The roles of the chemokine CCL5 and its receptor CCR5 in breast cancer progression remain unclear. Here, we conducted microarray analysis on 2,254 human breast cancer specimens and found increased expression of CCL5 and its receptor CCR5, but not CCR3, in the basal and HER-2 genetic subtypes. The subpopulation of human breast cancer cell lines found to express CCR5 displayed a functional response to CCL5. In addition, oncogene transformation induced CCR5 expression, and the subpopulation of cells that expressed functional CCR5 also displayed increased invasiveness. The CCR5 antagonists maraviroc or vicriviroc, developed to block CCR5 HIV coreceptor function, reduced in vitro invasion of basal breast cancer cells without affecting cell proliferation or viability, and maraviroc decreased pulmonary metastasis in a preclinical mouse model of breast cancer. Taken together, our findings provide evidence for the key role of CCL5/CCR5 in the invasiveness of basal breast cancer cells and suggest that CCR5 antagonists may be used as an adjuvant therapy to reduce the risk of metastasis in patients with the basal breast cancer subtype. Cancer Res; 72(15); 3839–50. ©2012 AACR.

Breast cancer causes the death of 40,000 women in the United States and 410,000 women in the world annually (1). Despite advances in the treatment of the disease, 20% to 30% of patients with early breast cancers will experience relapse with distant metastatic disease (2). In those patients, metastasis is the main cause of death. Patients with basal tumors have increased risk of metastasis and lower survival rate (3, 4). Kennecke and colleagues studied 3,726 patients with breast cancer and reported that the basal tumors have higher frequencies of metastases and reduced time from identification of metastases to death than that of patients with luminal A or B tumors (4). The absence of androgen receptor, estrogen receptor (ER), and HER-2 commonly found in basal breast tumors (5) means that they are unlikely to respond to hormone therapies or HER-2–targeted therapies. Currently, chemotherapy, radiation, and surgery are the only choices for patients with basal breast cancers, but all show poor outcomes (6). The need for a specific targeted therapy for basal breast cancer remains urgent.

Cancer metastasis is regulated by chemokines in the microenvironment (7). Chemokines bind to cell surface receptors that belong to the G-protein–coupled receptor family (GPCR), controlling diverse biologic and pathologic processes from immunosurveillance, inflammation, and cancer. Previous studies of human breast cancer and breast cancer cell lines showed that the chemokine receptors CXCR4 and CCR7 are expressed in breast cancer cells, malignant breast tumors, and metastasis. Their related ligands, CXCL12 (SDF1) and CCL21, are also expressed at the site of metastasis (8). Subsequent studies identified altered expression of CCL5 (RANTES) in patients with breast cancer, correlating with disease progression (9–11).

CCL5 can be expressed and secreted either by breast cancer cells (9–12) or by nonmalignant stromal cells at the primary or metastatic sites (13). However, the roles of CCL5 and its receptors in breast cancer are not fully understood. CCL5 facilitates disease progression by recruiting and modulating the activity of inflammatory cells, which subsequently remodel the tumor microenvironment (14, 15). Accordingly, inhibition of CCR5 by a peptide antagonist reduced leukocyte infiltration and reduced tumor growth after subcutaneous injection of 410.4 cells into immunocompetent mice (16). Studies analyzing the role of CCR5 in breast cancer cell proliferation have used mainly luminal MCF-7 cells and suggest a role for p53. Manes and colleagues reported that abrogation of cell surface CCR5 expression enhanced MCF-7 cell proliferation in vivo in the presence of p53 but did not affect proliferation in xenografts encoding a p53 mutation (17). Murooka and colleagues reported that in CCR5-transfected MCF-7 cells, CCL5 promotes proliferation and survival in an mTOR-dependent manner (18). Autocrine secretion of CCL5 controls migration and invasiveness of human breast cancer cells in vitro (12, 19). Forced expression of CCL5 increased tumor metastasis approximately 1.8-fold in one study of MDA-MB-231 cells but had no effect in the 168 breast cancer cell line (13, 20). Furthermore, other authors have shown that shRNA inhibition of CCL5 expression in murine mammary cancer cells does not affect growth rate or metastatic frequency in vivo (20).

Discrepancies in the reported effects of CCL5 and CCR5 in breast cancer may be due to the different models used. To study whether CCL5/CCR5 signaling is active in specific genetic subtypes of breast cancer, we analyzed the combined expression of CCL5 and CCR5 using a microarray database comprising 2,254 human breast cancer samples from 27 independent studies (21). This report is the first to show that CCL5/CCR5 signaling is preferentially active in the basal and HER-2 subtypes. Using basal-like breast cancer cell lines, we showed that only a subpopulation of cells express CCR5 and respond to CCL5. Importantly, CCR5+ cells displayed increased invasiveness, indicating that CCR5 contributes to the metastatic phenotype of basal breast cancer cells. Therefore, we examined the possibility that CCR5 inhibition may block the invasion and metastasis of basal-like breast cancer cells. We used drugs originally developed to prevent the interaction of CCR5 with the gp120 from HIV-1. The CCR5 antagonist maraviroc (Selzentry) received full U.S. Food and Drug Administration (FDA) approval for use in treatment-naive adults with CCR5-trophic HIV. Vicriviroc (SCH 417690), another CCR5 antagonist, has shown good tolerance and partial therapeutic success in phase II clinical trials for HIV (22). CCR5 inhibition blocked breast cancer cell invasiveness in vitro and efficiently reduced metastatic colonization in vivo. The antimetastatic effect produced by CCR5 inhibition was associated with reduced arrival of breast cancer cells to the target tissue but was independent of changes in cell proliferation or tumor growth. Our findings suggest that CCR5 antagonists may be used to reduce the risk of metastasis in patients with the basal subtype of breast cancer.

Breast cancer patients data set and statistical analysis

A microarray data set that was previously compiled (21) from the public repositories Gene Expression Omnibus (23) and ArrayExpress (24) was used to evaluate CCR5 and CCL5 expression in the context of clinical samples. Samples in this data set were assigned to 5 canonical breast cancer subtypes, including luminal A, luminal B, normal-like, basal, and HER-2–overexpressing disease. The classification of microarray samples among these 5 subtypes was achieved by computing their correlation against an expression profile centroid representative of each subtype and assigning samples to the subtype with the highest corresponding correlation coefficient (25). Samples with a maximum correlation coefficient below 0.3 were considered unclassified. Analysis of CCL5 and CCR5 transcript was then conducted specifically among the luminal A, luminal B, basal, normal-like, and HER-2 subtypes. Differential expression of the averaged gene signature magnitude among these sample subsets was evaluated using 2-tailed Student t test. Kaplan–Meier analysis was used to evaluate survival trends within the sample subsets. Scatter plots of CCL5 versus CCR5 samples were also generated to observe coregulation patterns specific to each subtype. For these scatter plots, gene profiles were median-centered and scaled to unitary SD.

Cell lines and cell culture

MDA-MB-231, MCF-7, and Hs578T cells were maintained in Dulbecco's Modified Eagle's Medium (DMEM) supplemented with 10% FBS. SUM-159 cells (kindly provided by Dr. Stephen Ethier, Wayne State University, Detroit, MI) were maintained in Ham's F-12 supplemented with 4 μg/mL of insulin, 1 μg/mL of hydrocortisone, and 5% FBS. Oncogene-transformed derivatives of MCF-10A cells (MCF10A-NeuT, MCF10A-Src, and MCF10A-Ras; ref. 26) were maintained in DMEM:Ham's F-12 (50 of 50) supplemented with 4 mg/mL of insulin, 10 ng/mL of EGF, and 1 mg/mL of hydrocortisone. A total of 100 μg/mL of each penicillin and streptomycin were included in all media. Cells were cultured in 5% CO2 at 37°C. For in vitro treatments, maraviroc was dissolved in dimethyl sulfoxide (DMSO) and diluted in culture medium. The final concentration of DMSO in treated and control cultures was 0.5%. Vicriviroc was dissolved in culture medium.

Fluorescence-activated cell-sorting analysis

Cell labeling and fluorescence-activated cell-sorting (FACS) analysis for CCR5 were based on prior publications (27) with minor modifications. Before labeling, the cells were blocked with normal mouse IgG (1 of 100) and purified rat anti-mouse Fcγ III/II receptor antibody (1 of 100; Pharmingen) for 30 minutes and then incubated with allophycocyanin (APC)-labeled CCR5 antibody (R&D Systems). All experiments were conducted at 4°C. Sample analysis was conducted on FACSCalibur flow cytometer (BD Biosciences). These data were analyzed with FlowJo software (Tree Star, Inc.).

Invasion assay

The 3-dimensional invasion assay was conducted as previously reported (12). Briefly, 100 μL of 1.67 mg/mL Rat Tail collagen type I (BD Biosciences) was pipetted into the top chamber of a 24-well 8-μm pore Transwell (Corning). The Transwell was incubated at 37°C overnight to allow the collagen to solidify. A total of 30,000 cells were then seeded on the bottom of the Transwell membrane and allowed to attach. Serum-free growth medium was placed into the bottom chamber, whereas 15 ng/mL CCL5 or 5% FBS was used as a chemoattractant in the medium of the upper chamber. The cells were then chemoattracted across the filter through the collagen above for 3 days. Cells were fixed in 4% formaldehyde, permeabilized with 0.2% Triton-X in PBS, and then stained with 40 μg/mL propidium iodide (PI) for 2 hours. Fluorescence was analyzed by confocal z-sections (one section every 20 μm) at ×10 magnification from the bottom of the filter using a Zeiss LSM 510 Meta inverted confocal microscope at the Kimmel Cancer Center Bioimaging Facility.

Intracellular calcium assay

Calcium responses induced either by CCL5 or FBS in human cancer cell lines were monitored under fluorescence confocal microscope as previously reported (28). Briefly, breast cancer cells were seeded in 4-well labtek chambers (Nunc) at 104 cells/cm2 and incubated for 1 day. After 12-hour starvation, cells were labeled by incubating them with 2 mmol/L Fluo-4-AM (Molecular Probes) in HBSS for 30 minutes, washed twice, and incubated for additional 30 minutes before imaging under the microscope. Time-lapse images were collected using a Zeiss LSM 510 Meta inverted confocal microscope with the incubator at 37°C. Relative intracellular Ca2+ concentration was determined by the changes in fluorescent intensity (FI) of Fluo-4-AM upon the addition of CCL5 (60 ng/mL) or FBS (5%) and was calculated as (FIt − FI0)/FI0.

MTT assay

The effects of CCR5 antagonists on cell viability and proliferation rate were estimated using the soluble tetrazolium salt MTT assay (29). MTT is reduced by the mitochondria of viable cells, and the amount of reduced formazan is proportional to the number of viable cells. After 72 hours of exposure to the drugs, cells were incubated with 1 mg/mL of MTT for 90 minutes. Then, the reduced (insoluble and colored) formazan was dissolved in DMSO and measured spectrophotometrically at 570 nm. The effect of CCR5 overexpression in breast cancer cell proliferation was studied in MDA-MB-231 cells transfected with full-length human CCR5 subcloned into pcDNA3.1+/Zeo+ vector (kindly provided by Dr. Eleanor Fish, University of Toronto, Toronto, ON, Canada) and selected with Zeocin (200 μg/mL) as previously described (18). MTT assays were conducted in sextuplicate using 96-well microplates.

Viral cell transduction

A lentiviral vector encoding firefly luciferase 2 (Luc2)-eGFP fusion protein was a generous gift from Dr. Sanjiv S. Gambhir (School of Medicine, Stanford University, Stanford, CA; ref. 30). Lentivirus propagation was conducted following the protocol described by Zahler and colleagues (31). Breast cancer cell lines were transduced at a multiplicity of infection of 20 in the presence of 8 mg/mL polybrene (Sigma) for 24 hours (30, 31).

Experimental metastasis assay and bioluminescence imaging

MB-MDA-231 cells expressing Luc2-eGFP (called MDA.pFLUG for the rest of the article) were detached with a nonenzymatic cell dissociation buffer (4 mmol/L EDTA in Ca2+ and Mg2+-free PBS), resuspended in Dulbecco's PBS without Ca2+ and Mg2+ and immediately injected into the tail vein of 8-week-old, female nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice (NCI, Bethesda MD). Each mouse received 106 cells. Mice were treated by oral gavage with maraviroc (8 mg/kg every 12 hours) or vehicle (5% DMSO in acidified water; ref. 32). Treatment was started immediately after injection or 10 days later for the experiments analyzing the proliferation of established metastasis. For in vivo bioluminescence imaging (BLI), mice were given an intraperitoneal (i.p.) injection with 200 μL of d-luciferin (30 mg/mL). Mice were anesthetized with isoflurane (2% in 1 L/min oxygen), and bioluminescence images were acquired 10 to 15 minutes after d-luciferin injection using the IVIS XR system (Caliper Life Sciences). Acquisition times ranged from 10 seconds (for later time points) to 5 minutes (for early time points). Data are expressed as total photon flux and were analyzed using Living Image 3.0 software (Caliper Life Sciences). For ex vivo BLI, d-luciferin was diluted in PBS to a final concentration of 300 μg/mL and used to soak freshly isolated lungs for 2 to 3 minutes before imaging. Some lungs were stained with India ink, as previously reported (33), or processed and stained with hematoxylin and eosin to corroborate the presence of pulmonary tumors. For homing assays, mice were euthanized 24 hours after the intravenous injection of MDA.pFULG cells. Lungs were perfused with PBS, fixed with freshly prepared formaldehyde (4% in PBS), and frozen in optimum cutting temperature (Sakura Finetek). Cryosections (10 μm) were counterstained with 4′,6-diamidino-2-phenylindole analyzed by confocal microscopy. Animal experiments were approved by the Thomas Jefferson University's Institutional Animal Care and Use Committee.

Reagents and antibodies

CCL5 (catalog no. 278-RN) and anti-CCR5 APC antibody (catalog no. FAB1802A) were purchased from R&D Systems. A rabbit anti-human CCR5 polyclonal antibody (GenScipt; catalog no. A00979) was used for immunohistochemical staining. Rat tail collagen type I was purchased from BD Biosciences. Vicriviroc and maraviroc were obtained from Selleck Chemicals. Luciferin was obtained from Gold Biotechnology.

Active CCL5/CCR5 signaling in basal breast cancer

To examine the relative abundance of CCL5 and its receptor CCR5 by genetic subtype, interrogation was conducted within a combined microarray database comprising 2,254 human breast cancer samples from 27 independent studies (21). The relative abundances of CCL5 and CCR5 were significantly increased in the basal and HER-2 subtypes compared with the normal-like, luminal A and luminal B subtypes (Fig. 1A). The increased expression of CCL5 and CCR5 correlated positively in individual breast cancer samples and the correlation was highly significant in the basal and HER-2 subtypes (Fig. 1B). The proportion of patients with a CCL5/CCR5-positive signature was more than 58% in the basal and HER-2 subtypes (Fig. 1C). In agreement with previous reports, clinical information of the cases in this database showed that the probability to develop metastasis is increased in the basal, luminal B, and HER-2 subtypes (Fig. 1D).

Figure 1.

CCL5 and CCR5 are overexpressed in the basal breast cancer subtype. A, heatmap of the expression of CCL5 and its receptor CCR5 in samples from patients with breast cancer divided by genetic subtype based on their gene expression pattern. The relative abundances of CCL5 and CCR5 are increased in patients with the basal and HER-2 subtypes. B, scatter plots and correlation analysis (Student t test) of the expression of CCL5 and CCR5 among the breast cancer molecular subtypes. C, quantification of the proportions of the samples overexpressing CCL5 and CCR5 (red fraction of the bar) displayed in B. The number of samples in each subtype is indicated at the top of the bar. D, metastasis-free Kaplan–Meier plots and log-rank analysis for the different genetic subtypes in the analyzed database (described in Materials and Methods). As reported elsewhere, patients with the basal or HER-2 subtypes display increased probability to form metastasis.

Figure 1.

CCL5 and CCR5 are overexpressed in the basal breast cancer subtype. A, heatmap of the expression of CCL5 and its receptor CCR5 in samples from patients with breast cancer divided by genetic subtype based on their gene expression pattern. The relative abundances of CCL5 and CCR5 are increased in patients with the basal and HER-2 subtypes. B, scatter plots and correlation analysis (Student t test) of the expression of CCL5 and CCR5 among the breast cancer molecular subtypes. C, quantification of the proportions of the samples overexpressing CCL5 and CCR5 (red fraction of the bar) displayed in B. The number of samples in each subtype is indicated at the top of the bar. D, metastasis-free Kaplan–Meier plots and log-rank analysis for the different genetic subtypes in the analyzed database (described in Materials and Methods). As reported elsewhere, patients with the basal or HER-2 subtypes display increased probability to form metastasis.

Close modal

To determine the gene expression signaling pathway associated with enrichment of CCR5 and CCL5, GSEA analysis using KEGG and GO was conducted of these tumor samples (Supplementary Fig. S1A). These studies showed enrichment for gene expression of pathways including lymphocyte activation, Janus-activated kinase (JAK)-STAT signaling, and Toll-like receptor activation (Supplementary Fig. S1A). The receptors for CCL5 include CCR1 and CCR3. Increased expression of CCL5 associated with increased CCR1, but not CCR3, in the basal and HER-2 genetic tumor type (Supplementary Fig. S1B and S1C). In ER-negative patients receiving chemotherapy, there was an insignificant trend toward reduced metastasis-free survival and relapse-free survival in the increased CCR5 population, compared with the population with reduced CCR5 expression (Supplementary Fig. S1D). A comparison of expression levels for CCL5 versus CCR5, CCR1, and CCR3, comparing normal breast with breast cancer showed increased correlation between receptor and ligand expression levels in tumors compared with healthy breast tissue (Supplementary Fig. S2).

CCL5 promotes breast cancer Ca2+ signaling and cellular invasion

We chose to use as models in our studies 3 human breast cancer cell lines with a basal phenotype and molecular signature: MDA-MB-231, Hs578T, and SUM-159 (34–37). Analysis of CCR5 expression by FACS showed that a small subpopulation of cells were positive for the receptor in all 3 cell lines (Fig. 2A for MDA-MB-231 and Supplementary Fig. S3A and S3C for Hs578T and SUM-159). Because CCR5 activation induces calcium flux (38, 39), we assessed the activation of calcium signaling by CCL5. Addition of CCL5 to the cultures induced immediate calcium fluxes in a subpopulation of cells (Fig. 2B for MDA-MB-231 and Supplementary Fig. S3B and S3D for Hs578T and SUM-159), providing evidence that CCR5 is functional in basal breast cancer cells. As a positive control, the same cultures were exposed to 5% FBS (40). Calcium flux, assessed by relative fluorescence intensity, increased in more than 95% of the cells after FBS addition (Fig. 2B and Supplementary Fig. S3B and S3D). To further distinguish CCL5-dependent signaling, SUM159 cells were stably transduced with a CCR5 expression vector and the Ca+2 response to CCL5 versus FBS was conducted (Supplementary Figs. S4C and D vs. S4E and F). CCR5 induced Ca+2 signaling in the CCR5-overexpressing cells, whereas both lines responded similarly to FBS induced Ca+2 activation (Supplementary Fig. S4).

Figure 2.

Human breast cancer cell lines that express CCR5 respond to CCL5. A, flow cytometric histograms of the CCR5 expression in MDA-MB-231 breast cancer cells identified a subpopulation of CCR5+ cells. B, induction of calcium signaling in cells loaded with Fluo-4-AM before the sequential addition of CCL5 (60 μg/mL) and FBS (5%). A fraction of cells responded to CCL5 (closed arrowheads in the middle of micrographs) whereas the rest did not (open arrowheads). The average changes in fluorescence on 5 responsive (green line) and 5 nonresponsive (red line) cells are represented in the far right graphs. Data shown are representative of 3 to 5 independent experiments for each cell line. Bar, 100 μm. C, 3D invasion into collagen gels by breast cancer cell lines, using CCL5 (15 μg/mL) as chemoattractant. D, mean distances of invasion ± SEM from 3 independent experiments. E, 3D invasion assays, and their corresponding quantification (F, mean ± SEM, n = 3), for MCF-10A cells and MCF-10A-NeuT, -Ras, and -Src derivatives showing that CCL5-induced invasion is activated by oncogenic transformation. G, CCR5+ cells display increased invasiveness. CCR5+ and CCR5 subpopulations from SUM-159 cell line were isolated by FACS and invasion into collagen gels was evaluated using FBS as chemoattractant. Quantification is shown in H as mean ± SEM of 2 independent experiments. Statistical analysis was conducted using the Student t test.

Figure 2.

Human breast cancer cell lines that express CCR5 respond to CCL5. A, flow cytometric histograms of the CCR5 expression in MDA-MB-231 breast cancer cells identified a subpopulation of CCR5+ cells. B, induction of calcium signaling in cells loaded with Fluo-4-AM before the sequential addition of CCL5 (60 μg/mL) and FBS (5%). A fraction of cells responded to CCL5 (closed arrowheads in the middle of micrographs) whereas the rest did not (open arrowheads). The average changes in fluorescence on 5 responsive (green line) and 5 nonresponsive (red line) cells are represented in the far right graphs. Data shown are representative of 3 to 5 independent experiments for each cell line. Bar, 100 μm. C, 3D invasion into collagen gels by breast cancer cell lines, using CCL5 (15 μg/mL) as chemoattractant. D, mean distances of invasion ± SEM from 3 independent experiments. E, 3D invasion assays, and their corresponding quantification (F, mean ± SEM, n = 3), for MCF-10A cells and MCF-10A-NeuT, -Ras, and -Src derivatives showing that CCL5-induced invasion is activated by oncogenic transformation. G, CCR5+ cells display increased invasiveness. CCR5+ and CCR5 subpopulations from SUM-159 cell line were isolated by FACS and invasion into collagen gels was evaluated using FBS as chemoattractant. Quantification is shown in H as mean ± SEM of 2 independent experiments. Statistical analysis was conducted using the Student t test.

Close modal

We next assessed the effect of CCR5 activation on breast cancer cell invasion using 3D migration assays. CCL5 induced invasion of the basal MDA-MB-231, Hs578T, SUM-159 but not the luminal MCF-7 cells (Fig. 2C and D). CCL5 promoted invasion of MCF-10A cells engineered to express either NeuT, H-Ras, or c-Src oncogenes, compared with MCF10A vector-transduced cells (Fig. 2E and F), suggesting that CCL5 responsiveness may be acquired during transformation and requires specific cooperative oncogenic signals. The finding that CCL5 induced cellular invasion led us to examine the migratory capacity of CCR5+ cells versus that of CCR5 cells. Within the same SUM-159 breast cancer cell line, CCR5+ cells showed an approximately 40-fold greater cellular invasiveness (Fig. 2G and H), indicating that the expression of CCR5 correlates with a proinvasive phenotype.

CCR5 antagonists block breast cancer calcium signaling and cell invasion

The importance of CCR5 in HIV infection led to the development of different drugs that target this receptor. We therefore examined whether the CCR5 antagonists maraviroc and vicriviroc were capable of blocking the CCL5/CCR5 signaling in basal breast cancer cells. Both CCR5 antagonists blocked CCL5-induced calcium mobilization. In MDA-MB-231 cells, maraviroc and vicriviroc inhibited calcium responses by 65% and 90%, respectively (Fig. 3A and B). Similar observations were made with both drugs in Hs578T cells (Fig. 3C and D), indicating that CCR5 expressed in different basal breast cancer cells is sensitive to pharmacologic inhibition.

Figure 3.

CCR5 antagonists block CCL5-induced calcium signaling. A, intensity versus time analysis of Fluo-4 AM–loaded MDA-MB-231 cells treated with the CCR5 antagonists maraviroc or vicriviroc (100 nmol/L) for 30 minutes before the addition of CCL5 (60 μg/mL). Micrographs illustrate the axis (x-x') of the pseudoline scan plot. Those axes were used to construct the adjacent intensity versus time plots. B, comparison of the fraction of cells with increased fluorescence intensity upon addition of CCL5. C, CCL5-induced calcium signaling was also blocked by CCR5 antagonists in Hs578T cells. The corresponding quantification is shown in D. Data in B and D are mean ± SEM of 3 to 4 independent experiments. Statistical analysis was conducted using the Student t test.

Figure 3.

CCR5 antagonists block CCL5-induced calcium signaling. A, intensity versus time analysis of Fluo-4 AM–loaded MDA-MB-231 cells treated with the CCR5 antagonists maraviroc or vicriviroc (100 nmol/L) for 30 minutes before the addition of CCL5 (60 μg/mL). Micrographs illustrate the axis (x-x') of the pseudoline scan plot. Those axes were used to construct the adjacent intensity versus time plots. B, comparison of the fraction of cells with increased fluorescence intensity upon addition of CCL5. C, CCL5-induced calcium signaling was also blocked by CCR5 antagonists in Hs578T cells. The corresponding quantification is shown in D. Data in B and D are mean ± SEM of 3 to 4 independent experiments. Statistical analysis was conducted using the Student t test.

Close modal

To evaluate the functional relevance of CCR5 in cellular migration and invasion, we tested the effects of maraviroc and vicriviroc in 3D invasion assays. Using 2 different cell lines, we found that both CCR5 antagonists inhibited FBS-induced breast cancer cell invasion at the clinically relevant concentration of 100 nmol/L (Fig. 4A–D). Thus, the proinvasive effect of CCR5 can be abrogated by using specific antagonists.

Figure 4.

CCR5 antagonists block FBS-induced breast cancer cell invasion. 3D reconstruction of FBS-induced invasion into collagen gels by Hs578T (A) or SUM-159 (C) breast cancer cells in presence of CCR5 antagonists (100 nmol/L). The corresponding quantifications (mean ± SEM, n = 3) and analysis (Bonferroni t test) are displayed in B and D.

Figure 4.

CCR5 antagonists block FBS-induced breast cancer cell invasion. 3D reconstruction of FBS-induced invasion into collagen gels by Hs578T (A) or SUM-159 (C) breast cancer cells in presence of CCR5 antagonists (100 nmol/L). The corresponding quantifications (mean ± SEM, n = 3) and analysis (Bonferroni t test) are displayed in B and D.

Close modal

CCR5 inhibition blocks breast cancer metastasis in vivo

In view of the finding that CCR5 inhibition by CCR5 antagonists reduced calcium signaling and cell invasion, we determined the in vivo effect of maraviroc on lung metastasis. We used MDA-MB-231 cells transduced within the Luc2-eGFP lentiviral vector (MDA.pFULG cells) in an experimental metastasis model. The Luc2 gene is a codon-optimized version of Luc and cells expressing this reporter were 10 to 100 times brighter than the unmodified Luc gene (30). After injection of MDA.pFULG cells into the tail vein of mice, noninvasive BLI enabled the early detection of breast cancer metastasis (41). Weekly BLI was conducted for 5 weeks and the radiance antemortem was used as a surrogate measurement of tumor burden. Mice treated with maraviroc (8 mg/kg twice daily) showed a significant reduction in both the number and the size of pulmonary metastases compared with vehicle-treated mice (Fig. 5A and B, Supplementary Fig. S5). To avoid the possibility that metastases were missed because of inappropriate imaging, we conducted ex vivo imaging, India ink staining (Fig. 5C), and histology (Fig. 5E) of the lungs. Histologic analysis corroborated that tumor burden corresponds to bioluminescence, as previously shown (30). Metastatic tumors were still detectable in 50% of the maraviroc-treated mice, but their mean size was reduced by 65% (Fig. 5D and F). Interestingly, analysis of CCR5 expression in lungs from control mice showed an 8-fold enrichment of the CCR5+ fraction (Supplementary Fig. S6). Collectively, these results provide evidence that CCR5 antagonists reduce breast cancer metastasis in vivo.

Figure 5.

The CCR5 antagonist maraviroc inhibits lung metastases in vivo. A, MDA-MB-231 cells transduced with Luc2-eGFP fusion protein were injected into the tail vein of NOD/SCID mice and the in vivo bioluminescent signal was quantified weekly. Representative in vivo images of vehicle- or maraviroc-treated (8 mg/kg every 12 hours) mice are shown in A. Quantification (mean ± SEM, n = 6) of BLI in the control (red line) and treated groups (blue line) are portrayed in B. Statistical comparison (*, P = 0.048) was carried out using Student t test with Welch correction for heterogeneous variances. C, the presence of pulmonary tumors and the differences between treatments were corroborated by ex vivo imaging (left) and India ink staining (right). D, the fraction of mice with metastatic tumors was significantly larger in the control group (P < 0.0001, Fisher exact test). E, histologic analysis (hematoxylin and eosin staining, ×100) and the corresponding quantification (F) of the area covered by metastatic tumors in lung slides. Tumor area was quantified with the Nikon Elements BR 3.0 software analyzing at low magnification (×40) 2 random fields of 2 different histologic sections (separated 600 μm from each other) per mouse. Statistical analysis was conducted using the Student t test with Welch correction for heterogeneous variances (n = 33 and 12 for control and treated groups, respectively).

Figure 5.

The CCR5 antagonist maraviroc inhibits lung metastases in vivo. A, MDA-MB-231 cells transduced with Luc2-eGFP fusion protein were injected into the tail vein of NOD/SCID mice and the in vivo bioluminescent signal was quantified weekly. Representative in vivo images of vehicle- or maraviroc-treated (8 mg/kg every 12 hours) mice are shown in A. Quantification (mean ± SEM, n = 6) of BLI in the control (red line) and treated groups (blue line) are portrayed in B. Statistical comparison (*, P = 0.048) was carried out using Student t test with Welch correction for heterogeneous variances. C, the presence of pulmonary tumors and the differences between treatments were corroborated by ex vivo imaging (left) and India ink staining (right). D, the fraction of mice with metastatic tumors was significantly larger in the control group (P < 0.0001, Fisher exact test). E, histologic analysis (hematoxylin and eosin staining, ×100) and the corresponding quantification (F) of the area covered by metastatic tumors in lung slides. Tumor area was quantified with the Nikon Elements BR 3.0 software analyzing at low magnification (×40) 2 random fields of 2 different histologic sections (separated 600 μm from each other) per mouse. Statistical analysis was conducted using the Student t test with Welch correction for heterogeneous variances (n = 33 and 12 for control and treated groups, respectively).

Close modal

CCR5 antagonist impairs lung colonization but not cell proliferation or tumor growth

We determined whether the reduction in metastatic tumors by maraviroc involved changes in cellular proliferation and/or target organ colonization. We analyzed the effect of CCR5 inhibition on cell viability and proliferation both in vitro and in vivo. Maraviroc or vicriviroc treatment of MDA-MB-231 cells for 48 hours did not affect the MTT reduction, which was used as a surrogate measurement of cancer cell number (Fig. 6A). In agreement, overexpression of CCR5 in MDA-MB-231 cells did not modify their proliferation rate compared with cells transfected with the empty vector (Fig. 6B and C). Finally, maraviroc treatment of mice with established pulmonary metastasis did not modify tumor growth (Fig. 6D and E), indicating that CCR5 activation does not promote the proliferation of basal breast cancer cells in vitro nor in the pulmonary microenvironment of immunocompromised mice.

Figure 6.

Maraviroc reduces lung colonization but does not modify cell proliferation. A, effect of CCR5 antagonist on breast cancer cell viability. MDA-MB-231 cells were exposed to increasing concentrations of maraviroc (inverted triangles) or vicriviroc (squares) for 48 hours and the cell viability was evaluated by MTT assay. Graph is from a representative experiment carried out by sextuplicate. No statistical differences were found (ANOVA) in 3 independent experiments. B, CCR5 expression in MDA-MB-231 cells stably transfected with pcDNA3.1+/Zeo+ (MDA.Vector) or human CCR5 cloned into pCDNA3+Zeo+ (MDA.CCR5). C, comparison of in vitro proliferation rates of MDA.Vector versus MDA.CCR5 showed no differences (ANOVA). Representative experiment from 2 carried out by sextuplicate. D, to evaluate the in vivo effect of maraviroc on growth of established metastasis, treatment of mice was initiated 10 days after injection of MDA.pFULG cells as illustrated. E, quantification (mean ± SEM, n = 5) of in vivo BLI in the control (red) and treated groups (blue) showed no differences in the growth rate. F, schema of the experimental design used to evaluate CCR5 role in lung colonization. G, representative confocal images and quantification (H) of the number of eGFP+ cells in lungs 24 hours after injection of MDA.pFULG cells. Cells expressing eGFP were counted in 3 random fields of 2 different histologic sections (separated 700 μm from each other) per mouse (n = 5 mice per group). Statistical analysis was conducted using Student t test. Bar in micrographs, 100 μm.

Figure 6.

Maraviroc reduces lung colonization but does not modify cell proliferation. A, effect of CCR5 antagonist on breast cancer cell viability. MDA-MB-231 cells were exposed to increasing concentrations of maraviroc (inverted triangles) or vicriviroc (squares) for 48 hours and the cell viability was evaluated by MTT assay. Graph is from a representative experiment carried out by sextuplicate. No statistical differences were found (ANOVA) in 3 independent experiments. B, CCR5 expression in MDA-MB-231 cells stably transfected with pcDNA3.1+/Zeo+ (MDA.Vector) or human CCR5 cloned into pCDNA3+Zeo+ (MDA.CCR5). C, comparison of in vitro proliferation rates of MDA.Vector versus MDA.CCR5 showed no differences (ANOVA). Representative experiment from 2 carried out by sextuplicate. D, to evaluate the in vivo effect of maraviroc on growth of established metastasis, treatment of mice was initiated 10 days after injection of MDA.pFULG cells as illustrated. E, quantification (mean ± SEM, n = 5) of in vivo BLI in the control (red) and treated groups (blue) showed no differences in the growth rate. F, schema of the experimental design used to evaluate CCR5 role in lung colonization. G, representative confocal images and quantification (H) of the number of eGFP+ cells in lungs 24 hours after injection of MDA.pFULG cells. Cells expressing eGFP were counted in 3 random fields of 2 different histologic sections (separated 700 μm from each other) per mouse (n = 5 mice per group). Statistical analysis was conducted using Student t test. Bar in micrographs, 100 μm.

Close modal

On a different in vivo experiment, we examined the effect of maraviroc on breast cancer cell homing to lungs. To reach a steady-state concentration in plasma and tissues, mice were given 10 administrations of maraviroc (twice a day for 5 days) before the intravenous injection of MDA.pFULG cells (Fig. 6F). Inoculation of equal numbers of MDA.pFULG cells in control and treated groups was corroborated by BLI immediately after injection. Maraviroc reduced the number of eGFP+ cells in the lungs by 40% (Fig 6G and H), suggesting that the in vivo antimetastatic effect of maraviroc is caused by a reduction in the number of cancer cells that colonize the target organ from the circulation.

The current studies show for the first time that: (i) enrichment of CCL5/CCR5 expression occurs in patients with basal and Her2 positive genetic subtypes of breast cancer; (ii) oncogenic transformation of immortalized human breast cells by distinct oncogenes induces CCL5 responsiveness; and (iii) maraviroc, an FDA-approved drug for the treatment of CCR5-trophic HIV infection, reduce metastatic tumor burden in vivo.

Previous studies showed that CCL5 levels are elevated in breast primary and metastatic tumors (9–11), suggesting a role of CCL5 in the acquisition of malignancy. We report here that increased expression of CCL5 and CCR5 are associated and that CCL5/CCR5 expression levels are different among the different genetic subtypes of breast cancer. Increased expression of CCL5 and CCR5 is found in the basal and HER-2 subtypes. In agreement, increased CCL5 expression has been found predominantly in ER-negative patients (42). Increased CCL5 also correlated with increased CCR1 in basal and Her2 genetic subtypes of breast cancer. A trend toward reduced metastasis-free survival and relapse-free survival was observed among the CCR5-overexpressing tumors in patients who received chemotherapy.

Given the aggressive clinical behavior of basal breast cancer and the lack of targeted therapies for it, we evaluated the importance of the CCL5/CCR5 axis in invasion and metastasis in the human breast cancer cell lines MDA-MB-231, Hs578T and SUM-159. These cell lines reflect the clinicopathologic features of the basal subtype of breast cancer (including the lack of HER-2, ER, and progesterone receptor), a basal-like molecular signature, the activation of specific signaling pathways (e.g., hypoxic or EGF receptor responses) and overexpression of epithelial–mesenchymal transition proteins (FN, VIM, and matrix metalloproteinase 2; refs. 34–37). Only a small fraction of cells within the cell lines used in this study expressed CCR5 as evaluated by FACS analysis. Our findings are consistent with studies by Müller and colleagues who showed CCR5 expression in MDA-MB-231 by quantitative real-time PCR (8). Our studies confirmed the expression of CCR5 in MDA-MB-23 cells by reverse transcriptase PCR and showed the presence of the CCR5 protein by FACS analysis (Supplementary Fig. S7), and showed that CCR5 immunohistochemical staining was localized primarily to the breast cancer epithelial cell, compared with normal breast tissue (Supplementary Fig. S8).

We showed that CCL5 activates calcium flux in basal-like human breast cancer cells, as previously described in cells of the immune system (39, 43) and CCR5-transfected cells (27, 44, 45). By using the selective CCR5-antagonists maraviroc and vicriviroc (both with IC50 below 30 nmol/L; refs. 44, 45), we showed that CCL5-activated signaling is mediated by CCR5. However, the fraction of CCL5-responsive cells (10% and 12% for MDA-MB-231 and Hs578T cells, respectively) is higher than the percentage of CCR5-expressing cells determined by FACS. This may be due to the greater sensitivity of the Ca2+ activation assays compared with the sensitivity of analysis by FACS. In addition, CCL5-induced calcium redistribution is not completely blocked by CCR5 antagonists. This may be caused by the expression of other receptors to CCL5, namely CCR1 and CCR3. CCR5 has been identified as the main CCL5 receptor in MDA-MB-231 cells (13) and CCR1 and CCR3 transcripts are absent in both MDA-MB-231 or Hs578T cell lines (8) and breast tumor samples (11). We were able to detect CCR1 and CCR3 by FACS (Supplementary Fig. S7), suggesting a possible mechanism for the incomplete response to the CCR5 antagonist.

We observed that the subpopulation of CCR5+ cells displayed increased invasiveness, indicating that CCR5 favors cell migration and invasion in basal-like breast cancer cells. The failure of luminal-like MCF-7 cells to respond to CCL5 is in agreement with previous publications (12). Our studies also showed that CCR5 inhibition with either maraviroc or vicriviroc reduced in vitro FBS-induced breast cancer cellular invasion without affecting cellular viability. The finding that CCR5 antagonists block FBS-induced invasion is novel and suggested that CCR5 activation contribute to the production of metastasis in vivo where different chemotactic and growth signals are present. The mechanisms involved in CCR5 regulation of FBS-activated invasiveness are uncharacterized but they may include heterodimerization and ligand affinity regulation of other GPCRs (46), or the transactivation of growth factor receptor- (47) or integrin-mediated signaling (48), as described in noncancerous cells.

The in vivo antimetastatic effect of maraviroc was shown by injecting MDA.pFULG cells into the circulation of immunodeficient mice and treating them with clinically relevant doses of the drug. In humans, oral doses of 300 mg produce an average Cmax of 1,200 nmol/L (49), whereas in mice 16 mg/kg produce an average Cmax of 1,045 nmol/L (32). Because the drug is taken twice a day in the clinical setting, we administered 16 mg/kg/d divided into 2 doses during our experiments. Maraviroc significantly reduced the pulmonary tumor burden. Although it has been proposed that pharmacologic CCR5 inhibition may be beneficial for patients with breast cancer, to our knowledge this is the first study showing that systemic administration of a CCR5 antagonist reduces metastatic colonization of basal breast cancer cells.

The antimetastatic effect of maraviroc is not caused by alterations in growth of established metastasis. CCR5 activation by CCL5 drives proliferation in CCR5-transfected MCF-7 breast cancer cells (18) and prostate cancer cells (50), but this study and others (13) showed that the CCL5/CCR5 axis does not play a role in cell proliferation or survival in the basal-like MDA-MB-231 cells. Furthermore, inhibition of CCR5 surface expression through a dominant-negative form of CCR5 (CCR5Δ32) in MDA-MB-231 cells does not change in vivo proliferation or apoptotic response (17). On the other hand, we found that maraviroc reduces lung colonization by MDA.pFULG cancer cells. This result is consistent with previous studies in which inhibition of CCR5 expression within breast cancer cells or administration of anti-CCL5 neutralizing antibody to tumor-bearing mice reduced the enhanced metastatic capability induced by coinjection of mesenchymal stem cells (MSC; ref. 13). The authors identified cancer cell extravasation as the crucial metastatic step affected by CCL5/CCR5 inhibition (13). Together, these data support a role for CCR5 antagonists in blocking the ability of basal breast cancer cells to reach the metastatic sites instead of inhibiting their proliferation or survival after arrival. Blocking the homing of cancer cells to metastatic sites is a desirable characteristic in a true antimetastatic drug (51). Therefore, CCR5 antagonists may be useful as adjuvant therapy for breast basal tumors with CCR5 overexpression or other tumor types where CCR5 promotes metastasis, such as prostate cancer (50) or gastric cancer (52). Our preclinical studies will expedite the path toward the clinical use of CCR5 antagonists as new treatments for dissemination of basal breast cancer.

R.G. Pestell has ownership interest (including patents) in Prostagene, LLC. No potential conflicts of interest were disclosed by the other authors. The Department specifically disclaims responsibility for any analysis, interpretations, or conclusions.

Conception and design: M. Velasco-Velazquez, X. Jiao, M.P. Lisanti, R.G. Pestell

Development of methodology: M. Velasco-Velazquez, X. Jiao

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Velasco-Velazquez, X. Jiao, M. De La Fuente, T.G. Pestell

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Velasco-Velazquez, X. Jiao, M. De La Fuente, T.G. Pestell, A. Ertel, M.P. Lisanti, R.G. Pestell

Writing, review, and/or revision of the manuscript: M. Velasco-Velazquez, T.G. Pestell, M.P. Lisanti, R.G. Pestell

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Velasco-Velazquez, X. Jiao, R.G. Pestell

Study supervision: M. Velasco-Velazquez, X. Jiao, R.G. Pestell

The authors thank Jeannine Moore and David Strahan for assistance in the preparation of the manuscript.

This work was supported in part by PASPA-UNAM (M. Velasco-Velazquez), NIH grants R01CA070896, R01CA075503, R01CA132115, R01CA107382, R01CA086072 (R.G. Pestell), R01CA120876 (M.P. Lisanti), the Kimmel Cancer Center NIH Cancer Center Core grant P30CA056036 (R.G. Pestell), generous grants from the Dr. Ralph and Marian C. Falk Medical Research Trust and the Margaret Q. Landenberger Research Foundation, and a grant from Pennsylvania Department of Health (R.G. Pestell).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Parkin
DM
,
Fernandez
LM
. 
Use of statistics to assess the global burden of breast cancer
.
Breast J
2006
;
12
Suppl 1
:
S70
80
.
2.
Group EBCTC
. 
Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials
.
Lancet
2005
;
365
:
1687
717
.
3.
Meyers
MO
,
Klauber-Demore
N
,
Ollila
DW
,
Amos
KD
,
Moore
DT
,
Drobish
AA
, et al
Impact of breast cancer molecular subtypes on locoregional recurrence in patients treated with neoadjuvant chemotherapy for locally advanced breast cancer
.
Ann Surg Oncol
2011
;
18
:
2851
7
.
4.
Kennecke
H
,
Yerushalmi
R
,
Woods
R
,
Cheang
MC
,
Voduc
D
,
Speers
CH
, et al
Metastatic behavior of breast cancer subtypes
.
J Clin Oncol
2010
;
28
:
3271
7
.
5.
Perou
CM
,
Sorlie
T
,
Eisen
MB
,
van de Rijn
M
,
Jeffrey
SS
,
Rees
CA
, et al
Molecular portraits of human breast tumours
.
Nature
2000
;
406
:
747
52
.
6.
Reis-Filho
JS
,
Lakhani
SR
. 
Breast cancer special types: why bother?
J Pathol
2008
;
216
:
394
8
.
7.
Kakinuma
T
,
Hwang
ST
. 
Chemokines, chemokine receptors, and cancer metastasis
.
J Leukoc Biol
2006
;
79
:
639
51
.
8.
Muller
A
,
Homey
B
,
Soto
H
,
Ge
N
,
Catron
D
,
Buchanan
ME
, et al
Involvement of chemokine receptors in breast cancer metastasis
.
Nature
2001
;
410
:
50
6
.
9.
Luboshits
G
,
Shina
S
,
Kaplan
O
,
Engelberg
S
,
Nass
D
,
Lifshitz-Mercer
B
, et al
Elevated expression of the CC chemokine regulated on activation, normal T cell expressed and secreted (RANTES) in advanced breast carcinoma
.
Cancer Res
1999
;
59
:
4681
7
.
10.
Niwa
Y
,
Akamatsu
H
,
Niwa
H
,
Sumi
H
,
Ozaki
Y
,
Abe
A
. 
Correlation of tissue and plasma RANTES levels with disease course in patients with breast or cervical cancer
.
Clin Cancer Res
2001
;
7
:
285
9
.
11.
Zhang
Y
,
Yao
F
,
Yao
X
,
Yi
C
,
Tan
C
,
Wei
L
, et al
Role of CCL5 in invasion, proliferation and proportion of CD44+/CD24- phenotype of MCF-7 cells and correlation of CCL5 and CCR5 expression with breast cancer progression
.
Oncol Rep
2009
;
21
:
1113
21
.
12.
Jiao
X
,
Katiyar
S
,
Willmarth
NE
,
Liu
M
,
Ma
X
,
Flomenberg
N
, et al
c-Jun induces mammary epithelial cellular invasion and breast cancer stem cell expansion
.
J Biol Chem
2010
;
285
:
8218
26
.
13.
Karnoub
AE
,
Dash
AB
,
Vo
AP
,
Sullivan
A
,
Brooks
MW
,
Bell
GW
, et al
Mesenchymal stem cells within tumour stroma promote breast cancer metastasis
.
Nature
2007
;
449
:
557
63
.
14.
Locati
M
,
Deuschle
U
,
Massardi
ML
,
Martinez
FO
,
Sironi
M
,
Sozzani
S
, et al
Analysis of the gene expression profile activated by the CC chemokine ligand 5/RANTES and by lipopolysaccharide in human monocytes
.
J Immunol
2002
;
168
:
3557
62
.
15.
Robinson
SC
,
Scott
KA
,
Balkwill
FR
. 
Chemokine stimulation of monocyte matrix metalloproteinase-9 requires endogenous TNF-alpha
.
Eur J Immunol
2002
;
32
:
404
12
.
16.
Robinson
SC
,
Scott
KA
,
Wilson
JL
,
Thompson
RG
,
Proudfoot
AE
,
Balkwill
FR
. 
A chemokine receptor antagonist inhibits experimental breast tumor growth
.
Cancer Res
2003
;
63
:
8360
5
.
17.
Manes
S
,
Mira
E
,
Colomer
R
,
Montero
S
,
Real
LM
,
Gomez-Mouton
C
, et al
CCR5 expression influences the progression of human breast cancer in a p53-dependent manner
.
J Exp Med
2003
;
198
:
1381
9
.
18.
Murooka
TT
,
Rahbar
R
,
Fish
EN
. 
CCL5 promotes proliferation of MCF-7 cells through mTOR-dependent mRNA translation
.
Biochem Biophys Res Commun
2009
;
387
:
381
6
.
19.
Stormes
KA
,
Lemken
CA
,
Lepre
JV
,
Marinucci
MN
,
Kurt
RA
. 
Inhibition of metastasis by inhibition of tumor-derived CCL5
.
Breast Cancer Res Treat
2005
;
89
:
209
12
.
20.
Jayasinghe
MM
,
Golden
JM
,
Nair
P
,
O'Donnell
CM
,
Werner
MT
,
Kurt
RA
. 
Tumor-derived CCL5 does not contribute to breast cancer progression
.
Breast Cancer Res Treat
2008
;
111
:
511
21
.
21.
Ertel
A
,
Dean
JL
,
Rui
H
,
Liu
C
,
Witkiewicz
AK
,
Knudsen
KE
, et al
RB-pathway disruption in breast cancer: differential association with disease subtypes, disease-specific prognosis and therapeutic response
.
Cell Cycle
2010
;
9
:
4153
63
.
22.
Wilkin
TJ
,
Su
Z
,
Krambrink
A
,
Long
J
,
Greaves
W
,
Gross
R
, et al
Three-year safety and efficacy of vicriviroc, a CCR5 antagonist, in HIV-1-infected treatment-experienced patients
.
J Acquir Immune Defic Syndr
2010
;
54
:
470
6
.
23.
Barrett
T
,
Troup
DB
,
Wilhite
SE
,
Ledoux
P
,
Rudnev
D
,
Evangelista
C
, et al
NCBI GEO: mining tens of millions of expression profiles–database and tools update
.
Nucleic Acids Res
2007
;
35
:
D760
5
.
24.
Brazma
A
,
Parkinson
H
,
Sarkans
U
,
Shojatalab
M
,
Vilo
J
,
Abeygunawardena
N
, et al
ArrayExpress–a public repository for microarray gene expression data at the EBI
.
Nucleic Acids Res
2003
;
31
:
68
71
.
25.
Hu
Z
,
Fan
C
,
Oh
DS
,
Marron
JS
,
He
X
,
Qaqish
BF
, et al
The molecular portraits of breast tumors are conserved across microarray platforms
.
BMC Genomics
2006
;
7
:
96
.
26.
Liu
M
,
Casimiro
MC
,
Wang
C
,
Shirley
LA
,
Jiao
X
,
Katiyar
S
, et al
p21CIP1 attenuates Ras- and c-Myc-dependent breast tumor epithelial mesenchymal transition and cancer stem cell-like gene expression in vivo
.
Proc Natl Acad Sci U S A
2009
;
106
:
19035
9
.
27.
Nguyen
DH
,
Taub
D
. 
Cholesterol is essential for macrophage inflammatory protein 1 beta binding and conformational integrity of CC chemokine receptor 5
.
Blood
2002
;
99
:
4298
306
.
28.
Janowski
E
,
Jiao
X
,
Katiyar
S
,
Lisanti
MP
,
Liu
M
,
Pestell
RG
, et al
c-Jun is required for TGF-beta-mediated cellular migration via nuclear Ca(2) signaling
.
Int J Biochem Cell Biol
2011
;
43
:
1104
13
.
29.
Velasco-Velazquez
MA
,
Agramonte-Hevia
J
,
Barrera
D
,
Jimenez-Orozco
A
,
Garcia-Mondragon
MJ
,
Mendoza-Patino
N
, et al
4-Hydroxycoumarin disorganizes the actin cytoskeleton in B16-F10 melanoma cells but not in B82 fibroblasts, decreasing their adhesion to extracellular matrix proteins and motility
.
Cancer Lett
2003
;
198
:
179
86
.
30.
Liu
H
,
Patel
MR
,
Prescher
JA
,
Patsialou
A
,
Qian
D
,
Lin
J
, et al
Cancer stem cells from human breast tumors are involved in spontaneous metastases in orthotopic mouse models
.
Proc Natl Acad Sci U S A
2010
;
107
:
18115
20
.
31.
Zahler
MH
,
Irani
A
,
Malhi
H
,
Reutens
AT
,
Albanese
C
,
Bouzahzah
B
, et al
The application of a lentiviral vector for gene transfer in fetal human hepatocytes
.
J Gene Med
2000
;
2
:
186
93
.
32.
Walker
DK
,
Abel
S
,
Comby
P
,
Muirhead
GJ
,
Nedderman
AN
,
Smith
DA
. 
Species differences in the disposition of the CCR5 antagonist, UK-427,857, a new potential treatment for HIV
.
Drug Metab Dispos
2005
;
33
:
587
95
.
33.
Wu
K
,
Katiyar
S
,
Li
A
,
Liu
M
,
Ju
X
,
Popov
VM
, et al
Dachshund inhibits oncogene-induced breast cancer cellular migration and invasion through suppression of interleukin-8
.
Proc Natl Acad Sci U S A
2008
;
105
:
6924
9
.
34.
Charafe-Jauffret
E
,
Ginestier
C
,
Monville
F
,
Finetti
P
,
Adelaide
J
,
Cervera
N
, et al
Gene expression profiling of breast cell lines identifies potential new basal markers
.
Oncogene
2006
;
25
:
2273
84
.
35.
Riaz
M
,
Elstrodt
F
,
Hollestelle
A
,
Dehghan
A
,
Klijn
JG
,
Schutte
M
. 
Low-risk susceptibility alleles in 40 human breast cancer cell lines
.
BMC Cancer
2009
;
9
:
236
.
36.
Hollestelle
A
,
Nagel
JH
,
Smid
M
,
Lam
S
,
Elstrodt
F
,
Wasielewski
M
, et al
Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines
.
Breast Cancer Res Treat
2010
;
121
:
53
64
.
37.
Kao
J
,
Salari
K
,
Bocanegra
M
,
Choi
YL
,
Girard
L
,
Gandhi
J
, et al
Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery
.
PLoS ONE
2009
;
4
:
e6146
.
38.
Mueller
A
,
Mahmoud
NG
,
Goedecke
MC
,
McKeating
JA
,
Strange
PG
. 
Pharmacological characterization of the chemokine receptor, CCR5
.
Br J Pharmacol
2002
;
135
:
1033
43
.
39.
Petkovic
V
,
Moghini
C
,
Paoletti
S
,
Uguccioni
M
,
Gerber
B
. 
I-TAC/CXCL11 is a natural antagonist for CCR5
.
J Leukoc Biol
2004
;
76
:
701
8
.
40.
Miyashita
M
,
Smith
MW
,
Willey
JC
,
Lechner
JF
,
Trump
BF
,
Harris
CC
. 
Effects of serum, transforming growth factor type beta, or 12-O-tetradecanoyl-phorbol-13-acetate on ionized cytosolic calcium concentration in normal and transformed human bronchial epithelial cells
.
Cancer Res
1989
;
49
:
63
7
.
41.
Prescher
JA
,
Contag
CH
. 
Guided by the light: visualizing biomolecular processes in living animals with bioluminescence
.
Curr Opin Chem Biol
2010
;
14
:
80
9
.
42.
Yaal-Hahoshen
N
,
Shina
S
,
Leider-Trejo
L
,
Barnea
I
,
Shabtai
EL
,
Azenshtein
E
, et al
The chemokine CCL5 as a potential prognostic factor predicting disease progression in stage II breast cancer patients
.
Clin Cancer Res
2006
;
12
:
4474
80
.
43.
Shideman
CR
,
Hu
S
,
Peterson
PK
,
Thayer
SA
. 
CCL5 evokes calcium signals in microglia through a kinase-, phosphoinositide-, and nucleotide-dependent mechanism
.
J Neurosci Res
2006
;
83
:
1471
84
.
44.
Dorr
P
,
Westby
M
,
Dobbs
S
,
Griffin
P
,
Irvine
B
,
Macartney
M
, et al
Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity
.
Antimicrob Agents Chemother
2005
;
49
:
4721
32
.
45.
Strizki
JM
,
Tremblay
C
,
Xu
S
,
Wojcik
L
,
Wagner
N
,
Gonsiorek
W
, et al
Discovery and characterization of vicriviroc (SCH 417690), a CCR5 antagonist with potent activity against human immunodeficiency virus type 1
.
Antimicrob Agents Chemother
2005
;
49
:
4911
9
.
46.
Isik
N
,
Hereld
D
,
Jin
T
. 
Fluorescence resonance energy transfer imaging reveals that chemokine-binding modulates heterodimers of CXCR4 and CCR5 receptors
.
PLoS ONE
2008
;
3
:
e3424
.
47.
Mira
E
,
Lacalle
RA
,
Gonzalez
MA
,
Gomez-Mouton
C
,
Abad
JL
,
Bernad
A
, et al
A role for chemokine receptor transactivation in growth factor signaling
.
EMBO Rep
2001
;
2
:
151
6
.
48.
Thirkill
TL
,
Lowe
K
,
Vedagiri
H
,
Blankenship
TN
,
Barakat
AI
,
Douglas
GC
. 
Macaque trophoblast migration is regulated by RANTES
.
Exp Cell Res
2005
;
305
:
355
64
.
49.
Abel
S
,
van der Ryst
E
,
Rosario
MC
,
Ridgway
CE
,
Medhurst
CG
,
Taylor-Worth
RJ
, et al
Assessment of the pharmacokinetics, safety and tolerability of maraviroc, a novel CCR5 antagonist, in healthy volunteers
.
Br J Clin Pharmacol
2008
;
65
Suppl 1
:
5
18
.
50.
Vaday
GG
,
Peehl
DM
,
Kadam
PA
,
Lawrence
DM
. 
Expression of CCL5 (RANTES) and CCR5 in prostate cancer
.
Prostate
2006
;
66
:
124
34
.
51.
Perret
GY
,
Crepin
M
. 
New pharmacological strategies against metastatic spread
.
Fundam Clin Pharmacol
2008
;
22
:
465
92
.
52.
Sugasawa
H
,
Ichikura
T
,
Tsujimoto
H
,
Kinoshita
M
,
Morita
D
,
Ono
S
, et al
Prognostic significance of expression of CCL5/RANTES receptors in patients with gastric cancer
.
J Surg Oncol
2008
;
97
:
445
50
.