MicroRNAs in the diagnosis and prognosis of breast cancer and their therapeutic potential (Review)

  • Authors:
    • Kaiyuan Zhang
    • Yanlei Zhang
    • Chang Liu
    • Ying Xiong
    • Jiqiang Zhang
  • View Affiliations

  • Published online on: June 10, 2014     https://doi.org/10.3892/ijo.2014.2487
  • Pages: 950-958
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are non-coding single-stranded RNAs in eukaryotes and are involved in the regulation of the post-transcriptional expression of specific genes. Studies have demonstrated that miRNAs play important roles in regul­ating diverse physiological events such as cell proliferation, differentiation and embryo development. In recent decades, considerable attention has been given to the relationship between miRNA and the pathology of cancers, particularly breast cancer. A large number of miRNAs have been shown to be involved in the pathophysiology of breast cancer. Studies have revealed that some miRNAs might regulate the oncogenesis and growth of breast cancer by acting on breast tumor-initiating cells or other downstream targets. Studies have also demonstrated that some miRNAs act as suppressors of metastasis or promoters of breast cancer. Additionally, certain miRNAs are involved in cancer tissue angiogenesis (one of the most important mechanisms of tumor growth and metastasis). Clinical evidence indicates that some miRNAs can be used as diagnostic and prognostic biomarkers for breast cancer due to their significantly increased or decreased expression in cancer tissue. Moreover, certain miRNAs may have therapeutic potential for targeting ER-α/HER, breast tumor-initiating cells and metastasis as well as multidrug resistance. In this review, we discuss the relationship between miRNAs and the pathogenesis of breast cancer as well as the progress of current research on the miRNA-specific diagnosis, prognosis and treatment of breast cancer.

1. Introduction

Breast cancer is one of the most common malignant cancers in women, and it has become a worldwide public health issue, with reports of more than 500,000 deaths per year (1). The pathological progression of breast cancer is multistep and complicated, consisting of oncogenesis, which is primarily due to the self-renewal and differentiation of breast tumor-initiating cells (BT-ICs), tumor growth (proliferation and apoptosis), invasion, metastasis, angiogenesis and possible post-treatment relapse (Fig. 1). As breast tissue is steroid responsive, extensive effort has been directed at identifying steroids and their receptors as well as the nuclear receptor coactivators (2). Endocrine therapy using the estrogen receptor antagonist tamoxifen and aromatase inhibitors for the treatment of breast cancer has been clinically applied. However, many problems, such as drug resistance and the adverse effects of these drugs, still exist. Therefore, novel and effective therapeutic methods for breast cancer are urgently needed.

MicroRNAs (miRNAs) are non-coding single-stranded RNAs with a length of approximately 22 nucleotides, and they function as key post-transcriptional regulators of eukaryotic gene expression through inhibiting translation or targeting mRNAs for degradation (3). Recent studies have uncovered the important roles of miRNAs in diverse physiological and pathological events such as cell proliferation, differentiation, embryo development and cancer progression (4). Accumulating evidence indicates that the putative functions of miRNAs might have important clinical significance. For example, they might be regarded as tumor suppressors and/or promoters (5,6), and their abnormal expression is highly correlated with the progression and pathology of breast cancer (7,8), supporting their diagnostic, prognostic and therapeutic potentials in breast cancer (4).

2. miRNAs in the pathogenesis and progression of breast cancer

miRNAs and breast tumor-initiating cells

Studies have shown that breast tumor-initiating cells (BT-ICs) are the key contributors to the oncogenesis of breast cancer; thus, understanding the regulation of BT-ICs is of great importance. As BT-ICs have gained mutations causing them to be more tumorigenic, they possess stem-like properties, such as self-renewal, unlimited replication potential, differentiation and metastasis.

At present, approximately 40 miRNAs have been found to be differentially expressed between human BT-ICs and non-tumorigenic breast cancer cells. Some of them, such as Let-7 (9), miR-16 (10), miR-34c (11), miR-200c (12), miR-183 and miR-203 (13), have been shown to promote the self-renewal of BT-ICs by targeting different signaling pathways. Let-7 miRNAs are members of the heterochronic pathway, and they are necessary for BT-ICs to undergo the correct progression of stage-specific events at the correct time, including differentiation (9). In contrast, other miRNAs such as miR-30 have been shown to inhibit BT-IC self-renewal by reducing Ubc9 and inducing apoptosis through silencing ITGB3 (14). Additionally, three genomic clusters, miR-183-96-182, miR-200b-200a-429 and miR-200c-141, are downregulated in BT-ICs, suggesting that they may play key roles in the regulation of self-renewal (12). Moreover, miR-145 has been reported to inhibit the differentiation of BT-ICs via the 3′-UTR of insulin receptor substrate-1 (IRS-1) (15).

Furthermore, miRNAs might also participate in the metastasis of breast cancers mediated by BT-ICs. The miR-200 family has been shown to play pivotal roles in the process of the epithelial-mesenchymal transition (EMT), which is the initial event of BT-IC-associated metastasis. Two pathways, the p53-miR-200c pathway (16) and the ZEB1-miR-200c-BMI1 pathway (17), regulate both EMT and BT-IC-associated metastasis. Furthermore, the downregulation of miR-34c (11) and the overexpression of the miR-106b-25 cluster (18) are also sufficient to induce EMT. Recently, miR-30a (19) and miR-30c (20), targeting the cytoskeleton network genes encoding twinfilin 1 (TWF1) and vimentin (VIM), have also been shown to be involved in EMT.

miRNAs and breast cancer oncogenesis

Studies have revealed that miRNAs associated with genomic changes can act as either breast cancer oncogenes (oncomiRs) or suppressors (5). The oncomiRs are upregulated in breast cancer and may hinder the expression of many genes related to tumor suppression, cell cycle regulation, apoptosis and differentiation. For example, the overexpression of miR-21 in breast cancers influences several targets, including the gene expression of tumor suppressor tropomyosin 1 (TPM1) (21), programmed cell death 4 (PDCD4) (22), phosphatase and tensin homolog (PTEN) (23) and TIMP metallopeptidase inhibitor 3 (TIMP3) (24), in breast cancer. Additionally, miR-27a, miR-96 and miR-182 can target the 3′-UTR of the mRNA that encodes the putative tumor suppressor transcription factor FOXO1 (25). Another miRNA, miR-155, has been reported to act as an oncomiR, targeting caspase-3 as a potent suppressor of apoptosis (26).

Compared to the oncomiRs, cancer-suppressing miRNAs have been reported to target and inhibit oncogenes, and their dysfunction may lead to the development of cancerous cells (27). Both in vitro (from MCF-7 cells) and in vivo (from analysis of cyclin D1-transgenic mice) analyses have shown that the miR17-5p (miR-91)/miR-20a cluster can inhibit breast cancer cell proliferation by suppressing the proliferative effect of cyclin D1 (28). miR-27b has been reported to play a suppressive role in breast cancer cells and to post-transcriptionally regulate cytochrome P4501B1 (CYP1B1), a key enzyme in the metabolism of 17β-estradiol, which promotes the growth of breast cancer (29). Recently, HER2 and HER3 (erbB2 and erbB3), which significantly correlate with breast cancer and poor prognosis, have been shown to be suppressed by miR-125a, miR-125b and miR-205 in breast cancer cells, leading to a reduction in cell proliferation and migration and increased apoptosis (30). Moreover, the suppressive role of miR-205 is mediated through the direct targeting of oncogenes such as Zeb1 (31). It has been shown that the overexpression of miR-145 inhibits estrogen receptor-α (ER-α) protein expression (32) and reduces RTKN protein expression (33), thus reducing breast cancer cell growth and inducing apoptosis, indicating its tumor-suppressive functions. Similarly, miR-22 (34), miR-206 (35) and miR-375 (36) negatively affect the expression of ER-α.

miRNAs and breast cancer growth

Studies have recently uncovered the roles of miRNAs in cancer cell proliferation and apoptosis, the dysregulation of which is the most important mechanism of cancer growth. For example, oncomiRs, including miR-21 (24), miR-27a, miR-96, miR-182 (25) and miR-155 (37), can target the 3′-UTR of mRNAs that encode tumor suppressor genes. Moreover, cell growth suppression is achieved by the overexpression of cancer suppressor miRNAs such as miR17-5p (miR-91)/miR-20a (28), miR-27b (29), miR-125a, miR-125b and miR-205 (30), suggesting their roles in growth suppression. Furthermore, the upregulation of ER-α accounts for the abnormal cell proliferation observed in approximately two-thirds of breast cancer cases. miR-22 (34), miR-145 (32), miR-206 (35) and miR-375 (36) are reported to negatively regulate ER-α expression, thus inhibiting breast cancer cell proliferation.

Several studies have addressed the roles that miRNAs play in the modulation of apoptosis in breast cancer cells. The overexpression of miR-145 has been demonstrated to induce apoptosis by directly binding to the 3′-UTR of RTKN (33). miR-26b can also impair the viability and trigger the apoptosis of human MCF-7 cells by directly targeting SLC7A11 (38). Moreover, miR-290 was shown to target Arid4b while simultaneously enhancing ER signaling and inducing apoptosis, thereby suppressing breast cancer growth (46). The overexpression of miR-335 decreases cell viability and increases apoptosis by simultaneously regulating the known BRCA1 activators ER-α, IGF1R and SP1 and the repressor ID4 (39).

miRNAs and breast cancer invasion and metastasis

Invasion and metastasis are the major factors underlying the poor prognosis or even death of breast cancer patients. In the past few years, many types of miRNAs have been identified, and their roles have been explored in cancer invasion and metastasis; some of these miRNAs function as metastasis promoters, whereas others function as suppressors (Fig. 2).

miRNAs as suppressors of metastasis

miR-23b has been shown to be a metastatic suppressor miRNA that directly inhibits a number of genes implicated in cytoskeletal remodeling, focal adhesion, cell spreading, cell-cell junction formation and the formation of lamellipodia in breast cancer cells (40). miR-31 has also been shown to trigger metastatic regression in the lungs by eliciting cell cycle arrest and apoptosis, which can be explained by the miR-31-mediated suppression of integrin-α5, radixin, RhoA (41), the actin cytoskeleton remodeling protein WAVE3 (42) and protein kinase C epsilon (PKC-ɛ) (43). Other cancer-suppressing miRNAs such as miR-125a, miR-125b and miR-205 significantly inhibit breast cancer cell invasion by directly targeting HER2 and HER3 (30), whereas miR-205 can target Zeb1 (31). Additionally, miR-145 significantly suppresses cell invasion and lung metastasis by directly targeting the metastasis gene mucin 1 (MUC1), leading to a reduction of β-catenin and adherin 11 (44). Moreover, miR-146 may be involved in the inhibition of the breast cancer metastasis suppressor 1 (BRMS1), a predominant nuclear protein, leading to metastasis suppression (45); miR-146a/b functions to negatively regulate NF-κB activity through targeting IL-1 receptor-associated kinase (IL-1-RSK) and TNF receptor-associated factor 6 (TNFRSF-6), thus functioning as a suppressor of breast cancer metastasis (46).

The initiation of metastasis, EMT, has been shown to be significantly regulated by the miR-200 family (miR-200a, miR-200b, miR-200c, miR-141 and miR-429), the members of which are all dramatically downregulated in breast cancer cells that have undergone the EMT induced by tumor growth factor-β (TGF-β), and the E-cadherin transcriptional repressors ZEB1 and ZEB2 have been established as their targets (47). In addition, the miR-200 family also enhances metastatic colonization, partially through directly targeting Sec23a, which is involved in the secretion of metastasis suppressor proteins (48). Furthermore, the miR-200b/c/429 cluster is a stronger regulator of EGF-driven invasion than the miR-200a/141 cluster, and phospholipase C gamma 1 (PLCG1) might be a potential candidate contributing to these differences (49). Moreover, the overexpression of miR-200a has been reported to decrease anchorage-independent growth and migration in breast cancer, targeting the oncogene silent information regulator 1 (SIRT1) (50); miR-200c greatly impacts the regulation of several EMT-related processes in breast cancer by directly targeting the actin cytoskeleton (51).

Studies have reported that in highly metastatic breast cancer cells, the expression of miR-203 is significantly downregulated and that its upregulation inhibits tumor cell invasion and metastatic colonization through the SNAI2 and miR-203 regulatory loop (52). Moreover, miR-335 has been identified as the first selective metastasis suppressor in human breast cancer by targeting SOX4 and tenascin-C (53). Additionally, miR-206 suppresses the invasion and migration of MDA-MB-231 cells in vitro, partially via regulating the remodeling of the actin cytoskeleton, with CDC42 as its potential target (54). Another study has shown that the overexpression of miR-224 inhibits cell division cycle 42 (CDC42) and chemokine receptor 4 (CXCR4), accounting for the inhibition of Ubc9-mediated invasion (55). Furthermore, let-7b and the miR-17-5p/miR-20a cluster are significantly decreased in lymph node metastases of breast cancer cells. The forced expression of let-7b significantly inhibits breast cancer cell migration by targeting four genes that are correlated with the actin cytoskeleton pathway, including PAK1, DIAPH2, RDX and ITGB8 (56). The miR-17-5p/miR-20a cluster plays a suppressive role in metastatic breast cancer through heterotypic secreted signaling (57).

miRNAs as metastasis promoters

miR-9 is upregulated in breast cancer and increases cell motility and invasiveness by directly targeting CDH1 and E-cadherin-encoding mRNAs (58). miR-10b expression is increased in metastatic breast cancer cells, leading to cell migration, invasion and metastasis through indirectly activating the prometastatic gene RHOC by suppressing the homeobox D10 (HOXD10) tumor suppressor signaling pathway (59). The regulation of T lymphoma invasion and metastasis 1 (Tiam1)-mediated Rac activation in breast cancer cells is a novel target for miR-10b to regulate the invasion and migration of breast cancer cells (60). miR-21, as an oncogenic miRNA, plays important roles in breast cancer invasion and metastasis by inhibiting multiple metastasis suppressor genes. The upregulated expression of miR-181a in metastatic breast cancer cells has been shown to enhance breast cancer metastasis by promoting EMT and migratory and invasive phenotypes, targeting the proapoptotic molecule Bim, which is involved in metastatic cancer cell anoikis (61). Similarly, miR-182 promotes breast cancer metastasis by suppressing the missing in metastasis (MIM), which activates the Ras homolog family member A (RhoA) (62). miR-373 and miR-520c can also promote the invasion and migration of breast cancer cells both in vitro and in vivo (63) by directly targeting and inhibiting the expression of RELA, TGFBR2 (63) and CD44 (64).

miRNAs and breast cancer angiogenesis

Angiogenesis is one of the most important mechanisms of tumor growth and metastasis (Fig. 1). Much effort has been directed toward the development of anti-angiogenic tumor therapies. miRNAs appear to have considerable potential as the gene resource for the gene therapy of breast cancer. Endothelial cells (ECs) are one of the key components of the tunica intima, playing vital roles in tumor angiogenesis. It has been shown that many miRNAs such as miR-216a, miR-330 and miR-608, which can bind to both the CD44 and CDC42 (CD44 downstream target mRNA) 3′-UTRs, can modulate EC activities (3). miR-126, which is particularly expressed in vascular endothelial cells, is reported to play a vital role in suppressing breast cancer angiogenesis through regulating the VEGF/PI3K/AKT signaling cascade (65) and targeting IGFBP2, PITPNC1 and MERTK (66).

Moreover, miR-9 contributes to angiogenesis through the upregulation of VEGF expression in breast cancer by directly targeting CDH1 (58). Another report has demonstrated that VEGF expression in breast cancer cells is mediated by hypoxia inducible factor 1 (HIF-1) and signal transducer and activator of transcription 3 (STAT3) in a miR-20b-dependent manner (67). The overexpression of miR-10b and miR-196b has been detected in high-grade breast cancer vasculature and, notably, to be responsive to VEGF stimulation (68). Moreover, miR-27a suppresses breast cancer angiogenesis by inhibiting specificity protein (Sp)-dependent angiogenic gene expression, including that of survivin, VEGF and VEGFR1, as it targets the zinc finger ZBTB10 gene (69). The overexpression of miR-19 downregulates tissue factor (TF) expression, which is an important factor in the regulation of tumor angiogenesis, suggesting the potential of miR-19 for regulating breast cancer angiogenesis (70).

Taken together, the involvement of these miRNAs in the pathogenesis and progression of breast cancer has been elucidated, with some of them acting as suppressors and others as tumor promoters, as summarized in Fig. 2. These studies are crucial to evaluating their use as clinical biomarkers and their therapeutic potential.

3. Clinical implications of miRNAs in breast cancer

Many miRNAs have been shown to contribute to the pathogenesis and progression of breast cancer, which supports the potential of miRNA-specific strategies for the treatment of breast cancer (Fig. 3).

Diagnostic and prognostic biomarkers of breast cancer

The increasing number of reports indicating the significance of miRNAs in the pathogenesis and progression of breast cancer has revealed the potential relevance of aberrantly expressed miRNAs as biomarkers for the detection, diagnosis, classification and therapy of breast cancer. First, the expression profile of miRNAs can be used to distinguish breast cancer tissue from normal tissue. For example, the significant overexpression of miR-21, miR-106a and miR-155 and the decreased expression of miR-126, miR-199a and miR-335 have been reported in breast cancer tissue when compared with normal tissue (71). A study by Iyevleva et al has reported higher levels of miR-21, miR-10b and miR-31 in bilateral patients than in unilateral patients (72). Additionally, a number of miRNAs are differentially expressed in the luminal A, luminal B, basal-like, HER2+ and normal-like breast cancer subtypes. For example, the upregulation of miR-17-92 clusters has been shown to account for the great distinction of triple-negative breast cancer from other subtypes (6).

The miRNA expression profile has also been correlated with pathological features such as tumor stage, proliferation index, lymph node metastasis, resistance to breast cancer treatment and poor survival, suggesting their prognostic potential. The most promising prognostic miRNAs for breast cancer are miR-21 (24), miR-181a (61) and the miR-221/miR-222 cluster (73) because their overexpression has been correlated with advanced tumor stage, lymph node metastasis and poor patient survival. Moreover, a high level of the miR-106b-25 cluster in breast cancer can significantly predict a shortened time to relapse (18). Additionally, in ER-positive cases, high miR-767-3p, miR-128a and/or miR-769-3p expression is associated with a poor prognosis, whereas high miR-135a expression is associated with a good prognosis; in contrast, in ER-negative cases, high levels of miR-27b, miR-144 and/or miR-210 are associated with a poor prognosis, and high miR-342, miR-150 and/or miR-30c expression is associated with a good prognosis (74). Moreover, miR-342, miR-27b and miR-150 are prognostic in triple receptor-negative breast cancers (74).

Therapeutic miRNAs

The traditional therapies for breast cancer, such as surgery, chemotherapy and/or radiotherapy, appear to be powerful; however, regardless of the level of damage directly caused to the human body, problems such as drug resistance and adverse effects still exist, making the treatment of breast cancer more difficult than expected. Therefore, novel, feasible and effective therapeutic methods for breast cancer are urgently needed. Because of the distinct roles that miRNAs play in the pathology of breast cancer, many studies have investigated their therapeutic potential in its treatment.

Anti-BT-ICs

One of the greatest challenges for breast cancer research is relapses that occur in patients undergoing chemotherapy and radiotherapy. Recent studies have shown that targeting BT-ICs is promising to prevent relapse and provides a new strategy for breast cancer prevention. Many types of miRNAs are involved in regulating the self-renewal and differentiation of BT-IC as well as BT-IC-induced relapse (Fig. 2). As mentioned above, one attractive candidate for this purpose is let-7 because the administration of let-7 has been found to be effective in mouse models of breast cancer (75).

Anti-ER-α/HER

As breast cancer is steroid-responsive, much attention has been given to steroids (e.g., estradiol and progesterone) and their receptors [e.g., ER-α, ER-β and the progesterone receptor (PR)] as well as the nuclear receptor coactivators, particularly steroid receptor coactivator-1 (SRC-1) and SRC-3 (2). It has been shown that ER-α upregulation is responsible for the abnormal cell proliferation observed in approximately two-thirds of breast cancer cases, and anti-ER-α therapy may be of great value for breast cancer treatments. Studies have revealed that the levels of miR-22 (34), miR-145 (32), miR-206 (35) and miR-375 (36) are significantly decreased in ER-α-positive human breast cancer tissues and inversely correlated with ER-α mRNA expression in breast cancer tissues; thus, they have been suggested as novel therapeutic agents for anti-multidrug resistance (MDR) therapies that target only ER-α in breast cancer. Additionally, the overexpression of HER2 and HER3 (erbB2 and erbB3) is significantly correlated with breast cancer grade and poor prognosis, and this overexpression can be suppressed by miR-125a, miR-125b and miR-205 (30), thus resulting in the abrogation of HER2- and HER3-mediated resistance and restoring potent proapoptotic activity.

Anti-metastasis

miR-21 plays a role in invasion and metastasis by targeting multiple tumor/metastasis suppressor genes such as PDCD4 (22), PTEN (23) and TIMP3 (24), suggesting its potential for anti-metastasis treatments. miR-31 has also been shown to regulate a number of metastasis-related genes in breast cancer cells and tissues, and its expression is inversely correlated with the cell’s ability to invade and metastasize. Activation of miR-31 in established metastases elicits metastatic regression and prolongs patient survival (41). The overexpression of miR-145 has been shown to suppress breast cancer cell growth and motility, and a treatment combining Ad-miR-145 with 5-FU has shown significant antitumor effects (76). miR-146 is also involved in suppressing metastasis, and miR-146a/b-expressing cells show markedly impaired invasion and migration capacities when compared with controls (45).

Anti-multidrug resistance (MDR)

Drug resistance and multidrug resistance (MDR) are the main causes of treatment failure, and these are the most challenging problems in the treatment of breast cancer. Studies have demonstrated that some miRNAs might be involved in the occurrence of MDR, and therefore, targeting these miRNAs might be of great significance regarding the development of novel strategies against breast cancer. For example, the expression of miR-155 (77) and miR-663 (78) has been demonstrated to induce breast cancer cell survival and MDR, whereas their knockdown renders breast cancer cells susceptible to apoptosis and enhances their chemosensitivity. miR-326 is downregulated in advanced breast cancer tissues and is inversely correlated with the expression of multidrug resistance-associated protein-1 (MRP-1); its upregulation in VP-16-resistant MCF-7 cell lines can downregulate the expression of MRP-1, making them sensitive to VP-16 and doxorubicin (DOX) (79). These findings indicate that miR-326 may be an efficient and powerful agent for preventing and reversing MDR in breast cancer cells. Moreover, the miR-328-mediated downregulation of ABCG2 (a molecular determinant of the pharmacokinetic properties of many drugs in humans) in MCF-7/MX100 cells may result in increased mitoxantrone sensitivity (80). Recently, the multidrug resistance 1 gene has been identified as a novel target of miR-451, and transfection of MCF-7/DOX-resistant cells with miR-451 results in increased sensitivity to DOX, indicating that increased miR-451 expression may have significant implications for overcoming the MDR of breast cancer cells (81).

The above results are summarized in Fig. 3 and Table I.

Table I

Selected miRNAs and their clinical applications in breast cancer.

Table I

Selected miRNAs and their clinical applications in breast cancer.

ApplicationmicroRNAPutative clinical significanceRefs.
DiagnosismiR-21↑, miR-106a↑, miR-155↑, miR-199a↓, miR-126↓, miR-335↓Potential biomarkers for breast cancer diagnosis, grading and prognosis(70)
miR-21↑, miR-10b↑, miR-31↑Biomarkers for the diagnosis bilateral or unilateral breast cancer(71)
miR-17-92 cluster↑Identifies the subtype of breast cancer: distinction of triple- negative breast cancer from other subtypes(72)
Biomarker
 PrognosismiR-21↑, miR-181a↑, miR-221/miR 222 cluster↑The overexpression is correlated with advanced tumor stage, lymph node metastasis and poor survival of the patients(6,23,60)
miR-767-3p, miR-128a, miR-769-3p, miR-135aAssociated with prognosis in ER-positive cases: poor, miR-767-3p↑, miR-128a↑, miR-769-3p↑; good, miR-135a↑(73)
miR-27b, miR-144, miR-210
miR-342, miR-150, miR-30c
Associated with prognosis in ER-negative cases: poor, miR-27b↑, miR-144↑, miR-210↑; good, miR-342↑, miR-150↑, miR-30c↑(73)
miR-27b, miR-150, miR-342Prognostic also in triple receptor-negative breast cancers(73)
miR-106b-25 cluster↑Significantly predicted shortened time to relapse(17)
miR-22↓, miR-145↓, miR-206↓ and miR-375↓Significantly decreased in ER-α-positive human breast cancer tissues and is inversely correlated with ER-α mRNA expression in breast cancer tissues in a dose-and time-dependent manner(31,3335)
 Anti-ER-α/HERmiR-125a↓, miR-125b↓
miR-205↓
Directly suppressed the HER2 and HER3 (erbB2 and erbB3)(29)
 Anti-BT-IClet-7↑Effective in suppressing metastatic breast cancer(74)
Therapy
 Anti-metastasismiR-21↓Significantly reduced invasion and lung metastasis ability(2123)
miR-31↑Elicits the regression of metastasis and prolongs the survival of patients(40)
miR-145↑Ad-miR-145 suppressed breast cancer cell motility and invasiveness; combining Ad-miR-145 with 5-FU significantly showed anti-tumor effects(2)
miR-146a/b↑Inhibited both invasion and migration, and suppressed experimental lung metastasis(44)
 Anti-MDRmiR-21↑miR-21 inhibitor gene therapy combining with taxol chemotherapy(81)
miR-24-2↓Combining with an anticancer drug such as cisplatin provides a new avenue for overcoming drug resistance(86)
miR-155↑Knockdown of it renders breast cancer cells to apoptosis and enhance chemosensitivity(76)
miR-326↑Downregulated MRP-1 expression and sensitize cancer cells to VP-16 and doxorubicin(78)
miR-328↑Increased mitoxantrone sensitivity(79)
miR-451↑Increased sensitivity of cancer cells to DOX(80)

[i] This table shows only some representative miRNAs and their pathological and clinical significance in breast cancer. MDR, multidrug resistance; ↑, increased expression; ↓, decreased expression.

4. Conclusions

In the past few years, the correlations between miRNAs and breast cancer have been widely investigated, contributing greatly to studies of breast cancer pathogenesis and their clinical implications. These small molecules play significant roles in the oncogenesis, growth, invasion, metastasis and angiogenesis of breast cancer; thus, altered expression of miRNAs can be regarded as a target for the diagnosis and/or treatment of breast cancer based on the miRNA expression profile. However, many of the remaining problems urgently require further exploration. For example, we know little about the upstream, intrinsic factors that regulate these specific miRNAs; there are tens or hundreds of miRNAs within a cell, and which is the most prominent indicator for the diagnosis and treatment as well as the prognosis of cancer remains to be elucidated. Future work should aim to identify the limited miRNAs involved in each pathological stage as well as the therapeutic targets of breast cancers. These under-explored issues may be the greatest challenges in the future.

Acknowledgements

This study was supported by the National Science Foundation of China (NSFC, No. 81171035).

References

1 

Zoon CK, Starker EQ, Wilson AM, Emmert-Buck MR, Libutti SK and Tangrea MA: Current molecular diagnostics of breast cancer and the potential incorporation of microRNA. Expert Rev Mol Diagn. 9:455–467. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Johnson AB and O’Malley BW: Steroid receptor coactivators 1, 2, and 3: critical regulators of nuclear receptor activity and steroid receptor modulator (SRM)-based cancer therapy. Mol Cell Endocrinol. 348:430–439. 2012. View Article : Google Scholar

3 

Jeyapalan Z, Deng Z, Shatseva T, Fang L, He C and Yang BB: Expression of CD44 3′-untranslated region regulates endogenous microRNA functions in tumorigenesis and angiogenesis. Nucleic Acids Res. 39:3026–3041. 2011.

4 

Piva R, Spandidos DA and Gambari R: From microRNA functions to microRNA therapeutics: novel targets and novel drugs in breast cancer research and treatment (Review). Int J Oncol. 43:985–994. 2013.PubMed/NCBI

5 

Iorio MV, Ferracin M, Liu CG, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Farazi TA, Horlings HM, Ten Hoeve JJ, et al: MicroRNA sequence and expression analysis in breast tumors by deep sequencing. Cancer Res. 71:4443–4453. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Andorfer CA, Necela BM, Thompson EA and Perez EA: MicroRNA signatures: clinical biomarkers for the diagnosis and treatment of breast cancer. Trends Mol Med. 17:313–319. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Melo SA and Esteller M: Dysregulation of microRNAs in cancer: playing with fire. FEBS Lett. 585:2087–2099. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Nimmo RA and Slack FJ: An elegant miRror: microRNAs in stem cells, developmental timing and cancer. Chromosoma. 118:405–418. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Zhang X, Wan G, Mlotshwa S, et al: Oncogenic Wip1 phosphatase is inhibited by miR-16 in the DNA damage signaling pathway. Cancer Res. 70:7176–7186. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Yu F, Jiao Y, Zhu Y, et al: MicroRNA 34c gene down-regulation via DNA methylation promotes self-renewal and epithelial-mesenchymal transition in breast tumor-initiating cells. J Biol Chem. 287:465–473. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Shimono Y, Zabala M, Cho RW, et al: Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell. 138:592–603. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Greene SB, Herschkowitz JI and Rosen JM: Small players with big roles: microRNAs as targets to inhibit breast cancer progression. Curr Drug Targets. 11:1059–1073. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Yu F, Deng H, Yao H, Liu Q, Su F and Song E: Mir-30 reduction maintains self-renewal and inhibits apoptosis in breast tumor-initiating cells. Oncogene. 29:4194–4204. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Rubin R, Arzumanyan A, Soliera AR, Ross B, Peruzzi F and Prisco M: Insulin receptor substrate (IRS)-1 regulates murine embryonic stem (mES) cells self-renewal. J Cell Physiol. 213:445–453. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Chang CJ, Chao CH, Xia W, et al: p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol. 13:317–323. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Ahmad A, Aboukameel A, Kong D, et al: Phosphoglucose isomerase/autocrine motility factor mediates epithelial-mesenchymal transition regulated by miR-200 in breast cancer cells. Cancer Res. 71:3400–3409. 2011. View Article : Google Scholar

18 

Smith AL, Iwanaga R, Drasin DJ, et al: The miR-106b-25 cluster targets Smad7, activates TGF-beta signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer. Oncogene. 31:5162–5171. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Cheng CW, Wang HW, Chang CW, et al: MicroRNA-30a inhibits cell migration and invasion by downregulating vimentin expression and is a potential prognostic marker in breast cancer. Breast Cancer Res Treat. 134:1081–1093. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Bockhorn J, Yee K, Chang YF, et al: MicroRNA-30c targets cytoskeleton genes involved in breast cancer cell invasion. Breast Cancer Res Treat. 137:373–382. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Zhu S, Si ML, Wu H and Mo YY: MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 282:14328–14336. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Li J, Zhang Y, Zhang W, et al: Genetic heterogeneity of breast cancer metastasis may be related to miR-21 regulation of TIMP-3 in translation. Int J Surg Oncol. 2013:8750782013.PubMed/NCBI

25 

Guttilla IK and White BA: Coordinate regulation of FOXO1 by miR-27a, miR-96, and miR-182 in breast cancer cells. J Biol Chem. 284:23204–23216. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Ovcharenko D, Kelnar K, Johnson C, Leng N and Brown D: Genome-scale microRNA and small interfering RNA screens identify small RNA modulators of TRAIL-induced apoptosis pathway. Cancer Res. 67:10782–10788. 2007. View Article : Google Scholar

27 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Yu Z, Wang C, Wang M, et al: A cyclin D1/microRNA 17/20 regulatory feedback loop in control of breast cancer cell proliferation. J Cell Biol. 182:509–517. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Tsuchiya Y, Nakajima M, Takagi S, Taniya T and Yokoi T: MicroRNA regulates the expression of human cytochrome P450 1B1. Cancer Res. 66:9090–9098. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Wang S, Huang J, Lyu H, et al: Functional cooperation of miR-125a, miR-125b, and miR-205 in entinostat-induced downregulation of erbB2/erbB3 and apoptosis in breast cancer cells. Cell Death Dis. 4:e5562013. View Article : Google Scholar : PubMed/NCBI

31 

Wu H and Mo YY: Targeting miR-205 in breast cancer. Expert Opin Ther Targets. 13:1439–1448. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Spizzo R, Nicoloso MS, Lupini L, et al: miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-alpha in human breast cancer cells. Cell Death Differ. 17:246–254. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Wang S, Bian C, Yang Z, et al: miR-145 inhibits breast cancer cell growth through RTKN. Int J Oncol. 34:1461–1466. 2009.PubMed/NCBI

34 

Xiong J, Yu D, Wei N, et al: An estrogen receptor alpha suppressor, microRNA-22, is downregulated in estrogen receptor alpha-positive human breast cancer cell lines and clinical samples. FEBS J. 277:1684–1694. 2010. View Article : Google Scholar

35 

Adams BD, Furneaux H and White BA: The micro-ribonucleic acid (miRNA) miR-206 targets the human estrogen receptor-alpha (ERalpha) and represses ERalpha messenger RNA and protein expression in breast cancer cell lines. Mol Endocrinol. 21:1132–1147. 2007. View Article : Google Scholar

36 

de Souza Rocha Simonini P, Breiling A, Gupta N, et al: Epigenetically deregulated microRNA-375 is involved in a positive feedback loop with estrogen receptor alpha in breast cancer cells. Cancer Res. 70:9175–9184. 2010.

37 

Rather MI, Nagashri MN, Swamy SS, Gopinath KS and Kumar A: Oncogenic microRNA-155 down-regulates tumor suppressor CDC73 and promotes oral squamous cell carcinoma cell proliferation: implications for cancer therapeutics. J Biol Chem. 288:608–618. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Liu XX, Li XJ, Zhang B, et al: MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett. 585:1363–1367. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Heyn H, Engelmann M, Schreek S, et al: MicroRNA miR-335 is crucial for the BRCA1 regulatory cascade in breast cancer development. Int J Cancer. 129:2797–2806. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Pellegrino L, Stebbing J, Braga VM, et al: miR-23b regulates cytoskeletal remodeling, motility and metastasis by directly targeting multiple transcripts. Nucleic Acids Res. 41:5400–5412. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Valastyan S, Chang A, Benaich N, Reinhardt F and Weinberg RA: Activation of miR-31 function in already-established metastases elicits metastatic regression. Genes Dev. 25:646–659. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Sossey-Alaoui K, Downs-Kelly E, Das M, Izem L, Tubbs R and Plow EF: WAVE3, an actin remodeling protein, is regulated by the metastasis suppressor microRNA, miR-31, during the invasion-metastasis cascade. Int J Cancer. 129:1331–1343. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Korner C, Keklikoglou I, Bender C, Worner A, Munstermann E and Wiemann S: MicroRNA-31 sensitizes human breast cells to apoptosis by direct targeting of protein kinase C epsilon (PKCepsilon). J Biol Chem. 288:8750–8761. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Sachdeva M and Mo YY: MicroRNA-145 suppresses cell invasion and metastasis by directly targeting mucin 1. Cancer Res. 70:378–387. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Hurst DR, Edmonds MD, Scott GK, Benz CC, Vaidya KS and Welch DR: Breast cancer metastasis suppressor 1 up-regulates miR-146, which suppresses breast cancer metastasis. Cancer Res. 69:1279–1283. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J and Benz CC: Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene. 27:5643–5647. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Gregory PA, Bert AG, Paterson EL, et al: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 10:593–601. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Korpal M, Ell BJ, Buffa FM, et al: Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nat Med. 17:1101–1108. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Uhlmann S, Zhang JD, Schwäger A, et al: miR-200bc/429 cluster targets PLCgamma1 and differentially regulates proliferation and EGF-driven invasion than miR-200a/141 in breast cancer. Oncogene. 29:4297–4306. 2010. View Article : Google Scholar

50 

Eades G, Yao Y, Yang M, Zhang Y, Chumsri S and Zhou Q: miR-200a regulates SIRT1 expression and epithelial to mesenchymal transition (EMT)-like transformation in mammary epithelial cells. J Biol Chem. 286:25992–26002. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Jurmeister S, Baumann M, Balwierz A, et al: MicroRNA-200c represses migration and invasion of breast cancer cells by targeting actin-regulatory proteins FHOD1 and PPM1F. Mol Cell Biol. 32:633–651. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Ding X, Park SI, McCauley LK and Wang CY: Signaling between transforming growth factor beta (TGF-beta) and transcription factor SNAI2 represses expression of microRNA miR-203 to promote epithelial-mesenchymal transition and tumor metastasis. J Biol Chem. 288:10241–10253. 2013. View Article : Google Scholar

53 

Tavazoie SF, Alarcón C, Oskarsson T, et al: Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 451:147–152. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Liu H, Cao YD, Ye WX and Sun YY: Effect of microRNA-206 on cytoskeleton remodelling by downregulating Cdc42 in MDA-MB-231 cells. Tumori. 96:751–755. 2010.PubMed/NCBI

55 

Zhu S, Sachdeva M, Wu F, Lu Z and Mo YY: Ubc9 promotes breast cell invasion and metastasis in a sumoylation-independent manner. Oncogene. 29:1763–1772. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Hu X, Guo J, Zheng L, et al: The heterochronic microRNA let-7 inhibits cell motility by regulating the genes in the actin cytoskeleton pathway in breast cancer. Mol Cancer Res. 11:240–250. 2013. View Article : Google Scholar

57 

Yu Z, Willmarth NE, Zhou J, et al: microRNA 17/20 inhibits cellular invasion and tumor metastasis in breast cancer by heterotypic signaling. Proc Natl Acad Sci USA. 107:8231–8236. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Ma L, Young J, Prabhala H, et al: miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 12:247–256. 2010.PubMed/NCBI

59 

Haque I, Banerjee S, Mehta S, et al: Cysteine-rich 61-connective tissue growth factor-nephroblastoma-overexpressed 5 (CCN5)/Wnt-1-induced signaling protein-2 (WISP-2) regulates microRNA-10b via hypoxia-inducible factor-1alpha-TWIST signaling networks in human breast cancer cells. J Biol Chem. 286:43475–43485. 2011. View Article : Google Scholar

60 

Moriarty CH, Pursell B and Mercurio AM: miR-10b targets Tiam1: implications for Rac activation and carcinoma migration. J Biol Chem. 285:20541–20546. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Taylor MA, Sossey-Alaoui K, Thompson CL, Danielpour D and Schiemann WP: TGF-beta upregulates miR-181a expression to promote breast cancer metastasis. J Clin Invest. 123:150–163. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Lei R, Tang J, Zhuang X, et al: Suppression of MIM by microRNA-182 activates RhoA and promotes breast cancer metastasis. Oncogene. 33:1287–1296. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Huang Q, Gumireddy K, Schrier M, et al: The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol. 10:202–210. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Yu Z, Baserga R, Chen L, Wang C, Lisanti MP and Pestell RG: microRNA, cell cycle, and human breast cancer. Am J Pathol. 176:1058–1064. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Zhu N, Zhang D, Xie H, et al: Endothelial-specific intron-derived miR-126 is down-regulated in human breast cancer and targets both VEGFA and PIK3R2. Mol Cell Biochem. 351:157–164. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Png KJ, Halberg N, Yoshida M and Tavazoie SF: A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Nature. 481:190–194. 2012. View Article : Google Scholar

67 

Cascio S, D’Andrea A, Ferla R, et al: miR-20b modulates VEGF expression by targeting HIF-1 alpha and STAT3 in MCF-7 breast cancer cells. J Cell Physiol. 224:242–249. 2010.PubMed/NCBI

68 

Plummer PN, Freeman R, Taft RJ, et al: MicroRNAs regulate tumor angiogenesis modulated by endothelial progenitor cells. Cancer Res. 73:341–352. 2013. View Article : Google Scholar : PubMed/NCBI

69 

Mertens-Talcott SU, Chintharlapalli S, Li X and Safe S: The oncogenic microRNA-27a targets genes that regulate specificity protein transcription factors and the G2-M checkpoint in MDA-MB-231 breast cancer cells. Cancer Res. 67:11001–11011. 2007. View Article : Google Scholar

70 

Zhang X, Yu H, Lou JR, et al: MicroRNA-19 (miR-19) regulates tissue factor expression in breast cancer cells. J Biol Chem. 286:1429–1435. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Wang F, Zheng Z, Guo J and Ding X: Correlation and quantitation of microRNA aberrant expression in tissues and sera from patients with breast tumor. Gynecol Oncol. 119:586–593. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Iyevleva AG, Kuligina E, Mitiushkina NV, Togo AV, Miki Y and Imyanitov EN: High level of miR-21, miR-10b, and miR-31 expression in bilateral vs. unilateral breast carcinomas. Breast Cancer Res Treat. 131:1049–1059. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Chen WX, Hu Q, Qiu MT, et al: miR-221/222: promising biomarkers for breast cancer. Tumour Biol. 34:1361–1370. 2013. View Article : Google Scholar

74 

Buffa FM, Camps C, Winchester L, et al: microRNA-associated progression pathways and potential therapeutic targets identified by integrated mRNA and microRNA expression profiling in breast cancer. Cancer Res. 71:5635–5645. 2011. View Article : Google Scholar

75 

Barh D, Malhotra R, Ravi B and Sindhurani P: MicroRNA let-7: an emerging next-generation cancer therapeutic. Curr Oncol. 17:70–80. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Kim SJ, Oh JS, Shin JY, et al: Development of microRNA-145 for therapeutic application in breast cancer. J Control Release. 155:427–434. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Kong W, He L, Coppola M, et al: MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J Biol Chem. 285:17869–17879. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Hu H, Li S, Cui X, et al: The overexpression of hypomethylated miR-663 induces chemotherapy resistance in human breast cancer cells by targeting heparin sulfate proteoglycan 2 (HSPG2). J Biol Chem. 288:10973–10985. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Liang Z, Wu H, Xia J, et al: Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem Pharmacol. 79:817–824. 2010. View Article : Google Scholar

80 

Pan YZ, Morris ME and Yu AM: MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol Pharmacol. 75:1374–1379. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Kovalchuk O, Filkowski J, Meservy J, et al: Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol Cancer Ther. 7:2152–2159. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 45 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang K, Zhang Y, Liu C, Xiong Y and Zhang J: MicroRNAs in the diagnosis and prognosis of breast cancer and their therapeutic potential (Review). Int J Oncol 45: 950-958, 2014
APA
Zhang, K., Zhang, Y., Liu, C., Xiong, Y., & Zhang, J. (2014). MicroRNAs in the diagnosis and prognosis of breast cancer and their therapeutic potential (Review). International Journal of Oncology, 45, 950-958. https://doi.org/10.3892/ijo.2014.2487
MLA
Zhang, K., Zhang, Y., Liu, C., Xiong, Y., Zhang, J."MicroRNAs in the diagnosis and prognosis of breast cancer and their therapeutic potential (Review)". International Journal of Oncology 45.3 (2014): 950-958.
Chicago
Zhang, K., Zhang, Y., Liu, C., Xiong, Y., Zhang, J."MicroRNAs in the diagnosis and prognosis of breast cancer and their therapeutic potential (Review)". International Journal of Oncology 45, no. 3 (2014): 950-958. https://doi.org/10.3892/ijo.2014.2487