VPCC team

VPCC team members are PhD, Masters and Honours students, and early-career researchers from across our network of partners. VPCC staff are also part of the team. They support and create crucial platforms that underpin our work. Together, the VPCC team represents the next generation of paediatric cancer research that will make a difference in the lives of children with cancer.

Hudson Institute of Medical Research

Dr Paul Daniel

Ihara Shazia Adjumain

Ihara Shazia Adjumain is a PhD candidate co-funded by the Robert Connor Dawes Foundation and Hudson Institute of Medical Research. She completed a Bachelor of Biomedical Science at Deakin University and moved to Monash University to pursue an Honours year within the next-generation precision medicine program. Under the supervision of Professor Ron Firestein and Dr Paul Daniel, she is focussing on identifying novel therapeutic targets in paediatric high-grade gliomas with the aim of changing the odds of children fighting this debilitating disease.

Project description
High grade gliomas are malignant tumours that arise from the central nervous system. Despite advances in treatment, the five-year survival rate is still ~20%. Hence, there is an urgent need to discover novel therapies that could be used to treat patients with the disease. Shazia’s PhD project will use an integrative genomic approach consisting of CRISPR screens, Drug screens and “omics” data to identify novel targets in paediatric high-grade gliomas. This will be achieved through a comparative analysis process with adult high-grade gliomas. She is currently evaluating the role of BCL-2 family of proteins in driving tumourgenesis in paediatric high grade gliomas.

Nicole Alexia

Nicole Alexia is a final-year biomedicine student from Indonesia International Institute for Life Sciences (i3L), with a specialisation in tumor biology. She is intrigued by the complexity of cancers and aspires to decipher therapeutic means against cancers. This drove her to visit the Hudson Institute for Medical Research for her undergraduate internship.

Project description

Nicole studies paediatric brain cancers such as the rare atypical teratoid rhabdoid tumor (ATRT) with the Next Generation Precision Medicine Program team. Her work mainly involves growing various patient-derived ATRT cell lines and then subjecting them to high-throughput drug screening against a library of 700 anticancer drugs, with an aim of identifying robust drug candidates.

 

Nicole Alexia
Vikesh Ajith

Vikesh Ajith

Vikesh Ajith is a Bioinformatics Research Assistant for the Hudson Next Generation Precision Medicine Program. He completed his Masters of Biotechnology and Bioinformatics At La Trobe University in 2022 where he gained a strong interest in the development and implementation of reproducible and robust bioinformatics pipelines.

Project description
Vikesh’s main role is to support the team with their bioinformatics analyses, through data management, data cleaning, primary data analysis on High-Performance Computing (HPC) clusters, the development of data visualisation tools such as the analytical portal of the Childhood Cancer Model Atlas, and assisting researchers and students with troubleshooting technical issues in bioinformatics.

Samitha Amarapathy

Samitha Amarapathy leads the agile driven software development capability at Monash eResearch Centre, delivering productive outcomes for research-led and research infrastructure-led projects. In addition to that he leads national-level IT projects in partnership with ARDC, Australian universities and other research institutes.

From a biology and IT academic background, he was keen to integrate the biology and IT knowledge to deliver outcomes that create a global impact for social good. Samitha was passionately exploring the arena of data science and AI in recent years in the context of its applications on precision medicine, developed a PhD proposal and initiated a PhD project in collaboration with Faculty of IT at Monash University and Hudson Medical Research Institute.

Project description

Samitha’s PhD project aims to build a machine learning/deep learning-based decision support tools that provides recommendations on precision medicine for paediatric brain cancer patients based on clinical, genomic and functional dependency data (CRISPR, drug screens). He closely works with Next Generation Precision Medicine Program to deliver positive outcomes from an AI perspective.

 

Samitha Amarapathy

Dr Nicole Chew

Dr Nicole Chew is the Children’s Cancer Foundation senior organoid specialist for the Next Generation Precision Medicine Program, she joined the program in 2021. Dr Chew graduated with first-class Honours in BSc and completed her PhD at Monash University under the supervision of Professor Roger Daly. During this time, she investigated therapeutic targets and drug combinations for particular subtypes of breast cancer and liver cancer, utilising in vitro (including generation of inhibitor-resistant cell lines and organoids) and in vivo (PDX) models. Her research has identified novel targetable fusions and drug combinations as improved treatment strategies.

Project description
Paediatric brain cancer is the most common solid tumour cancer affecting children and young adults. By building cohorts of specific and rare brain cancers, these models would help identify therapeutic targets and biomarkers to improve patient prognosis. Dr Chew’s research work involves generating in vitro and in vivo models such as primary cell lines, xenografts and organoids derived from paediatric brain tumours to identify therapeutic targets and biomarkers as potential treatment options using pharmacological and functional genomic approaches.

Nicole Chew

Hui Kheng Chua

Hui Kheng Chua is a Senior Research Assistant with the Cancer Genetics and Functional Genomics lab, and the Lab Manager for the Centre for Cancer Research.

Hui Kheng also works with the Next Generation Precision Medicine program, led by Prof Ron Firestein.

 

Hui Kheng Chua
Samiksha Dhananjay

Samiksha Dhananjay

Samiksha Dhananjay is a Master of Biomedical and Health Science student at Monash University. Before embarking on her Masters, Samiksha completed a Bachelor of Biomedical Science degree at Monash University in 2018 and worked as a research assistant for 3 years in the Nervous System Repair and Development Laboratory (Monash Biomedicine Discovery Institute), assisting and overseeing research projects interrogating nervous system and muscle regeneration. 

Project description
Samiksha joined the Leukaemia Modelling and Therapeutic Discovery group at the Hudson Institute of Medical Research in late 2023 for her Masters research project and is working on developing childhood AML models for use in large scale drug and genetic screens to identify unique functional dependencies and new therapeutic targets.

Dr Paul Daniel

Dr Paul Daniel

Dr Paul Daniel is a Senior Postdoctoral Research Fellow for the Hudson Monash Next Generation Precision Medicine Program. After completing his PhD with Dr Theo Mantamadiotis at Melbourne University, Dr Daniel undertook a three year postdoctoral fellowship at McGill University in Canada, looking into therapeutic opportunities in adult glioblastoma which emerge following exposure to chemotherapy. At the conclusion of Dr Daniel’s time in Canada, he then joined the Next Generation Program, led by Prof Ron Firestein, where he is involved in building the first paediatric-focused functional dependency map and expanding the library of known therapeutic opportunities for paediatric cancers.

Project description
Prolonged exposure of tumour cells to therapy results in progressive adaptation and acquisition of resistance, ultimately leading to treatment failure and disease recurrence. While preventing the acquisition of resistance has proved elusive thus far, we now know that adaptation of tumour cells to one type of therapy often results in the emergence of alternate therapeutic opportunities. Dr Daniel is currently interested in defining the underlying mechanisms driving acquired resistance to therapy in paediatric and adult brain cancers and how we can modulate evolutionary outcomes to favour emergence of states with predictable vulnerabilities. His research involves use of clinically relevant in vitro and in vivo tumour models, such as patient-derived cell lines and xenografts.

Shaye Game

Shaye Game is an Isabella and Marcus Foundation PhD scholar in the Developmental and Cancer Biology lab, Centre for Cancer Research at the Hudson Institute of Medical Research. Shaye graduated with first-class honours in Bachelor of Biomedical Science (Honours) from Monash University in 2020.

During honours Shaye investigated the epigenetic dysregulation in Diffuse Intrinsic Pontine Glioma (DIPG), utilising patient-derived cell line models and CRISPR/CAS9 under Dr Jason Cain and Professor Ron Firestein. Shaye found a passion and desire to further the research and therapeutic opportunity for DIPG patients she continued her project into a PhD.

Project description
Diffuse intrinsic pontine glioma (DIPG) is a devastating and fatal paediatric high-grade glioma that develops in the pons of the brain stem. Next generation sequencing has revealed ~80% of DIPG tumours exhibit an exclusive and highly recurrent heterozygous mutation in genes encoding histone variants, H3.3 (H3F3A) and H3.1 (HIST1H3B and HIST1H3C), resulting in lysine to methionine substitution (H3K27M), which is essential for epigenetic control of gene expression during development. Shaye’s project hopes to investigate the precise impact of H3K27M and subsequent epigenetic dysregulation on DIPG tumorigenesis to identify novel therapeutic opportunities.

Shaye Game
Hugh Gao

Dr Hugh Gao

Dr Hugh Gao is a medical doctor currently undertaking his PhD under the supervision of Professor Ron Firestein, Dr Dan Garama, and Dr Chunhua Wan. During his PhD, Hugh designed a kinase-targeted combinatorial CRISPR-cas12 library which enables the simultaneous evaluation of almost 100,000 synthetic lethal combinations of genes in cancer cell models. Using this unique resource, he is currently seeking to identify novel druggable synergies in childhood cancer which can be translated across to pre-clinical and clinical trials

Dilru Habarakada

Dilru Habarakada

Dilru Habarakada is the Biobank Manager of the Monash Children’s Paediatric Biobank, a partnership between the VPCC Next Generation Precision Medicine (HMPPM) and Monash Health. She leads in collaborating the Next Generation program and global scientists to establish a ‘living biobank’ of childhood brain/CNS and solid cancer – including living organoids and lab grown mini tumours to trial and develop targeted treatments. 

Her focus on a career in paediatrics and oncology, sees her develop collaborative professional relationships with clinical and non-clinical teams to facilitate the delivery and improvement in clinical outcomes, survival rates and quality of life (limiting side-effects) for childhood cancer patients. As the Biobank Manager, she brings technical and operational knowledge to the development of the ‘living biobank’ platforms by providing strategic guidance and project management support for clinical trials and clinical projects within the program.

Bao Le

Bao Le is a Junior Research Assistant with the Next Generation Precision Medicine Program (NGPMP) at the Hudson Institute of Medical Research. He graduated with first-class Honours in the Bachelor of Medical Science at Flinders University. Throughout his studies, Bao gained extensive experiences with molecular and cell biology techniques as well as developing a passion for translational research.

Project description
Bao’s work revolves around developing and characterising primary cell lines, organoids, and patient-derived xenograft models from paediatric tumours. These models will contribute to various projects in the NGPMP, the wider Victorian Pediatric Cancer Consortium (VPCC), and external collaborators. Additionally, he will contribute to high-throughput drug screens, helping to identify new therapeutic targets.

Bao Le
Hanbyeol Lee

Dr Hanbyeol Lee

Dr Hanbyeol Lee is a postdoctoral scientist involved in functional genomic approaches to identify new therapeutic targets in childhood brain cancers.

She undertook a Bachelor of Science (Biomedical technology major) and completed her phD at Kangwon National University in South Korea, where she investigated the therapeutic application of stem cells in human pulmonary disease. She had experience working in academia and bioindustry and focused widespread application of living organoids from stem cells to study infectious disease and genetic disorders in human cardiovascular disease. At the conclusion of Dr Lee’s time in Korea, she then joined the Next Generation Program in 2023, led by Prof Ron Firestein, where she is expanding a strong interest in functional genomic screening to identify new therapeutic targets for children with brain cancers.

Project description

Currently, she is involved in conducting a project for high throughput functional genomics screens using CRISPR/Cas systems to map functional dependencies in diverse cell lines from human paediatric brain tumours.

Dr Yuqing Liang

Yuqing Liang is a PhD student at Hudson Institute of Medical Research in the Next Generation Precision Medicine Program. Yuqing completed a Bachelor of Medicine Oncology at Sun Yat-sen University in China. She then majored in paediatric oncology at Sun Yat-sen University Cancer Centre, where her research was focused on identifying prognostic markers in children with high-risk neuroblastoma via clinical features and CT radiomics. To pursue her interest in precision medicine, Yuqing began her PhD under the supervision of Professor Ron Firestein and Professor David Eisenstat in 2021, focusing on identifying novel therapeutic targets in diffuse midline glioma (DMG). Her aim is to create a positive impact on therapeutic options for this vulnerable population.

Project description
Yuqing’s research is centred on diffuse midline glioma (DMG), a debilitating form of childhood brain cancer for which the overall prognosis has remained poor for decades. Hence there is an urgent need to discover novel therapies that could be used to treat patients with the disease. As part of Yuqing’s PhD project, she will perform innovative genetic screens on DMG patient models to identify new therapeutic targets and biomarkers that predict response.

Yuxing Liang
Steve Lin

Dr Steve Lin

Dr Steve Lin is a senior research assistant in the Leukaemia Modelling and Therapeutic Discovery group at the Hudson Institute of Medical Research. Steve has a strong background in biological sciences and obtained a Master of Science in Biochemistry and Molecular Biology from Nanjing University in China and a PhD in Physiology from Victoria University.

Project description

Steve assists with developing novel models of childhood AML using unique genetic and cellular approaches. Once developed, these models will be incorporated into the Next Generation Precision Oncology pipeline established within the Hudson Institute’s Centre for Cancer Research.

Melissa Loi

Melissa Loi is the technical sequencing specialist for the Next Generation Precision Medicine Program at the Hudson Institute of Medical Research. She completed her Bachelor in Biotechnology and received Honours in Nanotechnology from the University of South Australia in 2009. Prior to joining the Hudson Institute, Melissa had over 10 years’ experience in the field of Infectious diseases, Immunology and therapeutics quality control.

Project description
Her role involves using molecular methods to assist in generating an encyclopaedia of genomic profiles for childhood brain tumours. Using in vitro models of paediatric brain tumour, she uses CRISPR-Cas9 technology to perform knock-out screens and generate DNA libraries for sequencing. Her work also involves preparing samples to decode the epigenetics modifications that regulate gene expression using methylation analysis and chromatin accessibility sequencing.

Melissa Loi

Amanda Nguyen

Amanda Nguyen is a medical student who is currently doing an honours year at the VPCC. 

Project description
Her work is focused upon identifying novel synergistic drug combinations for childhood brain cancers such as DIPG. Amanda’s work makes use of the VPCCs childhood cancer focused compound library with which she routinely performs high-throughput combinatorial screens using leading drug candidates currently in clinical trials. Amanda hopes to find effective combination therapies which can be rapidly translated across from preclinical studies to clinical trials where they can improve patient outcomes for devastating childhood cancers.

Amanda Nguyen
Monty Panday

Monty Panday

Monty Panday completed his Masters of IT at Swinburne University in 2022, specialising in software development. He previously graduated from Deakin University in 2018 as Bachelor of Information Technology majoring in software development and cloud computing. He works closely with researchers and clinical oncology staff to manage and update a number of databases including scientific, clinical and biobanking databases.

Project description
Monty manages the database systems for VPCC Next Generation Precision Medicine Program and was tasked with developing the new database for the Next Generation Precision Medicine Program. In his role, he manages the delivery of data analytics and reporting to help achieve the program’s operation objectives and funding milestones.

Niveditha

Nivedhitha Selvakumar

Nivedhitha is a Master of Biotechnology student at Monash University who is currently undertaking a research project at the Translational Antigen Discovery Lab under the supervision of Dr. Faridi, as part of her final year laboratory placement. She completed her Bachelor of Engineering in Biotechnology from India.

Project description
Her project aims to identify and characterise novel classes of tumour neo-antigens that are presented on the surface of the Diffuse Intrinsic Pontine Glioma cancer cell lines specifically focussing on the peptides presented by HLA-G and HLA-F. She aims to find immunotherapeutic targets for DIPG patients.

Christie Sun

Christie Sun is a PhD candidate in the Centre of Cancer Research, Developmental and Cancer Biology Research group at the Hudson Institute of Medical Research. Christie completed a Master of Reproductive Sciences with first class honours from Monash University.

Her interest in cancer research was enhanced by her experiences during her Masters research into female infertility and has formed the basis of her career path. The research title of Christie’s research is improving childhood osteosarcoma risk stratification and outcomes for recurrent disease.

Project description
Osteosarcoma is the most common malignant primary bone tumour, accounting for approximately one in five bone sarcomas worldwide and more than one in three bone cancers in Australia. To date, although all great efforts have been made in the treatment management, clinical outcomes stay stagnant and remain poor for the osteosarcoma patient. In this project, we will assess the primary tissue of sarcoma patients to determine molecular and functional pathways predictive of therapeutic response, metastasis and survival outcomes. Identifying predictive biomarkers of therapeutic response and survival would represent a major development in the field and enable the future risk stratification of patients and appropriate adaptation of therapy to minimise and improve side effects overall.

Ms Christie Sun
Priya Sundaravel

Dr Priya Sundaravel

Dr Sundaravel undertook a Bachelor of Medicine and Bachelor of Surgery at Monash University. She is currently a Paediatric Fellow at the Royal Children’s Hospital, and an Honorary Registrar in the Paediatric Haematology-Oncology Unit at Monash Children’s Hospital. Dr Sundaravel’s research focuses on the identification of novel prognostic biomarkers for paediatric and adolescent sarcomas.

At the Hudson Institute of Medical Research, she volunteers as part of Dr Jason Cain’s program of sarcoma research.

Dr Xin (Claire) Sun

Dr Claire Sun is a highly skilled computational biologist and provider of bioinformatics support. Dr Sun obtained a Master of Science from Melbourne University (2013) and PhD from Monash University (2018) using next generation sequencing technologies to identify non-coding RNA biomarkers in human urine and blood samples and undertake pioneering work in epigenetic regulation of mitochondrial DNA in tumorigenesis. In 2019, Dr Sun joined the Centre for Cancer Research where she has developed a keen interest in developing and applying state-of-the-art computational approaches to identify new therapeutic targets and biomarkers for low-survival paediatric cancers.

In the past five years, Dr Sun has been leading the efforts to develope the computational framework and analytical methods underlying the Victorian Paediatric Cancer Consortiums Childhood Cancer Model Atlas. The goal of this atlas is to facilitate ‘big’ data- driven discovery of new therapeutic targets for paediatric cancers of lowest survival. As the lead bioinformatician of the program, Dr Sun works as part of a 25-person multi-disciplinary team, in collaboration with wet lab scientists and paediatric oncologists, and directly supervises Honours (x3) and PhD students (x2). Dr Sun’s leadership in this team-based effort is highlighted by a first-author data-resource paper in Cancer Cell. Dr Sun is the current recipient of a Victorian Cancer Agency Early-Career Fellowship and has authored 17 publications in high impact journals including Cancer Cell, Molecular Cell, Journal of Clinical Investigation and Science Advances.

Project description
In the VPCC Next Generation Precision Medicine Program, Dr Sun is working with cutting-edge bioinformatics tools to translate basic research findings into clinical applications.

Xin Cliare Sun
Shanuki Suriyasena

Shanuki Suriyasena

Shanuki Suriyasena is a summer intern at the VPCC who is helping to perform high-throughput drug screens across the cohort of childhood cancer models in the childhood cancer model atlas (CCMA). 

Project description
She has a specific focus on identifying novel therapeutics which target cancer-associated fibroblasts (CAFs), a key population of cells which support the growth and survival of many cancers. Using her experience at the VPCC, Shanuki is applying to pharmaceutical science masters programs which she hopes will ultimately enable her to design more effective treatments for childhood cancers.

Erwin Tanuwidjaya

Erwin Tanuwidjaya

Erwin is a PhD candidate with immunology and proteomics background. He came in contact with the -omics field while finishing his Master of Biotechnology at Monash University in 2021, on antibody engineering. He joined Monash Proteomics and Metabolomics Facility as Research Officer to develop his expertise in proteomics and gain experience in liquid chromatography/mass spectrometry (LC/MS), before joining the Faridi Lab in 2023 for his PhD studies.

Project description
Erwin’s project focuses on unravelling the potential the soluble HLA (sHLA) peptidome in liquid biopsy samples in the context of cancer, by utilising Immunopeptidomics and LC/MS.

Lynda Truong

Lynda Truong is a junior research assistant in the Leukaemia Modelling and Therapeutic Discovery group at the Hudson Institute of Medical Research. Lynda completed her Bachelor of Science Advanced Research with first class Honours from Monash University in 2022, and is currently working as a research assistant to enhance her laboratory skills prior to beginning her PhD studies in 2024. 

Project description
She is helping to develop novel models of childhood AML using cutting edge gene manipulation technologies in umbilical cord blood stem cells.

Lynda Truong
Vanessa Tsui

Dr Vanessa Tsui

Dr Vanessa Tsui is a Robert Connor Dawes Postdoctoral Research Fellow focusing on functional genomic approaches to identify new therapeutic targets in childhood brain cancers. She undertook a Bachelor of Science (Genetics major) and Master of Biomedical Science at the University of Melbourne. During her Masters, she investigated the role of interleukin-11 in adult glioblastoma. Dr Tsui completed her PhD with A/Prof Wayne Crismani at St Vincent’s Institute of Medical Research, where she studied Fanconi anaemia, a disease that predisposes patients to cancer. At the conclusion of her PhD in 2022, she joined Prof Firestein’s lab where she is currently applying innovative genetic technologies to map the functional dependencies in rare forms of childhood brain cancers, funded by the Robert Connor Dawes Foundation.

Project description
Paediatric brain tumours are the leading cause of death among children and young adult cancer patients. There is a need to improve prognosis and identify new therapeutic targets. Utilising paediatric brain tumour cell lines, Dr Tsui will conduct high throughput functional genomics screens using CRISPR/Cas9 technology to map functional dependencies in rare forms of childhood brain cancers.

Janette Vanessa

Janette Vanessa is a final year Master of Pharmaceutical Science student at Monash University completing her research placement in the Leukaemia Modelling and Therapeutic Discovery group at the Hudson Institute of Medical Research. 

Project description
Janette is working with Lynda Truong to develop novel models of childhood AML using cutting edge gene manipulation technologies in umbilical cord blood stem cells.

Janette Vanessa
Dr Vijesh Vaghjiani

Dr Vijesh Vaghjiani

Dr Vijesh Vaghjiani is a Postdoctoral Scientist with the Developmental and Cancer Biology research group and the Sarcoma program.

Project description
Dr Vaghjiani’s research program focuses on understanding the oncogenic roles of embryonic signalling pathways in cancer. He is particularly interested in the Hedgehog signalling pathway that is strongly implicated in a variety of both paediatric and adult cancers.

Dr Yichen Zhou

Dr Yichen Zhou is a postdoctoral researcher focusing on functional genomic approaches to identify new therapeutic targets in childhood brain cancers.

She completed her PhD with Prof Charles Mackay at Monash University in 2022. During her PhD, she investigated the role of gut microbiota and microbial metabolites in Alzheimer’s disease. After her PhD, Dr Zhou joined iCamuno Biotherapeutics and her project focus on cancer cell therapies utilising induced pluripotent stem cell-derived natural killer cells. In 2023, she joined the Next Generation Precision Medicine program, where she is currently involved in combinatorial CRISPR/Cas12 screening in human paediatric brain tumours.

Project description
Diffuse midline gliomas are the second most common type of primary high grade brain tumour in children. They are a type of glioma that grows in the midline between the two halves of the brain. There is an urgent need to discover novel therapies that could be used to treat patients with the disease. Dr Zhou will conduct a project in high throughput functional genomics screens using CRISPR/Cas12 systems to map functional dependencies in diverse cell lines from human paediatric diffuse midline gliomas.

Dr Yichen Zhou
Danxi Zhu

Dr Danxi Zhu

Dr Danxi Zhu is a medical graduate who recently completed her PhD under Prof Ron Firestein. 

Project description
She is currently working in the VPCC to generate a comprehensive phospho- and total- proteomics dataset across the rich cohort of cell line models in the Childhood Cancer Model Atlas (CCMA). She envisions that this work will enable more accurate prediction of therapeutic efficacy which will ultimately enhance current efforts to use precision medicine approaches in childhood cancers.

Monash University

Liesl Bramberger

Liesl Bramberger

Liesl Bramberger is a PhD student working with Dr Pouya Faridi in his Translational Antigen Discovery Lab at Monash Medical Centre. She completed her Bachelor of Science at Monash University and continued with an Honours degree in the Purcell Lab at Monash University. After Dr Faridi started his own lab, she followed to help set up the new lab at Monash Medical Centre. Here she started working with Dr Jason Cain, her co-supervisor, on histone deacetylase inhibitors in the pediatric brain cancer, Diffuse Intrinsic Pontine Glioma (DIPG).

Project description
Liesl Bramberger’s project is about developing combination therapies in DIPG to stimulate immunotherapeutic vulnerabilities. She is studying the effect of treatments such as epigenetic modulators on the expression of different immunotherapy targets in DIPG cell lines using mass spectrometry techniques.

Dr Kasey Chan

Kasey Chan is a research officer in Professor Lee Wong’s lab at Monash Biomedicine Discovery Institute. She completed her PhD at the Murdoch Children’s Research Institute on novel therapies for thalassaemia. Following the completion of her PhD, she worked as a research fellow at the Melbourne Research and Development department of the Australian Red Cross Lifeblood, focusing on blood components and transfusion research.

Project description

Recent research has revealed that histone gene mutations are commonly found in paediatric gliomas. Histones are crucial proteins responsible for packaging our genetic material (DNA) and directing the normal differentiation and maturation of cells into various cell types. In my project, I employ advanced genomics and proteomics techniques to investigate how histone mutations alter the packaging of DNA.

We have gathered evidence indicating that the presence of mutated H3.3 alters the composition of proteins in PML nuclear bodies. Our hypothesis suggests that mutant H3.3 may drive the development of cancer by disrupting the protein composition, organization, and function of PML bodies, thereby inhibiting cell differentiation and promoting the formation of brain tumours. This disruption could be attributed to the differing binding affinities of mutant H3.3 with chromatin modifiers within the PML bodies. My study aims to utilize a CRISPR Cas9 screening approach to identify chromatin factors that bind to mutant histones within the PML bodies. I also seek to determine whether these proteins could serve as potential therapeutic targets for treating paediatric brain cancers.

 

Kasey Chan
Andrew Garvie

Dr Andrew Garvie

Andrew Garvie is a research assistant who recently received his PhD in the Wong Laboratory at Monash Biomedicine Discovery Institute. His PhD focused on uncovering key factors involved with the Alternative Lengthening of Telomeres (ALT) telomere maintenance pathway and identifying new therapeutic strategies to treat these cancers using the ALT pathway to gain cellular immortality. Currently, he is investigating how histone H3.3 and ATRX mutations drive cellular immortality in paediatric glioblastomas. In addition, he is testing out a new drug to treat H3.3/ATRX-mutated glioblastomas. He has helped publish several highly cited papers on how H3.3/ATRX mutations lead to paediatric brain tumours and is currently working on a first-author publication that focuses on a treatment strategy for H3.3/ATRX mutated glioblastomas.

Project description
High-grade paediatric glioblastomas are the most aggressive and common solid cancer in children, with a median survival of 1.5 years. A seminal discovery was the identification of recurrent H3.3 mutations in paediatric glioblastomas: H3.3K27M and H3.3G34R. These H3.3-mutated tumours frequently show concurrent mutations in ATRX and activation of the Alternative Lengthening of Telomeres (ALT) pathway to evade telomere loss and enable cell immortality. Based on the previous data from his host lab and new, unpublished data from Andrew’s PhD study, he has compelling evidence that paediatric glioblastomas with histone H3.3 and ATRX mutations show severe ribosomal DNA (rDNA) repeat instability and, thus, are compromised in their ability to produce ribosomal RNA (rRNA). Therefore. they are hypersensitive to RNA Polymerase I (Pol I) inhibition. Our collaborator, Prof Ross Hannan (ANU), has developed the “best-in-class” Pol I inhibitor PMR-116, which is able to cross the blood-brain barrier. Andrew thus proposes to test PMR-116 efficacy on patient-derived glioma cell lines. Outcomes from this study will confirm the potential use of PMR-116 as a new therapeutic approach for treating incurable brain cancers.

Gabriel Goncalves

Gabriel Goncalves is a PhD candidate in the Translational Antigen Discover Lab (Faridi Lab) at Monash Medical Centre. Gabriel completed his Bachelor of Science with a first class honours degree at Monash University under the supervision of Professor Anthony Purcell and Dr Pouya Faridi. He is currently a third year PhD student focused on non classical HLA peptide presentation in cancer.

 

Gabriel Goncalves

Grace Huang

Grace Huang is a PhD candidate in the Translational Antigen Discover Lab (Faridi Lab) at Monash Medical Centre. She completed a Bachelor of Biomedical Science and a Bachelor of Commerce at Monash University in 2021 and then pursued an Honours degree in the Faridi Lab. Under the supervision of Dr Pouya Faridi and Dr Jason Cain, head of Developmental and Cancer Biology Research group and co-supervisor, she is focussing on identifying HLA-peptides that can be targeted in Ewing’s Sarcoma.

Project description
Grace’s project involves understanding the classes of HLA complexes that are presented on tumour cell surface, specifically on Ewing’s sarcoma, and what peptides are presented on these complexes. She hopes to identify novel immunotherapeutic targets for paediatric patients with Ewing’s Sarcoma.

Grace Huang
Linda Hii

Linda Hii

Linda Hii is a research officer/lab manager in the Wong laboratory at Monash Biomedicine Discovery Institute. She graduated from Lambuth University, Tennessee, USA majoring in Biology and Chemistry. She then obtained a master’s degree from Monash University under Prof Peter Rogers’ supervision defining integrin alpha V beta 3 expression during the menstrual cycle in the endometrium to potentially develop a new fertility indicator. She has since worked at Murdoch Children’s Research Institute developing novel gene therapy using artificial mini-chromosomes; Peter MacCallum Cancer Centre looking at the function of transcription factor IFI16 to develop chemotherapy for mouse cancer model; and focusing on treating in vitro and in vivo mouse tumour models with various novel antibodies developed against the Eph receptor EphA3 and Adam10 metalloproteases.

Project description
Mutations in proteins that package DNA are frequent events in malignant paediatric brain cancers. One of the most common mutations occurs in a specialised protein known called histone H3.3. Histone H3.3 has an amino acid at position 31 (serine 31) which distinguishes H3.3 from canonical H3.1/2 and lies close to common paediatric brain cancer mutations (lysine to methionine at position 27; glycine to arginine at position 34). We are investigating if the serine 31 residue may contribute to the effects of the brain cancer mutations, and potentially explain the high rates of H3.3 mutations in paediatric brain cancer. To test this, we are creating targeted substitution mutations of the serine 31 residue and testing the capacity for self-renewal and cell differentiation in cellular and animal models. This project is a key part of a multi-pronged investigation into the importance of H3.3 mutations in paediatric brain cancers.

Dr Dongbin Jin

Dr Jin completed his Bachelor of Science in Biology in China and Master of Science in Immunology in South Korea, before undertaking his PhD and Postdoctoral research on cancer vaccine development at Garvan Institute in Sydney Australia, under the supervision of Prof Jonathan Sprent. His expertise is on animal models of anti-tumour studies, cellular immunology, dendritic cells, cell culture, T cell functional assays, and flow cytometry.

Project description
At Garvan, Dr Jin used preclinical models to develop novel vaccine technologies that can expand cytotoxic T cells in an unprecedented way. These technologies are broadly applicable to different types of cancer and infectious diseases. He plans to extend these exciting findings into more clinically relevant settings in the Translational Antigen Discovery Laboratory at Hudson Institute of Medical Research.

Dongbin KJin

Samia Khan

Samia is a master’s student of Biomedical and Health Science at Monash University. As a part of her final year thesis, she is working on a project at the Faridi Lab and Cain Lab. She commenced her undergraduate degree in Biomedical Engineering from the Military Institute of Science and Technology, Bangladesh. She was awarded the Bangabandhu Science and Technology Fellowship by the Ministry of Science and Technology, Government of the People’s Republic of Bangladesh, to conduct her master’s degree at Monash University. She has prior experience in research work as a part of her undergraduate thesis and worked at a research institute as a research assistant.

Project description

Lung cancer is the leading cause of cancer mortality worldwide, accounting for >19% of all cancer-related deaths. Lung adenocarcinoma (LUAD) accounts for approximately 40% of all lung cancers. Mutually exclusive recurrent mutations in the SWI/SNF subunits SMARCA4 and ARID1A are reported in human LUAD with a frequency of 10% and 8%, respectively, and are associated with poor prognosis. Samia’s project primarily aims at identifying differential protein and phospho-protein expression between SMARAC4-deficient and wild-type lung adenocarcinoma isogenic cell lines. Additionally, the project aims at determining the effect of low-dose panobinostat treatment on global protein and phospho-protein expression in those cell lines.

 

Samia Khan
Terry Lim

Dr Terry Lim

Dr Terry Lim graduated with a Bachelor of Biotechnology with first-class Honours (specialising in medical biotechnology) and subsequently completed a PhD at Monash University under the supervision of Professor Anthony Purcell. His PhD work involved cutting-edge proteomics and immunopeptidomics mass spectrometry techniques to understand the immunology of the human leukocyte antigen (HLA)-B27 in the development of the autoimmune disease Ankylosing Spondylitis.

Dr Lim’s interests in cancer biology has led a Postdoctoral Research Fellow in Professor Roger Daly’s laboratory. During this time, he applied phosphoproteomics techniques as a tool to dissect the molecular changes in pancreatic, brain and breast cancer patient-derived organoids (PDO) and xenografts (PDX) models, to identify novel biomarkers and therapeutic targets for personalised cancer treatments.

Project description
Dr Lim currently holds a joint research fellow position in VPCC researcher Dr Pouya Faridi’s laboratory and the Monash Proteomics and Metabolomics Facility. Using both mouse and human models of diffuse midline glioma (DMG), his project involves developing immunotherapy applying his expertise in (phospho)proteomics and immunopeptidomics, with a focus in developing high-throughput platforms for large scale clinical proteomics studies.

Murray Manning

Murray Manning

Murray Manning is the Platform Manager at the Monash Functional Genomics Platform. This platform provides the facilities and expertise to support genomics research at Monash University and externally. He completed his Masters in Molecular Biology at the Katholieke Universiteit Leuven in Belgium, achieving Cum Laude honours for his thesis in the developmental biology field. Environmental biology and developing his talent for the use of CRISPR for answering questions in molecular biology led him to continue his career at the Monash Biomedicine Discovery Institute in 2019. He continued carrying out research in the Rosenbluh lab using pooled CRISPR screening to elucidate the links between breast cancer genes, before joining the Genomics Platform in 2021. His current focus is on providing cutting-edge techniques to enable researchers in their experiments and data acquisition.

Project description
As part of the Victorian Paediatric Cancer Consortium, he will undertake combination CRISPR screens in paediatric cell lines, aimed at identifying new drug targets and mechanisms of action.

Cody Mutch

Cody Mutch is a PhD student in A/Prof Lee Wong’s lab at the Monash University Biomedicine Discovery Institute.

Project description
For his PhD, Cody is focused on characterising the effect of H3.3 oncohistone mutations and PML nuclear bodies on paediatric glioma cell differentiation.

Cody Mutch
Matt Neve

Matthew Neve

Matt Neve is a final year PhD student in the Cancer Program at the Monash Biomedicine Discovery Institute. Matt’s interested using in innovative techniques to identify the cellular machinery that drives cancer growth. This will enable the development of new cancer targeting drugs.

Project description
In my project I am studying the effect of knocking down certain circular RNA molecules cancer cells. We use assays to measure the functional consequence of this knockdown on cancer cell growth. Identifying which circular RNA are contributing to paediatric brain cancer will then allow to us to begin targeting them therapeutically.

 

Lachlan Nott

Lachlan Nott is an Honours student in Dr Pouya Faridi’s Translational Antigen Laboratory team.

Project description
Lachlan’s honours project focuses on using cell surface proteomics to identify potential immunotherapy targets in Ewing sarcoma and osteosarcoma. His project description provides a clear framework for studying the surface proteins of these aggressive bone cancers, aiming to pinpoint targets crucial for tumour progression and immune evasion. This precise approach supports the development of personalized treatments and the discovery of biomarkers for patient stratification.

Lachlan Nott
Murray Manning

Dr Laura Perlaza-Jiménez

Laura Perlaza-Jiménez (PhD) is a Bioinformatician in a split role between Rosenbluh Laboratory and Monash Genomics and Bioinformatics Platform (MGBP). She completed her doctoral study in Bioinformatics at Max Planck Institute (MPIMP, Germany) and has a master’s in science and microbiology (Andes University, Colombia) and a Biology Major (Javeriana University, Colombia). Laura has experience in NGS analysis, comparative genomics, phylogenetics, metagenomics, transcriptomics, and single cell, as well as different scripting languages.

Project description

Laura’s tasks are supporting the team with bioinformatics analysis, mainly on single cell analysis and development of customised scripts for data wrangling, visualization and statistical analysis.\

Dr Tima Shamekhi

Dr Tima Shamekhi received her PhD in Nanobiotechnology in 2018. In 2021, she joined Translational Antigen Discovery-Faridi Laboratory at Monash Medical Centre, to pursue her interest in identifying HLA-bound peptide targets for the development of paediatric brain tumours precision immunotherapies. She is honoured to have been awarded the Kye Funch scholar, supported by the Australian Communities Foundation through the Isabella and Marcus Foundation, to investigate diffuse intrinsic pontine glioma (DIPG), the most aggressive of all childhood cancers.

Project description
Dr Shamekhi’s project is about using multi-omics technologies, particularly genomics, transcriptomics, and immunopeptidomics, to identify novel targets for DIPG and other childhood brain tumour immunotherapy.

Fatemeh Shamekhi

Tamara Tongoi

Tamara Tongoi is a PhD student at the Monash Biomedical discovery Institute in the Rosenbluh lab. Tamara completed a Bachelor of Science at Monash University majoring in Biochemistry and Genetics, followed by an honours year at the Rosenbluh lab. Under the supervision of A/Prof Sefi Rosenbluh, she began functional genomics work investigating cancer-related variants affecting hereditary diffuse gastric cancer.

Project description
Tamara’s research is focused on using CRISPR base editors to investigate and classify cancer-related variants that affect development of hereditary diffuse gastric cancer (HDGC). The project involves using high-throughput genomic screens on candidate cell lines to identify functional variants.

Tamara Tongoi
Simon Tsang

Simon Yung Shing Tsang

Yung Shing Tsang is a Masters student who has completed a Bachelor in Science (Biology) at The Chinese University of Hong Kong in 2016 and is currently studying Master of Biomedical and Health Science in Monash University. He has completed a research project investigating factors driving Alternative Lengthening of Telomeres (ALT) maintenance pathway in the Wong laboratory at Monash Biomedicine Discovery Institute.

Project description
Alternative Lengthening of Telomeres (ALT) is a key telomere maintenance pathway used in human cancers. ALT cancers are often linked to inactivating mutations in ATRX; however, ATRX loss alone isn’t sufficient to trigger ALT. This suggests the involvement of other essential factors in activating the ALT pathway. Recent research from our lab has revealed that mutations in histone H3.3 (H3.3 G34R) and the Krebs cycle gene Isocitrate dehydrogenase I (IDH1 R132H) are two such factors that, when combined with ATRX loss, activate ALT in gliomas. Both H3.3 G34R and IDH1 R132H mutations inhibit H3K9/K36 KDM4 demethylases, with KDM4B being the critical demethylase deactivated in ALT cancers.

ATRX mutations are also prevalent in paragangliomas and pheochromocytomas (PGL/PCC), and these neuroendocrine tumors frequently feature mutations in Krebs cycle genes like fumarate hydratase (FH) and succinate dehydrogenase (SDHx), leading to the accumulation of fumarate and succinate. In PGL/PCC, ATRX mutations are associated with ALT activation and aggressive clinical behavior. My project aims to investigate whether FH and SDHx mutations also inhibit KDM4B and whether they, in combination with ATRX loss, contribute to ALT-related telomere maintenance phenotypes.

Ms Maheshi Udugama

Maheshi Udugama

Maheshi Udugama is working as a research officer in the Wong laboratory at Monash Biomedicine Discovery Institute. She earned her Bsc (Hons) in Biochemistry and Molecular Biology from University of Colombo, Sri Lanka and then moved to the USA to continue with her PhD at Southern Illinois University. Her PhD focussed on the mechanism of ATP dependent chromatin remodelling of INO80 complex. She also investigated the phenomenon of mitotic bookmarking in erythroid progenitors during her postdoc at Children’s Hospital of Philadelphia, USA. Currently she is investigating how histone H3.3 mutations drive the activation of telomere maintenance mechanism and cellular immortality in paediatric glioblastomas. She has been an author on 11 publications including in Cell, Genome Research, Nature Communications, PNAS.

Project description
Brain cancers are the leading cause of cancer-related mortality in children and young adults. DNA sequencing studies have identified two common point mutations in histone variant H3.3, in paediatric glioblastomas (pGBMs). The first mutation replaces Lysine 27 with a Methionine (K27M). The second one replaces Glycine 34 with an Arginine (G34R). H3.3G34R mutations always overlap with ATRX mutations, and these pGBMs are activated in the alternative lengthening of telomeres (ALT) telomere maintenance pathway. One of my research aims is to explore how H3.3 G34R and ATRX act together to drive ALT activation, and use this information to develop novel therapeutic strategies for treating H3.3/ATRX-mutated pGBMs. My other aim is to investigate the effects of H3.3 and ATRX mutations on ribosomal gene organisation and transcription in ALT-positive pGBMs. The outcome of this work will be the identification of new potential targets in pGBMs vulnerable to therapeutic interventions.

Dr Hsiao Voon

Dr Hsiao Voon is a postdoc in the Wong lab at the Biomedical Discovery Institute (Monash University), where she studies chromatin biology and genome regulation. Dr Voon completed her PhD at the Murdoch Children’s Research Institute (University of Melbourne) on gene therapy for thalassaemias. She then completed a postdoc at the Weatherall Institute (Oxford University) where she studied the role of ATRX in gene and chromatin regulation. ATRX/H3.3 were subsequently found to be frequently mutated in paediatric brain tumours. She is currently a postdoc in the Wong lab at Monash University where she is investigating how mutations in ATRX/H3.3 promote the formation of brain tumours. She has published a number of highly cited papers on the biology of ATRX and H3.3, and how mutations in this complex lead to paediatric brain tumours.

Project description
Lab engineers in the Wong lab develop precise genetic mutations to create accurate cell models which we use to understand brain cancers. These mutant cell lines are analysed using high-throughput sequencing to create chromatin and transcription profiles. Hsiao conducts bioinformatics analyses of these profiles to pinpoint how mutations act individually and in combination to promote brain cancers. We have successfully used these techniques to show that H3.3 G34R acts through inhibition of a chromatin modifier (KDM4). We have further shown this mutation acts in concert with ATRX mutations to facilitate telomere maintenance in cancer. We are currently working on how paediatric glioma H3.3 mutations disrupt the formation and localisation of nuclear compartments, with the aim of targeting these compartments for therapy.

Ms Hsiao Voon

WEHI (Walter and Eliza Hall Institute of Medical Research)

Valeria Arcucci

Dr Valeria Arcucci

Dr Valeria Arcucci is a postdoctoral researcher in A/Prof Misty Jenkins’ laboratory at WEHI (Walter and Eliza Hall Institute of Medical Research). She completed her PhD in 2021 at the Peter MacCallum Cancer Centre and moved to WEHI to pursue her interest in immune-oncology and the biology of the tumour microenvironment in brain tumours. Her project focuses on the characterising the interactions between immune cells and tumour cells within the tumour microenvironment of paediatric brain cancers in order to identify biomarkers and shape immunotherapies for children with brain tumours.

Project description
Paediatric brain tumours are the most common type of solid tumours in children and also the main cause of death from cancer. Immunotherapies have recently proven to be incredibly efficacious in treating a variety of human cancers and could be the right approach in treating paediatric brain tumours that do not respond to current therapies. Response to immune therapies has been shown to closely correlate with the tumour immune microenvironment (TME) and therefore an accurate knowledge of the TME of paediatric brain malignancies is crucial in predicting responses to the immunotherapies being tested. Importantly, this information could shape the type of immunotherapies that should be used to treat different central nervous systmn malignancies.
Dr Arcucci’s project aims to perform in-depth analysis of the TME of paediatric brain tumours using cutting-edge “omics” technologies including spatial transcriptomics and MIBI-TOF, a multiplex staining technique that allows staining of 40 markers at once. With this project, she ultimately aims to define in detail the characteristics of the TME of paediatric brain tumours so that she can help shape immunotherapies for children with brain tumours.

Dr Alexander Davenport

Dr Alexander Davenport is a Postdoctoral scientist at WEHI (the Walter and Eliza Hall Institute). After completing his PhD in fundamental CAR-T cell biology at the Peter MacCallum Cancer Centre he moved to the Cambridge Institute for Medical Research, University of Cambridge UK, focussing on the fundamentals of T-cell biology. He then moved to a start-up where he led a team in the creation of immune cells from iPSCs which is seeking to solve to problem of cellular supply in immuno-oncology. Alex was recruited back to Australia as a post doc at the start of 2022 under the supervision of A/Prof Misty Jenkins to discover novel protein targets and create new immunotherapies for paediatric brain cancers.

Project description

Diffuse Midline Glioma (DMG) is a rare but lethal paediatric brain tumour with a median survival of one year. Less than 10% of cases survive past two years. The only current proven treatment is radiotherapy. With the success of CAR-T cell therapy in childhood leukaemia we are looking to expand this into a range of childhood brain cancers, most notably DMG. One of the greatest challenges is finding surface targets that are expressed on the tumour but not on healthy cells. This project seeks to discover a range of potential tumour antigens and re-direct the immune system to eradicate brain tumours.

 

Alex Davenport

Krishneel Prasad

Krishneel Prasad is an Honours student at WEHI (the Walter and Eliza Hall Institute). He completed a Bachelor of Biomedicine at the University of Melbourne in 2021 where he undertook short projects in cancer-focused labs that fostered his passion to step into research. Complementing this, Krishneel enjoys communicating cancer science as part of the student run ‘Young Australian’s Cancer Initiative’. Today, with co-supervision by A/Prof Misty Jenkins, Dr Alexander Davenport and Dr Ryan Cross, he will investigate novel immunotherapeutic solutions for paediatric high-grade gliomas.

Project description
Diffuse midline glioma (DMG) is a lethal high-grade glioma that has seen minimal therapeutic advances in children for decades. Complications with its infiltration into brainstem structures has rendered current therapies largely ineffective, accounting for its poor prognosis. The Jenkins lab aims to utilise a new immunotherapeutic approach, termed CAR-T cell therapy, to re-engineer patient T cells to specifically kill brain cancers. Krishneel will utilise proteomic and transcriptomic profiles of DMG to identify novel CAR-T cell targets and validate their function in DMG models.

 

Krishneel Prasad

Murdoch Children's Research Institute

Dr Michael Assis

Michael Assis is a mathematical physicist, having received his PhD in statistical mechanics and enumerative combinatorics from Stony Brook University in 2014.

Afterwards he moved to the University of Melbourne for a research fellowship in computational statistical mechanics, followed by a research fellowship at the University of Newcastle in computational mathematics.

In 2021 Michael became the program manager of the Fertility Preservation Taskforce at the University of Melbourne and The Royal Children’s Hospital, joining MCRI in 2022 as a research assistant for a pharmacogenomics study on premature ovarian insufficiency. Aside from his interest in fertility research, he continues to be involved in mathematics, teaching at RMIT and La Trobe University in 2022.

Ms Sarah Grimshaw

Paddy Barry

Originally from Adelaide (SA), Paddy is undertaking Bachelor of Science (Honours) majoring in Neuroscience at the University of Melbourne and Murdoch Children’s Research Institute (MCRI). Paddy has previously interned at the Florey Institute with Anthony Hannan’s Laboratory. Working on a project studying the effects of SAPAP3 gene knockout in mice as a model for obsessive compulsive disorder (OCD). Paddy started with the Neuro-Oncology Lab in February 2023 with primary supervisors Prof David Eisenstat and Dr Maree Faux.

Project description

Neuroblastoma is the most common solid tumour of childhood excluding brain tumours. Sometimes, babies with this tumor show that it gets smaller on its own, especially if a certain gene isn’t amplified. There’s a protein called GMFb that might help with the tumor cells to grow out and become more like normal cells. This protein is also changed by adding a special molecule. We want to study this protein in the lab using tools like gene editing and microscopes. Figuring out how this protein works could help us find new treatments for this serious cancer in children.

Paddy Barry
Lane Collier

Lane Collier

Lane Collier completed her Bachelor of Biomedicine and Bachelor of Science (Honours) at Victoria University in December 2019.

Upon completing university, she worked at Department of Health in the COVID-19 Response team as a Senior Data Manager. In June 2022, she moved to Murdoch Children’s Research Institute (MCRI) to work with A/Prof Rachel Conyers and A/Prof David Elliott under the Australian Cardio-oncology Registry (ACOR) umbrella of research work. Lane is the ACOR National Project Manager and will be working on multiple research projects looking at the effect of cardio-toxic chemotherapy on heart health. In addition, she hopes to go back to university to complete her Doctor of Medicine and become an oncologist.

James Cooper

James Cooper is a recent biomedical graduate from the University of Adelaide with a deep passion for medical research. Throughout his Bachelor’s and Master’s degrees, James developed strong skills in molecular and cellular biology that he has continued to build upon in his subsequent research positions at the Centre for Cancer Biology (CCB) and Adelaide Health and Medical Sciences (AHMS) labs.

James’ interest in research stems from his love of science and his desire for discovery. He is particularly interested in the development of novel cell lines and their use in cutting-edge research in the field of paediatric oncology. James values collaboration and seeks opportunities to work closely with other passionate scientists. He strives to bring enthusiasm and a positive work ethic to the projects in which he is involved. Overall, James’ passion for medical research and his expertise in molecular and cellular biology make him an asset to any research team. He is committed to sharing his knowledge and contributing to the advancement of medical research.

Project description

James works to develop and characterise childhood solid/CNS tumour models, such as primary cell lines, organoids, and patient-derived xenograft models from paediatric tumours. These models will feed into various projects, including those within the MCRI Cancer Flagship, the Hudson-Monash Paediatric Precision Medicine Program (HMPPMP) and the Victorian Paediatric Cancer Consortium (VPCC). Additionally, he contributes to high-throughput drug screens, CRISPR-cas9 screens, and molecular biology techniques including DNA and RNA extractions. He will work closely with colleagues at Hudson Institute of Medical Research (HIMR) to further develop skills in cell line development and multi-omics analysis and to strengthen collaborations with HIMR, the Centre for Cancer Research (CCR), and the Next Generation Preciosn Medicine program.

James Cooper

Ben Felmingham

Ben Felmingham has focused his career in paediatric oncology pharmacy with 10 years experience in the Cancer Pharmacy at The Royal Children’s Hospital Melbourne.

In late 2021 he moved to the Murdoch Children’s Research Institute (MCRI) to work with A/Prof Rachel Conyers and A/Prof David Elliott under the Australian Cardio-oncology Registry (ACOR) umbrella of research work. He has since led the development of the first paediatric cardio-oncology guidelines and implemented and coordinates the multidisciplinary cardio-oncology clinic at The Royal Children’s Hospital, Melbourne.

Ben is now expanding his expertise into pharmacogenomics and will be working on multiple research projects looking at the effects of pharmacogenomic screening for potential gene-drug interactions, and thus providing and investigating therapeutic recommendations for actionable variants in the paediatric population.

Ben Felmingham
Liam Furst

Liam Furst

Liam Furst is a PhD student co-supervised by Prof David Eisenstat and Dr Maree Faux in the Murdoch Children’s Research Institute Neuro-oncology group. He completed a Bachelor of Science at the University of Toronto in 2019, and a Master of Biomedical Science at the University of Melbourne in 2021 under Dr Theo Mantamadiotis. His Masters work focussed on characterising the presence of cancer stem cells in paediatric high-grade glioma patient tissue, obtained in collaboration with the Children’s Hospital of Philadelphia.

Project description
Diffuse midline glioma (previously called DIPG) is a uniformly fatal paediatric cancer that arises in the brainstem. Due to the location of the tumour, treatment options are extremely limited, and ultimately the median survival is only 9-11 months post diagnosis. Approximately 80% of DMG tumours harbour H3K27M mutations and appear to resemble oligodendroglial progenitor cells (OPCs). The Eisenstat lab has previously shown that the DLX transcription factors directly repress other transcription factors necessary for differentiation towards an OPC lineage, and direct differentiation toward a GABAergic interneuron lineage. This project hypothesises that by altering DLX2 expression, DMG cell lines cells will have an altered cell fate leading to a reduction in tumourigenicity. This will be evaluated using xenograft animal models of DMG.

Ankita George

Ankita George completed her Bachelor of Science, majoring in Neuroscience from the University of Western Australia, which included an exchange year at the University of New Mexico. After which, she completed a stand-alone Bachelor of Science with Honours from the University of Melbourne, based at the Peter Doherty Institute for Immunity and Infection under Professor Fabienne Mackay.

Project description
Ankita George has worked as both a Research Assistant at the former Department of Anatomy and Neuroscience within the University of Melbourne as well as a Medical Scientist at the WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute. She then joined the Neuro-oncology Group at MCRI to work with both Professor David Eisenstat and Dr Maree Faux, where she will work across several projects looking at the links between DLX homeobox genes and Diffuse Midline Glioma (DMG), as well as other genetic diseases.

Ankita George

Brittany Grantham

Britt’s background is in allied health as a physiotherapist and research assistant. In 2022, she moved into a project officer role and completed a Master of Public Health. This led her to a clinical research coordinator role at the Murdoch Children’s Research Institute in the neuro-oncology group.

Project description
Britt’s role in the neuroncology group is on the radiogenomics project. There, she seeks to answer the following question: Can we identify those at risk for serious adverse effects following radiation therapy? The radiogenomics project will develop genetic assays to predict those patients at risk for radiation induced neurocognitive impairments and/or second malignant neoplasm (SMN).

Britta Grantham
Dhrita Khatri

Dhrita Khatri

Dhrita Khatri completed her Bachelor of Pharmacy with Honours in Auckland, New Zealand. Dhrita has 17 years of experience as a pharmacist with over 12 years’ experience in paediatric pharmacy. She has covered all aspects of pharmacy from management and operational aspects to clinical and even compounding roles. She has created, updated and reviewed several monographs, protocols and guidelines in her career. Dhrita completed her post-graduate Diploma in Clinical Pharmacy (Distinction) with the University of Tasmania and a short course with RMIT on Project Management for Professionals. Dhrita has always been passionate about research and is very excited to join the pharmacogenomics/discovery team at the Murdoch Children’s Research Institute (MCRI) led by A/Prof Rachel Conyers and A/Prof David Elliott.

Project description

She will be looking at the effects of pharmacogenomic screening for potential gene-drug interactions, and thus providing and investigating therapeutic recommendations for actionable variants in the paediatric population. Dhrita will also be leading a project looking at genotype informed Bayesian dosing of tacrolimus in solid organ transplant together with Dr David Metz.

 

Mr Ryan Leung

Ryan Leung

Ryan Leung was born and raised in Hong Kong, and has been living in Melbourne for seven years. Growing up, he always dreamt of studying veterinary science, however the excitement of research and lab work caught his attention during his undergraduate studies, and he decided to pursue a career in research instead. After completing his bachelor degree, Ryan did a Masters degree to gain some research experience in studying the importance of alternative splicing in malaria parasites, although his research was somewhat compromised due to COVID-19. Currently, he is undertaking a PhD in the neuro-oncology lab at Murdoch Children’s Research Institute, under the supervision of Professor David Eisenstat and Dr Maree Faux.

Project description
Diffuse intrinsic pontine glioma (DIPG) is highly aggressive and difficult to treat; ~80% of these tumours have histone H3 K27M mutations. Most DIPG cells resemble oligodendroglial progenitor cells (OPC). DLX homeobox genes are necessary for GABAergic interneuron differentiation and migration. DLX transcription factors directly repress genes required for OPC development. Through using advanced methods in molecular, cell and development biology, the project aims to identify the DLX2 gene regulatory network in the forebrain development, and to investigate the interaction of DLX2 and other transcription factors and its functions. By using RNAseq, the project will assess expression level of DLX2 and its target genes in DIPG tumours, to examine the relations between DIPG and brain development regulated by DLX2 and/or its target genes.

Jason Li

Jason graduated with a Bachelor of Science at the University of Melbourne in 2021. Subsequently, he graduated with first-class honours in a standalone Bachelor of Biomedical Sciences (Honours) from the University of Melbourne in 2022.
Throughout his honours, he investigated the enteric nervous system development to Hirschsprung Disease in children under the guidance of Prof David Eisenstat and Dr Maree Faux in the Neuro-Oncology laboratory. Currently, he is working as a Research Assistant in the Neuro-Oncology laboratory in the Murdoch Children’s Research Institute.

Project description
Hirschsprung Disease is a congenital birth defect presenting with a pseudo-obstruction of the intestine. Children with Hirschsprung Disease experience postoperative problems after surgery, including enterocolitis, constipation and faecal incontinence. These postoperative problems results in a decreased quality of life for the child. Jason’s project involves assessing the link between Dlx1/Dlx2 and Ret expression during intestinal development of embryonic mice and in human Hirschsprung Disease tissues. He hopes to further understand the link between the enteric nervous system and Hirschsprung Disease in children. He plans finish up his Hirschsprung Disease project from honours, publish his findings and assisting the laboratory’s day to day activities.

Jason LI
Morgan Marshall

Morgan Marshall

Morgan Marshall is an early career cancer researcher with a passion for understanding the developmental origins of disease. She completed her undergraduate degree majoring in Developmental Biology at Monash University, then moved to the Monash University Department of Biochemistry to begin her Honours and PhD studies under the supervision of Dr Kylie Wagstaff, Professor David Jans and Dr Kim Fung. Morgan’s research to date has focussed on finding and characterising the functions of proteins identified as targets of tumour-specific DNA aptamer KW16-13, which recognises Triple Negative Breast Cancer cell biomarkers for precise drug delivery.

Project description
Morgan has recently been appointed to a post-doctoral position at the Murdoch Children’s Research Institute (MCRI) as the recipient of the Lily Hester Early Career Cancer Research Award, thanks to the Live for Lily Foundation. She will join Professor David Eisenstat’s team in the Neuro-oncology group at MCRI to study the biochemical signalling mechanisms that regulate the distal-less (DLX) transcription factors in brain development and childhood brain cancers such as Diffuse Midline Glioma (DMG, formerly Diffuse Intrinsic Pontine Glioma (DIPG)). DMG is a devastating cancer that mainly affects children, usually aged between five and seven years old. It makes up 10-15 per cent of all brain tumours in children, with about 25 new diagnoses per year in Australia. Currently, there are no recognised treatment options for DMG, and current therapies are aimed to reduce symptoms vs. eradicating the tumours. Using proteomics and phosphoproteomics methodology Morgan’s overarching project aim is to find ways to use the newly discovered biochemical signalling mechanisms of DLX proteins to develop new therapies for aggressive paediatric cancers.

Claire Moore

Claire Moore is a PhD candidate at the Murdoch Children’s Research Institute (MCRI) under the supervision of A/Prof Rachel Conyers and A/Prof David Elliott. She is a clinical pharmacist who has worked at The Royal Children’s Hospital for over 15 years in a variety of roles including education and leadership. She has sub-specialised as a paediatric oncology pharmacist, obtaining skills in clinical trials and research, project work and quality improvement.
Claire graduated from LaTrobe University in 2006 (Bachelor of Pharmacy with Honours) and in 2018 completed a Master of Pharmacy Practice (Monash University). In 2021 she received a scholarship to study Principles of Healthcare Quality Improvement (Monash University).
Claire is the recipient of a strategic Melbourne University RTP scholarship to take part in a PhD as part of the Victorian Paediatric Cancer Consortium MRFF-funded grants. This project is additionally funded by The Kids’ Cancer Project.

Project description
Claire’s PhD project – ‘Priority-P – Pharmacogenomics’ is a randomised control trial implementing pre-emptive pharmacogenomic screening in paediatric oncology patients receiving polypharmacy. The primary endpoint of the study is to determine if pre-emptive knowledge of patients’ pharmacogenomic metaboliser states enables clinicians to make informed medication choices, and ultimately reduces adverse drug reactions. Priority-P will include a full health economics analysis using inpatient, outpatient and PBS data paired with quality of life studies to determine, in real terms, the benefits of a pre-emptive pharmacogenomic approach. In addition, Claire will co-design a paediatric pharmacogenomics education package for the stakeholders involved in delivering precision medicine within the Royal Children’s Hospital Children’s Cancer Centre. The project will enrol 440 patients across 3 years and be the first study of its kind internationally in paediatric oncology to prove the utility of pharmacogenomic intervention both at an ADR and health economic level.

Claire Moore
Tayla Stenta

Tayla Stenta

Tayla Stenta completed her Bachelor of Science at North Carolina State University in 2018, prior to an honours year at the University of Melbourne and Murdoch Children’s Research Institute under A/Prof Shireen Lamande and Prof John Bateman.

Her honours project and subsequent years as a research assistant focused on using induced pluripotent stem cells to model human genetic diseases. In particular, aiming to understand the genetic basis of TRPV4- associated skeletal dysplasias. In June 2022, she moved to the Heart Regeneration group at MCRI to work with A/Prof Rachel Conyers and A/Prof David Elliott, where she will work across multiple projects looking at the real time implementation of pharmacogenomics and a predictive approach to identifying infertility risks in paediatric cancer patients.

 

Peter MacCallum Cancer Centre

Dr Charlotte Chen

Dr Charlotte Chen

Charlotte Chen is a postdoctoral scientist in A/Prof Paul Neeson’s laboratory at Peter MacCallum Cancer Centre, working within the cancer immunology program. She completed her PhD at the Hudson Institute of Medical Research, Monash University, in 2020. From three-and-half years of study, she obtained experience in adult and paediatric brain tumour research. Her PhD project led to understanding the working mechanism and anti-tumour efficacy of a first-in-class small molecule inhibitor for specific therapeutic targeting of high-grade glioma. After completing her PhD, she had experience working in academia and industry and developed a strong interest in cancer immunology and immunotherapies. She joined A/Prof Neeson’s lab in 2022 and is currently undertaking research towards a deep understanding of the tumour microenvironment in paediatric cancers.

Project description
The therapeutic outcomes for paediatric cancers have not been appreciably improved for decades, especially for high-risk cancers. Among the novel therapeutic strategies, immunotherapies have been generating promising results. However, most of them are developed for adult cancer. Given that childhood cancers are biologically and molecularly distinct entities from adult cancer, the current main problem for immunotherapy relies on the lack of specific biomarkers or clinical grade assays to predict those childhood patient responses, which is particularly an issue when selecting patients who are most likely to benefit in clinical trials. A comprehensive understanding of the tumour immune microenvironment (TIME) will provide significant predictive value to the clinical management of those cancers and, importantly, will guide the further development of novel therapies. Our lab explores the human immune system responses to tumour development and immunotherapy using patient samples from cohort studies or clinical trials. With the collaboration of our VPCC partners, this project is a clinical trial investigating paediatric cancer TIME using advanced spatial multi-omics studies.

Fatimah Jalud

Fatimah Jalud

Fatimah Jalud earned her Bachelor of Science in Biology from De La Salle University in 2018, followed by the completion of her Masters in Biotechnology and Bioinformatics at La Trobe University in 2021. Her primary interest lies in applying molecular biology to gain insights into the genomic factors contributing to cancer development. Her aspiration is to leverage this knowledge to advance personalized medicine, making it not only accessible for treating cancer patients but also as a tool for genomic screening to aid in the early detection of cancer in the broader population. Fatimah became a research assistant in October 2021, joining the research team led by Associate Professor Paul Ekert at the Peter MacCallum Cancer Centre.

Project description
Fatimah works on several projects that focus on investigating the biological mechanisms that drive cancer formation, growth, resistance and relapse in paediatric Acute Lymphoblastic Leukaemia.

Dr Deborah Meyran

Dr Deborah Meyran is a paediatric oncologist who specialised in leukaemia and cellular therapies at The Royal Children’s Hospital (RCH), and a post-doctoral researcher in the Cancer Immunology Program at PeterMac Callum Cancer Centre (PMCC). Dr Deborah Meyran leads the collaboration between PMCC, RCH and CCI for the development of T cell-based immunotherapy with an emphasis on the generation of memory chimeric antigen receptor engineered T cells (CAR T cells) to treat paediatric cancers. Her career as a paediatric oncologist and her expertise in cancer immunotherapy have ranked her as a leader in the field of immunotherapy dedicated to children with cancer. As evidence of her international recognition and expertise, she received invitations to presentations at more than 20 international and national conferences. She has authored or co-authored 17 articles, with 90% of them published in high-rank journals. She has also received several awards including the best early researcher presentation at PMCC scientific retreat in 2022, and the Picchi Award for Excellence in Cancer Research that acknowledge the most talented PhD candidate in the VCCC (2020).

Project description

The outcome for patients with high-risk extracranial solid paediatric cancer such as MYCN-amplified neuroblastoma and sarcoma remains dismal. There is an urgent need to identify better treatments that do not suffer from detrimental long-term health effects such as those caused by currently used chemotherapies. There is considerable interest in advancing immunotherapies such as chimeric antigen receptor (CAR)-T cells towards the clinic for solid paediatric cancers. However, the remarkable success of CAR-T cells in childhood B-cell acute lymphoblastic leukaemia has so far not been recapitulated in solid child cancers. Important barriers for CAR-T cells in solid child cancers are: 1) the immunosuppressive tumour microenvironment (TME) present in many solid child cancers; 2) suboptimal CAR-T cell functioning.

For CAR-T cells to be effective in extracranial solid child cancers, novel strategies to enhance their proliferative capacity and long-term persistence must be combined with approaches to overcome TME immunosuppression. Our strategy is to first study the TME of solid tumours to target and reverse the immunosuppressive TME of solid tumours. Then we will combine a new generation of early-lineage CAR-T cells with TSTEM properties developed at PMCC with novel anti-cancer drugs currently tested at CCI, that inhibit tumour growth in preclinical models of paediatric cancers as well as rewire the immunosuppressive TME.  This project has the potential to shift the treatment paradigm for extracranial solid paediatric cancers towards implementation of combination CAR-T cell immunotherapy in standard of care.

 

Deborah Meyran

Lachlan McAloney

Lachlan McAloney is a PhD student at the Peter MacCallum Cancer Centre within the Cancer Immunology Program under the supervision of A/Prof Paul Ekert and Dr Teresa Sadras as well as Professor David Eisenstat from the Murdoch Children’s Research Institute. He holds a Bachelor of Science and a Bachelor of Biomedicine (Honours) from the University of Melbourne, completed 2017-2021. His honours project focused on using multiplex immunohistochemistry to define the tumour microenvironment of nestin-derived brain and germ cell tumours. He realised his passion for cancer immunology during his undergraduate studies and wanted the opportunity to complete his PhD candidature at the Peter MacCallum Cancer Centre.

Project description
Lachlan’s project aims to shed light on the molecular basis of the oncogenic function of a new class of fusion oncogenes which deregulate the expression of homeobox (Hox) genes normally expressed in early embryogenesis. This class of fusions have been seen in cases of acute lymphoblastic leukaemia (ALL) in paediatrics. He will focus on a group of previously uncharacterized Hox-fusions which were identified by his lab group (RUNX1-EVX1, ETV6-MNX1 and ETV6-CRX) with the end goal of discovering if this class of fusions can be therapeutically targeted.

Lachlan Maloney
Dr Teresa Sadras

Dr Teresa Sadras

Teresa Sadras is a molecular biologist with a longstanding research interest in the genetic, signalling and microenvironmental networks which are deregulated in blood cancers, and how these impact on disease progression, prognosis and response to therapy. After the completion of her PhD from the University of Adelaide in 2014, Dr Sadras undertook two post-doctoral positions, firstly at the South Australian Health and Medical Research Institute (SAHMRI), followed by four years at the Beckman Research Institute in Los Angeles in the laboratory of Dr Markus Muschen. During this time, Dr Sadras was awarded a Lymphoma Research Foundation Fellowship for her research in B-cell leukaemia and lymphoma. Teresa joined the research group of A/Prof Paul Ekert at the Peter MacCallum Cancer Centre in December 2020 as a senior post-doctoral scientist.

Project description
Acute Lymphoblastic Leukaemia (ALL) is the most common childhood cancer, and while advances in chemotherapy allow long-term survival rates approaching 85%, disease relapse remains one of the leading causes of cancer-related death in children and young adults. The work of Dr Sadras is focused on developing new models and experimental pipelines to investigate the biological mechanisms that drive cancer growth and therapy resistance in children with ALL.

Dr Carolyn Shembrey

Dr Carolyn Shembrey is a postdoctoral researcher working within the Cancer Immunology Program at the Peter MacCallum Cancer Centre. Carolyn completed her PhD at the University of Melbourne Centre for Cancer Research. By combining traditional wet lab approaches with computational biology, she aimed to interrogate the extent of natural killer cell involvement in metastatic colorectal cancer liver metastases and determine whether particular patient characteristics could be harnessed for innate immunotherapy. Carolyn complements her research skills with several years of experience in tutoring undergraduate pathology in the Department of Microbiology & Immunology at the University of Melbourne.

Project description
Neuroblastoma is the most common solid extracranial malignancy in children. Patients who present with amplification of the MYCN gene, encoding the transcription factor n-Myc, have particularly poor prognosis. Using novel CRISPR-based RNA engineering tools, Carolyn’s research focusses on developing personalised RNA-directed therapeutics to help target tumour drivers such as MYCN in this high-risk childhood cancer.

Dr Carolyn Shembrey

The Royal Children's Hospital

Diane Hanna

Dr Diane Hanna

Dr Hanna obtained her Bachelor of Medicine and Surgery (MBBS Hon.) from Monash University in 2007. She commenced paediatric training at The Royal Children’s Hospital in 2009 and became a Fellow of the Australasian College of Physicians (FRACP), Paediatrics and Child Health: Oncology in 2015. In 2021, she was awarded her Doctorate of Philosophy (PhD) through The University of Melbourne, under the supervision of Prof Andrew Roberts, A/Prof Paul Ekert, Dr Seong Lin Khaw and Dr Ashley Ng. Dr Hanna is currently a full-time paediatric oncologist at RCH, where she is an Australian clinical leader in paediatric leukaemia, haematopoietic stem cell transplant (HSCT) and cellular and novel therapies. Dr Hanna chairs the Victorian Paediatric Integrated Cancer Service (PICS) Leukaemia Multi-Disciplinary Meeting (MDM) and authored the oncology chapters of the 9th and 10th edition of the RCH’s Paediatric Handbook. Dr Hanna has built strong collaborations within the Victorian Comprehensive Cancer Centre Alliance as a steering group member for the Accelerate Novel Therapies Program in 2021. In 2021, she co-authored the first National Optimal Care Pathway (OCP) for children, adolescents and young adults with acute leukaemia, an initiative endorsed by the Australian Government. In 2022, Dr Hanna was selected to receive the Helen MacPherson Smith Trust Social Purpose Scholarship to undertake the Women in Senior Leadership (WISL) course at the Melbourne Business School.

Project description

Dr Hanna’s PhD, funded by The Kid’s Cancer Project, informed the clinical utility of BH3-mimetic (drugs targeting cell death) combinations in HR-ALL by identifying synergistic combinations with each other as well as standard and targeted agents in vitro, evaluated the efficacy and tolerability of combination venetoclax (BCL-2 inhibitor) and S63845 (MCL-1 inhibitor) in vivo, and investigated mechanisms of therapeutic resistance using an unbiased genome-wide CRISPR-Cas9 loss-of-function screen and mass cytometry. 

Dr Hanna is a passionate clinician-researcher who continues to integrate cutting-edge cancer research into clinical care to improve the outcomes for patients by building on what is known about the biology of treatment failure and leading appropriate clinical trials.  She has a strong faciliatory role in encouraging and leading research in the clinical environment as the Oncology Lead for the Research Innovation and Implementation Committee and Deputy Director of the Clinical Trials Unit (CTU) at the Children’s Cancer Centre (CCC) at RCH, where she is the local principle investigator (PI) for 10 international collaborative trials in childhood leukaemia. She holds honorary post-doctoral appointments at The Walter and Eliza Hall Institute (WEHI) and the Murdoch Children’s Research Institute (MCRI) as part of the broader Victorian Paediatric Cancer Consortium (VPCC) where she continues to study mechanisms of therapeutic resistance (collaborations with A/Prof Paul Ekert, A/Prof Meg Wall), understand toxicities of chemotherapy (collaborations with A/Prof Rachel Conyers) and HSCT (collaborations with A/Prof Gabrielle Haeusler, Dr Melanie Neeland, and A/Prof Theresa Cole) and build translational research into the use of novel therapeutics (including BH3-mimetics) to improve outcomes for children with poor prognosis acute leukaemias (collaborations with Prof David Ziegler and Dr Seong Lin Khaw).

Dr Lorna McLeman

Dr Lorna McLeman is a PhD student with the University of Melbourne at St Vincent’s Institute of Medical Research in the genome stability unit and a clinician paediatric oncologist trained at The Royal Children’s Hospital in Melbourne and Evelina Children’s Hospital in London. She is researching novel gene editing techniques to treat patients with Fanconi Anaemia. She has a special interest in bone marrow transplant and gene therapies, and a passion to continue her work as a clinician scientist in the field of gene editing and therapy.

Project description
Fanconi anaemia is the most common inherited bone marrow failure syndrome with a 700-fold increased risk of developing cancer. The phenotypes of Fanconi anaemia are caused by a genetic deficiency in one of 23 known FANC genes that normally suppress cancer-causing mutations. Accumulation of DNA damage in blood stem cells cause bone marrow failure in children, which can be salvaged by bone marrow transplant. However, transplant has a high risk of death for patients with Fanconi anaemia due to toxic side effects of chemotherapy used in transplant preparation and acceleration of cancer onset. Lorna’s PhD project is based across St Vincent’s Institute Genome Stability Unit, Murdoch Children’s Research Institute Blood Development Unit and The Royal Children’s Hospital, Melbourne under expert supervision of A/Prof Andrew Deans, Prof Andrew Elefanty and A/Prof Rachel Conyers. She will be investigating novel gene editing techniques with the aim of correcting Fanconi anaemia-causing mutations. Gene editing is a therapeutic approach to cure the bone marrow failure of Fanconi anaemia without the damaging effects of bone marrow transplant. The goal of this work is to provide the basis for a gene editing clinical trial for patients with Fanconi anaemia in Australia.

Dr Lorna McLeman
Dr Claudia Toro

Dr Claudia Toro

Dr Claudia Toro is a paediatric oncologist who is an associate investigator on the Australian Cardio-Oncology Registry study at The Royal Children’s Hospital (RCH) in Melbourne. She commenced her role at the RCH as the My Room Children’s Cancer Centre Clinical Research Fellow in February 2019 and joined the ACOR research team at that time. Dr Toro completed her medical degree at Flinders University in 2009.

Her passion for paediatric oncology led her to complete her training at the RCH, where she currently works as leukaemia/bone marrow transplant consultant. Her main interests are in supportive care and survivorship.

Project description
Recent advances in paediatric oncology have seen overall survival rates increase to over 85% for patients across the developed world. These improvements have led to a growing number of children now becoming long-term survivors. However, cancer therapy is not without consequence with many survivors developing long-term sequelae. One of the most significant causes of morbidity and mortality in survivors is cardiovascular disease. Currently, screening for cardiac dysfunction relies on the use of 2D echocardiography although novel approaches, such as the use of cardiac MRI, are emerging. The Extended Cardiac Evaluation Study (ECES), will aim to evaluate whether exercise and cardiac MRI can be used to uncover subclinical cardiotoxicity in paediatric oncology patients. Furthermore, through the work of the Australian Cardio-oncology Registry (ACOR), genetic variations that may predispose to cardiac dysfunction will be investigated.

Dr Hannah Walker

Dr Hannah Walker is a PhD candidate at Murdoch Children’s Research Institute, The Royal Children’s Hospital and Melbourne University, funded by a My Room Clinician fellowship. Hannah is also a paediatric oncologist at The Royal Children’s Hospital, with a special interest in bone marrow transplant and cellular therapies. She is also currently completing a fellowship year in apheresis at The Royal Children’s Hospital. She completed her postgraduate medical training at The University of Wollongong in 2013 and received The University of Wollongong Medal for the highest academic achievement.

Project description
Survival for children undergoing allogenic HSCT has improved dramatically over several decades through optimising supportive care and managing complications. Pulmonary complications post-HSCT continue to be devastating for children and their families and have very poor long-term outcomes. Better treatments are urgently required. To achieve this goal, we must understand the biological drivers and identify the patients at highest risk.

Hannah’s research project is entitled ‘BREATH; Breathe Easier After Allogeneic Transplantation; Haematopoietic. The project involves a prospective longitudinal study of children undergoing allogeneic bone marrow transplantation (BMT) as a cure for a range of malignant and non-malignant conditions. Patients undergoing general anaesthetic for part of their clinical care will have additional airway sampling and peripheral blood samples taken at multiple timepoints pre and post HSCT. This study involves immune and microbiological profiling of children pre and post BMT to understand what is occurring within the lungs and peripheral blood. The results of this study will allow analysis of inflammation in affected individuals prior to clinical detection, allowing identification of the very earliest changes associated with pulmonary complications. These results will identify early, potentially novel, biomarkers which could have several translational impacts. These include use as diagnostic biomarkers and as novel therapeutic targets. In particular, it is possible that existing treatments for inflammatory lung conditions, that have already proven to be safe in children, could be repurposed for use in pulmonary complications of allo-HSCT

Hannah Walker
Dr Stacie Wang

Dr Stacie Wang

Dr Stacie Wang is a paediatric oncologist at the Children’s Cancer Centre, The Royal Children’s Hospital. She grew up in Adelaide and moved to Melbourne for her medical studies at the University of Melbourne, obtaining her MBBS/BMedSci in 2009. She undertook basic paediatric training at The Royal Children’s Hospital (RCH) and Monash Children’s Hospital, prior to completing her fellowship in training in paediatric haematology/oncology at the RCH. During her fellowship, she cultivated a keen interest in immunotherapy, and became very interested in the concept of harnessing a patient’s own immune system to fight their own cancer. As a result, she undertook a PhD in applying immunotherapy to paediatric brain cancer at WEHI in 2018 and aims to become an active translational researcher and clinician-scientist in the future. Stacie is now working as a post-doctoral researcher at WEHI focusing on expanding the use of CAR-T cell therapy in paediatric cancer.

Project description
Stacie’s PhD focused on chimeric antigen receptor (CAR)-T cell therapy, a branch of immunotherapy that engineers a patient’s T cells to be able to specifically recognise and kill the cancer cells in their body. CAR-T cell therapy has been extremely successful in leukaemia but applying this revolutionary ‘fourth pillar’ of cancer therapy to brain cancers has been less successful to date. Although survival rates for childhood cancers overall have markedly improved over recent decades, outcomes in paediatric brain tumours have lagged behind the dramatic gains achieved in blood cancers. Stacie is working on a novel CAR-T cell that aims to kill a specific type of paediatric brain tumour which currently has no cure, diffuse intrinsic pontine glioma (DIPG).that engineers a patient’s T cells to be able to specifically recognise and kill the cancer cells in their body. CAR-T cell therapy has been extremely successful in leukaemia but applying this revolutionary ‘fourth pillar’ of cancer therapy to brain cancers has been less successful to date. Although survival rates for childhood cancers overall have markedly improved over recent decades, outcomes in paediatric brain tumours have lagged behind the dramatic gains achieved in blood cancers. Stacie is working on a novel CAR-T cell that aims to kill a specific type of paediatric brain tumour which currently has no cure, diffuse intrinsic pontine glioma (DIPG).

Monash Children's Hospital

Dr Caroline Lamont

Caroline Lamont is a Clinical Research Associate involved in precision medicine trials at the Monash Children’s Cancer Centre. She holds a Bachelor of Biomedical Science (Honours) and Master of Public Health, with a speciality in Health Economic Evaluation.

Caroline brings with her over 10-years of industry experience in adult and paediatric oncology research support, having worked previously in both management and senior laboratory roles with the state-wide Victorian Cancer Biobank, the Hudson Institute of Medical Research and the Monash Children’s Cancer Biobank.

She has co-authored papers published in Cancer Cell and Neuro-Oncology, and has presented at multiple events including the Australasian Biobanking Network Association and the Childhood Cancer Research Symposium.

Caroline Lamont
Leanne Super

Dr Leanne Super

Dr Leanne Super is an experienced Paediatric Oncologist with a special interest in AYA Oncology, sarcomas, Hodgkin lymphoma, histiocytosis.

Dr Super completed medical school at Monash University and trained in general paediatrics and oncology at The Royal Children’s Hospital and Monash Health, becoming a consultant across both sites in 2007. Dr Super has done further training at University College Hospital and Great Ormond Street in London. Dr Super is an affiliate lecturer at Monash University involved with undergraduate medical training, supervises basic and advanced trainees with the RACP, and is involved in clinical trials as PI and coPI for national and local studies in various types of oncology. Dr Super is a member of many medical groups locally and internationally.

Dr Nataliya Zhukova

Dr Nataliya Zhukova

Dr Nataliya Zhukova is an early career researcher in the Developmental and Cancer Biology lab and Hudson Institute of Medical Research. Dr Zhukova holds an Adjunct Research Fellow appointment in the Department of Paediatrics, School of Clinical Sciences at Monash Health, Faculty of Medicine Nursing and Health Sciences at Monash University, and is also a Paediatric Oncology Fellow at the Children’s Cancer Centre of the Monash Children’s Hospital. She was the recipient of a prestigious “My Room” Clinical Research Fellowship, awarded by the Children’s Cancer Foundation, which concluded in April 2022.

Dr Zhukova commenced a PhD in July 2022, with project title “Identification of molecular prognostic and therapy resistance signatures in paediatric sarcomas”. She has been awarded the Victorian Paediatric Cancer Consortium (VPCC) PhD Student Scholarship, sponsored by Monash University, to support her PhD. She has also been awarded an Early Career Practitioner Fellowship through the School of Clinical Sciences at Monash Health, Monash University, which also commenced in July 2022.

Project description
Dr Zukhova is focused on combining biological, clinical, and translational knowledge at the junction of paediatric oncology, molecular oncology, and cancer genetics. She studies molecular and genetic markers to understand molecular determinants driving tumourogenesis to improve risk-stratification and allow for augment/decrease of therapies to maximize survival and decrease short- and long-term side-effects. Malignant brain and solid tumours are among the most common types of cancer in children and adolescents after leukemias with cure rates remaining poor despite multimodal therapies. There is a lack of reliable tests to predict which patients are likely to respond well to treatment, and which will not. In her research she is using next generation sequencing techniques, including DNA methylation-based classification, proteomics, and immune environment profiling to identify subgroups of patients who are likely to respond to standard of care therapies and those who are predicted to relapse and thus require different treatment approaches. Comprehensive genomic and molecular analysis of patients’ samples and tumour derived cell lines and xenografts, complemented with functional genomics, will allow for identification of gene target candidates for novel therapies. This then will transition to animal model testing and subsequently to clinical trials with the ultimate goal of improving patients’ outcomes.

Children's Cancer Institute

Ms Chelsea Mayoh

Chelsea Mayoh

Chelsea Mayoh is a senior bioinformatician at the Children’s Cancer Institute and is also undertaking her PhD. Chelsea studied at the University of British Columbia in Canada and started working as a computational biologist at the Genome Sciences Centre in affiliation with the BC Cancer Agency before coming to Australia in 2015. She currently leads the bioinformatics team at CCI and is a key bioinformatician on the Zero Childhood Cancer program, having developed the pipelines for the transcriptome, methylome and in vitro drug response analysis. She is also currently undergoing her PhD part-time under the supervision of A/Prof Mark Cowley (computation biology team) and A/Prof Paul Ekert (translational biology team). Chelsea’s research interests are increasing the utility of bulk RNA-seq in precision medicine and the integration of multi-omics data for the identification of better targeted treatment options for high-risk paediatric patients.

Project description
Chelsea’s PhD focuses on the integration of whole genome sequencing (WGS), RNA sequencing (RNA-seq) and methylomics to identify targetable aberrations, molecular drivers, aid in the diagnosis and prognosis of disease, and exploring the immune microenvironment of high-risk paediatric patients in a precision medicine setting. Presently, precision medicine programs in paediatrics focus on the aberrations identified in either adult-based panels or from whole genome sequencing (WGS) and fusions from RNA-seq. However, expanding analysis beyond fusion detection in RNA-seq allows for the potential functional interpretation of the observed WGS aberrations and a deeper analysis into the transcriptome (ie alternate splicing events, aberrantly expressed genes, RNA specific point mutations) to identify additional actionable alterations. In addition, expanding the utility of bulk RNA-sequencing allows for exploration of the tumour microenvironment to identify immune-inflamed patients providing additional treatment options for these high-risk, high-need patients.

Andrea Zhao

Andrea is a current PhD candidate undertaking paediatric cancer research under A/Prof Paul Ekert’s supervision in the Translational Tumour Biology (TTB) group at Children’s Cancer Institute (CCIA). She graduated from a Bachelor of Medical Science with first-class honours at the University of Sydney in 2019. Her honours research project involved screening compounds in vitro in order to characterise drug-binding affinity and anti-cancer function against glioblastoma multiforme. After honours she became interested in pursuing a PhD in paediatric cancer research, based on her curiosity about paediatric cancer as well as the small number of studies available in comparison to adult cancers. Her interest in translational research led to joining TTB and acquiring a skill set in functional genomics.

Project description
The project will utilise CRISPR-Cas13b as a novel functional genomic tool to characterise aberrant fusion gene drivers in paediatric cancer. CRISPR-Cas13 is a new programmable RNA editing system with a higher target specificity than CRISPR-Cas9 and RNA interference systems. Unlike CRISPR-Cas9, the system is reversible with fewer off-target effects which ideally enables a more robust readout. The project will involve silencing oncogenic fusions with target-specific CRISPR-Cas13 guides. RNA sequencing and phosphoproteomics will be applied for a non-biased approach to driver characterisation and identification of novel molecular targets. Hit genes and proteins identified will be silenced with CRISPR-Cas13 in order to validate the gene dependency between the target and fusion driver. This system will serve as a widely applicable functional genomic tool to any oncogenic driver for characterisation and therapeutic development.

Ms Andrea Zhao