Brain Tumor Res Treat. 2022 Oct;10(4):221-225. English.
Published online Oct 27, 2022.
Copyright © 2022 The Korean Brain Tumor Society, The Korean Society for Neuro-Oncology, and The Korean Society for Pediatric Neuro-Oncology
Review

Medical Treatment of Pediatric Low-Grade Glioma

Yeon Jung Lim
    • Department of Pediatrics, Chungnam National University College of Medicine, Deajeon, Korea.
Received September 19, 2022; Accepted October 12, 2022.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Low-grade glioma (LGG) is the most common brain tumor in children and has excellent long-term survival. With an excellent survival rate, the choice of treatment involves careful consideration of minimizing late toxicity from surgery, radiation, and chemotherapy. Surgery, radiation therapy, and chemotherapy can be used as monotherapy or in combination, providing different therapeutic ratios and complications. As a result, establishing the selection of ideal therapies has been a controversial area, presenting challenges. Recent advances in understanding molecular characteristics of pediatric LGG affect classification and treatment approaches. This review aims to overview recent developments in medical treatment in pediatric LGG.

Keywords
Low grade glioma; Pediatric brain tumor; Chemotherapy; Target therapy

INTRODUCTION

Low-grade glioma (LGG) is the most common brain tumor in children, accounting for approximately 30% of pediatric brain tumors [1, 2, 3]. Pediatric LGGs (pLGG) are heterogeneous neoplasms, including tumors of primarily glial histology and tumors of mixed neuronal-glial morphology [4]. Most LGGs in children are pilocytic astrocytoma (65%), followed by LGGs not otherwise specified in 21% of cases. The long-term survival is excellent, over 90%, but the progression-free survival (PFS) is only approximately 50%, and these patients require adjuvant therapy [5]. Surgical resection is important for the management of pLGG, and complete resection is the most favorable predictor of survival in patients with pLGG. In patients where gross tumor removal cannot be achieved, the progression of the tumor has been treated with adjuvant chemotherapy and radiation. Therefore conventional therapies, including surgical resection, radiotherapy, and chemotherapy, often provide long-term neurological and endocrine complications [6].

Recent molecular data has emerged to suggest that pLGG near universally upregulates the RAS-mitogen-activated protein kinase (RAS/MAPK) pathway [7, 8, 9]. The 5th edition of WHO Classification of Central Nervous System Tumors, published in 2021, introduces the change in the classification of gliomas into adult-type and pediatric-type diffuse gliomas according to molecular and genetic differences, and the pediatric glioma is further divided into low-grade and high-grade gliomas [3]. Despite clinical and molecular distinctions between those diffuse gliomas that primarily occur in adults (termed “adult-type”) and those that occur primarily in children (termed “pediatric-type”), pediatric-type tumors may sometimes occur in adults, particularly young adults, and adult-type tumors may more rarely occur in children. The pediatric low-grade group includes 4 entities that feature diffuse growth in the brain, and molecular work-up helps to characterize the lesion as one type or the other. Pediatric type diffuse LGGs expected to have good prognoses has three new tumors: 1) diffuse astrocytoma, MYB or MYBL1-altered, 2) polymorphous low grade neuroepithelial tumor of the young (PLNTY), and 3) diffuse LGG-MAPK altered. Five well-established tumors are classified to circumscribed astrocytic gliomas in which the tumor–non-tumor border is comparatively sharper, including 1) pilocytic astrocytoma, 2) pleomorphic xanthoastrocytoma, 3) subependymal giant cell astrocytoma, 4) chordoid glioma, and 5) astroblastoma, MN1-altered. Many of the circumscribed astrocytic gliomas share several morphological and molecular features with pediatric-type diffuse LGGs such as MAPK-activating alterations. Circumscribed astrocytic gliomas are relatively common in children, but can also occur in young adults [3].

TREATMENT OF pLGG

The mainstay of therapy for progressive or symptomatic pLGG is surgical resection [10]. Gross total resection (GTR) of tumors often requires no further therapy, and even subtotal resection may lead volume reduction and long-term tumor quiescence [8].

Several studies have shown that GTR correlates with increased PFS and overall survival (OS) in pLGG [5, 11]. Around 40% of all LGG patients can be cured by complete neurosurgical resection and second-look operation is highly recommended for patients with a postoperative residual or recurrent tumor [12, 13]. For the recurrent pLGGs, two thirds required subsequent therapies such as surgery, radiotherapy or chemotherapy. Unfavorable prognostic factors for OS include subtotal resection, young age, and unfavorable tumor location as brainstem or optic pathway [14, 15, 16].

Historically, radiation therapy has been used in the up-front and salvage treatment of pLGG. However, traditional photon radiotherapy is associated with several long-term adverse effects including cognitive decline, endocrine dysfunction, growth abnormalities, vascular damage, and secondary malignancies [6, 17, 18, 19, 20]. Thus, radiotherapy is generally reserved for older pLGG patients and for whom have progressive or refractory diseases after surgery, chemotherapy and/or targeted agents.

Newer radiation modalities including conformal radiation and proton beam radiotherapy have allowed sparing of normal brain regions that is exposed to low and intermediate doses of radiation compared to conventional photon radiotherapy.

CONVENTIONAL CHEMOTHERAPY OF pLGG

Chemotherapy has been considered for young children with progressive or incompletely resected pLGG to delay radiotherapy and recurrent tumors in unfavorable location infeasible surgery. This is especially important in children with neurofibromatosis type-1 (NF-1) who are at increased risk of pLGG, typically optic pathway glioma for high risk of secondary malignancy due to their germline mutation. The most commonly used chemotherapy regimens for pLGGs are carboplatin plus vincristine and/or etoposide, a combination of thioguanine, procarbazine, lomustine (CCNU) and vincristine (TPCV) or vinblastine monotherapy. Temozolomide, the treatment of choice for adult diffuse gliomas, is not effective standard therapy for pLGG [21]. Chemotherapy regimens in newly-diagnosed pLGG showed 50%–80% of 3-year PFS [14, 15, 22, 23, 24, 25]. The use of chemotherapy to manage newly diagnosed LGG was first introduced in the 1980s. Procarbazine, 6-thioguanine, dibromodulcitol, CCNU, and vincristine regimen given to 42 pediatric patients showed 78% of 5-year survival rate [24]. Twenty-three patients of this group had juvenile pilocytic astrocytomas, 11 had astrocytomas, one had oligodendroglioma, one had ganglioglioma, and six had radiographically diagnosed LGGs. Phase II study of carboplatin single therapy in 81 children with progressive LGG had 64% of 3-year PFS and 84% of OS [25]. One of the largest studies, the German multicenter, cooperative Hirntumorstudien (HIT)-LGG-1996 study reported long-term follow-up result of 1,032 patients, and 668 children were under observation and 363 had started adjuvant treatment, as either chemotherapy or radiotherapy [12]. Vincristine+carboplatin (VC) chemotherapy was administered to 216 children including 55 patients with NF-1. Compared with the radiotherapy group, the patients in the chemotherapy group were younger and more often had NF-1. Best tumor responses were complete remission (CR) in 3.8%, partial remission (PR) in 31.6%, and overall response/stable disease (SD) in 56.5%, while 8.1% had tumor progression during follow-up period. In this report, NF-1 status was positive predictor for OS in children treated with chemotherapy. The European comprehensive treatment strategy for childhood LGG, the International Society of Pediatric Oncology Low Grade Glioma (SIOP LGG) Committee reported a randomized controlled study in the non-NF1 group by adding etoposide to the standard VC combination and the addition of etoposide to VC did not improve PFS or OS [15]. The other randomized trial comparing VC versus TPCV regimen showed a trend of higher 5-year PFS without statistical significance [14]. Through these two studies, VC recommended as standard first-line therapy [22]. Vinblastine monotherapy is also recommended according to the result with low toxicity and comparable PFS to previous chemotherapy studies including VC regimen [23]. The two currently recommended standard therapies are VC for 81 weeks or a weekly IV administration of vinblastine for 70 weeks [26].

THE GENOMIC LANDSCAPE OF pLGG

Genomic profiling studies have confirmed pLGGs to be distinct from adult LGGs and proved pLGGs have somatic driver genetic alterations that result in activation of the MAPK pathway [7, 27, 28]. Diffuse astrocytoma, MYB-altered or MYBL1-altered is defined as having MYB or MYBL1 alterations, and angiocentric gliomas also commonly have MYB fusions. In both tumors, these alterations trigger MAPK pathway activation. Polymorphic PLNTY neoplasm includes histological components including oligodendrogliomas, gangliomas, pilocytic astrocytomas, dysembryoplastic neuroepithelial tumors, and pleomorphic xanthoastrocytomas. PLNTY is characterized by MAPK-activating mutations in BRAF, FGFR2, FGFR3 or even NTRK alterations. Diffuse LGG, MAPK pathway alterations are similar to the other previously mentioned tumors, and while other MAPK activating driver mutations are also found, tumor progression is mainly caused by BRAF mutations or FGFR1 alterations [4, 29]. They are all amalgamated by MAPK pathway activation and IDH mutations or lack of IDH mutations in histone-encoding genes.

MAPK activation resulting in downstream activation of the mTOR pathway is predominant in pLGG and offers a useful target for therapy [30]. Rearrangements affecting the genes BRAF and KIAA1549 are the most frequent alterations in all pLGGs [7, 27, 28]. KIAA1549-BRAF rearrangements results in constitutive activation of the BRAF kinase with downstream activation of MAPK signaling. BRAFV600E mutations are also tumorigenic driver particularly in the group of ganglioglioma and pilocytic astrocytoma [7, 29] and are associated with worse outcome when accompanying alterations of CDKN2A [31, 32].

TARGETED TREATMENTS FOR pLGG

Various agents that target the MAPK pathway, such as MEK or BRAF inhibitors are currently attempted in pLGGs and encouraging results has been recently reported [33, 34, 35, 36, 37]. Dabrafenib and vemurafenib of those agents, is expected to improve clinical outcomes with few adverse events and good tolerance in patients with BRAF-mutated gliomas [35, 36, 38, 39, 40, 41, 42].

Dabrafenib and vemurafenib are both orally bioavailable, potent and selective inhibitors of BRAF kinases that harbor V600 mutations, binding to the ATP binding domain of mutant BRAF. Early reports showed dramatic responses to dabrafenib and vemurafenib in infants and children with recurrent LGG that harbor BRAFV600E mutations [38, 39, 40, 42], and led to multi-institutional phase I/II trials of these agents in children with recurrent BRAFV600E mutated LGG. Thirty-two children with pLGG on dabrafenib have been revealed 13 PR and 1 CR with an overall RR of 44% and 85% of 1-year PFS [36, 43]. Phase I trial of vemurafenib in children with recurrent or refractory gliomas containing the BRAFV600E mutation also showed 1 CR, 5 PR, and 13 SD [44].

Despite high initial response rates, developing resistance to BRAF inhibitors has reported from melanoma trials, and the reason of this resistance is explained by reactivation of the MAPK pathway [45]. Combination therapy of BRAF inhibitors and MEK inhibitors that inhibit MAPK pathway demonstrated overcome of BRAF inhibitor resistance, and superiority over a BRAF inhibitor alone in CNS tumors including glioma [35, 46].

To evaluate the efficacy and safety of dabrafenib in combination with trametinib for pediatric gliomas, a nationwide phase II pediatric study is progress (NCT02684058) and the result of interim analysis is reported [47]. In the LGG cohort, median follow-up was 32.2 months and 9 (69%) of 13 patients had an objective response including one complete response, six partial responses, and two minor responses. The most common grade 3 or worse adverse events was fatigue, headache, and neutropenia. Dabrafenib plus trametinib showed clinically meaningful activity in patients with BRAFV600E mutation-positive recurrent or refractory LGG and BRAFV600E testing could potentially be adopted in clinical practice for patients with glioma.

The other drug that has been actively studied is selumetinib, MEK1/2 inhibitor. Promising preclinical data, led to the phase I and II trial of selumetinib in children with recurrent and refractory pLGG [48, 49]. In the phase 1 study, 37% of 38 eligible patients completed all 26 cycles of protocol treatment with at least SD and 2-year PFS was 69%±9.8%. Rash, increased amylase/lipase and mucositis were the main dose limiting toxicities [48]. In the next phase II study (NCT01089101), the result of the children with PA harboring either one of the two most common BRAF aberrations (KIAA1549-BRAF fusion or the BRAFV600E mutation), and NF-1 associated LGG was reported [49]. Selumetinib was given orally at the recommended phase 2 dose of 25 mg/m2 twice daily in 28-day courses for up to 26 courses. Among 25 eligible and evaluable patients with PA, 9 (36%) of 25 patients achieved a sustained partial response, and the median follow-up for the 11 patients who had not had a progression event was 36 months. In the group of NF-1 associated LGG, 10 (40%) of 25 patients achieved a sustained partial response and median follow-up was 48 months for the 17 patients without a progression event. The most common grade 3 or worse adverse events were elevated creatine phosphokinase and maculopapular rash. Throughout these trials, selumetinib was considered being an alternative to standard chemotherapy for the patients with recurrent or progressive BRAF aberrated PA and NF-1 associated LGG. Two Children’s Oncology Group phase 3 studies (NCT03871257 and NCT04166409) comparing standard chemotherapy (carboplatin and vincristine) to selumetinib in patients with newly diagnosed pLGG both with and without NF1 are ongoing.

Trametinib is another oral MEK-1/2 inhibitor, and several clinical trials are ongoing as single therapy or combination therapy (NCT05180825, phase II study comparing a daily trametinib to weekly vinblastine during 18 courses of 4 weeks each; NCT04485559, phase I trial studying the side effects and best dose of trametinib and everolimus in treating pediatric and young adult patients with LGG; NCT03363217, a phase 2, open-label, oral administration of trametinib). Another oral MEK inhibitors, binimetinib (MEK-162) and cobemitinib have completed or progressed phase 1 trials in children with recurrent, refractory or progressive pLGG and other BRAF-mutated recurrent pediatric solid tumors which are Ras/Raf pathway activated malignancies [50]. Therapies targeting the mTOR pathway, such as everolimus, are also currently being tested evaluating everolimus in children with recurrent/progressive pLGG [51, 52]. The phase 2 study of everolimus showed the results of 2 PR, 10 SD without CR in 23 children with recurrent and/or progressive pLGGs.

CONCLUSION

Aside from ‘watch and waiting’ when the tumor is low-grade and has gross total or near-total resection, the optimal adjuvant treatments for those pLGG remain unclear. Early results of new radiation therapy and MAPK-targeted agents have been quite promising in the children with LGG. Novel target agents such as BRAF inhibitors and MEK inhibitors have demonstrated high response rates, good PFS and relatively tolerable toxicity. Nevertheless, many questions regarding targeted therapies remain unanswered, such as the proper duration of therapy, durability of response, late toxicities and the use of these agents as up-front therapy or in combination with other agents. Hence, until now, chemotherapy with CV or vinblastine or focal radiotherapy in selected cases (in older children with relatively localized tumors) remains standard therapy. Further clinical trials will respond better answers to standardize and customize treatment protocols for pLGGs.

Notes

Ethics Statement:Not applicable

Conflicts of Interest:The author has no potential conflicts of interest to disclose.

Funding Statement:None

Availability of Data and Material

The data generated or analyzed during the current study are available in the PubMed database.

References

    1. Ward E, DeSantis C, Robbins A, Kohler B, Jemal A. Childhood and adolescent cancer statistics, 2014. CA Cancer J Clin 2014;64:83–103.
    1. Wen PY, Packer RJ. The 2021 WHO classification of tumors of the central nervous system: clinical implications. Neuro Oncol 2021;23:1215–1217.
    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;23:1231–1251.
    1. Ryall S, Tabori U, Hawkins C. Pediatric low-grade glioma in the era of molecular diagnostics. Acta Neuropathol Commun 2020;8:30
    1. 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;68:1548–1554.
      discussion 1554-5.
    1. 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 2010;13:223–234.
    1. 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;45:602–612.
    1. Northcott PA, Pfister SM, Jones DT. Next-generation (epi)genetic drivers of childhood brain tumours and the outlook for targeted therapies. Lancet Oncol 2015;16:e293–e302.
    1. Collins VP, Jones DT, Giannini C. Pilocytic astrocytoma: pathology, molecular mechanisms and markers. Acta Neuropathol 2015;129:775–788.
    1. Greuter L, Guzman R, Soleman J. Pediatric and adult low-grade gliomas: where do the differences lie? Children (Basel) 2021;8:1075
    1. Fisher BJ, Leighton CC, Vujovic O, Macdonald DR, Stitt L. Results of a policy of surveillance alone after surgical management of pediatric low grade gliomas. Int J Radiat Oncol Biol Phys 2001;51:704–710.
    1. 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;14:1265–1284.
    1. Gnekow AK, Kandels D, Tilburg CV, Azizi AA, Opocher E, Stokland T, et al. SIOP-E-BTG and GPOH guidelines for diagnosis and treatment of children and adolescents with low grade glioma. Klin Padiatr 2019;231:107–135.
    1. 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;30:2641–2647.
    1. Gnekow AK, Walker DA, Kandels D, Picton S, Giorgio Perilongo, 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;81:206–225.
    1. Stokland T, Liu JF, Ironside JW, Ellison DW, Taylor R, Robinson KJ, 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;12:1257–1268.
    1. Krishnatry R, Zhukova N, Guerreiro Stucklin AS, Pole JD, Mistry M, Fried I, et al. Clinical and treatment factors determining long-term outcomes for adult survivors of childhood low-grade glioma: a population-based study. Cancer 2016;122:1261–1269.
    1. 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. J Clin Oncol 2009;27:3691–3697.
    1. 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;68:932–938.
    1. 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;61:1173–1179.
    1. Nicholson HS, Kretschmar CS, Krailo M, Bernstein M, Kadota R, Fort D, et al. Phase 2 study of temozolomide in children and adolescents with recurrent central nervous system tumors: a report from the Children’s Oncology Group. Cancer 2007;110:1542–1550.
    1. Ater JL, Xia C, Mazewski CM, Booth TN, Freyer DR, Packer RJ, et al. Nonrandomized comparison of neurofibromatosis type 1 and non-neurofibromatosis type 1 children who received carboplatin and vincristine for progressive low-grade glioma: a report from the Children’s Oncology Group. Cancer 2016;122:1928–1936.
    1. 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. J Clin Oncol 2016;34:3537–3543.
    1. Prados MD, Edwards MS, Rabbitt J, Lamborn K, Davis RL, Levin VA. Treatment of pediatric low-grade gliomas with a nitrosourea-based multiagent chemotherapy regimen. J Neurooncol 1997;32:235–241.
    1. Gururangan S, Cavazos CM, Ashley D, Herndon JE 2nd, Bruggers CS, Moghrabi A, et al. Phase II study of carboplatin in children with progressive low-grade gliomas. J Clin Oncol 2002;20:2951–2958.
    1. Horbinski C, Berger T, Packer RJ, Wen PY. Clinical implications of the 2021 edition of the WHO classification of central nervous system tumours. Nat Rev Neurol 2022;18:515–529.
    1. Jones DT, Gronych J, Lichter P, Witt O, Pfister SM. MAPK pathway activation in pilocytic astrocytoma. Cell Mol Life Sci 2012;69:1799–1811.
    1. Ahrendsen JT, Sinai C, Meredith DM, Malinowski SW, Cooney TM, Bandopadhayay P, et al. Molecular alterations in pediatric low-grade gliomas that led to death. J Neuropathol Exp Neurol 2021;80:1052–1059.
    1. 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;37:569–583.e5.
    1. Pachow D, Wick W, Gutmann DH, Mawrin C. The mTOR signaling pathway as a treatment target for intracranial neoplasms. Neuro Oncol 2015;17:189–199.
    1. 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. J Clin Oncol 2017;35:2934–2941.
    1. Coutant M, Lhermitte B, Guérin E, Chammas A, Reita D, Sebastia C, et al. Retrospective and integrative analyses of molecular characteristics and their specific imaging parameters in pediatric grade 1 gliomas. Pediatr Blood Cancer 2022;69:e29575
    1. Hofer S, Berthod G, Riklin C, Rushing E, Feilchenfeldt J. BRAF V600E mutation: a treatable driver mutation in pleomorphic xanthoastrocytoma (PXA). Acta Oncol 2016;55:122–123.
    1. Lee EQ, Ruland S, LeBoeuf NR, Wen PY, Santagata S. Successful treatment of a progressive BRAF V600E-mutated anaplastic pleomorphic xanthoastrocytoma with vemurafenib monotherapy. J Clin Oncol 2016;34:e87–e89.
    1. Brown NF, Carter T, Kitchen N, Mulholland P. Dabrafenib and trametinib in BRAFV600E mutated glioma. CNS Oncol 2017;6:291–296.
    1. 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;25:7303–7311.
    1. Fangusaro JR, Onar-Thomas A, Poussaint TY, Wu S, Ligon AH, Lindeman NI, et al. Corrigendum to: LTBK-01. Updates on the phase Ii and re-treatment study of Azd6244 (Selumetinib) for children with recurrent or refractory pediatric low grade glioma: a pediatric brain tumor consortium (PBTC) study. Neuro Oncol 2022;24:1404
    1. del Bufalo F, Carai A, Figà-Talamanca L, Pettorini B, Mallucci C, Giangaspero F, et al. Response of recurrent BRAFV600E mutated ganglioglioma to vemurafenib as single agent. J Transl Med 2014;12:356
    1. Del Bufalo F, Ceglie G, Cacchione A, Alessi I, Colafati GS, Carai A, et al. BRAF V600E inhibitor (vemurafenib) for BRAF V600E mutated low grade gliomas. Front Oncol 2018;8:526
    1. van Tilburg CM, Selt F, Sahm F, Bächli H, Pfister SM, Witt O, et al. Response in a child with a BRAF V600E mutated desmoplastic infantile astrocytoma upon retreatment with vemurafenib. Pediatr Blood Cancer 2018;65:e26893
    1. Lassaletta A, Guerreiro Stucklin A, Ramaswamy V, Zapotocky M, McKeown T, Hawkins C, et al. Profound clinical and radiological response to BRAF inhibition in a 2-month-old diencephalic child with hypothalamic/chiasmatic glioma. Pediatr Blood Cancer 2016;63:2038–2041.
    1. Bavle A, Jones J, Lin FY, Malphrus A, Adesina A, Su J. Dramatic clinical and radiographic response to BRAF inhibition in a patient with progressive disseminated optic pathway glioma refractory to MEK inhibition. Pediatr Hematol Oncol 2017;34:254–259.
    1. Kieran MW, Geoerger B, Dunkel IJ, Broniscer A, Hargrave D, Hingorani P, et al. A phase I and pharmacokinetic study of oral dabrafenib in children and adolescent patients with recurrent or refractory BRAF V600 mutation-positive solid tumors. Clin Cancer Res 2019;25:7294–7302.
    1. Nicolaides T, Nazemi KJ, Crawford J, Kilburn L, Minturn J, Gajjar A, et al. Phase I study of vemurafenib in children with recurrent or progressive BRAFV600E mutant brain tumors: pacific pediatric neuro-oncology consortium study (PNOC-002). Oncotarget 2020;11:1942–1952.
    1. Solit DB, Rosen N. Resistance to BRAF inhibition in melanomas. N Engl J Med 2011;364:772–774.
    1. Migliorini D, Aguiar D, Vargas MI, Lobrinus A, Dietrich PY. BRAF/MEK double blockade in refractory anaplastic pleomorphic xanthoastrocytoma. Neurology 2017;88:1291–1293.
    1. 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. Lancet Oncol 2022;23:53–64.
    1. Banerjee A, Jakacki RI, Onar-Thomas A, Wu S, Nicolaides T, Young Poussaint T, et al. A phase I trial of the MEK inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or refractory low-grade glioma: a pediatric brain tumor consortium (PBTC) study. Neuro Oncol 2017;19:1135–1144.
    1. 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;20:1011–1022.
    1. de Blank P, Fouladi M, Huse JT. Molecular markers and targeted therapy in pediatric low-grade glioma. J Neurooncol 2020;150:5–15.
    1. Cacchione A, Lodi M, Carai A, Miele E, Tartaglia M, Megaro G, et al. Upfront treatment with mTOR inhibitor everolimus in pediatric low-grade gliomas: a single-center experience. Int J Cancer 2021;148:2522–2534.
    1. Wright KD, Yao X, London WB, Kao PC, Gore L, Hunger S, et al. A POETIC Phase II study of continuous oral everolimus in recurrent, radiographically progressive pediatric low-grade glioma. Pediatr Blood Cancer 2021;68:e28787

Metrics
Share
ORCID IDs
PERMALINK