Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers
Review Article

Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers

Bin Bao1, Asfar S. Azmi1, Shadan Ali2, Feras Zaiem1, Fazlul H. Sarkar1,2

1Department of Pathology, 2Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA

Correspondence to: Fazlul H. Sarkar. Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, 740 Hudson Webber Cancer Research Center, 4100 John R Street, Detroit, MI 48201, USA. Email: fsarkar@med.wayne.edu.

Abstract: Metformin is one of the most used diabetic drugs for the management of type II diabetes mellitus (DM) in the world. Increased numbers of epidemiological and clinical studies have provided convincing evidence supporting the role of metformin in the development and progression of a variety of human tumors including breast and pancreatic cancer. Substantial pre-clinical evidence from in vitro and in vivo experimental studies strongly suggests that metformin has an anti-cancer activity mediated through the regulation of several cell signaling pathways including activation of AMP kinase (AMPK), and other direct and indirect mechanisms; however, the detailed mechanism(s) has not yet been fully understood. The concept of cancer stem cells (CSCs) has gained significant attention in recent years due its identification and defining its clinical implications in many different tumors including breast cancer and pancreatic cancer. In this review, we will discuss the protective role of metformin in the development of breast and pancreatic cancers. We will further discuss the role of metformin as an anti-cancer agent, which is in part mediated through targeting CSCs. Finally, we will discuss the potential role of metformin in the modulation of tumor-associated or CSC-associated microRNAs (miRNAs) as part of the novel mechanism of action of metformin in the development and progression of breast and pancreatic cancers.

Keywords: Metformin; cancer stem cells (CSCs); microRNAs (miRNAs); breast cancer and pancreatic cancer


Submitted Feb 19, 2014. Accepted for publication May 27, 2014.

doi: 10.3978/j.issn.2305-5839.2014.06.05


Introduction

Metformin is one of the most commonly used drugs as the first line of medication for the treatment of diabetes mellitus (DM), especially type-II DM. This drug is inexpensive and very safe for DM patients. Great numbers of clinical and epidemiological reports revealed that the oral administration of metformin reduces the risk of the development and progression of various tumors including pancreatic cancer and breast cancer. The in vitro and in vivo experimental reports also revealed that metformin exhibits an anti-cancer activity, mediated through regulation of AMP kinase (AMPK)/mammalian target of rapamycin (mTOR) and insulin/IGF-1 signaling pathways. These data strongly suggest that metformin may have a protective role against the development and progression various human malignancies; however, the detailed mechanism of action of metformin against tumors has been not been fully elucidated.

The concept of tumor-initiating cells or cancer stem cells (CSCs) has gained significant attention in the field of cancer research. The existence of CSCs have been found in many different malignant diseases, which has been shown to exhibit aggressive features such as longer lifespan, greater capacity of self-renewal, higher potential of resistance to apoptosis, and ability of unlimited differentiation into multiple cell lineages driving proliferation of daughter cells, resulting in rapid tumor growth, migration/invasion, and drug resistance. These aggressive phenotypes of CSCs provide a good reasonable explanation for their clinical implication such as increased tumor relapse/recurrence, a shorter overall disease-free survival rate, and higher cancer-specific mortality. However, the pathogenesis of CSCs during tumorigenesis and tumor progression is still not fully understood.

Mounting evidence suggest that microRNAs (miRNAs), a group of small non-protein-coding RNAs, discovered as the critical regulators of gene expression and critically involved in the regulation of many biological processes such as cell growth/proliferation, differentiation, apoptosis/survival, energy metabolism and homeostasis, all of which may play an important role in the regulation of CSC phenotypes and functions during the development and progression of tumors. In this review article, we will discuss the protective role of metformin in the development of breast and pancreatic cancers. We will further discuss the role of metformin as a potential anti-tumor agent mediated through the inhibition of CSC phenotypes and functions. Finally, we will discuss the potential role of metformin in the regulation of tumor- and CSC-associated miRNAs in the development and progression of breast and pancreatic cancers.


Metformin and DM

It is well known that metformin (chemically named as 1,1-dimethylbiguanide hydrochloride) is widely used as the first line anti-diabetic drug for the management of DM, especially type-II DM in the world. Metformin is one member of the biguanide group of oral hypoglycenic chemicals, which is originally derived from French lilac or goat’s rue Galega officinalis, which was first used for the relief of polyuria in ancient Egypt and medieval European areas (1-3). Several biguanide family members including metformin, phenformin, and buformin were identified as the active components of Galega officinalis in the 1920s, and developed as therapeutic drugs for the management of DM in European countries in the 1950. However, due to the toxicity of lactic acidosis induced by phenformin and buformin, only metformin has been approved to be safe and most effective anti-diabetic drug for the the management of DM in UK in 1958 and USA in the 1990s (1). Occasionally, the metformin administration in DM patients is related to the low incidence rate of lactic acidosis with less than 1/10,000 cases, which is predominately reported in the DM patients who have poor kidney function (4,5), and may be due its impurity of the drug. However, metformin is still considered as a very safe and well-tolerated anti-diabetic drug.

In summary, due to its very cost-effective and safe properties, metformin has been widely prescribed as the first line of medication as an anti-diabetic drug for the management of DM, especially type-II DM in the world after failure of blood sugar control by diets and body weight loss. Although metformin has been widely used as a very effective anti-diabetic drug for many decades, the exact molecular mechanism(s) of metformin action against DM is not completely understood; and its primary systemic action is considered to be the reduction of blood sugar levels via the down-regulation of liver glucose synthesis and up-regulation of glucose intake in peripheral tissues such as fat tissues and skeletal muscles by the activation of AMPK, a central energy sensor located at the cytoplasm of the cells (5-9). Metformin has also been identified to enhance insulin sensitivity and reduce glucose intolerance in the tissues, which is attributed to relief/elimination of insulimia, finally attributing to relief of DM (5,8-10).


DM, breast and pancreatic cancers

DM, especially type II, is one of the most common metabolic diseases with high levels of blood sugar, eventually impairing multiple systems and tissues such as cardiovascular, immunological, and neurological systems in the body. In adults, type-II DM contributes to 90-95% of all DM patients in the world. It is estimated that greater than 1.5 million of subjects are diagnosed as new DM patients each year in the USA; and this figure increases every year and will double in 15-20 years (11). Therefore, DM is still a great challenging health problem in the US and in the world (estimating over 350 million DM patients). One major consequence of DM is attributed to the damage of cardiovascular system together with retinopathy, neuropathy, nephropathy, and immunological deregulation, leading to higher morbidity and mortality. Moreover, DM has been widely considered as high risk for the development and progression of human malignancies including breast and pancreatic cancers (12-15).

Evidence from increased numbers of epidemiological studies reveals that type-II DM is highly associated with high risk of pancreatic cancer development and progression. The interrelationship of DM to pancreatic cancer is somehow tricky because DM may cause the pancreatic cancer development, or displays as the early symptoms and signs of pancreatic cancer (11). However, pancreatic cancer-associated DM is associated with the development of insulin-dependence (16,17). DM-associated pancreatic cancer patients have a longer duration of more than four years of DM history before the onset of pancreatic cancer (16,17). Therefore, the investigation of the course duration of DM before the onset of pancreatic cancer appears to be an important approach to explore the cause-effect relationship between DM and pancreatic cancer. Sufficient data from epidemiological studies provides convincing evidence that DM is positively linked with higher risk of pancreatic cancer, as reviewed elsewhere (11). Several meta-analysis reports also support this hypothesis (18) although this issue still remains controversial. In one large prospective report recruiting 278,761 DM patients and 836,283 control subjects explored the link of DM to pancreatic cancer. The results indicate that DM is associated with a three-fold higher risk for the development of pancreatic cancer (19). Other clinical reports also provide supportive evidence indicating DM as an independent high risk factor for the development of pancreatic cancer (18,20-23).

Increased numbers of epidemiological and clinical data have revealed that type-II DM is linked with increased risk of breast cancer among other human malignancies. Several early epidemiological studies showed strong relationship between DM history and breast cancer mortality (24-27). One recent clinical study confirms that DM is an independent risk factor of lower breast cancer-specific survival and overall survival rates (28). These data suggest that DM plays an important role in the development and progression of breast cancer as well. Further evidence in support of the role of DM in the development and progression of both breast and pancreatic cancer is discussed in the following sections.


Metformin as an anti-tumor agent in breast and pancreatic cancers

Due to the role of DM in the development and progression of various tumors including breast cancer and pancreatic cancer together with current knowledge on the pro-oncogenic insulin/IGF-1 and signaling pathway in tumor development and progression (29,30), several anti-diabetic drugs including metformin has been widely investigated for the treatment and/or prevention of various tumors including breast cancer and pancreatic cancer for more than a decade. A great number of epidemiological and clinical reports have revealed that metformin treatment of DM patients shows a protective effect by reducing tumor incidence, and further improvement of clinical prognosis of tumor patients (24-26,31-33).

Metformin and breast cancer

The data from early epidemiological reports in both DM and non-DM patients indicate that metformin treatment might reduce the risk of the development and progression of breast cancer. Moreover, several case-control clinical studies have revealed a protective role of metformin administered with the adjuvant therapies including chemotherapy and radiotherapy in pancreatic cancer and breast cancer patients (9,34-40).

In 2009, Jiralerspong and associates performed a clinical trial in 2,529 breast cancer patients including 155 DM patients where 68 DM patients received metformin treatment while 87 DM patients did not receive metformin. The results showed that breast cancer patients who took metformin had a 24% complete pathologic response (pCR) rate vs. 8% pCR rate, compared to the breast cancer patients who did not take metformin (41). Other recent clinical studies show that the administration of metformin exhibited a similar protective result in breast cancer patients (34,39,40,42). These reports clearly suggest a protective role of metformin in the development and progression of breast cancer.

Metformin and pancreatic cancer

Several early clinical reports have revealed that metformin may have a protective function in the development and progression of pancreatic cancer. Moreover, one recent meta-analysis report including 6 cohort, 3 case-control, and 2 randomized control studies showed that metformin treatment might have a greater role in the reduction in the risk of pancreatic cancer although there was no statistical association between metformin and the risk of pancreatic cancer (n=9 studies; adjusted OR 0.76, 95% CI: 0.57-1.03, P=0.073), which could have been due to considerable heterogeneity across studies (43). A recent large case-control clinical trial recruiting 973 pancreatic cancer patients (259 DM patients included) and 863 non-tumor control subjects (109 DM patients included) was performed for assessing the role of metformin in the risk for the development and progression of pancreatic cancer. The results showed that DM patients treated with metformin had greatly reduced risk of pancreatic cancer, compared to those who did not receive metformin (44). These data clearly suggest a protective role of metformin in the development and progression of pancreatic cancer although this field needs further clarification.


Molecular mechanism of metformin as an anti-tumor agent

Although metformin has been shown to possess anti-tumor activity in certain tumors including pancreatic cancer and breast cancer, the detailed mechanism(s) of metformin action against tumor is yet to be fully understood. Greater numbers of experimental reports suggest that the anti-tumor effect of metformin could be linked with its direct and indirect mechanisms (11). Its indirect mechanisms, which are considered as insulin dependent, are to enhance insulin sensitivity and to reduce the blood levels of glucose and insulin, resulting in the inhibition of insulin/IGF-1 signaling pathway, and eventually resulting in the inhibition of tumor cell growth. It’s direct mechanisms, which are considered as insulin-independent, is to primarily activate AMPK, a central energy sensor, in turn leading to the inhibition of mTOR signaling pathway (45,46). Specifically, when the level of cellular ATP decreases, resulting in an increase in the ratio of AMP/ATP, AMPK is activated, attributing to the inactivation of Akt/mTOR signaling mediated through the inhibition of mTOR by activation of LKB1, a known tumor suppressor. The inhibition of mTOR activity leads to the inhibition of glucose synthesis in the liver and protein synthesis, attributing to the inhibition of cell growth/proliferation of tumor cells mediated through the reduction of bioavailability of intracellular energy and nutrients such as glucose and amino acids (34,46,47). Moreover, metformin can also directly inhibit tumor cell growth/proliferation via modulation of cyclin D1-medicated cell cycle and the expression of tumor suppressor p53 in different tumor cells including pancreatic cancer and breast cancer (11).

It is known that metformin is able to diminish the pro-inflammatory cytokines TNF-α and IL-6, as well as angiogenic cytokine VEGF by inhibition of pro-oncogenic pathway such as NF-κB and HIF-1α, resulting in the inhibition of cell growth/proliferation (11). It is also known that the metformin-mediated down-regulation of glucose synthesis in the liver and skeletal muscles and up-regulation of glucose uptake in the peripheral tissues is fundamentally mediated through the activation of AMPK, which can lead to the reduction of blood sugar and insulin levels, resulting in the inhibition of insulin/IGF-1 pathway, which leads to the inhibition of cell growth and proliferation (11). One recent experimental study demonstrates that metformin treatment in xenograft mouse model improves tumor oxygenation and increase radiotherapy sensitivity, which may be highly associated with a decrease in early biochemical relapse rates in the clinical setting (48). These findings indicate that metformin has an anti-tumor effect partially through the regulation of tumor micro-environment, resulting in an increase in the treatment sensitivity.


CSC concept, tumor aggressiveness, and breast and pancreatic cancers

Although the CSC concept was proposed several decades ago, it has received more and more attentions since Bonnet and his team, for the first time, identified and characterized a small subpopulation of CSCs from the bone marrow of patients in 1997 (49). Subsequent clinical and experimental studies provided additional evidence supporting the critical role of CSC in tumor aggressive phenotypes such as treatment resistance, tumor metastasis and tumor recurrence, attributing to poorer clinical prognosis of patients diagnosed with many different types of tumors including breast cancer and pancreatic cancer (50-55).

By sharing common properties with normal stem cells in the body, CSCs have several unique features such as longer life span, higher potential for apoptotic resistance, and greater capacities of self-renewal coupled with highly uncontrolled differentiation capacity into multiple different cell lineages. The CSC concept has an important clinical implication because they have been identified in many different tumors including pancreatic cancer and breast cancer, and they are associated with progression of these tumors (54,55). For example, breast CSCs have been identified as a rare sub-population (0.1% to 1%) of breast cancer cells in the primary tumor tissue. These rare sub-groups of breast CSCs have a greater ability of CSC self-renewal and have the high capacity of initiation of tumor formation when implanted into immunologically compromised animals such as NOD/SCID mice (56,57).

Several stem cell markers such as CD44+/CD24–, CD133, and ALDH+ have been used to identify the small sub-populations of several different breast CSCs from breast cancer (58). For example, the implantation of 500-1,000 of CSC cells with the CD44+/CD24– phenotype was able to initiate 80% of tumor formation in xenograft mouse models. However, the implantation of more than 10,000 of non-CSC-like breast tumor cells failed to initiate tumor formation (56,57), which is consistent with similar results from other reports by using other CSC marker-positive phenotypes such as CD133+ CSC-like and CD44+/CD24–/ALDH+ CSC-like cells (54,59). These reports suggest that breast CSC plays an important role in tumorigenesis and progression of breast cancer.

The small sub-population of pancreatic CSC cells has also been identified from the primary pancreatic cancer tissues, comprised of less than 1% of pancreatic tumor cells in the tumor tissues (54,55,60). These CSC cells have a greater ability of self-renewal and higher capacity of unlimited differentiation into multiple cell lineages. Several CSC markers such as CD44+, CD24+, CD133+ ESA+/EpCAM+ have been used to identify the CSC sub-population (55,56,61-63). It has been reported that when less than 500-1,000 of CSC cells with the CD44+/CD24+/ESA+ phenotype isolated from human primary pancreatic cancer were implanted into NOD/SCID mice (54,55,61), these sub-population of CSCs showed higher potential of tumor formation. However, the implantation of non-CSC tumor cells (namely CD44-/CD24–/ESA– parental tumor cells) failed to initiate the tumor formation (54,55,61). These CSCs exhibit a 100-fold higher potential of tumor formation, and further revealed that the histo-morphology of these tumors was similar to those of its primary tumor. Additionally, these sub-populations of CSCs can sustain CSC signatures after many passages as xenograft tumor in NOD/SCID mice (55,60).

Our recent data from the in vitro and in vivo experimental studies demonstrates that the implantation of 5,000-10,000 of human pancreatic cancer MiaPaCa-2 sphere-forming (CSC-like) cells can initiate tumor formation in SCID mice, compared to the implantation of 106-107 of its parental MiaPaCa-2 cells for the initiation of tumor formation in the same animal models for similar time period. The CSC-like cells derived from MiaPaCa-2 pancreatospheres isolated from mouse xenograft tumor tissues displayed higher tumor cell aggressive phenotypes such as higher potential of self-renewal and migration/inversion capacity consistent with remarkably higher levels of CD44+ and EpCAM, and enhancement of zeste homolog 2 (EZH2), a known epigenetic mediator that is involved in the enrichment of CSC characteristics (63).

Our unpublished data also showed that the CSC-like (CD44+/CD33+/EpCAM+) cells isolated from pancreatic cancer MiaPaCa-2 and L3.6pl cells by FACS technique display very highly aggressive phenotypes of CSC cells such as increased capacities of colony formation, CSC self-renewal, cell migration/invasion, and apoptotic resistance, all of which was consistent with significantly higher expression of CSC markers/mediators such as Notch-1, Gli, EZH2, FoxQ1, snail, and HIF-1α, as well as increased production of angiogenic cytokine VEGF. These CSC-like cells (CD44+/CD33+/EpCAM) cells also displayed highly tumorigenic potential (at least 100-fold higher) in xenograft mouse models, compared to its parental MiaPaCa-2 cells. Histological study revealed that xenograft tumors derived from these triple positive CSC-like cells exhibited remarkably higher cell proliferation, and increased expression of EpCAM, CD44, VEGF, Notch-1, EZH2, and FoxQ1, which was consistent with tumor cell dedifferentiated state, compared to the xenograft tumor tissues derived from its parental MiaPaCa-2 cells. These data clearly indicate that the CSC cells of pancreatic cancer display an important role in tumor development and progression of pancreatic cancer. Therefore, targeting or elimination of these CSCs will likely provide a newer avenue for the treatment of tumors including breast cancer and pancreatic cancer.


Metformin and CSCs

Recently, the inhibitory activity of metformin on CSC phenotypes and functions have received more attention. We showed that metformin treatment attenuates the CSC phenotypes and functions such as CSC self-renewal capacity and CSC signature and its mediators, consistent with attenuation of cell proliferation and migration in pancreatic cancer drug-resistant cells (64). It has also been reported that metformin selectively diminished the expansions of CSC clones through induction of apoptotic function and the inhibition of CSC mediators and markers, consistent with attenuation of tumor growth in animal models. However, non-CSC tumor cells only showed cell cycle arrest, but was unable to eliminate the cells when treated with metformin (65). These results may indicate that CSCs or CSC-like cells appear to be more vulnerable to metformin treatment than non-CSC tumor cells. The reason why the CSC cells appear to be more sensitive to metformin may be due to its primary mechanism on glucose metabolism. It has been noted that under the conditions of glucose deprivation, metformin treatment increases apoptotic cell death in breast cancer cells, while under the conditions of high level of glucose, metformin treatment mostly caused cell cycle arrest without the signs of apoptotic cell death in breast cancer cells (66). High levels of glucose has also been reported to increase the percentage of the side population (SP, CSC-like) cells, consistent with the activation of Akt pathway and suppression of AMPK activation, leading to the elevation of ABCG2 expression (67). These data suggest that CSC cells may be more sensitive to metformin, potentially due to its modulation of glucose homeostasis.

One recent experimental study showed that low concentration of metformin selectively inhibits cells proliferation of pancreatic CSC-like (CD133+) cells, and suppresses tumor growth, which was in direct agreement with the inactivation of ERK and mTOR networks independent of AKT and AMPK activity (68). Another experimental study also revealed that metformin treatment profoundly inhibits the number of EpCAM+ hepatocellular carcinoma (HCC) cells and significantly inhibits CSC self-renewal ability in vitro and tumor burden in vivo through the AMPK/mTOR independent pathways (69). These results indicate that metformin might inhibit the CSC phenotypes and functions; however, the precise mechanism(s) of action of metformin has not yet been fully elucidated. It has been noted that the anti-tumor activity of metformin may be associated with its regulations of several tumor suppressor genes, which may attribute to the modulation of CSC phenotypes and functions. For example, one recent experimental study has revealed that metformin treatment increases AMPK activation as well as the expressions of PARP, PTEN, and SPRY2 (70), accompanied by the attenuation of cell migration and promotion of apoptotic cell death in tumor cells (70).

Although the evidence from experimental reports suggests that metformin exert an inhibitory effect on CSC phenotypes and functions in HCC and pancreatic cancer cells by the mechanism of the AMPK activation independent pathway (68,69), AMPK has been known as a potent regulator of the differentiation of various stem cells such as mesenchymal cells, and exerts critical functions in the determination of cell fate of normal stem cells such as MSC (71). It has been found that the activation of AMPK causes promotion of the differentiation of MSC into osteogenesis and the inhibition of adipogenesis via the activation of SIRT1 (71), suggesting that the role of AMPK activation in the modulation of stem cell homeostasis appears to be cell-type specific.

One recent study reveals that stem cell signature Sox2 over-expressing CSC-like (called as induced pluripotent stem cells, iPSC) cells of breast cancer show a remarkable down-regulation of the gene expressions of PRKAA1 encoding the catalytic alpha 1 subunit of AMPK, DDIT4/REDD1 encoding a stress response protein acting as a negative regulator of mTOR, and DEPTOR encoding a protein acting as an endogenous inhibitor of mTOR activity, as well as the up-regulation of insulin receptor gene in CSC-like cells (iPSC) (72). Sox2 over-expressing CSC-like cells also have increased phosphorylation of mTOR (72). Thus metformin might exert pivotal roles in the attenuation of CSC phenotypes and functions, mediated through the activation of AMPK signaling pathway. However, further investigations are warranted to explore how metformin inhibits CSC phenotypes and functions.


The role of microRNAs (miRNAs) in the modulation of CSC phenotypes

The discovery of miRNAs, a large group of non-protein coding RNA molecules (average 22 nucleotides in length), has provided a newer insight on cancer biology. The miRNAs have been widely considered to act as endogenous mediators of gene expression by their site-specific binding at the 3' un-translated region (3'-UTR) or other sites of the genes, attributing to either the degradation of their target mRNAs or inactivation of protein synthesis (73,74). The recent evidence suggest that miRNAs may also mediate gene expression by their site-specific binding to the DNA open reading frames of the target genes, attributing to the inhibition of gene transcription. Currently, near three thousands different miRNAs have been discovered in humans and animals that take part in the regulation of gene expression of more than one third of all expressed genes, which are typically involved in a variety of biological processes such as cell proliferation, differentiation, survival/apoptosis, immune response, and energy homeostasis/metabolism (75,76). The data from numerous clinical reports revealed that altered expression of many different miRNAs which is associated with poor clinical outcome of a variety of tumors including breast cancer and pancreatic cancer, suggesting the pivotal functions of miRNAs in tumorigenesis and tumor progression (77-81). The evidence from a great number of experimental studies in vitro and in vivo has supported that miRNAs exert pivotal functions in the tumorigenesis and progression via the regulation of pro-oncogenic/anti-oncogenic signaling pathways via targeting different genes. More importantly, miRNAs have been more and more considered as potential mediators of CSC phenotypes and functions via the modulation of several pro-oncogenic pathways, attributing to tumor aggressive phenotypes such as rapid cell proliferation, invasiveness, treatment resistance, and metastatic potential. In the following paragraphs, we will discuss several well-established tumor-associated miRNAs, which are documented to be associated with the modulation of CSC characteristics attributing to tumor aggressive phenotypes.

Let-7

Let-7 is one of the most studied miRNA and may exert pivotal roles during tumorigenesis and tumor progression by targeting several pro-oncogenic/anti-oncogenic pathways. The deregulation of let-7 expression has been reported to be highly linked with poor clinical prognosis of many different tumors including breast cancer and pancreatic cancer. Substantial evidence from experimental studies in vitro and in vivo suggests that let-7 acts as a potent tumor suppressor molecule. For example, It has been reported that let-7 acts as negative mediator of the acquisition of epithelial-to-mesenchymal transition (EMT) of tumor cells, which is partially modulated by the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a well-established tumor suppressor which diminishes the activation of Akt/mTOR signalings, and the expression of stem cell signature gene Lin-28/Lin-28B which controls cell fate and dormancy of CSCs in several different tumors including breast cancer and pancreatic cancer cells (82-86). The acquisition of EMT characteristics of tumor cells has been considered as an important biological process during the development and growth of certain organs such as teeth, and wound healing after birth, which is considered to be reminiscent of CSCs, leading to high tumor aggressive phenotypes such as cell migration/invasion, drug resistance, and metastasis.

It has been noted that the expression of let-7 is either lost or significantly down-regulated in normal stem cells and in CSC cells of various tumor cells (87-89), and is inversely linked with the expression of stem cell signature gene Lin-28/Lin-28B. Furthermore, let-7 expression has been found to bi-directionally inhibited by Lin-28/Lin-28B (89), consistent with the results from other reports in which let-7 negatively targets several pro-oncogenic genes including Lin-28/Lin-28B (90-93). It has also been found that the re-expression of let-7 in breast CSCs decreased cell proliferation, CSC self-renewal capacity, tumorigenic potential, and tumor metastasis in animal xenograft tumor model, which was consistent with attenuation of the expansions of undifferentiated tumor cells in cell culture (88). Moreover, our published data showed that let-7 can inhibit the expression of EZH2, a key epigenetic mediator of the expression of the genes that have important functions in normal stem cells to regulate the expression of genes that are preferentially and selectively up-regulated during development and differentiation via a direct inhibition of anti-oncogenic molecules (94). Currently, EZH2 has been increasingly considered as a potential mediator for promoting the CSC phenotypes and functions via the modulation of several pro-oncogenic networks (95-97). Our unpublished data showed that the CSC-like (CD44+/CD133+/EpCAM+) cells derived from pancreatic cancer cells have very low levels of let-7f and i, compared to the triple marker negative non-CSC tumor cells and its parental cells. Therefore, let-7 expression may exert key roles in the modulation of CSC phenotypes and functions by targeting several cell signaling pathways.

The role of metformin in the modulation of the expression of let-7 during tumorigenesis and progression has been not fully elucidated. One recent experimental study reveals that metformin treatment significantly increased the levels of let-7a in breast cancer MCF-7 cells, consistent with the inhibition of TGF-β-induced mammosphere-forming potential (98). Emerging evidence suggests that the anti-tumor effect of metformin could be linked with attenuation in the expression and/or activity of Lin28/Lin28B mediated through the up-regulation of let-7 expression via an AMPK activation-dependent pathway (84). Other experimental studies also reveals that metformin induces the expression of let-7c in pancreatic cancer cells (99). Our published data confirmed that metformin treatment induced the expression of let-7b and c in the CSC-like pancreatosphere cells, consistent with attenuation of CSC phenotypes and function of drug-resistant pancreatic tumor cells (64). These reports clearly suggest that metformin may have a regulatory function on the expression of let-7 family during the development and progression of tumors including breast cancer and pancreatic cancer.

miR-21

Currently, the miR-21 is another most studied miRNA in cancer biology, and it is widely considered as a potential oncogene molecule which is involved in the deregulation of multiple cell signaling pathways. The clinical data have shown increased levels of miR-21 in many different tumors including breast cancer and pancreatic cancer. The altered expression of miR-21 has been observed to be highly linked with the clinical prognosis of cancer patients (100,101). The evidence from our recent study suggests that the high levels of miR-21 expression in pancreatic cancer cells and drug-resistant tumor cells may cause the down-regulation of PTEN expression, attributing to the activation of Akt/ERK networks, further attributing to the promotion of tumor cell growth and progression (63,102). Recently, the evidence indicates that miR-21 might exert pivotal functions in the regulation of CSC phenotypes and functions. It has been found that the expression of miR-21 is very high in CSC cells in the comparison with non-CSC tumor cells (87,103). The miR-21 overexpression can increase the potential of hypoxia-mediated survival of the bone marrow mesenchymal stem cells. The loss of miR-21 expression induces apoptotic cell death of these stem cells (104). Additionally, the loss of miR-21 expression enhances cell differentiation, and inhibits the CSC self-renewal capacity of drug-resistant colon cancer cells, which is in direct agreement with the down-regulation of stem cell surface marker CD44, and TCF/LEF, a positive regulator of CSC phenotypes and functions (105). We have also demonstrated that the inhibition of miR-21 by the functional loss technique causes the attenuation of CSC self-renewal capacity through the down-regulation in the expression of CD44 and EpCAM in the CSC-like cells of prostate and pancreatic cancers under hypoxia (106,107). These results clearly indicate an important role of miR-21 in the modulation of CSC phenotypes and functions which is mediated via the regulation of several cell signaling networks. However, the role of metformin in the regulation of miR-21 expression has been not fully elucidated. The limited data show that metformin treatment enhances miR-21 expression in gastric tumor cells in vitro and in vivo as demonstrated by micro-array analysis (108). Additional studies are warranted to appreciate the role of metformin in the modulation of miR-21 expression in tumor development and progression of pancreatic cancer and breast cancer.

miR-26a

Mounting evidence suggests that miR-26a might function as a potent tumor suppressor by targeting multiple cell signaling pathways such as IL-6/STAT, Myc, and MCL-1. The loss or lower expression of miR-26a has been identified in a variety of tumors including breast cancer and pancreatic cancer, and it has been found to be associated with poor clinical prognosis of cancer patients (77,109). Recently, several experimental studies have revealed a regulatory role of miR-26a in the modulation of cancer epigenome via the inhibition in the expression of EZH2, a potential CSC mediator (110-115).

It has been observed that miR-26a over-expression by functional gain technique can decrease the expression of EZH2 in tumor cells, attributing to the suppression of tumor invasive and metastatic potentials in vitro and in vivo (110,113,115). Therefore, targeting miR-26a would represent a newer avenue toward the treatment and/or prevention of aggressive tumors. Our published data showed that miR-26a re-expression by the functional gain technique decreased CSC signatures and its mediators EpCAM, EZH2, Oct4, and Notch-1 in pancreatic cancer cells (64), which strongly suggests the important function of miR-26a in the modulation of CSC phenotypes and functions through regulation of CSC signature/mediator genes.

Emerging evidence also suggest that metformin may target the expression of miR-26a, consistent with inhibition of CSC phenotypes and function in tumor development and progression. One recent experimental study showed that metformin treatment increased the expression of miR-26a, consistent with the inhibition of cell proliferation in gastric cancer (108). Moreover, It has been reported that metformin treatment could enhance miR-26a expression in a dose-dependent fashion by negatively targeting HMGA1, a known oncogene in pancreatic tumor cells, consistent with the inhibition of tumor aggressive phenotypes (99). Our published data confirmed that metformin treatment could enhance miR-26a expression in the CSC-like pancreatosphere cells, consistent with attenuation of CSC phenotypes and function of drug-resistant pancreatic cancer cells (64). However, one experimental study showed that metformin treatment could decrease the expression of miR-26a in prostate cancer cells (116), clearly suggesting that more in-depth mechanistic studies are warranted for delineating the role of metformin in the deregulation miR-26a and others in breast and pancreatic cancer.

miR-146a

Increased numbers of clinical reports indicate that miR-146a might play a pivotal role during the tumor development and progression of various tumors including breast cancer and pancreatic cancer (117). The loss of expression or reduced expression of miR-146a has been discovered which was tightly associated with poor clinical outcome of patients diagnosed with a variety of cancers (77,117-119). The data from several experimental reports showed that miR-146a might act as a tumor suppressor molecule mediated through the suppression of aggressive phenotypes of various tumors including breast cancer and pancreatic cancer through the regulation of various cell signaling networks such as NF-κB, MMP, CXCR4, STAT, K-ras, and Notch-1 (118,120-122). For example, miR-146a has been found to suppress NF-κB activation, attributing to the repression of gene expression of NF-κB-mediated proinflammatory molecules interleukin-1β (IL-1β), IL-6, IL-8, and TNF-α via the inhibition of IL-1 receptor associated kinase 1 (IRAK1) and TNF receptor associated factor 6 (TRAF6) (123). The NF-κB activation pathway is widely considered to be involved in the promotion of CSC phenotypes and functions via up-regulation of CSC signatures Nanog, Sox2, and Lin-28/Lin-28B (124). It has been noted that the loss of miR-146a expression results in the activation of NF-κB, accompanied by increased resistance to apoptosis of gastric cancer cells (125). Our recent findings confirmed that miR-146a expression was significantly decreased in pancreatic tumor cells and Kras/Cre-transgenic animals of pancreatic tumor, accompanied by increased tumor aggressive phenotypes (118,120). The functional gain analysis studies showed that re-expression of miR-146a attenuates EGFR and NF-κB activity, attributing to the inhibition of NF-κB downstream targets, accompanied by the suppression of tumor cell invasiveness (118). These findings clearly indicate that miR-146a might function as a suppressor of tumorigenesis partially mediated by targeting NF-κB signaling pathway.

Recently, the data from experimental studies have revealed that miR-146a might display a key role in the regulation of the hemostasis of normal stem cells by targeting several cell signaling networks such as NF-kB and Notch-1 (121,126). For example, miR-146a has been observed to suppress the development of glioma by down-regulation of Notch-1 activity (121). However, one experimental report revealed that the CD133+ sphere-forming cells of ovarian cancer have increased expression of miR-146a (127). In general, these findings suggest that miR-146a might exert pivotal functions in the modulation of CSC phenotypes and functions which clearly suggest that further in-depth mechanistic studies are warranted to ascertain the role of miR-146a in human malignancies including breast and pancreas cancers.

The role of metformin in the regulation of miR-146a during tumorigenesis is not clear. One experimental report showed that metformin treatment leads to decreased expression of miR-146a in prostate cancer PC-3 cells as documented by micro-array analysis (116). However, more studies will be needed to explore the role of metformin in the modulation of miR-146a in breast and pancreatic cancer.

miR-200

The miR-200 is another most studied miRNA family which has been widely recognized to play pivotal roles in human tumors including breast cancer and pancreatic cancer. It has been reported that the alterations in the expression of miR-200 are important in many different tumors, and the expression is highly linked with poorer clinical outcomes of cancer patients including those diagnosed with breast cancer and pancreatic cancer. The evidence from numerous experimental reports clearly suggests that miR-200 acts as potential tumor suppressor molecule, which is partially mediated through the suppression of EMT characteristics by directly targeting ZEB1/2, a known EMT marker. The data from early experimental studies have revealed that miR-200, for example, miR-200b and c, can inhibit the expression of ZEB1/2 by its direct binding to the 3'UTR of ZEB1/2 genes, attributing to the inhibition in the induction of EMT characteristics in tumor cells, resulting in the reversal of EMT phenotype (128). The data from our experimental studies have shown that gemcitabine-resistant pancreatic cancer cells have very low levels of miR-200 including a, b, and c and it was correlated with the mesenchymal morphology consistent with EMT phenotype and tumor cell aggressive phenotypes (102). The functional gain analysis studies showed that the re-expression of miR-200 in these drug-resistant tumor cells or PDGF-induced EMT phenotypic PC-3 prostate tumor cells resulted in the down-regulation in the expression of ZEB1/2 and Slug, and it was also associated with increased expression of E-cadherin, an epithelial marker, suggesting reversal of EMT phenotype (102,129). These findings are in direct agreement with the findings reported by other investigators (85,86,130). Additionally, it has also been noted that miR-200 over-expression can decrease Bmi1, Suz12, and Notch-1, the well-established mediators of CSC and EMT characteristics in a variety of tumor cells, which are accompanied by the inhibition of CSC phenotypes and functions (131-133). Importantly, the loss or lower expression of miR-200 has been found in CSC-like cells with the CD44+/CD24– phenotype of breast tumor (134), which is consistent with our unpublished data from miRNA profiling studies showing that the CSC-like (CD44+/CD133+/EpCAM+) cells derived from MiaPaCa-2 cells display alternations in the expression of miR-200 compared to non-CSC triple marker negative cells and its parental MiaPaCa-2 cells. These findings strongly suggest that miR-200 may have a pivotal role in the modulation of CSC phenotypes and functions mediated via targeting several cell signaling networks.

It has been noted that metformin treatment could lead to the up-regulation of miR-200, accompanied by the reduction of the number and size of induced pluripotent stem cell (CSC-like) colonies as well as the inhibition in the expression of pluripotent marker alkaline phosphatase, accompanied by stress-induced senescence in human diploid fibroblasts (135). One recent experimental study showed that metformin treatment could lead to increased expression of miR-200, accompanied by the inhibition of cell proliferation in gastric cancer (108). Our published data showed that metformin treatment could lead to increased expression of miR-200b and c in CSC-like pancreatosphere cells, consistent with the suppression of CSC phenotypes and function of drug-resistant pancreatic tumor cells (64). These findings indicate that metformin might have inhibitory effects on CSC phenotypes and functions in the development and progression of breast cancer and pancreatic cancer, in part, mediated through up-regulation of miR-200.

miR-451

A significant number of clinical reports provided supportive evidence showing that miR-451 may play a key role in tumor development and progression (136). It has been noted that the loss or lower expression of miR-451 has been identified in several different tumors such as gastric cancer, colorectal cancer, NSCLC, renal cell carcinoma, glioblastoma and pre-B-ALL (childhood B-cell precursor acute lymphoblastic leukemia) and was found to be associated with poor clinical prognosis (136-140). Evidence from experimental studies suggests that miR-451 may have an anti-tumor function; however, one clinical report showed that the level of miR-451 was higher in glioblastoma multiforme tissue compared to the adjacent normal brain tissue (141,142), suggesting organ specific role of miR-451. Another report also showed that breast cancer patients have increased levels of plasma miR-451, compared to that of normal control subject (143). These data suggest the potential roles of miR-451 in tumor development and tumor progression although it may function in an organ specific manner. Increased numbers of experimental studies supports the potential function of miR-451 as an anti-tumor molecule, which is in part mediated via targeting several pathways such as MIF, RAS-related protein 14 (RAB14), calcium binding protein 39 (CAB39), Akt, and Bcl-2. For example, re-expression of miR-451 diminishes cell proliferation and promotes apoptotic cell death of NSCLC cells by negatively targeting RAB14, a pro-oncogenic molecule. The high level of miR-451 led to induced sensitivity of tumor cells to chemotherapy, via the inhibition of Akt signaling pathway (144). It has been observed that the re-expression of miR-451 led to increased suppression of cell survival, growth/proliferation, and migration/invasion, accompanied by the inhibition of Akt and Bcl-2 in esophageal cancer cells (145). In the tumor xenograft mouse model study, miR-451 has been shown to inhibit tumor growth of human esophageal carcinoma (145). It has also been found that miR-451 displays an anti-tumor function in cell culture and xenograft tumor models, which is in direct agreement with the suppression of PI3K/Akt pathway targeting CAB39 in glioma (140).

The role of miR-451 in the modulation of CSC characteristics has not been fully elucidated. Limited data suggest that miR-451 might have regulatory roles in CSC characteristics. One experimental study showed that miR-451 was down-regulated in CSC-like cells (colonosphere cells) compared to its parental cells. Over-expression of miR-451 caused a decrease in the CSC self-renewal capacity, tumorigenic potential, drug resistance in CSC-like colon cancer cells, which was consistent with the inhibition of ABCB1 (ATP-binding cassette drug transporter) by indirectly targeting COX2/Wnt pathways (146). Clinical studies showed that the colorectal cancer patients who did not respond to chemotherapy have lower expression of miR-451, compared to those patients who responded to the chemotherapy (146).

The role of metformin in the regulation of miR-451 during tumor development and progression has not yet been fully elucidated although the data clearly suggest that the expression of miR-451 is glucose-dependent. High glucose level increased miR-451 while glucose deprivation decreased the expression of miR-451 (141,142). The expression of miR-451 has been found to be associated with glucose metabolism, and it may act as a sensor of glucose level in the cells. It has been noted that high levels of glucose increases the expression of miR-451, resulting in the inactivation of AMPK by negatively targeting LKB1, a positive regulator of AMPK (147,148). Glucose withdrawal decreases the expression of miR-451, resulting in the activation of AMPK (147,148). Metformin apparently regulates glucose homeostasis via the activation of AMPK. However, further studies are needed to firmly establish the interrelationship between metformin, AMPK, and miR-451 in the regulation of CSC phenotypes and functions in breast and pancreas cancers.


Conclusions

We have tried to make efforts to summarize the “state-of-our-knowledge” on the potential actions of metformin on the expression of tumor-related miRNAs in the regulation of CSC characteristics during the development and progression of various tumors including breast cancer and pancreatic cancer. We have presented evidence as concisely as possible due to the limitation of the space/pages; during such effort we could not cite all the published data, and thus we sincerely apologize to those whose work could not be cited here. In summary, metformin is an inexpensive and safe anti-diabetic drug which has been widely used for the management of DM. Numerous epidemiological, clinical, and experimental reports have revealed that metformin may also function as an anti-tumor agent in a variety of tumors including breast cancer and pancreatic cancer, suggesting that metformin may provide a newer avenue for the treatment and/or prevention of human malignancies including breast and pancreas cancers. However, the precise molecular mechanisms of action of metformin have not yet been fully elucidated, which opens newer areas of cutting-edge investigations. The identification of CSCs in many different tumors including breast cancer and pancreatic cancer and its characteristics such as longer lifer-span, higher capacity of CSC self-renewal, increased resistance to apoptosis, and greater potential of differentiation into multiple cell lineages and unlimited proliferation of its daughter tumor cells are very important because these have been clinically linked with rapid tumor growth and metastasis, chemo-radio-therapy resistance, and tumor recurrence/relapse, all of which leads to poor clinical outcome of patients diagnosed with breast cancer and pancreatic cancer. The evidence from recent experimental studies has shown that metformin may selectively inhibit the CSC phenotypes and functions, which may be responsible for inhibition of tumor development and progression. Moreover, emerging evidence suggests that metformin may play important roles in the modulation of tumor-associated or CSC-associated miRNAs that are critically important in the development and progression of a variety of tumors including breast cancer and pancreatic cancer. However, more studies are needed for appreciating the molecular role of metformin in the regulation of miRNAs as well as the functions of CSCs, and further realizing the potential of metformin for the prevention and/or treatment of human malignancies including breast and pancreas cancers.


Acknowledgements

Disclosure: The authors declare no conflict of interest.


References

  1. Bailey CJ, Turner RC. Metformin. N Engl J Med 1996;334:574-9. [PubMed]
  2. Hadden DR. Goat’s rue-French lilac-Italian fitch-Spanish sainfoin: gallega officinalis and metformin: the Edinburgh connection. J R Coll Physicians Edinb 2005;35:258-60. [PubMed]
  3. Witters LA. The blooming of the French lilac. J Clin Invest 2001;108:1105-7. [PubMed]
  4. Lalau JD, Race JM. Lactic acidosis in metformin-treated patients. Prognostic value of arterial lactate levels and plasma metformin concentrations. Drug Saf 1999;20:377-84. [PubMed]
  5. Lalau JD. Lactic acidosis induced by metformin: incidence, management and prevention. Drug Saf 2010;33:727-40. [PubMed]
  6. Brouwers MC, de Graaf J, van Greevenbroek MM, et al. Novel drugs in familial combined hyperlipidemia: lessons from type 2 diabetes mellitus. Curr Opin Lipidol 2010;21:530-8. [PubMed]
  7. Ibarra-Drendall C, Dietze EC, Seewaldt VL. Metabolic Syndrome and Breast Cancer Risk: Is There a Role for Metformin? Curr Breast Cancer Rep 2011;3:142-50. [PubMed]
  8. Vona-Davis L, Rose DP. Type 2 Diabetes and Obesity Metabolic Interactions: Common Factors for Breast Cancer Risk and Novel Approaches to Prevention and Therapy. Curr Diabetes Rev 2012;8:116-30. [PubMed]
  9. Shaw RJ, Lamia KA, Vasquez D, et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 2005;310:1642-6. [PubMed]
  10. Scheen AJ. Pharmacokinetic and pharmacodynamic evaluation of sitagliptin plus metformin. Expert Opin Drug Metab Toxicol 2010;6:1265-76. [PubMed]
  11. Bao B, Wang Z, Li Y, et al. The complexities of obesity and diabetes with the development and progression of pancreatic cancer. Biochim Biophys Acta 2011;1815:135-46.
  12. Huxley R, Ansary-Moghaddam A, Berrington de González A, et al. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer 2005;92:2076-83. [PubMed]
  13. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. Diabetes Care 2010;33:1674-85. [PubMed]
  14. De Bruijn KM, Arends LR, Hansen BE, et al. Systematic review and meta-analysis of the association between diabetes mellitus and incidence and mortality in breast and colorectal cancer. Br J Surg 2013;100:1421-9. [PubMed]
  15. Anothaisintawee T, Wiratkapun C, Lerdsitthichai P, et al. Risk factors of breast cancer: a systematic review and meta-analysis. Asia Pac J Public Health 2013;25:368-87. [PubMed]
  16. Gullo L, Pezzilli R, Morselli-Labate AM. Diabetes and the risk of pancreatic cancer. N Engl J Med 1994;331:81-4. [PubMed]
  17. Pezzilli R, Casadei R, Morselli-Labate AM. Is type 2 diabetes a risk factor for pancreatic cancer? JOP 2009;10:705-6. [PubMed]
  18. Malek M, Aghili R, Emami Z, et al. Risk of Cancer in Diabetes: The Effect of Metformin. ISRN Endocrinol 2013;2013:636927. eCollection 2013.
  19. Jamal MM, Yoon EJ, Vega KJ, et al. Diabetes mellitus as a risk factor for gastrointestinal cancer among American veterans. World J Gastroenterol 2009;15:5274-8. [PubMed]
  20. Chari ST, Leibson CL, Rabe KG, et al. Pancreatic cancer-associated diabetes mellitus: prevalence and temporal association with diagnosis of cancer. Gastroenterology 2008;134:95-101. [PubMed]
  21. Pannala R, Leirness JB, Bamlet WR, et al. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology 2008;134:981-7. [PubMed]
  22. Pannala R, Basu A, Petersen GM, et al. New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol 2009;10:88-95. [PubMed]
  23. Chodick G, Heymann AD, Rosenmann L, et al. Diabetes and risk of incident cancer: a large population-based cohort study in Israel. Cancer Causes Control 2010;21:879-87. [PubMed]
  24. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. Diabetes Care 2010;33:1674-85. [PubMed]
  25. Grote VA, Becker S, Kaaks R. Diabetes mellitus type 2 - an independent risk factor for cancer? Exp Clin Endocrinol Diabetes 2010;118:4-8. [PubMed]
  26. Schott S, Schneeweiss A, Sohn C. Breast Cancer and Diabetes Mellitus. Exp Clin Endocrinol Diabetes 2010;118:673-7. [PubMed]
  27. Peairs KS, Barone BB, Snyder CF, et al. Diabetes mellitus and breast cancer outcomes: a systematic review and meta-analysis. J Clin Oncol 2011;29:40-6. [PubMed]
  28. Chen WW, Shao YY, Shau WY, et al. The impact of diabetes mellitus on prognosis of early breast cancer in Asia. Oncologist 2012;17:485-91. [PubMed]
  29. Khandwala HM, McCutcheon IE, Flyvbjerg A, et al. The effects of insulin-like growth factors on tumorigenesis and neoplastic growth. Endocr Rev 2000;21:215-44. [PubMed]
  30. Simon D, Balkau B. Diabetes mellitus, hyperglycaemia and cancer. Diabetes Metab 2010;36:182-91. [PubMed]
  31. Chowdhury TA. Diabetes and Cancer. QJM 2010;103:905-15. [PubMed]
  32. Giaginis C, Katsamangou E, Tsourouflis G, et al. Peroxisome proliferator-activated receptor-gamma and retinoid X receptor-alpha expression in pancreatic ductal adenocarcinoma: association with clinicopathological parameters, tumor proliferative capacity, and patients’ survival. Med Sci Monit 2009;15:BR148-56. [PubMed]
  33. Bosco JL, Antonsen S, Sorensen HT, et al. Metformin and incident breast cancer among diabetic women: a population-based case-control study in Denmark. Cancer Epidemiol Biomarkers Prev 2011;20:101-11. [PubMed]
  34. Martin-Castillo B, Vazquez-Martin A, Oliveras-Ferraros C, et al. Metformin and cancer: Doses, mechanisms and the dandelion and hormetic phenomena. Cell Cycle 2010;9:1057-64. [PubMed]
  35. Cazzaniga M, Bonanni B, Guerrieri-Gonzaga A, et al. Is it time to test metformin in breast cancer clinical trials? Cancer Epidemiol Biomarkers Prev 2009;18:701-5. [PubMed]
  36. Goodwin PJ, Ligibel JA, Stambolic V. Metformin in breast cancer: time for action. J Clin Oncol 2009;27:3271-3. [PubMed]
  37. Goodwin PJ, Stambolic V. Obesity and insulin resistance in breast cancer--chemoprevention strategies with a focus on metformin. Breast 2011;20 Suppl 3:S31-5. [PubMed]
  38. Hosono K, Endo H, Takahashi H, et al. Metformin suppresses azoxymethane-induced colorectal aberrant crypt foci by activating AMP-activated protein kinase. Mol Carcinog 2010;49:662-71. [PubMed]
  39. Martin-Castillo B, Dorca J, Vazquez-Martin A, et al. Incorporating the antidiabetic drug metformin in HER2-positive breast cancer treated with neo-adjuvant chemotherapy and trastuzumab: an ongoing clinical-translational research experience at the Catalan Institute of Oncology. Ann Oncol 2010;21:187-9. [PubMed]
  40. Li D, Yeung SC, Hassan MM, et al. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009;137:482-8. [PubMed]
  41. Jiralerspong S, Palla SL, Giordano SH, et al. Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol 2009;27:3297-302. [PubMed]
  42. Guppy A, Jamal-Hanjani M, Pickering L. Anticancer effects of metformin and its potential use as a therapeutic agent for breast cancer. Future Oncol 2011;7:727-36. [PubMed]
  43. Singh S, Singh PP, Singh AG, et al. Anti-diabetic medications and risk of pancreatic cancer in patients with diabetes mellitus: a systematic review and meta-analysis. Am J Gastroenterol 2013;108:510-9. [PubMed]
  44. Li D, Yeung SC, Hassan MM, et al. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009;137:482-8. [PubMed]
  45. Dowling RJ, Zakikhani M, Fantus IG, et al. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res 2007;67:10804-12. [PubMed]
  46. Zakikhani M, Dowling R, Fantus IG, et al. Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res 2006;66:10269-73. [PubMed]
  47. Goodwin PJ, Ligibel JA, Stambolic V. Metformin in breast cancer: time for action. J Clin Oncol 2009;27:3271-3. [PubMed]
  48. Zannella VE, Dal PA, Muaddi H, et al. Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response. Clin Cancer Res 2013;19:6741-50. [PubMed]
  49. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997;3:730-7. [PubMed]
  50. Lawson JC, Blatch GL, Edkins AL. Cancer stem cells in breast cancer and metastasis. Breast Cancer Res Treat 2009;118:241-54. [PubMed]
  51. Lee HE, Kim JH, Kim YJ, et al. An increase in cancer stem cell population after primary systemic therapy is a poor prognostic factor in breast cancer. Br J Cancer 2011;104:1730-8. [PubMed]
  52. McDermott SP, Wicha MS. Targeting breast cancer stem cells. Mol Oncol 2010;4:404-19. [PubMed]
  53. Rasheed ZA, Matsui W. Biological and clinical relevance of stem cells in pancreatic adenocarcinoma. J Gastroenterol Hepatol 2012;27 Suppl 2:15-8. [PubMed]
  54. Klonisch T, Wiechec E, Hombach-Klonisch S, et al. Cancer stem cell markers in common cancers-therapeutic implications. Trends Mol Med 2008;14:450-60. [PubMed]
  55. Lee CJ, Dosch J, Simeone DM. Pancreatic cancer stem cells. J Clin Oncol 2008;26:2806-12. [PubMed]
  56. Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983-8. [PubMed]
  57. Patrawala L, Calhoun T, Schneider-Broussard R, et al. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and A. Cancer Res 2005;65:6207-19. [PubMed]
  58. Bao B, Ahmad A, Azmi AS, et al. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol 2013;Chapter 14:Unit 14.25.
  59. Ginestier C, Hur MH, Charafe-Jauffret E, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007;1:555-67. [PubMed]
  60. Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res 2007;67:1030-7. [PubMed]
  61. Hermann PC, Huber SL, Herrler T, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 2007;1:313-23. [PubMed]
  62. Hermann PC, Mueller MT, Heeschen C. Pancreatic cancer stem cells--insights and perspectives. Expert Opin Biol Ther 2009;9:1271-8. [PubMed]
  63. Bao B, Ali S, Banerjee S, et al. Curcumin Analogue CDF Inhibits Pancreatic Tumor Growth by Switching on Suppressor microRNAs and Attenuating EZH2 Expression. Cancer Res 2012;72:335-45. [PubMed]
  64. Bao B, Wang Z, Ali S, 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;5:355-64. [PubMed]
  65. Lonardo E, Cioffi M, Sancho P, et al. Metformin targets the metabolic achilles heel of human pancreatic cancer stem cells. PLoS One 2013;8:e76518. [PubMed]
  66. Menendez JA, Oliveras-Ferraros C, Cufi S, et al. Metformin is synthetically lethal with glucose withdrawal in cancer cells. Cell Cycle 2012;11:2782-92. [PubMed]
  67. Liu PP, Liao J, Tang ZJ, et al. Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ 2014;21:124-35. [PubMed]
  68. Gou S, Cui P, Li X, et al. Low concentrations of metformin selectively inhibit CD133(+) cell proliferation in pancreatic cancer and have anticancer action. PLoS One 2013;8:e63969. [PubMed]
  69. Saito T, Chiba T, Yuki K, et al. Metformin, a diabetes drug, eliminates tumor-initiating hepatocellular carcinoma cells. PLoS One 2013;8:e70010. [PubMed]
  70. Feng YH, Wu CL, Shiau AL, et al. MicroRNA-21-mediated regulation of Sprouty2 protein expression enhances the cytotoxic effect of 5-fluorouracil and metformin in colon cancer cells. Int J Mol Med 2012;29:920-6. [PubMed]
  71. Chen H, Liu X, Chen H, et al. Role of SIRT1 and AMPK in mesenchymal stem cells differentiation. Ageing Res Rev 2014;13:55-64. [PubMed]
  72. Corominas-Faja B, Cufi S, Oliveras-Ferraros C, et al. Nuclear reprogramming of luminal-like breast cancer cells generates Sox2-overexpressing cancer stem-like cellular states harboring transcriptional activation of the mTOR pathway. Cell Cycle 2013;12:3109-24. [PubMed]
  73. Garzon R, Pichiorri F, Palumbo T, et al. MicroRNA gene expression during retinoic acid-induced differentiation of human acute promyelocytic leukemia. Oncogene 2007;26:4148-57. [PubMed]
  74. Liu C, Tang DG. MicroRNA regulation of cancer stem cells. Cancer Res 2011;71:5950-4. [PubMed]
  75. DeSano JT, Xu L. MicroRNA regulation of cancer stem cells and therapeutic implications. AAPS J 2009;11:682-92. [PubMed]
  76. Perera RJ, Ray A. MicroRNAs in the search for understanding human diseases. BioDrugs 2007;21:97-104. [PubMed]
  77. Pang Y, Young CY, Yuan H. MicroRNAs and prostate cancer. Acta Biochim Biophys Sin (Shanghai) 2010;42:363-9. [PubMed]
  78. Rachagani S, Kumar S, Batra SK. MicroRNA in pancreatic cancer: pathological, diagnostic and therapeutic implications. Cancer Lett 2010;292:8-16. [PubMed]
  79. Kurisetty VV, Lakshmanaswamy R, Damodaran C. Pathogenic and therapeutic role of miRNAs in breast cancer. Front Biosci (Landmark Ed) 2014;19:1-11. [PubMed]
  80. Serpico D, Molino L, Di Cosimo S. microRNAs in breast cancer development and treatment. Cancer Treat Rev 2014;40:595-604. [PubMed]
  81. D’Ippolito E, Iorio MV. MicroRNAs and triple negative breast cancer. Int J Mol Sci 2013;14:22202-20. [PubMed]
  82. Kong D, Banerjee S, Ahmad A, et al. Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PLoS One 2010;5:e12445. [PubMed]
  83. Chang CJ, Hsu CC, Chang CH, et al. Let-7d functions as novel regulator of epithelial-mesenchymal transition and chemoresistant property in oral cancer. Oncol Rep 2011;26:1003-10. [PubMed]
  84. McCarty MF. Metformin may antagonize Lin28 and/or Lin28B activity, thereby boosting let-7 levels and antagonizing cancer progression. Med Hypotheses 2012;78:262-9. [PubMed]
  85. Li Y, VandenBoom TG, Kong D, et al. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res 2009;69:6704-12. [PubMed]
  86. Peter ME. Let-7 and miR-200 microRNAs: guardians against pluripotency and cancer progression. Cell Cycle 2009;8:843-52. [PubMed]
  87. Golestaneh AF, Atashi A, Langroudi L, et al. miRNAs expressed differently in cancer stem cells and cancer cells of human gastric cancer cell line MKN-45. Cell Biochem Funct 2012;30:411-8. [PubMed]
  88. Yu F, Yao H, Zhu P, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 2007;131:1109-23. [PubMed]
  89. Zhong X, Li N, Liang S, et al. Identification of microRNAs regulating reprogramming factor LIN28 in embryonic stem cells and cancer cells. J Biol Chem 2010;285:41961-71. [PubMed]
  90. De Vito C, Riggi N, Suvà ML, et al. Let-7a is a direct EWS-FLI-1 target implicated in Ewing’s sarcoma development. PLoS One 2011;6:e23592. [PubMed]
  91. Hua S, Xiaotao X, Renhua G, et al. Reduced miR-31 and let-7 maintain the balance between differentiation and quiescence in lung cancer stem-like side population cells. Biomed Pharmacother 2012;66:89-97. [PubMed]
  92. Huang Y. A mirror of two faces: Lin28 as a master regulator of both miRNA and mRNA. Wiley Interdiscip Rev RNA 2012;3:483-94. [PubMed]
  93. Li N, Zhong X, Lin X, et al. Lin-28 homologue A (LIN28A) promotes cell cycle progression via regulation of cyclin-dependent kinase 2 (CDK2), cyclin D1 (CCND1), and cell division cycle 25 homolog A (CDC25A) expression in cancer. J Biol Chem 2012;287:17386-97. [PubMed]
  94. Kong D, Heath E, Chen W, et al. Loss of Let-7 Up-Regulates EZH2 in Prostate Cancer Consistent with the Acquisition of Cancer Stem Cell Signatures That Are Attenuated by BR-DIM. PLoS One 2012;7:e33729. [PubMed]
  95. Chang CJ, Hung MC. The role of EZH2 in tumour progression. Br J Cancer 2012;106:243-7. [PubMed]
  96. Chou RH, Yu YL, Hung MC. The roles of EZH2 in cell lineage commitment. Am J Transl Res 2011;3:243-50. [PubMed]
  97. Crea F, Fornaro L, Bocci G, et al. EZH2 inhibition: targeting the crossroad of tumor invasion and angiogenesis. Cancer Metastasis Rev 2012;31:753-61. [PubMed]
  98. Oliveras-Ferraros C, Cufi S, Vazquez-Martin A, et al. Micro(mi)RNA expression profile of breast cancer epithelial cells treated with the anti-diabetic drug metformin: induction of the tumor suppressor miRNA let-7a and suppression of the TGFbeta-induced oncomiR miRNA-181a. Cell Cycle 2011;10:1144-51. [PubMed]
  99. Li W, Yuan Y, Huang L, et al. Metformin alters the expression profiles of microRNAs in human pancreatic cancer cells. Diabetes Res Clin Pract 2012;96:187-95. [PubMed]
  100. Dillhoff M, Liu J, Frankel W, et al. MicroRNA-21 is overexpressed in pancreatic cancer and a potential predictor of survival. J Gastrointest Surg 2008;12:2171-6. [PubMed]
  101. Moriyama T, Ohuchida K, Mizumoto K, et al. MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol Cancer Ther 2009;8:1067-74. [PubMed]
  102. Ali S, Ahmad A, Banerjee S, et al. Gemcitabine sensitivity can be induced in pancreatic cancer cells through modulation of miR-200 and miR-21 expression by curcumin or its analogue CDF. Cancer Res 2010;70:3606-17. [PubMed]
  103. Han M, Liu M, Wang Y, et al. Re-expression of miR-21 contributes to migration and invasion by inducing epithelial-mesenchymal transition consistent with cancer stem cell characteristics in MCF-7 cells. Mol Cell Biochem 2012;363:427-36. [PubMed]
  104. Nie Y, Han BM, Liu XB, et al. Identification of MicroRNAs involved in hypoxia- and serum deprivation-induced apoptosis in mesenchymal stem cells. Int J Biol Sci 2011;7:762-8. [PubMed]
  105. Yu Y, Sarkar FH, Majumdar AP. Down-regulation of miR-21 Induces Differentiation of Chemoresistant Colon Cancer Cells and Enhances Susceptibility to Therapeutic Regimens. Transl Oncol 2013;6:180-6. [PubMed]
  106. Bao B, Ahmad A, Kong D, et al. Hypoxia induced aggressiveness of prostate cancer cells is linked with deregulated expression of VEGF, IL-6 and miRNAs that are attenuated by CDF. PLoS One 2012;7:e43726. [PubMed]
  107. Bao B, Ali S, Ahmad A, et al. Hypoxia-Induced Aggressiveness of Pancreatic Cancer Cells Is Due to Increased Expression of VEGF, IL-6 and miR-21, Which Can Be Attenuated by CDF Treatment. PLoS One 2012;7:e50165. [PubMed]
  108. Kato K, Gong J, Iwama H, et al. The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol Cancer Ther 2012;11:549-60. [PubMed]
  109. Lü L, Tang D, Wang L, et al. Gambogic acid inhibits TNF-alpha-induced invasion of human prostate cancer PC3 cells in vitro through PI3K/Akt and NF-kappaB signaling pathways. Acta Pharmacol Sin 2012;33:531-41. [PubMed]
  110. Banerjee R, Mani RS, Russo N, et al. The tumor suppressor gene rap1GAP is silenced by miR-101-mediated EZH2 overexpression in invasive squamous cell carcinoma. Oncogene 2011;30:4339-49. [PubMed]
  111. Bracken AP, Pasini D, Capra M, et al. EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer. EMBO J 2003;22:5323-35. [PubMed]
  112. Chen Y, Xie D, Yin LW, et al. RNAi targeting EZH2 inhibits tumor growth and liver metastasis of pancreatic cancer in vivo. Cancer Lett 2010;297:109-16. [PubMed]
  113. Friedman JM, Liang G, Liu CC, et al. The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2. Cancer Res 2009;69:2623-9. [PubMed]
  114. Fujii S, Ito K, Ito Y, et al. Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J Biol Chem 2008;283:17324-32. [PubMed]
  115. Lu J, He ML, Wang L, et al. MiR-26a Inhibits Cell Growth and Tumorigenesis of Nasopharyngeal Carcinoma through Repression of EZH2. Cancer Res 2011;71:225-33. [PubMed]
  116. Avci CB, Harman E, Dodurga Y, et al. Therapeutic Potential of an Anti-diabetic Drug, Metformin: Alteration of miRNA expression in Prostate Cancer Cells. Asian Pac J Cancer Prev 2013;14:765-8. [PubMed]
  117. Xu X, Yang X, Ru G, et al. miR-146a gene polymorphism rs2910164 and the risk of digestive tumors: A meta-analysis of 21 case-control studies. Oncol Rep 2014;31:472-9. [PubMed]
  118. Li Y, VandenBoom TG, Wang Z, et al. miR-146a suppresses invasion of pancreatic cancer cells. Cancer Res 2010;70:1486-95. [PubMed]
  119. Rong M, He R, Dang Y, et al. Expression and clinicopathological significance of miR-146a in hepatocellular carcinoma tissues. Ups J Med Sci 2014;119:19-24. [PubMed]
  120. Ali S, Banerjee S, Logna F, et al. Inactivation of Ink4a/Arf leads to deregulated expression of miRNAs in K-Ras transgenic mouse model of pancreatic cancer. J Cell Physiol 2012;227:3373-80. [PubMed]
  121. Mei J, Bachoo R, Zhang CL. MicroRNA-146a inhibits glioma development by targeting Notch1. Mol Cell Biol 2011;31:3584-92. [PubMed]
  122. Vimalraj S, Miranda PJ, Ramyakrishna B, et al. Regulation of breast cancer and bone metastasis by microRNAs. Dis Markers 2013;35:369-87. [PubMed]
  123. Bhaumik D, Scott GK, Schokrpur S, et al. Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene 2008;27:5643-7. [PubMed]
  124. Liu M, Sakamaki T, Casimiro MC, et al. The canonical NF-kappaB pathway governs mammary tumorigenesis in transgenic mice and tumor stem cell expansion. Cancer Res 2010;70:10464-73. [PubMed]
  125. Sha M, Ye J, Zhang LX, et al. Celastrol induces apoptosis of gastric cancer cells by miR-146a inhibition of NF-kappaB activity. Cancer Cell Int 2013;13:50. [PubMed]
  126. Zhao JL, Rao DS, O’Connell RM, et al. MicroRNA-146a acts as a guardian of the quality and longevity of hematopoietic stem cells in mice. Elife 2013;2:e00537. [PubMed]
  127. Nam EJ, Lee M, Yim GW, et al. MicroRNA profiling of a CD133(+) spheroid-forming subpopulation of the OVCAR3 human ovarian cancer cell line. BMC Med Genomics 2012;5:18. [PubMed]
  128. Brabletz S, Brabletz T. The ZEB/miR-200 feedback loop--a motor of cellular plasticity in development and cancer? EMBO Rep 2010;11:670-7. [PubMed]
  129. Kong D, Li Y, Wang Z, et al. miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells 2009;27:1712-21. [PubMed]
  130. Kent OA, Mullendore M, Wentzel EA, et al. A resource for analysis of microRNA expression and function in pancreatic ductal adenocarcinoma cells. Cancer Biol Ther 2009;8:2013-24. [PubMed]
  131. Bao B, Wang Z, Ali S, et al. Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett 2011;307:26-36. [PubMed]
  132. Iliopoulos D, Lindahl-Allen M, Polytarchou C, et al. Loss of miR-200 inhibition of Suz12 leads to polycomb-mediated repression required for the formation and maintenance of cancer stem cells. Mol Cell 2010;39:761-72. [PubMed]
  133. Leal JA, Lleonart ME. MicroRNAs and cancer stem cells: Therapeutic approaches and future perspectives. Cancer Lett 2013;338:174-83. [PubMed]
  134. Shimono Y, Zabala M, Cho RW, et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 2009;138:592-603. [PubMed]
  135. Cufí S, Vazquez-Martin A, Oliveras-Ferraros C, et al. Metformin lowers the threshold for stress-induced senescence: a role for the microRNA-200 family and miR-205. Cell Cycle 2012;11:1235-46. [PubMed]
  136. Pan X, Wang R, Wang ZX. The potential role of miR-451 in cancer diagnosis, prognosis, and therapy. Mol Cancer Ther 2013;12:1153-62. [PubMed]
  137. Redova M, Poprach A, Nekvindova J, et al. Circulating miR-378 and miR-451 in serum are potential biomarkers for renal cell carcinoma. J Transl Med 2012;10:55. [PubMed]
  138. Solomides CC, Evans BJ, Navenot JM, et al. MicroRNA profiling in lung cancer reveals new molecular markers for diagnosis. Acta Cytol 2012;56:645-54. [PubMed]
  139. Xie P, Xu F, Cheng W, et al. Infiltration related miRNAs in bladder urothelial carcinoma. J Huazhong Univ Sci Technolog Med Sci 2012;32:576-80. [PubMed]
  140. Tian Y, Nan Y, Han L, et al. MicroRNA miR-451 downregulates the PI3K/AKT pathway through CAB39 in human glioma. Int J Oncol 2012;40:1105-12. [PubMed]
  141. Godlewski J, Bronisz A, Nowicki MO, et al. microRNA-451: A conditional switch controlling glioma cell proliferation and migration. Cell Cycle 2010;9:2742-8. [PubMed]
  142. Godlewski J, Nowicki MO, Bronisz A, et al. MicroRNA-451 regulates LKB1/AMPK signaling and allows adaptation to metabolic stress in glioma cells. Mol Cell 2010;37:620-32. [PubMed]
  143. Ng EK, Li R, Shin VY, et al. Circulating microRNAs as specific biomarkers for breast cancer detection. PLoS One 2013;8:e53141. [PubMed]
  144. Bian HB, Pan X, Yang JS, et al. Upregulation of microRNA-451 increases cisplatin sensitivity of non-small cell lung cancer cell line (A549). J Exp Clin Cancer Res 2011;30:20. [PubMed]
  145. Wang T, Zang WQ, Li M, et al. Effect of miR-451 on the biological behavior of the esophageal carcinoma cell line EC9706. Dig Dis Sci 2013;58:706-14. [PubMed]
  146. Bitarte N, Bandres E, Boni V, et al. MicroRNA-451 is involved in the self-renewal, tumorigenicity, and chemoresistance of colorectal cancer stem cells. Stem Cells 2011;29:1661-71. [PubMed]
  147. Kim Y, Roh S, Lawler S, et al. miR451 and AMPK mutual antagonism in glioma cell migration and proliferation: a mathematical model. PLoS One 2011;6:e28293. [PubMed]
  148. Kim Y. Regulation of cell proliferation and migration in glioblastoma: new therapeutic approach. Front Oncol 2013;3:53. [PubMed]
Cite this article as: Bao B, Azmi AS, Ali S, Zaiem F, Sarkar FH. Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. Ann Transl Med 2014;2(6):59. doi: 10.3978/j.issn.2305-5839.2014.06.05

Download Citation