Familial glioma

2018-09-01   Riccardo Bazzoni , Angela Bentivegna 

School of Medicine and Surgery, University of Milan-Bicocca, via Cadore, Monza, Italy (RB,AB); 2 NeuroMI, Milan center of Neuroscience, University of Milan-Bicocca, Dept. of Neurology and Neuroscience, San Gerardo Hospital, via Pergolesi, Monza, Italy (AB)r.bazzoni@campus.unimib.it; angela.bentivegna@unimib.it

Identity

Name

Familial glioma

Note

Primary central nervous system (CNS) tumors can be divided into gliomas and non-gliomas. For the more recent classification of gliomas (2016 WHO classification), see Table 1.
 , 
Glioma entityWHO Grade , 
Diffuse astrocytic and oligodendroglial tumors Diffuse astrocytoma, IDH-mutantII , 
- Gemistocytic astrocytoma, IDH-mutantII , 
Diffuse astrocytoma, IDH-wildtypeII , 
Diffuse astrocytoma, NOSII , 
Anaplastic astrocytoma, IDH-mutantIII , 
Anaplastic astrocytoma, IDH-wildtypeIII , 
Anaplastic astrocytoma, NOSIII , 
Glioblastoma, IDH-wildtypeIV , 
- Giant cell glioblastomaIV , 
- GliosarcomaIV , 
- Epithelioid glioblastomaIV , 
Glioblastoma, IDH-mutantIV , 
Glioblastoma, NOSIV , 
Diffuse midline glioma, H3K27M-mutantIV , 
Oligodendroglioma, IDH-mutant and 1p\/19q-codeletedII , 
Oligodendroglioma, NOSII , 
Anaplastic oligodendroglioma, IDH-mutant and 1p\/19q-codeleted   III , 
Anaplastic oligodendroglioma, NOSIII , 
Oligoastrocytoma, NOSII , 
Anaplastic oligoastrocytoma, NOSIII , 
Other (astrocytic) gliomasPilocytic astrocytoma I , 
- Pilomyxoid astrocytomaII , 
Subependymal giant cell astrocytomaI , 
Pleomorphic xanthoastrocytomaII , 
Anaplastic pleomorphic xanthoastrocytomaIII , 
Ependymal tumors SubependymomaI , 
Myxopapillary ependymomaI , 
EpendymomaII , 
- Papillary ependymomaII , 
- Clear cell ependymomaII , 
- Tanycytic ependymomaII , 
Ependymoma, RELA fusion-positiveII or III , 
Anaplastic ependymomaIII , 
Other gliomasAngiocentric glioma I , 
Chordoid glioma of third ventricleII , 
AstroblastomaLow\/high grade

Genes involved

TERT , EGFR , CCDC26 , CDKN2A , CDKN2B , PHLDB1 , RTEL1 , POT1 , SPAG9 , RUNDC1 , PTEN , TP53 , CDK4 , ETFA , ZBTB16 , POLR3B , NF1 , NF2 , TSC1 , TSC2 , MSH2 , MLH1 , MSH6 , PMS2 , APC , IDH1 , IDH2 , RB1 , BRCA2 , MYO19 , KIF18B

Clinics

Note

Gliomas represent 30% of all brain and central nervous system (CNS) tumors and 80% of all malignant brain tumors. The most common and malignant glioma is glioblastoma multiforme (GBM) (Goodenberger and Jenkins, 2012). Although there are several histologic types of gliomas, the incidence rates for all sporadic gliomas range from 4.67 to 5.73 per 100,000 persons (Barbagallo et al., 2016). Gliomas are more common in men than in women and in white rather than in black population (Ostrom et al., 2013). Anyway, familial glioma cases are similar to sporadic ones in terms of gender distribution, age, morphology and grade as shown in Table 2 (results from Gliogene Consortium https:\/\/www.bcm.edu\/centers\/cancer-center\/research\/gliogene\/) (Sadetzki et al., 2013).
 TotalIncident* (diagnosed from 2007)Prevalent* (diagnosed before 2007)
  TotalEurope & IsraelUSTotal
n=376 familiesn=219 familiesn=73 familiesn=146 familiesn=157 families
n=841 gliomasn=481 gliomasn=159 gliomasn=322 gliomasn=360 gliomas
n%n%n%n%n%
Gender    
  Males45053.526755.59358.51745418350.8
  Females39146.521444.56614.51484617749.2
  M\/F ratio (all)1.15 1.25 1.14 1.18 1.10 
Age at diagnosis ** (n=831)          
  Mean SD49.4 18.7 51.0 19.051.1 16.450.9 20.147.2 18.1 
  Range1-92 1-924-821-921-91 
  <20597.0347.142.5309.4256.9
  20-3918622.29620.13421.46219.49025.1
  40-5931637.716233.86239.010031.315442.9
  60-6916920.211624.24025.2762.8514.8
  70+10812.97114.81911.95216.33710.3
Tumor grade *** (n=739)          
  I354.7204.764145.1154.7
  II16522.57718.236244115.08827.8
  III12617.07016.530204014.75617.7
  IV41355.925660.5785217865.215749.7
Age at diagnosis by grade *****           
  Grades I-II37.2 16.1 38.5 17.442.7 16.235.3 17.835.9 14.8 
  Grades III-IV54.8 16.4 55.8 16.4 55.1 14.8 56.1 17.2 53.4 16.3 
Tumor grade by gender (n=739) ****          
  Grades I-II total200100971004210055100103100
  Males9949.544450.421502341.8555.4
  Females10150.55354.621503258.24846.6
  Grades III-IV total53910032610010810021100213100
  Males30055.719158.6686312356.410951.2
  Females23944.313541.440379543.610448.8

Table 2 | Distribution of glioma cases by date of diagnosis* and selected demographic and clinical characteristics. *When the glioma in the proband was diagnosed from 2007 all gliomas in the family were included in the incident cases column; when the glioma in the proband was diagnosed before 2007 all gliomas in the family were included in the prevalent cases column; **Excluding three cases with unknown age at diagnosis; comparison between mean age at diagnosis of incident and prevalent p = 0.003; ***p-value = 0.00009 (total incidents versus prevalent); ****For 92 cases of the 831 verified tumours, tumour histological behavior was unknown, and for 58 cases of the 481 verified tumours from the total incident cases, tumour histological behavior was unknown; *****For grades I-II p-value = 0.3 (total incident versus total prevalent). [Modified from Sadetzki et al., 2013]

Neoplastic risk

ENVIRONMENTAL: Some epidemiologic risk factors might lead to development of glioma such as therapeutic ionizing radiation, pesticides, smoking, petroleum refining or production work and employment in synthetic rubber manufacturing (Alifieris and Trafalis, 2015). An inverse association between glioma incidence and allergies, atopic diseases and systemic infections has been reported by multiple groups (Goodenberger and Jenkins, 2012).
FAMILIARITY: Excluding those gliomas known to be due to rare hereditary cancer syndromes such as Turcots and Li-Fraumeni syndromes as well as neurofibromatosis ( NF1, NF2) or tuberous sclerosis (Melin et al., 2017), there is evidence that gliomas cluster in families. Most familial gliomas appear to comprise clusters of two cases, suggesting low penetrance and a low risk of developing additional gliomas (Sadetzki et al., 2013). It is currently thought that approximately 5-10% of patients have a family history of glioma (Lindor et al., 2008, Robertson et al., 2010). An increased risk of developing primary brain tumors among first-degree relatives of patients with gliomas has been shown (Robertson et al., 2010), and there is a greater risk for first-degree relatives of probands with a younger age of onset than for first-degree relatives of probands with later onset (Malmer et al., 2003, Blumenthal and Cannon-Albright, 2008), as shown in Table 3.
 Cancer in probandCancer in relativeNo. relativesObservedExpectedRRp Value
AAstrocytoma\/GBMAstrocytoma\/GBM  11,4983811.63.29 <0.00001
AstrocytomaAstrocytoma5,637102.53.820.0004
GBMGBM5,93983.52.290.026
BAstrocytoma\/GBMAstrocytoma\/GBM  36,6503125.31.220.15
AstrocytomaAstrocytoma17,163126.31.910.03
GBMGBM19,94086.31.260.30
CAstrocytoma\/GBM <20y (n=214)  Astrocytoma\/GBM  1,05940.66.440.004
Astrocytoma <15y (n=161)Astrocytoma80130.39.650.004
GBM <55y (n=187)GBM1,47000.7--

Table 3 | Relative risks (RR) for brain tumor among: first-degree relatives of patients (A), second-degree relatives of patients (B), first-degree relatives of patients with early onset brain tumor (C). [Modified from Blumenthal and Cannon-Albright, 2008].
Anyway, the third-degree relative risks were not significantly elevated for astrocytoma, GBM or for the two types combined (Blumenthal and Cannon-Albright, 2008). However, familial aggregation of cancer can indicate a genetic etiology but may also indicate shared familial environmental exposures. Unfortunately, a multifactorial inheritance model could not be clearly rejected (Table 4) (de Andrade et al., 2001, Malmer et al., 2001, Shete et al., 2011).
Observed CancersPatients NumbersRelatives NumbersEtiology of Familial CancersStudiess
Clustering of multiple cancers in relatives of glioma patients.639 (under age 65 years)5088(first degree:3810, second degree: 1278)Multigenic action (unknown environmental exposure)de Andrade et al. (2001)
SIR 5.08 (FDRs, 45 years) melanoma, brain tumors, sarcoma; SIR 0.95 (FDRs, 45 years).1476 (under age 75 years)8746(all first degree)Unknown similar genetic contributionScheurer et al. (2007)
SIR 1.1, 95% CI 0.8-1.4 for all cancers (melanoma:SIR 4.0, 95% CI 1.5-8.8; meningioma: SIR 5.5, 95% CI 1.1-16).Multiple adult glioma patients in 17 Finnish families Unknown cancer susceptibility traitPaunu et al. (2002)
RR 3.29, 95% CI: 2.33-4.51, P 0.00001
RR 1.22, 95% CI: 0.83-1.74, P 0.15.
UPDB* in 1401 primary brain tumor cases with at least 3 generations of genealogy dataFirst degree: 11 498
Second degree: 36 650
Heritable glioma risk and shared environmentBlumenthal and Cannon-Albright (2008)

Table 4 | Epidemologic studies in families with gliomas and other tumors. *Utah Population Data Base; RR=risk relative; CI=confidential interval; SIR=standardized incidence ratio; FDR=first-relative degree [Modified from Kyritsis et al., 2010]
The variation in inherited risk of glioma could be related to combinations of multiple risk variants. Here, we reported the most significant variants (SNPs) figured out from GWASs (Table 5 and 6).
Gene and\/or
chromosome location
SNPOdds RatioRisk Allele
Frequency (controls)
Associated Glioma SubtypeOther Association
TERT
(5p15.33)
rs27361001.350.50All glioma subtypesIncreases risk of cancer at other sites, including
lung, testis, pancreas and colon
EGFR
(7p11.2)
rs2252586
rs11979158
1.20
1.25
0.28
0.83
All glioma subtypes 
CCDC26 (8q24.21)rs557058575.000.05Oligodendroglial
tumors\/IDH-mutant
astrocytic tumors
 
CDKN2B (9p21.3)rs1412829

rs4977756
1.30.41Astrocytic tumors,
WHO grades II-IV
Glioma
 
PHLDB1 (11q23.3)rs4988721.500.32IDH-mutant gliomas 
TP53 (17p13.1)rs783782222.700.01All glioma subtypesIncreases risk of several Li-Fraumeni tumors, including
basal cell carcinoma, prostate cancer, GBM and colorectal Adenoma
RTEL1 (20q13.33)rs60106201.400.75All glioma subtypes 
ETFA (15q24.2)rs180591 1.20 All glioma subtypes 
1p31.3rs1275525521.180.87All glioma subtypes 
1q32.1rs42527071.120.22All glioma subtypes 
1q44rs120763731.090.84All glioma subtypes 
2q33.3rs75722631.110.76All glioma subtypes 
3p14.1rs117068321.080.46All glioma subtypes 
10q24.33rs115980181.100.46All glioma subtypes 
11q14.1rs112332501.140.87All glioma subtypes 
11q21rs71077851.070.48All glioma subtypes 
14q12rs101310321.170.92All glioma subtypes 
16p13.3rs2562152
rs3751667
1.09
1.14
0.85
0.21
All glioma subtypes 
16q.12.1rs108526061.140.71All glioma subtypes 
22q13.1rs22355731.090.51All glioma subtypes 
VTI1A
(10q25.2) 
rs1116960670.89 All glioma subtypes 
ZBTB16 (11q23.2)rs6480441.10 All glioma subtypes 
Intergenic (12q21.2) rs122301720.88 All glioma subtypes 
POLR3B (12q23.3) rs38516340.87 All glioma subtypes 

Table 5 | Heritable variants associated with glioma risk from GWASs. Data from Kinnersley et al., 2015, Kinnersley et al., 2015, Melin et al., 2017, Ostrom et al., 2014.
Type of polymorphismGenetic LocusGlioma riskStudies
1013 glioma cases, 1016 controls, 1127 SNPs, and 388 putative functional SNPs in 136 DNA repair genes.rs243356 (intron 3 CHAF1A gene).OR 1.32Bethke et al. (2008)
217 cases, 1171 controls IL-4Rα, IL-13, Cyclooxygenase-2.rs1805015, rs1801275 (T-G IL-4Rα haplotype).OR 2.26Schwartzbaum et al. (2007)
456 cases and 541 controls IL-4 and IL-13 pathways.A IL-4 haplotype, borderline increased risk
A rare IL-4 haplotype, decreased risk
A common IL-13 haplotype, decreased risk.
OR 1.5
OR 0.23
OR 0.73
Wiemels et al. (2007)
309 patients with newly diagnosed glioma; 342 control subjects; XRCC1, XRCC3, RAD51, XRCC7, p53.XRCC7 G6721T (GT heterozygotes)
TT genotype increased in cases.
OR 1.78
OR 1.86
Wang et al. (2004)
1005 glioma cases; 1101 controls; MTHFR C677A and A1298C, MTRR A66G, and MTR A2756G variants.
MTHFR C677T A1298C diplotypes, increased risk.OR 1.23Bethke et al. (2009)
771 glioma patients; 752 controls; LIG4 and XRCC4 SNPs.Single locus: variant LIG4 SNP2 rs3093739:T > C, increased risk;
3 locus: LIG4 SNP4 rs1805388:C > T, XRCC4 SNP12 rs7734849:A > T, SNP15 rs1056503:G > T; more than additive increased risk P = .001
 Liu et al. (2008)
373 Caucasian glioma patients; 365 Caucasian controls; ERCC1, XRCC1, APEX1, PARP1, MGMT, LIG1, SNPs.6 SNPs (ERCC1 3UTR, XRCC1 R399Q, APEX1 E148D, PARP1 A762V, MGMT F84L, and LIG1 5UTR) increased glioma risk; MGMT F84L, main risk factor; MGMT F84L plus PARP1 A762V, dramatic increase glioma risk.OR 5.95Liu et al. (2009)
701 glioma cases; 1560 controls; XRCC1 and XRCC3 SNPs.Studied SNPs, not increased risk; SNP combinations: homozygous genotypes,
XRCC1 Gln399Gln and XRCC3 Met241Met, 3-fold glioma risk.
OR 3.18Kiuru et al. (2008)
1005 cases; 1011 controls; CASP8 D302H polymorphism.Carriers, 1.37 increased risk.OR 1.37Bethke et al. (2008)
CASP8, CCND1, CCNH, CDKN1A, CDKN2A, CHEK1, CHEK2, MDM2, PTEN, TP53 polymorphisms.CCND1 Ex4-1G > A and CCNH Ex8 + 49T > C variants, increased glioma risk;
MDM2 Ex12 + 162A > G, reduced glioma risk.
 Rajaraman et al. (2007)
236 glioma patients; 366 controls; MMP-1, MMP-3, MMP-9 polymorphisms.MMP-1 -1607 1G\/1G genotype and MMP-1 1G-MMP-3 6A haplotype may play protective role in the development of adult astrocytoma.OR 0.45Lu et al. (2007)
771 glioma patients; 752 healthy controls XRCC5, XRCC6, XRCC7 polymorphisms.XRCC5 haplotype CAGTT, 40% reduction in glioma risk.OR 0.60Liu et al. (2007)

Table 6 | Representative recent studies describing genetic polymorphism linked to glioma risk. OD= odd ratio [Modified from Kyritsis et al., 2010]
A particular attention goes to POT1 gene, which belongs to the telomere-shelterin complex. Indeed, Bainbridge et al. found two different mutations in POT1 in two families (A and B) (Bainbridge et al., 2015). In family A, six individuals had POT1 mutation (NM_015450:p.G95C, HG19:chr7:g.124503667C>A), of whom three developed glioma. In family B, also six individuals had POT1 mutation (NM_015450:p.E450X, HG19:chr7:g.124481048C>A) and two developed glioma. Moreover, they identified, in a third family (C), a third protein-changing mutation (NM_015450:p.D617Efs*8, HG19:chr7:g.124464068TTA>T). In families with POT1 mutations, they reported that the affected members suffered from oligodendroglioma, which is substantially sensitive to irradiation. Anyway, the association between familial glioma and POT1 mutations still needs to be validated.
Jalali et al. figured out that MYO19 and KIF18B genes and rare variants in SPAG9 and RUNDC1 are potentially involved in familial gliomas (Jalali et al., 2015).
MENDELIAN CANCER SYNDROMES: A heritable genetic contribution to gliomagenesis was initially suggested by the increased incidence of these tumors in families with Mendelian cancer syndromes (Table 7). Although numerous familial cancer syndromes are associated with increased glioma risk, monogenic Mendelian disorders account for only a small proportion of adult glioma incidence at the population level (Ostrom et al., 2014). However, germline mutations of PTEN, TP53, CDKN2A p16(INK4A)\/p14(ARF), and CDK4 are not common events in familial glioma, but occasionally they may account for a subset of familial glioma cases (Tachibana et al., 2000). Several syndromes are associated to pediatric glioma (Ripperger et al., 2017).
Disorder\/Syndrome (OMIM code)Gene name (chromosome location)Mode of inheritanceAssociated gliomas
Neurofibromatosis 1 (#162200)NF1 (17q11.2)DominantAstrocytoma
Neurofibromatosis 2 (#607379)NF2 (22q12.2)DominantEpendymoma
Tuberous sclerosis(#191100; #613254)TSC1 (9q34.14)
TSC2 (16p13.3)
DominantGiant cell astrocytoma
Lynch syndrome\/Turcots syndrome (type 1)(#120435; #276300)
constitutional mismatch repair deficiency syndrome( CMMRDS#276300)
MSH2 (2p21)
MLH1 (3p22.2)
MSH6 (2p16.3)
PMS2 (7p22.1)
Dominant and recessiveGlioblastoma, other gliomas , childhood cancer for recessive form
Turcots syndrome (type 2)(#175100)APC (5q22.2)Probably recessivePrimary brain tumor
Li-Fraumeni syndrome\/Families with patients with multifocal glioma, glioma + second cancer (#137800)  TP53 (17p13.1)DominantGlioblastoma, other gliomas
Melanoma-neural system tumor syndrome (#155755)p16\/CDKN2A (9p21.3)DominantGlioma
Ollier disease\/Maffucci syndrome (#166000;#614569)IDH1 (2q33.3)
IDH2 (15q26.1)
Acquired postzygotic mosaicism;
dominant with reducedpenetrance
Glioma
Retinoblastoma (#180200)RB1 (13q14.2)DominantGlioblastoma, other gliomas
FANCD1(# 605724), GLM3(#613029)BRCA2 (13q13.1) RecessiveGlioma, multicentric GBM

Table 7 | Known germline gene mutations associated with increased risk of glioma. Data from Ostrom et al., 2014, Kyritsis et al., 2010.
To date, no sex-linked disorders have been associated with increased glioma risk, nor has any SNP on the X chromosome been identified as a glioma risk factor in previous genome-wide association studies. However, somatic loss of-function mutations in the X chromosome gene Alpha thalassemia\/mental retardation syndrome X-linked (ATRX) have been observed in 20 % of adult oligodendroglioma tumors and in 80 % of grade 2 and 3 astrocytomas (Osorio et al., 2015).

Treatment

Multimodal therapies including surgical resection, radio- and chemotherapy (Bush et al., 2017).

Evolution

The lower-grade gliomas can evolve towards higher-grade ones.

Prognosis

Except for pilocytic astrocytomas ID: 5773>, the median survival of glioma patients is still poor (12-14 months). The 5-years survival of GBM patients is

Cytogenetics

Note

Here, we reported the most karyotype abnormalities associated with familial gliomas found in literature (Table 8)
 Sex (age at diagnosis)RelationshipKaryotype abnormalitiesNote
Duhaime et al. (1989)1) Female (2,5)
2) Male (5)
Siblings. Patient 1: The stem line karyotype of the tumor showed translocation t(11;14) and was often seen in a tetraploid version of the basic karyotype (48, XX, -14, +der(11)t(11;14) (p11.2-3;q11), +marker, +marker). Chromosomes from peripheral blood lymphocytes were normal
Patient 2: tumor contained normal 46, XY cell as well as cells with both numerical and structural abnormalities, even if a consistent stem line could not be discerned.
Both patients presented GBM.
Family history showed no genetic syndromes or cancers.
Authors suggested some possible agents in environment to which the siblings were exposed, causing the formation of their tumors.
Arruda et al. (1995)1) Female (7)Fourth proband generation. Sister of two brothers. One of these two brothers died by brain tumor.Tumor karyotype: 46, XX, 7q-\/ 46 ,XX ,-2 ,4p- ,7p- ,+15 \/ 46, XX
Peripheral blood lymphocytes were normal: 46, XX.
Patient presented GBM.
Patient was in a family with several members having GBM or other malignant tumors in other areas (breast, larynx and colon). 
Dirven et al. (1995)1) Male (22)
2) Female (29)
3) Female (33)
4) Female (41)
Siblings.Kayotypes from peripheral blood lymphocytes for all patients were normal.
Patient 4 showed mutations in codon 220 (T-->G; exon 6) and in codon 273 (C-->T; exon 8) of TP53 in tumor cells.
Patients 1 and 2 presented glioma.
Patients 3 and 4 presented GBM.
No other family members in three generations were affected by malignant brain tumors.
Patel et al. (1998)1) Male (6)
2) Male (13)
3) Male (73)
Patient 3 is the granduncle of patient 1.
Patient 3 is the grandfather of patient 2
CNAs from tumor specimens:
Patient 1: No detectable abnormality.
Patient 2: -Y.
Patient 3: +7q, -10, -13q(21-->33), -21.
Patient 1 presented an anaplastic astrocytoma.
Patient 2 presented an astrocytoma.
Patient 3 presented GBM.
In the family, there were other three cases of GBM and of other brain tumors.
4) Female (25)
5) Male (52)
Patient 5 is the paternal uncle of patient 4.CNAs from tumor specimens:
Patient 4:  -1p, +3q(13.3-->9), -4q, +12, -15, -19q, -X
Patient 5: ++7p(11.1-->12), +19.
Patient 4 presented anaplastic oligodendroglioma.
Patient 5 presented GBM.
6) Male (44)
7) Male (77)
Patient 7 is the paternal uncle of patient 6.CNAs from tumor specimens:
Patient 6: +7, -10, +12p, ++12p(11-->12), ++12q(13.2-->14), -13q(21-->33).
Patient 7: ++7p(11.21), ++7p(11.1-->12), ++(21.233), -9q34, -10, -16p, -19, -22.
Both patients presented GBM.
8) Male (68)Patient has one first cousin with GBM and one with an astrocytoma.CNAs from tumor specimen:
Patient 8: +7, -10, 18.
Patient presented GBM.
Ugonabo et al. (2011)1) Male (63)
2) Male (81)
Brothers.Tumor karyotype of patient 2 revealed trisomies of 4, 8, 12, 22 and LOH of 1p, 9p, and 10.Both patients presented GBM.
Chromosomal abnormalities not found in all tumor cells.
Osorio et al. (2014)1) Male (17)
2) Male (21)
Brothers.Tumor karyotype: Both patients had 1p and 19q deleted.Both patients presented oligodendroglioma.
3) Male (55)
4) Male (59)
Brothers.Tumor karyotype of patient 3 showed 1p and 19q deleted.
Tumor karyotype of patient 4 showed 1p\/19q intact.
Patient 3 presented anaplastic oligodendroglioma
Patient 4 presented GBM w\/oligo features.

Table 8 | Summary of karyotype abnormalities associated with familial gliomas found in literature. = copy number alterations

Genes involved and Proteins

Bibliography

Pubmed IDLast YearTitleAuthors
259445282015Glioblastoma multiforme: Pathogenesis and treatment.Alifieris C et al
74875461995Familial glioblastoma.Arruda WO et al
254825302015Germline mutations in shelterin complex genes are associated with familial glioma.Bainbridge MN et al
183980422008The common D302H variant of CASP8 is associated with risk of glioma.Bethke L et al
184833422008Functional polymorphisms in folate metabolism genes influence the risk of meningioma and glioma.Bethke L et al
188098382008Familiality in brain tumors.Blumenthal DT et al
270858592017Current and future strategies for treatment of glioma.Bush NA et al
75957551995Glioblastoma multiforme in four siblings: a cytogenetic and molecular genetic study.Dirven CM et al
25387721989Simultaneous presentation of glioblastoma multiforme in siblings two and five years old: case report.Duhaime AC et al
232382842012Genetics of adult glioma.Goodenberger ML et al
256521572015Targeted sequencing in chromosome 17q linkage region identifies familial glioma candidates in the Gliogene Consortium.Jalali A et al
266259492015Quantifying the heritability of glioma using genome-wide complex trait analysis.Kinnersley B et al
183305152008XRCC1 and XRCC3 variants and risk of glioma and meningioma.Kiuru A et al
201503732010Inherited predisposition to glioma.Kyritsis AP et al
185593312008Concise handbook of familial cancer susceptibility syndromes - second edition.Lindor NM et al
191244992009Association and interactions between DNA repair gene polymorphisms and adult glioma.Liu Y et al
173896092007Tagging SNPs in non-homologous end-joining pathway genes and risk of glioma.Liu Y et al
181659452008Polymorphisms of LIG4 and XRCC4 involved in the NHEJ pathway interact to modify risk of glioma.Liu Y et al
175029982007Polymorphisms in the matrix metalloproteinase-1, 3, and 9 promoters and susceptibility to adult astrocytoma in northern China.Lu Z et al
128002032003Familial brain tumours-genetics or environment? A nationwide cohort study of cancer risk in spouses and first-degree relatives of brain tumour patients.Malmer B et al
111614122001Genetic epidemiology of glioma.Malmer B et al
283464432017Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors.Melin BS et al
252084782015Familial gliomas: cases in two pairs of brothers.Osorio JA et al
248429562014The epidemiology of glioma in adults: a "state of the science" review.Ostrom QT et al
241370152013CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010.Ostrom QT et al
94065861998Gliomas in families: chromosomal analysis by comparative genomic hybridization.Patel A et al
120972922002A novel low-penetrance locus for familial glioma at 15q23-q26.3.Paunu N et al
176841422007Polymorphisms in apoptosis and cell cycle control genes and risk of brain tumors in adults.Rajaraman P et al
281688332017Childhood cancer predisposition syndromes-A concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology.Ripperger T et al
204550252010Survey of familial glioma and role of germline p16INK4A/p14ARF and p53 mutation.Robertson LB et al
269815072015Recurrent Glioblastoma: Where we stand.Roy S et al
232904252013Description of selected characteristics of familial glioma patients - results from the Gliogene Consortium.Sadetzki S et al
180069422007Aggregation of cancer in first-degree relatives of patients with glioma.Scheurer ME et al
180069352007An international case-control study of interleukin-4Ralpha, interleukin-13, and cyclooxygenase-2 polymorphisms and glioblastoma risk.Schwartzbaum JA et al
220378772011Genome-wide high-density SNP linkage search for glioma susceptibility loci: results from the Gliogene Consortium.Shete S et al
107974392000Investigation of germline PTEN, p53, p16(INK4A)/p14(ARF), and CDK4 alterations in familial glioma.Tachibana I et al
221406382011Familial glioblastoma: A case report of glioblastoma in two brothers and review of literature.Ugonabo I et al
153138912004Polymorphisms of DNA repair genes and risk of glioma.Wang LE et al
175486902007Allergy-related polymorphisms influence glioma status and serum IgE levels.Wiemels JL et al
111804512001Segregation analysis of cancer in families of glioma patients.de Andrade M et al

External Links