Introduction

Chemokines are small secretory chemo-attractant cytokines (8–14 kDa), which mainly regulate the directional migration of leukocytes, epithelial and endothelial cells in a concentration-dependent manner [1, 2]. The whole chemokine network is comprised of about 50 ligands and 20 receptors, where they demonstrate a phenomenon of redundancy as multiple chemokine ligands can interact with a single receptor and multiple receptors could be the target of a single chemokine ligand [35]. High levels of inflammatory cytokines, hypoxia, polarization/differentiation of immune cells and production of various growth factors can induce expressional changes of chemokines [68]. The expression patterns of the chemokine network are of vital importance in different autoimmune diseases, pathogenic situations, tumorigenesis and metastasis of malignant cancers [8, 9]. Significant alterations in the expressional profile of chemokine ligands and their corresponding receptors have been observed in different human cancers, which, in turn, alter the immune response and effect the progression of tumor mass [10, 11]. Nevertheless, the final outcomes of these modulations depend on the type of chemokines involved, cancerous tissue, leukocyte infiltrate and surrounding microenvironment [12, 13].

Colorectal cancer (CRC) is the third leading type of cancer in the developed countries and the second major cause of cancer-related mortalities worldwide [14, 15]. Significant alterations in the chemokines and their corresponding effects on disease progression have been identified in primary and metastatic CRC. Being an important component of chemokine network, understanding the role of chemokine receptor 5 (CCR5) in CRC is of paramount importance. CCR5 is a well-characterized chemokine receptor, which was identified for the first time about two decades ago [16, 17]. Three chemokine ligands for this receptor, described in detail, are CCL3 (MIP-1α), CCL4 (MIP-1β) and CCL5 (RANTES). CCR5 is a G-protein-coupled receptor with seven transmembrane loops, which interacts with the corresponding ligands and activates the intracellular bound heterotrimeric Gαβγ unit for subsequent downstream signaling [18, 19]. Noteworthy alterations in the expression of CCR5 have been correlated with the prognosis of primary CRC including metastasis to potential target organs. Regarding the expression level of CCR5 on CRC cells, contradicting reports have been published. Some authors reported higher CCR5 expression in primary CRC with increased local growth [20, 21], while others have found that high CCR5 expression is required for metastasis [22, 23]. In addition to this, it is presumed that variations in expressional pattern of CCR5 can alter the immune response system in CRC, which in turn will favor the further progression of the disease [2428].

In the present study, we aimed at investigating the role of CCR5 receptor in CRC cell lines following its blockage with an available antagonist. For this purpose, we selected primary (SW480) and metastatic (SW620) human CRC cell lines for detecting possible differences between primary and metastatic CRC malignancies. CCR5 receptor was blocked by maraviroc (MVC), which is a FDA-approved antagonist for this receptor [29]. Following the blockage of CCR5 by MVC, we investigated the impact on cellular proliferation, cell cycle distributions and apoptosis-related activities/pathways in the selected cell lines. This study illustrates the systematic impact of blocking CCR5 on various biological properties of CRC cells. Furthermore, the results led us to search into the importance of this receptor as a therapeutic for treatment of CRC.

Materials and methods

Cell lines

Two human colon adenocarcinoma cell lines, SW480 (primary) and SW620 (metastatic), were obtained from American Type Culture Collection (ATCC) and cultured in RPMI-1640 medium (Invitrogen, Darmstadt, Germany) supplemented with fetal bovine serum (10 %) and l-glutamine (2 mM). The cell lines, free of pathogenic contaminations, were maintained under standard incubation conditions (5 % CO2, 37 °C) with a humidified atmosphere and passaged routinely to maintain the logarithmically growing cell populations.

Cell viability assay

Viability of selected cell lines, following the treatment with MVC (UK-427857, Selleck Chemical Co. China), was assessed by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide) dye reduction assay. In brief, the cells were seeded at pre-optimized density (5 × 103 cells/well) in 96-well plates and treated with increasing concentrations of MVC (1–750 µM) for three time points (24, 48 and 72 h). Surviving cell fractions from the treated and control groups (eight replicates/sample) were determined by adding 10 µl/well MTT solution (10 mg/ml in PBS) and dissolving newly formed formazan crystals with 100 µl of acidic 2-propanol (0.04 N HCl). Optical density was measured by an ELISA plate reader (Anthos Mikrosysteme GmbH, Krefeld, Germany) at 540 nm wavelength with 690-nm reference filter. Cell survival rates were calculated as the percentages of untreated controls, and inhibitory concentrations (IC) were determined by GraphPad Prism 5 software.

Cell cycle assay

Effects of antagonizing CCR5 by MVC on cell cycle were determined by propidium iodide (PI) fluorescent staining and flow cytometry analysis (FACS). Briefly, the cells were seeded (3.5 × 105 cells/flask) in 25-cm2 cell culture flasks and treated with MVC (IC25, IC50 or IC75). After 48 h of drug exposure, the cells were harvested and re-suspended in 0.1 ml of PBS (2 × 105 cells) followed by the addition of ice-cold ethanol (70 %) for fixation. After an incubation period of 2 h at 4 °C, the cells were washed, re-suspended in PBS containing RNaseA (1 mg/ml) to digest their RNA and incubated for 30 min at 37 °C. Afterwards, PI (50 µg/ml) was added to the cells, and analysis was done immediately (≤30 min) in a FACS Canto (BD Biosciences, San Jose, CA, USA). Ten thousand cells (events) were analyzed from each sample, and the cell’s distributions in G0/G1, S and G2/M phases of cell cycle were calculated by ModFit LT software.

Hoechst 33324 staining

The cells were seeded (1.5 × 105 cells/well) in 6-well plates having sterilized cover slips inside. Next day, treatment was done with increasing concentrations of MVC (IC25, IC50 or IC75) for 48 h followed by the fixation of cells with 4 % formaldehyde and permeabilization with 0.3 % Triton X-100 (Sigma, Munich, Germany). Then, the cells were stained with 1.6 mM Hoechst 33342 dye (Invitrogen, Karlsruhe Germany), placed top down on glass slides and kept in the dark until analyses. Prepared slides were randomly photographed with a fluorescence Axiophot microscope (Carl Zeiss, Oberkochen, Germany).

Annexin V-FITC flow cytometry assay

Early signs of apoptosis, in response to MVC treatment, were investigated by Annexin V-FITC Apoptosis Detection Kit (eBioscience, Frankfurt, Germany). In brief, 3.5 × 105 cells/flask were seeded in 25-cm2 cell culture flasks and next day treated with MVC (IC25, IC50 or IC75) for 48 h. The cells were harvested with EDTA free trypsin (0.25 %) and washed with PBS followed by a second wash with 1X binding buffer provided with the kit. Then, 2 × 105 cells were re-suspended in 100 µl of 1X binding buffer, and 5 µl of fluorochrome-conjugated Annexin V dye was added before samples were incubated for 15 min at room temperature in dark. After the incubation period, the cells were washed again with 1X binding buffer to remove additional unbound Annexin V dye and re-suspended in 200 µl of 1X binding buffer. PI (5 µl), provided along the kit, was added to each sample, and analyses was done with a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA, USA).

RNA isolation and cDNA synthesis

The CRC cells were seeded in 6-well plates (1.5 × 105 cells/well) and treated with MVC (IC25, IC50 or IC75) on next day. After 48 h of drug exposure, the cells were harvested and washed and total RNA was extracted from the cell pellets with RNeasy Mini kit (Qiagen, Hilden, Germany) following the manufacturer’s protocol. Concentrations of the extracted RNA were measured by a GeneQuant pro spectrophotometer (GE Healthcare, Munich, Germany), and 1 µg RNA was used to synthesize complementary DNA (cDNA) by using Maxima reverse transcriptase enzyme (Thermo Scientific, Schwerte, Germany). Prepared cDNA samples were used in the subsequent real-time PCR analysis.

Quantitative real-time RT-PCR

Modulation of mRNA expression in response to blocking CCR5 with MVC was studied for cell cycle-relevant genes (cyclins) by qRT-PCR methodology in both selected cell lines. For this purpose, the cDNA samples were subjected to qRT-PCR amplification by using 2X LC480 Master Mix along with the appropriate probes from the Human Universal Probe library (Roche, Mannheim, Germany). The samples were amplified in triplicate, and the expression level of a reference gene (GAPDH) was used for normalizing the data. After the amplification procedures, fold changes were calculated by the 2-∆∆CT method [30]. Primer sequences used for the amplifications of cyclins and GAPDH are shown in Supplementary Table 1.

Human apoptosis panel, 96

To have a deeper insight about the effects of MVC on apoptosis-related genes, we used a ready-made Human Apoptosis Panel, 96 (Roche, Mannheim, Germany). This panel included the primers for 84 apoptosis-relevant genes along with their corresponding probes. In addition to this, two RT-negative controls to monitor the residual genomic DNA from RNA extraction step, three RT-positive controls to check the quality of reverse transcription and seven reference genes for normalization of the data were also included in this panel. For this analysis, SW480 cells were exposed to increasing concentrations of MVC (IC50 and IC75) for 48 h, followed by extraction of total RNA and cDNA synthesis as described above. RT-PCR was performed by adding 0.5 µl cDNA/sample (50 µl transcribed from 2.5 µg extracted RNA) along with 2X LC480 Master Mix (Roche, Mannheim, Germany) in a LightCycler 480 Real-Time PCR System.

Western blot analysis

The two cell lines (3 × 105 cells/flask) were treated in 25-cm2 cell culture flasks with MVC (IC25, IC50 or IC75) for 48 h. After the treatment, cells were harvested, transferred to 1.5-ml microcentrifuge tubes and washed in PBS and pellets were lysed with RIPA buffer (150 mM sodium chloride, 1.0 % NP-40, 0.5 % sodium deoxycholate, 0.1 % sodium dodecyl sulfate, 50 mM Tris, pH 8.0) and supplemented with complete protease inhibitor cocktail tablets (Roche, Mannheim, Germany). The lysates were maintained at constant agitation for 30 min at 4 °C, and later the supernatant was collected after spinning at 14,000 rpm at 4 °C for 20 min. The supernatant was quantified for protein concentration using the Pierce Protein Assay. The total protein lysates (30–50 µg) were subjected to electrophoresis on 4–12 % gradient polyacrylamide SDS gels. Proteins were transferred onto PVDF membranes, and the membranes were probed for different proteins (cleaved PARP, cleaved caspase 3, cleaved caspase 7 and cleaved caspase 9) using specific antibodies (Cell Signaling Technologies, Frankfurt, Germany) as per manufacturer’s instructions. Immunoblots were developed using a HRP-conjugated anti-mouse or anti-rabbit IgG (Cell Signaling Technologies, Frankfurt, Germany) and ECL-System (Amersham Pharmacia Biotech, Munich, Germany). Levels of β-actin were used to normalize the protein expression. Relative concentrations were assessed by densitometric analysis of digitized autographic images using the ImageJ Program.

Results

Blocking the CCR5 induces cytotoxicity in CRC cells

SW480 and SW620 cells were treated with increasing concentrations of MVC (1–750 µM), and resulting effects on cell viability were investigated by MTT dye reduction assay. MVC reduced the cell proliferation and led to cytotoxic effects in a concentration-dependent manner in both cell lines. At lower concentrations of MVC (≤50 µM), there were stimulatory effects on cell proliferation, while above 100 µM concentrations, inhibitory effects were found as calculated by GraphPad Prism 5 software. Concentrations from 100 to 750 µM caused a steady decline in survival rates for both cell lines in a dose–response curve. IC50 values, with 95 % confidence limits, were calculated for both cell lines for the three time points (Fig. 1).

Fig. 1
figure 1

In vitro cytotoxic effects induced by maraviroc in colorectal cancer cells: a, b SW480 and SW620 cells, respectively, were treated with 1–750 µM of maraviroc for 24, 48 and 72 h. Cytotoxicity was measured by MTT dye reduction assay. IC50 values with 95 % confidence limits are given below the respective curves

Maraviroc induces significant arrest in G1 phase of the cell cycle

Both CRC cell lines were analyzed after treatment with MVC to see the effects of CCR5 blockage on cell cycle. In the control samples, both cell lines illustrated comparable and almost equal distributions in G0/G1 (51–54 %) and S phase (44–45 %) cells as well as significant lower percentages of G2/M phase cells (2–4 %). In response to MVC exposure, both cell lines showed an increase in G0/G1 fractions, followed by a subsequent decrease in S phase cells. The effects were more profound in SW620 cells, where maximum increase in G0/G1 phase was 16 % as compared to 3 % in SW480 cells. With regard to S phase, both cell lines demonstrated a decrease in cell’s % in a concentration-dependent format as shown by a maximum reduction of 5 and 18 % for SW480 and SW620 cells, respectively. In response to the IC75 concentrations of MVC, a mild G2/M phase arrest was observed as shown by an increase of 5 and 2 % for SW480 and SW620 cells, respectively (Fig. 2).

Fig. 2
figure 2

Effects of maraviroc on cell cycle: a, b SW480 and SW620 cells, respectively, show G1 phase arrest at lower maraviroc concentrations (IC25, IC50), while additional G2 arrest is visible at higher concentrations (IC75). c, d In SW480 and SW620 cells, respectively, expression levels of CCND1 and CCNE2 were down-regulated concentration dependently (except for IC25 concentration in SW620 cells), while CCNE1 was slightly up-regulated

CCR5 blockage modulates the expression of cyclins

Modulations in different phases of cell cycle provided us the base to investigate expressional profiles of the cell cycle-relevant genes (cyclins). For this purpose, after treatment with MVC (IC25, IC50 or IC75), we studied the expression patterns of cyclin family genes at mRNA level by qPCR. Following normalization of the results, we noticed a consistent, concentration-dependent down-regulation of cyclin D1 (CCND1) for SW480 cells (maximum −2.69-fold), while SW620 cells showed a moderate down-regulation of CCND1 (maximum −1.45-fold) with higher concentrations of MVC (IC75). Both selected cell lines illustrated slight induction of cyclin E1 (CCNE1) and inhibition of cyclin E2 (CCNE2) as shown by a maximum change of 1.32- and −1.46-fold, respectively (Fig. 2).

Nuclear/DNA fragmentation by maraviroc

To determine whether the CCR5 blockage exerts any apoptotic activities at morphological level, we studied the ability of MVC to induce oligonucleosomal DNA/nuclear fragmentation in the selected cell lines. Exposure of both cell lines to increasing concentrations of MVC (IC25–IC75) for 48 h led to disruptive changes in nuclei. At lower concentrations of the CCR5 antagonist (IC25), chromatin condensation was observed, while at higher concentrations (IC50 and IC75), significant nuclear shrinkage and fragmentation were also noticed (Fig. 3).

Fig. 3
figure 3

Morphological signs of apoptosis in colorectal cancer cells treated with maraviroc: a Nuclear shrinkage, chromatin condensation and fragmentation (indicated by white arrows) are visible in SW480 and SW620 cells treated with increasing concentrations of maraviroc and subsequent staining with Hoechst 33342. b Corresponding sub-G1 (hypodiploid) DNA fractions obtained from flow cytometry analysis after the treatment with maraviroc are shown

CCR5 blockage by maraviroc stimulates apoptosis

Destructive morphological changes in the nuclei/DNA by MVC led to further investigations about apoptosis induction by CCR5 blockage. For this purpose, following treatment with MVC, we examined annexin V-FITC-based labeling of the inner membrane-bound phosphatidylserine (PS) of the cells. Both cell lines showed a significant increase in annexin V-positive cells with increasing concentrations of MVC as shown in Fig. 4. Maximum increases in annexin V-positive cells, in response to the highest concentrations of MVC (IC75), were 27 and 24 % for SW480 and SW620 cells, respectively. The continuous increase in the percentages of annexin V-bound cells in a concentration-dependent format showed the ability of MVC to induce apoptosis in CRC cell lines. Furthermore, the steady increase in apoptotic cells corroborated our cytotoxicity and nuclei/DNA fragmentation data from MTT and Hoechst 33342 staining assays, respectively.

Fig. 4
figure 4

Effects of maraviroc on phosphatidylserine translocation. Induction of apoptosis in colorectal cancer cells by maraviroc exposure was examined by annexin V-FITC binding assay. Cells were treated with increasing concentrations of maraviroc for 48 h. Concentration-dependent induction of early apoptosis was observed in both colorectal cancer cell lines as indicated by the corresponding percentages in lower right quadrants

Maraviroc induces multiple apoptosis-relevant genes

To figure out mechanistic aspects of CCR5 blockage by MVC and its correlation with apoptotic effects, we investigated the apoptosis-relevant genes by a ready-made apoptosis panel (see materials and methods). SW480 cells, treated with two different concentrations of MVC (IC50 and IC75), showed significant induction (≥twofold) of 15 pro- and anti-apoptosis-relevant genes at mRNA level. Out of the 84 genes included in this panel, 68 genes were not altered significantly (≤twofold), while one gene (NGFR) was down-regulated to a maximum fourfold. Nevertheless, significant induction was observed in a variety of gene families like caspases (1, 4, 5 and 10), stress-related BCL2 family (BCL2, BBC3, BIRC3 and BCL2L10) and death receptor signaling molecules (FAS, FASLG, TNF, TNFRSF1A, TNFRSF25) as shown in Fig. 5a. In addition to this, based on the results from the apoptosis panel, a signaling pathway for apoptosis was predicted by using the Ingenuity Software (Redwood, CA, USA) as shown in Fig. 5b.

Fig. 5
figure 5

Modulation of multiple apoptosis-related genes by maraviroc exposure: a SW480 cells were treated with maraviroc, and apoptosis-related genes were evaluated by Human Apoptosis Panel, 96. CP (crossing point) values, indicating the linear amplification of transcribed cDNA, were calculated by the Second Derivative Maximum Method, and the ∆∆CT method was used to calculate the relative expressional levels of the genes as compared to the respective untreated control samples. Averages of seven reference genes, incorporated in the panel, were used to normalize the data. b A schematic apoptosis pathway (Ingenuity Pathway Analysis) was modified to represent the modulation of signaling chain members caused by maraviroc (IC75, 48 h) in SW480 cells. For abbreviations used, see the supplementary data

Activation of cleaved caspases by maraviroc

To support and confirm the idea of induction of apoptosis in CRC cells by MVC exposure, we examined the expressional levels of caspases by western blot analysis. Both cell lines were exposed to varying concentrations of MVC (IC25, IC50 or IC75) for 48 h, and expression levels of activated/cleaved caspases 3, 7, and 9 were examined at protein level. The three caspases, which regulate the final steps of apoptosis, showed significant boosts in expression after MVC treatment. The utmost increases in expressions for caspases 3, 7 and 9 were 4.9-, 8.6- and 2.5-fold for SW480 cells, whereas SW620 cells showed 51.8-, 7.1- and 2.1-fold inductions, respectively. In addition to this, expression levels of cleaved PARP protein, which is a key hallmark of apoptosis, were also found to be significantly elevated with a maximum induction of 4.3- and 17.6-fold for SW480 and SW620 cells, respectively (Fig. 6).

Fig. 6
figure 6

Induction of activated caspases by maraviroc exposure: Following treatment with increasing concentrations of maraviroc for 48 h, western blot analysis for the expression levels of cleaved caspases 3, 7, 9 and cleaved PARP was performed with β-actin as a loading control. a, c Maraviroc induced the expression of cleaved caspases and PARP in SW480 and SW620 cells, respectively. b, d Fold inductions are calculated for both cell lines by the ImageJ Program

Discussion

MVC is the only clinically available and well-tolerated non-competitive (allosteric) CCR5 antagonist, which has been used for the treatment of CCR5-dependant immunodeficiency virus type 1 (HIV-1) infection. In recent years, MVC was also assessed for its potential as antineoplastic agent in a variety of primary and metastatic cancers. In this regard, animal models have been used to study the impact of CCR5 blockage by MVC and subsequent effects on tumor progression [3133]. Promising outcomes from these cancer-related studies (other than CRC) involving MVC as a CCR5 blocker provoked us to study the effects of this antagonist in CRC cells.

In this study, we examined the role of CCR5 in two CRC cell lines (SW480 and SW620) by blocking the receptor with MVC. In MTT assay, MVC exposure showed cytotoxic effects in both cell lines at higher concentrations (≥100 µM), while there were initially some stimulatory effects at lower concentrations (≤50 µM). Although, clinically used dosages of MVC are not associated with serum levels higher than 100 µM, the antineoplastic success of MVC in mice-bearing breast, gastric or hepatocellular cancers indicates a potential of this drug in vivo [3134]. We next aimed at finding the mechanism causing reduced cell proliferation in response to CCR5 blockage by MVC. For this purpose, we studied the possible effects on cell cycle in CRC cells by PI staining and flow cytometry. MVC exposure resulted in G1 phase arrest in both CRC cell lines, and a G2 phase arrest was found only for the highest MVC concentrations (IC75). We focused at the major arrested G1 phase at molecular levels and noticed the inhibition of cell cycle-relevant genes in both cell lines. Most prominent down-regulation was observed for CCND1, which interacts and regulates cyclin-dependent kinases (CDKs). In this regard, two important CDKs are CDK4 and CDK6, which are required for G1/S phase transition of the cell cycle [35, 36]. Thus, inhibition of CCND1 by MVC exposure hypothetically led to the decreased activity of CDK4 and CDK6, which ultimately hampered G1/S phase transition leading to G1 phase arrest. In addition to this, we found a moderate inhibition of CCNE2, which predominantly interacts with CDK2 and also regulates the G1/S transition of the cell cycle [37].

We further studied the cytotoxic mechanisms resulting from CCR5 blockage by MVC and found clear signs of apoptosis at morphological level. MVC induced the typical features of apoptosis like detachment of cells from the culture dish, blebbing, shrinkage in size and fragmentation of DNA/nuclei [38]. These signs of apoptosis, examined by general observations and staining protocols, were further studied by annexin V-FITC staining. Apoptosis is considered as programmed cell death, and one of the preliminary events in this process is translocation of membrane-bound PS molecules from the inner side of the plasma membrane to the outer surface [39]. Annexin V is a Ca2+-dependent phospholipid binding protein and has high affinity to bind these exposed PS molecules. In response to increasing concentrations of MVC, there were profound and almost similar increases of annexin V-positive cells in both CRC cell lines. This indicates and confirmed the ability of MVC to compel the tumor cells into the apoptotic process, regardless of their primary or metastatic nature of origin.

The significant induction of apoptosis prompted us to further investigate the respective signaling pathways. For this purpose, we exposed SW480 cells with MVC and examined the apoptosis-relevant genes by a ready-made panel (see materials and methods). MVC treatment showed significant modulation (≥twofold) of 16 genes, out of which significant proportions were comprised of the death receptors and their corresponding ligands. These included the cell surface FAS and tumor necrosis factor (TNF) superfamily receptors. Induction of death receptors and their ligands in this study indicates the ability of MVC to induce the extrinsic arm of apoptosis in CRC cells [40]. The other two dominating groups of gene families, which were altered significantly in response to MVC exposure, were caspases and BCL2. Considerable modulations of BCL2 family members, which cover the intrinsic arm of apoptosis [41], revealed the ability of MVC to exert stress in CRC cells through mitochondrial involvement. This could occur in response to chemokine deprivation as well, which, in turn, will cause alteration of BID and subsequent activation of BAX and BAK [42]. Regarding the caspase family, we found the induction of initiator caspases (8 and 10), which are required to initiate the apoptosis signaling in response to death receptor stimulation [43, 44]. The utmost induction at mRNA level was found for caspase 5 (27-fold), which is known to be involved in activation of caspase 1, which, in turn, processes the pro-inflammatory interleukins-1β and 18 [45].

Significant induction of initiator caspases led us to study the expressional profiles of the cleaved executer caspases (3, 7 and 9) and PARP at protein levels. Interestingly, we found a slight decrease at mRNA level of these caspases in the apoptosis panel (see pathway), but significant inductions were observed for cleaved proteins in both CRC cell lines after exposure to increasing concentrations of MVC. This indicates a high turnover of the relevant transcripts into proteins, followed by the amplification of cleaved (activated) forms for the subsequent apoptotic effects. The expression levels of cleaved caspases and PARP did not follow a concentration-dependent format for lower concentrations of MVC (IC25 and IC50), which indicates a plateau of cleaved protein levels, as the differences between IC25 and IC50 were not significant. In addition to this, induction of cleaved proteins was also partially cell line specific, as caspase 7 was the most effectively cleaved caspase in SW480 cells, while caspase 3 was most altered in SW620 cells.

In conclusion, blocking the CCR5 receptor by MVC induces cytotoxic and apoptotic effects in CRC cells. These antineoplastic effects were observed in CRC cells regardless of their primary or metastatic origin. These findings led us to identify MVC as antineoplastic lead compound which can serve as proof of concept for the anticancer effects of blocking the CCR5 receptor and will foster the development of CCR5 antagonists for the treatment of colorectal cancer.