Paediatrics (RCH) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 38
  • Item
    Thumbnail Image
    ETMR-06. Molecular and clinical characteristics of CNS tumors withBCOR(L1) fusion/internal tandem duplication
    Gojo, J ; Schmitt-Hoffner, F ; Mauermann, M ; von Hoff, K ; Sill, M ; Korshunov, A ; Stichel, D ; Capper, D ; Tauziede-Espariat, A ; Varlet, P ; Aldape, K ; Abdullaev, Z ; Donson, A ; Pahnke, J ; Schüller, U ; Tran, I ; Galbraith, K ; Snuderl, M ; Alexandrescu, S ; Brandner, S ; Łastowska, M ; Miele, E ; Lugt, JV ; Meijer, L ; Bunt, J ; Kramm, C ; Hansford, JR ; Krskova, L ; Zapotocky, M ; Nobusawa, S ; Solomon, D ; Haberler, C ; Jones, B ; Sturm, D ; Sahm, F ; Jäger, N ; Pfister, SM ; Kool, M (Oxford University Press (OUP), 2022-06-03)
    Abstract Central nervous system (CNS) tumor with BCOR internal tandem duplication (BCOR-ITD) have recently been introduced in the 5th edition of the WHO classification of CNS tumors, however, their molecular makeup and clinical characteristics remain widely enigmatic. This is further complicated by the recent discovery of tumors characterized by gene fusions involving BCOR or its homologue BCORL1. We identified a cohort of 206 BCOR altered CNS tumors via DNA methylation profiling and conducted in-depth molecular and clinical characterization in an international effort. By performing t-SNE clustering analysis we found that BCOR-fusion tumors form a distinct cluster (n=61), adjacent to BCOR-ITD cases (n=145). The identified fusion partners of BCOR(L1) included EP300 (n=20), CREBBP (n=5), and NUTM2HP (n=1). Notably, three cases within the BCOR-ITD cluster harbored a c-terminal intragenic deletion within BCOR. With respect to clinical characteristics gender ratio was balanced in BCOR-fusion cases (m/f, 1.1), whereas predominance of male patients was observed in the BCOR-ITD group (m/f, 1.5). Moreover, age at diagnosis of BCOR-fusion patients was higher as compared to BCOR-ITD cases (15 vs 4.5 years). Interestingly, BCOR-fusion tumors were exclusively found in the supratentorial region being originally diagnosed as ependymomas or gliomas whereas BCOR-ITD emerged across the entire CNS with diverse original diagnoses. 8% of BCOR-ITD and none of BCOR-fusion cases were disseminated at diagnosis. In line with this observation, 40% of first relapses within the BCOR-ITD group were metastatic which was less frequent in BCOR-fusion tumors. Survival estimates demonstrated no differences, generally showing short median PFS (BCOR-fusion, 2 years, n=15; BCOR-ITD, 1.8 years, n=55) and intermediate OS rates (BCOR-fusion, 6.8 years, n=18; BCOR-ITD 6.3 years, n=60). Further molecular and clinical characterization is ongoing potentially revealing first therapeutic leads for these highly aggressive CNS tumor types.
  • Item
    Thumbnail Image
    HGG-11. Clinical characteristics and clinical evolution of a large cohort of pediatric patients with primary central nervous system (CNS) tumors and tropomyosin receptor kinase (TRK) fusion.
    Lamoureux, A-A ; Fisher, M ; Lemelle, L ; Pfaff, E ; Kramm, C ; De Wilde, B ; Kazanowska, B ; Hutter, C ; Pfister, SM ; Sturm, D ; Jones, D ; Orbach, D ; Pierron, G ; Raskin, S ; Drilon, A ; Diamond, E ; Harada, G ; Zapotocky, M ; Ellezam, B ; Weil, AG ; Venne, D ; Barritault, M ; Leblond, P ; Coltin, H ; Hammad, R ; Tabori, U ; Hawkins, C ; Hansford, JR ; Meyran, D ; Erker, C ; McFadden, K ; Sato, M ; Gottardo, NG ; Dholaria, H ; Nørøxe, DS ; Goto, H ; Ziegler, DS ; Lin, FY ; Parsons, DW ; Lindsay, H ; Wong, T-T ; Liu, Y-L ; Wu, K-S ; Franson, AF ; Hwang, E ; Aguilar-Bonilla, A ; Cheng, S ; Cacciotti, C ; Massimino, M ; Schiavello, E ; Wood, P ; Hoffman, LM ; Cappellano, A ; Lassaletta, A ; Van Damme, A ; Llort, A ; Gerber, NU ; Ceruso, MS ; Bendel, AE ; Skrypek, M ; Hamideh, D ; Mushtaq, N ; Walter, A ; Jabado, N ; Alsahlawi, A ; Farmer, J-P ; Abadi, CC ; Mueller, S ; Mazewski, C ; Aguilera, D ; Robison, N ; O’Halloran, K ; Abbou, S ; Berlanga, P ; Geoerger, B ; Øra, I ; Moertel, CL ; Razis, ED ; Vernadou, A ; Doz, F ; Laetsch, TW ; Perreault, S (Oxford University Press (OUP), 2022-06-03)
    Abstract BACKGROUND: TRK fusions are detected in less than 3% of CNS tumors. Given their rarity, there are limited data on the clinical course of these patients. METHODS: We contacted 166 oncology centers worldwide to retrieve data on patients with TRK fusion-driven CNS tumors. Data extracted included demographics, histopathology, NTRK gene fusion, treatment modalities and outcomes. Patients less than 18 years of age at diagnosis were included in this analysis. RESULTS: Seventy-three pediatric patients with TRK fusion-driven primary CNS tumors were identified. Median age at diagnosis was 2.4 years (range 0.0–17.8) and 60.2 % were male. NTRK2 gene fusions were found in 37 patients (50.7%), NTRK1 and NTRK3 aberrations were detected in 19 (26.0%) and 17 (23.3%), respectively. Tumor types included 38 high-grade gliomas (HGG; 52.1%), 20 low-grade gliomas (LGG; 27.4%), 4 embryonal tumors (5.5%) and 11 others (15.1%). Median follow-up was 46.5 months (range 3-226). During the course of their disease, a total of 62 (84.9%) patients underwent surgery with a treatment intent, 50 (68.5%) patients received chemotherapy, 35 (47.9%) patients received radiation therapy, while 34 (46.6%) patients received NTRK inhibitors (3 as first line treatment). Twenty-four (32.9%) had no progression including 9 LGG (45%) and 9 HGG (23.6%). At last follow-up, only one (5.6%-18 evaluable) patient with LGG died compared to 11 with HGG (35.5%-31 evaluable). For LGG the median progression-free survival (PFS) after the first line of treatment was 17 months (95% CI: 0.0-35.5) and median overall survival (OS) was not reached. For patients with HGG the median PFS was 30 months (95% CI: 11.9-48.1) and median OS was 182 months (95% CI 20.2-343.8). CONCLUSIONS: We report the largest cohort of pediatric patients with TRK fusion-driven primary CNS tumors. These results will help us to better understand clinical evolution and compare outcomes with ongoing clinical trials.
  • Item
    Thumbnail Image
    LGG-58. Understanding the transcriptional heterogeneity of pediatric low-grade gliomas and its implication for tumor pathophysiology
    Boisvert, M ; Perera, AA ; Condurat, AL ; Jeang, J ; Tsai, JW ; Novikov, D ; Zhou, K ; Chacon, M ; DiGiacomo, J ; Kumbhani, R ; Wang, D ; Taylor, MD ; Hansford, JR ; Ludlow, L ; Jabado, N ; Ligon, KL ; Beroukhim, R ; Bandopadhayay, P ; Jones, DTW (Oxford University Press (OUP), 2022-06-03)
    Abstract Pediatric low-grade gliomas (pLGGs) are the most frequent brain tumors in children and comprise a heterogeneous group of tumors with different locations, histologic subtypes, ages at presentation, and clinical behavior. Tumors frequently respond to treatment with chemotherapy or surgical removal, but they can regrow after a period of quiescence, requiring further therapy. Thus, a deeper understanding of the molecular processes involved in these tumors is required to develop therapeutic strategies that are effective against their disease mechanisms. To better understand the cellular behaviors of this heterogenous group of tumors, we have employed single-cell and single-nuclei RNA sequencing technologies to analyze a large-scale dataset (>250,000 cells) of pLGGs. Analysis of this data identified a heterogenous population of cell types and cell states, detecting mature and progenitor-like astrocytes and oligodendrocytes, as well as cells exhibiting senescence or cycling programs. Moreover, we identify a significant immune infiltrate, comprised primarily of microglia. In addition to heterogeneity within pLGG tumors, heterogeneity between LGG subtypes represents another layer that stratifies pLGG biology. We performed a compositional analysis of the cell types present in these tumors and compared transcription signatures and gene expression programs across shared cellular populations of histologically and genetically distinct pLGGs. Finally, we optimized our integration and batch correction analyses by using external 293T cells as spike in controls during our single-cell and single-nuclei data generation steps to determine the most suitable method for batch-effect removal. Our analysis of human pLGGs at the single-cell and single-nuclei resolution provides critical insight into the heterogenous biological activities that constitute these tumors.
  • Item
    Thumbnail Image
    MEDB-49. Relapsed SHH medulloblastomas in young children. Are there alternatives to full-dose craniospinal irradiation?
    Erker, C ; Craig, B ; Bailey, S ; Massimino, M ; Larouche, V ; L Finlay, J ; Kline, C ; Michaiel, G ; Margol, A ; Cohen, K ; Cacciotti, C ; Harrods, V ; Doris, K ; AbdelBaki, M ; Amayiri, N ; Wang, Z ; Hansford, J ; Hukin, J ; Salloum, R ; Hoffman, L ; Muray, J ; Ginn, K ; Zapotocky, Z ; Baroni, L ; Ramaswamy, V ; Gilheens, S ; Aguiera, D ; Mazewski, C ; Shah, S ; Strother, D ; Muller, S ; Gajjar, A ; Northcott, P ; Clifford, S ; Robinson, G ; Bouffet, E ; Lafay-Cousin, L (Oxford University Press (OUP), 2022-06-03)
    Abstract BACKGROUND/RATIONAL: Following initial irradiation sparing therapy, many young children with relapsed medulloblastoma can be salvaged with craniospinal irradiation (CSI). However, the interval to relapse is short and neurocognitive sequelae remain a major concern. The contribution of molecular subgrouping may help refine indications and modalities of salvage strategies in this population. METHOD: From a cohort of 151 young children with molecularly characterized relapsed medulloblastoma, subset analysis of the SHH medulloblastoma was conducted to describe the practice of salvage radiotherapy and associated post-relapse survival (PRS). RESULTS: Sixty-seven SHH medulloblastoma patients (46 M0; 54 GTR; 11 non-ND/MBEN) received salvage therapy with curative intent. Before relapse, 54 (80.6%) received conventional chemotherapy (CC), 13 (19.4%) high-dose chemotherapy (HDC), while seven had additional focal radiotherapy (fRT). Median time to relapse was 11.1 months (range 3.8-41.0) and 43.3% were localized. Thirty patients (16 localized relapse) underwent surgery. Forty-seven (71.2%) received salvage radiotherapy (20 with CC; 10 with HDC; 15 alone, two unknown). CSI and fRT accounted for 82% and 18% respectively. CSI median dose was 36Gy (range 18-39Gy). Ten patients (eight with localized relapse) received CSI doses ≤23.4Gy. Nineteen patients (28.8%) did not receive any radiotherapy (nine HDC; 10 CC only). Radiotherapy was associated with better 3-year PRS (73.0% versus 36.1%; p=0.001). All patients treated with CSI ≤ 23.4Gy were alive at median follow-up of 69 months(24-142). Six of nine patients treated with HDC without irradiation were alive at last follow-up. Sixty-three percent of patients received reduced dose CSI(≤23.4Gy), fRT, or no radiotherapy, and their PRS did not significantly differ from those who received CSI ≥ 30.6Gy (p = 0.54). CONCLUSION: While salvage CSI provided PRS benefit in this SHH medulloblastoma cohort, we report the use of reduced salvage radiotherapy and irradiation avoidance in 63% of the patients, with 60% alive at last follow-up.
  • Item
    Thumbnail Image
    IMMU-08. Nivolumab with or without ipilimumab in pediatric patients with high-grade CNS malignancies: efficacy, safety, biomarker, and pharmacokinetic results from Checkmate 908
    Dunkel, IJ ; Cohen, K ; Foreman, NK ; Hargrave, D ; Lassaletta, A ; André, N ; Hansford, JR ; Hassall, T ; Eyrich, M ; Gururangan, S ; Bartels, U ; Gajjar, A ; Howell, L ; Warad, D ; Pacius, M ; Tam, R ; Wang, Y ; Zhu, L ; Doz, F (Oxford University Press (OUP), 2022-06-03)
    Abstract BACKGROUND: Limited data exist regarding checkpoint inhibitor efficacy for pediatric CNS malignancies. METHODS: CheckMate 908 is an open-label, sequential-arm, phase 1b/2 study investigating nivolumab (NIVO) and NIVO + ipilimumab (IPI) in 5 cohorts of pediatric patients previously treated with standard-of-care (NCT03130959). Patients received NIVO-3mg/kg Q2W or NIVO-3mg/kg + IPI-1mg/kg Q3W (4 doses) followed by NIVO-3mg/kg Q2W. Primary endpoints included OS (newly diagnosed DIPG) and PFS (other CNS cohorts); secondary endpoints included other efficacy metrics/safety. Exploratory endpoints included pharmacokinetics/biomarker analyses. Comparisons between treatments/cohorts were not planned. RESULTS: At data cutoff (13-Jan-2021), 166 patients received NIVO (n=85) or NIVO+IPI (n=81) at median (m) ages of 10.0yrs (range, 1-21) and 11.0yrs (1-21), respectively. In newly diagnosed DIPG, mOS (80% CI) was 11.7mos (10.3-16.5) with NIVO (n=23) and 10.8mos (9.1-15.8) with NIVO+IPI (n=22). In recurrent/progressive HGG, mPFS (80% CI) was 1.7mos (1.4-2.7) with NIVO (n=16) and 1.3mos (1.2-1.5) with NIVO+IPI (n=15). In relapsed/resistant medulloblastoma, mPFS (80% CI) was 1.4mos (1.2-1.4) with NIVO (n=15) and 2.8mos (1.5-4.5) with NIVO+IPI (n=15). In relapsed/resistant ependymoma, mPFS (80% CI) was 1.4mos (1.4-2.6) with NIVO (n=12) and 4.6mos (1.4-5.4) with NIVO+IPI (n=10). In other recurrent/progressive CNS tumors, mPFS (95% CI) was 1.2mos (1.1-1.3) with NIVO (n=19) and 1.6mos (1.3-3.5) with NIVO+IPI (n=19). Median treatment duration was 2.1mos (range, 0-41.7+ [NIVO]/0-29.6+ [NIVO+IPI]). Grade 3/4 treatment-related AEs occurred in 14.1% (NIVO) and 27.2% (NIVO+IPI) of patients. NIVO and IPI first dose trough concentrations were lower in youngest and lowest-weight patients. Baseline tumor PD-L1 expression was not associated with survival. Tumor mutational burden was high in 1 patient (NIVO+IPI) with HGG (OS=11.0mos). CONCLUSIONS: NIVO±IPI demonstrated no clinical benefit in pediatric patients with high-grade CNS malignancies, consistent with available historical data. The safety profiles were manageable.
  • Item
    Thumbnail Image
    A PHASE II STUDY OF CONTINUOUS LOW DOSE PANOBINOSTAT IN PAEDIATRIC PATIENTS WITH MALIGNANT RHABDOID TUMOURS AND ATYPICAL TERATOID RHABDOID TUMOURS
    Wood, P ; Desai, J ; Waldeck, K ; Cain, J ; Gottardo, N ; Strong, R ; Kinross, K ; Carr, M ; Jones, J ; Wong, L ; Ziegler, D ; Hansford, J ; Michael, M ; Ashley, D (OXFORD UNIV PRESS INC, 2022-06)
    Abstract BACKGROUND: Panobinostat treatment has been shown to terminally differentiate malignant rhabdoid tumours (MRT) and atypical teratoid rhabdoid tumours (ATRT) in pre-clinical models. We report results of the open label, phase II study of oral panobinostat in patients with newly diagnosed or relapsed MRT/ATRT. AIMS: To assess the anti-tumour activity of low dose, continuous oral panobinostat as well as its associated toxicities. To assess the biological activity of low dose panobinostat by measuring histone H4 acetylation status in peripheral mononuclear cells (PMNC), and differentiation markers. METHODS: Following primary institutional standard of care induction and consolidation chemotherapy and/or radiation treatment, patients were enrolled and commenced on panobinostat as a continuous daily oral dose starting at 10mg/m2/day, with a three-week wash out period between therapies. Real-time acetylation status, measuring acetylated H4 on PMNC, was performed to determine the pharmacodynamics of panobinostat at different dosing levels. Patients were monitored for toxicity; dose reductions were in decrements of 2mg/m2/day. RESULTS: A total of 13 patients with newly diagnosed ATRT/MRT and one patient with relapsed MRT have been enrolled. The average age at enrollment was 3.6 years (range 0.8-6.8 years). The mean treatment duration was 206 days (13-344 days). Currently, six patients (42.9%) remain on study with a mean study duration of 531 days (range 13-895 days). 6/14 patients (42.9%) were removed due to disease progression at a mean study duration of 245 days (44-560 days). 2/14 patients (14.3%) withdrew due to toxicity. 12/14 patients (85.7%) required dose reductions. The main toxicities were thrombocytopaenia and leukopaenia (Grade III-IV). Real-time pharmacodynamic assessment of panobinostat, at a dose as low as 6mg/m2/day resulted in significant acetylation of histone H4 in PMNC. CONCLUSIONS: Treatment with low dose panobinostat is well tolerated in infants and children with MRT/ATRT, with significant acetylation of histone H4 in PMNC.
  • Item
    Thumbnail Image
    Pediatric pineoblastoma: A pooled outcome study of North American and Australian therapeutic data.
    Hansford, JR ; Huang, J ; Endersby, R ; Dodgshun, AJ ; Li, BK ; Hwang, E ; Leary, S ; Gajjar, A ; Von Hoff, K ; Wells, O ; Wray, A ; Kotecha, RS ; Raleigh, DR ; Stoller, S ; Mueller, S ; Schild, SE ; Bandopadhayay, P ; Fouladi, M ; Bouffet, E ; Huang, A ; Onar-Thomas, A ; Gottardo, NG (Oxford University Press (OUP), 2022)
    BACKGROUND: Pineoblastoma is a rare brain tumor usually diagnosed in children. Given its rarity, no pineoblastoma-specific trials have been conducted. Studies have included pineoblastoma accruing for other embryonal tumors over the past 30 years. These included only occasional children with pineoblastoma, making clinical features difficult to interpret and determinants of outcome difficult to ascertain. PATIENTS AND METHODS: Centrally or independently reviewed series with treatment and survival data from North American and Australian cases were pooled. To investigate associations between variables, Fisher's exact tests, Wilcoxon-Mann-Whitney tests, and Spearman correlations were used. Kaplan-Meier plots, log-rank tests, and Cox proportional hazards models were used in survival analyses. RESULTS: We describe a pooled cohort of 178 pineoblastoma cases from Children's Oncology Group (n = 82) and institutional series (n = 96) over 30 years. Children <3 years of age have significantly worse survival compared to older children, with 5-year progression-free survival (PFS) and overall survival (OS) estimates of 13.5 ± 5.1% and 16.2 ± 5.3%, respectively, compared with 60.8 ± 5.6% and 67.3 ± 5.0% for ≥3 years old (both P < .0001). Multivariable analysis showed male sex was associated with worse PFS in children <3 years of age (hazard ratio [HR] 3.93, 95% CI 1.80-8.55; P = .0006), suggestive of sex-specific risks needing future validation. For children ≥3 years of age, disseminated disease at diagnosis was significantly associated with an inferior 5-year PFS of 39.2 ± 9.7% (HR 2.88, 95% CI 1.52-5.45; P = .0012) and 5-year OS of 49.8 ± 9.1% (HR 2.87, 95% CI 1.49-5.53; P = .0016). CONCLUSION: Given the rarity of this tumor, prospective, collaborative international studies will be vital to improving the long-term survival of these patients.
  • Item
    Thumbnail Image
    Emergence and maintenance of actionable genetic drivers at medulloblastoma relapse
    Richardson, S ; Hill, RM ; Kui, C ; Lindsey, JC ; Grabovksa, Y ; Keeling, C ; Pease, L ; Bashton, M ; Crosier, S ; Vinci, M ; Andre, N ; Figarella-Branger, D ; Hansford, JR ; Lastowska, M ; Zakrzewski, K ; Jorgensen, M ; Pickles, JC ; Taylor, MD ; Pfister, SM ; Wharton, SB ; Pizer, B ; Michalski, A ; Joshi, A ; Jacques, TS ; Hicks, D ; Schwalbe, EC ; Williamson, D ; Ramaswamy, V ; Bailey, S ; Clifford, SC (OXFORD UNIV PRESS INC, 2022-01)
    BACKGROUND: Less than 5% of medulloblastoma (MB) patients survive following failure of contemporary radiation-based therapies. Understanding the molecular drivers of medulloblastoma relapse (rMB) will be essential to improve outcomes. Initial genome-wide investigations have suggested significant genetic divergence of the relapsed disease. METHODS: We undertook large-scale integrated characterization of the molecular features of rMB-molecular subgroup, novel subtypes, copy number variation (CNV), and driver gene mutation. 119 rMBs were assessed in comparison with their paired diagnostic samples (n = 107), alongside an independent reference cohort sampled at diagnosis (n = 282). rMB events were investigated for association with outcome post-relapse in clinically annotated patients (n = 54). RESULTS: Significant genetic evolution occurred over disease-course; 40% of putative rMB drivers emerged at relapse and differed significantly between molecular subgroups. Non-infant MBSHH displayed significantly more chromosomal CNVs at relapse (TP53 mutation-associated). Relapsed MBGroup4 demonstrated the greatest genetic divergence, enriched for targetable (eg, CDK amplifications) and novel (eg, USH2A mutations) events. Importantly, many hallmark features of MB were stable over time; novel subtypes (>90% of tumors) and established genetic drivers (eg, SHH/WNT/P53 mutations; 60% of rMB events) were maintained from diagnosis. Critically, acquired and maintained rMB events converged on targetable pathways which were significantly enriched at relapse (eg, DNA damage signaling) and specific events (eg, 3p loss) predicted survival post-relapse. CONCLUSIONS: rMB is characterised by the emergence of novel events and pathways, in concert with selective maintenance of established genetic drivers. Together, these define the actionable genetic landscape of rMB and provide a basis for improved clinical management and development of stratified therapeutics, across disease-course.
  • Item
    Thumbnail Image
    Systems-Level Change to Alleviate Barriers to Cancer Clinical Trial Access for Adolescents and Young Adults in Australia
    Ellis, JA ; Malalasekera, VS ; Allan, C ; Choong, PF ; Hansford, JR ; Hehir, R ; Morello, N ; O'Callaghan, S ; Orme, L ; Phillipson, N ; Rosenthal, MA ; Sawyer, S ; Strong, R ; Super, L ; Watt, A ; Williams, C ; Woollett, A ; Robertson, A ; Lewin, J (MARY ANN LIEBERT, INC, 2022-04-01)
    Purpose: International data demonstrate association between clinical trial participation and reduced cancer mortality. Adolescents and young adults (AYA) have low clinical trial enrollment rates. We established a program to understand local barriers and develop targeted solutions that lead to greater AYA clinical trial participation. Methods: A steering committee (SC) with expertise in adult and pediatric oncology, research ethics, and consumer representation was formed. The SC mapped barriers related to AYA trial access and established working groups (WGs) around three themes. Results: The Regulatory Awareness WG identified a lack of understanding of processes that support protocol approval for clinical trials across the AYA age range. A guideline to raise awareness was developed. The Access WG identified challenges for young adults (18-25 years) to access a pediatric hospital to enroll in a pediatric trial. A procedure was developed to streamline applications for access. The first six applications using this procedure have been successful. The Availability WG identified lack of pediatric-adult oncology reciprocal relationships as a barrier to awareness of open trials, and future collaboration. An AYA Craft Group Framework was established to grow relationships within tumor streams across institutions; two craft groups are now operating locally. An additional achievement was a successful request to the Therapeutic Goods Administration for Australian adoption of the Food and Drug Administration Guidance on Considerations for the Inclusion of Adolescent Patients in Adult Oncology Clinical Trials. Conclusion: This multipronged approach to improving AYA clinical trial access has relevance for other health environments. Our knowledge products are available as an online toolkit.
  • Item
    Thumbnail Image
    Clinical Outcomes and Patient-Matched Molecular Composition of Relapsed Medulloblastoma
    Kumar, R ; Smith, KS ; Deng, M ; Terhune, C ; Robinson, GW ; Orr, BA ; Liu, APY ; Lin, T ; Billups, CA ; Chintagumpala, M ; Bowers, DC ; Hassall, TE ; Hansford, JR ; Khuong-Quang, DA ; Crawford, JR ; Bendel, AE ; Gururangan, S ; Schroeder, K ; Bouffet, E ; Bartels, U ; Fisher, MJ ; Cohn, R ; Partap, S ; Kellie, SJ ; McCowage, G ; Paulino, AC ; Rutkowski, S ; Fleischhack, G ; Dhall, G ; Klesse, LJ ; Leary, S ; Nazarian, J ; Kool, M ; Wesseling, P ; Ryzhova, M ; Zheludkova, O ; Golanov, A ; McLendon, RE ; Packer, RJ ; Dunham, C ; Hukin, J ; Fouladi, M ; Faria, CC ; Pimentel, J ; Walter, AW ; Jabado, N ; Cho, Y-J ; Perreault, S ; Croul, SE ; Zapotocky, M ; Hawkins, C ; Tabori, U ; Taylor, MD ; Pfister, SM ; Klimo, P ; Boop, FA ; Ellison, DW ; Merchant, TE ; Onar-Thomas, A ; Korshunov, A ; Jones, DTW ; Gajjar, A ; Ramaswamy, V ; Northcott, PA (LIPPINCOTT WILLIAMS & WILKINS, 2021-03-01)
    PURPOSE: We sought to investigate clinical outcomes of relapsed medulloblastoma and to compare molecular features between patient-matched diagnostic and relapsed tumors. METHODS: Children and infants enrolled on either SJMB03 (NCT00085202) or SJYC07 (NCT00602667) trials who experienced medulloblastoma relapse were analyzed for clinical outcomes, including anatomic and temporal patterns of relapse and postrelapse survival. A largely independent, paired molecular cohort was analyzed by DNA methylation array and next-generation sequencing. RESULTS: A total of 72 of 329 (22%) SJMB03 and 52 of 79 (66%) SJYC07 patients experienced relapse with significant representation of Group 3 and wingless tumors. Although most patients exhibited some distal disease (79%), 38% of patients with sonic hedgehog tumors experienced isolated local relapse. Time to relapse and postrelapse survival varied by molecular subgroup with longer latencies for patients with Group 4 tumors. Postrelapse radiation therapy among previously nonirradiated SJYC07 patients was associated with long-term survival. Reirradiation was only temporizing for SJMB03 patients. Among 127 patients with patient-matched tumor pairs, 9 (7%) experienced subsequent nonmedulloblastoma CNS malignancies. Subgroup (96%) and subtype (80%) stabilities were largely maintained among the remainder. Rare subgroup divergence was observed from Group 4 to Group 3 tumors, which is coincident with genetic alterations involving MYC, MYCN, and FBXW7. Subgroup-specific patterns of alteration were identified for driver genes and chromosome arms. CONCLUSION: Clinical behavior of relapsed medulloblastoma must be contextualized in terms of up-front therapies and molecular classifications. Group 4 tumors exhibit slower biological progression. Utility of radiation at relapse is dependent on patient age and prior treatments. Degree and patterns of molecular conservation at relapse vary by subgroup. Relapse tissue enables verification of molecular targets and identification of occult secondary malignancies.