ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Risk factors for brain tumors

Risk factors for brain tumors
Literature review current through: Jan 2024.
This topic last updated: Sep 21, 2023.

INTRODUCTION — Primary brain tumors are a diverse group of neoplasms arising from different cells of the central nervous system (CNS). (See "Classification and pathologic diagnosis of gliomas, glioneuronal tumors, and neuronal tumors", section on 'Histopathologic and molecular classification'.)

Although incidence rates for primary brain and CNS cancers were increasing through the late 1980s, the rates have been decreasing by approximately 0.2 percent annually over the past 15 years [1]. Among the factors that contributed to the early increase in rates were the introduction of noninvasive diagnostic technology including computed tomography (CT) in the 1970s and magnetic resonance imaging (MRI) in the 1980s, as well as better health care access for older adults [2].

These data, in conjunction with evidence suggesting the increase may have been occurring for many decades, leave open the possibility that environmental exposures may account for the increasing incidence of brain tumors.

General risk factors that have been associated with brain tumors are discussed here. A number of putative risk factors for brain tumors have been examined. A shortcoming in many reports is the tendency to group all brain tumors together; this approach may miss important exposures for specific histopathologic types of tumors [3].

The epidemiology of meningioma and its associated risk factors are discussed separately. (See "Epidemiology, pathology, clinical features, and diagnosis of meningioma".)

GENETIC PREDISPOSITION SYNDROMES — Approximately 1 to 5 percent of brain tumors are due to genetic syndromes that confer an increased risk of developing tumors of the nervous system [4-6]. Some of these tumors are associated with neurofibromatosis and several other inherited syndromes.

Neurofibromatosis type 1 — Neurofibromatosis type 1 (NF1) occurs in 1 of 3000 persons and is linked to a gene on chromosome 17. The NF1 gene encodes a protein called neurofibromin that restricts cell proliferation by activating guanosine triphosphate (GTP) hydrolysis on Ras proteins. (See "Neurofibromatosis type 1 (NF1): Pathogenesis, clinical features, and diagnosis".)

Multiple neurofibromas are seen, and some undergo malignant change to neurofibrosarcoma. (See "Pathogenetic factors in soft tissue and bone sarcomas".)

Other malignancies that develop in up to 5 to 10 percent of patients with NF1 include other malignant nerve sheath tumors such as malignant schwannomas and astrocytomas. The astrocytomas are usually low grade and frequently have a pilocytic histology. These lesions have a predilection for the optic pathways, hypothalamus, and cerebellum.

It has been proposed that malignant degeneration in NF1 reflects the two-hit hypothesis in which one allele is constitutionally inactivated in the germline while the other allele undergoes somatic inactivation (the second hit) [7]. Animal models are consistent with this hypothesis but suggest that the second hit can be a mutation in the p53 gene [8,9].

NF2-related schwannomatosis — NF2-related schwannomatosis (NF2) is an autosomal dominant disorder predisposing to multiple neoplastic lesions. This disorder is due to a mutation in the NF2 gene, a tumor suppressor gene on chromosome 22 that encodes a membrane cytoskeletal protein called merlin or schwannomin [10] that appears to be involved in actin-cytoskeleton organization [11]. Other modifier genes may also be involved [12]. (See "NF2-related schwannomatosis (formerly neurofibromatosis type 2)", section on 'Molecular pathogenesis'.)

The pathognomonic findings are bilateral vestibular schwannomas (acoustic neuromas). Vestibular schwannomas are seen in 90 to 95 percent of patients with NF2 and generally develop by 30 years of age. (See "Vestibular schwannoma (acoustic neuroma)" and "NF2-related schwannomatosis (formerly neurofibromatosis type 2)", section on 'Vestibular schwannomas'.)

Other types of brain tumors are also seen, the most frequent of which are meningiomas. Approximately one-half of individuals with NF2 have meningiomas, and multiple meningiomas are often present [13]. The incidence of meningioma increases with age, and the lifetime risk may be as high as 75 percent [14]. Patients with NF2 tend to develop meningiomas at an earlier age than those with sporadic meningiomas. The meningiomas seen in patients with NF2 are more frequently atypical or anaplastic compared with sporadic tumors [15,16]. (See "NF2-related schwannomatosis (formerly neurofibromatosis type 2)", section on 'Vestibular schwannomas'.)

von Hippel-Lindau syndrome — The von Hippel-Lindau syndrome is an autosomal dominant disorder associated with hemangioblastomas, pancreatic cysts and neuroendocrine tumors, renal tumors, and pheochromocytomas. The gene on chromosome 3p25 normally functions as a tumor suppressor gene. (See "Clinical features, diagnosis, and management of von Hippel-Lindau disease" and "Molecular biology and pathogenesis of von Hippel-Lindau disease".)

Li-Fraumeni syndrome — The Li-Fraumeni syndrome (LFS) is inherited as an autosomal dominant trait and is usually associated with a germline mutation in the tumor protein 53 (TP53) gene. (See "Li-Fraumeni syndrome" and "Gene test interpretation: TP53".)

LFS is primarily characterized by sarcomas, breast cancer, leukemia, adrenocortical cancer, and brain tumors occurring before the age of 45 years. A variety of brain tumors are seen in LFS, most commonly choroid plexus tumors in infants, medulloblastomas in children, and astrocytomas in young adults [17]. Of particular note, a high percentage of choroid plexus carcinomas are associated with germline mutations in TP53 even in the absence of another cancer or a positive family history. (See "Uncommon brain tumors", section on 'Choroid plexus carcinoma'.)

Outside of choroid plexus carcinoma, which is included in LFS criteria, recommendations for TP53 germline testing in patients with brain tumors have not been established. Some experts have proposed that several other brain tumors prompt consideration of germline testing, based on findings suggesting high prevalence of germline TP53 mutations in specific histologic and molecular subgroups, independent of personal and family history of other tumors [17]. These include:

Lower-grade choroid plexus tumors in children with a mutant p53 pattern in tumor by immunohistochemistry (IHC). (See "Uncommon brain tumors", section on 'Choroid plexus tumors'.)

Sonic hedgehog (SHH) medulloblastoma in children – Large sequencing efforts have found that germline TP53 mutations are present in 20 percent (13 out of 63) of SHH medulloblastomas in children aged 5 to 16 years and 8 percent (13 out of 170) of all pediatric SHH medulloblastomas [18]. Genetic testing should be considered for pediatric SHH medulloblastomas (table 1). (See "Histopathology, genetics, and molecular groups of medulloblastoma", section on 'Genetic predisposition'.)

Astrocytomas with rare isocitrate dehydrogenase type 1 (IDH1) R132C mutation – A strong association between germline TP53 and a rare IDH1 mutation (R132C) has been identified in LFS families [19] and in two germline TP53 mutations identified in The Cancer Genome Atlas (TCGA) database [20], suggesting that patients with this mutation might be recommended for germline TP53 testing. (See "Classification and pathologic diagnosis of gliomas, glioneuronal tumors, and neuronal tumors", section on 'IDH1/IDH2 mutation'.)

Clinical criteria for LFS, testing recommendations, and cancer surveillance strategies are reviewed in detail separately. (See "Li-Fraumeni syndrome".)

Familial adenomatous polyposis — Familial adenomatous polyposis (FAP) is an autosomal dominant condition caused by a mutation to the adenomatous polyposis coli gene on chromosome 5. The majority of FAP-associated brain tumors are medulloblastomas, but gliomas have also been described. (See "Clinical manifestations and diagnosis of familial adenomatous polyposis".)

Mismatch repair deficiency

Lynch syndrome – Lynch syndrome, previously called hereditary nonpolyposis colorectal cancer (HNPCC), is a cancer predisposition syndrome caused by a germline mutation that impairs deoxyribonucleic acid (DNA) nucleotide mismatch repair (MMR). Lynch syndrome genes include MLH1, MSH2, MSH6, PMS2, and EPCAM. Patients with Lynch syndrome are at increased risk for high-grade gliomas, in addition to colorectal cancer and other solid tumors (table 2) [21]. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis".)

Although MMR deficiency and Lynch syndrome are rare among patients with glioma as a whole, the prevalence is higher in younger patients with IDH-wildtype glioblastoma in particular. In a study that included 1225 adult gliomas referred to a single neuropathology department for diagnosis and next-generation sequencing, nine gliomas were MMR deficient (0.73 percent), including eight IDH-wildtype glioblastomas and one IDH-mutant astrocytoma [22]. Five of nine patients had Lynch syndrome confirmed by germline testing (overall prevalence of Lynch syndrome, 0.41 percent). In a complementary analysis including 257 additional IDH-wildtype glioblastomas enriched for early-onset cases, MMR deficiency was present in 12.5 percent of tumors in patients between 18 and 39 years of age and 2.6 percent of tumors in patients between 40 and 49 years of age. These data support neuropathologic testing for MMR deficiency in IDH-wildtype glioblastomas in patients younger than 50 years of age at diagnosis.

All patients with MMR deficiency identified on somatic (tumor) testing should be offered genetic counseling and germline testing for Lynch syndrome (see "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis", section on 'When to suspect Lynch syndrome'). A diagnosis of Lynch syndrome has important implications for health maintenance, reproductive counseling, and cancer screening (table 3), as reviewed separately. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Cancer screening and management".)

Constitutional mismatch repair-deficiency (CMMR-D) – Biallelic mutations in mismatch repair genes (eg, MLH1, MSH2, MSH6, PMS2) cause the CMMR-D syndrome. Patients with CMMR-D are at risk for brain tumors in late childhood (primarily glioblastoma) and may have an NF1-like phenotype, with café-au-lait macules and axillary freckling [23,24]. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis" and "Neurofibromatosis type 1 (NF1): Pathogenesis, clinical features, and diagnosis", section on 'Constitutional mismatch repair-deficiency syndrome'.)

Glioblastomas and other tumors associated with CMMR-D have a high mutational load compared with sporadic tumors, predicting that they may be responsive to immune checkpoint inhibition, and at least one case report describes a favorable response to nivolumab in two children with recurrent glioblastoma related to CMMR-D [25]. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors".)

Basal cell nevus syndrome — Persons with the basal cell nevus syndrome (Gorlin syndrome, nevoid basal cell cancer syndrome) have an increased risk of medulloblastoma. The syndrome is caused by germline mutations of the patched 1 (PTCH1) gene, a tumor suppressor gene. (See "Nevoid basal cell carcinoma syndrome (Gorlin syndrome)".)

OTHER GENETIC FACTORS

Familial glioma — The above inherited syndromes explain only a fraction of families with an aggregation of gliomas [26-28]. Little is known about the genetic factors underlying the apparent susceptibility to glioma in families without one of the recognized genetic syndromes. Shared environmental exposures may contribute to risk in some families as well.

In one population-based study that included 2141 first-degree relatives of 297 individuals with astrocytoma diagnosed over an eight-year period in Sweden, 5 percent had a familial aggregation of glioma [29]. Segregation analysis of 14 families with at least two affected first-degree relatives suggested an autosomal recessive pattern of inheritance in 65 percent (nine families), while an autosomal dominant inheritance pattern was suspected in only 21 percent (three families).

In a multinational study that included 376 families with two or more verified gliomas, the majority of families (83 percent) had only two gliomas, of which 57 percent were in first-degree relatives and 32 percent were in second-degree relatives [30]. The age, sex, and grade distribution of familial cases was similar to that of sporadic cases in the literature. An autosomal dominant pattern of inheritance with low penetrance was suggested by the large number of families with only two glioma cases in consecutive generations, but a clear mode of inheritance was not identified.

Large-scale sequencing efforts will likely identify causative germline mutations in some of these families. As an example, whole-exome sequencing of 90 affected individuals in 55 glioma families identified protein-changing variants in the protection of telomeres 1 (POT1) gene in three families with mostly oligodendrogliomas [31]. POT1 encodes a component of the shelterin complex, which participates in telomere maintenance and response to DNA damage. Incomplete penetrance was demonstrated in the two larger families, with approximately half of individuals known to carry the mutation developing a glioma. Additional cancers in these families included lung cancer (n = 3), leukemia, colon cancer, and kidney cancer (one each). Rare loss-of-function mutations in POT1 have also been described in familial melanoma [32,33].

Single nucleotide polymorphisms — Several genome-wide association studies (GWAS) have examined the risk of brain tumors and identified genetic polymorphisms associated with glioma, including TERT, CCDC26, CDKN2A/CDKN2B, RTEL1, PHLDB1, and EGFR [34-37]. A large meta-analysis identified 13 new susceptibility loci for glioma; while some susceptibility loci are common to all glioma subtypes (eg, TP53), others appear to be specific to either glioblastoma (eg, EGFR) or non-glioblastoma glioma (eg, PHLDB1) [38]. A single variant in the 8q24.21 region has been strongly associated with risk of IDH-mutant oligodendrogliomas and astrocytomas [39], with a magnitude of risk comparable to BRCA1 gene mutations and breast cancer. This single nucleotide polymorphism (rs55705857) has been shown to be causally linked to IDH-mutant low-grade gliomas through the organic cation transporter (OCT)-mediated regulation of MYC expression [40].

Two susceptibility loci have been identified for meningioma (10q12.31 and 11p15.5) [41,42].

Genetic polymorphisms of interest have also been identified using candidate gene approaches. For example, individuals with variants in genes involved in carcinogen metabolism [43], DNA repair [43,44], inflammation/allergies [45-48], and other immune responses [49] may be at higher risk of brain tumors. While collectively these gene variants may not provide strong enough associations to identify high-risk groups (for screening), they may provide new insights into the pathways involved in carcinogenesis.

IONIZING RADIATION — Exposure to ionizing radiation, as occurs from therapeutic radiation therapy or among atomic bomb survivors, is an established cause of brain tumors, including meningiomas, gliomas, and nerve sheath tumors [50]. The latency between irradiation and the development of brain tumors may be as short as five years or as long as many decades. Higher doses of irradiation are associated with both an increased risk of developing a secondary brain tumor and a shorter latency period.

Therapeutic brain radiation used for the treatment of primary brain tumors, acute leukemia, and other tumors has allowed patients to survive for extended periods, thereby placing them at risk for secondary malignancies. This association has been demonstrated for both meningiomas and gliomas. The risk is higher for meningioma than glioma and does not appear to plateau over time [51].

In the Childhood Cancer Survivor Study, glioma was diagnosed in 40 patients (0.3 percent), with the diagnosis being made at a median follow-up of nine years [52]. Gliomas occurred in 24 patients who had leukemia and eight with a previous brain tumor. The primary risk factor was antecedent radiation (odds ratio [OR] 6.8, compared with survivors who had not received cranial radiation). The risk of developing glioma was greatest in children who were irradiated before age five years, and was not related to other forms of treatment or the underlying diagnosis.

The combination of prophylactic cranial irradiation with antimetabolite therapy has resulted in a much higher incidence of second tumors in survivors of acute lymphocytic leukemia. As an example, brain tumors occurred in 6 of 52 children (13 percent) treated with combined therapy, compared with no brain tumors in 101 children who did not receive radiotherapy [53]. Three of the six children had a genetic defect in thiopurine metabolism that resulted in higher levels of the active metabolite of 6-mercaptopurine.

Lower doses of radiation are also important. The relationship of exposure of the brain and skull to lower doses of irradiation (eg, treatment of tinea capitis) is clearly linked to the subsequent development of meningiomas and other brain tumors. The relationship of irradiation and meningioma is discussed separately. (See "Epidemiology, pathology, clinical features, and diagnosis of meningioma", section on 'Ionizing radiation'.)

Childhood exposure to diagnostic head computed tomography (CT) may also be associated with an increased risk of brain tumors [54,55]. (See "Radiation-related risks of imaging", section on 'Children and adolescents'.)

NONIONIZING RADIATION

Low-frequency electromagnetic fields — Low-frequency electromagnetic field exposure has been postulated as a potential risk factor for brain tumors, and several positive associations were reported in historical literature. Biologic plausibility has not been established, however, and studies using more rigorous methodology, including direct, in-home measurement of electromagnetic fields, have concluded that a large effect of this type of radiation on the risk of adult brain tumors can be excluded [3,56]. The International Agency for Research on Cancer (IARC) has concluded that data are insufficient to classify low-frequency electromagnetic fields as a risk factor for brain tumors [57].

Similarly, there is no evidence that exposure of children or pregnant women to magnetic fields from high-current power lines, electric heating sources, or electric appliances is associated with the subsequent occurrence of brain tumors in children [56,58-61]. Analyses of occupational exposure to magnetic fields have also failed to establish a relationship to the risk of brain tumors [62,63].

Cellular phones and radiofrequency fields — Cellular telephones are a source of radiofrequency fields that have received coverage in the popular media as potential risk factors for brain tumors, presumably due to exposure of the head of the user to radiofrequency energy. Other sources of radiofrequency field exposure include microwave and radar equipment and occupational exposures (sealers, plastic welders, amateur radio operators, medical personnel, and telecommunications workers). Exposures to radiofrequency energy are difficult to quantify, even under laboratory conditions [64].

Numerous epidemiologic studies have looked for a possible relationship between cellular telephone use and the development of brain tumors [65,66]. A meta-analysis that included data from 22 case control series concluded that there was a slight increase in risk associated with cell phone use in those studies where investigators were blinded to whether the participant was a case or a control [65]. Furthermore, the risk appeared to be associated with an induction period of 10 years or longer. Experimental studies suggest that increased levels of oxidation with radiofrequency electromagnetic exposures may explain the association [67].

However, many of the retrospective studies could have biases, such as recall bias, that could explain the positive findings. A prospective cohort study that included over 775,000 females and 3268 brain tumors reported no increase in the incidence of brain tumors in cellular telephone users compared with nonusers, overall or by tumor subtype (including glioma and vestibular schwannoma), over 14 years of follow-up [68]. Similarly, analyses considering changes in incidence rates over time, prevalence of mobile phone use, and latency period provide no support for causal associations [69-71].

Due to inconsistencies observed in studies and potential biases in case-control studies, the World Health Organization (WHO)/IARC classified radiofrequency electromagnetic fields as possibly carcinogenic to humans (Group 2B) in 2011 [72].

OCCUPATIONAL EXPOSURES — A large number of studies have been conducted to examine whether occupational exposures are associated with the risk of brain tumors. Although some positive associations have been reported, there are many inconsistencies in the literature and results are often difficult to interpret given numerous methodologic issues.

Evidence suggests that professionals tend to have a higher risk of brain tumors, although the elevated risk may be a consequence of detection bias due to better access to care [73,74]. Agriculture workers, especially those exposed to herbicides and pesticides, may also have elevated risk of brain tumors; farming as an occupation and place of residence have been associated with a 1.3- to 3.6-fold increased risk of brain tumors [75-80].

Many studies have focused on electrical, rubber, and petroleum workers, where earlier studies had detected strong positive associations; however, the overall findings from these studies do not provide strong support for positive associations. No excess mortality from brain cancer was detectable in a meta-analysis summarizing risk estimates from 20 studies of rubber workers (relative risk [RR] 0.90, 95% CI 0.79-1.02) [81]. Similarly, at least two meta-analyses have found no significant overall increase in brain cancer mortality in petroleum workers [82,83]. Finally, for electrical workers, while a number of studies suggested that individuals in electrical occupations were at increased risk of brain tumors, with relative risks ranging between 1.5 and 2.5 [84-90], other studies did not confirmed these findings [2,4,91].

HEAD TRAUMA — Anecdotal descriptions of brain tumors arising after head trauma date back to the reports of Harvey Cushing in 1922 [92]. A number of other observations have implicated head trauma as a potential risk factor for brain tumors. The evidence is strongest for meningiomas and less convincing for gliomas. (See "Epidemiology, pathology, clinical features, and diagnosis of meningioma", section on 'Others'.)

Evidence for other types of brain tumors includes the following:

In a cohort study of 228,055 Danish residents hospitalized for head injury and followed for an average of eight years, there were increases in the risk of brain tumor with only hemangioblastoma and hemangioma reaching significance (standardized incidence ratio [SIR] = 2.6) [93].

Childhood brain tumors may be more common in firstborn children (higher risk of birth trauma) and in children with documented history of birth trauma (forceps delivery, prolonged labor, cesarean section) [94]. However, other studies of childhood brain tumors have not confirmed an increased risk among these groups [3].

Studies of head trauma and brain tumors may be confounded by ionizing radiation since individuals with a history of head trauma are more likely to have had exposure to ionizing radiation in the form of skull x-rays. Recall bias is another factor complicating interpretation of case-control studies of head trauma and risk of brain tumors [95]. Persons with brain tumors may be more likely to recall minor and major episodes of head injury than controls. As an example, one study reporting an overall positive association between head trauma and the risk of brain tumors found no association when the definition of head trauma was restricted to episodes that required medical attention [96].

ALLERGIES — There is increasing consensus for the inverse association between allergies (eg, asthma, eczema, hay fever) and glioma risk based on consistent findings across different populations and study designs [6,97]. Increased immune surveillance in patients with allergies has been postulated as a possible explanation of these observations, and more research is needed to understand the underlying mechanisms.

This relationship was initially observed in large observational studies [98-101]. A systematic review of the literature and meta-analysis based upon 3450 patients diagnosed with glioma provided further support for this relationship [102]. There was a decrease in the incidence of glioma in patients with a history of any form of allergy (relative risk [RR] 0.61, 95% CI 0.55-0.67). Similar, statistically significant risk reductions were observed when the risk of glioma was analyzed for individuals with either asthma or eczema. In comparison, significant relationship to allergy was not observed among 1070 patients with meningioma.

Several large epidemiologic studies have examined the relationship between serum levels of immunoglobulin E (IgE) and the risk of malignant brain tumors [103-106]. In the largest prospective study to date, samples from 594 blood donors who subsequently developed a glioma (including 374 with glioblastoma) were compared with 1177 paired controls [105]. An inverse relationship was present for total IgE and risk of glioma among males and females; elevated total IgE was associated with a 25 percent decrease in risk of glioma, compared with low total IgE level (odds ratio [OR] 0.75, 95% CI 0.56-0.99, comparing >100 to <100 international units/mL total IgE). Furthermore, the association was present for at least 20 years before the diagnosis of glioma. These findings are consistent with previous studies [103,104,106] and provide strong support for the allergy hypothesis.

DIET — Dietary constituents associated with an increased risk of brain tumors include N-nitroso compounds and possibly dietary fat intake. Aspartame ingestion has been suggested as a possible risk, but there are at present insufficient data to support this claim. Ingestion of antioxidants, fruits, and vegetables may reduce the risk of brain tumors.

N-nitroso compounds — N-nitroso compounds are potent neurocarcinogens in animal models [107]. Human exposure to these agents occurs from both endogenous and exogenous sources.

Exogenous – The major exogenous sources of population exposures to N-nitroso compounds include tobacco smoke, cosmetics, automobile interiors, and cured meats [4]. Other sources include rubber products (baby pacifiers, bottle nipples) and certain drugs including antihistamines, diuretics, oral hypoglycemic agents, antibiotics, tranquilizers, and opiates. N-nitrosodiethanolamine, a carcinogen in animal models, occurs mainly as a contaminant in cosmetic products, soaps, shampoos, and hand lotions.

Endogenous – Endogenous formation of N-nitroso compounds is a complex process that occurs in the stomach and is dependent upon the presence of N-nitroso compound precursors, gastric pH, the presence of bacteria, and other physiologic parameters [3]. Thus, measurement of exposure to endogenous N-nitroso compounds is extremely difficult.

The many epidemiologic studies that have examined the relationship between meat consumption and brain tumors in adults have produced inconsistent results.

In the four largest case-control studies, all of which included over 200 patients diagnosed with glioma and appropriate controls, and some assessment of meat intake [108-111], two reported a significant two- to threefold increased risk of glioma for high consumers of cured meat or bacon, as compared with those with a low intake [109,110]. However, excess risks were only observed among men and, in one, the relative risks were for high intake of cured meat in combination with low fruit and vegetable intake [109]. A meta-analysis that included nine observational studies (primarily case-control studies) reported a relative risk of 1.48 (95% CI 1.20-1.83) for adult glioma among individuals with a high intake of cured meat [112].

Two more recent publications using data from prospective cohort studies found no associations with meat intake or dietary N-nitroso compounds [113,114]. Both studies had over 300 glioma cases and detailed dietary assessment to examine these exposures and their potential relation to glioma risk. The lack of association in these two large prospective studies cast doubt on the N-nitroso compound hypothesis, at least in relation to adult glioma risk.

Antioxidants, fruits, and vegetables — Indirect support for the N-nitroso compound hypothesis includes the observation that certain inhibitors of the nitrosation process, vitamins C and E, appear to reduce brain tumor risk in adults and children [4,96,115]. Dietary studies have demonstrated a reduced risk of brain tumors in children who consume increased amounts of fruits and fruit juices [4]. Prenatal vitamin supplementation (including vitamins A and C and folate) and increased maternal intake of vegetables have been associated with a lowered brain tumor risk in the offspring [115-117].

However, most epidemiologic studies on adult brain tumors contain limited dietary assessment and few questions to explore the relationship between intake of fruits, vegetables, and vitamins and the risk of glioma. Of the nine case-control studies that include data on vitamin C intake from dietary sources and/or supplements, two reported a statistically significant inverse association between intake of supplemental vitamin C and glioma risk [111,118]. In a third report, patients who reported ever using supplemental vitamin C had a relative risk of glioma of 0.2, but this association did not reach the level of statistical significance, and vitamin C intake from foods was unrelated to risk [119]. Others report an interaction between intake of vitamin C and cured foods, such that men with high intake of cured meat who had a low intake of foods rich in vitamin C had a significant twofold increase in glioma risk relative to those with low cured meat intake and high intake of foods rich in vitamin C [109]. Findings were similar in women, although the increased risk (1.5) did not reach statistical significance.

Studies examining vitamin E intake and glioma risk have been inconsistent. A meta-analysis summarizing the evidence from 12 observational studies with a total of 3180 glioma cases reported a 12 percent decrease in risk, but the reduction was not statistically significant (relative risk [RR] 0.88, 95% CI 0.69-1.12) [120]. Further, combined results from two cohort studies with 920 glioma cases showed no association (RR 1.00, 95% CI 0.77-1.31), highlighting potential biases in case-control studies.

As with vitamin C and E intake, data regarding glioma risk and intake of fruits and vegetables are inconsistent. In some studies, a significant inverse association was observed between glioma risk and total fruit and/or vegetable intake [108,121] (which in one study was limited to women only [110]), but three others report no association [119,122,123]. No associations were observed for intake of total or individual fruits and vegetables and the risk of glioma in an analysis of three prospective cohort studies, which included 296 incident glioma cases [124]. In another large prospective study, a positive association was observed for fruit and vegetable intake and glioma risk [113]. As prospective studies are the least prone to recall and selection bias, these findings suggest that fruits and vegetables are unlikely to reduce the risk of glioma.

TOBACCO — Although the presence of nitrosamines in tobacco smoke has stimulated interest in tobacco exposure as a potential risk factor for brain tumors, there is little evidence that either active or passive smoking is a significant risk factor for brain tumors.

Although case control [4,94,125,126] and prospective cohort [127,128] studies have yielded some conflicting results, the majority of studies do not support an association between tobacco exposure and subsequent development of gliomas or meningiomas. A large, contemporary prospective study examining smoking history in detail, including dose, duration, and latency, found no association between smoking and risk of glioma.

Studies looking at the role of active smoking or passive smoking in mothers and the risk of childhood brain tumors have yielded conflicting results [4,115,129].

ALCOHOL — There has been speculation that consumption of alcoholic beverages may increase the risk of brain tumors since both beer and liquor contain nitrosamines. However, no consistent association between consumption of different types of alcoholic beverages and the risk of gliomas or meningiomas in childhood (maternal consumption) or adulthood has been demonstrated [125,130,131].

INFECTION — Most reported associations of infection with brain tumor have been inconsistent. As an example, prior infection with tuberculosis was suggested as a possible risk factor for glioma in one study [132] but not in another [133]. In one large study, subjects who reported a history of infectious diseases (eg, colds, flu), compared with those with none, had a 30 percent reduction in risk (relative risk [RR] = 0.72, 95% CI 0.61-0.85) [98]. Other proposed infectious risk factors have included the polyoma virus, simian virus 40 (SV40), neonatal viral infections, and infection with Toxoplasma gondii. At present, however, there is no compelling epidemiologic evidence establishing infectious agents as important factors in the etiology of brain tumors.

Viral infection — The relationship between antecedent viral infections and subsequent development of brain tumors appears to be complex. Evidence for possible interactions comes from the identification of viruses and virus-like particles in brain tumor specimens, as well as from epidemiologic studies.

Polyomaviruses – Interest in simian virus 40 (SV40), a polyomavirus, was stimulated by animal studies documenting brain tumor development after intracerebral inoculation with SV40 and by human studies that isolated SV40 from brain tumor tissue [134]. It was unclear, however, if SV40 contributed significantly to malignant transformation or whether certain tumors provided a microenvironment that favored replication in patients with latent SV40 infection.

Poliomyelitis vaccine administered between 1955 and 1962 was contaminated with SV40, and vaccination cohorts have been the subject of study over subsequent decades [135-137]. However, elevated brain tumor rates have not been observed in these cohorts. In a nested case-control study, no significant association was reported between antibodies to SV40 (or two other polyomaviruses, JC virus and BK virus) as measured in prediagnostic serum and incident primary malignant brain tumors [138]. Similar null findings were reported for JC and BK viruses in two prospective cohorts, but a different polyomavirus, the Merkel cell polyomavirus, was associated with a higher risk of glioma [139].

CMV – Data concerning a possible etiologic role for cytomegalovirus (CMV) are conflicting and controversial. While several studies have reported that a high percentage of gliomas are infected with CMV [140,141], other groups have not been able to replicate these findings [142-146], or have reported inverse associations for CMV and other herpesviruses (Epstein-Barr virus and herpes simplex virus) [147].

In utero viral exposure – Whether exposure to maternal viral infection while in utero is a risk factor for brain tumors is unclear. A large case-control study found an increased risk (RR = 2.4) for all types of brain tumors after different maternal and perinatal infections [148]. In addition, an association between influenza infection in pregnant women and childhood brain tumors (odds ratio [OR] = 3.15) was suggested in a study in which mothers of 94 children with brain tumors or neuroblastomas and 210 controls were interviewed [146]. However, others have failed to confirm an increased risk of brain tumors in the offspring of mothers infected with varicella, rubella, or mumps during pregnancy [149].

Varicella-zoster – By contrast, a protective role for antecedent infection with varicella-zoster was suggested by an analysis of 229 adults with glioma and 289 controls from the San Francisco Bay Area Adult Glioma Study [150]. Individuals with gliomas were significantly less likely than controls to have a self-reported history of chickenpox (OR 0.59), and they also had lower levels of immunoglobulins directed against varicella-zoster. A similar inverse association was observed for self-report of history of chickenpox in a case-control study of 325 adults with glioma and 600 controls (OR = 0.52) [147]. In the Glioma International Case-Control Study, a history of varicella-zoster virus infection was associated with a 21 percent reduction in risk of glioma, and the association was slightly stronger among high-grade gliomas [97]. Based upon these observations and those for allergies, immune modulation has been postulated to have a role brain tumorigenesis. (See 'Allergies' above.)

Toxoplasma infection — Infection with T. gondii has been associated with an increased risk of astrocytoma and meningioma in two case-control studies [151,152]. In one, a significantly increased risk of meningioma (OR = 2.1), but not glioma, was associated with the presence of immunoglobulin G (IgG) antibodies to T. gondii as measured by ELISA [152]. Although this parasite has a propensity to infect the nervous system, it has not been established as a major risk factor for brain tumors.

SUMMARY

Genetic predisposition syndromes – A small proportion of brain tumors are due to genetic syndromes that confer an increased risk of developing tumors of the nervous system. These include neurofibromatosis type 1 (NF1), NF2-related schwannomatosis (NF2), von Hippel-Lindau syndrome, Li-Fraumeni syndrome (LFS), familial adenomatous polyposis, Lynch syndrome, and the basal cell nevus syndrome. (See 'Genetic predisposition syndromes' above.)

Other genetic factors – Genetic susceptibility has also been noted to play a role in determining risk of brain tumors; a single variant in chromosomal region 8q24.21 has been causally linked to isocitrate dehydrogenase (IDH)-mutant low-grade glioma. (See 'Single nucleotide polymorphisms' above.)

Ionizing radiation – Ionizing radiation is the only firmly established environmental risk factor for brain tumors. Cohort studies of atomic bomb survivors and childhood cancer survivors have demonstrated that cranial radiation is associated with increased risk for a variety of brain tumors, including meningiomas, gliomas, and nerve sheath tumors. (See 'Ionizing radiation' above.)

Nonionizing radiation – The association between forms of nonionizing radiation, such as low-frequency electromagnetic fields and radiofrequency fields, and cancer is less clear, and the data do not support an important role for these types of radiation as risk factors for brain tumors. (See 'Low-frequency electromagnetic fields' above and 'Cellular phones and radiofrequency fields' above.)

Other possible factors – Possible causative factors that require further investigation include allergies, physical and acoustic trauma, and certain infections. (See 'Allergies' above and 'Head trauma' above and 'Infection' above.)

  1. Howlader N, Noone AM, Krapcho M, et al. SEER Cancer Studies Review, 1975-2012, National Cancer Institute, Bethesda 2015.
  2. Polednak AP. Interpretation of secular increases in incidence rates for primary brain cancer in Connecticut adults, 1965-1988. Neuroepidemiology 1996; 15:51.
  3. Davis FG, Preston-Martin S. Epidemiology. In: Russell and Rubinstein's Pathology of Tumors of the Nervous System, 6th ed, Bigner DD, McLendon RE, Bruner JM (Eds), Arnold, London 1998.
  4. Preston-Martin S, Mack WJ. Neoplasms of the nervous system. In: Cancer Epidemiology and Prevention, 2nd ed, Shottenfeld D, Fraumeni JF (Eds), Oxford University Press, New York 1996.
  5. Bondy ML, Lustbader ED, Buffler PA, et al. Genetic epidemiology of childhood brain tumors. Genet Epidemiol 1991; 8:253.
  6. Ostrom QT, Adel Fahmideh M, Cote DJ, et al. Risk factors for childhood and adult primary brain tumors. Neuro Oncol 2019; 21:1357.
  7. Colman SD, Williams CA, Wallace MR. Benign neurofibromas in type 1 neurofibromatosis (NF1) show somatic deletions of the NF1 gene. Nat Genet 1995; 11:90.
  8. Cichowski K, Shih TS, Schmitt E, et al. Mouse models of tumor development in neurofibromatosis type 1. Science 1999; 286:2172.
  9. Vogel KS, Klesse LJ, Velasco-Miguel S, et al. Mouse tumor model for neurofibromatosis type 1. Science 1999; 286:2176.
  10. Rouleau GA, Merel P, Lutchman M, et al. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature 1993; 363:515.
  11. Scoles DR, Huynh DP, Morcos PA, et al. Neurofibromatosis 2 tumour suppressor schwannomin interacts with betaII-spectrin. Nat Genet 1998; 18:354.
  12. Bruder CE, Ichimura K, Blennow E, et al. Severe phenotype of neurofibromatosis type 2 in a patient with a 7.4-MB constitutional deletion on chromosome 22: possible localization of a neurofibromatosis type 2 modifier gene? Genes Chromosomes Cancer 1999; 25:184.
  13. Goutagny S, Kalamarides M. Meningiomas and neurofibromatosis. J Neurooncol 2010; 99:341.
  14. Evans DG, Huson SM, Donnai D, et al. A clinical study of type 2 neurofibromatosis. Q J Med 1992; 84:603.
  15. Perry A, Giannini C, Raghavan R, et al. Aggressive phenotypic and genotypic features in pediatric and NF2-associated meningiomas: a clinicopathologic study of 53 cases. J Neuropathol Exp Neurol 2001; 60:994.
  16. Larson JJ, van Loveren HR, Balko MG, Tew JM Jr. Evidence of meningioma infiltration into cranial nerves: clinical implications for cavernous sinus meningiomas. J Neurosurg 1995; 83:596.
  17. Orr BA, Clay MR, Pinto EM, Kesserwan C. An update on the central nervous system manifestations of Li-Fraumeni syndrome. Acta Neuropathol 2020; 139:669.
  18. Waszak SM, Northcott PA, Buchhalter I, et al. Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort. Lancet Oncol 2018; 19:785.
  19. Watanabe T, Vital A, Nobusawa S, et al. Selective acquisition of IDH1 R132C mutations in astrocytomas associated with Li-Fraumeni syndrome. Acta Neuropathol 2009; 117:653.
  20. Hayes J, Yu Y, Jalbert LE, et al. Genomic analysis of the origins and evolution of multicentric diffuse lower-grade gliomas. Neuro Oncol 2018; 20:632.
  21. Hamilton SR, Liu B, Parsons RE, et al. The molecular basis of Turcot's syndrome. N Engl J Med 1995; 332:839.
  22. Benusiglio PR, Elder F, Touat M, et al. Mismatch Repair Deficiency and Lynch Syndrome Among Adult Patients With Glioma. JCO Precis Oncol 2023; 7:e2200525.
  23. Stark Z, Campbell LJ, Mitchell C, et al. Clinical problem-solving. Spot diagnosis. N Engl J Med 2014; 370:2229.
  24. Bruwer Z, Algar U, Vorster A, et al. Predictive genetic testing in children: constitutional mismatch repair deficiency cancer predisposing syndrome. J Genet Couns 2014; 23:147.
  25. Bouffet E, Larouche V, Campbell BB, et al. Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair Deficiency. J Clin Oncol 2016; 34:2206.
  26. Wrensch M, Lee M, Miike R, et al. Familial and personal medical history of cancer and nervous system conditions among adults with glioma and controls. Am J Epidemiol 1997; 145:581.
  27. Malmer B, Grönberg H, Bergenheim AT, et al. Familial aggregation of astrocytoma in northern Sweden: an epidemiological cohort study. Int J Cancer 1999; 81:366.
  28. Blumenthal DT, Cannon-Albright LA. Familiality in brain tumors. Neurology 2008; 71:1015.
  29. Malmer B, Iselius L, Holmberg E, et al. Genetic epidemiology of glioma. Br J Cancer 2001; 84:429.
  30. Sadetzki S, Bruchim R, Oberman B, et al. Description of selected characteristics of familial glioma patients - results from the Gliogene Consortium. Eur J Cancer 2013; 49:1335.
  31. Bainbridge MN, Armstrong GN, Gramatges MM, et al. Germline mutations in shelterin complex genes are associated with familial glioma. J Natl Cancer Inst 2015; 107:384.
  32. Robles-Espinoza CD, Harland M, Ramsay AJ, et al. POT1 loss-of-function variants predispose to familial melanoma. Nat Genet 2014; 46:478.
  33. Shi J, Yang XR, Ballew B, et al. Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma. Nat Genet 2014; 46:482.
  34. Shete S, Hosking FJ, Robertson LB, et al. Genome-wide association study identifies five susceptibility loci for glioma. Nat Genet 2009; 41:899.
  35. Wrensch M, Jenkins RB, Chang JS, et al. Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility. Nat Genet 2009; 41:905.
  36. Rajaraman P, Melin BS, Wang Z, et al. Genome-wide association study of glioma and meta-analysis. Hum Genet 2012; 131:1877.
  37. Sanson M, Hosking FJ, Shete S, et al. Chromosome 7p11.2 (EGFR) variation influences glioma risk. Hum Mol Genet 2011; 20:2897.
  38. Melin BS, Barnholtz-Sloan JS, Wrensch MR, 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.
  39. Jenkins RB, Xiao Y, Sicotte H, 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.
  40. Yanchus C, Drucker KL, Kollmeyer TM, et al. A noncoding single-nucleotide polymorphism at 8q24 drives IDH1-mutant glioma formation. Science 2022; 378:68.
  41. Dobbins SE, Broderick P, Melin B, et al. Common variation at 10p12.31 near MLLT10 influences meningioma risk. Nat Genet 2011; 43:825.
  42. Claus EB, Cornish AJ, Broderick P, et al. Genome-wide association analysis identifies a meningioma risk locus at 11p15.5. Neuro Oncol 2018; 20:1485.
  43. Gu J, Liu Y, Kyritsis AP, Bondy ML. Molecular epidemiology of primary brain tumors. Neurotherapeutics 2009; 6:427.
  44. Jiang J, Quan XF, Zhang L, Wang YC. The XRCC3 Thr241Met polymorphism influences glioma risk - a meta-analysis. Asian Pac J Cancer Prev 2013; 14:3169.
  45. Backes DM, Siddiq A, Cox DG, et al. Single-nucleotide polymorphisms of allergy-related genes and risk of adult glioma. J Neurooncol 2013; 113:229.
  46. Schwartzbaum J, Ahlbom A, Malmer B, et al. Polymorphisms associated with asthma are inversely related to glioblastoma multiforme. Cancer Res 2005; 65:6459.
  47. Amirian E, Liu Y, Scheurer ME, et al. Genetic variants in inflammation pathway genes and asthma in glioma susceptibility. Neuro Oncol 2010; 12:444.
  48. Wiemels JL, Wiencke JK, Kelsey KT, et al. Allergy-related polymorphisms influence glioma status and serum IgE levels. Cancer Epidemiol Biomarkers Prev 2007; 16:1229.
  49. Michaud DS, Siddiq A, Cox DG, et al. Mannose-binding lectin 2 gene and risk of adult glioma. PLoS One 2013; 8:e61117.
  50. Braganza MZ, Kitahara CM, Berrington de González A, et al. Ionizing radiation and the risk of brain and central nervous system tumors: A systematic review. Neuro Oncol 2012; 14:1316.
  51. Bowers DC, Nathan PC, Constine L, et al. Subsequent neoplasms of the CNS among survivors of childhood cancer: A systematic review. Lancet Oncol 2013; 14:e321.
  52. Neglia JP, Robison LL, Stovall M, et al. New primary neoplasms of the central nervous system in survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Natl Cancer Inst 2006; 98:1528.
  53. Relling MV, Rubnitz JE, Rivera GK, et al. High incidence of secondary brain tumours after radiotherapy and antimetabolites. Lancet 1999; 354:34.
  54. Pearce MS, Salotti JA, Little MP, et al. Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet 2012; 380:499.
  55. Davis F, Il'yasova D, Rankin K, et al. Medical diagnostic radiation exposures and risk of gliomas. Radiat Res 2011; 175:790.
  56. NIEHS. Health effects from exposure to power-line frequency electric and magnetic fields. NIH publication no 99-4493, National Institute of Environmental Health Sciences; 1999.
  57. Non-ionizing radiation, part 1: Static and extremely low-frequency (ELF) electric and magnetic fields. In: IARC Monographs on the Identification of Carcinogenic Hazards to Humans, International Agency for Research on Cancer, Lyon, France 2002. p.80.
  58. Kheifets LI, Sussman SS, Preston-Martin S. Childhood brain tumors and residential electromagnetic fields (EMF). Rev Environ Contam Toxicol 1999; 159:111.
  59. Preston-Martin S, Navidi W, Thomas D, et al. Los Angeles study of residential magnetic fields and childhood brain tumors. Am J Epidemiol 1996; 143:105.
  60. Gurney JG, Mueller BA, Davis S, et al. Childhood brain tumor occurrence in relation to residential power line configurations, electric heating sources, and electric appliance use. Am J Epidemiol 1996; 143:120.
  61. Sorahan T, Hamilton L, Gardiner K, et al. Maternal occupational exposure to electromagnetic fields before, during, and after pregnancy in relation to risks of childhood cancers: findings from the Oxford Survey of Childhood Cancers, 1953-1981 deaths. Am J Ind Med 1999; 35:348.
  62. Coble JB, Dosemeci M, Stewart PA, et al. Occupational exposure to magnetic fields and the risk of brain tumors. Neuro Oncol 2009; 11:242.
  63. Turner MC, Benke G, Bowman JD, et al. Occupational exposure to extremely low-frequency magnetic fields and brain tumor risks in the INTEROCC study. Cancer Epidemiol Biomarkers Prev 2014; 23:1863.
  64. Rothman KJ, Chou CK, Morgan R, et al. Assessment of cellular telephone and other radio frequency exposure for epidemiologic research. Epidemiology 1996; 7:291.
  65. Myung SK, Ju W, McDonnell DD, et al. Mobile phone use and risk of tumors: a meta-analysis. J Clin Oncol 2009; 27:5565.
  66. Corle C, Makale M, Kesari S. Cell phones and glioma risk: a review of the evidence. J Neurooncol 2012; 106:1.
  67. Schuermann D, Mevissen M. Manmade Electromagnetic Fields and Oxidative Stress-Biological Effects and Consequences for Health. Int J Mol Sci 2021; 22.
  68. Schüz J, Pirie K, Reeves GK, et al. Cellular Telephone Use and the Risk of Brain Tumors: Update of the UK Million Women Study. J Natl Cancer Inst 2022; 114:704.
  69. Little MP, Rajaraman P, Curtis RE, et al. Mobile phone use and glioma risk: comparison of epidemiological study results with incidence trends in the United States. BMJ 2012; 344:e1147.
  70. Deltour I, Auvinen A, Feychting M, et al. Mobile phone use and incidence of glioma in the Nordic countries 1979-2008: consistency check. Epidemiology 2012; 23:301.
  71. Elwood JM, Win SS, Aye PS, Sanagou M. Trends in brain cancers (glioma) in New Zealand from 1995 to 2020, with reference to mobile phone use. Cancer Epidemiol 2022; 80:102234.
  72. IARC monographs on the evaluation of carcinogenic risks to humans, IARC, Lyon 2013. Vol 102.
  73. Preston-Martin S. Descriptive epidemiology of primary tumors of the brain, cranial nerves and cranial meninges in Los Angeles County. Neuroepidemiology 1989; 8:283.
  74. McLaughlin JK, Malker HS, Blot WJ, et al. Occupational risks for intracranial gliomas in Sweden. J Natl Cancer Inst 1987; 78:253.
  75. Wong O, Raabe GK. Critical review of cancer epidemiology in petroleum industry employees, with a quantitative meta-analysis by cancer site. Am J Ind Med 1989; 15:283.
  76. Musicco M, Sant M, Molinari S, et al. A case-control study of brain gliomas and occupational exposure to chemical carcinogens: the risk to farmers. Am J Epidemiol 1988; 128:778.
  77. Littorin M, Attewell R, Skerfving S, et al. Mortality and tumour morbidity among Swedish market gardeners and orchardists. Int Arch Occup Environ Health 1993; 65:163.
  78. Heineman EF, Gao YT, Dosemeci M, McLaughlin JK. Occupational risk factors for brain tumors among women in Shanghai, China. J Occup Environ Med 1995; 37:288.
  79. Khuder SA, Mutgi AB, Schaub EA. Meta-analyses of brain cancer and farming. Am J Ind Med 1998; 34:252.
  80. Piel C, Pouchieu C, Tual S, et al. Central nervous system tumors and agricultural exposures in the prospective cohort AGRICAN. Int J Cancer 2017; 141:1771.
  81. Borak J, Slade MD, Russi M. Risks of brain tumors in rubber workers: a metaanalysis. J Occup Environ Med 2005; 47:294.
  82. International Agency for Research on Cancer. Occupational exposures in petroleum refining; crude oil and major petroleum fuels. In: IARC Monographs on the evaluation of carcinogenic risks to humans, IARC, Lyon 1989. Vol 45.
  83. Wong O, Raabe GK. A critical review of cancer epidemiology in the petroleum industry, with a meta-analysis of a combined database of more than 350,000 workers. Regul Toxicol Pharmacol 2000; 32:78.
  84. Lin RS, Dischinger PC, Conde J, Farrell KP. Occupational exposure to electromagnetic fields and the occurrence of brain tumors. An analysis of possible associations. J Occup Med 1985; 27:413.
  85. Thomas TL, Stolley PD, Stemhagen A, et al. Brain tumor mortality risk among men with electrical and electronics jobs: a case-control study. J Natl Cancer Inst 1987; 79:233.
  86. Loomis DP, Savitz DA. Mortality from brain cancer and leukaemia among electrical workers. Br J Ind Med 1990; 47:633.
  87. Juutilainen J, Läärä E, Pukkala E. Incidence of leukaemia and brain tumours in Finnish workers exposed to ELF magnetic fields. Int Arch Occup Environ Health 1990; 62:289.
  88. Schlehofer B, Kunze S, Sachsenheimer W, et al. Occupational risk factors for brain tumors: results from a population-based case-control study in Germany. Cancer Causes Control 1990; 1:209.
  89. Bates MN. Extremely low frequency electromagnetic fields and cancer: the epidemiologic evidence. Environ Health Perspect 1991; 95:147.
  90. Savitz DA, Loomis DP. Magnetic field exposure in relation to leukemia and brain cancer mortality among electric utility workers. Am J Epidemiol 1995; 141:123.
  91. Sahl JD, Kelsh MA, Greenland S. Cohort and nested case-control studies of hematopoietic cancers and brain cancer among electric utility workers. Epidemiology 1993; 4:104.
  92. Cushing H. The meningiomas (dural endotheliomas). Their source and favored seats of origin. Brain 1922; 45:282.
  93. Inskip PD, Mellemkjaer L, Gridley G, Olsen JH. Incidence of intracranial tumors following hospitalization for head injuries (Denmark). Cancer Causes Control 1998; 9:109.
  94. Choi NW, Schuman LM, Gullen WH. Epidemiology of primary central nervous system neoplasms. II. Case-control study. Am J Epidemiol 1970; 91:467.
  95. Wrensch M, Miike R, Lee M, Neuhaus J. Are prior head injuries or diagnostic X-rays associated with glioma in adults? The effects of control selection bias. Neuroepidemiology 2000; 19:234.
  96. Burch JD, Craib KJ, Choi BC, et al. An exploratory case-control study of brain tumors in adults. J Natl Cancer Inst 1987; 78:601.
  97. Amirian ES, Zhou R, Wrensch MR, et al. Approaching a Scientific Consensus on the Association between Allergies and Glioma Risk: A Report from the Glioma International Case-Control Study. Cancer Epidemiol Biomarkers Prev 2016; 25:282.
  98. Schlehofer B, Blettner M, Preston-Martin S, et al. Role of medical history in brain tumour development. Results from the international adult brain tumour study. Int J Cancer 1999; 82:155.
  99. Brenner AV, Linet MS, Fine HA, et al. History of allergies and autoimmune diseases and risk of brain tumors in adults. Int J Cancer 2002; 99:252.
  100. Wiemels JL, Wiencke JK, Sison JD, et al. History of allergies among adults with glioma and controls. Int J Cancer 2002; 98:609.
  101. Schoemaker MJ, Swerdlow AJ, Hepworth SJ, et al. History of allergies and risk of glioma in adults. Int J Cancer 2006; 119:2165.
  102. Linos E, Raine T, Alonso A, Michaud D. Atopy and risk of brain tumors: a meta-analysis. J Natl Cancer Inst 2007; 99:1544.
  103. Wiemels JL, Wiencke JK, Patoka J, et al. Reduced immunoglobulin E and allergy among adults with glioma compared with controls. Cancer Res 2004; 64:8468.
  104. Calboli FC, Cox DG, Buring JE, et al. Prediagnostic plasma IgE levels and risk of adult glioma in four prospective cohort studies. J Natl Cancer Inst 2011; 103:1588.
  105. Schwartzbaum J, Ding B, Johannesen TB, et al. Association between prediagnostic IgE levels and risk of glioma. J Natl Cancer Inst 2012; 104:1251.
  106. Schlehofer B, Siegmund B, Linseisen J, et al. Primary brain tumours and specific serum immunoglobulin E: a case-control study nested in the European Prospective Investigation into Cancer and Nutrition cohort. Allergy 2011; 66:1434.
  107. National Research Council. The health effects of nitrate, nitrite, and N-nitroso compounds. Part 1. National Academy Press; Washington DC.
  108. Chen H, Ward MH, Tucker KL, et al. Diet and risk of adult glioma in eastern Nebraska, United States. Cancer Causes Control 2002; 13:647.
  109. Lee M, Wrensch M, Miike R. Dietary and tobacco risk factors for adult onset glioma in the San Francisco Bay Area (California, USA). Cancer Causes Control 1997; 8:13.
  110. Giles GG, McNeil JJ, Donnan G, et al. Dietary factors and the risk of glioma in adults: results of a case-control study in Melbourne, Australia. Int J Cancer 1994; 59:357.
  111. Preston-Martin S, Mack W. Gliomas and meningiomas in men in Los Angeles County: investigation of exposures to N-nitroso compounds. IARC Sci Publ 1991; :197.
  112. Huncharek M, Kupelnick B, Wheeler L. Dietary cured meat and the risk of adult glioma: a meta-analysis of nine observational studies. J Environ Pathol Toxicol Oncol 2003; 22:129.
  113. Dubrow R, Darefsky AS, Park Y, et al. Dietary components related to N-nitroso compound formation: a prospective study of adult glioma. Cancer Epidemiol Biomarkers Prev 2010; 19:1709.
  114. Michaud DS, Holick CN, Batchelor TT, et al. Prospective study of meat intake and dietary nitrates, nitrites, and nitrosamines and risk of adult glioma. Am J Clin Nutr 2009; 90:570.
  115. McCredie M, Maisonneuve P, Boyle P. Perinatal and early postnatal risk factors for malignant brain tumours in New South Wales children. Int J Cancer 1994; 56:11.
  116. Preston-Martin S, Pogoda JM, Mueller BA, et al. Prenatal vitamin supplementation and risk of childhood brain tumors. Int J Cancer Suppl 1998; 11:17.
  117. Bunin GR, Kuijten RR, Buckley JD, et al. Relation between maternal diet and subsequent primitive neuroectodermal brain tumors in young children. N Engl J Med 1993; 329:536.
  118. Ahlbom A, Navier IL, Norell S, et al. Nonoccupational risk indicators for astrocytomas in adults. Am J Epidemiol 1986; 124:334.
  119. Blowers L, Preston-Martin S, Mack WJ. Dietary and other lifestyle factors of women with brain gliomas in Los Angeles County (California, USA). Cancer Causes Control 1997; 8:5.
  120. Qin S, Wang M, Zhang T, Zhang S. Vitamin E intake is not associated with glioma risk: evidence from a meta-analysis. Neuroepidemiology 2014; 43:253.
  121. Hu J, La Vecchia C, Negri E, et al. Diet and brain cancer in adults: a case-control study in northeast China. Int J Cancer 1999; 81:20.
  122. Boeing H, Schlehofer B, Blettner M, Wahrendorf J. Dietary carcinogens and the risk for glioma and meningioma in Germany. Int J Cancer 1993; 53:561.
  123. Kaplan S, Novikov I, Modan B. Nutritional factors in the etiology of brain tumors: potential role of nitrosamines, fat, and cholesterol. Am J Epidemiol 1997; 146:832.
  124. Holick CN, Giovannucci EL, Rosner B, et al. Prospective study of intake of fruit, vegetables, and carotenoids and the risk of adult glioma. Am J Clin Nutr 2007; 85:877.
  125. Hurley SF, McNeil JJ, Donnan GA, et al. Tobacco smoking and alcohol consumption as risk factors for glioma: a case-control study in Melbourne, Australia. J Epidemiol Community Health 1996; 50:442.
  126. Filippini G, Maisonneuve P, McCredie M, et al. Relation of childhood brain tumors to exposure of parents and children to tobacco smoke: the SEARCH international case-control study. Surveillance of Environmental Aspects Related to Cancer in Humans. Int J Cancer 2002; 100:206.
  127. Silvera SA, Miller AB, Rohan TE. Cigarette smoking and risk of glioma: a prospective cohort study. Int J Cancer 2006; 118:1848.
  128. Holick CN, Giovannucci EL, Rosner B, et al. Prospective study of cigarette smoking and adult glioma: dosage, duration, and latency. Neuro Oncol 2007; 9:326.
  129. Milne E, Greenop KR, Scott RJ, et al. Parental smoking and risk of childhood brain tumors. Int J Cancer 2013; 133:253.
  130. Preston-Martin S, Yu MC, Henderson BE, Roberts C. Risk factors for meningiomas in men in Los Angeles County. J Natl Cancer Inst 1983; 70:863.
  131. Galeone C, Malerba S, Rota M, et al. A meta-analysis of alcohol consumption and the risk of brain tumours. Ann Oncol 2013; 24:514.
  132. Ward DW, Mattison ML, Finn R. Association between Previous Tuberculous Infection and Cerebal Glioma. Br Med J 1973; 1:83.
  133. Davis FG, Boice JD Jr, Hrubec Z, Monson RR. Cancer mortality in a radiation-exposed cohort of Massachusetts tuberculosis patients. Cancer Res 1989; 49:6130.
  134. Huang H, Reis R, Yonekawa Y, et al. Identification in human brain tumors of DNA sequences specific for SV40 large T antigen. Brain Pathol 1999; 9:33.
  135. Strickler HD, Rosenberg PS, Devesa SS, et al. Contamination of poliovirus vaccines with simian virus 40 (1955-1963) and subsequent cancer rates. JAMA 1998; 279:292.
  136. Engels EA, Katki HA, Nielsen NM, et al. Cancer incidence in Denmark following exposure to poliovirus vaccine contaminated with simian virus 40. J Natl Cancer Inst 2003; 95:532.
  137. Strickler HD, Goedert JJ. Exposure to SV40-contaminated poliovirus vaccine and the risk of cancer--a review of the epidemiological evidence. Dev Biol Stand 1998; 94:235.
  138. Rollison DE, Helzlsouer KJ, Alberg AJ, et al. Serum antibodies to JC virus, BK virus, simian virus 40, and the risk of incident adult astrocytic brain tumors. Cancer Epidemiol Biomarkers Prev 2003; 12:460.
  139. Arzuza O, García-Villalón D, Tabarés E, et al. Inhibition of African swine fever virus DNA synthesis by (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl)adenine. Biochem Biophys Res Commun 1988; 154:27.
  140. Cobbs CS, Harkins L, Samanta M, et al. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Res 2002; 62:3347.
  141. Mitchell DA, Xie W, Schmittling R, et al. Sensitive detection of human cytomegalovirus in tumors and peripheral blood of patients diagnosed with glioblastoma. Neuro Oncol 2008; 10:10.
  142. Sabatier J, Uro-Coste E, Pommepuy I, et al. Detection of human cytomegalovirus genome and gene products in central nervous system tumours. Br J Cancer 2005; 92:747.
  143. Lau SK, Chen YY, Chen WG, et al. Lack of association of cytomegalovirus with human brain tumors. Mod Pathol 2005; 18:838.
  144. Holdhoff M, Guner G, Rodriguez FJ, et al. Absence of Cytomegalovirus in Glioblastoma and Other High-grade Gliomas by Real-time PCR, Immunohistochemistry, and In Situ Hybridization. Clin Cancer Res 2017; 23:3150.
  145. Taha MS, Abdalhamid BA, El-Badawy SA, et al. Expression of cytomegalovirus in glioblastoma multiforme: Myth or reality? Br J Neurosurg 2016; 30:307.
  146. Linos A, Kardara M, Kosmidis H, et al. Reported influenza in pregnancy and childhood tumour. Eur J Epidemiol 1998; 14:471.
  147. Scheurer ME, El-Zein R, Thompson PA, et al. Long-term anti-inflammatory and antihistamine medication use and adult glioma risk. Cancer Epidemiol Biomarkers Prev 2008; 17:1277.
  148. Linet MS, Gridley G, Cnattingius S, et al. Maternal and perinatal risk factors for childhood brain tumors (Sweden). Cancer Causes Control 1996; 7:437.
  149. Adelstein AM, Donovan JW. Malignant disease in children whose mothers had chickenpox, mumps, or rubella in pregnancy. Br Med J 1972; 4:629.
  150. Wrensch M, Weinberg A, Wiencke J, et al. History of chickenpox and shingles and prevalence of antibodies to varicella-zoster virus and three other herpesviruses among adults with glioma and controls. Am J Epidemiol 2005; 161:929.
  151. Schuman LM, Choi NW, Gullen WH. Relationship of central nervous system neoplasms to Toxoplasma gondii infection. Am J Public Health Nations Health 1967; 57:848.
  152. Ryan P, Hurley SF, Johnson AM, et al. Tumours of the brain and presence of antibodies to Toxoplasma gondii. Int J Epidemiol 1993; 22:412.
Topic 5214 Version 37.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟