Upregulation of EZH2 is associated with advanced stage and poor prognosis of prostate cancer; therefore, it is likely to be a promising therapeutic target. Metformin, a drug that has been used to treat type 2 diabetes, was found to have antineoplastic activity in different cancers. Herein, we report that the combination of metformin and the EZH2 inhibitor GSK126 exerts synergistic inhibition on prostate cancer cell growth, both in vitro and in vivo. Mechanistically, we identify that metformin can reduce EZH2 expression through upregulating miR-26a-5p, which is antagonized by androgen receptor (AR). Furthermore, we show that AR binds to the promoter of miR-26a-5p and suppresses its transcription. Although metformin can remove AR from the miR-26a-5p promoter, the interaction between AR and EZH2, which usually exists in androgen-refractory prostate cancer cells, strongly impedes the removal. However, GSK126 can inhibit the methyltransferase-dependent interaction between AR and EZH2, thus restoring metformin's efficacy in androgen-refractory prostate cancer cells. Collectively, our finding suggests that the combination of metformin and GSK126 would be an effective approach for future prostate cancer therapy, and particularly effective for AR-positive castration-resistant prostate cancer.

Prostate cancer is the second leading cause of cancer-related death in males in the United States, with 174,650 new cases and 31,620 deaths estimated in 2019 (1). Despite the benefit from local therapy and initial hormone therapy, the majority of patients will relapse with castration-resistant prostate cancer (CRPC), which might progress to metastatic disease. Some drugs, such as abiraterone acetate, enzalutamide, docetaxol, sepuleucel-T, have been approved by the FDA to treat advanced prostate cancer, but they can only prolong patients' life for 3 to 6 months (2–5). Therefore, discovery of new therapy strategies is urgently needed.

Metformin (N, N-dimethylbiguianide), the most commonly used oral drug to treat type II diabetes, has a good safety profile and limited side effects. Increasing observational and cohort studies have shown that patients with diabetes who were treated with metformin usually exhibited lower risk of cancer (6), indicating that it is feasible to repurpose metformin as an anti-cancer drug. In prostate cancer, metformin was observed to reduce prostate cancer incidence and slow down the development of CRPC (7, 8). Moreover, a phase II trial observed prostate-specific antigen (PSA) secretion was decreased by the use of a high-dose metformin in progressive metastatic CRPC (9). Recently, a population-based cohort study showed encouraging results that metformin use after diagnosis of prostate cancer might increase survival of patients (10). All of these suggest that metformin could be a useful medication for prostate cancer therapy, but more studies are still needed to further evaluate such a notion.

Enhancer of zeste homolog 2 (EZH2), the catalytic subunit of polycomb-repressive complex 2, suppresses expression of a number of genes via catalyzing histone-3 lysine 27 trimethylation (H3K27me3; ref. 11). Accumulating evidence has shown that EZH2 plays an important role in tumor oncogenesis and progression (12). GSK126 (GSK2816126; ref. 13) is an S-adenosyl-methionine–competitive inhibitor targeting EZH2, and it has recently been shown effective and well tolerated in lymphoma (14). However, EZH2 also acts independently of its methyltransferase activity. For instance, EZH2 can activate NK-κB by forming a complex with RelA and RelB, in which methylation is not involved (15). In prostate cancer, EZH2 can increase AR's expression by binding to its promoter, which is also methylation-independent (16). Therefore, although several EZH2 methyltransferase inhibitors, including GSK126, are proved effective, suppression of EZH2's protein level by drugs should be considered to enhance therapy targeting EZH2.

In our study, we found that the combination of metformin and GSK126 exerts a synergistic antiproliferative effect in prostate cancer cell lines and in human prostate tumor explants. Furthermore, we demonstrate that metformin can induce downregulation of EZH2 via upregulating miR-26a-5p in LNCaP. Although metformin's role was strongly hampered by the interaction between androgen receptor (AR) and EZH2 in 22Rv1, it can be restored when combined with GSK126. All of these suggest that the combination of metformin and GSK126 is effective, and acts, at least partially, through inhibition on both EZH2's expression and methyltransferase activity.

Cell culture, chemicals, and reagents

LNCaP, 22Rv1, PC3-Neo, and PC3-AR were used in this study. LNCaP and 22Rv1 were purchased from the ATCC. PC3-Neo and PC3-AR were kindly provided by Dr. Kerry Burnstein (University of Miami). All cells were grown in RPMI-1640 medium supplemented with 10% FBS in a humidified incubator at 37°C with 5% CO2. All cells were within 50 passages and Mycoplasma were detected every 3 months using MycoAlert PLUS Mycoplasma Detection Kit (Lonza, LT07-705). R1881 was purchased from Sigma, metformin and GSK126 were obtained from Selleckchem.

Antibodies

Whereas antibodies against caspase-9, cleaved caspase-9, caspase-3, cleaved caspase-3, PARP, cleaved PARP, H3, β-actin were purchased from Cell Signaling Technology, antibodies against AR, EZH2, and H3K27me3 were obtained from Active Motif. Antibody against Ki-67 was purchased from Abcam.

Immunoblotting

Upon harvest, cells were suspended with TBSN buffer (20 mmol/L Tris-HCl, pH 8.0, 0.5% NP-40, 5 mmol/L EGTA, 1.5 mmol/L EDTA, 0.5 mmol/L sodium vanadate, and 150 mmol/L NaCl) with protease inhibitors and phosphatase inhibitors, sonicated and then collected, followed by protein concentration measurement by Protein Assay Dye Reagent from Bio-Rad. Equal amounts of protein lysates from each sample were mixed with SDS loading buffer, resolved by SDS-PAGE, transferred to PVDF membranes, followed by incubations with appropriate primary and secondary antibodies.

RNA isolation and quantitative real-time PCR

Total RNA was extracted from tissues or cells using RNeasy mini kit (Qiagen) and reverse transcribed into cDNA using miScript II RT kit (Qiagen). FastStart Universal SYBR Green Master (Roche) was used to measure the expression level of mRNA. The primers used are: EZH2, tccctagtcccgcgcaatgagc (forward), ttgttggcggaagcgtgtaaaatc (reverse); β-actin, agaactggcccttcttggagg (forward), gtttttatgttctatggg (reverse). For the detection of microRNAs, specific primers (MS0029239, MS00008372, MS00031220, MS00003122, MS00003129) were purchased from Qiagen, and cDNA was amplified using miScript SYBR Green PCR Master Mix (Qiagen). The relative expression level of miRNA or mRNA was normalized to RNU6-2 or β-actin, respectively.

Colony formation assay

Cells (0.5 × 103) were seeded in 6-well plates with 2 mL RPMI-1640 supplemented with 10% FBS. One day later, the medium was replaced with new medium containing different drugs. After 12 days, the colonies were fixed by 10% formalin and stained with 5% crystal violet. Colony numbers were counted by using ImageJ software.

Cell viability assay

A total of 5,000 prostate cancer cells were seeded into 96-well plates, and treated with different drugs with indicated concentrations for 72 hours, followed by incubation with the tetrazolium dye MTT 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide for 4 hours. After the purple formazan was dissolved by DMSO, absorbance at 570 nm was measured by a plate reader.

Combination index

Cytotoxicity of the drugs was evaluated via determining viability by MTT assay. Combination indices (CI) were calculated by the multiple drug effect equation of Chou and colleagues (17). CI = (D)1/(Dx)1 + (D)2/(Dx)2, where (Dx)1 and (Dx)2 in the denominators are the doses for metformin and GSK126 alone that gives x% inhibition, whereas (D)1 and (D)2 in the numerators are the doses of metformin and GSK126 in combination that also inhibited x%. Antagonism is indicated when CI > 1, CI = 1 indicates an additive effect, and CI < 1 means synergy.

ChIP and Re-ChIP

Chromatin immunoprecipitation (ChIP) assay was performed by using a commercial kit (Millipore, #17-10085) following the manufacturer's instruction. AR-binding sites were predicted by PROMO (18, 19), and the PCR generated approximately 200 bp products from the miR-26a-5p proximal (<2,000 bp) promoter-containing sites. Antibodies against AR (#39781) and EZH2 (#39901) were purchased from Active Motif. For Re-ChIP, the immunoprecipitated protein–DNA complexes were eluted with Re-ChIP elution buffer (1 × TE, 2% SDS, 15 mmol/L DTT) at 37°C for 30 minutes, and the elutes were diluted 20-fold with ChIP dilution buffer for further incubation with the secondary antibodies and beads. The primers used are: P1, gttgtgggtccaagtacaaatagttttcc (forward), caatatcacctgcctggcctcaa (reverse); P2, gaatttcagaagtttccgtatcccccac (forward), cttttggggtgggtatttgctaaagat (reverse); P3, aattaaaatgaaaattccagtctcctgcctcc (forward), gatggcttttaaaagcatgaagtgtgga (reverse); P4, gcaatagaatgcagaccgatggg (forward), ctatgggagctttctgtccttggc (reverse).

Luciferase assay

HEK293T or PC3 cells were transfected with the indicated plasmids using lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions. Briefly, 0.1 μg of reporter plasmids containing the sequence of interest, together with 5 ng of an internal control plasmid pRL-TK, were transfected into the cells cultured in a 24-well plate. For the expression of AR, EZH2 (S21D) and EZH2 (S21A), the indicated amounts of plasmids were co-transfected into the cells. Total plasmid DNA was normalized to 0.6 μg per well by using an empty plasmid. Luciferase activity was assayed after 24 hours of transfection using a Dual Luciferase reporter assay system (Promega). The firefly luciferase activities were corrected by the corresponding Renilla luciferase activities and presented as means ± S.D.

22Rv1-derived mouse xenograft model

All the animal experiments were approved by the Purdue University Animal Care and Use Committee. 22Rv1 cells (2.5 × 105/mouse) were mixed with Matrigel (Collaborative Biomedical Products), and the mixture was injected subcutaneously into right flanks of castrated nude mice (Harlan Laboratories). After 2 weeks, the tumor-bearing mice were randomized into control and treatment groups (four mice/group). Metformin was dissolved in water and administered to mice via oral gavage (30 mg/kg body weight/d). GSK126 in 20% captisol with PH adjusted to 4–4.5 was injected intraperitoneally into mice (50 mg/kg body weight/d; refs. 20, 21). Tumor volumes were calculated from the formula V = L × W2/2 [where V is volume (cubic millimeters), L is length (millimeters), and W is width (millimeters)].

LuCaP35CR xenograft model

Mice bearing LuCaP35CR tumors were obtained from Dr. Robert Vessella at the University of Washington. Tumors were implanted and amplified in pre-castrated NSG mice. When tumor size was big enough, tumors were harvested and cut into approximately 25 mm3 pieces, followed by implantation into 16 pre-castrated NSG mice. After tumor size reached approximately 200 mm3, mice were randomized into four groups, followed by similar treatment and measurement as described above.

Histology and IHC

Xenograft tumors were fixed in 10% neutral-buffered formalin, paraffin-embedded, sectioned to 5 mm, and stained using conventional hematoxylin and eosin (H&E) staining. Immunofluorescent chemistry staining was accomplished with the M.O.M.TM kit from Vector Laboratories.

PSA measurement

Blood was collected from mice by retro-orbital bleeding, followed by centrifuge to collect serum. PSA levels were determined using a PSA ELISA kit (Abnova KA0208) as manufacturer instructed.

Statistical analysis

All numerical data are presented as mean ± SD. The statistical significance of the results was analyzed by using unpaired two-tailed Student t test. The P values of <0.05 indicate statistical significance.

Metformin and GSK126 synergistically inhibit growth of prostate cancer cells

To investigate whether metformin and GSK126 act synergistically to inhibit the growth of prostate cancer cells, colony formation assay was conducted with LNCaP, 22Rv1, and RWPE-1. Compared with treatment of metformin or GSK126 alone, combination of the two drugs exerted a stronger inhibitory effect on colony formation by LNCaP and 22Rv1 (Fig. 1A and B), but not the colony formation by RWPE-1 (Fig. 1C), a non-transformed prostate epithelial cell line. In comparison with mono-treatments, the combination of metformin and GSK126 also led to a greater inhibitory effect on cell survival of LNCaP and 22Rv1 (Fig. 1D and E), but RWPE1 cells were not affected (Fig. 1F). Moreover, the role of apoptosis was investigated upon different treatments. In LNCaP, the treatment with metformin or GSK126 as a single agent could induce slight apoptosis, but no combinational effect was observed (Fig. 1G; Supplementary Fig. S1A). In contrast, neither metformin nor GSK126 alone induced apoptosis in 22Rv1, but there was a dramatic increase of apoptosis induced by the combination (Fig. 1H; Supplementary Fig. S1B). Finally, CIs were measured to determine the types of drug interactions. As shown in Fig. 1I and J, the combinations exhibited slight to moderate synergy in LNCaP (CI range, 0.87–0.72), and moderate to strong synergy in 22Rv1 (CI range, 0.67–0.33). Altogether, these results demonstrate that the combination of metformin and GSK126 exerts synergistic inhibitory effect on prostate cancer cell growth.

Figure 1.

Metformin and GSK126 in combination synergistically inhibit growth of prostate cancer cells. A–C, LNCaP, 22Rv1, or RWPE1 cells were plated into 6-well plates and treated with metformin (0.5 mmol/L), GSK126 (2.5 μmol/L), or both for 12 days, followed by crystal violet staining to monitor colony formation. Data shown are representative of data from three repeats. The number of colonies was quantified by using ImageJ software (means ± SD; n = 3 independent experiments). *, P ≤ 0.05; **, P ≤ 0.01. D–F, LNCaP, 22Rv1, or RWPE1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 72 hours, followed by MTT assay. The results represent the mean of three independent experiments. *, P ≤ 0.05; **, P ≤ 0.01. G and H, LNCaP and 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by immunoblotting against pro- and cleaved-PARP and caspases. I and J, Combination indices of metformin and GSK126 in 22Rv1 and LNCaP cells.

Figure 1.

Metformin and GSK126 in combination synergistically inhibit growth of prostate cancer cells. A–C, LNCaP, 22Rv1, or RWPE1 cells were plated into 6-well plates and treated with metformin (0.5 mmol/L), GSK126 (2.5 μmol/L), or both for 12 days, followed by crystal violet staining to monitor colony formation. Data shown are representative of data from three repeats. The number of colonies was quantified by using ImageJ software (means ± SD; n = 3 independent experiments). *, P ≤ 0.05; **, P ≤ 0.01. D–F, LNCaP, 22Rv1, or RWPE1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 72 hours, followed by MTT assay. The results represent the mean of three independent experiments. *, P ≤ 0.05; **, P ≤ 0.01. G and H, LNCaP and 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by immunoblotting against pro- and cleaved-PARP and caspases. I and J, Combination indices of metformin and GSK126 in 22Rv1 and LNCaP cells.

Close modal

Metformin is capable of suppressing EZH2 expression in prostate cancer cells

Increasing evidence shows that EZH2 is usually upregulated in prostate cancer, and closely associated with progression, invasion, and metastasis (22, 23). Therefore, we were prompted to investigate the role of EZH2 in the antiproliferative effect induced by the treatments. As shown in Fig. 2A, metformin alone significantly suppressed EZH2's expression in androgen-sensitive LNCaP cells, and notably, the combined treatment resulted in enhanced inhibition on EZH2's activity indicated by the level of H3K27me3. In contrast, 22Rv1 cells, which are androgen-independent, displayed a limited reduction of EZH2 when treated with metformin alone, but it was decreased significantly by co-treatment of metformin and GSK126 (Fig. 2B), indicating that GSK126 restored metformin's ability of downregulating EZH2. To further explore how EZH2 was degraded, the relative mRNA level was measured by qRT-PCR. Consistently, metformin alone significantly reduced EZH2 mRNA level in LNCaP (Fig. 2C), but the combination of metformin and GSK126, instead of mono-treatments, was required to decrease EZH2 mRNA level in 22Rv1 cells (Fig. 2D). It is worthy noticing that the EZH2 expression displayed a similar changing trend with the cell growth upon indicated treatments in both cell lines, so we wondered whether the growth inhibition of prostate cancer cells is indeed due to EZH2 reduction. To test this hypothesis, we expressed exogenous EZH2 in LNCaP and 22Rv1, and then treated them with indicated drugs. We found that overexpression of EZH2 partially rescued the cells from the growth inhibition induced by the treatments (Fig. 2E and F). In summary, these results indicate metformin-induced downregulation of EZH2 expression is antagonized in 22Rv1 cells, but such an ability can be restored by co-treatment with GSK126. Also, downregulation of EZH2 is one of the reasons contributing to the antiproliferative effect induced by the treatments.

Figure 2.

Metformin downregulates EZH2 expression by regulating miR-26a-5p. A and B, LNCaP or 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by immunoblotting (IB). C and D, LNCaP and 22Rv1 cells were treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by qRT-PCR. E and F, LNCaP and 22Rv1 cells were transfected with EZH2 and pcDNA3.0, followed by 72-hour cell viability assay with indicated treatments (metformin: 1 mmol/L; GSK126: 5 μmol/L; means ± SD; n = 3). *, P ≤ 0.05; **, P ≤ 0.01. Meanwhile, some cells were harvested for Western blot to test EZH2 level after the treatments. G–I, qRT-PCR shows the expression of miR-26a-5p, miR-101-3p, let-7a-5p, let-7b-5p, and let-7c-5p of LNCaP cells treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or combination for 48 hours, with all microRNA expressions being normalized to RNU6-2. J, LNCaP cells were transfected with the miR-26a-5p inhibitor or negative control miRNA inhibitor, then treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by IB. K, LNCaP cells were transfected with the miR-101-3p inhibitor or the negative control miRNA inhibitor, then treated with metformin (1 mmol/L) for 48 hours and harvested for IB. L, LNCaP cells were transfected with inhibitors targeting let-7a-5p, let-7b-5p, and let-7c-5p or the negative control miRNA inhibitor, then treated with metformin (1 mmol/L) for 48 hours and harvested for IB. M, qRT-PCR shows the expression of miR-26a-5p or 22Rv1 cells treated with metformin (1 mmol/L), GSK126 (5 μmol/L) or both for 48 hours, with miR-26a-5p expression being normalized to RNU6-2. N, 22Rv1 cells were transfected with miR-26a-5p inhibitor or negative control miRNA inhibitor, then treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours and harvested for IB. O, 22Rv1 cells were transfected with miR-26a-5p inhibitor or negative control miRNA inhibitor, treated with DMSO or the combination of metformin (1 mmol/L) and GSK126 (5 μmol/L) for 72 hours, followed by MTT assay (means ± SD; n = 3). *, P ≤ 0.05; **, P ≤ 0.01.

Figure 2.

Metformin downregulates EZH2 expression by regulating miR-26a-5p. A and B, LNCaP or 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by immunoblotting (IB). C and D, LNCaP and 22Rv1 cells were treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by qRT-PCR. E and F, LNCaP and 22Rv1 cells were transfected with EZH2 and pcDNA3.0, followed by 72-hour cell viability assay with indicated treatments (metformin: 1 mmol/L; GSK126: 5 μmol/L; means ± SD; n = 3). *, P ≤ 0.05; **, P ≤ 0.01. Meanwhile, some cells were harvested for Western blot to test EZH2 level after the treatments. G–I, qRT-PCR shows the expression of miR-26a-5p, miR-101-3p, let-7a-5p, let-7b-5p, and let-7c-5p of LNCaP cells treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or combination for 48 hours, with all microRNA expressions being normalized to RNU6-2. J, LNCaP cells were transfected with the miR-26a-5p inhibitor or negative control miRNA inhibitor, then treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours, followed by IB. K, LNCaP cells were transfected with the miR-101-3p inhibitor or the negative control miRNA inhibitor, then treated with metformin (1 mmol/L) for 48 hours and harvested for IB. L, LNCaP cells were transfected with inhibitors targeting let-7a-5p, let-7b-5p, and let-7c-5p or the negative control miRNA inhibitor, then treated with metformin (1 mmol/L) for 48 hours and harvested for IB. M, qRT-PCR shows the expression of miR-26a-5p or 22Rv1 cells treated with metformin (1 mmol/L), GSK126 (5 μmol/L) or both for 48 hours, with miR-26a-5p expression being normalized to RNU6-2. N, 22Rv1 cells were transfected with miR-26a-5p inhibitor or negative control miRNA inhibitor, then treated with metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours and harvested for IB. O, 22Rv1 cells were transfected with miR-26a-5p inhibitor or negative control miRNA inhibitor, treated with DMSO or the combination of metformin (1 mmol/L) and GSK126 (5 μmol/L) for 72 hours, followed by MTT assay (means ± SD; n = 3). *, P ≤ 0.05; **, P ≤ 0.01.

Close modal

Metformin suppresses EZH2 expression via upregulating miR-26a-5p

Next, we aimed to further dissect the underlying mechanism for metformin-mediated EZH2 downregulation. Metformin has been reported to target a variety of microRNAs (24, 25), and some of them, including miR-26a, miR-101, let-7a, let-7b, and let-7c, directly regulate EZH2 expression in prostate cancer (26). To examine whether these microRNAs are responsible for metformin-induced inhibition on EZH2's expression, RT-PCR was used to determine their expression levels in LNCaP treated with metformin. As shown in Fig. 2G and H, metformin, instead of GSK126, enhanced the expression of miR-26a-5p and miR-101-3p. However, LNCaP cells treated with metformin exhibited decreased expression levels of let-7a-5p, let-7b-5p, and let-7c-5p (Fig. 2I), which could not explain metformin-induced EZH2 downregulation. To further verify these observations, LNCaP cells were transfected with miRNA inhibitors, which are small, double-stranded RNA molecules designed to inhibit specific mature miRNAs, and then treated with metformin. As shown in Fig. 2J, metformin-induced reduction of EZH2 was restored by miR-26a-5p inhibitor, but not by the inhibitors targeting miR-101-3p (Fig. 2K), let-7a-5p, let-7b-5p, and let-7c-5p (Fig. 2L), indicating that miR-26a-5p is the mediator of metformin-induced EZH2 downregulation. Moreover, the miR-26a-5p level was also assessed in 22Rv1 upon indicated treatments, and we found that although metformin alone failed to upregulate miR-26a-5p, the combined treatment enhanced it (Fig. 2M). To further validate this finding, we applied the miRNA inhibitor to block the function of miR-26a-5p. We found that the reduction of EZH2 expression level induced by the combination treatment was restored upon addition of miR-26a-5p inhibitor (Fig. 2N). Also, as EZH2 increased, more 22Rv1 cells survived upon the combination treatment (Fig. 2O). Collectively, we conclude that metformin-induced downregulation of EZH2 is through upregulating miR-26a-5p in LNCaP and 22Rv1.

Metformin-induced EZH2 downregulation is affected by AR

To investigate which factors regulate metformin's effect on EZH2 level in prostate cancer cells, we compared the responses with metformin in several prostate cancer cell lines, including PC3, DU145, LNCaP, and 22Rv1. As indicated, the low-concentration metformin was capable to significantly decrease EZH2 protein levels in PC3 and DU145 (Fig. 3A and B), both of which are AR negative (Supplementary Fig. S1A). In LNCaP, which is AR positive and androgen sensitive, metformin inhibited EZH2 expression as well, but with a lower inhibition efficiency (Fig. 3C). However, metformin failed to reduce EZH2 of 22Rv1, which is AR positive and androgen-refractory (Fig. 3D). Because AR, as well as its cofactors, plays a key role in prostate cancer progression and acquisition of drug resistance, we hypothesized that AR might impede metformin's ability of downregulating EZH2. To test this hypothesis, we assessed the effect of synthetic androgen (R1881) stimulation of AR on EZH2 expression upon metformin treatment. As shown in Fig. 3E, EZH2 protein level was significantly decreased by metformin, accompanied by a reduction of AR activity in LNCaP. However, metformin-induced downregulation of EZH2 in LNCaP was partially restored by addition of R1881. Meanwhile, we also detected the level of miR-26a-5p, and found that treatment of LNCaP with R1881 significantly abolished metformin-induced re-expression of miR-26a-5p (Fig. 3F). To further confirm this, two engineered PC3 cell lines were used. PC3-AR contains the coding region of human AR and stably expresses it, whereas PC3-Neo contains the same vector without the AR cDNA sequence. PC3-AR and PC3-Neo cells were treated with metformin, followed by western blot to determine EZH2 protein levels. As shown in Fig. 3G, PC3-Neo, rather than PC3-AR, displayed a remarkable decreasing trend of EZH2 expression as metformin concentration increases. Accordingly, miR-26a-5p was upregulated upon the treatment of metformin in PC3-Neo, but not in PC3-AR (Fig. 3H). Finally, we constructed two 22Rv1 cell lines with stable knockdown of AR, and treated them with metformin. As expected, depletion of AR led to a significant decrease of EZH2 protein level (Fig. 3I) and an increase of miR-26a-5p in response to metformin treatment (Fig. 3J), further supporting the notion that AR affects metformin-induced EZH2 downregulation.

Figure 3.

AR affects prostate cancer cells' response to metformin. A–D, PC3, DU145, LNCaP, and 22Rv1 cells were treated with metformin of indicated concentrations for 48 hours and harvested for immunoblotting (IB). E and F, LNCaP cells were treated with 10 nmol/L R1881 or metformin or metformin plus R1881 for 48 hours, followed by IB. Meanwhile, mRNA was extracted for the detection of levels of miR-26a-5p. G, PC3 (-AR or -Neo) cells were treated with metformin of indicated concentrations, as well as 10 nmol/L R1881 to activate AR, and subjected to IB. H, mRNA was extracted from PC3-Neo and PC3-AR cells treated with 1 mmol/L metformin for 48 hours, followed by qRT-PCR to test the levels of miR-26a-5p. I and J, 22Rv1 cells were stably transfected with sh-control, sh-AR #3, and sh-AR #4, and treated with 1 mmol/L metformin for 48 hours, followed by IB to test EZH2 protein level and qRT-PCR to detect miR-26a-5p level.

Figure 3.

AR affects prostate cancer cells' response to metformin. A–D, PC3, DU145, LNCaP, and 22Rv1 cells were treated with metformin of indicated concentrations for 48 hours and harvested for immunoblotting (IB). E and F, LNCaP cells were treated with 10 nmol/L R1881 or metformin or metformin plus R1881 for 48 hours, followed by IB. Meanwhile, mRNA was extracted for the detection of levels of miR-26a-5p. G, PC3 (-AR or -Neo) cells were treated with metformin of indicated concentrations, as well as 10 nmol/L R1881 to activate AR, and subjected to IB. H, mRNA was extracted from PC3-Neo and PC3-AR cells treated with 1 mmol/L metformin for 48 hours, followed by qRT-PCR to test the levels of miR-26a-5p. I and J, 22Rv1 cells were stably transfected with sh-control, sh-AR #3, and sh-AR #4, and treated with 1 mmol/L metformin for 48 hours, followed by IB to test EZH2 protein level and qRT-PCR to detect miR-26a-5p level.

Close modal

AR directly suppresses miR-26a transcription by binding to its promoter

Next, we investigated whether AR could bind to the promoter of miR-26a-5p and directly regulate its expression in prostate cancer cells. After assessing the 2-kb region of genomic DNA upstream of miR-26a-5p using PROMO, we identified eight potential binding motifs for AR lying within −1,955 to −1,903, −1,136 to −1,128, −488 to −449 regions on chromosome 3, and −189 to −12 regions on chromosome 12 (Fig. 4A). To examine whether AR could physically bind to the promoter of miR-26a-5p, ChIP-PCR assays were performed in 22Rv1 and LNCaP. Two AR-binding sites, P1 and P2, of miR-26a-5p promoter regions exhibited significant enrichment upon immunoprecipitation with the AR antibody, but no bands were evident for other two sites, P3 and P4 (Fig. 4B). We then sub-cloned the promoter region, including both P1 and P2 upstream of luciferase gene into a reporter plasmid. The dual-luciferase assay showed that the transcriptional activity was reduced when AR bound to the sites, and treatment of R1881 further decreased it (Fig. 4C and D), indicating that AR binds to the promoter of miR-26a-5p and inhibits its expression as predicted. Furthermore, we asked whether metformin and GSK126 affected the AR binding to the promoter of miR-26a-5p. As shown in Fig. 4E and F, either metformin or GSK126 alone reduced the AR binding to the promoter within limited extent, but the combination of metformin and GSK126 almost completely removed AR from the regions.

Figure 4.

MiR-26a-5p is directly regulated by AR. A, Scheme representing the binding sequences within the miR-26a promoter relative to the designed primers. B, ChIP analysis of AR binding to the miR-26a promoter region in 22Rv1 and LNCaP cells. C and D, HEK293T cells and PC3-Neo or PC3-AR cells were cotransfected with the miR-26a promoter construct with empty vector or AR for 24 hours, treated with R1881 (10 nmol/L) for additional 24 hours, and harvested for luciferase assays. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. E and F, 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours and harvested for anti-AR ChIP using qPCR to measure the binding of AR to the promoter of miR-26a. Values are means ± standard deviations; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. G, HEK293T cells were transfected with the miR-26a promoter construct in the presence of AR, EZH2-S21D, or EZH2-S21A, and harvested for luciferase assays. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. H and I, Chromatin was precipitated with anti-AR antibody and re-precipitated with anti-AR or anti-EZH2 antibody or IgG, followed by qPCR. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. NS, not significant.

Figure 4.

MiR-26a-5p is directly regulated by AR. A, Scheme representing the binding sequences within the miR-26a promoter relative to the designed primers. B, ChIP analysis of AR binding to the miR-26a promoter region in 22Rv1 and LNCaP cells. C and D, HEK293T cells and PC3-Neo or PC3-AR cells were cotransfected with the miR-26a promoter construct with empty vector or AR for 24 hours, treated with R1881 (10 nmol/L) for additional 24 hours, and harvested for luciferase assays. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. E and F, 22Rv1 cells were treated with DMSO, metformin (1 mmol/L), GSK126 (5 μmol/L), or both for 48 hours and harvested for anti-AR ChIP using qPCR to measure the binding of AR to the promoter of miR-26a. Values are means ± standard deviations; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. G, HEK293T cells were transfected with the miR-26a promoter construct in the presence of AR, EZH2-S21D, or EZH2-S21A, and harvested for luciferase assays. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. H and I, Chromatin was precipitated with anti-AR antibody and re-precipitated with anti-AR or anti-EZH2 antibody or IgG, followed by qPCR. Values are means ± SD; n = 3. *, P ≤ 0.05; **, P ≤ 0.01. NS, not significant.

Close modal

It is well known that metformin can reduce AR recruitment to the promoters of its target genes (27), but we asked how GSK126 could also reduce it. Recently, EZH2 was shown to interact with AR to regulate its binding to its target genes, in a manner dependent on EZH2's methyltransferase activity (28). In addition, the interaction between AR and EZH2 is mediated by phosphorylation at S21 of EZH2 (28). Therefore, we further hypothesized that AR cooperates with EZH2 to suppress miR-26a-5p expression. To test this, the miR-26a promoter construct was co-transfected with empty vector, AR, EZH2-S21D and EZH2-S21A either alone or in combination with AR plus EZH2-S21D and AR plus EZH2-S21A in HEK293T cells. We found that EZH2-S21D, instead of EZH2-S21A, promoted AR's inhibition on the transcriptional activity (Fig. 4G), suggesting that EZH2 interacts with AR to regulate the expression of miR-26a-5p. To further validate this point, re-ChIP assay was performed, and revealed that AR, bound to the miR-26a-5p promoter region, was significantly associated with EZH2 in 22Rv1 cells (Fig. 4H), but not in LNCaP cells (Fig. 4I). Altogether, these results suggest that expression of miR-26a-5p was suppressed by AR in prostate cancer cells, and that the AR/EZH2 complex reinforced the suppression in advanced androgen-refractory prostate cancer cells.

Metformin and GSK126 act synergistically in a 22Rv1-derived xenograft model

To further validate our in vitro finding, we evaluated the effect of metformin and GSK126 alone or in combination with a 22Rv1-derived xenograft mouse model. After a 24-day treatment, we found that metformin alone could barely affect the tumor growth, and GSK126 alone only exerted a limited inhibitory effect on it (Fig. 5AC). In contrast, the combination of metformin and GSK126 significantly blocked the tumor growth and decreased the tumor weight. Meanwhile, no significant body weight loss was observed among all four groups (Fig. 5D), implying that the combination of the two drugs with the indicated doses has little toxic side effect. To confirm the responses, we conducted histological analyses of these tumor samples. H&E staining showed that single-agent metformin, as well as GSK126, slightly reduced the tumor cell content (Fig. 6E); however, tumor cell content was markedly decreased after the combination therapy with metformin and GSK126 (Fig. 5E). Furthermore, the immunostaining of Ki67 and cleaved caspase-3 also showed that the combination of metformin and GSK126 led to a significant decrease in overall proliferation and a dramatic increase of apoptosis (Fig. 5FI). Finally, proteins were extracted from the harvested tumors and subjected to western blot against EZH2. We also found that co-therapy of metformin and GSK126 significantly lowered EZH2 expression than the mono-therapies (Fig. 5JL). These results are consistent with what we observed in the cell-based experiments, thus confirming the synergistic effect between metformin and GSK126.

Figure 5.

The combination of metformin and GSK126 reduced cell proliferation, increased apoptosis, and inhibited EZH2 expression in 22Rv1-derived xenograft tumors. A, Tumor growth curves of 22Rv1-derived mouse xenografts. After nude mice were inoculated with 22Rv1 cells (2.5 × 105/mouse) for 2 weeks, the mice bearing tumors were treated with drugs as described in Materials and Methods. The sizes of the tumors in each group were measured every 4 days (mean ± SD; n = 9 mice for each group). *, P ≤ 0.05; **, P ≤ 0.01. B, Images of the 22Rv1-derived xenograft tumors at the end of study. C, Measurement of tumor weight upon harvest. D, Measurement of mice body weight upon tumor harvest. E, Representative images of H&E staining on formaldehyde-fixed, paraffin-embedded, 22Rv1-derived tumor sections. F, Representative images of anti-Ki67 IHC staining of tumor sections. G, Quantification of Ki67 signals as percentages of Ki67-positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± standard deviations; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. H, Representative images of anti-cleaved caspase-3 IHC staining of tumor sections. I, Quantification of cleaved caspase-3 signals as percentages of cleaved caspase-3–positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. J and K, Protein lysates extracted from 22Rv1-derived tumors were subjected to Western blot for EZH2 and H3K23me3, as well as H3 and β-actin expression. L, Quantification of EZH2 protein levels in J and K.

Figure 5.

The combination of metformin and GSK126 reduced cell proliferation, increased apoptosis, and inhibited EZH2 expression in 22Rv1-derived xenograft tumors. A, Tumor growth curves of 22Rv1-derived mouse xenografts. After nude mice were inoculated with 22Rv1 cells (2.5 × 105/mouse) for 2 weeks, the mice bearing tumors were treated with drugs as described in Materials and Methods. The sizes of the tumors in each group were measured every 4 days (mean ± SD; n = 9 mice for each group). *, P ≤ 0.05; **, P ≤ 0.01. B, Images of the 22Rv1-derived xenograft tumors at the end of study. C, Measurement of tumor weight upon harvest. D, Measurement of mice body weight upon tumor harvest. E, Representative images of H&E staining on formaldehyde-fixed, paraffin-embedded, 22Rv1-derived tumor sections. F, Representative images of anti-Ki67 IHC staining of tumor sections. G, Quantification of Ki67 signals as percentages of Ki67-positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± standard deviations; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. H, Representative images of anti-cleaved caspase-3 IHC staining of tumor sections. I, Quantification of cleaved caspase-3 signals as percentages of cleaved caspase-3–positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. J and K, Protein lysates extracted from 22Rv1-derived tumors were subjected to Western blot for EZH2 and H3K23me3, as well as H3 and β-actin expression. L, Quantification of EZH2 protein levels in J and K.

Close modal
Figure 6.

The combination of metformin and GSK126 reduced cell proliferation, increased apoptosis, and inhibited EZH2 expression in LuCaP35CR xenograft tumors. A, Tumor growth curves of LuCaP35CR xenografts (mean ± SD; n = 4 mice for each group). *, P ≤ 0.05; **, P ≤ 0.01. B, Images of the LuCaP35CR xenograft tumors at the end of study. C, Measurement of tumor weight upon harvest. D, Blood was collected immediately when the mice were sacrificed, and a PSA enzyme–linked immunosorbent assay kit was used to measure the serum PSA levels. E, Representative images of H&E staining on formaldehyde-fixed, paraffin-embedded, LuCaP35CR tumor sections. F, Representative images of anti-Ki67 IHC staining of tumor sections. G, Quantification of Ki67 signals as percentages of Ki67-positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. H, Representative images of anti-cleaved caspase-3 IHC staining of tumor sections. I, Quantification of cleaved caspase-3 signals as percentages of cleaved caspase-3–positive cells compared with the total number of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. J and K, Protein lysates extracted from LuCaP35CR tumors were subjected to EZH2 Western blotting. L, Quantification of EZH2 protein levels in J and K. M, Proposed working model based on the results of this study.

Figure 6.

The combination of metformin and GSK126 reduced cell proliferation, increased apoptosis, and inhibited EZH2 expression in LuCaP35CR xenograft tumors. A, Tumor growth curves of LuCaP35CR xenografts (mean ± SD; n = 4 mice for each group). *, P ≤ 0.05; **, P ≤ 0.01. B, Images of the LuCaP35CR xenograft tumors at the end of study. C, Measurement of tumor weight upon harvest. D, Blood was collected immediately when the mice were sacrificed, and a PSA enzyme–linked immunosorbent assay kit was used to measure the serum PSA levels. E, Representative images of H&E staining on formaldehyde-fixed, paraffin-embedded, LuCaP35CR tumor sections. F, Representative images of anti-Ki67 IHC staining of tumor sections. G, Quantification of Ki67 signals as percentages of Ki67-positive cells compared with the total numbers of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. H, Representative images of anti-cleaved caspase-3 IHC staining of tumor sections. I, Quantification of cleaved caspase-3 signals as percentages of cleaved caspase-3–positive cells compared with the total number of cells. Multiple tumor sections were calculated (means ± SD; n = 4). *, P ≤ 0.05; **, P ≤ 0.01. J and K, Protein lysates extracted from LuCaP35CR tumors were subjected to EZH2 Western blotting. L, Quantification of EZH2 protein levels in J and K. M, Proposed working model based on the results of this study.

Close modal

Metformin and GSK126 act synergistically in a patient-derived xenograft model

To better mimic the growth and situation of CRPC, a patient-derived xenograft study was conducted with LuCaP35CR, which could recapitulate the major genomic and phenotypic features of the disease in humans. As predicted, the combination of GSK126 and metformin resulted in a stronger tumor-inhibitory effect than did monotherapies (Fig. 6AC). Of note, the serum PSA, which is often elevated with prostate cancer progression, displayed significant reduction upon the combination treatment (Fig. 6D). Furthermore, histological analyses were performed with the tumor samples. As shown in Fig. 6E, GSK126-treated tumors exhibited some apoptotic bodies and morphological changes, including cytoplasm reduction, nuclear pyknosis, and karyorrhexis. Remarkably, the tumors treated with both metformin and GSK126 showed increasing number of apoptotic bodies with pyknotic or fragmented nuclei, as well as condensed cytoplasm (Fig. 6E). Immunostaining of Ki67 and cleaved caspase-3 also revealed that the combination of metformin and GSK126 led to a significant decrease of overall proliferation and a dramatic increase of apoptosis (Fig. 6FI). Finally, we also analyzed EZH2 protein levels in all the tumor samples, and found that co-treatment of metformin and GSK126 led to a lower expression of EZH2 compared with either drug alone (Fig. 6JL). In summary, these results are consistent with that of 22Rv1-derived xenograft study, further confirming that metformin and GSK126 can act synergistically in CRPC.

We have demonstrated that the combination of metformin and GSK126 synergistically inhibited proliferation of LNCaP and 22Rv1, but not RWPE1 whose EZH2 is not affected by metformin (Supplementary Fig. S2B). Downregulation of EZH2 is one of the reasons contributing to the antiproliferative effect induced by the treatments. However, the cell viability could not be completely rescued by overexpression of exogenous EZH2. This could be because (i) transduction efficiency of Myc-EZH2 is not high enough; (ii) other molecules besides EZH2 may also be involved in the process. Compared with LNCaP, the combination is more synergistic in 22Rv1; meanwhile, 22Rv1 is much more resistant to metformin, suggesting that GSK126 strongly enhances the potency of metformin and re-sensitizes 22Rv1 to it. Previous studies of our laboratory have showed that different prostate cancer cells exhibit different sensitivities to metformin, and advanced AR-positive prostate cancer cells are usually resistant (29, 30), which is concordant with our finding. Similar to most malignancies, prostate tumors are usually composed of multiple cell types, with complexed characteristics and biological features, resulting in intratumoral heterogeneity; therefore, it seems that metformin alone is not a promising anti-cancer therapy for CRPC, and combinations of metformin with other drugs potentiating its function should be more viable.

It has been well documented that EZH2 is critical for prostate cancer growth, development, and progression (12). Recent studies revealed that EZH2 could act independently of its methyltransferase activity (16), indicating that additional approaches inhibiting EZH2 expression should be considered. In our study, we have demonstrated that metformin downregulated EZH2 through upregulating miR-26a-5p by using of microRNA inhibitors. Because EZH1 contains the same miR-26a–targeting sequence (UACUUGA) in its 3′-UTR region, it is likely that the miR-26a-5p inhibitor can stabilize H3K27me3 through regulating EZH1, consequently resulting in discordant change at the levels of EZH2 and H3K27me3. Furthermore, we have found that metformin's effect on EZH2 was actually antagonized by AR, which impeded metformin-induced re-expression of miR-26a-5p, and modification of AR changed metformin's effect on EZH2. Metformin could decrease EZH2 by eliminating AR's inhibitory effect on miR-26a-5p in LNCaP, but it could also reduce EZH2 in PC3 and DU145 as well, indicating that additional molecules besides AR are involved in metformin's regulation on EZH2. However, AR is still considered to be the determinant of the combination synergy, as we did not observe significant synergy using PC3 or DU145 (Supplementary Fig. S2C–S2F).

It was reported that AR inhibited gene expression by directly binding to its promoter (31). Moreover, AR was showed to regulate miRNAs' expression by directly binding to their promoters (32, 33). In our study, we observed AR bound to the promoter of miR-26a and suppressed its expression. Furthermore, the gene inhibition induced by AR could be reinforced by EZH2-S21D, which is consistent with previous studies demonstrating that EZH2 promoted AR recruitment to its sites via directly methylating it (28, 34). Also, we found that GSK126 decreased AR's recruitment at the promoter of miR-26a, further supporting the interaction between AR and EZH2 we observed.

We also tested the combination of the two drugs by in vivo experiments, and found that it could inhibit tumor growth, induce apoptosis, and downregulate EZH2 in both 22Rv1-derived tumors and LuCaP35CR xenografts. Meanwhile, the dosage we used did not cause any toxic effect. In the phase I study, GSK126 was escalated to maximum dose of 3,000 mg twice a week for patients with no dose limiting toxicity observed (14). However, the dosage used in our experiments is 50 mg/kg body weight/d, which was slightly higher and more frequent than that in the clinical trial. Till now, the safety limit of GSK126 is still undetermined, so our results are valuable to define the appropriate dosage range of medication of GSk126 for clinical use. The safe dosage of metformin for patients should be below 3,000 mg per day (35, 36), and the dosage used in our mice experiments is 30 mg/kg body weight/d, which is within the safe medication range. Although plasma concentrations of metformin in the vein may only reach tens of micromoles in the mice (37), it is enough to induce a significant lower cell survival rate (Supplementary Fig. S3A), as well as a stronger inhibition on EZH2 (Supplementary Fig. S3B), when combined with GSK126.

In summary, as shown in Fig. 6M, miR-26a-5p is negatively regulated by AR in LNCaP, which can be easily eliminated by metformin. Moreover, EZH2 reinforces AR's inhibition on miR-26a-5p expression in 22Rv1, which results in the resistance. However, inhibition of EZH2's methyltransferase activity with GSK126 can inhibit the interaction between AR and EZH2, restoring metformin's effect in 22Rv1. Therefore, our results suggest that the combination of metformin and GSK126 would be an effective approach targeting EZH2 for future prostate cancer therapy, in particular, for patients with AR-positive CRPC.

No potential conflicts of interest were disclosed.

Y. Kong: Conceptualization, data curation, methodology, writing-original draft. Y. Zhang: Investigation. F. Mao: Methodology. Z. Zhang: Methodology. Z. Li: Investigation, visualization. R. Wang: Methodology. J. Liu: Methodology. X. Liu: Conceptualization, supervision, funding acquisition, investigation, visualization, methodology, project administration, writing-review and editing.

This work was funded by NIH R01 CA157429, R01 CA192894, R01 CA196835, and R01 CA196634 (to X. Liu) and the Chinese Scholarship Council (to Y. Kong). This research is also supported by the University of Kentucky Markey Cancer Center (P30CA177558).

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.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2019
.
CA Cancer J Clin
2019
;
69
:
7
34
.
2.
Ryan
CJ
,
Smith
MR
,
Fizazi
K
,
Saad
F
,
Mulders
PF
,
Sternberg
CN
, et al
Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study
.
Lancet Oncol
2015
;
16
:
152
60
.
3.
Beer
TM
,
Armstrong
AJ
,
Rathkopf
DE
,
Loriot
Y
,
Sternberg
CN
,
Higano
CS
, et al
Enzalutamide in metastatic prostate cancer before chemotherapy
.
N Engl J Med
2014
;
371
:
424
33
.
4.
Kantoff
PW
,
Higano
CS
,
Shore
ND
,
Berger
ER
,
Small
EJ
,
Penson
DF
, et al
Sipuleucel-T immunotherapy for castration-resistant prostate cancer
.
N Engl J Med
2010
;
363
:
411
22
.
5.
Tannock
IF
,
de Wit
R
,
Berry
WR
,
Horti
J
,
Pluzanska
A
,
Chi
KN
, et al
Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer
.
N Engl J Med
2004
;
351
:
1502
12
.
6.
Kasznicki
J
,
Sliwinska
A
,
Drzewoski
J
. 
Metformin in cancer prevention and therapy
.
Ann Transl Med
2014
;
2
:
57
.
7.
Spratt
DE
,
Zhang
C
,
Zumsteg
ZS
,
Pei
X
,
Zhang
Z
,
Zelefsky
MJ
. 
Metformin and prostate cancer: reduced development of castration-resistant disease and prostate cancer mortality
.
Eur Urol
2013
;
63
:
709
16
.
8.
Murtola
TJ
,
Tammela
TL
,
Lahtela
J
,
Auvinen
A
. 
Antidiabetic medication and prostate cancer risk: a population-based case-control study
.
Am J Epidemiol
2008
;
168
:
925
31
.
9.
Rothermundt
C
,
Hayoz
S
,
Templeton
AJ
,
Winterhalder
R
,
Strebel
RT
,
Bartschi
D
, et al
Metformin in chemotherapy-naive castration-resistant prostate cancer: a multicenter phase 2 trial (SAKK 08/09)
.
Eur Urol
2014
;
66
:
468
74
.
10.
Li
K
,
Si-Tu
J
,
Qiu
J
,
Lu
L
,
Mao
Y
,
Zeng
H
, et al
Statin and metformin therapy in prostate cancer patients with hyperlipidemia who underwent radiotherapy: a population-based cohort study
.
Cancer Manag Res
2019
;
11
:
1189
97
.
11.
Simon
JA
,
Kingston
RE
. 
Mechanisms of polycomb gene silencing: knowns and unknowns
.
Nat Rev Mol Cell Biol
2009
;
10
:
697
708
.
12.
Shen
L
,
Cui
J
,
Liang
S
,
Pang
Y
,
Liu
P
. 
Update of research on the role of EZH2 in cancer progression
.
Onco Targets Ther
2013
;
6
:
321
4
.
13.
McCabe
MT
,
Ott
HM
,
Ganji
G
,
Korenchuk
S
,
Thompson
C
,
Van Aller
GS
, et al
EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
.
Nature
2012
;
492
:
108
12
.
14.
Yap
TA
,
Winter
JN
,
Leonard
JP
,
Ribrag
V
,
Constantinidou
A
,
Giulino-Roth
L
, et al
A phase I study of GSK2816126, an enhancer of zeste homolog 2(EZH2) inhibitor, in patients (pts) with relapsed/refractory diffuse large B-cell lymphoma (DLBCL), other non-Hodgkin lymphomas (NHL), transformed follicular lymphoma (tFL), solid tumors and multiple myeloma (MM)
.
Blood
2016
;
128
:
4203
.
15.
Gonzalez
ME
,
DuPrie
ML
,
Krueger
H
,
Merajver
SD
,
Ventura
AC
,
Toy
KA
, et al
Histone methyltransferase EZH2 induces Akt-dependent genomic instability and BRCA1 inhibition in breast cancer
.
Cancer Res
2011
;
71
:
2360
70
.
16.
Kim
J
,
Lee
Y
,
Lu
X
,
Song
B
,
Fong
KW
,
Cao
Q
, et al
Polycomb- and methylation-independent roles of ezh2 as a transcription activator
.
Cell Rep
2018
;
25
:
2808
20
.
17.
Chou
TC
,
Talalay
P
. 
Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors
.
Adv Enzyme Regul
1984
;
22
:
27
55
.
18.
Messeguer
X
,
Escudero
R
,
Farre
D
,
Nunez
O
,
Martinez
J
,
Alba
MM
. 
PROMO: detection of known transcription regulatory elements using species-tailored searches
.
Bioinformatics
2002
;
18
:
333
4
.
19.
Farre
D
,
Roset
R
,
Huerta
M
,
Adsuara
JE
,
Rosello
L
,
Alba
MM
, et al
Identification of patterns in biological sequences at the ALGGEN server: PROMO and MALGEN
.
Nucleic Acids Res
2003
;
31
:
3651
3
.
20.
Serresi
M
,
Siteur
B
,
Hulsman
D
,
Company
C
,
Schmitt
MJ
,
Lieftink
C
, et al
Ezh2 inhibition in Kras-driven lung cancer amplifies inflammation and associated vulnerabilities
.
J Exp Med
2018
;
215
:
3115
35
.
21.
Dudakovic
A
,
Camilleri
ET
,
Riester
SM
,
Paradise
CR
,
Gluscevic
M
,
O'Toole
TM
, et al
Enhancer of zeste homolog 2 inhibition stimulates bone formation and mitigates bone loss caused by ovariectomy in skeletally mature mice
.
J Biol Chem
2016
;
291
:
24594
606
.
22.
Ren
G
,
Baritaki
S
,
Marathe
H
,
Feng
J
,
Park
S
,
Beach
S
, et al
Polycomb protein EZH2 regulates tumor invasion via the transcriptional repression of the metastasis suppressor RKIP in breast and prostate cancer
.
Cancer Res
2012
;
72
:
3091
104
.
23.
Yang
YA
,
Yu
J
. 
EZH2, an epigenetic driver of prostate cancer
.
Protein Cell
2013
;
4
:
331
41
.
24.
Li
W
,
Yuan
Y
,
Huang
L
,
Qiao
M
,
Zhang
Y
. 
Metformin alters the expression profiles of microRNAs in human pancreatic cancer cells
.
Diabetes Res Clin Pract
2012
;
96
:
187
95
.
25.
Bao
B
,
Wang
Z
,
Ali
S
,
Ahmad
A
,
Azmi
AS
,
Sarkar
SH
, et al
Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells
.
Cancer Prev Res
2012
;
5
:
355
64
.
26.
Yamaguchi
H
,
Hung
MC
. 
Regulation and role of EZH2 in cancer
.
Cancer Res Treat
2014
;
46
:
209
22
.
27.
Wang
Y
,
Liu
G
,
Tong
D
,
Parmar
H
,
Hasenmayer
D
,
Yuan
W
, et al
Metformin represses androgen-dependent and androgen-independent prostate cancers by targeting androgen receptor
.
Prostate
2015
;
75
:
1187
96
.
28.
Xu
K
,
Wu
ZJ
,
Groner
AC
,
He
HH
,
Cai
C
,
Lis
RT
, et al
EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent
.
Science
2012
;
338
:
1465
9
.
29.
Shao
C
,
Ahmad
N
,
Hodges
K
,
Kuang
S
,
Ratliff
T
,
Liu
X
. 
Inhibition of polo-like kinase 1 (Plk1) enhances the antineoplastic activity of metformin in prostate cancer
.
J Biol Chem
2015
;
290
:
2024
33
.
30.
Chen
L
,
Ahmad
N
,
Liu
X
. 
Combining p53 stabilizers with metformin induces synergistic apoptosis through regulation of energy metabolism in castration-resistant prostate cancer
.
Cell Cycle
2016
;
15
:
840
9
.
31.
Bishop
JL
,
Thaper
D
,
Vahid
S
,
Davies
A
,
Ketola
K
,
Kuruma
H
, et al
The master neural transcription factor BRN2 is an androgen receptor-suppressed driver of neuroendocrine differentiation in prostate cancer
.
Cancer Discov
2017
;
7
:
54
71
.
32.
Pasqualini
L
,
Bu
H
,
Puhr
M
,
Narisu
N
,
Rainer
J
,
Schlick
B
, et al
miR-22 and miR-29a are members of the androgen receptor cistrome modulating LAMC1 and Mcl-1 in prostate cancer
.
Mol Endocrinol
2015
;
29
:
1037
54
.
33.
Takayama
KI
,
Misawa
A
,
Inoue
S
. 
Significance of microRNAs in androgen signaling and prostate cancer progression
.
Cancers
2017
;
9
:
102
.
34.
Kim
E
,
Kim
M
,
Woo
DH
,
Shin
Y
,
Shin
J
,
Chang
N
, et al
Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells
.
Cancer Cell
2013
;
23
:
839
52
.
35.
Graham
GG
,
Punt
J
,
Arora
M
,
Day
RO
,
Doogue
MP
,
Duong
JK
, et al
Clinical pharmacokinetics of metformin
.
Clin Pharmacokinet
2011
;
50
:
81
98
.
36.
Vecchio
S
,
Giampreti
A
,
Petrolini
VM
,
Lonati
D
,
Protti
A
,
Papa
P
, et al
Metformin accumulation: lactic acidosis and high plasmatic metformin levels in a retrospective case series of 66 patients on chronic therapy
.
Clin Toxicol
2014
;
52
:
129
35
.
37.
He
L
,
Wondisford
FE
. 
Metformin action: concentrations matter
.
Cell Metab
2015
;
21
:
159
62
.