1. Introduction
Atypical teratoid rhabdoid tumor (ATRT) is a highly malignant central nervous system (CNS) neoplasm of early childhood [1]. Approximately 70% of ATRTs are diagnosed in the first year of life, and over 90% of cases occur in children younger than three years of age. Although ATRT accounts for only 1–2% of all pediatric CNS tumors, it comprises 40–50% of all CNS malignancies in children younger than one year of age. ATRT is slightly more common in males, with a male-to-female ratio of approximately 1.5, and is most often diagnosed in Caucasians. It may arise anywhere in the CNS and should be considered as a diagnosis when evaluating any aggressive-appearing tumor of the brain or spinal cord in a very young pediatric patient. Metastatic dissemination at diagnosis is common and may be found in 20–40% of cases [2–6].
ATRT is characterized by a primitive histologic appearance with multi-lineage differentiation, including varying components of glial, mesenchymal, and epithelial differentiation [1]. Histologically, ATRT is an embryonal tumor that displays loss of nuclear SMARCB1 or SMARCA4 expression or, for unresolved lesions, a DNA methylation profile that aligns with ATRT [1,7]. Seminal molecular studies over the past 30 years have revolutionized our understanding of the molecular basis of ATRT, facilitating a transition from a descriptive, morphology-based assessment toward a molecular (epi) genetic understanding that has revealed potential targeted treatments for this lethal childhood cancer [8–10]. Here, we focus our discussions on disease mechanisms of ATRT and how its unique molecular changes may be exploited using novel therapeutics, ultimately offering hope for patients with ATRT and their families.
2. Basic biology
Pioneering research of rhabdoid tumor predisposition syndrome (RTPS) in the late 1990s and early 2000s identified the loss of part or all of the long arm of chromosome 22 as a recurrent molecular event in rhabdoid tumors including ATRT [11–13]. Subsequent molecular analyses demonstrated biallelic SMARCB1 loss-of-function alterations as the key genetic event in ATRT pathogenesis [14–16]. In its role as a core subunit of the canonical SWItch/sucrose non-fermentable (SWI/SNF) chromatin-remodeling complex (also known as the canonical BAF [cBAF] complex), SMARCB1 is required for cell lineage determination and normal cell differentiation [17–19]. Functional deficiency of SMARCA4, a shared BAF ATPase, in the few SMARCB1 wild-type cases further underscores the importance of BAF in ATRT tumorigenesis [8,20]. Despite their highly malignant and heterogeneous appearance, the genomes of ATRTs are remarkably stable and contain limited exomic alterations [15,21,22].
BAF complexes destabilize histone-DNA interactions in an ATP-dependent manner, opening neighboring stretches of DNA and allowing transcription factors and chromatin-remodeling enzymes to either activate or repress target genes [23]. Loss of BAF has profound effects on cell viability, proliferation, and differentiation depending on cell context and the exact BAF perturbation. Pertinent to ATRT, SMARCB1 loss results in histone subunit 3 lysine 27 acetylation (H3K27Ac) loss at promoter-distal enhancers necessary for cell-lineage specification and differentiation [18,24]. Conversely, promoter-proximal, cell lineage-specific super-enhancers associated with non-canonical BAF (ncBAF) are retained in ATRT and are essential for cell survival [19,25,26]. Deranged polycomb-related signaling as a result of SMARCB1 deficiency has been identified as a key tumorigenic driver in ATRT by multiple studies [21,27,28]. Abundant research further implicates SMARCB1 in the regulation of numerous other pathways of cancer and development, including RhoA-dependent regulation of cell migration [29,30], cell cycle regulation via Aurora Kinase A [31], P16INK4a [32], TP53 [33,34], and RB [35,36], and downstream activation of other oncogenic pathways such as SHH [37], WNT/β-catenin [38,39], and c-MYC [40–42] (Figure 1).
Figure 1.

Basic BAF complex composition and epigenetic function. The canonical SWItch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complex (canonical BAF [cBAF] complex) is a multi-subunit modulator of gene transcription implicated in vital cellular functions such as cell differentiation, lineage determination, and self-renewal. In the absence of a functional SMARCB1 subunit, less BAF complex is recruited to typical enhancers, which is accompanied by loss of histone acetylase activity and deranged polycomb-signaling at these sites of transcriptional regulation. Super enhancer activity is selectively retained at genes that contribute to immortality and cell proliferation in the absence a functional SMARCB1 subunit (created with BioRender.com).
In contrast to the chromosomal stability of ATRT, recent comprehensive molecular studies have demonstrated that SMARCB1-deficient ATRTs are comprised of at least three molecularly distinct, clinically relevant subtypes with shared and subtype-specific epigenomic features, while SMARCA4-deficient ATRT constitutes a fourth subgroup that does not harbor SMARCB1 alterations [20,21,43,44]. While multiple naming paradigms have been proposed, recent consensus defines the three SMARCB1-deficient subgroups as ATRT-SHH, ATRT-TYR, and ATRT-MYC [3]. ATRT-SHH tumors tend to occur in toddlers predominantly in the supratentorial compartment. They contain focal SMARCB1 alterations, have a hypermethylated phenotype, and are defined by a primitive neuronal gene expression pattern and NOTCH pathway activation [21,43,44]. ATRT-TYR tumors usually occur in the posterior fossa of infants and tend to have broad deletions affecting chr22q11.2, are hypermethylated, and display a mesenchymal gene expression profile characterized by deranged bone morphogenic protein (BMP) and melanogenesis signaling pathways, including micropthalmia-associated transcription factor (MITF) and tyrosinase (TYR) [21,43,44]. ATRT-MYC tumors are more common in older children and may occur in either the supra- or infratentorial compartment or the spinal cord. These tumors usually contain focal mutations or deletions in SMARCB1, but they display a hypomethylated phenotype, and are marked by dysregulated MYC signaling and derepression of HOX family genes [21,43,44]. SMARCA4-mutant ATRT occurs in young infants and is associated with a higher frequency of germline mutations and a distinct DNA methylation signature in comparison to SMARCB1 mutated tumors [20,45].
Clinically, prognosis appears to differ by molecular subgroup, but this is heavily influenced by other clinical factors that are not evenly distributed among molecular subgroups, namely patient age and the presence of metastatic disease at diagnosis. Two separate studies have demonstrated TYR-subtype tumors have an improved overall prognosis, either when analyzed independently or compared to all non-TYR-subtype ATRT. Frühwald et al. identified age less than one year as an independent risk factor for overall survival in addition to non-TYR subgrouping [46]. Upadhyaya et al. showed that ATRT-SHH is most commonly associated with metastatic disease at diagnosis, leading to inferior outcomes compared to ATRT-TYR and ATRT-MYC. In this study, given that patients with ATRT-TYR are often the youngest, infants without metastatic disease were analyzed as a separate subgroup, and in this analysis, outcomes did not differ by molecular subtype [47]. These observations demonstrate the complex interplay between clinicopathologic factors and molecular subtype in ATRT and highlight the need for further investigation into the unique role, if any, of molecular subgrouping in prognosis.
3. State of the art treatment
The mainstay of treatment in ATRT is an aggressive multi-modal approach consisting of maximal safe surgical resection with subsequent adjuvant chemotherapy and radiotherapy. Advances in each of these modalities have improved four-year overall survival to just under 50%, on average, leaving substantial room for improvement in future therapeutic avenues [5,48–50]. Most referral centers in the USA would use the Children’s Oncology Group (COG) ACNS0333 protocol for newly diagnosed ATRT, which utilizes two courses of multiagent chemotherapy followed by three courses of high-dose chemotherapy (HDCT) followed by peripheral blood stem cell rescue and craniospinal irradiation (CSI) after maximal safe surgical resection [6]. ACNS0333 resulted in a significant survival benefit over a historical cohort, but both event-free and overall survival remained below 50% at four years, and survivors suffered typical chemo- and radiotherapy-associated adverse effects. Similar outcomes have been achieved in some smaller studies using conventional and intrathecal chemotherapy in combination with radiotherapy, calling into question the need for HDCT in selected patients [48]. Given the associations of older age and ATRT-TYR subgrouping with improved prognosis, the risk-benefit ratio of HDCT in these patients must be carefully weighed, given the potential for similar therapeutic benefit using regimens that spare ablative chemotherapy. While radiotherapy remains a mainstay of contemporary treatment for ATRT and has generally been associated with improved tumor control [4,46], the known association with poor neurodevelopmental outcomes in very young patients demands a careful consideration of treatment risk in this modality compared to HDCT [5]. To assess the non-inferiority of HDCT compared to conventional chemotherapy and focal radiotherapy, the European Society for Pediatric Oncology recently initiated a randomized phase-III clinical trial for children with ATRT (EudraCT 2018–003335-29), which also includes the comparison of neurocognitive outcomes among treatment arms as a secondary study objective.
Many patients with ATRT who survive the early stage of the disease will remain refractory to conventional chemo- and radiotherapy, and for these patients, novel targeted treatments are on the horizon that may offer promise as a therapeutic option. Current work is aimed at defining both highly efficacious as well as molecular subtype-specific therapies that target underlying molecular drivers of tumorigenesis (Figure 2). Histone deacetylase inhibitors (HDACis) have demonstrated efficacy across molecular subgroups of ATRT. HDACis such as suberoylanilide hydroxamic acid (SAHA, also known as vorinostat) and LAQ824 were shown to be effective against multiple ATRT cell lines in vitro [44,51], and vorinostat demonstrated a radiosensitizing effect in a rhabdoid tumor xenograft model [52]. Recruitment for two early phase clinical trials investigating vorinostat in a variety of cancers including ATRT has been completed with myelosuppression being the dose-limiting toxicity (NCT00217412, NCT01076530 [53]). As described above, SMARCB1 loss leads to cell cycle progression, and both the Aurora Kinase A inhibitor alisertib and the CDK4/6 inhibitor ribociclib have been and are currently being used in clinical studies of children with ATRT (NCT02114229, NCT03387020, NCT03434262) [54,55].
Figure 2.

Summary of potential therapeutic strategies. Groundbreaking findings in the fields of BAF and enhancer biology have revealed a central role for epigenetic dysregulation in the pathogenesis of ATRT. A variety of different therapies targeting secondary epigenetic vulnerabilities are currently being investigated in preclinical studies and clinical trials. Among others, these pathophysiologically relevant and potentially subgroup-specific therapeutic approaches include inhibition of AURK, CDK4/6, MYC, and polycomb, which may be therapeutically targeted by alisertib, ribociclib, dasatinib, tazemetostat, respectively. An alternative strategy is to target residual BAF, and a few select compounds (e.g. FHD-609 and FHD-286) are likewise in clinical trial for cancers that contain a mutation in a BAF complex subunit (created with BioRender.com).
To date, only a few, limited-scale drug screens have been performed for ATRT. Torchia et al. screened a variety of small-molecule inhibitors targeting multiple molecular subtype-specific pathways on cell lines that transcriptionally correspond to ATRT-SHH and ATRT-MYC subtypes [44]. In this study, ATRT-SHH cell lines were more critically dependent on epigenetic regulators involved in chromatin remodeling with EZH2 and bromodomain inhibition being selectively toxic to ATRT-SHH cell lines. Tumors of ATRT-TYR and ATRT-MYC subgroups (formerly ‘Group 2’ subtypes) were more critically dependent on PDGFR and other receptor tyrosine kinase pathways. The multi-tyrosine kinase inhibitors dasatinib and nilotinib were selectively toxic to group-2 cells in vitro, and in an orthotopic xenograft model of a group-2 cell line, dasatinib prolonged survival [44]. Further cementing the importance of RTK signaling in ATRT-MYC, a CRISPR screen of extracranial rhabdoid tumors, which are most similar to ATRT-MYC by DNA methylation, similarly demonstrated RTK dependence [56]. A phase I/II study of dasatinib in combination with cytotoxic chemotherapy for CNS tumors is ongoing (NCT00788125). Taken together, results from laboratory research and early clinical studies underscore the therapeutic potential of ATRT-targeted therapies and suggest selective vulnerabilities in each molecular subtype.
4. Expert opinion
The benefit of aggressive multi-modal cytoreductive and cytotoxic therapy for ATRT has cemented the role of surgical resection, craniospinal radiotherapy, and ablative chemotherapy in the upfront treatment of ATRT. Nevertheless, the numerous developmental adverse effects of this treatment and high relapse rate underscore the importance of identifying highly efficacious, targeted therapies for this disease.
ATRT is fundamentally a disease of selective enhancer loss, so it stands to reason that targeting secondary epigenetic vulnerabilities is a pathophysiologically relevant and potentially tumor-specific therapeutic approach. However, the road to targeted epigenetic therapy in ATRT is not straightforward and is limited not only by our understanding of rhabdoid tumor biology but also by available therapeutic options.
Polycomb inhibition using the clinically available EZH2 inhibitor tazemetostat is one approach that has gained traction in the pediatric oncology community. EZH2 inhibition is attractive not only due to its targetability but its biological relevance: in developmental systems, BAF has been shown to antagonize polycomb-mediated gene repression [57], and EZH2 inhibition using RNA interference or pharmacologic inhibition has been shown to variably inhibit ATRT cell growth in vitro and in vivo [28,58]. In a phase I clinical trial or relapsed or refractory SMARCB1-deficient tumors, tazemetostat treatment resulted in a clinically measurable response in 19% of patients with ATRT [59]. Final results of clinical trials of tazemetostat in ATRT and other tumors (NCT02601937, NCT03213665) are necessary in determining the long-term efficacy of this treatment.
An alternative approach to targeting epigenetic vulnerabilities in ATRT is to inhibit residual, ncBAF-dependent enhancer activity that is thought to contribute to tumorigenesis. The oncogenic role of the ncBAF-specific subunit BRD9 in a range of malignancies and its demonstrated role in maintaining potentially proto-oncogenic, promoter-proximal enhancers in ATRT makes this protein an appealing therapeutic target [25,26]. A small molecule inhibitor of BRD9 (FHD-609) is now in clinical trial for SS18-SSX fusion-containing synovial sarcoma (NCT NCT04965753), in which SS18-SSX fusions preclude cBAF assembly. An inhibitor of the BAF ATPase subunits SMARCA2 and SMARCA4 (FHD-286) is likewise in clinical trial for uveal melanoma (NCT04879017) and multiple hematologic malignancies (NCT04891757). The results of these trials will be imperative in determining whether targeting BAF is a viable therapeutic strategy in the wide range of cancers that contain a mutation in a BAF complex subunit.
In sum, a wealth of research over the past decade has revealed a central role for epigenetic dysregulation in the pathogenesis of ATRT. Clinically, ablative chemotherapy, craniospinal radiotherapy, and aggressive surgical resection are the mainstay of upfront treatment of ATRT, which has substantially improved outcomes compared to historical cohorts. Moving forward, the next generation of therapeutic approaches should implement recent groundbreaking findings in the fields of BAF and enhancer biology into clinically efficacious treatments for early refractory and recurrent disease.
Funding
This paper was not funded.
Declaration of interest
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
Footnotes
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 1.Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ostrom QT, Chen Y, de Blank P, et al. The descriptive epidemiology of atypical teratoid/rhabdoid tumors in the United States, 2001–2010. Neuro Oncol. 2014;16(10):1392–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.•.Ho B, Johann PD, Grabovska Y, et al. Molecular subgrouping of atypical teratoid/rhabdoid tumors-a reinvestigation and current consensus. Neuro Oncol. 2020;22(5):613–624. [DOI] [PMC free article] [PubMed] [Google Scholar]; A 2020 consensus statement and integrated analysis of all available DNA methylation and gene expression data. Differences in nomenclature between study groups and minor discrepancies in key findings were resolved.
- 4.Fossey M, Li H, Afzal S, et al. Atypical teratoid rhabdoid tumor in the first year of life: the Canadian ATRT registry experience and review of the literature. J Neurooncol. 2017;132(1):155–162. [DOI] [PubMed] [Google Scholar]
- 5.Lafay-Cousin L, Hawkins C, Carret AS, et al. Central nervous system atypical teratoid rhabdoid tumours: the Canadian Paediatric Brain Tumour Consortium experience. Eur J Cancer. 2012;48(3):353–359. [DOI] [PubMed] [Google Scholar]
- 6.••.Reddy AT, Strother DR, Judkins AR, et al. Efficacy of high-dose chemotherapy and three-dimensional conformal radiation for atypical teratoid/rhabdoid tumor: a report from the Children’s Oncology Group Trial ACNS0333. J Clin Oncol. 2020;38 (11):1175–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]; Recent study demonstrating survival benefit of aggressive multi-modal treatment of ATRT using the ACNS033 protocol over a historical cohort.
- 7.Ellison D, Love S, and Chimelli LMC, et al. Neuropathology E-book: a reference text of CNS pathology. London, United Kingdom: Elsevier Health Sciences; 2012. [Google Scholar]
- 8.Hasselblatt M, Gesk S, Oyen F, et al. Nonsense mutation and inactivation of SMARCA4 (BRG1) in an atypical teratoid/rhabdoid tumor showing retained SMARCB1 (INI1) expression. Am J Surg Pathol. 2011;35(6):933–935. [DOI] [PubMed] [Google Scholar]
- 9.Bookhout C, Bouldin TW, Ellison DW. Atypical teratoid/rhabdoid tumor with retained INI1 (SMARCB1) expression and loss of BRG1 (SMARCA4). Neuropathology. 2018;38(3):305–308. [DOI] [PubMed] [Google Scholar]
- 10.Nemes K, Frühwald MC. Emerging therapeutic targets for the treatment of malignant rhabdoid tumors. Expert Opin Ther Targets. 2018;22(4):365–379. [DOI] [PubMed] [Google Scholar]
- 11.Sévenet N, Sheridan E, Amram D, et al. Constitutional mutations of the hSNF5/INI1 gene predispose to a variety of cancers. Am J Hum Genet. 1999;65(5):1342–1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Biegel JA, Tan L, Zhang F, et al. Alterations of the hSNF5/INI1 gene in central nervous system atypical teratoid/rhabdoid tumors and renal and extrarenal rhabdoid tumors. Clin Cancer Res. 2002;8 (11):3461–3467. [PubMed] [Google Scholar]
- 13.Sredni ST, Tomita T. Rhabdoid tumor predisposition syndrome. Pediatr Dev Pathol. 2015;18(1):49–58. [DOI] [PubMed] [Google Scholar]
- 14.Versteege I, Sévenet N, Lange J, et al. Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature. 1998;394 (6689):203–206. [DOI] [PubMed] [Google Scholar]
- 15.Lee RS, Stewart C, Carter SL, et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest. 2012;122(8):2983–2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kieran MW, Roberts CW, Chi SN, et al. Absence of oncogenic canonical pathway mutations in aggressive pediatric rhabdoid tumors. Pediatr Blood Cancer. 2012;59(7):1155–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kadoch C, Hargreaves DC, Hodges C, et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat Genet. 2013;45(6):592–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.••.Nakayama RT, Pulice JL, Valencia AM, et al. SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters. Nat Genet. 2017;49(11):1613–1623. [DOI] [PMC free article] [PubMed] [Google Scholar]; One of multiple important studies demonstrating a key role for the canonical BAF subunit SMARCB1 in defining promoter-distal, lineage-specific enhancers.
- 19.Wang X, Lee RS, Alver BH, et al. SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet. 2017;49(2):289–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Holdhof D, Johann PD, Spohn M, et al. Atypical teratoid/rhabdoid tumors (ATRTs) with SMARCA4 mutation are molecularly distinct from SMARCB1-deficient cases. Acta Neuropathol. 2021;141 (2):291–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.••.Johann PD, Erkek S, Zapatka M, et al. Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell. 2016;29(3):379–393. [DOI] [PubMed] [Google Scholar]; One of two seminal 2016 studies that identified three molecular subgroups of SMARCB1-deficient ATRT based on an integrated analysis of DNA methylation and gene expression.
- 22.Hasselblatt M, Isken S, Linge A, et al. High-resolution genomic analysis suggests the absence of recurrent genomic alterations other than SMARCB1 aberrations in atypical teratoid/rhabdoid tumors. Genes Chromosomes Cancer. 2013;52(2):185–190. [DOI] [PubMed] [Google Scholar]
- 23.Clapier CR, Iwasa J, Cairns BR, et al. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat Rev Mol Cell Biol. 2017;18(7):407–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Alver BH, Kim KH, Lu P, et al. The SWI/SNF chromatin remodelling complex is required for maintenance of lineage specific enhancers. Nat Commun. 2017;8:14648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Michel BC, D’Avino AR, Cassel SH, et al. A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nat Cell Biol. 2018;20(12):1410–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wang X, Wang S, Troisi EC, et al. BRD9 defines a SWI/SNF sub-complex and constitutes a specific vulnerability in malignant rhabdoid tumors. Nat Commun. 2019;10(1):1881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Erkek S, Johann PD, Finetti MA, et al. Comprehensive analysis of chromatin states in atypical teratoid/rhabdoid tumor identifies diverging roles for SWI/SNF and polycomb in gene regulation. Cancer Cell. 2019;35(1):95–110.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.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 U S A. 2013;110 (19):7922–7927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Caramel J, Quignon F, Delattre O. RhoA-dependent regulation of cell migration by the tumor suppressor hSNF5/INI1. Cancer Res. 2008;68(15):6154–6161. [DOI] [PubMed] [Google Scholar]
- 30.Medjkane S, Novikov E, Versteege I, et al. The tumor suppressor hSNF5/INI1 modulates cell growth and actin cytoskeleton organization. Cancer Res. 2004;64(10):3406–3413. [DOI] [PubMed] [Google Scholar]
- 31.Lee S, Cimica V, Ramachandra N, et al. Aurora A is a repressed effector target of the chromatin remodeling protein INI1/hSNF5 required for rhabdoid tumor cell survival. Cancer Res. 2011;71 (9):3225–3235. [DOI] [PubMed] [Google Scholar]
- 32.Betz BL, Strobeck MW, Reisman DN, et al. Re-expression of hSNF5/INI1/BAF47 in pediatric tumor cells leads to G1 arrest associated with induction of p16ink4a and activation of RB. Oncogene. 2002;21(34):5193–5203. [DOI] [PubMed] [Google Scholar]
- 33.Isakoff MS, Sansam CG, Tamayo P, et al. Inactivation of the Snf5 tumor suppressor stimulates cell cycle progression and cooperates with p53 loss in oncogenic transformation. Proc Natl Acad Sci U S A. 2005;102(49):17745–17750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.DelBove J, Kuwahara Y, Mora-Blanco EL, et al. Inactivation of SNF5 cooperates with p53 loss to accelerate tumor formation in Snf5(±); p53(±) mice. Mol Carcinog. 2009;48(12):1139–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Strobeck MW, Fribourg AF, Puga A, et al. Restoration of retinoblastoma mediated signaling to Cdk2 results in cell cycle arrest. Oncogene. 2000;19(15):1857–1867. [DOI] [PubMed] [Google Scholar]
- 36.Zhang HS, Gavin M, Dahiya A, et al. Exit from G1 and S phase of the cell cycle is regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb-hSWI/SNF. Cell. 2000;101(1):79–89. [DOI] [PubMed] [Google Scholar]
- 37.Jagani Z, Mora-Blanco EL, Sansam CG, et al. Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nat Med. 2010;16(12):1429–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Barker N, Hurlstone A, Musisi H, et al. The chromatin remodelling factor Brg-1 interacts with beta-catenin to promote target gene activation. Embo J. 2001;20(17):4935–4943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mora-Blanco EL, Mishina Y, Tillman EJ, et al. Activation of β-catenin/TCF targets following loss of the tumor suppressor SNF5. Oncogene. 2014;33(7):933–938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cheng SW, Davies KP, Yung E, et al. c-MYC interacts with INI1/hSNF5 and requires the SWI/SNF complex for transactivation function. Nat Genet. 1999;22(1):102–105. [DOI] [PubMed] [Google Scholar]
- 41.Park J, Wood MA, Cole MD. BAF53 forms distinct nuclear complexes and functions as a critical c-Myc-interacting nuclear cofactor for oncogenic transformation. Mol Cell Biol. 2002;22(5):1307–1316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Weissmiller AM, Wang J, Lorey SL, et al. Inhibition of MYC by the SMARCB1 tumor suppressor. Nat Commun. 2019;10(1):2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Torchia J, Picard D, Lafay-Cousin L, et al. Molecular subgroups of atypical teratoid rhabdoid tumours in children: an integrated genomic and clinicopathological analysis. Lancet Oncol. 2015;16 (5):569–582. [DOI] [PubMed] [Google Scholar]
- 44.••.Torchia J, Golbourn B, Feng S, et al. Integrated (epi)-genomic analyses identify subgroup-specific therapeutic targets in CNS rhabdoid tumors. Cancer Cell. 2016;30(6):891–908. [DOI] [PMC free article] [PubMed] [Google Scholar]; One of two landmark 2016 manuscripts detailing three molecular subgroups of SMARCB1-altered ATRT using DNA methylation, transcriptional profiling by array and ATAC-Seq. A limited in vitro and in vivo drug screen indicated molecular subtype-specific therapeutic vulnerabilities.
- 45.Hasselblatt M, Nagel I, Oyen F, et al. SMARCA4-mutated atypical teratoid/rhabdoid tumors are associated with inherited germline alterations and poor prognosis. Acta Neuropathol. 2014;128 (3):453–456. [DOI] [PubMed] [Google Scholar]
- 46.Frühwald MC, Hasselblatt M, Nemes K, et al. Age and DNA methylation subgroup as potential independent risk factors for treatment stratification in children with atypical teratoid/rhabdoid tumors. Neuro Oncol. 2020;22(7):1006–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Upadhyaya SA, Robinson GW, Onar-Thomas A, et al. Relevance of molecular groups in children with newly diagnosed atypical teratoid rhabdoid tumor: results from prospective St. Jude multi-institutional trials. Clin Cancer Res. 2021;27(10):2879–2889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chi SN, Zimmerman MA, Yao X, et al. Intensive multimodality treatment for children with newly diagnosed CNS atypical teratoid rhabdoid tumor. J Clin Oncol. 2009;27(3):385–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Underiner RM, Eltobgy M, Stanek JR, et al. Meta-analysis of treatment modalities in metastatic atypical teratoid/rhabdoid tumors in children. Pediatr Neurol. 2020;108:106–112. [DOI] [PubMed] [Google Scholar]
- 50.Tekautz TM, Fuller CE, Blaney S, et al. Atypical teratoid/rhabdoid tumors (ATRT): improved survival in children 3 years of age and older with radiation therapy and high-dose alkylator-based chemotherapy. J Clin Oncol. 2005;23(7):1491–1499. [DOI] [PubMed] [Google Scholar]
- 51.Kerl K, Ries D, Unland R, et al. The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells. BMC Cancer. 2013;13:286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Thiemann M, Oertel S, Ehemann V, et al. In vivo efficacy of the histone deacetylase inhibitor suberoylanilide hydroxamic acid in combination with radiotherapy in a malignant rhabdoid tumor mouse model. Radiat Oncol. 2012;7:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hummel TR, Wagner L, Ahern C, et al. A pediatric phase 1 trial of vorinostat and temozolomide in relapsed or refractory primary brain or spinal cord tumors: a Children’s Oncology Group phase 1 consortium study. Pediatr Blood Cancer. 2013;60(9):1452–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wetmore C, Boyett J, Li S, et al. Alisertib is active as single agent in recurrent atypical teratoid rhabdoid tumors in 4 children. Neuro Oncol. 2015;17(6):882–888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Geoerger B, Bourdeaut F, DuBois SG, et al. A phase I study of the CDK4/6 inhibitor ribociclib (LEE011) in pediatric patients with malignant rhabdoid tumors, neuroblastoma, and other solid tumors. Clin Cancer Res. 2017;23(10):2433–2441. [DOI] [PubMed] [Google Scholar]
- 56.Oberlick EM, Rees MG, Seashore-Ludlow B, et al. Small-molecule and CRISPR screening converge to reveal receptor tyrosine kinase dependencies in pediatric rhabdoid tumors. Cell Rep. 2019;28 (9):2331–44.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Weber CM, Hafner A, Kirkland JG, et al. mSWI/SNF promotes Polycomb repression both directly and through genome-wide redistribution. Nat Struct Mol Biol. 2021;28(6):501–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Alimova I, Birks DK, Harris PS, et al. Inhibition of EZH2 suppresses self-renewal and induces radiation sensitivity in atypical rhabdoid teratoid tumor cells. Neuro Oncol. 2013;15 (2):149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Chi SN, Bourdeaut F, Laetsch TW, et al. Phase I study of tazemetostat, an enhancer of zeste homolog-2 inhibitor, in pediatric pts with relapsed/refractory integrase interactor 1-negative tumors. J Clin Oncol. 2020;38(15_suppl):10525. [Google Scholar]
