Abstract Background Pancreatic angiosarcoma is a rare and highly aggressive tumor originating from lymphatic or vascular endothelial cells, with poor prognosis and few effective treatments. In this study, we aimed to characterize the tumor ecosystem of metastatic pancreatic angiosarcoma, along with its potential treatment strategies. Methods Single-cell RNA-sequencing and bioinformatics analysis were performed on samples obtained from one patient, including at total of 16,841 cells from pancreatic angiosarcoma liver metastasis and adjacent normal liver tissue. Results Pancreatic angiosarcoma cells exhibited marked upregulation of nuclear factor kappa-B (NF-κB), hypoxia-inducible factor 1 (HIF-1), and myelocytomatosis oncogene (MYC) proto-oncogene signaling pathways, while presenting limited upregulation of actionable therapeutic targets except for cyclin-dependent kinase 4 (CDK4) and epidermal growth factor receptor. Several immune checkpoint genes, including cytotoxic T-lymphocyte-associated protein 4 (CTLA4), lymphocyte-activation gene 3 (LAG3), programmed cell death protein 1 (PDCD1), and cluster of differentiation 86 (CD86), were upregulated in tumor-infiltrating T cells, natural killer (NK) cells, and myeloid cells. Furthermore, intercellular interaction profiling demonstrated enhanced activity of the programmed death-ligand 1 (PD-L1) and CD86 signaling pathways within the tumor microenvironment. The gene-set scores of T/NK-cell exhaustion, regulatory T cell, and macrophage angiogenesis were significantly higher in tumor tissues compared with adjacent normal tissues. However, the phagocytosis scores of macrophages within the tumor-infiltrating region were significantly lower than those in the adjacent normal tissues. Conclusions Our findings outlined an immunosuppressive and angiogenic tumor ecosystem in pancreatic angiosarcoma liver metastasis, suggesting that pancreatic angiosarcoma may be insensitive to most targeted therapies. Conversely, immunotherapies targeting LAG3, PD-L1, and CD86 (e.g. isatuximab, Opdualag, and abatacept) and anti-angiogenic agents may be therapeutically effective and worthy of subsequent exploration. Keywords: pancreatic angiosarcoma, single-cell RNA sequencing, immunotherapy, PD-L1, CD86 Introduction Angiosarcoma is a rare and highly aggressive tumor originating from lymphatic or vascular endothelial cells [[40]1], comprising <1% of all sarcomas [[41]2]. With an annual incidence rate of two to three per million [[42]3], angiosarcoma occurs at any age (most frequently in adults and the elderly), presenting no significant difference in incidence between males and females [[43]4]. Angiosarcoma consists of a clinically and genetically heterogeneous subgroup of sarcomas that can appear in any location of body, mostly in cutaneous tissues (∼60% of cases), especially head and neck, but also in soft tissues, parenchymatous organs, bone, and retroperitoneal areas [[44]5]. As a rare form of angiosarcoma that occurs in the pancreas, pancreatic angiosarcoma accounts for only 0.1% of all primary pancreatic tumors, with only ∼11 cases reported by investigators to date [[45]6–16]. Angiosarcoma is prone to metastasis and recrudescence, which leads to a very poor prognosis, with an overall survival ranging from 6 to 16 months [[46]17]. The diagnosis of pancreatic angiosarcoma remains a big challenge. Physicians have trouble in quickly distinguishing angiosarcoma from other tumors such as hemangiomas and neuroendocrine tumors due to its non-specific morphology and clinical manifestations [[47]17]. Ultrasound, computed tomography (CT), and magnetic resonance imaging have limitations and ultimately histopathology would be required for the final diagnosis, which shows spindle, polygonal, epithelioid, and undifferentiated round cells, and immunohistochemistry reveals the expression of both vascular and endothelial antigens, including Factor-VIII-related antigen (Factor-VIIIRA), cluster of differentiation 34 (CD34), platelet and endothelial-cell adhesion molecule 1 (PECAM1/CD31), and vascular endothelial growth factor (VEGF) [[48]17]. The rarity of pancreatic angiosarcoma cases and the delay in diagnosis have led to a dilemma in its treatment and prognostic assessment. Currently, there are no standardized guidelines for the management of patients with pancreatic angiosarcoma, and radical surgery and adjuvant radiotherapy are currently the mainstay treatment modalities [[49]6, [50]7, [51]17, [52]18]. Cytotoxic chemotherapy is also put into use in the face of inoperable tumors. However, these treatments are also controversial because of the difficulty in obtaining negative margins in surgery and the high recurrence rate after surgery or the severe side effects of drugs [[53]17, [54]18]. With the advent of gene-sequencing technology in recent years, targeted therapy and immunotherapy have also been studied but all gain only mild effects [[55]18]. In terms of prognosis, current gene sequencing in angiosarcomas has reflected several upregulated cancer-driver genes such as ATM serine (ATM), tumor protein p53 (TP53), B-Raf proto-oncogene (BRAF), patched 1 (PTCH1), and APC regulator of wingless-type mouse mammary tumor virus (MMTV) integration site (WNT) signaling pathway (APC) in some patients [[56]19], but characteristic markers for pancreatic angiosarcoma remain unidentified. Histological grading is also difficult for accurately predicting patient prognosis due to the heterogeneity in histologic behavior among the lesions and the presence of tiny, undetectable satellite lesions. In this study, we analysed the gene-expression landscape of pancreatic angiosarcoma liver metastasis based on single-cell RNA-sequencing (scRNA-seq) data from a rare case of the disease. By comparing tumor tissues with adjacent normal counterparts and analysing the tumor-immune microenvironment, we investigated the mechanisms underlying the limited efficacy of current targeted therapies in pancreatic angiosarcoma, identified drivers of immune evasion and metastasis, and explored potential immunotherapeutic targets. Materials and methods Human-specimen sampling A schematic diagram of the overall tissue-sample-collection and processing workflow is shown in [57]Figure 1A. One patient diagnosed with pancreatic angiosarcoma liver metastasis was included in this study. The patient presented a clear history of constant pain in the left upper abdomen for 7 days, with CT imaging demonstrating multiple tumor lesions involving both the pancreas and the liver. In order to clarify the diagnosis, the patient underwent a fine-needle biopsy within the tumor site. A tumor tissue sample within the liver (T) and its paired adjacent normal liver tissue sample (N) were obtained during the biopsy under the supervision of a pathologist. Pathological findings revealed a patchy distribution of malignant cells in the liver tissue, some of which exhibited enlarged hyperchromatic nuclei with marked cytologic atypia, accompanied by lumen formation of varying sizes. Immunohistochemistry presented erythroblast transformation-specific transcription factor ERG (ERG) (+), CD31 (+), CD34 (+), Kiel 67 antigen (Ki-67) (30%+), chromogranin A (CgA) (−), somatostatin receptor 2 (SSTR2) (−), synaptophysin (Syn) (−), hepatocyte (−), and glypican-3 (−), as shown in [58]Figure 1B, and the final diagnosis was considered to be pancreatic angiosarcoma liver metastasis. Prior to needle biopsy of the tumor, the patient did not receive any form of chemotherapy, radiation, or drug treatment. Adjacent non-malignant liver tissues were procured at a distance of ≥5 cm from the tumor. All clinical samples were obtained from The First Affiliated Hospital, Sun Yat-sen University (Guangzhou, China) and the study has received ethical approval from the Ethics Committee of The First Affiliated Hospital, Sun Yat-sen University (Approval No. [2023]701). Written informed consent was obtained from the patient for both sample collection and data analyses. Figure 1. [59]Figure 1. [60]Open in a new tab Single-cell transcriptomic analysis in pancreatic angiosarcoma liver metastasis and adjacent normal tissues. (A) Workflow of sample collection and data analysis in this study. One patient diagnosed with pancreatic angiosarcoma with liver metastasis was included in this study. Fresh liver metastasis and adjacent normal liver tissues were processed and single-cell sequenced to obtain single-cell transcriptomic data. (B) Immunohistochemistry (IHC) and hematoxylin–eosin staining of angiosarcoma tissue sample. (C) Uniform Manifold Approximation and Projection (UMAP) plot of the 16,841 cells from two samples of the tumor site, colored by Seurat clusters. (D) UMAP plot of the 16,841 cells from two samples of the tumor site, colored by manually annotated cell clusters. Each point indicates a single cell. (E) Feature plot displaying the expression distributions of housekeeping genes, including actin beta (ACTB), beta-2-microglobulin (B2M), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and the immune-cell-specific marker protein tyrosine phosphatase receptor type C (PTPRC). Each dot represents a single cell, with cells exhibiting the highest expression levels visually demarcated. (F) Heat map showing scaled, normalized expression of top five differentially expressed genes in each cell cluster. (G) Feature plot showing the expression distributions of cluster-specific markers used to distinguish individual cell clusters. Each dot represents a single cell, with peak expression intensity clearly reflected in the visual gradient. (H) UMAP showing cell-cluster distribution in tumor sample (T) and adjacent normal liver tissue sample (N), and stacked histogram shows the proportion of six cell clusters in each sample. HE = hematoxylin and eosin staining, NK = natural killer cell, UMAP = Uniform Manifold Approximation and Projection, CK = cytokeratin. Single-cell-suspension preparation Tumor tissues and adjacent normal liver tissues were snap-frozen in liquid nitrogen within 15 min after sampling for subsequent single-cell RNA-sequencing procedures [[61]20]. A total of 2.5 mL of tumor digestive enzyme mix (#130–095-929, Tumor Dissociation Kit; Miltenyi Biotech, Germany) was added before digestion for 10 min in a 37°C water bath. The single-cell suspension of dissociated tissues was then filtered through 70- and 30-μm magnetic-activated cell-sorting strainers (#130–098-462/130–098-458; Miltenyi Biotech) to remove the cell mass. Next, dissociated cells were pelleted (4°C, 400 g, 6 min) and resuspended by using a modest red blood cell (RBC) lysis buffer (#00430054; eBioscience, USA) for 5–10 min. Two or three times volume of phosphate buffer saline (PBS; #C10010500CP; Gibco, USA) was added to terminate the RBC lysis. The single-cell suspension was then pelleted again and washed once with cold PBS, after which the cells were resuspended with RPMI1640 (#01–103-1A; BI, Israel) or cryopreserved in 90% Fetal Bovine Serum (#FND500; ExCell Bio, China) with 10% dimethyl sulfoxide (#D8371; Solarbio, China) for long-term preservation. The whole process of tissue dissociation was carried out on ice [[62]21]. Single-cell RNA-seq library preparation Single cells were processed with the 10x Chromium Controller (10x Genomics, USA) based on the 10x gel bead-in-emulsion code (GEMCode) proprietary technology. An appropriate volume of single-cell suspension with a cell concentration of 700–1,200 cells/µL was loaded into each channel. About 8,000 cells were captured per sample, which were further mixed with barcoded beads. After reverse transcription reaction and complementary DNA (cDNA) amplification, libraries were constructed following the standard 10x Genomics protocol (Single Cell 5′ Reagent Kits v5.2 User Guide) and sequenced on Novaseq™ 6000 (Ilumina, USA) [[63]21]. Single-cell RNA-seq data preprocessing and cell-type annotation The CellRanger software pipeline (Version 7.0.1, 10x Genomics) was used to demultiplex cellular barcodes and map reads to the human genome (build hg38), and quantify the gene expression in single cells, generating a digital gene-expression matrix [unique molecular identifier (UMI) counts per gene per cell] for each sample. Expression matrices for all samples were then processed by using the R Seurat package (version 4.3.0) [[64]22]. As a quality-control (QC) step, we filtered out cells with: (i) the number of detected genes <200 or >6,000; (ii) the proportion of mitochondrial gene expression of >15%; and removed genes detected in fewer than three cells. After that, we obtained a total of 16,841 high-quality single-cell transcriptomes from all samples. To mitigate the impact of data sources on the analysis, we normalized the data by using canonical correlation analysis. Subsequently, the “ScaleData” function was employed to perform linear regression against the normalized expression levels, total UMI counts, and mitochondrial RNA content per cell for each gene. Principal component analysis was then performed by using “RunPCA”. The gene-expression and clustering outcomes were visualized on a Uniform Manifold Approximation and Projection (UMAP) plot by using the top 10 principal components, obtained through “RunUMAP”. To further cluster the cells, we employed the functions “FindNeighbors” and “FindClusters”. Cell types were determined by analysing the gene expression of established markers. T cells and natural killer (NK) cells were identified by using protein tyrosine phosphatase receptor type C (PTPRC), CD3D, CD3E, CD8A, and CD4; myeloid cells were identified by using CD68, S100 calcium binding protein A8 (S100A8), S100A9, complement C1q A chain (C1QA), and B chain (C1QB); B cells were identified by using CD19, CD79A, CD79B, and membrane spanning 4-domains A1 (MS4A1); endothelial cells were identified by using PECAM1, von Willebrand factor (VWF), and selectin E (SELE); fibroblasts were identified by using integrin subunit alpha 8 (ITGA8), periostin (POSTN), and collagen type I alpha chain 2 (COL1A2); and hepatic cells were identified by using albumin (ALB), apolipoprotein A2 (APOA2), epithelial cell adhesion molecule (EPCAM), and Keratin 19 (KRT19). Gene-set signature scores In order to probe the status and distribution of immune cells within the tumor-immune microenvironment, we assessed the expression levels of the several gene sets for naive, cell killing, exhausted, co-stimulate, and antigen presentation of immune cells in tumor tissues and adjacent normal liver tissues [[65]23]. In addition, tip-like gene-set scores were evaluated among endothelial cells [[66]24]. The R package AUCell (version 1.24.0) was utilized to assess the average expression of our target gene set at the single-cell level [[67]25]. Copy-number variation analysis Endothelial-cell copy-number variation (CNV) evaluation was performed by using the infercnv R package (version 1.18.1) [[68]21]. Amplifications and deletions at the single-cell level were predicted base on the scRNA-seq data. The gene-expression matrix of tumor cells was extracted from the Seurat object to calculate its CNVs and fibroblasts, T/NK cells, and myeloid cells were selected as normal references. The inferCNV analysis was conducted with parameters