Repositioning of aripiprazole, an anti‑psychotic drug, to sensitize the chemotherapy of pancreatic cancer
- Authors:
- Published online on: November 12, 2024 https://doi.org/10.3892/ijmm.2024.5458
- Article Number: 17
-
Copyright: © Cho et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Pancreatic cancer is an aggressive malignant tumor and pancreatic ductal adenocarcinoma (PDAC) is the most common form, representing ~90% of all types of pancreatic cancer (1). PDAC is one of the most lethal types of cancer, with a 5-year survival rate of <10%. Of patients with PDAC, ~50% are diagnosed with locally advanced disease, but 85-90% are unsuitable for surgical resection; so the majority receive systemic treatment (2). Despite recent advances in combination chemotherapy regimens based on gemcitabine or 5-FU, the median overall survival for patients with metastatic PDAC is <1 year (3). This underscores the urgent need for novel and effective treatments for PDAC. The standard therapeutic approach for newly diagnosed PDAC involves a combination of surgery and platinum-based chemotherapy.
Cisplatin, a potent platinum-based chemotherapeutics, has been used to treat various solid tumors, including lung, breast, ovarian and PDAC (4). Cisplatin interferes with DNA replication and transcription, leading to cell death, which is particularly effective against rapidly dividing cells such as cancer cells (5). Cisplatin- and platinum-based therapies are commonly employed to treat PDAC, either alone or in combination with other chemotherapeutic agents such as gemcitabine (6). However, despite its clinical efficacy, cisplatin often causes severe side effects, including peripheral neuropathy, hearing loss and kidney damage (7). Therefore, there is an urgent need to develop new pancreatic cancer treatment strategies that do not cause these side effects.
Drug repositioning is a promising strategy for developing new cancer treatments. This approach involves identifying new therapeutic applications for drugs already used in clinical practice, including those approved for other diseases or those unsuccessful for their original indications (8). Repositioned drugs have undergone extensive testing, including safety, toxicity and pharmacokinetic evaluations, which can expedite their clinical use in cancer therapy at a lower discovery cost (9). More importantly, they can be used in combination with existing cancer therapies to enhance efficacy, overcome drug resistance and improve patient outcomes, thereby providing a route for optimizing and personalizing cancer treatment (10). The synergistic use of repositioned drugs with current treatment regimens offers an avenue to target rare and neglected cancers, providing new hope to patients with limited treatment options.
Markedly, a lower incidence of cancer has been noted in patients with schizophrenia who are prescribed neuroleptics. Some antipsychotic drugs including chlorpromazine and aripiprazole, have demonstrated anticancer effects in preclinical studies (11). Aripiprazole, the second-generation antipsychotic drug for mental disorders, exhibits unique pharmacological activities as a serotonin 5-HT1A and 5-HT2A antagonist. Studies have shown that aripiprazole possesses anticancer properties and enhances radiosensitizing effects in various cancer cells (12-18). It also inhibits the growth of cancer stem cells and overcame the chemoresistance (12). Given the recent emphasis on the importance of sensitization and improving chemotherapeutic resistance in cancer treatment (19), along with the low toxicity of aripiprazole and previous studies suggesting its role in enhancing resistance, it was hypothesized that it could reduce cisplatin toxicity and exert synergistic anticancer effects when combined with cisplatin. To test this hypothesis, the present study investigated the effects of aripiprazole combined with cisplatin on apoptotic cell death and proliferation in pancreatic cancer cells and elucidated its molecular mechanisms involved.
Materials and methods
Ethics statement
Animal protocols were approved by the INHA Institutional Animal Care and Use Committee (INHA IACUC) at the College of Medicine, Inha University (Incheon, Korea; approval nos. INHA 2211124-848 and INHA 230731-883).
Cell culture
MIA PaCa-2 (cat. no. CRL-1420) and Capan-1 cell lines (cat. no. HTB-79) were obtained from the American Type Culture Collection. MIA PaCa-2 cell lines were cultured in Dulbecco's Modified Eagle's Medium (DMEM; Welgene, Inc.), while Capan-1 cell lines were cultured in Roswell Park Memorial Institute 1640 (RPMI-1640) medium (Gibco; Thermo Fisher Scientific, Inc.). The medium for MIA PaCa-2 was supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin. The medium for Capan-1 was supplemented with 15% fetal bovine serum (FBS) and 1% penicillin/streptomycin. All cell culture reagents, including FBS and penicillin/streptomycin, were purchased from Gibco (Thermo Fisher Scientific, Inc.). The cells were maintained in a CO2 incubator with 95% air and 5% CO2 at 37°C.
Chemicals
Aripiprazole and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were obtained from MilliporeSigma. For in vitro studies, aripiprazole was dissolved in dimethyl sulfoxide (DMSO) at a 20 mM stock concentration and freshly diluted to working concentrations. For in vivo experiments, the desired doses were prepared by dissolving in a DMSO:Cremophor:DW (1:2:7) mixture. Cisplatin was purchased from Selleck Chemicals.
Cell viability and proliferation assay
Cell viability was assessed using the MTT assay. Briefly, MIA PaCa-2 and Capan-1 cells were seeded at densities of 2,500 cells and 5,000 cells per well, respectively, in 96-well plates and treated with aripiprazole and/or cisplatin. After 48 h of incubation, 20 µl of MTT solution (2 mg/ml) was added and incubated for an additional 4 h at 37°C. The plates were read using a microplate reader at a wavelength of 540 nm. The combination index for cisplatin and aripiprazole was calculated using CompuSyn v1.0 (Biosoft), where combination index (CI)<1, CI=1 and CI>1 indicate synergistic, additive and antagonistic effects, respectively (10). Cell proliferation was monitored using the JULI Stage real-time image recording system (NanoEntek).
Colony formation assay
MIA PaCa-2 and Capan-1 cells (1×106) were treated with cisplatin (1 or 10 mM) and/or aripiprazole (10 mM) for 48 h. The treated cells were seeded in six-well plates at densities of 1×103 and 3×103 cells per well with a monolayer and incubated for 14 days. Colonies were washed twice with Dulbecco's phosphate-buffered saline (DPBS), fixed with 4% paraformaldehyde for 15 min at 4°C and stained with 1% crystal violet for 15 min at room temperature.
Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay
The TUNEL assay was conducted using the ApopTag peroxidase in situ apoptosis detection kit (MilliporeSigma). MIA PaCa-2 and Capan-1 cells were treated with cisplatin (1 or 10 µM) and aripiprazole (10 µM) for 48 h, fixed and washed three times with phosphate-buffered saline (PBS). After the terminal deoxynucleotidyl transferase enzyme was activated, the reaction was stopped, and the digoxigenin-conjugated antibody was attached overnight. After washing, 10% 3,3′-diaminobenzidine staining was performed to visualize apoptotic cells and stained for 15 min at room temperature.
Immunofluorescence
The cells were fixed in an acetic acid solution (1:2) for 10 min at 4°C and permeabilized with 0.5% Triton X-100 for 10 min and incubated in CAS block solution (Life Technologies) for 1 h at room temperature. Next, the cells were incubated overnight at 4°C with a primary antibody against cleaved caspase-3 (1:50; cat. no. 9661; Cell Signaling Technology). After several washes with PBS, the cells were incubated with fluorescently labeled secondary antibodies (1:60; cat. no. 31460; Invitrogen) for 2 h at room temperature and stained with 4,6-diamidino-2-phenylindole (Antifade mounting medium with DAPI; Vectashield; Vector Laboratories, Inc.) at a 1:100 dilution for 1 h at room temperature. The cells were viewed using a confocal laser scanning microscope (FluoView 1000; Olympus Corporation) at wavelengths of 488 and 568 nm.
Western blotting
The cells were washed three times with ice-cold DPBS before the lysis. Total cellular proteins were extracted using sterile RIPA lysis buffer (Biosesang; 150 mM sodium chloride, 1% triton X-100, 1% sodium deoxycholate, 0.1% SDS, 50 mM Tris-HCl, pH 7.5, and 2 mM EDTA), 1X Phosphatase inhibitor (GenDEPOT, LLC; sodium fluoride, sodium orthovanadate, sodium pyrophosphate and sodium glycerophosphate) and 1X Protease inhibitor (GenDEPOT, LLC; PMSF; pepstatin A, leupeptin, benzamidine, bestatin). Equal amounts of proteins (50 µg) were separated by 8% or 12% SDS gel electrophoresis using BCA assay and transferred to PVDF membranes (MilliporeSigma). The membranes were blocked with PBS containing 5% skimmed milk for 1 h at room temperature and incubated overnight at 4°C with primary antibodies. After washing, the membranes were incubated with secondary antibodies (1:2,000, Mouse (cat. no. 7076S), Rabbit (cat. no. 7074S); Cell signaling Technology, Inc.) for 1 h at room temperature. Proteins were visualized using Clarity Western ECL Substrate (Amersham Biosciences; Cytiva). Primary antibodies from Cell Signaling Technology, Inc. included cleaved caspase-3 (1:1,000; cat. no. 9661), AKT (1:1,000; cat. no. 9272), phosphorylated (p)-AKT (1:2,000; cat. no. 4060), p-SRC (1:1,000; cat. no. 6943), STAT3 (1:1,000; cat. no. 9139), p-STAT3 (1:1,000; cat. no. 94994) and β-actin (1:10,000; cat. no. 4967). The primary antibody for myeloid cell leukemia-1 (MCL-1, 1:500; cat. no. sc-819) and XIAP (1:500; cat. no. sc-55550) was obtained from Santa Cruz Biotechnology, Inc. The densitometry was quantified using ImageJ software (version 1.54, National Institutes of Health).
Measurement of mitochondrial membrane potential by JC-1 Staining
The cells were treated with cisplatin (1 or 10 µM) and/or aripiprazole (10 µM) for 48 h at 37°C, followed by incubation with JC-1 solution (Cayman Chemical) for 45 min at 37°C. Following staining with DAPI, the slides were washed twice with DPBS, mounted with Antifade Mounting Medium with DAPI (Vectashield; Vector Laboratories, Inc.) and viewed using confocal laser-scanning microscopy (FluoView 1000; Olympus Corporation).
Two-chamber migration and invasion assay
Cell invasion was analyzed using Transwell permeable support systems (Corning Life Sciences). The inserts were coated with 10% Matrigel for 30 min at 37°C before cell seeding. Cells were seeded at a density of 2×104 cells/well in a serum-free medium. The lower chambers were treated with cisplatin (1 or 10 µM) and/or aripiprazole (10 µM) for 48 h at 37°C. After 48 h, the cells that invaded the PET membrane were stained with 0.5% crystal violet for 15 min at room temperature. The migration assay followed a similar protocol, except Matrigel was not used. The purple regions were quantified using ImageJ software (version 1.54; National Institutes of Health).
Fluid shear stress assay
The cells were treated with various concentrations of cisplatin and aripiprazole for 48 h. Cells were then trypsinized and resuspended to a concentration of 1×106 cells/ml in culture medium and subjected to three repeated exposures to shear stress through a 30-gauge needle, followed by a constant flow of 100 µl/s. Briefly, 1×106 cells were resuspended in 4 ml PBS and loaded into a 5 ml plastic syringe. A Luer lock fitting was attached to the end of the syringe and connected to a polyether ether ketone tubing (inner diameter, 125 µm) which was connected to a 50 ml centrifuge tube. The syringe plunger was pushed by a syringe pump to flow the cell solution at flow rates corresponding to wall shear stresses of 5, 20, or 60 dyn/cm2. The syringe was regularly agitated to maintain cell suspension. After shearing, the cells were centrifuged at 300 × g for 3 min at 4°C and resuspended in DMEM/10% FBS. The cells were then incubated for 11 days at 37°C in 96-well ultra-low cluster round-bottom plates (Costar; Corning, Inc.) in a complement medium supplemented with basic fibroblast growth factor, human epidermal growth factor, N-2 and B-27 at 37°C to form three-dimensional (3D) tumor spheroids. After 11 days, the cells were observed under a phase-contrast microscope and captured images. The size and shape of the 3D tumor spheroids were recorded using an inverted light microscope from day 2 to day 11. Additionally, the cells treated for 48 h were seeded at 8×104 cells per well in collagen-coated 48-well plates, incubated for 1 h at 37°C, washed twice with DPBS, fixed with 4% paraformaldehyde and incubated for 1 h at room temperature with a 1:100 dilution of DAPI.
Phospho-kinase array
Relative phosphorylation levels of 43 human protein kinases were determined using a human phospho-kinase array kit (R&D Systems, Inc.). MIA PaCa-2 cells were treated with aripiprazole for 12 h at 37°C and then lysed. After blocking for 1 h at 37°C with Array Buffer 1 (R&D systems, Inc), the membranes were incubated overnight at 4°C with 600 µg of protein lysates, washed and then incubated with a streptavidin-HRP detection antibody for 30 min rocking at room temperature. Membranes were developed using ECL Western blotting detection reagents and protein expression levels were quantified using ImageJ software (version 1.54; National Institutes of Health).
Xenograft animal model
Male BALB/c nude mice (five-weeks-old; 18-20 g) were purchased from Orient Bio, Inc. All animal experiments were conducted in accordance with the guidelines of the INHA IACUC (approval no. INHA 2211124-848). The mice were acclimated for one week and then injected with 3×106 MIA PaCa-2 cells into the flank. When the tumor size reached 50-100 mm3, the mice were randomly assigned to four groups (control, cisplatin, aripiprazole, cisplatin and aripiprazole; n=9). The treatment group received cisplatin [2 mg/kg, intraperitoneally (i.p.)] once a week and/or aripiprazole [10 mg/kg, orally (p.o.)] three times a week. Tumor size was calculated using Vernier calipers with the formula: 0.5x length × (width)2. After 28 days, the tumors were carefully dissected out and fixed in 10% paraformaldehyde at 4°C overnight, embedded in paraffin and sectioned at 3 µm. All animals had ad libitum access to food and water and were maintained in a stable environment at 25±1°C, 60±5% humidity and a 12-h light/dark cycle. The health of the mice was monitored by observing the temperature, humidity, noise and lighting conditions in the animal room by monitoring the weight and general condition of mice to assess the health and behavioral status of the mice. At the end of study, the mice were then anesthetized using Ketamin:Rompun (9:1) mixture and sacrificed by collecting blood sample from the heart. The mice were confirmed dead by no spontaneous breathing for 2-3 min and no blink reflex. The blood, lung, liver, pancreas, spleen and tumors of mice were collected for sample preparation. During the experiment, all mice were anesthetized and sacrificed according to the experimental plan and all measures were taken to decrease the pain of the experimental animals. If the mice showed persistent pain behavior, severe dehydration, inability to eat, extreme fatigue or even severe infection during the research process, they were sacrificed; however, no animals in the present study reached these humane endpoints.
Orthotopic mouse model
Male C57BL/6 mice aged 5 weeks (18-20 g) were purchased from Orient Bio Animal, Inc. All animal experiments were conducted in accordance with the guidelines of the INHA IACUC (approval no. INHA 230731-883). The mice (n=7) were injected with 1×104 KPC cells into the pancreas. After two weeks, the treatment groups received cisplatin (2 mg/kg, i.p.) once a week and/or aripiprazole (10 mg/kg, p.o.) three times a week. After 25 days, the tumors were carefully dissected to avoid contamination from surrounding tissues. At the end of study, the mice were then anesthetized using Ketamin:Rompun (9:1) mixture and sacrificed by collecting blood sample from the heart. The mice were confirmed dead by no spontaneous breathing for 2-3 min and no blink reflex. The blood, lung, liver, pancreas, spleen and tumors of mice were collected for sample preparation. During the experiment, all mice were anesthetized and sacrificed according to the experimental plan and all measures were taken to decrease the pain of the experimental animals. If the mice showed persistent pain behavior, severe dehydration, inability to eat, extreme fatigue or even severe infection during the research process, they were sacrificed; however, no animals in the present study reached these humane endpoints.
Immunohistochemistry
Tumor samples were fixed in 10% buffered formaldehyde at 4°C overnight, embedded in paraffin, and sectioned, Immunostaining was performed on 3-µm sections of tumor samples after deparaffinization. Antigen retrieval was performed by incubation of 0.2 mg/ml Proteinase K (Thermo Fisher Scientific, Inc.) in PBS for 15 min at room temperature. After gently washing twice with PBS, tissue sections were permeabilized with 0.5% Triton X-100 for 10 min and endogenous peroxidase was blocked with 0.3% H2O2 in distilled water for 15 min at room temperature, followed by incubation in CAS block solution (Life Technologies) for 1 h at room temperature. The tissue sections were incubated with primary antibodies (1:50 dilution) following antigen retrieval at 4°C overnight. Subsequently, sections were incubated with a biotinylated secondary antibody (1:60 dilution; cat. no. 31460) for 1 h at room temperature. Primary antibodies from Cell Signaling Technology, Inc. included cleaved caspase-3 (1:50; cat. no. 9661), BCL-2 (1:50; cat. no. 15071), p-STAT3 (1:50; cat. no. 94994). Primary antibody from Abcam included Ki-67 (1:50; cat. no. ab16667). Immunoreactive proteins were detected by incubating sections with an avidin-biotin peroxidase complex solution ABC kit (Vector Laboratories, Inc.). After washing with PBS, proteins were visualized by incubating sections with DAB for 15 min at room temperature, followed by counterstaining with hematoxylin solution for 40 sec at room temperature. At least three random fields of each section were examined at ×400 magnification and analyzed using a computer image analysis system (Olympus Corporation).
Statistical Analysis
All experiments were repeated three times. The data were analyzed using one-way ANOVA with Tukey's post hoc tests for multiple comparisons. Results were presented as mean ± standard deviation (SD). All analyses were performed with GraphPad Prism (version 8.00, GraphPad Software, Inc.; Dotmatics). P<0.05 was considered to indicate a statistically significant difference.
Results
Aripiprazole enhances the anticancer efficacy of cisplatin in PDAC cells
To identify the synergistic effects of cisplatin and aripiprazole, PDAC cells (MIA PaCa-2 and Capan-1) were treated with both drugs for 48 h. The co-treatment exhibited significant synergistic effects, with CI<1 for the combination of 10 µM cisplatin and 10 µM aripiprazole in MIA PaCa-2 cells (CI=0.884) and 1 µM cisplatin and 10 µM aripiprazole in Capan-1 cells (CI=0.867; Fig. 1A). The cell proliferation curves showed similar trends (Fig. 1B). Additionally, the clonogenic survival of PDAC cells treated with this combination was inhibited in a dose-dependent manner compared with single-agent treatments (Fig. 1C). These findings indicated that aripiprazole enhanced the anticancer efficacy of cisplatin and synergistically inhibits the proliferation of PDAC cells.
Induction of apoptosis in PDAC cells by combination treatment of cisplatin and aripiprazole
Since the co-treatment significantly reduced cell proliferation, it was investigated whether it could induce apoptosis in PDAC cells. TUNEL staining revealed an increase in the percentage of TUNEL-positive cells following treatment (Fig. 2A and B). Furthermore, the combination treatment significantly increased the expression levels of cleaved caspase-3 (Fig. 2C and D). These apoptotic effects were confirmed by the increase in cleaved caspase-3 compared with single-agent treatments, as demonstrated by western blotting (Fig. 2E). Taken together, these results indicated that the combination of cisplatin and aripiprazole synergistically induced apoptosis in pancreatic cancer cells.
Combining of cisplatin and aripiprazole synergistically induces mitochondria-mediated apoptosis
Given that the combined treatment of cisplatin and aripiprazole markedly increased apoptotic cell death, the present study next investigated whether it affects the mitochondrial membrane potential (MMP), which is associated with apoptosis. MMP was examined using JC-1 staining. The control cells exhibited strong red fluorescence in the cytoplasm, whereas the combined treatment showed a clear decrease in red fluorescence and an increase in green fluorescence, indicating changes in MMP associated with apoptosis (Fig. 3A and B). Additionally, the combination treatment decreased the expression of XIAP and MCL-1, prominent anti-apoptotic proteins (Fig. 3C). The results suggested that the synergistic effects of aripiprazole and cisplatin are mediated by mitochondria-mediated apoptosis in PDAC cells.
Combining cisplatin and aripiprazole inhibits cell migration and invasion
Cancer cell migration and invasion are critical steps in metastasis. To investigate the potential inhibitory effects of co-treatment with aripiprazole and cisplatin on these processes, migration and invasion assays were conducted using MIA PaCa-2 and Capan-1 cell lines. Transwell assays demonstrated that the combination of aripiprazole and cisplatin synergistically inhibited both cell migration and invasion compared with the effects of either agent alone (Fig. 4A and B). Additionally, cell spheroid growth and attachment to collagen-coated plates were significantly diminished by the combined treatment, in contrast to the effects of single-agent treatments (Fig. 4C and D).
Aripiprazole inhibits the STAT3 pathway
Among the 43 kinases analyzed, aripiprazole inhibited STAT3 expression by 40% (Fig. 5A). To validate these findings, cells were treated with aripiprazole (10 µM) for 12 h, followed by western blot analysis to examine the expression of p-STAT3, p-AKT and p-SRC. As shown in Fig. 5B, aripiprazole decreased p-STAT3 expression, and the combination of aripiprazole and cisplatin almost completely abolished its expression in MIA PaCa-2 cells. These findings suggested that the combination treatment could induce pancreatic cancer cell death by inhibiting STAT3 signaling.
Aripiprazole inhibits tumor growth in mouse pancreatic cancer models
Tumor growth was delayed in mice treated with either aripiprazole or cisplatin alone compared with the control group. Notably, the combination treatment significantly reduced tumor volume by 78% relative to the control (Fig. 6A). These results were corroborated using an orthotopic pancreatic cancer model (Fig. 6B). All treatments were well-tolerated, with no significant differences between groups. Tumor formation incidence was >95%. Treatment with aripiprazole or cisplatin inhibited primary pancreatic tumor growth by 33 and 23%, respectively, compared with the control. The combination treatment markedly enhanced the anti-tumor effect, with a 73% growth inhibition observed compared with control. Furthermore, the combined treatment reduced the expression of Ki-67 and Bcl-2, markers of cell proliferation and apoptosis, respectively and increased the expression of cleaved caspase-3 and the number of TUNEL-positive cells. The combination treatment also significantly reduced p-STAT3 expression (Fig. 6C).
Discussion
Of patients with pancreatic cancer, ~80% are diagnosed at a locally advanced or metastatic stage, where combination chemotherapy is generally preferred over single-agent chemotherapy. However, the effectiveness of these treatments remains limited, prompting increased interest in combination therapies with fewer side effects and higher efficacy. Currently, cisplatin is used to treat various solid tumors, including PDAC. Despite its usefulness, cisplatin has shown severe side effects drug resistance, potentially contributing to treatment failure in PDAC (4,20). Nevertheless, cisplatin continues to be evaluated in clinical trials for its potential in combination chemotherapy for PDAC. Therefore, combination treatment with sensitizing agents that have fewer side effects may be an effective strategy for improving anticancer efficacy and overcoming cisplatin resistance. The present study found that the combination of aripiprazole and cisplatin synergistically induced apoptosis and inhibited cell growth by blocking STAT3 signaling. Moreover, this combination significantly reduced tumor growth in animal models without notable side effects, suggesting that the combination of aripiprazole and cisplatin could enhance PDAC treatment outcomes.
Aripiprazole, an atypical antipsychotic, which is effective in treating schizophrenia and schizoaffective disorders, exhibits unique pharmacological activities as a serotonin 5-HT1A and 5-HT2A antagonist. During tumor development, cancer not only evades the body's regulatory mechanisms, but also affects local and systemic homeostasis. It has been shown in human and animal cancer models that tumors affect the production of classic neurotransmitters such as hypothalamic and pituitary hormones (21). Indeed, the neurotransmitter regulator aripiprazole shows anti-cancer effects across different types of cancer cells (13,14). This drug repositioning strategy has recently demonstrated potent radiosensitizing activity in various cancer cells (18,22). Therefore, it was hypothesized that combining cisplatin with aripiprazole could result in synergistic effects in PDAC cells. The combined treatment significantly inhibited PDAC cell growth compared with individual agent treatments. Cotreatment with cisplatin and aripiprazole exhibited the highest synergistic effects in PDAC cells, as indicated by the CI values. The present study investigated whether this combination induced synergistic apoptotic effects. Indeed, it led to significant apoptosis, evidenced by increased TUNEL-positive nuclear fragmentation and elevated cleaved caspase-3 expression. While studies have confirmed the apoptotic effect of aripiprazole in combination with other antipsychotic drugs such as fluphenazine, halaven and thioridazine in cancer (14,18), few have explored its combination with anticancer drugs for clinical use. Therefore, the apoptotic effect observed with aripiprazole combined with cisplatin appears to be substantial.
Apoptosis occurs through two distinct pathways: The extrinsic pathway, activated by death receptors, and the intrinsic pathway, involving mitochondrial factors. Given reports that the combination of cisplatin with other agents induces the intrinsic mitochondrial apoptotic pathway (23,24), the present study extended its analysis to investigate whether aripiprazole affected mitochondrial membrane potential when combined with cisplatin, using JC-1 staining. As expected, the combined treatment synergistically reduced mitochondrial membrane potential. To validate these findings, the expression of XIAP and MCL-1, members of the inhibitor of apoptosis proteins (IAPs) family crucial in mitochondria-mediated apoptosis, were examined. The analyses demonstrated a significant reduction in XIAP and MCL-1 expression alongside increased levels of cleaved caspase-3 in PDAC cells following combined treatment. Overall, the results suggested that the synergistic apoptotic effects of this combination therapy may operate through the mitochondria-mediated apoptotic pathway in PDAC cells, potentially contributing to the inhibition of tumor growth in animal models.
To date, aripiprazole has been reported to modulate the cAMP/PKA, ERK/C-Fos and AKT/GSK3β signaling pathways in brain diseases (14,25,26). Studies have indicated that aripiprazole enhances cancer cell sensitivity to ionizing radiation by increasing reactive oxygen species production (17) and that SRC serves as a primary target for aripiprazole's anti-tumor activity in glioma cells (13). However, there has been limited research exploring the precise mechanisms by which aripiprazole affects cancer signaling pathways. To further investigate the potential mechanism underlying the anticancer effects of aripiprazole, a phosphokinase array analysis was conducted. The present study demonstrated that aripiprazole effectively inhibited STAT3 phosphorylation. Notably, contrary to previous reports (13,27), increased expression of AKT and SRC was observed in response to aripiprazole in pancreatic cancer cells, suggesting differential effects on AKT and SRC signaling pathways. Given that STAT3 is frequently overexpressed in tumors and its increased signaling promotes cancer cell survival and chemoresistance, the inhibition of p-STAT3 expression by the combination of aripiprazole and cisplatin, mediated by aripiprazole, holds significant therapeutic promise.
In conclusion, the present study is the first, to the best of the authors' knowledge, to demonstrate that combined treatment of aripiprazole and cisplatin markedly inhibited PDAC cell growth and synergistically exhibited anticancer activities by suppressing cell proliferation and inducing apoptosis both in vitro and in vivo via STAT3 pathway inhibition (Fig. 7). Aripiprazole is an FDA approved antidepressant currently in clinical use; therefore, it is expected to be useful as an excellent adjuvant that can markedly increase the efficacy of cisplatin therapy for pancreatic cancer. These findings suggested that the combination of aripiprazole and cisplatin represents an innovative therapeutic approach for pancreatic cancer in humans.
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
YJC performed all the experiments with the assistance of BSH, SK, MSP, YJL, SEK, PL and HGG. HL, SK, SP and ERP interpreted the results. YJC, KHJ and SH wrote the manuscript. SSH and KHJ contributed to the design of the study and assembled data. SSH and KHJ confirmed the authenticity of all the raw data. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Animal protocols were approved by the INHA Institutional Animal Care and Use Committee (approval no. INHA IACUC) at the College of Medicine, Inha University (Incheon, Korea; approval nos. INHA 2211124-848 and INHA 230731-883).
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Use of artificial intelligence tools
During the preparation of this work, the authors used DeepL (www.deepl.com) to assist in translating the original text into English to check for grammatical errors and confirm alternative expressions. After using this tool, the authors reviewed and edited the content as necessary and therefore they take full responsibility for the ultimate content of the present manuscript.
Acknowledgments
Not applicable.
Funding
The present study was supported by the Basic Science Research Program through the National Research Foundation of Korea (grant nos. 2021R1A5A2031612, 2024M3A9J4006509 and 2022R1A2C1092933) and the Technology development Program (RS-2022-TI023745) funded by the Ministry of SMEs and Startups (MSS; Korea).
References
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, et al: The molecular biology of pancreatic adenocarcinoma: Translational challenges and clinical perspectives. Sig Transduct Target Ther. 6:2492021. View Article : Google Scholar | |
Muaddi H, Kearse L and Warner S: Multimodal approaches to patient selection for pancreas cancer surgery. Curr Oncol. 31:2260–2273. 2024. View Article : Google Scholar : PubMed/NCBI | |
Awais N, Satnarine T, Ahmed A, Haq A, Patel D, Hernandez GN, Seffah KD, Zaman MA and Khan S: A systematic review of chemotherapeutic regimens used in pancreatic cancer. Cureus. 15:e466302023.PubMed/NCBI | |
Du J, Wang X, Li Y, Ren X, Zhou Y, Hu W, Zhou C, Jing Q, Yang C, Wang L, et al: DHA exhibits synergistic therapeutic efficacy with cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism. Cell Death Dis. 12:7052021. View Article : Google Scholar : PubMed/NCBI | |
Ouyang G, Liu Z, Huang S, Li Q, Xiong L, Miao X and Wen Y: Gemcitabine plus cisplatin versus gemcitabine alone in the treatment of pancreatic cancer: A meta-analysis. World J Surg Onc. 14:592016. View Article : Google Scholar | |
Principe DR, Underwood PW, Korc M, Trevino JG, Munshi HG and Rana A: The current treatment paradigm for pancreatic ductal adenocarcinoma and barriers to therapeutic efficacy. Front Oncol. 11:6883772021. View Article : Google Scholar : PubMed/NCBI | |
Santos NAGD, Ferreira RS and Santos ACD: Overview of cisplatin-induced neurotoxicity and ototoxicity, and the protective agents. Food Chem Toxicol. 136:1110792020. View Article : Google Scholar : PubMed/NCBI | |
Low ZY, Farouk IA and Lal SK: Drug repositioning: New approaches and future prospects for life-debilitating diseases and the COVID-19 pandemic outbreak. Viruses. 12:10582020. View Article : Google Scholar : PubMed/NCBI | |
Islam S, Wang S, Bowden N, Martin J and Head R: Repurposing existing therapeutics, its importance in oncology drug development: Kinases as a potential target. Br J Clin Pharmacol. 88:64–74. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shankar E, Subramaniam V and Allimuthu D: Editorial: Adopting drug repurposing to overcome drug resistance in cancer. Front Cell Dev Biol. 11:11916822023. View Article : Google Scholar : PubMed/NCBI | |
Stępnicki P, Kondej M and Kaczor AA: Current concepts and treatments of schizophrenia. Molecules. 23:20872018. View Article : Google Scholar : PubMed/NCBI | |
Suzuki S, Okada M, Kuramoto K, Takeda H, Sakaki H, Watarai H, Sanomachi T, Seino S, Yoshioka T and Kitanaka C: Aripiprazole, an antipsychotic and partial dopamine agonist, inhibits cancer stem cells and reverses chemoresistance. Anticancer Res. 36:5153–5161. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kim MS, Yoo BC, Yang WS, Han SY, Jeong D, Song JM, Kim KH, Aravinthan A, Kim JH, Kim JH, et al: Src is the primary target of aripiprazole, an atypical antipsychotic drug, in its anti-tumor action. Oncotarget. 9:5979–5992. 2017. View Article : Google Scholar | |
Kim JY, Tae IH, Lee BM, Kim HS and Yoon SP: Low doses of the anti-psychotic drug aripiprazole have strong P-gp-inhibitory activity and sensitize anti-mitotic drug-resistant cancer cells. Anticancer Res. 38:5101–5108. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhuo C, Xun Z, Hou W, Ji F, Lin X, Tian H, Zheng W, Chen M, Liu C, Wang W and Chen C: Surprising anticancer activities of psychiatric medications: Old drugs offer new hope for patients with brain cancer. Front Pharmacol. 10:12622019. View Article : Google Scholar : PubMed/NCBI | |
Badran A, Wahab AT, Zafar H, Mohammad N, Imad R, Khan MA, Baydoun E and Choudhary MI: Antipsychotics drug aripiprazole as a lead against breast cancer cell line (MCF-7) in vitro. PLoS One. 15:e02356762020. View Article : Google Scholar : PubMed/NCBI | |
Jiang C, Lee SH, Park JH, Lee JS, Park JW, Kim JR, Lee SH, Kim HS and Yoon SP: A low dose of aripiprazole has the strongest sensitization effect among 19 repositioned bipolar drugs in P-gp-overexpressing drug-resistant cancer cells. Anticancer Res. 41:687–697. 2021. View Article : Google Scholar : PubMed/NCBI | |
Lee HJ, Kang SM, Sonn JK and Lim YB: Dopamine receptor D2 activation suppresses the radiosensitizing effect of aripiprazole via activation of AMPK. FEBS Open Bio. 9:1580–1588. 2019. View Article : Google Scholar : PubMed/NCBI | |
Singh P, Patel M, Bhowmik D, Kumari N, Prajapati SK and Gupta R: Identification of common biomarkers affecting patient survival in cancers. World Acad Sci J. 6:1–17. 2024. View Article : Google Scholar | |
de Oliveira G, Freire PP, Cury SS, de Moraes D, Oliveira JS, Dal-Pai-Silva M, Reis PP and Carvalho RF: An integrated meta-analysis of secretome and proteome identify potential biomarkers of pancreatic ductal adenocarcinoma. Cancers (Basel). 12:7162020. View Article : Google Scholar : PubMed/NCBI | |
Slominski RM, Ramen C, Chen JY and Slominski AT: How cancer hijacks the body's homeostasis through the neuroendocrine system. Trends Neurosci. 4:263–275. 2023. View Article : Google Scholar | |
Jeong HJ, Jung CW, Kim HJ, Park BH, Moon Y, Kim JY and Park MJ: Aripiprazole sensitizes head and neck cancer cells to ionizing radiation by enhancing the production of reactive oxygen species. Pharmacol Res Perspect. 10:e009892022. View Article : Google Scholar : PubMed/NCBI | |
Rodrigues T and Ferraz L: Therapeutic potential of targeting mitochondrial dynamics in cancer. Biochem Pharmacol. 182:1142822020. View Article : Google Scholar : PubMed/NCBI | |
Li X, Guo S, Xiong XK, Peng BY, Huang JM, Chen MF, Wang FY and Wang JN: Combination of quercetin and cisplatin enhances apoptosis in OSCC cells by downregulating xIAP through the NF-κB pathway. J Cancer. 10:4509–4521. 2019. View Article : Google Scholar : | |
Pan B, Chen J, Lian J, Huang XF and Deng C: Unique effects of acute aripiprazole treatment on the dopamine D2 receptor downstream cAMP-PKA and Akt-GSK3β signalling pathways in rats. PLoS One. 10:e01327222015. View Article : Google Scholar | |
Pereira A, Zhang B, Malcolm P, Sugiharto-Winarno A and Sundram S: Quetiapine and aripiprazole signal differently to ERK, p90RSK and c-Fos in mouse frontal cortex and striatum: role of the EGF receptor. BMC Neurosci. 15:302014. View Article : Google Scholar : PubMed/NCBI | |
Pan B, Huang XF and Deng C: Aripiprazole and haloperidol activate GSK3β-dependent signalling pathway differentially in various brain regions of rats. Int J Mol Sci. 17:4592016. View Article : Google Scholar |