Introduction
Keratinocyte carcinomas including cSCC and BCC are the most common NMSC, and their incidences continue to increase [1, 2]. Although both cSCC and BCC originate from keratinocytes and primarily result from mutations induced by UV exposure [3], they exhibit distinct characteristics. While BCC is associated with uncontrolled activation of the hedgehog signaling pathway [4], cSCC is characterized by cumulative gene mutations affecting suppressor genes like CDKN2A and NOTCH, as well as oncogenes like RAS. Additionally, several signaling pathways, such as NF-kB, MAPK, and PI3K/AKT/mTOR, play roles in the increased expression of the epidermal growth factor receptor (EGFR) [5, 6]. Surgical excision is commonly advised for both BCC and cSCC, resulting in high rates of successful treatment. In contrast to BCC, metastasis occurs in 1.2%–5% of patients with cSCC, leading to a 3-year mortality rate of 46% for metastatic disease [4, 7]. Given the complexities of excision in these cases, alternative therapies are imperative. EGFR inhibitors and immunotherapy such as anti-PD-1 therapy are now established as standard care in advanced or metastatic cSCC [8]. Nevertheless, cSCC's pronounced heterogeneity, stemming from cumulative mutations, complicates patient stratification. Hence, identifying biomarkers for cSCC treatment requires conducting gene expression profiling analysis in each individual cSCC. Previous traditional bulk transcriptome sequencing (bulk RNA-seq) has offered valuable insights into the genetic basis of BCC and cSCC [9, 10]. However, these approaches, involving a mixture of various cells, often conceal significant aspects of intra-tumor heterogeneity, thereby limiting our insight into cSCC biology. The development of single-cell RNA sequencing (scRNA-seq) enables the elucidation of complex tumor diversity and dissects the dynamic communication network between the tumor and its microenvironment [11]. Utilizing scRNA, several groups have documented the gene expression patterns in human BCC and cSCC. Chistian defined BCC cellular heterogeneity and identified the heat shock protein (HSP) pathway as a potential therapeutic option for BCC [12]. Andrew and Li discovered a tumor-specific keratinocyte (TSK) population unique to cSCC, serving as a hub for intercellular communication and exhibiting remarkable epithelial–mesenchymal transition (EMT) features [13, 14]. Despite this, deciphering both tumor heterogeneity and its interactions with surrounding cells through high-resolution methods remains imperative. Here, we first characterize the differences between BCC and cSCC at the single-cell transcriptomic level combined with spatial transcriptome and identify potential invasive biomarkers and immunosuppressive mechanisms in SCC.
Materials and Methods
Human Samples
Human cancer tissues (BCC, n = 5; SCCIS, n = 3; SCC, n = 3) were collected from skin cancer patients at the Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Hospital (Institutional Review Board No. 30–2017-11), using wide excision surgery. The wide excision was designed to include both the cancerous and adjacent skin tissues with a safety margin of 5 mm to 30 mm, varying by cancer type, and was completely excised. These freshly excised biopsies were split into two sections: one half of each sample was promptly dissociated into a single-cell suspension for scRNA-seq, and the other half was preserved in formalin for immunohistochemical analysis and Spatial Transcriptomics analysis. All diagnoses of cancer types were verified histologically by a board-certified dermatopathologist.
Single-Cell
Single-cell cDNA and library construction were performed following the 10 × Genomics guidelines (10 × Genomics 5′ v2, CG000331, Rev. D). Cells were captured using a Chromium controller (10 × Genomics) at the Seoul National University College of Medicine, Republic of Korea. Gel beads containing 10 × barcodes, UMI, and oligo dTs were used to form GEMs (Gel bead-in EMulsions) with the cells. After generating GEMs, the captured mRNA underwent reverse transcription via PCR to produce cDNAs. During cDNA synthesis, each cDNA was tagged at the 5− end with a UMI and a barcode indicating its cell of origin. The full-length cDNA was then amplified via PCR to generate sufficient mass for library construction and quantified using the ScreenTape on a 4150 TapeStation instrument (Agilent). Then, the libraries were sequenced on a NovaSeq6000 (Illumina).
Preprocessing of
The FASTQ files generated from Illumina sequencing output were aligned to the human reference transcriptome (GRCh38) using 10× Genomics Cell Ranger v7.0.1. Raw expression data were loaded into Seurat (v4.9.9) R (4.2.1) for downstream analysis of our scRNA-seq [15]. Background transcript contamination in retained true cells was eliminated using SoupX with the contamination fraction set to 20% (setContaminationFraction = 0.2) [16]. Doublets or multiplets were removed using scDblFinder with default parameters [17], and cells within each dataset were filtered to include only those with more than 300 genes and less than 10% of mitochondrial genes, resulting in a total of 117,663 cells: SCCIS (ID: SCCIS1, SCCIS2, SCCIS3; n = 19,085 cells, 4310 cells, and 13,269 cells, respectively), SCC (ID: SCC1, SCC2; n = 19,277 cells and 13,987 cells), and BCC (ID: BCC1-5; total: 68,782 cells). To eliminate technical variations in samples derived from different experiments, 10 samples were merged and integrated to remove the batch effect by scVI [18]. Normalized expression, which was normalized using default parameters, was applied to detect highly variable genes (HVGs) using the Seurat algorithm. Each library was used as the batch key for calculating HVGs and the batch variable in the scVI modeling. Unsupervised clustering was conducted using the Louvain algorithm followed by visualization by Uniform Manifold Approximation and Projection (UMAP). The cluster markers were found using the FindAllMarkers function, and the cell types were manually annotated based on the markers. Gene signature was performed using the “GSVA” package with default parameters. The data visualization and statistical analysis were performed in the licensed version of GraphPad Prism v10 and R (4.2.1) along with the following packages: ggplot2, ggpubr (v0.6.0). All the statistical tests were two-tailed, and a p-value of 0.05 was considered significant.
Spatial transcriptomics was performed using the GeoMx DSP platform of the whole-transcriptome atlas (NanoString Technologies, Seattle, WA, USA). Tissue sections from formalin-fixed paraffin-embedded (FFPE) blocks were mounted on a charged Leica Bond slide. Tissue slides were deparaffinized and subjected to antigen retrieval procedures and RNA hybridization according to the manufacturer's user manuals. These slides were stained with morphology markers containing SYTO13 (NanoString), PanCK (Novus, AE1 + AE3), CD68 (Santa Cruz, KP1), and CD3 (Novus, C3e/1308). The stained slides were loaded into the GeoMx DSP instrument. Next, selected regions of interest (ROIs) were chosen within the combined stroma/tumor areas to represent distinct sections of the tumor. These ROIs were then segmented by applying PanCK staining, which separated the tumor and stroma areas of illumination (AOI). Oligonucleotides from AOI were released upon exposure to ultraviolet light and collected. These oligonucleotides were amplified and finally pooled. The generated libraries were paired-end sequenced on Illumina's NextSeq 6000.
Preprocessing of
The FASTQ files processed from the library were converted to Digital Count Conversion (DCC) files by NanoString's GeoMx NGS pipeline software (V.2.1). DCC files were imported back into the GeoMx DSP platform to generate an expression count matrix. After QC according to NanoString's recommendations, the GeoMx dataset was loaded into the StandR package (v1.9.0) for downstream analysis [19]. The segments were maintained with at least 100 “NucleiCount,” resulting in 16 segments, and 18,677 target genes were used for analysis. UMAP visualizations were generated after TMM normalization was applied to each ROI, and integration was performed to remove batch effects using RUV4 with default parameters in StandR. The data visualization and statistical analysis were performed in the licensed version of GraphPad Prism v10 and R (4.2.1) along with the ggplot2 package.
More materials and methods can be found online as Appendix S1.
Results
The Heterogeneity of Cellular Composition Between cSCC and BCC
We dissociated surgical biopsy samples taken from a cohort of patients diagnosed with BCC (n = 5) and cSCC (SCCIS, n = 3; SCC, n = 2) to acquire single-cell suspensions and subjected them to the 10× Genomics Chromium platform. After the removal of doublets, ambient RNA, and dead cells, a total of 117,663 single cells were obtained (Figure 1A). Based on their cell-specific markers, we identified 11 cell types, including Keratinocyte, Sweat gland, Fibroblast, T cell, Myeloid cell, B cell, Plasmablast, Mast cell, Melanocyte, Endothelial cell, and Lymphatic endothelial cell, respectively (Figure 1B) [12, 13]. To integrate the BCC and cSCC datasets, we used scVI [18]. In contrast to non-epithelial cell types, keratinocytes exhibited distinct clustering patterns across tumor types and donors (Figure 1C,D). Comparative analysis of the proportion of each cell type revealed no significant differences between BCC and SCC (Data not shown).
[IMAGE OMITTED. SEE PDF]
Defining and Characterizing Malignant Epithelial Cells
To define malignant cells from normal cells, we performed single-cell CNV analysis by using “CopyKAT” package [20]. The result indicated a distinction between malignant and normal cells among the 117,663 cells and inferred that malignant cells belong to the keratinocyte (KC) population. Malignant KC (aneuploid, n = 43,866 cells), referred to as carcinoma, and normal KC (diploid, n = 10,382 cells) were successfully identified (Figure 2A). To define which skin cancer types the Carcinoma clusters originated from, we labeled carcinoma clusters according to cancer type as BCC (n = 23,842), SCCIS (n = 2659), and SCC (n = 15,365), along with normal keratinocytes (NKC) (Figures 2B and S1). Since BCC and cSCC originate from NKC, we utilized the “Monocle2” package to delineate their differentiation pathway. NKC were positioned at the initial point of the trajectory, subsequently branching into two distinct pathways referred to as cell fate 1 and cell fate 2. Remarkably, SCCIS and SCC resided at the final segment of trajectory 1, whereas BCC populated the endpoint of trajectory 2 (Figure 2C). We observed the accumulation of molecules related to BCC progression such as SPON2, and components of the hedgehog pathway, including PDGFA, PTCH1, and PTCH2, along trajectory 2. Conversely, CDKN2A, a molecule possibly associated with the development of cSCC, coincided with trajectory 1 [6, 9]. Additionally, several oncogenes (MYC, TUBA1C, and TPM3), cyclin-dependent genes (CCNB1, CDC20, and CDKN3), and genes associated with proliferation (MKI67 and TOP2A) were enriched in cSCC progression (Figure 2D). Moreover, to characterize genes during the evolution of these cancer types, we acquired the upregulated genes in BCC, SCCIS, and SCC compared to NKC using differentially expressed genes (DEGs) analysis (Figure 2E). As a result, we identified 36 SCC-specific upregulated genes, of which 16 were highlighted. Consistent with previous findings, SCC exhibited increased CDKN2A compared to NKC [6, 9], and, as known, TNC and LAMB3 associated with the PI3K/AKT/mTOR pathway were upregulated [21, 22]. Furthermore, six tumor progression genes (GNAI2, ODC1, APP, TNFRSF12A, BASP1, and LARP6) and nine migration and metastasis-related genes (MSN, FLOT1, LAMB3, MYL9, ITGB1, FTH1, TPM4, and PMEPA1), which could explain the invasive characteristics of SCC, were identified. These genes represent potential key drivers of SCC development from NKC and serve as potential targets for the treatment of SCC.
[IMAGE OMITTED. SEE PDF]
Characteristics of
To address which clones are associated with enhanced aggressive traits of SCC, we subclustered 43,886 malignant epithelial-derived cells and identified 15 carcinoma cell clusters (Figure 3A). The proportion of Carcinoma 3 significantly increased within the carcinoma in cSCC compared to BCC (Figure 3B) with specific expression of ITGA5 and COL6A1 in SCC (Figure 3C,D). ITGA5 has been previously reported as a marker of tumor-specific keratinocytes (TSK) in SCC and associated with recurrence and migration [13, 14]. COL6A is a major component of the extracellular matrix (ECM), mainly found in the basement membrane region. Recently, COL6A1 has been recognized as important for tumor growth and metastasis, and it has been reported to be expressed in a variety of cancers including cervical squamous cell carcinoma and pancreatic carcinoma [23, 24]. We next explored DEGs between Carcinoma 3 and other carcinoma clusters using volcano plot analysis. Carcinoma 3 exhibited increased expression of S100-protein related genes (S100A2, S100A9, S100A11, and S100A13), interferon related genes (STAT1, IFITM1, IFITM3, and IFI44L), and metastasis related genes (TPM3, TPM4, MMP13, PLAU, and PLAUR) (Figure 3D). STAT1 promotes IFNAR and JAK pathway which potentially drives the invasion of cSCC [25]. S100A9 activates the NFkB signaling pathway, while S100A11 activates the mTOR pathway, inducing progression and invasion in cSCC [26, 27]. These findings indicate that Carcinoma 3 might have local invasive and metastatic potential. To evaluate the migration potential of Carcinoma 3, we performed EMT gene signature scoring using the Hallmark EMT gene set from MSigDB and observed that Carcinoma 3 exhibited the highest EMT activity. To further investigate this finding, we compared EMT scores between SCC and other cancer types, confirming that SCC tumors showed significantly elevated EMT gene scores (Figure 3E). Moreover, we identified disease-specific upregulated genes in SCC, particularly in Carcinoma 3, and discovered immune cell migration and proliferation-associated genes, such as IL15, IL32, CXCL16, and TNFSF9 (Figure 3F) [28, 29]. These results indicate that the specific cluster identified in SCC drives the invasive characteristics of SCC and the recruitment of specific immune cells.
[IMAGE OMITTED. SEE PDF]
Identifying Tumor-Associated
To define tumor-associated lymphocytes interacting with Carcinoma 3, we subclustered T lymphocytes based on the expression of CD3D and TRAC, yielding a total of 21,093 cells (nBCC = 6191, nSCCIS = 7981, and nSCC = 6921) (Figure 4A). Based on well-established markers, we identified seven T cell subpopulations: (1) Tregs (FOXP3+, CTLA4+, and TIGIT+), (2) CXCL13_CD4T (CD4+ and CXCL13+), (3) CD8_Tex (CD8A+, TIGIT+, and PDCD1+), (4) CD4T (CD4+ and IL7R+), (5) CD8T (CD8+ and GZMK+), (6) NK_gdT (TRDC+ and XCL2+), and (7) Prolif_Tcell (MKI67+ and TOP2A+) (Figure 4B) [30]. We next identified cells expressing the receptors CXCR6 and TNFRSF9, which correspond to their ligands CXCL16 and TNFSF9 in Carcinoma 3, particularly in SCC (Figure 4C). CXCR6 serves as a central indicator for resident memory T (Trm) cells, contributing to immune surveillance by interacting with epithelial cells [31]. TNFRSF9 (commonly referred to as 4-1BB or CD137) fosters T cell proliferation and enhances the immune-suppressive functions of Tregs within tumors [32]. Both CXCR6 and TNFRSF9 are predominantly expressed in Tregs, suggesting the potential interaction of specific Tregs and Carcinoma 3. Within our dataset, no significant differences were observed in the proportion of Tregs across different cancers or clone sizes. However, the prevalence of CXCR6 expression among Tregs was highest in SCC samples characterized by larger clone sizes (Figure 4D). Next, we examined the DEGs in Tregs based on CXCR6 expression. In CXCR6+ Tregs, we observed elevated expression of genes related to immune suppression such as FOXP3, IL2RA, IL2RB, LGALS1, and LGALS3, along with increased expression of costimulatory molecules like TIGIT, CTLA4, ICOS, TNFRSF4, TNFRSF9, and TNFRSF18 (Figure 4E) [33]. To further understand their functional state, we performed gene set enrichment analysis (GSEA) comparing CXCR6+ versus CXCR6− Tregs. Notably, CXCR6+ Tregs were significantly enriched for gene sets associated with TCR-activated CD4+ T cells (GSE13738) and activated or induced Treg signatures (e.g., GSE14415, GSE24634, and GSE7852) (Figure 4F). Taken together, these results imply that CXCR6+ clonally expanded Tregs in SCC may possess migratory properties and a transcriptional profile indicative of immunoregulatory activity, potentially contributing to the immunosuppressive tumor microenvironment.
[IMAGE OMITTED. SEE PDF]
Potential Receptor–Ligand Interaction Between Carcinoma 3 and Tregs
To identify communication between Carcinoma 3 and Tregs, we performed receptor–ligand interaction analysis among Carcinoma 3 and T cell populations using NicheNet. Consistent with a Carcinoma 3–T cell niche in SCC, robust signaling to Tregs was facilitated through multiple ligand–receptor interactions, encompassing TNFSF9-TNFRSF9 and CXCL16-CXCR6, as well as PTN-SDC4, PVR-CD96, and PVR-TIGIT (Figure 5A). Carcinoma 3 specifically expressed PTN and PVR in SCC compared to BCC and SCCIS (Figure 5B). PTN is a heparin-binding growth factor and has been reported to play an important role in cell–cell adhesion, cell motility, cell division, immune cell migration, and angiogenesis in various cancers [34, 35]. PVR predominantly interacts with TIGIT and is likely to foster an immunosuppressive phenotype in cells expressing TIGIT [36]. PVR is known for its ability to attach to CD96, which triggers an immunosuppressive reaction in mouse models. However, a direct inhibitory effect through CD96 has yet to be established in human research [37]. These receptors, identified in Tregs, were also significantly upregulated in SCC (Figure 5C). Conversely, ligand–receptor pairs including LGALS3-ANXA2, NAMPT-ITGA5, and GRN-TNFRSF1A were identified in Tregs (Figure 5D). LGALS3 promoted tumor progression, invasiveness, and immune suppression, and blocking LGALS3-ANXA2 has been documented to initiate cell apoptosis by suppressing all downstream EGFR signaling pathways [38]. NAMPT/Visfatin can modulate various downstream signaling pathways, including AKT/PI3K and ERK/MAPK, potentially increasing cell survival and proliferation in cSCC [39]. GRN overactivity occurs in many types of cancers and imparts an aggressive phenotype on poorly tumorigenic carcinomas [40]. Overall, our findings suggest that the SCC-specific carcinoma cluster may engage in transcriptionally inferred interactions with Tregs, potentially contributing to an immunosuppressive microenvironment that supports tumor progression.
[IMAGE OMITTED. SEE PDF]
Spatial Transcriptomic Analysis Supports Enrichment of Carcinoma 3 at the Tumor–Immune Boundary
To examine the localization of Carcinoma 3 in SCC specimens from a spatial perspective, we performed spatial transcriptome analysis on SCC samples (SCC1 and SCC3) using the NanoString GeoMx Digital Spatial Profiler (DSP) platform (Figure 1A). Based on histological features and immunostaining for SYTO13, PanCK, CD3D, and CD68, we selected eight regions of interest (ROIs) and defined segments representing tumor cells (SCC_Tumor, PanCK+, n = 8) and the surrounding TME (SCC_TME, PanCK-, n = 8) (Figure 6A,B). These ROIs were selected based on the presence of PanCK+ tumor regions directly adjacent to dense CD3D+ and CD68+ immune infiltrates, identified through both H&E and multiplex immunofluorescence staining, to capture tumor margins involved in immune interaction. To compare SCC-specific transcriptomic characteristics with a nonmalignant inflammatory skin condition, we utilized internal GeoMx data from pemphigus vulgaris (PV) [41], which is a keratinocyte-derived autoimmune disease characterized by strong immune cell infiltration and intraepidermal blistering. We included PV regions of interest (ROIs) categorized as either PanCK+ (PV_epi, n = 10) or PanCK− (PV_immune, n = 9), analogous in compartment identity to the SCC_Tumor and SCC_TME groups. All 37 AOIs (SCC and PV) passed quality control and were included in downstream analyses. UMAP analysis demonstrated a clear separation between SCC and PV samples, indicating distinct transcriptomic profiles between malignant and nonmalignant epithelial inflammation (Figure 6C). We compared the expression of Carcinoma 3-associated genes between SCC_Tumor and PV epithelial regions (PV_epi) (Figure 6D). Genes such as COL6A1, ITGA5, FTH1, TNC, LAMB3, S100A2, S100A11, STAT1, IFITM1, TPM3, TPM4, and IL32 were significantly upregulated in SCC_Tumor compared to PV_epi. To further investigate how Carcinoma 3 may participate in spatially coordinated tumor–immune interactions, we evaluated ligand–receptor gene pairs between SCC_Tumor and SCC_TME regions. For each ligand–receptor pair preselected from the NicheNet analysis (Figure 5A), we calculated the paired Spearman correlation coefficient across spatially matched SCC_Tumor and SCC_TME segments (n = 8 AOI pairs) (Figure 6E). Several ligand–receptor pairs exhibited relatively strong correlations, suggesting spatially organized interactions between tumor and immune compartments. Among immune-related interactions, TNFSF9–TNFRSF9 displayed a strong spatial correlation. Moreover, we compared multiple ligand–receptor pairs highlighted within the Carcinoma 3–Treg niche in SCC (Figure 6F). To validate the spatial association between Carcinoma 3 cells and CXCR6+ Tregs at the protein level, we performed multiplex immunofluorescence staining on serial sections of SCC (Figure S2). ITGA5+ PanCK+ tumor cells, corresponding to the Carcinoma 3 cluster, were localized at the tumor–stroma boundary, and CXCR6+ FOXP3+ CD3D+ regulatory T cells were observed in close proximity within the adjacent stroma. These findings provide direct spatial evidence supporting the Carcinoma 3–Treg niche and reinforce the transcriptionally inferred ligand–receptor interactions, suggesting that Carcinoma 3 cells at the tumor edge may actively shape the local immune landscape.
[IMAGE OMITTED. SEE PDF]
Discussion
The inter- and intra-tumor heterogeneity of NMSC hampers the general interpretation of tumor progression and the shaping of the tumor microenvironment. However, current research on NMSC has primarily focused on single cancer types, resulting in a lack of comprehensive explanations regarding general observation and tumor progression. Therefore, we conducted the comparative analysis of BCC and cSCC using scRNA-seq, which elucidated general principles and fostered a deeper, more specific understanding of each cancer type.
To understand how individual tumor cells shape the tumor microenvironment, we need to separate the tumor clones from the normal tissue cells from which they originated. However, distinguishing keratinocyte-derived carcinoma from human skin was challenging due to the expression of some oncogenes in normal cells as well. To address this issue, we estimated copy number variation driving clonal evolution of tumor cells and distinguished cancer cells from normal cell types using the “CopyKAT” package. We successfully annotated cancer cells, enabling us to conduct trajectory analysis from normal to tumor cells. Our results confirmed that BCC and cSCC undergo different evolution processes. In BCC, we were able to confirm a BCC-specific gene expression pattern, including BCAM and EPCAM, as identified in previous single-cell studies [12]. Moreover, while mutations in the PTCH gene are known to occur in familial BCC, our results also demonstrated an increase in PTCH-related genes in sporadic BCC. Conversely, in cSCC, we observed an increase in predominantly proliferative characteristics and confirmed that SCCIS and SCC share a common lineage. To understand which gene expressions influence the malignancy of SCC during its progression from SCCIS to SCC, we conducted a comparative analysis of cancer clones from SCC and SCCIS. Through this analysis, we defined 16 SCC-specific genes, including FTH1, CDKN2A, BASP1, APP, ITGB1, GNAI2, MSN, LARP6, FLOT1, TPM4, PMEPA1, TNFRSF12A, LAMB3, TNC, ODC1, and MYL9, which are related to tumor progression and migration. This finding aligns with the known invasive characteristics of SCC, suggesting that these genes could serve as potential biomarkers or therapeutic targets for SCC.
Next, we performed subclustering of tumor cells to understand intra-tumor heterogeneity. Interestingly, the Carcinoma 3 cluster with high expression of COL6A1 and ITGA5 was predominantly observed in SCC. COL6A1 encodes the α1 chain of type VI collagen, a fundamental ECM protein essential for maintaining the structural integrity of various tissues. Recent studies indicate that COL6A1 is involved in several biological functions including cell migration, differentiation, wound healing, and preserving cell stemness [42]. Furthermore, increased expression of COL6A1 has been linked to enhanced motility and metastasis in lung, pancreatic, and cervical cancer cells, whereas reducing COL6A1 expression has been associated with decreased metastatic capabilities [23, 24]. ITGA5 primarily serves as a receptor for fibronectin, partnering with integrin beta 1 to form a heterodimer. Research has established that ITGA5 is crucial for cancer cell proliferation, migration, invasion, and metastasis [43, 44]. Collectively, we propose that COL6A1 and ITGA5 expression in SCC enhances adhesion to the ECM, thereby promoting the malignancy of the cancer. Notably, the COL6A1+/ITGA5+ malignant epithelium cluster (Carcinoma 3) exhibited increased expression of IL15, IL32, CXCL16, and TNFSF9. IL15, IL32, and TNFSF9 have been reported to promote the survival and function of T cells and NK cells. CXCL16 induces immune cell homing and accumulation. These findings suggest that Carcinoma 3 in SCC leads to increased immune cell infiltration, thereby characterizing SCC as a hot tumor.
To determine which immune cells interact with Carcinoma 3, we performed subclustering of immune cells. In SCC, we observed increased expression of CXCR6, the receptor for Carcinoma 3's CXCL16, specifically in Tregs. This suggests a new potential migration mechanism of Tregs to the tumor site in SCC. Interestingly, among Tregs, those expressing CXCR6 showed increased levels of functional markers such as FOXP3, IL2RA, IL2RB, and CD7, as well as immune checkpoint proteins, including CTLA4, TIGIT, and ICOS, and Tregs activity-associated genes such as TNFRSF4, TNFRSF9, and TNFRSF18 [33]. These findings indicate that although Carcinoma 3 in SCC recruits diverse immune cells, since it also drives the migration of functional Tregs via the CXCL16/CXCR6 axis, thereby creating a tumor microenvironment favorable for their survival. To further characterize the cellular communication between Carcinoma 3 and Tregs in SCC, we utilized the “NicheNet” package and focused SCC-specific receptor–ligand pairs. We identified three ligands within Carcinoma 3 in SCC involving TNFSF9, PTN, and PVR, which promote the activation of Tregs [34–36]. This suggests that Carcinoma 3 in SCC not only promotes the migration of Tregs to tumor site but also enhances their immunosuppressive capabilities. Additionally, we focused the ligands of Tregs in the SCC niche. We confirmed increased levels of LGALS3, NAMPT, and GRN in Tregs within SCC. These pathways induce cancer proliferation, progression, and invasion [1, 38–40, 45]. This implies a novel mechanism whereby certain SCC clusters trigger Treg migration and activation, thus fostering an immunosuppressive microenvironment and SCC progression.
To investigate whether our observations from the scRNA-seq study were being recapitulated and to explore the spatial locations of the Carcinoma 3 cluster in target tissues, we performed ST analysis on SCC slides from independent patients. To specifically assess Carcinoma 3 at the tumor–immune boundary, we selected regions of interest (ROIs) enriched for PanCK+ tumor areas adjacent to immune cell infiltrates based on histology and marker expression. First, using the GeoMx DSP (NanoString platform), we compared the genes related to Carcinoma 3 discovered by scRNA-seq between SCC and PV to determine whether these tumor-associated transcriptional programs could be distinguished from nonmalignant epithelial inflammation. We observed that the expression of COL6A1 and ITGA5, as well as FTH1, TNC, LAMB3, S100A9, S100A11, STAT1, IFITM1, TPM3, and TPM4, which were expected to influence the progression of SCC, was significantly increased in SCC. These data suggest that Carcinoma 3 is preferentially located at the tumor–immune boundary and may possess invasive characteristics. Although COL6A1 was not clearly detected in tumor regions by immunofluorescence, its expression was consistently enriched in Carcinoma 3 across scRNA-seq and spatial transcriptomic data. The strong fluorescence signal in the surrounding stroma may have masked or diluted the detection of COL6A1 specifically in tumor areas. Additionally, we focused on ligand–receptor interactions predicted from NicheNet analysis of SCC–Treg communication. The expression of CXCL16, a chemokine implicated in Treg migration, was significantly upregulated in SCC_Tumor regions. Although its corresponding receptor CXCR6 was not differentially expressed, the CXCL16–CXCR6 pair exhibited a positive spatial correlation, suggesting potential coordination between tumor cells and neighboring Tregs. In contrast, TNFSF9 expression levels remained comparable between SCC and control samples, but its receptor TNFRSF9 was markedly elevated in the TME and showed a significant correlation with TNFSF9, indicating spatial co-regulation. These findings suggest that Carcinoma 3 may contribute to immune evasion in SCC by engaging ligand–receptor signaling pathways associated with Treg recruitment and activation. This potential interaction is further supported by spatial proximity between Carcinoma 3 cells and CXCR6+ regulatory T cells, as shown in serial immunofluorescence staining (Figure S2). In summary, we propose that Carcinoma 3, a tumor subpopulation enriched at the tumor–immune boundary, facilitates the formation of an immunosuppressive microenvironment. Furthermore, specific interaction between the SCC and Treg axis could potentially lead to the development of novel therapeutics for SCC.
Author Contributions
Hyun Seung Choi: data curation (lead), formal analysis (lead), investigation (equal), visualization (lead), writing – original draft (lead). Sunyoung Jung: conceptualization (equal), investigation (lead), validation (lead), writing – original draft (equal). Ki-Myo Kim: investigation (equal). Mihyun Lee: investigation (equal). Jun Ho Park: investigation (equal). Sanha Hwang: investigation (equal). Seung Min Cha: investigation (equal). Jade N. Young: writing – review and editing (equal). Dina Poplausky: investigation (equal). Hyunsung Nam: writing – review and editing (equal). Nicholas Gulati: writing – review and editing (equal). Chung-Gyu Park: writing – review and editing (equal). Hyun Je Kim: conceptualization (equal), funding acquisition (equal), investigation (equal), supervision (equal), validation (supporting), writing – review and editing (lead). Ji-Ung Park: conceptualization (equal), funding acquisition (lead), investigation (equal), supervision (equal), writing – review and editing (lead).
Acknowledgments
The authors express their gratitude to all patients who consented to participate in this study.
Disclosure
Integration analysis of scRNA-seq and ST of NMSC reveals mechanisms by which specific SCC clusters promote metastasis and shape an immunosuppressive TME, providing new approaches for targeted therapies to prevent SCC progression.
Ethics Statement
This study was approved by the institutional review boards at the Seoul National University Boramae Hospital (IRB number: 30–2017-11).
Consent
Consent obtained directly from patient(s).
Conflicts of Interest
The authors declare no conflicts of interest.
Data Availability Statement
The data generated in this study are available upon request from the corresponding author.
A. C. Geller and G. D. Annas, “Epidemiology of Melanoma and Nonmelanoma Skin Cancer,” Seminars in Oncology Nursing 19, no. 1 (2003): 2–11, https://doi.org/10.1053/sonu.2003.50000.
K. S. Nehal and C. K. Bichakjian, “Update on Keratinocyte Carcinomas,” New England Journal of Medicine 379, no. 4 (2018): 363–374, https://doi.org/10.1056/NEJMra1708701.
M. S. Chang, M. Azin, and S. Demehri, “Cutaneous Squamous Cell Carcinoma: The Frontier of Cancer Immunoprevention,” Annual Review of Pathology 17 (2022): 101–119, https://doi.org/10.1146/annurev‐pathol‐042320‐120056.
M. C. Cameron, E. Lee, B. P. Hibler, et al., “Basal Cell Carcinoma: Epidemiology; Pathophysiology; Clinical and Histological Subtypes; and Disease Associations,” Journal of the American Academy of Dermatology 80, no. 2 (2019): 303–317, https://doi.org/10.1016/j.jaad.2018.03.060.
M. C. G. Winge, L. N. Kellman, K. Guo, et al., “Advances in Cutaneous Squamous Cell Carcinoma, Nature Reviews,” Cancer 23, no. 7 (2023): 430–449, https://doi.org/10.1038/s41568‐023‐00583‐5.
P. Cammareri, A. M. Rose, D. F. Vincent, et al., “Inactivation of TGFβ Receptors in Stem Cells Drives Cutaneous Squamous Cell Carcinoma,” Nature Communications 7 (2016): 12493, https://doi.org/10.1038/ncomms12493.
Z. C. Venables, P. Autier, T. Nijsten, et al., “Nationwide Incidence of Metastatic Cutaneous Squamous Cell Carcinoma in England,” JAMA Dermatology 155, no. 3 (2019): 298–306, https://doi.org/10.1001/jamadermatol.2018.4219.
A. Boutros, F. Cecchi, E. T. Tanda, et al., “Immunotherapy for the Treatment of Cutaneous Squamous Cell Carcinoma,” Frontiers in Oncology 11 (2021): 733917, https://doi.org/10.3389/fonc.2021.733917.
G. J. Inman, J. Wang, A. Nagano, et al., “The Genomic Landscape of Cutaneous SCC Reveals Drivers and a Novel Azathioprine Associated Mutational Signature,” Nature Communications 9, no. 1 (2018): 3667, https://doi.org/10.1038/s41467‐018‐06027‐1.
J. Wan, H. Dai, X. Zhang, et al., “Distinct Transcriptomic Landscapes of Cutaneous Basal Cell Carcinomas and Squamous Cell Carcinomas,” Genes Dis 8, no. 2 (2021): 181–192, https://doi.org/10.1016/j.gendis.2019.10.004.
P. H. Li, X. Y. Kong, Y. Z. He, et al., “Recent Developments in Application of Single‐Cell RNA Sequencing in the Tumour Immune Microenvironment and Cancer Therapy,” Military Medicine Research 9, no. 1 (2022): 52, https://doi.org/10.1186/s40779‐022‐00414‐y.
C. F. Guerrero‐Juarez, G. H. Lee, Y. Liu, et al., “Single‐Cell Analysis of Human Basal Cell Carcinoma Reveals Novel Regulators of Tumor Growth and the Tumor Microenvironment,” Science Advances 8, no. 23 (2022): eabm7981, https://doi.org/10.1126/sciadv.abm7981.
A. L. Ji, A. J. Rubin, K. Thrane, et al., “Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma,” Cell 182, no. 2 (2020): 497–514, https://doi.org/10.1016/j.cell.2020.05.039.
X. Li, S. Zhao, X. Bian, et al., “Signatures of EMT, Immunosuppression, and Inflammation in Primary and Recurrent Human Cutaneous Squamous Cell Carcinoma at Single‐Cell Resolution,” Theranostics 12, no. 17 (2022): 7532–7549, https://doi.org/10.7150/thno.77528.
T. Stuart, A. Butler, P. Hoffman, et al., “Comprehensive Integration of Single‐Cell Data,” Cell 177, no. 7 (2019): 1888–1902, https://doi.org/10.1016/j.cell.2019.05.031.
M. D. Young and S. Behjati, “SoupX Removes Ambient RNA Contamination From Droplet‐Based Single‐Cell RNA Sequencing Data,” GigaScience 9, no. 12 (2020): 1–10, https://doi.org/10.1093/gigascience/giaa151.
P. L. Germain, A. Lun, C. Garcia Meixide, W. Macnair, and M. D. Robinson, “Doublet Identification in Single‐Cell Sequencing Data Using scDblFinder,” F1000Res 10 (2021): 979, https://doi.org/10.12688/f1000research.73600.2.
R. Lopez, J. Regier, M. B. Cole, M. I. Jordan, and N. Yosef, “Deep Generative Modeling for Single‐Cell Transcriptomics,” Nature Methods 15, no. 12 (2018): 1053–1058, https://doi.org/10.1038/s41592‐018‐0229‐2.
N. Liu, D. D. Bhuva, A. Mohamed, et al., “standR: Spatial Transcriptomic Analysis for GeoMx DSP Data,” Nucleic Acids Research 52, no. 1 (2024): e2, https://doi.org/10.1093/nar/gkad1026.
R. Gao, S. Bai, Y. C. Henderson, et al., “Delineating Copy Number and Clonal Substructure in Human Tumors From Single‐Cell Transcriptomes,” Nature Biotechnology 39, no. 5 (2021): 599–608, https://doi.org/10.1038/s41587‐020‐00795‐2.
Y. Liu, L. Y. Yang, D. X. Chen, et al., “Tenascin‐C as a Potential Biomarker and Therapeutic Target for Esophageal Squamous Cell Carcinoma,” Translational Oncology 42 (2024): 101888, https://doi.org/10.1016/j.tranon.2024.101888.
H. Zhang, Y. Z. Pan, M. Cheung, et al., “LAMB3 Mediates Apoptotic, Proliferative, Invasive, and Metastatic Behaviors in Pancreatic Cancer by Regulating the PI3K/Akt Signaling Pathway,” Cell Death & Disease 10, no. 3 (2019): 230, https://doi.org/10.1038/s41419‐019‐1320‐z.
T. Hou, C. Tong, G. Kazobinka, et al., “Expression of COL6A1 Predicts Prognosis in Cervical Cancer Patients,” American Journal of Translational Research 8, no. 6 (2016): 2838–2844.
K. G. Owusu‐Ansah, G. Song, R. Chen, et al., “COL6A1 Promotes Metastasis and Predicts Poor Prognosis in Patients With Pancreatic Cancer,” International Journal of Oncology 55, no. 2 (2019): 391–404, https://doi.org/10.3892/ijo.2019.4825.
Y. Kumagai, J. Nio‐Kobayashi, S. Ishihara, et al., “The Interferon‐β/STAT1 Axis Drives the Collective Invasion of Skin Squamous Cell Carcinoma With Sealed Intercellular Spaces,” Oncogene 11, no. 1 (2022): 27, https://doi.org/10.1038/s41389‐022‐00403‐9.
Y. Du, Y. Cai, Y. Lv, et al., “Single‐Cell RNA Sequencing Unveils the Communications Between Malignant T and Myeloid Cells Contributing to Tumor Growth and Immunosuppression in Cutaneous T‐Cell Lymphoma,” Cancer Letters 551 (2022): 215972, https://doi.org/10.1016/j.canlet.2022.215972.
F. Teng, J. Jiang, J. Zhang, et al., “The S100 Calcium‐Binding Protein A11 Promotes Hepatic Steatosis Through RAGE‐Mediated AKT‐mTOR Signaling,” Metabolism 117 (2021): 154725, https://doi.org/10.1016/j.metabol.2021.154725.
A. C. Pichler, N. Carrie, M. Cuisinier, et al., “TCR‐Independent CD137 (4‐1BB) Signaling Promotes CD8(+)‐Exhausted T Cell Proliferation and Terminal Differentiation,” Immunity 56, no. 7 (2023): 1631–1648, https://doi.org/10.1016/j.immuni.2023.06.007.
J. D. French and B. R. Haugen, “Thyroid Cancer: CAR T Cell Therapy ‐ Potential in Advanced Thyroid Cancer?,” Nature Reviews. Endocrinology 14, no. 1 (2018): 10–11, https://doi.org/10.1038/nrendo.2017.160.
A. Magen, P. Hamon, N. Fiaschi, et al., “Intratumoral Dendritic Cell‐CD4(+) T Helper Cell Niches Enable CD8(+) T Cell Differentiation Following PD‐1 Blockade in Hepatocellular Carcinoma,” Nature Medicine 29, no. 6 (2023): 1389–1399, https://doi.org/10.1038/s41591‐023‐02345‐0.
R. Muthuswamy, A. R. McGray, S. Battaglia, et al., “CXCR6 by Increasing Retention of Memory CD8(+) T Cells in the Ovarian Tumor Microenvironment Promotes Immunosurveillance and Control of Ovarian Cancer,” Journal for Immunotherapy of Cancer 9, no. 10 (2021): e003329, https://doi.org/10.1136/jitc‐2021‐003329.
J. W. Cho, J. Son, S. J. Ha, and I. Lee, “Systems Biology Analysis Identifies TNFRSF9 as a Functional Marker of Tumor‐Infiltrating Regulatory T‐Cell Enabling Clinical Outcome Prediction in Lung Cancer,” Computational and Structural Biotechnology Journal 19 (2021): 860–868, https://doi.org/10.1016/j.csbj.2021.01.025.
S. Sakaguchi, M. Miyara, C. M. Costantino, and D. A. Hafler, “FOXP3+ Regulatory T Cells in the Human Immune System,” Nature Reviews. Immunology 10, no. 7 (2010): 490–500, https://doi.org/10.1038/nri2785.
X. Long, Z. Zhang, Y. Li, et al., “ScRNA‐Seq Reveals Novel Immune‐Suppressive T Cells and Investigates CMV‐TCR‐T Cells Cytotoxicity Against GBM,” Journal for Immunotherapy of Cancer 12, no. 4 (2024), https://doi.org/10.1136/jitc‐2024‐008967.
J. Zhou, Y. Yang, Y. Zhang, H. Liu, and Q. Dou, “A Meta‐Analysis on the Role of Pleiotrophin (PTN) as a Prognostic Factor in Cancer,” PLoS One 13, no. 11 (2018): e0207473, https://doi.org/10.1371/journal.pone.0207473.
L. Gorvel and D. Olive, “Targeting the “PVR‐TIGIT Axis” With Immune Checkpoint Therapies,” F1000Res 9 (2020): 354, https://doi.org/10.12688/f1000research.22877.1.
H. Georgiev, I. Ravens, G. Papadogianni, and G. Bernhardt, “Coming of Age: CD96 Emerges as Modulator of Immune Responses,” Frontiers in Immunology 9 (2018): 1072, https://doi.org/10.3389/fimmu.2018.01072.
Y. Ma and H. Wang, “Clinical Significance of Annexin A2 Expression in Oral Squamous Cell Carcinoma and Its Influence on Cell Proliferation, Migration and Invasion,” Scientific Reports 11, no. 1 (2021): 5033, https://doi.org/10.1038/s41598‐021‐84675‐y.
P. K. Rajput, J. F. Varghese, A. K. Srivastava, U. Kumar, and U. C. S. Yadav, “Visfatin‐Induced Upregulation of Lipogenesis via EGFR/AKT/GSK3beta Pathway Promotes Breast Cancer Cell Growth,” Cellular Signalling 107 (2023): 110686, https://doi.org/10.1016/j.cellsig.2023.110686.
Z. He and A. Bateman, “Progranulin (Granulin‐Epithelin Precursor, PC‐Cell‐Derived Growth Factor, Acrogranin) Mediates Tissue Repair and Tumorigenesis,” Journal of Molecular Medicine (Berlin, Germany) 81, no. 10 (2003): 600–612, https://doi.org/10.1007/s00109‐003‐0474‐3.
J. H. Suh, S. Y. Choi, Y. J. Huh, et al., “Spatial Transcriptomics of Pemphigus Vulgaris and Bullous Pemphigoid: Insights Into Pathogenesis and Therapy on Bullous Formation,” Journal of the European Academy of Dermatology and Venereology 38, no. 10 (2024): e842–e845, https://doi.org/10.1111/jdv.19924.
M. Cescon, F. Gattazzo, P. Chen, and P. Bonaldo, “Collagen VI at a Glance,” Journal of Cell Science 128, no. 19 (2015): 3525–3531, https://doi.org/10.1242/jcs.169748.
N. Tani, S. Higashiyama, N. Kawaguchi, et al., “Expression Level of Integrin Alpha 5 on Tumour Cells Affects the Rate of Metastasis to the Kidney,” British Journal of Cancer 88, no. 2 (2003): 327–333, https://doi.org/10.1038/sj.bjc.6600710.
K. Sawada, A. K. Mitra, A. R. Radjabi, et al., “Loss of E‐Cadherin Promotes Ovarian Cancer Metastasis via Alpha 5‐Integrin, Which Is a Therapeutic Target,” Cancer Research 68, no. 7 (2008): 2329–2339, https://doi.org/10.1158/0008‐5472.CAN‐07‐5167.
P. Shetty, A. Bargale, B. R. Patil, et al., “Cell Surface Interaction of Annexin A2 and Galectin‐3 Modulates Epidermal Growth Factor Receptor Signaling in Her‐2 Negative Breast Cancer Cells,” Molecular and Cellular Biochemistry 411, no. 1–2 (2016): 221–233, https://doi.org/10.1007/s11010‐015‐2584‐y.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025. This work is published under http://creativecommons.org/licenses/by/4.0/ (the "License"). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
ABSTRACT
Background
Cutaneous squamous cell carcinoma (cSCC) and basal cell carcinoma (BCC) are the most prevalent types of nonmelanoma skin cancer (NMSC) and exhibit significant inter‐ and intra‐tumor heterogeneity. cSCC has a higher metastatic potential than BCC, accompanied by a considerable mortality rate. However, the detailed mechanisms of tumor evolution in cSCC have not yet been described.
Methods
We performed single‐cell RNA sequencing (scRNA‐seq) and T cell receptor (TCR) clonal analysis of skin biopsies from five BCCs, three squamous cell carcinomas in situ (SCCIS), and two invasive squamous cell carcinomas (SCC). Independent SCC specimens were used for spatial transcriptomic (ST) analysis using GeoMx Digital Spatial Profiler (DSP).
Result
Using scRNA‐seq, we analyzed a total of 117,663 cells. We distinguished cancer cells using copy number variation and identified SCC‐specific genes that potentially contribute to tumor progression. Analysis of tumor clones revealed SCC‐specific
Conclusion
We suggest COL6A1 and ITGA5 promote the invasive and metastatic property of SCC. We also uncovered how SCC recruits Tregs via the CXCL16/CXCR6 axis to create a TME favorable for its survival. These molecules can be used as potential therapeutic targets for treatment of SCC.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details

1 Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, the Republic of Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, the Republic of Korea
2 Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, the Republic of Korea
3 Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, the Republic of Korea
4 Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA
5 Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, the Republic of Korea
6 Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, the Republic of Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, the Republic of Korea, Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, the Republic of Korea
7 Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, the Republic of Korea, Cancer Research Institute, Seoul National University College of Medicine, Seoul, the Republic of Korea, Department of Dermatology, Seoul National University Hospital, Seoul, the Republic of Korea, Interdisciplinary Program in Artificial Intelligence (IPAI), Seoul National University, Seoul, the Republic of Korea