Back to Journals » OncoTargets and Therapy » Volume 14

High Tumor Mutation Burden and DNA Repair Gene Mutations are Associated with Primary Resistance to Crizotinib in ALK-Rearranged Lung Cancer

Authors Xiao D, Deng Q, He D, Huang Y, Liang W, Wang F, Yang H

Received 18 June 2021

Accepted for publication 31 August 2021

Published 15 September 2021 Volume 2021:14 Pages 4809—4817

DOI https://doi.org/10.2147/OTT.S325443

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Arseniy Yuzhalin



Dakai Xiao,1,* Qiuhua Deng,1,* Dongyun He,2,* Ying Huang,2 Wenchi Liang,2 Fengnan Wang,2 Haihong Yang2

1Research Center forTranslational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People’s Republic of China; 2Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Haihong Yang
Department of Thoracic Oncology, State Key Laboratory of Respiratory Diseases, National Clinical Research Center of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, People’s Republic of China
Email [email protected]

Background: About 20% of patients with ALK-rearranged non-small cell lung cancer (NSCLC) develop acquired resistance to tyrosine kinase inhibitor (TKI) during the first 6 months. This study aimed to examine the molecular mechanisms of early TKI resistance and prognosis in ALK-rearranged NSCLC.
Methods: Ten patients with ALK-rearranged NSCLC were included: five who developed rapid resistance to crizotinib (progression-free survival (PFS) ≤ 3 months) and five who exhibited a good response to crizotinib (PFS ≥ 36 months). The tumor specimens were subjected to whole-exome sequencing (WES). The validation cohort included 19 patients with ALK-rearranged NSCLC who received crizotinib; targeted sequencing of 43 selected genes was performed. The effect of the TP53 G245S mutation on crizotinib sensitivity was tested in H3122 cells.
Results: Mutations in DNA repair-associated genes were identified in primary resistance to crizotinib. Patients with a poor response to crizotinib harbored a greater burden of somatic mutations than those with a good response [median somatic mutations, 136 (range, 72– 180) vs 31 (range, 10– 48)]. Compared with the patients carrying wild-type TP53 or TP53 exon 3 deletion, 29 patients with TP53 G245S mutation showed a shorter survival time (P < 0.05), with a median PFS of 3 (95% CI: 1.9– 4.1) months and a median overall survival of 7 (95% CI: 3.4– 10.5) months. TP53 mutation promoted the proliferation of EML4-ALK-rearranged H3122 cells by approximately 3 folds (P < 0.001). H3122 cells with TP53 mutant were more sensitive to crizotinib compared with control cells.
Conclusion: A higher mutation burden and mutations in DNA repair gene, including TP53, were potentially associated with primary resistance to crizotinib in ALK-rearranged NSCLC. An immune-checkpoint inhibition strategy could be examined, which might overcome primary resistance to crizotinib in ALK-rearranged NSCLC.

Keywords: ALK, non-small cell lung cancer, TKI, resistance, prognosis

Introduction

Anaplastic lymphoma kinase (ALK)‑fusion genes represent a small but important part of oncogenic driver mutations in NSCLC, accounting for approximately 3%‑7% of all cases worldwide.1,2 Small molecule tyrosine kinase inhibitors (TKIs) are the standard therapy for ALK-rearranged NSCLC. Crizotinib, a first-generation TKI, is the most widely used targeted drug to treat ALK-rearranged NSCLC. Patients receiving first-line crizotinib can survive for more than 4 years,3 even 5–7 years after sequential treatment with next-generation TKIs.4,5 Nevertheless, based on the ALEX study,6 about 20% of patients with ALK-rearranged NSCLC developed acquired resistance to first-generation TKI (crizotinib) or second-generation TKI (alectinib) during the first 6 months of treatment and survived no more than 12 months. The ALEX study6 suggested that patients with early resistance to TKIs had a poor prognosis.

The resistance to first- and second-generation TKIs is mediated by a variety of mechanisms, including secondary ALK mutations in 20–30% of cases, ALK-rearranged gene amplification in 10% of cases, and activation of alternative signaling pathways and wild-type resistance in other cases.7–9 The three main ALK resistance mutations are L1196M, C1156Y, and G1202R, and not all ALK TKIs are active against them. In addition, other mutations may also lead to resistance to specific TKIs.10,11 Crizotinib resistance can be overcome in many cases by using next-generation TKIs, and the overall response rate (ORR) is as high as 70%–80%.12,13 Nevertheless, patients with early acquired resistance to crizotinib often fail to respond to next-generation TKIs and display a short survival. EML4-ALK variant 3a/b and high levels of p‑c‑Kit might be associated with TKI resistance and short survival,14,15 but little is known about the molecular mechanism of early TKI resistance or poor prognosis in patients with ALK fusion NSCLC.

Whole-exome sequencing (WES) is a powerful tool for identifying genetic variants related to cancer and treatment outcomes. It can provide biologically relevant information about genetic predisposition to cancer occurrence,16,17 development of metastasis,18 resistance to treatments,19 and the best treatment options.20 On the other hand, WES has the disadvantage of revealing many genetic variants with unknown biological meanings. It is necessary to process a large amount of data to determine the pathogenicity of specific genetic variants, and a lot of effort has been made to achieve this goal.20,21 One study used WES to examine genetic variants associated with the inflammatory microenvironment of NSCLC.22

Therefore, this study aimed to gain a comprehensive understanding of the possible molecular mechanisms of early TKI resistance and prognosis in ALK-rearranged NSCLC by using WES in lung adenocarcinoma specimens with early failure (≤3 months) to crizotinib vs very good response (≥36 months) to crizotinib. In addition, the identified genetic variants were validated by targeted sequencing in specimens from other NSCLC patients who received crizotinib.

Materials and Methods

Patients and Specimens

Patients with ALK-rearranged NSCLC who received crizotinib as first-line treatment were included. Nineteen validation patients came from a cohort of 87 ALK-positive NSCLC patients who received crizotinib. All patients were admitted to the First Affiliated Hospital of Guangzhou Medical University from August 2014 to February 2018 and were pathologically confirmed with stage IV NSCLC. The study was approved by the ethics committee of the First Affiliated Hospital of Guangzhou Medical University and conducted in accordance with the Declaration of Helsinki.

HE-stained sections from each patient specimen were subjected to an independent pathology review to confirm that the tumor specimen was histologically consistent with NSCLC (>70% tumor cells). ALK rearrangement was determined using VENTANA immunohistochemical system (Roche Diagnostics, Basel, Switzerland) or PCR (Amoy Diagnostics Company, Haicang, China). WES was performed on specimens from five patients with very early failure (≤3 months) to crizotinib and five patients with a very good response (≥36 months) to crizotinib. Probe capture sequencing was conducted on specimens from 19 validation patients who received crizotinib.

The progression-free time (PFS) was measured from the first treatment until progression or death. The overall survival (OS) was measured from the first treatment until death.

DNA Extraction and Next-Generation Sequencing

DNA was extracted from paraffin-embedded lung tissues using the QIAamp DNA Kit (Qiagen, Venlo, The Netherlands). The qualified genomic DNA samples were randomly fragmented using the Covaris technology, and the size of the library fragments was mainly distributed between 200 and 300 bp. The extracted DNA was amplified and purified. Genomic DNA was captured on the IDT xGen Lockdown probe for WES (Integrated DNA Technologies, Inc., Coralville, IA, USA) or IDT Individually Synthesized Panel for target region sequencing, according to the manufacturer’s instructions. Then, high-throughput sequencing of the library was performed using the Illumina Novaseq 6000 platform (Illumina, Inc., San Diego, CA, USA).

BWA (version 0.7.17-r1188) was used to align the short sequence reads to the hg19 human reference genome (NCBI build 37). The generated .sam file was converted to the .bam format using the SAMtools software (version 1.7). The raw .bam file was deduped using fgbio (version 0.12) and a unique molecular barcode sequence. After obtaining the deduped .bam file, fgbio was used to call the genetic variants from the .bam file with the recommended parameters.

The tumor mutational burden (TMB) was calculated as previously reported.23 The signaling pathways of the variants were analyzed using the KEGG database. The predicted protein functions were subjected to the online STRING database analysis (https://string-db.org/).

Cell Culture and P53 G245S Transfection

The lung cancer cell line H3122 (EML4-ALK fusion) was obtained from Dana–Farber Cancer Institute (Boston, MA), which was cultured in RPMI-1640 medium supplemented with 10% FBS and kanamycin/penicillin. Crizotinib was purchased from Selleck (Houston, TX, USA) and dissolved in DMSO. The wild-type TP53 fragment was amplified from normal lung tissue using the forward primer 5ʹ-TCT AGA GCC ACC ATG GAG GAG CCG CAG TCA GAT CC-3ʹ and reverse primer 5ʹ-GGA TCC TCA CTT ATC GTC GTC ATC CTT GTA ATC GTC TGA GTC AGG CCC TTC TGT C-3ʹ. The fragment was ligated into pGEM-T easy (Promega, Madison, WI, USA) to generate pGEM-T-TP53. The TP53 mutant p53 G245S was generated by site-directed mutagenesis (Agilent Technologies, Santa Clara, CA, USA). PCR was performed with pGEM-T-TP53 as a template and by using the mutagenic primers: 5ʹ-Cct ccg gtt cat gct gcc cat gca gga ac-3ʹ and 5ʹ-gtt cct gca tgg gca gca tga acc gga gg-3ʹ. The purified fragments of WT and mutant TP53 were inserted into the lentiviral expression vector pCDH-CMV-MCS-EF1-GFP-Puro (System Biosciences, Palo Alto, CA, USA). The lentiviral-based expression vector and the packaging vectors psPAX2 and pMD2.G were transiently transfected into 293T human embryonic kidney cells using Lipofectamine 2000 (Life Technologies Co., Grand Island, NY, USA) (Supplemental Methods). The lentiviruses were transduced into H3122 cells in the presence of polybrene (8 μg/mL).

MTT Assay

Cell proliferation was determined using the MTT assay. Cells seeded in 96-well plates (3×103 cells/well) were treated with various concentrations of crizotinib. After 48 h, the cells were incubated with MTT (1 mg/mL) for 3 h. The absorbance was determined at 570 nm with the cell imaging multi-mode microplate reader (Cytation 3, Bio-Tek, Winooski, VT, USA).

Statistical Analysis

Continuous data were analyzed using the Mann–Whitney U-test. Fisher’s exact test or chi-square test was used to compare the categorical data between the two groups. The Kaplan–Meier method was used to analyze the PFS and OS. Statistical analyses were carried out using SPSS 22.0 (IBM Corp., Armonk, NY, USA). Statistical significance was defined as a two-tailed P-value <0.05.

Results

Characteristics of the Patients

Eighty-seven NSCLC patients with ALK rearrangement who received crizotinib were included. In order to investigate the molecular mechanism underlying the primary or rapid resistance to crizotinib, ten ALK-rearranged NSCLC patients were selected based on their response to crizotinib. Five patients showed an extremely poor response, while the other five patients exhibited a good response to crizotinib. As shown in Table S1, the five poor responders (median PFS 2±1 months) had a dramatically shorter PFS than those with a good response (median PFS 36±12.8 months). There were no significant differences in age, sex, and other clinical characteristics between the two groups. Only 19 NSCLC patients had complete follow-up data and qualified samples for the validation study. For the validation cohort of 19 NSCLC patients with ALK rearrangement, the median PFS and OS were 15 (range, 1–44) and 19 (range, 2–44) months, respectively (Table 1).

Table 1 Clinical Characteristics of Patients in the Validation Cohort (n=19)

Identification of Somatic Genetic Alterations Involved in Primary Resistance to Crizotinib by WES

WES was performed on the tumor specimens from 10 NSCLC patients with a good or poor response to crizotinib. The whole-exome was sequenced to a mean depth of 828×, covering approximately 0.6–0.8 million small indels and 3.9–4.1 million single nucleotide variants (SNVs). The specimens harbored an average of 264 somatic mutations, which were differently distributed between the good and poor response groups. The number of somatic coding mutations was higher in the poor response group (P < 0.001). The median number of co-mutations was 31 (range, 10 to 48) in the good response group and 136 (range, 72 to 180) in the poor response group (Figure 1). The poor response group had a higher TMB than the good response group (median TMB, 18 vs 8, P=0.002) (Table S1). Hence, NSCLC that responded poorly to crizotinib harbored more mutations than NSCLC with a good response to the drug.

Figure 1 The number of co-mutations in groups with a poor or good response to crizotinib.

Some genes that were frequently mutated were identified. According to the KEGG database, the affected pathways included mitochondrial apoptosis, vascular endothelial growth factor (VEGF) angiogenesis, DNA repair, and platinum resistance (Figure S1). DNA mismatch repair-related gene variations such as TP53, MLH1, MSH2, and XPA were associated with survival or drug responsiveness. Interestingly, the results of the pathway analysis were in accordance with the functional protein analysis in the STRING database (Figure S2). It suggested that these genetic variations in patients with ALK-positive NSCLC were involved in different responses to crizotinib treatment or survival, and TP53 was the key player in these variations.

Next-Generation Sequencing for Validation

In order to validate the gene mutations identified by WES, 43 frequently mutated genes associated with tumor growth, metastasis, and drug therapy (Table S2) were selected for hybrid-recapture-based targeted sequencing in a cohort of 19 NSCLC patients with ALK-rearrangement. A total of 774 genomic variations could be matched to the crizotinib responses and were located within regions of DNA repair, mitochondrial apoptosis, and tumor angiogenesis-target genes. Among these variations, 20 were located in the coding regions of 18 genes, including SNV, frameshift deletion, and stopgain, and were identified in 19 patients treated with crizotinib (Figure 2). The TP53 exon 3 G245S mutation and frameshift deletion were among them.

Figure 2 Next-generation sequencing (NGS) was performed on an individually synthesized panel containing 43 target gene regions in 19 validation patients. A total of 774 genomic variations could be matched to the crizotinib responses and were located within regions of DNA repair, mitochondrial apoptosis, and tumor angiogenesis target genes.

TP53 Mutation and Survival

Compared with patients with wild type TP53 or exon 3 deletion, 29 patients carrying the TP53 G245S mutation showed a shorter survival time after crizotinib treatment (P < 0.05), with a median PFS of 3 (95% CI: 1.9–4.1) months (Figure 3A), and a median OS of 7 (95% CI:3.4–10.5) months (Figure 3B). None of these patients survived more than 12 months. There was no significant difference in PFS or OS between the TP53 exon 3 deletion and wild-type group.

Figure 3 Survival analysis. The Kaplan–Meier curve showed the survival of three subgroups of NSCLC patients: patients with wild-type TP53, TP53 G245S mutation, and TP53 exon 3 deletion. (A) Progression-free survival (PFS) curve. (B) Overall survival (OS) curve. The statistical difference was shown on the graph.

Functional Analysis of TP53 G245S Mutation in Lung Cancer Cell

In order to determine the effect of TP53 mutation on the response of EML4-ALK rearranged H3122 cells to crizotinib, cells stably expressing wild type or mutant TP53 (Figure 4A) were established. The TP53 mutation promoted the proliferation of H3122 cells by approximately 3 folds (Figure 4B) (P < 0.001). When the cells were treated with crizotinib, the TP53 mutant cells were more sensitive to crizotinib compared with the control cells (Figure 4C).

Figure 4 The effect of TP53 G245S mutation on the response of H3122 cells to crizotinib. Statistical significance was determined by unpaired t-tests, ***P<0.001. (A) Western blot of p53 expression in H3122 cells stably expressing wild type TP53 (control, EV) or TP53 G245S mutant. (B) Cell viability assay in control and mutant H3122 cells. (C) Sensitivity of control and mutant H3122 cells to crizotinib.

Discussion

About 20% of patients with ALK-rearranged NSCLC develop rapid TKI primary resistance during the first 6 months of treatment.6 This study aimed to examine the molecular mechanisms of early TKI resistance and prognosis in ALK-rearranged NSCLC. The results suggested that high TMB and mutations in DNA repair genes (including TP53 G245S) in ALK-positive NSCLC conferred rapid resistance to crizotinib.

To date, the mechanisms of primary resistance to ALK-TKIs have mainly focused on secondary mutations in the ALK gene. In this study, we observed that a subset of patients had an extremely poor response to crizotinib (PFS <3 months) with poor survival (no longer than 6 months) compared with the literature.2–6 Although they did not receive subsequent next-generation TKIs, their survival rate was worse than patients who only received crizotinib followed by chemotherapy, with a median survival of 20 months.3 In the ALEX study, among patients who received crizotinib and alectinib, patients with early resistance to TKI had shorter survival (no longer than 12 months).6 Therefore, we hypothesized that patients with early resistance to ALK-TKI had a poor prognosis, and some cases might be caused by mutations in genes other than ALK.

In the WES analysis, 264 somatic coding mutations were identified. The results showed that tumors that responded poorly to crizotinib exhibited a higher number of somatic mutations and higher TMB compared with tumors that responded well. In many cancer types, higher TMB was associated with poorer survival, in contrast to ICI-treated patients in whom higher TMB was associated with longer survival, reported by Valero et al.24 Similar findings have previously been shown in EGFR-mutant lung cancer. For instance, in TKI treatment, a higher number of concurrent driver gene mutations in patients with EGFR-mutant NSCLC was reported to be associated with poor PFS.25 As a novel biomarker for immune checkpoint inhibitors, TMB is lower in EGFR-mutant or ALK-rearranged NSCLC. Using the Cancer Genome Atlas (TCGA) database, TMB has also been identified as a negative prognostic biomarker for OS in EGFR-mutated patients, especially those with P53 mutations.26

We selected 43 genes covering 774 genomic variations for validation. Those genes were selected because of their known involvement in drug resistance, tumor prognosis, mitochondrial apoptosis, VEGF angiogenesis, DNA repair, and platinum-resistance pathway.27–31 It is worthwhile to note that TP53 mutations are often associated with high TMB.26 DNA repair-related gene variations, including MLH1, MSH2, and XPA, are associated with chemotherapy resistance. As previously reported, variations in these genes are important predictors of superior response to immune checkpoint inhibitors.30–33 It is plausible that TP53 or DNA repair-related mutations and high TMB are associated with the early resistance to crizotinib or poor prognosis. The results suggested that the subset of those patients might benefit from immune checkpoint inhibitors combined with TKI, which warranted further investigation.

In the validation analysis, the TP53 G245S mutation indicated extremely poor PFS and OS after crizotinib treatment. It was still not clear whether the TP53 G245S mutation affected TKI sensitivity or prognosis, or both. In this context, we sought to investigate the function of the TP53 G245S mutation in EML4-ALK fusion lung cancer cells. The results demonstrated that the TP53 G245S mutation promoted the proliferation of H3122 cells but did not show resistance to crizotinib compared with controls. Those results indicated that early TKI resistance is probably a complex event involving intrinsic and extrinsic factors. Only TP53 G245S is not enough to cause TKI resistance. We need to perform more research on DNA repair deficiency and TP53 mutations in TKI-resistant tumors. The TP53 G245S mutant has been associated with a poor prognosis in colon cancer.34 This mutation can cause decreased levels of the 53BP1 protein and destabilize several structural regions of the protein that are crucial for DNA binding.35,36

Conclusions

Although the sample size was small and the patients came from a single hospital, this study suggests that a high mutation burden and mutations in DNA repair genes, including TP53, might be associated with primary resistance to crizotinib in ALK-rearranged NSCLC, leading to poor survival outcomes. Further prospective clinical studies are needed to confirm the role of TP53 or DNR repair-related gene variations in responses to ALK-TKI treatment or an immune-checkpoint inhibition strategy for ALK-rearranged NSCLC.

Data Sharing Statement

The raw data supporting the conclusions of this article will be made available by the authors without undue reservation.

Ethics and Consent Statement

The studies involving human participants were reviewed and approved by the Medical Ethics Committee in the First Affiliated Hospital of Guangzhou Medical University and carried out in accordance with the Declaration of Helsinki. The patients/participants provided their written informed consent to participate in this study.

Acknowledgments

This work was supported by the Natural Science Foundation of Guangdong Province (D. X. No. 2017A030313484) and Guangdong Municipal Health Science and Technology Plan Project (Q. D. No. 20201A010050), and sponsored by Shanghai Tongshu Biotechnology Co., Ltd. In addition, the authors wish to thank Hao Gong, Ph.D., for the analysis of NGS data.

Author Contributions

All authors contributed to data analysis, drafting or revising the article, have agreed on the journal to which the article will be submitted, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

Disclosure

The authors declare no conflict of interest.

References

1. Zhou JX, Yang H, Deng Q, et al. Oncogenic driver mutations in patients with non-small-cell lung cancer at various clinical stages. Ann Oncol. 2013;24(5):1319–1325. doi:10.1093/annonc/mds626

2. Auliac JB, Monnet I, Dubos-Arvis C, et al. Non-Small-Cell Lung Cancer (NSCLC) harboring ALK translocations: clinical characteristics and management in a real-life setting: a French retrospective analysis (GFPC 02-14 Study). Target Oncol. 2017;12(6):833–838. doi:10.1007/s11523-017-0520-7

3. Solomon BJ, Kim DW, Wu YL, et al. Final overall survival analysis from a study comparing first-line crizotinib versus chemotherapy in ALK-mutation-positive non-small-cell lung cancer. J Clin Oncol. 2018;36(22):2251–2258. doi:10.1200/JCO.2017.77.4794

4. Duruisseaux M, Besse B, Cadranel J, et al. Overall survival with crizotinib and next-generation ALK inhibitors in ALK-positive non-small-cell lung cancer (IFCT-1302 CLINALK): a French nationwide cohort retrospective study. Oncotarget. 2017;8(13):21903–21917. doi:10.18632/oncotarget.15746

5. Ito K, Hataji O, Kobayashi H, et al. Sequential therapy with crizotinib and alectinib in ALK-rearranged non-small cell lung cancer-a multicenter retrospective study. J Thorac Oncol. 2017;12(2):390–396. doi:10.1016/j.jtho.2016.07.022

6. Camidge DR, Dziadziuszko R, Peters S, et al. Updated efficacy and safety data and impact of the EML4-ALK fusion variant on the efficacy of alectinib in untreated ALK-positive advanced non-small cell lung cancer in the global Phase III ALEX study. J Thorac Oncol. 2019;14(7):1233–1243. doi:10.1016/j.jtho.2019.03.007

7. Shaw AT, Solomon BJ, Besse B, et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol. 2019;37(16):1370–1379. doi:10.1200/JCO.18.02236

8. Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers. Sci Transl Med. 2012;4(120):120ra117. doi:10.1126/scitranslmed.3003316

9. Katayama R. Drug resistance in anaplastic lymphoma kinase-rearranged lung cancer. Cancer Sci. 2018;109(3):572–580. doi:10.1111/cas.13504

10. Sullivan I, Planchard D. ALK inhibitors in non-small cell lung cancer: the latest evidence and developments. Ther Adv Med Oncol. 2016;8(1):32–47. doi:10.1177/1758834015617355

11. Okada K, Araki M, Sakashita T, et al. Prediction of ALK mutations mediating ALK-TKIs resistance and drug re-purposing to overcome the resistance. EBioMedicine. 2019;41:41105–41119.

12. DiBonaventura MD, Wong W, Shah-Manek B, Schulz M. Real-world usage and clinical outcomes of alectinib among post-crizotinib progression anaplastic lymphoma kinase positive non-small-cell lung cancer patients in the USA. Onco Targets Ther. 2018;11:1175–1182.

13. Cadranel J, Cortot AB, Lena H, et al. Real-life experience of ceritinib in crizotinib-pretreated ALK+ advanced non-small cell lung cancer patients. ERJ Open Res. 2018;4(1):00058–2017. doi:10.1183/23120541.00058-2017

14. Lin JJ, Zhu VW, Yoda S, et al. Impact of EML4-ALK variant on resistance mechanisms and clinical outcomes in ALK-positive lung cancer. J Clin Oncol. 2018;36(12):1199–1206. doi:10.1200/JCO.2017.76.2294

15. Yang H, Wang F, Deng Q, et al. Predictive and prognostic value of phosphorylated c-KIT and PDGFRA in advanced non-small cell lung cancer harboring ALK fusion. Oncol Lett. 2019;17(3):3071–3076.

16. Shahi RB, De Brakeleer S, Caljon B, et al. Identification of candidate cancer predisposing variants by performing whole-exome sequencing on index patients from BRCA1 and BRCA2-negative breast cancer families. BMC Cancer. 2019;19(1):313. doi:10.1186/s12885-019-5494-7

17. Rusch M, Nakitandwe J, Shurtleff S, et al. Clinical cancer genomic profiling by three-platform sequencing of whole genome, whole exome and transcriptome. Nat Commun. 2018;9(1):3962. doi:10.1038/s41467-018-06485-7

18. Feng L, Hong S, Gao J, Li J. Whole-exome sequencing characterized the landscape of somatic mutations and pathways in colorectal cancer liver metastasis. J Oncol. 2019;20192684075. doi:10.1155/2019/2684075

19. Beltran H, Eng K, Mosquera JM, et al. Whole-exome sequencing of metastatic cancer and biomarkers of treatment response. JAMA Oncol. 2015;1(4):466–474. doi:10.1001/jamaoncol.2015.1313

20. Suwinski P, Ong C, Ling MHT, et al. Advancing personalized medicine through the application of whole exome sequencing and big data analytics. Front Genet. 2019;10:49.

21. Tuna M, Amos CI. Genomic sequencing in cancer. Cancer Lett. 2013;340(2):161–170. doi:10.1016/j.canlet.2012.11.004

22. Bailey MH, Tokheim C, Porta-Pardo E, et al. Comprehensive characterization of cancer driver genes and mutations. Cell. 2018;174(4):1034–1035.

23. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34. doi:10.1186/s13073-017-0424-2

24. Valero C, Lee M, Hoen D, et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat Commun. 2021;12(1):729. doi:10.1038/s41467-021-20935-9

25. Chen M, Xu Y, Zhao J, et al. Concurrent driver gene mutations as negative predictive factors in epidermal growth factor receptor-positive non-small cell lung cancer. EBioMedicine. 2019;42:304–42310.

26. Jiao XD, He X, Qin BD, et al. The prognostic value of tumor mutation burden in EGFR-mutant advanced lung adenocarcinoma, an analysis based on cBioPortal data base. J Thorac Dis. 2019;11(11):4507–4515. doi:10.21037/jtd.2019.11.04

27. Xie LL, Shi F, Tan Z, et al. Mitochondrial network structure homeostasis and cell death. Cancer Sci. 2018;109(12):3686–3694. doi:10.1111/cas.13830

28. Kingnate C, Charoenkwan K, Kumfu S, Chattipakorn N, Chattipakorn SC. Possible roles of mitochondrial dynamics and the effects of pharmacological interventions in chemoresistant ovarian cancer. EBioMedicine. 2018;34:256–266.

29. Danish Q, Mokhdomi TA, Bukhari S, Ahmad R. The ensemble of genetic factors and angiogenic signals via VEGF receptors in lung cancer progression. Cancer Biomark. 2015;15(5):619–633. doi:10.3233/CBM-150502

30. Perez-Ramirez C, Canadas-Garre M, Alnatsha A, et al. Pharmacogenetics of platinum-based chemotherapy: impact of DNA repair and folate metabolism gene polymorphisms on prognosis of non-small cell lung cancer patients. Pharmacogenomics J. 2019;19(2):164–177. doi:10.1038/s41397-018-0014-8

31. Romano FJ, Rossetti S, Conteduca V, et al. Role of DNA repair machinery and p53 in the testicular germ cell cancer: a review. Oncotarget. 2016;7(51):85641–85649. doi:10.18632/oncotarget.13063

32. Mandal R, Samstein RM, Lee KW, et al. Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response. Science. 2019;364(6439):485–491. doi:10.1126/science.aau0447

33. Wang Z, Zhao J, Wang G, et al. Comutations in DNA damage response pathways serve as potential biomarkers for immune checkpoint blockade. Cancer Res. 2018;78(22):6486–6496. doi:10.1158/0008-5472.CAN-18-1814

34. Samowitz WS, Curtin K, Ma KN, et al. Prognostic significance of p53 mutations in colon cancer at the population level. Int J Cancer. 2002;99(4):597–602. doi:10.1002/ijc.10405

35. Lepre MG, Omar SI, Grasso G, et al. Insights into the effect of the G245S single point mutation on the structure of p53 and the binding of the protein to DNA. Molecules. 2017;22(8):1358. doi:10.3390/molecules22081358

36. Suchankova J, Legartova S, Ruckova E, et al. Mutations in the TP53 gene affected recruitment of 53BP1 protein to DNA lesions, but level of 53BP1 was stable after gamma-irradiation that depleted MDC1 protein in specific TP53 mutants. Histochem Cell Biol. 2017;148(3):239–255. doi:10.1007/s00418-017-1567-3

Creative Commons License © 2021 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.