Background and Objectives:
Esophageal cancer is a major global malignancy, with particularly high incidence and mortality rates in East Asia. In China, esophageal squamous cell carcinoma (ESCC) is the predominant histological subtype, accounting for over 90% of cases. Due to its aggressive behavior and strong metastatic potential, ESCC is frequently diagnosed at an advanced stage, leading to a five-year survival rate of only 15–25%. Although cancer treatments have progressed significantly in recent years, therapeutic advancements for esophageal cancer remain relatively lagging behind other malignancies. Therefore, exploring more effective treatment strategies for ESCC is of critical importance.
ESCC is characterized by profound genomic and epigenetic heterogeneity, which plays a pivotal role in tumor progression, drug resistance, and metastasis. The immune landscape of ESCC featuring exhausted T cells, regulatory T cells (Tregs), and activated macrophages contributes significantly to tumor progression and immune evasion. This study aims to comprehensively profile the immune microenvironment of ESCC by analyzing peripheral blood, adjacent non-tumor tissue, and tumor tissue using state-of-the-art single-cell technologies. By identifying key immune cell subsets and correlating them with clinical outcomes, the findings aim to support the development of novel immunotherapeutic approaches.
Methods:
Two prospective clinical cohorts were collected and analyzed to evaluate the clinical efficacy and survival outcomes of neoadjuvant chemoradiotherapy and chemotherapy combined with immunotherapy. Further mechanistic studies were conducted to explore the underlying immune-related mechanisms. A total of 25 patients with ESCC were enrolled in this study. Tumor tissues, adjacent normal tissues, and peripheral blood samples were collected. Mass cytometry (CyTOF) was employed to perform an in-depth analysis of over 10 million cells using a panel of 42 immune markers. Single-cell analysis was used to characterize the immune cell composition and functional states across different tissue types. In addition, single-cell RNA sequencing (scRNA-seq) and multiplex immunohistochemistry (mIHC) were used to investigate the spatial distribution of immune cells within ESCC tissues. To explore the therapeutic potential of central memory T cells (TCM), TCM cells were isolated from tumor samples of four patients, expanded ex vivo, and co-cultured with ESCC cells to assess their cytotoxic function. Integrated analysis of single-cell transcriptomic and proteomic data helped identify key immunosuppressive markers in peripheral blood, adjacent tissues, and tumor tissues. Public cancer databases were used to evaluate the predictive and prognostic value of these markers in the context of immunotherapy. Additionally, a 4NQO-induced ESCC mouse model was established to examine differences in immunosuppressive marker expression between control and anti-PD-1 treatment groups.
Results:
Analysis of clinical benefits from neoadjuvant chemoradiotherapy or chemoimmunotherapy revealed that both strategies provided survival advantages. However, given the remarkable success of immunotherapy in esophageal cancer, its incorporation into neoadjuvant regimens may offer superior clinical value. Characterizing the tumor immune microenvironment is essential for optimizing immunotherapeutic strategies in esophageal cancer. CyTOF analysis revealed distinct differences in immune cell composition between tumor tissues, adjacent normal tissues, and peripheral blood in ESCC patients. Tumor tissues showed a significantly higher abundance of CD4+ T cells compared to adjacent tissues. Moreover, patients with lymph node metastases exhibited a notable increase in γδT cells in peripheral blood, suggesting that γδT cells could serve as biomarkers for metastatic disease and may play a role in systemic immune responses against tumor spread.
TCM cells exhibited potent tumor-killing effects, indicating their critical role in modulating anti-tumor immunity. Tumor-associated macrophages demonstrated high PD-L1 expression, which was associated with responsiveness to immunotherapy, highlighting their importance in shaping the immune response. Additionally, elevated CD39 expression in tumor-infiltrating T cells was linked to favorable prognosis and enhanced response to immunotherapy across multiple cancer types, suggesting CD39 as a promising biomarker for predicting patient outcomes and treatment efficacy.
Conclusion:
This study offers a comprehensive high-resolution immune profile of ESCC, revealing distinct immune cell compositions in blood, adjacent tissues, and tumor environments. The data suggest that enhancing TCM cell levels may boost the body’s anti-tumor capabilities. Furthermore, high PD-L1 expression in macrophages and increased CD39 and CD103 expression in tumor-infiltrating T cells may serve as reliable biomarkers for predicting responses to immunotherapy. These findings shed light on the immunological mechanisms underlying ESCC and lay a robust foundation for refining immunotherapeutic strategies. Future studies will focus on validating these results in clinical settings and exploring the potential for leveraging immune cell profiles to guide personalized ESCC treatment.