Abstract Background Breast cancer remains the most common malignancy among women worldwide. While traditional computed tomography (CT) scans and image-guided radiotherapy are widely used for breast cancer diagnosis and treatment, their efficacy is often limited. Methods and results In this study, we successfully synthesized human serum albumin (HSA)-based KBiF[4] nanoclusters through a simple one-pot biomimetic mineralization strategy. Compared to the clinical contrast agent iohexol, KBiF[4]@HSA significantly enhances dual-energy CT (DECT) imaging contrast at high keV levels, offering improved diagnostic accuracy for breast cancer. Furthermore, KBiF[4]@HSA exhibits a remarkable ability to scavenge elevated glutathione (GSH) levels and promote reactive oxygen species (ROS) generation. When combined with radiotherapy, KBiF[4]@HSA substantially increases X-ray dose deposition at tumor sites, leading to enhanced DNA damage and suppression of breast cancer progression. Importantly, KBiF[4]@HSA demonstrates excellent biocompatibility in vivo, with no significant tissue damage or inflammation observed. Conclusions This study presents a novel approach for the development of biocompatible DECT contrast agents and radiosensitizers, offering a promising strategy to enhance breast cancer diagnosis and treatment. However, the efficacy of this approach needs to be further validated across diverse breast cancer subtypes to ensure its broad applicability, which emphasizes the necessity for continued research to fully translate this innovative technology into clinical practice. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03530-8. Keywords: Bismuth, Dual-energy CT, Contrast agents, Breast cancer, Radiotherapy Graphical Abstract [42]graphic file with name 12951_2025_3530_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03530-8. Introduction Breast cancer is becoming the most frequent malignancy among women worldwide, but it is curable with early-stage, nonmetastatic diagnosis [[43]1]. With the development of imaging diagnosis technology, earlier and smaller breast cancers can be diagnosed early through advanced imaging tools, and the five-year survival rate of breast cancer has improved in the past decades [[44]2, [45]3]. Radiotherapy serves as a noninvasive approach for eradicating breast cancer and significantly diminishing tumor burden. Despite its high efficacy in the majority of breast cancers, the nonspecific nature of irradiation can lead to off-target toxicity affecting adjacent tissues. To address this issue, image-guided radiotherapy has been developed, which integrates cancer imaging with radiotherapy to provide crucial functional information for radiotherapy planning. Dual-energy computed tomography (DECT), as a groundbreaking CT imaging technique, facilitates material differentiation and improved diagnostic capabilities compared to conventional single-energy CT. Furthermore, DECT provides distinctive qualitative and quantitative information about tissue composition, which is particularly beneficial in image-guided radiotherapy [[46]4–[47]6]. However, iodine-based CT contrast agents (CAs), routinely used clinically, are inadequate for DECT imaging due to their low K-edge energy (33.2 keV) [[48]7, [49]8]. Consequently, there is an urgent requirement to develop high-performance CAs to fulfill the demands of DECT imaging in breast cancer diagnosis and image-guided radiotherapy. Radiosensitizers have been developed to amplify the effects of radiation within high-grade tumors, such as triple-negative breast cancer (TNBC) [[50]9]. However, the therapeutic efficacies of radiotherapy are often impaired by the radioresistance and antioxidant defense system relevant to elevated glutathione (GSH) within tumors [[51]10]. Recently, various strategies have been developed to improve the effectiveness of these sensitizers. For instance, novel synthetic compounds, such as phenethyl isothiocyanate [[52]11] and epigallocatechin gallate [[53]12], have been identified to specifically deplete GSH and overcome radioresistance in cancer cells. In addition, metallic nanoparticle-based radiosensitizers, such as gold nanoparticles, have gained attention due to their ability to increase the dose of radiation absorbed by tumor tissues, thereby improving the overall therapeutic index of radiotherapy [[54]13]. However, these radiosensitizers may increase the accumulation of heavy metals in the normal tissues and induce potential bio-toxicity [[55]14]. More recently, tumor-specific radiosensitizers have been developed to amplify the effects of radiation within cancer cells, which helps mitigate the risks associated with high radiation doses by improving the precision of tumor targeting and minimizing exposure to adjacent organs at risk. Bismuth (Bi), as an element with a high atomic number (Z = 83) and a high K-edge energy (K-edge = 90.5 keV), exhibits X-ray attenuation capacity that possesses outstanding radiosensitizing and constant DECT imaging capabilities at various X-ray energies [[56]15, [57]16]. For instance, the CT contrast of Bi is superior to iodine at higher energy range (80–150 keV), making it more suitable for DECT imaging. Over the past decades, the rapid advancement of nanotechnology has facilitated the development of various Bi-based CAs, including Bi [[58]17–[59]20], Bi[2]S[3] [[60]21–[61]23], Bi[2]Se[3] [[62]24–[63]27], Bi[2]O[3] [[64]28–[65]31], BiF[3] [[66]32–[67]34], BiOI [[68]35–[69]37] Bi-DTPA [[70]38–[71]41], Bi[2]WO[6] [[72]42–[73]44] and other Bi-based nanomaterials [[74]45–[75]47]. However, the synthesis of Bi-based CAs presents significant challenges, such as elevated temperatures, an oxygen-free atmosphere, and the use of toxic organic solvents [[76]48, [77]49]. Furthermore, post-synthetic surface modifications are generally required prior to biomedical applications, potentially elevating systemic toxicity risks. Additionally, these Bi-based CAs demonstrate limitations related to effective degradation in biological environments, which is essential for minimizing potential toxicity and issues of long-term retention. Looking toward clinical translation, focus should be put on biocompatible and biodegradable Bi-based CAs that are compatible with DECT imaging and radiosensitization. Human serum albumin (HSA), the most abundant protein in human plasma, demonstrates reversible binding capabilities with both organic molecules and metal ions [[78]50–[79]52]. Due to their superior biocompatibility and non-immunogenicity, HSA-based drug formulations, such as Abraxane, have been clinically employed in the treatment of breast cancer [[80]53–[81]56]. Recently, HSA-mediated biomimetic mineralization of inorganic nanoparticles has garnered significant interest owing to milder reaction conditions and high reproducibility [[82]57–[83]60]. Scheme 1. [84]Scheme 1 [85]Open in a new tab Scheme of the synthesis and degradation of HSA-based KBiF[4] nanoclusters for DECT imaging and GSH-scavenging radiotherapy for breast cancer In this study, we present biocompatible KBiF[4] nanoclusters using the HSA-mediated bioinspired mineralization strategy. Compared to iohexol, the synthesized KBiF[4]@HSA demonstrates a significant enhancement in CT value and DECT imaging contrast at 180 keV, thereby improving the diagnostic accuracy for breast cancer. In addition, the precise tumor localization achieved through DECT imaging with these nanoclusters could enhance image-guided radiotherapy, thereby minimizing off-target toxicity in adjacent tissues. Moreover, these nanoclusters could be rapidly degraded by intracellular GSH in tumor cells, which could efficiently consume radioresistance-related GSH and sensitize tumor cells to radiotherapy. Furthermore, the KBiF[4]@HSA could deposit more X-ray energy, which significantly induces DNA damage and reactive oxygen species (ROS) generation under radiation. These nanoclusters demonstrated remarkable radiosensitizing properties, effectively suppressing tumor growth at reduced radiation doses. Importantly, the biodegradable KBiF[4]@HSA exhibited no detectable tissue damage or inflammation, underscoring their superior biocompatibility and minimal in vivo toxicity. Therefore, this study investigates biocompatible and biodegradable Bi-based CAs for DECT imaging and radiosensitizing via an HSA-mediated bioinspired mineralization strategy, which shows considerable potential for clinical translation. Materials and methods Materials Human serum albumin (HSA), Bismuth nitrate (Bi[NO[3]][3]·5H[2]O), Ethylene glycol (EG, 99%, Shanghai Chemical Reagent, China), and Potassium fluoride (KF·2H[2]O) were purchased from Aladdin Reagent Co., Ltd. (Shanghai, China). Iohexol was obtained from Beijing BEILU Pharmaceutical Co., Ltd, China (Beijing, China). The deionized distilled water (ddH[2]O) was purified by a Millipore filtration system. Synthesis of KBiF[4]@HSA KBiF[4]@HSA was synthesized by the one-pot method. 400 mg of HSA was dissolved in 5 mL of deionized water under a 37 °C water bath heating, and 5 mL of EG was added. After stirring for 1 min, 5 mL of Bi[NO[3]][3]·5H[2]O (242 mg) dissolved in EG was added, and 12.5 mL of KF·2H[2]O (1.13 g) dissolved in EG was added with stirring for 30 min. The mixture was stirred for 15 min, and the light-yellow solution was observed to gradually change to white. It was precipitated by adding 10 mL of deionized water and separated by centrifugation (4000 rpm, 10 min). Subsequently, the KBiF[4]@HSA was washed twice with deionized water and resuspended in deionized water to obtain KBiF[4]@HSA. Characterization The morphology of the prepared materials was examined using transmission electron microscopy (TEM, FEI Tecnai F20) and scanning electron microscopy (SEM, Zeiss Gemini 300). Elemental distribution was analyzed through elemental mapping. X-ray photoelectron spectroscopy (XPS, Thermo Kalpha) was performed with a Thermo Kalpha instrument. X-ray diffraction profiles were obtained using an X’Pert PRO MPD system. Fourier transform infrared spectroscopy (FT-IR) spectra were recorded with a Nicolet iS 10. Absorption spectra were acquired using a Cary 100 UV − visible spectrophotometer (Agilent Technologies) at room temperature. Dynamic light scattering (Nano ZS, Malvern) was used to record the hydrodynamic size distribution. GSH consumption KBiF[4]@HSA samples at the same concentrations were added to solutions containing 0 mM, 1 mM, 2 mM, 5 mM, 10 mM, and 15 mM of GSH. After 2 h of reaction, the KBiF[4]@HSA samples treated with varying GSH concentrations were placed in clear glass vials for comparative photography and subjected to UV-Vis spectroscopy after 24 h. Additionally, the KBiF[4]@HSA samples were analyzed using DLS and TEM at 0 min, 5 min, 15 min, 30 min, and 60 min. To detect intracellular GSH, 4T1 cells were incubated with KBiF[4]@HSA for a period of 24 h using the cellular glutathione peroxidase assay kit with DTNB. Cell lysates were then added to the test solution. The absorbance at 412 nm was measured with an enzyme marker to determine the concentration of GSH. Cell culture Rat mammary carcinoma (SHZ-88) cells, normal rat kidney (NRK) cells, and 4T1 cells were provided by the Chinese Academy of Sciences Cell Bank (Shanghai, China) and cultured in RPMI-1640 medium (Gibco, Thermo Fisher, USA) with 10% fetal bovine serum (Gibco, Thermo Fisher, USA) and 1% penicillin-streptomycin (Gibco, Thermo Fisher, USA). The fresh medium was changed every two days. Cells were washed with phosphate buffer solution (Biosharp, China), and cells were digested with 0.25% trypsin (Gibco, ThermoFisher, USA). The cells were incubated at 37 °C, 5% CO[2] incubator. Cell viability test To assess the safety of KBiF[4]@HSA, NRK cells, SHZ-88 cells or 4T1 cells were seeded in 96-well plates. After 24 h, the cells were exposed to media containing varying concentrations of KBiF[4]@HSA and incubated at 37 °C with 5% CO[2] for an additional 24 h. Cell viability was then measured using the CCK-8 assay. Hemolysis test Healthy rat blood was mixed with phosphate-buffered saline (PBS) to create a 2% red blood cell suspension, then treated with varying concentrations of KBiF[4]@HSA and incubated at 37 °C for 3 h. The blood was centrifuged at 5000 rpm for 10 min at 4 °C. The supernatant was then transferred to a 96-well plate, and its UV absorbance was measured at 540 nm. For the calculation of hemolysis, PBS at pH 7.4 served as the negative control, while 2% red blood cell lysate was the positive control: Hemolysis =(𝐴𝑠−𝐴𝑛)/(𝐴𝑝−𝐴𝑛)×100%. The absorbance value of the sample is denoted as As, while the absorbance value of the positive control is represented as Ap, and the absorbance of the negative control is indicated as An. Clone formation assay To assess the radiosensitizing effects of KBiF[4]@HSA, 4T1 cells were inoculated into 6-well plates, respectively, and cultured in RPMI-1640 medium overnight. Next, the cells were incubated with KBiF[4]@HSA co-incubation for a further 4 h, then irradiated with X-rays and cultured in normal medium. After incubation for 7 days, cells were fixed by paraformaldehyde (4%) and stained with crystal violet. Detection of intracellular GSH levels 4T1 cells in logarithmic growth phase (5 × 10^5 cells/well) were seeded into 6-well plates and dynamically monitored for cell adhesion under conditions of 37 °C and 5% CO₂. When the cell confluence reached 80%, complete medium containing KBiF[4]@HSA was added for co-incubation for 6 h. Subsequently, the medium was replaced with fresh medium, and the experimental group was exposed to X-rays (4 Gy) while the control group was not. After treatment, the cells were cultured for 24 h. The cells were then rinsed three times with PBS solution, incubated with cell lysis buffer for 30 min, scraped off, and centrifuged at 12,000 rpm for 15 min at 4 °C. The supernatant was collected as the sample to be tested. The protein concentration of the samples was determined using the BCA method. A 96-well plate was set up with blank control and experimental groups according to the instructions of the Cellular Glutathione Peroxidase Assay Kit with DTNB (Beyotime, S0057S). Glutathione peroxidase assay buffer, 10 mM GSH solution, 15 mM peroxide reagent solution, and the samples to be tested were added sequentially and mixed well. After incubating at room temperature for 30 min, 6.6 µL of DTNB solution was added to each well, mixed well, and incubated for 10 min. The absorbance was measured at a wavelength of 412 nm using a microplate reader, and the glutathione peroxidase activity was calculated. Intracellular ROS detection Intracellular ROS production was detected by 2’,7’-dichlorodihydrofluorescein diacetate (DCFH-DA). The 4T1 cells were co-incubated with KBiF[4]@HSA for 12 h. After washing with PBS for 3 times, the cells of different treatments with/without X-ray irradiation were incubated with a DCFH-DA probe for 15 min, washed with PBS and imaged with an inverted fluorescence microscope. This study successfully performed quantitative analysis of fluorescence intensity using the ImageJ software. Two-photon digital scanned light-sheet microscopy 4T1 cells were plated in a 35 mm petri dish and incubated for 24 h. To study the distribution within cells, after a 4-hour incubation with KBiF[4]@HSA, cells were additionally stained with mito-tracker green (20 nM) or lyso-tracker green (50 nM) following standard procedures. After washing with PBS twice, cells were observed using the two-photon digital scanned light-sheet microscopy. Live/dead cell staining The cells that adhered well were cultured in 24-well plates with or without KBiF[4]@HSA for 24 h. Subsequently, cells for different treatments were irradiated with or without X-rays. After this period, the cells were incubated with calcein acetoxymethyl ester (calcein-AM) and propidium iodide (PI) for 30 min at 37 °C in the dark. Intracellular fluorescence was then observed using an inverted fluorescence microscope. Animal study All animal studies were approved by the Ethics Committee of Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine. Female Sprague-Dawley (SD) rats (4–5 weeks) were supplied by the Shanghai Experimental Animal Center and housed in a specific pathogen-free environment. Mammary carcinoma tumors in situ were established by injecting 2 × 10^7 SHZ-88 cells between the right axillary mammary fat pads of each rat. Tumor generally formed about 1 week after cell injection. DECT imaging experiments were performed when the breast in situ tumor reached a diameter of about 100 mm. In addition, 4-week-old female BALB/c mice were used for the radiotherapy experiment. Firstly, 4T1 cells (1 × 10^6) were subcutaneously injected into the right side of each mouse, and tumor formed after ten days. Then, these mice were randomly divided into four groups: (1) PBS, (2) KBiF[4]@HSA, (3) PBS + X, (4) KBiF[4]@HSA + X. After injection of KBiF[4]@HSA into the tumors, the mice were exposed to X-rays. X-ray biological irradiators were obtained using RAD SOURCE RS 2000. After different treatments, measurements of the body weights and tumor volumes for mice were conducted every 2 days. The tumor volumes were calculated by the formula: V = (length × width^2)/2. DECT imaging in vitro A DECT scanner (SIEMENS SOMATOM FORCE) was employed to collect HU values and CT images using split-filter mode at 120 kVp and sequential-spiral mode at 80 keV and 150 keV for various concentrations of iohexol and KBiF[4]@HSA. Data were analyzed to generate single-energy CT images and X-ray attenuation curves. The imaging parameters were set as follows: tube voltages of 80 kVp and 150 kVp, a section thickness of 0.5 mm, a field of view (FOV) of 70 mm, and a matrix size of 512 × 512. DECT imaging in vivo DECT imaging of SD rats prior to injection was captured using a DECT system, followed by scanning imaging 5 min after injection within the in situ mammary tumors of SD rats. Prior to injection, vascular and cardiac imaging of SD rats was conducted using a DECT system. Subsequently, an indwelling needle was placed in the tail vein of the rat, and a whole-body DECT scan was performed immediately following the injection of KBiF[4]@HSA. Histology examination The rats were split into two groups at random: one group was given a tail vein injection of PBS, while the other group received KBiF[4]@HSA. After a period of 7 days, major organs, including the heart, liver, spleen, lungs, kidneys, and tumor, were collected for hematological analysis, hematoxylin and eosin (H&E) staining. In addition, mice in radiotherapy experiment were randomized into four groups: (1) PBS, (2) KBiF[4]@HSA, (3) PBS + X, and (4) KBiF[4]@HSA + X. After 14 days, tumors were harvested and subjected to H&E staining, TUNEL staining, Ki67 staining, and γ-H2AX staining. RNA sequencing methods for transcriptome analysis From cryopreserved murine tumor tissues, total RNA was extracted using TRIzol reagent (Invitrogen, CA, USA), and its purity and concentration were measured afterward. Using the VAHTS Universal V6 RNA-seq library prep kit, libraries were created and sequenced on an Illumina Novaseq 6000 platform. DESeq2 was used for differential expression analysis, with significant gene expression differences defined by a Q value < 0.05 and a fold change > 2. R (v3.2.0) was employed to examine Gene Ontology (GO) terms and assess functional enrichment, covering biological processes, molecular functions, and cellular components. Transcriptome sequencing was conducted by OE Biotech Co., Ltd. (Shanghai, China). Assessment of blood biochemical parameters The serums were obtained from whole blood after centrifugation at a speed of 3240 rpm for 15 min. The serum biochemical levels, including alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), serum total protein (TP), globulin (GLOB), albumin-globulin ratio (A/G), triglyceride (TG), creatinine (CREA), urea, calcium (Ca), potassium (K), phosphate (P), sodium (Na) and chloride (Cl), were assayed by an automatic biochemical analyzer (C501, Cobas, USA). Statistical analysis Statistical analysis of the experimental data was performed using the one-way ANOVA, two-way ANOVA and three-way ANOVA followed by post hoc test. In detail, a minimum of three repetitions was conducted for each experiment. The levels of statistical significance for the P values were represented as follows: *, P < 0.05; **, P < 0.01; ***, P < 0.005 and ****, P < 0.0001. Results and discussion The synthesis and characterization of KBiF[4]@HSA The KBiF[4] nanoclusters were synthesized in a one-pot synthesis method using the HSA-mediated bioinspired mineralization strategy. First, HSA was dissolved in EG followed by the addition of Bi[NO[3]][3]·5H[2]O at 37 °C. Carboxyl groups exhibit a strong attraction to Bi³⁺ ions, enabling HSA to firmly anchor them. After stirring for 30 min, KF·2H[2]O was added and reacted for 15 min to produce a milky white solution as KBiF[4]@HSA. After centrifugal purification, the KBiF[4]@HSA were characterized by TEM and SEM images, and the results showed that the prepared KBiF[4]@HSA is monodisperse with an average size of ∼100 nm (Fig. [86]1A and B). As shown in Fig. [87]1C, elemental mapping analysis further indicated the existence and the homogeneous distribution of Bi, F, K, C, N, O, and S elements in the KBiF[4]@HSA, suggesting the KBiF[4] nanoclusters are generated with HSA. Furthermore, the atomic force microscope (AFM) images of the KBiF[4]@HSA revealed a spherical shape with a height of 139 nm (Fig. [88]1D). The maximum height of KBiF[4]@HSA in the three-dimensional AFM image was around 140 nm, but most of the particles were also present in small uniform nanoparticles of 100 nm and below. Fig. 1. [89]Fig. 1 [90]Open in a new tab (A) TEM image of KBiF[4]@HSA (scale bar: 200 nm). (B) SEM image of KBiF[4]@HSA. (C) EDX mapping of KBiF[4]@HSA. (scale bar: 0.5 μm). (D) AFM images of KBiF[4]@HSA. (E) TEM images and DLS analysis of KBiF[4]@HSA at different timepoints during reaction (scale bar: 200 nm) The formation process of KBiF[4]@HSA was explored by TEM images and DLS measurements at reaction times of 1, 5, 10, 15, and 20 min. During the preparation process, it was found that the uniform and monodispersed KBiF[4]@HSA was created in 1 min, resulting in spherical nanoparticles with an average size of approximately 48 nm (Figure [91]S1). Moreover, the TEM images showed that KBiF[4]@HSA at 5 min are uniformly dispersed particles of about 70 nm, indicating the size of KBiF[4]@HSA increased continuously during the reaction and the formation of KBiF[4]@HSA was fast (Fig. [92]1E). Interestingly, a significant change in morphology from sphere to cluster was evidenced by the TEM image of KBiF[4]@HSA at 10 min. Furthermore, DLS analysis showed KBiF[4]@HSA at 10 min has an average hydrodynamic size of 222 nm, and the aggregation may be due to the rapid formation and untimely dispersion of KBiF[4]@HSA. With the end of the reaction, the KBiF[4]@HSA dispersed again at 15 min and remained unchanged at 20 min. To evaluate the long-term stability of KBiF[4]@HSA, we conducted a 14-day colloidal stability study after synthesis. As consolidated in Supplementary Figure [93]S2, the hydrated diameters of KBiF[4]@HSA remained stable at 79.4 ± 11.61 nm (Day 1) to 80.73 ± 21.88 nm (Day 10), with a polydispersity index (PDI) of 0.12–0.18 across all timepoints. Additionally, the hydrated diameter was measured to be 75.83 ± 13.39 on day 3, 80.73 ± 21.88 on day 5, and 76.4 ± 11.61 on day 7. This confirms excellent dispersion stability for at least 10 days. Therefore, KBiF[4]@HSA could be efficiently synthesized via a simple one-pot HSA-mediated biomimetic mineralization method. The formation of the KBiF[4]@HSA was verified through XPS, XRD and FT-IR spectroscopy. The presence of the primary elements Bi, F, and K was confirmed by XPS analysis (Fig. [94]2A). XPS survey spectrum exhibited characteristic peaks corresponding to F 1s, K 2p, Bi 4f, C 1s, N 1s, and O 1s, which reveals the integration of KBiF[4] and HSA. In the high-resolution XPS spectrum, peaks at 164 eV and 158.65 eV confirmed the presence of Bi^3+ in the KBiF[4] nanoclusters (Fig. [95]2B), while the peak at 682.7 eV signified the presence of F 1s (Fig. [96]2C). In addition, the peak at 291.55 eV indicated the presence of K 2p, and the peak at 283.75 eV confirmed the presence of C 1s (Fig. [97]2D). Furthermore, absorbance spectra of the KBiF[4]@HSA demonstrated an increased absorbance within the 500–900 nm range with the increasing KBiF[4]@HSA concentration (Fig. [98]2E). Moreover, the peaks in the XRD patterns of the lyophilized KBiF[4]@HSA powder aligned with the characteristic peaks of the standard KBiF[4]@HSA crystal structure (JCPDS 48–0667), providing evidence for the successful synthesis of KBiF[4]@HSA (Fig. [99]2F). The XRD elemental distribution spectra demonstrated that the predominant elemental composition of KBiF[4]@HSA comprised K, Bi, and F in an atomic ratio of approximately 1.58: 1.43: 7.64, closely aligning with the stoichiometric ratio of the molecular formula (Fig. [100]2G and Figure [101]S3). FT-IR spectroscopy confirmed the modification of HSA within KBiF[4]@HSA, indicating enhanced biocompatibility (Fig. [102]2H). Fig. 2. [103]Fig. 2 [104]Open in a new tab (A) High-resolution Bi 4f spectrum of KBiF[4]@HSA. (B) High-resolution F 1s spectrum of KBiF[4]@HSA. (C) High-resolution K 2p and C 1s spectrum of KBiF[4]@HSA. (D) XPS survey spectra of KBiF[4]@HSA. (E) UV − vis absorption spectra of KBiF[4]@HSA. (F) XRD pattern of KBiF[4]@HSA. (G) Spectra of the main elements of KBiF[4]@HSA. (H) FT-IR of KBiF[4]@HSA. (I) TEM of KBiF[4]@HSA incubated with GSH for 0 min, 5 min, 15 min, 30 min, 60 min (scale bar: 200 nm). (J) DLS analysis and photographs of KBiF[4]@HSA with different concentrations of GSH The GSH-responsive degradation of KBiF[4]@HSA Recently, studies have demonstrated that GSH concentrations in tumor tissue are 100–1000 times higher than those in blood and normal tissues, with notable intracellular GSH concentrations in tumor cells ranging from 1 to 15 mM [[105]49]. The GSH-responsive degradation behavior of KBiF[4]@HSA was investigated using TEM, DLS, and UV − vis absorption following incubation with GSH-containing PBS. TEM analysis revealed minimal morphological changes in KBiF[4]@HSA during the initial 15 min. However, upon extension with prolonged incubation beginning at 30 min, a progressive degradation of KBiF[4]@HSA was detected (Fig. [106]2I). To further verify the degradation process, 1 mM of KBiF[4]@HSA was exposed to varying concentrations of GSH (0 mM, 1 mM, 2 mM, 5 mM, 10 mM, and 15 mM) for 2 h. The results demonstrated a transition of KBiF[4]@HSA from a milky-white suspension to a clarified solution with increasing GSH concentrations. As depicted in Fig. [107]2J, a GSH concentration of 15 mM resulted in a reduction of the particle size to approximately 10 nm, which may facilitate the excretion of KBiF[4]@HSA in vivo. Furthermore, the increased absorption of KBiF[4]@HSA after incubation with GSH indicated that GSH may degrade KBiF[4]@HSA into bismuth ions and form multicoordinated GSH-bismuth chelates, as further evidenced by the displacement of absorption peaks (Figure [108]S4A and B). Therefore, the degradable properties may not only improve imaging outcomes, but also enhance the biocompatibility and safety profile, which is ultimately eliminated through the body’s metabolic processes. DECT-guided radiotherapy in vitro To evaluate the biocompatibility of KBiF[4]@HSA, the viability of NRK cells was assessed at varying concentrations of KBiF[4]@HSA using the CCK-8 assay. The results demonstrated cell survival rates exceeding 80% following incubation with KBiF[4]@HSA, suggesting excellent biocompatibility in normal cells (Fig. [109]3A). Moreover, given the potential of nanomaterials to induce acute hemolysis upon direct contact with blood, the hemolytic activity of KBiF[4]@HSA was further evaluated. Rat erythrocytes were incubated with varying concentrations of KBiF[4]@HSA, and hemoglobin release was quantified by measuring absorbance at 540 nm. After incubation for 24 h, the positive control group, which utilized ddH[2]O as the erythrocyte dispersion medium, demonstrated significant hemolysis, as indicated by the presence of a red supernatant (Fig. [110]3B). In contrast, the negative control group, which employed PBS, along with the groups treated with different concentrations of KBiF[4]@HSA, showed no visible hemolysis. In addition, to enhance the accuracy of hemolysis quantification, the supernatants from all groups were subjected to centrifugation. After 3 h, hemoglobin absorbance was measured using an enzyme marker to evaluate the degree of erythrocyte hemolysis. The positive controls exhibited nearly complete hemolysis, whereas negative controls and the KBiF[4]@HSA group showed no hemolysis, which indicates that KBiF[4]@HSA exhibits favorable in vivo safety characteristics for DECT imaging and radiotherapy. Fig. 3. [111]Fig. 3 [112]Open in a new tab (A) Cell viability of SHZ-88 cells and NRK cells after incubation with KBiF[4]@HSA for 24 h. (B) Hemolysis experiment of KBiF[4]@HSA. (C) Cell viability of 4T1 cells after incubation with KBiF[4]@HSA for 24 h. (D) Typical images of colony formation of 4T1 cells treated with radiotherapy. (E) Two-photon digital scanned light-sheet microscopy images of 4T1 cells with mito-tracker and lyso-tracker. (F) Fluorescence images of 4T1 cells stained with DCFH-DA were taken to observe intracellular ROS after treatments (scale bar: 200 μm). (G) Changes in GSH levels in 4T1 cells after PBS or KBiF[4]@HSA treatments. (H) Fluorescence images of 4T1 cells stained with calcein-AM/PI following diverse treatments. (I) Fluorescence quantitative analysis of calcein-AM/PI The potential therapeutic efficacy of KBiF[4]@HSA as a radiosensitizer was further investigated. The viability of 4T1 cells treated with KBiF[4]@HSA and radiotherapy was assessed using the CCK-8 assay. After 24 h of co-incubation with KBiF[4]@HSA alone, the cell survival rate of 4T1 cells was 80%. Notably, when KBiF[4]@HSA was combined with X-ray treatment, substantial cytotoxicity was observed (Fig. [113]3C). In addition, the clonal proliferation assays illustrated that KBiF[4]@HSA alone did not significantly impact cell colony formation (Fig. [114]3D). Conversely, KBiF[4]@HSA and X-rays exposure led to a marked reduction in cell colony formation. Moreover, as shown in Fig. [115]3E, the red fluorescence of squaraine dye (SQ) labeled on KBiF[4]@HSA appeared in cells, exhibiting mitochondria-localization (Pearson’s correlation coefficient, PCC: 0.73 ± 0.03) and lysosome-localization tendency (PCC: 0.85 ± 0.03). Furthermore, utilizing DCFH-DA as an intracellular ROS indicator, cells treated with PBS and X-rays exhibited mild fluorescence, suggesting a limited ROS generation by PBS + X treatment (Fig. [116]3F). In contrast, the KBiF[4]@HSA + X group demonstrated the most intense fluorescence, providing direct evidence of the radiosensitizing effects of KBiF[4]@HSA. Notably, cells treated with KBiF[4]@HSA alone also exhibited strong fluorescence, indicating ROS generation. To elucidate the underlying mechanism, GSH levels were subsequently measured, revealing a significant decrease in the KBiF[4]@HSA group (Fig. [117]3G). These findings indicate that KBiF[4]@HSA can effectively scavenge the elevated GSH and disrupt the redox balance in cancer cells, resulting in increased ROS levels. In addition, calcein-AM/PI staining revealed that the majority of cells in PBS and KBiF[4]@HSA groups, suggesting robust cell viability and confirming the safety profile of KBiF[4]@HSA (Fig. [118]3H). Notably, the KBiF[4]@HSA + X group had significant tumor cell death compared to irradiation alone. This conclusion is supported by fluorescence quantitative analysis (Fig. [119]3I). In vitro DECT imaging with KBiF[4]@HSA The DECT imaging efficacy of KBiF[4]@HSA was evaluated, with iohexol clinical CAs utilized as a comparative control. The KBiF[4]@HSA and iohexol at different concentrations (0, 0.3125, 0.625, 1.25, 2.5, 5, and 10 mM) were placed in tubes and scanned by a CT imaging system. At the same concentration of radioactive elements, the CT signal of KBiF[4]@HSA was stronger than that of iohexol with increasing energy, especially at high keV energy imaging, where KBiF[4]@HSA maintained stable imaging. The monochromatic image showed that the CT values for both KBiF[4]@HSA and iohexol decrease as keV energy increases. However, the decrease in KBiF[4]@HSA was significantly slower than that of iohexol (Fig. [120]4A). This disparity can be attributed to the enhanced X-ray attenuation characteristics of bismuth elements in comparison to iodine elements at elevated energy levels. Monochrome energy images displayed across the energy range of 40–180 keV showed that the signal from iohexol decreased progressively with increasing energy. In contrast, the CT signal of KBiF[4]@HSA consistently exhibited greater intensity and persistence compared to iohexol across the entire spectrum of energy levels. This finding aligns with the trends observed in the Hounsfield unit (HU) curves. In comparison, the CT signal of KBiF[4]@HSA was persistently higher and more potent than iohexol across the entire energy levels. This findings align with the HU curves (Fig. [121]4B-F). In higher single-energy images, KBiF[4]@HSA shows superior X-ray attenuation properties compared to iohexol, indicating the advanced DECT imaging capabilities of KBiF[4]@HSA for higher diagnostic precision and contrast resolution. Fig. 4. [122]Fig. 4 [123]Open in a new tab (A) In vitro CT imaging at different keV and energy spectrum curves of KBiF[4]@HSA and iohexol. (B-F) In vitro CT imaging of KBiF[4]@HSA and iohexol at (B) 40 keV, (C) 60 keV, (D) 80 keV, (E) 150 keV, (F) 180 keV. The data are shown as the mean ± standard deviation (SD), as calculated using two-way ANOVA followed by post hoc tests with *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 In vivo Dect imaging with KBiF[4]@HSA To evaluate the DECT imaging efficacy of KBiF[4]@HSA, SHZ-88 cells were injected into the mammary gland of 5-week-old SD rats to establish in situ breast cancer tumors. Then, KBiF[4]@HSA (10 mM, 20 µL) and iohexol (10 mM, 20 µL) were administered via intratumoral injection. The CT signals of the tumor prior to injection were utilized as a control, and subsequent CT signals of the tumor were monitored post-injection. As depicted in Fig. [124]5A and B, the signal intensity within the tumor region exhibited significant variation in the monochrome energy image before and after injection. Specifically, the CT value of the tumor imaged before injection was approximately 56.07 HU at 80 keV and 34.06 HU at 150 keV (Figure [125]S5A). Subsequent to the administration of KBiF[4]@HSA, there was a significant enhancement in the CT signal within the tumor region at 80 keV, with a measured CT value of 491.35 HU, representing an approximate 9-fold increase compared to the pre-injection imaging. At 150 keV, the tumor region exhibited a high CT signal, with a recorded CT value of 368.66 HU, and this elevated signal intensity was maintained as the X-ray energy increased (Figure [126]S5B). Fig. 5. [127]Fig. 5 [128]Open in a new tab (A) DECT imaging of a tumor with iohexol at different radiation energies. (B) DECT imaging of a tumor with KBiF[4]@HSA at different radiation energies. (C) DECT imaging of the tumor at different anatomical positions with KBiF[4]@HSA and iohexol. (D) DECT imaging of the tumor at the effective atomic numbers (Z[eff]) with KBiF[4]@HSA. (E) DECT values at different keV in the tumor region with KBiF[4]@HSA. (The blue circle indicates the tumor location.) In contrast, for tumors subjected to intratumoral injection with iohexol, the CT value of the tumor region prior to injection was 41.29 HU at 80 keV and 29.11 HU at 150 keV (Figure [129]S6A). After the injection, the CT value of the tumor area rose to about double its pre-injection level. Specifically, at 80 keV, the tumor region showed slight visualization with a measured CT value of 127.12 HU. The CT signal of the tumor was notably attenuated at 150 keV, with no observable contrast-enhanced CT signal upon further increases in X-ray energy, as evidenced by a measured CT value of 47.96 HU (Figure [130]S6B). Quantitative evaluation and statistical analysis of in vivo imaging signals were conducted for KBiF[4]@HSA and iohexol, further confirming the increased signal intensity of KBiF[4]@HSA compared to iohexol (Figure [131]S7-[132]8). These findings suggest that iohexol is effective exclusively in contrast-enhanced imaging at lower kilovoltage levels, with its contrast-enhancing properties diminishing progressively as X-ray energy levels increase. The observed phenomenon may stem from the inherent characteristics of iohexol. As a water-soluble contrast agent, its low density and rapid diffusion following in vivo injection prevent adequate contrast enhancement in DECT scans at low dosage levels. Conversely, KBiF[4]@HSA demonstrates elevated K-edge energy, increased density, and enhanced stability, along with heightened sensitivity. Subsequently, a comparative analysis of CT signal intensities between KBiF[4]@HSA and iohexol was conducted in tumor regions across various anatomical positions. KBiF[4]@HSA exhibited a significantly higher CT signal intensity than iohexol (Fig. [133]5C and D). For image reconstruction, the Z[eff] mode was selected using the dual-energy mode on the Siemens postprocessing workstation. KBiF[4]@HSA incorporates the high atomic number element Bi, which is capable of producing significant signals at the tumor site post-injection, as evidenced with a measured Z[eff] value of 10.13 HU (Fig. [134]5E and [135]S9). These findings indicate that KBiF[4]@HSA exhibits consistent X-ray attenuation properties, thereby establishing its reliability as a contrast agent for DECT imaging in the context of disease diagnosis. To explore the imaging capabilities of KBiF[4]@HSA, their efficacy in cardiac imaging was also evaluated. Firstly, vasculature imaging was performed on 5-week-old healthy rats via tail-vein injection, using the same concentration of iohexol and KBiF[4]@HSA (10 mM, 1.5 mL) for comparison. In rats injected with KBiF[4]@HSA, a significant increase in the CT signal of the abdominal aorta was observed, with an increased CT signal as the blood circulated to the cardiac region (Figure [136]S10A). Notably, upon increasing the energy of X-ray radiation, the cardiac CT signal intensity remained consistent across the energy range of 40–180 keV. In contrast, rats injected with iohexol exhibited diminished CT signals, lacking contrast enhancement at elevated radiation energies (Figure [137]S10B). Quantitative evaluation and statistical analysis of in vivo cardiac imaging signals were conducted for KBiF[4]@HSA and iohexol, further confirming the increased signal intensity of KBiF[4]@HSA compared to iohexol (Figure [138]S11-[139]12). These findings indicate that vascular contrast enhancement can be effectively achieved using KBiF[4]@HSA at low concentrations, whereas iohexol necessitates a significantly higher concentration to achieve visualization, which consequently elevates its toxicity within the body. Furthermore, the imaging characteristics of KBiF[4]@HSA within the vasculature were evaluated at multiple anatomical sites and Z[eff] values both pre- and post-injection. The results demonstrated a significant enhancement of the CT signal within the vascular system (Figure [140]S10C and D). This finding highlights the potential of KBiF[4]@HSA for imaging ventricles and major blood vessels at low concentrations, thereby achieving improved contrast. This capability is anticipated to have potential applications in the imaging of vascular tumors, arterial stenosis, and other related pathologies. The efficiency of radiotherapy with KBiF[4]@HSA in vivo To assess in vivo radiotherapy efficiency, 4T1 cells were injected into the dorsal skin of mice to establish a subcutaneous tumor model. According to the procedures shown in Fig. [141]6A, the mice underwent two sessions of irradiation, with or without 6 Gy of X-ray exposure at the tumor sites, after intratumoral injection of either PBS or KBiF[4]@HSA. As shown in Fig. [142]6B, the KBiF[4]@HSA and PBS + X groups did not exhibit significant tumor growth inhibition compared to the PBS group. Significantly, tumor growth was delayed in the KBiF[4]@HSA + X group. In addition, no significant weight changes were detected across any of the treatment groups (Fig. [143]6C). At the end of therapy, the excellent therapeutic efficacy in the KBiF[4]@HSA + X group was further confirmed by the quantitative tumor weights (Fig. [144]6D). To elucidate the potential therapeutic mechanism of KBiF[4]@HSA in radiotherapy at the genetic level, a transcriptome analysis was performed to examine the expression of messenger RNA (mRNA). The analysis identified 307 differentially expressed genes (DEGs) when comparing the KBiF[4]@HSA group with the PBS group, including 240 upregulated and 67 downregulated genes. Conversely, when comparing the KBiF[4]@HSA group, the KBiF[4]@HSA + X group showed 288 gene differences, including 46 upregulated and 242 downregulated genes (Figure [145]S13A and B). Furthermore, the heatmap analysis demonstrated a notable difference in mRNA expression (Fig. [146]6E and F). Considering the variability in RNA sequencing outcomes, we performed Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO)analysis to attain a more comprehensive understanding of the changes in the biological functions of mRNAs and their related pathways (Fig. [147]6G and H). According to the KEGG enrichment analysis, the DEGs are chiefly enriched in the JAK-STAT signaling pathway, Th1 and Th2 cell differentiation, and the IL-17 signaling pathway. Fig. 6. [148]Fig. 6 [149]Open in a new tab Radiosensitizing effects of KBiF[4]@HSA in vivo. (A) Flow chart of the radiotherapy experiment in vivo. (B) Tumor growth curves of 4T1-bearing tumor mice. (C) Body weights and (D) tumor weights of mice after various treatments. (E) Heatmap diagram of differentially expressed mRNAs screened from PBS and KBiF[4]@HSA group (n = 3). (F) Heatmap diagram of differentially expressed mRNAs screened from KBiF[4]@HSA group and KBiF[4]@HSA + X group (n = 3). (G) KEGG enrichment analysis of the PBS group versus the KBiF[4]@HSA group (Top 20 pathways). (H) The results of KEGG pathway enrichment analysis of the PBS group versus the KBiF[4]@HSA + X group (Top 20 pathways). (I) H&E, TUNEL, Ki67, γ-H2AX staining images of tumors after treatments (scale bar: 100 μm). The data are shown as the mean ± SD, calculated using three-way ANOVA, followed by one-way ANOVA and post hoc tests with *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 Subsequently, H&E staining of tumor sections was conducted to further assess the extent of cellular damage and necrosis (Fig. [150]6I). The KBiF[4]@HSA + X group exhibited significant histopathological alterations. To detect DNA fragmentation, a hallmark of apoptosis, TUNEL staining was employed as both a molecular biological and morphological method. Enhanced green fluorescence was observed in the KBiF[4]@HSA + X groups compared to the PBS group, indicating increased apoptotic activity. Additionally, Ki67 immunofluorescence analysis was performed to evaluate the proliferative capacity of the tumor cells. As expected, mice in the KBiF[4]@HSA + X group exhibited effective inhibition of tumor invasion. Moreover, γ-H2AX staining was employed to examine DNA double-strand breaks, revealing that cancer cells treated with the KBiF[4]@HSA + X group exhibited significant fluorescence compared to other groups, confirming the greatest DNA damage for inhibition. In vivo safety and metabolism assessment of KBiF[4]@HSA In the development of nanobiomaterials, in vivo biocompatibility and biodegradability are critical considerations. Following intravenous administration of KBiF[4]@HSA, no significant abnormal behavior or weight loss was observed in rats over a 7-day period, indicating an absence of acute toxicity. To further assess the in vivo biocompatibility of KBiF[4]@HSA nanoclusters, hematological analyses and histological examination were conducted. The liver and kidneys, being the primary organs for the metabolism and excretion of KBiF[4]@HSA, were specifically evaluated. To assess whether KBiF[4]@HSA impacts liver and kidney function, the blood of rats was taken for the important hepatic and renal function indices tests, including ALT, AST, ALP, TP, ALB, GLOB, Ca, K, P, Na, Cl, A/G, CREA, UREA, and TG. One day and seven days post-injection, the aforementioned biochemical indices remained within the normal range, exhibiting no significant deviations from standard values, thereby confirming minimal side effects on renal and hepatic functions (Fig. [151]7A-D). In addition to blood analyses, histopathological examinations were performed to assess the potential toxicity of KBiF[4]@HSA on major organs using H&E staining. As illustrated in Fig. [152]7E, no discernible tissue damage, inflammation, or lesions were observed in the primary organs of rats treated with KBiF[4]@HSA. As a result, these findings provide an initial validation of the biocompatibility of KBiF[4]@HSA for in vivo biomedical applications. Fig. 7. [153]Fig. 7 [154]Open in a new tab Liver function indicators: ALT, ALP (A); Ca, K, P, Na, Cl (B); ALB, TP, GLOB, A/G (C) with KBiF[4]@HSA pre-injection and post-injection at 1 and 7 days. Kidney function indicators: CREA, UREA, TG (D) with KBiF[4]@HSA pre-injection and post-injection at 1 and 7 days. (E) H&E staining of tumor sections from tumor-bearing rats (scale bar: 50 μm) Conclusion In summary, the Bi-based DECT CAs, KBiF[4]@HSA could be efficiently synthesized using a straightforward one-pot HSA-mediated biomimetic mineralization strategy. Compared to the clinical CAs iohexol, KBiF[4]@HSA significantly enhances the CT value and imaging contrast at high keV, thereby improving the diagnostic accuracy for breast cancer. Furthermore, our study illustrates that KBiF[4]@HSA effectively scavenges the elevated GSH and increases the ROS levels in cancer cells, which augments the sensitivity of radiotherapy. Meanwhile, the combination of KBiF[4]@HSA with radiotherapy effectively inhibits the progression of breast cancer. Notably, the KBiF[4]@HSA exhibits no detectable tissue damage or inflammation, highlighting their superior biocompatibility in vivo. This study introduces a novel approach for developing biocompatible DECT CAs and radiosensitizers, offering a promising strategy for improving breast cancer diagnosis and treatment. However, further studies are needed to validate efficacy in diverse breast cancer subtypes, which highlights the need for continued research to fully translate this technology into clinical practice. Electronic supplementary material Below is the link to the electronic supplementary material. [155]Supplementary Material 1^ (2.3MB, docx) Author contributions Yuelin Huang: Writing–original draft, Conceptualization, Data curation, Formal analysis, Investigation, Validation, Visualization. Zhenghai Lu: Conceptualization, Data curation, Formal analysis, Investigation, Validation, Visualization. Huanhuan Liu: Formal analysis, Methodology, Software, Validation, Visualization. Ningxin Yang: Data curation, Formal analysis, Investigation, Validation; Yanhong Chen: Data curation, Formal analysis, Investigation, Visualization. Chunting Wang: Data curation, Validation, Visualization. Weixi Huang and Jingjing Hu: Formal analysis, Resources; Dengbin Wang: Funding acquisition, Project administration, Supervision. Defan Yao: Writing-original draft, Writing-review & editing, Project administration, Supervision, Funding acquisition Conceptualization, Formal analysis. All authors reviewed the manuscript. Funding This work was supported by the National Natural Science Foundation of China (92159203, 82471976, 82472039, 82071870), the Explorer Program of the Science and Technology Commission of Shanghai Municipality (24TS1415300, 24TS1415200), the Construction Project of the "Discipline Peak-Climbing Plan" of Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (XKPF2024C200, XKPF2024C201, XKPF2024C202), Shanghai Municipal Health Youth Talent Program (2022YQ042), and the Shanghai Rising-Star Program (22QA1406000). Data availability No datasets were generated or analysed during the current study. Declarations Ethical approval The ethical review board of the XinHua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, China, approved the experiment protocol and strictly followed its guidelines (Ethical approval number: XHEC-F-2024-014). Consent for publication All authors read and agreed to submit the manuscript. Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Yuelin Huang and Zhenghai Lu contributed equally to this work. Contributor Information Huanhuan Liu, Email: liuhuanhuan@xinhuamed.com.cn. Dengbin Wang, Email: wangdengbin@xinhuamed.com.cn. Defan Yao, Email: yaodefan@xinhuamed.com.cn. References