Next Article in Journal
Priming of Immune System in Tomato by Treatment with Low Concentration of L-Methionine
Previous Article in Journal
Noradrenaline and Adrenoreceptors Are Involved in the Regulation of Prostaglandin I2 Production in the Porcine Endometrium after Experimentally Induced Inflammation
Previous Article in Special Issue
Rapid Determination of Kinetic Constants for Slow-Binding Inhibitors and Inactivators of Human Histone Deacetylase 8
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Beyond Psychotropic: Potential Repurposing of Fluoxetine toward Cancer Therapy

by
Sultan F. Kadasah
1,
Abdulaziz M. S. Alqahtani
1,
Abdullah Alkhammash
2,* and
Mohamed O. Radwan
3,*
1
Department of Biology, Faculty of Science, University of Bisha, P.O. Box 551, Bisha 61922, Saudi Arabia
2
Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
3
Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(12), 6314; https://doi.org/10.3390/ijms25126314
Submission received: 16 May 2024 / Revised: 5 June 2024 / Accepted: 5 June 2024 / Published: 7 June 2024
(This article belongs to the Special Issue Techniques and Strategies in Drug Design and Discovery, 2nd Edition)

Abstract

:
Drug repurposing, rebranding an existing drug for a new therapeutic indication, is deemed a beneficial approach for a quick and cost-effective drug discovery process by skipping preclinical, Phase 1 trials and pharmacokinetic studies. Several psychotropic drugs, including selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs), were studied for their potential application in different diseases, especially in cancer therapy. Fluoxetine (FLX) is one of the most prescribed psychotropic agents from the SSRIs class for the treatment of several neuropsychiatric disorders with a favorable safety profile. FLX exhibited different oncolytic effects via mechanisms distinct from its main serotonergic activity. Taking advantage of its ability to rapidly penetrate the blood–brain barrier, FLX could be particularly useful in brain tumors. This was proved by different in vitro and in vivo experiments using FLX as a monotherapy or combination with temozolomide (TMZ) or radiotherapy. In this review of the literature, we summarize the potential pleiotropic oncolytic roles of FLX against different cancers, highlighting the multifaceted activities of FLX and its ability to interrupt cancer proliferation via several molecular mechanisms and even surmount multidrug resistance (MDR). We elaborated on the successful synergistic combinations such as FXR/temozolomide and FXR/raloxifene for the treatment of glioblastoma and breast cancer, respectively. We showcased beneficial pharmaceutical trials to load FLX onto carriers to enhance its safety and efficacy on cancer cells. This is the first review article extensively summarizing all previous FLX repurposing studies for the management of cancer.

Graphical Abstract

1. Introduction

Drug repurposing or repositioning represents an attractive approach for finding new applications for old drugs [1,2]. This kind of drug recycling introduces a batch of pros, including a shorter production time by eliminating Phase I clinical trials and a lower cost as a consequence in comparison to de novo drug discovery [1]. A pronounced merit of repurposing is that the pharmacokinetic and toxicity profiles of the repurposed entities were already identified. Previously, repurposing was dependent on serendipity; however, recently, it has been based on advanced omics technologies and computational tools [3]. One of the most outstanding examples is aspirin repurposing from a non-steroidal anti-inflammatory drug to an anti-platelet aggregation drug. Concomitantly, owing to the potential relationship between COX-2 and cancer, aspirin could be further reused for cancer therapy [4]. Drug repurposing modality played a role in trials to find drugs for COVID-19. Remdesivir and favipiravir are examples of existing drugs co-crystalized with SARS-CoV2 RNA-dependent RNA polymerase, whereas telaprevir and baicalein were co-crystalized SARS-CoV2 main protease (Mpro) [5,6,7,8].
Cancer is still a major cause of death globally, accounting for one-sixth of global mortality [9,10,11]. Available drugs encounter resistance and sometimes possess intolerable undesirable effects [12]. The scientific community is usually urged to pursue alternative cancer chemotherapeutic agents to address the issues and resistance of existing drugs. One obstacle is the lengthy period of time required to develop one new drug, in addition to the huge cost and high possibility of failing clinical trials or facing pharmacokinetic issues [13].
There are continuous trials demonstrating the repurposing of non-oncology drugs toward cancer therapy on both basic and clinical levels. Figure 1 demonstrates the remarkable increase in publications connecting cancer to repurposing, as found by a search in the Web of Science database. Sildenafil is a phosphodiesterase-5 inhibitor designed for the treatment of ischemic heart diseases and repurposed to treat erectile dysfunction. In addition, sildenafil is a sensitizer of cancer cells toward chemotherapy and radiation therapy [14,15]. Metformin, the widely used antidiabetic drug, exhibited anticancer and chemo-sensitization properties in preclinical and clinical studies [16,17].
The antimalarial drug quinacrine was found to be a dual target antiproliferative agent via the inhibition of Topo II and Hsp90 [18]. In addition, quinacrine was repurposed for managing cancer by several other mechanisms [19,20,21]. We have successful stories in the field of drug repurposing toward cancer management. Recently, we repositioned anti-HIV substituted benzimidazole derivatives for cell migration inhibition targeting heterogeneous nuclear ribonucleoprotein-M (hnRNP-M) [22]. Additionally, we repurposed S-trityl L-cysteine and S-trityl cysteamine derivatives from kinesin Eg5 inhibitors to Sirtuins 2 inhibitors [23,24]. A batch of repurposed non-oncology drugs for cancer management was extensively reviewed elsewhere [13,25].
Antidepressant drugs have a remarkable role in the therapy of cancer patients who are prone to depression disorders [26,27]. Early observations showed conflicting findings on the antidepressants’ effect on cancer promotion and growth [26,28]. Later studies revealed the great potential of antidepressant drugs, including tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors (SSRIs), for repurposing to cancer therapy via several mechanisms of action [2,8,27,29,30]. Different psychotropic drugs, including fluoxetine, fluspirilene, ebastine, and aripiprazole, demonstrated anticancer activity in glioblastoma, colorectal carcinoma, and breast cancer cell lines by disrupting lysosomal and mitochondrial function and triggering cell death but not inducing apoptosis [31]. In fact, among all classes of antidepressants, SSRIs have the highest repurposing potential for the management of cancer [2,32].
Owing to their favorable safety profile, SSRIs are the most prescribed antidepressant drugs, and they are used as adjuvant therapy for the treatment of other neuropsychiatric disorders [33,34]. Basically, a three-year study on a larger sample of patients using SSRIs ruled out any breast cancer risk due to their administration [35]. Concomitantly, several SSRIs proved oncopreventive and/or oncolytic properties against cancers of the lung [36], colorectal [37,38], and breast [39]. Accumulating pieces of evidence confirmed that SSRIs’ oncolytic activity is mainly through independent actions of their primary serotonergic-mediated mechanisms [30].
Indeed, all SSRIs showed various oncolytic activities except for vilazodone [2]. For example, sertraline (Zoloft®) induces apoptosis in colon cancer cells [40], suppresses tumor growth by blocking the 5′ adenosine monophosphate-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway, promotes autophagic flux in non-small cell lung cancer (NSCLC) cells [41], shows synergistic effects with sorafenib against hepatocellular carcinoma (HCC) cells proliferation [42], and reduces breast cell growth by interrupting serine/glycine synthesis [43,44].
Paroxetine (Paxil®) induces apoptosis in NSCLC via the ROS-MAPK pathway [45] in colon cancer cells by suppressing MET and HER3 kinases [46], and in MCF-7 by increasing extracellular calcium ion (Ca2+) and p38 [32]. Citalopram (CeleXA®) has a pro-apoptotic effect on acute myeloid leukemia (AML) via caspase-3 activation. Notably, it lowers invasion and metastasis of colon cancer cells by inhibiting the transforming growth factor-β (TGF-β) signaling pathway. The S-(+)-enantiomer of citalopram, escitalopram (Lexapro®), induces apoptosis and autophagy in glioblastoma [47] and NSCLC [48] and suppresses breast cancer cell growth [49].
Fluoxetine (FLX; Prozac®; see Figure 1) was the first approved SSRI and is still one of the most prescribed antidepressants worldwide. The literature is full of interesting studies on the potential rebranding of FLX for the management of different cancer types. Among different antidepressants, only FLX improved the overall survival of patients receiving FLX/PD-1/L1 immunotherapy compared to only PD-1/L1, according to a cohort study on cancer patients who receive checkpoint inhibitors [50]. For the first time, we introduce a review article to emphasize the potential role of FLX in the management of neoplasms, either solely or in combination with other chemotherapeutic agents. Additionally, the role of FLX in overcoming multidrug resistance (MDR) is discussed. Figure 2 summarizes the different cancer types that can be modulated by FLX. This review further includes pharmaceutical trials to load FLX on carriers to control its cellular release and enhance its efficacy.

2. Materials and Methods

We explored the literature in PubMed, Google Scholar, and Web of Science databases using three keywords, “fluoxetine”, “repurposing”, and “cancer”, to conduct a comprehensive search. Our search was carried out without year limitations because there was no previous review article on this topic. The outcome was around 92 research and review articles, of which 51 were extensively considered for the current review article. The residual articles were not investigated thoroughly because they mainly focused on repurposing other antidepressants or antipsychotics. We divided the results into sections based on the target cancer type. To recognize the potential cancer-related targets of FLX, the Swiss Target Prediction tool (http://www.swisstargetprediction.ch/) was accessed on 10 March 2024 [51].

3. Results

3.1. FLX in Brain Tumors

For brain tumors like glioblastoma multiforme (GBM), the blood–brain barrier (BBB) may hinder the development of some drugs that efficiently stop GBM cell proliferation and trigger apoptosis in vitro [30,52]. In other words, the efficacy of GBM drugs is often faced by poor drug delivery due to the presence of the BBB [53]. As an antidepressant drug, FLX has favorable physicochemical and pharmacokinetic properties to bypass BBB; hence, its repurposing for GBM therapy is substantially useful [54]. De facto, previous medical records showed remarkably enhanced life expectancy in GBM patients taking fluoxetine, but not other SSRIs [55].
FLX exhibited in vitro apoptotic effects on human GBM cells, viz. U87 and GMB8401, by increasing the intracellular Ca2+ concentration, damaging mitochondrial membranes, and releasing apoptogenic factors at 25–30 μM. This effect was reversed by the coadministrations of (2,3-dioxo-6-nitro-7-sulfamoyl-benzo[f]quinoxaline) (NBQX), a blocker of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR), which is highly expressed in GBM. In silico calculations supported by experimental tools emphasized that FLX binds to the GLUR1 subunit of AMPAR [56,57]. The apoptotic effect was consistent in vivo in a tumor xenograft using U87 GBM cells, using 10 mg/kg of FLX. Of note, the apoptotic effect was significant in brain tumors but not in normal brain cells of mice, reflecting a favorable safety profile of FLX [53].
Temozolomide (TMZ) is one of the most effective chemotherapeutics for GBM. Due to its common resistance, Wang’s group studied FLX activity in GBM and its synergistic effect with TMZ. Only FLX was able to inhibit the growth of different rat and human GBM cells, including C6, U87-MG, U373, and U251 with respective IC50 14.7, 21.8, 48.5, and 22.9 μM. FLX clearly enhanced C6 apoptosis by increasing the caspase-3 concentration and endoplasmic reticulum stress (ERS). Mechanistically, FXL elevated the expression of C/EBP homologous protein (CHOP) and autophosphorylation of its upstream and downstream signaling pathways, as identified by Western blotting. Notably, FLX sensitized C6 cells to TMZ treatment, showing a synergistic antiproliferative effect at lower inhibitory concentrations. The latter effect is also mediated by the CHOP pathway, as the knockdown of CHOP omitted FXR/TMZ synergism [58].
Considering that the TMZ resistance is mainly caused by DNA-repair O6-methylguanine-DNA methyltransferase (MGMT), Song et al. studied the role of FLX in suppressing MGMT expression. The authors concluded that 10 μM of FLX significantly reduced MGMT concentrations by disrupting NF-kB/p65 signaling and, hence, sensitizing GBM cells to TMZ. Those in vitro results were further validated in vivo in a murine subcutaneous xenograft model of human GBM cells U-138 MG [59].
An elegant research work by Bi et al. in 2021 extensively showed the effect of FLX on the signaling and metabolism of GBM. They identified acid sphingomyelinase (ASM; sphingomyelin phosphodiesterase 1 (SMPD1)) as an attractive GBM target that is necessary for cancer cell survival. In GBM cells, fluoxetine inhibited the SMPD1 enzymatic-activity formation of ceramide from sphingomyelin, resulting in a dose-dependent death in three different types of GBM cells. Consistently, this effect was reproduced in orthotopic xenografts implanted in nude mice brains. Interestingly, this effect is accompanied by lysosomal stress and suppression of the overexpressed oncogenic epidermal growth factor receptor VIII (EGFRvIII), a constitutively active form mutation of EGFR [55]. The authors confirmed in vitro and in vivo FLX/TMZ synergism in inducing DNA damage and cell death in multiple GBM models. This is in accordance with the recorded higher survival rates of GBM patients who co-administer FLX/TMZ [55].
Owing to its favorable safety profile, a research group studied the possibility of FLX repurposing for fatal neuroblastoma in children, which accounts for 7% of cancer diagnoses of children under 15 years old. In neuroblastoma, the Myc oncoprotein amplification is used as a negative prognostic marker and is a hallmark of a high-risk disease. Myc regulates the CKS1/SKP2/p27kip1 axis, which can be interrupted by FLX. The latter inhibited CKS1, increased p27Kip1 expression, and triggered neuroblastoma cell death at 10 μM concentration. Furthermore, small doses of FLX reduced the invasiveness and metastasis of neuroblastoma cells. This effect was further validated in a xenograft mice model [60].
FLX is not only effective as a monotherapy or combination therapy with TMZ, but it could also sensitize GBM cells (U-87 MG) to radiotherapy. Surprisingly, FLX showed a radioprotective effect on normal fibroblast cells HFFF2 in vitro [61]. Conclusively, FLX (10 μM) represents the best SSRI in terms of brain cancer management. Its effect is remarkable when combined with TMZ or radiotherapy.

3.2. FLX in Breast Cancer

Breast cancer is the most spreading cancer among women, with potential fatal metastasis to the bones, liver, lungs, and brain [62,63]. The literature is loaded with considerable results showing the sensitivity of different types of breast cancer to FLX. The group of Chan took advantage of FLX to circumvent MDR in the MCF-7/ADM (doxorubicin-resistant human breast carcinoma) cells; they used stealth liposome co-encapsulation of doxorubicin and FLX for prolonged circulation half-life and improved safety in vivo. The used formulation demonstrated promising anticancer properties, under both in vitro and in vivo conditions, capable of the effective reversal of doxorubicin resistance [64].
The mechanism of FLX for reversing MDR was not fully understood. Thus, another group studied the synergism of FLX with Adriamycin (ADM) and paclitaxel (PXL). FLX-ADM combination—(5 μg/mL) FLX with various concentrations of ADM—enhanced apoptosis significantly in MCF-7/ADM-resistant cells but not MCF-7 cells. The IC50 values of sole ADM and ADM with FLX were 13.62 µM and 2.71 µM, respectively, implying a significant positive effect of FLX addition. The authors found that the chemo-sensitizing effect of FLX occurs via simultaneous upregulation of the tumor suppressor protein p53 and downregulation of B-cell lymphoma 2 (Bcl-2) [65].
Raloxifene (RAL) is widely used for the treatment of estrogen receptor-positive breast cancer, with fewer side effects than the parent tamoxifen [66,67]. Kabel et al. investigated the potential synergistic effect in the case of using a FLX/RAL combination in experimentally 7,12-Dimethyl Benzanthracene (DMBA)-induced breast cancer in female Wistar rats. They found that either FLX or RAL is an effective pro-apoptotic agent, but the RAL/FLX combination had a better outcome than either of them, as shown by the tumor volume size. RAL (3 mg/kg)/FLX (30 mg/kg) administration improved tumor antioxidant status via the elevation of catalase (CAT) and superoxide dismutase (SOD) and reduction of malondialdehyde (MDA) in tumor tissue compared to the control group. The titled combination reduced tissue concentration of the pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) and suppressed TGF-β [68].
A complementary study to the abovementioned one regarding the FLX/RAL combination showed that it can suppress invasion, metastasis, and angiogenesis of DMBA-induced breast cancer. The combination lowered mammary tissue vascular endothelial growth factor (VEGF), macrophage colony-stimulating factor (M-CSF), and matrix metalloproteinase-9 (MMP-9) levels, as determined by an enzyme-linked immunosorbent assay (ELISA). In accordance with the previous study, the FLX/RAL oncolytic effect significantly surpasses each sole drug [69].
According to Duarte et al., the IC50 of FLX against MCF-7 cells is 7.78 µM, as detected by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assay. When combined with PXL, a significant synergistic antiproliferative effect was observed using both MTT and SRB assays [70]. The same research group found significant synergism for the combination of FXL and doxorubicin against MCF-7 in another study [71]. They also found that cotreatment of FXL and honeybee venom demonstrated a better cytotoxic effect than either of them [72].
In a study testing different antidepressants, including amitriptyline, bupropion, FLX, paroxetine, and tianeptine, as monotherapy for the induction of apoptotic cell death in breast cancer cells MCF-7, only paroxetine, followed by FLX (10 µM), demonstrated significant activity in a dose-dependent manner [32]. Another study benchmarked standalone FLX on triple-negative breast cancer (TNBC) in vivo. Surprisingly, FLX suppressed tumor growth by downregulating STAT3 signaling transduction and triggering a caspase-mediated apoptotic pathway [73].
By virtue of the aggressiveness of TNBC, Bowie et al. extensively studied the effect of FLX on TNBC cells SUM149PT. They found that treatment with 10 µM FLX induced apoptosis and enhanced ER stress and autophagy (IC50 µM 7.9). This is accompanied by cell-cycle arrest at the G1 phase and caspase-7-mediated cell death. Those effects are less prominent in the non-transformed MCF-10A cells, reflecting a favorable safety profile [74].
Another research group studied the FLX effect on other TNBC cells, MDA-MB-231 and MDA-MB-436. Sun et al. revealed that 1 μmol/L FLX induced both apoptosis and autophagic cell death in the titled cell lines. The apoptotic effect is attributed to the upregulation of the expression levels of caspase-3 and caspase-8 and poly (ADP-ribose) polymerase (PARP). In addition, FLX significantly reduced the phosphorylation of eukaryotic elongation factor-2 kinase (eEF2K), which is overexpressed in different cancers and plays an indispensable role in the crosstalk between apoptosis and autophagy in TNBC. Furthermore, FLX modulates autophagic proteins; it decreases phosphorylation of mTOR, activates AMPK, and increases ULK1 phosphorylation [75].

3.3. FLX in Hepatocellular Carcinoma (HCC)

Hepatocellular carcinoma (HCC) is the most common form of primary liver malignancy, with 906,000 new cases and 830,000 deaths worldwide yearly, making it the third leading cause of cancer death [76,77]. Several SSRIs and SNRIs, not including FLX, were tested for their possible cytotoxic effect on HCC [78]. FLX reduced the viability of the human HCC cell line Hep3B and induced apoptosis, as detected by MTT assay and staining, where at 10, 30, and 100 μM of FLX treatment for 24 h, the viability values were 98.3 ± 15.2, 80.4 ± 4.3, and 30.4 ± 3.4%, respectively. Disruption of mitochondrial membrane potential (MMP) is an early indicator of reactive nitrogen species-induced apoptosis. The authors found that the treatment of Hep3B with FLX resulted in a loss of MMP and enhanced the formation of reactive oxygen species (ROS). This was accompanied by the suppression of anti-apoptotic phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) protein and an increase in pro-apoptotic c-JUN N-terminal kinase (c-JNK) and p38 mitogen-activated protein kinases (MAPKs) [79].
An in vivo study showed that FLX significantly suppressed tumor growth of Hep3B cells in a xenograft mice model without inducing liver pathology or general toxicity at a 10 mg/kg FLX dose. The oncolytic effect is attributed to the upregulation of the extrinsic and intrinsic apoptosis-associated proteins, including caspase-3, caspase-8, and caspase-9, and suppression of metastasis-associated protein VEGF, MMP-9, urokinase-type plasminogen activator (uPA), and Cyclin- D1, as validated by immunohistochemistry (IHC) staining. This indicated that FLX could reduce Hep3B angiogenesis and cell invasion. Additionally, FLX attenuated phosphorylation of nuclear factor-kappa B (NF-κB) p65 on ser276, AKT, and ERK in the titled cells as validated by IHC [80]. A similar action was induced by 30 µM FLX treatment of HCC SK-Hep1 cells for 48 h in another study for the same research group [81].
Hend et al. prepared FLX-loaded hexosome (HEX) using the hot emulsification method to prolong the release and enhance the activity of FXR on HCC cells HepG2. Successfully, the optimized HEX was able to prolong FLX release, where only 19.5% was released in PBS pH 7.4 after 24 h, which was highly increased in acidic medium pH 5.5, which imitates the cancer microenvironment. Accordingly, HEX improved/enhanced the cellular delivery and cytotoxic activity of FLX against HepG2 compared to the drug solution [82].

3.4. FLX in Colon Cancer

Colorectal cancer is ranked as the third most common cancer in the United States [83,84]. MTT assay showed a significant antiproliferative effect of FLX on colon cancer cells HT-29 with an IC50 value of 6.12 µM. However, the cotreatment of FLX and 5-fluorouracil (5-FU) (IC50 3.79 µM) did not show a synergistic effect at different concentrations [70]. Nevertheless, the combination of fluoxetine and honeybee venom demonstrated a significant synergistic effect against HT-29 cells [72]. Another study showed that (20 µM) FLX remarkably augmented the antiproliferative effect of either cisplatin or carboplatin but not oxaliplatin on colon cancer cells HCT116. The authors attributed this synergistic effect to calmodulin inhibition by FLX [85].
Another effective combination of metformin/Efavirenz/FLX (5 mM/1.5 µM/0.9 µM) against HCT116 human colon cancer cells was recently reported by Kang et al. [86]. The three-drug combination triggered a massive increase in ROS levels and exhibited a dramatic antiproliferative effect on HCT116 but not human dermal fibroblasts (HDF) cells. The treated HCT116 cells showed increased DNA damage, apoptosis, autophagy, and necroptosis-related factors, as detected by Western blotting. In the HCT116 xenograft mice model, the titled combination showed consistent results by reducing tumor weight and size compared to the control group [86].
Another validating study showed that FLX-induced concentration-dependent apoptosis and DNA fragmentation in HCT116+/+ and p53 gene-depleted HCT116/ human colorectal cancer cells with respective IC50 values of 3.19 ± 0.23 μM and 4.73 ± 0.5 μM and less effect on normal cells. Mechanistically, FLX treatment resulted in cell-cycle arrest at Sub-G1 and G0/G1 phases in both cell lines, as detected by a FACS analysis [87]. In summary, the molecular mechanism of FLX cytotoxicity against HCT116 cells is independent of p53 modulation.

3.5. FLX in Cervical Cancer

Cervical cancer is one of the most spreading cancers in females worldwide [10]. The resistance of cervical cancer cells to cisplatin is a real challenge that leads to a decline in the survival rate [88]. In cervical-cancer HeLa cells, 50 μM of FLX reduced viability via energy depletion and increased cytosolic Ca2+ concentration by emptying the endoplasmic reticulum (ER) through the translocon, an ER Ca2+ leakage structure [89].
In a dose-dependent manner, FLX promoted HeLa cellular chemo-sensitivity to cisplatin in vitro and in xenograft mice models compared to FLX or cisplatin alone. The titled combination, 16 µM FLX/16 µM cisplatin, triggered G0/G1 phase arrest (73%) compared with FLX (58%) or cisplatin (60%), as observed by FACS analysis. Additionally, the percentage of apoptotic cells was 48.3% compared with FLX (38.1%) or cisplatin (31.5%). In vivo assay showed that oral dose of FXR (1 mg/kg), cisplatin (3 mg/kg), and FXR/cisplatin combination effectively reduced the tumor weight with inhibition rates of 10.9%, 46.6%, and 53.7%, respectively. Mechanistically, FXR/cisplatin upregulated apoptotic proteins caspase-9 and p17 and suppressed MDR proteins glutathione S-transferase π (GSTπ) and P-gp [90].
Naz et al. developed FLX–dextran nanoparticles conjugates for better efficacy of FLX. Dextran was oxidized by sodium iodate to form the corresponding aldehyde, which was readily reacted with FLX, forming a Schiff base [91,92]. FLX–dextran nanoparticles were stable at physiological blood circulation and normal tissues with remarkably higher release in an acidic environment, i.e., pH 5, which resulted in high specificity toward cancer cells and reduced systemic undesirable effects. Indeed, only FLX showed higher toxicity to normal mouse embryo fibroblast cells 3T3 than FLX–dextran nanoparticles. However, the latter showed a similar anticancer effect on HeLa cells by completely inhibiting cellular growth at 30 µM concentration [91].

3.6. FLX in NSCLC

NSCLC represents 85% of lung cancer cases, and it is characterized by an invasive and metastatic nature; the acquired resistance to receptor tyrosine kinase inhibitors is a major therapeutic obstacle [93]. A research group has extensively studied the effect of FLX on NSCLC cells CL1-5-F4. FLX reduced their viability via induction of apoptosis in a dose-dependent manner with IC50 40 µM, as shown by MTT assay. A Western blot analysis revealed that FLX significantly reduced the level of DNA repair-associated proteins, including levels of mediator of DNA damage checkpoint 1 (MDC1), O6-methylguanine DNA methyltransferase MGMT, and 14-3-3. Reporter gene assay showed that FLX remarkably inhibited NF-ĸB activation in CL1-5-F4 cells at the IC50 concentration of FLX. The related level of metastasis-associated proteins MMP-2, MMP-9, uPA, and VEGF was suppressed. Accordingly, CL1-5-F4 metastasis and invasion were attenuated [94].
In light of the above data, Hsua et al. benchmarked FLX in vivo using a CL1-5-F4-bearing mice model. FLX significantly suppressed tumor growth and size in the established model without general toxicity at 10 mg/kg FLX dose. The anticancer effect is ascribed to promoting the level of extrinsic and intrinsic apoptosis-associated proteins, including caspase-3, caspase-8, and caspase-9, and reduction of metastasis-associated protein VEGF, MMP-9, urokinase-type plasminogen activator (uPA), and Cyclin-D1. This validated the previous conclusion of FLX’s ability to attenuate NSCLC angiogenesis and invasion [94]. Additionally, FLX attenuated phosphorylation of NF-κB p65 on ser276, AKT, and ERK [80].
Another in vivo study by Yang et al. revealed that FLX (20 mg/kg) could suppress tumor growth as represented by weight and size in a xenograft mice model bearing subcutaneous transplanted lung cancer cells A549. While exposure to chronic unpredictable mild stress reduced serotonin and tryptophane concentrations and suppressed immunity, FLX treatment significantly reversed those effects. FLX promoted cellular immunity and elevated tryptophan/kynurenine ration. A biochemical assessment of kynurenine cascade-related genes tryptophan-2,3-dioxygenase (TDO), indoleamine-2,3-dioxygenase 1 (IDO1), and IDO2 showed that FLX (15 μmol/L) suppressed their upregulation due to chronic stress exposure [95].

3.7. FLX in Pancreatic Cancer

Pancreatic cancer is a relatively uncommon cancer that is characterized by a complex microenvironment. Its incidence is increasing, and it is expected to become the second leading cause of cancer-related death by 2030 [96]. As a hallmark of FLX safety to normal pancreatic cells, its administration did not induce acute pancreatitis compared to citalopram and other SSRIs [97]. A rough in vivo study on a subcutaneous xenograft model of human pancreatic cancer cell line SW1990 in mice revealed that FLX 10 mg/kg did not induce a significant reduction in tumor growth [98].
However, another impressive study by Schneider et al. showed a promising role of FLX in the management of pancreatic cancer via a serotonergic pathway. De facto, platelet-derived peripheral serotonin enhances the growth of murine pancreatic cancer cells Panc02. This is mediated by the enhanced expression of PD-L1 on mouse and human cancer cells in vitro via serotonylation reaction by covalently binding to glutamine amino acid, resulting in the activation of small G proteins. In turn, this impairs the accumulation of immune defense cells CD8+T and upregulates PD-L1 expression in the pancreatic cancer tumor microenvironment. Inhibition of serotonin cargo in platelets by FLX 20 mg/kg or TPH1 inhibitor telotristat reduced colon and pancreatic tumor growth in an established C57BL/6 mice model by increasing CD8+T accumulation within the tumor [99]. The authors concluded that combining FLX with anti-PD-1 therapy could be a potential way to treat solid tumors [99].

3.8. FLX in Lymphoma

Lymphomas are a heterogeneous group of malignancies that are attributed to the inadequate proliferation of lymphocytes at different maturation stages and account for around 5% of malignancies [100,101]. Human Burkitt lymphoma (BL) is an aggressive form of lymphoma due to the rapid proliferation rate, and it needs immediate intervention with strongly efficient chemotherapeutics [102]. FLX 50 µM exhibited quick apoptotic-mediated cytotoxicity against the chemo-sensitive BL cells MUTU-I, accompanied by an insignificant effect on normal blood cells. Independent of the serotonergic effect of FLX, it induced apoptosis via the caspase pathway, DNA cleavage, and PARP cleavage [103].
Later on, the same group explored the detailed mechanism using chemoresistant BL cell line DG-75 alongside MUTU-I. The former lacks Bax and Bak, making it difficult for any chemotherapy to induce apoptosis. FLX at 50 µM induced autophagy programmed cell death in DG-75 independently from the caspase pathway, DNA cleavage, and PARP cleavage. In DG-75 cells, Western blot showed upregulation of the autophagy marker Beclin 1, and the cytotoxic effect of FLX was reversed by cotreatment with the autophagy inhibitor 3-Methyladenine, contrary to MUTU-I cells. Extracellular Ca2+ influx was found to be crucial in FLX effect on DG-75 but not MUTU-I cells. Furthermore, the authors ruled out ROS involvement in BL cell death. Conclusively, in the chemo-sensitive cells MUTU-I, FLX elicits classic type I cell death apoptosis; nevertheless, it triggers type II cell death autophagy in the chemo-resistant cells DG-75 [104].
In EL4 lymphoma C57BL/6J mice model, FLX 15 mg/kg attenuated tumor growth accelerated by mice exposure to chronic stress conditions. Cell cycle regulators cyclins A2, D1, and D3 were elevated, whereas FLX treatment restored their mRNA expression levels to control values. Furthermore, FLX reduced invasiveness and metastasis to the liver and kidney. Once again, FLX reversed the effect of chronic stress by reducing MMP-2 and MMP-9 and increasing tissue inhibitors of metalloproteases TIMPs levels. Intriguingly, treatment with FLX promoted the antitumor immune response in animals [105]. Finally, It is worth noting that one recent report showed that FLX induced autophagy and apoptosis in lymphocytic leukemia targeting Akt phosphorylation dose-dependently [106]. The key findings in this review were summarized in Table 1.

3.9. FLX in Multidrug Resistance (MDR)

MDR operated by extrusion pumps such as P-glycoprotein is one of the main reasons for chemotherapy failure [107,108,109]. One of the earliest studies found that FLX works as a highly effective chemo-sensitizer by elevating the cytotoxicity of the anticancer drugs doxorubicin, mitomycin C, vinblastine, and paclitaxel in drug-resistant cancer cells. FLX modulated MDR in vitro and in vivo by slowing down the efflux rate of the titled chemotherapeutics [110].
Recent reports showcased that MDR is caused by the overexpression of transporters on the cancer cell membrane that expel chemotherapy out of the tumor cells. These pumps are members of the ATP-Binding Cassette (ABC) transporters superfamily [111]. There are seven subfamilies belonging to ABC viz., ABCA—ABCG, where ABCC includes 13 proteins, 9 of which are indicated as MDR proteins (MRPs). Recently, ABCC1/MRP1 and ABCC10/MRP7 were identified as the main drug transporters in MDR, and their inhibition is a potential tool for reversing MDR [112,113].
An interesting new study by Kanner et al. assessed three SSRIs, including FLX, for the binding and inhibition of MRP1 and MRP7 in silico. They found that FLX is well adopted in the binding sites of MRP1 and MRP7, with a slightly higher affinity toward MRP7. Experimentally, they found that FLX reversed the MDR resistance of MRP1 overexpressing human epidermoid carcinoma cell line KB/CV60 to vincristine and doxorubicin. In parallel, the same sensitizing effect was observed for MRP7-overexpressing human ovarian adenocarcinoma cell line SKOV3/MRP7, which is resistant to paclitaxel [114]. Conclusively, FLX could be an efficient chemo-sensitizer for overcoming MDR.

3.10. Potential Anticancer Mechanisms of FLX

Although FLX showed a vast array of anticancer activities, so far, its serotonin-independent molecular mechanism is yet to be elucidated. This provoked us to retrieve potential targets of FLX using the Swiss Target Prediction tool [51]. Other than CNS targets, FLX showed a high possibility of targeting four different kinases, namely serine/threonine-protein kinase PIM1, tyrosine-protein kinase receptor FLT3, glycogen synthase kinase-3 beta (GSK-3β), and serine/threonine-protein kinase Aurora-A. The titled kinases are valid targets in cancer therapy [115,116,117,118].
Indeed, FLX exhibited high 2D/3D similarities to inhibitors of those kinases, as predicted by SwissSimilarity [119]. Additionally, FLX showed structure similarity to vascular endothelial growth factor receptor 1 (VEGFR1) and vascular endothelial growth factor receptor 2 (VEGFR2) inhibitors, which are implicated in cancer angiogenesis and metastasis [120]. Other than kinases, FLX is predicted to inhibit the inhibitor of apoptosis protein 3 (IOA3) as well. Herein, we showcase an example of a reported molecule, CHEMBL437889, with a 0.86 3D similarity to FLX. According to SciFinder databases, CHEMBL437889 was reported to inhibit VEGFR1 and VEGFR2 with respective IC50 0.4 µM and 0.3 µM [121,122]. In addition, a notable compound, CHEMBL3642580 [123], is a multitarget anticancer kinase inhibitor with a 3D similarity value of 0.83 to FLX (Figure 3). Conclusively, FLX has a high considerable similarity to oncolytic compounds, and its pharmacophores can be clues for medicinal chemists to be repurposed for cancer therapy.

4. Discussion and Future Perspectives and Challenges

This review of the literature showed that FLX has versatile oncolytic and chemo-sensitization effects with different mechanisms of action. In brain tumors, FLX showed activity against GBM and neuroblastoma. The effect of FLX against GBM was shown either as a monotherapy or as a sensitizer for another chemotherapy or radiotherapy. Seemingly, FLX sensitized GBM cells to TMZ via the CHOP and MGMT pathways. The coadministration of TMZ/FLX enhanced life expectancy in GBM patients. FLX showed pronounced chemo-sensitization in luminal breast cancer and TNBC. In doxorubicin-resistant MCF-7 cells, FLX simultaneously upregulated bcl-2 and p53. FLX enhances the cytotoxic effect of RAL by lowering VEGF, M-CSF, and MMP-9. In the same context, FLX proved to have a synergistic effect when combined with PXL and honeybee venom. Notably, the FLX effect extends to different TNBC phenotypes, such as SUM149PT, MDA-MB-231, and MDA-MB-436.
FLX showed in vitro and in vivo apoptotic effects on HEP3B HCC cells via increasing ROS generation and the suppression of related tyrosine kinases and metalloproteinases. HEX-loaded FLX was successfully used against HepG2 HCC cells. As per colorectal cancer, standalone FLX had an IC50 value of 6.12 µM against HT-29 cells with a significant synergetic effect with honeybee venom. Furthermore, FLX solely induced apoptosis in HCT116 cells via the p53-independent mechanism, whereas its combination with cisplatin or metformin/Efavirenz was more effective.
FLX reduced the viability of cervical cancer HeLa cells by increasing the cytosolic Ca2+ concentration and enhanced their sensitivity to cisplatin in vitro and in vivo by upregulating apoptotic proteins and downregulating MDR proteins. Using FLX–dextran nanoparticles conjugates improved its selectivity toward cancer cells over normal cells. In addition, FLX induced apoptosis and reduced invasiveness and angiogenesis in NSCLC cells CL1-5-F4 in a dose-dependent manner both in vitro and in vivo by upregulating different caspases and suppressing VEGF, MMP-9, urokinase-uPA, and Cyclin-D1. NSCLC A549 cells were sensitive to FLX treatment in vivo by promoting cellular immunity and suppressing TDO, IDO1, and IDO2 genes.
Surprisingly, a serotonin-dependent pathway was involved in the anticancer effect of FLX on murine pancreatic cancer cells Panc02. A serotonylation reaction triggers PD-L1 overexpression and increases CD8+T-cell accumulation in the cancer microenvironment, implying that FLX might be useful in combination with anti-PD-1 therapy. In another instance, FXR reduced the viability of both chemo-sensitive and chemo-resistant BL cells MUTU-I and DG-75, respectively. In the latter cells, FXR induced autophagy and extracellular Ca2+ influx without the induction of ROS or apoptosis.
Being an existing drug with an established pharmacokinetic and safety profile, FLX is a possible candidate for drug repurposing. However, most of its oncolytic properties are non-serotonin dependent and require high doses. The majority of in vivo studies used 10 mg/kg/day, which highly exceeds its recommended therapeutic dose, which is 20 mg and up to 80 mg per day. High doses of FLX can lead to severe and sometimes fatal serotonin syndrome [124]. Therefore, there is a long road ahead regarding FLX repurposing for oncolytic purposes. This comprises the elucidation of its molecular mechanism(s) and improving its potency to be more effective at lower doses.

5. Conclusions

Drug repurposing represents an indispensable tool for modern drug discovery that paves the way for a short-term and cost-effective bench-to-bedside drug transition. Several SSRIs have been benchmarked against a multitude of cancer types. FLX is distinguished by its highly favorable safety profile; compared to other SSRIs, it is unlikely to cause pancreatitis. Among different antidepressants, only FLX enhanced the overall survival of patients receiving antidepressant/PD-1/L1 immunotherapy combination compared to PD-1/L1 alone. Of note, FLX is characterized by its high repurposing potential, especially against GBM and neuroblastoma cells, which it can access readily due to its rapid BBB penetration ability. FLX exhibited cytotoxic effects against cancers from different origins, including breast, liver, colon, cervix, lung, pancreas, and lymph system cancers. We showed that FLX/TMZ combination action significantly surpasses monotherapy against GBM on both in vitro and in vivo levels. Similarly, the FLX/RAL combination outperforms monotherapy for the treatment of DMBA-induced breast cancer. Additionally, a combination of cisplatin with fluoxetine could be a judicious choice for the treatment of cervical cancer with a better outcome than cisplatin alone.
In this review, we further explained that FLX is a promising chemo-sensitizer that can circumvent the MDR of chemotherapy. We also discussed pharmaceutical preparation that loaded fluoxetine on carriers to improve its delivery and selectivity toward cancer cells that have a characteristic acidic environment. During the writing of this article, a new study was released showcasing the ability of FLX to inhibit the cell proliferation, invasion, metastasis, and angiogenesis of osteosarcoma cells by suppressing the phosphorylation of signal transducer and activator of transcription 3 (STAT3) [125]. Taken together, FLX repurposing could be a potential avenue for the cotreatment of cancer patients.

Author Contributions

Conceptualization, M.O.R.; methodology, S.F.K., A.M.S.A. and M.O.R.; software, S.F.K., A.M.S.A. and M.O.R.; investigation, S.F.K., A.M.S.A. and A.A.; resources, S.F.K., A.M.S.A. and A.A.; data curation, S.F.K. and A.M.S.A.; writing—original draft preparation, S.F.K. and M.O.R.; writing—review and editing, S.F.K., A.M.S.A., A.A. and M.O.R.; visualization, S.F.K. and A.M.S.A.; supervision, M.O.R.; project administration, M.O.R.; funding acquisition, S.F.K., A.M.S.A. and A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are thankful to the Deanship of Graduate Studies and Scientific Research at University of Bisha for supporting this work through the Fast-Track Research Support Program. The authors would like to thank the Deanship of Scientific Research at Shaqra University for supporting this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Oliveira, E.A.M.D.; Lang, K.L. Drug Repositioning: Concept, Classification, Methodology, and Importance in Rare/Orphans and Neglected Diseases. J. Appl. Pharm. Sci. 2018, 8, 157–165. [Google Scholar] [CrossRef]
  2. Song, Y.; Yang, X.; Yu, B. Repurposing Antidepressants for Anticancer Drug Discovery. Drug Discov. Today 2022, 27, 1924–1935. [Google Scholar] [CrossRef] [PubMed]
  3. Rascol, O.; Fabbri, M.; Poewe, W. Amantadine in the Treatment of Parkinson’s Disease and Other Movement Disorders. Lancet Neurol. 2021, 20, 1048–1056. [Google Scholar] [CrossRef]
  4. Menter, D.G.; Schilsky, R.L.; DuBois, R.N. Cyclooxygenase-2 and Cancer Treatment: Understanding the Risk Should Be Worth the Reward. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 1384–1390. [Google Scholar] [CrossRef] [PubMed]
  5. Sezer, A.; Halilović-Alihodžić, M.; Vanwieren, A.R.; Smajkan, A.; Karić, A.; Djedović, H.; Šutković, J. A Review on Drug Repurposing in COVID-19: From Antiviral Drugs to Herbal Alternatives. J. Genet. Eng. Biotechnol. 2022, 20, 78. [Google Scholar] [CrossRef] [PubMed]
  6. Rodrigues, L.; Bento Cunha, R.; Vassilevskaia, T.; Viveiros, M.; Cunha, C. Drug Repurposing for COVID-19: A Review and a Novel Strategy to Identify New Targets and Potential Drug Candidates. Molecules 2022, 27, 2723. [Google Scholar] [CrossRef] [PubMed]
  7. Hijikata, A.; Shionyu, C.; Nakae, S.; Shionyu, M.; Ota, M.; Kanaya, S.; Shirai, T. Current Status of Structure-Based Drug Repurposing against COVID-19 by Targeting SARS-CoV-2 Proteins. Biophys. Physicobiol. 2021, 18, 226–240. [Google Scholar] [CrossRef] [PubMed]
  8. Lenze, E.J.; Reiersen, A.M.; Zorumski, C.F.; Santosh, P.J. Beyond “Psychotropic”: Repurposing Psychiatric Drugs for COVID-19, Alzheimer’s Disease, and Cancer. J. Clin. Psychiatry 2023, 84, 22r14494. [Google Scholar] [CrossRef] [PubMed]
  9. Radwan, M.O.; Abd-Alla, H.I.; Alsaggaf, A.T.; El-Mezayen, H.; Abourehab, M.A.S.; El-Beeh, M.E.; Tateishi, H.; Otsuka, M.; Fujita, M. Gypsogenin Battling for a Front Position in the Pentacyclic Triterpenes Game of Thrones on Anti-Cancer Therapy: A Critical Review—Dedicated to the Memory of Professor Hanaa M. Rady. Molecules 2023, 28, 5677. [Google Scholar] [CrossRef]
  10. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  11. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
  12. Hamdy, A.K.; Sakamoto, T.; Toma, T.; Sakamoto, M.; Abourehab, M.A.S.; Otsuka, M.; Fujita, M.; Tateishi, H.; Radwan, M.O. New Insights into the Structural Requirements of Isatin-Derived Pro-Apoptotic Agents against Acute Myeloid Leukemia. Pharmaceuticals 2022, 15, 1579. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, Z.; Zhou, L.; Xie, N.; Nice, E.C.; Zhang, T.; Cui, Y.; Huang, C. Overcoming Cancer Therapeutic Bottleneck by Drug Repurposing. Signal Transduct. Target. Ther. 2020, 5, 113. [Google Scholar] [CrossRef] [PubMed]
  14. Cruz-Burgos, M.; Losada-Garcia, A.; Cruz-Hernández, C.D.; Cortés-Ramírez, S.A.; Camacho-Arroyo, I.; Gonzalez-Covarrubias, V.; Morales-Pacheco, M.; Trujillo-Bornios, S.I.; Rodríguez-Dorantes, M. New Approaches in Oncology for Repositioning Drugs: The Case of PDE5 Inhibitor Sildenafil. Front. Oncol. 2021, 11, 627229. [Google Scholar] [CrossRef]
  15. Di, X.; Gennings, C.; Bear, H.D.; Graham, L.J.; Sheth, C.M.; White, K.L.; Gewirtz, D.A. Influence of the Phosphodiesterase-5 Inhibitor, Sildenafil, on Sensitivity to Chemotherapy in Breast Tumor Cells. Breast Cancer Res. Treat. 2010, 124, 349–360. [Google Scholar] [CrossRef]
  16. He, X.-K.; Su, T.-T.; Si, J.-M.; Sun, L.-M. Metformin Is Associated with Slightly Reduced Risk of Colorectal Cancer and Moderate Survival Benefits in Diabetes Mellitus: A Meta-Analysis. Medicine 2016, 95, e2749. [Google Scholar] [CrossRef]
  17. Urpilainen, E.; Puistola, U.; Boussios, S.; Karihtala, P. Metformin and Ovarian Cancer: The Evidence. Ann. Transl. Med. 2020, 8, 1711. [Google Scholar] [CrossRef]
  18. Pan, X.; Mao, T.-Y.; Mai, Y.-W.; Liang, C.-C.; Huang, W.-H.; Rao, Y.; Huang, Z.-S.; Huang, S.-L. Discovery of Quinacrine as a Potent Topo II and Hsp90 Dual-Target Inhibitor, Repurposing for Cancer Therapy. Molecules 2022, 27, 5561. [Google Scholar] [CrossRef] [PubMed]
  19. Oien, D.B.; Pathoulas, C.L.; Ray, U.; Thirusangu, P.; Kalogera, E.; Shridhar, V. Repurposing Quinacrine for Treatment-Refractory Cancer. Semin. Cancer Biol. 2021, 68, 21–30. [Google Scholar] [CrossRef]
  20. Gurova, K.V.; Hill, J.E.; Guo, C.; Prokvolit, A.; Burdelya, L.G.; Samoylova, E.; Khodyakova, A.V.; Ganapathi, R.; Ganapathi, M.; Tararova, N.D.; et al. Small Molecules That Reactivate P53 in Renal Cell Carcinoma Reveal a NF-kappaB-Dependent Mechanism of P53 Suppression in Tumors. Proc. Natl. Acad. Sci. USA 2005, 102, 17448–17453. [Google Scholar] [CrossRef]
  21. Park, S.; Oh, A.-Y.; Cho, J.-H.; Yoon, M.-H.; Woo, T.-G.; Kang, S.-M.; Lee, H.-Y.; Jung, Y.-J.; Park, B.-J. Therapeutic Effect of Quinacrine, an Antiprotozoan Drug, by Selective Suppression of p-CHK1/2 in P53-Negative Malignant Cancers. Mol. Cancer Res. MCR 2018, 16, 935–946. [Google Scholar] [CrossRef] [PubMed]
  22. Radwan, M.O.; Toma, T.; Arakaki, Y.; Kamo, M.; Inoue, N.; Koga, R.; Otsuka, M.; Tateishi, H.; Fujita, M. New Insight into the Bioactivity of Substituted Benzimidazole Derivatives: Repurposing from Anti-HIV Activity to Cell Migration Inhibition Targeting hnRNP M. Bioorg. Med. Chem. 2023, 86, 117294. [Google Scholar] [CrossRef] [PubMed]
  23. Radwan, M.O.; Ciftci, H.I.; Ali, T.F.S.; Ellakwa, D.E.; Koga, R.; Tateishi, H.; Nakata, A.; Ito, A.; Yoshida, M.; Okamoto, Y.; et al. Antiproliferative S-Trityl-l-Cysteine -Derived Compounds as SIRT2 Inhibitors: Repurposing and Solubility Enhancement. Molecules 2019, 24, 3295. [Google Scholar] [CrossRef] [PubMed]
  24. Radwan, M.O.; Ciftci, H.I.; Ali, T.F.S.; Koga, R.; Tateishi, H.; Nakata, A.; Ito, A.; Yoshida, M.; Fujita, M.; Otsuka, M. Structure Activity Study of S-Trityl-Cysteamine Dimethylaminopyridine Derivatives as SIRT2 Inhibitors: Improvement of SIRT2 Binding and Inhibition. Bioorg. Med. Chem. Lett. 2020, 30, 127458. [Google Scholar] [CrossRef] [PubMed]
  25. Singhal, S.; Maheshwari, P.; Krishnamurthy, P.T.; Patil, V.M. Drug Repurposing Strategies for Non-Cancer to Cancer Therapeutics. Anticancer Agents Med. Chem. 2022, 22, 2726–2756. [Google Scholar] [CrossRef] [PubMed]
  26. Sternbach, H. Are Antidepressants Carcinogenic? A Review of Preclinical and Clinical Studies. J. Clin. Psychiatry 2003, 64, 1153–1162. [Google Scholar] [CrossRef] [PubMed]
  27. Bielecka, A.M.; Obuchowicz, E. Antidepressant Drugs as a Complementary Therapeutic Strategy in Cancer. Exp. Biol. Med. 2013, 238, 849–858. [Google Scholar] [CrossRef] [PubMed]
  28. Tutton, P.J.; Barkla, D.H. Influence of Inhibitors of Serotonin Uptake on Intestinal Epithelium and Colorectal Carcinomas. Br. J. Cancer 1982, 46, 260–265. [Google Scholar] [CrossRef] [PubMed]
  29. Zheng, Y.; Chang, X.; Huang, Y.; He, D. The Application of Antidepressant Drugs in Cancer Treatment. Biomed. Pharmacother. 2023, 157, 113985. [Google Scholar] [CrossRef]
  30. Radin, D.P.; Patel, P. A Current Perspective on the Oncopreventive and Oncolytic Properties of Selective Serotonin Reuptake Inhibitors. Biomed. Pharmacother. 2017, 87, 636–639. [Google Scholar] [CrossRef]
  31. Varalda, M.; Antona, A.; Bettio, V.; Roy, K.; Vachamaram, A.; Yellenki, V.; Massarotti, A.; Baldanzi, G.; Capello, D. Psychotropic Drugs Show Anticancer Activity by Disrupting Mitochondrial and Lysosomal Function. Front. Oncol. 2020, 10, 562196. [Google Scholar] [CrossRef] [PubMed]
  32. Cho, Y.-W.; Kim, E.-J.; Nyiramana, M.; Shin, E.-J.; Jin, H.; Ryu, J.; Kang, K.; Lee, G.-W.; Kim, H.; Han, J.; et al. Paroxetine Induces Apoptosis of Human Breast Cancer MCF-7 Cells through Ca2+-and P38 MAP Kinase-Dependent ROS Generation. Cancers 2019, 11, 64. [Google Scholar] [CrossRef] [PubMed]
  33. Gillman, P.K. Tricyclic Antidepressant Pharmacology and Therapeutic Drug Interactions Updated. Br. J. Pharmacol. 2007, 151, 737–748. [Google Scholar] [CrossRef] [PubMed]
  34. Edinoff, A.N.; Akuly, H.A.; Hanna, T.A.; Ochoa, C.O.; Patti, S.J.; Ghaffar, Y.A.; Kaye, A.D.; Viswanath, O.; Urits, I.; Boyer, A.G.; et al. Selective Serotonin Reuptake Inhibitors and Adverse Effects: A Narrative Review. Neurol. Int. 2021, 13, 387–401. [Google Scholar] [CrossRef] [PubMed]
  35. Ashbury, J.E.; Lévesque, L.E.; Beck, P.A.; Aronson, K.J. A Population-Based Case-Control Study of Selective Serotonin Reuptake Inhibitors (SSRIs) and Breast Cancer: The Impact of Duration of Use, Cumulative Dose and Latency. BMC Med. 2010, 8, 90. [Google Scholar] [CrossRef]
  36. Toh, S.; García Rodríguez, L.A.; Hernández-Díaz, S. Use of Antidepressants and Risk of Lung Cancer. Cancer Causes Control 2007, 18, 1055–1064. [Google Scholar] [CrossRef]
  37. Xu, W.; Tamim, H.; Shapiro, S.; Stang, M.R.; Collet, J.-P. Use of Antidepressants and Risk of Colorectal Cancer: A Nested Case-Control Study. Lancet Oncol. 2006, 7, 301–308. [Google Scholar] [CrossRef] [PubMed]
  38. Coogan, P.F.; Strom, B.L.; Rosenberg, L. Antidepressant Use and Colorectal Cancer Risk. Pharmacoepidemiol. Drug Saf. 2009, 18, 1111–1114. [Google Scholar] [CrossRef] [PubMed]
  39. Wernli, K.J.; Hampton, J.M.; Trentham-Dietz, A.; Newcomb, P.A. Antidepressant Medication Use and Breast Cancer Risk. Pharmacoepidemiol. Drug Saf. 2009, 18, 284–290. [Google Scholar] [CrossRef]
  40. Gil-Ad, I.; Zolokov, A.; Lomnitski, L.; Taler, M.; Bar, M.; Luria, D.; Ram, E.; Weizman, A. Evaluation of the Potential Anti-Cancer Activity of the Antidepressant Sertraline in Human Colon Cancer Cell Lines and in Colorectal Cancer-Xenografted Mice. Int. J. Oncol. 2008, 33, 277–286. [Google Scholar] [CrossRef]
  41. Jiang, X.; Lu, W.; Shen, X.; Wang, Q.; Lv, J.; Liu, M.; Cheng, F.; Zhao, Z.; Pang, X. Repurposing Sertraline Sensitizes Non–Small Cell Lung Cancer Cells to Erlotinib by Inducing Autophagy. JCI Insight 2018, 3, e98921. [Google Scholar] [CrossRef] [PubMed]
  42. Ozunal, Z.G.; Cakil, Y.D.; Isan, H.; Saglam, E.; Aktas, R.G. Sertraline in Combination with Sorafenib: A Promising Pharmacotherapy to Target Both Depressive Disorders and Hepatocellular Cancer. Biol. Futura 2019, 70, 341–348. [Google Scholar] [CrossRef] [PubMed]
  43. Geeraerts, S.L.; Kampen, K.R.; Rinaldi, G.; Gupta, P.; Planque, M.; Louros, N.; Heylen, E.; De Cremer, K.; De Brucker, K.; Vereecke, S.; et al. Repurposing the Antidepressant Sertraline as SHMT Inhibitor to Suppress Serine/Glycine Synthesis–Addicted Breast Tumor Growth. Mol. Cancer Ther. 2021, 20, 50–63. [Google Scholar] [CrossRef] [PubMed]
  44. Baldissera, A.B.; Boia-Ferreira, M.; Basílio, A.B.C.; Resende, J.S.D.S.; Castro, M.A.A.; Chaim, O.M.; Gremski, L.H.; Veiga, S.S.; Senff-Ribeiro, A. Sertraline as a Potential Cancer Therapeutic Approach: Biological Relevance of TCTP in Breast Cancer Cell Lines and Tumors. Adv. Med. Sci. 2023, 68, 227–237. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, K.; Gong, Q.; Zhan, Y.; Chen, B.; Yin, T.; Lu, Y.; Zhang, Y.; Wang, H.; Ke, J.; Du, B.; et al. Blockage of Autophagic Flux and Induction of Mitochondria Fragmentation by Paroxetine Hydrochloride in Lung Cancer Cells Promotes Apoptosis via the ROS-MAPK Pathway. Front. Cell Dev. Biol. 2019, 7, 397. [Google Scholar] [CrossRef] [PubMed]
  46. Jang, W.-J.; Jung, S.K.; Vo, T.T.L.; Jeong, C.-H. Anticancer Activity of Paroxetine in Human Colon Cancer Cells: Involvement of MET and ERBB3. J. Cell. Mol. Med. 2019, 23, 1106–1115. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, V.C.-H.; Hsieh, Y.-H.; Chen, L.-J.; Hsu, T.-C.; Tzang, B.-S. Escitalopram Oxalate Induces Apoptosis in U-87MG Cells and Autophagy in GBM8401 Cells. J. Cell. Mol. Med. 2018, 22, 1167–1178. [Google Scholar] [CrossRef] [PubMed]
  48. Yuan, I.; Horng, C.-T.; Chen, V.C.-H.; Chen, C.-H.; Chen, L.-J.; Hsu, T.-C.; Tzang, B.-S. Escitalopram Oxalate Inhibits Proliferation and Migration and Induces Apoptosis in Non-Small Cell Lung Cancer Cells. Oncol. Lett. 2018, 15, 3376–3382. [Google Scholar] [CrossRef] [PubMed]
  49. Bavadekar, S.; Panchal, P.; Hanbashi, A.; Vansal, S. Cytotoxic Effects of Selective Serotonin- and Serotonin-Norepinephrine Reuptake Inhibitors on Human Metastatic Breast Cancer Cell Line, MCF-7 (842.3). FASEB J. 2014, 28, 842.3. [Google Scholar] [CrossRef]
  50. Magagnoli, J.; Narendran, S.; Pereira, F.; Cummings, T.H.; Hardin, J.W.; Sutton, S.S.; Ambati, J. Association between Fluoxetine Use and Overall Survival among Patients with Cancer Treated with PD-1/L1 Immunotherapy. Pharmaceuticals 2023, 16, 640. [Google Scholar] [CrossRef]
  51. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated Data and New Features for Efficient Prediction of Protein Targets of Small Molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [PubMed]
  52. Ciftci, H.I.; Radwan, M.O.; Sever, B.; Hamdy, A.K.; Emirdağ, S.; Ulusoy, N.G.; Sozer, E.; Can, M.; Yayli, N.; Araki, N.; et al. EGFR-Targeted Pentacyclic Triterpene Analogues for Glioma Therapy. Int. J. Mol. Sci. 2021, 22, 10945. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, K.-H.; Yang, S.-T.; Lin, Y.-K.; Lin, J.-W.; Lee, Y.-H.; Wang, J.-Y.; Hu, C.-J.; Lin, E.-Y.; Chen, S.-M.; Then, C.-K.; et al. Fluoxetine, an Antidepressant, Suppresses Glioblastoma by Evoking AMPAR-Mediated Calcium-Dependent Apoptosis. Oncotarget 2014, 6, 5088–5101. [Google Scholar] [CrossRef]
  54. You, F.; Zhang, C.; Liu, X.; Ji, D.; Zhang, T.; Yu, R.; Gao, S. Drug Repositioning: Using Psychotropic Drugs for the Treatment of Glioma. Cancer Lett. 2022, 527, 140–149. [Google Scholar] [CrossRef] [PubMed]
  55. Bi, J.; Khan, A.; Tang, J.; Armando, A.M.; Wu, S.; Zhang, W.; Gimple, R.C.; Reed, A.; Jing, H.; Koga, T.; et al. Targeting Glioblastoma Signaling and Metabolism with a Re-Purposed Brain-Penetrant Drug. Cell Rep. 2021, 37, 109957. [Google Scholar] [CrossRef] [PubMed]
  56. Dingledine, R.; Borges, K.; Bowie, D.; Traynelis, S.F. The Glutamate Receptor Ion Channels. Pharmacol. Rev. 1999, 51, 7–61. [Google Scholar] [PubMed]
  57. Sobolevsky, A.I.; Rosconi, M.P.; Gouaux, E. X-Ray Structure, Symmetry and Mechanism of an AMPA-Subtype Glutamate Receptor. Nature 2009, 462, 745–756. [Google Scholar] [CrossRef] [PubMed]
  58. Ma, J.; Yang, Y.-R.; Chen, W.; Chen, M.-H.; Wang, H.; Wang, X.-D.; Sun, L.-L.; Wang, F.-Z.; Wang, D.-C. Fluoxetine Synergizes with Temozolomide to Induce the CHOP-Dependent Endoplasmic Reticulum Stress-Related Apoptosis Pathway in Glioma Cells. Oncol. Rep. 2016, 36, 676–684. [Google Scholar] [CrossRef] [PubMed]
  59. Song, T.; Li, H.; Tian, Z.; Xu, C.; Liu, J.; Guo, Y. Disruption of NF-κB Signaling by Fluoxetine Attenuates MGMT Expression in Glioma Cells. OncoTargets Ther. 2015, 8, 2199–2208. [Google Scholar] [CrossRef]
  60. Bibbo’, S.; Lamolinara, A.; Capone, E.; Purgato, S.; Tsakaneli, A.; Panella, V.; Sallese, M.; Rossi, C.; Ciufici, P.; Nieddu, V.; et al. Repurposing a Psychoactive Drug for Children with Cancer: p27Kip1-Dependent Inhibition of Metastatic Neuroblastomas by Prozac. Oncogenesis 2020, 9, 1–8. [Google Scholar] [CrossRef]
  61. Hosseinimehr, S.J.; Najafi, S.H.; Shafiee, F.; Hassanzadeh, S.; Farzipour, S.; Ghasemi, A.; Asgarian-Omran, H. Fluoxetine as an Antidepressant Medicine Improves the Effects of Ionizing Radiation for the Treatment of Glioma. J. Bioenerg. Biomembr. 2020, 52, 165–174. [Google Scholar] [CrossRef] [PubMed]
  62. Patanaphan, V.; Salazar, O.M.; Risco, R. Breast Cancer: Metastatic Patterns and Their Prognosis. South. Med. J. 1988, 81, 1109–1112. [Google Scholar] [CrossRef] [PubMed]
  63. Nelson, H.D.; Smith, M.E.B.; Griffin, J.C.; Fu, R. Use of Medications to Reduce Risk for Primary Breast Cancer: A Systematic Review for the U.S. Preventive Services Task Force. Ann. Intern. Med. 2013, 158, 604–614. [Google Scholar] [CrossRef] [PubMed]
  64. Ong, J.C.-L.; Sun, F.; Chan, E. Development of Stealth Liposome Coencapsulating Doxorubicin and Fluoxetine. J. Liposome Res. 2011, 21, 261–271. [Google Scholar] [CrossRef] [PubMed]
  65. Zhou, T.; Duan, J.; Wang, Y.; Chen, X.; Zhou, G.; Wang, R.; Fu, L.; Xu, F. Fluoxetine Synergys with Anticancer Drugs to Overcome Multidrug Resistance in Breast Cancer Cells. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2012, 33, 1299–1306. [Google Scholar] [CrossRef] [PubMed]
  66. Radwan, M.O.; Kadasah, S.F.; Aljubiri, S.M.; Alrefaei, A.F.; El-Maghrabey, M.H.; El Hamd, M.A.; Tateishi, H.; Otsuka, M.; Fujita, M. Harnessing Oleanolic Acid and Its Derivatives as Modulators of Metabolic Nuclear Receptors. Biomolecules 2023, 13, 1465. [Google Scholar] [CrossRef] [PubMed]
  67. Kadasah, S.F.; Radwan, M.O. Overview of Ursolic Acid Potential for the Treatment of Metabolic Disorders, Autoimmune Diseases, and Cancers via Nuclear Receptor Pathways. Biomedicines 2023, 11, 2845. [Google Scholar] [CrossRef] [PubMed]
  68. Kabel, A.M.; Elkhoely, A.A. Ameliorative Potential of Fluoxetine/Raloxifene Combination on Experimentally Induced Breast Cancer. Tissue Cell 2016, 48, 89–95. [Google Scholar] [CrossRef] [PubMed]
  69. Tatar, O.; Ilhan, N.; Ilhan, N.; Susam, S.; Ozercan, I.H. Is There Any Potential Anticancer Effect of Raloxifene and Fluoxetine on DMBA-induced Rat Breast Cancer? J. Biochem. Mol. Toxicol. 2019, 33, e22371. [Google Scholar] [CrossRef]
  70. Duarte, D.; Cardoso, A.; Vale, N. Synergistic Growth Inhibition of HT-29 Colon and MCF-7 Breast Cancer Cells with Simultaneous and Sequential Combinations of Antineoplastics and CNS Drugs. Int. J. Mol. Sci. 2021, 22, 7408. [Google Scholar] [CrossRef]
  71. Duarte, D.; Rêma, A.; Amorim, I.; Vale, N. Drug Combinations: A New Strategy to Extend Drug Repurposing and Epithelial-Mesenchymal Transition in Breast and Colon Cancer Cells. Biomolecules 2022, 12, 190. [Google Scholar] [CrossRef] [PubMed]
  72. Duarte, D.; Falcão, S.I.; El Mehdi, I.; Vilas-Boas, M.; Vale, N. Honeybee Venom Synergistically Enhances the Cytotoxic Effect of CNS Drugs in HT-29 Colon and MCF-7 Breast Cancer Cell Lines. Pharmaceutics 2022, 14, 511. [Google Scholar] [CrossRef] [PubMed]
  73. Liao, P.-A.; Chu, P.-Y.; Tan, Z.-L.; Hsu, F.-T.; Lee, Y.-C.; Wu, H.-J. STAT3 Inactivation and Induction of Apoptosis Associate with Fluoxetine-Inhibited Epithelial-Mesenchymal Transition and Growth of Triple-Negative Breast Cancer In Vivo. Anticancer Res. 2022, 42, 3807–3814. [Google Scholar] [CrossRef] [PubMed]
  74. Bowie, M.; Pilie, P.; Wulfkuhle, J.; Lem, S.; Hoffman, A.; Desai, S.; Petricoin, E.; Carter, A.; Ambrose, A.; Seewaldt, V.; et al. Fluoxetine Induces Cytotoxic Endoplasmic Reticulum Stress and Autophagy in Triple Negative Breast Cancer. World J. Clin. Oncol. 2015, 6, 299–311. [Google Scholar] [CrossRef] [PubMed]
  75. Sun, D.; Zhu, L.; Zhao, Y.; Jiang, Y.; Chen, L.; Yu, Y.; Ouyang, L. Fluoxetine Induces Autophagic Cell Death via eEF2K-AMPK-mTOR-ULK Complex Axis in Triple Negative Breast Cancer. Cell Prolif. 2018, 51, e12402. [Google Scholar] [CrossRef] [PubMed]
  76. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  77. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  78. Kuwahara, J.; Yamada, T.; Egashira, N.; Ueda, M.; Zukeyama, N.; Ushio, S.; Masuda, S. Comparison of the Anti-Tumor Effects of Selective Serotonin Reuptake Inhibitors as Well as Serotonin and Norepinephrine Reuptake Inhibitors in Human Hepatocellular Carcinoma Cells. Biol. Pharm. Bull. 2015, 38, 1410–1414. [Google Scholar] [CrossRef] [PubMed]
  79. Mun, A.-R.; Lee, S.-J.; Kim, G.-B.; Kang, H.-S.; Kim, J.-S.; Kim, S.-J. Fluoxetine-Induced Apoptosis in Hepatocellular Carcinoma Cells. Anticancer Res. 2013, 33, 3691–3697. [Google Scholar]
  80. Hsu, L.-C.; Tu, H.-F.; Hsu, F.-T.; Yueh, P.-F.; Chiang, I.-T. Beneficial Effect of Fluoxetine on Anti-Tumor Progression on Hepatocellular Carcinoma and Non-Small Cell Lung Cancer Bearing Animal Model. Biomed. Pharmacother. 2020, 126, 110054. [Google Scholar] [CrossRef]
  81. Chen, W.-T.; Hsu, F.-T.; Liu, Y.-C.; Chen, C.-H.; Hsu, L.-C.; Lin, S.-S. 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. 2019, 20, 757. [Google Scholar] [CrossRef] [PubMed]
  82. Abdel-Bar, H.M.; Khater, S.E.; Ghorab, D.M.; Al-mahallawi, A.M. Hexosomes as Efficient Platforms for Possible Fluoxetine Hydrochloride Repurposing with Improved Cytotoxicity against HepG2 Cells. ACS Omega 2020, 5, 26697–26709. [Google Scholar] [CrossRef] [PubMed]
  83. Siegel, R.L.; Wagle, N.S.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 233–254. [Google Scholar] [CrossRef] [PubMed]
  84. Islami, F.; Goding Sauer, A.; Miller, K.D.; Siegel, R.L.; Fedewa, S.A.; Jacobs, E.J.; McCullough, M.L.; Patel, A.V.; Ma, J.; Soerjomataram, I.; et al. Proportion and Number of Cancer Cases and Deaths Attributable to Potentially Modifiable Risk Factors in the United States. CA Cancer J. Clin. 2018, 68, 31–54. [Google Scholar] [CrossRef] [PubMed]
  85. Engelmann, B.J.; Ryan, J.J.; Farrell, N.P. Antidepressants and Platinum Drugs. Anticancer Res. 2014, 34, 509–516. [Google Scholar] [PubMed]
  86. Kang, B.-G.; Shende, M.; Inci, G.; Park, S.-H.; Jung, J.-S.; Kim, S.B.; Kim, J.H.; Mo, Y.W.; Seo, J.-H.; Feng, J.-H.; et al. Combination of Metformin/Efavirenz/Fluoxetine Exhibits Profound Anticancer Activity via a Cancer Cell-Specific ROS Amplification. Cancer Biol. Ther. 2023, 24, 2161803. [Google Scholar] [CrossRef] [PubMed]
  87. Marcinkute, M.; Afshinjavid, S.; Fatokun, A.A.; Javid, F.A. Fluoxetine Selectively Induces P53-Independent Apoptosis in Human Colorectal Cancer Cells. Eur. J. Pharmacol. 2019, 857, 172441. [Google Scholar] [CrossRef] [PubMed]
  88. Minagawa, Y.; Kigawa, J.; Itamochi, H.; Kanamori, Y.; Shimada, M.; Takahashi, M.; Terakawa, N. Cisplatin-resistant HeLa Cells Are Resistant to Apoptosis via P53-dependent and -independent Pathways. Jpn. J. Cancer Res. Gann 1999, 90, 1373–1379. [Google Scholar] [CrossRef] [PubMed]
  89. Charles, E.; Hammadi, M.; Kischel, P.; Delcroix, V.; Demaurex, N.; Castelbout, C.; Vacher, A.-M.; Devin, A.; Ducret, T.; Nunes, P.; et al. The Antidepressant Fluoxetine Induces Necrosis by Energy Depletion and Mitochondrial Calcium Overload. Oncotarget 2016, 8, 3181–3196. [Google Scholar] [CrossRef]
  90. Liu, Y.; Li, T.; Xu, M.; Che, X.; Jiang, X. Fluoxetine Enhances Cellular Chemosensitivity to Cisplatin in Cervical Cancer. Int. J. Clin. Exp. Med. 2017, 10, 10521–10527. [Google Scholar]
  91. Naz, A.; Hashim, F.; Ali, S.A.; Badshah, M. Fabrication, Characterization and Therapeutic Evaluation of Fluoxetine-Dextran Nanoparticles. ChemistrySelect 2023, 8, e202204110. [Google Scholar] [CrossRef]
  92. Sagnella, S.M.; Duong, H.; MacMillan, A.; Boyer, C.; Whan, R.; McCarroll, J.A.; Davis, T.P.; Kavallaris, M. Dextran-Based Doxorubicin Nanocarriers with Improved Tumor Penetration. Biomacromolecules 2014, 15, 262–275. [Google Scholar] [CrossRef] [PubMed]
  93. Yamaoka, T.; Kusumoto, S.; Ando, K.; Ohba, M.; Ohmori, T. Receptor Tyrosine Kinase-Targeted Cancer Therapy. Int. J. Mol. Sci. 2018, 19, 3491. [Google Scholar] [CrossRef] [PubMed]
  94. Wu, J.-Y.; Lin, S.-S.; Hsu, F.-T.; Chung, J.-G. Fluoxetine Inhibits DNA Repair and NF-ĸB-Modulated Metastatic Potential in Non-Small Cell Lung Cancer. Anticancer Res. 2018, 38, 5201–5210. [Google Scholar] [CrossRef] [PubMed]
  95. Yang, Z.; Li, Z.; Guo, Z.; Ren, Y.; Zhou, T.; Xiao, Z.; Duan, J.; Han, C.; Cheng, Y.; Xu, F. Antitumor Effect of Fluoxetine on Chronic Stress-Promoted Lung Cancer Growth via Suppressing Kynurenine Pathway and Enhancing Cellular Immunity. Front. Pharmacol. 2021, 12, 685898. [Google Scholar] [CrossRef]
  96. Park, W.; Chawla, A.; O’Reilly, E.M. Pancreatic Cancer: A Review. JAMA 2021, 326, 851–862. [Google Scholar] [CrossRef] [PubMed]
  97. Aakjær, M.; Kristiansen, S.B.; Pape, K.; Sessa, M.; Dalhoff, K.P.; De Bruin, M.L.; Andersen, M. Investigation of the Potential Association between the Use of Fluoxetine and Occurrence of Acute Pancreatitis: A Danish Register-Based Cohort Study. Int. J. Epidemiol. 2022, 51, 1656–1665. [Google Scholar] [CrossRef] [PubMed]
  98. Jia, L.; Shang, Y.-Y.; Li, Y.-Y. Effect of Antidepressants on Body Weight, Ethology and Tumor Growth of Human Pancreatic Carcinoma Xenografts in Nude Mice. World J. Gastroenterol. 2008, 14, 4377–4382. [Google Scholar] [CrossRef] [PubMed]
  99. Schneider, M.A.; Heeb, L.; Beffinger, M.M.; Pantelyushin, S.; Linecker, M.; Roth, L.; Lehmann, K.; Ungethüm, U.; Kobold, S.; Graf, R.; et al. Attenuation of Peripheral Serotonin Inhibits Tumor Growth and Enhances Immune Checkpoint Blockade Therapy in Murine Tumor Models. Sci. Transl. Med. 2021, 13, eabc8188. [Google Scholar] [CrossRef]
  100. Matasar, M.J.; Zelenetz, A.D. Overview of Lymphoma Diagnosis and Management. Radiol. Clin. N. Am. 2008, 46, 175–198. [Google Scholar] [CrossRef]
  101. Mugnaini, E.N.; Ghosh, N. Lymphoma. Prim. Care 2016, 43, 661–675. [Google Scholar] [CrossRef] [PubMed]
  102. Thomas, D.A.; Faderl, S.; O’Brien, S.; Bueso-Ramos, C.; Cortes, J.; Garcia-Manero, G.; Giles, F.J.; Verstovsek, S.; Wierda, W.G.; Pierce, S.A.; et al. Chemoimmunotherapy with Hyper-CVAD plus Rituximab for the Treatment of Adult Burkitt and Burkitt-Type Lymphoma or Acute Lymphoblastic Leukemia. Cancer 2006, 106, 1569–1580. [Google Scholar] [CrossRef] [PubMed]
  103. Cloonan, S.M.; Drozgowska, A.; Fayne, D.; Williams, D.C. The Antidepressants Maprotiline and Fluoxetine Have Potent Selective Antiproliferative Effects against Burkitt Lymphoma Independently of the Norepinephrine and Serotonin Transporters. Leuk. Lymphoma 2010, 51, 523–539. [Google Scholar] [CrossRef] [PubMed]
  104. Cloonan, S.M.; Williams, D.C. The Antidepressants Maprotiline and Fluoxetine Induce Type II Autophagic Cell Death in Drug-Resistant Burkitt’s Lymphoma. Int. J. Cancer 2011, 128, 1712–1723. [Google Scholar] [CrossRef] [PubMed]
  105. Di Rosso, M.E.; Sterle, H.A.; Cremaschi, G.A.; Genaro, A.M. Beneficial Effect of Fluoxetine and Sertraline on Chronic Stress-Induced Tumor Growth and Cell Dissemination in a Mouse Model of Lymphoma: Crucial Role of Antitumor Immunity. Front. Immunol. 2018, 9, 306460. [Google Scholar] [CrossRef]
  106. Chomchoei, C.; Brimson, J.M.; Brimson, S. Repurposing Fluoxetine to Treat Lymphocytic Leukemia: Apoptosis Induction, Sigma-1 Receptor Upregulation, Inhibition of IL-2 Cytokine Production, and Autophagy Induction. Expert Opin. Ther. Targets 2022, 26, 1087–1097. [Google Scholar] [CrossRef] [PubMed]
  107. Tan, B.; Piwnica-Worms, D.; Ratner, L. Multidrug Resistance Transporters and Modulation. Curr. Opin. Oncol. 2000, 12, 450–458. [Google Scholar] [CrossRef] [PubMed]
  108. Emran, T.B.; Shahriar, A.; Mahmud, A.R.; Rahman, T.; Abir, M.H.; Siddiquee, M.F.-R.; Ahmed, H.; Rahman, N.; Nainu, F.; Wahyudin, E.; et al. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front. Oncol. 2022, 12, 891652. [Google Scholar] [CrossRef]
  109. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug Resistance in Cancer: Role of ATP–Dependent Transporters. Nat. Rev. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef] [PubMed]
  110. Peer, D.; Dekel, Y.; Melikhov, D.; Margalit, R. Fluoxetine Inhibits Multidrug Resistance Extrusion Pumps and Enhances Responses to Chemotherapy in Syngeneic and in Human Xenograft Mouse Tumor Models. Cancer Res. 2004, 64, 7562–7569. [Google Scholar] [CrossRef]
  111. Wilkens, S. Structure and Mechanism of ABC Transporters. F1000Prime Rep. 2015, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  112. Peña-Solórzano, D.; Stark, S.A.; König, B.; Sierra, C.A.; Ochoa-Puentes, C. ABCG2/BCRP: Specific and Nonspecific Modulators. Med. Res. Rev. 2017, 37, 987–1050. [Google Scholar] [CrossRef] [PubMed]
  113. Stefan, S.M.; Wiese, M. Small-Molecule Inhibitors of Multidrug Resistance-Associated Protein 1 and Related Processes: A Historic Approach and Recent Advances. Med. Res. Rev. 2019, 39, 176–264. [Google Scholar] [CrossRef] [PubMed]
  114. Bin Kanner, Y.; Teng, Q.-X.; Ganoth, A.; Peer, D.; Wang, J.-Q.; Chen, Z.-S.; Tsfadia, Y. Cytotoxicity and Reversal Effect of Sertraline, Fluoxetine, and Citalopram on MRP1- and MRP7-Mediated MDR. Front. Pharmacol. 2023, 14, 1290255. [Google Scholar] [CrossRef] [PubMed]
  115. Panchal, N.K.; Sabina, E.P. A Serine/Threonine Protein PIM Kinase as a Biomarker of Cancer and a Target for Anti-Tumor Therapy. Life Sci. 2020, 255, 117866. [Google Scholar] [CrossRef] [PubMed]
  116. Du, R.; Huang, C.; Liu, K.; Li, X.; Dong, Z. Targeting AURKA in Cancer: Molecular Mechanisms and Opportunities for Cancer Therapy. Mol. Cancer 2021, 20, 15. [Google Scholar] [CrossRef] [PubMed]
  117. Kiyoi, H.; Kawashima, N.; Ishikawa, Y. FLT3 Mutations in Acute Myeloid Leukemia: Therapeutic Paradigm beyond Inhibitor Development. Cancer Sci. 2020, 111, 312–322. [Google Scholar] [CrossRef] [PubMed]
  118. Domoto, T.; Uehara, M.; Bolidong, D.; Minamoto, T. Glycogen Synthase Kinase 3β in Cancer Biology and Treatment. Cells 2020, 9, 1388. [Google Scholar] [CrossRef]
  119. Zoete, V.; Daina, A.; Bovigny, C.; Michielin, O. SwissSimilarity: A Web Tool for Low to Ultra High Throughput Ligand-Based Virtual Screening. J. Chem. Inf. Model. 2016, 56, 1399–1404. [Google Scholar] [CrossRef]
  120. Ceci, C.; Atzori, M.G.; Lacal, P.M.; Graziani, G. Role of VEGFs/VEGFR-1 Signaling and Its Inhibition in Modulating Tumor Invasion: Experimental Evidence in Different Metastatic Cancer Models. Int. J. Mol. Sci. 2020, 21, 1388. [Google Scholar] [CrossRef]
  121. Kiselyov, A.S.; Semenova, M.; Semenov, V.V.; Piatnitski, E. 2-((1H-Azol-1-Yl)Methyl)-N-Arylbenzamides: Novel Dual Inhibitors of VEGFR-1/2 Kinases. Bioorg. Med. Chem. Lett. 2006, 16, 1726–1730. [Google Scholar] [CrossRef]
  122. Zhang, Y.; Liu, H.; Jiao, Y.; Yuan, H.; Wang, F.; Lu, S.; Yao, S.; Ke, Z.; Tai, W.; Jiang, Y.; et al. De Novo Design of N-(Pyridin-4-Ylmethyl)Aniline Derivatives as KDR Inhibitors: 3D-QSAR, Molecular Fragment Replacement, Protein-Ligand Interaction Fingerprint, and ADMET Prediction. Mol. Divers. 2012, 16, 787–802. [Google Scholar] [CrossRef]
  123. Okram, B.; Uno, T.; Ding, Q.; Liu, Y.; Jin, Y.; Jin, Q.; Wu, X.; Che, J.; Yan, S.F.; Hao, X. Compounds and Compositions as Kinase Inhibitors. U.S. Patent US8546370B2, 28 August 2009. [Google Scholar]
  124. Patel, D.D.; Galarneau, D. Serotonin Syndrome with Fluoxetine: Two Case Reports. Ochsner J. 2016, 16, 554–557. [Google Scholar]
  125. Chen, W.T.; Tsai, Y.H.; Tan, P.; Hsu, F.T.; Wang, H.M.D.; Lin, W.C.; Lin, F.H.; Wu, C.T. Fluoxetine Inhibits STAT3-Mediated Survival and Invasion of Osteosarcoma Cells. Anticancer Res. 2023, 43, 1193–1199. [Google Scholar] [CrossRef]
Figure 1. The number of scientific publications comprising scientific output linking repurposing studies with cancer over the period 2005–2023. Data were retrieved from the Web of Science database by using the keywords “repurposing” and “cancer”.
Figure 1. The number of scientific publications comprising scientific output linking repurposing studies with cancer over the period 2005–2023. Data were retrieved from the Web of Science database by using the keywords “repurposing” and “cancer”.
Ijms 25 06314 g001
Figure 2. Chemical structure of fluoxetine (FLX) and summary of different sensitive cancers to fluoxetine treatment.
Figure 2. Chemical structure of fluoxetine (FLX) and summary of different sensitive cancers to fluoxetine treatment.
Ijms 25 06314 g002
Figure 3. Similarity of fluoxetine (FLX) to a VEGFR2 inhibitor, CHEMBL437889; and a multitarget kinase inhibitor, CHEMBL3642580.
Figure 3. Similarity of fluoxetine (FLX) to a VEGFR2 inhibitor, CHEMBL437889; and a multitarget kinase inhibitor, CHEMBL3642580.
Ijms 25 06314 g003
Table 1. Summary of fluoxetine effect on cancer cells.
Table 1. Summary of fluoxetine effect on cancer cells.
Cancer OriginCellsConditionsMain Findings
BrainU87In vitro (25 μM) and in vivo (10 mg/kg)Apoptosis by increasing the intracellular Ca2+ concentration via binding to AMPAR [53]
C6, U373 and U251In vitro Respective IC50 14.7, 21.8, 48.5, and 22.9 μM. Apoptosis can be promoted by enhancing caspase-3 and CHOP concentrations and ERS [58]
C6In vitro, combined with TMZSynergistic antiproliferative effect mediated by CHOP pathway [58]
U-138 MGIn vitro (10 μM) and in vivo, combined with TMZSynergistic antiproliferative effect by suppressing MGMT expression and disrupting NF-kB/p65 signaling [59]
U87 RPE1, IMR90, and U87EGFRvIIIIn vitro and in vivo (10, 15, and 16.4 mg/kg), alone or combined with TMZFLX reduced cell viability by suppressing SMPD1 and EGFRvIII dose-dependently. The combination is synergistic [55]
Kelly cellsIn vitro (10 μM), in vivo (20 mg/kg)Inhibited CKS1, increased p27Kip1 expression, and triggered neuroblastoma cell death [60]
U-87 MGIn vitro (10 μM), combined with radiotherapyRadiosensitizer on cancer cells and radioprotective on normal cells HFFF2 [61]
BreastMCF-7In vitroIC50 7.78 µM [70]
MCF-7In vitro (10 µM) combined with PXL, doxorubicin, or honeybee venom,Synergistic reduction of cell viability [70,71,72]
MCF-7/ADMIn vitro (5 μg/mL) and combined with ADMSignificant enhancement of ADM cytotoxic effect with IC50 change from 13.62 µm to 2.7 µm. Effective reversal of ADM resistance and synergistic apoptosis via upregulation of p53 and downregulation of (Bcl-2) [65]
(DMBA)-induced breast cancerIn vivo (30 mg/kg), combined with RALSynergistic antitumor effect and reduced concentration of TNF-α, IL-6, and TGF-β [68]
SUM149PTIn vitroInduced cytotoxicity (IC50 μM 7.9) and apoptosis and enhanced ER stress and autophagy with less effect on normal MCF-10A cells [74]
MDA-MB-231 and MDA-MB-436In vitro (1 μmol/L FLX)Induced apoptotic and autophagic cell death via caspases, mTOR, and AMPK pathways [75]
LiverHep3BIn vitro (10, 30, and 100 µM)Induced apoptosis, enhanced ROS formation, suppression of pERK1/2, and increased c-JNK dose-dependently [79]
Hep3BIn vivo (10 mg/kg)Antitumor effect by promoting caspases and suppressing VEGF, MMP-9, and NF-kB p65 [80]
SK-Hep1In vitro (30 μM)Induction of apoptosis and reduced invasiveness through ERK/NF-κB suppression [81]
HepG2In vitro, FLX-loaded hexosomeProlonged release with better efficacy than FLX [82]
ColonHT-29In vitroIC50 6.12 µM [70]
HT-29In vitro (10 µM), combined with honeybee venomSynergistic antiproliferative effect [72]
HCT116In vitro (20 µM), combined with cisplatin or carboplatinSynergistic antiproliferative effect by calmodulin inhibition [85]
HCT116In vitro (0.9 μM) and in vivo (2.67 mg/kg), combined with metformin/EfavirenzSynergism, enhanced apoptosis, and autophagy with massive increase in ROS levels [86]
p53 gene-depleted HCT116/In vitroIC50 4.73 µM, cell-cycle arrest [87]
CervixHeLa cellsIn vitro (50 μM)Reduce viability by increasing cytosolic Ca2+ concentration [89]
HeLa cellsIn vitro (16 µM) and in vivo (3 mg/kg), combined with cisplatin (16 µM)Synergism is achieved by upregulating apoptotic proteins and downregulating MDR proteins [90]
HeLa cellsFLX–dextran nanoparticlesSimilar cytotoxic effect of 30 μM FLX HeLa with less toxicity to normal cells 3T3 [91]
LungCL1-5-F4In vitroIC50 40 µM by suppression of MMP-2, MMP-9, uPA, and VEGF [94]
CL1-5-F4In vivo (10 mg/kg)Suppression of tumor growth and impeding angiogenesis and invasion [80]
A549In vivo (20 mg/kg)Suppressed stress-induced tumor growth and kynurenine pathway and enhanced cellular immunity [95]
PancreasSW1990In vivo (10 mg/kg)Insignificant tumor growth suppression [98]
Panc02In vivo (20 mg/kg)Suppression of tumor growth by upregulation of PD-L1 expression [99]
LymphMUTU-IIn vitro (50 μM)Apoptosis via caspase pathway, DNA cleavage, and PARP cleavage [103]
DG-75In vitro (50 μM)Autophagy via increasing Beclin-1 and extracellular Ca2+ influx [104]
EL4In vivo (15 mg/kg)Attenuated tumor growth, invasiveness, and metastasis targeting cyclins, MMP-2 and MMP-9, and TIMPs [105]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kadasah, S.F.; Alqahtani, A.M.S.; Alkhammash, A.; Radwan, M.O. Beyond Psychotropic: Potential Repurposing of Fluoxetine toward Cancer Therapy. Int. J. Mol. Sci. 2024, 25, 6314. https://doi.org/10.3390/ijms25126314

AMA Style

Kadasah SF, Alqahtani AMS, Alkhammash A, Radwan MO. Beyond Psychotropic: Potential Repurposing of Fluoxetine toward Cancer Therapy. International Journal of Molecular Sciences. 2024; 25(12):6314. https://doi.org/10.3390/ijms25126314

Chicago/Turabian Style

Kadasah, Sultan F., Abdulaziz M. S. Alqahtani, Abdullah Alkhammash, and Mohamed O. Radwan. 2024. "Beyond Psychotropic: Potential Repurposing of Fluoxetine toward Cancer Therapy" International Journal of Molecular Sciences 25, no. 12: 6314. https://doi.org/10.3390/ijms25126314

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop