Next-generation sequencing analysis of lung and colon carcinomas reveals a variety of genetic alterations

  • Authors:
    • Sandy Chevrier
    • Laurent Arnould
    • François Ghiringhelli
    • Bruno Coudert
    • Pierre Fumoleau
    • Romain Boidot
  • View Affiliations

  • Published online on: June 27, 2014     https://doi.org/10.3892/ijo.2014.2528
  • Pages: 1167-1174
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The development of targeted therapies in cancer has accelerated the development of molecular diagnosis. This new cancer discipline is booming, with an increasing number of gene alterations to analyze in a growing number of patients. To deal with this fast-developing activity, current analysis techniques (Sanger sequencing, allelic discrimination and high resolution melting) take more and more time. In recent years, next generation sequencing (NGS) technologies have appeared and given new perspectives in oncology. In this study, we analyzed FFPE lung and colon carcinomas using the Truseq Cancer Panel, which analyzes the mutation hotspots of 48 genes. We also tested the use of whole-genome amplification before NGS analysis. NGS results were compared with the data obtained from routine diagnosis. All of the alterations routinely observed were identified by NGS. Moreover, NGS revealed mutations in the KRAS and EGFR genes in patients diagnosed as wild-type by routine techniques. NGS also identified concomitant mutations in EGFR and KRAS or BRAF mutations, and a 15-nt deletion in exon 19 of EGFR in colon carcinomas. The study of the other genes sequenced in the Panel revealed 14 genes altered by 27 different mutations and three SNP with a possible role in cancer susceptibility or in the response to treatment. In conclusion, this study showed that NGS analysis could be used for the analysis of gDNA extracted from FFPE tissues. However, given the high sensitivity of this technology, high-throughput clinical trials are needed to confirm its reliability for the molecular diagnosis of cancer.

Introduction

Lung and colon cancers are among the main causes of death in developed countries. The life expectancy of patients is very limited, especially in metastatic disease. Nevertheless, in recent years, the development of targeted therapies (Tyrosine Kinase inhibitors or inhibitors of receptors which hyperactivate survival pathways) has shown great therapeutic promise. For example, patients with a mutated EGFR gene and wild-type KRAS gene lung tumor are eligible for gefitinib therapy (1). In colon cancer, patients with wild-type KRAS and BRAF could be treated with panitumumab or cetuximab (2). In skin cancer, vemurafenib (3) or imatinib (4) can be used to treat mutated BRAF (V600E) or c-KIT melanoma, respectively. As the efficacy of these targeted therapies depends on specific genetic abnormalities, molecular diagnosis has become essential for the treatment of cancers. Since 2008, molecular biology platforms have screened for genetic alterations in EGFR (exons 18–21), KRAS (codons 12 and 13) and BRAF (codon 600). At the beginning, the gold standard was Sanger sequencing, but the technique has low sensitivity and is expensive. With the increasing number of samples and gene alterations to screen for, alternon-amplified techniques were developed. For example, allelic discrimination (5) was developed to screen for a specific mutation quickly at a relatively low price. In parallel, screening technologies, such as High Resolution Melting (6) or fragment analysis (for indel alterations), were developed to use with Sanger sequencing, but only in the presence of potential mutations in the region of interest. Over the years, the number of genes with genetic alterations that could be targeted by therapies has increased rapidly. This medical progress has led to the need for more and more molecular diagnoses. This need has now been met by the recent development of Next Generation Sequencing (NGS), which has revolutionized molecular diagnosis. Indeed, the sequencing capacity allows the analysis of dozens of genes on multiplexed samples. In this paper, we describe the results we obtained with the Truseq Cancer Panel. In addition to the routinely detected mutations, NGS analysis, thanks to its high sensitivity, revealed new mutations in routinely analyzed genes.

Materials and methods

Patients and DNA samples

Eighteen tissue samples with >400 ng gDNA (Table I) from patients treated at the Centre Georges-François Leclerc between 2009 and 2013 were randomly chosen. Genomic DNA was extracted from FFPE tissues with either the QIAamp DNA mini kit (Qiagen, Heidelberg, Germany) or the Maxwell 16 FFPE Plus LEV DNA purification kit (Promega, Madison, USA). The samples had already been genotyped by allelic discrimination, fragment analysis and Sanger sequencing. Written consent was provided by all patients, and the researchers obtained authorization from the diagnostic centers to use the tumor samples.

Table I

Clinical details of studied patients.

Table I

Clinical details of studied patients.

PatientsOrgan of originHistologyAge (years)
L1LungKeratinizing poorly differentiated squamous carcinoma61
L2LungModerately differentiated adenocarcinoma67
L3LungPoorly differentiated adenocarcinoma62
L4LungAdenocarcinoma50
L5LungAdenocarcinoma69
L6LungAdenocarcinoma55
L7LungMucus-secreting adenocarcinoma68
L8LungAcinar differentiated mucus-secreting adenocarcinoma86
C1ColonModerately differentiated adenocarcinoma55
C2ColonAdenocarcinoma64
C3ColonModerately differentiated adenocarcinoma59
C4ColonAdenocarcinoma72
C5ColonAdenosquamous adenocarcinoma70
C6ColonPoorly differentiated adenocarcinoma55
C7ColonAdenocarcinoma80
C8ColonPoorly differentiated adenocarcinoma72
C9ColonWell differentiated infiltrating lieberkunien adenocarcinoma58
C10ColonColloidal adenocarcinoma67
Whole genome amplification

The Repli-g FFPE kit (Qiagen) was used to amplify 300 ng of gDNA from patients L7 and L8: 10 μl of gDNA solution were mixed with 8 μl of FFPE buffer, 1 μl of ligation enzyme and 1 μl of FFPE enzyme. The solution was then incubated at 24°C for 30 min, at 95°C for 5 min and then kept at 4°C. The Repli-g master mix was prepared by mixing, per sample, 29 μl of Repli-g Midi reaction buffer and 1 μl of Repli-g Midi DNA Polymerase. This second mixture (30 μl) was added to the gDNA solution. This solution was then incubated at 30°C for 2 h, 95°C for 10 min and kept at 4°C. Amplified DNA was stored at −20°C. Thanks to this protocol, we obtained 6900 and 6400 ng of amplified gDNA.

Preparation of libraries

Libraries were prepared with the Truseq Cancer Panel (Illumina, San Diego, USA) by following the manufacturer protocol. Briefly, 400–1250 ng of gDNA in 5 μl water was hybridized with an oligo pool. Then, unbound oligos were removed, and extension-ligation of bound oligos was followed by PCR amplification. PCR products were cleaned and checked for quality using Tapestation analysis (Agilent). The PCR product size had to be around 350 bp. Before sequencing, the libraries were normalized by the normalization process of the Truseq Cancer Panel.

Sequencing with MiSeq device

As each library possessed a specific primer index combination (i5 and i7), the libraries were pooled for 2 sequencing runs (pool no. 1, 10 libraries; pool no. 2, 9 libraries). For the MiSeq sample sheet, each sample was identified by its specific index combination. Libraries were paired-end sequenced with 2×151 bp cycles.

Analysis of obtained sequences

At the end of the run, sequences were aligned to the human genome reference hg19. Generated BAM files were analyzed with the Genome Golden Helix software (Golden Helix, Bozeman, USA). A genetic variation was defined by a Q-score above 30 (except for indel alteration).

Results

All mutations detected with standard methods were detected with NGS

In the routine diagnosis of lung or colon carcinomas, mutations in KRAS (exon 2), EGFR (exon 18–21), BRAF (exon 15) and HER2 (exon 20) genes are analyzed using three different methods: allelic discrimination for targeted mutations, fragment analysis for the screening of indel variations and Sanger sequencing for non-targeted mutations or characterization of indel abnormalities detected by fragment analysis. All mutation hotspots are analyzed one by one. Over the years, more and more genes and mutation hotspots will need to be explored. For example, exons 3 and 4 of KRAS, and exons 2–4 of NRAS and HRAS need to be analyzed before anti-EGFR antibody can be prescribed for colon cancer (7,8). In the first step of our study, we used the Truseq Cancer Panel kit to sequence samples that had already been analyzed in routine diagnosis. We then compared the results obtained by NGS with the results of the routine diagnosis at the same mutational hotspots. All the mutations detected in the routine diagnosis were also detected by NGS (Fig. 1 and Table II). Moreover, mutations not found in routine diagnosis were detected by NGS. These included a 15-nt deletion (c.2235_2249delGGAATTAAGAGAAGC) in two lung carcinomas classified as wild-type using routine methods (patients L1 and L6). In the L1 sample, another mutation in the KRAS gene (G12D) was also identified. In patient L6, this 15-nt deletion in EGFR was concomitant with a V600E BRAF mutation.

Table II

Comparison of results obtained routinely and with NGS.

Table II

Comparison of results obtained routinely and with NGS.

KRASBRAFEGFRHER2




PatientsRoutineaNGSRoutinebNGSRoutinecNGSRoutinedNGS
L1WTG12DWTWTWT15-nt E19WTWT
L2WTWTWTWT15-nt E1915-nt E19WTWT
L3WTWTWTWT24-nt E1924-nt E19WTWT
L4WTWTWTWTL858RL858RWTWT
L5WTWTWTWT15-nt E19 T790M15-nt E19 T790MWTWT
L6WTWTV600EV600EWT15-nt E19WTWT
L7G12CG12CWTWTWTWTWTWT
L8WTWTWTWTWTWTWTWT
C1WTWTWTV600RNDND
C2WTWTWTWTNDND
C3G13DG13DWTWTNDND
C4G12DG12DWTWTNDND
C5WTWTWTWTNDND
C6WTWTWTWTNDND
C7G13DG13DWTWTNDND
C8WTWTWTWTNDND
C9WTWTWTWTNDND
C10WTWTWTWTNDND

a Allelic discrimination (codons 12 and 13),

b Allelic discrimination (codon 600),

c Allelic discrimination (codons 719, 790, 858 and 861), fragment analysis and Sanger sequencing (exons 19, 20 and 21)

d fragment analysis and Sanger sequencing (exon 20).

{ label (or @symbol) needed for fn[@id='tfn5-ijo-45-03-1167'] } ND, not determined.

{ label (or @symbol) needed for fn[@id='tfn6-ijo-45-03-1167'] } Characters in bold show the discordance between routine and NGS analyses.

In colon cancer, a ‘common’ 15-nt deletion in the EGFR gene was detected only with NGS

Up to now, rare mutations of the KRAS gene have not been routinely analyzed in lung carcinomas. In our small population, a Q61H mutation in the KRAS gene was found in the sample L8. This mutation was localized in exon 3, which is not routinely analyzed in lung cancer. No other alteration was found in the routinely analyzed genes (HRAS and NRAS).

In colon cancer, only the genes KRAS, BRAF and very recently NRAS and HRAS are studied. Concerning rare mutations of KRAS, NRAS and HRAS, we detected a Q61K mutation in the NRAS gene in patient C5. As numerous genes were sequenced by the Cancer Panel kit, we analyzed the results obtained for the PIK3CA, HER2 and EGFR genes, which are routinely analyzed in lung carcinomas. No mutations were detected in exon 20 of HER2, or in exons 18, 20 or 21 of EGFR. In exon 20 of PIK3CA, an H1047L mutation was detected in patient C9. Concerning exon 19 of EGFR, a 15-nt deletion (the same as that observed in lung carcinomas) was detected in three patients (C4, C6 and C7). As this region was not routinely analyzed for colon cancer, we decided to perform both fragment analysis and Sanger sequencing. Neither fragment analysis, nor Sanger sequencing was able to detect the 15-nt deletion in exon 19 of EGFR in colon cancer (Fig. 2A). In contrast, NGS sequencing detected the deletion in >8% of sequenced fragments (Fig. 2B) for one patient. The two other patients harbored the mutation in approximately 4% of read sequences. Among these three patients, only one did not present a concomitant KRAS mutation.

WGA does not alter the NGS sequencing results

An important limitation in routine diagnosis is the quantity of gDNA extracted from FFPE samples and another paraffin block cannot be obtained in most cases. To counteract this limitation, we tested the impact of Whole Genome Amplification (WGA) on two samples of gDNA obtained from FFPE tissues. We then performed allelic discrimination on non-amplified gDNA and amplified gDNA (Fig. 3A). A KRAS G12C mutation was detected in both the amplified and non-amplified sample from patient L7. For patient L8, no KRAS G12C mutation was observed in either sample. In NGS analysis, the KRAS G12C mutation was also observed in patient L7 (non-amplified and WGA) but not in patient L8 (Fig. 3B). We also analyzed other routinely studied genes to compare the sequences before and after WGA. Whatever the gene analyzed, no point mutation was induced by the WGA (e.g., with the V600E BRAF and L858R EGFR hotpoint mutations in Fig. 3C). Even the rare mutation Q61H of KRAS was detected in both non-amplified and WGA gDNA from patient L8 (Fig. 3D). Moreover, the variant allele fraction was not modified after amplification.

NGS analysis revealed cancer susceptibility SNP and genetic alterations in some genes

The Illumina Cancer Panel kit studies exons with mutation hotpoints of 48 genes. We therefore analyzed all covered sequences for the 8 lung carcinomas and 10 colon carcinomas. Twenty-eight genetic alterations and three SNP related to cancer susceptibility or different protein activities were found (Table III). The most frequently altered gene was TP53 with nine alterations detected in nine patients. Double mutations in the colon cancer susceptibility genes APC and SMAD4 were detected in two patients (C3 and C8, respectively), suggesting a familial risk of colon cancer in these patients. For the patient with the APC mutation, we detected a concomitant c-MET activating mutation E168D. Concerning patient C8, we found a large number of alterations in different genes (c-KIT, c-MET, FBXW7, FGFR3, FLT3, IDH1, KRAS, RB1, SMAD4 and TP53), suggesting high genetic instability in this SMAD4 mutated tumor. Moreover, thanks to the non-targeted analysis, we detected two BRAF exon 15 mutations, N581S and V600R, which induce intermediate and strong activation of the protein, respectively. Furthermore, two mutations with unknown impact were detected in PIK3CA and PTEN. Concerning SNP, two patients (L7 and C4) harbored the breast cancer susceptibility ATM F858L SNP, and one patient (C10) had the rare c-KIT M541L SNP, which may influence the response to imatinib. Finally, 10 patients (8 heterozygotes and 2 homozygotes H472H) harbored the KDR Q472H polymorphism, which has been reported to increase tumor microvasculature.

Table III

Exonic SNP and genetic alterations in other analyzed genes.

Table III

Exonic SNP and genetic alterations in other analyzed genes.

GenesNucleotide variationProtein sequence variationPatientsImpact
APCc.2626C→TR876XC3Loss of function (familial mutation)
c.3944C→TS1315XC3Loss of function (somatic mutation) (26)
ATMc.2572T→CF858LL7, C4Breast cancer susceptibility SNP (23)
BRAFc.1742A→GN581SC2Intermediate activated (27)
c.1798_1799GT→AGV600RC1Strongly activated (28)
c-KITc.1621A→CM541LC10SNP with a potential effect on imatinib response (29)
c.2146G→AD716NC8Possible resistance to imatinib (24)
c-METc.504G→TE168DC3Activated (19)
c.1156C→AL386IC8Unknown (never observed)
FBXW7c.832C→TR278XC8Uncertain significance (30)
FGFR3 c.1196_1197GC→AGR399HC8Unknown (31)
FLT3c.2039C→TA680VC8Activated (32)
IDH1c.290G→AG97VC8Loss of wild-type function (33)
KDRc.1416A→TQ472HH: L1-L3, L5, L7-L8, C5-C6
O: C7, C9
Increased activity SNP (25)
KRASc.408T→AS136KC8Unknown
PIK3CAc.2176G→AE726KC2Unknown (34)
PTENc.563A→TD187VL1Unknown
RB1 c.2074_2075insATGAY692FsX2L2, L5Loss of function
c.2119T→CS707PC8Unknown
SMAD4c.1009G→AE337KC8Unknown
c.1082G→AR361HC8Loss of function (35)
TP53c.310C→TQ104XL1Unknowna
c.523C→TR175VC8Unknown
c.527G→TC176FL5Partially functional/deleteriousa
c.536A→GH179RC3 Non-functional/deleteriousa
IVS5+2T→GG187FsC1Unknown
c.709A→CM237LC10Partially functional/deleteriousa
c.743G→AR248QC6 Non-functional/deleteriousa
c.742C→TR248WC5 Non-functional/deleteriousa
c.830G→TC277FC7 Non-functional/deleteriousa

a From IARC database (36).

Discussion

Molecular diagnosis is the current challenge in cancer management. Indeed, with the increased number of targeted therapies and resistance mechanisms developed by cancer cells, the molecular analysis of tumors is a very important task to achieve optimal cancer therapy. Sanger sequencing, even when accompanied by alternon-amplified technologies, such as allelic discrimination or high resolution melting technology, has shown its limits. Today, next-generation sequencing is providing exciting new perspectives. In this study, we tested Truseq Amplicon technology for the analysis of mutation hotspots of 48 genes in gDNA extracted from FFPE samples. All of the mutations detected by routine Sanger sequencing, allelic discrimination or fragment analysis (in KRAS, BRAF, EGFR genes) were also identified with NGS analysis. Moreover, other alterations at the mutation hotspots of the routinely analyzed genes were also detected. These additional alterations included a G12D KRAS, a V600R BRAF and a 15-nt deletion in exon 19 of EGFR. The additional deletion in the EGFR gene was concomitant with the G12D KRAS mutation in one patient, and with a V600E BRAF mutation in another patient. KRAS, BRAF and EGFR mutations are normally exclusive (9) but concomitant KRAS and EGFR mutations have already been described (10,11). The identification of concomitant mutations should increase with the higher sensitivity of NGS technologies. Nevertheless, as it may be impossible to confirm these ‘new’ mutations using routine techniques, their clinical relevance and even their existence may be debatable (12). In the same way, some mutations are detected by NGS in very few of the read sequences. For example, the 15-nt deletion found in three colon carcinomas was detected in less than 8% of the read sequences, and among these three colon carcinomas, one had no KRAS/BRAF mutation, one had a concomitant G12D KRAS mutation, and one also had a G13D KRAS mutation. This observation is quite disturbing, and raises two questions: was the 15-nt deletion true, and if so, was this alteration clinically relevant given the small number detected. Today, the only way to have an answer would be to treat these patients with EGFR tyrosine kinase inhibitors or to observe anti-EGFR antibody resistance in these patients. To date, only patients with KRAS (13), HRAS or NRAS mutations (7,8) can be diagnosed as immediately resistant. Concerning our three colon carcinomas, two may benefit from treatment with EGFR TKI as the G13D KRAS mutation does not seem to interfere with the inhibition of the EGFR pathway (14).

With the increase in the number of genes to be analyzed for molecular diagnosis, the quantity of gDNA obtained from FFPE tissues will rapidly become a major problem, especially for lung carcinomas. In this work, we tested Whole Genome Amplification in two lung carcinomas and analyzed the resulting samples by NGS. The genetic profile obtained before and after WGA was qualitatively the same and quantitatively close. Indeed, only the intensity of the G12C KRAS mutation in patient L7 was slightly lower in the amplified sample. The strong similarity between amplified and non-amplified samples is in accordance in very recent studies, which showed that WGA can be safely used for diagnosis (15,16). Moreover, through this experiment, we showed that Truseq Amplicon technology is compatible with samples treated by WGA.

Among the genes or codons studied in the panel but not analyzed in routine molecular diagnosis, we detected 27 different alterations in 14 genes. Of these, 9 were detected in the TP53 gene, which is the most frequently altered gene in cancer (17). Eight mutations were in the DNA binding domain of the protein, indicating that these mutations are deleterious. One mutation occurred in a splice site, inducing a frameshift that may not be deleterious (18). We detected 2 genes with a double mutation, APC and SMAD4. The presence of two mutations in these two colon cancer predisposition genes indicated that these patients could have been members of families with a high risk of colon cancer. Both patients harbored mutations in the c-MET genes. The APC mutated patient had the activating mutation E168D (19), making him/her eligible for crizotinib therapy, which is generally used in lung cancer (20). Concerning the SMAD4 mutated tumor, we detected eight other altered genes, suggesting high genetic instability in this tumor type (21,22). The impact of most of these alterations is unknown. Nevertheless, the activating A680V mutation of FLT3 may be targeted by anti-FLT3 therapies currently in clinical development for the treatment of leukemia.

Three SNP modifying protein sequences were found. ATM F858L polymorphism, detected in two patients, is associated with an increased risk of breast cancer (23), but the small number of patients in our study does not allow us to draw any conclusions with regard to the predisposition for colon and lung cancer. Then, c-KIT M541L polymorphism was found in only one patient. The impact of this polymorphism in not known, but it has been suggested that it may affect the response to imatinib (24). Finally, KDR Q472H polymorphism was the most interesting alteration. Indeed, tumors with histidine show higher vascularization than do tumors with glutamine (25). In the Caucasian population, the frequency of each genotype is 58, 36 and 6% for Q472Q, Q472H and H472H, respectively. In our small population of tumors (n=18), we found enrichment of the histidine allele in 55.5% of our tumors (44.5% Q/Q, 44.5% Q/H and 11% H/H). In lung carcinomas we observed an enrichment of heterozygous tumors (75%), whereas in colon, the enrichment concerned homozygous H/H tumors. Moreover, during the analysis, we found that the presence of each allele was not 50/50 in heterozygous tumors, but varied from 9 to 96% of the read sequences. This raises the question of true polymorphism or a selection of tumor cells with a high angiogenic capacity. To answer this question, it would be necessary to analyze the constitutive DNA of each patient. According to the study by Glubb et al, patients with heterozygous or homozygous histidine tumors could be more sensitive to inhibiting treatments of VEGFR2 or to bevacizumab.

In conclusion, the analysis by NGS of FFPE lung and colon carcinomas identified the alterations highlighted by routine molecular diagnosis techniques. Thanks to its higher sensitivity, NGS analysis revealed new mutations that were not detected routinely. The impossibility to confirm the presence of these mutations by another technology is problematic, and the only way to answer this question is by conducting clinical trials that compare treatments of patients diagnosed by routine techniques or by NGS. Finally, the use of NGS in routine practice could revolutionize the management of cancer patients. Indeed, simultaneous analysis of numerous genes could identify drug-sensitive alterations generally observed in other cancer types (for example a c-MET alteration in a colon carcinoma that would be treated with crizotinib in lung cancer). Nevertheless, high throughput studies that combine NGS analysis and clinical trials need to be performed before NGS analysis can be generalized in routine molecular diagnosis.

Acknowledgements

We thank Philip Bastable for editing the manuscript.

References

1 

Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J and Haber DA: Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 350:2129–2139. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S and Bardelli A: Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 26:5705–5712. 2008.

3 

Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O’Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT and McArthur GA; BRIM-3 Study Group. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 364:2507–2516. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Carvajal RD, Antonescu CR, Wolchok JD, Chapman PB, Roman RA, Teitcher J, Panageas KS, Busam KJ, Chmielowski B, Lutzky J, Pavlick AC, Fusco A, Cane L, Takebe N, Vemula S, Bouvier N, Bastian BC and Schwartz GK: KIT as a therapeutic target in metastatic melanoma. JAMA. 305:2327–2334. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Lee LG, Connell CR and Bloch W: Allelic discrimination by nick-translation PCR with fluorogenic probes. Nucleic Acids Res. 21:3761–3766. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Liew M, Pryor R, Palais R, Meadows C, Erali M, Lyon E and Wittwer C: Genotyping of single-nucleotide polymorphisms by high-resolution melting of small amplicons. Clin Chem. 50:1156–1164. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, Rivera F, Kocákova I, Ruff P, Błasińska-Morawiec M, Šmakal M, Canon JL, Rother M, Williams R, Rong A, Wiezorek J, Sidhu R and Patterson SD: Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 369:1023–1034. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Fornaro L, Lonardi S, Masi G, Loupakis F, Bergamo F, Salvatore L, Cremolini C, Schirripa M, Vivaldi C, Aprile G, Zaniboni A, Bracarda S, Fontanini G, Sensi E, Lupi C, Morvillo M, Zagonel V and Falcone A: FOLFOXIRI in combination with panitumumab as first-line treatment in quadruple wild-type (KRAS, NRAS, HRAS, BRAF) metastatic colorectal cancer patients: a phase II trial by the Gruppo Oncologico Nord Ovest (GONO). Ann Oncol. 24:2062–2067. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Reddi HV: Mutations in the EGFR pathway: clinical utility and testing strategies. Clin Lab News. 39:14–16. 2013.

10 

Schmid K, Oehl N, Wrba F, Pirker R, Pirker C and Filipits M: EGFR/KRAS/BRAF mutations in primary lung adenocarcinomas and corresponding locoregional lymph node metastases. Clin Cancer Res. 15:4554–4560. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Tuononen K, Mäki-Nevala S, Sarhadi VK, Wirtanen A, Rönty M, Salmenkivi K, Andrews JM, Telaranta-Keerie AI, Hannula S, Lagström S, Ellonen P, Knuuttila A and Knuutila S: Comparison of targeted next-generation sequencing (NGS) and real-time PCR in the detection of EGFR, KRAS, and BRAF mutations on formalin-fixed, paraffin-embedded tumor material of non-small cell lung carcinoma-superiority of NGS. Genes Chromosomes Cancer. 52:503–511. 2013. View Article : Google Scholar

12 

McCourt CM, McArt DG, Mills K, Catherwood MA, Maxwell P, Waugh DJ, Hamilton P, O’Sullivan JM and Salto-Tellez M: Validation of next generation sequencing technologies in comparison to current diagnostic gold standards for BRAF, EGFR and KRAS mutational analysis. PLoS One. 8:e696042013. View Article : Google Scholar : PubMed/NCBI

13 

Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D and Bardelli A: Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 486:532–536. 2012.PubMed/NCBI

14 

Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C and Van Cutsem E: Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 30:3570–3577. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Della Starza I, De Novi LA, Nunes V, Del Giudice I, Ilari C, Marinelli M, Negulici AD, Vitale A, Chiaretti S, Foà R and Guarini A: Whole-genome amplification for the detection of molecular targets and minimal residual disease monitoring in acute lymphoblastic leukaemia. Br J Haematol. 165:341–348. 2014.PubMed/NCBI

16 

Hasmats J, Gréen H, Orear C, Validire P, Huss M, Käller M and Lundeberg J: Assessment of whole genome amplification for sequence capture and massively parallel sequencing. PLoS One. 9:e847852014. View Article : Google Scholar : PubMed/NCBI

17 

Vogelstein B, Sur S and Prives C: p53: the most frequently altered gene in human cancers. Nat Educat. 3:62010.

18 

Végran F, Rebucci M, Chevrier S, Cadouot M, Boidot R and Lizard-Nacol S: Only missense mutations affecting the DNA binding domain of p53 influence outcomes in patients with breast carcinoma. PLoS One. 8:e551032013.PubMed/NCBI

19 

Ma PC, Jagadeeswaran R, Jagadeesh S, Tretiakova MS, Nallasura V, Fox EA, Hansen M, Schaefer E, Naoki K, Lader A, Richards W, Sugarbaker D, Husain AN, Christensen JG and Salgia R: Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Res. 65:1479–1488. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Tanizaki J, Okamoto I, Okamoto K, Takezawa K, Kuwata K, Yamaguchi H and Nakagawa K: MET tyrosine kinase inhibitor crizotinib (PF-02341066) shows differential antitumor effects in non-small cell lung cancer according to MET alterations. J Thorac Oncol. 6:1624–1631. 2011. View Article : Google Scholar

21 

Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, Fan RS, Zborowska E, Kinzler KW, Vogelstein B, et al: Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 268:1336–1338. 1995. View Article : Google Scholar : PubMed/NCBI

22 

Woodford-Richens KL, Rowan AJ, Gorman P, Halford S, Bicknell DC, Wasan HS, Roylance RR, Bodmer WF and Tomlinson IP: SMAD4 mutations in colorectal cancer probably occur before chromosomal instability, but after divergence of the microsatellite instability pathway. Proc Natl Acad Sci USA. 98:9719–9723. 2001. View Article : Google Scholar

23 

Struewing JP: Genomic approaches to identifying breast cancer susceptibility factors. Breast Dis. 19:3–9. 2004.PubMed/NCBI

24 

Debiec-Rychter M, Cools J, Dumez H, Sciot R, Stul M, Mentens N, Vranckx H, Wasag B, Prenen H, Roesel J, Hagemeijer A, Van Oosterom A and Marynen P: Mechanisms of resistance to imatinib mesylate in gastrointestinal stromal tumors and activity of the PKC412 inhibitor against imatinib-resistant mutants. Gastroenterology. 128:270–279. 2005. View Article : Google Scholar

25 

Glubb DM, Cerri E, Giese A, Zhang W, Mirza O, Thompson EE, Chen P, Das S, Jassem J, Rzyman W, Lingen MW, Salgia R, Hirsch FR, Dziadziuszko R, Ballmer-Hofer K and Innocenti F: Novel functional germline variants in the VEGF receptor 2 gene and their effect on gene expression and microvessel density in lung cancer. Clin Cancer Res. 17:5257–5267. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Albuquerque C, Breukel C, van der Luijt R, Fidalgo P, Lage P, Slors FJ, Leitão CN, Fodde R and Smits R: The ‘just-right’ signaling model: APC somatic mutations are selected based on a specific level of activation of the beta-catenin signaling cascade. Hum Mol Genet. 11:1549–1560. 2012.

27 

Fratev FF and Jónsdóttir SO: An in silico study of the molecular basis of B-RAF activation and conformational stability. BMC Struct Biol. 9:472009. View Article : Google Scholar : PubMed/NCBI

28 

Klein O, Clements A, Menzies AM, O’Toole S, Kefford RF and Long GV: BRAF inhibitor activity in V600R metastatic melanoma. Eur J Cancer. 49:1073–1079. 2013. View Article : Google Scholar

29 

Grabellus F, Worm K, Sheu SY, Siffert W, Schmid KW and Bachmann HS: The prevalence of the c-kit exon 10 variant, M541L, in aggressive fibromatosis does not differ from the general population. J Clin Pathol. 64:1021–1024. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Davis H, Lewis A, Behrens A and Tomlinson I: Investigation of the atypical FBXW7 mutation spectrum in human tumours by conditional expression of a heterozygous propellor tip missense allele in the mouse intestines. Gut. 63:792–799. 2014. View Article : Google Scholar

31 

Imielinski M, Berger AH, Hammerman PS, Hernandez B, Pugh TJ, Hodis E, Cho J, Suh J, Capelletti M, Sivachenko A, Sougnez C, Auclair D, Lawrence MS, Stojanov P, Cibulskis K, Choi K, de Waal L, Sharifnia T, Brooks A, Greulich H, Banerji S, Zander T, Seidel D, Leenders F, Ansén S, Ludwig C, Engel-Riedel W, Stoelben E, Wolf J, Goparju C, Thompson K, Winckler W, Kwiatkowski D, Johnson BE, Jänne PA, Miller VA, Pao W, Travis WD, Pass HI, Gabriel SB, Lander ES, Thomas RK, Garraway LA, Getz G and Meyerson M: Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell. 150:1107–1120. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Piccaluga PP, Bianchini M and Martinelli G: Novel FLT3 point mutation in acute myeloid leukaemia. Lancet Oncol. 4:6042003. View Article : Google Scholar : PubMed/NCBI

33 

Ward PS, Cross JR, Lu C, Weigert O, Abel-Wahab O, Levine RL, Weinstock DM, Sharp KA and Thompson CB: Identification of additional IDH mutations associated with oncometabolite R(−)-2-hydroxyglutarate production. Oncogene. 31:2491–2498. 2012.PubMed/NCBI

34 

Jaiswal BS, Janakiraman V, Kljavin NM, Chaudhuri S, Stern HM, Wang W, Kan Z, Dbouk HA, Peters BA, Waring P, Dela Vega T, Kenski DM, Bowman KK, Lorenzo M, Li H, Wu J, Modrusan Z, Stinson J, Eby M, Yue P, Kaminker JS, de Sauvage FJ, Backer JM and Seshagiri S: Somatic mutations in p85alpha promote tumorigenesis through class IA PI3K activation. Cancer Cell. 16:463–474. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Miyaki M, Iijima T, Konishi M, Sakai K, Ishii A, Yasuno M, Hishima T, Koike M, Shitara N, Iwama T, Utsunomiya J, Kuroki T and Mori T: Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene. 18:3098–3103. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P and Olivier M: Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 28:622–629. 2007. View Article : Google Scholar

Related Articles

Journal Cover

September-2014
Volume 45 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chevrier S, Arnould L, Ghiringhelli F, Coudert B, Fumoleau P and Boidot R: Next-generation sequencing analysis of lung and colon carcinomas reveals a variety of genetic alterations. Int J Oncol 45: 1167-1174, 2014
APA
Chevrier, S., Arnould, L., Ghiringhelli, F., Coudert, B., Fumoleau, P., & Boidot, R. (2014). Next-generation sequencing analysis of lung and colon carcinomas reveals a variety of genetic alterations. International Journal of Oncology, 45, 1167-1174. https://doi.org/10.3892/ijo.2014.2528
MLA
Chevrier, S., Arnould, L., Ghiringhelli, F., Coudert, B., Fumoleau, P., Boidot, R."Next-generation sequencing analysis of lung and colon carcinomas reveals a variety of genetic alterations". International Journal of Oncology 45.3 (2014): 1167-1174.
Chicago
Chevrier, S., Arnould, L., Ghiringhelli, F., Coudert, B., Fumoleau, P., Boidot, R."Next-generation sequencing analysis of lung and colon carcinomas reveals a variety of genetic alterations". International Journal of Oncology 45, no. 3 (2014): 1167-1174. https://doi.org/10.3892/ijo.2014.2528