ArticlePDF Available

Epidemiology, risk factors, and prognostic factors of gliomas

Authors:

Abstract

PurposeDiffuse gliomas are the most common primary malignant brain tumors in adults. Many studies analyzed their epidemiology, such as the incidence and mortality. Moreover, the identification of their risk factors is still controversial and to date we have only a few accepted and confirmed risk factors. In the last few years, many molecular markers have been analyzed and correlated to the prognosis of gliomas.We performed a review aiming to collect and clarify data on epidemiology, risk factors and prognostic factors regarding glioma patients.Methods We performed a comprehensive literature review of research studies focusing on epidemiology of gliomas and their risk factors. At the same time, we collected studies analyzing the most important and validated prognostic factors in glioma patients.ResultsGlioblastoma represents the most common primary malignant brain tumor with an incidence rate of 3.23 per 100,000 population. Diffuse astrocytoma and oligodendroglioma tend to peak in young adults with a median age of 46 and 43 years, respectively). Overall, the incidence rate of gliomas is higher in male patients than in females. Rates of overall survival vary widely, ranging from 5 year survival rates of 94.7% for pilocytic astrocytoma to 6.8% for glioblastoma. About 5% of gliomas can be classified as familial and associated with hereditary syndromes, such as the Li-Fraumeni, the Turcot and neurofibromatosis. Ionizing radiation remains the only ascertained environmental risk factor associated with glioma. In addition to the clinical characteristics, such as the age, performance status and the extent of resection, also mutational status of some genes such as IDH, TERT, CDKN2A and the MGMT methylation status can be correlated with the glioma patient survival.Conclusions Gliomas represent rare tumors and can be defined as a heterogenous group of primitive brain tumors. In recent years, new data emerged regarding the etiology of these tumors as well as the knowledge of new prognostic factors.
Vol.:(0123456789)
1 3
Clinical and Translational Imaging
https://doi.org/10.1007/s40336-022-00489-6
EXPERT REVIEW
Epidemiology, risk factors, andprognostic factors ofgliomas
AlessiaPellerino1· MarioCaccese2,3· MartaPadovan2,3· GiuliaCerretti2,4· GiuseppeLombardi2
Received: 27 January 2022 / Accepted: 4 March 2022
© The Author(s), under exclusive licence to Italian Association of Nuclear Medicine and Molecular Imaging 2022
Abstract
Purpose Diffuse gliomas are the most common primary malignant brain tumors in adults. Many studies analyzed their
epidemiology, such as the incidence and mortality. Moreover, the identification of their risk factors is still controversial and
to date we have only a few accepted and confirmed risk factors. In the last few years, many molecular markers have been
analyzed and correlated to the prognosis of gliomas.
We performed a review aiming to collect and clarify data on epidemiology, risk factors and prognostic factors regarding
glioma patients.
Methods We performed a comprehensive literature review of research studies focusing on epidemiology of gliomas and
their risk factors. At the same time, we collected studies analyzing the most important and validated prognostic factors in
glioma patients.
Results Glioblastoma represents the most common primary malignant brain tumor with an incidence rate of 3.23 per 100,000
population. Diffuse astrocytoma and oligodendroglioma tend to peak in young adults with a median age of 46 and 43years,
respectively). Overall, the incidence rate of gliomas is higher in male patients than in females. Rates of overall survival vary
widely, ranging from 5year survival rates of 94.7% for pilocytic astrocytoma to 6.8% for glioblastoma. About 5% of gliomas
can be classified as familial and associated with hereditary syndromes, such as the Li-Fraumeni, the Turcot and neurofi-
bromatosis. Ionizing radiation remains the only ascertained environmental risk factor associated with glioma. In addition to
the clinical characteristics, such as the age, performance status and the extent of resection, also mutational status of some
genes such as IDH, TERT, CDKN2A and the MGMT methylation status can be correlated with the glioma patient survival.
Conclusions Gliomas represent rare tumors and can be defined as a heterogenous group of primitive brain tumors. In recent
years, new data emerged regarding the etiology of these tumors as well as the knowledge of new prognostic factors.
Keywords Glioma· Epidemiology· Risk factors· Prognostic factors
Introduction
Diffuse gliomas are the most common primary malignant
brain tumors. The typical features of gliomas, such as high
malignancy, significant mortality rate, and increased risk of
recurrence, represent a challenge for neuro-oncologists and
are a considerable burden on society and families.
However, in the last few years, new studies analyzing
potential risk factors and new molecular markers were
performed.
In this work, we reviewed recent papers focusing on epi-
demiology, risk factors and prognostic factors regarding
glioma patients.
Alessia Pellerino, Mario Caccese, Marta Padovan, Giulia Cerretti
and Giuseppe Lombardi have contributed equally to this work.
* Giuseppe Lombardi
giuseppe.lombardi@iov.veneto.it
1 Department ofNeuro-Oncology, University andCity
ofHealth andScience Hospital, via Cherasco 15,
10126Turin, Italy
2 Department ofOncology, Oncology 1, Veneto Institute
ofOncology IOV-IRCCS, Via Gattamelata 64, 35128Padua,
Italy
3 Clinical andExperimental Oncology andImmunology
PhD Program, Department ofSurgery, Oncology
andGastroenterology, University ofPadua, 35128Padua,
Italy
4 Department ofSurgery, Oncology andGastroenterology
(DiSCOG), University ofPadua, 35128Padua, Italy
Clinical and Translational Imaging
1 3
Methods
We performed a literature review and the most important
studies on epidemiology, risk factors and prognostic fac-
tors on gliomas were identified by searching the Medline
electronic database. The search strategy included terms
used to describe gliomas, their etiology and clinical and
molecular factors impacting on survival. For further rel-
evant studies this search was supplemented by reviewing
the bibliographies of key papers.
Epidemiology
The incidence and mortality rate of gliomas vary accord-
ing to age, sex, race, and geographical region. Overall,
the broad category of gliomas (ICD-O-3 histology codes
9380–9384 and 9391–9460) represents approximately
24.5% of all primary brain tumors and 80.9% of all malig-
nant tumors in adults. Sixty-two percent of gliomas occur
in the supratentorial compartment: 27.0% in the frontal
lobe, 20.2% in the temporal lobe, 11.6% in the parietal
lobe, and 2.8% in the occipital lobe. A small proportion
may occur in other CNS sites, including the brainstem
(4.3%), spinal cord and cauda equina (4.0%), the cerebel-
lum (2.8%), and other brain sites (20.0%) [1]. Based on the
United States’ Central Brain Tumor Registry (CBTRUS)
report, astrocytic tumors, including glioblastoma, account
for 77.5% of all gliomas. Glioblastoma prevails among
malignant gliomas (58.4%), followed by diffuse astrocy-
toma (7.3%), anaplastic astrocytoma (6.8%), oligoden-
droglioma (3.5%), anaplastic oligodendroglioma (1.7%),
pilocytic astrocytoma (5.0%), and malignant gliomas not
otherwise specified (NOS) (7.9%). The histology group-
ing used in this report is based on the 2016 WHO Clas-
sification of CNS Tumors and certain data will be updated
accordingly once the 2021 WHO Classification is released
[2].
The incidence rate in the adult population is highest
for glioblastoma (3.23 per 100,000 population), followed
by diffuse astrocytoma and anaplastic astrocytoma (0.46
and 0.42 per 100,000 population, respectively), and oligo-
dendroglioma and anaplastic oligodendroglioma (0.23 and
0.11 per 100,000 population, respectively).
Diffuse astrocytoma and oligodendroglioma tend to
peak in young adults (median age of 46 and 43years,
respectively), while anaplastic astrocytoma and oligoden-
droglioma occur approximately 5–9years later (median
age of 53 and 49years, respectively). Glioblastoma is
the most common tumor in adult and elderly patients
(median age of 65years), while it is a rare entity in pedi-
atric patients. In fact, gliomas account for 45% of all CNS
malignant pediatric tumors. Diffuse midline glioma rep-
resents 31.1% of all childhood gliomas [3, 4], followed
by pilocytic astrocytoma (18.3%), diffuse astrocytoma and
anaplastic astrocytoma (5.3%), and glioblastoma (2.6%).
In the population aged 0–19years, the incidence rate for
diffuse midline gliomas is 0.31 per 100,000 population
[3], followed by diffuse astrocytoma (0.23 per 100,000
population), and glioblastoma (0.17 per 100,000 popula-
tion). Anaplastic astrocytoma, oligodendrogliomas, and
anaplastic oligodendrogliomas are all rare entities (0.09,
0.04, and 0.01 per 100,000 population, respectively) [1].
Overall, the incidence rate of gliomas is higher in males
(5.51 per 100,000 population) than in females (3.65 per
100,000 population), except for diffuse midline gliomas,
which prevail among females (0.324 versus 0.288) [3].
The incidence of gliomas may be influenced by ethnic-
ity. The incidence rate is about 2times higher in American
and northern European populations than in the Asian pop-
ulation [5]. Furthermore, from 2000 to 2014, glioblastoma
accounted for 80% of astrocytic tumors in the 40–99year
age group in North America, Europe, and Oceania, but
only 60% or less in Central and South America [6]. Epide-
miological studies have shown a different incidence trend
based on geographic regions. In this regard, a Japanese
study found that the incidence rate of malignant gliomas
in the elderly increased significantly between 1998 and
2008 [7], whereas the CBTRUS registry reported that
the incidence of glioma in patients 40years remained
relatively stable between 2000 and 2016 [8]. Recently, the
CONCORD-3 population-based study found that the pro-
portion of glioblastomas only rose in Europe (from 46 to
56%) and Oceania (from 57 to 65%) from 2000 to 2014,
while increasing trends for glioma NOS were reported in
Central and South America, due to disparities in access
to cancer registries and the lack of quality in neuropatho-
logical diagnostic workup for the identification of different
glioma subtypes [6].
Incidence rates for glioblastoma and diffuse midline
glioma are remarkably higher in non-Hispanic Whites than
in Blacks and Hispanics, suggesting that some genetic risk
susceptibility or environmental factors are more frequent
in populations of predominantly European ancestry [3, 8].
However, the aging population is closely linked to industri-
alized countries, such as Europe and North America, and
could partly explain the increased incidence of glioblastoma
in these regions. Other factors, including a higher socioeco-
nomic status, easier access to the health care system, and the
improvement of diagnostic tools and neuroimaging allow for
early detection in more asymptomatic or mildly symptomatic
patients, contributing to a rapid increase in the incidence of
glioma of 2.9% per year registered between 1978 and 1992
[9]. Glioma has been on the decline since 1987, with an
increase of 0.2% per year occurring only after 1992.
Clinical and Translational Imaging
1 3
Rates of overall survival (OS) vary widely, ranging from
5year OS rates of 94.7% for pilocytic astrocytoma to 6.8%
for glioblastoma, depending upon different factors that may
influence glioma patient survival [9]. Treatment patterns, the
ability to access the health care system, the type of facilities,
where patients receive treatments, physicians’ expertise, and
the availability of clinical trials are some of the potential
factors that may influence patient outcomes. Importantly,
glioma histological and molecular profiles, including the
assessment of IDH 1–2 mutations, 1p19q codeletion, TERT
mutations, EGFR amplification, the gain of chromosome
7 and loss of chromosome 10, CDKN2A/B deletions, and
MGMT methylation status, help neuro-oncologists iden-
tify patients with different outcomes and to tailor their
treatments.
Risk factors
Although gliomas are considered rare in terms of incidence
[1], they represent one of the greatest challenges in the
oncology field due to the burden of the high level of care
required by patients and poor survival. The identification
of risk factors for the onset of gliomas could be extremely
useful for the purposes of early diagnosis and prevention, but
to date, data available in this regard are controversial, with
only a few accepted and confirmed risk factors.
Genetic risk factors
Most gliomas develop without a family history, but a small
percentage (5%) can be classified as familial [10], with an
even lower percentage (1–2%) transmitted in a Mendelian
pattern or as part of hereditary syndromes [1, 11]. The Li-
Fraumeni syndrome, Turcot syndrome, and neurofibrama-
tosis type 1 disorders are now known to be associated with
the highest risk of developing glioma [11]. Several genome-
wide association studies have explored the presence of many
genomic variants linked to an increased risk of developing
brain tumors, particularly malignant gliomas [12]. There
are 25 known genomic susceptibility variants for gliomas in
adults and the proportion of glioma incidence variance that
can be attributed to genetic factors is 25%, with about 30%
of this being explained by the variants identified by these
studies. Approximately 70% of the remaining genetic risk is
currently unexplained [12, 13]. In addition to the presence
of these variants, however, additional somatic mutations
are required for gliomatous tumorigenesis [14, 15]. Neu-
rofibromatosis type 1 (NF1) is an autosomal dominant syn-
drome of the NF1 gene and is mainly associated with brain-
stem astrocytoma, pilocytic astrocytoma, and other brain
tumors [16]. Neurofibromatosis type 2 syndrome caused
by germline or somatic mutations of the NF2 tumor sup-
pressor gene may also be associated with higher incidences
of central and peripheral nervous system tumors, such as
cranial nerve schwannomas and cranial meningiomas [16].
Li-Fraumeni syndrome is an autosomal dominant syndrome
caused by a germline mutation in the tumor suppressor gene
TP53; this condition is associated with a higher incidence of
various cancers, such as adrenocortical carcinomas, breast
cancer, soft tissue sarcomas, and even central nervous sys-
tem neuroepithelial tumors, and it appears to be more com-
mon in females than in males [16]. Turcot syndrome appears
to be a condition caused by the loss of one of the two arms
of chromosome 17, caused by a germline mutation of APC
(isochromosome 17). There is a higher incidence of glioblas-
toma and medulloblastoma, in addition to the increased risk
of developing colorectal cancer, [17].
Radiation
Moderate or high exposure to ionizing radiation is, to
date, the only ascertained environmental risk factor for
the onset of glioma with the most evidence in the litera-
ture, yet only represented by a small percentage of patients
[18]. The carcinogenic effect of ionizing radiation with an
associated increased risk of glioma diagnosis was assessed
to be more present in children than in adults, particularly
in those treated for acute lymphoblastic leukemia [19]. A
study involving 14,000 pediatric patients who had previ-
ously received radiotherapy found 40 cases of glioma dur-
ing follow-up. These occurred on average 9years after the
main diagnosis, and a case–control analysis found an odds
ratio of 6.78 for glioma diagnosis in children who received
radiotherapy versus those who did not (95% CI 1.54–29.7)
[15, 20]. Some studies evaluated the relationship between
the intensity of radiation received in childhood and the sub-
sequent development of tumors of the central nervous sys-
tem; the data obtained demonstrates that there is a sevenfold
increase in the risk of developing gliomas following any
exposure to radiation [16, 20, 21]. The high use of radiologi-
cal techniques in pediatric and prenatal age also seems to be
correlated with a greater risk of developing brain tumors,
with a relative risk of 1.29 [22]. All studies agree that the
risk of developing a brain tumor increases with the amount
of radiation administered and the younger the age of the
patient receiving radiotherapy [12]. As regards exposure to
non-ionizing radiation, the correlation between the develop-
ment of brain tumors and exposure from the use of mobile
phones has apparently not yet been confirmed due to contra-
dictory studies and differing selection and methodological
biases. The INTERPHONE study evaluated 2708 cases of
glioma and 2409 cases of meningioma and matched con-
trols in 13 countries: the results showed that there is a 40%
increase in the risk of developing glioma for the highest
decile of mobile phone use and an increased risk of develop-
ing tumors on the side of the head, where the phone is placed
Clinical and Translational Imaging
1 3
most frequently [23]. However, these findings have not been
confirmed by additional large studies, which have found no
correlation between mobile phone use and an increased risk
of developing gliomas [24, 25]. Given the non-conclusive
findings in the literature and the increasingly widespread use
of these devices, further studies with adequate follow-up are
certainly necessary to clarify any correlations present.
Socioeconomic status
Socioeconomic status (SES) has always been considered a
possible risk factor in various fields of oncology and for
different types of cancer. As far as neuro-oncology is con-
cerned, a high SES has always been associated with an
increased risk of tumors of the central nervous system and,
in particular, of gliomas. Some studies have shown that
population samples with a higher SES have a greater risk
of developing brain tumors than those with a lower socio-
economic status, regardless of age and ethnicity [2628].
Although there may be several explanations for this type of
disparity, a precise reason has not yet been identified with
regard to this data. Some studies have hypothesized a diag-
nostic bias as a possible explanation. However, this does
not make the disparities between a low and a high socio-
economic status perfectly explainable if data from different
health systems are taken into consideration [26, 29, 30]. Cer-
tainly, it is more conceivable for people living in areas with
a low SES to experience undiagnosed or incorrectly clas-
sified cancers than populations with a high socioeconomic
status. Another plausible explanation appears to be related
to the different environmental and occupational risk factors
between populations with a high and low SES, making the
incidence of central nervous system tumors greater in people
with a higher socioeconomic status.
Microbiological andimmunological factors
The correlation between viruses and cancer has always been
studied, to the point, where the presence or absence of viral
infection is also considered as a potential prognostic factor
and risk factor in some types of cancer [31, 32]. As far as
gliomas are concerned, some studies have looked into the
possibility of a link between Herpes Simplex 1 or 2 infection
and the onset of brain tumors, but the results were found to
be contradictory and difficult to interpret [33]. The same
may be said for human Cytomegalovirus infection, where,
similar to the above, the results of correlation studies failed
to establish a clear link [34]. Data on the Varicella-Zoster
Virus (VZV) are different; in this case, some studies have
shown that having a history of VZV infection is associated
with an approximate 20% reduction in the risk of develop-
ing a glioma. This is most likely due to the virus’ latency
at the nervous level and the balance that is established
with the immune system, which appears to offer protection
against the development of gliomas [35, 36]. As with the
Varicella-Zoster Virus, a history of allergy or atopy seems
to be associated with a reduction in the risk of developing
brain tumors, particularly gliomas. Two large studies [37,
38] explored this aspect, and the results show that having
a history of allergy or atopy reduces the risk of develop-
ing gliomas by 22%. This inverse risk is also, in this case,
explained by allergic people’s heightened immunosurveil-
lance, which could make development and neoplastic trans-
formation more difficult. However, the exact mechanism is
currently unknown.
Chemical agents
Due to their capacity to be fat-soluble and propensity to pen-
etrate the blood–brain barrier, exposure to chemical agents
has always been considered, along with specific mutational
signatures related to them, a possible source of risk for the
development of various tumors, such as lung cancer and
breast cancer [39]. As for central nervous system tumors,
and particularly for gliomas, there does not seem to be such
a precise correlation. In this type of disease, this confirms
that the first hit is due to an endogenous process, not altered
by external exposure [39, 40]. However, in a relatively
recent study [40], an exposure-related mutational signature
was identified for substances known as haloalkanes. These
substances are derived from alkanes which contain one or
more halogens, and are often used commercially as refrig-
erants, propellants, in fire-fighting, and in pharmaceutical
products. These preliminary data undoubtedly need to be
confirmed by epidemiological and genetic studies to confirm
whether or not there is an association between exposure to
these agents and a higher incidence of tumors of the central
nervous system.
Lifestyle factors
Lifestyle risk factors for various types of cancer have long
been investigated in the field of oncology and particu-
larly close correlations have been found in some cases, for
instance, cigarette smoking with lung cancer. As is the case
for CNS tumors, there is a dearth of available data and, typi-
cally, no correlation between lifestyle factors and the onset
of these types of tumors has been identified, probably due to
their rarity and short survival, which makes these kinds of
epidemiological studies difficult. Alcohol consumption has
been investigated as a possible risk factor for the incidence
of CNS tumors, particularly gliomas and glioblastomas.
Data in the literature appear to be contradictory: some stud-
ies show an increased risk associated with higher alcohol
consumption [4143], while other studies appear to confirm
a lower risk of developing these tumors with higher alcohol
Clinical and Translational Imaging
1 3
consumption [44, 45]. Unlike other types of cancer, cigarette
smoking does not seem to be associated with an increased
risk of developing CNS tumors. This is the conclusion drawn
from several studies and meta-analyses which evaluated the
intensity and duration of the smoking habit [45, 46]. Sleep
duration has also been investigated as a possible risk factor
for the onset of various cancers, including CNS cancers.
A published study established that there is no correlation
between sleep duration and the onset of brain tumors, par-
ticularly by comparing sleep durations of less than 7h and
more than 8h [47]. As regards diet, there does not appear
to be a correlation with the incidence of gliomas: a pooled
analysis of three studies demonstrated the absence of this
correlation when food groups (fruits and vegetables), indi-
vidual foods (fish and meat), and nutrients (fats, carbohy-
drates, and vitamins) were considered [48].
Prognostic factors
Age, performance status, andpre‑operative tumor size
Diagnosis at 40years is a strong and well-known unfavora-
ble prognostic factor for survival in all gliomas [49, 50] and
categorizes patients in a subgroup, where more aggressive
treatment should be considered. In addition, older patients
are typically affected by IDH wild-type high-grade gliomas,
which have a worse prognosis [51]. PFS (progression-free
survival) and OS (overall survival) are negatively influenced
by a worse baseline neurological status (e.g., the presence
of neurological and/or cognitive deficits) and a lower Kar-
nofsky performance status (KPS) before surgery in both
high-grade (HGG) [49] and low-grade glioma (LGG) [52],
whereas a history of seizures at diagnosis is associated with
an increased PFS and OS in LGG [53]. Several retrospective
reviews have reported the tumoral diameter size as a factor
associated with lower survival, but different cutoffs have
been proposed (4 cm [54], 5 cm [52], and 6 cm [50]).
Extent ofresection
The procedure for surgery should be individualized based
on suspected tumor type and grade, location, and operabil-
ity. Complete or gross total resection without compromising
neurological function is the better choice when it is con-
sidered safe and feasible. If complete resection cannot be
obtained, stereotactic biopsy for deep-seated tumors can
lead to diagnosis. Surgery has a debulking role on lesions
with mass effects, which aids in the improvement of symp-
toms and seizure control [55]. The extent of resection (EOR)
should be assessed within 24–48h after surgery through
MRI with contrast (or CT if MRI is not possible) [55]. Its
estimation is based on the volume of residual tumor accord-
ing to the following formula: “EOR = preoperative tumor
volume postoperative tumor volume/preoperative tumor
volume”. The prognostic role of EOR is still not fully under-
stood, because there are no randomized controlled trials that
have compared the outcome benefits of biopsy versus gross
resection [51]. Retrospective and uncontrolled data sug-
gest that the extent of surgery and the subsequent absence
of residual tumor on imaging have a favorable prognostic
effect in increasing overall survival and time to progression
in anaplastic glioma [56] and in glioblastoma (GBM) [57].
A metanalysis [58] of 19 retrospective studies and 1 rand-
omized control trial observed that patients with LLG, both
with and without 1p/19q codeletion, who undergo gross total
resection have a better 5- and 10year OS rate than those
who undergo subtotal resection. Moreover, patients who
undergo subtotal resection obtain a similar benefit to those
with biopsy alone. In clinical practice, the scenario for LGG
is more complicated than for HGG, because a watch-and-
wait strategy may be appropriate, particularly for patients
who only have seizures (without focal neurological signs or
symptoms or signs of increased intracranial pressure) and at
a young age (< 40), with the possibility of surgery without
further delay at the time of radiological progression. How-
ever, more recent data suggest that early surgical resection
can result in a clinically relevant survival benefit [59] in
LGG patients too. Furthermore, the association between the
extent of resection and OS may be explained by the pres-
ence of mutations in isocitrate dehydrogenase (IDH) 1/2:
in a large retrospective series [60], IDH mutant anaplastic
astrocytomas and GBM (according to the 2016 WHO clas-
sification of central nervous system tumors [11]) resulted
more amenable to surgical resection, with a survival benefit
associated with maximal surgical resection.
Moreover, also the possibility to receive a radiochemo-
therapy treatment could influence the outcome regardless of
other prognostic factors and neurocognitive functions [61].
In particular, patients treated with new targeted therapy such
as regorafenib or anti-BRAF may have a longer overall sur-
vival [62, 63].
Histology andgrade
The recently updated [2] World Health Organization (WHO)
classification of tumors of the central nervous system,
defines 15 entities of adult diffuse gliomas based on molecu-
lar and histological diagnosis. Astrocytoma histology sub-
type is associated with an unfavorable prognosis compared
to oligodendroglioma [64]. Oligodendroglial tumors, charac-
terized both by codeletion of 1p and 19q (1p/19q) and IDH
1/2 mutation, are classified as WHO grade 2 or 3. They carry
a better prognosis than astrocytomas [52]. IDH mutant astro-
cytic tumors are considered as a single entity (IDH mutant
astrocytoma) and are graded as WHO grade 2, 3, or 4. WHO
grade 2 or 3 gliomas are invasive and tend to progress to
Clinical and Translational Imaging
1 3
higher grade lesions (WHO grade 4 astrocytomas) that have
the worst prognosis. However, the most aggressive form is
IDH wild-type GBM; in a recent retrospective multicenter
study, the reported median OS was 19.2–23.8months [49].
Molecular characteristics
An assessment of the IDH mutational status and 1p/19q
codeletion is required for the diagnosis of diffuse gliomas.
Several molecular alterations have been established as
markers of high grade tumors despite the histology, such
as CDKN2A/Bhomozygous deletion in IDH mutant grade
4 astrocytoma and TERTpromoter mutation,EGFRgene
amplification and/or combined gain of entire chromosome
7, and loss of entire chromosome 10 [+ 7/ 10] in IDH
wildtype GBM 265.
Isocitratedehydrogenase (IDH) 1/2 mutation
IDH1 and IDH2 encode important enzymes in the Krebs
cycle. IDH1 and IDH2 mutations reside at the enzymes’
catalytic domains and a-ketoglutarate (a-KG) is reduced to
D-2-hydroxyglutarate (2HG), an oncometabolite. IDH1 and
IDH2 mutations are mutually exclusive; they are much more
common in WHO grade 2 and 3 gliomas (60–80%) than in
glioblastomas (5–10%) 66(p2). IDH1/2 mutations in glioma
are associated with a significantly prolonged PFS and OS
than comparable tumors without an IDH mutation, mak-
ing IDH mutation the most important prognostic factor for
survival. In a large clinical data set of GBM [66], it was
observed that GBM patients harboring IDH 1/2 mutations
tend to have a prolonged median OS and median PFS in
comparison to patients with IDH wild-type GBM (median
OS 31 versus 15 months). Moreover, this tendency was con-
firmed among patients with anaplastic astrocytoma (median
OS of 65 months for patients with IDH mutant tumors ver-
sus 20 months for wild-type tumors). IDH mutation status
is an independent predictor of favorable outcomes among
glioma patients [67]. Furthermore, in the large randomized
phase III CATNON trial (“Concurrent and Adjuvant Temo-
zolomide chemotherapy in non-1p/19q deleted anaplastic
glioma”), the median overall survival was 19.9 months (95%
CI 16.8–22.7) for patients with IDH1 and IDH2 wild-type
anaplastic astrocytoma and 98.4 months (85.2–116.6) for
patients with IDH1 or IDH2 mutant tumors (HR 0.14 [95%
CI 0.12–0.18], p < 0.0001), regardless of treatment [68]. In
a recent study [69] involving a cohort of 433 patients harbor-
ing IDH1 mutated grade 2–3 glioma (90.1% with the canoni-
cal IDH1 R132H mutation and 9.9% with a non-canonical
IDH1 mutation), it was observed that non-canonical muta-
tions could be associated with improved survival; the pres-
ence of non-canonical IDH1 mutations is associated with a
younger age at diagnosis and a less frequent presence of the
1p19q codeletion. In addition, an analysis of the CATNON
trial’s 1p/19q non-codeleted astrocytoma samples found that
patients harboring non-R132H mutated tumors have a better
outcome (HR 0.41 [95% CI 0.24, 0.71], p = 0.0013) because
of higher levels of genome-wide DNA-methylation [70].
IDH mutation status can also be used to differentiate
between primary and secondary glioblastomas [66]: the
majority of primary GBMs, which are frequently diagnosed
in older patients, are IDH wild type in the absence of a lower
grade precursor, while virtually all secondary GBMs that
arise from lower grade gliomas have IDH mutation. This
finding, associated with the prognostic role of IDH1/2 muta-
tion, indicates that many IDH wild-type lower grade gliomas
can exhibit aggressive behavior comparable to GBM and
have a similar prognosis [71].
Codeletion of1p and19q (1p/19q)
The combined loss of chromosome arms 1p and 19q,
together with IDH mutation, are pathognomonic markers
of oligodendroglioma. As demonstrated by the results of
large randomized clinical trials, codeletion is a powerful
predictor of favorable therapeutic response and improved
survival among patients with diffuse gliomas. In summary,
in the RTOG 9402 trial, the presence of 1p/19q codeletion
in anaplastic oligodendrogliomas and oligoastrocytomas
was associated with longer OS when the patient was treated
with radiation (7.3 versus 2.7years with intact 1p/19q) [72].
Furthermore, the addition of subsequent PCV (procarbazine,
lomustine, and vincristine) chemotherapy led to an increase
in OS to 14.7years in the presence of 1p/19q codeletion,
but did not change survival for 1p/19q intact patients. The
EORTC 26,951, a large EORTC (European Organization for
Research and Treatment of Cancer) Brain Tumor Group pro-
spective phase III study, revealed similar chemosensitivity
and a favorable prognosis for 1p/19q codeleted tumors [73].
O‑6‑methylguanine–DNA methyltransferase promoter
(MGMTp) methylation (in GBM)
MGMT, O6-Methylguanine–DNA methyltransferase, gene
located on chromosome 10q26.3, is a nuclear enzyme
responsible for DNA mismatch repair; indeed, MGMT
repairs damaged guanine nucleotides due to alkylating
agents, such as temozolomide and nitrosourea derivatives.
MGMT gene has a CpG island with a length of 762bp.
MGMTp methylation can increase the sensitivity of high-
grade gliomas to chemotherapy [74].
MGMTp methylation has been observed in approximately
50% of GBM. It is identified as a strong prognostic factor in
GBM, both in the “Stupp” trial [75] and in following rand-
omized trials, better designed for subgroup analysis accord-
ing to theMGMTstatus; for instance, in the RTOG 0525
Clinical and Translational Imaging
1 3
trial, the median OS was 21.2 months and 14.0 months with
or without MGMTp methylation, respectively [76]. We have
known since 2005 that patients withMGMTp-methylated
GBM had a longer survival when treated with temozolomide
chemotherapy than patients with MGMTp-unmethylated
tumors [77]. Its role as a predictive marker to select patients
for treatment was also confirmed in elderly patients with
GBM who, in the presence of MGMTp methylation, had
better outcomes with temozolomide chemotherapy than with
radiotherapy [78] (NORDIC and NOA-08 trials). In contrast,
patients withMGMTp-unmethylated GBM had reduced sur-
vival when treated with chemotherapy.However, in a recent
analysis by van den Bent etal, MGMTp methylation deter-
mined by the MGMT–STP27 algorithm was not predictive
for outcome in patients with IDH-mutant anaplastic astro-
cytomas enrolled in the CATNON trial [79].
Several studies have demonstrated that MGMTp meth-
ylation is also prognostic for the recurrence of GBM,
with an examined longer post-progression survival (about
3–4 months longer in the presence than in the absence of
MGMTp methylation) [80].
Conclusions
Gliomas represent a large group of heterogeneous entities.
Overall, gliomas are about 81% of all malignant tumors in
adults. Ionizing radiation remains the only ascertained envi-
ronmental risk factor associated with glioma; the role of
mobile phones is unclear due to contradictory studies. In
addition to the clinical characteristics, such as the age, per-
formance status and the extent of resection, also molecular
alterations, such as IDH and TERT gene mutations, 1p/19q
codeletion, CDKN2A/B homozygous deletion and the
MGMT methylation status can correlate with the survival
in specific subgroups of glioma patients.
Declarations
Conflict of interest Alessia Pellerino, Mario Caccese, Marta Padovan,
Giulia Cerretti, Giuseppe Lombardi declare no conflict of interest.
Ethical approval This article does not contain any studies with human
or animal subjects performed by the any of the authors.
References
1. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-
Sloan JS (2020) CBTRUS statistical report: primary brain and
other central nervous system tumors diagnosed in the United
States in 2013–2017. Neuro Oncol 22(12 Supp 2):iv1–iv96.
https:// doi. org/ 10. 1093/ neuonc/ noaa2 00
2. Louis DN, Perry A, Wesseling P etal (2021) The 2021 WHO
classification of tumors of the central nervous system: a sum-
mary. Neuro Oncol 23(8):1231–1251. https:// doi. org/ 10. 1093/
neuonc/ noab1 06
3. Patil N, Kelly ME, Yeboa DN etal (2021) Epidemiology of
brainstem high-grade gliomas in children and adolescents in the
United States, 2000–2017. Neuro Oncol 23(6):990–998. https://
doi. org/ 10. 1093/ neuonc/ noaa2 95
4. Hoffman LM, Veldhuijzen van Zanten SEM, Colditz N etal
(2018) Clinical, radiologic, pathologic, and molecular char-
acteristics of long-term survivors of diffuse intrinsic pontine
glioma (DIPG): a collaborative report from the international
and European society for pediatric oncology DIPG registries.
J Clin Oncol 36(19):1963–1972. https:// doi. org/ 10. 1200/ JCO.
2017. 75. 9308
5. Leece R, Xu J, Ostrom QT, Chen Y, Kruchko C, Barnholtz-Sloan
JS (2017) Global incidence of malignant brain and other central
nervous system tumors by histology, 2003–2007. Neuro Oncol
19(11):1553–1564. https:// doi. org/ 10. 1093/ neuonc/ nox091
6. Girardi F, Rous B, Stiller CA etal (2021) The histology of brain
tumors for 67 331 children and 671 085 adults diagnosed in 60
countries during 2000–2014: a global, population-based study
(CONCORD-3). Neuro Oncol 23(10):1765–1776. https:// doi. org/
10. 1093/ neuonc/ noab0 67
7. Nakamura H, Makino K, Yano S, Kuratsu JI (2011) Kumamoto
brain tumor research group epidemiological study of primary
intracranial tumors: a regional survey in kumamoto prefecture in
southern Japan–20-year study. Int J Clin Oncol 16(4):314–321.
https:// doi. org/ 10. 1007/ s10147- 010- 0178-y
8. Ostrom QT, Egan KM, Nabors LB etal (2020) Glioma risk associ-
ated with extent of estimated European genetic ancestry in African
Americans and Hispanics. Int J Cancer 146(3):739–748. https://
doi. org/ 10. 1002/ ijc. 32318
9. Lin D, Wang M, Chen Y etal (2021) Trends in intracranial glioma
incidence and mortality in the United States, 1975–2018. Front
Oncol 11:748061. https:// doi. org/ 10. 3389/ fonc. 2021. 748061
10. Wrensch M, Lee M, Miike R etal (1997) Familial and personal
medical history of cancer and nervous system conditions among
adults with glioma and controls. Am J Epidemiol 145(7):581–593.
https:// doi. org/ 10. 1093/ oxfor djour nals. aje. a0091 54
11. Louis DN, Perry A, Reifenberger G etal (2016) The 2016 world
health organization classification of tumors of the central nervous
system: a summary. Acta Neuropathol 131(6):803–820. https://
doi. org/ 10. 1007/ s00401- 016- 1545-1
12. Ostrom QT, Fahmideh MA, Cote DJ etal (2019) Risk factors
for childhood and adult primary brain tumors. Neuro Oncol
21(11):1357–1375. https:// doi. org/ 10. 1093/ neuonc/ noz123
13. Kinnersley B, Mitchell JS, Gousias K etal (2015) Quantifying the
heritability of glioma using genome-wide complex trait analysis.
Sci Rep 5:17267. https:// doi. org/ 10. 1038/ srep1 7267
14. Rice T, Lachance DH, Molinaro AM etal (2016) Understanding
inherited genetic risk of adult glioma - a review. Neurooncol Pract
3(1):10–16. https:// doi. org/ 10. 1093/ nop/ npv026
15. Howell AE, Zheng J, Haycock PC etal (2018) Use of mendelian
randomization for identifying risk factors for brain tumors. Front
Genet 9:525. https:// doi. org/ 10. 3389/ fgene. 2018. 00525
16. Francis SS, Ostrom QT, Cote DJ, Smith TR, Claus E, Barnholtz-
Sloan JS (2022) The epidemiology of central nervous system
tumors. Hematol Oncol Clin North Am 36(1):23–42. https:// doi.
org/ 10. 1016/j. hoc. 2021. 08. 012
17. Khattab A, Monga DK. (2021) Turcot Syndrome. In: StatPearls.
StatPearls Publishing. http:// www. ncbi. nlm. nih. gov/ books/
NBK53 4782/. Accessed 6 Jan 2022
18. Braganza MZ, Kitahara CM, Berrington de González A, Inskip
PD, Johnson KJ, Rajaraman P (2012) Ionizing radiation and the
risk of brain and central nervous system tumors: a systematic
Clinical and Translational Imaging
1 3
review. Neuro Oncol 14(11):1316–1324. https:// doi. org/ 10. 1093/
neuonc/ nos208
19. Davis F, Il’yasova D, Rankin K, McCarthy B, Bigner DD (2011)
Medical diagnostic radiation exposures and risk of gliomas.
Radiat Res 175(6):790–796. https:// doi. org/ 10. 1667/ RR2186.1
20. Neglia JP, Robison LL, Stovall M etal (2006) New primary neo-
plasms of the central nervous system in survivors of childhood
cancer: a report from the childhood cancer survivor study. J Natl
Cancer Inst 98(21):1528–1537. https:// doi. org/ 10. 1093/ jnci/ djj411
21. Inskip PD, Sigurdson AJ, Veiga L etal (2016) Radiation-related
new primary solid cancers in the childhood cancer survivor study:
comparative radiation dose response and modification of treatment
effects. Int J Radiat Oncol Biol Phys 94(4):800–807. https:// doi.
org/ 10. 1016/j. ijrobp. 2015. 11. 046
22. Sheppard JP, Nguyen T, Alkhalid Y, Beckett JS, Salamon N, Yang
I (2018) Risk of brain tumor induction from pediatric head CT
procedures: a systematic literature review. Brain Tumor Res Treat
6(1):1–7. https:// doi. org/ 10. 14791/ btrt. 2018.6. e4
23. INTERPHONE Study Group (2010) Brain tumour risk in relation
to mobile telephone use: results of the INTERPHONE interna-
tional case-control study. Int J Epidemiol 39(3):675–694. https://
doi. org/ 10. 1093/ ije/ dyq079
24. Benson VS, Pirie K, Schüz J etal (2013) Mobile phone use and
risk of brain neoplasms and other cancers: prospective study. Int
J Epidemiol 42(3):792–802. https:// doi. org/ 10. 1093/ ije/ dyt072
25. Frei P, Poulsen AH, Johansen C, Olsen JH, Steding-Jessen M,
Schüz J (2011) Use of mobile phones and risk of brain tumours:
update of Danish cohort study. BMJ 343:d6387. https:// doi. org/
10. 1136/ bmj. d6387
26. Porter AB, Lachance DH, Johnson DR (2015) Socioeconomic
status and glioblastoma risk: a population-based analysis. Can-
cer Causes Control 26(2):179–185. https:// doi. org/ 10. 1007/
s10552- 014- 0496-x
27. Demers PA, Vaughan TL, Schommer RR (1991) Occupation, soci-
oeconomic status, and brain tumor mortality: a death certificate-
based case-control study. J Occup Med 33(9):1001–1006
28. Plascak JJ, Fisher JL (2013) Area-based socioeconomic position
and adult glioma: a hierarchical analysis of surveillance epidemi-
ology and end results data. PLoS ONE 8(4):e60910. https:// doi.
org/ 10. 1371/ journ al. pone. 00609 10
29. Khanolkar AR, Ljung R, Talbäck M etal (2016) Socioeco-
nomic position and the risk of brain tumour: a Swedish national
population-based cohort study. J Epidemiol Community Health
70(12):1222–1228. https:// doi. org/ 10. 1136/ jech- 2015- 207002
30. Cote DJ, Ostrom QT, Gittleman H etal (2019) Glioma incidence
and survival variations by county-level socioeconomic measures.
Cancer 125(19):3390–3400. https:// doi. org/ 10. 1002/ cncr. 32328
31. Dlamini Z, Alaouna M, Mbatha S etal (2021) Genetic drivers of
head and neck squamous cell carcinoma: aberrant splicing events,
mutational burden, HPV infection and future targets. Genes
(Basel) 12(3):422. https:// doi. org/ 10. 3390/ genes 12030 422
32. Araldi RP, Sant’Ana TA, Módolo DG, etal (2018) The human
papillomavirus (HPV)-related cancer biology: an overview.
Biomed Pharmacother 106:1537–1556. https:// doi. org/ 10. 1016/j.
biopha. 2018. 06. 149
33. Neves AM, Thompson G, Carvalheira J etal (2008) Detection and
quantitative analysis of human herpesvirus in pilocytic astrocy-
toma. Brain Res 1221:108–114. https:// doi. org/ 10. 1016/j. brain res.
2008. 05. 009
34. Garcia-Martinez A, Alenda C, Irles E etal (2017) Lack of cyto-
megalovirus detection in human glioma. Virol J 14(1):216. https://
doi. org/ 10. 1186/ s12985- 017- 0885-3
35. Wrensch M, Weinberg A, Wiencke J etal (1997) Does prior infec-
tion with varicella-zoster virus influence risk of adult glioma? Am
J Epidemiol 145(7):594–597. https:// doi. org/ 10. 1093/ oxfor djour
nals. aje. a0091 55
36. Amirian ES, Scheurer ME, Zhou R etal (2016) History of chick-
enpox in glioma risk: a report from the glioma international case-
control study (GICC). Cancer Med 5(6):1352–1358. https:// doi.
org/ 10. 1002/ cam4. 682
37. Amirian ES, Zhou R, Wrensch MR etal (2016) Approaching a sci-
entific consensus on the association between allergies and glioma
risk: a report from the glioma international case-control study.
Cancer Epidemiol Biomarkers Prev 25(2):282–290. https:// doi.
org/ 10. 1158/ 1055- 9965. EPI- 15- 0847
38. Linos E, Raine T, Alonso A, Michaud D (2007) Atopy and risk
of brain tumors: a meta-analysis. J Natl Cancer Inst 99(20):1544–
1550. https:// doi. org/ 10. 1093/ jnci/ djm170
39. Alexandrov LB, Kim J, Haradhvala NJ etal (2020) The repertoire
of mutational signatures in human cancer. Nature 578(7793):94–
101. https:// doi. org/ 10. 1038/ s41586- 020- 1943-3
40. Claus EB, Cannataro VL, Gaffney SG, Townsend JP (2022) Envi-
ronmental and sex-specific molecular signatures of glioma causa-
tion. Neuro Oncol 24(1):29–36. https:// doi. org/ 10. 1093/ neuonc/
noab1 03
41. Hu J, La Vecchia C, Negri E etal (1999) Diet and brain cancer
in adults: a case-control study in northeast China. Int J Cancer
81(1):20–23. https:// doi. org/ 10. 1002/ (sici) 1097- 0215(19990 331)
81:1% 3c20:: aid- ijc4% 3e3.0. co;2-2
42. Giles GG, McNeil JJ, Donnan G etal (1994) Dietary factors and
the risk of glioma in adults: results of a case-control study in
Melbourne. Australia Int J Cancer 59(3):357–362. https:// doi. org/
10. 1002/ ijc. 29105 90311
43. Lee M, Wrensch M, Miike R (1997) Dietary and tobacco risk
factors for adult onset glioma in the San Francisco Bay Area (Cali-
fornia, USA). Cancer Causes Control 8(1):13–24. https:// doi. org/
10. 1023/a: 10184 70802 969
44. Baglietto L, Giles GG, English DR, Karahalios A, Hopper JL,
Severi G (2011) Alcohol consumption and risk of glioblastoma;
evidence from the melbourne collaborative cohort study. Int J
Cancer 128(8):1929–1934. https:// doi. org/ 10. 1002/ ijc. 25770
45. Braganza MZ, Rajaraman P, Park Y etal (2014) Cigarette smok-
ing, alcohol intake, and risk of glioma in the NIH-AARP diet and
health study. Br J Cancer 110(1):242–248. https:// doi. or g/ 10. 1038/
bjc. 2013. 611
46. Shao C, Zhao W, Qi Z, He J (2016) Smoking and glioma risk:
evidence from a meta-analysis of 25 observational studies. Medi-
cine (Baltimore) 95(2):e2447. https:// doi. org/ 10. 1097/ MD. 00000
00000 002447
47. Gu F, Xiao Q, Chu LW etal (2016) Sleep duration and cancer
in the NIH-AARP diet and health study cohort. PLoS ONE
11(9):e0161561. https:// doi. org/ 10. 1371/ journ al. pone. 01615 61
48. Kuan AS, Green J, Kitahara CM etal (2019) Diet and risk of
glioma: combined analysis of 3 large prospective studies in the
UK and USA. Neuro Oncol 21(7):944–952. https:// doi. org/ 10.
1093/ neuonc/ noz013
49. Tesileanu CMS, Dirven L, Wijnenga MMJ etal (2020) Survival
of diffuse astrocytic glioma, IDH1/2 wildtype, with molecular
features of glioblastoma, WHO grade IV: a confirmation of the
cIMPACT-NOW criteria. Neuro Oncol 22(4):515–523. https://
doi. org/ 10. 1093/ neuonc/ noz200
50. Pignatti F, van den Bent M, Curran D etal (2002) Prognostic fac-
tors for survival in adult patients with cerebral low-grade glioma.
JCO 20(8):2076–2084. https:// doi. org/ 10. 1200/ JCO. 2002. 08. 121
51. van den Bent MJ, Smits M, Kros JM, Chang SM (2017) Dif-
fuse infiltrating oligodendroglioma and astrocytoma. JCO
35(21):2394–2401. https:// doi. org/ 10. 1200/ JCO. 2017. 72. 6737
52. Gorlia T, Wu W, Wang M etal (2013) New validated prognos-
tic models and prognostic calculators in patients with low-grade
gliomas diagnosed by central pathology review: a pooled analysis
of EORTC/RTOG/NCCTG phase III clinical trials. Neuro Oncol
15(11):1568–1579. https:// doi. org/ 10. 1093/ neuonc/ not117
Clinical and Translational Imaging
1 3
53. Pallud J, Audureau E, Blonski M etal (2014) Epileptic seizures
in diffuse low-grade gliomas in adults. Brain 137(Pt 2):449–462.
https:// doi. org/ 10. 1093/ brain/ awt345
54. Chang EF, Smith JS, Chang SM etal (2008) Preoperative prog-
nostic classification system for hemispheric low-grade gliomas in
adults: clinical article. J Neurosurg 109(5):817–824. https:// doi.
org/ 10. 3171/ JNS/ 2008/ 109/ 11/ 0817
55. Weller M, van den Bent M, Preusser M etal (2021) EANO guide-
lines on the diagnosis and treatment of diffuse gliomas of adult-
hood. Nat Rev Clin Oncol 18(3):170–186. https:// doi. org/ 10. 1038/
s41571- 020- 00447-z
56. Gorlia T, Delattre JY, Brandes AA etal (2013) New clinical,
pathological and molecular prognostic models and calculators
in patients with locally diagnosed anaplastic oligodendroglioma
or oligoastrocytoma. A prognostic factor analysis of European
organisation for research and treatment of cancer brain tumour
group study 26951. Eur J Cancer 49(16):3477–3485. https:// doi.
org/ 10. 1016/j. ejca. 2013. 06. 039
57. Molinaro AM, Hervey-Jumper S, Morshed RA etal (2020) Asso-
ciation of maximal extent of resection of contrast-enhanced and
non-contrast-enhanced tumor with survival within molecular
subgroups of patients with newly diagnosed glioblastoma. JAMA
Oncol 6(4):495–503. https:// doi. org/ 10. 1001/ jamao ncol. 2019.
6143
58. Xia L, Fang C, Chen G, Sun C (2018) Relationship between the
extent of resection and the survival of patients with low-grade
gliomas: a systematic review and meta-analysis. BMC Cancer
18(1):48. https:// doi. org/ 10. 1186/ s12885- 017- 3909-x
59. Jakola AS, Skjulsvik AJ, Myrmel KS etal (2017) Surgical resec-
tion versus watchful waiting in low-grade gliomas. Ann Oncol
28(8):1942–1948. https:// doi. org/ 10. 1093/ annonc/ mdx230
60. Beiko J, Suki D, Hess KR etal (2014) IDH1 mutant malignant
astrocytomas are more amenable to surgical resection and have a
survival benefit associated with maximal surgical resection. Neuro
Oncol 16(1):81–91. https:// doi. org/ 10. 1093/ neuonc/ not159
61. Bergo E, Lombardi G, Guglieri I, Capovilla E, Pambuku A, Zago-
nel V (2015) Neurocognitive functions and health-related quality
of life in glioblastoma patients: a concise review of the literature.
EurJCancerCare (Engl). https:// doi. org/ 10. 1111/ ecc. 12410
62. Lombardi G, De Salvo GL, Brandes AA etal (2019) Regorafenib
compared with lomustine in patients with relapsed glioblastoma
(REGOMA): a multicentre, open-label, randomised, controlled,
phase 2 trial. Lancet Oncol 20(1):110–119
63. Birzu C, French P, Caccese M etal (2020) Recurrent glioblas-
toma: from molecular landscape to new treatment perspectives.
Cancers (Basel). https:// doi. org/ 10. 3390/ cance rs130 10047
64. Pignatti F, van den Bent M, Curran D etal (2002) Prognostic fac-
tors for survival in adult patients with cerebral low-grade glioma.
J Clin Oncol 20(8):2076–2084. https:// doi. org/ 10. 1200/ JCO. 2002.
08. 121
65. Di Stefano AL, Labussiere M, Lombardi G etal (2015) VEGFA
SNP rs2010963 is associated with vascular toxicity in recurrent
glioblastomas and longer response to bevacizumab. JNeurooncol
121(3):499–504. https:// doi. org/ 10. 1007/ s11060- 014- 1677-x
66. Yan H, Parsons DW, Jin G etal (2009) IDH1 and IDH2 Mutations
in gliomas. N Engl J Med. https:// doi. org/ 10. 1056/ NEJMo a0808
710
67. Sanson M, Marie Y, Paris S etal (2009) Isocitrate dehydrogenase
1 codon 132 mutation is an important prognostic biomarker in
gliomas. J Clin Oncol 27(25):4150–4154. https:// doi. org/ 10. 1200/
JCO. 2009. 21. 9832
68. van den Bent MJ, Tesileanu CMS, Wick W etal (2021) Adju-
vant and concurrent temozolomide for 1p/19q non-co-deleted
anaplastic glioma (CATNON; EORTC study 26053–22054): sec-
ond interim analysis of a randomised, open-label, phase 3 study.
Lancet Oncol 22(6):813–823. https:// doi. org/ 10. 1016/ S1470-
2045(21) 00090-5
69. Franceschi E, De Biase D, Di Nunno V etal (2021) IDH1 Non-
canonical mutations and survival in patients with glioma. Diag-
nostics (Basel) 11(2):342. https:// doi. org/ 10. 3390/ diagn ostic s1102
0342
70. Tesileanu CMS, Vallentgoed WR, Sanson M etal (2021) Non-
IDH1-R132H IDH1/2 mutations are associated with increased
DNA methylation and improved survival in astrocytomas, com-
pared to IDH1-R132H mutations. Acta Neuropathol 141(6):945–
957. https:// doi. org/ 10. 1007/ s00401- 021- 02291-6
71. Dunn GP, Andronesi OC, Cahill DP (2013) From genomics to the
clinic: biological and translational insights of mutant IDH1/2 in
glioma. Neurosurg Focus 34(2):E2. https:// doi. org/ 10. 3171/ 2012.
12. FOCUS 12355
72. Cairncross G, Wang M, Shaw E etal (2013) Phase III trial of
chemoradiotherapy for anaplastic oligodendroglioma: long-term
results of RTOG 9402. J Clin Oncol 31(3):337–343. https:// doi.
org/ 10. 1200/ JCO. 2012. 43. 2674
73. van den Bent MJ, Brandes AA, Taphoorn MJB etal (2013) Adju-
vant procarbazine, lomustine, and vincristine chemotherapy in
newly diagnosed anaplastic oligodendroglioma: long-term fol-
low-up of EORTC brain tumor group study 26951. J Clin Oncol
31(3):344–350. https:// doi. org/ 10. 1200/ JCO. 2012. 43. 2229
74. Wick W, Weller M, van den Bent M etal (2014) MGMT testing–
the challenges for biomarker-based glioma treatment. Nat Rev
Neurol 10(7):372–385. https:// doi. org/ 10. 1038/ nrneu rol. 2014. 100
75. Stupp R, Mason WP, van den Bent MJ etal (2005) Radiotherapy
plus concomitant and adjuvant temozolomide for glioblastoma.
N Engl J Med 352(10):987–996. https:// doi. org/ 10. 1056/ NEJMo
a0433 30
76. Gilbert MR, Wang M, Aldape KD etal (2013) Dose-dense temo-
zolomide for newly diagnosed glioblastoma: a randomized phase
III clinical trial. J Clin Oncol 31(32):4085–4091. https:// doi. org/
10. 1200/ JCO. 2013. 49. 6968
77. Hegi ME, Diserens AC, Gorlia T etal (2005) MGMT gene silenc-
ing and benefit from temozolomide in glioblastoma. N Engl J Med
352(10):997–1003. https:// doi. org/ 10. 1056/ NEJMo a0433 31
78. Malmström A, Grønberg BH, Marosi C etal (2012) Temozo-
lomide versus standard 6-week radiotherapy versus hypofrac-
tionated radiotherapy in patients older than 60 years with glio-
blastoma: the Nordic randomised, phase 3 trial. Lancet Oncol
13(9):916–926. https:// doi. org/ 10. 1016/ S1470- 2045(12) 70265-6
79. Tesileanu CMS, Gorlia T, Golfinopoulos V, French PJ, van den
Bent MJ (2022) MGMT promoter methylation determined by the
MGMT-STP27 algorithm is not predictive for outcome to temo-
zolomide in IDH-mutant anaplastic astrocytomas. Neuro Oncol.
https:// doi. org/ 10. 1093/ neuonc/ noac0 14 (Published online Janu-
ary 31)
80. Wick W, Gorlia T, Bendszus M et al (2017) Lomustine and
Bevacizumab in Progressive Glioblastoma. N Engl J Med
377(20):1954–1963. https:// doi. org/ 10. 1056/ NEJMo a1707 358
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... Table 3 highlights the anti-cancer effects of curcumin against different cancers mediated through the PI3K/AKT/mTOR pathway. [82]. The most common and aggressive malignant brain tumor in adults is glioblastoma (GBM), with an incidence rate of about 3-4 people per 100,000 [83,84]. ...
Article
Full-text available
Simple Summary Cancer has a major impact on societies across the world. In an attempt to find new treatment options for cancer, attention has shifted to natural compounds. Curcumin is a polyphenol isolated from the roots of turmeric that possesses many biological properties. It acts on the regulation of different aspects of tumor development and interconnects with major signaling pathways that are dysregulated in cancer, such as the phosphatidylinositol-3-kinase/protein kinase B pathway. In this review, the diverse effects of curcumin on the regulation of this pathway in different malignancies will be discussed. Abstract Cancer is a life-threatening disease and one of the leading causes of death worldwide. Despite significant advancements in therapeutic options, most available anti-cancer agents have limited efficacy. In this context, natural compounds with diverse chemical structures have been investigated for their multimodal anti-cancer properties. Curcumin is a polyphenol isolated from the rhizomes of Curcuma longa and has been widely studied for its anti-inflammatory, anti-oxidant, and anti-cancer effects. Curcumin acts on the regulation of different aspects of cancer development, including initiation, metastasis, angiogenesis, and progression. The phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) pathway is a key target in cancer therapy, since it is implicated in initiation, proliferation, and cancer cell survival. Curcumin has been found to inhibit the PI3K/Akt pathway in tumor cells, primarily via the regulation of different key mediators, including growth factors, protein kinases, and cytokines. This review presents the therapeutic potential of curcumin in different malignancies, such as glioblastoma, prostate and breast cancer, and head and neck cancers, through the targeting of the PI3K/Akt signaling pathway.
... The size of intensity bins was fixed for all modalities to the average interquartile range (IQR) divided by 4 [46,47]. To take into account the typical prevalence of IDH wild-type and IDH mutant gliomas within a standard population [48], a weight of 0.6 was applied to the average IQR/4 from IDH wild-type gliomas, while the average IQR/4 from IDH mutant gliomas received a weight of 0.4. This yielded the following bin widths: 0.15 for TBR FET5-15 , 0.13 for TBR FET20-40 , 0.16 for TBR GE-180 , 0.08 for TBR T1CE , and 0.17 for TBR T2 . ...
Article
Full-text available
Purpose According to the World Health Organization classification for tumors of the central nervous system, mutation status of the isocitrate dehydrogenase (IDH) genes has become a major diagnostic discriminator for gliomas. Therefore, imaging-based prediction of IDH mutation status is of high interest for individual patient management. We compared and evaluated the diagnostic value of radiomics derived from dual positron emission tomography (PET) and magnetic resonance imaging (MRI) data to predict the IDH mutation status non-invasively. Methods Eighty-seven glioma patients at initial diagnosis who underwent PET targeting the translocator protein (TSPO) using [¹⁸F]GE-180, dynamic amino acid PET using [¹⁸F]FET, and T1-/T2-weighted MRI scans were examined. In addition to calculating tumor-to-background ratio (TBR) images for all modalities, parametric images quantifying dynamic [¹⁸F]FET PET information were generated. Radiomic features were extracted from TBR and parametric images. The area under the receiver operating characteristic curve (AUC) was employed to assess the performance of logistic regression (LR) classifiers. To report robust estimates, nested cross-validation with five folds and 50 repeats was applied. Results TBRGE-180 features extracted from TSPO-positive volumes had the highest predictive power among TBR images (AUC 0.88, with age as co-factor 0.94). Dynamic [¹⁸F]FET PET reached a similarly high performance (0.94, with age 0.96). The highest LR coefficients in multimodal analyses included TBRGE-180 features, parameters from kinetic and early static [¹⁸F]FET PET images, age, and the features from TBRT2 images such as the kurtosis (0.97). Conclusion The findings suggest that incorporating TBRGE-180 features along with kinetic information from dynamic [¹⁸F]FET PET, kurtosis from TBRT2, and age can yield very high predictability of IDH mutation status, thus potentially improving early patient management.
... Diffuse astrocytomas are innately invasive tumors that often emigrate along white matter networks deeply inside the normal brain. WHO classifies it as a grade II diffuse astrocytic neoplasm with a peak age of 30-39 years and a five-year survival rate ranging from 30 to 80% [2]. The regional impact of astrocytoma involves direct invasion and oxygen competition, which causes hypoxia of normal brain parenchyma, resulting in the release of free radicals, neurotransmitters, and inflammatory mediators, interfering with homeostasis and inducing clinical signs and symptoms [3]. ...
Article
Full-text available
Diffuse astrocytoma is a slow, progressive, and invasive tumor that develops from astrocytes and there is no discernible boundary between tumor and brain cells. We present a case of a 48-year-old woman with diffuse astrocytoma who experienced sudden left-sided weakness, multiple convulsive episodes, and vomiting. The patient underwent surgery for a left occipital mini craniotomy with complete tumor removal through a titanium burr hole. Postoperatively, the patient complained of bilateral upper and lower extremities weakness, and decreased muscular tone was found; hence, she was referred to undergo neurophysiotherapy. A four-week rehabilitative protocol was started. Physiotherapy is critical in these patients for ensuring early and rapid recovery and treating the condition's clinical manifestations. The outcome measures employed were the tone grading scale, the Brunnstrom recovery stage, and the Functional Independence Measure (FIM). This case study concludes that physiotherapy rehabilitation for an operated case of grade 2 diffuse astrocytoma led to improved lower limb strength, normal tone, and improved functional independence, which helped the patient achieve better functional activities and a greater quality of life.
... Several risk factors have been identified in relation to gliomas [27]. Notably, being exposed to ionizing radiation is connected with an increased risk, whereas having a family history of allergic diseases or atopy is associated with a lower risk [28]. More research should be directed toward discovering more effective treatments and gaining a better understanding of the genetic and molecular elements that contribute to the development and progression of this disease. ...
Article
Glioblastoma (GBM) is a highly aggressive type of primary brain cancer with a poor prognosis, and despite intensive research, survival rates have not significantly improved. Non-coding RNAs (ncRNAs) are emerging as critical regulators of GBM pathogenesis, including angiogenesis, which is essential for tumor growth and invasion. MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) have been identified as regulators of angiogenesis in GBM. miRNAs such as miR-21, miR-10b, and miR-26a promote angiogenesis by targeting anti-angiogenic factors, while lncRNAs such as H19 and MALAT1 inhibit angiogenesis by regulating pro-angiogenic factors. CircRNAs, such as circSMARCA5 and circBACH2, also regulate angiogenesis through various mechanisms. Similarly, signaling pathways such as the vascular endothelial growth factor (VEGF) pathway play critical roles in angiogenesis and have been targeted for GBM therapy. However, resistance to anti-angiogenic therapies is a significant obstacle in clinical practice. Developing novel therapeutic strategies targeting ncRNAs and angiogenesis is a promising approach for GBM. Potential targets include miRNAs, lncRNAs, circRNAs, and downstream signaling pathways that regulate angiogenesis. This review highlights the critical roles of ncRNAs and angiogenesis in GBM pathogenesis and the potential for new therapeutic strategies targeting these pathways to improve the prognosis and quality of life for GBM patients
... Gliomas represent a diverse group of primary brain tumors that arise from glial cells, which provide crucial support and protection to neurons in the central nervous system [1]. These tumors are associated with a spectrum of neurological symptoms, depending on their location within the brain. ...
Article
Full-text available
Background: Ubiquitin-related proteins have garnered increasing attention for their roles in tumorigenesis. Transmembrane and ubiquitin-like domain-containing 1 (TMUB1) is a recently discovered protein in the ubiquitin-like domain family, yet its involvement in glioma remains poorly understood. This study is aimed at investigating the functional significance and clinical relevance of TMUB1 in glioma. Methods: We conducted a comprehensive analysis using two cohorts: a retrospective glioma cohort from our hospital and The Cancer Genome Atlas (TCGA) cohort. The mRNA levels of TMUB1 were assessed through reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Clinical associations of TMUB1 in these cohorts were evaluated using correlation tests, chi-square tests, and survival analyses. Additionally, we performed TMUB1 knockdown in U87 and LN-229 human glioma cell lines, and cellular growth was assessed through the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Results: Our results revealed that TMUB1 expression was elevated in glioma tissues compared to normal brain tissues. Notably, lower TMUB1 expression correlated with favorable characteristics such as lower World Health Organization (WHO) grade and 1p/19q codeletion. Moreover, patients with higher TMUB1 levels in glioma tissues exhibited worse prognosis in both TCGA cohort and our retrospective cohort, underscoring its prognostic significance in gliomas. Cellular experiments demonstrated that TMUB1 silencing suppressed the growth of glioma cells. Conclusions: TMUB1 emerges as a novel and clinically relevant prognostic biomarker for gliomas. Targeting TMUB1 holds promise as a potential strategy for glioma treatment. This study contributes valuable insights into the multifaceted role of TMUB1 in glioma pathogenesis and its potential as a diagnostic and therapeutic target.
Article
Against the background of modest successes in the development of new diagnostic and therapeutic tools to improve the survival of patients with glial brain tumors, early diagnosis of this pathology remains relevant. Endogenous noncoding miRNAs that regulate the expression of target mRNAs have become attractive targets for the development of circulating biomarker-based assays, because sample acquisition does not require invasive sampling such as biopsy. Purpose of the study . To determine the levels of circulating microRNAs in the blood plasma of patients with glial tumors, meningiomas and apparently healthy donors, using high-output sequencing. Material and methods. 26 blood plasma samples were selected from the biobank data base of the National Medical Research Center for Oncology, and the total RNA was studied using the NGS sequencing method. The sample included: 2 cases of oligodendroglioma (grades 2–3), 6 – astrocytomas of 2–4 degrees of malignancy, 7 – glioblastomas of 4 degrees of malignancy, 7 – benign neoplasms (meningiomas), 4 – control (conditionally healthy donors). Results. During the primary analysis, a pool of 71 differentially expressed microRNAs was identified, the expression of which was tumor-specific: 20 microRNAs for glioblastoma, 4 microRNAs for astrocytoma, 23 microRNAs for oligodendroglioma, 24 microRNAs for meningioma. At the same time, 47 microRNAs showed increased levels in the blood plasma compared to the control group, 15 showed a corresponding decrease in levels. A comparative analysis identified microRNAs that specifically differentiate each tumor type. Conclusion . The results obtained seem promising and set the vector for further research, which will include expanding the sample and validating the identified biomarkers to determine their diagnostic value.
Chapter
1. Gliomas are categorized according to the International Classification of Diseases -Oncology, version 3 (ICD 9380–9384, 9391–9460) and the World Health Organization grade and originate from astrocytes, oligodendrocytes, a mix of these two cell types, or ependymal cells (Table 1) (Komori, Brain Tumor Pathol 39:47–50; Louis et al., Neuro Oncol 23:1231–51; ICD O 3 2013.pdf). 2. The average annual age-adjusted incidence rate (AAAIR) of all malignant and non-malignant brain and other central nervous system (CNS) tumors was 24.71 per 100,000 population (malignant 7.02, non-malignant 17.69) (Ostrom et al., Neuro Oncol 24:v1–v95). 3. Global incidence of glioma varies, with the highest rates in the United States, Canada, Australia, and Northern Europe (Ostrom et al., JAMA Oncol 4:1254–62). 4. Malignant brain tumors make up about 28.3% of all brain and other CNS tumors while non-malignant are 71.7% (Ostrom et al., Neuro Oncol 24:v1–v95). 5. Gliomas account for 81% of malignant brain tumors with glioblastoma being the most common glioma histology (Ostrom et al., Neuro Oncol 16:896–913). 6. The average annual mortality rate for malignant brain and CNS tumors is 4.41 per 100,000 (Ostrom et al., Neuro Oncol 24:v1–v95). 7. The current 5-year relative survival rate following diagnosis of a malignant brain and other CNS tumor is 35.7%, while for non-malignant is 91.8% (Ostrom et al., Neuro Oncol 24:v1–v95). 8. The 5-year survival for glioblastoma, the most common occurring glioma, is 6.9% (Ostrom et al., Neuro Oncol 24:v1–v95). 9. Estimated deaths in 2022 from malignant brain and other CNS tumors is 18,280 or 3% of all cancer deaths (https://seer.cancer.gov/statfacts/html/brain.html). 10. There have been no substantial changes in incidence of malignant brain tumors amongst the adult population.
Preprint
Full-text available
Background Diffuse intrinsic pontine glioma (DIPG), a malignant brain tumor in children, lacks effective treatment options, often presents with multiple complications during treatment, and has a poor prognosis. Objective To define the correlation between nutritional status, complications, and prognosis in pediatric patients with DIPG. Methods Clinical data were retrieved from the hospital database and follow-up, and the following clinical data of patients were organized and analyzed: age, gender, Karnofsky performance status (KPS) score at admission, treatment received, occurrence of pneumonia, onset of bed rest, overall survival (OS), 12-month survival rate, time to progression, occurrence of venous thrombosis, and prognostic nutritional index (PNI) at three stages after onset, within one week after radiotherapy, and in the last follow-up. Results A total of 34 patients met the inclusion criteria from January 2017 to June 2022. The average age was 9.0 years, and 47.1% were female. The median KPS score was 70 at admission. 29.4% of the patients were definitively diagnosed with pneumonia during the treatment of the disease, 32.4% had upper extremity venous thrombosis, and 29.4% had lower extremity venous thrombosis. The median OS of the patients was 9.2 months, and the median progression time was 4.7 months. The PNI was correlated at three stages, and it was the highest after radiotherapy (43.6 ± 8.2). Through COX survival analysis, we found that the occurrence of venous thrombosis was a disadvantageous factor for patient prognosis. The prolongation of the median progression time and the increase of the PNI at the three stages were positively correlated with the good prognosis of the patients. Conclusion High PNI sore and prevention of complications exert positive role in the prognosis of DIPG patients.
Article
Full-text available
Purpose Glioma incidence in the US seems to have stabilized over the past 20 years. It’s also not clear whether changes in glioblastoma incidence are associated with glioma mortality trends. Our study investigated trends in glioma incidence and mortality according to tumor characteristics. Methods This study obtained data from the Surveillance, Epidemiology, and End Results-9 (SEER-9) registries to calculate glioma incidence and mortality trends. Annual percent changes (APC) and 95% CIs were calculated using the Joinpoint program. Results 62,159 patients (34,996 males and 55,424 whites) were diagnosed with glioma during 1975-2018, and 31,922 deaths occurred from 1995-2018. Glioblastoma (32,893 cases) and non-glioblastoma astrocytoma (17,406 cases) were the most common histologic types. During the study period, the incidence of glioma first experienced a significant increase (APC=1.8%, [95% CI, 1.3% to 2.3%]) from 1975 to 1987, and then experienced a slight decrease (APC=-0.4%, [95% CI, -0.5% to -0.3%]) from 1987 to 2018, while the APC was 0.8% for glioblastoma, -2.0% for non-glioblastoma astrocytoma, 1.1% for oligodendroglial tumors, 0.7% for ependymoma and -0.3% for glioma NOS during the study period. Glioblastoma incidence increased for all tumor size and tumor extension except for distant. From 1995 to 2018, glioma mortality declined 0.4% per year (95% CI: -0.6% to -0.2%) but only increased in patients older than 80 years [APC=1.0%, (95% CI, 0.4% to 1.6%)]. Conclusion Significant decline in glioma incidence (1987-2018) and mortality (1995-2018) were observed. Epidemiological changes in non-glioblastoma astrocytoma contributed the most to overall trends in glioma incidence and mortality. These findings can improve understanding of risk factors and guide the focus of glioma therapy.
Article
Full-text available
The fifth edition of the WHO Classification of Tumors of the Central Nervous System (CNS), published in 2021, is the sixth version of the international standard for the classification of brain and spinal cord tumors. Building on the 2016 updated fourth edition and the work of the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy, the 2021 fifth edition introduces major changes that advance the role of molecular diagnostics in CNS tumor classification. At the same time, it remains wedded to other established approaches to tumor diagnosis such as histology and immunohistochemistry. In doing so, the fifth edition establishes some different approaches to both CNS tumor nomenclature and grading and it emphasizes the importance of integrated diagnoses and layered reports. New tumor types and subtypes are introduced, some based on novel diagnostic technologies such as DNA methylome profiling. The present review summarizes the major general changes in the 2021 fifth edition classification and the specific changes in each taxonomic category. It is hoped that this summary provides an overview to facilitate more in-depth exploration of the entire fifth edition of the WHO Classification of Tumors of the Central Nervous System.
Article
Full-text available
Background The relative importance of genetic and environmental risk factors in gliomagenesis remains uncertain. Methods Using whole-exome sequencing data from 1105 adult gliomas, we evaluate the relative contribution to cancer cell lineage proliferation and survival of single-nucleotide mutations in tumors by IDH mutation subtype and sex. We also quantify the contributions of COSMIC cancer mutational signatures to these tumors, identifying possible risk exposures. Results IDH-mutant tumors exhibited few unique recurrent substitutions—all in coding regions, while IDH-wildtype tumors exhibited many substitutions in non-coding regions. The importance of previously reported mutations in IDH1/2, TP53, EGFR, PTEN, PIK3CA and PIK3R1 was confirmed; however, the largest cancer effect in IDH wildtype tumors was associated with mutations in the low-prevalence BRAF V600E. Males and females exhibited mutations in a similar set of significantly overburdened genes, with some differences in variant sites—notably in the phosphoinositide 3-kinase (PI3K) pathway. In IDH-mutant tumors, PIK3CA mutations were located in the helical domain for females and the kinase domain for males; variants of import also differed by sex for PIK3R1. Endogenous age-related mutagenesis was the primary molecular signature identified; a signature associated with exogenous exposure to haloalkanes was identified and noted more frequently in males. Conclusions Cancer-causing mutations in glioma primarily originated as a consequence of endogenous rather than exogenous factors. Mutations in helical versus kinase domains of genes in the phosphoinositide 3-kinase (PI3K) pathway are differentially selected in males and females. Additionally, a rare environmental risk factor is suggested for some cases of glioma— particularly in males.
Article
Full-text available
Somatic mutations in the isocitrate dehydrogenase genes IDH1 and IDH2 occur at high frequency in several tumour types. Even though these mutations are confined to distinct hotspots, we show that gliomas are the only tumour type with an exceptionally high percentage of IDH1R132H mutations. Patients harbouring IDH1R132H mutated tumours have lower levels of genome-wide DNA-methylation, and an associated increased gene expression, compared to tumours with other IDH1/2 mutations (“non-R132H IDH1/2 mutations”). This reduced methylation is seen in multiple tumour types and thus appears independent of the site of origin. For 1p/19q non-codeleted glioma (astrocytoma) patients, we show that this difference is clinically relevant: in samples of the randomised phase III CATNON trial, patients harbouring tumours with IDH mutations other than IDH1R132H have a better outcome (hazard ratio 0.41, 95% CI [0.24, 0.71], p = 0.0013). Such non-R132H IDH1/2-mutated tumours also had a significantly lower proportion of tumours assigned to prognostically poor DNA-methylation classes (p < 0.001). IDH mutation-type was independent in a multivariable model containing known clinical and molecular prognostic factors. To confirm these observations, we validated the prognostic effect of IDH mutation type on a large independent dataset. The observation that non-R132H IDH1/2-mutated astrocytomas have a more favourable prognosis than their IDH1R132H mutated counterpart indicates that not all IDH-mutations are identical. This difference is clinically relevant and should be taken into account for patient prognostication.
Article
Full-text available
Head and neck cancers include cancers that originate from a variety of locations. These include the mouth, nasal cavity, throat, sinuses, and salivary glands. These cancers are the sixth most diagnosed cancers worldwide. Due to the tissues they arise from, they are collectively named head and neck squamous cell carcinomas (HNSCC). The most important risk factors for head and neck cancers are infection with human papillomavirus (HPV), tobacco use and alcohol consumption. The genetic basis behind the development and progression of HNSCC includes aberrant non-coding RNA levels. However, one of the most important differences between healthy tissue and HNSCC tissue is changes in the alternative splicing of genes that play a vital role in processes that can be described as the hallmarks of cancer. These changes in the expression profile of alternately spliced mRNA give rise to various protein isoforms. These protein isoforms, alternate methylation of proteins, and changes in the transcription of non-coding RNAs (ncRNA) can be used as diagnostic or prognostic markers and as targets for the development of new therapeutic agents. This review aims to describe changes in alternative splicing and ncRNA patterns that contribute to the development and progression of HNSCC. It will also review the use of the changes in gene expression as biomarkers or as the basis for the development of new therapies.
Article
Full-text available
Background: Non-canonical mutations of the isocitrate dehydrogenase (IDH) genes have been described in about 20-25% and 5-12% of patients with WHO grade II and III gliomas, respectively. To date, the prognostic value of these rare mutations is still a topic of debate. Methods: We selected patients with WHO grade II and III gliomas and IDH1 mutations with available tissue samples for next-generation sequencing. The clinical outcomes and baseline behaviors of patients with canonical IDH1 R132H and non-canonical IDH1 mutations were compared. Results: We evaluated 433 patients harboring IDH1 mutations. Three hundred and ninety patients (90.1%) had a canonical IDH1 R132H mutation while 43 patients (9.9%) had a non-canonical IDH1 mutation. Compared to those with the IDH1 canonical mutation, patients with non-canonical mutations were younger (p < 0.001) and less frequently presented the 1p19q codeletion (p = 0.017). Multivariate analysis confirmed that the extension of surgery (p = 0.003), the presence of the 1p19q codeletion (p = 0.001), and the presence of a non-canonical mutation (p = 0.041) were variables correlated with improved overall survival. Conclusion: the presence of non-canonical IDH1 mutations could be associated with improved survival among patients with IDH1 mutated grade II-III glioma.
Article
O ⁶-methylguanine-DNA methyltransferase (MGMT) promoter methylation is an important predictor of response to alkylating chemotherapy in glioblastomas.¹ A common method to determine MGMT promoter status is with the MGMT-STP27 algorithm which is calculated from the methylation levels of two specific CpGs (cg12434587 and cg12981137) on Illumina DNA methylation arrays.² This algorithm was constructed with data from predominantly isocitrate dehydrogenase 1 and 2 (IDH)-wildtype glioblastomas but is often extrapolated to IDH-mutant astrocytomas. However, IDH-wildtype glioblastomas usually exhibit loss of heterozygosity (LOH) of chromosome 10, whereas this copy number change is uncommon in IDH-mutant astrocytomas.³ This LOH is relevant because the MGMT gene is situated on chromosomal band 10q26, meaning that only one intact copy is left in most IDH-wildtype glioblastoma while two copies are present in IDH-mutant astrocytomas. Complete silencing of MGMT is most likely a prerequisite for efficacy of temozolomide treatment in high-grade glioma, since a reduced DNA repair (from O⁶-methylguanine to guanine) makes tumor cells more susceptible to treatment with alkylating agents that induce these defaults (from guanine to O⁶-methylguanine).4,5 The presence of two intact alleles in IDH-mutant astrocytomas, therefore, may indicate that MGMT gene methylation and subsequent temozolomide effectiveness might differ from IDH-wildtype glioblastomas. The correlation of MGMT expression with the MGMT-STP27 algorithm in tumors likely to be IDH-mutant has been assessed before, but without correlation with clinical outcome.
Article
This article reviews the current epidemiology of central nervous system tumors. Population-level basic epidemiology, nationally and internationally, and current understanding of germline genetic risk are discussed, with a focus on known and well-studied risk factors related to the etiology of central nervous system tumors.
Article
Background The CATNON trial investigated the addition of concurrent, adjuvant, and both current and adjuvant temozolomide to radiotherapy in adults with newly diagnosed 1p/19q non-co-deleted anaplastic gliomas. The benefit of concurrent temozolomide chemotherapy and relevance of mutations in the IDH1 and IDH2 genes remain unclear. Methods This randomised, open-label, phase 3 study done in 137 institutions across Australia, Europe, and North America included patients aged 18 years or older with newly diagnosed 1p/19q non-co-deleted anaplastic gliomas and a WHO performance status of 0–2. Patients were randomly assigned (1:1:1:1) centrally using a minimisation technique to radiotherapy alone (59·4 Gy in 33 fractions; three-dimensional conformal radiotherapy or intensity-modulated radiotherapy), radiotherapy with concurrent oral temozolomide (75 mg/m² per day), radiotherapy with adjuvant oral temozolomide (12 4-week cycles of 150–200 mg/m² temozolomide given on days 1–5), or radiotherapy with both concurrent and adjuvant temozolomide. Patients were stratified by institution, WHO performance status score, age, 1p loss of heterozygosity, the presence of oligodendroglial elements on microscopy, and MGMT promoter methylation status. The primary endpoint was overall survival adjusted by stratification factors at randomisation in the intention-to-treat population. A second interim analysis requested by the independent data monitoring committee was planned when two-thirds of total required events were observed to test superiority or futility of concurrent temozolomide. This study is registered with ClinicalTrials.gov, NCT00626990. Findings Between Dec 4, 2007, and Sept 11, 2015, 751 patients were randomly assigned (189 to radiotherapy alone, 188 to radiotherapy with concurrent temozolomide, 186 to radiotherapy and adjuvant temozolomide, and 188 to radiotherapy with concurrent and adjuvant temozolomide). Median follow-up was 55·7 months (IQR 41·0–77·3). The second interim analysis declared futility of concurrent temozolomide (median overall survival was 66·9 months [95% CI 45·7–82·3] with concurrent temozolomide vs 60·4 months [45·7–71·5] without concurrent temozolomide; hazard ratio [HR] 0·97 [99·1% CI 0·73–1·28], p=0·76). By contrast, adjuvant temozolomide improved overall survival compared with no adjuvant temozolomide (median overall survival 82·3 months [95% CI 67·2–116·6] vs 46·9 months [37·9–56·9]; HR 0·64 [95% CI 0·52–0·79], p<0·0001). The most frequent grade 3 and 4 toxicities were haematological, occurring in no patients in the radiotherapy only group, 16 (9%) of 185 patients in the concurrent temozolomide group, and 55 (15%) of 368 patients in both groups with adjuvant temozolomide. No treatment-related deaths were reported. Interpretation Adjuvant temozolomide chemotherapy, but not concurrent temozolomide chemotherapy, was associated with a survival benefit in patients with 1p/19q non-co-deleted anaplastic glioma. Clinical benefit was dependent on IDH1 and IDH2 mutational status. Funding Merck Sharpe & Dohme.
Article
Introduction: Global variations in survival for brain tumours are very wide when all histological types are considered together. Appraisal of international differences should be informed by the distribution of histology, but little is known beyond Europe and North America. Patients and methods: The source for the analysis was the CONCORD data base, a programme of global surveillance of cancer survival trends, which includes the tumour records of individual patients from more than 300 population-based cancer registries. We considered all patients aged 0-99 years who were diagnosed with a primary brain tumour during 2000-2014, whether malignant or non-malignant. We presented the histology distribution of these tumours, for patients diagnosed during 2000-2004, 2005-2009, and 2010-2014. Results: Records were submitted from 60 countries on five continents, 67,331 for children and 671,085 for adults. After exclusion of irrelevant morphology codes, the final study population comprised 60,783 children and 602,112 adults. Only 59 of 60 countries covered in CONCORD-3 were included, because none of the Mexican records were eligible. We defined 12 histology groups for children, and 11 histology groups for adults. In children (0-14 years), the proportion of low-grade astrocytomas ranged between 6% and 50%. Medulloblastoma was the most common sub-type in countries where low-grade astrocytoma was less commonly reported. In adults (15-99 years), the proportion of glioblastomas varied between 9% and 69%. International comparisons were made difficult by wide differences in the proportion of tumours with unspecified histology, which accounted for up to 52% of diagnoses in children and up to 65% in adults. Conclusions: To our knowledge, this is the first account of the global histology distribution of brain tumours, in children and adults. Our findings provide insights into the practices and the quality of cancer registration worldwide.