AKT1 and CTNNB1 mutations as drivers of paclitaxel resistance in breast cancer cells

  • Authors:
    • Gulsum Altiparmak-Ulbegi
    • Gozde Hasbal-Celikok
    • Pinar Aksoy-Sagirli
  • View Affiliations

  • Published online on: May 2, 2025     https://doi.org/10.3892/ol.2025.15070
  • Article Number: 324
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer (BC) is the most prevalent cancer type in the world, with increasing incidence rates. Drug resistance is a notable factor that limits the effectiveness of BC therapy. Paclitaxel (PTX), a chemotherapeutic agent belonging to the taxane class, is commonly used in BC; however, its efficacy is often compromised by drug resistance, which is primarily attributed to genetic alterations. The phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (AKT) and wingless‑type MMTV integration site family/β‑catenin signaling pathways are involved in essential cellular processes, such as proliferation, apoptosis and maintenance of homeostasis. Dysregulated activation of these pathways is strongly associated with carcinogenesis and drug resistance. In the present study, the potential effects of AKT1 (E17K/E49K/L52R) and catenin β‑1 (CTNNB1; S33P/T41A/S45F) mutations on PTX resistance in BC were investigated in vitro using site‑directed mutagenesis, transient transfection, MTS assay and western blot analyses. The results of the present study indicated that AKT1‑E17K/E49K and CTNNB1‑S45F/T41A mutations induced PTX resistance compared with AKT1‑wild‑type (WT) and CTNNB1‑WT in MCF‑7 cells, respectively. In MDA‑MB‑231 cells, all three AKT1 mutations (E17K/E49K/L52R) triggered PTX resistance compared with AKT1‑WT, while none of the CTNNB1 mutations exhibited such an effect. In conclusion, AKT1 mutations may serve as a biomarker for PTX resistance in both estrogen receptor (ER)(+)/progesterone receptor (PR)(+)/HER2(‑) and triple negative BC, while CTNNB1 mutations may be a potential biomarker for PTX resistance in ER(+)/PR(+)/HER2(‑) BC.

Introduction

According to global cancer statistics for 2022, breast cancer (BC) is the second most frequently diagnosed cancer type, with ~2.3 million new cases (1). As a natural consequence of the growing and aging of the world population, the burden of BC is expected to exceed 3 million new cases and 1 million mortalities in the next two decades (2). BC comprises a group of diseases, exhibiting different clinical behaviors (treatment response/resistance, prognosis and metastatic potential) between subtypes, which reflects its heterogeneous nature (3). Although there are numerous approaches such as hormonal, targeted drugs and chemotherapy in BC treatment, drug resistance is a common phenomenon that limits the treatment efficacy. However, the mechanisms underlying drug resistance are complex and not yet fully understood (4).

The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway is key for numerous cellular processes, including cell cycle progression, proliferation and apoptosis (5). Dysregulation of this pathway is associated with tumor progression and drug resistance in BC. Mutations in genes involved in the PI3K/AKT pathway, such as PI3K, AKT1/2/3 and PTEN, may disturb the regulation of tumor growth, angiogenesis and cell survival, contributing to cancer development (6). As a result, this pathway is a promising target for overcoming drug resistance in BC (7). AKT1, a serine/threonine kinase encoded by the AKT1 gene, is an important signaling protein that serves a key role in the PI3K/AKT pathway (8). AKT1 phosphorylates several targets, such as the mammalian target of rapamycin (mTOR), matrix metalloproteinases, cyclin-dependent kinases and vascular endothelial growth factor, to promote cell survival (9). During the AKT1 activation process, PI3K is primarily activated through the binding of ligands, including growth factors, hormones and cytokines, to receptors. Activated PI3K leads to the formation of phosphatidylinositol-(3,4,5)-trisphosphate (PIP3) from phosphatidylinositol-(4,5)-bisphosphate, facilitating the recruitment of AKT1 to the plasma membrane. The pleckstrin homology (PH) domain is key for the activation of AKT1, which binds to PIP3 via its PH domain and is subsequently phosphorylated by 3′-phosphoinositide-dependent protein kinase 1 and mTOR complex 2 (10). Missense mutations in the PH domain of AKT1, including AKT1-E17K (49G>A), -E49K (145G>A) and -L52R (155T>G), have been demonstrated to increase AKT1 activity (1113). The AKT1-E17K and -L52R mutations have also been observed in patients with BC (11,14). While the AKT1-E49K mutation has been reported in bladder cancer and pulmonary sclerosing hemangioma (12,15), to the best of our knowledge, its presence in BC has not been reported.

The canonical wingless-type MMTV integration site family (WNT) pathway (WNT/β-catenin) is an evolutionarily conserved signaling cascade that regulates key biological processes, including cell fate determination, organogenesis, tissue homeostasis and pathological conditions (such as cancer, osteoporosis and coronary artery disease). Dysregulation of the WNT/β-catenin pathway is associated with carcinogenesis through multiple mechanisms of action (16). In BC, various components of the WNT/β-catenin pathway undergo alteration due to factors such as mutations, amplifications and post-transcriptional/-translational modification. These alterations lead to changes in the localization of β-catenin within the cell (17). The phosphorylation status of cytosolic β-catenin is key to the WNT/β-catenin pathway (18). When the pathway is inactivated, β-catenin binds to the destruction complex that includes axis inhibition protein (AXIN), adenomatous polyposis coli, casein kinase 1 (CK-1) and glycogen synthase kinase 3β (GSK-3β). β-catenin is sequentially phosphorylated by CK-1 on Ser45 and then by GSK-3β on Thr41, Ser37 and Ser33. This phosphorylation leads to ubiquitination and proteasomal degradation of β-catenin (19). In physiological/pathological conditions such as embryogenesis and carcinogenesis, the WNT/β-catenin pathway is activated by binding of WNT family members (WNT1, WNT2, WNT3a, etc.) to the Frizzled (FZD) receptor and its coreceptor, low-density lipoprotein receptor-related protein (LRP) 6 or LRP5. The formation of the WNT-FZD-LRP6/LRP5 complex mediates the assembly of the destruction complex around the receptor, resulting in inhibition of destruction complex-mediated β-catenin phosphorylation and hence accumulation of β-catenin in the cytosol. This accumulation triggers the translocation of β-catenin to the nucleus, where it initiates the expression of target genes essential for cell survival, proliferation, differentiation and migration (20).

Mutations in the catenin β-1 (CTNNB1) gene, which encodes β-catenin, are observed in numerous types of cancer and are predominantly all localized in exon 3. The mutations are primarily found in the phosphorylation sites of β-catenin (N-terminal region; S33, S37, T41 and S45), leading to resistance to phosphorylation. As a result, β-catenin accumulates in the cell, eventually translocating to the nucleus (21). Missense mutations in the phosphorylation sites of β-catenin include CTNNB1-S33P (97T>C), -T41A (121A>G) and -S45F (134C>T). The presence of these mutations is associated with an increase in cellular proliferation (2229). CTNNB1-T41A and -S45F have been reported in BC (30), whereas the presence of CTNNB1-S33P remains unclear.

Paclitaxel (PTX) is a microtubule-stabilizing agent from the taxane class of plant alkaloids that is commonly used in the treatment of BC. PTX induces apoptosis in cancer cells through various mechanisms of action, including activation of antitumor immunity, the regulation of mitochondrial function, cell cycle arrest, downregulation of the PI3K/AKT pathway and production of reactive oxygen species (31,32). Despite its potent apoptotic effects via these mechanisms, the efficacy of PTX is limited, primarily due to the development of drug resistance (33).

Understanding the molecular mechanisms underlying PTX resistance and identifying predictive biomarkers for PTX resistance in BC treatment is key for improving therapeutic outcomes. To the best of our knowledge, no established biomarker exists to predict PTX resistance. Therefore, the present study explored the potential effects of AKT1 (E17K/E49K/L52R) and CTNNB1 (S33P/T41A/S45F) mutations, which are known to enhance AKT1 activity (1113) and β-catenin stability (21), on PTX resistance in in vitro models of BC. The results of the present study may serve as a guide for clinical studies associated with PTX resistance.

Materials and methods

Site-directed mutagenesis

The AKT1 and CTNNB1 mutations were created using a site-directed mutagenesis kit (cat. no. 200519; Agilent Technologies, Inc.) on the pCMV6 vectors with C-terminal Myc-DDK tag carrying AKT1 (cat. no. RC220361; OriGene Technologies, Inc.) and CTNNB1 (cat. no. RC208947; OriGene Technologies, Inc.) following the manufacturer's protocol. Mutagenic primers were designed using the specific design program accessible online at agilent.com/genomics/qcpd and synthesized by Integrated DNA Technologies, Inc. (Table I).

Table I.

Mutagenic primers used to generate AKT1 and CTNNB1 mutations.

Table I.

Mutagenic primers used to generate AKT1 and CTNNB1 mutations.

MutationPrimer sequence (5′-3′)
AKT1
  E17K (Glu17Lys)Forward, GCACAAACGAGGGAAGTACATCAAGAC
  49G>AReverse, GTCTTGATGTACTTCCCTCGTTTGTGC
  E49K (Glu49Lys)Forward, GATGTGGACCAACGTAAGGCTCCCCTCAAC
  145G>AReverse, GTTGAGGGGAGCCTTACGTTGGTCCACATC
  L52R (Leu52Arg)Forward, CGTGAGGCTCCCCGCAACAACTTCTCTG
  155T>GReverse, CAGAGAAGTTGTTGCGGGGAGCCTCACG
CTNNB1
  S33P (Ser33Pro)Forward, CAGTCTTACCTGGACCCTGGAATCCATTC
  97T>CReverse, GAATGGATTCCAGGGTCCAGGTAAGACTG
  T41A (Thr41Ala)Forward, CATTCTGGTGCCACTGCCACAGCTCCTTCTC
  121A>GReverse, GAGAAGGAGCTGTGGCAGTGGCACCAGAATG
  S45F (Ser45Phe)Forward, CTACCACAGCTCCTTTTCTGAGTGGTAAAG
  134C>TReverse, CTTTACCACTCAGAAAAGGAGCTGTGGTAG

[i] CTNNB1, catenin β-1; AKT, protein kinase B.

Transformation and plasmid isolation

Escherichia coli cells (cat. no. C3030-03; Invitrogen; Thermo Fisher Scientific, Inc.) were used to transform mutant and wild-type (WT) AKT1/CTNNB1 pCMV6 vectors, as well as an empty pCMV6 vector (EV) (cat. no. PS100001; OriGene Technologies, Inc.), following the manufacturer's protocol based on the heat-shock method (34). The transformed cell mixtures were plated onto lysogeny broth (LB) agar (cat. no. L3147; Sigma-Aldrich; Merck KGaA) containing 25 µg/ml kanamycin (KAN) (cat. no. 60615; Sigma-Aldrich; Merck KGaA) in Petri dishes and incubated at 37°C overnight. Since the vectors contained the KAN-resistance gene, the cells transformed with these vectors proliferated and formed colonies on LB agar containing KAN. Then the colonies containing transformed cells were collected by a loop and cultured in LB at 37°C for 8 h (cat. no. L3522; Sigma-Aldrich; Merck KGaA). The DNA was purified using an isolation kit (cat. no. 12123; Qiagen, Inc.), according to the manufacturer's instructions. The DNA samples were sequenced using next-generation sequencing by RefGen Gene Research and Biotechnology Company to verify the efficiency of the transformation.

Cell culture

MCF-7 (cat. no. HTB-22) and MDA-MB-231 (cat. no. HTB-26) BC cell lines were obtained from the American Type Culture Collection. MCF-7 cells were maintained in EMEM (cat. no. 320-026-CL; Wisent Biotechnology) supplemented with 10% fetal bovine serum (FBS; cat. no. FBS-11A; Capricorn Scientific GmbH), 100 U/ml penicillin, 100 µg/ml streptomycin (cat. no. 450-201-EL; Wisent Biotechnology) and 0.01 mg/ml recombinant human insulin (cat. no. I9278; Sigma-Aldrich; Merck KGaA). MDA-MB-231 cells were maintained in RPMI-1640 (cat. no. 350-007-CL; Wisent Biotechnology) supplemented with 10% FBS and 100 U/ml penicillin/100 µg/ml streptomycin. The cells were cultured in a humidified incubator with 5% CO2 at 37°C. The cell lines were passaged four times prior to use, with transfection performed at passage four. Following transfection, the cells were passaged a fifth time prior to drug treatment. To minimize cellular stress and ensure more reliable results, a cell scraper was used during passaging.

Plasmid DNA transfection

Transient transfection of the plasmid DNA (5 µg) into the cells was performed using the MegaTran 2.0 polymer-based plasmid DNA transfection agent (cat. no. TT210003; OriGene Technologies, Inc.) following the manufacturer's recommendations. MCF-7 (3×106) and MDA-MB-231 (1.5×106) cells were seeded in Petri dishes at 37°C for 24 h. Subsequently, the cells were transfected at 37°C for 17–18 h. Afterward, the medium containing the transfection complex was removed, fresh medium was added and the cells were incubated at 37°C for 24 h to allow for proliferation. At the end of 24 h, the cells were collected to perform subsequent experiments. The EV served as the transfection control.

Cell viability assay

The viability of cells was analyzed using MTS (cat. no. G111; Promega Corporation). MCF-7 (5×104) and MDA-MB-231 (1×104) cells were seeded into 96-well plates. PTX (cat. no. T7402; Sigma-Aldrich; Merck KGaA) was dissolved in DMSO and serially diluted (1:1) to obtain 16 different PTX doses (100–0.003 µg/ml). The cells were exposed to PTX at 37°C for 24, 48 and 72 h. DMSO was used as the negative control. A total of 20 µl MTS/phenazine methosulfate (cat. no. P9625; Sigma-Aldrich; Merck KGaA) mixture (20:1, volume:volume) was added and the plates were maintained for 1–4 h. The absorbance values were determined at 490 nm and cell death was determined using the following formula: Cell death (%)=[1-(absorbance of the sample/absorbance of the control)] ×100.

Western blot analysis

MCF-7 and MDA-MB-231 cells were lysed with RIPA [50 mM Tris-HCl (pH 8), 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate and 0.1% SDS] and the concentration of the total protein was assessed by the bicinchoninic acid protein assay kit (cat. no. BCA1; Sigma-Aldrich; Merck KGaA). A total of 20 µg/lane total protein was loaded onto the Any kD Mini-PROTEAN tris-glycine eXtreme (TGX) precast gels (cat. no. 4569034; Bio-Rad Laboratories, Inc.) and transferred onto a polyvinylidene difluoride membrane (cat. no. 1704272; Bio-Rad Laboratories, Inc.). The membrane was blocked in Tris-buffered saline with 0.1% Tween 20 containing 5% non-fat dry milk at room temperature for 1 h, and was incubated with anti-Myc-DDK (1:2,000; cat. no. TA50011; OriGene Technologies, Inc.) and anti-β-actin (1:10,000; cat. no. M01263; Boster Biological Technology) primary antibodies for 12 h at 4°C. The anti-Myc-DDK antibody was used due to the C-terminal Myc-DDK tag on pCMV6 vectors carrying the AKT1 and the CTNNB1 genes. The membrane was incubated with secondary antibodies (1:10,000; anti-mouse IgG-HRP; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.; 1:20,000; anti-rabbit IgG-HRP; cat. no. ab205718; Abcam) for 1 h at room temperature. The membrane was incubated with a chemiluminescence substrate (cat. no. 1705060; Bio-Rad Laboratories, Inc.) and imaged using Fusion FX (Vilber Lourmat Sté). The immunoreactive protein (IRP) bands were quantified using ImageJ 1.59 software (National Institutes of Health).

Statistical analysis

The experiments were performed in triplicate. Statistical analysis was performed using SPSS Statistics version 21 (IBM Corp.). The normality of the variables was evaluated using the Shapiro-Wilk test and all variables were found to be normally distributed. Descriptive statistics, including the mean and the standard deviation, were calculated. To compare differences among >2 groups, one-way ANOVA was performed followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Western blot analysis of AKT1/CTNNB1-WT and mutant proteins

BC cells were transfected with the EV and vectors carrying AKT1/CTNNB1-WT and mutations, followed by western blot analysis. As expected, no IRP band was observed for the EV. The presence of IRP bands in the samples from AKT1/CTNNB1-WT and mutant-transfected cells confirmed the success of the transfection process (Fig. 1). IRP levels of AKT1-E17K/E49K/L52R were significantly lower compared with those of AKT1-WT in both cell lines (P<0.05). AKT1-L52R mutant IRP levels were significantly lower than those of AKT1-E17K and -E49K (P<0.05; Fig. 1A and B). However, no significant changes were observed between the IRP levels of CTNNB1-WT/S33P/T41A/S45F (P>0.05; Fig. 1C and D).

Impact of AKT1 mutations on PTX resistance in BC cells

BC cells were incubated with PTX for 24, 48 and 72 h. The PTX responses were similar in both EV-transfected and non-transfected BC cells (P>0.05; Table SI, Table SII, Table SIII, Table SIV, Table SV, Table SVI), indicating that the EV did not have an effect.

PTX induced cell death in EV-transfected MCF-7 cells at 24, 48 and 72 h; however, PTX induced cell death in MCF-7 cells transfected with AKT1-WT only at 72 h. The death of AKT1-WT-transfected MCF-7 cells was significantly decreased compared with EV-transfected MCF-7 cells at all exposure times (P<0.05; Table SI, Table SII, Table SIII). These findings reveal that AKT1-WT is associated with PTX resistance in MCF-7 cells.

PTX exhibited no cytotoxic effects on the cells transfected with AKT1 mutations at 24 and 48 h. Following 72 h, death was observed in the cells carrying AKT1-L52R, while PTX remained ineffective in cells expressing AKT1-E17K and -E49K mutations. In addition, the death of cells expressing AKT1-E17K and -E49K was significantly lower compared with that of cells expressing AKT1-WT (P<0.05); however, no significant difference was noted between AKT1-L52R and AKT1-WT (P>0.05; Table SI, Table SII, Table SIII). These findings indicate that AKT1-E17K and -E49K conferred enhanced resistance to PTX in MCF-7 cells compared with AKT1-WT (Fig. 2A).

In both EV-transfected and AKT1-WT-transfected MDA-MB-231 cells, PTX induced death at 24, 48 and 72 h. In AKT1-WT-transfected cells, cell death was reduced compared with EV-transfected cells at 48 and 72 h (P<0.05; Table SIV, Table SV, Table VI). These results indicated that AKT1-WT was associated with PTX resistance in MDA-MB-231 cells, which was similar to the findings noted in MCF-7 cells.

The effects of AKT1 mutations on the PTX response in MDA-MB-231 cells were also evaluated. The PTX-induced death was significantly higher in cells carrying AKT1-WT compared with those harboring AKT1-E17K/E49K/AKT1-L52R (P<0.05; Table SIV, Table SV, Table VI). These results indicated that AKT1-E17K/E49K/L52R induced higher PTX resistance in MDA-MB-231 cells than AKT1-WT (Fig. 2B).

Impact of CTNNB1 mutations on PTX resistance in BC cells

BC cells were incubated with PTX for 24, 48 and 72 h, and cell death was evaluated. The similar PTX responses in both EV-transfected and non-transfected BC cells confirmed there was no effect exerted by EV on cell death (P>0.05; Table SVII, Table SVIII, Table SIX, Table SX, Table SXI, Table XII).

PTX induced death in transfected MCF-7 cells with CTNNB1-WT at 24, 48 and 72 h. The death of MCF-7 cells carrying CTNNB1-WT was significantly lower compared with that of EV-transfected MCF-7 cells (P<0.05; Table SVII, Table SVIII, Table SIX). These findings revealed that CTNNB1-WT was associated with PTX resistance in MCF-7 cells. Furthermore, the effects of CTNNB1 mutation on the PTX resistance in MCF-7 cells were assessed. PTX exhibited cytotoxic effects on cells expressing CTNNB1-S45F and -T41A only at 72 h. PTX-induced death was observed in cells transfected with CTNNB1-S33P at 24, 48 and 72 h (Table SVII, Table SVIII, Table SIX). PTX-induced cell death of CTNNB1-WT was higher compared with that of CTNNB1-S45F and -T41A (P<0.05); however, no significant difference (P>0.05) was noted between CTNNB1-WT and -S33P (Fig. 3A). These results indicated that CTNNB1-T41A and -S45F display higher PTX resistance compared with CTNNB1-WT in MCF-7 cells.

In MDA-MB-231 cells, PTX induced different death in cells transfected with CTNNB1-WT. The death of cells expressing CTNNB1-WT was significantly decreased compared with that of EV-transfected cells (P<0.05; Table SX, Table SXI, Table SXII). These findings suggest that CTNNB1-WT was associated with PTX resistance in MDA-MB-231 cells.

The impact of CTNNB1 mutations on PTX resistance in MDA-MB-231 cells was also evaluated (Table SX, Table SXI, Table SXII). The death levels of the cells expressing CTNNB1-S33P/S45F/T41A were comparable with those of cells expressing CTNNB1-WT (P>0.05; Fig. 3B). These findings demonstrated that CTNNB1-S33P/S45F/T41A mutations did not confer additional PTX resistance in MDA-MB-231 cells compared with CTNNB1-WT.

Discussion

BC is a heterogeneous group of diseases with different morphology and molecular structures, exhibiting varying biological behaviors and prognoses. Given the heterogeneous nature of BC, the primary challenge in its treatment is drug resistance. The identification of the molecular mechanisms underlying drug resistance and potential predictive biomarkers are key for overcoming this (35,36). The majority of the commonly used predictive biomarkers in BC treatment are estrogen receptor (ER)α, progesterone receptor (PR) and epidermal growth factor 2 (HER2). ERα and PR are used to guide endocrine therapy, while HER2 is used to determine the potential efficacy of anti-HER2 targeted therapies (37).

In the present study, cell lines (MCF-7 and MDA-MB-231) with different genetic profiles were used. The MCF-7 cell line represents a typical luminal subtype of BC characterized by ER(+)/PR(+)/HER2(−) (38). By contrast, the MDA-MB-231 cell line is a typical example of triple negative (TN)BC [ER(−)/PR(−)/HER2(−)] (39). The molecular pathophysiology of TNBC remains poorly understood and the primary strategy of the treatment is taxane-based chemotherapy. By contrast, endocrine therapy is the primary treatment strategy for ER(+)/HER2(−) BC; however, resistance is a common issue in patients with advanced BC, leading to recurrence. To decrease the risk of recurrence, taxane-based chemotherapy is often recommended. Nevertheless, the response rates are generally lower in ER(+)/HER2(−) patients with BC compared with those with HER2(+) or TNBC (40,41).

PTX is a taxane-class chemotherapeutic agent commonly used in BC treatment; however, its effectiveness is limited by drug resistance. The underlying mechanisms of PTX resistance remain unclear. To date, mutations in genes such as multidrug resistance 1, β-tubulin and p53 have been implicated in PTX resistance (33). In addition, elevated PI3K/AKT signaling in BC contributes to PTX resistance via specific mechanisms, such as promoting cell survival, enhancing anaerobic metabolism and inhibiting apoptosis (42). Genetic alterations in the PI3K/AKT pathway are frequently observed in BC, including gain-of-function mutations in PIK3CA and AKT1, as well as mutations in PIK3R1, AKT2, AKT3 and PTEN. These alterations highlight the key role of this pathway in breast carcinogenesis (11,43,44).

AKT1-E17K/E49K/L52R are the PH domain mutations resulting in AKT1 activation in a PI3K-independent manner (1113). The elevated levels of PI3K/AKT signaling due to these mutations lead to enhanced cell survival and oncogenic transformation (12,13,4548). In the present study, western blot analysis indicated that AKT1-E17K/E49K/L52R protein levels were lower compared with those of AKT1-WT in both cell lines. Also, AKT1-L52R protein levels were lower than those of AKT1-E17K and -E49K. The decreased levels of mutant proteins may be due to several factors, including altered protein stability, improper folding or differential interactions with cellular components (such as chaperones and downstream signaling partners). These mutations may affect the overall structural integrity or function of the protein, resulting in a decreased expression level. Additionally, the L52R mutation may disrupt critical interactions within the AKT1 protein, affecting its stability or trafficking within the cell.

The potential effects of AKT1-E17K/E49K/L52R mutations on PTX resistance in BC cells were also investigated in vitro, as well as their impact on protein activity. AKT1-E17K and -E49K mutations conferred increased PTX resistance in MCF-7 cells compared with AKT1-WT. In MDA-MB-231 cells, all three AKT1 mutations (E17K/E49K/L52R) were associated with increased PTX resistance. In our previous study, the association between AKT1-E17K/E49K/L52R mutations and cetuximab, 5-fluorouracil, 7-ethyl-10-hydroxycamptothecin (SN-38), oxaliplatin and irinotecan resistance in colorectal cancer cells was identified (49). The present study indicated similarities to those observed in colorectal cancer regarding drug resistance. Studies performed on patients with BC harboring an AKT1-E17K mutation have reported that the use of AKT inhibitors alone (50,51) or in combination with PTX (52) or fulvestrant (53) increases the response to treatment. The findings of the present study regarding the AKT1-E17K mutation and PTX resistance in BC are consistent with those reported in the literature (4953). To the best of our knowledge, the association between AKT1-E49K and -L52R mutations and drug resistance has not been previously reported in BC.

In addition to the PI3K/AKT pathway, the WNT/β-catenin pathway is associated with BC. Elevated levels of nuclear and/or cytoplasmic β-catenin are commonly observed in BC tissue and are associated with poor clinical outcomes and chemoresistance (5457). Genetic and epigenetic alterations in the WNT/β-catenin pathway lead to enhanced transcription of WNT-targeted genes (such as AXIN2, cyclin D1, epidermal growth factor receptor and myelocytomatosis viral oncogene homolog) in BC (58).

CTNNB1-S33P/T41A/S45F mutations (in exon 3) decrease ubiquitination-mediated proteolysis of β-catenin, causing its nuclear accumulation and activation (2224,27,28). Therefore, in the present study, the association between PTX resistance and the WNT/β-catenin pathway was explored. The results indicate that CTNNB1-S45F and -T41A induce PTX resistance, whereas CTNNB1-S33P did not affect the PTX response in MCF-7 cells. By contrast, CTNNB1-S33P/T41A/S45F mutations did not cause changes in PTX response in MDA-MB-231 cells. The varying effects of CTNNB1-T41A and -S45F mutations on PTX resistance may be attributed to the different genetic profiles of these cell lines. To the best of our knowledge, the relationship between CTNNB1/S33P/T41A/S45F mutations and drug resistance has rarely been investigated (49). A comprehensive analysis of patients with BC performed by Ozcan (59) identified CTNNB1 as a reliable biomarker for predicting the response to taxane-based chemotherapy in ER(+)/HER2(−) BC. The findings of the present study regarding MCF-7 are consistent with previous studies (49,59).

Yoo et al (60) reported that while high mutation rates in TP53, PIK3CA, BRCA2 and ATM serine/threonine kinase genes are observed in patients with TNBC, CTNNB1 mutations are rare. Furthermore, Geyer et al (61) reported that activation of the WNT/β-catenin pathway is notably associated with the TNBC phenotype with poor clinical outcomes but is not triggered by the CTNNB1 exon 3 mutations. The aforementioned studies support the findings of the present study on the association between CTNNB1 mutation and PTX response in MDA-MB-231 cells and suggest that CTNNB1 mutations do not drive PTX resistance in TNBC. Therefore, the activation of the WNT/β-catenin pathway in TNBC may not be linked to CTNNB1 exon 3 mutations, or this activation could be driven by mutations in other exons of CTNNB1 or genes associated with the WNT/β-catenin pathway.

In conclusion, to the best of our knowledge, the present study is the first to demonstrate the association of AKT1 and CTNNB1 mutations with PTX resistance in BC cells. AKT1-E17K/E49K and CTNNB1-S45F/T41A mutations may serve as predictive biomarkers for PTX resistance in patients with BC and ER(+)/PR(+)/HER2(−), while AKT1-E17K/E49K/L52R mutations may be used as indicators of PTX resistance in patients with TNBC. The emergence of resistance to cancer therapies highlights the necessity for personalized treatment approaches.

Although the association between AKT1/CTNNB1 mutations and PTX resistance has been established in the present study, the underlying molecular mechanisms, particularly the roles of these mutations in PTX resistance via the PI3K/AKT and WNT/β-catenin pathways, remain unclear. To the best of our knowledge, the phosphorylation status of AKT1 and β-catenin, as well as their downstream effectors such as mTOR and GSK-3β, have not been investigated. Furthermore, considering the highly heterogeneous nature of BC, the use of only MCF-7 and MDA-MB-231 cell lines limits the generalizability of the present findings. Finally, the absence of combination therapy involving PTX and other widely used chemotherapeutics, and the lack of in vivo experiments are key limitations of the present study. Future studies should incorporate additional HER2(+) BC cell lines, such as BT474 and SK-BR-3, to represent the various BC subtypes. Future studies should also include gene silencing/overexpression experiments, protein-protein interaction assays and phosphorylation-specific analyses to explore the roles of these mutations in PTX resistance via the PI3K/AKT and WNT/β-catenin pathways. To validate the clinical implications of the in vitro results, future studies should also incorporate in vivo xenograft tumor models.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Scientific Research Projects Coordination Unit of Istanbul University (grant no. TDK-2018-30153).

Availability of data and materials

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

Authors' contributions

GAU and PAS designed the study, analyzed and interpreted data and wrote the manuscript. GAU and GHC performed the experiments. GAU and PAS 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.

Glossary

Abbreviations

Abbreviations:

BC

breast cancer

PTX

paclitaxel

PI3K

phosphatidylinositol 3-kinase

AKT

protein kinase B

WT

wild-type

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, Vignat J, Gralow JR, Cardoso F, Siesling S and Soerjomataram I: Current and future burden of breast cancer: Global statistics for 2020 and 2040. Breast. 66:15–23. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Turashvili G and Brogi E: Tumor heterogeneity in breast cancer. Front Med (Lausanne). 4:2272017. View Article : Google Scholar : PubMed/NCBI

4 

Tang Y, Wang Y, Kiani MF and Wang B: Classification, treatment strategy, and associated drug resistance in breast cancer. Clin Breast Cancer. 16:335–343. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Shi X, Wang J, Lei Y, Cong C, Tan D and Zhou X: Research progress on the PI3K/AKT signaling pathway in gynecological cancer. Mol Med Rep. 19:4529–4535. 2019.PubMed/NCBI

6 

Noorolyai S, Shajari N, Baghbani E, Sadreddini S and Baradaran B: The relation between PI3K/AKT signaling pathway and cancer. Gene. 698:120–128. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Dong C, Wu J, Chen Y, Nie J and Chen C: Activation of PI3K/AKT/mTOR pathway causes drug resistance in breast cancer. Front Pharmacol. 12:6286902021. View Article : Google Scholar : PubMed/NCBI

8 

Cidado J and Park BH: Targeting the PI3K/Akt/mTOR pathway for breast cancer therapy. J Mammary Gland Biol Neoplasia. 17:205–216. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Nicholson KM and Anderson NG: The protein kinase B/Akt signaling pathway in human malignancy. Cell Signal. 14:381–395. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Basu A and Lambring CB: Akt isoforms: A family affair in breast cancer. Cancers. 13:34452021. View Article : Google Scholar : PubMed/NCBI

11 

Carpten JD, Faber AL, Horn C, Donoho GP, Briggs SL, Robbins CM, Hostetter G, Bpguslawski S, Moses TY, Savage S, et al: A transforming mutation in the plekstrin homology domain of AKT1 in cancer. Nature. 448:439–444. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Askham JM, Platt F, Chambers PA, Snowden H, Taylor CF and Knowles MA: AKT1 mutations in bladder cancer: Identification of a novel oncogenic mutation that can co-operate with E17K. Oncogene. 29:150–155. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Parikh C, Janakiraman V, Wu WI, Foo CK, Kljavin NM, Chaudhuri S, Stawiski E, Lee B, Lin J, Li H, et al: Disruption of PH-kinase domain interactions leads to oncogenic activation of AKT in human cancers. Proc Natl Acad Sci USA. 109:19368–19373. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Hinz N and Jücker M: Distinct functions of AKT isoforms in breast cancer: A comprehensive review. Cell Commun Signal. 17:1–29. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Jung SH, Kim MS, Lee SH, Park HC, Choi HJ, Maeng L, Min KO, Kim J, Park TI, Shin OR, et al: Whole-exome sequencing identifies recurrent AKT1 mutations in sclerosing hemangioma of lung. Proc Natl Acad Sci USA. 113:10672–10677. 2016. View Article : Google Scholar : PubMed/NCBI

16 

MacDonald BT, Tamai K and He X: Wnt/β-catenin signaling: Components, mechanisms, and diseases. Dev Cell. 17:9–26. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Xu X, Zhang M, Xu F and Jiang S: Wnt signaling in breast cancer: Biological mechanisms, challenges and opportunities. Mol Cancer. 19:1652020. View Article : Google Scholar : PubMed/NCBI

18 

Clevers H: Wnt/β-catenin signaling in development and disease. Cell. 127:469–480. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Kimelman D and Xu W: β-Catenin destruction complex: Insights and questions from a structural perspective. Oncogene. 25:7482–7491. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, Zhou Z, Shu G and Yin G: Wnt/β-catenin signaling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduc Target Ther. 7:32022. View Article : Google Scholar

21 

Gao C, Wang Y, Broaddus R, Sun L, Xue F and Zhang W: Exon 3 mutations of CTNNB1 drive tumorigenesis: A review. Oncotarget. 9:5492–5508. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Van Nhieu JT, Renard CA, Wei Y, Cherqui D, Zafrani ES and Buendia MA: Nuclear accumulation of mutated β-catenin in hepatocellular carcinoma is associated with increased cell proliferation. Am J Pathol. 155:703–710. 1999. View Article : Google Scholar

23 

Tanaka Y, Kato K, Notohara K, Hojo H, Ijiri R, Miyake T, Nagahara N, Sasaki F, Kitagawa N, Nakatani Y and Kobayashi Y: Frequent β-catenin mutation and cytoplasmic/nuclear accumulation in pancreatic solid-pseudo papillary neoplasm. Cancer Res. 61:8401–8404. 2001.PubMed/NCBI

24 

Machin P, Catasus L, Pons C, Muñoz J, Matias-Guiu X and Prat J: CTNNB1 mutations and β-catenin expression in endometrial carcinomas. Hum Pathol. 33:206–212. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Lévy L, Neuveut C, Renard CA, Charneau P, Branchereau S, Gauthier F, Nhieu JTV, Cherqui D, Petit-Bertron AF, Mathieu D and Buendia MA: Transcriptional activation of interleukin-8 by β-catenin-Tcf4. J Biol Chem. 277:42386–42393. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Austinat M, Dunsch R, Wittekind C, Tannapfel A, Gebhardt R and Gaunitz F: Correlation between β-catenin mutations and expression of Wnt-signaling target genes in hepatocellular carcinoma. Mol Cancer. 7:21–30. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Hamada S, Futamura N, Ikuta K, Urakawa H, Kozawa E, Ishiguro N and Nishida Y: CTNNB1 S45F mutation predicts poor efficacy of meloxicam treatment for desmoid tumors: A pilot study. PLoS One. 9:e963912014. View Article : Google Scholar : PubMed/NCBI

28 

An J, Woo HY, Lee Y, Kim HS, Jeong J and Kim SK: Clinicopathological features of 70 desmoid-type fibromatoses confirmed by β-catenin immunohistochemical staining and CTNNB1 mutation analysis. PLoS One. 16:e02506192021. View Article : Google Scholar : PubMed/NCBI

29 

Oulès B, Mourah S, Baroudjian B, Jouenne F, Delyon J, Louveau B, Gruber A, Lebbe C and Battistella M: Clinicopathologic and molecular characterization of melanomas mutated for CTNNB1 and MAPK. Virchows Arch. 480:475–480. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Norkowski E, Masliah-Planchon J, Le Guellec S, Trassard M, Courrèges JB, Charron-Barra C, Terrier P, Bonvalot S, Coindre JM and Laé M: Lower rate of CTNNB1 mutations and higher rate of APC mutations in desmoid fibromatosis of the breast: A series of 134 tumors. Am J Surg Pathol. 44:1266–1273. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Li G, Xu D, Sun J, Zhao S and Zheng D: Paclitaxel inhibits proliferation and invasion and promotes apoptosis of breast cancer cells by blocking activation of the PI3K/AKT signaling pathway. Adv Clin Exp Med. 29:1337–1345. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Zhao S, Tang Y, Wang R and Najafi M: Mechanisms of cancer cell death induction by paclitaxel: An updated review. Apoptosis. 27:647–667. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Alalawy AI: Key genes and molecular mechanisms related to Paclitaxel Resistance. Cancer Cell Int. 24:2442024. View Article : Google Scholar : PubMed/NCBI

34 

Froger A and Hall JE: Transformation of plasmid DNA into E. coli using the heat shock method. J Vis Exp. 2532007.doi: 10.3791/253. PubMed/NCBI

35 

Ke X and Shen L: Molecular targeted therapy of cancer: The progress and future prospect. Front Med. 1:69–75. 2017.

36 

Tarighati E, Keivan H and Mahani H: A review of prognostic and predictive biomarkers in breast cancer. Clin Exp Med. 23:1–16. 2023.PubMed/NCBI

37 

Duffy MJ and Crown J: A personalized approach to cancer treatment: How biomarkers can help. Clin Chem. 54:1770–1779. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Beaver JA, Gustin JP, Yi KH, Rajpurohit A, Thomas M, Gilbert SF, Rosen DM, Park BH and Lauring J: PIK3CA and AKT1 mutations have distinct effects on sensitivity to targeted pathway inhibitors in an isogenic luminal breast cancer model system. Clin Cancer Res. 19:5413–5422. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Welsh J: Animal models for studying prevention and treatment of breast cancer. Animal Models for the Study of Human Disease. Conn PM: Academic Press; Cambridge: pp. 997–1018. 2013, View Article : Google Scholar

40 

Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Burguin A, Diorio C and Durocher F: Breast cancer treatments: Updates and new challenges. J Pers Med. 11:8082021. View Article : Google Scholar : PubMed/NCBI

42 

Kaboli PJ, Imani S, Jomhori M and Ling KH: Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am J Cancer Res. 11:51552021.PubMed/NCBI

43 

Kim MS, Jeong EG, Yoo NJ and Lee SH: Mutational analysis of oncogenic AKT E17K mutation in common solid cancers and acute leukaemias. Br J Cancer. 98:1533–1535. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Lauring J, Park BH and Wolff AC: The phosphoinositide-3-kinase-Akt mTOR pathway as a therapeutic target in breast cancer. J Natl Compre Cancer Netw. 11:670–678. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Guo G, Qiu X, Wang S, Chen Y, Rothman PB, Wang Z and Chen JL: Oncogenic E17K mutation in the plekstrin homology domain of AKT1 promotes v-Abl-mediated pre-B-cell transformation and survival of Pim-deficient cells. Oncogene. 29:3845–3853. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Yang WL, Zhang X and Lin HK: Emerging role of Lys-63 ubiquitination in protein kinase and phosphatase activation and cancer development. Oncogene. 29:4493–4503. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Yi KH, Axtmayer J, Gustin JP, Rajpurohit A and Lauring J: Functional analysis of non-hotspot AKT1 mutants found in human breast cancers identifies novel driver mutations: Implications for personalized medicine. Oncotarget. 4:29–34. 2013. View Article : Google Scholar : PubMed/NCBI

48 

De Marco C, Malanga D, Rinaldo N, De Vita F, Scrima M, Lovisa S, Fabris L, Carriero MV, Franco R, Rizzuto A, et al: Mutant AKT1-E17K is oncogenic in lung epithelial cells. Oncotarget. 6:39634–39650. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Hasbal-Celikok G, Aksoy-Sagirli P, Altiparmak-Ulbegi G and Can A: Identification of AKT1/β-catenin mutations conferring cetuximab and chemotherapeutic drug resistance in colorectal cancer treatment. Oncol Lett. 21:1–10. 2021. View Article : Google Scholar : PubMed/NCBI

50 

Davies BR, Guan N, Logie A, Crafter C, Hanson L, Jacobs V, James N, Dudley P, Jacques K, Ladd B, et al: Tumors with AKT1 E17K mutations are rational targets for single agent or combination therapy with AKT inhibitors. Mol Can Ther. 14:2441–2451. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Hyman DM, Smyth LM, Donoghue MTA, Westin SN, Bedard PL, Dean EJ, Bando H, El–Khoueiry AB, Pérez–Fidalgo JA, Mita A, et al: AKT inhibition in solid tumors with AKT1 mutations. J Clin Oncol. 35:2251–2259. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Schmid P, Abraham J, Chan S, Wheatley D, Brunt AM, Nemsadze G, Baird RD, Park YH, Hall PS, Perren T, et al: Capivasertib plus paclitaxel versus placebo plus paclitaxel as first-line therapy for metastatic triple-negative breast cancer: The PAKT trial. J Clin Oncol. 38:423–433. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Smyth LM, Tamura K, Oliveira M, Ciruelos EM, Mayer IA, Sablin MP, Biganzoli L, Ambrose HJ, Ashton J, Barnicle A, et al: Capivasertib, an AKT kinase inhibitor, as monotherapy or in combination with fulvestrant in patients with AKT1 E17K-Mutant, ER-positive metastatic breast cancer. Clin Cancer Res. 26:3947–3957. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Lin SY, Xia W, Wang JC, Kwong KY, Spohn B, Wen Y, Pestell RG and Hung MC: Beta-catenin, a novel prognostic marker for breast cancer: Its roles in cyclin D1 expression and cancer progression. Proc Natl Acad Sci USA. 97:4262–4266. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Ryo A, Nakamura M, Wulf G, Liou YC and Lu KP: Pin1 regulates turnover and subcellular localization of beta-catenin by inhibiting its interaction with APC. Nat Cell Biol. 3:793–801. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Li S, Li S, Sun Y and Li L: The expression of β-catenin in different subtypes of breast cancer and its clinical significance. Tumor Biol. 35:7693–7698. 2014. View Article : Google Scholar

57 

Samant C, Kale R, Pai KSR, Nandakumar K and Bhonde M: Role of Wnt/β-catenin pathway in cancer drug resistance: Insights into molecular aspects of major solid tumors. Biochem Biophys Res Commun. 729:1503482024. View Article : Google Scholar : PubMed/NCBI

58 

Mukherjee N and Panda CK: Wnt/β-catenin signaling pathway as chemotherapeutic target in breast cancer: An update on pros and cons. Clin Breast Cancer. 20:361–370. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Ozcan G: PTCH1 and CTNNB1 emerge as pivotal predictors of resistance to neoadjuvant chemotherapy in ER+/HER2-breast cancer. Front Oncol. 13:12164382023. View Article : Google Scholar : PubMed/NCBI

60 

Yoo TK, Lee WS, Kim J, Kim MK, Park IA, Kim JH and Han W: Mutational analysis of triple-negative breast cancer using targeted kinome sequencing. J Breast Cancer. 25:1642022. View Article : Google Scholar : PubMed/NCBI

61 

Geyer FC, Lacroix-Triki M, Savage K, Arnedos M, Lambros MB, MacKay A, Natrajan R and Reis-Filho JS: β-catenin pathway activation in breast cancer is associated with triple-negative phenotype but not with CTNNB1 mutation. Mod Pathol. 24:209–231. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 30 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Altiparmak-Ulbegi G, Hasbal-Celikok G and Aksoy-Sagirli P: <em>AKT1</em> and <em>CTNNB1</em> mutations as drivers of paclitaxel resistance in breast cancer cells. Oncol Lett 30: 324, 2025.
APA
Altiparmak-Ulbegi, G., Hasbal-Celikok, G., & Aksoy-Sagirli, P. (2025). <em>AKT1</em> and <em>CTNNB1</em> mutations as drivers of paclitaxel resistance in breast cancer cells. Oncology Letters, 30, 324. https://doi.org/10.3892/ol.2025.15070
MLA
Altiparmak-Ulbegi, G., Hasbal-Celikok, G., Aksoy-Sagirli, P."<em>AKT1</em> and <em>CTNNB1</em> mutations as drivers of paclitaxel resistance in breast cancer cells". Oncology Letters 30.1 (2025): 324.
Chicago
Altiparmak-Ulbegi, G., Hasbal-Celikok, G., Aksoy-Sagirli, P."<em>AKT1</em> and <em>CTNNB1</em> mutations as drivers of paclitaxel resistance in breast cancer cells". Oncology Letters 30, no. 1 (2025): 324. https://doi.org/10.3892/ol.2025.15070