Open Access

Paclitaxel mediates the PI3K/AKT/mTOR pathway to reduce proliferation of FLT3‑ITD+ AML cells and promote apoptosis

  • Authors:
    • Yanyun Su
    • Meiqing Wu
    • Baowen Zhou
    • Ziwen Bai
    • Ruli Pang
    • Zhenfang Liu
    • Weihua Zhao
  • View Affiliations

  • Published online on: February 23, 2024     https://doi.org/10.3892/etm.2024.12449
  • Article Number: 161
  • Copyright: © Su et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute myeloid leukemia (AML) with internal tandem duplication (ITD) mutations in the FLT3 tyrosine kinase tend to have a poor prognosis. FLT3‑ITD can promote the progress of AML by activating the PI3K/AKT/mTOR pathway. Paclitaxel (PTX) is a natural anticancer drug that has been widely used in chemotherapy for multiple malignancies. The present study used the CCK‑8 assay, flow cytometry, PCR and western blotting to explore the anti‑leukemia effect and possible mechanisms of PTX on MV4‑11 cells with the FLT3‑ITD mutation and the underlying mechanism. As a result, it was found that PTX could inhibit proliferation of MV4‑11 cells and promoted apoptosis by inhibiting the PI3K/AKT/mTOR pathway.

Introduction

Acute myeloid leukemia (AML) is a common malignancy of the blood system and has characteristics of genetic diversity, aggression and high heterogeneity (1). Most AML patients have gene mutations associated with the occurrence, development and prognosis of leukemia (2-5), including the internal tandem duplication (ITD) mutations of the FLT3 tyrosine kinase (FLT3-ITD) gene. Of AML patients ~30% carry these mutations (6,7) with adverse effects on treatment outcomes (8,9). The FLT3-ITD mutation causes autophosphorylation of the FLT3 receptor in the absence of its ligand (10), resulting in activation of downstream PI3K/AKT, RAS/MAPK/ERK and STAT5 signaling pathways (11,12). Ultimately, proliferation and survival of leukemia cells are both enhanced (13-15). Therefore, the FLT3-ITD has been considered a therapeutic target (16) and several FLT3 inhibitors explored (4). However, inhibitory effects are limited (17,18) by acquired resistance caused by a secondary point mutation in the activation loop of the FLT3, producing FLT3-TKD, which often causes patients to relapse after remission (16,19,20). Thus, the search continues for new drugs and treatment strategies for AML patients.

The anti-tumor drug, paclitaxel (PTX) (21,22), promotes tubulin aggregation, stabilizes microtubule structure, inhibits cell mitosis and arrests cells in the G2/M phase of the cell cycle (23). The drug also regulates Bcl-2 and Caspase-3 to promote apoptosis of cancer cells (24,25). Increasing evidence suggests that PTX mediates the PI3K/AKT signaling pathway to inhibit proliferation of solid tumor cells and induce apoptosis. Such actions have been reported for nasopharyngeal carcinoma (26), cervical cancer (27) and lung cancer (28). Paclitaxel also enhances the sensitivity of cancer cells to other antitumor drugs through regulation of this pathway (28,29). Inhibitory effects have also been found for PTX with leukemia cell lines, such as HL-60(30) and K562 (31,32). Our previous study (33) showed that PTX combined with quizartinib synergistically inhibited proliferation and induced apoptosis of the FLT3-ITD+ AML cell-line, MV4-11, but how PTX acts on the MV4-11 cells remains to be elucidated.

The current study sought to clarify the effect of PTX on proliferation and apoptosis of MV4-11 cells and to investigate the underlying mechanism. Paclitaxel was found to have anti-proliferative and apoptosis-inducing effects on MV4-11 cells via an underlying mechanism connected with the PI3K/AKT/mTOR pathway.

Materials and methods

Cell culture

MV4-11 (FLT3-ITD+ AML cell-line) cells were obtained from Nanjing Kebai Biotechnology Co., Ltd. and THP-1 and K562 (FLT3-ITD- leukemia cell-lines) cells from Guangzhou Saiku Biotechnology Co., Ltd. and Procell Life Science & Technology Co., Ltd., respectively. MV4-11 cells were cultured in Iscove's Modified Dulbecco's Medium (Corning, Inc.) and THP-1 and K562 cells in RPMI-1640 medium (Corning, Inc.) both containing penicillin-streptomycin solution and 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) 37˚C, 5% CO2 in a humidified incubator.

Reagents

Paclitaxel and sorafenib were obtained from Shanghai Yuanye Biotechnology Co., Ltd. and PKI-587 from Shanghai Macklin Biochemical Co., Ltd. Cell Counting Kit-8 (CCK8) was from Dojindo Laboratories, Inc. and Annexin V-FLTC/propidium iodide (PI) Apoptosis Assay Kit (cat. no. 56584) from BD Biosciences. Reverse transcription and PCR kits were from Takara Biotechnology Co., Ltd. Anti-FLT3 antibody (1:1,000; cat. no. 3462), anti-AKT antibody (1:1,000; cat. no. 4691), anti-phosphorylated (p-)AKT antibody (1:2,000; cat. no. 4060), anti-S6K antibody (1:1,000; cat. no. 2708) and anti-p- S6K antibody (1:1,000; cat. no. 9234) were all purchased from Cell Signaling Technology, Inc. and anti-GAPDH antibody (1:5,000; cat. no. 10494-1-AP) was from Proteintech Group, Inc. HRP-conjugated secondary antibodies goat anti-rabbit (1:2,000; cat. no. M21002S) was purchased from Abmart Pharmaceutical Technology Co., Ltd.

CCK8 assay

MV4-11, THP-1 and K562 cells were seeded into 96-well plates and 10 µl per well CCK-8 added at time-points indicated for 1-4 h before the absorbance (OD) value was measured at 450 nm by microplate reader. Mean OD values at each concentration were used to calculate the proliferation inhibition rate=[1-(experimental group OD value-blank group OD value)/(negative control group OD value-blank group OD value)] x100%. Experiments were performed in triplicate. Half-maximal inhibitory concentration (IC50) values were calculated by GraphPad Prism version 5.0 software (Dotmatics).

Effects of Paclitaxel plus PKI-587 on proliferation of MV4-11 cells

The synergistic index, q, was calculated using the Kingsdale formula (34) to assess the combined effect of the two drugs. The formula was: q=E (D1+2)/(D1+ D2-D1xD2), where D1 and D2 are individual rates of inhibition by the two drugs and D1+2 is the combined rate of inhibition. Values of q>1.15, 0.85≤q≤1.15 and q<0.85 indicate synergistic, additive and antagonistic effects, respectively.

Flow cytometry assay

Aliquots of 1x106 cells/ml of MV4-11, THP-1 and K562 cells were treated for 48 h before harvesting and washing once with PBS precooled at 4˚C. Cells were resuspended with 1X Binding Buffer and 5 µl Annexin V added with incubation at room temperature for 15 min in the dark. 5 µl PI and 300 µl 1X Binding Buffer were added, at room temperature for 5 min. FACSCalibur (BD Biosciences) was used to perform flow cytometry within 5 min. FlowJo 10.0.7 software (FlowJo LLC) was used for analysis. Apoptotic rate=the percentage of early + late apoptotic cells.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted by TRIzol® reagent (Thermo Fisher Scientific, Inc.) from 1x106 cells/ml of MV4-11 cells which had been exposed to various concentrations of PTX for 48 h. Samples were dried and RNA dissolved in DEPC water. The concentration was adjusted to give an OD260/OD280 ratio between 1.8-2.0 and Prime Script RT Master Mix reverse transcription kit used to perform the reverse transcription into DNA at 37˚C for 15 min followed by inactivation at 85˚C for 5 min, according to the manufacturer's protocol. RT-qPCR was performed using TB Green Premix Ex Taq II kit with GAPDH as internal reference, according to the manufacturer's instructions. Fold change in expression levels was calculated using the 2-IICq method (35). The Fluorescent PCR instrument, Bio-Rad CFX Manager 2.1 was purchased from Bio-Rad Laboratories, Inc. All RT-qPCR primers were designed by Sangon Biotech Co., Ltd. and the primer sequences were as follows: FLT3, forward, 5'-GCAATCATAAGCACCAGCCAGGA-3' and reverse, 5'-TTCTGCGAGCACTTGAGGTTTCC-3'; PI3K, forward, 5'-CTTTGCGACAAGACTGCCGAGAG-3' and reverse, 5'-CGCCTGAAGCTGAGCAACATCC-3'; AKT, forward, 5'-ATGGAGTATGCCAACGGGGG-3' and reverse, 5'-TGTCGCGGTATACCACGTC-3'; mTOR, forward, 5'-CTTGCTGAACTGGAGGCTGATGG-3' and reverse, 5'-CCGTTTTCTTATGGGCTGGCTCT-3'; S6K, forward, 5'-TGCTGTGGATTGGTGGAGTTTGG-3' and reverse, 5'-TCTGGCTTCTTGTGTGAGGTAGGG-3'; GAPDH, forward, 5'-CTCTGCTCCTCCTGTTCGAC-3' and reverse, 5'-TAAAAAGCAGCCCTGGTGAC-3'. The thermocycling conditions were: Initial denaturation at 95˚C for 30 sec, denaturation at 95˚C for 5 sec, annealing and elongation at 60˚C for 30 sec for a total of 40 cycles.

Western blotting

Total protein was extracted from 1x106 cells/ml MV4-11 cells which had been exposed to various concentration of PTX for 48 h. In brief, cells were washed twice with pre-cooled PBS and radioimmunoprecipitation assay lysis buffer (RIPA; Beyotime Institute of Biotechnology) added before protein concentrations were determined by Enhanced Bicinchoninic acid Protein Assay Kit (BCA; Beyotime Institute of Biotechnology). Protein samples (10 µl per lane) were separated with 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) before transfer to PVDF membrane (Millipore, USA). Membranes were blocked for 1 h at room temperature with 5% BSA blocking buffer (Beijing Solarbio Science & Technology Co., Ltd.) and incubated with primary antibodies overnight at 4˚C. Membranes were incubated with secondary antibodies for 1 h at room temperature and ECL luminescence substrate kit (Biosharp Life Sciences) and ImageJ version 1.48 software (National Institutes of Health) were used to visualize and quantify protein bands.

Statistical analysis

SPSS17.0 software (SPSS, Inc.) was used to perform all statistical analyses. Data are presented as mean ± standard deviation. When groups=3, differences were analyzed by one-way ANOVA with the Least-Significant Difference (LSD) for post hoc multiple comparisons. When groups >3, differences were analyzed by one-way ANOVA with Tukey's for post hoc multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of PTX on proliferation of leukemia cells

To assess the effect of PTX on the FLT3-ITD+ AML cells, MV4-11, the present study first confirmed its effect on cell viability and compared it with that of FLT3 inhibitor sorafenib, which has been widely used in treatment of AML patients. It used various concentrations of PTX and sorafenib to treat MV4-11 cells for 24, 48 and 72 h. The results showed that PTX inhibited proliferation of MV4-11 cells in a dose-dependent manner and with increased potency with prolonged exposure (Fig. 1A). Sorafenib also had a significant inhibitory effect on cell proliferation (Fig. 1B). By comparing the IC50 values of both drugs for inhibition of MV4-11 cell proliferation, it was found that the inhibitory effect of PTX was stronger than that of sorafenib (Table I). To further test and compare the anti-proliferative effect of PTX on the FLT3-ITD- leukemia cells, THP-1 and K562, the present study used the same concentrations of PTX to treat THP-1 and K562 cells, respectively. It was observed that there was a more moderate inhibitory effect on proliferation (Fig. 1C and D) and PTX IC50 values for inhibition of THP-1 and K562 cell proliferation were larger than that for MV4-11 cells (Table I). These results indicated that PTX could clearly inhibit proliferation of MV4-11 cells.

Table I

IC50 values for growth rate inhibition by paclitaxel and sorafenib for leukemia cells.

Table I

IC50 values for growth rate inhibition by paclitaxel and sorafenib for leukemia cells.

 Paclitaxel (nM)Sorafenib (nM)
Drug action durationMV4-11THP-1K562MV4-11
24 h14.35±1.2235.84±8.77 99.29±2.59a16.84±1.41
48 h5.65±0.35 11.53±1b 33.13±3.85b 11.41±0.9c
72 h3.92±0.18 7.01±0.12a 17.65±0.95a 8.95±1.67c

[i] Various concentrations of paclitaxel were used to treat MV4-11, THP-1 or K562 cells and sorafenib was used to treat MV4-11 for 24, 48 and 72 h. Cell growth was measured by CCK8 assay and the IC50 values were calculated by GraphPad Prism Version 5.0 software. Data is presented as mean ± standard deviation (n=3). At the same time of action, vs. MV4-11 cells,

[ii] aP<0.01,

[iii] bP<0.001. At the same time of action, for MV4-11cells, vs. paclitaxel,

[iv] cP<0.05. IC50, half-maximal inhibitory concentration.

Effect of PTX on apoptosis in leukemia cells

MV4-11, THP-1 and K562 cells were treated with a range of PTX concentrations for 48 h before evaluation of apoptosis by flow cytometry. Rates of apoptosis elicited by PTX in the different cell-lines were: MV4-11, 10 nM PTX; 93.53±0.31% and 20 nM PTX: 95.37±0.67%; THP-1 cells, 10 nM PTX: 7.31±0.23% and 20 nM PTX: 53.23±0.9%; K562 cells, 10 nM PTX: 11.67±0.43% and 20n M PTX: 12.68±0.58%. PTX induced apoptosis in leukemia cells at higher rates for MV4-11 cells compared with THP-1 and K562 cells (Fig. 2). Then, in order to compare the pro-apoptotic effects of PTX and sorafenib on MV4-11 cells, the same concentration of PTX and sorafenib was used to treat MV4-11 cells respectively for 48 h. It was found that the apoptosis rate of sorafenib at 4 nM was slightly higher than that of PTX, but at 8 nM, the apoptosis rate of PTX, 91.2±0.82%, was significantly higher compared with that of sorafenib, 48.03±0.15% (Fig. 3). Therefore, it was hypothesized that the pro-apoptotic effect of PTX on MV4-11 cells was stronger than sorafenib.

Effect of PTX on expression of FLT3 mRNA and protein in MV4-11 cells

Given that PTX reduced proliferation and promoted apoptosis in MV4-11 cells, the underlying mechanism was investigated by treating MV4-11 cells with 10 or 20 nM PTX for 48 h, followed by PCR and western blotting. FLT3 mRNA (Fig. 4A) and protein (Fig. 4B and C) expression were significantly downregulated compared with control cells and with the increase of PTX concentration, FLT3 gene expression decreased significantly. Thus, downregulation of the FLT3 gene may be linked to decreased growth of MV4-11 cells.

Effect of PTX on PI3K/AKT/mTOR pathway in MV4-11 cells

The FLT3 receptor activates the downstream PI3K/AKT/mTOR signaling pathway which is known to be involved in growth of leukemia cells (36). According to the results of the previous experiment, PTX can inhibit the expression of FLT3 gene so, to test whether PTX have any effect on the PI3K/AKT/mTOR pathway, MV4-11 cells were treated with 0, 10 or 20 nM PTX for 48 h and expression of genes dependent on PI3K/AKT/mTOR signaling measured by PCR and western blotting. Production of PI3K, AKT, mTOR and S6K mRNA were all downregulated compared with the control cells (Fig. 5). Compared with the control group, the expression of p-AKT (Fig. 6A) and p-S6K (Fig. 6B) protein were also decreased significantly, but the changes of AKT, S6K protein were not evident (Fig. 6B). Based on this experiment, it was hypothesized that PTX might mainly inhibit the phosphorylation of AKT and S6K. Thus, the inhibitory effect of PTX on MV4-11 cell growth may be linked to inhibition of the PI3K/AKT/mTOR pathway.

Effect of the PI3K/AKT/mTOR pathway inhibitor, PKI-587, in inhibiting growth of MV4-11 cells

The above experiments showed that PTX could not only have anti-proliferative and pro-apoptotic effect on MV4-11 cells, but also inhibited PI3K/AKT/mTOR pathway in the cells. To test whether the effect of PTX on MV4-11 cells depend on this signaling pathway, cells were treated with PI3K/AKT/mTOR pathway inhibitor PKI-587, which also known as gedatolisib or PF 05212384. PKI-587 could inhibit cell viability (Fig. 7). Taken together, it was hypothesized that the inhibitory effect of PTX on MV4-11 cells proliferation was related to the PI3K/AKT/mTOR pathway.

Effect of PTX combined with PKI-587 on cell proliferation and apoptosis of MV4-11 cells

Abnormal activation of PI3K/AKT/mTOR pathway is not only related to the occurrence and development of AML, but also drug resistance (37). As a dual PI3K/AKT/mTOR inhibitor, PKI-587 has been shown to efficiently inhibit cell proliferation, block colony formation and induce apoptosis of sorafenib-resistant AML cells (38). The results of the present study confirmed that PKI-587 also had clear anti-proliferative on MV4-11 cells. As it was observed that PTX could inhibit the PI3K/AKT/mTOR pathway, it was hypothesized that PKI-587 might enhance the effect of PTX in MV4-11 cells, or the two drugs might have synergistic effect. To verify this hypothesis, first, 2 nM PTX combined with 10, 25 or 50 nM PKI-587 was used to treated MV4-11 cells, followed by CCK-8 assay to detect cell viability. As shown in Table II, comparing with PTX or PKI-587 monotherapy, the cell proliferation inhibition rate of the combination group was significantly increased. The synergistic index q value calculated by Kingsdale formula suggested that the combination of PTX and PKI-587 had synergistic or additive effect.

Table II

Effect of the combination of paclitaxel and PKI-587 on MV4-11 cell proliferation.

Table II

Effect of the combination of paclitaxel and PKI-587 on MV4-11 cell proliferation.

GroupInhibition rate (%)q-valueInteraction
Control3.11±0.63  
2 nM PTX10.5±3.3  
10 nM PKI-58714.63±5.93  
25 nM PKI-58766.68±3.44  
50 nM PKI-58769.65±5.63  
2 nM PTX + 10 nM PKI-587 35.41±2.63a1.50Synergy
2 nM PTX + 25 nM PKI-587 83.09±0.56a1.18Synergy
2 nM PTX + 50 nM PKI-587 82.07±5.87a1.13Additive

[i] MV4-11 cells were treated with 2 nM paclitaxel alone or in combination with 10, 25 or 50 nM PKI-587 for 48 h followed by CCK8 assay. The Kingsdale formula was used to determine the existence of synergism. Values of q>1.15, 0.85≤ q ≤1.15 and q<0.85 are considered to indicate synergistic, additive and antagonistic effects, respectively. Data is presented as mean ± standard deviation (n=3).

[ii] aP<0.05 vs. paclitaxel monotherapy group.

Second, its effect on cells apoptosis was confirmed. From Table II, it can be seen that the synergistic index q value was the largest at 2 nM PTX combined with 10 nM. Therefore, 2 nM PTX combined with 10 nM PKI-587 was selected to treat cells. The results showed that the apoptosis rate of PTX or PKI-587 alone was 27.69±7.45% and 26.42±5.24%, respectively, but at the combination group, the apoptosis rate was up to 76.23±2.55% (Fig. 8). Taken together, it was concluded that PKI-587 might enhance effect of PTX on MV4-11 cells, the two drugs having a synergistic or additive effect. It was also further proved that the proliferation inhibition and pro-apoptosis effect of PTX on MV4-11 cells were related to the PI3K/AKT/mTOR signaling pathway.

Discussion

The prognosis of AML patients carrying the FLT3-ITD mutation remains poor and its 5-year survival rate is only ~20%, despite advances in treatments (39). Thus, there is a pressing need to identify new and effective treatment strategies for these patients (39). PTX is considered an effective antitumor treatment for a variety of malignancies. The present study found that PTX had a time- and dose-dependent anti-proliferative and apoptosis-inducing effect on MV4-11 cells carrying the FLT3-ITD mutation. PTX also inhibited expression of FLT3 mRNA and protein and exerted a regulatory effect on the PI3K/AKT/mTOR pathway.

Leukemia is a common malignant tumor derived from the hematopoietic system during which bone marrow, peripheral blood and other organs are filled with uncontrollably proliferating primitive white blood cells (40,41). Previous studies have demonstrated anti-proliferative effects of PTX on the leukemia cells, HL-60(30) and K562 (31,32) and a similar inhibitory effect on MV4-11 cells was predicted. The results showed that PTX had a significant anti-proliferative and apoptosis-inducing effect on MV4-11 cells. To observe and compare its effect on the FLT3-ITD negative leukemia cell lines, THP-1 and K562, both cell lines were treated under the same conditions. PTX could also inhibit cell proliferation and induce apoptosis of THP-1 or K562 cells; however, compared with MV4-11 cells, its effect was weaker. Therefore, it was hypothesized that the anti-proliferative and pro-apoptosis effect of PTX might depend on the leukemia cell type and that it had a more potent effect on FLT3-ITD+ AML cells.

As the FLT3-ITD mutation often indicates high risk in AML patients, several FLT3 inhibitors have been developed and used in clinics for AML patients, such as sorafenib, midostaurin and gilteritinib (42,43). The most widely used is sorafenib, which has been used to treat relapsed or refractory AML patients, or as an adjunct to transplantation, although it is merely a multi-kinase inhibitor (44). To further identify the effect of PTX on MV4-11 cells, its inhibitory and pro-apoptotic effect were compared with sorafenib. The results showed that the IC50 values of sorafenib were greater than PTX, indicating the anti-proliferative effect of PTX was stronger than sorafenib. Moreover, the apoptosis rate of sorafenib was lower compared with PTX. However, sorafenib is a multi-kinase inhibitor; in order to improve our understanding of the anti-leukemia effect of PTX, it is necessary to further compare the efficacy of PTX with gilteritinib, a new FLT3/AXL dual inhibitor which has a relatively definite efficacy in relapsed or refractory AML patients (45). Taken together, it was suggested that PTX had distinct effect on MV4-11 cells.

FLT3 is often overexpressed in hematopoietic tumors (6), affecting the development, growth and differentiation of blood cells and promoting leukemia progression. The FLT3-ITD mutation can decrease treatment efficacy and result in adverse outcomes (46) by activating downstream signaling pathways (11,12), such as the PI3K/AKT signaling pathway, which has a great influence on survival, proliferation and differentiation of hematopoietic cells (36). Abnormal activation of the PI3K/AKT signaling pathway associated with reduced overall survival, has been identified in ~60% of AML patients (47,48). A number of studies have shown that mutations or activation of receptor tyrosine kinases, for example, the FLT3-ITD mutation, can stimulate the PI3K/AKT signaling pathway in most human malignancies, including AML (13,49,50). PTX is a potent anticancer drug and several studies have shown that it can inhibit cell proliferation and induce apoptosis by regulating the PI3K/AKT pathway in various solid tumors, such as nasopharyngeal carcinoma (26,51), cervical cancer (27) and lung cancer. Furthermore, the pathway inhibitors or other drugs inactivating the PI3K/AKT pathway can enhance the chemosensitivity of PTX in various cancer cells (52-54). The present study observed that PTX could downregulate the expression of FLT3 mRNA and protein in MV4-11 cells. In addition, upon PTX treatment, the mRNA expression level of PI3K, AKT, mTOR and S6K and the proteins of p-AKT, p-S6K all declined distinctly. However, the changes of AKT, S6K protein were not clear and it was hypothesized that PTX might mainly influence the phosphorylation of AKT and S6K, perhaps through inactivating the PI3K/AKT/mTOR pathway, to affect cell proliferation and apoptosis. It has been reported that PI3K/mTOR pathway inhibitors can inhibit cell proliferation and induce apoptosis of MV4-11 cells and another FLT3-ITD+ AML cell line, MOLM-13 cells. The present study also demonstrated that the resistance of AML cells to sorafenib might have resulted from overexpression of p-AKT and p-S6K proteins of the PI3K/mTOR pathway. Inhibition of this pathway enhanced the anti-leukemia effect of sorafenib, lengthening the life of mice with transplanted tumors (38). The present study found that PKI-587, a dual PI3K/mTOR inhibitor did have cytotoxic effect on MV4-11 cells, which was consistent with a previous study (38). It was also combined with PTX to treat the cells and it was found that this combination showed a significant synergistic or additive effect in inhibiting cell proliferation and inducing apoptosis. These findings further demonstrated that the effect of PTX on MV4-11 cells might be associated with the PI3K/AKT/mTOR pathway. Future studies will further detect whether the effect of PTX increases when the expression of p-AKT and p-S6K are downregulated. However, there is an important question; how PTX regulates the FLT3 is unclear. On the basis of previous studies, it is hypothesized that PTX might inhibit heat shock protein 90 (Hsp90) to downregulate the expression of FLT3.

FLT3 as an important therapeutic target and its expression can be inhibited by a variety of small molecule inhibitors, such as Hsp90, proteasome, RET and other inhibitors (55). Hsp90 is a molecular chaperone that is overexpressed in leukemia cells and its overexpression is associated with indefinite cells proliferation and drug resistance, mainly through forming multiprotein complexes to stabilize its client proteins (55,56). It was hypothesized that PTX might, through inhibition of Hsp90, downregulate the expression of FLT3 for the following reasons. First, several studies have confirmed that FLT3 is one of the Hsp90 client proteins; Hsp90 inhibitors, such as 17-AAG, can reduce its expression and inhibit proliferation of leukemia cells and induce apoptosis (57-59). Second, AKT is also an Hsp90 client protein; Hsp90 inhibitors can inactivate AKT and downregulate the expression of p-AKT. The present study verified that PTX could reduce the expression of both FLT3 and p-AKT. Third, triptolide and tripterine, isolated from Tripeterygium wilfordii Hook f, both are terpenoid compounds. Previous studies have demonstrated that tripterine demonstrates anti-proliferation and pro-apoptosis effect on various leukemia cells by inhibiting the expression of Hsp90 (60-62). It has been reported that triptolide can also inhibit the expression of Hsp90(63). Considering that PTX is also a terpenoid compound, such as triptolide and tripterine, it was hypothesized that it might also inhibit the expression of Hsp90. For these reasons, it was further hypothesized that PTX might inhibit the expression of FLT3 and p-AKT by reducing the expression of Hsp90. This remains a hypothesis and further work will be conducted to identify its veracity.

In conclusion, the present study revealed the anti-proliferative and apoptosis-inducing effect of PTX on MV4-11 cells and its molecular mechanism, which was connected to the inhibition of PI3K/AKT/mTOR pathway. The aim of the present study was to establish the potential utility of PTX as a therapy for FLT3-ITD+ AML patients, supported by a theoretical basis. However, the present study was limited by its in vitro nature and did not establish the efficacy of PTX in patients. In the future, we will further explore the effects of PTX on primary cells and further animal experiments will be planned which may pave the way for clinical trials.

Acknowledgements

The authors thank Dr Luo Bin from Guangxi Medical University (Guangxi Zhuang Autonomous Region, China) for technical assistances with the CCK-8 and PCR.

Funding

Funding: The present study was funded by National Natural Science Foundation of China (grant no. 81700172).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YS and WZ conceived and designed the study. YS, BZ and ZB performed the experiment. RP was responsible for cultivating cells. MW and ZL provided technical guidance. YS wrote the manuscript. WZ revised and finalized the manuscript. YS and WZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Estey E and Döhner H: Acute myeloid leukaemia. Lancet. 368:1894–1907. 2006.PubMed/NCBI View Article : Google Scholar

2 

Yang F, Anekpuritanang T and Press RD: Clinical utility of next-generation sequencing in acute myeloid leukemia. Mol Diagn Ther. 24:1–13. 2020.PubMed/NCBI View Article : Google Scholar

3 

Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson A, Hoadley K, Triche TJ Jr, Laird PW, Baty JD, et al: Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 368:2059–2074. 2013.PubMed/NCBI View Article : Google Scholar

4 

Daver N, Schlenk RF, Russell NH and Levis MJ: Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia. 33:299–312. 2019.PubMed/NCBI View Article : Google Scholar

5 

Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, et al: Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 129:424–447. 2017.PubMed/NCBI View Article : Google Scholar

6 

Sun YM, Wang WT, Zeng ZC, Chen TQ, Han C, Pan Q, Huang W, Fang K, Sun LY, Zhou YF, et al: CircMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood. 134:1533–1546. 2019.PubMed/NCBI View Article : Google Scholar

7 

De Kouchkovsky I and Abdul-Hay M: ‘Acute myeloid leukemia: A comprehensive review and 2016 update’. Blood Cancer J. 6(e441)2016.PubMed/NCBI View Article : Google Scholar

8 

Whitman SP, Ruppert AS, Radmacher MD, Mrózek K, Paschka P, Langer C, Baldus CD, Wen J, Racke F, Powell BL, et al: FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood. 111:1552–1559. 2008.PubMed/NCBI View Article : Google Scholar

9 

Whitman SP, Archer KJ, Feng L, Baldus C, Becknell B, Carlson BD, Carroll AJ, Mrózek K, Vardiman JW, George SL, et al: Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: A cancer and leukemia group b study. Cancer Res. 61:7233–7239. 2001.PubMed/NCBI

10 

Almatani MF, Ali A, Onyemaechi S, Zhao Y, Gutierrez L, Vaikari VP and Alachkar H: Strategies targeting FLT3 beyond the kinase inhibitors. Pharmacol Ther. 225(107844)2021.PubMed/NCBI View Article : Google Scholar

11 

Levis M and Small D: FLT3: It does matter in leukemia. Leukemia. 17:1738–1752. 2003.PubMed/NCBI View Article : Google Scholar

12 

Takahashi S: Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: Biology and therapeutic implications. J Hematol Oncol. 4(13)2011.PubMed/NCBI View Article : Google Scholar

13 

Brandts CH, Sargin B, Rode M, Biermann C, Lindtner B, Schwäble J, Buerger H, Müller-Tidow C, Choudhary C, McMahon M, et al: Constitutive activation of Akt by FLT3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res. 65:9643–9650. 2005.PubMed/NCBI View Article : Google Scholar

14 

Rocnik JL, Okabe R, Yu JC, Lee BH, Giese N, Schenkein DP and Gilliland DG: Roles of tyrosine 589 and 591 in STAT5 activation and transformation mediated by FLT3-ITD. Blood. 108:1339–1345. 2006.PubMed/NCBI View Article : Google Scholar

15 

Döhner H, Weisdorf DJ and Bloomfield CD: Acute myeloid leukemia. N Engl J Med. 373:1136–1152. 2015.PubMed/NCBI View Article : Google Scholar

16 

Smith CC, Wang Q, Chin CS, Salerno S, Damon LE, Levis MJ, Perl AE, Travers KJ, Wang S, Hunt JP, et al: Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 485:260–263. 2012.PubMed/NCBI View Article : Google Scholar

17 

Piloto O, Levis M, Huso D, Li Y, Li H, Wang MN, Bassi R, Balderes P, Ludwig DL, Witte L, et al: Inhibitory anti-FLT3 antibodies are capable of mediating antibody-dependent cell-mediated cytotoxicity and reducing engraftment of acute myelogenous leukemia blasts in nonobese diabetic/severe combined immunodeficient mice. Cancer Res. 65:1514–1522. 2005.PubMed/NCBI View Article : Google Scholar

18 

Alvarado Y, Kantarjian HM, Luthra R, Ravandi F, Borthakur G, Garcia-Manero G, Konopleva M, Estrov Z, Andreeff M and Cortes JE: Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer. 120:2142–2149. 2014.PubMed/NCBI View Article : Google Scholar

19 

Weisberg E, Barrett R, Liu Q, Stone R, Gray N and Griffin JD: FLT3 inhibition and mechanisms of drug resistance in mutant FLT3-positive AML. Drug Resist Updat. 12:81–89. 2009.PubMed/NCBI View Article : Google Scholar

20 

Alotaibi AS, Yilmaz M, Kanagal-Shamanna R, Loghavi S, Kadia TM, DiNardo CD, Borthakur G, Konopleva M, Pierce SA, Wang SA, et al: Patterns of resistance differ in patients with acute myeloid leukemia treated with type I versus type II FLT3 inhibitors. Blood Cancer Discov. 2:125–134. 2021.PubMed/NCBI View Article : Google Scholar

21 

Zhu L and Chen L: Progress in research on paclitaxel and tumor immunotherapy. Cell Mol Biol Lett. 24(40)2019.PubMed/NCBI View Article : Google Scholar

22 

Tamburin S, Park SB, Alberti P, Demichelis C, Schenone A and Argyriou AA: Taxane and epothilone-induced peripheral neurotoxicity: From pathogenesis to treatment. J Peripher Nerv Syst. 24 (Suppl 2):S40–S51. 2019.PubMed/NCBI View Article : Google Scholar

23 

Leung JC and Cassimeris L: Reorganization of paclitaxel-stabilized microtubule arrays at mitotic entry: Roles of depolymerizing kinesins and severing proteins. Cancer Biol Ther. 20:1337–1347. 2019.PubMed/NCBI View Article : Google Scholar

24 

Vassileva V, Allen CJ and Piquette-Miller M: Effects of sustained and intermittent paclitaxel therapy on tumor repopulation in ovarian cancer. Mol Cancer Ther. 7:630–637. 2008.PubMed/NCBI View Article : Google Scholar

25 

Al-Mahayri ZN, AlAhmad MM and Ali BR: Current opinion on the pharmacogenomics of paclitaxel-induced toxicity. Expert Opin Drug Metab Toxicol. 17:785–801. 2021.PubMed/NCBI View Article : Google Scholar

26 

Li T: Pacilitaxel induces human nasopharyngeal carcinoma cell line CNE2 apoptosis and growth inhibition by suppressing PI3K/AKT/P53 signaling pathway. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 29:2147–2150. 2015.PubMed/NCBI(In Chinese).

27 

Guertin DA and Sabatini DM: Defining the role of mTOR in cancer. Cancer Cell. 12:9–22. 2007.PubMed/NCBI View Article : Google Scholar

28 

Ma D, Li S, Cui Y, Li L, Liu H, Chen Y and Zhou X: Paclitaxel increases the sensitivity of lung cancer cells to lobaplatin via PI3K/AKT pathway. Oncol Lett. 15:6211–6216. 2018.PubMed/NCBI View Article : Google Scholar

29 

Moschetta M, Pretto F, Berndt A, Galler K, Richter P, Bassi A, Oliva P, Micotti E, Valbusa G, Schwager K, et al: Paclitaxel enhances therapeutic efficacy of the F8-IL2 immunocytokine to EDA-fibronectin-positive metastatic human melanoma xenografts. Cancer Res. 72:1814–1824. 2012.PubMed/NCBI View Article : Google Scholar

30 

Ying J, Yang W, Xie CY, Ni QC, Pan XD, Dong JH, Liu ZM and Wang XS: Induction of caspase-3-dependent apoptosis in human leukemia HL-60 cells by δ-elemene. Yakugaku Zasshi. 131:1383–1394. 2011.PubMed/NCBI View Article : Google Scholar

31 

Xia RL, Lu Y, Zhu LN, Zhang SF, Zhao FK and Fu CY: Different regulatory pathways are involved in the proliferative inhibition of two types of leukemia cell lines induced by paclitaxel. Oncol Rep. 30:1853–1859. 2013.PubMed/NCBI View Article : Google Scholar

32 

Meshkini A and Yazdanparast R: Involvement of oxidative stress in taxol-induced apoptosis in chronic myelogenous leukemia K562 cells. Exp Toxicol Pathol. 64:357–365. 2012.PubMed/NCBI View Article : Google Scholar

33 

Bai ZW, Wu MQ, Zhou BW, Shi ZY, Yao YB, Liu ZF, Pang RL and Zhao WH: Effects of paclitaxel and quizartinib alone and in combination on aml cell line MV4-11 and Its STAT5 signal pathway. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 30:671–676. 2022.PubMed/NCBI View Article : Google Scholar : (In Chinese).

34 

Jin ZJ and Zhang XW: Equal probability and curve with ‘q50’-A new method to estimate the effect of drug combination. Journal of Shanghai Second Medical College. (01)(15-18+86)1981.(In Chinese).

35 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

36 

Nepstad I, Hatfield KJ, Grønningsæter IS and Reikvam H: The PI3K-AKT-mTOR signaling pathway in human acute myeloid leukemia (AML) cells. Int J Mol Sci. 21(2907)2020.PubMed/NCBI View Article : Google Scholar

37 

Martelli AM, Evangelisti C, Chappell W, Abrams SL, Bäsecke J, Stivala F, Donia M, Fagone P, Nicoletti F, Libra M, et al: Targeting the translational apparatus to improve leukemia therapy: Roles of the PI3K/PTEN/Akt/mTOR pathway. Leukemia. 25:1064–1079. 2011.PubMed/NCBI View Article : Google Scholar

38 

Lindblad O, Cordero E, Puissant A, Macaulay L, Ramos A, Kabir NN, Sun J, Vallon-Christersson J, Haraldsson K, Hemann MT, et al: Aberrant activation of the PI3K/mTOR pathway promotes resistance to sorafenib in AML. Oncogene. 35:5119–5131. 2016.PubMed/NCBI View Article : Google Scholar

39 

Port M, Böttcher M, Thol F, Ganser A, Schlenk R, Wasem J, Neumann A and Pouryamout L: Prognostic significance of FLT3 internal tandem duplication, nucleophosmin 1, and cebpa gene mutations for acute myeloid leukemia patients with normal karyotype and younger than 60 years: A systematic review and meta-analysis. Ann Hematol. 93:1279–1286. 2014.PubMed/NCBI View Article : Google Scholar

40 

Almond LM, Charalampakis M, Ford SJ, Gourevitch D and Desai A: Myeloid sarcoma: Presentation, diagnosis, and treatment. Clin Lymphoma Myeloma Leuk. 17:263–267. 2017.PubMed/NCBI View Article : Google Scholar

41 

Ganzel C and Douer D: Extramedullary disease in APL: A real phenomenon to contend with or not? Best Pract Res Clin Haematol. 27:63–68. 2014.PubMed/NCBI View Article : Google Scholar

42 

Levis M: Midostaurin approved for FLT3-mutated AML. Blood. 129:3403–3406. 2017.PubMed/NCBI View Article : Google Scholar

43 

Pulte ED, Norsworthy KJ, Wang Y, Xu Q, Qosa H, Gudi R, Przepiorka D, Fu W, Okusanya OO, Goldberg KB, et al: FDA approval summary: Gilteritinib for relapsed or refractory acute myeloid leukemia with a FLT3 mutation. Clin Cancer Res. 27:3515–3521. 2021.PubMed/NCBI View Article : Google Scholar

44 

Antar A, Otrock ZK, El-Cheikh J, Kharfan-Dabaja MA, Battipaglia G, Mahfouz R, Mohty M and Bazarbachi A: Inhibition of FLT3 in AML: A focus on sorafenib. Bone Marrow Transplant. 52:344–351. 2017.PubMed/NCBI View Article : Google Scholar

45 

Cucchi DGJ, Denys B, Kaspers GJL, Janssen J, Ossenkoppele GJ, de Haas V, Zwaan CM, van den Heuvel-Eibrink MM, Philippé J, Csikós T, et al: RNA-based FLT3-ITD allelic ratio is associated with outcome and ex vivo response to FLT3 inhibitors in pediatric AML. Blood. 131:2485–2489. 2018.PubMed/NCBI View Article : Google Scholar

46 

Mrózek K, Marcucci G, Paschka P, Whitman SP and Bloomfield CD: Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: Are we ready for a prognostically prioritized molecular classification? Blood. 109:431–448. 2007.PubMed/NCBI View Article : Google Scholar

47 

Min YH, Eom JI, Cheong JW, Maeng HO, Kim JY, Jeung HK, Lee ST, Lee MH, Hahn JS and Ko YW: Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia: Its significance as a prognostic variable. Leukemia. 17:995–997. 2003.PubMed/NCBI View Article : Google Scholar

48 

Chen W, Drakos E, Grammatikakis I, Schlette EJ, Li J, Leventaki V, Staikou-Drakopoulou E, Patsouris E, Panayiotidis P, Medeiros LJ and Rassidakis HZ: Mtor signaling is activated by FLT3 kinase and promotes survival of FLT3-mutated acute myeloid leukemia cells. Mol Cancer. 9(292)2010.PubMed/NCBI View Article : Google Scholar

49 

Nepstad I, Hatfield KJ, Grønningsæter IS, Aasebø E, Hernandez-Valladares M, Hagen KM, Rye KP, Berven FS, Selheim F, Reikvam H and Bruserud Ø: Effects of insulin and pathway inhibitors on the PI3K-Akt-mTOR phosphorylation profile in acute myeloid leukemia cells. Signal Transduct Target Ther. 4(20)2019.PubMed/NCBI View Article : Google Scholar

50 

Watanabe D, Nogami A, Okada K, Akiyama H, Umezawa Y and Miura O: FLT3-ITD activates RSK1 to enhance proliferation and survival of AML cells by activating mTORC1 and eIF4B cooperatively with PIM or PI3K and by inhibiting BAD and BIM. Cancers (Basel). 11(1827)2019.PubMed/NCBI View Article : Google Scholar

51 

Dong P, Hao F, Dai S and Tian L: Combination therapy eve and pac to induce apoptosis in cervical cancer cells by targeting PI3K/AKT/mTOR pathways. J Recept Signal Transduct Res. 38:83–88. 2018.PubMed/NCBI View Article : Google Scholar

52 

Ding Z, Xu F, Li G, Tang J, Tang Z, Jiang P and Wu H: Knockdown of Akt2 expression by shRNA inhibits proliferation, enhances apoptosis, and increases chemosensitivity to paclitaxel in human colorectal cancer cells. Cell Biochem Biophys. 71:383–388. 2015.PubMed/NCBI View Article : Google Scholar

53 

Lin YH, Chen BY, Lai WT, Wu SF, Guh JH, Cheng AL and Hsu LC: The Akt inhibitor MK-2206 enhances the cytotoxicity of paclitaxel (Taxol) and cisplatin in ovarian cancer cells. Naunyn Schmiedebergs Arch Pharmacol. 388:19–31. 2015.PubMed/NCBI View Article : Google Scholar

54 

Liu X, Xie C, Li A, Zhang Y, Liu X, Zhou S, Shen J, Huo Z, Cao W, Ma Y, et al: BEZ235 enhances chemosensitivity of paclitaxel in hepatocellular carcinoma through inhibiting the PI3K/Akt/mTOR pathway. Am J Transl Res. 11:7255–7271. 2019.PubMed/NCBI

55 

Hacıhanefioglu A, Gonullu E, Mehtap O, Keski H, Yavuz M and Ercin C: Effect of heat shock protein-90 (HSP90) and vascular endothelial growth factor (VEGF) on survival in acute lymphoblastic leukemia: An immunohistochemical study. Med Oncol. 28:846–851. 2011.PubMed/NCBI View Article : Google Scholar

56 

Han SY: Small molecule induced FLT3 degradation. Pharmaceuticals (Basel). 15(320)2022.PubMed/NCBI View Article : Google Scholar

57 

Yao Q, Nishiuchi R, Li Q, Kumar AR, Hudson WA and Kersey JH: FLT3 expressing leukemias are selectively sensitive to inhibitors of the molecular chaperone heat shock protein 90 through destabilization of signal transduction-associated kinases. Clin Cancer Res. 9:4483–4493. 2003.PubMed/NCBI

58 

Ly BT, Chi HT, Yamagishi M, Kano Y, Hara Y, Nakano K, Sato Y and Watanabe T: Inhibition of FLT3 expression by green tea catechins in FLT3 mutated-AML cells. PLoS One. 8(e66378)2013.PubMed/NCBI View Article : Google Scholar

59 

Al Shaer L, Walsby E, Gilkes A, Tonks A, Walsh V, Mills K, Burnett A and Rowntree C: Heat shock protein 90 inhibition is cytotoxic to primary AML cells expressing mutant FLT3 and results in altered downstream signalling. Br J Haematol. 141:483–493. 2008.PubMed/NCBI View Article : Google Scholar

60 

Hieronymus H, Lamb J, Ross KN, Peng XP, Clement C, Rodina A, Nieto M, Du J, Stegmaier K, Raj SM, et al: Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell. 10:321–330. 2006.PubMed/NCBI View Article : Google Scholar

61 

Peng B, Xu L, Cao F, Wei T, Yang C, Uzan G and Zhang D: HSP90 inhibitor, celastrol, arrests human monocytic leukemia cell U937 at G0/G1 in thiol-containing agents reversible way. Mol Cancer. 9(79)2010.PubMed/NCBI View Article : Google Scholar

62 

Lu Z, Jin Y, Qiu L, Lai Y and Pan J: Celastrol, a novel HSP90 inhibitor, depletes Bcr-Abl and induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation. Cancer Lett. 290:182–191. 2010.PubMed/NCBI View Article : Google Scholar

63 

Zhang FZ, Ho DH and Wong RH: Triptolide, a HSP90 middle domain inhibitor, induces apoptosis in triple manner. Oncotarget. 9:22301–22315. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Su Y, Wu M, Zhou B, Bai Z, Pang R, Liu Z and Zhao W: Paclitaxel mediates the PI3K/AKT/mTOR pathway to reduce proliferation of FLT3‑ITD<sup>+</sup> AML cells and promote apoptosis. Exp Ther Med 27: 161, 2024
APA
Su, Y., Wu, M., Zhou, B., Bai, Z., Pang, R., Liu, Z., & Zhao, W. (2024). Paclitaxel mediates the PI3K/AKT/mTOR pathway to reduce proliferation of FLT3‑ITD<sup>+</sup> AML cells and promote apoptosis. Experimental and Therapeutic Medicine, 27, 161. https://doi.org/10.3892/etm.2024.12449
MLA
Su, Y., Wu, M., Zhou, B., Bai, Z., Pang, R., Liu, Z., Zhao, W."Paclitaxel mediates the PI3K/AKT/mTOR pathway to reduce proliferation of FLT3‑ITD<sup>+</sup> AML cells and promote apoptosis". Experimental and Therapeutic Medicine 27.4 (2024): 161.
Chicago
Su, Y., Wu, M., Zhou, B., Bai, Z., Pang, R., Liu, Z., Zhao, W."Paclitaxel mediates the PI3K/AKT/mTOR pathway to reduce proliferation of FLT3‑ITD<sup>+</sup> AML cells and promote apoptosis". Experimental and Therapeutic Medicine 27, no. 4 (2024): 161. https://doi.org/10.3892/etm.2024.12449