Paediatrics (RCH) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 7 of 7
  • Item
    No Preview Available
    HGG-20. DIAGNOSTIC AND BIOLOGICAL ROLE OF METHYLATION PATTERNS IN REPLICATION REPAIR DEFICIENT HIGH GRADE GLIOMAS
    Dodgshun, A ; Fukuoka, K ; Edwards, M ; Bianchi, V ; Sexton-Oates, A ; Larouche, V ; Magimairajan, V ; Lindhorst, S ; Yalon, M ; Mason, G ; Crooks, B ; Constantini, S ; Massimino, M ; Chiaravalli, S ; Ramdas, J ; Mason, W ; Shamvil, A ; Farah, R ; Van Damme, A ; Opocher, E ; Hamid, SA ; Ziegler, D ; Samuel, D ; Cole, KA ; Tomboc, P ; Stearns, D ; Thomas, G ; Lossos, A ; Sullivan, M ; Hansford, JR ; Jones, D ; Mackay, A ; Jones, C ; Ramaswamy, V ; Hawkins, C ; Bouffet, E ; Tabori, U (Oxford University Press (OUP), 2020-12-04)
    Abstract Replication repair deficiency (RRD) is an important driving mechanism of pediatric high grade glioma (pHGG) occurring predominantly in the context of germline mutations in RRD-associated genes. Although pHGG present specific patterns of DNA methylation corresponding to driving oncogenic processes, methylation patterns have not been well studied in RRD tumors. We analyzed 52 RRD pHGG using either 450k or 850k methylation arrays. These arrays were compared with 234 PHGG driven by other genetic or epigenetic mechanisms and 10 additional pHGG samples known to be hypermutant. RRD pHGG displayed a methylation pattern corresponding to specific secondary mutations such as IDH1 and H3K27M. Strikingly, RRD pHGG lacking these known secondary mutations largely clustered together with a poorly described group previously labelled Wild type-C. Most of the hypermutant tumors clustered in a similar location suggesting undiagnosed RRD may be a driving force for tumors clustering in this location. Analysis of methylation patterns revealed that RRD pHGG displayed a unique CpG Island Demethylator Phenotype in contrast to the Methylator Phenotype described in other cancers. This effect was most concentrated at gene promotors. Prominent demethylation was observed in genes and pathways critical to cellular survival including cell cycle, gene expression, cellular metabolism and cellular organization. These data suggest that methylation profiles may provide diagnostic information for the detection of RRD pHGG. Furthermore, our findings highlight the unique natural selection pressures in these highly dysregulated, hypermutant cancers and provide novel impact of hypermutation and RRD on the cancer epigenome.
  • Item
    Thumbnail Image
    RARE-15. EARLY PSEUDOPROGRESSION POST-RADIATION IN PAEDIATRIC HIGH-GRADE GLIOMA PATIENTS WITH CONSTITUTIONAL MISMATCH REPAIR DEFICIENCY: TWO CASE REPORTS FROM A SINGLE CENTRE
    Teng, A-J ; Gilmartin, B ; Campbell, M ; Bhatia, K ; Wheeler, G ; Sullivan, M ; Hansford, J ; Khuong-Quang, D-A (Oxford University Press (OUP), 2020-12-04)
    Abstract BACKGROUND Constitutional Mismatch Repair Deficiency (CMMRD) is a cancer predisposition syndrome caused by biallelic mutations in the mismatch repair pathway, and high-grade glioma (HGG) constitute the most prevalent brain tumours. Pseudoprogression alludes to radiological changes that mimic tumour progression, but are in fact due to other causes such as therapy related inflammation. It can occur as early as three months post treatment. To our knowledge, its characteristics in CMMRD patients has not been reported. METHODS We retrospectively identified seven patients with CMMRD and history of HGG at The Royal Children’s Hospital, Melbourne from 2005 to 2019. Our objective was to review the characteristics of pseudoprogression in this cohort. RESULTS Out of the seven patients, two with constitutional loss of PMS2 demonstrated evidence of pseudoprogression. Patient 1 presented at 16 years old with a cerebellar anaplastic astrocytoma. She developed clinical and radiological progression within two weeks of starting radiotherapy, persisting up to four months after completion. However, six months post radiation she improved without intervention and the tumour remains stable five years post therapy. Patient 2 presented at 17 years old with a midbrain anaplastic astrocytoma, and showed signs of progression four weeks after completion of radiotherapy. She was then treated with Bevacizumab, an anti-VEGFA antibody with remarkable response. She subsequently received Nivolumab, a checkpoint inhibitor with ongoing stable disease for four months. CONCLUSION Our findings showed that pseudoprogression can occur early in the treatment course in CMMRD patients. Identification of this entity is important for appropriate clinical management.
  • Item
    Thumbnail Image
    PATH-09. SJMB12 CLINICAL TRIAL: DISCREPANCY BETWEEN LOCAL AND CENTRAL PATHOLOGY IN ASSESSING ANAPLASTIC MEDULLOBLASTOMA – REPORT FROM A SINGLE SITE EXPERIENCE
    Khuong-Quang, D-A ; Williamson, K ; MacGregor, D ; Orr, BA ; Gajjar, A ; Robinson, GW ; Sullivan, MJ ; Hansford, JR (Oxford University Press (OUP), 2020-12-04)
    Abstract INTRODUCTION SJMB12 is a phase 2 clinical trial led by the St. Jude Children’s Research Hospital (St. Jude) that enrolls patients with medulloblastoma based on their biological subgroup. The large cell/anaplastic (LCA) histologic variant has been identified as an important independent risk factor associated with poor outcome. However, the histologic criteria for LCA is subjective, making the distinction between anaplastic and non-anaplastic medulloblastoma difficult in some cases. METHODS Pathological central review was performed at St. Jude. For all patients enrolled in the study to date, concordance was assessed between the initial and central review diagnosis and histologic variant calls made at the Royal Children’s Hospital Melbourne (RCH) and at St. Jude, respectively. RESULTS Since the SJMB12 clinical trial opened locally in 2014, 34 patients were enrolled, and 31 were eligible for this retrospective study. A total of 12 (39%) cases with discordance were identified. The most frequent disagreement was between the designation of LCA (10 cases, 32%). In five cases the tumour was not designated as LCA variant locally. In five cases the initial designation of LCA was refuted centrally. Overall, this led to a change of treatment stratum for four patients (13%). CONCLUSION A high discordance rate exists between neuropathologists in the designation of LCA variant. Differences in interpretation of the subjective histologic criteria and inconsistencies in the material submitted for central review contributed to the discordance. Incorporation of more objective histologic criteria and implementation of unbiased diagnostic tools may improve the generalisability of future risk stratification.
  • Item
    Thumbnail Image
    EPEN-36. THE TREATMENT OUTCOME OF PAEDIATRIC SUPRATENTORIAL C11ORF95-RELA FUSED EPENDYMOMA: A COMBINED REPORT FROM E-HIT SERIES AND AUSTRALIAN NEW ZEALAND CHILDREN’S HAEMATOLOGY/ONCOLOGY GROUP
    Ng, CH ; Obrecht, D ; Buntine, M ; Wells, O ; Campbell, MA ; Bhatia, K ; Sullivan, M ; Williams, M ; Quang, DAK ; Kinross, K ; White, C ; Algar, E ; Witt, H ; Schuller, U ; Mynarek, M ; Pietsch, T ; Gerber, NU ; Benesch, M ; Warmuth-Metz, M ; Kortmann, R ; Bison, B ; Taylor, MD ; Ramaswamy, V ; Rutkowski, S ; Pfister, SM ; Jones, DTW ; Gottardo, NG ; Von Hoff, K ; Pajtler, KW ; Hansford, JR (Oxford University Press (OUP), 2020-12-04)
    Abstract AIM Advances in molecular classification of paediatric ependymoma have been pivotal in improving risk stratification and understanding of this disease. C11orf95-RELA fused supratentorial ependymoma (ST-EPN) have been reported to have a poor outcome, with 10-year overall survival (OS) of 49% and progression free survival (PFS) of 19%. A cohort of patients from multiple international institutions with molecularly confirmed C11orf95-RELA fused ST-EPN were reviewed to assess their disease behaviour. METHOD: We reviewed patients with molecularly determined C11orf95-RELA supratentorial ependymoma diagnosed between 1999 – 2019. Demographic information, extent of surgical resection, use of radiotherapy and/or chemotherapy, disease recurrence, treatment at recurrence and clinical outcome data was collected. PFS and OS of all patients were estimated using Kaplan-Meier method. RESULTS A total of 76 ST-EPN patients with C11orf95-RELA fusion were identified (median age: 7 years3 months, range: 5 months – 18 years7 months). 58 patients (76.3%) had complete surgical resection. 70 patients(92.1%) received radiotherapy. 55 patients(72.3%) received chemotherapy. The 10-year OS of C11orf95-RELA fused ST-EPN was 72.4% and PFS was 63.8%. In contrast, ST-EPN at a single institution with unconfirmed molecular status had an OS of 61.1% and PFS of 34.9%. CONCLUSION Detailed molecular analysis identified distinct subgroups of patients with ST-EPN. Patients from this cohort with C11orf95-RELA methylation profiles had a significantly higher OS compared to previous reports and those with unconfirmed fusion status, emphasising the critical importance of complete molecular profiling to assist in treatment decision making. Complete molecular analysis in future prospective cohorts is essential for accurate risk stratification and treatment selection.
  • Item
    Thumbnail Image
    Outcomes by Clinical and Molecular Features in Children With Medulloblastoma Treated With Risk-Adapted Therapy: Results of an International Phase III Trial (SJMB03)
    Gajjar, A ; Robinson, GW ; Smith, KS ; Lin, T ; Merchant, TE ; Chintagumpala, M ; Mahajan, A ; Su, J ; Bouffet, E ; Bartels, U ; Schechter, T ; Hassall, T ; Robertson, T ; Nicholls, W ; Gururangan, S ; Schroeder, K ; Sullivan, M ; Wheeler, G ; Hansford, JR ; Kellie, SJ ; McCowage, G ; Cohn, R ; Fisher, MJ ; Krasin, MJ ; Stewart, CF ; Broniscer, A ; Buchhalter, I ; Tatevossian, RG ; Orr, BA ; Neale, G ; Klimo, P ; Boop, F ; Srinivasan, A ; Pfister, SM ; Gilbertson, RJ ; Onar-Thomas, A ; Ellison, DW ; Northcott, PA (American Society of Clinical Oncology, 2021-03-01)
    Purpose: SJMB03 (ClinicalTrials.gov identifier: NCT00085202) was a phase III risk-adapted trial that aimed to determine the frequency and clinical significance of biological variants and genetic alterations in medulloblastoma. Patients and Methods: Patients 3-21 years old were stratified into average-risk and high-risk treatment groups based on metastatic status and extent of resection. Medulloblastomas were molecularly classified into subgroups (Wingless [WNT], Sonic Hedgehog [SHH], group 3, and group 4) and subtypes based on DNA methylation profiles and overlaid with gene mutations from next-generation sequencing. Coprimary study end points were (1) to assess the relationship between ERBB2 protein expression in tumors and progression-free survival (PFS), and (2) to estimate the frequency of mutations associated with WNT and SHH tumors. Clinical and molecular risk factors were evaluated, and the most robust were used to model new risk-classification categories. Results: Three hundred thirty eligible patients with medulloblastoma were enrolled. Five-year PFS was 83.2% (95% CI, 78.4 to 88.2) for average-risk patients (n = 227) and 58.7% (95% CI, 49.8 to 69.1) for high-risk patients (n = 103). No association was found between ERBB2 status and PFS in the overall cohort (P = .74) or when patients were stratified by clinical risk (P = .71). Mutations in CTNNB1 (96%), DDX3X (37%), and SMARCA4 (24%) were most common in WNT tumors and PTCH1 (38%), TP53 (21%), and DDX3X (19%) in SHH tumors. Methylome profiling classified 53 WNT (17.4%), 48 SHH (15.7%), 65 group 3 (21.3%), and 139 group 4 (45.6%) tumors. A comprehensive clinicomolecular risk factor analysis identified three low-risk groups (WNT, low-risk SHH, and low-risk combined groups 3 and 4) with excellent (5-year PFS > 90%) and two very high-risk groups (high-risk SHH and high-risk combined groups 3 and 4) with poor survival (5-year PFS < 60%). CONCLUSION These results establish a new risk stratification for future medulloblastoma trials.
  • Item
    Thumbnail Image
    Growth hormone and targeted oncological agents: Are we stopping children with brain tumours from reaching their true height potential?
    Walker, H ; Khuong Quang, DA ; Campbell, M ; Bhatia, K ; Williams, M ; Sullivan, M ; Eisenstat, DD ; Kao, K-T ; Ng, J ; White, M ; Zacharin, M ; Hansford, JR (WILEY, 2021-08)
    Children with low-grade gliomas have excellent long-term survival outcomes. The development of therapies targeted to the driver mutations along the Mitogen Activated Protein (MAP) kinase signalling pathway are providing long-term stability for many patients with these tumours. Given the frequency of these tumours residing within or near the suprasellar region, our patients commonly suffer from hormone deficiencies. In Australia, the Pharmaceutical Benefits Scheme currently restricts growth hormone therapy to patients who are not being actively treated for cancer, including those receiving targeted therapies. This viewpoint hopes to facilitate an important discussion amongst our colleagues as to whether this should be changed to allow growth hormone to become available to children on chronic tumour suppressive therapy.
  • Item
    Thumbnail Image
    Medulloblastoma Down Under 2013: a report from the third annual meeting of the International Medulloblastoma Working Group
    Gottardo, NG ; Hansford, JR ; McGlade, JP ; Alvaro, F ; Ashley, DM ; Bailey, S ; Baker, DL ; Bourdeaut, F ; Cho, Y-J ; Clay, M ; Clifford, SC ; Cohn, RJ ; Cole, CH ; Dallas, PB ; Downie, P ; Doz, F ; Ellison, DW ; Endersby, R ; Fisher, PG ; Hassall, T ; Heath, JA ; Hii, HL ; Jones, DTW ; Junckerstorff, R ; Kellie, S ; Kool, M ; Kotecha, RS ; Lichter, P ; Laughton, SJ ; Lee, S ; McCowage, G ; Northcott, PA ; Olson, JM ; Packer, RJ ; Pfister, SM ; Pietsch, T ; Pizer, B ; Pomeroy, SL ; Remke, M ; Robinson, GW ; Rutkowski, S ; Schoep, T ; Shelat, AA ; Stewart, CF ; Sullivan, M ; Taylor, MD ; Wainwright, B ; Walwyn, T ; Weiss, WA ; Williamson, D ; Gajjar, A (SPRINGER, 2014-02)
    Medulloblastoma is curable in approximately 70% of patients. Over the past decade, progress in improving survival using conventional therapies has stalled, resulting in reduced quality of life due to treatment-related side effects, which are a major concern in survivors. The vast amount of genomic and molecular data generated over the last 5-10 years encourages optimism that improved risk stratification and new molecular targets will improve outcomes. It is now clear that medulloblastoma is not a single-disease entity, but instead consists of at least four distinct molecular subgroups: WNT/Wingless, Sonic Hedgehog, Group 3, and Group 4. The Medulloblastoma Down Under 2013 meeting, which convened at Bunker Bay, Australia, brought together 50 leading clinicians and scientists. The 2-day agenda included focused sessions on pathology and molecular stratification, genomics and mouse models, high-throughput drug screening, and clinical trial design. The meeting established a global action plan to translate novel biologic insights and drug targeting into treatment regimens to improve outcomes. A consensus was reached in several key areas, with the most important being that a novel classification scheme for medulloblastoma based on the four molecular subgroups, as well as histopathologic features, should be presented for consideration in the upcoming fifth edition of the World Health Organization's classification of tumours of the central nervous system. Three other notable areas of agreement were as follows: (1) to establish a central repository of annotated mouse models that are readily accessible and freely available to the international research community; (2) to institute common eligibility criteria between the Children's Oncology Group and the International Society of Paediatric Oncology Europe and initiate joint or parallel clinical trials; (3) to share preliminary high-throughput screening data across discovery labs to hasten the development of novel therapeutics. Medulloblastoma Down Under 2013 was an effective forum for meaningful discussion, which resulted in enhancing international collaborative clinical and translational research of this rare disease. This template could be applied to other fields to devise global action plans addressing all aspects of a disease, from improved disease classification, treatment stratification, and drug targeting to superior treatment regimens to be assessed in cooperative international clinical trials.