Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer

Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of... Hindawi Journal of Oncology Volume 2020, Article ID 8811487, 7 pages https://doi.org/10.1155/2020/8811487 Research Article Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer Dongfang Chen, Jianlin Xu, Rong Qiao, Yizhuo Zhao, Tianqing Chu, Baohui Han , and Runbo Zhong Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China Correspondence should be addressed to Baohui Han; hanxkyy@aliyun.com and Runbo Zhong; tonic_chung@139.com Received 28 July 2020; Revised 2 November 2020; Accepted 6 November 2020; Published 19 November 2020 Academic Editor: Ozkan Kanat Copyright © 2020 Dongfang Chen et al. 1is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Some studies have revealed that specific genetic mutations could be associated with chemotherapy response or even survival in small-cell lung cancer (SCLC). Our retrospective study aimed to identify the correlation between genetic mutations and progression-free survival (PFS) in extensive-stage SCLC after first-line chemotherapy. A total of 75 patients with extensive- stage SCLC confirmed by histopathology from February 2018 to February 2019 were retrospectively analyzed. 1e biopsy specimens of all patients were analyzed by Next-Generation Sequencing (NGS). All patients received first-line chemotherapy and follow-up at Shanghai Chest Hospital. Eleven genes were mutated in, at least, 10% of the 75 patients, including TP53 (96%), RB1 (77%), SMAD4 (32%), NOTCH1 (21%), PTEN (16%), FGFR1 (16%), KDR (15%), PIK3CA (15%), ROS1 (15%), BRCA2 (13%), and ERBB4 (10%). 1e median number of mutated genes among all patients was 5. Patients with more than 5 mutated genes (PFS � 6.7 months, P � 0.004), mutant TP53 (PFS � 5.0 months, P � 0.011), and mutant BRCA2 (PFS � 6.7 months, P � 0.046) had better PFS after first-line chemotherapy than other patients. Multivariate Cox regression analysis showed that patients who achieved a PR (HR 3.729, 95% CI 2.038–6.822), had more than 5 mutated genes (HR 1.929, 95% CI 1.096–3.396), had BRCA2 mutations (HR 4.581, 95% CI 1.721–12.195), and had no liver metastasis (HR 0.415, 95% CI 0.181–0.951) showed improvements in PFS after first-line chemotherapy. In conclusion, the number of mutated genes and BRCA2 mutation status in extensive-stage SCLC were significantly related to PFS after first-line chemotherapy. chemotherapy is between 7 and 10 months; the one-year 1. Introduction overall survival (OS) rate is approximately 20% to 40% [1]. Small-cell lung cancer (SCLC) is a malignant neuroendo- Except for the ALTER 1202 and IMpower133 trials, there crine tumor with an epithelial source and accounts for have been a few advances in targeted therapy and immu- approximately 15% to 17% of all diagnosed lung cancers [1]. notherapy for SCLC over the past 30 years. 1e novel 1e unique biological characteristics of SCLC include a close multitarget tyrosine kinase inhibitor (TKI) anlotinb which association with smoking, rapid proliferation, and early targets vascular endothelial growth factor receptor type 2 hematogenous metastasis [2]. 1us, 80% of patients are at an and 3 (VEGF 2, 3), platelet-derived growth factor β extensive stage when first diagnosed. 1e liver, bone, kidney, (PDGFRβ), fibroblast growth factor receptor (FGFR), and and brain are several common distant metastasis sites. For stem cell-factor receptor (c-Kit) has been approved as a these extensive-stage patients, the current first-line standard third-line or beyond therapy for SCLC by the National chemotherapy is etoposide plus carboplatin or cisplatin [3]. Medical Products Administration (NMPA) based on the 1e median survival of patients with advanced SCLC with ALTER 1202 trial in 2019 [4]. In the IMpower133 trial, OS 2 Journal of Oncology following the atezolizumab-based combination was only two inclusion criteria were as follows: (1) patients who were months longer than that following chemotherapy alone [5]. diagnosed with SCLC histopathologically and at an extensive However, chemotherapy is still an important component of stage clinically; (2) patients who had, at least, one measurable standard first-line therapy for extensive-stage SCLC. tumor lesion; (3) patients whose biopsy specimens were 1e emergence of NGS has revolutionized the detection analyzed by NGS; (4) patients who were between 18 and 70 of genetic mutations. 1e advantages of NGS are that it years of age; (5) patients whose Eastern Cooperative On- allows high throughput analysis, has good sensitivity, and cology Group performance status was 0 or 1; and (6) patients provides abundant information [6]. With the application of who received etoposide plus carboplatin or cisplatin as first- NGS technology, many studies have revealed that genetic line chemotherapy until disease progression or intolerance mutation rates are extremely high in SCLC [7–9]. Several to chemotherapy. 1e key exclusion criteria were as follows: driver genes or signaling pathways may be simultaneously (1) patients who were diagnosed with limited-stage SCLC; activated during SCLC development, which promotes tumor (2) patients who had severe and/or uncontrolled diseases progression through different mechanisms. TP53 and RB1 before first-line chemotherapy; and (3) patients who suffered mutations exist in almost all SCLCs [10]. Other common from other malignant tumors simultaneously. mutated genes include MYC family members, FGFR1, SOX2, PIK3CA, PTEN, and NOTCH family members [11]. 2.2. Clinical Assessments. Patients were given etoposide Most patients with extensive-stage SCLC have relatively (100 mg/m ) plus carboplatin (AUC � 5) or cisplatin (75 mg/ good responses to first-line chemotherapy, but they will m ) at every course. Clinical follow-up exams included a eventually experience chemotherapy resistance and relapse. physical examination and laboratory tests, which were According to NCCN guidelines, SCLC patients who relapse performed at every course. Efficacy was evaluated every two beyond three months after first-line chemotherapy are courses by computed tomography according to Response categorized as chemosensitive, whereas those who relapse Evaluation Criteria in Solid Tumors (RECIST v1.1) and within three months of initial chemotherapy are considered included complete response (CR), partial response (PR), chemoresistant. Only a few studies have revealed that certain stable disease (SD), and progressive disease (PD). If the first- mutated genes are associated with chemotherapy sensitivity. line treatment failed, a second-line treatment was given. All Chinese scholars identified NDRG4 as a new tumor sup- patients were regularly followed up after receiving first-line pressor gene that plays a tumor suppressive role in SCLC. chemotherapy at Shanghai Chest Hospital. 1e follow-up SCLC patients with NDRG4 mutations are more sensitive to ended on April 10, 2019. chemotherapy and have a longer survival time than those without such mutations, which suggests that the NDRG4 protein could be used as a biomarker to predict the clinical 2.3. NGS. 1e llumina TruSeq Amplicon Cancer Panel kit prognosis of SCLC. Another study focused on cisplatin- (Burning Rock Company, China) and the MiSeq instrument resistant target genes and genes associated with poor were used for sequencing. 1e sequencing targeted the prognosis in SCLC, and the results indicated that DNAH10 mutation hotspots of 68 genes with high mutation rates in mutations were significantly associated with cisplatin re- lung cancer (Table 1). 1e main steps were as follows: (1) an sistance, poor OS, and worse PFS in SCLC [12]. 1erefore, NGS library was prepared by fragmenting a gDNA sample DNAH10 mutations may have potential value in predicting and ligating specialized adapters to both fragment ends; (2) cisplatin resistance and poor survival in SCLC. the library was loaded into a flow cell and the fragments 1e abovementioned studies suggest that specific genetic hybridized to the flow cell surface, and each bound fragment mutations may be associated with chemotherapy response or was amplified into a clonal cluster through bridge ampli- survival in SCLC. Since gene-related research on SCLC is fication; (3) sequencing reagents, including fluorescently limited, the relationship between genetic mutations and labeled nucleotides, were added, the first base was incor- survival in SCLC is worth further study. 1is study may be porated, the flow cell was imaged, the emission from each helpful for identifying SCLC patients who could benefit from cluster was recorded, the emission wavelength and intensity chemotherapy, which is of great practical significance. 1us, were used to identify the base, and this cycle was repeated the main purpose of our retrospective study is as follows: “n” times to create a read length of “n” bases; and (4) data first, to investigate the mutation status of extensive-stage analysis. SCLC patients, including nonsmoking and female patients, and second, to identify the correlation between genetic mutations and PFS in extensive-stage SCLC patients after 2.4. Statistical Analysis. Progression-free survival (PFS) was first-line chemotherapy. measured from the date of initiation of first-line chemo- therapy to the date of disease progression or the last follow- up visit. SPSS22.0 statistical software (IBM, Armonk, NY, 2. Materials and Methods USA) was used for data processing. Only genes that were 2.1. Patients. We identified and reviewed the clinical data of mutated in, at least, 10% of the enrolled patients were patients who were diagnosed with extensive-stage SCLC at considered for statistical analysis. 1e Mann–Whitney U test Shanghai Chest Hospital from February 2018 to February was used to identify differences in the number of mutated 2019. 1e study protocol was approved by the Ethics genes between groups. 1e Kaplan–Meier method and log- Committee of Shanghai Chest Hospital (KS1934). 1e key rank test were used to identify the correlation between Journal of Oncology 3 Table 1: 1e 68 genes detected by NGS. ALK BRAF EGFR ERBB2 KRAS MET RET ROS1 AKT1 APC ARAF ATM AXL BCL2L11 BRCA1 BRCA2 CCND1 CD74 CDK4 CDK6 CDKN2A CTNNB1 DDR2 ERBB3 ERBB4 ESR1 FGF19 FGF3 FGF4 FGFR1 FGFR2 FGFR3 FLT3 HRAS IDH1 IDH2 IGF1R JAK1 JAK2 KDR KIT MAP2K1 MTOR MYC NF1 NOTCH1 NRAS NRG1 NTRK1 NTRK2 NTRK3 PDGFRA PIK3CA PTCH1 PTEN RAF1 RB1 SMAD4 SMO STK11 TOP2A TP53 TSC1 TSC2 AR CYP2D6 DPYD UGT1A1 genetic mutations and PFS. Multivariable Cox regression Among the mutated genes listed in Table 2, patients with mutant TP53 had a better PFS than those with wild-type was used to identify significant factors related to PFS. All tests were two sided, and P values <0.05 were considered TP53 (5.0 versus 3.4 months, P � 0.011). Similarly, patients statistically significant. with BRCA2 mutations had a better PFS than patients with wild-type BRCA2 (6.7 versus 4.5 months, P � 0.046) 3. Results (Table 5). Given the high frequencies of TP53 and RB1 mutations, 3.1. Patient Characteristics. 1e study sample consisted of 75 the correlation between mutation abundance and PFS was extensive-stage SCLC patients, 65 (87%) of which were further analyzed. Patients with mutant TP53 were divided smokers and 10 (13%) of which were never-smokers. After into two groups according to the mutation abundance first-line chemotherapy, 44 (59%) patients achieved a PR, 23 (≤80% and >80%, median: 80%), and there was no signif- (31%) patients had SD, and the remaining 8 (10%) patients icant difference in PFS between the two groups (P � 0.803). had PD. 1e proportions of patients with bone metastasis, Additionally, there was no significant difference in PFS brain metastasis, and liver metastasis were 24%, 13%, and related to the abundance of mutant RB1 (≤77% and >77%, 15%, respectively. Demographic data for all patients are median: 77%; P � 0.372). shown in Table 2. Multivariate Cox regression analysis showed that the response to first-line chemotherapy, the number of mutated genes, BRCA2 mutation status, and liver metastasis had 3.2. Genetic Mutations. Eleven genes were mutated in, at significant effects on PFS after first-line chemotherapy least, 10% of the 75 patients. 1e top two common mutated (Table 6). Patients who achieved a PR (HR 3.729, 95% CI genes were TP53 (96%) and RB1 (77%). SMAD4 and 2.038–6.822), had more than 5 mutated genes (HR 1.929, NOTCH1 were mutated in 32% and 21% of the 75 patients, 95% CI 1.096–3.396), had BRCA2 mutations (HR 4.581, 95% respectively. 1e frequencies of mutated genes related to the CI 1.721–12.195), and had no liver metastasis (HR 0.415, PIK3-AKT-mTOR signaling pathway (PTEN, FGFR1, and 95% CI 0.181–0.951) showed improvements in PFS. PIK3CA) were similar. Other mutated genes are shown in Table 3. 1e most frequent mutated genes in nonsmoking pa- 4. Discussion tients were TP53 (100%), RB1 (73%), SMAD4 (35%), KDR (23%), NOTCH1 (20%), and PTEN (20%). 1e most fre- 1ere are only few studies that have investigated the asso- quently mutated genes in female patients were TP53 (100%), ciation of genetic mutations with clinical prognosis in ex- RB1 (78%), SMAD4 (56%), NOTCH1 (44%), KDR (44%), tensive-stage SCLC. 1erefore, we collected data from 75 NTRK1 (33%), and RET (22%). extensive-stage SCLC patients for further study. Our study 1e number of mutated genes among all patients ranged revealed the mutation status of extensive-stage SCLC pa- from 2 to 15, and the median was 5. 1ere were no sig- tients, especially nonsmoking and female patients. In ad- nificant differences between the gender, age, smoking status, dition, we also found that the genetic mutations of extensive- and anatomy type (central type/peripheral type) subgroups stage SCLC were related to PFS after first-line (Table 4). chemotherapy. In our study, the genes that mutated in more than 20% of the total patients were TP53, RB1, SMAD4, and NOTCH1. In 3.3. PFS. 1e median PFS of the 75 patients was 4.7 months previous reports, TP53 and RB1 mutations were shown to (95% CI 3.8–5.5). 1e median PFSs of male and female affect up to 90% and up to 65% of SCLC patients, respec- patients were 4.6 months (95% CI 3.6–5.6) and 6.0 months tively [13]. 1e importance of these two mutated genes in (95% CI 4.0–8.0), respectively (P � 0.777). 1e median PFSs SCLC tumorigenesis has been highlighted by numerous of never-smokers and smokers were 5.2 months (95% CI functional studies [14]. 1e frequency of SMAD4 mutation 3.9–6.5) and 4.6 months (95% CI 3.1–6.1), respectively was surprisingly high in our study. SMAD4 mediates the (P � 0.285). 1e relationship between genetic mutations and signaling of transforming growth factor beta and bone PFS was investigated. Surprisingly, patients with more than 5 morphogenic protein ligands and is a well-defined tumor mutated genes had a better PFS than patients with less than 5 suppressor in pancreatic and colon cancer [15]. SMAD4 mutated genes (6.7 versus 3.6 months, P � 0.004) (Figure 1). 4 Journal of Oncology Table 2: Characteristics of the 75 extensive-stage SCLC patients. Characteristic N (%) Gender Male/female 66 (88)/9 (12) Age <65/≥65 39 (52)/36 (48) Smoking status Never-smoker/smoker 10 (13)/65 (87) Anatomy type Central type/peripheral type 55 (74)/20 (26) Response to first-line chemotherapy PR/SD/PD 44 (59)/23 (31)/8 (10) Metastatic sites Bone 18 (24) Brain 10 (13) Liver 11 (15) Lung (contralateral) 28 (37) Pleura 20 (27) 0.00 2.00 4.00 6.00 8.00 10.00 12.00 Pericardium 3 (4) Months Kidney 3 (4) Less than 5 (n = 41) median Censored SCLC, small-cell lung cancer; PR, partial response; SD, stable disease; PD, PFS = 3.6 months progressive disease. More than 5 (n = 34) median Censored PFS = 6.7 months Table 3: Frequency of genes mutated in, at least, 10% of the 75 P = 0.004 patients. Figure 1: Kaplan–Meier curves of progression-free survival based Gene Mutant N (%) on the number of mutated genes. TP53 72 (96) RB1 58 (77) SMAD4 24 (32) mutation status of this special population. We found that NOTCH1 16 (21) TP53, RB1, SMAD4, KDR, and NOTCH1 were mutated PTEN 12 (16) frequently in both nonsmoking and female patients. 1is FGFR1 12 (16) could be explained by the fact that most of the female pa- KDR 11 (15) tients were never-smokers. In addition, nonsmoking pa- PIK3CA 11 (15) tients had a high prevalence of PTEN mutations, while ROS1 11 (15) female patients had a high prevalence of NTRK1 and RET BRCA2 10 (13) mutations. Cardona et al. reported that, among 10 never-/ ERBB4 8 (10) ever-smokers, the most frequent genetic mutations detected by NGS were TP53 (80%), RB1 (40%), CYLD (30%), EGFR Table 4: Number of mutated genes in the different subgroups. (30%), MET (20%), SMAD4 (20%), and BRIP1 (20%) [17]. In Subgroup N (median, range) P value a study by Sun et al., among 28 genetically evaluable never- smokers, the most common mutations included TP53 Gender (93%), RB1 (25%), PTEN (18%), EGFR (14%), MET (14%), Male/female 5 (2–15)/5 (3–12) 0.216 Age and SMAD4 (11%) [18]. Our data and previous findings are <65/≥65 5 (2–13)/6 (2–15) 0.209 not entirely consistent with each other, since differences Smoking status exist in terms of sample size, sequencing panel, and tumor Never-smoker/smoker 5 (3–12)/5 (2–15) 0.857 stage. Anatomy type Smoking status and gender were not shown to be related Central type/peripheral type 5 (2–15)/5 (3–13) 0.884 to PFS in our study. In an analysis of 20 SCLC patients, an improvement in survival in terms of PFS in response to first- mutations are associated with lymph node metastases, in- line treatment according to smoking status was not ob- creased angiogenesis, and more aggressive cellular behavior served, but never-smokers achieved an improvement in OS in vitro [15]. NOTCH signaling is critical for the regulation compared to smokers [17]. In another study, in 394 ex- of neuroendocrine differentiation. A previous study iden- tensive-stage SCLC patients who received first-line che- tified the presence of mutations in NOTCH family members motherapy, both PFS and OS were correlated with smoking in a quarter of analyzed SCLC samples [16], which was history [19]. Overall, according to previous studies, smoking confirmed in our study. seems to be a negative factor for survival outcomes, espe- SCLC is strongly correlated with a history of smoking cially shorter OS. While gender-related differences in PFS and mainly occurs in males, but a small portion of patients were not seen according to our data, gender may play an are nonsmoking or female. It is meaningful to explore the important role in the prognosis of SCLC. Dowlati et al. Progression-free survival rate (%) Journal of Oncology 5 Table 5: Effect of mutation status on PFS after first-line male patients [21]. Another study indicated that female sex is chemotherapy. useful as a predictor for better long-term survival [22]. Tumor mutational burden (TMB), a quantification of Gene Median PFS in months (95% CI) P value tumoral mutations, has been associated with the response TP53 0.011 to immunotherapy. In the CheckMate 032 trial, 401 pa- WT 3.4 (1.2–5.6) tients received treatment with nivolumab or a combina- Mutant 5.0 (4.1–5.9) tion of nivolumab and ipilimumab [23]. Among patients RB1 0.576 treated with combination therapy, a high TMB was related WT 6.0 (2.9–9.1) Mutant 4.7 (4.0–5.4) to better ORR and OS. In the CheckMate 227 trial, first- SMAD4 0.077 line nivolumab plus ipilimumab significantly prolonged WT 4.1 (3.1–5.1) PFS versus chemotherapy in advanced NSCLC patients Mutant 6.3 (4.9–7.7) with a high TMB (≥10 mutations/megabase) [24]. 1us, NOTCH1 0.191 TMB may be a prognostic factor for lung cancer immu- WT 4.6 (3.4–5.8) notherapy. In contrast to previous studies, blood-based Mutant 6.0 (3.1–8.9) TMB showed no value in predicting benefit with atezo- PTEN 0.666 lizumab in the IMpower133 trial, and the possible ex- WT 4.7 (3.8–5.6) planation was that the combination of platinum and Mutant 5.0 (3.0–7.0) etoposide was active and myelosuppressive [5]. 1e FGFR1 0.472 WT 4.7 (4.0–5.4) KEYNOTE 604 trial showed that pembrolizumab com- Mutant 5.7 (3.2–8.2) bined with standard first-line EP significantly improved KDR 0.522 PFS in extensive-stage SCLC patients, but TMB was not WT 4.6 (3.7–5.5) further analyzed [25]. Notably, our study showed that an Mutant 5.4 (3.3–7.5) improvement in PFS was observed in patients who had PIK3CA 0.613 more than 5 mutated genes. We may further make an WT 4.6 (3.7–5.5) assessment of TMB to verify this conclusion. Additionally, Mutant 6.0 (3.0–9.0) more basic medical research is needed to reveal the ROS1 0.608 specific mechanism of our finding. WT 4.7 (3.8–5.6) In our retrospective study, among 11 common mutated Mutant 5.7 (2.8–8.6) genes, mutant TP53 and BRCA2 were associated with better BRCA2 0.046 WT 4.5 (3.6–5.4) PFS. Using multivariate analysis, only BRCA2 was significant Mutant 6.7 (5.0–8.4) in predicting PFS. Dowlati et al. reported that patients with ERBB4 0.660 TP53 mutations had similar PFS and OS as patients with WT 4.6 (3.9–5.3) wild-type TP53 [20]. However, only 3 (4%) patients had Mutant 6.0 (4.5–7.5) wild-type TP53, and this result should be confirmed in a PFS, progression-free survival; WT, wild type; CI, confidence interval. larger sample. Dowlati et al. also reported that patients with mutant RB1 had both better OS and PFS than patients with wild-type RB1, but this was not validated in the multivariate Table 6: Multivariate Cox regression analysis of PFS after first-line analysis. BRCA2 helps repair damaged DNA and plays a chemotherapy. crucial role in ensuring the stability of genome. Patients with Factor P value HR (95% CI) BRCA2 mutations may develop genetic alterations leading to Age 0.201 cancer. Specific inherited mutations in BRCA2 may notably Bone metastasis 0.353 increase the risk of female breast and ovarian cancers, but TP53 0.068 they have also been associated with increased risks of several SMAD4 0.412 additional types of cancer. 1ere is little research indicating a NOTCH1 0.073 relationship between mutant BRCA2 and SCLC, but our Response to first-line <0.0001 3.729 (2.038–6.822) finding suggests that there might be interactions between chemotherapy mutant BRCA2 and SCLC. Number of mutated genes 0.023 1.929 (1.096–3.396) 4.581 Moreover, other similar studies are worth referencing. A BRCA2 0.002 (1.721–12.195) group of researchers found that CREBBP/EP300, TP73, or Liver metastasis 0.038 0.415 (0.181–0.951) NOTCH mutations had no influence on the survival of SCLC PFS, progression-free survival; HR, hazard ratio;CI, confidence interval. patients treated with surgery and chemotherapy [14]. A chromogenic in situ hybridization study showed that MYC amplification was associated with poor survival in SCLC and found that female sex was a positive factor for response to might be an independent prognostic factor for SCLC [26]. chemotherapy in extensive-stage SCLC patients [20]. A Our study has some limitations. First, the sample size is pooled analysis of randomized SCLC chemotherapy trials small. Second, the OS data are not shown because of the high showed that female patients survived modestly longer than loss ratio of follow-up. 6 Journal of Oncology lung cancers defined by whole exome/RNA sequencing,” 5. Conclusions Carcinogenesis, vol. 36, pp. 616–621, 2015. [8] E. Arriola, I. Canadas, M. Arumi et al., “Genetic changes in In our retrospective study, the number of mutated genes and small cell lung carcinoma,” Clinical and Translational On- BRCA2 mutation status in extensive-stage SCLC were sig- cology, vol. 10, pp. 189–197, 2008. nificantly related to PFS after first-line chemotherapy. [9] B. Tong, J. Zhao, and M. Wang, “Advances on driver mu- tations of small cell lung cancer,” Zhongguo Fei Ai Za Zhi, Data Availability vol. 19, pp. 236–240, 2016, in Chinese. [10] L. Meder, K. Konig, L. Ozretic et al., “NOTCH, ASCL1, p53 1e data used to support the findings of this study are and RB alterations define an alternative pathway driving available from the first author and the corresponding author neuroendocrine and small cell lung carcinomas,” Interna- upon request. tional Journal of Cancer, vol. 138, pp. 927–938, 2016. [11] V. Foy, M. W. Schenk, K. Baker et al., “Targeting DNA damage in SCLC,” Lung Cancer, vol. 114, pp. 12–22, 2017. Disclosure [12] M. Li, A. Lin, P. Luo et al., “DNAH10 mutation correlates with cisplatin sensitivity and tumor mutation burden in small-cell Jianlin Xu is the co-first author of this article. lung cancer,” Aging (Albany NY), vol. 12, pp. 1285–1303, 2020. [13] M. Peifer, L. Fernandez-Cuesta, M. L. Sos et al., “Integrative Conflicts of Interest genome analyses identify key somatic driver mutations of small-cell lung cancer,” Nature Genetics, vol. 44, pp. 1104– 1e authors declare that there are no conflicts of interest 1110, 2012. regarding the publication of this paper. [14] J. George, J. S. Lim, S. J. Jang et al., “Comprehensive genomic profiles of small cell lung cancer,” Nature, vol. 524, pp. 47–53, Authors’ Contributions [15] S. M. Haeger, J. J. 1ompson, S. Kalra et al., “Smad4 loss (i) Dongfang Chen and Runbo Zhong were involved in promotes lung cancer formation but increases sensitivity to conception and design. (ii) Administrative support was DNA topoisomerase inhibitors,” Oncogene, vol. 35, pp. 577– provided by Baohui Han. (iii) Jianlin Xu was responsible for 586, 2016. [16] E. A. Semenova, R. Nagel, and A. Berns, “Origins, genetic provision of study materials or patients. (iv) Collection and landscape, and emerging therapies of small cell lung cancer,” assembly of data were performed by Yizhuo Zhao. (v) Data Genes & Development, vol. 29, pp. 1447–1462, 2015. analysis and interpretation were conducted by Rong Qiao [17] A. F. Cardona, L. Rojas, Z. L. Zatarain-Barron et al., “Mul- and Tianqing Chu. (vi) All authors were involved in man- tigene mutation profiling and clinical characteristics of small- uscript writing. (vii) All authors approved the final version cell lung cancer in never-smokers vs. heavy smokers of the manuscript. (Geno1.3-CLICaP),” Frontiers in Oncology, vol. 9, p. 254, Acknowledgments [18] J. M. Sun, Y. L. Choi, J. H. Ji et al., “Small-cell lung cancer detection in never-smokers: clinical characteristics and 1e authors would like to thank all of the investigators for multigene mutation profiling using targeted next-generation their involvement in this study. 1is work was supported by sequencing,” Annals of Oncology, vol. 26, pp. 161–166, 2015. the Natural Science Foundation of Shanghai (19ZR1449700). [19] M. Ma, M. Wang, Y. Xu et al., “First-line chemotherapy and its survival analysis of 394 patients with extensive-stage small cell lung cancer in a single institute,” Zhongguo Fei Ai Za Zhi, References vol. 17, pp. 8–14, 2014, in Chinese. [20] A. Dowlati, M. B. Lipka, K. McColl et al., “Clinical correlation [1] K. Kahnert, D. Kauffmann-Guerrero, and R. M. Huber, of extensive-stage small-cell lung cancer genomics,” Annals of “SCLC-state of the art and what does the future have in store?” Oncology, vol. 27, pp. 642–647, 2016. Clinical Lung Cancer, vol. 17, pp. 325–333, 2016. [21] P. Wheatley-Price, C. Ma, L. F. Ashcroft et al., “1e strength of [2] J. P. van Meerbeeck, D. A. Fennell, and D. K. De Ruysscher, female sex as a prognostic factor in small-cell lung cancer: a “Small-cell lung cancer,” Lancet, vol. 378, pp. 1741–1755, 2011. pooled analysis of chemotherapy trials from the Manchester [3] J. W. Neal, M. A. Gubens, and H. A. Wakelee, “Current lung group and medical research council clinical trials unit,” management of small cell lung cancer,” Clinics in Chest Annals of Oncology, vol. 21, pp. 232–237, 2010. Medicine, vol. 32, pp. 853–863, 2011. [22] N. Karachaliou, A. E. Sosa, and R. Rosell, “Unraveling the [4] S. Yang, Z. Zhang, and Q. Wang, “Emerging therapies for genomic complexity of small cell lung cancer,” Translational small cell lung cancer,” Journal of Hematology & Oncology, Lung Cancer Research, vol. 5, pp. 363–366, 2016. vol. 12, p. 47, 2019. [23] S. J. Antonia, J. A. Lopez-Martin, J. Bendell et al., “Nivolumab [5] L. Horn, A. S. Mansfield, A. Szczesna et al., “First-line ate- alone and nivolumab plus ipilimumab in recurrent small-cell zolizumab plus chemotherapy in extensive-stage small-cell lung cancer,” New England Journal of Medicine, vol. 379, lung cancer (Checkmate 032): a multicentre, open-label, phase 1/2 trial,” =e Lancet Oncology, vol. 17, pp. 883–895, pp. 2220–2229, 2018. [6] M. Le Gallo, F. Lozy, and D. W. Bell, “Next-generation se- 2016. [24] M. Reck, M. Schenker, K. H. Lee et al., “Nivolumab plus quencing,” Advances in Experimental Medicine and Biology, vol. 943, pp. 119–148, 2017. ipilimumab versus chemotherapy as first-line treatment in advanced non-small-cell lung cancer with high tumour [7] R. Iwakawa, T. Kohno, Y. Totoki et al., “Expression and clinical significance of genes frequently mutated in small cell mutational burden: patient-reported outcomes results from Journal of Oncology 7 the randomised, open-label, phase III Checkmate 227 trial,” European Journal of Cancer, vol. 116, pp. 137–147, 2019. [25] C. M. Rudin, M. M. Awad, A. Navarro et al., “Pembrolizumab or placebo plus etoposide and platinum as first-line therapy for extensive-stage small-cell lung cancer: randomized, double-blind, phase III Keynote-604 study,” Journal of Clinical Oncology, vol. 38, pp. 2369–2379, 2020. [26] C. Alves Rde, R. T. Meurer, and A. V. Roehe, “MYC am- plification is associated with poor survival in small cell lung cancer: a chromogenic in situ hybridization study,” Journal of Cancer Research and Clinical Oncology, vol. 140, pp. 2021– 2025, 2014. http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Journal of Oncology Hindawi Publishing Corporation

Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer

Loading next page...
 
/lp/hindawi-publishing-corporation/detection-of-genetic-mutations-by-next-generation-sequencing-for-ppC1E6HzAk

References (27)

Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2020 Dongfang Chen et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
ISSN
1687-8450
eISSN
1687-8469
DOI
10.1155/2020/8811487
Publisher site
See Article on Publisher Site

Abstract

Hindawi Journal of Oncology Volume 2020, Article ID 8811487, 7 pages https://doi.org/10.1155/2020/8811487 Research Article Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer Dongfang Chen, Jianlin Xu, Rong Qiao, Yizhuo Zhao, Tianqing Chu, Baohui Han , and Runbo Zhong Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China Correspondence should be addressed to Baohui Han; hanxkyy@aliyun.com and Runbo Zhong; tonic_chung@139.com Received 28 July 2020; Revised 2 November 2020; Accepted 6 November 2020; Published 19 November 2020 Academic Editor: Ozkan Kanat Copyright © 2020 Dongfang Chen et al. 1is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Some studies have revealed that specific genetic mutations could be associated with chemotherapy response or even survival in small-cell lung cancer (SCLC). Our retrospective study aimed to identify the correlation between genetic mutations and progression-free survival (PFS) in extensive-stage SCLC after first-line chemotherapy. A total of 75 patients with extensive- stage SCLC confirmed by histopathology from February 2018 to February 2019 were retrospectively analyzed. 1e biopsy specimens of all patients were analyzed by Next-Generation Sequencing (NGS). All patients received first-line chemotherapy and follow-up at Shanghai Chest Hospital. Eleven genes were mutated in, at least, 10% of the 75 patients, including TP53 (96%), RB1 (77%), SMAD4 (32%), NOTCH1 (21%), PTEN (16%), FGFR1 (16%), KDR (15%), PIK3CA (15%), ROS1 (15%), BRCA2 (13%), and ERBB4 (10%). 1e median number of mutated genes among all patients was 5. Patients with more than 5 mutated genes (PFS � 6.7 months, P � 0.004), mutant TP53 (PFS � 5.0 months, P � 0.011), and mutant BRCA2 (PFS � 6.7 months, P � 0.046) had better PFS after first-line chemotherapy than other patients. Multivariate Cox regression analysis showed that patients who achieved a PR (HR 3.729, 95% CI 2.038–6.822), had more than 5 mutated genes (HR 1.929, 95% CI 1.096–3.396), had BRCA2 mutations (HR 4.581, 95% CI 1.721–12.195), and had no liver metastasis (HR 0.415, 95% CI 0.181–0.951) showed improvements in PFS after first-line chemotherapy. In conclusion, the number of mutated genes and BRCA2 mutation status in extensive-stage SCLC were significantly related to PFS after first-line chemotherapy. chemotherapy is between 7 and 10 months; the one-year 1. Introduction overall survival (OS) rate is approximately 20% to 40% [1]. Small-cell lung cancer (SCLC) is a malignant neuroendo- Except for the ALTER 1202 and IMpower133 trials, there crine tumor with an epithelial source and accounts for have been a few advances in targeted therapy and immu- approximately 15% to 17% of all diagnosed lung cancers [1]. notherapy for SCLC over the past 30 years. 1e novel 1e unique biological characteristics of SCLC include a close multitarget tyrosine kinase inhibitor (TKI) anlotinb which association with smoking, rapid proliferation, and early targets vascular endothelial growth factor receptor type 2 hematogenous metastasis [2]. 1us, 80% of patients are at an and 3 (VEGF 2, 3), platelet-derived growth factor β extensive stage when first diagnosed. 1e liver, bone, kidney, (PDGFRβ), fibroblast growth factor receptor (FGFR), and and brain are several common distant metastasis sites. For stem cell-factor receptor (c-Kit) has been approved as a these extensive-stage patients, the current first-line standard third-line or beyond therapy for SCLC by the National chemotherapy is etoposide plus carboplatin or cisplatin [3]. Medical Products Administration (NMPA) based on the 1e median survival of patients with advanced SCLC with ALTER 1202 trial in 2019 [4]. In the IMpower133 trial, OS 2 Journal of Oncology following the atezolizumab-based combination was only two inclusion criteria were as follows: (1) patients who were months longer than that following chemotherapy alone [5]. diagnosed with SCLC histopathologically and at an extensive However, chemotherapy is still an important component of stage clinically; (2) patients who had, at least, one measurable standard first-line therapy for extensive-stage SCLC. tumor lesion; (3) patients whose biopsy specimens were 1e emergence of NGS has revolutionized the detection analyzed by NGS; (4) patients who were between 18 and 70 of genetic mutations. 1e advantages of NGS are that it years of age; (5) patients whose Eastern Cooperative On- allows high throughput analysis, has good sensitivity, and cology Group performance status was 0 or 1; and (6) patients provides abundant information [6]. With the application of who received etoposide plus carboplatin or cisplatin as first- NGS technology, many studies have revealed that genetic line chemotherapy until disease progression or intolerance mutation rates are extremely high in SCLC [7–9]. Several to chemotherapy. 1e key exclusion criteria were as follows: driver genes or signaling pathways may be simultaneously (1) patients who were diagnosed with limited-stage SCLC; activated during SCLC development, which promotes tumor (2) patients who had severe and/or uncontrolled diseases progression through different mechanisms. TP53 and RB1 before first-line chemotherapy; and (3) patients who suffered mutations exist in almost all SCLCs [10]. Other common from other malignant tumors simultaneously. mutated genes include MYC family members, FGFR1, SOX2, PIK3CA, PTEN, and NOTCH family members [11]. 2.2. Clinical Assessments. Patients were given etoposide Most patients with extensive-stage SCLC have relatively (100 mg/m ) plus carboplatin (AUC � 5) or cisplatin (75 mg/ good responses to first-line chemotherapy, but they will m ) at every course. Clinical follow-up exams included a eventually experience chemotherapy resistance and relapse. physical examination and laboratory tests, which were According to NCCN guidelines, SCLC patients who relapse performed at every course. Efficacy was evaluated every two beyond three months after first-line chemotherapy are courses by computed tomography according to Response categorized as chemosensitive, whereas those who relapse Evaluation Criteria in Solid Tumors (RECIST v1.1) and within three months of initial chemotherapy are considered included complete response (CR), partial response (PR), chemoresistant. Only a few studies have revealed that certain stable disease (SD), and progressive disease (PD). If the first- mutated genes are associated with chemotherapy sensitivity. line treatment failed, a second-line treatment was given. All Chinese scholars identified NDRG4 as a new tumor sup- patients were regularly followed up after receiving first-line pressor gene that plays a tumor suppressive role in SCLC. chemotherapy at Shanghai Chest Hospital. 1e follow-up SCLC patients with NDRG4 mutations are more sensitive to ended on April 10, 2019. chemotherapy and have a longer survival time than those without such mutations, which suggests that the NDRG4 protein could be used as a biomarker to predict the clinical 2.3. NGS. 1e llumina TruSeq Amplicon Cancer Panel kit prognosis of SCLC. Another study focused on cisplatin- (Burning Rock Company, China) and the MiSeq instrument resistant target genes and genes associated with poor were used for sequencing. 1e sequencing targeted the prognosis in SCLC, and the results indicated that DNAH10 mutation hotspots of 68 genes with high mutation rates in mutations were significantly associated with cisplatin re- lung cancer (Table 1). 1e main steps were as follows: (1) an sistance, poor OS, and worse PFS in SCLC [12]. 1erefore, NGS library was prepared by fragmenting a gDNA sample DNAH10 mutations may have potential value in predicting and ligating specialized adapters to both fragment ends; (2) cisplatin resistance and poor survival in SCLC. the library was loaded into a flow cell and the fragments 1e abovementioned studies suggest that specific genetic hybridized to the flow cell surface, and each bound fragment mutations may be associated with chemotherapy response or was amplified into a clonal cluster through bridge ampli- survival in SCLC. Since gene-related research on SCLC is fication; (3) sequencing reagents, including fluorescently limited, the relationship between genetic mutations and labeled nucleotides, were added, the first base was incor- survival in SCLC is worth further study. 1is study may be porated, the flow cell was imaged, the emission from each helpful for identifying SCLC patients who could benefit from cluster was recorded, the emission wavelength and intensity chemotherapy, which is of great practical significance. 1us, were used to identify the base, and this cycle was repeated the main purpose of our retrospective study is as follows: “n” times to create a read length of “n” bases; and (4) data first, to investigate the mutation status of extensive-stage analysis. SCLC patients, including nonsmoking and female patients, and second, to identify the correlation between genetic mutations and PFS in extensive-stage SCLC patients after 2.4. Statistical Analysis. Progression-free survival (PFS) was first-line chemotherapy. measured from the date of initiation of first-line chemo- therapy to the date of disease progression or the last follow- up visit. SPSS22.0 statistical software (IBM, Armonk, NY, 2. Materials and Methods USA) was used for data processing. Only genes that were 2.1. Patients. We identified and reviewed the clinical data of mutated in, at least, 10% of the enrolled patients were patients who were diagnosed with extensive-stage SCLC at considered for statistical analysis. 1e Mann–Whitney U test Shanghai Chest Hospital from February 2018 to February was used to identify differences in the number of mutated 2019. 1e study protocol was approved by the Ethics genes between groups. 1e Kaplan–Meier method and log- Committee of Shanghai Chest Hospital (KS1934). 1e key rank test were used to identify the correlation between Journal of Oncology 3 Table 1: 1e 68 genes detected by NGS. ALK BRAF EGFR ERBB2 KRAS MET RET ROS1 AKT1 APC ARAF ATM AXL BCL2L11 BRCA1 BRCA2 CCND1 CD74 CDK4 CDK6 CDKN2A CTNNB1 DDR2 ERBB3 ERBB4 ESR1 FGF19 FGF3 FGF4 FGFR1 FGFR2 FGFR3 FLT3 HRAS IDH1 IDH2 IGF1R JAK1 JAK2 KDR KIT MAP2K1 MTOR MYC NF1 NOTCH1 NRAS NRG1 NTRK1 NTRK2 NTRK3 PDGFRA PIK3CA PTCH1 PTEN RAF1 RB1 SMAD4 SMO STK11 TOP2A TP53 TSC1 TSC2 AR CYP2D6 DPYD UGT1A1 genetic mutations and PFS. Multivariable Cox regression Among the mutated genes listed in Table 2, patients with mutant TP53 had a better PFS than those with wild-type was used to identify significant factors related to PFS. All tests were two sided, and P values <0.05 were considered TP53 (5.0 versus 3.4 months, P � 0.011). Similarly, patients statistically significant. with BRCA2 mutations had a better PFS than patients with wild-type BRCA2 (6.7 versus 4.5 months, P � 0.046) 3. Results (Table 5). Given the high frequencies of TP53 and RB1 mutations, 3.1. Patient Characteristics. 1e study sample consisted of 75 the correlation between mutation abundance and PFS was extensive-stage SCLC patients, 65 (87%) of which were further analyzed. Patients with mutant TP53 were divided smokers and 10 (13%) of which were never-smokers. After into two groups according to the mutation abundance first-line chemotherapy, 44 (59%) patients achieved a PR, 23 (≤80% and >80%, median: 80%), and there was no signif- (31%) patients had SD, and the remaining 8 (10%) patients icant difference in PFS between the two groups (P � 0.803). had PD. 1e proportions of patients with bone metastasis, Additionally, there was no significant difference in PFS brain metastasis, and liver metastasis were 24%, 13%, and related to the abundance of mutant RB1 (≤77% and >77%, 15%, respectively. Demographic data for all patients are median: 77%; P � 0.372). shown in Table 2. Multivariate Cox regression analysis showed that the response to first-line chemotherapy, the number of mutated genes, BRCA2 mutation status, and liver metastasis had 3.2. Genetic Mutations. Eleven genes were mutated in, at significant effects on PFS after first-line chemotherapy least, 10% of the 75 patients. 1e top two common mutated (Table 6). Patients who achieved a PR (HR 3.729, 95% CI genes were TP53 (96%) and RB1 (77%). SMAD4 and 2.038–6.822), had more than 5 mutated genes (HR 1.929, NOTCH1 were mutated in 32% and 21% of the 75 patients, 95% CI 1.096–3.396), had BRCA2 mutations (HR 4.581, 95% respectively. 1e frequencies of mutated genes related to the CI 1.721–12.195), and had no liver metastasis (HR 0.415, PIK3-AKT-mTOR signaling pathway (PTEN, FGFR1, and 95% CI 0.181–0.951) showed improvements in PFS. PIK3CA) were similar. Other mutated genes are shown in Table 3. 1e most frequent mutated genes in nonsmoking pa- 4. Discussion tients were TP53 (100%), RB1 (73%), SMAD4 (35%), KDR (23%), NOTCH1 (20%), and PTEN (20%). 1e most fre- 1ere are only few studies that have investigated the asso- quently mutated genes in female patients were TP53 (100%), ciation of genetic mutations with clinical prognosis in ex- RB1 (78%), SMAD4 (56%), NOTCH1 (44%), KDR (44%), tensive-stage SCLC. 1erefore, we collected data from 75 NTRK1 (33%), and RET (22%). extensive-stage SCLC patients for further study. Our study 1e number of mutated genes among all patients ranged revealed the mutation status of extensive-stage SCLC pa- from 2 to 15, and the median was 5. 1ere were no sig- tients, especially nonsmoking and female patients. In ad- nificant differences between the gender, age, smoking status, dition, we also found that the genetic mutations of extensive- and anatomy type (central type/peripheral type) subgroups stage SCLC were related to PFS after first-line (Table 4). chemotherapy. In our study, the genes that mutated in more than 20% of the total patients were TP53, RB1, SMAD4, and NOTCH1. In 3.3. PFS. 1e median PFS of the 75 patients was 4.7 months previous reports, TP53 and RB1 mutations were shown to (95% CI 3.8–5.5). 1e median PFSs of male and female affect up to 90% and up to 65% of SCLC patients, respec- patients were 4.6 months (95% CI 3.6–5.6) and 6.0 months tively [13]. 1e importance of these two mutated genes in (95% CI 4.0–8.0), respectively (P � 0.777). 1e median PFSs SCLC tumorigenesis has been highlighted by numerous of never-smokers and smokers were 5.2 months (95% CI functional studies [14]. 1e frequency of SMAD4 mutation 3.9–6.5) and 4.6 months (95% CI 3.1–6.1), respectively was surprisingly high in our study. SMAD4 mediates the (P � 0.285). 1e relationship between genetic mutations and signaling of transforming growth factor beta and bone PFS was investigated. Surprisingly, patients with more than 5 morphogenic protein ligands and is a well-defined tumor mutated genes had a better PFS than patients with less than 5 suppressor in pancreatic and colon cancer [15]. SMAD4 mutated genes (6.7 versus 3.6 months, P � 0.004) (Figure 1). 4 Journal of Oncology Table 2: Characteristics of the 75 extensive-stage SCLC patients. Characteristic N (%) Gender Male/female 66 (88)/9 (12) Age <65/≥65 39 (52)/36 (48) Smoking status Never-smoker/smoker 10 (13)/65 (87) Anatomy type Central type/peripheral type 55 (74)/20 (26) Response to first-line chemotherapy PR/SD/PD 44 (59)/23 (31)/8 (10) Metastatic sites Bone 18 (24) Brain 10 (13) Liver 11 (15) Lung (contralateral) 28 (37) Pleura 20 (27) 0.00 2.00 4.00 6.00 8.00 10.00 12.00 Pericardium 3 (4) Months Kidney 3 (4) Less than 5 (n = 41) median Censored SCLC, small-cell lung cancer; PR, partial response; SD, stable disease; PD, PFS = 3.6 months progressive disease. More than 5 (n = 34) median Censored PFS = 6.7 months Table 3: Frequency of genes mutated in, at least, 10% of the 75 P = 0.004 patients. Figure 1: Kaplan–Meier curves of progression-free survival based Gene Mutant N (%) on the number of mutated genes. TP53 72 (96) RB1 58 (77) SMAD4 24 (32) mutation status of this special population. We found that NOTCH1 16 (21) TP53, RB1, SMAD4, KDR, and NOTCH1 were mutated PTEN 12 (16) frequently in both nonsmoking and female patients. 1is FGFR1 12 (16) could be explained by the fact that most of the female pa- KDR 11 (15) tients were never-smokers. In addition, nonsmoking pa- PIK3CA 11 (15) tients had a high prevalence of PTEN mutations, while ROS1 11 (15) female patients had a high prevalence of NTRK1 and RET BRCA2 10 (13) mutations. Cardona et al. reported that, among 10 never-/ ERBB4 8 (10) ever-smokers, the most frequent genetic mutations detected by NGS were TP53 (80%), RB1 (40%), CYLD (30%), EGFR Table 4: Number of mutated genes in the different subgroups. (30%), MET (20%), SMAD4 (20%), and BRIP1 (20%) [17]. In Subgroup N (median, range) P value a study by Sun et al., among 28 genetically evaluable never- smokers, the most common mutations included TP53 Gender (93%), RB1 (25%), PTEN (18%), EGFR (14%), MET (14%), Male/female 5 (2–15)/5 (3–12) 0.216 Age and SMAD4 (11%) [18]. Our data and previous findings are <65/≥65 5 (2–13)/6 (2–15) 0.209 not entirely consistent with each other, since differences Smoking status exist in terms of sample size, sequencing panel, and tumor Never-smoker/smoker 5 (3–12)/5 (2–15) 0.857 stage. Anatomy type Smoking status and gender were not shown to be related Central type/peripheral type 5 (2–15)/5 (3–13) 0.884 to PFS in our study. In an analysis of 20 SCLC patients, an improvement in survival in terms of PFS in response to first- mutations are associated with lymph node metastases, in- line treatment according to smoking status was not ob- creased angiogenesis, and more aggressive cellular behavior served, but never-smokers achieved an improvement in OS in vitro [15]. NOTCH signaling is critical for the regulation compared to smokers [17]. In another study, in 394 ex- of neuroendocrine differentiation. A previous study iden- tensive-stage SCLC patients who received first-line che- tified the presence of mutations in NOTCH family members motherapy, both PFS and OS were correlated with smoking in a quarter of analyzed SCLC samples [16], which was history [19]. Overall, according to previous studies, smoking confirmed in our study. seems to be a negative factor for survival outcomes, espe- SCLC is strongly correlated with a history of smoking cially shorter OS. While gender-related differences in PFS and mainly occurs in males, but a small portion of patients were not seen according to our data, gender may play an are nonsmoking or female. It is meaningful to explore the important role in the prognosis of SCLC. Dowlati et al. Progression-free survival rate (%) Journal of Oncology 5 Table 5: Effect of mutation status on PFS after first-line male patients [21]. Another study indicated that female sex is chemotherapy. useful as a predictor for better long-term survival [22]. Tumor mutational burden (TMB), a quantification of Gene Median PFS in months (95% CI) P value tumoral mutations, has been associated with the response TP53 0.011 to immunotherapy. In the CheckMate 032 trial, 401 pa- WT 3.4 (1.2–5.6) tients received treatment with nivolumab or a combina- Mutant 5.0 (4.1–5.9) tion of nivolumab and ipilimumab [23]. Among patients RB1 0.576 treated with combination therapy, a high TMB was related WT 6.0 (2.9–9.1) Mutant 4.7 (4.0–5.4) to better ORR and OS. In the CheckMate 227 trial, first- SMAD4 0.077 line nivolumab plus ipilimumab significantly prolonged WT 4.1 (3.1–5.1) PFS versus chemotherapy in advanced NSCLC patients Mutant 6.3 (4.9–7.7) with a high TMB (≥10 mutations/megabase) [24]. 1us, NOTCH1 0.191 TMB may be a prognostic factor for lung cancer immu- WT 4.6 (3.4–5.8) notherapy. In contrast to previous studies, blood-based Mutant 6.0 (3.1–8.9) TMB showed no value in predicting benefit with atezo- PTEN 0.666 lizumab in the IMpower133 trial, and the possible ex- WT 4.7 (3.8–5.6) planation was that the combination of platinum and Mutant 5.0 (3.0–7.0) etoposide was active and myelosuppressive [5]. 1e FGFR1 0.472 WT 4.7 (4.0–5.4) KEYNOTE 604 trial showed that pembrolizumab com- Mutant 5.7 (3.2–8.2) bined with standard first-line EP significantly improved KDR 0.522 PFS in extensive-stage SCLC patients, but TMB was not WT 4.6 (3.7–5.5) further analyzed [25]. Notably, our study showed that an Mutant 5.4 (3.3–7.5) improvement in PFS was observed in patients who had PIK3CA 0.613 more than 5 mutated genes. We may further make an WT 4.6 (3.7–5.5) assessment of TMB to verify this conclusion. Additionally, Mutant 6.0 (3.0–9.0) more basic medical research is needed to reveal the ROS1 0.608 specific mechanism of our finding. WT 4.7 (3.8–5.6) In our retrospective study, among 11 common mutated Mutant 5.7 (2.8–8.6) genes, mutant TP53 and BRCA2 were associated with better BRCA2 0.046 WT 4.5 (3.6–5.4) PFS. Using multivariate analysis, only BRCA2 was significant Mutant 6.7 (5.0–8.4) in predicting PFS. Dowlati et al. reported that patients with ERBB4 0.660 TP53 mutations had similar PFS and OS as patients with WT 4.6 (3.9–5.3) wild-type TP53 [20]. However, only 3 (4%) patients had Mutant 6.0 (4.5–7.5) wild-type TP53, and this result should be confirmed in a PFS, progression-free survival; WT, wild type; CI, confidence interval. larger sample. Dowlati et al. also reported that patients with mutant RB1 had both better OS and PFS than patients with wild-type RB1, but this was not validated in the multivariate Table 6: Multivariate Cox regression analysis of PFS after first-line analysis. BRCA2 helps repair damaged DNA and plays a chemotherapy. crucial role in ensuring the stability of genome. Patients with Factor P value HR (95% CI) BRCA2 mutations may develop genetic alterations leading to Age 0.201 cancer. Specific inherited mutations in BRCA2 may notably Bone metastasis 0.353 increase the risk of female breast and ovarian cancers, but TP53 0.068 they have also been associated with increased risks of several SMAD4 0.412 additional types of cancer. 1ere is little research indicating a NOTCH1 0.073 relationship between mutant BRCA2 and SCLC, but our Response to first-line <0.0001 3.729 (2.038–6.822) finding suggests that there might be interactions between chemotherapy mutant BRCA2 and SCLC. Number of mutated genes 0.023 1.929 (1.096–3.396) 4.581 Moreover, other similar studies are worth referencing. A BRCA2 0.002 (1.721–12.195) group of researchers found that CREBBP/EP300, TP73, or Liver metastasis 0.038 0.415 (0.181–0.951) NOTCH mutations had no influence on the survival of SCLC PFS, progression-free survival; HR, hazard ratio;CI, confidence interval. patients treated with surgery and chemotherapy [14]. A chromogenic in situ hybridization study showed that MYC amplification was associated with poor survival in SCLC and found that female sex was a positive factor for response to might be an independent prognostic factor for SCLC [26]. chemotherapy in extensive-stage SCLC patients [20]. A Our study has some limitations. First, the sample size is pooled analysis of randomized SCLC chemotherapy trials small. Second, the OS data are not shown because of the high showed that female patients survived modestly longer than loss ratio of follow-up. 6 Journal of Oncology lung cancers defined by whole exome/RNA sequencing,” 5. Conclusions Carcinogenesis, vol. 36, pp. 616–621, 2015. [8] E. Arriola, I. Canadas, M. Arumi et al., “Genetic changes in In our retrospective study, the number of mutated genes and small cell lung carcinoma,” Clinical and Translational On- BRCA2 mutation status in extensive-stage SCLC were sig- cology, vol. 10, pp. 189–197, 2008. nificantly related to PFS after first-line chemotherapy. [9] B. Tong, J. Zhao, and M. Wang, “Advances on driver mu- tations of small cell lung cancer,” Zhongguo Fei Ai Za Zhi, Data Availability vol. 19, pp. 236–240, 2016, in Chinese. [10] L. Meder, K. Konig, L. Ozretic et al., “NOTCH, ASCL1, p53 1e data used to support the findings of this study are and RB alterations define an alternative pathway driving available from the first author and the corresponding author neuroendocrine and small cell lung carcinomas,” Interna- upon request. tional Journal of Cancer, vol. 138, pp. 927–938, 2016. [11] V. Foy, M. W. Schenk, K. Baker et al., “Targeting DNA damage in SCLC,” Lung Cancer, vol. 114, pp. 12–22, 2017. Disclosure [12] M. Li, A. Lin, P. Luo et al., “DNAH10 mutation correlates with cisplatin sensitivity and tumor mutation burden in small-cell Jianlin Xu is the co-first author of this article. lung cancer,” Aging (Albany NY), vol. 12, pp. 1285–1303, 2020. [13] M. Peifer, L. Fernandez-Cuesta, M. L. Sos et al., “Integrative Conflicts of Interest genome analyses identify key somatic driver mutations of small-cell lung cancer,” Nature Genetics, vol. 44, pp. 1104– 1e authors declare that there are no conflicts of interest 1110, 2012. regarding the publication of this paper. [14] J. George, J. S. Lim, S. J. Jang et al., “Comprehensive genomic profiles of small cell lung cancer,” Nature, vol. 524, pp. 47–53, Authors’ Contributions [15] S. M. Haeger, J. J. 1ompson, S. Kalra et al., “Smad4 loss (i) Dongfang Chen and Runbo Zhong were involved in promotes lung cancer formation but increases sensitivity to conception and design. (ii) Administrative support was DNA topoisomerase inhibitors,” Oncogene, vol. 35, pp. 577– provided by Baohui Han. (iii) Jianlin Xu was responsible for 586, 2016. [16] E. A. Semenova, R. Nagel, and A. Berns, “Origins, genetic provision of study materials or patients. (iv) Collection and landscape, and emerging therapies of small cell lung cancer,” assembly of data were performed by Yizhuo Zhao. (v) Data Genes & Development, vol. 29, pp. 1447–1462, 2015. analysis and interpretation were conducted by Rong Qiao [17] A. F. Cardona, L. Rojas, Z. L. Zatarain-Barron et al., “Mul- and Tianqing Chu. (vi) All authors were involved in man- tigene mutation profiling and clinical characteristics of small- uscript writing. (vii) All authors approved the final version cell lung cancer in never-smokers vs. heavy smokers of the manuscript. (Geno1.3-CLICaP),” Frontiers in Oncology, vol. 9, p. 254, Acknowledgments [18] J. M. Sun, Y. L. Choi, J. H. Ji et al., “Small-cell lung cancer detection in never-smokers: clinical characteristics and 1e authors would like to thank all of the investigators for multigene mutation profiling using targeted next-generation their involvement in this study. 1is work was supported by sequencing,” Annals of Oncology, vol. 26, pp. 161–166, 2015. the Natural Science Foundation of Shanghai (19ZR1449700). [19] M. Ma, M. Wang, Y. Xu et al., “First-line chemotherapy and its survival analysis of 394 patients with extensive-stage small cell lung cancer in a single institute,” Zhongguo Fei Ai Za Zhi, References vol. 17, pp. 8–14, 2014, in Chinese. [20] A. Dowlati, M. B. Lipka, K. McColl et al., “Clinical correlation [1] K. Kahnert, D. Kauffmann-Guerrero, and R. M. Huber, of extensive-stage small-cell lung cancer genomics,” Annals of “SCLC-state of the art and what does the future have in store?” Oncology, vol. 27, pp. 642–647, 2016. Clinical Lung Cancer, vol. 17, pp. 325–333, 2016. [21] P. Wheatley-Price, C. Ma, L. F. Ashcroft et al., “1e strength of [2] J. P. van Meerbeeck, D. A. Fennell, and D. K. De Ruysscher, female sex as a prognostic factor in small-cell lung cancer: a “Small-cell lung cancer,” Lancet, vol. 378, pp. 1741–1755, 2011. pooled analysis of chemotherapy trials from the Manchester [3] J. W. Neal, M. A. Gubens, and H. A. Wakelee, “Current lung group and medical research council clinical trials unit,” management of small cell lung cancer,” Clinics in Chest Annals of Oncology, vol. 21, pp. 232–237, 2010. Medicine, vol. 32, pp. 853–863, 2011. [22] N. Karachaliou, A. E. Sosa, and R. Rosell, “Unraveling the [4] S. Yang, Z. Zhang, and Q. Wang, “Emerging therapies for genomic complexity of small cell lung cancer,” Translational small cell lung cancer,” Journal of Hematology & Oncology, Lung Cancer Research, vol. 5, pp. 363–366, 2016. vol. 12, p. 47, 2019. [23] S. J. Antonia, J. A. Lopez-Martin, J. Bendell et al., “Nivolumab [5] L. Horn, A. S. Mansfield, A. Szczesna et al., “First-line ate- alone and nivolumab plus ipilimumab in recurrent small-cell zolizumab plus chemotherapy in extensive-stage small-cell lung cancer,” New England Journal of Medicine, vol. 379, lung cancer (Checkmate 032): a multicentre, open-label, phase 1/2 trial,” =e Lancet Oncology, vol. 17, pp. 883–895, pp. 2220–2229, 2018. [6] M. Le Gallo, F. Lozy, and D. W. Bell, “Next-generation se- 2016. [24] M. Reck, M. Schenker, K. H. Lee et al., “Nivolumab plus quencing,” Advances in Experimental Medicine and Biology, vol. 943, pp. 119–148, 2017. ipilimumab versus chemotherapy as first-line treatment in advanced non-small-cell lung cancer with high tumour [7] R. Iwakawa, T. Kohno, Y. Totoki et al., “Expression and clinical significance of genes frequently mutated in small cell mutational burden: patient-reported outcomes results from Journal of Oncology 7 the randomised, open-label, phase III Checkmate 227 trial,” European Journal of Cancer, vol. 116, pp. 137–147, 2019. [25] C. M. Rudin, M. M. Awad, A. Navarro et al., “Pembrolizumab or placebo plus etoposide and platinum as first-line therapy for extensive-stage small-cell lung cancer: randomized, double-blind, phase III Keynote-604 study,” Journal of Clinical Oncology, vol. 38, pp. 2369–2379, 2020. [26] C. Alves Rde, R. T. Meurer, and A. V. Roehe, “MYC am- plification is associated with poor survival in small cell lung cancer: a chromogenic in situ hybridization study,” Journal of Cancer Research and Clinical Oncology, vol. 140, pp. 2021– 2025, 2014.

Journal

Journal of OncologyHindawi Publishing Corporation

Published: Nov 19, 2020

There are no references for this article.