J Breast Cancer. 2020 Apr;23(2):129-140. English.
Published online Jan 16, 2020.
© 2020 Korean Breast Cancer Society
Review

The Role of Long Noncoding RNAs in Antiestrogen Resistance in Breast Cancer: An Overview and Update

Lan Huang,1,* Guohua Liang,1,* Qingyuan Zhang,1,2,3 and Wenhui Zhao1
    • 1Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China.
    • 2Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China.
    • 3Heilongjiang Academy of Medical Sciences, Harbin, China.
Received August 29, 2019; Accepted January 09, 2020.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

As a standard treatment, endocrine therapy has dramatically enhanced the prognosis of patients with estrogen receptor (ER)-positive breast cancer, which accounts for nearly 70% of all breast cancers. Antiestrogen drugs such as tamoxifen and aromatase inhibitors are the standard treatment options for ERα-positive breast cancer. However, acquired antiestrogen resistance is still the leading cause of disease recurrence and progression. Evidence has shown that long noncoding RNAs (lncRNAs) play an essential role in the development of antiestrogen resistance in ER-positive breast cancer and can serve as biomarkers or potential therapeutic targets. This review highlights the role of lncRNAs in the development of antiestrogen resistance in breast cancer.

Keywords
Breast neoplasms; Drug resistance; Estrogen receptor modulators; RNA, long noncoding

INTRODUCTION

Breast cancer is the most prevalent malignancy and a major cause of cancer mortality in women worldwide. Up to 70% of breast cancer patients upregulate estrogen receptor (ER) and/or progesterone receptor, which indicates that the growth of these cancers is dependent on estrogen [1, 2]. Endocrine therapies targeting ERs, including selective ER modulators (SERMs), selective ER downregulators (SERDs), and aromatase inhibitors (AIs), as the dominant therapeutic approach, have dramatically improved hormone-dependent patient survival [3].

Tamoxifen, an antiestrogen drug, is commonly used in ER-positive (ERα+) breast cancer treatment and significantly improves overall survival [4]. A recent meta-analysis, that included 21,457 breast cancer female patients from 20 trials showed that tamoxifen treatment reduced mortality by 15 years in at least a third of them [5]. The third-generation AIs (i.e., exemestane, anastrozole, and letrozole) were observed to inhibit circulating estrogen levels by more than 97% in post-menopausal women with early-stage ERα+ breast cancer, and are thus the preferred treatment options [6]. However, de novo or acquired endocrine resistance, which occurred in approximately 50% of early-stage breast cancer patients and almost all patients with advanced disease, impairs patient survival and abrogates the initial beneficial response. The related mechanisms for the development of endocrine resistance have been proposed to include the following: mutations in ERα, the overactivation of growth factors or their corresponding receptors, the overexpression of oncogenes, and aberrant crosstalk between hormone receptors and signaling pathways, have been proposed [7]. Although the addition of the mammalian target of rapamycin (mTOR) complex-1 inhibitor everolimus or cyclin-dependent kinase 4/6 inhibitors to standard endocrine therapy has further extended recurrence-free survival, results remain unsatisfactory.

Long noncoding RNAs (lncRNAs) are a class of non-coding RNAs that are greater than 200 nucleotides in length and do not encode functional proteins. Studies have found that lncRNAs play roles in multiple cellular maintenance functions, such as protein scaffolding, chromatin looping, and the regulation of messenger RNA (mRNA) stability [8]. Although the exact functions of lncRNAs are still not fully understood, most of them were found to be critical regulators of gene expression. They alter chromatin or epigenetic modifications, transcriptional, and posttranscriptional gene regulation by interacting with RNAs and proteins [9]. The abnormal expression of lncRNAs has been detected in various malignant tumors [10]. In addition, studies have shown that changes in lncRNAs might be responsible for drug resistance, a major obstacle in cancer treatment.

The related mechanisms of lncRNA involvement in drug resistance are as follows: 1) the regulation of apoptosis-related proteins or transcription factors inhibiting tumor cell apoptosis; 2) the promotion of epithelial-mesenchymal transition (EMT) in tumor cells; 3) interaction with related microRNAs (miRNAs) to influence drug resistance; 4) improved DNA repair; 5) the regulation of cell membrane efflux and 6) the regulation of drug metabolism [11]. Since differential expression of lncRNAs was detected in sensitive and resistant tumors, the roles of lncRNAs in tamoxifen-resistant (TamR) ER+ breast cancer have been explored. Here, we reviewed the roles of specific lncRNAs involved in antiestrogen-resistant breast cancers and suggest that lncRNAs may serve as potential therapeutic targets for improvement of the clinical benefits of antiestrogen treatment.

SERMs: TAMOXIFEN

LncRNA breast cancer antiestrogen resistance 4 (BCAR4)

The lncRNA BCAR4 was first screened by Meijer et al. [12] in ZR-75-1 breast cancer TamR cells. The lncRNA BCAR4 is located at 16p13.13 and is 9017 bp long. It is normally expressed in the human placenta and oocytes [13]. Thus far studies have demonstrated that the lncRNA BCAR4 is abnormally expressed in various malignant tumors and is substantially related to the degree of malignancy [14]. It has been reported that the lncRNA BCAR4 is overexpressed in nearly 27% of primary breast cancers [12]. Overexpression of the lncRNA BCAR4 in endocrine-sensitive ZR-75-1 cells was observed to enhance cell invasion and proliferation [15].

It is well-established that the amplification of ERBB2 in breast cancer is a significant cause of tamoxifen treatment failure. The ERBB family, a group of receptor tyrosine kinases receptors, plays an essential role in many critical physiological processes that include, development, cell growth, differentiation, and tumorigenesis. Godinho et al. [15] predicted the amino acid sequence of the lncRNA BCAR4 and found 2 transmembrane domains in its molecular structure, suggesting that it may be located on the cell membrane. Considering that the lncRNA BCAR4 is overexpressed in TamR cells and is generally co-expressed with the human epidermal growth factor receptor 2 (HER2) molecule (the ERBB2 gene product) [16], the authors proposed that the lncRNA BCAR4 may act as a ligand for ERBB3—potentially by activating the ERBB2/ERBB3 pathways—to drive tamoxifen resistance [13]. As a critical transcription factor in the Hedgehog (Hh) pathway, glioma-associated oncogene homolog 2 (GLI2) is involved in tumor development, proliferation, and metastasis. Additionally, studies have shown that the lncRNA BCAR4 can promote endocrine therapy resistance via the non-canonical Hh/GLI2 pathway [14, 17]. Importantly, the authors further demonstrated that overexpression of the lncRNA BCAR4, independent of estrogen receptor 1 (ESR1) function, induced the conversion of estrogen-dependent breast cancer cells into an estrogen-independent phenotype.

Furthermore, high expression of the lncRNA BCAR4 may be linked to resistance to multiple drugs, such as raloxifene and fulvestrant (Faslodex) [13]. Thus, the lncRNA BCAR4 may act as a potential clinical biomarker for tamoxifen resistance [15]. Since lncRNA BCAR4-induced tamoxifen resistance may rely on the co-expression of HER2 [18], the specific targeting of the HER2 signaling pathway might be useful for patients with positive BCAR4 expression [15]. Further investigation is required to identify the mechanisms of this action.

HOX antisense intergenic RNA (HOTAIR)

The lncRNA HOTAIR is transcribed from the antisense strand of the homeobox C locus, a 2.2 kb gene located on chromosome 12. HOTAIR was the first identified lncRNA involved in trans-regulated gene transcription [19]. Studies have indicated that the lncRNA HOTAIR is upregulated in breast cancer, gastrointestinal stromal tumors, hepatocellular carcinoma, colorectal cancer, and pancreatic cancer. Moreover, high levels of the lncRNA HOTAIR increase the invasiveness of tumor cells, resulting in poor patient survival. Mechanistically, the lncRNA HOTAIR reprograms the chromatin status and promotes tumor metastasis through interaction with polycomb repressive complex 2 [20]. The lncRNA HOTAIR is a robust predictor of adverse outcomes in cancer, the high expression of the lncRNA HOTAIR is linked to breast cancer invasion, metastasis, and drug resistance, especially in ER+ breast cancer. Xue et al. [21] observed that the expression of the lncRNA HOTAIR was increased in TamR cells. Conversely, the downregulation of the lncRNA HOTAIR inhibited the colony-forming abilities of TamR cells.

The lncRNA HOTAIR is negatively regulated by estrogen. Evidence has shown that its expression is significantly increased under estrogen starvation or tamoxifen treatment. Importantly, the lncRNA HOTAIR can upregulate nuclear ER and further influence the expression of estrogen-responsive genes. This indicates that the lncRNA HOTAIR might stimulate endocrine therapy resistance in an estrogen-independent manner. In addition, the lncRNA HOTAIR is also regulated by many breast cancer-related transcription factors, such as forkhead box protein A1 (FOXA1) and forkhead box protein M1 (FOXM1). FOXA1 and FOXM1 are critical components of the ER signaling pathway in breast cancer and are closely associated with tamoxifen resistance and unfavorable outcomes [22, 23]. Therefore, the combination of HOTAIR and FOXM1 can better distinguish endocrine therapy responders from non-responders in antiestrogen therapy [24]. Recent studies have found that increased chromosomal instability (CIN) can induce tamoxifen resistance in ER+ breast cancer, and that the activation of ER signaling with high levels of CIN is likely to be a strong predictor of patient survival [25]. According to previous reports, increased FOXM1 can also have a positive impact on CIN levels [26]. Thus, Milevskiy et al. [24] proposed that HOTAIR might play a regulatory function between the CIN and ER pathways. It was found that the level of HOTAIR is increased while the CIN-related gene is amplified when ER signaling is suppressed (e.g. ER deletion). Given that high levels of CIN can affect the effectiveness of endocrine therapy and chemotherapy, CIN inhibitors such as threonine and tyrosine kinase and polo-like kinase 1 can potentially overcome drug resistance [27]. Consistent with this finding, studies have reported that breast cancer cells might be sensitive to CIN inhibitors in cases of ER receptor mutations or estrogen deficiency [25].

It is well known that EMT is involved in the development of multidrug resistance, and that HOTAIR overexpression induces EMT [27, 28]. Tian et al. [29] observed that TamR cells display a mesenchymal/fibroblast-like morphology, which is similar to cells undergoing the EMT process. Thus, researchers have proposed that HOTAIR promotes tamoxifen resistance by inducing EMT.

H19

The lncRNA H19 is located in the H19/IGF2 cluster on human chromosome 11p15.5. It was the first imprinted gene to be discovered [30]. The lncRNA H19 can function as a miRNA molecular sponge in genomic imprinting, transcriptional activation, and transcriptional interference [31]. High H19 expression is observed in an estimated 72.5% of breast cancers, and can increase the tumorigenicity and resistance to endocrine therapy in breast cancer.

In a study by Gao et al. [31], the knockout of H19 downregulated the expression of EMT-related transcription factors in TamR breast cancer cells by inhibiting Wnt/β-catenin pathway activation. This reversed the sensitivity of TamR cells, reduced cell proliferation and increased apoptosis. In concordance with this result, Basak et al. [32] also demonstrated that H19 is upregulated in both tamoxifen- and fulvestrant-resistant cells compared to endocrine-sensitive cells. In addition, the authors noted that the lncRNA H19 is regulated by the Notch and c-Met receptor pathways. When pharmacological inhibitors were applied to block Notch and c-Met receptor signaling, tumor cells lost their resistance to fulvestrant and tamoxifen in an H19-dependent manner. Therefore, patients who are resistant to fulvestrant or tamoxifen may benefit from treatments using Notch and c-Met inhibitors. Notably, H19 protects ERα from the degradation of fulvestrant. In addition, the lncRNA H19 is downregulated after ERα knockdown, indicating that they are mutually regulated each other.

Down syndrome cell adhesion molecule-antisense RNA (DSCAM-AS1)

The lncRNA DSCAM-AS1 has a total length of approximately 1.4 kb and is transcribed from the gene located on chromosome 21q22.3 (GRCh38/hg38). It was first described in 2002 by Liu et al. [33], who found its abnormally high expression in breast cancer. To date, studies have shown that the lncRNA DSCAM-AS1 is involved in vital biological processes in tumorigenicity, including DNA replication, G1/S phase transformation, sister chromatid condensation, chromosome separation, protein localization to the chromosome and DNA recombination [34]. Transcriptome sequencing data from 6,503 cancer samples, healthy tissues and cell lines from The Cancer Genome Atlas (http://cancergenome.nih.gov/) and Michigan Center for Translational Pathology showed that lncRNA DSCAM-AS1 is overexpressed specifically in breast and lung adenocarcinoma [35]. It has been proven that the knockdown of DSCAM-AS1 in nude mice can reduce the ability of liver metastasis in breast cancer cells. This suggests that DSCAM-AS1 may contribute to the liver metastasis of breast cancer [36].

Recent studies have suggested that the lncRNA DSCAM-AS1 is highly expressed in breast cancer cell lines with ER+ and HER-2 overexpression and weakly expressed in triple-negative breast cancer. As reported, the lncRNA DSCAM-AS1 sequence has an ER binding site, indicating that its expression is regulated by estrogen. Based on these results, Miano et al. [37] and Sun et al. [34] further found that DSCAM-AS1 is the most abundant lncRNA in ER+ breast cancer cells and is directly regulated by ERα. Thus, there may be positive feedback regulation between ERα and DSCAM-AS1.

Furthermore, data from Oncomine (https://www.oncomine.org) revealed that DSCAM-AS1 was associated with malignant biological behaviors linked to endocrine therapy resistance, high breast cancer grade, early recurrence, and metastasis. In a study by Niknafs et al. [36], the high expression of DSCAM-AS1 in MCF-7 TamR cells was detected. When DSCAM-AS1 was knocked down, the sensitivity to tamoxifen treatment in MCF-7 TamR cells was restored. It was also shown that lncRNA DSCAM-AS1 may enhance carcinogenicity and promote drug resistance through its interaction with heterogeneous nuclear ribonucleoprotein. Similarly, Ma et al. [38] demonstrated the overexpression of lncRNA DSCAM-AS1 in TamR breast cancer tissues. They suggested that DSCAM-AS1 regulates the EPS8 expression in breast cancer cells through miR-137, to promote cell proliferation and metastasis, inhibit apoptosis, and induce tamoxifen resistance. Therefore, the DSCAM-AS1/miR-137/EPS8 axis might be a potential therapeutic target for ER+ breast cancer [38].

Taken together, these results suggest that increased lncRNA DSCAM-AS1 expression predicts a poor prognosis and a high risk of endocrine therapy resistance in patients receiving endocrine therapy [34].

Urothelial carcinoma-associated 1 (UCA1)

The lncRNA UCA1 was first discovered in bladder cancer and is located on human chromosome 19p13.12. It is 1,439 bp in length and contains 3 exons and 2 introns [39]. According to reports, UCA1 is associated with resistance to a variety of drugs, such as cisplatin, gemcitabine, fluorouracil, tamoxifen, imatinib, and epidermal growth factor receptor tyrosine kinase inhibitors.

The lncRNA UCA1, is involved in carcinogenesis and is overexpressed in a number of drug-resistant malignant cells. Li et al. [40] suggested that UCA1 knockout in TamR breast cancer LCC2/LCC9 cell lines increases apoptosis in drug-resistant cells. In addition, the upregulation of UCA1 in TamR breast cancer cells was found to be hypoxia-inducible factor 1α (HIF-1α)-dependent. Consistent with these findings, Xu et al. [41] noted that the introduction of exosomes carrying bioactive lncRNA UCA1 into tamoxifen-sensitive MCF-7 breast cancer cells and significantly increased tamoxifen resistance. Functionally, UCA1 acts as a molecular sponge to adsorb miR-18a, a negative regulator of HIF-1α. The upregulation of HIF-1α hence enhances UCA1 expression and induces tamoxifen resistance [42].

The activation of the Wnt/β-catenin signaling pathway promotes proliferation and survival and maintains the stem-like characteristics of breast cancer cells, associated with the development of resistance to various antitumor drugs, including tamoxifen [43]. Evidence has shown that the lncRNA UCA1 induces tamoxifen resistance by activating Wnt/β-Catenin signaling [44]. It is well documented that the PI3K/AKT/mTOR pathway plays an important role in the promotion of tamoxifen resistance in ER+ breast cancer. Everolimus (an mTOR inhibitor) is effective in reversing tamoxifen resistance. Previous studies have demonstrated that elevated UCA1 enhances the activation of the AKT/mTOR pathway in various types of tumors [45]. Targeting mTOR significantly inhibited tamoxifen resistance induced by UCA1 overexpression, suggesting that the lncRNA UCA1 partially reduces breast cancer cell sensitivity to tamoxifen by activating the mTOR pathway [46]. Finally, UCA1 regulates the cell cycle by affecting the expression of the p21 protein by enhancer of zeste homolog 2 to control G2/M phase transition or to regulate the cell cycle by altering PI3K/AKT pathway activity and cAMP response element-binding protein transcription factors [40].

Regulator of reprogramming (ROR)

The lincRNA ROR was discovered by Loewer et al. [47] in embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). It is located on chromosome 18 and is a 2.6 kb-long transcript with 4 exons expressed both in the nucleus and cytoplasm. Functionally, the lincRNA ROR modulates the formation of iPSCs by regulating pluripotency transcription factors, such as octamer binding transcription factor 4, SRY-box 2, and Nanog homeobox, to maintain ESC self-renewal. The overexpression of the lincRNA ROR has been reported to contribute to tumorigenesis and progression. In addition, the lincRNA ROR acts as an important regulator of EMT and promotes the invasion and migration of several tumors, including breast cancer [48]. In contrast, silencing lncRNA ROR inhibited breast cancer cell growth and lung metastasis.

Due to the role of lncRNAs in drug resistance, Zhang et al. [49] found that the expression of the lincRNA ROR was positively correlated with tamoxifen resistance. It is well known that the transition from estrogen-dependence to estrogen-independence in ER+ breast cancer cells is a key process in the development of endocrine therapy resistance [50, 51]. It is also well-established that the mitogen-activated protein kinase/extracellular regulated protein kinase (MAPK/ERK) pathway is involved in the estrogen-independent growth of breast cancer. In the absence of estrogen, it was found that the lincRNA ROR, as a regulator of ER signaling, upregulates the phosphorylated MAPK/ERK pathway and activates ER signaling [50, 52].

Moreover, studies have confirmed that lincRNA ROR knockdown restores tamoxifen sensitivity in MCF7-TamR cells. It was also observed that the downregulation of the lincRNA ROR increased miR-205 levels, which inhibited the expression of zinc finger E-box binding homeobox (ZEB) 1 and ZEB2, and further reversed EMT [49]. Furthermore, Li et al. [53] found that inhibition of the lincRNA ROR reversed tamoxifen resistance by inducing autophagy.

Other lncRNAs

The luminal lncRNA (LOL) is a novel lncRNA highly expressed in breast cancers, especially ER+ breast cancer. It can thus be an independent prognostic factor for a poor prognosis. LOL represents an enhancer-associated lncRNA, which is extremely sensitive to enhancer-regulating factors ZMYND8 and BRD4. Bioinformatics analysis and GEO database evaluation revealed that LOL might not be directly regulated by ERα. Either estrogen deprivation or ERα signaling pathway blockage can stimulate LOL expression, which can in turn promote tumor progression [54]. The lncRNA LOL is significantly upregulated in MCF-7 TamR cells, which acts as a natural sponge for let-7 to promote tumor growth and tamoxifen resistance by enhancing the expression of let-7 target genes (including CCND1, CDC25A, DICER, PBX1, MYC, and ESR1) [54]. LncRNA CCAT2 is a non-coding RNA located at 8q24, and functions as an oncogene in breast cancer development. Cai et al. [55] showed that the high level of the lncRNA CCAT2 present in TamR cells was related to cell proliferation, inhibition of apoptosis, and tamoxifen resistance. In addition, they observed that the expression of CCAT2 was suppressed when the hyper-activated ERK/MAPK signaling pathway was blocked.

LncRNAs also inhibit the development of drug resistance. LINC00894-002 is significantly downregulated in MCF-7 TamR cells was the first lncRNA discovered to inhibit the development of tamoxifen resistance. It is transcribed from a locus on the X chromosome and may act as a tumor suppressor in various cancers. There is crosstalk between the ER and transforming growth factor-β (TGF-β) pathways and they are both critical pathways for the development of tamoxifen resistance. LINC00894 suppresses TGF-β2/ZEB1 signaling by adsorbing miR-200, thereby lowering the occurrence of drug resistance. In addition, LINC00894-002 can be upregulated by ERα activation and positively modulates the expression of miR-200a-3p and miR-200b-3p to inhibit the downstream TGF-β2/ZEB1 signaling pathway [56]. Moreover, Zhang et al. [57] found that lncRNA uc.57 and its downstream gene B-cell lymphoma/leukaemia 11A (BCL11A) were differentially expressed in MCF-7 and MCF-7 TamR cells. They suggested that the expression levels of BCL11A, as the target gene of uc.57, was positively correlated with the development of TamR. Mechanistically, uc.57 downregulated BCL11A and inhibited drug resistance by inhibiting the PI3K/AKT and MAPK pathways.

As reported, the activation of nuclear factor kappa-B (NF-κB) activation promotes tamoxifen resistance in breast cancer patients [58]. Wang et al. [59] found that the overexpression of LINC00472 regulated the interaction between NF-κB and ERα and reduced the growth, invasion, and drug resistance associated with breast cancer. Since ERα binds to the LINC00472 promoter and upregulates the expression of LINC00472, LINC00472 targets NF-κB and negatively regulates its expression. Therefore, the long-term usage of tamoxifen may reduce the suppression of NF-κB phosphorylation through LINC00472, inducing endocrine resistance and tumor progression.

Finally, lncRNA growth arrest-specific transcript 5 (GAS5) is localized at chromosome 1q25 and was originally isolated from a subtraction complementary DNA library. It was found to be markedly downregulated in MCF-7 TamR (MCF-7R) cells [60]. The lncRNA GAS5 reduces miR-222 levels by sponge adsorption and leads to the upregulation of phosphatase tensin homologs (PTEN). Thus, the lncRNA GAS5 partially restores the sensitivity of MCF-7R cells to tamoxifen via the lncRNA GAS5/miR-222/PTEN pathway [61]. Similarly, the lncRNA ADAMTS9AS2 (an antisense transcript of tumor suppressor ADAMTS9) enhances PTEN expression by targeting miRNA-130a-5p and restores cell sensitivity to tamoxifen [62, 63].

AIs

To date, few studies have focused on the role of lncRNA in AI resistance. In the study by Ingle et al. [64], lncRNA MIR2052HG was defined as a functionally polymorphic gene, which increases the risk of breast cancer recurrence in women treated with AI. They identified 2 single nucleotide polymorphisms (SNPs) (rs4476990 and rs3802201) in the gene, MIR2052HG. Their results showed that estrogen and AI induced MIR2052HG and ERα expression in a SNP-dependent manner. Overexpression of MIR2052HG promotes cell proliferation, colony formation, and ERα expression. Mechanistic investigations revealed that MIR2052HG sustained ERα expression both by enhancing AKT/FOXO3-mediated ESR1 (gene, encoding ERα) transcription and by preventing ubiquitin-mediated proteasome-dependent degradation of ERα.

The important role of lemur tyrosine kinase 3 (LMTK3) in de novo and acquired endocrine resistance in breast cancer has been reported [65]. Cairns et al. [66] found that MIR2052HG enhanced LMTK3 transcription by directly interacting with early growth response protein 1. High levels of LMTK3 in turn, sustained ESR1 expression and stabilized ERα protein. Mechanistically, MIR2052HG regulates LMTK3 in a SNP- and AI-dependent manner and LMTK3 regulates ERα stability via the PKC/MEK/ERK/RSK1 axis. Therefore, MIR2052HG plays a key role in regulating ERα and endocrine resistance.

SERDs: FULVESTRANT

At the time of writing, only 1 study reported a lncRNA associated with fulvestrant resistance. As described above, the overexpression of H19 can also induce fulvestrant resistance through Notch and HGF signaling. It was demonstrated that H19 regulates ERα expression at the mRNA and protein levels, and in turn, protects ERα proteins from fulvestrant-mediated downregulation. The combination of pharmacological inhibitors of Notch and c-MET with fulvestrant significantly restored the sensitivity of drug-resistant cells to fulvestrant in an H19-dependent manner [32]. A summary is provided in Table 1.

FUTURE PROSPECTS

In this review, we summarized the lncRNAs that are differentially expressed between breast cancers resistant and sensitive to antiestrogens. Although the role of lncRNAs in reversing tamoxifen resistance is undeniable, there are still some fundamental issues that need to be further addressed. First, given that breast cancer is heterogeneous, different lncRNAs may have different regulatory effects on tamoxifen resistance. Second, the study of lncRNA involvement in drug resistance is still limited to in vitro experiments, and further validation using in vivo experiments is necessary. Finally, lncRNAs have been confirmed to be closely related to the development of cancer, and most of the exosomal lncRNAs are stably present in human body fluids. The study of exosomal lncRNAs (as a kind of liquid biopsy) may be valuable in cancer diagnosis, prognosis assessment, the prediction of drug resistance and treatment outcome [67].

CONCLUSION

Considering the extensive clinical application of endocrine therapies, there is an urgent need for the prevention, early prediction and management of antiestrogen resistance, which will contribute to prolonged patient survival. LncRNAs may serve as a potential therapeutic target for the improvement of antiestrogen treatments.

Notes

Funding:This study was supported by the Postdoctoral Scientific Research Developmental Fund of Heilongjiang Province, China (grant No. LBH-Q16148), the provincial natural science foundation project of Heilongjiang Province, China (grant No. H2018044), and the Foundation of Science and Technology Research Project of the Heilongjiang Education Department.

Conflict of Interest:The authors declare that they have no competing interests.

Author Contributions:

  • Funding acquisition: Zhao W.

  • Investigation: Huang L.

  • Supervision: Zhao W.

  • Writing - original draft: Huang L, Liang G.

  • Writing - review & editing: Zhang Q, Zhao W.

References

    1. Pritchard KI, Gelmon KA, Rayson D, Provencher L, Webster M, McLeod D, et al. Endocrine therapy for postmenopausal women with hormone receptor-positive her2-negative advanced breast cancer after progression or recurrence on nonsteroidal aromatase inhibitor therapy: a Canadian consensus statement. Curr Oncol 2013;20:48–61.
    1. Gradishar W, Salerno KE. NCCN guidelines update: breast cancer. J Natl Compr Canc Netw 2016;14:641–644.
    1. Razavi P, Chang MT, Xu G, Bandlamudi C, Ross DS, Vasan N, et al. The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell 2018;34:427–438.e6.
    1. Jordan VC. Tamoxifen as the first targeted long-term adjuvant therapy for breast cancer. Endocr Relat Cancer 2014;21:R235–46.
    1. Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Davies C, Godwin J, Gray R, Clarke M, Cutter D, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 2011;378:771–784.
    1. Khosrow-Khavar F, Yin H, Barkun A, Bouganim N, Azoulay L. Aromatase inhibitors and the risk of colorectal cancer in postmenopausal women with breast cancer. Ann Oncol 2018;29:744–748.
    1. Ziauddin MF, Hua D, Tang SC. Emerging strategies to overcome resistance to endocrine therapy for breast cancer. Cancer Metastasis Rev 2014;33:791–807.
    1. Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 2011;472:120–124.
    1. Han P, Chang CP. Long non-coding RNA and chromatin remodeling. RNA Biol 2015;12:1094–1098.
    1. Zhang Q, Su M, Lu G, Wang J. The complexity of bladder cancer: long noncoding RNAs are on the stage. Mol Cancer 2013;12:101.
    1. Majidinia M, Yousefi B. Long non-coding RNAs in cancer drug resistance development. DNA Repair (Amst) 2016;45:25–33.
    1. Meijer D, van Agthoven T, Bosma PT, Nooter K, Dorssers LC. Functional screen for genes responsible for tamoxifen resistance in human breast cancer cells. Mol Cancer Res 2006;4:379–386.
    1. Godinho M, Meijer D, Setyono-Han B, Dorssers LC, van Agthoven T. Characterization of BCAR4, a novel oncogene causing endocrine resistance in human breast cancer cells. J Cell Physiol 2011;226:1741–1749.
    1. Yang H, Yan L, Sun K, Sun X, Zhang X, Cai K, et al. LncRNA BCAR4 increases viability, invasion, and migration of non-small cell lung cancer cells by targeting glioma-associated oncogene 2 (GLI2). Oncol Res 2019;27:359–369.
    1. Godinho MF, Sieuwerts AM, Look MP, Meijer D, Foekens JA, Dorssers LC, et al. Relevance of BCAR4 in tamoxifen resistance and tumour aggressiveness of human breast cancer. Br J Cancer 2010;103:1284–1291.
    1. Hurtado A, Holmes KA, Geistlinger TR, Hutcheson IR, Nicholson RI, Brown M, et al. Regulation of ERBB2 by oestrogen receptor-PAX2 determines response to tamoxifen. Nature 2008;456:663–666.
    1. Chen F, Mo J, Zhang L. Long noncoding RNA BCAR4 promotes osteosarcoma progression through activating GLI2-dependent gene transcription. Tumour Biol 2016;37:13403–13412.
    1. Godinho MF, Wulfkuhle JD, Look MP, Sieuwerts AM, Sleijfer S, Foekens JA, et al. BCAR4 induces antioestrogen resistance but sensitises breast cancer to lapatinib. Br J Cancer 2012;107:947–955.
    1. Gupta S, Iljin K, Sara H, Mpindi JP, Mirtti T, Vainio P, et al. FZD4 as a mediator of ERG oncogene-induced WNT signaling and epithelial-to-mesenchymal transition in human prostate cancer cells. Cancer Res 2010;70:6735–6745.
    1. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010;464:1071–1076.
    1. Xue X, Yang YA, Zhang A, Fong KW, Kim J, Song B, et al. LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast cancer. Oncogene 2016;35:2746–2755.
    1. Bergamaschi A, Madak-Erdogan Z, Kim YJ, Choi YL, Lu H, Katzenellenbogen BS. The forkhead transcription factor FOXM1 promotes endocrine resistance and invasiveness in estrogen receptor-positive breast cancer by expansion of stem-like cancer cells. Breast Cancer Res 2014;16:436.
    1. Sanders DA, Ross-Innes CS, Beraldi D, Carroll JS, Balasubramanian S. Genome-wide mapping of FOXM1 binding reveals co-binding with estrogen receptor alpha in breast cancer cells. Genome Biol 2013;14:R6.
    1. Milevskiy MJ, Al-Ejeh F, Saunus JM, Northwood KS, Bailey PJ, Betts JA, et al. Long-range regulators of the lncRNA HOTAIR enhance its prognostic potential in breast cancer. Hum Mol Genet 2016;25:3269–3283.
    1. Al-Ejeh F, Simpson PT, Saunus JM, Klein K, Kalimutho M, Shi W, et al. Meta-analysis of the global gene expression profile of triple-negative breast cancer identifies genes for the prognostication and treatment of aggressive breast cancer. Oncogenesis 2014;3:e124
    1. Cheng WY, Ou Yang TH, Anastassiou D. Biomolecular events in cancer revealed by attractor metagenes. PLOS Comput Biol 2013;9:e1002920
    1. Al-Ejeh F, Shi W, Miranda M, Simpson PT, Vargas AC, Song S, et al. Treatment of triple-negative breast cancer using anti-EGFR-directed radioimmunotherapy combined with radiosensitizing chemotherapy and PARP inhibitor. J Nucl Med 2013;54:913–921.
    1. Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, et al. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 2013;32:1616–1625.
    1. Tian M, Schiemann WP. TGF-β Stimulation of EMT programs elicits non-genomic ER-α activity and anti-estrogen resistance in breast cancer cells. J Cancer Metastasis Treat 2017;3:150–160.
    1. Brannan CI, Dees EC, Ingram RS, Tilghman SM. The product of the H19 gene may function as an RNA. Mol Cell Biol 1990;10:28–36.
    1. Gao H, Hao G, Sun Y, Li L, Wang Y. Long noncoding RNA H19 mediated the chemosensitivity of breast cancer cells via Wnt pathway and EMT process. Onco Targets Ther 2018;11:8001–8012.
    1. Basak P, Chatterjee S, Bhat V, Su A, Jin H, Lee-Wing V, et al. Long non-coding RNA H19 acts as an estrogen receptor modulator that is required for endocrine therapy resistance in ER+ breast cancer cells. Cell Physiol Biochem 2018;51:1518–1532.
    1. Liu D, Rudland PS, Sibson DR, Barraclough R. Identification of mRNAs differentially-expressed between benign and malignant breast tumour cells. Br J Cancer 2002;87:423–431.
    1. Sun W, Li AQ, Zhou P, Jiang YZ, Jin X, Liu YR, et al. DSCAM-AS1 regulates the G1 /S cell cycle transition and is an independent prognostic factor of poor survival in luminal breast cancer patients treated with endocrine therapy. Cancer Med 2018;7:6137–6146.
    1. Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet 2015;47:199–208.
    1. Niknafs YS, Han S, Ma T, Speers C, Zhang C, Wilder-Romans K, et al. The lncRNA landscape of breast cancer reveals a role for DSCAM-AS1 in breast cancer progression. Nat Commun 2016;7:12791.
    1. Miano V, Ferrero G, Reineri S, Caizzi L, Annaratone L, Ricci L, et al. Luminal long non-coding RNAs regulated by estrogen receptor alpha in a ligand-independent manner show functional roles in breast cancer. Oncotarget 2016;7:3201–3216.
    1. Ma Y, Bu D, Long J, Chai W, Dong J. LncRNA DSCAM-AS1 acts as a sponge of miR-137 to enhance tamoxifen resistance in breast cancer. J Cell Physiol 2019;234:2880–2894.
    1. Wang XS, Zhang Z, Wang HC, Cai JL, Xu QW, Li MQ, et al. Rapid identification of UCA1 as a very sensitive and specific unique marker for human bladder carcinoma. Clin Cancer Res 2006;12:4851–4858.
    1. Li Z, Yu D, Li H, Lv Y, Li S. Long non-coding RNA UCA1 confers tamoxifen resistance in breast cancer endocrinotherapy through regulation of the EZH2/p21 axis and the PI3K/AKT signaling pathway. Int J Oncol 2019;54:1033–1042.
    1. Xu CG, Yang MF, Ren YQ, Wu CH, Wang LQ. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur Rev Med Pharmacol Sci 2016;20:4362–4368.
    1. Li X, Wu Y, Liu A, Tang X. Long non-coding RNA UCA1 enhances tamoxifen resistance in breast cancer cells through a miR-18a-HIF1α feedback regulatory loop. Tumour Biol 2016;37:14733–14743.
    1. Wang H, Guan Z, He K, Qian J, Cao J, Teng L. LncRNA UCA1 in anti-cancer drug resistance. Oncotarget 2017;8:64638–64650.
    1. Liu H, Wang G, Yang L, Qu J, Yang Z, Zhou X. Knockdown of long non-coding RNA UCA1 increases the tamoxifen sensitivity of breast cancer cells through inhibition of Wnt/β-catenin pathway. PLoS One 2016;11:e0168406
    1. Li Z, Li X, Wu S, Xue M, Chen W. Long non-coding RNA UCA1 promotes glycolysis by upregulating hexokinase 2 through the mTOR-STAT3/microRNA143 pathway. Cancer Sci 2014;105:951–955.
    1. Wu C, Luo J. Long non-coding RNA (lncRNA) urothelial carcinoma-associated 1 (UCA1) enhances tamoxifen resistance in breast cancer cells via inhibiting mTOR signaling pathway. Med Sci Monit 2016;22:3860–3867.
    1. Loewer S, Cabili MN, Guttman M, Loh YH, Thomas K, Park IH, et al. Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet 2010;42:1113–1117.
    1. Hou P, Zhao Y, Li Z, Yao R, Ma M, Gao Y, et al. LincRNA-ROR induces epithelial-to-mesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis 2014;5:e1287
    1. Zhang HY, Liang F, Zhang JW, Wang F, Wang L, Kang XG. Effects of long noncoding RNA-ROR on tamoxifen resistance of breast cancer cells by regulating microRNA-205. Cancer Chemother Pharmacol 2017;79:327–337.
    1. Kümler I, Knoop AS, Jessing CA, Ejlertsen B, Nielsen DL. Review of hormone-based treatments in postmenopausal patients with advanced breast cancer focusing on aromatase inhibitors and fulvestrant. ESMO Open 2016;1:e000062
    1. Sachdeva M, Wu H, Ru P, Hwang L, Trieu V, Mo YY. MicroRNA-101-mediated Akt activation and estrogen-independent growth. Oncogene 2011;30:822–831.
    1. Peng WX, Huang JG, Yang L, Gong AH, Mo YY. Linc-RoR promotes MAPK/ERK signaling and confers estrogen-independent growth of breast cancer. Mol Cancer 2017;16:161.
    1. Li Y, Jiang B, Zhu H, Qu X, Zhao L, Tan Y, et al. Inhibition of long non-coding RNA ROR reverses resistance to tamoxifen by inducing autophagy in breast cancer. Tumour Biol 2017;39:1010428317705790
    1. Sun W, Xu X, Jiang Y, Jin X, Zhou P, Liu Y, et al. Transcriptome analysis of luminal breast cancer reveals a role for LOL in tumor progression and tamoxifen resistance. Int J Cancer 2019;145:842–856.
    1. Cai Y, He J, Zhang D. Suppression of long non-coding RNA CCAT2 improves tamoxifen-resistant breast cancer cells' response to tamoxifen. Mol Biol (Mosk) 2016;50:821–827.
    1. Zhang X, Wang M, Sun H, Zhu T, Wang X. Downregulation of LINC00894-002 contributes to tamoxifen resistance by enhancing the TGF-β signaling pathway. Biochemistry (Mosc) 2018;83:603–611.
    1. Zhang CH, Wang J, Zhang LX, Lu YH, Ji TH, Xu L, et al. Shikonin reduces tamoxifen resistance through long non-coding RNA uc.57. Oncotarget 2017;8:88658–88669.
    1. Riggins RB, Zwart A, Nehra R, Clarke R. The nuclear factor kappa B inhibitor parthenolide restores ICI 182,780 (Faslodex; fulvestrant)-induced apoptosis in antiestrogen-resistant breast cancer cells. Mol Cancer Ther 2005;4:33–41.
    1. Wang Z, Katsaros D, Biglia N, Shen Y, Loo L, Yu X, et al. ERα upregulates the expression of long non-coding RNA LINC00472 which suppresses the phosphorylation of NF-κB in breast cancer. Breast Cancer Res Treat 2019;175:353–368.
    1. Schneider C, King RM, Philipson L. Genes specifically expressed at growth arrest of mammalian cells. Cell 1988;54:787–793.
    1. Gu J, Wang Y, Wang X, Zhou D, Shao C, Zhou M, et al. Downregulation of lncRNA GAS5 confers tamoxifen resistance by activating miR-222 in breast cancer. Cancer Lett 2018;434:1–10.
    1. Shi YF, Lu H, Wang HB. Downregulated lncRNA ADAMTS9-AS2 in breast cancer enhances tamoxifen resistance by activating microRNA-130a-5p. Eur Rev Med Pharmacol Sci 2019;23:1563–1573.
    1. Yao J, Zhou B, Zhang J, Geng P, Liu K, Zhu Y, et al. A new tumor suppressor lncRNA ADAMTS9-AS2 is regulated by DNMT1 and inhibits migration of glioma cells. Tumour Biol 2014;35:7935–7944.
    1. Ingle JN, Xie F, Ellis MJ, Goss PE, Shepherd LE, Chapman JW, et al. Genetic polymorphisms in the long noncoding RNA MIR2052HG offer a pharmacogenomic basis for the response of breast cancer patients to aromatase inhibitor therapy. Cancer Res 2016;76:7012–7023.
    1. Stebbing J, Filipovic A, Lit LC, Blighe K, Grothey A, Xu Y, et al. LMTK3 is implicated in endocrine resistance via multiple signaling pathways. Oncogene 2013;32:3371–3380.
    1. Cairns J, Ingle JN, Kalari KR, Shepherd LE, Kubo M, Goetz MP, et al. The lncRNA MIR2052HG regulates ERα levels and aromatase inhibitor resistance through LMTK3 by recruiting EGR1. Breast Cancer Res 2019;21:47.
    1. Fan Q, Yang L, Zhang X, Peng X, Wei S, Su D, et al. The emerging role of exosome-derived non-coding RNAs in cancer biology. Cancer Lett 2018;414:107–115.

Metrics
Share
Tables

1 / 1

PERMALINK