Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 8 of 8
  • Item
    No Preview Available
    Tucatinib promotes immune activation and synergizes with programmed cell death-1 and programmed cell death-ligand 1 inhibition in HER2-positive breast cancer
    Li, R ; Sant, S ; Brown, E ; Caramia, F ; Nikolic, B ; Clarke, K ; Byrne, A ; Gonzalez, LEL ; Savas, P ; Luen, SJ ; Teo, ZL ; Virassamy, B ; Neeson, PJ ; Darcy, PK ; Loi, S (OXFORD UNIV PRESS INC, 2023-07-06)
    BACKGROUND: Programmed cell death-1 (PD-1) and programmed cell death-ligand 1 (PD-L1) inhibitors have poor efficacy in patients with trastuzumab-resistant advanced HER2-positive breast cancer. Tucatinib is a potent, selective anti-HER2 tyrosine kinase inhibitor with proven clinical benefit in the advanced setting in patients with trastuzumab resistance. We investigated if tucatinib can alter the tumor microenvironment and if this could be harnessed for therapeutic efficacy. METHODS: We investigated the antitumor efficacy and contribution of the immune response of tucatinib using 2 immunocompetent, HER2-positive murine breast cancer models (trastuzumab-sensitive H2N113; trastuzumab-resistant Fo5) and the efficacy of tucatinib with trastuzumab and PD-1 or PD-L1 checkpoint inhibitors. RESULTS: In both models, tucatinib statistically significantly inhibited tumor growth and demonstrated dose-dependent efficacy. Ex vivo analysis by flow cytometry of tumor-infiltrating lymphocytes in mice treated with tucatinib showed increased frequency, higher proliferation, and enhanced effector function of CD8+ effector memory T cells. Tucatinib treatment also increased frequency of CD8+PD-1+ and CD8+TIM3+ T cells, CD49+ natural killer cells, monocytes, and major histocompatibility complex II expression on dendritic cells and macrophages and a decrease in myeloid-derived suppressor cells. Gene expression analysis revealed statistically significant enrichment in pathways associated with immune activation, type I and II interferon response, adaptive immune response, and antigen receptor signaling. In vivo, tucatinib and α-PD-L1 or α-PD-1 demonstrated statistically significantly increased efficacy and improved survival of mice compared with tucatinib alone. CONCLUSION: Tucatinib modulates the immune microenvironment favorably, and combination treatment with α-PD-L1 or α-PD-1 demonstrated increased efficacy in preclinical HER2-positive tumor models. These findings provide a rationale for investigation of tucatinib and immune checkpoint inhibition in the clinic.
  • Item
    No Preview Available
    Alpelisib Monotherapy for PI3K-Altered, Pretreated Advanced Breast Cancer: A Phase II Study
    Savas, P ; Lo, LL ; Luen, SJ ; Blackley, EF ; Callahan, J ; Moodie, K ; van Geelen, CT ; Ko, Y-A ; Weng, C-F ; Wein, L ; Silva, MJ ; Bujak, AZ ; Yeung, MM ; Ftouni, S ; Hicks, RJ ; Francis, PA ; Lee, CK ; Dawson, S-J ; Loi, S (AMER ASSOC CANCER RESEARCH, 2022-09-02)
    UNLABELLED: There is limited knowledge on the benefit of the α-subunit-specific PI3K inhibitor alpelisib in later lines of therapy for advanced estrogen receptor-positive (ER+) HER2- and triple-negative breast cancer (TNBC). We conducted a phase II multicohort study of alpelisib monotherapy in patients with advanced PI3K pathway mutant ER+HER2- and TNBC. In the intention-to-treat ER+ cohort, the overall response rate was 30% and the clinical benefit rate was 36%. A decline in PI3K pathway mutant circulating tumor DNA (ctDNA) levels from baseline to week 8 while on therapy was significantly associated with a partial response, clinical benefit, and improved progression-free-survival [HR 0.24; 95% confidence interval (CI), 0.083-0.67, P = 0.0065]. Detection of ESR1 mutations at baseline in plasma was also associated with clinical benefit and improved progression-free survival (HR 0.22; 95% CI, 0.078-0.60, P = 0.003). SIGNIFICANCE: Alpelisib monotherapy displayed efficacy in heavily pretreated ER+ breast cancer with PIK3CA mutations. PIK3CA mutation dynamics in plasma during treatment and ESR1 mutations detected in plasma at baseline were candidate biomarkers predictive of benefit from alpelisib, highlighting the utility of ctDNA assays in this setting. This article is highlighted in the In This Issue feature, p. 2007.
  • Item
    Thumbnail Image
    Combining Radiotherapy and Immunotherapy in Metastatic Breast Cancer: Current Status and Future Directions
    David, S ; Tan, J ; Siva, S ; Karroum, L ; Savas, P ; Loi, S (MDPI, 2022-04)
    The role of radiotherapy and immunotherapy with immune checkpoint inhibitors (ICI) is of emerging interest in many solid tumours, including breast cancer. There is increasing evidence that the host's immune system plays an important role in influencing the response to treatment and prognosis in breast cancer. Several pre-clinical studies and clinical trials have reported on the 'abscopal effect-regression of distant untreated tumour sites, mediated by an immunological response following ionizing radiation to a targeted tumour site. Stereotactic Ablative Body Radiotherapy (SABR) is a non-invasive technique used to augment various immune responses with an ablative tumoricidal dose when compared to conventional radiotherapy. SABR is characterized by typically 1-5 precision radiotherapy treatments that simultaneously deliver a high dose, whilst sparing normal tissues. Following SABR, there is evidence of systemic immune activation in patients with increased PD1 expression on CD8+ and CD4+ T cells. Studies continue to focus on metastatic triple-negative disease, a highly immunogenic subtype of breast cancer with poor prognosis. In this review, we discuss the immunological effect of SABR, alone and in combination with immunotherapy, and the importance of dose and fractionation. We also propose future strategies for treating oligometastatic disease, where this approach may be most useful for producing durable responses.
  • Item
    Thumbnail Image
    Stereotactic ablative body radiotherapy (SABR) for bone only oligometastatic breast cancer: A prospective clinical trial
    David, S ; Tan, J ; Savas, P ; Bressel, M ; Kelly, D ; Foroudi, F ; Loi, S ; Siva, S (CHURCHILL LIVINGSTONE, 2020-02)
    BACKGROUND: Stereotactic ablative body radiotherapy (SABR) is an emerging noninvasive approach for the treatment of oligometastases. Limited prospective evidence is available in breast cancer. OBJECTIVES: To determine the safety and feasibility of single fraction SABR for patients with bone only oligometastatic breast cancer. Secondary endpoints were local and distant progression-free survival (LPFS and DPFS), toxicity and response assessment. METHODS AND MATERIALS: In this single institution prospective trial we screened patients with computed tomography, bone scan, and sodium fluoride positron emission tomography. Eligible patients had one to three bone only oligometastases. All patients were treated at a dose of 20Gy in 1 fraction to each metastasis. Kaplan-Meier methods were used to determine local and distant progression free survival (LPFS and DPFS). Toxicity was graded using Common Terminology Criteria for Adverse Event version 4.0. RESULTS: 15 eligible patients were recruited to the study. Median follow-up time was 24 months. The treatment was feasible in 12 (80%) of patients with 3 (20%) of patients having treatment delayed by more than 3 days. 10 (67%) of patients experienced grade 1 treatment related toxicity, 4 (27%) experienced grade 2 toxicity and no patients experienced grade 3 or 4 treatment related toxicity. The two-year LPFS was 100%, DPFS was 67%. CONCLUSION: We observed that SABR is feasible, well tolerated and effective in this cohort with two thirds of patients disease-free at two years. In selected patients with bone-only oligometastatic disease, SABR could be considered a treatment option. Randomised trials are required to assess the impact of SABR on overall survival when compared to the standard of care.
  • Item
    Thumbnail Image
    Pitfalls in assessing stromal tumor infiltrating lymphocytes (sTILs) in breast cancer
    Kos, Z ; Roblin, E ; Kim, RS ; Michiels, S ; Gallas, BD ; Chen, W ; van de Vijver, KK ; Goel, S ; Adams, S ; Demaria, S ; Viale, G ; Nielsen, TO ; Badve, SS ; Symmans, WF ; Sotiriou, C ; Rimm, DL ; Hewitt, S ; Denkert, C ; Loibl, S ; Luen, SJ ; Bartlett, JMS ; Savas, P ; Pruneri, G ; Dillon, DA ; Cheang, MCU ; Tutt, A ; Hall, JA ; Kok, M ; Horlings, HM ; Madabhushi, A ; van der Laak, J ; Ciompi, F ; Laenkholm, A-V ; Bellolio, E ; Gruosso, T ; Fox, SB ; Araya, JC ; Floris, G ; Hudecek, J ; Voorwerk, L ; Beck, AH ; Kerner, J ; Larsimont, D ; Declercq, S ; Van den Eynden, G ; Pusztai, L ; Ehinger, A ; Yang, W ; AbdulJabbar, K ; Yuan, Y ; Singh, R ; Hiley, C ; al Bakir, M ; Lazar, AJ ; Naber, S ; Wienert, S ; Castillo, M ; Curigliano, G ; Dieci, M-V ; Andre, F ; Swanton, C ; Reis-Filho, J ; Sparano, J ; Balslev, E ; Chen, I-C ; Stovgaard, EIS ; Pogue-Geile, K ; Blenman, KRM ; Penault-Llorca, F ; Schnitt, S ; Lakhani, SR ; Vincent-Salomon, A ; Rojo, F ; Braybrooke, JP ; Hanna, MG ; Soler-Monso, MT ; Bethmann, D ; Castaneda, CA ; Willard-Gallo, K ; Sharma, A ; Lien, H-C ; Fineberg, S ; Thagaard, J ; Comerma, L ; Gonzalez-Ericsson, P ; Brogi, E ; Loi, S ; Saltz, J ; Klaushen, F ; Cooper, L ; Amgad, M ; Moore, DA ; Salgado, R (NATURE RESEARCH, 2020-05-12)
    Stromal tumor-infiltrating lymphocytes (sTILs) are important prognostic and predictive biomarkers in triple-negative (TNBC) and HER2-positive breast cancer. Incorporating sTILs into clinical practice necessitates reproducible assessment. Previously developed standardized scoring guidelines have been widely embraced by the clinical and research communities. We evaluated sources of variability in sTIL assessment by pathologists in three previous sTIL ring studies. We identify common challenges and evaluate impact of discrepancies on outcome estimates in early TNBC using a newly-developed prognostic tool. Discordant sTIL assessment is driven by heterogeneity in lymphocyte distribution. Additional factors include: technical slide-related issues; scoring outside the tumor boundary; tumors with minimal assessable stroma; including lymphocytes associated with other structures; and including other inflammatory cells. Small variations in sTIL assessment modestly alter risk estimation in early TNBC but have the potential to affect treatment selection if cutpoints are employed. Scoring and averaging multiple areas, as well as use of reference images, improve consistency of sTIL evaluation. Moreover, to assist in avoiding the pitfalls identified in this analysis, we developed an educational resource available at www.tilsinbreastcancer.org/pitfalls.
  • Item
    Thumbnail Image
    Report on computational assessment of Tumor Infiltrating Lymphocytes from the International Immuno-Oncology Biomarker Working Group
    Amgad, M ; Stovgaard, ES ; Balslev, E ; Thagaard, J ; Chen, W ; Dudgeon, S ; Sharma, A ; Kerner, JK ; Denkert, C ; Yuan, Y ; AbdulJabbar, K ; Wienert, S ; Savas, P ; Voorwerk, L ; Beck, AH ; Madabhushi, A ; Hartman, J ; Sebastian, MM ; Horlings, HM ; Hudecek, J ; Ciompi, F ; Moore, DA ; Singh, R ; Roblin, E ; Balancin, ML ; Mathieu, M-C ; Lennerz, JK ; Kirtani, P ; Chen, I-C ; Braybrooke, JP ; Pruneri, G ; Demaria, S ; Adams, S ; Schnitt, SJ ; Lakhani, SR ; Rojo, F ; Comerma, L ; Badve, SS ; Khojasteh, M ; Symmans, WF ; Sotiriou, C ; Gonzalez-Ericsson, P ; Pogue-Geile, KL ; Kim, RS ; Rimm, DL ; Viale, G ; Hewitt, SM ; Bartlett, JMS ; Penault-Llorca, F ; Goel, S ; Lien, H-C ; Loibl, S ; Kos, Z ; Loi, S ; Hanna, MG ; Michiels, S ; Kok, M ; Nielsen, TO ; Lazar, AJ ; Bago-Horvath, Z ; Kooreman, LFS ; van der Laak, JAWM ; Saltz, J ; Gallas, BD ; Kurkure, U ; Barnes, M ; Salgado, R ; Cooper, LAD (NATURE RESEARCH, 2020-05-12)
    Assessment of tumor-infiltrating lymphocytes (TILs) is increasingly recognized as an integral part of the prognostic workflow in triple-negative (TNBC) and HER2-positive breast cancer, as well as many other solid tumors. This recognition has come about thanks to standardized visual reporting guidelines, which helped to reduce inter-reader variability. Now, there are ripe opportunities to employ computational methods that extract spatio-morphologic predictive features, enabling computer-aided diagnostics. We detail the benefits of computational TILs assessment, the readiness of TILs scoring for computational assessment, and outline considerations for overcoming key barriers to clinical translation in this arena. Specifically, we discuss: 1. ensuring computational workflows closely capture visual guidelines and standards; 2. challenges and thoughts standards for assessment of algorithms including training, preanalytical, analytical, and clinical validation; 3. perspectives on how to realize the potential of machine learning models and to overcome the perceptual and practical limits of visual scoring.
  • Item
    Thumbnail Image
    Clinical implications of prospective genomic profiling of metastatic breast cancer patients
    van Geelen, CT ; Savas, P ; Teo, ZL ; Luen, SJ ; Weng, C-F ; Ko, Y-A ; Kuykhoven, KS ; Caramia, F ; Salgado, R ; Francis, PA ; Dawson, S-J ; Fox, SB ; Fellowes, A ; Loi, S (BMC, 2020-08-18)
    BACKGROUND: Metastatic breast cancer remains incurable. Next-generation sequencing (NGS) offers the ability to identify actionable genomic alterations in tumours which may then be matched with targeted therapies, but the implementation and utility of this approach is not well defined for patients with metastatic breast cancer. METHODS: We recruited patients with advanced breast cancer of any subtype for prospective targeted NGS of their most recent tumour samples, using a panel of 108 breast cancer-specific genes. Genes were classified as actionable or non-actionable using the European Society of Medical Oncology Scale for Clinical Actionability of Molecular Targets (ESCAT) guidelines. RESULTS: Between February 2014 and May 2019, 322 patients were enrolled onto the study, with 72% (n = 234) of patients successfully sequenced (n = 357 samples). The majority (74%, n = 171) of sequenced patients were found to carry a potentially actionable alteration, the most common being a PIK3CA mutation. Forty-three percent (n = 74) of patients with actionable alterations were referred for a clinical trial or referred for confirmatory germline testing or had a change in therapy outside of clinical trials. We found alterations in AKT1, BRCA2, CHEK2, ESR1, FGFR1, KMT2C, NCOR1, PIK3CA and TSC2 to be significantly enriched in our metastatic population compared with primary breast cancers. Concordance between primary and metastatic samples for key driver genes (TP53, ERBB2 amplification) was > 75%. Additionally, we found that patients with a higher number of mutations had a significantly worse overall survival. CONCLUSION: Genomic profiling of patients with metastatic breast cancer can have clinical implications and should be considered in all suitable patients.
  • Item
    Thumbnail Image
    Application of a risk-management framework for integration of stromal tumor-infiltrating lymphocytes in clinical trials
    Hudecek, J ; Voorwerk, L ; van Seijen, M ; Nederlof, I ; de Maaker, M ; van den Berg, J ; van de Vijver, KK ; Sikorska, K ; Adams, S ; Demaria, S ; Viale, G ; Nielsen, TO ; Badve, SS ; Michiels, S ; Symmans, WF ; Sotiriou, C ; Rimm, DL ; Hewitt, SM ; Denkert, C ; Loibl, S ; Loi, S ; Bartlett, JMS ; Pruneri, G ; Dillon, DA ; Cheang, MCU ; Tutt, A ; Hall, JA ; Kos, Z ; Salgado, R ; Kok, M ; Horlings, HM (NATURE RESEARCH, 2020-05-12)
    Stromal tumor-infiltrating lymphocytes (sTILs) are a potential predictive biomarker for immunotherapy response in metastatic triple-negative breast cancer (TNBC). To incorporate sTILs into clinical trials and diagnostics, reliable assessment is essential. In this review, we propose a new concept, namely the implementation of a risk-management framework that enables the use of sTILs as a stratification factor in clinical trials. We present the design of a biomarker risk-mitigation workflow that can be applied to any biomarker incorporation in clinical trials. We demonstrate the implementation of this concept using sTILs as an integral biomarker in a single-center phase II immunotherapy trial for metastatic TNBC (TONIC trial, NCT02499367), using this workflow to mitigate risks of suboptimal inclusion of sTILs in this specific trial. In this review, we demonstrate that a web-based scoring platform can mitigate potential risk factors when including sTILs in clinical trials, and we argue that this framework can be applied for any future biomarker-driven clinical trial setting.