Abstract Objective To enhance the therapeutic effect of atezolizumab on osteosarcoma (OS) by constructing a pH-responsive hydrogel nanocomplex (Gel@PLGA@FA) as a delivery platform for Shikonin. Methods First, Shikonin was initially employed to analyze the [28]GSE14359 dataset, leading to the identification of 28 differentially expressed genes (DEGs). Based on this, a risk score model was constructed and molecular dynamics simulations were performed to assess the binding ability between Shikonin and cyclin-dependent kinase 1 (CDK1). In addition, the in vitro antiproliferative effect of Shikonin on MG63 and Saos-2 OS cell lines and its selective toxicity on normal cells were assessed. In order to overcome the disadvantages of poor water solubility and normal cytotoxicity towards Shikonin, a complex loaded with Shikonin by pH-responsive intelligent hydrogel nanomaterials was synthesized and its anti-programmed death ligand-1 (PD-L1) therapeutic effect on OS cells was evaluated. Results Molecular dynamics simulation showed that Shikonin showed strong binding ability to CDK1, showing stable conformation, enhanced structural stability and other characteristics. In vitro experiments showed that Shikonin had a significant anti-proliferative effect on OS cells, while it had selective toxicity on normal liver, kidney and osteoblasts. The pH-responsive hydrogel nanomaterial (Gel@PLGA@FA) loaded with Shikonin showed good drug release characteristics at different pH conditions, especially in the tumor microenvironment to achieve controllable drug release. Combined use of Gel@PLGA@Shikonin@FA and atezolizumab effectively down-regulated CDK1 and PD-L1 expression, inhibited cell proliferation and promoted apoptosis, significantly enhancing the anti-PD-L1 therapeutic effect on OS cells. JC-1 staining experiments further confirmed that this combination therapy could perturb mitochondrial membrane potential and lead to stronger apoptosis. Conclusion This study reveals the unique mechanism of action of Shikonin as a potential anticancer drug and demonstrates the potential of pH-responsive hydrogel nanomaterials as efficient and safe delivery systems for targeted cancer therapeutics. The strategy of Gel@PLGA@Shikonin@FA combined with atezolizumab provides a new idea and experimental basis for OS treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-03742-2. Keywords: Osteosarcoma, Shikonin, Nanomaterial, Hydrogels, PD-L1, CDK1 Introduction Osteosarcoma (OS), the most common primary malignant bone tumor in adolescents, is characterized by high invasiveness and early metastasis [[29]1]. Despite advancements in surgical and chemotherapeutic techniques, the 5-year survival rate for patients with recurrent or metastatic disease remains at approximately 20% [[30]2], highlighting an urgent need for more effective treatment strategies. The advent of immune checkpoint inhibitors has brought breakthroughs in the treatment of solid tumors [[31]3]. However, the single-agent response rate of programmed death ligand 1 (PD-L1) inhibitors such as atezolizumab in OS remains extremely low. The core reason lies in the fact that OS is classified as a “cold tumor”-characterized by sparse T-cell infiltration in the tumor microenvironment and a strong immunosuppressive state, which makes it difficult to elicit an effective immune response [[32]4]. Shikonin, a natural naphthoquinone compound extracted from the traditional Chinese medicinal herb Lithospermum erythrorhizon [[33]5], possesses multiple biological activities including anti-inflammatory [[34]6], antioxidant [[35]7], and anti-tumor properties [[36]8]. In particular, it can improve the tumor microenvironment by downregulating immunosuppressive factors such as TGF-β [[37]9] and regulating the ratio of CD8⁺ T cells to Treg cells, thereby showing potential for combination immunotherapy [[38]3, [39]10]. However, its clinical application is limited by drawbacks such as poor water solubility and certain toxicity to normal cells. In recent years, the development of nanotechnology has provided new ideas for tumor combination therapy [[40]11]. PH-responsive hydrogel nanocomplexes are widely used in targeted drug delivery systems because of their unique tumor microenvironment response ability, good biocompatibility, and controllable drug release characteristics [[41]12]. Such materials are able to trigger structural changes in the acidic microenvironment of tumor tissue and achieve site-directed release of the drug, thereby improving therapeutic efficiency and reducing systemic toxicity [[42]13]. Li et al. [[43]14] used pH-sensitive polymeric micelles to load chemotherapeutic drugs to achieve effective accumulation and controlled release at the tumor site, significantly improving the therapeutic effect and reducing side effects. Based on this, the present study constructs a pH-responsive hydrogel nanocomposite (Gel@PLGA@FA) as a shikonin delivery platform, aiming to enhance the immunomodulatory effect of shikonin on OS through targeted delivery and controlled release, thereby improving the therapeutic efficacy of atezolizumab. Materials and methods Differential gene screening and functional analysis of Shikonin targeting osteosarcoma First, we normalized the OS-related gene data (3501 genes in total) from GEO database ([44]GSE14359) using GEOquery toolkit and intersected it with 84 potential targets of Shikonin predicted by prediction databases, and Venn diagrams were drawn to show the intersection. To identify differentially expressed genes (DEGs), we used the limma algorithm package to analyze [45]GSE14359 transcriptome data (log2 |FC| ≥ 1, P < 0.05) and used the ggplot2 package to draw volcano plots to visually display the distribution of DEGs. Subsequently, we performed multidimensional clustering and heatmap analysis of selected differential genes by the ComplexHeatmap package, thereby revealing heterogeneous gene expression profiles between different sample groups. Further combined with Venn diagram analysis, we identified a common set of genes between potential targets of Shikonin and OS-related genes. Finally, we performed GO functional enrichment and KEGG pathway enrichment analysis of these key target genes using clusterProfiler package to systematically annotate the functions of related genes and the biological processes involved. Establishment of a key target prognostic risk model for Shikonin targeting osteosarcoma By combining network pharmacology with transcriptomics, we selected 28 key targets of Shikonin acting on OS and applied them to a LASSO regression model (glmnet package and survival packages) to calculate risk coefficients and then construct a risk score model. Subsequently, we used the key genes included in this model to establish nomograms (rms and survival packages) for predicting the survival prognosis of OS patients. To assess the predictive accuracy of the model, we used the concordance index (C-index) and the calibration curve for validation. At the same time, the clinical utility of the model was further evaluated by decision curve analysis (DCA) to ensure its potential value in guiding clinical decision-making. Clinical relevance and prognostic analysis of key targets targeted by Shikonin in osteosarcoma The four key targets finally identified for Shikonin-targetied OS were validated for prognostic relevance with the survival analysis toolkit. Kaplan-Meier survival analysis was used to plot survival curves to reveal a significant association between the target gene and patient survival. Further, the regulatory relationships between core genes and clinicopathological characteristics (such as gender, metastatic status, etc.) were analyzed using multivariate regression models to determine their predictive value for disease progression. Finally, based on the core algorithm of CIBERSORT (CIBERSORT.R script analysis), using the CIBERSORTx website ([46]https://cibersortx.stanford.edu/), 22 immune cell markers were used to calculate cyclin-dependent kinase 1 (CDK1) immune infiltration in OS. Molecular dynamics assessment of Shikonin molecular binding to CDK1 Based on the CHARMM36 force field, we constructed the CDK1 crystal structure and the three-dimensional conformation of Shikonin, and completed the initialization setup of the complex system by generating topology files, using the TIP3P water model, and adding 0.15 M NaCl for ion neutralization. In order to eliminate steric hindrance, we first implemented the energy optimization of the steepest descent method of 5000 steps. Then, in order to reach the equilibrium state, we sequentially performed the equilibrium treatment of 10 nanoseconds each for the NVT ensemble at a temperature of 310 K (temperature controlled using the V-rescale method) and the NPT ensemble with a pressure of 1 bar (pressure controlled using the Parrinello-Rahman method). Subsequently, all-atom explicit solvent simulations were performed for a total of 70 nanoseconds with a time step of 2 femtoseconds, and trajectory frames were acquired every 10 picoseconds. At the analysis stage, we cluster trajectories using the gmx cluster tool to identify dominant binding conformations. At the same time, using the built-in tool in the Gromacs software package, a series of indicators were calculated to evaluate the system stability, including root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Gyrate), accessible surface area (SASA), and hydrogen bond formation (HBond). In addition, we calculated the binding free energy between Shikonin and CDK1 using MM/GBSA method in order to further understand the interaction between the two. Design and synthesis of pH responsive hydrogel nanomaterial complex Shikonin (50 mg, HY-NO882, MCE, China) was co-dissolved with the amphiphilic copolymer PLGA-PEG-NH[2] (100 mg) in 5 mL chloroform to prepare an oily phase solution. A 1% aqueous solution of polyvinyl alcohol (PVA) (50 mL) was also prepared, and the oil phase was evenly dispersed in the aqueous phase to form a microemulsion by probe sonication (20 kHz, power 300 W for 5 min). The emulsion was subsequently transferred to a three-necked flask and organic solvents were continuously volatilized for 4 h at 500 rpm under stirring. The resulting nanoparticles were purified by centrifuged three times at 8000 × g for 15 min using 100 kDa ultrafiltration tubes to finally obtain a concentrated suspension of Shikonin@PLGA-PEG-NH[2] nanoparticles. In the folic acid coupling step, 10 mg of folic acid was dispersed in 10 mL MES buffer at pH 5.5, 60 mg EDC and 100 mg NHS were added, and the carboxyl group was activated for 30 min at 37 ℃ with shaking. The activated mixture was subsequently mixed with the nanoparticle suspension described above and supplemented with borate buffer (pH 8.5) to a total volume of 20 mL, and the reaction was continued for 12 h at room temperature to complete conjugation. At the end of the reaction, unbound folic acid molecules were removed by centrifugation using 100 kDa ultrafiltration to finally obtain a Shikonin@PLGA@PEG@FA targeted drug delivery system. For construction of pH-responsive hydrogels, 2 g dextran (DEX) was first dissolved in 15 mL deionized water and 1.2 g NaIO[4] was added after stirring until complete dissolution and the reaction was performed for 6 h under nitrogen atmosphere protection. The reaction was subsequently stopped by adding 1.6 g glycerol and the product was transferred to a 10 kDa dialysis bag for 24 h dialysis, followed by freeze-drying and reconstitution in 20 mL deionized water. During magnetic stirring, 0.5 mL ethylenediamine was added to continue the reaction for 6 h, and the reaction solution was dialyzed again in a 10 kDa dialysis bag for 24 h, and finally freeze-dried and reconstituted in 10 mL deionized water to obtain hydrogel material with pH response characteristics. In order to characterize the structural features of the constructed composite system, scanning electron microscopy (SEM) was used to observe its micromorphology and confirm that it had a significant three-dimensional network structure; transmission electron microscopy (TEM) was also used to analyze the morphology of nanoparticles. Effective encapsulation of Shikonin and successful implementation of folic acid modification were assessed by zeta potential detection. In addition, dynamic light scattering (DLS) was used to measure particle size changes under different treatment conditions, further verifying drug loading and surface modification. Cell culture The human OS cell lines Saos-2 and MG63 were from Wuhan Servicebio (Wuhan, China), while the human renal tubular epithelial cell HK-2, human liver parenchymal cell L-02, and human osteoblast cell line hFOB1.19 were obtained from China Center for Type Culture Collection. All cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM, Gibco, Carlsbad, CA) containing 10% fetal bovine serum (FBS, PAN, Germany) and maintained in a 5% CO[2] incubator at 37℃. Cell counting kit-8 (CCK-8) proliferation assay OS cells were seeded in 96-well plates at a density of 2 × 10³ cells per well. After the cells completed adherent growth, the following treatments were administered: Shikonin (500 nM), PD-L1 inhibitor atezolizumab (10 µg/mL), targeted nanocomplex Gel@PLGA@Shikonin@FA at 10 µM, and the combination of Gel@PLGA@Shikonin@FA and PD-L1 inhibitor. After 6 h of treatment in each group, fresh complete medium was replaced and cells were maintained in a 37 ℃ incubator containing 5% CO[2] and tested at three time points: 24 h, 48 h, and 72 h. After reaching the designated time point, CCK-8 solution prepared with complete medium was changed to react for 2 h in the dark. Subsequently, absorbance values at 450 nm wavelength were measured using a full-wavelength microplate (Synergy H1, Bio Tek, USA) reader to assess the viability of cells under different treatment conditions. EdU proliferation assay First, treated OS cells were seeded in 6-well plates and grown to approximately 70% confluency under routine culture conditions. Subsequently, EdU solution (KTA2031, Abbkine) was added to the culture system at a final concentration of 10 µM and incubation was continued for 2 h to allow efficient integration of EdU into the DNA strand being synthesized. Following completion of labeling, cells were washed twice with PBS buffer to remove unincorporated EdU. Cells were then fixed in 4% paraformaldehyde for 15 min and subsequently permeabilized in 0.5% Triton X-100 in PBS for 10 min. According to the instructions for use of EdU detection Kit (KTA2031, Abbkine), add Apollo staining reaction solution and incubate for 30 min in the dark to make Apollo fluorescent dye click chemical reaction with labeled EdU to form a stable fluorescent complex. At the end of the staining reaction, cells were again washed twice with PBS and subsequently stained for nuclei using Hoechst 33,342 staining solution for 10 min. Finally, images were collected using fluorescence microscopy (CKX53, Olympus Corporation, Japan) for assessment of cell proliferation. Evaluation of uptake efficiency and cytotoxicity in osteosarcoma cells To systematically assess the uptake kinetics and biocompatibility of pH-responsive hydrogel nanodelivery systems, especially the uptake differences between the targeted modified Gel@PLGA@DiO@FA and the non-targeted form Gel@PLGA@DiO in OS cells, we used a DiO fluorescent probe labeling strategy. The intracellular uptake efficiency of the two nanocarriers was quantitatively analyzed by confocal laser scanning microscopy (LSM880, Carl Zessi, Germany) to detect the intracellular green fluorescence intensity (excitation wavelength 488 nm), so as to evaluate the effect of targeted modification on the cellular uptake ability. In addition, to comprehensively understand the biosafety of the constructed Gel@PLGA@Shikonin@FA thermosensitive hydrogel nanocomplex, we performed CCK-8 cell viability assays in a variety of normal human cell lines (including tubular epithelial cell HK-2, hepatic parenchymal cell L-02, and osteoblast hFOB1.19) to assess their concentration-dependent cytotoxic responses. We systematically assessed the biosafety of pH-responsive hydrogel nanosystems by calculating the half maximal inhibitory concentration (IC50) and determining the maximum non-toxic effect concentration. Quantitative reverse transcription PCR (qRT-PCR) According to the manufacturer ‘s instructions, we used TRIzol solution (Acuracy) to extract total RNA. Subsequently, reverse transcription and real-time quantitative PCR analysis were performed using PrimeScript RT Master Mix (Takara Bio) and SYBR Premix Ex Taq Kit (Takara Bio). All results for gene expression were normalized against GAPDH and relative quantitation was performed by the 2^−ΔΔCT method. Apoptosis by flow cytometry MG63 cells were seeded at a density of 2 × 10^5 cells per well in 6-well plates, and after complete attachment of the cells, the following treatments were administered: Shikonin (500 nM), PD-L1 inhibitor atezolizumab (10 µg/mL), Gel@PLGA@Shikonin@FA (10µM), and Gel@PLGA@Shikonin@FA combined with PD-1 inhibitor treatment groups, while DMSO treatment was used as the control group. All treatments were performed in the presence of complete medium for 24 h. At the end of treatment, cells from each group were collected and washed with pre-chilled PBS to remove residual medium. Subsequently, cells were digested with 0.25% trypsin without EDTA for 2 min at 37 °C to prepare single cell suspensions. According to the operating instructions of Annexin V-FITC/PI Apoptosis Detection Kit, cells were resuspended in 100 µL binding buffer, 5 µL Annexin V-FITC and 10 µL PI staining solution were added sequentially and incubated for 5 min in the dark. Following completion of the reaction, 400 µL of buffer was added to stop staining. Finally, the samples were detected by flow cytometry (Cytoflex, Beckman, USA), and the distribution of subsets at each stage of apoptosis was analyzed by FITC and PI dual fluorescence channels to assess the effect of different treatments on apoptosis. JC-1 assay for detecting changes in mitochondrial membrane potential First, 1 × 10^6 MG63 cells were seeded in confocal dishes for culture. The following day, confocal dishes were supplemented with 0.5 mL of JC-1 dye (C2005, Beyotime Bio) and Hoechst 33,342 stain solution (C0031, Solaibao) and incubated at 37℃ for 25 min. Afterwards, stained cells were observed and images were taken using confocal microscopy (LSM880, Carl Zessi, Germany). Statistical analysis All data were presented as mean ± standard deviation (Mean ± SD) from at least three independent experiments, each performed in triplicate. Statistical differences between groups were compared using unpaired Student’s t-test; differences between multiple groups were assessed using one-way or two-way analysis of variance (ANOVA), respectively. Differences were considered statistically significant when P < 0.05 and were marked as “*”; P < 0.05 were marked as “**”; P < 0.001 were marked as “***”; and P < 0.0001 were marked as “#”. Results Differential gene screening of Shikonin targeting osteosarcoma Through analysis of the [47]GSE14359 dataset, we identified a series of DEGs. First, DEGs identified in the [48]GSE14359 dataset (|LogFC| >1& P < 0.05) clearly revealed a large number of upregulated (n = 944) and downregulated (n = 775) genes by volcano plots (Fig. [49]1A). Next, a heatmap of the top 50 DEGs (Fig. [50]1B) demonstrates the differences in expression levels of these genes between OS and normal samples. To further explore the relationship between Shikonin and OS, we showed the intersection of DEGs in [51]GSE14359 with Shikonin targeted genes by Venn diagram (Fig. [52]1C) and found that a total of 28 genes overlapped in these two types of genes. These 28 intersection genes were subsequently ranked according to their log2FC and presented as a ranked difference plot to highlight the most significant changes in these genes across the cross-set (Fig. [53]1D). To show the expression changes of these genes more visually, we visualized these 28 intersection genes to hight differences (15 upregulated and 13 downregulated genes) via a gradient volcano plot (Fig. [54]1E). Finally, the heat map (Fig. [55]1F) compares the expression levels of these 28 intersection genes in OS tissues versus normal tissues, showing their differential levels between tumor and normal tissues (Fig. [56]1F). By performing functional analysis of 28 intersection genes, we first constructed protein-protein interaction (PPI) networks for these genes (Supplementary Fig. 1A), revealing interaction relationships between the proteins they encode. Critical hub proteins in the network include CDK1, PIK3R1 and PDGFRB, indicating that these proteins may play an important role in the development of OS. Next, we performed cellular component (CC) enrichment analysis of these genes and showed that they were mainly enrich in cellular components such as “transferase complex” and “protein kinase complex”, which implied that these genes may be involved in the formation and function of a variety of important intracellular complexes (Supplementary Fig. 1B). In addition, enrichment analysis of biological processes (BP) showed that these genes significantly involved biological processes such as “calcium homeostasis” and “ERK1 and ERK2 cascades”, suggesting that they may play a key role in regulating cell signaling and ion balance (Supplementary Fig. 1C). The enrichment results of molecular function (MF) showed that these genes mainly had the function of “protein serine/threonine/tyrosine kinase activity”, which showed their important roles in cell signal transduction and metabolism (Supplementary Fig. 1D).Further, KEGG pathway enrichment analysis revealed that these genes were significantly enriched in signaling pathways such as “EGFR tyrosine kinase inhibitor resistance” and “Rap1”, providing new insights into cancer therapy and drug resistance (Supplementary Fig. 1E). Finally, through GO classification and circular mapping maps of KEGG pathways (Supplementary Fig. 1F), we can visually see the functional annotation and pathway information of these genes and thus understand their roles in different biological processes and molecular functions more comprehensively. Fig. 1. [57]Fig. 1 [58]Open in a new tab Shikonin targeted differential gene screening for OS. A [59]GSE14359 dataset volcano plot; B [60]GSE14359 dataset difference ranking top 50 differential gene heat map; (C) Shikonin targeted OS differential gene intersection Venn plot; D 28 intersection gene difference ranking; E 28 intersection gene gradient volcano plot; F 28 intersection gene difference heat map in OS and normal tissues Construction of Shikonin targeting prognostic model in patients with osteosarcoma In this study, we first screening for prognostic differential genes in OS patients treated with Shikonin using a LASSO regression model (Supplementary Fig. 2A). The partial likelihood deviance plot showed a decreasing trend with increasing log(λ), reaching a minimum at approximately log(λ) = − 3, which suggests an optimal λ value of approximately 0.001, indicating that the genes selected at this point are most relevant for predicting the prognosis of OS patients treated with Shikonin (Supplementary Fig. 2B). As λ decreases, more genes are selected into the model, and the coefficients of some genes increase or decrease dramatically, highlighting their importance for prognostic models. Based on the four selected key genes, we constructed a risk score model for patients and divided patients into high-risk and low-risk groups according to the risk score (Supplementary Fig. 2C). Kaplan–Meier survival curves showed a significant difference in survival between the two groups, with the high-risk group having a shorter median survival time. In addition, we also plotted a heatmap to show key target genes (SCD, CCR2, NR3C2, CDK1) and their expression patterns in patients with different risk groups, revealing clear differences in gene expression levels (Supplementary Fig. 2D). To provide a quantitative tool for individualizing prognosis, we developed a nomogram prediction model that combined multiple clinical factors such as age, sex, metastatic status, and expression levels of key genes (Supplementary Fig. 2E). The total number of points can be transformed into the probability of survival at 1, 3, and 5 years. The calibration curves showed good agreement between the observed partial survival probability and the nomogram predicted survival probability, indicating that our prediction model had satisfactory predictive accuracy for survival outcomes at 1 year (Supplementary Fig. 2F), 3 years (Supplementary Fig. 2G), and 5 years (Supplementary Fig. 2H). In summary, through this study, we not only identified the key genes of Shikonin targeted therapy for OS, but also established a prognostic prediction model of Shikonin targeted therapy for OS, providing a scientific basis for clinical application. To assess the prognosis of key targets in OS, we first assessed the clinical utility of Shikonin targeting as a key target for OS (SCD, CCR2, NR3C2, CDK1) by decision curve analysis (DCA) at 1, 3 ,5 years (Supplementary Fig. 3A-C). The results showed that the net benefit using the risk score model was higher than the strategy of “all patients treated” or “none treated” over a wide range of threshold probabilities. The curves for each key target also showed significant net benefit compared to these two extreme strategies, indicating their clinical value in predicting short- and long-term survival. In addition, we also plotted receiver operating characteristic (ROC) curves for these key targets to assess their predictive performance. The results showed areas under the curve (AUC) of 0.840 for SCD, 0.852 for CCR2, 0.973 for NR3C2, and 0.977 for CDK1. These high AUC values indicate a strong discriminatory power of these targets in distinguishing different survival outcomes (Supplementary Fig. 3D). Based on selected key targets, we constructed an overall survival risk score for patients and compared overall survival between the low-risk group and the high-risk group by Kaplan-Meier survival curves. The results showed that patients in the high-risk group had significantly worse survival outcomes (HR = 2.96, 95% CI = 1.71–5.10, P < 0.001; Supplementary Fig. 3E). Further, we performed survival analysis separately for each key target. High SCD expression levels were associated with worse survival (Supplementary Fig. 3F). Similarly, high CDK1 expression levels were also associated with worse survival (Supplementary Fig. 3G). In contrast, high expression of CCR2 (Supplementary Fig. 3H) and NR3C2 (Supplementary Fig. 3I) was associated with better survival. In order to better investigate the role of targeted genes that are highly expressed and have a poor prognosis in OS, SCD and CDK1 were selected for in-depth study. Clinical characteristics of key targets of Shikonin targeted therapy for osteosarcoma To investigate the role of CDK1, a key target of Shikonin targeted therapy for OS, and SCD in OS, we first analyzed the pan-cancer differential expression of CDK1, a key target, in a variety of cancer types. The results showed that CDK1 was significantly overexpressed in tumor samples from multiple cancer types (e.g., ACC, BLCA, BRCA, etc.) compared to normal tissues. These results suggest that CDK1 may play an important role in tumorigenesis in multiple cancer types (Fig. [61]2A). Next, we investigated the relationship between the expression levels of key targets (SCD and CDK1) and sex in OS patients. The analysis found that SCD expression levels were not significantly different between male and female patients, while CDK1 expression was higher in female patients than in male patients, suggesting that sex may have some effect on CDK1 expression (Fig. [62]2B). Further, we investigated the relationship between expression levels of key targets (SCD and CDK1) and metastatic status. The results showed that the expression level of CDK1 was significantly upregulated in patients with metastasis, which suggests that it may play a role in promoting OS metastasis (Fig. [63]2C). In addition, we analyzed the expression levels of key targets (SCD and CDK1) in relation to tumor residual status and found that CDK1 expression levels were higher in patients with tumor residual (R1 and R2) than in patients with complete resection (R0). This finding suggests that these targets may be involved in tumor persistence and may serve as markers to predict residual disease after surgery (Fig. [64]2D). Therefore, we chose CDK1 for further investigation. Finally, we investigated the association between CDK1 expression and immune cell infiltration in OS. The heatmap showed that the proportion of activated NK cells, M1 macrophages, and activated dendritic cells increased, while the proportion of resting immune cells (such as B cells and resting NK cells) was relatively low in the high CDK1 expression group. This suggests that CDK1 may modulate the immune microenvironment by promoting activation of certain immune cell types, thereby potentially influencing immune responses in OS (Fig. [65]2E). In summary, this study not only reveals the clinical features of CDK1, a key target of Shikonin targeted therapy for OS, and its association with immune cells, but also provides a new perspective for understanding the role of CDK1 in tumor biology. These findings lay the foundation for further exploration of Shikonin as a novel strategy for OS treatment. Fig. 2. [66]Fig. 2 [67]Open in a new tab Clinical features and immunoassay of key targets of Shikonin targeting OS. A Pan-cancer differential expression of the key target CDK1; B Correlation of the key target with sex in OS; C Correlation of the key target with metastasis in OS; D Correlation of the key target with residual tumor in OS; E Correlation of the key target CDK1 with immune cells. *, P < 0.05; **, P < 0.01; ***, P < 0.001 Shikonin interacts with the CDK1 molecule To assess the binding ability between Shikonin and CDK1, we performed molecular dynamics simulation analysis. First, the root mean square deviation of the Shikonin-CDK1 complex suggests that Shikonin maintains a stable conformation in the CDK1 binding pocket throughout the simulation, which suggests a strong and sustained interaction between Shikonin and CDK1 (Fig. [68]3A). Next, the results of structural stability of Shikonin-CDK1 complexes over time showed that both control (CDK1 alone) and Shikonin-bound CDK1 were relatively structurally stable, but Shikonin-bound CDK1 showed lower RMSD values, indicating that ligand binding enhanced its stability (Fig. [69]3B). In addition, dynamic stability of certain atomic regions in CDK1 was shown by RMSF analysis, and it was found that the motility of some specific residues decreased when CDK1 was combined with Shikonin, suggesting that this ligand was able to stabilize these regions (Fig. [70]3C). Meanwhile, the tightness of the overall structure of the Shikonin-CDK1 complex was assessed by radius of Gyrate analysis, which showed that Gyrate values remained relatively constant throughout the simulation, indicating that the complex maintained a stable and compact structure (Fig. [71]3D). Further, the XVG-box comparison provides a thermodynamic comparison of the Shikonin-CDK1 complex under different conditions, and the boxplot shows that the presence of Shikonin makes the complex more stable, with fewer thermal fluctuations and better thermodynamic properties than the unbound state (Fig. [72]3E). Meanwhile SASA curves showed that binding of Shikonin reduced the exposed surface area of CDK1 and contributed to stabilization of the complex (Fig. [73]3F). The number of hydrogen bonds between Shikonin and CDK1 varied over time, and hydrogen bond analysis showed multiple stable hydrogen bonds formed throughout the simulation, supporting strong interactions between ligand and protein (Fig. [74]3G). In addition, the evolution of the secondary structure of the Shikonin-CDK1 complex over time suggests that binding of Shikonin does not significantly alter the secondary structure of CDK1, indicating that the ligand achieves binding without causing major conformational changes (Fig. [75]3H). The contribution of each residue to the total energy identifies that some key residues play an important role in stabilizing the complex, highlighting the importance of specific interactions for overall binding affinity (Fig. [76]3I). The energy landscape of the Gibbs free energy of the Shikonin-CDK1 complex as a function of the two principal components shows that the complex is in a low-energy state, indicating that it has a stable and favorable binding mode (Fig. [77]3J). Finally, 3D free energy topography provides a visual representation of the most stable conformation of the complex, further confirming a strong and stable interaction between Shikonin and CDK1 (Fig. [78]3K). Taken together, the results of molecular dynamics simulations show that Shikonin exhibits strong binding ability to CDK1, as evidenced by stable conformations, enhanced structural stability, reduced atomic fluctuations, compact structures, reduced surface exposure, sustained hydrogen bonding, unchanged secondary structures, significant residue energy contributions, low-energy states, and overall structural stability. These findings provide an important basis for an in-depth understanding of the molecular interaction mechanism between Shikonin and CDK1 and also support its potential as a therapeutic agent targeting CDK1. Fig. 3. [79]Fig. 3 [80]Open in a new tab Molecular dynamics assessment of Shikonin binding ability to CDK1. A Frequency profile of Shikonin-CDK1 complexes; B Structural stability of Shikonin-CDK1 complexes over time; C Dynamic stability of atoms in some regions of CDK1; D Closeness of the overall structure of Shikonin-CDK1 complexes; E Comparison of XVG-box thermodynamics of Shikonin-CDK1 complexes under different conditions; F Degree of molecular surface exposure of Shikonin-CDK1 complexes; G Number of hydrogen bonds in Shikonin-CDK1 complexes over time; H Secondary structure of Shikonin-CDK1 complexes over time; I Contribution of each residue to total energy in Shikonin-CDK1 complexes; J Gibbs energy of Shikonin-CDK1 complexes changes with the two principal components; K 3D structure of Shikonin-CDK1 complexes Inhibitory effect of Shikonin on proliferation and toxicity of osteosarcoma cells To investigate the effect of Shikonin in OS cells in vitro, we first determined the half maximal inhibitory concentration (IC50) of Shikonin in MG63 cells and showed that its IC50 value was 1.73 µM. Cell viability decreased significantly with increasing concentration of Shikonin, indicating its strong inhibitory effect on MG63 cells (Fig. [81]4A). Similarly, Shikonin showed significant cell growth inhibition in Saos-2 cells with IC50 value of 1.38 µM (Fig. [82]4B). CCK-8 assay was used to assess the time-dependent effect of Shikonin on MG63 and Saos-2 cell proliferation. For MG63 cells, cell viability in the Shikonin treated group was significantly reduced compared to the DMSO control group at 48 and 72 h, indicating that it effectively inhibited cell proliferation (Fig. [83]4C).Similar results were observed in Saos-2 cells, showing that Shikonin significantly inhibited proliferation of these cells in a time-dependent manner (Fig. [84]4D).To assess the potential toxicity of Shikonin on normal cells, we examined its effects on L0-2 liver epithelial cells, HK-2 tubular epithelial cells, as well as hFOB1.19 osteoblasts. The results showed that although Shikonin showed some cytotoxicity to these normal cells, its IC50 values were 5.69 µM (L0-2 cells, Fig. [85]4E), 10.28 µM (HK-2 cells, Fig. [86]4F) and 6.16 µM (hFOB1.19 cells, Fig. [87]4G), which were much higher than their IC50 values for OS cells, suggesting that Shikonin has some selective toxicity to cancer cells. Finally, the inhibitory effect of Shikonin on Saos-2 cell proliferation was visually observed and quantified by EdU assay. After 24 h of Shikonin treatment, the number of EdU positive cells was significantly reduced, confirming its ability to inhibit DNA synthesis and cell proliferation. Quantitative analysis showed that the proportion of EdU positive cells in the Shikonin-treated group was significantly lower than that in the DMSO control group (Fig. [88]4H). In summary, this study not only revealed that Shikonin had a significant antiproliferative effect on OS cells (MG63 and Saos-2), but also assessed its toxicity to normal liver, kidney and osteoblasts cells, finding that it exhibited some toxicity to these normal cells. Fig. 4. [89]Fig. 4 [90]Open in a new tab Effect and toxicity assessment of Shikonin in OS cells. A IC50 of Shikonin in MG63 cells, n = 5; B IC50 of Shikonin in Saos-2 cells, n = 5; C CCK-8 to assess the inhibition of proliferation of MG63 cells by Shikonin, n = 5; D CCK-8 to assess the inhibition of proliferation of Saos-2 cells by Shikonin, n = 5; E assessment of the toxic effect of Shikonin on normal epithelial cells of the liver (L0-2), n = 5; F assessment of the toxic effect of Shikonin on normal epithelial cells of the renal tubules (HK-2), n = 5; G assessment of the toxic effect of Shikonin on osteoblasts (hFOB1.19), n = 5; and H EdU to assess the inhibitory effect of Shikonin on the proliferation of OS cell lines (Saos-2). ***, P < 0.001 Synthesis of Shikonin complex containing pH-response intelligent hydrogel nanomaterial In order to solve the problem of poor water solubility and normal cytotoxicity of Shikonin, we synthesized a complex loaded with Shikonin in pH-responsive intelligent hydrogel nanomaterial (Gel@PLGA@FA). To verify the successful synthesis, we first analyzed the particle size distribution of PLGA nanoparticles by DLS, which showed that their average diameter was about 72 nm, and they had uniform particle size distribution and good synthesis efficiency (Fig. [91]5A). Further, DLS analysis of PLGA@Shikonin nanoparticles revealed a slight increase in particle size to about 91.6 nm, indicating that Shikonin was successfully encapsulated in PLGA matrix without significantly altering the morphology of the nanoparticles (Fig. [92]5B). For PLGA@Shikonin@FA nanoparticles, DLS data showed a further increase in particle size to approximately 132 nm, which suggests that FA was successfully attached to the nanoparticle surface (Fig. [93]5C). To assess the stability of these nanoparticles, we measured their zeta potential. PLGA nanoparticles showed good colloidal stability at a zeta potential of approximately 2.15 mV (Fig. [94]5D). However, the zeta potential of PLGA@Shikonin nanoparticles was slightly reduced to about 7.95 mV, indicating that the encapsulation of Shikonin did not significantly affect the overall charge of nanoparticles and still maintained good colloidal stability (Fig. [95]5E). The zeta potential of PLGA@Shikonin@FA nanoparticles was about − 44.35 mV, indicating that the attachment of folic acid enhances the negative electricity of the nanoparticle surface and may help to improve its stability and targeting efficiency (Fig. [96]5F). SEM images showed that the Gel@PLGA@Shikonin@FA composite had a porous internal structure, in which the nanoparticles were evenly distributed and showed a smooth spherical morphology, indicating that the nanoparticles were successfully embedded in the hydrogel network (Fig. [97]5G). Higher magnification TEM images showed the morphology of individual nanoparticles more clearly, showed consistent sizes and shapes, and confirmed the uniform distribution of nanoparticles in the hydrogel (Fig. [98]5H). Subsequently, we characterized PLGA using a fourier transform infrared (FTIR) spectrometer. The results revealed a distinct absorption peak in the range of 1701 cm^− 1, which corresponds to the infrared absorption of the carbonyl (C = O) bond within the ester groups (-COO-) in the PLGA molecule. This observation confirms the successful synthesis of PLGA. Then, the drug loading capacity and encapsulation efficiency of the hydrogel nanocomposite were determined using UV-vis spectrophotometry, yielding values of 5.6% and 85.8%, respectively. Finally, the pH - responsive drug release profiles revealed the drug release behavior of the Gel@PLGA@Shikonin@FA composite under different pH conditions. The results showed that Shikonin exhibited a continuous release trend within a certain period of time. Specifically, the release was faster at pH 5 compared to other pH values (6.0, 6.5, 7.0, and 7.4). This pH - responsive characteristic indicated that the composite could be used for local tumor drug delivery and achieve controlled drug release in the tumor microenvironment (low pH) (Fig. [99]5J). In summary, this study not only showed that PLGA-based nanocomposites successfully loaded Shikonin and achieved folic acid functionalization, but also verified that they had ideal physicochemical properties, including appropriate particle size, charge, morphology and pH-responsive drug release behavior. These findings support the potential application value of this nanocomposite as a targeted drug delivery system in cancer therapy. Fig. 5. [100]Fig. 5 [101]Open in a new tab Hydrogel nanomaterial complex synthesis characterization. A DLS plot of PLGA, 72 ± 2.31, n = 3; B DLS plot of PLGA@Shikonin, 91.6 ± 2.87, n = 3; C DLS plot of PLGA@ Shikonin@FA, 132 ± 4.54, n = 3; D Zeta plot of PLGA, 2.15 ± 0.25, n = 3; E Zeta plot of PLGA@Shikonin, 7.95 ± 0.32, n = 3; F Zeta plot of PLGA@Shikonin@FA, -44.35 ± 0.87, n = 3; G SEM plot of Gel@PLGA@Shikonin@FA; H TEM plot of Gel@PLGA@Shikonin@FA; I Infrared spectroscopy chart of PLGA; J PH drug release curve of Gel@PLGA@Shikonin@FA Targeted uptake capacity and cytotoxicity assessment of pH- responsive hydrogel nanomaterial complexes We further assessed the targeted uptake capacity and cytotoxicity of pH-responsive hydrogel nanomaterial complexes. First, we observed the uptake of Gel@PLGA@DIO in Saos-2 cells using confocal microscopy. The results showed that at 0 h, very little green fluorescence was observed, indicating low initial uptake (Fig. [102]6A). After 24 h, the green fluorescence of Gel@PLGA@DIO was slightly enhanced, indicating that only a small amount of nanoparticles were taken up. When Saos-2 cells were further treated with FA-functionalized nanoparticles (Gel@PLGA@DIO@FA), their uptake efficiency was significantly enhanced at 24 h (Fig. [103]6B). Saos-2 cells treated with Gel@PLGA@DIO@FA showed a significant increase in green fluorescence intensity at 24 h compared to unmodified Gel@PLGA@DIO. This suggests that FA modification improves the targeting ability of nanoparticles to cancer cells, due to folate receptors highly expressed on the surface of Saos-2 cells. Next, we evaluated the toxicity of this pH-responsive hydrogel nanomaterial complex (Gel@PLGA@Shikonin@FA) on normal cell lines. In L-02 liver epithelial cells, cell survival remained above 90% even at the highest concentration tested (200 µM/mL), indicating that this material was minimally toxic to normal hepatocytes (Fig. [104]6C). Similarly, no significant decrease in cell viability was observed in HK-2 tubular epithelial cells exposed to Gel@PLGA@Shikonin@FA for up to 48 h at any concentration tested (Fig. [105]6D), suggesting that the composite did not produce significant toxicity to tubular epithelial cells. Finally, we investigated its effect on hFOB1.19 osteoblasts. The results showed that cell viability remained stable and approached 100% at all time points and concentrations, indicating that the pH-responsive hydrogel nanomaterial complex was non-toxic to osteoblasts (Fig. [106]6E). In summary, the results of targeted uptake studies and toxicity assessment showed that the pH-responsive hydrogel nanomaterial complex (Gel@PLGA@Shikonin@FA) showed excellent targeting ability for Saos-2 cells while maintaining low toxicity in normal hepatocytes, renal cells, and osteocytes. These findings highlight their potential as efficient and safe delivery systems for targeted cancer therapeutics. Fig. 6. [107]Fig. 6 [108]Open in a new tab Targeted uptake capacity and toxicity assessment of pH responsive hydrogel nanomaterial complexes. A Targeted uptake capacity of Gel@PLGA@DIO in OS cell line (Saos-2); B Targeted uptake capacity of Gel@PLGA@DIO@FA in OS cell line (Saos-2); C Toxicity assessment of pH responsive hydrogel nanomaterial complexes to normal liver epithelial cells; D Toxicity assessment of pH responsive hydrogel nanomaterial complexes to tubular epithelial cells; E Toxicity assessment of pH responsive hydrogel nanomaterial complexes to osteoblasts. ns, P > 0.05 pH-responsive hydrogel nanomaterial complex equipped with Shikonin enhances anti-PD-L1 therapeutic effect in osteosarcoma We further evaluated the enhancing effect of Shikonin-loaded (Gel@PLGA@Shikonin@FA) pH-responsive hydrogel nanomaterial complexes on the anti-PD-L1 effect of OS cells. The results showed that in Saos-2 and MG63 cells, CDK1 mRNA expression levels were significantly decreased in the Gel@PLGA@Shikonin@FA treatment group compared with Gel@PLGA alone (Fig. [109]7A). This suggests that Shikonin loaded in folate-functionalized nanoparticles is able to effectively downregulate CDK1 expression. Similarly, Gel@PLGA@Shikonin@FA significantly reduced the mRNA expression level of CD279 in these two cell lines (Fig. [110]7B), suggesting that this nanocomplex has the ability to regulate immune checkpoint molecules and may enhance the effect of immunotherapy. The inhibitory effect on CDK1 expression was further enhanced when Gel@PLGA@Shikonin@FA was combined with the anti-PD-L1 inhibitor atezolizumab (Fig. [111]7C). In Saos-2 and MG63 cells, CDK1 mRNA expression was significantly reduced in the Gel@PLGA@Shikonin@FA plus atezolizumab group compared with the Gel@PLGA plus atezolizumab group alone, showing a synergistic effect and highlighting the potential of targeted drug delivery systems in combination with immunotherapeutic agents. In addition, this combined treatment strategy also showed a significant inhibitory effect on the proliferation of OS cells. Both MG63 (Fig. [112]7D) and Saos-2 (Fig. [113]7E) cell lines showed a significant decrease in cell viability after treatment with Gel@PLGA@Shikonin@FA combined with atezolizumab, with OD450 values significantly lower than other treatment groups at all time points, indicating that this combination therapy potently inhibited the proliferation of tumor cells. Finally, flow cytometry analysis showed that Gel@PLGA@Shikonin@FA combined with atezolizumab promoted apoptosis of Saos-2 cells (Fig. [114]7F). Compared with the DMSO control group (apoptosis rate of 2.17%), the apoptosis rate of the combined treatment group increased to 33.45% (which was also significantly higher than that of the single group), indicating that the combined treatment had a significant effect in inducing tumor cell apoptosis. In summary, the pH-responsive hydrogel nanomaterial complex (Gel@PLGA@Shikonin@FA), especially in combination with atezolizumab, effectively enhances the anti-PD-L1 effect on OS cells by mechanisms such as down-regulation of CDK1 and PD-1 expression, inhibition of cell proliferation, and promotion of apoptosis. These findings provide a strong experimental basis and offer new strategies for improving OS treatment by targeted drug delivery systems combined with immunotherapy. Fig. 7. [115]Fig. 7 [116]Open in a new tab pH-responsive hydrogel nanomaterial complexes augment anti-PD-L1 effects in OS cells. A Gel@PLGA@Shikonin@FA decreased the expression level of CDK1, n = 3; B Gel@PLGA@Shikonin@FA decreased the expression level of CD279, n = 3; C Gel@PLGA@Shikonin@FA combined with atezolizumab decreased the expression level of CDK1, n = 3; D Gel@PLGA@Shikonin@FA combined with atezolizumab inhibited the proliferation of MG63 cells, n = 5; E Gel@PLGA@Shikonin@FA combined with atezolizumab inhibited the proliferation of Saos-2 cells, n = 5; F Gel@PLGA@Shikonin@FA combined with atezolizumab promoted the apoptosis of Saos-2, n = 3. **, P < 0.01; ***, P < 0.001; #, P < 0.0001 To further investigate the mechanism by which Gel@PLGA@Shikonin@FA combined with PD-L1 inhibitor atezolizumab promoted apoptosis in OS cells, we assessed changes in mitochondrial membrane potential using JC-1 staining. In DMSO-treated controls, most cells showed high mitochondrial membrane potential as indicated by red fluorescence emitted by JC-1 polymers. This suggests that these cells are in a healthy state and have intact mitochondria. However, a significant increase in green fluorescence from JC-1 monomers was observed when treated with Gel@PLGA@Shikonin@FA alone, indicating a decrease in mitochondrial membrane potential and signs of early apoptosis. Similarly, treatment with atezolizumab alone also resulted in an increase in green fluorescence, suggesting that PD-L1 inhibitors can induce some degree of mitochondrial dysfunction and apoptosis. However, the effect was more pronounced when Gel@PLGA@Shikonin@FA was combined with atezolizumab. In this combined treatment group, green fluorescence intensity was significantly higher than in the other groups, while red fluorescence was significantly reduced, suggesting substantial loss of mitochondrial membrane potential and strong apoptosis induction (Fig. [117]8). In summary, the results of JC-1 staining experiments showed that the combination of Gel@PLGA@Shikonin@FA and atezolizumab was able to effectively perturb mitochondrial membrane potential in OS cells, which resulted in more intense apoptosis. This synergistic effect highlights the potential of combining targeted drug delivery systems with immunotherapeutic agents for improving cancer treatment outcomes. Fig. 8. [118]Fig. 8 [119]Open in a new tab JC-1 detection of pH response to hydrogel nanomaterial complex (Gel@PLGA@Shikonin@FA) combined with PD-L1 inhibitor (Atezolizumab) promotes apoptosis of OS cells Discussion This study represents the first attempt to encapsulate Shikonin into a tumor-targeted, pH-responsive nanocarrier, specifically designed to enhance the efficacy of PD-L1 inhibitors in osteosarcoma. Previous research on Shikonin combined with immunotherapy has mostly relied on free drug administration, which is plagued by issues such as poor solubility, rapid clearance, and off-target toxicity [[120]9]. In contrast, our nanoplatform significantly improves the bioavailability and tumor targeting of shikonin. Studies by Wang [[121]15] and Yu et al. [[122]16] further supported that the combination of Shikonin with PD-L1 inhibitors significantly improved antitumor efficacy and prolonged the survival of tumor-bearing mice. In contrast, studies have shown limited efficacy of atezolizumab alone in the treatment of liver cancer [[123]17], particularly in “cold tumors” such as OS, which highlight the importance of combined treatment strategies. Through analysis of the [124]GSE14359 dataset, we identified a series of DEGs and found that 28 of them overlapped among Shikonin targeted genes. Functional analysis of these genes has revealed their critical role in regulating cell signaling, ion balance, and so forth, and is potentially important in multiple cancer types. Based on this, we constructed a patient’s risk score model combining expression levels of key genes as well as multiple clinical factors to provide a scientific basis for individualized prognosis. However, the bibliometric analysis of the role of autophagy in the diagnosis and treatment of OS by Zhu et al. [[125]1] emphasizes the necessity of exploring OS treatment strategies from multiple perspectives. In addition, we investigated the molecular interaction mechanism between Shikonin and CDK1. Molecular dynamics simulation results showed that Shikonin showed strong binding ability to CDK1, with stable conformation, enhanced structural stability, and decreased atomic fluctuations. This lays the foundation for further exploration of Shikonin as a novel strategy for OS treatment. In contrast, recent advances on Shikonin in the treatment of immune-related diseases by Guo [[126]5] and others point out that the mechanism of action of Shikonin is not limited to direct anticancer effects, but also includes a wide range of anti-inflammatory and immunomodulatory functions. In assessing the effect of Shikonin in OS in vitro, we found that it had a significant antiproliferative effect on MG63 and Saos-2 cell lines. However, Shikonin also showed some toxicity to normal liver, kidney, and osteoblasts, but its IC50 value was much higher than that for OS cells, indicating that Shikonin has some selective toxicity to cancer cells. This phenomenon is similar to that observed by Wang et al. [[127]15] in studies investigating Shikonin promoting apoptosis of HCC cells to enhance anti-PD-L1 efficacy, although they focused on different biomarkers and mechanisms of action. In order to overcome the disadvantages of poor water solubility and normal cytotoxicity of Shikonin, we successfully synthesized a pH-responsive intelligent hydrogel nanomaterial (Gel@PLGA@FA) equipped with Shikonin complex. By enabling the local release of Shikonin, our nanocomposite enhances anti-tumor immune responses and exerts a synergistic effect with atezolizumab, achieving superior tumor suppression efficacy and prolonged survival in osteosarcoma models. Importantly, compared with free Shikonin, this system significantly reduces toxicity. Similarly, Zhang et al. [[128]18] used pH-sensitive polymer micelles to load chemotherapeutic drugs to achieve effective accumulation and controlled release at the tumor site of pancreatic cancer, significantly improving the therapeutic effect and reducing side effects. Finally, we evaluated the anti-PD-L1 therapeutic efficacy of this pH-responsive hydrogel nanomaterial complex on OS cells. The results showed that Gel@PLGA@Shikonin@FA combined with atezolizumab could effectively downregulate CDK1 and PD-1 expression, inhibit cell proliferation, and promote apoptosis, thereby significantly enhancing the anti-PD-L1 effect on OS cells. JC-1 staining experiments further confirmed that this combination therapy was able to effectively perturb mitochondrial membrane potential in OS cells, leading to more intense apoptosis. This conclusion echoes the findings of Li et al. [[129]19] regarding the simultaneous regulation of ROS generation and copper metabolism by nanomedicine for sonodynamic amplification tumor therapy, demonstrating the great potential of combined therapeutic strategies. In summary, this study not only reveals the unique mechanism of action of Shikonin as a potential anticancer drug, but also demonstrates the potential of pH-responsive hydrogel nanomaterial complexes as efficient and safe delivery systems for targeted cancer therapeutics. These findings provide a strong experimental rationale and novel strategies for improving OS treatment by targeted drug delivery systems combined with immunotherapy. Conclusion Shikonin can significantly down-regulate the expression of CDK1 and PD-L1, inhibit OS cell proliferation and promote apoptosis through strong and stable binding to the key target, CDK1. In addition, we successfully constructed a pH-responsive hydrogel nanocomplex (Gel@PLGA@FA) equipped with Shikonin, which showed good drug release characteristics at different pH conditions, especially in the tumor microenvironment to achieve controllable drug release. The combined use of Gel@PLGA@Shikonin@FA and PD-L1 inhibitor (atezolizumab) not only effectively enhanced the anti-PD-L1 therapeutic effect on OS cells, but also led to stronger apoptosis by disrupting the mitochondrial membrane potential. The combination of Shikonin and atezolizumab provides an innovative and efficient treatment approach for OS patients. Future studies will further optimize the design of this complex and bring new hope for OS treatment. Supplementary Information [130]Supplemenatary Material 1.^ (1.2MB, docx) Acknowledgments