Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 17
  • Item
    Thumbnail Image
    Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial
    Lin, NU ; Borges, V ; Anders, C ; Murthy, RK ; Paplomata, E ; Hamilton, E ; Hurvitz, S ; Loi, S ; Okines, A ; Abramson, V ; Bedard, PL ; Oliveira, M ; Mueller, V ; Zelnak, A ; DiGiovanna, MP ; Bachelot, T ; Chien, AJ ; O'Regan, R ; Wardley, A ; Conlin, A ; Cameron, D ; Carey, L ; Curigliano, G ; Gelmon, K ; Loibl, S ; Mayor, J ; McGoldrick, S ; An, X ; Winer, EP (AMER SOC CLINICAL ONCOLOGY, 2020-08-10)
    PURPOSE: In the HER2CLIMB study, patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer with brain metastases (BMs) showed statistically significant improvement in progression-free survival (PFS) with tucatinib. We describe exploratory analyses of intracranial efficacy and survival in participants with BMs. PATIENTS AND METHODS: Patients were randomly assigned 2:1 to tucatinib or placebo, in combination with trastuzumab and capecitabine. All patients underwent baseline brain magnetic resonance imaging; those with BMs were classified as active or stable. Efficacy analyses were performed by applying RECIST 1.1 criteria to CNS target lesions by investigator assessment. CNS-PFS (intracranial progression or death) and overall survival (OS) were evaluated in all patients with BMs. Confirmed intracranial objective response rate (ORR-IC) was evaluated in patients with measurable intracranial disease. RESULTS: There were 291 patients with BMs: 198 (48%) in the tucatinib arm and 93 (46%) in the control arm. The risk of intracranial progression or death was reduced by 68% in the tucatinib arm (hazard ratio [HR], 0.32; 95% CI, 0.22 to 0.48; P < .0001). Median CNS-PFS was 9.9 months in the tucatinib arm versus 4.2 months in the control arm. Risk of death was reduced by 42% in the tucatinib arm (OS HR, 0.58; 95% CI, 0.40 to 0.85; P = .005). Median OS was 18.1 versus 12.0 months. ORR-IC was higher in the tucatinib arm (47.3%; 95% CI, 33.7% to 61.2%) versus the control arm (20.0%; 95% CI, 5.7% to 43.7%; P = .03). CONCLUSION: In patients with HER2-positive breast cancer with BMs, the addition of tucatinib to trastuzumab and capecitabine doubled ORR-IC, reduced risk of intracranial progression or death by two thirds, and reduced risk of death by nearly half. To our knowledge, this is the first regimen to demonstrate improved antitumor activity against BMs in patients with HER2-positive breast cancer in a randomized, controlled trial.
  • Item
    Thumbnail Image
    The path to a better biomarker: application of a risk management framework for the implementation of PD-L1 and TILs as immuno-oncology biomarkers in breast cancer clinical trials and daily practice
    Gonzalez-Ericsson, P ; Stovgaard, ES ; Sua, LF ; Reisenbichler, E ; Kos, Z ; Carter, JM ; Michiels, S ; Le Quesne, J ; Nielsen, TO ; Laenkholm, A-V ; Fox, SB ; Adam, J ; Bartlett, JMS ; Rimm, DL ; Quinn, C ; Peeters, D ; Dieci, M ; Vincent-Salomon, A ; Cree, I ; Hida, A ; Balko, JM ; Haynes, HR ; Frahm, I ; Acosta-Haab, G ; Balancin, M ; Bellolio, E ; Yang, W ; Kirtani, P ; Sugie, T ; Ehinger, A ; Castaneda, CA ; Kok, M ; McArthur, H ; Siziopikou, K ; Badve, S ; Fineberg, S ; Gown, A ; Viale, G ; Schnitt, SJ ; Pruneri, G ; Penault-Llorca, F ; Hewitt, S ; Thompson, EA ; Allison, KH ; Symmans, WF ; Bellizzi, AM ; Brogi, E ; Moore, DA ; Larsimont, D ; Dillon, DA ; Lazar, A ; Lien, H ; Goetz, MP ; Broeckx, G ; El Bairi, K ; Harbeck, N ; Cimino-Mathews, A ; Sotiriou, C ; Adams, S ; Liu, S-W ; Loibl, S ; Chen, I-C ; Lakhani, SR ; Juco, JW ; Denkert, C ; Blackley, EF ; Demaria, S ; Leon-Ferre, R ; Gluz, O ; Zardavas, D ; Emancipator, K ; Ely, S ; Loi, S ; Salgado, R ; Sanders, M (WILEY, 2020-04)
  • Item
    Thumbnail Image
    Inhibition of RANK signaling in breast cancer induces an anti-tumor immune response orchestrated by CD8+T cells
    Gomez-Aleza, C ; Nguyen, B ; Yoldi, G ; Ciscar, M ; Barranco, A ; Hernandez-Jimenez, E ; Maetens, M ; Salgado, R ; Zafeiroglou, M ; Pellegrini, P ; Venet, D ; Garaud, S ; Trinidad, EM ; Benitez, S ; Vuylsteke, P ; Polastro, L ; Wildiers, H ; Simon, P ; Lindeman, G ; Larsimont, D ; Van den Eynden, G ; Velghe, C ; Rothe, F ; Willard-Gallo, K ; Michiels, S ; Munoz, P ; Walzer, T ; Planelles, L ; Penninger, J ; Azim, HA ; Loi, S ; Piccart, M ; Sotiriou, C ; Gonzalez-Suarez, E (NATURE RESEARCH, 2020-12-10)
    Most breast cancers exhibit low immune infiltration and are unresponsive to immunotherapy. We hypothesized that inhibition of the receptor activator of nuclear factor-κB (RANK) signaling pathway may enhance immune activation. Here we report that loss of RANK signaling in mouse tumor cells increases leukocytes, lymphocytes, and CD8+ T cells, and reduces macrophage and neutrophil infiltration. CD8+ T cells mediate the attenuated tumor phenotype observed upon RANK loss, whereas neutrophils, supported by RANK-expressing tumor cells, induce immunosuppression. RANKL inhibition increases the anti-tumor effect of immunotherapies in breast cancer through a tumor cell mediated effect. Comparably, pre-operative single-agent denosumab in premenopausal early-stage breast cancer patients from the Phase-II D-BEYOND clinical trial (NCT01864798) is well tolerated, inhibits RANK pathway and increases tumor infiltrating lymphocytes and CD8+ T cells. Higher RANK signaling activation in tumors and serum RANKL levels at baseline predict these immune-modulatory effects. No changes in tumor cell proliferation (primary endpoint) or other secondary endpoints are observed. Overall, our preclinical and clinical findings reveal that tumor cells exploit RANK pathway as a mechanism to evade immune surveillance and support the use of RANK pathway inhibitors to prime luminal breast cancer for immunotherapy.
  • Item
    Thumbnail Image
    53. TUCATINIB VS PLACEBO ADDED TO TRASTUZUMAB AND CAPECITABINE FOR PATIENTS WITH PREVIOUSLY TREATED HER2+ METASTATIC BREAST CANCER (MBC) WITH BRAIN METASTASES (BM) (HER2CLIMB)
    Lin, N ; Murthy, R ; Anders, C ; Borges, V ; Hurvitz, S ; Loi, S ; Abramson, V ; Bedard, P ; Oliveira, M ; Zelnack, A ; DiGiovanna, M ; Bachelot, T ; Chien, AJ ; O’Regan, R ; Wardley, A ; Mueller, V ; Carey, L ; McGoldrick, S ; An, X ; Winer, E (Oxford University Press (OUP), 2020-08-04)
    Abstract BACKGROUND HER2CLIMB (NCT02614794) primary results have been reported previously (Murthy, NEJM 2019). We report results of exploratory efficacy analyses in pts with brain metastases (BM). METHODS All HER2+ MBC pts enrolled had a baseline brain MRI. Pts with BM were eligible and randomized 2:1 to receive tucatinib (TUC) or placebo, in combination with trastuzumab and capecitabine. Efficacy analyses were performed by applying RECIST 1.1 to the brain based on investigator evaluation. CNS-PFS and OS were evaluated in BM pts overall. Intracranial (IC) confirmed ORR-IC and DOR-IC were evaluated in BM pts with measurable IC disease. After isolated brain progression, pts could continue study therapy until second progression, and time from randomization to second progression or death was evaluated. RESULTS Overall, 291 pts (48%) had BM at baseline: 198 (48%) in the TUC arm and 93 (46%) in the control arm. There was a 68% reduction in risk of CNS-PFS in the TUC arm (HR: 0.32; P&lt;0.0001). Median CNS-PFS was 9.9 mo in the TUC arm vs 4.2 mo in the control arm. Risk of overall death was reduced by 42% in the TUC arm (OS HR: 0.58; P=0.005). Median OS was 18.1 mo vs 12.0 mo. ORR-IC was higher in the TUC arm (47.3%) vs the control arm (20.0%). Median DOR-IC was 6.8 mo vs 3.0 mo. In pts with isolated brain progression who continued study therapy after local treatment (n=30), risk of second progression or death was reduced by 71% (HR: 0.29), and median time from randomization to second progression or death was 15.9 mo vs 9.7 mo, favoring the TUC arm. CONCLUSIONS In pts with previously treated HER2+ MBC with BM, TUC in combination with trastuzumab and capecitabine doubled the ORR-IC, reduced risk of IC progression or death by two-thirds and reduced risk of death by nearly half.
  • Item
    No Preview Available
    Identifying oncogenic drivers associated with increased risk of late distant recurrence in postmenopausal, estrogen receptor-positive, HER2-negative early breast cancer: results from the BIG 1-98 study
    Luen, SJ ; Asher, R ; Lee, CK ; Savas, P ; Kammler, R ; Dell'Orto, P ; Biasi, OM ; Demanse, D ; Hackl, W ; Thuerlimann, B ; Viale, G ; Di Leo, A ; Colleoni, M ; Regan, MM ; Loi, S (ELSEVIER, 2020-10)
    BACKGROUND: In postmenopausal, estrogen receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative early breast cancer, the risk for distant recurrence can extend beyond 5 years of adjuvant endocrine therapy. This study aims to identify genomic driver alterations associated with late distant recurrence. PATIENTS AND METHODS: Next generation sequencing was used to characterize driver alterations in primary tumors from a subset of 764 postmenopausal estrogen receptor-positive/HER2-negative patients from the BIG 1-98 randomized trial. Late distant recurrence events were defined as ≥5 years from time of randomization). The association of driver alterations with distant recurrence-free interval in early and late time periods was assessed using Cox regression models. Multivariable analyses were carried out to adjust for clinicopathological factors. Weighted analysis methods were used in order to correct for over-sampling of distant recurrences. RESULTS: A total of 538 of 764 (70%) samples were successfully sequenced including 88 (63%) early and 52 (37%) late distant recurrence events after a median follow up of 8.1 years. In univariable analysis for late distant recurrence, PIK3CA mutations (58.8%) were significantly associated with reduced risk [hazard ratio (HR) 0.40, 95% confidence interval (CI) 0.20-0.82, P = 0.012], whereas amplifications on chromosome 8p11 (10.9%) (HR 4.79, 95% CI 2.30-9.97, P < 0.001) and BRCA2 mutations (2.3%) (HR 5.39, 95% CI 1.51-19.29, P = 0.010) were significantly associated with an increased risk. In multivariable analysis, only amplifications on 8p11 (P = 0.002) and BRCA2 mutations (P = 0.013) remained significant predictors. CONCLUSIONS: In estrogen receptor-positive/HER2-negative postmenopausal early breast cancer, PIK3CA mutations were associated with reduced risk of late distant recurrence, whereas amplifications on 8p11 and BRCA2 mutations were associated with increased risk of late distant recurrence. The characterization of oncogenic driver alterations may aid in refining treatment choices in the late disease setting, and help identify potential drug targets for testing in future trials.
  • Item
    No Preview Available
    Geospatial immune variability illuminates differential evolution of lung adenocarcinoma
    AbdulJabbar, K ; Raza, SEA ; Rosenthal, R ; Jamal-Hanjani, M ; Veeriah, S ; Akarca, A ; Lund, T ; Moore, DA ; Salgado, R ; Al Bakir, M ; Zapata, L ; Hiley, CT ; Officer, L ; Sereno, M ; Smith, CR ; Loi, S ; Hackshaw, A ; Marafioti, T ; Quezada, SA ; McGranahan, N ; Le Quesne, J ; Swanton, C ; Yuan, Y (NATURE PORTFOLIO, 2020-07)
    Remarkable progress in molecular analyses has improved our understanding of the evolution of cancer cells toward immune escape1-5. However, the spatial configurations of immune and stromal cells, which may shed light on the evolution of immune escape across tumor geographical locations, remain unaddressed. We integrated multiregion exome and RNA-sequencing (RNA-seq) data with spatial histology mapped by deep learning in 100 patients with non-small cell lung cancer from the TRACERx cohort6. Cancer subclones derived from immune cold regions were more closely related in mutation space, diversifying more recently than subclones from immune hot regions. In TRACERx and in an independent multisample cohort of 970 patients with lung adenocarcinoma, tumors with more than one immune cold region had a higher risk of relapse, independently of tumor size, stage and number of samples per patient. In lung adenocarcinoma, but not lung squamous cell carcinoma, geometrical irregularity and complexity of the cancer-stromal cell interface significantly increased in tumor regions without disruption of antigen presentation. Decreased lymphocyte accumulation in adjacent stroma was observed in tumors with low clonal neoantigen burden. Collectively, immune geospatial variability elucidates tumor ecological constraints that may shape the emergence of immune-evading subclones and aggressive clinical phenotypes.
  • Item
    No Preview Available
    The T cell differentiation landscape is shaped by tumour mutations in lung cancer
    Ghorani, E ; Reading, JL ; Henry, JY ; de Massy, MR ; Rosenthal, R ; Turati, V ; Joshi, K ; Furness, AJS ; Aissa, AB ; Saini, SK ; Ramskov, S ; Georgiou, A ; Sunderland, MW ; Wong, YNS ; De Mucha, MV ; Day, W ; Galvez-Cancino, F ; Becker, PD ; Uddin, I ; Ismail, M ; Ronel, T ; Woolston, A ; Jamal-Hanjani, M ; Veeriah, S ; Birkbak, NJ ; Wilson, GA ; Litchfield, K ; Conde, L ; Guerra-Assuncao, JA ; Blighe, K ; Biswas, D ; Salgado, R ; Lund, T ; Al Bakir, M ; Moore, DA ; Hiley, CT ; Loi, S ; Sun, Y ; Yuan, Y ; AbdulJabbar, K ; Turajilic, S ; Herrero, J ; Enver, T ; Hadrup, SR ; Hackshaw, A ; Peggs, KS ; McGranahan, N ; Chain, B ; Swanton, C ; Quezada, SA (NATURE PORTFOLIO, 2020-05)
    Tumour mutational burden (TMB) predicts immunotherapy outcome in non-small cell lung cancer (NSCLC), consistent with immune recognition of tumour neoantigens. However, persistent antigen exposure is detrimental for T cell function. How TMB affects CD4 and CD8 T cell differentiation in untreated tumours, and whether this affects patient outcomes is unknown. Here we paired high-dimensional flow cytometry, exome, single-cell and bulk RNA sequencing from patients with resected, untreated NSCLC to examine these relationships. TMB was associated with compartment-wide T cell differentiation skewing, characterized by loss of TCF7-expressing progenitor-like CD4 T cells, and an increased abundance of dysfunctional CD8 and CD4 T cell subsets, with significant phenotypic and transcriptional similarity to neoantigen-reactive CD8 T cells. A gene signature of redistribution from progenitor-like to dysfunctional states associated with poor survival in lung and other cancer cohorts. Single-cell characterization of these populations informs potential strategies for therapeutic manipulation in NSCLC.
  • Item
    No Preview Available
    Reactive stroma and trastuzumab resistance in HER2-positive early breast cancer
    Sonnenblick, A ; Salmon-Divon, M ; Salgado, R ; Dvash, E ; Ponde, N ; Zahavi, T ; Salmon, A ; Loibl, S ; Denkert, C ; Joensuu, H ; Ameye, L ; Van den Eynden, G ; Kellokumpu-Lehtinen, P-L ; Azaria, A ; Loi, S ; Michiels, S ; Richard, F ; Sotiriou, C (WILEY, 2020-07-01)
    We investigated the value of reactive stroma as a predictor for trastuzumab resistance in patients with early HER2-positive breast cancer receiving adjuvant therapy. The pathological reactive stroma and the mRNA gene signatures that reflect reactive stroma in 209 HER2-positive breast cancer samples from the FinHer adjuvant trial were evaluated. Levels of stromal gene signatures were determined as a continuous parameter, and pathological reactive stromal findings were defined as stromal predominant breast cancer (SPBC; ≥50% stromal) and correlated with distant disease-free survival. Gene signatures associated with reactive stroma in HER2-positive early breast cancer (N = 209) were significantly associated with trastuzumab resistance in estrogen receptor (ER)-negative tumors (hazard ratio [HR] = 1.27 p interaction = 0.014 [DCN], HR = 1.58, p interaction = 0.027 [PLAU], HR = 1.71, p interaction = 0.019 [HER2STROMA, novel HER2 stromal signature]), but not in ER-positive tumors (HR = 0.73 p interaction = 0.47 [DCN], HR = 0.71, p interaction = 0.73 [PLAU], HR = 0.84; p interaction = 0.36 [HER2STROMA]). Pathological evaluation of HER2-positive/ER-negative tumors suggested an association between SPBC and trastuzumab resistance. Reactive stroma did not correlate with tumor-infiltrating lymphocytes (TILs), and the expected benefit from trastuzumab in patients with high levels of TILs was pronounced only in tumors with low stromal reactivity (SPBC <50%). In conclusion, reactive stroma in HER2-positive/ER-negative early breast cancer tumors may predict resistance to adjuvant trastuzumab therapy.
  • Item
    Thumbnail Image
    Circulating tumour DNA in metastatic breast cancer to guide clinical trial enrolment and precision oncology: A cohort study
    Zivanovic Bujak, A ; Weng, C-F ; Silva, MJ ; Yeung, M ; Lo, L ; Ftouni, S ; Litchfield, C ; Ko, Y-A ; Kuykhoven, K ; Van Geelen, C ; Chandrashekar, S ; Dawson, MA ; Loi, S ; Wong, SQ ; Dawson, S-J ; Swanton, C (PUBLIC LIBRARY SCIENCE, 2020-10)
    BACKGROUND: Metastatic breast cancer (mBC) is a heterogenous disease with increasing availability of targeted therapies as well as emerging genomic markers of therapeutic resistance, necessitating timely and accurate molecular characterization of disease. As a minimally invasive test, analysis of circulating tumour DNA (ctDNA) is well positioned for real-time genomic profiling to guide treatment decisions. Here, we report the results of a prospective testing program established to assess the feasibility of ctDNA analysis to guide clinical management of mBC patients. METHODS AND FINDINGS: Two hundred thirty-four mBC patients (median age 54 years) were enrolled between June 2015 and October 2018 at the Peter MacCallum Cancer Centre, Melbourne, Australia. Median follow-up was 15 months (range 1-46). All patient samples at the time of enrolment were analysed in real time for the presence of somatic mutations. Longitudinal plasma testing during the course of patient management was also undertaken in a subset of patients (n = 67, 28.6%), according to clinician preference, for repeated molecular profiling or disease monitoring. Detection of somatic mutations from patient plasma was performed using a multiplexed droplet digital PCR (ddPCR) approach to identify hotspot mutations in PIK3CA, ESR1, ERBB2, and AKT1. In parallel, subsets of samples were also analysed via next-generation sequencing (targeted panel sequencing and low-coverage whole-genome sequencing [LC-WGS]). The sensitivity of ddPCR and targeted panel sequencing to identify actionable mutations was compared. Results were discussed at a multidisciplinary breast cancer meeting prior to treatment decisions. ddPCR and targeted panel sequencing identified at least 1 actionable mutation at baseline in 80/234 (34.2%) and 62/159 (39.0%) of patients tested, respectively. Combined, both methods detected an actionable alteration in 104/234 patients (44.4%) through baseline or serial ctDNA testing. LC-WGS was performed on 27 patients from the cohort, uncovering several recurrently amplified regions including 11q13.3 encompassing CCND1. Increasing ctDNA levels were associated with inferior overall survival, whether assessed by ddPCR, targeted sequencing, or LC-WGS. Overall, the ctDNA results changed clinical management in 40 patients including the direct recruitment of 20 patients to clinical trials. Limitations of the study were that it was conducted at a single site and that 31.3% of participants were lost to follow-up. CONCLUSION: In this study, we found prospective ctDNA testing to be a practical and feasible approach that can guide clinical trial enrolment and patient management in mBC.
  • Item
    Thumbnail Image
    Immune system and angiogenesis-related potential surrogate biomarkers of response to everolimus-based treatment in hormone receptor-positive breast cancer: an exploratory study
    Schettini, F ; Sobhani, N ; Ianza, A ; Triulzi, T ; Molteni, A ; Lazzari, MC ; Strina, C ; Milani, M ; Corona, SP ; Sirico, M ; Bernocchi, O ; Giudici, F ; Cappelletti, MR ; Ciruelos, E ; Jerusalem, G ; Loi, S ; Fox, SB ; Generali, D (SPRINGER, 2020-11)
    PURPOSE: mTOR inhibitor everolimus is used for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC). No reliable predictive biomarker of response is available. Following evidences from other solid tumors, we aimed to assess the association between treatment-associated immune system features and everolimus activity. METHODS: We retrospectively explored a correlation with the therapeutic activity of everolimus and tumor-associated immune pathways with ingenuity pathway analysis (IPA), neutrophil-to-lymphocyte ratio (NLR), circulating lymphocytes, and endothelial cells (CECs) in 3 different HR+ mBC studies, including the BALLET phase IIIb study. RESULTS: The circulating levels of CD3+/CD8+, CD3+/CD4+, and overall T lymphocytes were higher in responders versus non-responders at baseline (p = 0.017, p < 0.001, p = 0.034) and after treatment (p = 0.01, p = 0.003, p = 0.023). Reduced CECs, a tumor neoangiogenesis marker, were observed in responders after treatment (p < 0.001). Patients with low NLR (≤ 4.4) showed a better progression-free survival compared to patients with high NLR (> 4.4) (p = 0.01). IPA showed that the majority of immunity-related genes were found upregulated in responders compared to non-responders before treatment, but not after. CONCLUSIONS: Lymphocytes subpopulations, CECs and NLR could be interesting biomarkers predictive of response to everolimus-based regimens, potentially useful in daily clinical practice to select/monitor everolimus-based treatment in mBC. Further studies to confirm such hypotheses are warranted.