Open Access

Utility of exome sequencing in routine care for metastatic colorectal cancer

  • Authors:
    • Melchior De Giraud D'Αgay
    • Loïck Galland
    • Zoe Tharin
    • Caroline Truntzer
    • Francois Ghiringhelli
  • View Affiliations

  • Published online on: September 10, 2021     https://doi.org/10.3892/mco.2021.2392
  • Article Number: 229
  • Copyright: © D'Αgay 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

Metastatic colorectal cancer (mCRC) is a heterogenous disease and its prognosis depends on clinical features, such as tumor sidedness, and whether it is metachronous or synchronous. However, little is known about the overall genomic characterization of mCRC in these clinical subtypes. This single‑center observational study included 77 patients with mCRC who underwent somatic and germline exome analysis during the first or second line of therapy in 2018. Somatic and germline variants were determined in addition to tumor mutational burden, ploidy, clonality, human leucocyte antigen typing, neoantigens, and mutational and copy number signatures. Variables associated with sidedness, synchronous status and RAS status were determined using Fisher's test; and variables associated with overall survival were determined using univariate Cox survival models. The present study successfully generated whole exome sequencing analysis in 77 mCRC cases. Among them, 50 were left‑ and rectal‑sided, while 27 were right‑sided. Furthermore, 27 were metachronous and 46 were RAS‑mutated. The median OS was 3.75 years. It was observed that signature single nucleotide variation (SNV) 26, oncogenic alterations in receptor tyrosine kinase and nucleotide excision repair pathways were associated with tumor sidedness. SNV signature 3, Hedgehog signaling and mismatch repair pathways were associated with synchronous status. Phosphatidylinositol signaling system, ERK signaling and chromatin organization pathways were associated with RAS mutant status. In the whole cohort, metachronous metastasis was associated with improved survival. On gene variation, PTEN, PDGFRA, MYCN and SMAD4 were associated with poor prognosis, as was SNV signature 15. In conclusion, this study highlighted that structural and pathway genomic features are associated with sidedness, synchronous status, RAS status and overall survival and could be helpful to improve the stratification of patients with colorectal cancer.

Introduction

Colorectal cancer (CRC) is the second leading cause of cancer death worldwide (1). The estimated number of deaths due to CRC in Europe was 243,000 in 2018(2). Approximately 25% of patients present metastases at the time of diagnosis (synchronous disease) and about half of the remaining patients will develop metachronous metastases, contributing to the high mortality rates reported for CRC (3).

The treatment of metastatic CRC (mCRC) is based on chemotherapy when metastases cannot be removed by surgery. Treatment is guided by molecular information. Microsatellite unstable tumors are treated with immunotherapy (4), while microsatellite stable tumors, which account for 95% of mCRC, are treated by cytotoxic agents and targeted therapies. The principal cytotoxic agents currently used are fluoropyrimidines, irinotecan and oxaliplatin. The chemotherapy regimen often consists of fluorouracil and folinic acid combined with either oxaliplatin (FOLFOX) or irinotecan (FOLFIRI) or both (FOLFOXIRI). These chemotherapies are associated with two different classes of target therapies: Anti-angiogenesis drugs, and anti-epidermal growth factor receptor (EGFR). While antiangiogenics can be used broadly (5,6), anti EGFR therapy is only effective in patients with RAS wild type status (3,7).

It is currently established that mCRC is a heterogeneous disease. Tumor sidedness carries strong prognostic value (8) while synchronous mCRC is associated with a particularly poor prognosis. These data suggest that the biological characteristics of these tumors are different. Several papers have introduced CRC molecular subtyping systems, which are currently summarized in consensus molecular subtypes (9). This transcriptomic typology classifies patients into 4 consensual subtypes with different prognostic behavior. Using large panel sequencing data, a recent analysis detailed the molecular landscape of mCRC (10). This study observed different gene variations between left and right-sided tumors. However, the genomic structural pattern was not assessed in this study.

Here, using exome analysis of 77 mCRC patients, we aimed to determine genomic structural patterns and gene or pathway variations associated with overall survival, sidedness, RAS mutation status and synchronous disease.

Materials and methods

Study population

Seventy-seven patients with mCRC in whom WES (Whole Exome Sequencing) analysis was performed in 2018 as part of routine care, and interpreted according to the Molecular Tumor Board of the Georges François Leclerc Cancer Center, were included in this single-centre, retrospective study. WES analyses were performed during first or second line therapy. WES analysis is performed as part of routine care in our center in order to find potential targetable mutations for second line therapy. Before patients consented to WES of their tumoral tissue, they were informed by their oncologist. Germline testing was performed after counseling by a clinical geneticist.

Only patients from whom informed consent was obtained and recorded in the medical chart were included in this retrospective study. The study was approved by the CNIL (French national commission for data privacy) and the local ethics committee, and was performed in accordance with the Helsinki Declaration and European legislation.

Sample selection

Physicians selected an archival tumor sample (primary or metastasis) for genomic analysis. At the discretion of the physician, a new tumor biopsy could be proposed to the patient. Tumor cellularity was assessed by a senior pathologist on a hematoxylin and eosin slide from the same biopsy core used for nucleic acid extraction and molecular analysis.

DNA isolation

DNA was isolated from archival tumor tissue using the Maxwell 16 FFPE Plus LEV DNA purification kit (Promega Corp.). DNA from whole blood (germline DNA) was isolated using the Maxwell 16 Blood DNA Purification kit (Promega Corp.) following the manufacturer's instructions. The quantity of extracted genomic DNA was assessed by a fluorometric method with a Qubit device.

Whole exome capture and sequencing

Two hundred ng of genomic DNA were used for library preparation, using the Agilent SureSelectXT reagent kit (Catalog number G9642B, Agilent Technologies, Inc.)and the All Exon v5 probeset (5190-8863, Agilent Technologies, Inc.). Following hybridization, the libraries were purified according to the manufacturer's recommendations and amplified by polymerase chain reaction (12 cycles). DNA integrity was verified using TapeStation (SCREENTAPE D1000 tapestation 5067-5582, reagents D1000 tapestation 5067-5583). Concentrations were measured using Qubit® dsDNA BR Assay Q32853. Loading concentrations were 22 nM for fragmentation and 6 pM for NextSeq injection. Normalized libraries were pooled, and DNA was sequenced on an Illumina NextSeq500 device using 2x111-bp paired-end reads and multiplexed. Names, catalog numbers and suppliers of the Illumina sequencing kit were following: NextSeq 500 High Output Kit FC404-2004/2140817 and NextSeq 500 Mid Output Kit FC404-2003/2140816. More than 90% of the target sequence was covered with a read depth of at least 10X for somatic DNA.

Exome analysis pipeline

Reads in FASTQ format were aligned to the reference human genome GRCh37 using the Burrows-Wheeler aligner (BWA v.0.7.15). Local realignment was performed using the Genome Analysis Toolkit (GATK v.3.6). Duplicate reads were removed using Picard v.2.5. To identify somatic single-nucleotide variants (SNVs), a validated pipeline was used that integrates mutation calls from three different mutation callers. Single Nucleotide Variants (SNVs) were called with VarScan (v2.4.3) (11) and Mutect (v1.1.7) (12) insertion/deletions (indels) were called with VarScan and Strelka (v2.9.2) (13). Tumor Mutational Burden (TMB) was calculated using the number of significant SNV (UTRs, synonyms, introns and intergenic SNVs filtered out) divided by the number of megabases covered at a defined level. TMB was calculated with and without splicing sites mutations. Splicing site mutations were excluded because it has been demonstrated that variants present in splice regions have predominantly no impact (14). To identify tumor-specific mutant peptides, pVAC-Seq v4.0.3(15) (personalized Variant Antigens by Cancer Sequencing) was used; pVAC-Seq is based on HLA typing obtained by HLAminer (16). TITAN (17) was used to infer the number of copy number alterations (CNA) subclones, the number of large deletions, as well as loss of heterogeneity (LOH)>15 Mb from whole-exome sequencing data. It was also used to estimate tumor ploidy. SNV signatures were generated using DeconstructSigs (v1.8.0) (18) and COSMIC signatures identified by Alexandrov et al (19). CNV signatures were inferred according to the methodology of Macintyre et al (20). MSI score was computed using MSIsensor (21) HRD score was obtained through scarHRD (22) pipeline.

Statistical analysis

Patient and disease characteristics were compared across the different groups of interest using the Chi-2 or Fisher's exact test for qualitative variables and the Wilcoxon test for continuous variables, as appropriate. Enrichr analysis using KEGG database was performed on genes differentially mutated given sidedness, metastases and KRAS mutation status (23). Genes with a P-value <0.1 were selected for this analysis. Enrichr is a web-based tool for analysing gene sets; it returns any enrichment of common annotated biological features, here KEGG database.

Survival analysis was performed using the survival R library. Continuous variables were dichotomised using Lausen et al (24) methodology through the maxstat library (25). The prognostic value of the different variables was tested using univariate Cox regression for overall (OS) survival. OS was defined as the time from diagnosis to death from mCRC. Survivors were censored at the end of study. Survival probabilities were estimated using the Kaplan-Meier method and survival curves were compared using the log-rank test.

Statistical analyses were performed using the R software (http://www.R-project.org/) and graphs were drawn using GraphPad Prism version 7.03 (GraphPad Software, LLC).

Results

Patients' clinical characteristics

We included 27 (35%) patients with right primary colon cancer and 50 (65%) with rectal or left primary colon cancer. Twenty-seven (35%) patients had metachronous metastasis and 50 (65%) were synchronous. Forty-six (60%) patients were RAS mutated. The most frequent metastasis site was the liver (54 patients, 70%). Liver metastasis were more frequently synchronous (44 patients, 57%), than metachronous (10 patients, 12%) (P-value=0.01). The most common sites of first metastasis were the liver (74%) and the lung (27%), two metastatic sites that are potentially curable by resection. The other identified sites of metastases were the lymph nodes, peritoneal, adrenal and bone metastasis.

The presence of lung or liver metastases at time of diagnosis of metastatic disease did not vary significantly by primary tumour site (19% of right-sided tumors versus 32% of left-sided mCRC for lung metastasis, and 18% of right-sided tumors versus 36% of left-sided mCRC for liver metastasis). In contrast, peritoneal and omental metastases were more frequent among right-sided primary tumors (P-value=0.001). The main differences between clinical variables according to tumor sidedness are presented in Table SI, according to synchronous or metachronous status in Table SII, and between RAS/WT mutations in Table SIII.

All patients received a doublet or triplet of chemotherapy as first-line therapy; fluorouracil and folinic acid were used consistently. Patients with synchronous metastasis significantly more frequently received oxaliplatin in their chemotherapy regimens (P-value=0.01). Thirty patients were treated with anti-epidermal growth factor receptor (EGFR), without any significant difference concerning the sidedness or the time to metastasis (P-values 0.13 and 0.61 respectively). Sixty-four patients (83%) were treated with anti-angiogenesis drugs (bevacizumab or aflibercept). Fifty-four patients (70%) received a therapeutic proposition from the molecular tumor board. Only 18 patients (23%) received a treatment based on the molecular tumour board recommendations, most frequently using an oral MEK-inhibitor called Trametinib (Table I). Three patients yielded significant clinical benefit from these therapeutic strategies, with more than 6 months of progression-free survival. Two of these three patients had unstable microsatellite status and were treated with immunotherapy.

Table I

Description of treatment based on molecular tumour board recommendation.

Table I

Description of treatment based on molecular tumour board recommendation.

SampleSomatic mutationNucleotide variantProtein variantImpactTreatment
1BARD1, BRIP1c.266C>T, c.2665C>Ap.Pro89Leu, p.Gln889LysUnknownOLAPARIB
2NF1c.1007G>Ap.Trp336TerUnknownTRAMETINIB
3BRAFc.1799T>Ap.Val600GluActivating functionVEMURAFENIB
4RAD51Cc.859A>Gp.Thr287AlaLoss of functionOLAPARIB
5MSH6c.2017C>Ap.Pro673ThrUnknownNIVOLUMAB
6TOP1c.852G>Ap.Lys284LysUnknownIRINOTECAN
7TP53c.450_451delAC p.Pro152AlafsTer28Loss of functionAFLIBERCEPT
8KRASc.35G>Cp.Gly12AlaActivating functionTRAMETINIB
9MSH6.3254delC p.Phe1088SerfsTer2Loss of functionDURVAUMAB
10TP53c.638G>Tp.Arg213LeuLoss of functionBEVACIZUMAB
11MSH6c.2731C>Tp.Arg911TerUnknownNIVOLUMAB
12High number of variant---NIVOLUMAB
13KRASc.35G>Tp.Gly12ValActivating functionTRAMETINIB
14MTORc.6352C>Tp.Leu2118PheUnknownEVEROLIMUS
15TP53c.637C>Tp.Arg213XLoss of functionREGORAFENIB
16KRASc.38G>Ap.Gly13AspActivating functionTRAMETINIB
Patients' genomic characteristics

The most frequent mutation in the whole cohort was APC followed by TP53 and RAS (Fig. 1A). A summary of the genomic characteristics is presented in Table II. Thirty one patients presented WT mutational status, 44 patients presented KRAS and 2 patients NRAS mutational status. We identified 8 patients (10%) with BRAF mutation. The most frequent RAS mutation was KRAS (60%).

Table II

Summary of genomic characteristics by primary side, metachronous/synchronous and RAS/WT status.

Table II

Summary of genomic characteristics by primary side, metachronous/synchronous and RAS/WT status.

 Primary sideMoment of metastasis diagnosisRAS status
VariableWhole population (n=77)Right side (n=27)Left side (n=50)P-valueMetachronous group (n=27)Synchronous group (n=50)P-valueRAS mutated (n=46)RAS WT (n=31)P-value
MSI categories, n (%)   >0.99  0.46  >0.99
     MSS (score ≤5)72 (93.5)25(92)47(94) 24(89)48(98) 44(96)29(94) 
     MSI low and high5 (6.5)2(8)3(6) 3(11)2(2) 2(4)2(6) 
TMB without splicing categories, n (%)   0.41  0.66  >0.99
     Low (score ≤40)71(92)26(96)45(90) 24(89)47(94) 42(91)29(94) 
     High6(8)1(4)5(10) 3(11)3(6) 4(9)2 6) 
RAS mutation, n (%)46(60)18(69)28(56)0.6517(63)29(58)0.4646(100)--
BRAF mutation, n (%)8(10)5(19)3(6)0.185(19)3(6)0.12-8(10)>0.99
MSI score, median (IQR)0.08 (0.25)0.03 (0.27)0.09 (0.22)0.350.23 (0.43)0.09 (0.22)0.350.06 (0.21)0.10 (0.24)0.40
TMB without splicing regions score, median (IQR)5.5 (3.1)5.7 (2.8)5.4 (2.9)0.555.7 (2.8)5.4 (2.9)0.55.6 (3.1)5.4 (2.7)0.7
HRD score, median (IQR)23(13)23(14)23. (11.7)0.8723(14)23(12)0.8721(13)28(14)0.07
Ploidy, median (IQR)2.1 (0.5)2.1 (0.5)2.1 (0.5)0.732.1 (0.4)2.1 (0.5)0.732.1 (0.2)2.1 (0.6)0.3
Clonality, n (%)   0.06  0.062 (1.75)1 (1.75)0.3
     139(51)9(33)30(60) 9(33)30(60) -- 
     >137(49)17(63)20(40) 17(63)20(40) -- 
Deletions with LOH, median (IQR)3(8)1(7)4(9)0.211(7)4(9)0.215(9)1(5)0.1
Microdeletions, median (IQR)3(4)3 (3.)3(5)0.643(3)3(5)0.643(5)2(4)0.1
Neoantigens, median (IQR)11(8)11(8)11(7)0.711(8)11(7)0.8811(6)11(10)0.9
Strong neoantigens, median (IQR)1(2)1(1)1(2)0.071(2)1(2)0.631(2)1(2)0.9

[i] MSI, microsatellite instability; MSS, microsatellite stability; TMB, tumor mutational burden; HRD, homologous recombination deficiency; LOH, loss of heterozygosity; IQR, interquartile range; WT, wild-type.

Mean tumor mutational burden (TMB) without splicing regions in the whole cohort was 8.87 (median=5.50, IQR=3.07). The mean number of neoantigens was 13 (median=11, IQR=7.75). Five patients had a high HRD score, using the classical cut off value 42(26). The two most frequent SNV signatures were 1 and 25. The two most frequent CNV signatures were 3 and 5.

Seventy-two (94%) patients had proficient mismatch repair status (pMMR) and 5 (6%) had deficient mismatch repair status (dMMR) on immunohistological analysis (Table SIV). Median MSI score and TMB score were significantly higher in the dMMR group (respectively P-value=0.02 and P-value=0.01) (Fig. 1B). There was no significant enrichment of signature 6 in patients with dMMR status (P-value=0.7).

Association of genomic variables with sidedness

To evaluate whether we could isolate a genetic basis for the difference in survival between disease that originates in the right versus left side of the colon, we analysed genomic structural and gene alterations for the primary tumor site. The only difference in structural variants was significant enrichment of signature 26 in left-sided primary tumors (Fig. 2A). Signature 26 is associated with homologous repair deficiency (defective DNA mismatch repair).

For gene base analysis (Fig. 2B), there was significant enrichment of oncogenic alterations in AMER1, SMARCB1, ERBB4, FUBP1 and PTEN in right-sided primary tumors. Nevertheless, we did not identify any significant differences according to sidedness or frequencies of KRAS/BRAF mutations. Mutations observed in these genes are further described in Table SV. Results of Enrichr analysis using KEGG database are described in Table SVI.

Beyond the gene-level associations, analysis at the level of oncogenic pathways showed that mutations related to certain pathways differed according to primary tumor site. These pathways consisted in significant enrichment of Nucleotide excision repair for right-sided tumors, and RTK for left-sided ones (Fig. 2C). No significant association was found between sidedness and TP53/ATM, WNT/CTNNB1, TGF-beta or IGF2/PI-3-kinase pathways, which are frequently mutated in CRC.

Association of genomic variables with synchronous versus metachronous presentation

Using structural genome analysis, SNV signature 3, which is associated with failure of DNA double-strand break repair by homologous recombination, was significantly associated with synchronous presentation (Fig. 3A). We did not find additional differences at this genomic level.

P53 loss of function mutations were significantly more frequent in synchronous metastasis. In contrast, metachronous metastasis was associated with PTCH, PTPRD and CSF1R mutations (Fig. 3B). Mutations observed in these genes are further described in Table SV. Results of Enrichr analysis using KEGG database are described in Table SVI.

When pooling mutations in gene pathways, we observed that Mismatch repair and Hedgehog signaling were significantly more frequently affected in metachronous tumors (Fig. 3C).

Association of genomic variables with RAS status

In our series, the prevalence of RAS mutations was not significantly different according to tumor location. On gene variation, KMT2B and RET were significantly associated with RAS wild type tumors. In contrast, KMT2C and GNAS were significantly more frequent with RAS mutated tumors (Fig. S1A). Mutations observed in these genes are further described in Table SV. Results of Enrichr analysis using KEGG database are described in Table SVI.

An analysis at the level of oncogenic pathways demonstrated that ERK signaling and chromatin organisation pathways were significantly associated with RAS mutation, while Phosphatidylinositol signaling was associated with WT tumors (Fig. S1B).

Association of genomic variables with survival

Median OS was 3.75 years. Regarding genomic structural variants, low SNV signature 15 was associated with poor overall survival (Fig. 4A). In gene variation, PDGFRA, SMAD4, PTEN, MYCN mutations were significantly associated with poor prognosis (Fig. S2A-D). We also estimated a multivariate model involving the 4 genes; all genes remain significant. At the level of oncogenic pathways, cell-cycle, NFKappaB signaling and mismatch repair pathways were significantly associated with better survival (Fig. 4B-D). We did not observe a significant association between any WNT-signaling pathway and CRC survival.

Overall survival was significantly better in patients with metachronous metastatic disease, but this difference was no longer significant after removing MMR deficient tumors, thus suggesting a link between the prognosis of metachronous disease and enrichment in MMR deficient tumors (Fig. 4E and F).

Discussion

CRC is caused by multiple risk factors, including environmental, lifestyle and genetic risks. All these elements cause mutations and epigenetic alterations, conferring on cells the capacity to transform and grow, with aberrant DNA editing and defective DNA maintenance.

Next-generation sequencing (NGS) has identified a diversity of driver mutations in genes and altered signaling pathways in CRC (10,27). In addition, genomic structural patterns within gene pathway mutations could be used to identify prognostic features. Recent studies have revealed a mutational landscape of colorectal cancer and defined different subtypes that could guide therapeutic decisions (28). Increasing access to WES and the increasing rapidity of analysis offers new opportunities to implement such tests in the care of mCRC. WES analysis is performed in routine care in our center in order to find potentially targetable mutations for second line therapy.

Like in previous studies of precision medicine, we found that only very few mCRC patients yielded a benefit from precision medicine. In our study, we used WES analysis to identify genomic mutational profiles linked to tumor sidedness and metastatic occurrences, as well as genomic features related to prognosis.

In a systemic review and metanalysis (8), left-sided primary tumors were associated with improved prognosis, for both localized and metastatic tumors, in comparison to right-sided disease. Peritoneal and omental metastases, which are metastatic sites known to be associated with poor survival (29), were more frequent among right-sided primary tumors. Moreover, the survival differences seen between patients with right-sided vs. left-sided primary tumor sites in mCRC are supported by differences in transcriptomic patterns (8,30).

Like other authors (30,31), we showed that metachronous metastatic status is associated with better outcome. However, the molecular mechanism remains obscure, and the prognostic role seems to be less impactful than that of MMR status.

Our findings indicate that differences in survival could also be explained by genomic differences. Our analysis relied on structural information and on mutated genes grouped in pathways. We showed in particular that SNV signature 26, associated with MMR deficiency 29 (https://cancer.sanger.ac.uk/cosmic/signatures_v2.tt), was surprisingly associated with left-sided mCRC, while SNV signature 3 was associated with synchronous metastasis. Signature 3 is associated with better overall survival in ovarian (32) and breast cancer (33), by the better response to platinum therapy. At present, no data link signature 3 to oxaliplatin efficacy in mCRC. This question may be relevant for further clinical trials.

Moreover, somatic mutations in TP53 genes, which are present in the majority of cancers, are associated with poorer clinical outcomes, in several cancer types, including CRC (28). P53 loss of function mutations were significantly more frequent in synchronous metastasis.

In contrast, when we looked at more classical genomic features such as tumor mutational burden, there was no significant difference between tumors according to sidedness or time to metastasis occurrence. Conversely, we observed that MMR deficiency was associated with better outcome. MMR deficiency is more frequent in metachronous tumors and this genetic event may explain the better prognosis of these patients.

Oncogenic alterations-KRAS, NRAS and BRAF, which exhibit resistance to EGFR therapy with panitumumab and cetuximab, did not differ in our cohort, as previously reported (34), according to sidedness or metastatic occurrence, thus suggesting that these parameters are probably weak prognostic factors. The RAS pathway is a cell signaling pathway that plays a key role in regulation of cellular proliferation, apoptosis, cellular differentiation and migration and angiogenesis. This pathway is often dysregulated in mCRC. The association between RAS mutations and activation of the ERK signaling pathway is already well known.

When we looked at classical somatic mutations in mCRC, we observed that APC was frequently mutated in our series. However, APC was not associated with survival. The WNT signaling pathway is also frequently mutated in CRC and we did not observe a significant association between any WNT-signaling pathway and CRC survival. SMAD4 loss was found to be associated with poor outcome in our series. Similarly, a recent report also showed an association between SMAD4 loss and poor CRC survival, resistance to chemotherapy and decreased tumor immune infiltration (35).

Both PDGF and PDGFR families play an important role in colorectal carcinogenesis, and PDGFR is frequently overexpressed in CRC. Activation of this pathway is frequently related to angiogenesis, invasion, metastasis and poor survival (36). In our study, mutations in PDGFRA were associated with poor prognosis.

The main limitation of our work is the low number of patients included, which may impact the statistical significance, and precluded multivariate analysis. Moreover, WES was made during first or second line of treatment, so mutation profiles were generated under therapeutic pressure, which changed the cancer characterizations. Unfortunately, we do not dispose of samples before treatments as a baseline. In addition, considering the sequencing result of PTEN, PDGFRA, MYCN or SMAD4, the lack of PCR verification step is a limitation of this study and should be done in future works. Thus, our results should be considered as descriptive and exploratory, and warrant confirmation in further studies including larger sample sizes.

In conclusion, with the development of targeted therapies, it seems necessary to be able to rapidly identify the molecular status of mCRC tumors. Large panel or exome sequencing is slightly effective in improving patient care with precision medicine; however, such analyses could reveal structural and pathway genomic features that are associated with sidedness, synchronous status, and overall survival. Although the cost of NGS is steadily declining, WES nonetheless remains expensive, and currently, this is one of the major limitations on the routine use of exome sequencing. Moreover, the number of tumour mutations that could be used to treat a given patient is limited in conventional clinical practice or clinical trials, and it is difficult to determine which patients will clinically benefit from exome sequencing. In all likelihood, before being used in routine practice, exome sequencing will be reserved for patients with advanced mCRC, after one or more lines of treatment, or for patients with very poor outcomes, who fail to respond to classical targeted therapy and chemotherapy. Indeed, the knowledge of the molecular status could lead to inclusion in therapeutic clinical trials with a direct benefit for our patients. More generally, patients with right-sided mCRC, synchronous metastasis or peritoneal and omental metastasis may benefit the most from such analysis. This important information could be used to improve patient stratification for clinical trials, and will lead to a new molecular classification of patients that could be helpful to finetune the future of mCRC therapy.

Supplementary Material

Genetic characteristics according to RAS status. (A) Proportion of patients presenting at least one mutation in the most frequently mutated genes, by RAS status. (B) Distribution of patients presenting an alteration in significant signaling pathways according to RAS status. Only pathways for which the Fisher tests were considered significant (P<0.05) are presented. WT, wild-type.
Gene mutation and overall survival. Kaplan Meier curves for genes with significantly different overall survival according to their mutation status, namely (A) PDGFRA, (B) SMAD4, (C) PTEN and (D) MYCN. HR, hazard ratio; WT, wild-type.
Summary of clinical characteristics according to primary tumour side (right/left).
Summary of clinical characteristics according to metachronous/synchronous status.
Summary of clinical characteristics according to RAS mutational status.
Description of genomic characteristics according to MSI/MSS groups.
Description of mutations observed in genes differentially mutated in function of sidedness, synchronous or metachronous status, and KRAS mutated status.
Results of Enrichr analysis based on Kyoto Encyclopedia of Genes and Genomes pathways and genes differentially mutated in function of sidedness, synchronous or metachronous status, and KRAS mutated status.

Acknowledgements

The authors would like to thank Ms. Sandy Chevrier (Department of Medical Oncology, Georges François Leclerc Cancer Center-UNICANCER, Dijon, France), Dr Romain Boidot (Department of Medical Oncology, Georges François Leclerc Cancer Center-UNICANCER, Dijon, France) for providing whole exome sequencing data, Mr. Hugo Mananet (Department of Medical Oncology, Georges François Leclerc Cancer Center-UNICANCER, Dijon, France) for pre-processing data, and Dr Juliette Albuisson (Department of Medical Oncology, Georges François Leclerc Cancer Center-UNICANCER, Dijon, France) for her valuable comments. The authors would also like to thank Dr Fiona Ecarnot (EA3920, University of Franche-Comté, Besançon, France) for English correction and helpful comments.

Funding

No funding was received.

Availability of data and materials

The datasets generated and analyzed during the current study are available in the Sequence Read Archive repository, (https://www.ncbi.nlm.nih.gov/sra/?term=SRP318854).

Authors' contributions

MDGDA, LG, ZT, CT and FG contributed to the design and implementation of the research, to the analysis of the results and to the writing of the manuscript. CT and FG confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the CNIL (French national commission for data privacy) and the Georges François Leclerc Cancer Center (Dijon, France) local ethics committee (13.085), and was performed in accordance with the Helsinki Declaration and European legislation. Written informed consent was obtained from all subjects involved in the study. Only patients from whom informed consent was obtained and recorded in the medical chart were included in this retrospective study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

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.PubMed/NCBI View Article : Google Scholar

2 

Ferlay J, Colombet M, Soerjomataram I, Dyba T, Randi G, Bettio M, Gavin A, Visser O and Bray F: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries and 25 major cancers in 2018. Eur J Cancer. 103:356–387. 2018.PubMed/NCBI View Article : Google Scholar

3 

Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, Aranda Aguilar E, Bardelli A, Benson A, Bodoky G, et al: ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 27:1386–1422. 2016.PubMed/NCBI View Article : Google Scholar

4 

Andre T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt CJA, Smith DM, Garcia-Carbonero R, Benavides M, Gibbs P, et al: Pembrolizumab versus chemotherapy for microsatellite instability-high/mismatch repair deficient metastatic colorectal cancer: The phase 3 KEYNOTE-177 study. J Clin Oncol. 38 (Suppl 18)(LBA4)2020.PubMed/NCBI View Article : Google Scholar

5 

Giantonio BJ, Catalano PJ, Meropol NJ, O'Dwyer PJ, Mitchell EP, Alberts SR, Schwartz MA and Benson AB III: Eastern Cooperative Oncology Group Study E3200. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the eastern cooperative oncology group study E3200. J Clin Oncol. 25:1539–1544. 2007.PubMed/NCBI View Article : Google Scholar

6 

Van Cutsem E, Rivera F, Berry S, Kretzschmar A, Michael M, DiBartolomeo M, Mazier MA, Canon JL, Georgoulias V, Peeters M, et al: Safety and efficacy of first-line bevacizumab with FOLFOX, XELOX, FOLFIRI and fluoropyrimidines in metastatic colorectal cancer: The BEAT study. Ann Oncol. 20:1842–1847. 2009.PubMed/NCBI View Article : Google Scholar

7 

Van Cutsem E, Lenz HJ, Köhne CH, Heinemann V, Tejpar S, Melezínek I, Beier F, Stroh C, Rougier P, van Krieken JH and Ciardiello F: Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 33:692–700. 2015.PubMed/NCBI View Article : Google Scholar

8 

Petrelli F, Tomasello G, Borgonovo K, Ghidini M, Turati L, Dallera P, Passalacqua R, Sgroi G and Barni S: Prognostic survival associated with left-sided vs right-sided colon cancer: A systematic review and meta-analysis. JAMA Oncol. 3:211–219. 2017.PubMed/NCBI View Article : Google Scholar

9 

Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, et al: The consensus molecular subtypes of colorectal cancer. Nat Med. 21:1350–1356. 2015.PubMed/NCBI View Article : Google Scholar

10 

Yaeger R, Chatila WK, Lipsyc MD, Hechtman JF, Cercek A, Sanchez-Vega F, Jayakumaran G, Middha S, Zehir A, Donoghue MTA, et al: Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell. 33:125–136.e3. 2018.PubMed/NCBI View Article : Google Scholar

11 

Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L and Wilson RK: VarScan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22:568–576. 2012.PubMed/NCBI View Article : Google Scholar

12 

Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, Gabriel S, Meyerson M, Lander ES and Getz G: Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 31:213–219. 2013.PubMed/NCBI View Article : Google Scholar

13 

Kim S, Scheffler K, Halpern AL, Bekritsky MA, Noh E, Källberg M, Chen X, Kim Y, Beyter D, Krusche P and Saunders CT: Strelka2: Fast and accurate calling of germline and somatic variants. Nat Methods. 15:591–594. 2018.PubMed/NCBI View Article : Google Scholar

14 

Nykamp K, Anderson M, Powers M, Garcia J, Herrera B, Ho YY, Kobayashi Y, Patil N, Thusberg J, Westbrook M, et al: Sherloc: A comprehensive refinement of the ACMG-AMP variant classification criteria. Genet Med. 19:1105–1117. 2017.PubMed/NCBI View Article : Google Scholar

15 

Hundal J, Carreno BM, Petti AA, Linette GP, Griffith OL, Mardis ER and Griffith M: pVAC-Seq: A genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 8(11)2016.PubMed/NCBI View Article : Google Scholar

16 

Warren RL, Choe G, Freeman DJ, Castellarin M, Munro S, Moore R and Holt RA: Derivation of HLA types from shotgun sequence datasets. Genome Med. 4(95)2012.PubMed/NCBI View Article : Google Scholar

17 

Ha G, Roth A, Khattra J, Ho J, Yap D, Prentice LM, Melnyk N, McPherson A, Bashashati A, Laks E, et al: TITAN: Inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 24:1881–1893. 2014.PubMed/NCBI View Article : Google Scholar

18 

Rosenthal R, McGranahan N, Herrero J, Taylor BS and Swanton C: DeconstructSigs: Delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 17(31)2016.PubMed/NCBI View Article : Google Scholar

19 

Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ and Stratton MR: Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 3:246–259. 2013.PubMed/NCBI View Article : Google Scholar

20 

Macintyre G, Goranova TE, De Silva D, Ennis D, Piskorz AM, Eldridge M, Sie D, Lewsley LA, Hanif A, Wilson C, et al: Copy number signatures and mutational processes in ovarian carcinoma. Nat Genet. 50:1262–1270. 2018.PubMed/NCBI View Article : Google Scholar

21 

Middha S, Zhang L, Nafa K, Jayakumaran G, Wong D, Kim HR, Sadowska J, Berger MF, Delair DF, Shia J, et al: Reliable pan-cancer microsatellite instability assessment by using targeted next-generation sequencing data. JCO Precis Oncol. 2017(PO.17.00084)2017.PubMed/NCBI View Article : Google Scholar

22 

Sztupinszki Z, Diossy M, Krzystanek M, Reiniger L, Csabai I, Favero F, Birkbak NJ, Eklund AC, Syed A and Szallasi Z: Migrating the SNP array-based homologous recombination deficiency measures to next generation sequencing data of breast cancer. NPJ Breast Cancer. 4(16)2018.PubMed/NCBI View Article : Google Scholar

23 

Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR and Ma'ayan A: Enrichr: Interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 14(128)2013.PubMed/NCBI View Article : Google Scholar

24 

Lausen B, Hothorn T, Bretz F and Schumacher M: Assessment of optimal selected prognostic factors. Biom J. 46:364–374. 2004.

25 

Hothorn T and Lausen B: On the exact distribution of maximally selected rank statistics. Comput Stat Data Anal. 43:121–137. 2003.

26 

Takaya H, Nakai H, Takamatsu S, Mandai M and Matsumura N: Homologous recombination deficiency status-based classification of high-grade serous ovarian carcinoma. Sci Rep. 10(2757)2020.PubMed/NCBI View Article : Google Scholar

27 

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

28 

Zaidi SH, Harrison TA, Phipps AI, Steinfelder R, Trinh QM, Qu C, Banbury BL, Georgeson P, Grasso CS, Giannakis M, et al: Landscape of somatic single nucleotide variants and indels in colorectal cancer and impact on survival. Nat Commun. 11(3644)2020.PubMed/NCBI View Article : Google Scholar

29 

Franko J, Shi Q, Goldman CD, Pockaj BA, Nelson GD, Goldberg RM, Pitot HC, Grothey A, Alberts SR and Sargent DJ: Treatment of colorectal peritoneal carcinomatosis with systemic chemotherapy: A pooled analysis of north central cancer treatment group phase III trials N9741 and N9841. J Clin Oncol. 30:263–267. 2012.PubMed/NCBI View Article : Google Scholar

30 

Boeckx N, Koukakis R, Op de Beeck K, Rolfo C, Van Camp G, Siena S, Tabernero J, Douillard JY, André T and Peeters M: Primary tumor sidedness has an impact on prognosis and treatment outcome in metastatic colorectal cancer: Results from two randomized first-line panitumumab studies. Ann Oncol. 28:1862–1868. 2017.PubMed/NCBI View Article : Google Scholar

31 

Ghiringhelli F, Hennequin A, Drouillard A, Lepage C, Faivre J and Bouvier AM: Epidemiology and prognosis of synchronous and metachronous colon cancer metastases: A French population-based study. Dig Liver Dis. 46:854–858. 2014.PubMed/NCBI View Article : Google Scholar

32 

Pennington KP, Walsh T, Harrell MI, Lee MK, Pennil CC, Rendi MH, Thornton A, Norquist BM, Casadei S, Nord AS, et al: Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin Cancer Res. 20:764–775. 2014.PubMed/NCBI View Article : Google Scholar

33 

Zhao EY, Shen Y, Pleasance E, Kasaian K, Leelakumari S, Jones M, Bose P, Ch'ng C, Reisle C, Eirew P, et al: Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clin Cancer Res. 23:7521–7530. 2017.PubMed/NCBI View Article : Google Scholar

34 

Bylsma LC, Gillezeau C, Garawin TA, Kelsh MA, Fryzek JP, Sangaré L and Lowe KA: Prevalence of RAS and BRAF mutations in metastatic colorectal cancer patients by tumor sidedness: A systematic review and meta-analysis. Cancer Med. 9:1044–1057. 2020.PubMed/NCBI View Article : Google Scholar

35 

Wasserman I, Lee LH, Ogino S, Marco MR, Wu C, Chen X, Datta J, Sadot E, Szeglin B, Guillem JG, et al: SMAD4 loss in colorectal cancer patients correlates with recurrence, loss of immune infiltrate, and chemoresistance. Clin Cancer Res. 25:1948–1956. 2019.PubMed/NCBI View Article : Google Scholar

36 

Manzat Saplacan RM, Balacescu L, Gherman C, Chira RI, Craiu A, Mircea PA, Lisencu C and Balacescu O: The Role of PDGFs and PDGFRs in colorectal cancer. Mediators Inflamm. 2017(4708076)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2021
Volume 15 Issue 5

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
D'Αgay MD, Galland L, Tharin Z, Truntzer C and Ghiringhelli F: Utility of exome sequencing in routine care for metastatic colorectal cancer. Mol Clin Oncol 15: 229, 2021
APA
D'Αgay, M.D., Galland, L., Tharin, Z., Truntzer, C., & Ghiringhelli, F. (2021). Utility of exome sequencing in routine care for metastatic colorectal cancer. Molecular and Clinical Oncology, 15, 229. https://doi.org/10.3892/mco.2021.2392
MLA
D'Αgay, M. D., Galland, L., Tharin, Z., Truntzer, C., Ghiringhelli, F."Utility of exome sequencing in routine care for metastatic colorectal cancer". Molecular and Clinical Oncology 15.5 (2021): 229.
Chicago
D'Αgay, M. D., Galland, L., Tharin, Z., Truntzer, C., Ghiringhelli, F."Utility of exome sequencing in routine care for metastatic colorectal cancer". Molecular and Clinical Oncology 15, no. 5 (2021): 229. https://doi.org/10.3892/mco.2021.2392