Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2023 Feb 9;37:100880. doi: 10.1016/j.neo.2023.100880

Recent progress and novel approaches to treating atypical teratoid rhabdoid tumor

Elizabeth Alva a, Jeffrey Rubens b, Susan Chi c, Tom Rosenberg c, Alyssa Reddy d, Eric H Raabe b,, Ashley Margol e,
PMCID: PMC9929860  PMID: 36773516

Abstract

Atypical teratoid rhabdoid tumors (AT/RT) are malignant central nervous system (CNS) tumors that occur mostly in young children and have historically carried a very poor prognosis. While recent clinical trial results show that this tumor is curable, outcomes are still poor compared to other central nervous system embryonal tumors. We here review prior AT/RT clinical trials and highlight promising pre-clinical results that may inform novel clinical approaches to this aggressive cancer.

Keywords: INI1, MYC, epigenetics, adaptive, clinical trial, mTOR, MEK

Introduction

While only representing 3% of all pediatric CNS tumors, AT/RT is the most common malignant CNS tumor in children less than one year of age and represents 20% of CNS tumors in children less than three years of age [1], [2], [3], [4],. The World Health Organization recognized AT/RT as a formal diagnostic category in 2000 [5,6]. Bi-allelic loss of function mutations in the SMARCB1 gene define the majority of cases of AT/RT [7], [8], [9]. The remaining cases carry loss of function mutations in the SMARCA4 gene [10]. Although the morphologic appearance of AT/RT varies significantly, the loss of INI1 (SMARCB1) or BRG1 (SMARCA4) by immunohistochemical staining allows for rapid histologic diagnosis [10,11]. Approximately one-third of patients with AT/RT have an underlying germline SMARCB1 alteration resulting in rhabdoid tumor predisposition syndrome, carrying a risk of multiple CNS and non-CNS rhabdoid tumors [12]. Although AT/RT harbor no recurrent genetic alterations outside of SMARCB1 and SMARCA4, AT/RT can be grouped into three consensus molecular subgroups defined by DNA methylation and RNA expression [13], [14], [15].

Following the recognition of AT/RT as a distinct entity, disease specific approaches using intensive, multimodal therapies have led to improved survival outcomes [16], [17], [18]. Toxicity of current treatment options is high [16,17,[19], [20], [21]] and there is still much to understand about how molecular subgroups and other prognostic factors may be used in future clinical trials to stratify treatment. Novel therapies and innovative approaches to clinical trial design are potential opportunities to continue to improve outcomes for these patients.

Methods

We performed a search of PubMed using the following combinations of keywords: “teratoid,” “rhabdoid,” and “therapeutic.” This retrieved a total of 347 articles that were reviewed for applicability for AT/RT. We prioritized for inclusion prospective clinical trials including AT/RT patients as well as large registry data. Abstracts from recent scientific meetings were also included if reporting results of a recently completed or ongoing clinical trial including AT/RT patients. In addition, we prioritized for inclusion publications describing novel therapeutics that demonstrated efficacy in animal models. We obtained additional references from initially selected articles.

Discussion

Completed trials for newly diagnosed AT/RT patients

Due to the rarity of this tumor type, the majority of historical data on treatment of AT/RT arises from retrospective reviews and small studies. Though historical “baby” protocols for young children with CNS tumors included patients with AT/RT, only recently have larger, prospective studies been completed that were designed specifically for AT/RT (Table 1).

Table 1.

Prospective Clinical Trials in AT/RT

Protocol Number of Patients (n) Chemotherapy Regimen Timing of Radiation: (n) Outcomes
CCG921 28 Regimen A: VCR, CDDP, CPM, ETOP x 5 cycles
Regimen B: VCR, CBP, IFOS, ETOP x 5 cycles
Maintenance: VCR, ETOP, CBP, ETOP x 8 cycles
Adjuvant: 2
Salvage: 9
5-yr EFS: 14%±7%
5-yr OS: 29%±9%
Head Start III 19 Induction Cycle 1, 3, 5: CDDP, VCR, ETOP, CPM, HDMTX
Induction Cycle 2, 4: TEM, ETOP, VCR, CPM
Consolidation with HDCT: CBP, Thiotepa, ETOP
Salvage: 5 3-yr EFS: 21%±9%
3-yr OS: 26%±10%
DFCI IRS-III 20 VCR, ACT-D, CPM, CDDP, DOX, TEM
IT chemotherapy: MTX, HCT, ARA-C
Adjuvant: 15
2-yr PFS: 53%±13%
2-yr OS: 70%±10%
ACNS0333 65 Induction: MTX, VCR, ETOP, CPM, CDDP x 2 cycles
Consolidation with HDCT: CBP, Thiotepa, ETOP x 3 cycles
Adjuvant: 42
4-yr EFS: 37%
4-yr OS: 43%
SJYC07 52 IR:
Induction: HDMTX, VCR, CPM, CDDP x 4 cycles
Maintenance: PO CTX/TOPO alternating with PO ETOP
HR:
Induction: HDMTX, VCR, CPM, CDDP, VBL x 4 cycles
Consolidation: CPM/TOPO x 2 cycles
Maintenance: PO CPM/TOPO alternating with PO ETOP
Adjuvant: 34 (IR only) IR:
5-yr PFS: 31.4%±9.2%
5-yr OS: 43.9%±9.5%
HR:
5yr PFS and OS: 0%
SJMB03 22 CDDP, VCR, CPM followed by PBSC x 4 cycles Adjuvant: 22 AR:
5-yr PFS: 72.7%±12.7%
5-yr OS: 81.8% ±11.0%
HR:
5-yr PFS and OS: 18.2%±9.5%

Abbreviations: ACT-D, dactinomycin; AR, average risk; ARA-C, cytarabine; CBP, carboplatin; CDDP, cisplatin; CPM, cyclophosphamide; DFCI, Dana-Farber Cancer Institute; DOX, doxorubicin; EFS, event free survival; ETOP, etoposide; HCT, hydrocortisone; HDCT, high dose chemotherapy; HDMTX, high dose methotrexate; HR; high risk; IFOS, ifosfamide; IR, intermediate risk; IRS, Intergroup Rhabdomyosarcoma Study; MTX, methotrexate; OS, overall survival; PBSC, peripheral blood stem cells; PO, oral; TEM, temozolomide; TOPO, topotecan; VCR, vincristine;

CCG9921

One early study through the Children's Cancer Group, CCG9921, randomized subjects to one of two intensive platinum-based multi-agent chemotherapy regimens, followed by maintenance chemotherapy with the goal of avoiding radiation therapy [22]. This study was designed for malignant brain tumors of varying histologies in infants < 36 months of age and included 28 AT/RT patients. Eleven of the 28 AT/RT patients received radiation therapy on study, with nine of those receiving irradiation at the time of disease recurrence or progression. Patients with AT/RT experienced worse outcomes compared to infants with tumors of other histologies, and the 5-year event free survival (EFS) and overall survival (OS) were 14% and 29%, respectively, which was not improved compared to historical controls.

Head Start

Efforts to intensify chemotherapy to avoid radiation therapy in these young patients continued with the Head Start protocols, where sequential prospective studies included 13 patients on Head Start 1 and II and 19 patients on Head Start III [19,23]. After initial surgical resection, patients received five cycles of multi-agent chemotherapy followed by one cycle of high dose chemotherapy (HDC) with autologous stem cell rescue. On Head Start III, owing to toxicity and progressive disease in patients during induction, only four of 19 patients completed all five cycles of chemotherapy and only three proceed to consolidation. The 3-year EFS and OS were 21% and 26%, respectively.

DFCI-AT/RT

The first prospective trial designed specifically for AT/RT patients, a multi-institutional trial (NCT00084838) led by Dana-Farber Cancer Institute (DFCI), consisted of a chemotherapy backbone based on the Intergroup Rhabdomyosarcoma Group-III protocol for rhabdomyosarcoma with parameningeal extension. Upfront surgical resection was followed by 51 weeks of multi-agent systemic anthracycline-based chemotherapy with concurrent intrathecal chemotherapy. After the initial six weeks of induction chemotherapy, patients received radiation with concurrent chemotherapy. Patients with M0 disease received focal radiation therapy. Patients older than three years with M+ disease received craniospinal irradiation (CSI). Survival was encouraging with 2-year progression free survival (PFS) and OS of 53% and 70%, respectively. Extent of resection was prognostic by univariate analyses, with patients who achieved gross total resection demonstrating a 2-year OS of 91%. Tumor location also influenced OS, with posterior fossa location being favorable over supratentorial location. As expected, this intensive regimen was associated with frequent grade 3-4 toxicities. There was one case of toxic death during induction, one case of transverse myelitis that required therapy cessation and two patients that experienced radiation recall, one near therapy completion and another following it.

EU-RHAB

The European Registry for rhabdoid tumors (EU-RHAB) has published results on 31 patients uniformly treated with systemic and intrathecal chemotherapy [24]. Additional therapy modalities included radiotherapy in 23 patients, HDCT in 8 patients and additional maintenance chemotherapy for 17 patients. For the entire study cohort, the reported 6-year event-free survival (EFS) and OS were 46% and 45%, respectively. Age > 3 years and metastatic status were found to be prognostic factors of statistical significance in this study. In addition, the ability to reach a complete remission and the incorporation of radiotherapy were statistically significant positive prognostic factors. Major toxicities reported in this study included one treatment related death secondary to VP-shunt failure and three instances of treatment discontinuation due to pneumonia, recurrent infections and methotrexate induced leukoencephalopathy. In a more recent publication from the EU-RHAB group, results from 143 patients are reported. Outcomes reported for this larger cohort were 5-year EFS and OS of 31% and 35%, respectively. Age remained an important determinant of outcome, as did metastatic disease and synchronous tumors. Methylation profiling was available for 84/143 patients, showing a survival advantage for patients with ATRT-TYR subgroup when compared to non-TYR ATRT [18].

ACNS0333

The largest prospective trial specifically for patients with AT/RT, the Children's Oncology Group (COG) trial ACNS0333, combined two cycles of intensive multi-agent chemotherapy including high dose methotrexate with three tandem cycles of HDC with autologous stem cell rescue [17]. Second-look surgery was encouraged prior to consolidation. Timing and extent of radiation therapy ware based on age and extent of disease at presentation. Sixty-five evaluable patients with AT/RT were enrolled on study and two year EFS was 42%. Four-year EFS and OS were 37% and 43% respectively. Approximately a third of patients had disease relapse on treatment with a cumulative incidence of relapse (CIR) of 22% at 6 months and 41% at one year [25]. Age at diagnosis, location, extent of initial resection, and presence of metastatic disease were not found to be prognostic for EFS, OS, or CIR. Patients with SMARCB1 germline mutation or tumors that involved the infratentorial and supratentorial compartments simultaneously had a worse outcome, but the number of patients in each category was too small to reach significance. Main toxicities of ACNS0333 included myelosuppression and infectious complications. There were four treatment-related deaths: one from sepsis during induction, one from pulmonary fibrosis, and two from CNS necrosis. Of note, the study was amended to decrease the risk of pulmonary toxicity, with no further cases following these changes. Retrospective molecular subtyping was performed on tumors of children enrolled on this study, and each was identified as one of the 3 molecular subtypes (TYR, SHH and MYC) previously reported [13,14]. While not powered to evaluate the effect of subtype on outcome, it is notable that patients with SHH subtype had a 6-month EFS of 100%. Thirty-four of the 40 patients who received RT had focal RT only. Patients with no evidence of disease at the time of radiation had a 4-year OS of 79% (Reddy, personal communication). There was no difference in EFS/OS/CIR based on the order of radiation and consolidation, but there was increased neurotoxicity when radiation was administered between induction and consolidation. The investigators recommend that patients receive radiation after consolidation on future trials that use this backbone.

SJYC07 and SJMB03

The St. Jude Children's Research Hospital prospective risk-adapted trials SJYC07 and SJMB03 also showed that multimodal therapy can achieve long-term survival in some children with AT/RT [26]. The SJYC07 cohort included 55 infants less than 36 months of age and stratified patients into intermediate risk and high risk based on the absence or presence of metastatic disease. After maximal safe upfront resection, intermediate risk patients were treated with 4 cycles of multi-agent induction chemotherapy followed by focal radiation and maintenance chemotherapy for 24 weeks. Second-look surgery was performed in patients with residual disease after 2-4 cycles of induction chemotherapy. High-risk patients received 4 cycles of induction chemotherapy similar to intermediate risk patients but with the addition of vinblastine, followed by consolidation with 2 cycles of cyclophosphamide and topotecan, and oral maintenance therapy. Craniospinal irradiation (CSI) was optional for patients who were ≥ 3 years of age at the end of induction. The 5-year PFS and OS for intermediate risk patients were 31% and 44%, respectively. Unfortunately, 5-year PFS and OS for high risk patients was 0%. SJMB03 enrolled 22 children ≥ 36 months of age and stratified patients to average risk (M0 and < 1.5cm2 of residual tumor) or high risk (M+ or ≥ 1.5cm2 of residual tumor). All patients received risk-adapted CSI with boost to primary and metastatic sites. CSI was followed by 4 cycles of multi-agent chemotherapy with peripheral blood stem cell support. Five-year PFS and OS for average-risk patients were 73% and 82%, respectively. For high-risk patients, 5-year PFS and OS were 18%. There was no evidence that presence of a germline SMARCB1 or SMARCA4 mutation influenced outcome in these cohorts. For both SJYC07 and SJMB03, the most common toxicity was febrile neutropenia, with one reported case of death following febrile neutropenia and a respiratory infection. Molecular subtypes were retrospectively evaluated and suggest that infants with TYR subgroup AT/RT treated on SJYC07 had improved outcomes compared to those with SHH or MYC subgroups. However, outcomes for patients without metastases at diagnosis were similar between the different AT/RT molecular subgroups.

Impact of extent of resection

Evaluation of these clinical trials shows a mixed picture of the impact of GTR on survival. While the DFCI trial showed in univariate analysis that GTR significantly improved survival [27], in the ACNS0333 cohort GTR vs STR vs biopsy did not affect survival [17]. Similarly, in the European EU-RHAB AT/RT cohort, extent of resection did not significantly affect outcome [24]. In the St. Jude series, in the SJYC07 cohort of children less than 3 years, second-look surgery was encouraged to achieve a GTR. In this cohort, outcomes did not differ by extent of resection. In the SJMB03 older cohort of children greater than 3 years of age, the extent of resection was incorporated into risk stratification, and patients with greater than 1.5 cm2 of residual tumor were classified as high risk. This group, which also included patients with metastatic disease at outset, did significantly worse than average risk patients [26].

Influence of epigenetic subtype

There are 3 consensus epigenetic subgroups of AT/RT: TYR, SHH, and MYC [3,[28], [29], [30]]. These subgroups differ in gene expression profile, location, method of inactivating SMARCB1 and in susceptibility to targeted therapy [29], [30], [31]. In the ACNS0333 cohort, there was a trend towards increasing risk of relapse in patients with the MYC subgroup, but this has not reached statistical significance [25]. A close examination of the SJYC07 data shows that TYR patients were less likely to be metastatic at onset, and when only M0 patients were included, there was no difference in outcome based on epigenetic subtype [26]. Similarly, in the EU-RHAB cohort, there is no significant difference in OS when TYR vs SHH vs MYC subgroups are compared [18]. Therefore, we conclude that there is no definitive evidence to date that any methylation group does better or worse. With larger cohorts, a trend for differential outcomes by subgroup may become more apparent.

Effect of radiation therapy

The timing, dose and field of radiation prescribed by each regimen vary. The DFCI protocol included focal XRT for patients with M0 disease and for children with M+ disease < 3yrs, while those ≥3 yrs of age with M+ disease received CSI. ACNS0333 recommended CSI for children with metastatic disease while children with localized disease received focal XRT. Of note, there were 3 children treated on ACNS0333 who were <3yrs of age with metastatic disease and received focal XRT only – all 3 were alive at last study follow-up suggesting that focal XRT may be sufficient for patients with M+ disease [32]. For children ≥3yrs of age without metastatic or residual disease, SJMB03 employs 23.4Gy CSI with a boost to the primary site.

The treatment-related toxicity associated with these regimens is significant, making it unlikely that further intensification of conventional therapy will lead to additional survival improvement for patients [16,17,[19], [20], [21]]. Additionally, given the devastating effects of radiation therapy in this very young patient population, strategies to avoid radiation should be developed whenever possible. As previously mentioned, patients treated on Head Start with no radiation therapy had a dismal prognosis, however most of the patients progressed early during chemotherapy. A multi-institutional investigation of patients treated on or as per ACNS0333 found that in a cohort of 80 patients, children who received radiation therapy had a higher EFS and OS than children who did not experience disease progression on therapy and did not receive radiation therapy due to parent or physician preference [33]. When using the ACNS0333 regimen, induction and consolidation chemotherapy alone are not sufficient. While these studies demonstrate that AT/RT can be cured with aggressive, multimodal therapy, the relatively poor outcomes and therapy- associated toxicity highlight the need for novel treatment modalities to improve outcomes for children with AT/RT.

Summary of upfront AT/RT clinical trials

Although the aforementioned trials have shown improvement in outcomes for patients with AT/RT, they used varied approaches. Therefore, there is no established standard of care for newly diagnosed patients with AT/RT. Patients treated on ACNS0333 and the DFCI protocol had similar characteristics and outcomes. For children <3yrs of age, we recommend treatment as per ACNS0333 or the DFCI protocol. SJMB03 should also be considered for patients ≥3yrs of age with no residual or metastatic disease. Decisions regarding therapy must consider the age of the patient, size and location of the tumor, resources of the treating facility, and family preferences.

Completed trials for patients with relapsed/recurrent AT/RT

There have been only a few studies designed specifically for patients with recurrent AT/RT, although patients with relapsed/refractory AT/RT have been included in a variety of other early phase clinical trials.

The Pediatric Brain Tumor Consortium (PBTC) conducted an open-label phase II studies of oxaliplatin that included a stratum for recurrent AT/RT, along with medulloblastoma and supratentorial primitive neuroectodermal tumor (PNET) [34]. Of the 43 patients included on the trial, 5 had AT/RT. No AT/RT patients responded. However, one patient with AT/RT had prolonged stable disease for 17 courses while on study and received an additional 17 courses of oxaliplatin off protocol.

Based on a preclinical data suggesting activity of the cyclin-dependent kinase (CDK) 4/6 inhibitor ribociclib in malignant rhabdoid tumors (MRTs), a multi-center phase I study was pursued with this agent [35]. The study included an initial dose-escalation phase, followed by an expansion phase in patients with neuroblastoma and rhabdoid tumors, including AT/RT. Of the 32 patients enrolled on trial, 13 had AT/RT. Ribociclib was given once daily on a 3-weeks-on/1-week-off schedule in 28-day cycles. Although no responses were seen, 2 patients with AT/RT were found to have prolonged stable disease, remaining on treatment for 20 and 24 months.

Another targeted agent of interest in the relapse setting has been the Aurora A kinase inhibitor, alisertib. In a phase II study conducted by the COG in recurrent or refractory solid tumors and leukemia, 2 patients with AT/RT were included and treated. Although the drug was tolerable, no responses were seen in the AT/RT patients on trial. The objective response rate for all patients on trial (n=137) was less than 5% [36]. St. Jude Children's Research Hospital also conducted a phase II trial of single-agent alisertib specifically for recurrent or progressive AT/RT [37]. Thirty patients, representing all three molecular subgroups, were included. Only eight of 29 evaluable patients were without progressive disease by 12 weeks and thus the study did not meet the efficacy endpoint. However, PFS was 31%±8.2% at 6 months and 15.8%±6.5% at 1 year. One-third of patients had stable disease for greater than 6 months and two patients remained on therapy for greater than one year.

SMARCB1 or SMARCA4 deletions in AT/RT lead to an abnormal epigenetic landscape that transforms tumors and drives their rapid growth and survival. Therefore, targeting the abnormal epigenetics of these tumors is highly attractive. The deletion of SMARCB1 or SMARCA4 in AT/RT releases SWI/SNF inhibition of EZH2 methyltransferase activity leading to increased H3K27me3 repressive marks at the promoter regions of tumor suppressor and neuronal differentiating genes [38]. Knockdown of EZH2 in rhabdoid tumors prevents orthotopic tumor formation, demonstrating its importance in the establishment and growth of SMARCB1-deficient tumors [39]. The EZH2 inhibitor tazemetostat was studied in a phase 1 pediatric trial. Tazemetostat demonstrated one complete response and 5/21 objective responses in patients with AT/RT, with a median duration of 6.5 months [40,41]. In the COG-NCI Pediatric MATCH trial, tazemetostat demonstrated no objective responses in patients with AT/RT, however, 2/8 patients had stable disease at 6 and 13 months [42].

The current landscape of clinical trials for children with AT/RT

Currently, there is one ongoing clinic trial for newly diagnosed AT/RT patients at St Jude Children's Research Hospital, evaluating the Aurora A kinase inhibitor, alisertib, in combination with age-and risk-adaptive chemotherapy along with radiation therapy (NCT02114229). There are also ongoing early phase clinical trials for children with progressive or recurrent AT/RT investigating molecularly targeted agents as well as immunotherapies (Table 2). These studies are ongoing, and preliminary data are not yet available.

Table 2.

Ongoing Clinical Trials for Relapsed/Refractory AT/RT

Phase Title Therapeutic Category AT/RT or INI-1 deficient tumor specific Status
II Phase 2 Proof of Concept Study of Nivolumab and Ipilimumab in Children and Young Adults With Relapsed or Refractory INI1-negative Cancers (NCT04416568) Immunotherapy Yes Recruiting
I/II TAZNI: A Phase I/II Combination Trial of Tazemetostat With Nivolumab and Ipilimumab for Children With INI-1 Negative or SMARCA4-Deficient Tumors (NCT05407441) Immunotherapy + targeted agent Yes Not yet recruiting
II A Phase 2 Study of Tiragolumab (NSC# 827799) and Atezolizumab (NSC# 783608) in Patients With Relapsed or Refractory SMARCB1 or SMARCA4 Deficient Tumors (NCT05286801) Immunotherapy Yes Not yet recruiting
I Combination Intraventricular Chemotherapy Pilot Study: Methotrexate and Etoposide Infusions Into the Fourth Ventricle or Resection Cavity in Children With Recurrent Posterior Fossa Brain Tumors (NCT02905110) IT chemotherapy No Recruiting
I A Phase 1 Study of Modified Measles Virus (MV-NIS) for the Treatment of Children and Young Adults With Recurrent Medulloblastoma or Recurrent Atypical Teratoid Rhabdoid Tumors (NCT02962167) immunotherapy No Recruiting
I Phase I Study to Evaluate the Safety and Tolerability of the CD40 Agonistic Monoclonal Antibody APX005M in Pediatric Subjects With Recurrent/Refractory Brain Tumors and Newly Diagnosed Brain Stem Glioma (NCT03389802) Immunotherapy No Recruiting
I Phase 1 Study of HER2-Specific CAR T Cell Locoregional Immunotherapy for HER2 Positive Recurrent/Refractory Pediatric Central Nervous System Tumors (NCT03500991) Immunotherapy No Recruiting
I Phase 1 Study of EGFR806-specific CAR T Cell Locoregional Immunotherapy for EGFR-positive Recurrent or Refractory Pediatric Central Nervous System Tumors (NCT03638167) Immunotherapy No Recruiting
I Phase 1 Study of B7-H3-Specific CAR T Cell Locoregional Immunotherapy for Diffuse Intrinsic Pontine Glioma/Diffuse Midline Glioma and Recurrent or Refractory Pediatric Central Nervous System Tumors (NCT04185038) Immunotherapy No Recruiting
I Phase Ib Study of Oncolytic Polio/Rhinovirus Recombinant Against Recurrent Malignant Glioma in Children (NCT03043391) Immunotherapy No Active, not recruiting
I Phase 1 Trial of Engineered HSV G207 in Children With Recurrent or Refractory Cerebellar Brain Tumors (NCT03911388) Immunotherapy No Recruiting
II A Phase II Study of Metronomic and Targeted Anti-angiogenesis Therapy for Children With Recurrent/Progressive Medulloblastoma, Ependymoma and ATRT (NCT01356290) Metronomic chemotherapy No Recruiting
II AflacST1502: A Phase II Study of Sirolimus in Combination With Metronomic Chemotherapy in Children With Recurrent and/or Refractory Solid and CNS Tumors (NCT02574728) Metronomic chemotherapy No Recruiting
I Molecularly-Driven Doublet Therapy for All Children With Refractory or Recurrent CNS Malignant Neoplasms and Young Adults With Refractory or Recurrent SHH Medulloblastoma (NCT03434262) Targeted agents No Recruiting
I Phase 1 Study of 9-ING-41, a Glycogen Synthase Kinase 3 Beta (GSK 3β) Inhibitor, as a Single Agent or With Irinotecan, Irinotecan Plus Temozolomide, or With Cyclophosphamide Plus Topotecan in Pediatric Patients With Refractory Malignancies (NCT04239092) Targeted agent + chemotherapy No Recruiting
I Abemaciclib in Children With Newly Diagnosed Diffuse Intrinsic Pontine Glioma, and in Children With Recurrent and Refractory Solid Tumors Including Malignant Brain Tumors (NCT02644460) Targeted agent No Recruiting
I An Open-Label, Dose Escalation, Efficacy, and Safety Study of CLR 131 in Children, Adolescents, and Young Adults With Select Solid Tumors, Lymphoma, and Malignant Brain Tumors (NCT03478462) Radioisotope No Recruiting

The Pacific Pediatric Neuro-oncology Consortium (PNOC) is conducting a phase I study of modified measles virus (MV-NIS) directly into the tumor bed for locally recurrent disease or into the subarachnoid space via lumbar puncture for those with disseminate recurrence (NCT02962167). This trial is based on preclinical data showing survival benefit in in vivo models of both focal and disseminated AT/RT when treated with measles virus injected either intratumorally or intraventricularly [43].

A multi-institutional trial based at Dana-Farber Cancer Institute is investigating the safety and efficacy of checkpoint inhibitor therapy (nivolumab in combination with ipilimumab) for children with relapsed/recurrent INI1-negative tumors (NCT04416568). Although tumor mutational burden is low in AT/RT, preclinical data supports the potential immunogenicity of AT/RT and use of immunotherapy in treatment [44].

The recent identification of B7-H3/CD276 as being highly expressed in pediatric brain tumors (including 100% of human AT/RT tumors with moderate to high level expression), in contrast to this antigen being expressed at low levels elsewhere in the body, suggests B7-H3 as a potential immunotherapeutic target [45]. Previous studies in human B7-H3-expressing colon, breast, and ovarian cancer cell lines showed that B7-H3 antibody-drug conjugates (ADC) killed B7-H3 expressing tumor cells as well as B7-H3 expressing tumor vasculature and spared normal cells, significantly extending the life of mice bearing flank or orthotopic tumors [46]. Similarly, the ADC m276-SL-PBD demonstrated broad activity against pediatric solid malignancies, including flank tumors of AT/RT [47]. B7-H3 generated CAR-T cell therapy when injected intratumorally or intraventricularly eradicated BT16 AT/RT orthotopic xenografts and persisted in the brain, preventing growth of tumors on rechallenge [48]. B7-H3 CAR-T cell clinical trials for children with relapsed/refractory solid tumors are open at several sites (NCT04897321, NCT04185038).

Potential novel agents for use in AT/RT

While in vitro high-throughput or hypothesis-based screening can provide an indication of activity of an agent against AT/RT, validating agents in pre-clinical models in general provides the best predictor of success in humans [49]. Embryonal cancer cells change their metabolism and epigenetics when grown as orthotopic tumors in mice compared to cells in culture and even flank tumors, making pre-clinical study in orthotopic models essential to justify the risks of testing novel agents within a clinical trial setting, especially for young children [50]. A number of promising clinical agents have advanced from the in vitro setting to demonstrating a survival advantage in pre-clinical in vivo models. Fig. 1 shows a summary cartoon of the mechanism of action of potential novel agents for treating AT/RT. Table 3 summarizes the clinical development of these potential novel agents.

Fig. 1.

Fig 1

Cartoon summarizing potential novel therapeutic agents for use in AT/RT.Potential agents are color coded, with signal transduction inhibitors in green, stress activators in blue, epigenetic agents in purple, and cell cycle acting agents indicated in red. Some agents act in multiple ways to inhibit AT/RT proliferation and promote death. Epigenetic alterations downstream of loss of SMARCB1 help AT/RT tumors to access developmentally significant growth and resistance programs that allow tumor cells to tolerate replicative, epigenetic and endoplasmic reticulum (ER) stress. Upregulation of platelet derived growth factor receptor B (PDGFB) may drive cell proliferation, and this may be blocked by the multi-kinase inhibitor dasatinib. Other agents targeting downstream effectors such as PI3K/mTOR, MEK and MELK may deprive AT/RT cells of key growth and survival pathways. Metabolic targeting agents such as 6-diazo-5-oxo-l-norleucine (DON) and its prodrug DRP-104 disrupt the metabolic dependencies of fast-growing AT/RT cells and synergize with traditional chemotherapy. Blocking the proteasome with inhibitors such as ixazomib may lead to further activation of the unfolded protein response (UPR) and ER stress. In combination with nutrient deprivation and induction of mitochondrial stress, these multiple stressors may lead to persistent activation of the integrated stress response and cell death. Epigenetic altering drugs such as histone deaceylase (HDAC) inhibitors panobinostat and RG2833 as well as enhancer of zest homologue 2 (EZH2) inhibitors such as tazemetostat and ribavirin may restore epigenetic balance to AT/RT. Similarly, JQ1 and other bromodomain inhibitors may block MYC activation, leading to decreased cell growth. The aurora kinase A inhibitor alisertib may block the mitotic activity of fast-growing AT/RT cells. Acting at the G1 cell cycle checkpoint, cyclin dependent kinase inhibitors such as palbociclib or abemaciclib also prevent AT/RT proliferation. DNA binding agents such as quinacrine and the curaxin CBL-0137 block transcription downstream of nuclear factor kappa B (NFkB) and separately lead to the accumulation and activation of the tumor suppressor TP53. Idasanutlin releases TP53 from MDM2 inhibition, allowing wild type TP53 to become active.

Table 3.

Summary of novel agents under consideration for treatment of AT/RT

Agent Target Mouse in vivo data? Adult RP2D? Pediatric RP2D?
Signal transduction pathway inhibitors
Paxalisib PI3K/mTOR Yes Yes Yes
Sapanisertib/TAK228 TORC1/2 Yes Yes No
Selumetinib MEK No Yes Yes
Binimetinib MEK Yes Yes Yes
OTSSP167 MELK Yes Yes No
Dasatinib PDGFR Yes Yes Yes
Epigenetic modifiers
Tazemetostat EZH2 Yes Yes Yes
Panobinostat Pan HDAC Yes Yes Yes
RG2833 HDAC 1,3 No No No
Quinacrine/CBL0137 FACT Yes Yes Yes
JQ1/TEN-10 BRD4/MYC Yes Yes No
Cellular stress activators
Ribavirin eIF4e Yes Yes Yes
DON/DRP104 Glutamine Yes Yes Yes
Bortezomib/Marizomib/Ixazomib Proteasome Yes Yes Yes
Obatoclax BCL-2, BCL-XL Yes Yes No
Cell cycle targeting
Alisertib Aurora Kinase A Yes Yes Yes
Palbociclib/Abemaciclib CDK4/6 Yes Yes Yes
Idasanutlin TP53 Yes Yes Yes
Immunotherapies
Nivolumab/Ipilimumab PD-1/CTLA-4 Yes Yes Yes
B7-H3 CAR-T cells B7-H3 Yes Yes In progress
m276-SL-PBD B7-H3 Yes No No
MV-NIS CD46 Yes Yes In progress

Agents targeting mTOR

Several groups have demonstrated a vulnerability of AT/RT to mTOR inhibition, specifically using the TORC1/2 kinase inhibitor sapanisertib (also known as TAK228, MLN0128 and INK128) [51], [52], [53]. TAK228 increased survival as a single agent in mice bearing BT12 or CHLA06 orthotopic xenografts, but more impressively suppressed glutathione, and when combined with cisplatin, led to near doubling of median survival [51]. TAK228 and cisplatin combination treatment led to the long-term survival of nearly 40% of the treated mice.

One of the key targets of dual TORC1/2 inhibition is downregulation of the NRF2 transcription factor, which regulates the expression of genes involved in the response to cellular stress, redox homeostasis, metabolism, DNA repair, proliferation, and survival [54]. Inhibition of TORC1/2 and subsequent down-regulation of NRF2 disrupts AT/RT defenses against oxidative stress and cell death. Obatoclax is a brain-penetrant pan-BH3 inhibitor, which induces high levels of oxidative stress and apoptosis in AT/RT. TAK228 combined with obatoclax to increase median survival in mice bearing BT37 and CHLA06 orthotopic xenografts [54].

While the clinical development of sapanisertib is currently uncertain, these results led to the advancement of the highly brain penetrant PI3K/mTOR inhibitor paxalisib as a potential agent of interest. Paxalisib as a single agent led to an increase in survival of 20-30% in mice bearing orthotopic xenografts of CHLA-06 and BT12 [55]. Studies are currently ongoing, evaluating paxalisib's efficacy in combination with targeted as well as traditional chemotherapy. The PNOC AT/RT clinical trial working group is therefore considering advancing paxalisib as a combination therapy in the PNOC AT/RT adaptive clinical trial.

RTK inhibitors

An unbiased ATAC-seq-based approach led to the identification of an open region of chromatin in group 2A (corresponding to MYC subgroup of AT/RT tumors) near the PDGFRB gene promoter [29]. These tumors had increased expression of PDGFRB and sensitivity to the multi-kinase inhibitors nilotinib and dasatinib. Orthotopic xenografts of BT12 treated with dasatinib had modest but significantly longer survival compared to controls [29]. This data aligns with prior studies showing that some AT/RT cell lines demonstrate in vitro sensitivity to dasatinib [56].

MEK inhibitors

AT/RT primary tumors have increased expression of maternal embryonic leucine zipper kinase (MELK) as well as MAP kinase [57,58]. As monotherapy, the MEK inhibitors selumetinib and binimetinib inhibited the growth of AT/RT cells and induced apoptosis [58,59]. Binimetinib suppressed the growth of AT/RT flank xenograft but did not have efficacy against orthotopic xenografts [59]. Neither the MEK inhibitor trametinib nor the MELK inhibitor OTSSP167 significantly increased the survival of mice bearing AT/RT orthotopic xenografts, however combination therapy increased median survival from 22 to 32 days [57].

Metabolic targeting in AT/RT

Rapid proliferation in AT/RT drives dependencies on distinct metabolic pathways to support greater energetic needs and protect against toxic by-products of metabolism as compared to normal brain. Comprehensive metabolic profiling showed that AT/RT cells expressing high levels of MYC protein were dependent on glutamine signaling and were sensitive to glutamine metabolic inhibitors such as 6-diazo-5-oxo-L-norleucine (DON) [31]. DON as a single agent nearly doubled survival in mice bearing BT12 orthotopic xenografts. DON depleted glutathione and combined synergistically with carboplatin in mice bearing orthotopic xenografts of BT12 and CHLA06 [31]. Although DON is not currently available for clinical use, DON prodrugs are currently in clinical trials for adults with solid tumors [60], [61], [62] (NCT04471415).

Proteasome inhibitors

The high growth rate of rhabdoid tumors suggested that these cells might exhibit increased cellular stress. Mouse-derived malignant rhabdoid tumors have increased expression of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress [63]. Treatment of these tumors with proteasome inhibitors such as bortezemib or ixazomib led to regression of tumors and extension of mouse survival [63]. Analysis of AT/RT showed an increase in proteasomal genes compared to normal brain. Targeting AT/RT with the proteasome inhibitor bortezomib killed tumor cells in vitro and extended the survival of mice bearing AT/RT orthotopic xenografts [64].

An unbiased drug screen of AT/RT identified proteasome inhibitors as being highly effective in vitro. The brain-penetrant proteasome inhibitor marizomib demonstrated activity against BT16 and MAF-737A AT/RT orthotopic xenografts [65]. Clinical development of marizomib is currently halted, however, the orally bioavailable and brain-penetrant proteasome inhibitor ixazomib also demonstrated activity against malignant rhabdoid tumor (MRT) in vitro and in vivo and killed AT/RT cells [63]. Ixazomib is currently in pediatric clinical trials in conjunction with anthracyclines for recurrent pre-B ALL (NCT03817320). Ixazomib was also noted in an unbiased drug screen of AT/RT primary cell lines, suppressing growth significantly across the board (J. Cain, personal communication). The positive preclinical results for ixazomib and other proteasome inhibitors suggest that this class may have activity against AT/RT in human patients and may warrant further exploration in a clinical trial.

CDK4/6 inhibitors

Cyclin-dependent kinase 4/6 (CDK 4/6) inhibitors such as ribociclib, palbociclib, and abemaciclib disrupt the proliferation of aggressive tumors with intact RB signaling. CDK 4/6 inhibitors prevent the growth of embryonal tumors in vitro and in orthotopic xenografts [49,66]. In AT/RT, palbociclib alone increased median survival of mice bearing orthotopic BT12 xenografts by 60 percent [67]. Combining palbociclib with radiation therapy led to a more than doubling of median survival compared to control in BT16 orthotopic xenografts [67]. Mark et al., using a CRISPR-genome-wide screen, identified CDK4/6 as a dependency in AT/RT and showed that single-agent abemeciclib significantly extended the life of mice bearing orthotopic BT16 AT/RT xenografts [68]. While single agent ribociclib did not show response in AT/RT patients in a phase I study [35], a phase 1A/1B clinical trial of abemaciclib in combination with temozolomide and irinotecan is currently enrolling pediatric patients with brain and other solid tumors (NCT04238819).

Epigenetic therapies

The anti-viral drug ribavirin has structural similarity to the EZH2 inhibitor 3-deazaneplanocin A (DZNep) [69] and efficiently downregulates EZH2 in AT/RT cells [70]. Ribavirin kills AT/RT tumor cells in part by blocking eIF4E and inducing apoptosis. Ribavirin penetrates the brain, and ribavirin treatment of mice bearing BT12 orthotopic xenografts led to an increase in median survival of 56 compared to 37 days for vehicle-treated mice [70].

Despite increases in EZH2 methyltransferase activity in AT/RT, there are important regions of the genome activated by H3K27 acetylation [38]. The bromodomain and extraterminal (BET) domain family of proteins are required for this H3K27 acetylation. JQ1 is a highly specific inhibitor of BET and BET bromodomain-containing protein 4 (BRD4) [14]. JQ1 is especially effective at targeting MYC driven AT/RT, extending survival in a MYC-driven BT12 orthotopic xenograft [71]. JQ1 and the EZH2 inhibitor GSK126 combined to extend median survival by more than 50% of mice bearing BT12 orthotopic xenografts [72]. These drugs in combination also significantly extended survival in a BT16 orthotopic xenograft model of AT/RT [72]. TEN-010/RO6870810 is a JQ1 analog that was explored in several clinical trials in adult patients (NCT01987362, NCT03068351).

The histone deaceytlase inhibitor (HDAC) panobinostat suppressed the growth of AT/RT cell lines at nanomolar concentrations and induced the differentiation of these cell lines along a neuronal lineage [73]. Combinations of HDAC and mTOR inhibitors drive embryonal cell differentiation in rhabdoid tumors [74]. Treatment of mice bearing BT12 orthotopic xenografts with daily low dose panobinostat for 28 days led to decreased tumor growth as measured by bioluminescent imaging and statistically significant extension of overall survival [73]. Interestingly, when panobinostat treatment stopped after 28 days, the tumors rapidly progressed, underscoring the epigenetic plasticity inherent in AT/RT and other epigenetically driven tumors. These results were concordant with those observed in a prior study in which panobinostat treatment of malignant rhabdoid tumors led to suppression of tumor growth and differentiation along an osteoblastic program [75]. A clinical trial of panobinostat as maintenance therapy in AT/RT recently closed due to drug supply issues (NCT04897880).

The HDAC 1,3, brain-penetrant inhibitor RG2833 also suppressed AT/RT tumor cell growth, promoted apoptosis and upregulated tumor suppressor expression [55]. This inhibitor was originally designed to penetrate the brain for treatment of neuro-degenerative diseases and was well tolerated in humans in early clinical trials [76,77]. RG2833 induced histone de-acetylation in AT/RT orthotopic xenografts and may synergize with other targeted or traditional chemotherapy in AT/RT [58].

Curaxin (CBL0137) is a DNA minor groove binding drug that is related to the anti-malarial drug quinacrine [78]. Curaxin disrupts the FAcilitates Chromatin Transcriiption (FACT) complex and as monotherapy extends the life of mice bearing medulloblastoma orthotopic xenografts [79]. Curaxin also has activity against AT/RT cells in vitro and suppresses the growth of MRT flank xenografts [80]. Quinacrine itself kills AT/RT cells, and in conjunction with the ATP-Binding Cassette transporter inhibitor Elacridar extends the life of mice with bearing BT37 orthotopic xenografts [81]. CBL0137 has improved brain penetration and is less likely to be a substrate for multi-drug resistance (MDR) proteins, making it an improvement on quinacrine [78]. A phase I/II clinical trial of CBL0137 in pediatric patients with solid tumors, including brain tumors, is currently ongoing (NCT04870944).

Other cell cycle inhibitors

In addition to the previously mentioned Aurora Kinase A inhibitor alisertib, drugs that activate p53 are also active in AT/RT preclinical models. Mutations in TP53 are rare in AT/RT, but the TP53 binding protein MDM2 is highly expressed [82]. Treatment with the MDM2 inhibitor idasnutlin killed AT/RT tumor cells and extended survival of mice bearing orthotopic MAF737 xenografts [82]. The DNA-binding drug quinacrine also induces expression of activated p53 in AT/RT, suggesting that activation of p53 may contribute, along with disruption of FACT, to the activity of quinacrine and Curaxin/CBL-0137 [81].

Immunotherapies

AT/RT has the lowest mutational burden in coding genes of any aggressive human malignancy, and other mutations are rare and inconsistent [29], suggesting that there is a lack of suitable neoantigens for immune attack. However, immune profiling of AT/RT primary tumors has shown that they are richly infiltrated with macrophages, CD8+, CD4+ T-cell and NK cells [44]. Tumor resident and exhausted memory T-cells were increased in AT/RT tumors. Perhaps due to their epigenetic dysregulation, human AT/RT tumors express increased levels of endogenous retroviral RNA, leading to increased interferon-regulated gene expression [44]. Treatment of a syngeneic mouse model of MYC-driven AT/RT growing in flanks with PD-1 immune checkpoint inhibitor led to delayed tumor growth and increased tumor rejection on rechallenge [44].

Landscape of upcoming clinical trials for children with AT/RT

Due to the rarity of this disease and still unacceptably high rate of recurrence and death, novel clinical trial designs are needed to investigate novel agents more efficiently and to provide new treatment options to as many children with AT/RT as possible. PNOC is developing a multi-arm clinical trial for patients with relapsed/recurrent/refractory AT/RT where novel agents will be incorporated into the study schema as preclinical data emerge about their efficacy against AT/RT. The trial will have multiple treatment arms based on the agents under investigation with some arms enrolling patients to determine safety/tolerability and recommended dosing, while other arms will interrogate the efficacy of agents with pre-established pediatric dosing. Some arms will be investigating single agents while others will be designed as combination studies. Patients enrolled on the trial will be able to be treated sequentially on different arms of the study based on their response to the different agents. This will allow patients to be treated with a number of different agents/combinations of agents without having to experience prolonged delays between therapies. This type of design also allows investigators to quickly move novel therapies into the clinic without undo delays associated with the development of a new clinical trial. In parallel, PNOC is also creating a comprehensive registry of patients with AT/RT inclusive of complete clinical, radiographic, and molecular data. These data will be used to create an external control cohort that will serve as the comparator cohort for determination of the trial's outcome measures.

Conclusion

While current available therapies utilizing intensive multimodal therapies have led to improvement in outcomes for patients with AT/RT, the risk of recurrence and death remains at about 50%, and survivors carry a high burden of treatment related toxicity, including neurocognitive impairment [83]. As such, continued efforts to improve treatment options for patients with AT/RT in the upfront and relapsed setting is imperative. Bringing novel therapies into prospective clinical trials for AT/RT is needed as part of the efforts to improve outcomes and decrease toxicity. There are numerous novel therapeutics with biologic rationale and supportive pre-clinical data that are either currently being evaluated or soon to be incorporated into clinical trials for AT/RT. As we learn more about the molecular and epigenetic drivers of AT/RT, more data will likely emerge in the future. Given the rarity of AT/RT, as well as the emerging understanding of the molecular heterogeneity of the subgroups and their correlation to response and outcomes, the use of innovative clinical trial designs and international collaborations will be paramount in making further progress in the treatment of AT/RT.

Author contributions

Elizabeth Alva: – writing – original draft, editing, data curation. Jeffrey Rubens: writing – original draft, review and editing, data curation. Susan Chi: review and editing, data curation. Tom Rosenberg: review and editing, data curation. Alyssa Reddy: review and editing, data curation. Eric Raabe: writing – original draft, review and editing, conceptualization, data curation. Ashley Margol: writing – original draft, review and editing, editing, conceptualization, data curation.

Declaration of Competing Interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Footnotes

Funding support: National Cancer Institute Core Grant to the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center (P30CA006973) (ER and JR).

Contributor Information

Eric H. Raabe, Email: eraabe2@jhmi.edu.

Ashley Margol, Email: amargol@chla.usc.edu.

References

  • 1.Hilden JM, Meerbaum S, Burger P, Finlay J, Janss A, Scheithauer BW, Walter AW, Rorke LB, Biegel JA. Central nervous system atypical teratoid/rhabdoid tumor: results of therapy in children enrolled in a registry. J. Clin. Oncol. 2004;22:2877–2884. doi: 10.1200/JCO.2004.07.073. [DOI] [PubMed] [Google Scholar]
  • 2.Woehrer A, Slavc I, Waldhoer T, Heinzl H, Zielonke N, Czech T, Benesch M, Hainfellner JA, Haberler C. Incidence of atypical teratoid/rhabdoid tumors in children: a population-based study by the Austrian Brain Tumor Registry, 1996-2006. Cancer. 2010;116:5725–5732. doi: 10.1002/cncr.25540. [DOI] [PubMed] [Google Scholar]
  • 3.Nesvick CL, Lafay-Cousin L, Raghunathan A, Bouffet E, Huang AA, Daniels DJ. Atypical teratoid rhabdoid tumor: molecular insights and translation to novel therapeutics. J. Neurooncol. 2020;150:47–56. doi: 10.1007/s11060-020-03639-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ostrom QT, de Blank PM, Kruchko C, Petersen CM, Liao P, Finlay JL, Stearns DS, Wolff JE, Wolinsky Y, Letterio JJ, et al. Alex's lemonade stand foundation infant and childhood primary brain and central nervous system tumors diagnosed in the United States in 2007-2011. Neuro. Oncol. 2015;16(Suppl 10):x1–x36. doi: 10.1093/neuonc/nou327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803–820. doi: 10.1007/s00401-016-1545-1. [DOI] [PubMed] [Google Scholar]
  • 6.Rorke LB, Packer RJ, Biegel JA. Central nervous system atypical teratoid/rhabdoid tumors of infancy and childhood: definition of an entity. J. Neurosurg. 1996;85:56–65. doi: 10.3171/jns.1996.85.1.0056. [DOI] [PubMed] [Google Scholar]
  • 7.Hasselblatt M, Isken S, Linge A, Eikmeier K, Jeibmann A, Oyen F, Nagel I, Richter J, Bartelheim K, Kordes U, 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;52:185–190. doi: 10.1002/gcc.22018. [DOI] [PubMed] [Google Scholar]
  • 8.Lee RS, Stewart C, Carter SL, Ambrogio L, Cibulskis K, Sougnez C, Lawrence MS, Auclair D, Mora J, Golub TR, et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J. Clin. Invest. 2012;122:2983–2988. doi: 10.1172/JCI64400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Biegel JA, Zhou JY, Rorke LB, Stenstrom C, Wainwright LM, Fogelgren B. Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res. 1999;59:74–79. [PubMed] [Google Scholar]
  • 10.Saunders J, Ingley K, Wang XQ, Harvey M, Armstrong L, Ng T, Dunham C, Bush J. Loss of BRG1 (SMARCA4) Immunoexpression in a pediatric non-central nervous system tumor cohort. Pediatr. Dev. Pathol. 2020;23:132–138. doi: 10.1177/1093526619869154. [DOI] [PubMed] [Google Scholar]
  • 11.Haberler C, Laggner U, Slavc I, Czech T, Ambros IM, Ambros PF, Budka H, Hainfellner JA. Immunohistochemical analysis of INI1 protein in malignant pediatric CNS tumors: Lack of INI1 in atypical teratoid/rhabdoid tumors and in a fraction of primitive neuroectodermal tumors without rhabdoid phenotype. Am. J. Surg. Pathol. 2006;30:1462–1468. doi: 10.1097/01.pas.0000213329.71745.ef. [DOI] [PubMed] [Google Scholar]
  • 12.Nemes K, Bens S, Bourdeaut F, Johann P, Kordes U, Siebert R, Frühwald MC (1993). Rhabdoid Tumor Predisposition Syndrome. Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW and Amemiya A (eds). University of Washington, Seattle [PubMed]
  • 13.Torchia J, Picard D, Lafay-Cousin L, Hawkins CE, Kim SK, Letourneau L, Ra YS, Ho KC, Chan TS, Sin-Chan P, et al. Molecular subgroups of atypical teratoid rhabdoid tumours in children: an integrated genomic and clinicopathological analysis. Lancet Oncol. 2015;16:569–582. doi: 10.1016/S1470-2045(15)70114-2. [DOI] [PubMed] [Google Scholar]
  • 14.Johann PD, Erkek S, Zapatka M, Kerl K, Buchhalter I, Hovestadt V, Jones DTW, Sturm D, Hermann C, Segura Wang M, et al. Atypical teratoid/rhabdoid tumors are comprised of three epigenetic subgroups with distinct enhancer landscapes. Cancer Cell. 2016;29:379–393. doi: 10.1016/j.ccell.2016.02.001. [DOI] [PubMed] [Google Scholar]
  • 15.Ho B, Johann PD, Grabovska Y, De Dieu Andrianteranagna MJ, Yao F, Frühwald M, Hasselblatt M, Bourdeaut F, Williamson D, Huang A, et al. Molecular subgrouping of atypical teratoid/rhabdoid tumors—a reinvestigation and current consensus. Neuro-Oncology. 2019;22:613–624. doi: 10.1093/neuonc/noz235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chi SN, Zimmerman MA, Yao X, Cohen KJ, Burger P, Biegel JA, Rorke-Adams LB, Fisher MJ, Janss A, Mazewski C, et al. (2009). Intensive multimodality treatment for children with newly diagnosed CNS atypical teratoid rhabdoid tumor J. Clin. Oncol. 27, 385-389. [DOI] [PMC free article] [PubMed]
  • 17.Reddy AT, Strother DR, Judkins AR, Burger PC, Pollack IF, Krailo MD, Buxton AB, Williams-Hughes C, Fouladi M, Mahajan A, 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;38:1175–1185. doi: 10.1200/JCO.19.01776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fruhwald MC, Hasselblatt M, Nemes K, Bens S, Steinbugl M, Johann PD, Kerl K, Hauser P, Quiroga E, Solano-Paez P, 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;22:1006–1017. doi: 10.1093/neuonc/noz244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zaky W, Dhall G, Ji L, Haley K, Allen J, Atlas M, Bertolone S, Cornelius A, Gardner S, Patel R, et al. Intensive induction chemotherapy followed by myeloablative chemotherapy with autologous hematopoietic progenitor cell rescue for young children newly-diagnosed with central nervous system atypical teratoid/rhabdoid tumors: the Head Start III experience. Pediatr. Blood. Cancer. 2014;61:95–101. doi: 10.1002/pbc.24648. [DOI] [PubMed] [Google Scholar]
  • 20.Mulhern RK, Merchant TE, Gajjar A, Reddick WE, Kun LE. Late neurocognitive sequelae in survivors of brain tumours in childhood. Lancet Oncol. 2004;5:399–408. doi: 10.1016/S1470-2045(04)01507-4. [DOI] [PubMed] [Google Scholar]
  • 21.Fouladi M, Gilger E, Kocak M, Wallace D, Buchanan G, Reeves C, Robbins N, Merchant T, Kun LE, Khan R, et al. Intellectual and functional outcome of children 3 years old or younger who have CNS malignancies. J. Clin. Oncol. 2005;23:7152–7160. doi: 10.1200/JCO.2005.01.214. [DOI] [PubMed] [Google Scholar]
  • 22.Geyer JR, Sposto R, Jennings M, Boyett JM, Axtell RA, Breiger D, Broxson E, Donahue B, Finlay JL, Goldwein JW, et al. Multiagent chemotherapy and deferred radiotherapy in infants with malignant brain tumors: a report from the Children's Cancer Group. J. Clin. Oncol. 2005;23:7621–7631. doi: 10.1200/JCO.2005.09.095. [DOI] [PubMed] [Google Scholar]
  • 23.Gardner SL, Asgharzadeh S, Green A, Horn B, McCowage G, Finlay J. Intensive induction chemotherapy followed by high dose chemotherapy with autologous hematopoietic progenitor cell rescue in young children newly diagnosed with central nervous system atypical teratoid rhabdoid tumors. Pediatr. Blood. Cancer. 2008;51:235–240. doi: 10.1002/pbc.21578. [DOI] [PubMed] [Google Scholar]
  • 24.Bartelheim K, Nemes K, Seeringer A, Kerl K, Buechner J, Boos J, Graf N, Durken M, Gerss J, Hasselblatt M, et al. Improved 6-year overall survival in AT/RT - results of the registry study Rhabdoid 2007. Cancer Med. 2016;5:1765–1775. doi: 10.1002/cam4.741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Reddy A, Biegel J, Huang A, Strother D, Judkins A, Pollack I, Buxton A, Mahajan A, Ho B, Mazewski C, et al. Correlation of clinicopathologic features and cumulative incidence of relapse for patients with atypical teratoid rhabdoid tumor on ACNS0333: a report from the children's oncology group. Neuro. Oncol. 2021;23:i1. [Google Scholar]
  • 26.Upadhyaya SA, Robinson GW, Onar-Thomas A, Orr BA, Johann P, Wu G, Billups CA, Tatevossian RG, Dhanda SK, Srinivasan 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;27:2879–2889. doi: 10.1158/1078-0432.CCR-20-4731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chi SN, Zimmerman MA, Yao X, Cohen KJ, Burger P, Biegel JA, Rorke-Adams LB, Fisher MJ, Janss A, Mazewski C, et al. Intensive multimodality treatment for children with newly diagnosed CNS atypical teratoid rhabdoid tumor. J. Clin. Oncol. 2009;27:385–389. doi: 10.1200/JCO.2008.18.7724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hoffman LM, Richardson EA, Ho B, Margol A, Reddy A, Lafay-Cousin L, Chi S, Slavc I, Judkins A, Hasselblatt M, et al. Advancing biology-based therapeutic approaches for atypical teratoid rhabdoid tumors. Neuro. Oncol. 2020;22:944–954. doi: 10.1093/neuonc/noaa046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Torchia J, Golbourn B, Feng S, Ho KC, Sin-Chan P, Vasiljevic A, Norman JD, Guilhamon P, Garzia L, Agamez NR, et al. Integrated (epi)-genomic analyses identify subgroup-specific therapeutic targets in CNS rhabdoid tumors. Cancer Cell. 2016;30:891–908. doi: 10.1016/j.ccell.2016.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ho B, Johann PD, Grabovska Y, De Dieu Andrianteranagna MJ, Yao F, Fruhwald M, Hasselblatt M, Bourdeaut F, Williamson D, Huang A, et al. Molecular subgrouping of atypical teratoid/rhabdoid tumors-a reinvestigation and current consensus. Neuro. Oncol. 2020;22:613–624. doi: 10.1093/neuonc/noz235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang SZ, Poore B, Alt J, Price A, Allen SJ, Hanaford AR, Kaur H, Orr BA, Slusher BS, Eberhart CG, et al. Unbiased metabolic profiling predicts sensitivity of high myc-expressing atypical Teratoid/Rhabdoid tumors to glutamine inhibition with 6-Diazo-5-Oxo-L-Norleucine. Clin. Cancer Res. 2019;25:5925–5936. doi: 10.1158/1078-0432.CCR-19-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Reddy AT, Krailo MD, Buxton AB, Strother DR, Huang A, Zhou T, Judkins AR, Burger PC, Pollack IF, Williams-Hughes C, et al. (2020). Reply to S.A. Upadhyaya J. Clin. Oncol. 38, 3353-3354. [DOI] [PMC free article] [PubMed]
  • 33.Margol A, Hoffman L, Tran J, Malvar J, Chi Y, Leary S, Stevens J, Faulk K, Wu C, Salloum R, et al. Radiation therapy is an important component of multimodal therapy for children with atypical teratoid rhabdoid tumors. Pediatr. Blood. Cancer. 2022;69(Suppl 5):e29952. [Google Scholar]
  • 34.Fouladi M, Blaney SM, Poussaint TY, Freeman BB, 3rd, McLendon R, Fuller C, Adesina AM, Hancock ML, Danks MK, Stewart C, et al. Phase II study of oxaliplatin in children with recurrent or refractory medulloblastoma, supratentorial primitive neuroectodermal tumors, and atypical teratoid rhabdoid tumors: a pediatric brain tumor consortium study. Cancer. 2006;107:2291–2297. doi: 10.1002/cncr.22241. [DOI] [PubMed] [Google Scholar]
  • 35.Geoerger B, Bourdeaut F, DuBois SG, Fischer M, Geller JI, Gottardo NG, Marabelle A, Pearson ADJ, Modak S, Cash T, 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;23:2433–2441. doi: 10.1158/1078-0432.CCR-16-2898. [DOI] [PubMed] [Google Scholar]
  • 36.Mossé YP, Fox E, Teachey DT, Reid JM, Safgren SL, Carol H, Lock RB, Houghton PJ, Smith MA, Hall D, et al. A phase II study of alisertib in children with recurrent/refractory solid tumors or leukemia: children's oncology group phase I and pilot consortium (ADVL0921) Clin. Cancer Res. 2019;25:3229–3238. doi: 10.1158/1078-0432.CCR-18-2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Upadhyaya S, Campagne O, Robinson GW, Onar-Thomas A, Orr B, Billups CA, Tatevossian RG, Broniscer A, Kilburn LB, Baxter PA, et al. Phase II study of alisertib as a single agent in recurrent or progressive atypical teratoid rhabdoid tumors. J. Clinic. Oncol. 2020;38:10542. [Google Scholar]
  • 38.Erkek S, Johann PD, Finetti MA, Drosos Y, Chou HC, Zapatka M, Sturm D, Jones DTW, Korshunov A, Rhyzova M, 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 doi: 10.1016/j.ccell.2018.11.014. 95-110 e118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wilson BG, Wang X, Shen X, McKenna ES, Lemieux ME, Cho YJ, Koellhoffer EC, Pomeroy SL, Orkin SH, Roberts CW. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell. 2010;18:316–328. doi: 10.1016/j.ccr.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chi SN, Bourdeaut F, Casanova M, Kilburn LB, Hargrave DR, McCowage GB, Pinto NR, Yang J, Chadha R, Kahali B, et al. Update on phase 1 study of tazemetostat, an enhancer of zeste homolog 2 inhibitor, in pediatric patients with relapsed or refractory integrase interactor 1–negative tumors. J. Clinic. Oncol. 2022;40:10040. [Google Scholar]
  • 41.Chi SN, Bourdeaut F, Laetsch TW, Fouladi M, Macy ME, Makin GW, Shukla NN, Wetmore C, Margol AS, Casanova M, 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. Clinic. Oncol. 2020;38:10525. [Google Scholar]
  • 42.Chi SN, Yi JS, Williams PM, Roy-Chowdhuri S, Patton DR, Coffey B, Reid JM, Piao J, Saguilig L, Alonzo TA, et al. Tazemetostat in patients with tumors with alterations in EZH2 or the SWI/SNF complex: results from NCI-COG Pediatric MATCH trial Arm C (APEC1621C) J. Clinic. Oncol. 2022;40:10009. [Google Scholar]
  • 43.Studebaker AW, Hutzen B, Pierson CR, Shaffer TA, Raffel C, Jackson EM. Oncolytic measles virus efficacy in murine xenograft models of atypical teratoid rhabdoid tumors. Neuro. Oncol. 2015;17:1568–1577. doi: 10.1093/neuonc/nov058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Leruste A, Tosello J, Ramos RN, Tauziede-Espariat A, Brohard S, Han ZY, Beccaria K, Andrianteranagna M, Caudana P, Nikolic J, et al. (2019). Clonally Expanded T Cells Reveal Immunogenicity of Rhabdoid Tumors Cancer Cell 36, 597-612 e598. [DOI] [PubMed]
  • 45.Maachani UB, Tosi U, Pisapia DJ, Mukherjee S, Marnell CS, Voronina J, Martinez D, Santi M, Dahmane N, Zhou Z, et al. (2020). B7-H3 as a Prognostic Biomarker and Therapeutic Target in Pediatric central nervous system Tumors Transl Oncol 13, 365-371. [DOI] [PMC free article] [PubMed]
  • 46.Seaman S, Zhu Z, Saha S, Zhang XM, Yang MY, Hilton MB, Morris K, Szot C, Morris H, Swing DA, et al. Eradication of tumors through simultaneous ablation of CD276/B7-H3-positive tumor cells and tumor vasculature. Cancer Cell. 2017;31 doi: 10.1016/j.ccell.2017.03.005. 501-515 e508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kendsersky NM, Lindsay J, Kolb EA, Smith MA, Teicher BA, Erickson SW, Earley EJ, Mosse YP, Martinez D, Pogoriler J, et al. The B7-H3-targeting antibody-drug conjugate m276-SL-PBD is potently effective against pediatric cancer preclinical solid tumor models. Clin. Cancer Res. 2021;27:2938–2946. doi: 10.1158/1078-0432.CCR-20-4221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Theruvath J, Sotillo E, Mount CW, Graef CM, Delaidelli A, Heitzeneder S, Labanieh L, Dhingra S, Leruste A, Majzner RG, et al. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors. Nat. Med. 2020;26:712–719. doi: 10.1038/s41591-020-0821-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hanaford AR, Archer TC, Price A, Kahlert UD, Maciaczyk J, Nikkhah G, Kim JW, Ehrenberger T, Clemons PA, Dancik V, et al. Discovering Innovative therapies for rare tumors: combining genetically accurate disease models with in silico analysis to identify novel therapeutic targets. Clin. Cancer Res. 2016;22:3903–3914. doi: 10.1158/1078-0432.CCR-15-3011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pham K, Hanaford AR, Poore BA, Maxwell MJ, Sweeney H, Parthasarathy A, Alt J, Rais R, Slusher BS, Eberhart CG, et al. Comprehensive metabolic profiling of MYC-amplified medulloblastoma tumors reveals key dependencies on amino acid, tricarboxylic acid and hexosamine pathways. Cancers (Basel) 2022:14. doi: 10.3390/cancers14051311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rubens JA, Wang SZ, Price A, Weingart MF, Allen SJ, Orr BA, Eberhart CG, Raabe EH. The TORC1/2 inhibitor TAK228 sensitizes atypical teratoid rhabdoid tumors to cisplatin-induced cytotoxicity. Neuro. Oncol. 2017;19:1361–1371. doi: 10.1093/neuonc/nox067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Singh A, Lun X, Jayanthan A, Obaid H, Ruan Y, Strother D, Chi SN, Smith A, Forsyth P, Narendran A. Profiling pathway-specific novel therapeutics in preclinical assessment for central nervous system atypical teratoid rhabdoid tumors (CNS ATRT): favorable activity of targeting EGFR- ErbB2 signaling with lapatinib. Mol. Oncol. 2013;7:497–512. doi: 10.1016/j.molonc.2013.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jozwiak J, Bikowska B, Grajkowska W, Sontowska I, Roszkowski M, Galus R. Activation of Akt/mTOR pathway in a patient with atypical teratoid/rhabdoid tumor. Folia Neuropathol. 2010;48:185–189. [PubMed] [Google Scholar]
  • 54.Parkhurst A, Wang SZ, Findlay TR, Malebranche KJ, Odabas A, Alt J, Maxwell MJ, Kaur H, Peer CJ, Figg WD, et al. Dual mTORC1/2 inhibition compromises cell defenses against exogenous stress potentiating Obatoclax-induced cytotoxicity in atypical teratoid/rhabdoid tumors. Cell Death. Dis. 2022;13:410. doi: 10.1038/s41419-022-04868-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Findlay T., Malebranche K., Raabe E., Eberhart C., Rubens J. The PI3k inhibitor Paxalisib combines with the novel HDAC1/3 inhibitor RG2833 to improve survival in mice bearing orthotopic xenografts of atypical teratoid/rhabdoid tumors. Neuro. Oncol. 2022;24 [Google Scholar]
  • 56.Kolb EA, Gorlick R, Houghton PJ, Morton CL, Lock RB, Tajbakhsh M, Reynolds CP, Maris JM, Keir ST, Billups CA, et al. Initial testing of dasatinib by the pediatric preclinical testing program. Pediatr. Blood. Cancer. 2008;50:1198–1206. doi: 10.1002/pbc.21368. [DOI] [PubMed] [Google Scholar]
  • 57.Meel MH, Guillen Navarro M, de Gooijer MC, Metselaar DS, Waranecki P, Breur M, Lagerweij T, Wedekind LE, Koster J, van de Wetering MD, et al. MEK/MELK inhibition and blood-brain barrier deficiencies in atypical teratoid/rhabdoid tumors. Neuro. Oncol. 2020;22:58–69. doi: 10.1093/neuonc/noz151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Weingart MF, Roth JJ, Hutt-Cabezas M, Busse TM, Kaur H, Price A, Maynard R, Rubens J, Taylor I, Mao XG, et al. Disrupting LIN28 in atypical teratoid rhabdoid tumors reveals the importance of the mitogen activated protein kinase pathway as a therapeutic target. Oncotarget. 2015;6:3165–3177. doi: 10.18632/oncotarget.3078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Shahab S, Rubens J, Kaur H, Sweeney H, Eberhart CG, Raabe EH. MEK inhibition suppresses growth of atypical teratoid/rhabdoid tumors. J. Neuropathol. Exp. Neurol. 2020;79:746–753. doi: 10.1093/jnen/nlaa042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pham K, Maxwell MJ, Sweeney H, Alt J, Rais R, Eberhart CG, Slusher BS, Raabe EH. Novel glutamine antagonist JHU395 suppresses MYC-driven medulloblastoma growth and induces apoptosis. J. Neuropathol. Exp. Neurol. 2021;80:336–344. doi: 10.1093/jnen/nlab018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hanaford AR, Alt J, Rais R, Wang SZ, Kaur H, Thorek DLJ, Eberhart CG, Slusher BS, Martin AM, Raabe EH. Orally bioavailable glutamine antagonist prodrug JHU-083 penetrates mouse brain and suppresses the growth of MYC-driven medulloblastoma. Transl. Oncol. 2019;12:1314–1322. doi: 10.1016/j.tranon.2019.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rais R, Lemberg KM, Tenora L, Arwood ML, Pal A, Alt J, Wu Y, Lam J, Aguilar JMH, Zhao L, et al. Discovery of DRP-104, a tumor-targeted metabolic inhibitor prodrug. Sci. Adv. 2022;8:eabq5925. doi: 10.1126/sciadv.abq5925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Carugo A, Minelli R, Sapio L, Soeung M, Carbone F, Robinson FS, Tepper J, Chen Z, Lovisa S, Svelto M, et al. p53 Is a master regulator of proteostasis in SMARCB1-deficient malignant Rhabdoid tumors. Cancer Cell. 2019;35 doi: 10.1016/j.ccell.2019.01.006. 204-220 e209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tran HM, Wu KS, Sung SY, Changou CA, Hsieh TH, Liu YR, Liu YL, Tsai ML, Lee HL, Hsieh KL, et al. Upregulation of protein synthesis and proteasome degradation confers sensitivity to proteasome inhibitor bortezomib in myc-atypical teratoid/rhabdoid tumors. Cancers (Basel) 2020:12. doi: 10.3390/cancers12030752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Morin A, Soane C, Pierce A, Sanford B, Jones KL, Crespo M, Zahedi S, Vibhakar R, Mulcahy Levy JM. Proteasome inhibition as a therapeutic approach in atypical teratoid/rhabdoid tumors. Neurooncol. Adv. 2020;2:vdaa051. doi: 10.1093/noajnl/vdaa051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cook Sangar ML, Genovesi LA, Nakamoto MW, Davis MJ, Knobluagh SE, Ji P, Millar A, Wainwright BJ, Olson JM. Inhibition of CDK4/6 by palbociclib significantly extends survival in medulloblastoma patient-derived xenograft mouse models. Clin. Cancer Res. 2017;23:5802–5813. doi: 10.1158/1078-0432.CCR-16-2943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hashizume R, Zhang A, Mueller S, Prados MD, Lulla RR, Goldman S, Saratsis AM, Mazar AP, Stegh AH, Cheng SY, et al. Inhibition of DNA damage repair by the CDK4/6 inhibitor palbociclib delays irradiated intracranial atypical teratoid rhabdoid tumor and glioblastoma xenograft regrowth. Neuro. Oncol. 2016;18:1519–1528. doi: 10.1093/neuonc/now106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Merk DJ, Hirsch S, Tsiami F, Walter B, Haeusser LA, Babaei S, Admar J, Casadei N, Roggia C, Spohn M, et al. (2020). Genome-wide CRISPR and small-molecule screens uncover targetable dependencies in ATRT bioRxiv, 2020.2012.2009.417378.
  • 69.De la Cruz-Hernandez E, Medina-Franco JL, Trujillo J, Chavez-Blanco A, Dominguez-Gomez G, Perez-Cardenas E, Gonzalez-Fierro A, Taja-Chayeb L, Duenas-Gonzalez A. Ribavirin as a tri-targeted antitumor repositioned drug. Oncol. Rep. 2015;33:2384–2392. doi: 10.3892/or.2015.3816. [DOI] [PubMed] [Google Scholar]
  • 70.Casaos J, Huq S, Lott T, Felder R, Choi J, Gorelick N, Peters M, Xia Y, Maxwell R, Zhao T, et al. Ribavirin as a potential therapeutic for atypical teratoid/rhabdoid tumors. Oncotarget. 2018;9:8054–8067. doi: 10.18632/oncotarget.23883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Alimova I, Pierce A, Danis E, Donson A, Birks DK, Griesinger A, Foreman NK, Santi M, Soucek L, Venkataraman S, et al. Inhibition of MYC attenuates tumor cell self-renewal and promotes senescence in SMARCB1-deficient Group 2 atypical teratoid rhabdoid tumors to suppress tumor growth in vivo. Int. J. Cancer. 2019;144:1983–1995. doi: 10.1002/ijc.31873. [DOI] [PubMed] [Google Scholar]
  • 72.Ishi Y, Zhang Y, Zhang A, Sasaki T, Piunti A, Suri A, Watanabe J, Abe K, He X, Katagi H, et al. Therapeutic targeting of EZH2 and BET BRD4 in pediatric rhabdoid tumors. Mol. Cancer Ther. 2022;21:715–726. doi: 10.1158/1535-7163.MCT-21-0646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chong WC, Jayasekara WSN, Vaghjiani VG, Parackal S, Sun C, Popovski D, Algar EM, Firestein R, Wood PJ, Khan S, et al. Atypical teratoid rhabdoid tumours are susceptible to panobinostat-mediated differentiation therapy. Cancers (Basel) 2021:13. doi: 10.3390/cancers13205145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Custers L, Khabirova E, Coorens THH, Oliver TRW, Calandrini C, Young MD, Vieira Braga FA, Ellis P, Mamanova L, Segers H, et al. Somatic mutations and single-cell transcriptomes reveal the root of malignant rhabdoid tumours. Nat. Commun. 2021;12:1407. doi: 10.1038/s41467-021-21675-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Muscat A, Popovski D, Jayasekara WS, Rossello FJ, Ferguson M, Marini KD, Alamgeer M, Algar EM, Downie P, Watkins DN, et al. Low-Dose histone deacetylase inhibitor treatment leads to tumor growth arrest and multi-lineage differentiation of malignant Rhabdoid tumors. Clin. Cancer Res. 2016;22:3560–3570. doi: 10.1158/1078-0432.CCR-15-2260. [DOI] [PubMed] [Google Scholar]
  • 76.Soragni E, Miao W, Iudicello M, Jacoby D, De Mercanti S, Clerico M, Longo F, Piga A, Ku S, Campau E, et al. Epigenetic therapy for Friedreich ataxia. Ann. Neurol. 2014;76:489–508. doi: 10.1002/ana.24260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Soragni E, Chou CJ, Rusche JR, Gottesfeld JM (2015). Mechanism of Action of 2-Aminobenzamide HDAC Inhibitors in Reversing Gene Silencing in Friedreich's Ataxia Front Neurol 6, 44. [DOI] [PMC free article] [PubMed]
  • 78.Gasparian AV, Burkhart CA, Purmal AA, Brodsky L, Pal M, Saranadasa M, Bosykh DA, Commane M, Guryanova OA, Pal S, et al. Curaxins: anticancer compounds that simultaneously suppress NF-kappaB and activate p53 by targeting FACT. Sci. Transl. Med. 2011;3:95ra74. doi: 10.1126/scitranslmed.3002530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wang J, Sui Y, Li Q, Zhao Y, Dong X, Yang J, Liang Z, Han Y, Tang Y, Ma J. Effective inhibition of MYC-amplified group 3 medulloblastoma by FACT-targeted curaxin drug CBL0137. Cell Death. Dis. 2020;11:1029. doi: 10.1038/s41419-020-03201-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lock R, Carol H, Maris JM, Kolb EA, Gorlick R, Reynolds CP, Kang MH, Keir ST, Wu J, Purmal A, et al. (2017). Initial testing (stage 1) of the curaxin CBL0137 by the pediatric preclinical testing program Pediatr. Blood. Cancer 64. [DOI] [PMC free article] [PubMed]
  • 81.Kaur H, Parthasarathy A, Guo H, Green PC, Akhtarkhavari S, Eberhart CG, Raabe EH (2021). Repurposed anti-malarial Quinacrine activates P53 and inhibits ATRT tumorigenicity Neuro. Oncol. 23, i1-i2.
  • 82.Alimova I, Wang D, Danis E, Pierce A, Donson A, Serkova N, Madhavan K, Lakshmanachetty S, Balakrishnan I, Foreman NK, et al. Targeting the TP53/MDM2 axis enhances radiation sensitivity in atypical teratoid rhabdoid tumors. Int. J. Oncol. 2022;60 doi: 10.3892/ijo.2022.5322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lafay-Cousin L, Fay-McClymont T, Johnston D, Fryer C, Scheinemann K, Fleming A, Hukin J, Janzen L, Guger S, Strother D, et al. Neurocognitive evaluation of long term survivors of atypical teratoid rhabdoid tumors (ATRT): the Canadian registry experience. Pediatr. Blood. Cancer. 2015;62:1265–1269. doi: 10.1002/pbc.25441. [DOI] [PubMed] [Google Scholar]

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES