Role of tissue resident memory CD8+ T cells in triple negative breast cancer

Pathology ◽  
2019 ◽  
Vol 51 ◽  
pp. S98
Author(s):  
Dominique Yuan Bin Seow ◽  
Johnathan Lim ◽  
Clara Ong ◽  
Jeffrey Lim ◽  
Aye Aye Thike ◽  
...  
2019 ◽  
Vol 5 (1) ◽  
Author(s):  
Ana S. Leal ◽  
Kayla Zydeck ◽  
Sarah Carapellucci ◽  
Lyndsey A. Reich ◽  
Di Zhang ◽  
...  

Abstract Despite numerous therapeutic advances in the past decade, breast cancer is expected to cause over 42,000 deaths in the United States in 2019. Breast cancer had been considered an immunologically silent tumor; however recent findings suggest that immune cells play important roles in tumor growth even in the breast. Retinoid X receptors (RXRs) are a subclass of nuclear receptors that act as ligand-dependent transcription factors that regulate a variety of cellular processes including proliferation and differentiation; in addition, they are essential for macrophage biology. Rexinoids are synthetic molecules that bind and activate RXRs. Bexarotene is the only rexinoid approved by the FDA for the treatment of refractory cutaneous T-cell lymphoma. Other more-potent rexinoids have been synthesized, such as LG100268 (LG268). Here, we report that treatment with LG 268, but not bexarotene, decreased infiltration of myeloid-derived suppressor cells and CD206-expressing macrophages, increased the expression of PD-L1 by 50%, and increased the ratio of CD8/CD4, CD25 T cells, which correlates with increased cytotoxic activity of CD8 T cells in tumors of MMTV-Neu mice (a model of HER2-positive breast cancer). In the MMTV-PyMT murine model of triple negative breast cancer, LG268 treatment of established tumors prolonged survival, and in combination with anti-PD-L1 antibodies, significantly (p = 0.05) increased the infiltration of cytotoxic CD8 T cells and apoptosis. Collectively, these data suggest that the use of LG268, a RXR agonist, can improve response to immune checkpoint blockade in HER2+ or triple-negative breast cancer.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A365-A365
Author(s):  
Aaron Stevens ◽  
Joyce O’Shaughnessy ◽  
Subing Cao ◽  
Jessica Sorrentino ◽  
Janet Horton ◽  
...  

BackgroundTrilaciclib is an intravenous cyclin-dependent kinase 4/6 inhibitor approved to reduce the incidence of chemotherapy-induced myelosuppression in patients with extensive-stage small cell lung cancer (myeloprotection). In a randomized, open-label phase 2 trial in patients with metastatic triple-negative breast cancer (mTNBC), adding trilaciclib prior to gemcitabine/carboplatin (GCb) increased overall survival in both PD-L1–positive and –negative populations versus GCb alone.1 2 We investigated potential immune mechanisms of anti-tumor efficacy among patients who received trilaciclib plus GCb.MethodsPeripheral blood was collected prior to and on treatment for flow cytometric analysis, and total RNA isolated from diagnostic tumor biopsies for sequencing. Differential gene expression analysis between responders and non-responders was based on negative binomial distribution and related pathways identified by Kyoto Encyclopedia of Genes and Genomes pathway analysis. Tumor inflammation signatures and deconvolution-based approaches were used to assess the tumor immune microenvironment. PD-L1 expression was considered positive if ≥1% of the total tumor area contained PD-L1–labelled immune cells (Ventana SP142 assay). Patients were defined as responders (confirmed complete or partial response) or non-responders (stable or progressive disease) according to RECIST criteria.ResultsOf 68 patients who received trilaciclib prior to GCb, tumor response status and RNA sequencing data were available for 51 patients, comprising 24 responders and 27 non-responders. Tumors from responders had 253 differentially expressed genes compared with non-responders. Analysis of immune gene signatures revealed a higher T-cell exhaustion score at baseline among responders versus non-responders (P=0.044).Among patients with PD-L1–positive tumors, responders had a greater peripheral immune response at baseline compared with non-responders, including more T cells (P=0.037; particularly memory CD8 T cells [P=0.042]), and a trend toward fewer myeloid-derived suppressor cells (MDSCs). Additionally, tumors from responders had more dendritic cells (P=0.044) and a trend toward enriched tumor inflammation signatures compared with non-responders.By contrast, among patients with PD-L1–negative tumors, responders had similar peripheral immune populations at baseline compared with PD-L1–negative non-responders, but fewer MDSCs (P=0.016), and a trend toward increased T-cell numbers after two cycles of trilaciclib plus GCb.Responders with both PD-L1–positive and –negative tumors had increased numbers of naïve CD8 T cells after two treatment cycles compared with non-responders.ConclusionsThe data suggest that adding trilaciclib prior to GCb enhances antitumor efficacy by modulating the composition of immune cell subsets. The impact of trilaciclib on changes to the tumor-infiltrating immune response is being further investigated in a phase 3 trial in patients with mTNBC (NCT04799249).AcknowledgementsFlow cytometry and RNA sequencing analyses were performed by Covance, Inc., and Q2 Laboratory Solutions, respectively.Trial Registration www.clinicaltrials.govNCT02978716ReferencesTan AR, Wright GS, Thummala AR, Danso MA, Popovic L, Pluard TJ, Han HS, Vojnović Ž, Vasev N, Ma L, Richards DA, Wilks ST, Milenković D, Yang Z, Antal JM, Morris SR, O’Shaughnessy J. Trilaciclib plus chemotherapy versus chemotherapy alone in patients with metastatic triple-negative breast cancer: a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol 2019;20(11):1587–1601.O’Shaughnessy J, Wright GS, Thummala AR, Danso MA, Popovic L, Pluard TJ, Han HS, Vojnović Ž, Vasev N, Ma L, Richards DA, Wilks ST, Milenković D, Xiao J, Sorrentino JA, Horton J, Tan AR. Abstract PD1-06: trilaciclib improves overall survival when given with gemcitabine/carboplatin in patients with metastatic triple-negative breast cancer: final analysis of a randomized phase 2 trial. Cancer Res 2021;81(4 Supplement):PD1-06.Ethics ApprovalThe study protocol and all associated amendments and study-related materials were approved by the institutional review board or independent ethics committee of each investigational site.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A838-A838
Author(s):  
Erika Crosby ◽  
Hiroshi Nagata ◽  
Melinda Telli ◽  
Chaitanya Acharya ◽  
Irene Wapnir ◽  
...  

BackgroundTriple-negative breast cancer (TNBC) is an aggressive disease with limited therapeutic options. Immune checkpoint inhibitors (ICI) have entered the therapeutic landscape in TNBC, but only a minority of patients benefit. Interleukin-12 (IL-12) is a pro-inflammatory cytokine involved in the generation of an inflammatory tumor microenvironment and is critical in eliciting a productive anti-tumor immune response. It has been investigated as an anti-cancer therapeutic using various delivery routes, but intratumoral injection of plasmid IL-12 (tavokinogene telseplasmid; tavo) followed by electroporation is a gene therapy approach with minimal systemic immune-related toxicity.MethodsIntratumoral injection of tavo was tested in several preclinical models of TNBC and single cell RNA sequencing (scRNAseq) was used to evaluate changes in the tumor microenvironment following treatment. These findings were then applied to the analysis of patient samples from a single arm, prospective clinical trial of tavo monotherapy (OMS-I140; NCT02531425).ResultsA comprehensive analysis of cellular networks using ligand-receptor interactions identified CXCR3 (expressed by APCs) to be positively correlated with CXCL9/10/11 secreted by CD8 T cells. Further investigation of tavo treated murine tumors resulted in a 50-gene CXCR3 gene expression signature that is associated with a decrease in granulocytes, enhanced antigen presentation, increased T cell infiltration, and induction of PD-1/PD-L1. A deeper look at paired TCR alpha and beta chains on tumor infiltrating T cells (TILs) demonstrated a dramatic shift in TIL clonality and frequency following tavo treatment. There was a significant increase in not only the number of expanded (>10) clones, but also a robust activation signature that was absent in control tumors. Treatment of mice with tavo prior to anti-PD1 therapy led to complete tumor regression and long-term survival in a significant proportion of mice, while none of the mice treated with anti-PD1 alone exhibited this therapeutic efficacy. As a proof of concept, we utilized nanostring data from OMS-I140 to show a significant enhancement in this signature in patients who demonstrated a greater than 2-fold increase in CD8 TILS by IHC post-treatment. Further, we show a single patient who had previously been non-responsive to ICI that went on to receive anti-PD1 as their immediate next treatment after participating in OMS-I140, and demonstrated a significant clinical response.ConclusionsTogether these data identify a clinically relevant CXCR3-associated gene signature that represents both a potential biomarker for response to ICIs and a potentially targetable pathway for therapeutic intervention in TNBC.Ethics ApprovalAll animal studies described were approved by the Duke University Medical Center Institutional Animal Care & Use Committee (A198-18-08) and performed in accordance with established guidelines.


2017 ◽  
Vol 5 (6) ◽  
pp. 439-445 ◽  
Author(s):  
Michiko Harao ◽  
Marie-Andrée Forget ◽  
Jason Roszik ◽  
Hui Gao ◽  
Gildy V. Babiera ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document