Since the first reports in 2009, genome-wide association studies (GWAS) have been successful in identifying germline variants associated with glioma susceptibility. In this review, we describe a chronological history of glioma GWAS, culminating in the most recent study comprising 12,496 cases and 18,190 controls. We additionally summarize associations at the 27 glioma-risk SNPs that have been reported so far. Future efforts are likely to be principally focused on assessing association of germline-risk SNPs with particular molecular subgroups of glioma, as well as investigating the functional basis of the risk loci in tumor formation. These ongoing studies will be important to maximize the impact of research into glioma susceptibility, both in terms of insight into tumor etiology as well as opportunities for clinical translation. Cancer Epidemiol Biomarkers Prev; 27(4); 418–28. ©2018 AACR.

See all articles in this CEBP Focus section, “Genome-Wide Association Studies in Cancer.”

Gliomas account for approximately 80% of all malignant primary brain tumors with approximately 26,000 individuals diagnosed in Europe each year (1, 2). Diffuse gliomas have traditionally been classified into oligodendroglial and astrocytic tumors and graded II to IV, with the revised 2016 World Health Organization classification additionally incorporating the tumor molecular features of IDH1/IDH2 (IDH) mutation and 1p/19q codeletion (3). The most common tumor type, glioblastoma multiforme (GBM), typically has a median survival of only 15 months (2).

To date the only recognized environmental risk factor for glioma is exposure to ionizing radiation, which is responsible for only a very small number of cases (2). Evidence for a genetic component to glioma risk is provided by observations of an approximately 2-fold elevated risk in relatives of glioma patients (4). Furthermore, an increased risk of glioma is recognized to be associated with a number of Mendelian cancer predisposition syndromes, notable neurofibromatosis (NF1 and NF2), Li-Fraumeni, and Turcot (5–16). In addition, germline mutation of CDKN2A has been reported to be a cause of the astrocytoma–melanoma syndrome (17, 18). More recently, germline POT1 variants have been implicated in development of oligodendroglioma (19); however, their penetrance is yet to be established. Collectively, these syndromes are very rare and explain little of the risk of glioma.

Attempts to study the contribution of low-frequency variants of moderate effect have generally been through sequencing of candidate genes in glioma cases and controls; however, they have failed to identify any genes associated with glioma. Indeed, a recent study of 1,662 cases and 1,301 controls failed to replicate 52 variants previously implicated by candidate gene studies (20). In combination with more systematic approaches such as those making use of the exome array (21), these studies do not currently provide support for this class of susceptibility allele playing a major role in glioma predisposition.

More fruitful have been efforts over the past decade to investigate the contribution of small-effect variants that are common in the general population to many traits including glioma through the genome-wide association study (GWAS). The combination of technological advancements and collaborative efforts in establishment of consortia such as the Glioma International Case–Control (GICC) study (22) has enabled genotyping of hundreds of thousands of variants in thousands of glioma cases and controls. It is now recognized that a substantial component of glioma genetic risk is explained by combinations of common polymorphisms of modest effect (23), with 27 loci in total identified so far from glioma GWAS (24–33).

This review provides a brief overview of discoveries from glioma GWAS, as well as a short discussion of future perspectives.

The first glioma GWAS was carried out in 2009 by Shete and colleagues (30), comprising a discovery case–control series of UK and European-American individuals (totaling 1,878 cases and 3,670 controls) and replication series of French, German, and Swedish individuals (totaling 2,545 cases and 2,953 controls). This study identified five susceptibility loci at 5p15.33, 8q24.21, 9p21.3, 11q23.3, and 20q13.33 (30). The loci at 9p21.3 and 20q13.33 were independently confirmed by Wrensch and colleagues (26) in a contemporaneous study of European-American individuals comprising a discovery phase of 692 high-grade glioma cases and 3,992 controls as well as a replication phase of 176 high-grade glioma cases and 174 controls (26). In this study, the top SNP rs1412829 at 9p21.3 is in linkage disequilibrium (LD) with the SNP rs4977756 reported by Shete and colleagues (ref. 30; r2 = 0.75, D′ = 0.89). At 20q13.33, the two reported SNPs rs6010620 and rs4809324 were reported to represent independent association signals (r2 = 0.028, D′ = 1.0).

A further GWAS by Sanson and colleagues (31) was carried out in 2011, making use of data from the United Kingdom and European-American studies previously reported by Shete and colleagues (30) as well as two additional case–control series from France and Germany (totaling 4,147 cases and 7,435 controls). This study led to the identification of 7p11.2 as a susceptibility locus for glioma, which contained two statistically independent SNP associations with glioma risk (31). More recently, a GWAS was carried out in 2014 by Walsh and colleagues (29) comprising a United Kingdom and European-American discovery series of 1,013 high-grade glioma cases and 6,595 controls (in part overlapping with the study of Wrensch and colleagues; ref. 26), as well as a European-American replication series of 631 GBM cases and 1,141 controls. This study reported a novel glioma risk locus at 3q26.2 (near TERC) (29).

In addition to this, a sequence-based association study in the Icelandic population led to the discovery of rs78378222 at 17p13.1 (TP53) as a risk variant for several cancers including glioma (27). The association with glioma was confirmed in an independent European study (32). To refine the association signal at 8q24.21 in glioma, the region was fine-mapped by sequencing as well as statistical imputation of preexisting GWAS datasets. This led to the identification of rs55705857 as being responsible for the 8q24.21 glioma association, with the SNP exhibiting a much larger effect size than the initial GWAS tagSNPs and being highly restricted to low-grade IDH-mutated glioma (28, 34). Following on from this, in 2015, a GWAS was carried out by Kinnersley and colleagues (25), making use of datasets from Sanson and colleagues (31) as well as an independent replication series, in total comprising 5,637 cases and 9,158 controls. This study identified five new glioma risk loci, one for GBM at 12q23.33 (POLR3B), and four for non-GBM gliomas at 10q25.2 (VTI1A), 11q23.2 (ZBTB16), 12q21.2, and 15q24.2 (ETFA).

The largest and most recent glioma GWAS was carried out in 2017 by Melin and colleagues (24). This featured a meta-analysis of six existing GWAS and two new GWAS making use of the Illumina OncoArray (35), in total comprising 12,496 cases and 18,190 controls. The study identified 13 new glioma risk loci, five for GBM at 1p31.3 (JAK1), 11q14.1, 16p13.3 (near MPG), 16q12.1 (HEATR3) and 22q13.1 (SLC16A8), and 8 for non-GBM glioma at 1q32.1 (MDM4), 1q44 (AKT3), 2q33.3 (near IDH1), 3p14.1 (LRIG1), 10q24.33 (OBFC1), 11q21 (MAML2), 14q12 (AKAP6), and 16p13.3 (LMF1). At previously identified loci, with the exception of associations at 3q26.2 and 12q23.33 all associations retained genome-wide significance. Many of the loci demonstrated specificity for GBM or non-GBM glioma (Fig. 1).

Figure 1.

Relative impact of SNP associations at glioma-risk loci for GBM and non-GBM tumors. Glioma risk estimates are from Melin and colleagues, 2017 (24). ORs were derived with respect to the risk allele.

Figure 1.

Relative impact of SNP associations at glioma-risk loci for GBM and non-GBM tumors. Glioma risk estimates are from Melin and colleagues, 2017 (24). ORs were derived with respect to the risk allele.

Close modal

A brief overview of the glioma susceptibility loci identified by GWAS as well as the datasets used is provided in Table 1.

Table 1.

Glioma susceptibility loci identified by GWAS in European populations

Glioma association studyReported glioma-risk variants
ReferenceStudySample size(cases/controls)Glioma subtypeLocusGeneSNPAllelesPOR (95% CI)
Shete et al., 2009 (30) UK-GWAS 631/1,434 All 5p15.33 TERT rs2736100 T/G 1.50 × 10−17 1.27 (1.19–1.37) 
 MDA-GWAS 1,247/2,236 All 8q24.21 CCDC26 rs4295627 G/T 2.34 × 10−18 1.36 (1.29–1.43) 
 French-replication 1,392/1,602 All 8q24.21 CCDC26 rs891835 G/T 7.54 × 10−11 1.24 (1.17–1.30) 
 German-replication 504/573 All 9p21.3 CDKN2A/B rs4977756 A/G 7.24 × 10−15 1.24 (1.19–1.30) 
 Swedish-replication 649/778 All 11q23.3 PHLDB1 rs498872 C/T 1.07 × 10−8 1.18 (1.13–1.24) 
 Total 4,423/6,623 All 20q13.33 RTEL1 rs6010620 G/A 2.52 × 10−12 1.28 (1.21–1.35) 
Wrensch et al., 2009 (26) SFAGS 692/3,992 High-grade 9p21.3 CDKN2A/B rs1412829 C/T 1.85 × 10−10 1.42 (1.27–1.58) 
 Mayo-replication 176/174 High-grade 20q13.33 RTEL1 rs6010620 G/A 3.40 × 10−9 1.52 (1.32–1.75) 
 Total 868/4,166 High-grade 20q13.33 RTEL1 rs4809324 C/T 1.70 × 10−9 1.60 (1.37–1.87) 
Sanson et al., 2011 (31) UK-GWAS 631/2,699 All 7p11.2 EGFR rs11979158 A/G 7.72 × 10−8 1.23 (1.15–1.35) 
 French-GWAS 1,423/1,190 All 7p11.2 EGFR rs2252586 T/C 2.09 × 10−8 1.18 (1.11–1.25) 
 German-GWAS 846/1,310        
 MDA-GWAS 1,247/2,236        
 Total 4,147/7,435        
Stacey et al., 2011 (27) Iceland imputation 135/>37,881 All 17p13.1 TP53 rs78378222 T/G 1.0 × 10−5 2.35 (1.61–3.44) 
 Iceland genotyped 72/7,200        
 USA UCSF 658/573        
 USA Mayo Clinic 530/283        
 Total 1,395/>45,937        
Rajaraman et al., 2012 (33) GliomaScan 1,856/4,955        
Jenkins et al., 2012 (28) Mayo Clinic 860/795 Oligodendroglioma 8q24.21 CCDC26 rs55705857 A/G 2.2 × 10−28 6.3 (4.6–8.8) 
 UCSF AGS 953/1,079        
 Total 1,813/1,874        
Walsh et al., 2014 (29) SFAGS 692/3,992 High-grade 3q26.2 Near TERC rs1920116 G/A 8.3 × 10−9 1.30 (1.19–1.42) 
 TCGA cases, WTCCC controls 323/2,603        
 Mayo-UCSF replication 631/1,141        
 Total 1,646/7,736        
Kinnersley et al., 2015 (25) UK-GWAS 631/2,699 Non-GBM 10q25.2 VTI1A rs11196067 A/T 4.32 × 10−8 1.19 (1.12–1.27) 
 French-GWAS 1,423/1,190 Non-GBM 11q23.2 ZBTB16 rs648044 C/T 6.26 × 10−11 1.25 (1.17–1.34) 
 German-GWAS 846/1,310 Non-GBM 12q21.2 Intergenic rs12230172 G/A 7.53 × 10−11 1.23 (1.16–1.32) 
 MDA-GWAS 1,247/2,236 GBM 12q23.3 POLR3B rs3851634 T/C 3.02 × 10−9 1.23 (1.15–1.32) 
 UK Replication 1,490/1,723 Non-GBM 15q24.2 ETFA rs1801591 G/A 5.71 × 10−9 1.36 (1.23–1.51) 
 Total 5,637/9,158        
Melin et al., 2017 (24) UK-GWAS 631/2,699 New loci       
 French-GWAS 1,423/1,190 GBM 1p31.3 JAK1 rs12752552 T/C 2.04 × 10−9 1.22 (1.15–1.31) 
 German-GWAS 846/1,310 Non-GBM 1q32.1 MDM4 rs4252707 G/A 3.34 × 10−9 1.19 (1.12–1.26) 
 MDA-GWAS 1,175/2,236 Non-GBM 1q44 AKT3 rs12076373 G/C 2.63 × 10−10 1.23 (1.16–1.32) 
 SFAGS 677/3,940 Non-GBM 2q33.3 Near IDH1 rs7572263 A/G 2.18 × 10−10 1.20 (1.13–1.26) 
 GliomaScan 1,653/2,725 Non-GBM 3p14.1 LRIG1 rs11706832 A/C 7.66 × 10−9 1.15 (1.09–1.20) 
 GICC 4,572/3,286 Non-GBM 10q24.33 OBFC1 rs11598018 C/A 3.39 × 10−8 1.14 (1.09–1.20) 
 UCSF/Mayo 1,519/804 GBM 11q14.1 Intergenic rs11233250 C/T 9.95 × 10−10 1.24 (1.16–1.33) 
 Total 12,496/18,190 Non-GBM 11q21 MAML2 rs7107785 T/C 3.87 × 10−10 1.16 (1.11–1.21) 
   Non-GBM 14q12 AKAP6 rs10131032 G/A 5.07 × 10−11 1.33 (1.22–1.44) 
   GBM 16p13.3 Near MPG rs2562152 A/T 1.93 × 10−8 1.21 (1.13–1.29) 
   Non-GBM 16p13.3 LMF1 rs3751667 C/T 2.61 × 10−9 1.18 (1.12–1.25) 
   GBM 16q12.1 HEATR3 rs10852606 T/C 1.29 × 10−11 1.18 (1.13–1.24) 
   GBM 22q13.1 SLC16A8 rs2235573 G/A 1.76 × 10−10 1.15 (1.10–1.20) 
   Existing loci       
   All glioma 3q26.2 Near TERC rs3772190 G/A 2.25 × 10−6 1.11 (1.06–1.15) 
   GBM 5p15.33 TERT rs10069690 C/T 8.33 × 10−74 1.61 (1.53–1.69) 
   GBM 7p11.2 EGFR rs75061358 T/G 4.94 × 10−34 1.63 (1.50–1.76) 
   GBM 7p11.2 EGFR rs723527 A/G 4.79 × 10−23 1.25 (1.20–1.31) 
   Non-GBM 8q24.21 CCDC26 rs55705857 A/G 7.28 × 10−149 3.39 (3.09–3.71) 
   GBM 9p21.3 CDKN2A/B rs634537 T/G 7.23 × 10−45 1.37 (1.31–1.43) 
   Non-GBM 10q25.2 VTI1A rs11599775 G/A 3.44 × 10−9 1.16 (1.10–1.22) 
   Non-GBM 11q23.2 ZBTB16 rs648044 A/G 4.66 × 10−12 1.19 (1.13–1.25) 
   Non-GBM 11q23.3 PHLDB1 rs12803321 G/C 6.33 × 10−43 1.42 (1.35–1.49) 
   Non-GBM 12q21.2 Intergenic rs1275600 T/A 3.72 × 10−9 1.16 (1.10–1.21) 
   GBM 12q23.33 RFX4 rs12227783 A/T 1.60 × 10−5 1.16 (1.08–1.24) 
   Non-GBM 15q24.2 ETFA rs77633900 G/C 1.60 × 10−13 1.35 (1.25–1.46) 
   All glioma 17p13.1 TP53 rs78378222 T/G 8.64 × 10−38 2.53 (2.19–2.91) 
   GBM 20q13.3 RTEL1 rs2297440 T/C 3.66 × 10−46 1.48 (1.40–1.56) 
Glioma association studyReported glioma-risk variants
ReferenceStudySample size(cases/controls)Glioma subtypeLocusGeneSNPAllelesPOR (95% CI)
Shete et al., 2009 (30) UK-GWAS 631/1,434 All 5p15.33 TERT rs2736100 T/G 1.50 × 10−17 1.27 (1.19–1.37) 
 MDA-GWAS 1,247/2,236 All 8q24.21 CCDC26 rs4295627 G/T 2.34 × 10−18 1.36 (1.29–1.43) 
 French-replication 1,392/1,602 All 8q24.21 CCDC26 rs891835 G/T 7.54 × 10−11 1.24 (1.17–1.30) 
 German-replication 504/573 All 9p21.3 CDKN2A/B rs4977756 A/G 7.24 × 10−15 1.24 (1.19–1.30) 
 Swedish-replication 649/778 All 11q23.3 PHLDB1 rs498872 C/T 1.07 × 10−8 1.18 (1.13–1.24) 
 Total 4,423/6,623 All 20q13.33 RTEL1 rs6010620 G/A 2.52 × 10−12 1.28 (1.21–1.35) 
Wrensch et al., 2009 (26) SFAGS 692/3,992 High-grade 9p21.3 CDKN2A/B rs1412829 C/T 1.85 × 10−10 1.42 (1.27–1.58) 
 Mayo-replication 176/174 High-grade 20q13.33 RTEL1 rs6010620 G/A 3.40 × 10−9 1.52 (1.32–1.75) 
 Total 868/4,166 High-grade 20q13.33 RTEL1 rs4809324 C/T 1.70 × 10−9 1.60 (1.37–1.87) 
Sanson et al., 2011 (31) UK-GWAS 631/2,699 All 7p11.2 EGFR rs11979158 A/G 7.72 × 10−8 1.23 (1.15–1.35) 
 French-GWAS 1,423/1,190 All 7p11.2 EGFR rs2252586 T/C 2.09 × 10−8 1.18 (1.11–1.25) 
 German-GWAS 846/1,310        
 MDA-GWAS 1,247/2,236        
 Total 4,147/7,435        
Stacey et al., 2011 (27) Iceland imputation 135/>37,881 All 17p13.1 TP53 rs78378222 T/G 1.0 × 10−5 2.35 (1.61–3.44) 
 Iceland genotyped 72/7,200        
 USA UCSF 658/573        
 USA Mayo Clinic 530/283        
 Total 1,395/>45,937        
Rajaraman et al., 2012 (33) GliomaScan 1,856/4,955        
Jenkins et al., 2012 (28) Mayo Clinic 860/795 Oligodendroglioma 8q24.21 CCDC26 rs55705857 A/G 2.2 × 10−28 6.3 (4.6–8.8) 
 UCSF AGS 953/1,079        
 Total 1,813/1,874        
Walsh et al., 2014 (29) SFAGS 692/3,992 High-grade 3q26.2 Near TERC rs1920116 G/A 8.3 × 10−9 1.30 (1.19–1.42) 
 TCGA cases, WTCCC controls 323/2,603        
 Mayo-UCSF replication 631/1,141        
 Total 1,646/7,736        
Kinnersley et al., 2015 (25) UK-GWAS 631/2,699 Non-GBM 10q25.2 VTI1A rs11196067 A/T 4.32 × 10−8 1.19 (1.12–1.27) 
 French-GWAS 1,423/1,190 Non-GBM 11q23.2 ZBTB16 rs648044 C/T 6.26 × 10−11 1.25 (1.17–1.34) 
 German-GWAS 846/1,310 Non-GBM 12q21.2 Intergenic rs12230172 G/A 7.53 × 10−11 1.23 (1.16–1.32) 
 MDA-GWAS 1,247/2,236 GBM 12q23.3 POLR3B rs3851634 T/C 3.02 × 10−9 1.23 (1.15–1.32) 
 UK Replication 1,490/1,723 Non-GBM 15q24.2 ETFA rs1801591 G/A 5.71 × 10−9 1.36 (1.23–1.51) 
 Total 5,637/9,158        
Melin et al., 2017 (24) UK-GWAS 631/2,699 New loci       
 French-GWAS 1,423/1,190 GBM 1p31.3 JAK1 rs12752552 T/C 2.04 × 10−9 1.22 (1.15–1.31) 
 German-GWAS 846/1,310 Non-GBM 1q32.1 MDM4 rs4252707 G/A 3.34 × 10−9 1.19 (1.12–1.26) 
 MDA-GWAS 1,175/2,236 Non-GBM 1q44 AKT3 rs12076373 G/C 2.63 × 10−10 1.23 (1.16–1.32) 
 SFAGS 677/3,940 Non-GBM 2q33.3 Near IDH1 rs7572263 A/G 2.18 × 10−10 1.20 (1.13–1.26) 
 GliomaScan 1,653/2,725 Non-GBM 3p14.1 LRIG1 rs11706832 A/C 7.66 × 10−9 1.15 (1.09–1.20) 
 GICC 4,572/3,286 Non-GBM 10q24.33 OBFC1 rs11598018 C/A 3.39 × 10−8 1.14 (1.09–1.20) 
 UCSF/Mayo 1,519/804 GBM 11q14.1 Intergenic rs11233250 C/T 9.95 × 10−10 1.24 (1.16–1.33) 
 Total 12,496/18,190 Non-GBM 11q21 MAML2 rs7107785 T/C 3.87 × 10−10 1.16 (1.11–1.21) 
   Non-GBM 14q12 AKAP6 rs10131032 G/A 5.07 × 10−11 1.33 (1.22–1.44) 
   GBM 16p13.3 Near MPG rs2562152 A/T 1.93 × 10−8 1.21 (1.13–1.29) 
   Non-GBM 16p13.3 LMF1 rs3751667 C/T 2.61 × 10−9 1.18 (1.12–1.25) 
   GBM 16q12.1 HEATR3 rs10852606 T/C 1.29 × 10−11 1.18 (1.13–1.24) 
   GBM 22q13.1 SLC16A8 rs2235573 G/A 1.76 × 10−10 1.15 (1.10–1.20) 
   Existing loci       
   All glioma 3q26.2 Near TERC rs3772190 G/A 2.25 × 10−6 1.11 (1.06–1.15) 
   GBM 5p15.33 TERT rs10069690 C/T 8.33 × 10−74 1.61 (1.53–1.69) 
   GBM 7p11.2 EGFR rs75061358 T/G 4.94 × 10−34 1.63 (1.50–1.76) 
   GBM 7p11.2 EGFR rs723527 A/G 4.79 × 10−23 1.25 (1.20–1.31) 
   Non-GBM 8q24.21 CCDC26 rs55705857 A/G 7.28 × 10−149 3.39 (3.09–3.71) 
   GBM 9p21.3 CDKN2A/B rs634537 T/G 7.23 × 10−45 1.37 (1.31–1.43) 
   Non-GBM 10q25.2 VTI1A rs11599775 G/A 3.44 × 10−9 1.16 (1.10–1.22) 
   Non-GBM 11q23.2 ZBTB16 rs648044 A/G 4.66 × 10−12 1.19 (1.13–1.25) 
   Non-GBM 11q23.3 PHLDB1 rs12803321 G/C 6.33 × 10−43 1.42 (1.35–1.49) 
   Non-GBM 12q21.2 Intergenic rs1275600 T/A 3.72 × 10−9 1.16 (1.10–1.21) 
   GBM 12q23.33 RFX4 rs12227783 A/T 1.60 × 10−5 1.16 (1.08–1.24) 
   Non-GBM 15q24.2 ETFA rs77633900 G/C 1.60 × 10−13 1.35 (1.25–1.46) 
   All glioma 17p13.1 TP53 rs78378222 T/G 8.64 × 10−38 2.53 (2.19–2.91) 
   GBM 20q13.3 RTEL1 rs2297440 T/C 3.66 × 10−46 1.48 (1.40–1.56) 

NOTE: ORs are reported with respect to the risk allele, highlighted in bold.

Details of each glioma risk locus so far identified are presented in the following sections.

1p31.3

At the GBM locus 1p31.3, the risk allele of rs12752552 (T) is associated with increased expression of JAK1 in brain cortex and cerebellar hemisphere (24). JAK1 is involved in actomyosin contractility in tumor cells and stroma (36), and dysregulation of JAK–STAT signaling is thought to play an important role in development of high-grade glioma (37), providing an attractive mechanistic basis for the 1p31.3 association.

1q32.1

The non-GBM risk SNP rs4252707 is within the eighth intron of MDM4, with the risk allele (A) associated with increased expression of this gene (24). MDM4 is amplified in glioma tumors lacking TP53 mutation, consistent with its role in inactivation of p53-dependent growth control (38). Another SNP in the region, rs4245739, has been reported to be associated with ER-negative breast (39) and prostate cancer (40), and it is unclear if the association signals are fully independent (r2 = 0.10, D′ = 1).

1q44

At 1q44, rs12076373 maps to the eighth intron of AKT3 (v-akt murine thymoma viral oncogene homolog 3) one of the major downstream effectors of PI3K that is highly expressed during active neurogenesis (41). AKT3 is highly expressed in glioma and plays an important role in tumor viability by activating DNA repair (42).

2q33.3

The non-GBM risk SNP at 2q33.3 (rs7572263) is 50-kb telomeric to IDH1 (isocitrate dehydrogenase 1). Mutation of IDH1 is a driver for gliomagenesis (43, 44) and is responsible for the CpG island methylator (G-CIMP) phenotype (45, 46). IDH mutation predominates in non-GBM glioma (47, 48); therefore, dysregulation of IDH1 is highly plausible as a basis for susceptibility of these tumors.

3p14.1

The 3p14.1 association marked by rs11706832 localizes to intron 2 of LRIG1 (leucine-rich repeats- and immunoglobulin-like domains-containing protein 1). LRIG1 is highly expressed in the brain and negatively regulates the EGFR signaling pathway (49). Reduced LRIG1 expression correlates with tumor aggressiveness as well as resistance to temozolomide and radiotherapy (50, 51). An additional association at 3p14.1 is with rs812481 and colorectal cancer risk (52), as this SNP is correlated with rs11706832 the association signals appear to overlap (r2 = 0.67, D′ = 0.83).

3q26.2

The GWAS tagSNP rs1920116 is in strong LD with variants at the nearby gene TERC (telomerase RNA component), leading Walsh and colleagues to suggest that this could be a plausible basis for the high-grade glioma association (29). The risk allele of rs1920116 was found to be positively associated with leukocyte telomere length (LTL, P = 5.5 × 10−20), consistent with the functional basis of the 3q26.2 association involving TERC (29, 53). The strongest association with telomere length at 3q26.2 was exhibited by rs10936599, which is also a known colorectal cancer risk locus (53, 54); there is moderate correlation between rs10936599 and rs1920116 (r2 = 0.44, D′ = 0.72) suggesting these are not independent signals. In the latest glioma GWAS, the 3q26.2 locus does not attain genome-wide significance (24).

5p15.33

The original glioma tagSNP rs2736100 is intronic within TERT, the gene coding for telomerase reverse transcriptase that is primarily involved in telomere length regulation (55). Consistent with a TERT-mediated role in telomere length maintenance, the risk allele of rs2736100 was found to be positively associated with LTL (P = 4.4 × 10−19; refs. 29, 53). 5p15.33 is a pleiotropic cancer risk locus harboring associations with multiple cancers including breast cancer, prostate cancer, and testicular germ cell tumor (56–59). In the latest glioma meta-analysis, the strongest association at 5p15.33 is with rs10069690 and GBM. Intriguingly, the rs10069690 risk allele (T) has been shown to create a splice-donor site leading to an alternate TERT splice isoform lacking telomerase activity (60), providing direct evidence for its functional effects on tumor development.

7p11.2

The two statistically independent glioma-risk SNPs identified (rs11979158 and rs2252586) lie just outside EGFR, a receptor tyrosine kinase (RTK) commonly activated in glioma (61). Association between these SNPs and gliomas has been reported to be independent of EGFR amplification, p16 deletion, and IDH1 mutation status (31). The authors note that the absence of a particular subtype association is consistent with EGFR activation playing a role in both initiation of primary GBM and progression from lower-grade glioma to secondary GBM (61). While the latest glioma meta-analysis has identified more strongly associated SNPs (rs75061358 for rs2252586 and rs723527 for rs11979158), the fine-mapped locus is still consistent with two independent signals.

8q24.21

After identification through fine-mapping rs55705857 represents the likely causal variant at the 8q24.21 glioma risk locus (28, 34). Jenkins and colleagues (28) and Enciso-Mora and colleagues (34) suggest that the association confined to IDH-mutated gliomas, in particular those with 1p/19q co-deletion (i.e., classical oligodendroglioma). rs55705857 is located within CCDC26, a long intergenic noncoding RNA (lincRNA), which has been shown to be a retinoic acid–dependent modulator of myeloid cell differentiation and death (62). The SNP additionally resides within a conserved unmethylated CpG cluster. These motifs are sufficient for recruitment of repressive polycomb complex components that are implicated in control of cell fate, development, and cancer through gene silencing (63–65). 8q24.21 is a pleiotropic cancer risk locus with different cancers mapping to distinct subregions within the locus (66–69). The locus contains the oncogene Myc, regulation of which provides a plausible basis of predisposition at 8q24.21.

9p21.3

rs497756 lies within the intron of CDKN2B-AS1 (ANRIL), a long noncoding RNA involved in transcriptional silencing of CDKN2A (70, 71), providing an attractive functional basis for the 9p21.3 glioma association. Consistent with the known importance of mutations at this locus in melanoma tumors, rs7023329 at 9p21.3 has been identified as a susceptibility locus for malignant melanoma (72). rs7023329 and rs497756 are not at all correlated, suggesting these associations represent distinct risk loci within 9p21.3. p16/CDKN2A is a tumor suppressor gene commonly deleted in a number of cancer types including glioma (73, 74). Interestingly, allelic loss of 9p21.3 has been recently shown to be present in 41.7% of anaplastic 1p/19q co-deleted oligodendrogliomas, with these tumors being associated with shorter overall survival (75).

10q24.33

The 10q24.33 association marked by rs11598018 lies intronic to OBFC1 (oligonucleotide/oligosaccharide-binding fold-containing protein 1), which functions in a telomere-associated CST complex that competes with shelterin to regulate telomere length (76, 77). The risk allele of rs11598018 (C) is associated with increased leukocyte telomere length thereby supporting a relationship between genotype and biology (78–80). Variation at 10q24.33 is additionally associated with risk of melanoma (81), basal cell carcinoma (82), thyroid cancer (83), and renal cell carcinoma (84). The SNPs defined by these associations do not appear to be correlated with rs11598018 (r2 = 0.11–0.12, D′ = 0.72–1.0).

10q25.2

At 10q25.2, the originally reported SNP rs1196067 as well as the most associated SNP in the latest GWAS are intronic within VTI1A. As well as participating in regulating insulin-stimulated trafficking of secretory vesicles (85), VTI1A plays a key role in neuronal development and in selectively maintaining spontaneous neurotransmitter release (86). While the glioma-risk signal appears to be within VTI1A, the possibility of other nearby genes such as TCF7L2 playing a role cannot be excluded. At 10q25.2, there are additional associations with colorectal (87), lung (88), and breast cancer (89). These appear to be uncorrelated, perhaps indicative of tissue-specific effects.

11q14.1

The GBM SNP rs11233250 was discovered in the latest glioma GWAS and lies in an intergenic region, with no obvious mechanistic candidates nearby.

11q21

At 11q21, the non-GBM SNP rs7107785 lies within MAML2, encoding Mastermind-like protein 2. The mastermind-like protein family has been reported to function as coactivators of Notch signaling (90).

11q23.2

The risk SNP rs648044 is specific to non-GBM glioma and is intronic within ZBTB16. ZBTB16 is highly expressed in undifferentiated, multipotential progenitor cells and its expression has been shown to influence resistance to retinoid-mediated redifferentiation in t(11;17)(q23;21) acute promyelocytic leukaemia (APL; ref. 91). The BTB domain of ZBTB16 has transcriptional repression activity and interacts with components of the histone deacetylase complex thereby linking the transcription factor with regulation of chromatin conformation (92). Additional associations have been reported at 11q23.2 with risk of prostate cancer (93) and chronic lymphocytic leukemia (CLL; ref. 94); however, they appear to represent distinctive association signals.

11q23.3

Both the original tag-SNP rs498872 and most associated SNP from the latest glioma GWAS rs12803321 are intronic within PHLDB1. rs498872 has been shown to exhibit an association with IDH-mutated glioma but not IDH-wild-type glioma that is independent of histology or grade (95). Pleckstrin homology-like domain family B member 1 (PHLDB1) is an insulin-responsive protein that enhances Akt activation (96). Insulin signaling and the Akt pathway often play an important role in cancer progression (97) providing a rationale for the functional basis of the 11q23.3 association; however, the explanation for the specificity of association with IDH-mutated glioma remains to be established. Additional signals at 11q23.3 with other cancers include follicular lymphoma (ref. 98; rs4938573) and lung adenocarcinoma (ref. 99; rs1056562), which appear to represent distinct signals (LD metrics with respect to rs12803321: r2 = 0.04/0.03, D′ = 0.55/0.24, respectively).

12q21.2

The non-GBM glioma association signal at 12q21.2 appears to map within RP11-114H23.1, a lincRNA of currently unknown function. Although only lying adjacent to PHLDA1, the known 11q23.3 association maps to the related gene PHLDB1 that is also specific to non-GBM tumors (30). Although a role for PHLDA1 in glioma has yet to be established downregulation of PHLDA1 in neuronal cells has been shown to enhance cell death without Fas induction (100). In addition, PHLDA1 expression may be involved in regulation of antiapoptotic effects of IGF1 (101).

12q23.3

The original published GBM SNP at 12q23.3 was rs3851634, which is intronic within POLR3B (25). RNA polymerase III (POLR3B) is involved in the transcription of small noncoding RNAs and short interspersed nuclear elements, as well as all transfer RNAs (102). Although mutations in POLR3B have been shown to cause recessive hypomyelinating leukoencephalopathy (103) thus far there is no evidence implicating the gene in the development of glioma. No variant at 12q23.3 exhibited genome-wide significance in the latest glioma GWAS, and therefore its status as a glioma-risk locus is uncertain.

14q12

At 14q12, the non-GBM glioma risk SNP rs10131032 resides within AKAP6. Protein Kinase A–Anchoring Protein 6 targets protein kinase A to the sarcoplasmic reticulum (104) and is commonly deleted in GBM tumors (105).

15q24.2

The non-GBM association signal at 15q24.2 resides within ETFA, which participates in mitochondrial fatty acid beta oxidation; shuttling electrons between flavoprotein dehydrogenases and the membrane-bound electron transfer flavoprotein ubiquinone oxidoreductase (106). Mutations of ETFA have been reported to be a cause of recessive Glutaric acidemia IIA (107, 108) that features gliosis.

16p13.3

This locus appears to harbor two independent association signals: one for GBM defined by rs2562152 and one for non-GBM defined by rs3751667. In contrast with 7p11.2, where both signals appear to be involved in EGFR regulation, the signals at 16p13.3 may have distinct molecular mechanisms. The non-GBM SNP rs3751667 is intronic within LMF1, whereas the GBM SNP rs2562152 maps 3-kb telomeric to MPG that encodes a N-methylpurine DNA glycosylase whose expression is linked to temozolomide resistance in glioma (109).

16q12.1

At 16q12.1, the C-risk allele of rs10852606 is associated with reduced HEATR3 expression in brain tissues, consistent with differential expression of HEATR3 being the functional basis of the 16q12.1 association. Variation at 16q12.1 is associated with risk of testicular (ref. 110; rs8046148) and esophageal (ref. 111; rs4785204) cancer (pairwise r2 and D' with rs10852606, 0.67, 1.0, and 0.16, 1.0, respectively) suggestive of pleiotropic effects on tumor risk.

17p13.1

A GWAS for cutaneous basal cell carcinoma in Icelandic individuals identified rs78378222 (P = 5.2 × 10−17, OR = 2.36) as being associated with disease risk (27). rs7837822 lies within the 3′-untranslated region (3′-UTR) of TP53, a tumor suppressor gene highly mutated in many cancer types including glioma (112). The authors additionally showed that the variant affected the polyadenylation signal of TP53, impairing processing of TP53 mRNA (27). This is highly suggestive of rs78378222 representing the functional variant at 17p13.1 through disrupting the normal functioning of TP53. The study additionally demonstrated a relationship between rs78378222 and risk of prostate cancer (OR = 1.44, P = 2.4 × 10−6), glioma (OR = 2.35, P = 1.0 × 10−5), colorectal adenoma (OR = 1.39, P = 1.6 × 10−4), but not breast cancer (OR = 1.06, P = 0.57; ref. 27). A number of additional studies have replicated the association between rs78378222 and glioma risk (20, 32, 113, 114).

20q13.33

Both the original tag-SNP rs6010620 and top SNP in the most recent GWAS rs2297440 are intronic within RTEL1 (regulator of telomere elongation helicase 1), encoding a DNA helicase involved in telomere and genome-wide replication, with mutations leading to genetic instability (115). A negative, albeit modest association has been demonstrated between the risk allele of rs6010620 and LTL (P = 1.1 × 10−3; refs. 29, 53). However, the glioma risk and telomere length association signals at 20q13.33 do not fully overlap, raising the possibility of extra-telomeric functions at this glioma locus. Variation at 20q13.33 has additionally been implicated in prostate cancer (ref. 40; rs6062509), lung adenocarcinoma (ref. 99; rs41309931), and colorectal cancer (ref. 54; rs4925386). The prostate cancer SNP rs6062509 appears correlated with rs2297440 (r2 = 0.466, D′ = 0.838), indicative of a shared association signal, whereas the other associations appear to be independent.

22q13.1

The GBM association at 22q13.1 marked by rs2235573 is intronic within SLC16A8. Variation at 22q13.1 is additionally associated with melanoma (ref. 72; rs2284063), breast cancer (ref. 116; rs738321), bladder cancer (ref. 117; rs1014971), and multiple myeloma (ref. 118; rs139371), which appear to represent independent signals.

The glioma GWAS risk loci so far provide support for a polygenic model of disease susceptibility, with currently identified risk SNPs for glioma account for at best around 27% and 37% of the familial risk of GBM and non-GBM tumors, respectively (24). Aside from the fine-mapped associations at 8q24.21 and 17p13.1, the glioma GWAS SNPs identified are relatively common (European MAF > ∼9%) and have modest effect sizes (1.11 < OR < 1.63). The loci implicate genes and pathways known to be important in glioma and cancer biology; for example, IDH1 at 2q33.3, EGFR–AKT signaling (EGFR at 7p11.2, LRIG1 at 3p14.1, JAK1 at 1q32.1, PHLDB1 at 11q23.3), CDKN2A/B at 9p21.3, Myc at 8q24.21, TP53 at 17p13.1. In addition, through identification of risk loci at TERC (3q26.2), TERT (5p15.33), OBFC1 (10q24.33), and RTEL1 (20q13.33) GWAS associations reveal telomere maintenance as an important feature in glioma progression.

Subtype specificities

While pragmatically within large case–control consortia glioma cases were divided into GBM and non-GBM classifications, it is becoming increasingly clear that these tumors comprise a number of distinctive molecular subtypes, principally involving various combinations of IDH mutation, 1p/19q co-deletion, and TERT promoter mutation (48, 119). A number of loci have been assessed for association with molecular features. Notably, rs55705857 at 8q24.21 is highly associated with IDH-mutated gliomas, in particular, those with 1p/19q co-deletion (i.e., classical oligodendroglioma; refs. 28, 34). In contrast, the 5p15.33, 9p21.3, and 20q13.3 loci are associated with IDH wild-type gliomas (120), and rs7837822 at 17p13.1 is reported to be associated with gliomas with TERT promoter mutation and IDH mutation without 1p/19q co-deletion (48). The majority of loci, particularly those discovered more recently, await molecular characterization.

Glioma risk loci in non-European populations

Glioma incidence varies across population group (121). While the majority of susceptibility studies have been carried out in the northern European population, a number have been carried out in different population groups. In a study of 976 glioma cases and 1,057 controls, Chen and colleagues (122) investigated previously identified risk SNPs in the Chinese population, where generally incidence is lower than in Europe. They reported significant associations at 5p15.33 [rs2736100, P = 3.69 × 10−4, OR = 1.26 (1.11–1.43), 11q23.3 (rs498872, P = 3.80 × 10−6, OR = 1.38 (1.20–1.57) and 20q13.3 (rs6010620, P = 2.79 × 10−6, OR = 1.39 (1.20–1.59)], but not at 8q24.21 or 9p21.3. An additional study of 980 cases and 990 controls reported an association between rs1920116 at 3q26.2 and glioma in the Chinese population (123). Aside from these studies there is a paucity of data currently available for glioma genetic risk in non-European populations.

Investigating nongenetic glioma risk factors

Glioma GWAS data can additionally be used in investigation of proposed risk factors not necessarily thought to have a genetic component. For example, risk SNPs at 5p15.33, 8q24.21, 9p21.3, 11q23.3, and 20q13.3 were not shown to be associated with allergy (124). In addition, there is limited evidence for variants implicated in Vitamin D metabolism being associated with glioma (125). While Kitahara and colleagues (126) report association between diabetes and reduced risk of glioma, there was no association in glioma with diabetes-related SNPs. Investigating the relationship between risk factors such as these and glioma can be carried out in a relatively unbiased fashion using a Mendelian Randomization approach (127).

Functional investigation of mechanistic effect at glioma-risk loci

For the vast majority of glioma risk loci identified so far, the molecular mechanisms underlining the associations have not yet been deciphered. Notable exceptions are the 17p13.1 locus, where rs78378222 has been demonstrated to disrupt TP53 polyadenylation (27) and 5p15.33, where the risk SNP rs10069690 is reported to create a splice-donor site leading to an alternate TERT splice isoform lacking telomerase activity (60). As many of the risk SNPs are noncoding an attractive mechanism of action is through regulating expression of nearby genes. Regulatory targets at risk loci can be explored through Hi-C methodologies as exemplified by Jaeger and colleagues (128) for colorectal cancer and Dryden and colleagues (129) for breast cancer. Of vital importance is the use of appropriate model systems to investigate potential tissue- and developmental stage-specific differences in chromatin architecture, from neuronal progenitor cells (130) through to IDH mutated and wild-type tumors (131).

Exploring the relationship between telomere length and glioma risk

Recent reports assessing associations with glioma at telomere-risk loci (53) suggest that increased telomere length is a risk factor for glioma (132). However, these telomere length estimates were based on data from blood cells, and it is unclear whether this correlates with telomere length in cells in the brain. This can be addressed in the near future by projects such as enhancing GTEx (eGTEx) that aim to investigate genetic influences on telomere length across different tissues (133). In addition, the majority of telomere length loci also exhibit strong associations with glioma (i.e., 3q26.2 – TERC, 5p15.33 – TERT, 10q24.33 – OBFC1, 20q13.33 – RTEL1). This raises the issue of to what extent risk of glioma is being driven by telomere length at these individual loci, or whether there are any extra-telomeric effects.

The past decade has overseen a transformation in our understanding of inherited predisposition to glioma, with the majority of the genetic component explained by a polygenic contribution from at least 25 risk polymorphisms identified through GWAS. It is increasingly recognized that glioma tumors comprise multiple distinctive molecular subtypes, with which germline predisposition variants show particular preferences. Understanding the molecular basis of glioma predisposition loci is still in its relative infancy but is ultimately likely to lead to insight into the etiology of glioma and potentially have therapeutic applications.

No potential conflicts of interest were disclosed.

The work of B. Kinnersley and R.S. Houlston is funded by Cancer Research UK. As part of GICC, M.L. Bondy was funded by the U.S. NIH grants R01CA139020 and R01CA52689.

1.
Crocetti
E
,
Trama
A
,
Stiller
C
,
Caldarella
A
,
Soffietti
R
,
Jaal
J
, et al
Epidemiology of glial and non-glial brain tumours in Europe
.
Eur J Cancer
2012
;
48
:
1532
42
.
2.
Bondy
ML
,
Scheurer
ME
,
Malmer
B
,
Barnholtz-Sloan
JS
,
Davis
FG
,
Il'yasova
D
, et al
Brain tumor epidemiology: consensus from the Brain Tumor Epidemiology Consortium
.
Cancer
2008
;
113
(
7 Suppl
):
1953
68
.
3.
Louis
DN
,
Perry
A
,
Reifenberger
G
,
von Deimling
A
,
Figarella-Branger
D
,
Cavenee
WK
, et al
The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary
.
Acta Neuropathol
2016
;
131
:
803
20
.
4.
Hemminki
K
,
Tretli
S
,
Sundquist
J
,
Johannesen
TB
,
Granstrom
C
. 
Familial risks in nervous-system tumours: a histology-specific analysis from Sweden and Norway
.
Lancet Oncol
2009
;
10
:
481
8
.
5.
Li
FP
,
Fraumeni
JF
 Jr
,
Mulvihill
JJ
,
Blattner
WA
,
Dreyfus
MG
,
Tucker
MA
, et al
A cancer family syndrome in twenty-four kindreds
.
Cancer Res
1988
;
48
:
5358
62
.
6.
Srivastava
S
,
Tong
YA
,
Devadas
K
,
Zou
ZQ
,
Sykes
VW
,
Chen
Y
, et al
Detection of both mutant and wild-type p53 protein in normal skin fibroblasts and demonstration of a shared ‘second hit’ on p53 in diverse tumors from a cancer-prone family with Li-Fraumeni syndrome
.
Oncogene
1992
;
7
:
987
91
.
7.
Iavarone
A
,
Matthay
KK
,
Steinkirchner
TM
,
Israel
MA
. 
Germ-line and somatic p53 gene mutations in multifocal osteogenic sarcoma
.
Proc Natl Acad Sci USA
1992
;
89
:
4207
9
.
8.
Malkin
D
,
Li
FP
,
Strong
LC
,
Fraumeni
JF
 Jr
,
Nelson
CE
,
Kim
DH
, et al
Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms
.
Science
1990
;
250
:
1233
8
.
9.
Malkin
D
,
Jolly
KW
,
Barbier
N
,
Look
AT
,
Friend
SH
,
Gebhardt
MC
, et al
Germline mutations of the p53 tumor-suppressor gene in children and young adults with second malignant neoplasms
.
N Engl J Med
1992
;
326
:
1309
15
.
10.
Sidransky
D
,
Tokino
T
,
Helzlsouer
K
,
Zehnbauer
B
,
Rausch
G
,
Shelton
B
, et al
Inherited p53 gene mutations in breast cancer
.
Cancer Res
1992
;
52
:
2984
6
.
11.
Kyritsis
AP
,
Bondy
ML
,
Xiao
M
,
Berman
EL
,
Cunningham
JE
,
Lee
PS
, et al
Germline p53 gene mutations in subsets of glioma patients
.
J Natl Cancer Inst
1994
;
86
:
344
9
.
12.
Chen
P
,
Iavarone
A
,
Fick
J
,
Edwards
M
,
Prados
M
,
Israel
MA
. 
Constitutional p53 mutations associated with brain tumors in young adults
.
Cancer Genet Cytogenet
1995
;
82
:
106
15
.
13.
Evans
DG
,
Sainio
M
,
Baser
ME
. 
Neurofibromatosis type 2
.
J Med Genet
2000
;
37
:
897
904
.
14.
Gutmann
DH
,
James
CD
,
Poyhonen
M
,
Louis
DN
,
Ferner
R
,
Guha
A
, et al
Molecular analysis of astrocytomas presenting after age 10 in individuals with NF1
.
Neurology
2003
;
61
:
1397
400
.
15.
Poley
JW
,
Wagner
A
,
Hoogmans
MM
,
Menko
FH
,
Tops
C
,
Kros
JM
, et al
Biallelic germline mutations of mismatch-repair genes: a possible cause for multiple pediatric malignancies
.
Cancer
2007
;
109
:
2349
56
.
16.
Hamilton
SR
,
Liu
B
,
Parsons
RE
,
Papadopoulos
N
,
Jen
J
,
Powell
SM
, et al
The molecular basis of Turcot's syndrome
.
N Engl J Med
1995
;
332
:
839
47
.
17.
Randerson-Moor
JA
,
Harland
M
,
Williams
S
,
Cuthbert-Heavens
D
,
Sheridan
E
,
Aveyard
J
, et al
A germline deletion of p14(ARF) but not CDKN2A in a melanoma-neural system tumour syndrome family
.
Hum Mol Genet
2001
;
10
:
55
62
.
18.
Tachibana
I
,
Smith
JS
,
Sato
K
,
Hosek
SM
,
Kimmel
DW
,
Jenkins
RB
. 
Investigation of germline PTEN, p53, p16INK4A/p14ARF, and CDK4 alterations in familial glioma
.
Am J Med Genet
2000
;
92
:
136
41
.
19.
Bainbridge
MN
,
Armstrong
GN
,
Gramatges
MM
,
Bertuch
AA
,
Jhangiani
SN
,
Doddapaneni
H
, et al
Germline mutations in shelterin complex genes are associated with familial glioma
.
J Natl Cancer Inst
2015
;
107
:
384
.
20.
Walsh
KM
,
Anderson
E
,
Hansen
HM
,
Decker
PA
,
Kosel
ML
,
Kollmeyer
T
, et al
Analysis of 60 reported glioma risk SNPs replicates published GWAS findings but fails to replicate associations from published candidate-gene studies
.
Genet Epidemiol
2013
;
37
:
222
8
.
21.
Kinnersley
B
,
Kamatani
Y
,
Labussiere
M
,
Wang
Y
,
Galan
P
,
Mokhtari
K
, et al
Search for new loci and low-frequency variants influencing glioma risk by exome-array analysis
.
Eur J Hum Genet
2016
;
24
:
717
24
.
22.
Amirian
ES
,
Armstrong
GN
,
Zhou
R
,
Lau
CC
,
Claus
EB
,
Barnholtz-Sloan
JS
, et al
The Glioma International Case-Control Study: A report from the Genetic Epidemiology of Glioma International Consortium
.
Am J Epidemiol
2016
;
183
:
85
91
.
23.
Kinnersley
B
,
Mitchell
JS
,
Gousias
K
,
Schramm
J
,
Idbaih
A
,
Labussiere
M
, et al
Quantifying the heritability of glioma using genome-wide complex trait analysis
.
Sci Rep
2015
;
5
:
17267
.
24.
Melin
BS
,
Barnholtz-Sloan
JS
,
Wrensch
MR
,
Johansen
C
,
Il'yasova
D
,
Kinnersley
B
, et al
Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors
.
Nat Genet
2017
;
49
:
789
94
.
25.
Kinnersley
B
,
Labussiere
M
,
Holroyd
A
,
Di Stefano
AL
,
Broderick
P
,
Vijayakrishnan
J
, et al
Genome-wide association study identifies multiple susceptibility loci for glioma
.
Nat Commun
2015
;
6
:
8559
.
26.
Wrensch
M
,
Jenkins
RB
,
Chang
JS
,
Yeh
RF
,
Xiao
Y
,
Decker
PA
, et al
Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility
.
Nat Genet
2009
;
41
:
905
8
.
27.
Stacey
SN
,
Sulem
P
,
Jonasdottir
A
,
Masson
G
,
Gudmundsson
J
,
Gudbjartsson
DF
, et al
A germline variant in the TP53 polyadenylation signal confers cancer susceptibility
.
Nat Genet
2011
;
43
:
1098
103
.
28.
Jenkins
RB
,
Xiao
Y
,
Sicotte
H
,
Decker
PA
,
Kollmeyer
TM
,
Hansen
HM
, et al
A low-frequency variant at 8q24.21 is strongly associated with risk of oligodendroglial tumors and astrocytomas with IDH1 or IDH2 mutation
.
Nat Genet
2012
;
44
:
1122
5
.
29.
Walsh
KM
,
Codd
V
,
Smirnov
IV
,
Rice
T
,
Decker
PA
,
Hansen
HM
, et al
Variants near TERT and TERC influencing telomere length are associated with high-grade glioma risk
.
Nat Genet
2014
;
46
:
731
5
.
30.
Shete
S
,
Hosking
FJ
,
Robertson
LB
,
Dobbins
SE
,
Sanson
M
,
Malmer
B
, et al
Genome-wide association study identifies five susceptibility loci for glioma
.
Nat Genet
2009
;
41
:
899
904
.
31.
Sanson
M
,
Hosking
FJ
,
Shete
S
,
Zelenika
D
,
Dobbins
SE
,
Ma
Y
, et al
Chromosome 7p11.2 (EGFR) variation influences glioma risk
.
Hum Mol Genet
2011
;
20
:
2897
904
.
32.
Enciso-Mora
V
,
Hosking
FJ
,
Di Stefano
AL
,
Zelenika
D
,
Shete
S
,
Broderick
P
, et al
Low penetrance susceptibility to glioma is caused by the TP53 variant rs78378222
.
Br J Cancer
2013
;
108
:
2178
85
.
33.
Rajaraman
P
,
Melin
BS
,
Wang
Z
,
McKean-Cowdin
R
,
Michaud
DS
,
Wang
SS
, et al
Genome-wide association study of glioma and meta-analysis
.
Hum Genet
2012
;
131
:
1877
88
.
34.
Enciso-Mora
V
,
Hosking
FJ
,
Kinnersley
B
,
Wang
Y
,
Shete
S
,
Zelenika
D
, et al
Deciphering the 8q24.21 association for glioma
.
Hum Mol Genet
2013
;
22
:
2293
302
.
35.
Amos
CI
,
Dennis
J
,
Wang
Z
,
Byun
J
,
Schumacher
FR
,
Gayther
SA
, et al
The OncoArray Consortium: A network for understanding the genetic architecture of common cancers
.
Cancer Epidemiol Biomarkers Prev
2017
;
26
:
126
35
.
36.
Sanz-Moreno
V
,
Gaggioli
C
,
Yeo
M
,
Albrengues
J
,
Wallberg
F
,
Viros
A
, et al
ROCK and JAK1 signaling cooperate to control actomyosin contractility in tumor cells and stroma
.
Cancer Cell
2011
;
20
:
229
45
.
37.
Nicolas
CS
,
Amici
M
,
Bortolotto
ZA
,
Doherty
A
,
Csaba
Z
,
Fafouri
A
, et al
The role of JAK-STAT signaling within the CNS
.
JAKSTAT
2013
;
2
:
e22925
.
38.
Riemenschneider
MJ
,
Buschges
R
,
Wolter
M
,
Reifenberger
J
,
Bostrom
J
,
Kraus
JA
, et al
Amplification and overexpression of the MDM4 (MDMX) gene from 1q32 in a subset of malignant gliomas without TP53 mutation or MDM2 amplification
.
Cancer Res
1999
;
59
:
6091
6
.
39.
Garcia-Closas
M
,
Couch
FJ
,
Lindstrom
S
,
Michailidou
K
,
Schmidt
MK
,
Brook
MN
, et al
Genome-wide association studies identify four ER negative-specific breast cancer risk loci
.
Nat Genet
2013
;
45
:
392
8
.
40.
Eeles
RA
,
Al Olama
AA
,
Benlloch
S
,
Saunders
EJ
,
Leongamornlert
DA
,
Tymrakiewicz
M
, et al
Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array
.
Nat Genet
2013
;
45
:
385
91
.
41.
Boland
E
,
Clayton-Smith
J
,
Woo
VG
,
McKee
S
,
Manson
FD
,
Medne
L
, et al
Mapping of deletion and translocation breakpoints in 1q44 implicates the serine/threonine kinase AKT3 in postnatal microcephaly and agenesis of the corpus callosum
.
Am J Hum Genet
2007
;
81
:
292
303
.
42.
Turner
KM
,
Sun
Y
,
Ji
P
,
Granberg
KJ
,
Bernard
B
,
Hu
L
, et al
Genomically amplified Akt3 activates DNA repair pathway and promotes glioma progression
.
Proc Natl Acad Sci USA
2015
;
112
:
3421
6
.
43.
Watanabe
T
,
Nobusawa
S
,
Kleihues
P
,
Ohgaki
H
. 
IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas
.
Am J Pathol
2009
;
174
:
1149
53
.
44.
Yan
H
,
Parsons
DW
,
Jin
G
,
McLendon
R
,
Rasheed
BA
,
Yuan
W
, et al
IDH1 and IDH2 mutations in gliomas
.
N Engl J Med
2009
;
360
:
765
73
.
45.
Noushmehr
H
,
Weisenberger
DJ
,
Diefes
K
,
Phillips
HS
,
Pujara
K
,
Berman
BP
, et al
Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma
.
Cancer Cell
2010
;
17
:
510
22
.
46.
Christensen
BC
,
Smith
AA
,
Zheng
S
,
Koestler
DC
,
Houseman
EA
,
Marsit
CJ
, et al
DNA methylation, isocitrate dehydrogenase mutation, and survival in glioma
.
J Natl Cancer Inst
2011
;
103
:
143
53
.
47.
Sanson
M
,
Marie
Y
,
Paris
S
,
Idbaih
A
,
Laffaire
J
,
Ducray
F
, et al
Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas
.
J Clin Oncol
2009
;
27
:
4150
4
.
48.
Eckel-Passow
JE
,
Lachance
DH
,
Molinaro
AM
,
Walsh
KM
,
Decker
PA
,
Sicotte
H
, et al
Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors
.
N Engl J Med
2015
;
372
:
2499
508
.
49.
Gur
G
,
Rubin
C
,
Katz
M
,
Amit
I
,
Citri
A
,
Nilsson
J
, et al
LRIG1 restricts growth factor signaling by enhancing receptor ubiquitylation and degradation
.
EMBO J
2004
;
23
:
3270
81
.
50.
Yang
JA
,
Liu
BH
,
Shao
LM
,
Guo
ZT
,
Yang
Q
,
Wu
LQ
, et al
LRIG1 enhances the radiosensitivity of radioresistant human glioblastoma U251 cells via attenuation of the EGFR/Akt signaling pathway
.
Int J Clin Exp Pathol
2015
;
8
:
3580
90
.
51.
Wei
J
,
Qi
X
,
Zhan
Q
,
Zhou
D
,
Yan
Q
,
Wang
Y
, et al
miR-20a mediates temozolomide-resistance in glioblastoma cells via negatively regulating LRIG1 expression
.
Biomed Pharmacother
2015
;
71
:
112
8
.
52.
Schumacher
FR
,
Schmit
SL
,
Jiao
S
,
Edlund
CK
,
Wang
H
,
Zhang
B
, et al
Genome-wide association study of colorectal cancer identifies six new susceptibility loci
.
Nat Commun
2015
;
6
:
7138
.
53.
Codd
V
,
Nelson
CP
,
Albrecht
E
,
Mangino
M
,
Deelen
J
,
Buxton
JL
, et al
Identification of seven loci affecting mean telomere length and their association with disease
.
Nat Genet
2013
;
45
:
422
7
.
54.
Houlston
RS
,
Cheadle
J
,
Dobbins
SE
,
Tenesa
A
,
Jones
AM
,
Howarth
K
, et al
Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33
.
Nat Genet
2010
;
42
:
973
7
.
55.
Weinrich
SL
,
Pruzan
R
,
Ma
L
,
Ouellette
M
,
Tesmer
VM
,
Holt
SE
, et al
Reconstitution of human telomerase with the template RNA component hTR and the catalytic protein subunit hTRT
.
Nat Genet
1997
;
17
:
498
502
.
56.
Haiman
CA
,
Chen
GK
,
Vachon
CM
,
Canzian
F
,
Dunning
A
,
Millikan
RC
, et al
A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor-negative breast cancer
.
Nat Genet
2011
;
43
:
1210
4
.
57.
Wang
Y
,
Broderick
P
,
Webb
E
,
Wu
X
,
Vijayakrishnan
J
,
Matakidou
A
, et al
Common 5p15.33 and 6p21.33 variants influence lung cancer risk
.
Nat Genet
2008
;
40
:
1407
9
.
58.
McKay
JD
,
Hung
RJ
,
Gaborieau
V
,
Boffetta
P
,
Chabrier
A
,
Byrnes
G
, et al
Lung cancer susceptibility locus at 5p15.33
.
Nat Genet
2008
;
40
:
1404
6
.
59.
Turnbull
C
,
Rapley
EA
,
Seal
S
,
Pernet
D
,
Renwick
A
,
Hughes
D
, et al
Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer
.
Nat Genet
2010
;
42
:
604
7
.
60.
Killedar
A
,
Stutz
MD
,
Sobinoff
AP
,
Tomlinson
CG
,
Bryan
TM
,
Beesley
J
, et al
A common cancer risk-associated allele in the hTERT locus encodes a dominant negative inhibitor of telomerase
.
PLoS Genet
2015
;
11
:
e1005286
.
61.
Huang
PH
,
Xu
AM
,
White
FM
. 
Oncogenic EGFR signaling networks in glioma
.
Sci Signal
2009
;
2
:
re6
.
62.
Yin
W
,
Rossin
A
,
Clifford
JL
,
Gronemeyer
H
. 
Co-resistance to retinoic acid and TRAIL by insertion mutagenesis into RAM
.
Oncogene
2006
;
25
:
3735
44
.
63.
Sparmann
A
,
van Lohuizen
M
. 
Polycomb silencers control cell fate, development and cancer
.
Nat Rev Cancer
2006
;
6
:
846
56
.
64.
Lynch
MD
,
Smith
AJ
,
De Gobbi
M
,
Flenley
M
,
Hughes
JR
,
Vernimmen
D
, et al
An interspecies analysis reveals a key role for unmethylated CpG dinucleotides in vertebrate Polycomb complex recruitment
.
EMBO J
2012
;
31
:
317
29
.
65.
Khalil
AM
,
Guttman
M
,
Huarte
M
,
Garber
M
,
Raj
A
,
Rivea Morales
D
, et al
Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression
.
Proc Natl Acad Sci USA
2009
;
106
:
11667
72
.
66.
Tomlinson
I
,
Webb
E
,
Carvajal-Carmona
L
,
Broderick
P
,
Kemp
Z
,
Spain
S
, et al
A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21
.
Nat Genet
2007
;
39
:
984
8
.
67.
Yeager
M
,
Orr
N
,
Hayes
RB
,
Jacobs
KB
,
Kraft
P
,
Wacholder
S
, et al
Genome-wide association study of prostate cancer identifies a second risk locus at 8q24
.
Nat Genet
2007
;
39
:
645
9
.
68.
Crowther-Swanepoel
D
,
Broderick
P
,
Di Bernardo
MC
,
Dobbins
SE
,
Torres
M
,
Mansouri
M
, et al
Common variants at 2q37.3, 8q24.21, 15q21.3 and 16q24.1 influence chronic lymphocytic leukemia risk
.
Nat Genet
2010
;
42
:
132
6
.
69.
Easton
DF
,
Pooley
KA
,
Dunning
AM
,
Pharoah
PD
,
Thompson
D
,
Ballinger
DG
, et al
Genome-wide association study identifies novel breast cancer susceptibility loci
.
Nature
2007
;
447
:
1087
93
.
70.
Yap
KL
,
Li
S
,
Munoz-Cabello
AM
,
Raguz
S
,
Zeng
L
,
Mujtaba
S
, et al
Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a
.
Mol Cell
2010
;
38
:
662
74
.
71.
Kotake
Y
,
Nakagawa
T
,
Kitagawa
K
,
Suzuki
S
,
Liu
N
,
Kitagawa
M
, et al
Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15INK4B tumor suppressor gene
.
Oncogene
2011
;
30
:
1956
62
.
72.
Bishop
DT
,
Demenais
F
,
Iles
MM
,
Harland
M
,
Taylor
JC
,
Corda
E
, et al
Genome-wide association study identifies three loci associated with melanoma risk
.
Nat Genet
2009
;
41
:
920
5
.
73.
Kyritsis
AP
,
Zhang
B
,
Zhang
W
,
Xiao
M
,
Takeshima
H
,
Bondy
ML
, et al
Mutations of the p16 gene in gliomas
.
Oncogene
1996
;
12
:
63
7
.
74.
Foulkes
WD
,
Flanders
TY
,
Pollock
PM
,
Hayward
NK
. 
The CDKN2A (p16) gene and human cancer
.
Mol Med
1997
;
3
:
5
20
.
75.
Alentorn
A
,
Dehais
C
,
Ducray
F
,
Carpentier
C
,
Mokhtari
K
,
Figarella-Branger
D
, et al
Allelic loss of 9p21.3 is a prognostic factor in 1p/19q codeleted anaplastic gliomas
.
Neurology
2015
;
85
:
1325
31
.
76.
Miyake
Y
,
Nakamura
M
,
Nabetani
A
,
Shimamura
S
,
Tamura
M
,
Yonehara
S
, et al
RPA-like mammalian Ctc1-Stn1-Ten1 complex binds to single-stranded DNA and protects telomeres independently of the Pot1 pathway
.
Mol Cell
2009
;
36
:
193
206
.
77.
Chen
LY
,
Redon
S
,
Lingner
J
. 
The human CST complex is a terminator of telomerase activity
.
Nature
2012
;
488
:
540
4
.
78.
Zhang
C
,
Doherty
JA
,
Burgess
S
,
Hung
RJ
,
Lindstrom
S
,
Kraft
P
, et al
Genetic determinants of telomere length and risk of common cancers: a Mendelian randomization study
.
Hum Mol Genet
2015
;
24
:
5356
66
.
79.
Walsh
KM
,
Wiencke
JK
,
Lachance
DH
,
Wiemels
JL
,
Molinaro
AM
,
Eckel-Passow
JE
, et al
Telomere maintenance and the etiology of adult glioma
.
Neuro Oncol
2015
;
17
:
1445
52
.
80.
Walsh
KM
,
Codd
V
,
Rice
T
,
Nelson
CP
,
Smirnov
IV
,
McCoy
LS
, et al
Longer genotypically-estimated leukocyte telomere length is associated with increased adult glioma risk
.
Oncotarget
2015
;
6
:
42468
77
.
81.
Law
MH
,
Bishop
DT
,
Lee
JE
,
Brossard
M
,
Martin
NG
,
Moses
EK
, et al
Genome-wide meta-analysis identifies five new susceptibility loci for cutaneous malignant melanoma
.
Nat Genet
2015
;
47
:
987
95
.
82.
Chahal
HS
,
Wu
W
,
Ransohoff
KJ
,
Yang
L
,
Hedlin
H
,
Desai
M
, et al
Genome-wide association study identifies 14 novel risk alleles associated with basal cell carcinoma
.
Nat Commun
2016
;
7
:
12510
.
83.
Gudmundsson
J
,
Thorleifsson
G
,
Sigurdsson
JK
,
Stefansdottir
L
,
Jonasson
JG
,
Gudjonsson
SA
, et al
A genome-wide association study yields five novel thyroid cancer risk loci
.
Nat Commun
2017
;
8
:
14517
.
84.
Cao
Y
,
Saygili
Y
,
Ummadisingu
A
,
Teuscher
J
,
Luo
J
,
Pellet
N
, et al
11% efficiency solid-state dye-sensitized solar cells with copper(II/I) hole transport materials
.
Nat Commun
2017
;
8
:
15390
.
85.
Bose
A
,
Guilherme
A
,
Huang
S
,
Hubbard
AC
,
Lane
CR
,
Soriano
NA
, et al
The v-SNARE Vti1a regulates insulin-stimulated glucose transport and Acrp30 secretion in 3T3-L1 adipocytes
.
J Biol Chem
2005
;
280
:
36946
51
.
86.
Ramirez
DM
,
Khvotchev
M
,
Trauterman
B
,
Kavalali
ET
. 
Vti1a identifies a vesicle pool that preferentially recycles at rest and maintains spontaneous neurotransmission
.
Neuron
2012
;
73
:
121
34
.
87.
Zhang
B
,
Jia
WH
,
Matsuda
K
,
Kweon
SS
,
Matsuo
K
,
Xiang
YB
, et al
Large-scale genetic study in East Asians identifies six new loci associated with colorectal cancer risk
.
Nat Genet
2014
;
46
:
533
42
.
88.
Lan
Q
,
Hsiung
CA
,
Matsuo
K
,
Hong
YC
,
Seow
A
,
Wang
Z
, et al
Genome-wide association analysis identifies new lung cancer susceptibility loci in never-smoking women in Asia
.
Nat Genet
2012
;
44
:
1330
5
.
89.
Michailidou
K
,
Hall
P
,
Gonzalez-Neira
A
,
Ghoussaini
M
,
Dennis
J
,
Milne
RL
, et al
Large-scale genotyping identifies 41 new loci associated with breast cancer risk
.
Nat Genet
2013
;
45
:
353
61
.
90.
Wu
L
,
Sun
T
,
Kobayashi
K
,
Gao
P
,
Griffin
JD
. 
Identification of a family of mastermind-like transcriptional coactivators for mammalian notch receptors
.
Mol Cell Biol
2002
;
22
:
7688
700
.
91.
Chen
SJ
,
Zelent
A
,
Tong
JH
,
Yu
HQ
,
Wang
ZY
,
Derre
J
, et al
Rearrangements of the retinoic acid receptor alpha and promyelocytic leukemia zinc finger genes resulting from t(11;17)(q23;q21) in a patient with acute promyelocytic leukemia
.
J Clin Invest
1993
;
91
:
2260
7
.
92.
Ahmad
KF
,
Engel
CK
,
Prive
GG
. 
Crystal structure of the BTB domain from PLZF
.
Proc Natl Acad Sci USA
1998
;
95
:
12123
8
.
93.
Al Olama
AA
,
Kote-Jarai
Z
,
Berndt
SI
,
Conti
DV
,
Schumacher
F
,
Han
Y
, et al
A meta-analysis of 87,040 individuals identifies 23 new susceptibility loci for prostate cancer
.
Nat Genet
2014
;
46
:
1103
9
.
94.
Law
PJ
,
Berndt
SI
,
Speedy
HE
,
Camp
NJ
,
Sava
GP
,
Skibola
CF
, et al
Genome-wide association analysis implicates dysregulation of immunity genes in chronic lymphocytic leukaemia
.
Nat Commun
2017
;
8
:
14175
.
95.
Rice
T
,
Zheng
S
,
Decker
PA
,
Walsh
KM
,
Bracci
P
,
Xiao
Y
, et al
Inherited variant on chromosome 11q23 increases susceptibility to IDH-mutated but not IDH-normal gliomas regardless of grade or histology
.
Neuro Oncol
2013
;
15
:
535
41
.
96.
Zhou
QL
,
Jiang
ZY
,
Mabardy
AS
,
Del Campo
CM
,
Lambright
DG
,
Holik
J
, et al
A novel pleckstrin homology domain-containing protein enhances insulin-stimulated Akt phosphorylation and GLUT4 translocation in adipocytes
.
J Biol Chem
2010
;
285
:
27581
9
.
97.
Denduluri
SK
,
Idowu
O
,
Wang
Z
,
Liao
Z
,
Yan
Z
,
Mohammed
MK
, et al
Insulin-like growth factor (IGF) signaling in tumorigenesis and the development of cancer drug resistance
.
Genes Dis
2015
;
2
:
13
25
.
98.
Skibola
CF
,
Berndt
SI
,
Vijai
J
,
Conde
L
,
Wang
Z
,
Yeager
M
, et al
Genome-wide association study identifies five susceptibility loci for follicular lymphoma outside the HLA region
.
Am J Hum Genet
2014
;
95
:
462
71
.
99.
McKay
JD
,
Hung
RJ
,
Han
Y
,
Zong
X
,
Carreras-Torres
R
,
Christiani
DC
, et al
Large-scale association analysis identifies new lung cancer susceptibility loci and heterogeneity in genetic susceptibility across histological subtypes
.
Nat Genet
2017
;
49
:
1126
32
.
100.
Johnson
EO
,
Chang
KH
,
de Pablo
Y
,
Ghosh
S
,
Mehta
R
,
Badve
S
, et al
PHLDA1 is a crucial negative regulator and effector of Aurora A kinase in breast cancer
.
J Cell Sci
2011
;
124
(
Pt 16
):
2711
22
.
101.
Sellheyer
K
,
Nelson
P
,
Kutzner
H
,
Patel
RM
. 
The immunohistochemical differential diagnosis of microcystic adnexal carcinoma, desmoplastic trichoepithelioma and morpheaform basal cell carcinoma using BerEP4 and stem cell markers
.
J Cutan Pathol
2013
;
40
:
363
70
.
102.
Dieci
G
,
Fiorino
G
,
Castelnuovo
M
,
Teichmann
M
,
Pagano
A
. 
The expanding RNA polymerase III transcriptome
.
Trends Genet
2007
;
23
:
614
22
.
103.
Saitsu
H
,
Osaka
H
,
Sasaki
M
,
Takanashi
J
,
Hamada
K
,
Yamashita
A
, et al
Mutations in POLR3A and POLR3B encoding RNA Polymerase III subunits cause an autosomal-recessive hypomyelinating leukoencephalopathy
.
Am J Hum Genet
2011
;
89
:
644
51
.
104.
McCartney
S
,
Little
BM
,
Langeberg
LK
,
Scott
JD
. 
Cloning and characterization of A-kinase anchor protein 100 (AKAP100). A protein that targets A-kinase to the sarcoplasmic reticulum
.
J Biol Chem
1995
;
270
:
9327
33
.
105.
Leiserson
MD
,
Blokh
D
,
Sharan
R
,
Raphael
BJ
. 
Simultaneous identification of multiple driver pathways in cancer
.
PLoS Comput Biol
2013
;
9
:
e1003054
.
106.
Frerman
FE
. 
Acyl-CoA dehydrogenases, electron transfer flavoprotein and electron transfer flavoprotein dehydrogenase
.
Biochem Soc Trans
1988
;
16
:
416
8
.
107.
Indo
Y
,
Glassberg
R
,
Yokota
I
,
Tanaka
K
. 
Molecular characterization of variant alpha-subunit of electron transfer flavoprotein in three patients with glutaric acidemia type II–and identification of glycine substitution for valine-157 in the sequence of the precursor, producing an unstable mature protein in a patient
.
Am J Hum Genet
1991
;
49
:
575
80
.
108.
Freneaux
E
,
Sheffield
VC
,
Molin
L
,
Shires
A
,
Rhead
WJ
. 
Glutaric acidemia type II. Heterogeneity in beta-oxidation flux, polypeptide synthesis, and complementary DNA mutations in the alpha subunit of electron transfer flavoprotein in eight patients
.
J Clin Invest
1992
;
90
:
1679
86
.
109.
Xipell
E
,
Aragon
T
,
Martinez-Velez
N
,
Vera
B
,
Idoate
MA
,
Martinez-Irujo
JJ
, et al
Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51
.
Neuro Oncol
2016
;
18
:
1109
19
.
110.
Ruark
E
,
Seal
S
,
McDonald
H
,
Zhang
F
,
Elliot
A
,
Lau
K
, et al
Identification of nine new susceptibility loci for testicular cancer, including variants near DAZL and PRDM14
.
Nat Genet
2013
;
45
:
686
9
.
111.
Wu
C
,
Kraft
P
,
Zhai
K
,
Chang
J
,
Wang
Z
,
Li
Y
, et al
Genome-wide association analyses of esophageal squamous cell carcinoma in Chinese identify multiple susceptibility loci and gene-environment interactions
.
Nat Genet
2012
;
44
:
1090
7
.
112.
Petitjean
A
,
Achatz
MI
,
Borresen-Dale
AL
,
Hainaut
P
,
Olivier
M
. 
TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes
.
Oncogene
2007
;
26
:
2157
65
.
113.
Egan
KM
,
Nabors
LB
,
Olson
JJ
,
Monteiro
AN
,
Browning
JE
,
Madden
MH
, et al
Rare TP53 genetic variant associated with glioma risk and outcome
.
J Med Genet
2012
;
49
:
420
1
.
114.
Wang
Z
,
Rajaraman
P
,
Melin
BS
,
Chung
CC
,
Zhang
W
,
McKean-Cowdin
R
, et al
Further confirmation of germline glioma risk variant rs78378222 in TP53 and its implication in tumor tissues via integrative analysis of TCGA data
.
Hum Mutat
2015
;
36
:
684
8
.
115.
Vannier
JB
,
Sandhu
S
,
Petalcorin
MI
,
Wu
X
,
Nabi
Z
,
Ding
H
, et al
RTEL1 is a replisome-associated helicase that promotes telomere and genome-wide replication
.
Science
2013
;
342
:
239
42
.
116.
Michailidou
K
,
Lindstrom
S
,
Dennis
J
,
Beesley
J
,
Hui
S
,
Kar
S
, et al
Association analysis identifies 65 new breast cancer risk loci
.
Nature
2017
;
551
:
92
4
.
117.
Rothman
N
,
Garcia-Closas
M
,
Chatterjee
N
,
Malats
N
,
Wu
X
,
Figueroa
JD
, et al
A multi-stage genome-wide association study of bladder cancer identifies multiple susceptibility loci
.
Nat Genet
2010
;
42
:
978
84
.
118.
Weinhold
N
,
Johnson
DC
,
Chubb
D
,
Chen
B
,
Forsti
A
,
Hosking
FJ
, et al
The CCND1 c.870G>A polymorphism is a risk factor for t(11;14)(q13;q32) multiple myeloma
.
Nat Genet
2013
;
45
:
522
5
.
119.
Cancer Genome Atlas Research Network
,
Brat
DJ
,
Verhaak
RG
,
Aldape
KD
,
Yung
WK
,
Salama
SR
, et al
Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas
.
N Engl J Med
2015
;
372
:
2481
98
.
120.
Di Stefano
AL
,
Enciso-Mora
V
,
Marie
Y
,
Desestret
V
,
Labussiere
M
,
Boisselier
B
, et al
Association between glioma susceptibility loci and tumour pathology defines specific molecular etiologies
.
Neuro Oncol
2013
;
15
:
542
7
.
121.
Dubrow
R
,
Darefsky
AS
. 
Demographic variation in incidence of adult glioma by subtype, United States, 1992–2007
.
BMC Cancer
2011
;
11
:
325
.
122.
Chen
H
,
Chen
Y
,
Zhao
Y
,
Fan
W
,
Zhou
K
,
Liu
Y
, et al
Association of sequence variants on chromosomes 20, 11, and 5 (20q13.33, 11q23.3, and 5p15.33) with glioma susceptibility in a Chinese population
.
Am J Epidemiol
2011
;
173
:
915
22
.
123.
Wang
D
,
Hu
E
,
Wu
P
,
Yuan
W
,
Xu
S
,
Sun
Z
, et al
Genetic variant near TERC influencing the risk of gliomas with older age at diagnosis in a Chinese population
.
J Neurooncol
2015
;
124
:
57
64
.
124.
Safaeian
M
,
Rajaraman
P
,
Hartge
P
,
Yeager
M
,
Linet
M
,
Butler
MA
, et al
Joint effects between five identified risk variants, allergy, and autoimmune conditions on glioma risk
.
Cancer Causes Control
2013
;
24
:
1885
91
.
125.
Anic
GM
,
Thompson
RC
,
Nabors
LB
,
Olson
JJ
,
Browning
JE
,
Madden
MH
, et al
An exploratory analysis of common genetic variants in the vitamin D pathway including genome-wide associated variants in relation to glioma risk and outcome
.
Cancer Causes Control
2012
;
23
:
1443
9
.
126.
Kitahara
CM
,
Linet
MS
,
Brenner
AV
,
Wang
SS
,
Melin
BS
,
Wang
Z
, et al
Personal history of diabetes, genetic susceptibility to diabetes, and risk of brain glioma: a pooled analysis of observational studies
.
Cancer Epidemiol Biomarkers Prev
2014
;
23
:
47
54
.
127.
Pierce
BL
,
Ahsan
H
,
Vanderweele
TJ
. 
Power and instrument strength requirements for Mendelian randomization studies using multiple genetic variants
.
Int J Epidemiol
2011
;
40
:
740
52
.
128.
Jager
R
,
Migliorini
G
,
Henrion
M
,
Kandaswamy
R
,
Speedy
HE
,
Heindl
A
, et al
Capture Hi-C identifies the chromatin interactome of colorectal cancer risk loci
.
Nat Commun
2015
;
6
:
6178
.
129.
Dryden
NH
,
Broome
LR
,
Dudbridge
F
,
Johnson
N
,
Orr
N
,
Schoenfelder
S
, et al
Unbiased analysis of potential targets of breast cancer susceptibility loci by Capture Hi-C
.
Genome Res
2014
;
24
:
1854
68
.
130.
Dixon
JR
,
Jung
I
,
Selvaraj
S
,
Shen
Y
,
Antosiewicz-Bourget
JE
,
Lee
AY
, et al
Chromatin architecture reorganization during stem cell differentiation
.
Nature
2015
;
518
:
331
6
.
131.
Flavahan
WA
,
Drier
Y
,
Liau
BB
,
Gillespie
SM
,
Venteicher
AS
,
Stemmer-Rachamimov
AO
, et al
Insulator dysfunction and oncogene activation in IDH mutant gliomas
.
Nature
2016
;
529
:
110
4
.
132.
Telomeres Mendelian Randomization C
,
Haycock
PC
,
Burgess
S
,
Nounu
A
,
Zheng
J
,
Okoli
GN
, et al
Association between telomere length and risk of cancer and non-neoplastic diseases: a mendelian randomization study
.
JAMA Oncol
2017
;
3
:
636
51
.
133.
e GP
. 
Enhancing GTEx by bridging the gaps between genotype, gene expression, and disease
.
Nat Genet
2017
;
49
:
1664
70
.