Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 21
  • Item
    Thumbnail Image
    SUN-LB75 The Anti-Tumor Activity of the Selective Ret Inhibitor Selpercatinib (LOXO-292) in Medullary Thyroid Cancer Is Independent of the Specific RET Mutation
    Cabanillas, ME ; Wirth, LJ ; Sherman, EJ ; Drilon, A ; Solomon, B ; Robinson, BG ; Lorch, JH ; McCoach, C ; Patel, J ; Leboulleux, S ; Worden, F ; Owonikoko, TK ; Brose, MS ; Taylor, MH ; Subbiah, V ; Rothenberg, SM ; Huang, X ; Zhu, E ; French, PP ; Shah, MH (The Endocrine Society, 2020-05-08)
    Abstract The RET receptor tyrosine kinase proto-oncogene is activated by somatic or germline mutations in a majority of medullary thyroid cancers (MTC). However, treatment of MTC has been challenging due to the lack of effective and tolerable RET-specific therapy, thus testing tumors for the presence of somatic RET mutation has not been warranted. In a first-in-human, phase 1/2 clinical trial (LIBRETTO-001, NCT03157128), selpercatinib (LOXO-292), an investigational, highly selective, potent small molecule RET kinase inhibitor, demonstrated significant and durable anti-tumor activity in patients with advanced RET-mutant MTC or with diverse RET fusion-positive cancers (1). Among the primary analysis set of patients with RET-mutant MTC previously treated with cabozantinib and/or vandetanib (N=55), the investigator-assessed objective response rate (ORR) per RECIST 1.1 was 56% (95% CI 42.3-69.7, n=31/55). Duration of response was not reached with a 10.6-months median follow-up (data cutoff date 17-Jun-2019). Here, we evaluated investigator-assessed ORR per RECIST 1.1 and clinical benefit rate (CBR) in this previously treated patient population by RET alteration and by germline or somatic testing used for enrollment. The ORR remained consistent across subgroups with RET M918T (49%, 95% CI 30.8-66.5, n=16/33), V804M/L gatekeeper mutations (60%, 95% CI 14.7-94.7, n=3/5), extracellular cysteine mutations (43%, 95% CI 9.9-81.6, n=3/7), other mutations (90%, 95% CI 55.5-99.7, n=9/10), and germline (50%, 95% CI 6.8-93.2, n=2/4) or somatic (57%, 95% CI 42.2-70.7, n=29/51) testing. The CBR, defined as the proportion of patients with best overall response of confirmed complete response, confirmed or unconfirmed partial response, or stable disease lasting 16 weeks or more, in this patient set was 87% (95% CI 75.5-94.7, n=48/55). The CBR remained consistent across subgroups with RET M918T (88%, 95% CI 71.8-96.6, n=29/33), V804M/L gatekeeper mutations (80%, 95% CI 28.4-99.5, n=4/5), extracellular cysteine mutations (71%, 95% CI 29.0-96.3, n=5/7), other mutations (100%, 95% CI 69.2-100.0, n=10/10), and germline (75%, 95% CI 19.4-99.4, n=3/4) or somatic (88%, 95% CI 76.1-95.6, n=45/51) testing. The primary technologies used to identify RET alterations were tumor next-generation sequencing (n=43) and polymerase chain reaction (n=9). As previously reported, selpercatinib was well tolerated with an acceptable safety profile (1). These results indicate broad anti-tumor activity for selpercatinib in patients with RET-mutant MTC irrespective of the specific RET mutation, and support implementation of RET mutation testing for patients with advanced MTC, including somatic testing, to identify patients who may benefit from selpercatinib. Reference: (1) Wirth et al., Ann Oncol. 2019 Oct; 30(supplement 5): v933.
  • Item
    Thumbnail Image
    Phase 2 Study of Cemiplimab in Patients with Advanced Cutaneous Squamous Cell Carcinoma (CSCC): Longer Follow-Up
    Rischin, D ; Khushalani, N ; Schmults, C ; Guminski, A ; Chang, AL ; Lewis, K ; Lim, A ; Hernandez-Aya, L ; Hughes, B ; Schadendorf, D ; Hauschild, A ; Stankevich, E ; Booth, J ; Yoo, S-Y ; Chen, Z ; Okoye, E ; Lowy, I ; Fury, M ; Migden, M (National Society for Cutaneous Medicine, 2020-10-27)
    Abstract not available.
  • Item
    Thumbnail Image
    Rapid-Fire Presentation Abstracts
    Lehmann, J ; Hussein, M ; Siva, S ; Moore, A ; Standen, T ; Subramanian, B ; Greer, P ; Clark, CH (WILEY, 2020-11)
  • Item
    No Preview Available
    Patient derived organoid model of penile squamous cell carcinoma
    Teh, J ; Guerra, G ; Heriot, A ; Ramsay, R ; Lawrentschuk, N (Elsevier BV, 2020-07)
  • Item
    No Preview Available
    PRECISION MEDICINE FOR PAEDIATRIC HIGH-GRADE DIFFUSE MIDLINE GLIOMAS - RESULTS FROM THE ZERO CHILDHOOD CANCER COMPREHENSIVE PRECISION MEDICINE PROGRAM
    Dong-Anh, K-Q ; Nagabushan, S ; Manoharan, N ; Arndt, G ; Barahona, P ; Cowley, MJ ; Ekert, PG ; Failes, T ; Bolanos, NF ; Gauthier, M ; Gifford, AJ ; Haber, M ; Kumar, A ; Lock, RB ; Marshall, GM ; Mayoh, C ; Mould, E ; Norris, MD ; Gopalakrishnan, A ; Omer, N ; Trebilcock, P ; Trahair, TN ; Tsoli, M ; Tucker, K ; Wong, M ; Tyrrell, V ; Lau, L ; Ziegler, DS (OXFORD UNIV PRESS INC, 2020-12)
    Abstract The Australian Zero Childhood Cancer (ZERO) program aims to assess the feasibility of a comprehensive precision medicine approach to improve outcomes for patients with an expected survival <30%. ZERO combines molecular profiling (whole genome sequencing, whole transcriptome sequencing, DNA methylation profiling) with in vitro high-throughput drug screening (HTS) and patient-derived xenograft drug efficacy testing. We report on the cohort of patients with midline high-grade glioma (HGG), including H3-K27M DMG, enrolled on the pilot study (TARGET) and on the ongoing ZERO clinical trial (PRISM). We identified 48 patients with midline HGG. Fresh or cryopreserved samples were submitted in 37 cases and cell culture was attempted in 30/37 cases with 45% success rate. The most commonly mutated genes/pathways identified by molecular profiling include H3-K27M mutations, DNA repair pathway, and PI3K/mTOR pathway. Two targetable fusions (NTRK and FGFR1) were reported. Five patients with germline alterations were identified. Thirty-five (72%) patients received a therapeutic recommendation from the ZERO molecular tumour board and the main recommended therapies were mTOR inhibitors, PARP inhibitors or tyrosine kinase inhibitors. HTS added evidence for the recommended therapy (n=3) or identified novel potential therapy (n=1). Out of the 35 patients, 16 received a recommended drug. Response to treatment was complete response for five months (n=1), partial response for nine months (n=1), stable disease (n=4), and progressive disease (n=10). These results highlight the feasibility of the ZERO platform and the value of fresh biopsy, necessary for pre-clinical drug testing. Targetable alterations were identified leading to clinical benefit in six patients.
  • Item
    No Preview Available
    Integrating Shared Survivorship Care into an Allogeneic Bone Marrow Transplant Long Term Follow up Service
    Panek-Hudson, Y ; Ritchie, DS ; Hookey, S ; Wright, T ; Masons, K ; O'Leary, S ; Chard, L (ELSEVIER SCIENCE INC, 2020-03)
  • Item
    No Preview Available
    Quantitation of CMV Specific T-Cell Expansion Using T Cell Receptor Beta Locus Deep Sequencing to Identify Patients at Risk of Viral Complications
    Kuzich, JA ; Kankanige, Y ; Guinto, J ; Ryland, G ; McBean, M ; Thompson, E ; Wong, E ; Koldej, R ; Collins, J ; Westerman, D ; Ritchie, DS ; Blombery, P (ELSEVIER SCIENCE INC, 2020-03)
  • Item
    Thumbnail Image
    HIGH DOSE-RATE BRACHYTHERAPY OF LOCALIZED PROSTATE CANCER CONVERTS TUMORS FROM COLD TO HOT
    Keam, S ; Halse, H ; ThuNgoc, N ; Wang, M ; Losio, NVK ; Mitchell, C ; Caramia, F ; Byrne, D ; Haupt, S ; Ryland, G ; Darcy, P ; Sandhu, S ; Blombery, P ; Haupt, Y ; Williams, S ; Neeson, P (BMJ PUBLISHING GROUP, 2020-11)
    Background Prostate cancer is frequently cured with high dose-rate brachytherapy (HDRBT) radiation as a front-line treatment. Although considered to be an immune-excluded tissue, immune responses to radiation are implicated in driving tumour-eradication in prostate cancer.1 This has not been proven, and yet is used as the rationale for clinical trials combining radiation and immunotherapies.2 We hypothesise that there is a predictable relationship between radiation and the immune responses in prostate cancer that could be used to provide sound rationale for specific immune interventions in solid tumours that are made possible by radiation therapy. Methods We present here new results stemming from our recently published immunoprofiling study of world-unique pre- and post-radiation tissues from 24 prostate cancer patients (figure 1A), RadBank cohort).3 These samples were assessed using immune cell multiplex IHC, gene expression profiling, digital spatial profiling (DSP) and computational analysis of cell distribution. Results This study unequivocally revealed that high dose-rate radiation converts predominately ‘cold’ prostate tumour tissue to a more activated ‘hot’ state comprised of two sub-types (high and a less activated intermediate state). These changes were evident in increased tumour inflammation gene signatures and immune checkpoint expression, immune cell composition changes, and alterations in spatial interactions. However, as 20% of the patients did not respond, we also explored pre-treatment gene signatures of patient responses to radiation – identifying potential mechanisms that prime tissues to respond more favourably. Most recently, we have explored three other important facets of the immune response to HDRBT: (i) putative differential drivers of high and intermediate responses (figure 1B), (ii) TCR clonality changes (figure 1C), and (iii) the influence of clinical features (e.g. Gleason grade) and treatment (e.g. androgen deprivation) (figure 1D). Differential expression analysis has identified key molecules (e.g. CD40LG and Lck expression) which are associated with higher activation responses. TCR sequencing of pre- and post-HDRBT tissue and peripheral circulating cells is also suggestive of engagement of the adaptive immune system and the emergence of tumor-specific T cells. Finally, multivariate analysis has also revealed that higher grade tumours exhibit higher basal levels of activation and IC expression – making them less sensitive to immune activation by HDRBT. Abstract 580 Figure 1The effect of prostate brachytherapy on immune contexts(A) Study of immune response in 24 patients treated with HDRBT at Peter MacCallum Cancer Center ((DOI:10.1136/jitc2020-000792). Examples of new insights including (B) molecules associated with higher activation levels (e.g. Lck and CD40LG/CD154), (C) changes in T cell receptor dominance and diversity in tissue and peripheral circulation, and (D) effects of clinical attributes on immune modulators (e.g. TGFbeta) and TIS activation states. Conclusions We have begun to resolve clear patient and clinical classifiers based on immune responses to radiation, and identified patient groups likely to benefit from immune therapy alongside radiation. Importantly, these classifications are associated with baseline gene expression profiles that may be used for pre-clinical stratification and more sophisticated treatment paradigms. Ethics Approval All participants provided consent covering tissue research as part of a prospective tissue collection study for prostate radiobiology research, approved by the Human Research Ethics Committee at the Peter MacCallum Cancer Centre (PMCC; HREC approvals 10/68, 13/167, 18/204). Consent Written informed consent was obtained from the patient for publication of this abstract and any accompanying images. A copy of the written consent is available for review by the Editor of this journal. References Dudzinski SO, et al., Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J Immunother Cancer 2019. 7(1): p. 218. Kwon E.D., et al., Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15(7): p. 700–12. Keam SP, et al., High dose-rate brachytherapy of localized prostate cancer converts tumors from cold to hot. J Immunother Cancer 2020;8(1).
  • Item
    Thumbnail Image
    NOVEL COMBINATION IMMUNOTHERAPY FOR BOOSTING AND PRIMING IMMUNE RESPONSES IN PANCREATIC CANCER: STRONG ANTI-TUMOUR EFFECTS WITH INTERLEUKIN-15 AND CD40 AGONIST TREATMENT
    Van Audenaerde, J ; Marcq, E ; von Scheidt, B ; Davey, A ; Oliver, A ; De Waele, J ; Quatannens, D ; Van Loenhout, J ; Pauwels, P ; Roeyen, G ; Lardon, F ; Slaney, C ; Peeters, M ; Kershaw, M ; Darcy, P ; Smits, E (BMJ PUBLISHING GROUP, 2020-11)
    Background With the poorest 5-year survival of all cancers, improving treatment for pancreatic cancer is one of the biggest challenges in cancer research. In this era of combination immunotherapies, we sought to explore the potential of combining both priming and activation of the immune system. To achieve this, we combined a CD40 agonist with interleukin-15 and tested its potential in pancreatic cancer. Methods Two different mouse models of pancreatic cancer were used to assess the potential of this combination regimen. Therefore, effects on tumour growth kinetics and survival were charted. Differential effects on immune signatures was investigated using RNA sequencing. Functional immune subset involvement was tested using different immune depletion experiments and multicolour flow cytometry in different relevant immune sites. Immune memory was checked using re-challenge experiments. Results We demonstrated profound reduction in tumour growth and increased survival of mice with the majority of mice being cured when both agents were combined, including an unprecedented dose reduction of CD40 agonist without losing any efficacy (fig 1). RNA sequencing analysis showed involvement of natural killer cell and T cell mediated anti-tumour responses and the importance of antigen-presenting cell pathways. This combination resulted in enhanced infiltration of tumours by both cytotoxic T cells and natural killer cells, as well as a striking increase in the ratio of CD8+ T cells over T regulatory cells. We also observed a significant increase in numbers of dendritic cells in tumour draining lymph nodes, particularly CD103+ dendritic cells with cross-presentation potential. A critical role for CD8+ T cells and involvement of natural killer cells in the anti-tumour effect was highlighted. Importantly, strong immune memory was established, with an increase in memory CD8+ T cells only when both interleukin-15 and the CD40 agonist were combined. Abstract 453 Figure 1Tumour kinetics and survival in Panc02 (left) and KPC (right) pancreatic cancer mouse models Conclusions We demonstrated profound synergistic anti-tumour effects upon combination of CD40 agonist and interleukin-15 treatment in mouse models of pancreatic cancer. This preclinical data supports initiation of a first-in-human clinical trial with this combination immunotherapy strategy in pancreatic cancer.
  • Item
    Thumbnail Image
    POTENTIAL MECHANISMS OF RESISTANCE IDENTIFIED THROUGH ANALYSIS OF MULTIPLE BIOMARKERS IN IMMUNE HOT NON-RESPONDERS WITH NON-SMALL CELL LUNG CANCER (NSCLC) TREATED WITH TISLELIZUMAB
    Desai, J ; Zhou, Q ; Deva, S ; Zhao, J ; Wang, J ; Tan, W ; Ma, X ; Zhang, Y ; Shen, Z ; Wu, X ; Leaw, S ; Zhang, J ; Wu, Y-L (BMJ PUBLISHING GROUP, 2020-11)
    Background Tislelizumab, an anti-PD-1 monoclonal antibody, has demonstrated clinical benefit for patients with NSCLC. The underlying response and resistance mechanisms to tislelizumab treatment remain unknown. Methods Baseline tumor samples from 59 nonsquamous (NSQ) and 41 squamous (SQ) NSCLC patients treated with tislelizumab monotherapy (NCT02407990 and NCT04068519) were tested for gene mutations using large panel next generation sequencing and RNA expression using gene expression profiling (GEP; Precision Immuno-Oncology Panel, HTG Molecular Diagnostics). GEP analyses of NSQ and SQ NSCLC were performed separately due to different gene expression patterns. Results The ORR, mPFS, and mOS in this pooled NSCLC cohort were 15.2% (95% CI: 9.0, 23.6), 4.1 months (95% CI: 2.20, 6.11), and 15.1 months (95% CI: 11.20, NE), respectively, with a median study follow-up of 15.3 months (95% CI: 14.06, 15.90). Non-responders (NRs) exhibited distinct tumor and immune gene signature profiles and could be clustered into two subgroups: NR1 and NR2. Compared with responders, NR1 had elevated cell cycle signatures in both NSQ (P=0.2) and SQ (P=0.03) cohorts, and a trend of decreased inflamed gene signature profiles. However, NR2 showed comparable or even higher tumor inflammation (18-gene), and CD8+ T-cell signature scores in both NSQ and SQ cohorts and could be classified as immune hot. To explore the resistance mechanisms of immune hot NRs, differentially expressed gene analyses between immune hot NR2 and responders were performed. M2 macrophage and Treg signature scores were higher in NR2 in both NSQ (M2, P=0.05; Treg, P=0.03) and SQ (M2, P=0.05 [subgroup of NR2]; Treg, P=0.03) cohorts; significantly higher expression of immune regulatory genes included PIK3CD, CCR2, CD244, IRAK3, and MAP4K1 (P<0.05) in NSQ and PIK3CD, CCR2, CD40, CD163, and MMP12 (P<0.05) in SQ. Significantly higher epithelial–mesenchymal transition (EMT) and angiogenesis gene expression, including SNAI1, FAP, VEGFC, and TEK (P<0.05) genes, were also observed in SQ NR2. Moreover, gene mutation analysis identified seven immune hot NR patients harboring either driver mutations (RET fusion, ROS1 fusion, BRAF, and PIK3CA amp) or well-established resistance mutations (loss of function mutation in JAK2, STK11, and MDM2 amplification). Conclusions Despite the presence of immune hot features, a subgroup of tislelizumab NRs with NSCLC were identified. High levels of immune suppressive factors, such as M2 macrophage and Treg signatures, angiogenesis, and EMT genes, as well as the existence of driver/resistance mutations, may indicate mechanisms of resistance of immune hot NRs, highlighting potential novel treatment targets. Acknowledgements Editorial assistance was provided by Agnieszka Laskowski, PhD, and Elizabeth Hermans, PhD (OPEN Health Medical Communications, Chicago, IL), and funded by the study sponsor. Trial Registration NCT02407990 and NCT04068519