Integrating multi-modal transcriptomics identifies cellular subtypes with distinct roles in PDAC progression.

Cell Oncol (Dordr)

Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.

Published: August 2025


Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Purpose: Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest malignancies, largely due to its highly immunosuppressive and fibrotic tumor microenvironment (TME). However, the spatial and functional organization of its cellular components remains poorly understood.

Methods: We present an integrated transcriptomic atlas of the PDAC TME by combining single-cell RNA sequencing (n = 88; 187,520 cells), Visium spatial transcriptomics (n = 20; 67,933 spots), bulk RNA sequencing (n = 1,383), and high-resolution Xenium spatial transcriptomics (n = 2; 307,679 cells). Key findings were validated using scRNA-seq, bulk datasets, multiplex immunohistochemistry, and spatial imaging.

Results: POSTN⁺ fibroblasts and SPP1⁺ macrophages consistently co-infiltrated across 12 independent bulk RNA-seq cohorts, and showed spatial correlation in both Visium and Xenium platforms. Those tumor-promoting cell states were enriched in hypoxic, angiogenesis, and epithelial-mesenchymal transition, and were linked to poor prognosis. In contrast, CCL4⁺ CD8⁺ effector T cells and IGHG1⁺ plasma cells co-occurred within immune-active niches, were enriched for cytotoxic and activation-related pathways, and were associated with improved patient survival. Notably, these protective immune subsets remained detectable despite the immunosuppressive nature of the PDAC TME. Expression specificity of POSTN, SPP1, CCL4, and IGHG1 was validated at the transcriptomic and protein levels.

Conclusions: We delineate two opposing cellular programs in the PDAC TME-tumor-promoting stromal remodeling and anti-tumor immune activation-spatially organized in distinct niches. Those findings suggest that targeting POSTN⁺ fibroblasts and SPP1⁺ macrophages-mediated stromal interactions while promoting CCL4⁺ T cell and IGHG1⁺ plasma cell immunity, may offer new therapeutic strategies for PDAC.

Download full-text PDF

Source
http://dx.doi.org/10.1007/s13402-025-01100-6DOI Listing

Publication Analysis

Top Keywords

pdac tme
8
rna sequencing
8
spatial transcriptomics
8
postn⁺ fibroblasts
8
fibroblasts spp1⁺
8
ighg1⁺ plasma
8
pdac
6
spatial
5
integrating multi-modal
4
multi-modal transcriptomics
4

Similar Publications

Sequential YAP-1/FOSL-1 silencing and epigenetic therapy to overcome stromal barriers in pancreatic cancer.

Int J Pharm

September 2025

CINBIO, Immunology Group, Universidade de Vigo 36310 Vigo, Spain; Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain. Electronic address:

Pancreatic ductal adenocarcinoma (PDAC) remains a highly aggressive malignancy with poor therapeutic outcomes due to its desmoplastic tumor microenvironment (TME), hindering drug and activated immune cell penetration. Cancer-associated fibroblasts (CAFs) are central in supporting tumor growth and forming a protective stroma. We propose a novel dual-therapy targeting the Hippo pathway and histone deacetylation, both involved in tumor progression, resistance, and stromal interactions, to overcome PDAC therapeutic resistance.

View Article and Find Full Text PDF

Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is highly aggressive with limited curative options, primarily surgical resection. However, only about 20% of the tumors are resectable at diagnosis. Immunotherapies have largely failed in PDAC due to its immunosuppressive tumor microenvironment (TME).

View Article and Find Full Text PDF

Pancreatic ductal adenocarcinoma (PDAC) remains a devastating malignancy characterized by profound lethality, aggressive local invasion, dismal prognosis, and significant resistance to existing therapies. Two critical biological features underpin the challenges in treating PDAC: extensive perineural invasion (PNI), the process by which cancer cells infiltrate and migrate along nerves, and a profoundly immunosuppressive, or "cold," tumor microenvironment (TME). PNI is not only a primary route for local tumor dissemination and recurrence but also a major contributor to the severe pain often experienced by patients.

View Article and Find Full Text PDF

The absence of Peroxiredoxin-1 in human pancreatic ductal adenocarcinoma (PDAC) markedly reduces cell survival and tumor growth when coupled with the inhibition of Ref-1 redox signaling.

Redox Biol

September 2025

Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University School of Medicine, Departme

Pancreatic ductal adenocarcinoma (PDAC) remains highly resistant to therapy, surviving despite hypoxia, oxidative stress, and nutrient deprivation. Redox effector factor-1 (Ref-1) regulates several oncogenic transcription factors (TFs) and is controlled by peroxiredoxins (PRDX). We investigated how Ref-1 inhibition by APX2014, combined with PRDX expression, affects pancreatic cancer cells from multiple patient lines.

View Article and Find Full Text PDF

is among the most frequently mutated oncogenes in cancer, and for decades, efforts at pharmacological blockade of its function in solid cancers have been unsuccessful. A notable advance in this endeavor is the recent development of small-molecule KRAS inhibitors, which enable direct targeting of the mutant oncoprotein. Here, we comprehensively evaluated the preclinical efficacy of BI-2493, a first-in-class allele-agnostic mutant-KRAS inhibitor (panKRASi), in pancreatic ductal adenocarcinoma (PDAC).

View Article and Find Full Text PDF