Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 12
  • Item
    Thumbnail Image
    CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity
    Burr, ML ; Sparbier, CE ; Chan, Y-C ; Williamson, JC ; Woods, K ; Beavis, PA ; Lam, EYN ; Henderson, MA ; Bell, CC ; Stolzenburg, S ; Gilan, O ; Bloor, S ; Noori, T ; Morgens, DW ; Bassik, MC ; Neeson, PJ ; Behren, A ; Darcy, PK ; Dawson, S-J ; Voskoboinik, I ; Trapani, JA ; Cebon, J ; Lehner, PJ ; Dawson, MA (NATURE RESEARCH, 2017-09-07)
    Cancer cells exploit the expression of the programmed death-1 (PD-1) ligand 1 (PD-L1) to subvert T-cell-mediated immunosurveillance. The success of therapies that disrupt PD-L1-mediated tumour tolerance has highlighted the need to understand the molecular regulation of PD-L1 expression. Here we identify the uncharacterized protein CMTM6 as a critical regulator of PD-L1 in a broad range of cancer cells, by using a genome-wide CRISPR-Cas9 screen. CMTM6 is a ubiquitously expressed protein that binds PD-L1 and maintains its cell surface expression. CMTM6 is not required for PD-L1 maturation but co-localizes with PD-L1 at the plasma membrane and in recycling endosomes, where it prevents PD-L1 from being targeted for lysosome-mediated degradation. Using a quantitative approach to profile the entire plasma membrane proteome, we find that CMTM6 displays specificity for PD-L1. Notably, CMTM6 depletion decreases PD-L1 without compromising cell surface expression of MHC class I. CMTM6 depletion, via the reduction of PD-L1, significantly alleviates the suppression of tumour-specific T cell activity in vitro and in vivo. These findings provide insights into the biology of PD-L1 regulation, identify a previously unrecognized master regulator of this critical immune checkpoint and highlight a potential therapeutic target to overcome immune evasion by tumour cells.
  • Item
    Thumbnail Image
    Tumor-derived exosomes modulate T cell function through transfer of RNA
    House, IG ; Petley, EV ; Beavis, PA (WILEY, 2018-03)
    Tumor cells can develop a variety of mechanisms to evade and subvert the immune system for their survival. Bland et al., in this edition of The FEBS Journal, make the novel finding that the tumor line B16F0 can deliver mRNA/miRNA loaded exosomes to cytotoxic T lymphocytes and alter their metabolic function and interferon gamma production.
  • Item
    Thumbnail Image
    A2A blockade enhances anti-metastatic immune responses
    Beavis, PA ; Milenkovski, N ; Stagg, J ; Smyth, MJ ; Darcy, PK (TAYLOR & FRANCIS INC, 2013-12)
    The specific targeting of tumor-elicited immunosuppression is a promising strategy for the treatment of cancer. We have recently demonstrated that targeting the immunosuppressive pathway mediated by CD73-derived adenosine through the blockade of A2A/A2B adenosine receptors significantly reduced the metastatic potential of CD73+ breast carcinomas and melanomas via both immunological and non-immunological mechanisms.
  • Item
    Thumbnail Image
    Relevance of tumor-infiltrating lymphocytes in breast cancer
    Dushyanthen, S ; Beavis, PA ; Savas, P ; Teo, ZL ; Zhou, C ; Mansour, M ; Darcy, PK ; Loi, S (BMC, 2015-08-24)
    While breast cancer has not been considered a cancer amenable to immunotherapeutic approaches, recent studies have demonstrated evidence of significant immune cell infiltration via tumor-infiltrating lymphocytes in a subset of patient tumors. In this review we present the current evidence highlighting the clinical relevance and utility of tumor-infiltrating lymphocytes in breast cancer. Retrospective and prospective studies have shown that the presence of tumor-infiltrating lymphocytes is a prognostic marker for higher responses to neoadjuvant chemotherapy and better survival, particularly in triple negative and HER2-positive early breast cancer. Further work is required to determine the immune subsets important in this response and to discover ways of encouraging immune infiltrate in tumor-infiltrating lymphocytes-negative patients.
  • Item
    Thumbnail Image
    Agonist immunotherapy restores T cell function following MEK inhibition improving efficacy in breast cancer
    Dushyanthen, S ; Teo, ZL ; Caramia, F ; Savas, P ; Mintoff, CP ; Virassamy, B ; Henderson, MA ; Luen, SJ ; Mansour, M ; Kershaw, MH ; Trapani, JA ; Neeson, PJ ; Salgado, R ; McArthur, GA ; Balko, JM ; Beavis, PA ; Darcy, PK ; Loi, S (NATURE PUBLISHING GROUP, 2017-09-19)
    The presence of tumor-infiltrating lymphocytes in triple-negative breast cancers is correlated with improved outcomes. Ras/MAPK pathway activation is associated with significantly lower levels of tumor-infiltrating lymphocytes in triple-negative breast cancers and while MEK inhibition can promote recruitment of tumor-infiltrating lymphocytes to the tumor, here we show that MEK inhibition adversely affects early onset T-cell effector function. We show that α-4-1BB and α-OX-40 T-cell agonist antibodies can rescue the adverse effects of MEK inhibition on T cells in both mouse and human T cells, which results in augmented anti-tumor effects in vivo. This effect is dependent upon increased downstream p38/JNK pathway activation. Taken together, our data suggest that although Ras/MAPK pathway inhibition can increase tumor immunogenicity, the negative impact on T-cell activity is functionally important. This undesirable impact is effectively prevented by combination with T-cell immune agonist immunotherapies resulting in superior therapeutic efficacy.MEK inhibition in breast cancer is associated with increased tumour infiltrating lymphocytes (TILs), however, MAPK activity is required for T cells function. Here the authors show that TILs activity following MEK inhibition can be enhanced by agonist immunotherapy resulting in synergic therapeutic effects.
  • Item
    Thumbnail Image
    Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity
    Davenport, AJ ; Cross, RS ; Watson, KA ; Liao, Y ; Shi, W ; Prince, HM ; Beavis, PA ; Trapani, JA ; Kershaw, MH ; Ritchie, DS ; Darcy, PK ; Neeson, PJ ; Jenkins, MR (NATL ACAD SCIENCES, 2018-02-27)
    Chimeric antigen receptor T (CAR-T) cells are effective serial killers with a faster off-rate from dying tumor cells than CAR-T cells binding target cells through their T cell receptor (TCR). Here we explored the functional consequences of CAR-mediated signaling using a dual-specific CAR-T cell, where the same cell was triggered via TCR (tcrCTL) or CAR (carCTL). The carCTL immune synapse lacked distinct LFA-1 adhesion rings and was less reliant on LFA to form stable conjugates with target cells. carCTL receptors associated with the synapse were found to be disrupted and formed a convoluted multifocal pattern of Lck microclusters. Both proximal and distal receptor signaling pathways were induced more rapidly and subsequently decreased more rapidly in carCTL than in tcrCTL. The functional consequence of this rapid signaling in carCTL cells included faster lytic granule recruitment to the immune synapse, correlating with faster detachment of the CTL from the target cell. This study provides a mechanism for how CAR-T cells can debulk large tumor burden quickly and may contribute to further refinement of CAR design for enhancing the quality of signaling and programming of the T cell.
  • Item
    Thumbnail Image
    Switching on the green light for chimeric antigen receptor T-cell therapy
    Mardiana, S ; Lai, J ; House, IG ; Beavis, PA ; Darcy, PK (WILEY, 2019)
    Adoptive cellular therapy involving genetic modification of T cells with chimeric antigen receptor (CAR) transgene offers a promising strategy to broaden the efficacy of this approach for the effective treatment of cancer. Although remarkable antitumor responses have been observed following CAR T-cell therapy in a subset of B-cell malignancies, this has yet to be extended in the context of solid cancers. A number of promising strategies involving reprogramming the tumor microenvironment, increasing the specificity and safety of gene-modified T cells and harnessing the endogenous immune response have been tested in preclinical models that may have a significant impact in patients with solid cancers. This review will discuss these exciting new developments and the challenges that must be overcome to deliver a more sustained and potent therapeutic response.
  • Item
    Thumbnail Image
    CAR T cells take centre stage
    Beavis, PA ; Darcy, PK (WILEY, 2019)
  • Item
    Thumbnail Image
    Tissue-specific tumor microenvironments influence responses to immunotherapies
    Oliver, AJ ; Davey, AS ; Keam, SP ; Mardiana, S ; Chan, JD ; von Scheidt, B ; Beavis, PA ; House, IG ; Van Audernaerde, JRM ; Darcy, PK ; Kershaw, MH ; Slaney, CY (WILEY, 2019)
    OBJECTIVES: Investigation of variable response rates to cancer immunotherapies has exposed the immunosuppressive tumor microenvironment (TME) as a limiting factor of therapeutic efficacy. A determinant of TME composition is the tumor location, and clinical data have revealed associations between certain metastatic sites and reduced responses. Preclinical models to study tissue-specific TMEs have eliminated genetic heterogeneity, but have investigated models with limited clinical relevance. METHODS: We investigated the TMEs of tumors at clinically relevant sites of metastasis (liver and lungs) and their impact on αPD-1/αCTLA4 and trimAb (αDR5, α4-1BB, αCD40) therapy responses in the 67NR mouse breast cancer and Renca mouse kidney cancer models. RESULTS: Tumors grown in the lungs were resistant to both therapies whereas the same tumor lines growing in the mammary fat pad (MFP), liver or subcutaneously could be completely eradicated, despite greater tumor burden. Assessment of tumor cells and drug delivery in 67NR lung or MFP tumors revealed no differences and prompted investigation into the immune TME. Lung tumors had a more immunosuppressive TME with increased myeloid-derived suppressor cell infiltration, decreased T cell infiltration and activation, and decreased NK cell activation. Depletion of various immune cell subsets indicated an equivalent role for NK cells and CD8+ T cells in lung tumour control. Thus, targeting T cells with αPD-1/αCTLA4 or trimAb was not sufficient to elicit a robust antitumor response in lung tumors. CONCLUSION: Taken together, these data demonstrate that tissue-specific TMEs influence immunotherapy responses and highlight the importance in defining tissue-specific response patterns in patients.
  • Item
    Thumbnail Image
    Targeting Adenosine Receptor Signaling in Cancer Immunotherapy
    Sek, K ; Molck, C ; Stewart, GD ; Kats, L ; Darcy, PK ; Beavis, PA (MDPI, 2018-12)
    The immune system plays a major role in the surveillance and control of malignant cells, with the presence of tumor infiltrating lymphocytes (TILs) correlating with better patient prognosis in multiple tumor types. The development of 'checkpoint blockade' and adoptive cellular therapy has revolutionized the landscape of cancer treatment and highlights the potential of utilizing the patient's own immune system to eradicate cancer. One mechanism of tumor-mediated immunosuppression that has gained attention as a potential therapeutic target is the purinergic signaling axis, whereby the production of the purine nucleoside adenosine in the tumor microenvironment can potently suppress T and NK cell function. The production of extracellular adenosine is mediated by the cell surface ectoenzymes CD73, CD39, and CD38 and therapeutic agents have been developed to target these as well as the downstream adenosine receptors (A₁R, A2AR, A2BR, A₃R) to enhance anti-tumor immune responses. This review will discuss the role of adenosine and adenosine receptor signaling in tumor and immune cells with a focus on their cell-specific function and their potential as targets in cancer immunotherapy.