Next Article in Journal
Translational Application of Circulating DNA in Oncology: Review of the Last Decades Achievements
Next Article in Special Issue
Identification of a Diagnostic Set of Endomyocardial Biopsy microRNAs for Acute Cellular Rejection Diagnostics in Patients after Heart Transplantation Using Next-Generation Sequencing
Previous Article in Journal
Mechanisms Underlying Hepatitis C Virus-Associated Hepatic Fibrosis
Previous Article in Special Issue
miR-371a-3p, miR-373-3p and miR-367-3p as Serum Biomarkers in Metastatic Testicular Germ Cell Cancers Before, During and After Chemotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Theragnostic Relationship between Drug-Resistance Specific miRNA Expressions, Chemotherapeutic Resistance, and Sensitivity in Breast Cancer: A Systematic Review and Meta-Analysis

1
College of Health and Human Sciences, Charles Darwin University, Darwin 0810, Australia
2
Vellore Institute of Technology (VIT), School of Bio-Sciences and Technology, Vellore 632014, India
3
CHIRI, School of Pharmacy and Biomedical Research, Faculty of Health Sciences, Curtin University, Bently campus, Western Australia
4
North Terrace Campus, University of Adelaide, Adelaide 5005, Australia
5
Department of Biochemistry, Bharathiyar University, Coimbatore, Tamil Nadu 641046, India
6
National Heart Institute, New Delhi 110065, India
7
John Flynn Private Hospital, Genesis Cancer Care, Tugun 4224, Australia
8
Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
These authors have contributed equally.
Cells 2019, 8(10), 1250; https://doi.org/10.3390/cells8101250
Submission received: 26 July 2019 / Revised: 20 September 2019 / Accepted: 21 September 2019 / Published: 14 October 2019
(This article belongs to the Special Issue microRNA as Biomarker)

Abstract

:
Awareness of breast cancer has been increasing due to early detection, but the advanced disease has limited treatment options. There has been growing evidence on the role of miRNAs involved in regulating the resistance in several cancers. We performed a comprehensive systematic review and meta-analysis on the role of miRNAs in influencing the chemoresistance and sensitivity of breast cancer. A bibliographic search was performed in PubMed and Science Direct based on the search strategy, and studies published until December 2018 were retrieved. The eligible studies were included based on the selection criteria, and a detailed systematic review and meta-analysis were performed based on PRISMA guidelines. A random-effects model was utilised to evaluate the combined effect size of the obtained hazard ratio and 95% confidence intervals from the eligible studies. Publication bias was assessed with Cochran’s Q test, I2 statistic, Orwin and Classic fail-safe N test, Begg and Mazumdar rank correlation test, Duval and Tweedie trim and fill calculation and the Egger’s bias indicator. A total of 4584 potential studies were screened. Of these, 85 articles were eligible for our systematic review and meta-analysis. In the 85 studies, 188 different miRNAs were studied, of which 96 were upregulated, 87 were downregulated and 5 were not involved in regulation. Overall, 24 drugs were used for treatment, with doxorubicin being prominently reported in 15 studies followed by Paclitaxel in 11 studies, and 5 drugs were used in combinations. We found only two significant HR values from the studies (miR-125b and miR-4443) and our meta-analysis results yielded a combined HR value of 0.748 with a 95% confidence interval of 0.508–1.100; p-value of 0.140. In conclusion, our results suggest there are different miRNAs involved in the regulation of chemoresistance through diverse drug genetic targets. These biomarkers play a crucial role in guiding the effective diagnostic and prognostic efficiency of breast cancer. The screening of miRNAs as a theragnostic biomarker must be brought into regular practice for all diseases. We anticipate that our study serves as a reference in framing future studies and clinical trials for utilising miRNAs and their respective drug targets.

1. Introduction

Breast cancer is the most prevalent type of cancer in women worldwide [1]. This makes it a cause of increasing concern, and it is important to address this issue. It was estimated that 41,070 breast cancer deaths occurred in women during 2017 in the USA alone, making it the second-leading cause of cancer-related death in women [1]. A large number of breast cancer patients are from developing countries as compared to Western countries, mainly due to their increasing populations [2]. In developed countries, breast cancer is often diagnosed early and treated accordingly; developing countries have higher death rates due to delayed diagnosis and improper access to healthcare [2]. Regardless of this, in developed countries breast cancer is second to lung cancer for cancer-related deaths in women [2]. Asia has 44% of the world’s breast cancer deaths, with 39% of overall new breast cancer cases diagnosed [2]. In India, breast cancer has been ranked as the foremost cancer among the Indian female population [3]. Approximately 25% of female cancer cases in the country are breast cancer [4,5]. The rate of incidence was found to be 25.8 in 100,000 women, and the mortality rate was 12.7 per 100,000 women (2017 statistics) [3]. The highest rate of incidence was found to be in Delhi (41 per 100,000 women) followed by Chennai (37.9 per 100,000 women), Bangalore (34.4 per 100,000 women) and Thiruvananthapuram district (33.7 per 100,000 women) [3]. When the mortality-to-incidence ratio was analysed, it was found to reach 66 in rural registries and 8 in urban registries [3]. Another troubling concern about the scenario of breast cancer in India is the increased incidence of disease in younger Indian women (between the ages of 30 and 40) [3,4,5]. Presently, almost 48% of breast cancer patients in India are below 50 years of age [4,5]. There is an increasing trend of breast cancer in women between the ages of 25 to 40 in the past 25 years [4,5].
At present, breast cancer is classified into four types: (1) Luminal A (classical hormone-positive tumours); (2) Luminal B (hormone-positive with higher ki 67 and poorer prognosis); (3) Triple-negative (ER/PR/HER neu negative); and (4) Her 2 neu overexpressing [6,7]. Currently, several treatments are available for breast cancer, and these include: surgical resection [8], which is often followed by radiotherapy [9], hormone replacement therapy (differs in pre-menopausal and post-menopausal women) [10], targeted therapies [11], immunotherapy [12] and chemotherapeutic drugs [13]. There are a number of chemotherapeutic drugs that are commonly in use and have distinct mechanisms of action, such as anthracyclines (e.g., doxorubicin [14] and epirubicin [15]), taxanes (e.g., Paclitaxel [16,17], docetaxel [16]), alkylating agents (e.g., cyclophosphamide (CTX) [18], carboplatin [17]), trastuzumab—a monoclonal antibody targeted against Her 2 neu [17], anti-metabolites (e.g., 5-fluorouracil (5-FU)) [18], and hormonal agents (e.g., tamoxifen, estradiol (E2), fulvestrant, anastrazole, letrozole).
Conventional chemotherapeutics for breast cancer treatment comprise cytotoxic [19], hormonal [20], and immunotherapeutic agents [21]. Both in neoadjuvant and adjuvant instances, the effectiveness of the chemotherapeutics is limited by resistance developed in the tumour tissue. This is mainly due to the various genetic and epigenetic changes found in cancer cells, and the resistance thus conferred may be intrinsic or acquired [22]. Like most other tumour cells, breast cancer cells exhibit the phenomenon of multi-drug resistance (MDR) [23]. MDR is characterized by a combination of mechanisms including, P-glycoprotein (P-gp) [20], multidrug-resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP) of the ATP-binding cassette (ABC) membrane transporter family, which efflux a diverse range of anticancer drugs from the tumour cells [23,24]. Other notable mechanisms that simultaneously contribute to MDR are enhanced aldehyde dehydrogenase (ALDH) activity, up-regulation of anti-apoptotic B-cell lymphoma-2 (Bcl-2) family proteins and abnormal activation of signalling pathways such as PI3K (phosphatidylinositol 3-kinase)/Akt, Notch, Hedgehog and Wnt pathways [25,26,27]. These mechanisms are predominantly showcased in CSCs (cancer stem cells).
The recent surge in the number of cancer cases along with the development of drug resistance in a large number of tumours has pushed the direction of cancer research towards new arenas that provide the grounds for the development of more effective personalised medicine treatment. MicroRNAs (miRNAs) pave the way for this by being potential biomarkers for early cancer detection, and could also help in designing a more specific treatment plan by helping in the analysis of drug resistance and sensitivity [28]. Various studies have been conducted highlighting the effect of miRNAs in chemotherapeutic resistance in cancers such as gastric cancer [29], breast cancer [30], cervical cancer [31], colorectal cancer [32], lung cancer [33], oral cancer [34], ovarian cancer [35], pancreatic cancer [36], prostate cancer [37] and skin cancer [38].
In one study it was found that there was increased resistance to docetaxel in breast cancer tissues having decreased expression of miR-638, and the restoration of miR-638 in these tissues led to apoptosis and enhanced sensitivity to docetaxel [39]. Microarray miRNA expression analysis in OHT (4-hydroxytamoxifen) showed the overexpression of eight miRNA genes, namely, miR-221, miR-222, miR-181, miR-203, miR-375, miR-32, miR-171, and miR-213, as compared to regular MCF-7 cell line conferring resistance [40]. Furthermore, seven miRNAs were under-expressed in OHT cells: miR-342, miR-484, miR-21, miR-24, miR-27, miR-23 and miR-200. miR-221 and miR-222 were also found to be up-regulated in HER2/neu-positive primary human breast cancer cells [40].
When an MCF7 (Michigan Cancer Foundation-7 cell line treated with VP-16 (etoposide) was compared with the untreated parent MCF7 cell line, it was observed that 17 miRNAs had abnormal levels of expression; the majority of them were up-regulated, whereas miR-326, miR-429, miR-187, miR-7, and miR-92-2 showed decreased expression [41]. The results were verified by RT-PCR, and it was concluded that these miRNAs could be specific regulators of MRP1 (multidrug-resistance-associated protein) and play a critical role in MDR (multiple drug resistance) [41].
A clinical study comparing the effects of the drug tamoxifen versus tamoxifen plus breast radiotherapy, carried out on 71 lymph-node-negative (LNN) breast cancer patients, revealed that the up-regulation of miRNA-301 in co-operation with SKA2 (spindle kinetochore-associated complex subunit 2) increased proliferation, migration, invasion and tumour formation through the regulation of key signalling pathways including PTEN, FOXF2 and Col2A1 [42]. According to another study, high levels of miRNA-210 expression in plasma was observed to be associated with trastuzumab resistance in HER-2 (human epidermal growth factor receptor 2)-positive breast cancer patients [43]. Xiang Ao and his colleagues examined 55 pairs of breast cancer tissues and adjacent normal tissues in total, and found that resistance to taxol in breast cancer patients increased with the loss of miRNA-17 and miRNA-20b, by the up-regulation of nuclear receptor co-activator 3 (NCOA3) levels [44].
Over the years, several studies have focused on the role of various miRNAs in the chemotherapeutic resistance or sensitivity in breast cancer. However, none of these studies have been able to conclusively define the exact mechanism by which these miRNAs are involved in chemo-sensitivity/resistance. Through this study, we aim to provide insight into the association of the expression of specific miRNAs with breast-cancer-related chemotherapeutic drug resistance and sensitivity, thereby making it relevant in a clinical setting. Further, this study paves the way to devise new treatment strategies targeting these miRNAs, and developing alternate ways to counter the occurrence of chemo-resistance in breast cancer. This study was carried out with the aid of tools including meta-analysis and systematic review.

2. Methods

To obtain studies to perform the meta-analysis, two databases were extensively used: PubMed and Science Direct. This systematic review required articles related to the chemotherapeutic resistance specific to miRNA in breast cancer. To obtain relevant papers, the selection was performed using of the following MeSH (Medical Subject Heading) terms: “miRNA” or “microRNA”, “drug resistance” and “breast cancer”. To further refine the process of selection, only papers published within 2012–2018 were selected. This systematic review and meta-analysis study adheres to the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines [45].
The study search ended on (31 December 2018). After the initial screening process, additional studies were obtained via the reference section of relevant articles. The relevance of articles was determined by reading the title and abstract followed by the analysis of the complete text. The search was conducted in an orderly and elaborate manner, and was designed to meet the requirements of the study.

2.1. Selection Criteria

Studies that were to be used in the systematic review and meta-analysis had to adhere to certain selection criteria. These criteria were of two types: inclusion criteria and exclusion criteria. The inclusion criteria set the guidelines for the studies that could be included in the analysis process and included the following factors:
  • An analysis of the association between miRNA and breast cancer;
  • Studies with both breast cancer patients as well as in vitro studies with cell lines;
  • Studies that focused on cancer tissues that had resistance to some form of therapy;
  • Reporting of miRNA profiling platforms;
  • Information about the genes or pathways involved in chemotherapeutic resistance or sensitivity;
  • Inclusion of some in vitro assays to analyse the expression of miRNA or gene-related studies.
Some studies were not considered because of certain exclusion criteria. These included studies that were not in the English language, did not involve drug resistance in breast cancer, studies involving microbes and those focusing only on long non-coding (lnc) RNA. Additionally, review articles, editorials and studies with only in vitro or only breast cancer patient samples were excluded.

2.2. Data Analysis

The studies were evaluated separately by both authors (RJ and MRM), and further elaboration was performed with the help of corresponding authors. All articles were subject to the exclusion and inclusion criteria. An MS Excel worksheet (Master) was used to structurally store all the information obtained from the studies that qualified for final inclusion. After a complete survey of full-text and supplementary material, the data from all the studies were broken down under the following important headings: First author, Year of publication, Patients information, Location of the study, Ethnicity, Gender, Drug used, Clinical stages, Number of samples, Lymph node metastasis, Cell lines used, miRNA(s) involved, miRNA profiling platform and Drug pathways or gene associated. A number of biochemical and molecular assays were used to qualitatively and quantitatively analyse the miRNA expression in various studies. The frequency of their usage in all studies were compared and duly represented in a graphical form.
For further qualification of the studies, they had to pass a set of criteria that ensured a degree of quality control [46,47,48]. Two of the authors (RJ and MRM) critically assessed the quality of eligible articles for epidemiological studies based on some checklists derived from Dutch Cochrane Centre represented by Meta-analysis Of Observational Studies in Epidemiology (MOOSE) [49]. The studies that were finally selected had to meet all the criteria as determined by the authors. This process of sorting all the information obtained was a step that was crucial to ensure efficient examination of the studies.

2.3. Publication Bias

On the basis of a few distinct methods, two of the authors (RJ and MRM) individually assessed the risk of bias [50,51,52,53,54]. This included the number of patients, year of publication, study period, study location and diagnostic procedure. With the information obtained from the eligible studies, the reviewers arrived at a decision [55,56,57,58,59]. Egger’s and Begg’s bias indicator tests were employed to infer the publication bias along with the inverted funnel plot [60,61,62,63]. The effect size of statistically non-significant, unpublished and small studies was addressed using classic [64] and Orwin [65] fail-safe N tests. Duval and Tweedie’s trim and fill calculation was also performed to compute the new size effect, after the removal of an extremely positive and small study, until a symmetric funnel plot was obtained [66]. A third reviewer was consulted to resolve any disagreement regarding the decision of the team.

2.4. Statistical Analysis

We used the Comprehensive Meta-Analysis (CMA) 3.0 software for the meta-analysis and calculated the hazard ratios (HRs) with 95% confidence intervals (CIs). Cochran’s Q test and Higgins’ I2 statistic [67] were used to obtain the heterogeneity, and statistical significance was defined as a p-value less than 0.01. A fixed-effect model [67] or random-effects model [68] was used to calculate 95% CI in cases where significant heterogeneity was not observed. The overall standard deviation (SD) of each sample from the main sample was calculated using the statistical Z-test.

3. Results

The eligible studies for our systematic review and meta-analysis through search results identified are shown in the form of the flow chart in Figure 1. Of the 4584 potential studies, 600 were screened for further proceeding and 92 articles were analysed in depth. Finally, 85 studies were found to be confined to the inclusion and exclusion criteria and the eligible studies involved 5159 tissues. The main characteristics of the patients are represented in Table 1. The systematically reviewed articles met all the criteria, and of the 85 articles included only 6 had hazard ratios and 95% confidence intervals and among these 3 articles denoted them directly in the article and 3 were extracted from Kaplan–Meier curves through online software. Between the 85 articles published, 57 were from China, 9 were from the USA, 5 were from Japan, 3 were from India, 2 each were from France, Italy and Taiwan, and there was 1 from each of Argentina, Canada, Finland, South Korea and Spain. Thirty studies used frozen tissues samples, 15 studies used formalin fixed paraffin embedded (FFPE) samples, 6 studies used core needle biopsy and 1 used blood sample. Meanwhile, 33 studies did not mention the type of material used.
A total of 22 cell lines were used in the 85 studies, and MCF-7, SKBR3, T47D and MDA-MB-231 cell lines were the most frequently included, with MCF-7 used in 33 studies. Zhao Y et al. (2011) used the highest number of cell lines in a single study [135].
Overall, 188 miRNAs were studied in our systematic review and meta-analysis, conjointly 96 miRNAs were upregulated and 87 miRNAs were downregulated. Elevated expression of miR- 18a, 21, 21-3p, 29a, 31, 34a, 34c-5p, 124, 125b, 130b, 137, 138, 138-5p, 139, 139-5p, 140, 140-3p, 141, 149, 149-3p, 155-5p, 181a-5p, 181b, 181b-5p, 181d, 183-5p, 197, 197-3p, 200a-5p, 200c, 205, 210, 210-3p, 221, 222, 378a-3p, 423, 423-5p, 520h, 574, 574-3p, 663, 671, 671-5p, 744, 744-5p, 944, 1246, 1268a, 3178, 3613, 3613-5p, 4258, 4298, 4438, 4443, 4644, 6780b, 6780b-3p, 7107, 7107-5p, 7847, 7847-3p, Let-7a and Lin28 and redundant expression of miR-7, 10b-5p, 17, 20a, 20b, 21, 24-2, 25, 25-3p, 27b, 31-5p, 34a-3p, 103, 125a-3p, 125, 125b-5p, 128, 134, 145, 148a, 149, 181a, 191, 195, 195-5p, 200c, 210, 221, 222, 301a, 320a, 375, 424, 451, 489, 520b-5p, 532-3p, 548n, 574-3p, 708-3p, 873 and Let7a were associated with chemotherapeutic resistance and increased expression of miR- 16, 27a, 34a, 128, 148a, 152, 155, 210, 221, 346, 484 and Let-7 and reduced expression of miR- 21, 24, 23b, 26a, 26b, 27b, 27b-3p, 34a, 100, 125a-3p, 125b-1, 130a-3p, 139, 145, 181a, 181b, 195, 200, 200c, 205, 214, 216b, 218, 301, 320a, 326, 342, 370, 378a-3p, 451a, 489, 576-3p, 638, 760, 765, 1254, Let-7 and Let-7a were associated with chemosensitivity.
Five miRNAs were differentially regulated and four miRNAs (i.e., miR- 90b, 130a, 200b and 452) contributed to chemoresistance. miR-491-3p did not have any impact on chemoresistance or sensitivity. Chemotherapeutic resistance and chemosensitivity were boosted by the miRNAs through drug-regulated cellular pathways. In total, 26 drugs were studied in the included articles: 5-FU, anastrozole, cisplatin, cyclophosphamide, docetaxel, doxorubicin, E2, epirubicin, etoposide, fulvestrant, gemcitabine, lapatinib, letrozole, methotrexate, mitoxantrone, Paclitaxel, PiB, tamoxifen, topotecan, trastuzumab, vinorelbine and combinations such as cisplatin plus doxorubicin, epirubicin plus Paclitaxel, Paclitaxel plus carboplatin, taxol plus doxorubicin plus Cyclophosphamide, Methotrexate, Fluorouracil (CMF), and anthracycline plus taxane were studied, and radiotherapy was also observed in one study.

miRNA Pathway Relation

The miRNA and pathways involved in chemoresistance and chemosensitivity are represented in Table 2 and Table 3, respectively.
The relationship between miRNA expression and patient survival was assessed by meta-analysis. Breast cancer (BC) patients had elevated expressions of miR-125b (HR = 6.350, 95% CI = 1.211–33.297), 484 (HR = 0.375, 95% CI = 0.193–0.730), 520h (HR = 1.233, 95% CI = 0.890–1.707), 4443 (HR = 0.721, 95% CI = 0.529–0.983) and downregulated expression of miR-200c (HR = 0.433, 95% CI = 0.102–1.829), 489 (HR = 0.703, 95% CI = 0.415–1.191). An extensive examination found that 89 out of 95 articles did not mention the HR and 95% confidence interval values and of the six remaining articles, only three mentioned them in their manuscript and three HR values were obtained from Kaplan–Meier curve through online software. So, 89 studies were excluded from our meta-analysis due to insufficient data. Cumulatively, a meta-analysis was done for six studies encompassing 852 samples (Figure 2).
An unbiased correlation was observed from Begg and Mazumdar rank collection test results. Regarding Duval and Tweedie’s trim and fill calculation for the fixed-effect model, the point estimate and 95% confidence interval for the combined studies was 0.83921 (0.69115–1.01899). Under the random-effects model, the point estimate and 95% confidence interval for the combined studies was 0.79909 (0.50575–1.26256). Using trim and fill, these values were unchanged. Egger’s regression intercepted at −0.132 with 95% CI from −5.141 to 4.877; t = 0.07, p = 0.945. The 1-tailed p-value was 0.47237, and the 2-tailed p-value was 0.94473. The funnel plot is represented in Figure 3.

4. Discussion

This systematic meta-analysis of “the miRNAs that influence the chemoresistance or chemosensitivity to drugs in breast cancer” carefully reviewed over 400 research articles through a systematic PubMed search query from which 80 research articles were scrutinized based on the inclusion criteria.
From the meta-analysis, the results indicate that many miRNAs could intricately orchestrate cellular functions including chemosensitivity/resistance through post-transcriptional control on target gene expression, either canonically or non-canonically. Of the studies included in this meta-analysis, anthracyclines like doxorubicin and epirubicin were predominantly tested in patients/cell lines to study the differential expression of miRNAs followed by tamoxifen in the case of Estrogen Receptor (ER) positive subjects and trastuzumab in the case of Human Epidermal growth factor Receptor (HER) positive subjects. A major limitation in our research is that less than 10% of the 80 papers (6 papers) had direct hazard values that could be utilized for the meta-analysis, reducing the accuracy of the results obtained since only a small fraction of papers were used to give results of the whole, leading to the biasing of the results. There is a possibility of our interpretation being wrong in the context of heterogeneous disease.

4.1. Role of miRNAs in Guiding Diagnosis and Prognosis

We extracted the prognosis results of six miRNAs from six different studies. Among the selected miRNAs, two miRNAs (miR200c and miR489) were downregulated and the remaining four miRNAs (miR484, miR4443, miR520h and miR125b) were upregulated. Both downregulated miRNAs were associated with better prognosis; similarly, both miRNAs (miR484 and miR4443) from the overexpressed miRNAs were expressed as better prognosis whereas miR520h and miR125b were associated with poor prognosis.
The overall hazard ratio (95% CI) of the prognostic significance was 0.78 (0.508–1.100) at a p-value of 0.140 which was analysed by random-effect model. This overall combined sized effect estimate indicates that the miRNAs decreased the likelihood of death of breast cancer patients by 22%. This means an HR value >1 indicates an increased risk of breast cancer survival whereas an HR <1 indicates a decreased risk of breast cancer patient survival. The Z-value of the overall effect size was −1.476. The individual overall hazard ratios (95% CIs) of upregulated and downregulated miRNAs were estimated 0.662 (0.403–1.087) and 0.904 (0.487–1.678), respectively. On observing the overall effect size of the individual subgroups, the significant prognosis was associated with a good prognosis, and hence the miRNAs could be considered as better prognostic biomarkers for breast cancer patients.
The Z-value of upregulated and downregulated miRNAs for the null hypothesis test (the mean risk ratio of which is 1.0) were −1.636 and −0.319, respectively. Both the differently expressed miRNA subgroups were associated with lower risk of death in breast cancer patients and hence we cannot accept the null hypothesis that the risk is lower in both differently expressed miRNAs. Similar to our study, two other studies have studied the subgroup analysis of higher and lower expressed miRNAs in meta-analysis studies of the prognosis of melanoma and nasopharyngeal carcinoma patient survival. Those studies demonstrated different risk levels among the subgroups, whereas in our study both subgroups exhibited better prognosis for cancer patients. More studies are required to obtain better prognostic significance of miRNAs in breast cancer patients [147].

4.2. Current Challenges

Systematic reviews and meta-analytic studies face a number of challenges when investigating the theragnostic relationship between miRNA and chemotherapeutic response in breast cancer. The primary limiting factor for detailed analysis and clinically applicable insights/results is the scarcity of data. The literature in this specific niche of breast cancer treatment is sparse, with few high-quality studies being available for comparison and analysis. This challenge is exacerbated by the lack of homogeneity between similar studies. The variance in study parameters and the methodology makes assessment difficult by introducing uncertainty in the reliability of the results. Furthermore, a large number of studies have explored this topic via the use of in-vitro models, which cannot be directly applied to clinical theragnostics. The lack of well-documented, large-scale, patient-based clinical studies is a significant challenge faced by this study. Furthermore, the mechanisms of miRNA and chemotherapeutic response are not currently understood in detail, requiring further assessment in the future if meta-analytic studies are to provide conclusions viable for application in the clinical sphere.
The strengths of our paper include its large set of research papers, varied results in terms of miRNAs and pathways that show a change in function in cancerous cells. The result of this exhaustive analysis has provided us with a large number of miRNAs that can be focused on for prognostic or diagnostic purposes. Many miRNAs play a role in regulating many vital cellular pathways, and these regulations are observed to be significantly potentiated or deregulated during treatment with chemotherapeutics. A single miRNA can regulate multiple genes, and this regulation down the cascade can affect many pathways. Many reports have independently observed several genes or pathways as targets of many miRNAs. Of those, the treatment of doxorubicin has been frequently observed to affect the PTEN/Akt and MAPK signalling pathways, and increases chemoresistance (Table 3). In the case of miRNA 21 which is also an oncomiR, treatment with Fulvestrant; Selective Estrogen Receptor Degrader (SERD) or trastuzumab (HER2 antagonist) leads to downregulation, affecting the EMT. Whereas treatment with tamoxifen; Selective Estrogen Receptor Modulators (SERM) downregulates the expression of miRNA-21 via estrogen-dependent functions, leading to chemosensitivity.
In case of miRNAs 221 and 222, the treatment with fulvestrant, doxorubicin or trastuzumab also leads to the downregulation with increased expression of ABC transporters. The treatment with Paclitaxel leads to the downregulation of miRNA 320a with downregulation of TRPC5, NFATC3 and the FTS-1 genes, ultimately causing chemoresistance. miRNA 125b is upregulated when treated with tamoxifen, letrozole, anastrazole or fulvestrant due to its interaction with the Akt/mTOR pathway, leading to chemoresistance. The same pattern is observed when treatment of 5-FU, Paclitaxel and cyclophosphamide is applied, which affects the EMT pathway; or when 5-FU is used, which affects the transcription factor E2F3.
miRNAs Let-7, 181a and 145 are also majorly downregulated when treated with drugs like doxorubicin, tamoxifen, or epirubicin, with increases in chemosensitivity. Thus, myriad miRNAs take centre stage in the search for theragnostic miRNAs indicating drug resistance. However, Our study has tried its best to bridge the gaps, and serves as a benchmark for further clinical studies in personalized treatment research.

Author Contributions

R.J. contributed to the conceptualisation, study design, search strategy, protocol development and review by revising different versions. R.J., M.R.M., A.K., S.S. (Shubhangi Sathyakumar), H.M., M.S., S.S. (Shanthi Sabarimurugan), C.K., S.G.N., N.R., K.M.G., S.K., T.P., S.S. (Suja Swamiappan), A.G. and S.B. provided input into the study design, supervision, ensured the absence of errors and arbitrated in case of disagreement. M.R.M., A.K., S.S. (Shubhangi Sathyakumar), H.M., M.S., S.S. (Shanthi Sabarimurugan) and S.G.N. engaged in initial searches to determine the feasibility, data collection, analysis and drafting the manuscript. All authors have read and approved the final version of the manuscript.

Funding

This study did not receive any funding from any organisation.

Acknowledgments

We would like to acknowledge the Meta-Analysis Concepts and Applications workshop manual by Michael Borenstein for his guidelines on reporting meta-analysis, subgroup analysis and publication bias (www.meta-analysis-workshops.com).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Society, A.C. Cancer Facts & Figures 2017. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2017.html (accessed on 12 June 2018).
  2. Berry, J. Worldwide statistics on breast cancer. Available online: https://www.medicalnewstoday.com/articles/317135.php (accessed on 13 January 2019).
  3. Malvia, S.; Bagadi, S.A.; Dubey, U.S.; Saxena, S. Epidemiology of breast cancer in Indian women. Asia-Pac. J. Clin. Oncol. 2017, 13, 289–295. [Google Scholar] [CrossRef] [PubMed]
  4. Research, I.C.o.M. Three-Year Report of Population Based Cancer Registries 2012–2014. Available online: http://www.ncdirindia.org/ncrp/ALL_NCRP_REPORTS/PBCR_REPORT_2012_2014/ALL_CONTENT/PDF_Printed_Version/Preliminary_Pages_Printed.pdf (accessed on 13 January 2019).
  5. Research, I.C.o.M. Consolidated Report of Hospital Based Cancer Registries 2012–2014. Available online: http://www.ncdirindia.org/ncrp/ALL_NCRP_REPORTS/HBCR_REPORT_2012_2014/ALL_CONTENT/PDF_Printed_Version/Preliminary_Pages.pdf (accessed on 13 January 2019).
  6. Weigelt, B.; Geyer, F.C.; Reis-Filho, J.S. Histological types of breast cancer: How special are they? Mol. Oncol. 2010, 4, 192–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Cheang, M.C.; Chia, S.K.; Voduc, D.; Gao, D.; Leung, S.; Snider, J.; Watson, M.; Davies, S.; Bernard, P.S.; Parker, J.S. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J. Natl. Cancer. Inst. 2009, 101, 736–750. [Google Scholar] [CrossRef] [PubMed]
  8. Gärtner, R.; Jensen, M.-B.; Nielsen, J.; Ewertz, M.; Kroman, N.; Kehlet, H.J.J. Prevalence of and factors associated with persistent pain following breast cancer surgery. JAMA 2009, 302, 1985–1992. [Google Scholar] [CrossRef]
  9. Early Breast Cancer Trialists’ Collaborative Group. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: An overview of the randomised trials. Lancet 2005, 366, 2087–2106. [Google Scholar] [CrossRef]
  10. Fournier, A.; Berrino, F.; Clavel-Chapelon, F. Unequal risks for breast cancer associated with different hormone replacement therapies: Results from the E3N cohort study. Breast Cancer Res. Treat. 2008, 107, 103–111. [Google Scholar] [CrossRef]
  11. Higgins, M.J.; Baselga, J. Targeted therapies for breast cancer. J. Clin. Invest. 2011, 121, 3797–3803. [Google Scholar] [CrossRef]
  12. Miles, D.; Towlson, K.; Graham, R.; Reddish, M.; Longenecker, B.; Taylor-Papadimitriou, J.; Rubens, R.D. A randomised phase II study of sialyl-Tn and DETOX-B adjuvant with or without cyclophosphamide pretreatment for the active specific immunotherapy of breast cancer. Brit. J. Cancer 1996, 74, 1292–1296. [Google Scholar] [CrossRef]
  13. Hassan, M.; Ansari, J.; Spooner, D.; Hussain, S.J.O. Chemotherapy for breast cancer. Oncol. Rep. 2010, 24, 1121–1131. [Google Scholar] [CrossRef]
  14. Kovalchuk, O.; Filkowski, J.; Meservy, J.; Ilnytskyy, Y.; Tryndyak, V.P.; Vasyl’F, C.; Pogribny, I.P. Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol. Cancer Therap. 2008, 7, 2152–2159. [Google Scholar] [CrossRef] [Green Version]
  15. Buzdar, A.U.; Valero, V.; Ibrahim, N.K.; Francis, D.; Broglio, K.R.; Theriault, R.L.; Pusztai, L.; Green, M.C.; Singletary, S.E.; Hunt, K.K. Neoadjuvant therapy with Paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophosphamide chemotherapy and concurrent trastuzumab in human epidermal growth factor receptor 2–positive operable breast cancer: An update of the initial randomized study population and data of additional patients treated with the same regimen. Clin. Cancer Res. 2007, 13, 228–233. [Google Scholar] [PubMed]
  16. Jones, S.; Erban, J.; Overmoyer, B.; Budd, G.; Hutchins, L.; Lower, E.; Laufman, L.; Sundaram, S.; Urba, W.; Pritchard, K.I. Randomized phase III study of docetaxel compared with Paclitaxel in metastatic breast cancer. J. Clin. Oncol. 2005, 23, 5542–5551. [Google Scholar] [CrossRef] [PubMed]
  17. Robert, N.; Leyland-Jones, B.; Asmar, L.; Belt, R.; Ilegbodu, D.; Loesch, D.; Raju, R.; Valentine, E.; Sayre, R.; Cobleigh, M. Randomized phase III study of trastuzumab, Paclitaxel, and carboplatin compared with trastuzumab and Paclitaxel in women with HER-2–overexpressing metastatic breast cancer. J. Clin. Oncol. 2006, 24, 2786–2792. [Google Scholar] [CrossRef] [PubMed]
  18. Joensuu, H.; Bono, P.; Kataja, V.; Alanko, T.; Kokko, R.; Asola, R.; Utriainen, T.; Turpeenniemi-Hujanen, T.; Jyrkkiö, S.; Möykkynen, K. Fluorouracil, epirubicin, and cyclophosphamide with either docetaxel or vinorelbine, with or without trastuzumab, as adjuvant treatments of breast cancer: Final results of the FinHer Trial. J. Clin. Oncol. 2009, 27, 5685–5692. [Google Scholar] [CrossRef] [PubMed]
  19. Hanahan, D.; Bergers, G.; Bergsland, E. Less is more, regularly: Metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J. Clin. Investig. 2000, 105, 1045–1047. [Google Scholar] [CrossRef]
  20. Gonzalez-Angulo, A.M.; Morales-Vasquez, F.; Hortobagyi, G.N. Overview of resistance to systemic therapy in patients with breast cancer. Adv. Exp. Med. Biol. 2007, 608, 1–22. [Google Scholar]
  21. Early Breast Cancer Trialists’ Collaborative Group. Systemic treatment of early breast cancer by hormonal, cytotoxic, or immune therapy: 133 randomised trials involving 31 000 recurrences and 24 000 deaths among 75 000 women. Lancet 1992, 339, 1–15. [Google Scholar]
  22. Niero, E.L.; Rocha-Sales, B.; Lauand, C.; Cortez, B.A.; de Souza, M.M.; Rezende-Teixeira, P.; Urabayashi, M.S.; Martens, A.A.; Neves, J.H.; Machado-Santelli, G.M.; et al. The multiple facets of drug resistance: One history, different approaches. J Exp. Clin. Cancer Res.. 2014. [Google Scholar] [CrossRef]
  23. Coley, H.M. Mechanisms and strategies to overcome chemotherapy resistance in metastatic breast cancer. Cancer Treat. Rev. 2008, 34, 378–390. [Google Scholar] [CrossRef]
  24. Baguley, B.C. Multiple drug resistance mechanisms in cancer. Mol. Biotechnol. 2010, 46, 308–316. [Google Scholar] [CrossRef]
  25. Hu, Y.; Guo, R.; Wei, J.; Zhou, Y.; Ji, W.; Liu, J.; Zhi, X.; Zhang, J.J. Effects of PI3K inhibitor NVP-BKM120 on overcoming drug resistance and eliminating cancer stem cells in human breast cancer cells. Cell Death Dis. 2015. [Google Scholar] [CrossRef] [PubMed]
  26. Tanei, T.; Morimoto, K.; Shimazu, K.; Kim, S.J.; Tanji, Y.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin. Cancer Res. 2009, 15, 4234–4241. [Google Scholar] [CrossRef] [PubMed]
  27. Teixeira, C.; Reed, J.C.; Pratt, M.C. Estrogen promotes chemotherapeutic drug resistance by a mechanism involving Bcl-2 proto-oncogene expression in human breast cancer cells. Cancer Res. 1995, 55, 3902–3907. [Google Scholar]
  28. Garofalo, M.; Croce, C.M. MicroRNAs as therapeutic targets in chemoresistance. Drug Resist. Updat. 2013, 16, 47–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Yang, W.; Ma, J.; Zhou, W.; Cao, B.; Zhou, X.; Yang, Z.; Zhang, H.; Zhao, Q.; Fan, D.; Hong, L. Molecular mechanisms and theranostic potential of miRNAs in drug resistance of gastric cancer. Exp. Opinion Therap. Tar. 2017, 21, 1063–1075. [Google Scholar] [CrossRef]
  30. Lin Teoh, S.; Das, S. The role of MicroRNAs in diagnosis, prognosis, metastasis and resistant cases in breast cancer. Curr. Pharmaceut. Des. 2017, 23, 1845–1859. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, W.; Zhou, J.; Zhu, X.; Yuan, H.J.G. MiR-126 reverses drug resistance to TRAIL through inhibiting the expression of c-FLIP in cervical cancer. Gene 2017, 627, 420–427. [Google Scholar] [CrossRef]
  32. Zhang, Y.; Wang, J. MicroRNAs are important regulators of drug resistance in colorectal cancer. Biol. Chem. 2017, 398, 929–938. [Google Scholar] [CrossRef] [Green Version]
  33. Võsa, U.; Vooder, T.; Kolde, R.; Vilo, J.; Metspalu, A.; Annilo, T. Meta-analysis of microRNA expression in lung cancer. Internat. J. Cancer 2013, 132, 2884–2893. [Google Scholar] [CrossRef]
  34. Zhuang, Z.; Hu, F.; Hu, J.; Wang, C.; Hou, J.; Yu, Z.; Wang, T.T.; Liu, X.; Huang, H. MicroRNA-218 promotes cisplatin resistance in oral cancer via the PPP2R5A/Wnt signaling pathway. Oncol. Rep. 2017, 38, 2051–2061. [Google Scholar] [CrossRef]
  35. Tung, S.; Huang, W.; Hsu, F.; Yang, Z.; Jang, T.; Chang, J.; Chuang, C.; Lai, C.; Wang, L.J.O. miRNA-34c-5p inhibits amphiregulin-induced ovarian cancer stemness and drug resistance via downregulation of the AREG-EGFR-ERK pathway. Oncogenesis 2017. [Google Scholar] [CrossRef] [PubMed]
  36. Amponsah, P.S.; Fan, P.; Bauer, N.; Zhao, Z.; Gladkich, J.; Fellenberg, J.; Herr, I. microRNA-210 overexpression inhibits tumor growth and potentially reverses gemcitabine resistance in pancreatic cancer. Cancer Lett. 2017, 388, 107–117. [Google Scholar] [CrossRef] [PubMed]
  37. Armstrong, C.M.; Liu, C.; Lou, W.; Lombard, A.P.; Evans, C.P.; Gao, A.C. MicroRNA-181a promotes docetaxel resistance in prostate cancer cells. Prostate 2017, 77, 1020–1028. [Google Scholar] [CrossRef] [PubMed]
  38. Fattore, L.; Sacconi, A.; Mancini, R.; Ciliberto, G.J.C. MicroRNA-driven deregulation of cytokine expression helps development of drug resistance in metastatic melanoma. Cytokine Growth Factor Rev. 2017, 36, 39–48. [Google Scholar] [CrossRef]
  39. Zhao, G.; Li, Y.; Wang, T.J.B. Potentiation of docetaxel sensitivity by miR-638 via regulation of STARD10 pathway in human breast cancer cells. Biochem. Biophys. Res. Commun. 2017, 487, 255–261. [Google Scholar] [CrossRef]
  40. Miller, T.E.; Ghoshal, K.; Ramaswamy, B.; Roy, S.; Datta, J.; Shapiro, C.L.; Jacob, S.; Majumder, S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J. Biol. Chem. 2008, 283, 29897–29903. [Google Scholar] [CrossRef]
  41. Liang, Z.; Wu, H.; Xia, J.; Li, Y.; Zhang, Y.; Huang, K.; Wagar, N.; Yoon, Y.; Cho, H.T.; Scala, S. Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem. Pharmacol. 2010, 79, 817–824. [Google Scholar] [CrossRef] [Green Version]
  42. Shi, W.; Gerster, K.; Alajez, N.M.; Tsang, J.; Pintilie, M.; Hui, A.B.; Sykes, J.; P’ng, C.; Miller, N.; McCready, D. MicroRNA-301 mediates proliferation and invasion in human breast cancer. Cancer Res. 2011, 71, 2926–2937. [Google Scholar] [CrossRef]
  43. Jung, E.J.; Santarpia, L.; Kim, J.; Esteva, F.J.; Moretti, E.; Buzdar, A.U.; Di Leo, A.; Le, X.F.; Bast Jr, R.C.; Park, S.T. Plasma microRNA 210 levels correlate with sensitivity to trastuzumab and tumor presence in breast cancer patients. Cancer 2012, 118, 2603–2614. [Google Scholar] [CrossRef]
  44. Ao, X.; Nie, P.; Wu, B.; Xu, W.; Zhang, T.; Wang, S.; Chang, H.; Zou, Z. Decreased expression of microRNA-17 and microRNA-20b promotes breast cancer resistance to taxol therapy by upregulation of NCOA3. Cell Death Dis. 2016. [Google Scholar] [CrossRef]
  45. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Ann. Intern. Med. 2009, 151, 264–269. [Google Scholar] [CrossRef] [PubMed]
  46. Madhav, M.R.; Nayagam, S.G.; Biyani, K.; Pandey, V.; Kamal, D.G.; Sabarimurugan, S.; Ramesh, N.; Gothandam, K.M.; Jayaraj, R. Epidemiologic analysis of breast cancer incidence, prevalence, and mortality in India: Protocol for a systematic review and meta-analyses. Medicine 2018. [Google Scholar] [CrossRef] [PubMed]
  47. Poddar, A.; Aranha, R.R.; Muthukaliannan, G.K.; Nachimuthu, R.; Jayaraj, R. Head and neck cancer risk factors in India: Protocol for systematic review and meta-analysis. BMJ Open 2018. [Google Scholar] [CrossRef] [PubMed]
  48. Jayaraj, R.; Kumarasamy, C.; Piedrafita, D. Systematic review and meta-analysis protocol for Fasciola DNA vaccines. J. Vet. Res. 2018, 22, 517–524. [Google Scholar]
  49. Stroup, D.F.; Berlin, J.A.; Morton, S.C.; Olkin, I.; Williamson, G.D.; Rennie, D.; Moher, D.; Becker, B.J.; Sipe, T.A.; Thacker, S.B. Meta-analysis of observational studies in epidemiology: A proposal for reporting. JAMA 2000, 283, 2008–2012. [Google Scholar] [CrossRef] [PubMed]
  50. Jayaraj, R.; Kumarasamy, C.; Madhav, M.R.; Pandey, V.; Sabarimurugan, S.; Ramesh, N.; Gothandam, K.M.; Baxi, S. Comment on “Systematic Review and Meta-Analysis of Diagnostic Accuracy of miRNAs in Patients with Pancreatic Cancer”. Dis. Markers 2018. [Google Scholar] [CrossRef]
  51. Jayaraj, R.; Kumarasamy, C. Comment on ’Prognostic biomarkers for oral tongue squamous cell carcinoma: A systematic review and meta-analysis’. Br. J. Cancer 2018. [Google Scholar] [CrossRef]
  52. Jayaraj, R.; Kumarasamy, C. Comment on,” Survival for HPV-positive oropharyngeal squamous cell carcinoma with surgical versus non-surgical treatment approach: A systematic review and meta-analysis”. J. Oral Oncol. 2018, 90, 137–138. [Google Scholar] [CrossRef]
  53. Jayaraj, R.; Kumarasamy, C.; Sabarimurugan, S.; Baxi, S. Commentary: Blood-Derived microRNAs for Pancreatic Cancer Diagnosis: A Narrative Review and Meta-Analysis. Front. Physiol. 2018. [Google Scholar] [CrossRef]
  54. Jayaraj, R.; Kumarasamy, C. Conceptual interpretation of analysing and reporting of results on systematic review and meta-analysis of optimal extent of lateral neck dissection for well-differentiated thyroid carcinoma with metastatic lateral neck lymph nodes. Oral Oncol. 2019, 89, 153–154. [Google Scholar] [CrossRef]
  55. Jayaraj, R.; Kumarasamy, C.; Gothandam, K.M. Letter to the editor “Prognostic value of microRNAs in colorectal cancer: A meta-analysis”. Cancer Manag. Res. 2018, 10, 3501–3503. [Google Scholar] [CrossRef] [PubMed]
  56. Jayaraj, R.; Kumarasamy, C. Letter to the Editor about the Article: “Performance of different imaging techniques in the diagnosis of head and neck cancer mandibular invasion: A systematic review and meta-analysis”. J. Oral Oncol. 2018, 89, 159–160. [Google Scholar] [CrossRef] [PubMed]
  57. Jayaraj, R.; Kumarasamy, C.; Sabarimurugan, S.; Baxi, S. Letter to the Editor in response to the article, “The epidemiology of oral human papillomavirus infection in healthy populations: A systematic review and meta-analysis”. Oral Oncol. 2018, 84, 121–122. [Google Scholar] [CrossRef] [PubMed]
  58. Jayaraj, R.; Kumarasamy, C.; Samiappan, S.; Swaminathan, P. Letter to the Editor regarding, “The prognostic role of PD-L1 expression for survival in head and neck squamous cell carcinoma: A systematic review and meta-analysis”. Oral Oncol. 2019, 90, 139–140. [Google Scholar] [CrossRef] [PubMed]
  59. Jayaraj, R.; Kumarasamy, C.; Madurantakam Royam, M.; Devi, A.; Baxi, S. Letter to the editor: Is HIF-1alpha a viable prognostic indicator in OSCC? A critical review of a meta-analysis study. World J. Surg. Oncol. 2018. [Google Scholar] [CrossRef] [PubMed]
  60. Kumarasamy, C.; Devi, A.; Jayaraj, R. Prognostic value of microRNAs in head and neck cancers: A systematic review and meta-analysis protocol. Syst. Rev. 2018. [Google Scholar] [CrossRef]
  61. Jayaraj, R.; Kumarasamy, C. Systematic review and meta-analysis of cancer studies evaluating diagnostic test accuracy and prognostic values: Approaches to improve clinical interpretation of results. Cancer Manag. Res. 2018, 10, 4669–4670. [Google Scholar] [CrossRef]
  62. Jayaraj, R.; Kumarasamy, C.; Ramalingam, S.; Devi, A. Systematic review and meta-analysis of risk-reductive dental strategies for medication related osteonecrosis of the jaw among cancer patients: Approaches and strategies. Oral Oncol. 2018, 86, 312–313. [Google Scholar] [CrossRef]
  63. Sabarimurugan, S.; Madurantakam Royam, M.; Das, A.; Das, S.; Gothandam, K.M.; Jayaraj, R. Systematic Review and Meta-analysis of the Prognostic Significance of miRNAs in Melanoma Patients. Mol. Diagn. Ther. 2018, 22, 653–669. [Google Scholar] [CrossRef]
  64. Rosenthal, R. The file drawer problem and tolerance for null results. Psychol. Bull. 1979, 86, 638–641. [Google Scholar] [CrossRef]
  65. Orwin, R.G. A fail-safe N for effect size in meta-analysis. J. Educ. Stat. 1983, 8, 157–159. [Google Scholar] [CrossRef]
  66. Duval, S.; Tweedie, R. Trim and fill: A simple funnel-plot–based method of testing and adjusting for publication bias in meta-analysis. Biometrics 2000, 56, 455–463. [Google Scholar] [CrossRef] [PubMed]
  67. Higgins, J.P.; Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 2002, 21, 1539–1558. [Google Scholar] [CrossRef] [PubMed]
  68. DerSimonian, R.; Laird, N. Meta-analysis in clinical trials. Control. Clin. Trials 1986, 7, 177–188. [Google Scholar] [CrossRef]
  69. Lin, X.; Chen, W.; Wei, F.; Zhou, B.P.; Hung, M.-C.; Xie, X. Nanoparticle Delivery of miR-34a Eradicates Long-term-cultured Breast Cancer Stem Cells via Targeting C22ORF28 Directly. Theranostics 2017, 7, 4805–4824. [Google Scholar] [CrossRef] [Green Version]
  70. Nakano, M.; Fukami, T.; Gotoh, S.; Nakajima, M. A-to-I RNA editing up-regulates human dihydrofolate reductase in breast cancer. JBC 2017. [Google Scholar] [CrossRef]
  71. Miao, Y.; Zheng, W.; Li, N.; Su, Z.; Zhao, L.; Zhou, H.; Jia, L. MicroRNA-130b targets PTEN to mediate drug resistance and proliferation of breast cancer cells via the PI3K/Akt signaling pathway. Sci. Rep. 2017. [Google Scholar] [CrossRef]
  72. Chen, M.-J.; Cheng, Y.-M.; Chen, C.-C.; Chen, Y.-C.; Shen, C.-J. MiR-148a and miR-152 reduce tamoxifen resistance in ER+ breast cancer via downregulating ALCAM. Biochem. Biophys. Res. Commun. 2017, 483, 840–846. [Google Scholar] [CrossRef]
  73. Yang, F.; Luo, L.-j.; Zhang, L.; Wang, D.-d.; Yang, S.-j.; Ding, L.; Li, J.; Chen, D.; Ma, R.; Wu, J.-z. MiR-346 promotes the biological function of breast cancer cells by targeting SRCIN1 and reduces chemosensitivity to docetaxel. Gene 2017, 600, 21–28. [Google Scholar] [CrossRef]
  74. Gong, J.P.; Yang, L.; Tang, J.W.; Sun, P.; Hu, Q.; Qin, J.W.; Xu, X.M.; Sun, B.C.; Tang, J.H. Overexpression of microrna-24 increases the sensitivity to Paclitaxel in drug-resistant breast carcinoma cell lines via targeting abcb9. Oncol. Lett. 2016, 12, 3905–3911. [Google Scholar] [CrossRef]
  75. Zhu, J.; Zou, Z.; Nie, P.; Kou, X.; Wu, B.; Wang, S.; Song, Z.; He, J. Downregulation of microRNA-27b-3p enhances tamoxifen resistance in breast cancer by increasing NR5A2 and CREB1 expression. Cell Death Dis. 2016, 7, e2454. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, X.; Lu, P.; Wang, D.-d.; Yang, S.-j.; Wu, Y.; Shen, H.-Y.; Zhong, S.-l.; Zhao, J.-h.; Tang, J.-h. The role of miRNAs in drug resistance and prognosis of breast cancer formalin-fixed paraffin-embedded tissues. Gene 2016, 595, 221–226. [Google Scholar] [CrossRef] [PubMed]
  77. Damiano, V.; Brisotto, G.; Borgna, S.; di Gennaro, A.; Armellin, M.; Perin, T.; Guardascione, M.; Maestro, R.; Santarosa, M. Epigenetic silencing of miR-200c in breast cancer is associated with aggressiveness and is modulated by ZEB1. Gene. Chromosome. Cancer 2017, 56, 147–158. [Google Scholar] [CrossRef] [PubMed]
  78. Jana, S.; Sengupta, S.; Biswas, S.; Chatterjee, A.; Roy, H.; Bhattacharyya, A.J.B. miR-216b suppresses breast cancer growth and metastasis by targeting SDCBP. Biochem. Biophys. Res. Commun. 2017, 482, 126–133. [Google Scholar] [CrossRef]
  79. Wang, D.-d.; Yang, S.-j.; Chen, X.; Shen, H.-Y.; Luo, L.-j.; Zhang, X.-h.; Zhong, S.-l.; Zhao, J.-h.; Tang, J.-h. miR-222 induces Adriamycin resistance in breast cancer through PTEN/Akt/p27 kip1 pathway. Tumor Biol. 2016, 37, 15315–15324. [Google Scholar] [CrossRef]
  80. Xu, X.; Lv, Y.-g.; Yan, C.-y.; Yi, J.; Ling, R. Enforced expression of hsa-miR-125a-3p in breast cancer cells potentiates docetaxel sensitivity via modulation of BRCA1 signaling. Biochem. Biophys. Res. Commun. 2016, 479, 893–900. [Google Scholar] [CrossRef]
  81. Chen, X.; Zhong, S.-l.; Lu, P.; Wang, D.-d.; Zhou, S.-y.; Yang, S.-j.; Shen, H.-y.; Zhang, L.; Zhang, X.-h.; Zhao, J.-h. miR-4443 Participates in the Malignancy of Breast Cancer. PLoS ONE 2016. [Google Scholar] [CrossRef]
  82. Gao, M.; Miao, L.; Liu, M.; Li, C.; Yu, C.; Yan, H.; Yin, Y.; Wang, Y.; Qi, X.; Ren, J. miR-145 sensitizes breast cancer to doxorubicin by targeting multidrug resistance-associated protein-1. Oncotarget 2016, 7, 59714–59726. [Google Scholar] [CrossRef] [Green Version]
  83. Thakur, S.; Grover, R.K.; Gupta, S.; Yadav, A.K.; Das, B.C. Identification of specific miRNA signature in paired sera and tissue samples of Indian women with triple negative breast cancer. PLoS ONE 2016. [Google Scholar] [CrossRef]
  84. Hu, Y.; Qiu, Y.; Yagüe, E.; Ji, W.; Liu, J.; Zhang, J. miRNA-205 targets VEGFA and FGF2 and regulates resistance to chemotherapeutics in breast cancer. Cell Death Dis. 2016. [Google Scholar] [CrossRef]
  85. Sha, L.; Zhang, Y.; Wang, W.; Sui, X.; Liu, S.; Wang, T.; Zhang, H. MiR-18a upregulation decreases Dicer expression and confers Paclitaxel resistance in triple negative breast cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2201–2208. [Google Scholar] [PubMed]
  86. Chen, X.; Wang, Y.W.; Xing, A.Y.; Xiang, S.; Shi, D.B.; Liu, L.; Li, Y.X.; Gao, P. Suppression of SPIN1-mediated PI3K–Akt pathway by miR-489 increases chemosensitivity in breast cancer. J. Pathol. 2016, 239, 459–472. [Google Scholar] [CrossRef] [PubMed]
  87. Venturutti, L.; Russo, R.C.; Rivas, M.A.; Mercogliano, M.F.; Izzo, F.; Oakley, R.; Pereyra, M.; De Martino, M.; Proietti, C.; Yankilevich, P. MiR-16 mediates trastuzumab and lapatinib response in ErbB-2-positive breast and gastric cancer via its novel targets CCNJ and FUBP1. Oncogene 2016, 35, 6189–6202. [Google Scholar] [CrossRef] [PubMed]
  88. Gu, X.; Xue, J.-Q.; Han, S.-J.; Qian, S.-Y.; Zhang, W.-H. Circulating microRNA-451 as a predictor of resistance to neoadjuvant chemotherapy in breast cancer. Cancer Biomarkers 2016, 16, 395–403. [Google Scholar] [CrossRef]
  89. Zhong, S.; Chen, X.; Wang, D.; Zhang, X.; Shen, H.; Yang, S.; Lv, M.; Tang, J.; Zhao, J. MicroRNA expression profiles of drug-resistance breast cancer cells and their exosomes. Oncotarget 2016, 7, 19601–19609. [Google Scholar] [CrossRef]
  90. Zhang, B.; Zhao, R.; He, Y.; Fu, X.; Fu, L.; Zhu, Z.; Fu, L.; Dong, J.-T. Micro RNA 100 sensitizes luminal A breast cancer cells to Paclitaxel treatment in part by targeting mTOR. Oncotarget 2016, 7, 5702–5714. [Google Scholar] [CrossRef]
  91. Shen, R.; Wang, Y.; Wang, C.-X.; Yin, M.; Liu, H.-L.; Chen, J.-P.; Han, J.-Q.; Wang, W.-B. MiRNA-155 mediates TAM resistance by modulating SOCS6-STAT3 signalling pathway in breast cancer. Am. J. Transl. Res. 2015, 7, 2115–2126. [Google Scholar]
  92. Yu, X.; Luo, A.; Liu, Y.; Wang, S.; Li, Y.; Shi, W.; Liu, Z.; Qu, X. MiR-214 increases the sensitivity of breast cancer cells to tamoxifen and fulvestrant through inhibition of autophagy. Mol. Cancer 2015. [Google Scholar] [CrossRef]
  93. Zhou, S.; Huang, Q.; Zheng, S.; Lin, K.; You, J.; Zhang, X. miR-27a regulates the sensitivity of breast cancer cells to cisplatin treatment via BAK-SMAC/DIABLO-XIAP axis. Tumor Biol. 2016, 37, 6837–6845. [Google Scholar] [CrossRef]
  94. Zheng, Y.; Lv, X.; Wang, X.; Wang, B.; Shao, X.; Huang, Y.; Shi, L.; Chen, Z.; Huang, J.; Huang, P. MiR-181b promotes chemoresistance in breast cancer by regulating Bim expression. Oncol. Rep. 2016, 35, 683–690. [Google Scholar] [CrossRef]
  95. Ye, Z.; Hao, R.; Cai, Y.; Wang, X.; Huang, G. Knockdown of miR-221 promotes the cisplatin-inducing apoptosis by targeting the BIM-Bax/Bak axis in breast cancer. Tumor Biol. 2016, 37, 4509–4515. [Google Scholar] [CrossRef] [PubMed]
  96. 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. 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, 37269–37280. [Google Scholar] [CrossRef] [PubMed]
  97. Lu, L.; Ju, F.; Zhao, H.; Ma, X. MicroRNA-134 modulates resistance to doxorubicin in human breast cancer cells by downregulating ABCC1. Biotechnol. Lett. 2015, 37, 2387–2394. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, D.-w.; Mao, L.; Zhang, J.; Jiang, L.-h.; Li, J.; Wu, Y.; Ji, H.; Chen, W.; Wang, J.; Ma, R. MiR-139-5p inhibits the biological function of breast cancer cells by targeting Notch 1 and mediates chemosensitivity to docetaxel. Biochem. Biophys. Res. Commun. 2015, 465, 702–713. [Google Scholar]
  99. He, H.; Tian, W.; Chen, H.; Jiang, K. MiR-944 functions as a novel oncogene and regulates the chemoresistance in breast cancer. Tumor Biol. 2016, 37, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
  100. Ikeda, K.; Horie-Inoue, K.; Ueno, T.; Suzuki, T.; Sato, W.; Shigekawa, T.; Osaki, A.; Saeki, T.; Berezikov, E.; Mano, H. miR-378a-3p modulates tamoxifen sensitivity in breast cancer MCF-7 cells through targeting GOLT1A. Sci. Rep. 2015. [Google Scholar] [CrossRef]
  101. Wu, J.; Li, S.; Jia, W.; Deng, H.; Chen, K.; Zhu, L.; Yu, F.; Su, F. Reduced Let-7a is associated with chemoresistance in primary breast cancer. PLoS ONE 2015. [Google Scholar] [CrossRef]
  102. Takahashi, R.-u.; Miyazaki, H.; Takeshita, F.; Yamamoto, Y.; Minoura, K.; Ono, M.; Kodaira, M.; Tamura, K.; Mori, M.; Ochiya, T. Loss of microRNA-27b contributes to breast cancer stem cell generation by activating ENPP1. Nature Commun. 2015. [Google Scholar] [CrossRef]
  103. Niu, J.; Xue, A.; Chi, Y.; Xue, J.; Wang, W.; Zhao, Z.; Fan, M.; Yang, C.H.; Shao, Z.; Pfeffer, L.M. Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer. Oncogene 2016, 35, 1302–1313. [Google Scholar] [CrossRef]
  104. Su, C.-M.; Wang, M.; Hong, C.; Chen, H.-A.; Su, Y.-H.; Wu, C.-H.; Huang, M.-T.; Chang, Y.W.; Jiang, S.S.; Sung, S.-Y. miR-520h is crucial for DAPK2 regulation and breast cancer progression. Oncogene 2016, 35, 1134–1142. [Google Scholar] [CrossRef]
  105. Boulbes, D.R.; Chauhan, G.B.; Jin, Q.; Bartholomeusz, C.; Esteva, F.J. CD44 expression contributes to trastuzumab resistance in HER2-positive breast cancer cells. Breast Cancer Res. Treat 2015, 151, 501–513. [Google Scholar] [CrossRef] [PubMed]
  106. Manvati, S.; Mangalhara, K.C.; Kalaiarasan, P.; Srivastava, N.; Bamezai, R. miR-24-2 regulates genes in survival pathway and demonstrates potential in reducing cellular viability in combination with docetaxel. Gene 2015, 567, 217–224. [Google Scholar] [CrossRef] [PubMed]
  107. Kang, L.; Mao, J.; Tao, Y.; Song, B.; Ma, W.; Lu, Y.; Zhao, L.; Li, J.; Yang, B.; Li, L. Micro RNA-34a suppresses the breast cancer stem cell-like characteristics by downregulating Notch 1 pathway. Cancer Sci. 2015, 106, 700–708. [Google Scholar] [CrossRef] [PubMed]
  108. Lü, M.; Ding, K.; Zhang, G.; Yin, M.; Yao, G.; Tian, H.; Lian, J.; Liu, L.; Liang, M.; Zhu, T. MicroRNA-320a sensitizes tamoxifen-resistant breast cancer cells to tamoxifen by targeting ARPP-19 and ERRγ. Sci. Rep. 2015. [Google Scholar] [CrossRef] [PubMed]
  109. Ye, F.-G.; Song, C.-G.; Cao, Z.-G.; Xia, C.; Chen, D.-N.; Chen, L.; Li, S.; Qiao, F.; Ling, H.; Yao, L. Cytidine deaminase axis modulated by miR-484 differentially regulates cell proliferation and chemoresistance in breast cancer. Cancer Res. 2015, 75, 1504–1515. [Google Scholar] [CrossRef] [PubMed]
  110. Vilquin, P.; Donini, C.F.; Villedieu, M.; Grisard, E.; Corbo, L.; Bachelot, T.; Vendrell, J.A.; Cohen, P.A. MicroRNA-125b upregulation confers aromatase inhibitor resistance and is a novel marker of poor prognosis in breast cancer. Breast Cancer Res. 2015. [Google Scholar] [CrossRef]
  111. Ujihira, T.; Ikeda, K.; Suzuki, T.; Yamaga, R.; Sato, W.; Horie-Inoue, K.; Shigekawa, T.; Osaki, A.; Saeki, T.; Okamoto, K. MicroRNA-574-3p, identified by microRNA library-based functional screening, modulates tamoxifen response in breast cancer. Sci. Rep. 2015. [Google Scholar] [CrossRef]
  112. Cui, J.; Yang, Y.; Li, H.; Leng, Y.; Qian, K.; Huang, Q.; Zhang, C.; Lu, Z.; Chen, J.; Sun, T. MiR-873 regulates ERα transcriptional activity and tamoxifen resistance via targeting CDK3 in breast cancer cells. Oncogene 2015, 34, 3895–3907. [Google Scholar] [CrossRef]
  113. Lv, J.; Xia, K.; Xu, P.; Sun, E.; Ma, J.; Gao, S.; Zhou, Q.; Zhang, M.; Wang, F.; Chen, F. miRNA expression patterns in chemoresistant breast cancer tissues. Biomed. Pharmacother. 2014, 68, 935–942. [Google Scholar] [CrossRef]
  114. He, X.; Xiao, X.; Dong, L.; Wan, N.; Zhou, Z.; Deng, H.; Zhang, X. MiR-218 regulates cisplatin chemosensitivity in breast cancer by targeting BRCA1. Tumor Biol. 2015, 36, 2065–2075. [Google Scholar] [CrossRef]
  115. Winsel, S.; Mäki-Jouppila, J.; Tambe, M.; Aure, M.; Pruikkonen, S.; Salmela, A.; Halonen, T.; Leivonen, S.; Kallio, L.; Børresen-Dale, A. Excess of miRNA-378a-5p perturbs mitotic fidelity and correlates with breast cancer tumourigenesis in vivo. Brit. J. Cancer 2014, 111, 2142–2151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hu, J.; Xu, J.; Wu, Y.; Chen, Q.; Zheng, W.; Lu, X.; Zhou, C.; Jiao, D. Identification of microRNA-93 as a functional dysregulated miRNA in triple-negative breast cancer. Tumor Biol. 2015, 36, 251–258. [Google Scholar] [CrossRef] [PubMed]
  117. He, D.-X.; Gu, X.-T.; Jiang, L.; Jin, J.; Ma, X. A methylation-based regulatory network for microRNA 320a in chemoresistant breast cancer. Mol. Pharmacol. 2014, 86, 536–547. [Google Scholar] [CrossRef] [PubMed]
  118. He, D.X.; Gu, X.T.; Li, Y.R.; Jiang, L.; Jin, J.; Ma, X. Methylation-regulated miR-149 modulates chemoresistance by targeting Glc NA c N-deacetylase/N-sulfotransferase-1 in human breast cancer. FEBS J. 2014, 281, 4718–4730. [Google Scholar] [CrossRef]
  119. Ouyang, M.; Li, Y.; Ye, S.; Ma, J.; Lu, L.; Lv, W.; Chang, G.; Li, X.; Li, Q.; Wang, S. MicroRNA profiling implies new markers of chemoresistance of triple-negative breast cancer. PLoS ONE 2014. [Google Scholar] [CrossRef]
  120. Luo, M.-L.; Gong, C.; Chen, C.-H.; Lee, D.Y.; Hu, H.; Huang, P.; Yao, Y.; Guo, W.; Reinhardt, F.; Wulf, G. Prolyl isomerase Pin1 acts downstream of miR200c to promote cancer stem–like cell traits in breast cancer. Cancer Res. 2014, 74, 3603–3616. [Google Scholar] [CrossRef]
  121. Jiang, L.; He, D.; Yang, D.; Chen, Z.; Pan, Q.; Mao, A.; Cai, Y.; Li, X.; Xing, H.; Shi, M. MiR-489 regulates chemoresistance in breast cancer via epithelial mesenchymal transition pathway. FEBS Lett. 2014, 588, 2009–2015. [Google Scholar] [CrossRef]
  122. 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. [Google Scholar] [CrossRef]
  123. Zhu, Y.; Wu, J.; Li, S.; Ma, R.; Cao, H.; Ji, M.; Jing, C.; Tang, J. The function role of miR-181a in chemosensitivity to adriamycin by targeting Bcl-2 in low-invasive breast cancer cells. Cell. Physiol. Biochem. 2013, 32, 1225–1237. [Google Scholar] [CrossRef]
  124. Yang, G.; Wu, D.; Zhu, J.; Jiang, O.; Shi, Q.; Tian, J.; Weng, Y. Upregulation of miR-195 increases the sensitivity of breast cancer cells to Adriamycin treatment through inhibition of Raf-1. Oncol. Rep. 2013, 30, 877–889. [Google Scholar] [CrossRef]
  125. Pichiorri, F.; Palmieri, D.; De Luca, L.; Consiglio, J.; You, J.; Rocci, A.; Talabere, T.; Piovan, C.; Lagana, A.; Cascione, L. In vivo NCL targeting affects breast cancer aggressiveness through miRNA regulation. J. Exp. Med. 2013, 210, 951–968. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Ji, S.; Shao, G.; Lv, X.; Liu, Y.; Fan, Y.; Wu, A.; Hu, H. Downregulation of mi RNA-128 sensitises breast cancer cell to chemodrugs by targeting Bax. Cell Biol. Int. 2013, 37, 653–658. [Google Scholar] [CrossRef] [PubMed]
  128. Hu, H.; Li, S.; Cui, X.; Lv, X.; Jiao, Y.; Yu, F.; Yao, H.; Song, E.; Chen, Y.; Wang, M. The overexpression of hypomethylated miR-663 induces chemotherapy resistance in human breast cancer cells by targeting heparin sulfate proteoglycan 2 (HSPG2). Int. J. Biol. Chem. 2013, 288, 10973–10985. [Google Scholar] [CrossRef] [PubMed]
  129. Masuda, M.; Miki, Y.; Hata, S.; Takagi, K.; Sakurai, M.; Ono, K.; Suzuki, K.; Yang, Y.; Abe, E.; Hirakawa, H. An induction of microRNA, miR-7 through estrogen treatment in breast carcinoma. J. Transl. Med. 2012. [Google Scholar] [CrossRef]
  130. Li, X.-j.; Ji, M.-h.; Zhong, S.-l.; Zha, Q.-b.; Xu, J.-j.; Zhao, J.-h.; Tang, J.-h. MicroRNA-34a modulates chemosensitivity of breast cancer cells to adriamycin by targeting Notch 1. Arch. Med. Res. 2012, 43, 514–521. [Google Scholar] [CrossRef]
  131. Lv, K.; Liu, L.; Wang, L.; Yu, J.; Liu, X.; Cheng, Y.; Dong, M.; Teng, R.; Wu, L.; Fu, P. Lin28 mediates Paclitaxel resistance by modulating p21, Rb and Let-7a miRNA in breast cancer cells. PLoS ONE 2012. [Google Scholar] [CrossRef]
  132. 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. [Google Scholar] [CrossRef]
  133. Chen, J.; Tian, W.; Cai, H.; He, H.; Deng, Y. Down-regulation of microRNA-200c is associated with drug resistance in human breast cancer. Med. Oncol. 2012, 29, 2527–2534. [Google Scholar] [CrossRef]
  134. Zhu, Y.; Yu, F.; Jiao, Y.; Feng, J.; Tang, W.; Yao, H.; Gong, C.; Chen, J.; Su, F.; Zhang, Y. Reduced miR-128 in breast tumor–initiating cells induces chemotherapeutic resistance via Bmi-1 and ABCC5. Clin. Cancer Res. 2011, 17, 7105–7115. [Google Scholar] [CrossRef]
  135. Zhao, Y.; Deng, C.; Lu, W.; Xiao, J.; Ma, D.; Guo, M.; Recker, R.R.; Gatalica, Z.; Wang, Z.; Xiao, G.G. let-7 microRNAs induce tamoxifen sensitivity by downregulation of estrogen receptor α signaling in breast cancer. Mol. Med. 2011, 17, 1233–1241. [Google Scholar] [CrossRef] [PubMed]
  136. 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] [PubMed]
  137. Cittelly, D.M.; Das, P.M.; Spoelstra, N.S.; Edgerton, S.M.; Richer, J.K.; Thor, A.D.; Jones, F.E. Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol. Cancer 2010. [Google Scholar] [CrossRef] [PubMed]
  138. Maillot, G.; Lacroix-Triki, M.; Pierredon, S.; Gratadou, L.; Schmidt, S.; Bénès, V.; Roché, H.; Dalenc, F.; Auboeuf, D.; Millevoi, S. Widespread estrogen-dependent repression of micrornas involved in breast tumor cell growth. Cancer Res. 2009, 69, 8332–8340. [Google Scholar] [CrossRef]
  139. Iorio, M.V.; 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]
  140. Yu, F.; Yao, H.; Zhu, P.; Zhang, X.; Pan, Q.; Gong, C.; Huang, Y.; Hu, X.; Su, F.; Lieberman, J. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell 2007, 131, 1109–1123. [Google Scholar] [CrossRef]
  141. Li, G.; Wu, X.; Qian, W.; Cai, H.; Sun, X.; Zhang, W.; Tan, S.; Wu, Z.; Qian, P.; Ding, K. CCAR1 5′ UTR as a natural miRancer of miR-1254 overrides tamoxifen resistance. Cell Res. 2016, 26, 655–673. [Google Scholar] [CrossRef]
  142. Yu, S.-J.; Yang, L.; Hong, Q.; Kuang, X.-Y.; Di, G.-H.; Shao, Z.-M. MicroRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer. BMC Cancer 2018. [Google Scholar] [CrossRef]
  143. Lee, J.W.; Guan, W.; Han, S.; Hong, D.K.; Kim, L.S.; Kim, H. Micro RNA-708-3p mediates metastasis and chemoresistance through inhibition of epithelial-to-mesenchymal transition in breast cancer. Cancer Sci. 2018, 109, 1404–1413. [Google Scholar] [CrossRef]
  144. Si, W.; Shen, J.; Du, C.; Chen, D.; Gu, X.; Li, C.; Yao, M.; Pan, J.; Cheng, J.; Jiang, D. A miR-20a/MAPK1/c-Myc regulatory feedback loop regulates breast carcinogenesis and chemoresistance. Cell Death Differ. 2018, 25, 406–420. [Google Scholar] [CrossRef]
  145. Cheng, S.; Huang, Y.; Lou, C.; He, Y.; Zhang, Y.; Zhang, Q. FSTL1 enhances chemoresistance and maintains stemness in breast cancer cells via integrin β3/Wnt signaling under miR-137 regulation. Cancer Biol. Ther. 2019, 20, 328–337. [Google Scholar] [CrossRef] [PubMed]
  146. Hu, G.; Zhao, X.; Wang, J.; Lv, L.; Wang, C.; Feng, L.; Shen, L.; Ren, W. miR-125b regulates the drug-resistance of breast cancer cells to doxorubicin by targeting HAX-1. Oncol. Lett. 2018, 15, 1621–1629. [Google Scholar] [CrossRef] [PubMed]
  147. Sabarimurugan, S.; Kumarasamy, C.; Baxi, S.; Devi, A.; Jayaraj, R. Systematic review and meta-analysis of prognostic microRNA biomarkers for survival outcome in nasopharyngeal carcinoma. PLoS ONE 2019, 14, e0209760. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of our literature search.
Figure 1. Flowchart of our literature search.
Cells 08 01250 g001
Figure 2. Forest plot of the studies included in our meta-analysis. BC: breast cancer.
Figure 2. Forest plot of the studies included in our meta-analysis. BC: breast cancer.
Cells 08 01250 g002
Figure 3. Funnel plot of the studies included in our meta-analysis.
Figure 3. Funnel plot of the studies included in our meta-analysis.
Cells 08 01250 g003
Table 1. Main characteristics of the included studies.
Table 1. Main characteristics of the included studies.
AuthorEthnicity (Patient)Period of StudyDrug(s)Clinical Stages No. of Samples (Cancer/Normal)miRNAmiRNA Profiling Platform
Total stagesIIIIIIIV
Lin X et al. (2017) [69]Chinese2001 to 2006 and 2015docetaxel2 stages (I–II and III)744600138/8334aGeneSpring GX (Agilent Technologies, Capital Biochip Corporation)
Zhao G et al. (2017) [39]ChineseJanuary 2012 to November 2015docetaxelNMNMNMNMNM78/78638qRT-PCR- SYBR Premix ExTaqTM (Takara, USA)
Nakano M et al. (2017) [70]JapaneseNMmethotrexate3 stages (I, I–II, II, II–III)1211NM19/1925-3p and 125a-3pMx3000P (Stratagene, La Jolla, CA)
Miao Y et al. (2017) [71]ChineseJanuary 2014 to March 2016doxorubicinNMNMNMNMNM29/29130bSYBR Green qRT-PCR master mix (TaKaRa, Otsu, Shiga, Japan)
Chen M-J et al. (2017) [72]TaiwaneseNMtamoxifenNMNMNMNMNM36a148a, 152ABI 7900 and SYBR® Select Master Mix (Applied Biosystems).
Yang F et al. (2017) [73]Chinese2012–2015docetaxelNMNMNMNMNM24/24346ABI 7300 real-time PCR machine (Applied Biosystems, USA)
Gong J-P et al. (2016) [74]ChineseJuly 2010 to June 2014PaclitaxelNMNMNMNMNM40a24TaqMan™ MicroRNA Assays (Applied Biosystems; Thermo Fisher Scientific, Inc.)
Ao X et al. (2016) [44]Chinese2009–2011taxol3 stages (II, III and III-IV)012182555/5517 and 20bSYBR on the CFX96 system (Bio-Rad).
Zhu J et al. (2016) [75]Chinese2005–2009tamoxifen3 stages (II, III and III–IV)08222273/1927b-3pSYBR on the CFX96 system (Bio-Rad)
Chen X et al. (2016) [76]ChineseJanuary 2010 to February 2015docetaxel, epirubicin and vinorelbineNMNMNMNMNM55/2629a, 34a, 90b, 130a, 138, 139, 140, 149, 197, 200b, 210, 222, 423, 452, 574, 671, 744, 1246, 1268a, 3178, 3613, 4258, 4298, 4644, 6780b, 7107 and 7847SYBR® Advantage® qPCR Premix, Light cycler system (Roche, Australia)
Damiano V et al. [77]Italian2000–2010anthracycline, anthracycline + taxane and CMF2 stages (I–II and III)2 48051a200cTaqMan normalizer (Applied Biosystems, ThermoFisher Scientific)
Jana S et al. (2016) [78]IndianNMNMNMNMNMNMNM35/35216bSYBR green detection system
Wang D et al. (2016) [79]Chinese2010–2015doxorubicinNMNMNMNMNM21a222SYBR Premix Ex Taq system (Roche, Australia)
Xu X et al. (2016) [80]NM2011–2014docetaxelNMNMNMNMNM37/37125a-3pSYBR Premix ExTaqTM (Takara, USA)
Chen X et al. (2016) [81]ChineseJanuary 2010 to February 2015epirubicin3 stages (I, II and III)10324076a4443MiR-X miRNA qRT-PCR SYBR Kit (638314; Clontech Laboratories, USA)
Gao M et al. (2016) [82]ChineseNMdoxorubicinNMNMNMNMNM55/21145NCode VILO miRNA cDNA Synthesis Kit and the EXPRESS SYBR GreenER miRNA qRT-PCR Kit, respectively (Invitrogen, Carlsbad, CA, USA)
Thakur S et al. (2016) [83]IndianNMNM2 stages (I–II and III–IV)47 38 100/10021, 145, 195, 210, 221 and Let-7aTaqMan Universal Master Mix kit (Applied Biosystems, USA)
Hu Y et al. (2016) [84]ChineseJune 2014 to June 2015docetaxel, doxorubicin and cyclophosphamide3 stages (II, III and III–IV)0719430a205TaqMan assays (Life Technologies)
Sha L-Y et al. (2016) [85]ChineseNMepirubicin plus PaclitaxelNMNMNMNMNM20/2018aTaqMan MicroRNA Assay Kit (Applied Biosystems)
Chen X et al. (2016) [86]Chinese2008–2013doxorubicin4 stages (I, II, III and IV)3764123114/114489SYBR Primescript miRNA RT PCR Kit (TaKaRa, Dalian, China)
Venturutti L et al. (2016) [87]Argentinians2008–2014trastuzumab and lapatinib4 stages (I, II, III and IV)593219a16TaqMan® MicroRNA assay (Ambion)
Gu X et al. (2016) [88]ChineseJanuary 2010 to December 2013epirubicin and docetaxel2 stages (II and III)NMNMNMNM82/60451miScript SYBR Green PCR Kit (QIAGEN, Hilden, Germany) and a real-time LightCycler PCR (Roche Molecular Biochemicals, Mannheim, Germany)
Zhong S et al. (2016) [89]ChineseJanuary 2010 to February 2015docetaxel, epirubicin and vinorelbine3 stages (I, II and III)689023a138-5p, 139-5p, 140-3p, 149-3p, 197-3p, 210-3p, 423-5p, 574-3p, 744-5p, 1246, 1268a, 3178, 4258, 4298, 4443, 4644, 6780b-3p, 7107-5p and 7847-3p Affymetrix GeneChip miRNA 4.0 Array
Zhang B et al. (2015) [90]ChineseNMPaclitaxel NMNMNMNMNM36/36100Realplex Real-time PCR Detection System (Eppendorf, Beijing, China)
Shen R et al. (2015) [91]ChineseBetween January 2006 to December 2011tamoxifenNMNMNMNMNM18a155SYBR Green PCR master mix (TaKaRa) on the ABI 7500HT System
Yu X et al. (2015) [92]ChineseNMtamoxifen and fulvestrantNMNMNMNMNM20/20214MiScript SYBR Green PCR kit (Qiagen)
Zhou S et al. (2015) [93]ChineseMarch 2014 to June 2015cisplatinNMNMNMNMNM40/4027aFastStart Universal STBR Green Master (Roche, Switzerland)
Zheng Y et al. (2015) [94]ChineseNMdoxorubicinNMNMNMNMNM30/30181bTaqMan MicroRNA assays kit (Applied Biosystems, USA)
Ye Z et al. (2015) [95]ChineseNMcisplatinNMNMNMNMNM85/85221SYBR Green (Takara)
Mattos-Arruda L-D et al. (2015) [96]Spaniards2005–2011trastuzumab, anthracyclines, taxanes NMNMNMNMNM85a21LightCycler 480 Real-Time PCR System (Roche)
Lu L et al. (2015) [97]ChineseNot mentioneddoxorubicin, cyclophosphamide and fluorouracil2 stages (II–III)NMNMNMNM40a134SYBR PrimeScript miRNA RT-PCR Kit (Takara, Japan)
Zhang H-d et al. (2015) [98]Chinese2012–2015docetaxel2 stages (I–II and III)18 17035a139TaqMan MicroRNA Assay Kit (assay ID: miR-139-5p: 002289, and RNU6B: 001093), (Applied Biosystems, Life Technologies)
He H et al. (2015) [99]ChineseOctober 2012 to January 2015cisplatinNMNMNMNMNM70/70944ABI PRISM 7900 Sequence Detection System (Applied Biosystems) with SYBR Green (TaKaRa, Japan)
Ikeda K et al. (2015) [100]JapaneseNot mentionedtamoxifenNMNMNMNMNM40/16378a-3pTaqMan microRNA assays (Applied Biosystems, CA, USA)
Wu J et al. (2015) [101]ChineseJanuary 2005 to December 2006before therapyNMNMNMNMNM39aLet7aReal-time quantitative reverse transcription PCR (qRT-PCR)
January 2008 to December 2009epirubicinNMNMNMNMNM31a
Takahashi R et al. (2015) [102]Japanese1996–2000docetaxel1 stage (II–III)NM26 NM26/927bTaqMan MicroRNA Assays (Applied Biosystems)
Niu J et al. (2015) [103]Chinese1 January 2009 to 31 December 2010doxorubicin2 stages (I–II and III–IV)49 13 62a181aMyiQ Real-Time PCR Detection System (Bio-Rad)
Su C-M et al. (2015) [104]TaiwaneseNMPaclitaxel2 stages (I and I–II)36110NMNM146a520hApplied Biosystems 7900 Fast Real-Time PCR
Boulbes D et al. (2015) [105]AmericanNMtrastuzumab, fluorouracil, epirubicin and cyclophosphamideNMNMNMNMNM50ahas-520b-5p, 532-3p, 548n and 34a-3pmiRNA microarray (version 4.0, microRNACHIPv4)
Manvati S et al. (2015) [106]IndianNMdocetaxel3 stages (I, II and III)NMNMNMNM46/4624-2TaqMan microRNA assays (Applied Biosystems)
Kang L et al. (2015) [107]ChineseNMPaclitaxel4 stages (I, II, III and IV)111812445a34aTaqMan MicroRNA Assay kit (Applied Biosystems, Foster City, CA, USA)
Lu M et al. (2015) [108]Chinese2009–2010tamoxifenNMNMNMNMNM31/27320aApplied Biosystems Step One real-time PCR system using an SYBR Premix Ex Taq II Kit (Takara Bio, Inc., Shiga, Japan)
Ye F-G et al. (2015) [109]ChineseSeptember 2013gemcitabine3 stages (I, II and III)159 32NM400/243484SYBR Premix Ex Taq System (TaKaRa)
Vilquin P et al. (2015) [110]FrenchNMletrozole, anastrazole, tamoxifen and fulvestrant3 stages (I, II and III)41823065/65125bExiLENT SYBR Green Master Mix and CFX96 (BioRad, Marne-laCoquette, France)
Ujihira T et al. (2015) [111]JapaneseNMtamoxifenNMNMNMNMNM19a574-3ptriplicate TaqMan microRNA assays (Applied Biosystems, CA, USA)
Cui J et al. (2014) [112]ChineseNMtamoxifenNMNMNMNMNMNM873RNeasy Mini kit (Qiagen, Hilden, Germany) or TRIzol (Invitrogen) reagent. SYBR Green PCR Master Mix reagents using an ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, CA, USA)
Lv J et al. (2014) [113]Chinese2008–2009doxorubicinNMNMNMNMNMNM31, 125b-1, 141, 145, 196b, 200a, 200c, 370, 429, 491-3p, 576, 760, 765 and Let-7a ABI 7900 PCR System (Applied Biosystems, USA) using Power SYBR Green PCR Master Mix (2X, Applied Biosystems)
He X et al. (2014) [114]ChineseNMcisplatin4 stages (I, II, III and IV)15 + 1715 + 1730 + 2330 + 2385a218TRIzol reagent (Invitrogen) miRNA microarray chip (v.10.0, Exiqon, Vedbaek, Denmark)
Winsel S et al. (2014) [115]NorwegiansMay 1995 to December 1998taxolNMNMNMNMNM101a378a-3pRNeasy Mini Kit (Qiagen) TaqMan Universal Master Mix II, no PNG (Applied Biosystems, Foster City, CA, USA)
Hu J et al. (2014) [116]ChineseNMNM4 stages (I, II, III and IV)2025314119a93TRIzol Reagent (Invitrogen) and the miRNeasy Mini Kit (QIAGEN)
He DX et al. (2014) [117]ChineseNMdoxorubicin, PaclitaxelNMNMNMNMNMNM320aAll-in-One miRNA qRT-PCR detection kit (GeneCopoeia, Rockville, MD, USA)
He DX et al. (2014) [118]ChineseNMdoxorubicin, PaclitaxelNMNMNMNMNMNM149All-in-One miRNA qRT-PCR detection kit (GeneCopoeia, Rockville, MD, USA). Briefly, total RNA was extracted from MCF-7/WT and ADM cells with TRIzol (Invitrogen, Carlsbad, CA, USA)
Ouyang M et al. (2014) [119]Chinese2011 (January–October)doxorubicinNMNMNMNMNMNM10b-5p, 21-3p, 31-5p, 125b-3p, 130a-3p, 155-5p, 181a-5p, 181b-5p, 183-5p, 195-5p and 451aTotal RNA was harvested using TRIzol (Invitrogen) and miRNAeasy mini kit (QIAGEN). SYBR Premix EX TaqTM II kit (Takara, Dalian, China)
Luo ML et al. (2014) [120]ChineseNMPiBNMNMNMNMNMNM200Total RNA was isolated from miRNeasy kit (Qiagen) and reversely transcribed by miScript PCR starter kit
Jiang L et al. (2014) [121]ChineseNMdoxorubicinNMNMNMNMNMNM489Total RNA was prepared using TRIzol (Beyotime, China) according to the manufacturer’s instructions.
Ye XM et al. (2014) [122]ChineseNMtrastuzumab/HerceptinNMNMNMNMNMNM375Total RNA was extracted from each cell line using TRIzol reagent (Invitrogen, USA)
Zhu Y et al. (2013) [123]ChineseNMdoxorubicin2 stages (I and II)349NMNM43a181aTotal RNA was extracted from each cell line using TRIzol reagent (Invitrogen, Carlsbad, CA, USA)
Ye X et al. (2014) [122]ChineseNMtrastuzumabNMNMNMNMNMNM221Total RNA from each cell line was extracted by TRIzol reagent (Invitrogen, USA)
Yang G et al. (2013) [124]ChineseNMdoxorubicin2 stages (I and II)98NMNM30a195Total cellular RNA from tissues and cultured cells were isolated using a TRIzol Reagent (Invitrogen)
Pichiorri F et al. (2013) [125]AmericansNMfulvestrantNMNMNMNMNM183/5721, 103, 221 and 222TaqMan PCR kit (Applied Biosystems) and 7900HT Sequence Detection System (Applied Biosystems)
Wang H-J et al. (2013) [126]ChineseJanuary 2010 to December 2011Paclitaxel, 5-FU, epirubicin and cyclophosphamideNMNMNMNMNM19/19125bABI 7900HT system (Applied Biosystems)
Ji S et al. (2013) [127]Chinese2007–2009taxol + doxorubicin + cyclophosphamideNMNMNMNMNM67/67128QRT-PCR
Hu H et al. (2013) [128]ChineseOctober 2003 to July 2010topotecan, etoposide, doxorubicin, docetaxel and cyclophosphamideNMNMNMNMNM39/39663Conventional TaqMan PCR (Bio-Rad)
Masuda M et al. (2011) [129]JapaneseNM estradiol (E2)NMNMNMNMNM41a7PCR was performed in ABI7500 Real-Time PCR System (Applied Biosystems, Foster city, CA, USA)
Li X et al. (2012) [130]Chinese2008–2010doxorubicin, cyclophosphamide (CTX) and 5-fluorouracil (5-FU)1 stage (II)0380038/3834aSYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA)
Lv K et al. (2012) [131]Chinese2002–2010Paclitaxel, vincristineNMNMNMNMNM9/9Lin28Real-time PCR was performed using the TaqMan MicroRNA Reverse Transcription Kit and the Fast Real-Time PCR System (Applied Biosystems, Carlsbad, CA, USA)
Wang H et al. (2012) [132]Chinese2009–20105-FU (5-fluorouracil)2 stages (II and III)03521056/1010b, 34a, 125b and 155miRNA-specific TaqMan MicroRNA Assays (Applied Biosystems)
Jung E-J et al. (2012) [43]Americans, KoreansNMtrastuzumab, Paclitaxel, fluorouracil, cyclophosphamide and epirubicin3 stages (I, II and III)33318072/7221, 29a, 126 and 210TaqMan MicroRNA Assay kit (Applied Biosystems, Foster City, Calif)
Chen J et al. (2011) [133]Chinese2007–2011doxorubicinNMNMNMNMNM39a200c Real-time PCR was performed using SYBR Green PCR Master Mix (Applied Biosystems, USA) on the Stepone plus system (Applied Biosystems, USA)
Zhu Y et al. (2011) [134]Chinese2004–2011NM3 stages (II, III and IV)NM4429477a128 Mature miRNA expression analysis was conducted using a TaqMan MicroRNA Assays (Applied Biosystems)
Zhao Y et al. (2011) [135]NMNMtamoxifenNMNMNMNMNM29/15Let-7mirVana miRNA isolation kit (Ambion Inc., Austin, TX, USA) or from FFPE tissues using the miRNeasy FFPE Kit (Qiagen, Valencia, CA, USA)
Gong C et al. (2011) [136]Chinese2008–2009trastuzumab (Herceptin)NMNMNMNMNM32a21Total RNA was harvested using TRIzol (Invitrogen) and the RNeasy minikit (Qiagen) according to the manufacturer’s instructions.
Shi W et al. (2011) [42]NMNMNM3 stages (I, II and III)83330NM71a301Standard TaqMan MicroRNA Assay (Applied Biosystems)
Cittelly D et al. (2010) [137]Americans1978–1993tamoxifen3 stages (I, II and III)72346322NM791a342miRVANA RNA Isolation System (Ambion)
Liang Z et al. (2010) [41]AmericansNMVP-16, mitoxantrone3 stages (I, III and IV)5NM10 (III and IV)10 (III and IV)35a326Total RNA was extracted from 70% to 85% confluence of MCF-7 and MCF-7/VP cells with TRIzol (Invitrogen, Carlsbad, CA, USA)
Maillot G et al. (2009) [138]NMNMtamoxifen2 stages (III and IV)NMNM51015a21, 23b, 26a, 26b, 27b, 181a, 181b and 200c miRNA microarray analysis was performed as described by Castoldi and colleagues
Iorio M et al. (2009) [139]ItaliansNMNMNMNMNMNMNMNM205TaqMan MicroRNA Reverse Transcription kit and TaqMan MicroRNA Assay were used to detect and quantify mature microRNA-205 (Applied Biosystems)
Miller T et al. (2008) [40]AmericansNMtamoxifenNMNMNMNMNM76a221 and 222The miRNA microarray was performed at the Ohio State University Comprehensive Cancer Center Microarray Core Facility
Yu F et al. (2007) [140]ChineseNMepirubicinNMNMNMNMNM25aLet-7NM
Li G et al. (2016) [141]Chinese2001–2002tamoxifenNMNMNMNMNM57/571254mirVana miRNA isolation kit (Ambion) using stem-loop RT primers and analysed by qPCR (TaqMan, TaKaRa)
Yu S-J et al. (2018) [142]Chinese2003–2009Paclitaxel and carboplatin2 stages (II and III)NM2844NM110/110200a-5p7900HT Fast Real-Time PCR System (Applied Biosystems)
Lee J-W et al. (2017) [143]South KoreanNMdoxorubicin2 stages (I–II and III–IV)28NM21NM50/50708-3pHigh-Capacity cDNA Reverse Transcription Kit (Life Technologies)
Si W et al. (2018) [144]ChineseNMPaclitaxel3 stages (I, II and III)1538530106/10620aSYBR Premix Ex Taq (TaKaRa, RR420A)
Cheng S et al. (2018) [145]ChineseNMcisplatin and doxorubicinNMNMNMNMNM57/31137ABI Prism 7900HT thermal cycler (Applied Biosystems, Foster City, CA, USA)
Hu G et al. (2018) [146]ChineseAugust 2013 to December 2015doxorubicinNMNMNMNMNM30a 125bABI PRISM 7900 Sequence Detection system (Applied Biosystems)
NM: Not Mentioned; a: only cancer tissue; CMF: Cyclophosphamide, Methotrexate, Fluorouracil.
Table 2. Pathways involved in chemoresistance.
Table 2. Pathways involved in chemoresistance.
DownregulatedUpregulated
DrugmiRNAGene/PathwayDrugmiRNAGene/Pathway
5-FU134ABCC15-FU125bEMT
anastrozole424Akt/mTOR pathway5-FU125bTranscription factor E2F3
anthracycline200cZEB1anthracycline21IL-6/STAT3/NF-κB/PI3K pathway.
anthracycline + taxane200cZEB1cisplatin944Bcl2/BNIP3
CMF200cZEB1cisplatin and doxorubicin137FSTL1/integrin β3/Wnt
CTX134ABCC1CTX125bEMT
docetaxel451NMCTX663HSPG2
docetaxel24-2YWHAZ, TP53, SMAD3, ESR1 and CREBBPdocetaxel663HSPG2
doxorubicin145MRP1doxorubicin130bPTEN/PI3K/Akt
doxorubicin320aTRPC5, NFATC3 and ETS-1 genedoxorubicin222PTEN/Akt/cyclin-dependent kinase (p27) pathway
doxorubicin149GlcNAc-NDST1doxorubicin181bMMP/caspase pathway
doxorubicin103NCLdoxorubicin663HSPG2
doxorubicin222NCLdoxorubicin31MAPK signalling pathway, cytokine–cytokine receptor interaction
doxorubicin134ABCC1doxorubicin141MAPK signalling pathway, cytokine–cytokine receptor interaction
doxorubicin181aSTAT3/NF-kB/MSK1doxorubicin200cMAPK signalling pathway, cytokine–cytokine receptor interaction
doxorubicin10b-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genesdoxorubicin181b-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
doxorubicin125b-3pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genesdoxorubicin183-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
doxorubicin155-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genesdoxorubicin195-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
doxorubicin181a-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genesdoxorubicin21-3pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
doxorubicin31-5pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genesE2124EGFR
doxorubicin200cMDR1 mRNAE229aEGFR
doxorubicin708-3pZEB1/CDH2/vimentinE221EGFR
doxorubicin125bHAX-1E2181dEGFR
E2301aEGFRE234c-5pEGFR
E220aEGFRepirubicin4443TIMP2
E2149EGFRepirubicin + Paclitaxel18aDicer
E217EGFRepirubicin125bEMT
E225EGFRetoposide663HSPG2
E2191EGFRfulvestrant125bAkt/mTOR pathway
E227bEGFRletrozole205Akt/mTOR pathway
E2148aEGFRPaclitaxel520hDAPK2
E2210EGFRPaclitaxelLin28p21, RB, cyclin B1, Akt and Let-7 miRNA
E27EGFRPaclitaxel125bEMT
epirubicinLet7aH-RAS/HMGA2Paclitaxel and carboplatin200a-5pTP53INP1/YAP1
epirubicinLet7aH-RAS/HMGA2tamoxifen222p27Kip1
epirubicin451NMtamoxifen221p27Kip1
fulvestrant21NCLtaxanes21IL-6/STAT3/NF-κB/PI3K pathway
methotrexate25-3pADAR1/DHFRtaxol378a-3pTriggered receptor tyrosine kinase–MAP kinase pathway signalling, suppression of Aurora B kinase
methotrexate125a-3pADAR1/DHFRtopotecan663HSPG2
Paclitaxel320aTRPC5 gene; NFATC3gene; ETS-1 genetrastuzumab21IL-6/STAT3/NF-κB/PI3K pathway
Paclitaxel149GlcNAc-NDST1trastuzumab221PTEN
Paclitaxel20aMAPK1/c-Myctrastuzumab21PTEN
tamoxifen574-3pCLTCvincristineLin28p21, RB, cyclin B1
tamoxifen873CDK3, Erα
tamoxifen424Akt/mTOR pathway
taxol17NCOA3
taxol20bNCOA3
trastuzumab221NCL
trastuzumab375IGF1R
anthracyclin: epirubicin/doxorubicin; EMT: Epithelial-Mesenchymal Transition.
Table 3. Pathways involved in chemosensitivity.
Table 3. Pathways involved in chemosensitivity.
DownregulationUpregulation
DrugmiRNAGene/PathwayDrugmiRNAGene/Pathway
CTX205VEGF/FGF25-FU34aNotch 1
cisplatin218BRCA1CTX34aNotch 1
doxorubicin489Smad3, EMTcisplatin27aBAK-SMAC/DIABLO-XIAP Pathway
doxorubicin181aBcl-2cisplatin221BIM/Bcl-2/Bax/Bak
docetaxel34aC22ORF28docetaxel346SRCIN1
docetaxel638STARD10doxorubicin196bMAPK signalling pathway, cytokine–cytokine receptor interaction
docetaxel125a-3pBRCA1doxorubicin200aMAPK signalling pathway, cytokine–cytokine receptor interaction
doxorubicin195Raf-1doxorubicin34aNotch 1
docetaxel139Notch 1doxorubicin451aPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
docetaxel27bENPP1doxorubicin429MAPK signalling pathway, cytokine–cytokine receptor interaction
docetaxel205VEGF/FGF2gemcitabine484CDA/Cyclin-dependent kinase
doxorubicin145MAPK signalling pathway, cytokine–cytokine receptor interactionlapatinib16CCNJ/FUBP1
doxorubicin370MAPK signalling pathway, cytokine–cytokine receptor interactiontamoxifen148aALCAM
doxorubicin576-3pMAPK signalling pathway, cytokine–cytokine receptor interactiontamoxifen152ALCAM
doxorubicin760MAPK signalling pathway, cytokine–cytokine receptor interactiontamoxifenLet-7MAPK/Akt, ER-α36
doxorubicin765MAPK signalling pathway, cytokine–cytokine receptor interactiontamoxifen155SOCS6-STAT3 signalling pathway
doxorubicin125b-1MAPK signalling pathway, cytokine–cytokine receptor interactiontaxol + doxorubicin + cyclophosphamide128Bax
doxorubicinLet-7aMAPK signalling pathway, cytokine–cytokine receptor interactiontrastuzumab16CCNJ/FUBP1
doxorubicin130a-3pPTEN/Akt, MAPK, RhoA, FOXO3 and PDCD4 genes
doxorubicin205VEGF/FGF2
epirubicinLet-7HMGA2
fulvestrant214UCP2/PI3K-Akt-mTOR pathway
mitoxantrone326MRP-1
Paclitaxel24ABCB9
Paclitaxel34aNotch 1
Paclitaxel100mTOR
PiB200Pin1
tamoxifen342Cyclin B1, p53, BRCA1 gene
tamoxifen27b-3pNR5A2/CREB1
tamoxifen378a-3pGOLT1A
tamoxifen320aARPP-19/ERRᵧ, c-Myc, Cyclin D1
tamoxifen21Estrogen-dependent cellular functions
tamoxifen181aEstrogen-dependent cellular functions
tamoxifen181bEstrogen-dependent cellular functions
tamoxifen200cEstrogen-dependent cellular functions
tamoxifen23bEstrogen-dependent cellular functions
tamoxifen26aEstrogen-dependent cellular functions
tamoxifen26bEstrogen-dependent cellular functions
tamoxifen27bEstrogen-dependent cellular functions
tamoxifen1254CCAR1
tamoxifen214UCP2/PI3K-Akt-mTOR pathway
VP-16326MRP-1
Anthracyclin: epirubicin/doxorubicin.

Share and Cite

MDPI and ACS Style

Jayaraj, R.; Madhav, M.R.; Nayagam, S.G.; Kar, A.; Sathyakumar, S.; Mohammed, H.; Smiti, M.; Sabarimurugan, S.; Kumarasamy, C.; Priyadharshini, T.; et al. Clinical Theragnostic Relationship between Drug-Resistance Specific miRNA Expressions, Chemotherapeutic Resistance, and Sensitivity in Breast Cancer: A Systematic Review and Meta-Analysis. Cells 2019, 8, 1250. https://doi.org/10.3390/cells8101250

AMA Style

Jayaraj R, Madhav MR, Nayagam SG, Kar A, Sathyakumar S, Mohammed H, Smiti M, Sabarimurugan S, Kumarasamy C, Priyadharshini T, et al. Clinical Theragnostic Relationship between Drug-Resistance Specific miRNA Expressions, Chemotherapeutic Resistance, and Sensitivity in Breast Cancer: A Systematic Review and Meta-Analysis. Cells. 2019; 8(10):1250. https://doi.org/10.3390/cells8101250

Chicago/Turabian Style

Jayaraj, Rama, Madurantakam Royam Madhav, Sankaranarayanan Gomathi Nayagam, Ananya Kar, Shubhangi Sathyakumar, Hina Mohammed, Maria Smiti, Shanthi Sabarimurugan, Chellan Kumarasamy, T. Priyadharshini, and et al. 2019. "Clinical Theragnostic Relationship between Drug-Resistance Specific miRNA Expressions, Chemotherapeutic Resistance, and Sensitivity in Breast Cancer: A Systematic Review and Meta-Analysis" Cells 8, no. 10: 1250. https://doi.org/10.3390/cells8101250

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