Agonists to A3 adenosine receptor (A3AR) have been reported to inhibit cell growth and/or induce apoptosis in various tumors. We tested the effect of a novel A3AR agonist generically known as LJ-529 in breast cancer cells. Anchorage-dependent cell growth and in vivo tumor growth were attenuated by LJ-529, independently of its estrogen receptor (ER) α status. In addition, apoptosis was induced as evidenced by the activation of caspase-3 and c–poly(ADP)ribose polymerase. Furthermore, the Wnt signaling pathway was down-regulated and p27kip was induced by LJ-529. In ER-positive cells, the expression of ER was down-regulated by LJ-529, which might have additionally contributed to attenuated cell proliferation. In ER-negative, c-ErbB2-overexpressing SK-BR-3 cells, the expression of c-ErbB2 and its downstream extracellular signal-regulated kinase pathway were down-regulated by LJ-529. However, such effect of LJ-529 acted independently of its receptor because no A3AR was detected by reverse transcription-PCR in all four cell lines tested. In conclusion, our novel findings open the possibility of LJ-529 as an effective therapeutic agent against both ER-positive and ER-negative breast cancers, particularly against the more aggressive ER-negative, c-ErbB2-overexpressing types. [Mol Cancer Ther 2006;5(3):685–92]

Synthetic agonists to the Gi-protein-coupled A3 adenosine receptor (A3AR) have recently been suggested as a possible therapeutic agent to various cancers (1, 2). The A3AR agonist N6-(3-iodobenzyl)-5′-N-methylcarbamoyladenosine (IB-MECA) and its 2-chloro derivative, 2-chloro-N6-(3-iodobenzyl)-5′-N-methylcarbamoyladenosine (Cl-IB-MECA), inhibit the in vitro and in vivo growth of B16-F10 melanoma cells (3, 4). IB-MECA has also been reported to inhibit prostate carcinoma cell growth, breast cancer cell proliferation, and primary colon carcinoma growth and liver metastasis (58). Cl-IB-MECA is known to induce apoptosis of HL-60 and MOLT-4 leukemic and Li-7A hepatoma cells (9, 10). Although the action is not directly on cancer cells, Cl-IB-MECA is also known to potentiate natural killer cells, which is strongly implicated to have tumor surveillance activity (11).

Despite many reports suggesting synthetic A3AR agonists as a promising therapeutic agent against various cancers, the underlying molecular mechanisms seem to vary considerably depending on the tumor system. In melanoma cells, the A3AR internalization/externalization and the deregulation of the Wnt signaling pathway by IB-MECA has been reported. IB-MECA in melanoma cells rapidly internalizes, sorts, resynthesizes, and externalizes the receptor to the cell surface (12). In addition, it decreases cyclic AMP, prevents the activation of both protein kinase A and Akt, diminishes the phosphorylation of glycogen synthase kinase 3β (GSK-3β), and by maintaining GSK-3β in its active form, inhibits cell proliferation via deregulation of the Wnt pathway (13). Furthermore, Cl-IB-MECA has been reported to inhibit human melanoma cell proliferation via phosphatidylinositol 3-kinase/Akt–dependent inhibition of the extracellular signal-regulated kinase (ERK) 1/2 phosphorylation (14). The suppression of the Wnt signaling pathway by IB-MECA has also been reported in prostate carcinoma cells (5). Cl-IB-MECA induces Fas and apoptosis in a p53-independent manner in human leukemia cell lines (9), and apoptosis most likely by activating of caspase-3 in human hepatoma cells (10). In breast cancer cells, IB-MECA suppresses cell proliferation by down-regulating estrogen receptor (ER) α (6) and inhibits anchorage-dependent cell growth in both ER-positive and ER-negative cell lines (7).

We have recently reported that the 4′-thio analogues of the Cl-IB-MECA are associated with higher potency and selectivity than IB-MECA and Cl-IB-MECA (15). In this study, we tested the effect of one of the 4′-thio analogues of the Cl-IB-MECA generically known as LJ-529 [2-chloro-N6-(3-iodobenzyl)-5′-N-methylcarbamoyl-4′-thioadenosine; previously reported as thio-Cl-IB-MECA in ref. 15] in breast cancer cells. LJ-529 is reported to have a higher binding affinity to the human A3AR than Cl-IB-MECA (Ki = 0.38 versus 1.00 nmol/L; ref. 15). We report that LJ-529 was effective on both ER-positive and ER-negative breast cancer cell lines, both in vitro and in vivo, by affecting various signaling pathways. It is of significance that LJ-529 was effective in ER-negative, c-ErbB2-overexpressing breast cancer cells because this type of breast cancer is resistant to hormonal therapies and tends to have a more aggressive phenotype (16, 17). However, such effect was independent of the A3AR because none of the cells tested expressed endogenous A3AR.

Cell Culture and Drug Treatment

MCF7, T47D, SK-BR-3, and MDA-MB-231 were from American Type Culture Collection (Manassas, VA). Cells were maintained in DMEM (Welgene, Daegu, Republic of Korea) supplemented with 10% fetal bovine serum (Welgene) in a humidified atmosphere with 5% CO2 at 37°C. For drug treatment on cells, a stock of 100 mmol/L was prepared in DMSO. When cells reached 60% to 70% confluency, cells were treated with LJ-529 diluted in DMEM supplemented with 5% fetal bovine serum at indicated concentrations for indicated times.

3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide Assay

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays were done with CellTiter 96 Assay (Promega, Madison, WI) according to the instructions of the manufacturer. The absorbance at 570 nm was recorded using a 96-well plate reader (Bio-Rad, Hercules, CA).

Propidium Iodide Staining and Flow Cytometry Analysis

After drug treatment, cells were detached by trypsinization, collected by centrifugation, and washed once with PBS. Staining of cells with propidium iodide and analyzing them by flow cytometry (FACSCalibur and CellQuest software, BD Biosciences, San Jose, CA) were done as described (18).

4′,6-Diamidino-2-Phenylindole Staining

After drug treatment, cells were lifted by trypsinization and subjected to nuclear staining with 4′,6-diamidino-2-phenylindole as previously described (19), except that the cells were mounted with 50 μL VectaShield (Vector Laboratories, Burlingame, CA).

Immunoblotting

Cells were lysed in lysis buffer [50 mmol/L Tris (pH 7.4), 150 mmol/L NaCl, 1 mmol/L EDTA, 1% NP40, 0.25% sodium deoxycholate] supplemented with phosphatase inhibitors (Phosphatase Inhibitor Cocktail I and II, Sigma, St. Louis, MO) and protease inhibitors (Complete, Mini, EDTA-free, Roche Applied Science; Indianapolis, IN) and the cleared lysates by centrifugation were used for immunoblotting. Fifty to 100 μg total protein were loaded per lane, separated by SDS-PAGE, transferred to nitrocellulose filter, and subjected to immunoblotting with appropriate antibodies. Antibodies against caspase-3, A3AR, GSK-3β, cyclin D1, p27kip, and ERα were from Santa Cruz Biotechnologies (Santa Cruz, CA); Akt, phospho-Akt, and phospho-GSK-3β were from Cell Signaling Technology (Beverly, MA); c–poly(ADP)ribose polymerase was from BD Biosciences; and β-actin was from Sigma. Bands were detected with ECL Plus Western Blotting Detection Reagents from GE Healthcare (Chalfont St. Giles, United Kingdom).

Reverse Transcription-PCR

Total RNA was extracted with TRIzol (Invitrogen, Carlsbad, CA) and reverse transcription-PCR (RT-PCR) was done with Access RT-PCR System (Promega) according to the instructions of the manufacturer. PCR primers and thermal profiles for A3AR, glyceraldehyde-3-phosphate dehydrogenase, and β-actin were as previously described (6, 20). Specific primers used for PCR were as follows: A1AR (forward, 5′-GATGCCACCTTCTGCTTCAT-3′; reverse, 5′-AGACCATGTACTCCATGCTG-3′), A2aAR (forward, 5′-CTCCGGTACAATGGCTTGGT-3′; reverse, 5′-CTCCATCTGCTTCAGCTGTC-3′), and A2bAR (forward, 5′-TCCTCTGGGTCCTTGCCTTT-3′; reverse, 5′-GGGATTGACAACTGAATTGGC-3′).

Xenograft in Murine Model

Experiments were done in accordance with the guidelines established by the USPHS. Female nude BALB/cAnNCrjBgi-nu mice (Orient, Seoul, Republic of Korea) were used at 7 to 8 weeks of age and maintained in the specific pathogen-free laboratory animal facility at Hanyang University (Seoul, Republic of Korea). A xenograft was established by s.c. injection of in vitro cultured T47D (3 × 106 cells/200 μL) or SK-BR-3 (4 × 106 cells/150 μL) into the flank of mice. For xenograft with T47D cells, mice were supplemented with estradiol pellets (0.72 mg, released over 60 days; Innovative Research of America, Sarasota, FL). Tumors were measured in two diameters with calibers to permit calculation of tumor volume, V = {(D + d) / 2}3, where D and d were the larger and smaller diameters, respectively. For drug treatment in mice, a stock of 0.5 mg/mL was prepared by sonication in water. Five to seven days after injection, when the tumor width reached 1 to 3 mm, 20 animals were randomly selected, grouped, and treated with the indicated doses of LJ-529 daily and p.o. for 28 days. Each group contained five mice. The first day of drug treatment was set as day 0 and the tumor size and body weight were measured twice a week up to day 28.

Time- and Dose-Dependent Attenuation of Anchorage-Dependent Cell Proliferation by LJ-529 in Breast Cancer Cells

Because several A3AR agonists are known to attenuate cancer cell growth, we examined the effect of a novel A3AR agonist, LJ-529, in breast cancer cell lines. MCF7 and T47D cells were tested as ER-positive cells, whereas MDA-MB-231 and SK-BR-3 were examined as ER-negative breast cancer cell lines. As shown in Fig. 1, all of the cells tested yielded attenuated anchorage-dependent cell growth in a dose- and time-dependent manner regardless of its ER status.

Figure 1.

Time- and dose-dependent attenuation of anchorage-dependent cell growth by LJ-529 in breast cancer cells assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cells were treated with different concentrations of LJ-529 for indicated times. Points, mean percentage of corresponding control values; bars, SD. Three repeats were each done in triplicate.

Figure 1.

Time- and dose-dependent attenuation of anchorage-dependent cell growth by LJ-529 in breast cancer cells assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cells were treated with different concentrations of LJ-529 for indicated times. Points, mean percentage of corresponding control values; bars, SD. Three repeats were each done in triplicate.

Close modal

Enhanced Apoptosis by LJ-529 in Breast Cancer Cells

Next, we examined whether the attenuated growth upon LJ-529 treatment was due to accumulation of cells at a certain stage during the cell cycle. As seen in Fig. 2, a significant population of cells accumulated at the sub-G1 phase, suggesting enhanced apoptosis by LJ-529. To confirm that LJ-529 indeed induced apoptosis, nuclear fragmentation was quantified in cells after 4′,6-diamidino-2-phenylindole staining (Fig. 3A). As shown in Fig. 3B, both ER-positive and ER-negative cells showed enhanced apoptosis by LJ-529 in a dose-dependent manner. Biochemical evidence also support that LJ-529 activated the apoptotic pathway as seen by cleavages of caspase-3 and c–poly(ADP)ribose polymerase (Fig. 3C).

Figure 2.

Enhanced apoptosis by LJ-529 in breast cancer cells assessed by propidium iodide staining and flow cytometry analysis. A, cells were treated with vehicle (DMSO) or indicated concentrations of LJ-529 for 2 d. Cells were stained with propidium iodide and subjected to flow cytometry analysis. Representative fluorescence histograms of three independent experiments. B, two axes presented in A (left). Y axis, cell count; X axis, fluorescence intensity. Right, gatings that correspond to the phases of the cell cycle.

Figure 2.

Enhanced apoptosis by LJ-529 in breast cancer cells assessed by propidium iodide staining and flow cytometry analysis. A, cells were treated with vehicle (DMSO) or indicated concentrations of LJ-529 for 2 d. Cells were stained with propidium iodide and subjected to flow cytometry analysis. Representative fluorescence histograms of three independent experiments. B, two axes presented in A (left). Y axis, cell count; X axis, fluorescence intensity. Right, gatings that correspond to the phases of the cell cycle.

Close modal
Figure 3.

Enhanced apoptosis by LJ-529 in breast cancer cells assessed by 4′,6-diamidino-2-phenylindole staining and immunoblotting. A, representative photographs of cells treated with vehicle (DMSO) or 100 μmol/L LJ-529 for 2 d. B, quantified results of 4′,6-diamidino-2-phenylindole staining. Columns, average of three independent experiments with similar results; bars, SD. P values indicate statistical significance of induction of apoptosis by LJ-529 (Student's t test): *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, cell lysates were prepared from cells treated with 50 μmol/L LJ-529 for indicated times and subjected to immunoblotting for caspase-3 and c–poly(ADP)ribose polymerase (c-PARP) as described in Materials and Methods. Immunoblots for β-actin were shown as loading controls. Representative of three independent experiments.

Figure 3.

Enhanced apoptosis by LJ-529 in breast cancer cells assessed by 4′,6-diamidino-2-phenylindole staining and immunoblotting. A, representative photographs of cells treated with vehicle (DMSO) or 100 μmol/L LJ-529 for 2 d. B, quantified results of 4′,6-diamidino-2-phenylindole staining. Columns, average of three independent experiments with similar results; bars, SD. P values indicate statistical significance of induction of apoptosis by LJ-529 (Student's t test): *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, cell lysates were prepared from cells treated with 50 μmol/L LJ-529 for indicated times and subjected to immunoblotting for caspase-3 and c–poly(ADP)ribose polymerase (c-PARP) as described in Materials and Methods. Immunoblots for β-actin were shown as loading controls. Representative of three independent experiments.

Close modal

A3AR-Independent Effect of LJ-529 in Breast Cancer Cells

Because there are reports that A3AR agonists at high doses (micromolar concentrations) act independently of its receptor, A3AR (6, 9), we examined the expression of A3AR in breast cancer cells by immunoblotting and RT-PCR. As seen in Fig. 4A and B, A3AR expression was not detected in any of the breast cancer cells tested. Similar results were obtained regardless of the growth conditions, such as in culture medium supplemented with 10% or 5% fetal bovine serum with LJ-529 (data not shown). Therefore, we conclude that the effect of LJ-529 occurred independently of the A3AR in breast cancer cells. Intriguingly, when the expression of other adenosine receptors was examined, only the A2 isoforms (A2a and A2b) were detected by RT-PCR (Fig. 4C).

Figure 4.

The expression of adenosine receptors assessed by immunoblotting and/or RT-PCR. The expression levels of various adenosine receptors in rat brain (+) and breast cancer cell lines were assayed using immunoblotting and/or RT-PCR as described in Materials and Methods. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) or β-actin was used as controls. Results are obtained from cells treated with vehicle (DMSO) in culture medium supplemented with 5% fetal bovine serum for 2 d. A, Western analysis done with 100 μg cell lysate and anti-A3AR antibody. B, RT-PCR done with A3AR-specific primers. C, RT-PCR done with primers specific for various adenosine receptors. Representative of three independent experiments.

Figure 4.

The expression of adenosine receptors assessed by immunoblotting and/or RT-PCR. The expression levels of various adenosine receptors in rat brain (+) and breast cancer cell lines were assayed using immunoblotting and/or RT-PCR as described in Materials and Methods. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) or β-actin was used as controls. Results are obtained from cells treated with vehicle (DMSO) in culture medium supplemented with 5% fetal bovine serum for 2 d. A, Western analysis done with 100 μg cell lysate and anti-A3AR antibody. B, RT-PCR done with A3AR-specific primers. C, RT-PCR done with primers specific for various adenosine receptors. Representative of three independent experiments.

Close modal

Down-Regulation of the Wnt Signaling Pathway by LJ-529 in Breast Cancer Cells

As certain A3AR agonists are reported to deregulate the Wnt signaling pathway in cancer cells (5, 13), we tested the effect of LJ-529 treatment on the Wnt signaling pathway in T47D (ER-positive) and SK-BR-3 (ER-negative) cells. The total amount of Akt was reduced in both T47D and SK-BR-3 cells (Fig. 5A). Furthermore, the phosphorylated form of Akt at serine-473 was reduced in both cells in a dose-dependent manner (Fig. 5A). Whereas the total GSK-3β protein level remained unchanged, the phosphorylated form at serine-9 was reduced in a dose-dependent manner (Fig. 5A). The expression of cyclin D1 was examined because its transcription is regulated as a result of downstream β-catenin and T-cell factor/lymphoid enhancer–binding factor transcription factors (21). The protein level of cyclin D1 was also significantly reduced in a dose-dependent manner (Fig. 5A). Therefore, as evidenced from the changes of Akt, GSK-3β, to cyclin D1, the canonical Wnt signaling pathway was down-regulated by LJ-529 in breast cancer cells regardless of their ER status.

Figure 5.

Signaling pathways affected by LJ-529 treatment. Cell lysates were prepared from cells treated with 50 μmol/L LJ-529 for indicated times and subjected to immunoblotting with indicated antibodies as described in Materials and Methods. Immunoblots for β-actin were shown as loading controls. Representative of at least three independent experiments.

Figure 5.

Signaling pathways affected by LJ-529 treatment. Cell lysates were prepared from cells treated with 50 μmol/L LJ-529 for indicated times and subjected to immunoblotting with indicated antibodies as described in Materials and Methods. Immunoblots for β-actin were shown as loading controls. Representative of at least three independent experiments.

Close modal

Down-Regulation of the p27kip Expression by LJ-529 in Breast Cancer Cells

Because Akt activity was reduced upon LJ-529 treatment, and Akt is reported to regulate p27kip (22, 23), we tested the expression of p27kip by immunoblotting. As seen in Fig. 5B, the expression level of p27kip was induced by LJ-529 in a dose-dependent manner. This is intriguing because activated Akt upon phosphorylation at serine-473 is known to phosphorylate p27kip at threonine-157 (22, 23). As a result, the phosphorylated p27kip is excluded from the nucleus and remains in the cytoplasm, where protein degradation occurs ultimately. Therefore, it is plausible to conclude that total p27kip protein accumulated as a consequence of reduced phospho-Akt (at serine-473) by LJ-529 treatment in breast cancer cell lines.

Down-Regulation of ERα by LJ-529 in ER-Positive Breast Cancer Cells

Because an adenosine analogue, IB-MECA, is reported to down-regulate ERα and suppress human breast cancer cell proliferation in MCF7 cells (6), we examined whether LJ-529 acted in a similar manner. As seen in Fig. 5C, down-regulation of ERα occurred by LJ-529 in a time-dependent manner in MCF7 and T47D cell lines. Thus, we conclude that LJ-529 down-regulates ERα, whose signaling is critical in cell proliferation of ER-positive breast cancer cells (6).

Down-Regulation of ErbB2 Signaling by LJ-529 in SK-RB-3 Cells

We further investigated the additional biochemical pathway that may be responsible for growth-suppressive effect in ER-negative breast cancer cells. As SK-BR-3 cells express high levels of c-ErbB2 (24), we examined the expression level of c-ErbB2 in LJ-529-treated SK-BR-3 cells. As seen in Fig. 5D, the expression level of c-ErbB2 was significantly reduced by LJ-529. Furthermore, the expression of total and phosphorylated forms of both ERK1 (p44) and ERK2 (p42) was also significantly reduced by LJ-529. Therefore, we conclude that LJ-529 down-regulated ErbB2 signaling in c-ErbB2-positive SK-BR-3 cells.

Inhibition of Breast Cancer Growth by LJ-529 in Xenograft Models

To examine the in vivo effect of LJ-529 in breast cancer cells, T47D and SK-BR-3 cells were engrafted s.c. into nude mice. When tumor reached the width of 1 to 3 mm, the mice were treated daily p.o. with the indicated doses of LJ-529. Tumor growth was clearly suppressed in the LJ-529-treated group compared with the vehicle-treated group as examined by Student's t test (P < 0.05 at indicated time points; Fig. 6). When one-way ANOVA was done to examine the group differences between tumor growth rates of T47D xenografts, statistical significance was seen between the groups [F(3, 10) = 11.536, P = 0.001]. Upon post hoc test using Scheffe, significant differences between vehicle- and all three LJ-529-treated groups were revealed (the mean differences were significant at the 0.05 level), but not within the drug-treated groups. The average tumor sizes are shown in Table 1. The results show that LJ-529 inhibits breast cancer growth in xenograft models.

Figure 6.

Inhibition of breast cancer growth by LJ-529 in mouse xenograft models. Xenograft experiments were done as described in Materials and Methods. Points, mean calculated as percentage of corresponding tumor sizes of day 0; bars, SE. Each group was treated daily p.o. with vehicle (water; control), 50 μg/mL LJ-529 (low), 500 μg/kg LJ-529 (medium), and 5 mg/kg LJ-529 (high). Results are from at least three animals. P values indicate statistical significance of inhibition of tumor growth by LJ-529 (Student's t test): *, P < 0.05; **, P < 0.01; ***, P < 0.001. A, xenograft tumors of T47D. *, for data points with low doses of LJ-529 at days 24 and 28, medium and high doses at day 17. **, for data points with medium and high doses of LJ-529 at days 21, 24, and 28. B, xenograft tumors of SK-BR-3. **, for data points with medium dose of LJ-529 at days 14 and 18. ***, for data points with low, medium, and high doses of LJ-529 at days 21, 25, and 28.

Figure 6.

Inhibition of breast cancer growth by LJ-529 in mouse xenograft models. Xenograft experiments were done as described in Materials and Methods. Points, mean calculated as percentage of corresponding tumor sizes of day 0; bars, SE. Each group was treated daily p.o. with vehicle (water; control), 50 μg/mL LJ-529 (low), 500 μg/kg LJ-529 (medium), and 5 mg/kg LJ-529 (high). Results are from at least three animals. P values indicate statistical significance of inhibition of tumor growth by LJ-529 (Student's t test): *, P < 0.05; **, P < 0.01; ***, P < 0.001. A, xenograft tumors of T47D. *, for data points with low doses of LJ-529 at days 24 and 28, medium and high doses at day 17. **, for data points with medium and high doses of LJ-529 at days 21, 24, and 28. B, xenograft tumors of SK-BR-3. **, for data points with medium dose of LJ-529 at days 14 and 18. ***, for data points with low, medium, and high doses of LJ-529 at days 21, 25, and 28.

Close modal
Table 1.

The effect of LJ-529 in a xenograft model of breast cancer cells in nude mice

Cell lineTreatmentDay 0Day 7Day 14Day 21Day 28
T47D Control 3.4 ± 0.2 15.1 ± 3.5 38.1 ± 4.0 101.1 ± 13.0 135.4 ± 15.1 
 Low 3.7 ± 0.3 7.1 ± 2.4 20.1 ± 7.3 39.5 ± 21.1 53.9 ± 21.3 
 Medium 3.5 ± 0.3 4.8 ± 0.1 9.1 ± 2.6 21.6 ± 4.7 25.9 ± 10.3 
 High 2.3 ± 0.3 2.6 ± 0.4 9.8 ± 2.7 12.9 ± 0.5 19.8 ± 3.6 
SK-BR-3 Control 22.6 ± 8.1 23.0 ± 8.0 32.1 ± 10.8 34.1 ± 10.4 34.8 ± 10.3 
 Low 19.9 ± 7.9 20.6 ± 7.7 22.8 ± 8.3 16.2 ± 6.6 7.6 ± 3.1 
 Medium 16.3 ± 5.2 17.3 ± 5.1 18.0 ± 5.7 11.8 ± 3.7 5.6 ± 1.8 
 High 16.4 ± 4.6 18.9 ± 4.6 19.5 ± 5.2 12.8 ± 3.4 6.1 ± 1.6 
Cell lineTreatmentDay 0Day 7Day 14Day 21Day 28
T47D Control 3.4 ± 0.2 15.1 ± 3.5 38.1 ± 4.0 101.1 ± 13.0 135.4 ± 15.1 
 Low 3.7 ± 0.3 7.1 ± 2.4 20.1 ± 7.3 39.5 ± 21.1 53.9 ± 21.3 
 Medium 3.5 ± 0.3 4.8 ± 0.1 9.1 ± 2.6 21.6 ± 4.7 25.9 ± 10.3 
 High 2.3 ± 0.3 2.6 ± 0.4 9.8 ± 2.7 12.9 ± 0.5 19.8 ± 3.6 
SK-BR-3 Control 22.6 ± 8.1 23.0 ± 8.0 32.1 ± 10.8 34.1 ± 10.4 34.8 ± 10.3 
 Low 19.9 ± 7.9 20.6 ± 7.7 22.8 ± 8.3 16.2 ± 6.6 7.6 ± 3.1 
 Medium 16.3 ± 5.2 17.3 ± 5.1 18.0 ± 5.7 11.8 ± 3.7 5.6 ± 1.8 
 High 16.4 ± 4.6 18.9 ± 4.6 19.5 ± 5.2 12.8 ± 3.4 6.1 ± 1.6 

NOTE: Control, low, medium, and high groups were treated daily with vehicle (water), 50 μg/kg LJ-529, 500 μg/kg LJ-529, and 5 mg/kg LJ-529, respectively. The tumor volumes (mm3) were presented as average ± SE.

This study presents data showing that the novel A3AR agonist, LJ-529, attenuated in vitro proliferation and in vivo tumor growth of breast cancer cells by inducing apoptosis and deregulating the Wnt signaling pathway. Such effect was seen in both ER-positive and ER-negative breast cancer cells. In addition, ERα expression was down-regulated by LJ-529 in ER-positive cells, which may partially account for the suppression of cell proliferation as IB-MECA does in MCF7 cells (6). Furthermore, c-ErbB2 expression and its downstream ERK activities were down-regulated by LJ-529 in c-ErbB2-overexpressing, ER-negative SK-BR-3 cells. Finally, such effect of LJ-529 occurred independently of the A3AR, because no A3AR expression was detected by RT-PCR in all four breast cancer cell lines used in this study.

The requirement for the signaling via the A3AR has been examined by using the A3AR-specific antagonists, by quantifying the expression by RT-PCR or immunoblotting or by stably overexpressing A3AR. The anticancer effect of A3AR agonists occurs via A3AR in melanoma, prostate, and hepatoma cells (5, 10, 1214). In agreement with these results, A3AR is reported to be highly expressed in primary tumors, such as colon carcinoma, breast carcinoma, small cell lung carcinoma, pancreatic carcinoma, and melanoma (25). However, the action of A3AR agonist has been reported to be independent of the A3AR in leukemia cells (9). The requirement for A3AR in breast cancer cells is controversial because two research groups have drawn opposite conclusions with the identical A3AR agonist (IB-MECA) on identical cells (MCF7). One group has detected A3AR by RT-PCR and observed partial attenuation of anchorage-dependent cell growth by addition of the A3AR antagonist (7), whereas the other detected no A3AR by RT-PCR and no change in colony formation by stably overexpressing A3AR (6). The reasons for such discrepancies are not known, but for the RT-PCR we found that the primers and the reaction conditions are quite different. The former group (7) ran PCR for 40 cycles with the primers as previously reported (26), whereas the latter group (6) did PCR for 33 cycles with newly designed primers. When nucleotide-nucleotide Basic Local Alignment Search Tool was ran to search for short, nearly exact matches, we found that the antisense primer used by the former group significantly lacks the specificity to A3AR sequence and that it only matches a stretch of 13 nucleotides (from the 8th to 20th nucleotide) out of a 20-mer. In this study, we adopted the conditions of the latter group because the primers were highly specific to A3AR upon Basic Local Alignment Search Tool analysis and detected no A3AR expression in all four cell lines tested.

Despite elucidating several signaling pathways affected by LJ-529 in breast cancer cells, it is not clear how LJ-529 triggers the effect on proliferation and apoptosis in breast cancer cells. One possibility is that LJ-529 at high concentrations binds to a yet unidentified membrane receptor(s) and triggers downstream signaling. Another possibility would be that LJ-529 is transported into the cell via a nucleoside transporter, and the transported compound signals through direct interaction with intracellular targets as reported for adenosine (27) and 2-chloroadenosine (28). Because the growth inhibitory effect of IB-MECA in MCF7 cells was not prevented using the nucleoside transporter inhibitor, Lu et al. (6) have suggested that IB-MECA at high concentrations may compete for the transporter or enter the cell by a nucleoside transporter–independent mechanism. The signaling pathways from the membrane to the cytosol or the transporters of A3AR agonists in cells that do not express A3AR are intriguing and remain to be explored.

The antiproliferative and apoptotic effect of LJ-529 was seen with relatively high doses of the drug at tens-of-micromolar concentrations. Similarly, high doses of IB-MECA are used to attenuate cell growth of MCF7 breast cancer cells (6, 7). Because LJ-529 was effective on all four breast cancer cell lines tested, it is possible that the results were due to toxic effects of the drug at high concentrations. However, we believe this is not the case because we did not observe any toxicity as evidenced by no change in body weight in the animals p.o. treated with LJ-529 for up to 1 month. Therefore, we present the effect of LJ-529 as genuine.

The c-ERBB2 gene is amplified and c-ErbB2 is overexpressed in 25% to 30% of breast cancers, increasing the aggressiveness of the tumor (16). It has also been implicated in the development of resistance to the antiestrogen tamoxifen in both advanced disease and in an adjuvant setting (17). Herceptin, a humanized monoclonal antibody against c-ErbB2, is used clinically in breast cancers that overexpress c-ErbB2 because it seems to block growth signals transmitted by c-ErbB2 to the nucleus and enhances response to chemotherapeutic agents (29, 30). Here, we showed that the expression of c-ErbB2 and it downstream ERK signaling were down-regulated by LJ-529 in c-ErbB2-overexpressing SK-BR-3 cells. Although more study needs to be done to generalize the effect of LJ-529 in c-ErbB2-overexpressing breast cancers, such as testing the effectiveness of the drug in c-ErbB2 transgenic mice, our study strongly suggest LJ-529 as a possible therapeutic agent for this type of aggressive breast cancer.

Recent report of Merighi et al. (14) has shown that Cl-IB-MECA inhibits phosphatidylinositol 3-kinase/Akt–dependent ERK phosphorylation in melanoma cells. Although we have not explored this signaling pathway in breast cancer cells with LJ-529, we reported down-regulation of total and phospho-Akt by LJ-529 in T47D and SK-BR-3 cells. It remains to be seen if ERK phosphorylation is down-regulated in these cells and it is dependent on the inhibition of phosphatidylinositol 3-kinase/Akt by using phosphatidylinositol 3-kinase inhibitors. Apart from the phosphatidylinositol 3-kinase/Akt–dependent regulation of ERKs, we report a novel pathway of c-ErbB2-dependent down-regulation of ERK phosphorylation in SK-BR-3 breast cancer cells by LJ-529.

In summary, we presented LJ-529, an A3AR agonist, as an effective drug that acted independently of the A3AR activity, but with antiproliferative and apoptotic activity in breast cancer cells. We also presented several molecular mechanisms responsible for the effects, and identified a novel pathway of down-regulation of c-ErbB2 by an A3AR agonist in SK-BR-3 breast cancer cells both in vitro and in vivo. Therefore, we suggest LJ-529 as a novel drug that may be effective in the treatment of breast cancer.

Grant support: Hanyang University grant HY-2003-T (H. Chung).

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
Yan L, Burbiel JC, Maass A, Muller CE. Adenosine receptor agonists: from basic medicinal chemistry to clinical development.
Expert Opin Emerg Drugs
2003
;
8
:
537
–76.
2
Fishman P, Bar-Yehuda S, Madi L, Cohn I. A3 adenosine receptor as a target for cancer therapy.
Anticancer Drugs
2002
;
13
:
437
–43.
3
Fishman P, Bar-Yehuda S, Barer F, Madi L, Multani AS, Pathak S. The A3 adenosine receptor as a new target for cancer therapy and chemoprotection.
Exp Cell Res
2001
;
269
:
230
–6.
4
Bar-Yehuda S, Barer F, Volfsson L, Fishman P. Resistance of muscle to tumor metastases: a role for A3 adenosine receptor agonists.
Neoplasia
2001
;
3
:
125
–31.
5
Fishman P, Bar-Yehuda S, Ardon E, et al. Targeting the A3 adenosine receptor for cancer therapy: inhibition of prostate carcinoma cell growth by A3AR agonist.
Anticancer Res
2003
;
23
:
2077
–83.
6
Lu J, Pierron A, Ravid K. An adenosine analogue, IB-MECA, down-regulates estrogen receptor α and suppresses human breast cancer cell proliferation.
Cancer Res
2003
;
63
:
6413
–23.
7
Panjehpour M, Karami-Tehrani F. An adenosine analog (IB-MECA) inhibits anchorage-dependent cell growth of various human breast cancer cell lines.
Int J Biochem Cell Biol
2004
;
36
:
1502
–9.
8
Ohana G, Bar-Yehuda S, Arich A, et al. Inhibition of primary colon carcinoma growth and liver metastasis by the A3 adenosine receptor agonist CF101.
Br J Cancer
2003
;
89
:
1552
–8.
9
Kim SG, Ravi G, Hoffmann C, et al. p53-Independent induction of Fas and apoptosis in leukemic cells by an adenosine derivative, Cl-IB-MECA.
Biochem Pharmacol
2002
;
63
:
871
–80.
10
Wen LT, Knowles AF. Extracellular ATP and adenosine induce cell apoptosis of human hepatoma Li-7A cells via the A3 adenosine receptor.
Br J Pharmacol
2003
;
140
:
1009
–18.
11
Harish A, Hohana G, Fishman P, Arnon O, Bar-Yehuda S. A3 adenosine receptor agonist potentiates natural killer cell activity.
Int J Oncol
2003
;
23
:
1245
–9.
12
Madi L, Bar-Yehuda S, Barer F, Ardon E, Ochaion A, Fishman P. A3 adenosine receptor activation in melanoma cells: association between receptor fate and tumor growth inhibition.
J Biol Chem
2003
;
278
:
42121
–30.
13
Fishman P, Madi L, Bar-Yehuda S, Barer F, Del Valle L, Khalili K. Evidence for involvement of Wnt signaling pathway in IB-MECA mediated suppression of melanoma cells.
Oncogene
2002
;
21
:
4060
–4.
14
Merighi S, Benini A, Mirandola P, et al. A3 adenosine receptor activation inhibits cell proliferation via phosphatidylinositol 3-kinase/Akt-dependent inhibition of the extracellular signal-regulated kinase 1/2 phosphorylation in A375 human melanoma cells.
J Biol Chem
2005
;
280
:
19516
–26.
15
Jeong LS, Jin DZ, Kim HO, et al. N6-substituted d-4′-thioadenosine-5′-methyluronamides: potent and selective agonists at the human A3 adenosine receptor.
J Med Chem
2003
;
46
:
3775
–7.
16
Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer.
Science
1989
;
244
:
707
–12.
17
De Placido S, Carlomagno C, De Laurentiis M, Bianco AR. c-erbB2 expression predicts tamoxifen efficacy in breast cancer patients.
Breast Cancer Res Treat
1998
;
52
:
55
–64.
18
Sun S, Ravid K. Role of a serine/threonine kinase, Mst1, in megakaryocyte differentiation.
J Cell Biochem
1999
;
76
:
44
–60.
19
Choi BY, Kim HY, Lee KH, Cho YH, Kong G. Clofilium, a potassium channel blocker, induces apoptosis of human promyelocytic leukemia (HL-60) cells via Bcl-2-insensitive activation of caspase-3.
Cancer Lett
1999
;
147
:
85
–93.
20
Chung H, Hong D-P, Jung J-Y, et al. Comprehensive analysis of differential gene expression profiles on carbon tetrachloride-induced rat liver injury and regeneration.
Toxicol Appl Pharmacol
2005
;
206
:
27
–42.
21
Tetsu O, McCormick F. β-catenin regulates expression of cyclin D1 in colon carcinoma cells.
Nature
1999
;
398
:
422
–6.
22
Viglietto G, Motti ML, Bruni P, et al. Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer.
Nat Med
2002
;
8
:
1136
–44.
23
Shin I, Yakes FM, Rojo F, et al. PKB/Akt mediates cell-cycle progression by phosphorylation of p27(Kip1) at threonine 157 and modulation of its cellular localization.
Nat Med
2002
;
8
:
1145
–52.
24
deFazio A, Chiew YE, Sini RL, Janes PW, Sutherland RL. Expression of c-erbB receptors, heregulin and oestrogen receptor in human breast cell lines.
Int J Cancer
2000
;
87
:
487
–98.
25
Madi L, Ochaion A, Rath-Wolfson L, et al. The A3 adenosine receptor is highly expressed in tumor versus normal cells: potential target for tumor growth inhibition.
Clin Cancer Res
2004
;
10
:
4472
–9.
26
Gessi S, Varani K, Merighi S, et al. Pharmacological and biochemical characterization of A3 adenosine receptors in Jurkat T cells.
Br J Pharmacol
2001
;
134
:
116
–26.
27
Schrier SM, van Tilburg EW, van der Meulen H, Ijzerman AP, Mulder GJ, Nagelkerke JF. Extracellular adenosine-induced apoptosis in mouse neuroblastoma cells: studies on involvement of adenosine receptors and adenosine uptake.
Biochem Pharmacol
2001
;
61
:
417
–25.
28
Barbieri D, Abbracchio MP, Salvioli S, et al. Apoptosis by 2-chloro-2′-deoxy-adenosine and 2-chloro-adenosine in human peripheral blood mononuclear cells.
Neurochem Int
1998
;
32
:
493
–504.
29
Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2.
N Engl J Med
2001
;
344
:
783
–92.
30
Pietras RJ, Pegram MD, Finn RS, Maneval DA, Slamon DJ. Remission of human breast cancer xenografts on therapy with humanized monoclonal antibody to HER-2 receptor and DNA-reactive drugs.
Oncogene
1998
;
17
:
2235
–49.