- Research
- Open access
- Published:
Cisplatin-encapsulated TRAIL-engineered exosomes from human chorion-derived MSCs for targeted cervical cancer therapy
Stem Cell Research & Therapy volume 15, Article number: 396 (2024)
Abstract
Background
Cisplatin (DDP) is an efficacious and widely applied chemotherapeutic drug for cervical cancer patients who are diagnosed as metastatic and inoperable, or desiring fertility preservation. Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) selectively triggers cancer cells apoptosis by binding to cognate death receptors (DR4 and DR5). Mesenchymal stem cells-derived exosomes (MSCs-Exo) have been regarded as ideal drug carriers on account of their nanoscale, low toxicity, low immunogenicity, high stability, biodegradability, and abundant sources.
Methods
Human chorion-derived mesenchymal stem cells (hCD-MSCs) were isolated by adherent culture method. TRAIL-engineered hCD-MSCs (hCD-MSCsTRAIL) were constructed by lentivirus transfection, and its secreted Exo (hCD-MSCs-ExoTRAIL) were acquired by differential centrifugation and confirmed to overexpress TRAIL by western blotting. Next, nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) were fabricated by loading DDP into hCD-MSCs-ExoTRAIL via electroporation. The CCK-8 assay and flow cytometry were conducted to explore the proliferation and apoptosis of cervical cancer cells (SiHa and HeLa), respectively. Cervical cancer-bearing nude mice were constructed to examine the antitumor activity and biosafety of DDP & hCD-MSCs-ExoTRAIL in vivo.
Results
Compared with hCD-MSCs-Exo, hCD-MSCs-ExoTRAIL weakened proliferation and enhanced apoptosis of cervical cancer cells. DDP & hCD-MSCs-ExoTRAIL were proved to retard cervical cancer cell proliferation and propel cell apoptosis more effectively than DDP or hCD-MSCs-ExoTRAIL alone in vitro. In cervical cancer-bearing mice, DDP & hCD-MSCs-ExoTRAIL evidently hampered tumor growth, and its role in inducing apoptosis was mechanistically associated with JNK/p-c-Jun activation and survivin suppression. Moreover, DDP & hCD-MSCs-ExoTRAIL showed favorable biosafety in vivo.
Conclusions
DDP & hCD-MSCs-ExoTRAIL nanoparticles exhibited great promise for cervical cancer treatment as an Exo-based chemo-gene combinational therapy in clinical practice.
Graphical abstract

Introduction
Cervical cancer induces the fourth highest carcinoma-related mortality in females, presenting with about 661,021 newly diagnosed patients as well as 348,189 deaths in 2022 worldwide [1]. Current conventional treatments for cervical cancer consist of surgical resection, radiotherapy and chemotherapy [2]. Metastatic and inoperable recurrent cervical cancer holds serious threats to patients clinical prognosis [3], necessitating platinum-based chemotherapy as the preferentially recommended therapeutic regimen [4].
Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) initiates programmed cell death in extrinsic pathway, and optionally mediates apoptosis of malignant cells while renders no toxicity on normal cells [5]. TRAIL triggers the apoptosis of cancer cells via specific binding to cognate death receptors (DR4 and DR5) [6]. However, clinical application of soluble TRAIL protein variant is restricted by its short plasma half-life, low biostability, and limited bioavailability [7]. Delivery of TRAIL by nanoparticle-based carriers is potentially considered as a promising approach [8].
Exosomes (Exo), are disc-shaped nanoparticles naturally released by cells, with a diameter of 30–150 nm [9]. Exo serve as promising nano-sized drug delivery carriers and their phospholipid bilayer membrane protect the loaded small-molecule drugs from degradation [10]. Cell-free therapy using mesenchymal stem cells-derived exosomes (MSCs-Exo) as drug transporters potentially avoids some adverse reactions of MSCs transplantation, such as immune rejection, teratogenicity, and tumorigenicity [11]. And MSCs-Exo exhibit stem cell-like tumor-homing capacity [12]. Therefore, MSCs-Exo have been regarded as ideal drug carriers with nanoscale, low immunogenicity, low toxicity, biodegradability, high stability, and rich sources [13]. Researches implicated that TRAIL delivery by MSCs-Exo was more effective in killing tumor cells than recombinant soluble TRAIL protein [14].
Our previous research outlined that human chorion-derived mesenchymal stem cells (hCD-MSCs) exhibited good performance on chemotaxis towards cervical cancer cells [15]. On the basis of this premise, hCD-MSCs were selected as the cell source to manufacture Exo (hCD-MSCs-Exo) for co-loading with TRAIL and Cisplatin (DDP) to treat cervical cancer in the present study. Firstly, lentivirus packaging technology was employed to construct TRAIL-engineered hCD-MSCs (hCD-MSCsTRAIL), and a novel pattern of Exo expressing TRAIL (hCD-MSCs-ExoTRAIL) was further isolated. Secondly, chemotherapeutic agent DDP was encapsulated into hCD-MSCs-ExoTRAIL by electroporation method, thus developing a drug delivery system of hCD-MSCs-Exo co-loaded with TRAIL and DDP (DDP & hCD-MSCs-ExoTRAIL). Subsequently, DDP & hCD-MSCs-ExoTRAIL were administered in cervical cancer cells and cervical cancer-bearing nude mice to observe its antitumor activity. Collectively, application of DDP & hCD-MSCs-ExoTRAIL drug delivery systems may be a feasible scheme for cervical cancer treatment.
Materials and methods
The work has been reported in line with the ARRIVE guidelines 2.0.
Cell lines and culture
The cervical squamous carcinoma cell line SiHa and cervical adenocarcinoma cell line HeLa were acquired from Shanghai Cell Biology Medical Research Institute, Chinese Academy of Sciences. The normal cervical epithelial cell line ECT1 was obtained from Shanghai BinSui Biological Technology Co., Ltd. High-glucose Dulbecco’s Modified Eagle’s Medium (DMEM; Gibco, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, USA) and 1% penicillin-streptomycin solution (Pen-Strep, 100 mg/mL; Invitrogen, UK) was employed to cultivate SiHa and HeLa cells. And Eagle’s minimum essential medium (EMEM) was used to incubate ECT1 cells. Cells were cultured at 37 °C in a humidity-controlled atmosphere with 5% CO2.
Compounds
Cisplatin (DDP, #HY-17394; MCE, New Jersey, USA) was solubilized in distilled water to generate 2 mM solution and preserved at -20 °C, which was diluted to specific concentrations with complete DMEM medium before use.
Separation and incubation of hCD-MSCs
The separation and incubation of hCD-MSCs were performed based on our previously mentioned protocol [15]. Placental chorionic tissues from healthy and full-term pregnant women (25–30 years of age) were obtained following cesarean section, which received approval from ethics committee of the Second Affiliated Hospital of Wenzhou Medical University. The chorionic tissues were washed with phosphate-buffered saline solution (PBS) and chopped up completely, and subsequently dissociated with 1 mg/mL collagenase type II (Solarbio, China) for 1 h at 37 °C in a water-bath shaker. Thereafter, cell suspension underwent filtration utilizing a 100-µm nylon mesh cell filter to remove the tissue and cellular debris. Next, cell suspension was gathered and subjected to centrifugation at 1000 rpm for 10 min, and the supernatant was removed carefully. Afterwards, isolated hCD-MSCs were resuspended in low-glucose DMEM medium consisting of 10% FBS and 1% Pen-Strep. The cell culture medium was replaced at 2-day intervals and cells were passaged at a confluence density ranging from 80 to 90%.
Multipotent differentiation and immunophenotype characterizations of hCD-MSCs
The hCD-MSCs at passage 3 (P3) were selected to for multipotent differentiation and immunophenotype characterizations. An appraisement of the multipotency of hCD-MSCs towards adipogenic, osteogenic, and chondrogenic lineages was performed. Concisely, cells were inoculated in 24-well plates at a density of 5000 cells per well. When cell confluence reached 60-70%, cell culture medium was replaced with adipogenic differentiation medium (Gibco, USA), osteogenic differentiation medium (Gibco, USA), and chondrogenesis differentiation medium (Gibco, USA), respectively. 4% paraformaldehyde fixation for 30 min was administrated in cells 2 weeks later. Subsequently, cells were subjected to Oil red O staining (Solarbio, China), Alkaline phosphatase staining (Beyotime, China), and Alcian blue staining (Solarbio, China), respectively, for confirming the positive induction.
The immunophenotype of hCD-MSCs was characterized by flow cytometry with fluorescent-labeled monoclonal antibodies against CD90, CD73, CD44, CD45, CD34, HLA-ABC, and HLA-DR (ThermoFisher, USA). Cells were subjected to trypsinization and PBS washing, followed by incubation with respective antibodies for 20 min at room temperature in light-protecting condition. Afterwards, the samples were processed by CytoFLEX flow cytometry (Beckman Coulter, Fullerton, USA).
Immunohistochemistry (IHC)
The cervical cancer tissues and paired adjacent non-cancerous cervical tissues were incubated with primary antibody DR4 (1:200, Affinity Biosciences, #AF0304) following the guidelines of immunohistochemistry. Concisely, deparaffinized and rehydrated tissue sections were subjected to microwave oven for antigen retrieval. Next, the tissue sections were incubated with primary antibodies overnight at 4 ℃ followed by incubation with appropriate secondary antibodies. A freshly-prepared DAB solution was employed for chromogenic reaction and nuclei were counterstained with hematoxylin. Positive staining appeared in brown, and representative pictures were captured by microscope (Leica Microsystems, Wetzlar, Germany).
Western blotting assay
Total proteins were extracted from cell lysates and phenylmethanesulfonylfluoride (PMSF) was applied to prevent proteins from degradation. The protein quantification was done by bicinchoninic acid (BCA) protein assay kit (Beyotime, China). The prepared protein (40 µg/path) was resolved on polyacrylamide sodium dodecyl sulfate (SDS-PAGE) gels and subsequently electro-transferred onto polyvinylidene difluoride (PVDF) membranes. Following blocking with 5% milk, membranes were incubated with primary antibodies overnight at 4 ℃. Primary antibody of DR4 (1:2000, Affinity Biosciences, #AF0304) were employed and the anti-rabbit secondary antibodies conjugated with horseradish peroxidase (HRP) were employed to detect the protein bands. The protein bands were visualized with enzyme-linked chemiluminescence detection kit (ECL) under the Chemiluminescence Imaging System (ChemiScope 6000, CLiNX, China).
Lentivirus and transfection
The pL-CMV-GFP-TRAIL-LAMP2b lentiviral vector and the negative control vector pL-CMV-GFP-blank-LAMP2b were acquired from Shanghai Nuobai Biological Technology Co., Ltd. (China), which were co-cultured with HEK-293T cells by using Lipofectamine 2000 (Invitrogen, USA) transfection reagent with a lentivirus transfection system, respectively. Then, the resulting virus particles were applied to transfect hCD-MSCs. The cell model stably overexpressing TRAIL (hCD-MSCsTRAIL) was generated by selection with 10 µg/mL Blasticidin-S (Solarbio, China), and the protein expression of TRAIL (1:1000, CST, #3219) was examined by western blotting.
Exo extraction and characterization
To obtain hCD-MSCs and hCD-MSCsTRAIL secreted Exo, cells were first maintained in low-glucose DMEM medium consisting of 10% FBS and 1% Pen-Strep until cell density reached 60–70% confluence. Cells were subsequently cultured in low-glucose DMEM medium containing 10% exosome-free FBS and 1% Pen-Strep for an additional 48 h. Then, culture supernatant was collected followed by a differential centrifugation at 4℃, including 3,000 g for 10 min and 10,000 g for 30 min to eliminate cells and debris. Exo was extracted at 100,000 g for 90 min at 4℃ in a Type Ti32 rotor by ultracentrifugation (Beckman Coulter, USA). After meticulously eliminating the supernatant, Exo was washed in PBS followed by a 90 min spin at 100,000 g in a Type Ti41 rotor at 4℃, and ultimately resuspended in PBS for preservation at -80℃ for follow-up usage. Herein, Exo derived from hCD-MSCs and hCD-MSCsTRAIL were referred as hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL, respectively.
The morphology of Exo was visualized and photographed with transmission electron microscopy (TEM; Tecnai G2 Spirit, Fei, USA). The size distribution of Exo was discerned by nanoparticle tracking analysis (NTA; ZetaView PMX 110, Particle Metrix, Germany). The protein content in Exo was determined by BCA protein assay according to manufacturer’s instructions. Moreover, representative markers (Alix, Annexin-V and Flotillin-1) and negative marker (GM130) of Exo were identified by Western blotting. Primary antibodies against Alix (1:1000, CST, #2171), Annexin-V (1:1000, CST, #8555), Flotillin-1 (1:1000, CST, #18634) and GM130 (1:1000, CST, #12480) were employed. The corresponding secondary antibodies conjugated with horseradish peroxidase (HRP) were visualized to inspect the protein bands.
Uptake of Exo in cervical cancer cells
The purified hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL were labeled with PKH26 red fluorescent labeling kit (Umibio, China). Briefly, Exo suspension was mixed with 100 µM PKH26 staining working solution at room temperature for 30 min shielded from light, and the reaction was ceased by adding 100 µL low-glucose DMEM medium containing 10% exosome-free FBS. The PKH26-labeled Exo was resuspended with PBS and centrifuged at 100,000 g in a Type Ti41 rotor for 90 min at 4 ℃ to remove the excess dye. The collected Exo were named as PKH26-hCD-MSCs-Exo or PKH26-hCD-MSCs-ExoTRAIL.
The cervical cancer cells (SiHa and HeLa) were seeded onto coverslips in 6-well plates (2 × 105 cells/well) and cultured for 24 h at 37°C with 5% CO2. Afterwards, cells were grown with medium consisting of 4 µg/mL PKH26-hCD-MSCs-Exo or 4 µg/mL PKH26-hCD-MSCs-ExoTRAIL for 24 h shielded from light, at 37°C with 5% CO2. Subsequently, cells were treated with PBS rinsing, 4% paraformaldehyde fixing, and 4’, 6-diamidino-2-phenylindole (DAPI; Abcam, ab104139) dyeing. Eventually, cells were photographed with the laser scanning confocal microscope (Leica, Germany).
Cell viability assay
Cells were inoculated at a proper density in 96-well plates overnight and subsequently incubated with different vehicle or therapeutic regimens for 48 h. Thereafter, CCK-8 reagent (10 µL/well) was added for another incubation of 2 h, and the optical density (OD) at 450 nm was acquired with Micro-plate Reader (Bio-Rad, USA).
Cell apoptosis assay
Cells were inoculated at a proper density in 60-mm dish overnight and subsequently incubated with vehicle or therapeutic regimens for 48 h. Next, cells were gathered through trypsinization, resuspended in binding buffer, and double stained with Annexin V conjugated with phycoerythrin (PE) and 7-amino-actinomycin D (7-AAD) for 20 min avoiding light. Stained cells were evaluated with CytoFLEX flow cytometry (Beckman Coulter, Fullerton, USA).
Fabrication of DDP & hCD-MSCs-ExoTRAIL
To entrap drug DDP into hCD-MSCs-ExoTRAIL, 32 µg/mL of hCD-MSCs-ExoTRAIL and 6 µM of DDP were mingled with ice-cold PBS in 0.4 cm electroporation cuvette (Bio-Rad, USA). Afterwards, the mixture was electroporated by once pulse at 400 V, 150 µF capacitance using Gene Pulser Xcell (Bio-Rad, USA), and subsequently maintained at 37 ℃ for 30 min for the recovery of Exo membrane. Following the reaction termination, the mixture was ultracentrifugated at 100,000 g for 90 min to eliminate free DDP and obtain DDP & hCD-MSCs-ExoTRAIL nanoparticles. The platinum encapsulated in Exo was quantified using inductively coupled plasma mass spectrometry (ICP-MS) and DDP loading efficiency (%) in DDP & hCD-MSCs-ExoTRAIL nanoparticles was calculated. The traits of the fabricated DDP & hCD-MSCs-ExoTRAIL nanoparticles were described with TEM and NTA. For stability evaluation, nanoparticles were dispersed in pure PBS at -80 ℃ or 4 ℃. And the size distribution of nanoparticles was measured by dynamic light scattering (DLS) using a nanoparticle size analyzer at the days 0, 1, 2, 3, 5, 7.
Drug administration in tumor-bearing nude mice model
All animal experiments were in line with the regulations promulgated by the Institutional Animal Care and Use Committee of Wenzhou Medical University (No.wydw2021-0176). Female BALB/c nude mice (5 weeks of age) were ordered from Beijing Vital River Laboratory Animal Technology Co., Ltd., and housed in standard rearing condition. To establish cervical cancer-bearing mice, 1.5 × 106 SiHa cells in 100 µL PBS were transplanted subcutaneously in the right buttock of mice aged 7 weeks. When tumors grow to around 30 mm3 (volume = ½ × length × width2, as examined with vernier caliper [16]), mice were randomized into 4 clusters with 4 mice per cluster and subsequently intratumorally administrated with PBS, DDP (2 mg/kg), hCD-MSCs-ExoTRAIL (2 µg/g), and DDP & hCD-MSCs-ExoTRAIL (as obtained from electroporation with 2 mg/kg DDP and 2 µg/g hCD-MSCs-EXOTRAIL), respectively. The administration was repeated at 4-day intervals for 4 cycles. The tumor size and mice body weight were monitored before administration every time, and the last measurement was done 4 days after the fourth treatment.
Four days post the fourth treatment, mice were anesthetized intraperitoneally with pentobarbital sodium (100 mg/kg, #P3761; Sigma, USA). Peripheral blood collection of mice via submandibular vein puncture was applied for blood routine and biochemistry inspection. Whereafter, mice were euthanized by cervical dislocation method, and major organs (heart, liver, spleen, lung, kidney) and tumors were harvested, 4% paraformaldehyde-fixed, dehydrated, paraffin-embedded and sectioned. Tissue sections of major organs were deparaffinized and subjected to hematoxylin-eosin (HE) staining for histopathological lesions assessment. Tumor slices were processed for fluorescent terminal-deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) staining to evaluate tumor cells apoptosis. Additionally, total proteins were extracted from frozen tumor tissues, and protein expressions of JNK (1:1000, abcam, #ab179461), c-Jun (1:1000, CST, #9165), p-c-Jun (1:1000, CST, #3270) and survivin (1:1000, CST, #2808) in tumors were detected by Western blotting.
Statistical analysis
Experimental data were analyzed and plotted with GraphPad Prism 8.0 software, and delineated as mean ± standard deviation (SD). Statistical criteria were validated by Student’s t-test for two groups and One-way analysis of variance (ANOVA) for three or more groups. The least significance method was employed for data exhibiting homogeneous variances, and data with nonhomogeneous variances were analyzed with Dunnett’s T3 method. Moreover, non-parametric data were compared by Mann-Whitney U tests for two groups and Kruskal-Wallis tests for three or more groups. Significant probability (P)-values are represented as****P < 0.0001, ***P < 0.001, **P < 0.01, and *P < 0.05.
Results
Isolation and characterization of hCD-MSCs
Firstly, IHC staining presented that DR4 protein was higher expressed in cervical cancer tissues than adjacent non-cancerous cervical tissues (Fig. 1A). In comparison with ECT1 cells, Western blotting revealed that DR4 protein was highly expressed in cervical cancer cells SiHa and HeLa (Fig. 1B).
Characterizations of the cultured hCD-MSCs. (A) Immunohistochemistry (IHC) depicted the DR4 protein expression in cervical cancer tissues and adjacent normal tissues (original magnification, 200x). (B) Western blotting revealed DR4 protein expression in cervical cancer cells and normal cervical epithelial cells. (C) Flow cytometry analysis of specific surface markers of hCD-MSCs. (D-F) Morphological observation of the trilineage differentiation of hCD-MSCs (original magnification, 200x). Oil red staining reflected the adipogenic differentiation in (D). Alkaline phosphatase esterase staining reflected the osteogenic differentiation in (E). Alcian blue staining reflected the chondrogenic differentiation in (F). (G) Western blotting revealed the TRAIL protein expression in hCD-MSCsTRAIL. Full-length blots are presented in Supplementary Fig. 1E
Microscopic observations displayed that the cultured hCD-MSCs were spindle-shaped and adherent growth as monolayer (Fig. s1). Moreover, hCD-MSCs were positive for mesenchymal surface markers CD90, CD73, CD44, and HLA-ABC (>95%), while negative for hematopoietic surface markers CD45, CD34, and myeloid marker HLA-DR (<2%) (Fig. 1C). After selective induction, Oil red O staining verified the intracellular lipid accumulation (Fig. 1D), and Alkaline phosphatase staining confirmed the cytoplasm calcium deposits (Fig. 1E), and Alcian blue staining validated the proteoglycan production (Fig. 1F), reflecting that hCD-MSCs presented multipotent differentiation properties comprising of adipogenesis, osteogenesis, and chondrogenesis, respectively. The features addressed above complied with the classical characteristics of MSCs, which revealed the successful extraction and propagation of hCD-MSCs from placental chorionic tissues.
Moreover, Western blotting (Fig. 1G) validated the successful transfection of pL-CMV-GFP-TRAIL-LAMP2b plasmid into hCD-MSCs, with a higher expression of TRAIL protein in transfected cells (hCD-MSCsTRAIL).
Identification and characterization of hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL
Firstly, TEM exhibited that hCD-MSCs-Exo (Fig. 2A) and hCD-MSCs-ExoTRAIL (Fig. 2B) both manifested as a canonical saucer-shaped morphology. Secondly, NTA revealed that hCD-MSCs-Exo (Fig. 2C) and hCD-MSCs-ExoTRAIL (Fig. 2D) showed a similar size distribution and presented with a mean diameter of 136.3 ± 46.7 nm and 138.6 ± 47.4 nm, respectively. Thirdly, Western blotting confirmed that TRAIL protein was enriched in hCD-MSCs-ExoTRAIL in comparison with hCD-MSCs-Exo (Fig. 2E). Furthermore, both hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL positively expressed exosome biomarker proteins Alix, Annexin-V and Flotillin-1, whereas the negative extracellular vesicles surface marker GM130 was not presented (Fig. 2E). Collectively, these findings validated the successful isolation and purification of hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL.
Characterizations of the isolated hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL. (A-B) Transmission. electron microscope (TEM) images of hCD-MSCs-Exo in (A) and hCD-MSCs-Exo TRAIL in (B). (C-D) Nanoparticle tracking analysis (NTA) of the size and distribution of hCD-MSCs-Exo in (C) and hCD-MSCs-Exo TRAILin (D). (E) Western blotting revealed TRAIL protein and specific molecular markers expressions in hCD-MSCs-Exo and hCD-MSCs-Exo TRAIL. Full-length blots are presented in Supplementary Fig. 2E
Internalization of Exo by cervical cancer cells
After co-culturing of cervical cancer cells (SiHa and HeLa) with PKH26-hCD-MSCs-Exo or PKH26-hCD-MSCs-ExoTRAIL for 24 h, laser scanning confocal microscope (Fig. 3A for SiHa; Fig. 3B for HeLa) exhibited that red fluorescence (PKH26 labeled Exo) concentrated in cytoplasm surrounding the nuclei (stained as blue by DAPI). The results confirmed the uptake of hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL by cervical cancer cells.
Impacts of hCD-MSCs-ExoTRAIL on proliferation and apoptosis of cervical cancer cells. (A-B) Laser scanning confocal microscope images of Exo internalization by SiHa cells (A) and HeLa cells (B) (original magnification, 1000x). (C-D) Effects of Exo (0, 0.5, 1, 2, 4, 8, 16, 32 µg/mL) on proliferative abilities of SiHa cells (C) and HeLa cells (D), n = 3. (E) Effects of Exo (8 µg/mL) on apoptosis of SiHa cells, n = 3. (F) Quantitative analysis of the results in (E) of SiHa cells. (G) Effects of Exo (8 µg/mL) on apoptosis of HeLa cells, n = 3. (H) Quantitative analysis of the results in (G) of HeLa cells. (****P < 0.0001)
Impact of hCD-MSCs-ExoTRAIL on proliferation and apoptosis of cervical cancer in vitro
The proliferative ability of cervical cancer cells with the administration of various concentrations of hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL (0, 0.5, 1, 2, 4, 8, 16, 32 µg/mL) for 48 h was assessed by CCK-8 assay. As exhibited in Fig. 3C, SiHa cells treated with hCD-MSCs-ExoTRAIL had a lower survival rate with respect to hCD-MSCs-Exo in a concentration-dependent pattern (P<0.05). Moreover, resembled results were detected in HeLa cells (P<0.05) (Fig. 3D).
The apoptosis of cervical cancer cells with the treatment of 8 µg/mL hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL for 48 h was appraised by flow cytometry. As illustrated in Fig. 3E-F, hCD-MSCs-ExoTRAIL induced higher apoptosis of SiHa cells relative to hCD-MSCs-Exo (P<0.05). Additionally, similar results were obtained in HeLa cells (P<0.05) (Fig. 3G-H).
Above findings presented that the engineered hCD-MSCs-ExoTRAIL showed a significant advantage for attenuating proliferation and facilitating apoptosis of cervical cancer cells. Therefore, hCD-MSCs-ExoTRAIL was selected to pack with DDP.
Effects of DDP on proliferation of cervical cancer cells
CCK-8 assay was employed to evaluate the antiproliferative capacity of DDP with various concentrations (0, 2, 4, 6, 8, 10, 12, 14, 16, 18 µM) for 48 h against cervical cancer cells. DDP displayed concentration-dependent cytotoxicity to SiHa and HeLa cells (Fig. 4A-B). As shown in Fig. 4A, the IC50 value was 12 µM and the IC30 value was 6 µM for DDP in SiHa cells. As exhibited in Fig. 4B, the IC50 value was 8 µM and the IC30 value was 6 µM for DDP in HeLa cells. Therefore, 6 µM DDP at 48 h administration was applied in the following experiments.
Characterizations and impacts of DDP & hCD-MSCs-ExoTRAIL nanopaticles on proliferation and apoptosis of cervical cancer cells. (A-B) Effects of DDP (0, 2, 4, 6, 8, 10, 12, 14, 16, 18 µM) on proliferative abilities of SiHa cells (A) and HeLa cells (B), n = 3. (C) TEM images of DDP & hCD-MSCs-ExoTRAIL nanopaticles. (D) NTA of the size and distribution of DDP & hCD-MSCs-ExoTRAIL nanopaticles. (E) DLS analysis of DDP & hCD-MSCs-ExoTRAIL nanoparticles at the days 0, 1, 2, 3, 5, 7 stored at -80℃ or 4 ℃. (F-G) Proliferative abilities of SiHa cells (F) and HeLa cells (G) with the treatment of control, DDP, hCD-MSCs-ExoTRAIL and DDP & hCD-MSCs-ExoTRAIL, n = 3. (H-I) Apoptosis of SiHa cells (H) and HeLa cells (I) with the treatment of control, DDP, hCD-MSCs-ExoTRAIL and DDP & hCD-MSCs-ExoTRAIL, n = 3. (J) Quantitative analysis of the results in (H) of SiHa cells. (K) Quantitative analysis of the results in (I) of HeLa cells. (***P < 0.001, ****P < 0.0001)
Characterization of DDP & hCD-MSCs-ExoTRAIL
Electroporation method was implemented to synthesize the drug delivery systems of hCD-MSCs-Exo co-loaded with TRAIL and DDP (DDP & hCD-MSCs-ExoTRAIL). As presented in Fig. 4C, TEM image exhibited that DDP & hCD-MSCs-ExoTRAIL remained the typical saucer-shaped appearance, demonstrating that Exo membrane structure showed no damage after DDP package by electroporation. Moreover, NTA described that the average diameter of DDP & hCD-MSCs-ExoTRAIL was 128.5 ± 36.3 nm (Fig. 4D). Next, DLS analysis exhibited that DDP & hCD-MSCs-ExoTRAIL appeared negligible size change at -80 ℃ or 4 ℃ for 7 days (Fig. 4E). In addition, ICP-MS analysis determined that the DDP loading efficiency of DDP & hCD-MSCs-ExoTRAIL was 32.12 ± 2.74%.
Effects of DDP & hCD-MSCs-ExoTRAIL on proliferation and apoptosis of cervical cancer in vitro
The CCK-8 and flow cytometry assay were performed by incubating cervical cancer cells with DMEM (control group), DDP (6 µM), hCD-MSCs-ExoTRAIL (32 µg/mL), and DDP & hCD-MSCs-ExoTRAIL (as obtained from electroporation with 6 µM DDP and 32 µg/mL hCD-MSCs-EXOTRAIL) for 48 h, to evaluate cell proliferation and apoptosis, respectively.
In comparison to control group, survival rates of SiHa cells in DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL groups were lower, of which survival rate ranked the lowest in SiHa cells treated with DDP & hCD-MSCs-ExoTRAIL (P<0.05) (Fig. 4F). Moreover, similar results were revealed in HeLa cells (P<0.05) (Fig. 4G).
In contrast to control group, apoptosis of SiHa cells in DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL groups were higher, of which apoptosis ranked the highest in SiHa cells treated with DDP & hCD-MSCs-ExoTRAIL (P<0.05) (Fig. 4H and J). Moreover, similar results were observed in HeLa cells (P<0.05) (Fig. 4I and K).
These results illustrated that DDP & hCD-MSCs-ExoTRAIL exhibited the greatest proliferation inhibiting and apoptosis promoting capacities of cervical cancer cells.
In vivo therapeutic antitumor activity and biosafety evaluation
The therapeutic antitumor activity was evaluated in cervical cancer-bearing nude mice with the treatment of PBS, DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL, respectively. The tumors continuously grew in PBS group, which displayed as the most rapidly growth pattern among these four groups (P<0.05) (Fig. 5A). Moreover, tumors grew slowly with varying degrees in DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL groups, of which tumors grew slowest in DDP & hCD-MSCs-ExoTRAIL group (P<0.05) (Fig. 5A). After treatment completion, the ex vivo tumor images and tumor weights from these four groups visually demonstrated the antitumor efficacy, of which tumor weights were the smallest in the DDP & hCD-MSCs-ExoTRAIL group (P<0.05) (Fig. 5B-C). These results described that DDP & hCD-MSCs-ExoTRAIL exerted the most potently inhibitory function on cervical cancer growth.
Therapeutic activity of DDP & hCD-MSCs-ExoTRAIL nanoparticles in cervical cancer-bearing nude mice. (A) Tumor growth curves of mice with the treatment of control, DDP, hCD-MSCs-ExoTRAIL and DDP & hCD-MSCs-ExoTRAIL, n = 4 mice per group. (B-C) Tumor tissue images (B) and tumor weights (C) of mice with the treatment of control, DDP, hCD-MSCs-ExoTRAIL and DDP & hCD-MSCs-ExoTRAIL, n = 4 mice per group. (D) TUNEL-DAPI fluorescence staining of tumor tissues reflecting the cell apoptosis (original magnification, 200x). (E) Western blotting revealed the expression of apoptotic-related proteins in tumor tissues. Full-length blots are presented in Supplementary Fig. 5E. (*P < 0.05, ***P < 0.001, ****P < 0.0001)
Afterwards, TUNEL staining illustrated the apoptotic cells (stained as green fluorescence) in tumor tissues. As shown in Fig. 5D, the apoptotic cells were largely augmented in tumors treated with DDP & hCD-MSCs-ExoTRAIL versus to PBS, DDP, and hCD-MSCs-ExoTRAIL groups. Next, apoptosis-related proteins in tumor tissues were further investigated by Western blotting. In contrast with PBS group, the expression level of JNK and p-c-Jun proteins were elevated in DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL groups, with the highest expression in DDP & hCD-MSCs-ExoTRAIL group (Fig. 5E). Survivin protein expression was decreased in DDP, hCD-MSCs-ExoTRAIL, and DDP & hCD-MSCs-ExoTRAIL groups in comparison with PBS group, with the lowest expression in DDP & hCD-MSCs-ExoTRAIL group (Fig. 5E). These results suggested that DDP & hCD-MSCs-ExoTRAIL exhibited the strongest induction of apoptosis in cervical cancer.
Additionally, body weight, histopathology of major organs, and primary blood parameters of mice were assessed to systematically monitor the biosafety profiles of DDP & hCD-MSCs-ExoTRAIL treatment in vivo. No notable differences were revealed regarding the body weights of mice following administration with different drug regimens (P>0.05) (Fig. 6A). Moreover, HE staining presented that no noticeable histopathological alterations or impairments were inspected in major organ tissues (heart, liver, spleen, lung, kidney) of mice treated with different drug regimens (Fig. 6B). Furthermore, blood routine (WBC, Lym, Neu, RBC, HB, MCV, HCT, PLT) (Fig. 6C-D) and plasma biochemistry inspection (Fig. 6E) indicating liver (ALT, AST, TP, ALB, GLO), kidney (BUN, CREA), and heart ( LDH) damages were implemented to perceive slight lesions that missed to be reflected by HE staining. More importantly, these biomarker levels among mice receiving different drug treatments displayed no obvious differences (P>0.05) and the CK-MB concentrations were less than 0.18 µg/L of all mice. These results documented that DDP & hCD-MSCs-ExoTRAIL showed favorable biosafety in vivo.
Biosafety evaluation of DDP & hCD-MSCs-ExoTRAIL nanoparticles in cervical cancer-bearing nude mice. (A) Body weight curves of mice with the treatment of control, DDP, hCD-MSCs-ExoTRAIL and DDP & hCD-MSCs-ExoTRAIL, n = 4 mice per group. (B) Hematoxylin-eosin (HE) staining reflected the pathological morphology of major organs (Heart, Liver, Spleen, Lung, and Kidney) of mice (original magnification, 200x). (C-D) Blood routine parameters of mice, n = 4 mice per group. (E) Plasma biochemistry parameters of mice, n = 4 mice per group
Discussion
The most common pathological types of cervical cancer are cervical squamous cell carcinoma, which constitutes approximately 75% of cases, and cervical adenocarcinoma, accounting for roughly 20% of cases [17]. Lately, the demographic profile of cervical cancer patients has exhibited a noticeable shift towards a younger age group [18]. Neoadjuvant chemotherapy has emerged as a crucial and alternative therapeutic approach, particularly for reproductive-age cervical cancer patients aiming to preserve fertility [19]. DDP is an efficacious and widely applied chemotherapeutic drug for cervical cancer [20]. However, its non-selective cytotoxicity and dose-dependent toxic side effects necessitate the exploration of more efficient therapeutic regimens with lower-dose DDP [21]. Nanoscale drug delivery systems potentially benefit in ameliorating therapeutic performances of drugs because of the enhanced permeability and retention (EPR) effect [22]. Among these, MSCs-Exo have garnered significant attention as naturally occurring nano-sized drug delivery vehicles [23].
Shamili et al. [24] applied a non-viral polyplex, consisting of polyethylenimine 25 kDa and GFP-tagged TRAIL plasmid, to engineer TRAIL-expressing murine bone marrow-derived MSCs. The resultant Exo containing TRAIL (Exo-TRAIL) decreased cell viability and increased apoptosis of B16F0 mouse melanoma cells in contrast to Exo [24]. In the present study, DR4 protein was highly expressed in cervical cancer, indicating that DR4 could serve as target of nanoscale drug delivery systems for cervical cancer therapy. Then, we succeeded in isolating and culturing hCD-MSCs and subsequently obtained Exo from hCD-MSCs. Next, genetically engineered hCD-MSCsTRAIL were constructed by ligating TRAIL with exosomal transmembrane protein Lamp2b [25], and its secreted Exo were proved to highly express TRAIL, indicating that TRAIL loaded hCD-MSCs-Exo were successfully generated. In more detail, our study examined and depicted the shape, size, and specific biomarker proteins of the purified hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL, and these characterizations fulfilled the classical criteria for authentication of Exosomes [26]. Next, hCD-MSCs-Exo and hCD-MSCs-ExoTRAIL were confirmed to be effectively taken up by cervical cancer cells. Moreover, hCD-MSCs-ExoTRAIL exerted tumor-suppressive functions by weakening proliferative capacity and enhancing apoptotic capacity of cervical cancer cells, suggesting that hCD-MSCs-ExoTRAIL were ideal drug transport vehicles.
As it stands, exogenous loading of therapeutic agents following Exo purification can be performed by co-cultivation, sonication, electroporation, extrusion, and freeze-thaw cycles [27]. Moreover, the drug loading efficacy is of fundamental importance for evaluating the effectiveness of the established nanoscale drug delivery systems [28]. Zhang et al. [29] documented that the loading efficiency of DDP in umbilical cord blood-derived M1 macrophage Exo was estimated to around 30% with mild sonication approach. Considering the dual antitumor functions of chemo-gene combinational therapy [30], we established the nanoscale drug delivery systems by means of co-loading of TRAIL and DDP with hCD-MSCs-Exo. Our study successfully developed DDP & hCD-MSCs-ExoTRAIL by encapsulating DDP into hCD-MSCs-ExoTRAIL, which maintained the saucer shape with a diameter averaging 128.5 ± 36.3 nm. Additionally, the DDP loading capacity was approximately 32.12 ± 2.74%, achieved by employing electroporation technique.
Jiang et al. [31] designed nanoscale drug delivery systems via entrapping triptolide in TRAIL-engineered exosomes (TRAIL-Exo) secreted by TRAIL-overexpressing macrophage Raw264.7 cells, which was confirmed to play a stronger impact on cytotoxicity and pro-apoptosis compared with triptolide in malignant melanoma. Our study further assessed the therapeutic functions of DDP & hCD-MSCs-ExoTRAIL in cervical cancer. The DDP & hCD-MSCs-ExoTRAIL provided evident advantages in retarding cell growth and propelling cell apoptosis with respect to single DDP or hCD-MSCs-ExoTRAIL in cervical cancer in vitro, which revealed that the fabricated DDP & hCD-MSCs-ExoTRAIL exhibited remarkable tumoricidal activity in both cervical squamous cell carcinoma and cervical adenocarcinoma.
The mitogen-activated protein kinase (MAPK) signaling pathway comprises ERK1/2, p38, c-Jun N-terminal kinase (JNK) and MEK cascades, which modulating multiple cellular activities like apoptosis, autophagy, proliferation and differentiation [32]. Although exceptions exist, p38 MAPK and JNK pathways are generally relative to apoptosis promotion [33]. Frensemeier et al. [34] revealed that DDP facilitated apoptosis of cervical cancer cells through activation of JNK and c-Jun phosphorylation. Moreover, Mahalingam et al. [35] illustrated that recombinant human TRAIL protein-mediated pro-apoptotic function on colon carcinoma cells was related to an increase of JNK and c-Jun phosphorylation. Additionally, Xu et al. [36] documented that DDP plus TRAIL protein exerted their function in promoting cell apoptosis via reducing the expression of antiapoptotic protein Survivin in triple-negative breast tumor cells. In our study, the results acquired from these cervical cancer-bearing nude mice revealed that nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) were able to lessen tumor burden and hamper tumor growth more efficiently than DDP or hCD-MSCs-ExoTRAIL alone by xenograft volumetric and weight analysis. Additionally, TUNEL assay observed that DDP & hCD-MSCs-ExoTRAIL exhibited a greater capacity to prompt apoptosis in cervical cancer tissues than DDP or hCD-MSCs-ExoTRAIL alone. Moreover, DDP & hCD-MSCs-ExoTRAIL displayed maximum upregulation of JNK and p-c-Jun protein and downregulation of Survivin protein in contrast to DDP or hCD-MSCs-ExoTRAIL alone in the cervical cancer of mice. Therefore, the activation of JNK/p-c-Jun and suppression of Survivin potentially was one mechanism by which DDP & hCD-MSCs-ExoTRAIL performed its role in inducing apoptosis of cervical cancer.
The biological safety of nanoscale drug delivery systems is a crucial precondition for application in vivo and future clinical translation [37]. Next, our current study inspected the biosafety profile of DDP & hCD-MSCs-ExoTRAIL in mice. Firstly, the body weight of mice received four different treatments revealed no apparent difference. Secondly, histopathological and blood paraments evaluation reflected that DDP & hCD-MSCs-ExoTRAIL showed no toxic side effect in major organs of mice. Therefore, our present findings provided evidence that the nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) displayed superior anti-tumor efficacy and favorable biosafety in cervical cancer.
As widely known, cisplatin is extensively employed as a chemotherapeutic agent for the treatment of cervical cancer, especially for advanced or metastatic or recurrent cervical cancer [38]. Unfortunately, cisplatin administration is generally restricted by insufficient tumor chemotherapy response [39]. Therefore, clinical treatment requires higher dosage of cisplatin to achieve the anticipated therapeutic effect, while generating dose-related toxicities as nephrotoxicity, neurotoxicity and other serious undesirable effects [40]. In our present study, a low dosage of cisplatin delivered by hCD-MSCs-ExoTRAIL exerted superior ability to inhibit tumor growth than free cisplatin. Moreover, the nanoscale drug delivery system DDP & hCD-MSCs-ExoTRAIL appeared no apparent systemic toxicity. Based on the above results, we will continue to explore the long-term effects of DDP & hCD-MSCs-ExoTRAIL on tumor recurrence and metastasis, and elaborate the potential immune responses and immunogenicity associated with DDP or hCD-MSCs-ExoTRAIL in mice in future studies.
Certainly, potential challenges in translating DDP & hCD-MSCs-ExoTRAIL from preclinical studies to clinical practice need to be considered and addressed in follow-up investigations. Firstly, the preparation of exosomes is a time-consuming and labor-intensive process. Therefore, the primary bottleneck for clinical trial is the large-scale and rapid production of exosomes. The method for isolating high purity exosomes efficiently and economically is urgently needed in the future. Secondly, although DDP & hCD-MSCs-ExoTRAIL delivery system exhibits great potential for cervical cancer treatment, there is still a long way to go for clinical application. In present study, only one dosage and treatment regimen was evaluated in cervical cancer in vitro and in vivo, and additional experiments are required to meliorate the drug delivery system. We need to improve DDP encapsulation efficiency, and explore the most appropriate drug concentration to ascertain the most effective combination of DDP and exosomes. Moreover, we need to optimize the administration mode and schedule of DDP & hCD-MSCs-ExoTRAIL. Thirdly, individualized characterization of cervical cancer patients influences the therapeutic efficacy of DDP & hCD-MSCs-ExoTRAIL. We need to conduct multi-center prospective clinical trials to validate applicability of DDP & hCD-MSCs-ExoTRAIL in cervical cancer patients. Moreover, we need to obtain the approval of regulatory agencies for clinical applications.
Conclusions
Collectively, the rich source of human chorion will guarantee an adequate amount of hCD-MSCs and hCD-MSCs-Exo. The TRAIL-engineered hCD-MSCs-Exo featured as a tumor-suppressor, and acted as an appropriate nano-carrier platform for DDP encapsulation. Compared with single DDP or hCD-MSCs-ExoTRAIL, the developed nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) empowered improved therapeutic efficacy in hampering cervical cancer progression both in vitro and in vivo. Moreover, the DDP & hCD-MSCs-ExoTRAIL nanoparticles possessed an acceptable biosafety profile, holding great promise for its employment in cervical cancer as chemo-gene combinational therapy in clinical practice. Fortunately, Exo-based drug delivery systems present bright prospects for cervical cancer treatment.
Data availability
The data and materials that support the findings of this study are available from the corresponding author upon reasonable request.
Abbreviations
- WBC:
-
White blood cell
- Lym:
-
Lymphocyte
- Neu:
-
Neutrophil
- RBC:
-
Red blood cell
- HB:
-
Hemoglobin
- MCV:
-
Mean corpuscular volume
- HCT:
-
Hematocrit
- PLT:
-
Platelet
- ALT:
-
Alanine aminotransferase
- AST:
-
Aspartate aminotransferase
- TP:
-
Total protein
- ALB:
-
Albumin
- GLO:
-
Globulin
- BUN:
-
Blood urea nitrogen
- CREA:
-
Creatinine
- CK-MB:
-
Creatine kinase-MB isoenzyme
- LDH:
-
Lactate dehydrogenase
References
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–63.
Ferrall L, Lin KY, Roden RBS, Hung C-F, Wu TC. Cervical Cancer immunotherapy: facts and hopes. Clin Cancer Res. 2021;27(18):4953–73.
Tewari KS, Monk BJ. New strategies in Advanced Cervical Cancer: from angiogenesis blockade to Immunotherapy. Clin Cancer Res. 2014;20(21):5349–58.
Theobald L, Stroeder R, Melchior P, Iordache II, Tanzer T, Port M, et al. Chemoradiotherapy-induced increase in Th17 cell frequency in cervical cancer patients is associated with therapy resistance and early relapse. Mol Oncol. 2021;15(12):3559–77.
Montinaro A, Walczak H. Harnessing TRAIL-induced cell death for cancer therapy: a long walk with thrilling discoveries. Cell Death Differ. 2023;30(2):237–49.
Nguyen TTK, Woo SM, Seo SU, Banstola A, Kim H, Duwa R, et al. Enhanced anticancer efficacy of TRAIL-conjugated and odanacatib-loaded PLGA nanoparticles in TRAIL resistant cancer. Biomaterials. 2025;312:122733.
Huang RY, Lin YH, Lin SY, Li YN, Chiang CS, Chang CW. Magnetic ternary nanohybrids for nonviral gene delivery of stem cells and applications on cancer therapy. Theranostics. 2019;9(8):2411–23.
Jyotsana N, Zhang Z, Himmel LE, Yu F, King MR. Minimal dosing of leukocyte targeting TRAIL decreases triple-negative breast cancer metastasis following tumor resection. Sci Adv. 2019;5(7):eaaw4197.
Han QF, Li WJ, Hu KS, Gao J, Zhai WL, Yang JH, et al. Exosome biogenesis: machinery, regulation, and therapeutic implications in cancer. Mol Cancer. 2022;21(1):207.
Li MY, Liu LZ, Dong M. Progress on pivotal role and application of exosome in lung cancer carcinogenesis, diagnosis, therapy and prognosis. Mol Cancer. 2021;20(1):22.
Xie Y, Sun Y, Liu Y, Zhao J, Liu Q, Xu J, et al. Targeted delivery of RGD-CD146 + CD271 + human umbilical cord mesenchymal stem cell-derived Exosomes promotes blood–spinal cord barrier repair after spinal cord Injury. ACS Nano. 2023;17(18):18008–24.
Liang L, Zhao L, Wang Y, Wang Y. Treatment for Hepatocellular Carcinoma is enhanced when Norcantharidin is encapsulated in Exosomes derived from bone marrow mesenchymal stem cells. Mol Pharm. 2021;18(3):1003–13.
Fan R, Su L, Zhang H, Jiang Y, Yu Z, Zhang X, et al. Enhanced therapeutic effect of PEDF-loaded mesenchymal stem cell-derived small extracellular vesicles against oxygen-induced retinopathy through increased stability and penetrability of PEDF. J Nanobiotechnol. 2023;21(1):327.
Yuan Z, Kolluri KK, Gowers KH, Janes SM. TRAIL delivery by MSC-derived extracellular vesicles is an effective anticancer therapy. J Extracell Vesicles. 2017;6(1):1265291.
Song Y, Li R, Ye M, Pan C, Zheng L, Wang Z-w, et al. Differences in chemotaxis of human mesenchymal stem cells and cervical cancer cells. Apoptosis. 2022;27(11–12):840–51.
Sudhan DR, Guerrero-Zotano A, Won H, González Ericsson P, Servetto A, Huerta-Rosario M, et al. Hyperactivation of TORC1 drives resistance to the Pan-HER tyrosine kinase inhibitor Neratinib in HER2-Mutant cancers. Cancer Cell. 2020;37(2):183–e995.
Liu C, Zhang M, Yan X, Ni Y, Gong Y, Wang C, et al. Single-cell dissection of cellular and molecular features underlying human cervical squamous cell carcinoma initiation and progression. Sci Adv. 2023;9(4):eadd8977.
Arbyn M, Weiderpass E, Bruni L, de Sanjosé S, Saraiya M, Ferlay J, et al. Estimates of incidence and mortality of cervical cancer in 2018: a worldwide analysis. Lancet Glob Health. 2020;8(2):e191–203.
Buda A, Borghese M, Puppo A, Perotto S, Novelli A, Borghi C, et al. Neoadjuvant Chemotherapy prior fertility-sparing surgery in women with FIGO 2018 Stage IB2 Cervical Cancer: a systematic review. Cancers. 2022;14(3):797.
Mountzios G, Soultati A, Pectasides D, Pectasides E, Dimopoulos M-A, Papadimitriou CA. Developments in the systemic treatment of metastatic cervical cancer. Cancer Treat Rev. 2013;39(5):430–43.
Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4(4):307–20.
Davoudi M, Jadidi Y, Moayedi K, Farrokhi V, Afrisham R. Ameliorative impacts of polymeric and metallic nanoparticles on cisplatin-induced nephrotoxicity: a 2011–2022 review. J Nanobiotechnol. 2022;20(1):504.
Lin Z, Wu Y, Xu Y, Li G, Li Z, Liu T. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: recent advances and therapeutic potential. Mol Cancer. 2022;21(1):179.
Shamili FH, Bayegi HR, Salmasi Z, Sadri K, Mahmoudi M, Kalantari M, et al. Exosomes derived from TRAIL-engineered mesenchymal stem cells with effective anti-tumor activity in a mouse melanoma model. Int J Pharm. 2018;549(1–2):218–29.
Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383–90.
Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.
Tenchov R, Sasso JM, Wang X, Liaw WS, Chen CA, Zhou QA. Exosomes - Nature’s lipid nanoparticles, a rising star in Drug Delivery and Diagnostics. ACS Nano. 2022;16(11):17802–46.
Gutierrez-Millan C, Calvo Diaz C, Lanao JM, Colino CI. Advances in Exosomes-based drug Delivery systems. Macromol Biosci. 2021;21(1):e2000269.
Zhang X, Wang J, Liu N, Wu W, Li H, Lu W, et al. Umbilical cord blood-derived M1 macrophage exosomes loaded with cisplatin target ovarian Cancer in vivo and reverse cisplatin resistance. Mol Pharm. 2023;20(11):5440–53.
Shrestha B, Wang L, Zhang H, Hung CY, Tang L. Gold nanoparticles mediated drug-gene combinational therapy for breast Cancer Treatment. Int J Nanomed. 2020;15:8109–19.
Jiang L, Gu Y, Du Y, Tang X, Wu X, Liu J. Engineering Exosomes endowed with targeted delivery of Triptolide for Malignant Melanoma Therapy. ACS Appl Mater Interfaces. 2021;13(36):42411–28.
Sheng R, Liu J, Zhang W, Luo Y, Chen Z, Chi J, et al. Material stiffness in Cooperation with macrophage paracrine signals determines the tenogenic differentiation of mesenchymal stem cells. Adv Sci (Weinh). 2023;10(17):e2206814.
Yue J, López JM. Understanding MAPK signaling pathways in apoptosis. Int J Mol Sci. 2020;21(7):2346.
Frensemeier K, Holzer A, Hoppe-Seyler K, Hoppe-Seyler F. Dickkopf-1 expression is repressed by oncogenic human papillomaviruses (HPVs) and regulates the cisplatin sensitivity of HPV-positive cancer cells in a JNK-dependent manner. Int J Cancer. 2022;151(12):2215–28.
Mahalingam D, Keane M, Pirianov G, Mehmet H, Samali A, Szegezdi E. Differential activation of JNK1 isoforms by TRAIL receptors modulate apoptosis of colon cancer cell lines. Br J Cancer. 2009;100(9):1415–24.
Xu L, Yin S, Banerjee S, Sarkar F, Reddy KB. Enhanced anticancer effect of the combination of cisplatin and TRAIL in triple-negative breast tumor cells. Mol Cancer Ther. 2011;10(3):550–7.
Yang L, Sun J, Liu Q, Zhu R, Yang Q, Hua J, et al. Synergetic functional nanocomposites enhance immunotherapy in solid tumors by remodeling the Immunoenvironment. Adv Sci (Weinh). 2019;6(8):1802012.
Gennigens C, Jerusalem G, Lapaille L, De Cuypere M, Streel S, Kridelka F, et al. Recurrent or primary metastatic cervical cancer: current and future treatments. ESMO Open. 2022;7(5):100579.
Mann M, Singh VP, Kumar L. Cervical cancer: a tale from HPV infection to PARP inhibitors. Genes Dis. 2023;10(4):1445–56.
Xu L, Zhang K, Ma X, Li Y, Jin Y, Liang C, et al. Boosting cisplatin chemotherapy by nanomotor-enhanced tumor penetration and DNA adducts formation. J Nanobiotechnol. 2022;20(1):429.
Funding
This work was supported by National Natural Science Foundation of China (Grant No. 82172088) and Wenzhou Major Science and Technology Innovation Project (Grant No. ZY2022012). The sponsors of the study had no involvement in the data collection, analysis, or interpretation, or in the writing of the manuscript. The authors declare that they have not used Artificial Intelligence in this study.
Author information
Authors and Affiliations
Contributions
Conceptualization, X.Z., J.Z., and M.Y.; methodology, M.Y., T.L., L.M., and H.J.; formal analysis, M.Y., T.L., H.J. and L.M.; investigation, J.Z., M.Y., and Z.X.; writing-original draft, M.Y. and T.L.; writing-review and editing, M.Y., T.L., L.M., H.J., Z.X., H.W., J.Z. and X.Z.; visualization, T.L., L.M., and H.W. supervision, X.Z. and J.Z.; funding acquisition, X.Z. All authors have read and agreed to the published version of the manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
This study has been approved by the ethics committee of the Second Affiliated Hospital of Wenzhou Medical University. And informed consent was obtained from all subjects involved in the study. The project was titled as “The establishment of a co-delivery Nano-granular system based on TRAIL-exosome, siRNA and cisplatin and its application in drug-resistant cervical cancer (No. 2016kykt31)”, and was approved on 03/10/2016. The animal experiments were in line with the regulations promulgated by the Institutional Animal Care and Use Committee of Wenzhou Medical University. The project was titled as “The mechanistic exploration of a novel biomimetic nanozyme based on TRAIL target for reversing radio-resistance of cervical cancer to synergistically improve immunotherapy (No. wydw2021-0176)”, and was approved on 03/10/2021. Shanghai Cell Biology Medical Research Institute and Shanghai BinSui Biological Technology Co. have confirmed that there was initial ethical approval for collection of human cells (SiHa, HeLa, ECT1), and that the donors had signed informed consent.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Ye, M., Liu, T., Miao, L. et al. Cisplatin-encapsulated TRAIL-engineered exosomes from human chorion-derived MSCs for targeted cervical cancer therapy. Stem Cell Res Ther 15, 396 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-04006-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-024-04006-6