Abstract

BackgroundTumor progression is accompanied by dramatic remodeling of the surrounding extracellular matrix leading to the formation of a tumor-specific ECM, which is often more collagen-rich and of increased stiffness. The altered ECM of the tumor supports cancer growth and metastasis, but it is unknown if this effect involves modulation of T cell activity. To investigate if a high-density tumor-specific ECM could influence the ability of T cells to kill cancer cells, we here studied how T cells respond to 3D culture in different collagen densities.MethodsT cells cultured in 3D conditions surrounded by a high or low collagen density were imaged using confocal fluorescent microscopy. The effects of the different collagen densities on T cell proliferation, survival, and differentiation were examined using flow cytometry. Cancer cell proliferation in similar 3D conditions was also measured. Triple-negative breast cancer specimens were analyzed for the number of infiltrating CD8+ T cells and for the collagen density. Whole-transcriptome analyses were applied to investigate in detail the effects of collagen density on T cells. Computational analyses were used to identify transcription factors involved in the collagen density-induced gene regulation. Observed changes were confirmed by qRT-PCR analysis.ResultsT cell proliferation was significantly reduced in a high-density matrix compared to a low-density matrix and prolonged culture in a high-density matrix led to a higher ratio of CD4+ to CD8+ T cells. The proliferation of cancer cells was unaffected by the surrounding collagen-density. Consistently, we observed a reduction in the number of infiltrating CD8+ T-cells in mammary tumors with high collagen-density indicating that collagen-density has a role in regulating T cell abundance in human breast cancer.Whole-transcriptome analysis of 3D-cultured T cells revealed that a high-density matrix induces downregulation of cytotoxic activity markers and upregulation of regulatory T cell markers. These transcriptional changes were predicted to involve autocrine TGF-β signaling and they were accompanied by an impaired ability of tumor-infiltrating T cells to kill autologous cancer cells.ConclusionsOur study identifies a new immune modulatory mechanism, which could be essential for suppression of T cell activity in the tumor microenvironment.

Highlights

  • Tumor progression is accompanied by dramatic remodeling of the surrounding extracellular matrix leading to the formation of a tumor-specific ECM, which is often more collagen-rich and of increased stiffness

  • As an important way for the cancer cells to evade immune destruction, tumors can develop a strongly immunosuppressive tumor microenvironment [4,5,6]. This includes the accumulation of cell types with immunosuppressive activity, such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory target cells [43]. Effector cells (T cells) (Tregs) [4]

  • Analysis of the distribution of the different T cell differentiation subsets after 5 days of 3D culture in a high- or low-density collagen matrix or regular 2D culture (Additional file 1: Figure S2) suggested an increase in effector memory T cells and a decrease in central memory T cells after 3D culture in a high-density collagen matrix compared to a low-density collagen matrix

Read more

Summary

Introduction

Tumor progression is accompanied by dramatic remodeling of the surrounding extracellular matrix leading to the formation of a tumor-specific ECM, which is often more collagen-rich and of increased stiffness. To investigate if a high-density tumor-specific ECM could influence the ability of T cells to kill cancer cells, we here studied how T cells respond to 3D culture in different collagen densities. Solid tumors consist of cancer cells interacting with the tumor microenvironment, which includes stromal cells, immune cells, and the extracellular matrix. As an important way for the cancer cells to evade immune destruction, tumors can develop a strongly immunosuppressive tumor microenvironment [4,5,6]. This includes the accumulation of cell types with immunosuppressive activity, such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs) [4]. Antibody-mediated blockade of the PD1-PD-L1 interaction has demonstrated remarkable clinical efficacy for many cancer patients and stimulated the research aiming at identifying additional targetable immunosuppressive mechanisms in the tumor microenvironment [7]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call