1. Introduction
Cancer remains one of the leading causes of death worldwide and requires continuous improvement in treatment strategies [
1]. A hallmark of cancer is aberrant oncogenic signaling triggered by both genetic and non-genetic alterations [
2]. Novel targeted therapies that selectively inhibit activated oncogenic signaling pathways have led to more effective treatments and better outcomes [
3]. Unfortunately, these targeted therapies in advanced cancers often do not result in a long-lasting response due to the emergence of resistance. Secondary mutations that lead to reactivation of the oncogenic pathway in the presence of the drug result in resistance to targeted therapies [
4,
5]. This underscores the need for alternative approaches to effectively combat cancer that are fundamentally different from inhibiting oncogenic signaling.
Recent studies demonstrate that hyperactivation of oncogenic signaling pathways can be as lethal to cancer cells as inhibition of these pathways [
6,
7]. Indeed, oncogenic signaling in cancer cells is associated with increased activation of stress response pathways that enable cells to cope with the stress caused by oncogenic activity [
8]. As a result, further activation of mitogenic signaling could destabilize the fragile homeostasis of cancer cells and overload the stress response pathways [
9].
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that plays an important role in the progression of mitosis and the cellular response to DNA damage [
10]. Due to its ability to control various oncogenic signaling pathways, PP2A has been recognized as a tumor suppressor, leading to the development of drugs designed to reactivate its activity [
11]. However, inhibition of PP2A has gained interest as a strategy to target tumors that are resistant to conventional treatments [
12]. Namely, the proper function of PP2A during the G2 phase of the cell cycle is essential for cell senescence [
13]. Cancer cell senescence is a major contributor to resistance to therapy, especially to conventional DNA-damaging agents that preferentially affect actively dividing cells [
14,
15]. Recent studies have shown that inhibition of PP2A can help drive senescent cancer cells into mitosis, leading to cell death by mitotic catastrophe [
16,
17].
LB-100, a small molecule inhibitor of PP2A, induces mitotic catastrophe in several tumor models, especially in combination with DNA-damaging agents [
18]. A recent study has shown that LB-100 enhances oncogenic signaling and activates stress response pathways [
16]. Lu et al. discovered that LB100 caused an inappropriate entry into mitosis and increased cell death when combined with doxorubicin in glioblastoma cells [
19]. In vivo, the combination therapy showed a better response, exhibiting either growth inhibition or regression, than each agent used alone. In addition, the combination of LB-100 with doxorubicin effectively prevented tumor recurrence [
19]. It has also been shown that simultaneous inhibition of PP2A by LB-100 and WEE1 kinase by adavosertib is highly lethal in several cancer models, leading to DNA replication breakdown and triggering premature mitosis with subsequent cell death [
16]. Importantly, in a phase I clinical trial, LB-100 demonstrated a favorable toxicity profile at doses associated with clinical response [
20]. These properties of LB-100 make it an attractive candidate to test the concept of activating oncogenic signaling for cancer treatment.
LB-100 is an emerging pharmaceutical agent with significant potential for overcoming multidrug resistance (MDR) in cancer treatment. Its unique mechanism enables the sensitization of cancer cells to conventional therapies, including radio and chemotherapy [
21]. Research has demonstrated that LB-100 can effectively reverse resistance to cisplatin in models of ovarian carcinoma and medulloblastoma [
20]. Furthermore, its efficacy in combination with immunotherapy underscores its potential in diverse treatment strategies [
22]. The combination approaches with LB-100 target both primary tumors and address emerging resistance mechanisms. Due to its efficacy and safety profile in combination therapies [
21], LB-100 is a promising option for combating MDR in cancer.
Glioblastoma (GBM) is the most common primary brain tumor and accounts for about 80% of all tumors of the central nervous system [
23]. GBM represents a major challenge in the field of oncology due to its aggressive nature and resistance to conventional therapies [
24]. In addition, the low biodistribution of most cancer therapeutics in the brain due to the high expression of P-glycoprotein (P-gp) in the blood–brain barrier (BBB) is one of the limitations in GBM treatment. P-gp or the ATP-binding cassette (ABC) transporter (ABCB1) prevents cellular uptake of a variety of anticancer drugs, leading to cancer drug resistance [
25]. Standard treatment options for GBM include maximal surgical resection followed by radiotherapy with concomitant and adjuvant temozolomide administration [
26]. Although the standard treatment for GBM is associated with poor response rates, it has remained essentially unchanged over the past decades due to the lack of therapeutic alternatives [
27]. As no therapy shows a promising response, there is an urgent need for innovative therapeutic approaches for GBM.
Using glioblastoma cells, patient-derived cells and pairs of sensitive and MDR cancer cell lines, we investigated the anticancer effect of LB-100 in combination with adavosertib, an inhibitor of WEE1 kinase, and doxorubicin, an inhibitor of topoisomerase II. These drugs were selected for their ability to target stress response pathways, particularly DNA damage stress, by either inducing DNA damage stress (doxorubicin) or inhibiting the response to DNA damage (adavosertib). The combined treatment of LB-100 with adavosertib or doxorubicin was additionally investigated in non-small cell lung carcinoma cells. For the first time, we report that LB-100 synergizes with doxorubicin in simultaneous and subsequent treatment by inhibiting P-gp expression.
2. Materials and Methods
2.1. Compounds
LB-100 and adavosertib were purchased from Selleckchem, Houston, TX, USA. Doxorubicin (DOX) was obtained from Sigma–Aldrich Chemie GmbH, Hamburg, Germany. LB-100 was diluted in sterile deionized water and kept as 10 mM aliquots at −20 °C. Adavosertib and DOX were dissolved as 10 mM stocks in dimethyl sulfoxide (DMSO) and kept as aliquots at −20 °C. Prior to administration, the compounds were dissolved in sterile deionized water.
2.2. Chemicals and Reagents
The following chemicals and reagents were used in the experimental work: minimum essential medium (MEM), Ham’s F12 growth media, Dulbecco’s modified minimal essential medium (DMEM), L-glutamine, hydrocortisone, insulin, adenine, DMSO, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT), Hoechst 33342 (Sigma–Aldrich Chemie GmbH, Taufkirchen, Germany), a mixture of antibiotics penicillin-streptomycin, a mixture of antibiotics and antimycotics: penicillin, streptomycin, amphotericin B, MEM non-essential amino acids (Capricorn Scientific GmbH, Ebsdorfergrund, Germany), trypsin/EDTA (Gibco, Thermo Fisher Scientific, Waltham, MA, USA), RPMI 1640 medium, fetal bovine serum (FBS) (Corning, NY, USA), propidium iodide (PI) (Molecular Probes, Thermo Fisher Scientific, USA), ribonuclease A (Invitrogen, Thermo Fisher Scientific, USA), Anti-phospho-histone H2A.X (Ser139) rabbit (Cell Signaling Technology®, USA), Tumor Dissociation Kit (Miltenyi Biotec, Bergisch Gladbach, Germany), epidermal growth factor (BioLegend, San Diego, CA, USA), anti-cytokeratin 8/18 (CK8/18) primary antibody cocktail (clone SU0338, #MA5-32118), anti-ABCB1 monoclonal antibody (clone C219, #MA1-26528), isotype control IgG2bk (Abcam, Cambridge, UK), Alexa Fluor 555 goat anti-mouse secondary antibody (#A-21422), Alexa Fluor 488 goat anti-rabbit secondary antibody (#A-11008) (Thermo Fisher Scientific, Waltham, MA, USA), polyclonal rabbit anti-glial fibrillary acidic protein antibody (anti-GFAP, Z0334) (Agilent Dako, Santa Clara, CA, USA) and PE-conjugated anti-P-gp antibody (BD Biosciences, Plymouth, UK).
2.3. Cells and Cell Culture
Glioblastoma cell line U87 and human non-small cell lung carcinoma cells (NSCLCs) NCI-H460 were purchased from the American Type Culture Collection (Rockville, MD, USA). Multidrug-resistant (MDR) U87-TxR cell line was selected from U87 by continuous exposure to the stepwise increasing concentrations of paclitaxel during nine months [
28], while MDR NCI-H460/R cells were selected from NCI-H460 cells by continuous exposure to the stepwise increasing concentrations of DOX during three months [
29]. Both MDR models are well-characterized and detailed in several publications [
30,
31,
32]. A key characteristic contributing to the MDR phenotype is the overexpression of P-glycoprotein (P-gp). U87 and U87-TxR were cultivated in MEM, supplemented with 10% FBS, 2 mM L-glutamine, 5000 U/mL penicillin and 5 mg/mL streptomycin mixture, and 1% non-essential amino acids. NCI-H460 and NCI-H460/R were grown in RPMI 1640 medium, supplemented with 10% FBS, 2 mM L-glutamine, and 10,000 U/mL penicillin, 10 mg/mL streptomycin, 25 mg/mL amphotericin B solutions. All cell lines were grown at 37 °C in a humid atmosphere with 5% CO
2. The cells were grown in 25 cm
2 flasks (Sarstedt, Nümbrecht, Germany) until they reached 80–90% confluence. Then, cell passage was carried out using 0.25% trypsin/EDTA. The cells were counted using a Bürker–Türk hemocytometer on an inverted microscope. Once counted, the cells were seeded at the following densities for further experimentation or maintenance in culture: 8000 cells/ cm
2 for NCI-H460 and NCI-H460/R, and 16,000 cells/cm
2 for U87 and U87-TxR.
2.4. Patient-Derived Cell Cultures
A sample DT7 from patient with WHO grade 4, IDH-wildtype glioblastoma was collected from the Clinic for Neurosurgery at the Clinical Center of Serbia after obtaining the patient’s informed consent and the approval from the Ethics Committee of the Clinical Center of Serbia (ref. number 187/13). The histological grade was determined by correlating neuroradiological examination and histopathological analysis of the surgical specimen. The expression of GFAP, OLIG2, MAP2, vimentin and p53 were confirmed by histopathological analysis. TR159, an NSCLC sample, was obtained from the Clinic for Thoracic Surgery at the Clinical Center of Serbia after obtaining informed consent from the patients and approval from the Ethics Committee of the Clinical Center of Serbia (ref. number 623/4). The sample was collected during surgery, and histopathologic analysis determined NSCLC diagnosis, histologic grade, stage, necrosis and lymph node invasion status. The histological stage of the collected NSCLC sample is IIA. The surgical specimens DT7 and TR159 were placed in a sterile tube containing antibiotic-antimycotic solution and immediately transported to the research laboratory for further processing.
To establish primary cultures, the tissue was manually minced using a surgical blade in a sterile Petri dish immediately upon arrival at the laboratory. The samples were then sectioned into pieces measuring 3–5 mm and dissociated with the Tumor Dissociation Kit (Miltenyi Biotec, Bergisch Gladbach, Germany) as per the manufacturer’s protocol. The tissue was incubated on a 37 °C orbital shaker (KS 4000 ic control, IKA, Königswinter, Germany) at 300 rpm for 90 min. Following incubation, the dissociated material was transferred to DMEM/Ham’s F12 medium (1:3 ratio) enriched with 5% FBS, antibiotic-antimycotic solution, 4 μg/mL hydrocortisone, 1 μg/mL insulin, 10 ng/mL epidermal growth factor and 24 μg/mL adenine. The tissue was cultured in 25 cm2 cell culture flasks, with the medium replaced after cell attachment was observed. Successfully established patient-derived cultures were maintained in a 37 °C humidified atmosphere with 5% CO2 and grown to confluence before experiments.
2.5. Real-Time Primary Culture Cells Growth Monitoring
Growth of DT7 and TR159 primary culture cells was monitored in real time using the xCELLigence system (Roche, Basel, Switzerland) with a 96-well E-plate. To establish a baseline, 100 µL of medium was added to each well, and the background was measured before seeding the cells. The cells were then seeded, and the total volume in each well was adjusted to 200 µL with media. Twenty-four hours after seeding, cells were treated with 250 and 500 nM DOX and adavosertib, either individually or in combination with 2.5 µM and 5.0 µM LB-100. Drug treatments were refreshed every 2 to 3 days for a total of 10 days. After the treatment period, the TR159 cells were allowed to recover for another 5 days. The xCELLigence system automatically recorded impedance values for each well every 30 min and displayed the results as a cell index (CI). Antiproliferative effects are characterized by a reduced growth rate while maintaining a similar growth trajectory to untreated cells. Cytostatic effects are identified by a growth profile that remains consistent with the initial seeding baseline, whereas cytotoxic effects refer to a decline in the growth profile below the baseline established during seeding.
2.6. Immunofluorescence Assay
The immunofluorescence assay was optimized to distinguish cancer cells from stromal cells using GFAP antibody and CK8/18 antibody mixture cocktail in glioblastoma and NSCLC primary cell cultures, respectively. In the glioblastoma primary cell culture, GFAP-positive cells were considered as glioblastoma cells. In the NSCLC primary cell culture, CK8/18-positive cells were considered as cancer cells. The immunofluorescence assay was additionally optimized to quantify P-gp expression using ABCB1 antibody. Patient-derived cells were seeded in black, clear-bottom 384-well cell culture microplates (Thermo Fisher Scientific, Waltham, MA, USA) in 50 µL of cell growth medium at a density of 4000 cells per well for glioblastoma primary cells and 1000 cells per well for NSCLC primary cells. The glioblastoma primary cells were treated 24 h after seeding with 250, 500, 1000 and 2500 nM adavosertib and DOX alone or in combination with 0.5, 1.0 and 2.5 µM LB-100 for 5 days. NSCLC primary cells were treated with the same concentrations of adavosertib and DOX alone or in combinations with 250, 500 and 1000 nM LB-100 for 7 days. After the treatments, the cells were fixed in 4% paraformaldehyde for 20 min at RT and washed using the Wellwash™ Versa microplate washer (Thermo Fisher Scientific, Waltham, MA, USA). The cells were then blocked with 2% bovine serum albumin (BSA) in PBS for 1 h at RT. After blocking, the glioblastoma and NSCLC primary cells were incubated overnight at 4 °C with a primary rabbit GFAP antibody and primary rabbit CK8/18 antibody cocktail, respectively. Both primary culture cells were co-immunostained with mouse antibody against ABCB1 to detect the presence of P-gp in both the cancer and stromal cells. The cells were washed three times with PBS using the microplate washer before incubation with the secondary Alexa Fluor 555 goat anti-mouse antibody and the secondary Alexa Fluor 488 goat anti-rabbit antibody at RT for 2 h in the dark. The cell nuclei were counterstained with 1 µg/mL Hoechst 33342 at RT for 2 h. The cells were kept at 4 °C in the dark until imaging. Fluorescently labeled cells were visualized using the ImageXpress Pico Automated Cell Imaging System (Molecular Devices, San Jose, CA, USA) with a 4× objective lens, after determining optimal exposure times for each illumination filter (
Figure S1). Image analysis was conducted using CellReporterXpress software v. 2.8.2.669 (Molecular Devices). Cytotoxicity of the compounds was assessed using the Cell Scoring Analysis Protocol, as previously described [
33]. The Multi-Wavelength Cell Scoring Analysis Protocol was used to evaluate ABCB1 expression [
33].
2.7. Drug Interaction Assessment
SynergyFinder+ is an advanced online tool used to analyze interactions between drugs, specifically LB-100 combined with adavosertib and LB-100 combined with DOX, to identify potential synergies. We collected dose–response data from immunofluorescence assays performed on DT7 and TR159 patient-derived cells for various drug combinations. These data were organized into a matrix, and SynergyFinder+ utilized several models to evaluate the synergies between the drugs. The Zero Interaction Potency (ZIP) model enhances the analysis by comparing the drugs’ dose–response curves. It assumes that there is no interaction between the drugs and calculates the expected effect accordingly. Any deviations from this expected effect are interpreted as a measure of synergy [
34]. Next, the Bliss Independence model assesses the scenario where the drugs act independently. It estimates the expected effect based on the probability of unrelated events occurring simultaneously. If the observed effects of the combination exceed this expected value, it indicates a synergistic interaction [
34]. Additionally, the Loewe Additivity model is based on the principle that drugs in combination behave like a single drug administered at varying doses. This model helps to determine whether the combined doses result in an additive effect or provide evidence of synergy [
34]. Finally, the Highest Single Agent (HSA) model compares the effects of the drug combination to the maximum effects of the individual drugs. If the combination’s effect surpasses that of the most effective single agent, it suggests a synergistic interaction [
34]. For all the four models, observing synergism is indicated by a positive value above zero.
2.8. MTT Assay
The combined effects of LB-100 with DOX were studied in MDR U87-TxR glioblastoma and NCI-H460/R NSCLC cell lines by the MTT assay. The assay works by measuring the metabolic activity of the cell, which indirectly measures the viability of living cells. The cells were seeded in 96-well microtiter plates at the density of 2000 cells per well for NCI-H460/R and 4000 cells per well for U87-TxR in 100 μL of the appropriate medium. In simultaneous treatments that lasted 72 h, two concentrations of LB-100 (2.5 and 5 µM) were combined with DOX (100, 250, 500, 1000 and 2500 nM) in MDR glioblastoma cells, as well as with DOX (50, 100, 250, 500 and 1000 nM) in MDR NSCLCs. In subsequent treatment, both MDR cancer cell lines were pre-treated with 2.5 µM and 5 µM LB-100. After 24 h, the cells were treated with increasing concentrations of DOX in the same concentrations range like in simultaneous treatments. The experiment was carried out for further 48 h. At the end of the treatment period, MTT was introduced to each well at a final concentration of 0.2 mg/mL, and the plates were stored for 3 h at 37 °C in a 5% CO2 environment. The formazan crystals formed in the cells with functioning mitochondria were dissolved in 100 µL of DMSO, and absorbance readings were taken at 570 nm with a reference wavelength of 690 nm using the Multiskan Sky microplate reader (Thermo Scientific, Waltham, MA, USA). The IC50 value, representing the drug concentration required to inhibit cell growth by 50%, was calculated through non-linear regression analysis using the log (inhibitor) vs. normalized response model in GraphPad Prism 8.0.2 software.
2.9. Flow Cytometric Detection of P-gp Expression
The P-gp expression levels in U87-TxR and NCI-H460/R cells were measured by flow cytometry. The cells were seeded in adherent 6-well plates, incubated overnight and treated with 2.5 µM and 5.0 µM LB-100. After 24 h, 48 h and 72 h, cells were collected by trypsinization, washed in PBS and directly immunostained by PE-conjugated anti-P-gp antibody according to the manufacturer’s protocol. In all experiments, unstained untreated sensitive (U87 and NCI-H460) cancer cells were utilized as negative controls. Each experiment also incorporated an isotype control IgG2bk to assess the background fluorescence levels. The fluorescence intensity was measured on a CytoFLEX flow cytometer (Beckman Coulter, Indianapolis, IN, USA) in the red (585 nm) channel. Each sample recorded at least 10,000 events, and the obtained results were analyzed using CytExpert 2.4 software (Beckman Coulter, Indianapolis, IN, USA).
2.10. qRT-PCR
Quantitative real-time PCR (qPCR) was used to determine the mRNA expression level of
MDR1 (
ABCB1) [
35],
HIF-1α [
36],
MGMT [
37],
PARP1 and
PARP2 in U87-TxR and NCI-H460/R cells. The primer sets used to amplify
PARP1 and
PARP2 were generously provided by the Department of Molecular Biology at the Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade.
PARP1: forward 5′-GTGGATCCTGATTCTGGACTGG-3′ and reverse 5′-TCCTTGGACGGCATCTGTTC-3′ and
PARP2: forward 5′-CTACACCAGGATTCCGCATGA-3′ and reverse 5′-GTGTGGGAGCATGGGTAGATT-3′. The reactions were performed using a Maxima SYBR Green/ROX qPCR Master Mix (Thermo Scientific, Waltham, MA, USA) on an ABI PRISM 7000 Sequence Detection System (Applied Biosystems, Waltham, MA, USA) according to the manufacturer’s recommendations. Each sample was tested in triplicate, and relative gene expression levels were analyzed by the 2
−ΔΔCt method [
38]. The difference between the Ct values of specific genes and the endogenous control (
ACTB) [
39] represents the Δ
Ct value. All statistical analyses and data visualizations were performed using the R statistical computing environment (Version 4.2.2; R Core Team). Graphs and figures were generated with the ggplot2 package (Version 3.5.1), and statistical comparisons were conducted using the rstatix package (Version 0.7.2).
2.11. Rho123 and DOX Accumulation Assays
The accumulation of fluorescent Rho123 was analyzed by flow cytometry. Since Rho123 is a P-gp substrate, the intensity of fluorescence is proportional to Rho123 accumulation in the cell. NCI-H460/R and U87-TxR cells were counted, suspended in a growth medium and treated with increasing concentrations of LB-100 (2.5, 5, 10, 25 and 50 µM) and Rho123 (2 µM) simultaneously. The samples were then incubated at 37 °C in 5% CO2 for 30 min. At the end of the accumulation period, the samples were pelleted by centrifugation, washed with PBS and placed in 1 mL PBS. The green channel fluorescence (525 nm) of the samples was read on a CytoFLEX flow cytometer (Beckman Coulter, Indianapolis, IN, USA), and the results were analyzed by CytExpert software (Beckman Coulter, Indianapolis, IN, USA).
DOX fluorescence was utilized to examine the uptake of DOX in U87-TxR and NCI-H460/R cells after a 72 h treatment with 2.5 and 5.0 µM LB-100, using flow cytometry. DOX is a substrate for P-gp and emits fluorescence, with the intensity indicating the amount of DOX accumulated by the cells. For the analysis, both untreated and LB-100 treated cells were counted, and 100,000 cells per sample were placed in a growth medium containing 10 µM DOX after the 72 h treatment. The samples were incubated at 37 °C with 5% CO2 for 60 min. After the accumulation period, the samples that underwent centrifugation were washed twice with cold PBS and then resuspended in 1 mL of PBS for further analysis. The red channel fluorescence at 610 nm was measured using a CytoFLEX flow cytometer (Beckman Coulter, Indianapolis, IN, USA), and the results were analyzed with CytExpert software (Beckman Coulter, Indianapolis, IN, USA).
2.12. SwissADME Online Tool
The pharmacokinetic and drug-likeness properties of LB-100 were analyzed using the SwissADME website. Using this platform, values such as WLOGP (water partition coefficient, which indicates the lipophilicity of a molecule) and TPSA (topological polar surface area, which represents the polar surface area of a molecule) were calculated for LB-100 [
40]. The resulting “boiled egg” model indicates the position of the molecule on a WLOGP-versus-TPSA diagram and facilitates the assessment of passive gastrointestinal absorption and brain penetration of small molecules such as LB-100. The white region of the diagram corresponds to a high probability of passive absorption through the human intestine (HIA), while the yellow region, the yolk, indicates a high probability of blood–brain barrier (BBB) penetration. These regions are not mutually exclusive. Blue-colored dots indicate molecules that are expected to be actively secreted by P-gp (PGP+), whereas red-colored dots represent molecules that are not considered P-gp substrates (PGP−).
2.13. Cell Cycle Analysis
The MDR cancer cells were seeded in 6-well plates at a density of 200,000 cells per well for U87-TxR, and 100,000 cells per well for NCI-H460/R in 2 mL of the appropriate medium. The effects and the aforementioned subsequent treatment of 2.5 μM LB-100 and 250 nM DOX in U87-TxR cells, and 5 μM LB-100 and 500 nM DOX in NCI-H460/R cells on cell cycle distribution were studied after 72 h. Following treatment, cells were trypsinized, pelleted by centrifugation, rinsed with PBS and fixed in 70% ethanol for 24 h at −20 °C. After fixation, the cells were rinsed with PBS and pretreated with 50 μg/mL ribonuclease A, followed by a 15 min incubation at 37 °C. Next, PI was added to a final concentration of 2 μg/mL. Flow cytometry was performed using the CytoFLEX flow cytometer (Beckman Coulter, Indianapolis, IN, USA), collecting data from at least 10,000 events per sample. The cell cycle distribution was analyzed using CytExpert software (Beckman Coulter, Indianapolis, IN, USA).
2.14. Double Strand DNA Breaks Detection
To assess DNA damage in U87-TxR and NCI-H460/R cells, the fluorescence intensity of the anti-phospho-histone H2A.X antibody was employed. This antibody identifies endogenous phosphorylated H2A.X, which accumulates at the sites of double-strand breaks in DNA. The cells were initially plated at a concentration of 100,000 cells per well in 6-well tissue culture plates and allowed to grow overnight. The effects of the aforementioned subsequent treatment of 2.5 μM LB-100 and 250 nM DOX in U87-TxR cells, and 5 μM LB-100 and 500 nM DOX in NCI-H460/R cells were assessed. After treatment, cells were harvested, rinsed with PBS and fixed in 4% paraformaldehyde for 15 min at room temperature. Next, they were permeabilized with ice-cold 90% methanol for 90 min at −20 °C. After another PBS rinse, the cells were blocked with 0.5% BSA in PBS for 1 h and incubated overnight at 4 °C with an anti-phospho-histone H2A.X antibody (1:500 dilution in 0.5% BSA in PBS). After another PBS wash, the cells were incubated for 30 min at room temperature with the fluorescent Alexa Fluor 488 anti-rabbit IgG (H + L) secondary antibody (1:1000 dilution in 0.5% BSA in PBS). The cells were washed again and resuspended in 1 mL of PBS, and green fluorescence intensity was measured at 525 nm using a CytoFLEX flow cytometer (Beckman Coulter, Indianapolis, IN, USA). A minimum of 10,000 events were recorded for each sample, and the results were analyzed using CytExpert software (Beckman Coulter, Indianapolis, IN, USA).
2.15. Statistical Analysis
The IC50 values obtained from the immunofluorescence and MTT assays were calculated using nonlinear regression analysis in GraphPad Prism 8.0.2 for Windows (San Diego, CA, USA). The results regarding cytotoxicity and the percentage of P-gp positive cells in patient-derived samples, assessed by the immunofluorescence assay, as well as P-gp expression in MDR cancer cell lines, were analyzed using a two-way ANOVA followed by Dunnett’s multiple comparisons test, also using GraphPad Prism 8.0.2. A one-way ANOVA with Dunnett’s multiple comparisons test was applied for the DOX accumulation assay, while the qRT-PCR results were analyzed using a paired t-test. p values below 0.05 were considered significant (* p < 0.05; ** p < 0.01; *** p < 0.001; n.s. = non-significant). All the results represent the means ± SEM from at least three samples or experiments.
4. Discussion
LB-100, a PP2A inhibitor, can counteract cancer cell senescence, promote the differentiation of progenitor cells, enhance drug penetration and induce mitotic catastrophe and cell death [
12]. In this study, we explored the effects of LB-100 in combination with adavosertib and doxorubicin (DOX) in patient-derived cell cultures of glioblastoma and NSCLCs. Additionally, we conducted studies using MDR cellular models of glioblastoma and NSCLCs to investigate the potential mechanisms behind the effects of LB-100 in combination with other drugs, using DOX as a reference for this study.
Our results from multiple applications of LB-100 in combination with adavosertib and DOX in patient-derived glioblastoma and NSCLCs align with findings from previous research that demonstrated synergy between adavosertib and LB-100 in various models of colon cancer, pancreatic cancer and cholangiocarcinoma [
16]. However, single applications of this combined treatment, evaluated through an ex vivo immunofluorescence assay and the SynergyFinder+ online tool, indicated that LB-100 enhances the effects of adavosertib and DOX but without synergistic interaction.
The combination of LB-100 and adavosertib induces replication stress, which leads to premature mitosis and cell death. Prior studies have shown that LB-100 downregulates DNA repair signaling and deregulates several mitotic proteins, resulting in replication stress and mitotic catastrophe [
19,
41]. Additionally, adavosertib drives cells with unrepaired DNA damage into mitosis by inhibiting WEE1, leading to mitotic catastrophe [
42]. Thus, the synergistic effect of LB-100 and adavosertib stems from their complementary impacts on cell cycle regulation, DNA damage repair and apoptosis. However, our experimental setting indicates that the combination must be applied multiple times to achieve synergistic interaction.
DOX intercalates into DNA, disrupts DNA replication and transcription and inhibits topoisomerase II, inducing DNA double-strand breaks [
43]. Since LB-100 downregulates DNA repair signaling, it may inhibit the repair of DOX-induced DNA damage, forcing cells with damaged DNA into mitosis and ultimately causing cell death. Similar to the LB-100 and adavosertib combination, our results indicate that achieving synergistic interaction between LB-100 and DOX also requires multiple applications.
We analyzed the expression of the MDR marker P-gp in patient-derived cells of glioblastoma and NSCLCs after combined treatment with LB-100 and adavosertib/DOX, using a modified immunoassay developed in our laboratory [
33]. LB-100 alone tended to increase P-gp expression, particularly at higher concentrations. The PP2A modulates P-gp expression by influencing signaling pathways, transcription factors and posttranslational modifications associated with P-gp expression and activity [
44]. As a result, dysregulation of PP2A activity may lead to elevated P-gp expression through various mechanisms. When LB-100 was added to adavosertib, there was a further increase in P-gp expression compared to adavosertib alone in both primary cell cultures. However, the combined treatment of LB-100 and DOX presented different outcomes. In glioblastoma primary cells, LB-100 further increased P-gp expression at lower concentrations of DOX. In contrast, in NSCLC primary cells, LB-100 resulted in a significant decrease in P-gp expression compared to DOX alone.
The ability of LB-100 to modify P-gp levels was examined at the transcriptional and protein levels in glioblastoma and NSCLC MDR models, which exhibit overexpression of P-gp, compared to their sensitive counterpart cells. Treatment with LB-100 led to a time-dependent reduction in P-gp expression in both MDR models. Notably, LB-100 induced a cell type-dependent effect on
ABCB1 gene expression, which encodes P-gp. Specifically, LB-100 decreased
HIF1α mRNA expression in MDR glioblastoma U87-TxR cells. The inhibition of PP2A by LB-100 may increase the phosphorylation of proteins involved in regulating HIF-1α degradation [
45], leading to a reduction in HIF-1α levels, subsequently suppressing its transcriptional activation of target genes, including
ABCB1. The observed decrease in
ABCB1 mRNA expression after 24 h of treatment with LB-100 is followed by a decline in P-gp levels after 48 h and 72 h.
Our findings revealed increased
ABCB1 and
HIF1α mRNA expression following LB-100 treatment in MDR NSCLC NCI-H460/R cells. HIF-1α is a transcription factor tightly regulated at the transcriptional level by multiple signaling pathways. The PI3K-Akt-mTOR and ERK signaling pathways promote
HIF-1α mRNA expression [
46,
47]. PP2A dephosphorylates and inactivates Akt, mTOR and the ERK signaling pathway [
48]. Thus, the inhibition of PP2A hyperactivates mTOR and maintains ERK activation, leading to enhanced
HIF-1α mRNA expression. HIF-1α binds directly to hypoxia response elements (HREs) in the promoter of the
ABCB1 gene, driving its transcription [
49]. Despite the increase in
ABCB1 gene transcription after LB-100 treatment, P-gp expression shows a concentration-dependent decrease maintained over time, indicating that the post-translational regulation of P-gp is also affected. It is known that the activity and stability of P-gp are influenced by its phosphorylation status. The reduction in P-gp protein levels by LB-100 may result from altered protein stability, likely due to PP2A inhibition disrupting the phosphorylation states involved in the turnover or degradation of P-gp [
44,
50] suggesting that while
ABCB1 transcription is upregulated, along with its transcriptional regulator
HIF-1α, P-gp becomes less stable or is degraded more rapidly, possibly due to changes in cellular stress or protein quality control mechanisms activated by PP2A inhibition.
HIF-1α also plays a crucial role in regulating the transcription of the
MGMT gene, which encodes the DNA repair protein O6-methylguanine-DNA-methyl-transferase [
51]. Consequently, the LB-100-induced decrease in
HIF-1α mRNA expression was followed by reduced
MGMT gene transcription in MDR glioblastoma cells. This reduction in
MGMT transcription levels, prompted by LB-100, could be advantageous for glioblastoma patients, as the absence of
MGMT expression is regarded as a favorable prognostic indicator for temozolomide-treated glioblastoma patients [
52].
LB-100 can influence DNA repair processes and disrupt several proteins that are involved in cell-cycle checkpoints [
19,
41]. This effect may increase the dependence on DNA damage repair proteins such as poly (ADP-ribose) polymerases (PARPs), potentially altering their expression levels. Our results showed decreased gene expression of
PARP1 and
PARP2 in MDR glioblastoma cells, while MDR NSCLCs exhibited increased mRNA expression of these genes.
We also conducted a cell cycle analysis and assessed DNA damage by detecting double-strand breaks to explore whether changes in the cell cycle or the induction of DNA damage contribute to the increased efficacy of DOX when combined with LB-100. Our findings indicated that adding LB-100 does not change the cell cycle dynamics or induce DNA damage compared to DOX alone.
P-gp plays a critical role in eliminating various anticancer drugs, including DOX [
53]. Since LB-100 has shown potential in suppressing P-gp expression, we hypothesized that combining LB-100 with DOX could be beneficial in sensitizing MDR cancer cells. Therefore, we assessed the cytotoxicity of simultaneous and subsequent combined treatments. In U87-TxR cells, only the subsequent administration of LB-100 enhanced the cytotoxicity of DOX. In NCI-H460/R cells, LB-100 improved the efficacy of DOX with both treatment schedules, although the subsequent treatment demonstrated a more pronounced effect. Our findings highlight the importance of the sequence, in which LB-100 is administered alongside other drugs. Additionally, a DOX accumulation assay conducted after 72 h with LB-100 revealed a concentration-dependent increase in DOX accumulation in both MDR cancer cell models. This result aligns with the observed impact of LB-100 on DOX efficacy in simultaneous and subsequent treatments and could also be due to LB-100 ability to enhance drug penetration [
12]. The Rho123 accumulation assay demonstrated that LB-100 does not affect P-gp activity, while the SwissADME analysis indicated that LB-100 is not a P-gp substrate. Therefore, our findings suggest that LB-100 sensitizes MDR cancer cells by altering P-gp expression.
The impact of LB-100 on P-gp expression and activity has not been studied previously. According to the SwissADME tool, LB-100 cannot penetrate the BBB, which includes P-gp as a key component. While the SwissADME tool offers predictive insights, validating these findings through experimental studies is essential to determine whether LB-100 can indeed cross the BBB. Additionally, this tool suggests that LB-100 is not a substrate for P-gp. To confirm this, we used the Rho123 accumulation assay in MDR cancer cells expressing P-gp, which supported the prediction.
Mechanistically, LB-100 reduces P-gp expression in MDR cancer cells. However, the transcriptional pattern of the ABCB1 gene varies between MDR glioblastoma and NSCLCs, suggesting that regulation may occur at the post-transcriptional level. HIF-1α, a transcriptional regulator of ABCB1, also exhibited a different pattern in MDR glioblastoma and NSCLCs that mirrors ABCB1 expression.
Furthermore, SwissADME shows that LB-100 does not interact with enzymes involved in drug metabolism, making it a promising candidate for combination therapy. The impact of LB-100 on P-gp expression is most significant after 72 h, which correlates with increased DOX accumulation and enhanced sensitivity of MDR cancer cells to DOX treatment. Notably, LB-100 does not appear to affect DNA damage or disrupt the cell cycle, indicating that MDR cancer cells possess robust DNA repair mechanisms that may counteract the typical effects of LB-100 on DNA damage induction and alterations in cell cycle dynamics.