Radioactive hybrid semiconducting polymer nanoparticles for imaging-guided tri-modal therapy of breast cancer

Junhao Gu a, Danling Cheng a, Haiyan Li b, Tao Yu a, Zhenghe Zhang a, Yue Liu a, Xiaoying Wang *c, Xia Lu *b and Jingchao Li *a
aState Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China. E-mail: jcli@dhu.edu.cn
bDepartment of Nuclear Medicine, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, P. R. China. E-mail: lxgf2222@163.com
cOffice of Hospital Infection and Disease Control and Prevention, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China. E-mail: goodwxyz@163.com

Received 17th April 2024 , Accepted 17th May 2024

First published on 20th May 2024


Abstract

Due to the rapid progression and aggressive metastasis of breast cancer, its diagnosis and treatment remain a great challenge. The simultaneous inhibition of tumor growth and metastasis is necessary for breast cancer to obtain ideal therapeutic outcomes. We herein report the development of radioactive hybrid semiconducting polymer nanoparticles (SPNH) for imaging-guided tri-modal therapy of breast cancer. Two semiconducting polymers are used to form SPNH with a diameter of around 60 nm via nano-coprecipitation and they are also labeled with iodine-131 (131I) to enhance the imaging functions. The formed SPNH show good radiolabeling stability and excellent photodynamic and photothermal effects under 808 nm laser irradiation to produce singlet oxygen (1O2) and heat. Moreover, SPNH can generate 1O2 with ultrasound irradiation via their sonodynamic properties. After intravenous tail vein injection, SPNH can effectively accumulate in the subcutaneous 4T1 tumors of living mice as verified via fluorescence and single photon emission computed tomography (SPECT) imaging. With the irradiation of tumors using an 808 nm laser and US, SPNH mediate photodynamic therapy (PDT), photothermal therapy (PTT) and sonodynamic therapy (SDT) to kill tumor cells. Such a tri-modal therapy leads to an improved efficacy in inhibiting tumor growth and suppressing tumor metastasis compared to the sole SDT and combinational PDT–PTT. This study thus demonstrates the applications of SPNH to diagnose tumors and combine different therapies for effective breast cancer treatment.


image file: d4tb00834k-p1.tif

Jingchao Li

Jingchao Li received his PhD degree in Materials Science and Engineering from the University of Tsukuba (Japan) in 2017. He then worked as a postdoctoral research fellow in the School of Chemical and Biomedical Engineering, Nanyang Technological University. He is currently a Professor in the College of Biological Science and Medical Engineering at Donghua University. Dr Li has published more than 150 SCI papers in famous international journals such as Nat. Commun., Adv. Mater. and Angew. Chem., Int. Ed. with an H-index of 60. His research focuses on the development of advanced nanomaterials for cancer theranostics.


Introduction

Breast cancer is a malignant tumor that has seriously affected the survival rate of female patients.1,2 For decades, the traditional treatment methods including surgery, radiotherapy,3,4 chemotherapy5 and their combined therapy have failed to achieve ideal therapeutic outcomes. Surgical excision is the major option for breast cancer, but it may cause the issue of incomplete resection, leading to tumor recurrence.6,7 Although chemotherapy and radiotherapy can improve the therapeutic effect to a certain extent, there are some inevitable problems such as poor tumor specificity and serious side effects.8,9 Accordingly, it is important to explore some more effective and safe therapeutic strategies for breast cancer treatment.10–14

In recent years, some therapeutic strategies have shown promising opportunities for cancer treatment, such as photothermal therapy (PTT), photodynamic therapy (PDT) and sonodynamic therapy (SDT).15 PTT has shown promise as an approach for cancer treatment because the photothermal agents can absorb the optical energy and convert the photoenergy into heat to ablate cancer cells.16,17 PDT usually utilizes photosensitizers to produce a certain amount of reactive oxygen species (ROS) under light irradiation at an appropriate power to induce manifest and irreversible damage to cancer cells.18,19 Both PTT and PDT have unique advantages, such as low side effects, high therapeutic selectivity and negligible drug resistance.20–24 Besides, SDT utilizes sonosensitizers to generate ROS via the sonodynamic effect under ultrasound (US) irradiation.25–27 Similar to PDT, SDT has been reported to have slight toxicity to adjacent normal tissues and thus widely used for tumor ablation.28–31 In addition, because US penetrates deeply into the tumor tissues, SDT can overcome the tissue penetration depth limitations.32 However, tumors show a complicated microenvironment that will compromise the efficacies of various therapies.33 For example, the expression of heat shock proteins limits the PTT effect for ablating tumor cells.33,34 The hypoxic tumor microenvironment with high levels of glutathione can obviously restrict the efficacies of PDT and SDT.35,36 Therefore, a sole therapeutic strategy usually fails to achieve satisfactory outcomes, and the combination of multiple therapies is highly desired.37

Semiconducting polymer nanoparticles (SPN) formed from semiconducting polymers (SPs) with a π-conjugated aromatic framework have been used for fluorescence imaging, PTT, PDT and SDT due to their good optical absorbance, structural stability and high biosafety.38 For example, near-infrared (NIR)-absorbing poly[2,6-(4,4-bis-(2-ethylhexyl)-4H-cyclopenta[2,1-b;3,4b′]dithiophene)-alt-4,7(2,1,3-benzothiadiazole)] (PCPDTBT) can be activated by NIR light to produce 1O2 and heat for both PDT and PTT.39–41 Another SP, poly[2,7-(9,9-di-octyl-fluorene)-alt-4,7-bis(thiophen-2-yl)benzo-2,1,3-thiadiazole] (PFODBT), has been used as a sonosensitizer to mediate SDT via generating 1O2.42–47 Therefore, the integration of two different SPs into one therapeutic system will enable combinational therapy with high efficacy, which however has been poorly reported.

In this study, radioactive hybrid semiconducting polymer nanoparticles (SPNH) are fabricated to enable imaging-guided PDT–PTT–SDT combination therapy for tumor ablation and metastasis inhibition. SPNH are formed via the nano-coprecipitation of PCPDTBT, PFODBT and an amphiphilic triblock copolymer F127 and their surface is labeled with 131I for single photon emission computed tomography (SPECT) imaging (Fig. 1a). Under 808 nm laser and US irradiation, SPNH mediate the generation of heat by PCPDTBT for PTT, and also abundant 1O2 by PCPDTBT and PFODBT for PDT and SDT. Such a tri-modal therapy can greatly ablate tumor cells to inhibit tumor growth and effectively suppress tumor metastasis in the lungs and liver (Fig. 1b). The therapeutic efficacy of PDT, PTT and SDT combinational therapy is higher than that of sole SDT and combinational PDT–PTT.


image file: d4tb00834k-f1.tif
Fig. 1 Design of SPNH for imaging-guided tri-modal therapy. (a) The chemical structure formulas of PCPDTBT and PFODBT, and the formation of SPNHvia nanoprecipitation and labeling of 131I. (b) Schematic illustration of SPNH for fluorescence/SPECT imaging-guided tri-modal therapy.

Experimental section

Materials

PCPDTBT and PFODBT were obtained from Sigma-Aldrich (USA). The cell counting kit-8 (CCK-8) was obtained from Molecular Probes Inc. Chemical reagents used in all experiments were provided by Sinopharm (China). 2′,7′-Dichlorodihydrofluorescein diacetate (H2DCFDA) and Pluronic® F-127 were purchased from Shanghai Macklin Biochemical Technology Co., Ltd (China). A singlet oxygen sensor green (SOSG) fluorescent probe was obtained from Thermo Fisher Scientific (USA).

Characterization

Morphological characteristics were analyzed using a Tecnai transmission electron microscope (JEOL, TEM JEM-2100, Japan). An IVIS Lumina Series III system was used to obtain the fluorescence images of living animals (IVIS machine, PerkinElmer, USA). An inverted fluorescence microscope (Leica, DMi8, Germany) was used to obtain cell fluorescence images. An absorption spectrophotometer (Persee, TU-1810, Beijing, China) was utilized to detect ultraviolet absorption characteristics. The fluorescence properties were evaluated using a fluorescence spectrophotometer (SHIMADZU, RF-6000, Kyoto, Japan). A zeta potential and particle sizer (Malvern, Zetasizer Nano S90, UK) was used to measure the hydrodynamic sizes and zeta potential values.

Synthesis of SPNH

SPN were synthesized by a nano-precipitation method.47–49 In general, 0.25 mg of PCPDTBT and 0.25 mg of PFODBT were dissolved in 2 mL of tetrahydrofuran (THF), respectively. 50 mg of Pluronic® F-127 dissolved in 2 mL of THF was blended with the above solution. Then the solution was added into 10 mL of THF/water (V/V = 1[thin space (1/6-em)]:[thin space (1/6-em)]9) and sonicated. THF was evaporated overnight, and the solution was filtered through a 200 nm membrane and ultrafiltrated to obtain SPN. SPNH were obtained by labeling the SPN with 131I. In brief, 1 mg of SPN, 20 mCi Na131I and 1 mg of chloramine-T were mixed and reacted at room temperature for 2 h, and then the solution was centrifuged 5 times to obtain SPNH. A TU-1810 Persee spectrophotometer was used to determine the concentrations of SPNH.

Photothermal effect evaluation

The SPNH solution was diluted with ultrapure water to prepare samples with SP concentrations of 12.5, 25, 50, and 100 μg mL−1, respectively. An 808 nm laser with a power density of 1.0 W cm−2 was used to irradiate the sample solutions for 6 min. A Fotric 220s thermal camera was utilized to record temperature changes every 5 s. After irradiation, the temperatures of SPNH solutions were measured every 5 s for another 360 s. For the evaluation of the photothermal stability, SPNH solutions at SP concentrations of 12.5, 25, 50, and 100 μg mL−1 were irradiated under the same conditions, and the changes of temperature were monitored after five laser on and laser off cycles.

Evaluation of 1O2 generation efficacy

A 1O2 fluorescence probe (SOSG) was utilized to measure 1O2 generation efficacy under laser and US irradiation. A SPNH solution at a SP concentration of 50 μg mL−1 (3 mL) was mixed with SOSG (3 μL, 500 μM) and the initial fluorescence intensity (F0) was measured. The solutions were divided into sole laser (1.0 W cm−2, 10 min), sole US (1.0 W cm−2, 1.0 MHz, 50% cycle, 10 min) and laser (1.0 W cm−2, 5 min) plus US (1.0 W cm−2, 1.0 MHz, 50% cycle, 5 min) groups. The fluorescence intensities of SOSG in solutions (F) after various treatments were recorded. The 1O2 generating efficiency was calculated using F/F0.

Evaluation of cytotoxicity in vitro

4T1 cancer cells were seeded in 96-well cell culture plates and then incubated with SPNH. The cell viability was measured via cell counting kit 8 (CCK-8) analysis.

Evaluation of the therapeutic effect in vitro

4T1 cells were treated with PBS (control) or SPNH at a SP concentration of 50 μg mL−1 for 12 h. The cells were then treated with US at a power density of 1.0 W cm−2 (1.0 MHz, 50% cycle, 2.5 min) and an 808 nm laser at a power density of 1.0 W cm−2 (2.5 min). To confirm the PTT effect, the treated cells were also incubated with N-acetyl-L-cysteine (NAC) to scavenge ROS.50–52 During laser irradiation, the temperatures of cells were controlled below 43 °C to verify the PDT effect. After the cells were cultured for another 12 h, the CCK-8 assay was used to measure the cell viability.

In vitro dead/live staining analysis

4T1 cells were incubated with SPNH at a SP concentration of 50 μg mL−1 for 12 h and then treated with US at a power density of 1.0 W cm−2 (1.0 MHz, 50% cycle, 2.5 min) and an 808 nm laser at a power density of 1.0 W cm−2 (2.5 min). After that, the cells were cultured for 12 h and stained with 0.1% PI and 0.1% Calcein for 15 min, and the dead/live staining images of cells were then obtained.

Detection of ROS in cells

After 4T1 cells were incubated with SPNH (50 μg mL−1) for 12 h, the fluorescent dye H2DCFDA solution was added into each well for cell incubation. The cells were then treated with US (1.0 W cm−2, 1.0 MHz, 50% cycle, 2.5 min) and an 808 nm laser (1.0 W cm−2, 2.5 min). The generation of ROS in cells was evaluated by detecting fluorescence signals using an inverted fluorescence microscope.

Establishment of mouse tumor models

All the animal experiments were approved by the Animal Care and Use Committee of Donghua University. BALB/c mice (5-week-old) were used to establish subcutaneous 4T1 tumor-bearing mouse models. In brief, 2 × 106 4T1 cells suspended in PBS were injected into the right flank of mice to build the mouse tumor models.

In vivo fluorescence and SPECT imaging

After systemic administration of SPNH (SP concentration = 250 μg mL−1, 200 μL) into subcutaneous 4T1 tumor-bearing mice, the fluorescence images of mice at predetermined time points were captured using an IVIS Lumina Series III imaging system. After 27 h of systemic administration, the mouse tumor models were euthanized to extract tumors and main organs for detecting their fluorescence signals.

The in vivo SPECT imaging of 4T1 tumor-bearing mice was performed using an Infinia GE SPECT equipped with a Xeleris workstation and a high energy general purpose detector (GE Inc., Fairfield, CT). The mice were intravenously injected with SPNH at a SP concentration of 250 μg mL−1 (200 μL). At different time points post injection, SPECT images of tumor-bearing mice were captured and the signal values of tumor sites were measured. At 27 h post-injection, the main organs and tumors were extracted to measure the SPECT signals.

In vivo PTT effect evaluation

4T1 tumor-bearing BALB/c mice were intravenously injected with PBS or SPNH (200 μL, 250 μg mL−1). At 12 h after post-injection, the tumor sites of mice were exposed to 808 nm laser irradiation for 300 s (1.0 W cm−2). An infrared thermographic camera (Fotric 225, USA) was used to record the temperature changes in the tumor area during laser irradiation.

In vivo tumor therapy

4T1 tumor-bearing BALB/c mice were intravenously injected with PBS or SPNH (250 μg mL−1, 200 μL). The mice for in vivo therapeutic evaluation were divided into six groups: PBS (control), PBS + laser + US, SPNH, SPNH + US, SPNH + laser, and SPNH + US + laser (n = 5). The tumors were irradiated by US (1.0 W cm−2, 1.0 MHz, 50% cycle, 5 min) and/or using an 808 nm laser (1.0 W cm−2, 5 min) in different groups. The tumor volumes were calculated based on the measured tumor sizes and the body weights of mice were recorded. After 14 days of treatment, the mice in each group were sacrificed to extract the tumors and main organs for histological analysis via hematoxylin and eosin (H&E) staining. The Kaplan–Meier survival curves of mice were established by recording the survival of mice after various treatments.

In vivo metastasis inhibition analysis

4T1 tumor-bearing BALB/c mice were randomly divided into six groups: PBS (control), PBS + laser + US, SPNH, SPNH + US, SPNH + laser and SPNH + US + laser (n = 5). These mice were injected with PBS or SPNH (250 μg mL−1, 200 μL) and the tumors were exposed to US (1.0 W cm−2, 1.0 MHz, 50% cycle, 5 min) and 808 nm laser (1.0 W cm−2, 5 min) irradiation at 24 h post-injection. After 30 days of treatment, the treated mice were euthanized to obtain the lungs and livers for H&E staining. The metastasis inhibition efficacies in the lungs and livers were analyzed by calculating the areas of metastatic tumor nodules.

Statistical analysis

All the experiments were repeated at least three times, and mean ± standard deviation (SD) was used to represent the data. The experimental data were statistically analyzed by one-way ANOVA. The values of p less than 0.05 indicated statistical significance, that is, (*)p < 0.05, (**)p < 0.01, and (***)p < 0.001.

Results and discussion

Synthesis and characterization of SPNH

SPN were synthesized via a nanoprecipitation method using two SPs (PCPDTBT and PFODBT) and F127.47–49 At the optimized feeding weight of PCPDTBT, PFODBT and F127 of 1[thin space (1/6-em)]:[thin space (1/6-em)]1[thin space (1/6-em)]:[thin space (1/6-em)]200, the hydrodynamic size was the smallest (62.1 nm), while it was larger than 110 nm at other feeding weights (Table S1, ESI). Then SPN were labeled with 131I to form the final SPNH. 131I was covalently immobilized onto the aromatic backbone of SPs via electrophilic substitution of protons, which enabled stable labeling. The transmission electron microscopy (TEM) image demonstrated that SPNH displayed a homogeneous granular morphology with a diameter of about 60 nm (Fig. 2a). Subsequently, the hydrodynamic size of SPNH was measured to be 62.1 nm (Fig. 2b). The zeta potential of SPNH was −25.7 mV (Fig. 2c), and this negative charge was conducive to in vivo stability and biosafety.53 Furthermore, the average sizes of SPNH in water, PBS and DMEM cell culture medium did not change significantly (Fig. 2d). Their polydispersity index (PDI) was measured to be around 0.15 and did not obviously change in water, PBS and DMEM cell culture medium within 14 days (Fig. S1, ESI), demonstrating that they had good stability in different systems. The absorbance and fluorescence properties of SPNH were detected. SPNH displayed the definite characteristic peaks of PCPDTBT and PFODBT at 380, 540 and 665 nm. The characteristic absorption peaks of SPNH at 380 and 665 nm overlapped with those of PCPDTBT, while the characteristic absorption peak at 540 nm was assigned to PFODBT (Fig. 2e). The fluorescence spectrum showed that SPNH had a fluorescence peak at 845 nm (Fig. 2f). The radiolabeling efficacy of 131I in SPNH was as high as 99.0% after purification by centrifugation. After 24 h incubation in fetal bovine serum (FBS) and PBS at 37 °C, the radiochemical purity of SPNH remained higher than 90.8% (Fig. S2, ESI), which indicated the good radiochemical stability of SPNH.
image file: d4tb00834k-f2.tif
Fig. 2 Characterization of SPNH. (a) Morphology characterization of SPNH using TEM imaging. (b) Hydrodynamic diameter profiles of SPNH measured in aqueous solution. (c) Determination of the zeta potential of SPNH in aqueous solution (n = 3). (d) Hydrodynamic diameters of SPNH measured in water on days 1, 3, 5, 7, 10 and 14 (n = 5). (e) Ultraviolet–visible absorption spectra of PFODBT, PCPDTBT and SPNH. (f) Fluorescence spectrum of SPNH. (g) Temperature curves of aqueous solutions containing SPNH under 808 nm laser irradiation for 360 s. (h) The photothermal stability study of SPNH after five laser on/off cycles at different concentrations. (i) The 1O2 generation efficacies of SPNH under US and laser irradiation (n = 3).

The photothermal effect of SPNH under laser irradiation was evaluated. Thermal imaging showed that the temperatures of solutions containing SPNH under laser irradiation were positively correlated with SP concentration (Fig. S3, ESI). The temperature of SPNH solutions was found to increase to 36.2, 40.6, 47.7 and 59.4 °C for irradiated groups at concentrations of 12.5, 25, 50 and 100 μg mL−1, respectively (Fig. 2g). At concentrations of 12.5, 25, 50, and 100 μg mL−1, the maximum temperatures of SPNH solutions after five laser on–off cycles did not have obvious changes (Fig. 2h), suggesting the excellent photothermal stability of SPNH. In the photothermal cycle experiment, the photothermal conversion efficiency of SPNH was measured to be 31.9% under 808 nm laser irradiation.54 The photodynamic effect and sonodynamic effect of SPNH were then evaluated by using singlet oxygen sensor green (SOSG) as a 1O2 probe. The fluorescence intensities of SOSG in the aqueous solution containing SPNH showed a gradual increase under both laser and US irradiation (Fig. S4, ESI). After 10 min, the fluorescence intensities of SOSG increased 9.3 times for both laser and US irradiation, which was higher than those in the case of sole US (3.0 times) and laser (7.2 times) irradiation (Fig. 2i). The results illustrated that the amount of 1O2 produced by photodynamic and sonodynamic combinational effects was higher than that of the sole photodynamic effect or sonodynamic effect.

Evaluation of in vitro therapeutic efficacy and ROS generation

The in vitro cytotoxicity of SPNH was determined using the CCK-8 assay. 4T1 cells were incubated with SPNH at various concentrations (0, 6, 12, 25, 50 and 100 μg mL−1) for 24 h, and the results showed that the cell viability remained higher than 90.0% (Fig. 3a), which suggested that SPNH had low cytotoxicity. The in vitro therapeutic efficacy of SPNH at a concentration of 50 μg mL−1 was studied. The viability of 4T1 cells in the SPNH + US + laser group was only 9.6% due to the PDT, PTT and SDT combinational therapy (Fig. 3b). In addition, the 4T1 cell viability in SPNH + US and SPNH + laser groups was 68.2% and 38.1%, respectively. In order to distinguish the toxic effects of PDT and PTT in the SPNH + laser group, NAC was used to eliminate the generation of ROS for only the PTT effect or the temperatures were controlled below 43 °C for the sole PDT effect. Both SPNH + laser + NAC and SPNH + laser (<43 °C) groups showed certain therapeutic effects, and the cell viability was 73.4% for the SPNH + laser + NAC group and 60.8% for the SPNH + laser (<43 °C) group (Fig. S5, ESI). These results demonstrated that the cell viability in the SPNH + US + laser group decreased much more significantly compared to that in SPNH + US and SPNH + laser groups, revealing the better in vitro therapeutic efficacy of PDT–PTT–SDT combinational therapy.
image file: d4tb00834k-f3.tif
Fig. 3 Evaluation of in vitro therapeutic efficacy and ROS generation. (a) Cell viability analysis of 4T1 cells after 24 h incubation with different concentrations of SPNH (n = 5). (b) Cell viability of 4T1 cancer cells in each group. (c) Fluorescence images of stained 4T1 cells to show live/apoptotic cells. (d) Quantitative analysis of live and apoptotic cell ratios after various treatments. (e) Fluorescence images of intracellular ROS generation after various treatments. (f) The mean fluorescence intensities of ROS stained signals in treated 4T1 cells.

The in vivo therapeutic efficacy was also verified using Calcein-AM/PI staining to show the live and apoptotic cells.55 The apoptotic cells were remarkably observed in SPNH + laser, SPNH + US and SPNH + US + laser groups (red fluorescence signals), while nearly no apoptotic cells could be found in the other groups (Fig. 3c). More importantly, the signals of red fluorescence of apoptotic cells in the SPNH + US + laser group were much stronger compared to those in SPNH + laser and SPNH + US groups. Through quantitative analysis of the green/red fluorescence intensity, we found that the proportions of apoptotic cells in the SPNH + US + laser group were as high as 83.8%, while those were 48.4% and 38.9% in SPNH + laser and SPNH + US groups, respectively (Fig. 3d). These results revealed that the SPNH + US + laser group had the highest efficacy in killing cancer cells.

Intracellular ROS production after different treatments was also detected using H2DCFDA as the indicator. Conspicuous ROS generation occurred in SPNH + US + laser, SPNH + laser, and SPNH + US groups, while the ROS levels in PBS, SPNH and PBS + laser + US groups were negligible (Fig. 3e). The quantitative analysis showed that the fluorescence intensity of ROS generation in the SPNH + US + laser group was approximately 1.5- and 1.6-fold higher than those in SPNH + laser and SPNH + US groups, respectively (Fig. 3f). These results confirmed the abundant ROS generation in the SPNH + US + laser group, which should be due to the SDT and PDT effects.

In vivo fluorescence and SPECT imaging

Mouse models with subcutaneous 4T1 tumors were utilized for evaluating in vivo fluorescence and SPECT imaging. After intravenous injection of SPNH, the fluorescence signals of tumor sites gradually increased and reached the maximum value in 12 h (Fig. 4a). At 12 h post-injection, the fluorescence intensity of tumors was nearly 3000 times higher compared to the initial fluorescence intensity (Fig. 4b). These results suggested the effective diagnosis of 4T1 tumors using fluorescence imaging. In addition, manifest fluorescence signals could be observed in isolated livers and tumors, which were barely detected in the heart, spleen, lungs and kidneys (Fig. 4c). The quantitative data illustrated that the fluorescence intensity of the liver and tumors was much higher than those of the other organs (Fig. 4d).
image file: d4tb00834k-f4.tif
Fig. 4 In vivo fluorescence and SPECT imaging. (a) The fluorescence images of tumor-bearing mice at different time points after SPNH injection through the tail vein. (b) Intensity of fluorescence signals in the tumor areas of mice (n = 3). (c) The fluorescence images of the tumors and main organs of mice at 27 h after intravenous injection of SPNH. (d) Quantitative analysis of fluorescence intensity in tumors and major organs (n = 3). (e) The SPECT images of 4T1 tumor-bearing mice post intravenous injection of SPNH (131I, 1 mCi). (f) The quantitative analysis of the relative signal intensity of tumor areas. (g) The quantitative analysis of the signal intensity of major organs and tumors.

As for SPECT imaging, the signals in tumor sites could be clearly detected at 2 h post-injection of SPNH and the signal gradually increased until 12 h (Fig. 4e). The maximum signal value at 12 h post-injection increased by around 2.4-fold compared to the initial value (Fig. 4f). The ex vivo SPECT imaging illustrated that SPNH inclined to accumulate in the liver and tumors because the signals could be observed in the liver and tumors (Fig. S6, ESI). The signal intensities of the liver and tumors were overall higher than those in the heart, spleen, lungs and kidneys (Fig. 4g). These results illustrated that SPNH could be used for SPECT imaging of tumors, and the accumulation effect of SPNH observed via fluorescence and SPECT imaging was similar. After intravenous injection, SPNH exhibited good pharmacokinetic properties as they still could be detected in the bloodstream for 84 h (Fig. S7, ESI).56

In vivo antitumor efficacy evaluation

After treatment, the tumors were used to confirm the ROS production using a fluorescence probe. The ROS signal (green fluorescence) was observed only in SPNH + US, SPNH + laser, and SPNH + US + laser groups (Fig. S8, ESI), confirming the generation of 1O2. Compared with the PBS group, the ROS green fluorescence intensity was increased 7.8, 16.1 and 28.5 times in SPNH + US, SPNH + laser, and SPNH + US + laser groups, respectively.

Because the maximum accumulation of SPNH at the tumor sites occurred at 12 h post-injection, the tumors were irradiated by a laser and/or US at this time point to enable therapeutic actions. Due to the photothermal effect of PCPDTBT, the in vivo PTT effect of SPNH was investigated.57,58 Two groups of mice with 4T1 tumors were randomly assigned to PBS and SPNH groups. The real-time temperatures of the tumor areas under 808 nm laser irradiation (1.0 W cm−2) were monitored. The tumor temperature in the SPNH + laser group gradually increased under laser irradiation, while in the PBS + laser group it did not have obvious elevation (Fig. 5a), which verified the in vivo PTT effect. The temperature curves of tumor sites illustrated that the temperature in the SPNH + laser group could obviously increase to 55 °C after 300 s of laser irradiation (Fig. 5b).


image file: d4tb00834k-f5.tif
Fig. 5 Evaluation of in vivo antitumor efficacy. (a) Thermal imaging of tumor-bearing mice under 808 nm laser irradiation for 300 s (1 W cm−2) at 12 h after systemic administration of PBS or SPNH (250 μg mL−1, 200 μL). (b) Mean temperatures of tumors at 0, 60, 120, 180, 240 and 300 s under 808 nm laser irradiation (n = 3). (c) Tumor volumes from the six groups after injection of PBS or SPNH at a concentration of 250 μg mL−1 (200 μL) without or with US, laser and US plus laser irradiation (n = 5). (d) The tumor photographs from mice in various treatment groups (n = 5). (e) The tumor weights in various treatment groups (n = 5). (f) The tumor inhibition efficacy in various treatment groups (n = 5). (g) Survival curves of mice after treatment (n = 5). (h) H&E staining images of tumors in various treatment groups.

The mouse models with 4T1 tumors were used to evaluate the in vivo antitumor efficacy of SPNH by monitoring tumor volumes and measuring tumor weights.59,60 After 14 days of observation, the tumor volumes in the SPNH + laser + US group were 3.8-, 3.4-, 3.5-, 2.5- and 1.9-fold lower than those in PBS, SPNH, PBS + US + laser, SPNH + US and SPNH + laser groups, respectively (Fig. 5c). Tumor photographs showed that the sizes of all five tumors in the SPNH + laser group were smaller than those in the SPNH + US group (Fig. 5d). This should be because the nanoparticles mediated both PTT and PDT in the SPNH + laser group to ablate the tumor cells, while they only enabled SDT in the SPNH + US group. In the SPNH + US + laser group, all five tumors showed the smallest sizes. Additionally, the mean tumor weight in the SPNH + US + laser group was only 0.06 g, which was 6.3- and 3.9-fold lower compared to those in the SPNH + US and SPNH + laser groups, respectively (Fig. 5e). Tumors in the SPNH + US + laser group showed a conspicuous tumor inhibition rate (91.5%), which was higher than those in SPNH + US (46.4%) and SPNH + laser (66.7%) groups (Fig. 5f). These results verified the highest in vivo antitumor efficacy in the SPNH + US + laser group via the PDT–PTT–SDT combinational effect.

Next, the mouse survival rates were recorded for 24 days after different treatments. All mice in PBS and PBS + US + laser groups died, while the survival rate of mice in the SPNH + US + laser group remained 80% (Fig. 5g). The H&E staining results further indicated that tumor cell necrosis/apoptosis could be observed in SPNH + US, SPNH + laser and SPNH + US + laser groups, but the tumors in the SPNH + US + laser group displayed the most manifest cell necrosis/apoptosis (Fig. 5h).

Evaluation of the antimetastatic effect

The metastasis inhibition of breast cancer is pivotal to ensure ideal therapeutic effects.61–63 The in vivo antimetastatic effect of SPNH was evaluated using H&E staining after treatment for 22 days. The combinational therapy in the SPNH + US + laser group conspicuously suppressed tumor metastasis in the lungs because nearly no tumor nodules were observed in H&E staining images (Fig. 6a). However, the tumor nodules could be found in all other groups. The areas of metastatic nodules in the lung tissues were 0.43, 0.35 and 0.43 cm2 for PBS, SPNH and PBS + US + laser groups, respectively, while the area of metastatic nodules was only 0.02 cm2 for the SPNH + US + laser group (Fig. 6b). Obvious metastatic tumor nodules were also found in the liver tissues of PBS, SPNH and PBS + US + laser groups, while very small numbers of tumor nodules could be observed in the SPNH + US + laser group (Fig. 6c). The area of liver metastatic tumor nodules for the SPNH + US + laser group was as low as 0.08 cm2 (Fig. 6d). These results revealed that the treatment in the SPNH + US + laser group could remarkably restrain metastases in lung and liver tissues with the highest antimetastatic effect.
image file: d4tb00834k-f6.tif
Fig. 6 Evaluation of the in vivo antimetastatic effect. (a) Representative H&E staining images of the lungs for various groups (the black arrow shows the location of the metastasis tumor nodules; the black dotted line represents the metastasis tumor nodules). (b) The areas of metastasis tumor nodules in isolated lung tissues in various groups (n = 5). (c) H&E staining images of the livers after treatment (blue arrows indicate the location of metastasis tumor nodules). (d) The areas of tumor metastasis nodules in the liver tissues of various groups (n = 5).

After various treatments, there were no manifest changes in the body weights of mice (Fig. S9, ESI). As observed in the H&E staining images, the other isolated organs (heart, spleen and kidneys) in the SPNH + US + laser group showed normality that was consistent with those in the PBS group (Fig. S10, ESI). These results verified the good biosafety of SPNH in living subjects.

Conclusions

We herein have designed radioactive hybrid semiconducting polymer nanoparticles (SPNH) for imaging-guided tri-modal therapy of breast cancer. SPNH containing two semiconducting polymers and labeled 131I showed good radiolabeling stability and photodynamic, photothermal and sonodynamic effects, allowing for fluorescence and SPECT imaging and combinational therapy. The formed SPNH had a small size of around 60 nm, enabling their effective accumulation in subcutaneous 4T1 tumors. Through in vivo fluorescence and SPECT imaging, subcutaneous 4T1 tumors could be clearly diagnosed after injection of SPNH. Under laser and US irradiation, SPNH mediated the PDT–PTT–SDT combination effect by generating ROS and heat, which enabled a much better antitumor efficacy in inhibiting tumor growth and metastasis compared to sole SDT and combinational PDT–PTT. In consideration of the simple preparation processes, other antitumor drugs can be integrated into SPNH to further improve the antitumor efficacy. Thus, this research can provide a theranostic nanomedicine for imaging-guided effective treatment of tumors.

Author contributions

J. Gu, D. Cheng and H. Li performed all experimental works and data analyses and wrote the original draft. T. Yu, Z. Zhang and Y. Liu analysed the data. X. Wang, X. Lu and J. Li supervised the project and revised the manuscript.

Conflicts of interest

The authors declare no conflicts of interest.

Acknowledgements

This study was financially supported by the Science and Technology Commission of Shanghai Municipality (22490760700, 22ZR1401100 and 20DZ2254900), the Foundation of Jiangsu Shuangchuang Talents Plan (SC0001) and Jiangsu Provincial Health Commission General Project (H2023055).

References

  1. S. Wang, K. Li, Y. Chen, H. Chen, M. Ma, J. Feng, Q. Zhao and J. Shi, Biomaterials, 2015, 39, 206–217 CrossRef CAS PubMed.
  2. X. Sun, P. K. Chintakunta, A. A. Badachhape, R. Bhavane, H.-J. Lee, D. S. Yang, Z. Starosolski, K. B. Ghaghada, P. G. Vekilov, A. V. Annapragada and E. A. Tanifum, Adv. Sci., 2023, 10, 2206435 CrossRef CAS PubMed.
  3. G. Song, C. Ji, C. Liang, X. Song, X. Yi, Z. Dong, K. Yang and Z. Liu, Biomaterials, 2017, 112, 257–263 CrossRef CAS PubMed.
  4. D. Wang, T. Nie, C. Huang, Z. Chen, X. Ma, W. Fang, Y. Huang, L. Luo and Z. Xiao, Small, 2022, 18, 2203227 CrossRef CAS PubMed.
  5. L. Ge, C. Qiao, Y. Tang, X. Zhang and X. Jiang, Nano Lett., 2021, 21, 3218–3224 CrossRef CAS PubMed.
  6. I. Kola and J. Landis, Nat. Rev. Drug Discov., 2004, 3, 711–716 CrossRef CAS PubMed.
  7. S. Gao, T. Li, Y. Guo, C. Sun, B. Xianyu and H. Xu, Adv. Mater., 2020, 32, 1907568 CrossRef CAS PubMed.
  8. N. Ogrinc, P. Saudemont, Z. Takats, M. Salzet and I. Fournier, Trends Mol. Med., 2021, 27, 602–615 CrossRef CAS PubMed.
  9. X. Guan, L. Sun, Y. Shen, F. Jin, X. Bo, C. Zhu, X. Han, X. Li, Y. Chen, H. Xu and W. Yue, Nat. Commun., 2022, 13, 2834 CrossRef CAS PubMed.
  10. C. Jian, T. Wu, L. Wang, C. Gao, Z. Fu, Q. Zhang and C. Shi, Small, 2024, 2309279 CrossRef PubMed.
  11. S. Fu, Y. Li, L. Shen, Y. Chen, J. Lu, Y. Ran, Y. Zhao, H. Tang, L. Tan, Q. Lin and Y. Hao, Small, 2024, 2309537 CrossRef PubMed.
  12. T. Liu, X. Si, L. Liu, S. Ma, Z. Huang, Y. Zhang, W. Song, Y. Zhang and X. Chen, ACS Nano, 2024, 18, 3087–3100 CrossRef CAS PubMed.
  13. S. S. Lucky, Y. Soo Kc, F. Zhang and Y. Zhang, Chem. Rev., 2015, 115, 1990–2042 CrossRef CAS PubMed.
  14. S. Geng, P. Guo, J. Wang, Y. Zhang, Y. Shi, X. Li, M. Cao, Y. Song, H. Zhang, Z. Zhang, K. Zhang, H. Song, J. Shi and J. Liu, Adv. Mater., 2024, 2307923 CrossRef CAS PubMed.
  15. F. Liu, C. Guo, X. Li, Y. Li, S. Xu, T. D. James and L. Wang, Nano Today, 2024, 54, 102116 CrossRef CAS.
  16. D. Cheng, J. Gong, P. Wang, J. Zhu, N. Yu, J. Zhao, Q. Zhang and J. Li, J. Mater. Chem. B, 2021, 9, 9316–9323 RSC.
  17. M. Santha Moorthy, G. Hoang, B. Subramanian, N. Q. Bui, M. Panchanathan, S. Mondal, V. P. Thi Tuong, H. Kim and J. Oh, J. Mater. Chem. B, 2018, 6, 5220–5233 RSC.
  18. Y. Wang, F. Zhang, H. Lin and F. Qu, ACS Appl. Mater., 2019, 11, 43964–43975 CrossRef CAS PubMed.
  19. X. Zhang, J. Cui, J. Liu, X. Chen, M. Chen and J. Wang, J. Mater. Chem. B, 2023, 11, 9516–9524 RSC.
  20. R. R. Allison, G. H. Downie, R. Cuenca, X.-H. Hu, C. J. H. Childs and C. H. Sibata, Photodiagn. Photodyn. Ther., 2004, 1, 27–42 CrossRef CAS PubMed.
  21. Y. Yang, Z. Zeng, E. Almatrafi, D. Huang, C. Zhang, W. Xiong, M. Cheng, C. Zhou, W. Wang, B. Song, X. Tang, G. Zeng, R. Xiao and Z. Li, Coord. Chem. Rev., 2022, 458, 214427 CrossRef CAS.
  22. H. Yang, R. Liu, Y. Xu, L. Qian and Z. Dai, Nano-Micro Lett., 2021, 13, 35 CrossRef PubMed.
  23. X.-X. Lu, C. Xue, J.-H. Dong, Y.-Z. Zhang and F. Gao, J. Mater. Chem. B, 2024, 12, 3209–3225 RSC.
  24. M. Shi, X. Liu, W. Pan, N. Li and B. Tang, J. Mater. Chem. B, 2023, 11, 6478–6490 RSC.
  25. X. Wang, X. Wang, Q. Yue, H. Xu, X. Zhong, L. Sun, G. Li, Y. Gong, N. Yang, Z. Wang, Z. Liu and L. Cheng, Nano Today, 2021, 39, 101170 CrossRef CAS.
  26. B. Liu, F. Du, Z. Feng, X. Xiang, R. Guo, L. Ma, B. Zhu and L. Qiu, J. Mater. Chem. B, 2024, 12, 3636–3658 RSC.
  27. F. Wang, Y. Fan, Y. Liu, X. Lou, L. Sutrisno, S. Peng and J. Li, Exploration, 2024, 20230100 CrossRef.
  28. Y. Liu, Y. Wang, W. Zhen, Y. Wang, S. Zhang, Y. Zhao, S. Song, Z. Wu and H. Zhang, Biomaterials, 2020, 251, 120075 CrossRef CAS PubMed.
  29. S. Li, F. Yang, Y. Wang, L. Jia and X. Hou, Mater. Horiz., 2023, 10, 5734–5752 RSC.
  30. Y. Jiang, X. Zhao, J. Huang, J. Li, P. K. Upputuri, H. Sun, X. Han, M. Pramanik, Y. Miao, H. Duan, K. Pu and R. Zhang, Nat. Commun., 2020, 11, 1857 CrossRef CAS PubMed.
  31. Z. Xiao, Q. Chen, Y. Yang, S. Tu, B. Wang, Y. Qiu, Y. Jiang, Q. Huang and K. Ai, Chem. Eng. J., 2022, 449, 137889 CrossRef CAS.
  32. F. Gong, L. Cheng, N. Yang, O. Betzer, L. Feng, Q. Zhou, Y. Li, R. Chen, R. Popovtzer and Z. Liu, Adv. Mater., 2019, 31, 1900730 CrossRef PubMed.
  33. X. Zhang, L. An, Q. Tian, J. Lin and S. Yang, J. Mater. Chem. B, 2020, 8, 4738–4747 RSC.
  34. Q. Sun, F. Liu, Z. Wen, J. Xia, H. Li, Y. Xu and S. Sun, J. Mater. Chem. B, 2022, 10, 1369–1377 RSC.
  35. L. Lei, W. Dai, J. Man, H. Hu, Q. Jin, B. Zhang and Z. Tang, J. Nanobiotechnol., 2023, 21, 482 CrossRef CAS PubMed.
  36. M.-M. Pan, P. Li, Y.-P. Yu, M. Jiang, X. Yang, P. Zhang, J. Nie, J. Hu, X. Yu and L. Xu, Adv. Healthcare Mater., 2023, 12, 2300821 CrossRef CAS PubMed.
  37. Y. Wang, G. Song, S. Liao, Q. Qin, Y. Zhao, L. Shi, K. Guan, X. Gong, P. Wang, X. Yin, Q. Chen and X.-B. Zhang, Angew. Chem., Int. Ed., 2021, 60, 19779–19789 CrossRef CAS PubMed.
  38. P. R. Neumann, F. Erdmann, J. Holthof, G. Hädrich, M. Green, J. Rao and L. A. Dailey, Adv. Healthcare Mater., 2021, 10, 2001089 CrossRef CAS PubMed.
  39. J. Feng, C. Zhu, H. Yuan, S. Liu, F. Lv and S. Wang, Chem. Soc. Rev., 2013, 42, 6620 RSC.
  40. C. Qian, J. Yu, Y. Chen, Q. Hu, X. Xiao, W. Sun, C. Wang, P. Feng, Q.-D. Shen and Z. Gu, Adv. Mater., 2016, 28, 3313–3320 CrossRef CAS PubMed.
  41. N. Yu, M. Ding, F. Wang, J. Zhou, X. Shi, R. Cai and J. Li, Nano Today, 2022, 46, 101600 CrossRef CAS.
  42. Y. Pan, J. Li, X. Wang, Y. Sun, J. Li, B. Wang and K. Zhang, Adv. Funct. Mater., 2022, 32, 2110207 CrossRef CAS.
  43. Q. Miao, C. Xie, X. Zhen, Y. Lyu, H. Duan, X. Liu, J. V. Jokerst and K. Pu, Nat. Biotechnol., 2017, 35, 1102–1110 CrossRef CAS PubMed.
  44. S. Liao, Y. Wang, Z. Li, Y. Zhang, X. Yin, S. Huan, X.-B. Zhang, S. Liu and G. Song, Theranostics, 2022, 12, 6883–6897 CrossRef CAS PubMed.
  45. M. Ding, Y. Zhang, N. Yu, J. Zhou, L. Zhu, X. Wang and J. Li, Adv. Mater., 2023, 35, 2302508 CrossRef CAS PubMed.
  46. W. Chen, Y. Zhang, Q. Li, Y. Jiang, H. Zhou, Y. Liu, Q. Miao and M. Gao, J. Am. Chem. Soc., 2022, 144, 6719–6726 CrossRef CAS PubMed.
  47. N. Yu, M. Li, Y. Zhang, F. Wang, X. Yu, R. Cai and J. Li, Nano Today, 2023, 52, 101944 CrossRef CAS.
  48. D. Cheng, J. He, C. Zhang, Y. Zhang, N. Yu, R. Cai, X. Wang and J. Li, ACS Appl. Nano Mater., 2023, 6, 421–430 CrossRef CAS.
  49. X. Chen, D. Cheng, N. Yu, J. Feng, J. Li and L. Lin, J. Mater. Chem. B, 2024, 12, 1296–1306 RSC.
  50. L. S. Gomez-Aparicio, J. Bernáldez-Sarabia, T. A. Camacho-Villegas, P. H. Lugo-Fabres, N. E. Díaz-Martínez, E. Padilla-Camberos, A. Licea-Navarro and A. B. Castro-Ceseña, Biomater. Sci., 2021, 9, 726–744 RSC.
  51. H. Huang, C. Dong, M. Chang, L. Ding, L. Chen, W. Feng and Y. Chen, Exploration, 2021, 1, 50–60 CrossRef PubMed.
  52. J. Wang, M. Liu, X. Zhang, X. Wang, M. Xiong and D. Luo, Exploration, 2024, 20230027 CrossRef.
  53. P. Ge, B. Niu, Y. Wu, W. Xu, M. Li, H. Sun, H. Zhou, X. Zhang and J. Xie, Chem. Eng. J, 2020, 383, 123228 CrossRef CAS.
  54. Y. Lyu, C. Xie, S. A. Chechetka, E. Miyako and K. Pu, J. Am. Chem. Soc., 2016, 138, 9049–9052 CrossRef CAS PubMed.
  55. X. Zhen, N. K. Pandey, E. Amador, W. Hu, B. Liu, W. Nong, W. Chen and L. Huang, Mater. Today Phys., 2022, 27, 100838 CrossRef CAS.
  56. Y. Li, J. Lin, Y. He, K. Wang, C. Huang, R. Zhang and X. Liu, Exploration, 2024, 4, 20230054 CrossRef.
  57. Z. Liu, C. Hong, C. Pan, Y. Sun, Y. Lv, Y. Wei, X. Wang, W. Zang, Q. Mao, X. Deng, P. Wang, W. Zhu, T. Chen, M. Wu, J. Li and A. Wu, ACS Mater. Lett., 2024, 1593–1605 CrossRef CAS.
  58. T. F. Abelha, P. R. Neumann, J. Holthof, C. A. Dreiss, C. Alexander, M. Green and L. A. Dailey, J. Mater. Chem. B, 2019, 7, 5115–5124 RSC.
  59. C. Song, X. Wu, J. Wang, R. Liu and Y. Zhao, Nano Today, 2023, 51, 101913 CrossRef CAS.
  60. Z. Wu, H. Chen, H. Zhang, L. Ye, J. Ke, Y. Liu, P. Sun and M. Hong, Nano Today, 2024, 55, 102214 CrossRef CAS.
  61. H. Hu, J. Zhao, K. Ma, J. Wang, X. Wang, T. Mao, C. Xiang, H. Luo, Y. Cheng, M. Yu, Y. Qin, K. Yang, Q. Li, Y. Sun and S. Wang, J. Controlled Release, 2023, 359, 188–205 CrossRef CAS PubMed.
  62. W. Zeng, Y. Xu, W. Yang, K. Liu, K. Bian and B. Zhang, Adv. Healthcare Mater., 2020, 9, 2000560 CrossRef CAS PubMed.
  63. L. Li, M. Zhen, H. Wang, Z. Sun, X. Cao, J. Li, S. Liu, Z. Zhao, C. Zhou, C. Wang and C. Bai, Nano Today, 2023, 48, 101702 CrossRef CAS.

Footnotes

Electronic supplementary information (ESI) available. See DOI: https://doi.org/10.1039/d4tb00834k
These authors contributed equally to this work.

This journal is © The Royal Society of Chemistry 2024