www.fgks.org   »   [go: up one dir, main page]

Centre for Cancer Research, 2023 Student Research Projects

Page 1

CENTRE FOR CANCER RESEARCH 2023 Student Research Projects


Contents Welcome to Hudson Institute

3

Centre for Cancer Research

4

Hudson Monash Paediatric Precision Medicine Program (HMPPMP)

5

Cancer Genetics and Functional Genomics 6 Developmental and Cancer Biology

8

STAT Cancer Biology

9

Ovarian Cancer Biomarkers

10

Immunohaematology

11

Structural Biology of Inflammation and Cancer 13 Leukaemia Modelling and Therapeutic Discovery

14

Contact our supervisors

15

Connect with us

15

The Translational Research Facility is connected via a link bridge to Monash Health and provides a crucial link between our scientific discoveries and medical treatments. The facility houses six worldleading technology platforms and an eight-bed, 21-chair Clinical Trials Centre that support the transition of discoveries from initial Phase I testing through to Phase IV primary health trials. Centre for Cancer Research | Student Research Projects 2023

2


Welcome to Hudson Institute Hudson Institute specialises in discoveries in five areas of medical need • Inflammation • Reproductive health and pregnancy • Infant and child health • Hormones and health Our 443 scientists and students focus on laboratory discovery science and translational research – taking discoveries to patients and industry for real-world impact.

STAFF

Join regular networking and learning and development programs, including the off-site Institute student retreat

Learn a range of dynamic and transferable skills for careers in the biomedical and clinical research sectors including commercialisation

All work and no play … Our students can join in a range of student networking and social events organised by Hudson Institute Student Society (HISS), including being part of the management committee.

Our precinct

• Cancer

267

176

45

257

STUDENTS

RESEARCH GROUPS

RESEARCH PUBLICATIONS

Hudson Institute is a leading Australian medical research institute recognised internationally for discovery science and translational research into inflammation, reproductive health and pregnancy, infant and child health, cancer, hormones and health. Our Institute is home to 443 world-class scientists, who push the boundaries of scientific knowledge to answer complex questions about human disease, including prevention and treatment. We are a founding member of the Monash Health Translation Precinct (MHTP) with partners Monash Health and Monash University. Our close ties with clinicians and industry enable us to translate our discoveries into new preventative approaches, therapies and devices for patients. Our location at Monash Medical Centre means our research is informed by patient need and our discoveries are transitioned into practical treatments.

Students at a glance 2021

68

176

26

POSTGRADUATE AND HONOURS STUDENTS COMPLETED

STUDENTS 125 PhD 1 MASTERS 50 HONOURS

STUDENTS WITH MEDICAL TRAINING

Working alongside clinicians in Melbourne hospitals for more than 50 years, our scientists pioneered IVF and stem cell discoveries and are now leading developments in cell therapies, paediatric cancer and the human microbiome. Our worldwide scientific and medical collaborations provide a foundation for transformative healthcare programs across the globe.

We educate and train more than 170 students through our academic affiliation with Monash University. Our postgraduate training is predominantly through the School of Clinical Sciences at Monash Health, part of the Faculty of Medicine, Nursing and Health Sciences at Monash University.

Our students •

Are exposed to university, institute, and hospital research

Attend national and international conferences

Publish their research in high impact journals

Are mentored by leading supervisors and their teams

Win prestigious prizes and awards

Centre for Cancer Research | Student Research Projects 2023

3


Centre for Cancer Research Location: Hudson Institute of Medical Research 27–31 Wright Street Clayton VIC 3168 t: +61 3 8572 2773 e: stephanie.forman@hudson.org.au w: hudson.org.au/research-centre/centre-forcancer-research/

Centre Head Prof Ron Firestein

Research Group Heads / Project Supervisors Hudson Monash Paediatric Precision Medicine Program (HMPPMP) Research Group Head Assoc/Prof Ron Firestein Cancer Genetics and Functional Genomics Research Group Head Assoc/Prof Ron Firestein Postdoctoral Scientist Dr Chunhua Wan Developmental and Cancer Biology Research Group Head Dr Jason Cain STAT Cancer Biology Research Group Head Dr Daniel Gough

Scientists working in the Centre for Cancer Research undertake basic research into the molecular mechanisms underlying the development, growth and metastasis of tumours, as well as the relationship between the innate immune system and cancer. The discovery and development of novel therapies for the treatment of cancers is also an important aspect of the team’s work. Current key areas of interest include:

Links between innate immunity, inflammatory processes and cancer‒ Role of embryonic signalling pathways in cancer, and the targeting of these pathways with novel therapies Cell signalling pathways involved in tumour survival and growth, and the development of monoclonal antibodies to treat glioma and other cancers

Ovarian Cancer Biomarkers Research Group Head Dr Andrew Stephens Research Scientist Dr Maree Bilandzic Immunohaematology Research Group Head Dr George Grigoriadis Research Group Head Dr Jim Vadolas Structural Biology of Inflammation and Cancer Research Group Head Dr Wilson Wong Leukaemia Modelling & Therapeutic Discovery

Role of integrin-linked kinase in cell migration and oncogenesis

Research Group Head Dr Catherine Carmichael

Molecular pathways involved in the metastasis of tumours, including colorectal, ovarian, prostate and bladder cancers

Cancer and Innate Immunity

Role of peptidase activity on inflammatory signalling and tumour microenvironment in ovarian cancer

Role of the microenvironment in tumour progression, chemoresistance and metastasis

Cancer precision medicine, including childhood brain cancer and solid tumours

Centre for Cancer Research | Student Research Projects 2023

Research Group Head Prof Bryan Williams Functional RNAomics Research Group Head Dr Minni Änkö

4


Hudson Monash Paediatric Precision Medicine Program (HMPPMP) Precision Medicine for Childhood Brain Cancer Suitability: PhD/Doctorate, Masters, Honours Project leader: Prof Ron Firestein e: ron.firestein@hudson.org.au Project description: The Hudson Monash Paediatric Precision Medicine program marks a significant investment in future clinical management and novel research discovery in childhood cancer. The program includes: The development of a living tumour biobank for paediatric solid tumours At present, very few reliable patient-derived preclinical models are available to researchers. To bridge this gap, our program will establish and bank organoid, cell lines, and xenograft models directly from childhood tumour tissue. The establishment of a living biobank for paediatric solid tumours will provide a critical renewable resource for local, national and international researchers. The establishment of a functional genomics pipeline We capitalise on the living biobank tumour samples to integrate genomic data (next generation sequencing) with functional data obtained from highthroughput genetic (Cas9/CRISPR) and results from global pharmacological drug screens. Translation of genomic data into targeted therapy The comprehensive molecular analysis of individual patient tumours will help identify both new and existing therapies that can be rapidly implemented in the clinic. This approach will facilitate clinical implications of data from the functional genomics pipeline for individual paediatric patients. Unique national and global collaborations The establishment of a living biobank and functional genomic testing for paediatric solid tumours provides a critical resource for local, national and international researchers. Thus, a key element of the program includes national and international stakeholders’ involvement to build expertise, share resources and disseminate results that will advance the field of precision medicine for paediatric cancer patients. Keywords: cancer, genetics, paediatrics, brain cancer, CRISPR, drug screens, genomics, personalised medicine, precision therapy

Centre for Cancer Research | Student Research Projects 2023

5


Cancer Genetics and Functional Genomics Functional genomic screens to identify new therapeutic targets for bowel cancer

KMT2A as a druggable therapeutic target against β-catenin-driven colorectal cancer Suitability: PhD/Doctorate, Masters, Honours, Short Projects Project leader: Dr Chunhua Wan

Suitability: PhD/Doctorate, Masters, Honours

e: chunhua.wan@monash.edu

Project leader: Prof Ron Firestein

Project description: Colorectal cancer (CRC) is the third most common cancer and the fourth leading cause of cancer-related death in the world. Australia leads the incidence rates of CRC worldwide that the lifetime risk of developing CRC is 1 in 13 among Australians. What’s worse, CRC was ranked the most (2016) and then the second most common (2017-2018) cause of cancer death in Australia, with an estimated death of 5,500 people annually. Unfortunately, due to lack of effective targeted therapy, the 5-year overall survival rate of CRC is limited to roughly 69%, lagging well behind other common cancers such as breast, melanoma and prostate (over 90% survival rate). Hence, there is a critical unmet need to develop new targeted therapies for colorectal cancer.

e: ron.firestein@hudson.org.au Project description: Bowel/colon cancer is a major cause of cancer related morbidity worldwide. We will use novel genomic technologies (e.g. CRISPR, shRNAs) to screen the cancer genome in an effort to identify novel therapeutic targets to colon cancer patients. Keywords: genetics, genomics, cancer, screen, personalised medicine

Development of new 3-dimensional models of cancer to model drug resistance and develop new cancer treatment Suitability: PhD/Doctorate, Masters, Honours Project leader: Prof Ron Firestein e: ron.firestein@hudson.org.au Project description: The development of clinically relevant cancer models that recapitulate human cancer is key to both understanding biological mechanisms of cancer growth as well fine tuning therapeutic cancer treatments. In this project, the student will work with both human tissues and animal models to develop 3-dimensional organotypic culture of genetically defined cancer models. Using CRISPR and other technologies we will genetically manipulate these models, and assess the contribution of new targets in mediating cancer growth. Keywords: colon cancer, organoids, models

Transcriptional regulators as cancer targets: new models and therapeutic approaches Suitability: PhD/Doctorate, Masters, Honours Project leader: Prof Ron Firestein e: ron.firestein@hudson.org.au Project description: Transcriptional regulators play a key role in activating oncogenic pathways that impinge on tumour growth, invasion and mestasasis. We study the role of Mediator kinases CDK8/19 as transcriptional regulators in normal tissue homeostasis and cancer (Sooraj et al., Molecular Cell 2022). Keywords: genetics, genomics, cancer, oncogenes, transcription Centre for Cancer Research | Student Research Projects 2023

Over 90% of CRCs arise from genetic mutations that activate a tumour-initiating pathway called Wnt/βcatenin pathway. Abrogating β-catenin function results in the loss of tumorigenic potential of Wntactive CRC cells. This makes β-catenin a most prominent treatment target in CRC. However, targeting β-catenin so far eludes clinical success, as pharmacologically blocking β-catenin remains technically infeasible. β-catenin mainly promotes the transformation of normal cells into cancer cells through activating the expression of various tumourinitiating genes. This process requires the recruitment of assorted transcriptional coactivators, especially epigenetic enzymes, and these epigenetic enzymes are ideal drug targets. Epigenetic enzymes are responsible for the reading of genetic code via introducing chemical modifications on DNA and its associated proteins, whose reprograming is indispensable for the initiation and progression of virtually all colorectal tumours. The recruitment of epigenetic enzymes is essential for β-catenin to initiate the expression of tumour-promoting genes and CRC development. Thus, targeting epigenetic enzymes that are essential for β-catenin function is a promising therapeutic strategy against CRC. However, the epigenetic enzymes that are selectively required for β-catenin function remain largely unknown. Methods and preliminary results: We employed a state-of-the-art gene editing tool (CRISPR-Cas9) to systemically investigate β-catenin-related therapeutic targets on a whole genome scale and validate the results using a confirmatory screen that specifically targeting epigenetic modifiers. Both screenings identified an epigenetic enzyme, namely lysine methyltransferase 2A (KMT2A), as a key player of β-catenin-mediated transcription. We have validated that the impacts of deleting KMT2A on β6


catenin targets are almost comparable to deleting βcatenin itself, suggesting that targeting KMT2A can diminish the activity of β-catenin in CRC cells. Furthermore, ablation of KMT2A or pharmacological inhibition of KMT2A led to evidently reduced expression of β-catenin targets and impaired colony and organoid formation in β-catenin-active CRC cells, but not in β-catenin-inactive cells. More importantly, β-catenin-active CRC cells exhibit significantly higher sensitivity towards KMT2A inhibitors that are under clinical development. These findings implicate KMT2A as an epigenetic enzyme highly specifically required for β-catenin function in whole human genome, highlighting the potential of KMT2A therapy as a CRC treatment. Expected outcomes: 1. The precise mechanisms underlying KMT2Afacilitated β-catenin function. 2. The unique role of KMT2A in the development of CRC. 3. The efficacy of pharmacologically inhibiting KMT2A on CRC treatment. Keywords: colorectal cancer; β-catenin; targeted therapy; epigenetics; translational medicine; cancer treatment; KMT2A

Targeting colorectal cancer stem cells using genome-scale CRISPR screens

Methods: The present project plans to establish a reporter system of CSCs in colorectal cancer cell lines and organoids (a 3D in vitro tissue culture system), and screen the genes that are selectively required for colorectal cancer stemness at a wholegenome scale. We will build reporter systems using fluorescence proteins (EGFP, RFP) to monitor key markers for cancer stemness (LGR5, ASCL2) and differentiation (KRT20, CEACAM1). A whole genome CRISPR library screen will be performed to identify genes whose knockout may cause the differenitation of CSCs. Following genome-wide screening, we will clarify the key druggable regulators that are required for the maintainance of CSCs using organoid and xenograft studies. The molecular mechanisms underpinning CSCs may also be investigated using biochemical approaches. Overall Goals: 1. Clarify the regulators of CSCs at a whole genome scale; 2. Identify of druggable targets of CSCs; 3. Investigate the translational merit of pharmaceutical inhibitors in colorectal cancer treatment. Keywords: colorectal cancer; translational medicine; cancer stem cells; CRISPR-Cas9 screen; druggable targets

Suitability: PhD/Doctorate, Masters, Honours, Short Projects Project leader: Dr Chunhua Wan e: chunhua.wan@monash.edu Project description: Colorectal cancer (CRC) is a most common cancer and the 4th leading cause of cancer-related death worldwide. Australia has one of the highest incidence rates of CRC in the world. According to Australian statistics, the lifetime risk of developing CRC in the general populationis 1 in 13. Due to lack of effectivetargeted therapy, the 5-year overall survival rate of CRC is roughly 69%, lagging well behind other common cancers such as breast, melanoma and prostate (over 90 % survival rate). Thus, the development of novel targeted therapies is a most urgent needin the fight against colorectal cancer. Colorectal cancer stem cells (CSCs) play a determinant role in colorectal cancer initiation and progression. The presence of LGR5+ CSCs is essential for colon tumors to grow and disseminate remotely. Elimination of CSCs causes colorectal cancer regression and long-term survival in experimental animals. Thus, finding the drugtargetable regulators that are selectively required for CSCs may pave the paths for novel targeted therapy of colorectal cancer. Our recent work has successfully uncovered the key drivers of wntinitiated colorectal cancer using genome-wide CRISPR screen (Chunhua Wan et al. Science Advances. 2021). This powerful strategy may dissect the regulators of CSCs at a whole genome level. Centre for Cancer Research | Student Research Projects 2023

7


Developmental and Cancer Biology Identification of targetable pathway dependencies in childhood sarcoma

epigenetic-mediated differentiation and apply these findings to a broader clinical context. Keywords: epigenetics, cancer, paediatrics, brain, lung

Identification of novel combination therapy for diffuse midline glioma Suitability: PhD/Doctorate, Honours

Suitability: PhD/Doctorate, Honours

Project leader: Dr Jason Cain

Project leader: Dr Jason Cain

e: jason.cain@hudson.org.au

e: jason.cain@hudson.org.au

Project description: Diffuse midline glioma is a highly aggressive cancer that arises in the midline brain structures of children and is universally fatal. Using next-generation sequencing strategies, significant advancement has been made in understanding the genetic profile of these tumours. Mutations in either of two genes encoding the Histone H3 protein converge on a critical lysine residue resulting in substitution with a methionine residue (K27M) have been described in the vast majority of DMG patients, suggesting a pathogenic role in this disease. The purpose of this project is to elucidate potential mechanisms of H3K27M tumorigenesis and likely therapeutic interventions that could be rapidly progressed into the clinic.

Project description: Soft tissue and bone sarcomas represent ~13% of all childhood cancer diagnosis and collectively are the second highest cause of childhood cancer related death, accounting for 20% of all mortalities. Despite the use of neoadjuvant chemotherapy and surgery, survival rates for these patients have remained stagnant for the last four decades. Curative treatment, effective in <70% of all sarcoma patients, leads to lifelong morbidity. For the remaining >30% there is no effective treatment. Whilst molecular markers of disease prognosis at diagnosis are revolutionising the clinical treatment and outcomes of other paediatric cancer types, this approach is largely lacking in childhood sarcoma. This highlights the urgent need for new and improved therapies for these diseases. In this project, we will utilize comprehensive functional and molecular datasets derived from patient-derived models to identify targetable sarcoma pathways. These pathways will be validated using clinically relevant cell and animal models of disease. The identification of targetable pathways underlying sarcoma growth and therapy resistance would represent a major development in the field and enable the future risk stratification of patients and appropriate application of targeted therapy to minimise side effects and improve overall survival.

Utilizing large functional and molecular datasets we will determine critical pathways required for DMG progression and therapy resistance. A combination approach to targeting selected pathways in clinically relevant DMG cell and animal models will inform on potential new therapeutic approaches that can be rapidly translated into clinical trials to improve patient outcomes. Keywords: DIPG, Histone H3, epigenetics, mouse models

Keywords: childhood cancer, sarcoma, cancer biology, osteosarcoma

Exploiting Epigenetic Dysregulation in SWI/SNF-Deficient Solid Tumours Suitability: PhD/Doctorate, Honours Project leader: Dr Jason Cain e: jason.cain@hudson.org.au Project description: Impaired differentiation is a common feature of cancer. We have recently demonstrated the differentiation potential of histone deacetylase inhibitors (HDACi) in paediatric (rhabdoid tumours) and adult (lung adenocarcinoma) solid tumours that are genetically defined by mutations in the SWI/SNF chromatin remodelling complex. Recent genomic studies have shown that mutations in subunits of this complex occur in at least 20% of all cancer. Using preclinical models of SWI/SNFdeficient and intact cancers, the successful candidate will investigate the mechanisms of Centre for Cancer Research | Student Research Projects 2023

8


STAT Cancer Biology

STAT3, the one transcription factor to rule them all Suitability: PhD/Doctorate

How does STAT3 enter the mitochondria? Suitability: PhD/Doctorate, Honours Project leader: Dr Daniel Gough e: daniel.gough@hudson.org.au Project description: The JAK-STAT3 signaling pathway is engaged by many cytokines and growth factor stimuli to control diverse biological processes including proliferation, angiogenesis, survival, immune modulation, and metabolism. For over two decades it has been accepted that STAT3-dependent biology is due to its potency as a transcription factor capable of regulating the expression of many hundreds of genes. However, recent evidence of noncanonical and non-genomic activities of STAT3 has emerged. The most exciting of these activities is its capacity to translocate into the mitochondria where it regulates the activity of the electron transport chain and the opening of the mitochondrial permeability transition pore. These have broad consequences including cell survival and the production of reactive oxygen species and ATP in both normal tissue and under pathological conditions. Despite these fascinating observations there are many key unanswered questions about the mechanism of STAT mitochondrial activity foremost of these is how does STAT3 get into the mitochondria. Keywords: STAT3, mitochondria, biochemistry

Leveraging the innate immune system to improve lung cancer treatments Suitability: PhD/Doctorate Project leader: Dr Daniel Gough e: daniel.gough@hudson.org.au Project description: Lung cancer is the leading cause of cancer related deaths worldwide. One of the greatest advances in cancer treatment has been the development of immune augmenting therapies that reactivate the anti-tumour T-cell response. However, despite their promise, around 80% of lung cancer patients do not respond or relapse following immunotherapy. What gets lost, or underappreciated in highlighting these approaches however, it the critical role of the innate immune system in the maturation and activity of the adaptive immune system. In this PhD project you will interrogate the potential of targeting the innate immune system to augment current lung cancer treatments. You will take advantage of sophisticated mouse models of cancer, functional genomics, proteomics, molecular biology and biochemistry to identify novel therapeutic strategies.

Project leader: Dr Daniel Gough e: daniel.gough@hudson.org.au Project description: STAT3, is a potent transcription factor that is essential for life. It is required to control the biological responses to many growth factors and cytokines. Whilst the basic mechanism by which STAT3 elicits is biological response has been known for 40 years, it is still entirely unclear how a single transcription factor can control opposing biological outcomes. A prime example of this is that IL-6 is a potent immune stimulator and IL-10 a potent immune suppressor, yet both require STAT3. New advances in technology have made it possible to address this critical question. In this PhD project you will use functional genomics, quantitative mass spectrometry, cell biology, biochemistry and animal models to define the importance of proteins recruited by STAT3 to sculpt its biological response. Keywords: STAT3, signal transduction, biochemistry, cancer, immunity

Targeting purine and pyrimidine synthesis to treat Small Cell Lung Cancer Suitability: Masters, Honours Project leader: Dr Daniel Gough e: daniel.gough@hudson.org.au Project description: Small cell lung cancer is an aggressive and highly metastatic disease that represents around 15% of all lung cancer patients. The majority of patients (70%) present in the clinic with advanced disease that has spread beyond the lung. The treatment options available to these patients are limited to platinum-based chemotherapy. This is effective in the majority of patients, however almost all will rapidly relapse with platinum resistant disease. There is no effective second line therapies which has meant these patients have an appalling overall survival rate of 25% which has not improved over the past three decades. We have developed mouse models of platinum-resistant small cell lung cancer and performed extensive RNA-sequencing and metabolomics analysis on the primary tumour tissue from these animals which has revealed a dramatic increase in purine and pyrimidine synthesis. Importantly we have shown that inhibitors of these pathways kill small cell lung cancer cell lines. This project will take advantage of our mouse models of small cell lung to investigate the efficacy of these agents in vitro and in vivo as well as their capacity to kill primary or metastatic tumours or both. Keywords: Small Cell Lung Cancer, therapy, mouse models of cancer

Keywords: lung cancer, immunotherapy, treatment

Centre for Cancer Research | Student Research Projects 2023

9


Ovarian Cancer Biomarkers

Keywords: photodynamic therapy, cancer, PDT, veterinary, chemoresistance, tumour

Identifying New Drug Targets in Ovarian Cancer Stem-Like Cells

Suitability: Honours

Suitability: PhD/Doctorate, Honours

e: maree.bilandzic@hudson.org.au

Project leader: Dr Andrew Stephens

Project description: We have identified novel molecules expressed during the early events of ovarian cancer invasion to healthy tissue. We hypothesize that these molecules are key to the metastatic process, and by specifically disrupting their expression we will disrupt the invasion process. This work will seek to develop new therapeutic strategies to block ovarian cancer metastasis and the formation of metastatic nodules.

e: andrew.n.stephens@hudson.org.au Project description: Ovarian cancers are the most lethal of all gynaecological malignancies, with <30% 5-year survival. Cancer progression requires cells to orchestrate a highly co-ordinated program of attachment, migration and invasion into healthy tissues. We have identified that a specialized subset of stem-like cancer cells, termed “Leader Cells”, control these processes in ovarian tumours. Leader cells are also enriched by chemotherapy and exert immune suppressive effects in vivo. Existing therapies do not kill or inhibit the leader cell population, resulting in their enrichment over time and ultimately leading to a poor prognosis for patients. We hypothesize that therapies targeting leader cells will synergize effectively with standard chemotherapy to achieve stable, long-term disease regression. This project will use a combination of molecular, biochemical and precision medicine approaches to investigate molecular pathways and identify “druggable” targets in ovarian cancer leader cells, and develop therapeutic strategies for translation into clinical practice. Keywords: cancer, stem cell, therapeutic, metastasis, ovarian, translation

Photodynamic Therapy for Cancer Treatment Suitability: PhD/Doctorate, Honours Project leader: Dr Andrew Stephens e: andrew.n.stephens@hudson.org.au Project description: Photodynamic therapy (PDT) uses light-sensitising agents to directly destroy tumour tissue and initiate anti-tumour immune responses. We have developed a series of novel photosensitising molecules and are currently progressing these through pharmaceutical approvals and phase I clinical trials.

Mechanisms of ovarian cancer metastasis- characterizing molecules expressed during early cancer invasion Project leader: Dr Maree Bilandzic

Keywords: cancer, ovarian cancer, metastasis

Preventing disease recurrence and increasing disease free survival rates for ovarian cancer patients Suitability: Honours Project leader: Dr Maree Bilandzic e: maree.bilandzic@hudson.org.au Project description: Majority of ovarian cancer patients are diagnosed with widespread disease on first clinical presentation. The initial response to chemotherapy and surgery is promising, however close to 90% develop recurrent, resistant disease at which point therapeutic options are limited. This project is focused on finding ways to prevent the spread of tumour cells following initial diagnosis and treatment, with the ultimate aim of increasing disease free lifespan and preventing disease recurrence in ovarian cancer patients. The project utilizes novel models of ovarian cancer metastasis to examine the early interactions between cancer cells and healthy tissue. You will investigate key changes that occur at the earliest point of ovarian cancer spread and devise ways to target molecules that drive the spread of ovarian cancer cells to stabilise disease, prevent tumour spread and recurrence. Keywords: ovarian cancer, cancer, therapies, treatment, real time modelling, 3D culture

This project will examine the use of two new compounds (INV043 and INV082) to treat ovarian and breast cancers, as a mechanism to improve response to checkpoint inhibition. The data will inform additional phase I human trials, and potentially also translation into veterinary medicine.

Centre for Cancer Research | Student Research Projects 2023

10


Immunohaematology Epigenetic modifications of the human βglobin locus: new therapeutic targets for haemoglobin disorders Suitability: PhD/Doctorate, Masters, Honours Project leaders: Dr Jim Vadolas, Dr George Grigoriadis e: jim.vadolas@hudson.org.au, george.grigoriadis@monash.edu Project description: Haemoglobin disorders, such as sickle cell disease and β-thalassaemia are the result of mutations in the adult β-globin gene. When these disorders are co-inherited with hereditary persistence of fetal haemoglobin, (high levels of γglobin gene expression in adult life) the disease phenotype is much reduced. Understanding the mechanism of γ-globin globin gene regulation through development has been the subject of intense investigation for many years. These studies led to an appreciation of the role of epigenetic modifications such as DNA methylation and histone acetylation in globin gene expression and regulation. Networks of regulatory proteins interact with epigenetic complexes to regulate DNA accessibility and histone modifications, thereby determining appropriate patterns of globin gene expression, giving rise to several developmental stage-specific hemoglobin variants. This study will investigate the potential impact of epigenetic regulators on globin gene expression. Functional genomic screening strategies will be performed using RNA interference (RNAi) or CRISPR/Cas9 genome editing to either suppress or knockout the expression of specific epigenetic regulators in erythroid cells modified to express fluorescent reporter genes under the control of the γ-globin promoter. Further studies will also be conducted in vivo using unique humanised βthalassaemia mouse models. Positive outcomes of such studies could pave the way for better treatment strategies for sickle cell anaemia and β thalassaemia patients by targeting epigenetic regulators to increase fetal globin expression. Keywords: epigentics, RNA interference, CRISPR/Cas9 genome editing

Harnessing RNA interference in gene therapy vectors for β-thalassaemia Suitability: PhD/Doctorate, Masters, Honours Project leaders: Dr Jim Vadolas, Dr George Grigoriadis e: jim.vadolas@hudson.org.au, george.grigoriadis@monash.edu Project description: The β-haemoglobin disorders such as β thalassaemia, haemoglobin E (HbE), and sickle cell disease (SCD) are among the most prevalent inherited disorders worldwide. The conditions are the result of mutations in the adult βCentre for Cancer Research | Student Research Projects 2023

globin gene, leading to production of either aberrant or insufficient β-globin protein. Symptoms appear in the first year of life, the period when fetal haemoglobin (HbF) is replaced by the adult form (HbA), leaving the patient dependent upon the mutated adult β-globin gene. Much of the pathology of this disease is due to excess α-globin chains forming toxic insoluble precipitates in erythroid cells resulting in cell death, ineffective erythropoiesis and severe anaemia. Interestingly, restoration of balanced globin protein synthesis through the reduction of α-globin expression can ameliorate the disease phenotype, exemplified by individuals who co-inherit α- and β-thalassaemia. This definitive observation forms the basis of a novel therapeutic strategy for β-thalassaemia, involving not an elimination but a targeted reduction of complementary α-globin chains, to mimic coinheritance of α- and β thalassaemia. While the benefits of increased β-globin expression in the context of β-thalassaemia are very clear, decreasing α-globin expression has not yet been extensively investigated. This project aims to develop novel gene therapy strategies harnessing RNAi in gene therapy vectors for β-thalassaemia. Initial studies will be conducted in vitro using both cell lines and primary haematopoietic stem cells. Further studies will also be conducted in vivo using our unique humanised βthalassaemia mouse models and patient-derived cells. Keywords: gene therapy, RNA interferance, anaemia

Impact of impaired immune function in haemoglobin disorders Suitability: PhD/Doctorate, Masters, Honours Project leaders: Dr Jim Vadolas, Dr George Grigoriadis e: jim.vadolas@hudson.org.au, george.grigoriadis@monash.edu Project description: Haemoglobin disorders, such as sickle cell disease and β-thalassaemia are the result of mutations in the adult β-globin gene. Patients suffering with the most severe form of the disease require chronic blood transfusion for survival. Ongoing transfusion therapy to counteract anaemia exacerbates iron overload, and necessitates iron chelation therapy. One important clinical feature of these conditions is the increased frequency of infectious complications such as pneumonia and sepsis, which are significantly associated with an increased rate of morbidity and mortality. The increased susceptibility to pathogenic organisms has been attributed to multiple deficiencies affecting both innate and adaptive immune systems. What has become apparent, is that iron overload in chronically anaemic patients contributes to aberrant neutrophil effector functions resulting in increased susceptibility to infection and inflammation-related organ damage. This knowledge, combined with the emergence of novel immunomodulatory function and phenotypes for neutrophils has helped to reinvigorate interest in the field. To further understand the clinical significance of aberrant immune function 11


in β-thalassaemia, we will undertake a comprehensive evaluation of the molecular and cellular mechanisms responsible for aberrant innate immune effector functions in β-thalassaemic mice and β-thalassaemia patients. The work proposed in this project will generate a better understanding of the mechanism underlying aberrant immune functions and provide novel insights into disease progression. Positive outcomes of such studies could pave the way for better treatment strategies for βthalassaemia and related patients. Keywords: thalassaemia, chronic anemia, immune response, iron overload

Centre for Cancer Research | Student Research Projects 2023

12


Structural Biology of Inflammation and Cancer Structural Biology of Inflammation Suitability: PhD/Doctorate, Honours Project leader: Dr Wilson Wong e: wilson.wong@hudson.org.au Project description: My laboratory utilises structural biology methods to investigate the structure and function of protein and protein-nucleic acid complexes important in inflammation and cancer. Students undertaking this project will learn a variety of experimental techniques and obtain expertise in protein chemistry, biochemistry and structural biology/cryo-EM, while contributing to research which will lead to new knowledge required for therapeutic designs. It is anticipated students will generate data that will contribute to manuscripts suitable for publication. Inflammation An important part of innate immunity is the ability of infected cells to sense and respond to infections by eliciting pro-inflammatory processes. Inflammasomes are key cytoplasmic sensors that detect pathogen components to undergo conversion from inactive to active signalling inflammasomes to drive inflammatory signalling. The activation status of inflammasomes is under tight-regulations to maintain healthy homeostasis, but mutations in inflammasome components can result in imbalance of inflammatory signalling which is the underlying basis of many autoinflammatory diseases. Our interest is focused on understanding how the activity of inflammasome is controlled and the mechanisms by which inflammasome sensors detect infections and the molecular details of inflammasome activation. In this project, the student will investigate the AIM2 inflammasome sensor and its binding with interacting partners and characterise the role of these interactions in AIM2 inflammasome activation. Specific aims will include characterisation on the mechanism of AIM2 oligomerisation and recruitment of core components of the inflammasome including ASC and caspase1 to form signalling inflammasome. Students will use protein expression and purification, functional biochemistry and structural biology method (cryo-EM) to understand AIM2 inflammasome signalling. Keywords: Cryo-EM, structural biology, biochemistry, inflammation, inflammasome, CryoEM, Cryo-electron microscopy

Centre for Cancer Research | Student Research Projects 2023

13


Leukaemia Modelling and Therapeutic Discovery Genetic modelling of Acute Erythroleukaemia Suitability: PhD/Doctorate, Masters, Honours Project leader: Dr Catherine Carmichael e: catherine.carmichael@hudson.org.au Project description: Background: Acute Erythroleukemia (AEL) is an aggressive and poor outcome subtype of AML that is largely resistant to standard treatments. Unlike other more common AML subtypes, very little is known of the genetic lesions that drive erythroid transformation, and there is a scarcity of in vivo genetic models in which to study AEL pathogenesis.

expression of EMT modulators has also been linked to the acquisition of cancer stem cell properties and enhanced therapeutic resistance in epithelial tumour cells. Intriguingly, we and others have more recently discovered that altered expression of EMT modulators, such as members of the SNAIL and ZEB families, also plays a role in the development and pathogenesis of haematological malignancies. Broadly targeting EMT processes therefore, is an attractive alternative therapeutic approach for both epithelial and haematopoietic tumours. Project: In this project, we will screen therapeutic compounds that putatively target regulators of EMT for their ability to inhibit malignant EMT processes, and negatively impact survival and migration capabilities of cancer cells – both epithelial and haematopoietic. Keywords: Epithelial to Mesenchymal Transition (EMT), leukaemia, tumour development, cancer metastasis, therapeutic targeting

We have recently contributed to the world’s first comprehensive genome wide analysis of the genetic lesions that define AEL (Iacobucci I et al, Nature Genetics, 2019), and now have the unique opportunity to generate much needed pre-clinical models that faithfully recapitulate the genetic landscape of the human disease. The project: Utilising novel genetic models of AEL, this project aims to study the molecular mechanisms driving transformation of the red blood cell lineage. Briefly, CRISPR/Cas9 gene editing approaches will be employed in both mouse and human cells to generate in vitro and in vivo models of AEL that faithfully recapitulate the underlying genetics identified in human AEL patients. Large scale genomic studies will be performed using these models to identify key mechanisms of erythroid transformation and AEL development. Finally, putative drug targets will be identified and tested in these model systems, with the ultimate aim of identifying novel therapeutic approaches for this very poor outcome malignancy. Keywords: erythroleukaemia, genetics, therapeutics, red blood cells, disease modelling, leukaemia

Therapeutic targeting of EMT modulators in cancer Suitability: PhD/Doctorate, Masters, Honours Project leader: Dr Catherine Carmichael e: catherine.carmichael@hudson.org.au Project description: Background: The epithelialmesenchymal transition (EMT) is a key developmental process that plays an important role during epithelial tumour development and pathogenesis. Activation of EMT in tumour cells contributes to the development of the migratory and invasive phenotype required for effective tumour cell metastasis. Furthermore, Centre for Cancer Research | Student Research Projects 2023

14


Contact our supervisors Students are encouraged to contact supervisors to discuss projects, arrange a time to visit the laboratories and view our facilities. Simply email the supervisor to arrange a time.

STEP 1: Find a project that you are interested in. STEP 2: Email the supervisor: “I am interested in your student project. Could I please arrange a time to visit you in your lab?”

All the information you need to enrol is on Hudson Institute’s website, or the project supervisor can help you to enrol. w: hudson.org.au/students/courses-available/

Keep up-to-date with our research news. Sign up for our e-newsletter at hudson.org.au/news/newsletters

Connect with us hudson.org.au HUDSONResearchAu @Hudson_Research Hudson-research hudson_research

27-31 Wright Street Clayton VIC 3168 Australia t: +61 3 8572 2700 w: hudson.org.au e: info@hudson.org.au

Centre for Cancer Research | Student Research Projects 2023

15


Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.