Abstract Breast cancers present intricate microenvironments comprising heterotypic cellular interactions, yet a comprehensive spatial map remained to be established. Here, we employed the DNA nanoball-based genome-wide in situ sequencing (Stereo-seq) to visualize the geospatial architecture of 30 primary breast tumors and metastatic lymph nodes across different molecular subtypes. This unprecedented high-resolution atlas unveils the fine structure of the tumor vasculature, highlighting heterogeneity in phenotype, spatial distribution, and intercellular communication within both endothelial and perivascular cells. In particular, venular smooth muscle cells are identified as the primary source of CCL21/CCL19 within the microenvironment. In collaboration with ACKR1-positive endothelial cells, they create a chemokine-rich venular niche to synergistically promote lymphocyte extravasation into tumors. High venule density predicts increased immune infiltration and improved clinical outcomes. This study provides a detailed spatial landscape of human breast cancer, offering key insights into the venular regulation of tumor immune infiltration. Subject terms: Breast cancer, Cancer microenvironment, Immunoediting, Next-generation sequencing __________________________________________________________________ This study utilizes Stereo-seq to comprehensively map the geospatial architecture of breast cancer, revealing venular niches that promote immune infiltration and better clinical outcomes, offering new insights into tumor microenvironment regulation. Introduction Breast cancer is a highly heterogeneous collection of tumors that are characterized by distinct molecular subtypes^[86]1. Advances in the subtype-specific therapies have yielded significant survival benefits for patients, primarily through the administration of hormone and HER2-targeted therapy^[87]2. In recent years, there has been a growing interest in the combination of immune checkpoint inhibitors (ICIs) and chemotherapy as a treatment for breast cancer^[88]3. The success of several clinical trials, including IMpassion130^[89]4, KEYNOTE-355^[90]5, and KEYNOTE-522 trials^[91]6, has led to the approval of atezolizumab and pembrolizumab for triple-negative breast cancer patients with PD-L1 expression. Despite these promising achievements, the efficacy of ICIs in treating other molecular subtypes and PD-L1-negative patients remains limited, partly due to inadequate immune infiltration in the tumor microenvironment (TME)^[92]3,[93]7. A deeper understanding of TME heterogeneity and the mechanisms dictating immune infiltration is required to optimize immunotherapy efficacy. The post-capillary venules have been identified as the major channel for lymphocytes extravasation through the endothelial barrier^[94]8,[95]9. Mechanistically, circulating lymphocytes engage venular endothelial cells (ECs) under shear stress via the binding of endothelial selectins, P-selectin and E-selectin, leading to the tethering and rolling of lymphocytes along the vessel wall^[96]8. Rolling lymphocytes are then activated through the interaction between CCR7 and homeostatic chemokines, including CCL21 and CCL19, that are immobilized on the EC surface^[97]10. Subsequently, activation of lymphocytes mediates firm adhesion and enables lymphocytes to crawl on the luminal surface of venules until they transmigrate through the barriers of ECs, associated pericytes/vascular smooth muscle cells (SMCs), and the basement membrane^[98]11. Previous single-cell and experimental studies have provided valuable insights into the interaction between ECs and lymphocytes^[99]12–[100]15, but ECs present significant plasticity and spatial heterogeneity that are coordinated with different functional states^[101]12. The spatial organization of venular ECs and other EC functional subtypes within intact tumors from patients in a clinical setting remains unclear. More importantly, our understanding of their interaction and coordination with perivascular cells in mediating immune infiltration is still limited, primarily due to limitations in technical precision and a lack of suitable experimental models^[102]11,[103]16. A high-resolution, unbiased spatial map of the tumor ecosystem and its vasculature could help to elucidate the complex spatial and functional networks between the immune and vascular systems. The application of spatial transcriptomics (ST) technologies has revolutionized our ability to retrieve spatial information that is lost in single-cell RNA sequencing (scRNA-seq)^[104]17. Wu et al. performed ST on six breast cancers to reveal the coordination of tumor and host cell phenotypes^[105]18. However, conventional ST methods, such as Visium, exhibit insufficient resolution due to their reliance on 55 μm barcoded spots, with a center-to-center distance of 100 μm, that encompass dozens of cells, thus impeding precise localization of dispersed ECs and perivascular cells within the TME. An innovative technology, SpaTial Enhanced REsolution Omics-sequencing (Stereo-seq)^[106]19, has recently been developed that combines 220 nm DNA nanoball (DNB)-patterned arrays and in situ tissue RNA capture to create spatial maps at cellular resolution, making it an ideal tool to depict the intricate tumor vasculature^[107]20. In this study, we applied Stereo-seq to characterize the geospatial architecture and cell-cell communication in 30 surgically resected specimens of primary breast tumors and paired lymph node metastases. We unveiled that tumor vasculature displayed distinct heterogeneity of biological phenotype, spatial organization, and interactions with surrounding cells. CCL21+ venular SMCs and ACKR1+ venular ECs cooperated through multiple chemokines to promote lymphocyte infiltration in breast cancer. Results Depicting a high-resolution spatial atlas of human breast cancer To comprehensively characterize the spatial landscape of breast cancer, we employed Stereo-seq on 30 fresh frozen tissues derived from 23 breast cancer patients, including 23 primary breast cancers and 7 paired metastatic lymph nodes that collectively covered all four molecular subtypes (luminal A: n = 8; luminal B: n = 9; HER2: n = 8; triple-negative: n = 5). Adjacent tumor tissues were subjected to scRNA-seq to facilitate cell type identification in the ST slides. We further applied immunohistochemistry (IHC) and multiplex immunofluorescence (mIF) on the adjacent slides of all 30 ST slides and on the validation cohort to confirm our discovery (Fig. [108]1a). Detailed clinical and pathological information of patients is provided in the Supplementary Data [109]1. Fig. 1. A spatially resolved atlas of human breast cancer slides across different molecular subtypes and tumor sites. [110]Fig. 1 [111]Open in a new tab a Schematic diagram illustrating the acquisition and analysis workflow of the ST maps. Stereo-seq was performed on 30 slides of primary breast tumors and paired lymph node metastases from 23 patients, yielding a total of 4,894,305,510 DNBs. ScRNA-seq was performed on the fresh tumor samples from 6 out of 23 patients and identified 78,848 qualified cells. Each block represents a ST slide and is colored by molecular subtype. Red, blue, orange, and green spots represent venular endothelial cells (vECs), venular smooth muscle cells (vSMCs), tumor cells, and immune cells, respectively, in the projection of scRNA-seq and ST data. Schematic plots were created using bioRender. b Uniform manifold approximation and projection (UMAP) plot of the scRNA-seq dataset. Each cluster is colored by its corresponding cell type. EC: endothelial cell; SMC: smooth muscle cell, including vascular SMC and pericyte; T/NK: T or natural killer cell. c Differential expression of canonical markers in each cell type cluster. Macro: macrophage; Fibro: fibroblast; Melano: melanocyte; Granulo: granulocyte; PB: plasmablast. d–f Hematoxylin and eosin (H&E) staining (d), gene count maps (e), and cell type annotation maps (f) of the ST slides of the primary tumor (PT) and metastatic lymph node (LN) from the patient NCC-P05. ST spots are mapped to the H&E staining of adjacent slides. Raw spatial expression matrix of each bin/spot is convoluted into pseudo 50 spot with 25 µm squares (50 × 50 bins/spot, bin50 spot for short). IDC: invasive ductal carcinoma; DCIS: ductal carcinoma in situ. g The cell type annotation, H&E staining, gene counts, and unique molecular identifiers (UMI) counts in the selected area of NCC-BR5 slide highlighted in Fig. 1d–f. h Relative proportions of cell types across 30 ST slides. ST slides are grouped according to the molecular subtype and tumor site. After quality control (Supplementary Fig. [112]1a-c), the scRNA-seq analysis of 6 breast cancer samples identified 78,848 qualified cells, which were categorized into 13 dominant cell types based on the canonical cell markers, including breast tumor cell (EPCAM), T or natural killer (T/NK) cell (TRBC2), B cell (CD79A), plasma cell (JCHAIN), plasmablast (GZMB and JCHAIN), macrophage (CD163), fibroblast (DCN), EC (VWF), vascular SMC/pericyte (referred to as SMC; NOTCH3), basal cell (COL17A1), neuron (CNTN5), granulocyte (CPA3), and melanocyte (CDH19) (Fig. [113]1b, c). In order to validate the robustness of cell classification, we conducted cell clustering and cell type annotation on an external scRNA-seq dataset of 26 breast cancer samples consisting of 88,876 cells^[114]18, which produced similar clustering results and feature genes for the dominant cell types (Supplementary Fig. [115]1d, e). For the ST data, Stereo-seq of 30 slides yielded a total of 4,894,305,510 DNBs (220 nm × 220 nm), with an average of 163,143,517 DNBs per slide. Utilizing a resolution similar to that of Visium, Stereo-seq captured an average of 7565 pseudo-spots (bin200 spot, 200 × 200 DNB bins) per slide. Each spot, on average, quantified approximately 6120 genes (Supplementary Fig. [116]1f). To gain a more detailed view of slides with sufficient genes for spatial annotation, we convoluted the raw spatial expression data of each slide into smaller pseudo-spots (bin50 spot, 50 × 50 DNB bins, 25 μm diameter) (Fig. [117]1d-g), with each bin50 spot covering approximately 3-5 tumor cells and having an average detection of 921 genes and 2058 UMIs per spot (Fig. [118]1g) (Supplementary Fig. [119]1g). Random sampling of 10,000 bin50 spots taken from each slide showed few batch effects (Supplementary Fig. [120]1h). Using the internal and external scRNA-seq results as independent reference datasets, we utilized the Robust Cell Type Decomposition (RCTD) algorithm to infer the cell composition for each bin50 spot^[121]21. Annotation of eight major cell types, including tumor cell, T/NK cell, B cell, macrophage, fibroblast, EC, SMC, and basal cell, produced highly correlated results in both references