MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib

  • Authors:
    • A.L.R. Bordinhão
    • A.F. Evangelista
    • R.J.S. Oliveira
    • T. Macedo
    • H.C. Silveira
    • R.M. Reis
    • M.M. Marques
  • View Affiliations

  • Published online on: October 7, 2016     https://doi.org/10.3892/or.2016.5153
  • Pages: 3197-3206
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cediranib, a pan-tyrosine kinase inhibitor is showing promising results for the treatment of several solid tumours. In breast cancer, its effects remain unclear, and there are no predictive biomarkers. Several studies have examined the expression profiles of microRNAs (miRNAs) in response to different chemotherapy treatments and found that the expression patterns may be associated with the treatment response. Therefore, our aim was to evaluate the cellular behaviour and differential expression profiles of miRNAs in breast cancer cell lines exposed to cediranib. The biological effect of this drug was measured by viability, migration, invasion and cell death in in vitro assays. Signaling pathways were assessed using a human phospho-receptor tyrosine kinase array. Furthermore, using a miRNA array and quantitative real-time PCR (qRT‑PCR), we assessed the relative expression of miRNAs following cediranib treatment. The breast cancer cell lines exhibited a distinct cytotoxic response to cediranib treatment. Cediranib exposure resulted in a decrease in the cell migration and invasion of all the breast cancer cell lines. Treatment with cediranib appeared to be able to modulate the activation of several RTKs that are targets of cediranib such as EGFR and a new potential target ROR2. Furthermore, this drug was able to modulate the expression profile of different microRNAs such as miR-494, miR-923, miR-449a, miR-449b and miR-886-3 in breast cancer cell lines. These miRNAs are reported to regulate genes involved in important molecular processes, according to bioinformatics prediction tools.

Introduction

Breast cancer is the second most prevalent neoplastic disease among women and is characterised by a complex aetiology and chemoresistant behaviour (1,2). Although most women are treated with tamoxifen, the current standard adjuvant therapy in women with estrogen receptor (ER)-positive breast cancer, they develop resistance to the drug (3). Cediranib (AZD2171, Recentin, AstraZeneca) is a potent inhibitor of several receptor tyrosine kinases (RTKs), such as VEGFR, KIT and PDGFRA (4,5). Significant results have been obtained for patients with advanced solid tumours, such as glioblastoma, lung and prostate tumours, after cediranib treatment (69). Recently, novel targets of cediranib have been identified by in vitro and in vivo studies with glioblastoma cell lines (10). Furthermore, it was shown that cediranib alone or in combination with temozolomide is an effective drug in anti-angiogenic therapies due to its greater antitumour activity (10). Animal models of breast cancer revealed that cediranib affected vessel density and cell proliferation, but not with chronic administration (11,12). Phase I/II studies have been conducted with cediranib for breast cancer treatment, especially in combination with other drugs, such as fulvestrant, olaparib and RO4929097, a γ-secretase inhibitor (1315). The promise of cediranib treatment is an improvement in overall survival of up to six months in several tumour types (6,7), and while it has not yet been approved by the FDA, cediranib, alone or in combination with other drugs, represents a new potential therapy for breast cancer.

Despite these observations, in breast cancer cell lines, the effects of cediranib on cellular processes such as proliferation, migration and invasion are unknown. Therefore, exploring the biological effect of cediranib on breast cancer cell lines and revealing the pathways that can be predictive biomarkers of cediranib response are necessary. MicroRNAs (miRNAs) are small noncoding RNAs approximately 22–26 nucleotides (nt) long that are involved in post-transcriptional repression or mRNA degradation in a sequence-specific manner (16,17). The role of miRNAs as new regulatory molecules that are involved in different cancer processes have recently emerged (18). Several studies on miRNA profiling have shown that these molecules can act as oncogenes and tumour suppressors (19). Accordingly, strategies to correct the deficiencies associated with miRNA dysregulation have been proposed as future clinical interventions for cancer patients (20). In breast cancer, several miRNAs have been described in several steps of tumorigenesis, angiogenesis and metastasis (2123). Furthermore, miRNAs have been considered as potential biomarkers in breast cancer metastasis (24).

miRNAs also play a role in anticancer drug resistance (25). Several miRNAs have been associated with chemoresistance in breast cancer, including miR-34a with docetaxel, miR-125b with anthracycline, miR-451 and miR-27 with doxorubicin and miR-326 with doxorubicin and VP-16 resistance (2631). Several reports have shown that deletions of miRNA regions can affect the regulation of RTKs and the response to gefitinib or imatinib (32,33), reinforcing a possible role for miRNAs in the mechanism of the response and/or resistance to RTK inhibitors.

Overall, miRNAs can affect breast cancer drug sensitivity with implications in clinical management and in understanding the molecular mechanisms that may contribute to drug susceptibility and resistance (17). Therefore, in the present study, we aimed to evaluate the miRNA expression profiles of breast cancer cell lines exposed to cediranib.

Materials and methods

Cell lines and determination of the half-maximal inhibitory concentration (IC50)

In the present study, the breast carcinoma cell lines Hs578T, MDA-MB-231 and T47D were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). Cells were cultured in Dulbeccos modified Eagles medium (DMEM 1X, high glucose), supplemented with 10% FBS (both from Gibco, Invitrogen Life Technologies Grand Island, NY, USA) and 1% penicillin and streptomycin solution (Sigma-Aldrich) at 37°C and 5% CO2.

Authentication of cell lines was performed by short tandem repeat (STR) DNA typing according to the International Reference Standard for Authentication of Human Cell Lines using a panel of eight (D5S818, D13S317, D7S820, D16S539, vWA, TH01, TPOX and CSF1P0) STR loci plus gender determination (AMEL), using the fluorescent labeling primers as reported by Dirks et al (34). Briefly, 50 ng of DNA was amplified in multiplex PCR reaction carried out in a total volume of 10 µl with Qiagen Multiplex PCR kit (Qiagen) comprising 0.5 µM of all fluorescent primer pairs plus 1 µM of TH01 primer reinforcement, performed in a Veriti® 96-well Thermal Cycler with an initial denaturation at 95°C for 15 min, amplified for 30 cycles of denaturation at 95°C for 30 sec, annealing at 55°C for 1 min 30 sec, extension at 72°C for 1 sec and a final cycle at 72°C for 30 min. The DNA amplification products were diluted 1:100 in Nuclease-Free Water Ultrapure (USB, Cleveland, OH, USA) and combined with 0.3 µl of internal size standard GeneScan 500 ROX (Applied Biosystems, Foster City, CA, USA) in 8.7 µl formamide and loaded automatically in a capillary electrophoresis using an Genetic Analyzer ABI PRISM 3500 (Applied Biosystems). The analysis was performed in GeneMapper software version 4.1 (Applied Biosystems). Genotyping confirmed the identity of all three cell lines.

The drug used, cediranib (Selleck Chemicals, Houston, TX, USA), was diluted in 1% DMSO. To determine the IC50, the cells were plated in increasing concentrations of the drug from 0.1 to 100 µM, and an MTS proliferation assay was performed. The IC50 concentration was calculated using the drc package in R (35,36), and the best model for each cell type was selected according to the Akaike information Criterion (AIC).

Invasion and migration assays

The invasion assays were performed using BD BioCoat Matrigel invasion chambers (BD Biosciences) according to the manufacturers instructions and as previously described (10,37). Briefly, 2.5×104 cells were plated in the Matrigel-coated 24-well Transwell inserts in DMEM-0.5% containing fixed concentrations of the drug. DMEM-10% was used as a chemoattractant, and the cells that attached to the inserts were fixed with methanol and stained with haematoxylin. The cells were photographed at a ×40 magnification level and counted on a pixel-by-pixel basis using ImageJ software. The invasiveness of the cells exposed to cediranib (IC50 dose at 72 h) was expressed in relation to the DMSO control (considered 100% invasion) as the mean percentage of invasion ± SD. The assays were performed in triplicate.

The migration capacity of the cells after cediranib exposure was assessed using a wound-healing assay as previously described (10,38). The cells were seeded in 6-well plates and cultured until reaching 95% confluency. The monolayer of cells was washed with PBS; a wound was created, and the cells were incubated with the IC50 concentrations of cediranib for varying times. The selected areas were photographed at ×40 magnification at 0 to 24 h for the Hs578T and MDA-MB-231 cells and at 0 to 72 h for T47D cells. The relative migration distance was calculated using the following formula: Percentage of wound closure (%) = 100(A − B)/A, where A is the width of the cell wound before incubation and B is the width of the cell wound after different lengths of cediranib exposure. The assay was performed in triplicate with four measurements for each well.

Western blot analysis and human phospho-RTK array

To evaluate the inhibition of the intracellular signalling pathways, Hs578T, MDA-MB-231 and T47D cells were cultured in T25 culture flasks in DMEM 10% FBS. After reaching 90% confluency, the cell lines were starved and exposed to cediranib (IC50 values) for 12 h. Analysis of apoptosis was conducted in 6-well culture plates at 70% confluency and incubated for 12, 24 and 72 h. For all experiments, after time points, the cell lines were washed and scraped in cold PBS and lysed in buffer, containing 50 mM Tris, 150 mM NaCl, 5 mM EDTA, 1 mM Na3VO4, 10 mM NaF, 10 mM sodium pyrophosphate, 1% NP-40 and protease cocktail inhibitors. Western blot analysis was performed using standard 10% sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) loading 30 µg protein per line. To assess apoptosis and the activation of intracellular signaling pathways, the antibodies used were the following: primary antibody PARP total/cleaved [Cell Signaling Technology (CST), no. 9532]; total p44/42 MAPK (CST, no. 137F5) and phospho-p44/42 MAPK (CST, no. 8544); pan AKT (CST, no. C67E7) and phospho-Akt (Ser473) (CST, no. 4060); β-actin (CST, no. 12262). Secondary antibodies were used according to the manufacturers instructions. Immune blots were performed using ECL western blotting detection reagent (GE Healthcare) and finally the bands were detected and images were captured using an Automatic ImageQuant Mini LAS 4000 (GE Healthcare).

Human phospho-RTK array (PN no. 894042, R&D Systems) was used according to the manufacturers instructions. In brief, after the blocking step, 750 µg of protein were incubated overnight at 4°C with nitrocellulose membranes containing 49 different anti-RTK Abs spots in duplicate. Next they were incubated with anti-phospho-tyrosine-HRP for 2 h and detection was performed with a Chemi Reagent Mix in Automatic ImageQuant Mini LAS 4000 (GE Healthcare).

RNA isolation and quality control

Total RNA was isolated using an miRNeasy kit (Qiagen) according to the manufacturers instructions. Quantification was performed using a NanoDrop ND-1000 spectrophotometer (NanoDrop Products, Wilmington, DE, USA), and the RNA quality was assessed using an Agilent Nano RNA chip with a bioanalyser device (Agilent Technologies), as previously described (39).

miRNA microarrays

To assess the expression of miRNAs after 24 h of treatment with an IC50 dose of cediranib in Hs578T, MDA-MB-231 and T47D cells and the same cell lines without treatment were considered as controls, the Agilent Human miRNA Microarray (8×15K-G4471A, Agilent Technologies) was used. Total RNA samples (200 ng) were hybridised using an miRNA complete labelling and a Hyb kit (Agilent Technologies) according to the manufacturers instructions. The reactions followed a two-step preparation; an initial dephosphorylation and denaturation of the total RNA was performed, and the Cy3 fluorochrome was then incorporated with T4 ligase. The next steps included standard washing procedures and hybridisation with the microarray slides. Images were scanned with an Agilent DNA microarray scanner with SureScan technology (Agilent Technologies), as previously described (40).

miRNA microarray data analysis

The raw data were obtained using the Feature Extraction software v.11.0 (Agilent Technologies) and submitted to R environment version 3.0.1 (36) for additional analyses. Median signals (gMedianSignal and gBGMedianSignal) were used as intensity values. Normalisation was performed using the quantile method with the Bioconductor ‘aroma.light’ package (41). miRNAs differentially expressed between the cediranib-treated and control cells were obtained by rank product analysis considering a P-value and pfp (positive false predictions) ≤0.05 using the RankProd package (42). Heatmaps of the differentially expressed miRNAs were constructed using hclust R and the distance and average Pearson linkage were determined using the gplots R package (43).

Target prediction and functional analysis

Target prediction was performed using the mirDIP interface (44). At least 3 of the 12 available algorithms were selected for prediction. To perform a functional enrichment analysis, all the targets were separated according to up- or downregulation and submitted to the Database for Annotation, Visualization, and Integrated Discovery (DAVID) (45). This approach was used to identify significant biological processes in Gene Ontology level 2 that may be shared among the targets of the miRNAs of interest. A biological process or pathway was considered significant if it contained a minimum of 3 genes per category that had score values <0.05 after a Benjamini-Hochberg correction. A compilation of the categories according to a GoSlim summarisation was performed using the REVIGO tool (46).

Confirmation by quantitative real-time PCR (qRT-PCR)

qRT-PCR using TaqMan miRNA assays (Life Technologies, Foster City, CA, USA) was used to confirm the expression of five miRNAs from the microarray data that were differentially expressed between cells exposed to cediranib compared with the control group. The criterion for selecting the miRNAs for confirmation was the level of expression. All of the samples used in the microarray experiments were performed in technical triplicates for the RT-PCR reactions. A total of 10 ng of RNA was used in the reverse transcription reaction with miRNA-specific primers in an Eppendorf Mastercycler (Eppendorf). The real-time reactions were performed in a 7900 HT Fast Real-time PCR system (Applied Biosystems).

All of the analysis procedures and graphs were constructed using the R environment v.3.0.1. The normalisation step was performed according to the 2−ΔΔCt method (47) using the minimum value of expression of the untreated group as a calibrator. The cycle threshold (Ct) values from the selected miRNA targets were subtracted from the Ct values of the endogenous small noncoding RNA controls RNU44 and RNU48 (control miRNA assay; Applied Biosystems). The criterion for selecting the best endogenous RNA was the stability of expression according to the NormFinder software (48). Differences in miRNA expression between the treated and non-treated cell lines (Hs578T, MDA-MB-231 and T47D) were evaluated using Mann-Whitney U test, and differences with a P≤0.05 were considered significant.

Results

Following an initial assessment of the basal viability conditions for all the three breast cell lines, a total density of 4×103 cells/well for Hs578T, 6×103 cells/well for MDA-MB-231 and 1×104 cells/well for the T47D cell line was used. The IC50 was then determined for the breast cancer cells at 24, 48 and 72 h, as shown in Table I and Fig. 1. Our results showed that the most sensitive breast cancer cell line was Hs578T, followed by MDA-MB-231. The T47D cell line was the most resistant, with an IC50 value 4-fold higher than that of the Hs578T cells and almost 2-fold higher than that of the MDA-MB-231 cells after a 24-h treatment.

Table I.

Half-maximal inhibitory concentration (IC50) at 24, 48 and 72 h of cediranib exposure in the Hs578T, MDA-MB-231 and T47D cells.

Table I.

Half-maximal inhibitory concentration (IC50) at 24, 48 and 72 h of cediranib exposure in the Hs578T, MDA-MB-231 and T47D cells.

IC50 (µM)

Cell line24 h48 h72 h
Hs578T10.66±0.662.51±0.622.08±0.77
MDA-MB-23130.77±2.0115.57±3.032.52±0.81
T47D38.69±2.9026.54±2.7918.85±3.21

To determine whether decreased viability was due to cytotoxic effects, we analyzed poly(ADP ribose) polymerase (PARP) cleavage. We observed a striking effect on PARP cleavage using low doses (0.5 and 2.0 µM) of cediranib in the Hs578T cell line. Similar effects were demonstrated in the MDA-MB-231 cell line that exhibited higher cleaved PARP levels. In contrast, the T47D cell line showed lower PARP cleaved levels (8 and 18 µM) using a high dose of cediranib drug, representing a resistant profile. Cytotoxic effects demonstrated by cleaved PARP were not found at times less than 24 h for both cell lines (Fig. 2).

In order to assess the functional impact of cediranib in migration, we used the wound-healing migration assay, and observed the inhibition of cell migration in all of the cell lines. After 24 h of cediranib exposure, the Hs578T cells exhibited the most inhibition (80%), followed by the T47D cells (70%) and MDA-MB-231 cells (54%) (Fig. 3). Even the adhesion of this cell type appeared to be affected by the drug (data not shown), leading to negative values, i.e., the wound was extended because of the detachment of these cells. Due to this behaviour, we studied the T47D migration ability for 72 h. Similarly, cediranib treatment significantly inhibited cell invasion in all breast cancer cell lines. The reduction in the percentage of cell invasion at 24 h was ~70% in T47D, 60% in Hs578T and 30% in the MDA-MB-231 cells (Fig. 4).

To identify the RTKs that are targets of cediranib, we used a phospho-RTK array that assesses the levels of 49 RTKs, in the most sensitive cell line, Hs578T, which was exposed to 2 µM cediranib. Under basal levels, we observed the presence of the active forms of EGFR, Tie-2, RYK, FGFR2α, m-CSFR, c-RET, AXL and ROR2 (RTK-like orphan receptor 2) tyrosine kinase receptors (Fig. 5A). Following cediranib treatment, we found a slight increase in EGFR and Tie-2 phosphorylation levels (Fig. 5A-C) and diminished phosphorylation levels of FGFR2α and ROR2. We did not detect any significant changes in the phosphorylation of the other RTKs after treatment with cediranib (Fig. 5C).

We further addressed the inhibitory effect of cediranib in intracellular pathways, using higher concentrations of the drug for 12 h in both cell lines (Fig. 5D). We observed a decrease in ERK phosphorylation at the concentrations analyzed in the Hs578T cells. However, this cell line showed that the inhibition of AKT seemed to be dose-dependent (2 µM) after cediranib treatment. Both cell lines, HS578T and MDA-MB-231, were exposed to equal cediranib concentrations (0.5 and 2 µM), and only the MDA-MB-231 cell line did not exhibit decreased ERK phosphorylation levels. The absence of AKT phosphorylation was detected in the MDA-MB-231 cell line. The phosphorylation levels of ERK and AKT were unchanged in the T47D cell line after exposure to cediranib (Fig. 5D).

To further identify potential miRNAs in response to cediranib, we performed miRNA expression profiles in each breast cancer cell line and compared them with the controls. The results revealed 31 differentially expressed miRNAs in the Hs578T cells, 13 miRNAs in the MDA-MB-231 cells and 7 miRNAs in the T74D cell line (Fig. 6). The targets of these identified miRNAs are summarised according to their biological processes. The Hs578T cell line shared several processes with the MDA-MB-231 cells, including the induction of a response to endogenous/chemical stimuli, biosynthesis, vesicle-mediated transport, the movement of cellular components, the repression of the ubiquitin cycle, and the regulation of anti-apoptosis and cell death (Fig. 7). The T74D cell line presented specific biological processes; only biosynthesis was upregulated in the other cell lines in a similar manner, while vesicle-mediated transport and cellular component movement were repressed (Fig. 7).

Among the miRNAs identified, five miRNAs, miR-494, miR-923, miR-449a, miR-449b and miR-886-3p, were selected for further confirmation of expression due to the greater change in expression and the biological relevance of these miRNAs (Fig. 8). The endogenous miRNA RNU-48 was the most stable in our results, and this miRNA was thus used for all the analyses. The results confirmed the overexpression of miR-494 and miR-923 in the Hs578T cells exposed to cediranib compared to the control cells. In the MDA-MB-231 cells, the overexpression of miR-923 and the decreased expression of miR-886-3p after exposure to cediranib were confirmed. Both miR-449a and miR-449b were dowregulated in the T47D cell line after cediranib treatment.

Discussion

Cediranib is a tyrosine kinase inhibitor that represents promise in the treatment of several tumours (6). The main targets of this pharmacological drug are the VEGFR proteins, which regulate blood vessel formation in tumours and are associated with the anti-angiogenic effects of cediranib (4). In the present study, we showed that cediranib exhibited an effect on breast cancer cells, affecting cell migration and invasion, and the molecular processes associated with the miRNA expression altered in response to treatment was also studied.

Regarding the sensitivity to the agent, the IC50 value varied according to the cell line used. The most sensitive cell line (Hs578T) had IC50 values (~2 µM) similar to those of glioma cells after 72 h of cediranib exposure (10). Notably, PARP is an abundant, chromatin-associated enzyme which responds to DNA damage (49). Our results showed that when exposed to cediranib (according to the IC50 values) the cell line Hs578T displayed cleavage of PARP at 48 h of exposure, followed by MDA-MB-231 and T47D cell lines. Based on this result, we demonstrated that the Hs578T cell line had greater sensitivity to cediranib. ERK inhibition was detected only in sensitive cell line Hs578T, showing the efficient inhibition in the conserved RAS-mitogen activated protein kinase (MAPK) signalling pathway which may affect the cellular growth, survival, and differentiation (50). In the MDA-MB-231 and T47D cell lines, the phosphorylation of ERK was not significantly blocked. A recent study showed similar results in another breast cancer cell line, which presented a resistance profile when exposed to cediranib at 1–10 µM/l (51). The persistent AKT activation in some solid cancers, such as non-small cell lung carcinomas (NSCLCs), is associated with TKI response (52). We observed a total blockade of the AKT phosphorylation in the Hs578T cell line when exposed to different cediranib concentrations. However the MDA-MB-231 cell line showed absence of phosphorylated proteins and T47D displayed no significant changes. Therefore, we may hypothesized that AKT inhibition may be related with cediranib response in breast cancer cell lines.

The proteome profiler of phospho-RTKs showed that although Hs578T cells did not express the described cediranib targets under baseline conditions, such as VGFRs, KIT or PDGFRA, this cell line exhibited other important RTKs, such as EGFR, Tie-2, RYK, FGFR2α, m-CSFR, c-RET, AXL and ROR2, and upon cediranib exposure we observed reduction of the ROR2 and FGFR2α RTKs. ROR2 is a novel Wnt receptor recently discovered and is associated with progression of solid tumours such as melanoma, osteosarcoma and prostate cancer (53). An immunohistochemistry study conducted in patients with breast cancer showed ROR2 overexpression in 87% of the cases, and its association with a worse outcome (54). Further studies are needed to validate our findings, and more importantly to determine whether the expression levels of ROR2 and FGFR2α in human breast cancer tissue may predict the response to cediranib.

Regarding the expression of miRNAs, the Hs578T cell line presented a greater number of modulated miRNAs than the MDA-MB-231 and T47D cell lines, as an indicator of sensitivity to the drug. Although the Hs578T cell line presented 22 specific miRNAs, the most significant biological processes, such as biological regulation, development, metabolic processes, cell motility and homeostasis, were related to targets that were shared with the MDA-MB-231 cell line, including VEGFA, VEGFC, and PDGFRA. Several studies support the roles of these molecules in these processes, especially roles in tissue metabolism and homeostasis in breast cancer (55,56).

Of the targets shared between the Hs578T and MDA-MB-231 cell lines, miR-923 upregulation was observed. Moreover, few miRNAs were upregulated in our findings. miR-923 dysregulation was highly associated with drug resistance. The upregulation of this miRNA was also observed in breast cancer cells that are resistant to Taxol (57). In the same study, miR-125b was upregulated, which was associated with Bcl-2 antagonist killer 1 (Bak1) expression. In our findings, miR-125b was also modulated but was downregulated. The interaction between specific targets may further be explored. miR-923 and miR-886-3p, which were also validated in the present study, were shown to be involved in cisplatin resistance in bladder cancer; changes in expression were also correlated with survival (58). The upregulation of miR-923 was also associated with multidrug resistance in the HEp-2 cell line (59).

Among the specific miRNAs found in the MDA-MB-231 cell line, miR-886 has been recently proposed as a vault RNA (vtRNA2-1), which may form a complex that is implicated in cancer drug resistance. Recently, Lee et al (60) identified pre-miR-886 as a 102-nt, abundant cytoplasmic RNA that is neither a pre-miRNA nor a vault RNA but is a noncoding molecule (nc886). Some evidence indicates that nc886 in an immature state is physically associated with PKR (protein kinase RNA-activated), a double-stranded RNA-dependent kinase (61).

The involvement of miR-886 in the drug response and the modulation of this molecule by cediranib in the sensitive cell line MDA-MB-231 supports evidence that some mechanisms related to apoptosis could be activated in this context and affect the sensitivity of these cells, as observed in the present study. In addition, miR-886-3p repression was recently described as being mediated by methylation processes in lung cancer cell lines (62) and can affect cell proliferation, migration, and invasion in lung and non-medullary thyroid cancer (63,64). This fact is in concordance with our findings because we determined that migration and invasion processes were highly affected by cediranib in MDA-MB-231 cells.

In contrast to the other cell lines studied, few miRNAs were modulated in the T47D cell line. The T47D-specific miRNAs miR-449a and miR-449b were highly downregulated, and these results were confirmed via qRT-PCR. The miRNA miR-449 has been identified in a wide range of tumours (6569) and has several identified targets, including c-Myc and c-Met (66,67). This miRNA induces cell cycle progression and apoptosis by regulating CDK6 and CDC25A, direct targets that lead to the phosphorylation of E2F1 (7072). In contrast to the Hs578T and MDA-MB-231 cells, no target associated with VEGFR or PDGFRA was identified in the T47D cells with the target prediction. Considering the difference in the T47D resistance to cediranib, this finding suggests that other RTK targets are possibly affected by cediranib, in addition to VEGFR, the most potent RTK affected, and these other targets may activate other downstream pathways that affect the invasion and migration processes, instead of cell death. Further studies are necessary to confirm this hypothesis.

The crosstalk of the EGFR pathways and certain miRNAs has been described; for example, EGFR mutations can regulate miR-21 in lung cancer and glioblastoma cell lines (73). Similarly, miR-145 was found to inhibit cell proliferation by targeting EGFR in lung adenocarcinoma and in a model of colon cancer (74,75). In the present study, both miRNAs were modulated in the Hs578T cells, and only miR-21 was downregulated in MDA-MB-231 cells. However, some different processes seem to occur in the T47D cells. Considering that miR-449 (specific to T47D) can also modulate EGFR, the ambiguities of this process should be further explored.

The present study assessed for the first time the anti-neoplastic effects of cediranib in breast cancer cell lines. We observed a distinct degree of response in all the cell lines. Cediranib impaired mechanisms leading to cancer cell invasion and migration. The present study revealed an anti-neoplastic role of cediranib in human breast cancer cell lines. Importantly, we identified ROR2 and FGFR2α as novel RTK targets of cediranib. The miRNA expression profiling revealed the effect of this drug on several miRNAs involved in several molecular processes. Finally, more studies are necessary to validate these miRNA signatures as indicators of the therapeutic response in human breast cancer, and these signatures may lead to the selection of patients who would be most effectively treated with cediranib.

Acknowledgements

We would like to thank Olga Martinho and Celine Pinheiro for assisting in the cellular experiments. This study received financial support from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP Proc. no. 2010/16796-0, São Paulo, Brazil).

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Lønning PE: Molecular basis for therapy resistance. Mol Oncol. 4:284–300. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Arpino G, Green SJ, Allred DC, Lew D, Martino S, Osborne CK and Elledge RM: HER-2 amplification, HER-1 expression, and tamoxifen response in estrogen receptor-positive metastatic breast cancer: A southwest oncology group study. Clin Cancer Res. 10:5670–5676. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Wedge SR, Kendrew J, Hennequin LF, Valentine PJ, Barry ST, Brave SR, Smith NR, James NH, Dukes M, Curwen JO, et al: AZD2171: A highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res. 65:4389–4400. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Brave SR, Ratcliffe K, Wilson Z, James NH, Ashton S, Wainwright A, Kendrew J, Dudley P, Broadbent N, Sproat G, et al: Assessing the activity of cediranib, a VEGFR-2/3 tyrosine kinase inhibitor, against VEGFR-1 and members of the structurally related PDGFR family. Mol Cancer Ther. 10:861–873. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Drevs J, Siegert P, Medinger M, Mross K, Strecker R, Zirrgiebel U, Harder J, Blum H, Robertson J, Jürgensmeier JM, et al: Phase I clinical study of AZD2171, an oral vascular endothelial growth factor signaling inhibitor, in patients with advanced solid tumors. J Clin Oncol. 25:3045–3054. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Batchelor TT, Duda DG, di Tomaso E, Ancukiewicz M, Plotkin SR, Gerstner E, Eichler AF, Drappatz J, Hochberg FH, Benner T, et al: Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol. 28:2817–2823. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Goss GD, Arnold A, Shepherd FA, Dediu M, Ciuleanu TE, Fenton D, Zukin M, Walde D, Laberge F, Vincent MD, et al: Randomized, double-blind trial of carboplatin and paclitaxel with either daily oral cediranib or placebo in advanced non-small-cell lung cancer: NCIC clinical trials group BR24 study. J Clin Oncol. 28:49–55. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Dahut WL, Madan RA, Karakunnel JJ, Adelberg D, Gulley JL, Turkbey IB, Chau CH, Spencer SD, Mulquin M, Wright J, et al: Phase II clinical trial of cediranib in patients with metastatic castration-resistant prostate cancer. BJU Int. 111:1269–1280. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Martinho O, Silva-Oliveira R, Miranda-Gonçalves V, Clara C, Almeida JR, Carvalho AL, Barata JT and Reis RM: In vitro and in vivo analysis of RTK inhibitor efficacy and identification of its novel targets in glioblastomas. Transl Oncol. 6:187–196. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Miller KD, Miller M, Mehrotra S, Agarwal B, Mock BH, Zheng QH, Badve S, Hutchins GD and Sledge GW Jr: A physiologic imaging pilot study of breast cancer treated with AZD2171. Clin Cancer Res. 12:281–288. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Denduluri N, Tan AR, Walshe J, Berman A, Yang SX, Chow CK and Swain SM: A pilot study to evaluate the vascular endothelial growth factor receptor tyrosine kinase inhibitor AZD2171 and chemotherapy in locally advanced and inflammatory breast cancer. Clin Breast Cancer. 6:460–463. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Hyams DM, Chan A, de Oliveira C, Snyder R, Vinholes J, Audeh MW, Alencar VM, Lombard J, Mookerjee B, Xu J, et al: Cediranib in combination with fulvestrant in hormone-sensitive metastatic breast cancer: A randomized Phase II study. Invest New Drugs. 31:1345–1354. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Liu JF, Tolaney SM, Birrer M, Fleming GF, Buss MK, Dahlberg SE, Lee H, Whalen C, Tyburski K, Winer E, et al: A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer. 49:2972–2978. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Sahebjam S, Bedard PL, Castonguay V, Chen Z, Reedijk M, Liu G, Cohen B, Zhang WJ, Clarke B, Zhang T, et al: A phase I study of the combination of ro4929097 and cediranib in patients with advanced solid tumours (PJC-004/NCI 8503). Br J Cancer. 109:943–949. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Almeida MI, Reis RM and Calin GA: MicroRNA history: Discovery, recent applications, and next frontiers. Mutat Res. 717:1–8. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, et al: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–789. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Harquail J, Benzina S and Robichaud GA: MicroRNAs and breast cancer malignancy: An overview of miRNA-regulated cancer processes leading to metastasis. Cancer Biomark. 11:269–280. 2012.PubMed/NCBI

22 

Singh R and Mo YY: Role of microRNAs in breast cancer. Cancer Biol Ther. 14:201–212. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Valastyan S: Roles of microRNAs and other non-coding RNAs in breast cancer metastasis. J Mammary Gland Biol Neoplasia. 17:23–32. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Marino ALF, Evangelista AF, Vieira RAC, Macedo T, Kerr LM, Abrahão-Machado LF, Longatto-Filho A, Silveira HC and Marques MM: MicroRNA expression as risk biomarker of breast cancer metastasis: A pilot retrospective case-cohort study. BMC Cancer. 14:7392014. View Article : Google Scholar : PubMed/NCBI

25 

Wiemer EAC: Role of microRNAs in anti-cancer drug resistanceMicroRNAs in Cancer Translational Research. Cho WCS: Springer; Netherlands: pp. 449–483. 2011, http://link.springer.com/chapter/10.1007/978-94-007-0298-1_19Accessed November 20, 2013. View Article : Google Scholar

26 

Kastl L, Brown I and Schofield AC: miRNA-34a is associated with docetaxel resistance in human breast cancer cells. Breast Cancer Res Treat. 131:445–454. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Climent J, Dimitrow P, Fridlyand J, Palacios J, Siebert R, Albertson DG, Gray JW, Pinkel D, Lluch A and Martinez-Climent JA: Deletion of chromosome 11q predicts response to anthracycline-based chemotherapy in early breast cancer. Cancer Res. 67:818–826. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Zhu H, Wu H, Liu X, Evans BR, Medina DJ, Liu CG and Yang JM: Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem Pharmacol. 76:582–588. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Kovalchuk O, Filkowski J, Meservy J, Ilnytskyy Y, Tryndyak VP, Chekhun VF and Pogribny IP: Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol Cancer Ther. 7:2152–2159. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Liang Z, Wu H, Xia J, Li Y, Zhang Y, Huang K, Wagar N, Yoon Y, Cho HT, Scala S, et al: Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem Pharmacol. 79:817–824. 2010. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Mahadevan D, Cooke L, Riley C, Swart R, Simons B, Della Croce K, Wisner L, Iorio M, Shakalya K, Garewal H, et al: A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors. Oncogene. 26:3909–3919. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Weiss GJ, Bemis LT, Nakajima E, Sugita M, Birks DK, Robinson WA, Varella-Garcia M, Bunn PA Jr, Haney J, Helfrich BA, et al: EGFR regulation by microRNA in lung cancer: Correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Ann Oncol. 19:1053–1059. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Dirks WG, Faehnrich S, Estella IAJ and Drexler HG: Short tandem repeat DNA typing provides an international reference standard for authentication of human cell lines. ALTEX. 22:103–109. 2005.PubMed/NCBI

35 

drc R Package. http://cran.r-project.org/web/packages/drc/

36 

The R project for statistical computing. http://www.r-project.org

37 

Pinto F, Pértega-Gomes N, Pereira MS, Vizcaíno JR, Monteiro P, Henrique RM, Baltazar F, Andrade RP and Reis RM: T-box transcription factor brachyury is associated with prostate cancer progression and aggressiveness. Clin Cancer Res. 20:4949–4961. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Martinho O, Zucca LE and Reis RM: AXL as a modulator of sunitinib response in glioblastoma cell lines. Exp Cell Res. 332:1–10. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Viana CR, Neto CS, Kerr LM, Palmero EI, Marques MMC, Colaiacovo T, de Queiroz Junior AF, Carvalho AL and Siqueira SA: The interference of cold ischemia time in the quality of total RNA from frozen tumor samples. Cell Tissue Bank. 14:167–173. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Macedo C, Evangelista AF, Marques MM, Octacílio-Silva S, Donadi EA, Sakamoto-Hojo ET and Passos GA: Autoimmune regulator (Aire) controls the expression of microRNAs in medullary thymic epithelial cells. Immunobiology. 218:554–560. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Aroma-light. http://www.bioconductor.org/packages/2.12/bioc/html/aroma.light.html

42 

Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL and Chory J: RankProd: A bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics. 22:2825–2827. 2006. View Article : Google Scholar : PubMed/NCBI

43 

gplots: Various R programming tools for plotting data. http://cran.r-project.org/web/packages/gplots/index.html

44 

mirDIP: microRNA data integration portal. http://ophid.utoronto.ca/mirDIP/

45 

Bioinformatics Resouces DAVID: 6.7. http://david.abcc.ncifcrf.gov

46 

Supek F, Bošnjak M, Škunca N and Šmuc T: REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS One. 6:e218002011. View Article : Google Scholar : PubMed/NCBI

47 

Pfaffl MW: A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29:e452001. View Article : Google Scholar : PubMed/NCBI

48 

Andersen CL, Jensen JL and Ørntoft TF: Normalization of real-time quantitative reverse transcription-PCR data: A model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 64:5245–5250. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Lindahl T, Satoh MS, Poirier GG and Klungland A: Post-translational modification of poly(ADP-ribose) polymerase induced by DNA strand breaks. Trends Biochem Sci. 20:405–411. 1995. View Article : Google Scholar : PubMed/NCBI

50 

Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3:11–22. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Tao LY, Liang YJ, Wang F, Chen LM, Yan YY, Dai CL and Fu LW: Cediranib (recentin, AZD2171) reverses ABCB1- and ABCC1-mediated multidrug resistance by inhibition of their transport function. Cancer Chemother Pharmacol. 64:961–969. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Tetsu O, Phuchareon J, Eisele DW, Hangauer MJ and McCormick F: AKT inactivation causes persistent drug tolerance to EGFR inhibitors. Pharmacol Res. 102:132–137. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Ford CE, Ma SS Qian, Quadir A and Ward RL: The dual role of the novel Wnt receptor tyrosine kinase, ROR2, in human carcinogenesis. Int J Cancer. 133:779–787. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Henry C, Quadir A, Hawkins NJ, Jary E, Llamosas E, Kumar D, Daniels B, Ward RL and Ford CE: Expression of the novel Wnt receptor ROR2 is increased in breast cancer and may regulate both β-catenin dependent and independent Wnt signalling. J Cancer Res Clin Oncol. 141:243–254. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Malavaki CJ, Roussidis AE, Gialeli C, Kletsas D, Tsegenidis T, Theocharis AD, Tzanakakis GN and Karamanos NK: Imatinib as a key inhibitor of the platelet-derived growth factor receptor mediated expression of cell surface heparan sulfate proteoglycans and functional properties of breast cancer cells. FEBS J. 280:2477–2489. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Thanigaimani S, Kichenadasse G and Mangoni AA: The emerging role of vascular endothelial growth factor (VEGF) in vascular homeostasis: Lessons from recent trials with anti-VEGF drugs. Curr Vasc Pharmacol. 9:358–380. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Zhou M, Liu Z, Zhao Y, Ding Y, Liu H, Xi Y, Xiong W, Li G, Lu J, Fodstad O, et al: MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression. J Biol Chem. 285:21496–21507. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Nordentoft I, Birkenkamp-Demtroder K, Agerbæk M, Theodorescu D, Ostenfeld MS, Hartmann A, Borre M, Ørntoft TF and Dyrskjøt L: miRNAs associated with chemo-sensitivity in cell lines and in advanced bladder cancer. BMC Med Genomics. 5:402012. View Article : Google Scholar : PubMed/NCBI

59 

Yin W, Wang P, Wang X, Song W, Cui X, Yu H and Zhu W: Identification of microRNAs and mRNAs associated with multidrug resistance of human laryngeal cancer Hep-2 cells. Braz J Med Biol Res. 46:546–554. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Lee K, Kunkeaw N, Jeon SH, Lee I, Johnson BH, Kang GY, Bang JY, Park HS, Leelayuwat C and Lee YS: Precursor miR-886, a novel noncoding RNA repressed in cancer, associates with PKR and modulates its activity. RNA. 17:1076–1089. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Jeon SH, Lee K, Lee KS, Kunkeaw N, Johnson BH, Holthauzen LMF, Gong B, Leelayuwat C and Lee YS: Characterization of the direct physical interaction of nc886, a cellular non-coding RNA, and PKR. FEBS Lett. 586:3477–3484. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Jeon SH, Johnson BH and Lee YS: A tumor surveillance model: A non-coding RNA senses neoplastic cells and its protein partner signals cell death. Int J Mol Sci. 13:13134–13139. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Cao J, Song Y, Bi N, Shen J, Liu W, Fan J, Sun G, Tong T, He J, Shi Y, et al: DNA methylation-mediated repression of miR-886-3p predicts poor outcome of human small cell lung cancer. Cancer Res. 73:3326–3335. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Xiong Y, Zhang L, Holloway AK, Wu X, Su L and Kebebew E: MiR-886-3p regulates cell proliferation and migration, and is dysregulated in familial non-medullary thyroid cancer. PLoS One. 6:e247172011. View Article : Google Scholar : PubMed/NCBI

65 

Noonan EJ, Place RF, Basak S, Pookot D and Li LC: miR-449a causes Rb-dependent cell cycle arrest and senescence in prostate cancer cells. Oncotarget. 1:349–358. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Miao LJ, Huang SF, Sun ZT, Gao ZY, Zhang RX, Liu Y and Wang J: MiR-449c targets c-Myc and inhibits NSCLC cell progression. FEBS Lett. 587:1359–1365. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Luo W, Huang B, Li Z, Li H, Sun L, Zhang Q, Qiu X and Wang E: MicroRNA-449a is downregulated in non-small cell lung cancer and inhibits migration and invasion by targeting c-Met. PLoS One. 8:e647592013. View Article : Google Scholar : PubMed/NCBI

68 

Fang Y, Gu X, Li Z, Xiang J and Chen Z: miR-449b inhibits the proliferation of SW1116 colon cancer stem cells through downregulation of CCND1 and E2F3 expression. Oncol Rep. 30:399–406. 2013.PubMed/NCBI

69 

Bou Kheir T, Futoma-Kazmierczak E, Jacobsen A, Krogh A, Bardram L, Hother C, Grønbæk K, Federspiel B, Lund AH and Friis-Hansen L: miR-449 inhibits cell proliferation and is down-regulated in gastric cancer. Mol Cancer. 10:292011. View Article : Google Scholar : PubMed/NCBI

70 

Feng M and Yu Q: miR-449 regulates CDK-Rb-E2F1 through an auto-regulatory feedback circuit. Cell Cycle. 9:213–214. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Yan F, Liu H, Hao J and Liu Z: Dynamical behaviors of Rb-E2F pathway including negative feedback loops involving miR449. PLoS One. 7:e439082012. View Article : Google Scholar : PubMed/NCBI

72 

Yang X, Feng M, Jiang X, Wu Z, Li Z, Aau M and Yu Q: miR-449a and miR-449b are direct transcriptional targets of E2F1 and negatively regulate pRb-E2F1 activity through a feedback loop by targeting CDK6 and CDC25A. Genes Dev. 23:2388–2393. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Zhou X, Ren Y, Moore L, Mei M, You Y, Xu P, Wang B, Wang G, Jia Z, Pu P, et al: Downregulation of miR-21 inhibits EGFR pathway and suppresses the growth of human glioblastoma cells independent of PTEN status. Lab Invest. 90:144–155. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Cho WCS, Chow ASC and Au JSK: MiR-145 inhibits cell proliferation of human lung adenocarcinoma by targeting EGFR and NUDT1. RNA Biol. 8:125–131. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Zhu H, Dougherty U, Robinson V, Mustafi R, Pekow J, Kupfer S, Li YC, Hart J, Goss K, Fichera A, et al: EGFR signals downregulate tumor suppressors miR-143 and miR-145 in Western diet-promoted murine colon cancer: Role of G1 regulators. Mol Cancer Res. 9:960–975. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 36 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bordinhão A, Evangelista A, Oliveira R, Macedo T, Silveira H, Reis R and Marques M: MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib. Oncol Rep 36: 3197-3206, 2016
APA
Bordinhão, A., Evangelista, A., Oliveira, R., Macedo, T., Silveira, H., Reis, R., & Marques, M. (2016). MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib. Oncology Reports, 36, 3197-3206. https://doi.org/10.3892/or.2016.5153
MLA
Bordinhão, A., Evangelista, A., Oliveira, R., Macedo, T., Silveira, H., Reis, R., Marques, M."MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib". Oncology Reports 36.6 (2016): 3197-3206.
Chicago
Bordinhão, A., Evangelista, A., Oliveira, R., Macedo, T., Silveira, H., Reis, R., Marques, M."MicroRNA profiling in human breast cancer cell lines exposed to the anti-neoplastic drug cediranib". Oncology Reports 36, no. 6 (2016): 3197-3206. https://doi.org/10.3892/or.2016.5153