Research Paper Volume 11, Issue 8 pp 2369—2377

Additional data support the role of LINC00673 rs11655237 C>T in the development of neuroblastoma

Yong Li1, , Zhen-Jian Zhuo2, , Haiyan Zhou3, , Jiabin Liu2, , Zan Liu1, , Jiao Zhang4, , Jiwen Cheng5, , Suhong Li6, , Haixia Zhou7, , Rong Zhou8, , Jing He2, , Yaowang Zhao1, ,

  • 1 Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha 41004, China
  • 2 Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
  • 3 Department of Pathology, Xiang-ya School of Medicine, Central South University, Changsha 410013, China
  • 4 Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
  • 5 Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
  • 6 Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan 030013, China
  • 7 Department of Hematology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
  • 8 Science and Education Section, Hunan Children’s Hospital, Changsha 410004, China
* Equal contribution

Received: February 18, 2019       Accepted: April 14, 2019       Published: April 20, 2019      

https://doi.org/10.18632/aging.101920
How to Cite

Copyright: Li et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Neuroblastoma is the most frequently diagnosed neural tumor of childhood. Abnormal function of the long intergenic non-coding RNA (lincRNA) LINC00673 has been implicated in various human malignancies. Genome-wide association studies revealed the LINC00673 rs11655237 C>T polymorphism to be associated with the risk of neuroblastoma, though the effect was not well defined, in part due to the small sample size in our earlier study. Herein, we verified the impact of LINC00673 rs11655237 C>T on the risk of neuroblastoma in 700 cases and 1516 controls from six centers in China. After pooling all enrolled patients, we observed a significant association between LINC00673 rs11655237 C>T and risk of neuroblastoma (TT vs. CC: adjusted odds ratio [OR]=1.58, 95% confidence interval [CI]=1.06–2.35, P=0.024; additive model: adjusted OR=1.20, 95% CI=1.03–1.39, P=0.020; recessive model: adjusted OR=1.50, 95% CI=1.02–2.22, P=0.040). Stratification analysis revealed a significant relationship between rs11655237 CT/TT and neuroblastoma risk in subgroups of males, patients whose tumor originated in the adrenal gland, and patients with clinical stage IV disease. These findings add new evidence of the importance of LINC00673 rs11655237 C>T to the risk of developing neuroblastoma.

Introduction

Neuroblastoma is the most commonly seen solid childhood tumor outside the brain [1, 2]. It generally results from a differentiation failure of neural crest cell precursors [3]. Despite being relatively rare (nearly 8% of all pediatric cancer diagnoses), neuroblastoma disproportionately contributes to about 15% of all childhood cancer-related mortality [46]. Outcomes greatly vary among neuroblastoma patients, spanning a range from spontaneous regression with little or no treatment to progression despite aggressive, multimodal therapy [7]. At present, the five-year survival rate for patients with metastatic high-risk neuroblastoma is less than 50% after multimodal therapy [8].

Neuroblastoma is a heterogenous cancer influenced by both environmental and genetic factors. Previous studies have shown that children and child-bearing women exposed to certain kinds of environmental factors are more likely to develop neuroblastoma [911]. In addition, genetic factors, such as ALK [12, 13] and PHOX2B [14, 15] mutations, have also been implicated in neuroblastoma progression. Recent progress in genome-wide association studies (GWASs) has helped to identify single nucleotide polymorphisms (SNPs) that contribute to neuroblastoma risk. So far, it appears that SNPs in LMO1 [16], TP53 [17], LIN28B [18], BARD1 [19], DUSP12 [20], HACE1 [18], NEFL [21] and CDKN1B [22] contribute to the risk of neuroblastoma. However, the currently identified variants do not fully account for the etiology of neuroblastoma.

Long intergenic non-coding RNAs (lincRNAs) are a class of transcripts that are longer than 200 nucleotides but do not code for a protein [23]. LincRNAs modulate cellular activities through multiple mechanisms, including transcriptional regulation, epigenetic regulation, imprinting, genome rearrangement, and chromatin modification [24]. Over the past decade, a variety of lincRNAs have been shown to play key roles in human disorders, including cancers [25, 26]. For example, LINC00673 (OMIM No. 617079) has been implicated in the development and prognosis of several malignancies [2729], and polymorphism in LINC00673, rs11655237 C>T (also reported as G>A elsewhere), was identified as being significantly associated with susceptibility to pancreatic cancer [30]. In an earlier investigation, we observed a significantly increased risk of neuroblastoma in subjects carrying the T allele of LINC00673 rs11655237 [31]. This prompted us to further evaluate the relationship between LINC00673 rs11655237 C>T and the risk of neuroblastoma in a larger sample.

Results

Association between the LINC00673 rs11655237 C>T and neuroblastoma risk

The baseline characteristics of the neuroblastoma patients and controls are summarized in Supplementary Table 1 and in our previously published articles [3235]. There were no significant differences in the age or gender distributions between the cases and controls, which indicates that the frequency matching was adequate. The genotype frequencies of LINC00673 rs11655237 C>T for all the subjects and their contributions to neuroblastoma risk are listed in Table 1. The observed genotype distributions of the rs11655237 C>T polymorphism among the controls were consistent with the Hardy-Weinberg equilibrium (HWE) for both the Hunan subjects (HWE=0.268) and the combined patient sample (HWE=0.824). We detected no significant association between LINC00673 rs11655237 C>T and neuroblastoma risk among the Hunan subjects. After pooling all the subjects, however, we found that those harboring the T allele were significantly more likely to develop neuroblastoma (TT vs. CC: adjusted odds ratio [OR]=1.58, 95% confidence interval [CI]=1.06–2.35, P=0.024; additive model: adjusted OR=1.20, 95% CI=1.03–1.39, P=0.020; recessive model: adjusted OR=1.50, 95% CI=1.02–2.22, P=0.040).

Table 1. LINC00673 rs11655237 C>T polymorphism and neuroblastoma susceptibility.

GenotypeCases (N=698)Controls (N=1516)PaCrude OR (95% CI)PAdjusted OR (95% CI)bPb
Hunan subjects (HWE=0.268)
CC101 (62.35)165 (61.11)1.001.00
CT54 (33.33)96 (35.56)0.92 (0.61–1.39)0.6900.91 (0.60–1.38)0.648
TT7 (4.32)9 (3.33)1.27 (0.46–3.52)0.6451.57 (0.56–4.39)0.395
Additive0.8040.99 (0.70–1.40)0.9651.02 (0.72–1.45)0.906
Dominant61 (37.65)105 (38.89)0.7980.95 (0.64–1.42)0.7990.96 (0.64–1.43)0.830
Recessive155 (95.68)261 (96.67)0.5991.31 (0.48–3.59)0.6001.62 (0.58–4.50)0.355
Combined (HWE=0.824)
CC401 (57.45)935 (61.68)1.001.00
CT252 (36.10)513 (33.84)1.15 (0.95–1.39)0.1631.14 (0.94–1.38)0.175
TT45 (6.45)68 (4.49)1.54 (1.042.29)0.0311.58 (1.062.35)0.024
Additive0.0571.19 (1.031.38)0.0221.20 (1.031.39)0.020
Dominant297 (42.55)581 (38.32)0.0591.19 (0.99–1.43)0.0591.19 (0.99–1.43)0.059
Recessive653 (93.55)1448 (95.51)0.0511.47 (0.996–2.16)0.0531.50 (1.022.22)0.040
OR, odds ratio; CI, confidence interval; HWE, Hardy-Weinberg equilibrium.
aχ2 test for genotype distributions between neuroblastoma cases and cancer-free controls.
bAdjusted for age and gender.

Stratification analysis

To identify vulnerable subgroups, we stratified the subjects based on selected variables (Table 2). This enabled us to detect a significant association between rs11655237 CT/TT and neuroblastoma risk in males (adjusted OR=1.34, 95% CI=1.05–1.71, P=0.019). Taking sites of tumor origin into consideration, we observed that carriers of the rs11655237 CT/TT genotype were more likely to develop tumors originating in the adrenal gland (adjusted OR=1.36, 95% CI=1.02–1.81, P=0.039). Moreover, carriers of the CT/TT genotypes had a significantly higher risk of INSS clinical stage IV disease than carriers of the CC genotype (adjusted OR=1.50, 95% CI=1.11–2.04, P=0.009).

Table 2. Stratified results for LINC00673 rs11655237 C>T polymorphism and neuroblastoma susceptibility in the combined patient sample.

Variablesrs11655237 (cases/controls)OR (95% CI)PAOR (95% CI)aPa
CCCT/TT
Age, month
 ≤18161/378112/2371.11 (0.83–1.48)0.4841.12 (0.83–1.49)0.459
 >18240/557185/3441.25 (0.99–1.58)0.0641.25 (0.99–1.58)0.065
Gender
 Females182/393125/2631.03 (0.78–1.35)0.8541.03 (0.78–1.35)0.853
 Males219/542172/3181.34 (1.051.71)0.0191.34 (1.051.71)0.019
Sites of origin
 Adrenal gland117/93598/5811.35 (1.011.80)0.0421.36 (1.021.81)0.039
 Retroperitoneal137/935101/5811.19 (0.90–1.57)0.2271.19 (0.90–1.57)0.218
 Mediastinum107/93570/5811.05 (0.77–1.45)0.7521.04 (0.76–1.44)0.790
 Others38/93522/5810.93 (0.55–1.59)0.7960.93 (0.55–1.59)0.795
Clinical stages
 I123/93593/5811.22 (0.91–1.62)0.1831.22 (0.91–1.62)0.187
 II78/93551/5811.05 (0.73–1.52)0.7861.04 (0.72–1.50)0.833
 III88/93546/5810.84 (0.58–1.22)0.3620.84 (0.58–1.21)0.345
 IV101/93594/5811.50 (1.112.02)0.0081.50 (1.112.04)0.009
 4s6/93510/5812.68 (0.97–7.42)0.0572.80 (0.98–7.95)0.054
 I+II+4s201/935144/5811.15 (0.91–1.46)0.2411.15 (0.91–1.46)0.248
 III+IV189/935140/5811.19 (0.94–1.52)0.1551.19 (0.93–1.52)0.165
OR, odds ratio; CI, confidence interval; AOR, adjusted odds ratio.
aAdjusted for age and gender, omitting the corresponding stratification factor.

False-positive report probability analysis

In a false-positive report probability analysis (Table 3) at a prior probability level of 0.1, a significant association with neuroblastoma risk was noteworthy for males and clinical stage IV patients carrying CT/TT genotypes. At a prior probability level of 0.25, the increased neuroblastoma risk remains noteworthy in carriers of the rs11655237 TT and rs12587 CT/TT genotypes for the male, adrenal tumor, and stage IV subgroups.

Table 3. False-positive report probability analysis for significant findings in the combined patient sample.

GenotypeCrude OR (95% CI)PaStatistical powerbPrior probability
0.250.10.010.0010.0001
TT vs. CC1.54 (1.04–2.29)0.0310.4690.1660.3740.8680.9850.998
CT/TT vs. CC
Males1.34 (1.05–1.71)0.0190.8230.0630.1680.6900.9570.996
Adrenal gland1.35 (1.01–1.80)0.0420.7690.1410.3290.8440.9820.998
Stage IV1.50 (1.11–2.02)0.0080.5090.0460.1270.6150.9410.994
OR, odds ratio; CI, confidence interval.
aChi-square test was used to calculate the genotype frequency distributions.
bStatistical power was calculated using the number of observations in the subgroup and the OR and P values in this table.

Discussion

In the present study, we further evaluated the association between LINC00673 rs11655237 C>T and the risk of neuroblastoma using a larger sample. Our results indicate that LINC00673 rs11655237 C>T is significantly associated with increased neuroblastoma susceptibility, which further highlights the important contribution of LINC00673 rs11655237 C>T to the risk of neuroblastoma in Chinese children.

LINC00673 is located on chromosome 17q24.3, which exhibits a high frequency of loss of heterozygosity [36]. In 2011, a rs11655237 C>T variant was first documented by Cabiliet et al. [37]. Later, a GWAS showed rs11655237 C>T to be associated with pancreatic cancer susceptibility [30]. A subsequent study by Zheng et al. [38] confirmed the relationship between rs11655237 and the risk of pancreatic ductal adenocarcinoma (PDAC) in a Chinese population and shed light on the molecular mechanism by which the polymorphism confers PDAC risk. They found that the C-to-T shift at rs11655237 creates a target site for miR-1231 binding and interferes with ubiquitination and degradation of PTPN11. They also demonstrated that the rs11655237 T allele can impair LINC00673 activity, thereby triggering SRC-ERK oncogenic signaling and ultimately resulting in a higher risk for developing PDAC. Intriguingly, LINC00673 functions as a tumor suppressor or promoter in different cancer types. Huang et al. [29] found that LINC00673 is upregulated in gastric cancer and is associated with a poor prognosis. Investigation into the mechanism suggested that LINC00673 is activated by SP1 and exerts oncogenic effects in part through interaction with LSD1 and EZH2. Lu et al. [39] also identified LINC00673 as an oncogenic mediator in non-small cell lung cancer. They demonstrated that LINC00673 promotes non-small cell lung cancer cell proliferation, migration, invasion, and epithelial mesenchymal transition by sponging miR-150-5p. In addition, research conducted by Yu et al. [40] showed that LINC00673 is highly expressed in human tongue squamous cell carcinoma and correlates with a poor prognosis. Up to now, however, the possible activity of LINC00673 in neuroblastoma remained unexplored.

Considering the important functional role of LINC00673 in malignancies, we conducted the first case-control study to investigate the association between LINC00673 rs11655237 C>T polymorphism and neuroblastoma risk in a Chinese population [31]. In that two-center study with 393 neuroblastoma patients and 812 healthy controls, we found that LINC00673 rs11655237 C>T polymorphism confers a high risk of neuroblastoma. More recently, the epidemiological role of LINC00673 rs11655237 C>T was examined within the context of cervical cancer [41]. Wang et al. observed that rs11655237 contributes significantly to a higher susceptibility to cervical cancer in a Chinese population, which they speculated may reflect decreased LINC00673 expression caused by the T allele. In the present study, we further investigated the association between LINC00673 rs11655237 C>T and neuroblastoma risk using a larger sample from multiple centers in China. Unexpectedly, we did not detect a significant contribution of LINC00673 rs11655237 C>T to neuroblastoma risk in the patient sample from Hunan. However, significant associations were revealed after combining the patients from all six centers. This finding is biologically plausible, as the relatively small sample size and low-penetrance of a single polymorphism could account for the null association. Interestingly, the impact of LINC00673 rs11655237 C>T on susceptibility to neuroblastoma was similar to that for pancreatic cancer. This suggests LINC00673 rs11655237 C>T may confer neuroblastoma and pancreatic cancer risk via similar molecular mechanisms.

This is a pioneer multi-center case-control study examining the contribution of LINC00673 rs11655237 C>T to neuroblastoma risk. The study has several minor limitations. First, despite our relatively large sample size, the numbers in some stratified analyses were not sufficient to provide convincing statistical power. Second, because all the included subjects were from hospitals located in China, inherent selection bias cannot be completely excluded and the conclusions drawn may not be representative for other populations. Third, the single SNP analyzed here is far from enough to elucidate the full spectrum of neuroblastoma etiologies. Analysis of additional SNPs and use of additional methods of analysis (e.g., find mapping) will help to identify more neuroblastoma-associated loci [42]. Finally, SNP-SNP and SNP-environmental factor interaction analyses are absent. The role of LINC00673 rs11655237 C>T in neuroblastoma may be modified by such interactions.

In summary, our findings support the notion that LINC00673 rs11655237 C>T polymorphism may predispose one to neuroblastoma. That conclusion needs to be by further substantiated by research exploring the possible mechanisms by which LINC00673 rs11655237 C>T could modulate neuroblastoma risk.

Materials and Methods

Study subjects

In brief, we carried out a six-hospital-based case-control study in China. A total of 162 cases and 270 controls from Hunan Children’s Hospital were freshly genotyped in the present study. The other study subjects have been described in detail elsewhere [32, 34, 43]. In all, 700 cases and 1516 controls were recruited from six regional hospitals in China (275 cases and 531 controls from Guangzhou Women and Children’s Medical Center, 76 cases and 186 controls from The Second Affiliated Hospital of Xi’an Jiaotong University, 36 cases and 72 controls from The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, 118 cases and 281 controls from The First Affiliated Hospital of Zhengzhou University, 33 cases and 176 controls from Children’s Hospital of Shanxi, and 162 cases and 270 controls from Hunan Children’s Hospital). Eligible controls, frequency-matched to cases with respect to age, sex, and study center, were recruited from the same region as the cases during the same period. Informed consent was obtained from all subjects or their guardians, and the study was approved by the institutional review boards of all the participating hospitals.

Genotyping

DNA was extracted from blood samples collected from the study participants. For genotyping, TaqMan methodology was according to the manufacturer’s instructions [4447]. For quality control, technicians were blind to the status of the samples, and 10% of the samples were genotyped twice to ensure genotyping accuracy. The results for the random duplicate samples were 100% concordant.

Statistical analysis

The χ2 test was used to determine whether the observed genotype frequencies among the control subjects were in line with the Hardy-Weinberg equilibrium. The χ2 test was also adopted to compare differences in selected demographic variables and genotype frequencies between the cases and controls. Multivariable logistic regression analysis adjusted for age and gender was conducted to determine the association between rs11655237 C>T and neuroblastoma risk based on ORs and their 95% CIs. Values of P<0.05 were considered significant. We also performed a false-positive report probability analysis, as described previously [4850]. SAS ver. 9.1 (SAS Institute Inc., Cary, NC, USA) was used for all statistical analyses.

Supplementary Materials

Supplementary Table 1

Conflicts of Interest

There are no competing interests to declare.

Funding

This study was supported by grants from Pearl River S&T Nova Program of Guangzhou (No: 201710010086), and Hunan Provincial Natural Science Foundation Project (No: 2018JJ2210), and Hunan provincial key laboratory of Pediatric emergency medicine (No: 2018TP1028).

References

  • 1. Matthay KK, Maris JM, Schleiermacher G, Nakagawara A, Mackall CL, Diller L, Weiss WA. Neuroblastoma. Nat Rev Dis Primers. 2016; 2:16078. https://doi.org/10.1038/nrdp.2016.78 [PubMed]
  • 2. Capasso M, Diskin SJ. Genetics and genomics of neuroblastoma. Cancer Treat Res. 2010; 155:65–84. https://doi.org/10.1007/978-1-4419-6033-7_4 [PubMed]
  • 3. Maris JM, Matthay KK. Molecular biology of neuroblastoma. J Clin Oncol. 1999; 17:2264–79. https://doi.org/10.1200/JCO.1999.17.7.2264 [PubMed]
  • 4. Irwin MS, Park JR. Neuroblastoma: paradigm for precision medicine. Pediatr Clin North Am. 2015; 62:225–56. https://doi.org/10.1016/j.pcl.2014.09.015 [PubMed]
  • 5. Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, Burchill S, Boubaker A, McHugh K, Nuchtern JG, London WB, Seibel NL, Lindwasser OW, et al. Revisions to the International Neuroblastoma Response Criteria: A Consensus Statement From the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol. 2017; 35:2580–87. https://doi.org/10.1200/JCO.2016.72.0177 [PubMed]
  • 6. Esposito MR, Aveic S, Seydel A, Tonini GP. Neuroblastoma treatment in the post-genomic era. J Biomed Sci. 2017; 24:14. https://doi.org/10.1186/s12929-017-0319-y [PubMed]
  • 7. Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet. 2007; 369:2106–20. https://doi.org/10.1016/S0140-6736(07)60983-0 [PubMed]
  • 8. Whittle SB, Smith V, Doherty E, Zhao S, McCarty S, Zage PE. Overview and recent advances in the treatment of neuroblastoma. Expert Rev Anticancer Ther. 2017; 17:369–86. https://doi.org/10.1080/14737140.2017.1285230 [PubMed]
  • 9. Cook MN, Olshan AF, Guess HA, Savitz DA, Poole C, Blatt J, Bondy ML, Pollock BH. Maternal medication use and neuroblastoma in offspring. Am J Epidemiol. 2004; 159:721–31. https://doi.org/10.1093/aje/kwh108 [PubMed]
  • 10. Menegaux F, Olshan AF, Neglia JP, Pollock BH, Bondy ML. Day care, childhood infections, and risk of neuroblastoma. Am J Epidemiol. 2004; 159:843–51. https://doi.org/10.1093/aje/kwh111 [PubMed]
  • 11. Ferrís i Tortajada J, Ortega García JA, García i Castell J, López Andreu JA, Berbel Tornero O, Crehuá Gaudiza E. [Risk factors for neuroblastoma]. . An Pediatr (Barc). 2005; 63:50–60. https://doi.org/10.1157/13076768 [PubMed]
  • 12. George RE, Sanda T, Hanna M, Fröhling S, Luther W2nd, Zhang J, Ahn Y, Zhou W, London WB, McGrady P, Xue L, Zozulya S, Gregor VE, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature. 2008; 455:975–78. https://doi.org/10.1038/nature07397 [PubMed]
  • 13. Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M, Wang L, Soda M, Kikuchi A, Igarashi T, Nakagawara A, Hayashi Y, Mano H, Ogawa S. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008; 455:971–74. https://doi.org/10.1038/nature07399 [PubMed]
  • 14. Trochet D, Bourdeaut F, Janoueix-Lerosey I, Deville A, de Pontual L, Schleiermacher G, Coze C, Philip N, Frébourg T, Munnich A, Lyonnet S, Delattre O, Amiel J. Germline mutations of the paired-like homeobox 2B (PHOX2B) gene in neuroblastoma. Am J Hum Genet. 2004; 74:761–64. https://doi.org/10.1086/383253 [PubMed]
  • 15. Perri P, Bachetti T, Longo L, Matera I, Seri M, Tonini GP, Ceccherini I. PHOX2B mutations and genetic predisposition to neuroblastoma. Oncogene. 2005; 24:3050–53. https://doi.org/10.1038/sj.onc.1208532 [PubMed]
  • 16. Oldridge DA, Wood AC, Weichert-Leahey N, Crimmins I, Sussman R, Winter C, McDaniel LD, Diamond M, Hart LS, Zhu S, Durbin AD, Abraham BJ, Anders L, et al. Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature. 2015; 528:418–21. https://doi.org/10.1038/nature15540 [PubMed]
  • 17. Diskin SJ, Capasso M, Diamond M, Oldridge DA, Conkrite K, Bosse KR, Russell MR, Iolascon A, Hakonarson H, Devoto M, Maris JM. Rare variants in TP53 and susceptibility to neuroblastoma. J Natl Cancer Inst. 2014; 106:dju047. https://doi.org/10.1093/jnci/dju047 [PubMed]
  • 18. Diskin SJ, Capasso M, Schnepp RW, Cole KA, Attiyeh EF, Hou C, Diamond M, Carpenter EL, Winter C, Lee H, Jagannathan J, Latorre V, Iolascon A, et al. Common variation at 6q16 within HACE1 and LIN28B influences susceptibility to neuroblastoma. Nat Genet. 2012; 44:1126–30. https://doi.org/10.1038/ng.2387 [PubMed]
  • 19. Latorre V, Diskin SJ, Diamond MA, Zhang H, Hakonarson H, Maris JM, Devoto M. Replication of neuroblastoma SNP association at the BARD1 locus in African-Americans. Cancer Epidemiol Biomarkers Prev. 2012; 21:658–63. https://doi.org/10.1158/1055-9965.EPI-11-0830 [PubMed]
  • 20. Nguyen B, Diskin SJ, Capasso M, Wang K, Diamond MA, Glessner J, Kim C, Attiyeh EF, Mosse YP, Cole K, Iolascon A, Devoto M, Hakonarson H, et al. Phenotype restricted genome-wide association study using a gene-centric approach identifies three low-risk neuroblastoma susceptibility Loci. PLoS Genet. 2011; 7:e1002026. https://doi.org/10.1371/journal.pgen.1002026 [PubMed]
  • 21. Capasso M, Diskin S, Cimmino F, Acierno G, Totaro F, Petrosino G, Pezone L, Diamond M, McDaniel L, Hakonarson H, Iolascon A, Devoto M, Maris JM. Common genetic variants in NEFL influence gene expression and neuroblastoma risk. Cancer Res. 2014; 74:6913–24. https://doi.org/10.1158/0008-5472.CAN-14-0431 [PubMed]
  • 22. Capasso M, McDaniel LD, Cimmino F, Cirino A, Formicola D, Russell MR, Raman P, Cole KA, Diskin SJ. The functional variant rs34330 of CDKN1B is associated with risk of neuroblastoma. J Cell Mol Med. 2017; 21:3224–30. https://doi.org/10.1111/jcmm.13226 [PubMed]
  • 23. Ransohoff JD, Wei Y, Khavari PA. The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 2018; 19:143–57. https://doi.org/10.1038/nrm.2017.104 [PubMed]
  • 24. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014; 15:7–21. https://doi.org/10.1038/nrg3606 [PubMed]
  • 25. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010; 464:1071–76. https://doi.org/10.1038/nature08975 [PubMed]
  • 26. Chung S, Nakagawa H, Uemura M, Piao L, Ashikawa K, Hosono N, Takata R, Akamatsu S, Kawaguchi T, Morizono T, Tsunoda T, Daigo Y, Matsuda K, et al. Association of a novel long non-coding RNA in 8q24 with prostate cancer susceptibility. Cancer Sci. 2011; 102:245–52. https://doi.org/10.1111/j.1349-7006.2010.01737.x [PubMed]
  • 27. Xia E, Shen Y, Bhandari A, Zhou X, Wang Y, Yang F, Wang O. Long non-coding RNA LINC00673 promotes breast cancer proliferation and metastasis through regulating B7-H6 and epithelial-mesenchymal transition. Am J Cancer Res. 2018; 8:1273–87. https://doi.org/10.3892/ijo.2018.4524 [PubMed]
  • 28. Roth A, Boulay K, Groß M, Polycarpou-Schwarz M, Mallette FA, Regnier M, Bida O, Ginsberg D, Warth A, Schnabel PA, Muley T, Meister M, Zabeck H, et al. Restoring LINC00673 expression triggers cellular senescence in lung cancer. RNA Biol. 2018; 15:1499–511. https://doi.org/10.1080/15476286.2018.1553481 [PubMed]
  • 29. Huang M, Hou J, Wang Y, Xie M, Wei C, Nie F, Wang Z, Sun M. Long Noncoding RNA LINC00673 Is Activated by SP1 and Exerts Oncogenic Properties by Interacting with LSD1 and EZH2 in Gastric Cancer. Mol Ther. 2017; 25:1014–26. https://doi.org/10.1016/j.ymthe.2017.01.017 [PubMed]
  • 30. Childs EJ, Mocci E, Campa D, Bracci PM, Gallinger S, Goggins M, Li D, Neale RE, Olson SH, Scelo G, Amundadottir LT, Bamlet WR, Bijlsma MF, et al. Common variation at 2p13.3, 3q29, 7p13 and 17q25.1 associated with susceptibility to pancreatic cancer. Nat Genet. 2015; 47:911–16. https://doi.org/10.1038/ng.3341 [PubMed]
  • 31. Zhang Z, Chang Y, Jia W, Zhang J, Zhang R, Zhu J, Yang T, Xia H, Zou Y, He J. LINC00673 rs11655237 C>T confers neuroblastoma susceptibility in Chinese population. Biosci Rep. 2018; 38:BSR20171667. https://doi.org/10.1042/BSR20171667 [PubMed]
  • 32. Wang J, Zhuo Z, Chen M, Zhu J, Zhao J, Zhang J, Chen S, He J, Zhou H. RAN/RANBP2 polymorphisms and neuroblastoma risk in Chinese children: a three-center case-control study. Aging (Albany NY). 2018; 10:808–18. https://doi.org/10.18632/aging.101429 [PubMed]
  • 33. Zhuo ZJ, Zhang R, Zhang J, Zhu J, Yang T, Zou Y, He J, Xia H. Associations between lncRNA MEG3 polymorphisms and neuroblastoma risk in Chinese children. Aging (Albany NY). 2018; 10:481–91. https://doi.org/10.18632/aging.101406 [PubMed]
  • 34. Cheng J, Zhuo Z, Xin Y, Zhao P, Yang W, Zhou H, Zhang J, Gao Y, He J, Li P. Relevance of XPD polymorphisms to neuroblastoma risk in Chinese children: a four-center case-control study. Aging (Albany NY). 2018; 10:1989–2000. https://doi.org/10.18632/aging.101522 [PubMed]
  • 35. Zhuo ZJ, Liu W, Zhang J, Zhu J, Zhang R, Tang J, Yang T, Zou Y, He J, Xia H. Functional Polymorphisms at ERCC1/XPF Genes Confer Neuroblastoma Risk in Chinese Children. EBioMedicine. 2018; 30:113–19. https://doi.org/10.1016/j.ebiom.2018.03.003 [PubMed]
  • 36. Tseng RC, Chang JW, Hsien FJ, Chang YH, Hsiao CF, Chen JT, Chen CY, Jou YS, Wang YC. Genomewide loss of heterozygosity and its clinical associations in non small cell lung cancer. Int J Cancer. 2005; 117:241–47. https://doi.org/10.1002/ijc.21178 [PubMed]
  • 37. Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011; 25:1915–27. https://doi.org/10.1101/gad.17446611 [PubMed]
  • 38. Zheng J, Huang X, Tan W, Yu D, Du Z, Chang J, Wei L, Han Y, Wang C, Che X, Zhou Y, Miao X, Jiang G, et al. Pancreatic cancer risk variant in LINC00673 creates a miR-1231 binding site and interferes with PTPN11 degradation. Nat Genet. 2016; 48:747–57. https://doi.org/10.1038/ng.3568 [PubMed]
  • 39. Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen HM, Wu H, Xia D, Wu Y. Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer. 2017; 16:118. https://doi.org/10.1186/s12943-017-0685-9 [PubMed]
  • 40. Yu J, Liu Y, Gong Z, Zhang S, Guo C, Li X, Tang Y, Yang L, He Y, Wei F, Wang Y, Liao Q, Zhang W, et al. Overexpression long non-coding RNA LINC00673 is associated with poor prognosis and promotes invasion and metastasis in tongue squamous cell carcinoma. Oncotarget. 2017; 8:16621–32. https://doi.org/10.18632/oncotarget.14200 [PubMed]
  • 41. Wang Y, Luo T. LINC00673 rs11655237 Polymorphism Is Associated With Increased Risk of Cervical Cancer in a Chinese Population. Cancer Control. 2018; 25:1073274818803942. https://doi.org/10.1177/1073274818803942 [PubMed]
  • 42. Cimmino F, Avitabile M, Diskin SJ, Vaksman Z, Pignataro P, Formicola D, Cardinale A, Testori A, Koster J, de Torres C, Devoto M, Maris JM, Iolascon A, Capasso M. Fine mapping of 2q35 high-risk neuroblastoma locus reveals independent functional risk variants and suggests full-length BARD1 as tumor-suppressor. Int J Cancer. 2018; 143:2828–37. https://doi.org/10.1002/ijc.31822 [PubMed]
  • 43. He J, Zou Y, Liu X, Zhu J, Zhang J, Zhang R, Yang T, Xia H. Association of Common Genetic Variants in Pre-microRNAs and Neuroblastoma Susceptibility: A Two-Center Study in Chinese Children. Mol Ther Nucleic Acids. 2018; 11:1–8. https://doi.org/10.1016/j.omtn.2018.01.003 [PubMed]
  • 44. Chang J, Tian J, Yang Y, Zhong R, Li J, Zhai K, Ke J, Lou J, Chen W, Zhu B, Shen N, Zhang Y, Gong Y, et al. A Rare Missense Variant in TCF7L2 Associates with Colorectal Cancer Risk by Interacting with a GWAS-Identified Regulatory Variant in the MYC Enhancer. Cancer Res. 2018; 78:5164–72. https://doi.org/10.1158/0008-5472.CAN-18-0910 [PubMed]
  • 45. Chang J, Tian J, Zhu Y, Zhong R, Zhai K, Li J, Ke J, Han Q, Lou J, Chen W, Zhu B, Shen N, Zhang Y, et al. Exome-wide analysis identifies three low-frequency missense variants associated with pancreatic cancer risk in Chinese populations. Nat Commun. 2018; 9:3688. https://doi.org/10.1038/s41467-018-06136-x [PubMed]
  • 46. Chang J, Zhong R, Tian J, Li J, Zhai K, Ke J, Lou J, Chen W, Zhu B, Shen N, Zhang Y, Zhu Y, Gong Y, et al. Exome-wide analyses identify low-frequency variant in CYP26B1 and additional coding variants associated with esophageal squamous cell carcinoma. Nat Genet. 2018; 50:338–43. https://doi.org/10.1038/s41588-018-0045-8 [PubMed]
  • 47. Li J, Chang J, Tian J, Ke J, Zhu Y, Yang Y, Gong Y, Zou D, Peng X, Yang N, Mei S, Wang X, Cheng L, et al. A Rare Variant P507L in TPP1 Interrupts TPP1-TIN2 Interaction, Influences Telomere Length, and Confers Colorectal Cancer Risk in Chinese Population. Cancer Epidemiol Biomarkers Prev. 2018; 27:1029–35. https://doi.org/10.1158/1055-9965.EPI-18-0099 [PubMed]
  • 48. He J, Wang MY, Qiu LX, Zhu ML, Shi TY, Zhou XY, Sun MH, Yang YJ, Wang JC, Jin L, Wang YN, Li J, Yu HP, Wei QY. Genetic variations of mTORC1 genes and risk of gastric cancer in an Eastern Chinese population. Mol Carcinog. 2013 (Suppl 1); 52:E70–79. https://doi.org/10.1002/mc.22013 [PubMed]
  • 49. Fu W, Zhu J, Xiong SW, Jia W, Zhao Z, Zhu SB, Hu JH, Wang FH, Xia H, He J, Liu GC. BARD1 Gene Polymorphisms Confer Nephroblastoma Susceptibility. EBioMedicine. 2017; 16:101–05. https://doi.org/10.1016/j.ebiom.2017.01.038 [PubMed]
  • 50. He J, Zhang X, Zhang J, Zhang R, Yang T, Zhu J, Xia H, Zou Y. LMO1 super-enhancer polymorphism rs2168101 G>T correlates with decreased neuroblastoma risk in Chinese children. J Cancer. 2018; 9:1592–97. https://doi.org/10.7150/jca.24326 [PubMed]