Abstract Background & Aims Despite atezolizumab plus bevacizumab being a standard treatment for advanced hepatocellular carcinoma (HCC), a significant proportion of patients do not achieve durable benefit. This study aimed to identify predictive biomarkers for this therapy by investigating the role of immune activation within the tumor microenvironment (TME). Methods We characterized the intratumoral TME of patients with advanced HCC treated with atezolizumab plus bevacizumab using single cell transcriptomics on pretreatment tumor biopsies from 12 patients. To complement and support these findings, we integrated our single cell data with publicly available bulk RNA-sequencing data from independent clinical trial cohorts. Results Patients who responded to combination therapy with atezolizumab plus bevacizumab demonstrated an immune-activated TME, marked by enhanced cytotoxicity and a tumor-specific T cell response. These patients also exhibited an increased proportion of inflammatory cytokine-enriched tumor-associated macrophage clusters with stronger interactions with T cells, an increased population of conventional dendritic cells, and activated antigen-presenting function in tumor endothelial cells. When publicly available bulk RNA-sequencing data from independent clinical trial cohorts were analyzed, these immune activation features were associated with improved progression-free survival (median 10.8 months, 95% CI: 7.3–not reached versus 5.5 months, 95% CI: 4.0–6.7; p <0.001). Conclusions These findings suggest that the existence of an activated immune TME before treatment is crucial for a favorable clinical response in patients with HCC treated with atezolizumab plus bevacizumab. Impact and implications: Only a subset of patients with HCC benefit from combination therapy with atezolizumab plus bevacizumab, limiting its clinical utility. In this study, we used single cell RNA analysis to identify TME features associated with a clinical response to this therapy. Our findings suggest that a pre-existing immune-activated TME is crucial for predicting the response to atezolizumab plus bevacizumab. Specifically, features such as enhanced T cell cytotoxicity, inflammatory cytokine-enriched macrophage clusters, active antigen presentation in endothelial cells, and an increased presence of dendritic cells may aid patient selection and inform therapeutic strategies. Keywords: RNA, Single cell, Immunotherapy, Immune activation, Biomarkers, Macrophages, Tumor associated Graphical abstract [49]Image 1 [50]Open in a new tab Highlights: * • An activated immune microenvironment in advanced HCC is associated with treatment response to atezolizumab plus bevacizumab combination therapy. * • Responders exhibit increased cytotoxic T cells, inflammatory cytokine-rich TAMs, cDC2 cells, and antigen presentation by TECs. * • The 10-gene sc_ABRS signature, identified through single-cell analysis, is associated with PFS in the GO30140/IMBRAVE150 database. Introduction The combination of atezolizumab and bevacizumab is now a standard first-line therapy for advanced hepatocellular carcinoma (HCC).[51]^1 This immunotherapy and anti-angiogenic combination has shown significant survival benefits compared with previous standards of care,[52]^2^,[53]^3 because atezolizumab enhances antitumor immunity and bevacizumab targets angiogenesis, a key driver of tumor growth and immune evasion in HCC.[54]^4^,[55]^5 However, the objective response rate to this combination therapy is only ∼30%, highlighting the crucial need to identify biomarkers that can predict response and optimize treatment strategies in advanced HCC. Although several biomarkers, such as programmed death-ligand 1 (PD-L1) expression, tumor mutational burden, and microsatellite instability, have been proposed as predictors of immunotherapy response in various cancers,[56][6], [57][7], [58][8] their predictive value in HCC has yet to be fully established.[59]^9 This uncertainty may stem from the inherent heterogeneity of HCC, characterized by diverse systemic immunity, pathogenesis, histopathology, and molecular features.[60]^10^,[61]^11 Moreover, the complex interplay within the tumor microenvironment (TME) can modulate treatment response and potentially contribute to drug resistance.[62][12], [63][13], [64][14] Therefore, a thorough understanding of these intricate interactions is essential for accurately predicting immunotherapy response in HCC. Single cell-level analysis is a highly effective method for evaluating the heterogeneity of cellular components in the TME.[65]^15 However, a comprehensive investigation of the correlation between treatment response to atezolizumab plus bevacizumab and TME at the single cell level in advanced HCC has been hindered by the challenging nature of procuring fresh tumor tissue. In this study, we conducted single cell RNA sequencing (scRNA-seq) of freshly acquired biopsy samples from patients with advanced HCC to analyze cellular and molecular mechanisms and identify predictive markers for clinical response to atezolizumab plus bevacizumab. Materials and methods Sample collection This study was approved by the Institutional Review Board of the Samsung Medical Center. Written informed consent was obtained from each patient (IRB No. 2021-01-021-009). Between June 2020 and February 2023, tumor biopsy samples were collected from 18 patients with advanced HCC, including patients with Barcelona Clinic Liver Cancer (BCLC) stage B HCC with diffuse, bilobar involvement for whom locoregional treatments were not feasible, and patients with BCLC stage C HCC. Pretreatment core-needle biopsies were performed on 17 patients. For one patient who had undergone surgical resection but subsequently developed pulmonary metastases, tumor-infiltrating lymphocytes (TILs) were collected for analysis. ScRNA and T cell receptor V(D)J sequencing The Chromium Single Cell V(D)J Solution from 10x Genomics was used for single-cell 5′ gene expression and V(D)J sequencing. Cell suspensions were loaded onto the Chromium Single Cell Controller to create single cell gel beads in emulsion (GEMs) using Chromium Single Cell V(D)J Reagent Kits. The resulting libraries for single-cell 5′ gene expression and V(D)J were sequenced on the Illumina Novaseq 6000 platform with 150 paired-end reads. Alignment of the scRNA and V(D)J sequencing data to the hg38 genome and the generation of gene-barcode metrics were performed using Cell Ranger software (v.5.0.0). ScRNA-seq analysis To ensure high-quality cells, those expressing fewer than 300 genes and those with >20% unique molecular identifier (UMI) tags from mitochondria gene-expressed cells were filtered out. After this quality-filtering step, the remaining counts underwent normalization, dimension reduction using principial components analysis (PCA), integration, shared nearest neighbor (SNN) clustering, and uniform manifold approximation and projection (UMAP) visualization of scRNA-sequencing data using the Seurat R package (v4.3.0.1) with default options. Cluster annotations were based on marker gene lists, including PTPRC, CD3D, LYZ, VWF, COL1A2, and other relevant genes. For the analysis of differentially expressed genes (DEGs), the FindAllMarkers or FindMarkers function from Seurat was used with the MAST test method. TCR V(D)J analysis and clonotype identification Using filtered annotation T cell receptor (TCR) data from Cell Ranger (v5.0.0), only productive CDR3 sequences matched with UMIs from scRNA-seq data were selected for subsequent analysis. New clonotype IDs were generated by concatenating the CDR3 protein sequences of alpha and beta chains, taking into account all possible chain combinations. Instances of multiple chains, such as two beta chains and one alpha chain, or two alpha chains and one beta chain, were all considered in the subsequent analysis. To quantify the abundance of clonality, the number of cells with the same clonotype ID was counted. The Gini index, used as a measure of clonality, was calculated using the Vegan package in R. Gene set enrichment pathway analysis For functional pathway analysis, we performed gene set enrichment pathway analysis (GSEA) using the DEG list. This analysis was conducted using the clusterProfiler and fgsea package in R, with the curated gene sets (C2) or ontology gene sets (C5) from the Molecular Signature Database v7.2 ([66]www.gsea-msigdb.org/gsea/msigdb). For specific gene sets, such as tissue-resident T cell markers[67]^16 or tumor-specific markers,[68]^17 we used gene sets listed in related publications. Cell–cell interaction analysis To investigate cell–cell interactions between cell types, we used CellphoneDB[69]^18 (v4) to identify significant ligand–receptor combinations using CellphoneDB Data (v4.1.0). The interaction score was calculated based on the mean expression values of individual ligand–receptor pairs across corresponding cell types within clinical groups, where at least 10% of cells expressed both the ligand and receptor genes. Statistical significance was assessed through a permutation test with 1,000 iterations, with other parameters set to their default options. Gene expression signature analysis The phagocyte, M1, M2, and angiogenesis signature scores in the scRNA-seq analysis were calculated using the gene sets described in [70]Table S1 and the AddModuleScore function in the Seurat package. To examine the correlation between specific cell clusters and patient survival in bulk RNA-seq data, a mean of marker gene expression was used, and the groups were categorized based on the 50th percentile. Multiplex immunofluorescence Formalin-fixed paraffin-embedded (FFPE) tissue sections (n = 9) from biopsy samples collected for single cell analysis were subjected to multiplex immunofluorescence (mIF) staining. Sections were cut at 4 μm, deparaffinized, rehydrated, and stained following the manufacturer’s protocol. mIF staining targeted DAPI, CD68, CD163, and pan-cytokeratin (CK) using the Opal 7-color fluorescence IHC kit (PerkinElmer, Waltham, MA, USA) and the BOND RX system (Leica Microsystems, Vista, CA, USA).[71]^19 Antibodies and fluorophores included CK (AE1/AE3, Dako; Opal 620), CD68 (KP1, Dako; Opal 690), CD163 ([72]EPR19518, Abcam; Opal 570), and CCL3 (1H20L19, Invitrogen; Opal 520). Spectral images of multiplex-stained slides were captured with the Vectra Polaris Automated Quantitative Pathology Imaging System (PerkinElmer). Of the nine slides evaluated, one was excluded because of suboptimal staining, leaving eight slides for analysis. For these, 15 regions of interest (ROIs) were selected (1–3 ROIs per slide). Cell segmentation and annotations for CD68, CD163, CCL3, and CK were performed in the ROIs, with 8–19 cells annotated per antibody per ROI to train the software in cell classification. The inForm software (version 3.0.0, Akoya Biosciences, Marlborough, MA, USA) determined the positivity or negativity of each antibody. Cells positive for CD163 and negative for CK were classified as tumor-associated macrophages (TAMs), and the proportion of CCL3-positive TAMs was calculated. All analyses were performed by experienced pathologists. Integration of the publicly available bulk RNA-seq dataset from the GO30140 phase Ib and IMbrave150 phase III clinical trials To further explore the clinical relevance of our single cell-derived findings on progression-free survival (PFS) and overall survival (OS), we analyzed a publicly available bulk RNA-seq dataset. This included 311 tumor samples[73]^20 from patients with advanced HCC treated with either atezolizumab plus bevacizumab (n = 253) or sorafenib (n = 58) in the phase Ib (GO30140)[74]^5 and phase III (IMbrave150)[75]^2 clinical trials. Raw fastq data (EGAD00001008128) and corresponding clinical data (EGAD00001008130) were obtained from the European Genome-Phenome Archive. The raw reads were aligned to the human reference genome (GRCh37) using STAR v2.5.1b with default parameters, and gene expression quantification was performed using RSEM software. Samples with low library sizes (<1 million reads) were excluded from the analysis, resulting in 217 patients treated with atezolizumab plus bevacizumab being included in the final dataset. Clinical characteristics are summarized in [76]Table S2. Survival and statistical analysis Survival analyses were conducted using the Cox proportional hazards model with a log-likelihood ratio test to evaluate overall model significance. Kaplan-Meier survival analysis stratified samples into high- and low-signature groups. Statistical analyses were performed in R (version 4.1.2). The Wilcoxon test compared continuous variables between two groups. Statistical significance was indicated as follows: ∗p <0.05, ∗∗p <0.01, and ∗∗∗p <0.001. Results Profiling of 10 major cell types in advanced hepatocellular carcinoma To delineate the single cell landscape of advanced HCC, we analyzed 18 tissue samples, including 17 tumor biopsies and one TIL sample, with scRNA-seq using the 10x Genomics platform ([77]Fig. 1A). Clinical characteristics are detailed in [78]Table S3; 12 patients were treated with first-line atezolizumab plus bevacizumab. All but two of these patients were treatment naive. In total, 63,936 high-quality cells (average 3,552 per sample) were analyzed, revealing 10 distinct cell-type clusters based on prior HCC studies[79]^21^,[80]^22 ([81]Fig. 1B,C): C1 myeloid cells (PTPRC and CD68); C2 plasmacytoid dendritic cells (DCs; CLEC4C and CD74); C3 B cells (PTPRC, CD79A, and MS4A1); C4 plasma cells (CD79A and MZB1); C5 T/natural killer (NK) cells (PTPRC, CD3D, CD4, CD8A, and KLRF1); C6 fibroblasts (ACTA2 and COL1A1); C7 endothelial cells (FCN3, PECAM1, and VWF); C8 malignant hepatocytes (HP and KRT7); C9 mast cells (KIT); and C10 cycling cells (STMN1 and MKI67). Fig. 1. [82]Fig. 1 [83]Open in a new tab Single-cell transcriptomic profiling and tumor response to atezolizumab plus bevacizumab in HCC. (A) Flowchart of the study. (B) UMAP visualization displaying 10 distinct cell clusters, each represented by a unique color. (C) Dot plot of canonical markers for each cluster. Circle size indicates the percentage of cells expressing each marker, and color represents the scaled average expression level. (D) Waterfall plot of the maximum change in tumor size. Statistical analysis was performed using the Wilcoxon test. (E) Proportion of major clusters by immunotherapy response group, excluding malignant clusters (hepatocytes). The x-axis represents the response group, with statistical analysis performed using the Wilcoxon test. UMAP, uniform manifold approximation and projection. To identify biomarkers, we performed transcriptomic analysis on 12 patients treated with atezolizumab plus bevacizumab. Patients were classified as responders (partial response [PR], n = 4) or nonresponders (stable disease [SD]/progressive disease [PD], n = 8) based on modified Response Evaluation Criteria in Solid Tumors (mRECIST) criteria[84]^23 ([85]Fig. 1D). The overall response rate was 33.3% (complete response [CR]: 0; PR: 4), with 25.0% SD and 41.7% PD. The proportions of cell types varied substantially across samples, possibly reflecting underlying differences in tumor phenotypes or biopsy locations ([86]Figs. S1A and B). No significant differences were observed in major cell type proportions between responders and nonresponders ([87]Fig. 1E), although immune cell density tended to be higher in responders ([88]Fig. S1C). Enriched cytotoxic and tissue-resident T cell signatures in responders to atezolizumab plus bevacizumab To further investigate the mechanisms underlying the response to atezolizumab plus bevacizumab therapy in HCC, we focused on dissecting T cell clusters. A total of 27,717 T or NK cells were identified and categorized into 14 clusters, including five CD4 T cell subsets, four CD8 T cell subsets, two gamma delta T cell subsets, one mucosal-associated invariant T, and two NK cell subsets ([89]Fig. 2A). We also performed single cell TCR sequencing (scTCR-seq) to assess T cell diversity and clonality, yielding 41,884 TCR sequences with a 90% match to T cells ([90]Fig. S2A). The analysis of number of T cells (abundance) sharing unique CDR3 (Clone ID) indicated an increase in clonality score (Gini index) within CD8 T cells (C6–C9 clusters) in the responder group, although this did not reach statistical significance (p = 0.11) because of the small sample size ([91]Fig. 2B,C). Fig. 2. [92]Fig. 2 [93]Open in a new tab Enhanced cytotoxicity and tissue-resident T cell presence in immunotherapy responders. (A) UMAP visualization of T cells from 18 patients with HCC, showing 14 T and NK cell clusters, each represented by a distinct color. (B) UMAP visualization of clonal abundance, indicating the number of cells with the same TCR clone ID. (C) Boxplots showing the degree of clonality (Gini index) across different response groups. Statistical comparisons were performed using the wilcoxon tests. (D) Heatmap of relative expression levels for canonical T and NK cell markers. (E) Volcano plot displaying differential gene expressions in CD8 T cells between responder and nonresponders, specifically for clusters 6–9. (F) Enrichment plot of GO terms related to cell killing, leukocyte-mediated cytotoxicity, tissue-resident memory gene signatures, and tumor-specific T cell signatures (Melanoma antigen vs. EBV or CMV virus) in the responder group compared with the nonresponder group in CD8 T cells. CMV, cytomegalovirus; EBV, Epstein–Barr virus; HCC, hepatocellular carcinoma; NK, natural killer; TCR, T cell receptor; TRM, tissue-resident memory T cell; UMAP, uniform manifold approximation and projection. Within the CD8+ T cell clusters, distinct subpopulations were delineated, including central memory T cells (Tcm) expressing IL7R (C6, CD8_Tcm_IL7R^+), effector memory T cells (Tem) expressing CCL4 (C7, CD8_Tem_CCL4^+), Tem cells expressing GZMK and exhaustion markers, such as PDCD1, ICOS, and HAVCR2 (C8, CD8_Tem_GZMK^+), and cytotoxic T cells (Tcyt) expressing GZMH (C9, CD8_Tcyt_GZMH^+) ([94]Fig. 2D). When comparing T cell subclusters between responders and nonresponders, the CD4_Tcm_IL7R+ (C2) and CD8_Tcm_IL7R+ (C6) clusters were significantly lower in responders than in nonresponders (p <0.05) ([95]Fig. S2B). To further explore the differences between responders and nonresponders, we performed DEG analysis within CD8+ T cell clusters. Responders exhibited higher expression of exhaustion molecules (such as PDCD1, ICOS, and HAVCR2) and cytotoxic molecules (such as FGFBP2, GNLY, and GZMB) compared with nonresponders ([96]Fig. 2E). GSEA revealed significant enrichment of tumor cell killing-related pathways, such as leukocyte-mediated cytotoxicity, in responders ([97]Fig. 2F). Moreover, gene sets associated with tissue-resident T cells were significantly enriched in the responder group, suggesting a higher presence of such cells in responder patients (Normalized Enrichment Score [NES] = 1.74, p = 0.034). In addition, considering a predominant infection by HBV in our cohort, we validated that these differential gene sets were tumor specific rather than related to viral infection (NES = 1.53, p = 0.004). These findings collectively emphasize T cell activation of cytotoxicity-related pathways, abundance of tissue-resident T cells, and enrichment of tumor-specific signatures in the responder group ([98]Fig. 2F). When we computed signature scores for cytotoxicity and exhaustion, we found that responders had significantly higher cytotoxicity and exhaustion scores in CD8+ T cells (both p <0.001) ([99]Fig. S2C). Proinflammatory TAM_activating_CCL^+ cluster enhances T cell recruitment and predicts response to atezolizumab plus bevacizumab We next performed myeloid cell clustering and generated merged UMAP plots ([100]Fig. 3A), identifying two distinct C1Q+TAM clusters. The resting TAM cluster (C1, TAM_resting_C1Q^+) expressed C1Q genes (C1QA, C1QB, and C1QC) and GPNMB, whereas the activating TAM cluster (C2, TAM_activating_CCL^+) showed proinflammatory chemokine expression (CCL3, CCL4, and CXCL2) along with C1Q genes ([101]Fig. S3A). Additional clusters included MDSC-like_EREG^+ cells (C3) expressing EREG, FCN1, and S100A4, and Neutrophil_CSF3R+ cells (C4) with high neutrophil marker expression (CSF3R, G0S2, and NAMPT) ([102]Fig. 3B). Both TAM_resting_C1Q^+ and TAM_activating_CCL^+ clusters expressed phagocytosis-associated genes (MRC1, CD163, MERTK, and C1QB), although TAM_activating_CCL^+ showed distinctly higher M1-associated features ([103]Fig. 3C). Fig. 3. [104]Fig. 3 [105]Open in a new tab Enhanced inflammatory features in myeloid cells of responders. (A) UMAP of the distribution of myeloid cells, including macrophages, dendritic cells, and other myeloid clusters. Each cluster is indicated by a unique color. (B) Heatmap illustrating the expression levels of marker genes across different myeloid cell clusters. Colors represent the relative expression levels within each cell. (C) Gene-set signatures related to phagocytosis, M1 (proinflammatory), M2 (anti-inflammatory), and angiogenesis scores within the four macrophage/monocyte-related clusters (clusters 1–4). (D) Boxplot of the proportion of each cell cluster within macrophage-related clusters (clusters 1–4). Statistical significance was evaluated using the Wilcoxon test: ∗p <0.05, ∗∗p <0.01. (E) Violin plot depicting the expression of inflammation-related genes (CCL3, CCL4, IL1B, and KLF2) according to the response group in cluster 1 (TAM_resting_C1Q^+) and cluster 2 (TAM_activating_CCL^+). (F) Dot plot of the KEGG pathway enrichment analysis results for the top 100 upregulated genes associated with each macrophage-related cluster (clusters 1–4). Dot color represents FDR values, whereas circle size indicates the gene ratio for each KEGG pathway. (G) Bubble plot of ligand–receptor pairs of CCL3 chemokines between myeloid cells and other cell types. CCL, C–C motif chemokine ligand; FDR, false discovery rate; KEGG, Kyoto Encyclopedia of Genes and Genomes; KLF2, Kruppel-like factor 2; TAM, tumor-associated macrophage; UMAP, Uniform Manifold Approximation and Projection. Analysis of the myeloid cell profiles between responder and nonresponder groups revealed a distinct distribution of myeloid cell clusters. The responder group had a higher proportion of the TAM_activating_CCL^+ cluster (C2), whereas the nonresponder group had higher proportions of the resting TAM cluster (C1) and the Neutrophil_CSF3R^+ cluster (C4) ([106]Figs. S3B and D). Responders also showed significantly elevated expression of proinflammatory mediators, including chemokines CCL3, CCL4, the cytokine IL1B, and the transcriptional regulator KLF2 ([107]Fig. 3E). Further analysis via Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment demonstrated activation of immune-recruiting pathways, including chemokine signaling and NF-κB signaling, in the TAM_activating_CCL+ cluster ([108]Fig. 3F). Cell–cell interaction analysis identified CCL3–CCR5 as a key ligand–receptor pair between myeloid cells and T cells, with stronger interactions observed in responders ([109]Fig. 3G). This was further supported by CCL3 expression analysis in an independent cohort (GO30140 + IMbrave150, n = 217), where strong positive correlations were observed between CCL3 and markers of CD8^+ T cells (CD8A; correlation coefficient [R] = 0.71, p = 2.2e-12) and cytotoxic T cells (GZMH; Pearson’s r [R] = 0.67, p = 8.4e-11) in responders. In nonresponders, weaker correlations were observed between CCL3 and these markers (CD8A: R = 0.46, p = 5.2e-05; GZMH: R = 0.38, p = 0.0011) ([110]Fig. S3C). mIF analysis demonstrated that responders exhibited a high degree of colocalization between CD163^+ and CCL3^+ cells, whereas this coexistence was significantly reduced in nonresponders ([111]Figs. S4A and B). The number of CD163+CCL3^+ TAMs was significantly higher in responders than in nonresponders ([112]Fig. S4C), and a strong correlation with PFS was observed (R = 0.998, p <0.0001; [113]Fig. S4D). cDC2_CD1C^+ dendritic cells are enriched in responders Within the DC population, we identified four distinct clusters: (1) cDC1_CLEC9A^+ expressing markers such as IDO1, XCR1, and CLEC9A; (2) cDC2_CD1C^+ expressing CD1C, CLEC10A, and FCER1A; (3) mDC_LAMP3^+ representing migrating DCs and expressing markers such as CCR7, BIRC3, and LAMP3; and (4) plasmacytoid DCs (pDC) expressing markers IRF7 and LILRA4 ([114]Fig. 3B). Upon comparing DC populations, we observed a higher proportion of cDC2_CD1C+ in responders than in nonresponders ([115]Fig. S3D). Tumor-associated endothelial cells exhibit enhanced antigen presentation in responders To delineate the distinct features of endothelial cells, we performed subcluster analysis on the 1,360 endothelial cells identified. This analysis revealed two normal endothelial clusters: Endothelial_PROX1^+ and liver sinusoidal endothelial cells (LSECs), along with a third cluster representing tumor-associated endothelial cells (TECs) ([116]Fig. 4A). The Endothelial_PROX1^+ cluster expressed TFF3 and PROX1, whereas the LSEC cluster expressed CLEC4G. The TEC cluster exhibited high expression of VWF, PLVAP, and PECAM1 ([117]Fig. 4B). Fig. 4. [118]Fig. 4 [119]Open in a new tab Antigen-presenting characteristics in tumor-associated endothelial cells. (A) UMAP visualization of endothelial cells, color coded by endothelial cell subclusters. (B) Expression levels of endothelial marker genes. (C) Bar plot of significant GSEA results in GO pathways in TEC. Bar color represents the significance of enrichment, measured by the log p value. (D) Expression of antigen-presenting related genes, cell projection, and angiogenesis based on response in TEC. Statistical significance was evaluated using the Student’s t test: ∗∗p <0.01, ∗∗∗p <0.001, ∗∗∗∗ p <0.0001. GSEA, gene set enrichment analysis; GO, Gene Ontology; LSEC, liver sinusoidal endothelial cells; TEC, tumor endothelial cells; UMAP, uniform manifold approximation and projection. To further understand the function of TECs in atezolizumab and bevacizumab combination therapy, GSEA was performed using ranked DEG lists. Among 2,421 significantly enriched pathways (p <0.05), key pathways included antigen processing, cell projection, and angiogenesis ([120]Fig. 4C; [121]Table S4). Notably, antigen-presenting pathways were enriched in the responder group, with high expression of genes crucial for antigen presentation, such as CD74, HLA-DRA, B2M, and HLAC ([122]Fig. 4D). By contrast, the nonresponders showed enrichment in pathways related to cell projection, including processes such as assembly, organization, and regulation. Angiogenesis pathways were also enriched in nonresponders, particularly those involved in the positive regulation of blood vessel endothelial cell proliferation, sprouting angiogenesis, and blood vessel maturation. Genes such as EMP1, ICAM1, SEPTIN2, and TLN1, known to have roles in cell projection, were highly expressed in the nonresponder group. In addition, genes contributing to angiogenesis, including CD40, HSPG2, RAMP2, and TGFBR3, exhibited higher expression in nonresponders compared with responders. Integration of bulk RNA-seq data reveals an activating TME-associated gene signature for predicting response to atezolizumab plus bevacizumab in HCC To evaluate the predictive value of gene signatures from single cell analysis, we calculated cytotoxicity, exhaustion, activated TAM, and cDC2 scores using transcriptomic data from 217 pretreatment tumor biopsies (GO30140 + IMbrave150) and assessed associations with PFS and OS. Patients with high cytotoxic or exhaustion scores (top 50th percentile) had significantly longer PFS under atezolizumab plus bevacizumab (p = 0.014 and p = 0.027, respectively; [123]Fig. 5A,B). High activated TAM and DC scores were similarly linked to longer PFS (p = 0.0039 and p = 0.0022, respectively; [124]Fig. 5C,D). These signatures were not predictive for PFS in patients treated with sorafenib ([125]Fig. 5E–H). For OS, only the cytotoxic T cell signature was significantly associated with this measure (p = 0.024; [126]Figs. S5A–D). Based on these findings, we developed a 10-gene atezolizumab plus bevacizumab response signature (sc_ABRS) encompassing cytotoxic/exhausted T cells, activated TAMs, and cDC2 cells ([127]Table S1). High sc_ABRS expression was associated with an extended PFS of a median of 10.8 months (95% CI: 7.3 months–not reached) compared with 5.5 months (95% CI: 4.0–6.7 months; p <0.001) ([128]Fig.5I), but did not correlate with OS (p = 0.29; S5E). With an AUC of 0.75 for PFS, sc_ABRS performed comparably to established biomarkers, such as CD274 (AUC = 0.72), effective T cell signature (AUC = 0.76) and atezolizumab + bevacizumab response signature[129]^20 (AUC = 0.76) ([130]Fig. 5J). Although sc_ABRS did not outperform these markers, its single cell origin provides a more detailed view of the tumor immune microenvironment. Fig. 5. [131]Fig. 5 [132]Open in a new tab Predictive value of single-cell derived gene signatures for response to atezolizumab plus bevacizumab in HCC. (A–D) Kaplan-Meier plots of PFS in 217 patients from the IMBRAVE150 + GO30140 trial treated with atezolizumab plus bevacizumab, stratified by high versus low (median split) expression of the following gene signatures: (A) cytotoxic, (B) exhaustion, (C) activated TAM, and (D) cDC2. Patients with higher expression of these signatures showed significantly longer PFS. (E–H) Kaplan-Meier plots of PFS in 58 patients treated with sorafenib in the IMBRAVE150/GO30140 trial, showing no significant differences between high and low expression groups for the same gene signatures: (E) cytotoxic, (F) exhaustion, (G) activated TAM, and (H) cDC2. (I) Violin plots comparing sc_ABRS (a 10-gene signature derived from single cell analysis) scores across different radiological response categories (CR, PR, SD, and PD) based on RECIST criteria, with p values from two-sided Student’s t tests. (J) Kaplan-Meier plot showing PFS stratified by high versus low sc_ABRS scores in patients treated with atezolizumab plus bevacizumab. (K) ROC curve analysis demonstrating the predictive performance of the sc_ABRS score compared with previously reported biomarkers, including effective T cell signatures, CD274, and ABRS. ABRS, atezolizumab + bevacizumab response signature; cDC2, type 2 conventional dendritic cells; CR, complete response; PFS, progression-free survival; PD, progressive disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; ROC, receiver operating characteristic; sc_ABRS, single cell atezolizumab plus bevacizumab response signature; SD, stable disease; TAM, tumor-associated macrophage. Discussion In this comprehensive single cell analysis of the immune landscape in advanced HCC, we identified immune cell populations and molecular signatures associated with favorable outcomes, particularly an activated TME characterized by tumor-specific cytotoxic T cells, cytokine-enriched TAMs, increased cDC2 cells, and enhanced antigen presentation by TECs. This was further supported by findings from an independent cohort. Consistent with previous findings from the GO30140 and IMbrave150 trials,[133]^20 we observed that responders to atezolizumab–bevacizumab exhibited higher levels of exhausted and cytotoxic gene signatures compared with nonresponders. Upregulation of these genes is crucial for identifying patients likely to benefit from immune checkpoint inhibitor therapy in HCC. By inhibiting the PD-1/PD-L1 axis, these therapies can restore exhausted CD8^+ T cells and enhance their effector function.[134]^24 Although baseline levels of cytotoxic CD8^+ T cells were not significantly different, responders demonstrated enhanced clonal expansion, particularly within the C9 cluster of terminally differentiated CD8^+ T cells expressing GZMH and CX3CR1. This cluster aligns with CD8 TEMRA cells, which have been reported to exhibit patrolling activity in the antitumor response under atezolizumab–bevacizumab therapy.[135]^5 Beyond CD8^+ T cells, TAMs have a crucial role in modulating the response to atezolizumab and bevacizumab combination therapy. Spatial transcriptomic analysis of HCC surgical specimens revealed an immunosuppressive TME characterized by SPP1^+ TAMs interacting with cancer-associated fibroblasts, which limit immune cell infiltration and reduce anti-PD-1 efficacy.[136]^22 However, other studies have shown that PD-L1^+ CXCL10^+ TAMs are enriched in responders, suggesting a role in recruiting T cells into the TME.[137]^25 These conflicting results illustrate the multifaceted role of TAMs within the TME and emphasize the need for further investigation into the diverse TAM populations and their impact on treatment responses. We found a unique TAM subtype characterized by chemokines, such as CXCL2, CCL3, CCL4, and IL1B, which was enriched in responders. This TAM_activating_CCL^+ subtype closely interacts with T cells, suggesting a role in CD8^+ T cell recruitment. These features align with the inflammatory cytokine-enriched TAM subtype described by Ma et al.,[138]^26 characterized by expression of inflammatory cytokines, such as IL1B, CXCL1/2/3/8, and CCL3, which are involved in active immune cell recruitment and regulation during the tumor-associated inflammatory response. In addition, this TAM subtype exhibited a high myeloid inflammation signature, previously associated with improved PFS in patients treated with atezolizumab plus bevacizumab compared with atezolizumab monotherapy.[139]^20 However, additional research is required to elucidate the function of bevacizumab in modulating these TAMs to improve the antitumor response. In contrast to the findings of Cappuyns et al.,[140]^25 we observed minimal PD-L1 and CXCL10 expression in TAMs ([141]Figs. S6A–C), possibly because of limited myeloid cell numbers in our analysis or differences in patient characteristics. Our cohort primarily comprised advanced viral HCC cases (83.3%) with high-risk features (50%), whereas Cappuyns et al.[142]^25 investigated nonviral HCC (93%) with less advanced disease (32% with macrovascular invasion). These results suggest that TAM interactions in the TME vary by HCC etiology and stage, potentially influencing response to atezolizumab plus bevacizumab. Growing evidence suggests that TECs have a multifaceted role in the TME, not only promoting tumor angiogenesis, but also mediating immune regulation.[143]^27 In this study, we observed that TECs in responders were enriched for antigen-presenting pathways, indicating their active involvement in T cell priming and activation. However, we were unable to identify specific TEC subpopulations associated with treatment response because of the limited number of TECs in the biopsy samples. These findings underscore the importance of an immunologically activated TME that can effectively recruit and prime T cells, contributing to favorable responses to atezolizumab plus bevacizumab in advanced HCC. This study has several limitations. First, although RNA scope provided valuable single cell gene expression data, it lacked spatial resolution and protein quantification. Second, the small sample size restricted analysis of non-immune cells and differentiation between viral and nonviral HCC. In addition, we could not validate findings with other scRNA-seq datasets because of time and resource constraints, requiring cautious interpretation of our results. Third, although radiological reviews confirmed that most biopsies were representative, they inherently capture only a small portion of the tumor, potentially underestimating intratumor heterogeneity. Lastly, longitudinal biopsies were not feasible because of bleeding risks in patients with HCC. Despite these limitations, our findings offer a detailed transcriptional view of advanced HCC treated with atezolizumab plus bevacizumab, supported by bulk transcriptome data from an independent cohort. In conclusion, single cell analysis of the TME in advanced HCC reveals that an activated immune microenvironment before treatment is crucial for a favorable response to combination therapy with atezolizumab plus bevacizumab. These insights highlight the potential for identifying patients with advanced HCC most likely to benefit from such treatment; however, further research with larger, longitudinal cohorts is needed to explore dynamic changes in the TME. Abbreviations Atezolizumab plus bevacizumab, atezolizumab and bevacizumab combination therapy; BCLC, Barcelona Clinic Liver Cancer; CCL, C–C motif chemokine ligand; CK, pan-cytokeratin; CMV, cytomegalovirus.CR, complete response; DC, dendritic cell; DEG, differentially expressed gene; EBV, Epstein–Barr virus; FDR, false discovery rate; FFPE, Formalin-fixed paraffin-embedded; GEM, gel bead in emulsion; GO, gene ontology; GSEA, gene set enrichment analysis; HCC, hepatocellular carcinoma; KEGG, Kyoto Encyclopedia of Genes and Genomes; KLF2, Kruppel-like factor 2; LSEC, liver sinusoidal endothelial cell; mIF, Multiplex immunofluorescence; mRECIST, modified Response Evaluation Criteria in Solid Tumors; NES, Normalized Enrichment Score; NK, natural killer; OS, overall survival; PCA, principial components analysis; PD-L1, programmed death-ligand 1; PD, progressive disease; pDC, plasmacytoid dendritic cell; PFS, progression-free survival; PR, partial response; ROI, regions of interest; ScRNA-seq, single-cell RNA sequencing; scTCR-seq, single-cell T cell receptor sequencing; SD, stable disease; SNN, shared nearest neighbor; TAM, tumor-associated macrophage; Tcm, central memory T cell; TCR, T cell receptor; Tcyt, cytotoxic T cell; TEC, tumor-associated endothelial cell; Tem, effector memory T cell; TIL, tumor-infiltrating lymphocyte; TME, tumor microenvironment; TRM, tissue-resident memory T cell; UMAP, uniform manifold approximation and projection; UMI, unique molecular identifier. Financial support statement This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number:RS-2020-[144]KH088687). Authors’ contributions Conceptualization: Y-HP and WP. Methodology: JL, W-YP, Y-HP, MJG, BGS, SYH, IH. Formal analyses: JL, MJG, WK, DIC, KG, SYH, IH. Resources: JL, DHS, G-YG, DIC, KG. Writing - original draft: JL, MJG. Writing - review & editing: JL, MJG, Y-HP, W-YP, BGS, DHS, WK, G-YG, DIC, KG, SYH, IH. Supervision: Y-HP, W-YP, JHL, MSC. Project administration: Y-HP, W-YP, JHL, MSC. Funding acquisition: Y-HP, WK. All authors read and approved the final manuscript. Data availability statement The datasets used in this study are available from the corresponding authors upon reasonable request. All datasets and software used for analysis are listed in the CTAT table. Further inquiries can be directed to the corresponding authors. Conflicts of interest The authors declare no competing interests. Please refer to the accompanying ICMJE disclosure forms for further details. Footnotes Author names in bold designate shared co-first authorship Supplementary data to this article can be found online at [145]https://doi.org/10.1016/j.jhepr.2024.101304. Contributor Information Woong-Yang Park, Email: woongyang.park@samsung.com. Yong-Han Paik, Email: yh.paik@samsung.com. Supplementary data The following are the supplementary data to this article: Multimedia component 1 [146]mmc1.pdf^ (1.5MB, pdf) Multimedia component 2 [147]mmc2.docx^ (39KB, docx) Multimedia component 3 [148]mmc3.pdf^ (619.5KB, pdf) Multimedia component 4 [149]mmc4.pdf^ (5.9MB, pdf) References