Profiles

Kate Brettingham-Moore

UTAS Home Doctor Kate Brettingham-Moore

Kate Brettingham-Moore

Lecturer in Pathology

Room 243-07 (Level 2) , Medical Science 2

+61 3 6226 4609 (phone)

+61 3 6226 7704 (fax)

Kate.BrettinghamMoore@utas.edu.au

Dr Kate Brettingham-Moore is a lecturer in the School of Medicine in the College of Health and Medicine. Her research involves molecular profiling of prostate cancer cells to identify drivers of treatment resistance and potential predictive biomarkers.

Biography

Kate obtained a PhD in molecular biology from the University of Tasmania in 2007. Her thesis investigated the molecular mechanisms involved in GM-CSF gene activation. During her post-doc at the Murdoch Children’s Research Institute in Melbourne (2007-2008), she examined the role of transcription in centromere formation. This was followed by a post-doc at the Peter MacCallum Cancer Centre in Melbourne (2008-2010) where she studied the potential of pre-treatment transcriptional biomarkers in predicting response to chemoradiotherapy. Kate returned to the University of Tasmania at the end of 2010 and is now a lecturer in the School of Medicine. Her research involves molecular profiling of prostate cancer cells to identify drivers of treatment resistance and potential predictive biomarkers.

Career summary

Qualifications

  • PhD, University of Tasmania, Australia, 2007, Defining the molecular events in GM-CSF gene activation
  • BSc (1st Class Hons), University of Tasmania, Australia, 2002, Chromatin remodelling of the GM-CSF promoter
  • BSc, University of Tasmania, Australia, 2001

Memberships

Professional practice

  • Australian Society for Molecular Biology and Biochemistry (ASBMB)- State Representative
  • Cancer Council Tasmania Scientific and Research Committee member

Teaching

Cell Biology, Biochemistry, Molecular Biology, Cancer Genetics, Epigenetics, Research Skills

Teaching expertise

Kate has teaching expertise in undergraduate molecular biology. She teaches cell biology and cancer genetics into the MBBS and is unit coordinator for the BMedRes unit, Human Genetics (CEA301). In addition, Kate was involved in designing and developing the Research Skills units (CAA209 and CAM103). She has supervised a number of Honours students (as primary supervisor) and PhD students (as co-supervisor).

Teaching responsibility

Research Invitations

External Reviewer NHMRC

Invited reviewer for a number of journals including BMC cancer & Journal of Experimental and Clinical Cancer Research.

View more on Dr Kate Brettingham-Moore in WARP

Expertise

  • Transcription
  • Cancer biology
  • Epigenetics
  • Radiotherapy response
  • Gene expression and chromatin biology
  • Genome structure and regulation

Research Themes

Kate's research aligns to the University's research theme of Better Health. Her research interests lie in personalised medicine and uncovering functionally relevant biomarkers to predict response to cancer therapies. Treatment resistance and disease recurrence are significant problems following chemo/radiotherapy for cancer patients. The development of predictive biomarkers would enable identification of patients unlikely to respond to treatment and help direct them towards better treatment options. In terms of searching for a predictor of response, the most successful predictive tests rely on the marker being functionally relevant and involved in the therapeutic mechanism. Current work is focussed on radiation resistance in prostate cancer and characterising the molecular differences in resistant versus sensitive cells.

Collaboration

Kate is involved in a number projects investigating molecular drivers of prostate cancer and leukaemia. She collaborates with Associate Professor Adele Holloway and Dr Phillippa Taberlay within the School of Medicine, along with collaborators from the Royal Hobart Hospital and University of Melbourne.

Awards

  • Cancer Council Tasmania/Chemist warehouse Emerging Researcher Scholarship 2012

Current projects

DNA methylation changes following radiotherapy in prostate cancer cells: investigating the epigenetic changes prostate cancer cells experience following radiotherapy and the implications these may have in terms of treatment response.

Fields of Research

  • Biochemistry and cell biology (310199)
  • Cancer genetics (321103)
  • Epigenetics (incl. genome methylation and epigenomics) (310504)
  • Cancer cell biology (321101)
  • Molecular targets (321108)
  • Gene expression (incl. microarray and other genome-wide approaches) (310505)
  • Radiation therapy (321110)
  • Genome structure and regulation (310508)
  • Medical biochemistry - nucleic acids (320505)
  • Medical bacteriology (320701)
  • Cellular immunology (320404)
  • Genomics (310509)
  • Oncology and carcinogenesis (321199)
  • Genetics (310599)
  • Bioinformatic methods development (310201)
  • Cellular interactions (incl. adhesion, matrix, cell wall) (310105)
  • Solid tumours (321111)
  • Predictive and prognostic markers (321109)
  • Medical biochemistry and metabolomics (320599)
  • Cell development, proliferation and death (310102)

Research Objectives

  • Clinical health (200199)
  • Expanding knowledge in the environmental sciences (280111)
  • Expanding knowledge in the health sciences (280112)
  • Expanding knowledge in the biological sciences (280102)
  • Treatment of human diseases and conditions (200105)
  • Efficacy of medications (200102)
  • Expanding knowledge in the biomedical and clinical sciences (280103)
  • Diagnosis of human diseases and conditions (200101)

Publications

Total publications

26

Journal Article

(16 outputs)
YearCitationAltmetrics
2019Sutton LP, Jeffreys SA, Phillips JL, Taberlay PC, Holloway AF, et al., 'DNA methylation changes following DNA damage in prostate cancer cells', Epigenetics, 14, (10) pp. 989-1002. ISSN 1559-2294 (2019) [Refereed Article]

DOI: 10.1080/15592294.2019.1629231 [eCite] [Details]

Citations: Scopus - 16Web of Science - 13

Co-authors: Sutton LP; Jeffreys SA; Phillips JL; Taberlay PC; Holloway AF; Ambrose M; Young A; Berry R

Tweet

2018Phillips JL, Taberlay PC, Woodworth AM, Hardy K, Brettingham-Moore KH, et al., 'Distinct mechanisms of regulation of the ITGA6 and ITGB4 genes by RUNX1 in myeloid cells', Journal of Cellular Physiology, 233, (4) pp. 3439-3453. ISSN 0021-9541 (2018) [Refereed Article]

DOI: 10.1002/jcp.26197 [eCite] [Details]

Citations: Scopus - 10Web of Science - 10

Co-authors: Phillips JL; Taberlay PC; Woodworth AM; Dickinson JL; Holloway AF

Tweet

2015Brettingham-Moore KH, Taberlay PC, Holloway AF, 'Interplay between transcription factors and the epigenome: insight from the role of RUNX1 in leukemia', Frontiers in Immunology, 6 Article 499. ISSN 1664-3224 (2015) [Refereed Article]

DOI: 10.3389/fimmu.2015.00499 [eCite] [Details]

Citations: Scopus - 23Web of Science - 22

Co-authors: Taberlay PC; Holloway AF

Tweet

2015Patchett AL, Latham R, Brettingham-Moore KH, Tovar C, Lyons AB, et al., 'Toll-like receptor signaling is functional in immune cells of the endangered Tasmanian devil', Developmental and Comparative Immunology, 53 pp. 123-133. ISSN 0145-305X (2015) [Refereed Article]

DOI: 10.1016/j.dci.2015.07.003 [eCite] [Details]

Citations: Scopus - 15Web of Science - 14

Co-authors: Patchett AL; Latham R; Tovar C; Lyons AB; Woods GM

Tweet

2015Qadi A, Taberlay PC, Phillips JL, Young A, West AC, et al., 'The leukemia inhibitory factor receptor gene is a direct target of RUNX1', Journal of Cellular Biochemistry, 117, (1) pp. 49-58. ISSN 0730-2312 (2015) [Refereed Article]

DOI: 10.1002/jcb.25246 [eCite] [Details]

Citations: Scopus - 6Web of Science - 5

Co-authors: Taberlay PC; Phillips JL; Young A; West AC; Dickinson JL; Holloway AF

Tweet

2014Young A, Berry R, Holloway AF, Blackburn NB, Dickinson JL, et al., 'RNA-seq profiling of a radiation resistant and radiation sensitive prostate cancer cell line highlights opposing regulation of DNA repair and targets for radiosensitization', BMC Cancer, 14 Article 808. ISSN 1471-2407 (2014) [Refereed Article]

DOI: 10.1186/1471-2407-14-808 [eCite] [Details]

Citations: Scopus - 26Web of Science - 25

Co-authors: Young A; Berry R; Holloway AF; Blackburn NB; Dickinson JL; Phillips JL

Tweet

2013Tran TN, Brettingham-Moore KH, Duong CP, Mitchell C, Clemons NJ, et al., 'Molecular changes in the phosphatidylinositide 3-kinase (PI3K) pathway are common in gastric cancer', Journal of Surgical Oncology, 108, (2) pp. 113-120. ISSN 0022-4790 (2013) [Refereed Article]

DOI: 10.1002/jso.23357 [eCite] [Details]

Citations: Scopus - 11Web of Science - 11

Tweet

2012Poke FS, Upcher WR, Sprod OR, Young A, Brettingham-Moore KH, et al., 'Depletion of c-Rel from cytokine gene promoters is required for chromatin reassembly and termination of gene responses to T cell activation', PLoS One, 7, (7) Article e41734. ISSN 1932-6203 (2012) [Refereed Article]

DOI: 10.1371/journal.pone.0041734 [eCite] [Details]

Citations: Scopus - 5Web of Science - 5

Co-authors: Poke FS; Sprod OR; Young A; Holloway AF

Tweet

2011Brettingham-Moore KH, Duong C, Greenawalt DM, Heriot AG, Ellul J, et al., 'Pretreatment transcriptional profiling for predicting response to neoadjuvant chemoradiotherapy in rectal adenocarcinoma', Clinical Cancer Research, 17, (9) pp. 3039-3047. ISSN 1078-0432 (2011) [Refereed Article]

DOI: 10.1158/1078-0432.CCR-10-2915 [eCite] [Details]

Citations: Scopus - 45Web of Science - 42

Tweet

2011Brettingham-Moore KH, Duong CP, Heriot AG, Thomas RJS, Phillips WA, 'Using gene expression profiling to predict response and prognosis in gastrointestinal cancers-the promise and the perils', Annals of Surgical Oncology, 18, (5) pp. 1484-1491. ISSN 1068-9265 (2011) [Refereed Article]

DOI: 10.1245/s10434-010-1433-1 [eCite] [Details]

Citations: Scopus - 25Web of Science - 20

Tweet

2011Pilgrim CHC, Brettingham-Moore KH, Pham A, Murray W, Link E, et al., 'MRNA gene expression correlates with histologically diagnosed chemotherapy-induced hepatic injury', HPB, 13, (11) pp. 811-816. ISSN 1365-182X (2011) [Refereed Article]

DOI: 10.1111/j.1477-2574.2011.00365.x [eCite] [Details]

Citations: Scopus - 10Web of Science - 10

Tweet

2009Chueh AC, Northrop EL, Brettingham-Moore KH, Choo KHA, Wong LH, 'LINE retrotransposon RNA is an essential structural and functional epigenetic component of a core neocentromeric chromatin', PLoS Genetics, 5, (1) Article e1000354. ISSN 1553-7390 (2009) [Refereed Article]

DOI: 10.1371/journal.pgen.1000354 [eCite] [Details]

Citations: Scopus - 126Web of Science - 127

Tweet

2008Brettingham-Moore KH, Sprod OR, Chen X, Oakford PC, Shannon MF, et al., 'Determinants of a transcriptionally competent environment at the GM-CSF promoter', Nucleic Acids Research, 36, (8) pp. 2639-2653. ISSN 0305-1048 (2008) [Refereed Article]

DOI: 10.1093/nar/gkn117 [eCite] [Details]

Citations: Scopus - 17Web of Science - 14

Co-authors: Sprod OR; Oakford PC; Holloway AF

Tweet

2007Wong LH, Brettingham-Moore KH, Chan L, Quach JM, Anderson MA, et al., 'Centromere RNA is a key component for the assembly of nucleoproteins at the nucleolus and centromere', Genome Research, 17, (8) pp. 1146-1160. ISSN 1088-9051 (2007) [Refereed Article]

DOI: 10.1101/gr.6022807 [eCite] [Details]

Citations: Scopus - 212Web of Science - 213

Tweet

2006Shannon MF, Chen X, Brettingham-Moore KH, Holloway AF, 'Chromatin remodelling: Distinct molecular events during differentiation and activation of T cells', Current Immunology Reviews, 2, (3) pp. 273-289. ISSN 1573-3955 (2006) [Refereed Article]

DOI: 10.2174/157339506778018587 [eCite] [Details]

Citations: Scopus - 1

Co-authors: Holloway AF

Tweet

2005Brettingham-Moore KH, Rao S, Jeulich T, Shannon MF, Holloway AF, 'GM-CSF promoter chromatin remodelling and gene transcription display distinct signal and transcription factor requirements', Nucleic Acids Research, 33, (1) pp. 225-234. ISSN 0305-1048 (2005) [Refereed Article]

DOI: 10.1093/nar/gki161 [eCite] [Details]

Citations: Scopus - 31Web of Science - 28

Co-authors: Holloway AF

Tweet

Chapter in Book

(1 outputs)
YearCitationAltmetrics
2015Brettingham-Moore KH, Taberlay PC, 'Cancer Epigenetics', Drug Discovery in Cancer Epigenetics, Academic Press, G Egger & P Arimondo (ed), United Kingdom, pp. 41-62. ISBN 978-0128022085 (2015) [Research Book Chapter]

[eCite] [Details]

Co-authors: Taberlay PC

Tweet

Conference Publication

(9 outputs)
YearCitationAltmetrics
2005Brettingham-Moore KH, Ray SN, Holloway AF, 'A role for the BAF remodelling complex in GM-CSF gene regulation', ComBio 2005 Combined Conference Abstracts, 25-29 Sep 2005, Adelaide, pp. 51. (2005) [Conference Extract]

[eCite] [Details]

Co-authors: Ray SN; Holloway AF

2005Brettingham-Moore KH, Shannon MF, Holloway AF, 'GM-CSF gene activation; providing an insight into the role of BRG1 in gene regulation', Mechanisms of Eukaryotic Transcription, 31 Aug - 4 Sep 2005, pp. 44. (2005) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2005Brettingham-Moore KH, Shannon MF, Holloway AF, 'Transcription factors and chromatin remodelers involved in activating the GM-CSF gene', The Genome Conference, 13-17 Feb 2005, Victoria, pp. 101. (2005) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2004Brettingham-Moore KH, Holloway AF, 'A role for the NF-KB family member, Crel, in chromatin remodelling events at the GM-CSF promotor', ComBio 2004, 26-30 Sept 2004, Perth, pp. 82. (2004) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2004Brettingham-Moore KH, Shannon MF, Holloway AF, 'GM-CSF promotor remodeling and gene transcription in response to T cell activation are distinct events', The Genome Conference, 15-19 Feb 2004, Lorne, Vic (2004) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2004Brettingham-Moore KH, Shannon MF, Holloway AF, 'Distinct signals and factors are required for GM-CSF promotor chromatin remodelling and gene transcription', ComBio 2004, 26-30 Sept 2004, Perth, pp. 46. (2004) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2003Brettingham-Moore KH, Rao S, Shannon MF, Holloway AF, 'Disecting the molecular events involved in activation of GM-CSF gene expression in T cells', 24th Annual Conference on the Organisation and Expression of the Genome, 16-20 Feb 2003, Lorne, Victoria, pp. Poster 1-25. ISBN 0-9585259-0-0 (2003) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2003Brettingham-Moore KH, Shannon MF, Holloway AF, 'Defining the molecular events required for GM-CSF gene activation in T cells', ComBio 2003 Combined Conference Abstracts, 9 Sept-2 Oct 2003, pp. 161. (2003) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

2002Holloway AF, Rao S, Brettingham-Moore KH, Shannon MF, 'Selective remodelling of a single nucleosome precedes GM-CSF transcription in T cells', Proceedings of the Australian Society for Biochemistry and Molecular Biology, 29 September - 3 October, 2002, Sydney, pp. 4. ISSN 1328-4924 (2002) [Conference Extract]

[eCite] [Details]

Co-authors: Holloway AF

Grants & Funding

Funding Summary

Number of grants

13

Total funding

$433,626

Projects

Examining DNA hydroxymethylation as a radiosensitisation target in medulloblastoma (2021)$11,000
Description
Radiotherapy is frequently used in treating brain cancers such as medulloblastoma. Unfortunately, outcomes are poor for many patients. This study will investigate the potential of targeting DNA hydroxymethylation to sensitise medulloblastoma cells to radiotherapy. Aim 1 will profile levels of hydroxymethylation while Aim 2 will assess the impact of TET inhibition on response to radiotherapy.
Funding
Cancer Council of Tasmania ($11,000)
Scheme
Grant-Small
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH; Taberlay PC; Holloway AF
Year
2021
Identifying shared mechanisms in response to radiotherapy (2020)$18,980
Description
Radiotherapy remains one of the most frequently utilised therapies in cancer treatment today. However, despite the frequency of its use, many patients experience treatment resistance and disease recurrence. It is therefore essential to identify predictive markers of response that would enable patients unlikely to benefit from radiotherapy to be identified and spared an ineffective treatment and its associated side effects. This project will use an in vitro model to identify shared drivers of resistance and potential predictive biomarkers between cancers originating from different sites (prostate vs brain). It will involve RNA-seq, proteomics profiling and analysis of 3D spheroids in response to radiotherapy.
Funding
Cancer Council of Tasmania ($18,980)
Scheme
Grant-Small
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH; Azimi I; Wilson RR
Year
2020
Gaining new insights into metastatic bone tumours (2020)$24,925
Description
Molecular analysis of prostate cancer tumour specimen to identify new molecular targets for therapy.
Funding
Royal Hobart Hospital Research Foundation ($24,925)
Scheme
Grant-Project
Administered By
University of Tasmania
Research Team
Dickinson JL; Holloway AF; Nott LM; Liu G; FitzGerald LM; Brettingham-Moore KH; Hewitt A; Taberlay PC
Year
2020
The impact of repeated DNA damage on the molecular profile of prostate cancer cells (2019)$23,295
Description
Many cancer patients receive fractionated/recurrent dosages of DNA damaging agents and there is some evidence that this may drive future treatment resistance and metastasis. This project will profile prostate cancer cells after repeated DNA damage via RNA-seq and EPIC arrays
Funding
Royal Hobart Hospital Research Foundation ($23,295)
Scheme
Grant-Project
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH; Black A; Taberlay PC; Holloway AF
Year
2019
Mapping epigenomic information in a three-dimensional prostate cancer cell environment (2017)$20,990
Description
We lack a model that better replicates in vivo tumour development to study epigenomic mechanisms of prostate cancer. To date, epigenetic changes in cancer have been investigated in homogenous cell lines from two-dimensional laboratory cultures or end-point tumour samples (static) from patients. Without doubt these studies have provided great insight into epigenetic differences that distinguish cancer cells from normal cells. However, there is a glaring lack of information about dynamic and temporal changes to the epigenome and cell behaviour as the cells acquire invasive potential. This is not possible using currently available two-dimensional models and clinical samples. We will address this deficit by establishing a three-dimensional model of prostate cancer that allows us to map how epigenomes and cell behaviours change during cancer development.
Funding
Cancer Council of Tasmania ($20,990)
Scheme
Grant-Cancer Research
Administered By
University of Tasmania
Research Team
Taberlay PC; Brettingham-Moore KH; Holloway AF; Biro M
Year
2017
Developing CRISPR/Cas Library Screens for identifying Novel Cancer Therapies (2017)$63,845
Description
Our understanding of many cancers has improved dramatically over the past decade predominantly due to our ability to sequence entire genomes, at scale. Yet, we still require a better understanding of the underlying mechanisms that initiate and perpetuate cancers, as well as gene-based factors that initiate the transition from indolent to aggressive cancers with a propensity to metastasize. CRISPR/Cas is proving a robust, powerful and necessary tool in the laboratory that will undoubtedly underpin the next breakthrough in the field of cancer. As such, it is essential that we develop this capability at the University of Tasmania in a timely manner.The specific AIMS of this proposal are to:Aim 1: Establish CRISPR/Cas screening at the University of Tasmania as a tool to identify genesthat drive aggressive and metastatic cancers.Aim 2: Perform a genome-wide CRISPR/Cas negative selection (loss-of-function) screen toidentify genes essential for proliferation and survival, as well as metastatic behaviour.
Funding
University of Tasmania Foundation Inc ($63,845)
Scheme
Grant-Cancer Research
Administered By
University of Tasmania
Research Team
Taberlay PC; Hewitt A; Holloway AF; Liu G; Dickinson JL; Brettingham-Moore KH; Fitzgerald L; Eri RD; Cook AL
Year
2017
David Collins Leukaemia Foundation Elite Research Scholarship - Epigenetic and Transcriptional regulation of leukaemia (2016 - 2019)$153,000
Description
This project will investigate how epigenetic and transcription factors co-operate to control gene expression, how this is reprogrammed in leukaemia and how epigenetic drugs impact these gene expression programs.
Funding
David Collins Leukaemia Foundation ($153,000)
Scheme
Donation - Individual
Administered By
University of Tasmania
Research Team
Holloway AF; Dickinson JL; Brettingham-Moore KH
Period
2016 - 2019
Molecular profiling of the post radiotherapy chromatin landscape in prostate cancer cells (2016)$33,000
Description
This project will investigate the basal and post radiotherapy epigenetic changes in prostate cancer cells with divergent response to radiotherapy (RT). It will validate methylation data obtained from illumina Infinium 450 beadchip arrays, profile histone modifications post RT and investigate DNA damage post RT in cells treated with inhibitors of epigenetic enzymes (DNMT and HDAC).
Funding
Cancer Council of Tasmania ($33,000)
Scheme
Grant-Cancer Research
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH; Ambrose M; Holloway AF; Skala M; Taberlay PC
Year
2016
Investigating molecular determinants of radiation response in prostate cancer cells (2015)$11,550
Funding
University of Tasmania ($11,550)
Scheme
Grant-Research Enhancement (REGS)
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH
Year
2015
Epigenetic regulation of inegrin beta 4 as a predictor of prostate cancer progression (2015)$14,750
Description
Prostate cancer diagnoses continue to rise rapidly in Australia, but of most concern is ourcurrent inability to distinguish aggressive tumours with propensity to metastasize from moreindolent disease. This results in unnecessary treatment of many men whose cancer may neverprogress to clinically significant disease. However, progression of the disease results in aggressive, metastatic tumours which are difficult to treat and are associated with poor prognosis. We thus urgently require a better understanding of the underlying drivers of this disease, and particularly the factors and mechanisms that drive the transition to more aggressive tumours with a propensity to spread. These factors may represent biomarkers and potential therapeutic targets in prostate cancer progression.The aims of this study are therefore:1.To determine whether epigenetic changes at the ITGB4 gene are associated with its increased expression in prostate cancer progression, and 2. To investigate whether epigenetic changes at the ITGB4 gene in prostate cancer are driven by the RUNX transcription factors.
Funding
Cancer Council of Tasmania ($14,750)
Scheme
Grant-Cancer Research
Administered By
University of Tasmania
Research Team
Holloway AF; Dickinson JL; Brettingham-Moore KH; Rao S; Black A
Year
2015
Regulation of integrins by RUNX1 in leukaemia (2013)$30,000
Funding
David Collins Leukaemia Foundation ($30,000)
Scheme
Grant
Administered By
University of Tasmania
Research Team
Holloway AF; Dickinson JL; Brettingham-Moore KH
Year
2013
Role of NF-kB in radiosensitivity and resistance in prostate cancer (2012)$19,200
Funding
Cancer Council of Tasmania ($19,200)
Scheme
Grant-Cancer Research
Administered By
University of Tasmania
Research Team
Brettingham-Moore KH; Dickinson JL; Skala M
Year
2012
Transcriptome analysis for predicting response to radiotherapy in prostate cancer patients (2011)$9,091
Funding
Royal Hobart Hospital Research Foundation ($9,091)
Scheme
Grant-Research
Administered By
University of Tasmania
Research Team
Dickinson JL; Brettingham-Moore KH; Skala M; Holloway AF; Stankovich J
Year
2011

Research Supervision

Kate is currently co-supervising 3 PhD students and has been primary supervisor for 4 completed Honours students.

Current

1

Completed

2

Current

DegreeTitleCommenced
PhDEpigenetic Evolution During Cancer Treatment2020

Completed

DegreeTitleCompleted
PhDTranscriptomic and DNA Methylation Alterations in Prostate Cancer Metastasis
Candidate: Aparna Shree Raina
2021
PhDGene Regulation by RUNX1 in the Absence of Consensus Sequences
Candidate: Alexandra Morgan Woodworth
2020