Introduction: Previously, we reported that interleukin-8 (IL-8) was associated with poor prognosis of basal-like breast cancer patients and has been identified as a pro-tumorigenic factor, facilitating cell invasion and migration. Here, we investigated the pharmacological impact of inhibitors targeting the chemokine receptors, C-X-C chemokine receptor 1 (CXCR1) and C-X-C chemokine receptor 2 (CXCR2), which are activated by IL-8. Methods: The survival rates of triple-negative breast cancer (TNBC) patients by IL-8 were analyzed by the Kaplan-Meier plotter. The levels of mRNA and protein expression were analyzed by real-time PCR and Western blotting. The alteration of apoptotic cell death-related proteins by SB225002 was analyzed by the Proteome Profiler Human Apoptosis Array. Cell growth was analyzed by MTT and colony-forming assay. Apoptosis and cell cycle were analyzed by fluorescence-activated cell sorting. Results: Aberrant IL-8 expression is involved with the prognosis of TNBC patients. Basal IL-8 levels are markedly elevated in TNBC cells compared to those in HER2+ and/or ER+ breast cancer cells. Furthermore, recombinant human IL-8 treatment enhanced cell invasiveness in TNBC cells. To counteract the tumor-promoting effects of IL-8, we assessed the therapeutic potential of CXCR1 and CXCR2 inhibitors. Notably, while reparixin, a CXCR1-specific inhibitor, exhibited no impact on cell viability, SB225002, a CXCR2-specific inhibitor, significantly reduced cell viability in a dose-dependent manner. There was a noticeable reduction in the levels of anti-apoptotic biomarkers, including Bcl-2, cIAP-1, cIAP-2, Survivin, XIAP, HIF-1α, and HO-1, following SB225002 treatment. Our findings indicate an increase in the apoptotic cell population with SB225002 treatment in TNBC cells. In xenograft models, SB225002 effectively diminished the metastatic potential of 4T1 cells, which are known to metastasize to the lung and liver. Conclusion: Our results underscore the significant role of the IL-8/CXCR2 signaling axis in the metastasis of TNBC and suggest that CXCR2 inhibitors such as SB225002 may be promising therapeutic agents for TNBC patients.

Breast cancer is the most commonly diagnosed cancer, accounting for 32% of all cancers in women [1]. Over the past decade, the incidence rates of breast cancer in young women have increased steeply, and it has also been the leading cause of cancer deaths in women aged 20–49 [1]. In particular, triple-negative breast cancer (TNBC) is characterized by its lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Accounting for approximately 15–20% of all breast cancer cases, it is recognized as a heterogeneous and malignant disease marked by a challenging clinical prognosis [2]. Renowned for being the most aggressive and invasive breast cancer subtype, the scarcity of effective therapeutic targets for TNBC has spurred a wealth of research endeavors [3]. Efforts to tailor therapies for TNBC through the use of biomarkers have been pursued [4]. Investigating such biomarkers offers a critical theoretical framework for understanding drug efficacy and resistance mechanisms in TNBC patients [5]. Several types of solid tumors, including TNBC, are known to drive cell proliferation and invasion via IL-8 expression, positioning IL-8 as a prognostic and potentially predictive biomarker [6, 7].

Among chemokines, IL-8, also designated as C-X-C motif ligand 8, is identified as a small, soluble protein [8]. IL-8 has been implicated in various cancer types, including those of the breast, lung, melanoma, prostate, and pancreas-colon continuum [9, 10]. It exerts a direct influence on tumors by modulating the inherent characteristics of breast cancer cells [11]. Notably, IL-8 has been highlighted for its significant role in enhancing cancer cell proliferation, angiogenesis, infiltration, and metastatic potential [12]. Indirectly, IL-8 may foster tumor growth and metastasis by reshaping the tumor microenvironment [11]. Moreover, IL-8 is crucial in mediating the differentiation and function of stromal and immune cells within the tumor microenvironment, thereby influencing the effectiveness of immunotherapy [13]. Clinical investigations have established a correlation between serum IL-8 levels and tumor stage, invasiveness, and patient prognosis [14, 15].

IL-8, alongside its receptors C-X-C chemokine receptor 1 (CXCR1) and C-X-C chemokine receptor 2 (CXCR2), has been shown to encourage tumorigenesis in breast cancer cells [16]. Expressed within these cells, CXCR1 and CXCR2 are pivotal in activating the inflammatory response through their chemokines [17] and in modulating tumor cell proliferation, metastasis, and angiogenesis across various cancer types [18, 19]. Despite sharing 76% homology, CXCR1 and CXCR2 display distinct functionalities owing to differential regional regulation [20]. IL-8 has been recognized as an oncogenic factor in several cancers, triggering the activation of Src, signal transducer and activator of transcription (STAT-3), and extracellular signal-regulated kinase (ERK)1/2 pathways. The IL-8/CXCR1 axis, in particular, has recently been highlighted as a promising therapeutic target for combating breast cancer stem cells [21, 22].

This study aimed to explore the pharmacological implications of CXCR1 and CXCR2 inhibition in TNBC cell lines. Prior research has elucidated the role of IL-8 in the progression of breast cancer, with aberrant expression of IL-8 linked to TNBC patient survival rates. Consequently, our focus has been drawn toward CXCR1 and CXCR2 inhibitors as a means to obstruct the IL-8-dependent signaling cascade. Herein, we demonstrate that the specific CXCR2 inhibitor, SB225002, significantly reduces TNBC cell proliferation, invasion, and metastasis. Thus, our findings advocate for the superior efficacy of CXCR2 inhibitors, such as SB225002, over CXCR1 inhibitors in the treatment of TNBC.

Cell Culture

Human breast cancer cell lines were obtained from the ATCC and were checked STR profiling data. MCF7 (ER+/PR+/HER2−), Hs578T (ER−/PR−/HER2−), MDA-MB468 (MDA468; ER−/PR−/HER2−), MDA-MB453 (MDA453; ER−/PR−/HER2+), and MDA-MB231 (MDA231; ER−/PR−/HER2−), were cultured in Dulbecco’s Modified Eagle’s Medium (HyClone; Cytiva, Marlborough, MA, USA), whereas T47D (ER+/PR+/HER2−), BT474 (ER+/PR+/HER2+), HCC1419 (ER−/PR−/HER2+), HCC1143 (ER−/PR−/HER2−), and SKBR3 (ER−/PR−/HER2+) were maintained in RPMI1640 medium (HyClone; Cytiva). All culture media were supplemented with 10% fetal bovine serum (FBS; HyClone, Logan, UT, USA), 100 IU/mL penicillin, and 100 μg/mL streptomycin. We housed all cells in a humidified environment containing 95% air and 5% CO2 at 37°C [23].

Survival Analysis Using Kaplan-Meier Plotter Database

Survival analysis was performed in TNBC patients using the Kaplan-Meier plotter database based on IL-8 mRNA expression levels [24]. For this analysis, we utilized the “202859_at” probe set and employed the “Auto select best cutoff” feature to stratify patients based on IL-8 expression. Patient stratification by TNBC subtypes was performed using the “ER status – IHC: ER-negative, ER status – array: ER-negative, PR status – IHC: PR-negative, HER2 status – array: HER2-negative” setting. Calculations and displays included the hazard ratio, 95% confidence intervals, and log-rank p values. We deemed p values <0.05 statistically significant.

Quantitative Reverse Transcription PCR

In our previous study, we employed TRIzol (Thermo Fisher Scientific) for total RNA extraction from the cells [25]. Following the manufacturer’s guidelines, we synthesized cDNA using a cDNA Synthesis Kit (Thermo Fisher Scientific). The SYBR Green master mix (Bioline Ltd., London, UK) was used for quantitative reverse transcription PCR on an ABI 7900HT real-time PCR detection system (Foster City, CA, USA), normalizing target gene expression against beta-actin (ACTB) using the comparative CT method (2−ΔΔCT) to determine relative gene expression (fold change). Thermal cycling involved an initial step at 50°C for 2 min, followed by 95°C for 10 min and 40 cycles of 95°C for 15 s, 60°C for 15 s, and 72°C for 15 s. We employed primers for human ACTB (forward, F: TCA CCA ATT GGA TGA GCG GTT; reverse, R: AGT TTC GTG GAT GCC ACA GGA C) and IL-8 (F: AGG GTT GCC AGA TGC AAT AC; R: AAA CCA AGG CAC AGT GGA AC) [25].

IL-8 ELISA

To evaluate secreted IL-8 protein levels, we analyzed the conditioned culture media (200 μL) from breast cancer cells using the manufacturer’s protocol of ELISA kit (R&D Systems, MN, USA).

Boyden Chamber Assay

We executed cell migration analyses through the Boyden chamber assay, as previously described [25]. TNBC cells were placed in the upper chamber of Boyden-chamber inserts, having an 8-μm pore size, within 24-well plates (Becton-Dickinson, San Diego, CA, USA). To the lower chamber, fresh culture medium containing 10% FBS was added. The cells (5 × 104 cells/well) in the upper chamber were subjected to 20 ng/mL recombinant human IL-8 treatment for 24 h. Post-treatment, cells on the filter’s upper surface were eradicated, the filter underwent fixation with 100% methanol, followed by staining with 0.1% crystal violet solution. The migrated cells were examined using a CK40 inverted microscope (Olympus, Tokyo, Japan). The quantification of migrated cells was counted using Image J software.

Cell Viability

We evaluated sensitivities to CXCR1 and CXCR2 inhibitors, specifically reparixin and SB225002, using the MTT assay. TNBC cells (1.5 × 103 cells/well) were plated onto 96-well plates. Post 24-h incubation, cells were exposed to DMEM supplemented with 10% FBS, with or without specified concentrations of reparixin or SB225002 for 48 h. To assess cell viability, we mixed equal volumes of serum-free media and MTT solution in each well. Following a 3-h incubation at 37°C, DMSO was added to dissolve the MTT formazan. Absorbance at 590 nm was measured using a tunable microplate reader [26].

Colony Formation Assays

For colony formation assays, TNBC cells (2 × 103 cells/well) were seeded into 12 or 6-well plates and incubated overnight at 37°C. Subsequently, cells received treatment with specified concentrations of reparixin or SB225002, followed by an additional 10-day incubation. Fixed with 100% ethanol and stained with 0.1% crystal violet, cell colonies were visualized using a CK40 inverted microscope (Olympus, Tokyo, Japan) [25, 26].

Western Blots

Cells were disrupted using PRO-PREP™ Protein Extraction Solution (Intron Biotechnology, Inc., Gyeonggi-do, Korea) on ice, centrifuged (16,100 g) for 15 min, and the resulting supernatants were harvested. Prepared samples, consisting of equal amounts of proteins boiled for 5 min in Laemmli sample buffer, were deployed into SDS-polyacrylamide gels under denaturing conditions and subsequently transferred to polyvinylidene fluoride membranes (Millipore, Billerica, MA, USA). Blocked with 5% skim milk for 1 h at room temperature (RT), membranes were incubated overnight at 4°C with primary antibodies against β-actin (Abfrontier, Seoul, South Korea, LF-PA0207, 1:2,000), PARP-1 (CST, Danvers, MA, USA, 9542S, 1:1,000), pro-caspase3 (Abcam, Waltham, MA, USA, ab32351, 1:5,000), cleaved-caspase3 (CST, 9661S, 1:1,000), p-STAT3 (Abcam, ab76315, 1:10,000), t-STAT3 (Santa Cruz Biotechnology, CA, USA, sc-8019, 1:1,000), p-AKT (Abcam, ab81283, 1:5,000), t-AKT (CST, 9272S, 1:1,000), p-ERK (Santa Cruz, sc-7383, 1:1,000), and t-ERK (CST, 9102S, 1:1,000). Following washing, membranes were treated with horseradish peroxidase-conjugated secondary antibodies (Cell Signaling Technology, Danvers, MA, USA) at room temperature for 1 h. Visualization of transferred proteins was achieved using the ECL prime reagent (GE Healthcare, Bucks, UK), and the protein bands on the X-ray film were sensitized using a JPI automatic X-ray film processor (JPI Healthcare Co., Ltd, Seoul, South Korea, JP-33).

Cell Cycle Analysis

We conducted flow cytometry with propidium iodide-stained cells to analyze the cell cycle. Post-harvesting, cells were fixed with 70% ethanol at room temperature for 20 min. After fixation, cells were treated with 100 µg/mL DNase-free RNase A (Thermo Fisher Scientific, Waltham, MA, USA) at 37°C for 30 min. Following a PBS wash, cells were resuspended in fluorescence-activated cell sorting (FACS) buffer containing 50 µg/mL propidium iodide (Sigma-Aldrich, St. Louis, MO, USA) and analyzed using a BD FACSCalibur flow cytometer (BD, Becton-Dickinson, San Diego, CA, USA) [25].

Apoptosis Analysis

For apoptosis assays, we utilized the Annexin V PE Apoptosis Detection Kit (BioGems, Via Colinas, Westlake Village, USA), following the manufacturer’s protocol. In short, cells (1 × 106 cells/mL) were collected, washed twice with PBS, and then resuspended in 500 μL of staining solution containing 5 μL Annexin V-PE and PE-Cy7. Following a 15-min incubation at RT in a dark environment, cells were immediately subject to analysis on a flow cytometer. Apoptotic cells, identified by dual staining with Annexin V-PE and PE-Cy7, were assessed using the FACS Vantage system (Becton-Dickinson, San Diego, CA, USA), with the percentage of cells undergoing apoptosis quantified thereafter.

Apoptosis Protein Array

Utilizing the Proteome Profiler Human Apoptosis array kit (R&D Systems, Minneapolis, MN, USA), we analyzed cell lysates from 10 μm SB225002-treated and -untreated vehicles at 48 h post-treatment for apoptosis-related proteins, adhering strictly to the manufacturer’s guidelines. Cell lysates were prepared using lysis buffer from the kit, and protein concentration was ascertained through the Bradford method. Following a blockage phase in buffer, equilibrated proteins were incubated overnight at 4°C on a rocking platform shaker with precoated array membranes ready for analysis. Post-incubation, the membranes underwent three washes in 1× washing buffer to discard unattached proteins, followed by detection antibody cocktail treatment for 1 h at room temperature while shaking. After additional washes, the membranes were incubated with diluted streptavidin-HRP for 30 min at room temperature under shaking. Detection was performed by X-ray film exposure, leveraging enhanced chemiluminescence. Quantification of each protein level was relative to positive control spots on the same membrane.

Xenograft Study and Immunohistochemical Staining

Female BALB/c nude mice (Orient Bio, Seoul, Korea) at 6–8 weeks were used to establish a nude mouse xenograft model. All animal care and experimental procedures complied with guidelines approved by the Institutional Animal Care and Use Committee (IACUC) of Samsung Medical Center (Protocol No. 20240403001) [26].

To investigate the effect of SB225002 on lung metastasis, 4T1 cells (1.8 × 107 cells/120 μL) were resuspended in Matrigel (BD Biosciences, Bedford, MA, USA) and injected directly into the right secondary mammary fat pads. The mice were randomly divided into two groups (n = 4/group), which were treated with vehicle (25% PEG 400 and 5% Tween 80) or 10 mg/kg SB225002 starting on the day of inoculation and continuing until the end of the experiment. Once the tumors reached a volume of approximately 50 mm3, the tumor size was measured using digital calipers at set time points, and the volume was determined using the formula V = 1/2 × length × (width)2. Growth curves were calculated using the average relative tumor volume per group at the set time points [26].

For immunohistochemistry assays, paraffin-embedded lung tissue sections (4 μm) were deparaffinized in xylene, dehydrated in graded alcohol, and hydrated in water. Tissue sections were evaluated by H&E staining for detection of lung metastasis. The slides were analyzed using a ScanScope XT apparatus (Aperio Technologies, CA, USA) [26].

Statistical Analysis

Graph construction was carried out using Microsoft Excel 2016 (Microsoft, Redmond, WA, USA) and GraphPad Prism 8 software (GraphPad Software, La Jolla, CA, USA). We computed p values using the Student’s t test (unpaired, two-tailed) and one-way ANOVA. All experimental operations were independently replicated a minimum of three times. Data presentation adheres to the mean ± standard error of the mean (SEM) format, with significance ascribed to differences when p < 0.05.

Aberrant IL-8 Expression Is Associated with Poor Prognosis in TNBC Patients

To assess the role of IL-8 expression on survival rates across different breast cancer subtypes, we utilized the Kaplan-Meier plotter dataset. Our analysis revealed a significant correlation between IL-8 expression and survival outcomes in TNBC patients (Fig. 1a–c). Patients with elevated IL-8 levels exhibited substantially worse RFS (Fig. 1a, p = 0.00058), OS (Fig. 1b, p = 0.00049), and DMFS (Fig. 1c, p = 0.011) compared to those with lower IL-8 expression. Hence, these findings underscore the direct impact of IL-8 expression on the prognosis of TNBC.

Fig. 1.

Aberrant IL-8 expression is associated with poor prognosis in TNBC patients. Analysis of recurrence-free survival (RFS, low, n = 283; high, n = 109) (a), overall survival (OS, low, n = 93; high, n = 60) (b), and distant metastasis-free survival (DMFS, low, n = 160; high, n = 146) (c) in relation to IL-8 expression levels, evaluated through the Kaplan-Meier plotter database for TNBC patients. Statistical significance determined via the log-rank test.

Fig. 1.

Aberrant IL-8 expression is associated with poor prognosis in TNBC patients. Analysis of recurrence-free survival (RFS, low, n = 283; high, n = 109) (a), overall survival (OS, low, n = 93; high, n = 60) (b), and distant metastasis-free survival (DMFS, low, n = 160; high, n = 146) (c) in relation to IL-8 expression levels, evaluated through the Kaplan-Meier plotter database for TNBC patients. Statistical significance determined via the log-rank test.

Close modal

IL-8 Expression Is Increased in TNBC Cells

Evaluating the expression levels of IL-8 in breast cancer cells, we discovered a pronounced upregulation of IL-8 mRNA and protein expression in TNBC cells, in contrast to other breast cancer subtypes (Fig. 2a, b). To further comprehend the consequences of inherent IL-8 expression in TNBC cells, we analyzed the metastatic potential through a Boyden chamber migration assay in HCC1143, Hs578T, and MDA231 cells. Recombinant human IL-8 significantly heightened migration across cell culture inserts (Fig. 2c), indicating that aberrant IL-8 expression is critical for increasing TNBC cell migration ability. As shown in Figure 2c, cell invasiveness by IL-8 treatment was increased 2.8-fold (in HCC1143), 3.1-fold (in Hs578T), and 4.6-fold (MDA231) compared to the control groups, respectively.

Fig. 2.

The levels of IL-8 expression in breast cancer cells and the effect of IL-8 on cell invasiveness in TNBC cells. a Determination of IL-8 transcript levels across ER-α+ (ER+/PR+/HER2−: MCF7, T47D or ER+/PR+/HER2+: BT474), HER2+ (ER−/PR−/HER2+: HCC1419, SKBR3, MDA453), and TNBC (ER−/PR−/HER2−: HCC1143, Hs578T, MDA231, MDA468) cells through quantitative reverse transcription PCR. b Quantification of secreted IL-8 proteins in conditioned media from breast cancer cells using ELISA. c Assessment of cell invasion in TNBC cells with or without treatment of 20 ng/mL IL-8. Results presented are from three separate experiments.

Fig. 2.

The levels of IL-8 expression in breast cancer cells and the effect of IL-8 on cell invasiveness in TNBC cells. a Determination of IL-8 transcript levels across ER-α+ (ER+/PR+/HER2−: MCF7, T47D or ER+/PR+/HER2+: BT474), HER2+ (ER−/PR−/HER2+: HCC1419, SKBR3, MDA453), and TNBC (ER−/PR−/HER2−: HCC1143, Hs578T, MDA231, MDA468) cells through quantitative reverse transcription PCR. b Quantification of secreted IL-8 proteins in conditioned media from breast cancer cells using ELISA. c Assessment of cell invasion in TNBC cells with or without treatment of 20 ng/mL IL-8. Results presented are from three separate experiments.

Close modal

A Specific CXCR2 Inhibitor, SB225002, Dose-Dependently Decreases the Cell Viabilities in TNBC Cells

Figure 3a illustrates the chemical structures and functions of reparixin and SB225002. In our effort to curb IL-8-mediated signaling in TNBC cells, we administered a specific CXCR1 inhibitor (reparixin) or a CXCR2 inhibitor (SB225002) for 48 h at various concentrations. Interestingly, reparixin showed no effect on cell viability (Fig. 3b), whereas SB225002 distinctly reduced cell viability in a dose-dependent fashion (Fig. 3c). Additionally, we evaluated the pharmacological effectiveness of both inhibitors on cell growth using a colony-forming assay. As anticipated, our experiments confirmed that SB225002 notably hindered cell proliferation (Fig. 3d), spotlighting CXCR2 inhibitors like SB225002 as more efficacious than CXCR1 inhibitors in the treatment of TNBC.

Fig. 3.

A specific CXCR2 inhibitor, SB225002, dose-dependently decreases the cell viabilities in TNBC cells. a Depiction of chemical structures for reparixin and SB225002. b Analysis of cell viabilities post reparixin treatment via MTT assay. c Evaluation of cell viabilities following SB225002 treatment through MTT assay. d Investigation of cell proliferation following treatment with reparixin or SB225002 through colony forming assay. Data represent three independent trials. Values are expressed as mean ± SEM. Significance indicated by *p < 0.05, **p < 0.01 compared to the untreated group.

Fig. 3.

A specific CXCR2 inhibitor, SB225002, dose-dependently decreases the cell viabilities in TNBC cells. a Depiction of chemical structures for reparixin and SB225002. b Analysis of cell viabilities post reparixin treatment via MTT assay. c Evaluation of cell viabilities following SB225002 treatment through MTT assay. d Investigation of cell proliferation following treatment with reparixin or SB225002 through colony forming assay. Data represent three independent trials. Values are expressed as mean ± SEM. Significance indicated by *p < 0.05, **p < 0.01 compared to the untreated group.

Close modal

SB225002 Induces the Apoptotic Cell Death

Following promising results, we proceeded to analyze apoptosis-related protein expressions using the Proteome Profiler Human Apoptosis Array, post-SB225002 treatment. The treatment resulted in a visible decline in green square indicators (Fig. 4a). Key proteins associated with apoptosis, including Bcl-2, cIAP-1, cIAP-2, Survivin, XIAP, HIF-1α, and HO-1 experienced significant downregulation upon SB225002 treatment. Furthermore, Annexin V-PE and PE-Cy7 staining showcased a notable increase in apoptosis rates in SB225002-treated groups compared to the control, whereas reparixin showed no observable effect in TNBC cells (Fig. 4b, c). Additionally, we examined cell cycle alterations induced by SB225002 or reparixin. Our results showed that the subG1 population significantly increased in both HCC1143 and MDA231 cells following treatment with SB225002 (online supplement 1A, B; for all online suppl. material, see https://doi.org/10.1159/000545659), but not reparixin (online supplement 1C, D). As shown in Figure 4d, pro-PARP-1 expression was decreased by SB225002 while cleaved-PARP-1 expression was increased. Also, pro-caspase 3 and cleaved caspase 3 expression showed a similar pattern, as did PARP-1 expression in Hs578T cells. The phosphorylation of intracellular signaling molecules such as STAT-3, AKT, and ERK was also decreased by SB225002 but not by reparixin (Fig. 4e). Collectively, these findings illustrate that SB225002, unlike reparixin, can trigger apoptotic cell death in TNBC cells.

Fig. 4.

SB225002 induces the apoptotic cell death in Hs578T TNBC cells. a Proteomic array analysis of apoptosis-related proteins using Proteome Profiler Human Apoptosis Array Kit. We treated with 10 µm SB225002 for 24 h in Hs578T cells. After 24 h, whole cell lysates were harvested using lysis buffer from the kit. Dot blotting was performed as described in the methods. b FACS-based quantification of apoptotic cell death via Annexin V-PE and PE-Cy7 staining, as outlined in methods. c Graphical representation of data from b. d, e We treated with 10 µm SB225002 or reparixin for 24 h in Hs578T cells. After 24 h, whole cell lysates were harvested. Levels of pro-, cleaved-PARP-1, caspase3, total (t)-, phospho (p)-STAT-3, AKT, and ERK proteins were analyzed by Western blotting. β-actin was used as a loading control. Represented results are from three independent experiments. Values are mean ± SEM, with **p < 0.01 against control (Con).

Fig. 4.

SB225002 induces the apoptotic cell death in Hs578T TNBC cells. a Proteomic array analysis of apoptosis-related proteins using Proteome Profiler Human Apoptosis Array Kit. We treated with 10 µm SB225002 for 24 h in Hs578T cells. After 24 h, whole cell lysates were harvested using lysis buffer from the kit. Dot blotting was performed as described in the methods. b FACS-based quantification of apoptotic cell death via Annexin V-PE and PE-Cy7 staining, as outlined in methods. c Graphical representation of data from b. d, e We treated with 10 µm SB225002 or reparixin for 24 h in Hs578T cells. After 24 h, whole cell lysates were harvested. Levels of pro-, cleaved-PARP-1, caspase3, total (t)-, phospho (p)-STAT-3, AKT, and ERK proteins were analyzed by Western blotting. β-actin was used as a loading control. Represented results are from three independent experiments. Values are mean ± SEM, with **p < 0.01 against control (Con).

Close modal

SB225002 Suppresses the Metastatic Potential of TNBC Cells

Continuing our exploration, we assessed the pharmacological impact of SB225002 on 4T1 mouse TNBC cells. Treatment with SB225002 notably amplified the total apoptotic cell population, a contrast not seen with reparixin treatment (Fig. 5a). Specifically, the apoptotic cell population increased by 12.75 ± 0.59% (early) and 7.63 ± 0.36% (late) relative to control levels (Fig. 5b). Conforming to expectations, cell proliferation markedly decreased upon SB225002 treatment (Fig. 5c). Observations also included cell morphological changes toward roundness, swelling, and the formation of a balloon-like structure (Fig. 5c). Consistent with the results in Figure 4d, the expression of apoptosis-related proteins such as PARP-1 and caspase 3 was also regulated by SB225002 in 4T1 murine TNBC cells (Fig. 5d). To investigate the pharmacological efficacy of SB225002 in vivo, we performed xenograft model studies, which are schematically illustrated in Figure 5e. Moreover, examining the suppressive effects of SB225002 on tumor growth and metastatic potential via orthotopic xenograft models, we noted a significant reduction in primary tumor size at the second fat pad due to SB225002 treatment (Fig. 5f). The size of tumors in the SB225002-treated group was about half of that in the vehicle-treated group (Fig. 5f), and lung metastasis was dramatically reduced (Fig. 5g). This compilation of results highlights the efficacy of SB225002 as a potent antagonist in curbing growth and metastasis of TNBC cells.

Fig. 5.

SB225002 suppresses the metastatic potential of TNBC. a FACS-based analysis of apoptotic cell death in 4T1 cells employing Annexin V-PE and PE-Cy7 staining, following the described methods. b Quantification and graphical representation of findings from a. Data stem from three separate studies. Values presented as mean ± SEM, **p < 0.01 versus control (Con). c Examination of cell proliferation via colony forming assay post-treatment with reparixin or SB225002. d We treated with 10 µm SB225002 or reparixin for 24 h in 4T1 cells. After 24 h, whole cell lysates were harvested. Levels of pro-, and cleaved-PARP-1, caspase3 proteins were analyzed by Western blotting. β-actin was used as a loading control. e Illustrative scheme of the experimental approach. f Evaluation of primary tumor size in the 2nd fat pad following vehicle or SB225002 treatment, expressed as mean ± SEM, p = 0.0046 (vehicle vs. SB225002). g Analysis of lung metastasis of 4T1 cells post vehicle or SB225002 treatment. Control identified as Con.

Fig. 5.

SB225002 suppresses the metastatic potential of TNBC. a FACS-based analysis of apoptotic cell death in 4T1 cells employing Annexin V-PE and PE-Cy7 staining, following the described methods. b Quantification and graphical representation of findings from a. Data stem from three separate studies. Values presented as mean ± SEM, **p < 0.01 versus control (Con). c Examination of cell proliferation via colony forming assay post-treatment with reparixin or SB225002. d We treated with 10 µm SB225002 or reparixin for 24 h in 4T1 cells. After 24 h, whole cell lysates were harvested. Levels of pro-, and cleaved-PARP-1, caspase3 proteins were analyzed by Western blotting. β-actin was used as a loading control. e Illustrative scheme of the experimental approach. f Evaluation of primary tumor size in the 2nd fat pad following vehicle or SB225002 treatment, expressed as mean ± SEM, p = 0.0046 (vehicle vs. SB225002). g Analysis of lung metastasis of 4T1 cells post vehicle or SB225002 treatment. Control identified as Con.

Close modal

TNBC is characterized by a highly aggressive phenotype and an increased propensity for metastasizing to the central nervous system and visceral organs, such as the liver, lungs, and bones due to the absence of effective therapeutic targets [27]. Furthermore, TNBC patients often experience a significantly shortened survival period following metastasis, with an early recurrence rate of 25% after surgical intervention; notably, the predominant cause of mortality in breast cancer emanates from metastatic progression rather than primary tumor growth [27, 28]. Despite these challenges, the search for an effective targeted therapy for TNBC remains ongoing. In our investigation, we identified a marked reduction in survival rates among TNBC patients with aberrant induction of IL-8. Moreover, the expression levels of IL-8 mRNA and protein were found to be substantially elevated in TNBC cells compared to other subtypes, leading us to explore the potential of targeting IL-8’s binding receptors, specifically CXCR1 and CXCR2, as therapeutic strategies for TNBC.

Prior research by Kim et al. [29] elucidated that IL-8 is secreted by various cell types and exhibits highest expression within the TNBC stromal environment relative to other breast cancer subtypes [30]. The overexpression of IL-8 correlates with several oncogenic characteristics, including angiogenesis, invasion, and enhanced metastatic capacity across multiple carcinoma types, serving as a prognostic marker for adverse clinical outcomes [31, 32]. Zhang et al. [33] reported that the expression level of serum IL-8 was 79.68 ± 9.53 ng/L in benign group and 220.54 ± 12.49 ng/L in malignant group, and both were higher than the 54.31 ± 10.26 ng/L in healthy control group in ovarian cancer. The basal expression of IL-8 is modulated via a mitogen-activated protein kinase)/ ERK1/2-dependent pathway in TNBC cells [29]. Contrarily, employing a specific MEK inhibitor, UO126, led to reduced anchorage-independent growth, cell invasion, and migration in TNBC cultures [29]. Our results were also revealed that the levels of ERK, AKT, and STAT-3 phosphorylation were decreased by SB225002 but not by reparixin. Additionally, we observed an augmentation in cell invasiveness following treatment with recombinant human IL-8 in a spectrum of TNBC cell lines. Thus, it is posited that disrupting IL-8-mediated signaling pathways could offer a novel approach for curbing TNBC proliferation and metastasis.

IL-8 exerts its biological actions through binding to G-protein-coupled receptors, specifically the cysteine-X-cysteine receptors CXCR1 and CXCR2 [34]. The IL-8/CXCR complex initiates a cascade of intracellular signaling pathways, including phosphoinositide-3-kinase/protein kinase B Akt (PI3K/Akt), mitogen-activated protein kinase, and STAT-3, implicated in tumorigenesis [35‒37]. Although our results did not explicitly demonstrate this, we noted that the mRNA expression levels of CXCR1 and CXCR2 did not exhibit significant variance across different breast cancer cell types. Nonetheless, the abnormal induction of IL-8 and its interaction with respective receptors in TNBC prompted an investigation into the efficacy of specific inhibitors. Intriguingly, our data unveiled distinct responses to CXCR1 and CXCR2 inhibitors in TNBC cells, with prior studies highlighting CXCR2’s involvement in tumor growth, metastasis, and chemoresistance in various cancers, including breast carcinoma [34‒38]. Consequently, we provide evidence that the IL-8/CXCR2 signaling axis, unlike CXCR1, may represent a viable therapeutic target in TNBC by promoting cancer cell apoptosis to inhibit metastasis.

Many studies have revealed that CXCR2 expression is associated with their proliferation, invasion, and migration in a variety of cancer cells including lung and pancreatic cancers [38, 39]. In addition, the inhibition of CXCR1/2 by a IL-8 analog or the CXCR2 antagonists decreased lung tumor cell proliferation, angiogenesis, and metastasis in vivo [18, 40]. Prajapati et al. [41] reported that SCH-479833, a CXCR1/2 antagonist, significantly decreased cell proliferation of tumors derived from antagonist-treated mice were and increased apoptosis through immunostaining of Ki-67 and CC3. The anticancer agents induce the intrinsic pathway for apoptosis, whereas direct activation of extrinsic apoptotic pathway may not be needed to kill the cells downstream of antineoplastic agents [42]. Based on these results, our results revealed that mice treated with the SB225002 significantly reduced tumor volume and incidence of lung metastasis when compared to the control group in vivo study using xenografts model. These findings reflect that the apoptotic potential of CXCR2 antagonists such as SB225002 is effective in preventing tumor growth or lung metastasis in TNBC.

Elevated secretion levels of IL-8 were predominantly observed in TNBC cells compared to other subtypes. The aberrant induction of IL-8 was strongly linked with poor survival outcomes in TNBC patients. Leveraging these insights, our study evaluated the therapeutic potentials of CXCR1- and CXCR2-specific inhibitors using TNBC models. Notably, SB225002, a selective CXCR2 inhibitor, exhibited pronounced effectiveness in inducing apoptotic cell death in TNBC, whereas reparixin, a CXCR1 inhibitor, demonstrated negligible impact. Subsequent investigations confirmed that SB225002 substantially inhibited tumor growth and lung metastasis in 4T1 cell-based lung metastasis models. Here, we demonstrated that CXCR2 antagonist is effective for the treatment of TNBCs that are not subject to targeted therapy against ER, PR, and HER2. Based on these preclinical findings, the inhibitory effects of the CXCR2 antagonist resulted from decreased tumor growth, progression, and metastasis of TNBC cells. A significant point of further exploration includes the downstream signaling pathways of IL-8/CXCR2 complex or the mechanism of intrinsic or extrinsic apoptosis pathway in TNBC cells. Collectively, these findings underscore the critical role of the IL-8/CXCR2 signaling pathway in TNBC tumor proliferation and metastatic dissemination. We cautiously propose that therapies targeting CXCR2 may offer a more promising treatment modality compared to CXCR1-based strategies for TNBC management.

All animal care and experimental procedures were performed in accordance with the principles and guidelines of the Korea Council for Animal Care. This study protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Samsung Medical Center (Approval No. 20240403001). Experiments were performed in accordance with the relevant guidelines and regulations. Studies with humans were collected through the Kaplan-Meier plotter database (https://kmplot.com/analysis/index.php?p=service&cancer=breast). This database is open source for easy access for all researchers, so no separate ethical approval is required for data collection.

The authors declare no conflicts of interest.

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (RS-2024-00452849).

Y.J., S.Y.Y., and S.P.J. contributed to the investigation, data curation, formal analysis, and writing – original draft. S.K. and S.J.N contributed to the manuscript review and editing. S.K and J.E.L. contributed to the study conceptualization, data curation, formal analysis, and validation. All authors have read and agreed to the published version of the manuscript.

Additional Information

Yisun Jeong, Sun Young Yoon, and Seung Pil Jung contributed equally to this work.

The data that support the findings of the current study are not publicly available due to their containing information that could compromise the privacy of research participants but are available from the corresponding author (Sangmin Kim: [email protected]) on reasonable request.

1.
Siegel
RL
,
Giaquinto
AN
,
Jemal
A
.
Cancer statistics, 2024
.
CA Cancer J Clin
.
2024
;
74
(
1
):
12
49
.
2.
So
JY
,
Ohm
J
,
Lipkowitz
S
,
Yang
L
.
Triple negative breast cancer (tnbc): non-genetic tumor heterogeneity and immune microenvironment: emerging treatment options
.
Pharmacol Ther
.
2022
;
237
:
108253
.
3.
Liu
J
,
Chen
K
,
Jiang
W
,
Mao
K
,
Li
S
,
Kim
MJ
, et al
.
Chemotherapy response and survival of inflammatory breast cancer by hormone receptor- and her2-defined molecular subtypes approximation: an analysis from the national cancer database
.
J Cancer Res Clin Oncol
.
2017
;
143
(
1
):
161
8
.
4.
de Gramont
A
,
Watson
S
,
Ellis
LM
,
Rodón
J
,
Tabernero
J
,
de Gramont
A
, et al
.
Pragmatic issues in biomarker evaluation for targeted therapies in cancer
.
Nat Rev Clin Oncol
.
2015
;
12
(
4
):
197
212
.
5.
Fleisher
B
,
Clarke
C
,
Ait-Oudhia
S
.
Current advances in biomarkers for targeted therapy in triple-negative breast cancer
.
Breast Cancer (Dove Med Press)
.
2016
;
8
:
183
97
.
6.
Uddin
MM
,
Gaire
B
,
Deza
B
,
Vancurova
I
.
Interleukin-8-induced invasion assay in triple-negative breast cancer cells
.
Methods Mol Biol
.
2020
;
2108
:
107
15
.
7.
Wani
AK
,
Prakash
A
,
Sena
S
,
Akhtar
N
,
Singh
R
,
Chopra
C
, et al
.
Unraveling molecular signatures in rare bone tumors and navigating the cancer pathway landscapes for targeted therapeutics
.
Crit Rev Oncol Hematol
.
2024
;
196
:
104291
.
8.
Rossi
D
,
Zlotnik
A
.
The biology of chemokines and their receptors
.
Annu Rev Immunol
.
2000
;
18
:
217
42
.
9.
Todorović-Raković
N
,
Milovanović
J
.
Interleukin-8 in breast cancer progression
.
J Interferon Cytokine Res
.
2013
;
33
(
10
):
563
70
.
10.
Cheng
Y
,
Ma
XL
,
Wei
YQ
,
Wei
XW
.
Potential roles and targeted therapy of the cxcls/cxcr2 axis in cancer and inflammatory diseases
.
Biochim Biophys Acta Rev Cancer
.
2019
;
1871
(
2
):
289
312
.
11.
Fousek
K
,
Horn
LA
,
Palena
C
.
Interleukin-8: a chemokine at the intersection of cancer plasticity, angiogenesis, and immune suppression
.
Pharmacol Ther
.
2021
;
219
:
107692
.
12.
Waugh
DJ
,
Wilson
C
.
The interleukin-8 pathway in cancer
.
Clin Cancer Res
.
2008
;
14
(
21
):
6735
41
.
13.
Han
ZJ
,
Li
YB
,
Yang
LX
,
Cheng
HJ
,
Liu
X
,
Chen
H
.
Roles of the cxcl8-cxcr1/2 axis in the tumor microenvironment and immunotherapy
.
Molecules
.
2021
;
27
(
1
):
137
.
14.
Mishra
A
,
Suman
KH
,
Nair
N
,
Majeed
J
,
Tripathi
V
.
An updated review on the role of the cxcl8-cxcr1/2 axis in the progression and metastasis of breast cancer
.
Mol Biol Rep
.
2021
;
48
(
9
):
6551
61
.
15.
Habanjar
O
,
Bingula
R
,
Decombat
C
,
Diab-Assaf
M
,
Caldefie-Chezet
F
,
Delort
L
.
Crosstalk of inflammatory cytokines within the breast tumor microenvironment
.
Int J Mol Sci
.
2023
;
24
(
4
):
4002
.
16.
Zlotnik
A
,
Yoshie
O
.
The chemokine superfamily revisited
.
Immunity
.
2012
;
36
(
5
):
705
16
.
17.
Ha
H
,
Debnath
B
,
Neamati
N
.
Role of the cxcl8-cxcr1/2 axis in cancer and inflammatory diseases
.
Theranostics
.
2017
;
7
(
6
):
1543
88
.
18.
Khan
MN
,
Wang
B
,
Wei
J
,
Zhang
Y
,
Li
Q
,
Luan
X
, et al
.
Cxcr1/2 antagonism with cxcl8/interleukin-8 analogue cxcl8(3-72)k11r/g31p restricts lung cancer growth by inhibiting tumor cell proliferation and suppressing angiogenesis
.
Oncotarget
.
2015
;
6
(
25
):
21315
27
.
19.
Zuccari
DA
,
Leonel
C
,
Castro
R
,
Gelaleti
GB
,
Jardim
BV
,
Moscheta
MG
, et al
.
An immunohistochemical study of interleukin-8 (il-8) in breast cancer
.
Acta Histochem
.
2012
;
114
(
6
):
571
6
.
20.
Xu
C
,
Zhao
B
,
Xu
L
,
Wang
Y
,
Liu
B
,
Xu
M
, et al
.
Cxcr1 participates in bone cancer pain induced by walker 256 breast cancer cells in female rats
.
Mol Pain
.
2022
;
18
:
17448069221135743
.
21.
Brandolini
L
,
Cristiano
L
,
Fidoamore
A
,
De Pizzol
M
,
Di Giacomo
E
,
Florio
TM
, et al
.
Targeting cxcr1 on breast cancer stem cells: signaling pathways and clinical application modelling
.
Oncotarget
.
2015
;
6
(
41
):
43375
94
.
22.
Ahmed
S
,
Mohamed
HT
,
El-Husseiny
N
,
El Mahdy
MM
,
Safwat
G
,
Diab
AA
, et al
.
Il-8 secreted by tumor associated macrophages contribute to lapatinib resistance in her2-positive locally advanced breast cancer via activation of src/stat3/erk1/2-mediated egfr signaling
.
Biochim Biophys Acta Mol Cell Res
.
2021
;
1868
(
6
):
118995
.
23.
Dai
X
,
Cheng
H
,
Bai
Z
,
Li
J
.
Breast cancer cell line classification and its relevance with breast tumor subtyping
.
J Cancer
.
2017
;
8
(
16
):
3131
41
.
24.
Győrffy
B
.
Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer
.
Comput Struct Biotechnol J
.
2021
;
19
:
4101
9
.
25.
You
D
,
Kim
H
,
Jeong
Y
,
Yoon
SY
,
Lo
E
,
Kim
S
, et al
.
Tumorigenicity of egfr- and/or her2-positive breast cancers is mediated by recruitment of tumor-associated macrophages
.
Int J Mol Sci
.
2023
;
24
(
2
):
1443
.
26.
You
D
,
Jeong
Y
,
Yoon
SY
,
Kim
SA
,
Lo
E
,
Kim
SW
, et al
.
Entelon® (Vitis vinifera seed extract) prevents cancer metastasis via the downregulation of interleukin-1 alpha in triple-negative breast cancer cells
.
Molecules
.
2021
;
26
(
12
):
3644
.
27.
Al-Mahmood
S
,
Sapiezynski
J
,
Garbuzenko
OB
,
Minko
T
.
Metastatic and triple-negative breast cancer: challenges and treatment options
.
Drug Deliv Transl Res
.
2018
;
8
(
5
):
1483
507
.
28.
Jaradat
SK
,
Ayoub
NM
,
Al Sharie
AH
,
Aldaod
JM
.
Targeting receptor tyrosine kinases as a novel strategy for the treatment of triple-negative breast cancer
.
Technol Cancer Res Treat
.
2024
;
23
:
15330338241234780
.
29.
Kim
S
,
Lee
J
,
Jeon
M
,
Lee
JE
,
Nam
SJ
.
Mek-dependent il-8 induction regulates the invasiveness of triple-negative breast cancer cells
.
Tumour Biol
.
2016
;
37
(
4
):
4991
9
.
30.
Han
J
,
Bae
SY
,
Oh
SJ
,
Lee
J
,
Lee
JH
,
Lee
HC
, et al
.
Zerumbone suppresses IL-1β-induced cell migration and invasion by inhibiting IL-8 and MMP-3 expression in human triple-negative breast cancer cells
.
Phytother Res
.
2014
;
28
(
11
):
1654
60
.
31.
Li
XP
,
Yang
XY
,
Biskup
E
,
Zhou
J
,
Li
HL
,
Wu
YF
, et al
.
Co-expression of cxcl8 and hif-1α is associated with metastasis and poor prognosis in hepatocellular carcinoma
.
Oncotarget
.
2015
;
6
(
26
):
22880
9
.
32.
Peng
S
,
Chen
Y
,
Gong
Y
,
Li
Z
,
Xie
R
,
Lin
Y
, et al
.
Predictive value of intratumour inflammatory cytokine mrna levels of hepatocellular carcinoma patients and activation of two distinct pathways govern il-8 induced epithelial-mesenchymal transition in human hepatic cancer cell lines
.
Cytokine
.
2019
;
119
:
81
9
.
33.
Zhang
L
,
Liu
W
,
Wang
X
,
Wang
X
,
Sun
H
.
Prognostic value of serum il-8 and il-10 in patients with ovarian cancer undergoing chemotherapy
.
Oncol Lett
.
2019
;
17
(
2
):
2365
9
.
34.
Wang
Y
,
Tu
L
,
Du
C
,
Xie
X
,
Liu
Y
,
Wang
J
, et al
.
Cxcr2 is a novel cancer stem-like cell marker for triple-negative breast cancer
.
Onco Targets Ther
.
2018
;
11
:
5559
67
.
35.
Knall
C
,
Worthen
GS
,
Johnson
GL
.
Interleukin 8-stimulated phosphatidylinositol-3-kinase activity regulates the migration of human neutrophils independent of extracellular signal-regulated kinase and p38 mitogen-activated protein kinases
.
Proc Natl Acad Sci U S A
.
1997
;
94
(
7
):
3052
7
.
36.
Knall
C
,
Young
S
,
Nick
JA
,
Buhl
AM
,
Worthen
GS
,
Johnson
GL
.
Interleukin-8 regulation of the ras/raf/mitogen-activated protein kinase pathway in human neutrophils
.
J Biol Chem
.
1996
;
271
(
5
):
2832
8
.
37.
Wu
T
,
Wang
Z
,
Liu
Y
,
Mei
Z
,
Wang
G
,
Liang
Z
, et al
.
Interleukin 22 protects colorectal cancer cells from chemotherapy by activating the stat3 pathway and inducing autocrine expression of interleukin 8
.
Clin Immunol
.
2014
;
154
(
2
):
116
26
.
38.
Tazzyman
S
,
Barry
ST
,
Ashton
S
,
Wood
P
,
Blakey
D
,
Lewis
CE
, et al
.
Inhibition of neutrophil infiltration into a549 lung tumors in vitro and in vivo using a cxcr2-specific antagonist is associated with reduced tumor growth
.
Int J Cancer
.
2011
;
129
(
4
):
847
58
.
39.
Matsuo
Y
,
Ochi
N
,
Sawai
H
,
Yasuda
A
,
Takahashi
H
,
Funahashi
H
, et al
.
Cxcl8/il-8 and cxcl12/sdf-1alpha co-operatively promote invasiveness and angiogenesis in pancreatic cancer
.
Int J Cancer
.
2009
;
124
(
4
):
853
61
.
40.
Steele
CW
,
Karim
SA
,
Leach
JDG
,
Bailey
P
,
Upstill-Goddard
R
,
Rishi
L
, et al
.
Cxcr2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma
.
Cancer Cell
.
2016
;
29
(
6
):
832
45
.
41.
Prajapati
DR
,
Molczyk
C
,
Purohit
A
,
Saxena
S
,
Sturgeon
R
,
Dave
BJ
, et al
.
Small molecule antagonist of cxcr2 and cxcr1 inhibits tumor growth, angiogenesis, and metastasis in pancreatic cancer
.
Cancer Lett
.
2023
;
563
:
216185
.
42.
Wani
AK
,
Akhtar
N
,
Mir
TUG
,
Singh
R
,
Jha
PK
,
Mallik
SK
, et al
.
Targeting apoptotic pathway of cancer cells with phytochemicals and plant-based nanomaterials
.
Biomolecules
.
2023
;
13
(
2
):
194
.