1. Introduction
Breast cancer is the most prevalent malignancy globally, with triple-negative breast cancer (TNBC) accounting for 15–20% of all diagnoses [
1]. TNBC is characterized by the absence of estrogen and progesterone receptors, along with human epidermal growth factor receptor 2 (HER2) [
2], making it unresponsive to hormone and HER2-targeted therapies that benefit other breast cancer subtypes [
3]. As a result, treatment options for TNBC are largely confined to surgery and chemotherapy. Nonetheless, the high recurrence rates and limited overall survival (OS) associated with these approaches make the development of more effective therapeutic strategies a pressing priority [
4,
5].
This unmet clinical need is even more apparent if we consider that current advancements with poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint (IC) inhibitors show limited effectiveness in TNBC, beneficial only to specific patient subsets [
6]. Indeed, only 15–25% of TNBC cases harbor germline BRCA1/2 (gBRCA) mutations [
7], and PD-L1 protein is detected in about one-fifth of all TNBC patients [
8]. Furthermore, while these therapies demonstrate efficacy in early-stage TNBC, their impact on advanced disease remains largely insufficient [
6].
A hallmark of cancer metabolism is the shift from mitochondrial to aerobic glycolysis, a phenomenon known as the Warburg effect [
9]. This metabolic adaptation leads to the accumulation of acidic glycolytic byproducts, yet cancer cells maintain an alkaline intracellular pH [
10]. This apparent contradiction is explained by the increased activity or expression of pH-regulatory proteins, many of which are Na
+ transporters that induce a net sodium entry into the cell [
10]. Early studies from our group showed that mitochondrial dysfunction in primary, non-transformed rodent hepatocytes, triggers an irreversible rise in intracellular Na
+ levels which, altering the mechanisms of cell volume maintenance, causes hepatocyte death. Congruently, preventing this Na
+ accumulation conferred enhanced resistance to cellular damage [
11,
12,
13].
Based on these observations, we hypothesize that TNBC cancer cells, by inherently containing elevated intracellular Na+ levels compared with normal cells, would be energetically unable to compensate for further increases in Na+ concentration induced by pharmacological treatment with sodium ionophore compounds.
Consistently, our recent research on a mouse model of HCC [
14] demonstrated that liver cancer tissues exhibited significantly elevated levels of intracellular Na⁺ compared with normal tissues, a disparity that was further exacerbated by the treatment with the antibiotic cation ionophore monensin [
14]. Monensin selectively killed HCC cells and not healthy hepatocytes [
14] by inducing sodium-dependent mitochondrial alterations, ATP loss, and energy-dependent inhibition of the Na
+/K
+ pump with a consequent irreversible increase in intracellular sodium. This condition of permanent energetic stress made HCC cells unable to keep, unlike healthy hepatocytes, life-compatible levels of Na
+ with a consequent increase in the cellular osmotic pressure and death of the cancer cells [
14].
This research was the first to demonstrate that cancer cells are acutely sensitive to a pharmacologically induced Na+ overload via monensin.
The finding that the antibiotic monensin possessed a potent anticancer activity had been, however, previously established by several other studies [
15,
16,
17,
18,
19]. Monensin has been, in fact, shown to inhibit cell proliferation and induce apoptosis across different cancer cell types, including those exhibiting multidrug resistance [
15] and breast cancer cells [
19]. Such effects were found to be associated with the inhibition of signal mediators involved in the regulation of cell division and apoptosis, such those of the Wnt/β-Catenin, EGF receptor, or SUMO signal pathway [
15,
16,
17,
18,
19]. In the specific case of the breast cancer cells, monensin was reported to induce cytotoxicity and repress colony cell formation by inhibiting a component of the SUMO pathway, UBA2 [
19].
None of these previous studies, however, linked the anticancer activity of monensin to its primary chemical property as a cation ionophore [
20]. Monensin is, in fact, a polyether cation ionophore able to reversibly tie and transport Na
+ from the extracellular liquid ([Na
+]e ≃ 145 mM) to the cytoplasm ([Na
+]i ≃ 15 mM) along its concentration slope [
20].
Aerobic glycolytic metabolism and alkaline intracellular pH are nearly ubiquitous features of cancer tissues, with increased intracellular Na
+ levels likely arising as a consequence of these metabolic conditions [
9,
10]. Significantly, this relationship has been substantiated by recent 23Na-magnetic resonance imaging analyses of various human cancers [
21,
22,
23,
24,
25,
26], including breast cancer [
24,
25], that consistently identified aberrantly high intracellular Na
+ levels in cancerous tissues relative to adjacent normal tissues. These findings not only reveal Na
+ concentration as a novel diagnostic biomarker for human tumors [
26] but also provide a compelling scientific base for the hypothesis that triple-negative breast cancer (TNBC) could be specifically sensitive to a further sodium accumulation and that TNBC cells would be specifically killed upon exposure to the sodium ionophore monensin, thus inducing TNBC inhibition by a mechanism unrelated to cytostatic effects.
The present study investigated these issues, exploring, for the first time in a mouse in vivo model, the anticancer potential of monensin against TNBC by focusing on its ability to influence Na+ dynamics and cell viability and analyzing the possible conditions responsible for the inconsistencies of the results regarding the cytostatic or cytotoxic effects of monensin.
To this purpose, using both in vitro and in vivo imaging approaches, we measured Na+ accumulation and cancer cell death in murine TNBC cells (4T1-Luc2), alongside tumor mass reduction in mice injected with 4T1-Luc2 cells and intraperitoneally (i.p.) with monensin. We developed a novel methodology able, unlike the available techniques, to estimate in vivo the intracellular variations of the sodium level and explored the effects of monensin on cancer and healthy tissues, focusing on markers of proliferation.
Our findings confirmed that monensin selectively induced massive necrosis in TNBC tumors through a sodium-dependent mechanism without affecting the proliferative activity of transformed and normal tissues, thus distinguishing its cytotoxic from cytostatic effects as also enlightened by the role of differential in vitro settings in producing the discrepancies regarding its mechanism of action.
Collectively, these results underscore the potential of targeting Na⁺ homeostasis with sodium ionophores like monensin as innovative therapeutic agents for TNBC able to specifically kill TNBC cells without altering healthy tissues.
2. Materials and Methods
2.1. Chemicals and Reagents
Fetal bovine serum (FBS), penicillin (P), streptomycin (S), monensin, trypan blue, 2-hydroxypropyl-β-cyclodextrin (HBCD), acrylamide/bis-acrylamide (30% solution), Amersham Hybond PVDF Blotting Membrane, anti-PCNA (Ab-1) mouse mAb (PC10) 1:100, monoclonal anti-β-actin mouse antibody 1:1000, and all chemicals for buffer and reagent preparations were obtained from Sigma-Aldrich (MO, USA). PierceTM RIPA buffer, NovexTM tris-glycine transfer buffer, PierceTM BCA protein assay, PierceTM ECL Western blotting substrate, and all chemicals for buffer and reagent preparations for Western blot analysis were purchased from Thermo Fisher Scientific (MA, USA). Roswell Park Memorial Institute medium (RPMI), Dulbecco’s modified eagle medium (DMEM), and custom-made Na+-free DMEM (DMEM-Na+), with no Na+ constituents, were obtained from GIBCO (S.I.A.L. group, Rome, Italy). ION NaTRIUM Green-AM was obtained from Abcam (Cambridge, UK), and CoroNa™ Green, AM, was from Thermo Fisher Scientific (MA, USA). Ultra Rediject salt D-luciferin was obtained from Perkin Elmer (MA, USA).
2.2. Cell Culture
Murine 4T1-Luc2-Luc2 (4T1-Luc2) mammary tumor cells, murine B16-F10-Luc (B16) melanoma tumor cells, human SK-OV-3 (SKOV3) ovarian tumor cells, and murine Hepa-1Hepa-C1C7 (HEPA-C1C7) were supplied by LGC Ltd. (Middlesex, UK). Human MCF7 mammary tumor cells were obtained from American Type Culture Collection (ATCC; Manassas, VA, USA).
The 4T1-Luc2, B16, and SKOV3 cells were routinely cultured in RPMI supplemented with 10% FBS, 100 U/mL penicillin, 100 μg/mL streptomycin. HEPA-C1C7 and MCF7 cells were cultured in DMEM without glutamine, added with 10% FBS, 100 U/mL penicillin, 6 mM glutamine, and 100 μg/mL streptomycin (all materials were from Lonza Bioscience, Basel, Switzerland). Cells were kept under 5% CO
2/95% air atmosphere at 37 °C as previously described [
14]. All cell lines were tested for mycoplasma contamination using the MycoAlert™ mycoplasma detection kit.
2.3. Cell Treatments
For in vitro cell imaging analysis or for the measurement of cell viability, ATP, and Na+ content, 4T1-Luc2 or MCF7 cells were seeded (1.5 × 105/mL cell density) in RPMI and treated with 1 μM monensin (or vehicle) after 24 h in DMEM with normal (145 mM) sodium concentration (+Na+) or without any sodium component (−Na+) (GIBCO, S.I.A.L, Rome, Italy) in the absence of serum.
For WB analysis, 4T1-Luc2-Luc2 (4T1-Luc2), B16, SKOV3, and HEPA-C1C7 cells were seeded (1.25 × 105/mL cell density) in RPMI or DMEM (HEPA-C1C7) and treated with 5 μM monensin (or vehicle) after 16–24 h in DMEM/RPMI supplemented or not with fetal bovine serum (FBS, 10%).
2.4. Cell Viability, ATP, and Na+ Content
Cell viability and intracellular ATP were determined by using, respectively, the CellTox and Cell-Titer protocols (Promega; Madison, WI, USA) as previously reported [
14]. The ATP values were corrected excluding ATP released by dead cells, as determined by simultaneous CellTox and Cell-Titer analyses performed for each sample. Viability and ATP values were expressed as a % of corresponding controls.
The 4T1-Luc2-Luc 2 and MCF7 cells were loaded with the intracellular Na+-fluorescent dye ION NaTRIUM Green-AM (5 μM) in Hank’s Balanced Salt Solution (HBSS) with BSA (2%), pluronic acid (10 μM), and glucose (10 mM) for 1 h at RT. Intracellular Na+ variations upon monensin treatment were monitored using a Kontron SFM25 spectrofluorometer set at 525 nm excitation and at 545 nm emission wavelengths and expressed as a % of control values.
2.5. In Vitro Cell Imaging Analysis of CoroNa Green, AM/Cytotox NIR Dye Fluorescence
The 4T1-Luc2 cells were seeded in a 96-well plate for 24 h in DMEM supplemented with 10% FBS and subsequently loaded with the fluorescent dye CoroNa Green-AM. This dye acts as a sodium ion indicator requiring intracellular de-esterification to bind Na+, with Na+ binding leading to an increase in green fluorescence emission intensity with minimal wavelength shift. The 4T1-Luc2 cells were incubated with the fluorescent probe (5 μM) in HBSS containing BSA (4%) and pluronic acid (0.02%) at 37 °C for 1 h and then washed twice with HBSS. Cells were then exposed to monensin (1 μM) and the cell-impermeant cytotox NIR dye (0.6 μM) to visualize the nuclei of dead cells and maintained for 20 h in DMEM in the presence or absence of Na+. Intracellular Na+ variations were recorded every 3 min, while cytotoxicity was assessed hourly using a semi-automated analysis protocol of the IncuCyte SX5 Live® Cell Analysis System. The Sartorius® probe in the same imaging system facilitated pre-set parameters for optimal fluorescent outputs.
The in vitro (Incucyte) fluorescence intensity data were collected from the Sartorius Incucyte 2022B Rev2 software and analyzed through the GraphPad Prism 8 software. Raw fluorescence intensity data, measured by the instrument as total green object integrated intensity (GCU × um2/Image) or as total NIR object integrated intensity (NIRCU × um2/Image), were used for the graphs. The intracellular sodium probe (CoroNa Green-AM; max Ex.: 492 nm, max Em.: 516 nm) was captured by using the default green channel (Ex.: 453–485 nm, Em.: 494–533 nm), while the cytotoxicity probe (IncuCyte® Cytotox NIR Dye; max Ex.: 665 nm, max Em.: 695 nm) was captured by using the default near-infrared channel (Ex.: 648–674 nm, Em.: 685–756 nm). All the images were acquired through the included 20× (0.62 um/px) objective. The in vivo (IVIS Spectrum) fluorescence intensity data were collected from and analyzed through the Living Image ® 4.7.4 software (Revvity). Raw fluorescence intensity data, measured by the instrument as p/sec/cm2/sr and analyzed through the Living Image™ software, were used for the graphs. The luciferin images were captured using the protocol provided by the producer (Bioluminescence, open filter, automatic exposure), while the intracellular sodium probe was captured by using a 500/40 fluorescence filter and automatic exposure.
2.6. Quantitative Expression of Proliferating Cell Nuclear Antigen (PCNA)
Protein extracts from 4T1-Luc2, HEPA-C1C7, B16, and SKOV3 cells were electrophoresed by SDS/PAGE (10% gel), and after blotting them onto the PVDF membranes, the membranes were probed with antibodies against PCNA ( Sigma-Aldrich (MO, USA)). The β-actin monoclonal antibody (Sigma-Aldrich (MO, USA ) was used to assess equal protein loading. The antigens were detected by ECL Western Blotting Substrate (Thermo Fisher Scientific, MA, USA) and a ChemiDoc MP quantitative imaging system (BioRad Laboratories, Milan, Italy). The results were expressed as ratios.
2.7. In Vivo Experiments
The 4T1-Luc2-Luc2 tumor cells (100,000 cells in 100 μL of 0.9% NaCl per mouse) were orthotopically injected into the fourth mammary fat pad of female BALB/c mice aged six to eight weeks (Envigo, Inc., IN, USA). Tumor growth was monitored daily, and when the tumor size reached approximately 80 mm3, the mice were treated daily with intraperitoneal (i.p.) injections of 4, 8, or 12 mg/kg monensin, dissolved in DMSO/HBCD 10% in 0.9% NaCl (1:9, v/v) or with vehicle alone (DMSO/HBCD 10% in 0.9% NaCl–1:9, v/v). Groups of five (treatment with 4 or 12 mg/kg monensin) or fifteen animals (treatment with 8 mg/kg Momensin or vehicle) were employed. Tumors were measured in two dimensions using an analog caliper to estimate the tumor volume using the formula V = (L × W × W)/2.
Treatments were administered for 10 days, and mice were sacrificed 2 h after the last administration or upon signs of distress.
2.8. In Vivo Imaging Analysis of Tumor Development and Intratumoral Na+ Content
Bioluminescence imaging using the IVIS® Spectrum was employed to monitor the distribution of 4T1-Luc2-Luc2 tumor cells and the development of the tumor. The IVISbrite™ D-luciferin ultra bioluminescent substrate in RediJect™ solution (RediJect D-luciferin ultra K+ salt) was injected i.p. following the manufacturer’s guidelines (150 mg/kg). The bioluminescence signal, derived from living luciferase-expressing cells, directly represented the tumor mass, whereas a decrease in bioluminescence signals corresponded to necrotic areas. The fluorescent cytosolic Na+ indicator CoroNa™ Green-AM was used to assess the Na+ concentrations in tumor and healthy tissues. Just prior to imaging acquisition, mice were injected with a solution (100 μL) containing the intracellular Na+-fluorescent dye (67 μM) in 0.9% NaCl with BSA (4%) and pluronic acid (0.02%). Intracellular Na+ variations following monensin treatment were monitored using the IVIS® Spectrum in vivo imaging system set to excitation at 500 nm and emission at 540 nm.
2.9. Histology and Immunohistochemistry
Mouse tissues were routinely embedded in paraffin and fixed in 10% neutral buffered formalin. Bone marrow samples were decalcified for 8 h using an EDTA-based decalcifying solution, washed in running water for 1 h, and subsequently embedded in paraffin. Four-micron-thick sections were deparaffinized, rehydrated, and stained with hematoxylin and eosin for histopathologic analyses or with anti-Ki-67 (1:250, Ventana® Medical Systems, Roche, Monza, Italy) to evaluate cell proliferation by using an automated immunostainer (Ventana, Roche, Monza, Italy). Unstained or corresponding hematoxylin and eosin-stained sections were used as controls of anti-Ki-67-stained samples. Slide images were captured using the ZEISS Axioscan 7 slide scanner (objective: Plan-Apochromat 40x/0.95 corr M27; camera: ZEISS Axiocam 705 color). Ki-67-positive cells, cell density (cells/mm2), and cell size (EQPC: equivalent diameter of a circle with the same projection area as the cell) were quantified using QuPath’s built-in “Positive cell detection” and “Cell detection” tools (software version QuPath-0.5.1).
2.10. Statistics and Reproducibility
All experiments were independently repeated at least three times. The results were presented as the mean of three to eight independent experiments ± standard deviation. Statistical significance between two groups was determined by ordinary two-way ANOVA with Sidak’s multiple comparisons test. Data analysis was performed using GraphPad Prism 8.4.3 (GraphPad Software, Inc., La Jolla, CA, USA).
4. Discussion
The present study investigated the anticancer effects of monensin treatment in cellular and mouse TNBC models, focusing on the ability of this ionophore to induce a specific and cytotoxic increase in intracellular Na+ within cancerous cells. Using live cell imaging analysis, we showed that monensin induced an early increase in intracellular Na+, which preceded TNBC cell death, and that maintaining cells in Na+-free medium effectively blocked Na+ upregulation, hindering monensin-induced cytotoxicity. Employing both human and mice TNBC cell types, we evidenced that the cytotoxic action of monensin was preceded by sodium-dependent energy depletion. We finally found that systemic treatment with monensin in BALB/c mice bearing orthotopic TNBC allografts led to significant TNBC tumor shrinkage and induced a further and selective increase in intracellular Na+ content within cancer tissues, without affecting the integrity and sodium content of healthy organs.
In this research we employed vivo imaging methodologies [
28] to simultaneously and non-invasively monitor in mice TNBC development and cellular sodium variations of transformed and healthy cells, using luciferase-targeted TNBC cells along with bioluminescent and fluorescent dyes that needed intracellular processing for visualization. Using this approach, we addressed the limitations of available techniques [
21,
22,
23,
24,
25], which cannot discriminate between intra- and extra-cellular Na
+ that passively enters necrotic cells and demonstrated that the Na
+ increase precedes cancer cell killing, excluding epiphenomena due to cancer tissue necrosis.
The results obtained were consistent with our in vitro observations in TNBC and HCC cells [
14], marking the first in vivo demonstration that monensin increased the Na
+ content of TNBC cells and that such upregulation was associated with a reduction in TNBC expansion. Our in vivo analysis was also in good agreement with findings from 23Na MRI studies on human breast cancer [
20], confirming that TNBC cells displayed higher basal intracellular Na
+ concentration ([Na
+]i) compared with untransformed cells and indicating [Na
+]i as a distinct and actionable target characteristic of cancer cells. Notably, we showed that monensin treatment selectively enhanced Na
+ levels in TNBC cells without affecting healthy tissues, suggesting that unlike healthy cells, cancerous TNBC cells were unable to compensate for the monensin-driven Na
+ load, rendering them selectively vulnerable to its cytotoxic effects.
Consistent with prior studies involving a different strain of mice (i.e., NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ mice) and tumor type (HCC) [
14], our histological analysis revealed that monensin treatment in Balb-c TNBC mice did not induce morphological damage in vital organs, such as the liver, lung, spleen, kidney, heart, and brain. By contrast, monensin-treated TNBC tumors displayed extensive areas of necrosis and pronounced swelling of tumor cells. This observation aligned with our previous findings on primary hepatocyte death showing that mitochondrial dysfunction led to an irreversible increase in intracellular Na
+ that resulted in an abrupt increase in cellular volume with lysis and death of the cells [
11,
13]. Similarly, in HCC cells [
14], we demonstrated that monensin triggered a Na
+-dependent increase in intracellular water retention (k
io) and cell enlargement within the tumors [
14]. Supporting these observations, MRI and NMR relaxometry studies on human tumors [
29,
30,
31] have also reported significantly enhanced k
io in cancer cells, indicative of a heightened intracellular osmotic pressure during neoplastic development.
Our findings in HCC and TNBC models, along with the evidence of elevated Na
+ content [
21,
22,
23,
24,
25] and increased intracellular water retention in human cancers [
29,
30,
31], support the hypothesis that treatment with sodium ionophores may exacerbate these conditions, thereby inducing selective TNBC cell killing. This assumption indicated that the anti-tumor effect of monensin was primarily due to a specific cytotoxic action on tumor tissues rather than a cytostatic effect on rapidly proliferating tissues. Indeed, our previous report using an HCC allograft model provided evidence that monensin did not affect proliferative activity in either transformed or healthy tissues [
14]. Consistently, the present study further confirmed in the TNBC mice model the lack of cytostatic action of monensin in both tumor and normal proliferating tissues, such as BM and intestinal mucosa. These observations are particularly significant as they support the hypothesis that monensin exerts its anti-tumor effect in TNBC without relying on a cytostatic mechanism. They are, however, in contrast with the several in vitro studies that, employing different human or murine cancer cell types, had previously linked the anti-cancer activity of monensin to its inhibitory effects on pro-proliferative pathways [
15,
16,
17,
18,
19].
To investigate this question, we examined the effects of monensin on the expression of the proliferative marker PCNA in 4T1-Luc2 cells, comparing them with those observed in murine HCC cells (Hepa-C1C7), human ovarian carcinoma cells (SKOV-3), and murine melanoma cells (B16), with or without 10% FBS during incubation. The absence of serum, as used in our previous study on the cytotoxic and sodium-dependent activity of monensin in Hepa-C1C7 cells [
14], was chosen to better reflect in vivo conditions where cells of solid tumors, such as breast cancer, HCC, ovarian carcinoma, and melanoma were not directly in contact with serum and, more specifically, were not exposed to the components of fetal bovine serum (FBS) [
27]. Conversely, monensin treatment in the presence of FBS replicated the experimental conditions described in the articles reporting the cytostatic action of monensin [
15,
16,
17,
18,
19].
Our results revealed significant differences depending on the conditions applied. Treatment with monensin in the absence of FBS did not alter PCNA levels in HEPA-C1C7, SKOV-3, B16, or 4T1-Luc2 cells. In contrast, the same treatment in the presence of serum reduced PCNA levels in all cell types except 4T1-Luc2 cells. These data were consistent with previous reports highlighting the ability of this ionophore to inhibit pro-proliferative pathways, including EGF-R, SUMO, and Wnt/β-catenin signaling [
15,
16,
17,
18,
19]. It is, therefore, conceivable that monensin can produce a cytostatic effect in vitro only when cell proliferation is maintained by auxiliary exogenous pro-proliferative stimuli, such as those present in fetal bovine serum [
27]. In the absence of these stimuli, monensin does not appear capable of influencing the intrinsic proliferative capacity of Hepa-C1C7, SKOV-3, or B16 cells.
Regarding TNBC 4T1-Luc2 cells, monensin did not affect PCNA expression even in the presence of serum. This observation appeared in contrast with a previous report of an antiproliferative effect of monensin in MCF-7 and MCF-10A breast cancer cells [
25], where the cytostatic activity was evaluated as decreased cell colony formation. Such an effect, however, can be also visualized in our 4T1-Luc2 cell model (
Figure 2d) and is likely to represent the reflection of a reduced cell density due to the killing activity of the ionophore.
Several factors can be instead involved in the different or delayed responsiveness of the other cell types to the effects of monensin on PCNA expression in the presence of FBS. Cancer cells in the absence of FBS undergo proliferation inhibition and apoptosis with differential timing depending on the cell-specific resistance to this condition [
27]. Similarly, monensin might need dissimilar dosages to exert its cytotoxic action in different cancer cells (i.e., HCC cells [
14] vs. TNBC cells). Thus, in the presence of FBS, each cell lineage might require differential concentration or time of treatment with monensin to manifest the effects on PCNA expression, and this might explain the observed difference (
Figure 3).
The possible FBS components involved in this process are at the moment unclear. Dedicated methodological analysis would be necessary not only for their identification but also to establish alternative culture conditions able to ensure the long-term survival of cultured cells also in the absence of FBS. Our findings indicate, in fact, that the presence of FBS might represent a possible confounding factor when comparing in vitro and in vivo results related to proliferative processes.
Standard chemotherapy relays on a general cytostatic effect and chemotherapy treatments for TNBC patients inevitably interfere with physiological signaling pathways, often inhibiting necessary cell turnover in normal proliferating tissues and causing side effects that exacerbate the morbidities in TNBC patients [
32]. These include a wide spectrum of adverse reactions during and after treatment, which, as reported by meta-analysis studies involving 4474 participants [
33], results in serious symptoms, such as diarrhea, nausea, vomiting, hair loss, fatigue, weight loss, pain, and a decline in immune defenses [
32,
33].
The present study showed that monensin achieved its antitumor efficacy in mice TNBC through a sodium-dependent and cancer-specific cytotoxic mechanism, without altering the structural and functional integrity of healthy non-proliferating and proliferating tissues.
These observations point to the potential role of monensin as a therapeutic alternative to standard cytostatic treatments, without the detrimental side effects associated with their employment. Investigations with additional models and larger experimental studies will be, however, needed to further validate and more extensively analyze the anticancer effect of monensin against TNBC.
In this study we used immuno-competent mice and analyzed TNBC tumors derived from mice TNBC cells, to more closely represent physiological conditions. This model, however, could not differentiate the eventual and additional effects of monensin on the anti-cancer immune-inflammatory reactions. To overcome this limitation, it will be useful, in future research, to analyze the anticancer activity of monensin also in immuno-compromised mice models of TNBC. These models will also consent to evaluating in vivo the effects of monensin against TNBC developed from human TNBC cells such those used in vitro in the present study (
Table 1) or in previous studies [
19], thus increasing the clinical relevance of the findings.
The present study focused its investigation on a mice model of primary TNBC. Future research employing metastatic models of TNBC will be fundamental to extend the analysis of the anticancer activity of monensin to processes of main relevance for the treatment of the advanced stages of TNBC such as epithelial mesenchymal transition and migration TNBC cells and eventually metastasis formation.
Although mice studies have by now excluded adverse effects, three reported cases of severe human rhabdomyolysis after monensin ingestion [
34,
35], the variability of the toxic effects of monensin in different species [
36], and its previously described cytostatic effects [
15,
16,
17,
18,
19] might have, by now, prevented the direct employment of monensin in clinics.
Our findings on the specific anticancer activity of monensin unrelated to antiproliferative effects and the numerous previous reports on its potent anticancer activity [
15,
16,
17,
18,
19] represent, however, a solid scientific base for further investigations aimed at overcoming all potential translation barriers. Such investigations, possibly including the development of novel and more active monensin analogs, will help to identify the lower dosages for a possible safe employment of sodium-ionophore compounds in the therapy of TNBC and of cancer in general.