Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 7 of 7
  • 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)
    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
    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
    Clinical validity and Utility of Tumor-infiltrating Lymphocytes in Routine Clinical Practice for Breast Cancer Patients: Current and Future Directions
    Wein, L ; Savas, P ; Luen, SJ ; Virassamy, B ; Salgado, R ; Loi, S (FRONTIERS MEDIA SA, 2017-08-03)
    The interest in tumor-infiltrating lymphocytes (TILs) as a prognostic biomarker in breast cancer has grown in recent years. Biomarkers must undergo comprehensive evaluation in terms of analytical validity, clinical validity and clinical utility before they can be accepted as part of clinical practice. The International Immuno-Oncology Biomarker Working Group has developed a practice guideline on scoring TILs in breast cancer in order to standardize TIL assessment. The prognostic value of TILs as a biomarker in early-stage breast cancer has been established by assessing tumor samples in thousands of patients from large prospective clinical trials of adjuvant therapy. There is a strong linear relationship between increase in TILs and improved disease-free survival for triple-negative and HER2-positive disease. Higher levels of TILs have also been associated with increased rates of pathological complete response to neoadjuvant therapy. TILs have potential clinical utility in breast cancer in a number of areas. These include prediction of responders to immune checkpoint blockade, identification of primary HER2-positive and triple-negative patients who have excellent prognoses and may thus be appropriate for treatment de-escalation, and potentially incorporation into a neoadjuvant endpoint which may be a better surrogate maker for drug development.
  • 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
    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.