Next Article in Journal
Evolution of the Tumor Microenvironment toward Immune-Suppressive Seclusion during Brain Metastasis of Breast Cancer: Implications for Targeted Therapy
Next Article in Special Issue
Dynamics of Serum Thymidine Kinase 1 at the First Cycle of Neoadjuvant Chemotherapy Predicts Outcome of Disease in Estrogen-Receptor-Positive Breast Cancer
Previous Article in Journal
Anti-PD-1/Anti-PD-L1 Drugs and Radiation Therapy: Combinations and Optimization Strategies
Previous Article in Special Issue
A Novel Immunomodulatory 27-Gene Signature to Predict Response to Neoadjuvant Immunochemotherapy for Primary Triple-Negative Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Circulating and Intracellular miRNAs as Prognostic and Predictive Factors in HER2-Positive Early Breast Cancer Treated with Neoadjuvant Chemotherapy: A Review of the Literature

1
Division of Medical Oncology, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41124 Modena, Italy
2
Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
3
Division of Medical Oncology, Department of Oncology-Hematology, University Hospital of Modena, 41124 Modena, Italy
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(19), 4894; https://doi.org/10.3390/cancers13194894
Submission received: 4 August 2021 / Revised: 24 September 2021 / Accepted: 27 September 2021 / Published: 29 September 2021
(This article belongs to the Special Issue Neoadjuvant Therapy in Breast Cancer)

Abstract

:

Simple Summary

Breast cancer is a leading cause of female cancer-related death worldwide. Anti-HER2-targeted therapies dramatically improved prognosis for HER2-positive breast cancer patients. Despite that, growing drug resistance due to the pressure of therapy is relatively frequent. For that reason, it is necessary to find biomarkers able to predict treatment sensitivity and survival outcomes. Increasing research has shown how miRNAs, secreted by tumor cells, are strongly involved in cancer development. In this review, we will discuss the recent evidence on the predictive and prognostic value of miRNAs involved in HER2-positive early breast cancer progression.

Abstract

MicroRNAs (miRNA) are small noncoding RNAs that can act as both oncogene and tumor suppressors. Deregulated miRNA expression has been detected in human cancers, including breast cancer (BC). Considering their important roles in tumorigenesis, miRNAs have been investigated as potential prognostic and diagnostic biomarkers. Neoadjuvant setting is an optimal model to investigate in vivo the mechanism of treatment resistance. In the management of human epidermal growth factor receptor-2 (HER2)-positive early BC, the anti-HER2-targeted therapies have drastically changed the survival outcomes. Despite this, growing drug resistance due to the pressure of therapy is relatively frequent. In the present review, we focused on the main miRNAs involved in HER2-positive BC tumorigenesis and discussed the recent evidence on their predictive and prognostic value.

1. Introduction

Breast cancer (BC) is the most common type of cancer diagnosed in women worldwide, with around 2.3 million new cases in 2020 and 685,000 deaths globally according to the World Health Organization [1]. The human epidermal growth factor receptor-2 (HER2) is overexpressed in almost 20% of cases and is generally associated with poorer survival outcomes [2]. Thanks to the advent of anti-HER2-targeted therapies, prognosis for HER2-positive BC patients has dramatically improved [3,4,5,6,7,8,9,10,11,12]. Despite their objective activity, intrinsic and acquired resistance to anti-HER2 agents remains a major clinical challenge [13]. To address this problem of resistance, considerable efforts have been made to investigate the mechanism of treatment failure. The neoadjuvant setting is an optimal model for studying in vivo the mechanism of treatment resistance [14,15].
Growing evidence has indicated that microRNA (miRNA) profiling could be a promising biomarker for early diagnosis, treatment sensitivity/resistance, and prognosis in different cancer types, including BC [16,17,18]. MiRNAs are a class of small noncoding regulatory RNAs that are involved in both physiological and pathological processes, such as cancer proliferation [19]. miRNAs can be easily detected in blood and tumor tissue [19]. In particular, miRNAs secreted by cancer cells are packed into extracellular vesicles (EVs) and released into peripheral blood. EVs are largely involved in the cross-talk between cancer cells and the microenvironment [20,21]. The important involvement of EVs and miRNA in ontogenetic processes and their availability in biological fluids, make these molecules optimal biomarkers in oncology [20,22].
This review summarizes the main miRNAs involved in HER2-positive BC tumorigenesis, focusing on their prognostic and predictive role.

2. Extracellular Vesicles and MicroRNAs

EVs are small lipid bilayer-delimited particles that are naturally released by cells and involved in cell communication [23]. EVs can be detected in biological fluids such as serum, plasma, urine, saliva, breast milk, bile, amniotic fluid, and cerebrospinal fluid [24,25]. Once released into the extracellular fluid, EVs may link to the cell plasma membrane, fusing directly with the membrane or being endocytosed, resulting in delivery of proteins and RNA into the target cell [24]. EVs are involved in several biological processes such as antigen presentation, immune regulation, apoptosis evasion, drug resistance, and angiogenesis [25,26,27,28]. Based on their size, they are classified into apoptotic bodies (50–5000 nm) released by apoptotic cells, shedding microvesicles/ectosomes (100–1000 nm) produced by budding from the plasma membrane, and exosomes (30–150 nm) that originate from intracellular multivesicular bodies and are secreted in the extracellular space upon fusion with the plasma membrane [20,23,29]. They carry proteins, metabolites, lipids, mRNA, miRNA, long-non-coding RNA, and DNA [24,25,26,30,31]. A large body of evidence suggests that cancer cells release a higher amount of EVs compared to non-malignant cells [25,26]. The most common methods reported for isolating exosomes from serum or plasma are commercial kits available for the isolation of EVs the size of exosomes (e.g., Total Exosomes Isolation Kit—Invitrogen) [29]. Exosomes contain miRNAs that are strongly involved in the cross-talk between cancer cells and the microenvironment. miRNAs are short, noncoding, single-stranded RNAs of 21–25 nucleotides, which regulate gene expression at a post transcriptional level by binding to the 3′untranslated region (3′UTR) of its target messenger RNA [32]. miRNAs can be present in the peripheral blood or packed into EVs such as exosomes [29]. MiRNAs are a class of small, noncoding, regulatory RNAs that are involved in both physiological and pathological processes, such as cancer proliferation. In physiological processes, miRNA are produced by non-cancerous cells. The most common techniques reported for miRNA profiling are: microarray analysis; bead-based flow-cytometric techniques; and real-time PCR, SAGE, or RAKE assay, with peculiar strengths and weaknesses under investigation [33,34,35,36,37].

3. miRNAs in HER2-Positive Breast Cancer

Several preclinical and clinical studies have focused their attention on the research of miRNAs linked to HER2 pathways in BC. In most of these cases, miRNAs were isolated from tumor tissues (cell lines or formalin-fixed paraffin embedded samples). Table 1 summarizes all the miRNAs known to be involved in HER2-positive BC tumorigenesis. The most widely reported miRNAs in the literature are: miR-155, miR-21, miR-205, and miR-125 families. In the literature, the most widely cited miRNAs related to HER2-positive subtypes are the following:
miR-155 has been found upregulated in HER2-positive BC in both early and metastatic settings [38]. miR-155 has an oncogene function in the interaction with the MAPK pathway through constitutive activation of STATs proteins by inhibiting SOCS1 gene and activating AKT and Src [39,40].
miR-21 is one of the most investigated miRNAs in oncology. It has been found overexpressed in all BC subtypes, but mainly in the HER2-positive hormone receptors negative subtype [41,42]. In particular, high expression of miR-21 has been found to significantly correlate with more aggressive tumor behavior (larger tumor with high nuclear grade) and poor survival outcomes [43]. In vitro studies have shown that up-regulation of miR-21 induced by HER2/neu signaling via MAPK (ERK1/2) pathway promotes cell invasion through downregulation of PDCD4 (programmed cell death 4) [43].
miR-205 has been detected in serum of BC patients with a decreasing expression from the less aggressive BC subtypes to the more aggressive ones, mainly in HER2-positive and triple-negative tumors [44,45]. It targets HER3 directly, inhibiting SKBr3 in BC cell lines [46,47]. Its oncosuppressive role is also linked by EMT suppression [45].
Its downregulation in serum of BC patients compared to healthy people suggests a possible role as a marker for BC diagnosis [63,64].
miR-125b belongs to the miR-125 family. miR-125b is generated from two genes: miR-125b-1 (on chromosome 11q24) and miR-125b-2 (on chromosome 21q21). It can be up- or downregulated depending on the tumor type [69,70,71,72]. In BC patients, its upregulation seems to promote EMT expression with an important implication in metastasis development [73]. Other direct targets of miR-125b are the EGFR family genes, mainly ERBB2 and ERBB3. In particular, in BC cell lines, miR-125b seems to induce a suppression of these genes [74]. Evidence from paraffin-embedded BC tissue samples reported a significant correlation between miR125b expression and HER2 expression (p < 0.001), with an important prognostic implication for the patient’s overall survival (OS) [66].
Finally, Lowery et al, through the expression profiling analysis of 453 miRNAs from formalin-fixed paraffin-embedded samples of 29 early BC patients, using an artificial neural network (ANN) and RQ-PCR for validation, reported a signature of five miRNAs (miR-520d, miR-181c, miR-302c, miR-376b, and miR-30e-3p), which seemed to accurately predict HER2 status in early BC patients [51].

4. Predictive Biomarkers

The identification of biomarkers able to predict treatment sensitivity before any intervention remains one of the main goal for oncologists. Thanks to the easy availability of miRNAs, they can be an optimal biomarker. Several published papers investigated the predictive role of miRNAs isolated in both BC tissue and blood. Table 2 summarizes miRNAs known to be predictive of chemotherapy or anti-HER2-targeted therapy sensitivity. In particular, data from different clinical and preclinical studies identified miR-125b, miR21, miR-210, and miR-155 as predictive biomarkers of trastuzumab resistance. Circulating miR-125b in blood serum of early BC patients seems to be significantly associated with neoadjuvant chemotherapy response. In particular, higher expression levels of miR-125b have been reported in resistant patients compared to responsive ones [75]. Wang et al. demonstrated an association between higher miR-125b expression and a higher percentage of proliferating cells in BC tissue [75]. Biologically, it can be explained by the Bak1 (Bcl-2 antagonist Killer 1) downregulation induced by miR-125b, which suppresses drug efficacy [76]. BaK1 is also known to be involved in the epithelial–mesenchymal transition process through EMT dysregulation, resulting in an increased resistance to chemotherapy [77]. In addition, preclinical evidence has suggested that the upregulation of miR-21 induced trastuzumab resistance due to the reduction of PTEN gene expression [78,79,80]. Another possible biological mechanism of resistance is PI3K pathway dysregulation by miR-21 and EMT activation with the triggering of IL-6/STAT3/NF-kB-mediated signaling [81]. A study by Liu et al on circulating miR-21 (ser-mir21) in HER2-positive early BC patients treated with neoadjuvant chemotherapy plus trastuzumab showed a significant correlation between changes in ser-miR-21 expression and treatment sensitivity [78]. The authors reported a significant reduction of expression of ser-miR-21 levels at the end of the second cycle and at the end of neoadjuvant treatment compared to the baseline value (before start of treatment), mainly shown in clinical responders (p < 0.001). These results were confirmed by Rodríguez-Martínez et al., who observed lower levels of miR-21 in HER2-positive patients during neoadjuvant treatment with trastuzumab [79]. They also highlighted a significant association between higher levels of exosomal miR-21 and circulating tumor cells [79]. In a study by Jung et al. on plasma samples of BC patients who underwent neoadjuvant trastuzumab-based chemotherapy, levels of miR-210 inversely correlated with treatment response. Higher levels of miR-201 have been reported in patients with residual disease compared to those with pathologically complete response (p = 0.0359) [82]. An in vitro study confirmed these data, showing a higher expression of miR-21 in trastuzumab-resistant BT474 cells [82]. miR-155 was identified as a negative predictive marker too. In a cohort of 175 BC patients, 107 early stage and 68 in the metastatic setting, the upregulation of miR-155 was related to poor trastuzumab sensitivity [83].
On the contrary, miR-148a-3p and miR-205 seem to be positive predictive biomarkers for anti-HER2-targeted therapies. In particular, the early blood detection of miR-148a-3p seems to identify early responsive patients to neoadjuvant anti-HER2 treatments [100]. Indeed, the study by Di Cosimo et al. demonstrated a strong correlation between miR-148a-3p and a pathologically complete response in a BC population treated with primary trastuzumab (p = 0.008) [100]. Consistently, in vitro studies reported that miR-148a over-expression inhibits BC cell proliferation through inhibition of the MAPK/ERK signaling pathways by direct targeting of ERBB3 genes and angiogenesis inhibition [98]. Preclinical evidence has shown how the miR-205 restores a pro-apoptotic activity. In BC tissues, miR-205 directly targets HER3 receptor and inhibits the AKT-mediated survival pathway, increasing the responsiveness to both tyrosine kinase inhibitors (such as Lapatinb and Gefitinb) and trastuzumab [47,101]. Finally, a recent preclinical study showed a significant dysregulation of four miRNAs (miR-23b-3p, miR-195-5p, miR-656-5p, miR-340-5p) in HER2-positive BC-resistant BT-474 cells, suggesting potential involvement of these miRNAs in trastuzumab resistance mechanisms [91].

5. Prognostic Biomarkers

As well as the predictive role of miRNAs being widely investigated, their prognostic value has also been studied (Table 3). The most widely reported miRNAs as negative prognostic biomarkers are: miR-21, miR-155, miR-150-5p, and miR-4734. miR-21 demonstrated an important prognostic role in HER2-positive early BC patients receiving neoadjuvant chemotherapy combined with trastuzumab [78]. In particular, Liu et al. demonstrated a better OS and disease-free survival (DFS) in patients with decreased ser-miR-21 from the start to the end of neoadjuvant treatment [78]. Higher levels seemed to be associated with larger tumor size and high Ki67 expression, with a statistically significant correlation with higher stage (p = 0.008) and tumor grade (p = 0.005). In particular, HER2-positive BC subtype (p = 0.002) and negative estrogen receptor cancer (p = 0.002) had a significantly higher expression of miR-21 [41]. Moreover, in a study on serum samples from 127 patients with early HER2-positive BC undergoing neoadjuvant treatment, increased levels of circulating miR-21 before and after chemotherapy showed a significant association with poor OS [84]. In early BC, higher miR-155 expression showed a strong correlation with poor event-free survival too, in univariate and multivariate analyses [83]. Du et al. identified two intracellular miRNAs, miR-150-5p and miR-4734, as reliable prognostic biomarkers predicting development of recurrence disease in HER2-positive BC after adjuvant trastuzumab-based treatment [102].
Contrasting clinical roles have been reported for miR-125. In particular, miR-125-b was identified as a negative prognostic factor, associated with worse OS in HER2-positive BC patients [66]. On the contrary, miR-125a-5p was a positive prognostic factor. A low expression level was found in the serum of patients with shorter survival compared to long-term patients. It also had a negative correlation with tumor grade (p = 0.004), lymph-node status (p = 0.004), and tumor size (p < 0.001). [103].
Both miR-205 and miR-148a seem to be positive prognostic factors. In a clinical study on 52 HER2-positive BC patients treated with adjuvant trastuzumab, miR-205 expression on sample tissues significantly correlated to better DFS (p = 0.00168) [101]. In addition, in cell lines, lower levels of miR-205 were correlated to high histological grade biopsies and higher invasion rate [104]. In preclinical evidence, miR-148a inhibits BC cell migration and invasion, through wnt-βcatenin pathway inhibition [105] and MMP-13 downregulation [106], resulting in a pro-apoptotic activity [107]. Among positive prognostic factors, miR-145 has been reported by Quan et al. as downregulated in tumor samples more than in paracancerous tissue [108]. In agreement with its oncosuppressive function, higher miR-145 levels seem to be associated with a better survival rate than the lower expression group (p < 0.028) [48,108].
As expected, due to the close connection between predictive and prognostic value, some miRNAs overlap between Table 1 and Table 2, specifically, miR-210, miR-205, miR-21, miR-125b, and miR-148. High expression levels of miR-210, miR-21, and miR-125b correlate to trastuzumab and/or chemotherapy resistance and to poor survival, accordingly [66,75,78,82,84]. On the contrary, high expression of miR-205 and miR-148 correlated to an increase in sensitivity to cytotoxic treatments and a higher number of pathologically complete responses at the end of the neoadjuvant therapy, with an improvement in OS [104,105,106,107].

6. Conclusions

Emerging evidence highlights the key role in cell-to-cell communication and tumorigenesis of miRNAs. Due to their easy availability in blood and tumor tissue, they represent the optimal biomarker in oncology. Actually, a great deal of effort has been put into basic and clinical research to isolate a signature of miRNAs with predictive and prognostic value. This issue is particularly important in patients treated with targeted therapies, such as HER2-positive BC therapies. Data from our review identified a panel of miRNAs expressed in HER2-positive BC potentially able to predict treatment and survival outcomes. Unfortunately, available data are not enough to define whether some miRNAs are better predictors than others. In the available studies, the miRNA level changes were evaluated in different ways, using fold change in some cases and p-value in others, which made a comparison between the studies difficult. Moreover, further investigations would be necessary to properly define and validate these molecular tools as clinical biomarkers. Despite this, our literature review identified a triplet of miRNAs (155, 125b, 21), typically expressed in HER2-positive BC, that seem to be valid negative predictive and prognostic biomarkers. Available evidence is promising but requires further data from clinical trials. A future development could be the periodic assessment of a panel of HER2-specific miRNAs in patients on neoadjuvant treatment to enable early prediction of treatment resistance and to tailor patient management.

Author Contributions

C.O., M.D. and F.P., conceptualization. C.I., M.B., A.T. and C.P., writing—original draft preparation and review and editing. F.C., I.M. and V.M., original draft preparation and visualization. C.O., M.M. and L.M., supervision. M.D., project administration. All authors have read and agreed to the published version of the manuscript.

Funding

The authors received no financial support for the research.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BCbreast cancer
HER2human epidermal growth factor receptor-2
miRNAmicroRNA
EVsextracellular vesicles
OSoverall survival
DFSdisease-free survival

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Cronin, K.A.; Harlan, L.C.; Dodd, K.W.; Abrams, J.S.; Ballard-Barbash, R. Population-based Estimate of the Prevalence of HER-2 Positive Breast Cancer Tumors for Early Stage Patients in the US. Cancer Investig. 2010, 28, 963–968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Swain, S.M.; Miles, D.; Kim, S.-B.; Im, Y.-H.; Im, S.-A.; Semiglazov, V.; Ciruelos, E.; Schneeweiss, A.; Loi, S.; Monturus, E.; et al. Pertuzumab, Trastuzumab, and Docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): End-of-study results from a double-blind, randomised, placebo-controlled, phase 3 study. Lancet Oncol. 2020, 21, 519–530. [Google Scholar] [CrossRef]
  4. Piccart-Gebhart, M.J.; Procter, M.; Leyland-Jones, B.; Goldhirsch, A.; Untch, M.; Smith, I.; Gianni, L.; Baselga, J.; Bell, R.; Jackisch, C.; et al. Trastuzumab after Adjuvant Chemotherapy in HER2-positive Breast cancer. N. Engl. J. Med. 2005, 353, 1659–1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Romond, E.H.; Perez, E.A.; Bryant, J.; Suman, V.J.; Geyer, C.E.; Davidson, N.E.; Tan-Chiu, E.; Martino, S.; Paik, S.; Kaufman, P.A.; et al. Trastuzumab plus Adjuvant chemotherapy for Operable HER2-Positive Breast cancer. N. Engl. J. Med. 2005, 353, 1673–1684. [Google Scholar] [CrossRef] [Green Version]
  6. Cameron, D.; Piccart-Gebhart, M.J.; Gelber, R.D.; Procter, M.; Goldhirsch, A.; de Azambuja, E.; Castro Jr, G.; Untch, M.; Smith, I.; Gianni, L.; et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: Final analysis of the HERceptin Adjuvant (HERA) trial. Lancet 2017, 389, 1195–1205. [Google Scholar] [CrossRef] [Green Version]
  7. Perez, E.A.; Romond, E.H.; Suman, V.J.; Jeong, J.-H.; Sledge, G.; Geyer Jr, C.E.; Martino, S.; Rastogi, P.; Gralow, J.; Swain, S.M.; et al. Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: Planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J. Clin. Oncol. 2014, 32, 3744–3752. [Google Scholar] [CrossRef]
  8. Slamon, D.; Eiermann, W.; Robert, N.; Pienkowski, T.; Martin, M.; Press, M.; Mackey, J.; Glaspy, J.; Chan, A.; Pawlicki, M.; et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N. Engl. J. Med. 2011, 365, 1273–1283. [Google Scholar] [CrossRef] [Green Version]
  9. Von Minckwitz, G.; Huang, C.-S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef]
  10. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.-Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [Green Version]
  11. Krop, I.E.; Kim, S.-B.; Gonzáles-Martín, A.; LoRusso, P.M.; Ferrero, J.-M.; Smitt, M.; Yu, R.; Leung, A.C.F.; Wildiers, H. Trastuzumab emtansine versus treatment of physician’s choice for pretreated HER2-positive advanced breast cancer (TH3RESA): A randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 689–699. [Google Scholar] [CrossRef]
  12. Modi, S.; Saura, C.; Yamashita, T.; Park, Y.H.; Kim, S.-B.; Tamura, K.; Andre, F.; Iwata, H.; Ito, Y.; Tsurutani, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N. Engl. J. Med. 2020, 382, 610–621. [Google Scholar] [CrossRef]
  13. Vernieri, C.; Milano, M.; Brambilla, M.; Mennitto, A.; Maggi, C.; Cona, M.S.; Prisciandaro, M.; Fabbroni, C.; Celio, L.; Mariani, G.; et al. Resistance mechanisms to anti-HER2 therapies in HER2-positive breast cancer: Current knowledge, new research directions and therapeutic perspectives. Crit. Rev. Oncol. Hematol. 2019, 139, 53–66. [Google Scholar] [CrossRef] [PubMed]
  14. Gianni, L.; Pienkowski, T.; Im, Y.-H.; Roman, L.; Tseng, L.-M.; Liu, M.-C.; Lluch, A.; Staroslawska, E.; de la Haba-Rodriguez, J.; Im, S.-A.; et al. Efficacy and Safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced inflammatory or Early HER2 positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012, 13, 25–32. [Google Scholar] [CrossRef]
  15. Schneeweiss, A.; Chia, S.; Hickish, T.; Harvey, V.; Eniu, A.; Hegg, R.; Tausch, C.; Seo, J.H.; Tsai, Y.-F.; Ratnayake, J.; et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: A randomized phase II cardiac safety study (TRYPHAENA). Ann. Oncol. 2013, 24, 2278–2284. [Google Scholar] [CrossRef] [PubMed]
  16. Hannafon, B.N.; Trigoso, Y.D.; Calloway, C.L.; Zhao, Y.D.; Lum, D.H.; Welm, A.L.; Zhao, Z.J.; Blick, K.E.; Dooley, W.C.; Ding, W.Q. Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Res. 2016, 18, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Shimomura, A.; Shiino, S.; Kawauchi, J.; Takizawa, S.; Sakamoto, H.; Matsuzaki, J.; Ono, M.; Takeshita, F.; Niida, S.; Shimizu, C.; et al. Novel combination of serum microRNA for detecting breast cancer in the early stage. Cancer Sci. 2016, 107, 326–333. [Google Scholar] [CrossRef] [PubMed]
  18. Quan, J.; Liu, S.; Dai, K.; Jin, L.; He, T.; Pan, X.; Lai, Y. Micro-RNA-23a/24–2/27a as a potential diagnostic biomarker for cancer: A systematic review and meta-analysis. Mol. Clin. Oncol. 2018, 8, 159–169. [Google Scholar]
  19. Hannafon, B.N.; Ding, W.-Q. Intercellular communication by exosome-derived microRNAs in cancer. Int. J. Mol. Sci. 2013, 14, 14240–14269. [Google Scholar] [CrossRef] [Green Version]
  20. Piombino, C.; Mastrolia, I.; Omarini, C.; Candini, O.; Dominici, M.; Piacentini, F.; Toss, A. The Role of Exosomes in Breast Cancer Diagnosis. Biomedicines 2021, 9, 312. [Google Scholar] [CrossRef]
  21. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
  22. Wu, H.; Wang, Q.; Zhong, H.; Li, L.; Zhang, Q.; Huang, Q.; Yu, Z. Differentially expressed microRNAs in exosomes of patients with breast cancer revealed by next-generation sequencing. Oncol. Rep. 2020, 43, 240–250. [Google Scholar] [CrossRef]
  23. Kalra, H.; Drummen, G.P.C.; Mathivanan, S. Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int. J. Mol. Sci. 2016, 17, 170. [Google Scholar] [CrossRef] [Green Version]
  24. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  25. Yánez-Mo’, M.; Siljande, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  26. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
  27. Kormelink, T.G.; Mol, S.; de Jong, E.C.; Wauben, M.H.M. The role of extracellular vesicles when innate meets adaptive. Semin. Immunopathol. 2018, 40, 439–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Peinado, H.; AlečKović, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Joyce, D.P.; Kerin, M.J.; Dwyer, R.M. Exosome-encapsulated microRNAs as circulating biomarkers for breast cancer. Int. J. Cancer 2016, 139, 1443–1448. [Google Scholar] [CrossRef] [PubMed]
  30. Thierry, A.R.; Messaoudi, S.E.I.; Gahan, P.B.; Anker, P.; Stroun, M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016, 35, 347–376. [Google Scholar] [CrossRef] [Green Version]
  31. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  32. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  33. Liu, C.G.; Calin, G.A.; Meloon, B.; Gamliel, N.; Sevignani, C.; Ferracin, M.; Dumitru, C.D.; Shimizu, M.; Zupo, S.; Dono, M.; et al. An oligonucleotide microchip for genome wide miRNA profiling in human and mouse tissues. Proc. Natl. Acad. Sci. USA 2004, 101, 9740–9744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, C.; Ridzon, D.A.; Broomer, A.J.; Zhou, Z.; Lee, D.H.; Nguyen, J.T.; Barbisin, M.; Xu, N.L.; Mahuvakar, V.R.; Andersen, M.R.; et al. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005, 33, e179. [Google Scholar] [CrossRef] [PubMed]
  36. Cummins, J.M.; He, Y.; Leary, R.J.; Pagliarini, R.; Diaz Jr, L.A.; Sjoblom, T.; Barad, O.; Bentwich, Z.; Szafranska, A.E.; Labourier, E.; et al. The colorectal microRNAome. Proc. Natl. Acad. Sci. USA 2006, 103, 3687–3692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Nelson, P.T.; Baldwin, D.A.; Scearce, L.M.; Oberholtzer, J.C.; Tobias, J.W.; Mourelatos, Z. Microarray-based, high-throughput gene expression profiling of microRNAs. Nat. Methods 2004, 1, 155–161. [Google Scholar] [CrossRef]
  38. Song, C.G.; Wu, X.Y.; Fu, F.-M.; Han, Z.-H.; Wang, C.; Shao, Z.-M. Correlation of miR-155 on formalin-fixed paraffin embedded tissues with invasiveness and prognosis of breast cancer. Zhonghua Wai Ke Za Zhi 2012, 50, 1011–1014. [Google Scholar]
  39. Jiang, S.; Zhang, H.-W.; Lu, M.-H.; He, X.-H.; Li, Y.; Gu, H.; Liu, M.-F.; Wang, E.-D. MicroRNA-155 functions as an OncomiR in breast cancer by targeting the suppressor of cytokine signaling 1 gene. Cancer Res. 2010, 70, 3119–3122. [Google Scholar] [CrossRef] [Green Version]
  40. Nami, B.; Wang, Z. HER2 in Breast Cancer Stemness: A Negative Feedback Loop towards Trastuzumab Resistance. Cancers 2017, 9, 40. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, J.A.; Lee, H.Y.; Lee, E.S.; Kim, I.; Bae, J.W. Prognostic Implications of MicroRNA-21 Overexpression in Invasive Ductal Carcinomas of the Breast. J. Breast Cancer 2011, 14, 269–275. [Google Scholar] [CrossRef] [Green Version]
  42. Sempere, L.F.; Christensen, M.; Silahtaroglu, A.; Bak, M.; Heath, C.V.; Schwartz, G.; Wells, W.; Kauppinen, S.; Cole, C.N. Altered MicroRNA Expression Confined to Specific Epithelial Cell Subpopulations in Breast Cancer. Cancer Res. 2007, 67, 11612–11620. [Google Scholar] [CrossRef] [Green Version]
  43. Huang, T.-H.; Wu, F.; Loeb, G.B.; Hsu, R.; Heidersbach, A.; Brincat, A.; Horiuchi, D.; Lebbink, R.J.; Mo, Y.-Y.; Goga, A.; et al. Up-regulation of miR-21 by HER2/neu Signaling Promotes Cell Invasion. J. Biol. Chem. 2009, 284, 18515–18524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Xiao, Y.; Humphries, B.; Yang, C.; Wang, Z. MiR-205 Dysregulations in Breast Cancer: The Complexity and Opportunities. Non-Coding RNA 2019, 5, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Plantamura, I.; Cataldo, A.; Cosentino, G.; Iorio, M.V. miR-205 in Breast Cancer: State of the Art. Int. J. Mol. Sci. 2021, 22, 27. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, H.; Zhu, S.; Mo, Y.-Y. Suppression of cell growth and invasion by miR-205 in breast cancer. Cell Res. 2009, 19, 439–448. [Google Scholar] [CrossRef] [PubMed]
  47. Iorio, M.; Casalini, P.; Piovan, C.; Di Leva, G.; Merlo, A.; Triulzi, T.; Ménard, S.; Croce, C.M.; Tagliabue, E. microRNA-205 Regulates HER3 in Human Breast Cancer. Cancer Res. 2009, 69, 2195–2200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Patel, Y.; Shah, N.; Lee, J.S.; Markoutsa, E.; Jie, C.; Liu, S.; Botbyl, R.; Reisman, D.; Xu, P.; Chen, H. A novel double-negative feedback loop between miR-489 and the HER2-SHP2-MAPK signaling axis regulates breast cancer cell proliferation and tumor growth. Oncotarget 2016, 7, 18295–18308. [Google Scholar] [CrossRef] [Green Version]
  49. Savad, S.; Mahdipour, P.; Shirdast, H.; Nekoohesh, L.; Nekoohesh, L.; Shirkoohi, R.; Nikooee, V.; Miryounesi, M.; Modarresi, M.H. Expression of miR-520d in breast cancer. Basic Clin. Cancer Res. 2013, 5, 11–15. [Google Scholar]
  50. Tsukerman, P.; Yamin, R.; Seidel, E.; Khawaled, S.; Schmiedel, D.; Bar-Mag, T.; Mandelboim, O. MiR-520d-5p directly targets TWIST1 and downregulates the metastamiR miR-10b. Oncotarget 2014, 5, 12141–12150. [Google Scholar] [CrossRef] [Green Version]
  51. Lowery, A.J.; Miller, N.; Devaney, A.; McNeill, R.E.; Davoren, P.A.; Lemetre, C.; Benes, V.; Schmidt, S.; Blake, J.; Ball, G.; et al. MicroRNA signatures predict oestrogen receptor, progesterone receptor and HER2/neu receptor status in breast cancer. Breast Cancer Res. 2009, 11, R27. [Google Scholar] [CrossRef] [PubMed]
  52. An, N.; Luo, X.; Zhang, M.; Yu, R. MicroRNA-376b promotes breast cancer metastasis by targeting Hoxd10 directly. Exp. Ther. Med. 2017, 13, 79–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Lindholm, E.M.; Leivone, S.K.; Undlien, E.; Nebdal, D.; Git, A.; Caldas, C.; Børresen-Dale, A.-L.; Kleivi, K. miR-342-5p As a Potential Regulator of HER2 Breast Cancer Cell Growth. MicroRNA 2019, 8, 155–165. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, X.; Somlo, G.; Yu, Y.; Palomares, M.R.; Li, A.X.; Zhou, W.; Chow, A.; Yen, Y.; Rossi, J.J.; Gao, H.; et al. De novo sequencing of circulating miRNAs identifies novel markers predicting clinical outcome of locally advanced breast cancer. J. Transl. Med. 2012, 10, 1–10. [Google Scholar] [CrossRef] [Green Version]
  55. Zou, Q.; Yi, W.; Huang, J.; Fu, F.; Chen, G.; Zhong, D. MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells. Exp. Ther. Med. 2017, 14, 1198–1204. [Google Scholar] [CrossRef] [Green Version]
  56. Ye, X.M.; Zhu, H.Y.; Bai, W.-D.; Wang, T.; Wang, L.; Chen, Y.; Yang, A.-G.; Jia, L.-T. Epigenetic silencing of miR-375 induces trastuzumab resistance in HER2-positive breast cancer by targeting IGF1R. BMC Cancer 2014, 14, 1–12. [Google Scholar] [CrossRef] [Green Version]
  57. Tashkandi, H.; Shah, N.; Patel, Y.; Chen, H. Identification of new miRNA biomarkers associated with HER2-positive breast cancers. Oncoscience 2015, 2, 924–929. [Google Scholar] [CrossRef] [Green Version]
  58. Han, B.; Huang, J.; Han, Y.; Hao, J.; Wu, X.; Song, H.; Chen, X.; Shen, Q.; Dong, X.; Pang, H.; et al. The microRNA miR-181c enhances chemosensitivity and reduces chemoresistance in breast cancer cells via down-regulating osteopontin. Int. J. Biol. Macromol. 2019, 125, 544–556. [Google Scholar] [CrossRef]
  59. Zhao, L.; Wang, Y.; Jiang, L.; He, M.; Bai, X.; Yu, L.; Wei, M. MiR-302a/b/c/d cooperatively sensitizes breast cancer cells to adriamycin via suppressing P-glycoprotein(P-gp) by targeting MAP/ERK kinase kinase 1 (MEKK1). J. Exp. Clin. Cancer Res. 2016, 35, 1–14. [Google Scholar] [CrossRef] [Green Version]
  60. Xi, C.; Wang, J.; Sun, H.; Zhang, X.; Kang, H. Loss of microRNA-30e induced by extracellular vesicles from cancer-associated fibroblasts promotes breast cancer progression by binding to CTHRC1. Exp. Mol. Pathol. 2021, 118, 104586. [Google Scholar] [CrossRef]
  61. Shizhen, E.W.; Lin, R.-J. MicroRNA and HER2-overexpressing Cancer. MicroRNA 2013, 2, 137–147. [Google Scholar]
  62. Fontana, A.; Barbano, R.; Dama, E.; Pasculli, B.; Rendina, M.; Morritti, M.G.; Melocchi, V.; Castelvetere, M.; Valori, V.M.; Ravaioli, S.; et al. Combined analysis of miR-200 family and its significance for breast cancer. Sci. Rep. 2021, 11, 1–4. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, J.; Mao, Q.; Liu, Y.; Hao, X.; Zhang, S.; Zhang, J. Analysis of miR-205 and miR-155 expression in the blood of breast cancer patients. Chin. J. Cancer Res. 2013, 25, 46–54. [Google Scholar] [PubMed]
  64. Zhang, H.; Li, B.; Zhao, H.; Chang, J. The expression and clinical significance of serum miR-205 for breast cancer and its role in detection of human cancers. Int. J. Clin. Exp. Med. 2015, 8, 3034–3043. [Google Scholar]
  65. Ergün, S.; Ulasli, M.; Igci, Y.Z.; Igci, M.; Kırkbes, S.; Borazan, E.; Balik, A.; Yumrutaş, Ö.; Camci, C.; Cakmak, E.A.; et al. The association of the expression of miR-122-5p and its target ADAM10 with human breast cancer. Mol. Biol. Rep. 2015, 42, 497–505. [Google Scholar] [CrossRef]
  66. Luo, Y.; Wang, X.; Niu, W.; Wang, H.; Wen, Q.; Fan, S.; Zhao, R.; Li, Z.; Xiong, W.; Peng, S.; et al. Elevated microRNA-125b levels predict a worse prognosis in HER2-positive breast cancer patients. Oncol. Lett. 2017, 13, 867–874. [Google Scholar] [CrossRef]
  67. Uhlmann, S.; Mannsperge, H.; Zhang, J.D.; Horvat, E.Á.; Schmidt, C.; Küblbeck, M.; Henjes, F.; Ward, A.; Tschulena, U.; Zweig, K.; et al. Global microRNA level regulation of EGFR-driven cell-cycle protein network in breast cancer. Mol. Syst. Biol. 2012, 8, 570. [Google Scholar] [CrossRef]
  68. Fu, Y.; Xiong, J. MicroRNA-124 enhances response to radiotherapy in human epidermal growth factor receptor 2-positive breast cancer cells by targeting signal transducer and activator of transcription 3. Croat. Med. J. 2016, 57, 457–464. [Google Scholar] [CrossRef]
  69. Nakanishi, H.; Taccioli, C.; Palatini, J.; Fernandez-Cymering, C.; Cui, R.; Kim, T.; Violinia, S.; Croce, C.M. Loss of miR-125b-1 contributes to head and neck cancer development by dysregulating TACSTD2 and MAPK pathway. Oncogene 2014, 33, 702–712. [Google Scholar] [CrossRef] [Green Version]
  70. Xia, H.-F.; He, T.-Z.; Liu, C.-M.; Cui, Y.; Song, P.-P.; Jin, X.-H.; Ma, X. MiR-125b Expression Affects the Proliferation and Apoptosis of Human Glioma Cells by Targeting Bmf. Cell Physiol. Biochem. 2009, 23, 347–358. [Google Scholar] [CrossRef]
  71. Le, M.T.N.; Teh, C.; Shyh-Chang, N.G.; Xie, H.; Zhou, B.; Korzh, V.; Lodish, H.F.; Lim, B. MicroRNA-125b is a novel negative regulator of p53. Genes Dev. 2009, 23, 862–876. [Google Scholar] [CrossRef] [Green Version]
  72. Banzhaf-Strathmann, J.; Edbauer, D. Good guy or bad guy: The opposing roles of microRNA 125b in cancer. Cell Commun. Signal. 2014, 12, 1–13. [Google Scholar] [CrossRef] [Green Version]
  73. Tang, F.; Zhang, R.; He, Y.; Zou, M.; Guo, L.; Xi, T. MicroRNA-125b Induces Metastasis by Targeting STARD13 in MCF-7 and MDA-MB-231 Breast Cancer Cells. PLoS ONE 2012, 7, e35435. [Google Scholar] [CrossRef]
  74. Scott, G.K.; Goga, A.; Bhaumik, D.; Berger, C.E.; Sullivan, C.S.; Benz, C.C. Coordinate Suppression of ERBB2 and ERBB3 by Enforced Expression of Micro-RNA miR-125a or miR-125b. J. Biol. Chem. 2007, 282, 1479–1486. [Google Scholar] [CrossRef] [Green Version]
  75. Wang, H.; Tan, G.; Dong, L.; Cheng, L.; Li, K.; Wang, Z.; Luo, H. Circulating MiR-125b as a Marker Predicting Chemoresistance in Breast Cancer. PLoS ONE 2012, 7, e34210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zhou, M.; Liu, Z.; Zhao, Y.; Ding, Y.; Liu, H.; Xi, Y.; Xiong, W.; Li, G.; Lu, J.; Fodstad, O.; et al. MicroRNA-125b Confers the Resistance of Breast Cancer Cells to Paclitaxel through Suppression of Pro-apoptotic Bcl-2 Antagonist Killer 1 (Bak1) Expression. J. Biol. Chem. 2010, 285, 21496–21507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, H.-J.; Guo, Y.-Q.; Tan, G.; Dong, L.; Cheng, L.; Li, K.-J.; Wang, Z.-Y.; Luo, H.-F. miR-125b Regulates Side Population in Breast Cancer and Confers a Chemoresistant Phenotype. J. Cell. Biochem. 2013, 114, 2248–2257. [Google Scholar] [CrossRef] [PubMed]
  78. Liu, B.; Su, F.; Lv, X.; Zhang, W.; Shang, X.; Zhang, Y.; Zhang, J. Serum microRNA-21 predicted treatment outcome and survival in HER2-positive breast cancer patients receiving neoadjuvant chemotherapy combined with trastuzumab. Cancer Chemother. Pharmacol. 2019, 84, 1039–1049. [Google Scholar] [CrossRef]
  79. Rodríguez-Martínez, A.; de Miguel-Pérez, D.; Ortega, F.G.; García-Puche, G.L.; Robles-Fernández, I.; Exposito, J.; Martorell-Marugan, J.; Carmona-Sáez, P.; Garrido-Navas, M.D.C.; Rolfo, C.; et al. Exosomal miRNA profile as complementary tool in the diagnostic and prediction of treatment response in localized breast cancer under neoadjuvant chemotherapy. Breast Cancer Res. 2019, 21, 1–9. [Google Scholar] [CrossRef]
  80. Gong, C.; Yao, Y.; Wang, Y.; Liu, B.; Wu, W.; Chen, J.; Su, F.; Yao, H.; Song, E. Up-regulation of miR-21 Mediates Resistance to Trastuzumab Therapy for Breast Cancer. J. Biol. Chem. 2011, 286, 19127–19137. [Google Scholar] [CrossRef] [Green Version]
  81. De Mattos-Arruda, L.; Bottai, G.; Nuciforo, P.G.; Di Tommaso, L.; Giovannetti, E.; Peg, V.; Losurdo, A.; Pérez-Garcia, J.; Masci, G.; Corsi, F.; et al. MicroRNA-21 links epithelial-to-mesenchymal transition and inflammatory signals to confer resistance to neoadjuvant trastuzumab and chemotherapy in HER2-positive breast cancer patients. Oncotarget 2015, 6, 37269. [Google Scholar] [CrossRef] [Green Version]
  82. Jung, E.-J.; Santarpia, L.; Kim, J.; Esteva, F.J.; Moretti, E.; Buzdar, A.U.; Di Leo, A.; Le, X.-F.; Bast, R.C., Jr.; Park, S.-T.; et al. Plasma miR-210 levels correlate with sensitivity to trastuzumab and tumor presence in breast cancer patients. Cancer 2012, 118, 2603–2614. [Google Scholar] [CrossRef]
  83. Zhang, Z.; Zhang, L.; Yu, G.; Sun, Z.; Wang, T.; Tian, X.; Duan, X.; Zhang, C. Exosomal miR-1246 and miR-155 as predictive and prognostic biomarkers for trastuzumab-based therapy resistance in HER2-positive breast cancer. Cancer Chemother. Pharmacol. 2020, 86, 761–772. [Google Scholar] [CrossRef]
  84. Müller, V.; Gade, S.; Steinbach, B.; Loibl, S.; von Minckwitz, G.; Untch, M.; Schwedler, K.; Lübbe, K.; Schem, C.; Fasching, P.A.; et al. Changes in serum levels of miR-21, miR-210, and miR-373 in HER2 positive breast cancer patients undergoing neoadjuvant therapy: A translational research project within the Geparquinto trial. Breast Cancer Res. Treat. 2014, 147, 61–68. [Google Scholar] [CrossRef]
  85. Zhao, Q.; Liu, Y.; Wang, T.; Yang, Y.; Ni, H.; Liu, H.; Guo, Q.; Xi, T.; Zheng, L. MiR-375 inhibits the stemness of breast cancer cells by blocking the JAK2/STAT3 signaling. Eur. J. Pharmacol. 2020, 884, 173359. [Google Scholar] [CrossRef]
  86. Bockhorn, J.; Dalton, R.; Nwachukwu, C.; Huang, S.; Prat, A.; Yee, K.; Chang, Y.F.; Huo, D.; Wen, Y.; Swanson, K.E.; et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat. Commun. 2013, 4, 2393. [Google Scholar] [CrossRef] [Green Version]
  87. Fang, Y.; Shen, H.; Cao, Y.; Li, H.; Qin, R.; Chen, Q.; Long, L.; Zhu, X.L.; Xie, C.J.; Xu, W.L. Involvement of miR-30c in resistance to doxorubicin by regulating YWHAZ in breast cancer cells. Braz. J. Med. Biol. Res. 2014, 47, 60–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Tormo, E.; Adam-Artigues, A.; Ballester, S.; Pineda, B.; Zazo, S.; González-Alonso, P.; Albanell, J.; Rovira, A.; Rojo, F.; Lluch, A.; et al. The role of miR-26a and miR-30b in HER2+ breast cancer trastuzumab resistance and regulation of the CCNE2 gene. Sci. Rep. 2017, 7, 41309. [Google Scholar] [CrossRef] [PubMed]
  89. Ichikawa, T.; Sato, F.; Terasawa, K.; Tsuchiya, S.; Toi, M.; Tsujimoto, G.; Shimizu, K. Trastuzumab Produces Therapeutic Actions by Upregulating miR-26a and miR-30b in Breast Cancer Cells. PLoS ONE 2012, 7, e31422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Li, Z.H.; Weng, X.; Xiong, Q.Y.; Tu, J.H.; Xiao, A.; Qiu, W.; Gong, Y.; Hu, E.W.; Huang, S.; Cao, Y.L. miR-34a expression in human breast cancer is associated with drug resistance. Oncotarget 2017, 8, 106270–106282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Rezaei, Z.; Sebzari, A.; Kordi-Tamandani, D.M.; Dastjerdi, K. Involvement of the Dysregulation of miR-23b-3p, miR-195-5p, miR-656-5p, and miR-340-5p in Trastuzumab Resistance of HER2-Positive Breast Cancer Cells and System Biology Approach to Predict Their Targets Involved in Resistance. DNA Cell Biol. 2019, 38, 184–192. [Google Scholar] [CrossRef]
  92. Noyan, S.; Gurdal, H.; Gur Dedeoglu, B. Involvement of miR-770-5p in trastuzumab response in HER2 positive breast cancer cells. PLoS ONE 2019, 14, e0215894. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, F.; Fu, Z.; Yang, M.; Sun, C.; Li, Y.; Chu, J.; Zhang, Y.; Li, W.; Huang, X.; Li, J.; et al. Expression pattern of microRNAs related with response to trastuzumab in breast cancer. J. Cell Physiol. 2019, 234, 16102–16113. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Z.; Qin, Y.; Chen, P.; Luo, Q.; Shi, H.; Jiang, X. miR-135b-5p enhances the sensitivity of HER-2 positive breast cancer to trastuzumab via binding to cyclin D2. Int. J. Mol. Med. 2020, 46, 1514–1524. [Google Scholar] [PubMed]
  95. Li, Z.-H.; Xiong, Q.-Y.; Xu, L.; Duan, P.; Yang, Q.O.; Zhou, P.; Tu, J.H. miR-29a regulated ER-positive breast cancer cell growth and invasion and is involved in the insulin signaling pathway. Oncotarget 2017, 8, 32566–32575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Shen, H.; Li, L.; Yang, S.; Wang, D.; Zhong, S.; Zhao, J.; Tang, J. MicroRNA-29a contributes to drug-resistance of breast cancer cells to adriamycin through PTEN/AKT/GSK3β signaling pathway. Gene 2016, 593, 84–90. [Google Scholar] [CrossRef] [PubMed]
  97. Han, G.; Qiu, N.; Luo, K.; Liang, H.; Li, H. Downregulation of miroRNA-141 mediates acquired resistance to trastuzumab and is associated with poor outcome in breast cancer by upregulating the expression of ERBB4. J. Cell Biochem. 2019, 120, 11390–11400. [Google Scholar] [CrossRef] [PubMed]
  98. Yu, J.; Li, Q.; Xu, Q.; Liu, L.; Jiang, B. MiR-148a inhibits angiogenesis by targeting ERBB3. J. Biomed. Res. 2011, 25, 170–177. [Google Scholar] [CrossRef] [Green Version]
  99. Ye, X.; Bai, W.; Zhu, H.; Zhang, X.; Chen, Y.; Wang, L.; Yang, A.; Zhao, J.; Jia, L. MiR-221 promotes trastuzumab-resistance and metastasis in HER2-positive breast cancers by targeting PTEN. BMB Rep. 2014, 47, 268–273. [Google Scholar] [CrossRef] [Green Version]
  100. Di Cosimo, S.; Appierto, V.; Pizzamiglio, S.; Silvestri, M.; Baselga, J.; Piccart, M.; Huober, J.; Izquierdo, M.; de la Pena, L.; Hilbers, F.S.; et al. Early Modulation of Circulating MicroRNAs Levels in HER2-Positive Breast Cancer Patients Treated with Trastuzumab-Based Neoadjuvant Therapy. Int. J. Mol. Sci. 2020, 21, 1386. [Google Scholar] [CrossRef] [Green Version]
  101. Cataldo, A.; Piovan, C.; Plantamura, I.; D’Ippolito, E.; Camelliti, S.; Casalini, P.; Giussani, M.; Déas, O.; Cairo, S.; Judde, J.-G.; et al. MiR-205 as predictive biomarker and adjuvant therapeutic tool in combination with trastuzumab. Oncotarget 2018, 9, 27920–27928. [Google Scholar] [CrossRef] [Green Version]
  102. Du, F.; Yuan, P.; Zhao, Z.T.; Yang, Z.; Wang, T.; Zhao, J.D.; Luo, Y.; Ma, F.; Wang, J.Y.; Fan, Y.; et al. A miRNA-based signature predicts development of disease recurrence in HER2 positive breast cancer after adjuvant trastuzumab-based treatment. Sci. Rep. 2016, 6, 35509. [Google Scholar] [CrossRef] [Green Version]
  103. Hsieh, T.-H.; Hsu, C.-Y.; Tsai, C.-F.; Long, C.-Y.; Chai, C.-Y.; Hou, M.-F.; Lee, J.N.; Wu, D.-C.; Wang, S.-C.; Tsai, E.-M. miR-125a-5p is a prognostic biomarker that targets HDAC4 to suppress breast tumorigenesis. Oncotarget 2014, 6, 494. [Google Scholar] [CrossRef] [Green Version]
  104. Stankevicins, L.; Barat, A.; Dessen, P.; Vassetzky, Y.; de Moura Gallo, C.V. The microRNA-205-5p is correlated to metastatic potential of 21T series: A breast cancer progression model. PLoS ONE 2017, 12, e0173756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Jiang, Q.; He, M.; Ma, M.-T.; Wu, H.-Z.; Yu, Z.-J.; Guan, S.; Jiang, L.-Y.; Wang, Y.; Zheng, D.-D.; Jin, F.; et al. MicroRNA-148a inhibits breast cancer migration and invasion by directly targeting WNT-1. Oncol Rep. 2016, 35, 1425–1432. [Google Scholar] [CrossRef]
  106. Xue, J.; Chen, Z.; Gu, X.; Zhang, Y.; Zhang, W. MicroRNA-148a inhibits migration of breast cancer cells by targeting MMP-13. Tumour Biol. 2016, 37, 1581–1590. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, L.; Xing, M.; Wang, X.; Cao, W.; Wang, H. MiR-148a suppresses invasion and induces apoptosis of breast cancer cells by regulating USP4 and BIM expression. Int. J. Clin. Exp. Pathol. 2017, 10, 8361–8368. [Google Scholar]
  108. Quan, Y.; Huang, X.; Quan, X. Expression of miRNA-206 and miRNA-145 in breast cancer and correlation with prognosis. Oncol. Lett. 2018, 16, 6638–6642. [Google Scholar] [CrossRef] [PubMed]
  109. Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.; Li, S.; Chin, A.R.; et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Miao, Y.; Wang, L.; Zhang, X.; Xing, R.G.; Zhou, W.W.; Liu, C.R.; Zhang, X.L.; Tian, L. miR-30a inhibits breast cancer progression through the Wnt/β-catenin pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 241–250. [Google Scholar]
  111. Zhang, J.; Yang, J.; Zhang, X.; Xu, J.; Sun, Y.; Zhang, P. MicroRNA-10b expression in breast cancer and its clinical association. PLoS ONE 2018, 13, e0192509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Liu, Y.; Zhao, J.; Zhang, P.Y.; Zhang, Y.; Sun, S.Y.; Yu, S.Y.; Xi, Q.S. MicroRNA-10b targets E-cadherin and modulates breast cancer metastasis. Med. Sci. Monit. 2012, 18, BR299–BR308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Huang, Q.; Gumireddy, K.; Schrier, M.; le Sage, C.; Nagel, R.; Nair, S.; Egan, D.A.; Li, A.; Huang, G.; Klein-Szanto, A.J.; et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat. Cell Biol. 2008, 10, 202–210. [Google Scholar] [CrossRef] [PubMed]
  114. Ding, Y.; Zhang, C.; Zhang, J.; Zhang, N.; Li, T.; Fang, J.; Zhang, Y.; Zuo, F.; Tao, Z.; Tang, S.; et al. miR-145 inhibits proliferation and migration of breast cancer cells by directly or indirectly regulating TGF-β1 expression. Int. J. Oncol. 2017, 50, 1701–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Zhao, H.; Kang, X.; Xia, X.; Wo, L.; Gu, X.; Hu, Y.; Xie, X.; Chang, H.; Lou, L.; Shen, X. miR-145 suppresses breast cancer cell migration by targeting FSCN-1 and inhibiting epithelial-mesenchymal transition. Am. J. Transl. Res. 2016, 8, 3106–3114. [Google Scholar]
  116. Persson, H.; Kvist, A.; Rego, N.; Staaf, J.; Vallon-Christersson, J.; Luts, L.; Loman, N.; Jonsson, G.; Naya, H.; Hoglund, M.; et al. Identification of New MicroRNAs in Paired Normal and Tumor Breast Tissue Suggests a Dual Role for the ERBB2/Her2 Gene. Cancer Res. 2011, 71, 78–86. [Google Scholar] [CrossRef] [Green Version]
  117. Lu, Q.; Guo, Z.; Qian, H. Role of microRNA-150-5p/SRCIN1 axis in the progression of breast cancer. Exp. Ther. Med. 2019, 17, 2221–2229. [Google Scholar] [CrossRef]
  118. Li, F. Expression of miR-221 and miR-489 in breast cancer patients and their relationship with prognosis. Oncol Lett. 2020, 19, 1523–1529. [Google Scholar] [CrossRef] [PubMed]
  119. Leivonen, S.K.; Sahlberg, K.K.; Mäkelä, R.; Due, E.U.; Kallioniemi, O.; Børresen-Dale, A.L.; Perälä, M. High-throughput screens identify microRNAs essential for HER2 positive breast cancer cell growth. Mol. Oncol. 2014, 8, 93–104. [Google Scholar] [CrossRef] [PubMed]
Table 1. HER2-positive breast cancer miRNA expression.
Table 1. HER2-positive breast cancer miRNA expression.
miRNAExpression LevelGene Target #ActionEvidenceSampleReferences
miR-489downSMAD3, SHP2, HER2Cell growth, invasion and EMT inhibition (decreased expression of HER2)clinical/preclinicaltissue[48]
miR-520d downTWIST1Restoration of E-cadherine expression (reduction in invasiveness)clinical/preclinical tissue[49,50]
miR-376b upHoxd10Angiogenesisclinical/preclinicaltissue[51,52]
miR-342-5pdownERK, MAPK, SAPK/JNKHER2 downstream inhibiting effectpreclinicaltissue[53]
miR-375up/downPAX6, IGF1RTumor suppressor (HER2, HR *-negative and inflammatory BC §-related)clinical/preclinicaltissue/blood[54,55,56]
miR-155 upSOCS1, JAK2/STAT3, FOXO3HER2-positive status and TNBC correlation (cell proliferation)clinical/preclinicaltissue[38,39]
miR-181c downOPNIncreased HER2-positive signature, enhanced chemosensitivityclinical/preclinicaltissue[51,57,58]
miR-302cupMEKK1Increased HER2-positive signature, enhanced chemosensitivity clinical/preclinicaltissue[51,59]
miR-30e-3pup/downCTHRC1Increased HER2-positive signature, proliferation inhibition, and apoptosisclinical/preclinicaltissue[51,60]
miR-200up/downEGFR, SIRT1HER2-positive and HR * negative status association clinical/preclinicaltissue[61,62]
miR-205downEGFR, VEGF-A, HER3, ZEB1, SIP1, E2FTumor suppressor (cell cycle arrest and apoptosis)clinical/preclinicaltissue/blood[45,63,64]
miR-122-5pupADAM10Tumor suppressor (cell cycle arrest and apoptosis)preclinical/clinicaltissue[65]
miR-125bupERBB2,BAK1, BCL-2, STARD13Increased migration and invasionclinicaltissue[66]
miR-125a-5pdownEGFR family, AKT-ERKTumor suppressor (invasion reduction)clinical/preclinicaltissue[61]
miR-21 upPDCD4, PTEN, TPM1, MAPK, EGFR family HER2/neu upregulation correlation and apoptosis inhibitionclinical/preclinicaltissue[41,61]
miR-147downEGFR1, AKT2, CDK4, RB1EGFR-driven cell-cycle (cell proliferation inhibition)preclinicaltissue[67]
miR-124downEGFR1, STAT3Cell growth and differentiation inhibitionpreclinicaltissue[68]
# Gene targets included in the table were identified through an in silico analysis; * HR: hormone receptors; § BC: breast cancer.
Table 2. Predictive value for treatment outcomes of miRNAs expressed in HER2-positive early breast cancer.
Table 2. Predictive value for treatment outcomes of miRNAs expressed in HER2-positive early breast cancer.
miRNAExpression LevelGene Target #ActionEvidenceSampleReferences
miR-210upRAD52, FGFR1, E2F3, ephrin A3, MET, IGFR1, MUC4Trastuzumab resistancepreclinical/clinicaltissue/blood[82,84]
miR-375downIGF1R, JAK2/STAT3Trastuzumab, tamoxifene, and adriamycin resistancepreclinical/clinicaltissue/blood[55,56,85]
miR-205upEGFR, VEGF-A, HER3, AKTIncreased sensitivity to anti-HER2 therapypreclinicaltissue[61]
miR-125upERBB2, BAK1, BCL-2, STARD13Increased sensitivity to anti-HER2 therapypreclinicaltissue[61]
miR-21 upNFKB, MAPK, PTENTrastuzumab and taxol resistancepreclinical/clinicaltissue/blood[78,79,80,81]
miR-125bupERBB2, BAK1, BCL-2Resistance to anthracycline/taxol-based therapiespreclinical/clinicaltissue/blood[75]
miR-30cupTWF1, IL-11, YWHAZIncreased sensitivity to taxol, doxorubicin and tamoxifene preclinicaltissue[86,87]
miR-26aupCCNE2Increased sensitivity to trastuzumabpreclinicaltissue[88]
miR-30bupCCNE2Increased sensitivity to trastuzumabpreclinicaltissue[88,89]
miR-34a downBCL-2, CCND1, NOTCH1RT sensitivity, multidrug resistance preclinical/clinicaltissue[90]
miR-122-5pupADAM10Increased sensitivity to trastuzumabclinicaltissue[65]
miR-23b-3pupPTENTrastuzumab resistancepreclinicaltissue[91]
miR-195-5pdownVEGFA, RAF 1, FGF2, AKT3, CCND1, MYBTrastuzumab resistancepreclinicaltissue[91]
miR-656-5pdownCREB1Trastuzumab resistancepreclinicaltissue[91]
miR-340-5pdownC-METTrastuzumab resistancepreclinicaltissue[91]
miR-770-5pupPI3K, MAPKPotentiated trastuzumab effectpreclinicaltissue[92]
miR-200bupPTENTrastuzumab resistancepreclinical/clinicaltissue/blood[93]
miR-135bupCyclinD2Increased sensitivity to trastuzumabpreclinical/clinicaltissue/blood[93,94]
miR-29aupIGF-1R, PTENTrastuzumab and adryamicin resistancepreclinical/clinicaltissue/blood[95,96]
miR-141downERBB4Trastuzumab resistancepreclinicaltissue[97]
miR-148upMAPK/ERK (ERBB3 genes)Increased pathological complete responseclinicalblood[98]
miR-155upSOCS1,STAT3Trastuzumab resistancepreclinical/clinicaltissue/blood[39,83]
miR-221upPTENTrastuzumab resistancepreclinicaltissue[99]
# Gene targets included in the table were identified through an in silico analysis.
Table 3. Prognostic value of miRNAs expressed in HER2-positive early breast cancer.
Table 3. Prognostic value of miRNAs expressed in HER2-positive early breast cancer.
miRNAExpression LevelPos/NegActionEvidenceSampleReferences
miR-21upnegReduction during treatment: better OS and DFS (high level in high histological grade and invasion rate)preclinical/clinicaltissue/blood[41,78,84]
miR-122upnegCirculating higher levels associated with metastasisclinicaltissue/blood[54,109]
miR-155upnegHigh levels predicts poor event free/progression free survivalclinicalblood[83]
miR-205upposLow level associated with worse prognosis (high level in high histological grade and invasion rate)preclinicaltissue[104]
miR-30aupposLow level associated with higher grade and TNM stageclinicaltissue[110]
miR-210UPnegReduced RFSpreclinicalblood[84]
miR-125bupnegWorse OSclinicaltissue[66]
miR-125a-5pupposLow levels associated with poor survival, high grade, lymph node status, and tumor sizepreclinical/clinicaltissue/blood[103]
miR-10bupnegCorrelation with tumor stage and lymph node positivitypreclinical/clinicaltissue[111,112]
miR-373upnegCorrelation with advanced clinical stagepreclinicaltissue/blood[84,113]
miR-148aupposMigration and invasion cancer cell inhibition (pro-apoptotic function)preclinicaltissue[105,106,107]
miR-145upposLow levels correlate with poor survival outcomespreclinical/clinicaltissue[108,114,115]
miR-4734upnegDisease recurrence (ERBB2 amplification)preclinical/clinicaltissue[102,116]
miR-150-5pupnegDisease recurrence (enhanced cell proliferation, invasion, and migration)preclinical/clinicaltissue[102,117]
miR-489 downnegLow level correlates with shorter OSclinicalblood[118]
miR-770-5pupposCell invasion and motility inhibitionpreclinicaltissue[92]
miR-342-5pupposBetter OS and increased time to progression (cell proliferation inhibition)preclinical/clinicaltissue[53,119]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Isca, C.; Piacentini, F.; Mastrolia, I.; Masciale, V.; Caggia, F.; Toss, A.; Piombino, C.; Moscetti, L.; Barbolini, M.; Maur, M.; et al. Circulating and Intracellular miRNAs as Prognostic and Predictive Factors in HER2-Positive Early Breast Cancer Treated with Neoadjuvant Chemotherapy: A Review of the Literature. Cancers 2021, 13, 4894. https://doi.org/10.3390/cancers13194894

AMA Style

Isca C, Piacentini F, Mastrolia I, Masciale V, Caggia F, Toss A, Piombino C, Moscetti L, Barbolini M, Maur M, et al. Circulating and Intracellular miRNAs as Prognostic and Predictive Factors in HER2-Positive Early Breast Cancer Treated with Neoadjuvant Chemotherapy: A Review of the Literature. Cancers. 2021; 13(19):4894. https://doi.org/10.3390/cancers13194894

Chicago/Turabian Style

Isca, Chrystel, Federico Piacentini, Ilenia Mastrolia, Valentina Masciale, Federica Caggia, Angela Toss, Claudia Piombino, Luca Moscetti, Monica Barbolini, Michela Maur, and et al. 2021. "Circulating and Intracellular miRNAs as Prognostic and Predictive Factors in HER2-Positive Early Breast Cancer Treated with Neoadjuvant Chemotherapy: A Review of the Literature" Cancers 13, no. 19: 4894. https://doi.org/10.3390/cancers13194894

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop