Huihai
Zhong
ab,
Yuefei
Fang
bd,
Pengfei
Zhao
e,
Aihua
Wu
*f,
Guohui
Nie
*a,
Yongzhuo
Huang
*bc and
Bin
Zhang
*a
aShenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518035, China. E-mail: nieguohui@email.szu.edu.cn; binzhang@email.szu.edu.cn
bZhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China. E-mail: yzhuang@simm.ac.cn
cNMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
dState Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
eCenter of Clinical Pharmacology, the Second Affiliated Hospital Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, China
fDepartment of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3rd East Qingchun Road, Hangzhou 310016, China. E-mail: wuaihua2016@sina.com
First published on 31st July 2025
Cancer treatment faces challenges including drug resistance to intrinsic apoptosis pathways and the persistence of cancer stem cells (CSCs). The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces extrinsic apoptosis in tumor cells by binding to death receptors while sparing normal cells. TRAIL-based gene therapy has emerged as a promising solution to address the short half-life limitation of recombinant TRAIL proteins. To further enhance the therapeutic efficacy, combination strategies with TRAIL sensitizer salinomycin (Sali) appear to be particularly effective. Sali not only upregulates death receptor expression and reactive oxygen species (ROS) levels but also modulates CSC-related gene expression, contributing to its multifaceted antitumor activity. In this study, a micelle system was developed for co-delivery of TRAIL plasmids (pTRAIL) and Sali to tumor sites. γ-Polyglutamic acid (γ-PGA)-coating improved the in vivo stability profile of the co-delivery nanocomposites. The resulting co-delivery system (γ-PGA/Sali@P–S/pTRAIL) effectively induced apoptosis and suppressed CSC-associated gene expression in cervical cancer cells. Both in vitro and in vivo evaluations demonstrated the system's potent antitumor efficacy, highlighting its potential as a promising strategy for cervical cancer therapy.
Induction of tumor cell apoptosis is a key therapeutic mechanism of most treatments, including chemotherapy and radiotherapy. However, conventional treatments face two major limitations: systemic toxicity and resistance to intrinsic apoptosis pathways.7,8 Therefore, there is an urgent need to identify safer and more effective therapeutic approaches. The tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF family, selectively induces extrinsic apoptosis in tumor cells while sparing normal cells.9,10 The TRAIL exerts its effects by binding to death receptors (DRs), such as DR5, which are mainly expressed on the tumor cell surface, thereby activating downstream apoptotic pathways.11 Thus, effective TRAIL therapy relies on two critical factors: efficient delivery of the TRAIL and sufficient DR expression. While the development of TRAIL receptor agonists remains challenging, current clinical trials primarily focus on the delivery of recombinant TRAIL proteins,12 which are limited by their short half-life and ineffective delivery.13,14 With the advancement of interdisciplinary fields, particularly materials science and nanobiotechnology, combination therapies have provided targeted approaches for cancer treatment. Nanoparticle-mediated TRAIL gene delivery represents a promising alternative, with non-viral DNA vectors offering a safer and more convenient option.15–17 Various cationic polymers have been developed, including polyethyleneimine (PEI), poly(β-amino ester)s, poly[(2-dimethylamino)ethyl methacrylate], and dendrimers.18,19 PEI is one of the most widely used polymeric vectors for gene delivery due to its ability to facilitate endosomal escape through the “proton sponge” effect.20,21 For instance, PEI-grafted cationic liposomes demonstrate potent transfection efficacy.22 However, tumor cells can evade TRAIL-induced apoptosis by downregulating DR expression.23 Polyether ionophore antibiotics have been reported to sensitize TRAIL therapy by upregulating DR5 and inhibiting FLIP.24 Therefore, combining TRAIL delivery with effective sensitization strategies may enhance its antitumor efficacy and help overcome resistance, thereby improving therapeutic outcomes.
In addition, cancer stem cells (CSCs) are another major contributor to poor prognosis and are closely associated with therapy resistance.25 CSCs represent a small subpopulation of tumor cells with self-renewal and differentiation capabilities, capable of generating heterogeneous tumor cell populations.26,27 These cells play a pivotal role in tumor initiation, progression, recurrence, metastasis, and drug resistance.28,29 Thus, targeting CSCs in cancer therapy is of great clinical significance.30,31 Although our understanding of CSCs is still evolving, current evidence shows that their regulation involves multiple signaling pathways. The activation of stemness-related genes and oncogenes has been shown to transform original cells into tumorigenic CSCs.32 Targeted inhibition of aberrant Wnt/β-catenin signaling and c-Myc activation can effectively suppress proliferation and induce apoptosis in CSCs.33 Moreover, additional pathways, such as STAT3 signaling, contribute significantly to CSC maintenance and function.34,35 Precise modulation of these critical signaling networks offers a promising therapeutic strategy to impair CSC viability and potentially inhibit tumor progression and metastasis. These signaling pathways are also closely associated with TRAIL-based therapies.36–38 Salinomycin (Sali), a polyether ionophore antibiotic, has been reported as one of the most potent inhibitors of CSC fate regulators.30,39 The anti-CSC mechanisms of Sali involve the modulation of oncogenes and stemness-associated factors.40 Although Sali may serve as an effective sensitizer for TRAIL-based cancer therapy,41 the mechanistic basis underlying their combinatorial action remains incompletely defined.
Based on these considerations, we constructed a PEI-based amphiphilic material for the co-delivery of Sali and TRAIL plasmids (pTRAIL) (Scheme 1). Surface modification with poly-γ-glutamic acid (γ-PGA) reduced the zeta potential and improved stability for in vivo delivery.42–44 This study focuses on developing a dual-targeted therapeutic strategy that combines enhanced TRAIL-mediated apoptosis with the inhibition of CSC-related signaling pathways. While the expressed TRAIL activates the extrinsic apoptotic pathway in HeLa cells, Sali effectively sensitizes this process by upregulating DR5 expression. Beyond death receptor modulation, Sali exerts complementary anticancer effects by elevating reactive oxygen species (ROS) levels and coordinately downregulating CSC-associated signaling pathways, including Wnt/β-catenin, STAT3, and c-Myc, thereby synergizing with TRAIL therapy. Our co-delivery nanoplatform demonstrates remarkable therapeutic efficacy in murine HeLa xenograft models, achieving approximately 80% tumor growth inhibition.
![]() | ||
| Scheme 1 Schematic illustration of co-delivery of the pTRAIL and salinomycin for inducing apoptosis of cancer cells and CSC-associated gene downregulation. | ||
The gel retardation of the P–S/pDNA nanocomplexes was examined. In brief, the nanocomplexes with different mass ratios were freshly prepared before use as mentioned above. The electrophoresis experiment was performed in 1% agarose gel containing 0.1 mL mL−1 Gelred at room temperature in 1 × TAE buffer at 80 V for 45 min. Gel imaging was visualized using a chemiDOX system (Bio-Rad, USA). Particle sizes and zeta potentials were evaluated using a Zetasizer Nano ZS (Malvern Instruments Ltd, UK).
:
4 (v/v) ratio and reacted at 72 °C for 40 min, generating a salinomycin–vanillin derivative with characteristic absorption at 526 nm. 1 mg mL−1 salinomycin stock solution (in absolute ethanol) was diluted to prepare standard solutions at concentrations of 0, 6.25, 12.5, 25, 50, 100, and 200 μg mL−1. For the standard curve determination, 600 μL of each salinomycin standard solution was mixed with 400 μL of vanillin derivatization solution, followed by incubation at a constant temperature of 72 °C in a water bath for 40 minutes. After rapid cooling to room temperature, the absorbance of the reaction mixture was measured at 526 nm using a multimode microplate reader. For quantification of encapsulated salinomycin in Sali@P–S nanomicelles, 200 μL of the nanoparticle suspension was mixed with 800 μL of absolute ethanol, vortexed for 30 seconds, and subsequently sonicated for 10 minutes to ensure complete release of the encapsulated drug. From this solution, 600 μL was combined with 400 μL of vanillin solution and subjected to identical derivatization conditions (72 °C for 40 minutes in a water bath). Following rapid cooling to ambient temperature, the absorbance of the resulting derivative was similarly determined at 526 nm using the microplate reader. The drug-loading capacity was calculated using the following formula:| Drug-loading capacity (%) = weight of encapsulated drug/weight of nanoparticles × 100% |
:
5
:
1), utilized for cellular uptake analysis. The cells were seeded on glass bottom dishes. After being incubated with γ-PGA/C6@P–S/pDNA in different pH conditions, the cells were observed by CLSM. For flow cytometry analysis, the HeLa cells were seeded in a 24-well plate at 5 × 104 cells per well. After being incubated with γ-PGA/C6@P–S/pDNA in different pH conditions, the cells were detected by flow cytometry. The cells were stained with LysoTracker (Thermo) to investigate the uptake process of nanocomposites.
The cell viability (%) was calculated by the following formula:
| Cell viability (%) = (AE − AB)/(AC − AB) × 100% |
A E presented the absorbance of the samples with nanodrugs; AC was the absorbance of the samples without nanodrugs; AB was the absorbance of the blank control.
The HeLa cells were seeded at 2 × 105 cells per well in 6-well plates and incubated for 24 h. After the confluence reached about 70%, the nanodrugs were added to incubate for another 48 h. Afterwards, the cells were harvested and stained with annexin V-FITC and propidium iodide (PI) according to the manufacturer's protocol. Finally, the cells were detected by flow cytometry.
To assess the impact of Sali and TRAIL on CSC subpopulations, HeLa cells were seeded in 6-well plates at 2 × 105 cells per well and cultured for 24 h. Cells were then treated with either Sali alone or Sali + γ-PGA/Sali@P–S/pTRAIL nanocomplex (containing 2 μg pTRAIL per well) for an additional 24 h. Harvested cells were trypsinized, washed with PBS, and stained with anti-CD44-FITC and anti-CD133-APC antibodies for 30 min at 4 °C in the dark. After resuspension in PBS supplemented with 2% FBS, CD44+CD133+ dual-positive subpopulations were analyzed using a flow cytometer.
:
1) and γ-PGA/IR780@P–S/pTRAIL (12
:
5
:
1) were prepared for in vivo imaging study. After being injected intravenously into the tumor-bearing mice with IR780@P–S/pTRAIL and γ-PGA/IR780@P–S/pTRAIL, the mice were imaged by in vivo imaging system under isoflurane anesthesia at different time intervals (0.5 h, 1 h, 2 h, 6 h, 4 h, 8 h, 24 h, and 48 h). The mice were euthanized at the last time point, and the tumors were isolated for ex vivo imaging. The fluorescence intensities were analyzed by Carestream MI software.
| Tumor inhibition (%) = (PBS group tumor weight − Experimental group tumor weight)/PBS group tumor weight × 100% |
| Organ coefficient (%) = Organ weight/body weight × 100% |
The tissues were fixed with 4% paraformaldehyde for 24 h for immunohistochemical staining. The paraffin-embedded tissues were cut into 4 μm thick sections and dewaxed, and rehydrated. The epitope retrieval was performed by boiling in Tris/EDTA buffer (pH 9.0), followed by treated with 3% hydrogen peroxide and 5% BSA solution. The tissues were incubated with primary antibodies at 4 °C overnight. After washing three times with PBS, the HRP-conjugated secondary antibodies were incubated at room temperature for 1 h. Tissues were finally stained with a 3,3′-diaminobenzidine (DAB, brown) kit according to the manufacturer's instruction and counterstained with hematoxylin (blue). After dehydration and mounted with coverslips, tissues were observed for the proportion and brown intensity of the protein-positive cells under a bright-field microscope.
:
1 (w/w) ratio to form nanomicellar structures, designated as P–S nanomicelles. Agarose gel electrophoresis demonstrated that P–S could effectively condense plasmid DNA (pDNA) at a mass ratio of 2
:
1 (Fig. 1A and Fig. S4). When P–S was complexed with pEGFP at various mass ratios and transfected into HeLa cells under serum-free conditions (with PEI25K serving as the positive control), optimal GFP transfection efficiency was achieved at a 5
:
1 mass ratio, showing comparable performance to PEI25K-mediated transfection (Fig. 1B).
Based on the optimal ratio determined from transfection experiments, Sali@P–S was complexed with pTRAIL to prepare the nanocomposites Sali@P–S/pTRAIL. To shield the positive surface charge of the nanocomposites and improve in vivo applicability, the anionic polymer γ-PGA was coated onto the surface at a mass ratio of 2
:
1 (γ-PGA
:
Sali@P–S/pTRAIL), yielding γ-PGA/Sali@P–S/pTRAIL (12
:
5
:
1). Dynamic light scattering analysis revealed that Sali@P–S/pTRAIL exhibited a uniform particle size distribution centered at approximately 130 nm (Fig. 1C), which was corroborated by transmission electron microscopy (TEM) observations (Fig. 1D). Following γ-PGA coating, the particle size of γ-PGA/Sali@P–S/pTRAIL decreased to about 100 nm (Fig. 1C), likely due to charge compression induced by the negatively charged γ-PGA wrapping. TEM examination of γ-PGA/Sali@P–S/pTRAIL confirmed the formation of spherical nanoparticles (Fig. 1E). Zeta potential measurements demonstrated a complete surface charge reversal from highly positive (+45 mV) to negative (−30 mV) after γ-PGA coating (Fig. 1F), a modification expected to enhance colloidal stability for in vivo applications.
To evaluate the protective effect of γ-PGA coating on nanoparticles under serum-containing conditions, we investigated the P–S/pEGFP system at an optimal mass ratio of 5
:
1 (carrier to plasmid). Under these conditions, the γ-PGA-coated formulation demonstrated significantly improved performance. While naked P–S/pEGFP nanoparticles exhibited compromised transfection efficiency in the presence of serum, the γ-PGA-coated nanoparticles maintained enhanced transfection efficiency in HeLa cells (Fig. 2A). These results clearly indicate that the anionic γ-PGA coating effectively protects the cationic P–S/pEGFP nanocomposites from serum protein interference, thereby preserving their gene delivery capability. We next evaluated the drug loading capacity and stability of the nanocomposites. The P–S copolymer was first self-assembled with salinomycin (Sali) to form Sali@P–S nanomicelles, with the Sali encapsulation efficiency determined to be approximately 4.35% using the vanillin derivatization method (Fig. S5). Stability testing revealed that γ-PGA-coated nanoparticles maintained excellent colloidal stability in serum-containing media, whereas uncoated nanoparticles rapidly aggregated under serum conditions due to their strongly positive surface charge (Fig. 2B). We further investigated the pH-dependent drug release profile of the nanocomposites under different pH conditions (Fig. 2C). The free salinomycin control group released approximately 80% of the drug within 12 hours. In contrast, the γ-PGA/Sali@P–S nanoparticle system demonstrated pH-responsive release characteristics—sustained release under physiological pH (7.4) and accelerated release in acidic environments. This pH-sensitive behavior originates from the conformational changes of γ-PGA.45 At neutral pH, γ-PGA maintains a compact random coil structure that forms a dense protective layer around the micelles. Under acidic conditions, however, it undergoes a conformational transition to an extended helical structure, which facilitates its detachment from the micelle surface. This structural rearrangement promotes drug release from the hydrophobic core into the aqueous medium after the nanocomposites are internalized by tumor cells.
Lysosomal escape is crucial for effective intracellular drug delivery, as lysosomes contain hydrolytic enzymes that can degrade encapsulated plasmids if the nanocomposites remain trapped, thereby compromising their antitumor efficacy. To monitor this process, γ-PGA/C6@P–S/pTRAIL nanoparticles were used for uptake tracking, with lysosomes labeled using LysoTracker Red and nuclei stained with DAPI for cellular localization. When green fluorescent nanoparticles colocalize with red fluorescent lysosomes, merged yellow fluorescence is observed; isolated green fluorescence, in contrast, indicates successful lysosomal escape. Three time points (2, 4, and 6 hours) were examined to assess nanoparticle–lysosome colocalization. At 2 hours, nanoparticles were predominantly internalized and colocalized with lysosomes, confirming entry via the lysosomal pathway. By 4 hours, cellular uptake increased while lysosomal colocalization was still evident. However, by 6 hours, most nanoparticles had separated from lysosomes, with only minimal residual colocalization remaining (Fig. 3A). These results demonstrate that the formulated nanoparticles can effectively escape from lysosomes following endocytosis, thereby enabling their intended antitumor function. The temporal progression from initial uptake (2 h) to lysosomal accumulation (4 h), and subsequent escape (6 h), confirms the system's capability to overcome this critical intracellular barrier to gene delivery.
We further evaluated the cytotoxic effects of the nanoparticles on tumor cells, beginning with a comparative assessment of salinomycin monotherapy versus salinomycin/TRAIL combination therapy in HeLa cells. With the pTRAIL dosage fixed at 1 μg mL−1, the results demonstrated that salinomycin significantly potentiated TRAIL-induced cytotoxicity (Fig. 3B). Western blot analysis revealed that salinomycin upregulated DR5 expression in HeLa cells (Fig. 3C and Fig. S6A), and the combination treatment triggered caspase-3 cleavage, thereby inducing tumor cell apoptosis (Fig. 3D and Fig. S6B). The cytotoxic effects of the nanodelivery system were subsequently examined. After 24 hours of treatment, the dual-drug-loaded γ-PGA/Sali@P–S/pTRAIL nanocomposites exhibited superior tumor cell-killing efficacy, whereas blank nanoparticles showed minimal toxicity, indicating good biocompatibility (Fig. 3E). Furthermore, the nanoparticle system successfully mediated TRAIL protein expression in HeLa cells through efficient gene transfection (Fig. 3F and Fig. S6C).
Previous studies have reported that salinomycin exerts antitumor effects by inducing intracellular reactive oxygen species (ROS) generation. In our study, intracellular ROS levels were measured using a ROS detection kit after 24-hour treatment with either γ-PGA/Sali@P–S or γ-PGA/Sali@P–S/pTRAIL nanoparticles. The results demonstrated that both drug-loaded nanoparticles effectively induced ROS production, with the combination therapy group exhibiting significantly higher ROS levels (Fig. 3G and H). Consistent with prior findings on salinomycin's ability to downregulate cancer stem cell (CSC)-related pathways, our western blot analysis revealed significant suppression of Wnt/β-catenin signaling, accompanied by reduced expression of the downstream oncogene c-Myc (Fig. 3I and Fig. S6D) and the transcription factor STAT3 (Fig. 3J and Fig. S6), both of which are key regulators of tumor progression. Subsequent apoptosis assays showed that γ-PGA/Sali@P–S/pTRAIL induced the highest proportion of apoptotic cell death, confirming its superior therapeutic efficacy (Fig. 3K and L).
Given the established correlation between tumor cell stemness and metastatic potential, we performed Transwell migration assays demonstrating that Sali significantly inhibits cell migration at concentrations exceeding 200 nM (Fig. 4A). Subsequent flow cytometric analysis revealed a marked reduction in the CD44+CD133+ dual-positive fraction upon Sali treatment, indicating decreased CSCs prevalence. Notably, combinatorial treatment with TRAIL further diminished this subpopulation, suggesting that Sali sensitizes CSCs to TRAIL-mediated cytotoxicity (Fig. 4B and C). As downstream effectors co-regulated by Wnt/β-catenin signaling, transcription factor c-Myc and STAT3 frequently exhibit overexpression in advanced malignancies, where their synergistic activation drives tumor proliferation and metastasis. Thus, concurrent suppression of both molecules may hold therapeutic significance. Therefore, simultaneous suppression of these oncogenic factors may offer therapeutic benefit. Importantly, our data demonstrate that Sali concurrently inhibits the Wnt/β-catenin, c-Myc, and STAT3 pathways, while augmenting TRAIL-mediated CSC eradication.
Toxicity was preliminarily assessed through body weight monitoring and organ coefficient analysis at the study endpoint. No significant differences in body weight were observed among treatment groups (Fig. S8). However, organ coefficient analysis revealed splenomegaly in the Sali@P–S/pTRAIL group (Fig. S9), likely due to the unshielded positive charge of these nanoparticles, which may cause spleen-specific toxicity via electrostatic interactions with reticuloendothelial cells. This finding aligns with the known tendency of cationic nanoparticles to accumulate in and potentially damage splenic tissue. In contrast, γ-PGA-coated formulations exhibited no such adverse effects, confirming the importance of surface charge shielding in reducing organ-specific toxicity. Histopathological examination of major organs (heart, liver, spleen, lungs, kidneys) revealed no significant abnormalities across all treatment groups, indicating acceptable systemic tolerability (Fig. S10).
TUNEL assays conducted on tumor sections showed that the γ-PGA/Sali@P–S/pTRAIL group induced significantly higher levels of apoptosis compared to single-agent treatments, which induced only moderate apoptosis (Fig. 5E). Immunohistochemical analysis revealed differential TRAIL expression among the groups: while Sali@P–S/pTRAIL showed partial expression, both γ-PGA/P–S/pTRAIL and γ-PGA/Sali@P–S/pTRAIL groups demonstrated substantially higher TRAIL levels in tumor tissues (Fig. 5F). Evaluation of angiogenesis via CD31 staining showed the lowest expression levels in the γ-PGA/Sali@P–S/pTRAIL group (Fig. 5G), suggesting that extensive tumor cell apoptosis may suppress tumor vascularization. Similarly, Ki67 immunostaining indicated markedly reduced tumor cell proliferation in this group relative to all others (Fig. 5H). Collectively, these histopathological analyses confirm that the co-delivery nanocomposites enhance apoptosis, inhibit angiogenesis, and suppress proliferation through coordinated and synergistic mechanisms.
Despite promising results, translating such nanotherapeutics from bench to bedside remains a formidable challenge. TRAIL-based therapies, in particular, have encountered persistent limitations in clinical translation. Overcoming these barriers requires the convergence of multiple scientific disciplines. When such interdisciplinary synergies mature, they may ultimately unlock the full therapeutic potential of TRAIL-associated combination strategies.
| This journal is © The Royal Society of Chemistry 2025 |