Header image

SESSION 6.2 - Conference Abstracts Presentations

Tracks
Track 2
i. Basic science
ii. Translational
Friday, November 8, 2024
11:00 AM - 1:00 PM
Tyree Room, John Niland Scientia Building

Agenda Item Image
Dr Radek Szmyd
Postdoc
Children's Medical Research Institute

Chair

Biography

Radek is a mid-career researcher at Children’s Medical Research Institute (CMRI) in Sydney with over 10 years of experience in cancer cell biology, cell cycle and genome instability. Graduated with a MSc in Biotechnology from Jagiellonian University (Krakow, Poland) and PhD in Molecular and Cell Biology from National University of Singapore. During his PhD, Radek used a genetic mouse model to investigate spatial and temporal relationships between DNA replication and chromosome segregation, and the impact of their misregulation on cell death and tumorigenesis. Immediately after the PhD, he’s joined Prof Tony Cesare’s lab at CMRI as a lead postdoctoral researcher in the collaborative project between Prof Cesare and Assoc/Prof Harriet Gee. In this project, he has characterized molecular mechanisms of cell death in response to lethal DNA damage following ablative radiotherapy. Radek’s current research focuses on targeting DNA damage response in combination with radiotherapy to elicit synergy with immunotherapy for cancer patients benefit.
Dr Smadar Kahana-Edwin
Children's Hospital at Westmead, Sydney Children’s Hospitals Network

Harnessing liquid biopsy to unveil somatic RAS-MEK pathway variants in extracranial AVMs

11:00 AM - 11:15 AM

Abstract

Background: Arterio-venous malformations (AVMs) are rare congenital high-flow vascular anomalies characterized by abnormal direct artery-to-vein communications, lacking intervening capillaries. Complex extracranial AVMs, typically have a limited response to standard treatments such as sclerotherapy or surgery, leading to a progressive disabling pathology with lifelong consequences that include chronic pain, deformity, and disfunction. Somatic variants in the RAS-MEK pathway are implicated in AVMs, generating interest in the role of MEK inhibitors to treat this condition. However, open biopsy for molecular characterisation carries a potentially life-threatening bleeding risk.
We aimed to evaluate the effectiveness of liquid biopsy for somatic genotyping of AVMs in children and young adults. We hypotheses that liquid biopsy from efferent draining veins is a reproducible and feasible technique for obtaining DNA from AVM, and that detection of the underlying somatic mosaic variants from AVMs can assist in directing targeted pharmacotherapy.
Methods: 10 patients with complex AVMs for which endovascular treatment or diagnostic catheter angiography was being undertaken were enrolled and 15 blood samples collected (11 efferent vein, 3 synchronous peripheral vein, and 1 peripheral vein only). Ultra-deep panel next-generation sequencing was conducted on cell-free DNA (cfDNA), followed by error-correction sequencing data analysis and variant calling.
Results: Pathogenic variants were identified in 12 samples from 8 patients (levels 0.11%-20.06%). No procedural complications occurred from efferent sampling. Three patients with identified variants began genotype-directed pharmacotherapy, demonstrating marked clinical improvement.
Variant levels inversely correlated with the sampling distance from the AVM nidus (p=0.004). Similarly, variant levels in synchronous peripheral collections were reduced by 33-100% in 3 patients (p=0.035).
Conclusion: Somatic variants of the RAS-MEK pathway are common in extracranial AVMs and can be readily identified using efferent vein liquid biopsy, albeit with a moderate yield in peripheral samples. This approach reduces bleeding risk, enabling broader access to targeted pharmacotherapy for AVM patients.

Biography

Dr Smadar Kahana-Edwin is a molecular biologist and a cancer researcher with specific training and expertise in translational cancer research. She is an emerging leader in liquid biopsy research and has special interest in developing minimally invasive approaches for precision medicine to improve diagnostics and care. Smadar has been working at the Children’s Hospital at Westmead since 2017 to develop blood tests for monitoring the presence of circulating tumour material in patients’ blood as well as identifying targetable mutations in non-oncological conditions for oncological treatment repurposing.
Dr Pablo Acera
Children's Cancer Institute

Cryptico: A Computational Approach for Exploring Cryptons in Search of Novel Therapeutics"

11:15 AM - 11:30 AM

Abstract

Background: The development of new cancer therapies is constrained by the proteome's limited druggability, estimated at only 10-15%. Additionally, the increasing recognition of non-coding RNAs' roles in disease underscores the urgency for innovative therapeutics that move beyond conventional protein-targeting strategies. The emergence of RNA-targeting small molecules presents a promising avenue for novel therapeutic approaches, potentially expanding the range of actionable targets. Recent advancements include the use of splicing modulator compounds (SMCs) to suppress gene expression by introducing deleterious cryptic exons (cryptons) in a sequence specific manner.
Methods: In this study, we developed a computational method to systematically identify potential targets for SMCs, specifically cryptons within the human genome. This method combines deep learning with high-throughput RNA-sequencing data from human tissues to discover high confidence putative cryptons.
Results: We detected around 50k cryptons and predicted their therapeutic potential based on their ability to disrupt gene expression through PTCs, frameshift mutations, or the alterations of protein folding due to the introduction of novel peptide chains. Furthermore, our analysis revealed 41,664 previously unannotated cryptons, some of which are predicted to interfere with several oncogenes traditionally considered undruggable. We further validated the existence of several of cryptons in therapeutically relevant genes, by engineering one of the spliceosome components to match a specific cryptons 3’ splice sites.
Conclusion: Our findings provide a valuable resource for exploring potential therapeutic targets of SMCs and lay the groundwork for future drug screening efforts.

Biography

Pablo Acera is a Postdoctoral Researcher at the Computational Drug Discovery Lab, Children's Cancer Institute, where they have worked since 2022 under the supervision of Antoine de Weck. Their research focuses on developing advanced statistical and machine learning methods to enhance drug discovery, particularly through identifying novel RNA splicing modulators compounds. He earned his PhD in Bioinformatics from the Australian National University in 2022, with a dissertation on developing deep learning models for RNA modification detection using nanopore sequencing. Previously, he worked at the the European Bioinformatics Institute, developing algorithms for analyzing capture-HiC datasets. He holds a Master’s degree in Bioinformatics from Lund University and Utrecht University, and a Bachelor's degree in Biology from the University of Seville.
Agenda Item Image
Dr Piyushkumar Mundra
Children's Cancer Institute

Inferring the life history of NTRK-fusion paediatric tumours

11:30 AM - 11:45 AM

Abstract

Background: Many paediatric cancer tumours are driven by rearrangements in neurotrophic tyrosine receptor kinase (NTRK) genes (NTRK1, NTRK2 and NTRK3). While several inhibitors targeting tropomyosin receptor kinases family genes have been either approved or are at an advanced stage of clinical development, many paediatric cancer patients develop resistance to these inhibitors. Tumour evolution studies in adult cancers suggest polyclonal tumours likely to develop therapy resistance. We hypothesized that characterizing the tumour evolution of pediatric cancers with NTRK fusions might reveal insights into resistance mechanisms.
Methods: In our extended ZERO-Childhood Cancer cohort, we identified 26 tumours (from 24 children) with NTRK-fusion events. This cohort includes central nervous system tumours (CNS; n=13), sarcomas (n=8), neuroblastomas (n=1) and other solid tumours (n=4). We performed ~90X whole genome sequencing on these tumours and corresponding germline samples (at ~30X) and analysed these data using computational genomic tools, including SAGE, PURPLE and LINX to identify somatic single nucleotide variants (SNV), copy number variants (CNV) and structural variants. We then applied SNV based tumour evolution framework to identify clonal and subclonal populations. The order of mutation clusters was inferred using evolutionary principles. We also identified four tumour samples that have undergone whole-genome doubling (WGD). The timing of WGD was inferred using GRITIC pipeline.
Results: In this cohort of NTRK-fusion tumours, SNV based clonality analysis revealed that 23 out of the 26 tumours were polyclonal in nature, while the monoclonal tumours exhibited low purity. Multi-time point analyses from two patients showed evidence of branching evolution. One CNS tumour presented with founding whole genome doubling event, while in other tumours WGD was relatively late event.
Conclusion: Polyclonality is a common feature of NTRK-fusion paediatric tumours, while timing of WGD may explain evolution of some tumours. Such inherent features of clonal evolution may help delineate therapy resistance in these tumours.

Biography

Dr Piyushkumar Mundra has extensive experience in developing and applying bioinformatics and machine learning tools in the computational identification of biomarkers for cancer. Since being awarded his PhD in 2011, he has built an outstanding track record for mid-career researcher, with 45 peer- reviewed publication. He has gained extensive experience in cancer genomics, lipidomics and development of machine learning algorithm. His research is published in leading journals, such as Science, Nature Medicine, Nature Communications and Nature Cancer. He has extensive experience working in international laboratories in Australia, the United Kingdom and Singapore.
Agenda Item Image
Dr Lauren Brown
Children's Cancer Institute

CBL mutations in CNS and solid tumours - a new therapeutic opportunity?

11:45 AM - 12:00 PM

Abstract

Background: Molecularly targeted therapies improve outcomes for childhood cancer patients and are made possible by personalised medicine approaches like the Zero Childhood Cancer Program (ZERO). Mutations in CBL, an E3 ubiquitin ligase, represent one such molecular target in acute myeloid leukaemia, where CBL mutation mediates activation of the Receptor Tyrosine Kinase (RTK) FLT3 and sensitivity to FLT3-targeted Tyrosine Kinase Inhibitors (TKIs). We have identified novel CBL mutations in paediatric CNS and solid tumours, raising the possibility that CBL mutations mediate RTK activation in other tumour types and these patients may benefit from TKI therapy.
Methods: We analysed the ZERO cohort for somatic mutations in CBL. Molecular (whole genome, RNA sequencing and methylation) and high-throughput drug screening data from CBL mutated-patients, performed as part of ZERO, were interrogated. In silico pathogenicity analysis (ESM-2) was performed on novel variants. CBL mutations were cloned from patients and modelled in vitro to analyse the functional and signalling impacts of CBL mutation.
Results: We identified 26 somatic CBL variants in 22 individual patients, including 14 patients with CNS tumours. Thirteen non-haematological tumour samples harboured CBL mutations that are established drivers (exon 8/9Δ) or suspected to be oncogenic given their location in critical domains and in silico pathogenicity analysis. Over half of these samples had molecular indications of RTK activation (7/13). Most CNS tumours did not fall into a pre-defined DNA methylation-based subtype classification, suggesting CBL mutation may represent a distinct molecular entity. In vitro modelling of exon 8/9Δ in a neuroblastoma cell line demonstrated that this variant enhances proliferation through RTK activation in other tumour types.
Conclusions: CBL mutation may be a marker of RTK activation and therapeutic target in CNS and solid tumours. This finding will extend the benefit of TKIs, which have proven efficacy in paediatric cancer, to more patients.

Biography

Lauren is a Research Officer in the Translational Tumour Biology Research Group, under the supervision of Associate Professor Paul Ekert, at Children’s Cancer Institute and Conjoint Associate Lecturer at the University of New South Wales (UNSW). She completed her PhD at The University of Melbourne in 2019 where her research focussed on the identification and characterisation of novel kinase-activating fusion genes in paediatric acute lymphoblastic leukaemia (ALL). Her current research focusses on characterising novel mechanisms of receptor tyrosine kinase (RTK) activation in paediatric cancers broadly and is guided by novel genomic findings from the Zero Childhood Cancer Program (ZERO).
Agenda Item Image
Mr Steven He
Phd Student
Children's Cancer Institute

Identifying non-coding lncRNA dependencies in high-risk MLL-rearranged acute myeloid leukaemia

12:00 PM - 12:15 PM

Abstract

Background: CRISPR-Cas9 genome-wide screening has comprehensively mapped the dependency landscape in cancer, including paediatric subtypes. Whilst therapeutic discovery in certain cancers has benefitted from this, other high-risk subtypes, such as MLL-rearranged acute myeloid leukaemia (MLLr-AML), still lack therapeutic options. Existing work in the dependency-mapping space has focused predominantly on the coding genome, ignoring potential novel cancer dependencies in non-coding regions. Long non-coding RNAs (lncRNAs) in particular represent untranslated RNAs that are increasingly implicated in cancer development, including in MLLr-AML. In the current study, we perform both CRISPR-Cas9 screening and Cas13d secondary screening to identify novel lncRNA dependencies in MLLr-AML, with Cas13d as a validation tool that better emulates therapeutic conditions, where genes are often modulated instead of completely suppressed.
Methods: Stable Cas9-positive MLLr-AML cells were generated using lentiviral transduction, with enzymatic activity confirmed through single-target knockdown. In vitro genome-wide CRISPR-Cas9 lncRNA KO screening was performed in three Cas9-positive MLLr-AML cell lines transduced with single guide RNA (sgRNA) libraries at ~500x coverage. Following 21 days in culture, gDNA was isolated and sequenced to observe overall sgRNA representation. lncRNA dependencies were identified using both MAGeCK and CHRONOS analysis softwares. Stable Cas13d-positive MLLr-AML cells were similarly generated for pooled CRISPR-Cas13d secondary screening.
Results: MAGECK identified 5 lncRNA gene dependencies (fold-change < -1.5, FDR < 0.01) overlapping all three cell lines in the CRISPR-Cas9 screen. In CHRONOS, 18 genes showed strong dependency (gene-effect score < -1) across all three cell lines. A custom Cas13d library for secondary screening was designed containing sgRNAs targeting the identified dependencies. Three stable Cas13d-positive MLLr-AML cell lines were generated in preparation. Flow cytometry following transduction with CD45 sgRNAs showed successful knockdown in these lines.
Conclusion: Utilising a genome-wide screening approach we have identified lncRNA dependencies in MLLr-AML which will be further validated utilising a CRISPR-Cas13d secondary screening strategy.

Biography

Steven completed his B.Sc (Hons) at the University of Sydney before working as an RA in both academic and industry settings. He went on to complete his MRes in a Bioinformatics research group at Macquarie University before beginning his current PhD studies at the Children's Cancer Institute Australia in the Computationally-Enable Drug Discovery Group under the primary supervision of Antoine de Weck.
Dr Wenhan Chen
Children’s Cancer Institute

Individualized-tumor-informed panel approach enables ultra-sensitive ctDNA-based minimal disease monitoring for pediatric cancers

12:15 PM - 12:30 PM

Abstract

While the use of ctDNA-based minimal residual disease (MRD) monitoring is gaining popularity in adult cancers, this approach is not yet widely adopted in paediatric cancers (PCs). However, this approach is not widely implemented in pediatric cancers. Enabled by the ZERO program, Australian’s national Childhood Cancer Precision Medicine program, we developed a robust and sensitive detection method to monitor ctDNA in PCs. This method overcomes PC-specific challenges including low tumor mutational burden, lacking of hotspot mutations and limited blood from children. Specifically, our assay includes personalized panel design which targets primary tumor mutations from each patient, a fixed panel targeting hotspot mutations, error suppression from duplex deep sequencing and customized software for UMI de-duplexing (REDUX) and tumor fraction estimation (WISP).
We retrospectively analyzed 661 blood or cerebrospinal fluid (CSF) samples from 168 PC patients. We achieved an average detection limit of 0.005% ctDNA fraction and a minimal limit of 0.0001%. We detected ctDNA burdens ranged widely from 0.001% to 100% with brain (CNS) tumors consistently showing a low ctDNA burden (average 0.03%). When patients were identified with high disease burden, our approach identified ctDNA in over 93% of extra-cranial solid tumours, 90% of leukaemia, 89% of sarcoma, and 90% of Neuroblastoma. Our approach showed that ctDNA burden dynamics correlated with disease progression or remission. Importantly, we identified circulating disease in ~20% of clinically complete response assessments, predicting relapse 1–6 months before clinical manifestation. Finally, with the fixed panel, our assay identified emergence of novel variants, suggesting potential new treatment options during tumour evolution. Overall, these results demonstrated that our ctDNA-based disease monitoring approach for pediatric cancer is an important addition to standard of care, quantifying disease burden and tracking disease evolution for precision-guided treatment.

Biography

Wenhan Chen is a senior bioinformatician in the Computational Biology Group, Children's cancer institute. He is an enthusiastic tool developer. He created software tools for association studies, somatic variant calling, and recently for detecting and quantifying tumor burden from liquid biopsy.
Mr James Bradley
Children’s Cancer Institute

A Data Lakehouse for Childhood Cancer Precision Medicine

12:30 PM - 12:45 PM

Abstract

Background: The Zero Childhood Cancer Program (ZERO) is a precision medicine program with currently ~1600 patients enrolled to date, with the program forecasted to have 3500 patients by end of 2025. This poses two data challenges: how to store and analyse billions of germline variants from this cohort, and how to integrate different data sources and types from within and outside the program to interpret analysis outcomes. For these large and complex datasets, we selected a Data Lakehouse architecture for its combination of performance, data-type flexibility and cost-effectiveness.
Method: To address these challenges, we implemented a cloud Data Lakehouse underpinned by the Delta file format and distributed computation engine Spark, on the Databricks platform. Additionally, we created a scalable data pipeline to investigate pharmacogenic interactions with adverse drug reaction (ADR) in ZERO patients to demonstrate the ability to efficiently integrate heterogenous data.
Results: We successfully populated a distributed germline variant database with >1600 whole genomes for query and analysis; this achieved genome range cohort queries for over 8.3 billion variants in under 5 seconds and a 60:1 compression ratio from raw VCF (10TB to 165GB).
We then integrated PharmKGB’s drug-pharmacogenic interactions and ZERO’s ADR dataset with ZERO’s germline database, enabling detection of patients with ADR patients that had pharmacogene alleles related to their ADR-causing drugs. We identified 3 ADR patients out of 34 patient-drug combinations with possible pharmacogene links, one having a well-documented pharmacogene (UGT1A1*93) related to their ADR therapy.
Conclusion: This project demonstrates how the Data Lakehouse architecture can be successfully implemented to meet the scale of the ZERO program’s analytical needs.

Biography

James Bradley is a recent graduate of UNSW, obtaining a bachelors degree in Software Engineering and bachelors degree in Science majoring in Bioinformatics. He is currently employed as a junior bioinformatics data engineer at the Children's Cancer Institute. He specializes in data management of large amounts of genomic data as part of the Children's Cancer Institue's paediatric precision medicine program ZERO. He is currently working on implementing a production scaled version of his honours thesis, in building a Data Lakehouse to meet the analytic demands of the ZERO program as the program scales both nationwide and internationally.
Miss Safaa Al Haj Hussein
Children's Hospital at Westmead, Sydney Children’s Hospitals Network, The University of Sydney

Rapid Fire Presentation: PREDICT and Validate: Investigating Germline Alterations in Paediatric Cancer Predisposition

12:45 PM - 12:50 PM

Abstract

Germline alterations in cancer predisposition genes (CPGs) are associated with increased risk of developing certain cancers. Identifying underlying germline genetic contribution to cancer development not only assists in developing a personalised management plan for the patient, but also provides cancer risk clarification and guidance for family. Studies suggest 10-15% of paediatric cancer patients carry germline pathogenic variants in CPGs. However, a proportion of patients with features suggestive of underlying cancer predisposition, have no pathogenic alterations identified in known CPGs. The PREDICT study was conducted to identify germline variants in patients <21 years, newly diagnosed with cancer across NSW, together with biological parents where possible (trio analysis), using whole genome sequencing (WGS). Our team aimed to comprehensively interrogate the genome sequencing data to identify and functionally validate novel germline variants that may contribute to cancer predisposition.
We have established bioinformatics pipelines to analyse PREDICT germline WGS data against a panel of ~1250 cancer-related genes, to investigate genomic alterations within and beyond protein-coding regions, including exonic single nucleotide variants and small insertions/deletions, splicing variants, non-coding region variants (e.g. promoter regions), and structural alterations. Trio-based WGS data was run on The University of Sydney's High Performance Computing system, with analysis incorporating RStudio and online curation tools.
Clinically actionable pathogenic/likely pathogenic variants were detected in 51/230 (22.2%) enrolled patients (e.g. PMS2, NF1), and 62 novel candidate variants were identified in 53/230 (23.0%). Functional validation of prioritised variants-of-interest recurrently identified in B cell leukaemia/lymphoma, is currently being conducted in transduced murine pro-B cell line Ba/F3, which stably express wildtype and each mutant, to assess their functional impacts on the relevant pathway.
Our comprehensive WGS analysis has identified known and novel germline alterations. Further characterisation of novel findings may broaden our understanding regarding underlying mechanisms of paediatric cancer predisposition, and lead to newly defined genotype-phenotype associations.

Biography

Safaa Al Haj Hussein is a third year PhD student based at the Children's Cancer Research Unit, Kids Research at The Children's Hospital at Westmead. Through the PREDICT Study, her project focuses on discovering novel germline variants that predispose children to developing cancer. This includes using bioinformatics pipelines to detect, predict and prioritise variants which are then investigated using functional validation. Safaa has presented her PhD work at multiple conferences including the Australian Society for Medical Research NSW Annual Scientific Meeting (2023), Sydney Cancer Partners NSW Cancer Conference (2023, funding support received from the conference) and Kids Cancer Alliance Symposium (2023, first prize student oral presentation). Safaa has also received prestigious scholarship support from The University of Sydney and Tour De Cure.
loading