Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 3 of 3
  • Item
    Thumbnail Image
    IL-15 Preconditioning Augments CAR T Cell Responses to Checkpoint Blockade for Improved Treatment of Solid Tumors
    Giuffrida, L ; Sek, K ; Henderson, MA ; House, IG ; Lai, J ; Chen, AXY ; Todd, KL ; Petley, E ; Mardiana, S ; Todorovski, I ; Gruber, E ; Kelly, MJ ; Solomon, BJ ; Vervoort, SJ ; Johnstone, RW ; Parish, IA ; Neeson, PJ ; Kats, LM ; Darcy, PK ; Beavis, PA (CELL PRESS, 2020-11-04)
    Chimeric antigen receptor (CAR) T cell therapy has been highly successful in hematological malignancies leading to their US Food and Drug Administration (FDA) approval. However, the efficacy of CAR T cells in solid tumors is limited by tumor-induced immunosuppression, leading to the development of combination approaches, such as adjuvant programmed cell death 1 (PD-1) blockade. Current FDA-approved methods for generating CAR T cells utilize either anti-CD3 and interleukin (IL)-2 or anti-CD3/CD28 beads, which can generate a T cell product with an effector/exhausted phenotype. Whereas different cytokine preconditioning milieu, such as IL-7/IL-15, have been shown to promote T cell engraftment, the impact of this approach on CAR T cell responses to adjuvant immune-checkpoint blockade has not been assessed. In the current study, we reveal that the preconditioning of CAR T cells with IL-7/IL-15 increased CAR T cell responses to anti-PD-1 adjuvant therapy. This was associated with the emergence of an intratumoral CD8+CD62L+TCF7+IRF4- population that was highly responsive to anti-PD-1 therapy and mediated the vast majority of transcriptional and epigenetic changes in vivo following PD-1 blockade. Our data indicate that preservation of CAR T cells in a TCF7+ phenotype is crucial for their responsiveness to adjuvant immunotherapy approaches and should be a key consideration when designing clinical protocols.
  • Item
    Thumbnail Image
    MAIT cells regulate NK cell-mediated tumor immunity
    Petley, E ; Koay, H-F ; Henderson, MA ; Sek, K ; Todd, KL ; Keam, SP ; Lai, J ; House, IG ; Li, J ; Zethoven, M ; Chen, AXY ; Oliver, AJ ; Michie, J ; Freeman, AJ ; Giuffrida, L ; Chan, JD ; Pizzolla, A ; Mak, JYW ; McCulloch, TR ; Souza-Fonseca-Guimaraes, F ; Kearney, CJ ; Millen, R ; Ramsay, RG ; Huntington, ND ; McCluskey, J ; Oliaro, J ; Fairlie, DP ; Neeson, PJ ; Godfrey, D ; Beavis, PA ; Darcy, PK (NATURE PORTFOLIO, 2021-08-06)
    The function of MR1-restricted mucosal-associated invariant T (MAIT) cells in tumor immunity is unclear. Here we show that MAIT cell-deficient mice have enhanced NK cell-dependent control of metastatic B16F10 tumor growth relative to control mice. Analyses of this interplay in human tumor samples reveal that high expression of a MAIT cell gene signature negatively impacts the prognostic significance of NK cells. Paradoxically, pre-pulsing tumors with MAIT cell antigens, or activating MAIT cells in vivo, enhances anti-tumor immunity in B16F10 and E0771 mouse tumor models, including in the context of established metastasis. These effects are associated with enhanced NK cell responses and increased expression of both IFN-γ-dependent and inflammatory genes in NK cells. Importantly, activated human MAIT cells also promote the function of NK cells isolated from patient tumor samples. Our results thus describe an activation-dependent, MAIT cell-mediated regulation of NK cells, and suggest a potential therapeutic avenue for cancer treatment.
  • Item
    Thumbnail Image
    Generating CAR T cells from tumor-infiltrating lymphocytes.
    Mills, JK ; Henderson, MA ; Giuffrida, L ; Petrone, P ; Westwood, JA ; Darcy, PK ; Neeson, PJ ; Kershaw, MH ; Gyorki, DE (SAGE Publications, 2021)
    Background: Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapies have demonstrated promising, though limited, efficacy against melanoma. Methods: We designed a model system to explore the efficacy of dual specific T cells derived from melanoma patient TILs by transduction with a Her2-specific CAR. Results: Metastatic melanoma cells in our biobank constitutively expressed Her2 antigen. CAR-TIL produced greater amounts of IFN compared with parental TIL, when co-cultured with Her2 expressing tumor lines, including autologous melanoma tumor lines, although no consistent increase in cytotoxicity by TIL was afforded by expression of a CAR. Results of an in vivo study in NSG mice demonstrated tumor shrinkage when CAR-TILs were used in an adoptive cell therapy protocol. Conclusion: Potential limitations of transduced TIL in our study included limited proliferative potential and a terminally differentiated phenotype, which would need addressing in further work before consideration of clinical translation.