version]
  47. Beringer, M.; Pisano, P.; Di Carlo, V.; Blanco, E.; Chammas, P.; Vizán, P.; Gutiérrez, A.; Aranda, S.; Payer, B.; Wierer, M.; et al. EPOP Functionally Links Elongin and Polycomb in Pluripotent Stem Cells. Mol. Cell 2016, 64, 645–658. [Google Scholar] [CrossRef] [Green Version]
  48. Pajtler, K.W.; Wen, J.; Sill, M.; Lin, T.; Orisme, W.; Tang, B.; Hübner, J.-M.; Ramaswamy, V.; Jia, S.; Dalton, J.D.; et al. Molecular Heterogeneity and CXorf67 Alterations in Posterior Fossa Group A (PFA) Ependymomas. Acta Neuropathol. 2018, 136, 211–226. [Google Scholar] [CrossRef]
  49. Michealraj, K.A.; Kumar, S.A.; Kim, L.; Cavalli, F.; Przelicki, D.; Wojcik, J.B.; Delaidelli, A.; Bajic, A.; Saulnier, O.; MacLeod, G.; et al. Metabolic Regulation of the Epigenome Drives Lethal Infantile Ependymoma. Cell 2020, 181, 1329–1345.e24. [Google Scholar] [CrossRef]
  50. Hübner, J.M.; Müller, T.; Papageorgiou, D.N.; Mauermann, M.; Krijgsveld, J.; Russell, R.B.; Ellison, D.W.; Pfister, S.M.; Pajtler, K.W.; Kool, M. EZHIP/CXorf67 mimics K27M mutated oncohistones and functions as an intrinsic inhibitor of PRC2 function in aggressive posterior fossa ependymoma. Neuro Oncol. 2019, 21, 878–889. [Google Scholar] [CrossRef]
  51. Jain, S.U.; Do, T.J.; Lund, P.J.; Rashoff, A.Q.; Diehl, K.L.; Cieslik, M.; Bajic, A.; Juretic, N.; Deshmukh, S.; Venneti, S.; et al. PFA Ependymoma-Associated Protein EZHIP Inhibits PRC2 Activity through a H3 K27M-like Mechanism. Nat. Commun. 2019, 10, 2146. [Google Scholar] [CrossRef] [Green Version]
  52. Dewaele, B.; Przybyl, J.; Quattrone, A.; Finalet Ferreiro, J.; Vanspauwen, V.; Geerdens, E.; Gianfelici, V.; Kalender, Z.; Wozniak, A.; Moerman, P.; et al. Identification of a Novel, Recurrent MBTD1-CXorf67 Fusion in Low-grade Endometrial Stromal Sarcoma. Int. J. Cancer 2014, 134, 1112–1122. [Google Scholar] [CrossRef]
  53. Khuong-Quang, D.A.; Buczkowicz, P.; Rakopoulos, P.; Liu, X.Y.; Fontebasso, A.M.; Bouffet, E.; Bartels, U.; Albrecht, S.; Schwartzentruber, J.; Letourneau, L.; et al. K27M Mutation in Histone H3.3 Defines Clinically and Biologically Distinct Subgroups of Pediatric Diffuse Intrinsic Pontine Gliomas. Acta Neuropathol. 2012, 124, 439–447. [Google Scholar] [CrossRef] [Green Version]
  54. Karremann, M.; Gielen, G.H.; Hoffmann, M.; Wiese, M.; Colditz, N.; Warmuth-Metz, M.; Bison, B.; Claviez, A.; van Vuurden, D.G.; von Bueren, A.O.; et al. Diffuse High-Grade Gliomas with H3 K27M Mutations Carry a Dismal Prognosis Independent of Tumor Location. Neuro Oncol. 2018, 20, 123–131. [Google Scholar] [CrossRef] [Green Version]
  55. Zaytseva, M.A.; Shekhtman, A.P.; Papusha, L.I.; Valiakhmetova, E.F.; Yasko, L.A.; Druy, A.E. Analysis of genetic aberrations in pediatric high-grade gliomas. Advances in Molecular Oncology 2020, 7, 37–47. (In Russian) [Google Scholar] [CrossRef]
  56. Infinger, L.; Stevenson, C. Re-Examining the Need for Tissue Diagnosis in Pediatric Diffuse Intrinsic Pontine Gliomas: A Review. Curr. Neuropharmacol. 2016, 15, 129–133. [Google Scholar] [CrossRef] [Green Version]
  57. Lu, V.M.; Alvi, M.A.; McDonald, K.L.; Daniels, D.J. Impact of the H3K27M mutation on survival in pediatric high-grade glioma: A systematic review and meta-analysis. J. Neurosurg. Pediatr. 2018, 23, 308–316. [Google Scholar] [CrossRef] [Green Version]
  58. Mosaab, A.; El-Ayadi, M.; Khorshed, E.N.; Amer, N.; Refaat, A.; El-Beltagy, M.; Hassan, Z.; Soror, S.H.; Zaghloul, M.S.; El-Naggar, S. Histone H3K27M Mutation Overrides Histological Grading in Pediatric Gliomas. Sci. Rep. 2020, 10, 8368. [Google Scholar] [CrossRef]
  59. Rudà, R.; Reifenberger, G.; Frappaz, D.; Pfister, S.M.; Laprie, A.; Santarius, T.; Roth, P.; Tonn, J.C.; Soffietti, R.; Weller, M.; et al. EANO guidelines for the diagnosis and treatment of ependymal tumors. Neuro Oncol. 2018, 20, 445–456. [Google Scholar] [CrossRef] [Green Version]
  60. Merchant, T.E.; Boop, F.A.; Kun, L.E.; Sanford, R.A. A retrospective study of surgery and reirradiation for recurrent ependymoma. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 87–97. [Google Scholar] [CrossRef]
  61. Han, J.; Yu, M.; Bai, Y.; Yu, J.; Jin, F.; Li, C.; Zeng, R.; Peng, J.; Li, A.; Song, X.; et al. Elevated CXorf67 Expression in PFA Ependymomas Suppresses DNA Repair and Sensitizes to PARP Inhibitors. Cancer Cell 2020, 38, 844–856.e7. [Google Scholar] [CrossRef] [PubMed]
  62. Tsai, J.W.; Manoharan, N.; Alexandrescu, S.; Zimmerman, M.A.; Scully, J.; Chordas, C.; Clymer, J.; Wright, K.D.; Filbin, M.; Ullrich, N.J.; et al. Outcomes after first relapse of childhood intracranial ependymoma. Pediatr. Blood Cancer 2021, 68, e28930. [Google Scholar] [CrossRef] [PubMed]
  63. Adolph, J.E.; Fleischhack, G.; Mikasch, R.; Zeller, J.; Warmuth-Metz, M.; Bison, B.; Mynarek, M.; Rutkowski, S.; Schüller, U.; von Hoff, K.; et al. Local and systemic therapy of recurrent ependymoma in children and adolescents: Short- and long-term results of the E-HIT-REZ 2005 study. Neuro Oncol. 2021, 23, 1012–1023. [Google Scholar] [CrossRef] [PubMed]
  64. Cavalli, F.M.G.; Hübner, J.M.; Sharma, T.; Luu, B.; Sill, M.; Zapotocky, M.; Mack, S.C.; Witt, H.; Lin, T.; Shih, D.J.H.; et al. Heterogeneity within the PF-B ependymoma subgroup. Acta Neuropathol. 2018, 136, 227–237. [Google Scholar] [CrossRef]
  65. Keenan, C.; Graham, R.T.; Harreld, J.H.; Lucas, J.T., Jr.; Finkelstein, D.; Wheeler, D.; Li, X.; Dalton, J.; Upadhyaya, S.A.; Raimondi, S.C.; et al. Infratentorial C11orf95-fused gliomas share histologic, immunophenotypic, and molecular characteristics of supratentorial RELA-fused ependymoma. Acta Neuropathol. 2020, 140, 963–965. [Google Scholar] [CrossRef]
  66. Abdallah, A.; Emel, E.; Gündüz, H.B.; Sofuoğlu, Ö.E.; Asiltürk, M.; Abdallah, B.G. Long-Term Surgical Resection Outcomes of Pediatric Myxopapillary Ependymoma: Experience of Two Centers and Brief Literature Review. World Neurosurg. 2020, 136, e245–e261. [Google Scholar] [CrossRef]
  67. Bagley, C.A.; Wilson, S.; Kothbauer, K.F.; Bookland, M.J.; Epstein, F.; Jallo, G.I. Long term outcomes following surgical resection of myxopapillary ependymomas. Neurosurg. Rev. 2009, 32, 321–334. [Google Scholar] [CrossRef]
  68. Ahmad, O.; Chapman, R.; Storer, L.C.; Luo, L.; Heath, P.R.; Resar, L.; Cohen, K.J.; Grundy, R.G.; Lourdusamy, A. Integrative molecular characterization of pediatric spinal ependymoma: The UK Children’s Cancer and Leukaemia Group study. Neurooncol. Adv. 2021, 3, vdab043. [Google Scholar]
  69. Ghasemi, D.R.; Sill, M.; Okonechnikov, K.; Korshunov, A.; Yip, S.; Schutz, P.W.; Scheie, D.; Kruse, A.; Harter, P.N.; Kastelan, M.; et al. MYCN amplification drives an aggressive form of spinal ependymoma. Acta Neuropathol. 2019, 138, 1075–1089. [Google Scholar] [CrossRef] [Green Version]
  70. Swanson, A.A.; Raghunathan, A.; Jenkins, R.B.; Messing-Jünger, M.; Pietsch, T.; Clarke, M.J.; Kaufmann, T.J.; Giannini, C. Spinal Cord Ependymomas With MYCN Amplification Show Aggressive Clinical Behavior. J. Neuropathol. Exp. Neurol. 2019, 78, 791–797. [Google Scholar] [CrossRef]
  71. Raffeld, M.; Abdullaev, Z.; Pack, S.D.; Xi, L.; Nagaraj, S.; Briceno, N.; Vera, E.; Pittaluga, S.; Lopes Abath Neto, O.; Quezado, M.; et al. High level MYCN amplification and distinct methylation signature define an aggressive subtype of spinal cord ependymoma. Acta Neuropathol. Commun. 2020, 8, 101. [Google Scholar] [CrossRef]
  72. D’Amico, R.S.; Praver, M.; Zanazzi, G.J.; Englander, Z.K.; Sims, J.S.; Samanamud, J.L.; Ogden, A.T.; McCormick, P.C.; Feldstein, N.A.; McKhann, G.M.; et al. Subependymomas Are Low-Grade Heterogeneous Glial Neoplasms Defined by Subventricular Zone Lineage Markers. World Neurosurg. 2017, 107, 451–463. [Google Scholar] [CrossRef]
  73. Kweh, B.; Rosenfeld, J.V.; Hunn, M.; Tee, J.W. Tumor characteristics and surgical outcomes of intracranial subependymomas: A systematic review and meta-analysis. J. Neurosurg. 2021, 1, 1–13. [Google Scholar] [CrossRef]
  74. Korshunov, A.; Okonechnikov, K.; Schmitt-Hoffner, F.; Ryzhova, M.; Sahm, F.; Stichel, D.; Schrimpf, D.; Reuss, D.E.; Sievers, P.; Suwala, A.K.; et al. Molecular analysis of pediatric CNS-PNET revealed nosologic heterogeneity and potent diagnostic markers for CNS neuroblastoma with FOXR2-activation. Acta Neuropathol. Commun. 2021, 9, 20. [Google Scholar] [CrossRef]
  75. de Sousa, G.R.; Lira, R.C.P.; de Almeida Magalhães, T.; da Silva, K.R.; Nagano, L.F.P.; Saggioro, F.P.; Baroni, M.; Marie, S.K.N.; Oba-Shinjo, S.M.; Brandelise, S.; et al. A coordinated approach for the assessment of molecular subgroups in pediatric ependymomas using low-cost methods. J. Mol. Med. 2021, 99, 1101–1113. [Google Scholar] [CrossRef]
  76. Matsumoto, Y.; Ichikawa, T.; Kurozumi, K.; Otani, Y.; Date, I. Clinicopathological and Genetic Features of Supratentorial Cortical Ependymomas. World Neurosurg. 2019, 129, e417–e428. [Google Scholar] [CrossRef]
  77. Panwalkar, P.; Clark, J.; Ramaswamy, V.; Hawes, D.; Yang, F.; Dunham, C.; Yip, S.; Hukin, J.; Sun, Y.; Schipper, M.J.; et al. Immunohistochemical analysis of H3K27me3 demonstrates global reduction in group-A childhood posterior fossa ependymoma and is a powerful predictor of outcome. Acta Neuropathol. 2017, 134, 705–714. [Google Scholar] [CrossRef]
  78. Antin, C.; Tauziède-Espariat, A.; Debily, M.A.; Castel, D.; Grill, J.; Pagès, M.; Ayrault, O.; Chrétien, F.; Gareton, A.; Andreiuolo, F.; et al. EZHIP is a specific diagnostic biomarker for posterior fossa ependymomas, group PFA and diffuse midline gliomas H3-WT with EZHIP overexpression. Acta Neuropathol. Commun. 2020, 8, 183. [Google Scholar] [CrossRef]
  79. Nambirajan, A.; Sharma, A.; Rajeshwari, M.; Boorgula, M.T.; Doddamani, R.; Garg, A.; Suri, V.; Sarkar, C.; Sharma, M.C. EZH2 inhibitory protein (EZHIP/Cxorf67) expression correlates strongly with H3K27me3 loss in posterior fossa ependymomas and is mutually exclusive with H3K27M mutations. Brain Tumor Pathol. 2021, 38, 30–40. [Google Scholar] [CrossRef]
  80. Łastowska, M.; Matyja, E.; Sobocińska, A.; Wojtaś, B.; Niemira, M.; Szałkowska, A.; Krętowski, A.; Karkucińska-Więckowska, A.; Kaleta, M.; Ejmont, M.; et al. Transcriptional profiling of paediatric ependymomas identifies prognostically significant groups. J. Pathol. Clin. Res. 2021. online ahead of print. [Google Scholar] [CrossRef]
  81. Northcott, P.A.; Shih, D.J.H.; Remke, M.; Cho, Y.J.; Kool, M.; Hawkins, C.; Eberhart, C.G.; Dubuc, A.; Guettouche, T.; Cardentey, Y.; et al. Rapid, reliable, and reproducible molecular sub-grouping of clinical medulloblastoma samples. Acta Neuropathol. 2012, 123, 615–626. [Google Scholar] [CrossRef] [Green Version]
  82. Druy, A.E.; Yasko, L.A.; Konovalov, D.M.; Ektova, A.P.; Valiakhmetova, E.F.; Olshanskaya, Y.V.; Maschan, A.A.; Novichkova, G.A.; Papusha, L.I. Identification of medulloblastoma molecular subgroups by gene expression profiling. Pediatr. Hematol./Oncol. Immunopathol. 2017, 16, 85–89. (In Russian) [Google Scholar] [CrossRef]
  83. Łastowska, M.; Trubicka, J.; Sobocińska, A.; Wojtas, B.; Niemira, M.; Szałkowska, A.; Krętowski, A.; Karkucińska-Więckowska, A.; Kaleta, M.; Ejmontet, M.; et al. Molecular identification of CNS NB-FOXR2, CNS EFT-CIC, CNS HGNET-MN1 and CNS HGNET-BCOR pediatric brain tumors using tumor-specific signature genes. Acta Neuropathol. Commun. 2020, 8, 105. [Google Scholar] [CrossRef]
  84. Gururangan, S.; Fangusaro, J.; Young Poussaint, T.; Onar-Thomas, A.; Gilbertson, R.J.; Vajapeyam, S.; Gajjar, A.; Goldman, S.; Friedman, H.S.; Packer, R.J.; et al. Lack of efficacy of bevacizumab + irinotecan in cases of pediatric recurrent ependymoma--a Pediatric Brain Tumor Consortium study. Neuro Oncol. 2012, 14, 1404–1412. [Google Scholar] [CrossRef] [Green Version]
  85. DeWire, M.; Fouladi, M.; Turner, D.C.; Wetmore, C.; Hawkins, C.; Jacobs, C.; Yuan, Y.; Liu, D.; Goldman, S.; Fisher, P.; et al. An open-label, two-stage, phase II study of bevacizumab and lapatinib in children with recurrent or refractory ependymoma: A collaborative ependymoma research network study (CERN). J. Neurooncol. 2015, 123, 85–91. [Google Scholar] [CrossRef]
  86. Wetmore, C.; Daryani, V.M.; Billups, C.A.; Boyett, J.M.; Leary, S.; Tanos, R.; Goldsmith, K.C.; Stewart, C.F.; Blaney, S.M.; Gajjar, A. Phase II evaluation of sunitinib in the treatment of recurrent or refractory high-grade glioma or ependymoma in children: A children’s Oncology Group Study ACNS1021. Cancer Med. 2016, 5, 1416–1424. [Google Scholar] [CrossRef]
  87. Fouladi, M.; Stewart, C.F.; Blaney, S.M.; Onar-Thomas, A.; Schaiquevich, P.; Packer, R.J.; Goldman, S.; Geyer, J.R.; Gajjar, A.; Kun, L.E.; et al. A molecular biology and phase II trial of lapatinib in children with refractory CNS malignancies: A pediatric brain tumor consortium study. J. Neurooncol. 2013, 114, 173–179. [Google Scholar] [CrossRef] [Green Version]
  88. Milde, T.; Kleber, S.; Korshunov, A.; Witt, H.; Hielscher, T.; Koch, P.; Kopp, H.G.; Jugold, M.; Deubzer, H.E.; Oehme, I.; et al. A novel human high-risk ependymoma stem cell model reveals the differentiation-inducing potential of the histone deacetylase inhibitor Vorinostat. Acta Neuropathol. 2011, 122, 637–650. [Google Scholar] [CrossRef] [Green Version]
  89. Rahman, R.; Osteso-Ibanez, T.; Hirst, R.A.; Levesley, J.; Kilday, J.P.; Quinn, S.; Peet, A.; O’Callaghan, C.; Coyle, B.; Grundy, R.G. Histone deacetylase inhibition attenuates cell growth with associated telomerase inhibition in high-grade childhood brain tumor cells. Mol. Cancer Ther. 2010, 9, 2568–2581. [Google Scholar] [CrossRef] [Green Version]
  90. Bukowinski, A.; Chang, B.; Reid, J.M.; Liu, X.; Minard, C.G.; Trepel, J.B.; Lee, M.J.; Fox, E.; Weigel, B.J. A phase 1 study of entinostat in children and adolescents with recurrent or refractory solid tumors, including CNS tumors: Trial ADVL1513, Pediatric Early Phase-Clinical Trial Network (PEP-CTN). Pediatr. Blood Cancer 2021, 68, e28892. [Google Scholar] [CrossRef]
  91. Antonelli, R.; Jiménez, C.; Riley, M.; Servidei, T.; Riccardi, R.; Soriano, A.; Roma, J.; Martínez-Saez, E.; Martini, M.; Ruggiero, A.; et al. CN133, a Novel Brain-Penetrating Histone Deacetylase Inhibitor, Hampers Tumor Growth in Patient-Derived Pediatric Posterior Fossa Ependymoma Models. Cancers 2020, 12, 1922. [Google Scholar] [CrossRef] [PubMed]
  92. Servidei, T.; Meco, D.; Martini, M.; Battaglia, A.; Granitto, A.; Buzzonetti, A.; Babini, G.; Massimi, L.; Tamburrini, G.; Scambia, G.; et al. The BET Inhibitor OTX015 Exhibits In Vitro and In Vivo Antitumor Activity in Pediatric Ependymoma Stem Cell Models. Int. J. Mol. Sci. 2021, 22, 1877. [Google Scholar] [CrossRef] [PubMed]
  93. Witt, D.A.; Donson, A.M.; Amani, V.; Moreira, D.C.; Sanford, B.; Hoffman, L.M.; Handler, M.H.; Levy, J.; Jones, K.L.; Nellan, A.; et al. Specific expression of PD-L1 in RELA-fusion supratentorial ependymoma: Implications for PD-1-targeted therapy. Pediatr. Blood Cancer 2018, 65, e26960. [Google Scholar] [CrossRef] [PubMed]
  94. Nambirajan, A.; Malgulwar, P.B.; Sharma, A.; Boorgula, M.T.; Doddamani, R.; Singh, M.; Suri, V.; Sarkar, C.; Sharma, M.C. Clinicopathological evaluation of PD-L1 expression and cytotoxic T-lymphocyte infiltrates across intracranial molecular subgroups of ependymomas: Are these tumors potential candidates for immune check-point blockade? Brain Tumor Pathol. 2019, 36, 152–161. [Google Scholar] [CrossRef]
  95. Perruccio, K.; Mastronuzzi, A.; Lupattelli, M.; Arcioni, F.; Capolsini, I.; Cerri, C.; Gurdo, G.M.I.; Massei, M.S.; Mastrodicasa, E.; Caniglia, M. Targeted Therapy with Sirolimus and Nivolumab in a Child with Refractory Multifocal Anaplastic Ependymoma. Reports 2021, 4, 12. [Google Scholar] [CrossRef]
Figure 1. Basic classification of intracranial ependymal tumors.
Figure 1. Basic classification of intracranial ependymal tumors.
Cancers 13 04954 g001
Figure 2. The integrated approach for molecular group determination applied to ependymal tumors.
Figure 2. The integrated approach for molecular group determination applied to ependymal tumors.
Cancers 13 04954 g002
Table 1. Summary of major molecular markers of ependymal tumors.
Table 1. Summary of major molecular markers of ependymal tumors.
LocalizationMolecular GroupMajor Molecular MarkersPrevalence within the GroupPathogenic Impact
SupratentorialST-EPN-ZFTAZFTARELA fusion,
chromothripsis 11q13.1
90–95%NF-kB pathway activation
ZFTAMAML2
ZFTANCOA1
ZFTANCOA2
5–10%EP300/CREBBP gene expression pathway
ST-EPN-YAP1YAP1MAMLD195%Hippo pathway activation
YAP1FAM118B5%
InfratentorialPF-EPN-AEZHIP overexpression95%CpG-island methylator phenotype
HIST1H3C, HIST1H3B or H3F3A K27M substitution<5%
PF-EPN-BMajor cytogenetic aberrations Up to 100%Ciliogenesis deregulation
SpinalSp-MPEHOXB cluster genes amplificationUp to 100%Mitochondrial metabolism pathways activation
SP-EPN-MYCNMYCN amplification100%Proliferative signaling
Table 2. Subgroup-specific diagnostic marker and candidate genes involved in recurrent chromosomal abnormalities in pediatric EPNs.
Table 2. Subgroup-specific diagnostic marker and candidate genes involved in recurrent chromosomal abnormalities in pediatric EPNs.
Molecular GroupImplicated Gene *Gene NameCytogenetic BandPathogenic ImpactEvidence Level **Evidence-Based Categorization ***Hallmark of Cancer ****
PromotesSuppresses
ST-EPN-ZFTAZFTAZinc finger translocation associated11q13.15′-partner gene in ZFTARELA fusionITier I,
level A
Genome instability
RELAV-Rel avian reticuloendotheliosis viral oncogene homolog A11q13.13′-partner gene in ZFTARELA fusionITier I,
level A
Escaping programmed cell death; tumor promoting inflammation
MAML2Mastermind-like transcriptional coactivator 211q213′-partner gene in ZFT–MAML2 fusionIIITier II,
level C
Proliferative signaling; angiogenesis
NCOA1Nuclear receptor coactivator 12p23.33′-partner gene in ZFTA–NCOA1 fusionIIITier II,
level C
Proliferative signaling; change of cellular energetics
NCOA2Nuclear receptor coactivator 28q13.33′-partner gene in ZFTA–NCOA2 fusionIIITier II,
level C
Proliferative signaling; change of cellular energetics; escaping programmed cell death
ST-EPN-YAP1YAP1Yes1-associated transcriptional regulator11q22.15′-partner gene in YAP1–MAMLD1 fusionIITier I,
level A
Proliferative signaling; escaping programmed cell death; invasion and metastasisEscaping programmed cell death
MAMLD1Mastermind-like domain-containing 1Xq283′-partner gene in YAP1–MAMLD1 fusionIITier I,
level A
Proliferative signaling; angiogenesisEscaping programmed cell death
FAM118BFamily with sequence similarity 118 member B11q24.23′-partner gene in YAP1–FAM118B fusionIVTier II,
level D
Unknown
Non-ZFTA/Non-YAP1 ST-EPNsPLAGL1PLAG1-like zinc finger 16q24.23′-partner gene in EWSR1-PLAGL1 fusion; 5′-partner gene in PLAGL1–FOXO1 or PLAGL1–EP300 fusionIVTier II,
level D
Suppression of growthEscaping immunic response to cancer; tumor promoting inflammation; invasion and metastasis; angiogenesis
EWSR1EWS RNA binding protein 122q12.25′-partner gene in EWSR1–PLAGL1 or EWSR1–PATZ1 fusionIVTier II,
level D
Proliferative signaling; escaping programmed cell death; angiogenesis; invasion and metastasisGenome instability and mutations
FOXO1Forkhead box O113q14.113′-partner gene in PLAGL1–FOXO1 fusionIVTier II,
level D
Change of cellular energeticsEscaping programmed cell death
EP300E1A binding protein P30022q13.23′-partner gene in PLAGL1–EP300 fusionIVTier II,
level D
Suppression of growthEscaping programmed cell death
PATZ1POZ/BTB and AT hook-containing zinc finger 122q12.23′-partner gene in EWSR1–PATZ1 or MN1–PATZ1 fusionIVTier II,
level D
Proliferative signaling; escaping programmed cell death
MN1MN1 proto-oncogene, transcriptional regulator22q12.15′-partner gene in MN1-PATZ1 fusionIVTier II,
level D
Suppression of growthEscaping programmed cell death
PF-EPN-AEZHIPEZH inhibitory proteinXp11.22OverexpressionIVTier II,
level D
EZH1/EZH2-mediated trimethylation of H3K27
EPOPElongin BC and polycomb repressive complex 2-associated protein17q12OverexpressionIVTier II,
level D
EZH2-mediated trimethylation of H3K27
HIST1H3CH3 clustered histone 36p22.2Somatic mutationIVTier II,
level D
EZH2-mediated trimethylation of H3K27
HIST1H3BH3 clustered histone 26p22.2Somatic mutationIVTier II,
level D
EZH2-mediated trimethylation of H3K27
H3F3AH3.3 histone A1q42.12Somatic mutationIVTier II,
level D
EZH2-mediated trimethylation of H3K27
BCL9BCL9 transcription coactivator1q21.2Oncogene, involved in 1q gainVNAProliferative signaling; invasion and metastasis; angiogenesis
ARNTAryl hydrocarbon receptor nuclear translocator1q21.3Oncogene, involved in 1q gainVNAAngiogenesis; change of cellular energeticsInvasion and metastasis
SETDB1SET domain bifurcated histone lysine methyltransferase 11q21.3Oncogene, involved in 1q gainVNAEpigenetic transcriptional repression by recruiting HP1 (CBX1, CBX3 and/or CBX5) proteins to methylated histones
NTRK1Neurotrophic receptor tyrosine kinase 11q23.1Oncogene, involved in 1q gainVNAProliferative signaling; escaping programmed cell death; angiogenesis
FCRL4Fc receptor-like 41q23.1Oncogene, involved in 1q gainVNAEscaping immunic response to cancer
FCGR2BFc fragment of IgG receptor IIb1q23.3Oncogene, involved in 1q gainVNASuppression of growthEscaping programmed cell death
DDR2Discoidin domain receptor tyrosine kinase 21q23.3Oncogene, involved in 1q gainVNAInvasion and metastasis
PBX1PBX homeobox 11q23.3Oncogene, involved in 1q gainVNAAngiogenesis; escaping programmed cell death; change of cellular energetics
ABL2ABL proto-oncogene 2, non-receptor tyrosine kinase1q25.2Oncogene, involved in 1q gainVNAProliferative signaling; invasion and metastasis; angiogenesis; genome instability and mutations; change of cellular energeticsEscaping programmed cell death
MDM4MDM4 regulator of P531q32.1Oncogene, involved in 1q gainVNAProliferative signaling; invasion and metastasis; angiogenesis; escaping programmed cell deathSuppression of growth
ELK4ETS transcription factor ELK41q32.1Oncogene, involved in 1q gainVNAProliferative signaling; escaping programmed cell death
RGS7Regulator of G protein signaling 71q43Oncogene, involved in 1q gainVNAChange of cellular energetics
AKT3AKT serine/threonine Kinase 31q43-q44Oncogene, involved in 1q gainVNAProliferative signaling; suppression of growth; invasion and metastasis; angiogenesis; escaping programmed cell death; change of cellular energeticsInvasion and metastasis; angiogenesis; genome instability and mutations
EPHA7EPH receptor A76q16.1Tumor suppressor gene, involved in 6q lossVNA Escaping programmed cell death
CCNCCyclin C6q16.2Tumor suppressor gene, involved in 6q lossVNA Proliferative signaling
PRDM1PR/SET domain 16q21Tumor suppressor gene, involved in 6q lossVNASuppression of growthEscaping immunic response to cancer
FOXO3Forkhead box O36q21Tumor suppressor gene, involved in 6q lossVNAChange of cellular energeticsEscaping programmed cell death
PTPRKProtein tyrosine phosphatase receptor type K6q22.33Tumor suppressor gene, involved in 6q lossVNAEscaping immunic response to cancerProliferative signaling
BCLAF1BCL2-associated transcription factor 16q23.3Tumor suppressor gene, involved in 6q lossVNA Escaping programmed cell death
TNFAIP3TNF alpha-induced protein 36q23.3Tumor suppressor gene, involved in 6q lossVNA Escaping immunic response to cancer; tumor promoting inflammation
LATS1Large tumor suppressor kinase 16q25.1Tumor suppressor gene, involved in 6q lossVNASuppression of growthGenome instability and mutations; escaping programmed cell death
ESR1Estrogen receptor 16q25.1Tumor suppressor gene, involved in 6q lossVNAProliferative signaling; suppression of growth; escaping immunic response to cancer; invasion and metastasis Invasion and metastasis
ARID1BAT-rich interaction domain 1B6q25.3Tumor suppressor gene, involved in 6q lossVNASuppression of growth; cell replicative immortalityCell replicative immortality; invasion and metastasis; genome instability and mutations; escaping programmed cell death
QKIQKI, KH domain-containing RNA binding6q26Tumor suppressor gene, involved in 6q lossVNASuppression of growth; escaping programmed cell deathEscaping programmed cell death
PF-EPN-BLATS2Large tumor suppressor kinase 213q12.11Tumor suppressor gene, involved in 13 q lossVNASuppression of growth; invasion and metastasisInvasion and metastasis; genome instability and mutations; escaping programmed cell death
CDX2Caudal type homeobox 213q12.2Tumor suppressor gene, involved in 13 q lossVNA Proliferative signaling
BRCA2BRCA2 DNA repair associated13q13.1Tumor suppressor gene, involved in 13 q lossVNA Genome instability and mutations; escaping programmed cell death
RB1RB transcriptional corepressor 113q14.2Tumor suppressor gene, involved in 13 q lossVNASuppression of growth; escaping programmed cell death; change of cellular energeticsEscaping immunic response to cancer; invasion and metastasis; genome instability and mutations; escaping programmed cell death
GPC5Glypican 513q31.3Tumor suppressor gene, involved in 13 q lossVNASuppression of growth; invasion and metastasis
SOX21SRY-box transcription factor 2113q32.1Tumor suppressor gene, involved in 13 q lossVNASuppression of growthProliferative signaling
ERCC5ERCC excision repair 5, endonuclease13q33.1Tumor suppressor gene, involved in 13 q lossVNAGenome instability and mutations; escaping programmed cell deathGenome instability and mutations
SP-MPEHOXB13Homeobox B1317q21.32AmplificationIIITier II,
level C
Change of cellular energeticsEscaping programmed cell death
SP-EPN-MYCNMYCNMYCN proto-oncogene, BHLH transcription factor2p24.3AmplificationIITier I,
level A
Proliferative signaling; escaping immunic response to cancer; angiogenesis; genome instability and mutations; change of cellular energeticsCell replicative immortality; invasion and metastasis; escaping programmed cell death
* The list of genes is selected from the Catalogue of Somatic Mutations in Cancer (COSMIC) Cancer Gene Census (https://cancer.sanger.ac.uk/census, accessed on 20 September 2021). Oncogenes and tumor suppressor genes are viewed as candidates for recurrent aberrations resulting in gain-of-function (1q gains) and loss-of-function (6q losses, 13q losses), respectively. ** Strength of evidence for gene diagnostic value based on Strength-of-evidence rating scheme of the Centre for Evidence-Based Medicine (https://www.cebm.net, accessed on 20 September 2021). *** Evidence-based variant (nucleotide substitution, copy-number variation, and fusion) of listed genes categorization based on the Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists (AMP/ASCO/CAP recommendations). **** Potential roles of the cancer hallmark genes are annotated using COSMIC Cancer Gene Census (https://cancer.sanger.ac.uk/census, accessed on 20 September 2021), GeneCards: The Human Gene Database (https://www.genecards.org/, accessed on 20 September 2021), and KEGG: Kyoto Encyclopedia of Genes and Genomes (https://www.kegg.jp/kegg/, accessed on 20 September 2021). NA—nonapplicable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zaytseva, M.; Papusha, L.; Novichkova, G.; Druy, A. Molecular Stratification of Childhood Ependymomas as a Basis for Personalized Diagnostics and Treatment. Cancers 2021, 13, 4954. https://doi.org/10.3390/cancers13194954

AMA Style

Zaytseva M, Papusha L, Novichkova G, Druy A. Molecular Stratification of Childhood Ependymomas as a Basis for Personalized Diagnostics and Treatment. Cancers. 2021; 13(19):4954. https://doi.org/10.3390/cancers13194954

Chicago/Turabian Style

Zaytseva, Margarita, Ludmila Papusha, Galina Novichkova, and Alexander Druy. 2021. "Molecular Stratification of Childhood Ependymomas as a Basis for Personalized Diagnostics and Treatment" Cancers 13, no. 19: 4954. https://doi.org/10.3390/cancers13194954

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop