Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
NPJ Breast Cancer ; 9(1): 68, 2023 Aug 15.
Article En | MEDLINE | ID: mdl-37582853

Novel therapeutic strategies that can effectively combine with immunotherapies are needed in the treatment of triple-negative breast cancer (TNBC). We demonstrate that combined PARP and WEE1 inhibition are synergistic in controlling tumour growth in BRCA1/2 wild-type TNBC preclinical models. The PARP inhibitor (PARPi) olaparib combined with the WEE1 inhibitor (WEE1i) adavosertib triggered increases in anti-tumour immune responses, including STING pathway activation. Combinations with a STING agonist resulted in further improved durable tumour regression and significant improvements in survival outcomes in murine tumour models of BRCA1/2 wild-type TNBC. In addition, we have identified baseline tumour-infiltrating lymphocyte (TIL) levels as a potential predictive biomarker of response to PARPi, WEE1i and immunotherapies in BRCA1/2 wild-type TNBC.

2.
Nat Commun ; 12(1): 1434, 2021 03 04.
Article En | MEDLINE | ID: mdl-33664264

Although melanoma is initiated by acquisition of point mutations and limited focal copy number alterations in melanocytes-of-origin, the nature of genetic changes that characterise lethal metastatic disease is poorly understood. Here, we analyze the evolution of human melanoma progressing from early to late disease in 13 patients by sampling their tumours at multiple sites and times. Whole exome and genome sequencing data from 88 tumour samples reveals only limited gain of point mutations generally, with net mutational loss in some metastases. In contrast, melanoma evolution is dominated by whole genome doubling and large-scale aneuploidy, in which widespread loss of heterozygosity sculpts the burden of point mutations, neoantigens and structural variants even in treatment-naïve and primary cutaneous melanomas in some patients. These results imply that dysregulation of genomic integrity is a key driver of selective clonal advantage during melanoma progression.


Aneuploidy , DNA Copy Number Variations/genetics , Genome, Human/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Disease Progression , Exome/genetics , Humans , INDEL Mutation/genetics , Melanocytes/pathology , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Exome Sequencing , Whole Genome Sequencing , Melanoma, Cutaneous Malignant
3.
Nat Med ; 24(12): 1941, 2018 Dec.
Article En | MEDLINE | ID: mdl-30135555

In the version of this article originally published, the institution in affiliation 10 was missing. Affiliation 10 was originally listed as Department of Surgery, Royal Melbourne Hospital and Royal Womens' Hospital, Melbourne, Victoria, Australia. It should have been Department of Surgery, Royal Melbourne Hospital and Royal Womens' Hospital, University of Melbourne, Melbourne, Victoria, Australia. The error has been corrected in the HTML and PDF versions of this article.

4.
Nat Med ; 24(7): 986-993, 2018 07.
Article En | MEDLINE | ID: mdl-29942092

The quantity of tumor-infiltrating lymphocytes (TILs) in breast cancer (BC) is a robust prognostic factor for improved patient survival, particularly in triple-negative and HER2-overexpressing BC subtypes1. Although T cells are the predominant TIL population2, the relationship between quantitative and qualitative differences in T cell subpopulations and patient prognosis remains unknown. We performed single-cell RNA sequencing (scRNA-seq) of 6,311 T cells isolated from human BCs and show that significant heterogeneity exists in the infiltrating T cell population. We demonstrate that BCs with a high number of TILs contained CD8+ T cells with features of tissue-resident memory T (TRM) cell differentiation and that these CD8+ TRM cells expressed high levels of immune checkpoint molecules and effector proteins. A CD8+ TRM gene signature developed from the scRNA-seq data was significantly associated with improved patient survival in early-stage triple-negative breast cancer (TNBC) and provided better prognostication than CD8 expression alone. Our data suggest that CD8+ TRM cells contribute to BC immunosurveillance and are the key targets of modulation by immune checkpoint inhibition. Further understanding of the development, maintenance and regulation of TRM cells will be crucial for successful immunotherapeutic development in BC.


Breast Neoplasms/immunology , Immunologic Memory , Single-Cell Analysis/methods , Breast Neoplasms/pathology , CD3 Complex/metabolism , CD8 Antigens/metabolism , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Sequence Analysis, RNA , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
5.
Nat Commun ; 8(1): 606, 2017 09 19.
Article En | MEDLINE | ID: mdl-28928458

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.


4-1BB Ligand/agonists , Cell Proliferation/drug effects , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Mammary Neoplasms, Animal/immunology , OX40 Ligand/agonists , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , T-Lymphocyte Subsets/drug effects , Triple Negative Breast Neoplasms/immunology , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , T-Lymphocytes/drug effects
6.
Sci Transl Med ; 9(393)2017 06 07.
Article En | MEDLINE | ID: mdl-28592566

Immune checkpoint inhibitors have emerged as a potent new class of anticancer therapy. They have changed the treatment landscape for a range of tumors, particularly those with a high mutational load. To date, however, modest results have been observed in breast cancer, where tumors are rarely hypermutated. Because BRCA1-associated tumors frequently exhibit a triple-negative phenotype with extensive lymphocyte infiltration, we explored their mutational load, immune profile, and response to checkpoint inhibition in a Brca1-deficient tumor model. BRCA1-mutated triple-negative breast cancers (TNBCs) exhibited an increased somatic mutational load and greater numbers of tumor-infiltrating lymphocytes, with increased expression of immunomodulatory genes including PDCD1 (PD-1) and CTLA4, when compared to TNBCs from BRCA1-wild-type patients. Cisplatin treatment combined with dual anti-programmed death-1 and anti-cytotoxic T lymphocyte-associated antigen 4 therapy substantially augmented antitumor immunity in Brca1-deficient mice, resulting in an avid systemic and intratumoral immune response. This response involved enhanced dendritic cell activation, reduced suppressive FOXP3+ regulatory T cells, and concomitant increase in the activation of tumor-infiltrating cytotoxic CD8+ and CD4+ T cells, characterized by the induction of polyfunctional cytokine-producing T cells. Dual (but not single) checkpoint blockade together with cisplatin profoundly attenuated the growth of Brca1-deficient tumors in vivo and improved survival. These findings provide a rationale for clinical studies of combined immune checkpoint blockade in BRCA1-associated TNBC.


BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Mutation/genetics , Tumor Suppressor Proteins/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/metabolism , Breast Neoplasms/drug therapy , Cell Proliferation , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology
7.
Int J Oncol ; 43(1): 194-200, 2013 Jul.
Article En | MEDLINE | ID: mdl-23615977

Growing evidence suggests that the flavonoid epigallocatechin-3-gallate (EGCG), notably abundant in green tea, has health-promoting properties. We examined the effect of EGCG on cell survival and apoptosis in the prostate cancer cell line PC3. Cell survival was reduced and apoptosis increased significantly with a low dose of 1 µM EGCG. The ability of the anticancer drug cisplatin to promote apoptosis was enhanced by EGCG. Furthermore, EGCG, both alone and in combination with cisplatin, promoted the expression of the pro-apoptotic splice isoform of caspase 9.


Apoptosis/drug effects , Caspase 9/genetics , Catechin/analogs & derivatives , Prostatic Neoplasms/genetics , Caspase 9/biosynthesis , Catechin/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Isoforms/genetics
...