Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

High ECT2 expression is an independent prognostic factor for poor overall survival and recurrence-free survival in non-small cell lung adenocarcinoma

  • Shijie Zhou,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Software, Validation, Writing – original draft, Writing – review & editing

    Affiliations Research Center for Public Health and Preventive Medicine, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, China, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China

  • Ping Wang,

    Roles Methodology, Supervision, Visualization, Writing – review & editing

    Affiliations Research Center for Public Health and Preventive Medicine, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, China, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China

  • Xiaolan Su,

    Roles Investigation, Resources, Supervision, Validation

    Affiliation Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China

  • Jingxia Chen,

    Roles Data curation, Investigation, Methodology, Project administration, Software, Supervision, Writing – review & editing

    Affiliation Department of Emergency Medicine, the Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China

  • Hongfen Chen,

    Roles Data curation, Formal analysis, Investigation, Methodology, Software, Validation, Visualization, Writing – review & editing

    Affiliation Department of Emergency Medicine, the Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China

  • Hanbing Yang,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Project administration, Validation, Writing – review & editing

    Affiliation Department of Emergency Medicine, the Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China

  • Aiping Fang,

    Roles Data curation, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Research Center for Public Health and Preventive Medicine, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, China, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China

  • Linshen Xie,

    Roles Methodology, Validation

    Affiliation Research Center for Public Health and Preventive Medicine, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, China

  • Yuqin Yao ,

    Roles Funding acquisition, Methodology, Supervision, Validation, Writing – review & editing

    yuqin_yao@scu.edu.cn

    Affiliation Research Center for Public Health and Preventive Medicine, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, China

  • Jinliang Yang

    Roles Funding acquisition, Resources, Supervision

    Affiliation Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China

Correction

19 Apr 2018: Zhou S, Wang P, Su X, Chen J, Chen H, et al. (2018) Correction: High ECT2 expression is an independent prognostic factor for poor overall survival and recurrence-free survival in non-small cell lung adenocarcinoma. PLOS ONE 13(4): e0196354. https://doi.org/10.1371/journal.pone.0196354 View correction

Abstract

Different subtypes of non-small cell lung cancer (NSCLC) have distinct sites of origin, histologies, genetic and epigenetic changes. In this study, we explored the mechanisms of ECT2 dysregulation and compared its prognostic value in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). In addition, we also investigated the enrichment of ECT2 co-expressed genes in KEGG pathways in LUAD and LUSC. Bioinformatic analysis was performed based on data from the Cancer Genome Atlas (TCGA)-LUAD and TCGA-LUSC. Results showed that ECT2 expression was significantly upregulated in both LUAD and LUSC compared with normal lung tissues. ECT2 expression was considerably higher in LUSC than in LUAD. The level of ECT2 DNA methylation was significantly lower in LUSC than in LUAD. ECT2 mutation was observed in 5% of LUAD and in 51% of LUSC cases. Amplification was the predominant alteration. LUAD patients with ECT2 amplification had significantly worse disease-free survival (p = 0.022). High ECT2 expression was associated with unfavorable overall survival (OS) (p<0.0001) and recurrence-free survival (RFS) (p = 0.001) in LUAD patients. Nevertheless, these associations were not observed in patients with LUSC. The following univariate and multivariate analysis showed that the high ECT2 expression was an independent prognostic factor for poor OS (HR: 2.039, 95%CI: 1.457–2.852, p<0.001) and RFS (HR: 1.715, 95%CI: 1.210–2.432, p = 0.002) in LUAD patients, but not in LUSC patients. Among 518 genes co-expressed with ECT2 in LUAD and 386 genes co-expressed with ECT2 in LUSC, there were only 98 genes in the overlapping cluster. Some of the genes related KEGG pathways in LUAD were not observed in LUSC. These differences might help to explain the different prognostic value of ECT2 in LUAD and LUSC, which are also worthy of further studies.

Introduction

Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer death in the world [1]. NSCLC accounts for about 80% of all lung cancer cases and can be divided into three subtypes, including lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC) and large cell carcinoma (LCLC) [2]. Different subtypes have distinct sites of origin, histologies, genetic and epigenetic changes [3, 4]. These differences are closely related to their unique responses to therapy [5, 6]. Therefore, it is meaningful to investigate the difference in their molecular mechanisms.

Epithelial cell transforming sequence 2 (ECT2) is a guanine nucleotide exchange factor encoded by ECT2 gene in human [7]. In non-transformed cells, ECT2 is involved in the regulation of cytokinesis via catalyzing guanine nucleotide exchange on the small GTPases, RhoA, Rac1, and Cdc42 [7]. ECT2 is frequently upregulated in human cancers and acts as an oncogene [8, 9]. In the transformed growth of ovarian and lung cancer cells, ECT2 has distinct regulative effects from its role in cytokinesis [911]. Nuclear ECT2 can activate Rac1 in the cancer cells and recruit Rac effectors to the nucleus, which is required for tumor initiation and transformation [9, 11]. Knockdown of ECT2 can inhibit Rac1 activity and block transformed growth, invasion and tumorigenicity of LUAD cells [9, 12]. One recent study found that ECT2 upregulation was associated with worse disease-free survival and overall survival (OS) of patients with LUAD [13].

ECT2 maps to 3q26.31 in the human genome. In fact, broad 3q chromosome amplification is the most common chromosomal aberration found in LUSC [14, 15]. In this study, via bioinformatic analysis, we explored the mechanisms of ECT2 dysregulation in NSCLC and compared its prognostic value in LUAD and LUSC. In addition, we also investigated the enrichment of ECT2 co-expressed genes in KEGG pathways in LUAD and LUSC.

Materials and methods

Bioinformatic analysis using FireBrowse

ECT2 expression in some solid tumors and in corresponding normal tissues was analyzed by using data from the Cancer Genome Atlas (TCGA). Data analysis was performed by using FireBrowse (http://firebrowse.org/), which provides access to analyze data generated by TCGA.

Bioinformatic analysis using UCSC Xena browser

The level 3 data of patients with primary NSCLC in TCGA-NSCLC were obtained by using the UCSC Xena browser (https://xenabrowser.net/) [16]. ECT2 mRNA expression, exon expression and DNA methylation in patients with primary LUAD or LUSC were also examined using data in TCGA-LUAD and TCGA-LUSC, by UCSC Xena browser. Kaplan-Meier curves of OS and recurrence-free survival (RFS) after initial therapy were generated by GraphPad Prism v6.0.

Bioinformatic analysis using cBioPortal for Cancer Genomics and ClueGo

ECT2 genetic alteration in TCGA-LUAD and in TCGA-LUSC was examined by using cBioPortal for Cancer Genomics (http://www.cbioportal.org/) [17, 18]. The association between ECT2 DNA mutation and disease-free survival in LUAD and LUSC patients was assessed by generating Kaplan-Meier survival curves. The genes co-expressed with ECT2 in LUAD and LUSC (|Pearson’s r| ≥ 0.4 and |Spearman’s r| ≥ 0.4) were identified. Then, the genes were loaded into ClueGo in Cytoscape [19] for analysis of KEGG pathways. Only pathways with p-value ≤ 0.05 were included.

Statistical analysis

Statistical analysis was performed by using SPSS 19.0 (SPSS Inc., Chicago, IL, USA). The association between ECT2 RNA expression and the clinicopathological features was evaluated using χ2 tests. Receiver operating characteristic (ROC) curves for death and recurrence detection were constructed and the optimal cut-off value of ECT2 expression was determined based on Youden index. Log-rank test was performed to assess the difference between the survival curves. Prognostic values were analyzed by univariate and multivariate Cox regression models. Welch’s t-test was conducted to compare ECT2 RNA expression between LUAD and LUSC groups. p < 0.05 was considered to be statistically significant.

Results

ECT2 was significantly upregulated in both LUAD and LUSC compared with normal lung tissues

By data mining using FireBrowse, we characterized ECT2 mRNA expression in several types of solid tumors, including LUAD and LUSC. Results indicated that ECT2 expression was approximately 4-fold higher in LUAD tissues than in normal lung tissues, while was about 9-fold higher in LUSC tissues than in normal lung tissues (Fig 1). To further compare ECT2 expression in LUAD and LUSC, ECT2 mRNA RNAseq and exon RNAseq data in TCGA-LUAD and TCGA-LUSC were extracted for analysis. Heatmap and following comparison showed that ECT2 expression was significantly higher in LUSC than in LUAD tissues (Fig 2A and 2B).

thumbnail
Fig 1. ECT2 mRNA expression in different types of solid tumors and in corresponding normal tissues.

https://doi.org/10.1371/journal.pone.0187356.g001

thumbnail
Fig 2. ECT2 expression in LUSC and in LUAD.

A. Heatmap of ECT2 mRNA and exon expression in patients with primary LUSC or LUAD. Data were obtained from TCGA-LUSC and TCGA-LUAD. B. Box plots of ECT2 expression in LUSC and in LUAD tissues. The analysis was performed using UCSC Xena Browser.

https://doi.org/10.1371/journal.pone.0187356.g002

LUSC had a lower level of ECT2 DNA methylation and a higher level of ECT2 DNA amplification than LUAD

Then, we tried to investigate the underlying mechanisms of dysregulated ECT2 expression in LUSC and LUAD. By comparing ECT2 expression and its DNA methylation, we observed that the level of ECT2 DNA methylation was significantly lower in LUSC cases than in LUAD cases (Fig 3A). Some CpG loci were hypermethylated in LUAD, but not in LUSC (Fig 3A). Then, we examined copy number alterations (CNA) in TCGA-LUAD and TCGA-LUSC. ECT2 mutation was observed in 5% of LUAD and in 51% of LUSC cases (Fig 3B). Amplification was the predominant type of alteration and was associated with increased ECT2 mRNA expression in both LUAD and LUSC (Fig 3C and 3D).

thumbnail
Fig 3. ECT2 DNA methylation and copy number alteration (CNA) in LUSC and LUAD.

A. Heatmap of ECT2 mRNA expression, exon expression and DNA methylation in patients with primary LUSC or LUAD. B. Genetic alteration of ECT2 in 230 cases of LUAD and 177 cases of LUSC. C-D. Box plots of ECT2 expression in LUAD (C) and in LUSC (D) tissues with indicating genetic status. Data were obtained from TCGA-LUSC and TCGA-LUAD. The analysis was performed using UCSC Xena Browser and cBioPortal for Cancer Genomics.

https://doi.org/10.1371/journal.pone.0187356.g003

ECT2 DNA mutation was associated with worse disease-free survival in LUAD, but not in LUSC patients

Then, we studied the association between ECT2 DNA mutation and survival in LUAD and LUSC patients. Survival curves indicated that LUAD patients with ECT2 amplification had significantly worse disease-free survival (p = 0.023, Fig 4A). In comparison, although ECT2 amplification was common in LUSC patients, there was no significant association between ECT2 amplification and disease-free survival (Fig 4B).

thumbnail
Fig 4.

The association between ECT2 DNA mutation and disease-free survival in LUAD (A) and LUSC (B) patients.

https://doi.org/10.1371/journal.pone.0187356.g004

High ECT2 expression was an independent prognostic factor for poor OS and RFS in LUAD, but not in LUSC patients

The associations between ECT2 expression and the demographic and clinicopathological parameters in patients with primary LUAD and LUSC were summarized in Tables 1 and 2. In patients with LUAD, the high ECT2 expression group had significantly lower proportions of female (156/318, 49.1% vs. 115/184, 62.5%, p = 0.0036) and lifelong non-smoker (37/311, 11.9% vs. 35/177, 19.8%, p = 0.018) than the low ECT2 expression group (Table 1). Besides, the high ECT2 expression group also had a significantly higher ratio of death (136/318, 42.8% vs. 47/184, 25.5%, p<0.0001) compared with the low ECT2 expression group (Table 1). In contrast, these associations were not observed in LUSC patients (Table 2). In LUAD, ECT2 expression gradually increased with the increase of pathological stages (Fig 5A). In comparison, this trend was not observed in LUSC (Fig 5B). High ECT2 expression was associated with significantly worse OS (p<0.0001) and RFS (p = 0.001) in patients with LUAD (Fig 5C and 5D). Nevertheless, no significant association was observed in patients with LUSC (Fig 5E and 5F). By performing univariate analysis, we found that advanced stage (III/IV) and high ECT2 expression were associated with significantly shorter OS and RFS in LUAD patients (Table 3). Following multivariate analysis confirmed that the high ECT2 expression was an independent prognostic factor for poor OS (HR: 2.039, 95%CI: 1.457–2.852, p<0.001) and RFS (HR: 1.715, 95%CI: 1.210–2.432, p = 0.002) in LUAD patients (Table 3). In comparison, ECT2 had no prognostic value in LUSC patients (Table 4).

thumbnail
Fig 5. The association between ECT2 expression and survival in LUAD and LUSC patients.

A-B. ECT2 expression in different pathological stages of LUAD (A) and LUSC (B). C-F. The association between ECT2 expression and OS (C and E) or RFS (D and F) in LUAD (C-D) and LUSC (E-F) patients.

https://doi.org/10.1371/journal.pone.0187356.g005

thumbnail
Table 1. The association between ECT2 expression and the demographic and clinicopathological parameters of patients with primary LUAD in TCGA.

https://doi.org/10.1371/journal.pone.0187356.t001

thumbnail
Table 2. The association between ECT2 expression and the demographic and clinicopathological parameters of patients with primary LUSC in TCGA.

https://doi.org/10.1371/journal.pone.0187356.t002

thumbnail
Table 3. Univariate and multivariate analyses of OS/RFS in patients with primary LUAD.

https://doi.org/10.1371/journal.pone.0187356.t003

thumbnail
Table 4. Univariate analysis of OS/RFS in patients with primary LUSC.

https://doi.org/10.1371/journal.pone.0187356.t004

ECT2 was involved in different signaling pathways in LUAD and LUSC

By data mining using cBioPortal for Cancer Genomics, we identified the genes co-expressed with ECT2 in LUAD and LUSC (|Pearson’s r| ≥ 0.4 and |Spearman’s r| ≥ 0.4) (S1 Table). Results indicated that 518 genes were co-expressed with ECT2 in LUAD and 386 genes were co-expressed with ECT2 in LUSC (Fig 6A and S1 Table). However, only 98 genes were in the overlapping cluster (Fig 6A). To further investigate the possible signaling pathways in which ECT2 might be involved in, ECT2 co-expressed genes in LUAD and LUSC were subjected to KEGG pathway analysis respectively. In LUAD, the genes were enriched in Pyrimidine metabolism, Ribosome biogenesis in eukaryotes, p53 signaling pathway, HTLV-I infection, RNA transport, Base excision repair, Homologous recombination, Fanconi anemia pathway, Cell cycle, Oocyte meiosis, Progesterone-mediated oocyte maturation, DNA replication, Nucleotide excision repair and Mismatch repair (Fig 6B and S2 Table). In comparison, ECT2 co-expressed genes in LUSC were enriched in Glycerophospholipid metabolism, Cell cycle, p53 signaling pathway, DNA replication, Mismatch repair, Homologous recombination and Fanconi anemia pathway (Fig 6C and S3 Table).

thumbnail
Fig 6. KEGG pathway analysis of the genes co-expressed with ECT2 in LUAD and LUSC.

A. The genes co-expressed with ECT2 in LUAD and LUSC. B-C. KEGG pathway analysis of ECT2 co-expressed genes in TCGA-LUAD (B) and in TCGA-LUSC (C).

https://doi.org/10.1371/journal.pone.0187356.g006

Discussion

Aberrant ECT2 expression was observed in both LUAD and LUSC [13, 20, 21]. In the current study, via characterizing ECT2 expression based on data in TCGA-LUAD and TCGA-LUSC, we also confirmed significantly upregulated ECT2 expression in LUAD and LUSC compared with normal lung tissues. In addition, we found that ECT2 expression was considerably higher in LUSC than in LUAD tissues. Previous studies indicated that copy number gains (CNGs) are one of the most common mechanisms of dysregulated genes at chromosome 3q26 [15, 21]. In this study, we found that amplification is common in LUSC, but not in LUAD. Approximately 50% of LUSC cases had ECT2 amplification, but this rate was only around 5% in LUAD. These findings are consistent with the prevalence of chromosome 3q26 CNGs in LUSC and the relatively rare occurrence of 3q26 CNGs in LUAD [22]. In addition, we also observed that some CpG loci of ECT2 gene had higher levels of methylation in LUAD than in LUSC, suggesting that epigenetic alteration is also an important mechanism of dysregulated ECT2 in NSCLC. These results help to explain why ECT2 expression is significantly higher in LUSC than in LUAD.

As an oncogene, ECT2 upregulation also has prognostic values in some cancers. In patients with colorectal cancer, high expression level of ECT2 was significantly associated with tumor size, serum CEA levels and TNM stage [23]. Kaplan-Meier survival analysis indicated that patients with high ECT2 expression had a remarkably shorter OS [23]. High level of ECT2 expression was also associated with poor prognosis in patients with esophageal squamous cell carcinomas [20]. One study based on patients with LUAD indicated that high ECT2 expression was associated with unfavorable disease-free survival and overall survival [13]. However, the number of patients included in this study is relatively small (N = 88) [13]. In this study, based on large datasets in TCGA, we found that although ECT2 amplification is common in LUSC, its mutation had no influence on survival outcomes. Nevertheless, although ECT2 amplification was less frequent in LUAD, its mutation was associated with significantly worse disease-free survival. By generating Kaplan-Meier curves, we further demonstrated that in patients with LUAD, high ECT2 expression was related to unfavorable OS and RFS. But no significant association was observed in patients with LUSC. In addition, our univariate and multivariate analysis showed that high ECT2 expression was an independent prognostic factor for poor OS (HR: 2.039, 95%CI: 1.457–2.852, p<0.001) and RFS (HR: 1.715, 95%CI: 1.210–2.432, p = 0.002) in LUAD patients, but not in LUSC patients. Therefore, we hypothesized that ECT2 might play different roles in LUAD and LUSC.

In LUAD, one recent study indicated that ECT2 could activate rRNA synthesis by binding the nucleolar transcription factor upstream binding factor 1 (UBF1) on rDNA promoters and recruiting Rac1 and its downstream effector nucleophosmin (NPM) to rDNA [11]. However, whether other mechanisms are involved in the oncogenic properties of ECT2 in LUAD and whether ECT2 participates in different molecular pathways in LUAD and LUSC have not been fully revealed. By comparing ECT2 co-expressed genes in LUAD and LUSC, we found a considerable variation. Among 518 genes co-expressed with ECT2 in LUAD and 386 genes co-expressed with ECT2 in LUSC, there were only 98 genes in the overlapping cluster. The following KEGG pathway analysis of the enrichment of ECT2 co-expressed genes showed that Cell cycle, p53 signaling pathway, DNA replication, Mismatch repair, Homologous recombination and Fanconi anemia pathway are common in LUSC and LUAD. In LUAD, ECT2 co-expressed genes were additionally enriched in some cancer-related pathways, such as Pyrimidine metabolism, Ribosome biogenesis in eukaryotes, RNA transport and Base excision repair. Therefore, it is meaningful to further investigate the involvement of ECT2 in these pathways in LUAD in the future.

Conclusion

Both genetic and epigenetic alterations contributed to dysregulated ECT2 in NSCLC. High ECT2 expression was an independent prognostic factor for poor OS and RFS in LUAD patients, but not in LUSC patients. Some of the genes related KEGG pathways in LUAD were not observed in LUSC. These differences might help to explain the different prognostic value of ECT2 in LUAD and LUSC, which are also worthy of further studies.

Supporting information

S1 Table. The genes co-expressed with ECT2 in LUAD and LUSC.

https://doi.org/10.1371/journal.pone.0187356.s001

(XLSX)

S2 Table. KEGG pathway analysis of ECT2 co-expressed genes in TCGA-LUAD.

https://doi.org/10.1371/journal.pone.0187356.s002

(DOCX)

S3 Table. KEGG pathway analysis of ECT2 co-expressed genes in TCGA-LUSC.

https://doi.org/10.1371/journal.pone.0187356.s003

(DOCX)

References

  1. 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA: a cancer journal for clinicians. 2015;65(1):5–29. pmid:25559415.
  2. 2. Schmid-Bindert G. Update on antiangiogenic treatment of advanced non-small cell lung cancer (NSCLC). Targeted oncology. 2013;8(1):15–26. pmid:23371029.
  3. 3. Pierceall WE, Olaussen KA, Rousseau V, Brambilla E, Sprott KM, Andre F, et al. Cisplatin benefit is predicted by immunohistochemical analysis of DNA repair proteins in squamous cell carcinoma but not adenocarcinoma: theranostic modeling by NSCLC constituent histological subclasses. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2012;23(9):2245–52. pmid:22269178.
  4. 4. Saghaeian Jazi M, Samaei NM, Ghanei M, Shadmehr MB, Mowla SJ. Overexpression of the non-coding SOX2OT variants 4 and 7 in lung tumors suggests an oncogenic role in lung cancer. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine. 2016. pmid:26846097.
  5. 5. Pasini A, Paganelli G, Tesei A, Zoli W, Giordano E, Calistri D. Specific Biomarkers Are Associated with Docetaxeland Gemcitabine-Resistant NSCLC Cell Lines. Translational oncology. 2012;5(6):461–8. pmid:23397475; PubMed Central PMCID: PMC3567725.
  6. 6. Sun R, Liu Z, Wang L, Lv W, Liu J, Ding C, et al. Overexpression of stathmin is resistant to paclitaxel treatment in patients with non-small cell lung cancer. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine. 2015;36(9):7195–204. pmid:25894372.
  7. 7. Tatsumoto T, Xie X, Blumenthal R, Okamoto I, Miki T. Human ECT2 is an exchange factor for Rho GTPases, phosphorylated in G2/M phases, and involved in cytokinesis. The Journal of cell biology. 1999;147(5):921–8. pmid:10579713; PubMed Central PMCID: PMC2169345.
  8. 8. Fields AP, Justilien V. The guanine nucleotide exchange factor (GEF) Ect2 is an oncogene in human cancer. Advances in enzyme regulation. 2010;50(1):190–200. pmid:19896966; PubMed Central PMCID: PMC2863999.
  9. 9. Justilien V, Fields AP. Ect2 links the PKCiota-Par6alpha complex to Rac1 activation and cellular transformation. Oncogene. 2009;28(41):3597–607. pmid:19617897; PubMed Central PMCID: PMC2762483.
  10. 10. Huff LP, Decristo MJ, Trembath D, Kuan PF, Yim M, Liu J, et al. The Role of Ect2 Nuclear RhoGEF Activity in Ovarian Cancer Cell Transformation. Genes & cancer. 2013;4(11–12):460–75. pmid:24386507; PubMed Central PMCID: PMC3877668.
  11. 11. Justilien V, Ali SA, Jamieson L, Yin N, Cox AD, Der CJ, et al. Ect2-Dependent rRNA Synthesis Is Required for KRAS-TRP53-Driven Lung Adenocarcinoma. Cancer cell. 2017;31(2):256–69. pmid:28110998; PubMed Central PMCID: PMC5310966.
  12. 12. Justilien V, Jameison L, Der CJ, Rossman KL, Fields AP. Oncogenic activity of Ect2 is regulated through protein kinase C iota-mediated phosphorylation. The Journal of biological chemistry. 2011;286(10):8149–57. pmid:21189248; PubMed Central PMCID: PMC3048701.
  13. 13. Murata Y, Minami Y, Iwakawa R, Yokota J, Usui S, Tsuta K, et al. ECT2 amplification and overexpression as a new prognostic biomarker for early-stage lung adenocarcinoma. Cancer science. 2014;105(4):490–7. pmid:24484057; PubMed Central PMCID: PMC4317802.
  14. 14. Mendez P, Ramirez JL. Copy number gains of FGFR1 and 3q chromosome in squamous cell carcinoma of the lung. Translational lung cancer research. 2013;2(2):101–11. pmid:25806221; PubMed Central PMCID: PMC4369856.
  15. 15. Fields AP, Justilien V, Murray NR. The chromosome 3q26 OncCassette: A multigenic driver of human cancer. Advances in biological regulation. 2016;60:47–63. pmid:26754874; PubMed Central PMCID: PMC4729592.
  16. 16. Cline MS, Craft B, Swatloski T, Goldman M, Ma S, Haussler D, et al. Exploring TCGA Pan-Cancer data at the UCSC Cancer Genomics Browser. Scientific reports. 2013;3:2652. pmid:24084870; PubMed Central PMCID: PMC3788369.
  17. 17. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Science signaling. 2013;6(269):pl1. pmid:23550210; PubMed Central PMCID: PMC4160307.
  18. 18. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer discovery. 2012;2(5):401–4. pmid:22588877; PubMed Central PMCID: PMC3956037.
  19. 19. Bindea G, Galon J, Mlecnik B. CluePedia Cytoscape plugin: pathway insights using integrated experimental and in silico data. Bioinformatics. 2013;29(5):661–3. pmid:23325622; PubMed Central PMCID: PMC3582273.
  20. 20. Hirata D, Yamabuki T, Miki D, Ito T, Tsuchiya E, Fujita M, et al. Involvement of epithelial cell transforming sequence-2 oncoantigen in lung and esophageal cancer progression. Clinical cancer research: an official journal of the American Association for Cancer Research. 2009;15(1):256–66. pmid:19118053.
  21. 21. Qian J, Zou Y, Wang J, Zhang B, Massion PP. Global gene expression profiling reveals a suppressed immune response pathway associated with 3q amplification in squamous carcinoma of the lung. Genomics data. 2015;5:272–4. pmid:26484266; PubMed Central PMCID: PMC4583673.
  22. 22. Balsara BR, Sonoda G, du Manoir S, Siegfried JM, Gabrielson E, Testa JR. Comparative genomic hybridization analysis detects frequent, often high-level, overrepresentation of DNA sequences at 3q, 5p, 7p, and 8q in human non-small cell lung carcinomas. Cancer research. 1997;57(11):2116–20. pmid:9187106.
  23. 23. Luo Y, Qin SL, Mu YF, Wang ZS, Zhong M, Bian ZQ. Elevated expression of ECT2 predicts unfavorable prognosis in patients with colorectal cancer. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2015;73:135–9. pmid:26211594.