Abstract Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality, with high postoperative recurrence rates due to occult micrometastases or minimal residual disease, markedly worsening the prognosis for HCC patients. Current therapies lack effective strategies to prevent recurrence, while traditional Chinese medicine (TCM) shows potential in delaying HCC progression. Combining a hemostatic hydrogel with nanoparticle-based delivery of active TCM components provides a strategy to enhance tumor recurrence prevention. Herein, we develop a nanocomposite hydrogel (HPS@ZCJ) by encapsulating Jianpi-Huayu essential oils (JEO) into zein-based nanoparticles (zein@chondroitin sulfate@JEO, ZCJ) and embedding them in a hydroxymethyl cellulose/Pluronic F-127/sodium alginate (HPS) hydrogel matrix. HPS@ZCJ hydrogel enhances cytotoxic T-lymphocyte infiltration, inhibits the polarization of tumor-associated macrophages to M2 phenotype, induces tumor cell death, reverses immunosuppression, and inhibits angiogenesis within the tumor. The antitumor mechanism involves dual downregulation of GPNMB and DHCR7, key genes in HCC progression and immune evasion. In vitro and in vivo experiments demonstrate that HPS@ZCJ hydrogel-mediated targeted comprehensive therapy simultaneously achieves intraoperative hemostasis, impedes primary tumor growth and prevents HCC postoperative recurrence. This study provides a promising postoperative HCC treatment strategy, leveraging TCM's therapeutic potential with significant clinical translation prospects. Keywords: Nanoparticle drug delivery, Jianpi-Huayu decoction, Essential oils, Postoperative treatment, Hepatocellular carcinoma, Hydrogel Graphical abstract Image 1 [41]Open in a new tab 1. Introduction Hepatocellular carcinoma (HCC) ranks as the third-leading cause of cancer mortality worldwide, with a dismal 5-year survival rate below 18 % [[42]1]. While surgical resection remains the gold-standard treatment for early-stage HCC [[43]2], up to 70 % of patients, including those with small, solitary tumor nodules (≤2 cm), experience recurrence within 5 years [[44]3]. This high recurrence rate stems primarily from occult micrometastases and minimal residual disease that enter circulation during intraoperative bleeding [[45]4]. Notably, current clinical guidelines lack effective pharmacological interventions for postoperative recurrence prevention [[46]5], highlighting an urgent unmet need in HCC management. Traditional Chinese medicine (TCM) has shown promise in managing HCC, particularly in delaying progression and reducing recurrence, either alone or in conjunction with other conventional therapies. Among TCM formulas, Jianpi-Huayu decoction (JHD)-composed of Baizhu (Rhizoma Atractylodis Macrocephalae), Ezhu (Curcuma zedoaria Roscoe), Fuling (Poria cocos), Foshou (fingered citron), Kushen (Radix Sophorae Flavescentis), and Baihuasheshecao (Hedyotis diffusa Willd)-exerts direct cytotoxicity against HCC cells, inhibits angiogenesis, and modulates immunity [[47][6], [48][7], [49][8]]. Notably, the essential oils derived from JHD (JEO)-rich in monoterpenes and sesquiterpenes-are key bioactive components with demonstrated anticancer and immunoregulatory properties [[50][9], [51][10], [52][11], [53][12], [54][13], [55][14]]. However, the clinical translation of JEO is hindered by poor solubility, which limits its bioavailability and administration routes. Thus, developing advanced delivery systems for JEO could improve its bioavailability and enhance HCC therapy. Nanoparticle (NP) delivery systems have shown promise for overcoming the poor solubility of JEO [[56]15,[57]16]. Among them, zein NPs exhibit distinct advantages: (1) high loading capacity for lipophilic drugs, compatible with JEO's essential oils [[58]17]; (2) γ-zein's N-terminal proline-rich domain promotes cell membrane interaction, enhancing cellular uptake [[59]18]. However, conventional intravenous administration of NPs risks redistribution and side-effects, while locally injected particles can migrate or prematurely release therapeutic agents [[60]19,[61]20]. Therefore, engineering zein NPs for localized JEO delivery with prolonged tumor retention is essential to maximize therapeutic efficacy while minimizing off-target effects. Hydrogels have emerged as attractive platforms for local drug delivery due to their biocompatibility, biodegradability, and ability to adhere to target sites, yet loading hydrophobic agents remains problematic [[62]21,[63]22]. This challenge can be addressed through nanocomposite hydrogels that combine the advantages of drug-loaded NPs with hydrogel matrices [[64]19]. In the context of HCC treatment, this approach becomes particularly relevant given the clinical challenges associated with tumor resection, including intraoperative bleeding and high postoperative recurrence rates due to potential micrometastasis [[65]23]. In this study, we introduce an innovative solution through the development of a thermosensitive composite hydrogel system composed of hydroxypropyl methylcellulose (HMC), Pluronic F-127 (PF127), and sodium alginate (SA), which was termed HPS (HMC-PF127-SA). This formulation was specifically designed to address multiple clinical needs simultaneously. HMC contributes crucial hemostatic properties through its branched fiber network and ability to promote thrombus formation at vascular injury sites [[66][24], [67][25], [68][26]]. PF127 provides temperature-responsive gelation behavior, enabling convenient in situ application during surgical procedures [[69]27]. SA complements HMC's bioadhesive function while strengthening the hydrogel's structural integrity [[70]28]. In addition, the system was further optimized by incorporating JEO-loaded zein nanoparticles stabilized with chondroitin sulfate (CS), designated as Zein-CS@JEO (ZCJ). The CS modification maintains the NPs'cell-interaction capabilities while improving their stability within the hydrogel matrix [[71]29]. The complete hydrogel system, incorporating the ZCJ NPs, was termed HPS@ZCJ ([72]Fig. 1). The in-situ-formed HPS@ZCJ hydrogel enables intraoperative injection and provides three key clinical benefits for postoperative HCC management: (1) immediate hemostatic action to reduce the risk of tumor cell dissemination; (2) precise local delivery of therapeutic JEO components, minimizing loss and off-target toxicity; (3) controlled release kinetics through the combined nanoparticle-hydrogel architecture. This therapeutic strategy specifically targets early-stage HCC (BCLC stages 0 and A), where surgical resection remains the primary treatment option but faces significant challenges from postoperative recurrence [[73]30]. The high recurrence rates in these stages, often due to occult micrometastases or minimal residual disease, highlight the need for effective postoperative therapies to prevent recurrence [[74]31]. In summary, the HPS@ZCJ hydrogel presents a treatment strategy for HCC based on TCM-inspired therapy, synergistically combined with physical barriers against tumor cell spread to effectively prevent post-resection HCC recurrence. Fig. 1. [75]Fig. 1 [76]Open in a new tab Schematic of preparing HPS@ZCJ hydrogel and combination therapy for postoperative recurrence of HCC. Created with [77]BioRender.com. 2. Materials and methods 2.1. Preparation and component analysis of JEO The JHD is formulated from six traditional Chinese medicinal herbs: Ezhu (Curcuma Zedoaria Roscoe), Baizhu (Rhizoma Atractylodis Macrocephalae), Foshou (Fingered Citron), Fuling (Poria Cocos), Kushen (Radix Sophorae Flavescentis), Baihuasheshecao (Hedyotis Diffusa Willd). These herbs are combined in a proportion of 3:3:3:5:5:5. The herbs are then cut or ground into small particles using scissors or herbal grinder. Next, the herbal mixture is placed into a round-bottom flask and soaked in ultrapure water at a volume ratio of 1:8 (herbs: water) for 6 h. JEO is subsequently extracted via steam distillation. The extracted JEO is then sealed with a paraffin film and stored at 4 °C in a light-protected refrigerator for future use. The chemical constituents of JEO were analyzed using high-resolution liquid chromatography-mass spectrometry (HRLC-MS) and gas chromatography-mass spectrometry (GC-MS). Specifically, 100 μL of the sample was combined with 500 μL of methanol, mixed thoroughly, and centrifuged at 13,000 rpm for 10 min. The resulting supernatant was subsequently used for detection. The liquid chromatography system utilized was an UltiMate 3000 UHPLC. The chromatographic column was a Thermo Hypersil gold column (1.9 μm, 2.1 mm × 100 mm). The mobile phase was composed of 0.1 % formic acid in acetonitrile (B) and 0.1 % formic acid in water (A). The flow rate for chromatographic analysis was maintained at 0.3 mL/min. The gradient elution program was as follows: starting at 10 % B, the proportion of B was increased to 100 % over 10 min; 100 % B was held for 7 min, then returned to the initial condition within 0.1 min, followed by column equilibration for 2.9 min. The total runtime was 20 min. The mass spectrometry instrument used was a Q-Exactive (Thermo Fisher Scientific, CA, USA) equipped with the HESI source. The ion source temperature was set at 310 °C, the capillary temperature at 320 °C, the sheath gas flow rate at 30 units, and the auxiliary gas flow rate at 10 units. The spray voltage was 3 kV in positive ion mode and 2.8 kV in negative ion mode. Data-dependent acquisition (DDA) was employed with a loop count of 10. The HCD energy was configured with step-wise normalized collision energy values of 10, 28, and 35 eV. The first-order mass spectrometry scan range was 100–1500 m/z, with a resolution of 70,000, an AGC target of 3E6, and an injection time of 200 ms. For the second-order mass spectrometry, the resolution was 17,500, the AGC target was 1E5, and the injection time was 50 ms. The gas chromatograph employed was an Agilent 7890 B-5977, fitted with an HP-5MS column (30 m × 0.25 mm × 0.25 μm). Helium (He) was used as the carrier gas in constant flow mode at a rate of 1 mL/min. The column temperature was programmed as follows: the initial temperature was set to 60 °C, then ramped up to 100 °C at a rate of 20 °C/min; after holding at 100 °C for 1 min, the temperature was further increased to 300 °C at a rate of 12 °C/min and maintained for 1 min. The total runtime was 20.667 min. The injection port temperature was maintained at 280 °C. The injection volume was 1 μL, with a split ratio of 5:1. The mass spectrometer utilized an electron impact ionization source with an electron energy of 70 eV, an ion source temperature of 220 °C, and a transfer line temperature of 280 °C. The solvent delay was set to 2.5 min, and the mass spectrometer operated in full scan mode with a mass range of 10–650 amu. The HRLC-MS data were analyzed for potential chemical constituents using the Compound Discoverer software (V 3.2, Thermo Fisher Scientific, CA, USA) through an automated database search. The GC-MS data were processed using the Mass Hunter software (VB.07.00) to identify compounds via integration. The search parameters were set as follows: signal-to-noise ratio (SNR) at 2; sharpness threshold at 25 %; absolute height at 500 counts; and relative height at 0.1 %. The NIST 11 database was employed for automatic compound identification, with a score threshold of 65, absolute height set at 100 counts, and relative height at 0.5 %. To ensure batch-to-batch reproducibility, the JEO was extracted by the same steam-distillation protocol and analyzed with identical HRLC-MS and GC-MS conditions for every preparation. All approach aligns with the Chinese Pharmacopoeia guidelines for essential-oil standardization and support reproducible biological outcomes. 2.2. Preparation of ZCJ NPs ZCJ NPs were prepared at various zein-to-CS weight ratios using a solvent displacement method ([78]Table S1). Zein (10 mg, Gibco) and JEO (1 mg) were dissolved in a co-solvent mixture of ethanol and double-deionized water (DDW) (3:1 v/v, 2 mL). CS (0, 2, 5, or 10 mg, Sigma) was dissolved in DDW (2 mL) and added dropwise to the zein solution under probe sonication at 20 % amplitude for 20 s. The resulting NP suspensions were dialyzed against DDW using a dialysis bag with a molecular weight cut-off (MWCO) of 6–8 kDa (Seguin, USA) for 12 h to remove residual ethanol and unloaded JEO. The dialyzed products were then filtered through a syringe filter with a pore size of 0.45 μm (Goettingen, Germany) to remove any residual particulate matter or large aggregates. The final NP compositions are detailed in [79]Table S1. Blank NPs were prepared using the same method but without adding JEO. 2.3. Characterization of the ZCJ NPs The surface morphology of the ZCJ was investigated using Scanning electron microscopy (SEM, FEI, USA). The core–shell structure was visualized via Transmission Electron Microscope (TEM, Talos, USA). The mean particle diameter, PDI, and ζ potential of the nanospheres were assessed by dynamic light scattering at 25 °C using a Zetasizer Nano ZS90 (Malvern Instruments, UK). Each sample was analyzed at least three times, and the results are expressed as the mean ± standard deviation. Additionally, the chemical structures of the samples were documented using a Fourier Transform Infrared Spectroscopy (FTIR) spectrometer (Bruker, Horiba, Germany). 2.4. Preparation of HPS@ZCJ hydrogel To prepare a 20 % PF127 solution, 10 g of PF127 (Sigma, USA) was dissolved in 50 mL of sterile deionized water and stirred at 4 °C until completely dissolved. Subsequently, 10 mL of this solution was mixed with 400 mg of SA (96 kDa, with an M/G ratio of 1.2 as determined by FTIR analysis, Sigma), 400 mg of HMC (Macklin, China), and 200 mg of ZCJ. The mixture was stirred at 300 rpm for 1 h to form the HPS@ZCJ hydrogel. For the preparation of the blank hydrogel (HPS), the same procedure was followed, but ZCJ was omitted. In the study, the hydrogel was administered using a 1 mL syringe equipped with a 21G needle. The choice of a 21G needle is based on clinical practice, where 10 mL syringes with 21G needles are commonly used in HCC surgeries. This ensures compatibility with existing surgical procedures and equipment. 2.5. Characterization of the HPS@ZCJ hydrogel 2.5.1. Microstructure of HPS@ZCJ hydrogel The samples were freeze-dried with a lyophilizer after preparation. Following freeze-drying, liquid nitrogen was employed to fracture the samples, revealing a clean cross-section and the internal structure. Subsequently, the samples were affixed to a stage using conductive adhesive tape and coated with gold via a sputter coater at a current of 15 mA for 60 s. Ultimately, the internal structure of the samples was scrutinized with SEM (FEI, USA). 2.5.2. Rheological test The rheological characteristics of the samples were assessed utilizing a rheometer (HAAKE MARS III, Thermo Fisher Scientific, America). Hydrogel samples were situated between parallel plates (diameter 60 mm, depth 0.3 mm), and temperature and frequency sweeps were executed on the samples. The measurement parameters, including amplitude sweep, were defined according to the linear viscoelastic region of the storage modulus (G′) and loss modulus (G″). The temperature sweep spanned from 4 to 43 °C at a heating rate of 0.05 °C/s. The frequency sweep ranged from 0 to 100 rad/s. During viscosity assessments, a shear rate ranging from 0.01 to 100 s^−1 was applied to evaluate the shear-thinning characteristics of the gels. 2.5.3. FTIR analysis The samples underwent freeze-drying via a lyophilizer. Following this, the samples were pulverized into a fine powder using a mortar and pestle, after which the powder was gathered. The infrared spectra of the samples were captured using the FTIR spectrometer (Bruker Vertex 70v, Bruker, Germany) across the range of 4000–400 cm^−1, at a resolution of 4 cm^−1. 2.5.4. Swelling ratio The hydrogels were submerged in simulated body fluid (SBF, pH = 7.4) at a temperature of 37 °C. At set time points (0.5, 1, 2, 4, 8, 16, 24, and 32 h), the samples were extracted. The surface moisture was dabbed with filter paper, and the samples were subsequently weighed precisely to ascertain the swelling ratio. 2.5.5. Adhesive test of the hydrogels To examine the adhesive strength of the HPS@ZCJ hydrogel, wooden sticks were chosen as the substrate in this study. The detailed process is described as follows: Initially, wooden sticks (hydrophilic substrate) measuring 60 × 5 mm were prepared. Next, at room temperature, 20 μL of the hydrogel solution was applied to a 5 × 20 mm area at one end of each wooden stick. The hydrogel-coated ends of the two wooden sticks were then joined together and maintained at 37 °C for 10 min to allow curing. The shear strength was subsequently measured using a universal tensile testing machine (3365 Instron, USA) to evaluate the adhesive performance of the hydrogel. A PF127 hydrogel (20 %) was used as a control. Additionally, the experiment was repeated using iron blocks (hydrophobic substrate). Each test was conducted with three replicates. 2.5.6. In vitro degradation tests The HPS@ZCJ hydrogel was placed in SBF at 37 °C and immersed continuously for 21 days. At specified intervals, the hydrogels were taken out, cleaned with ultrapure water, subjected to freeze-drying, and weighed afterward. The extent of weight loss for the hydrogels was quantified using the formula below: [MATH: Weightlossrate(%)=Wt/Wo×100(%) :MATH] Here, Wo is the initial dry weight before degradation, while Wt is the dry weight after degradation. 2.5.7. Encapsulation efficiency (EE) and in vitro drug release test Precise amounts of JEO were dissolved in PBS (1 % DMSO) to formulate standard solutions with diverse concentrations. The absorbance of these solutions at distinct concentrations was assessed using a UV–Vis spectrophotometer (UV-2600, SHIMADZU, Japan), and calibration curves were generated. For JEO, the peak absorption wavelength was 274 nm. Freshly prepared ZCJ NPs were centrifuged at 15,000g in a refrigerated centrifuge (Thermo, USA) at 25 °C for 30 min. The supernatant was diluted with PBS containing 1 % DMSO, and the absorbance at 274 nm was measured using a UV-2600 spectrophotometer. The content of JEO was quantified against a standard curve of JEO dissolved in PBS (1 % DMSO). The EE was calculated using the following equations: [MATH: EE(%)=AmountofencapsulatedJEOofTotalJEOamount×100(%) :MATH] Moreover, at defined time intervals, 1 mL of the solution was extracted from the centrifuge tubes and placed into separate Eppendorf tubes, with 1 mL of fresh PBS (1 % DMSO) replenished into the solution. The absorbance of the extracted solutions at various time points was monitored using the UV–Vis spectrophotometer, and the outcomes were juxtaposed with the calibration curves. Drug release profiles were constructed to elucidate the kinetics of drug release. 2.6. H22 cancer cells killing effect and biocompatibility assay To begin with, ZCJ NPs at concentrations of 250, 500, 1000, 2000, and 4000 μg/mL were fully dissolved in corresponding volumes of RPMI-1640 (Gibco) culture medium with 10 % fetal bovine serum (FBS). Subsequently, the proliferation of H22 cells (3 × 10^3 cells per well) was evaluated using the Cell Counting Kit-8 (CCK-8) after 1, 2, and 3 days of incubation with ZCJ at these concentrations, with RPMI-1640 alone as the control. Briefly, the CCK-8 reagent was added to the culture medium and incubated with the cells for 1 h. The absorbance was then measured at 450 nm using a microplate reader (Bio-Tek, USA). Based on these findings, the ideal concentration of ZCJ for further cell proliferation studies was identified. H22 cells and human umbilical vein endothelial cells (HUVECs) were plated in 96-well plates (3 × 10^3 cells per well). Once the cells adhered the following day, they were treated with the growing concentration of HPS@ZCJ hydrogel for 48 h. In the same way, the IC[50] values were subsequently assessed using CCK-8 assays. 2.7. Live/dead staining The viability of H22 cells was further evaluated using the Calcein-AM/PI Double Stain Kit according to the manufacturer's protocol. Cells cultured in blank culture medium, ZCJ NPs, HPS, and HPS@ZCJ hydrogel were resuspended and plated into 96-well plates at a density of 3 × 10^3 cells per well. Following a 1, 2, 3-day culture period, the cells were stained with calcein-AM (acetoxymethyl, indicating live cells) and propidium iodide (PI, indicating dead cells) for 30 min, and subsequently imaged using confocal laser scanning microscopy (CLSM). 2.8. Cell apoptosis H22 cells (1 × 10^5 cells/well) were plated into a 6-well plate and exposed to 350 μg/mL of ZCJ, HPS hydrogel, and HPS@ZCJ hydrogel. For controls, cells were cultured in RPMI-1640 with 10 % FBS. Apoptosis in H22 cells was evaluated using the Annexin-V-FITC Apoptosis Detection Kit (Beyotime) according to the manufacturer's protocol. Cells were collected, washed twice with cold PBS (pH = 7.4), centrifuged, and then incubated with annexin-V-FITC/PI at 37 °C for 15 min as per the kit instructions. After staining, the cells were analyzed using a FACS-Calibur flow cytometer and CellQuest software (Becton Dickinson, San Jose, CA). 2.9. Cell cycle assay H22 cells (1 × 10^5 cells/well) were plated into 6-well plates and exposed to ZCJ, HPS hydrogel, and HPS@ZCJ hydrogel (350 μg/mL) at 37 °C for 48 h. Cells maintained in RPMI-1640 with 10 % FBS were used as controls. Post-incubation, the cells were dissociated with trypsin, carefully harvested, and centrifuged at 1000g. The supernatant was carefully aspirated, and the cells were resuspended in pre-cooled PBS (pH = 7.4) before being transferred to a 1.5 mL centrifuge tube. The cells were then fixed in 70 % cold ethanol and stored at 4 °C for 24 h. The cells were subsequently centrifuged, washed twice with cold PBS (pH = 7.4), treated with RNase A (0.1 mg/mL) for 1 h at 37 °C, and stained with PI (0.1 mg/mL) for 30 min in the dark. DNA content was analyzed by flow cytometry (FACSCalibur, BD, USA), and the distribution of cells across different cell cycle phases was evaluated using ModFit software. 2.10. Transwell assay The migration assay was carried out using transwells (8 μm pore size, 24-well plate, BD Biosciences, USA). H22 cells and HUVECs were deprived of serum by incubation in serum-free medium for 24 h. Subsequently, H22 cells and HUVECs (1 × 10^6 cells each) were introduced into the upper chamber of the transwell, with the lower chamber containing migration-inducing medium supplemented with 10 % FBS (HPS, HPS@ZCJ or ZCJ). The cells were allowed to migrate for 24 h. Cells that migrated to the lower chamber were harvested, fixed with 4 % paraformaldehyde for 30 min, and stained with 0.5 % crystal violet (Beyotime Biotechnology, China) for 20 min. Migrated cells were imaged using an optical microscope, and cell numbers were manually counted in each microscopic field. 2.11. Scratch assay HUVECs were plated in 35-mm petri dishes at a density of 1 × 10^6 cells per dish and cultured at 37 °C for 24 h. Once the cells formed a confluent monolayer, a uniform gap was introduced using a 200-μL sterile pipette tip. After PBS rinsing to remove cell debris, the cells were maintained in either plain medium, ZCJ NPs, or medium supplemented with HPS or HPS@ZCJ. The closure of the scratch was observed with an inverted microscope (Olympus IX-73, Japan) at 0-, 12-, and 24-h post-scratch. The scratch area was quantified using ImageJ software (version 1.8.0, NIH, USA), and the wound-healing rate was computed to evaluate the migratory potential of HUVECs. The healing rate was calculated using the formula: Wound-healing rate (%) = A[n]/A[0] × 100 %, where A[0] and A[n] denote the initial wound area and the remaining unhealed area, respectively. 2.12. Cell immunofluorescence staining experiment HUVECs were plated in confocal dishes (JingAn Biological, China) at a density of 1 × 10^3 cells/mL and incubated with 350 μg/mL of ZCJ, HPS, or HPS@ZCJ solutions. After 3 days of incubation, the cells were fixed using 4 % paraformaldehyde for 15 min. They were then permeabilized with 0.1 % Triton X-100 (Abcam, USA) and blocked with 3 % BSA/PBS (Aladdin, China) for 20 and 30 min, respectively, at room temperature. Following PBS (pH = 7.4) rinsing, the cells were stained with a rabbit anti-mouse primary antibody specific for CD31 (1:200 dilution, Abcam, USA) and incubated overnight at 4 °C. The cells were gently washed with PBS (pH = 7.4) and reacted with Cy3-conjugated anti-rabbit IgG secondary antibodies (1:200 dilution, Abcam, USA) for 2 h in the dark. In parallel, cell nuclei were stained with 4,6-diamidino-2-phenylindole dilactate (DAPI, Abcam, USA) for 15 min in the dark. The stained cells were imaged using a laser scanning confocal microscope (Nikon, Japan), and the relative expression levels of VEGF were quantified using ImageJ software. 2.13. Hemolysis test Red blood cells were extracted from BALB/c mice, washed with saline, and diluted. Next, 100 μL of a 4 % (v/v) red blood cell solution was combined with 0.9 mL of saline containing different concentrations of HPS@ZCJ hydrogel. In the negative control, 100 μL of 4 % (v/v) red blood cells was mixed with 0.9 mL of saline, whereas in the positive control, 100 μL of 4 % (v/v) red blood cells was mixed with 0.9 mL of deionized water. Following incubation at 37 °C for 2 h, the absorbance of the supernatant at 540 nm was determined using a microplate reader (iMark, BIO-RAD, USA). The hemolysis rate was calculated using the formula: [MATH: Hemolysisrate(%)=(OD0OD1)/(OD2OD1)×100% :MATH] where OD0 is the optical density (OD) of red blood cells in HPS@ZCJ hydrogel at various concentrations, OD1 is the OD of red blood cells in normal saline, and OD2 is the OD of red blood cells in deionized water. 2.14. In vitro and in vivo hemostasis properties Anticoagulated mouse blood (2000 μL) was combined with PBS, fibrinogen, or HPS@ZCJ in a test tube, followed by the addition of thrombin to induce clotting. After a 5-min incubation, the tubes were tilted to assess clot formation. To further assess the in vitro hemostatic effect of HPS@ZCJ hydrogel, an in vitro clotting test was performed with four groups: i) control; ii) HPS@ZCJ; iii) fibrin glue. The control group was treated with 100 μL of PBS, the HPS@ZCJ group with 100 μL of HPS@ZCJ hydrogel and the fibrin glue group with 100 μL of fibrin glue. Next, 900 μL of mouse blood was added to each group. After 1 min, all liquids were gently removed, and the samples were rinsed three times with PBS. Repeat the above steps, wait 2–9 min, absorb the liquid, and wash. The first time a blood clot appeared in each group was photographed and recorded. The blood clotting index (BCI) was then determined. For the BCI assay, fibrin hydrogel or HPS@ZCJ was prewarmed at 37 °C for 10 min. Then, 9 mL of anticoagulated blood and 1 mL of 0.1 M CaCl2 were applied to the hydrogel. Post a 5-min incubation, unclotted blood was dissolved in 5 mL of deionized water, and the optical density (OD) was measured at 540 nm. As a control, 50 μL of anticoagulated blood was mixed with deionized water. The BCI was calculated using the formula: BCI (%) = (OD of materials/OD of reference) × 100 %. Red blood cell (RBC) adhesion was evaluated by preparing 100 μL of hydrogel with HPS@ZCJ or fibrin in a 96-well plate, to which 50 μL of anticoagulated whole blood was added. The mixture was agitated at 37 °C for 10 min, followed by washing with PBS to remove non-adherent RBCs. The hydrogels were then incubated with deionized water at 37 °C for 30 min to lyse adherent RBCs, after which the OD was measured at 540 nm. RBC attachment was calculated as: RBC adhesion (%) = (OD of sample/OD of reference) × 100 %. All animal procedures were sanctioned by the Institutional Animal Care and Use Committee at Southern University of Science and Technology (SUSTech-JY202411104). In the standard round liver defect model, a 6 mm diameter biopsy needle was utilized to induce a circular wound in the rat liver, reaching a depth of 3 mm. Subsequently, 100 μl of each test sample was administered directly into the defect. The extent and duration of blood loss were then systematically evaluated. 2.15. Animals and ethics statement Male BALB/c mice, 8 weeks old and weighing 16–18g, were sourced from the Southern University of Science and Technology and maintained under Specific Pathogen Free (SPF) conditions. These mice were kept in a specialized facility with controlled temperature and humidity, exposed to a 12-h light/12-h dark cycle, and provided with unrestricted access to water and standard rodent diet. The study's protocols were sanctioned by the Institutional Animal Care and Animal Ethics Committee of the Southern University of Science and Technology (approval no. SUSTech-JY202411104). All animal experiments adhered to the local animal welfare regulations and guidelines set by the Southern University of Science and Technology. 2.16. In vivo antitumor effects of HPS@ZCJ hydrogel To assess the in situ antitumor activity of HPS@ZCJ hydrogel, Balb/c mice were randomly divided into six groups (n = 9): sham operation, control, ZCJ, HPS, HPS@ZCJ, and doxorubicin (DOX). H22 cells (1 × 10^6 cells/mL) were suspended in PBS (pH = 7.4), and 100 μL was injected into the livers of the mice (H22 cells were not injected in the sham operation group). The mice in each group were then treated as follows: i) and ii) no treatment; iii) application of 100 μL ZCJ NPs at the inoculation site; iv) application of 100 μL HPS hydrogel at the inoculation site; v) application of 100 μL HPS@ZCJ hydrogel at the inoculation site; vi) application of 100 μL DOX (2 mg/kg) at the inoculation site. The body weight of the animals was also recorded every 2 days. On day 7 after treatment, the mice (n = 3 in each group) were euthanized, and the tumor-bearing livers were photographed and weighed. After treatment, residual HPS@ZCJ hydrogel remained adhered to the liver surface. Moreover, the survival rates of mice in each group (with six mice per group) were tracked for 30 days, and Kaplan-Meier survival curves were generated. 2.17. In vivo recurrence prevention effect of HPS@ZCJ hydrogel after tumor resection To assess the antirecurrence potential of HPS@ZCJ hydrogel in vivo, a subcutaneous HCC recurrence model was developed using BALB/c mice. H22 cells (1 × 10^6 cells in 0.1 mL PBS) were injected subcutaneously into the lateral right lower abdominal wall of the mice. The body weight of the animals was recorded on days 0, 3, and 7. One week after tumor cell implantation, when the tumor volume reached approximately 150 mm^3, mice were anesthetized using isoflurane (up to 5 % for induction and 1–3 % for maintenance) in an induction chamber, with anesthesia maintained via a nose cone. Subsequently, all visible tumors were surgically excised using sterile instruments, and subsequent treatments were administered. Mice were randomly allocated into five groups (n = 12): i) resection only; ii) resection and HPS hydrogel treatment; iii) resection and ZCJ NPs treatment; iv) resection and DOX treatment; v) resection and HPS@ZCJ hydrogel treatment. Additionally, the amount of blood loss during the surgical removal of the tumor was recorded. Post-surgery, the body weight of the mice was tracked. On day 14 post-surgery, the heart, liver, spleen, lungs, kidneys and recurrent tumors from all mice (n = 6 in each group) were harvested for follow-up experiments. The tumors were photographed and weighed. Tumor size was calculated using the formula: width [[80]2] × length × 0.5. After treatment, a portion of the HPS@ZCJ hydrogel persisted at the tumor-resection site; because the hydrogel is fully biodegradable, it can be left in place, eliminating the need for secondary surgery to remove residual material and markedly reducing the associated operative risk. Additionally, the survival of mice in each group (n = 6 in each group) was monitored over 31 days, and Kaplan-Meier survival curves were constructed. 2.18. Hematoxylin and eosin (H&E) staining To determine the potential toxicity of HPS@ZCJ hydrogel on major organs and the pathological conditions of tumors in mice, histopathological evaluations were carried out on the heart, liver, spleen, lungs, kidneys, and tumors. Under a protocol approved by our Institutional Animal Care and Use Committee, mice from each group were humanely euthanized at the end of the experiment via cervical dislocation. The heart, liver, spleen, lungs, kidneys, and tumors were promptly excised and fixed in 4 % paraformaldehyde for 48 h. The fixed tissues were then dehydrated with 30 % sucrose solution and embedded in paraffin using standard protocols. Tissue sections (5 μm thick) were stained with hematoxylin and eosin (H&E; C0105M, Beyotime). The stained sections were examined under a light microscope (Leica Microsystems, Germany) to evaluate tissue morphology changes. 2.19. Immunohistochemical staining Tumor tissue samples were fixed with 10 % neutral buffered formalin (Sigma-Aldrich, USA) and embedded in paraffin. Subsequently, the samples were processed for deparaffinization and rehydration. They were then incubated overnight at 4 °C with primary antibodies targeting Ki-67 (1:200, Abcam, USA), TUNEL (1:200, Abcam, USA), DHCR7 (1:200, SAB, USA), GPNMB (1:200, Proteintech, China), and CD-31 (1:200, Abcam, USA). On the following day, the samples were treated with biotinylated secondary antibodies for 30 min at 37 °C. Tissue sections were visualized using the Pierce™ DAB Substrate Kit (34002, Thermo Fisher, USA) and examined under an optical microscope. 2.20. Immunofluorescent staining The tumor microenvironment significantly influences tumor growth. Consequently, we examined the impact of HPS@ZCJ hydrogel on immune cells in tumor tissues via immunofluorescence staining. Tumor tissues were sliced, affixed to slides, and stained with primary antibodies specific for CD206 and CD8 (1:200, Abcam, USA) for 12 h. Fluorescence-labeled secondary goat anti-rat antibodies were subsequently added to enhance signal detection. Nuclei were stained with DAPI. The sections were then imaged using a Zeiss fluorescence microscope. 2.21. Human tumor tissue samples The protocol for obtaining human hepatocellular carcinoma tissue samples was approved by the Ethics Review Committee of the Guangdong Provincial People's Hospital (No. KY2024-1105-01). All samples were collected with informed consent from patients, in accordance with the International Ethical Guidelines for Biomedical Research Involving Human Subjects (CIOMS). 2.22. Isolation and culture of human primary HCC cells Fresh tumor samples were rinsed three times with DMEM to remove blood and other impurities. The cleaned tumor samples were then minced into fragments of approximately 1 mm^3 and digested with Hank's Balanced Salt Solution (HBSS) (Gibco, Carlsbad, CA, USA) and 0.1 % Type IV collagenase (Gibco, Carlsbad, CA, USA) at 37 °C for 1–2 h. The digested samples were filtered through a 100 μm nylon filter and centrifuged at 4 °C for 3 min. The supernatant was discarded, and the cells were washed twice with HBSS and finally resuspended in hepatocyte culture medium. After seeding, the medium was replaced with fresh medium containing different concentrations of peptide after 24 h to remove dead cells and debris. 2.23. RNA sequencing analysis (RNA-seq) Human HCC cell samples (P1) were randomly divided into two groups and co-cultured with either blank culture medium or HPS@ZCJ hydrogel for 5 days. Total RNA was then extracted from the samples and its integrity was assessed using the RNA Nano 6000 Assay Kit (Agilent Technologies, CA, USA, 5067-1511). mRNA libraries were constructed following the standard operating procedure with Novogene software. Indexed samples were clustered on the cBot Cluster Generation System using the TruSeq PE Cluster Kit v3-cBot-HS (Illumina). After clustering, library preparation was sequenced on the Illumina Novaseq platform, generating 150 bp paired-end reads. To ensure the reliability and reproducibility of the results, three independent experiments were performed (n = 3). Differentially expressed genes (DEGs) were detected from RNA-seq data using the R package limma (V.3.56.2). DEGs were identified based on a log2 fold change (log2FC) exceeding 1 or below −1, and an adjusted p-value <0.05 after Bonferroni correction. Subsequently, these DEGs were analyzed for enrichment in Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) using the R package clusterProfiler (V.4.8.3). 2.24. Statistical analysis Data are presented as individual values with the mean ± standard error of the mean. Comparisons between multiple groups were made using one-way ANOVA, while Student's t-test was used for comparisons between two groups. Survival analysis was assessed by the log-rank test. All statistical analyses were performed using GraphPad Software, with P < 0.05 indicating statistical significance. 3. Results and discussion 3.1. Preparation and characterization of ZCJ NPs loaded with JEO The chemical constituents of JEO were analyzed using HRLC-MS and GC-MS techniques, with the results presented in [81]Fig. 2A–C. Based on existing research, Dehydrocostus lactone, Curdione, Germacrone, and Neocurdione—all identified within JEO—possess significant anticancer properties [[82][32], [83][33], [84][34]]. Thus, it is indicated that these compounds are the primary antitumor active components of JEO. Fig. 2. [85]Fig. 2 [86]Open in a new tab Characterization of ZCJ NPs. (A–C) Schematic illustration of JEO's chemical constituents analyzed via HRLC-MS and GC-MS. (D–F) Physicochemical properties of ZCJ NPs in different proportions, including particle size, PDI, and zeta potential. (G) Morphological examination of ZCJ NPs via SEM and TEM, demonstrating spherical shape and layered structure (Sscale bar: 120 nm). (H) Physicochemical properties of zein@JEO, zein-CS, and ZCJ NPs. (I) Particle size distribution of ZCJ NPs. (J) FTIR spectroscopy confirmed the successful combination of zein and CS in ZCJ NPs and the encapsulation of JEO. Zein-based NP delivery systems are widely utilized in tumor therapy due to their ability to encapsulate hydrophobic compounds [[87]35]. Zein, a natural protein carrier rich in hydrophobic amino acids, effectively encapsulates hydrophobic drugs [[88]36]. However, the stability of these NPs often requires enhancement. To address this, CS, a natural anionic polysaccharide, is employed as a stabilizer due to its negative charge, which helps protect the NPs. In the study, we developed a stable NP system by loading the hydrophobic compound JEO into zein and coating it with chondroitin sulfate through electrostatic interactions and hydrogen bonding, forming ZCJ NPs. The physicochemical properties of JEO-loaded NPs are summarized in [89]Fig. 2D–J. Our preliminary experiments investigated the effect of varying the zein/CS ratio on NP size. As shown in [90]Fig. 2D–F and [91]Table S1, the ZCJ-1 formulation (zein to CS ratio of 5:1) exhibited the smallest particle size (141.8 ± 1.1 nm) and polydispersity index (PDI, 0.233 ± 0.127). The negative charge of CS resulted in a negative zeta potential for ZCJ NPs (−30.11 ± 0.66 mV). SEM and TEM images revealed that the NPs were spherical, with distinct inner and outer layers ([92]Fig. 2G–I and [93]S1A). These ZCJ-1 NPs achieved high drug loading efficiency, with an EE (%) of 94.7 ± 2.8 % ([94]Fig. 2H). On the other, zein and CS had their characteristic peaks. Zein showed N-H bending vibration and C-N stretching vibration of the secondary amide at 1529 cm^−1 28, while CS showed C single bond O single bond C stretching vibration at 1031 cm^−1 37. Notably, the 1031 and 1529 cm^−1 absorption peaks could be detected in ZCJ and Zein@CS NPs ([95]Fig. 2J). In addition, CS exhibited a peak at 3244 cm^−1 due to overlapping vibrations of –NH and –OH groups, while zein showed a narrower peak at 3301 cm^−1 attributed to –OH vibrations [[96]28,[97]37]. In the spectrum of ZCJ NPs, the –OH groups exhibited a slight shift to 3282 cm^−1, likely due to the formation of hydrogen bonds between zein and CS [[98]38]. The peak shape of ZCJ was similar to that of Zein@CS, indicating that JEO was encased in the interior. 3.2. Preparation and characterization of HPS@ZCJ hydrogel In this study, we developed the HPS@ZCJ hydrogel based on PF127, HMC, and SA for hemostasis of liver hemorrhage, and integrated JEO-loaded CS-modified zein NPs (ZCJ) into the hydrogel system as a functional nanoreinforcing filler to prevent postoperative tumor recurrence. To fulfill the requirements of facile preparation, biocompatibility, robust drug-loading capacity, and sustained drug release, we selected PF127 hydrogel as the carrier material. PF127, a polymeric nonionic surfactant comprising 70 % polyethylene oxide and 30 % polypropylene oxide, exhibits thermosensitive behavior, transitioning from a sol to a gel state at its lower critical solution temperature due to hydrophobic-hydrophilic interactions between its components [[99]27]. To enhance the duration and effectiveness of hemostasis while improving sustained drug release and stability, we incorporated HMC into the hydrogel and introduced SA as a double-crosslinked network carrier with PF127, with HMC also functioning as a thickener. The HPS@ZCJ hydrogel was specifically designed for injection to ensure precise delivery to the surgical site. This method allows for controlled application and ensures that the hydrogel adheres to the tissue, providing sustained release of the therapeutic agents. The thermosensitive nature of the hydrogel ensures that it transitions from a liquid to a solid state at body temperature [[100]39], further enhancing its ability to remain in place and deliver the JEO effectively. The as-prepared HPS@ZCJ hydrogel was able to be extruded from a syringe through a 21G needle ([101]Fig. S1B). Characterization studies revealed that the HPS@ZCJ hydrogel transitions from a flowing sol state at 4 °C to a solid gel state at 37 °C. This thermoresponsive behavior was confirmed in vitro by injecting the HPS@ZCJ solution into a 37 °C water bath, where it rapidly formed a viscoelastic gel capable of adhering to surfaces and tissues without flowing ([102]Fig. 3A). To ensure a stable controlled-release microenvironment, hydrogels must possess adequate mechanical and adhesive strength. As shown in [103]Fig. 3B, the HPS@ZCJ hydrogel can adhere the liver tissue of a rat to the tip of a finger. We further evaluated these properties using a lap shear test. As shown in [104]Fig. 3C, the adhesive strength of hydrogels composed solely of PF127 was minimal. In contrast, the HPS@ZCJ hydrogel demonstrated remarkable adhesive strength, reaching up to 28.89 kPa on iron blocks (hydrophobic surfaces) and up to 37.17 kPa on wooden sticks (hydrophilic surfaces). These unique thermal and adhesive characteristics offer practical advantages. The hydrogel can be injected at low temperatures, uniformly covering the liver surface. Upon injection, it solidifies at body temperature, preventing displacement by wound exudate. This ensures secure mechanical adhesion and sustained drug release at the wound site, facilitating hemostasis and anti-tumor effects. [105]Fig. S1C illustrates a real application scenario. The image shows an isolated rat's liver, with the left side untreated and the right side uniformly coated with HPS@ZCJ. The bottom image depicts the in vivo application, with HPS@ZCJ stained red for visualization. Besides, SEM analysis revealed the microstructural differences between HPS@ZCJ and HPS. As shown in [106]Fig. 3D, HPS@ZCJ exhibited a porosity of 43.6 %, compared to 52.1 % for HPS, indicating smaller and more uniformly distributed pores in HPS@ZCJ, whereas HPS had larger and more numerous pores. This pore distribution pattern likely results from the incorporation of ZCJ NPS in HPS@ZCJ, which enhances intermolecular interactions and increases cross-linking density, thereby impeding water evaporation during freeze-drying. We next probed the synergistic interactions among PF127, HMC, and SA ([107]Fig. S2A). In the FTIR analysis of SA, the characteristic band at 1606 cm^−1 is indicative of the asymmetric stretching vibrations of carboxylate (–COO^-) groups [[108]40]. In the HPS hydrogel, the hydroxyl group peak of HMC, originally at 3460 cm^−1, broadens and shifts to 3458 cm^−1, suggesting that these hydroxyl groups are engaged in hydrogen bonding. Concurrently, the carboxylate ion peak at 1610 cm^−1 in SA is markedly attenuated in the HPS hydrogel spectrum, which implies the formation of hydrogen bonds between the hydroxyl groups of HMC and the carboxylate ions of SA. Moreover, PF127 exhibits a peak at 1103 cm^−1, corresponding to the C-O-C stretching, while HMC displays this peak at 1056 cm^−1 [[109]41]. Within the HPS hydrogel, the C-O-C peak shifts to 1095 cm^−1, indicating hydrogen bond formation between HPMC and PF127, which results in the observed peak shift. The presence of characteristic peaks for PF127 (1095 cm^−1) and SA (1610 cm^−1) in the hydrogel confirms the establishment of an interpenetrating double cross-linked hydrogel network. In addition, from the spectra of [110]Fig. 3E, it is important to highlight the absence of a band between 1505 and 1545 cm^−1 characteristic of amide II in the HPS group [[111]28]. This observation is attributable to the proteinaceous nature of zein: the sharp, intense band at 1523 cm^−1 present in zein ([112]Fig. 2J), ZCJ and HPS@ZCJ (1505 cm^−1) confirms successful encapsulation of ZCJ NPs within the hydrogel. Fig. 3. [113]Fig. 3 [114]Open in a new tab Characterization of the HPS@ZCJ hydrogel. (A) Assessed fluidity at 4 °C and 37 °C to demonstrate temperature-responsive behavior. (B) Photographs depicting HPS@ZCJ hydrogel adhesion to rat liver. (C) Schematic and quantitative results of hydrogel adhesion tests. (D) SEM images detailing the structural and morphological features of HPS@ZCJ hydrogel. (E) FTIR spectra comparing HPS@ZCJ, ZCJ, and HPS. (F) Rheological analysis highlighting the viscoelastic properties influenced by temperature. (G) Frequency scanning outcomes for HPS and HPS@ZCJ composites. (H) Cumulative in vitro release profile of JEO from HPS@ZCJ. (n = 3). Rheological analysis revealed that the critical phase transition temperature of the HPS@ZCJ hydrogel is approximately 25.6 °C ([115]Fig. 3F). The sol-to-gel transition is rapid and smooth, occurring swiftly once the gelation temperature is exceeded. Below this temperature, the hydrogel exhibits liquid-like behavior, characterized by a loss modulus (G″) greater than the storage modulus (G′). Above 25.6 °C, G′ exceeds G″, indicating solid-like behavior. Given that operating room temperatures are typically around 26 °C, the HPS@ZCJ hydrogel, stored at 4 °C, requires a brief period to reach its critical transition temperature. This delay prevents premature gelation, facilitating precise injection during clinical procedures. Frequency scanning results ([116]Fig. 3G) revealed that both HPS@ZCJ and HPS hydrogels exhibited G′ higher than their G″, with minimal changes observed with increasing frequency. This indicates a stable crosslinking network at room temperature (25 °C). Specifically, HPS@ZCJ had a G′ of approximately 6.3 kPa and a G″ of 1.3 kPa, whereas HPS, lacking ZCJ NPs, had lower values of 4.1 kPa and 0.8 kPa, respectively. These findings suggest that the incorporation of ZCJ NPs significantly alters the hydrogel structure, enhancing its mechanical properties. Furthermore, the HPS@ZCJ hydrogel exhibited shear-thinning behavior, indicating that its viscosity decreased under applied shear stress. This property facilitates the smooth extrusion of the HPS@ZCJ hydrogel through medical needles, ensuring ease of injection during clinical procedures ([117]Fig. S3A). This property complements the hydrogel's thermosensitive transition to a solid-like state at body temperature, which ensures stable application and sustained release of therapeutic agents. 3.3. JEO sustained release, swelling behavior, and in vitro degradation of HPS@ZCJ hydrogel To accurately quantify the sustained release capacity of the HPS@ZCJ hydrogel, standard curves of JEO were established ([118]Fig. S2B). These curves facilitated the determination that the cumulative release of JEO from HPS@ZCJ in a pH 7.4 SBF environment reached approximately 89.7 % within 6 days ([119]Fig. 3H). This substantial release profile confirms the hydrogel's efficacy in achieving sustained drug delivery, which is critical for maintaining therapeutic concentrations over extended periods. These results highlight the rational design of HPS@ZCJ hydrogel with its ideal release kinetics and sustain an enduring anti-HCC effect. Excessive swelling of hydrogels can compress surrounding tissues, blood vessels, and nerves, potentially causing discomfort and severe side effects. Moreover, such swelling can diminish the hydrogel's cohesion, resulting in inadequate mechanical and adhesive strength [[120]42]. Therefore, we evaluated the swelling behavior of the HPS@ZCJ hydrogel. As shown in [121]Fig. S3B, the swelling ratio of HPS@ZCJ gradually increased, reaching equilibrium at approximately 131 % after 16 h. This indicated that the HPS@ZCJ hydrogel has good anti-swelling properties and does not exert pressure on surrounding tissues post-injection. Furthermore, for optimal in vivo performance, the hydrogel should exhibit gradual biodegradability to sustain drug release effectively and be fully absorbed by the body. [122]Fig. S3C illustrates that the HPS@ZCJ hydrogel underwent gradual degradation in SBF, with 74.9 % remaining after 7 days and essentially degrading by day 21. This profile parallels the inflammatory and proliferative phases of hepatic wound healing, which typically span 7–21 days after partial hepatectomy [[123]43]. Additionally, degradation releases only naturally occurring metabolites-salts of alginate, hydroxymethyl cellulose oligosaccharides, and plant-derived essential-oil constituents-whose benign profiles are well documented. Consequently, the hydrogel is expected to clear from the surgical site before the onset of the remodeling phase, ensuring both safety and compatibility with the normal healing trajectory. 3.4. In vitro antitumor effects of HPS@ZCJ hydrogel We first studied the antitumor effects of ZCJ NPs on H22 cells by using CCK-8. ZCJ was at different concentrations (0–4000 μg/mL) and cocultured with H22 cells for 1–3 days. As shown in [124]Fig. 4A, ZCJ remarkably inhibited H22 cells in a dose-dependent manner. Moreover, the cytotoxicity of HPS@ZCJ hydrogel was assessed against H22 cells and HUVECs. The IC50 values (the dose required to inhibit 50 % cellular growth within 24 h) were found to be 1848.1 μg/mL for HUVECs and 361.6 μg/mL for H22 cells ([125]Fig. 4B and C). This indicated that HPS@ZCJ hydrogel is less cytotoxic to normal cells than to the tested cancerous cells. Consequently, guided by the IC50 results, a concentration of 350 μg/mL was selected for subsequent cellular experiments. The live/dead staining outcomes corroborated the aforementioned findings, indicating that the HPS@ZCJ hydrogel significantly impedes the proliferation of H22 cells ([126]Fig. 4D and E). Moreover, for biological materials applied directly to the human body, biocompatibility is a fundamental requirement. For hemostatic materials, hemocompatibility testing is crucial to ensure no hemolysis occurs during hemostasis. As shown in [127]Fig. 4F, the hemolysis ratio of HPS@ZCJ hydrogel was evaluated over a concentration range of 0–2000 μg/mL. The embedded image reveals that up to 1000 μg/mL, the solution color remained largely unchanged, indicating minimal hemolysis and aligning with the negative control. This concentration range is commonly accepted as indicative of good hemocompatibility. However, at 2000 μg/mL, the hemolysis ratio exceeded 10 %, suggesting a potential risk at higher concentrations. Fig. 4. [128]Fig. 4 [129]Open in a new tab (A) Cell cytotoxicity of H22 cells treated with ZCJ NPs in different concentrations by CCK-8 assay. (B, C) The IC50 values for HUVECs and H22 cells were calculated from nonlinear regression analyses plotting the percentage of specific cytotoxicity against the Log10 concentration of the HPS@ZCJ hydrogel. (D) The Live/Dead cell staining result of H22 cells treated with ZCJ NPs, HPS and HPS@ZCJ hydrogels after 3 days of culture. (E) Quantitative analysis of the Live/Dead cell staining. (F) Hemocompatibility evaluations of HPS@ZCJ hydrogel in different concentrations. (n = 3, ∗ and # represent P < 0.05 by comparing with the control and HPS groups, respectively). To further assess the antitumor efficacy of HPS@ZCJ hydrogel, three groups were established based on their distinct compositions: ZCJ NPs, HPS, and HPS@ZCJ hydrogel. Flow cytometry was initially employed to measure apoptosis rates in H22 cells across these groups. The HPS@ZCJ group exhibited an apoptosis rate of 37.55 ± 3.8 %, whereas ZCJ NPs achieved a rate of 42.3 ± 4.8 % ([130]Fig. 5A). This difference is likely due to the sustained-release profile of HPS@ZCJ hydrogel, which prevents the rapid release of the maximum dose within a short period. Given that HPS hydrogel alone does not induce apoptosis ([131]Fig. 5B), the cytotoxicity observed in [132]Fig. 4D and E indicates that HPS hydrogel may promote tumour-cell death via non-apoptotic pathways. We further explored the impact of HPS@ZCJ hydrogel on cell cycle progression in H22 cells ([133]Fig. 5C and D). Following treatment with HPS@ZCJ hydrogel, the proportion of H22 cells in the G0/G1 phase significantly increased from 41.01 % to 52.73 % (P < 0.05). Concurrently, the percentage of cells in the S phase decreased from 34.82 % to 29.18 % (P < 0.05), and that in the G2/M phase decreased from 24.04 % to 18.09 % (P < 0.05). These findings demonstrate that HPS@ZCJ hydrogel induces G0/G1 phase arrest in H22 cells. Transwell chamber assays were employed to evaluate and quantify the migratory behavior of H22 cells on hydrogels. The results, as shown in [134]Fig. 5, indicated that after 48 h, both the ZCJ and HPS@ZCJ groups significantly impeded H22 cell migration compared to the control and HPS groups (P < 0.05, [135]Fig. 5E–G). Collectively, these findings highlight the significant antitumor potential of HPS@ZCJ hydrogel, which can induce apoptosis, enhance cell cycle arrest, inhibit cell proliferation, and impede cell migration. Fig. 5. [136]Fig. 5 [137]Open in a new tab In vitro anti-tumor ability of HPS@ZCJ hydrogel. (A) Flow plot showing the cell apoptosis rate in different groups with an Annexin V/PI apoptosis detection kit. (B) Flow cytometry quantification of the proportion of cell apoptosis rate in different groups. (C) Flow cytometry analysis of the H22 cells cycle after 2 days of treatment with HPS@ZCJ hydrogel. (D) The quantification of the cell cycle test. (E, F) Transwell was used to detect the effect of HPS@ZCJ hydrogel on the migration of H22 cells and HUVECs, respectively. (G) Quantitative analysis of the transwell experiment. (H) Representative fluorescence images of differentially treated HUVECs after CD31 staining. (n = 3, Data are mean ± SD; ∗ and # represent P < 0.05 by comparing with the control and HPS groups, respectively). 3.5. HPS@ZCJ hydrogel inhibits endothelial cell functions Throughout the angiogenesis of tumor progression, the migration and invasion of endothelial cells are crucial processes [[138]44]. However, when stimulated by HPS@ZCJ, HUVECs exhibited minimal migration to the lower compartment of the filter ([139]Fig. 5F and G). In contrast, HPS hydrogel alone had negligible effects on cell migration. A wound healing assay also demonstrated that HPS@ZCJ hydrogel inhibited the migration of HUVECs, as indicated by shorter migration distances ([140]Fig. S4). We further investigated the expression of CD31 in HUVECs cultured with various samples. CD31, a key marker of angiogenesis, is implicated in the proliferation, migration, and vasculogenesis of HUVECs [[141]44]. Our results indicated that HUVECs cultured with HPS@ZCJ displayed weaker green fluorescence intensity, signifying a substantial reduction in CD31 protein expression compared to other groups ([142]Fig. 5H). Semi-quantitative analysis of CD31 expression further corroborated that the CD31 level in the HPS@ZCJ group was significantly lower than in other groups ([143]Fig. S5). Collectively, these findings demonstrate that HPS@ZCJ exerts anti-angiogenic effects by inhibiting CD31-mediated pathways. 3.6. Hemostatic effect of the HPS@ZCJ hydrogel The hemostatic properties of HPS@ZCJ were evaluated using a tube tilting experiment ([144]Fig. 6A). Both the fibrin hydrogel and HPS@ZCJ were able to coagulate blood and maintain their shapes in a solid state. This hemostatic ability was further confirmed by a clotting experiment in a 24-well plate ([145]Fig. 6B and C), which demonstrated significantly shorter clotting times for the fibrin and HPS@ZCJ hydrogels compared to the PBS group. As expected, the 5-min BCIs of the fibrin and HPS@ZCJ hydrogels were 26.71 ± 11.39 % and 28.13 ± 10.62 %, respectively ([146]Fig. 6D; P > 0.05). The red blood cell adhesion rates were 49.26 ± 2.04 % and 48.64 ± 2.29 %, respectively, further indicating comparable hemostatic abilities between the fibrin hydrogel and HPS@ZCJ ([147]Fig. 6D). Fig. 6. [148]Fig. 6 [149]Open in a new tab Hemostatic Evaluation of HPS@ZCJ Hydrogel. (A) Inversion test comparing PBS, fibrin gel, and HPS@ZCJ hydrogel. (B, C) Time-dependent clot formation for PBS, fibrin gel, and HPS@ZCJ hydrogel. (D) Blood-clotting index and red blood cell attachment for PBS, fibrin gel, and HPS@ZCJ hydrogel. (E, F) Schematic and photographic representation of HPS@ZCJ hydrogel application in a rat liver resection model. (G) Quantification of blood loss and hemostasis time across various treatment groups. Data are mean ± SD; ∗ <0.05. (For interpretation of the references to color