Metformin and prostate cancer stem cells: a novel therapeutic target REVIEW Metformin and prostate cancer stem cells: a novel therapeutic target MJ Mayer1,2, LH Klotz1,2 and V Venkateswaran1,2 Prostate cancer is the second most frequently diagnosed cancer in the world. Localized disease can be effectively treated with radiation therapy or radical prostatectomy. However, advanced prostate cancer is more difficult to treat and if metastatic, is incurable. There is a need for more effective therapy for advanced prostate cancer. One potential target is the cancer stem cell (CSC). CSCs have been described in several solid tumors, including prostate cancer, and contribute to therapeutic resistance and tumor recurrence. Metformin, a common oral biguanide used to treat type 2 diabetes, has been demonstrated to have anti- neoplastic effects. Specifically, metformin targets CSCs in breast cancer, pancreatic cancer, glioblastoma and colon cancer. Metformin acts directly on the mitochondria to inhibit oxidative phosphorylation and reduce mitochondrial ATP production. This forces tumor cells to compensate by increasing the rate of glycolysis. CSCs rely heavily on mitochondrial oxidative phosphorylation for energy production. The glycolytic switch results in an energy crisis in these cells. Metformin could be used to exploit this metabolic weakness in CSCs. This would increase CSC sensitivity to conventional cancer therapies, circumventing treatment resistance and enhancing treatment efficacy. This review will explore the characteristics of prostate CSCs, their role in tumor propagation and therapeutic resistance and the role of metformin as a potential prostate CSC sensitizer to current anticancer therapies. Prostate Cancer and Prostatic Diseases (2015) 18, 303–309; doi:10.1038/pcan.2015.35; published online 28 July 2015 INTRODUCTION Prostate cancer (PCa) has a long natural history. When prostate cancer is localized and is classified as a low Gleason grade tumor, it can be monitored with active surveillance. Higher-grade cancers are effectively treated by surgical resection of the prostate (radical prostatectomy) or radiation therapy.1,2 However, if the cancer invades through the capsule into surrounding tissue, or recurs after local therapy, it is much more difficult to treat. If the disease metastasizes, it is generally incurable.3,4 Advanced-stage PCa is usually treated with androgen-deprivation therapy (ADT). Most patients with metastatic disease managed with ADT eventually relapse with castration-resistant prostate cancer (CRPC) and die of the disease.3,4 CRPC can be treated with docetaxel, abiraterone plus prednisone, enzalutamide, and cabazitaxel, which provide significant survival benefits, but are not curative.5 Thus, there is still a need to improve the therapeutic options available for advanced-stage prostate cancer patients. Prostate cancer is a multifocal disease. Prostates often contain multiple independent and genetically distinct foci.6 One model that explains this heterogeneity is the ‘cancer stem cell’ model. This model postulates that only a small subset of cancer cells within a tumor have the ability to sustain tumorigenesis, resulting in a hierarchical organization of primary tumors.7,8 This subset of tumor-propagating cells are defined as cancer stem cells (CSCs), as they share a number of characteristics with normal stem cells, one such example being self-renewal.7–12 CSCs were first identified and characterized in hematological malignancies by Bonnet and Dick.13 Since then, CSCs have been identified in several solid tumors including breast, pancreas, colon, lung, brain and prostate.14–19 The CSC model is intriguing because it provides an explanation for tumor recurrence and resistance to conven- tional cancer therapies.8 The discovery of new agents that can sensitize CSCs to conventional cancer treatments holds therapeu- tic promise. Metformin, a common well-tolerated oral biguanide prescribed for type 2 diabetes, has been shown to selectively target cancer stem cells in several types of solid tumors and improve the efficacy of radiation and chemotherapy in breast cancer and colon cancer.20–23 Targeting therapy-resistant CSCs in prostate cancer provides a unique opportunity for novel therapeutic interventions. Metformin could be used to sensitize prostate CSCs to current conven- tional anticancer therapies and improve efficacy of treatment. This review evaluates the current evidence regarding the role of CSCs in prostate cancer and focuses on the following topics: (1) identification and characterization of prostate CSCs; (2) role that CSCs play in tumor propagation and therapeutic resistance; (3) role of metformin as a prostate CSC sensitizer for conventional therapy. PROSTATE STRUCTURE AND DEVELOPMENT The prostate is a glandular organ comprised of three distinct epithelial cell types. Basal cells are found along the basement membrane of each prostatic duct and express CK5, CK14, CD44, 1Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada and 2Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada. Correspondence: Dr V Venkateswaran, Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, S-118B, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5. E-mail: vasundara.venkateswaran@sunnybrook.ca Received 13 April 2015; revised 7 June 2015; accepted 13 June 2015; published online 28 July 2015 Prostate Cancer and Prostatic Diseases (2015) 18, 303–309 © 2015 Macmillan Publishers Limited All rights reserved 1365-7852/15 www.nature.com/pcan http://dx.doi.org/10.1038/pcan.2015.35 mailto:vasundara.venkateswaran@sunnybrook.ca http://www.nature.com/pcan CD133, p63, Bcl-2 and low or undetectable levels of androgen receptor (AR).1,6,7,24 Luminal cells form a layer above the basal cells and are the predominant cell type of the prostate.1,24 Luminal cells express high levels of AR, CK8, CK18, CK19, CD57 and produce proteins such as PSA and prostatic acid phosphatase (PAP) that are secreted into the luminal space.1,6,7,24 Neuroendocrine cells are a rare population, which is dispersed among the basal layer, are androgen-independent, express chromogranin A and secrete neuroendocrine peptides.1,6,24 The primary survival of basal cells following androgen depriva- tion led to the hypothesis that prostate stem cells reside within the basal layer.7,24 Collins et al.25 identified a small population of basal prostate epithelial stem cells, which expressed high levels of α2β1. This was further corroborated by the identification of a small (1%) population of human prostate basal stem cells, which expressed CD133 and were restricted to the α2β1 population. 26 Leong et al.27 demonstrated that a single stem cell with the Lin-/ Sca-1+/CD133+/CD44+/CD117+ phenotype was able to regener- ate a prostate after transplantation in vivo. Lineage tracing has shown that multipotent basal stem cells give rise to basal, luminal and neuroendocrine cells, as well as unipotent luminal progeni- tors, which cause the epithelial expansion observed during postnatal prostate development.28 Wang et al.29 also demon- strated that basal cells and luminal cells undergo different types of division. Basal cells can divide symmetrically to produce two daughter basal cells or asymmetrically to produce one basal cell and one luminal cell.29 However, luminal cells can only undergo symmetric divisions to produce two luminal cells and cannot produce any basal cells.29 These results lend support to the theory that there is a hierarchy of epithelial lineages containing both multipotent and unipotent stem cells in the developing prostate.29 ROLE OF CSCs IN PROSTATE CANCER CSCs are defined as a subset of tumor cells that have the capacity to self-renew and generate the heterogeneous cell lineages that comprise a tumor.10,30 The term ‘cancer stem cell’ was established for this unique subpopulation of tumor cells because they possess properties similar to those of normal tissue stem cells, such as self-renewal by symmetric or asymmetric division, high proliferative potential and the capacity to differentiate into multiple cell lineages.10,12,30,31 The CSC model postulates that CSCs give rise to a hierarchically organized, heterogeneous tumor, with the CSCs at the top of the hierarchy.30 The CSCs then proliferate and generate PCa progenitor cells, which despite only having limited self-renewal abilities, can differentiate into many different mature cells types, such as CD57+AR+PSA+ luminal secretory cells.12 The CSCs themselves are typically quiescent, whereas the majority of the proliferating population of tumor progenitor cells are generated by the CSCs.12 CSCs are also thought to have an important role in cancer progression. Colombel et al.32 demonstrated that primary prostate carcinomas contain a subset of CSCs, which have a role in local invasion, specifically seminal vesicle invasion and bone metastasis. The percentage of CSCs had a prognostic impact, particularly on the risk of progression of bone metastases.32 The percentage of cells expressing a stem cell phenotype in bone marrow metastases of prostate cancer patients has been shown to be predictive of bone metastases progression.33 There is also a small population of docetaxel-resistant cells, likely CSCs, that have been shown to be higher in metastasic compared to primary patient samples, and, in primary untreated samples, the percentage of these cells was associated with prognostic factors and time to biochemical relapse.34 The role of prostate CSCs in recurrence and development of CRPC is likely due to their intrinsic therapeutic resistance. Conventional anticancer therapies such as radiation, androgen deprivation and chemotherapy target the rapidly proliferating bulk of tumor cells, but they may not affect the small quiescent, androgen-independent CSC population. This reservoir of CSCs could then in the future, after therapy has been halted, begin proliferating again and give rise to another tumor, which would have survived selective pressure from therapy. The selection theory of androgen resistance postulates that there is a set of pre-existing ADT-resistant cancer cells in tumors.35 Androgen depletion, therefore, provides selective pressure, killing the androgen-dependent cancer cells while androgen-independent tumor cells survive and can then give rise to castration-resistant tumors.35 Prostate CSCs are candidates for these pre-existing ADT-resistant cells that could give rise to CRPC since they have self-renewal and tumor-propagating capabilities, as well as a lack of or very low AR expression.35 Recurrent PCa tissue samples are enriched in CSCs, which are likely responsible for chemoresistance.36 An important distinction should be made between CSCs and the cancer cell of origin. The cell of origin refers to the initial cell that is the target of genetic alteration(s) that result in malignant transformation.7,30 CSCs, in contrast, are tumor-propagating cells that sustain malignant growth.37 CSCs may arise from a number of different types of cells including normal stem cells, restricted progenitors or differentiated cells.30,38,39 IDENTIFICATION AND CHARACTERIZATION OF PROSTATE CSCs CSCs can only be identified and characterized experimentally.40 The gold standard for defining CSCs functionally is serial transplantation in immunodeficient mice. This approach confirms the properties of self-renewal and tumor propagation.4,10 Many CSC studies have used experimental methods that were established for enriching normal stem cells including transplanta- tion of flow cytometry-sorted CSCs, side-population and ALDE- FLUOR assays, and lineage-tracing studies.7 A number of different populations of prostate CSCs have been identified.41 A population of CD44+/α2β1 high/CD133+ cancer stem cells from human prostate tumor samples has been identified in human prostate tumor tissue.19,42 The CD44+/α2β1 high population was also shown to be enriched in CSCs in LAPC-9 cells.43 Populations of CD44+ prostate cancer stem cells have been shown to be more proliferative, tumorigenic and metastatic than CD44 − prostate cancer cells, which are all typical characteristics of CSCs.44,45 Aldehyde dehydrogenase (ALDH) has also been shown to be a marker for prostate CSCs. ALDH+ prostate cancer cells have been shown to exhibit several CSC characteristics and is considered a marker for prostate CSCs.46,47 ALDH+ prostate cancer cells have been shown to possess enhanced clonogenicity, migration, tumorigenicity and readily form metastases in vivo.47 Qin et al.48 also identified highly tumorigenic castration-resistant PSA− /lo CSCs, which can be further enriched to include a population of ALDH+/CD44+/α2β1+ cells. ATP-binding cassette (ABC) transporter ABCG2+ prostate CSCs have also been isolated using the side-population method, which allows for the isolation of cells that can efflux Hoechst 33342 dye more efficiently because of elevated ABC transporter expression.49,50 Stemness markers have also been used to characterize CSC populations in the prostate. Overexpression of Nanog in LNCaP and DU145 human prostate cancer cell lines promoted CSC properties and enhanced the expression of CD133, CD44, ALDH1A1 and ABCG2.51 Prostate CSCs have also been shown to express Oct4 and Sox2 along with Nanog.52,53 Guzel et al.36 also showed an increased expression of Sox2, Oct4, Nanog, and ABCG2 in recurrent prostate cancer tissue samples, indicating an enrichment in prostate CSCs. A number of other cell surface markers identify prostate CSCs. These are included in Table 1. Metformin and prostate cancer stem cells MJ Mayer et al 304 Prostate Cancer and Prostatic Diseases (2015), 303 – 309 © 2015 Macmillan Publishers Limited PROSTATE CSCs AND THERAPEUTIC RESISTANCE Prostate CSCs are resistant to most conventional cancer therapies. Although there is some evidence that CSCs are radioresistant in breast cancer and glioblastoma cell lines, this has not yet been demonstrated in prostate cancer.54–56 Prostate CSCs are thought to contribute to the development of CRPC. Seiler et al.57 demonstrated that ADT resulted in castration resistance and overexpression of stemness markers such as Sox2 and Oct4, indicating a role for prostate CSCs in ADT resistance. In vivo studies have demonstrated enhanced expression of stem cell markers in resistant tumors in castrated mice.58 The peak expression of these markers also occurred soon after ADT, suggesting that prostate CSCs may have a role as an adaptive survival mechanism.58 CSCs may confer resistance to chemotherapy.59 One contribut- ing factor to this chemoresistance is the expression of ABC transporters. ABC transporters are membrane bound and can pump various small molecules, such as cytotoxic drugs and dyes, out of cells at the expense of ATP hydrolysis.60 CSCs express high levels of ABC transporters. These pump chemotherapeutic drugs out of the cytoplasm, resulting in reduced intracellular drug concentrations.60,61 Zhang et al.62 demonstrated that tumor- spheres from PCa cell lines expressing high levels of CD44 and ABCG2 were chemoresistant, likely due to the elevated expression of ABCG2. A subset of cells with high tumor-initiating capacity, likely CSCs, have been identified in both DU145 and 22RV1 human prostate cancer cells, as well as in human PCa samples. These cells contribute to docetaxel resistance and tumor re-initiation.34 METFORMIN AND PROSTATE CANCER Metformin, a commonly prescribed and well-tolerated oral biguanide used to treat type 2 diabetes, has been shown to exert anti-neoplastic effects in several types of cancer. Metformin reduces hepatic gluconeogenesis, increases glucose uptake in peripheral tissue such as skeletal muscle and increases insulin sensitivity.63,64 Evans et al.65 showed a reduced cancer burden in diabetic patients treated with metformin compared with those treated with other diabetic therapies.63 Since that initial paper, metformin has been shown to have anti-neoplastic properties in breast cancer,66,67 ovarian cancer,68 pancreatic cancer69 and prostate cancer.70 In prostate cancer, metformin inhibits the proliferation of LNCaP, DU145 and PC3 human prostate cancer cell lines and also reduced tumor growth in LNCaP xenografts.70 In prostate cancer patients, metformin use is associated with a decreased risk of PCa diagnosis while other oral diabetes medications are not.71 Increased cumulative metformin exposure after PCa diagnosis has been associated with decreased all-cause and PCa-specific mortality in diabetic patients.72 A meta-analysis by Yu et al.73 showed that metformin was associated with a significant reduction in cancer risk and biochemical recurrence. METFORMIN TARGETS CSCs In breast cancer, metformin effectively targets breast CSCs and enhances the effectiveness of some therapies. Song et al.20 demonstrated that metformin was cytotoxic to radioresistant breast CSCs and increased the efficacy of radiation in suppressing tumor growth in vivo, likely by reducing the population of CSCs. Hirsch et al.21 also showed that metformin targeted breast CSCs in four cell lines, enhanced the tumor-suppressing effect of doxorubicin and prolonged remission in vivo. Furthermore, metformin combined with a fourfold lower dose of doxorubicin was shown to be as effective as the standard dose of doxorubicin treatment alone in vivo.74 Metformin has also been shown to enhance the effect of paclitaxel and carboplatin by targeting CSCs in breast cancer xenografts at doses comparable to the dose per kg used in type 2 diabetes patients.74 In pancreatic cancer, metformin has been shown to significantly decrease cell survival, clonogenicity, wound-healing and sphere-forming capacity in pancreatic CSCs and also decreased the expression of CSC markers such as CD44, CD133, ALDH1, Nanog and Oct4.22,75–78 In glio- blastoma cell lines, metformin effectively reduced the proliferation of CD133+ CSCs in an Akt-dependent manner.79 The CD133+ population of colon cancer CSCs have been shown to be signi- ficantly reduced by treatment with metformin and it enhanced the antiproliferative effect of 5-fluorouracil on colon CSCs.22 In addition, metformin acts synergistically with 5-fluorouracil and oxaliplatin to inhibit cell proliferation and tumor growth of chemoresistant colorectal cancer cells by reducing the viability of colorectal CSCs.23 MECHANISM OF ACTION OF METFORMIN IN CSCs Biguanides, such as metformin, exert their effects by decreasing oxidative phosphorylation which induces energetic stress.80 This leads to a number of secondary cell lineage-specific effects.81 Metformin action is mediated through direct effects on the Table 1. Markers for isolation of prostate CSCs Marker Expression level Characteristics Reference ALDH High Enzyme oxidizes aldehydes Li et al.,46 Qin et al.,48 Finones et al.92 ABCG2 High ATPase transporter Foster et al.,49 Gangavaparu et al.50 CD44 + Cell adhesion and signaling Patrawala et al.,44 Patrawala et al.,43 Collins et al.,19 Hurt et al.45 CD133 (Prominin-1) + Marker normal stem cells and CSCs Richardson et al.,26 Collins et al.19 c-Kit (CD117) + Receptor tyrosine kinase Finones et al.92 Integrin α2β1 (CD49b) High Collagen receptor Collins et al., 19 Patrawala et al.,44 Guzman-Ramirez et al.42 CD49f High Laminin binding Guzman-Ramirez et al.42 CD166 + Cell adhesion Jiao et al.93 PSA − /lo Glycoprotein Qin et al.48 CK5/14 + Cytokeratin Tokar et al.94 CK8/18 + Cytokeratin Tokar et al.94 Nestin + Intermediate filament protein Guzman-Ramirez et al.42 SCA-1 + Cell surface marker Lawson et al.,95 Xin et al.96 SMO (Smoothened) + G-protein-coupled receptor Patrawala et al.44 Sox2 + Transcription factor (self-renewal) Rybak and Tang53 Oct4 + Transcription factor (self-renewal) Patrawala et al.44 Nanog + Transcription factor (self-renewal) Jeter et al.51 Abbreviation: ALDH, aldehyde dehydrogenase; CSC, cancer stem cell. Metformin and prostate cancer stem cells MJ Mayer et al 305 © 2015 Macmillan Publishers Limited Prostate Cancer and Prostatic Diseases (2015), 303 – 309 mitochondria, either by inhibiting mitochondrial complex 1 or the redox shuttle enzyme mitochondrial glycerophosphate dehydro- genase.63,81–84 This causes inhibition of oxidative phosphorylation and reduced ATP production and oxygen consumption.63 The decline in mitochondrial ATP production triggers activation of the cellular energy regulator AMP-activated protein kinase (AMPK) thereby directing cells towards an antiproliferative ‘energy-saving’ phenotype characterized by the downregulation of ATP-consuming processes such as protein and fatty acid synthesis.63,80 The antiproliferative effect of this metabolic reprogramming by AMPK was thought to contribute to the anti- neoplastic effect of metformin. However, the activation of AMPK can enhance cell survival under the conditions of energetic stress and could in fact have a pro-survival effect.85 The effect of metformin in PCa cells has been shown to be independent of the AMPK pathway.86 Tumors with loss of LKB1, an upstream kinase required for AMPK activation, are hypersensitive to biguanides, which indicates that cancer cells deficient in AMPK will be less likely to reduce energy consumption due to biguanide-induced reduction in ATP production and would increase the likelihood of these cells experiencing an energy crisis.85 More research is still needed to elucidate the antiproliferative and pro-survival effect of AMPK in cancer cells and the specific downstream effects of metformin-mediated inhibition of mitochondrial oxidative phosphorylation. This mechanism of action of metformin has been evaluated in cancer. In human breast cancer cells, metformin directly inhibits mitochondrial complex 1 and citric acid cycle function, which reduces mitochondrial respiration.81 Reduced mitochondrial oxidative phosphorylation results in impaired mitochondrial ATP production and forces cancer cells to compensate by increasing aerobic glycolysis.81 Therefore, cells which rely more heavily on mitochondrial oxidative phosphorylation are more sensitive to metformin treatment and CSCs are included in this category.81 Pancreatic cancer stem cells have been reported to have a highly mitochondrial-dependent metabolic profile.78 This is quite differ- ent from the majority of cancer cells that proliferate uncontrollably and limit their metabolism primarily to glycolysis even in the presence of oxygen (i.e., aerobic glycolysis), known as the Warburg effect.87 CSCs do not rapidly divide and therefore do not undergo this metabolic shift. If CSCs continue to rely on mitochondrial ATP production, they would be more sensitive to metformin treatment and would not be able to compensate effectively by switching to aerobic glycolysis. There are two possible scenarios related to the role of AMPK as the expression of AMPK in CSCs has not been fully elucidated. If CSCs do have functional AMPK, the activation of AMPK would likely prevent CSCs from shifting to aerobic glycolysis, which would cause an energy crisis and render CSCs incapable of compensation.88 If the CSCs are deficient in AMPK, they would have an innate inability to compensate for an energy crisis and AMPK-deficient CSCs would, therefore, be more sensitive to metformin treatment.88 Both scenarios would ultimately result in making CSCs more sensitive to additional stressors after metformin treatment, suggesting that metformin could be used as a CSC sensitizer for various types of cancer therapy (Figure 1). The pharmacokinetics of metformin also play an important role in its mechanism of action as different tissues have varying levels of exposure to biguanides, such as metformin.80 This is due to the fact that metformin requires membrane transport proteins such as organic cation transporter 1 (OCT1) to enter cells.80 The varying expression level of transport proteins between cell types can affect the bioavailability of metformin. For example, hepatocytes express high levels of OCT1, which allows for enhanced cellular uptake of metformin and the accumulation of relatively high metformin concentrations in the liver.85 Once metformin has entered a cell, the mitochondrial membrane potential causes an increased uptake of metformin into the organelle and results in the mitochondria being the site of the highest concentration of the drug within a cell.85 The accumulation of metformin in the mitochondria may contribute to its ability to effectively inhibit oxidative phosphorylation and cause an energy crisis in CSCs. It is also possible that if CSCs have a higher OCT1 expression level, the effect of metformin on these cells would be enhanced. However, the OCT1 expression levels of CSCs have not been elucidated. ROLE OF METFORMIN IN PROSTATE CANCER THERAPY A few studies suggest that metformin may enhance the effectiveness of prostate cancer therapies. Metformin has been shown to enhance the effectiveness of ADT. Research conducted in our laboratory has shown that metformin combined with bicalutamide significantly reduces prostate cancer cell growth more effectively than either metformin or bicalutamide alone.89 In addition, the combined treatment affected AR-positive LNCaP cells by altering cell proliferation, whereas combined treatment in Figure 1. Mechanism of action of metformin in prostate cancer stem cells. ADT, androgen-deprivation therapy. Oct, organic cation transporter. Metformin and prostate cancer stem cells MJ Mayer et al 306 Prostate Cancer and Prostatic Diseases (2015), 303 – 309 © 2015 Macmillan Publishers Limited AR-negative PC3 cells promoted apoptosis.89 Nguyen et al.90 also showed that combined treatment of enzalutamide and metformin significantly reduced tumor size in enzalutamide-resistant LNCaP C4-2B xenografts, and combined treatment was more effective than enzalutamide or metformin alone. Iliopoulos et al.74 demonstrated that metformin combined with doxorubicin sup- pressed tumor growth in PC3 xenografts. Radiosensitization by targeting prostate CSCs is another potential benefit. Reducing oxygen consumption may have an impact on prostate CSCs as reducing oxygen supply would impair mitochondrial oxidative phosphorylation. This would result in CSCs compensating by using glycolysis, which would cause a cellular energy crisis that would make CSCs more sensitive to additional stressors such as radiation. In one study, prostate cancer patients taking metformin during radiotherapy had a significant reduction in early biochemical relapse.91 When considering future clinical trials, an important considera- tion is the potential benefit of conducting trials assessing the effect of metformin on prostate CSCs. Currently two trials are registered on clinicaltrials.gov assessing the effect of metformin combined with anticancer therapies for prostate cancer. TAXOMET is a Phase II trial recruiting patients to assess the effect of metformin combined with Taxotere (generic name of docetaxel) on PSA response rate, biochemical and clinical progression-free survival, overall survival and tolerance. The MetAb-Pro trial is a Phase II pilot study assessing the impact of combining metformin with abiraterone treatment on survival in patients with metastatic CRPC. This is indicative of the definite interest in the use of metformin as a sensitizing agent for anticancer therapies. However, there is no focus on prostate CSCs in either of the trials mentioned above. Currently, very few trials are registered that assess the effect of metformin on CSCs. A randomized clinical trial on colon cancer patients currently in the data analysis phase assessed the effect of metformin administered before surgery on the proportion of CSCs identified in tumors. There is also a Phase II trial currently recruiting ovarian, primary peritoneal and fallopian tube cancer patients to assess whether metformin combined with chemotherapy will prevent relapse by targeting CSCs. However, again there are no trials exploring the effect of metformin on prostate CSCs. If metformin does, in fact, have a CSC-specific effect when combined with other anticancer therapies, this would provide evidence for a novel therapeutic approach for prostate cancer that would warrant assessment in clinical trials. CONCLUSION Great strides have been made in establishing expression profiles for prostate CSC isolation and characterization. Therapeutic resistance continues to be a problem in the treatment of prostate cancer, and prostate CSCs are likely an important factor in treatment resistance, cancer recurrence and progression. The ability to selectively eliminate prostate CSCs holds therapeutic promise. Metformin has been shown to target CSCs in a number of different solid tumors although there is limited data in prostate cancer. Establishing the effect of metformin as a potential sensitizer for prostate CSCs and combining metformin with other anticancer therapies to enhance their effectiveness provides an opportunity to develop improved therapeutic options for prostate cancer patients. CONFLICT OF INTEREST The authors declare no conflict of interest. REFERENCES 1 Abate-Shen C, Shen MM. Molecular genetics of prostate cancer. Genes Dev 2000; 14: 2410–2434. 2 Maitland NJ, Collins AT. Prostate cancer stem cells: a new target for therapy. J Clin Oncol 2008; 26: 2862–2870. 3 Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer 2001; 1: 34–45. 4 Li H, Tang DG. Prostate cancer stem cells and their potential roles in metastasis. J Surg Oncol 2011; 103: 558–562. 5 Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T et al. EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol 2014; 65: 467–479. 6 Shen MM, Abate-Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev 2010; 24: 1967–2000. 7 Chen X, Rycaj K, Liu X, Tang DG. New insights into prostate cancer stem cells. Cell Cycle 2013; 12: 579–589. 8 Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving com- plexities. Cell Stem Cell 2012; 10: 717–728. 9 Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol 2013; Chapter 14; Unit 14.25. 10 Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 2006; 66: 9339–9344. 11 Ning X, Shu J, Du Y, Ben Q, Li Z. Therapeutic strategies targeting cancer stem cells. Cancer Biol Ther 2013; 14: 295–303. 12 Tang DG, Patrawala L, Calhoun T, Bhatia B, Choy G, Schneider-Broussard R et al. Prostate cancer stem/progenitor cells: identification, characterization and impli- cations. Mol Carcinog 2007; 46: 1–14. 13 Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3: 730–737. 14 Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 2003; 100: 3983–3988. 15 Li C, Lee CJ, Simeone DM. Identification of human pancreatic cancer stem cells. Methods Mol Biol 2009; 568: 161–173. 16 Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007; 445: 111–115. 17 Wang P, Gao Q, Zhenhe S, Munthe E, Solberg S, Ma L et al. Identification and characterization of cells with cancer stem cell properties in human primary lung cancer cell lines. PLoS One 2013; 8: e57020. 18 Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T et al. Identification of human brain tumour intiating cells. Nature 2004; 432: 396–401. 19 Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res 2005; 65: 10946–10951. 20 Song CW, Lee H, Dings RPM, Williams B, Powers J, Dos Santos T et al. Metformin kills and radiosensitizes cancer cells and preferentially kills cancer stem cells. Sci Rep 2012; 2: 1–9. 21 Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res 2009; 69: 7507–7511. 22 Zhang Y, Guan M, Zheng Z, Zhang Q, Gao F, Xue Y. Effects of metformin on CD133+ colorectal cancer cells in diabetic patients. PLoS One 2013; 8: e81264. 23 Nangia-Makker P, Yu Y, Vasudevan A, Farhana L, Rajendra SG, Lei E et al. Met- formin: a potential therapeutic agent for recurrent colon cancer. PLoS One 2014; 9: e84369. 24 Lawson DA, Witte ON. Stem cells in prostate cancer initiation and progression. J Clin Invest 2007; 117: 2044–2050. 25 Collins AT, Habib FK, Maitland NJ, Neal DE. Identification and isolation of human prostate epithelial stem cells based on alpha1beta1-integrin expression. J Cell Sci 2001; 114: 3865–3872. 26 Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci 2004; 117: 3539–3545. 27 Leong KG, Wang BE, Johnson L, Gao WQ. Generation of a prostate from a single adult stem cell. Nature 2008; 456: 804–810. 28 Ousset M, Van Keymeulen A, Bouvencourt G, Sharma N, Achouri Y, Simons BD et al. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat Cell Biol 2012; 14: 1131–1138. 29 Wang J, Zhu HH, Chu M, Liu Y, Zhang C, Liu G et al. Symmetrical and assymetrical division analysis provides evidence for a hierarchy of prostate epithelial cell lineages. Nat Commun 2014; 5: 4758. 30 Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evi- dence and unresolved questions. Nat Rev Cancer 2008; 8: 755–768. 31 Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell 2014; 14: 275–291. Metformin and prostate cancer stem cells MJ Mayer et al 307 © 2015 Macmillan Publishers Limited Prostate Cancer and Prostatic Diseases (2015), 303 – 309 32 Colombel M, Eaton CL, Hamdy F, Ricci E, van der Pluijm G, Cecchini M et al. Increased expression of putative cancer stem cell markers in primary prostate cancer is associated with progression of bone metastases. Prostate 2012; 72: 713–720. 33 Ricci E, Mattei E, Dumontet C, Eaton CL, Hamdy F, van der Pluijm G et al. Increased expression of putative cancer stem cell markers in the bone marrow of prostate cancer patients is associated with bone metastasis progression. Prostate 2013; 73: 1738–1746. 34 Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V, Castillo-Martin M, Quinn SA, Rodriguez-Barrueco R et al. Suppression of acquired docetaxel resistance in prostate cancer through depletion of notch- and hedgehog-dependent tumor- initiating cells. Cancer Cell 2012; 22: 373–388. 35 Zong Y, Goldstein AS. Adaptation or selection-mechanisms of castration-resistant prostate cancer. Nat Rev Urol 2013; 10: 90–98. 36 Guzel E, Karatas OF, Duz MB, Solak M, Ittmann M, Ozen M. Differential expression of stem cell markers and ABCG2 in recurrent prostate cancer. Prostate 2014; 74: 1498–1505. 37 Visvader JE. Cells of origin in cancer. Nature 2011; 469: 314–322. 38 Magee JA, Piskounova E, Morrison SJ. Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell 2012; 21: 283–296. 39 Collins AT, Maitland NJ. Prostate cancer stem cells. Eur J Cancer 2006; 42: 1213–1218. 40 Tang DG. Understanding cancer stem cell heterogeneity and plasticity. Cell Res 2012; 22: 457–472. 41 Klonisch T, Wiechec E, Hombach-Klonisch S, Ande SR, Wesselborg S, Schulze- Osthoff K et al. Cancer stem cell markers in common cancers-therapeutic impli- cations. Trends Mol Med 2008; 14: 450–460. 42 Guzman-Ramirez N, Voller M, Wetterwald A, Germann M, Cross NA, Rentsch CA et al. In vitro propagation and characterization of neoplastic stem/progenitor-like cells from human prostate cancer tissue. Prostate 2009; 69: 1683–1693. 43 Patrawala L, Calhoun-Davis T, Schneider-Broussard R, Tang DG. Hierarchical organization of prostate cancer cell sin xenograft tumors: the CD44+α2β1 + cell population is enriched in tumor initiating cells. Cancer Res 2007; 67: 6796–6805. 44 Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S et al. Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene 2006; 25: 1696–1708. 45 Hurt EM, Kawasaki BT, Klarmann GJ, Thomas SB, Farrar WL. CD44+CD24(-) prostate cells are early cancer progenitor/stem cells that provide a model for patients with poor prognosis. Br J Cancer 2008; 98: 756–765. 46 Li T, Su Y, Leng Q, Leng B, Liu Z, Stass SA et al. ALDH1A1 is a marker for malignant prostate stem cells and predictor of prostate cancer patients' outcome. Lab Invest 2010; 90: 234–244. 47 van den Hoogen C, van der Horst G, Cheung H, Buijs JT, Lippitt JM, Guzman- Ramirez N et al. High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-intiating cells in human prostate cancer. Cancer Res 2010; 70: 5162–5173. 48 Qin J, Liu X, Laffin B, Chen X, Choy G, Jeter CR et al. The PSA(-/lo) prostate cancer cell population harbors self-renewing long-term tumor-propagating cells that resist castration. Cell Stem Cell 2012; 10: 556–569. 49 Foster BA, Gangavarapu KJ, Mathew G, Azabdaftari G, Morrison CD, Miller A et al. Human prostate side population cells demonstrate stem cell properties in recombination with urogenital sinus mesenchyme. PLoS One 2013; 8: e55062. 50 Gangavarapu KJ, Azabdaftari G, Morrison CD, Miller A, Foster BA, Huss WJ. Aldehyde dehydrogenase and ATP binding cassette transporter G2 (ABCG2) functional assays isolate different populations of prostate stem cells where ABCG2 function selects for cells with increased stem cell activity. Stem Cell Res Ther 2013; 4: 132. 51 Jeter CR, Liu B, Liu X, Chen X, Liu C, Calhoun-Davis T et al. NANOG promotes cancer stem cell characteristics and prostate cancer resistance to androgen deprivation. Oncogene 2011; 30: 3833–3845. 52 Gu G, Yuan J, Wills M, Kasper S. Prostate cancer cells with stem cell characteristics reconstitute the original human tumor in vivo. Cancer Res 2007; 67: 4807–4815. 53 Rybak AP, Tang D. SOX2 plays a critical role in EGFR-mediated self-renewal of human prostate cancer stem-like cells. Cell Signal 2013; 25: 2734–2742. 54 Sharpe B, Beresford M, Bowen R, Mitchard J, Chalmers AD. Searching for prostate cancer stem cells: markers and methods. Stem Cell Rev 2013; 9: 721–730. 55 Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006; 444: 756–760. 56 Duru N, Fan M, Candas D, Menaa C, Liu HC, Nantajit D et al. HER2-associated radioresistance of breast cancer stem cells isolated from HER2-negative breast cancer cells. Clin Cancer Res 2012; 18: 6634–6647. 57 Seiler D, Zheng J, Liu G, Wang S, Yamashiro J, Reiter RE et al. Enrichment of putative prostate cancer stem cells after androgen deprivation: upregulation of pluripotency transactivators concurs with resistance to androgen deprivation in LNCaP cell lines. Prostate 2013; 73: 1378–1390. 58 Tang Y, Hamburger AW, Wang L, Khan MA, Hussain A. Androgen deprivation and stem cell markers in prostate cancers. Int J Clin Exp Pathol 2009; 3: 128–138. 59 Abdullah LN, Chow EK. Mechanisms of chemoresistance in cancer stem cells. Clin Trans Med 2013; 2: 3. 60 Chen K, Huang YH, Chen JL. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin 2013; 34: 732–740. 61 Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax JG, Gobel U et al. A distinct ‘side population’ of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci USA 2004; 101: 14228–14233. 62 Zhang L, Jiao M, Li L, Wu D, Wu K, Li X et al. Tumorspheres derived from prostate cancer cells possess chemoresistant and cancer stem cell properties. J Cancer Res Clin Oncol 2012; 138: 675–686. 63 Pollak MN. Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov 2012; 2: 778–790. 64 Kourelis TV, Siegel RD. Metformin and cancer: new applications for an old drug. Med Oncol 2012; 29: 1314–1327. 65 Evans JMM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. Br Med J 2005; 330: 1304–1305. 66 Zakikhani M, Blouin MJ, Piura E, Pollak MN. Metformin and rapamycin have distinct effects on the AKT pathway and proliferation in breast cancer cells. Breast Cancer Res Treat 2010; 123: 271–279. 67 Alimova IN, Liu B, Fan Z, Edgerton SM, Dillon T, Lind SE et al. Metformin inhibits breast cancer cell growth, colony formation, and induces cell cycle arrest in vitro. Cell Cycle 2009; 8: 909–915. 68 Rattan R, Giri S, Hartmann LC, Shridhar V. Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensible manner. J Cell Mol Med 2011; 15: 166–178. 69 Kisfalvi K, Moro A, Sinnett-Smith J, Eibl G, Rozengurt E. Metformin inhibits the growth of human pancreatic cancer xenografts. Pancreas 2013; 42: 781–785. 70 Sahra IB, Laurent K, Loubat A, Giorgietti-Peraldi S, Colosetti P, Auberger P et al. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene 2008; 27: 3576–3586. 71 Preston MA, Riis AH, Ehrenstein V, Breau RH, Batista JL, Olumi AF et al. Metformin use and prostate cancer risk. Eur Urol 2014; 66: 1012–1020. 72 Margel D, Urbach DR, Lipscombe LL, Bell CM, Kulkarni G, Austin PC et al. Metformin use and all-cause and prostate cancer-specific mortality among men with diabetes. J Clin Oncol 2013; 31: 3069–3075. 73 Yu H, Yin L, Jiang X, Sun X, Wu J, Tian H et al. Effect of metformin on cancer risk and treatment outcome of prostate cancer: a meta-analysis of epidemiological observational studies. PLoS One 2014; 9: e116327. 74 Iliopoulos D, Hirsch HA, Struhl K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res 2011; 71: 3196–3201. 75 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 (Phila) 2012; 5: 355–364. 76 Gou S, Cui P, Li X, Shi P, Liu T, Wang C. Low concentrations of metformin selec- tively inhibit CD133+ cell proliferation in pancreatic cancer and have anticancer action. PLoS One 2013; 8: e63969. 77 Mohammed A, Janakiram NB, Brewer M, Ritchie RL, Marya A, Lightfoot S et al. Antidiabetic drug metformin prevents progression of pancreatic cancer by target- ing in part cancer stem cells and mTOR signaling. Transl Oncol 2013; 6: 649–659. 78 Lonardo E, Cioffi M, Sancho P, Sanchez-Ripoli Y, Trabulo SM, Dorado J et al. Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells. PLoS One 2013; 8: e76518. 79 Wurth R, Pattarozzi A, Gatti M, Bajetto A, Corsaro A, Parodi A et al. Metformin selectively affects human glioblastoma tumor-initiating cell viability: a role for metformin-induced inhibition of Akt. Cell Cycle 2013; 12: 145–156. 80 Pollak M. Potential applications for biguanides in oncology. J Clin Invest 2013; 123: 3693–3700. 81 Andrzejewski S, Gravel SP, Pollak M, St-Pierre J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab 2014; 2: 12. 82 Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J 2000; 348 (Pt 3): 607–614. 83 Wheaton WW, Weinberg SE, Hamanaka RB, Soberanes S, Sullivan LB, Anso E et al. Metformin inhibits mitochondrial complex 1 of cancer cells to reduce tumor- igenesis. Elife 2014; 3: e02242. 84 Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, Albright RA et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycero- phosphate dehydrogenase. Nature 2014; 510: 542–546. 85 Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: from mechanisms of action to therapies. Cell Metab 2014; 20: 953–966. Metformin and prostate cancer stem cells MJ Mayer et al 308 Prostate Cancer and Prostatic Diseases (2015), 303 – 309 © 2015 Macmillan Publishers Limited 86 Sahra IB, Regazzetti C, Robert G. Metformin, independent of AMPK, induces mTOR inhibition and cell-cyle arrest through REDD1. Cancer Res 2011; 71: 4366–4372. 87 Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674. 88 Hardie DG. The LKB1-AMPK pathway-friend or foe in cancer? Cancer Cell 2013; 23: 131–132. 89 Colquhoun AJ, Venier NA, Vandersluis AD, Besla R, Sugar LM, Kiss A et al. Met- formin enhances the antiproliferative and apoptotic effect of bicalutamide in prostate cancer. Prostate Cancer Prostatic Dis 2012; 15: 346–352. 90 Nguyen HG, Yang JC, Kung HJ, Shi XB, Tilki D, Lara PNJ et al. Targeting autophagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model. Oncogene 2014; 33: 4521–4530. 91 Zannella VE, Dal Pra A, Muaddi H, McKee TD, Stapleton S, Sykes J et al. Repro- gramming metabolism with metformin improves tumor oxygenation and radio- therapy response. Clin Cancer Res 2013; 19: 6741–6750. 92 Finones RR, Yeargin J, Lee M, Kaur AP, Cheng C, Sun P et al. Early human prostate adenocarcinomas harbor androgen-independent cancer cells. PLoS One 2013; 8: e74438. 93 Jiao J, Hindoyan A, Wang S, Tran LM, Goldstein AS, Lawson D et al. Identification of CD166 as a surface marker for enriching prostate stem/progenitor and cancer initiating cells. PLoS One 2012; 7: e42564. 94 Tokar EJ, Ancrile BB, Cunha GR, Webber MM. Stem/progenitor and intermediate cell types and the origin of human prostate cancer. Differentiation 2005; 73: 463–473. 95 Lawson DA, Xin L, Lukacs R, Xu Q, Cheng D, Witte ON. Prostate stem cells and prostate cancer. Cold Spring Harb Symp Quant Biol 2005; 70: 187–196. 96 Xin L, Lawson DA, Witte ON. The Sca-1 cell surface marker enriches for a prostate- regenerating cell subpopulation that can initiate prostate tumorigenesis. Proc Natl Acad Sci USA 2005; 102: 6942–6947. Metformin and prostate cancer stem cells MJ Mayer et al 309 © 2015 Macmillan Publishers Limited Prostate Cancer and Prostatic Diseases (2015), 303 – 309 Metformin and prostate cancer stem cells: a novel therapeutic target Introduction Prostate structure and development Role of CSCs in prostate cancer Identification and characterization of prostate CSCs Prostate CSCs and therapeutic resistance Metformin and prostate cancer Metformin targets CSCs Mechanism of action of metformin in CSCs Role of metformin in prostate cancer therapy Conclusion References