Research Program Overview

In the Translational Oncology Laboratory, we are developing innovative new therapies for cancers including glioblastoma, melanoma and cancers of the lung and pancreas, as well as novel approaches to better guide the use of existing therapies. Much of our research is collaborative, working in close association with the Cancer Clinical Trials Unit at the Royal Adelaide Hospital, and partnering with other clinical sites, research laboratories and industry partners around Australia and internationally.

We are a large group with diverse interests, but unified by a common focus: the rapid translation of our research to the clinic, to enable our discoveries to contribute to better patient outcomes as soon as possible. Our two major research streams are described below.

Cancer Immunotherapy Group

The Cancer Immunotherapy group focuses on novel therapies that enhance a cancer patient’s immune system, to enable the patient’s own T cells to target and destroy their tumour. Such approaches include chimeric antigen receptor (CAR)-T cells, bi-specific T cell engagers (BiTE) and immune checkpoint inhibitors (ICI). Our main focus is on brain tumours (glioblastoma in adults and diffuse midline glioma in children) and melanoma.

Antibody Targeting

This team uses functionalised monoclonal antibodies, including antibody-drug conjugates (ADCs) and antibodies labelled with radioactive isotopes, to target tumour cells for therapeutic and diagnostic purposes.

Current Research Projects

Cancer Immunotherapy Group

Pre-clinical development of CAR-T cell and BiTE therapies

CAR-T cell and BiTE therapies are revolutionising the treatment of certain blood cancers, which has spurred intense interest in extending these successes to the treatment of solid tumours. We have a pre-clinical program to develop CAR-T cell and BiTE therapy for solid cancers, with a particular focus on aggressive brain cancers (glioblastoma and diffuse midline glioma). We use patient-derived tumour and blood cells, as well as advanced mouse models and a novel tumour organoid system, to develop, optimise and test CAR-T cells for their ability to control tumour growth. Research programs in this area focus on identifying novel target antigens; improving the homing of CAR-T cells to tumour tissues; testing combination therapies; and optimising CAR-T cell survival and function.

CAR-T cell clinical trials

Our clinical program manufactures CAR-T cells targeting the GD2 tumour antigen here in Adelaide, using a protocol optimised through our pre-clinical research program. The GD2 molecule is expressed by many solid tumour types but has limited expression on healthy cells and tissues, making it an excellent CAR-T cell target. We have completed recruitment to a 12-patient trial of GD2-specific CAR-T cell therapy for patients with GD2-positive malignancies including melanoma and sarcoma (www.anzctr.org.au: ACTRN12613000198729).

Now, we have two active phase 1 clinical trials testing these GD2-targeting CAR-T cells in cancer patients. In the KARPOS trial (ACTRN12622001514796) at the Royal Adelaide Hospital, GD2-CAR-T cells are administered to patients with glioblastoma whose tumour has recurred despite previous surgery, chemotherapy, and radiotherapy. In the LEVI’S CATCH trial (ACTRN12622000675729) these CAR-T cells are given to children with brain tumours through a collaboration with Prof David Ziegler at the Sydney Children’s Hospital and Children’s Cancer Institute.

Understanding and predicting patient responses to Immune Checkpoint Inhibitor (ICI) therapy

ICI therapy is a new therapeutic approach that is now approved in Australia for the treatment of several cancer types, including melanoma, lung, and kidney cancers. These medicines can re-activate dormant anti-tumour immune responses, leading to dramatic tumour shrinkage, and possibly cure, in a fraction of patients. However, many patients receive little to no benefit, yet are still exposed to the risk of severe side effects. Using blood and tumour samples from melanoma and lung cancer patients, we are investigating the immune responses that underpin successful clinical outcomes following ICI therapy. This research may identify novel strategies to improve response rates, and is being used to develop a simple blood test to help guide the optimal treatment for each patient.

Exploring the microenvironment of brain tumours
Solid tumours don’t just contain cancerous cells, but are also infiltrated with a complex network of immune cells, blood vessels and other cell types. We have a research program focussed on understanding these cellular ecosystems in brain tumour patients, using techniques including single cell RNA sequencing, spatial transcriptomics, and high-parameter flow cytometry. These insights will allow us to optimise our immunotherapies to function optimally within challenging tumour microenvironments.

Antibody Targeting Group

First-line therapy for lung cancer typically involves cytotoxic chemotherapy, which is DNA-damaging and causes cancer cell death. We have preclinical proof of concept for a novel method of detecting cancer cell death using the APOMAB® monoclonal antibody that is specific for the essential La ribonucleoprotein overexpressed in malignancy.

We are currently investigating the following applications of APOMAB®:

  • Antibody-drug conjugates (ADC) for bystander treatment of solid cancers: We are investigating ADC versions of APOMAB based on the premise that APOMAB-ADC-bound dead tumour cells are processed by both viable tumour cells and supporting leucocytes to produce bystander killing.
  • Preclinical and clinical development of an imaging agent for detection of cancer cell death:  We have adapted the APOMAB® monoclonal antibody for use in immuno-positron emission tomography (PET). Non-invasive methods for detecting cancer cell death can be useful for the early evaluation of therapeutic responses. In this respect, a 20-patient phase 1 proof-of-concept immunoPET/CT trial has recently been completed at RAH (ACTRN12620000622909). APOMAB was radiolabelled with the diagnostic isotope Zirconium-89 (89Zr). We found that its tumour uptake was greatest after prior treatment using cytotoxic chemotherapy or radiotherapy had induced cancer cell death.
  • Using ADCs to stimulate anti-tumour immune responses in models of cancer: We are using ADCs alone and in combination with immunotherapy agents to both directly kill tumour cells and to modulate the body’s immune system to target and eliminate any remaining resistant cancer cells more effectively.
  • Preclinical and clinical development of an APOMAB-CAR-T cell therapy: We have commenced preliminary experiments investigating the notion that APOMAB binding to dead cancer cells after a first step of standard anti-cancer treatment can provide a way of activating T cells.

Laboratory staff

Laboratory head

Group Leader (Cancer Immunotherapy Group)

  • Associate Professor Lisa Ebert

Team Members

Postdoctoral Research Scientists

Research Assistants

Students

  • Erica Yeo


  • Bryan Gardam


  • Eunwoo (Chris) Nam


  • Resty Nabeeta


  • Ali Nazarizadeh


Select Recent Publications

Cancer Immunotherapy Group

  1. Yu W*, Ebert LM*, Truong N, Polara R, Gargett T, Yeo E, Brown MP (2023). Endogenous bystander killing mechanisms enhance the activity of novel FAP-CAR-T cells against glioblastoma. Preprint in BioRxiv: DOI: https://doi.org/10.1101/2023.02.21.529331 *equal contribution
  2. Gargett T, Ebert LM, Truong NTH, Kollis PM, Sedivakova K, Yu W, Yeo ECF, Wittwer NL, Gliddon BL, Tea MN, Ormsby R, Poonnoose S, Nowicki J, Vittorio O, Ziegler DS, Pitson SM, Brown MP (2022). GD2-targeting CAR-T cells enhanced by transgenic IL-15 expression are an effective and clinically feasible therapy for glioblastoma. J Immunother Cancer. 10(9):e005187
  3. Kollis PM, Ebert LM, Toubia J, Bastow CR, Ormsby RJ, Poonnoose SI, Lenin S, Tea MN, Pitson SM, Gomez GA, Brown MP, Gargett T (2022). Characterising distinct migratory profiles of infiltrating T-cell subsets in human glioblastoma. Front Immunol; 13:850226.
  4. Ebert LM, Yu W, Gargett T, Toubia J, Kollis PM, Tea MN, Ebert BW, Bardy C, van den Hurk M, Bonder CS, Manavis J, Ensbey KS, Oksdath Mansilla M, Scheer KG, Perrin SL, Ormsby RJ, Poonnoose S, Koszyca B, Pitson SM, Day BW, Gomez GA, Brown MP (2020). Endothelial, pericyte and tumor cell expression in glioblastoma identifies fibroblast activation protein (FAP) as an excellent target for immunotherapy. Clin Transl Immunology. 9(10):e1191.
  5. Brown MP, Ebert LM, Gargett T (2019). Clinical chimeric antigen receptor-T cell therapy: a new and promising treatment modality for glioblastoma. Clin Transl Immunology; 8:e1050.
  6. Gargett T, Truong N, Ebert LM, Yu W, Brown MP (2019). Optimization of manufacturing conditions for chimeric antigen receptor T cells to favor cells with a central memory phenotype. Cytotherapy; 21:593-602.
  7. Perrin SL, Samuel MS, Koszyca B, Brown MP, Ebert LM*, Oksdath M*, Gomez GA* (2019). Glioblastoma heterogeneity and the tumour microenvironment: implications for preclinical research and development of new treatments. Biochem Soc Trans; 47(2):625-638. * Equal corresponding authors
  8. Ebert LM, Yu W, Gargett T, Brown MP (2018). Logic-gated approaches to extend the utility of chimeric antigen receptor T-cell technology. Biochem Soc Trans. 46:391

Antibody Targeting Group

  1. Wittwer NL, Staudacher AH, Liapis V, Cardarelli P, Warren H, Brown MP (2023). An anti-mesothelin targeting antibody drug conjugate induces pyroptosis and ignites antitumor immunity in mouse models of cancer. J Immunother Cancer. 11(3): e006274.
  2. Staudacher, A.H.*, Li, Y.*, Liapis, V. and Brown, M.P. (2022) The RNA-binding protein La/SSB associates with radiation-induced DNA double-strand breaks in lung cancer cell lines. Cancer Reports. Aug;5(8):e1543. doi: 10.1002/cnr2.1543. (*shared first authorship).
  3. Reid, P., Staudacher, A.H., Marcu, L.G., Olver, I., Moghaddasi, L., Brown, M.P., and Bezak, E. (2021). Characteristic differences in radiation-induced DNA damage response in HPV negative and HPV positive head and neck cancers with accumulation of fractional radiation dose. Head and Neck 46:10: 3086-3096
  4. Liapis, V., Tieu, W., Wittwer, N.L., Gargett, T., Evdokiou, A., Takhar, P., Rudd, S.E., Donnelly, P.S., Brown M.P. and Staudacher, A.H. (2021). Positron Emission Tomographic imaging of tumor cell death using Zirconium-89-labeled chimeric DAB4 following cisplatin chemotherapy in lung and ovarian cancer xenograft models.  Molecular Imaging and Biology, https://doi.org/10.1007/s11307-021-01640-x
  5. Liapis, V., Tieu, W., Rudd, S.E., Donnelly, P.S., Wittwer, N.L., Brown M.P. and Staudacher, A.H. (2020). Improved non-invasive positron emission tomographic imaging of chemotherapy-induced tumor cell death using Zirconium-89-labeled APOMAB®.  EJNMMI Radiopharmacy and Chemistry 5(27)
  6. Staudacher, A.H.*, Liapis, V.*, Tieu, W., Wittwer, N.L. and Brown M.P. (2020). Tumour-associated macrophages process drug and radio-conjugates of the dead tumour cell targeting APOMAB antibody. Journal of Controlled Release 327(10): 779-787, (*both authors contributed equally)  
  7. Staudacher, A.H., Li, Y., Liapis, V., Hou, J., Chin, D., Dolezal, O., Adams, T.E., van Berkel, P.H.  and Brown M.P. (2019).  APOMAB® antibody drug conjugates targeting dead tumor cells are effective in vivo.  Molecular Cancer Therapeutics 18(2): 335-345.
  8. Staudacher, A.H., Liapis, V. and Brown M.P. (2018). Therapeutic targeting of tumor hypoxia and necrosis with antibody α-radioconjugates.  Antibody Therapeutics 1:2 75-83.