232 The epithelial-to-mesenchymal transition (EMT) contributes to immunosuppression in breast carcinomas and regulates their response to immune checkpoint blockade

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A250-A250
Author(s):  
Anushka Dongre ◽  
Robert Weinberg ◽  
Mohammad Rashidian ◽  
Elinor Eaton ◽  
Ferenc Reinhardt ◽  
...  

BackgroundImmune checkpoint blockade (ICB) has generated some dramatic responses in certain types of human tumors, most notably, melanomas. However, the response of breast tumors has been largely limited. We have previously demonstrated that the residence of breast cancer cells in the epithelial or mesenchymal phenotypic states can itself be used as an important determinant of the success or failure of ICB. Specifically, we have shown that while epithelial tumors are sensitive to anti-CTLA4, mesenchymal tumors are highly resistant. Most strikingly, in tumors arising from a mixture of both cell types, a minority population (10%) of mesenchymal cells can cross-protect the vast majority (90%) of their epithelial neighbors from immune attack.1 However, the mechanisms underlying such cross-protection remain elusive. This is particularly important as most human breast cancers contain minority populations of such mesenchymal cells which can protect the tumor as a whole from immune attack.MethodsUsing a combination of transcriptomic and CRISPR/Cas9 approaches, we first identified that mesenchymal carcinoma cells express high levels of CD73, an ecto-enzyme that catalyzes the production of adenosine. Additionally, we used digital spatial profiling to determine whether CD73 expression differs across distinct epithelial and mesenchymal sectors in mixed tumors.ResultsAbrogation of CD73 from mesenchymal carcinoma cells prevented the assembly of an immunosuppressive tumor microenvironment and resulted instead in increased numbers of tumor-infiltrating lymphocytes and cross-presenting dendritic cells. Most strikingly, abrogation of CD73 sensitized previously refractory mesenchymal tumors completely to ICB. In the context of mixed tumors comprised of both epithelial and mesenchymal carcinoma cells, gradients in expression of CD73 were observed corresponding to the epithelial or mesenchymal sectors of these mixed tumors. Importantly, mixed tumors in which the minority population of mesenchymal carcinoma cells lacked the expression of CD73, were also sensitized partially to ICB. Thus, these mesenchymal carcinoma cells knocked out for CD73 could no longer protect their epithelial neighbors from immune attack.ConclusionsTaken together, our work suggests that mesenchymal carcinoma cells exert immune-suppressive effects which are also prominent in heterogeneous tumors. Furthermore, targeting the adenosinergic signaling pathway in mesenchymal carcinoma cells can potentiate the efficacy of ICB.ReferenceDongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, Ploegh HL, Weinberg RA, Epithelial-to-mesenchymal transition contributes to immunosuppression in breast carcinomas. Cancer Res 2017 Aug 1;77(15):3982–3989.

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Wenjing Qian ◽  
Mingfang Zhao ◽  
Ruoyu Wang ◽  
Heming Li

AbstractImmune checkpoint therapy has achieved significant efficacy by blocking inhibitory pathways to release the function of T lymphocytes. In the clinic, anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) monoclonal antibodies (mAbs) have progressed to first-line monotherapies in certain tumor types. However, the efficacy of anti-PD-1/PD-L1 mAbs is still limited due to toxic side effects and de novo or adaptive resistance. Moreover, other immune checkpoint target and biomarkers for therapeutic response prediction are still lacking; as a biomarker, the PD-L1 (CD274, B7-H1) expression level is not as accurate as required. Hence, it is necessary to seek more representative predictive molecules and potential target molecules for immune checkpoint therapy. Fibrinogen-like protein 1 (FGL1) is a proliferation- and metabolism-related protein secreted by the liver. Multiple studies have confirmed that FGL1 is a newly emerging checkpoint ligand of lymphocyte activation gene 3 (LAG3), emphasizing the potential of targeting FGL1/LAG3 as the next generation of immune checkpoint therapy. In this review, we summarize the substantial regulation mechanisms of FGL1 in physiological and pathological conditions, especially tumor epithelial to mesenchymal transition, immune escape and immune checkpoint blockade resistance, to provide insights for targeting FGL1 in cancer treatment.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A779-A779
Author(s):  
Michelle Williams ◽  
Jessica Christenson ◽  
Kathleen O’Neill ◽  
Sabrina Hafeez ◽  
Nicole Spoelstra ◽  
...  

BackgroundTo identify novel molecular mechanisms used by triple negative breast cancer (TNBC) to facilitate metastasis, we manipulated oncogenic epithelial-to-mesenchymal transition (EMT) by restoring the microRNA-200c (miR-200c), termed ‘the guardian of the epithelial phenotype.’ We identified several tumor cell catabolizing enzymes, including tryptophan 2,3-dioxygenase (TDO2) and heme oxygenase-1 (HO-1). The Richer lab has published that TDO2 promotes anchorage independent cell survival during TNBC metastasis via its catabolite kynurenine, which also induces CD8+ T cell death. Similarly, published studies have demonstrated that HO-1 supports BC anchorage independent survival. However, effects of the HO-1 catabolite bilirubin on the tumor microenvironment had not been studied. We postulated that TNBC utilize targetable catabolizing enzymes, like HO-1, to simultaneously support tumor cell survival and dampen the anti-tumor immune response.MethodsTo test our hypothesis in an immune competent mouse model, Met-1 mammary carcinoma cells from a late stage MMTV-PyMT tumor were engineered to inducibly express miR-200c. Tumor cell infiltrates were analyzed by immunohistochemistry (IHC), flow cytometry and multispectral fluorescence. RAW264.7 mouse macrophages were cultured with conditioned medium from carcinoma cells ± miR-200c or the HO-1 competitive inhibitor tin mesoporphyrin (SnMP). RAW264.7 macrophages were also treated with 0–20 µM bilirubin and macrophage polarization and efferocytic capacity, the ability to engulf dead tumor cells, were assessed using qRT-PCR and IncuCyte assays.ResultsMiR-200c restoration to Met-1 orthotopic tumors decreased growth by 45% and increased infiltration of CD11c+ dendritic cells and activation, determined by CD44 expression, of CD4+ and CD8+ T cells. While the number of F4/80+ macrophages was unchanged by miR-200c, the percent of M1 anti-tumor macrophages (F4/80+iNOS+/total cells) increased by >6-fold in miR-200c+tumors. RAW264.7 macrophages cultured with conditioned medium from miR-200c-restored mammary carcinoma cells had a 25–95% decrease in M2 pro-tumor genes (Arg1, Il4 and Il13) and a 15–55% increase in M1 genes (Nos2, Tnfa and Cxcl10). A similar decrease in M2 (30–50%) and increase M1 (35–160%) genes was seen in macrophages cultured with conditioned medium from SnMP treated mammary carcinoma cells. Conversely, bilirubin treatment alone enhanced M2 macrophage polarization and inhibited efferocytosis in a dose-dependent manner.ConclusionsUse of miR-200c to reverse EMT revealed that HO-1 promotes simultaneous TNBC cell survival and immune suppression. These studies are the first to show that tumor cell-HO-1 activity and subsequent bilirubin production may alter macrophage function in the tumor microenvironment. This finding could be clinically relevant since HO-1 inhibitors like SnMP are already FDA approved for treatment of other diseases.


Cells ◽  
2018 ◽  
Vol 7 (7) ◽  
pp. 78 ◽  
Author(s):  
Julien Guinde ◽  
Diane Frankel ◽  
Sophie Perrin ◽  
Valérie Delecourt ◽  
Nicolas Lévy ◽  
...  

Lung cancer represents the primary cause of cancer death in the world. Malignant cells identification and characterization are crucial for the diagnosis and management of patients with primary or metastatic cancers. In this context, the identification of new biomarkers is essential to improve the differential diagnosis between cancer subtypes, to select the most appropriate therapy, and to establish prognostic correlations. Nuclear abnormalities are hallmarks of carcinoma cells and are used as cytological diagnostic criteria of malignancy. Lamins (divided into A- and B-types) are localized in the nuclear matrix comprising nuclear lamina, where they act as scaffolding protein, involved in many nuclear functions, with regulatory effects on the cell cycle and differentiation, senescence and apoptosis. Previous studies have suggested that lamins are involved in tumor development and progression with opposite results concerning their prognostic role. This review provides an overview of lamins expression in lung cancer and the relevance of these findings for disease diagnosis and prognosis. Furthermore, we discuss the link between A-type lamins expression in lung carcinoma cells and nuclear deformability, epithelial to mesenchymal transition, and metastatic potential, and which mechanisms could regulate A-type lamins expression in lung cancer, such as the microRNA miR-9.


Sign in / Sign up

Export Citation Format

Share Document