Abstract
Purpose of review:
Pediatric low-grade gliomas and glioneuronal tumors (pLGG) account for approximately 30% of pediatric CNS neoplasms, encompassing a heterogeneous group of tumors of primarily glial or mixed neuronal-glial histology. This article reviews the treatment of pLGG with emphasis on an individualized approach incorporating multidisciplinary input from surgery, radiation oncology, neuroradiology, neuropathology and pediatric oncology to carefully weigh the risks and benefits of specific interventions against tumor-related morbidity. Complete surgical resection can be curative for cerebellar and hemispheric lesions, while use of radiotherapy is restricted to older patients or those refractory to medical therapy. Chemotherapy remains the preferred first line therapy for adjuvant treatment of the majority of recurrent or progressive pLGG.
Recent findings:
Technologic advances offer the potential to limit volume of normal brain exposed to low doses of radiation when treating pLGG with either conformal photon or proton RT. Recent neurosurgical techniques such as laser interstitial thermal therapy offer a ‘dual’ diagnostic and therapeutic treatment modality for pLGG in specific surgically inaccessible anatomical locations. The emergence of novel molecular diagnostic tools has enabled scientific discoveries elucidating driver alterations in mitogen activated protein kinase (MAPK) pathway components and enhanced our understanding of the natural history (oncogenic senescence). Molecular characterization strongly supplements the clinical risk stratification (age, extent of resection, histological grade) to improve diagnostic precision and accuracy, prognostication and can lead to the identification of patients who stand to benefit from precision medicine treatment approaches.
Summary:
The success of molecular targeted therapy (BRAF inhibitors and/or MEK inhibitors) in the recurrent setting has led to a gradual and yet significant paradigm shift in the treatment of pLGG. Ongoing randomized trials comparing targeted therapy to standard of care chemotherapy are anticipated to further inform the approach to upfront management of pLGG patients.
Keywords: Pediatric low-grade gliomas, glioneuronal tumors, neurosurgery, chemotherapy, radiotherapy, molecular targeted therapy, MEK inhibitors, BRAF inhibitors
INTRODUCTION
Pediatric low-grade gliomas and glioneuronal tumors (subsequently referred to as pLGG) represent the most frequently encountered brain tumors accounting for nearly 30% of pediatric CNS neoplasms overall. pLGG are defined as grade 1 or 2 per the recent World Health Organization (WHO) 2021 classification (CNS5) and encompass a heterogeneous group of tumors of primarily glial histology, including astrocytic and/or oligodendroglial, as well as tumors of mixed neuronal-glial morphology (see Table 1) [1].
Table 1.
WHO classification | Molecular alteration |
---|---|
Pediatric-type diffuse low-grade gliomas Diffuse astrocytoma, MYB- or MYBL1-altered Angiocentric glioma Polymorphous low-grade neuroepithelial tumor of the young Diffuse low-grade glioma, MAPK pathway-altered |
MYB- or MYBL1-altered MYB-altered FGFR2/3 Fusions (30-40%) BRAF p.V600E (30-40%) - |
Circumscribed astrocytic gliomas Pilocytic astrocytoma High-grade astrocytoma with piloid features Pleomorphic xanthoastrocytoma Subependymal giant cell astrocytoma Chordoid glioma Astroblastoma, MN1-altered |
KIAA1549-BRAF (70-80%) FGFR1-TACC1 (3-5%) FGFR1 SNV (3-5%) BRAF p.V600E (3-5%) Other BRAF Fusions (2-5%) CRAF Fusions (2-5%) PTPN11 SNV (2-5%) KRAS/HRAS SNV (2-5%) BRAF p.V600E TSC1/2 SNV (85-95%) PRKCA SNV (80-90%) MN1 |
Glioneuronal and neuronal tumors Ganglioglioma Desmoplastic infantile ganglioglioma/astrocytoma Dysembryoplastic neuroepithelial tumor Diffuse glioneuronal tumor with oligodendroglioma-like features and nuclear clusters Papillary glioneuronal tumor Rosette-forming glioneuronal tumor Myxoid glioneuronal tumor Diffuse leptomeningeal glioneuronal tumor Gangliocytoma Multinodular and vacuolating neuronal tumor Dysplastic cerebellar gangliocytoma (Lhermitte-Duclos disease) Central neurocytoma Extraventricular neurocytoma Cerebellar liponeurocytoma |
BRAF p.V600E (40-50%) KIAA1549-BRAF (10-15%) BRAF pV600E/D (40-60%) FGFR1 SNV (5-10%) KIAA1549-BRAF (2-5%) FGFR1-TKD duplication (20-30%) FGFR1 SNV (20-30%) FGFR1-TACC1 (10-15%) Other RTK SNV/Fusions (5-10%) BRAF p.V600E (5-10) SLC44A1-PRKCA (80-90%) PIK3CA SNV (20-30%) KIAA1549-BRAF (20-30%) FGFR1 SNV (20-30%) MAP2K1 SNV/Indel (50-60%) BRAF p.V600E (5-10%) Other BRAF SNV (5-10%) FGFR2 Fusions (3-5%) |
There are some important considerations which significantly impact the treatment approach to pLGG.
First, overall survival (OS) is excellent for the majority of pLGG [2], with 20-year overall survival rates up to 87% [3]. However, patients can experience significant morbidity including vision loss, epilepsy, endocrine dysfunction, motor disability, neurocognitive dysfunction, and decreased quality of life [3, 4]. Therapy is indicated only in the event of sub-totally resected tumors with clinical progression or unequivocal non enhancing (T2/FLAIR MRI sequences) radiographic progression with likelihood of significant morbidity. Treatment goals should not only include long-term tumor control (i.e., radiographic responses), but also minimization of treatment-related morbidity and improvement in functional outcomes (such as visual acuity and motor abilities), quality of life, and neuro-psychological assessments.
Devising an optimal clinical management plan is complex and requires multidisciplinary input from surgery, radiation oncology, neuroradiology and pediatric oncology and should carefully weigh the risks and benefits of tumor directed interventions against tumor related morbidity. Therefore, a one size fits all approach is unlikely to be effective.
For some patients, the pLGG maybe represent the initial manifestation of an underlying tumor predisposition syndrome (neurofibromatosis type 1 [NF1], tuberous sclerosis [TS]) which carries important implications for genetic counseling, tumor surveillance and treatment choices.
Extent of surgical resection, tumor location, histological grade/subtype and age at diagnosis were historically considered for clinical risk stratification and importantly, remain so to this day. However, it has long been recognized that differences in patient outcomes cannot be explained by clinical variables alone. The past two decades have witnessed the emergence of novel molecular diagnostic tools fueling seminal scientific discoveries which have shed light on the fundamental events contributing to gliomagenesis [5]. With few exceptions, pLGG are exclusively driven by a diverse array of genetic alterations in mitogen activated protein kinase (MAPK) pathway components, resulting in constitutive activation of downstream effector signaling pathways including RAF/MEK/ERK and PI3K/AKT/mTOR [6, 7]. Molecular characterization is now an integral component of contemporary neuropathology and can strongly supplement the clinical risk stratification to improve diagnostic precision and accuracy [8, 9], prognostication (see Table 2) [10] and lead to the identification of patients who stand to benefit from precision medicine treatment approaches [11].
Mutations in MAPK pathway component genes (RAS, RAF and NF1) can trigger cellular senescence. This phenomenon, termed oncogene induced senescence (OIS) likely accounts for the relatively benign behavior of pLGG and lack of transformation to higher-grade gliomas (with specific exceptions such as BRAFV600E) in the absence of additional cooperating mutations [12].
For pLGG, a specific genetic alteration may either be disease defining (MYB-QKI fusion in angiocentric glioma) or play only a supporting role in diagnosis because of its enrichment in specific tumor histologies (BRAFV600E mutation in PXA) (see Table 1).
- Despite the common theme of constitutively activated MAPK signaling, there exists substantial molecular heterogeneity. The specific underlying alteration may variably activate different MAPK downstream effector pathways (PI3K/mTOR pathway in SEGA and the RAF/MEK/ERK pathway in BRAF/NF1 altered pLGG). Even the mechanisms driving signaling via a common effector pathway (RAF/MEK/ERK) can be distinct based on -
- the type of molecular alteration (BRAFV600E mutant pLGG signal as monomers while BRAF fusion pLGG signal as dimers) which carries important therapeutic implications (BRAF monomer inhibitors cause paradoxical activation and accelerate tumor growth in BRAF fused pLGG and should be avoided)
- which specific nodes in the MAPK pathway are disrupted because of complex feedback loops that exist between these nodes and effector pathways
While pLGG usually appear fairly circumscribed on imaging compared to pediatric high-grade gliomas (pHGGs), some lesions demonstrate a more diffuse growth pattern. These “pediatric-type” diffuse gliomas [1] constitute a new group including 4 entities with some histological overlap but distinct molecular features [1, 13]. They should be differentiated from “adult-type” diffuse gliomas which are more aggressive and require a different approach to diagnosis and management.
Table 2.
Risk group | Alteration type | Outcomes |
---|---|---|
Low | Gene fusions (BRAF-KIAA1549, FGFR1-TACC1) Germline NF1 mutations |
10-year PFS of 67% and OS of 98% 20-year PFS and OS of 58% and 96%, respectively. [7] |
Intermediate |
BRAF p.V600E without CDKN2A deletion, FGFR1 SNV or MET mutations Co-occur with other alterations |
10-year PFS and OS of 35% and 90% 20-year PFS of 27% and 20-year OS of 81%, respectively. [7] |
High | H3.3 p.K27M, or BRAF p.V600E with CDKN2A deletion | 10-year PFS and OS of 0% and 35% 10-year PFS and OS of 0% and 60%, respectively. [7] |
Keeping these principles in mind, a brief overview of the management options, including surgery, radiation therapy (RT), and chemotherapy, is discussed in this review. In addition, our growing understanding of the molecular underpinnings of pLGG is leading to the increasing utilization of novel targeted molecular agents mostly in the recurrent, and in specific scenarios, in the upfront setting, signaling a gradual, yet significant paradigm shift [14, 15]. Therefore, we emphasize the relevant pathophysiology and genetics that significantly impact the clinical approach within the framework of the most frequently encountered pLGG.
Surgery
The surgical treatment of pLGG is complex and requires multi-disciplinary involvement to delineate the goals of surgery. Often, these lesions are discovered incidentally or in the setting of seizure or headache without significant neurological deficit present. In pLGG, maximal safe excision with an intent to cure is the preferred treatment approach and should always be considered. However, this is not always feasible due to location, infiltrative nature, or molecular phenotype of the lesion. Surgical objectives fall into the broad categories of total tumor extirpation, cytoreduction, or biopsy (stereotactic needle and endoscopic).
To address the questions of efficacy and specific type of surgical intervention, clinical, radiographic, diagnostic, and treatment factors must be considered. Currently, there is no validated diagnostic imaging or biomarker available that can predict the clinical grade of the lesion. In pLGG, goals of surgical intervention include symptom palliation and improvement of neurological condition, obtainment of tissue for molecular and histological diagnosis, and reduction in risk of continued tumor growth or malignant transformation.[16] For symptomatic or asymptomatic non-infiltrative and surgically accessible lesions with confidence around a WHO grade 1 diagnosis for which resection is associated with acceptable risk, maximal safe surgical intervention with intent to cure is the preferred treatment approach. Gross total resection of pLGG is significantly associated with improved overall survival (OS), with 10-year OS rates approximating 90%, and is the main predictor of PFS [2, 17–20]. Notably, over 50% of children with residual tumor volume after resection have no disease progression at 5 years, and these patients have excellent long-term survival. Therefore, even though complete resection should be a goal, the benefit of possibly prolonging PFS should be carefully weighed with the risk of neurologic deficit caused by an aggressive resection. While the rates of malignant transformation of pLGG are estimated to be less than 10%, this can be a source of lifelong concern for the patient and their families, and require frequent and longitudinal monitoring.[21]
For lesions that are surgically inaccessible principally being defined by an infiltrative growth pattern on imaging, have known biological behavior that would not warrant resection, or for which resection would leave an unacceptable functional outcome (optic pathway gliomas), the surgical objectives narrow to tissue sampling for diagnosis and molecular classification and debulking in the setting of mass effect and hydrocephalus. Further research is needed to quantify the minimal amount of tissue that is required for histological and molecular analysis in order to better understand if stereotactic or endoscopic biopsies are sufficient for providing diagnostic tissue samples. Laser interstitial thermal therapy is another surgical tool to target focal, surgically inaccessible tumors, such as lesions in the dominant temporal lobe, medial to the internal capsule, or cerebral peduncle [22, 23].
With molecular classification increasing in the role of therapeutic management, tissue sampling should be performed early in diagnosis. In general, the benefits of accurate molecular diagnosis far outweigh the risks of surgical intervention and can often act synergistically with other surgical goals of symptom palliation, restoration of neurological function, or decreasing risk of tumor growth or transformation. However, for certain slow growing pathologies with a well-understood natural history, exposing children to the risk of surgical intervention involving highly functional structures may not be necessary. A reappraisal of the need for surgical intervention in these cases is warranted. In the case of optic pathway and hypothalamic gliomas, surgical intervention is rarely indicated except when the treating team is requesting tissue for diagnosis prior to initiating treatment or to debulk the lesion due to significant mass effect. Given the risks involved with tissue sampling including vision loss, the field should investigate the role of upfront chemotherapy or molecular targeted therapy.[24, 25]
Radiation Therapy
Radiation therapy (RT) is an effective management strategy in both the upfront and salvage treatment settings for pLGG. Historically, RT was the preferred primary treatment for rapidly progressive or unresectable tumors, with 10-year progression-free (PFS) and overall survival (OS) rates of 70% and 80%, respectively.[26–29] RT has also been used in the adjuvant setting, particularly when surgery is limited to partial resection or biopsy for tumors located in the optic pathway, hypothalamus, deep midline structures, and brainstem.[18, 19] Since PFS is significantly reduced after incomplete resections,[18, 30, 31] adjuvant RT is considered for this scenario; however, there is a lack of consensus for this use due in part to the absence of randomized prospective trials.[18, 20, 32] One randomized prospective phase 3 trial sought to evaluate neurosurgical and radiotherapeutic treatments of pLGG;[33] however, the trial was closed early due to practitioner bias in treatment selection. Our current understanding is therefore built upon conflicting retrospective studies, which have suggested that while adjuvant RT may improve PFS, this benefit does not necessarily translate into improved OS.[18, 31, 34] For instance, while one study demonstrated that adjuvant RT improved PFS when complete resection was not achieved (89% vs. 49%, P < 0.003), others showed no improvement in PFS with postoperative RT.[20, 32] This unclear PFS benefit suggests that RT could be delayed until the time of progression.[35] However, Tsang et al found initial RT improved event-free survival for optic pathway and hypothalamic gliomas, suggesting that delaying RT with chemotherapy may not be without consequence for certain high risk patients.[36] Therefore, in the adjuvant setting, RT is used for patients with symptomatic residual tumors or for those who have radiologic or symptomatic progression.[18]
RT is also favored as part of the management strategy in older children who have failed multiple lines of systemic agents. Historically, the rationale for delaying RT revolved around concern for RT-associated toxicities, namely cognitive decline,[37, 38] endocrine dysfunction,[39] secondary malignancies,[36] vascular damage,[36, 40] and growth abnormalities,[41] the severity of which is highly dependent on the location of the tumor and patient age (<10-years-old).[36–38] Much of the concern about RT-associated toxicity is based on long-term toxicity data derived from trials performed in the 1970s-1990s, which used 2-dimensional RT techniques that did not enable conformal radiation dose delivery. Major technological advancements have since been made to reduce the radiation dose delivered to normal structures surrounding the tumor: first with 3-dimensional conformal external beam RT (3D-CRT) followed in the 2000s by intensity-modulated RT (IMRT). Importantly, the introduction of proton therapy minimized radiation exit dose,[42, 43] largely contributing to its increasing role in pediatric patients. Some studies suggest that proton therapy may even improve both patient quality of life and cost effectiveness of treatment for pediatric brain tumors.[44, 45] Moreover, outcomes with proton therapy are excellent, with one institutional review over a median follow-up of 11 years reporting 8-year PFS and OS rates of 83% and 100%, respectively.[46] In this study, neurocognitive function was not shown to decline overall; however, there was significant cognitive decline in young children (<7-years-old) and in patients who received significant dose to the left temporal lobe or hippocampus. Endocrine dysfunction was identified in those with higher doses to the hypothalamus or pituitary, and two patients developed moya moya disease. Second cancers are a rare, but feared, potential long-term toxicity after RT. In a large single-institutional study of 1,713 children treated with proton therapy (1,040 for central nervous system [CNS] tumors) over a median follow up of 3.3 years, 5- and 10-year cumulative incidences of second tumors were 0.8% and 3.1%, respectively.[47] Importantly, all but one patient who developed a second tumor were irradiated at age 5 years or younger, and there was a significant relationship between tumor predisposition syndromes and second tumor development. A recent report of 945 pediatric patients treated with proton therapy for CNS tumors reported a low 5-year cumulative incidence of secondary neoplasms of 2.4%.[48] In a recently published prospective study of 174 pediatric patients with LGG treated with proton therapy, 5-year PFS and OS rates were 84% and 92%, respectively, at a median follow-up of 4.4 years.[49] Severe late toxicities (brainstem necrosis, symptomatic vasculopathy, radiation retinopathy, and fatal secondary malignancy) occurred in four patients. While radiation-related toxicity must be acknowledged, studies performed in the modern era have encouraging results and the long latency of toxicity must therefore be appreciated within the context of rapid advances in the field.[50]
In addition to radiation planning advances, improvements in neuroimaging (such as multiparametric magnetic resonance imaging) and patient immobilization have further contributed to accurate delivery of highly conformal radiation dose with smaller target margins. Specifically, planning margins can be safely limited to 10 mm or less to protect adjacent normal structures when delivering the typical dose of 54 Gray (Gy) in 30 fractions.[38] Stereotactic RT is a technique that has demonstrated excellent local control for patients with small (<5 cm) tumors. Dana Farber Cancer Institute reported their 5-year results with this technique using a median dose of 52.5 Gy in 1.8 Gy fractions, demonstrating PFS and OS rates of 83% and 98%, respectively.[51] Importantly, in this study there were no marginal (“near miss”) disease recurrences, meaning that treatment margins can be minimized to reduce radiation-related toxicities without compromising local control.
In summary, technologic advances offer the potential to limit volume of normal brain exposed to low doses of radiation when treating pLGG with either conformal photon[52] or proton[49] RT. The severity of radiation-related toxicities is most often dependent on the age at the time of RT and the location of the tumor. Given the chronic nature of this disease and long latency of treatment-related toxicities, long-term data are needed, but RT in the modern era offers a safe, effective strategy for local tumor control.
Chemotherapy
Over the past 3 decades multiple prospective clinical trials (single arm or randomized) in children with progressive LGGs were completed (see Table 3). Most patients had diencephalic tumors (optic nerves/chiasm/hypothalamus/optic tracks/optic radiations) with more recent studies including a higher proportion of non-diencephalic tumors including brainstem tumors. Inconsistencies in the inclusion criteria and imaging criteria for response assessment (enhancing versus non enhancing, central vs local review), lack of correlation between radiographic responses and PFS and the long duration of follow up resulting in delayed reporting of results hamper the ability to directly compare results across studies. Nonetheless, these studies demonstrate very similar 5-year PFS rates and better 5-year disease control in children with NF1 compared to those without.
Table 3.
Study | Design | Chemotherapy | N | EFS/PFS |
---|---|---|---|---|
Packer et al [132] | Single arm, multicenter | CV | 63 NF1 (n=15) |
2-y 79 ± 11% 2-y 79 ± 11% |
POG [133] | Single arm | C | 29 NF1 (n=21) |
3-yr 51 ± 9% 5-y 61 ± 12% |
SFOP [134] | Single arm | PCV/CARBO; VP16/CPDD VCR/CYTOX |
62 NF1 (n=23) |
3-yr 42 ± 12% 3-y 62 ± 13% |
HIT-LGG-1996 [135] | Single arm | CV | 161 NF1 (n=55) |
5-yr 47% 5-y 68% |
COG A9952 [53] | Randomized multicenter | TPCV CV |
137 137 NF1 – single arm – CV only (n=127) |
5-yr 52% ± 5% 5-yr 39% ± 4% (ns) 5-yr 69 ± 4% |
COG (ACNS0223) [136] | Single arm | CV + TMZ | 66 | 5-yr 46 ± 13% |
(SIOP European Brain Tumor Committee) [137] | Randomized multicenter |
CV CV + VP-16 |
497 | 5-yr 46% 5-yr 45% (ns) |
[138] | Single arm | Vinblastine | 54 NF1 (n=15) |
5-yr 42% 5-yr 85% |
Chemotherapy regimens in newly-diagnosed pLGG achieve 3-year PFS between 50–80% depending on the regimen. Frequently used chemotherapy regimens include i) carboplatin alone or in combination with vincristine (CV), ii) thioguanine, procarbazine, CCNU and vincristine (TPCV) and iii) vinblastine alone. Although carboplatin and vincristine may offer slightly inferior PFS compared to TPCV (not statistically significant) [53], the combination avoids the risks of secondary malignancy and infertility posed by the TPCV regimen and is therefore preferred given the indolent nature of pLGG to mitigate long term sequelae. Carboplatin monotherapy affords the advantage of monthly administration (as opposed to weekly administration with CV regimen) obviating the need for a central line and carries reduced risk of chemotherapy hypersensitivity reactions and infections.
Molecular genetics and targeted therapy
The most common alterations in pLGG are loss of neurofibromin in the context of patients with NF1, and in non-NF1-related pLGG, the fusion and tandem duplication of BRAF with KIAA1549 (class II BRAF alteration which signals as BRAF dimers) and the BRAFV600E mutation (class I BRAF alteration which signals as BRAF monomers), respectively [5, 7, 54]. These seminal discoveries opened the door to precision oncology trials targeting the RAS-MAP kinase pathway with MEK inhibitors (MEKi) (which function downstream of RAF) and BRAF inhibitors (BRAFi) (first generation=BRAF monomer inhibitors and second generation=BRAF dimer inhibitors or pan-RAF inhibitors) [11, 55].
A). pLGG associated with tumor predisposition syndromes (TPS)
Neurofibromatosis type 1 (NF1)
NF1 is an autosomal dominant (AD) tumor predisposition syndrome and affected individuals develop a combination of dermatologic, skeletal, ophthalmic, and neurologic findings at typical ages of onset. Optic pathway gliomas (OPGs) and brainstem low-grade gliomas (LGGs) are the most common intracranial neoplasms found in NF1. Nearly a third of children with OPG have germline mutations in NF1. Conversely, OPGs are detectable in approximately 15% of NF1 patients, usually before the age of 7 years and bilateral OPGs are detected exclusively in NF1. Most of these tumors are WHO grade 1 pilocytic astrocytomas (PA), although most patients are diagnosed based on imaging without a biopsy. Brainstem gliomas present in late childhood (mean age 7 years), exhibit mass effect on T2 and increased signal on T1 weighted images (unlike UBOs), are more indolent than sporadic brainstem gliomas, and may regress spontaneously. Patients with NF1 also often exhibit multiple T2 hyperintense lesions, mainly in the basal ganglia and brainstem which are referred to as unidentified bright objects (UBOs) and undergo spontaneous regression.
The NF1 locus maps to chromosome 17q11.2 and encodes neurofibromin, a protein which harbors a GTPase–activating protein domain that functions to silence RAS in its activated form. Biallelic inactivation of the NF1 gene leads to deregulated RAS activity, which initiates downstream signaling by activating the RAF/MEK/ERK and the Akt/mTOR pathways.[56] A comprehensive molecular profiling study in NF1 patients using WES of tumor and matched blood germline DNA demonstrated that LGGs have a low mutational burden and primarily exhibit loss of heterozygosity (LOH) in the NF1 region along with alterations in genes (FGFR1, PIK3CA) encoding component of the MAPK pathway [57].
Treatment
Patients with asymptomatic OPG or LGG are managed conservatively, with imaging surveillance and close clinical/ophthalmological follow-up [58]. Surveillance neuroimaging in asymptomatic children with NF1 has not been shown to reduce the incidence of visual loss, with MRI recommended only for patients with ophthalmological findings suggestive of an OPG, such as proptosis, optic disc pallor or vision loss.[58] Only a third of patients with NF1-OPG require treatment, with the primary goal to preserve vision. Radiotherapy is generally avoided given the increased risk of secondary neoplasms [59] and moya moya disease [60], with carboplatin or vinblastine based regimens most commonly used. Recently, molecular targeted therapy with MEK inhibitors has demonstrated impressive anti-tumor effects and is increasingly being considered as second-line therapy for NF1-associated LGGs.[11] The current COG (children’s oncology group) study ACNS1831 is a randomized phase 3 trial testing anti-tumor efficacy and visual outcomes during treatment with selumetinib (MEK inhibitor) compared with standard chemotherapy and may alter the current treatment paradigm for NF1-associated LGG (NCT03871257).
Tuberous sclerosis complex (TSC)
TSC is an autosomal dominant multisystem condition characterized by the triad of adenoma sebaceum, epilepsy and mental retardation [61–63]. CNS lesions are the major cause for TSC-related morbidity and mortality.[64] The vast majority of TSC patients develop cortical tubers before birth and subependymal nodules during the first years of life [64–66]. Cortical tubers are benign hamartomas resulting from abnormal neuronal migration and disordered differentiation and demonstrate dysplastic neurons along with giant eosinophilic cells of mixed glioneuronal lineage. Subependymal nodules are hamartomatous lesions growing indolently along the walls of the lateral ventricles. These are WHO grade 1 tumors of mixed glioneuronal lineage and may cause symptomatic obstructive hydrocephalus secondary to their oft occurrence at the Foramen of Monroe. Subependymal giant cell astrocytoma (SEGA) is the most common brain tumor in patients with TSC, observed in 5–15% of confirmed cases and usually occurs in the first 2 decades of life. Size >10 mm, location, growth on serial neuroimaging studies, and development of hydrocephalus are helpful to distinguish SEGA from a subependymal nodule [66].
The genes responsible are TSC1, also known as Hamartin, located on chromosome 9q34 and TSC2 or Tuberin on chromosome 16p13 [67] and de novo mutations account for approximately 80% of cases [68]. The AKT/mTOR pathway is a key driver of tumorigenesis in TS patients and an important therapeutic target [69, 70].
Treatment
In TS patients, surveillance neuroimaging should be obtained annually during childhood and adolescence, when the risk for SEGA development is greatest.[71] The two main approaches to treatment include surgical resection and targeted medical therapy with mTOR inhibitors such as everolimus. Indications for surgical resection of SEGAs include obstructive hydrocephalus, increased intracranial pressure, tumor progression, and the presence of focal neurologic deficits. Although potentially curative, gross total resection (GTR) is seldom feasible given their intra-ventricular location [72, 73]. TS represents a prototype disease in which biological discoveries have led to the successful development of effective targeted therapies, with profound consequences on clinical management. First-generation mTOR inhibitors (termed rapamycin analogs or rapalogs, including rapamycin) are mTOR complex 1 (mTORC1) specific inhibitors, acting downstream of TSC 1 and 2. Clinical trials using rapalogs have revealed striking tumor regression of virtually all SEGAs in treated TS patients [74–76], and some benefit for neurological symptoms including improved seizure control [77, 78]. Everolimus is FDA approved for pediatric and adult patients with TSC associated SEGAs deemed unresectable. Furthermore, prevention strategies and protocols for long-term therapy with rapalogs are currently being developed for these patients.[79]
B). BRAF fused pLGG
Nearly 30–40% of pLGG harbor focal gains at 7q34 due to a tandem duplication leading to the formation of a novel oncogenic fusion, KIAA1549-BRAF, which represents the most frequent molecular alteration encountered [5, 80, 81]. This rearrangement results in loss of the N-terminal regulatory domain of BRAF and constitutive activation of the RAS/MAPK signaling pathway [5, 81]. KIAA1549-BRAF is enriched in specific histologies (pilocytic astrocytomas which are highly circumscribed) and in tumors arising in the posterior fossa/cerebellum (amenable to gross total resection) resulting in excellent PFS and OS [7, 82–84]. BRAF rearrangements involving non canonical fusion partners including SRGAP [80], FAM131B [85], among others [54, 86, 87] are frequently observed in hemispheric and/or brainstem lesions and tend to arise in older children and adolescents. Given their rarity, impact on patient outcome is difficult to ascertain with some data suggesting lower PFS [7] but requires validation in larger cohorts.
Molecular targeted therapy
Several studies have been completed evaluating MEKi (selumetinib, binimetinib, trametinib) which have demonstrated impressive anti-tumor efficacy across multiple pLGG subtypes [11, 88–92]. Importantly, visual outcomes were reported and improved or stabilized in most patients [89]. Consequently, the COG launched a randomized clinical trial to compare the ORR and functional outcomes for newly diagnosed pLGG treated upfront with either MEKi (Selumetinib) or standard of care (SOC) chemotherapy in non NF1 pLGG (NCT04166409).
Tovorafenib, a second generation BRAFi (blocks BRAF dimers and causes less paradoxical activation) demonstrated impressive ORR in recurrent/refractory BRAF fusion driven pLGG (FIREFLY-1, NCT04775485) and a global phase 3 randomized trial comparing Tovorafenib vs SOC chemotherapy for newly diagnosed BRAF fused pLGG is planned.
Another important question concerns the durability of the observed responses and whether acquired resistance developed off treatment. The PBTC conducted a re-treatment study (NCT01089101) evaluating selumetinib in patients who previously enrolled on PBTC-029 (MEKi naïve patients) and maintained SD for ≥12 courses or had a sustained PR or CR during their first exposure to selumetinib but later progressed after coming off treatment [93]. Re-treatment with selumetinib (n=35) appeared to be effective with 80% of patients again achieving response or prolonged stable disease.
At present, MEKi or BRAFi (second generation) appear similarly efficacious in BRAF fused pLGG. The results of the above referenced randomized phase III studies may potentially alter the standard treatment paradigm for upfront management of non NF1/BRAF fused pLGG.
C). BRAF V600E
Mutations in BRAF resulting from a single amino acid substitution (valine is replaced with a glutamic acid at position 600 (p.V600E) or infrequently, alternate codon 600 substitutions (V600K/R/D/L), located near the activation segment. These alterations act as a phosphomimetic resulting in constitutive activation of MAPK signaling [94, 95]. BRAF p.V600E mutations are histologically and spatially enriched in with pleomorphic xanthoastrocytoma (40–80%) [96, 97], diffuse astrocytoma (30– 40%) and ganglioglioma (25–45%) and supratentorial lesions demonstrating a high frequency of BRAF V600E alterations, respectively [96–98]. BRAF V600E mutant pLGG have worse PFS and OS compared to other pLGG [99, 100]. This is driven in part by the increased propensity for anaplastic/malignant transformation to HGG in specific histological entities (ganglioglioma and PXA) and may occur several years from initial diagnosis, especially when co-occurring with CDKN2A deletions [101, 102]. These anaplastic GGs and “pleomorphic xanthoastrocytoma like” HGG fare better compared to other HGG but are still significantly worse when compared to pLGG [102, 103].
Molecular targeted therapy
Based on impressive results noted in the phase II ROAR trial [104] and NCI-MATCH ‘basket’ trials [105], the Dabrafenib (BRAFi) and Trametinib (MEKi) combination received tumor agnostic approval for adult and pediatric patients with solid tumors harboring a BRAFV600E mutation. For patients with BRAFV600E mutant pLGG, preliminary results of a randomized phase II study (Dabrafenib/Trametinib versus chemotherapy) support the use of combined BRAFi and MEKi molecular targeted therapy for front line treatment in lieu of chemotherapy [106]. The combination is associated with less dermatological toxicity than that seen with MEKi alone.
Molecular targeted therapy versus chemotherapy
As is to be expected, BRAFi and MEKi offer distinct advantages to chemotherapy (oral administration, less myelosuppression) but harbor unique toxicity profiles which include rashes, skin and nail infections and rarely, but significantly, cardiac dysfunction and ocular retinal toxicity which require periodic monitoring, supportive care and drug interruption in severe cases. In addition, the importance of tailoring therapy for pLGG based on a thorough understanding of the distinct signaling mechanisms underlying different BRAF alterations cannot be overstated. First-generation BRAFi (vemurafenib, dabrafenib) which target the monomeric forms of BRAF should not be used for tumors with BRAF fusion which function as dimers given paradoxical ERK activation resulting in tumor progression as demonstrated in a prior study [107, 108].
Receptor tyrosine kinase (RTK) altered pLGG
Besides BRAF, additional fusion genes involving upstream receptor tyrosine kinases (RTKs) have been identified in pLGG including FGFR1/2/3 (fibroblast growth factor receptor), NTRK2 (neurotrophic tropomyosin-related kinase), ROS1 (protein tyrosine kinase encoded by the ROS1 gene), or ALK (anaplastic lymphoma kinase), RAF1, MET or PDGFRA (platelet derived growth factor alpha) [6, 87]. These kinase fusion positive tumors respond to targeted therapy clinically [109]. In pediatric glioma specifically, both entrectinib (ALK/ROS/TRK) and larotrectinib (TRK only) have shown potent anti-tumor effects (NCT02637687, NCT02576431) and the latter was recently approved in the treatment of pediatric and adult patients with TRK-altered cancers (NCT02122913).
FGFR altered pLGG
The molecular landscape of FGFR alterations in pLGG can be divided into 2 groups: 1) Single structural variants (SNVs) or 2) rearrangements that result in the expression of a fusion protein. FGFR1 mutations represent the second most common point mutations in pLGG after BRAF V600 and are most frequently reported in DNETs, RGNTs and a subset of PAs which occur predominantly in extracerebellar, midline locations [6, 87, 110]. These are hotspot alterations affecting p.N546 or p.K656 in the kinase domain and frequently co-occur with a second event in FGFR1 (“dual hit”) and NF1 alterations or additional mutations in components of RAS/MAPK/PI3K pathway [7, 111]. Rearrangement driven pLGG include fusions of FGFR genes with members of the TACC protein family (TACC1, TACC2, and TACC3) or other partners and internal tandem duplications (ITDs). FGFR3:TACC3 fusions are reported in pLGG [6, 112]. FGFR1-TACC1 fusions have been reported in extraventricular neurocytoma [EVN] while several fusions (FGFR2–KIAA1598, FGFR2–CTNNA3 and FGFR3–TACC3) have been observed in polymorphous low-grade neuroepithelial tumor of the young (PLNTY) [113]. Other novel mechanisms resulting in constitutive FGFR1 activation include duplication of the entire kinase domain (TKD) called ITD which is frequently demonstrated in DNET or tumors with oligodendroglial-like histology [6, 87, 114].
A small single center study reported the promising efficacy of FGFR targeted therapy in recurrent/refractory FGFR altered pediatric gliomas [115]. Interestingly, skeletal toxicities not encountered in adults were reported in skeletally immature patients including acceleration of linear growth velocity and slipped capital femoral epiphyses, both of which represent on target effects given the critical role of FGFR3 in bone growth [115].
MYB altered pLGG
MYB alterations are histologically restricted to angiocentric (87%) and diffuse gliomas (41%). Angiocentric gliomas demonstrate characteristic MYB-QKI gene fusion [114, 116] which function via a tripartite mechanism of MYB protein activation, MYB overexpression and the loss-of-function of QKI [116]. Within the same MYB gene family of transcriptional regulators is MYBL1 with similar structure and function [117, 118]. The category of “diffuse astrocytoma, MYB or MYBL1-altered” includes pLGG not bearing the characteristic histologic features of angiocentric glioma but demonstrating recurrent amplifications and structural variants of MYB and MYBL1 [6, 117], including fusions with various gene partners. These tumors arise in young children predominantly in the cerebral hemispheres, although infrequently they occurred in the diencephalon or brainstem [119–121]. Reported 10-year OS and PFS are 90% and 95%, respectively, suggesting that these lesions are indolent [120].
Tectal gliomas
Tectal gliomas arising in the dorsal midbrain typically cause aqueductal obstruction with resultant hydrocephalus [122–124]. These tumors are usually indolent [125, 126] and biopsy is not indicated unless atypical features are present. When biopsied, the majority are WHO grade I PA and frequently harbor KRAS mutations and/or BRAF alterations [127], while histone H3 K27M mutations are absent [124]. Patients can be safely observed post CSF diversion (VP shunt or endoscopic third ventriculostomy) to relieve hydrocephalus and remain progression free without further therapy. Ten-year progression-free and overall survival were 49 and 84 percent, respectively [124, 126, 128].
Cystic pLGG
Some pLGG present with cystic components wherein their biologic behavior may be independent of the solid component of the tumor. These cysts can be symptomatic, necessitating drainage. Approaches to treatment of reaccumulated fluid include repeated drainage via intracavitary ommaya placement, cyst fenestration, bevacizumab or focal RT [129, 130].
Spinal cord pLGG
A recent study reported outcomes in a large cohort (n = 128) of pediatric spinal LGG patients and reported favorable 10-year OS (93 ± 2%) but low 10-year EFS (38 ± 5%), demonstrating a high rate of tumor recurrence and treatment related morbidities resulting in a significant neurological and orthopedic sequela, including kyphoscoliosis, motor disability, pain, and decreased quality of life [131]. An important observation was the excellent disease control rate for patients with localized disease when treated with first-line RT (5-year PFS of 92 ± 8%), whereas patients receiving first-line chemotherapy had 5-year PFS rates of 62 ± 11% which suggest that RT merits serious consideration when adjuvant therapy is required, especially in older, skeletally mature children [131]. Whereas concerns of long-term neurocognitive and endocrine sequelae from RT are of great importance when choosing adjuvant therapy for intracranial LGGs, these are seemingly less relevant to patients with spinal cord tumors where proton irradiation may help limit exposure of unaffected tissues and subsequent morbidity.
CONCLUSIONS
It is important to individualize the timing and selection of tumor-directed interventions for each patient with pLGG based on clinical (age, extent of resection) and molecular characteristics, severity of clinical symptoms, and functional status at presentation. The low mortality but high morbidity rates highlight the need to focus on functional outcomes rather than survival alone. Accordingly, future clinical trials should include systematic evaluation of late toxicities (particularly with respect to molecular targeted therapies where such data is unavailable currently), while incorporating functional outcomes (such as motor abilities), quality of life, and neuro-psychological assessments. Moreover, several important questions remain unresolved, including the role, timing (front line versus relapse) and durability of responses with molecularly targeted agents. It is paramount that future prospective studies will build on the observations made, and ultimately lead to further improvements in both tumor control and functional outcomes for pLGG.
Funding:
This work has been supported in part by the NIH/NCI Cancer Center Support Grant P30 CA008748 to Memorial Sloan Kettering Cancer Center
Funding and/or Conflicts of Interests/Competing Interests
Dr. Karajannis reports grants from Y-mAbs Therapeutics (research support) and personal fees from Bayer, AstraZeneca, QED Therapeutics, CereXis, Recursion, Alexion, Cardinal Health and Medscape (medical advisory board and/or consultant). Dr. Tringale reports grants from Hopper Belmont Foundation and RSNA Resident Grant, as well as honoraria from GT Medical Technologies. Drs. Farouk Sait and Souweidane report no relationships, conditions or circumstances that present a potential conflict of interest.
REFERENCES
Papers of particular interest and published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1. •.Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021;23(8):1231–51. doi: 10.1093/neuonc/noab106. [DOI] [PMC free article] [PubMed] [Google Scholar]; The updated 2021 CNS WHO classification introduced a revised classification system for adult and pediatric-type gliomas.
- 2.Wisoff JH, Sanford RA, Heier LA, Sposto R, Burger PC, Yates AJ, et al. Primary neurosurgery for pediatric low-grade gliomas: a prospective multi-institutional study from the Children’s Oncology Group. Neurosurgery. 2011;68(6):1548–54; discussion 54–5. doi: 10.1227/NEU.0b013e318214a66e. [DOI] [PubMed] [Google Scholar]
- 3.Bandopadhayay P, Bergthold G, London WB, Goumnerova LC, Morales La Madrid A, Marcus KJ, et al. Long-term outcome of 4,040 children diagnosed with pediatric low-grade gliomas: an analysis of the Surveillance Epidemiology and End Results (SEER) database. Pediatr Blood Cancer. 2014;61(7):1173–9. doi: 10.1002/pbc.24958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Packer RJ, Pfister S, Bouffet E, Avery R, Bandopadhayay P, Bornhorst M, et al. Pediatric low-grade gliomas: implications of the biologic era. Neuro Oncol. 2017;19(6):750–61. doi: 10.1093/neuonc/now209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pfister S, Janzarik WG, Remke M, Ernst A, Werft W, Becker N, et al. BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas. J Clin Invest. 2008;118(5):1739–49. doi: 10.1172/JCI33656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhang J, Wu G, Miller CP, Tatevossian RG, Dalton JD, Tang B, et al. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas. Nat Genet. 2013;45(6):602–12. doi: 10.1038/ng.2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. •.Ryall S, Zapotocky M, Fukuoka K, Nobre L, Guerreiro Stucklin A, Bennett J, et al. Integrated Molecular and Clinical Analysis of 1,000 Pediatric Low-Grade Gliomas. Cancer Cell. 2020;37(4):569–83.e5. doi: 10.1016/j.ccell.2020.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]; International collaborative report representing the largest cohort of clinically and molecularly annotated cohort of pLGGs that sheds light on the pLGG molecular landscape and proposes a novel risk stratification system with the potential to improve prognostication and impact treatment.
- 8.Gajjar A, Bowers DC, Karajannis MA, Leary S, Witt H, Gottardo NG. Pediatric Brain Tumors: Innovative Genomic Information Is Transforming the Diagnostic and Clinical Landscape. J Clin Oncol. 2015;33(27):2986–98. doi: 10.1200/JCO.2014.59.9217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hwang EI, Kool M, Burger PC, Capper D, Chavez L, Brabetz S, et al. Extensive Molecular and Clinical Heterogeneity in Patients With Histologically Diagnosed CNS-PNET Treated as a Single Entity: A Report From the Children’s Oncology Group Randomized ACNS0332 Trial. J Clin Oncol. 2018;36(34):JCO2017764720. doi: 10.1200/JCO.2017.76.4720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Korshunov A, Ryzhova M, Hovestadt V, Bender S, Sturm D, Capper D, et al. Integrated analysis of pediatric glioblastoma reveals a subset of biologically favorable tumors with associated molecular prognostic markers. Acta Neuropathol. 2015;129(5):669–78. doi: 10.1007/s00401-015-1405-4. [DOI] [PubMed] [Google Scholar]
- 11. ••.Fangusaro J, Onar-Thomas A, Young Poussaint T, Wu S, Ligon AH, Lindeman N, et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol. 2019;20(7):1011–22. doi: 10.1016/S1470-2045(19)30277-3. [DOI] [PMC free article] [PubMed] [Google Scholar]; First prospective phase II study consortium based study demonstrating promising efficacy of MEK inhibition in recurrent/refractory pediatric low grade gliomas with and without NF1. Theses results led the COG to launch 2 prospective randomized phase III clinical trials for patients with newly diagnosed pediatric low grade gliomas comparing targeted therapy versus standard of care chemotherapy in patients with and without NF1.
- 12.Jacob K, Quang-Khuong D-A, Jones DTW, Witt H, Lambert S, Albrecht S, et al. Genetic Aberrations Leading to MAPK Pathway Activation Mediate Oncogene-Induced Senescence in Sporadic Pilocytic Astrocytomas. Clinical Cancer Research. 2011;17(14):4650–60. doi: 10.1158/1078-0432.Ccr-11-0127. [DOI] [PubMed] [Google Scholar]
- 13.Bale TA, Rosenblum MK. The 2021 WHO Classification of Tumors of the Central Nervous System: An update on pediatric low-grade gliomas and glioneuronal tumors. Brain Pathology. 2022;32(4):e13060. doi: 10.1111/bpa.13060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Packer RJ, Lange B, Ater J, Nicholson HS, Allen J, Walker R, et al. Carboplatin and vincristine for recurrent and newly diagnosed low-grade gliomas of childhood. J Clin Oncol. 1993;11(5):850–6. doi: 10.1200/JCO.1993.11.5.850. [DOI] [PubMed] [Google Scholar]
- 15.Jones DTW, Kieran MW, Bouffet E, Alexandrescu S, Bandopadhayay P, Bornhorst M, et al. Pediatric low-grade gliomas: next biologically driven steps. Neuro Oncol. 2018;20(2):160–73. doi: 10.1093/neuonc/nox141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Giantini-Larsen AM, Pannullo S, Juthani RG. Challenges in the diagnosis and management of low-grade gliomas. World Neurosurgery. 2022. [DOI] [PubMed] [Google Scholar]
- 17.Youland RS, Khwaja SS, Schomas DA, Keating GF, Wetjen NM, Laack NN. Prognostic factors and survival patterns in pediatric low-grade gliomas over 4 decades. J Pediatr Hematol Oncol. 2013;35(3):197–205. doi: 10.1097/MPH.0b013e3182678bf8. [DOI] [PubMed] [Google Scholar]
- 18.Sievert AJ, Fisher MJ. Pediatric low-grade gliomas. J Child Neurol. 2009;24(11):1397–408. doi: 10.1177/0883073809342005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gajjar A, Sanford RA, Heideman R, Jenkins JJ, Walter A, Li Y, et al. Low-grade astrocytoma: a decade of experience at St. Jude Children’s Research Hospital. J Clin Oncol. 1997;15(8):2792–9. doi: 10.1200/JCO.1997.15.8.2792. [DOI] [PubMed] [Google Scholar]
- 20.Fisher PG, Tihan T, Goldthwaite PT, Wharam MD, Carson BS, Weingart JD, et al. Outcome analysis of childhood low-grade astrocytomas. Pediatr Blood Cancer. 2008;51(2):245–50. doi: 10.1002/pbc.21563. [DOI] [PubMed] [Google Scholar]
- 21.Broniscer A, Baker SJ, West AN, Fraser MM, Proko E, Kocak M, et al. Clinical and Molecular Characteristics of Malignant Transformation of Low-Grade Glioma in Children. Journal of Clinical Oncology. 2007;25(6):682–9. doi: 10.1200/jco.2006.06.8213. [DOI] [PubMed] [Google Scholar]
- 22.Tovar-Spinoza Z, Choi H. MRI-guided laser interstitial thermal therapy for the treatment of low-grade gliomas in children: a case-series review, description of the current technologies and perspectives. Child’s Nervous System. 2016;32(10):1947–56. doi: 10.1007/s00381-016-3193-0. [DOI] [PubMed] [Google Scholar]
- 23.Easwaran TP, Lion A, Vortmeyer AO, Kingery K, Bc M, Raskin JS. Seizure freedom from recurrent insular low-grade glioma following laser interstitial thermal therapy. Child’s Nervous System. 2020;36(5):1055–9. doi: 10.1007/s00381-019-04493-6. [DOI] [PubMed] [Google Scholar]
- 24.Karajannis MA, Souweidane MM, Dunkel IJ. Letter to the Editor regarding clinical debate concerning treatment of pediatric LGG by Cooney et al et al. Neurooncol Pract. 2020;7(5):569–70. doi: 10.1093/nop/npaa019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Revere KE, Katowitz WR, Katowitz JA, Rorke-Adams L, Fisher MJ, Liu GT. Childhood Optic Nerve Glioma: Vision Loss Due to Biopsy. Ophthalmic Plast Reconstr Surg. 2017;33(3S Suppl 1):S107–S9. doi: 10.1097/IOP.0000000000000687. [DOI] [PubMed] [Google Scholar]
- 26.Taveras JM, Mount LA, Wood EH. The value of radiation therapy in the management of glioma of the optic nerves and chiasm. Radiology. 1956;66(4):518–28. doi: 10.1148/66.4.518. [DOI] [PubMed] [Google Scholar]
- 27.Erkal HS, Serin M, Cakmak A. Management of optic pathway and chiasmatic-hypothalamic gliomas in children with radiation therapy. Radiother Oncol. 1997;45(1):11–5. doi: 10.1016/s0167-8140(97)00102-3. [DOI] [PubMed] [Google Scholar]
- 28.Cappelli C, Grill J, Raquin M, Pierre-Kahn A, Lellouch-Tubiana A, Terrier-Lacombe MJ, et al. Long-term follow up of 69 patients treated for optic pathway tumours before the chemotherapy era. Arch Dis Child. 1998;79(4):334–8. doi: 10.1136/adc.79.4.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jahraus CD, Tarbell NJ. Optic pathway gliomas. Pediatr Blood Cancer. 2006;46(5):586–96. doi: 10.1002/pbc.20655. [DOI] [PubMed] [Google Scholar]
- 30.Armstrong GT, Conklin HM, Huang S, Srivastava D, Sanford R, Ellison DW, et al. Survival and long-term health and cognitive outcomes after low-grade glioma. Neuro Oncol. 2011;13(2):223–34. doi: 10.1093/neuonc/noq178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Oh KS, Hung J, Robertson PL, Garton HJ, Muraszko KM, Sandler HM, et al. Outcomes of multidisciplinary management in pediatric low-grade gliomas. Int J Radiat Oncol Biol Phys. 2011;81(4):e481–8. doi: 10.1016/j.ijrobp.2011.01.019. [DOI] [PubMed] [Google Scholar]
- 32.Mishra KK, Puri DR, Missett BT, Lamborn KR, Prados MD, Berger MS, et al. The role of up-front radiation therapy for incompletely resected pediatric WHO grade II low-grade gliomas. Neuro Oncol. 2006;8(2):166–74. doi: 10.1215/15228517-2005-011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Watson GA, Kadota RP, Wisoff JH. Multidisciplinary management of pediatric low-grade gliomas. Semin Radiat Oncol. 2001;11(2):152–62. doi: 10.1053/srao.2001.21421. [DOI] [PubMed] [Google Scholar]
- 34.Pollack IF, Claassen D, al-Shboul Q, Janosky JE, Deutsch M. Low-grade gliomas of the cerebral hemispheres in children: an analysis of 71 cases. J Neurosurg. 1995;82(4):536–47. doi: 10.3171/jns.1995.82.4.0536. [DOI] [PubMed] [Google Scholar]
- 35.Raikar SS, Halloran DR, Elliot M, McHugh M, Patel S, Gauvain KM. Outcomes of pediatric low-grade gliomas treated with radiation therapy: a single-institution study. J Pediatr Hematol Oncol. 2014;36(6):e366–70. doi: 10.1097/MPH.0000000000000142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tsang DS, Murphy ES, Merchant TE. Radiation Therapy for Optic Pathway and Hypothalamic Low-Grade Gliomas in Children. Int J Radiat Oncol Biol Phys. 2017;99(3):642–51. doi: 10.1016/j.ijrobp.2017.07.023. [DOI] [PubMed] [Google Scholar]
- 37.Packer RJ, Sutton LN, Atkins TE, Radcliffe J, Bunin GR, D’Angio G, et al. A prospective study of cognitive function in children receiving whole-brain radiotherapy and chemotherapy: 2-year results. J Neurosurg. 1989;70(5):707–13. doi: 10.3171/jns.1989.70.5.0707. [DOI] [PubMed] [Google Scholar]
- 38.Merchant TE, Conklin HM, Wu S, Lustig RH, Xiong X. Late effects of conformal radiation therapy for pediatric patients with low-grade glioma: prospective evaluation of cognitive, endocrine, and hearing deficits. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(22):3691–7. doi: 10.1200/JCO.2008.21.2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brauner R, Malandry F, Rappaport R, Zucker JM, Kalifa C, Pierre-Kahn A, et al. Growth and endocrine disorders in optic glioma. Eur J Pediatr. 1990;149(12):825–8. doi: 10.1007/BF02072067. [DOI] [PubMed] [Google Scholar]
- 40.Bowers DC, Mulne AF, Reisch JS, Elterman RD, Munoz L, Booth T, et al. Nonperioperative strokes in children with central nervous system tumors. Cancer. 2002;94(4):1094–101. [PubMed] [Google Scholar]
- 41.Armstrong GT, Liu Q, Yasui Y, Huang S, Ness KK, Leisenring W, et al. Long-term outcomes among adult survivors of childhood central nervous system malignancies in the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2009;101(13):946–58. doi: 10.1093/jnci/djp148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Harrabi SB, Bougatf N, Mohr A, Haberer T, Herfarth K, Combs SE, et al. Dosimetric advantages of proton therapy over conventional radiotherapy with photons in young patients and adults with low-grade glioma. Strahlenther Onkol. 2016;192(11):759–69. doi: 10.1007/s00066-016-1005-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Takizawa D, Mizumoto M, Yamamoto T, Oshiro Y, Fukushima H, Fukushima T, et al. A comparative study of dose distribution of PBT, 3D-CRT and IMRT for pediatric brain tumors. Radiat Oncol. 2017;12(1):40. doi: 10.1186/s13014-017-0775-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yock TI, Bhat S, Szymonifka J, Yeap BY, Delahaye J, Donaldson SS, et al. Quality of life outcomes in proton and photon treated pediatric brain tumor survivors. Radiother Oncol. 2014;113(1):89–94. doi: 10.1016/j.radonc.2014.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Verma V, Mishra MV, Mehta MP. A systematic review of the cost and cost-effectiveness studies of proton radiotherapy. Cancer. 2016;122(10):1483–501. doi: 10.1002/cncr.29882. [DOI] [PubMed] [Google Scholar]
- 46.Greenberger BA, Pulsifer MB, Ebb DH, MacDonald SM, Jones RM, Butler WE, et al. Clinical outcomes and late endocrine, neurocognitive, and visual profiles of proton radiation for pediatric low-grade gliomas. Int J Radiat Oncol Biol Phys. 2014;89(5):1060–8. doi: 10.1016/j.ijrobp.2014.04.053. [DOI] [PubMed] [Google Scholar]
- 47.Indelicato DJ, Bates JE, Mailhot Vega RB, Rotondo RL, Hoppe BS, Morris CG, et al. Second tumor risk in children treated with proton therapy. Pediatr Blood Cancer. 2021;68(7):e28941. doi: 10.1002/pbc.28941. [DOI] [PubMed] [Google Scholar]
- 48.Indelicato D, Tringale K, Bradley J, Vega RM, Morris C, Casey D, et al. RONC-03. Secondary Neoplasms in Children with Central Nervous System (CNS) Tumors Following Radiotherapy in the Modern Era. Neuro-Oncology. 2022;24(Suppl 1):i176. [Google Scholar]
- 49.Indelicato DJ, Rotondo RL, Uezono H, Sandler ES, Aldana PR, Ranalli NJ, et al. Outcomes Following Proton Therapy for Pediatric Low-Grade Glioma. Int J Radiat Oncol Biol Phys. 2019;104(1):149–56. doi: 10.1016/j.ijrobp.2019.01.078. [DOI] [PubMed] [Google Scholar]
- 50.Bitterman DS, MacDonald SM, Yock TI, Tarbell NJ, Wright KD, Chi SN, et al. Revisiting the Role of Radiation Therapy for Pediatric Low-Grade Glioma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2019;37(35):3335–9. doi: 10.1200/JCO.19.01270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Marcus KJ, Goumnerova L, Billett AL, Lavally B, Scott RM, Bishop K, et al. Stereotactic radiotherapy for localized low-grade gliomas in children: final results of a prospective trial. Int J Radiat Oncol Biol Phys. 2005;61(2):374–9. doi: 10.1016/j.ijrobp.2004.06.012. [DOI] [PubMed] [Google Scholar]
- 52.Cherlow JM, Shaw DWW, Margraf LR, Bowers DC, Huang J, Fouladi M, et al. Conformal Radiation Therapy for Pediatric Patients with Low-Grade Glioma: Results from the Children’s Oncology Group Phase 2 Study ACNS0221. Int J Radiat Oncol Biol Phys. 2019;103(4):861–8. doi: 10.1016/j.ijrobp.2018.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ater JL, Zhou T, Holmes E, Mazewski CM, Booth TN, Freyer DR, et al. Randomized study of two chemotherapy regimens for treatment of low-grade glioma in young children: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30(21):2641–7. doi: 10.1200/JCO.2011.36.6054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun. 2020;8(1):30. doi: 10.1186/s40478-020-00902-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hargrave DR, Bouffet E, Tabori U, Broniscer A, Cohen KJ, Hansford JR, et al. Efficacy and Safety of Dabrafenib in Pediatric 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. doi: 10.1158/1078-0432.Ccr-19-2177. [DOI] [PubMed] [Google Scholar]
- 56.Gutmann DH, Donahoe J, Brown T, James CD, Perry A. Loss of neurofibromatosis 1 (NF1) gene expression in NF1-associated pilocytic astrocytomas. Neuropathology and applied neurobiology. 2000;26(4):361–7. [DOI] [PubMed] [Google Scholar]
- 57.D’Angelo F, Ceccarelli M, Tala, Garofano L, Zhang J, Frattini V, et al. The molecular landscape of glioma in patients with Neurofibromatosis 1. Nature medicine. 2019;25(1):176–87. doi: 10.1038/s41591-018-0263-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Evans DGR, Salvador H, Chang VY, Erez A, Voss SD, Schneider KW, et al. Cancer and Central Nervous System Tumor Surveillance in Pediatric Neurofibromatosis 1. Clin Cancer Res. 2017;23(12):e46–e53. doi: 10.1158/1078-0432.Ccr-17-0589. [DOI] [PubMed] [Google Scholar]
- 59.Bhatia S, Chen Y, Wong FL, Hageman L, Smith K, Korf B, et al. Subsequent Neoplasms After a Primary Tumor in Individuals With Neurofibromatosis Type 1. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2019;37(32):3050–8. doi: 10.1200/jco.19.00114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ullrich NJ, Robertson R, Kinnamon DD, Scott RM, Kieran MW, Turner CD, et al. Moyamoya following cranial irradiation for primary brain tumors in children. Neurology. 2007;68(12):932–8. doi: 10.1212/01.wnl.0000257095.33125.48. [DOI] [PubMed] [Google Scholar]
- 61.Sampson JR, Scahill SJ, Stephenson JB, Mann L, Connor JM. Genetic aspects of tuberous sclerosis in the west of Scotland. Journal of medical genetics. 1989;26(1):28–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Osborne JP, Fryer A, Webb D. Epidemiology of tuberous sclerosis. Annals of the New York Academy of Sciences. 1991;615:125–7. [DOI] [PubMed] [Google Scholar]
- 63.Northrup H, Krueger DA. Tuberous sclerosis complex diagnostic criteria update: recommendations of the 2012 Iinternational Tuberous Sclerosis Complex Consensus Conference. Pediatric neurology. 2013;49(4):243–54. doi: 10.1016/j.pediatrneurol.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Northrup H, Koenig MK, Pearson DA, Au KS. Tuberous Sclerosis Complex. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, et al. , editors. GeneReviews(R). Seattle (WA): University of Washington, Seattle; [PubMed] [Google Scholar]
- 65.Crino PB. Molecular pathogenesis of tuber formation in tuberous sclerosis complex. J Child Neurol. 2004;19(9):716–25. doi: 10.1177/08830738040190091301. [DOI] [PubMed] [Google Scholar]
- 66.Grajkowska W, Kotulska K, Jurkiewicz E, Matyja E. Brain lesions in tuberous sclerosis complex. Review. Folia neuropathologica. 2010;48(3):139–49. [PubMed] [Google Scholar]
- 67.van Slegtenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, Verhoef S, et al. Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science (New York, NY). 1997;277(5327):805–8. [DOI] [PubMed] [Google Scholar]
- 68.Au KS, Williams AT, Roach ES, Batchelor L, Sparagana SP, Delgado MR, et al. Genotype/phenotype correlation in 325 individuals referred for a diagnosis of tuberous sclerosis complex in the United States. Genetics in medicine: official journal of the American College of Medical Genetics. 2007;9(2):88–100. doi: 10.1097/GIM.0b013e31803068c7. [DOI] [PubMed] [Google Scholar]
- 69.Sabatini DM. mTOR and cancer: insights into a complex relationship. Nature reviews Cancer. 2006;6(9):729–34. doi: 10.1038/nrc1974. [DOI] [PubMed] [Google Scholar]
- 70.Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC, Blenis J. Tuberous sclerosis complex-1 and −2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc Natl Acad Sci U S A. 2002;99(21):13571–6. doi: 10.1073/pnas.202476899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Roach ES, Gomez MR, Northrup H. Tuberous sclerosis complex consensus conference: revised clinical diagnostic criteria. J Child Neurol. 1998;13(12):624–8. doi: 10.1177/088307389801301206. [DOI] [PubMed] [Google Scholar]
- 72.Krueger DA. Management of CNS-related Disease Manifestations in Patients With Tuberous Sclerosis Complex. Current treatment options in neurology. 2013;15(5):618–33. doi: 10.1007/s11940-013-0249-2. [DOI] [PubMed] [Google Scholar]
- 73.Goh S, Butler W, Thiele EA. Subependymal giant cell tumors in tuberous sclerosis complex. Neurology. 2004;63(8):1457–61. [DOI] [PubMed] [Google Scholar]
- 74.Franz DN, Leonard J, Tudor C, Chuck G, Care M, Sethuraman G, et al. Rapamycin causes regression of astrocytomas in tuberous sclerosis complex. Ann Neurol. 2006;59(3):490–8. doi: 10.1002/ana.20784. [DOI] [PubMed] [Google Scholar]
- 75.Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. 2010;363(19):1801–11. doi: 10.1056/NEJMoa1001671. [DOI] [PubMed] [Google Scholar]
- 76.Franz DN, Belousova E, Sparagana S, Bebin EM, Frost M, Kuperman R, et al. Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2013;381(9861):125–32. doi: 10.1016/S0140-6736(12)61134-9. [DOI] [PubMed] [Google Scholar]
- 77.Ehninger D, Han S, Shilyansky C, Zhou Y, Li W, Kwiatkowski DJ, et al. Reversal of learning deficits in a Tsc2+/− mouse model of tuberous sclerosis. Nature medicine. 2008;14(8):843–8. doi: 10.1038/nm1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wiegand G, May TW, Ostertag P, Boor R, Stephani U, Franz DN. Everolimus in tuberous sclerosis patients with intractable epilepsy: a treatment option? Eur J Paediatr Neurol. 2013;17(6):631–8. doi: 10.1016/j.ejpn.2013.06.002. [DOI] [PubMed] [Google Scholar]
- 79.Krueger DA, Care MM, Agricola K, Tudor C, Mays M, Franz DN. Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma. Neurology. 2013;80(6):574–80. doi: 10.1212/WNL.0b013e3182815428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jones DT, Kocialkowski S, Liu L, Pearson DM, Backlund LM, Ichimura K, et al. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008;68(21):8673–7. doi: 10.1158/0008-5472.CAN-08-2097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ryall S, Arnoldo A, Krishnatry R, Mistry M, Khor K, Sheth J, et al. Multiplex Detection of Pediatric Low-Grade Glioma Signature Fusion Transcripts and Duplications Using the NanoString nCounter System. J Neuropathol Exp Neurol. 2017;76(7):562–70. doi: 10.1093/jnen/nlx042. [DOI] [PubMed] [Google Scholar]
- 82.Becker AP, Scapulatempo-Neto C, Carloni AC, Paulino A, Sheren J, Aisner DL, et al. KIAA1549: BRAF Gene Fusion and FGFR1 Hotspot Mutations Are Prognostic Factors in Pilocytic Astrocytomas. Journal of Neuropathology & Experimental Neurology. 2015;74(7):743–54. doi: 10.1097/nen.0000000000000213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Horbinski C, Hamilton RL, Nikiforov Y, Pollack IF. Association of molecular alterations, including BRAF, with biology and outcome in pilocytic astrocytomas. Acta neuropathologica. 2010;119(5):641–9. doi: 10.1007/s00401-009-0634-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Lassaletta A, Zapotocky M, Mistry M, Ramaswamy V, Honnorat M, Krishnatry R, et al. Therapeutic and Prognostic Implications of BRAF V600E in Pediatric Low-Grade Gliomas. Journal of Clinical Oncology. 2017;35(25):2934–41. doi: 10.1200/jco.2016.71.8726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cin H, Meyer C, Herr R, Janzarik WG, Lambert S, Jones DTW, et al. Oncogenic FAM131B–BRAF fusion resulting from 7q34 deletion comprises an alternative mechanism of MAPK pathway activation in pilocytic astrocytoma. Acta neuropathologica. 2011;121(6):763–74. doi: 10.1007/s00401-011-0817-z. [DOI] [PubMed] [Google Scholar]
- 86.Helgager J, Lidov HG, Mahadevan NR, Kieran MW, Ligon KL, Alexandrescu S. A novel GIT2-BRAF fusion in pilocytic astrocytoma. Diagnostic Pathology. 2017;12(1):82. doi: 10.1186/s13000-017-0669-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jones DT, Hutter B, Jager N, Korshunov A, Kool M, Warnatz HJ, et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013;45(8):927–32. doi: 10.1038/ng.2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Fangusaro J, Onar-Thomas A, Poussaint TY, Lensing S, Wu S, Ligon AH, et al. LGG-06. Selumetinib in pediatric patients with non-neurofibromatosis type 1-associated, non-optic pathway (OPG) and non-pilocytic recurrent/progressive low-grade glioma harboring BRAFV600E mutation or BRAF-KIAA1549 fusion: a multicenter prospective Pediatric Brain Tumor Consortium (PBTC) Phase 2 trial. Neuro-Oncology. 2022;24(Supplement_1):i88–i. doi: 10.1093/neuonc/noac079.322. [DOI] [Google Scholar]
- 89.Fangusaro J, Onar-Thomas A, Poussaint TY, Wu S, Ligon AH, Lindeman N, et al. LGG-02. A PHASE II PROSPECTIVE TRIAL OF SELUMETINIB IN CHILDREN WITH RECURRENT/PROGRESSIVE PEDIATRIC LOW-GRADE GLIOMA (PLGG) WITH A FOCUS UPON OPTIC PATHWAY/HYPOTHALAMIC TUMORS AND VISUAL ACUITY OUTCOMES: A PEDIATRIC BRAIN TUMOR CONSORTIUM (PBTC) STUDY, PBTC-029B. Neuro-Oncology. 2019;21(Supplement_2):ii98–ii9. doi: 10.1093/neuonc/noz036.145. [DOI] [Google Scholar]
- 90.Robison N, Pauly J, Malvar J, Gardner S, Allen J, Margol A, et al. LTBK-04. LATE BREAKING ABSTRACT: MEK162 (binimetinib) in children with progressive or recurrent low-grade glioma: a multi-institutional phase II and target validation study. Neuro-Oncology. 2022;24(Supplement_1):i191–i2. doi: 10.1093/neuonc/noac079.716. [DOI] [Google Scholar]
- 91.Perreault S, Sadat Kiaei D, Dehaes M, Larouche V, Tabori U, Hawkin C, et al. A phase 2 study of trametinib for patients with pediatric glioma or plexiform neurofibroma with refractory tumor and activation of the MAPK/ERK pathway. Journal of Clinical Oncology. 2022;40(16_suppl):2042-. doi: 10.1200/JCO.2022.40.16_suppl.2042. [DOI] [Google Scholar]
- 92.Trippett T, Toledano H, Campbell Hewson Q, Verschuur A, Langevin AM, Aerts I, et al. Cobimetinib in Pediatric and Young Adult Patients with Relapsed or Refractory Solid Tumors (iMATRIX-cobi): A Multicenter, Phase I/II Study. Target Oncol. 2022;17(3):283–93. doi: 10.1007/s11523-022-00888-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fangusaro J, Onar-Thomas A, Wu S, Poussaint TY, Packer R, Kilburn L, et al. LGG-04. A PHASE II RE-TREATMENT STUDY OF SELUMETINIB FOR RECURRENT OR PROGRESSIVE PEDIATRIC LOW-GRADE GLIOMA (pLGG): A PEDIATRIC BRAIN TUMOR CONSORTIUM (PBTC) STUDY. Neuro-Oncology. 2020;22(Supplement_3):iii367–iii. doi: 10.1093/neuonc/noaa222.389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Cantwell-Dorris ER, O’Leary JJ, Sheils OM. BRAFV600E: Implications for Carcinogenesis and Molecular Therapy. Molecular Cancer Therapeutics. 2011;10(3):385–94. doi: 10.1158/1535-7163.Mct-10-0799. [DOI] [PubMed] [Google Scholar]
- 95.Yao Z, Torres NM, Tao A, Gao Y, Luo L, Li Q, et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell. 2015;28(3):370–83. doi: 10.1016/j.ccell.2015.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gierke M, Sperveslage J, Schwab D, Beschorner R, Ebinger M, Schuhmann MU, et al. Analysis of IDH1-R132 mutation, BRAF V600 mutation and KIAA1549–BRAF fusion transcript status in central nervous system tumors supports pediatric tumor classification. Journal of Cancer Research and Clinical Oncology. 2016;142(1):89–100. doi: 10.1007/s00432-015-2006-2. [DOI] [PubMed] [Google Scholar]
- 97.Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold-Mende C, et al. Analysis of BRAF V600E mutation in 1,320 nervous system tumors reveals high mutation frequencies in pleomorphic xanthoastrocytoma, ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta neuropathologica. 2011;121(3):397–405. doi: 10.1007/s00401-011-0802-6. [DOI] [PubMed] [Google Scholar]
- 98.Schiffman JD, Hodgson JG, VandenBerg SR, Flaherty P, Polley M-YC, Yu M, et al. Oncogenic BRAF Mutation with CDKN2A Inactivation Is Characteristic of a Subset of Pediatric Malignant Astrocytomas. Cancer Research. 2010;70(2):512–9. doi: 10.1158/0008-5472.CAN-09-1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lassaletta A, Zapotocky M, Mistry M, Ramaswamy V, Honnorat M, Krishnatry R, et al. Therapeutic and Prognostic Implications of BRAF V600E in Pediatric Low-Grade Gliomas. Journal of Clinical Oncology. 2017;35(25):2934–41. doi: 10.1200/JCO.2016.71.8726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dahiya S, Haydon DH, Alvarado D, Gurnett CA, Gutmann DH, Leonard JR. BRAFV600E mutation is a negative prognosticator in pediatric ganglioglioma. Acta neuropathologica. 2013;125(6):901–10. doi: 10.1007/s00401-013-1120-y. [DOI] [PubMed] [Google Scholar]
- 101.Mistry M, Zhukova N, Merico D, Rakopoulos P, Krishnatry R, Shago M, et al. BRAF Mutation and CDKN2A Deletion Define a Clinically Distinct Subgroup of Childhood Secondary High-Grade Glioma. Journal of Clinical Oncology. 2015;33(9):1015–22. doi: 10.1200/jco.2014.58.3922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Tanaka S, Nakada M, Nobusawa S, Suzuki SO, Sabit H, Miyashita K, et al. Epithelioid glioblastoma arising from pleomorphic xanthoastrocytoma with the BRAF V600E mutation. Brain Tumor Pathology. 2014;31(3):172–6. doi: 10.1007/s10014-014-0192-2. [DOI] [PubMed] [Google Scholar]
- 103.Korshunov A, Ryzhova M, Hovestadt V, Bender S, Sturm D, Capper D, et al. Integrated analysis of pediatric glioblastoma reveals a subset of biologically favorable tumors with associated molecular prognostic markers. Acta neuropathologica. 2015;129(5):669–78. doi: 10.1007/s00401-015-1405-4. [DOI] [PubMed] [Google Scholar]
- 104.Wen PY, Stein A, van den Bent M, De Greve J, Wick A, de Vos FYFL, et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial. The Lancet Oncology. 2022;23(1):53–64. doi: 10.1016/S1470-2045(21)00578-7. [DOI] [PubMed] [Google Scholar]
- 105. •.Salama AKS, Li S, Macrae ER, Park JI, Mitchell EP, Zwiebel JA, et al. Dabrafenib and Trametinib in Patients With Tumors With BRAF(V600E) Mutations: Results of the NCI-MATCH Trial Subprotocol H. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2020;38(33):3895–904. doi: 10.1200/jco.20.00762. [DOI] [PMC free article] [PubMed] [Google Scholar]; Subprotocol H (EAY131-H) of the NCI-MATCH platform trial was a single-arm phase II histology agnostic trial investigating the combination of BRAF inhibitor dabrafenib and the MEK1/2 inhibitor trametinib in a biomarker-selected cohort of patients with recurrent/refractory solid tumors harboring a BRAFV600 mutation. Dabrafenib and trametinib therapy resulted in responses in 38% of patients and showed a high rate of disease control across a variety of disease histologies eventually culminating in the recent FDA approval of dabrafenib and trametinib for patients with BRAFV600E-mutant solid tumors.
- 106. ••.Bouffet E, Geoerger B, Moertel C, Whitlock JA, Aerts I, Hargrave D, et al. Efficacy and Safety of Trametinib Monotherapy or in Combination With Dabrafenib in Pediatric BRAF V600-Mutant Low-Grade Glioma. J Clin Oncol. 2023;41(3):664–74. doi: 10.1200/JCO.22.01000. [DOI] [PMC free article] [PubMed] [Google Scholar]; This randomized phase 2 trial tested dabrafenib with trametinib versus standard-ofcare chemotherapy (carboplatin/vincristine) and demonstrated improved overall response rate (ORR) and prolonged progression-free survival (PFS) with targeted therapy compared with standard chemotherapy. Dabrafenib with trametinib represents a new standard of care for pediatric patients with newly diagnosed BRAFV600-mutant low-grade glioma.
- 107.Karajannis MA, Legault G, Fisher MJ, Milla SS, Cohen KJ, Wisoff JH, et al. Phase II study of sorafenib in children with recurrent or progressive low-grade astrocytomas. Neuro Oncol. 2014;16(10):1408–16. doi: 10.1093/neuonc/nou059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Sievert AJ, Lang SS, Boucher KL, Madsen PJ, Slaunwhite E, Choudhari N, et al. Paradoxical activation and RAF inhibitor resistance of BRAF protein kinase fusions characterizing pediatric astrocytomas. Proc Natl Acad Sci U S A. 2013;110(15):5957–62. doi: 10.1073/pnas.1219232110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Clarke M, Mackay A, Ismer B, Pickles JC, Tatevossian RG, Newman S, et al. Infant High-Grade Gliomas Comprise Multiple Subgroups Characterized by Novel Targetable Gene Fusions and Favorable Outcomes. Cancer Discov. 2020;10(7):942–63. doi: 10.1158/2159-8290.CD-19-1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Gessi M, Moneim YA, Hammes J, Goschzik T, Scholz M, Denkhaus D, et al. FGFR1 mutations in Rosette-forming glioneuronal tumors of the fourth ventricle. J Neuropathol Exp Neurol. 2014;73(6):580–4. doi: 10.1097/nen.0000000000000080. [DOI] [PubMed] [Google Scholar]
- 111.Sievers P, Appay R, Schrimpf D, Stichel D, Reuss DE, Wefers AK, et al. Rosette-forming glioneuronal tumors share a distinct DNA methylation profile and mutations in FGFR1, with recurrent co-mutation of PIK3CA and NF1. Acta neuropathologica. 2019;138(3):497–504. doi: 10.1007/s00401-019-02038-4. [DOI] [PubMed] [Google Scholar]
- 112.Lasorella A, Sanson M, Iavarone A. FGFR-TACC gene fusions in human glioma. Neuro Oncol. 2017;19(4):475–83. doi: 10.1093/neuonc/now240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Huse JT, Snuderl M, Jones DT, Brathwaite CD, Altman N, Lavi E, et al. Polymorphous low-grade neuroepithelial tumor of the young (PLNTY): an epileptogenic neoplasm with oligodendroglioma-like components, aberrant CD34 expression, and genetic alterations involving the MAP kinase pathway. Acta neuropathologica. 2017;133(3):417–29. doi: 10.1007/s00401-016-1639-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Qaddoumi I, Orisme W, Wen J, Santiago T, Gupta K, Dalton JD, et al. Genetic alterations in uncommon low-grade neuroepithelial tumors: BRAF, FGFR1, and MYB mutations occur at high frequency and align with morphology. Acta neuropathologica. 2016;131(6):833–45. doi: 10.1007/s00401-016-1539-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Farouk Sait S, Gilheeney SW, Bale TA, Haque S, Dinkin MJ, Vitolano S, et al. Debio1347, an Oral FGFR Inhibitor: Results From a Single-Center Study in Pediatric Patients With Recurrent or Refractory FGFR-Altered Gliomas. JCO Precis Oncol. 2021;5. doi: 10.1200/po.20.00444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Bandopadhayay P, Ramkissoon LA, Jain P, Bergthold G, Wala J, Zeid R, et al. MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism. Nat Genet. 2016;48(3):273–82. doi: 10.1038/ng.3500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ramkissoon LA, Horowitz PM, Craig JM, Ramkissoon SH, Rich BE, Schumacher SE, et al. Genomic analysis of diffuse pediatric low-grade gliomas identifies recurrent oncogenic truncating rearrangements in the transcription factor MYBL1. Proc Natl Acad Sci U S A. 2013;110(20):8188–93. doi: 10.1073/pnas.1300252110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Cicirò Y, Sala A. MYB oncoproteins: emerging players and potential therapeutic targets in human cancer. Oncogenesis. 2021;10(2):19. doi: 10.1038/s41389-021-00309-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chan E, Bollen AW, Sirohi D, Van Ziffle J, Grenert JP, Kline CN, et al. Angiocentric glioma with MYB-QKI fusion located in the brainstem, rather than cerebral cortex. Acta neuropathologica. 2017;134(4):671–3. doi: 10.1007/s00401-017-1759-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Chiang J, Harreld JH, Tinkle CL, Moreira DC, Li X, Acharya S, et al. A single-center study of the clinicopathologic correlates of gliomas with a MYB or MYBL1 alteration. Acta neuropathologica. 2019;138(6):1091–2. doi: 10.1007/s00401-019-02081-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.D’Aronco L, Rouleau C, Gayden T, Crevier L, Décarie J-C, Perreault S, et al. Brainstem angiocentric gliomas with MYB–QKI rearrangements. Acta neuropathologica. 2017;134(4):667–9. doi: 10.1007/s00401-017-1763-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Li KW, Roonprapunt C, Lawson HC, Abbott IR, Wisoff J, Epstein F, et al. Endoscopic third ventriculostomy for hydrocephalus associated with tectal gliomas. Neurosurg Focus. 2005;18(6a):E2. [PubMed] [Google Scholar]
- 123.Epstein F, Constantini S. Practical decisions in the treatment of pediatric brain stem tumors. Pediatr Neurosurg. 1996;24(1):24–34. doi: 10.1159/000121011. [DOI] [PubMed] [Google Scholar]
- 124.Liu APY, Harreld JH, Jacola LM, Gero M, Acharya S, Ghazwani Y, et al. Tectal glioma as a distinct diagnostic entity: a comprehensive clinical, imaging, histologic and molecular analysis. Acta Neuropathol Commun. 2018;6(1):101. doi: 10.1186/s40478-018-0602-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Lapras C, Bognar L, Turjman F, Villanyi E, Mottolese C, Fischer C, et al. Tectal plate gliomas. Part I: Microsurgery of the tectal plate gliomas. Acta Neurochir (Wien). 1994;126(2–4):76–83. doi: 10.1007/bf01476414. [DOI] [PubMed] [Google Scholar]
- 126.May PL, Blaser SI, Hoffman HJ, Humphreys RP, Harwood-Nash DC. Benign intrinsic tectal “tumors” in children. J Neurosurg. 1991;74(6):867–71. doi: 10.3171/jns.1991.74.6.0867. [DOI] [PubMed] [Google Scholar]
- 127.Chiang J, Li X, Liu APY, Qaddoumi I, Acharya S, Ellison DW. Tectal glioma harbors high rates of KRAS G12R and concomitant KRAS and BRAF alterations. Acta Neuropathol. 2020;139(3):601–2. doi: 10.1007/s00401-019-02112-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Dağlioğlu E, Cataltepe O, Akalan N. Tectal gliomas in children: the implications for natural history and management strategy. Pediatr Neurosurg. 2003;38(5):223–31. doi: 10.1159/000069823. [DOI] [PubMed] [Google Scholar]
- 129.Disabato JA, Handler MH, Strain JD, Fleitz JM, Foreman NK. Successful use of intracavitary bleomycin for low-grade astrocytoma tumor cyst. Pediatr Neurosurg. 1999;31(5):246–50. doi: 10.1159/000028871. [DOI] [PubMed] [Google Scholar]
- 130.Giovanini MA, Mickle JP. Long-term access to cystic brain stem lesions using the Ommaya reservoir: technical case report. Neurosurgery. 1996;39(2):404–7; discussion 7–8. doi: 10.1097/00006123-199608000-00039. [DOI] [PubMed] [Google Scholar]
- 131.Perwein T, Benesch M, Kandels D, Pietsch T, Schmidt R, Quehenberger F, et al. High frequency of disease progression in pediatric spinal cord low-grade glioma (LGG): management strategies and results from the German LGG study group. Neuro Oncol. 2021;23(7):1148–62. doi: 10.1093/neuonc/noaa296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Packer RJ, Ater J, Allen J, Phillips P, Geyer R, Nicholson HS, et al. Carboplatin and vincristine chemotherapy for children with newly diagnosed progressive low-grade gliomas. J Neurosurg. 1997;86(5):747–54. doi: 10.3171/jns.1997.86.5.0747. [DOI] [PubMed] [Google Scholar]
- 133.Mahoney DH Jr, Cohen ME, Friedman HS, Kepner JL, Gemer L, Langston JW, et al. Carboplatin is effective therapy for young children with progressive optic pathway tumors: a Pediatric Oncology Group phase II study. Neuro Oncol. 2000;2(4):213–20. doi: 10.1093/neuonc/2.4.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Laithier V, Grill J, Le Deley MC, Ruchoux MM, Couanet D, Doz F, et al. Progression-free survival in children with optic pathway tumors: dependence on age and the quality of the response to chemotherapy--results of the first French prospective study for the French Society of Pediatric Oncology. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2003;21(24):4572–8. doi: 10.1200/jco.2003.03.043. [DOI] [PubMed] [Google Scholar]
- 135.Gnekow AK, Falkenstein F, von Hornstein S, Zwiener I, Berkefeld S, Bison B, et al. Long-term follow-up of the multicenter, multidisciplinary treatment study HIT-LGG-1996 for low-grade glioma in children and adolescents of the German Speaking Society of Pediatric Oncology and Hematology. Neuro Oncol. 2012;14(10):1265–84. doi: 10.1093/neuonc/nos202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chintagumpala M, Eckel SP, Krailo M, Morris M, Adesina A, Packer R, et al. A pilot study using carboplatin, vincristine, and temozolomide in children with progressive/symptomatic low-grade glioma: a Children’s Oncology Group study†. Neuro Oncol. 2015;17(8):1132–8. doi: 10.1093/neuonc/nov057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Gnekow AK, Walker DA, Kandels D, Picton S, Giorgio P, Grill J, et al. A European randomised controlled trial of the addition of etoposide to standard vincristine and carboplatin induction as part of an 18-month treatment programme for childhood (≤16 years) low grade glioma - A final report. Eur J Cancer. 2017;81:206–25. doi: 10.1016/j.ejca.2017.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Lassaletta A, Scheinemann K, Zelcer SM, Hukin J, Wilson BA, Jabado N, et al. Phase II Weekly Vinblastine for Chemotherapy-Naïve Children With Progressive Low-Grade Glioma: A Canadian Pediatric Brain Tumor Consortium Study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(29):3537–43. doi: 10.1200/jco.2016.68.1585. [DOI] [PubMed] [Google Scholar]