Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

STAT3 Inactivation and Induction of Apoptosis Associate With Fluoxetine-inhibited Epithelial-mesenchymal Transition and Growth of Triple-negative Breast Cancer In Vivo

PEN-AN LIAO, PEI-YI CHU, ZHAO-LIN TAN, FEI-TING HSU, YANG-CHENG LEE and HSING-JU WU
Anticancer Research August 2022, 42 (8) 3807-3814; DOI: https://doi.org/10.21873/anticanres.15871
PEN-AN LIAO
1Department of Radiology, Cathay General Hospital, Taipei, Taiwan, R.O.C.;
2School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
PEI-YI CHU
2School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, R.O.C.;
3Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan, R.O.C.;
4Department of Pathology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.;
5Department of Health Food, Chung Chou University of Science and Technology, Changhua, Taiwan, R.O.C.;
6National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ZHAO-LIN TAN
7Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
FEI-TING HSU
7Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YANG-CHENG LEE
8Division of Hematology/Oncology, Department of Internal Medicine, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: leeyangjason2@gmail.com hildawu09@gmail.com
HSING-JU WU
9Research Assistant Center, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.;
10Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua, Taiwan, R.O.C.;
11Department of Biology, National Changhua University of Education, Changhua, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: leeyangjason2@gmail.com hildawu09@gmail.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Breast cancer (BC) is the most common cancer and second leading cause of death in women worldwide. Triple-negative breast cancer (TNBC) is the most aggressive type of BC, while the treatment option is limited and has long been considered as a major unmet need. Meta-analysis indicated the anti-tumor potential of anti-depressants, especially selective serotonin-reuptake inhibitors (SSRIs). The SSRI fluoxetine has been shown to suppress BC and ovarian cancer cell growth; however, whether it suppresses tumor progression in vivo is unclear. Materials and Methods: We established an 4T1 bearing animal model, an orthotopic TNBC model, to identify the mechanism and therapeutic efficacy of fluoxetine. Results: Tumor growth evaluated by caliper and computed tomography scan demonstrated the inhibition effect by fluoxetine treatment. Immunohistochemistry showed that the expression of STAT3-mediated epithelial-to-mesenchymal transition (EMT) proteins and apoptosis-related proteins was decreased. Conclusion: Fluoxetine may induce an anti-TNBC effect via inactivating STAT3 signaling transduction and triggering the caspase-mediated apoptotic pathway.

Key Words:
  • Fluoxetine
  • triple-negative breast cancer
  • STAT3
  • apoptosis
  • epithelial-to-mesenchymal transition

Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of estrogen receptor (ER), progesterone receptor (ER), and human epidermal growth factor receptor 2 (HER2) expression. TNBC has poor outcome due to the high recurrence and metastasis rate (1, 2). The standard neoadjuvant chemotherapy regimen comprising adriamycin, cyclophosphamide, and taxane is used for the treatment of TNBC. For further improvement of pathological complete response of TNBC patients, potential drug combinations have been developing. New therapeutic strategies such as the combination of poly ADP-ribose polymerase 1 (PARP-1) inhibitor and/or immune check point inhibitor has been shown to increase anti-TNBC efficacy of chemotherapy (3, 4).

Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor that participates in tumor growth, angiogenesis, metastasis, and evasion of apoptosis by inducing the expression of downstream target genes (5). Constitutively activated STAT3 signaling is an essential mediator of TNBC. The inhibition of STAT3 signaling has been found to mediate TNBC suppression but also up-regulate the sensitivity of TNBC cells to therapeutic agents including olaparib (PARP-1 inhibitor), doxorubicin, and docetaxel. Therefore, STAT3 has been recognized as a potential therapeutic target for the treatment of TNBC (2, 6-8).

Efficient induction of apoptosis is required for therapeutic agents-elicited tumor regression. Caspase-mediated apoptosis is activated through extrinsic and intrinsic pathways (9). Several antidepressants have been associated with anti-cancer effects against TNBC (10, 11). For instance, imipramine, a tricyclic antidepressant, has been reported to induce tumor regression through the induction of apoptosis, cell cycle arrest, and disruption of DNA repair activity in TNBC both in vitro and in vivo (10). Fluoxetine, a selective serotonin reuptake inhibitor (SSRI), has been demonstrated to trigger intrinsic apoptosis, resulting in inhibition of TNBC cell growth (11). Previous studies demonstrated that fluoxetine, as a complementary agent, reduced tumor progression through attenuation of nuclear factor-kappaB (NF-Embedded ImageB) signaling or the induction of apoptosis in non-small cell lung cancer (NSCLC), hepatocellular carcinoma (HCC), and gastric adenocarcinoma (12-15). However, the anti-TNBC effect of fluoxetine has not yet been understood. The main purpose of the present study was to investigate the anticancer efficacy and mechanism of fluoxetine in TNBC in vivo.

Materials and Methods

Cell culture. 4T1 cells were purchased from the American Type Culture Collection (ATCC). Cells were cultured in DMEM, containing 10% fetal bovine serum, 1% penicillin and streptomycin, and 2 mM L-glutamine (Hyclone Laboratories LLC, Logan, UT, USA) in a humidified incubator at 37°C containing 5% CO2.

Animal experiments. All procedures followed the China Medical University IACUC guidelines (Approval number: CMUIACUC-2022-299). Six-week-old male BALB/cByJNarl mice were purchased from the National Laboratory Animal Center (Taipei, Taiwan). 1×105 4T1 cells were resuspended in PBS (containing 30% Matrigel) with the 29G needle tip turned upwards, to enter the skin subcutaneously at 10 mm from the upper inoculation nipple (16); treatment was started when the tumor size reached approximately 50 mm3 as measured by a digital caliper. Tumor volume was measured every three days and calculated using the following formula: Volume=Height×Weight2×0.523. Mice were randomly divided into three group: Control (0.1% DMSO in 100 μl distilled water/day), fluoxetine (5 mg/kg fluoxetine in 100 μl distilled water/day), and fluoxetine (10 mg/kg fluoxetine in 100 μl distilled water/day). All treatments were delivered through gavage. Mice were humanely sacrificed post treatment on day 22, and the tumor and organs were collected for further experiments.

In vivo computed tomography (CT) scanning. After twenty-one days of treatment, the tumor size of mice was assessed by CT scan (Mediso Ltd., Budapest, Hungary). 4T1 bearing mice were sedated by using 1-3% isoflurane. The scanning parameters were listed as follows: tube energy=55 kVp×145 μA; direction=360°; Voxel size=145×145×145 μm (17).

Immunohistochemistry (IHC) staining. After 22 days’ treatment, mice were sacrificed. Tumors were isolated and fixed with 10% neutral formalin solution (containing 4% paraformaldehyde) at 4°C. Paraffin-embedded tumor specimens were sliced into 5-μm thick specimens by Bio-Check Laboratories Ltd. (New Taipei City, Taiwan, ROC). IHC staining was used to evaluate protein expression according to the instructions of the IHC kit manufacturer (DAB500, Millipore, Burlington, VT, USA). Primary antibodies STAT3 (Tyr705) (#9145, Cell Signaling Technology, Danvers, MA, USA, Twist (#46702, Cell Signaling Technology), SNAIL (#3879, Cell Signaling Technology), SLUG (#9585, Cell Signaling Technology), E-cadherin (#3195, Cell Signaling Technology), N-cadherin (#13116, Cell Signaling Technology), ZEB1 (#70512, Cell Signaling Technology), ZEB2 (#97885, Cell Signaling Technology), cleaved caspase-3 (#9661, Cell Signaling Technology), cleaved caspase-8 (#9496, Cell Signaling Technology), cleaved caspase-9 (#9509, Cell Signaling Technology) and Ki-67(E-AB-63523, Elabscience) were used in the present experiment (18).

Hematoxylin & Eosin staining. Mice were sacrificed on day 22. The heart, lung, kidney, liver, spleen, and small intestine were fixed with 10% neutral formalin solution at 4°C. Paraffin-embedded tumor specimens were sliced into 5-μm thick specimens and H&E staining was performed by Bio-Check Laboratories (New Taipei City, Taiwan, ROC) (19).

Statistical analysis. Statistical analysis was performed by Microsoft Excel 2017 version (Redmond, WA, USA) and One-way ANOVA and student t-test was used to evaluate significant differences. A p-value <0.05 defined a statistically significant difference. Data are displayed as mean±standard.

Results

Fluoxetine inhibited tumor growth in a 4T1 orthotopic mice model. In order to investigate the anti-tumor effect of fluoxetine in vivo, a breast cancer model was established by orthotopic injection (4T1 cells) in the upper nipple (as indicated in methods). The design of the animal experiments is presented in Figure 1A. The tumor volume of mice treated with 10 mg/kg fluoxetine was significantly smaller than that of the CTRL group (p-value <0.01) after twelve days of treatment (Figure 1B). Tumor growth inhibition by fluoxetine was also dose-depended; 10 mg/kg fluoxetine showed greater tumor growth inhibition than 5 mg/kg fluoxetine. After humanely sacrificing the mice, tumors were isolated, imaged, and weighted (Figure 1C-E). As indicated in Figure 1C-E, the 10 mg/kg fluoxetine group showed superior tumor size and weight inhibition than the 5 mg/kg fluoxetine group (p-value <0.01). Furthermore, in order to provide a more accurate assessment of the 3D tumor growth pattern of each treatment group in vivo, we performed CT scanning on days 0 and 21. A lateral, frontal, and transverse view from one representative mouse of each group is displayed in Figure 1F. Notable tumor growth inhibition was observed in the 5 mg/kg and 10 mg/kg fluoxetine groups compared to the 0.1% CTRL group. Taken together, our results indicated that fluoxetine inhibited 4T1 tumor growth.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Fluoxetine inhibited the growth of 4T1 tumor. (A) Flow chart of orthotopic animal model is displayed. (B) Tumor growth, (C-D) isolated tumor photograph, and (E) tumor weight on day 22 are shown. (F) CT scanning results from one representative mouse of each group on days 0 and 21 are presented. (*p<0.05, **p<0.01, ***p<0.005 vs. CTRL; $p<0.05, $$p<0.01 vs. 5 mg/kg fluoxetine).

Fluoxetine induced expression of pro-apoptosis related proteins and inhibited expression of proliferation-related proteins in 4T1 orthotopic mice model. After 22 days of treatment, mice were sacrificed, and tumors were isolated and sections from them were analyzed by using IHC staining. The expression of pro-apoptotic proteins such as cleaved caspase-3, extrinsic signaling marker cleaved caspase-8, and intrinsic signaling marker cleaved caspase-9 were analyzed after treatment (Figure 2A and 2B). As indicated in Figure 2A and B, fluoxetine induced apoptosis of 4T1 tumor cells via activating both the extrinsic and intrinsic apoptosis signaling pathways. Additionally, expression of the tumor proliferation marker Ki-67 was examined in 4T1 tumor sections (Figure 2C and 2D). The expression levels of Ki-67 in the 4T1 tumor sections were significantly inhibited by fluoxetine treatment (p-value<0.01). These results indicated that fluoxetine may not only trigger apoptosis but may also reduce the proliferation capacity of 4T1 tumor cells.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Fluoxetine induced apoptosis and inhibited proliferation. (A, C) Protein expression of cleaved-caspase-3, -8, -9, and ki-67 in tumor sections from each group are displayed. (B, D) The quantified expression levels of cleaved-caspase-3, -8, -9, and ki-67 from each group is presented in a bar chart. (***p<0.005 vs. CTRL; $$$p<0.005 vs. 5 mg/kg fluoxetine).

Fluoxetine diminished STAT3 protein expression and its mediated epithelial mesenchymal transition (EMT)-related protein expression in a 4T1 orthotopic mice model. Next, we investigated whether STAT3 and its related EMT factors (18) were affected by fluoxetine treatment. As illustrated in Figure 3A and 3B, the phosphorylation of STAT3 (Tyr705) was suppressed by fluoxetine. Furthermore, E-cadherin, which suppresses EMT, was increased by fluoxetine (Figure 3C and 3D). In contrast, factors that may promote EMT, such as N-cadherin, Slug, Snail, ZEB1, and ZEB2 were decreased by fluoxetine (Figure 3C-F). Taken together, we suggest that fluoxetine-induced inhibition of EMT may associate with the reduction of the phosphorylation of STAT3 at the Tyr705 site (inactivation of STAT3).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Fluoxetine induced STAT3 and epithelial-mesenchymal transition-related proteins inhibition. (A, C, F) Protein expression pattern of STAT3 (Tyr705), E-cadherin, N-cadherin, Slug, Snail, ZEB1, and ZEB2 in tumor sections from each group are displayed. (B, D, E, G) The quantified expression levels of these proteins from each group are presented in a bar chart. (***p<0.005 vs. CTRL; $$p<0.01 vs. 5 mg/kg fluoxetine).

Mice body weight and normal organ morphology were not affected by fluoxetine treatment in the 4T1 orthotopic mice model. To investigate whether fluoxetine induces toxicity in mice, we weighted the mice every three days (Figure 4A). There was no statistically significant difference between the body weight of mice in the fluoxetine-treated mice and that of CTRL group (p-value> 0.05), which indicated no obvious acute toxicity induced by fluoxetine treatment. Furthermore, we examined the normal organ (heart, lung, liver, spleen, kidney, and small intestine) morphology using H&E staining, as shown in Figure 4B. Therefore, fluoxetine treatment is safe in the dosage used to inhibit tumor growth. No obvious general toxicity was observed in 4T1 orthotopic mice.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

No general toxicity was found after fluoxetine treatment in a 4T1 orthotopic mice model. (A) Body weight of mice on day 0 to 21. (B) H&E staining of the heart, lung, liver, spleen, kidney, and small intestine of each treatment group is shown.

Discussion

Extrinsic and intrinsic pathways mediate apoptosis through the activation of caspase-8 and caspase-9, respectively. Activated caspase-8 and -9 trigger expression of cleaved caspase-3, -6, and -7, which lead to cell death. Cleaved caspase-3 is a central player in apoptosis, specifically for the induction of apoptotic DNA fragmentation and the cleavage of poly [ADP-ribose] polymerase 1 (9, 21, 22). Bowie et al. found that the intrinsic pathway participated in fluoxetine-induced apoptosis in TNBC in vitro (11). Our data showed that the increased expression of cleaved caspase-3, -8, and -9 was significantly augmented by treatment with fluoxetine. According to these data, it is suggested that fluoxetine induces apoptosis through both the extrinsic and intrinsic pathways in TNBC in vivo (Figure 2A and B).

The EMT is the conversion of non-invasive epithelial cells to an invasive mesenchymal phenotype. EMT-related proteins such as N-cadherin, Slug, Snail, Twist, ZEB1, and ZEB2 facilitate tumor metastasis through up-regulation of EMT (23-25). E-cadherin, a crucial component of cell-cell adhesion junctions, restrains tumor invasion and metastasis by preventing tumor cell dissociation. Loss of E-cadherin has been recognized as an important hallmark of EMT (26, 27). The decreased expression of E-cadherin has been associated with unfavorable chemotherapy response and prognosis in TNBC (28). Our results indicated that fluoxetine not only effectively reduced tumor growth but also induced E-cadherin expression in TNBC in vivo (Figure 1B and Figure 3C).

Slug, Snail, Twist, ZEB1, and ZEB2 as E-cadherin repressors, inhibit E-cadherin expression to promote metastasis in various cancers (29-31). Constitutive STAT3 signaling is associated with a high expression of EMT-related proteins that correlate with metastasis and poor survival of breast cancers (32-36). The reduced STAT3 signaling down-regulates EMT and associates with a favorable TNBC survival (32, 37). Furthermore, inhibition of EMT-related proteins also contributes to a reduction in metastasis in TNBC (38-40). The results showed that the protein levels of STAT3(Try705), N-cadherin, slug, snail, Twist, ZEB1, and ZEB2 were significantly decreased by treatment with fluoxetine in TNBC in vivo (Figure 3C-G).

In conclusion, this study indicated that fluoxetine induces apoptosis through extrinsic/intrinsic pathways while suppresses tumor growth, STAT3 signaling, and expression of EMT-related proteins. We suggested that induction of apoptosis and STAT3 inactivation are associated with fluoxetine-induced inhibition of cell growth and EMT in TNBC.

Acknowledgements

The Authors thank the Medical Research Core Facilities Center, Office of Research & Development at China Medical University (Taichung, Taiwan, R.O.C) for the technical support.

Footnotes

  • Authors’ Contributions

    PAL, PYC, ZLT performed the experiments, derived the models and analyzed the data. ZLT and FTH prepared the initial version of the paper. FTH, YCL, and HJW conceived of the presented idea, supervised the findings of this work, performed the literature review, and prepared the final versions of the paper.

  • Conflicts of Interest

    The Authors declare no competing financial interests regarding this study.

  • Funding

    This study was supported by Tainan Municipal Hospital, Tainan, Taiwan (RD-108-04) and Show Chwan Memorial Hospital, Changhua, Taiwan (ID: SRD-110033), respectively.

  • Received June 6, 2022.
  • Revision received June 24, 2022.
  • Accepted June 25, 2022.
  • Copyright © 2022 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

References

  1. ↵
    1. Howard CM,
    2. Estrada M,
    3. Terrero D,
    4. Tiwari AK and
    5. Raman D
    : Identification of cardiac glycosides as novel inhibitors of eIF4A1-mediated translation in triple-negative breast cancer cells. Cancers (Basel) 12(8): 2169, 2020. PMID: 32759815. DOI: 10.3390/cancers12082169
    OpenUrlCrossRefPubMed
  2. ↵
    1. Qin JJ,
    2. Yan L,
    3. Zhang J and
    4. Zhang WD
    : STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review. J Exp Clin Cancer Res 38(1): 195, 2019. PMID: 31088482. DOI: 10.1186/s13046-019-1206-z
    OpenUrlCrossRefPubMed
  3. ↵
    1. Lee JS,
    2. Yost SE and
    3. Yuan Y
    : Neoadjuvant treatment for triple negative breast cancer: recent progresses and challenges. Cancers (Basel) 12(6): 1404, 2020. PMID: 32486021. DOI: 10.3390/cancers12061404
    OpenUrlCrossRefPubMed
  4. ↵
    1. Damaskos C,
    2. Garmpi A,
    3. Nikolettos K,
    4. Vavourakis M,
    5. Diamantis E,
    6. Patsouras A,
    7. Farmaki P,
    8. Nonni A,
    9. Dimitroulis D,
    10. Mantas D,
    11. Antoniou EA,
    12. Nikolettos N,
    13. Kontzoglou K and
    14. Garmpis N
    : Triple-negative breast cancer: The progress of targeted therapies and future tendencies. Anticancer Res 39(10): 5285-5296, 2019. PMID: 31570423. DOI: 10.21873/anticanres.13722
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Sun CY,
    2. Nie J,
    3. Huang JP,
    4. Zheng GJ and
    5. Feng B
    : Targeting STAT3 inhibition to reverse cisplatin resistance. Biomed Pharmacother 117: 109135, 2019. PMID: 31226634. DOI: 10.1016/j.biopha.2019.109135
    OpenUrlCrossRefPubMed
  6. ↵
    1. Marijon H,
    2. Lee DH,
    3. Ding L,
    4. Sun H,
    5. Gery S,
    6. de Gramont A and
    7. Koeffler HP
    : Co-targeting poly(ADP-ribose) polymerase (PARP) and histone deacetylase (HDAC) in triple-negative breast cancer: Higher synergism in BRCA mutated cells. Biomed Pharmacother 99: 543-551, 2018. PMID: 29902865. DOI: 10.1016/j.biopha.2018.01.045
    OpenUrlCrossRefPubMed
    1. Tzeng YT,
    2. Liu PF,
    3. Li JY,
    4. Liu LF,
    5. Kuo SY,
    6. Hsieh CW,
    7. Lee CH,
    8. Wu CH,
    9. Hsiao M,
    10. Chang HT and
    11. Shu CW
    : Kinome-wide siRNA screening identifies Src-enhanced resistance of chemotherapeutic drugs in triple-negative breast cancer cells. Front Pharmacol 9: 1285, 2018. PMID: 30473665. DOI: 10.3389/fphar.2018.01285
    OpenUrlCrossRefPubMed
  7. ↵
    1. Liu CY,
    2. Chen KF,
    3. Chao TI,
    4. Chu PY,
    5. Huang CT,
    6. Huang TT,
    7. Yang HP,
    8. Wang WL,
    9. Lee CH,
    10. Lau KY,
    11. Tsai WC,
    12. Su JC,
    13. Wu CY,
    14. Chen MH,
    15. Shiau CW and
    16. Tseng LM
    : Sequential combination of docetaxel with a SHP-1 agonist enhanced suppression of p-STAT3 signaling and apoptosis in triple negative breast cancer cells. J Mol Med (Berl) 95(9): 965-975, 2017. PMID: 28578456. DOI: 10.1007/s00109-017-1549-x
    OpenUrlCrossRefPubMed
  8. ↵
    1. Pfeffer CM and
    2. Singh ATK
    : Apoptosis: a target for anticancer therapy. Int J Mol Sci 19(2): 448, 2018. PMID: 29393886. DOI: 10.3390/ijms19020448
    OpenUrlCrossRefPubMed
  9. ↵
    1. Timilsina S,
    2. Rajamanickam S,
    3. Rao A,
    4. Subbarayalu P,
    5. Nirzhor S,
    6. Abdelfattah N,
    7. Viswanadhapalli S,
    8. Chen Y,
    9. Jatoi I,
    10. Brenner A,
    11. Rao MK,
    12. Vadlamudi R and
    13. Kaklamani V
    : The antidepressant imipramine inhibits breast cancer growth by targeting estrogen receptor signaling and DNA repair events. Cancer Lett 540: 215717, 2022. PMID: 35568265. DOI: 10.1016/j.canlet.2022.215717
    OpenUrlCrossRefPubMed
  10. ↵
    1. Bowie M,
    2. Pilie P,
    3. Wulfkuhle J,
    4. Lem S,
    5. Hoffman A,
    6. Desai S,
    7. Petricoin E,
    8. Carter A,
    9. Ambrose A,
    10. Seewaldt V,
    11. Yu D and
    12. Ibarra Drendall C
    : Fluoxetine induces cytotoxic endoplasmic reticulum stress and autophagy in triple negative breast cancer. World J Clin Oncol 6(6): 299-311, 2015. PMID: 26677444. DOI: 10.5306/wjco.v6.i6.299
    OpenUrlCrossRefPubMed
  11. ↵
    1. Chen WT,
    2. Hsu FT,
    3. Liu YC,
    4. Chen CH,
    5. Hsu LC and
    6. Lin SS
    : Fluoxetine induces apoptosis through extrinsic/intrinsic pathways and inhibits ERK/NF-κB-modulated anti-apoptotic and invasive potential in hepatocellular carcinoma cells in vitro. Int J Mol Sci 20(3): 757, 2019. PMID: 30754643. DOI: 10.3390/ijms20030757
    OpenUrlCrossRefPubMed
    1. Hsu LC,
    2. Tu HF,
    3. Hsu FT,
    4. Yueh PF and
    5. Chiang IT
    : Beneficial effect of fluoxetine on anti-tumor progression on hepatocellular carcinoma and non-small cell lung cancer bearing animal model. Biomed Pharmacother 126: 110054, 2020. PMID: 32145588. DOI: 10.1016/j.biopha.2020.110054
    OpenUrlCrossRefPubMed
    1. Wu JY,
    2. Lin SS,
    3. Hsu FT and
    4. Chung JG
    : Fluoxetine inhibits DNA repair and NF-κB-modulated metastatic potential in non-small cell lung cancer. Anticancer Res 38(9): 5201-5210, 2018. PMID: 30194168. DOI: 10.21873/anticanres.12843
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Khing TM,
    2. Po WW and
    3. Sohn UD
    : Fluoxetine enhances anti-tumor activity of paclitaxel in gastric adenocarcinoma cells by triggering apoptosis and necroptosis. Anticancer Res 39(11): 6155-6163, 2019. PMID: 31704843. DOI: 10.21873/anticanres.13823
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Bieniasz-Krzywiec P,
    2. Martín-Pérez R,
    3. Riera-Domingo C and
    4. Mazzone M
    : Isolation and separation of murine tumor-associated macrophages (TAMs) subpopulations from orthotopic 4T1 breast tumors. STAR Protoc 2(2): 100481, 2021. PMID: 33982015. DOI: 10.1016/j.xpro.2021.100481
    OpenUrlCrossRefPubMed
  14. ↵
    1. Weng MC,
    2. Li MH,
    3. Chung JG,
    4. Liu YC,
    5. Wu JY,
    6. Hsu FT and
    7. Wang HE
    : Apoptosis induction and AKT/NF-κB inactivation are associated with regroafenib-inhibited tumor progression in non-small cell lung cancer in vitro and in vivo. Biomed Pharmacother 116: 109032, 2019. PMID: 31163381. DOI: 10.1016/j.biopha.2019.109032
    OpenUrlCrossRefPubMed
  15. ↵
    1. Su CM,
    2. Lin SS,
    3. Wang HC,
    4. Hsu FT,
    5. Chung JG and
    6. Hsu LC
    : The inhibitory effect and mechanism of quetiapine on tumor progression in hepatocellular carcinoma in vivo. Environ Toxicol 37(1): 92-100, 2022. PMID: 34626444. DOI: 10.1002/tox.23380
    OpenUrlCrossRefPubMed
  16. ↵
    1. Lee KC,
    2. Chen WT,
    3. Liu YC,
    4. Lin SS and
    5. Hsu FT
    : Amentoflavone inhibits hepatocellular carcinoma progression through blockage of ERK/NF-κB activation. In Vivo 32(5): 1097-1103, 2018. PMID: 30150431. DOI: 10.21873/invivo.11351
    OpenUrlAbstract/FREE Full Text
    1. Lin WH,
    2. Chang YW,
    3. Hong MX,
    4. Hsu TC,
    5. Lee KC,
    6. Lin C and
    7. Lee JL
    : STAT3 phosphorylation at Ser727 and Tyr705 differentially regulates the EMT-MET switch and cancer metastasis. Oncogene 40(4): 791-805, 2021. PMID: 33262462. DOI: 10.1038/s41388-020-01566-8
    OpenUrlCrossRefPubMed
  17. ↵
    1. Chiang IT,
    2. Chen WT,
    3. Tseng CW,
    4. Chen YC,
    5. Kuo YC,
    6. Chen BJ,
    7. Weng MC,
    8. Lin HJ and
    9. Wang WS
    : Hyperforin inhibits cell growth by inducing intrinsic and extrinsic apoptotic pathways in hepatocellular carcinoma cells. Anticancer Res 37(1): 161-167, 2017. PMID: 28011486. DOI: 10.21873/anticanres.11301
    OpenUrlAbstract/FREE Full Text
  18. ↵
    1. Akpolat M,
    2. Oz ZS,
    3. Gulle K,
    4. Hamamcioglu AC,
    5. Bakkal BH and
    6. Kececi M
    : X irradiation induced colonic mucosal injury and the detection of apoptosis through PARP-1/p53 regulatory pathway. Biomed Pharmacother 127: 110134, 2020. PMID: 32361637. DOI: 10.1016/j.biopha.2020.110134
    OpenUrlCrossRefPubMed
  19. ↵
    1. Wang M,
    2. Ren D,
    3. Guo W,
    4. Huang S,
    5. Wang Z,
    6. Li Q,
    7. Du H,
    8. Song L and
    9. Peng X
    : N-cadherin promotes epithelial-mesenchymal transition and cancer stem cell-like traits via ErbB signaling in prostate cancer cells. Int J Oncol 48(2): 595-606, 2016. PMID: 26647992. DOI: 10.3892/ijo.2015.3270
    OpenUrlCrossRefPubMed
    1. Babaei G,
    2. Aziz SG and
    3. Jaghi NZZ
    : EMT, cancer stem cells and autophagy; The three main axes of metastasis. Biomed Pharmacother 133: 110909, 2021. PMID: 33227701. DOI: 10.1016/j.biopha.2020.110909
    OpenUrlCrossRefPubMed
  20. ↵
    1. Pinto F,
    2. Costa ÂM, Andrade RP and
    3. Reis RM
    : Brachyury is associated with glioma differentiation and response to temozolomide. Neurotherapeutics 17(4): 2015-2027, 2020. PMID: 32785847. DOI: 10.1007/s13311-020-00911-9
    OpenUrlCrossRefPubMed
  21. ↵
    1. Adhikary A,
    2. Chakraborty S,
    3. Mazumdar M,
    4. Ghosh S,
    5. Mukherjee S,
    6. Manna A,
    7. Mohanty S,
    8. Nakka KK,
    9. Joshi S,
    10. De A,
    11. Chattopadhyay S,
    12. Sa G and
    13. Das T
    : Inhibition of epithelial to mesenchymal transition by E-cadherin up-regulation via repression of slug transcription and inhibition of E-cadherin degradation: dual role of scaffold/matrix attachment region-binding protein 1 (SMAR1) in breast cancer cells. J Biol Chem 289(37): 25431-25444, 2014. PMID: 25086032. DOI: 10.1074/jbc.M113.527267
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Kaszak I,
    2. Witkowska-Piłaszewicz O,
    3. Niewiadomska Z,
    4. Dworecka-Kaszak B,
    5. Ngosa Toka F and
    6. Jurka P
    : Role of cadherins in cancer-a review. Int J Mol Sci 21(20): 7624, 2020. PMID: 33076339. DOI: 10.3390/ijms21207624
    OpenUrlCrossRefPubMed
  23. ↵
    1. Liu X,
    2. Jiang S,
    3. Tian X and
    4. Jiang Y
    : Expression of cleaved caspase-3 predicts good chemotherapy response but poor survival for patients with advanced primary triple-negative breast cancer. Int J Clin Exp Pathol 11(9): 4363-4373, 2018. PMID: 31949833.
    OpenUrlPubMed
  24. ↵
    1. Gao T,
    2. Li JZ,
    3. Lu Y,
    4. Zhang CY,
    5. Li Q,
    6. Mao J and
    7. Li LH
    : The mechanism between epithelial mesenchymal transition in breast cancer and hypoxia microenvironment. Biomed Pharmacother 80: 393-405, 2016. PMID: 27133080. DOI: 10.1016/j.biopha.2016.02.044
    OpenUrlCrossRefPubMed
    1. Galván JA,
    2. Zlobec I,
    3. Wartenberg M,
    4. Lugli A,
    5. Gloor B,
    6. Perren A and
    7. Karamitopoulou E
    : Expression of E-cadherin repressors SNAIL, ZEB1 and ZEB2 by tumour and stromal cells influences tumour-budding phenotype and suggests heterogeneity of stromal cells in pancreatic cancer. Br J Cancer 112(12): 1944-1950, 2015. PMID: 25989272. DOI: 10.1038/bjc.2015.177
    OpenUrlCrossRefPubMed
  25. ↵
    1. Vesuna F,
    2. van Diest P,
    3. Chen JH and
    4. Raman V
    : Twist is a transcriptional repressor of E-cadherin gene expression in breast cancer. Biochem Biophys Res Commun 367(2): 235-241, 2008. PMID: 18062917. DOI: 10.1016/j.bbrc.2007.11.151
    OpenUrlCrossRefPubMed
  26. ↵
    1. Wendt MK,
    2. Balanis N,
    3. Carlin CR and
    4. Schiemann WP
    : STAT3 and epithelial-mesenchymal transitions in carcinomas. JAKSTAT 3(1): e28975, 2014. PMID: 24843831. DOI: 10.4161/jkst.28975
    OpenUrlCrossRefPubMed
    1. Li X,
    2. Li Y,
    3. Du X,
    4. Wang X,
    5. Guan S,
    6. Cao Y,
    7. Jin F and
    8. Li F
    : HES1 promotes breast cancer stem cells by elevating Slug in triple-negative breast cancer. Int J Biol Sci 17(1): 247-258, 2021. PMID: 33390847. DOI: 10.7150/ijbs.53477
    OpenUrlCrossRefPubMed
    1. Chang HY,
    2. Tseng YK,
    3. Chen YC,
    4. Shu CW,
    5. Lin MI,
    6. Liou HH,
    7. Fu TY,
    8. Lin YC,
    9. Ger LP,
    10. Yeh MH and
    11. Liu PF
    : High snail expression predicts a poor prognosis in breast invasive ductal carcinoma patients with HER2/EGFR-positive subtypes. Surg Oncol 27(2): 314-320, 2018. PMID: 29937187. DOI: 10.1016/j.suronc.2018.05.002
    OpenUrlCrossRefPubMed
    1. Qiao W,
    2. Jia Z,
    3. Liu H,
    4. Liu Q,
    5. Zhang T,
    6. Guo W,
    7. Li P,
    8. Deng M and
    9. Li S
    : Prognostic and clinicopathological value of Twist expression in breast cancer: A meta-analysis. PLoS One 12(10): e0186191, 2017. PMID: 29016671. DOI: 10.1371/journal.pone.0186191
    OpenUrlCrossRefPubMed
  27. ↵
    1. Jang MH,
    2. Kim HJ,
    3. Kim EJ,
    4. Chung YR and
    5. Park SY
    : Expression of epithelial-mesenchymal transition-related markers in triple-negative breast cancer: ZEB1 as a potential biomarker for poor clinical outcome. Hum Pathol 46(9): 1267-1274, 2015. PMID: 26170011. DOI: 10.1016/j.humpath.2015.05.010
    OpenUrlCrossRefPubMed
  28. ↵
    1. Sirkisoon SR,
    2. Carpenter RL,
    3. Rimkus T,
    4. Anderson A,
    5. Harrison A,
    6. Lange AM,
    7. Jin G,
    8. Watabe K and
    9. Lo HW
    : Interaction between STAT3 and GLI1/tGLI1 oncogenic transcription factors promotes the aggressiveness of triple-negative breast cancers and HER2-enriched breast cancer. Oncogene 37(19): 2502-2514, 2018. PMID: 29449694. DOI: 10.1038/s41388-018-0132-4
    OpenUrlCrossRefPubMed
  29. ↵
    1. Rhodes LV,
    2. Tate CR,
    3. Segar HC,
    4. Burks HE,
    5. Phamduy TB,
    6. Hoang V,
    7. Elliott S,
    8. Gilliam D,
    9. Pounder FN,
    10. Anbalagan M,
    11. Chrisey DB,
    12. Rowan BG,
    13. Burow ME and
    14. Collins-Burow BM
    : Suppression of triple-negative breast cancer metastasis by pan-DAC inhibitor panobinostat via inhibition of ZEB family of EMT master regulators. Breast Cancer Res Treat 145(3): 593-604, 2014. PMID: 24810497. DOI: 10.1007/s10549-014-2979-6
    OpenUrlCrossRefPubMed
    1. Ferrari-Amorotti G,
    2. Chiodoni C,
    3. Shen F,
    4. Cattelani S,
    5. Soliera AR,
    6. Manzotti G,
    7. Grisendi G,
    8. Dominici M,
    9. Rivasi F,
    10. Colombo MP,
    11. Fatatis A and
    12. Calabretta B
    : Suppression of invasion and metastasis of triple-negative breast cancer lines by pharmacological or genetic inhibition of slug activity. Neoplasia 16(12): 1047-1058, 2014. PMID: 25499218. DOI: 10.1016/j.neo.2014.10.006
    OpenUrlCrossRefPubMed
  30. ↵
    1. Liu SS,
    2. Qi J,
    3. Teng ZD,
    4. Tian FT,
    5. Lv XX,
    6. Li K,
    7. Song YJ,
    8. Xie WD,
    9. Hu ZW and
    10. Li X
    : Resistomycin attenuates triple-negative breast cancer progression by inhibiting E3 ligase Pellino-1 and inducing SNAIL/SLUG degradation. Signal Transduct Target Ther 5(1): 133, 2020. PMID: 32728028. DOI: 10.1038/s41392-020-00255-y
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 42 (8)
Anticancer Research
Vol. 42, Issue 8
August 2022
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
STAT3 Inactivation and Induction of Apoptosis Associate With Fluoxetine-inhibited Epithelial-mesenchymal Transition and Growth of Triple-negative Breast Cancer In Vivo
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 18 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
STAT3 Inactivation and Induction of Apoptosis Associate With Fluoxetine-inhibited Epithelial-mesenchymal Transition and Growth of Triple-negative Breast Cancer In Vivo
PEN-AN LIAO, PEI-YI CHU, ZHAO-LIN TAN, FEI-TING HSU, YANG-CHENG LEE, HSING-JU WU
Anticancer Research Aug 2022, 42 (8) 3807-3814; DOI: 10.21873/anticanres.15871

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
STAT3 Inactivation and Induction of Apoptosis Associate With Fluoxetine-inhibited Epithelial-mesenchymal Transition and Growth of Triple-negative Breast Cancer In Vivo
PEN-AN LIAO, PEI-YI CHU, ZHAO-LIN TAN, FEI-TING HSU, YANG-CHENG LEE, HSING-JU WU
Anticancer Research Aug 2022, 42 (8) 3807-3814; DOI: 10.21873/anticanres.15871
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Fluoxetine Suppresses Osteosarcoma Progression In Vivo by Inducing Apoptosis and Inhibiting the AKT/mTOR/NF-{kappa}B Signaling Pathway
  • Imipramine Suppresses Tumor Growth and Induces Apoptosis in Oral Squamous Cell Carcinoma: Targeting Multiple Processes and Signaling Pathways
  • Anticancer Effects of Antidepressants in Hepatocellular Carcinoma Cells
  • Clinicopathological and Prognostic Significance of Programmed Death Ligand-1 SP142 Expression in 132 Patients With Triple-negative Breast Cancer
  • Google Scholar

More in this TOC Section

  • Association of Interleukin-12A Genotypes With Nasopharyngeal Carcinoma Risk
  • Immunogenicity of Neoantigens in Colorectal Cancer: Potential Influence of Tumor Mutation Burden, Stages, and Metastasis
  • Tissue Prognostic Markers for Clear Cell Renal Cell Carcinoma Tumor-stroma Interaction: Impact on TNM Staging Parameters
Show more Experimental Studies

Similar Articles

Keywords

  • fluoxetine
  • triple-negative breast cancer
  • STAT3
  • apoptosis
  • epithelial-to-mesenchymal transition
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire