Abstract

BackgroundGlioma is the most common type of primary brain tumor in adults. Patients with the most malignant form have an overall survival time of <16 months. Although considerable progress has been made in defining the adapted therapeutic strategies, measures to counteract tumor escape have not kept pace, due to the developed resistance of malignant glioma. In fact, identifying the nature and role of distinct tumor-infiltrating immune cells in glioma patients would decipher potential mechanisms behind therapy failure.MethodsWe integrated into our study glioma transcriptomic datasets from the Cancer Genome Atlas (TCGA) cohort (154 GBM and 516 LGG patients). LM22 immune signature was built using CIBERSORT. Hierarchical clustering and UMAP dimensional reduction algorithms were applied to identify clusters among glioma patients either in an unsupervised or supervised way. Furthermore, differential gene expression (DGE) has been performed to unravel the top expressed genes among the identified clusters. Besides, we used the least absolute shrinkage and selection operator (LASSO) and Cox regression algorithm to set up the most valuable prognostic factor.ResultsOur study revealed, following gene enrichment analysis, the presence of two distinct groups of patients. The first group, defined as cluster 1, was characterized by the presence of immune cells known to exert efficient antitumoral immune response and was associated with better patient survival, whereas the second group, cluster 2, which exhibited a poor survival, was enriched with cells and molecules, known to set an immunosuppressive pro-tumoral microenvironment. Interestingly, we revealed that gene expression signatures were also consistent with each immune cluster function. A strong presence of activated NK cells was revealed in cluster 1. In contrast, potent immunosuppressive components such as regulatory T cells, neutrophils, and M0/M1/M2 macrophages were detected in cluster 2, where, in addition, inhibitory immune checkpoints, such as PD-1, CTLA-4, and TIM-3, were also significantly upregulated. Finally, Cox regression analysis further corroborated that tumor-infiltrating cells from cluster 2 exerted a significant impact on patient prognosis.ConclusionOur work brings to light the tight implication of immune components on glioma patient prognosis. This would contribute to potentially developing better immune-based therapeutic approaches.

Highlights

  • Glioma is the most common type of primary brain tumors in adults, with the most aggressive form known as glioblastoma

  • Potent immunosuppressive components such as regulatory T cells, neutrophils, and M0/M1/ M2 macrophages were detected in cluster 2, where, in addition, inhibitory immune checkpoints, such as programmed-death receptor-1 (PD-1), cytotoxic T lymphocyte antigen-4 (CTLA-4), and TIM-3, were significantly upregulated

  • Our work brings to light the tight implication of immune components on glioma patient prognosis

Read more

Summary

Introduction

Glioma is the most common type of primary brain tumors in adults, with the most aggressive form known as glioblastoma. Glioma patients are known to be extremely resistant to chemotherapy. Based on histological and molecular features, glioma patients can be stratified into oligodendroglioma or astrocytoma (LGG) and glioblastoma (HGG) with clinically relevant molecular subtypes including mesenchymal, proneural, neural, and classical. These features can be associated with tumor niche (stromal cells, reactive astrocytes, tumor cells, and immune cells), patient survival, and prognosis [6, 7]. Identifying the nature and role of distinct tumorinfiltrating immune cells in glioma patients would decipher potential mechanisms behind therapy failure

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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