Abstract Bioenergetic therapy based on tumor glucose metabolism is emerging as a promising therapeutic modality. To overcome the poor bioavailability and toxicity of arenobufagin (ArBu), a MOF-derived intelligent nanosystem, ZIAMH, was designed to facilitate energy deprivation by simultaneous interventions of glycolysis, OXPHOS and TCA cycle. Herein, zeolitic imidazolate framework-8 was loaded with ArBu and indocyanine green, encapsulated within metal–phenolic networks for chemodynamic therapy and hyaluronic acid modification for tumor targeting. ZIAMH nanoparticles can release ArBu in the tumor microenvironment for chemtherapy, and ICG enables photothermal therapy under near-infrared laser irradiation. In vitro and in vivo mechanism studies revealed that the ZIAMH nanoplatform downregulated glucose metabolism related genes, resulting in the reduction of energy substances and metabolites in tumors. Additionally, it significantly promoted cell apoptosis by upregulating pro-apoptotic proteins such as Bax, Bax/Bcl-2, cytochrome C. Animal studies have shown that the tumor inhibition efficiency of ZIAMH nanomedicines was three fold higher than that of free drugs. Therefore, this study provides a new strategy for glucose metabolism-mediated bioenergetic therapy and PTT/CDT/CT combined therapy for tumors. Graphical Abstract [50]graphic file with name 12951_2024_3084_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-03084-1. Keywords: Arenobufagin, Nanomedicine, Glycolysis, Oxidative phosphorylation, Tricarboxylic acid cycle, Bioenergetic therapy, Integrated photothermal-chemodynamic-chemotherapy Introduction Metabolism is the essential characteristic and material basis of cell life. Unlike normal cells, tumor cells undergo metabolic reprogramming during differentiation and proliferation, such as changes in glucose metabolism. In 1920s Otto Warburg reported that tumor cells mainly produce adenosine triphosphate (ATP) through glycolysis even under aerobic conditions (Warburg effect) [[51]1]. Tumor cells have higher glucose uptake compared to normal cells [[52]2]. Metabolic intermediates produced by glycolysis promote tumor growth, invasion, and metastasis the produced lactic acid can damage normal tissue structure and lowers the pH of the tumor microenvironment (TME), preventing the tumor from being attacked by immune cells and enhancing the “fighting capacity” of tumor cells [[53]3–[54]5]. At the same time, glycolysis combines with the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in cancer cells to jointly provide energy for cancer cell proliferation [[55]6, [56]7]. Over the past few decades, inhibitors have been developed to target glucose metabolism pathway exhibiting prolonged survival rate of experimental animals, some of these drugs are in clinical trial stages [[57]8–[58]10]. However, due to the heterogeneity and complexity of tumors, these new drugs showed limited efficacy and cannot prevent the progression of cancer. In addition, traditional drug chemotherapy and emerging monotherapies such as photothermal and chemodynamic therapy have certain limitations in the TME [[59]11, [60]12]. For example, tumor cells treated with PTT abnormally express multiple heat shock proteins (HSPs), such as HSP70 and HSP90, to resist thermal damage, resulting in insufficient tumor cell apoptosis. These proteins are ATP-dependent and protect cancer cells from the cytotoxic effects of heat generated during PTT [[61]13]. Glycolysis is an important pathway for cancer cells to produce ATP, which can promote cancer cell metastasis [[62]14–[63]16]. Inhibition of glycolysis reduces the expression of HSPs, inhibits tumor growth and metastasis, and enhances the therapeutic effect of PTT. Therefore, development of multifunctional nanodrugs that target cancer cell for treatment may be a promising direction. In the few decades, more and more traditional Chinese medicine extracts are showing efficient activity in tumor treatment [[64]17–[65]19]. Arenobufagin (ArBu) is a monomer extracted from secretions of Bufonis Venenum, with a content up to 2%. It is the main active components of Chinese medical anti-tumor preparation Huachansu injection [[66]20, [67]21]. The potent anti-tumor effect of ArBu and the mode of action behind the cytotoxic characteristics have been reported in previous studies [[68]22–[69]24]. Zhang et al.[[70]25] found that ArBu can induce apoptosis in HepG2 and its drug-resistant strain HepG2/ADM via the mitochondrial pathway, namely, the down-regulation of some key proteins in the PI3K/Akt/mTOR signaling pathway. In addition to apoptosis induction, ArBu may also play an antagonistic role in metabolic reprogramming mediated by mTOR, which means that ArBu can not only directly kill tumor cells, but also can inhibit their growth by regulating the metabolic patterns of tumor cells [[71]25]. However, the potent Na/K-ATPase binding activities, low solubility and poor bioavailability of ArBu limited its anticancer function [[72]26, [73]27], which requires the development of appropriate cargo nanomaterials to explore its clinical applications. Zeolitic imidazolate framework-8 (ZIF-8) is the most typical metal–organic framework materials (MOFs) in the ZIF family, which is coordinated by Zn^2+ cations and 2-methylimidazole anions [[74]28]. Since zinc is an ample transition metal in human body and the side chain of histidine incorporates imidazole, ZIF-8 exhibited thermal stability and safety [[75]29]. In addition, due to its pH responsiveness, ZIF-8 is easy to decompose under the acidic conditions of the TME, leading to the release of incorporated drugs [[76]30]. To date, multiple nano-drug delivery systems containing ZIF-8 have been developed [[77]31–[78]33]. These systems can combine with existing tumor treatment methods such as photothermal and photodynamic therapies [[79]34]. PTT is an effective cancer treatment method, which can directly and precisely treat local areas in a non-invasive manner [[80]35]. Since tumor cells exhibit high thermal sensitivity, high temperature can destroy tumor cell proteins or increase natural drug release from nanocarriers, thereby achieving chemosensitization [[81]36, [82]37]. These combinations are expected to improve treatment outcomes and reduce side effects. Metal–phenolic networks (MPNs) are self-assembled from metal ions and polyphenol ligands. Polyphenols are widely found in natural plants, which have antioxidant and anti-tumor effects [[83]38, [84]39]. Due to their good biocompatibility and excellent adhesive ability, MPNs can effectively encapsulate nanoparticles with various structure [[85]40]. In tumor microenvironment, MPNs initiate Fenton or Fenton-like reactions to convert weak oxidizing H[2]O[2] into strong oxidizing hydroxyl radical (·OH) [[86]41, [87]42]. This conversion leads to increased intracellular oxidation levels, triggering a series of reactions such as DNA necrosis, protein inactivation and lipid oxidation, ultimately inducing apoptosis of cancer cells. Herein, based on our group’s previous review of nanomedicines modulating cancer metabolism [[88]43], we constructed a versatile ZIF-8@ArBu&ICG@MPN@HA (ZIAMH) nanosystem to regulate glucose metabolism in the TME, i.e., simultaneously suppress glycolysis, OXPHOS and tricarboxylic acid (TCA) cycle, thereby inhibiting tumor growth and metastasis. As shown in scheme [89]1, MOFs-derived ZIF-8 loaded with ArBu and indocyanine green (ICG) were first prepared. Subsequently, a metal-polyphenol network self-assembled from ferric iron and gossypol encapsulated the above material for chemodynamic therapy (CDT), and followed by coating with a hyaluronic acid (HA) film to confer the targeting abilities of ZIAMH. During cancer therapy, ZIAMH nanoparticles could target to the tumor sites under the action of HA to the overexpressed CD44 receptors of cancer cells. The ArBu encapsulated in ZIF-8 was able to exert chemotherapy (CT) while enhancing its solubility and reducing its toxicity. Besides, the Fe^3+ in the ZIAMH nanomaterials was released and reduced to Fe^2+ by gossypol, which exhibited enhanced CDT effects based on the Fenton reaction to generate ·OH in the tumor tissue. Under 808 nm near-infrared (NIR) laser irradiation, ICG-loaded ZIAMH nanometarials exhibited photothermal conversion capabilities and can kill cancer cells for photothermal therapy (PTT). Combining the above multiple advantages, ZIAMH could synchronously suppress tumor cell glycolysis and OXPHOS by inhibiting the activities of the key enzymes in the process of glucose metabolism, such as hexokinase 2 (HK2), pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA). At the same time, it can further damage mitochondria functions by depriving the TCA cycle of energy substances and metabolites. Consequently, this multifunctional ZIAMH nanoplatform is expected to combine the multiple therapeutic effects of bioenergetic therapy with PTT/CDT/CT therapy, providing an effective strategy for tumor treatment through precise energy deprivation and targeted therapy. Scheme 1. [90]Scheme 1 [91]Open in a new tab Schematic diagram of the synthesis process of ZIAMH and its therapeutic mechanism of interference tumor glucose metabolism (glycolysis, OXPHOS and TCA cycle) through enhanced bioenergetic therapy integrated with PTT/CDT/CT therapy Materials and methods Materials and animals Iron (III) chloride (FeCl[3]) and 2-methylimidazole (2-MI) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Zinc nitrate hexahydrate [Zn (NO[3])[2]·6H[2]O] were purchased from Aladdin Biochemical Technology Co., Ltd. (Shanghai, China). Gossypol (Gp) was obtained from Jinan Daigang Biomaterial Co., Ltd. (Jinan, China). Thiazolyl blue tetrazolium bromide, Annexin V-FITC, JC-1 and propidium iodide (PI) were provided by the Beyotime Institute of Biotechnology (Shanghai, China). Cytochrome C, caspase-9, caspase-3, Bcl-2, Bax and GAPDH were all obtained from Abcam (Cambridge, UK). PKM2, LDHA, SDHA, HSP70 and HSP90 were provided by Cell Signaling Technology (Massachusetts, USA). Dithiothreitol, glycine and Tris-base were from Biosharp (Anhui, China). PMSF, TEMED, bromophenol blue and acrylamid were purchased from Amresco (WA, USA). DMEM, FBS, PBS buffer and trypsin–EDTA solution (0.25%) were bought from Gibco (Carlsbad, CA). All other chemical reagents were of analytical or HPLC grade. HepG2 cells were provided by Cell Resource Center, Peking Union Medical College (PCRC) (Beijing, China). Male BALB/c nude mice (6–8 weeks, 19–23 g) were purchased from Beijing Vital River Laboratory Animal Technology Co. Ltd (Beijing, China). All animal experiment procedures were approved by the Institutional Animal Care and Use Committee of the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, and followed the Guide for the Care and Use of Laboratory Animals. Preparation and characterization of ZIAMH nanocomposites Synthesis of ZIF-8@ArBu&ICG 40 mg Zn (NO[3])[2]·6H[2]O, 0.77 g 2-methylimidazole, 1 mg ArBu and 1 mg ICG were dissolved in 5 mL methanol, stirring at room temperature for 5 min. After centrifugation, desired precipitate was washed with water and dried for further use. Synthesis of ZIF-8@ArBu&ICG@MPN (ZIAM) The material ZIF-8@ArBu &ICG, Gp and FeCl[3]·6H[2]O were mixed in defined ratio (1:0.75:0.36 w/w). After being stirred overnight at room temperature, the mixture was dialyzed in deionized water, and the dialysis melium was interplaced by fresh deionized water every 2 h. Nanoparticles were collected by centrifugation and washed three times with deionized water. ZIF-8@MPN of control group was synthesized in similar way. Synthesis of ZIAMH The HA solution (1.0 mg/mL) was stirred with ZIAM at a ratio of 1:50 in deionized water for 2 h, and ZIAMH was obtained by centrifugation, resuspended in PBS, and kept at 4 °C for further experiments. Characterization of ZIAMH The morphology of the nanoparticles was observed by transmission electron microscope (HITACHI, Tokyo, Japan). The particle size, dispersion index (PDI) and Zeta potential of the prepared ZIAMH nanoparticles were measured by a Malvern NanoZS90 (UK). The phase composition and microstructure of the nanoparticles were further analyzed using Powder X-ray diffraction (PXRD), The element distribution was analyzed by Energy Dispersive Spectrometer (EDS). The surface area of the nanoparticles was measured by Brunauer − Emmett − Teller (BET) method. The loading content (LC) and encapsulation efficacy (EE) of the ArBu and ICG in the nanoparticles was measured by UV − vis spectra, and was determined according to the following equations. [MATH: LC=weightofdruginthenanoparticl esweightofdrugloadedmicelles×100% :MATH] [MATH: EE=weightofdruginthenanoparticl esweightoftheaddeddrug×100% :MATH] Performance investigation of ZIAMH nanomaterials In vitro drug release The in vitro ArBu and ICG release from nanoparticles was determined at 37 °C in PBS containing 0.5% Tween-80 (pH 4.0, pH 5.6 or pH 6.5). The quantities of released ArBu and ICG drug were determined by UV–vis spectrophotometer, respectively. Each sample was measured for three times. Detection of ·OH Methylene blue (MB) was introduced to detect the hydroxyl radical (·OH) generated by ZIAMH. Using methylene blue (MB) as an indicator, the generation of ·OH was detected by UV–Vis spectrophotometer. 100 μg and 200 μg of ZIAMH was mixed with MB (10 μg/mL) and H[2]O[2] (150 μM), respectively, to investigate the effect of sample size on the chemical kinetic catalytic ability. The photothermal properties of ZIAMH The temperature elevation curve and photothermal stability of ZIAMH were measured under 808 nm NIR laser irradiation in vitro. Firstly, the thermal stability of ZIAMH was established. The temperature variation curves of ZIAMH solution at different concentrations (125, 250, 500, 1000 μg/mL) were measured under the irradiation of 808 nm NIR with a power density of 1.5 W/cm^2 according to the previous study [[92]44]. FLIR Thermal CAM (E50) was used to record the real-time thermal image of the sample, and FLIR Examiner software was used to quantify and draw the temperature elevation curve. Secondly, the thermal stability of ZIAMH was investigated. 1 mL of ZIAMH solution was irradiated with 808 nm NIR laser for 600 s and cooled down to room temperature, followed by three cycles of laser irradiation. The temperature change of the solution was recorded in real time curve. Hemolysis Assay The hemolysis of nanoparticles was determined by co-culture with human red blood cells according to previously reported method. Red blood cells were isolated by centrifugation at 5000 rpm, then were rinsed with tris buffer and finally diluted to 5% blood cell stock solution. The concentration of the material was started from 1000 μg/mL, which was diluted using a double dilution method. The material was then incubated with an equal amount of red blood cell samples in a 37% CO[2] incubator for 1 h. The PBS group was used as negative control. 1% Triton X-100 solution was defined as positive control (which induced 100% hemolysis). After centrifugation, the supernatant was collected and its absorbance was measured at 540 nm. Triplicates were made for each test, the data were recorded as mean ± standard deviation (SD, n = 3). The hemolytic percentage was estimated by the following equation: [MATH: Hemolysis%=Am-< msub>An/Ap-< msub>An×100% :MATH] where A[m] is the absorbance of cells treated by ZIAMH, A[n] is the absorbance of the negative control, and A[p] is the absorbance of the positive control. In vitro cellular uptake HepG2 cells were inoculated in confocal laser scanning microscope (CLSM) plates at a density of 1 × 10^5 cell/ well for 24 h incubation. Free ICG (0.5 μg/mL), ZIF-8@ArBu&ICG@MPN (ZIAM, 0.625 mg/mL) and ZIAMH (0.625 mg/mL) were added to the cells before incubation for 0, 0.5, 1, 2 or 4 h. HepG2 cells were washed with PBS and fixed with 4% paraformaldehyde for 30 min. Pre-cooled TritonX-100 was added to destroy the cell membrane. After washing with PBS, HepG2 cells were stained with DAPI for 10 min. The fluorescence intensity was imaged and analyzed by laser confocal microscopy at an excitation wavelength of 750 ± 10 nm and an emission wavelength of 810 ± 20 nm. Cytotoxicity evaluation HepG2 were seeded in 96-well plates with 5 × 10^3 cell/ well for 24 h, and then different gradient concentrations of ZIAMH were added to investigate the cytotoxicity. Next, ZIF-8@MPN@HA (ZMH), ArBu, ArBu&ICG + NIR and ZIAMH + NIR were added and incubated for 24 h. The cell viability value was detected using MTT colorimetry and calculated as follows: [MATH: Cell viability%=OD(treatment group)/OD(control group)×100% :MATH] Flow cytometry analysis of apoptosis The cell apoptosis mechanism was explored by flow cytometry using Annexin V-FITC/PI Apoptosis Detection Kit (BD Biosciences). HepG2 cells were inoculated into 6-well plate for 24 h, follow by 24 h incubation with PBS、ZMH (0.625 mg/mL), ArBu (90 μM), ArBu (90 μM)&ICG (50 μg) and ZIAMH (0.625 mg/mL), during which ArBu&ICG group and ZIAMH group were irradiated by 1.5 w/ 5 min/ well of 808 nm laser post 4-h incubation. Collected cell pellets were washed with cool PBS and resuspended up to 1 × 10^6 cells/mL with binding buffer. 100 μL of the cell suspensions and 5 μL of AnnexinV-FITC were then gently mixed in falcon tube, followed by 30 min dark incubation at room temperature, with 5 μL of PI added in the middle of incubation. The suspension was then replenished with 400 μL of binding buffer and analyzed immediately. Live/Dead assay HepG2 cells were planted in 6-well plates at a density of 3 × 10^5 cells/ well, and then treated with PBS, ZMH (0.625 mg/mL), ArBu (90 μM), ArBu (90 μM)&ICG (5 0 μg), ZIAMH (0.625 mg/mL) for 24 h, during which ArBu&ICG group and ZIAMH group were irradiated with 1.5 W/ well 808 nm laser for 5 min after 4 h of treatment. The harvested cells were stained with 10 nM of calcein AM and PI dye for 10 min at 37℃. Cells were washed with PBS before analysis by confocal microscopy. Measurement of mitochondrial membrane potential HepG2 cells were treated with PBS, ZMH (0.625 mg/mL), ArBu (90 μM), ArBu&ICG + NIR (90 μM ArBu & 50 μg ICG), ZIAMH + NIR (0.625 mg/mL) for 24 h, during which ArBu&ICG group and ZIAMH group were irradiated with 1.5 W/ well 808 nm laser for 5 min after 4 h of treatment. The harvested cells were then incubated with 1 mL JC-1 working solution for 20 min. After washing, the cells were resuspended with 500 μL of JC-1 working solution for analysis by BD FACSCalibur flow cytometer. Western blot analysis Total protein was extracted from HepG2 cells or nude mouse tumors using RIPA lysis buffer (Beyotime, China) with 1% protease inhibitor PMSF (Amresco, China). The protein content was determined using a BCA protein assay kit (Beyotime, China). 30 μg of total protein were taken for SDS-PAGE electrophoresis, and wet transferred to polyvinylidene fluoride (PVDF) membrane (Merck, Germany), based on the protein quantification results. After blocking with 5% skimmed milk/TBST solution at room temperature for 1 h, the PVDF membrane was incubated with the indicated primary antibodies: caspase-9 (1:500), caspase-3 (1:1000), cytochrome C (1:1000), Bcl-2 (1:1000), Bax (1:1000), PKM2 (1:500), LDHA (1:1000), SDHA (1:500), HSP70 (1:1000) and HSP90 (1:1000), and was shaken overnight at 4 °C. After washing with TBST, the PVDF membrane was incubated with the corresponding secondary antibody (1:10,000) for 45 min at room temperature. The protein band was acquired by Tanon 5200 Chemiluminescence Imaging System (Shanghai, China) and GAPDH was used as internal control. Seahorse cell energy metabolic analysis The oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of HepG2 cells were detected by Seahorse XF-96 system (Agilent, Santa Clara, USA) according to the instructions of Seahorse XF Cell Mito Stress Test Kit and Glycolysis Stress Test Kit. The HepG2 cells were seeded into Seahorse XF cell culture plates and were treated with PBS, ZMH, ArBu, ArBu&ICG + NIR and ZIAMH. The two indicators OCR and ECAR were measured to evaluate the energy metabolism status and mitochondrial function of cells, and the experimental data was analyzed using wave software and report generator. Cellular ROS analysis Cellular reactive oxygen species (ROS) levels were determined using a 2,7-dichloro-fluorescein diacetate (DCFH-DA) assay kit (Beyotime, #S0033M) following the manufacturer's protocol. HepG2 cells were seeded into confocal small dish cultured for 24 h at 37 °C and incubated with ZMH, ArBu, ArBu&ICG + NIR and ZIAMH + NIR for 4 h, respectively. Subsequently, the cells were washed with PBS three times and incubated with 10 μM DCFH-DA for 30 min. Then, the cells were observed by CLSM. In vivo NIR fluorescence imaging of ZIAMH For NIR fluorescence imaging and biodistribution analysis, HepG2 tumor-bearing mice were intravenously injected with ICG or ZIAMH. The tumor-bearing mice were imaged using an animal imaging system (Xtreme, Bruke) at 0.5, 4, 8, 24, and 48 h post-injection. Mice were sacrificed 48 h after administration, and tumor tissues and main organs (including heart, liver, spleen, lung and kidney) were isolated for fluorescence imaging and semi-quantitative analysis. The excitation wavelength of ICG was 750 ± 10 nm and the emission wavelength was 810 ± 20 nm. In vivo photothermal imaging of ZIAMH To evaluate the in vivo photothermal effect of ICG-loaded ZIAMH nanoparticles, PBS and ZIAMH were intravenously injected to tumor-bearing mice, the thermal change of tumor irradiated at 808 nm NIR laser was monitored. Moreover, 24 h after intravenous injection was selected as the irradiation time and ICG accumulation reached its peak at this time. The mice were anesthetized with isoflurane, at predetermined time, and then 808 nm NIR laser (1.5 W/cm^2) was injected to the tumor site. The mice were thermally imaged with an infrared thermal imager (FLIR E50) every 0.5 min for a total of 5 min. Quantitative analysis on imaging results and thermal variation data was performed by FLIR tools. In vivo tumor inhibition evaluation To establish a xenograft tumor nude mouse model, 6 × 10^6 HepG2 cells/well were subcutaneously injected into male BALB/c nude mice based on a previously described study with some minor changes [[93]16]. After the tumor volume reached approximately 100 mm^3, HepG2 tumor-bearing mice were randomly divided into five groups (n = 3): PBS group, ZMH group, ArBu group, ArBu + ICG + NIR group, PD-L1 group, ZIAMH + NIR group and ZIAMH + PD-L1 + NIR group, afterward the mice were intravenously injected with the above drug formulations. For photothermal therapy, 24 h after injection, the tumor site was irradiated with 808 nm NIR laser (1.5 W/cm^2) for 5 min. The tumor volume and mouse body weight (bw) were measured every two days for a total of 10 days, and the tumor volume was calculated according to the following formula: V (mm^3) = length × (width)^2/2. The tumor inhibition ratio percentage = (bw control group -bw treatment group)/ bw control group × 100%. The tumor-bearing mice were sacrificed 21 days after intravenous administration, and the heart, liver, lung, spleen and kidney of the mice were obtained for hematoxylin and eosin (H&E) staining to evaluate the morphology of cancer cells. Histochemical staining For hematoxylin and eosin (H&E) staining, tumor tissues and main organs (heart, liver, spleen, lung and kidney) of HepG2 tumor-bearing mice were fixed in 4% formaldehyde solution and then placed in different concentrations of ethanol for dehydration. Subsequently, the tissues were cleared with xylene, immersed and embedded in molten paraffin. The wax blocks were cut into sections and dewaxed with xylene, then rehydrated and rinsed with water, and stained with hematoxylin and eosin. The coverslips were covered with neutral balsam mounting medium for sealing. The tumor and major tissue sections were observed and photographed with a microscope. For CD4 immunohistochemical staining, the tissue fixation and section steps are similar to H&E staining, expected that a CD4 monoclonal antibody was added to specifically identify immune cell subpopulations that express CD4 molecules. RNA-sequencing analysis The tumor tissues were ground into powder using liquid nitrogen and total RNA was extracted by Trizol (Thermo Scientific, USA). RNA integrity was assessed using the Agilent 2100 Bioanalyzer (Agilent Technologies, USA). The transcriptome library was constructed using a TruSeq Stranded mRNA LTSample Prep Kit (Illumina, USA), and RNA-sequencing analysis was conducted by Shanghai OE Biotech Co., Ltd (Shanghai, China). Fastq software was used to perform quality control analysis on the preprocessed data. The clean data were mapped to Homo sapiens (GRCh38.p13) by HISAT2. The Fragments Per Kilobase Million (FPKM) value of each gene was calculated by Cufflinks, and the sequence counts for per gene were acquired by HTSeq-count. Differentially expressed genes (DEGs) analysis was carried out using an R package, DESeq2. Data with P < 0.05 and log[2] |(fold change)|> 1 was considered as significant DEGs. Principal Component Analysis (PCA), heatmap analysis, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was conducted using Omicshare cloud tools ([94]https://www.omicshare.com/tools/). Targeted metabolomics of glucose metabolic pathways The tumor tissues in each group were homogenized with threefold volume of saline solution. 100 μL of the homogenate were added with 0.2% formic acid-acetonitrile, then were vortexed for 60 s and sonicated for 10 min in an ice-cold sonication bath. The mixture was centrifuge at 12,000 r/min for 15 min at 4 °C. The supernatant was taken for UPLC-MS/MS targeted metabolomic analysis. The UPLC-6500 + triple quadrupole MS/MS system (SCIEX, CA, USA) was applied for quantitative analysis of TCA cycle metabolites in the above tumor tissues. A Waters UPLC^® HSS PFP column (2.1 mm × 100 mm, 1.8 μm) was used for sample separation at 35 ℃. The mobile phase consisted of (A) water containing 0.05% formic acid and (B) acetonitrile containing 0.05% formic acid with a flow rate of 0.3 mL/min. The following gradient program of chromatographic condition was performed: 0–4 min, 2% B; 4–6 min, 2%–98% B; 6–10 min, 98% B; 10–10.1 min, 98%–2% B; 10.1–14 min, 2% B. The injection volume was 3 µL. The MS spectrum conditions were optimized as follows: ESI source negative ion mode; multiple reaction monitoring (MRM) detection mode; spray voltage, ± 4500 V; atomization temperature, 550 ℃; curtain gas, 35 psi; collision air pressure, 9 psi; atomization gas pressure (N[2]) and gas pressure (N[2]) were both 55 psi. The parameters of metabolites including Q1 Mass, Q3 Mass, collision energy (CE) and decluttering potential (DP) were listed in Table S1. Statistical analysis All data were analyzed by GraphPad Prism software (Version 9.0, SanDiego, USA) and expressed as means ± standard deviation (SD). One-way ANOVA and t-test statistical analysis were used for all experimental data. The significant differences were defined as *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001. Results and discussion Synthesis and characterization of ZIAMH In order to fabricate the nanocomposites, ArBu and ICG were firstly loaded into ZIF-8 nanocarriers, gossypol and Fe^3+ were then added to establish a metal polyphenol network on the surface of ZIF-8 nanoparticles. HA was encapsulated on the outer layer of the nanoparticles to form the final structure of ZIAMH. As observed by transmission electron microscopy (TEM) (Fig. [95]1a), the morphology of ZIF-8, ZIF-8@MPN and ZIAMH exhibited regular polyhedral structures, and the particle size distribution was between 150–190 nm. The elemental mapping of ZIAMH nanoparticles were analyzed using a TECNAI F20 transmission electron microscope equipped with an energy dispersive X-ray spectrometer (EDX). The results clearly showed that Zn, Fe, C, O and N elements were uniformly distributed in ZIAMH (Figure S1), which confirmed the metal polyphenol network on the surface of ZIF-8 nanoparticles. Dynamic light scattering (DLS) test showed that the sizes of three particles fluctuated by about 10 nm around 190 nm, indicating that the outer layer of ZIF-8 did not alter its basic morphology (Fig. [96]1b). The Zeta potential of ZIAM was 16.5 ± 1.28 mV, while the Zeta potential of ZIAMH shifted to − 21.9 ± 1.78 mV after HA coating (Table S2). This change from positive to negative charge emphasizes the influence of the HA layer on the physical characteristics of ZIAMH. The PDI values of three particles ​​were all less than 0.2. The above results demonstrated the assembled nanoparticles have uniform morphology and good size distribution. Fig 1. [97]Fig 1 [98]Open in a new tab Synthesis and characterization of ZIAMH. a TEM images of ZIF-8, ZIF-8@MPN and ZIAMH (scale bar: 100 nm). b DLS results of ZIF-8, ZIF-8@MPN and ZIAMH. c XPS pattern of ZIAMH. d PXRD patterns of ZIF-8@ICG&Arbu, ZIF-8@MPN and ZIAMH. e UV–vis spectra of MB, MB after reaction with ZIAMH in different concentrations. f The cumulative release of ArBu and ICG from ZIAMH at pH 6.5. g 10-min photothermal temperature curves of ZIAMH dispersed in water at various concentrations. h Photothermal conversion curves of ZIAMH under three cycles of irradiation The assay on surface elements through X-ray photoelectron spectroscopy (XPS) also confirmed the desired construction of the material (Fig. [99]1c). Derived from the analysis of the phase composition and microstructure of nanoparticles using PXRD, it was found that the signature peaks of all nanoparticles were almost identical to those simulated of ZIF-8, the crystal structure of the nanoparticles remained intact post subsequent modifications (Fig. [100]1d). The evaluation on material's specific surface area, pore volume, and pore structure was conducted by N[2] adsorption measurement (Figure. S2). According to the BET model, the specific surface area of ZIAMH nanoparticles was calculated to be 76.7554 m^2/g, the pore volume and pore diameter were 0.0394 cm^3/g and 2.604 nm, respectively. The sharp increase in N[2] absorption by ZIAMH nanoparticles under low relative pressure (< 0.01) indicates the existence of mesoporous structure. Additionally, methylene blue (MB) was used as an indicator to test whether Fe^2+ in the material can effectively generate ∙OH through Fenton reaction. As shown in Fig. [101]1e, with the increase amount of material sample, the absorbance of MB significantly decreased, indicating that the amount of material sample was proportional to the catalytic chemical kinetic energy. The UV spectrophotometer assay was applied to determine that the material could load with 5.7% ArBu and 7.2% ICG, and it was also used to investigate the release of nanoparticles loaded with the two drugs at different pH values (Fig. [102]1f and S3). It can be seen that the cumulative release drug amount could be accelerated at lower pH. It was hypothesized that the structures of MPNs and ZIF-8 within the nanomaterials can be decomposed under acidic environments, which leaded to the breakage of the material structure and release of the incorporated drugs. TEM images confirmed that ZIAMH composites gradually decomposed in acidic PBS (pH 5.6 and 6.5), producing small-sized ZIAMH fragments, while they retained the regular morphology in neutral PBS (pH 7.4) within 8 h, which is consistent with the literature reports (Figure. S4). ICG can convert light energy into thermal energy under NIR laser irradiation due to its photothermal characteristic [[103]45]. In this study, different concentrations (0, 125, 250, 500, and 1000 μg/mL) of ZIAMH nanoparticles were exposed to 808 nm NIR light (1.5 W/cm^2). As shown in Fig. [104]1g, the temperature rise amplitude is positively correlated with the concentration and illumination time of the nanoparticles. The temperature of the aqueous solution rapidly increased from 27.1 ℃ to 52.5 ℃ after 600 s illumination even at a nanomaterial concentration as low as 125 μg/mL, suggesting that the ZIAMH nanomaterials had good photothermal conversion effects. Moreover, the thermal performance test also showed the material exhibited sustained photothermal ability at least post three cycles (Fig. [105]1h). To evaluate the biocompatibility of ZIAMH nanomaterials in vitro, hemolysis experiments were conducted using red blood cells. The results suggested that the hemolysis rate of the nanoparticles at all concentrations (15.6, 31.25, 125, 250, and 500 μg/mL) was less than 5%, which demonstrated that ZIAMH nanoparticles have wonderful hemo-compatibility (Figure. S5). In vitro cellular uptake and anti-tumor effects of ZIAMH nanoparticles HA is a tumor-targeting ligand and can actively target liver tumors by binding to the CD44 receptor [[106]46–[107]48]. HepG2 cell was applied as the tumor model to explore the cancer targeting performance of HA coated nanoparticles. The in vitro HepG2 cells uptake of ZIAMH nanoparticles was observed using confocal laser scanning microscopy. As shown in Fig. [108]2a, the blue fluorescence represented DAPI stained nuclei, while the red ones represented ICG molecules internalized into the cytoplasm of the nanoparticles. There was barely red signal in the cytoplasm after co-incubation with free ICG for 1 h, indicating the lower cellular uptake of free ICG by the tumor cells. ICG has strong hydrophobicity and poor dispersibility in water, making it hardly to permeate through the cell membrane through endocytosis. As a comparison, strong red fluorescence appeared in the cytoplasm treated with ZIAM nanoparticles, proving that the size effect and good water solubility of nanoparticles promoted the cellular internalization of ICG (Figure. S6). And due to the addition of HA, stronger ICG signal was observed in the cytoplasm of the ZIAMH group (Fig. [109]2b and c), suggesting that the presence of HA could help to enhance the targeting ability of the nanoparticles and further promote the internalization of ICG. Fig 2. [110]Fig 2 [111]Open in a new tab The cellular uptake assay and in vitro anticancer activity of various drug formulations. a The CLSM images of HepG2 cells treated with ICG. b The CLSM images of HepG2 cells treated with ZIAMH. (Blue channel: DAPI, Red channel: ICG, scale bar: 50 µm). c The mean fluorescence intensity of ICG in HepG2 cells treated with ICG, ZIAM or ZIAMH for 0–4 h (n = 3). d The viability of the HepG2 cells incubated with ZMH, ArBu, ArBu&ICG + NIR or ZIAMH + NIR for 24 h (n = 6). e, f Apoptosis analysis of HepG2 cells after different treatments (n = 3). All quantitative data are presented as mean ± SD. ***P < 0.001, ****P < 0.0001 The cytotoxicity of the carrier material was investigated using MTT assay. The cellular viability of HepG2 cells were measured after co-incubation with different concentrations of ZIAMH for 24 h. The exposure of ZIAMH to 808 nm NIR laser irradiation induced elevated temperature due to the photothermal effect of the loaded ICG, leading to the apoptosis of cancer cells. The survival rate of HepG2 cells gradually decreased with the increase of the concentration of ICG. These results demonstrated that ZIAMH nanoparticles can significantly inhibit the growth of cancer cells under NIR light irradiation. In addition, the cytotoxicity of HepG2 cells in different treatments was also evaluated (Fig. [112]2d and S7). The mechanism study of cell apoptosis Furthermore, the cell apoptosis of each treatment group was studied by Annexin V-FITC/PI assay (Fig. [113]2e and f). The cell apoptosis rate of ZIAMH nanoparticles increased to 26.7% after 24 h incubation, as compared with the 14.1% apoptosis rate of free ArBu. This result demonstrated that the ZIAMH nanoparticles could be effectively targeted for delivery to tumor cells due to the presence of HA molecules, which was consistent with previous studies on the tumor targeting ability of HA [[114]49, [115]50]. The apoptosis of HepG2 cells was also investigated after treatment with various therapeutic agents combined with NIR light irradiation, accompanied by calcein AM (2 μM) and PI solution (4 μM) staining. As shown in Fig. [116]3b, the absence of red signal in cells of PBS group demonstrated that the single laser irradiation treatment could not damage the cancer cells. As for the blank material group ZMH, the red signal in the cells after irradiation was shown as scattered and weak, indicating the partial apoptosis of cancer cells. The metal-polyphenol network in the blank material exhibited chemodynamic effect inducing cellular apoptosis, however, the therapeutic efficacy of chemodynamic catalysis was less remarkable than that of other groups. Interestingly, compared with the ICG group and free ArBu group, the ZIAMH group showed massive death of cancer cell, as reflected by the strong red signal. The results demonstrated that ZIAMH nanoparticles exhibited optimized in vitro anticancer effect under NIR light irradiation. Fig 3. [117]Fig 3 [118]Open in a new tab In vitro anticancer activity and mechanism of cell apoptosis. a Mitochondrial membrane protein JC-1 red/green florescence ratio (n = 3). b Live (green) and dead (red) cells staining after treatment with PBS, ZMH, ArBu, ArBu&ICG + NIR or ZIAMH + NIR. scale bar: 50 µm. c Western blot imaging of proteins caspase-9, caspase-3, cytochrome C, Bcl-2, Bax and GAPDH after incubation with PBS, ArBu and ZIAMH. d The relative expression level of caspase-9, caspase-3, cytochrome C, Bcl-2 and Bax by western blot analysis (n = 3). All quantitative data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 The cellular apoptosis is closely related to mitochondrial dysfunction [[119]51]. In order to investigate the mechanism of the tumor cell apoptosis, JC-1 staining was used to observe the change of mitochondrial membrane potential in each dosing group. As shown in Fig. [120]3a, ZIAMH nanoparticles exhibited significant anti-tumor effect, which could be seen by the highest green-to-red ratio after 24 h incubation, hinting the extensive mitochondrial damage of the cells. To elucidate the levels of mitochondrial apoptosis proteins such as anti-apoptotic Bcl-2 and pro-apoptotic Bax in HepG2 cells induced by ZIAMH nanoparticles, western blot analysis was performed on HepG2 cells treated with ArBu and ZIAMH nanoparticles. As exhibited in Fig. [121]3c and d, compared with the western blot results of free ArBu, ZIAMH nanomaterials significantly reduced the level of Bcl-2 in HepG2 cells, and promoted expressions of apoptosis-related proteins including Bax/Bcl-2 ratio, cytochrome C, caspase 9 and caspase 3. Therefore, it can be concluded that there is indeed a close relationship between tumor cell death and mitochondrial apoptosis pathways. In vitro mechanism study of tumor glucose metabolism by ZIAMH Most of the energy required for cell life activities originates from mitochondria, thus, the study of mitochondrial function is particularly important to understand the energy metabolism of cells. The seahorse cell energy metabolic analysis was performed in this study to explore the regulation effects of ZIAMH nanomaterials on tumor glycolytic capacity and mitochondrial OXPHOS pathway. Besides, ECAR and OCR were used to reflect glycolysis and mitochondrial respiratory functions respectively, thereby to quickly assess the energy metabolism status of intracellular mitochondria. As exhibited in Fig. [122]4a, the ECAR of each treatment group was significantly reduced compared with the control group, indicating that the glycolysis process was inhibited. As compared with the free drug ArBu, the supplement of ICG and NIR irradiation further suppressed the glycolysis process. In addition, it can be concluded from the OCR result that the level of mitochondrial OXPHOS decreased significantly in HepG2 cells after treatment with ZIAMH nanomaterials (Fig. [123]4b). To sum up, the regulation of glucose metabolism by ZIAMH nanocomposites combining with NIR irradiation was demonstrated in the dual inhibition pathway of glycolysis and mitochondrial OXPHOS. It was speculated that ArBu in the ZIAMH nanomaterial may inhibit the mitochondrial OXPHOS through the mitochondrial apoptosis pathway, thereby exerting anti-tumor effects. Fig 4. [124]Fig 4 [125]Open in a new tab In vitro antitumor mechanism based on glucose metabolism. a The ECAR (glycolysis indicator) of HepG2 cells by seahorse cell energy metabolic analysis. b The OCR (OXPHOS indicator) of HepG2 cells by seahorse cell energy metabolic analysis. c Western blot imaging of proteins PKM2, LDHA, SDHA, HSP70, HSP90 and GAPDH after incubation with PBS, ZMH, ArBu, ArBu&ICG + NIR or ZIAMH + NIR. d The relative expression level of PKM2, LDHA, SDHA, HSP70 and HSP90 by western blot analysis (n = 3). e The fluorescence images of intracellular ROS generation in HepG2 cells treated with PBS, ZMH, ArBu, ArBu&ICG + NIR or ZIAMH + NIR using DCFH-DA probe (Blue channel: Hoechst, Green channel: DCFH-DA, scale bar: 50 µm) and the quantitative analysis of intracellular ROS generation (n = 3). All quantitative data are presented as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 Furthermore, the expression of related proteins in the glucose metabolism pathway was detected to further investigate the regulatory effect of ZIAMH on tumor glucose metabolism through western blot assay (Fig. [126]4c and d). Compared with the control group, the protein expression of each treatment group was varied to some extent, especially in the ZIAMH + NIR group, where the proteins related to glucose metabolism were all decreased. Among them, PKM2 and LDHA are the main proteins related to the glycolysis pathway, which have the functions of generating pyruvate and converting pyruvate into lactate, respectively. The reduced expression of proteins PKM2 and LDHA indicated that the glycolytic pathway was inhibited, and there is no doubt that the content of lactate, the final product of glycolysis, will be reduced. Notably, the expression of SDHA, a marker of mitochondrial regulation, was also reduced. Decreased SDHA activity suggested that mitochondrial OXPHOS was also affected, as it regulates the cellular metabolic balance between OXPHOS and glycolytic metabolism. Heat shock proteins (HSP70 and HSP90) has been shown to control metabolic reprogramming of tumor cells [[127]52]. In terms of tumor metabolism, overexpression of Hsp70 enhances the glycolytic activity, leading to a shift in energy metabolism, while HSP90 is involved in metabolic regulation through interaction with SDHA and PKM2 [[128]53]. Currently, studies have shown that mitochondria are an important source of cellular ROS, and the production of ROS can break the mitochondrial electron transport chain, thereby inhibiting mitochondrial OXPHOS [[129]54, [130]55]. Based on this, intracellular ROS was detected by the DCFH-DA probe to assess its impact on tumor mitochondrial OXPHOS. As exhibited in Fig. [131]4e, compared with the PBS or free ArBu groups, the level of ROS in cell mitochondria increased significantly after treatment with ZIAMH. From this perspective, ZIAMH nanoparticles indirectly inhibited OXPHOS in tumor cells by generating a large amount of ROS, which is consistent with the OCR results by seahorse cell energy metabolic analysis. Biodistribution and in vivo antitumor efficacy of ZIAMH In vivo NIR fluorescence imaging of ZIAMH To investigate in vivo targeting ability of ZIAMH nanoparticles, PBS, free ICG, and ZIAMH were intravenously injected into the respective groups when HepG2 tumor size reached around 100 mm^3, followed by NIR fluorescence imaging on living mice at predefined time points. As shown in Fig. [132]5a, the fluorescence signal in tumor tissue of free ICG group gradually increased in the first 8 h post-injection, but decreased rapidly afterward. In contrast, the signal of that in the ZIAMH group continued to increase up t a peak at 24 h after injection, and then decreased slowly, which suggested that 24 h after injection is the best time for subsequent NIR light therapy. Fig 5. [133]Fig 5 [134]Open in a new tab The biodistribution and in vivo antitumor efficacy of ZIAMH nanomedicine. A: Control (PBS); B: ZMH; C: ArBu, D: ArBu&ICG + NIR; E: PD-L1; F: ZIAMH + NIR; G: ZIAMH + PD-L1 + NIR. a The in vivo distributions of ICG visualized by using an IVIS Spectrum system after intravenous administration. b The NIR thermographic images of tumor-bearing mice after receiving exposure to an 808 nm laser at a power density of 1.5 W/cm^2 in different groups, taken at 5 min. c Schematic illustration for the therapeutic model. d Representative images of the excised tumors after different treatments. e Tumor volume after different treatments at the 21th day. (n = 6, ***P < 0.001). f The body weight of mice among the different treatment groups presented no differences. g CD4 staining images of the dissected tumor tissues after sacrifice. Scale bar: 100 μm The encapsulation of ICG in ZIAMH also facilitated drug concentration in tumor site due to the active tumor-targeting ability of HA. Subsequently, each group of mice was sacrificed 48 h post injection to collect main organs (heart, liver, spleen, lungs, kidneys) and tumors. The biodistribution and concentration of ZIAMH in collected samples was detected using in vivo imaging technology. It has been shown that the ZIAMH group demonstrated stronger fluorescence signal in tumors and liver of the mice. Moreover, the ZIAMH nanoparticles may not induce cardiotoxicity because no significant fluorescence was observed. In vivo photothermal imaging of ZIAMH To evaluate the photothermal effect in vivo, ZIAMH nanoparticles were injected into tumor-bearing mice through tail vein injection, with the PBS group as a control. Them the tumor region was exposed to 808 nm NIR laser for 5 min after 24 h of injection. As manifested in Fig. [135]5b, the temperature of the tumor areas in the PBS group scarcely rise within 5 min, while that of the ZIAMH nanoparticles group increased significantly. Moreover, the maximum temperatures reached by the tumor sites in these two groups were 30.5 ℃ and 52.5 ℃, respectively. Importantly, the accumulation of ZIAMH nanoparticles at the tumor site was improved due to the active targeting effect of HA, thereby promoting photothermal conversion within tumor, which was consistent with the results of NIR imaging. These results demonstrated that ZIAMH nanoparticles can not only efficiently accumulate in tumor area, but can also generate sufficient heat to neutralize cancer cells under irradiation, which is a promising photothermal agent. In vivo anti-tumor evaluation Recently, great progress has been made in tumor immunotherapy for regulation of the patient's own immune cells to enhance the treatment outcome of tumor cells [[136]56, [137]57]. Programmed cell death 1-ligand 1 (PD-L1) is currently the most widely used immune checkpoint pathway in clinical practice, and the commercialized PD-L1 antibodies have shown tremendous success, especially in the treatment of advanced cancer [[138]58]. Thus, PD-L1 treatment group and ZIAMH&PD-L1 treatment group were added to evaluate the in vivo antitumor effect. The anti-tumor activity of ZIAMH with laser irradiation was studied by establishing mouse HepG2 xenograft tumor model. PBS, ZMH, Arbu, Arbu&ICG, ZIAMH, PD-L1 and ZIAMH&PD-L1 was intravenously injected into mice, respectively (Fig. [139]5c). The PTT treatment was conducted 24 h post administration, with tumor site being irradiated with 808 nm NIR laser (1.5 W/cm^2) for 5 min. The body weight and tumor volume of mice were recorded every other day and results were displayed in Fig. [140]5d–f. The tumor growth trend of each group is basically consistent regardless of irradiation and no significant reduction of weight was observed. Under NIR irradiation, the ZIAMH group containing ICG showed stronger antitumor effects on tumors than the free Arbu group. It should be noted that the tumor volume in the ZIAMH group was smaller than that of the PD-L1 group, indicating that the ZIAMH nanoparticles had a better antitumor effect. In addition, the combination of ZIAMH and PD-L1 therapy exhibited the most potent antitumor efficacy among all of the treatment groups. These results suggested that the combined use of ZIAMH and PD-L1 inhibitors may offer a promising therapeutic approach for cancer treatment. The tumor tissues in each group were further collected for histology analysis. From immunofluorescence results in Fig. [141]5g, we found that that CD4 cell was particularly high in ZIAMH added with PD-L1. Interestingly, CD4 cell in ZIAMH nanomedicines was higher than that in PD-L1, indicating that ZIAMH may be better than PD-L1 in immunotherapy of tumors, because CD4 cells play an important role in the immune response process. It can also be observed from the H&E staining results that the tumor pathological sections in the treatment group were severely damaged compared to the control group, especially in ZIAMH nanomaterial (Figure. S8a). Considering that ZIAMH with synergistic therapy may cause damage to the normal organs when exerting its anti-cancer effect, the major organs (heart, liver, spleen, lung, kidney) of the mice was extracted in the ZIAMH + NIR group for histology analysis. It can be seen that the health status of these organs in the ZIAMH + NIR group was basically the same as that in the PBS group (Figure. S8b), indicating that the combination of PPT, CDT and chemotherapy did not cause significant damage to the normal organs of the mice. In vivo mechanism study of tumor glucose metabolism by ZIAMH Subsequently, transcriptomics analysis was performed and the results indicated that the PBS group and ZMH, ArBu, ArBu&ICG + NIR, PD-L1, ZIAMH + NIR, ZIAMH + PD-L1 + NIR groups clustered into different categories in the PCA plot (Figure. S9), and a total of 686 common DEGs were obtained among these groups compared with the PBS group (Figure. S10). To better understand the in vivo mechanisms of tumor metabolic reprogramming by ZIAMH nanomedicines, we further investigate the transcriptomics data. As shown in Fig. [142]6a, PCA plot revealed that the ZIAMH + NIR group was significantly different from the PBS group. Then, a volcano diagram was plotted to provide a basic description of the DEGs between the ZIAMH + NIR group and the PBS group, and |log[2](fold change)|> 1 and Q-value < 0.05 were adopted as the filtering criteria for DEGs screening (Fig. [143]6b), 1470 genes were downregulated and 1226 genes were upregulated. Further heatmap analysis were conducted to uncover DEGs associated with glucose metabolism (Fig. [144]6c). After ZIAMH + NIR treatment, the expression of key enzymes in glycolysis, OXPHOS or the TCA cycle, such as genes HK2, SDHA, LDHB, PFKM, PDK1, were significantly downregulated, which suggested that ZIAMH + NIR treatment had a considerable influence on glucose metabolism pathway and played a key role in inhibiting tumor growth. Among them, gene HK2 plays an important role in allowing glucose to enter the glycolysis and OXPHOS pathway [[145]59]. SDHA is an enzyme involved in the TCA cycle and energy metabolism, which not only plays a role in mitochondrial energy production, but also regulates the cellular metabolic balance between the TCA cycle and glycolysis [[146]60]. To gain further insight into the potential targeting pathways of ZIAMH + NIR, GO and KEGG enrichment analysis were carried out. As can be seen in Fig. [147]6d, it was observed that the negative regulation of carboxylic acid metabolic process, cellular metabolic process and other metabolic process were significantly enriched after ZIAMH + NIR treatment. The KEGG enrichment analysis results indicated that ZIAMH nanomedicines could downregulated glycolysis and carbon metabolism, and achieve antitumor effects by regulating carbohydrate metabolism and energy metabolism (Fig. [148]6e and S11). Additionally, protein–protein interaction (PPI) network analysis was performed using the genes associated with glucose metabolism pathways that were significantly altered after the ZIAMH + NIR treatment (Fig. [149]6f). Key proteins in the PPT network, including ACLY, LDHB, ENO2, ADH6 and PFKM, were involved in a variety of biological processes, for example, glycolysis, OXPHOS and tumor growth. Fig 6. [150]Fig 6 [151]Open in a new tab The in vivo biological mechanism of ZIAMH nanomedicines by RNA-sequencing analysis. a The PCA plot of PBS (A) and ZIAMH + NIR (B) groups. b The volcano map of PBS and ZIAMH + NIR groups. c The heatmap analysis of DEGs associated with glucose metabolism in PBS (A) and ZIAMH + NIR (B) groups. d GO enriched in PBS and ZIAMH + NIR groups. e KEGG analysis of genes and genomes enriched in PBS and ZIAMH + NIR groups (red box represent ZIAMH relative pathways). f PPI network associated with glucose metabolism pathways Glycolysis is a common pathway for glucose degradation in all living organisms, a process that produces pyruvate, followed by decarboxylating to generate acetyl CoA, which is completely degraded to carbon dioxide and water through the TCA cycle, and releases large amounts of energy. Glycolysis is closely linked with the TCA cycle, and together they constitute an important aerobic oxidation process of sugar. Thus, we further conducted targeted metabolomic analysis related with glycolysis and the TCA cycle pathway to explore the underlying sugar metabolism mechanism of intelligent ZIAMH nanomaterials in the TME (Fig. [152]7a). To visualize these products, the parameters of 9 metabolites were optimized using UPLC-triple quadrupole MS/MS (Table S1). As shown in Fig. [153]7b, compared with the PBS group, the content of glycolysis metabolites pyruvate and lactate were both decreased significantly. Furthermore, the levels of the TCA cycle intermediate metabolites citrate, cis-aconitate, succinate, fumarate and malate were all significantly reduced, indicating that ZIAMH treatment weakened the activity of TCA cycle and may have a tumor suppression effect. Fig 7. [154]Fig 7 [155]Open in a new tab Targeted metabolomic analysis of tumor tissues from HepG2 xenograft mice. a The diagram of the TCA cycle. b The concentration of key metabolites involved in glycolysis and the TCA cycle. The statistical significance was analyzed by one-way ANOVA and data are presented as means ± SD (n = 6). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 Conclusion In summary, we successfully constructed an MOF-derived intelligent ZIAMH nanoplatform that might promote energy deprivation by synchronously interfering with glycolysis, OXPHOS and TCA cycle, thereby achieving antitumor bioenergetic therapy integrated with PTT/CDT/CT therapy. Encapsulated with ZIF-8, the ZIAMH nanomaterials have considerably better antitumor efficacy than free ArBu, as well as good biosafety and solubility. Furthermore, the ZIAMH nanoparticles loaded with HA can efficiently be delivered to the tumor area by targeting the CD44 receptor, followed by the generation of ·OH triggered via the Fenton reaction and exhibiting enhanced CDT effects. Owing to the photothermal properties of ICG, such ZIAMH nanoparticles can effectively killed tumor cells under NIR laser irradiation without obvious side effects. In addition, in vitro evaluation demonstrated that the ZIAMH nanomaterials promoted cellular mitochondrial apoptosis through facilitating the expression of apoptosis-related proteins, such as Bax, Bax/Bcl-2, cytochrome C, caspase 9 and caspase 3. Importantly, systematic in vivo and in vitro mechanism studies suggested that ZIAMH nanomedicines can simultaneously inhibited glycolysis and OXPHOS in tumor cells by suppressing the expression of HK2, PKM2, LDHA, SDHA and PDK1. Moreover, they can further impair mitochondria function and decrease the level of ATP by depriving the TCA cycle of energetic substances and metabolites. Therefore, this work provides a new strategy for tumor treatment through precise energy deprivation and targeted therapy, integrating multiple pathways of bioenergetic therapy for metabolic interference and PTT/CDT/CT therapy. Supplementary Information [156]Supplementary material 1^ (6.8MB, pdf) Acknowledgements