Open Access

Targeted molecular profiling of genetic alterations in colorectal cancer using next‑generation sequencing

  • Authors:
    • Jia Luo
    • Shengjun Zhang
    • Meihua Tan
    • Jia Li
    • Huadong Xu
    • Yanfei Tan
    • Yue Huang
  • View Affiliations

  • Published online on: December 10, 2019     https://doi.org/10.3892/ol.2019.11203
  • Pages: 1137-1144
  • Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Colorectal cancer (CRC) is a major contributor to cancer‑associated mortality in China and remains a vast challenge worldwide. Although the genetic basis of CRC has been investigated, the uncommonly mutated genes in CRC remain unknown, in particular in the Asian population. In the present study, targeted region sequencing on 22 CRC and 10 paired non‑cancerous tissues was performed to determine the genetic pattern of CRC samples in the Chinese population. Driver genes were detected by three distinct softwares, including MutSigCV, oncodriveFM and iCAGES. A total of 1,335 reliable somatic mutations were identified in tumour samples compared with normal samples. Furthermore, mismatch repair (MMR) mutant patients presented significantly higher mutation density compared with MMR wild‑type patients. The results from MutSigCV, oncodriveFM and iCAGES analyses simultaneously detected 29 unique driver genes. In addition, the genes APC regulator of WNT signaling pathway, SMAD family member 4, neurofibromin 1, AT‑rich interaction domain 5B and nuclear receptor corepressor 1 were the top five most frequently mutated genes in CRC samples, with mutation rates of 68, 36, 36, 32 and 27%, respectively. The findings from the present study may therefore serve as a basis for future investigation on the diagnosis and oncogenesis of CRC.

Introduction

Colorectal cancer (CRC) is the fourth most frequently diagnosed cancer and the second leading cause of cancer-associated mortality worldwide according to the global cancer statistics from 2018 (1). In the United States, approximately 135,430 new cases are diagnosed each year and approximately 50,260 patients die of CRC, which accounts for approximately 9% of all cancer-associated mortalities (2). In 2015, CRC was considered as the fifth most common type of cancer in terms of occurrence and mortality in China (3). Cancer stage is a major factor contributing to the overall survival of patients with CRC. The 3-year survival rate is 80–90% for patients with CRC who are diagnosed with stages I or II, whereas the 3-year survival rate is reduced to 20% for patients with CRC diagnosed with stage IV (4).

Over the past decade, large-scale sequencing studies have investigated the genetic basis of CRC and uncovered key pathways involved in the pathogenesis of CRC, including WNT, RAS-MAPK, PI3K, TGF-β, P53 and DNA mismatch repair pathways (58). The Cancer Genome Atlas program conducted a genome-scale analysis of 276 CRC samples using multi-omics sequencing results and discovered that 16% of hypermutated CRC samples are characterized by high microsatellite instability, hypermethylation, mutL homolog 1 (MLH1) silencing or somatic mutations in mismatch repair genes and polymerases. In addition to the known mutations in the genes APC regulator of WNT signaling pathway (APC), tumor protein 53 (TP53), SMAD family member 4 (SMAD4), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha and KRAS proto-oncogene GTPase, 19 new recurrently mutated genes have been detected, including AT-rich interaction domain 1A (ARID1A), SRY-box transcription factor 9 (SOX9) and APC membrane recruitment protein 1, and amplifications of the genes erb-b2 receptor tyrosine kinase 2 and insulin like growth factor 2 have been identified (9).

Although our understanding of genomic alterations of oncology has rapidly improved (59), the uncommonly mutated genes in CRC, especially in the Asian population, remain unknown. In the present study, targeted region sequencing (508 genes) on 22 CRC and 10 paired non-cancerous samples from patients with CRC was performed to study genetic changes. The conclusion of this study may provide further insight into genomic changes in CRC and determine potential therapeutic targets in CRC.

Materials and methods

Patients and samples

A total of 22 primary CRC and 10 paired normal colorectal tissues were surgically resected at Sanming Hospital between May 2014 and August 2015 and immediately frozen. Patients with secondary CRC were excluded from the present study. The mean age of patients at diagnosis was 66.7±10.3 years and ranged between 35 and 80 years old. Written informed consent was obtained from all patients and the study was approved by the Ethics Review Board of the Sanming First Hospital. The clinical characteristics of patients are presented in Table I.

Table I.

Clinical and demographic characteristics of the 22 patients with CRC.

Table I.

Clinical and demographic characteristics of the 22 patients with CRC.

VariableCRC (n=22)
Sex
  Male9
  Female13
Tumor/adenoma/Polyp location
  Rectum7
  Sigmoideum1
  Ascending colon7
  Transverse colon1
  Rectosigmoid1
  Descending colon5
Pathologic diagnosis
  Protruding adenocarcinoma14
  Protruding and mucinous adenocarcinoma7
  Protruding moderately differentiated adenocarcinoma1
Pathologic stage
  I6
  II11
  III3
  IV2
Tumor stage
  T16
  T212
  T32
  T42
Nodal stage
  N018
  N11
  N23
Metastasis stage
  M020
  M12
DNA extraction, library preparation and sequencing

Genomic DNA was extracted from the 22 CRC and 10 paired normal colorectal tissues using the QIAamp DNA Blood Midi Kit (Qiagen GmbH) according to the manufacturer's protocol. A NanoDrop 2000 (NanoDrop Technologies; Thermo Fisher Scientific, Inc.) was used to assess DNA quality and concentration. Total DNA samples (400 ng) were used to construct sequencing libraries following the improved protocols for Illumina sequencing (10). In brief, genomic DNA samples were fragmented and ligated with Illumina standard adapters to both fragments ends. Adapter-ligated fragments are then PCR-amplified and gel-purified. Libraries were pooled (11) and hybridized using the Oseq-T panel (508 genes; BGI Genomics Co. Ltd.) for capturing. The process of hybridization was performed as previously described (12). The hybridized product was sequenced using the Illumina MiSeq platform (Illumina, Inc.) using a 100 paired-end (PE) sequencing strategy.

Variant calling and filtering

High-quality reads were aligned to the human reference genome 19 (http://hgdownload.soe.ucsc.edu/goldenPath/hg19/chromosomes/) using MEM algorithms of Burrows-Wheeler-Aligner v.0.7.17 (http://bio-bwa.sourceforge.net) with default parameters, except that the number of threads was set to 2 to accelerate this mapping process (-t 2) (13). Read deduplication was performed utilizing Picard v.1.98 software (http://picard.sourceforge.net/), using the main parameters as follows: ASSUME_SORTED=true and VALIDATION_STRINGENCY=LENIENT. Local realignment around insertion-deletions were performed using GATK v3.3.0 RealignerTargetCreator and IndelRealigner model (https://software.broadinstitute.org/gatk/) (14). Base quality score recalibration was also performed by GATK v3.3.0 BaseRecalibrator and PrintReads model (https://software.broadinstitute.org/gatk/) (13). Single nucleotide variant and insertion-deletion (indel) mutations were detected by 3.3.0 HaplotypeCaller of GATK (https://software.broadinstitute.org/gatk/). New variants and variants with maximum allele frequency <1% reported in the 1000 genome project (15) and dbsnp v141 database (16) were considered as high confidence variants. High confidence variants that were detected specifically in CRC samples but not in normal colon tissues were regarded as somatic mutations. The SnpEff tool (http://snpeff.sourceforge.net/SnpEff_manual.html) was applied to annotate somatic mutations (17). The final somatic variants and annotation results were used in the downstream analysis.

Driver gene prediction and functional annotation

Driver genes were predicted using three distinct computational tools, including MutSigCV (v1.0) (https://cloud.genepattern.org/gp/pages/index.jsf) (18), oncodriveFM (v0.0.1) (https://www.intogen.org) (19) and Integrated CAncer GEnome Score (iCAGES) (http://icages.wglab.org) (20). All parameters were set to default values. Driver genes were determined according to the following criteria: i) Genes with q-value <0.1 in the MutSigCV analysis (21); ii) genes with q-value less <0.05 in the oncodriveFM analysis (19); and iii) genes predicted to be drivers by iCAGES, with icagesGeneScores of predicted drivers >0.5 (22). To functionally annotate these driver genes, the Database for Annotation, Visualization and Integrated Discovery (https://david.ncifcrf.gov) (23) was applied to analyse the significant Gene Ontology (GO; http://david.ncifcrf.gov) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://david.ncifcrf.gov) (24) pathways in which driver genes were enriched. Bonferroni adjusted P-value <0.05 indicated a statistical significance.

Copy number variation analysis

Focal copy number variations (CNVs) were detected using CONTRA v2.0.8 software (25) between CRC and paired normal samples using default parameters, (eg: contra.py -t *.bed -s *tumor.bam -c *normal.bam -f hg19.fasta -o output-removeDups), except for ‘-removeDups’. For CRC samples without paired normal tissues, deduplicated bam files of 10 normal samples were merged to form one bam file. Focal CNVs were detected between deduplicated bam files of CRC samples and the merged bam file of 10 normal samples. All parameters were set to default values. Focal CNVs with P-values <0.05 were considered statistically significant. Fraction of CNV (FCNV) was computed as follows: FCNV=(size of significant CNVs)/size of exons.

Statistical analyses

In the MutSigCV analysis, P-value was calculated for the gene by convoluting the background distributions of all the mutation types, and determining the probability of meeting or exceeding that score by background mutation alone. Benjamini-Hochberg false discovery rate procedure was used to correct for P-values of multiple tests. In the oncodrive FM analysis, the method randomly sampled one milliongroups of the same number of observed mutations. P-value referred to the fraction of functional impact scores equal to or greater than the observed average functional impact score of the gene. Bonferroni correction was applied to calculate the q-value for each gene. The Bonferroni correction was used to adjust the P-values computed by Fisher's exact test in the GO and KEGG pathway enrichment analyses. Difference of quantitative values was compared between two groups using Wilcoxon rank-sum test in R 3.2.0 (www.r-project.org).

Results

Clinical and demographic characteristics of the 22 patients with CRC

The 22 patients with CRC comprised 13 female and 9 male patients. The primary sites of CRC included rectum (7 cases), sigmoideum (1 case), ascending colon (7 cases), transverse colon (1 case), rectosigmoid (1 case) and descending colon (5 cases). The pathological types of the 22 patients comprised 14 cases of protruding adenocarcinoma, 7 cases of protruding and mucinous adenocarcinoma and 1 case of protruding moderately differentiated adenocarcinoma. In addition, 6, 11, 3 and 2 patients with CRC were diagnosed with stages I, II, III and IV, respectively. Furthermore, 2 and 4 patients presented positive metastasis in lymph nodes and distant organs, respectively (Table I).

Summary statistics of alignment to target regions

A 3.65 Mb target region comprising 508 genes was captured from genomic DNA and used to perform variant calling. On average, 98.87% reads were mapped successfully. The duplicated reads were removed, resulting in an average of 4,312,287 (425.73 Mb) effective reads. Of the total effective bases, 69.95% was mapped to target regions (capture specificity), and the mean sequencing depth in the target regions was 81.57-fold. On average, 99.37% of targeted bases had at least 1× coverage, and 97.15% of the targeted bases had at least 10× coverage (Table SI). In addition, the distributions of per-base sequencing depth, cumulative sequencing depth and insert size are presented in Fig. 1. The density of sequencing depth on the target region followed normal distribution, with a mean of 100X. As expected, the peak of the insertion size distribution of the sequencing library was ~200 bp.

Somatic mutations in colorectal cancer

To characterize the mutational spectrum, a targeted region sequencing (3.65 Mb) on the 22 CRC and 10 paired normal tissues was performed. A total of 1,335 somatic mutations were identified, including 122 missense mutations, 48 silent variants, 48 splicing site variants, 13 inframe deletions and 8 inframe insertions. Furthermore, 513, 404 and 113 variants were located in the untranslated region, intron and noncoding regions, respectively. In total, 52 variants caused a frame shift and 15 variants led to stop gain (Fig. 2A). The somatic mutation rates varied considerably among the samples. The average mutation density was 16.63 mutations/Mb in the 508-gene panel (range, 4.38–69.04 mutations/Mb). To analyse the cause for the different mutation densities, the mutation statuses in DNA mismatch repair (MMR) pathway genes was assessed, including MLH1, MLH3, mutS homolog 2 (MSH2), MSH3, MSH6 and PMS1 homolog 2. Five patients with CRC had mutations in any of the MMR genes, and the average mutation density in MMR mutant patients was significantly higher compared with MMR wild-type patients (31.73 vs. 12.18 mutations/Mb; P=0.01; Wilcoxon rank sum test; Fig. 2B). In addition, indels, C>T/G>A and A>G/T>C were the three most prevalent mutation types with mutation rates of 48, 18. 8, and 12.8%, respectively (Fig. 2C).

Driver genes in colorectal cancer

MutSigCV analysis identified four recurrently mutated genes, named APC, TP53, SUZ12 polycomb repressive complex 2 subunit (SUZ12) and AT-rich interaction domain 5B (ARID5B), with significant statistical evidence (q-value <0.2). Furthermore, OncodriveFM and iCAGES detected five and 24 driver genes, respectively. In total, 29 unique driver genes were detected using these three computational tools. TP53 was the overlapping gene among the three sets of driver genes. APC, SMAD4, neurofibromin 1 (NF1), ARID5B and nuclear receptor corepressor 1 (NCOR1) were the top five most frequently mutated genes in patients with CRC with mutation rates of 68, 36, 36, 32 and 27%, respectively (Fig. 3). In addition, the tumour-suppressor genes APC and SMAD4 had an excessively high number of frameshift or nonsense mutations. The average mutation rate was 18% in the 29 driver genes (Table SII). Certain driver genes, including mitogen-activated protein kinase kinase kinase 1 (MAP3K1), FGFR4 and PDGFR-β, were mutated at low frequencies (5% for all three genes), and firstly reported as driver genes in CRC samples.

To functionally annotate the 29 driver genes, GO term and KEGG pathway enrichment analyses were performed. The results from GO enrichment analysis demonstrated that the 29 driver genes were significantly enriched in 152 GO terms, including ‘positive regulation of cell proliferation’, ‘phosphatidylinositol-mediated signalling’, ‘cellular response to DNA damage stimulus’ and ‘MAPK cascade’ (Bonferroni-adjusted P<0.05; data not shown). Furthermore, the driver genes were significantly enriched in 9 KEGG pathways, including ‘Wnt signalling pathways’, ‘cell cycle’, ‘colorectal cancer’, ‘melanoma, glioma’, ‘pancreatic and endometrial cancer’ (Bonferroni-adjusted P<0.05; Table SIII).

Copy number variation analysis in CRC samples

To further analyse the copy number variation in driver genes, CONTRA was used to detect focal CNVs at the exon level. The results demonstrated that 12 driver genes had FCNV >20% in all CRC sample, including GNAS complex locus (GNAS), BRCA2 DNA repair associated (BRCA2), SOX9, fibroblast growth factor receptor 4 (FGFR4), SMAD4, SUZ12, FGFR3, epidermal growth factor receptor (EGFR), E1A binding protein p300, NCOR1, TP53 and insulin like growth factor 1 receptor. In particular, GNAS presented a significant focal gain >20% in 45% (10/22) of CRC samples compared with normal samples. In addition, tumour suppressor genes, including TP53, presented a significant focal loss >20% in 22.7 (5/22) and 4.5% (1/22) of CRC samples compared with normal samples. The EGFR oncogene exhibited a significant focal gain >20% in 9.1% (2/22) of CRC samples compared with normal samples (Table II).

Table II.

Fraction of significant CNVs of driver genes in colorectal cancer samples.

Table II.

Fraction of significant CNVs of driver genes in colorectal cancer samples.

GenesSampleFraction of CNVCNV type
GNAS13CB0.54Gain
GNAS15CB0.55Gain
GNAS16CB0.86Gain
GNAS18CB0.56Gain
GNAS19CB0.56Gain
GNAS20CB0.23Gain
GNAS21CB0.65Gain
GNAS31CB0.33Gain
GNAS36CB0.56Gain
GNAS41CB0.93Gain
BRCA217CB0.58Gain
BRCA228CB0.88Gain
BRCA230CB0.56Gain
BRCA232CB0.49Loss
BRCA235CB0.48Loss
SMAD419CB0.31Loss
SMAD430CB0.27Loss
SMAD431CB0.22Loss
SMAD440CD0.22Loss
SOX917CB0.29Loss
SOX918CB0.83Gain
SUZ1219CB0.23Loss
SUZ1230CB0.22Gain
FGFR311CB0.20Gain
FGFR318CB0.22Gain
EGFR16CB0.73Gain
EGFR31CB0.33Gain
EP30016CB0.42Gain
NCOR119CB0.23Loss
TP5339CB0.27Loss
IGF1R42CB0.25Loss
FGFR419CB0.20Gain

[i] BRCA2, BRCA2 DNA repair associated; CNV, copy number variation; EGFR, epidermal growth factor receptor; EP300, E1A binding protein p300; FGFR3, fibroblast growth factor receptor 3; FGFR4, fibroblast growth factor receptor 4; GNAS, GNAS complex locus; IGF1R, insulin like growth factor 1 receptor; NCOR1, nuclear receptor corepressor 1; SMAD4, SMAD family member 4; SOX9, SRY-box transcription factor 9; SUZ12, SUZ12 polycomb repressive complex 2 subunit; TP53, tumor protein p53.

Discussion

The activation of driver mutations might serve a crucial role in cancer development (26). Genes that carry these driver mutations are considered as driver genes and are essential to tumorigenesis (2731). The most common approach to predict driver genes in numerous cancer samples is the identification of genes that present significantly higher mutation frequencies compared with the background mutation rate (18,32), via the MutSigCV model for example. However, many driver genes may occur at a low frequency. (<1%) in tumours (8). Novel computational tools have therefore been developed to detect driver genes with middle or low mutation frequencies. OncodriveFM first evaluates the functional impact of a somatic mutation using the three different tools SIFT (33), PolyPhen2 (34) and MutationAssessor (35), and applies the transFIC method (http://bg.upf.edu/transfic) to transform the three functional scores into a uniform score (36). To identify driver genes, oncodriveFM (19) compares the actual functional impact with a null distribution model generated by 1,000,000 permutations and computes the bias towards the accumulation of variants with high functional impact.

The iCAGES is a novel statistical framework that can predict driver variants by integrating contributions from coding, noncoding and structural variants. iCAGES can therefore identify driver genes by combining genomic information and biological knowledge to generate prioritized drug treatments (20). iCAGES consists of three consecutive layers. The first layer prioritizes personalized cancer driver coding, noncoding and structural variations. The second layer associates these mutations to genes using a statistical model with prior biological knowledge of cancer driver genes for specific subtypes of cancer. The third layer generates a list of drugs targeting the repertoire of these potential driver genes. Three computational tools, including MutSigCV, oncodriveFM and iCAGES, were applied to detect driver genes based on complementary principles independently of somatic mutation recurrence. The combination of the three tools enables the detection of recurrently and rarely mutated driver genes in a more comprehensive manner than with MutSigCV alone (9).

Over the last decade, large-scale sequencing projects have been completed to characterize the mutation profiling of CRC (9,37,38). Recurrent gene mutations, including APC, KRAS proto-oncogene, GTPase and titin genes, and dysregulated signalling pathways, including the Wnt, tumour growth factor-β, phosphoinositide 3-kinase and P53 signalling pathways, have been reported to contribute to CRC carcinogenesis (9,37,38). Giannakis et al (37) identified recurrently mutated genes in CRC, including BCL9 like, RNA binding motif protein 10, CCCTC-binding factor and Kruppel like factor 5, which were not previously reported in CRC. R-spondin (RSPO) gene fusions are present in 10% of colon tumours and are mutually exclusive with APC mutations, which suggests that they might be involved in the activation of Wnt signalling pathway (38). In the present study, the use of three computational algorithms, including MutSigCV, oncodriveFM and iCAGES allowed the detection of 29 driver genes on 22 CRC samples using somatic mutations. The results demonstrated that APC, SMAD4, NF1, ARID5B and TP53 genes were frequently mutated in patients with CRC, which was similar to results from a previous study (9). Among the 29 driver genes, EGFR, kinase insert domain receptor and platelet derived growth factor receptor beta (PDGFR-β) are oncogenes, whereas von Hippel-Lindau tumor suppressor, NF1, SUZ12, BRCA1 and BRCA2 are tumour suppressor genes, according to the curated oncogene (39) and tumour suppressor gene (40) databases. In particular, certain driver genes, including mitogen-activated protein kinase kinase kinase 1 (MAP3K1), FGFR4 and PDGFR-β, had low mutation frequencies. To the best of our knowledge, these three genes were reported as driver genes in CRC samples for the first time. MAP3K1 is a Ser/Thr protein kinase that belongs to the mitogen-activated protein kinase kinase kinase 1 (MEKK)/serine/threonine protein kinase subgroup of the MEKK family. Silencing MAP3K1 expression significantly enhances paclitaxel-induced cell proliferation inhibition in breast cancer cells, (41) pancreatic cancer cells (42) and medulloblastoma cells (43) and inhibits the human pancreatic cancer cell invasive and migratory abilities (43). In accordance to previous studies (4144), this study suggests that MAP3K1 may have oncogenic role in cancers. It has been demonstrated that FGFR4 serves the role of oncogene and regulator of drug or radiation resistance in CRC (45). Furthermore, FGFR4 silencing can inhibit colon cancer cell line proliferation and induces caspase-dependent apoptosis (45). In addition, FGFR4 can regulate FLICE-like inhibitory protein expression via Signal transducer and activator of transcription 3, conferring resistance to 5-fluorouracil (5-FU) and oxaliplatin chemotherapy in colon cancer cell lines (45). Furthermore, it has been reported that FGFR induces resistance to radiation therapy in CRC by downregulating RAD51 recombinase level and inhibiting cancer cell proliferation (46). PDGFR-β is a cell surface tyrosine kinase receptor for members of the PDGF family (47). It has been demonstrated that PDGFR-β high expression is positively correlated with lymphatic metastasis and advanced UICC stages, and that PDGFR-β high expression might be considered as a negative factor for the prognosis of patients with colon cancer (4850). Decreased expression of PDGFR-β can inhibit the invasion and proliferation of CRC cell lines (HCT116 and DLD-1) in a dose-dependent manner (48). PDGFR-β expression may therefore be considered as a risk factor for recurrence in CRC, suggesting that PDGFR inhibitor could represent a useful therapeutic agent for CRC (48). These newly identified driver genes may be considered as novel candidates that could be used for functional validation in future investigation.

In conclusion, the present study screened over 500 genes in order to analyse the genetic alterations present in 22 CRC samples. A total of 29 driver genes were identified. Driver genes with significant copy number variations, such as GNAS and TP53, may be crucial in CRC oncogenesis. These findings discoveries may serve as basis for further investigation on CRC diagnosis and oncogenesis.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Mr. Licheng Cai and Mr. Yanyang Si from BGI-Shenzhen for their contribution on sample collection and project management.

Funding

The study was supported by the Natural Science Foundation of Fujian Province (grant nos. 2015J01568 and 2016J01656.

Availability of data and materials

The raw sequencing data that support the findings of this study have been deposited in the CNSA (https://db.cngb.org/cnsa/) of CNGBdb (accession number, CNP0000601: published on 2019/09/30).

Author's contributions

YH designed the entire study. HX, YT and SZ conducted library preparation and targeted region sequencing. JLuo, SZ, JLi and MT were responsible for the bioinformatics analyses, manuscript preparation and manuscript revision. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

This study was approved by the Ethics Review Board of the Sanming First Hospital [SFHRB(2014) No 3] and all patients provided informed consent prior to the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CNV

copy number variation

CRC

colorectal cancer

GO

Gene ontology

iCAGES

Integrated CAncer GEnome Score

KEGG

Kyoto Encyclopedia of Genes and Genomes

MMR

mismatch repair

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–32. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Benitez Majano S, Di Girolamo C, Rachet B, Maringe C, Guren MG, Glimelius B, Iversen LH, Schnell EA, Lundqvist K, Christensen J, et al: Surgical treatment and survival from colorectal cancer in Denmark, England, Norway, and Sweden: A population-based study. Lancet Oncol. 20:74–87. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Fearon ER: Molecular genetics of colorectal cancer. Annu Rev Pathol. 6:479–507. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Bass AJ, Lawrence MS, Brace LE, Ramos AH, Drier Y, Cibulskis K, Sougnez C, Voet D, Saksena G, Sivachenko A, et al: Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat Genet. 43:964–968. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Sjöblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ, Ptak J, Silliman N, et al: The consensus coding sequences of human breast and colorectal cancers. Science. 314:268–274. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, et al: The genomic landscapes of human breast and colorectal cancers. Science. 318:1108–1113. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Bronner IF, Quail MA, Turner DJ and Swerdlow H: Improved protocols for Illumina sequencing. Curr Protoc Hum Genet. 80:18.2.1–42. 2014. View Article : Google Scholar

11 

Wei X, Sun Y, Xie J, Shi Q, Qu N, Yang G, Cai J, Yang Y, Liang Y, Wang W and Yi X: Next-generation sequencing identifies a novel compound heterozygous mutation in MYO7A in a Chinese patient with Usher Syndrome 1B. Clin Chim Acta. 413:1866–1871. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Shao D, Lin Y, Liu J, Wan L, Liu Z, Cheng S, Fei L, Deng R, Wang J, Chen X, et al: A targeted next-generation sequencing method for identifying clinically relevant mutation profiles in lung adenocarcinoma. Sci Rep. 6:223382016. View Article : Google Scholar : PubMed/NCBI

13 

Li H and Durbin R: Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 25:1754–1760. 2009. View Article : Google Scholar : PubMed/NCBI

14 

McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M and DePristo MA: The genome analysis toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20:1297–1303. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Gibbs RA, Boerwinkle E, Doddapaneni H, Zhu H, Alkan C, Dal E, Kahveci F, Garrison EP, Kural D, Lee WP and Dermitzakis ET: A global reference for human genetic variation. Nature. 526:68–74. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Sherry ST, Ward MH, Kholodov M, Baker J, Phan L, Smigielski EM and Sirotkin K: dbSNP: The NCBI database of genetic variation. Nucleic Acids Res. 29:308–311. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, Land SJ, Lu X and Ruden DM: A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 6:80–92. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, et al: Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 499:214–218. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Gonzalez-Perez A and Lopez-Bigas N: Functional impact bias reveals cancer drivers. Nucleic Acids Res. 40:e1692012. View Article : Google Scholar : PubMed/NCBI

20 

Dong C, Guo Y, Yang H, He Z, Liu X and Wang K: iCAGES: Integrated CAncer GEnome Score for comprehensively prioritizing driver genes in personal cancer genomes. Genome Med. 8:1352016. View Article : Google Scholar : PubMed/NCBI

21 

Tokheim CJ, Papadopoulos N, Kinzler KW, Vogelstein B and Karchin R: Evaluating the evaluation of cancer driver genes. Proc Natl Acad Sci USA. 113:14330–14335. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Zhao X, Lei Y, Li G, Cheng Y, Yang H, Xie L, Long H and Jiang R: Integrative analysis of cancer driver genes in prostate adenocarcinoma. Mol Med Rep. 19:2707–2715. 2019.PubMed/NCBI

23 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Kanehisa M, Furumichi M, Tanabe M, Sato Y and Morishima K: KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45(D1): D353–D361. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Li J, Lupat R, Amarasinghe KC, Thompson ER, Doyle MA, Ryland GL, Tothill RW, Halgamuge SK, Campbell IG and Gorringe KL: CONTRA: Copy number analysis for targeted resequencing. Bioinformatics. 28:1307–1313. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, et al: Patterns of somatic mutation in human cancer genomes. Nature. 446:153–158. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van Allen E, Stransky N, et al: Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 44:685–689. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, et al: The mutational landscape of lethal castration-resistant prostate cancer. Nature. 487:239–243. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Collisson EA, Campbell JD, Brooks AN, Berger AH, Lee W, Chmielecki J, Beer DG, Cope L, Creighton CJ, Danilova L, et al: Comprehensive molecular profiling of lung adenocarcinoma. Nature. 511:543–550. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, Shimamura T, Sato-Otsubo A, Nagae G, Suzuki H, et al: Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 45:860–867. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Cancer Genome Atlas Research Network, . Integrated genomic characterization of papillary thyroid carcinoma. Cell. 159:676–690. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Dees ND, Zhang Q, Kandoth C, Wendl MC, Schierding W, Koboldt DC, Mooney TB, Callaway MB, Dooling D, Mardis ER, et al: MuSiC: Identifying mutational significance in cancer genomes. Genome Res. 22:1589–1598. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Sim NL, Kumar P, Hu J, Henikoff S, Schneider G and Ng PC: SIFT web server: Predicting effects of amino acid substitutions on proteins. Nucleic Acids Res. 40((Web Server Issue)): W452–W457. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS and Sunyaev SR: A method and server for predicting damaging missense mutations. Nat Methods. 7:248–249. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Reva B, Antipin Y and Sander C: Predicting the functional impact of protein mutations: Application to cancer genomics. Nucleic Acids Res. 39:e1182011. View Article : Google Scholar : PubMed/NCBI

36 

González-Pérez A and López-Bigas N: Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel. Am J Hum Genet. 88:440–449. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Giannakis M, Mu XJ, Shukla SA, Qian ZR, Cohen O, Nishihara R, Bahl S, Cao Y, Amin-Mansour A, Yamauchi M, et al: Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 15:857–865. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, Chaudhuri S, Guan Y, Janakiraman V, Jaiswal BS, et al: Recurrent R-spondin fusions in colon cancer. Nature. 488:660–664. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Liu Y, Sun J and Zhao M: ONGene: A literature-based database for human oncogenes. J Genet Genomics. 44:119–121. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Zhao M, Sun J and Zhao Z: TSGene: A web resource for tumor suppressor genes. Nucleic Acids Res. 41((Database Issue)): D970–D976. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Hu P, Huang Q, Li Z, Wu X, Ouyang Q, Chen J and Cao Y: Silencing MAP3K1 expression through RNA interference enhances paclitaxel-induced cell cycle arrest in human breast cancer cells. Mol Biol Rep. 41:19–24. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Hirano T, Shino Y, Saito T, Komoda F, Okutomi Y, Takeda A, Ishihara T, Yamaguchi T, Saisho H and Shirasawa H: Dominant negative MEKK1 inhibits survival of pancreatic cancer cells. Oncogene. 21:5923–5928. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Antonucci L, Di Magno L, D'Amico D, Manni S, Serrao SM, Di Pastena F, Bordone R, Yurtsever ZN, Caimano M, Petroni M, et al: Mitogen-activated kinase kinase kinase 1 inhibits hedgehog signaling and medulloblastoma growth through GLI1 phosphorylation. Int J Oncol. 54:505–514. 2019.PubMed/NCBI

44 

Su F, Li H, Yan C, Jia B, Zhang Y and Chen X: Depleting MEKK1 expression inhibits the ability of invasion and migration of human pancreatic cancer cells. J Cancer Res Clin Oncol. 135:1655–1663. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Turkington RC, Longley DB, Allen WL, Stevenson L, McLaughlin K, Dunne PD, Blayney JK, Salto-Tellez M, Van Schaeybroeck S and Johnston PG: Fibroblast growth factor receptor 4 (FGFR4): A targetable regulator of drug resistance in colorectal cancer. Cell Death Dis. 5:e10462014. View Article : Google Scholar : PubMed/NCBI

46 

Ahmed MA, Selzer E, Dörr W, Jomrich G, Harpain F, Silberhumer GR, Müllauer L, Holzmann K, Grasl-Kraupp B, Grusch M, et al: Fibroblast growth factor receptor 4 induced resistance to radiation therapy in colorectal cancer. Oncotarget. 7:69976–69990. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Heldin CH and Lennartsson J: Structural and functional properties of platelet-derived growth factor and stem cell factor receptors. Cold Spring Harb Perspect Biol. 5:a0091002013. View Article : Google Scholar : PubMed/NCBI

48 

Fujino S, Miyoshi N, Ohue M, Takahashi Y, Yasui M, Hata T, Matsuda C, Mizushima T, Doki Y and Mori M: Platelet-derived growth factor receptor-β gene expression relates to recurrence in colorectal cancer. Oncol Rep. 39:2178–2184. 2018.PubMed/NCBI

49 

Mezheyeuski A, Bradic Lindh M, Guren TK, Dragomir A, Pfeiffer P, Kure EH, Ikdahl T, Skovlund E, Corvigno S, et al: Survival-associated heterogeneity of marker-defined perivascular cells in colorectal cancer. Oncotarget. 7:41948–41958. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Schimanski C, Wehler T, Galle P, Gockel I and Moehler M: PDGFR-α/β expression correlates with the metastatic behavior of human colorectal cancer-A rationale for a molecular targeting strategy? J Clin Oncol. 26:220192008. View Article : Google Scholar

Related Articles

Journal Cover

February-2020
Volume 19 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo J, Zhang S, Tan M, Li J, Xu H, Tan Y and Huang Y: Targeted molecular profiling of genetic alterations in colorectal cancer using next‑generation sequencing. Oncol Lett 19: 1137-1144, 2020
APA
Luo, J., Zhang, S., Tan, M., Li, J., Xu, H., Tan, Y., & Huang, Y. (2020). Targeted molecular profiling of genetic alterations in colorectal cancer using next‑generation sequencing. Oncology Letters, 19, 1137-1144. https://doi.org/10.3892/ol.2019.11203
MLA
Luo, J., Zhang, S., Tan, M., Li, J., Xu, H., Tan, Y., Huang, Y."Targeted molecular profiling of genetic alterations in colorectal cancer using next‑generation sequencing". Oncology Letters 19.2 (2020): 1137-1144.
Chicago
Luo, J., Zhang, S., Tan, M., Li, J., Xu, H., Tan, Y., Huang, Y."Targeted molecular profiling of genetic alterations in colorectal cancer using next‑generation sequencing". Oncology Letters 19, no. 2 (2020): 1137-1144. https://doi.org/10.3892/ol.2019.11203