Back to Journals » OncoTargets and Therapy » Volume 11

Identification of the predictive genes for the response of colorectal cancer patients to FOLFOX therapy

Authors Lin HJ, Qiu XK, Zhang B, Zhang JC

Received 8 March 2018

Accepted for publication 12 July 2018

Published 17 September 2018 Volume 2018:11 Pages 5943—5955

DOI https://doi.org/10.2147/OTT.S167656

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Geoffrey Pietersz



Hengjun Lin, Xueke Qiu, Bo Zhang, Jichao Zhang

Department of Tumor, Anus and Intestine, Jinhua People’s Hospital, Jinhua, Zhejiang 321000, China

Background: Colorectal cancer is a malignant tumor with high death rate. Chemotherapy, radiotherapy and surgery are the three common treatments of colorectal cancer. For early colorectal cancer patients, postoperative adjuvant chemotherapy can reduce the risk of recurrence. For advanced colorectal cancer patients, palliative chemotherapy can significantly improve the life quality of patients and prolong survival. FOLFOX is one of the mainstream chemotherapies in colorectal cancer, however, its response rate is only about 50%.
Methods: To systematically investigate why some of the colorectal cancer patients have response to FOLFOX therapy while others do not, we searched all publicly available database and combined three gene expression datasets of colorectal cancer patients with FOLFOX therapy. With advanced minimal redundancy maximal relevance and incremental feature selection method, we identified the biomarker genes.
Results: A Support Vector Machine-based classifier was constructed to predict the response of colorectal cancer patients to FOLFOX therapy. Its accuracy, sensitivity and specificity were 0.854, 0.845 and 0.863, respectively.
Conclusion: The biological analysis of representative biomarker genes suggested that apoptosis and inflammation signaling pathways were essential for the response of colorectal cancer patients to FOLFOX chemotherapy.

Keywords:
colorectal cancer, FOLFOX therapy, support vector machine, minimal redundancy maximal relevance, incremental feature selection, chemotherapy response

Introduction

Colorectal cancer is a malignant tumor that seriously endangers people’s health. In recent years, the incidence of colorectal cancer has significantly increased and has become the third most common type of cancer. In the past few decades, due to the early detection and treatment, many countries have improved the survival rate of colorectal cancer. Especially in some developed countries, the 5-year survival rate has reached more than 65%.1

Treatment options for colorectal cancer include chemotherapy, radiotherapy and surgery.2 In general, surgical removal of the affected tumor and any adjacent intestines can effectively eliminate cancer cells and reduce the risk of cancer spreading. Chemotherapy also occupies an important role in the treatment of colorectal cancer. Postoperative adjuvant chemotherapy in early colorectal cancer can reduce the risk of recurrence. For patients with advanced colorectal cancer who are inoperable, palliative chemotherapy can significantly improve the life quality of patients and prolong survival.

Generally, the combination of chemotherapeutic agent results in significantly increased response rates and improved survival.3 Current combination chemotherapy includes 5-fluorouracil (5-FU)/leucovorin with oxaliplatin (FOLFOX), 5-FU/leucovorin and irinotecan (FOLFIRI), capecitabine and oxaliplatin (CAPEOX/XELOX) and 5-FU/leucovorin/oxaliplatin and irinotecan (FOLFOXIRI).

FOLFOX chemotherapy has proven to be effective in the treatment of unresectable metastatic colorectal cancer.4 Studies have suggested that patients with stage III colorectal cancer, who receive adjuvant FOLFOX chemotherapy, experience an improved disease-free and overall survival.5 However, about half of the patients were unable to benefit from the treatment and even suffered from neurotoxicity.6

There have been several studies that are trying to predict the FOLFOX chemotherapy response.7,8 It has been reported that MTHFR germinal polymorphism is a potential strong predictor of response to FOLFOX therapy, and the response rate to FOLFOX increases continuously with the number of favorable MTHFR alleles.7 Another reported biomarker is SMURF2. It was highly expressed in non-responders for FOLFOX therapy.8

To systematically investigate the response mechanisms of FOLFOX chemotherapy in colorectal cancer patients, we collected three gene expression datasets of colorectal cancer patients with FOLFOX therapy and identified the genes that can predict responders to FOLFOX therapy for colorectal cancer using advanced machine learning methods. The biological analysis of several representative signature genes, such as MLKL, CC2D1A, LPL, PAGE4 and SLC26A9, suggested that apoptosis and inflammation signaling pathways were the essential pathways that controlled the response of colorectal cancer patients to FOLFOX chemotherapy.

Methods

The gene expression profiles of colorectal cancer patients with FOLFOX therapy

We searched Gene Expression Omnibus (GEO) database and found three datasets of colorectal cancer patients with FOLFOX therapy.

The gene expression profiles of colorectal cancer patients with FOLFOX therapy were combined from three datasets downloaded from GEO with accession number of GSE19860, GSE28702 and GSE72970. The platform of these three datasets was the same. They all used Affymetrix Human Genome U133 Plus 2.0 Array.

These three datasets were generated by different researchers from different labs. To minimize the systemic bias, the raw CEL files were downloaded and processed together using R package affyPLM and affy.9 The gene expression levels of probes were quantified with MAS5 method10 and normalized with quantile method. The probe expression levels were transformed into gene expression levels using R package gahgu133plus2cdf and gahgu133plus2.db. There were 18,733 genes with expression levels that were used as features to predict whether a colorectal cancer patient will respond to FOLFOX therapy.

In GSE72970 dataset, there were 20 colorectal cancer patients with FOLFOX response and 12 colorectal cancer patients without FOLFOX response. In GSE28702, there were 42 colorectal cancer patients with FOLFOX response and 41 colorectal cancer patients without FOLFOX response. In GSE19860, there were nine colorectal cancer patients with FOLFOX response and 20 colorectal cancer patients without FOLFOX response. Together, there were 42 colorectal cancer patients with FOLFOX response who were considered as positive samples and 41 colorectal cancer patients without FOLFOX response who were considered as negative samples. The sizes of positive and negative samples are shown in Table 1. The clinical information of the 144 colorectal cancer patients from GEO is given in Table S1.

Table 1 The sizes of positive and negative samples
Notes: aPositive samples: colorectal cancer patients with FOLFOX response. bNegative samples: colorectal cancer patients without FOLFOX response.

Rank the discriminative genes using mRMR method

The minimal redundancy maximal relevance (mRMR) method11 is widely used to select discriminative features.1217 The mRMR software downloaded from http://home.penglab.com/proj/mRMR/ was used to perform the feature ranking.

It works as follows: first, let us represent all the 18,733 genes, the selected m genes and the to-be-selected n genes using Ω, ΩS and Ωt, respectively. The relevance I of gene g from Ωt with FOLFOX response r can be measured with mutual information (I):18,19

(1)

The redundancy R of the gene g from Ω with the selected genes in ΩS are


(2)

The algorithm tries to find the best gene gj from Ωt that has maximum relevance with FOLFOX response r and minimum redundancy with the selected genes in ΩS by maximizing the function below


(3)

After N rounds of evaluation procedure, all the genes from Ωt will be ranked

(4)

The mRMR rank represents the discriminating power of the gene.

To reduce the computational time, only the top 500 mRMR genes were analyzed in the following steps.

Identify the predictive genes using incremental feature selection (IFS) method

To evaluate the prediction performance of mRMR genes, IFS method2026 was applied to select the genes with greatest prediction power. The IFS method is a wrapped feature selection method that combines the feature selection with classifier construction. We used Support Vector Machine (SVM) as the classifier. To be specific, the SVM function in R package e1071 was used to construct the classifier.

IFS is a process of iteration that adds genes one by one based on the mRMR ranking and then evaluates the classification performance of the selected genes. Each time, the top k genes from the mRMR table were selected and used to build the classifier that predicts whether a colorectal cancer patient will respond to FOLFOX therapy. The performance of each classifier was evaluated with leave-one-out cross validation (LOOCV).

The three major measurements for a classifier, sensitivity (Sn), specificity (Sp) and accuracy (ACC), were calculated.


(5)


(6)


(7)

In these equations, TP, TN, FP and FN stand for true positive samples, true negative samples, false positive samples and false negative samples, respectively.

In this study, the colorectal cancer patients with FOLFOX response and the colorectal cancer patients without FOLFOX response were considered as positive and negative samples, respectively.

After 500 rounds of IFS evaluation, an IFS curve can be plotted. The x-axis was the number of used genes, and the y-axis was the LOOCV accuracy. Based on the IFS, we can easily see how many genes should be used to classify the colorectal cancer patients with FOLFOX response and the colorectal cancer patients without FOLFOX response.

The visualization of how predictive the genes are for FOLFOX response

After we identified the predictive genes using mRMR and IFS methods, we tried to visually investigate how good they can classify the colorectal cancer patients with FOLFOX response and the colorectal cancer patients without FOLFOX response.

Principal component analysis (PCA)27 was performed to extract the first and second principal component (PC) of the selected genes. PCA is a widely used multivariate statistical method and can capture most of the gene expression variability.27 With the dimensionality reduction via PCA, the high dimension gene expression profiles can be mapped onto two dimensions of PC1 and PC2, which can explain the most variance observed in the data. Since it is unsupervised, the 2D-PCA plot will give an intuitive view of how close each sample is to each other.

Another method that we applied was two-way hierarchical clustering of both colorectal cancer patients and selected genes. From the heatmap, we can not only explore whether the colorectal cancer patients with FOLFOX response and the colorectal cancer patients without FOLFOX response were clustered into different groups but also know which genes were highly expressed or lowly expressed in the colorectal cancer patients with FOLFOX response.

Results and discussion

The top discriminative genes ranked with mRMR method

The mRMR can rank the genes based on not only their relevance with the FOLFOX responses of colorectal cancer patients but also the redundancy with each other. Therefore, the discriminative genes identified by mRMR methods will be compact, which means the highly co-expressed genes will not all be selected, only the best representative gene will be chosen. We obtained the top 500 most discriminative genes using the mRMR method. These 500 genes will be further optimized using IFS method.

The predictive genes selected based on IFS method

We used different number of top mRMR genes to construct the SVM classifier. Based on how accurate the model can classify the colorectal cancer patients into the right FOLFOX response groups, we plotted the IFS curve in which the x-axis was the number of genes and the y-axis was the LOOCV accuracy. The IFS curve is shown in Figure 1.

Figure 1 The IFS curve of how the classifiers were based on different number of gene performance.
Notes: The x-axis was the number of genes used to build the classifier and y-axis was the prediction accuracy evaluated with LOOCV. The peak of IFS curve was accuracy of 0.854 when 138 genes were used. But even when only top ten genes were used, the accuracy was over 0.8.
Abbreviations: IFS, incremental feature selection; LOOCV, leave-one-out cross validation.

As shown in Figure 1, the peak located at the position of using top 138 genes. Its accuracy was 0.854, which was the highest. We also calculated its sensitivity and specificity, which were 0.845 and 0.863, respectively. The top 138 genes are given in Table S2. The confusion matrix of actual responses and predicted responses is given in Table 2. We calculated the CIs of prediction performance using function sensSpec from R package epibasix28 and the 95% CIs for sensitivity and specificity were (76.1, 92.9) and (78.4, 94.2), respectively.

Table 2 The confusion matrix of actual responses and predicted responses based on 138 genes

Although the performance of 138 genes was best, the accuracy of the top ten genes had already been over 0.8. The sensitivity and specificity for the ten gene classifier were 0.732 and 0.890, respectively. The top ten genes are given in Table 3.

Table 3 The top ten mRMR genes

The first gene was LOC100009676, which was under-studied and did not have too much known functions.

The second gene was Lnc-ZNF461, which has been reported to be associated with non-small-cell lung cancer (NSCLC).29 It was involved in immune response and can promote NSCLC progression by interacting with SLA2, DEFB4A, LAT and LIME1.29

The third gene was MLKL, a necroptosis kinase. It was reported that MLKL was involved in immune activation in cancer cells.30 Chemotherapy kills MLKL−/− cancer cells, and due to MLKL deficiency, the dying cancer cells will not cause immune response. MLKL may function through ICD signaling pathway. A recent publication by Sun et al31 found that small-molecule analogs of SMAC mimetic in association with MLKL-pDNA and z-VAD-fmk showed antitumor effects in colorectal cancer cells in vitro via induction of RIP3-dependent necroptosis. All these findings have confirmed MLKL as a good chemotherapy response biomarker.

Another interesting gene was CC2D1A, a remarkable member of various signaling pathways, such as nuclear factor kB, PDK1/Akt, cAMP/PKA and Notch. Notch pathway is a well-studied colorectal cancer pathway.32,33 It has also been reported to be involved in the antiviral pathway by interacting with TBK-1 and IKKε and acts as a transcriptional repressor of serotonin and dopamine receptor genes.34 CC2D1A silencing can induce apoptosis and increase chemotherapy sensitivity by decreasing Akt kinase activity.35

The responders and non-responders were different on the first PC

To intuitively explore the difference of responders and non-responders, we calculated the first and second PCs of the 138 genes and plotted the PCA of responders (blue dots) and non-responders (red dots) in Figure 2. PC1 represented 8.7% variance, while PC2 represented 4.7% variance.

Figure 2 The PCA plot of responders and non-responders.
Notes: The x-axis was the first PC and y-axis was the second PC. The red dots were NR and the blue dots were R. It can be seen that most responders were in area of PC1<0, while most non-responders were in the area of PC1>0. R and NR were different on the first PC.
Abbreviations: PCA, principal component analysis; PC, principal component; NR, non-responders; R, responders.

It can be seen that most responders were in area of PC1<0, while most non-responders were in the area of PC1>0. The responders and non-responders were different on the first PC.

The highly expressed genes in FOLFOX responders and non-responders

Although the PCA plot clearly demonstrated the difference of responders and non-responders, we were interested in identifying the highly expressed genes in FOLFOX responders and non-responders, which may reveal the biological mechanisms of FOLFOX response in colorectal cancer. Therefore, we plotted the heatmap of the 138 genes in the responder and non-responder colorectal cancer patients (Figure 3).

Figure 3 The heatmap of the 138 genes in the responder and non-responder colorectal cancer patients.
Notes: Each row corresponded to the scaled gene expressed level of a gene. The warmer colors indicated higher expression level and the colder colors indicate lower expression levels. Each column corresponded to a colorectal cancer patient who may be responder (red) and non-responder (green) to FOLFOX therapy. It can be seen that the responders and non-responders were clearly clustered into two groups and correspondingly, the 138 genes were also clustered into two groups. The top cluster of genes was highly expressed in responders and the bottom cluster of genes was highly expressed in non-responders.
Abbreviations: NR, non-responders; R, responders.

It can be seen that the responders and non-responders were clearly clustered into two groups and correspondingly, the 138 genes were also clustered into two groups. The top cluster of genes was highly expressed in responders, and the bottom cluster of genes was highly expressed in non-responders.

We have listed the highly expressed genes in FOLFOX responders whose fold change was greater than 1.5 and the lowly expressed genes in FOLFOX responders whose fold change was smaller than 0.67 in Tables 4 and 5, respectively.

Table 4 The highly expressed genes in FOLFOX responders
Notes: aNR, colorectal cancer patients without FOLFOX response. bR, colorectal cancer patients with FOLFOX response. cFold change, R/NR.

Table 5 The lowly expressed genes in FOLFOX responders
Notes: aNR, colorectal cancer patients without FOLFOX response. bR, colorectal cancer patients with FOLFOX response. cFold change, R/NR.

For the highly expressed genes in FOLFOX responders, CRYBB1 was one of the highly mutated genes in microsatellite instability colorectal cancers.36

NEUROG3 played important roles in intestinal enteroendocrine cells and was repressed by the growth factor-independent one transcription factor (GFI1) that was normally expressed in Paneth and goblet cells of colon.37

LPL is a crucial enzyme for intravascular catabolism of triglyceride-rich lipoproteins. The alteration of LPL may let the cell acquire growth advantage and develop malignancy.38 The LPL gene deficiency increases cancer risk. The tumor suppressive effects of LPL have been verified in animal models; due to its roles in inflammation, it is a great general target for chemotherapy.39

CYP4F is a member of the CYP/CYP450 superfamily of enzymes. It was highly expressed in prostate cancer and RNAi experiments, which suggested that CYP4F was important for cell growth and survival.40

PAGE4 is a member of GAGE family, which is highly expressed in various tumors.4143 It has been reported that PAGE4 expression can predict liver metastasis of colorectal cancer.44

For the lowly expressed genes in FOLFOX responders, SLC26A9 has colon-specific functions, such as transport of glucose, organic acids, metal ions and mineral absorption.45 Its low expression may affect the growth of tumor cells.

The limitations and potential improvements of this study

Although this study identified candidate genes for chemotherapy response for colorectal cancer and revealed highly possible mechanism, there were several limitations:

Since this was a bioinformatics study, we did not validate our results with biological experiments. This limited the discovery of novel mechanisms. To reduce the effects of lacking experimental validation, we did thoroughgoing literature survey and proposed the possible mechanisms based on confirmed biological functions of candidate genes from published papers.

The sample size of this study was small, even though we collected all publicly available gene expression profiles from the largest gene expression database, GEO. In the next step, we will collect colorectal cancer patients with chemotherapy from our hospital and build a large independent test dataset.

The number of genes was still too large. We will try more advanced feature selection methods to further reduce the number of selected genes. The exhaust search strategies can be applied within the 138 genes to find the optimal 3–5 genes.

The clinical information should be documented carefully. Since the data we analyzed were from GEO, much clinical information of the patients was unknown. Analyzing the clinical information may provide novel insight. For example, within the 141 colorectal cancer patients, 117 samples were from primary sites and 27 samples were from metastatic lesions. But, we found that all 27 metastatic samples were predicted with the correct responses, as shown in Table S1 in which the third and sixth columns are actual responses and predicted responses, respectively. There may be two reasons of why the metastatic lesions can predict chemotherapy response: 1) the gene expressions between primary tumors and metastatic lesions have strong correlation.46,47 Staub et al reported that the primary site of metastatic cancer can be predicted based on the similarity between metastatic cancer and primary tissue.46 2) Some of the candidate genes were general tumor genes, such as PAGE4, a member of the GAGE family that is expressed in a variety of tumors.4143

Genetic variations, such as single-nucleotide polymorphisms (SNPs) and copy number variations, have been proven to be a causal factor for tumorgenesis.4852 They can be used for cancer subtyping and drug response prediction.22,48 Unfortunately, our dataset did not include genetic data. But based on central dogma and previous studies, most SNPs function through expression quantitative trait loci (eQTL).17,18,53 The gene expression data can partially represent the effects of SNPs. If possible, we will preform DNA-Seq and RNA-Seq for the same patients and investigate the eQTL regulatory network of colorectal cancer patients with chemotherapy in the future.

Conclusion

Chemotherapy is a widely used treatment for cancers but not all cancer patients have expected responses to this treatment. In this study, we analyzed the gene expression profiles of FOLFOX responders and FOLFOX non-responders of colorectal cancer patients by combing several datasets. With advanced feature selection methods, we identified the biomarkers that can accurately predict the response of colorectal cancer patient to FOLFOX treatment. The biological analysis of selected genes revealed the possible mechanism of chemotherapy in colorectal cancer.

Disclosure

The authors report no conflicts of interest in this work.


References

1.

Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014;383(9927):1490–1502.

2.

Abdolahi HM, Asiabar AS, Azami-Aghdash S, Pournaghi-Azar F, Rezapour A. Cost-effectiveness of Colorectal Cancer Screening and Treatment Methods: Mapping of Systematic Reviews. Asia Pac J Oncol Nurs. 2018;5(1):57–67.

3.

Mohelnikova-Duchonova B, Melichar B, Soucek P. FOLFOX/FOLFIRI pharmacogenetics: the call for a personalized approach in colorectal cancer therapy. World J Gastroenterol. 2014;20(30):10316–10330.

4.

Suh KW, Kim JH, Kim DY, Kim YB, Lee C, Choi S. Which gene is a dominant predictor of response during FOLFOX chemotherapy for the treatment of metastatic colorectal cancer, the MTHFR or XRCC1 gene? Ann Surg Oncol. 2006;13(11):1379–1385.

5.

Kumar A, Peixoto RD, Kennecke HF, et al. Effect of Adjuvant FOLFOX Chemotherapy Duration on Outcomes of Patients With Stage III Colon Cancer. Clin Colorectal Cancer. 2015;14(4):262.e1–268.e1.

6.

Watanabe T, Kobunai T, Yamamoto Y, et al. Gene expression signature and response to the use of leucovorin, fluorouracil and oxaliplatin in colorectal cancer patients. Clin Transl Oncol. 2011;13(6):419–425.

7.

Etienne-Grimaldi MC, Milano G, Maindrault-Goebel F, et al. Methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms and FOLFOX response in colorectal cancer patients. Br J Clin Pharmacol. 2010;69(1):58–66.

8.

Tsuji S, Midorikawa Y, Takahashi T, et al. Potential responders to FOLFOX therapy for colorectal cancer by Random Forests analysis. Br J Cancer. 2012;106(1):126–132.

9.

Gautier L, Cope L, Bolstad BM, Irizarry RA. Affy – analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 2004;20(3):307–315.

10.

Hubbell E, Liu WM, Mei R. Robust estimators for expression analysis. Bioinformatics. 2002;18(12):1585–1592.

11.

Peng H, Long F, Ding C. Feature selection based on mutual information: criteria of max-dependency, max-relevance, and min-redundancy. IEEE Trans Pattern Anal Mach Intell. 2005;27(8):1226–1238.

12.

Zhou Y, Zhang N, Li BQ, Huang T, Cai YD, Kong XY. A method to distinguish between lysine acetylation and lysine ubiquitination with feature selection and analysis. J Biomol Struct Dyn. 2015;33(11):2479–2490.

13.

Zhao TH, Jiang M, Huang T, et al. A novel method of predicting protein disordered regions based on sequence features. Biomed Res Int. 2013;2013:414327.

14.

Niu B, Huang G, Zheng L, et al. Prediction of substrate-enzyme-product interaction based on molecular descriptors and physicochemical properties. Biomed Res Int. 2013;2013:674215.

15.

Zhang N, Wang M, Zhang P, Huang T. Classification of cancers based on copy number variation landscapes. Biochimica et Biophysica Acta (BBA) – General Subjects. 2016;1860(11):2750–2755.

16.

Liu L, Chen L, Zhang Y-H, et al. Analysis and prediction of drug–drug interaction by minimum redundancy maximum relevance and incremental feature selection. J Biomol Struct Dyn. 2017;35(2):312–329.

17.

Li J, Huang T. Predicting and analyzing early wake-up associated gene expressions by integrating GWAS and eQTL studies. Biochim Biophys Acta. 2018;1864(6 Pt B):2241–2246.

18.

Huang T, Cai Y-D. An Information-Theoretic Machine Learning Approach to Expression QTL Analysis. PLoS One. 2013;8(6):e67899.

19.

Sun L, Yu Y, Huang T, et al. Associations between Ionomic Profile and Metabolic Abnormalities in Human Population. PLoS One. 2012;7(6):e38845.

20.

Zhang N, Huang T, Cai YD. Discriminating between deleterious and neutral non-frameshifting indels based on protein interaction networks and hybrid properties. Mol Genet Genomics. 2015;290(1):343–352.

21.

Shu Y, Zhang N, Kong X, Huang T, Cai Y-D. Predicting A-to-I RNA Editing by Feature Selection and Random Forest. PLoS One. 2014;9(10):e110607.

22.

Li BQ, You J, Huang T, Cai YD. Classification of non-small cell lung cancer based on copy number alterations. PLoS One. 2014;9(2):e88300.

23.

Jiang Y, Huang T, Chen L, Gao Y-F, Cai Y, Chou K-C. Signal Propagation in Protein Interaction Network during Colorectal Cancer Progression. Biomed Res Int. 2013;2013(1):287019.

24.

Zhang P-W, Chen L, Huang T, Zhang N, Kong X-Y, Cai Y-D. Classifying Ten Types of Major Cancers Based on Reverse Phase Protein Array Profiles. PLoS One. 2015;10(3):e0123147.

25.

Huang T, Shu Y, Cai Y-D. Genetic differences among ethnic groups. BMC Genomics. 2015;16(1):1093.

26.

Chen L, Li J, Zhang YH, et al. Identification of gene expression signatures across different types of neural stem cells with the Monte-Carlo feature selection method. J Cell Biochem. 2018;119(4):3394–3403.

27.

Todorov H, Fournier D, Gerber S. Principal components analysis: theory and application to gene expression data analysis. Genom Comput Biol. 2018;4(2):e100041.

28.

Szklo M, Nieto FJ. Epidemiology Beyond the Basics. Boston, MA: Jones and Bartlett; 2007.

29.

Li J, Bi L, Shi Z, et al. RNA-Seq analysis of non-small cell lung cancer in female never-smokers reveals candidate cancer-associated long non-coding RNAs. Pathol Res Pract. 2016;212(6):549–554.

30.

Yang H, Ma Y, Chen G, et al. Contribution of RIP3 and MLKL to immunogenic cell death signaling in cancer chemotherapy. Oncoimmunology. 2016;5(6):e1149673.

31.

Sun D, Zhao L, Lin J, Zhao Y, Zheng Y. Cationic liposome co-encapsulation of SMAC mimetic and zVAD using a novel lipid bilayer fusion loaded with MLKL-pDNA for tumour inhibition in vivo. J Drug Target. 2018;26(1):45–54.

32.

Fender AW, Nutter JM, Fitzgerald TL, Bertrand FE, Sigounas G. Notch-1 Promotes Stemness and Epithelial to Mesenchymal Transition in Colorectal Cancer. J Cell Biochem. 2015;116(11):2517–2527.

33.

Vinson KE, George DC, Fender AW, Bertrand FE, Sigounas G. The Notch pathway in colorectal cancer. Int J Cancer. 2016;138(8):1835–1842.

34.

Deshar R, Cho E-B, Yoon SK, Yoon J-B. CC2D1A and CC2D1B regulate degradation and signaling of EGFR and TLR4. Biochem Biophys Res Commun. 2016;480(2):280–287.

35.

Nakamura A, Naito M, Tsuruo T, Fujita N. Freud-1/Aki1, a Novel PDK1-Interacting Protein, Functions as a Scaffold To Activate the PDK1/Akt Pathway in Epidermal Growth Factor Signaling. Mol Cell Biol. 2008;28(19):5996–6009.

36.

Tuupanen S, Hänninen UA, Kondelin J, et al. Identification of 33 candidate oncogenes by screening for base-specific mutations. Br J Cancer. 2014;111(8):1657–1662.

37.

Gerbe F, van Es JH, Makrini L, et al. Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium. J Cell Biol. 2011;192(5):767–780.

38.

Notarnicola M, Messa C, Caruso MG. A significant role of lipogenic enzymes in colorectal cancer. Anticancer Res. 2012;32(7):2585–2590.

39.

Takasu S, Mutoh M, Takahashi M, Nakagama H. Lipoprotein Lipase as a Candidate Target for Cancer Prevention/Therapy. Biochem Res Int. 2012;2012(2):398697.

40.

Vainio P, Gupta S, Ketola K, et al. Arachidonic Acid Pathway Members PLA2G7, HPGD, EPHX2, and CYP4F8 Identified as Putative Novel Therapeutic Targets in Prostate Cancer. Am J Pathol. 2011;178(2):525–536.

41.

Brinkmann U, Vasmatzis G, Lee B, Yerushalmi N, Essand M, Pastan I. PAGE-1, an X chromosome-linked GAGE-like gene that is expressed in normal and neoplastic prostate, testis, and uterus. Proc Natl Acad Sci U S A. 1998;95(18):10757–10762.

42.

Iavarone C, Wolfgang C, Kumar V, et al. PAGE4 is a cytoplasmic protein that is expressed in normal prostate and in prostate cancers. Mol Cancer Ther. 2002;1(5):329–335.

43.

Kong U, Koo J, Choi K, Park J, Chang H. The expression of GAGE gene can predict aggressive biologic behavior of intestinal type of stomach cancer. Hepatogastroenterology. 2004;51(59):1519–1523.

44.

Chen Z, Li M, Yuan Y, Wang Q, Yan L, Gu J. Cancer/Testis Antigens and Clinical Risk Factors for Liver Metastasis of Colorectal Cancer: A Predictive Panel. Dis Colon Rectum. 2010;53(1):31–38.

45.

Chen A-P, Chang M-H, Romero MF. Functional analysis of nonsynonymous single nucleotide polymorphisms in human SLC26A9. Hum Mutat. 2012;33(8):1275–1284.

46.

Staub E, Buhr H-J, Gröne J. Predicting the site of origin of tumors by a gene expression signature derived from normal tissues. Oncogene. 2010;29(31):4485–4492.

47.

Greco FA. Cancer of unknown primary site: still an entity, a biological mystery and a metastatic model. Nat Rev Cancer. 2014;14(1):3–4.

48.

Huang T. Copy Number Variations in Tumors. Elsevier: Reference Module in Biomedical Sciences; 2018. Available from: https://doi.org/10.1016/B978-0-12-801238-3.65047-X. Accessed September 07, 2018.

49.

Huang T, Li B-Q, Cai Y-D. The integrative network of gene expression, microRNA, methylation and copy number variation in colon and rectal cancer. Curr Bioinform. 2016;11(1):59–65.

50.

Chen L, Huang T, Zhang Y-H, Jiang Y, Zheng M, Cai Y-D. Identification of novel candidate drivers connecting different dysfunctional levels for lung adenocarcinoma using protein-protein interactions and a shortest path approach. Sci Rep. 2016;6(1):29849.

51.

Huang T, Yang J, Cai Y-D. Novel Candidate Key Drivers in the Integrative Network of Genes, MicroRNAs, Methylations, and Copy Number Variations in Squamous Cell Lung Carcinoma. Biomed Res Int. 2015;2015(2):358125.

52.

Zhang TM, Huang T, Wang RF. Cross talk of chromosome instability, CpG island methylator phenotype and mismatch repair in colorectal cancer. Oncol Lett. 2018;16(2):1736–1746.

53.

The GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans. Science. 2015;348(6235):648–660.

Supplementary materials

Table S1 The clinical information of the 144 colorectal cancer patients
Abbreviation: NA, not applicable

Table S2 The top 138 mRMR genes

Creative Commons License © 2018 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.