Abstract The spatial proteomic profiling of complex tissues is essential for investigating cellular function in physiological and pathological states. However, the imbalance among resolution, protein coverage, and expense precludes their systematic application to analyze whole tissue sections in an unbiased manner and with high resolution. Here, we introduce panoramic spatial enhanced resolution proteomics (PSERP), a method that combines tissue expansion, automated sample segmentation, and tryptic digestion with high-throughput proteomic profiling. The PSERP approach facilitates rapid quantitative profiling of proteomic spatial variability in whole tissue sections at sub-millimeter resolution. We demonstrated the utility of this method for determining the streamlined large-scale spatial proteomic features of gliomas. Specifically, we profiled spatial proteomic features for nine glioma samples across three different mutation types (IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT gliomas) at sub-millimeter resolution (corresponding to a total of 2,230 voxels). The results revealed over 10,000 proteins identified in a single slide, which helps us to portray the diverse proteins and pathways with spatial abundance patterns in the context of tumor heterogeneity and cellular features. Our spatial proteomic data revealed distinctive proteomic features of malignant and non-malignant tumor regions and depicted the distribution of proteins from tumor centers to tumor borders and non-malignant tumor regions. Through integrative analysis with single-cell transcriptomic data, we elucidated the cellular composition and cell–cell communications in a spatial context. Our PSERP also includes a spatially resolved tumor-specific peptidome identification workflow that not only enables us to elucidate the spatial expression patterns of tumor-specific peptides in glioma samples with different genomic types but also provides us with opportunities to select combinations of tumor-specific mutational peptides whose expression could cover the maximum tumor regions for future immune therapies. We further demonstrated that combining tumor-specific peptides might enhance the efficacy of immunotherapy in both patient-derived cell (PDC) and patient-derived xenograft (PDX) models. PSERP efficiently retains precise spatial proteomic information within the tissue context and provides a deeper understanding of tissue biology and pathology at the molecular level. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-025-01710-5. Keywords: Spatial proteomics, Tumor heterogeneity, Tumor microenvironment, Glioma Introduction Tissues are composed of various microscopic features, cell types, and phenotypically diverse subpopulations, and the spatial localization of cells within the tumor and their spatial neighborhood is crucial for elucidating their identity and functions [[54]1]. The cellular composition of tissue has a substantial influence on measured co-expression signals within the molecular profiles of bulk tissue and their microenvironment, contributing to the influence of cellular function, signaling, and different disease outcomes [[55]2]. Spatial molecular profiling of complex tissues is essential for elucidating cellular function in physiological and pathological states, providing critical insights into disease mechanisms. Spatial techniques such as spatial transcriptomics have undergone rapid progress, from early spatial detection methods, such as the 10 × Genomics Visium platform, serial microtomy sequencing (tomo-seq), to recent high-resolution spatially resolved transcriptomic methods, such as Stereo-seq, SeqScope, and PIXEL-seq [[56]3–[57]5]. Scientists can now profile transcript expression in an unbiased and panoramic manner. Similar to spatial transcriptomics, emerging methods involving spatial genomics, metabolomics and epigenomics are also being developed. For instance, spatial ATAC-seq can be performed by in situ Tn5 transposition and probe ligation via microfluidics devices, followed by digestion and sequencing to profile chromatin accessibility [[58]6]. Moreover, spatially resolved metabolic techniques, such as matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) [[59]7], hold significant potential for revealing the spatial distribution of metabolic species and molecular interactions. Despite the rapid development of spatial-resolved techniques for transcriptomics and metabolomics, spatial proteomics remains challenging due to the limited tools available. Currently, widely used spatial proteomic analysis methods include laser capture microdissection (LCM)-based methods and antibody-based spatial proteomics methods, such as MACSima Imaging Cyclic Staining (MICS) [[60]8], tissue-based cyclic immunofluorescence (t-CycIF) [[61]9], and codetection by IndEXing (CODEX) [[62]10]. While LCM-based spatially resolved proteomics is an elegant approach that enables the microscopic isolation of defined regions from tissue-preserving crucial spatial information, it is time-consuming, requires sophisticated equipment, and is limited by its efficiency. Consequently, its spatial coverage is inevitably limited to a comparatively small sample size, typically hundreds of micrometers. Antibody-based methods, on the other hand, depend on the affinity and specificity of antibodies, which compromises their reproducibility and multiplexing capacity (currently limited to approximately 50–100 proteins), thus impeding scalability. Therefore, there is an urgent need to develop an unbiased, high-throughput, and panoramic spatial proteomic approach that facilitates systematic and comprehensive analysis of tissue proteomics. The rapid development of spatial techniques has provided an opportunity to better decipher the complex tumor microenvironment (TME) and enhance immunotherapy efficacy [[63]1, [64]6]. Currently, immune checkpoint inhibitor (ICI) and chimeric antigen receptor T-cell (CAR-T) therapies, which leverage the human immune system to target and eliminate cancer cells, represent the most promising therapeutic strategies for treating malignant cancers. However, patients with an immune"desert"in their microenvironment benefit little from immunotherapy [[65]11]. Neoantigens, which are derived from nonsynonymous somatic mutations and are specific to tumor cells, offer the potential for targeted tumor destruction without toxic side effects on normal tissues [[66]12, [67]13]. The personalized neoantigen vaccine strategy can be a major step in improving the poor tumor microenvironment. However, identifying and selecting personalized neoantigens remains a significant challenge. Current methods for neoantigen prediction lack expression data [[68]13, [69]14], which limits their therapeutic potential and complicates the validation process. Developing an improved method for identifying and quantifying neoantigens, which is able to demonstrate their tumor-specific expression and spatial distribution, could enhance the efficacy of neoantigen-based immunotherapies. Spatially resolved proteomics using tissue expansion is a cutting-edge approach, in which samples are prepared by adhering a tissue section to a stretchable hydrogel [[70]15, [71]16]. Then, defined-sized tissue fragments can be isolated from regions of interest within the tissue section and subsequently subjected to mass spectrometry analysis [[72]15]. This approach enhances spatial resolution; however, it is technically complex, labor-intensive, prone to potential batch effects, and limited by insufficient throughput due to manual processing. Additionally, the limited throughput of current tissue expansion-based spatial proteomics techniques hinders their integration with spatial transcriptomics and single-cell transcriptomics data. These limitations hinder the large-scale implementation of unbiased spatial proteomic analysis in complex tumor ecosystems. Spatial proteomics via tissue expansion significantly enhanced the resolution, yet since the existing procedure for tissue expansion-based proteomics is very sophisticated and relies on manual labor, it is still technically fragile and challenging to implement and has insufficient throughput [[73]15]. Limitations in detection throughput and potential batch effects due to manual labor have prevented the utilization of this approach in the completion of unbiased spatial proteomics analysis of larger tumor ecosystems. In parallel, the throughput of the current tissue expansion spatial proteomics technique is still inadequate, making profiling results difficult to utilize for further integrative analysis with data from spatial transcriptomics and single-cell transcriptomics. To address these challenges, we developed panoramic spatial enhanced resolution proteomics (PSERP), a method that combines tissue expansion, automated sample segmentation using a three-dimensionally (3D) printed cutting toolkit, and tryptic digestion with high-throughput proteomic profiling. PSERP enables rapid quantitative profiling of proteomic spatial diversity across the entire tissue section at sub-millimeter resolution (Methods). In this proof-of-principle study, we validated the PSERP method using gliomas with different genomic alterations and demonstrated its capability for in-depth characterization of tumor ecosystems, including the cellular composition, distribution, and cell–cell communication within the TME. PSERP includes a spatially resolved tumor-specific peptidome identification workflow, enabling the systematic analysis of tumor-specific peptides and their impact on immune therapies. By utilizing PSERP, we delineated the distinctive proteomic signatures of malignant and nonmalignant regions, illustrating the dynamic expression patterns of proteins from tumor centers to borders and nonmalignant areas. Integrating this data with single-cell transcriptomics, we provided a comprehensive view of the tumor ecosystem and cell‒cell interactions in glioma. We also identified shared and distinct proteomic features across glioma samples with or without IDH1/EGFR mutations and analyzed the spatial expression of tumor-specific peptides (potential neoantigens), confirming their impact on the tumor microenvironment in both patient-derived cell (PDC) and patient-derived xenograft (PDX) models. PSERP has the potential to provide novel insights into the molecular landscape of tumors, thereby paving the way for future advancements in precision medicine and targeted immunotherapy. Results Spatially resolved proteomics of glioma using a panoramic spatial enhanced resolution proteomics (PSERP) approach To achieve high-resolution spatially resolved proteomics in tissue samples, we developed the PSERP approach, comprising the following systematic steps (Fig. [74]1A and S1 A, Methods). In Step 1, we performed tissue expansion process, enabling the physical enlargement of the tissue while preserving protein localization (Methods). In Step 2, we employed the custom-designed 3D-printed cutting toolkit, allowing the expanded tissue to be precisely sectioned into thousands of “voxels” of defined dimensions (Figure S1B-D, Methods). In Step 3, these divided voxels were transferred into 96-well plates, with their spatial coordinates meticulously recorded to maintain positional context (Figure S1E, Methods). Step 4 involved in-gel digestion and peptide extraction from each voxel, preparing the samples for downstream analysis. Finally, in Step 5, the extracted peptides were subjected to high-throughput data-independent acquisition (DIA) liquid chromatography-tandem mass spectrometry (LC–MS/MS) for comprehensive spatial proteomic profiling, phosphoproteomic profiling, and neoantigen profiling (Fig. [75]1A, Methods). To facilitate a comprehensive understanding of the PSERP workflow, we have included a detailed timeline from tissue preparation to LC–MS/MS analysis in Fig. [76]1B. This approach was optimized for time efficiency, and the majority of the workflow can be automated on a robotic workstation (Figure S1 A, Methods). Fig. 1. [77]Fig. 1 [78]Open in a new tab Overview of the panoramic spatial enhanced resolution proteomics (PSERP) workflow and its application. A A schematic representation of the PSERP workflow, encompassing the steps involved in neoantigen profiling. B Timeline illustrating the duration of each step in the PSERP process, from sample processing to LC–MS/MS data acquisition. C Hematoxylin and eosin (H&E) stained images and histological annotations of samples obtained from glioma patients. The samples include: G1, G4, and G5 (IDH1-WT/EGFR-mutant); G2, G6, and G7 (IDH1-mutant); G3, G8, and G9 (EGFR & IDH1 wild-type). The dotted line delineates the malignant tumor regions The primary objective of this proof-of-principle study was to validate the capabilities of our PSERP-based spatial proteomics method to capture the spatial heterogeneity of tumors and their corresponding microenvironments. The mutual exclusivity and microenvironment diversities of EGFR- and IDH1-mutant gliomas are well-documented in previous studies [[79]17]. EGFR mutations predominantly occur in IDH1-wildtype (WT) tumors. Thus, we focused on IDH1-mutant and two types of IDH1-wildtype (WT) gliomas: IDH1-WT/EGFR-mutant (G1, G4 and G5), IDH1-mutant (G2, G6 and G7), and IDH1/EGFR-double-WT (G3, G8 and G9) gliomas. For this proof-of-principle study, we sampled sections from a total of nine samples (G1-G9), and processed them with PSERP procedures (Fig. [80]1C, Methods). The isolated voxels were then processed following the PSERP workflow and analyzed using the DIA strategy (Methods). A total of 12,190 proteins were identified across 2,230 voxels from all glioma samples. The median number of proteins identified was 1,650 and the standard deviation was 562 across samples. Herein, we obtained the panoramic spatial proteomic profiles with comprehensive coverage of whole sections of glioma samples. The fidelity, quantification and reproducibility of PSERP In this proof-of-principle study, we consistently applied a sixfold expansion factor across all samples, resulting in an isotropic increase in length, width, and height. This corresponded to a 216-fold (6 by 6 by 6) volumetric increase in the sample. To illustrate the changes in tissue size before and after expansion, we have provided representative images of the samples in their pre-expanded and post-expanded states (Figure S2 A). Additionally, we applied PSERP to consecutive replicate sections of glioma tissue, and provided the images of the corresponding replicate samples after expansion to show the consistency (Figure S2B). The replicate samples expanded uniformly by the same expansion factor, indicating a high degree of consistency between replicate samples before and after expansion. Furthermore, to validate the consistency of the expanded tissues, we performed isotropic distortion analysis to assess the expansion distortion. Specifically, the pre- and post-expanded tissue images were recorded using the Biorad imaging system (ChemiDoc, Biorad), a B-spline-based image registration MATLAB package was used to register post-PSERP to pre-PSERP image by landmark selection [[81]15]. The root-mean-square (RMS) length measurement error of feature measurement was calculated to quantify the percentages of the distortion in the expanded tissue. In this way, we found that in our study, after tissue expansion over length scales up to 1,500 μm with the linear expansion factor of six fold, the average of feature RMS errors in our samples was 5% (Figure S2 C and S2D), demonstrating high fidelity in the preservation of tissue architecture. All samples in our study were processed using the same protocol, ensuring comparability and reliability across different samples. To evaluate the reproducibility, we utilized two consecutive tissue slides from one sample block (G1 sample) for the replicate experiment. These two consecutive slides were named G1-R1 and G1-R2, respectively (Figure S3 A and S3B). We processed these two tissue slides following the same protocol, including gel embedding, expanding, cutting, transferring, peptide extraction, and MS detection steps. During transferring process, we recorded the (x,y) coordinates of all voxels for both G1-R1 and G1-R2 samples to map them back to their original positions of the tissue slide (Methods). Then, we specifically selected 12 voxels within the malignant regions and 15 voxels within the non-malignant regions of the G1 and G1 replicate samples, respectively. The corresponding voxels with the same (x,y) coordinates in G1-R1 and G1-R2 samples were utilized to assess the reproducibility between samples (Figure S3 A and S3B). We generated proteomic data from selected voxels in G1-R1 and G1-R2 samples for further analysis (Figure S3 C). For the G1-R1 sample, we identified an average of 1,440 proteins per voxel in the non-malignant region, and 1,722 proteins per voxel in the malignant region. For the G1-R2 sample, we identified an average of 1,518 proteins per voxel in the non-malignant region, and 1,729 proteins per voxel in the malignant region. Additionally, 93% of the proteins identified in the non-malignant voxels of G1-R1 were consistently identified in their corresponding replicates of G1-R2, while 97% of proteins overlapped between malignant voxels. This substantial overlap supported the reproducibility of our method. We observed high correlation coefficients (R > 0.92) between corresponding voxels in G1-R1 and G1-R2 samples in both the malignant region and non-malignant region (Figure S3D). Clustering analysis revealed a clear distinction between voxels from malignant and non-malignant regions (Figure S3E). This finding indicated that the differential proteomic patterns were consistently preserved in both G1-R1 and G1-R2 samples. Additionally, the evaluation of protein diffusion between voxels was necessary for assessing the reliability of the method. In light of this, we assessed protein diffusion by sampling a homogeneous glioma tissue. After the same processing protocol mentioned above, we collected proteomic data from voxels containing the tissue sample and adjacent blank gel voxels without any tissue. Then, we compared the results between the tissue gel voxels and the adjacent blank gel voxels. The results showed that an average of 2,641 proteins were identified in tissue gel voxels (n = 10), while only an average of 57 proteins were identified in blank gel voxels (n = 10) (Figure S3 F). We observed that marker proteins, such as GFAP, EGFR, VIM, and PARP1, were significantly more abundant in glioma tissue voxels, while they were barely detectable in the blank gel voxels (Figure S3G). Although several proteins were identified in both blank gel voxels and tissue voxels, the abundance of proteins identified in blank voxels was significantly lower than that in tissue voxels (lower than 1%) (Figure S3H). These results further suggested that protein diffusion between voxels was nearly negligible. Moreover, voxels with fewer than 500 identified proteins were excluded from further analysis in our study (Methods). Collectively, by utilizing the PSERP procedure, we could obtain the panoramic spatial proteomic profiles covering whole pathological sections with deep coverage and high quantitative reproducibility, accurately illustrating the diverse proteomic features of malignant and nonmalignant regions within one tumor section. Spatial proteomic profiling of malignant and non-malignant regions in glioma To assess whether the PSERP method could capture spatial heterogeneity, we first conducted an unsupervised Louvain clustering analysis using the spatial proteomic data from the IDH1-WT/EGFR-mutant G1 sample (Methods). The results revealed six Louvain clusters among all voxels (Fig. [82]2A). These six clusters could be further partitioned into two major groups by applying hierarchical clustering (Fig. [83]2B and C). The principal component analysis (PCA) revealed clear separation between the malignant and non-malignant clusters (Fig. [84]2D). To explore the relationship between the clusters and the glioma sample’s spatial information, we mapped the clusters to their original tissue positions (Fig. [85]2E). Interestingly, when combined with H&E staining, the clusters corresponded precisely to the malignant and non-malignant regions of the sample. Specifically, cluster 2 (C2) and cluster 3 (C3) were identified as malignant clusters, while cluster 0 (C0), cluster 1 (C1), cluster 4 (C4), and cluster 5 (C5) were designated as non-malignant clusters (Fig. [86]2E). These results demonstrated the capability of identifying distinct spatial regions within a single sample section based solely on the spatial proteome. Fig. 2. [87]Fig. 2 [88]Open in a new tab Spatial proteomic dissection of malignant and non-malignant regions in IDH1-WT/EGFR-mutant gliomas using PSERP. A Uniform Manifold Approximation and Projection (UMAP) plots of spatial spots from the IDH1-WT/EGFR-mutant G1 sample, colored according to their Louvain clustering identities. B Phylogenetic tree illustrating proteomic similarities across clusters in the IDH1-WT/EGFR-mutant G1 sample. C UMAP plots of spots from the IDH1-WT/EGFR-mutant G1 sample, with colors representing malignant and non-malignant regions based on histological annotations. D Principal Component Analysis (PCA) plot comparing the proteomic profiles of malignant and non-malignant spots from the IDH1-WT/EGFR-mutant G1 sample. E H&E staining (left), spatial cluster distribution (middle), and spatial annotations of malignant versus non-malignant regions (right) for the IDH1-WT/EGFR-mutant G1 sample. F Volcano plot depicting differentially expressed proteins between malignant and non-malignant clusters from the IDH1-WT/EGFR-mutant G1 sample (FDR < 0.05). G Bar plots showing the enrichment pathways of upregulated proteins (FDR < 0.05, Log2 FC > 1.5) in malignant (left) and non-malignant (right) clusters from the IDH1-WT/EGFR-mutant G1 sample. H Heatmaps illustrating the spatial distribution of differentially expressed proteins between malignant and non-malignant regions in the IDH1-WT/EGFR-mutant G1 sample Further comparative analysis and enrichment analysis revealed that upregulated proteins in malignant clusters were enriched in cancer hallmark pathways, including MYC targets, MTORC1 signaling, E2 F targets, and mitotic spindle pathways (Fig. [89]2F-G, Table S1). In contrast, proteins upregulated in non-malignant clusters were enriched in coagulation, adipogenesis, and metabolism pathways (Fig. [90]2F-G, Table S1). In addition, spatial differential expression analysis revealed that glioma malignancy-related proteins, such as VIM, GFAP, PARP1 and EGFR, were significantly upregulated in the malignant regions. Conversely, immune-associated proteins, such as C1QB, and adipogenesis-associated proteins, such as APOA2, APOA4, and APOC2, were significantly upregulated in the non-malignant regions of the IDH1-WT/EGFR-mutant sample (Fig. [91]2H). Moreover, comparative and enrichment analyses validated the reproducibility of spatial expression profiles between corresponding voxels in G1-R1 and G1-R2 samples (Figure S3I and S3 J; Table S1). Specifically, proteins upregulated in the malignant region of replicates showed enrichment in the Myc target V1 and G2M checkpoint pathways (Figure S3 J). In contrast, proteins upregulated in the non-malignant region showed enrichment in coagulation and complement pathways (Figure S3 J). These findings demonstrated that the differential expression of proteins and pathways observed in the IDH1-WT/EGFR-mutant G1 sample was consistent and reproducible across replicates. Overall, the strong quantitative reproducibility across replicates underscored the robustness and reliability of our PSERP approach. To minimize the impact of interpatient heterogeneity on the diverse proteomic features of malignant and non-malignant regions identified by PSERP, we further analyzed the differences between malignant and non-malignant regions in three IDH1-WT/EGFR-mutant samples (G1, G4 and G5). The results of enrichment analysis and differentially expressed proteins were consistent across three distinct IDH1-WT/EGFR-mutant samples (Figure S4 A-B). We also performed paired comparisons between malignant and non-malignant clusters across the three IDH1-WT/EGFR-mutant samples (G1, G4, and G5 samples). As a result, the expression levels of tumor-related pathways, such as Myc targets, E2 F targets, and mitotic spindle, were increased in the malignant regions, while the expression levels of immune or metabolism-related pathways, such as coagulation, inflammatory response, and adipogenesis pathways, were increased in the non-malignant regions (Figure S4 C), indicating a consistent pattern of pathway alterations between malignant and non-malignant regions. The consistency of these findings across distinct IDH1-WT/EGFR-mutant samples highlighted the robustness of our PSERP method in capturing tumor heterogeneity, and further supporting the reproducibility of spatial proteomic variations in IDH1-WT/EGFR-mutant gliomas. High-resolution spatial proteomic heterogeneities in different regions of gliomas To evaluate whether our PSERP approach could comprehensively characterize the molecular diversity of distinct tumor regions at high resolution, we conducted a comparative analysis of the six clusters in the IDH1-WT/EGFR-mutant G1 sample (C0-C5) and performed pathway enrichment analysis on proteins that exhibited differential expression patterns within each cluster (Fig. [92]3A-3E and S5 A-C, Methods). The results revealed that immune-related pathways were significantly enriched in cluster C0, heme metabolism pathways were enriched in cluster C1, and clusters C2 and C3 exhibited similar patterns of pathway enrichment, primarily involving the G2M checkpoint and mRNA metabolic process pathways (Fig. [93]3B). Clusters C4 and C5 displayed enrichment in complement and coagulation cascades, complement activation pathways, and other related pathways (Fig. [94]3B). Fig. 3. [95]Fig. 3 [96]Open in a new tab Spatially resolved proteomic profiling of gliomas reveals intra-tumor heterogeneity. A H&E staining images and spatial proteomic cluster distribution in the IDH1-WT/EGFR-mutant G1 sample (left panel; scale bar, 1 mm). The details of each regions in the IDH1-WT/EGFR-mutant G1 sample (right panel; scale bar, 100 μm). B Dot plots illustrating the enrichment of pathways associated with significantly upregulated proteins across six spatial clusters in the IDH1-WT/EGFR-mutant G1 sample. C Heatmaps and boxplots illustrating significantly upregulated pathways in non-malignant clusters (C0, C1, C4, and C5) compared to malignant clusters (C2 and C3) in the IDH1-WT/EGFR-mutant G1 sample (adjusted p value < 0.05). D Heatmaps and boxplots showing significantly upregulated pathways in cluster C2 compared to cluster C3 in the IDH1-WT/EGFR-mutant G1 sample (adjusted p value < 0.05). E Heatmaps and boxplots showing significantly downregulated pathways in cluster C2 compared to cluster C3 in the IDH1-WT/EGFR-mutant G1 sample (adjusted p value < 0.05). F Heatmap demonstrating differentially expressed proteins related to key pathways between clusters C2 and C3 of the IDH1-WT/EGFR-mutant G1 sample (FDR < 0.05). G Heatmaps and boxplots illustrating significant differences in the immune score, immune cell types, and stromal score between clusters C3 and C2 in the IDH1-WT/EGFR-mutant G1 sample (FDR < 0.05) Importantly, by integrating pathological information from H&E images, we found that the spatial distributions of the six clusters exhibited significant diversity: clusters C2 and C3 were enriched in malignant regions, whereas clusters C0, C1, C4 and C5 were enriched in the non-malignant regions (Fig. [97]3A). Consistent with their regional distributions, we observed that more tumor-related pathways were significantly upregulated in malignant regions (clusters C2 and C3) than in non-malignant regions (clusters C0, C1, C4, and C5), whereas the tumor microenvironment and immune-related pathways were significantly upregulated in non-malignant regions (clusters C0, C1, C4, and C5) (Fig. [98]3B, C and S5 A). Although both clusters C2 and C3 were enriched in malignant tumor tissue regions, their separation into two distinct clusters suggested molecular heterogeneity between spatially different malignant tumor regions within the same sample. To further explore the heterogeneity between clusters C2 and C3, we compared the pathway ssGSEA scores of spots within these clusters to identify differentially enriched pathways (Figure S5B, Methods). The analysis revealed that Myc targets and G2M checkpoint pathways were significantly upregulated in cluster C2 (Fig. [99]3D), suggesting enhanced tumor cell proliferation in this cluster. This was further confirmed by the elevated multi-gene proliferation score (MGPS) in cluster C2 (Fig. [100]3D and F). In contrast, cluster C3 exhibited elevated expression of proteins involved in fatty acid metabolism, the interferon alpha response, and antigen processing and presentation pathways was detected (Fig. [101]3E and F). This result was consistent with the higher immune score and increased scores of immune cell types in cluster C3 (Fig. [102]3G, Methods). In parallel with the results of ssGSEA analysis, the upregulation of cell proliferation-associated molecules such as MRTOR4, CDK6, MCM5, MCM6, and AURKB was observed in cluster C2, while proteins related to fatty acid degradation (HADH, ACADVL, and ACAT2), the tricarboxylic acid (TCA) cycle (LDHA, PDHB, and ACO2), the interferon alpha response (PSMB8/9, PSME1/2, and STAT2), and antigen presentation (HLA-DQA1, DRA, and DRB1) were significantly upregulated in cluster C3 compared to those in cluster C2 (Fig. [103]3F, Methods). These results confirmed the spatial heterogeneity of glioma tumor regions, suggesting that cluster C2 might have a stronger proliferative capacity, whereas cluster C3 exhibited a more pronounced tumor immune response. In summary, our findings revealed glioma spatial heterogeneity through PSERP. By integrating the H&E pathology slides, we observed distinct histological characteristics within different clusters. The analysis of significantly upregulated proteins and pathway enrichment allowed us to identify molecular features associated with each cluster. Notably, the differences observed in the malignant and non-malignant regions on the H&E images and the pathway enrichment patterns among clusters were highly consistent. Furthermore, the heterogeneity between the two malignant tumor clusters, C2 and C3, was characterized by differences in proliferation- and immune-related pathways, suggesting the presence of distinct molecular subtypes within spatially distinct malignant tumor regions. These findings provided valuable insights into the spatial complexity and molecular characteristics of glioma, and contributed to a deeper understanding of this heterogeneous disease. Characterization of Tumor Border Characteristics and Interactions with the Tumor Microenvironment Using Spatial Proteomics Additionally, to illustrate the diverse proteomic features along the malignant-border-nonmalignant axis in the glioma sample, our high-resolution PSERP approach enabled a more precise exploration of unique protein expression patterns and microenvironmental features relative to the borders between malignant and non-malignant tumor regions. Based on the tumor borders identified by pathologists using the H&E-stained image of the IDH1-WT/EGFR-mutant sample, we divided all the voxels of the IDH1-WT/EGFR-mutant sample into three groups: tumor center, border, and paratumor (Fig. [104]4A, Methods). To assess the alignment between pathological-annotated tumor boundaries and those defined by PSERP data, we also applied Cottrazm (Construction of Tumor Transition Zone Microenvironment) [[105]18] using the spatial proteomic data to further delineate the malignant region, tumor border, and non-malignant region of the IDH1-WT/EGFR-mutant sample (Figure S5 C and S5D, Methods). We then compared the tumor border identified through H&E histological annotation with the border determined by Cottrazm. The results demonstrated a high degree of consistency between the tumor borders predicted by Cottrazm and those annotated by pathology (Figure S5E and S5 F). To understand the proteomic feature related to the tumor core spots, we calculated pathway enrichment scores based on spatial proteomic data of the IDH1-WT/EGFR-mutant G1 sample, and assessed their correlations with inferred copy number variation (CNV) scores. The results indicated that the hypoxia signaling pathway was the most significant correlated with inferred CNV scores (p-value = 1.6e-7, Figure S5G). Consistently, the hypoxia pathway scores increased at the tumor core within the Cottrazm-identified malignant regions and gradually decreased with the increasing distance from the tumor core (Figure S5H). Fig. 4. [106]Fig. 4 [107]Open in a new tab The spatial characteristics along the tumor border. A Strategy to quantify the Euclidean distance of spatial spots from the malignant tumor border. The red line delineates the malignant tumor border. B Heatmap illustrating the distance of each spot from the malignant tumor border. C Line graphs depicting distinct groups of proteins, clustered by Mfuzz, to show relative expression changes across the spatial proteomic landscape. D Enrichment analysis results of proteins within each Mfuzz cluster. E Variation in G2M checkpoint pathway scores of spatial spots with respect to the distance from the tumor region to the para-tumor region. F Variation in immune scores of spatial spots with respect to the distance from the tumor region to the para-tumor region. G Heatmap demonstrating the expression level changes of representative proteins significantly correlated with distance from the tumor border These findings prompted us to then compute the Euclidean distances from tumor and paratumor spots to border spots (Fig. [108]5A and B, Methods). We then applied the Mfuzz algorithm [[109]19] to identify different proteomic expression patterns associated with the distance from the border, and to explore potential crosstalk among the tumor, border and paratumor regions (Methods). This analysis revealed six distinct protein groups representing diverse distances from the border (Fig. [110]4C). Among these groups, group 2 represented proteins displaying specific higher expression levels at the tumor center region, and proteins enriched in group 4 exhibited the highest expression within the closest proximity to the border, implying that proteins in this group represent molecular features of tumor border regions. We subsequently performed gene set enrichment analysis (GSEA) on the proteins categorized within each group to characterize the proteomic features of the tumor center and tumor border (Fig. [111]4D). Specifically, proteins in group 2 were associated with cell cycle features, consistent with the elevated expression of G2M checkpoint pathway (Fig. [112]4E and G). In contrast, group 4 showed enrichment of proteins involved in coagulation, innate immune system and complement cascade pathways, suggesting a prominent role for immune-related processes in shaping expression dynamics at the tumor border (Fig. [113]4F and G). Based on these findings, we investigated immune cell infiltration in the IDH1-WT/EGFR-mutant sample using the xCell deconvolution algorithm (Methods). The results revealed a gradient of immune scores across the malignant and non-malignant regions of the tumor (Fig. [114]4F). As the distance from the border increased, the immune scores gradually decreased (Fig. [115]4F), suggesting the regulatory mechanisms in the border region contribute to an activated immune response. Furthermore, our analysis revealed that the expression levels of immune-related proteins such as C1QB, C3, IGHG1, IGLC2, F2, FGA, and FGB were upregulated with the increasing distance from the tumor region (Fig. [116]4G). These spatially dependent alterations in protein expression were shown on the spatial proteomic map of the IDH1-WT/EGFR-mutant sample (Figure S5 J). Fig. 5. [117]Fig. 5 [118]Open in a new tab Spatial heterogeneities across glioma tissues with distinct mutations (IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT). A H&E staining images, and proteomic cluster maps in different samples from glioma samples with distinct mutations: G1(IDH1-WT/EGFR-mutant), G2 (IDH1-mutant), and G3 (IDH1/EGFR-double-WT) samples. B Significantly upregulated marker proteins in the malignant regions compared to non-malignant regions for each sample (FDR < 0.05). C Boxplots and heatmaps showing ssGSEA scores of pathways upregulated in the malignant regions compared to non-malignant regions of the IDH1-mutant G2 sample. D Boxplots and heatmaps showing ssGSEA scores of pathways upregulated in the malignant regions compared to non-malignant regions of the IDH1/EGFR-double-WT G3 sample. E Heatmaps depicting significantly upregulated proteins in the malignant regions c non-malignant regions in the IDH1-mutant G2 sample (FDR < 0.05). F Heatmaps showing significantly upregulated proteins in the malignant region comparedompared to non-malignant regions in the IDH1/EGFR-double-WT G3 sample (FDR < 0.05). G Heatmaps and boxplots illustrating significantly downregulated xCell scores for immune cell types in the malignant regions compared to non-malignant regions of the IDH1-mutant G2 sample. H Heatmaps and boxplots illustrating significantly upregulated xCell scores for immune cell types in the malignant region compared to non-malignant regions of the IDH1/EGFR-double-WT G3 sample High-resolution spatial proteomic heterogeneities revealed intra- and inter-tumor heterogeneity in glioma To evaluate whether our PSERP approach could robustly reflect the diverse spatial proteomic heterogeneity of different samples, we conducted spatial proteomic profiling of glioma samples with distinct mutational statuses (IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT gliomas). We performed unsupervised Louvain clustering analysis for the IDH1-mutant G2 sample and EGFR & IDH1-WT G3 sample utilizing the same method as we used for the EGFR-mutant G1 sample (Methods). This analysis resulted in 7 clusters (G2 C0 to G2 C6) across 405 voxels of the IDH1-mutant G2 sample, and 6 clusters (G3 C0 to G3 C5) across 630 voxels of the IDH1/EGFR-double-WT G3 sample (Fig. [119]5A). Using the H&E image-based histological annotations of the G1, G2 and G3 samples, we found that in the IDH1-WT/EGFR-mutant G1 sample, clusters G1 C2 and G1 C3 were enriched in malignant regions. In the IDH1-mutant G2 sample, clusters G2 C1 and G2 C3 were enriched in malignant regions. In the IDH1/EGFR-double-WT G3 sample, clusters G3 C0, G3 C1, G3 C4, and G3 C5 were enriched in the malignant regions (Fig. [120]5A). These results indicated that the assigned clusters of the G1, G2, and G3 samples reflected the spatial diversity between malignant and non-malignant regions of gliomas. Previous proteogenomic studies have demonstrated that genomic alterations can influence downstream protein expression. For instance, IDH1 mutations can upregulate proteins related to the hypoxia and glutamate metabolism pathways [[121]20, [122]21]. In parallel, mutations in EGFR can increase the expression of its cognate protein and proteins genes involved in the TCA cycle [[123]22]. To explore the spatial distributions reflecting the distinctive or common proteomic features of gliomas with different mutational statuses, we examined the spatial expression patterns of proteins known to be altered, such as GFAP, EGFR, PARP1, and GLUD1 [[124]23–[125]26], among IDH1-WT/EGFR-mutant, IDH1-mutant and IDH1/EGFR-double-WT samples. Specifically, we observed significant upregulation of key proteins in the malignant regions across distinct samples (Fig. [126]5B), which could reflect the underlying mutation statuses and the tumor’s biological characteristics. A significant upregulation of EGFR was detected in the malignant regions of IDH1-WT/EGFR-mutant samples (G1, G4, and G5), consistent with the well-established role of EGFR mutations in glioma pathogenesis [[127]27] (Fig. [128]5B and Figure S6 A). In IDH1-mutant gliomas (G2, G6, and G7), we observed a significant upregulation of GLUD1 in the malignant regions (Fig. [129]5B and Figure S6B). GLUD1, a key enzyme in the glutamate-glutamine cycle, plays a crucial role in maintaining cellular metabolic homeostasis. Its upregulation in IDH1-mutant gliomas suggested a compensatory mechanism to maintain metabolic flux, further supporting the metabolic reprogramming in these tumors [[130]28, [131]29]. PARP1, a DNA repair enzyme involved in repairing single-strand DNA breaks, was significantly upregulated in the malignant regions of both IDH1-WT/EGFR-mutant gliomas (G1, G4, and G5) and IDH1/EGFR-double-WT gliomas (G3, G8, and G9) (Fig. [132]5B, Figure S6 A and Figure S6 C). The enhanced expression of PARP1 in these regions reflected the increased DNA damage and the reliance of tumor cells on DNA repair mechanisms to sustain their uncontrolled proliferation and survival [[133]30]. These findings suggested a possible vulnerability in these gliomas, which could be targeted for therapeutic intervention. Additionally, GFAP, a key protein of astrocytic differentiation [[134]31], was upregulated in the malignant regions of all three glioma mutation statuses, including IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT gliomas (Fig. [135]5B and Figure S6 A -S6 C). The increase in GFAP expression across all mutation statuses indicated its importance as a general marker of malignancy in gliomas, regardless of their mutational profiles. The upregulation of key proteins, such as EGFR, GLUD1, PARP1, and GFAP reflected critical aspects of glioma progression and tumor malignancy, including cell proliferation, metabolic reprogramming, DNA repair, and reactive gliosis. In addition, we conducted pathway enrichment analysis to more comprehensively depict inter-tumor heterogeneity across gliomas with different mutations (Table S4). We compared the differential expression of proteins between the malignant and non-malignant regions of the glioma samples characterized by distinct mutational profile. Specifically, we found that oxidative stress-induced senescence and glutathione catabolic process pathways were upregulated in the malignant regions of the IDH1-mutant samples (G2, G6, and G7) (Fig. [136]5C and Figure S7 A-B). In contrast, coagulation, interferon α response, and interferon γ response pathways were upregulated in the malignant regions of the IDH1/EGFR-double-WT samples (G3, G8, and G9) (Fig. [137]5D and Figure S7 C-D). Consistently, the expression levels of proteins, such as H2BC5, H2BC1, MINK1, H2 AC4, MAPK8, H2 AX, and CEACAM8, were significantly higher in the malignant regions compared with the non-malignant regions of the IDH1-mutant samples (G2, G6, and G7) (Fig. [138]5E and Figure S7E). Immune-related proteins, including CD9, ANXA1, C3, C9, HLA-A, HLA-B and HLA-C, were upregulated in the malignant regions of IDH1/EGFR-double-WT samples (G3, G8, and G9) (Fig. [139]5F and Figure S7 F). Furthermore, the xCell deconvolution results (Methods) revealed that the spatial distribution of immune cells between the non-malignant and malignant regions was distinct in IDH1/EGFR-double-WT samples compared to IDH1-mutant samples. Specifically, immune cells, including M2 macrophages, cDC, iDC, and CD8 + T effector memory cells, were significantly upregulated in the malignant regions of IDH1/EGFR-double-WT samples (G3, G8 and G9) (Fig. [140]5G and Figure S8 A). In contrast, the immune score and scores for immune cells, including B cell and plasma cells, were significantly downregulated in the malignant regions of the IDH1-mutant samples (G2, G6 and G7) (Fig. [141]5H and Figure S8B), consistent with the inhibitory effect of IDH1 mutations on immune infiltration [[142]32]. Given that PSERP approach can capture the proteomic characteristics of whole tissue sections without bias, we systematically compared the molecular features of both malignant and non-malignant regions in glioma samples with different genomic mutations. These results demonstrated that our PSERP approach accurately reflected the distinctive features of both intra- and inter-tumor heterogeneity in glioma samples. This approach also enabled us to analyze how different mutations influence the tumor and its surrounding microenvironment. Spatial phosphoproteomic profiling of gliomas revealed spatial heterogeneity of kinase-substrate signal transduction Besides elucidating the spatial proteomic features, our PSERP can also portray the spatial distributions of phosphoproteomic features (Methods). Specifically, based on phosphoproteomic data from our previously published article [[143]17], we built a phospho-spectral library using the FragPipe computational platform [[144]33]. The raw DIA data generated in our research by PSERP were then searched against the phospho-spectral library. The results revealed an average of 1,177 phosphosites in the IDH1-WT/EGFR-mutant sample, 1,447 phosphosites in the IDH1-mutant sample, and 1,310 phosphosites in the IDH1/EGFR-double-WT sample (Figure S8 C-S8H). Intriguingly, compared to the non-malignant regions, more phosphosites were identified in the malignant regions (Figure S8 C-S8H). To illustrate the most active kinases in the malignant regions among the three glioma samples, we conducted the KSEA analysis. The results revealed that, in the sample with EGFR mutation, the most significantly activated kinase in malignant regions was MAPK7 (Figure S8I and S8 J). Previously, we demonstrated that MAPK7 could promote IDH1-WT/EGFR-mutant gliomas’ tumor growth through phosphorylating enzymes (PRPS1, PRPS2) that catalyzed the phosphoribosylation of ribose 5-phosphate to 5-phosphoribosyl-1-pyrophosphate [[145]17]. We then evaluated the spatial distribution of phospho-substrates of MAPK7 and observed that phospho-substrates of MAPK7, including PRPS1_T255 and PRPS2_S41, which were involved in the nucleotide metabolism pathway, showed consistent expression patterns with the kinase activity of MAPK7 (Figure S8 K and S8L). Intriguingly, apart from MAPK7, CDK2 was also observed to be activated in the malignant regions of the IDH1-WT/EGFR-mutant sample. Importantly, we found that although both MAPK7 and CDK2 were activated in the malignant regions, they exhibited spatial heterogeneity in kinase activity and protein expression. Specifically, CDK2 was activated in the center of malignant regions, while MAPK7 was activated in the peritumoral regions. Along with these findings, the phospho-substrates of CDK2, including XRCC6_T455 and CDK7_T170, showed enhanced expression in the malignant regions, specifically in the tumor centroid of IDH1-WT/EGFR-mutant sample (Figure S8M and S8 N). Furthermore, the pathway enrichment analysis indicated that the tumor center exhibited heightened activity in CDK-related G2M cell cycle pathways, while the boundary regions showed increased activation of EGFR-MAPK-related pathways (Figure S8O and S8P). For the sample with IDH1 mutation, the GSK3B kinase showed enhanced activity in the malignant regions compared to the non-malignant regions. Accordingly, the spatial kinase activity of GSK3B was also observed to show elevation in the malignant regions (Figure S9 A and S9B). To elucidate the phospho-signaling pathways activated by GSK3B, we investigated the spatial expression of its phospho-substrates. As a result, the phospho-substrate that presented the most significant consistent spatial expression patterns with GSK3B was HIF-1A_T554 (Figure S9 C). Consistently, the hypoxia pathway was also observed to be activated in the malignant regions of the IDH1 mutant sample at the spatial proteomic level (Figure S9D). Thus, spatial phosphoproteomic data generated by PSERP revealed a mechanism through which the activation of the GSK3B-HIF-1 A phosphorylation signaling pathway could lead to the hypoxia features in the malignant regions of the IDH1-mutant sample. For the IDH1/EGFR-double-WT sample, we observed that the most significantly activated kinase in the malignant regions was PDGFRA (Figure S9E and S9 F). The activity of PDGFRA was also elevated in the malignant regions of the sample (Figure S9 F). Intriguingly, the phospho-substrates of PDGFRA, such as MUC1_Y1218 and ABL2_Y272, exhibited spatial expression patterns consistent with that of PDGFRA, and were enriched in the angiogenesis pathway (Figure S9G and S9H). Additionally, by calculating the micro-vessel density (MVD) score, we observed significantly higher MVD score in the malignant regions of the IDH1/EGFR-double-WT sample compared to the malignant regions of the IDH1-WT/EGFR-mutant and IDH1-mutant samples (Figure S9I), further confirming the angiogenesis characteristics of the malignant regions in the IDH1/EGFR-double-WT sample. In conclusion, our data revealed the concordance between spatial phosphoproteomic data and proteomic data in portraying the specific features of diverse tumor regions. These results underscored the ability of our PSERP method to systematically elucidate the spatial heterogeneity of tumors. Profiling of the glioma ecosystem reveals region-specific cellular composition and cell‒cell communications Previous single-cell transcriptomic studies of gliomas have provided insight into the intra-tumoral heterogeneity and dynamic cellular plasticity in gliomas [[146]34–[147]36]. Combining spatial omic data with single- nuclei transcriptomic data could offer a more comprehensive understanding of the glioma ecosystem. However, previous spatial proteomic studies were limited by insufficient protein coverage or detection throughput, which hindered their interactive analysis with single-cell transcriptomic data. Using our PSERP approach, which can provide in-depth proteomic profiling, we analyzed the multicellular ecosystems in different glioma samples. Specifically, we performed single-nuclei transcriptomic analysis on the same glioma samples (the IDH1-WT/EGFR-mutant sample, the IDH1-mutant sample, and the IDH1/EGFR-double-WT sample) used in our spatial proteomic analysis. After performing quality control (Methods), we profiled a total of 31,872 cells from three glioma specimens with different mutational statuses (Fig. [148]6A-B), with an average of 6,781 genes detected per cell. Fig. 6. [149]Fig. 6 [150]Open in a new tab The integrative analysis of spatial tumor ecosystem. A UMAP plot of all cells from the IDH1-WT/EGFR-mutant G1 sample, IDH1-mutant G2 sample, and IDH1/EGFR-double-WT G3 sample, colored by their sample source. B UMAP plot of major cell clusters. These plot were colored based on deconvoluted cell types using the sc-RNAseq data. C The compositions of diverse malignant cell types (upper) and non-malignant immune cell types (lower) in the IDH1-WT/EGFR-mutant G1 sample, IDH1-mutant G2 sample, and IDH1/EGFR-double-WT G3 sample, respectively. D, F, and H Heatmaps showing proteomic cluster maps of the IDH1-WT/EGFR-mutant G1 sample (D), IDH1-mutant G2 sample (F), and EGFR & IDHI-WT G3 sample (H). Spots was colored by their clusters. E, G, and I Heatmaps showing spatial distribution maps of cell types in the IDH1-WT/EGFR-mutant G1 sample (E), IDH1-mutant G2 sample (G), and IDH1/EGFR-double-WT G3 sample (I), colored by their cell types. J Heatmaps showing the spatial distribution of the levels of EGFR (left) and CS1RF (right) in the IDH1-WT/EGFR-mutant G1 sample. K Heatmaps showing the spatial distribution of the levels of HIF-1 A (left) and NeuN (right) in the IDH1-mutant G2 sample. L Heatmaps showing the spatial distribution of the levels of PDGFRA (left) and MPO (right) in the IDH1/EGFR-double-WT G3 sample. M, O, and Q The cell-cell interactions among cell types indicated by the scRNA-seq data from the IDH1-WT/EGFR-mutant sample (M), IDH1-mutant sample (O) and EGFR & IDH1-mutant sample (Q), respectively. The line thickness represented the number of significant ligand-receptor pairs. N, P, and Q Dot plots indicating the correlations between ligands and receptors in the IDH1-WT/EGFR-mutant G1 sample (N), the IDH1-mutant G2 sample (P), and the IDH1/EGFR-double-WT G3 sample (Q). Cell types enriched in malignant regions are labeled in red text, while those enriched in non-malignant regions are labeled in blue text. Ligands and receptors upregulated in cell types enriched in malignant regions are labeled in red text, while those upregulated in cell types enriched in non-malignant regions are labeled in blue text To explore tumor cellular composition, we performed UMAP analysis and classified cells from each sample (Methods). This analysis revealed substantial intratumor heterogeneity, identifying 13 clusters in the IDH1-WT/EGFR-mutant sample, 7 clusters in the IDH1-mutant sample, and 14 clusters in the IDH1/EGFR-double-WT sample. We also identified cluster-specific marker genes through differential gene expression analysis, which helped define the identity of each cell cluster (Fig. [151]6C, Methods). Comparative analysis revealed distinct malignant and immune cell compositions across samples. In the IDH1-WT/EGFR-mutant sample, the predominant malignant cells were AC-like (astrocytic-like), while the major non-malignant cells included tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), CD8 + T cells, and microglia. In the IDH1-mutant sample, MES-like (mesenchymal-like) cells predominated, with smaller proportions of NPC-like and OPC-like (oligodendrocytic precursor) cells. The major non-malignant cells were astrocytes, oligodendrocytes, and neurons. In the IDH1/EGFR-double-WT sample, the major malignant cells were OPC-like, and the major non-malignant cells were neutrophils, endothelial cells, and CD4 + T cells. To characterize the spatial cellular composition of each sample, we integrated the spatial proteomic data with single-nuclei transcriptomic data using the Bulk2Space algorithm (Methods). The results revealed distinct cellular compositions across different spatial clusters within each sample. Specifically, in the IDH1-WT/EGFR-mutant sample, we observed significant enrichment of malignant cell types, such as AC-like cells, in the malignant regions, while non-malignant cell types, such as TAMs, were predominantly enriched in the non-malignant regions (Fig. [152]6D and E). In the IDH1-mutant sample, MES-like cells were enriched in the malignant regions, while neurons were enriched in the non-malignant regions (Fig. [153]6F and G). In the IDH1/EGFR-double-WT sample, OPC-like cells were enriched in the malignant regions, while neutrophils were more abundant in the non-malignant regions (Fig. [154]6H and I). These findings suggested different cell type distribution patterns between malignant and non-malignant regions were observed across IDH1-WT/EGFR-mutant, IDH1-mutant and IDH1/EGFR-double-WT samples. We next portrayed the spatial distribution of cell type markers within each sample. In the IDH1-WT/EGFR-mutant sample, we observed elevated expression of the AC-like marker EGFR in the malignant regions, while the non-malignant regions showed enhanced expression of CSF1R (Fig. [155]6J). In the IDH1-mutant sample, we observed dominant expression of the MES-like marker HIF-1A in the malignant regions, whereas the neuronal marker NeuN was enriched in the non-malignant regions (Fig. [156]6K). In the IDH1/EGFR-double-WT sample, the OPC-like marker PDGFRA was predominantly expressed in the malignant regions, while the neutrophil marker MPO showed significantly higher expression in the non-malignant regions (Fig. [157]6L). To explore the cellular relationships between malignant and non-malignant cell types, we analyzed cell–cell interactions by examining ligand-receptor pairs using CellPhoneDB [[158]37] based on single-cell transcriptomic data (Methods). Notably, further analysis revealed key ligand-receptor interactions between malignant and non-malignant cells across samples with different mutational statuses. Specifically, in the IDH1-WT/EGFR-mutant sample, AC-like tumor cells were observed to interact with non-tumor cells, including CAFs, through ligand-receptor pairs such as PLG-ITGB2 and PLAU-ITGB2 (Fig. [159]6M and 6 N). AC-like tumor cells also interacted with TAMs via ligand-receptor pairs C3-CD74, C3-CD81, and EGF-EGFR (Fig. [160]6O and P). In the IDH1-mutant sample, MES-like tumor cells were found to interact with non-tumor cells, including oligodendrocytes, through ligand-receptor pairs like ANXA1-FPR1 A and PON2-HTR2 A (Fig. [161]6Q and R). In the IDH1/EGFR-double-WT sample, OPC-like tumor cells interacted with non-tumor cells, including endothelial cells, through ligand-receptor pairs VEGFA-PDGFRA and PDGFA-PDGFRA (Fig. [162]6S and T). Spatial integrative analysis revealed the impact of tumor microenvironment on glioma tumorigenesis Building on these findings, we sought to elucidate whether the spatial distribution of distinct cell types in the tumor microenvironments impacted tumor growth. First, we performed survival analysis using our previously published glioma multi-omic dataset [[163]17]. The results revealed the enrichment of TAMs was significantly correlated to glioma patient survival (Figure S10 A), suggesting that higher TAM infiltration contributes to more aggressive gliomas. In the IDH1-WT/EGFR-mutant sample, combining PSERP-based spatial proteomic data with single-nuclei transcriptomic data, we found that TAMs were dominantly enriched in the tumor border regions of the IDH1-WT/EGFR-mutant sample (Figure S10B and S9 C). The TAM-associated protein, CSF1R, was observed to have elevated expression in the tumor border regions of the IDH1-WT/EGFR-mutant sample (Figure S10D). We also conducted multiplexed IHC (mIHC) staining for CSF1R + TAMs (TAM marker: CSF1R) and EGFR + tumor cells (AC-like tumor cell marker: EGFR). As a result, significant enrichments of CSF1R + TAMs were observed in non-malignant regions, specifically in the tumor border regions of the IDH1-WT/EGFR-mutant sample (Figure S10E). Further analysis focused on how TAM influenced the tumor cell growth. To this end, we screened for TAM-secreted factors potentially associated with glioma malignant growth using single-nuclei transcriptomic data. A total of 281 transcripts were identified as specifically upregulated in TAMs, among which EGF was most significantly upregulated in TAMs (Figure S10 F-G). We then applied survival analysis using our glioma multi-omic data. The protein expression of EGF showed the most significantly negative correlation with patients’ overall survival (Figure S10H). These data suggested that EGF might be secreted by TAMs and that its expression was associated with poor survival in gliomas. In accordance with this finding, cell–cell interaction analysis also confirmed a positive bidirectional interaction between TAMs and AC-like glioma tumor cells through the EGF-EGFR ligand-receptor pair (Figure S10I). We then examined the spatial distributions of TAMs and AC-like glioma tumor cells using the Bulk2Space algorithm. The analysis revealed that the malignant tumor region near the tumor border were predominantly enriched with AC-like glioma tumor cells (Figure S10 J and S10 K). Furthermore, we investigated how EGF-EGFR signaling promoted glioma cell growth. In previous work, we demonstrated that EGFR could promote glioma tumor cell growth by activating MAPK7-mediated phosphorylation signaling pathway [[164]17]. We found that both EGFR and MAPK7 were enriched in the malignant tumor regions (Figure S10L and S10M). Correlation analysis revealed a positive correlation between the spatial expression levels of EGFR and MAPK7 (Figure S10 N). To further validate the impact of EGF-EGFR signaling on glioma tumor cell growth, we constructed EGFR overexpressed glioma tumor cells (EGFR-OE-U87). These cells were stimulated with EGF or left untreated as a control, and tumor growth was assessed using a CCK-8 assay. The results demonstrated that EGF stimulation significantly increased tumor cell growth in EGFR-OE-U87 cells (Figure S10O). These findings confirmed the role of EGF-EGFR signaling in promoting glioma tumor cell growth. In conclusion, the distribution of TAMs in tumor border regions of glioma may promote tumor cell growth through the EGF-EGFR signaling pathway. In addition, we also investigated the influence of the tumor microenvironment on glioma tumorigenesis in both the IDH1-mutant and IDH1/EGFR-double-WT samples. In the IDH1-mutant sample, the application of the Bulk2Space algorithm revealed a significant enrichment of neuronal cells in the non-malignant regions (Figure S11 A and S11B). To explore the impact of neuronal cell enrichment on the tumor microenvironment, we analyzed the spatial expression of neuron-specific proteins and found that the spatial expression of dopamine receptor DRD1 showed the most significantly positive correlation with the spatial distribution of neurons (Figure S11 C and S11D). Previous research on dopamine receptors has shown that their high expression can inhibit NLRP3, thereby reducing inflammation [[165]38]. We then examined the enrichment of inflammasomes in these regions and found that areas with higher neuronal enrichment had relatively lower inflammasome enrichment (Figure S11E). Correlation analysis revealed a negative correlation between inflammasome enrichment and DRD1 expression, suggesting that the elevated neuronal enrichment in this region might reduce inflammation by modulating NLRP3 (Figure S11 F). To further confirm the elevated expression of DRD1 + neuron cells in the non-malignant regions and validate the impact of DRD1 on inflammasome, we employed multiplexed IHC (mIHC) staining for DRD1 + neuronal cells, IDH1^R132H + tumor cells, and NLRP3 + inflammasome activated cells. The results confirmed significant enrichment of DRD1 + neuron cells in non-malignant regions, accompanied by a reduction in inflammasome-positive cells (Figure S11G). These findings indicated that the enrichment of dopaminergic-like neurons in the non-malignant regions was associated with reduced inflammasome activation. In the IDH1/EGFR-double-WT sample, neutrophils were the most abundant cell type in the non-malignant regions based on the single-nuclei transcriptomic data (Figure S11H). Consistent with this observation, PSERP-based spatial proteomic data also revealed elevated expression of neutrophil marker MPO in the non-malignant regions of the IDH1/EGFR-double-WT sample (Figure S11I). To explore the biological functions associated with the enrichment of neutrophils in the non-malignant regions, we identified pathways correlated with the spatial distribution of neutrophils. The results revealed that the neutrophil degranulation pathway had the strongest positive correlation with neutrophil enrichment, suggesting substantial activation of this pathway in the regions with high neutrophil content (Figure S11 K and S11L). Building on this finding, we screened for neutrophil-specific molecules and found that the spatial distribution of proteinase 3 (PR3) was strongly associated with neutrophil degranulation (Figure S11M and S11 N). This is consistent with previous reports indicating that PR3, a key enzyme on the neutrophil surface, plays a crucial role in modulating chemotactic migration and culminating in a process known as NETosis [[166]39]. Furthermore, we examined the spatial enrichment of the NETosis pathway and found it to be significantly upregulated in the neutrophil-enriched regions (Figure S11O). The significant positive correlation between PR3 expression and NETosis further highlighted the role of PR3 in neutrophil degranulation and NETosis in the non-malignant regions of the IDH1/EGFR-double-WT sample (Figure S11P). Prospective cohort analysis confirmed the effectiveness of PSERP To further demonstrate that our PSERP-based spatial proteomic methods could effectively decipher the tumor spatial heterogeneity, we collected additional FFPE samples of 20 glioma patients from Zhongshan Hospital, Fudan University (Methods). Samples were classified into two groups (10 IDH1-mutant, 10 IDH1-WT). No significant inter-group differences were observed in age distribution (median age: 50 years (IDH1-mutant group) vs. 52 years (IDH1-WT group); Wilcoxon rank-sum test, p = 0.75) and gender proportion (60% male in both groups). To uncover molecular heterogeneities of the tumor boundary between IDH1-mutant and IDH1-WT samples, we preformed both bulk proteomic analysis and spatial proteomic analysis on representative areas, including the tumor core, tumor boundary, non-malignant region near the tumor boundary and non-malignant region distal to the tumor. The detailed information of these samples were presented in Table S1. To be more specific, IDH1-mutant samples exhibited lower immune cell enrichment scores compared with IDH1-WT samples (Figure S12 A). The results also showed that NLRP3 and CXCR3 proteins exhibited reduced expression in IDH1-mutant samples (Figure S12 C). Conversely, IDH1-mutant samples showed higher neuron infiltration scores, with elevated expression of dopamine signaling pathway proteins (DRD1, DRD2, MAOA, MAOB) (Figure S12B-C). These results were consistent with our spatial proteomic observations, indicating that the immune microenvironment features identified via PSERP-based spatial proteomics may reflect a universal mechanism underlying IDH1 mutation-driven alterations in the tumor microenvironment. We then systematically characterized the spatial proteomic features of IDH1-mutant and IDH1-WT samples. In the malignant regions, IDH1-mutant protein was only detected in IDH1-mutant samples (Figure S12D-E). As for the non-malignant regions, consistent with the findings from the bulk proteomic analysis, the neuronal cell related protein, dopamine receptor DRD1, was significantly enriched in non-malignant regions of IDH1-mutant samples, especially near the tumor boundaries (Figure S12D-E). In contrast, the inflammasome-related protein, NLRP3, was predominantly localized in non-malignant regions adjacent to tumor boundaries in IDH1-WT samples (Figure S12D-E). Multiplexed IHC staining for DRD1 + neuron cells and IDH1^R132H + tumor cells confirmed pronounced enrichment of DRD1 + neurons in non-malignant regions (Figure S12 F-G). These results validated our hypothesis that the expanded population of DRD1 + neurons may establish an immunosuppressive niche in peritumoral regions through the suppression of NLRP3 inflammasome in IDH1-mutant gliomas. Specifically, dopamine signaling via DRD1 receptors inhibits NLRP3-mediated pyroptosis in myeloid cells. To investigate how neuronal cells at the tumor boundary impact tumor cell progression through neuron-tumor interactions, we performed further analysis and functional experiments. We analyzed neuron-secreted proteins and identified NLGN3 as the most significantly upregulated protein in non-malignant regions near tumor boundaries of IDH1-mutant gliomas (Figure S13 A-C). NLGN3 plays a important role in promoting glioma progression. In our study, correlation analysis revealed that NLGN3 expression was strongly associated with mTOR signaling activity (Figure S13D). Spatial mapping results further demonstrated the co-enrichment of NLGN3 and the mTORC1 complex at tumor boundaries (Figure S13E). Collectively, the spatial distribution of NLGN3 and its correlation with mTORC1 suggested its potential functional role in linking neuronal cells to glioma tumor cells, and implied that it might promote tumor cell progression via the mTOR signaling pathway. To validate this mechanism, IDH1-mutant PDCs were treated with NLGN3 (100 ng/mL) and/or the mTOR inhibitor rapamycin (20 nM). CCK-8 assays showed that NLGN3 significantly promoted PDC proliferation (p < 0.01), while rapamycin (m) completely abolished this effect (p = 0.92 vs. untreated controls, Figure S13 F-G). These results supported our assumption that elevated NLGN3 in the tumor boundary might enhance glioma tumor cell growth by activating the mTOR signaling pathway. Furthermore, these findings provided actionable insights into niche-specific therapeutic targeting for glioma in the future. Through the prospective cohort validation and functional experiments, we have demonstrated that our PSERP platform reliably resolves spatially organized proteomic patterns in gliomas, particularly the IDH1 mutation-dependent enrichment of neuronal signaling components (e.g., DRD1, NLGN3) at tumor boundaries. The identification of the NLGN3-mTORC1 axis as a driver of proliferation in IDH1-mutant gliomas not only validates PSERP’s analytical power but also reveals a targetable niche-specific mechanism. In summary, our PSERP approach provides comprehensive spatial proteomic coverage across the entire slide, enabling integrative analysis with single-cell transcriptomic data to elucidate the spatial distribution and interactions of distinct cell types. Using the PSERP approach, we elucidated both the cell type heterogeneity in gliomas with different mutational statuses and the role of spatially specific recruitment of cell types in promoting tumor cell communication and fostering a supportive microenvironment for tumor growth. Moreover, we observed several receptor-ligand interactions that primarily occurred at the protein level, highlighting the critical role of proteomic analysis in understanding cellular communication. Our PSERP-based spatial proteomic approach directly identified these interactions, enabling a more comprehensive understanding of biological pathways within the tumor microenvironment. By pinpointing the high expression of DRD1 and its correlation with inflammasome enrichment in the IDH1-mutant sample, as well as the observation of NETosis in the IDH1/EGFR-double-WT sample, we provided clear examples of how spatial proteomic data uncovers intricate molecular interactions within tumor microenvironments, which may not be fully captured by spatial transcriptomic data alone. Therefore, while spatial transcriptomics is a powerful tool for gene expression analysis, the PSERP-based spatial proteomic method provides a unique perspective on the functional molecular landscape of tissues, which is particularly valuable in understanding complex diseases with spatial heterogeneity. Spatial-resolved proteomics revealed the intra-tumor heterogeneity of tumor-specific mutational peptides Gene mutations can generate neoantigen candidates for personalized immunotherapy [[167]12, [168]13]. By integrating tumor-specific genomic events, our PSERP-based spatial proteomic approach can portray spatial distributions of tumor specific peptides at proteomic level. Specifically, in our study, we integrated tumor-specific genomic events from our previously published glioma multi-omics dataset, which included the glioma specimens utilized in this dataset [[169]17]. The tumor-specific genomic events for each tumor were translated into 1, 3, or 6 frames and used to build a glioma-specific spectral library. Based on this library, we searched for tumor-specific peptides using the raw LC–MS files of spatial proteomics. A total of 479 tumor-specific mutational peptides were identified (FDR < 0.1 at peptide level and protein level). The MHC-I binding affinities for all the detected tumor-specific mutational peptides were predicted using NetMHCpan v4.0, and 75 tumor-specific mutational peptides with a predicted binding affinity < 500 nM were identified (Fig. [170]7A; Methods). Fig. 7. [171]Fig. 7 [172]Open in a new tab The heterogeneity of mutated peptide identified by PRSEP. A The strategy of mutated peptide identification and quantification. B Mutant peptides whose distribution showed significant spatial-specificity in specific cluster of each sample. C, D Heatmaps (upper) showing spatial distribution of specific mutant peptides in the IDH1-WT/EGFR-mutant G1 sample (C) and IDH1-mutant G2 sample (D). Boxplots (bottom) showing differential expression levels among clusters in the IDH1-WT/EGFR-mutant G1 sample (C) and IDH1-mutant G2 sample (D), respectively. E, F IFNy ELISPOT output of PBMCs co-cultured with synthesized tumor-specific peptides identified in the IDH1-WT/EGFR-mutant G1 sample (E) and IDH1-mutant G2 sample (F). Scrambled peptides (control), tumor-specific peptides, or combined peptides were performed. G, H Line graphs showing progression rates of PBMCs co-cultured with synthesized tumor-specific peptides identified in the IDH1-WT/EGFR-mutant G1 sample (G) and IDH1-mutant G2 sample (H) by CCK-8 assay. Scrambled peptides (control), tumor-specific peptides, or combined peptides were performed. H Images of PDX tumors from mice treated with specific mutant peptide or control We then analyzed the spatial distribution of these 75 tumor-specific peptides, and focused on identifying those whose spatial distribution covered the maximal area of malignant regions in tumor samples. To this end, five tumor-specific peptides (CTNNA2_p. V401 A, EGFR_p. G598 VPTEN_p. K125E, SLC25 A5_p. T254M, and CD36_p. G326 V) were screened out for the IDH1-WT/EGFR-mutant sample, and five tumor-specific peptides (TP53_p. R248 W, IDH1_p. R132H, PML_p. P410L, EIF3 A_p. R700 K, and HDAC6_p. G518 V) were screened out for the IDH1-mutant sample (Fig. [173]7B-D). The combination of these selected peptides covered more than 95% of the malignant regions in their respective samples. T cells from healthy donors provide a rich source of T-cell repertoires that can specifically recognize neoantigens presented on tumors. To evaluate the immunogenicity of tumor-specific mutational peptides, we synthesized ten tumor-specific peptides (Methods), and evaluate T-cell responses by treating them with these candidate peptides, followed by assessment of the responses using an ELISpot assay (Methods). IFN-γ secretion was significantly increased when treated with the combination of peptides (Fig. [174]7E and F). Furthermore, an in vitro cytotoxicity assay (Methods) revealed that vaccines derived from the combination of peptides showed enhanced tumor cell growth inhibition efficacy (Fig. [175]7G and H). Based on these findings, we constructed a PDX mouse model via subcutaneous injection (Methods), to assess whether tumor-specific mutational peptides can activate T cells in vivo. Histological examination, proteomic profiling, and phosphoproteomic analysis demonstrated a high degree of biological similarity between the original tumors and the PDX tumors (Methods, Supplementary Data 1). A single type of tumor-specific peptide-derived vaccine or combination of peptide-derived vaccines was then intraperitoneally injected into the PDX mouse model (Methods). The results revealed that the combination of peptide-derived vaccines significantly delayed xenograft growth (Fig. [176]7I). In summary, our data highlighted that the PSERP approach could directly reveal the spatial expression profiles of tumor-specific mutational peptides (potential neoantigens) and their nearby microenvironments. This is important since current T-cell reactivity detection methods can only be applied to a limited set of neoantigens per tumor. Information on the spatial expression of neoantigens and tumor microenvironments can help us efficiently filter out low-expression or nonspecifically expressed peptides. Moreover, combinational targeting of tumor-specific mutational peptides is a trend in neoantigen-based treatments. Therefore, selecting appropriate neoantigen combinations and retaining highly confident and actionable candidates is crucial [[177]13]. Our approach can help identify tumor-specific mutational peptides with spatial expression specificity. Thus, we can select combinations of tumor-specific mutational peptides whose spatial expression covers the largest areas of tumor regions, providing insights into potential neoantigen combinations that could most efficiently target and kill tumor cells. Discussion Spatial omics is a rapidly developing research field that aims to enhance our understanding of health and disease within the spatial context of biological structures, such as organs, tissues, or tumors. The use of spatial technologies in disease research aids in understanding drug action, drug delivery, and immune system activities, while also providing critical location information [[178]1, [179]4]. Spatial transcriptomic studies have begun to derive diagnostic and prognostic value in several oncology contexts, demonstrating that spatial omic data can facilitate the pathological assessment of tumor tissue at the point of care [[180]40]. To date, spatial transcriptomic techniques have made significant progress, and several landmark methods, such as Stereo-seq [[181]41], Seq-Scope [[182]42], and ZipSeq [[183]43], have been employed to perform high-resolution spatial analysis across entire tissue sections. Despite the extraordinary achievements of spatial transcriptomics, it is important to note that proteins, rather than DNA or RNA, govern most cellular processes and execute biological functions. To accurately elucidate tissue heterogeneity and the underlying molecular mechanism, spatial proteomics is critical. To assess the advantages and limitations of various spatial proteomic methods, we compared PSERP with widely used techniques, including CODEX [[184]44], LCM [[185]45], and ProteomEX [[186]46]. CODEX, a multiplexed imaging technique, enables high-resolution analysis of tissue architecture by simultaneously detecting multiple proteins. However, it requires advanced equipment, is limited by available antibodies and markers, and has a restricted scanning range. LCM allows for the precise microdissection of specific tissue regions for proteomic analysis, but it is labor-intensive and requires significant histological expertise. ProteomEx combines tissue expansion with manual microdissection, but it cannot provide comprehensive spatial proteomic profiling across the entire tissue, potentially overlooking gradual molecular changes. Additionally, its manual approach may lead to reproducibility issues. In summary, although current spatial proteomic techniques have made significant progress, they still face limitations in terms of protein coverage and throughput (Supplementary Data 2). Under these circumstances, we developed PSERP, a matrix-based spatial proteomic technique. PSERP enables rapid, quantitative profiling of proteomic spatial variability across whole tissue sections with submillimeter resolution. Its key advantage lies in its ability to capture protein distributions without bias, offering flexible field-of-view selection and comprehensive protein coverage. PSERP also offers significant time savings by automating much of the process through robotic workstations. With standardized protocols, consistent quality control, and quantification metrics, PSERP ensures reproducible results across different laboratories and large-scale studies, enhancing its reliability for research applications. As a result, PSERP can effectively capture dynamic alterations in protein expression across entire tissue sections. Moreover, its in-depth protein coverage and flexible capturing capabilities facilitate subsequent integrative analyses with single-cell transcriptomic data or other spatial omic data. Gliomas exhibit complex intratumoral and intertumoral heterogeneity, making them an ideal model for a proof-of-principle study of PSERP. The selection of glioma samples was based on several scientific and clinical factors. The WHO 2021 classification integrates genetic mutations into glioma diagnosis, prompting us to focus on common mutations such as EGFR and IDH1, which are frequently observed in gliomas [[187]47–[188]49]. More importantly, EGFR and IDH1 mutations have been confirmed to be associated with patient’s prognosis, with EGFR mutations leading to poor clinical outcomes and IDH1 mutations associated with improved clinical outcomes [[189]17]. In addition to mutational frequencies, IDH1 and EGFR mutations have been shown to have distinct impacts on immune microenvironments. Specifically, compared to IDH1-WT/EGFR-mutant samples, IDH1-mutant samples displayed molecular characteristics indicative of an immunologically “desert-like” microenvironment [[190]17]. Additionally, patients with IDH1 mutations can be classified into those with and without1p/19q co-deletion, according to the WHO 2021 guidelines. Our preliminary research on IDH1 mutations included both samples with and without 1p/19q co-deletion [[191]17]. Comparative analysis revealed that IDH1-mutant samples, regardless of 1p/19q co-deletion status, exhibited a “desert-like” microenvironment in contrast to IDH1-WT samples, in contrast to IDH1-WT samples [[192]17]. Based on these findings, for this study, we specifically selected IDH1-mutant gliomas without 1p/19q co-deletion for further analysis. For this proof-of-principle study, we analyzed IDH1-mutant and IDH1-WT (IDH1-WT/EGFR-mutant and IDH1/EGFR-double-WT) glioma samples to analyze tumor spatial heterogeneity using PSERP-based spatial proteomic data. In this study, we used the PSERP approach to profile the spatial proteome of gliomas with varying mutational statuses. In addition to characterizing the distinct features of malignant and non-malignant tumor regions, we also illustrated the dynamic expression patterns of proteins from the tumor center to the border and paratumoral regions, a feature predicted by computational methods [[193]50]. The diverse proteomic patterns across the core tumor, tumor border, and paratumoral regions provide valuable clinical insights for identifying potential therapeutic targets in gliomas. Notably, the strong immune infiltration observed at the tumor border enhances our understanding of the mechanisms driving glioma metastasis and may inform the development of more effective therapeutic strategies. While previous single-cell transcriptome studies have highlighted the cellular heterogeneity of gliomas [[194]51], the spatial resolution of cellular interactions remains poorly understood. By integrating analysis of single-cell transcriptome and spatial proteome data, we not only depicted the spatial diversity of cellular distribution in gliomas with different genetic mutations but also demonstrated how specific cell types reshape the tumor ecosystem through cell–cell communication at spatial resolution. For instance, OPC-like glioma cells, characterized by elevated PDGFRA expression, were found to recruit endothelial cells and promote angiogenesis. This observation aligned with our previous findings that gliomas with increased expression of PDGFRA were characterized by angiogenesis [[195]52]. Additionally, we applied our PSERP-based spatial proteomic analysis tool to profile both proteomic and phosphoproteomic data, revealing distinct kinase activation patterns across samples. These findings underscored the robustness of our spatial proteomic and phosphoproteomic data in capturing the molecular features of different glioma regions, highlighting the potential of PSERP to elucidate tumor spatial heterogeneity. In addition, the brain is a complex organ with distinct regional functional characteristics that may influence tumor behavior. To explore whether the spatial proteomic patterns can reflect the characteristic distribution of tumors in the brain, we reviewed the clinical records of patients whose tumor samples were included in this study. The three IDH1-WT/EGFR-mutant samples and three IDH1-mutant samples in our study were derived from distinct brain regions. Specifically, for the IDH1-WT/EGFR-mutant samples (G1, G4 and G5), G1 sample originated from the temporal lobe, while G4 and G5 samples came from the frontal lobe. For the IDH1-mutant samples (G2, G6 and G7), G2 and G6 samples originated from the frontal lobe, while G7 sample came from the parietal lobe. We compared tumor characteristics among samples with the same mutation features in different brain regions. For IDH1-mutant tumors, those in the frontal lobe showed significant activation of the Notch signaling pathway, while tumors in the parietal lobe exhibited significant activation of the HIF-1 signaling pathway (Figure S14 A). Consistent with this finding, proteins such as NOTCH2, NOTCH1, and SLC4 A4 were elevated in the frontal lobe, while HIF-1 A, TP53, and PDK1 were elevated in the parietal lobe (Figure S14B). For IDH1-WT/EGFR-mutant tumors, those in the frontal lobe exhibited significant activation of the TGF signaling pathway, while tumors in the temporal lobe showed significant activation of the IGF pathway (Figure S14 C). Proteins such as SMAD3, SMAD4, and TGFβ were elevated in the frontal lobe, while IGF1R, IGF1, and IGF2R were elevated in the temporal lobe (Figure S14D). The differences in molecular characteristics were also observed in our previous glioma cohort studies (Figure S14E-F). These results indicated that tumors in different brain regions may exhibit distinct molecular characteristics. Furthermore, our methods enhance the understanding of glioma infiltration in the brain, potentially aiding in the precise surgical resection of tumors in a clinical context. Future research should focus on expanding the range of brain tumor regions studied and increasing the sample size for more comprehensive comparisons. Importantly, our PSERP includes a spatially resolved workflow for tumor-specific peptidome identification. Advanced neoantigen prediction algorithms can identify millions of potential mutation-derived neoantigens. For example, the TSNAdb database compiles neoantigens predicted by NetMHCpan from somatic mutations in The Cancer Genome Atlas (TCGA) tumor samples [[196]53]. Meanwhile, the GNIFdb database covers glioma-specific neoantigens across various subtypes [[197]54]. Despite these algorithmic advancements, most predicted neoantigens are still difficult to validate at the proteomic level, limiting their broader application in immunotherapy. To date, no neoantigen-directed therapy has been granted regulatory approval for use in cancer patients. Several reasons might explain this. One key reason is that early research on neoantigens primarily relied on genomic and computational approaches, which could not directly capture their expression patterns, tumor specificity, or spatial distributions. As a result, many predicted neoantigens are not stably expressed, compromising their therapeutic efficacy [[198]55]. Our PSERP workflow is a powerful tool with several advantages. First, it allows for the quantitative detection of tumor neoantigens. Second, it enables the spatial mapping of these neoantigens. This helps identify tumor-specific antigens expressed exclusively within the tumor region, thus minimizing potential damage to normal tissue during targeted therapy. Another reason is the diverse immune evasion mechanisms that tumors use to escape immune surveillance, which are closely linked to intratumoral heterogeneity [[199]56]. Thus, understanding the potential impact of neoantigens on the tumor microenvironment is crucial. Using our PSERP methods, we observed diverse spatial expression patterns of neoantigens. By combining this with the spatial distribution of immune cells, we were able to predict the impact of neoantigens on tumor immune environments, providing valuable information for therapeutic neoantigen selection. Moreover, this method can help select tumor neoantigens that cover the tumor most extensively, improving the targeted therapeutic effect. To our knowledge, our PSERP provides the first spatial proteomic landscape of neoantigens. Therefore, these findings could have a significant impact on the clinical translation and therapeutic application of neoantigens. Furthermore, our method can be integrated with the Tandem Mass Tag (TMT) multiplexing strategy, which enables the labeling of peptides extracted from each voxel. This integration has the potential to accelerate the detection process by allowing parallel detection of multiple samples. For example, when using 16-plex TMT (Tandem Mass Tag) for sample labeling, the detection time for 600 samples could be significantly shortened to 1–2 days. In future studies, by combining PSERP with TMT, we could enhance the throughput and efficiency of spatial proteomic profiling, reducing overall detection time without compromising data quality or resolution. Although we have highlighted many advantages of the PSERP method, there are still areas for improvement. While PSERP provides submillimeter spatial resolution, it may be less sensitive than methods like CODEX, particularly for detecting low-abundance proteins in very small or sparse tissue samples. This limitation could impact its ability to achieve single-cell or subcellular resolution in certain contexts, thus affecting its sensitivity in applications that demand ultra-high resolution. Furthermore, although PSERP minimizes the need for expensive specialized equipment compared to techniques like CODEX, it still requires access to mass spectrometry platforms and robotic workstations. These technical requirements may present a challenge for laboratories lacking such infrastructure, potentially limiting the broader adoption and application of PSERP in diverse research settings. In conclusion, we propose the PSERP approach, which integrates tissue expansion and matrix-based sample preparation with an optimized high-throughput mass spectrometry platform. The proposed framework offers an efficient and cost-effective approach for characterizing intra-tumoral cellular heterogeneity in both tumor and normal tissues. We demonstrated the value of our framework in exploring tumor ecosystems, making it possible to characterize the spatial heterogeneity present in cancer, which could inform the development of more precise and effective treatments for brain tumors and other solid tumors. Methods Specimens and clinical data Frozen primary tumor and germline blood samples from qualified patients were collected from Zhongshan Hospital. One 4 μm thick slide from each tissue sample was sectioned for hematoxylin and eosin (H&E) staining. Histopathologically defined adult glioma tumors were considered for analysis. Clinical data were obtained from Zhongshan Hospital and reviewed for correctness and completeness. The Research Ethics Committees of Zhongshan Hospital, Fudan University, approved this study (B2019-200R), and all patients provided written informed consent. Tissue expansion in PSERP method was performed following the previously published protocol [[200]15]. The 10 μm thick slides were deparaffinized with xylen and washed with ethanol for PSERP-based proteomic analysis. We used 10 μm thick slides from FFPE sample blocks for bulk proteomic analysis. We used 10 μm thick slides from FFPE sample blocks. The slides were macro-dissected, deparaffinized with xylene, and washed with ethanol. The extracted tissues were then lysed in a buffer comprising 0.1 M Tris–HCl (pH 8.0), 0.1 M DTT, and 4% SDS at 99 °C for 30 min. The crude extract was then clarified via centrifugation at 16,000 xg for 10 min, and the supernatant was loaded into a 10 kD Microcon filtration device (Millipore), centrifuged at 12,000 × g for 20 min, and then washed twice with Urea lysis buffer (8 M Urea, 100 mM Tris–HCl pH 8.0) and twice with 50 mM NH4HCO3. The samples were digested using trypsin at an enzyme to protein mass ratio of 1:25 overnight at 37 °C. Finally, the peptides were extracted and dried SpeedVac (Eppendorf). Cohort sample acquisition For cohort validation, glioma FFPE samples were obtained from Zhongshan Hospital, Fudan University. A total of 20 participants underwent surgical resection from January 2018 to December 2024. Clinical data, including gender, age, tumor grade, and IDH1 mutational status were obtained from Zhongshan Hospital and are summarized in Table S1. Sample processing for whole exome sequencing (WES) and RNA-seq Our study sampled a single site of the primary tumor from surgical resections due to the internal requirement to process a minimum of 125 mg of tumor tissue and 50 mg of adjacent normal tissue. DNA and RNA were extracted from normal tumor and blood specimens via a coisolation protocol using Qiagen QIAsymphony DNA Mini Kit and QIAsymphony RNA Kit. Genomic DNA was also isolated from peripheral blood (3–5 mL) to serve as a matched normal reference material. A Qubit™ dsDNA BR Assay Kit was used with a Qubit® 2.0 fluorometer to determine the concentration of dsDNA in the aqueous solution. Any sample that passed quality control and produced enough DNA to be subjected to various genomic assays was subjected to genomic characterization. RNA quality was quantified using a NanoDrop 8000, and quality was assessed using an Agilent Bioanalyzer. A sample that passed RNA quality control and had a minimum RNA integrity number (RIN) of 7 was subjected to RNA sequencing. Identity matches for germline tissue, normal adjacent tissue, and tumor tissue were assayed at the BCR using the Illumina Infinium QC array. Whole exome sequencing Library construction Library construction was performed as described in [[201]57], with the following modifications: initial genomic DNA input into shearing was reduced from 3 μg to 20–250 ng in 50 μL of solution. For adapter ligation, Illumina paired-end adapters were replaced with palindromic forked adapters purchased from Integrated DNA Technologies with unique dual-indexed molecular barcode sequences to facilitate downstream pooling. Kapa HyperPrep reagents in 96-reaction kit format were used for end repair/A-tailing, adapter ligation, and library enrichment PCR. In addition, during postenrichment SPRI cleanup, the elution volume was reduced to 30 μL to maximize the library concentration, and a vortexing step was added to maximize the amount of template eluted. In-solution hybrid selection After library construction, the libraries were pooled into groups of up to 96 samples. Hybridization and capture were performed using the relevant components of Illumina’s Nextera Exome Kit following the manufacturer’s suggested protocol, with the following exceptions. First, all libraries within a library construction plate were pooled prior to hybridization. Second, the Midi plate from Illumina’s Nextera Exome Kit was replaced with a skirted PCR plate to facilitate automation. All hybridization and capture steps were automated on an Agilent Bravo liquid handling system. Preparation of libraries for cluster amplification and sequencing After postcapture enrichment, library pools were quantified using qPCR (automated assay on the Agilent Bravo) using a kit purchased from KAPA Biosystems with probes specific to the ends of the adapters. For qPCR quantification, the concentrations of the libraries were normalized to 2 nM. Cluster amplification and sequencing Cluster amplification of DNA libraries was performed according to the manufacturer’s protocol (Illumina) using exclusion amplification chemistry and flow cells. Flow cells were sequenced utilizing sequencing-by-synthesis chemistry. The flow cells were then analyzed using RTA v.2.7.3 or later. Each pool of whole exome libraries was sequenced on 76 paired cycle runs with two 8 cycle index reads across the number of lanes needed to meet coverage for all libraries in the pool. Pooled libraries were subjected to HiSeq 4000 paired-end runs to achieve a minimum of 150 × target coverage per sample library. The raw Illumina sequence data were demultiplexed and converted to fastq files; adapter and low-quality sequences were trimmed. The raw reads were mapped to the hg38 human reference genome, and the validated BAMs were used for downstream analysis and variant calling. Genomic data analysis Somatic variant calling Somatic variants were called from WES tumor and normal paired BAMs using SomaticWrapper v1.5, a pipeline designed for the detection of somatic variants from tumor and normal exome data. The pipeline merges and filters variant calls from MuTect v1.1.7 [[202]58]. SNV calls were obtained from Strelka, Varscan, and Mutect. Indel calls were obtained from Stralka2, Varscan, and Pindel. The following filters were applied to obtain variant calls of high confidence: normal VAF ≤ 0.02 and tumor VAF ≥ 0.05; read depth in tumor ≥ 14 and normal ≥ 8; and indel length < 100 bp. All variants must be called by 2 or more callers; all variants must be exonic; and all variants must exclude variants in dbSNP but not in COSMIC. Single-nuclei RNA sequencing Single-nuclei RNA library preparation and sequencing Approximately 20–30 mg of cryopulverized powder from GBM specimens was resuspended in lysis buffer (10 mM Tris–HCl (pH 7.4), 10 mM NaCl, 3 mM MgCl2, and 0.1% NP-40). This suspension was pipetted gently 6–8 times, incubated on ice for 30 s, and pipetted again 4–6 times. The lysate containing free nuclei was filtered through a 40 μm cell strainer. We washed the filter with 1 mL of wash and resuspension buffer (1 × PBS + 2% BSA + 0.2 U/μL RNase inhibitor) and combined the mixture with the original filtrate. After a 6-min centrifugation at 500 × g and 4 °C, the nuclear pellet was resuspended in 500 μL of wash and resuspension buffer. After DRAQ5 staining, the nuclei were further purified by fluorescence-activated cell sorting (FACS). The FACS-purified nuclei were centrifuged again and resuspended in a small volume (approximately 30 μL). After counting and microscopic inspection of nuclear quality, the nuclei were diluted to approximately 1,000 nuclei/μL. Approximately 20,000 nuclei were subjected to single-nuclei RNA sequencing (snRNA-seq) on the 10X Chromium platform. We loaded single nuclei onto a Chromium Chip B Single Cell Kit, 48 rxns (10 × Genomics, PN-1000073), and processed them through a Chromium Controller to generate GEMs (Gel Beads in Emulsion). We then prepared the sequencing libraries with the Chromium Single Cell 3’ GEM, Library & Gel Bead Kit v3, 16 rxns (10 × Genomics, PN-1000075) following the manufacturer’s protocol. Sequencing was performed on an Illumina NovaSeq 6000 S4 flow cell. The libraries were pooled and sequenced using the XP workflow according to the manufacturer’s protocol with a 28 × 8x98 bp sequencing recipe. The resulting sequencing files were available as FASTQ files per sample after demultiplexing. snRNA-seq data preprocessing For each sample, we obtained the unfiltered feature-barcode matrix per sample by passing the demultiplexed FASTQs to the Cell Ranger v3.1.0 ‘count’ command using default parameters, and a customized pre-mRNA GRCh38 genome reference was built to capture both exonic and intronic reads. The customized genome reference modified the transcript annotation from the 10 × Genomics prebuilt human genome reference 3.0.0 (GRCh38 and Ensembl 93). Seurat v3.1.2 was used for all subsequent analyses. We constructed a Seurat object using the unfiltered feature-barcode matrix for each sample. A series of quality filters were applied to the data to remove those cell barcodes that fell into any one of the following categories recommended by Seurat: too few total transcript counts (< 300); possible debris with too few genes expressed (< 200) and too few UMIs (< 1,000); possibly more than one cell with too many genes expressed (> 10,000) and too many UMIs (> 10,000); possibly dead cells or a sign of cellular stress and apoptosis with too high of a proportion of mitochondrial gene expression relative to the total transcript count (> 10%). Each sample was scaled and normalized using Seurat’s ‘SCTransform’ function to correct for batch effects (with parameters: vars.to.regress = c("nCount_RNA","percent.mito"), variable.features.n = 3000). We then merged all the samples and repeated the same scaling and normalization method. All cells in the merged Seurat object were then clustered using the original Louvain algorithm [[203]59]: Fast unfolding of communities in large networks, J. Stat. Mech. Theor. Exp., 2008 (2008), p. P10008) and the top 30 PCA dimensions via Seurat’s ‘FindNeighbors’ and ‘FindClusters’ (with parameters: resolution = 0.5) functions. The resulting merged and normalized matrix was used for subsequent analysis. snRNA-seq cell type annotation Cell types were assigned to each cluster by manually reviewing the expression of marker genes. The marker genes used were referred to CellMarker2.0, and previously published glioma single cell researches. snRNA-seq analysis Differentially expressed genes within among different cell types were identified by the FindMarkers function, which compares cells belonging to one cluster to those belonging to the other clusters. The Wilcoxon statistical test was used. A log2 FC > 0.3 and an FDR < 0.05 were used to filter DEGs. Analysis of cell–cell interactions To analyze the cellular cross-talk between cell types in different regions, CellPhoneDB (version 2.0.1) [[204]60, [205]61], was utilized to identify significant ligand-receptor interactions. The interaction score refers to the mean of the average expression values for all individual ligand-receptor partners in corresponding interacting pairs for the for the different cell types. Expansion microscopy (ExM) for proteomics Tissue expansion Tissue expansion for proteomics was performed following a previously published protocol [[206]52, [207]62]. Here, we further explain the specific steps in the protocol. Firstly, we used paraformaldehyde-fixed tissues, which preserve proteins and tissue structure to prevent diffusion. Secondly, we applied the succinimidyl ester of 6 ((acryloyl) amino) hexanoic acid (acryloyl-X (AcX), 0.1 mg/mL in PBS) to the tissue, which was then incubated overnight in a humid chamber at room temperature. AcX can form a linker that covalently binds to the primary amine groups on proteins. This interaction facilitates the incorporation of AcX into the swellable hydrogel polymer during gelation. Consequently, the bonds formed by AcX on the proteins are tethered to the hydrogel polymer chains, preserving the spatial organization of biomolecules relative to one another. Thirdly, the tissue was incubated in the expansion gel solution. Initially, tissues were incubated in the gel solution on ice for 1 h, which included 0.01% (w/v) 4-hydroxy-TEMPO, 2 M NaCl, 8.6% (w/v) sodium acrylate, 30% (v/v) acrylamide/bisacrylamide (30% solution; 37.5:1), and 0.2% (w/v) APS in 1 × PBS. After removing the gel solution, tissues were then incubated with an activated gel solution containing the same components plus 0.2% (w/v) TEMED. The activated solution was spread into a gelation chamber and incubated at 37 °C for 2 h. The gelation chamber was constructed using a glass microscope slide and two coverslip spacers (Fisher Scientific, catalog no. 12-541B), forming a gel with a thickness of approximately 170 μm. After gelation, the gel was peeled off from the chamber and homogenized in denaturation buffer (200 mM SDS, 200 mM NaCl, 50 mM Tris in Milli-Q water) at 75 °C for 4 h. SDS served as a homogenization agent, aiding lipid removal while preserving protein integrity by maintaining their anchoring to the hydrogel. This step ensured that the sample maintained its structural integrity throughout the expansion process. The homogenized samples were then washed in on a shaker for three times (30 min per wash) at room temperature. To facilitate the visualization of the samples after expansion, we applied Coomassie staining for gel samples. For Coomassie staining, gels were washed 3 times with 50% methanol for 15 min per wash and 1 time with 100% methanol for 1 h at room temperature. Methanol was replaced with Coomassie stain solution and incubated for 1.5 h at 95 °C then replaced with fresh Coomassie stain solution and incubated for another 1.5 h at 95 °C. Although gels may shrink to the original size or smaller during Coomassie staining, the mechanical stability of the hydrogel in the expanded state was not effected. For gel destaining, samples were serially washed in 50 mM Tris, 25 mM Tris, 10 mM Tris and 2.5 mM Tris (all buffers pH = 8.8) for 1 h each at 60 °C. Finally, the samples were washed in Milli-Q water to initiate expansion, with the water changed every one hour until the samples reached a needed expansion factor, which is a sixfold expansion factor in this study. The expanded tissue gel can retain the spatial distribution of proteins. Technically, the expansion factor can be adjusted according to user requirements, such as threefold, sixfold, ninefold and so on. In our proof-of-principle study, we used the sixfold expansion factor for all samples, which was corresponded to a 216-fold (6 × 6 × 6) increase in volume of both the sample and the surrounding gel. The original sample thickness was 10 μm, which expanded to 60 μm after expansion. The initial gel thickness of approximately 170 μm increased to around 1,202 μm after the expansion process. In our study, we ensure that the gel is homogeneous during the hydrogel expansion procedures. Specifically, (1) as for the hydrogel composition, we referred to previously published articles [[208]15]. Our hydrogel composition includes acryloyl-X (AcX), a linker that has been reported to covalently binds to the primary amine groups on proteins. This interaction facilitates the incorporation of AcX into the swellable hydrogel polymer during the gelation phase [[209]15]. Consequently, the connections formed by AcX on the proteins are tethered to the hydrogel polymer chains, effectively preserving the spatial organization of biomolecules relative to one another. (2) In addition, we included a mechanical homogenization step, using sodium dodecyl sulfate (SDS) to maintain the structural integrity and organization of the sample throughout the expansion process. SDS serves as a homogenization agent that not only aids in lipid removal but also preserves protein integrity by affecting their conformation and charge without disrupting their anchoring to the hydrogel [[210]15]. As the hydrogel-tissue hybrid expands uniformly upon immersion in water, this physical magnification of the tissue ensures that the original localization of proteins and the overall organizational structure are retained. Taken together, the hydrogel composition and the hydrogel expansion procedures ensure the homogeneity of the gel and the tissue. Details on tissues before and after expansion In this proof-of-principle study, we applied a sixfold expansion factor consistently across all samples, resulting in an isotropic increase in length, width, and height. This corresponded to a 216-fold (6 × 6 × 6) increase in volume of both the sample and the gel. The original thickness of samples used in our study is 10 μm, and the sample thickness after expansion is 60 μm. For gel thickness, the gels started with a thickness of approximately 170 μm, which expanded to around 1,202 μm after the expansion process. Moreover, the images of samples before and after the expansion were presented to show the tissue sizes of the pre-expanded and post-expanded samples (Figure S2 A). Sample segmentation The expanded tissue gel retained the spatial distribution of proteins. Subsequently, the expanded tissue was physically segmented into numerous volumetric units utilizing a custom-designed 3D-printed cutting toolkit. This toolkit facilitated the precise division of the expanded tissue into thousands of cubic voxels, with an adjustable cutting pitch. The utilization of the 3D-printed cutting toolkit ensured uniform and stable sample segmentation. By precisely controlling the cutting parameters, the toolkit enabled the division of tissue into voxels of stable dimensions. Here, we divided sixfold expended tissues into 2 mm × 2 mm voxels (~ 333 μm × 333 μm original regions). The details of the cutting toolkit In our study, the cutting toolkit is used for cutting the expanded gel into voxels. To better explain the details of the cutting toolkit, we describe the preprocessing steps for the samples before cutting briefly. Firstly, the sample was embedded in the hydrogel, followed by homogenization of the tissue-hydrogel composite to ensure uniformity before expansion. Secondly, the tissue-hydrogel composite was expanded in water. Thirdly, the expanded tissue-hydrogel composite was placed in the cutting toolkit. Subsequently, the expanded tissue-hydrogel composite was cut into voxels of the specific size using the cutting toolkit. The cutting toolkit includes a specialized 3D-printed platform, which can support the gels and facilitate the cutting process (Figure S1 A-B). The 3D-printed platform is a grid base with evenly spaced grooves that divide the surface into multiple small squares. The users can place the gel onto the platform, and then the gel can be cut into voxels along these grid lines. The distance between the centers of two adjacent grooves is designed to match the specific voxel size (e.g., 2 mm in this proof-of-principle study). We can accomplish a precise division of the expanded tissue into hundreds or thousands of uniform cubic voxels during the cutting process. The specific size of the voxels can be adjusted by using cutting toolkit with different sizes. This cutting toolkit enables precise cutting of the gel into uniform voxels based on the defined sizes. After placing the gel onto the cutting platform, we utilized a robotic arm (Tecan Freedom EVO) equipped with a fine, tensioned stainless steel cutting wire (30 µm diameter) to uniformly divide the gel into specified voxels along the grooves of the cutting toolkit (Figure S1B). The cutting size can be customized by the researchers by adjusting the cutting toolkit and the robotic arm, allowing for the generation of voxels with varying sizes. The design of the cutting toolkit aimed to provide flexibility in accommodating both the original size of the target samples and the specific requirements for the resolution by researchers. To this end, we have utilized this customizable and replaceable 3D-printed cutting toolkit that was able to align with the cutting procedure and the gel samples, according to the specific experimental needs. The method for recording the position of voxels within the sample The expanded gel sample was first imaged using Biorad’s imaging system (ChemiDoc, Biorad) (Figure S15 A). The length and width of the image were recorded by image J (Biorad). The recorded image was then segmented according to the custom-determined voxel size. Each segmented voxel will be recorded with its corresponding coordinates (x,y), and marked with a barcode (Figure S15 A). The expanded gel sample was then segmented by the robotic arm according to the marked image. The segmented voxels were then transferred to a 96-well plate in the order of barcodes, and processed with further peptide extraction and LC–MS/MS analysis. The barcode of each voxel will then be utilized to locate the proteomic data of the voxel back to its location on the expanded gel sample as well as the pre-expanded sample. The process of transferring the cut tissue samples After cutting, the voxels were transferred using a robotic arm. The robotic arm was equipped with a vacuum suction tip that allowed for the gentle attachment and movement of the hydrogel. Since the tissue pieces remained embedded within the hydrogel, the transfer of gel pieces also transferred the cut tissue pieces. This suction mechanism ensured that the hydrogels were moved without compromising their integrity, minimizing any potential damage during the transfer process. Then, the tissue pieces were transferred to the multi-well plates, such as the 96-well plate, for further processing. The robotic arm can execute continuous transfers of the hydrogel in a predetermined sequence, enabling fast and high-throughput operations. During the transfer, the (x,y) coordinates of the voxels were assigned to the specific wells on the 96-well plate, ensuring that after transfer and subsequent analysis, the data could be mapped back to the original locations. The robotic arm was capable of executing continuous transfers in a predetermined sequence, enabling fast and high-throughput operations (Figure S1 C). We accurately recorded the positions of voxels within the sample to ensure that the spatial information of the original tissue was faithfully represented during analysis. After segmenting the expanded tissue image and the corresponding H&E image, each voxel was assigned a (x, y) coordinate. These coordinates were then mapped to the corresponding locations in the corresponding H&E image (Figure S15B-D). Proteomic analysis Protein extraction and tryptic digestion The excised tissue-gel samples were washed in ddH[2]O three times at 25 °C and incubated with 50%/50% (v/v) acetonitrile (ACN)/ddH[2]O for 30 min at 30 °C on a shaker. The samples were then washed in 50%/50% (v/v) ACN/100 mM ammonium bicarbonate (ABB) for 10 min and dried in a SpeedVac at 45 °C for 3 min. The dried samples were treated with 20 mM TCEP for 30 min in darkness at 32 °C, followed by alkylation by the addition of 55 mM iodoacetamide (IAA) and incubation for 30 min in the dark at 25 °C. The sample pieces were further washed with 100 mM ABB. The samples were dehydrated by washing with 50%/50% (v/v) ACN/100 mM ABB 2 times for 5 min each at 25 °C on a shaker and then dried in a SpeedVac. Protein digestion was performed with 10 ng/μL trypsin in 25 mM ABB (pH = 8.0) by incubating twice at 37 °C for 4 h and 8 h. The digested peptide solutions were collected in the following steps and combined: 1) 30–40 μL of the supernatant was collected; 2) 100 μL of 25 mM ABB was added and vortexed for 10 min at 25 °C, and the supernatant was collected; 3) 100 μL of 50% ACN/2.5% formic acid was added, and the mixture was vortexed for 10 min; the supernatant was collected, and the process was repeated three times; and 4) 100 μL of 100% ACN was added, and the mixture was vortexed until the gel pieces turned white and sticky. Peptide samples were placed under vacuum to reduce the volume to 20–30 μL. The peptides were then desalted using C18 spin columns (Pierce™ C18 Spin Tips, Thermo Fisher Scientific, US) and dried in a SpeedVac. The cleaned peptides were stored at –20 °C until further analysis. LC‐MS/MS acquisition For FFPE derived bulk proteomic analysis, peptide samples were analyzed on an Easy-nLC 1200 liquid chromatography system (Thermo Fisher Scientific) coupled to a Q Exactive HFX via a nano-electrospray ion source (Thermo Fisher Scientific). The dried peptides were redissolved in 10 μL loading buffer (5% methanol and 0.2% formic acid), and 5 μL of the sample was loaded onto a trap column (100 μm × 2 cm, home-made; particle size, 3 μm; pore size, 120 Å; SunChrom) with a maximum pressure of 280 bar using solvent A, then separated on home-made 150 μm × 12 cm silica microcolumn (particle size, 1.9 μm; pore size, 120 Å; SunChrom) with a gradient of 5–35% mobile phase B (acetonitrile and 0.1% formic acid) at a flow rate of 600 nL/min for 75 min. MS analysis was conducted with one full scan (300–1,400 m/z, R = 120,000 at 200 m/z) at an automatic gain control (AGC) target of 3e6 ions, followed by up to 20 data-dependent MS/MS scans with higher-energy collision dissociation (target 5e4 ions, max injection time 20 ms, isolation window 1.6 m/z, normalized collision energy of 27%). Detection was performed using Orbitrap (R = 7,500 at 200 m/z) and data were acquired using Xcalibur software (Thermo Fischer Scientific). For ExM derived proteomics, samples were measured using LC‐MS instrumentation consisting of an EASY‐nLC 1200 ultra‐high‐pressure system (Thermo Fisher Scientific) coupled via a nano‐electrospray ion source (Thermo Fisher Scientific) to a Fusion Lumos Orbitrap (Thermo Fisher Scientific). The high-performance liquid chromatography (HPLC) gradient was set to 15 min, which enabled efficient peptide separation tailored for our samples. For each voxel analyzed, one run was performed. The total number of runs depended on the specific paraments during sampling, such as the original size of the tissue section, the cutting size, and the expansion factor. In this study, for example, we collected around 600 voxels on average per sample, which resulted in a total of 600 runs on average for each sample. Given that each instrument in our platform can analyze approximately 40 samples per day, the estimated total measurement time for each sample, involving around 600 runs, was approximately 15 days in one instrument. It is important to note that the LC–MS/MS analysis time can be adjusted according to the desired protein coverage. In our study, we utilized a 15-min gradient to optimize the number of identified proteins within a relatively short time. If the objective is to increase the number of identified proteins, the gradient time can be extended. Conversely, for studies focusing on a larger number of samples with less stringent protein identification requirements, the gradient duration can be shortened to enhance throughput. MS analysis was performed in a data‐independent manner (DIA). The DIA method consisted of MS1 scans from 300–1,400 m/z at 60 k resolution (AGC target 4e5 or 50 ms). Then, 30 DIA segments were acquired at 15 k resolution with an AGC target of 5e4 or 22 ms for maximal injection time. The setting “inject ions for all available parallelizable time” was enabled. HCD fragmentation was set to a normalized collision energy of 30%. The spectra were recorded in profile mode. The default charge state for MS2 was set to 3. For quality control (QC) of the performance of the LC‒MS/MS proteomics system, standard HEK293 T QC samples were inserted into the sample test sequence at intervals of every 20 runs. Peptide identification and protein quantification For FFPE derived bulk proteomic analysis, MS raw files generated by LC–MS/MS were processed with. MaxQuant, version 2.3.1.0 with a false discovery rate set at 0.01 [[211]63]. Proteins were identified with at least 2 peptide of a minimum length of 6 amino acids by searching against the human Uniprot database (UniprotKB, downloaded in 2022). Protease was Trypsin/P. The maximum number of missed cleavages was set to two. A mass tolerance of ± 10 ppm for precursor was allowed. The fixed modification was Carbamidomethyl (C), and the variable modification was oxidation (M). LFQ quantification [[212]64] was used for protein quantification. For ExM derived proteomic analysis, the raw data were processed using DIA-NN 1.7.10 in the “robust LC (high precision)” mode with RT-dependent median-based cross-run normalization enabled Ref: DIA-NN: neural networks and interference correction enable deep proteome coverage in high throughput). The MS2 and MS1 mass accuracies were set to 20 and 12 ppm, respectively, and the scan window size was set to 6. Although DIA-NN can automatically optimize such parameters, we fixed them to these values to ensure comparability. To generate a project-specific spectral library, a 24-fraction high-pH reversed-phase fractionated precursor library was created from the same tissue specimen and other glioma tissue specimens from a previously published study and acquired in DDA mode. The raw files were analyzed with MSFragger [[213]65]: MSFragger: ultrafast and comprehensive peptide identification in mass spectrometry–based proteomics) under default settings (with the exception that cysteine carbamidomethylation was removed from fixed modifications) to generate the library file used in DIA-NN. The human UniProt (UniProt Consortium, 2022) isoform sequence database (3 AUP000005640) was used to annotate the library. The library was first automatically refined based on the dataset in question at a 0.01 global q-value (using the “Generate spectral library” option in DIA-NN). DIA-NN performs such refinement by identifying the highest score for each library precursor across all runs in the experiment and then replacing the library data with the empirically observed spectrum and retention time. The resulting report was stringently filtered at a 0.01 precursor-level q-value, 0.005 precursor-level library q-value and 0.05 protein group-level q-value. Protein quantification was performed using the MaxLFQ algorithm [[214]66]. In our study, 11,700 proteins were identified in G1 sample (EGFR-mutant sample), 11,329 in G2 sample (IDH1-mutant sample), and 11,465 in G3 sample (IDH1 and EGFR-wild type sample). Patient specific database construction and variant peptide identification The proteogenomic database tool CustomProDB [[215]67] was used to incorporate the somatic SNVs, indels from our previous published 213 patients’ glioma cohort [[216]68], into a searchable protein specific database with default parameters for variant peptide identification. The identified MS2 spectra were removed from the original raw files were searched against customized databases using DIA-NN. The database searching parameters were almost identical to those described above except that Oxidized methionine and protein N-term acetylation were set as variable modifications and the data were filtered at 1% PSM FDR. HLA typing and neoantigen prediction Neoantigen prediction was performed with NetMHCpan v4.0 (NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data), testing all novel 9-mer mutant peptides containing the mutated amino acids for binding to the patient’s germline HLA alleles. A peptide was defined as a putative neoantigen with a predicted binding affinity < 500 nM. The bulk multi-omics data utilized in the PSERP study We utilized the multi-omics data of 213 glioma patients from our previously published paper [[217]17] and built a glioma-specific spectral library. The detailed methods for library construction were as follows: We integrated tumor-specific genomic events (SNVs and InDels), from our previously published glioma multi-omics dataset, which also included the multi-omics data of the same glioma samples utilized for the PSERP-based spatial proteomic analysis in this study. The tumor-specific genomic events for each tumor were translated into 1, 3, or 6 frames and were constructed to the glioma-specific spectral library by CustomProDB [[218]67]. The raw files generated by PSERP-based spatial proteomic analysis were then searched against this glioma-specific spectral library. In this way, we identified 479 tumor-specific mutational peptides in total. These results proved that by including multi-omics data, PSERP could not only reveal the spatial heterogeneity of proteins but also could show the impact of mutations on proteins at the spatial level. Based on phosphoproteomic data from our previously published paper [[219]17], we built a phospho-spectral library, utilizing the FragPipe computational platform [[220]44]. The raw DIA data of the three glioma samples (IDH1-WT/EGFR-mutant G1, IDH1-mutant G2, and IDH1/EGFR-double-WT G3) generated in our research by PSERP were then searched against the phospho-spectral library. The criteria for selecting voxel sizes Based on the previous explanation, the voxel size used in our method can be adjusted according to the custom requirement. The selection of voxel size is primarily based on two factors: proteomic identification depth and detection throughput. 1. Regarding identification depth: during the experimental design, to evaluate proteomic identification depth at different voxel sizes, we performed expansion and sampling of a homogeneous glioma tumor sample. We selected samples with post-expansion sizes of 1 mm × 1 mm, 2 mm × 2 mm, and 5 mm × 5 mm (corresponding to actual sizes of approximately 166 μm, 333 μm, and 833 μm before expansion) for peptide extraction and mass spectrometry analysis. Then, we compared the number of identified peptides and proteins in voxels with different sizes. The results showed that on average, 946 (ranging from 759 to 1110), 2350 (ranging from 2022 to 2671), and 3079 (ranging from 2882 to 3311) proteins were identified at voxel sizes of 1 mm × 1 mm, 2 mm × 2 mm, and 5 mm × 5 mm (corresponding to 166 μm × 166 μm, 333 μm × 333 μm, and 833 μm × 833 μm before expansion), respectively. These results suggested that the number of identified proteins increased with tissue volume and plateaued at a voxel size of 2 mm × 2 mm after expansion (corresponding to 330 μm × 330 μm before expansion) (Figure S15E). Using larger voxel sizes may lead to a reduction in spatial resolution without a significant increase in the number of identified proteins. 2. Regarding detection throughput: in our method, the voxel size and the target sample area size together determine the number of voxels that need to be analyzed. In our study, for a target sample area of approximately 5 cm × 5 cm after expansion, selecting a 2 mm voxel size resulted in approximately 600 voxels. Further reducing the voxel size would significantly increase the number of voxels, thereby greatly increasing the time and equipment costs for detection. Therefore, voxel size selection also depended on the actual detection throughput and costs. Here, we presented the corresponding results for different voxel sizes from the sample after the six-fold expansion to provide researchers with a reference for the suitable selection (Table S1). Taken together, based on the considerations of both identification depth and detection throughput, we selected a voxel size of 2 mm × 2 mm (corresponding to the actual size of 333 × 333 μm) for our proof-of-principle study. For each voxel, we also evaluated its peptide concentration using BCA analysis. The result revealed that average 104 ng peptides could be extracted from each 2 mm × 2 mm voxel, corresponding to the actual size of 333 × 333 μm. Moreover, both the specific voxel size and the expansion factor can be adjusted according to the user’s specific needs for further studies. The flexible combinations of the voxel size and the expansion factor can result in different actual sizes of the voxel and different number of voxels (Table S1). The deal of the boarder of tissue To deal with the boarder of the tissue where no full squared voxels can be obtained, we used the two quality control procedures for border voxels based on two methods used in spatial transcriptomic studies [[221]60, [222]69–[223]72]. 1.The alignment with H&E image: in multiple spatial transcriptomic studies [[224]60, [225]69, [226]71], spots were automatically aligned to the paired H&E images by software, and all spots under tissue detected by the software were included in downstream analysis. According to this method, we captured the H&E image of each sample, and determine the tissue area according to the image (Figure S16 A). We recorded the coordinates of all voxels containing tissue as mentioned above. After that, all the voxels containing tissue, including the border voxels which were not fully populated with tissue, were processed for peptide extraction, and mass spectrometry analysis. 2. The cutoff of identification number for downstream analysis: several spatial transcriptomic studies also added a cutoff to filter out spots with low identification number. For instance, in Kuppe C et al.’s study [[227]71], spots with less than 300 measured genes and less than 500 UMIs were filtered out. In Kaufmann M et al.’s study [[228]70] spots transcriptomes consisting of < 300 expressed genes were removed. And in Qi et al.’s study [[229]72], spots were filtered for minimum detected gene count of 200 genes. In our study, voxels with fewer than 500 identified proteins were filtered out from the following analysis. Additionally, we compared the number of identified proteins among normal voxels which were fully populated with tissue, border voxels which were not fully populated with tissue, and blank gel voxels (Figure S16B). The result indicated that border voxels which were not fully populated with tissue had a relatively lower number of identified proteins compared to the fully populated voxels (Figure S16 C). However, the number of identified proteins in these boundary voxels was significantly higher than that of the control blank gel voxel located outside the tissue border that contained no tissue at all (Figure S16 C). Moreover, during our unsupervised clustering analysis, the malignant region boundary voxels with relatively low protein identification did not segregate clearly from other malignant voxels; instead, they clustered together, indicating that these boundary voxels retained meaningful biological information (Figure S16D-E). Similarly, non-tumor boundary voxels were also grouped within the non-tumor cluster (Figure S16D-E), further suggesting that the boundary voxels can contribute valuable insights into the biological context of the samples. In summary, to ensure the quality of data, we integrated the protein identification results with the image recognition outcomes to filter the border voxels, which allowed us to utilize high quality data for subsequent analysis and enhance the reliability of our findings. Bioinformatic analysis Proteomic data analysis was performed within the R environment ([230]https://www.r-project.org/). Proteins with more than 30% missing values (those displayed as 0 in the MaxQuant output) were removed. Missing values were subsequently imputed using a normal distribution approach with parameters set to a width of 0.3 and a downshift of 1.8 prior to statistical analysis. To assess differences in protein distribution between two groups, we employed the Wilcoxon rank-sum test to compare differences between two groups of voxels. The paired t-test was used to compare the means of two different groups from the same samples. Statistical significance was determined using a Bonferroni correction to account for multiple comparisons. In addition, we applied a permutation-based false discovery rate (FDR) adjustment with a significance threshold of 5% for multiple hypothesis testing. Functional enrichment analysis of multiomics data using GSVA/ssGSEA To further analyze the biological characteristics of the different samples, we performed single-sample gene set enrichment analysis (ssGSEA/GSVA). Gene expression data of proteome data across different samples were used to determine enrichment scores across gene sets (i.e., KEGG, Reactome, Gene Ontology and HALLMARK) downloaded from the MsigDB (v7.4, [231]https://data.broadinstitute.org/gseamsigdb/msigdb/release/7.4/) with at least 10 overlapping genes via the R/Bioconductor package GSVA. GSEA was performed with GSEA software ([232]https://www.gseamsigdb.org/gsea/index.jsp). Gene sets including KEGG, Reactome, Gene Ontology and HALLMARK downloaded from the MsigDB (v7.4, [233]https://data.broadinstitute.org/gsea-msigdb/msigdb/release/7.4/) were used as the background. Estimation of stromal and immune scores xCell analysis ([234]https://xcell.ucsf.edu/) [[235]73] was used to infer stromal and immune scores based on the RNA-seq data. The abundance of 64 different cell types was also computed via xCell. Application of Cottrazm The Cottrazm clustered spots of the G1 sample using the K-nearest neighbor (KNN) algorithm based on the normalized spatial protein expression matrix. The results revealed the six clusters (Figure S5 C). The CNV scores for each spot were calculated using the InferCNV algorithm. Two clusters with the greatest CNV scores were identified as the core spots of malignant spots (Figure S5D). Spatial spots were then arranged, and the Manhattan distance between spots was calculated. The neighboring spots of the tumor core where the Manhattan distance was less than a linear model-fitted radius were identified. Spots as malignant spots or tumor boundary spots were then determined based on the UMAP distance to the tumor centroid (Figure S5E and S5 F). Fuzzy C-means Clustering Proteins from spatial proteomic data were grouped into different clusters using Mfuzz package in R with fuzzy c-means algorithm [[236]74]. Integrated analysis of single-cell and spatial proteomic data by Bulk2Space We generated spatially resolved single-cell expression profiles from bulk proteomes via Bulk2Space ([237]https://github.com/ZJUFanLab/bulk2space) [[238]75] using high-quality scRNA-seq data and spatial proteomics as references.