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

Discovery of a Novel Immune Gene Signature with Profound Prognostic Value in Colorectal Cancer: A Model of Cooperativity Disorientation Created in the Process from Development to Cancer

  • Ning An,

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Xiaoyu Shi,

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Yueming Zhang,

    Affiliation Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China

  • Ning Lv,

    Affiliation Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China

  • Lin Feng,

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Xuebing Di,

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Naijun Han,

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Guiqi Wang,

    Affiliation Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China

  • Shujun Cheng ,

    Contributed equally to this work with: Shujun Cheng, Kaitai Zhang

    chengshj@263.net.cn (SC); zhangkt_bingyin@sina.cn (KZ)

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

  • Kaitai Zhang

    Contributed equally to this work with: Shujun Cheng, Kaitai Zhang

    chengshj@263.net.cn (SC); zhangkt_bingyin@sina.cn (KZ)

    Affiliation State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

Abstract

Immune response-related genes play a major role in colorectal carcinogenesis by mediating inflammation or immune-surveillance evasion. Although remarkable progress has been made to investigate the underlying mechanism, the understanding of the complicated carcinogenesis process was enormously hindered by large-scale tumor heterogeneity. Development and carcinogenesis share striking similarities in their cellular behavior and underlying molecular mechanisms. The association between embryonic development and carcinogenesis makes embryonic development a viable reference model for studying cancer thereby circumventing the potentially misleading complexity of tumor heterogeneity. Here we proposed that the immune genes, responsible for intra-immune cooperativity disorientation (defined in this study as disruption of developmental expression correlation patterns during carcinogenesis), probably contain untapped prognostic resource of colorectal cancer. In this study, we determined the mRNA expression profile of 137 human biopsy samples, including samples from different stages of human colonic development, colorectal precancerous progression and colorectal cancer samples, among which 60 were also used to generate miRNA expression profile. We originally established Spearman correlation transition model to quantify the cooperativity disorientation associated with the transition from normal to precancerous to cancer tissue, in conjunction with miRNA-mRNA regulatory network and machine learning algorithm to identify genes with prognostic value. Finally, a 12-gene signature was extracted, whose prognostic value was evaluated using Kaplan–Meier survival analysis in five independent datasets. Using the log-rank test, the 12-gene signature was closely related to overall survival in four datasets (GSE17536, n = 177, p = 0.0054; GSE17537, n = 55, p = 0.0039; GSE39582, n = 562, p = 0.13; GSE39084, n = 70, p = 0.11), and significantly associated with disease-free survival in four datasets (GSE17536, n = 177, p = 0.0018; GSE17537, n = 55, p = 0.016; GSE39582, n = 557, p = 4.4e-05; GSE14333, n = 226, p = 0.032). Cox regression analysis confirmed that the 12-gene signature was an independent factor in predicting colorectal cancer patient’s overall survival (hazard ratio: 1.759; 95% confidence interval: 1.126–2.746; p = 0.013], as well as disease-free survival (hazard ratio: 2.116; 95% confidence interval: 1.324–3.380; p = 0.002).

Introduction

Colorectal cancer (CRC) is the third most common cancer in men (746,000 cases, 10.0% of all cancers) and the second in women (614,000 cases, 9.2% of the all cancers) worldwide [1]. Despite significant advances in understanding its molecular mechanism, CRC remains a major cause of cancer mortality [2]. Previous researches suggested large-scale heterogeneity occurred in CRC [35], as well as in many other types of cancer [68]. Tumor heterogeneity develops through a sequence of events, guided by clonal selection, where genomic instability contributes to generating a diverse cell population that is subject to selection in a micro-environmental or therapeutic context [9]. Therefore, a novel model that shares similarities with cancer in terms of cell-behavioral and molecular attributes, but that is intrinsically more “organized” is urgently needed.

It has been more than 150 years since Rudolf Virchow first proposed that neoplasms arise “in accordance with the same law, which regulates embryonic development” in 1858. The association between embryonic development and carcinogenesis is widely reported. Furthermore, certain key developmental genes are also involved in carcinogenesis through mutational activation [10]. Through developmental animal models, molecular mechanisms of carcinogenesis have been unveiled and a variety of novel cancer-related molecules, pathways and biomarkers identified [1113]. Embryonic development and carcinogenesis also share many other similarities with respect to cellular behavior, including epithelial-to-mesenchymal transition (EMT) [14], mesenchymal-to-epithelial transition (MET) [15], and immune-surveillance evasion [16]. Taken together, these findings offer convincing evidence that tumor can be viewed as an aberrant organ that has acquired the capacity for indefinite proliferation through accumulated strikes [17], and that the molecular events which deviate tumor cells from the normal developmental path, are probably accountable for cancer initiation and progression.

Pairwise gene expression correlations (using Pearson correlation) are often used to determine associations between genes in transcriptomic studies [1820]. The pairwise gene expression correlations in development stage manifest the physiological close or distant associations of gene to gene regulation. Our study indicates that the correlations between genes within a given functional group (immune response) show a remarkably compact and synchronized pattern of gene expression that ensures tight regulation of colonic development. For a given gene, the rank order of its correlation with the remaining members of this gene group, representing the biological association topology, was probably disturbed during carcinogenesis (regulatory relations were stepwise switched from physiological to pathological status). We hypothesized that, if one views a tumor as an aberrant developing organ, the culprit genes responsible for disrupting the integrity of this coordinated gene expression correlation pattern and more specifically, disrupting the rank order of the correlation pattern within this particular gene group during carcinogenesis, probably hold profound prognostic information. We defined this concept as “cooperativity disorientation”, and originally constructed a Spearman transition model to quantify cooperativity disorientation that arises during the progression from colonic development to precancerous progression to cancer, rather than simply concentrate on differentially expressed genes by specific phenotypes.

MicroRNAs (miRNAs) are a class of small non-coding RNAs, ~22 nt in length, that regulate gene expression by binding to the 3′-untranslated region (3′-UTR) of target genes leading to degradation or protein translation inhibition of target genes [21]. MiRNAs are predicted to regulate more than 60% of all protein-coding genes in mammals [22], thereby regulating almost every cellular process [23, 24]. We hypothesized that miRNAs play a pivotal role in CRC patient survival and that the downstream targets of these miRNAs may have prognostic value; this strategy was also adopted by Yang et al. [25] who showed that the expression of 219 miRNA-associated genes was associated with a mesenchymal subtype of serous ovarian cancer associated with poor overall survival (OS) [25].

Although the relationship between cooperativity disorientation, embryonic development and carcinogenesis is still not clear, it is plausible that certain miRNA regulated genes, which play important roles in development stage and contribute to cooperativity disorientation during carcinogenesis, might have a substantial impact on cancer transformation. These genes could be promising candidate prognostic biomarkers.

In this study, we focused on immune response-related genes. The immune response and more specifically, inflammation, has a profound influence on carcinogenesis, which can either kill tumor cells, or, in some circumstances, can be mobilized to facilitate carcinogenesis [26]. The importance of the immune response in carcinogenesis prompted us to determine prognostic biomarkers for CRC. Ours is the first study to examine a range of samples, from human colon embryonic development, colorectal precancerous progression, to CRC samples, in order to simulate the trajectory of human colon development and carcinogenesis. The Spearman transition model we proposed here represents the first step in identifying the culprit genes [differentially expressed genes (DEGs) with a new interpretation on the basis of expression correlation pattern] responsible for disrupting the organized correlation pattern among immune-related genes during carcinogenesis. Using microarray technology and bioinformatics analyses, we identified a 12-gene signature with significant prognostic value, which may be clinically relevant in future.

Materials and Methods

A schematic for the study is depicted in Fig 1.

thumbnail
Fig 1. Schematic of the stepwise gene signature selection and evaluation workflow.

CRC colorectal cancer, DVIG development varying immune gene, OS overall survival, DFS disease-free survival.

https://doi.org/10.1371/journal.pone.0137171.g001

Patients and samples

In accordance with the principles of gastrointestinal developmental biology [27], developing colon samples were obtained from 20 abortion cases at the Maternal & Child Health Care Hospital of Hai Dian between 2007 and 2009. The samples included whole embryos (WE) at three to five postovulatory weeks (PWs), early embryonic colons (EEC) at eight to ten PWs and middle embryonic colons (MEC) at 14 to 22 PWs. Within 10 minutes of abortion, tissues were rinsed with normal saline, and whole embryos or embryonic colons were carefully separated from fetal tissues with the guidance of a Nikon stereo microscope SMZ1500 (Japan). Embryos/fetuses with known or suspected genetic disorders were excluded.

Normal colorectal mucosal samples were collected from patients with hemorrhoids who received surgical excision in the Department of Colon and Rectal Surgery of Beijing Shi Ji Tan Hospital between 2009 and 2010. Fifty-two CRC samples with OS information were obtained during surgical resection from Zhe Jiang University School of Medicine. Colonoscopy biopsy samples, including colorectal adenomas and adenocarcinomas were obtained from the Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, between 2008 and 2011. Patients with the history of familial adenomatous polyposis, hereditary non-polyposis CRC, or inflammatory bowel disease were excluded. Adenoma is defined as dysplasia, carcinoma in situ, suspicion for invasive carcinoma and intramucosal carcinoma; the adenocarcinoma is defined as submucosal invasion by adenocarcinoma [28]. Four to six areas were excised from colorectal neoplasia samples, including the edges and the center of the lesion, according to ASGE guideline [29]. Tissue samples were all snap-frozen in liquid nitrogen immediately after biopsy or surgery and stored at -80°C. A portion of all the samples was subjected to pathological analysis performed by two independent, blinded and experienced pathologists. Samples satisfied with the diagnostic criteria of normal mucosa and neoplasia (neoplastic cells >70%) were enrolled. If more than one biopsy sample was taken from the same patient, these samples were pooled. All donors signed informed consent forms. The use of human tissue samples and the experimental procedures for this study were reviewed and approved by the Ethics Committee of the Cancer Institute and Hospital, Chinese Academy of Medical Sciences.

RNA isolation

Total RNA was extracted from frozen tissues using TRIzol RNA isolation reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s specifications. RNA integrity was evaluated using a 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). If the RNA integrity number was ≥ 5, the total RNA was further purified using the RNeasy Mini Kit (Cat No.74106, Qiagen, Germany). RNA concentrations were determined with a NanoDrop ND-1000 Spectrophotometer (NanoDrop Technologies, Wilmington, USA).

Microarray expression profiling and data normalization

After histopathological evaluation and RNA integrity analysis, all the samples were analyzed using Agilent microarrays. Biopsy samples including 6 WE, 6 EEC, 8 MEC, 12 normal, 58 adenoma and 47 adenocarcinoma samples were used for mRNA microarray analysis; of these, 60 samples (2 WE, 6 EEC, 8 MEC, 11 normal, 9 adenoma and 24 adenocarcinoma samples) were also used for miRNA microarray analysis. Purified total RNA samples were labeled and hybridized to Agilent 4×44K Whole Human Genome Oligo Microarrays (G4112F) according to the manufacturer’s instructions. For the miRNA arrays, total RNA was analyzed with an Agilent 8×15K Human miRNA Microarray V3 (G4470C).

The mRNA and miRNA microarray raw data were normalized using the GeneSpring GX software, version 11.5 (Silicon Genetics, Redwood City, CA, USA). For the mRNA expression data, a total of 41,091 single probes were obtained according to GeneSpring’s default setting. The expression value for a particular gene was determined as the median value of all probes mapping to this gene. Eventually, the expression values of 18,986 genes were obtained. Measured miRNAs were deemed present if their signal could be detected in at least 50% of the samples within each sample type. The expression profiles were acquired for 96 miRNAs. The raw and processed data have been deposited in the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database with the series accession numbers GSE71187 (mRNA data) and GSE71130 (miRNA data).

Affymetrix microarray data collection, preprocessing, and normalization

The raw data for five human colorectal cancer mRNA microarray studies (Table 1) were downloaded from GEO. The combined data set contained a total of 1,094 samples was processed on Affymetrix HG-U133A Plus2 (GPL570) arrays, which contain 52,475 probes. Normalized expression values were obtained via the robust multi-array average (RMA) algorithm and further quantile normalized using the “affy” Bioconductor package. The ComBat algorithm was utilized to eliminate potential batch effects with the Bioconductor package “inSilicoMerging”. The expression levels of 20,184 genes were obtained as the median value of all probes mapping to a particular gene. All clinical information was extracted from the original publications. Among these five datasets, GSE17536, GSE17537 and GSE39582 contain both OS and disease-free survival (DFS) information. GSE39084 contains OS data only, while GSE14333 contains DFS information only.

thumbnail
Table 1. Affymetrix microarray datasets included in this study, used to evaluate the prognostic value of our 12-gene signature.

https://doi.org/10.1371/journal.pone.0137171.t001

Identifying “development-varying immune-related genes” (DVIGs)

WE, EEC, MEC and normal samples represented samples at different stages of human colonic development; adenomas were regarded as precancerous lesions; and adenocarcinomas represented the cancer stage. Genes that fell under the Gene Ontology (http://www.geneontology.org) term GO:0006955 were considered immune response-related genes; this resulted in 1028 genes of which 972 were present in our mRNA microarray data. ANOVA was used to retrieve 665 DVIGs that were differentially expressed during the developmental stage (FDR<0.0001).

Establishment of the Spearman transition model

A detailed description of the Spearman transition model is presented in S1 Methods.

Construction of a miRNA-mRNA regulatory network

A miRNA-mRNA regulatory network was generated based on sequence algorithms (miRanda [30], TargetScan [31], PicTar [32]) and microarray data (60 biopsy samples with both miRNA and mRNA microarray data). A miRNA-mRNA regulatory pair was regarded as solid only if they satisfied at least two sequence algorithms and if their expression levels were significantly and inversely correlated (FDR<0.01).

Establishing a CRC gene signature using machine learning applied to mRNA expression profiles of primary clinical samples

Of the 52 surgically excised CRC samples, we selected 19 and 22 cases where patients survived longer (“Good group”) or shorter (“Poor group”) than five years (after surgery) to train a random forests machine learning model. Briefly, genes were ordered by the mean decrease Gini (MDG) criterion, where genes are ranked by their level of influence on the performance of the random forests classification; leave one out cross validation (LOOCV) to estimate “Poor voting” proportion of the test case, which was further treated as predictor in receiver operating characteristic (ROC) test. Genes were then recursively eliminated based on the initial gene ranking, until the area under ROC curve (AUC) was optimized. This algorithm is clearly described in previous research as the AUC-RF algorithm[33].

Kaplan–Meier survival analysis and Cox regression analysis

Principal component analysis (PCA) was conducted using genes of interest in each downloaded Affymetrix dataset. The first principal component (PC1) captures the greatest amount of total variance in the profiles and was calculated for each patient. Patients were then divided into two groups of equal size based on the rank order of PC1 across their tumor profiles. Kaplan–Meier survival analysis and the log-rank test were used to evaluate the prognostic difference between the two PC1-assigned groups [12]. The Cox proportional hazards regression model was used to evaluate the independence of the prognostic factors in a stepwise manner. Samples in the combined Affymetrix dataset (1,094 samples) with complete information of age, sex, American Joint Committee on Cancer (AJCC) stage (stage), pathological grade (grade) and survival information were used (213 samples for OS Cox analysis and 213 samples for DFS Cox analysis), and a value of p < 0.05 was regarded as significant.

Gene signature validation using random gene sampling

Our strategy was to select a small gene signature with significant prognostic value by narrowing down genes of interest in a stepwise manner. To prove that this method truly predicted survival outcome, n-gene (where the final gene signature contains n genes) random samplings were performed 2000 times in each gene pool. Kaplan–Meier survival analysis was performed with randomly chosen n genes, and the number of times that randomly chosen genes could simultaneously discriminate all the target survival datasets was recorded.

Statistical Analyses

All statistical analyses were executed using R project software (Version 2.15.1), and Bioconductor (Version 2.11). The R packages “randomForest” (Version 4.6–7) [34] and “pROC” (Version 1.7.1) [35] were used to construct the AUC-RF model. Differentially expressed genes were obtained using the R package “samr”. Kaplan–Meier survival analysis was carried out using R package “survival”. The Bioconductor annotation package “org.Hs.eg.db” (Version 2.8.0) was used to retrieve immune-related genes [36]. Mappings between Affymetrix probes and Entrez gene identifiers were carried out using the Bioconductor package “hgu133plus2.db”. Meta-analysis was conducted with the R package “meta”, and forest plots were made using the R package “rmeta”. Network visualization was performed in Cytoscape (Version 3.2.0) [37].

Results

Genes differentially expressed between normal and CRC tissue are significantly enriched for the Reactome term “signaling in immune system”

Differentially expressed genes (DEGs) between normal and CRC tissues were identified using the SAM algorithm (FDR<1e-07). DEGs included 3,226 and 2,538 significantly up- and downregulated genes in CRC. Using Reactome enrichment analysis, conducted with DAVID Bioinformatics Resources 6.7 (http://david.abcc.ncifcrf.gov/), we found that the Reactome term “signaling in immune system” was significantly enriched in CRCs (Bonferroni-adjusted p value = 0.004), suggesting a significant association between carcinogenesis and immune-related genes (S1 Table).

Pearson correlation heatmaps

Pairwise Pearson correlations among the 665 DVIGs were calculated and adjusted to eliminate bias (adjusted method was described in S1 Methods). Pearson correlation heatmaps (665×665) were constructed for colon samples during colonic development (Fig 2A), progression (Fig 2B) and cancer (Fig 2C) stages. During the developmental stage, three distinct clusters were obtained. Distinct clusters were however not evident during the progression and cancer stages. By superimposing the three Pearson correlation density curves (Fig 2D), a clear bimodal distribution was seen for the developmental stage, in contrast to the unimodal distributions of the progression and cancer stages. Further, the cancer stage had a higher maximum density at a Pearson correlation of zero compared with the progression or developmental stages.

thumbnail
Fig 2. Pearson correlation heatmaps and density curve plot of 665 DVIGs.

Heatmaps of adjusted Pearson correlations for 665 DVIGs in (A) development, (B) precancerous progression and (C) cancer, respectively. Genes were clustered into three clusters (highlighted with different colors) by UCA. (D) Density plot of pairwise adjusted Pearson correlations for all three stages. The curve for the development stage is bimodal distribution, but unimodal for in progression and cancer stages. In order to render intra-immune vectors comparable, genes were reordered in the progression and cancer stage heatmaps to match the order in the development stage heatmap, to generate (E) reordered progression heatmap and (F) reordered cancer heatmap. DVIG, development varying immune gene; UCA, unsupervised clustering algorithm.

https://doi.org/10.1371/journal.pone.0137171.g002

“Obedient genes” were filtered out using Spearman transition model

Pearson correlation heatmaps of DVIGs during the progression and cancer stages were reordered to make all three stages (Fig 2E and 2F, described in S1 Methods). As shown in Fig 3A, the 665 DVIGs were projected onto a Spearman transition coordinate system, with the Spearman transition between development and progression (STD-P, S1 Methods) and between progression and cancer (STP-C, S1 Methods) as the x and y axis coordinates, respectively. Genes were colored in the same way as in the developmental heatmap clustering in Fig 2A. Of the 665 DVIGs, 385 (termed “obedient genes”) fell within the quarter circle’s arc, while the remaining 280 (termed “diversion genes”) fell outside this arc and were used as candidates for downstream selection procedures.

thumbnail
Fig 3. Gene signature optimization based on Spearman correlation transition model and AUC-RF algorithm.

(A) The 665 DVIGs were projected onto a Spearman correlation transition coordinate system based on their cooperativity disorientation between the consecutive stages. Genes were colored in the same way as in the development heatmap. (B) The AUC-RF algorithm was used for gene signature optimization. Genes were recursively removed from an importance-ordered gene list until the largest AUC value was met. (C) The biggest AUC of 0.904 (95% CI: 0.799~1.000) was obtained when the number of genes were reduced to 12, with 81.8% sensitivity (95% CI: 0.636–0.955) and 89.5% specificity (95% CI: 0.737–1.000). Dev, development; Prog, progression; TPS, theoretically stable point; AUC, area under curve; DVIG, development varying immune gene; CI, confidence interval.

https://doi.org/10.1371/journal.pone.0137171.g003

Diversion genes with one or more miRNA regulators stayed in the gene pool

Using the paired mRNA and miRNA data that were available for 60 of the CRC biopsy samples, we constructed a miRNA-mRNA regulatory network to select diversion genes that had at least one miRNA regulator (S1 Methods). This resulted in 59 diversion genes that were potentially regulated by 37 miRNAs (Fig 4, S2 Table).

thumbnail
Fig 4. miRNA-mRNA regulatory network.

Dark yellow nodes represent miRNAs. Red and sapphire nodes represent mRNAs, among which red ones are genes in the 12-gene signature. Directed solid edges represent miRNA-mRNA regulation.

https://doi.org/10.1371/journal.pone.0137171.g004

Gene signature optimization by AUC-RF algorithm

The 59 diversion genes were further narrowed down these 59 genes to obtain the subset of genes providing the best prognostic performance. These genes were first ordered according to their importance in discriminating cases by OS (longer or shorter than 5 years) using the random forest algorithm; genes were then recursively eliminated from the bottom of this list until the area under ROC curve (AUC) was optimized (AUC = 0.904, 95% CI: 0.799–1.000, Fig 3B). This resulted in an optimized 12-gene signature that had 81.8% sensitivity (95% CI: 0.636–0.955) and 89.5% specificity (95% CI: 0.737–1.000) in discriminating poor from good OS in 52 surgery samples with a “Poor” voting proportion of 0.560 (Fig 3C). This 12-gene signature is composed of AXL, BCL3, COLEC12, ABR, PXDN, EP300, JAM3, MAP3K1, CASP8, RPS6KA1, CHUK, and RPS6KA2, and is regulated by 16 miRNAs (Fig 4).

Kaplan–Meier survival and Cox regression analysis confirmed the validity of the 12-gene signature

Kaplan–Meier survival analysis was conducted to evaluate the prognostic value of the 12-gene signature in five Affymetrix datasets retrieved from the GEO database. The log-rank test results confirmed that the 12-gene signature was closely related to OS in four datasets (Fig 5A; GSE17536, n = 177, p = 0.0054; GSE17537, n = 55, p = 0.0039; GSE39582, n = 562, p = 0.13; GSE39084, n = 70, p = 0.11). Furthermore, this 12-gene signature was significantly associated with DFS in four datasets (Fig 5B; GSE17536, n = 177, p = 0.0018; GSE17537, n = 55, p = 0.016; GSE39582, n = 557, p = 4.4e-05; GSE14333, n = 226, p = 0.032). Cox regression analysis also confirmed that the 12-gene signature was an independent factor in predicting CRC patient’s OS [Table 2; hazard ratio (HR): 1.759; 95% CI: 1.126–2.746; p = 0.013], as well as DFS (Table 2; HR: 2.116; 95% CI: 1.324–3.380; p = 0.002). Meta-analysis was conducted to evaluate the correlation between each of the 12 genes and survival (OS: GSE17536, GSE17537, GSE39582 and GSE39084; and DFS: GSE17536, GSE17537, GSE39582 and GSE14333) of CRC patients with fixed-effect model (Fig 6A) and random-effect model (Fig 6B).

thumbnail
Fig 5. Kaplan–Meier survival analyses and log-rank tests of the 12-gene signature.

Kaplan–Meier survival analyses and log-rank tests were conducted to evaluate the prognostic value of the 12-gene signature. (A) The performance of the 12-gene signature in OS discrimination. Datasets with OS information were GSE17536, GSE17537, GSE39582 and GSE39084. (B) The performance of the 12-gene signature in DFS discrimination. Datasets with DFS information were GSE17536, GSE17537, GSE39582 and GSE14333. OS, overall survival; DFS, disease-free survival.

https://doi.org/10.1371/journal.pone.0137171.g005

thumbnail
Fig 6. Forest plot of the association between individual genes in the 12-gene signature and CRC survival.

(A) Forest plot of the association between individual genes and OS with a fixed-effect model in datasets containing OS information (GSE17536, GSE17537, GSE39582 and GSE39084). Meta-analysis of these 12 genes in four independent datasets was conducted, and HR, 95% CI of each gene and corresponding p value were calculated and plotted in the forest plot. (B) Forest plot of the association between individual genes and DFS with a random-effect model in four datasets containing DFS information (GSE17536, GSE17537, GSE39582 and GSE14333). CRC, colorectal cancer; HR, hazard ratio; CI; confidence interval; OS, overall survival; DFS, disease-free survival.

https://doi.org/10.1371/journal.pone.0137171.g006

thumbnail
Table 2. Univariate and multivariate analyses of overall survival and disease-free survival (Cox proportional hazards regression model) in 213 patients according to age, sex, stage and the first principal component (PC1) assigned groups calculated with the 12-gene signature.

https://doi.org/10.1371/journal.pone.0137171.t002

Random gene sampling verified the validity of our methodology

To confirm the validity of our signature selection process, a 12-gene panel was randomly sampled 2000 times across the 972 immune-related genes, 665 DVIGs, 280 diversion genes and 59 miRNA-regulated genes, respectively. The number of times that a randomly chosen 12-gene panel could simultaneously discriminate the survival datasets (OS and DFS in GSE17536 and GSE17537, DFS in GSE39582 and GSE14333), was 0, 0, 9 and 33 for the aforementioned four gene groups, respectively, providing strong evidence for the validity of our hypothesis and gene signature-selection pipeline (Fig 7A).

thumbnail
Fig 7. Random gene sampling verified the validity of our step- gene selection procedure.

(A) Bar plot of the number of times that 12 randomly chosen genes could simultaneously discriminate four survival datasets (OS and DFS in GES17536 and GSE17537, DFS in GSE39582 and GSE14333). (B) Heatmap of 137 biopsy samples established with mRNA expression profile of the 12-gene signature. The mRNA raw data were normalized and then filtered (see “Materials and Methods”). Rows represent genes, and columns represent biopsy samples. Rows, rather than columns, were reordered using UCA, whereas samples of the same type were placed together. DVIG, development varying immune gene; UCA, unsupervised clustering algorithm; OS, overall survival; DFS, disease-free survival.

https://doi.org/10.1371/journal.pone.0137171.g007

Discussion

The intimate association between embryonic development and carcinogenesis makes embryonic development a viable reference model to study cancer, which circumvents the potentially misleading complexity associated with tumor heterogeneity. The molecular resemblances between certain malignant tumors and developing tissues have been reported on the basis of transcription factor activity [38], regulation of chromatin structure [39] and cellular signaling [40]. Important molecules were reported to play a substantial role in both embryonic development and carcinogenesis: PTCH1 is a key regulator of embryonic development, whose overexpression could drive skin carcinogenesis [41]. Developmental animal models have been used to uncover the complicated molecular mechanisms of carcinogenesis, and a variety of novel and pivotal molecules, pathways and biomarkers have been discovered [12, 42, 43]. For instance, many important signaling pathways, including NOTCH1 signaling, play a key role in development and in carcinogenesis [44].

These findings prompted scientists to further examine the commonalities between embryonic development and cancer. The two most widely accepted theories are: cancers may, via mutational, epigenetic changes and genome instability, acquire a variety of critical phenotypic traits that compound to facilitate territorial expansion, via proliferative self-renewal, migration and invasion, which are also phenotypic characteristics of embryonic development [4547]; The second theory is that tumors originate from either tissue stem cells or from their immediate progeny through dysregulation of (normally tightly regulated) self-renewal, so that tumors retain certain key embryonic/stem cell properties [48].

The undeniable relationship between inflammation and cancer has been reported and is firmly established. Infection and chronic inflammation contribute to an estimated 25% of all cancers worldwide [49]. “Smoldering” inflammation refers to chronic, often subclinical inflammation, which has many tumor-promoting effects such as increased proliferation and survival of malignant cells, promotion of angiogenesis and metastasis, subversion of the adaptive immune response, and alteration in the response to hormones and chemotherapeutic agents [50]. Chronic inflammation is particularly relevant to CRC: patients suffering from inflammatory bowel disease (which includes ulcerative colitis and Crohn’s disease), for instance, are at increased risk of developing CRC [51]. Immuno-compromised patients, such as organ transplant recipients [52, 53] or patients with acquired immune deficiency syndrome (AIDS) patients [54, 55], have a remarkable propensity for cancer formation. In addition, certain anti-inflammatory drugs have the ability to reduce CRC risk [56, 57]. In developmental biology, the fetus, which in many ways behaves like an allogenic transplant also evades maternal immune-surveillance by adopting similar mechanisms with tumors [16]. Thus, immune-related genes play a very important role in carcinogenesis as well as in embryonic development.

Ours is the first study to use the mRNA and miRNA expression profiles of human colonic development, colorectal precancerous progression, and cancer samples, to recapitulate the trajectory of human colonic development and carcinogenesis, in an attempt to identify a gene signature with prognostic value. The close association between development and carcinogenesis and the significance of immune-related genes in both processes, substantiates our choice of using immune-related genes as the initial gene pool. The immune-related genes differentially expressed during development are more likely to contain prognostic information of CRC, so we collected 665 DVIGs during the first gene selection step. The correlation pattern within these immune genes was highly organized and synchronized, implying that gene among immune genes were tightly regulated to keep the whole developmental process safe and steady. During carcinogenesis, the culprit genes, which severely sabotage this subtle machinery and drag tumor cell away from normal developmental pathway on the basis of expression correlation, are probably promising indicators of CRC prognosis. In this regard, we initially proposed the Spearman correlation transition model to identify these aberrant genes causing tremendous cooperativity disorientation, and these genes may therefore have significant prognostic value.

In the first step of our Spearman correlation transition model, we calculated pairwise adjusted Pearson correlations for all the 665 DVIGs during development, progression and cancer. Suppose there are two genes A and B, if the correlation value between A and B is close to 1 or -1, the expressions of A and B are strongly synchronized; if the value is close to 0, the expression of A is random to B, and the chance of A and B having a biological association is relatively slim. The expressions of the 665 DVIGs were strongly correlated during colonic development, which implied coordinate regulation of these genes. However, in precancerous progression and cancer stages, the accurate cooperation was corrupted and the expression seems substantially randomized to each other, manifesting massive cooperativity disorientation (a concept we defined in this study) occurred throughout carcinogenesis in terms of immune gene’s expression.

Intra-immune vector (explained in S1 Methods) described the cooperative relation of a particular gene with other immune genes in each stage. A quantitative parameter is needed to describe the shift of this intra-immune relation between consecutive stages for each gene, so we chose Spearman correlation to fulfill the mission. The progression and cancer stage heatmaps were reordered according to the order of the development stage heatmap, to make the development intra-immune vector (DIV), progression intra-immune vector (PIV) and cancer intra-immune vector (CIV) comparable (Fig 2E and 2F). Suppose there is a DVIG named A, for which the STD-P (Spearman correlation between A’s DIV and PIV) is close to 1, indicating in the transition between development and progression stage, the rank order of Gene A’s association with other immune genes was nearly the same; STD-P is close to -1, meaning the rank order was nearly turned upside down; STD-P is close to 0, suggesting the rank order was randomly reshuffled. Likewise, STP-C of Gene A is the parameter denoting the transition of intra-immune cooperativity disorientation between progression and cancer stage. Therefore, we treated STD-P and STP-C as x-axis and y-axis coordinate of their corresponding gene, and in this way, projected all the 665 DVIGs onto the Spearman transition coordinate system.

Point (1, 1) was defined as theoretically stable point (TSP). Suppose a gene was projected onto this TSP, it inferred the cooperativity status (implying biological association) of this gene with other immune-related genes in the whole process of carcinogenesis was absolutely stable. The closer to the TSP a given gene is located in the Spearman correlation transition system, the more stable of this gene during carcinogenesis. Use one Euclidean distance from TSP as the threshold (S1 Methods), we excluded 385 obedient DVIGs and used 280 diversion DVIGs for further narrow-down procedures.

By constructing the miRNA-mRNA regulatory network, we identified 59 (of 280 diversion genes) that were potentially regulated by miRNAs. Using permutation analysis (Fig 7A), our gene signature selection pipeline (Spearman transition model and miRNA-associated genes) proved effective in identifying genes with prognostic significance.

The final 12-gene signature was selected (from the 59 miRNA-regulated genes) using the AUC-RF algorithm (Fig 7B). This 12-gene signature is closely associated with OS of CRC patients, and performed even better in predicting DFS of CRC patients, probably because OS is more likely to be affected by a variety of extraneous factors, such as cardiac diseases, malnutrition etc. Comparing with OS, DFS is much more related to the cancer itself, which might be a sensible explanation for the 12-gene signature (reflecting cooperativity disorientation of the tumor itself) performing better in DFS than OS.

Our results are supported by evidence from the literature. Among the predicted miRNA-mRNA interactions, the regulatory pair miR-34aAXL has been implicated in chronic lymphocytic leukemia, breast cancer and other cancer types [5860]; miR-200b and miR-200c were proven as major regulators of EP300, which suppresses metastasis in ductal adenocarcinomas of the pancreas [61]. Meanwhile, many of the genes in our 12-gene signature are involved in key cancer-related pathways (S3 Table), which have important implications for cancer formation, prognosis and clinical management. AXL is an important mediator of inherent and chemotherapy-induced invasion and a predictor of poor clinical outcome in early-stage CRC [60]. Frequent microsatellite instability and consequent loss of EP300 expression has been reported in gastric and colorectal cancers [62]. MAP3K1 regulates JNK activation and is altered in a variety of cancer types [63]. The 16 upstream miRNAs regulating the 12 genes in the miRNA-mRNA network also influence cancer phenotype and clinical outcome. For example, miR-23a antisense enhanced 5-FU-induced apoptosis in CRC cells [64], and was used in a triple miRNA classifier for CRC early detection [65]. Down-regulation of miR-20b was observed in various types of CRC, and occurs as an early event of colorectal carcinogenesis in familial adenomatous polyposis tumors [66]. MiR-17 is a predictive factor for chemotherapy response, a prognostic factor for OS in CRC [67], and also an oncogenic miRNA that regulates tumorigenesis and progression [68].

Conclusions

As far as we know, this is the first study to use mRNA and miRNA expression profiles of human colonic development, precancerous progression and cancer samples, and together with bioinformatics approaches aimed at stepwise selection of immune-related genes to identify a 12-gene signature with profound prognostic potential for CRC. Spearman transition model was originally constructed to quantify the level of cooperativity disorientation associated with progression from normal to precancerous to cancer tissue, which hopefully was able to reveal prognostic information that would probably have been missed by simply comparing gene expression levels between distinct sample types. Further investigation is needed to unravel the underlying molecular mechanism of these 12 genes, which may have diagnostic and therapeutic potential.

Supporting Information

S1 Methods. Spearman correlation transition model.

https://doi.org/10.1371/journal.pone.0137171.s001

(DOCX)

S1 Table. Reactome enrichment of differentially expressed genes (DEGs) in colorectal cancer.

https://doi.org/10.1371/journal.pone.0137171.s002

(DOCX)

S2 Table. Gene lists of the miRNA-mRNA regulatory network constructed using bioinformatics analysis and 60 paired mRNA and microRNA profiles from CRCs.

https://doi.org/10.1371/journal.pone.0137171.s003

(DOCX)

S3 Table. KEGG pathways containing at least one gene of the 12-gene signature.

https://doi.org/10.1371/journal.pone.0137171.s004

(XLSX)

Acknowledgments

We would like to thank Prof. Ying Hu for his helpful suggestions and assistance with the study design, Dr. Xue Yang for advice in manuscript writing and submission.

Author Contributions

Conceived and designed the experiments: KZ SC GW. Performed the experiments: NA XS YZ. Analyzed the data: NA LF NL. Contributed reagents/materials/analysis tools: XD NH. Wrote the paper: NA KZ SC.

References

  1. 1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86. Epub 2014/09/16. pmid:25220842.
  2. 2. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014;383(9927):1490–502. Epub 2013/11/15. pmid:24225001.
  3. 3. Kanthan R, Senger JL, Kanthan SC. Molecular events in primary and metastatic colorectal carcinoma: a review. Patholog Res Int. 2012;2012:597497. Epub 2012/09/22. pmid:22997602; PubMed Central PMCID: PMC3357597.
  4. 4. Troiani T, Martinelli E, Napolitano S, Morgillo F, Belli G, Cioffi L, et al. Molecular aspects of resistance to biological and non-biological drugs and strategies to overcome resistance in colorectal cancer. Curr Med Chem. 2014;21(14):1639–53. Epub 2013/09/03. pmid:23992332.
  5. 5. Schlicker A, Beran G, Chresta CM, McWalter G, Pritchard A, Weston S, et al. Subtypes of primary colorectal tumors correlate with response to targeted treatment in colorectal cell lines. BMC Med Genomics. 2012;5:66. Epub 2013/01/01. pmid:23272949; PubMed Central PMCID: PMC3543849.
  6. 6. Jeng KS, Chang CF, Jeng WJ, Sheen IS, Jeng CJ. Heterogeneity of hepatocellular carcinoma contributes to cancer progression. Crit Rev Oncol Hematol. 2015. Epub 2015/02/15. pmid:25680939.
  7. 7. Skibinski A, Kuperwasser C. The origin of breast tumor heterogeneity. Oncogene. 2015. Epub 2015/02/24. pmid:25703331.
  8. 8. Cheng X, Chen H. Tumor heterogeneity and resistance to EGFR-targeted therapy in advanced nonsmall cell lung cancer: challenges and perspectives. Onco Targets Ther. 2014;7:1689–704. Epub 2014/10/07. pmid:25285017; PubMed Central PMCID: PMC4181629.
  9. 9. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–45. Epub 2013/09/21. pmid:24048066.
  10. 10. Xie K, Abbruzzese JL. Developmental biology informs cancer: the emerging role of the hedgehog signaling pathway in upper gastrointestinal cancers. Cancer Cell. 2003;4(4):245–7. Epub 2003/10/31. pmid:14585350.
  11. 11. Kho AT, Zhao Q, Cai Z, Butte AJ, Kim JY, Pomeroy SL, et al. Conserved mechanisms across development and tumorigenesis revealed by a mouse development perspective of human cancers. Genes Dev. 2004;18(6):629–40. Epub 2004/04/13. pmid:15075291; PubMed Central PMCID: PMC387239.
  12. 12. Liu HY, Kho AT, Kohane IS, Sun Y. Predicting survival within the lung cancer histopathological hierarchy using a multi-scale genomic model of development. Plos Medicine. 2006;3(7):1090–102. doi: Artn E232 ISI:000239493300028.
  13. 13. Kaiser S, Park YK, Franklin JL, Halberg RB, Yu M, Jessen WJ, et al. Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer. Genome Biol. 2007;8(7):R131. Epub 2007/07/07. pmid:17615082; PubMed Central PMCID: PMC2323222.
  14. 14. Nieto MA. Epithelial Plasticity: A Common Theme in Embryonic and Cancer Cells. Science. 2013;342(6159):708–+. ISI:000326647600033.
  15. 15. Eastham AM, Spencer H, Soncin F, Ritson S, Merry CLR, Stern PL, et al. Epithelial-mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res. 2007;67(23):11254–62. ISI:000251444100023. pmid:18056451
  16. 16. Ridolfi L, Petrini M, Fiammenghi L, Riccobon A, Ridolfi R. Human embryo immune escape mechanisms rediscovered by the tumor. Immunobiology. 2009;214(1):61–76. ISI:000263689600007. pmid:19159828
  17. 17. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11. pmid:11689955
  18. 18. Chen Y, Mcgee J, Chen XM, Doman TN, Gong XQ, Zhang YY, et al. Identification of Druggable Cancer Driver Genes Amplified across TCGA Datasets. Plos One. 2014;9(5). doi: ARTN e98293 ISI:000336790800032.
  19. 19. Bockmayr M, Klauschen F, Gyorffy B, Denkert C, Budczies J. New network topology approaches reveal differential correlation patterns in breast cancer. Bmc Systems Biology. 2013;7. doi: Artn 78 ISI:000324816800001.
  20. 20. Teschendorff AE, Gomez S, Arenas A, El-Ashry D, Schmidt M, Gehrmann M, et al. Improved prognostic classification of breast cancer defined by antagonistic activation patterns of immune response pathway modules. Bmc Cancer. 2010;10. doi: Artn 604 ISI:000284575500001.
  21. 21. Bagga S, Bracht J, Hunter S, Massirer K, Holtz J, Eachus R, et al. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell. 2005;122(4):553–63. ISI:000231555100011. pmid:16122423
  22. 22. Friedman RC, Farh KKH, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Research. 2009;19(1):92–105. ISI:000262200000009. pmid:18955434
  23. 23. Shukla GC, Singh J, Barik S. MicroRNAs: Processing, Maturation, Target Recognition and Regulatory Functions. Mol Cell Pharmacol. 2011;3(3):83–92. Epub 2011/01/01. pmid:22468167; PubMed Central PMCID: PMC3315687.
  24. 24. Ghildiyal M, Zamore PD. Small silencing RNAs: an expanding universe. Nature Reviews Genetics. 2009;10(2):94–108. ISI:000263091500011. pmid:19148191
  25. 25. Yang D, Sun Y, Hu L, Zheng H, Ji P, Pecot CV, et al. Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell. 2013;23(2):186–99. Epub 2013/02/16. pmid:23410973; PubMed Central PMCID: PMC3603369.
  26. 26. Hanahan D, Weinberg RA. Hallmarks of Cancer: The Next Generation. Cell. 2011;144(5):646–74. ISI:000288007100007. pmid:21376230
  27. 27. Montgomery RK, Mulberg AE, Grand RJ. Development of the human gastrointestinal tract: twenty years of progress. Gastroenterology. 1999;116(3):702–31. Epub 1999/02/25. pmid:10029630.
  28. 28. Stolte M. The new Vienna classification of epithelial neoplasia of the gastrointestinal tract: advantages and disadvantages. Virchows Archiv. 2003;442(2):99–106. pmid:12596058
  29. 29. Davila RE, Rajan E, Adler D, Hirota WK, Jacobson BC, Leighton JA, et al. ASGE guideline: the role of endoscopy in the diagnosis, staging, and management of colorectal cancer. Gastrointestinal endoscopy. 2005;61(1):1–7. pmid:15672048
  30. 30. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–4. Epub 2005/12/31. pmid:16381832; PubMed Central PMCID: PMC1347474.
  31. 31. Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP. MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell. 2007;27(1):91–105. Epub 2007/07/07. pmid:17612493; PubMed Central PMCID: PMC3800283.
  32. 32. Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, et al. Combinatorial microRNA target predictions. Nat Genet. 2005;37(5):495–500. Epub 2005/04/05. pmid:15806104.
  33. 33. Calle ML, Urrea V, Boulesteix AL, Malats N. AUC-RF: a new strategy for genomic profiling with random forest. Hum Hered. 2011;72(2):121–32. Epub 2011/10/15. pmid:21996641.
  34. 34. Liaw A, Wiener M. Classification and Regression by randomForest. R news. 2002;2(3):18–22.
  35. 35. Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC bioinformatics. 2011;12(1):77.
  36. 36. Carlson M. org. Hs. eg. db: Genome wide annotation for Human. R package version; 2013.
  37. 37. Killcoyne S, Carter GW, Smith J, Boyle J. Cytoscape: a community-based framework for network modeling. Methods Mol Biol. 2009;563:219–39. Epub 2009/07/15. pmid:19597788.
  38. 38. Hartwell KA, Muir B, Reinhardt F, Carpenter AE, Sgroi DC, Weinberg RA. The Spemann organizer gene, Goosecoid, promotes tumor metastasis. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(50):18969–74. ISI:000242884200019. pmid:17142318
  39. 39. Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nature Reviews Cancer. 2006;6(11):846–56. ISI:000241493800009. pmid:17060944
  40. 40. Liu SL, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, et al. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Research. 2006;66(12):6063–71. ISI:000238379500015. pmid:16778178
  41. 41. Kang HC, Wakabayashi Y, Jen KY, Mao JH, Zoumpourlis V, Del Rosario R, et al. Ptch1 overexpression drives skin carcinogenesis and developmental defects in K14Ptch(FVB) mice. J Invest Dermatol. 2013;133(5):1311–20. Epub 2012/12/12. pmid:23223138; PubMed Central PMCID: PMC3610826.
  42. 42. Kho AT, Zhao Q, Cai ZH, Butte AJ, Kim JYH, Pomeroy SL, et al. Conserved mechanisms across development and tumorigenesis revealed by a mouse development perspective of human cancers. Genes & Development. 2004;18(6):629–40. ISI:000220794200005.
  43. 43. Kaiser S, Park YK, Franklin JL, Halberg RB, Yu M, Jessen WJ, et al. Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer. Genome Biol. 2007;8(7). doi: Artn R131 ISI:000249416400011.
  44. 44. Hu H, Zhou L, Awadallah A, Xin W. Significance of Notch1-signaling pathway in human pancreatic development and carcinogenesis. Appl Immunohistochem Mol Morphol. 2013;21(3):242–7. Epub 2012/12/14. pmid:23235341.
  45. 45. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481(7381):306–13. Epub 2012/01/20. pmid:22258609; PubMed Central PMCID: PMC3367003.
  46. 46. Watson IR, Takahashi K, Futreal PA, Chin L. Emerging patterns of somatic mutations in cancer. Nat Rev Genet. 2013;14(10):703–18. Epub 2013/09/12. pmid:24022702.
  47. 47. Stratton MR. Exploring the genomes of cancer cells: progress and promise. Science. 2011;331(6024):1553–8. Epub 2011/03/26. pmid:21436442.
  48. 48. Wicha MS, Liu SL, Dontu G. Cancer stem cells: An old idea—A paradigm shift. Cancer Res. 2006;66(4):1883–90. ISI:000235387200001. pmid:16488983
  49. 49. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121(11):2373–80. Epub 2007/09/26. pmid:17893866.
  50. 50. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44. Epub 2008/07/25. pmid:18650914.
  51. 51. Bruttel VS, Wischhusen J. Cancer stem cell immunology: key to understanding tumorigenesis and tumor immune escape? Front Immunol. 2014;5:360. Epub 2014/08/15. pmid:25120546; PubMed Central PMCID: PMC4114188.
  52. 52. Ulrich C, Schmook T, Sachse MM, Sterry W, Stockfleth E. Comparative epidemiology and pathogenic factors for nonmelanoma skin cancer in organ transplant patients. Dermatol Surg. 2004;30(4 Pt 2):622–7. Epub 2004/04/06. pmid:15061846.
  53. 53. Andres A. Cancer incidence after immunosuppressive treatment following kidney transplantation. Crit Rev Oncol Hematol. 2005;56(1):71–85. Epub 2005/06/28. pmid:15978827.
  54. 54. Bower M, Palmieri C, Dhillon T. AIDS-related malignancies: changing epidemiology and the impact of highly active antiretroviral therapy. Current Opinion in Infectious Diseases. 2006;19(1):14–9. ISI:000235095300003. pmid:16374212
  55. 55. Dal Maso L, Serraino D, Franceschi S. Epidemiology of AIDS-related tumours in developed and developing countries. European Journal of Cancer. 2001;37(10):1188–201. ISI:000169648500003. pmid:11423251
  56. 56. Farraye FA, Odze RD, Eaden J, Itzkowitz SH. AGA technical review on the diagnosis and management of colorectal neoplasia in inflammatory bowel disease. Gastroenterology. 2010;138(2):746–74, 74 e1–4; quiz e12-3. Epub 2010/02/10. pmid:20141809.
  57. 57. Chen GY, Nunez G. Inflammasomes in intestinal inflammation and cancer. Gastroenterology. 2011;141(6):1986–99. Epub 2011/10/19. pmid:22005480; PubMed Central PMCID: PMC3442608.
  58. 58. Boysen J, Sinha S, Price-Troska T, Warner SL, Bearss DJ, Viswanatha D, et al. The tumor suppressor axis p53/miR-34a regulates Axl expression in B-cell chronic lymphocytic leukemia: implications for therapy in p53-defective CLL patients. Leukemia. 2014;28(2):451–5. Epub 2013/11/13. pmid:24217154; PubMed Central PMCID: PMC3929965.
  59. 59. Mudduluru G, Ceppi P, Kumarswamy R, Scagliotti GV, Papotti M, Allgayer H. Regulation of Axl receptor tyrosine kinase expression by miR-34a and miR-199a/b in solid cancer. Oncogene. 2011;30(25):2888–99. Epub 2011/02/15. pmid:21317930.
  60. 60. Dunne PD, Mcart DG, Blayney JK, Kalimutho M, Greer S, Wang TT, et al. AXL Is a Key Regulator of Inherent and Chemotherapy-Induced Invasion and Predicts a Poor Clinical Outcome in Early-Stage Colon Cancer. Clinical Cancer Research. 2014;20(1):164–75. ISI:000329303100019. pmid:24170546
  61. 61. Mees ST, Mardin WA, Wendel C, Baeumer N, Willscher E, Senninger N, et al. EP300—a miRNA-regulated metastasis suppressor gene in ductal adenocarcinomas of the pancreas. Int J Cancer. 2010;126(1):114–24. Epub 2009/07/02. pmid:19569050.
  62. 62. Kim MS, Lee SH, Yoo NJ. Frameshift mutations of tumor suppressor gene EP300 in gastric and colorectal cancers with high microsatellite instability. Hum Pathol. 2013;44(10):2064–70. Epub 2013/06/14. pmid:23759652.
  63. 63. Pham TT, Angus SP, Johnson GL. MAP3K1: Genomic Alterations in Cancer and Function in Promoting Cell Survival or Apoptosis. Genes Cancer. 2013;4(11–12):419–26. Epub 2014/01/05. pmid:24386504; PubMed Central PMCID: PMC3877667.
  64. 64. Shang J, Yang F, Wang Y, Wang Y, Xue G, Mei Q, et al. MicroRNA-23a Antisense Enhances 5-Fluorouracil Chemosensitivity Through APAF-1/Caspase-9 Apoptotic Pathway in Colorectal Cancer Cells. J Cell Biochem. 2014;115(4):772–84. pmid:24249161
  65. 65. Yong FL, Law CW, Wang CW. Potentiality of a triple microRNA classifier: miR-193a-3p, miR-23a and miR-338-5p for early detection of colorectal cancer. BMC Cancer. 2013;13:280. Epub 2013/06/14. pmid:23758639; PubMed Central PMCID: PMC3691634.
  66. 66. Yamaguchi T, Iijima T, Wakaume R, Takahashi K, Matsumoto H, Nakano D, et al. Underexpression of miR-126 and miR-20b in Hereditary and Nonhereditary Colorectal Tumors. Oncology. 2014;87(1):58–66. ISI:000338974700007. pmid:24994098
  67. 67. Fang LK, Li HR, Wang L, Hu J, Jin TR, Wang JP, et al. MicroRNA-17-5p promotes chemotherapeutic drug resistance and tumour metastasis of colorectal cancer by repressing PTEN expression. Oncotarget. 2014;5(10):2974–87. ISI:000336967000009. pmid:24912422
  68. 68. Ma YL, Zhang P, Wang F, Zhang HZ, Yang YZ, Shi CZ, et al. Elevated oncofoetal miR-17-5p expression regulates colorectal cancer progression by repressing its target gene P130. Nature Communications. 2012;3. doi: Artn 1291 ISI:000316356700058.