Pediatricneuro-Oncology: Fatema Malbari
Pediatricneuro-Oncology: Fatema Malbari
Fatema Malbari, MD
KEYWORDS
Pediatric brain tumors Gliomas Medulloblastoma ATRT Ependymoma
Craniopharyngioma Cancer predisposition syndromes
KEY POINTS
Central nervous system (CNS) tumors are the most common solid tumor in pediatrics and
represent the largest cause of childhood cancer–related mortality.
With the advances in molecular characterization of tumors considerable developments
have occurred impacting diagnosis, management and refining prognostication of pediat-
ric CNS tumors.
Therapeutic approaches targeting the biology of these tumors are being investigated to
improve overall survival and decrease treatment-related morbidity.
INTRODUCTION
Central nervous system (CNS) tumors are the most common solid tumor in pediatrics
and represent the largest cause of childhood cancer–related mortality.1 The incidence
of CNS tumors in children and young adults, 0 to 19 years of age, is 6.06 per 100,000
population. Gliomas are the most common tumor histology reported in this age group
and embryonal tumors have the highest incidence in children 0 to 4 years of age. Pi-
tuitary and craniopharyngeal duct tumors are the most common tumor location. The
different pediatric CNS tumor types by histology and location are shown in Fig. 1.
Risk factors for developing these tumors remain unknown aside from prior ionizing ra-
diation exposure and genetic predisposition syndromes, such as neurofibromatosis
(NF), tuberous sclerosis (TSC), Li Fraumeni syndrome (LFS), Gorlin syndrome, familial
adenomatous polyposis (FAP) syndrome, and constitutional mismatch repair defi-
ciency (CMMRD).
With the advances in molecular characterization of tumors, considerable advances
have occurred in the field of pediatric neuro-oncology.2 The understanding of the
biology has impacted diagnosis and management, and refined prognostication of pe-
diatric CNS tumors. Advances in management have led to better survival, but mortality
remains high and significant morbidity persists. Patients often have deficits from the
Fig. 1. Distribution in children and young adults (0–19 years of age) with primary CNS tu-
mors based on (A) histology and (B) location from the Central Brain Tumor Registry in the
United States. a All others include oligodendrogliomas, including anaplastic oligodendro-
gliomas, other neuroepithelial tumors, tumors of the pineal region, choroid plexus tumors,
other tumors of cranial and spinal nerves, other tumors of the meninges, hemangioma,
mesenchymal tumors, melanocytic lesions, hematopoietic neoplasms, and neoplasms un-
specified. b Other astrocytomas includes diffuse astrocytoma, anaplastic astrocytoma and as-
trocytoma variants. c All other embryonal tumors include ICD-O3 histology codes 8963/3,
9364/3, 9480/0, 9480/3, 9490/0, 9490/3, 9500/3, 9501/3, 9502/3. (Modified from Ostrom, Q.,
Cioffi, G., Gittleman, H., Patil, N., Waite, K., Kruchko, C. and Barnholtz-Sloan, J., 2019.
CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diag-
nosed in the United States in 2012–2016. Neuro-Oncology, 21(Supplement_5), pp.v1-v100.)
LOW-GRADE GLIOMA
Low-grade gliomas (LGG), the most common pediatric CNS tumor, represent 30% to
40% of all CNS tumors.4,5 Histologically LGGs (WHO grade I and II) are composed of
both astrocytic and mixed glial-neuronal tumors, with PA being the most common of
all LGGs. Other histologies include pilomyxoid astrocytoma, pleomorphic xanthroas-
trocytoma (PXA), diffuse astrocytoma, ganglioma, dysembryoplastic neuroepithelial
tumor (DNET), and subependymal giant cell astrocytoma (SEGA).6 Clinical presenta-
tion is dependent on tumor location, which can arise anywhere in the CNS. Cortical
tumors typically manifest with focal neurologic deficits, seizures, and headache. Cere-
bellar signs and increased intracranial pressure are most commonly seen with tumors
in the posterior fossa. Diencephalic tumors can present with failure to thrive. Although
LGGs are often localized, approximately 5% of patients at initial diagnosis and up to
12% of patients at time of progression will have disseminated disease.7
Pediatric Neuro-oncology 831
Management of LGG is dependent on tumor location and age of the patient. Gross
total resection (GTR) can be curative and patients can be followed with serial imaging,
with a low risk of recurrence, 5-year posterior fossa syndrome (PFS) of 94%.8 Subtotal
resection is also beneficial as tumors can become quiescent for extended periods. For
patients in whom surgical resection cannot be achieved or if LGGs are progressive,
chemotherapy, radiation, and targeted therapy are other treatment options with 10-
year OS reported at approximately 90%.9 Patients with residual or progressive dis-
ease often have a decrease in quality of life (QOL) due to the chronic nature of the
disease and associated morbidity from the tumor and treatment. Radiation therapy
(RT) can achieve comparable results, with 5-year PFS and OS of 87% and 96%,
respectively.9 However, due to potential adverse effects from RT, including neurocog-
nitive effects, vasculopathies, secondary malignancies, endocrine and growth defi-
ciencies, radiation is often reserved for children older than 10 in whom
neurocognitive effects are not as significant, in children who have exhausted multiple
treatment options, or in patients in whom RT may cause the least toxicity.10 For these
reasons, chemotherapy is usually the preferred treatment option when GTR is not
feasible, although this may change with the recent advances in molecular character-
ization of pediatric CNS tumors and the availability of targeted therapy. Chemotherapy
is not as effective as surgery or radiation but can achieve approximately 50% PFS at
5 years.11,12 The most common chemotherapy regimens used are carboplatin and
vincristine or single-agent vinblastine.
Pediatric LGG (pLGG) molecular alterations involve the RAS-MAPK pathway, with
most involving the BRAF oncogene13–19 (Table 1). In 70% to 80% of pediatric PAs,
there is a tandem duplication on chromosome 7q34 resulting in a BRAF-KIAA1549
fusion, which is almost universally found in midline and infratentorial PAs.5 The other
common alteration seen in 15% to 20% of pLGGs is a BRAF V600E mutation, which is
frequently found in supratentorial hemispheric PAs, PXAs, gangliogliomas, and
DNETs.20 It suggests a poorer prognosis with an increased risk of tumor progres-
sion.4,20 IDH mutations often observed in adults with LGG infrequently occur in ado-
lescents with diffuse astrocytomas and the clinical significance in this population
remains unclear. MAP/ERK kinase (MEK) inhibitors and BRAF inhibitors have shown
promising results in patients with these alterations.9,14 Results from phase I and II clin-
ical trials using MEK and BRAF inhibitors in patients with recurrent or refractory pLGG
has led to the development of upfront phase III clinical trials randomizing standard of
care chemotherapy to these targeted therapies in patients with newly diagnosed or
previously untreated pLGG with BRAF fusion alterations (NCT04166409) or mutations
(NCT02684058).21,22
Patients with NF1 AND TSC have an increased risk of developing pLGGs (Table 2).
PA of the optic pathway can occur in 15% to 20% of patients with NF1.23–26 Treatment
for symptomatic tumors are similar to non NF-1 PAs, with chemotherapy as the first
option because surgery is generally not feasible and RT can cause an increased
risk for secondary malignancies and vasculopathy; it is therefore reserved for patients
with progressive tumors who have exhausted other therapeutic options. Targeted
therapy with MEK inhibitors has shown promising results leading to a phase III clinical
trial randomizing standard of care chemotherapy versus a MEK inhibitor
(NCT03871257).9 SEGAs can occur in 20% of patients with TSC. If a patient is symp-
tomatic with acute focal neurologic deficits or obstructive hydrocephalus, surgical
resection is indicated. Otherwise, targeted therapy with mammalian target of rapamy-
cin inhibitors is recommended for unresectable tumors or multisystem involvement of
TSC.25,27,28
832 Malbari
Table 1
Molecular alterations of specific pediatric central nervous system tumors
HIGH-GRADE GLIOMA
Pediatric Neuro-oncology
Li Fraumeni Syndrome Chromosome 17p13, TP53 High-grade glioma, choroid plexus carcinomas,
medulloblastoma SHH subgroup
Neurofibromatosis 2 Chromosome 22q12, NF2 Ependymoma, meningioma, schwannoma
833
834 Malbari
a greater than 5-year PFS.30,31 Infants and very young children with HGG have better
OS outcomes than older children.32,33 Multiple chemotherapy regimens have been
tried in pHGG without any significant improvement in OS.14,34–36 Molecular profiling
of pHGG has identified potential targetable therapeutic options that are currently be-
ing investigated.
Molecular characterization of pHGG has led to the discovery of unique characteris-
tics that separate these tumors from adult HGG and has allowed for further subgroup-
ing.14,18 pHGGs have a novel oncogenic mutation in histone 3 variants including H3.1
K27M and H3.3 K27M as well as H3.3 G34R/V (see Table 1). The H3.1 and H3.3 K27M
mutation are classically seen in diffuse midline HGGs.14 Within the K27M mutated
pHGG, the H3.3 K 27M mutation can be seen in all midline HGGs and in most DIPG
tumors and is associated with a poorer outcome, whereas the H3.1 K27M mutation
is unique to DIPG, frequently co-occurs with an ACVR1 mutation, and portends a
slightly better prognosis. H3.3 G34R/V mutations are seen in up to a third of hemi-
spheric HGGs. As opposed to adult HGG, less than 5% of pHGGs have somatic mu-
tations in IDH1 or IDH2.14,29,37–40 pHGG with wild-type Histone 3 and IDH1/2 GBM can
be subdivided further into 3 additional groups: (1) receptor tyrosine kinase, (2) mesen-
chymal, and (3) PXA-like, distinguished by BRAF V600E mutation and deletion of
CKDN2A.14,18,37,39 PXA-like pHGG, which represents 5% to 10% of pHGGs, have
BRAF V600E mutations similar to LGGs and tend to have a better prognosis with
slightly prolonged survival. They can be treated with targeted BRAF inhibitors.14,29,39
The discovery of these somatic mutations in pHGG has led to clinical trials using tar-
geted agents to different molecular alterations. Histone deacetylate inhibitors, dopa-
mine receptor 2 antagonist, and GD2 chimeric antigen therapy are being investigated
in H3K27M mutated tumors (NCT02717455, NCT04196413, NCT03416530,
NCT04099797).41–43 Tyrosine kinase inhibitors targeting epidermal growth factor re-
ceptor, platelet-derived growth factor receptor, vascular endothelial growth factor re-
ceptor, and c-met are being explored in addition to PD-1 inhibitors for hypermutant
gliomas.20 Other treatment options include immune therapy using vaccines with
glioma-associated antibodies and other different delivery approaches of novel agents
to bypass the blood brain barrier (intra-arterial, convection enhanced delivery, intra-
nasal, intracavitary).29
generally the standard therapy for sporadic cases. CMMRD and Lynch syndrome are
associated with the development of glioblastoma. Patients with pHGG due to
mismatch repair deficiencies are treated similar to patients with sporadic GBM but
are often resistant to therapy. However, clinical responses have been seen to check-
point inhibition.14,29,49,51
MEDULLOBLASTOMA
Table 3
Clinical risk groups for medulloblastoma
Intermediate-Risk 5-
Low-Risk 5-Year OS >90% Year OS 75%–90% High-Risk 5-Year OS <60%
WNT, M0, M1 SHH, TP53 wild type Group 3, MYC and non MYC
amplified Group 3, M1
SHH, MBEN SHH, not MBEN SHH b/I
SHH g/II Group 4 SHH, TP53 mutant
Group 4 with Group 4, M1
chromosome 11 loss or 17 gain
Abbreviations: M0, no evidence of metastases in MRI or CSF cytology; M1, microscopic tumor cells
in CSF; M2, gross intracranial metastasis beyond primary site; M3, gross metastasis in subarachnoid
space; M4, metastasis outside cerebrospinal axis; MBEN, medulloblastoma with extensive nodular-
ity; OS, overall survival; SHH, sonic hedgehog.
Data from Northcott PA, Robinson GW, Kratz CP, et al. Medulloblastoma. Nature reviews. Disease
primers. 2019;5(1):11.
Table 3).57 Molecular alterations specific to WNT include monosomy 6 and a mutation
in CTNNB1 (Fig. 2; see Table 1).56 APC mutations are often associated with FAP (see
Table 2). De-escalation of therapy consisting of decreased doses of both CSI and
chemotherapy are being investigated in this subgroup, given the overall excellent
prognosis, in attempt to decrease toxicity and late effects.52
Fig. 2. MB subgroups and risk adapted management. (Data from Northcott PA, Robinson
GW, Kratz CP, et al. Medulloblastoma. Nature reviews. Disease primers. 2019;5(1):11.)
Pediatric Neuro-oncology 837
The SHH subgroup accounts for approximately 30% of all MBs and has a bimodal
age distribution, occurring more frequently in children younger than 3 or older than
16 years. These tumors usually arise from the cerebellar hemispheres and present
with appendicular ataxia.18 The most common histology seen is desmoplastic or
MB with extensive nodularity.18 SHH MB can be further categorized into 4 different
subtypes: SHH b/I, SHH g/II, SHHa, and SHHd.20,52,58 SHH b/I and SHH g/II are the
2 predominant subtypes in infants. SHH b/I is associated with poorer outcomes, often
with metastatic disease and molecularly have PTEN loss and SUFU mutations,
whereas infants with SHH g/II subtype have excellent outcomes and SMO muta-
tions.59 SHHa is the most common subtype in older children and when associated
with germline p53 mutation, portend a significantly worse prognosis. SHHd is most
commonly seen in adults and adolescents and is associated with PTCH, SMO, and
TERT promoter mutations (see Fig. 2, Table 1).56,60 SHHa and SHHd patients will
receive surgery, CSI, and chemotherapy with the addition of an SHH inhibitor in the
setting of a clinical trial (NCT01878617).
Group 3 MB represents approximately 25% of all MBs and are seen in young infants
and children. Tumors are often midline near the fourth ventricle and metastatic in 40%
of patients at initial diagnosis.61 This subgroup is associated with the worst prognosis,
with 5-year OS less than 60%.52 Histology is frequently large cell, anaplastic, or
classic. Three different subtypes of group 3 are being proposed based on genetic al-
terations and outcomes.52,56,61 MYC amplification can be seen in 20% of patients,
which is associated with poor outcomes (see Table 3). Isochromosome 17q is also
seen frequently in this subgroup (see Fig. 2, Table 1). Treatment for high-risk patients
includes surgery, CSI, and chemotherapy, with the addition of novel agents through a
clinical trial (NCT01878617) and high-dose chemotherapy with ASCR for
infants.59,62,63
Group 4 MB, the most common subgroup, represents 35% of all MB and is seen in
late childhood, early adolescence. These tumors typically arise from the midline adja-
cent to the fourth ventricle and are metastatic in a third of patients. Histology is often
classic and based on molecular features, and can be further subdivided into 3 sub-
types. Prognosis is intermediate, with 5-year OS reported at 70%. The low-risk group
has chromosome 11 loss or 17 gain and the high-risk group has metastatic disease at
diagnosis (see Table 3).57 Isochrome 17q is seen in 80% of patients in this subgroup
(see Fig. 2, Table 1).20 MYCN amplification can be seen in a small subset and is asso-
ciated with poorer outcomes. Treatment includes surgery, CSI, and chemotherapy
with the potential addition of novel agents for high-risk patients (NCT01878617).
Gorlin, FAP, and LFS are cancer predisposition syndromes associated with the
development of MB. SHH MB typically develops in infants with Gorlin syndrome.
Treatment is the same as infant SHH MB, specifically avoiding radiation, as there is
a significantly higher risk for developing secondary basal cell carcinoma in the radia-
tion field. FAP is characterized by the development of multiple adenomatous polyps
and WNT MB in adolescence (see Table 2).24,64,65 Patients are treated the same as
sporadic cases of WNT MB.
Atypical teratoid rhabdoid tumors (ATRT) are highly aggressive tumors that affect chil-
dren younger than 3 years, with a 5-year OS of 30% to 40%.66 These tumors represent
about 15% of all CNS embryonal tumors (see Fig. 1).1,67 They can arise most
commonly in the infratentorial region, followed by supratentorial and rarely in the spi-
nal cord. Approximately 20% to 40% of patients will have metastatic disease at the
838 Malbari
time of diagnosis and require extent of disease evaluation with CSF analysis and MRI
of the spine. Clinical presentation is dependent on tumor location, with infratentorial
tumors presenting with increased intracranial pressure and cranial nerve deficits
and supratentorial tumors with headaches and focal deficits.
Management of ATRT has been challenging due to the aggressive nature and young
age of patients. Multimodal therapy with surgery, conventional chemotherapy, high-
dose chemotherapy with ASCR, and focal radiation has shown dramatic improvement
in OS compared with historical therapies that included dose-intensive multi-agent
chemotherapy without radiation.66 RT has been avoided due to age; however, some
studies have shown that focal RT may not have as significant neurocognitive affects.68
A recent Children’s Oncology Group (COG) trial used this multimodal approach in
addition to age-stratified RT, which resulted in a 4-year OS of 43%.
Mutations associated with ATRT include biallelic inactivation of SMARCB1 on chro-
mosome 22, which histologically can be identified as loss of INI1 nuclear staining. Less
frequently you can see a SMARCB4 mutation.69 In addition to these somatic muta-
tions, a third of patients will have germline mutations in SMARCB1 and less commonly
SMARCA4.70 Further molecular analyses of ATRT has identified 3 main subgroups.69
Targeted therapies are under investigation through clinical trials (NCT02114229).
EPENDYMOMA
Ependymoma is the third most common malignant CNS tumor and represents
approximately 5% of all pediatric CNS tumors.1 They can arise anywhere in the neuro-
axis, but 90% of pediatric cases occur intracranially, with two-thirds in the posterior
fossa and one-third supratentorially. There is a peak incidence in early childhood.
Fewer than 10% of patients will present with metastatic disease at diagnosis and
therefore require MRI spine and CSF analyses for further evaluation.71,72 Clinical
symptoms are dependent on tumor location with infratentorial tumors often presenting
with signs of increased intracranial pressure, cranial nerve palsies, and ataxia and
supratentorial tumors presenting with focal neurologic deficits, headaches, and sei-
zures. Histologically, ependymomas can be WHO grade I (myxopapillary ependy-
moma, subependymoma), grade II (classic ependymoma), or grade III (anaplastic
ependymoma).73
Treatment is similar for all tumor locations, with maximal safe surgical resection fol-
lowed by focal RT in children older than 1. Chemotherapy is currently used for children
younger than 1, as a bridge to focal RT. A Phase III COG study was recently completed
to determine if chemotherapy provides any additional benefit after surgery; these data
are currently being analyzed. The 10-year OS is approximately 60%, but patients diag-
nosed in infancy have the lowest survival.74
Nine molecular subgroups of ependymoma have been identified, 3 in each anatomic
location: supratentorial, posterior fossa, and spinal cord (Fig. 3). Sub-ependymomas,
which primarily occur in adults, represent one molecular subgroup in each anatomic
region. The other 2 molecular subgroups for spinal cord ependymomas are distin-
guished by pathology: myxopapillary, and WHO grade II/III ependymomas. Supraten-
torial ependymomas (ST-EPN) can be subdivided by the presence of fusion
oncoproteins: C11ORF95-RELA and YAP1 (ST-EPN-RELA and ST-EPN-YAP1,
respectively) (see Table 1). The ST-RELA subgroup represents 85% of supratentorial
ependymomas and occurs in all ages, whereas the ST-YAP1 subgroup is found mostly
in pediatric patients.73,74 The ST-RELA subgroup is believed to have a poorer prog-
nosis but prospective studies did not show any differences in event-free survival be-
tween the different subgroups.72–76 The ST-YAP1 subgroup is said to have an
Pediatric Neuro-oncology 839
Fig. 3. Ependymoma molecular subgroups. (Data from Pajtler KW, Witt H, Sill M, et al. Mo-
lecular Classification of Ependymal Tumors across All CNS Compartments, Histopathological
Grades, and Age Groups. Cancer cell. 2015;27(5):728-743.)
CRANIOPHARYNGIOMA
Molecular profiling of CNS tumors are changing the current tumor classification,
management, and prognostication of these patients.
The goal of management of pediatric brain tumors is to continue to improve survival,
decrease morbidity, and improve QOL.
In patients who have received RT, recommend monitoring for potential neurologic side
effects, including neurocognitive outcomes, radiation vasculopathy, and secondary CNS
malignancies.
Targeted therapy for pediatric brain tumors is currently being investigated for different
tumor types and may be the future direction of care for these patients.
DISCLOSURE
None.
REFERENCES
1. Ostrom QT, Cioffi G, Gittleman H, et al. CBTRUS statistical report: primary brain
and other central nervous system tumors diagnosed in the United States in 2012-
2016. Neuro Oncol 2019;21(Suppl 5):v1–100.
2. Kumar R, Liu APY, Orr BA, et al. Advances in the classification of pediatric brain
tumors through DNA methylation profiling: from research tool to frontline diag-
nostic. Cancer 2018;124(21):4168–80.
3. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization
classification of tumors of the central nervous system: a summary. Acta Neuropa-
thol 2016;131(6):803–20.
4. Jones DTW, Kieran MW, Bouffet E, et al. Pediatric low-grade gliomas: next biolog-
ically driven steps. Neuro Oncol 2018;20(2):160–73.
5. Packer RJ, Pfister S, Bouffet E, et al. Pediatric low-grade gliomas: implications of
the biologic era. Neuro Oncol 2017;19(6):750–61.
6. Bergthold G, Bandopadhayay P, Bi WL, et al. Pediatric low-grade gliomas: how
modern biology reshapes the clinical field. Biochim Biophys Acta 2014;
1845(2):294–307.
7. Chamdine O, Broniscer A, Wu S, et al. Metastatic low-grade gliomas in children:
20 years’ experience at St. Jude Children’s Research Hospital. Pediatr Blood
Cancer 2016;63(1):62–70.
8. Stokland T, Liu JF, Ironside JW, et al. A multivariate analysis of factors determining
tumor progression in childhood low-grade glioma: a population-based cohort
study (CCLG CNS9702). Neuro Oncol 2010;12(12):1257–68.
9. de Blank P, Bandopadhayay P, Haas-Kogan D, et al. Management of pediatric
low-grade glioma. Curr Opin Pediatr 2019;31(1):21–7.
10. Merchant TE, Conklin HM, Wu S, et al. Late effects of conformal radiation therapy
for pediatric patients with low-grade glioma: prospective evaluation of cognitive,
endocrine, and hearing deficits. J Clin Oncol 2009;27(22):3691–7.
Pediatric Neuro-oncology 841
11. Ater JL, Zhou T, Holmes E, et al. Randomized study of two chemotherapy regi-
mens for treatment of low-grade glioma in young children: a report from the Chil-
dren’s Oncology Group. J Clin Oncol 2012;30(21):2641–7.
12. Lassaletta A, Scheinemann K, Zelcer SM, et al. Phase II weekly vinblastine for
chemotherapy-naive children with progressive low-grade glioma: a Canadian Pe-
diatric Brain Tumor Consortium Study. J Clin Oncol 2016;34(29):3537–43.
13. Hoffman LM, Salloum R, Fouladi M. Molecular biology of pediatric brain tumors
and impact on novel therapies. Curr Neurol Neurosci Rep 2015;15(4):10.
14. Sturm D, Pfister SM, Jones DTW. Pediatric gliomas: current concepts on diag-
nosis, biology, and clinical management. J Clin Oncol 2017;35(21):2370–7.
15. Northcott PA, Pfister SM, Jones DT. Next-generation (epi)genetic drivers of child-
hood brain tumours and the outlook for targeted therapies. Lancet Oncol 2015;
16(6):e293–302.
16. Ichimura K, Nishikawa R, Matsutani M. Molecular markers in pediatric neuro-
oncology. Neuro Oncol 2012;14(Suppl 4):iv90–9.
17. Chen YH, Gutmann DH. The molecular and cell biology of pediatric low-grade gli-
omas. Oncogene 2014;33(16):2019–26.
18. Gajjar A, Bowers DC, Karajannis MA, et al. Pediatric brain tumors: innovative
genomic information is transforming the diagnostic and clinical landscape.
J Clin Oncol 2015;33(27):2986–98.
19. Pollack IF, Jakacki RI. Childhood brain tumors: epidemiology, current manage-
ment and future directions. Nat Rev Neurol 2011;7(9):495–506.
20. Cacciotti C, Fleming A, Ramaswamy V. Advances in the molecular classification
of pediatric brain tumors: a guide to the galaxy. J Pathol 2020;251(3):249–61.
21. Hargrave DR, Bouffet E, Tabori U, et al. Efficacy and safety of dabrafenib in pe-
diatric patients with BRAF V600 mutation-positive relapsed or refractory low-
grade glioma: results from a phase I/IIa study. Clin Cancer Res 2019;25(24):
7303–11.
22. Nicolaides T, Nazemi KJ, Crawford J, et al. Phase I study of vemurafenib in chil-
dren with recurrent or progressive BRAF(V600E) mutant brain tumors: Pacific Pe-
diatric Neuro-Oncology Consortium study (PNOC-002). Oncotarget 2020;11(21):
1942–52.
23. Daras M, Kaley TJ. Benign brain tumors and tumors associated with phakomato-
ses. Continuum (Minneap Minn) 2015;21(2 Neuro-oncology):397–414.
24. Ranger AM, Patel YK, Chaudhary N, et al. Familial syndromes associated with
intracranial tumours: a review. Childs Nerv Syst 2014;30(1):47–64.
25. Ullrich NJ. Neurocutaneous syndromes and brain tumors. J Child Neurol 2016;
31(12):1399–411.
26. Hottinger AF, Khakoo Y. Neurooncology of familial cancer syndromes. J Child
Neurol 2009;24(12):1526–35.
27. Franz DN, Belousova E, Sparagana S, et al. Efficacy and safety of everolimus for
subependymal giant cell astrocytomas associated with tuberous sclerosis com-
plex (EXIST-1): a multicentre, randomised, placebo-controlled phase 3 trial. Lan-
cet 2013;381(9861):125–32.
28. Franz DN, Belousova E, Sparagana S, et al. Long-term use of everolimus in pa-
tients with tuberous sclerosis complex: final results from the EXIST-1 study. PLoS
One 2016;11(6):e0158476.
29. Braunstein S, Raleigh D, Bindra R, et al. Pediatric high-grade glioma: current mo-
lecular landscape and therapeutic approaches. J Neurooncol 2017;134(3):
541–9.
842 Malbari
30. Pollack IF. The role of surgery in pediatric gliomas. J Neurooncol 1999;42(3):
271–88.
31. Wisoff JH, Boyett JM, Berger MS, et al. Current neurosurgical management and
the impact of the extent of resection in the treatment of malignant gliomas of
childhood: a report of the Children’s Cancer Group trial no. CCG-945.
J Neurosurg 1998;89(1):52–9.
32. Espinoza JC, Haley K, Patel N, et al. Outcome of young children with high-grade
glioma treated with irradiation-avoiding intensive chemotherapy regimens: final
report of the Head Start II and III trials. Pediatr Blood Cancer 2016;63(10):
1806–13.
33. Sanders RP, Kocak M, Burger PC, et al. High-grade astrocytoma in very young
children. Pediatr Blood Cancer 2007;49(7):888–93.
34. Turner CD, Chi S, Marcus KJ, et al. Phase II study of thalidomide and radiation in
children with newly diagnosed brain stem gliomas and glioblastoma multiforme.
J Neurooncol 2007;82(1):95–101.
35. Wolff JE, Wagner S, Reinert C, et al. Maintenance treatment with interferon-
gamma and low-dose cyclophosphamide for pediatric high-grade glioma.
J Neurooncol 2006;79(3):315–21.
36. Grill J, Massimino M, Bouffet E, et al. Phase II, open-label, randomized, multi-
center trial (HERBY) of bevacizumab in pediatric patients with newly diagnosed
high-grade glioma. J Clin Oncol 2018;36(10):951–8.
37. Korshunov A, Schrimpf D, Ryzhova M, et al. H3-/IDH-wild type pediatric glioblas-
toma is comprised of molecularly and prognostically distinct subtypes with asso-
ciated oncogenic drivers. Acta Neuropathol 2017;134(3):507–16.
38. Chamdine O, Gajjar A. Molecular characteristics of pediatric high-grade gliomas.
CNS Oncol 2014;3(6):433–43.
39. Jones C, Karajannis MA, Jones DTW, et al. Pediatric high-grade glioma: biolog-
ically and clinically in need of new thinking. Neuro Oncol 2017;19(2):153–61.
40. Panditharatna E, Yaeger K, Kilburn LB, et al. Clinicopathology of diffuse intrinsic
pontine glioma and its redefined genomic and epigenomic landscape. Cancer
Genet 2015;208(7–8):367–73.
41. Lin GL, Wilson KM, Ceribelli M, et al. Therapeutic strategies for diffuse midline gli-
oma from high-throughput combination drug screening. Sci Transl Med 2019;
11(519):eaaw0064.
42. Chi AS, Tarapore RS, Hall MD, et al. Pediatric and adult H3 K27M-mutant diffuse
midline glioma treated with the selective DRD2 antagonist ONC201. J Neurooncol
2019;145(1):97–105.
43. Mount CW, Majzner RG, Sundaresh S, et al. Potent antitumor efficacy of anti-GD2
CAR T cells in H3-K27M(1) diffuse midline gliomas. Nat Med 2018;24(5):572–9.
44. Grimm SA, Chamberlain MC. Brainstem glioma: a review. Curr Neurol Neurosci
Rep 2013;13(5):346.
45. Hoffman LM, Veldhuijzen van Zanten SEM, Colditz N, et al. Clinical, radiologic,
pathologic, and molecular characteristics 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
2018;36(19):1963–72.
46. Korones DN, Fisher PG, Kretschmar C, et al. Treatment of children with diffuse
intrinsic brain stem glioma with radiotherapy, vincristine and oral VP-16: a Chil-
dren’s Oncology Group phase II study. Pediatr Blood Cancer 2008;50(2):227–30.
Pediatric Neuro-oncology 843
a report from the children’s oncology group trial ACNS0333. J Clin Oncol 2020;
38(11):1175–85.
67. Nesvick CL, Lafay-Cousin L, Raghunathan A, et al. Atypical teratoid rhabdoid tu-
mor: molecular insights and translation to novel therapeutics. J Neurooncol 2020;
150(1):47–56.
68. Merchant TE, Mulhern RK, Krasin MJ, et al. Preliminary results from a phase II trial
of conformal radiation therapy and evaluation of radiation-related CNS effects for
pediatric patients with localized ependymoma. J Clin Oncol 2004;22(15):
3156–62.
69. Ho B, Johann PD, Grabovska Y, et al. Molecular subgrouping of atypical teratoid/
rhabdoid tumors-a reinvestigation and current consensus. Neuro Oncol 2020;
22(5):613–24.
70. Eaton KW, Tooke LS, Wainwright LM, et al. Spectrum of SMARCB1/INI1 mutations
in familial and sporadic rhabdoid tumors. Pediatr Blood Cancer 2011;56(1):7–15.
71. Fangusaro J, Van Den Berghe C, Tomita T, et al. Evaluating the incidence and util-
ity of microscopic metastatic dissemination as diagnosed by lumbar cerebro-
spinal fluid (CSF) samples in children with newly diagnosed intracranial ependy-
moma. J Neurooncol 2011;103(3):693–8.
72. Upadhyaya SA, Robinson GW, Onar-Thomas A, et al. Molecular grouping and
outcomes of young children with newly diagnosed ependymoma treated on the
multi-institutional SJYC07 trial. Neuro Oncol 2019;21(10):1319–30.
73. Pajtler KW, Mack SC, Ramaswamy V, et al. The current consensus on the clinical
management of intracranial ependymoma and its distinct molecular variants.
Acta Neuropathol 2017;133(1):5–12.
74. Pajtler KW, Witt H, Sill M, et al. Molecular classification of ependymal tumors
across all CNS compartments, histopathological grades, and age groups. Can-
cer Cell 2015;27(5):728–43.
75. Ramaswamy V, Hielscher T, Mack SC, et al. Therapeutic impact of cytoreductive
surgery and irradiation of posterior fossa ependymoma in the molecular era: a
retrospective multicohort analysis. J Clin Oncol 2016;34(21):2468–77.
76. Merchant TE, Bendel AE, Sabin ND, et al. Conformal radiation therapy for pedi-
atric ependymoma, chemotherapy for incompletely resected ependymoma,
and observation for completely resected, supratentorial ependymoma. J Clin On-
col 2019;37(12):974–83.
77. Mack SC, Witt H, Piro RM, et al. Epigenomic alterations define lethal CIMP-
positive ependymomas of infancy. Nature 2014;506(7489):445–50.
78. Pajtler KW, Wen J, Sill M, et al. Molecular heterogeneity and CXorf67 alterations in
posterior fossa group A (PFA) ependymomas. Acta Neuropathol 2018;136(2):
211–26.
79. Evans DGR, Salvador H, Chang VY, et al. Cancer and central nervous system tu-
mor surveillance in pediatric neurofibromatosis 2 and related disorders. Clin Can-
cer Res 2017;23(12):e54–61.
80. Muller HL, Merchant TE, Warmuth-Metz M, et al. Craniopharyngioma. Nat Rev Dis
Primers 2019;5(1):75.
81. Apps JR, Carreno G, Gonzalez-Meljem JM, et al. Tumour compartment transcrip-
tomics demonstrates the activation of inflammatory and odontogenic pro-
grammes in human adamantinomatous craniopharyngioma and identifies the
MAPK/ERK pathway as a novel therapeutic target. Acta Neuropathol 2018;
135(5):757–77.
Pediatric Neuro-oncology 845
82. Donson AM, Apps J, Griesinger AM, et al. Molecular analyses reveal inflamma-
tory mediators in the solid component and cyst fluid of human adamantinomatous
craniopharyngioma. J Neuropathol Exp Neurol 2017;76(9):779–88.
83. Grob S, Mirsky DM, Donson AM, et al. Targeting IL-6 is a potential treatment for
primary cystic craniopharyngioma. Front Oncol 2019;9:791.
84. Whelan R, Prince E, Gilani A, et al. The inflammatory milieu of adamantinomatous
craniopharyngioma and its implications for treatment. J Clin Med 2020;9(2):519.
85. Coy S, Rashid R, Lin JR, et al. Multiplexed immunofluorescence reveals potential
PD-1/PD-L1 pathway vulnerabilities in craniopharyngioma. Neuro Oncol 2018;
20(8):1101–12.