Anaplastic Thyroid Cancer (ATC) is an aggressive form of cancer with poor prognosis, heavily influenced by its tumor immune microenvironment (TIME). Understanding the cellular and gene expression dynamics within the TIME is crucial for developing targeted therapies. This study analyzes the immune microenvironment of ATC and Papillary Thyroid Cancer (PTC) using single-cell RNA sequencing (scRNA-seq). We performed a comprehensive scRNA-seq analysis on ATC and PTC samples, incorporating cell type annotation, marker gene identification, and clustering based on gene expression. A specific focus was on the prevalence and biomarkers of Pre-Exhausted CD8+ T cells in ATC, utilizing the single-cell tumor immune atlas for immune cell characterization. The scRNA-seq analysis identified distinct immune cell populations and differentially expressed genes in ATC and PTC samples. Notably, Pre-Exhausted CD8+ T cells were found to be prevalent in ATC datasets. Additional immunofluorescence staining and co-culture experiments with the ATC cell line identified GNLY, a member of the saposin-like protein family as a potential biomarker for Pre-Exhausted CD8+ T cells in ATC. This study provides valuable insights into the immune landscape of ATC, emphasizing the prevalence of Pre-Exhausted CD8+ T cells and identifying GNLY as a potential biomarker. Understanding the TIME composition and the role of specific immune cells in cancer progression can inform the development of targeted immunotherapies for ATC. Future research should explore the functional implications of GNLY and other identified biomarkers in modulating the immune response in thyroid cancer.

Download full-text PDF

Source
http://dx.doi.org/10.1530/ERC-24-0169DOI Listing

Publication Analysis

Top Keywords

pre-exhausted cd8+
20
cd8+ cells
20
thyroid cancer
16
tumor immune
12
immune microenvironment
12
atc
10
immune
9
anaplastic thyroid
8
gene expression
8
scrna-seq analysis
8

Similar Publications

Anaplastic Thyroid Cancer (ATC) is an aggressive form of cancer with poor prognosis, heavily influenced by its tumor immune microenvironment (TIME). Understanding the cellular and gene expression dynamics within the TIME is crucial for developing targeted therapies. This study analyzes the immune microenvironment of ATC and Papillary Thyroid Cancer (PTC) using single-cell RNA sequencing (scRNA-seq).

View Article and Find Full Text PDF

Lymphatic system regulation of anti-cancer immunity and metastasis.

Front Immunol

August 2024

Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.

Cancer dissemination to lymph nodes (LN) is associated with a worse prognosis, increased incidence of distant metastases and reduced response to therapy. The LN microenvironment puts selective pressure on cancer cells, creating cells that can survive in LN as well as providing survival advantages for distant metastatic spread. Additionally, the presence of cancer cells leads to an immunosuppressive LN microenvironment, favoring the evasion of anti-cancer immune surveillance.

View Article and Find Full Text PDF

Background: Neoadjuvant chemotherapy (NAC) combined with immunotherapy is increasingly used in non-small cell lung cancer (NSCLC). Tissue-resident memory T (T) cells are the primary subset responding to anti-cancer immunity. However, the immunomodulatory effects of NAC on T cells remain unknown.

View Article and Find Full Text PDF

A spatial architecture-embedding HLA signature to predict clinical response to immunotherapy in renal cell carcinoma.

Nat Med

June 2024

Laboratory of Cell Stress and Immunity (CSI), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.

An important challenge in the real-world management of patients with advanced clear-cell renal cell carcinoma (aRCC) is determining who might benefit from immune checkpoint blockade (ICB). Here we performed a comprehensive multiomics mapping of aRCC in the context of ICB treatment, involving discovery analyses in a real-world data cohort followed by validation in independent cohorts. We cross-connected bulk-tumor transcriptomes across >1,000 patients with validations at single-cell and spatial resolutions, revealing a patient-specific crosstalk between proinflammatory tumor-associated macrophages and (pre-)exhausted CD8 T cells that was distinguished by a human leukocyte antigen repertoire with higher preference for tumoral neoantigens.

View Article and Find Full Text PDF
Article Synopsis
  • TIGIT is an inhibitory receptor that competes with CD226 on immune cells, and targeting it may improve anti-tumor immunity, especially in exhausted CD8+ T cells and regulatory T cells.
  • Combining different types of anti-TIGIT antibodies (Fc-enabled and Fc-silent) with another treatment showed varied effects on tumors: Fc-enabled antibodies reduced regulatory T cell numbers, while Fc-silent antibodies did not deplete these cells but enhanced the activity of tumor-specific CD8+ T cells.
  • The study highlights that Fc-silent anti-TIGIT antibodies can drive antitumor responses by activating exhausted T cells without affecting regulatory T cells, indicating their potential for cancer therapy.
View Article and Find Full Text PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!