Open Access

Investigating a multigene prognostic assay based on significant pathways for Luminal A breast cancer through gene expression profile analysis

  • Authors:
    • Haiyan Gao
    • Mei Yang
    • Xiaolan Zhang
  • View Affiliations

  • Published online on: February 2, 2018     https://doi.org/10.3892/ol.2018.7940
  • Pages: 5027-5033
  • Copyright: © Gao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate potential recurrence‑risk biomarkers based on significant pathways for Luminal A breast cancer through gene expression profile analysis. Initially, the gene expression profiles of Luminal A breast cancer patients were downloaded from The Cancer Genome Atlas database. The differentially expressed genes (DEGs) were identified using a Limma package and the hierarchical clustering analysis was conducted for the DEGs. In addition, the functional pathways were screened using Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses and rank ratio calculation. The multigene prognostic assay was exploited based on the statistically significant pathways and its prognostic function was tested using train set and verified using the gene expression data and survival data of Luminal A breast cancer patients downloaded from the Gene Expression Omnibus. A total of 300 DEGs were identified between good and poor outcome groups, including 176 upregulated genes and 124 downregulated genes. The DEGs may be used to effectively distinguish Luminal A samples with different prognoses verified by hierarchical clustering analysis. There were 9 pathways screened as significant pathways and a total of 18 DEGs involved in these 9 pathways were identified as prognostic biomarkers. According to the survival analysis and receiver operating characteristic curve, the obtained 18‑gene prognostic assay exhibited good prognostic function with high sensitivity and specificity to both the train and test samples. In conclusion the 18‑gene prognostic assay including the key genes, transcription factor 7‑like 2, anterior parietal cortex and lymphocyte enhancer factor‑1 may provide a new method for predicting outcomes and may be conducive to the promotion of precision medicine for Luminal A breast cancer.

Introduction

Breast cancer is the most commonly diagnosed cancer in women and approximately accounts for 29% of all new cancers in women, and it is the leading cause of cancer-related death in women worldwide (1). Breast cancer is considered as a heterogeneous disease but not a single disease at molecular and clinical levels (2,3). The well-known characteristics of breast cancer-associated factors include pathologic and clinical characteristics of the primary tumor, tumor histology, axillary lymph node (ALN) status, estrogen receptor (ER) content, progesterone receptor (PR) content, content, tumor HER2 status, detectable metastatic disease, patient age, patient comorbid conditions, and menopausal status (3,4). Based on the determination of ER, PR, HER2, and Ki-67, breast cancer is often to be accepted as four subtypes, namely Luminal A, Luminal B, Erb-B2 overexpression and Basal-like (also known as Triple negative breast cancer) according to St Gallen (3). Luminal A (ER positive, PR positive, HER2/neu negative) is the most common subtype, accounting for more than 50% of all breast cancer patients (5,6).

Multigene predictors have been introduced by various technologies, including immunohistochemistry (IHC), reverse transcription-quantitative polymerase chain reaction (RT-qPCR), fluorescence in situ hybridization (FISH) and genomic microarrays (7). Nowadays, some microarray-based multigene predictors have been developed as predictors of response to hormonal therapy (8,9), predictors of response to multiagent cytotoxic chemotherapy (1013) and independent prognostic biomarkers (1416).

Studies have shown that the recurrence score based on a 21-gene assay is a recurrence predictor for breast cancer patients receiving adjuvant endocrine therapy (1719). Recurrence score is an independent predict factor for the response to adjuvant chemotherapy (20,21). Patients with high scores could benefit from adjuvant treatments, whereas those with low scores could not regardless of the pathologic and clinical characteristics. In ATAC trial (22), the risk of recurrence score obtained using a 50-gene assay was seen to have an obvious relationship with the 10 years distant recurrence risk in postmenopausal breast cancer women treated with tamoxifen or aromatase inhibitors. In addition, a first commercialized microarray-based multigene assay containing 70 genes, primarily associated with proliferation, metastasis, invasion, stromal integrity, and angiogenesis, is approved by the FDA's new in diagnostic multivariate index assay classification (7).

Though multigene predictors have been widely investigated and used for the breast cancer, there are still rare studies focusing on the prognosis of subtypes. According to retrospective analyses and authoritative guidelines, these subtypes are associated with different relapse-free survival and overall survival and the patients with different subtypes should be administrated with different systemic treatment strategies (3). In this study, we aimed to utilize microarray profiling to investigate potential biomarkers that are differentially expressed in women with Luminal A-like breast cancer based on significant pathways analysis through gene expression profiles analysis. To validate the ability of the candidate multigene assay for the prediction of clinical outcomes, the gene expression data and survival data of Luminal A breast cancer patients were downloaded from Gene Expression Omnibus for analysis.

Materials and methods

Gene expression data

The gene expression profiles of breast cancer patients were downloaded from The Cancer Genome Atlas (TCGA, https://cancergenome.nih.gov/) database with the deadline of December 27, 2016, including 20,501 genes obtained from 1,160 samples (1,041 tumor tissue samples, 112 normal tissue samples and 7 peripheral blood samples). According to the clinical information of ER, PR and HER2 information (23), 370 Luminal A breast cancer samples were screened.

Data processing and differentially expressed genes (DEGs) identifying

The expression profile data of Luminal A patients were normalized. Z-score correction method was utilized to rule out the difference at gene expression level (24). A total of 249 Luminal A samples from alive patients were assigned as good outcome group; whereas a total of 47 Luminal A samples from dead patients were assigned as poor outcome group. P<0.01 and |log2 Fold-Change (FC)| >1 were regarded as the cut-off criteria to screen out DEGs between the good and poor outcome groups using LIMMA package (25).

Hierarchical clustering analysis

Hierarchical clustering analysis was conducted for the DEGs using heatmap2 package in R language (26) and the result was visualized using the form of heatmap.

Identifying statistically significant pathways

The pathway information were download from Kyoto Encyclopedia of Genes and Genomes (KEGG) (http://www.kegg.jp/kegg/pathway.html) database on March 1, 2017. The KEGG pathway enrichment analyses were performed based on pathway feature vector calculation (27) and nearest shrunken centroids (28). Briefly, the KEGG pathway was scored using the expression values of the DEGs in all samples. The sample was projected by taking the upregulated score and downregulated score as coordinates. The accuracy of the good group and the poor group was evaluated by calculating the geometric center of the same sample and specifying the radius (27). The pathways with accuracy more than 80% in these two groups were screened. The statistically significant pathways were recognized by calculating the Ratio of rank and converting to P-value according to the random 10,000 times perturbation of the background library (train set samples) (28).

Identifying prognostic biomarkers and training

Survival analysis for Luminal A breast cancer samples in TCGA were performed using DEGs involved in the obtained significant pathways (29). The support vector machine (SVM) classification model was constructed using these DEGs. Meanwhile, the model was trained using the Luminal A samples and the receiver operating characteristic (ROC) curve was drawn.

Verification of multigene prognostic assay

The reliability and repeatability of the multigene assay were verified using the gene expression profiles of GSE2034 (https://www.ncbi.nlm.nih.gov/geo/geo2r/?acc=GSE2034) and the survival data of Luminal A breast cancer patients was downloaded from Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo) database. Besides, the SVM model was also utilized to test the multigene assay, and the accuracy of the model was analyzed using the ROC curve.

Results

DEGs for Luminal A breast cancer with good and poor outcome groups

Using LIMMA package, a total of 300 DEGs were identified between 249 samples from good outcome group and 47 samples from poor outcome group, including 176 upregulated genes and 124 downregulated genes (Fig. 1). It can be observed from the figure that the data are homogenized to eliminate the deviation and the deviation scores of most genes were concentrated in −1 to 1. The genes distributed in the two branches were the most significant DEGs.

Clustering of DEGs

The 300 DEGs identified between good and poor outcome groups were selected for hierarchical clustering analysis. As presented in Fig. 2, 68 of 74 dead samples were clustered together and only 6 samples were clustered to the alive group with a precision of 92%. Meanwhile, 248 of 249 alive samples were clustered together and only 1 sample was clustered to the dead group with a precision of 99.6%. This result indicated that the DEGs could be used to effectively distinguish Luminal A samples with different prognoses.

Statistically significant pathways

Total 9 pathways with accuracy more than 80% in the two groups were screened. The significance analysis for these 9 pathways was conducted using Nearest Shrunken Centroids and the results are shown in Table I. It was observed that all of the 9 pathways significantly distinguished cancer samples with good outcome from cancer samples with poor outcome (P<0.05).

Table I.

Nine significant pathways according to Kyoto Encyclopedia of Genes and Genomes analysis nearest shrunken centroids.

Table I.

Nine significant pathways according to Kyoto Encyclopedia of Genes and Genomes analysis nearest shrunken centroids.

PathwayInitial precisionAverage valueP-value
Colorectal cancer0.78140.57751.27E-12
Oocyte meiosis0.80500.62919.24E-09
Wnt signaling pathway0.70330.50883.38E-06
Terpenoid backbone biosynthesis0.62680.42985.80E-04
Endometrial cancer0.55820.47697.90E-04
Cell cycle0.58430.46482.81E-03
Proteasome0.54220.41781.13E-02
Basal cell carcinoma0.57150.41041.50E-02
Small cell lung cancer0.55670.39951.72E-02

[i] The average value column represents the average score of 100 iterations.

Identifying prognostic biomarkers based on the significant pathways

The DEGs involved in these 9 significant pathways were collected and a total of 18 DEGs were identified as prognostic biomarkers (Table II). Three genes [transcription factor 7-like 2 (TCF7L2), anterior parietal cortex (APC), and lymphocyte enhancer factor-1 (LEF1)], were involved in four pathways, one gene [cyclin E1 (CCNE1)] in three pathways, four [S-phase kinase-associated protein 2 (SKP2), human frizzled-7 (FZD7), polo-like kinase 1 (PLK1), and B cell lymphoma 2 (BCL2)] in two pathways and ten [proteasome activator subunit 4 (PSME4), prenyldiphosphate synthase, subunit 1 (PDSS1), promoters for human DNA-PK cs (PRKDC), TTK protein kinase (TTK), minichromosome maintenance deficient 4 (MCM4), progesterone receptor (PGR), proteasome subunit alpha 7 (PSMA7), MDM2 proto-oncogene (MDM2), laminin subunit beta 2 (LAMB2), and proteasome 26S subunit, non-ATPase 7 (PSMD7)] in one pathway. Meanwhile, the annotation results for the 18 genes based on the TCGA database are shown in Fig. 3A. Simultaneously, the heat map was shown for the changes of the 18 DEGs in TCGA breast cancer samples (Fig. 3B). It can be seen that the expression of these 18 DEGs in TCGA breast cancer samples are almost upregulated.

Table II.

DEGs identified as prognostic biomarkers based on the significant pathways.

Table II.

DEGs identified as prognostic biomarkers based on the significant pathways.

BiomarkersPathwayCounts
TCF7L2Wnt signaling pathway, colorectal cancer, endometrial cancer, basal cell carcinoma4
APCWnt signaling pathway, colorectal cancer, endometrial cancer, basal cell carcinoma4
LEF1Wnt signaling pathway, colorectal cancer, endometrial cancer, B cell carcinoma4
CCNE1Cell cycle, oocyte meiosis, small cell lung cancer3
SKP2Cell cycle, small cell lung cancer2
FZD7Wnt signaling pathway, basal cell carcinoma2
PLK1Cell cycle, oocyte meiosis2
BCL2Colorectal cancer, small cell lung cancer2
PSME4Proteasome1
PDSS1Terpenoid backbone biosynthesis1
PRKDCCell cycle1
TTKCell cycle1
MCM4Cell cycle1
PGROocyte meiosis1
PSMA7Proteasome1
MDM2Cell cycle1
LAMB2Small cell lung cancer1
PSMD7Proteasome1

[i] Counts stand for the number of significant pathways. TCF7L2, transcription factor 7-like 2; APC, anterior parietal cortex; LEF1, lymphocyte enhancer factor-1; CCNE1, cyclin E1; SKP2, S-phase kinase-associated protein 2; FZD7, human frizzled-7; PLK1, polo-like kinase 1; BCL2, B cell lymphoma 2; PSME4, proteasome activator subunit 4; PDSS1, prenyldiphosphate synthase, subunit 1; PRKDC, promoters for human DNA-PK cs; TTK, TTK protein kinase; MCM4, minichromosome maintenance deficient 4; PGR, progesterone receptor; PSMA7, proteasome subunit α7; MDM2, MDM2 proto-oncogene; LAMB2, laminin subunit β2; PSMD7, proteasome 26S subunit, non-ATPase 7.

Survival analysis for the 18 DEGs in train set

The survival analysis was conducted for the Luminal A breast patient samples with abnormal expression of these 18 genes and samples with normal expression of these 18 genes in TCGA database. As a result, the samples with abnormal expression of these 18 genes showed a significantly lower survival rate than samples with normal expression of these 18 genes (Fig. 4A, P=0.0319).

Additionally, the average AUC (area under curve) was 0.871 for the ROC of these 18 genes calculated from the random train set in TCGA database. The sensitivity was 0.913 and the specificity was 0.88 (Fig. 4B).

The verification of multigene prognostic assay in test set

The multigene assay was verified using the Luminal A breast patient samples with abnormal expression of these 18 genes and samples with normal expression of these 18 genes obtained from GSE2034 database. As a result, the samples with abnormal expression of these 18 genes showed a significantly lower survival rate than samples with normal expression of these 18 genes (Fig. 5A, P=0.0279). According to the ROC for the test set, the average AUC was 0.793 with sensitivity of 0.832 and specificity of 0.779 (Fig. 5B).

Discussion

With the promotion and put forward of precision medical, studies focusing on special subtypes of breast cancer are particularly meaningful. Regardless of the development of the multigene prognostic assay for breast cancer, our study still has a critical necessary for the prognostic study of breast cancer by focusing on the Luminal A subtype. According to our results, a total of 300 DEGs were identified between good prognosis group and poor prognosis group, including 176 upregulated genes and 124 downregulated genes. Based on the hierarchical clustering analysis, these DEGs could clearly distinguish the samples of the two groups. Meanwhile, the 18 genes predictors were involved in 9 significant pathways, including cancer-related pathways (colorectal cancer, endometrial cancer, basal cell carcinoma and small cell lung cancer), oocyte meiosis, Wnt signaling pathway, Terpenoid backbone biosynthesis, cell cycle and proteasome were selected. According to the survival analysis and ROC curve, the obtained 18-gene prognostic assay exhibited a good prognostic function with high sensitivity and specificity for the train set samples and verification set samples.

TCF7L2 was identified as a key gene in the multigene prognostic assay and it was involved in four significant pathways, namely Wnt signaling pathway, colorectal cancer, endometrial cancer and basal cell carcinoma, based on the pathway analysis. TCF7L2, located on chromosome 10q25.2, plays a critical role in cancer cell growth, apoptosis, invasion and metastasis by regulating Wnt signalling (30,31). The regulation roles of TCF7L2 gene and related Wnt signaling pathway in breast cancer and its special subtypes have been widely confirmed (3234). Several studies have exploited the association between the gene polymorphisms of TCF7L2 and the risk of breast cancer. Naidu et al (34) and Burwinkel et al (35) reported that TCF7L2 variants induced an increased breast cancer risk, and might be a potential candidate for the susceptibility of breast cancer. Additionally, the TCF7L2 protein is involved in the homeostasis of blood glucose and the gene polymorphisms of TCF7L2 are identified to increase the risk of type 2 diabetes (36). Diabetes have been reported to be associated with the increased risk of breast cancer and the similar results were seen in Luminal A and B subtypes (37). Consistent with these previous studies, TCF7L2 gene was screened as a key DEG between patients with good outcomes and poor outcomes in Luminal A breast cancer.

In addition to TCF7L2, APC and LEF1 are also involved in the above mentioned four significant pathways. The association between APC and the prognosis of breast cancer has also been reported. In a study conducted by Müller et al, the methylated APC DNA indicated the worst prognosis in breast cancer samples from the train set and the independent test set (P<0.001) and it was considered as an potentially independent prognostic factor for breast cancer with poor outcomes (38). The prognostic importance of APC was also been confirmed by a study which discovered that the deletion of APC was associated with a poor overall survival of breast cancer patients (39). According to the hierarchical analysis, the alterations of APC were significantly higher in ER-/PR-breast cancer compared with ER+/PR+ breast cancer samples (39). In general, patients with ER-/PR-have a worse outcome than patients with ER+/PR+. Thus, it is reasonable to speculate that the abnormal expression of APC might be associated poor outcome in Luminal A (ER+/PR+) breast cancer. LEF1 has been widely reported to promote cancer cell metastasis, mediate chemotactic invasion, and is associated with cancer progression (40). The LEF1 overexpression has been identified as a prognostic factor for poor outcome and increased risk of liver metastasis in primary colorectal cancer (41). Delaunay et al reported that the depletion of LEF1 strongly decreased the chemotactic potential of breast cancer cells and the expression level of LEF1 was associated with the risk of developing metastasis in breast cancer patients (42). As expected, the expression of LEF1 was significantly different between good and poor outcome groups and it was screened as a key DEG according to our analysis.

The 18-gene prognostic assay including the three key genes, TCF7L2, APC and LEF1, was also demonstrated with an accurate ability to distinguish good outcomes and poor outcomes in Luminal A breast cancer. To meet the background of the precision treatment of our study would enrich the research field of specific multi-gene prognosis for breast cancer subtypes. Further study with large samples should be conducted to verify the prognostic value of this 18-gene prognostic assay and prospective study is also needed.

By conducting survival analysis, the 18-multigene assay showed effective distinguish effect on patients with different prognosis status in the low risk and high risk groups. However, the prognosis in these two groups was extraordinarily poor. The 18-gene prognostic assay should be verified in more Luminal A breast cancer samples with more typical prognosis status in further experiment.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Aure MR, Vitelli V, Jernström S, Kumar S, Krohn M, Due EU, Haukaas TH, Leivonen SK, Vollan HK, Lüders T, et al: Integrative clustering reveals a novel split in the Luminal A subtype of breast cancer with impact on outcome. Breast Cancer Res. 19:442017. View Article : Google Scholar : PubMed/NCBI

3 

Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B and Senn HJ Panel members: 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. 24:2206–2223. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Jagsi R, Raad RA, Goldberg S, Sullivan T, Michaelson J, Powell SN and Taghian AG: Locoregional recurrence rates and prognostic factors for failure in node-negative patients treated with mastectomy: Implications for postmastectomy radiation. Int J Radiat Oncol Biol Phys. 62:1035–1039. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, Karaca G, Troester MA, Tse CK, Edmiston S, et al: Race, breast cancer subtypes, and survival in the Carolina breast cancer study. JAMA. 295:2492–2502. 2006. View Article : Google Scholar : PubMed/NCBI

6 

McDermott AM, Miller N, Wall D, Martyn LM, Ball G, Sweeney KJ and Kerin MJ: Identification and validation of oncologic miRNA biomarkers for Luminal A-like breast cancer. PLoS One. 9:e870322014. View Article : Google Scholar : PubMed/NCBI

7 

Ross JS, Hatzis C, Symmans WF, Pusztai L and Hortobágyi GN: Commercialized multigene predictors of clinical outcome for breast cancer. Oncologist. 13:477–493. 2008. View Article : Google Scholar : PubMed/NCBI

8 

van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 415:530–536. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Ma XJ, Wang Z, Ryan PD, Isakoff SJ, Barmettler A, Fuller A, Muir B, Mohapatra G, Salunga R, Tuggle JT, et al: A two-gene expression ratio predicts clinical outcome in breast cancer patients treated with tamoxifen. Cancer Cell. 5:607–616. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Chang JC, Wooten EC, Tsimelzon A, Hilsenbeck SG, Gutierrez MC, Elledge R, Mohsin S, Osborne CK, Chamness GC, Allred DC and O'Connell P: Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet. 362:362–369. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Chang JC, Wooten EC, Tsimelzon A, Hilsenbeck SG, Gutierrez MC, Tham YL, Kalidas M, Elledge R, Mohsin S, Osborne CK, et al: Patterns of resistance and incomplete response to docetaxel by gene expression profiling in breast cancer patients. J Clin Oncol. 23:1169–1177. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Cleator S, Tsimelzon A, Ashworth A, Dowsett M, Dexter T, Powles T, Hilsenbeck S, Wong H, Osborne CK, O'Connell P and Chang JC: Gene expression patterns for doxorubicin (Adriamycin) and cyclophosphamide (cytoxan) (AC) response and resistance. Breast Cancer Res Treat. 95:229–233. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Peintinger F, Anderson K, Mazouni C, Kuerer HM, Hatzis C, Lin F, Hortobagyi GN, Symmans WF and Pusztai L: Thirty-gene pharmacogenomic test correlates with residual cancer burden after preoperative chemotherapy for breast cancer. Clin Cancer Res. 13:4078–4082. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Liu R, Wang X, Chen GY, Dalerba P, Gurney A, Hoey T, Sherlock G, Lewicki J, Shedden K and Clarke MF: The prognostic role of a gene signature from tumorigenic breast-cancer cells. N Engl J Med. 356:217–226. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, d'Assignies MS, et al: Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 13:3207–3214. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F, Talantov D, Timmermans M, Meijer-van Gelder ME, Yu J, et al: Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 365:671–679. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T, et al: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 351:2817–2826. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Dowsett M, Cuzick J, Wale C, Forbes J, Mallon EA, Salter J, Quinn E, Dunbier A, Baum M, Buzdar A, et al: Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: A TransATAC study. J Clin Oncol. 28:1829–1834. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Mamounas EP, Tang G, Fisher B, Paik S, Shak S, Costantino JP, Watson D, Geyer CE Jr, Wickerham DL and Wolmark N: Association between the 21-gene recurrence score assay and risk of locoregional recurrence in node-negative, estrogen receptor-positive breast cancer: Results from NSABP B-14 and NSABP B-20. J Clin Oncol. 28:1677–1683. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Tang G, Shak S, Paik S, Anderson SJ, Costantino JP, Geyer CE Jr, Mamounas EP, Wickerham DL and Wolmark N: Comparison of the prognostic and predictive utilities of the 21-gene recurrence score assay and Adjuvant! for women with node-negative, ER-positive breast cancer: Results from NSABP B-14 and NSABP B-20. Breast Cancer Res Treat. 127:133–142. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Albain KS, Barlow WE, Shak S, Hortobagyi GN, Livingston RB, Yeh IT, Ravdin P, Bugarini R, Baehner FL, Davidson NE, et al: Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: A retrospective analysis of a randomised trial. Lancet Oncol. 11:55–65. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Dowsett M, Sestak I, Lopez-Knowles E, Sidhu K, Dunbier AK, Cowens JW, Ferree S, Storhoff J, Schaper C and Cuzick J: Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy. J Clin Oncol. 31:2783–2790. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Zhu X, Ying J, Wang F, Wang J and Yang H: Estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 status in invasive breast cancer: A 3,198 cases study at National cancer center, China. Breast Cancer Res Treat. 147:551–555. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Thomas JG, Olson JM, Tapscott SJ and Zhao LP: An efficient and robust statistical modeling approach to discover differentially expressed genes using genomic expression profiles. Genome Res. 11:1227–1236. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Y, Xie J, Yang J, Fennell A, Zhang C and Ma Q: QUBIC: A bioconductor package for qualitative biclustering analysis of gene co-expression data. Bioinformatics. 33:450–452. 2017.PubMed/NCBI

27 

Chen X, Liu L, Wang Y, Liu B, Zeng D, Jin Q, Li M, Zhang D, Liu Q and Xie H: Identification of breast cancer recurrence risk factors based on functional pathways in tumor and normal tissues. Oncotarget. 8:20679–20694. 2017.PubMed/NCBI

28 

Choi BY, Bair E and Lee JW: Nearest shrunken centroids via alternative genewise shrinkages. PLoS One. 12:e01710682017. View Article : Google Scholar : PubMed/NCBI

29 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

30 

Ravindranath A, O'Connell A, Johnston PG and El-Tanani MK: The role of LEF/TCF factors in neoplastic transformation. Curr Mol Med. 8:38–50. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Reya T and Clevers H: Wnt signalling in stem cells and cancer. Nature. 434:843–850. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Dey N, Barwick BG, Moreno CS, Ordanic-Kodani M, Chen Z, Oprea-Ilies G, Tang W, Catzavelos C, Kerstann KF, Sledge GW Jr, et al: Wnt signaling in triple negative breast cancer is associated with metastasis. BMC Cancer. 13:5372013. View Article : Google Scholar : PubMed/NCBI

33 

Vijaya Kumar A, Salem Gassar E, Spillmann D, Stock C, Sen YP, Zhang T, Van Kuppevelt TH, Hülsewig C, Koszlowski EO, Pavao MS, et al: HS3ST2 modulates breast cancer cell invasiveness via MAP kinase- and Tcf4 (Tcf7l2)-dependent regulation of protease and cadherin expression. Int J Cancer. 135:2579–2592. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Naidu R, Yip CH and Taib NA: Genetic variations in transcription factor 7-like 2 (TCF7L2) gene: Association of TCF7L2 rs12255372(G/T) or rs7903146(C/T) with breast cancer risk and clinico-pathological parameters. Med Oncol. 29:411–417. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Burwinkel B, Shanmugam KS, Hemminki K, Meindl A, Schmutzler RK, Sutter C, Wappenschmidt B, Kiechle M, Bartram CR and Frank B: Transcription factor 7-like 2 (TCF7L2) variant is associated with familial breast cancer risk: A case-control study. BMC Cancer. 6:2682006. View Article : Google Scholar : PubMed/NCBI

36 

Bodhini D, Radha V, Dhar M, Narayani N and Mohan V: The rs12255372(G/T) and rs7903146(C/T) polymorphisms of the TCF7L2 gene are associated with type 2 diabetes mellitus in Asian Indians. Metabolism. 56:1174–1178. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Crispo A, Augustin LS, Grimaldi M, Nocerino F, Giudice A, Cavalcanti E, Di Bonito M, Botti G, De Laurentiis M, Rinaldo M, et al: Risk differences between prediabetes and diabetes according to breast cancer molecular subtypes. J Cell Physiol. 232:1144–1150. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Müller HM, Widschwendter A, Fiegl H, Ivarsson L, Goebel G, Perkmann E, Marth C and Widschwendter M: DNA methylation in serum of breast cancer patients: An independent prognostic marker. Cancer Res. 63:7641–7645. 2003.PubMed/NCBI

39 

Mukherjee N, Bhattacharya N, Alam N, Roy A, Roychoudhury S and Panda CK: Subtype-specific alterations of the Wnt signaling pathway in breast cancer: Clinical and prognostic significance. Cancer Sci. 103:210–220. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Wang WJ, Yao Y, Jiang LL, Hu TH, Ma JQ, Liao ZJ, Yao JT, Li DF, Wang SH and Nan KJ: Knockdown of lymphoid enhancer factor 1 inhibits colon cancer progression in vitro and in vivo. PLoS One. 8:e765962013. View Article : Google Scholar : PubMed/NCBI

41 

Lin AY, Chua MS, Choi YL, Yeh W, Kim YH, Azzi R, Adams GA, Sainani K, van de Rijn M, So SK and Pollack JR: Comparative profiling of primary colorectal carcinomas and liver metastases identifies LEF1 as a prognostic biomarker. PLoS One. 6:e166362011. View Article : Google Scholar : PubMed/NCBI

42 

Delaunay S, Rapino F, Tharun L, Zhou Z, Heukamp L, Termathe M, Shostak K, Klevernic I, Florin A, Desmecht H, et al: Elp3 links tRNA modification to IRES-dependent translation of LEF1 to sustain metastasis in breast cancer. J Exp Med. 213:2503–2523. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao H, Yang M and Zhang X: Investigating a multigene prognostic assay based on significant pathways for Luminal A breast cancer through gene expression profile analysis. Oncol Lett 15: 5027-5033, 2018
APA
Gao, H., Yang, M., & Zhang, X. (2018). Investigating a multigene prognostic assay based on significant pathways for Luminal A breast cancer through gene expression profile analysis. Oncology Letters, 15, 5027-5033. https://doi.org/10.3892/ol.2018.7940
MLA
Gao, H., Yang, M., Zhang, X."Investigating a multigene prognostic assay based on significant pathways for Luminal A breast cancer through gene expression profile analysis". Oncology Letters 15.4 (2018): 5027-5033.
Chicago
Gao, H., Yang, M., Zhang, X."Investigating a multigene prognostic assay based on significant pathways for Luminal A breast cancer through gene expression profile analysis". Oncology Letters 15, no. 4 (2018): 5027-5033. https://doi.org/10.3892/ol.2018.7940