Abstract Background Osteosarcoma is a highly aggressive cancer, and the efficacy of existing therapies has plateaued. Multiomics integration analysis can identify novel therapeutic targets for various cancers and therefore shows potential toward osteosarcoma treatment. This study aimed to leverage multiomics integration to develop a new risk model, characterizing the immune features of osteosarcoma to uncover novel therapeutic targets. Methods Metabolomics profiling was conducted to identify key metabolites in osteosarcoma. Transcriptomic sequencing datasets were analyzed to identify prognostic genes related to key metabolic pathways and develop a prognostic risk model. Patients were then divided into high-risk and low-risk groups with distinct clinical outcomes based on the risk model. The single-sample gene set enrichment analysis, Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) algorithm, and xCell algorithms were used to evaluate the immune cell infiltration and activity. Single-cell RNA sequencing was used to explore cell-to-cell interactions within the tumor microenvironment. In vitro coculture functional assays were performed to validate the role of macrophage migration inhibitory factor (MIF) in macrophage polarization and chemotaxis. In vivo studies were used to evaluate the effectiveness of MIF inhibition in combination with immune checkpoint blockade in murine models. Results Elevated lactate levels in osteosarcoma patients correlated with poorer overall survival. We identified SLC7A7 and CYP27A1 as prognostic lactate metabolism genes and developed a risk model to stratify patients into high-risk and low-risk groups with distinct outcomes. Bioinformatics analyses highlighted the differences in immune infiltration patterns and activity between the groups. Notably, the infiltration and phenotype of macrophages varied significantly between the groups, and MIF was identified as a critical mediator in this process. In osteosarcoma cells, lactate regulated MIF expression through histone H3K9 lactylation. Combining the MIF inhibitor 4-IPP with a programmed cell death 1 (PD-1) monoclonal antibody treatment demonstrated a significant antitumor effect. Conclusion MIF acts as a novel therapeutic target by regulating macrophage polarization and chemotaxis. Lactate regulated MIF expression through histone lactylation. Targeting MIF holds promise for enhancing the efficacy of anti-PD-1 treatment. Keywords: Immune modulatory, Macrophage, Immunotherapy, Bone Cancer __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * Immune checkpoint inhibitors show limited efficacy in osteosarcoma treatment, which was due to the immunosuppressive tumor microenvironment. WHAT THIS STUDY ADDS * Lactate was correlated with macrophage migration inhibitory factor (MIF) expression in osteosarcoma and combined treatment with MIF inhibitor and programmed cell death 1 (PD-1) demonstrated a significant antitumor effect in osteosarcoma. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * This study offered a rationale for combining MIF inhibitors with PD-1, which may influence the clinical trials and treatment for osteosarcoma. Introduction Osteosarcoma is a highly malignant bone tumor predominantly affecting children and adolescents. The application of neoadjuvant chemotherapy combined with surgical treatment significantly improves the 5-year overall survival rate of patients with osteosarcoma. However, the efficacy of these conventional therapies for osteosarcoma has reached a plateau, with no significant recent improvements. Approximately 35% of patients develop recurrence and metastasis during treatment, leading to an unfavorable prognosis with an overall 5-year survival rate of less than 25%.[47]^1 2 Consequently, new promising treatments are required to enhance the long-term survival of patients with osteosarcoma. Cancer treatment has been revolutionized in the last decade with the advent of immune checkpoint inhibitors (ICIs) that show remarkable clinical responses across various tumor types.[48]^3 4 Regrettably, several phase II trials on the programmed cell death 1 (PD-1) inhibitor pembrolizumab in metastatic or advanced osteosarcoma demonstrated that only approximately 5% of patients achieved a significant response, highlighting its limited effectiveness.[49]^5 6 Osteosarcoma has been characterized as a “cold” tumor because of insufficient immune infiltration and lack of tumor immunogenicity.[50]^7 These factors limit the potential for meaningful responses to ICIs alone. Furthermore, identifying reliable biomarkers to guide enhanced ICI treatment continues to be a challenge. Accumulating evidence highlights cancer as a metabolic malady and metabolic reprogramming in cancer and immune cells is therefore essential for modulating the antitumor immune response.[51]^8 This modulation occurs through releasing metabolites that can affect immune molecule expression and effector function execution.[52]^9 The integration of metabolomics with other multiomics approaches, such as transcriptomics, has been widely adopted to unravel the intricate and key immune interactions within the tumor microenvironment (TME).[53]10,[54]12 This comprehensive approach has been proved to be valuable in revealing novel biomarkers that may serve as targets to enhance the efficacy of ICI therapies.[55]^13 14 To characterize osteosarcoma immune features and discover biomarkers for improving immunotherapy, we employed a multiomics approach combining metabolomics, bulk transcriptomics, and single-cell transcriptomics. Metabolomics profiling revealed elevated lactate levels in patients with osteosarcoma, which correlated to poorer prognosis. Subsequently, we developed a risk model incorporating prognostic lactate metabolism-related genes (LMRGs) SLC7A7 and CYP27A1. The integration of bulk and single-cell transcriptomics data into our risk model identified disparities in immune cell infiltration, and notable differences were observed in macrophage polarization and chemotaxis, regulated by macrophage migration inhibitory factor (MIF). Lactate regulated MIF expression through histone H3K9 lactylation in osteosarcoma cells. Consequently, we found that combining the MIF inhibitor 4-IPP with a PD-1 monoclonal antibody exerted a notable antitumor effect. This study offers new insights into the immune heterogeneity of osteosarcoma and may inform more targeted treatment strategies. Methods Serum non-targeted metabolomics analysis Previously, we applied serum non-targeted metabolomics using the ultrahigh-performance liquid chromatography coupled with high-resolution mass spectrometry platform to assess samples from 65 patients with osteosarcoma and 30 healthy adults.[56]^15 Metabolomics profiling in this study was based on further analysis of those data. Partial least squares discriminant analysis and metabolic pathway enrichment analysis were performed using MetaboAnalyst (widely used web-based platform dedicated for comprehensive metabolomics data analysis, interpretation and integration with other omics data. Website: [57]https://www.metaboanalyst.ca/. Bulk transcriptome sequence and public bulk transcriptome dataset download We used proprietary transcriptome data comprizing 45 osteosarcoma tissue samples and 25 paired peri-osteosarcoma tissue samples, which had been collected and processed at the First Affiliated Hospital of Sun Yat-sen University. We defined this as the Sun Yat-Sen Cohort (SYS cohort). For transcriptome sequencing, TRIzol was used to extract RNA from tumor tissues. For the qualified total RNA samples, 1–3 µg of total RNA was used as the starting material for each sample to construct a transcriptome sequencing library. Sequencing was performed using the BGISEQ-500 platform by running a paired-end sequencing program (PE), and 150 bp PE sequencing reads were obtained. For public bulk transcriptome datasets, the Therapeutically Applicable Research to Generate Effective Treatment-Osteosarcoma (TARGET-OS) dataset (n=85) was obtained from the UCSC Xena browser ([58]http://xena.ucsc.edu/). The Gene Expression Omnibus (GEO) dataset [59]GSE21257[60]^16 (n=53) was downloaded from the National Center for Biotechnology Information ([61]https://www.ncbi.nlm.nih.gov/). The Chromatin Immunoprecipitation sequence (ChIP-seq) and CUT&Tag data[62]17,[63]24 of histone lactylation was also downloaded from GEO. Single-cell transcriptome sequence data download and processing A single-cell RNA-sequencing (scRNA-seq) dataset comprizing six treatment-naïve osteosarcoma samples was obtained from GEO datasets ([64]GSE152048).[65]^25 Quality control (QC) and subsequent analysis were conducted using the “Seurat” R package (a widely used and powerful R package designed for QC, analysis, exploration and of scRNA-seq data) following standard protocols. Cells with <300 genes or >10% mitochondrial content were excluded as were genes detected in less than three cells. After filtering out housekeeping and mitochondrial genes, the study encompassed 25,640 cells. We pinpointed the 2,000 most variable genes for data normalization. Principal component analysis was applied to distil the data into 30 principal components, and the Harmony package was used to mitigate batch effects. Cells were visualized in a two-dimensional space via t-distributed stochastic neighbor embedding with the uniform Manifold Approximation and Projection (uMAP) algorithm. Seurat’s FindAllMarkers function identified signature genes for each cluster, aiding in cell type characterization. The FindAllMarkers function was employed with a logarithmic fold change threshold of 0.25 (logfc.threshold=0.25) and set to identify only positive markers (only.pos=TRUE) to detect markers for each cluster. CopyKAT (a widely-used and powerful tool identifying genome-wide aneuploidy in single cells, which can separate tumor cells from normal cells, and tumor subclones rapidly using high-throughput scRNA-seq data) was used to assess the benign or malignant nature of cells. Prognostic LMRG identification We used the term “lactic” as the search keyword in the Molecular Signatures Database (MSigDB; [66]https://www.gsea-msigdb.org/gsea/msigdb/index.jsp) and determined five priority lactate related gene (LRG) sets, namely, Gene Ontology Biological Process (GOBP) lactate metabolic process, human phenotype (HP) increased serum lactate, HP lactic acidosis, HP lactic aciduria, and HP severe lactic acidosis. All gene sets used in this research were listed in [67]online supplemental table 1. After deleting duplicates, 284 LMRGs were identified. The differential expression of LMRGs was assessed between normal and tumor groups in the SYS cohort using the “limma” package in R, with significance defined as p<0.05 and |log2FC|≥0.50. Significant differentially expressed LMRGs underwent subsequent univariate Cox regression to ascertain their association with overall survival. Variables with p<0.05 were evaluated by Least Absolute Shrinkage and Selection Operator (LASSO) analysis via the “glmnet” package (a widely used and convenient package that fits generalized linear and similar models), identifying candidate genes with the optimal penalty parameter λ determined by the 1—SE criterion. Finally, multivariate Cox analysis was used to determine the prognostic LMRGs for the construction of the optimal lactate metabolism-related risk model based on a minimum Akaike information criterion. Lactate metabolism risk model construction and related immune landscape analysis Risk scores were calculated by aggregating the expression levels and the corresponding coefficient of each prognostic LMRG. Risk scores were calculated using the following formula that incorporates the expression levels and the corresponding coefficient of two key genes: Risk score=SLC7 A7 × (−0.2948) + CYP27 A1 × (−0.3376). The “survminer” package (a widely used and powerful package that provides functions for facilitating survival analysis and visualization) was used for survival analysis and visualization. The risk group was stratifying patients into low-risk and high-risk groups according to the risk score. In addition, time-dependent receiver operating characteristic curves were used to assess the efficacy of the model in predicting 2-year, 3-year, and 5-year survival for patients. The [68]GSE21257 cohort served as an external validation set for the generalizability. The levels of cancer-related hallmarks raised by Hanahan and Weinberg,[69]^26 such as “Cell cycle,” “Hypoxia,” and “Epithelial–mesenchymal transition,” were quantified using the single-sample gene set enrichment analysis (ssGSEA) algorithm (a widely used and comprehensive tool to characterize cell state in terms of the activity levels of biological processes and pathways rather than through the expression levels of individual genes); this analysis was based on transcriptome data and gene sets from MSigDB and published literature. The ESTIMATE algorithm was employed to assess the infiltration ratio of tumor, immune, and stromal cells across all samples. In addition, the ssGSEA algorithm was used to evaluate the abundance of 29 immune cells and immune-related molecules. Then, the components of TME cells were analyzed using the xCell algorithm as previously reported. Intercellular communication analysis was conducted using the CellChat package (a widely used and convenient R package designed for inference, analysis, and visualization of cell-cell communication from single-cell data). Cancer Single-cell State Atlas (CancerSEA) was performed to assess 14 functional states of single-cell data from cancer cells.[70]^27 These states encompass stemness, invasion, metastasis, proliferation, epithelial–mesenchymal transition, angiogenesis, apoptosis, cell cycle, differentiation, DNA damage, DNA repair, hypoxia, inflammation, and quiescence. Immunohistochemistry Immunohistochemistry (IHC) staining was performed on 5 mm sections of paraffin-embedded osteosarcoma tissue samples. Briefly, slides were deparaffinized, treated for antigen retrieval, and then catalase-inactivated with 3% H[2]O[2]. Slides were then incubated overnight at 4°C with primary antibodies, including anti-CYP27A1 (1:1000, Cat. 14 739–1-AP, Proteintech), anti-SLC7A7 (1:1000, Cat. ab126785, Abcam), anti-MIF (1:500, Cat. 87501, Cell Signaling Technology), anti-CD68 (1:1000, Cat. ab303565, Abcam), anti-CD206 (1:2000, Cat. 81 525–1-RR, Proteintech), and anti-CD8α (1:400, Cat. 85 336S, Cell Signaling Technology). Antirabbit/mouse–horseradish peroxidase conjugate was then added as the secondary antibody, and immunoreactive proteins were detected using 3,3ʹ-diaminobenzidine. Finally, sections were counterstained with hematoxylin. The score for MIF, SLC7A7, and CYP27A1 staining was based on the integrated staining intensity and the proportion of positive cells. IHC-positive staining of CD68, CD206, and CD8α was used for the calculation of positive cell numbers per high field. All percentages/numbers of positive cells were expressed as the average of five randomly selected microscopic fields. Immunofluorescence Cells were seeded in chamber slides, then fixed with 4% paraformaldehyde, and permeabilized with 0.1% Triton X-100 for 15 min. Cell samples were then blocked with 5% bovine serum albumin (BSA) for 1 hour at 25°C, followed by incubation with primary antibodies overnight at 4°C, including anti-iNOS (1:500, Cat.18 985–1-AP, Proteintech) and anti-Arg1 (1:800, Cat.66 129–1-lg, Proteintech). Slides were then washed three times with phosphate-buffered saline (PBS) and incubated with secondary antibodies for 30 min at 25°C. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI) for visualization. Digital images were taken using a fluorescence microscope (Nikon, Japan). Western blotting analysis Total cellular proteins were extracted with radio immunoprecipitation (RIPA) lysis buffer (Biosharp) supplemented with protease and phosphatase inhibitors. Equal amounts of cellular proteins were resolved via sodium dodecyl sulfate–polyacrylamide electrophoresis followed by electrotransfer onto the 0.2 µm polyvinylidene fluoride membrane for 45 min at 4°C and 100 V. Subsequently, membranes were incubated overnight at 4°C with the specific primary antibody (anti-MIF, 1:2000, Cat. ab175189, Abcam). After washing the membranes three times with TBST, membranes were incubated with the corresponding secondary antibodies. The immunoreactive signal was visualized using an enhanced chemiluminescence kit. RNA isolation, reverse transcription, and real-time PCR Total RNA from cultured cell lines was isolated using the TRIzol reagent (Takara), and complementary DNA was synthesized using primers and the Takara PrimeScript reverse transcription reagent kit. Real-time PCR was performed using the RT-qPCR SYBR Green Kit (Takara) with a 7500 Fast Real-Time PCR System (Applied Biosystem). GAPDH expression was used as an internal reference. The following primers were used: CD86-RE, 5ʹ-CTGCTCATCTATACACGGTTACC-3ʹ and 5ʹ-GGAAACGTCGTACAGTTCTGTG-3ʹ; CD163-RE, 5ʹ-TTTGTCAACTTGAGTCCCTTCAC-3ʹ and 5ʹ-TTTGTCAACTTGAGTCCCTTCAC-3ʹ; ARG1-RE, 5ʹ-TGGACAGACTAGGAATTGGCA-3ʹ and 5ʹ-CCAGTCCGTCAACATCAAAACT-3ʹ; IL10-RE, 5ʹ-GACTTTAAGGGTTACCTGGGTTG-3ʹ and 5ʹ-TCACATGCGCCTTGATGTCTG-3ʹ; iNOS-RE, 5ʹ-AGGGACAAGCCTACCCCTC-3ʹ and 5ʹ-CTCATCTCCCGTCAGTTGGT-3ʹ; and TNFa-RE, 5ʹ-CCTCTCTCTAATCAGCCCTCTG-3ʹ and 5ʹ-GAGGACCTGGGAGTAGATGAG-3ʹ. MIF-ChIP-RE 5ʹ- CGTCACAAAAGGCGGGACCACA-3ʹ and 5ʹ-CTTCCTGTCCCCTCCCGGCAAA-3ʹ ELISA assay The concentration of MIF, CXCL9 and CXCL10 in the culture media was detected using an ELISA kit (Cat.E-EL-H6170, Cat.E-EL-H6062, Cat. E-EL-H0050 Elabscience). Culture media was centrifuged at 1000 rcf for 10 min, and the ELISA assay was performed as per the provided protocol. The average of three independent experiments was used in the analysis. Macrophage generation and differentiation THP-1 cells were pretreated with 100 ng/mL Phorbol 12-myristate 13-acetate (PMA) for 24 hours (MedChemExpress, USA) to generate M0 macrophages. To mimic the generation of tumor-associated macrophages (TAMs), M0 macrophages were cocultured with osteosarcoma cells in a 24-well Transwell coculture system (0.4 µm pore size, Corning, USA), where 2.5×10^5 THP-1 cells were seeded in the bottom well while 2.5×10^5 osteosarcoma cells were seeded in the upper chamber. After 48 hours, cocultured macrophages were collected to obtain TAMs for further analysis. Phalloidin-staining assay After discarding media, cells were fixed with 4% paraformaldehyde for 15 min and then washed with PBS three times. Cells were then incubated with 0.1% Triton X-100 for 15 min to increase permeability. Finally, cells were stained with 200 nM FITC-Phalloidin, sealed with DAPI Fluoromount-G, and observed under a laser scanning microscope (Nikon, Tokyo, Japan). Migration assay THP-1 cells (1×10^5) were suspended in a serum-free medium and seeded into the upper chambers of 24-well inserts (8.0 µm), and osteosarcoma cells were added to the lower chambers. Cells were co-cultured for 48 hours, and migrated macrophages were fixed with 10% formaldehyde, stained with crystal violet, and counted under a microscope. Isolation and activation of peripheral blood mononuclear cells Peripheral blood mononuclear cells (PBMCs) of three healthy donors were isolated via Ficoll–Hypaque density gradient centrifugation and then cultured in Roswell Park Memorial Institute (RPMI)-1640 medium supplemented with 10% fetal bovine serum (FBS). In addition, T cells in PBMCs were activated by adding 25 µL/mL of human CD3/CD28 T-cell activator (Cat.10991, STEMCELL Technologies) in combination with 1.5 ng/mL IL-2 (Cat.791902, BioLegend) for 72 hours at 37°C. T-cell activation assay Macrophages were differentiated from THP-1 cells and precultured with osteosarcoma cells before coculturing with T cells. Following 48 hours of coculture, osteosarcoma cells were removed, and preactivated T cells were seeded in six-well plates at 1×10^6 cells per well. After an additional 48 hours of coculture, suspended T cells were harvested and resuspended in a staining buffer (Dulbecco's Phosphate-Buffered Saline, DPBS containing 3% FBS) along with the indicated antibodies: APC antihuman CD8 (Cat.344721, BioLegend), PE antihuman/mouse granzyme B (Cat.372207, BioLegend), and Brilliant Violet 605 antihuman interferon-gamma (IFN-γ) (Cat.502535, BioLegend), followed by flow cytometry analysis. T-cell proliferation assay Preactivated T cells were labeled with fluorescent violet dye and then cultured with macrophages for 1 week in RPMI-1640 medium with 10% FBS, 100 mM L-glutamine, and 100 mM sodium pyruvate. T cells in suspension were harvested and resuspended in the staining buffer (DPBS containing 3% FBS). The violet or mean fluorescence intensity was detected as an indicator of T-cell proliferation via flow cytometry. ChIP-qPCR assay The ChIP assays were performed using the Pierce Magnetic ChIP kit (Thermo Fisher Scientific, #26157) with Anti-L-Lactyl-Histone H3 (Lys9) (PTM-1419RM) antibody. In brief, cells were incubated with 1% formaldehyde in PBS for 10 min, then washed two times with PBS. Cells were lysed for 10 min at 4°C in lysis buffer with protease inhibitors, followed by centrifugation at 12,000 rpm for 10 min at 4°C to remove debris. Samples were incubated overnight at 4°C with the antibody. Immunoprecipitated complexes were enriched using protein A/G Sepharose. DNA from immunoprecipitates was amplified by PCR and analyzed quantitatively with qPCR. Animal models All animal experiments were conducted in accordance with established guidelines for the Use and Care of Laboratory Animals and approved by the After receiving approval from the Institutional Animal Care and Use Committee of Guangzhou LingFu TopBiotech (LFTOP-IACUC-2024–0069). All mice were randomly divided into per groups. The murine osteosarcoma K7M2 cell line was purchased from Zhongqiaoxinzhou Biotechnology (Shanghai, China). A tumor-bearing mouse model was established by inoculating 2×10^6 K7M2 cells with Matrigel matrix (Sigma-Aldrich E1270) into the right flank of 4-week-old female BALB/c mice. Macrophages were depleted via tail-vein injection, once every 3 days with 200 µL PBS (control) or clodronate liposome (5 mg/mL, Cat.CP-005–005, Liposoma, Holland). Mice were intraperitoneally administered a vehicle control (5% dimethyl sulfoxide/40% polyethylene glycol 300/5% Tween-80/45% saline) or 4-IPP at doses of 10 mg/kg once every 3 days. Mice were monitored every 3 days for 3 weeks. Five mice from each group were euthanized at random when tumors attained an approximately spherical shape. Subcutaneous and xenograft tumors were harvested, photographed, and weighed. Tumor size was calculated using the formula width[71]^2×length × π/6 as described previously.[72]^28 Statistical analysis Statistical analyses and graph visualization were conducted using R V.4.2.1 ([73]http://www.r-project.org) or GraphPad Prism (V.9.0). All experiments were performed in triplicate, and representative data from one experiment are presented. Statistical analyses were conducted using Student’s t-test and one-way analysis of variance with Dunnett’s multiple comparisons test or Tukey’s multiple comparisons test. Overall survival was analyzed using Kaplan-Meier (KM) analysis and compared using the log-rank test. Differences with p<0.05 were considered significant. Results Elevated lactate predicts poor prognosis in patients with osteosarcoma To investigate prognostic metabolic signature of osteosarcoma, we analyzed the metabolomics data of serum samples from osteosarcoma patients and healthy individuals in our previous study.[74]^15 Integrating clinical information from the patients, we identified significant differences in serum metabolite profiles between patients with osteosarcoma and healthy adults ([75]figure 1A), with lactate showing the highest Variable Importance in Projection (VIP) score among 120 analyzed metabolites ([76]figure 1B). The pathway enrichment analysis revealed lactate metabolism pathways—particularly the Warburg effect, pyruvate metabolism, and gluconeogenesis—were significantly enriched in patients with osteosarcoma ([77]figure 1C). Serum lactate levels significantly elevated in patients with osteosarcoma and positively correlated with tumor size (R^2=0.3379, p<0.0001) ([78]figure 1D and E). Patients with elevated lactate levels had significantly reduced 5-year overall survival compared with those with lower levels ([79]figure 1F). Multivariate analysis confirmed that the serum lactate level was an independent prognostic indicator for adverse outcomes in patients with osteosarcoma ([80]figure 1G). Further quantification of lactate in fresh tumors and adjacent tissues from a cohort of 30 patients with osteosarcoma revealed a significant increase in lactate within tumor tissues (p<0.001) ([81]figure 1H). Similarly, lactate levels positively correlated with tumor size (R^2=0.4074, p=0.0001; [82]figure 1I). Multivariate Cox regression analysis identified hypoxia as a significant risk factor for overall survival among various hallmarks of cancer defined by both Hanahan and Weinberg in the TARGET-OS dataset ([83]figure 1J). Consistently, GSEA analysis revealed that glycolysis—a lactate metabolism-related pathway—was enriched, whereas oxidative phosphorylation was downregulated in osteosarcoma tissues compared with adjacent tissues based on the SYS cohort ([84]figure 1K). These results suggested that elevated lactate levels serve as a prognostic indicator and reflect enhanced lactate metabolism in patients with osteosarcoma. Figure 1. Elevated lactate levels in patients with osteosarcoma correlate with poor prognosis. (A) Scores plot from orthogonal partial least squares discriminant analysis showing differences in serum metabolite patterns between patients with osteosarcoma and healthy adults. (B) VIP scores indicate the serum metabolite differences between osteosarcoma patients and healthy adults. (C) Enrichment analysis of serum metabolites between patients with osteosarcoma and healthy adults. (D) Comparison of serum lactate levels between patients with osteosarcoma and healthy adults. (E) Correlation between serum lactate levels and tumor size. (F) Overall survival analysis was generated for patients stratified according to serum lactate levels, grouping based on the median of lactate (G) Multivariate analysis of prognostic factors for overall survival in patients with osteosarcoma. (H) Comparison of lactate levels in osteosarcoma tumors and adjacent tissues using a colorimetric assay (n=30). (I) Correlation analysis between the lactate concentration in osteosarcoma tissues and tumor size. (J) Multivariate Cox regression analysis among various hallmarks of cancer defined by both Hanahan and Weinberg in the TARGET-OS dataset. (K) GSEA analysis of glycolysis and oxidative phosphorylation pathways between the tumor and normal tissues based on the SYS cohort. Data are presented as the mean±SD, ***p<0.001 and ****p<0.0001, by Student’s t-test (D and H). CMPF, 2-(2-carboxyethyl)-4-methyl-5-propylfuran-3-carboxylic acid; HA, healthy adult;EMT, epithelial–mesenchymal transition; GSEA, gene set enrichment analysis; NES, normalized enrichment score; OPLS-DA, orthogonal partial least squares discriminant analysis; OS, osteosarcoma; SYS cohort, Sun Yat-Sen Cohort; TARGET-OS, Therapeutically Applicable Research to Generate Effective Treatment-Osteosarcoma; VIP, variable importance in projection. [85]Figure 1 [86]Open in a new tab The development and validation of a risk score model based on LMRGs To further investigate the role of lactate metabolism in osteosarcoma prognosis, we constructed a prognostic model prioritizing LMRG. First, five LMRG sets, comprizing 284 genes, were collected from the MSigDB database. Principal component analysis using these genes successfully distinguished osteosarcoma tissues from normal controls in the SYS cohort ([87]online supplemental figure S1A). Among these genes, 68 LMRGs exhibited significant differential expression ([88]figure 2A and [89]online supplemental figure S1B). Univariate Cox regression analysis identified eight genes significantly associated with overall survival in the TARGET-OS dataset ([90]figure 2B, [91]online supplemental figure S1C and D). LASSO regression combined with multivariate Cox analysis further narrowed down the candidate genes, identifying CYP27A1 and SLC7A7 as the most suitable genes for constructing a lactate metabolism-related prognostic risk score model ([92]figure 2C). KM analysis further confirmed that patients with low expression levels of SLC7A7 and CYP27A1 were associated with poor overall survival ([93]figure 2C and [94]online supplemental figure S1E). IHC staining of 62 osteosarcoma specimens from our department consistently demonstrated that lower expression of these genes correlated with poorer 5-year overall survival and lung metastasis-free survival ([95]figure 2D–K). Figure 2. Identification of prognostic lactate metabolism-related genes and construction of a risk model. (A) Volcano plots showing differentially expressed lactate metabolism-related genes between osteosarcoma and normal tissues in the SYS cohort, with important prognostic genes labeled. (B) Univariate Cox regression analysis in the TARGET-OS dataset identified eight prognostic genes associated with patient survival. (C) Multivariate Cox regression analysis and Kaplan-Meier survival analysis identified SLC7A7 and CYP27A1 retained to construct the risk model for TARGET-OS dataset. (D) Representative immunohistochemistry staining images of high and low SLC7A7 expression in tumor specimens scale bars: 100 µm (left) and 50 µm(right). (E and F) Analysis of overall survival (E) and lung metastasis-free survival (F), grouping based on median SLC7A7 expression levels. (G) Univariate analysis in the TARGET-OS dataset. (H) Representative immunohistochemistry staining images of high and low CYP27A1 expression in tumor specimens scale bars: 100 µm (left) and 50 µm(right). (I and J) Analysis of overall survival (I) and lung metastasis-free survival (J) based on CYP27A1 expression levels. (K) Multivariable analysis in the TARGET-OS dataset. (L) KM and receiver operating characteristic analysis for predicting 2-year, 3-year, and 5-year overall survival based on the stratified risk model in the TARGET-OS dataset. (M) KM and receiver operating characteristic analysis for predicting 2-year, 3-year, and 5-year overall survival based on the stratified risk model in the [96]GSE21257 cohort. Data are presented as the mean±SD, by Log-rank test (C, E, F, I, J, L and M). FC, fold change; FPR, false positive rate; KM, Kaplan-Meier; SYS cohort, Sun Yat-Sen Cohort; TARGET-OS, Therapeutically Applicable Research to Generate Effective Treatment-Osteosarcoma; TPR, true positive rate. [97]Figure 2 [98]Open in a new tab We then integrated clinical parameters, including age, gender, tumor site, and metastasis status, alongside the expression levels of SLC7A7 and CYP27A1, into multivariable Cox regression analyses. The analysis revealed that the risk score exhibited the highest hazard ratio (HR=6.37, p=0.002), demonstrating its prognostic value for patient survival outcomes ([99]figure 2K). Using the optimal cut-off value, patients were classified into low-risk and high-risk groups. In the TARGET-OS dataset, low-risk patients exhibited significantly better survival, with area under the curve (AUC) values of 0.68, 0.73, and 0.76 for 2-year, 3-year, and 5-year survival predictions, respectively ([100]figure 2L). Similarly, in the [101]GSE21257 cohort, application of the same formula successfully stratified patients into high-risk and low-risk groups, with survival analysis indicating significantly better survival in the low-risk group ([102]figure 2M). The AUC values in this cohort were 0.69, 0.68, and 0.69 for 2-year, 3-year, and 5-year survival, respectively, further validating the model’s prognostic accuracy ([103]figure 2M). Furthermore, the risk model exhibited prognostic power for overall survival across several cancer types among the 33 pancancer types, including Lung adenocarcinoma (LUAD, p=0.026; [104]Online supplemental figure S2A), Sarcoma (SARC, p=0.096; [105]Online supplemental figure S2B), Adrenocortical carcinoma (ACC, p=0.022; [106]Online supplemental figure S2C), Mesothelioma (MESO, p=0.016; [107]Online supplemental figure S2D), Lymphoid Neoplasm Diffuse Large B-cell Lymphoma (DLBC, p=0.012; [108]Online supplemental figure S2E), and Skin Cutaneous Melanoma (SKCM, p=0.00025; [109]Online supplemental figure S2F). These findings suggest that our LMRG-based model has prognostic potential for osteosarcoma and possibly other cancer types. Distinct immune microenvironment features in LMRG-derived osteosarcoma risk groups To dissect the distinct immune microenvironmental traits associated with different risk groups in osteosarcoma, we performed gene set variation analysis (GSVA) for pathway enrichment. Risk scores were negatively correlated with pathways involved in lymphocyte-mediated immune regulation, immune response initiation, B cell immune modulation, lymphocyte and macrophage chemotaxis, effector CD8+ T cell activation, T-cell cytotoxicity regulation, T-cell differentiation, macrophage inflammatory cytokine production, and T-cell costimulation in the SYS cohort ([110]online supplemental figure S3A). Similarly, in the TARGET-OS and [111]GSE21257 cohorts, these immune-related pathways showed inverse correlation with risk scores and were more enriched in the low-risk group than in the high-risk group ([112]online supplemental figure S3B and C). In addition, ESTIMATE scores revealed that the high-risk group had significantly lower estimates for immune, stromal, and overall ESTIMATE scores (p<0.001) while the tumor purity was significantly higher (p<0.001), in contrast to the low-risk group ([113]figure 3A). Similar results were observed in the [114]GSE21257 cohort ([115]online supplemental figure S4A). Using the ssGSEA algorithm, we scored 29 immune-related gene sets across samples, which reflects the diverse immune cell populations and their functional activities. The analysis showed that as risk scores decreased, the abundance of various immune cells—including plasmacytoid dendritic cells (DCs), immature DCs, B cells, and CD8^+ T cells—increased significantly, along with enhanced immune response activity ([116]figure 3B). This pattern was consistently observed in both the SYS and TARGET-OS datasets ([117]figure 3C; [118]Online supplemental figure S4B). In addition, xCell deconvolution analysis revealed a notable enrichment of macrophages and monocytes, which were the most differentially abundant immune cells in the low-risk group ([119]figure 3D). Macrophage infiltration was higher in the low-risk group within the [120]GSE21257 cohort ([121]online supplemental figure S4C). KM survival analysis demonstrated that the higher macrophage infiltration in osteosarcoma tissues was associated with improved survival outcomes in both the TARGET-OS and [122]GSE21257 cohorts ([123]figure 3E and F). Figure 3. Characterization of the immune microenvironment disparities associated with the LMRG-based risk model. (A) Comparison of ESTIMATE scores between high-risk and low-risk groups. (B and C) ssGSEA analysis of immune cell infiltration and immune function activity in the SYS and TARGET-OS datasets. (D) Comparison of the immune cell relative abundance between high-risk and low-risk groups in the TARGET-OS dataset using xCell algorithms. (E and F) Macrophage infiltration on patient survival in the TARGET-OS dataset, grouping based on median macrophage infiltration score. (G) Representative immunohistochemistry staining images of high and low CD68+ macrophage infiltration in tumor specimens, 100 µm (left) and 50 µm(right). (H and I) Analysis of overall survival (H) and lung metastasis-free survival (I) based on CD68+ macrophage infiltration levels, grouping based on median IHC score. (J) Comparison of proinflammatory TAM and anti-inflammatory TAM macrophage phenotypes between high-risk and low-risk groups in the TARGET-OS dataset. (K and L) Correlation analysis of risk scores with total macrophage abundance and proinflammatory TAM macrophage abundance in the SYS cohort and the TARGET-OS dataset. (M and N) Correlation of risk scores with expression of macrophage function, T-cell function, and immune-related genes in the SYS and TARGET-OS datasets. Data are presented as the mean±SD, by Student’s t-test (A and J), by Log-rank test (E and F). DCs, dendritic cells; aDCs, activated dendritic cells; APC, Antigen-presenting cell; CCR, C-C motif chemokine receptor; ESTIMATE, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data; FC, fold change; IFN, interferon; IHC, immunohistochemistry; LMRG, lactate metabolism-related gene; MHC, major histocompatibility complex; MSC, mesenchymal stem cell; NK, natural killer; pDCs, plasmacytoid dendritic cells; ssGSEA, single-sample gene set enrichment analysis; SYS cohort, Sun Yat-Sen Cohort; TAMs, tumor-associated macrophages; TARGET-OS, Therapeutically Applicable Research to Generate Effective Treatment-Osteosarcoma; Th1, T helper cell 1; Th2, T helper cell 2; TIL, tumor-infiltrating lymphocyte; Treg, regulatory T cell; Tcm, central emory memory T cell; Tem, effective memory T Cell. [124]Figure 3 [125]Open in a new tab To further examine the effect of macrophage infiltration on prognosis, we performed IHC on 62 osteosarcoma specimens from our institution ([126]figure 3G). These findings confirmed that patients with high levels of macrophage infiltration had significantly better 5-year overall survival and lung metastasis-free survival (p<0.001) ([127]figure 3H and I). Macrophage polarization analysis revealed a predominance of the anti-inflammatory TAMs phenotype in the high-risk group, whereas the proinflammatory TAMs phenotype dominated in the low-risk group[128]^29 30 ([129]figure 3J). Spearman correlation analysis in the SYS cohort demonstrated an inverse correlation between risk scores and macrophage infiltration, particularly with the proinflammatory TAMs phenotype (R=−0.68 for total macrophages, p<0.001; R=−0.47 for proinflammatory TAMs macrophages, p=0.0011) ([130]figure 3K), a trend that was also observed in the TARGET-OS and [131]GSE21257 datasets ([132]figure 3L; [133]Online supplemental figure S4D). The analysis further indicated that as risk scores decreased, the expression of genes associated with macrophage activation and antigen presentation increased, enhancing the capacity of macrophages to stimulate cytotoxic T lymphocytes and thereby strengthen antitumor immunity ([134]figure 3M and N, [135]online supplemental figure S4E). This was supported by the increased expression of T-cell activation and trafficking genes in the low-risk group, consistently observed across both cohorts ([136]figure 3M and N, [137]online supplemental figure S4E). Our findings revealed distinct macrophage profiles between risk groups. Specifically, the high-risk group exhibited reduced macrophage infiltration with predominant anti-inflammatory TAM polarization, while the low-risk group showed increased infiltration of proinflammatory TAMs. Insights from single-cell transcriptomics on immune microenvironment diversity between LMRG-based risk groups To further investigate the immune microenvironment differences between risk groups, we performed a single-cell transcriptomic analysis on data from six chemotherapy-naïve primary patients with osteosarcoma from the [138]GSE162454 dataset. After QC, a total of 25,640 cells were analyzed using Harmony for batch correction, CopyKAT for malignant cell prediction and UMAP for dimensionality reduction and visualization ([139]figures5A[140]D). We identified 10 distinct cell subtypes based on their signature gene expression: macrophages (C1QC, APOE, APOC1, C1QA, and C1QB), monocytes (FCN1, S100A9, S100A8, and CXCL8), DCs (CST3, HLA-DRA, and HLA-DQA2), natural killer (NK)/T cells (CD2, CD3D, CD3E, CD3G, and NKG7), osteoblastic cells (ALPL, RUNX2, and IBSP), osteoclasts (ACP5, CTSK, and MMP9), cancer-associated fibroblasts (COL1A1, ACTA2, and DCN), plasmocytes (IGHG1 and MZB1), endothelial cells (eg,FL7 and PLVAP), and B cells (MS4A1 and CD79A) ([141]figure 4A). Figure 4. Single-cell transcriptomic analysis of immune microenvironment disparities between LMRG-based risk score groups. (A) UMAP plot illustrating the identified 10 cell types in the [142]GSE162454 dataset. (B) Columns and pie charts show the distribution of cell types in high-risk and low-risk groups related to (A). (C) Gene set variation enrichment analysis pathway of differential enrichment between macrophages in high-risk and low-risk score groups. (D–O) Comparison of the functional status score of hypoxia (D), proliferation (E), metastasis (F), invasion (G), stemness (H), angiogenesis (I), EMT (J), inflammation (K), apoptosis (L), differentiation (M), DNA damage (N), DNA repair (O) in tumor cells between high-risk and low-risk groups based on the CancerSEA database. Data are presented as the mean±SD, by Student’s t-test (D–O). CAFs, cancer-associated fibroblasts; CancerSEA, Cancer Single-cell State Atlas; DC, dendritic cell; EMT, epithelial–mesenchymal transition; GO, Gene Ontology; LMRG, lactate metabolism-related gene; NK, natural killer; NES, normalized enrichment score; UMAP, uniform manifold approximation and projection. [143]Figure 4 [144]Open in a new tab Figure 5. Effects of osteosarcoma cells on macrophage phenotype polarization and migration capacity. (A and B) Interaction strength among various cell types within the osteosarcoma microenvironment. (C) Classification of osteosarcoma cell lines into high-risk and low-risk cells using the NTP method. (D) Schematic of a co-culture system between osteosarcoma cells and macrophages. (E) Phalloidin-FITC staining of macrophage morphology after coculture with 143B or U2OS cells. +143B: macrophages cocultured with 143B cells; +U2OS: macrophages cocultured with U2OS cells. Scale bar: 50 µm. (F) RT-qPCR analysis of mRNA levels for iNOS, CD86, TNFa, ARG1, IL10, and CD163 in macrophages (n=3). (G) Immunofluorescence detection of ARG-1 and iNOS protein expression in macrophages, scale bar: 100 µm. (H) Relative fluorescence quantification of ARG-1 protein expression (n=3). (I) Relative fluorescence quantification of iNOS protein expression. (J) Migration assay comparing the chemotactic effect of osteosarcoma cells on macrophages, scale bar: 200 µm. (K) Relative quantification of migrated macrophages. Control: medium alone in the lower chamber; +143B: macrophages cocultured with 143B cells; +U2OS: macrophages cocultured with U2OS cells (n=3). Data are presented as the mean±SD. *p<0.05, **p<0.01, ***p<0.001 and ****p<0.0001, by one-way ANOVA with Tukey’s multiple comparisons test (F, H, I and K). ANOVA, analysis of variance; CAFs, cancer-associated fibroblasts; DAPI, 4′,6-diamidino-2-phenylindole; IL, interleukin; DC, dendritic cell; mRNA, messenger RNA; NK, natural killer; ns, not significant; NPT, nearest template prediction; RT-qPCR, reverse-transcription quantitative PCR; TAMs, tumor-associated macrophages. [145]Figure 5 [146]Open in a new tab Patients were then categorized into high-risk score (n=3) and low-risk score (n=3) groups based on the risk scores. The high-risk score group exhibited a higher proportion of tumor cells and tumor-associated fibroblasts, whereas the low-risk score group had higher proportions of macrophages (predominantly), monocytes, B cells, DCs, and NK/T cells ([147]figure 4B). GSVA revealed that macrophages in the low-risk score group were enriched in pathways related to antigen processing, T-cell activation, and lymphocyte proliferation, suggesting a proinflammatory TAM phenotype. In contrast, macrophages in the high-risk group were enriched in pathways associated with an anti-inflammatory TAM phenotype, including leukocyte-mediated immunity suppression and vasculature development ([148]figure 4C). These findings were consistent with bulk transcriptomic analyses, which also revealed the elevated infiltration of immune cells, particularly proinflammatory TAMs, in the low-risk group. Conversely, the high-risk score group exhibited decreased immune cell infiltration, with a predominance of anti-inflammatory TAMs. Further analysis using the CancerSEA database revealed that tumor cells in the high-risk group exhibited greater activity in pathways related to hypoxia, invasion, stemness, differentiation, proliferation, metastasis, angiogenesis, apoptosis, DNA damage response, quiescence, and inflammation, compared with those in the low-risk group ([149]figure 4D–O). Both bulk and single-cell sequencing analyses consistently demonstrated that differences in LMRG-based risk scores are primarily reflected in macrophage infiltration and polarization, which may underlie the observed disparities in immune microenvironment and tumor aggressiveness between risk groups. Heterogeneity of osteosarcoma cell–macrophage crosstalk among LMRG-based risk groups To elucidate the interaction between different cell types, we analyzed the communication between macrophages and other cell types using the CellChat algorithm. The results revealed significant differences in signaling interactions between tumor cells and macrophages ([150]figure 5A and B and [151]online supplemental figure S5E). We next investigated whether tumor cells influence macrophage infiltration and polarization. Using the NTP method, we categorized osteosarcoma cell lines into groups resembling high-risk and low-risk score patients based on transcriptomic similarities.[152]^25 Cell lines such as HUO9, NOS1, 143B, G292, PSS008, and HSOS1 exhibited gene expression patterns similar to the high-risk group. While, cell lines including C396, HS860T, PSS131R, MG63, OS252, HOS, CAL72, SAOS2, and U2OS exhibited expression patterns typical of the low-risk group ([153]figure 5C). After 48 hours of coculture, macrophages exhibited distinct morphologies in response to the osteosarcoma cell lines they were exposed to. Macrophages cocultured with 143B cells developed elongated projections, a morphology associated with anti-inflammatory TAMs. Conversely, macrophages cocultured with U2OS cells showed a round morphology, characteristic of proinflammatory TAMs[154]^29 30 ([155]figure 5D and E). RT-qPCR analysis confirmed a gene expression profile consistent with anti-inflammatory TAM polarization in macrophages cocultured with 143B cells, characterized by increased levels of ARG1, IL10, and CD163 mRNA ([156]figure 5F). Conversely, macrophages cocultured with U2OS cells showed a proinflammatory TAM phenotype, evidenced by elevated expression of iNOS, CD86, and TNFa ([157]figure 5F). Protein expression supported these findings, with elevated expression of ARG-1 in the 143B coculture system and iNOS in the U2OS coculture system ([158]figure 5G–I). Notably, neither 143B nor U2OS cell lines expressed ARG-1 or iNOS ([159]online supplemental figure 6A). Transwell assays further revealed the contrasting chemotactic effects of osteosarcoma cells on macrophages, as 143B cells exerted a stronger inhibitory effect on macrophage migration compared with that of U2OS cells ([160]figure 5J and K). These cell communication analysis and coculture experiments consistently demonstrated the significant effect of diverse osteosarcoma cells on macrophage polarization and chemotaxis. Osteosarcoma-derived MIF is a key differential mediator in tumor–macrophage crosstalk between LMRG-based risk groups In order to identify the essential mediators of tumor–macrophage crosstalk. We analyzed the cytokine-mediated ligand–receptor interactions between tumor cells and macrophages in high-risk and low-risk groups. This study revealed significant disparities in MIF signaling, particularly in the MIF-(CD74+CXCR4) and MIF-(CD74+CD44) axes ([161]figure 6A). KM analysis showed that higher MIF expression was associated with poorer overall survival in patients with osteosarcoma ([162]figure 6B). Immunoblotting and ELISA assays further confirmed that high-risk osteosarcoma cell lines exhibited higher expression and secretion of MIF than those in low-risk cell lines ([163]figure 6C and D). To investigate the clinical relevance of MIF expression in osteosarcoma, we examined the expression of MIF in osteosarcoma tissues via IHC staining of paraffin sections. Patients with high MIF expression had significantly lower 5-year overall survival and lung metastasis-free survival (p<0.001) ([164]figure 6E). Spearman’s correlation analysis revealed a significant positive correlation between MIF levels and anti-inflammatory TAMs infiltration (R=0.31, p=0.0037), with no correlation observed for pro-inflammatory TAMs (R=0.16, p=0.14) ([165]figure 6F). We then knocked out MIF (SgMIF) in 143B cells and co-cultured them with macrophages. Macrophages showed distinct morphological changes depending on MIF expression ([166]figure 6G–I). The analysis of gene expression indicated increased levels of proinflammatory markers (iNOS, CD86, and TNFa) in macrophages co-cultured with MIF-knockout cells, whereas those treated with rhMIF expressed higher levels of anti-inflammatory markers (ARG1, CD163, and IL10) ([167]figure 6J). These results were further supported at the protein level, showing increased iNOS expression in MIF-knockout co-cultures and elevated ARG-1 levels following rhMIF treatment ([168]figure 6K–M). MIF is a multifunctional cytokine that regulates macrophage migration, participating in multiple immune responses through ligand–receptor interactions.[169]^31 We performed a migration assay using a co-culture model. Results demonstrated that MIF knockout alleviated tumor cell-mediated suppression of macrophage chemotaxis, while rhMIF treatment restored the inhibitory effect ([170]figure 6N and O). Overall, these results suggested that osteosarcoma cells promote anti-inflammatory macrophage polarization and inhibit chemotaxis through MIF secretion. Figure 6. Osteosarcoma-derived MIF contributes to anti-inflammatory TAM macrophage polarization and chemotaxis. (A) Analysis of ligand–receptor interactions between tumor cells and macrophages in high-risk and low-risk score groups. (B) Survival analysis of MIF expression in the TARGET-OS dataset, grouping based on median MIF expression. (C) MIF protein levels in osteosarcoma cell lines. (D) Quantification of MIF in osteosarcoma cell culture supernatants (n=3). (E) Representative immunohistochemistry staining images of high and low MIF expression in tumor specimens and survival analysis 100 µm (left) and 50 µm(right), grouping based on median IHC score. (F) Correlation analysis between MIF expression and macrophage phenotype in TARGET-OS patients. (G) Knockout of MIF in 143B cell lines. (H) MIF levels in 143B cells measured via ELISA (n=3). (I) Phalloidin-FITC staining of macrophage morphology post coculture with MIF-knockout 143B cells, with rhMIF supplementation; scale bar: 20 µm. (J) RT-qPCR analysis of iNOS, CD86, TNFa, ARG1, IL10, and CD163 in macrophages after coculture (n=3). (K) Immunofluorescence detection of ARG-1 and iNOS protein expression in macrophages, scale bar: 100 µm. (L) Relative fluorescence quantification of ARG-1 protein expression (n=3). (M) Relative fluorescence quantification of iNOS protein expression (n=3). (N and O) Migration assay assessing chemotaxis of macrophages (N) and quantification (O) (n=3), scale bar: 200 µm. Data are presented as the mean±SD. *p<0.05, **p<0.01, ***p<0.001 and ****p<0.0001, by Student’s t-test (H), by one-way ANOVA with Tukey’s multiple comparisons test (D, J, L, M and O), by Spearman correlation analysis (F), by Log-rank test (B and E). ANOVA, analysis of variance; DAPI, 4′,6-diamidino-2-phenylindole; IHC, immunohistochemistry; IL, interleukin; MIF, macrophage migration inhibitory factor; mRNA, messenger RNA; RT-qPCR, reverse-transcription quantitative PCR; TAM, tumor-associated macrophage; TARGET-OS, Therapeutically Applicable Research to Generate Effective Treatment-Osteosarcoma. [171]Figure 6 [172]Open in a new tab Lactate regulates MIF expression through histone lactylation To further investigate the role of lactate in the immune microenvironment, we first explored the effect of lactate on MIF expression in osteosarcoma cells with 2-Deoxy-D-glucose (2-DG, glycolytic inhibitor) treatment. Treatment with 2-DG significantly suppressed MIF expression in osteosarcoma cells and reduced the level of protein lactylation ([173]figure 7A and B). Histone lactylation is a vital mechanism by which lactate regulates gene expression.[174]^32 We further investigated whether lactate modulates MIF expression in osteosarcoma through histone lactylation. After exploring the ChIP and CUT&Tag data related to histone lactylation from the GEO database, we found that lactylation at histone H3K9 (H3K9la) was enriched at the region of MIF ([175]online supplemental figure S6B), indicating that histone lactylation could regulate MIF expression. Then we assess the level of H3K9la in osteosarcoma cells and the results showed 2-DG treatment significantly suppressed the H3K9la level ([176]figure 7C). ChIP-qPCR using anti-H3K9la antibody was used to explore the role of this histone-lactylation site in MIF expression and results exhibited H3K9la regulated MIF expression in 143B and G292 cell lines ([177]figure 7D). Moreover, we found the lactic acid level was positively correlated with MIF expression in patients with osteosarcoma ([178]figure 7E). Additionally, 2-DG treatment reduced the secretion of MIF by tumor cells ([179]figure 7F). In the tumor immune microenvironment, macrophage-derived CXCL9 and CXCL10 play a crucial role in recruiting CD8^+ T cells, which are essential for antitumor immune responses and the efficacy of anti-PD-1 therapy.[180]33,[181]37 Then, using the co-culture model, we further examined the effect of osteosarcoma-derived MIF on the expression of CXCL9 and CXCL10 in macrophages. The results showed that MIF knockdown significantly upregulated the expression of CXCL9 and CXCL10, while supplementation with rhMIF suppressed their expression ([182]online supplemental figure S6C). We then used the co-culture model to investigate whether lactate-regulated osteosarcoma cells influence CD8^+ T cell function via macrophages. The results showed that osteosarcoma cells treated with 2-DG promoted CXCL9 and CXCL10 expression in macrophages ([183]figure 7G) and enhanced the expression of granzyme B and IFN-γ in CD8^+ T cells ([184]figure 7H). These results demonstrated that lactate promotes the MIF expression in osteosarcoma cells through histone lactylation modification, which suppresses the secretion of CXCL9 and CXCL10 in macrophages, resulting in impairment of CD8^+ T cell function. Figure 7. Lactate regulated MIF expression through histone H3K9 lactylation (A) RT-qPCR analysis of MIF mRNA expression levels in 143B and G292 cells treated with 2-DG (n=3). (B) MIF and protein lactylation levels in 143B and G292 cells treated with 2-DG measured by immunoblotting. (C) H3K9 lactylation levels in 143B and G292 cells treated with 2-DG measured by immunoblotting. (D) RT-qPCR analysis of MIF signal from ChIP (using anti-H3K9la antibody) in 143B cell (n=3). (E) Correlation of lactic acid level with MIF expression in tumor samples from SYS cohort (n=30). (F) MIF secretion in 143B and G292 cells treated with 2-DG (n=3). (G) CXCL9 (left) and CXCL10 (right) level in macrophage cocultured with 2-DG treated 143B cells measured by ELISA (n=3). (H) Flow cytometry analysis of the effect of 143B-co-cultured macrophages on T-cell granzyme B and interferon-γ expression levels (n=3). Data are presented as the mean±SD, *p<0.01, ***p<0.001 and ****p<0.0001, by Student’s t-test (A, F and G), by one-way ANOVA with Tukey’s multiple comparisons test (D and H), by Pearson correlation (E). ANOVA, analysis of variance; CHIP, chromatin immunoprecipitation; 2-DG, 2-Deoxy-D-glucose; IFN-γ, interferon-gamma; MIF, macrophage migration inhibitory factor; mRNA, messenger RNA; RT-qPCR, reverse-transcription quantitative PCR; SYS cohort, Sun Yat-Sen Cohort; SSC, side scatter. [185]Figure 7 [186]Open in a new tab MIF-mediated macrophage polarization regulates T-cell activation and infiltration in osteosarcoma Previous studies showed that macrophage polarization affects T-cell proliferation and activation. To further explore the role of MIF-mediated macrophage polarization in regulating T cell responses, we used a co-culture model. Macrophages were first co-cultured for 48 hours with SgNC 143B, SgMIF 143B, or rhMIF-supplemented SgMIF 143B osteosarcoma cells. Tumor cells were then removed, and PBMC-derived T cells were added for subsequent co-culture ([187]online supplemental figure S7A). The CellTrace assay demonstrated that T cell proliferation was partially restored when cocultured with macrophages previously exposed to SgMIF 143B cells, compared with those exposed to SgNC 143B cells. However, the addition of rhMIF significantly inhibited this proliferation ([188]online supplemental figure S7B). Flow cytometry further revealed increased expression of granzyme B and IFN-γ in T cells cocultured with SgMIF 143B–induced macrophages, which was significantly reduced on rhMIF treatment ([189]online supplemental figure S7C and D). In addition, immunostaining of osteosarcoma tissues revealed that patients with high MIF expression had significantly fewer tumor-infiltrating CD68^+ macrophages and CD8a^+ T cells, while markers associated with immunosuppressive macrophages, including CD206 and CD163, were notably increased ([190]online supplemental figure S7E-I). These results showed MIF-mediated macrophage polarization suppressed the function and proliferation of T cells. Antitumor effects of MIF inhibition combined with anti-PD-1 therapy in osteosarcoma To further explore the therapeutic potential of MIF in osteosarcoma treatment, we then used K7M2 syngeneic mice to investigate the role of MIF in the osteosarcoma immune microenvironment ([191]figure 8A). Mif knockout significantly reduced tumor weight and volume ([192]figure 8A–C). Immunofluorescence staining revealed the increased infiltration of CD8a^+ cells ([193]figure 8D) and F4/80^+ cells ([194]figure 8E), along with a decrease in CD206^+ cells ([195]figure 8F) in the TME following Mif deletion. Moreover, the expression of CXCL9 and CXCL10 was also increased in the Mif-knockout group ([196]figure 8G and H). We then administered the MIF inhibitor 4-iodo-6-phenylpyrimidine (4-IPP) (10 mg/kg) intraperitoneally to syngeneic mice every 3 days,[197]^38 which also led to a significant reduction in tumor weight and volume ([198]figure 8I–K). Flow cytometry confirmed the increased macrophage infiltration following MIF inhibition ([199]figure 8L and [200]online supplemental figure S7J and K). In addition, depleting macrophages with clodronate liposomes alongside 4-IPP treatment partially attenuated the antitumor effects of 4-IPP, highlighting the critical role of macrophages in mediating the efficacy of 4-IPP ([201]figure 8L). Given the established association between F4/80^+ and CD8^+ cell infiltration and improved outcomes in immunotherapy, we further examined the synergy between 4-IPP and anti-PD-1 therapy ([202]figure 8M–P). The combination treatment significantly enhanced therapeutic efficacy, demonstrated by the increased infiltration of CD8^+ and F4/80^+ cells ([203]figure 8Q and R), along with a decrease in that of CD206^+ cells ([204]online supplemental figure S8)([205]figure 8S). The expression of CXCL9 and CXCL10 was also increased ([206]figure 8T and U) in the TME. Overall, these results indicated that the combination treatment of 4-IPP and anti-PD-1 therapy significantly boosted the immune response in osteosarcoma. Figure 8. Antitumor effects of MIF inhibition and anti-PD-1 therapy in osteosarcoma. (A–C) General view, tumor volume, and tumor weight in K7M2 syngeneic tumors with Mif knockout (n=5). (D–H) IF staining of CD8a+, F4/80+, CD206+, CXCL9 and CXCL10 following Mif knockout (n=5). (I–K) General view, tumor volume, and tumor weight in K7M2 syngeneic tumors with 4-IPP or clodronate liposomes treatment (n=5). (L) Flow cytometry analysis of the macrophage infiltration ratio (n=5). (M) Schematic showing steps for anti-PD-1 treatment combining 4-IPP treatment. (N–P) General view, tumor volume, and tumor weight in K7M2 syngeneic tumors with 4-IPP and anti-PD-1 treatment (n=5). (Q–U) IF staining of CD8a+, CD68+, CD206+, and CXCL9 and CXCL10 cells following 4-IPP and anti-PD-1 treatment. Data are presented as the mean±SD, ns means no significant, **p<0.01, ***p<0.001 and ****p<0.0001, by one-way ANOVA with Tukey’s multiple comparisons test (C, D, E, F, G, H, K, L, P, Q, R, S, T and U), by Pearson correlation (E), two-way ANOVA with Tukey’s multiple comparisons test (B, J and O). ANOVA, analysis of variance; DAPI, 4′,6-diamidino-2-phenylindole; FOV, field of view; IF, immunofluorescence; 4-IPP, 4-iodo-6-phenylpyrimidine; MIF, macrophage migration inhibitory factor; PD-1, programmed cell death 1. [207]Figure 8 [208]Open in a new tab Discussion Metabolic reprogramming has emerged as a hallmark of cancer and is crucial for the survival and functional execution of both cancer and immune cells.[209]^9 39 40 Accumulating evidence highlights the role of metabolic reprogramming as a key determinant of cancer development and antitumor immune response.[210]^41 42 Lactate, a glycolysis byproduct, functions as a crucial carbon source and signaling molecule. Research has indicated a significant correlation between elevated lactate levels and increased angiogenesis, tumor growth, nodal and distant metastases, and poorer survival across various cancer types.[211]43,[212]45 Consistent with these findings, our study revealed that high lactate levels in patients with osteosarcoma were associated with larger tumors and poorer survival, emphasizing the role of lactate metabolism in osteosarcoma progression. LMRGs are essential for maintaining lactate homeostasis in the TME by regulating synthesis, catabolism, and transport within this.[213]^46 Here, we developed a risk model based on the prognostic LMRGs SLC7A7 and CYP27A1, serving as an independent survival predictor for patients with osteosarcoma. These findings suggested that exploring disparities based on the risk model may reveal new insights into the lactate metabolism network and highlight previously overlooked aspects of osteosarcoma progression. In the TME, cancer cells, T cells, NK cells, DCs, and macrophages all respond to extracellular lactate levels.[214]47,[215]50 This triggers intracellular signals that regulate cellular activities and significantly affect their infiltration. Lactate has been shown to induce ARG-1 expression in macrophages, promoting an anti-inflammatory TAM phenotype and suppressing NK cell function.[216]^48 Moreover, regulatory T cells rely on lactate uptake to maintain their proliferation and immunosuppressive functions. In addition, lactate inhibits T-cell receptor-triggered production of IFN-γ, TNF-α, and IL-2, thereby impairing the function of cytotoxic T lymphocytes. These findings suggest that the disparities associated with the LMRG-based risk score may reflect differences in immune cell infiltration and activity. In this study, we applied the risk model to differentiate patients with osteosarcoma into high-risk and low-risk groups with distinct clinical outcomes. The analysis of independent transcriptomic datasets revealed that the high-risk group had lower immune cell infiltration and immune activation than the low-risk group. The combination of bulk and scRNA transcriptomic analyses confirmed notable differences in macrophage abundance and polarization: the low-risk group had more macrophages with a proinflammatory TAM phenotype, whereas the high-risk group had fewer macrophages with an anti-inflammatory phenotype. These findings implied that disparities in macrophage infiltration and polarization probably contribute to the overall survival differences between the high-risk and low-risk groups. TAMs are crucial components in the TME.[217]^51 Reprogramming TAMs from the immunosuppressive anti-inflammatory TAM phenotype to the proinflammatory TAM phenotype is key to improving antitumor immunity.[218]^52 53 Macrophages modify their activation in response to microenvironmental signals such as cytokines and pathogen-associated molecular patterns. Tumor-secreted cytokines such as IL-4, IL-10, IL-13, and M-CSF activate transcription factors including STAT3/6, KLF2/4, and IRF3/5, which drive arginine-dependent metabolic pathways and macrophage polarization.[219]^54 It is worth noting that while our study highlights the immunosuppressive role of MIF via modulation of macrophage polarization, TAM in the TME exhibit substantial phenotypic and functional heterogeneity beyond the classical anti/proinflammatory classification.[220]^29 30 Future studies using single-cell transcriptomics or spatial profiling may help further resolve the complexity of TAM subsets and their dynamic roles in osteosarcoma progression and response to immunotherapy. Our study found notable differences in macrophage abundance and phenotype between the high-risk and low-risk groups, mainly because of the secretion of MIF by tumor cells. Osteosarcoma cells with high MIF levels promoted anti-inflammatory TAM polarization in macrophages and reduced their chemotactic activity, highlighting MIF as a key mediator in the immunomodulatory environment of osteosarcoma. MIF is a multifunctional cytokine that regulates cell migration, activation, and differentiation in both immune and non-immune cells.[221]^31 Elevated MIF levels are commonly found in various cancers, including osteosarcoma, and are associated with increased tumor burden, advanced grades, and poor prognosis.[222]^55 56 In brain tumors, MIF engages CD74 receptors on microglial cells, reducing interferon-γ production and shifting them from an anti-inflammatory TAM to an anti-inflammatory TAM phenotype. In addition, MIF supports macrophage survival and maintains homeostasis by upregulating the expression of inflammation resolution genes.[223]^57 Furthermore, monocyte-derived MIF drives alternative activation in mouse melanoma TAMs, and its inhibition reduces TAM-mediated and myeloid-derived suppressor cell-mediated immune suppression.[224]^58 Similarly, in the 4T1 murine model of breast cancer, tumor-derived MIF promotes myeloid-derived suppressor cell accumulation and immunosuppressive activity. In osteosarcoma, MIF promoted tumor cell proliferation and metastasis via the RAS/MAPK pathway.[225]^59 Our study extended this knowledge, demonstrating lactate promoted MIF expression through histone H3K9 lactylation. We also found that MIF expression not only induced anti-inflammatory TAM polarization in macrophages but also reduced their chemotactic activity in vitro and in vivo. Moreover, MIF-regulated TAM polarization influenced the infiltration of CD8^+ T cells through CXCL9 and CXCL10. These findings highlighted MIF as a critical factor in osteosarcoma, suggesting that targeting MIF could inhibit tumor growth and metastasis while fostering a supportive antitumor immune environment, representing a promising therapeutic strategy. ICIs have transformed the treatment landscape for chemoresistant malignancies, such as melanoma.[226]^60 A notable success of ICIs is achieving long-term remission even after treatment discontinuation, raising significant hope for potential cures in some patients. Given this, the use of immunotherapy to improve survival outcomes in osteosarcoma has become an attractive strategy. However, clinical trials of ICIs targeting the PD-1 pathway in advanced osteosarcoma have yet to yield the expected results. Our study supported these findings, showing that monotherapy with a PD-1 monoclonal antibody did not significantly reduce tumor growth. Previous studies have shown that the clinical benefits and prognostic outcomes of immunotherapy are largely influenced by the TME status of the patient.[227]^61 Tumors with low levels of tumor-infiltrating lymphocytes are referred to as “cold tumors” and respond poorly to ICIs.[228]^62 Our findings highlighted that knocking out Mif from murine tumor cells increased effector CD8^+ T-cell infiltration in the TME. This suggested that targeting MIF in osteosarcoma could enhance the effectiveness of immune checkpoint blockade. Supporting this, our in vivo studies combining 4-IPP, a MIF inhibitor, with a PD-1 monoclonal antibody produced a significant reduction in tumor growth, surpassing the effects of either agent alone. In addition, this combination approach increased effector CD8^+ T-cell infiltration in the TME. These results indicated that 4-IPP could enhance the antitumor efficacy of PD-1 blockade by improving immune cell penetration into the tumor, potentially offering a significant therapeutic advantage for patients with osteosarcoma with elevated MIF expression. Conclusions Overall, we developed an LMRG-based risk score that distinguishes patients with osteosarcoma into two groups with notable differences in survival outcomes. Our findings illuminated the disparities in TME within the groups, with a focus on immune cell infiltration and functional activity. We identified MIF-induced macrophage polarization and chemotaxis as crucial factors contributing to the differences between the groups and confirmed MIF as a potential target for enhancing ICI treatment. Supplementary material online supplemental file 1 [229]jitc-13-8-s001.jpg^ (1.9MB, jpg) DOI: 10.1136/jitc-2024-011091 online supplemental file 2 [230]jitc-13-8-s002.docx^ (15.6MB, docx) DOI: 10.1136/jitc-2024-011091 online supplemental file 3 [231]jitc-13-8-s003.docx^ (35KB, docx) DOI: 10.1136/jitc-2024-011091 Acknowledgements