#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Molecular characterization of lung adenocarcinoma from Korean patients using next generation sequencing


Authors: You Jin Chun aff001;  Jae Woo Choi aff002;  Min Hee Hong aff001;  Dongmin Jung aff002;  Hyeonju Son aff004;  Eun Kyung Cho aff005;  Young Joo Min aff006;  Sang-We Kim aff007;  Keunchil Park aff008;  Sung Sook Lee aff009;  Sangwoo Kim aff004;  Hye Ryun Kim aff001;  Byoung Chul Cho aff001
Authors place of work: Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea aff001;  Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea aff002;  Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea aff003;  Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea aff004;  Division of Hematology-Oncology, Department of Internal Medicine, Gachon Medical School, Gil Medical Center, Incheon, Korea aff005;  Division of Hematology and Oncology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea aff006;  Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea aff007;  Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea aff008;  Department of Hematology-Oncology, Inje University Haeundae Paik Hospital, Busan, Korea aff009
Published in the journal: PLoS ONE 14(11)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0224379

Summary

The treatment of Lung adenocarcinoma (LUAD) could benefit from the incorporation of precision medicine. This study was to identify cancer-related genetic alterations by next generation sequencing (NGS) in resected LUAD samples from Korean patients and to determine their associations with clinical features. A total of 201 tumors and their matched peripheral blood samples were analyzed using targeted sequencing via the Illumina HiSeq 2500 platform of 242 genes with a median depth of coverage greater than 500X. One hundred ninety-two tumors were amenable to data analysis. EGFR was the most frequently mutated gene, occurring in 106 (55%) patients, followed by TP53 (n = 67, 35%) and KRAS (n = 11, 6%). EGFR mutations were strongly increased in patients that were female and never-smokers. Smokers had a significantly higher tumor mutational burden (TMB) than never-smokers (average 4.84 non-synonymous mutations/megabase [mt/Mb] vs. 2.84 mt/Mb, p = 0.019). Somatic mutations of APC, CTNNB1, and AMER1 in the WNT signaling pathway were highly associated with shortened disease-free survival (DFS) compared to others (median DFS of 89 vs. 27 months, p = 0.018). Patients with low TMB, annotated as less than 2 mt/Mb, had longer DFS than those with high TMB (p = 0.041). A higher frequency of EGFR mutations and a lower of KRAS mutations were observed in Korean LUAD patients. Profiles of 242 genes mapped in this study were compared with whole exome sequencing genetic profiles generated in The Cancer Genome Atlas Lung Adenocarcinoma. NGS-based diagnostics can provide clinically relevant information such as mutations or TMB from readily available formalin-fixed paraffin-embedded tissue.

Keywords:

Human genetics – Lung and intrathoracic tumors – Cancer genomics – Mutation databases – Next-generation sequencing – Nonsense mutation – Deletion mutation – Somatic mutation

Introduction

Lung adenocarcinoma (LUAD) is the leading cause of cancer death worldwide. In particular, the incidence of LUAD is increasing in both never-smokers and females[1]. This means that prognosis and treatment of each patient can differ widely at the molecular level based on their gene expression patterns, copy number alterations, and mutations. Previous genomic studies of LUAD have shown that patients with driver gene mutations, such as those in epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK), receive a significant survival benefit from personalized therapy for LUAD [2, 3]. The recent discoveries of C-Ros oncogene 1, receptor tyrosine kinase (ROS1) and Ret proto-oncogene (RET1) fusions have raised expectations for the development of new targeted agents in LUAD. In molecularly selected patients, response rates to the appropriate targeted treatment can reach 60–70% or more, compared to the 20–30% response rate in an unselected population treated with conventional chemotherapy [4].

Ethnicity plays a distinct role in the prevalence of some genetic markers[5]. Asian patients with LUAD have a longer survival (11.0 vs. 8.9 months, p < 0.001), higher response rates (32.7 vs. 29.8%, p = 0.027), and greater toxicity in response to targeted therapy than Caucasian patients [6]. However, there is still a limited understanding of the genetic features of LUAD in Asian patients based on a lack of representation in existing public databases. Therefore, it is worthwhile to investigate whether these ethnic differences are due to genetic variation among ethnic groups. In this study, we investigated these variations in a Korean LUAD cohort. As we were able to sequence individual genomes, we examined these markers via next generation sequencing (NGS) technology, which can determine the profile of genetic changes in tumors, including single-nucleotide variations (SNVs), copy number variations (CNVs), and complex chromosomal rearrangements. NGS technology can provide a fast turnaround time and cost-effective sequencing for high numbers of targets. Given this, we sought to delineate a comprehensive characterization of the genomic landscape in Korean patients with LUAD using formalin-fixed paraffin-embedded (FFPE) surgical tissues and NGS technology. We have rendered to provide NGS results in a relevant time with simple FFPE samples rather than fresh tissue by targeted sequencing analysis, which is feasible to apply in clinical practice. Our data may serve as a reference in the development of precision medicine for Korean LUAD patients.

Materials and methods

Patients and data collection

A total of 201 LUAD patients with surgically resected primary lung cancer were prospectively enrolled from the Yonsei Cancer Center and Ulsan University Hospital between 2014 and 2016. All patients provided prior written informed consent, and this study was conducted with the approval of Institutional Review Board of Yonsei University Health System, Severance Hospital. A predesigned data collection format was used to review the patients’ electronic medical records for evaluation of clinicopathological characteristics and survival outcomes. Never-smokers were defined as those with a lifetime smoking dose of < 100 cigarettes. Ten tumor tissue sections (at least 10 μm thick) and patient blood samples (5 ml) were collected from prospectively recruited patients to differentiate between germline and somatic genetic aberrancy. Genetic analyses were performed in routine practice and included EGFR mutation and ALK/ROS1 rearrangement. We uploaded raw NGS data to National Center for Biotechnology Information Sequence Read Archive (NCBI SRA) website for public access. (https://trace.ncbi.nlm.nih.gov/Traces/sra/, SRA accession ID is SRP200786.)

Targeted sequencing of tumors

Genomic DNA was isolated from FFPE samples using the QIAamp DNA FFPE Tissue Kit (Qiagen, Hilden, Germany) for the targeted sequencing of 242 lung cancer-related genes selected based on a literature search (S1 Table) [2, 7, 8]. The genomic regions of the 242 genes were captured by the customized SureSelectXT Target Enrichment library generation kit (Agilent, Santa Clara, CA, USA) and sequenced on the Illumina HiSeq 2500 platform with a depth of coverage > 500X and a read length of 100 bp.

To do FFPE quality control and analysis, two cross validations were performed. First, we checked up and confirmed that FFPE precisely detect EGFR hotspot mutations which are the main target of LUAD therapy. We compared the NGS result with the PCR result regarding the EGFR hotspot mutations in the same sample. Another is to compare the results of the known frozen fresh (FF) data in public dataset. We first evaluated how similar the overall pattern of LUAD results of current study with that of The Cancer Genome Atlas (TCGA) dataset which was conducted by FF [2]. To evaluate the overall pattern of our data, we compared that of TCGA dataset [2]. This TCGA data set is composed of a total of 230 patients, of which the majorities (173 patients) were Caucasian [2].

Variant calling and functional annotation

By default, base quality trimming for short reads from the targeted sequences was performed using Sickle[9]. Filtered reads were mapped to the human reference genome (GRCh37/hg19) using BWA[10]. All reads with a mapping quality score < 20 were discarded. The aligned reads (BAM file) were further processed with the Genome Analysis Tool Kit v3.5[11], including Mark Duplicate, Local Realignment, and Base Quality Score Recalibration. Candidates for somatic mutations were called by MuTect ver. 1.17[12] with default parameters. Somatic insertions/deletions were called by Scalpel [13] with default parameters. During somatic mutation calling, FoxoG sequencing artifacts [14] were removed using the Oxidative Damage Detection and Removal Tools (https://github.com/migbro/IGSB_oxoG_tools) to discard skewed read-orientation variants with the FoxoG parameter 0.625. Even after FoxoG filtration, nine samples had unexpectedly large numbers of mutations (Z-score of tumor mutational burden (TMB) > 1) and thus were excluded from further analysis under suspicion of potential damage to DNA. Somatic variants that passed all filters were considered high-confidence variants. CNVs were called using a CNV kit [15]. CNVs in genes were defined as follows: deletion, 0 copies; loss, 1 copy; gain, 3 copies; and amplification, ≥ 4 copies. The functional impacts of high confidence variants were annotated with ANNOVAR software[16], based on the consequences, predicted impacts, and reported allele frequencies in the population. In particular, non-rare variants (minor allele frequency > 0.05 in gnomAD database [17]) were discarded to remove non-pathogenic variants. Finally, CIVic and DoCM databases were used for clinical interpretation of variants in cancer. TMB was measured by the number of non-synonymous missense mutations per megabase (Mb) within the range of the targeted capture region. An ‘Oncoprint’ is a way to visualize overall genomic alteration events using a heatmap. Mutations of each sample on the Oncoprint are aligned in a mutually exclusive manner. For example, the samples with the highest frequency in the entire sample are aligned on the top left, and the samples with the next highest frequency are aligned on the back. This is a kind of clustering that can easily distinguish between co-occurrence and mutually exclusive patterns on the major genes. It was drawn using the bioconductor package ‘Complex Heatmap’ [18] in R ver. 3.4. Using the package ‘maftools’ [19], lollipop plots were drawn for frequently mutated genes to check the recurrence of genomic loci with variants, and somatic interactions between mutually exclusive or co-occurring sets of genes were investigated. Mutations and putative CNVs stored in cBioportal[20, 21] were used for the above genomic analysis. Pathway diagrams were depicted using Pathway Mapper[22]. To identify the clinical importance of mutations, we created a mutation classification system based on knowledgebase databases and a computational prediction algorithm. Clinical importance was ranked using CIVic(criteria: predictive & sensitive & evidence level = {A, B, C} & supports categories), Cancer Genome Interpreter (CGI) (criteria: drug prescription & responsive & alteration match–complete categories)[23], and CRAVAT(criteria: CHASM FDR ≤ 0.1 & TARGET DB only categories), sequentially. To confirm how many ranked mutations were included, Venn diagrams were drawn using Venny [24].

Statistical methods

All statistical analyses were performed using R and Python (Scipy and Seaborn packages). Student's t-test or Fisher's exact test was used for group comparisons. Disease-free survival (DFS) was measured from the date of diagnosis to tumor recurrence or death, while overall survival (OS) was measured from the date of diagnosis until the date of death. Patients were censored on October 2017 if alive and recurrence free. Patients without a known date of death were censored at the time of last follow-up. A log rank test for mutations of each gene, signaling pathways, and TMB was used to compare the DFS between groups. Two-sided p-values < 0.05 were considered significant.

Results

Clinical characteristics

We enrolled 201 patients with LUAD, and their characteristics are summarized in Tables 1 and S2. This entire cohort included 87 men and 114 women; the median age was 63 years (range, 34–83), and 157 patients (78.1%) had stage I or II disease at initial diagnosis. One hundred twenty-five patients (62.2%) were never-smokers; never-smokers were defined as those with a lifetime smoking dose of < 100 cigarettes. One hundred nineteen patients (59.2%) were urban residents and 68 patients (33.8%) were non-urban residents. Ninety-four patients (46.8%) had adjuvant platinum-based chemotherapy as a standard treatment. EGFR mutations (51.8%, 57/110), ALK rearrangements (4.1%, 4/98), and ROS1 rearrangements (0.7%, 1/140) were identified based on genetic mutation testing in routine practice. The mean follow-up period was 42 months (range, 7–114 months). During follow-up, recurrence was observed in 55 patients (27.4%), and median DFS was 89 months (95% confidence interval [CI], range 63.68–114.32 months). DFS was separated by each stage (S1 Fig). Since only 10 patients died, the median OS was not yet reached (Table 2).

Tab. 1. Baseline patient characteristics of the analytic population.
Baseline patient characteristics of the analytic population.
Tab. 2. Clinical outcomes of the analytic population.
Clinical outcomes of the analytic population.

Genomic landscape of LUAD

We analyzed 192 of 201 samples, after excluding nine with excessive FoxoG artifacts. To confirm FFPE quality control, we compared the NGS result with their PCR result on the well-known EGFR hotspot mutations (i.e.S768I, L858R, L861Q, E19 DEL) and identified that about 90% (45/51) were identical to each other. Next, we evaluated the overall pattern of our data compared with that of TCGA data set as positive control (S2 Fig) [2]. We confirmed that the mutation patterns of EGFR, TP53, KRAS, and PIK3CA of our data are the comparable with TCGA (S2A Fig). In TCGA LUAD dataset, EGFR hotspot mutations were observed in L858R, exon19 deletion and other hotspot mutations order, which is similar pattern to our result (S2B Fig). TP53 tends to be widely distributed in the DNA binding domain (S2C Fig). KRAS (G12, Q61) and PIK3CA (E542, E545, H1047) were observed in TCGA dataset, which are comparable with ours (S2D and S2E Fig). In terms of CNV, we found out that the oncogene and tumor suppressor gene (TSG) are similar to TCGA and COSMIC (https://cancer.sanger.ac.uk/cosmic) (S2F Fig). Collectively, based on the quality control process, we analyzed genetic analysis in this study.

A total of 761 somatic non-silent SNVs and 388 insertions and deletions (indels) were identified from the targeted sequencing regions of the 192 tumors, corresponding to a median of 2.08 SNVs per Mb. The Oncoprint demonstrated that SNV alterations included missense, nonsense, frameshift indel, in-frame indel, and splice site mutations. We found that EGFR was the most frequently mutated gene (n = 106, 55%), followed by TP53 (n = 67, 35%) and KRAS (n = 11, 6%) (Fig 1A). EGFR and KRAS mutations were mutually exclusive (S3A Fig). EGFR mutation were strongly enriched in patients that were female and never-smokers. In female and never-smokers, between patients with EGFR mutation and EGFR wild type are statistically significant (p = 0.005). Rates of mutations in other genes were 8% for ADGRV1 (n = 15), 8% for SMARCA2 (n = 15), 7% for BIRC6 (n = 13), 7% for NF1 (n = 13), 6% for RELN (n = 12), 6% for KMT2C (n = 12), 6% for FAT3 (n = 12), 6% for ATM (n = 11), and 5% for RB1 (n = 9) (Fig 1A). Copy number gain or amplification was detected in HRAS (n = 38, 20%), FGFR3 (n = 36, 19%), TERT (n = 34, 18%), CREBBP (n = 30, 16%), MYC (n = 26, 14%), AKT (n = 25, 13%), and EGFR (n = 24, 12%). Copy number loss or deletion was observed in CDKN2A (n = 26, 14%), SMAD4 (n = 17, 9%), VHL (n = 15, 8%), STK11 (n = 16, 8%), PTEN (n = 13, 7%), and KMT2D (n = 13, 7%) (Fig 1B). We found that smokers had a significantly higher TMB than never-smokers (average 4.84 vs. 2.84 mt/Mb, respectively, p = 0.019) (S3B Fig).

Genomic landscape of lung adenocarcinoma (LUAD).
Fig. 1. Genomic landscape of lung adenocarcinoma (LUAD).
(A) This is the oncoprint of the somatic single-nucleotide variations (SNVs) in 192 LUAD patients. SNV alterations included missense, nonsense, frameshift indel, in-frame indel, and splice site mutations. (Red, oncogene; Blue, tumor suppressor gene) The upper bar chart is the total number of mutations or CNVs. The order of the genes is performed by the frequency of the mutations or CNVs across all samples. (B) This is the oncoprint of the somatic copy number variations (CNVs) in 192 LUAD patients. CNV alterations included copy number gain, amplification, loss, or deletion.

Mutation mapper plot and pathway diagram

In the mapper plot for EGFR, L858R and exon 19 deletion were the most common alterations, observed in 35 samples (18%) and 55 samples (29%), respectively. This was followed by L861Q, observed in 5 samples (3%). In the mapper plot for TP53, the P112S (n = 2, 1%), V118F (n = 2, 1%), R136H (n = 2, 1%), N171fs (n = 2, 1%), H175R (n = 2, 1%), G206C/V (n = 2, 1%), R234H (n = 2, 1%), and E246K (n = 2, 1%) mutations were identified at similar rates. In PIK3CA, the established canonical E542K missense mutation was the most common (n = 3, 2%). In addition, among KRAS mutations in codon 12, G12D/V/C/S/A was the most common (n = 10, 5%) (Fig 2). We also depicted pathway diagrams of four canonical pathways: canonical WNT, cell cycle, PI3K, and RTK-RAS (S4 Fig) [25]. In the canonical WNT pathway, we observed rates of 3% for SNVs in CTNNB1 (n = 6, 5 missense mutations, 1 nonsense mutation), 4% for SNVs in APC (n = 8, 2 missense mutations, 2 nonsense mutations, 3 frameshift indels, 1 splice site mutation), 1% for SNVs in AMER1 (n = 1 missense mutation), and 1% for CNVs in APC (n = 2, 1 gains with 3 copies, 1 loss with 1 copy) (S4A Fig).

Gene mutation mapper plot of <i>EGFR</i>, <i>TP53</i>, <i>PIK3CA</i>, and <i>KRAS</i>.
Fig. 2. Gene mutation mapper plot of EGFR, TP53, PIK3CA, and KRAS.
(A) Among EGFR mutations, L858R/M and exon 19 deletion were the most common alterations, followed by L861Q/R. (B) In TP53, P112S, V118F, R136H, N171fs, H175R, G206C/V, R234H, and E246K mutations were identified at similar rates. (C) In PIK3CA, the established canonical E542K missense mutation was the most common. (D) Among KRAS mutations in codon 12, G12V/D/C/S/A was the most common.

Clinical implication with somatic mutation classification system for LUAD

We attempted to implement a precision medicine approach for application in the clinical field. The purpose of precision medicine through NGS is to determine the link between each mutation with an associated targeted therapy and the clinical outcome in cancer patients. Although there are many clinical annotation databases for various somatic mutations, the determination of which mutations have clinical implications differs slightly in each. Hence, a harmonized system for a meta-knowledgebase of clinical interpretations of cancer genomic variants is required to reliably determine clinical implications for as many patients as possible[26]. Of 192 LUAD patient samples, 121 samples (63%) were clinically annotated in CIVic[27], validated through various publications and clinical trials, and annotated through CGI[23], resulting in the annotation of 151 samples (79%). Potential targets that still remain are annotated with CRAVAT[28] (155 samples, 81%), which involves computational prediction (Fig 3A). There were a total of 86 samples annotated in the three databases, 3 of which were annotated only in CIVic, 11 only in CGI, and 4 only in CRAVAT (Fig 3B). The somatic mutations reported in CIVic were the well-known EGFR L858R, exon 19 deletion, and T790M mutations; the G12V/D/C/S/A mutation in KRAS; E542K in PIK3CA; and Y220C and R175H in TP53. Genes with CNVs included CDKN2A, EGFR, and PTEN, among others. Somatic mutations annotated only in CGI were ARID1A, BRAF, BRCA, STK11, and BAP1, while SETD2 and STK11 were the annotated somatic CNVs. The somatic mutations independently estimated by CRAVAT were H179R and G245C for TP53 and P750R for DNMT3A. Prospective application of this approach should be assessed in a future umbrella trial of lung cancer patients.

Mutation classification system.
Fig. 3. Mutation classification system.
(A) Hierarchical mutation classification system based on knowledgebase database and the computational prediction algorithm. (B) Venn diagram to confirm how many ranked mutations were included.

Clinical correlation

Among 192 patients with available NGS and survival data, TP53 (p = 0.062), EGFR (p = 0.299), the RTK-RAS pathway (p = 0.089), and the PI3K pathway (p = 0.149) were not associated with shorter DFS (S5 Fig); however, mutations in APC (8 samples with 2 missense mutations, 2 nonsense mutations, 3 frame shift indels, and 1 splice site mutation; 2 samples with 1 gain and 1 loss), CTNNB1 (6 samples with 5 missense mutations and 1 nonsense mutation), and AMER1 (1 sample with 1 missense mutation) in the canonical WNT pathway were associated with shorter DFS (p = 0.018) (Fig 4). In addition, based on TMB annotations, cut-offs were used to divide patients into tertiles of low (≤ 2 mt/Mb, n = 88), intermediate (> 2 to ≤ 7 mt/Mb, n = 81), and high TMB (> 7 mt/Mb, n = 23) groups, and these were associated with differences in DFS. Patients with a low TMB showed better prognosis than those with high or intermediate TMB (p = 0.041) (Fig 5A). The low TMB group had more EGFR exon 19 deletions than the other groups (36%). In the intermediate TMB group, EGFR L858R was the most common mutation (30%) (Fig 5B).

Disease-free survival (DFS) by somatic mutation in the canonical WNT pathway (<i>p</i> = 0.018).
Fig. 4. Disease-free survival (DFS) by somatic mutation in the canonical WNT pathway (p = 0.018).
APC, CTNNB1, and AMER1 were mutated in the WNT pathway.
Disease-free survival (DFS) by tumor mutation burden (TMB).
Fig. 5. Disease-free survival (DFS) by tumor mutation burden (TMB).
(A) Kaplan-Meier plots showing prognostic effect of nonsynonymous TMB on disease-free survival (p = 0.041). (B) The low TMB group had more EGFR exon 19 deletions among various mutations than other groups (36%). In the intermediate TMB group, EGFR L858R was the most common (30%). Fisher’s exact test was used to compare low and intermediate TMB groups for the EGFR exon 19 deletion (p = 0.041) and EGFR L858R variants (p < 0.001). No significant differences were found for other comparisons.

Discussion

Our study shows that it is feasible to incorporate NGS into the clinical care of lung cancer patients. Through our NGS analysis, the most common genomic alterations (EGFR, TP53, ADGRV1, and SMARCA2) were slightly different from those observed in present investigations of LUAD in Caucasian [2]. In The Cancer Genome Atlas (TCGA), the rate of KRAS mutation in LUAD is 33%, while that of EGFR mutation is only 14%[2]. It should be noted that we have a higher proportion of female (56.2%) and non-smokers (62.2%) than is found in TCGA. However, the most prominent difference is the ethnicity of the patients. Only eight Asian patients are included in TCGA[2]. We analyzed only ethnic Korean patient samples and can conclude that EGFR mutation is the most common (55%) in Koreans, based on the current study and a rate of 59% among Asian patients with LUAD in previous reports [29]. Since KRAS mutation occurs exclusive of EGFR mutation, KRAS mutations are slightly less frequent in ethnic Koreans than in Caucasian patients. Luo published the results of whole genome sequencing for young never-smoked Asian with lung adenocarcinoma[30]. Compared with this study, we conducted in a more practical way by targeted sequencing with FFPE. In Luo study, EGFR mutation was found to be somatic SNV 25% and CNV 19% but ours was 64%, 15% respectively. And KRAS of Luo study was sSNV 11% but ours was 2%. Interestingly, several genes showed almost the same ratio (TP53 sSNV, Luo 28% vs. ours 31%; MYC sCNV, 14% vs. 10%; TERT sCNV, 17% vs. 17%, respectively).

Similar to the discovery of EGFR, ALK, and ROS1, various studies for identifying molecular characterizations of LUAD are under way, and our study is also part of this effort. Mutations in specific genes affect not only the carcinogenic process but also dysfunction of signaling pathways and can be important mediators in tumorigenesis[31]. The WNT pathway is involved in the formation of lung homeostasis and tumor angiogenesis[32]. WNT pathway aberrations are potential therapeutic targets in lung cancer patients[33]. The most studied WNT pathway mutations in cancers include sporadic mutations in APC and β-catenin genes. Since APC is part of the degradation scaffold for β-catenin, mutations of APC result in reduced degradation and increased nuclear accumulation of β-catenin, leading to activation of target oncogenes including cyclin D1 and c-Myc[33]. Clinical trials of WNT signaling pathway inhibitors have been conducted in advanced solid tumors (NCT03355066). Our analysis also shows that patients with APC, CTNNB1, and AMER1 mutations in the WNT pathway show shorter DFS compared to wild-type patients (Fig 4). In addition, we investigated the clinical significance of TMB in patients with LUAD and examined the relationship between TMB and prognosis. TMB is thought to be associated with the amount of tumor neoantigen and to have an important role in predicting the effect of immune checkpoint inhibitors[34]. We found that smokers had a significantly higher TMB than never-smokers (average 4.84 vs. 2.84 mt/Mb, respectively, p = 0.019). Devarakonda et al. also annotated a TMB greater than 8 mt/Mb as high and reported a better prognosis in this group[35]. On the other hand, Owada-Ozaki reported that shorter OS and DFS was associated with high TMB in stage I NSCLC[34]. In our data, patients with a TMB < 2 mt/Mb showed longer DFS than patients with a TMB ≥ 7 mt/Mb (p = 0.041) (Fig 5A). Since there are still many conflicting results, further studies are needed to validate TMB as a prognostic marker. Notably, exon 19 deletion was the most common mutation in the low TMB group, which exhibited good prognosis. It is already known that exon 19 deletion results in a better prognosis than other EGFR mutations[36] (Fig 5B).

In order for the above analyses to be applied to clinical practice, appropriate use of a meta-knowledgebase of clinical implications of cancer genomic variants is necessary[37]. A meta-knowledge-based framework of holistic interpretation comprehensively covers hundreds of genes, disease and drugs. Hence, we included predicted target mutations in CRAVAT, as well as providing annotations via CIVic and CGI. Overall, this methodology may expedite the widespread implementation of an umbrella trial of lung cancer patients.

Several technical limitations were identified in this study. First, a low tumor cellularity in samples, owing to normal cell contaminants, and high levels of intra-tumor heterogeneity make it difficult to accurately call SNVs and CNVs. For this reason, the variant allele frequency was lower than the theoretical value of 0.5 (S6 Fig). Second, targeted sequencing for the identification of CNVs remains a secondary option when more sensitive methods, such as whole-genome sequencing or specialized array-based methods, are unavailable. As targeted sequencing-based CNV analysis generally performs better in a larger cohort, the size and sustainability of clinical trials should be considered when they are designed. Third, the NGS platform used in this study detected only SNVs and CNVs although diverse structural variations and epigenetic events exist outside of the captured exons. Active participation of genome analysis experts is strongly recommended to manage these technical issues. Finally, since we used only 242 genes in this study, other factors including genetic alteration in other genes, epigenetic alterations, gene and protein expression may be related to LUAD risk. There are recent reports that exposure to outdoor particulate matter (PM10)[38, 39] or indoor secondhand smoke and high temperature cooking oil fumes[40] are associated with lung cancer. Since, there were inadequate information for patient’s dwelling or occupation, it was precluded to analyze environmental factor.

Conclusions

In conclusion, targeted sequencing using NGS can provide clinically relevant mutation profiling information from readily available FFPE tissues. EGFR was the most frequently mutated gene (55%), followed by TP53 (35%) and KRAS (6%). This may assist in decision to the use of innovative clinical trials of genotype-matched drugs and provide benefits to many cancer patients.

Supporting information

S1 Fig [tif]
Disease-free survival (DFS) by stage in lung adenocarcinoma (LUAD) patients ( < 0.001).

S2 Fig [a]
Comparison of genetic alteration pattern between current study and TCGA data.

S3 Fig [a]
Co-occurrence and tumor mutation burden (TMB) test.

S4 Fig [a]
Pathway diagrams.

S5 Fig [dfs]

S6 Fig [tif]
Variant allele frequency (VAF).

S1 Table [xls]
A list of 242 genes and information of genomic location used for targeted sequencing.

S2 Table [xlsx]
Individual sample lists including detailed clinical information.


Zdroje

1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59(4):225–49. Epub 2009/05/29. doi: 10.3322/caac.20006 19474385.

2. The Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543. doi: 10.1038/nature13385 https://www.nature.com/articles/nature13385#supplementary-information. 25079552

3. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350(21):2129–39. Epub 2004/05/01. doi: 10.1056/NEJMoa040938 15118073.

4. Chan BA, Hughes BGM. Targeted therapy for non-small cell lung cancer: current standards and the promise of the future. Translational lung cancer research. 2015;4(1):36–54. doi: 10.3978/j.issn.2218-6751.2014.05.01 25806345.

5. Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Annals of Oncology. 2013;24(9):2371–6. doi: 10.1093/annonc/mdt205 23723294

6. Soo RA, Kawaguchi T, Loh M, Ou SH, Shieh MP, Cho BC, et al. Differences in outcome and toxicity between Asian and caucasian patients with lung cancer treated with systemic therapy. Future Oncol. 2012;8(4):451–62. Epub 2012/04/21. doi: 10.2217/fon.12.25 22515448.

7. Lim SM, Kim HR, Cho EK, Min YJ, Ahn JS, Ahn M-J, et al. Targeted sequencing identifies genetic alterations that confer primary resistance to EGFR tyrosine kinase inhibitor (Korean Lung Cancer Consortium). Oncotarget. 2016;7(24):36311–20. doi: 10.18632/oncotarget.8904 27121209.

8. Ahn JW, Kim HS, Yoon J-K, Jang H, Han SM, Eun S, et al. Identification of somatic mutations in EGFR/KRAS/ALK-negative lung adenocarcinoma in never-smokers. Genome Medicine. 2014;6(2):18. doi: 10.1186/gm535 24576404

9. Joshi NA, Fass JN. Sickle: A sliding-window, adaptive, quality-based trimming tool for FastQ files (Version 1.33) [Software]. 2011.

10. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60. Epub 2009/05/20. doi: 10.1093/bioinformatics/btp324 19451168; PubMed Central PMCID: PMC2705234.

11. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303. Epub 2010/07/21. doi: 10.1101/gr.107524.110 20644199; PubMed Central PMCID: PMC2928508.

12. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31(3):213–9. Epub 2013/02/12. doi: 10.1038/nbt.2514 23396013; PubMed Central PMCID: PMC3833702.

13. Narzisi G, O'Rawe JA, Iossifov I, Fang H, Lee YH, Wang ZH, et al. Accurate de novo and transmitted indel detection in exome-capture data using microassembly. Nature Methods. 2014;11(10):1033–6. doi: 10.1038/nmeth.3069 PubMed PMID: WOS:000342719100021. 25128977

14. Costello M, Pugh TJ, Fennell TJ, Stewart C, Lichtenstein L, Meldrim JC, et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Research. 2013;41(6):e67–e. doi: 10.1093/nar/gks1443 PubMed PMID: PMC3616734. 23303777

15. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS Comput Biol. 2016;12(4):e1004873. Epub 2016/04/23. doi: 10.1371/journal.pcbi.1004873 27100738; PubMed Central PMCID: PMC4839673.

16. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164. Epub 2010/07/06. doi: 10.1093/nar/gkq603 20601685; PubMed Central PMCID: PMC2938201.

17. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285–91. Epub 2016/08/19. doi: 10.1038/nature19057 27535533; PubMed Central PMCID: PMC5018207.

18. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847–9. doi: 10.1093/bioinformatics/btw313 27207943

19. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28(11):1747–56. Epub 2018/10/21. doi: 10.1101/gr.239244.118 30341162; PubMed Central PMCID: PMC6211645.

20. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4. Epub 2012/05/17. doi: 10.1158/2159-8290.CD-12-0095 22588877; PubMed Central PMCID: PMC3956037.

21. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1. Epub 2013/04/04. doi: 10.1126/scisignal.2004088 23550210; PubMed Central PMCID: PMC4160307.

22. Bahceci I, Dogrusoz U, La KC, Babur O, Gao J, Schultz N. PathwayMapper: a collaborative visual web editor for cancer pathways and genomic data. Bioinformatics. 2017;33(14):2238–40. Epub 2017/03/24. doi: 10.1093/bioinformatics/btx149 28334343; PubMed Central PMCID: PMC5859976.

23. Tamborero D, Rubio-Perez C, Deu-Pons J, Schroeder MP, Vivancos A, Rovira A, et al. Cancer Genome Interpreter annotates the biological and clinical relevance of tumor alterations. Genome Med. 2018;10(1):25. Epub 2018/03/30. doi: 10.1186/s13073-018-0531-8 29592813; PubMed Central PMCID: PMC5875005.

24. Oliveros JC. VENNY. An interactive tool for comparing lists with Venn diagrams. 2007.

25. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173(2):321-37.e10. Epub 2018/04/07. doi: 10.1016/j.cell.2018.03.035 29625050; PubMed Central PMCID: PMC6070353.

26. Van Allen EM, Wagle N, Stojanov P, Perrin DL, Cibulskis K, Marlow S, et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat Med. 2014;20(6):682–8. Epub 2014/05/20. doi: 10.1038/nm.3559 24836576; PubMed Central PMCID: PMC4048335.

27. Griffith M, Spies NC, Krysiak K, McMichael JF, Coffman AC, Danos AM, et al. CIViC is a community knowledgebase for expert crowdsourcing the clinical interpretation of variants in cancer. Nat Genet. 2017;49(2):170–4. Epub 2017/02/01. doi: 10.1038/ng.3774 28138153; PubMed Central PMCID: PMC5367263.

28. Douville C, Carter H, Kim R, Niknafs N, Diekhans M, Stenson PD, et al. CRAVAT: cancer-related analysis of variants toolkit. Bioinformatics. 2013;29(5):647–8. Epub 2013/01/18. doi: 10.1093/bioinformatics/btt017 23325621; PubMed Central PMCID: PMC3582272.

29. Li S, Choi YL, Gong Z, Liu X, Lira M, Kan Z, et al. Comprehensive Characterization of Oncogenic Drivers in Asian Lung Adenocarcinoma. J Thorac Oncol. 2016;11(12):2129–40. Epub 2016/09/13. doi: 10.1016/j.jtho.2016.08.142 27615396.

30. Luo W, Tian P, Wang Y, Xu H, Chen L, Tang C, et al. Characteristics of genomic alterations of lung adenocarcinoma in young never-smokers. International Journal of Cancer. 2018;143(7):1696–705. doi: 10.1002/ijc.31542 29667179

31. Tsimberidou AM. Targeted therapy in cancer. Cancer Chemother Pharmacol. 2015;76(6):1113–32. Epub 2015/09/24. doi: 10.1007/s00280-015-2861-1 26391154; PubMed Central PMCID: PMC4998041.

32. Stewart DJ. Wnt signaling pathway in non-small cell lung cancer. J Natl Cancer Inst. 2014;106(1):djt356. Epub 2013/12/07. doi: 10.1093/jnci/djt356 24309006.

33. Rapp J, Jaromi L, Kvell K, Miskei G, Pongracz JE. WNT signaling–lung cancer is no exception. Respiratory Research. 2017;18:167. doi: 10.1186/s12931-017-0650-6 PubMed PMID: PMC5584342. 28870231

34. Owada-Ozaki Y, Muto S, Takagi H, Inoue T, Watanabe Y, Fukuhara M, et al. Prognostic Impact of Tumor Mutation Burden in Patients With Completely Resected Non-Small Cell Lung Cancer: Brief Report. J Thorac Oncol. 2018;13(8):1217–21. Epub 2018/04/15. doi: 10.1016/j.jtho.2018.04.003 29654927.

35. Devarakonda S, Rotolo F, Tsao M-S, Lanc I, Brambilla E, Masood A, et al. Tumor Mutation Burden as a Biomarker in Resected Non–Small-Cell Lung Cancer. Journal of Clinical Oncology. 2018;36(30):2995–3006. doi: 10.1200/JCO.2018.78.1963 30106638

36. Choi YW, Jeon SY, Jeong GS, Lee HW, Jeong SH, Kang SY, et al. EGFR Exon 19 Deletion is Associated With Favorable Overall Survival After First-line Gefitinib Therapy in Advanced Non-Small Cell Lung Cancer Patients. Am J Clin Oncol. 2018;41(4):385–90. Epub 2016/03/12. doi: 10.1097/COC.0000000000000282 26967328.

37. Wagner AH, Walsh B, Mayfield G, Tamborero D, Sonkin D, Krysiak K, et al. A harmonized meta-knowledgebase of clinical interpretations of cancer genomic variants. bioRxiv. 2018:366856. doi: 10.1101/366856

38. Lamichhane DK, Kim HC, Choi CM, Shin MH, Shim YM, Leem JH, et al. Lung Cancer Risk and Residential Exposure to Air Pollution: A Korean Population-Based Case-Control Study. Yonsei Med J. 2017;58(6):1111–8. Epub 2017/10/20. doi: 10.3349/ymj.2017.58.6.1111 29047234; PubMed Central PMCID: PMC5653475.

39. Consonni D, Carugno M, De Matteis S, Nordio F, Randi G, Bazzano M, et al. Outdoor particulate matter (PM10) exposure and lung cancer risk in the EAGLE study. PLoS One. 2018;13(9):e0203539. Epub 2018/09/15. doi: 10.1371/journal.pone.0203539 30216350; PubMed Central PMCID: PMC6157824.

40. Jin ZY, Wu M, Han RQ, Zhang XF, Wang XS, Liu AM, et al. Household ventilation may reduce effects of indoor air pollutants for prevention of lung cancer: a case-control study in a Chinese population. PLoS One. 2014;9(7):e102685. Epub 2014/07/16. doi: 10.1371/journal.pone.0102685 25019554; PubMed Central PMCID: PMC4097600.


Článok vyšiel v časopise

PLOS One


2019 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#