Abstract
Breast cancer remains one of the leading cancers among women. Cancer stem cells (CSCs) are tumor-initiating cells which drive progression, metastasis, and reoccurrence of the disease. CSCs are resistant to conventional chemo- and radio-therapies and their ability to survive such treatment enables tumor reestablishment. Metastasis is the main cause of mortality in women with breast cancer, thus advances in treatment will depend on therapeutic strategies targeting CSCs. Salinomycin (SAL) is a naturally occurring polyether ionophore antibiotic known for its anticancer activity towards several types of tumor cells. In the present work, a library of 17 C1-single and C1/C20-double modified SAL analogs was screened to identify compounds with improved activity against breast CSCs. Six single- and two double-modified analogs were more potent (IC50 range of 1.1 ± 0.1 to 1.4 ± 0.2 μM) toward the breast cancer cell line MDA-MB-231 compared to SAL (IC50 of 4.9 ± 1.6 μM). Double-modified compound 17 was found to be more efficacious than SAL against the majority of cancer cell lines in the NCI-60 Human Tumor Cell Line Panel. Compound 17 was more potent than SAL in inhibiting cell migration and cell renewal properties of MDA-MB-231 cells, as well as inducing selective loss of the CD44+/CD24−/low stem-cell-like subpopulation in both monolayer (2D) and organoid (3D) culture. The present findings highlight the therapeutic potential of SAL analogs towards breast CSCs and identify select compounds that merit further study and clinical development.
Graphical abstract

1. Introduction
Breast cancer remains one of the leading cancers among women, with an estimated 2.1 million cases worldwide in 2018 [1]. Approximately 90% of breast cancer deaths are due to metastasis and the consequent impaired functioning of vital organs (e.g., lung, liver, brain), regardless of whether the cancer was metastatic at diagnosis or developed later [2][3]. Despite the widespread availability of screening mammography for early detection, the rate of metastatic breast cancer at initial diagnosis in the U.S. has remained unchanged since 1975 [4]. Despite recent progresses in understanding mechanisms leading to breast cancer recurrence, there is no known cure for metastatic breast cancer.
Recently, it has become apparent that tumors are a heterogeneous population of cell types that in many instances display a distinct cellular hierarchy. At the apex of this hierarchy are tumor-initiating cancer stem cells (CSCs) that represent a subpopulation of cells with the dual capacity of self-renewal and differentiation [5][6][7][8]. They drive initiation, progression, metastasis, and tumor reoccurrence [9][2][3]. CSCs exhibit resistance to conventional chemo- and radio-therapies and their ability to survive treatment facilitates tumor reestablishment [10]. Therefore, therapeutic strategies targeting CSCs hold great potential for novel advances in cancer treatment [11].
With more than 120 structures reported to date, polyether ionophores represent a class of natural products displaying a broad spectrum of pharmacological activities, including cytotoxic activity against various types of cancer cells [12][13][14]. Of particular interest is salinomycin (SAL, 1, Figure 1), a carboxyl ionophore with selective activity toward multidrug resistant (MDR) cancer cells and CSCs [15]. Gupta and co-workers [16] used high-throughput screening of about 16,000 bioactive molecules to show that SAL markedly and selectively reduced the viability of breast CSCs, with a potency nearly 100-times that of the conventional drug paclitaxel [16]. In addition, SAL has attracted attention as a selective inhibitor of CSCs from other cancer types, including leukemia, colorectal cancer, osteosarcoma, and uveal melanoma [17][18][19][20]. Mechanistically, many stemness-associated transcription factors, such as β-catenin, c-Myc, Snail, SOX2, and Twist1, were found to be critical for SAL-mediated CSC elimination [16][17][21][22][23][24][25][26]. Changes in phenotype composition after SAL treatment [27] appears to be due to modulation of the Hedgehog, K-Ras, and Wnt signaling pathways [18][26][28]. In the recent study by Mai et al. SAL, and it’s analog – ironmycin (AM 5), were shown to selectively target breast CSCs through accumulating and sequestering iron in lysosomes in vitro and in vivo [24]. In response to cytoplasmic iron deficiency, the SAL-treated cells induced the degradation of ferritin in the lysosomes, resulting in further iron loading in these organelles [24]. Iron-mediated generation of reactive oxygen species induced the permeability of the lysosomal membrane, triggering ferroptosis [24]. This is important, since iron endocytosis mediated by CD44 is enhanced during epithelial-mesenchymal transition, making mesenchymal cells more vulnerable to ferroptotic cell death, such as induced by SAL [5].
Figure 1.
Synthesis of salinomycin analogs
It is important to note that in limited clinical trials SAL was well tolerated by cancer patients without any long-term acute adverse effects when used at doses of 200–250 μg kg−1 [25].
The chemical modification of the SAL biomolecule through the rational design and synthesis of semisynthetic derivatives has been explored in an effort to obtain compounds with superior therapeutic properties. A series of SAL analogs has been synthesized to date via synthetic manipulation of either the C1 carboxyl [29][30][31][32][33] or the C20 hydroxyl of SAL [24][34][35][36][37][38][39] which in turn has led to the synthesis of a library of C1/C20 doubly modified products [40][41][42][43][44]. Among these SAL analogs, we identified select C1 amides and esters, especially compound 4 (Figure 1), as effective anticancer agents against drug-sensitive and drug-resistant cancer cells of various origins, and demonstrated their ability to overcome MDR [31][32][33][44][45][46]. Furthermore, we have recently identified select derivatives of SAL or C20-oxosalinomycin (19) (compounds 15–18, Figure 1) as lead structures for further antibacterial and antiparasitic drug development [41][43]. Interestingly, compounds that exhibit significant activity towards bacteria or parasites are often also active on cancer cells [32][47][48]. While there have been reports of C20 singly modified SAL analogs as selective agents towards CSCs [24][34][49][50], there has not been a comprehensive study to examine the ability of a large selection of C1-single and C1/C20-double modified analogs to target breast CSCs. A library of 17 C1-single and C1/C20-double modified SAL analogs was screened against breast cancer stem-like cells using both 2D and 3D models and several compounds have been identified that merit further study and clinical development.
2. Materials and methods
2.1. Compounds
Salinomycin (SAL) derivatives were synthesized as shown in Figure 1 and characterized by Huczyński and co-workers as reported previously [31][32][41] [43] [51][52]. SAL conjugates with hydroxamic acids (compounds 15–16) were resynthesized following the protocol published by Wu and co-workers [29], while C20-oxosalinomycin (compound 19) was generated by the chemoselective oxidation of the C20 allylic hydroxyl of SAL, as previously reported by Rodriguez and co-workers [24]. The NMR data of all semisynthetic analogs of SAL were in good agreement with those found in the reference literature [24][29][31][32][41][43][51][52].
2.2. Cells and culturing conditions
Human breast adenocarcinoma cell line MDA-MB-231, human primary acute lymphoblastic leukemia ALL-5 cells [53], and human breast epithelial cell line MCF 10A, were employed in the study. MDA-MB-231 cells were cultured in DMEM/Ham’s Nutrient Mixture F12 1:1 (cat. no. 10-090-CV, Corning, Manassas, VA, USA) supplemented with 5% (v/v) heat-inactivated fetal bovine serum (FBS) (cat. no. FP-0500-A, Atlas Biologicals, Fort Collins, CO, USA) and 1% penicillin/streptomycin solution 100x (cat. no. 30-002-Cl, Corning, Manassas, VA, USA). Primary ALL-5 cells were derived from the bone marrow of a 37-year old patient and can be cultured for up to 6 months retaining their original properties, as previously described [53][54]. They were cultured in IMDM Modified (HyClone) media supplemented with 10 μg ml−1 cholesterol, 6 mg ml−1 human serum albumin, 2 mM L-glutamine, 2% v/v amphotericin-B/penicillin/streptomycin, 1 μg ml−1 insulin, 200 μg ml−1 apo-transferrin, and 50 μM β-mercaptoethanol, and were subcultured to maintain a density of 1–3 x 106 cells ml−1. MCF 10A cell line was cultured in Mammary Epithelial Cell Basal Medium (cat. no. C-21215, Promo Cell, Heidelberg, Germany) supplemented with Mammary Epithelial Cell Supplement Pack (cat. no. C-39110, Promo Cell, Heidelberg, Germany), 100 μg ml−1 gentamicin (cat. no. G1397, Sigma-Aldrich, Deisenhofen, Germany) and 0.05 μg ml−1 amphotericin B (cat. no. A2942, Sigma-Aldrich, Deisenhofen, Germany). All cells were routinely maintained at 37°C in a humidified 5% CO2 incubator. Mycoplasma testing and cell validation were performed via short tandem repeat profiling in February 2020 by Genetica DNA Laboratories (Burlington, NC, USA). Cell lines were reported to be negative for mycoplasma contamination and verified as authentic, giving a 100% match when compared to the known reference profile [55].
2.3. Cell viability assay
In order to assess cell viability, a 3-(4,5-dimethylthiazol-2-yl)-2,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)-based assay was employed [56][57]. Compounds were dissolved in cell culture grade DMSO (cat. no. BP231-1, Fisher). MDA-MB-231 and MCF 10A cells lines were seeded in 96-well plates (TPP, Trasadingen, Switzerland) at a density of 2 x 103 or 2 x 104 cells/well, respectively, in 100 μl of corresponding complete medium. After 24 h of incubation cells were treated with compounds at concentrations up to 10 μM for 72 h (MDA-MB-231) or 96 h (MCF 10A) with control cells receiving vehicle (0.1% DMSO) alone. After treatment, 10 μl of MTT solution (5 mg ml−1) was added to each well, and the plate was incubated at 37°C for 4 h in a humidified 5% CO2 incubator. Medium was then aspirated and 150 μl of DMSO was added to each well and the plate agitated on a shaking platform (150 rpm) for 10 min. Absorbance was read at 540 nm using a BioTek Plate Reader. Inhibition of formation of colored MTT formazan was taken as an index of cytotoxicity activity. ALL-5 cells were seeded at a density of 105 cells/well in a 96-well plate and treated with different concentrations of compounds or vehicle (0.1% DMSO) for 120 h. Notably, ALL cells have a relatively long doubling time of ~60 h [53] and the assay was designed to encompass two doubling times for untreated control cells. At the end of the incubation, 10 μl of MTT solution (5 mg ml−1) was added to each well, and the plate was incubated at 37°C for 24 h in a humidified 5% CO2 incubator. Then 100 μl of 10% SDS in 0.01 M HCl was added to each well and the plate was incubated at 37°C for a further 24 h. Absorbance was recorded at 540 nm using a BioTek Plate Reader. IC50 values were determined by non-linear regression analysis using GraphPad Prism 6 for Windows (GraphPad Software).
2.4. NCI-60 human tumor cell line screen
Anticancer screening assay was performed by the National Cancer Institute (NCI) employing previously published methodology of Rubinstein and co-workers [58]. Tumor cell lines were cultured in RPMI 1640 medium containing 5% fetal bovine serum and 2 mM L-glutamine. After 24 h of culture, two microtiter plates of each cell line were fixed in situ with trichloroacetic acid (TCA) to obtain readings at time-zero (Tz) prior to compound addition. Additional plates of cells were treated with compounds for 48 h and after sulforhodamine B staining absorbance was read at 515 nm to determine control growth in the presence of vehicle (C) and test growth in the presence of compound at the different concentrations (Ti). The percentage growth was calculated for every compound concentration according to the following equations: [(Ti-Tz)/(C-Tz)] x 100 for concentrations for which Ti≥Tz, or [(Ti-Tz)/Tz] x 100 for concentrations for which Ti<Tz. GI50 is defined as the drug concentration resulting in a 50% decrease in the net absorbance increase during 48 h growth (C-Tz), and LC50 as the drug concentration that reduces Tz, the initial absorbance, by 50%, reflecting net loss, i.e. death, of cells.
2.5. Cell cycle analysis by flow cytometry
MDA-MB-231 cells (2 x 105/well for compound treatment and 105/well for DMSO treatment) were seeded in 6-well plates (TPP, Trasadingen, Switzerland) and allowed to attach for 48 h in a 37°C humidified 5% CO2 incubator. The medium was replaced and cells were treated with 0.1% DMSO or SAL (compound 1), or compounds 2, 15, 16, 17 or 18, at concentrations equal to 5 x IC50 values (Table 1), for 24, 48 or 72 h. Cells were then harvested, washed with Dulbecco’s phosphate-buffer saline (DPBS) (cat. no. 21-030-CM, Corning, Manassas, VA, USA), fixed with 3 ml of 70% ice-cold ethanol and stored at 4°C prior to flow cytometric analysis. Cells were centrifuged, treated with 400 μl propidium iodide/RNase Staining buffer (BD Biosciences, San Jose, CA, USA) and stored in the dark for 1 h at room temperature (RT). DNA content was quantitated with a FacsAria IIIu Flow Cytometer (BD Biosciences, San Jose, CA, USA) and data analysed using FlowJo software.
Table 1.
Antiproliferative activity (IC50) values of salinomycin (1), and its analogs.
| Compound | MDA-MB-231 | ALL-5 | MCF 10A |
|---|---|---|---|
|
|
|
|
|
| IC50 (μM) | IC50 (μM) | IC50 (μM) | |
| SAL (1) | 4.9 ± 1.6& | 5.8 ± 1.7 | 0.1 ± 0.02 |
| 2 | 2.7 ± 0.1** | 1.5 ± 0.5 [46] | 2.9 ± 0.1 |
| 3 | 3.9 ± 0.4 | 1.9 ± 0.7 [46] | 3.1 ± 0.1 |
| 4 | 3.6 ± 0.5 | 1.2 ± 0.02 [46] | 2.3 ± 0.4 |
| 5 | 2.7 ± 0.1** | 2.9 ± 0.7 [46] | 1.2 ± 0.2 |
| 6 | > 10 | 5.7 ± 0.5 | 2.0 ± 0.4 |
| 7 | > 10 | 9.4 ± 0.3* | > 10 |
| 8 | 2.6 ± 0.5** | 1.7 ± 0.5 [46] | 0.9 ± 0.3 |
| 9 | 1.9 ± 0.5*** | 4.3 ± 3.1 [46] | 0.2 ± 0.01 |
| 10 | > 10 | 3.6 ± 0.4 | > 10 |
| 11 | > 10 | 1.8 ± 0.9* | > 10 |
| 12 | > 10 | 9.7 ± 0.3* | 8.7 ± 5.0 |
| 13 | > 10 | 9.6 ± 3.0 | 3.7 ± 0.8 |
| 14 | > 10 | 2.3 ± 0.4* | 0.4 ± 0.1 |
| 15 | 1.7 ± 0.2*** | 1.1 ± 0.2* | 0.2 ± 0.05 |
| 16 | 1.4 ± 0.1*** | 1.5 ± 0.2* | 0.2 ± 0.05 |
| 17 | 1.1 ± 0.2*** | 4.8 ± 0.3 | 0.2 ± 0.1 |
| 18 | 2.8 ± 0.2** | 1.0 ± 0.1* | 0.2 ± 0.1 |
The value is an average from two independent experiments (n=4 each).
IC50 value is defined as the concentration of the compound which induces 50% growth inhibition. Values are given as mean ± SD (n = 4)
p ≤ 0.05
p ≤ 0.01
p ≤ 0.001.
2.6. Colony formation assay
MDA-MB-231 cells were seeded in 6-well plates (1.5 x 105/well) and allowed to attach for 48 h in a 37°C humidified 5% CO2 incubator. After the medium was replaced, cells were treated with 0.1% DMSO or SAL (compound 1) (2.5 μM), 2 (1.4 μM), or 17 (0.5 μM) (concentrations equal to ½ IC50 values) (Table 1), for 6, 24, 48 or 72 h. Cells were harvested, re-seeded at the density of 250 cells/well (n=6) in 6-well plates in compound-free medium, and allowed to proliferate for 13 days. The medium was removed, and cells were washed twice with 1 ml DPBS prior to staining with 0.5% crystal violet in methanol for 40 min at RT with agitation. Colonies of at least 25 cells were counted manually and graphed as percent relative to the vehicle treated cells.
2.7. Wound healing assay
MDA-MB-231 cells were seeded in 60 mm petri dishes (3 x 105 cells/dish) and allowed to attach in a 37°C humidified 5% CO2 incubator for 72 h resulting in a confluent monolayer of cells. After removing the medium, cells were washed with DPBS and incubated overnight in the absence of FBS. Next, the medium was removed, and cross shaped scratches (wound areas) were made in the cell monolayer with a sterile 1250 μl pipette tip. After washing twice with DPBS, cells were incubated in medium containing 0.1% DMSO or SAL (compound 1) (2.5 μM), 2 (1.4 μM) or 17 (0.5 μM) at concentrations equal to respective ½ IC50 values (Table 1). The scratch area was photographed by the inverted phase contrast microscopy with an EVOS FL Auto Cell Imaging System (Thermo Fisher Scientific) directly (time 0), as well as after 2, 4, 24, 26 and 48 h. Seven measurements for each condition and time point were taken and mean ± SD was calculated. The wound area at each time point was divided with the area at 0 h to obtain the measure of wound closure. The wound area was defined as 100% at 0 h for each treatment condition.
2.8. Organoids generation and culturing
Droplets of a 1:4 mixture of complete Mammocult medium (cat. no. 05620, Stemcell Technologies) and Matrigel (cat. no. 354277, Corning), containing 104 MDA-MB-231 cells/droplet, were formed at 0°C by pipetting 20 μl of the cold mixture onto a sheet of Parafilm. Droplets were solidified for 1 h in a 37°C humidified 5% CO2 incubator, and subsequently grown in complete Mammocult medium without agitation for four days. Half of the volume of fresh complete Mammocult medium was added every other day. On the fourth day, organoids were transferred to the shaking platform at 90 rpm (Orbi-Shaker Jr, Benchmark Scientific, USA). In order to propagate and maintain the organoid cultures, when organoids reached approximately 3 mm diameter they were minced into approximately 0.5–1 mm diameter pieces using Excelta scissors (cat. no. 17-467-493, 17-456-004, 17-467-497, Fisher Scientific) to avoid necrotic cell death in the inner core [59].
2.9. Cell surface markers identified by flow cytometry
MDA-MB-231 cells were seeded in 6-well plates (0.5 x 105/well) and allowed to attach for 48 h in a 37°C humidified 5% CO2 incubator. After the medium had been replaced, cells were treated with 0.1% DMSO or SAL (compound 1) (2.5 μM), 2 (1.4 μM) or 17 (0.5 μM) at concentrations equal to respective ½ IC50 values (Table 1) for 48 h. For the organoid treatment, one dissected organoid was incubated in 1 ml of complete Mammocult medium in 24-well plates and treated with vehicle or test compound as described above for 48 h. After incubation cells were harvested with enzyme-free cell dissociation buffer (cat. no. 13151014, Gibco), and organoids with enzyme-free cell dissociation buffer containing 0.2% of anti-clumping agent, 2-naphtol-6,8-disulfonic acid dipotassium salt (NDA) (cat. no. 439013, Frontier Scientific) and dispersed by gentle pipetting with a 25 ml tip. Cells were then washed with flow cytometry buffer, and re-suspended in flow cytometry buffer containing FITC conjugated anti-CD24 (cat. no. ab30350, Abcam) 1:100 and PE conjugated anti-CD44 (cat. no. ab46793, Abcam) 1:200, and incubated for 30 min in the dark. Cells were washed twice with flow cytometry buffer and re-suspended in 300 μl propidium iodide/RNase Staining buffer and subjected to a FacsAria IIIu Flow Cytometer. Data were analysed using FlowJo software. The threshold lines were set according to the isotype control. The accuracy of the triple immunostaining was confirmed by the comparison with single immunostaining for CD44, CD24 and PI respectively.
2.10. Combination assay
In order to determine whether the activity of compound 17 was attributed to the molecule itself, or to the possible synergistic effect between its constituent components, C20-oxosalinomycin (19) and benzhydroxamic acid potentially released during enzymatic hydrolysis, MDA-MB-231 cells were treated with 19 at ¼, ½, 1, 2 or 4 x IC50 concentrations in the absence or presence of benzhydroxamic acid at the following range of concentrations: 35.5 μM, 71 μM, 142 μM, 284 μM or 568 μM (where IC50 value is 142 μM). The experiment was performed in triplicate and standard MTT viability assays were conducted as described above. Results were evaluated quantitatively by the median effect principle of Chou-Talalay [60]. This method uses the IC50 values for individual compounds and the IC50 for their combination to calculate a Combination Index, CI, where CI = D1/Dx1 + D2/Dx2; and where Dx1 = IC50 of compound 1, Dx2 = IC50 of compound 2, D1 = IC50 of compound 1 in the presence of compound 2 at a concentration of D2, D2 = concentration of compound 2.
2.11. Immunoblot analysis
MDA-MB-231 cells (2.2 x 106 cells/100 mm petri dish) were treated for 24 or 48 h with compound 1, 2 or 17 at 5 x IC50 values concentration (see Table 1 for respective IC50 values) or with vehicle (0.1% DMSO) in the absence or presence of 100 μM Z-VAD-FMK (cat. No. A1902, APExBIO). Cells were washed with PBS, released with 0.05% trypsin (cat. no. 25-052-CI, Corning), centrifuged and washed with PBS. Cell pellets were snap frozen in liquid nitrogen and stored in −80°C until processing. MDA-MB-231 organoids were generated as described in 2.8. Organoids generation and culturing. Two cut organoids/well were cultured in 24-well plates and treated for 24 or 48 h with compound 1, 2 or 17 at 5 x IC50 values concentration or with vehicle (0.1% v/v DMSO). Organoids were washed with PBS, incubated with enzyme-free cell dissociation buffer with 0.2% (v/v) NDA and dispersed by gentle pipetting with a 1250 μl tip. Cells released from organoids were washed and frozen as described above. Cells were further thawed and lysed in lysis buffer (25 mM HEPES, pH 7.5, 300 mM NaCl, 0.1% w/v Triton X-100, 1.5 mM MgCl2, 0.2 mM EDTA, 0.5 mM DTT, EDTA-free complete protease inhibitor tablets (Roche), 20 μg ml−1 aprotinin, 50 μg ml−1 leupeptin, 10 μM pepstatin, 1 mM phenylmethylsulfonyl fluoride, 20 mM β-glycerophosphate, 1 mM Na3VO4, and 1 μM okadaic acid). Protein content was measured by Bradford assay and equal amounts (20 μg) were separated by electrophoresis using 12% (w/v) acrylamide Mini-PROTEAN® precast gels (Bio-Rad). Proteins were electrophoretically transferred onto a PVDF membrane (Immobilon-FL, Merck Millipore) and next stained with Ponceau S to assess transfer efficiency and verify equal loading. The membrane was blocked with 5% (w/v) non-fat milk in Tris-buffered saline containing 0.1% (w/v) TWEEN-20 (TBS-T) for 1 h at RT and incubated overnight at 4°C with primary antibodies against PARP (cat. no. 9532, Cell Signaling Technology) (1:1000 dilution), Bcl-2 (cat. no. sc-509, Santa Cruz Biotechnology) (1:200 dilution), Mcl-1 (cat. no. sc-12756, Santa Cruz Biotechnology) (1:200 dilution), Bak (cat no. 12105, Cell Signaling Technology) (1:1000 dilution), LC3B (cat. no. 2775, Cell Signaling Technology) (1:1000 dilution), and GAPDH (cat. no. 2118, Cell Signaling Technology) (1:10000 dilution). After washing with TBS-T for 5 x 5 min the membrane was incubated with secondary HRP-conjugated goat anti-rabbit IgG (H+L) antibody (1:5000 dilution) (cat. no. 170-6515, Bio-Rad) or goat anti-mouse (cat. no. 1706516, Bio-Rad) for 1 h at RT. After washing in TBS-T the membrane was exposed to ClarityTM Western ECL Substrate luminol enhancer solution and peroxide solution (Bio-Rad) for 5 min and visualized and quantified using Image J software. Uncropped immunoblots are shown in Supplemental Figures S1-S3.
2.12. qRT-PCR analysis
MDA-MB-231 cells (0.7–2.2 x 106 cells/100mm petri dish) were treated for 24 h with compound 1, or 17 at ½ IC50 values concentration (see Table 1 for respective IC50 values) or with vehicle (0.1% v/v DMSO). Cells were washed in PBS, released with 0.05% trypsin (cat. no. 25-052-CI, Corning), spun down and washed with PBS. Cell pellets were snap frozen in liquid nitrogen and stored in −80°C until processing. Total RNA was isolated with RNeasy Mini Kit (cat no. 74104, Qiagen). Genomic DNA removal and reverse transcription ware performed with iScript gDNA Clear cDNA Synthesis Kit (cat no. 1725035, Rio-Rad). The level of specific transcripts was quantitated by qPCR using TaqMan assays: CDH1 (Hs01023895_m1), SNAIL-2 (Hs00161904_m1), Vimentin (Hs00418522_m1), Fibronectin (Hs01549976_m1), ZEB1 (Hs01566408_m1), 18s (Hs99999901_s1), GAPDH (Hs02786624_g1). The mRNA transcript levels were calculated relative to that of GAPDH (endogenous control) using the ΔΔCt method.
2.13. Statistical analysis
Unpaired t test with Welch's correction was performed for the significance and p values of <0.05 were considered significant. For the analysis of large data sets with multiple replicates (cell cycle analysis and colony formation assay) one-way ANOVA was employed. Data are presented as a mean ± SD. GraphPad Prism 9 for Windows (GraphPad Software) was employed for statistical analysis.
3. Results
3.1. Analog design and synthesis
The synthesis of salinomycin (SAL) analogs studied in this work, both C1 singly (compounds 2–9 and 15–16, Figure 1) and C1/C20 doubly modified products (compounds 10–14 and 17–18, Figure 1), is described thoroughly elsewhere [24][29][31][32][41][43][51][52]. Briefly, a library of SAL analogs (Figure 1) was devised based on the most bioactive compounds obtained in our previous studies; it included SAL C1 esters (compounds 2, 4–6, 8–9 and 15–16), C1 amides (compounds 3 and 7) as well as products that were obtained through the double modification of the SAL molecule, i.e. C1 esters/amides of SAL with the chemoselectively oxidized C20 hydroxyl to the corresponding ketone group (compounds 10–14 and 17–18). For better structure-activity relationship (SAR) studies, we selected for SAL derivatives that contained identical ester/amide moieties at the C1 position, such as propargyl ester 6 and amide 7, together with their C20-oxo counterparts (compounds 12–13). The spectroscopic data of all resynthesized SAL derivatives were in line with previously published data [24][29][31][32][41][43][51][52].
3.2. Activity of salinomycin and its analogs in cell viability screening
Most commonly, cancer stem cells (CSCs) are identified according to the expression of characteristic cell-surface markers, which vary in different subtypes of breast cancer. High expression of CD44 and low expression of CD24 (CD44+/CD24−/low) in breast cancer is associated with cell proliferation and tumorigenesis [61][62]. The human triple negative basal-like MDA-MB-231 cell line expressing CD44+/CD24−/low is one of the well-established in vitro models for studying breast CSCs [61][63].
It has been previously shown in JIMT-1 breast cancer cell line that SAL has the highest selective activity towards stem cells at ~IC25 [66]. For this reason, and to investigate changes in stem-like properties without inducing significant cell death, all of the subsequent stem cell assays has been performed in concentrations equal to ½ of IC50 (Table 1) of SAL and select analogs.
SAL and its ten single- and seven double-modified analogs were evaluated for their in vitro antiproliferative effect on primary ALL-5, human MDA-MB-231 and mammary epithelial MCF 10A cells. The data presented as IC50 ± SD have been shown in Table 1 and the associated viability curves in Supplemental Figure S4.
In our previous study we identified six single-modified esters and amides of SAL potent against primary ALL-5 cells [46]. Here we expanded the library of investigated compounds and determined two additional single- (15 and 16) (IC50 = 1.1 ± 0.2 – 1.5 ± 0.2 μM) and three double-modified (11, 14, and 18) (IC50 = 1.0 ± 0.1 – 2.3 ± 0.4 μM) analogs of SAL (1) (IC50 = 5.8 ± 1.7 μM) more active (indicated by lower IC50 values) than parent molecule towards those cells (Table 1).
As SAL was previously shown to be a potent inhibitor of breast CSCs, it was of interest to investigate the activity of its analogs against MDA-MB-231 cell line which, due to the high ratio of CD24low/CD44high and expression of ALDH+, is an established model for studying breast CSCs [61]. Since SAL was employed as a positive control, we recorded two separate IC50 values of 4.0 and 5.8 μM. In Table 1 we present the average from those measurements 4.9 ± 1.6 μM, which is in agreement with previously published data [29][67]. In the present work, we identified six single- (2, 5, 8, 9, 15, and 16) and two double-modified (17 and 18) analogs of SAL (1) more potent towards MDA-MB-231 cells than parent compound 1 (Table 1). The most active analogs towards both primary ALL-5 and MDA-MB-231 cells were conjugates of SAL with hydroxamic acids, making this modification particularly interesting for further studies.
The IC50 value obtained for the parental SAL towards the non-cancerous, immortalized MCF 10A cell line was 0.1 ± 0.02 μM and stood in agreement with previously published data (Table 1) [42][68]. The MCF 10A IC50 values recorded for single and double-modified SAL analogs were higher than for the unmodified scaffold and formed in the range of 0.2 ± 0.05 – 3.1 ± 0.1 μM for single- and 0.2 ± 0.1 – 8.7 ± 5.0 μM for double-modified compounds (Table 1).
3.3. In vitro growth inhibition against the panel of NCI-60 cancer cell lines
Taking into consideration the results from our preliminary cell viability assay (Table 1) and structural diversity, we identified seven analogs, 10, 11, 12, 13, 14, 17 and 18, for further screening in the panel of NCI-60 cancer cell lines (Supplemental Table S1). Compounds 10, 11, 12, 13, 14, 17 and 18 were tested at a single concentration of 10−5 M on the panel of 60 human tumor cell lines including leukemia, non-small cell lung, colon, central nervous system (CNS), melanoma, ovarian, renal, prostate, and breast cancer. Data are summarized in Supplemental Table S1 and represent percent of growth of the compound treated cells relative to the control, and relative to “time zero” cell count. The consequent growth percent is employed to calculate growth inhibition (values between 0 and 100) and lethality (values <0) (Supplemental Table S1). Compounds which exhibited at least 60% or more growth inhibitory property in at least eight cell lines during this single concentration screening were directed for the assessment against the same panel in 5-dose concentration range (for the details see 2.4. NCI-60 human tumor cell line screen).
After the initial screening two compounds, 13 and 17, were selected for further testing in the concentration range of 10−4 to 10−8 M (Supplemental Table S2). Dose-response curves were generated for each cell line by plotting cytotoxic activity against the log10 of the compound concentration. Cytotoxic effect was determined as GI50 value for each compound, which represents the concentration inducing 50% of growth inhibition. Due to its potent activity compound 17 was selected for the repeat of 5-dose testing (Supplemental Table S2). As presented in Table S2 compound 17 was more potent than parent SAL (1) towards the majority of cell lines included in the screening. This was especially true to panels of leukemia, colon, CNS, ovarian, renal and the entire panel of breast cancer (Supplemental Table S2).
3.4. Salinomycin and its analogs induce DNA fragmentation in the MDA-MB-231 cell line
In order to further examine the mechanism behind the promising activity of SAL and its analogs against the MDA-MB-231 cell line, we measured DNA content and DNA fragmentation via flow cytometry. Briefly, propidium iodide (PI) staining was employed to evaluate DNA content and cells with sub-G1 (<2N) DNA were scored as dead. MDA-MB-231 cells were treated with parent SAL (1), or its select and the most potent analogs (characterized by the lowest IC50 values, see Table 1), 2, 15, 16, 17 and 18, each at concentrations equal to 5 x IC50 values (Table 1) for 24, 48 or 72 h. MDA-MB-231 cells treated with 0.1% DMSO at an equivalent durations served as a control. The full set of representative cytograms is presented in Figure 2A. A graphical summary of cells in the different phases of the cell cycle as a mean from three independent experiments is shown in Figure 2B. Statistically significant increases (p ≤ 0.0001) in sub-G1 DNA content control vs. treatment were observed after the treatment with all of the compounds for 72 h. However, compounds 2, 17 and 18 induced DNA fragmentation as early as 24 h after treatment. Since the cytotoxic activity of parent SAL was tested in two independent cell viability experiments (see 3.2.), MDA-MB-231 cells were treated with two separate doses of at 20 and 30 μM (corresponding to 5 x IC50 values). Only 30 μM of SAL (1) was able to induce statistically significant sub-G1 DNA content after 24 h. Importantly, since the concentrations were adjusted to five times of the respective IC50 values, SAL analogs were able to induce a statistically significant amount of DNA fragmentation after 24 h in approximately two to five times lower doses than SAL (1). The findings from this experiment and the data from Table 1, prompted us to select compound 2 as the most promising representative of single-modified analogs and compound 17 as the best candidate from double-modified agents for further studies.
Figure 2.
Salinomycin analogs induced DNA fragmentation in MDA-MB-231 cells. MDA-MB-231 cells were treated with 0.1% DMSO (control), 20 μM (indicated by grey circle) or 30 μM (indicated by black circle) of salinomycin (1) or specified analogs at 5 x IC50 values for 24, 48 or 72 h and subjected to propidium iodide staining and flow cytometry. A. Representative cytograms; B. Distribution of cells observed in different phases of the cell cycle or with Sub-G1 DNA content determined by PI staining. Data represent mean ± SD (n = 3) *p ≤ 0.05, **p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
3.5. Salinomycin and its analogs reduce colony formation potential of MDA-MB-231 cell line
The colony formation capacity of MDA-MB-231 cells was initially found to be significantly reduced by the treatment with compounds 2 (55%) and 17 (55%) as compared with both control (100%) and parent SAL (1) (92%) after 24 h (Figure 3A and B). In order to measure the effect on colony formation without inducing cell death, the dose of each compound was adjusted to 50% of the respective IC50 value (see Table 1) and colony formation was assessed relative to vehicle-treated controls. Compounds 2 and 17 inhibited colony growth by ~50% after 24 h, but this was not seen for parent SAL (1). After 48 h of treatment SAL (1) dramatically inhibited colony growth by 84% (the same value was noted for compound 17), while compound 2 still had ~50% inhibitory effect (Figure 3A and B). 72 h of treatment lead to further decreases as follows: SAL (1) (90% inhibition); compound 17 (96% inhibition), and compound 2 (68% inhibition) (Figure 3A and B). Of note, the slight decrease in the number of colonies formed under control conditions after 72 h vs. earlier treatments may be related to the increased cell confluence and decreased amount of nutrients upon prolonged culturing. Based on these results compound 17 was shown to act more rapidly and was more potent than SAL or 2.
Figure 3.
Effect of salinomycin (1) and analogs 2 and 17 on colony formation potential of MDA-MB-231 cells. MDA-MB-231 cells were treated with 0.1% DMSO (control), 1 (2.5 μM), 2 (1.4 μM) or 17 (0.5 μM) for 6, 24, 48 and 72 h. At the end of the incubation, cells were harvested and re-seeded in 6-well plates at 250 cells/well. After 13 days, cells were fixed and stained with 0.5% crystal violet methanol solution. Colonies, defined by group of more than 50 cells, were counted manually for each well and normalized to the control. A. Representative images of colonies formed; B. Colonies quantification. Data represent mean values ± SD (n = 6). *p ≤ 0.05, **p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
3.6. Salinomycin and its analogs attenuate MDA-MB-231 cell migration
A wound healing assay was performed to assess the effect of our selected compounds on the migration of MDA-MB-231 cells. The wound was created on a confluent cell monolayer and cells were allowed to migrate for 48 h in the absence or presence of the compound of interest (Figure 4A). Compound 17 significantly reduced the migration of MDA-MB-231 cells as early as after 24 h (83% vs. 26% of wound surface area treatment vs. control respectively) (Figure 4B). Statistically significant inhibition of migratory properties of MDA-MB-231 cells was observed for treatment with SAL (1) and its two analogs: compounds 2 and 17 after 48 h, with compound 17 showing the biggest inhibitory effect (80% vs. 0% of wound surface area treatment vs. control, respectively) (Figure 4B). Due to the differences in IC50 values (Table 1), it should be noted that compound 17 was used in this assay at a five-fold lower concentration then SAL (1). Taken together, these data suggest a superior anti-migratory effect of compound 17 over the parental SAL (1).
Figure 4.
Wound healing assay of MDA-MB-231 cells after treatment with 0.1% DMSO (control), 2.5 μM 1, 1.4 μM 2 or 0.5 μM 17 for 48 h. Wounds were created on a confluent cell monolayer and images were acquired at 0, 4, 24, and 48 h after wounding. The experiment was performed in duplicate. A. Representative images of wound healing; B. Quantification of wound healing. Data represents mean ± SD (n = 2), *p ≤ 0.05.
3.7. Salinomycin analogs reduce the CD44+/CD24− stem-like population in MDA-MB-231 cell monolayer and organoid cultures
We subsequently investigated the effect of treatment with SAL (1) and compounds 2 and 17 on CD44+/CD24−/low stem cell-like populations within the MDA-MB-231 cell line (Figure 5A). The quantification of CD44+/CD24−/low populations relative to the control is shown in Figure 5B. All of the compounds induced small, but significant reductions in the percentage of viable CD44+/CD24−/low cells, with a corresponding increase in number of cells with the CD44+/CD24+ phenotype (Figure 5A). Of these, compound 17 had a 2-fold greater effect than parent SAL (1) in reducing the stem cell-like population but at 1/5th the concentration.
Figure 5.
Analysis of the expression of CD44high/CD24low stem-like populations in MDA-MB-231 cells and organoids. A. MDA-MB-231 cells were triple stained with anti-CD44-PE, anti-CD24-FITC and propidium iodide (PI) and viable PI negative cells are shown in the representative cytograms with CD24 on y axis and CD44 on x axis; B. The ratio of CD44high/CD24low MDA-MB-231 cells after treatment with 0.1% DMSO (control), 2.5 μM 1, 1.4 μM 2 or 0.5 μM 17 for 48 h. Bar graph was prepared from the percentage of viable CD44high/CD24low MDA-MB-231 cells in the flow cytometry analysis. Data represents means ± SD (n = 3), **p ≤ 0.01, *** p ≤ 0.001; C. Schematic representation of organoids acquisition and treatment. Scale bar, 2 mm; D. Organoids dispersed to single-cells were triple stained with anti-CD44-PE, anti-CD24-FITC and PI and viable PI negative cells are shown in the representative cytograms with CD24 on y axis and CD44 on x axis; E. The ratio of CD44high/CD24low organoid cells after treatment with 0.1% DMSO (control), 2.5 μM 1, or 0.5 μM 17 for 48 h. Bar graph was prepared from the percentage of viable CD44high/CD24low organoid cells in the flow cytometry. Data represents means ± SD (n =3), *p ≤ 0.05,*** p ≤ 0.001.
In order to overcome the limitations associated with utilization of cell monolayers and to more appropriately model the 3-dimensional (3D) nature of tumors [69], we employed organoid culture. The culturing of organoids and treatment scheme is shown in Figure 5C (see also 2.8. Organoids generation and culturing). Representative cytograms of CD24/CD44 surface marker distribution of untreated (control), SAL (1) and compound 17 treated organoids are shown in Figure 5D. As evidenced in Figure 5E, which represents quantification of the percentage of viable CD44+/CD24−/low cells from treated organoids, both SAL (1) and compound 17 induced a modest, but significant reduction in the percentage of CD44+/CD24−/low cells. Similarly, as in 2D culture treatment (Figure 5A), an increase in the amount of cells with CD44+/CD24+ phenotype can be observed (Figure 5D). We conclude that compound 17 was superior over SAL in reducing CD44+/CD24−/low stem-like sub-population of MDA-MB-231 cells in monolayer culture and retained this activity in 3D organoid culture.
3.8. C20-oxosalinomycin shows antagonistic effect in combination with benzhydroxamic acid
In order to determine whether the activity of compound 17 was attributed to the molecule itself or the two individual synthetic precursors released during possible enzymatic hydrolysis in vitro, we assessed IC50 values in cell viability assay for C20-oxosalinomycin (19) and benzhydroxamic acid. The IC50 values found were 12.1 and 142.0 μM respectively, which corresponds to 11 and 129 X the IC50 value of compound 17 (Table 2), and stays in agreement with our previous finding [42]. To investigate the possibility of a synergistic effect between those two compounds leading to the decreased IC50 value seen with compound 17, we performed a Chou-Talalay combination assay. The results are shown in Table 2, and as previously described for a pair of drugs, CI > 1.3 indicates antagonism, CI = 1.1–1.3 indicates moderate antagonism, CI = 0.9 to 1.1 indicates an additive effect, CI = 0.8 to 0.9 indicates slight synergism, CI = 0.6 to 0.8 indicates moderate synergism, CI = 0.4 to 0.6 indicates synergism, and CI = 0.2 to 0.4 indicates strong synergism [60][70][46][71]. We found both compounds to have antagonistic effect, resulting in the increase of IC50 value (Table 2), therefore proving that the activity of compound 17 is attributed to the molecule itself rather than the unbounded components.
Table 2.
Combination Index (CI) and IC50 values of C20-oxosalinomycin (19) in combination treatment with benzhydroxamic acid against MDA-MB-231 cells.
| Compound | Benzhydroxamic acid, μM | IC50, μM | CI | Effect |
|---|---|---|---|---|
| 19 | - | 12.1 | - | - |
| 19 | 35.5 | 18.3 | 1.7 | Antagonism |
| 19 | 71.0 | 14.8 | 1.5 | Antagonism |
| 19 | 142.0 | 16.0 | 1.9 | Antagonism |
| 19 | 284.0 | 17.3 | 2.7 | Antagonism |
3.9. Salinomycin and its analogs induce features of apoptotic cell death in MDA-MB-231 cells and organoids
To examine the apoptotic effect of compounds 2 and 17, as well as the parental SAL (1), on MDA-MB-231 cells in adherent monolayer or in 3D organoid cultures we investigated the levels of PARP, Bak, Mcl-1 and Bcl-2 via immunoblotting. With the exception of analog 2, all of the compounds induced significant decrease of total PARP in cell monolayer after 48 h treatment as compared to the untreated control (Figure 6A and C). Interestingly, in organoid culture this effect was observed as early as after 24 h, and loss of total PARP further increased after 48 h in SAL (1) and compound 17 treatment (Figure 6B and D). The level of anti-apoptotic protein Bcl-2 was slightly reduced in MDA-MB-231 cells after 48 h of treatment with all of the compounds studied as compared to the control (Figure 6E). Similarly, in the organoid culture significant Bcl-2 degradation was observed after 48 h treatment with all of the compounds, with more robust effect after 24 h of incubation with SAL (1) and analog 17 (Figure 6F). Examination of another anti-apoptotic protein Mcl-1 in MDA-MB-231 monolayer revealed a significant increase in its level after 48h, as compared to the untreated control, with an even more robust increase induced by analog 17 (Figure 6I). A contrasting effect was observed in 3D organoid culture, where Mcl-1 protein was significantly downregulated after 24 h treatment with all of the compounds, to further decrease after 48 h (except analog 2) (Figure 6J). Investigation of the expression of pro-apoptotic protein Bak revealed its significant increase vs. control after treatment of MDA-MB-231 cells with analog 17 after 24 h and 48 h as well as a very modest, but statistically significant, increase in Bak with SAL (1) (Figure 6G). In organoid culture, treatment with all of the compounds significantly increased expression of Bak after 24 h and 48 h (Figure 6H).
Fig. 6.
A-B. Effect of compounds 1, 2 or 17 on PARP, Mcl-1, Bcl-2 and Bak expression. MDA-MB-231 cells or organoids were treated with 0.1% DMSO (control), or 25 μM 1, 14 μM 2, or 5 μM 17 for the times indicated, and extracts were prepared and subjected to immunoblotting for PARP, Mcl-1, Bcl-2 and Bak. GAPDH was employed as a loading control; .C-J Bar diagrams showing the fold changes of proteins normalized to GADPH. Images were quantified by measuring the band intensity using ImageJ software. Data represent mean ± S.D (n = 3). *p ≤ 0.05, **p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
In order to determine whether loss of Bcl-2 expression was the event leading to cell death, or its consequence, MDA-MB-231 cells were incubated with SAL (1) or analog 17 in the absence or presence of caspase-3 inhibitor – Z-VAD. Z-VAD was previously internally verified to be a caspase-3 inhibitor (M. Delgado, R.R. Rainwater, A. Urbaniak, K. Butler, M. Davidson, R.M. Protacio, B. Heflin, G. Baldini, A. Edwards, K.D. Raney, and T.C. Chambers, Microtubule depolymerization in primary acute lymphoblastic leukemia cells induces death in G1 and M phases which occurs through distinct cell death pathways, manuscript in preparation). Treatment with SAL (1) or its analog 17 in the presence of Z-VAD significantly inhibited loss of Bcl-2 protein as compared to incubation with compounds alone and untreated control (Figure 7A and C). Significant upregulation of Bcl-2 expression in cells incubated with Z-VAD alone vs. untreated controls suggests a decrease of physiologically occurring cell death, due to the inhibition of caspase-3 (Figure 7C). PARP was used as a control, and as expected, loss of its expression was protected by Z-VAD (Figure 7A and B).
Figure 7.
A. Inhibition of the degradation of PARP and Bcl-2 in the presence of caspase-3 inhibitor (Z-VAD). MDA-MB-231 cells were treated with 0.1% DMSO (control), 25 μM 1, or 5 μM 17 in the absence or presence of 100 μM Z-VAD for 48 h, and extracts were prepared and subjected to immunoblotting for PARP or Bcl-2. GAPDH was employed as a loading control; B. Bar diagram showing the fold changes of proteins normalized to GADPH. Images were quantified by measuring the band intensity using ImageJ software. Data represented as mean ± S.D. of three independent determinations (n = 3). **p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
We also examined the effect of the treatment with SAL (1), and its analogs, compounds 2 and 17 on the expression of autophagy pathway associated marker LC3 in monolayer and organoid cultures. There was a significant upregulation of LC3-II (14 kDa) expression after incubation of cell monolayers with SAL (1) and compound 17 for 24 h as compared to the untreated control, with further significant increase after 48 h of treatment (Figure 8A and C). Similarly treatment of organoids for 24 h significantly induced the expression of LC3-II (14 kDa) as compared to the control (Figure 8B and D). By contrast, compound 2 only induced a very modest increase in LC3-II in adherent and organoid culture.
Figure 8.
A. Effect of compounds on LC3B expression in MDA-MB-231 cells and organoids. Cells and organoids were treated with control
, 25 μM 1
, 14 μM 2
, or 5 μM 17
for the times indicated. Extracts were prepared and subjected to immunoblotting for LC3B (14 kDa). GAPDH was employed as a loading control; B. Bar diagram showing the fold changes of proteins normalized to GADPH. Images were quantified by measuring the band intensity using ImageJ software. Data represented as mean ± S.D. of three independent determinations (n = 3). *p ≤ 0.05, **p ≤ 0.01.
Taken together, we conclude that both SAL (1) and compound 17 induced apoptosis in a caspase-3 dependent manner, as well as upregulation of the expression of autophagy pathway associated marker LC3II, in both monolayer and 3D organoid culture.
3.10. Treatment with salinomycin and its analogs increased the expression of mesenchymal markers and decreased the expression of E-cadherin
We next examined the effect of SAL (1) and compound 17 on epithelial to mesenchymal transition (EMT) associated markers (E-cadherin, Fibronectin, Vimentin, Snail-2 and Zeb-1) in cell monolayer. Compared to untreated controls, E-cadherin was downregulated by SAL (1) and no significant changes in the expression of this gene were observed after treatment with compound 17 (Figure 9A). No statistically significant changes in control vs. treatment level of Fibronectin were observed (Figure 9B). Vimentin was upregulated by both SAL (1) and compound 17, with a bigger effect of the latter (Figure 9C). Zeb-1 was upregulated only by compound 17 (Figure 9D) while Snail-2 was upregulated only by SAL (1) (Figure 9E). We conclude that, other than a common upregulation of vimentin, treatment with SAL (1) and compound 17 had differential effects on EMT marker gene expression.
Figure 9.
Effect of compounds on EMT markers in MDA-MB-231 cell line. (A–E) Effect of 24 h treatment with salinomycin (1) (2.5 μM), or 17 (0.5 μM) on E-cadherin, Fibronectin, Vimentin, Zeb-1 and Snail-1 mRNA abundance. Bar graphs represented as mean ± S.D. of three independent determinations (n = 3). *p ≤ 0.05, **p ≤ 0.01.
4. Discussion
We and others demonstrated that the modification of the C1 carboxyl alone or in combination with derivatization of the C20 hydroxyl may improve the biological activity of the resulting semisynthetic derivatives of salinomycin (SAL) [29][32][41][42][43][44][45][46][72]. For instance, SAL C1 esters and amides, especially 3 and 4, were highly selective agents in vitro and met the activity criteria for hit compounds for parasites that are responsible for African trypanosomiasis in humans (sleeping sickness) and animals (nagana disease) [72]. Recently, conjugates of SAL and C20-oxosalinomycin (19) with benz- and salicylhydroxamic acid (compounds 15–18) were found to exhibit excellent inhibitory activity against planktonic Gram(+) bacteria and methicillin-resistant staphylococci, but also towards biofilm-forming bacterial strains [41]. As far as activity against cancer cells, certain C1 singly and C1/C20 doubly modified analogs of SAL were shown to possess selective anticancer effects towards a panel of human cancer cell lines, and the potential to overcome MDR at micromolar concentrations [29][32][42][44][45][46]. However, the ability of this class of compounds to target CSCs remained unclear.
In the current study, we investigated the anti-breast cancer stem-like cell activity of select C1 singly and C1/C20 doubly modified SAL analogs using several independent CSC assays. Cancer cell renewal and migration have a great impact on tumor metastasis. SAL was previously shown to affect self-renewal of MDA-MB-231 [73] and HCC1937 [63] breast cancer cell lines. Additionally, SAL inhibited cell migration in MDA-MB-231 [74] [73] [75], MDA-MB-436 [76], JIMT-1 [50] and MCF-7 [74] breast cancer cell lines. Herein, we have found a double-modified analog of SAL, compound 17 to be more potent than the parent SAL molecule in colony formation and wound healing assays, by inducing significant inhibition of self-renewal and migration faster (as early as 24 h) at only one fifth of the concentration of SAL (Figures 3 and 4).
Current models for studying CSCs typically include using mice in vivo which has inherent issues because adaptation to the mouse can alter the fundamental properties of human CSCs. The use of 3D tumor organoids derived from human cancer cells has provided a robust alternative experimental system to study CSC function and responses to drug treatment [69]. Organoids are stem-cell derived structures that self-organize and as such more closely resemble the tissue or tumor of origin [69]. Additionally, they offer advantages for pre-in vivo testing over mammosphere assays, including maintenance of cell-cell interactions, heterogeneity, microenvironment, morphology, histopathology, receptor status and drug response of the tumor sample they originated from [69][77][78]. Therefore, we employed a breast cancer organoid model to validate anti-breast cancer stem-like activity of our experimental compounds.
SAL was previously shown to reduce the CD44+/CD24−/low stem-like populations in breast cancer cells including HMLER [16][24] and JIMT-1 [66], as well as in MDA-MB-231 mammospheres [75]. Here we report that SAL (1) and its analog 17 significantly reduced the CD44+/CD24−/low stem-like population in both adherent (monolayer) and 3D organoids derived from MDA-MB-231 cells (Figure 5). Indeed, analog 17 was considerably more potent then parent SAL (1) in this regard. The compensatory increase in the non-stem CD44+/CD24+ population may be explained by recent findings of Kamlund and co-workers, where treatment of JIMT-1 cells with 0.5 μM of SAL resulted in selective inhibition of the CD24− subpopulation, consequently increasing the CD24+ population [79].
We were concerned whether the effects of compound 17 was a consequence of possible metabolic breakdown after enzymatic hydrolysis in vitro to its two synthetic precursors: 19 and benzhydroxamic acid. The Chau Talalay method was employed and it was found that the two compounds used simultaneously act in an antagonistic fashion, leading to the conclusion that the activity of analog 17 is attributed to this novel scaffold itself and not its breakdown products (Table 2).
Evasion of apoptosis is one of the major features by which a cancer cell population expands [80]. SAL was shown to induce an apoptotic cascade in various types of cancers including breast, hepatic, lung, prostate, leukemia and ovarian [81]. We therefore investigated whether SAL and its analogs 2 and 17 induced apoptosis in MDA-MB-231 cells and organoids by analyzing changes in apoptosis-related proteins such as PARP, Bcl-2, Mcl-1 and Bak (Figure 6). Our study shown that SAL (1) and its analogs not only down-regulated the expression of anti-apoptotic Bcl-2 and Mcl-1 (in organoid) and enhanced the expression of pro-apoptotic Bak but also increased the degradation of PARP in both 2D and 3D culture, which is consistent with induction of apoptosis. Interestingly, contrasting changes in Mcl-1 levels occurred between 2D and 3D cultures as a consequence of compound treatment. The increase of Mcl-1 in cell monolayers after treatment with SAL and its analogs may indicate the induction of initial defense mechanisms protecting cells from the toxic effects of these compounds. Such a protective effect is not evident in the 3D organoid model.
Autophagy is a self-degradable catabolic mechanism related to the breakdown of unnecessary or dysfunctional components [82]. It is also considered a survival mechanism to counteract stressors such as starvation, endoplasmic reticulum (ER) stress, hypoxia or infection [83]. Autophagy may participate in cell death when apoptotic processes are compromised or when cells are subjected to severe stressors [84]. Several studies have highlighted the role of autophagy in chemoresistant tumors, and provided an insight into the crosstalk between apoptosis and autophagy [85][86]. During starvation, cancer cells undergo autophagy, which may delay activation of the apoptotic pathway [87]. Despite its importance in cancer therapy, molecular pathways leading to autophagy are not well understood and precise therapeutic strategies to control autophagy have not yet been developed. SAL was previously shown to induce activation of autophagy in human cancer cells [88][89]. Therefore, it was of interest to investigate the status of the autophagy-associated marker LC3II after treatment of MDA-MB-231 cells and organoids with SAL (1) and its analogs, and this was found to be upregulated after treatment with SAL (1) and compound 17 in 2D and 3D cultures (Figure 8).
Breast CSCs have been shown to possess increased metastatic capacity that correlates with an ongoing EMT to promote cell migration [90]. SAL has been previously shown to upregulate epithelial marker E-cadherin in the MDA-MB-231 cell line [73] and downregulate various mesenchymal markers for example Fibronectin and Vimentin in MDA-MB-231 [73] and Vimentin in JIMT-1 cells [50]. We observed an inhibition of cell migration but an inconsistent effect on EMT marker gene expression (with some genes inhibited but others induced) after treatment with SAL and its analogs (Figures 4 and 9). These results could be explained by recent findings of Kamlund and co-workers, where treatment of breast cancer JIMT-1 cells with 0.5 μM of SAL resulted in increased cell motility, but at the same time it decreased average cell migration [79]. Since cell migration is related to the distance traveled by cells from a starting point, and motility reflects movement around starting point, therefore SAL treated cells appear to be “dancing” in circles in place to a greater degree than vehicle treated cells [79]. It should be noted however that in the JIMT-1 study [79] the authors observed an increase in E-cadherin (the opposite of our findings in MDA-MB-231 cells), which may reflect differences in the cell line used and/or the concentration of SAL employed.
Our results indicate that double-modified SAL analog 17 exhibited improved activity against breast cancer stem-like cells when compared to the native molecule using several well-accepted traits of CSCs. Although the single-modified analog 2 was less potent than SAL in CSCs assays, it still induced a significant effect as compared to the vehicle treatment at approximately half lower concentrations than the parent molecule. The differences in activity between double-modified analog 17 and single-modified compound 2 may be explained by an increased stability of molecule 17 achieved by the replacement of hydroxyl group by ketone in C20 position. This increase in stability translated to improved biological activity. Additionally, in the studies of Li et al. the conjugation with hydroxamic acid, such as we used with compound 17, was shown to increase membrane permeability, which corresponded with enhanced activity [29]. The same authors also pointed out to the ion-binding activity of some hydroxamic acids, which would lead to better biological properties [29].
Future studies will focus on the investigation of the metabolism and toxicity of these compounds in clinically relevant models, such as hepatic [91][92][93], cardiac [94][95] and kidney organoid [96] cultures. In addition, it will be important to address the selectivity of each compound on cancer vs. normal cells. To this end, an appropriate in vitro model which could be adapted for this study would be the HMLER (Human Mammary Epithelial Cells overexpressing hTERT, SV40 T/t and H-RasV12) cell line, which was also characterized by high CD44+/CD24−/low ratio and the ability to form non-adherent mammospheres [64]. The HMLER cell line (CD24low) and its isogenic control (CD24hlgh) was previously successfully used to study the mechanism of anti-breast cancer activity of SAL and its analogs [24][65].
5. Conclusions
In the present study we investigated anti-breast cancer stem-like cell activity of select single- and double-modified analogs of SAL in cell monolayer and organoid cultures. Several analogs were found to be more potent than the parent molecule towards breast cancer cell line MDA-MB-231. These compounds induced DNA fragmentation suggestive of apoptotic cell death. Further study revealed double-modified compound 17 to be more effective than SAL at inhibiting cell migration and renewal, as well as reducing thee CD44+/CD24− stem cell subpopulation in both monolayer and organoid cultures. Analog 17 was also found to be more potent than SAL in a majority of NCI-60 cancer cell lines, including the entire panel of breast tumor cell lines. Further investigation revealed that compound 17 induced features of apoptosis and upregulation of autophagy-associated marker LC3II in both cell monolayer and organoids. Our findings highlight the therapeutic potential of SAL scaffold analogs in the treatment of metastatic breast cancer and support further research towards clinical development of these compounds.
Supplementary Material
Highlights.
Select single- and double-modified salinomycin analogs were more potent towards MDA-MB-231 breast cancer cell line than salinomycin.
Double modified analog 17 induced DNA fragmentation and showed premises of apoptotic cell death in MDA-MB-231 cell monolayer and organoids.
Double modified analog 17 reduced colony formation potential and migration of MDA-MB-231 cells to the greater extent than salinomycin.
Double modified analog 17 more effectively than salinomycin reduced percentage of CD44+/CD24− breast cancer stem-like cells in MDA-MB-231 monolayer and organoid cultures.
Acknowledgements
This work was supported by a grant from the Arkansas Breast Cancer Research Program (to AMM, TCC, MCM and RLE), a Barton Pilot Grant from UAMS College of Medicine (to AU), a Team Science Award (to AMM and RLE) and NIH/R01 HD93461 (to AMM). We thank Ms. Andrea Harris from the UAMS Flow Cytometry Core and Mr. Eric R. Siegel from the Department of Biostatistics at UAMS. Testing was performed by the Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, http://dtp.cancer.gov. M.A. wishes to acknowledge the Polish Science Center (NCN) for financial support by a grant SONATA (2016/23/D/ST5/00242). M.A. also wishes to acknowledge the NCN and the Polish National Agency for Academic Exchange (NAWA) for the scholarships under the UWERTURA (2019/32/U/ST4/00092) and the BEKKER program (PPN/BEK/2019/1/00034), respectively, and the Polish Ministry of Science and Higher Education (MNiSW) for the scholarship for outstanding young scientists in the years 2020–2023 (STYP/15/1665/E-336/2020).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A, Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries., CA. Cancer J. Clin (2018). 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- [2].Jin X, Mu P, Targeting breast cancer metastasis, Breast Cancer Basic Clin. Res (2015). 10.4137/BCBCR.S25460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Gupta GP, Massagué J, Cancer Metastasis: Building a Framework, Cell. (2006). 10.1016/j.cell.2006.11.001. [DOI] [PubMed] [Google Scholar]
- [4].Narod SA, Iqbal J, Miller AB, Why have breast cancer mortality rates declined?, J. Cancer Policy (2015). 10.1016/j.jcpo.2015.03.002. [DOI] [Google Scholar]
- [5].Müller S, Sindikubwabo F, Cañeque T, Lafon A, Versini A, Lombard B, Loew D, Di Wu T, Ginestier C, Charafe-Jauffret E, Durand A, Vallot C, Baulande S, Servant N, Rodriguez R, CD44 regulates epigenetic plasticity by mediating iron endocytosis, Nat. Chem 12 (2020) 929–938. 10.1038/s41557-020-0513-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Jordan CT, Guzman ML, Noble M, Mechanism of Disease: Cancer Stem Cells, N. Engl. J. Med (2006). [DOI] [PubMed] [Google Scholar]
- [7].Clarke MF, Fuller M, Stem Cells and Cancer: Two Faces of Eve, Cell. (2006). 10.1016/j.cell.2006.03.011. [DOI] [PubMed] [Google Scholar]
- [8].Dalerba P, Cho RW, Clarke MF, Cancer stem cells: Models and concepts, Annu. Rev. Med (2007). 10.1146/annurev.med.58.062105.204854. [DOI] [PubMed] [Google Scholar]
- [9].International Agency for Research on Cancer, Latest global cancer data: Cancer burden rises to 18.1 million new cases and 9.6 million cancer deaths in 2018, Press Release. (2018).
- [10].Reya T, Morrison SJ, Clarke MF, Weissman IL, Stem cells, cancer, and cancer stem cells, Nature. (2001). 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
- [11].Chen K, Huang YH, Chen JL, Understanding and targeting cancer stem cells: Therapeutic implications and challenges, Acta Pharmacol. Sin (2013). 10.1038/aps.2013.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Liu H, Lin S, Jacobsen KM, Poulsen TB, Chemical Syntheses and Chemical Biology of Carboxyl Polyether Ionophores: Recent Highlights, Angew. Chemie - Int. Ed (2019). 10.1002/anie.201812982. [DOI] [PubMed] [Google Scholar]
- [13].Antoszczak M, A comprehensive review of salinomycin derivatives as potent anticancer and anti-CSCs agents, Eur. J. Med. Chem (2019). 10.1016/j.ejmech.2019.01.034. [DOI] [PubMed] [Google Scholar]
- [14].Versini A, Saier L, Sindikubwabo F, Müller S, Cañeque T, Rodriguez R, Chemical biology of salinomycin, Tetrahedron. (2018). 10.1016/j.tet.2018.07.028. [DOI] [Google Scholar]
- [15].Antoszczak M, A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent, Eur. J. Med. Chem (2019). 10.1016/j.ejmech.2018.12.057. [DOI] [PubMed] [Google Scholar]
- [16].Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, Lander ES, Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening, Cell. (2009). 10.1016/j.cell.2009.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Zhou J, Liu S, Wang Y, Dai W, Zou H, Wang S, Zhang J, Pan J, Salinomycin effectively eliminates cancer stem-like cells and obviates hepatic metastasis in uveal melanoma, Mol. Cancer (2019). 10.1186/s12943-019-1068-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Najumudeen AK, Jaiswal A, Lectez B, Oetken-Lindholm C, Guzmán C, Siljamäki E, Posada IMD, Lacey E, Aittokallio T, Abankwa D, Cancer stem cell drugs target K-ras signaling in a stemness context, Oncogene. (2016). 10.1038/onc.2016.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Zhang C, Tian Y, Song F, Fu C, Han B, Wang Y, Salinomycin inhibits the growth of colorectal carcinoma by targeting tumor stem cells, Oncol. Rep (2015). 10.3892/or.2015.4253. [DOI] [PubMed] [Google Scholar]
- [20].L Tang Q, Zhao ZQ, chun Li J, Liang Y, Yin JQ, Zou CY, Xie XB, Zeng YX, Shen JN, Kang T, Wang J, Salinomycin inhibits osteosarcoma by targeting its tumor stem cells, Cancer Lett. (2011). 10.1016/j.canlet.2011.07.016. [DOI] [PubMed] [Google Scholar]
- [21].Liu L, Wang Q, Mao J, Qin T, Sun Y, Yang J, Han Y, Li L, Li Q, Salinomycin suppresses cancer cell stemness and attenuates TGF-β-induced epithelial-mesenchymal transition of renal cell carcinoma cells, Chem. Biol. Interact (2018). 10.1016/j.cbi.2018.09.018. [DOI] [PubMed] [Google Scholar]
- [22].Lee HG, Shin SJ, Chung HW, Kwon SH, Do Cha S, Lee JE, Cho CH, Salinomycin reduces stemness and induces apoptosis on human ovarian cancer stem cell, J. Gynecol. Oncol (2017). 10.3802/JGO.2017.28.E14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Hamaï A, Cañeque T, Müller S, Mai TT, Hienzsch A, Ginestier C, Charafe-Jauffret E, Codogno P, Mehrpour M, Rodriguez R, An iron hand over cancer stem cells, Autophagy. (2017). 10.1080/15548627.2017.1327104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Mai TT, Hamaï A, Hienzsch A, Cañeque T, Müller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V, Ryo A, Ginestier C, Birnbaum D, Charafe-Jauffret E, Codogno P, Mehrpour M, Rodriguez R, Salinomycin kills cancer stem cells by sequestering iron in lysosomes, Nat. Chem (2017). 10.1038/nchem.2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Naujokat C, Steinhart R, Salinomycin as a drug for targeting human cancer stem cells, J. Biomed. Biotechnol (2012). 10.1155/2012/950658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Lu D, Choi MY, Yu J, Castro JE, Kipps TJ, Carson DA, Salinomycin inhibits wnt signaling and selectively induces apoptosis in chronic lymphocytic leukemia cells, Proc. Natl. Acad. Sci. U. S. A (2011). 10.1073/pnas.1110431108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Huang X, Borgströ2m B, Stegmayr J, Abassi Y, Kruszyk M, Leffler H, Persson L, Albinsson S, Massoumi R, Scheblykin IG, Hegardt C, Oredsson S, Strand D, The Molecular Basis for Inhibition of Stemlike Cancer Cells by Salinomycin, ACS Cent. Sci (2018). 10.1021/acscentsci.8b00257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Lu Y, Ma W, Mao J, Yu X, Hou Z, Fan S, Song B, Wang H, Li J, Kang L, Liu P, Liu Q, Li L, Salinomycin exerts anticancer effects on human breast carcinoma MCF-7 cancer stem cells via modulation of Hedgehog signaling, Chem. Biol. Interact (2015). 10.1016/j.cbi.2014.12.002. [DOI] [PubMed] [Google Scholar]
- [29].Li B, Wu J, Zhang W, Li Z, Chen G, Zhou Q, Wu S, Synthesis and biological activity of salinomycin-hydroxamic acid conjugates, Bioorganic Med. Chem. Lett (2017). 10.1016/j.bmcl.2017.01.080. [DOI] [PubMed] [Google Scholar]
- [30].Borgström B, Huang X, Chygorin E, Oredsson S, Strand D, Salinomycin Hydroxamic Acids: Synthesis, Structure, and Biological Activity of Polyether Ionophore Hybrids, ACS Med. Chem. Lett (2016). 10.1021/acsmedchemlett.6b00079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Antoszczak M, Maj E, Stefańska J, Wietrzyk J, Janczak J, Brzezinski B, Huczyński A, Synthesis, antiproliferative and antibacterial activity of new amides of salinomycin, Bioorganic Med. Chem. Lett (2014). 10.1016/j.bmcl.2014.02.042. [DOI] [PubMed] [Google Scholar]
- [32].Antoszczak M, Popiel K, Stefańska J, Wietrzyk J, Maj E, Janczak J, Michalska G, Brzezinski B, Huczyński A, Synthesis, cytotoxicity and antibacterial activity of new esters of polyether antibiotic - Salinomycin, Eur. J. Med. Chem (2014). 10.1016/j.ejmech.2014.02.031. [DOI] [PubMed] [Google Scholar]
- [33].Antoszczak M, Maj E, Napiórkowska A, Stefańska J, Augustynowicz-Kopeć E, Wietrzyk J, Janczak J, Brzezinski B, Huczyński A, Synthesis, anticancer and antibacterial activity of salinomycin N-benzyl amides, Molecules. (2014). 10.3390/molecules191219435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Versini A, Colombeau L, Hienzsch A, Gaillet C, Retailleau P, Debieu S, Müller S, Cañeque T, Rodriguez R, Salinomycin Derivatives Kill Breast Cancer Stem Cells by Lysosomal Iron Targeting, Chem. - A Eur. J (2020). 10.1002/chem.202000335. [DOI] [PubMed] [Google Scholar]
- [35].Li Y, Shi Q, Shao J, Yuan Y, Yang Z, Chen S, Zhou X, Wen S, Jiang ZX, Synthesis and biological evaluation of 20-epi-amino-20-deoxysalinomycin derivatives, Eur. J. Med. Chem (2018). 10.1016/j.ejmech.2018.02.004. [DOI] [PubMed] [Google Scholar]
- [36].Borgström B, Huang X, Hegardt C, Oredsson S, Strand D, Structure–Activity Relationships in Salinomycin: Cytotoxicity and Phenotype Selectivity of Semi-synthetic Derivatives, Chem. - A Eur. J (2017). 10.1002/chem.201603621. [DOI] [PubMed] [Google Scholar]
- [37].Zhang W, Wu J, Li B, Xia J, Wu H, Wang L, Hao J, Zhou Q, Wu S, Synthesis and biological activity evaluation of 20-: Epi -salinomycin and its 20- O -acyl derivatives, RSC Adv. (2016). 10.1039/c6ra08967d. [DOI] [Google Scholar]
- [38].Shi Q, Li Y, Bo S, Li X, Zhao P, Liu Q, Yang Z, Cong H, Deng H, Chen M, Chen S, Zhou X, Ding H, Jiang ZX, Discovery of a 19F MRI sensitive salinomycin derivative with high cytotoxicity towards cancer cells, Chem. Commun (2016). 10.1039/c6cc01508e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Borgström B, Huang X, Martin P, Hegardt C, Oredsson S, Strand D, Synthetic modification of salinomycin: Selective O-acylation and biological evaluation, Chem. Commun (2013). 10.1039/c3cc45983g. [DOI] [PubMed] [Google Scholar]
- [40].Czerwonka D, Barcelos Y, Steverding D, Cioch A, Huczyński A, Antoszczak M, Singly and doubly modified analogues of C20-epi-salinomycin: A new group of antiparasitic agents against Trypanosoma brucei, Eur. J. Med. Chem (2020). 10.1016/j.ejmech.2020.112900. [DOI] [PubMed] [Google Scholar]
- [41].Sulik M, Stępień K, Stefańska J, Huczyński A, Antoszczak M, Antibacterial activity of singly and doubly modified salinomycin derivatives, Bioorganic Med. Chem. Lett (2020). 10.1016/j.bmcl.2020.127062. [DOI] [PubMed] [Google Scholar]
- [42].Czerwonka D, Urbaniak A, Sobczak S, Piña-Oviedo S, Chambers TC, Antoszczak M, Huczyński A, Synthesis and Anticancer Activity of Tertiary Amides of Salinomycin and Their C20-oxo Analogues, ChemMedChem. (2020). 10.1002/cmdc.201900593. [DOI] [PubMed] [Google Scholar]
- [43].Antoszczak M, Steverding D, Sulik M, Janczak J, Huczyński A, Anti-trypanosomal activity of doubly modified salinomycin derivatives, Eur. J. Med. Chem (2019). 10.1016/j.ejmech.2019.03.061. [DOI] [PubMed] [Google Scholar]
- [44].Antoszczak M, Urbaniak A, Delgado M, Maj E, Borgström B, Wietrzyk J, Huczyński A, Yuan Y, Chambers TC, Strand D, Biological activity of doubly modified salinomycin analogs – Evaluation in vitro and ex vivo, Eur. J. Med. Chem (2018). 10.1016/j.ejmech.2018.07.021. [DOI] [PubMed] [Google Scholar]
- [45].Michalak M, Lach MS, Antoszczak M, Huczynski A, Suchorska WM, Overcoming resistance to platinum-based drugs in ovarian cancer by salinomycin and its derivatives—an in vitro study, Molecules. (2020). 10.3390/molecules25030537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Urbaniak A, Delgado M, Antoszczak M, Huczyński A, Chambers TC, Salinomycin derivatives exhibit activity against primary acute lymphoblastic leukemia (ALL) cells in vitro, Biomed. Pharmacother (2018). 10.1016/j.biopha.2018.01.081. [DOI] [PubMed] [Google Scholar]
- [47].Kapron B, Czarnomysy R, Paneth A, Wujec M, Bielawski K, Bielawska A, Swiatek L, Rajtar B, Polz-Dacewicz M, Plech T, Dual Antibacterial and Anticancer Activity of 4-Benzoyl-1-dichlorobenzoylthiosemicarbazide Derivatives, Anticancer. Agents Med. Chem (2018). 10.2174/1871520617666171023142958. [DOI] [PubMed] [Google Scholar]
- [48].Barrett SV, Barrett MP, Anti-sleeping sickness drugs and cancer chemotherapy, Parasitol. Today (2000). 10.1016/S0169-4758(99)01560-4. [DOI] [PubMed] [Google Scholar]
- [49].Klose J, Kattner S, Borgström B, Volz C, Schmidt T, Schneider M, Oredsson S, Strand D, Ulrich A, Semi-synthetic salinomycin analogs exert cytotoxic activity against human colorectal cancer stem cells, Biochem. Biophys. Res. Commun (2018). 10.1016/j.bbrc.2017.10.147. [DOI] [PubMed] [Google Scholar]
- [50].Huang X, Borgström B, Kempengren S, Persson L, Hegardt C, Strand D, Oredsson S, Breast cancer stem cell selectivity of synthetic nanomolar-active salinomycin analogs, BMC Cancer. (2016). 10.1186/s12885-016-2142-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Huczyński A, Janczak J, Antoszczak M, Stefańska J, Brzezinski B, X-ray, FT-IR, NMR and PM5 structural studies and antibacterial activity of unexpectedly stable salinomycin-benzotriazole intermediate ester, J. Mol. Struct (2012). 10.1016/j.molstruc.2012.05.019. [DOI] [Google Scholar]
- [52].Huczyński A, Janczak J, Stefańska J, Antoszczak M, Brzezinski B, Synthesis and antimicrobial activity of amide derivatives of polyether antibiotic - Salinomycin, Bioorganic Med. Chem. Lett (2012). 10.1016/j.bmcl.2012.05.081. [DOI] [PubMed] [Google Scholar]
- [53].Nijmeijer BA, Szuhai K, Goselink HM, van Schie MLJ, van der Burg M, de Jong D, Marijt EW, Ottmann OG, Willemze R, Falkenburg JHF, Long-term culture of primary human lymphoblastic leukemia cells in the absence of serum or hematopoietic growth factors, Exp. Hematol (2009). 10.1016/j.exphem.2008.11.002. [DOI] [PubMed] [Google Scholar]
- [54].Kothari A, Hittelman WN, Chambers TC, Cell cycle-dependent mechanisms underlie vincristine-induced death of primary acute lymphoblastic leukemia cells, Cancer Res. (2016). 10.1158/0008-5472.CAN-15-2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Robin T, Capes-Davis A, Bairoch A, CLASTR: The Cellosaurus STR similarity search tool - A precious help for cell line authentication, Int. J. Cancer (2020). 10.1002/ijc.32639. [DOI] [PubMed] [Google Scholar]
- [56].Alley MC, Scudiero DA, Monks A, Hursey M, Czerwinski MJ, Fine DL, Abbott BJ, Mayo JG, Shoemaker R, Boyd MR, Feasibility of Drug Screening with Panels of Human Tumor Cell Lines Using a Microculture Tetrazolium Assay, Cancer Res. (1988). [PubMed] [Google Scholar]
- [57].Skehan P, Storeng R, Scudiero D, Monks A, McMahon J, Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR, New colorimetric cytotoxicity assay for anticancer-drug screening., J. Natl. Cancer Inst 82 (1990) 1107–12. [DOI] [PubMed] [Google Scholar]
- [58].Rubinstein LV, Shoemaker RH, Pauli KD, Simon RM, Tosini S, Skehan P, Scudiero DA, Monks A, Boyd MR, Comparison of in vitro anticancer-drug-screening data generated with a tetrazolium assay versus a protein assay against a diverse panel of human tumor cell lines., J. Natl. Cancer Inst (1990). [DOI] [PubMed] [Google Scholar]
- [59].Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, Thokala R, Sheikh S, Saxena D, Prokop S, ao Liu D, Qian X, Petrov D, Lucas T, Chen HI, Dorsey JF, Christian KM, Binder ZA, Nasrallah M, Brem S, O’Rourke DM, li Ming G, Song H, A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity, Cell. (2020). 10.1016/j.cell.2019.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Chou TC, Drug combination studies and their synergy quantification using the chou-talalay method, Cancer Res. (2010). 10.1158/0008-5472.CAN-09-1947. [DOI] [PubMed] [Google Scholar]
- [61].Li W, Ma H, Zhang J, Zhu L, Wang C, Yang Y, Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis, Sci. Rep (2017). 10.1038/s41598-017-14364-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat-Nakshatri P, Turner CH, Goulet R, Badve S, Nakshatri H, CD44+/CD24-Breast cancer cells exhibit enhanced invase properties: An early step necessary for metastasis, Breast Cancer Res. (2006). 10.1186/bcr1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Kai M, Kanaya N, Wu SV, Mendez C, Nguyen D, Luu T, Chen S, Targeting breast cancer stem cells in triple-negative breast cancer using a combination of LBH589 and salinomycin, Breast Cancer Res. Treat (2015). 10.1007/s10549-015-3376-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Morel AP, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A, Generation of breast cancer stem cells through epithelial-mesenchymal transition, PLoS One. 3 (2008) 2888. 10.1371/journal.pone.0002888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Versini A, Colombeau L, Hienzsch A, Gaillet C, Retailleau P, Debieu S, Müller S, Cañeque T, Rodriguez R, Salinomycin Derivatives Kill Breast Cancer Stem Cells by Lysosomal Iron Targeting, Chem. - A Eur. J 26 (2020) 7416–7424. 10.1002/chem.202000335. [DOI] [PubMed] [Google Scholar]
- [66].Huang X, Borgström B, Månsson L, Persson L, Oredsson S, Hegardt C, Strand D, Semisynthesis of SY-1 for investigation of breast cancer stem cell selectivity of C-ring-modified salinomycin analogues, ACS Chem. Biol (2014). 10.1021/cb5002153. [DOI] [PubMed] [Google Scholar]
- [67].Czerwonka D, Urbaniak A, Sobczak S, Piña-Oviedo S, Chambers TC, Antoszczak M, Huczyński A, Synthesis and Anticancer Activity of Tertiary Amides of Salinomycin and Their C20-oxo Analogues, ChemMedChem. 15 (2020) 236–246. 10.1002/cmdc.201900593. [DOI] [PubMed] [Google Scholar]
- [68].Urbaniak A, Jousheghany F, Yuan Y, Piña-Oviedo S, Huczyński A, Delgado M, Kieber-Emmons T, Monzavi-Karbassi B, Chambers T, The response of phyllodes tumor of the breast to anticancer therapy: An in vitro and ex vivo study, Oncol. Lett (2019). 10.3892/ol.2019.10823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Drost J, Clevers H, Organoids in cancer research, Nat. Rev. Cancer (2018). 10.1038/s41568-018-0007-6. [DOI] [PubMed] [Google Scholar]
- [70].Ichite N, Chougule MB, Jackson T, Fulzele SV, Safe S, Singh M, Enhancement of docetaxel anticancer activity by a novel diindolylmethane compound in human non-small cell lung cancer, Clin. Cancer Res (2009). 10.1158/1078-0432.CCR-08-1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Alicja Urbaniak TCC, Jousheghany Fariba, Piña-Oviedo Sergio, Yuan Youzhong, Majcher-Uchańska Urszula, Klejborowska Greta, Moorjani Anika, Monzavi-Karbassi Behjatolah, Huczyński Adam, Carbamate derivatives of colchicine show potent activity towards primary acute lymphoblastic leukemia and primary breast cancer cells-in vitro and ex vivo study, J. Biochem. Mol. Toxicol (2020). 10.1002/jbt.22487. [DOI] [PubMed] [Google Scholar]
- [72].Steverding D, Antoszczak M, Huczyński A, In vitro activity of salinomycin and monensin derivatives against Trypanosoma brucei, Parasites and Vectors. (2016). 10.1186/s13071-016-1698-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Rai F, Suman S, Mishra S, Shukla Y, Evaluation of growth inhibitory response of Resveratrol and Salinomycin combinations against triple negative breast cancer cells, Biomed. Pharmacother (2017). 10.1016/j.biopha.2017.02.110. [DOI] [PubMed] [Google Scholar]
- [74].Dewangan J, Srivastava S, Mishra S, Divakar A, Kumar S, Rath SK, Salinomycin inhibits breast cancer progression via targeting HIF-1α/VEGF mediated tumor angiogenesis in vitro and in vivo, Biochem. Pharmacol (2019). 10.1016/j.bcp.2019.04.026. [DOI] [PubMed] [Google Scholar]
- [75].An F, Kim JY, Oh E, Lee N, Cho Y, Seo JH, Salinomycin promotes anoikis and decreases the CD44+/CD24− stem-like population via inhibition of STAT3 activation in MDA-MB-231 cells, PLoS One. (2015). 10.1371/journal.pone.0141919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Kopp F, Hermawan A, Oak PS, Herrmann A, Wagner E, Roidl A, Salinomycin treatment reduces metastatic tumor burden by hampering cancer cell migration, Mol. Cancer (2014). 10.1186/1476-4598-13-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Roelofs C, Hollande F, Redvers R, Anderson RL, Merino D, Breast tumour organoids: Promising models for the genomic and functional characterisation of breast cancer, Biochem. Soc. Trans (2019). 10.1042/BST20180375. [DOI] [PubMed] [Google Scholar]
- [78].Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H, A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity, Cell. (2018). 10.1016/j.cell.2017.11.010. [DOI] [PubMed] [Google Scholar]
- [79].Kamlund S, Janicke B, Aim K, Oredsson S, Salinomycin treatment specifically inhibits cell proliferation of cancer stem cells revealed by longitudinal single cell tracking in combination with fluorescence microscopy, Appl. Sci (2020). 10.3390/app10144732. [DOI] [Google Scholar]
- [80].Hanahan D, Weinberg RA, The hallmarks of cancer, Cell. (2000). 10.1016/S0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- [81].Dewangan J, Srivastava S, Rath SK, Salinomycin: A new paradigm in cancer therapy, Tumor Biol. (2017). 10.1177/1010428317695035. [DOI] [PubMed] [Google Scholar]
- [82].Yang Z, Klionsky DJ, Eaten alive: A history of macroautophagy, Nat. Cell Biol (2010). 10.1038/ncb0910-814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Glick D, Barth S, Macleod KF, Autophagy: Cellular and molecular mechanisms, J. Pathol (2010). 10.1002/path.2697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Kroemer G, Levine B, Autophagic cell death: The story of a misnomer, Nat. Rev. Mol. Cell Biol (2008). 10.1038/nrm2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Suman S, Das TP, Reddy R, Nyakeriga AM, Luevano JE, Konwar D, Pahari P, Damodaran C, The pro-apoptotic role of autophagy in breast cancer, Br. J. Cancer (2014). 10.1038/bjc.2014.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Young MM, Kester M, Wang HG, Sphingolipids: Regulators of crosstalk between apoptosis and autophagy, J. Lipid Res (2013). 10.1194/jlr.R031278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Jain MV, Paczulla AM, Klonisch T, Dimgba FN, Rao SB, Roberg K, Schweizer F, Lengerke C, Davoodpour P, Palicharla VR, Maddika S, Los M, Interconnections between apoptotic, autophagic and necrotic pathways: Implications for cancer therapy development, J. Cell. Mol. Med (2013). 10.1111/jcmm.12001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Jangamreddy JR, Ghavami S, Grabarek J, Kratz G, Wiechec E, Fredriksson BA, Pariti RKR, Cieślar-Pobuda A, Panigrahi S, Łos MJ, Salinomycin induces activation of autophagy, mitophagy and affects mitochondrial polarity: Differences between primary and cancer cells, Biochim. Biophys. Acta - Mol. Cell Res (2013). 10.1016/j.bbamcr.2013.04.011. [DOI] [PubMed] [Google Scholar]
- [89].Kim KY, Il Park K, Kim SH, Yu SN, Lee D, Kim YW, Noh KT, Ma JY, Seo YK, Ahn SC, Salinomycin induces reactive oxygen species and apoptosis in aggressive breast cancer cells as mediated with regulation of autophagy, Anticancer Res. (2017). 10.21873/anticanres.11507. [DOI] [PubMed] [Google Scholar]
- [90].Bozorgi A, Khazaei M, Khazaei MR, New findings on breast cancer stem cells: A review, J. Breast Cancer (2015). 10.4048/jbc.2015.18.4.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Meng Q, Three-dimensional culture of hepatocytes for prediction of drug-induced hepatotoxicity, Expert Opin. Drug Metab. Toxicol (2010). 10.1517/17425251003674356. [DOI] [PubMed] [Google Scholar]
- [92].Katsuda T, Kawamata M, Hagiwara K, Takahashi R. u., Yamamoto Y, Camargo FD, Ochiya T, Conversion of Terminally Committed Hepatocytes to Culturable Bipotent Progenitor Cells with Regenerative Capacity, Cell Stem Cell. (2017). 10.1016/j.stem.2016.10.007. [DOI] [PubMed] [Google Scholar]
- [93].Huch M, Gehart H, Van Boxtel R, Hamer K, Blokzijl F, Verstegen MMA, Ellis E, Van Wenum M, Fuchs SA, De Ligt J, Van De Wetering M, Sasaki N, Boers SJ, Kemperman H, De Jonge J, Ijzermans JNM, Nieuwenhuis EES, Hoekstra R, Strom S, Vries RRG, Van Der Laan LJW, Cuppen E, Clevers H, Long-term culture of genome-stable bipotent stem cells from adult human liver, Cell. (2015). 10.1016/j.cell.2014.11.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Eder A, Vollert I, Hansen A, Eschenhagen T, Human engineered heart tissue as a model system for drug testing, Adv. Drug Deliv. Rev (2016). 10.1016/j.addr.2015.05.010. [DOI] [PubMed] [Google Scholar]
- [95].Voges HK, Mills RJ, Elliott DA, Parton RG, Porrello ER, Hudson JE, Development of a human cardiac organoid injury model reveals innate regenerative potential, Dev. (2017). 10.1242/dev.143966. [DOI] [PubMed] [Google Scholar]
- [96].Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, Parton RG, Wolvetang EJ, Roost MS, De Sousa Lopes SMC, Little MH, Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis, Nature. (2015). 10.1038/nature15695. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.









