Next Article in Journal
A Role of DNA Methylation within the CYP17A1 Gene in the Association of Genetic and Environmental Risk Factors with Stress-Related Manifestations of Schizophrenia
Next Article in Special Issue
Systematic Review of the Diagnostic and Clinical Utility of Salivary microRNAs in Traumatic Brain Injury (TBI)
Previous Article in Journal
Epigenetic Changes in Prion and Prion-like Neurodegenerative Diseases: Recent Advances, Potential as Biomarkers, and Future Perspectives
Previous Article in Special Issue
Quantitative Prediction of Steatosis in Patients with Non-Alcoholic Fatty Liver by Means of Hepatic MicroRNAs Present in Serum and Correlating with Hepatic Fat
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Circulating Small EVs miRNAs as Predictors of Pathological Response to Neo-Adjuvant Therapy in Breast Cancer Patients

1
Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania
2
Department of Pharmaceutical Technology and Biopharmaceutics, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
3
Department of Molecular Biology and Biotechnology, “Babes-Bolyai” University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
4
Department of Radiology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania
5
Department of Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania
6
Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
7
Electron Microscopy Center “Prof. C. Craciun”, Faculty of Biology & Geology, “Babes-Bolyai” University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
8
Electron Microscopy Integrated Laboratory, National Institute for R&D of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors share the last authorship.
Int. J. Mol. Sci. 2022, 23(20), 12625; https://doi.org/10.3390/ijms232012625
Submission received: 15 September 2022 / Revised: 15 October 2022 / Accepted: 18 October 2022 / Published: 20 October 2022
(This article belongs to the Special Issue The Role of microRNA in Human Diseases)

Abstract

:
Neo-adjuvant therapy (NAT) is increasingly used in the clinic for the treatment of breast cancer (BC). Pathological response to NAT has been associated with improved patients’ survival; however, the current techniques employed for assessing the tumor response have significant limitations. Small EVs (sEVs)-encapsulated miRNAs have emerged as promising new biomarkers for diagnosis and prediction. Therefore, our study aims to explore the predictive value of these miRNAs for the pathological response to NAT in BC. By employing bioinformatic tools, we selected a set of miRNAs and evaluated their expression in plasma sEVs and BC biopsies. Twelve miRNAs were identified in sEVs, of which, miR-21-5p, 221-3p, 146a-5p and 26a-5p were significantly associated with the Miller–Payne (MP) pathological response to NAT. Moreover, miR-21-5p, 146a-5p, 26a-5p and miR-24-3p were independent as predictors of MP response to NAT. However, the expression of these miRNAs showed no correlation between sEVs and tissue samples, indicating that the mechanisms of miRNA sorting into sEVs still needs to be elucidated. Functional analysis of miRNA target genes and drug interactions revealed that candidate miRNAs and their targets, can be regulated by different NAT regimens. This evidence supports their role in governing the patients’ therapy response and highlights their potential use as prediction biomarkers.

1. Introduction

Early detection through mammographic screening and improved treatment options have increased BC survival in westernized countries [1]. However, around 30% of cancer patients fail to respond to conventional treatments, leading to tumor recurrence. The main prognostic factors associated with BC are the number of involved lymph nodes, tumor size, histological grade, and hormone receptor status, while the treatment decision is based on both clinical and molecular characteristics of the tumor [2]. NAT is generally the first option before surgical treatment of locally advanced tumors, as it can downstage the extent of the disease, increase the rate of breast-conserving therapy [3,4], and offer information regarding the tumor response to systemic therapy. This information can guide treatment decision in adjuvant settings and provide prognostic data regarding patients survival outcomes [5]. Tumor response to systemic therapy is evaluated by clinical and radiological examination followed by a histological assessment of the excised tumor tissue [1]. However, clinical evaluation is often under or overestimated, thus pathological examination of the tumor bed following NAT is the gold standard, being associated with overall and disease-free survival [6]. Consequently, several pathological evaluation systems have been proposed in order to better stratify patients’ prognoses, including MP system [7] and residual cancer burden (RCB) index [8]. The MP method comprises a five-grade pathological evaluation system, with grade 5 representing pathological complete response (pCR), with no malignant cells identifiable in sections from the site of the tumor and grades 1–4 representing partial responses, with variable reductions 0–90% in tumor cellularity [7]. Besides cellularity, the RCB index takes into account additional variables such as the bidimensional diameter of the tumor, lymph node status, and metastasis. The pCR using RCB index is defined as RCB-0 [8]. Both methods have shown associations with overall and disease-free survival, highlighting them as reliable methods for evaluating the pathological tumor response to neoadjuvant therapy [9]. However, the histological assessment of NAT response can be achieved only at the time of surgery, after the patient has been exposed to several rounds of treatment. Therefore, the identification of predictive biomarkers of pathological response that could be evaluated before therapy onset is vital for stratifying patients that are more likely to benefit from NAT and may prevent over or undertreatment.
microRNAs (miRNAs) have been described to play a significant role in cancer [10]. Numerous reports of altered miRNA expression have been linked to various pathologies [11,12,13], including BC [14,15]. MiRNAs are small non-coding RNAs that regulate gene expression in key biological processes such as proliferation, apoptosis, invasion, and metastasis [16,17,18] and have recently emerged as regulators of the complex mechanisms that drive BC therapeutic resistance or sensitivity. Extracellular vesicles (EVs) are small lipid bilayer-encapsulated vesicles that are released by living cells in the microenvironment. Based on their size, EVs can be classified into two main subtypes: ectosomes (100–1000 nm) and small extracellular vesicles (sEVs, 30–150 nm, also referred to as exosomes by the literature), that encapsulate various components from the cell of origin, such as small RNA species and proteins [19]. Recent data emphasized that sEVs which carry miRNAs possess higher stability in various body fluids and can be selectively delivered to tissues based on surface markers [20]. Thus, tumor cells shed miRNAs into the circulation which are delivered to and up-taken by recipient cells in adjacent or distant tissues [21]. This horizontal transfer of miRNA functions as endogenous signaling mechanisms to reshape the microenvironment as tumor favoring–drug resistant niches [22]. Previous studies suggest that circulating sEVs miRNAs represent specific and stable non-invasive molecular biomarkers in disease progression and treatment efficacy [23,24,25]. Few studies have investigated the predictive power of miRNAs in pathological response following NAT in BC. Most of them were only focused on triple negative BC (TNBC) [26,27,28], and were evaluated in tissue samples [29], or enrolled a smaller patient cohort [9,30]. Considering that sEVs miRNAs detected in liquid biopsies represent more accessible and less invasive biomarker sources, our study aims to explore the predictive value of these miRNAs for the pathological response to NAT in BC.

2. Results

2.1. Patients and Tumor Characteristics

The clinical-pathological data of the patients included in the study are summarized in Table 1. The patient cohort consisted of 43 patients with matched biopsies and plasma samples, for 29 patients only plasma samples were available, while for 10 only biopsy samples. The median age of the patients included in the study was roughly 61, most of the patients being over 50 years old at the time of diagnosis. Around 50% of the patients had moderately differentiated carcinomas, with higher than 20 proliferation index. Most of the patients had luminal tumors, over 85% received an advanced-stage diagnosis (>II). Of the patients that received neoadjuvant therapy (NAT), around 75% received only chemotherapy, 16–17% received only hormonal therapy, while the rest received combinatory regimens of chemotherapy and hormonal therapy. One patient with disease progression also received radiotherapy in combination with chemotherapy and hormonal therapy. TNM classification system of malignant tumors was retained for prognostic information (primary and post-NAT surgery), while MP and RCB systems were used to evaluate the pathological response of the patients to NAT. According to the MP evaluation, around 30% of the patients had no response, 11% presented minor response, less than 30% presented intermediate response, while over 25%, had almost complete pathological response. RCB system classification data revealed that only 15% of the patients reached pathological complete response, almost a quarter were therapy resistant, while half had partial responses.

2.2. Selection of miRNAs of Interest

GEO2R analysis of the GSE25066 dataset revealed a list of 250 genes (−1.5 > FR > 1.5, BH adjusted p-value < 0.05) differentially expressed between the resistant and responsive BC patients. MiRwalk analysis predicted this list to be targeted by 957 individual miRNAs. NCI-60 revealed 60 miRNAs associated with paclitaxel and 20 associated with doxorubicin responses (p-value < 0.05, Pearson > 0.85), while 60 miRNAs were previously validated as paclitaxel and doxorubicin targets according to Pharmaco-miR database (Table S1). Of these, we randomly selected and further investigated in plasma sEVs, 33 miRNAs as predictive biomarkers for therapy response. (Table 2).

2.3. Investigation of microRNA Expression in Plasma sEVs

Of the 33 investigated miRNAs, only 12 miRNAs presented clear exponential amplification curves, 10 miRNAs had low or unspecific amplification and 11 miRNAs were negative for amplification. The qPCR amplification efficiency for miRNA advanced assays was 1.935 (96.75%). The statistical data association between the 12 quantifiable sEVs miRNAs expression and the clinic-pathological data of the patients included in the study are presented in Table 3. No correlations were observed between the investigated miRNA expression and patients age, while only slight associations were observed between miRNAs expression and the rest of the clinical-pathological data. Of note, significant different expression was calculated for miR-143-3p, 146a-5p, 24-3p, 193b-3p, 92a-3p, 484, 185-5p and 26a-5p and clinical lymph node status. Most notably, miR-21-5p, 221-3p, 146a-5p and 26a-5p were significantly associated with the MP pathological response to NAT; in general, their expression is decreased in patients that have improved response compared to the ones that are resistant (Figure 1). No significant associations were observed for the RCB evaluation system.

2.4. Investigation of microRNA Expression in Tissue Biopsies

Four miRNAs were associated with MP pathological response and further investigated for their expression in BC tissues (Table 4). Of note, only miR221-3p and miR-146a-5p were significantly associated with pathological response to NAT, higher expression was observed in tissues with pathological complete response (G5) compared to partial (G3) or non-responsive samples (G1) (Figure 2). None of the miRNAs expression correlated between the biopsy and sEVs samples. (Table 4). Analysis of the miRNAs expression in the TCGA database was performed as an independent external cohort (Table S2).

2.5. Prediction Analysis of miRNA Expression in Relation with Pathological Response

Ordinal logistic regression was used to test the prediction value of the four miRNAs of interest in pathological response to NAT, in both tissue and plasma samples. We found the expression of circulating sEVS miR-21-5p (OR = 0.603, 95% CI = 0.386–0.986, p = 0.044), miR-146a-5p (OR = 0.144, 95% CI = 0.026–0.799, p = 0.027), miR-24-3p (OR = 0.019, 95% CI = 0–0.953, p = 0.047) and miR-26a-5p (OR = 0.407, 95% CI = 0.189–0.874, p = 0.021) expression to be independent predictors of MP response. In general, for every unit increase in miRNA expression, there is a decreased probability in the odds of patients having an improved pathological response. In multivariable analysis with the full set of the four miRNAs as independent variables, the fitted model showed significant improvement over the null model (p = 0.011); however, none of the independent variables were found to have a significant effect on the model. No significant predictors were calculated for the RCB response. Tissue evaluation showed no significant predictive value for the four miRNAs set.

2.6. Functional Analysis of miRNA Target Genes and Drug Interactions

In order to provide a biological context to our results (for our four miRNAs of interest), miRTarBase was interrogated for validated mRNA targets. In parallel, mRNA and miR-21, miR-221, miR-146a and miR-26a expression data from TCGA were correlated in the matching samples. The two lists were intersected to generate lists of validated miRNAs—mRNA targets, considering of interest only the anticorrelated genes (r < −0.2, p < 0.05). Fifty-seven genes were identified to be anticorrelated with miR-21, sicty-nine genes with miR-221, eighteen genes with miR-146a and six genes with miR-26a (Table S3). These lists were imported in miRWalk3 for subsequent characterization using the GSEA built-in option (Table 5). Gene ontology analysis depicted the association of the miRNAs of interest with the biological processes, molecular function and cellular components associated with the significant target genes. Thus, through their anticorrelated target genes, miR-21-5p was mainly associated with kinase and ubiquitin transfer activities. miR-221-3p targets were associated with protein processing such as ubiquitination, destabilization and peroxisome targeting and regulation of the proliferation and apoptotic processes possibly through transcription regulation. A reduced number of anticorrelated genes were identified for the miR-146a and miR-26a, the only significant processes identified were that miR-146a targets are part of the receptor complex compartments, while miR-26a targets belong to the glutamatergic synapse compartment indicating a kinase activity. While no significant pathways were identified using the KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis option, Reactome database revealed that miR-221-3p target genes are associated with protein localization and gene transcription pathways.
In order to comprehensively depict the role of the 4 miRNAs in drug response regulation, we interrogated the pharmaco-miR database for known drug-miRNA-mRNA interactions (Table 6). Through their mRNA targets, miR-21-5p, miR-221-3p and miR-146a-5p have been found to be known mediators of BC chemotherapeutics such as 5-fluorouracil, taxanes, cisplatin or doxorubicin, or targeted therapeutics such as tamoxifen and trastuzumab.

3. Discussion

Treatment response represents one of the most significant issues of BC management, since about 30% of BC patients develop resistance to the standard therapeutic scheme, leading to cancer progression and decreased survival [31]. Locally advanced BC patients are subjected to neo-adjuvant therapy in order to reduce the tumor size and improve the surgical decision (the ability to choose breast-conserving surgery) as well as a method for assessing the patient’s response to systemic therapy [32]. Recent data suggest that pathological complete response (pCR) after NAT is associated with better patient prognosis [6]. However, the pathological response to NAT can only be assessed at surgery, after several cycles of systemic therapy, exposing the non-responding patients to unnecessary toxicity. Significant efforts have been made for developing gene-expression based predictors which could better stratify patients that could benefit from chemotherapy or hormonal treatment (Oncotype Dx, MammaPrint) and anticipate their prognosis (MapQuant D, Endopredict, the BC Index (BCI), and PAM50-ROR) [33,34]. Even if these tests have proven their usefulness, they are difficult to implement in the standard clinical routine due to their high cost and infrastructure requirements. Moreover, these tests demand fresh tissue samples, which can be difficult to harvest and involve invasive procedures for the patient. Therefore, the identification of novel, less invasive and more easily-accessible biomarkers for predicting BC response to the available therapeutic scheme is essential for globally improving BC management.
Liquid biopsy emerged as a complementary method for investigating molecular biomarkers in various body fluids, that could be used to assess the dynamic phenotype of BC patients. Together with the increasing interest for liquid biopsy, tumor derived sEVs miRNAs arose as a promising new class of biomarkers for cancer progression and treatment efficacy [23,24,25,35]. Circulating exosomes are crucial mediators of intercellular communication, their cargos mirroring the originating cells physiological state. By transferring cargos between tumor cells and adjacent or distant sites, exosomes reshape the microenvironment to establish tumor favoring–drug resistant niches [36]. Therefore, this study aimed to determine the predictive value of circulating sEVs miRNAs regarding pathological response to NAT and their expression correlation with the clinico-pathological data of BC patients. While several evaluation methods have been established [37], herein, we used RCB index and the MP system to evaluate pathological response to NAT. Both systems have been shown to accurately stratify survival outcome of patients after NAT [9].
By employing multiple publicly available databases, we explored the associations between miRNA expression and therapy response, drug interactions and their predicted mRNA targets, and identified 33 miRNAs of interest of which 12 could be accurately quantified in BC circulating sEVs (Table 3).
Two sEVs miRNAs, miR-328-3p and miR-34a-5p that have been previously reported as predictive biomarkers of NAT response in BC [30], had low or no expression in our samples. Moreover, plasma circulating miRNAs that have been described to be associated with pathological response in Her2+ BC patients (rev in [38]) such as miR-140-3p, miR-195-5p, miR- 373-3p, and let-7a-3p were negative in our samples. Furthermore, plasma miR-375-3p association with NAT response is well documented [39,40], suggesting that the presence of these miRNAs in plasma samples is not as an EVs cargo. However, the inconsistencies between studies, could be related to the use of different isolation, quantification and normalization methods of circulating miRNA in plasma in general and exosomes and EVs in particular. As no optimal normalization strategy for exosomal and sEVs miRNA expression is consensually accepted so far, variations in data interpretation and biological predicted effects are possible, leading to limitations in comparing research studies, and resulting in misleading conclusions [41]. Thus, validation of miRNA measurement across laboratories still remains a stringent future aspiration.
Four circulating sEVs miRNAs, notably miR-21-5p, 221-3p, 146a-5p and 26a-5p were found to be significantly associated with the MP pathological response to NAT. When their expression was evaluated in tissue samples, only miR-221-5p and miR-146a-5p retained statistical significance. However, no correlation was observed, when we compared the miRNA expression levels between plasma sEVs and tissue samples.
Previous data pointed out that the miRNAs loading into exosomes is not a passive process, and the repertoire of miRNAs content into exosomes may differ from that of the producer cell [42], assorting of oncomiRs into exosomes is generally suppressed, while the sorting of tumor suppressor miRs (TS-miRs) into exosomes is increased [43].
Three of these miRNAs, mir-21-5p, miR-146a-5p and miR-26a -5p were found to be individual independent predictors of MP pathological response in ordinal logistic regression, higher sEVs expression predicting decreased odd ratios of reaching pathological complete response. Consistently with these results, sEVs expression of these miRNAs was significantly lower in patients with MP partial response (G3) compared to non-responding ones (G1). Moreover, lower sEVs miR-21-5p expression was associated with MP pathological complete response (G5).
MiR-21-5p is a well-established oncomiR, that has been associated with chemoresistance in various cancers, such as lymphoma, colorectal, ovarian, lung and BC (rev in [44]). The literature data regarding the role of miR-21-5p in BC are conflicting. Studies reported both increased and decreased expression associations with NAT response. Higher miR-21-5p expression in plasma exosomes was directly correlated with tumor size in patients undergoing chemotherapy [45] and increased expression in patient’s serum after NAT was predictive of overall survival (OS) [46]. On the other hand, Liu et al. [47] reported decreased miR-21-5p serum expression associated with clinical response to NAT, patients with increased levels of serum miR-21-5p during NAT had a worse disease-free survival than those with decreased levels (rev in [48]), while miR-21-5p overexpression on tumor tissues induced resistance to neoadjuvant therapy combined with Trastuzumab and chemotherapy [49]. We have found that lower circulating sEVs miR-21-5p levels are associated with better pathological response to NAT and it is an independent predictor of MP response, consistent with previous data reported by McGuire et al. [50]. Additionally, based on miRNA-mRNA correlation and target prediction data, we estimate that miR-21-5p has a role in protein kinase and transferase activity. MiR-21-5p has been previously reported to regulate chemosensitivity to classic BC treatment regimens by targeting tumor-suppressors such as PTEN and programmed Cell Death 4 (PDCD4) [51,52,53] in various cancers.
Conflicting data regarding the oncogenic or tumor suppressive role of miR-221-3p in different cancers indicate its dual role in tumorigenesis [54,55]. In BC, high expression of miR-221-3p has been related to lymph node metastasis, distant metastasis, and poor prognosis [56]. Consistently, our results show increased circulating sEVs miR-221-3p in positive lymph node patients and worse pathological response to NAT. However, these data are conflicting with those from tumor tissue, where higher miR-221-3p was associated with complete response (G5) compared to partial (G3) and non-responders (G1). Even so, our results are confirmed by the study of Hanna et al. [57], that revealed that patients with higher level of tissue miR-221-3p had better survival prognosis. GSEA analysis suggests that through its molecular targets, miR-221-3p acts as a transcription regulator in processes such as proliferation and apoptosis. MiR-221 has been reported to downregulate some tumor suppressor pathways such as PTEN/Akt/mTOR signaling and thus promote chemoresistance of BC cells [58]. Moreover, miR-221/222 cluster has been shown to regulate ER-α [59], and its suppression has been shown to sensitize BC cells to tamoxifen [60]. A phase I clinical trial with antisense oligonucleotide targeting miR-221-3p (NCT04811898) is currently undergoing to assess the safety and tolerability in advanced solid tumor patients.
The predictive role of miR146a-5p has been studied in various cancers. Higher levels of miR-146a-5p in serum and plasma have been associated with worse response to platinum-based treatment in lung cancer [61,62], Imatinib therapy in patients with chronic myeloid leukemia (CML) [63], hormone [64] and radio [65] therapy in prostate cancer. In contrast, in acute myeloid leukemia (AML), high miR-146a-5p expression and/or upregulation of this miRNA during granulocyte-colony stimulating factor (G-CSF) priming chemotherapy was predictive of better clinical outcomes [66]. Similarly, advanced non-small cell lung cancer (NSCLC) patients with low serum exosomal miR-146a-5p levels had higher recurrence rates than those with high levels, suggesting that miR-146a-5p increased the chemosensitivity of NSCLC to cisplatin [67]. The role of miR-146a-5p in BC remains largely unexplored. We found sEVs miR-146a-5p to be an independent predictive factor in MP pathological response higher sEVs miR-146a-5p expression in plasma patients being associated with worse response to NAT. Of interest, in tissue samples, higher miR-146a-5p expression were observed in nonresponsive patients (G1, G2) when compared to partial responders (G3), but also in patients with complete response (G5) compared to those with partial response (G3). Mechanistically, exosomal miR-146a-5p has been shown to confer prostate cancer cells metastatic abilities through EGFR/ERK pathway [65], downregulate BRCA1 in triple negative sporadic BCs [68] and play a central role within the STAT/IFN signaling axis to create an immunosuppressive microenvironment [69,70].
Consistent with our results, miR-26a-5p, has been previously shown to be involved in pathways related to drug sensitivity/resistance [71], being associated with poor prognosis and predictive of the therapeutic response [72,73]. MiR-26a-5p expression is regulated by a negative estrogen signaling feedback loop [72,73] and has been shown to be a trastuzumab-inducible microRNAs [74] that seems to play an important role in resistance to trastuzumab therapy [75].

4. Materials and Methods

4.1. BC Patients and Samples Collection

Eighty-two female patients diagnosed with invasive BC at The Oncology Institute “Ion Chiricuță”, Cluj-Napoca, Romania (IOCN) gave their written consent for participation in this study in accordance with the Declaration of Helsinki. The study was approved by the IOCN ethical committee (Approval No. 59/29 November 2016). Core biopsies (n = 53) and peripheral blood (n = 72) were collected at the time of diagnostic procedure.

4.2. Plasma Processing and RNA Extraction from sEVs

Peripheral blood from 72 BC (BC) patients was collected in EDTA collection tubes and processed for plasma separation by double centrifugation at 4000 and 12,000 rpm for 10 min at 4 °C. The samples were aliquoted and stored at −80 °C until sEVs RNA extraction. Small EVs isolation was conducted on 400 μL prefiltered plasma following the Total Exosome Isolation Kit from plasma (Thermo Fisher, Waltham, MA, USA) protocol. Our preliminary data (in Supplementary File S1, Figures S1–S4) indicated that including a supplementary step of filtration (0.8 μm filter) considerably improves the sample purity, by maintaining the small EVs and by eliminating the large EVs, as also demonstrated by Enderle et al. [76], for a column-based method. Briefly, 400 μL of 0.8-μm-filtered plasma was treated with 0.05 volume of Proteinase K and incubated at 37 °C for 10 min followed by a 30 min incubation at 4 °C with 120 ul of precipitation reagent. Small EVs were separated by centrifugation at 10,000× g, resuspended in 200 ul PBS and processed for RNA isolation with Total Exosome RNA and Protein Isolation Kit (Thermo Fisher, Waltham, MA, USA) according to the manufacturer’s instructions 2.5 μL of exogenous cel-miR-39 (2 × 108 transcripts) were used as spike-in for each sample for normalization of miRNA expression.

4.3. Biopsies Processing and RNA Extraction

Fifty-three frozen biopsies were homogenized in TriReagent Solution (Ambion, Thermo Fisher Scientific, Waltham, MA, USA) using a Miccra D-1 (Miccra GmbH, Mullheim, Germany) polytron and processed for total RNA extraction using the classic phenol-chloroform method. The RNAs were quantified using NanoDrop ND-1000 (Thermo Scientific, Wilmington, DE, USA) and 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA).

4.4. miRNA Selection

Three approaches were employed to explore for putative miRNA biomarkers of NAT pathological response. First, Gene Expression Omnibus (GEO: https://www.ncbi.nlm.nih.gov/geo/ (accessed on 17 April 2019)) was interrogated for gene expression data of BC patients that underwent neoadjuvant therapy and had documented data regarding the pathological response to treatment. Complete gene expression data for primary breast tissue from a cohort of 508 patients were identified under accession number GSE25066. GEO2R analysis software was used to compare for gene expression data between resistant- RCB III (n = 110) and responsive- RCB 0/I (n = 118) patients. Fold regulation (FR) of ±1.5 and Benjamini–Hochberg (BH) adjusted p-value < 0.05 was considered as significance threshold. Retrieved differentially expressed gene list was imported into the miRwalk webtool and the miRTarBase filter was used to identify validated miRNAs known to target the 3′-UTR, CDS or the 5′-UTR of identified genes. Second, Cellminer Cross DataBase (https://discover.nci.nih.gov/rsconnect/cellminercdb/ (accessed on 17 April 2019)) was used to retrieve BC cell line pharmacogenomic miRNA data in response to classic BC treatments. The Compare Patterns tool was used to compute Pearson correlations between the NCI-60 miRNA BC datasets and paclitaxel and doxorubicin treatments. Last, Pharmaco-mir database was explored for known miRNA-drug interactions. Of interest were miRNAs targets of paclitaxel and doxorubicin and their clinical derivatives.

4.5. miRNA Expression Evaluation

Considering that plasma samples contain low quantities of sEVs-miRNA, we used the one-step advanced miRNA system to simultaneously assess the expression levels of multiple miRNAs from the same sample. An amount of 4 μL of plasma sEVs RNAs or 10 ng of total RNA from biopsies were pre-amplified using universal RT miRNA primers to generate cDNAs following the TaqMan Advanced miRNA cDNA Synthesis Kit protocol. Next, 1:10 v/v diluted cDNAs and specific miRNA advanced assays were amplified with TaqMan Fast Advanced Master Mix (2X) using the LC480 device (Roche, Basel, Switzerland) with the following PCR settings: 55 °C for 2 min to remove RNA contaminants; 95 °C for 20 sec for Taq polymerase amplification; 40 cycles of 95 °C for 3 sec followed by 60 °C for 30 sec for PCR amplification. The efficiency of qPCR amplification was evaluated using the standard curve generated for cel-miR-39 exogenous normalizer, started with 2 × 108 transcripts. ∆∆Ct method was used for miRNA relative quantification by reporting the Ct values of the miRNAs of interest to miR-16-5p Ct values. For the sEVs samples, all Ct values were beforehand normalized to cel-miR-39 expression.

4.6. TCGA Data Analysis and Bioinformatics

The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) expression data (miRNA and mRNA) and their clinical information were obtained from National Cancer Institute Genomic Data Commons (NCI GDC) data portal (https://portal.gdc.cancer.gov/ (accessed on 17 April 2019)) and cBioPortal for Cancer Genomics (https://www.cbioportal.org/ (accessed on 17 April 2019)). The miRNA-seq data expressed as reads per million and fragments per kilobase millions mRNA-seq data were filtered and log2 (x + 1) transformed. After processing, a miRNA tissue dataset containing 916 tumoral samples and a mRNA tissue dataset of 983 tumoral samples were retained for subsequent analysis. Spearman correlation was used to test potential miRNAs—mRNA associations and intersected with validated miRNA-target interactions retrieved from miRTarBase.

4.7. Gene Set Enrichment and Drug Prediction Analysis

The GSEA built-in option of the miRWalk3 webtool was used to identify possible pathways, biological processes, cellular compartments and molecular functions of the miRNAs of interest. Each list of mRNAs that were anticorrelated with the miRNAs of interest in the TCGA cohort was analyzed for enrichment in the Reactome and KEGG Pathways or Gene Ontology (GO). MultiMiR package in R was used to interrogate Pharmaco-miR database for interactions between possible drugs and the miRNAs of interest and their mRNA targets.

4.8. Statistical Analysis

The association between clinico-pathological characteristics and miRNAs expression was evaluated with the Mann–Whitney U test for two categorical variables or the Kruskal–Wallis test, followed by Dunn’s multiple comparison post hoc test in the case of three or more categorical variables based on the data distribution. Fold regulation (FR) was calculated as the ratio between mean value of the interest group and the reference group. Spearman correlation was used to test for correlations between the miRNA expression in sEVs and tissue. Univariate and multivariable ordinal regression analysis was used to test for pathological response miRNA predictors using SPSS (IBM SPSS Statistics for Macintosh, Version 28.0; IBM Corp Armonk, NY, USA). All analyses were considered significant at p-value less than 0.05.

5. Conclusions

In summary, we investigated the predictive power of microRNAs from plasma sEVs of BC patients, and identified four miRNAs (miR-21-5p, 221-3p, 146a-5p and 26a-5p) that were associated with MP pathological response after neo-adjuvant therapy, of which, three acted as independent predictors (miR-21-5p, 146a-5p and 26a-5p). Bioinformatic analysis revealed that candidate miRNAs, and subsequently their target genes, can be regulated by different NAT regimens. This evidence supports their role in governing the patients’ therapy response and highlights their potential use as prediction biomarkers. The expression levels of these miRNAs in tissue and plasma sEVs s were not correlated, indicating that further functional studies to understand the specific sorting of sEVs miRNAs are needed to fully elucidate the underlying mechanisms involved in shaping the NAT response.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232012625/s1.

Author Contributions

Conceptualization, O.B. (Ovidiu Balacescu), O.T., O.B. (Oana Baldasici) and L.B.; methodology, O.B. (Ovidiu Balacescu), O.B. (Oana Baldasici), D.C., V.P.; validation, O.B. (Oana Baldasici), O.T., S.V., V.P. and D.C.; formal analysis, L.B.; investigation, O.B. (Oana Baldasici), D.C., V.P., S.V., C.L., A.R., A.C., B.F., L.B.-T. and A.J.; resources, O.B. (Ovidiu Balacescu), O.T.; data curation, L.B.; writing—original draft preparation, O.B. (Oana Baldasici), O.T. and L.B.; writing—review and editing, O.T., L.B., O.B. (Ovidiu Balacescu), and O.B. (Oana Baldasici); visualization, L.B.; supervision, O.T. and L.V.; project administration, O.B. (Ovidiu Balacescu); funding acquisition, O.B. (Ovidiu Balacescu) All authors have read and agreed to the published version of the manuscript.

Funding

This research and the APC was funded by Experimental-Demonstrative Project PN-III-P2-2.1-PED-2019-5128, grant number 556PED/2020, project title: Exploring the molecular mechanisms involved in the regulation of the migratory phenotype of primary breast cancer cells, by exosomes through microfluidic and genomics approaches (EXOMICROVEL). Patients’ samples were provided by Competitiveness Operational Programme 2014–2020, Contract no. 41/02.09.2016, MySMIS 103557, Project title: Genomic and microfluidic approaches towards blocking breast cancer cell invasion and metastasis—BREASTMINCROGENOMICS.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee of The Oncology Institute “Ion Chiricuță”, Cluj-Napoca, Romania (Approval No. 59/29 September 2016).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef]
  2. Godone, R.; Leitão, G.; Araújo, N.; Castelletti, C.; Lima-Filho, J.; Martins, D. Clinical and molecular aspects of breast cancer: Targets and therapies. Biomed. Pharmacother. 2018, 106, 14–34. [Google Scholar] [CrossRef]
  3. McDonald, E.S.; Clark, A.S.; Tchou, J.; Zhang, P.; Freedman, G.M. Clinical Diagnosis and Management of Breast Cancer. J. Nucl. Med. 2016, 57, 9S–16S. [Google Scholar] [CrossRef] [Green Version]
  4. Thompson, A.M.; Moulder-Thompson, S.L. Neoadjuvant treatment of breast cancer. Ann. Oncol. 2012, 23, x231–x236. [Google Scholar] [CrossRef]
  5. Sikov, W.; Boughey, J.; Al-Hilli, Z. UpToDate: General Principles of Neoadjuvant Management of Breast Cancer. UpToDate 2021, 1–21. [Google Scholar]
  6. Spring, L.M.; Fell, G.; Arfe, A.; Sharma, C.; Greenup, R.A.; Reynolds, K.L.; Smith, B.L.; Alexander, B.M.; Moy, B.; Isakoff, S.J.; et al. Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis. Clin. Cancer Res. 2020, 26, 2838–2848. [Google Scholar] [CrossRef]
  7. Ogston, K.N.; Miller, I.D.; Payne, S.; Hutcheon, A.W.; Sarkar, T.K.; Smith, I.; Schofield, A.; Heys, S.D. A new histological grading system to assess response of breast cancers to primary chemotherapy: Prognostic significance and survival. Breast 2003, 12, 320–327. [Google Scholar] [CrossRef]
  8. Symmans, W.F.; Peintinger, F.; Hatzis, C.; Rajan, R.; Kuerer, H.; Valero, V.; Assad, L.; Poniecka, A.; Hennessy, B.; Green, M.; et al. Measurement of Residual Breast Cancer Burden to Predict Survival After Neoadjuvant Chemotherapy. J. Clin. Oncol. 2007, 25, 4414–4422. [Google Scholar] [CrossRef]
  9. Wang, W.; Liu, Y.; Zhang, H.; Zhang, S.; Duan, X.; Ye, J.; Xu, L.; Zhao, J.; Cheng, Y.; Liu, Q. Prognostic value of residual cancer burden and Miller-Payne system after neoadjuvant chemotherapy for breast cancer. Gland Surg. 2021, 10, 3211–3221. [Google Scholar] [CrossRef]
  10. Carvalho, T.M.; Brasil, G.O.; Jucoski, T.S.; Adamoski, D.; de Lima, R.S.; Spautz, C.C.; Anselmi, K.F.; Ozawa, P.M.M.; Cavalli, I.J.; de Oliveira, J.C.; et al. MicroRNAs miR-142-5p, miR-150-5p, miR-320a-3p, and miR-4433b-5p in Serum and Tissue: Potential Biomarkers in Sporadic Breast Cancer. Front. Genet. 2022, 13. [Google Scholar] [CrossRef]
  11. Ryan, B.M.; Robles, A.I.; Harris, C.C. Genetic variation in microRNA networks: The implications for cancer research. Nat. Rev. Cancer 2010, 10, 389–402. [Google Scholar] [CrossRef] [Green Version]
  12. Mehrgou, A.; Ebadollahi, S.; Seidi, K.; Ayoubi-Joshaghani, M.H.; Yazdi, A.A.; Zare, P.; Jaymand, M.; Jahanban-Esfahlan, R. Roles of miRNAs in Colorectal Cancer: Therapeutic Implications and Clinical Opportunities. Adv. Pharm. Bull. 2021, 11, 233–247. [Google Scholar] [CrossRef]
  13. Gaál, Z. Implication of microRNAs in Carcinogenesis with Emphasis on Hematological Malignancies and Clinical Translation. Int. J. Mol. Sci. 2022, 23, 5838. [Google Scholar] [CrossRef]
  14. Loh, H.-Y.; Norman, B.P.; Lai, K.-S.; Rahman, N.M.A.N.A.; Alitheen, N.B.M.; Osman, M.A. The Regulatory Role of MicroRNAs in Breast Cancer. Int. J. Mol. Sci. 2019, 20, 4940. [Google Scholar] [CrossRef] [Green Version]
  15. Kim, M.W.; Park, S.; Lee, H.; Gwak, H.; Hyun, K.; Kim, J.Y.; Jung, H.; Kim, S.I. Multi-miRNA panel of tumor-derived extracellular vesicles as promising diagnostic biomarkers of early-stage breast cancer. Cancer Sci. 2021, 112, 5078–5087. [Google Scholar] [CrossRef]
  16. Ichimura, A.; Ruike, Y.; Terasawa, K.; Tsujimoto, G. miRNAs and regulation of cell signaling. FEBS J. 2011, 278, 1610–1618. [Google Scholar] [CrossRef]
  17. Inui, M.; Martello, G.; Piccolo, S. MicroRNA control of signal transduction. Nat. Rev. Mol. Cell Biol. 2010, 11, 252–263. [Google Scholar] [CrossRef]
  18. Stavast, C.J.; van Zuijen, I.; Erkeland, S.J. MicroRNA-139, an Emerging Gate-Keeper in Various Types of Cancer. Cells 2022, 11, 769. [Google Scholar] [CrossRef]
  19. Gao, Y.; Qin, Y.; Wan, C.; Sun, Y.; Meng, J.; Huang, J.; Hu, Y.; Jin, H.; Yang, K. Small Extracellular Vesicles: A Novel Avenue for Cancer Management. Front. Oncol. 2021, 11. [Google Scholar] [CrossRef]
  20. Wang, H.-X.; Gires, O. Tumor-derived extracellular vesicles in breast cancer: From bench to bedside. Cancer Lett. 2019, 460, 54–64. [Google Scholar] [CrossRef]
  21. Cardinali, B.; Tasso, R.; Piccioli, P.; Ciferri, M.C.; Quarto, R.; Del Mastro, L. Circulating miRNAs in Breast Cancer Diagnosis and Prognosis. Cancers 2022, 14, 2317. [Google Scholar] [CrossRef] [PubMed]
  22. Yi, Y.; Wu, M.; Zeng, H.; Hu, W.; Zhao, C.; Xiong, M.; Lv, W.; Deng, P.; Zhang, Q.; Wu, Y. Tumor-Derived Exosomal Non-Coding RNAs: The Emerging Mechanisms and Potential Clinical Applications in Breast Cancer. Front. Oncol. 2021, 11, 4173. [Google Scholar] [CrossRef]
  23. Manier, S.; Liu, C.-J.; Avet-Loiseau, H.; Park, J.; Shi, J.; Campigotto, F.; Salem, K.Z.; Huynh, D.; Glavey, S.V.; Rivotto, B.; et al. Prognostic role of circulating exosomal miRNAs in multiple myeloma. Blood 2017, 129, 2429–2436. [Google Scholar] [CrossRef] [Green Version]
  24. Schwarzenbach, H. The clinical relevance of circulating, exosomal miRNAs as biomarkers for cancer. Expert Rev. Mol. Diagn. 2015, 15, 1159–1169. [Google Scholar] [CrossRef] [PubMed]
  25. Nagamitsu, Y.; Nishi, H.; Sasaki, T.; Takaesu, Y.; Terauchi, F.; Isaka, K. Profiling analysis of circulating microRNA expression in cervical cancer. Mol. Clin. Oncol. 2016, 5, 189–194. [Google Scholar] [CrossRef] [Green Version]
  26. García-Vazquez, R.; Ruiz-García, E.; García, A.M.; la Vega, H.A.-D.; Lara-Medina, F.; Alvarado-Miranda, A.; Maldonado-Martínez, H.; González-Barrios, J.A.; Campos-Parra, A.D.; Cuevas, S.R.; et al. A microRNA signature associated with pathological complete response to novel neoadjuvant therapy regimen in triple-negative breast cancer. Tumor Biol. 2017, 39, 47–68. [Google Scholar] [CrossRef] [Green Version]
  27. Sueta, A.; Fujiki, Y.; Goto-Yamaguchi, L.; Tomiguchi, M.; Yamamoto-Ibusuki, M.; Iwase, H.; Yamamoto, Y. Exosomal miRNA profiles of triple-negative breast cancer in neoadjuvant treatment. Oncol. Lett. 2021, 22, 1–10. [Google Scholar] [CrossRef]
  28. Kahraman, M.; Röske, A.; Laufer, T.; Fehlmann, T.; Backes, C.; Kern, F.; Kohlhaas, J.; Schrörs, H.; Saiz, A.; Zabler, C.; et al. MicroRNA in diagnosis and therapy monitoring of early-stage triple-negative breast cancer. Sci. Rep. 2018, 8, 11584. [Google Scholar] [CrossRef] [Green Version]
  29. Xing, A.-Y.; Wang, B.; Li, Y.H.; Chen, X.; Wang, Y.-W.; Liu, H.-T.; Gao, P. Identification of miRNA Signature in Breast Cancer to Predict Neoadjuvant Chemotherapy Response. Pathol. Oncol. Res. 2021, 27, 1609753. [Google Scholar] [CrossRef]
  30. Todorova, V.K.; Byrum, S.D.; Gies, A.J.; Haynie, C.; Smith, H.; Reyna, N.S.; Makhoul, I. Circulating Exosomal microRNAs as Predictive Biomarkers of Neoadjuvant Chemotherapy Response in Breast Cancer. Curr. Oncol. 2022, 29, 613–630. [Google Scholar] [CrossRef]
  31. Tryfonidis, K.; Senkus, E.; Cardoso, M.J.; Cardoso, F. Management of locally advanced breast cancer—Perspectives and future directions. Nat. Rev. Clin. Oncol. 2015, 12, 147–162. [Google Scholar] [CrossRef] [PubMed]
  32. Mandilaras, V.; Bouganim, N.; Spayne, J.; Dent, R.; Arnaout, A.; Boileau, J.; Brackstone, M.; Meterissian, S.; Clemons, M. Concurrent Chemoradiotherapy for Locally Advanced Breast Cancer—Time for a New Paradigm? Curr. Oncol. 2015, 22, 25–32. [Google Scholar] [CrossRef] [PubMed]
  33. Prat, A.; Ellis, M.J.; Perou, C.M. Practical implications of gene-expression-based assays for breast oncologists. Nat. Rev. Clin. Oncol. 2011, 9, 48–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zeng, C.; Zhang, J. A Narrative Review of Five Multigenetic Assays in Breast Cancer. Transl. Cancer Res. 2022, 11, 897–907. [Google Scholar] [CrossRef] [PubMed]
  35. Dong, X.; Bai, X.; Ni, J.; Zhang, H.; Duan, W.; Graham, P.; Li, Y. Exosomes and breast cancer drug resistance. Cell Death Dis. 2020, 11, 987. [Google Scholar] [CrossRef]
  36. Dong, M.; Liu, Q.; Xu, Y.; Zhang, Q. Extracellular Vesicles: The Landscape in the Progression, Diagnosis, and Treatment of Triple-Negative Breast Cancer. Front. Cell Dev. Biol. 2022, 10, 1–14. [Google Scholar] [CrossRef]
  37. Sahoo, S.; Lester, S.C. Pathology of Breast Carcinomas after Neoadjuvant Chemotherapy: An Overview with Recommendations on Specimen Processing and Reporting. Arch. Pathol. Lab. Med. 2009, 133, 633–642. [Google Scholar] [CrossRef]
  38. Davey, M.G.; Davey, M.S.; Richard, V.; Wyns, W.; Soliman, O.; Miller, N.; Lowery, A.J.; Kerin, M.J. Overview of MicroRNA Expression in Predicting Response to Neoadjuvant Therapies in Human Epidermal Growth Receptor-2 Enriched Breast Cancer—A Systematic Review. Breast Cancer 2022, 16, 11782234221086684. [Google Scholar] [CrossRef]
  39. Shao, B.; Wang, X.; Zhang, L.; Li, D.; Liu, X.; Song, G.; Cao, H.; Zhu, J.; Li, H. Plasma MicroRNAs Predict Chemoresistance in Patients with Metastatic Breast Cancer. Technol. Cancer Res. Treat. 2019, 18, 1533033819828709. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, Z.; Zhang, H.; Li, C.; Xiang, Q.; Xu, L.; Liu, Q.; Pang, X.; Zhang, W.; Zhang, H.; Zhang, S.; et al. Circulating MicroRNAs as Indicators in the Prediction of Neoadjuvant Chemotherapy Response in Luminal B Breast Cancer. Thorac. Cancer 2021, 12, 3396–3406. [Google Scholar] [CrossRef]
  41. Schwarzenbach, H.; Da Silva, A.M.; Calin, G.; Pantel, K. Data Normalization Strategies for MicroRNA Quantification. Clin. Chem. 2015, 61, 1333–1342. [Google Scholar] [CrossRef] [PubMed]
  42. Squadrito, M.L.; Baer, C.; Burdet, F.; Maderna, C.; Gilfillan, G.D.; Lyle, R.; Ibberson, M.; De Palma, M. Endogenous RNAs Modulate MicroRNA Sorting to Exosomes and Transfer to Acceptor Cells. Cell Rep. 2014, 8, 1432–1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Teng, Y.; Ren, Y.; Hu, X.; Mu, J.; Samykutty, A.; Zhuang, X.; Deng, Z.; Kumar, A.; Zhang, L.; Merchant, M.L.; et al. MVP-Mediated Exosomal Sorting of MiR-193a Promotes Colon Cancer Progression. Nat. Commun. 2017, 8, 14448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Campos, A.; Sharma, S.; Obermair, A.; Salomon, C. Extracellular Vesicle-Associated MiRNAs and Chemoresistance: A Systematic Review. Cancers 2021, 13, 4608. [Google Scholar] [CrossRef]
  45. Rodríguez-Martínez, A.; De Miguel-Pérez, D.; Ortega, F.G.; García-Puche, J.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] [PubMed]
  46. 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] [PubMed]
  47. 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]
  48. Zhang, Z.; Zhang, H.; Yu, J.; Xu, L.; Pang, X.; Xiang, Q.; Liu, Q.; Cui, Y. MiRNAs as Therapeutic Predictors and Prognostic Biomarkers of Neoadjuvant Chemotherapy in Breast Cancer: A Systematic Review and Meta-Analysis. Breast Cancer Res. Treat. 2022, 194, 483–505. [Google Scholar] [CrossRef]
  49. 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, 37269–37280. [Google Scholar] [CrossRef] [Green Version]
  50. McGuire, A.; Casey, M.C.; Waldron, R.M.; Heneghan, H.; Kalinina, O.; Holian, E.; McDermott, A.; Lowery, A.J.; Newell, J.; Dwyer, R.M.; et al. Prospective Assessment of Systemic MicroRNAs as Markers of Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancers 2020, 12, 1820. [Google Scholar] [CrossRef]
  51. Sun, L.H.; Tian, D.; Yang, Z.C.; Li, J.L. Exosomal MiR-21 Promotes Proliferation, Invasion and Therapy Resistance of Colon Adenocarcinoma Cells through Its Target PDCD4. Sci. Rep. 2020, 10, 1–8. [Google Scholar] [CrossRef] [PubMed]
  52. Vandewalle, V.; Essaghir, A.; Bollaert, E.; Lenglez, S.; Graux, C.; Schoemans, H.; Saussoy, P.; Michaux, L.; Valk, P.J.M.; Demoulin, J.B.; et al. MiR-15a-5p and MiR-21-5p Contribute to Chemoresistance in Cytogenetically Normal Acute Myeloid Leukaemia by Targeting PDCD4, ARL2 and BTG2. J. Cell. Mol. Med. 2021, 25, 575–585. [Google Scholar] [CrossRef] [PubMed]
  53. Shi, G.H.; Ye, D.W.; Yao, X.D.; Zhang, S.L.; Dai, B.; Zhang, H.L.; Shen, Y.J.; Zhu, Y.; Zhu, Y.P.; Xiao, W.J.; et al. Involvement of MicroRNA-21 in Mediating Chemo-Resistance to Docetaxel in Androgen-Independent Prostate Cancer PC3 Cells. Acta Pharmacol. Sin. 2010, 31, 867–873. [Google Scholar] [CrossRef]
  54. Sharma, P.C.; Gupta, A. MicroRNAs: Potential Biomarkers for Diagnosis and Prognosis of Different Cancers. Transl. Cancer Res. 2020, 9, 5798–5818. [Google Scholar] [CrossRef] [PubMed]
  55. Di Martino, M.T.; Arbitrio, M.; Caracciolo, D.; Cordua, A.; Cuomo, O.; Grillone, K.; Riillo, C.; Caridà, G.; Scionti, F.; Labanca, C.; et al. MiR-221/222 as Biomarkers and Targets for Therapeutic Intervention on Cancer and Other Diseases: A Systematic Review. Mol. Ther. Nucleic Acids 2022, 27, 1191–1224. [Google Scholar] [CrossRef] [PubMed]
  56. Yan, L.X.; Huang, X.F.; Shao, Q.; Huang, M.Y.; Deng, L.; Wu, Q.L.; Zeng, Y.X.; Shao, J.Y. MicroRNA MiR-21 Overexpression in Human Breast Cancer Is Associated with Advanced Clinical Stage, Lymph Node Metastasis and Patient Poor Prognosis. RNA 2008, 14, 2348–2360. [Google Scholar] [CrossRef] [Green Version]
  57. Hanna, J.A.; Wimberly, H.; Kumar, S.; Slack, F.; Agarwal, S.; Rimm, D.L. Quantitative Analysis of MicroRNAs in Tissue Microarrays by in Situ Hybridization. Biotechniques 2012, 52, 235. [Google Scholar] [CrossRef]
  58. Yin, Y.; Wang, X.; Li, T.; Ren, Q.; Li, L.; Sun, X.; Zhang, B.; Wang, X.; Han, H.; He, Y.; et al. MicroRNA-221 Promotes Breast Cancer Resistance to Adriamycin via Modulation of PTEN/Akt/MTOR Signaling. Cancer Med. 2020, 9, 1544–1552. [Google Scholar] [CrossRef]
  59. Di Leva, G.; Gasparini, P.; Piovan, C.; Ngankeu, A.; Garofalo, M.; Taccioli, C.; Iorio, M.V.; Li, M.; Volinia, S.; Alder, H.; et al. MicroRNA Cluster 221-222 and Estrogen Receptor Alpha Interactions in Breast Cancer. J. Natl. Cancer Inst. 2010, 102, 706–721. [Google Scholar] [CrossRef]
  60. Gan, R.; Yang, Y.; Yang, X.; Zhao, L.; Lu, J.; Meng, Q.H. Downregulation of MiR-221/222 Enhances Sensitivity of Breast Cancer Cells to Tamoxifen through Upregulation of TIMP3. Cancer Gene Ther. 2014, 21, 290–296. [Google Scholar] [CrossRef] [Green Version]
  61. Yuwen, D.; Ma, Y.; Wang, D.; Gao, J.; Li, X.; Xue, W.; Fan, M.; Xu, Q.; Shen, Y.; Shu, Y. Prognostic Role of Circulating Exosomal MiR-425-3p for the Response of NSCLC to Platinum-Based Chemotherapy. Cancer Epidemiol. Biomarkers Prev. 2019, 28, 163–173. [Google Scholar] [CrossRef] [PubMed]
  62. Zhong, S.; Golpon, H.; Zardo, P.; Borlak, J. MiRNAs in Lung Cancer. A Systematic Review Identifies Predictive and Prognostic MiRNA Candidates for Precision Medicine in Lung Cancer. Transl. Res. 2021, 230, 164–196. [Google Scholar] [CrossRef] [PubMed]
  63. Habib, E.M.; Nosiar, N.A.; Eid, M.A.; Taha, A.M.; Sherief, D.E.; Hassan, A.E.; Abdel Ghafar, M.T. Circulating MiR-146a Expression Predicts Early Treatment Response to Imatinib in Adult Chronic Myeloid Leukemia. J. Investig. Med. 2021, 69, 333–337. [Google Scholar] [CrossRef] [PubMed]
  64. Kopcalic, K.; Petrovic, N.; Stanojkovic, T.P.; Stankovic, V.; Bukumiric, Z.; Roganovic, J.; Malisic, E.; Nikitovic, M. Association between MiR-21/146a/155 Level Changes and Acute Genitourinary Radiotoxicity in Prostate Cancer Patients: A Pilot Study. Pathol. Res. Pract. 2019, 215, 626–631. [Google Scholar] [CrossRef]
  65. Zhang, Y.; Zhao, J.; Ding, M.; Su, Y.; Cui, D.; Jiang, C.; Zhao, S.; Jia, G.; Wang, X.; Ruan, Y.; et al. Loss of Exosomal MiR-146a-5p from Cancer-Associated Fibroblasts after Androgen Deprivation Therapy Contributes to Prostate Cancer Metastasis. J. Exp. Clin. Cancer Res. 2020, 39, 1–19. [Google Scholar] [CrossRef]
  66. Li, X.; Xu, L.; Sheng, X.; Cai, J.; Liu, J.; Yin, T.; Xiao, F.; Chen, F.; Zhong, H. Upregulated MicroRNA-146a Expression Induced by Granulocyte Colony-Stimulating Factor Enhanced Low-Dosage Chemotherapy Response in Aged Acute Myeloid Leukemia Patients. Exp. Hematol. 2018, 68, 66–79. [Google Scholar] [CrossRef]
  67. Yuwen, D.L.; Sheng, B.B.; Liu, J.; Wenyu, W.; Shu, Y.Q. MiR-146a-5p Level in Serum Exosomes Predicts Therapeutic Effect of Cisplatin in Non-Small Cell Lung Cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2650–2658. [Google Scholar]
  68. Garcia, A.I.; Buisson, M.; Bertrand, P.; Rimokh, R.; Rouleau, E.; Lopez, B.S.; Lidereau, R.; Mikaélian, I.; Mazoyer, S. Down-Regulation of BRCA1 Expression by MiR-146a and MiR-146b-5p in Triple Negative Sporadic Breast Cancers. EMBO Mol. Med. 2011, 3, 279–290. [Google Scholar] [CrossRef]
  69. Mastroianni, J.; Stickel, N.; Andrlova, H.; Hanke, K.; Melchinger, W.; Duquesne, S.; Schmidt, D.; Falk, M.; Andrieux, G.; Pfeifer, D.; et al. MiR-146a Controls Immune Response in the Melanoma Microenvironment. Cancer Res. 2019, 79, 183–195. [Google Scholar] [CrossRef] [Green Version]
  70. Sun, X.; Zhang, J.; Hou, Z.; Han, Q.; Zhang, C.; Tian, Z. MiR-146a Is Directly Regulated by STAT3 in Human Hepatocellular Carcinoma Cells and Involved in Anti-Tumor Immune Suppression. Cell Cycle 2015, 14, 243–252. [Google Scholar] [CrossRef] [Green Version]
  71. Rizzo, S.; Cangemi, A.; Galvano, A.; Fanale, D.; Buscemi, S.; Ciaccio, M.; Russo, A.; Castorina, S.; Bazan, V. Analysis of MiRNA Expression Profile Induced by Short Term Starvation in Breast Cancer Cells Treated with Doxorubicin. Oncotarget 2017, 8, 71924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Tan, S.; Ding, K.; Li, R.; Zhang, W.; Li, G.; Kong, X.; Qian, P.; Lobie, P.E.; Zhu, T. Identification of MiR-26 as a Key Mediator of Estrogen Stimulated Cell Proliferation by Targeting CHD1, GREB1 and KPNA2. Breast Cancer Res. 2014, 16, R40. [Google Scholar] [CrossRef] [PubMed]
  73. Maillot, G.; Lacroix-Triki, M.; Pierredon, S.; Gratadou, L.; Schmidt, S.; Bénès, V.; Roché, H.; Dalenc, F.; Auboeuf, D.; Millevoi, S.; et al. Widespread Estrogen-Dependent Repression of Micrornas Involved in Breast Tumor Cell Growth. Cancer Res. 2009, 69, 8332–8340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. 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] [Green Version]
  75. 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, srep41309. [Google Scholar] [CrossRef] [Green Version]
  76. Enderle, D.; Spiel, A.; Coticchia, C.M.; Berghoff, E.; Mueller, R.; Schlumpberger, M.; Sprenger-Haussels, M.; Shaffer, J.M.; Lader, E.; Skog, J.; et al. Characterization of RNA from Exosomes and Other Extracellular Vesicles Isolated by a Novel Spin Column-Based Method. PLoS ONE 2015, 10, e0136133. [Google Scholar] [CrossRef]
Figure 1. The miR-21-5p, 221-3p, 146a-5p and 26a-5p expression in plasma-derived sEVs, according to MP staging. The differences between groups were evaluated with Kruskal–Wallis test, and pairwise comparisons were performed with Dunn’s test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Figure 1. The miR-21-5p, 221-3p, 146a-5p and 26a-5p expression in plasma-derived sEVs, according to MP staging. The differences between groups were evaluated with Kruskal–Wallis test, and pairwise comparisons were performed with Dunn’s test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Ijms 23 12625 g001
Figure 2. The miR-21-5p, 221-3p, 146a-5p and 26a-5p expression in tissue samples, according to MP staging. The differences between groups were evaluated with Kruskal–Wallis test, and pairwise comparisons were performed with Dunn’s test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Figure 2. The miR-21-5p, 221-3p, 146a-5p and 26a-5p expression in tissue samples, according to MP staging. The differences between groups were evaluated with Kruskal–Wallis test, and pairwise comparisons were performed with Dunn’s test (* p < 0.05, ** p < 0.01, *** p < 0.001).
Ijms 23 12625 g002
Table 1. Clinico-pathological data of the patients included in the study.
Table 1. Clinico-pathological data of the patients included in the study.
VariablePlasma StudyTissue Study
n = 72n = 53
Age
Median (range)61.5 (35–76)61 (29–76)
Grading biopsy
G19 (12.5%)6 (11.3%)
G239 (54.2%)26 (49.1%)
G324 (33.3%)21 (39.6%)
Estrogen Receptor
ER-20 (27.8%)14 (26.4%)
ER+52 (72.2%)39 (73.6%)
Progesteron receptor
PR-32 (44.4%)24 (45.3%)
PR+40 (55.6%)29 (54.7%)
HER2
HER2-64 (88.9%)47 (88.7%)
HER2+8 (11.1%)5 (9.4%)
NA-1 (1.9%)
KI67
2031 (43.1%)20 (37.7%)
>2041 (56.9%)33(62.3%)
Molecular Subtype
Luminal A23 (31.9%)15 (28.3%)
Luminal B26 (36.2%)22 (41.5%)
LuminalB_HER2+3 (4.2%)1 (1.9%)
HER2+5 (6.9%)4 (7.5%)
TNBC15 (20.8%)10 (18.9%)
NA-1 (1.9%)
Tumor size (c)
cT14 (5.6%)2 (3.8%)
cT236 (50%)25 (47.2%)
cT311 (15.3%)9 (17.0%)
cT416 (22.2%)13 (24.5%)
NA5 (6.9%)4 (7.5%)
Lymph nodes (c)
cN017 (23.6%)9 (17.0%)
cN122 (30.6%)16 (30.2%)
cN226 (36.1%)23 (43.4%)
cN32 (2.8%)1 (1.9%)
NA5 (6.9%)4 (7.5%)
Metastasis (c)
cM062 (86.1%)45 (84.9%)
cM12 (2.8%)1 (1.9%)
NA8 (11.1%)7 (13.2%)
Clinical stage
S-I3 (4.2%)2 (3.8%)
S-II23 (31.9%)15 (28.3%)
S-III36 (50.0%)29 (54.7%)
S-IV2 (2.8%)1 (1.9%)
NA8 (11.1%)6 (11.3%)
Tumor size (p)
pT011 (15.3%)7 (13.2%)
pT122 (30.5%)21 (39.6%)
pT228 (38.9%)19 (35.8%)
pT32 (2.8%)2 (3.8%)
NA9 (12.5%)4 (7.6%)
Lymph nodes (p)
pN029 (40.3%)25 (47.1%)
pN120 (27.8%)14 (26.4%)
pN212 (16.7%)8 (15.1%)
pN34 (5.6%)3 (5.7%)
NA7 (9.6%)3 (5.7%)
Metastasis (p)
pM01 (1.4%)0
pMx58 (80.5%)44 (83%)
NA13 (18.1%)9 (17%)
Lymphatic Invasion
L040 (55.6%)30 (56.6%)
L124 (33.3%)20 (37.7%)
NA8 (11.1%)3 (5.7%)
n = 62n = 53
Neodjuvant therapy
Only CT46 (74.2%)41 (77.4%)
Only HT10 (16.1%)9 (17%)
CT + HT5 (8.1%)3 (5.6%)
CT + HT + RTE1 (1.6%)-
MP System
Grade 119 (30.6%)18 (34%)
Grade 27 (11.3%)6 (11.3%)
Grade 317 (27.4%)16 (30.2%)
Grade 46 (9.7%)4 (7.5%)
Grade 513 (21.0%)9 (17%)
RCB
RCB 010 (16.1%)8 (15.1%)
RCB-I5 (8.1%)5 (9.4%)
RCB-II31 (50.0%)25 (47.2%)
RCB-III16 (25.8%)15 (28.3%)
ER—estrogen receptor, PR—progesterone receptor, TNBC—triple negative BC, cT—clinic tumor, cN—clinic lymph node, cM—clinic metastasis, pT—pathologic tumor, pN—pathologic lymph node, pM—pathologic metastasis, CT—chemotherapy, HT—hormone therapy, RTE—radiotherapy.
Table 2. Candidate biomarker miRNAs selection and evaluation in plasma sEVs.
Table 2. Candidate biomarker miRNAs selection and evaluation in plasma sEVs.
miRNAAmplification in Plasma sEVsGSE25066Pharmaco-miRNCI-60
hsa-miR-125b-5phighpredictedpaclitaxeldoxorubicin
hsa-miR-146a-5pdoxorubicindoxorubicin
hsa-miR-17-5p
hsa-miR-185-5p
hsa-miR-193b-3p
hsa-miR-21-5pdocetaxel, paclitaxel
doxorubicin
hsa-miR-221-3p
hsa-miR-24-3p doxorubicin
hsa-miR-26a-5p
hsa-miR-484
hsa-miR-92a-3p taxol
hsa-miR-143-3p docetaxel
hsa-let7c-5plowpredicted
hsa-miR-130b-3ptaxoltaxol
hsa-miR-140-3p and 5p
hsa-miR-18a-5p
hsa-miR-192-5p taxol
hsa-miR-328-3p taxol
hsa-miR-195-5p
hsa-let-7e-5p taxol
hsa-miR-31-5p doxorubicin
hsa-miR-197-3pnegativepredicted
hsa-miR-200b-5p
hsa-miR-203a-3p and 5ppaclitaxeldoxorubicin
hsa-miR-215-5p
hsa-miR-34a-5pdocetaxel, paclitaxel
hsa-miR-373-3p
hsa-miR-520h
hsa-let-7a-3p doxorubicin, paclitaxeltaxol
hsa-miR-375-3p doxorubicin
hsa-miR-589-5p
Table 3. Associations between clinical-pathological features and sEVs miRNA expression.
Table 3. Associations between clinical-pathological features and sEVs miRNA expression.
miR-21-5pmiR-125-5pmiR-221-3pmiR-143-3pmiR-146a-5pmiR-24-3pmiR-193b-3pmiR-92a-3pmiR-484miR-185-5pmiR-17-5pmiR-26a-5p
p-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Valuep-Value
Age0.5920.7220.3290.9820.7730.3410.9040.4040.8170.1100.3930.811
R = 0.06R = 0.04R = 0.12R = −0.003R = −0.03R = −0.11R = 0.02R = −0.10R = −0.03R = −0.19R = −0.10R = −0.03
Grading Biopsy0.1930.1380.3850.2150.2870.3200.5290.2530.6900.4180.6390.846
G3 vs. G2 vs. G1
Estrogen receptor0.8200.4150.3870.8020.1450.2220.7220.1490.2380.015 *0.9330.933
ER+ vs. ER−FR = −1.21
Progesteron receptor0.9390.1550.3780.5960.0590.0640.9350.1050.2280.039 *0.6140.828
PR+ vs. PR−FR = −1.02
HER20.1560.0600.8870.3160.0570.044 *0.7820.1990.2680.035 *0.7280.286
HER2+ vs. HER2−FR = 1.54FR = 2.11
KI670.6860.8010.7350.6080.3200.4490.2040.4690.7580.3780.8230.917
>20 vs. ≤20
Molecular Subtype0.9980.7360.6410.9340.3380.5190.9640.1090.4350.028 *0.7950.938
TNBC vs.FR TNBC vs. LumB = 1.56 **
LuminalB vs. LuminalA
cT0.1040.7990.4260.3990.2080.6520.4560.9860.6080.1490.5300.503
T4 vs. T3 vs. T1 + T2
cN0.0600.7180.2850.009 **0.025 *0.027 *0.025 *0.001 **0.005 **0.0006 ***0.0540.030 *
Positive (N1 + N2 + N3) vs. Negative (N0)FR = −1.47FR = −1.47FR = −1.64FR = −1.69FR = −2.13FR = −2.34FR = −2.19FR = −2.01
Clinical Stage0.3290.7470.7150.3600.4990.2330.1930.6220.5400.3510.037 *0.794
High (S3 + S4) vs. Low (S1 + S2)FR = −1.93
pT0.3410.7520.2090.7550.6340.7460.1270.8910.8450.4480.9150.673
T2 vs. T1 vs. T0
pN0.2860.6820.024 *0.6140.9990.4990.5370.5510.4070.6150.3290.183
Positive (N1 + N2 + N3) vs. Negative (N0)FR = 1.69 *
Lymphatic Invasion0.6370.8500.7890.8430.5150.5860.9090.5900.1030.1660.2530.915
L1 vs. L0
MP0.0004 ***
FR G5 vs. G1 =
−2.32 **
FR G3 vs. G1 =
−3.54 ****
FR G2 vs. G1 =
−3.31 **
0.1610.008 **
FR G5 vs. G1 =
−1.33 *
FR G3 vs. G1 =
−1.87 ***
0.1740.017 *
FR G4 vs. G3 =
2.27 *
FR G3 vs. G1 =
−3.79 **
0.7030.1750.1200.5280.1780.9940.045 *
FR G3 vs. G1 =
−2.32 **
FR G2 vs. G1 =
−1.88 *
G5 vs. G4 vs. G3 vs. G2 vs. G1
RCB0.3030.5960.7940.6860.7610.4420.4230.4970.6720.0970.4770.860
III vs. II vs.0 + I
ER—estrogen receptor, PR—progesterone receptor, TNBC—triple negative BC, cT—clinic tumor, cN—clinic lymph node, pT—pathologic tumor, pN—pathologic lymph node, RCB—residual cancer burden, FR—fold regulation. The table shows the p-values for all the compared groups (as indicated in the first column) and where these differences were statistically significant, the expression level of that miRNAs (FR) in the interest group versus the reference group was calculated. According to data distributions, the differences in expression in case of two groups was evaluated with Mann–Whitney test and for three groups with Kruskal–Wallis test, followed by Dunn’s multiple comparison post hoc test. The p-values were highlighted in red when a significant value (<0.05) was obtained, and asterisks indicate the magnitude of p-value (* p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001). In case of three or more groups, the asterisk next to the FR value is associated to the p-value obtained by the Dunn test.
Table 4. Associations of miRNAs expression in tumor tissue with clinical-pathological features and plasma sEVs.
Table 4. Associations of miRNAs expression in tumor tissue with clinical-pathological features and plasma sEVs.
miR-21-5pmiR-221-3pmiR-146a-5pmiR-26a-5p
p-Valuep-Valuep-Valuep-Value
Age0.8170.1830.2760.086
R = −0.03R = 0.19R = −0.15R = 0.24
Grading Biopsy0.045 *0.4350.0520.679
G3 vs. G2 vs. G1FR G3 vs. G2 = 1.39 *
FR G2 vs. G1 = −1.58 *
Estrogen receptor0.3440.3040.0003 ***0.573
ER+ vs. ER−FR = −3.11
Progesteron receptor0.6290.2650.042 *0.454
PR+ vs. PR−FR = −2.02
HER20.4280.034 *0.0610.034 *
HER2+ vs. HER2−FR = 1.60FR = 1.50
KI670.9460.9820.3140.158
>20 vs. ≤20
Molecular Subtype0.3950.6410.019 *0.042 *
TNBC vs.FR TNBC vs. LumB = 3.81 **FR TN vs. LumA = −1.87 *
LuminalB vs. LuminalA
cT0.1910.9070.1230.875
T4 vs. T3 vs. T1 + T2
cN0.0650.3770.3820.767
Positive (N1 + N2 + N3) vs. Negative (N0)
Clinical Stage0.1290.8830.0890.730
High (S3 + S4) vs. Low (S1 + S2)
pT0.6480.1510.0740.561
T2 vs. T1 vs. T0
pN0.6550.4440.3180.100
Positive (N1 + N2 + N3) vs. Negative (N0)
Lymphatic Invasion0.5860.8640.7350.139
L1 vs. L0
MP
G5 vs. G4 vs. G3 vs. G2 vs. G1
0.1490.020 *0.008 **0.230
FR G5 vs. G3 = 2.80 ***FR G5 vs. G3 = 5.00 ***
FR G5 vs. G1 = 1.58 *FR G3 vs. G2 = −2.90 *
FR G3 vs. G1 = −2.73 *
RCB0.1760.1970.1800.938
III vs. II vs.0 + I
Correlations between miRNAs expression in tumor tissue and sEVs
0.7810.4210.5430.953
R = 0.04R = 0.13R = 0.09R = −0.01
ER—estrogen receptor, PR—progesterone receptor, TNBC—triple negative BC, cT—clinic tumor, cN—clinic lymph node, pT—pathologic tumor, pN—pathologic lymph node, RCB—residual cancer burden, FR—fold regulation. The table shows the p-values for all the compared groups (as indicated in the first column) and where these differences were statistically significant, the expression level of that miRNAs (FR) in the interest group versus the reference group was calculated. According to data distributions, the differences in expression in case of two groups was evaluated with Mann–Whitney test and for three groups with Kruskal–Wallis test, followed by Dunn’s multiple comparison post hoc test. The p-values were highlighted in red when a significant value (<0.05) was obtained, and asterisks indicate the magnitude of p-value (* p < 0.05, ** p < 0.01, *** p < 0.001). In case of three or more groups, the asterisk next to the FR value is associated to the p-value obtained by the Dunn test.
Table 5. Candidate miRNAs target genes and processes.
Table 5. Candidate miRNAs target genes and processes.
NameGenesAdj p-Value *
miR-21-5p
GO:0004674 Protein serine/threonine kinase activityGSK3B; MAPK60
GO:0004842 Ubiquitin-protein transferase activityTOPORS; TRIM33; RMND5A0.0046
miR-221-3p
R-HSA-9609507 Protein localizationPEX1; CROT; PEX190.013
R-HSA-74160 Gene expression (Transcription)ESR1; TFAP2A; SIRT1; MDM2; APAF1; DICER1; BCL2L11; ZKSCAN8; ZNF571; WDR61; ZFP30; ARID1A; CSTF2T0.0136
R-HSA-73857 RNA Polymerase II TranscriptionESR1; TFAP2A; SIRT1; MDM2; APAF1; BCL2L11; ZKSCAN8; ZNF571; WDR61; ZFP30; ARID1A; CSTF2T0.0139
R-HSA-212436 Generic Transcription PathwayESR1; TFAP2A; SIRT1; MDM2; APAF1; BCL2L11; ZKSCAN8; ZNF571; ZFP30; ARID1A0.0179
R-HSA-5663202 Diseases of signal transduction by growth factor receptors and second messengersESR1; ERBB4; MDM2; BCL2L11; AP3B1; BRAP0.0335
GO:0006625 Protein targeting to peroxisomePEX1; CROT; PEX190
GO:0031648 Protein destabilizationSIRT1; MDM2; MYLIP0
GO:0006511 Ubiquitin-dependent protein catabolic processMDM2; MYLIP; RNF20; NDFIP10.003
GO:0016567 Protein ubiquitinationSIRT1; MDM2; WDR61; BRAP;
MYLIP; RNF20
0.0058
GO:0007399 Nervous system developmentERBB4; APAF1; EVL; ARID1A; MYLIP0.0101
GO:0043065 Positive regulation of apoptotic processSIRT1; APAF1; BCL2L110.0167
GO:0045893 Positive regulation of_transcription,_DNA-templatedESR1; TFAP2A; ERBB4; ARID1A; RNF200.019
GO:0008285 Negative regulation of cell_population_proliferationTFAP2A; ERBB4; EIF2AK1; DDAH1; BECN10.0196
GO:0045892 Negative regulation of transcription, DNA-templatedTFAP2A; PHF12; SIRT1; MDM20.0231
GO:0005777 PeroxisomePEX1; CROT; PEX190
GO:0005778 Peroxisomal membranePEX1; PEX19; ACSL30
GO:0032991 Protein-containing complexESR1; ACVR2B; MDM2; APAF1; PEX19; RBM39; SNX40.0006
GO:0004842 Ubiquitin-protein transferase activityMDM2; HECTD2; BRAP; MYLIP; RNF200.0002
GO:0061630 Ubiquitin protein ligase activityMDM2; BRAP; MYLIP; HECTD10.0002
GO:0003677 DNA bindingHMGXB4; TFAP2A; POGZ; DICER1; GLYR1; ARID1A; CERS2; PALB20.0032
miR-146a-5p
GO:0043235 Receptor complexERBB4; LRP20
miR-26a
R-HSA-162582_Signal TransductionGSK3B; KPNA2; MAPK60.05
GO:0098978 Glutamatergic synapseGSK3B; KPNA2; HSPA80
GO:0004674 Protein serine/threonine kinase activityGSK3B; MAPK60
* Benjamini-Hochberg (BH).
Table 6. Drug interaction predictions.
Table 6. Drug interaction predictions.
miRNATarget GeneTargeting Drug
hsa-miR-21-5pAP1topotecan
BCL2; PTEN; RECKgemcitabine
CDC25A3,3′-diindolylmethane
ICAM1; SERPINB5tretinoin
LRRFIP1teniposide
MSH2; PDCD2; PTEN; SPRY25-fluorouracil
PDCD4arsenic trioxide, cytarabine, cisplatin, docetaxel, doxorubicin, paclitaxel,
PTENcurcumin, daunorubicin, doxorubicin, transtuzumab,
SPRY2metformin
hsa-miR-221-3pABCB1trail
CDKN1Aglucocorticoids
CDKN1B; ESR1tamoxifen
CDKN4gemcitabine
hsa-miR-146a-5pCFHpyrollidine dithiocarbamate, resveratrol
ERBB4doxorubicin
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Baldasici, O.; Balacescu, L.; Cruceriu, D.; Roman, A.; Lisencu, C.; Fetica, B.; Visan, S.; Cismaru, A.; Jurj, A.; Barbu-Tudoran, L.; et al. Circulating Small EVs miRNAs as Predictors of Pathological Response to Neo-Adjuvant Therapy in Breast Cancer Patients. Int. J. Mol. Sci. 2022, 23, 12625. https://doi.org/10.3390/ijms232012625

AMA Style

Baldasici O, Balacescu L, Cruceriu D, Roman A, Lisencu C, Fetica B, Visan S, Cismaru A, Jurj A, Barbu-Tudoran L, et al. Circulating Small EVs miRNAs as Predictors of Pathological Response to Neo-Adjuvant Therapy in Breast Cancer Patients. International Journal of Molecular Sciences. 2022; 23(20):12625. https://doi.org/10.3390/ijms232012625

Chicago/Turabian Style

Baldasici, Oana, Loredana Balacescu, Daniel Cruceriu, Andrei Roman, Carmen Lisencu, Bogdan Fetica, Simona Visan, Andrei Cismaru, Ancuta Jurj, Lucian Barbu-Tudoran, and et al. 2022. "Circulating Small EVs miRNAs as Predictors of Pathological Response to Neo-Adjuvant Therapy in Breast Cancer Patients" International Journal of Molecular Sciences 23, no. 20: 12625. https://doi.org/10.3390/ijms232012625

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