Integrative analyses of single-cell and bulk RNA sequencing reveal tumour microenvironment features associated with neoadjuvant immunochemotherapy response in oesophageal squamous cell carcinoma

Xianxian Wu , Xiaoxing Ye , Wei Ji , Xiangyang Yu , Ahuan Xie , Zichang Xiang , Zhilin Sui , Jiquan Tang , Zhentao Yu

Clinical and Translational Discovery ›› 2025, Vol. 5 ›› Issue (4) : e70080

PDF
Clinical and Translational Discovery ›› 2025, Vol. 5 ›› Issue (4) : e70080 DOI: 10.1002/ctd2.70080
RESEARCH ARTICLE

Integrative analyses of single-cell and bulk RNA sequencing reveal tumour microenvironment features associated with neoadjuvant immunochemotherapy response in oesophageal squamous cell carcinoma

Author information +
History +
PDF

Abstract

Background: Neoadjuvant chemotherapy combined with immunotherapy (NACI) has shown promise in oesophageal squamous cell carcinoma (ESCC). However, a significant proportion of patients exhibit resistance to NACI, and the underlying mechanisms remain unresolved.

Methods: We integrated single-cell RNA sequencing data, including seven patients with ESCC treated with NACI and 69 patients with ESCC treated with surgery alone. Bulk RNA sequencing data were obtained from a public database. Immunohistochemistry and multiplexed immunofluorescence staining were performed to verify the role of important immune cells and molecules in clinical treatment outcomes.

Results: Here, we profiled the transcriptomes of 512 736 cells from 76 patients with ESCC, revealing that the nonresponder baseline tumour microenvironment exhibited a relative absence of major histocompatibility complex II molecules expressed on CD20+B cells and a low expression of CXCL13 on CD4_Tfh and CD8_Tex cells. We also identified CD68+CD163+ macrophages that highly expressed the immunosuppressive LGALS9 gene and preferentially accumulated in the nonresponders after NACI treatment. In addition, nonresponders had a higher baseline fraction of POSTN+fibroblasts, which is associated with higher infiltration of CD68+CD163+ macrophages and lower infiltration of germinal centre B cells. Finally, we described the different characteristics of malignant epithelial cells from different pathological responses to tumours.

Conclusions: This study has unveiled a potential regulatory network among immune cells, stromal cells and malignant epithelial cells under different pathological response conditions and provides a valuable resource for discovering novel targeted therapies for ESCC.

Keywords

immunotherapy / neoadjuvant chemotherapy / oesophageal squamous cell carcinoma / single-cell RNA sequencing / tumour microenvironment

Cite this article

Download citation ▾
Xianxian Wu, Xiaoxing Ye, Wei Ji, Xiangyang Yu, Ahuan Xie, Zichang Xiang, Zhilin Sui, Jiquan Tang, Zhentao Yu. Integrative analyses of single-cell and bulk RNA sequencing reveal tumour microenvironment features associated with neoadjuvant immunochemotherapy response in oesophageal squamous cell carcinoma. Clinical and Translational Discovery, 2025, 5(4): e70080 DOI:10.1002/ctd2.70080

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2021; 71: 209-249.

[2]

Thrift AP. Global burden and epidemiology of Barrett oesophagus and oesophageal cancer. Nat Rev Gastroenterol Hepatol. 2021; 18: 432-443.

[3]

Yi M, Zheng XL, Niu M, et al. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022; 21: 28.

[4]

Wu HX, Pan YQ, He Y, et al. Clinical benefit of first-line programmed death-1 antibody plus chemotherapy in low programmed cell death ligand 1-expressing esophageal squamous cell carcinoma: a post hoc analysis of JUPITER-06 and meta-analysis. J Clin Oncol. 2022; 41: 1735-1746.

[5]

Liu J, Yang Y, Liu ZC, et al. Multicenter, single-arm, phase II trial of camrelizumab and chemotherapy as neoadjuvant treatment for locally advanced esophageal squamous cell carcinoma. J Immunother Cancer. 2022; 10:e004291.

[6]

Chen XF, Xu X, Wang D, et al. Neoadjuvant sintilimab and chemotherapy in patients with potentially resectable esophageal squamous cell carcinoma (KEEP-G 03): an open-label, single-arm, phase 2 trial. J Immunother Cancer. 2023; 11:e005830.

[7]

Bejarano L, Jordāo MJC, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021; 11: 933-959.

[8]

Kim R An M, Lee H, et al. Early tumor-immune microenvironmental remodeling and response to first-line fluoropyrimidine and platinum chemotherapy in advanced gastric cancer. Cancer Discov. 2021; 12: 984-1001.

[9]

Qin P, Chen H, Wang Y, et al. Cancer-associated fibroblasts undergoing neoadjuvant chemotherapy suppress rectal cancer revealed by single-cell and spatial transcriptomics. Cell Rep Med. 2023; 4:101231.

[10]

Liu ZC, Zhang Y, Ma N, et al. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 2023; 41: 1852-1870.e9.

[11]

Zhang D, Jiang DX, Jiang L, et al. HLA-A(+) tertiary lymphoid structures with reactivated tumor infiltrating lymphocytes are associated with a positive immunotherapy response in esophageal squamous cell carcinoma. Br J Cancer. 2024; 131: 184-195.

[12]

Mao X, Xu J, Wang W, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021; 20: 131.

[13]

Niu B, Liao K, Zhou Y, et al. Application of glutathione depletion in cancer therapy: enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials. 2021; 277:121110.

[14]

Zhang W, Dai J, Hou G, et al. SMURF2 predisposes cancer cell toward ferroptosis in GPX4-independent manners by promoting GSTP1 degradation. Mol Cell. 2023; 83: 4352-4369.e8.

[15]

Ji G, Yang Q, Wang S, et al. Single-cell profiling of response to neoadjuvant chemo-immunotherapy in surgically resectable esophageal squamous cell carcinoma. Genome Med. 2024; 16: 49.

[16]

Wang Z, Zhao Y, Wo Y, et al. The single cell immunogenomic landscape after neoadjuvant immunotherapy combined chemotherapy in esophageal squamous cell carcinoma. Cancer Lett. 2024; 593:216951.

[17]

Butler A, Hoffman P, Smibert P, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018; 36: 411-420.

[18]

McGinnis CS, Murrow L, and Gartner Z. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 2019; 8: 329-337.e4.

[19]

Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 2019; 16: 1289-1296.

[20]

Yang Y, Li Y, Yu H, et al. Comprehensive landscape of resistance mechanisms for neoadjuvant therapy in esophageal squamous cell carcinoma by single-cell transcriptomics. Signal Transduct Target Ther. 2023; 8: 298.

[21]

Qiu X, Mao Q, Tang Y, et al. Reversed graph embedding resolves complex single-cell trajectories. Nat Methods. 2017; 14: 979-982.

[22]

Mandard AM, Dalibard F, Mandard JC, et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer. 1994; 73: 2680-2686.

[23]

Wu XX, Zhang HD, Sui ZL, et al. CXCR4 promotes the growth and metastasis of esophageal squamous cell carcinoma as a critical downstream mediator of HIF-1α. Cancer Sci. 2022; 113: 926-939.

[24]

Long H, Jia Q, Wang L, et al. Tumor-induced erythroid precursor-differentiated myeloid cells mediate immunosuppression and curtail anti-PD-1/PD-L1 treatment efficacy. Cancer Cell. 2022; 40: 674-693.e7.

[25]

Li Y, Azmi AS, Mohammad RM. Deregulated transcription factors and poor clinical outcomes in cancer patients. Semin Cancer Biol. 2022; 86: 122-134.

[26]

Yan R, Lin B, Jin W, et al. NRF2, a Superstar of Ferroptosis. Antioxidants (Basel). 2023: 12: 1739.

[27]

Yang W, Xing X, Yeung SC, et al. Neoadjuvant programmed cell death 1 blockade combined with chemotherapy for resectable esophageal squamous cell carcinoma. J Immunother Cancer. 2022; 10:e003497.

[28]

Yan X, Duan H, Ni Y, et al. Tislelizumab combined with chemotherapy as neoadjuvant therapy for surgically resectable esophageal cancer: a prospective, single-arm, phase II study (TD-NICE). Int J Surg. 2022; 103:106680.

[29]

Wu Z, Wu C, Zhao J, et al. Camrelizumab, chemotherapy and apatinib in the neoadjuvant treatment of resectable oesophageal squamous cell carcinoma: a single-arm phase 2 trial. EClinicalMedicine. 2024; 71:102579.

[30]

Liu B, Zhang Y, Wang D, et al. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13(+) T cells to immune-checkpoint blockade. Nat Cancer. 2022; 3: 1123-1136.

[31]

Yue P, Bie F, Zhu J, et al. Minimal residual disease profiling predicts pathological complete response in esophageal squamous cell carcinoma. Mol Cancer. 2024; 23: 96.

[32]

Nakamura S, Ohuchida K, Ohtsubo Y, et al. Single-cell transcriptome analysis reveals functional changes in tumour-infiltrating B lymphocytes after chemotherapy in oesophageal squamous cell carcinoma. Clin Transl Med. 2023; 13:e1181.

[33]

Zhang H, Wen H, Zhu Q, et al. Genomic profiling and associated B cell lineages delineate the efficacy of neoadjuvant anti-PD-1-based therapy in oesophageal squamous cell carcinoma. EBioMedicine. 2024; 100:104971.

[34]

Rudy GB, Lew AM. The nonpolymorphic MHC class II isotype, HLA-DQA2, is expressed on the surface of B lymphoblastoid cells. J Immunol. 1997; 158: 2116-2125.

[35]

Kropshofer H, Hämmerling GJ, Vogt AB. The impact of the non-classical MHC proteins HLA-DM and HLA-DO on loading of MHC class II molecules. Immunol Rev. 2000; 172: 267-278.

[36]

Coulie PG, Van den Eynde BJ, van der Bruggen P, et al. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014; 14: 135-146.

[37]

Paijens ST, Vledder A, de Bruyn M, et al. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021; 18(4): 842-859.

[38]

Ghosh D, Jiang W, Mukhopadhyay D, et al. New insights into B cells as antigen presenting cells. Curr Opin Immunol. 2021; 70: 129-137.

[39]

Silina K. B cell-rich niches support stem-like CD8(+) T cells in cancer microenvironment. Cancer Cell. 2023; 41: 824-825.

[40]

Wang X, Waschke BC, Woolaver RA, et al. HDAC inhibitors overcome immunotherapy resistance in B-cell lymphoma. Protein Cell. 2020; 11: 472-482.

[41]

Liu Z, Zhang Y, Ma N, et al. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 2023; 41: 1852-1870.

[42]

Li JP, Wu CY, Chen MY, et al. PD-1CXCR5CD4 Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer. 2021; 9:e002101.

[43]

Yang M, Lu J, Zhang G, et al. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J Immunother Cancer. 2021; 9:e001136.

[44]

Ukita M, Hamanishi J, Yoshitomi H, et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight. 2022; 7:e157215.

[45]

Li JP, Wu CY, Chen MY, et al. PD-1(+)CXCR5(-)CD4(+) Th-CXCL13 cell subset drives B cells into tertiary lymphoid structures of nasopharyngeal carcinoma. J Immunother Cancer. 2021; 9:e002101.

[46]

Magen A, Hamon P, Fiaschi N, et al. Intratumoral dendritic cell-CD4(+) T helper cell niches enable CD8(+) T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat Med. 2023; 29: 1389-1399.

[47]

Yang M, Lu J, Zhang G, et al. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J Immunother Cancer. 2021; 9.

[48]

Rimal R, Desai P, Daware R, et al. Cancer-associated fibroblasts: origin, function, imaging, and therapeutic targeting. Adv Drug Deliv Rev. 2022; 189:114504.

[49]

You T, Tang H, Wu W, et al. POSTN secretion by extracellular matrix cancer-associated fibroblasts (eCAFs) correlates with poor ICB response via macrophage chemotaxis activation of Akt signaling pathway in gastric cancer. Aging Dis. 2023; 14: 2177-2192.

[50]

Kundu M, Butti R, Panda VK, et al. Modulation of the tumor microenvironment and mechanism of immunotherapy-based drug resistance in breast cancer. Mol Cancer. 2024; 23: 92.

[51]

Li J, Yang J, Jiang S, et al. Targeted reprogramming of tumor-associated macrophages for overcoming glioblastoma resistance to chemotherapy and immunotherapy. Biomaterials. 2024; 311:122708.

[52]

Wang H, Yao L, Chen J, et al. The dual role of POSTN in maintaining glioblastoma stem cells and the immunosuppressive phenotype of microglia in glioblastoma. J Exp Clin Cancer Res. 2024; 43: 252.

[53]

Lin J, Zhang X, Cheng A, et al. Delivery of peptide-LYTAC via polyporus polysaccharide microneedles for targeted CD47 degradation and enhanced tumor immunotherapy. J Am Chem Soc. 2025; 147: 25004-25016.

[54]

Vadevoo SMP, Kang Y, Rangaswamy G, et al. IL4 receptor targeting enables nab-paclitaxel to enhance reprogramming of M2-type macrophages into M1-like phenotype via ROS-HMGB1-TLR4 axis and inhibition of tumor growth and metastasis. Theranostics. 2024; 14: 2605-2621.

[55]

Kang N, Son S, Min S, et al. Stimuli-responsive ferroptosis for cancer therapy. Chem Soc Rev. 2023; 52.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Discovery published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

8

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/