Abstract While cervical cancer (CC) prognosis depends on tumor staging, the spatiotemporal evolution of tumor microenvironment (TME) heterogeneity during metastatic progression remains poorly characterized at single-cell resolution. We employed an integrative multi-omics approach, combining single-cell RNA sequencing (scRNA-seq; n = 11), spatial transcriptomics (ST), and bulk RNA-seq data from the TCGA-CESC cohort (n = 304), to systematically map TME remodeling across CC progression stages. scRNA-seq was performed on primary lesions from patients with localized (n = 3), regional (n = 4), and metastatic (n = 4) diseases, with in-depth analyses focusing on cellular characteristics, cell type composition alterations, functional changes, differentiation trajectories, and cell-cell interaction networks. These findings were further validated using spatial transcriptomics, bulk RNA-seq data, and multiple immunohistochemistry (mIHC) experiments. ScRNA-seq data revealed that the TME of the metastatic group displayed a distinct immunosuppressive phenotype. Three key subclusters closely linked to TME remodeling in this group were identified. Notably, a novel metastasis-associated epithelial subpopulation (Epi0_AGR2), characterized by both epithelial-mesenchymal transition (EMT) and chemokine secretory phenotypes, was discovered. Gene Set Variation Analysis (GSVA) revealed that transforming growth factor β (TGF-β) signaling activation served as its primary transcriptional driver. Additionally, a neutrophil subset with pro-tumor and immunosuppressive properties, as well as a cancer-associated fibroblasts (CAFs) subset that promoted angiogenesis, were enriched in the metastatic group. Cell-cell interaction analysis and spatial mapping further revealed the formation of coordinated Epi0-neutrophil-CAFs niches, which established TGF-β-CXCL1/2/8-OSM/OSMR feedforward loops. Importantly, a computational model derived from the TME metastatic niche signature demonstrated significant prognostic stratification in the TCGA cohort (HR = 2.5179, p = 0.0144). In all, this study provides the first comprehensive delineation of stage-specific TME dynamics in CC, revealing TGF-β-driven cellular cooperativity as a metastatic switch. The joint framework establishes a potential clinically translatable tool for precision prognosis and therapeutic targeting. Keywords: cervical cancer, tumor progression, multi-omics analysis, TME, niche Introduction Cancer metastasis is associated with tumor progression and lethality in cases of cervical cancer (CC) [49]^1. The five-year survival rates of patients with stage III and stage IV CC were only 10-40% [50]^2. The recent introduction of immune checkpoint inhibitors (ICIs) as a treatment for advanced CC patients resulted in a significant improvement in their progression-free survival and overall survival [51]^3^-[52]^6. The response to ICIs has been shown to be closely associated with the initial tumor microenvironment (TME) [53]^7^, [54]^8. Therefore, a comprehensive understanding of the TME in advanced CC, and the exploration of the underlying mechanisms that promote cancer progression and immunosuppression, will offer new insights on the treatment and management of CC. Metastasis is not a simple linear cascade of events but involves multiple parallel and intersecting pathways [55]^9^-[56]^11. The phenotypes of metastasis-initiating cells (MICs) depend on the interaction among various cell types [57]^12^-[58]^14, as well as modulations of multiple signaling pathways during distinct metastatic phases, including local invasion, dissemination, and colonization [59]^15. Among multiple signaling pathways involved in the process, transforming growth factor β (TGF-β) signaling has a crucial role in metastasis and cancer progression. It can not only stimulate epithelial mesenchymal transition (EMT) and dissemination of tumor cells [60]^15^, [61]^16, but also create an immunosuppressive TME via inhibiting the expansion and function of immune cells [62]^17. Taniguchi et al. reported the presence of an invasion niche in a squamous cell carcinoma mouse model which creates a positive feedback loop promoting tumor cell invasion and malignant progression via TGF-β [63]^18. Thus, further exploration is required to unveil the multicellular interactions and spatiotemporal relevance of TGF-β signaling during the progression and metastasis of CC. Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for characterizing TME and dissecting intratumor heterogeneity [64]^19. Several scRNA-seq studies predominantly focused on investigating the diversity and heterogeneity of TME in primary CC or CC initiation [65]^20^-[66]^24. However, how tumor cells crosstalk with other important cell types to reprogram the TME of CC in both temporal and spatial dimensions during progression remain unclear. In this study, we integrated scRNA-seq profiling of localized, regional, and metastatic TME to elucidate the dynamic alterations in the proportion and the biological function of different cellular components. We identified a TGF-β signaling-responsive malignant subset, as well as pro-tumor neutrophils and cancer-associated fibroblasts (CAFs) that secreted TGF-β. These cells were found to be enriched in metastatic TME. Combining scRNA-seq data with spatial transcriptomics (ST) data further elucidated the colocalization and cell-cell interactions between these subsets. In addition, we provided a clustering strategy based on bulk RNA-seq that related these key clusters with metastasis and patients' prognosis. Materials and methods Data collection The raw scRNA-seq data were obtained from our previously deposited database (the GSA human genome sequence archive: HRA001742 and HRA007492). Primary CC patients receiving no anti-tumor treatment at the time of sampling were included, patients with secondary CC or associated with other primary tumors were excluded. Detailed sources and clinical information of the patients in the scRNA-seq cohort are listed in [67]Table S1. Our cohort included three localized primary tumors, four regional primary tumors and four metastatic primary tumors of CC. The processed spatial transcriptomics data were obtained from Science Data Bank ([68]https://doi.org/10.57760/sciencedb.11624), published by Fan et al. [69]^21. Normalized The Cancer Genome Atlas (TCGA) expression matrix and clinical information for cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) cohorts were obtained from the cBioPortal website ([70]http://www.cbioportal.org/). scRNA-seq data preprocessing Sequencing data were aligned to the GRCh38 human reference genome and then processed into the initial Unique Molecular Identifier (UMI) matrix using the Cell Ranger Single-Cell toolkit (version 7.0.1). The Seurat workflow (version 4.2.1) was utilized for downstream analyses with default parameters unless otherwise stated. Quality control measures were implemented to exclude cells with fewer than 1000 UMIs or fewer than 500 features or a mitochondrial gene fraction greater than 25%. The NormalizeData function was then used to normalize the library size and log-transform the expression matrix to prepare it for further downstream analyses. Data integration and cluster identification Two rounds of unsupervised clustering were performed to identify major cell types and subclusters, and to eliminate residual doublets. The FindVariableFeatures function was used to identify the highly variable genes (HVGs). After scaling the data, Principal Component Analysis (PCA) was performed using the RunPCA function with the top 2000 HVGs for dimensionality reduction. For visualization, Uniform Manifold Approximation and Projection (UMAP) was implemented to reduce dimensionality in the RunUMAP function with the top 20 principal components. As we observed batch effects between patients in the UMAP plot, we used the canonical correlation analysis (CCA) function to integrate expression data from different samples for batch effect correction. Cell clusters were annotated based on cluster-specific marker genes found by the FindAllMarkers function. Any clusters expressing more than two canonical cell type markers were considered as contaminated doublets and removed. Repeated dimension reduction and unsupervised clustering was performed if any doublets were removed. The first round of cell clustering and annotation identified major cell types including T and natural killer (T/NK) cells, myeloid cells, epithelial cells, B/plasma cells and stromal cells. To further analyze these major cell types, T/NK cells, myeloid cells, epithelial cells, B/plasma cells and stromal cells were reintegrated, re-clustered and re-annotated using the procedure described above. Distributed preference analysis To characterize the tissue distribution of different clusters, odds ratios (OR) were calculated to indicate preferences as previously