Next Article in Journal
Marginal Bone Level Evaluation of Fixed Partial Dental Prostheses Using Preformed Stock versus CAD/CAM Customized Abutments
Previous Article in Journal
Modeling Endometrium Biology and Disease
Previous Article in Special Issue
Endogenous Retroviral Elements in Human Development and Central Nervous System Embryonal Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Current State of Radiotherapy for Pediatric Brain Tumors: An Overview of Post-Radiotherapy Neurocognitive Decline and Outcomes

1
School of Medicine, Mercer University School of Medicine, Savannah, GA 31404, USA
2
Department of Surgery, Eisenhower Army Medical Center, Augusta, GA 30905, USA
3
Department of Neurosurgery, University of California San Diego, San Diego, CA 92103, USA
4
Department of Neurosurgery, University of California Irvine, Orange, CA 92868, USA
5
Johns Hopkins Whiting School of Engineering, Baltimore, MD 21218, USA
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(7), 1050; https://doi.org/10.3390/jpm12071050
Submission received: 30 April 2022 / Revised: 10 June 2022 / Accepted: 13 June 2022 / Published: 27 June 2022

Abstract

:
Tumors of the central nervous system are the most common solid malignancies diagnosed in children. While common, they are also found to have some of the lowest survival rates of all malignancies. Treatment of childhood brain tumors often consists of operative gross total resection with adjuvant chemotherapy or radiotherapy. The current body of literature is largely inconclusive regarding the overall benefit of adjuvant chemo- or radiotherapy. However, it is known that both are associated with conditions that lower the quality of life in children who undergo those treatments. Chemotherapy is often associated with nausea, emesis, significant fatigue, immunosuppression, and alopecia. While radiotherapy can be effective for achieving local control, it is associated with late effects such as endocrine dysfunction, secondary malignancy, and neurocognitive decline. Advancements in radiotherapy grant both an increase in lifetime survival and an increased lifetime for survivors to contend with these late effects. In this review, the authors examined all the published literature, analyzing the results of clinical trials, case series, and technical notes on patients undergoing radiotherapy for the treatment of tumors of the central nervous system with a focus on neurocognitive decline and survival outcomes.

1. Introduction

Central nervous system tumors are the leading cause of cancer mortality in childhood and are the second-most-common malignancy diagnosed in this patient population behind leukemia [1,2]. Intracranial tumors contribute to 20% of all pediatric malignancies with an incidence of 3.7 cases per 100,000 children [3,4,5]. Initial maximal safe resection is the standard of care for most of intracranial tumors, with prognosis dependent upon extent of resection for medulloblastoma [6], craniopharyngioma [7], ependymoma [8], low-grade glioma [9], and high-grade glioblastoma [10]. However, invasion into eloquent anatomy can prohibit maximal resection and, consequently, maximal progression-free survival [9]. Multimodal-treatment strategies potentially encompass early surgical resection, local radiation, complete craniospinal radiation, and chemotherapeutic treatments to achieve the maximal clinical outcome. Local control potentially improves with adjuvant radiotherapy for many pediatric brain tumors and is used in the management of a substantial fraction of solid malignancies [11].
While advances in multimodal therapy have improved outcomes [12], they may come at the cost of long-term, late effects [13], such as neurocognitive deficits [14,15], hearing loss [16], endocrine dysfunction [17], secondary malignancy [18,19], and radiation necrosis [19,20]. Neurocognitive dysfunction was recognized early on as a late effect of craniospinal irradiation when, in 1969, HJG Bloom noted a high rate of dementia in a cohort of 82 patients with medulloblastoma, who received cranial or craniospinal radiotherapy before the age of 2 [21]. In totality, late effects contribute to reduced direct and indirect health-related quality of life (HRQoL) [22,23,24,25,26,27], with neurocognitive dysfunction identified as the leading cause of reduced quality of life in long-term survivors of pediatric brain tumors [28].
There is well-established literature on the developmental consequences of these late effects, with children at increased risk for cognitive, emotional, and behavioral decline [14,29,30], regardless of focal [31,32,33] or whole-brain irradiation [29,30,34,35]. When compared to surgery or chemotherapy alone, radiotherapy is consistently associated with poorer neuropsychological outcomes [29,36]. Indeed, a cross-sectional survey of 342 patients with brain tumors treated with radiotherapy and 479 sibling controls showed the former patients were 28.8 times less likely to drive a car and 10.8 times less likely to be employed compared to their siblings [37]. In addition, these patients are found to have increased chances of being involved with theft, fraud, or assault [38,39]. The severity of these morbidities is directly related to dosage of radiation and volume of normal tissue exposed to radiation, while inversely related to age at time of irradiation [17,37,40,41,42,43,44,45]. Tumor size, location, baseline intellectual function, and enzyme polymorphisms with genetic predisposition also influence elements of cognitive function [46,47].
To help mediate against these long-term sequelae, approaches for prevention of and reduction in neurocognitive dysfunction include avoiding the use of radiotherapy in young children and reducing the dose and volume of brain that is irradiated [28]. Development of intensity-modulated radiotherapy (IMRT), volumetric-arc therapy (VMAT), and proton-beam radiotherapy (PBRT) over the past decades have sought to maximize the dose coverage of the tumor, while minimizing coverage to nearby normal tissue when compared to traditional techniques, such as conformal radiotherapy [48].
Improvements in long-term survival further underscore the impact of these late effects, as survivors now increasingly have longer lifespans, wherein, they must contend with these morbidities [48]. With the concomitant rise in prevalence, opportunities emerge to further understand the nature and consequences of the late effects of radiotherapy deeper into the patients’ course of disease. In this review, the authors examined all the published literature analyzing results of clinical trials, case series, and technical notes on patients undergoing radiotherapy for the treatment of tumors of the central nervous system, with an emphasis on neurocognitive decline and survival outcomes.

2. Survival Outcomes and Benefit of Radiotherapy

2.1. Medulloblastoma

With approximately 500 children being diagnosed each year in the United States, medulloblastoma is the most common malignant brain tumor of childhood and is the most common embryonal tumor [49,50,51]. As with other tumors, medulloblastoma usually presents with signs and symptoms of increased intracranial pressure secondary to obstructive hydrocephalus [50]. Additionally, 6%–32% of presentations are metastatic [52,53]. Initial work-up includes magnetic resonance imaging (MRI) that will classically reveal a contrast-enhancing cerebellar-midline mass [54,55]. Radiotherapy is an essential element in the treatment of pediatric medulloblastoma [51]. In the past two decades, advances in radiotherapy have included the development of a better-target volume delineation using imaging and use of adaptive techniques. IMRT, VMAT, image-guided radiotherapy, and particle-beam therapy [56,57,58]. Current dose-fractionation protocol includes 23.4 Gy in 13 fractions of craniospinal irradiation with a posterior fossa boost of 30.6 Gy in 17 fractions (Table 1) [59,60,61,62]. However, a new clinical trial has noted that current protocols lead to significant post-radiotherapy neurotoxicity. Thus, a reduction in dose or in some cases, elimination of craniospinal irradiation may be necessary. The clinical trial has proposed a new protocol for WNT 2-driven medulloblastoma-radiotherapy dosing: 18 Gy in 10 fractions plus a tumor-bed boost of 36 Gy in 20 fractions for a total primary site dose of 54 Gy in 30 fractions without concurrent chemotherapy followed by the standard six cycles of adjuvant systemic chemotherapy. By reducing the dosage to the craniospinal axis and reducing the total tumor-bed dose, the investigators are hoping this new protocol will decrease the late side effects of craniospinal irradiation and morbidity [63].
Prior treatment protocols emphasized higher radiation dosages to achieve higher cure rates, however contemporary protocols utilize risk-based dosing after accounting for the late effects of neurotoxicity; higher-risk patients will still receive high dosages [74], while standard risk indicates a lower dose [14,40,75,76,77,78]. Current treatment for medulloblastoma may involve gross total resection followed by radiotherapy and platinum-based chemotherapy [78,79,80,81]. These patients are classified postoperatively as standard risk or high risk (>1.5 cm2 residual disease after resection, known metastatic disease, or anaplastic large-cell histology) [51,82]. Children under 3–5 years are treated with chemotherapy only [83,84,85]. Prospective studies have shown 5-year event-free survival at 80–82% for average risk [59] and up to 70% for high-risk patients with current management protocol [86,87].
Risk stratification has classically involved evaluating the extent of the disease and the age at diagnosis, with younger age conferring a negative prognosis [52]. A study of 188 children with medulloblastoma found that those with no evidence of metastatic disease had a significantly higher 5-year progression-free survival (PFS) at 70%, compared to those with microscopic tumor cells in the CSF (57%) or those with gross nodular seeding (40%) [88]. This same study found the PFS of children between 18 months and 3 years to be 32% compared to 58% for those older than three. Decreased amounts of radiation therapy used in children younger than three was thought to play a role in the difference between the two groups.
The 2021 World Health Organization (WHO) Classification of Tumors of the Central Nervous System has separated medulloblastomas into two main types: molecularly defined and histologically defined [89]. The first main classification, molecularly defined medulloblastomas, are divided into four subtypes based on respective markers: WNT-activated, SHH-activated and TP53-wildtype, SHH-activated and TP53-mutant, and non-WNT/non-SHH. The second main classification, histologically defined medulloblastoma, is under its own subtype as “histologically defined” [89]. A 2010 meta-analysis with 270 children younger than five years assessed the prognostic role of clinical parameters and histology of early-childhood medulloblastoma and found that the 8-year event-free survival (EFS) and overall survival (OS) were both significantly higher in those with desmoplastic/nodular and desmoplastic with extensive nodularity, compared to those with either classic or anaplastic medulloblastoma [90]. The four molecular subtypes of medulloblastoma may be divided by genetic mutation for the purposes of risk stratification. The Wingless-related integration site (Wnt) pathway-activation group has the best prognosis, with an overall survival rate of 95%, and is mainly found in WNT-activated medulloblastoma. However, the Wnt-pathway activation is only present in ~10% of cases [91]. The activation pathway for sonic hedgehog (SHH), present in 30% of cases, is found chiefly in infants and adults. The overall survival for those with this mutation is ~75% [91]. The worst prognosis belongs to the non-WNT/non-SHH pathways, where some tumors display a high-level overactivation of the MYC protooncogene, yielding overall survival of ~50% at five years [92].
Treatment of medulloblastoma typically involves a combination of surgery, radiation, and multiagent chemotherapy [50,93,94,95,96]. The surgeon can achieve a margin-free excision or a near-total excision in most cases. No investigators have completed prospective, randomized trials evaluating survival in those with total versus near-total excision to date. A 2018 systematic review of retrospective studies, assessing the impact of the extent of resection on survival, found 16 articles with 1489 patients that did show a statistically significant association and 20 articles with 2335 patients that showed no significant association between the two groups. The authors concluded that the prognostic importance of the extent of resection for medulloblastoma is unclear [92]. Another retrospective study of 787 patients with medulloblastoma separated patients by their molecular subgroup, and the predictive value of the increased extent of resection decreased when the different subgroups were considered [97]. Thus, the standard of care for maximal, safe surgical resection remains. Surgical excision of small, residual portions of medulloblastoma is not recommended, when the likelihood of neurological morbidity is high. There is insufficient evidence that total resection imparts a survival benefit compared to subtotal resection [97].
Radiation therapy is started following surgical excision and is limited by its toxicity [94,95,96,98]. Proton-beam irradiation is a newer technique that is equally effective compared to conventional radiation therapy in treating posterior fossa and craniospinal disease, while significantly reducing the irradiation dose to the surrounding structures [98,99,100]. In a study comparing photon and PBRT in children with standard-risk medulloblastoma, Eaton et al. observed no significant difference between 6-year recurrence-free survival and overall survival [62]. In addition, patients at average risk are given a radiation boost to the primary site because 50–70% of recurrences develop in the posterior fossa, usually in the tumor bed and surrounding leptomeninges [101,102].
A Children’s Oncology Group study was a phase III trial of 379 children who have undergone total or near-total resection of a medulloblastoma, without evidence of disseminated disease [59,103]. Following resection, patients were treated with 23.4 Gy of craniospinal radiation, followed by a boost to the posterior fossa of 32.4 Gy. Weekly vincristine was given during radiation therapy, and a combination of vincristine, cisplatin, and either lomustine or cyclophosphamide was given for eight cycles. No significant difference in efficacy was found between the two chemotherapy regimens [59,103]. Infections were slightly higher in the cyclophosphamide group, while electrolyte disturbances were more common in the lomustine group. Event-free survival was higher in both groups receiving adjuvant radiation and chemotherapy than past surgery and radiation trials alone [59,103]. All patients undergoing this treatment developed hematologic toxicity. Approximately 25% developed ototoxicity. A cumulative cisplatin dose has not shown an increased event-free or overall survival, so lower doses are recommended in the treatment of medulloblastoma [104].
High-risk patients with metastatic or unresectable disease are usually treated with more aggressive chemotherapy and radiation, although there is no consensus about the most optimal therapy. Another Children’s Oncology Group study of 161 children ≥3 years of age with high-risk medulloblastoma were treated with postoperative radiation along with carboplatin and vincristine, followed by six maintenance cycles of cyclophosphamide, vincristine, and cisplatin [105]. Maintenance chemotherapy with cisplatin had a five-year progression-free and overall survival rates of 59% and 68%, respectively [105]. There were no treatment-related deaths reported. Another new strategy in the treatment of metastatic disease is hyperfractionated accelerated radiotherapy, which can be used in combination with high-intensity chemotherapy [106].
Therapies targeting molecular markers of medulloblastoma are currently under investigation. Vismodegib is an antagonist of the smoothened receptor that causes a disruption of transcription factors in the sonic-hedgehog pathway. It is currently only FDA-approved for the treatment of basal-cell carcinoma. Clinical trials are currently underway, and the phase 1 trials have shown that vismodegib is well-tolerated but is associated with teeth and bone abnormalities, including osteonecrosis in those taking concurrent steroids [107]. A phase II trial of 12 patients with sonic-hedgehog mutations had only 4 show a response to vismodegib, so further studies are needed to identify molecular patterns most susceptible to smoothened receptor antagonism [108].

2.2. Low-Grade Gliomas

Pilocytic astrocytoma (PA) is the most common primary brain tumor of childhood and is typically found in the cerebellum, but it may also present in the third ventricle, spinal cord, optic pathways, and cerebral hemispheres [109]. Maximal surgical resection is the standard of care; however, multimodal therapy is indicated with unresectable tumors in central locations, such as the optic pathway [110], and with recurrent tumors [111]. The prognosis for PA is among the best for primary brain tumors with a 5-year survival rate of 94% [112]. Grouped initially due to their similar gross and radiographic features, pilomyxoid astrocytoma (PMA) was distinguished from PA in 1999 [113]. They are now separated based on their histological appearance. PA has a biphasic architecture with protoplasmic cells, Rosenthal fibers, and eosinophilic granular bodies that are rare or absent in PMA. PMA has a monophasic architecture with a myxoid background and an angiocentric pattern [114]. PMA is critical to distinguish from PA as it has a more unpredictable course and is potentially more aggressive [113]. Maximal surgical resection is the mainstay of therapy for both PA and PMA, and radiation therapy is reserved for those who begin to progress after initial tumor resection [64]. The role of chemotherapy in low-grade astrocytoma is still uncertain. Still, it is generally limited to those with recurrent disease and young children in whom the risks of radiation therapy outweigh those of chemotherapy [64].

2.3. High-Grade Gliomas

High-grade gliomas (HGG) are divided into anaplastic glioma and glioblastoma. Both are aggressive, malignant tumors with poor prognoses. Length of survival in those with glioblastoma is dependent on tumor location and extent of resection [115]. A small retrospective study of 27 pediatric patients with glioblastoma found that those with superficial tumors amenable to complete resection had a median overall survival of 106 months compared to 11 months for those with incomplete resection [115]. As with other neoplasia of the central nervous system, maximal surgical resection is preferred at the outset to confirm the diagnosis, grade the tumor, and begin molecular analysis. When the tumor is not amenable to surgery, stereotactic biopsy is preferred. Once maximal resection is complete, the current standard of care in the treatment of glioblastoma is radiotherapy with concomitant temozolomide, an alkylating agent, followed by adjuvant temozolomide [65].
Radiotherapy is the most effective nonsurgical treatment for gliomas, however, glioma recurrence is common as glioma cells are highly radioresistant. It is thought that cancer stem cells contribute to radioresistance in gliomas through the Notch signaling pathway. A study by Wang et al. is discussed below in detail and provides insight on how inhibiting the Notch pathway leads to more radiosensitive glioma cells [116]. Furthermore, it has been shown that glioma stem cells are more radioresistant than the non-stem glioma cells [117]. Although temozolomide may increase radiotherapy sensitivity of tumor cells, glioblastomas remain highly resistant to radiation [118]. Due to the highly tumorigenic activity of these stem cells, enhancing their radiosensitivity has been a target for improving overall effectiveness of radiotherapy. Chemotherapy with molecular targeting has shown promise in enhancing radiosensitivity. Bao et al. has shown that inhibitors of two DNA-damage-checkpoint kinases, Chk1 and Chk2, increased the cell death of glioma stem cells in response to radiation [119]. However, this target is not exclusively expressed by tumor cells, limiting the therapeutic index of inhibitor therapy by increased radiosensitivity of normal cells as well [120]. In more detail, the Notch signaling pathway has emerged as a target of radioresistance in cancer stem cells. Normal notch signaling promotes self-renewal and dedifferentiation in many adult stem cells in breast [121,122], intestine [123,124], and neuronal tissue [125,126,127]. Dysfunctional Notch activity is seen in many human tumors, such as breast cancer [128,129], leukemia [130], and glioma [131]. The Notch pathway is dependent upon a final proteolytic step, wherein a gamma-secretase releases the intracellular domain of the Notch protein (NICD) [132,133]. Notch activation via NICD1 overexpression promotes tumorgenicity [120], whereas inhibition by gamma-secretase inhibitors (GSI’s), which are used to block Notch activity in vivo and in vitro, reduces tumorigenicity [134]. In a study that measured the tumor size of CD133+ gliomas both in vitro and in vivo in response to radiotherapy with Notch inhibition via GSI’s or knockdown expression, Wang et al. found significant increases in cell death at clinically relevant doses. The degree of cell death in non-stem cells was unaltered by Notch inhibition as compared to the CD133+ stem cells when irradiated. Additionally, constitutive expression of the active intracellular domains of Notch1 or Notch2 protects glioma stem cells against radiation. Reduction in Notch1 or Notch2 levels also leads to radio-sensitive glioma stem cells. Thus, overexpression of Notch in cancerous glioma cells may play a critical role in the radioresistance of glioma stem cells [116]. Lower doses of radiation are required when tumor cells are molecularly radiosensitive, thus, further research into molecular targeted therapies can ultimately contribute to reductions in the late effects of neurotoxicity.
While limited randomized trials on the treatment of HGG exist, a 2020 case report of a 3-year-old girl diagnosed with glioblastoma highlights the importance of molecular targeted therapy in the future of care [135]. She initially presented with progressive seizure activity. Neuroimaging revealed a significant, heterogeneously enhancing, mixed cystic and solid mass in the left frontal-parietal-temporal region. She underwent debulking surgery, and genomic analysis of the tumor showed a BRCA2 nonsense mutation. Standard adjuvant radiation and temozolomide were used initially. Nine months after surgery, treatment with a combination of olaparib, an inhibitor of poly ADP ribose polymerase, and temozolomide was used for 16 cycles. She remains neurologically intact and “continues to experience an exceptional and durable response (>2 years)” after her treatment, with no evidence of tumor recurrence in serial imaging [135]. This case underscores the importance of genomic sequencing of brain tumors and the role of progressing to more targeted molecular therapies instead of systemic chemotherapy in the future.

2.4. Brainstem Gliomas

Brainstem gliomas are a subgroup of astrocytoma that range from low-grade lesions that may not require any intervention to high-grade malignancies with poor prognoses [136,137,138,139]. Diffuse Midline Glioma (DMG) is important, since radiation therapy is the mainstay treatment. A subtype of particular interest, diffuse intrinsic pontine glioma (DIPG) is a rare, aggressive type that forms in the brainstem of pediatric patients. With most survival rates at less than one year, immediate treatment is imperative. The first-line treatment for DIPG is radiation therapy, as surgical resection is not a viable option [140]. The molecular etiology of DIPG may be attributed to the “H3 K27M-mutant” in ~80% of cases, as noted in WHO 2016. However, WHO 2021 has adapted a new name, “H3 K27M-altered”, to recognize other mechanisms where the pathogenic pathway may be altered in these tumors [89,141]. Approximately 500 new cases of brainstem glioma are diagnosed in the pediatric population each year [142,143]. Neurofibromatosis type I is one of the only known risk factors for developing a brainstem glioma [144,145,146]. Patients may present with cranial-nerve deficits corresponding to the location of the mass [138,139]. Exophytic tumors of the dorsal brainstem may cause obstructive hydrocephalus [147]. The site of these tumors provides a unique challenge for their treatment [40]. Maximal safe resection remains the standard of care when possible [9,66]. Radiation therapy may be used in those patients whose tumor burden is not amenable to surgery or those where gross total resection is not achieved [67,68,69]. Radiation therapy for brainstem gliomas has been used sparingly due to its side effects. One study has shown that children who received either conventionally fractionated or hyperfractionated radiation for brainstem gliomas had significantly lower IQs than those who received surgery alone [148]. However, this is likely confounded by those with more severe tumor burden being the ones who received radiation. Other side effects of radiation to the brainstem include mild nausea and vomiting, endocrinopathies from damage to the hypothalamic-pituitary axis, and brainstem necrosis, which is a rare but devastating outcome [149,150,151]. Over the past 30 years, multiple trials have shown that chemotherapy has activity against brainstem gliomas and could be a safe first-line therapy for those in which the consequences of brainstem radiation is desired to be avoided [152,153,154,155]. Combinations of vincristine and platinum-based chemotherapeutics have been studied, but no consensus on the optimal regimen has been reached [152,153,154].

2.5. Ependymoma

Ependymomas are tumors of the lining of the ventricular system that typically arise in the fourth ventricle in the first two decades of life [109]. Intracranial ependymoma contributes to 10% of all intracranial tumor presentations in children [112]. Five-year survival approaches 50–80% [70,71,72,156,157]. The 2021 World Health Organization Classification separates ependymomas according to a combination of histopathology, molecular markers, and anatomic sites. The molecular groups are now divided across the following anatomic sites: supratentorial, posterior fossa, and spinal compartments. WHO 2021 subcategorizes molecularly defined types of supratentorial ependymoma into two types: ZFTA fusion and YAP1 fusion. There are also two major molecular subtypes of posterior fossa ependymomas: group PFA and group PFB. The spinal tumor-molecular subtype is characterized by the presence of MYCN amplification. Lastly, the histological ependymoma subtypes include Myxopapillary ependymoma and subependymoma [89].
As with other surgeries in the posterior fossa, complications of ependymoma resection include cerebellar ataxia, lower cranial nerve damage, and posterior fossa syndrome [158]. Following maximal safe resection, further treatment is guided by the extent of resection and tumor grade. Local or craniospinal radiation with doses of 54–59.4 Gy is usually given to those with subtotal resection [70,71,72]. Extent of resection is the most determinant factor of prognosis [8,156,159,160,161,162,163,164,165]. Should recurrence occur, it is most commonly local [8,159,161,163]. A long-term prospective study evaluated the health-related quality of life (HRQoL) of 40 patients < 4 who received proton-radiation therapy for a central-nervous-system tumor [166]. Ependymoma was the most common tumor in the cohort (n = 22), followed by medulloblastoma (n = 9). The median age at radiotherapy was 2.5, and the median age at follow-up was 9.1. The authors found that the HRQoL was variable with just over a third of patients and families achieving levels equivalent to healthy children. In total, 90% of the patients were able to function in a regular classroom. Patients who developed hydrocephalus or those that required feeding-tube placement reported significantly lower HRQoL. Scores among the cohort remained stable from the baseline analysis until the last follow-up [166]. A similar retrospective study examining patients with posterior fossa brain tumors found that families with lower socioeconomic status also reported more inadequate measures for their quality of life [167].

2.6. Craniopharyngioma

Craniopharyngiomas are supratentorial tumors of childhood that arise from the remnants of Rathke’s pouch and may contain a cystic component [168]. Like pituitary adenomas, craniopharyngiomas may present with headaches and bitemporal hemianopia [169]. Disruption of the hypothalamic–pituitary axis may lead to diabetes insipidus or another endocrine dysfunction [169]. These symptoms may be present for over a year before a diagnosis is made, as this tumor is slow-growing [169]. Historically, the management of craniopharyngiomas has been controversial, with some advocating for more aggressive surgical management to achieve the maximal extent of resection and others advocating for a more conservative approach in the operating room, followed by radiation therapy [73]. Even when confirmed by imaging, gross total resection can result in recurrence up to 20–27% [7,170]. When treated with a conservative surgical resection with adjuvant radiotherapy, 10-year progression-free survival has been reported up to 84–100% [171,172,173]. While there is no absolute consensus today, most agree that optimal management includes removing the greatest tumor burden possible without introducing any iatrogenic deficits [174]. This is highlighted by the approximately 50% of survivors that have long-term sequelae that negatively impact their quality of life [175].

3. Neurocognitive Late Effects

3.1. Decline in Neurocognitive Development

Intelligence quotient (IQ) has been used as a metric to measure changes in neurocognitive development after the treatment of pediatric brain tumors with a mean score of 100 and a standard deviation of 15 to 16 [40]. Declines in IQ are likely attributed to failure to develop neurocognitively at a rate expected for the patients’ age, rather than a loss of previously established development. Rate of IQ decline is correlated with younger age at time of treatment, volume of brain irradiated, dose of irradiation, female sex, and hydrocephalus [29,35,40]. Merchant et al. showed that radiation dosimetry can be used to predict patient IQ after conformal radiotherapy treatment of localized ependymoma [176]. While the effects of radiotherapy are mainly implicated in explaining neurocognitive decline, additional factors related to diagnosis and treatment course, such as posterior fossa syndrome and hydrocephalus, are also known to negatively influence neurocognitive development [177]. Specifically, following surgical resection of medulloblastoma, up to 29% of patients experience posterior fossa syndrome, or cerebellar mutism syndrome, which is characterized by diminished or absent speech, ataxia, hypotonia, and emotional lability. These symptoms may be exacerbated by radiotherapy [178]. Declines are associated with similar declines in measures of basic academic achievement [14,179,180], which may present as differences in performance, with standardized testing as early as one-year post-treatment. Declines may continue as late as ten years post-treatment [29,181]. Certain deficits due to neurocognitive decline may be delayed in presentation, due to normal ability being expected later during childhood development, such as speech (Table 2) [182].

3.2. Mechanism of Neurocognitive Decline

While the pathophysiology of radiation induced long-term CNS damage is not fully understood, hypotheses generally fall under the primary effects or secondary effects of radiation [40]. Primary effects may be caused by inciting damage to progenitor cells, vasculature, inflammatory cells, and stromal cells [187]. Another mechanism consists of the radiation-induced destruction of oligodendrocytes, which causes inadequate myelination and white-matter necrosis. Secondary effects include radiation-induced oxidative stress in the myelin membrane [184]. This is extremely pertinent in pediatric tumors, as full myelination of the cerebral cortex is achieved in early adulthood [188]. This mechanism has been further substantiated over the past few decades by new technologies that quantify the volumes of brain tissue [189]. For example, in a study that measured post-operative IQ after treatment of medulloblastoma with age-matched controls, the survivors of medulloblastoma had statistically significant lower volumes of white matter and IQ scores. IQ scores had a statistically significant inverse association with volumes of white matter [179]. Additional longitudinal studies show regions receiving an increased dose of radiation are associated with greater rates of decline in white-matter volume [185,186]. This mechanism could have a direct effect on processing speed, memory, and, in severe cases, cause ataxia, urinary incontinence, and dementia [183].
Certain eloquent areas are also associated with memory creation and cognition. In the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal gyrus, neurons and glial cells are produced that contribute to memory production. These areas form part of the limbic system [190,191]. Additional structures of the limbic system include the hippocampus, parahippocampal gyrus, and amygdala, which is located in the temporal lobe. One study reports worsening neurocognitive dysfunction when targeting the left temporal lobe with chemotherapy in children <7 years of age [192]. In adults, an additional study shows neurological sequelae can be spared when avoiding the hippocampi during radiotherapy; in children this has yet to be studied [193]. Finally, cerebellar neurons form closed loops with neurons in the prefrontal cortex, temporal lobes, and limbic structures. Damage to these neurons can lead to posterior fossa syndrome in medulloblastoma and neurocognitive dysfunction and should be avoided if possible [178].

3.3. Management of Late Effects

There remains a shortage of published studies that investigate interventions for survivors of pediatric brain tumors suffering from neurocognitive late effects. Of the available studies, interventions fall under three categories: cognitive remediation, pharmacotherapy, and environmental modifications [40]. A case study involving the use of a memory notebook to remediate severe memory impairment in a survivor found that the patient’s academic achievement, classroom attendance, and completion of assignments increased slightly, despite a continued, significant memory impairment [194]. Stimulants, such as methylphenidate, have been shown to be effective medications for attention deficit hyperactivity disorder (ADHD), through improvements in cognitive function [195]. A randomized, double-blind trial that investigated the effects of methylphenidate on survivors of both pediatric leukemia and malignant brain tumors with impaired learning found that the group assigned methylphenidate showed significantly greater improvements in tests of vigilance, a proxy for attention, compared to the placebo group [196]. However, another study that exclusively tested survivors of brain tumors who received craniospinal irradiation 3–12 years earlier found no significant immediate, or delayed, benefit from methylphenidate therapy [197]. Environmental interventions in children with neurocognitive delay is often underestimated in its efficacy [198]. The family dynamic survivors are recovering in have been shown to play a role in neurocognitive recovery from traumatic brain injury, with more dysfunctional environments being associated with slower recovery when severity of brain injury and other medical factors are controlled for [199]. While the neurodevelopmental consequences of brain irradiation in pediatric patients are not fully elucidated, the interventions indicated for treatment of these effects seem to be just as, if not more, poorly established.
The risk factors for late effects of neurotoxicity with PBRT are similar to photon-based therapy: being of a younger age as well as the volume of the brain and spine that were irradiated [200,201,202].

4. Future Directions

While advancements in photon-based radiotherapy, such as IMRT and VMAT, reduce neurotoxicity to normal brain tissue, PBRT has emerged as a particularly promising modality for minimizing late toxicity due to reductions in the dose to vital organs (Table 3) [41,203,204,205].
Macdonald et al. found that initial control of pediatric ependymoma and sparing of normal tissue was favorable with PBRT compared to IMRT, a form of photon radiotherapy [206]. In addition, Mirabell et al. found a reduced incidence of secondary cancer resulting from radiotherapy with PBRT in rhabdomyosarcoma and medulloblastoma patients [207]. Indeed, Zhang et al. calculate a lifetime risk of 0.18 for secondary cancer attributable to PBRT, much lower than that of photon-based radiotherapy [209]. Eaton et al. found no significant difference in recurrence free survival or overall survival in standard-risk medulloblastoma patients treated with proton versus photon-based radiotherapy, however the need for endocrine replacement was significantly lower in those receiving PBRT [58]. A retrospective study of children with medulloblastoma treated with photon versus PBRT showed no difference in clinical outcome between groups, despite the cochlea receiving a lower mean dose in the PBRT group. It was hypothesized that the rates of ototoxicity did not differ between groups due to confounding ototoxicity from cisplatin therapy.
The initial biological effect of PBRT is considered to have the same theoretical relative biological effect (RBE) of 1.1 as photon-based therapy [210,211,212]. It is hypothesized that reduced incidence and/or intensity of late effects can be observed with PBRT due to the intrinsic physics of its dose deposition [41,213,214,215,216]. Proton beams exhibit a sharp peak in proton deposition called the Bragg peak, which is targeted to cover to tumor volume (Figure 1). The deposition before the peak is suppressed and the deposition distal to the peak is near zero, effectively minimizing the radiotherapy dose to normal tissue within the beam trajectory [41,217,218,219,220]. PBRT has been shown to reduce the dose to the brain, brainstem, optic nerve, and optic chiasm [221,222,223]. Even the small, critical organs that are typically susceptible to radiotherapy toxicity, such as the cochlea and hypothalamus, can be spared with PBRT, which preserves hearing, intelligence, and endocrine function [41,200].
Evidence of PBRT’s benefit towards reduced neurocognitive toxicity has grown over the past two decades [200,215,224,225,226,227,228,229,230,231,232]. Studies that measured neurocognitive outcomes post PBRT therapy did not find any significant neurocognitive impairment in survivors [200,201,202,208]. Processing speed was identified as the domain of greatest vulnerability [208,228]. A study by Kahelley et al. comparing cognitive outcomes after photon versus proton radiotherapy showed no significant decline in IQ among survivors receiving PBRT, while those treated with photon radiotherapy did show a significant decline [208]. The risk factors for late effects of neurotoxicity with PBRT are similar to photon-based therapy: being of a younger age as well as the volume of the brain and spine that were irradiated [200,201,202].
With the advent of these novel radiotherapeutic techniques coupled with developments in advanced MRI techniques, these novel approaches need to be tested in prospective studies of children with brain tumors. Primary outcomes should be survival as well as long-term neurocognitive function. Additionally, these studies should locate the tumor, gather its proximity to vital eloquent areas and tracks of the brain, and monitor outcomes as it pertains to location. With data from these prospective studies, optimal therapeutic strategies can be created for each specific type of brain tumor.

5. Conclusions

The management of pediatric central-nervous-system tumors with radiotherapy allows for greater lifetime survival but also lengthens the morbidity from late effects, especially neurocognitive decline. This is largely due to the unintended consequence of radiation-induced damage to the normal neural tissue caught in the beam trajectory. Advancements in photon-based radiotherapy confined the volume of the brain that was irradiated to fit the tumor margins more closely; however, the advent of PBRT further allows for greater sparing of normal neuronal tissue, by maximizing energy deposition at the tumor location while minimizing energy deposition both proximal and distal to the tumor target. While additional advancements in radiotherapy technology are certainly warranted for both an increase in survival and a decrease in neurocognitive late effects, further research is needed to develop interventions for those already suffering from these late effects. Current intervention options are limited and of uninspiring efficacy, mostly limited to environmental accommodations and cognitive remediations. Interventions for generalized developmental delay may attenuate the consequences of these late effects, but there remains an opportunity to implement specific strategies to intervene with survivors treated with radiotherapy, especially due to the emergence of consensus deficits in neurocognition, such as processing speed.

Author Contributions

Conceptualization, J.L.G.; methodology, N.M.; validation, N.A.P.; formal analysis, E.N.; data curation, M.A., N.J.B. and D.P.; writing—original draft preparation, N.M. and J.B.; writing—review and editing, all authors; supervision, R.S. and J.L.; project administration, J.L.G. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by the United States Department of Defense.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Linabery, A.M.; Ross, J.A. Trends in childhood cancer incidence in the U.S. (1992–2004). Cancer 2008, 112, 416–432. [Google Scholar] [CrossRef] [PubMed]
  2. Centers for Disease Control and Prevention. Trends in childhood cancer mortality--United States, 1990–2004. MMWR Morb. Mortal Wkly. Rep. 2007, 56, 1257–1261. [Google Scholar]
  3. Pollack, I.F.; Agnihotri, S.; Broniscer, A. Childhood brain tumors: Current management, biological insights, and future directions. J. Neurosurg. Pediatr. 2019, 23, 261–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Duffner, P.K.; Cohen, M.E.; Myers, M.H.; Heise, H.W. Survival of children with brain tumors: SEER Program, 1973–1980. Neurology 1986, 36, 597–601. [Google Scholar] [CrossRef]
  5. Young, J.L., Jr.; Ries, L.G.; Silverberg, E.; Horm, J.W.; Miller, R.W. Cancer incidence, survival, and mortality for children younger than age 15 years. Cancer 1986, 58, 598–602. [Google Scholar] [CrossRef]
  6. Massimino, M.; Giangaspero, F.; Garrè, M.L.; Gandola, L.; Poggi, G.; Biassoni, V.; Gatta, G.; Rutkowski, S. Childhood medulloblastoma. Crit. Rev. Oncol. Hematol. 2011, 79, 65–83. [Google Scholar] [CrossRef] [Green Version]
  7. Hoffman, H.J.; De Silva, M.; Humphreys, R.P.; Drake, J.M.; Smith, M.L.; Blaser, S.I. Aggressive surgical management of craniopharyngiomas in children. J. Neurosurg. 1992, 76, 47–52. [Google Scholar] [CrossRef]
  8. Rousseau, P.; Habrand, J.L.; Sarrazin, D.; Kalifa, C.; Terrier-Lacombe, M.J.; Rekacewicz, C.; Rey, A. Treatment of intracranial ependymomas of children: Review of a 15-year experience. Int. J. Radiat. Oncol. Biol. Phys. 1994, 28, 381–386. [Google Scholar] [CrossRef]
  9. Wisoff, J.H.; Sanford, R.A.; Heier, L.A.; Sposto, R.; Burger, P.C.; Yates, A.J.; Holmes, E.J.; Kun, L.E. Primary neurosurgery for pediatric low-grade gliomas: A prospective multi-institutional study from the Children’s Oncology Group. Neurosurgery 2011, 68, 1548–1554. [Google Scholar] [CrossRef]
  10. Wisoff, J.H.; Boyett, J.M.; Berger, M.S.; Brant, C.; Li, H.; Yates, A.J.; McGuire-Cullen, P.; Turski, P.A.; Sutton, L.N.; Allen, J.C.; et al. Current neurosurgical management and the impact of the extent of resection in the treatment of malignant gliomas of childhood: A report of the Children’s Cancer Group trial no. CCG-945. J. Neurosurg. 1998, 89, 52–59. [Google Scholar] [CrossRef]
  11. Shibui, S. Brain Tumor Registry of Japan (2001–2004). Neurol. Med. Chir. 2014, 54, 1–102. [Google Scholar]
  12. Howlander, N.; Noone, A.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.; et al. SEER Cancer Statistics Review, 1975–2016. Available online: https://seer.cancer.gov/archive/csr/1975_2016/ (accessed on 21 February 2021).
  13. Armstrong, G.T.; Stovall, M.; Robison, L.L. Long-term effects of radiation exposure among adult survivors of childhood cancer: Results from the childhood cancer survivor study. Radiat. Res. 2010, 174, 840–850. [Google Scholar] [CrossRef] [Green Version]
  14. Ris, M.D.; Packer, R.; Goldwein, J.; Jones-Wallace, D.; Boyett, J.M. Intellectual outcome after reduced-dose radiation therapy plus adjuvant chemotherapy for medulloblastoma: A Children’s Cancer Group study. J. Clin. Oncol. 2001, 19, 3470–3476. [Google Scholar] [CrossRef]
  15. Mulhern, R.K.; Palmer, S.L.; Merchant, T.E.; Wallace, D.; Kocak, M.; Brouwers, P.; Krull, K.; Chintagumpala, M.; Stargatt, R.; Ashley, D.M.; et al. Neurocognitive consequences of risk-adapted therapy for childhood medulloblastoma. J. Clin. Oncol. 2005, 23, 5511–5519. [Google Scholar] [CrossRef]
  16. Hua, C.; Bass, J.K.; Khan, R.; Kun, L.E.; Merchant, T.E. Hearing loss after radiotherapy for pediatric brain tumors: Effect of cochlear dose. Int. J. Radiat. Oncol. Biol. Phys. 2008, 72, 892–899. [Google Scholar] [CrossRef]
  17. Laughton, S.J.; Merchant, T.E.; Sklar, C.A.; Kun, L.E.; Fouladi, M.; Broniscer, A.; Morris, E.B.; Sanders, R.P.; Krasin, M.J.; Shelso, J.; et al. Endocrine outcomes for children with embryonal brain tumors after risk-adapted craniospinal and conformal primary-site irradiation and high-dose chemotherapy with stem-cell rescue on the SJMB-96 trial. J. Clin. Oncol. 2008, 26, 1112–1118. [Google Scholar] [CrossRef]
  18. Neglia, J.P.; Robison, L.L.; Stovall, M.; Liu, Y.; Packer, R.J.; Hammond, S.; Yasui, Y.; Kasper, C.E.; Mertens, A.C.; Donaldson, S.S.; et al. New primary neoplasms of the central nervous system in survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. J. Natl. Cancer Inst. 2006, 98, 1528–1537. [Google Scholar] [CrossRef]
  19. Miké, V.; Meadows, A.T.; D’Angio, G.J. Incidence of second malignant neoplasms in children: Results of an international study. Lancet 1982, 2, 1326–1331. [Google Scholar] [CrossRef]
  20. Murphy, E.S.; Merchant, T.E.; Wu, S.; Xiong, X.; Lukose, R.; Wright, K.D.; Qaddoumi, I.; Armstrong, G.T.; Broniscer, A.; Gajjar, A. Necrosis after craniospinal irradiation: Results from a prospective series of children with central nervous system embryonal tumors. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, e655–e660. [Google Scholar] [CrossRef] [Green Version]
  21. Bloom, H.J.; Wallace, E.N.; Henk, J.M. The treatment and prognosis of medulloblastoma in children. A study of 82 verified cases. Am. J. Roentgenol. Radium Ther. Nucl. Med. 1969, 105, 43–62. [Google Scholar] [CrossRef]
  22. Meeske, K.A.; Patel, S.K.; Palmer, S.N.; Nelson, M.B.; Parow, A.M. Factors associated with health-related quality of life in pediatric cancer survivors. Pediatr. Blood Cancer 2007, 49, 298–305. [Google Scholar] [CrossRef]
  23. Packer, R.J. Childhood brain tumors: Accomplishments and ongoing challenges. J. Child Neurol. 2008, 23, 1122–1127. [Google Scholar] [CrossRef] [Green Version]
  24. Meeske, K.; Katz, E.R.; Palmer, S.N.; Burwinkle, T.; Varni, J.W. Parent proxy-reported health-related quality of life and fatigue in pediatric patients diagnosed with brain tumors and acute lymphoblastic leukemia. Cancer 2004, 101, 2116–2125. [Google Scholar] [CrossRef]
  25. Brice, L.; Weiss, R.; Wei, Y.; Satwani, P.; Bhatia, M.; George, D.; Garvin, J.; Morris, E.; Harrison, L.; Cairo, M.S.; et al. Health-related quality of life (HRQoL): The impact of medical and demographic variables upon pediatric recipients of hematopoietic stem cell transplantation. Pediatr. Blood Cancer 2011, 57, 1179–1185. [Google Scholar] [CrossRef]
  26. Johnson, D.L.; McCabe, M.A.; Nicholson, H.S.; Joseph, A.L.; Getson, P.R.; Byrne, J.; Brasseux, C.; Packer, R.J.; Reaman, G. Quality of long-term survival in young children with medulloblastoma. J. Neurosurg. 1994, 80, 1004–1010. [Google Scholar] [CrossRef] [Green Version]
  27. Reimers, T.S.; Mortensen, E.L.; Nysom, K.; Schmiegelow, K. Health-related quality of life in long-term survivors of childhood brain tumors. Pediatr. Blood Cancer 2009, 53, 1086–1091. [Google Scholar] [CrossRef] [PubMed]
  28. Ajithkumar, T.; Price, S.; Horan, G.; Burke, A.; Jefferies, S. Prevention of radiotherapy-induced neurocognitive dysfunction in survivors of paediatric brain tumours: The potential role of modern imaging and radiotherapy techniques. Lancet Oncol. 2017, 18, e91–e100. [Google Scholar] [CrossRef] [Green Version]
  29. Ris, M.D.; Noll, R.B. Long-term neurobehavioral outcome in pediatric brain-tumor patients: Review and methodological critique. J. Clin. Exp. Neuropsychol. 1994, 16, 21–42. [Google Scholar] [CrossRef] [PubMed]
  30. Mulhern, R.K.; Hancock, J.; Fairclough, D.; Kun, L. Neuropsychological status of children treated for brain tumors: A critical review and integrative analysis. Med. Pediatr. Oncol. 1992, 20, 181–191. [Google Scholar] [CrossRef] [PubMed]
  31. Chadderton, R.D.; West, C.G.; Schuller, S.; Quirke, D.C.; Gattamaneni, R.; Taylor, R. Radiotherapy in the treatment of low-grade astrocytomas. II. The physical and cognitive sequelae. Childs Nerv. Syst. 1995, 11, 443–448. [Google Scholar] [CrossRef] [PubMed]
  32. Dennis, M.; Spiegler, B.J.; Obonsawin, M.C.; Maria, B.L.; Cowell, C.; Hoffman, H.J.; Hendrick, E.B.; Humphreys, R.P.; Bailey, J.D.; Ehrlich, R.M. Brain tumors in children and adolescents--III. Effects of radiation and hormone status on intelligence and on working, associative and serial-order memory. Neuropsychologia 1992, 30, 257–275. [Google Scholar] [CrossRef]
  33. North, C.A.; North, R.B.; Epstein, J.A.; Piantadosi, S.; Wharam, M.D. Low-grade cerebral astrocytomas. Survival and quality of life after radiation therapy. Cancer 1990, 66, 6–14. [Google Scholar] [CrossRef]
  34. Palmer, S.L.; Goloubeva, O.; Reddick, W.E.; Glass, J.O.; Gajjar, A.; Kun, L.; Merchant, T.E.; Mulhern, R.K. Patterns of intellectual development among survivors of pediatric medulloblastoma: A longitudinal analysis. J. Clin. Oncol. 2001, 19, 2302–2308. [Google Scholar] [CrossRef]
  35. Walter, A.W.; Mulhern, R.K.; Gajjar, A.; Heideman, R.L.; Reardon, D.; Sanford, R.A.; Xiong, X.; Kun, L.E. Survival and neurodevelopmental outcome of young children with medulloblastoma at St Jude Children’s Research Hospital. J. Clin. Oncol. 1999, 17, 3720–3728. [Google Scholar] [CrossRef]
  36. Reimers, T.S.; Ehrenfels, S.; Mortensen, E.L.; Schmiegelow, M.; Sønderkaer, S.; Carstensen, H.; Schmiegelow, K.; Müller, J. Cognitive deficits in long-term survivors of childhood brain tumors: Identification of predictive factors. Med. Pediatr. Oncol. 2003, 40, 26–34. [Google Scholar] [CrossRef]
  37. Mostow, E.N.; Byrne, J.; Connelly, R.R.; Mulvihill, J.J. Quality of life in long-term survivors of CNS tumors of childhood and adolescence. J. Clin. Oncol. 1991, 9, 592–599. [Google Scholar] [CrossRef]
  38. Howard, A.F.; Hasan, H.; Bobinski, M.A.; Nurcombe, W.; Olson, R.; Parkinson, M.; Goddard, K. Parents’ perspectives of life challenges experienced by long-term paediatric brain tumour survivors: Work and finances, daily and social functioning, and legal difficulties. J. Cancer Surviv. 2014, 8, 372–383. [Google Scholar] [CrossRef]
  39. De Ruiter, M.A.; Schouten-van Meeteren, A.Y.N.; van Vuurden, D.G.; Maurice-Stam, H.; Gidding, C.; Beek, L.R.; Granzen, B.; Oosterlaan, J.; Grootenhuis, M.A. Psychosocial profile of pediatric brain tumor survivors with neurocognitive complaints. Qual. Life Res. 2016, 25, 435–446. [Google Scholar] [CrossRef] [Green Version]
  40. Mulhern, R.K.; Merchant, T.E.; Gajjar, A.; Reddick, W.E.; Kun, L.E. Late neurocognitive sequelae in survivors of brain tumours in childhood. Lancet Oncol. 2004, 5, 399–408. [Google Scholar] [CrossRef]
  41. Merchant, T.E.; Hua, C.H.; Shukla, H.; Ying, X.; Nill, S.; Oelfke, U. Proton versus photon radiotherapy for common pediatric brain tumors: Comparison of models of dose characteristics and their relationship to cognitive function. Pediatr. Blood Cancer 2008, 51, 110–117. [Google Scholar] [CrossRef]
  42. Merchant, T.E.; Kiehna, E.N.; Li, C.; Shukla, H.; Sengupta, S.; Xiong, X.; Gajjar, A.; Mulhern, R.K. Modeling radiation dosimetry to predict cognitive outcomes in pediatric patients with CNS embryonal tumors including medulloblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2006, 65, 210–221. [Google Scholar] [CrossRef]
  43. Grill, J.; Renaux, V.K.; Bulteau, C.; Viguier, D.; Levy-Piebois, C.; Sainte-Rose, C.; Dellatolas, G.; Raquin, M.A.; Jambaqué, I.; Kalifa, C. Long-term intellectual outcome in children with posterior fossa tumors according to radiation doses and volumes. Int. J. Radiat. Oncol. Biol. Phys. 1999, 45, 137–145. [Google Scholar] [CrossRef]
  44. Dennis, M.; Spiegler, B.J.; Hetherington, C.R.; Greenberg, M.L. Neuropsychological sequelae of the treatment of children with medulloblastoma. J. Neurooncol. 1996, 29, 91–101. [Google Scholar] [CrossRef]
  45. Nathan, P.C.; Patel, S.K.; Dilley, K.; Goldsby, R.; Harvey, J.; Jacobsen, C.; Kadan-Lottick, N.; McKinley, K.; Millham, A.K.; Moore, I.; et al. Guidelines for identification of, advocacy for, and intervention in neurocognitive problems in survivors of childhood cancer: A report from the Children’s Oncology Group. Arch. Pediatr. Adolesc. Med. 2007, 161, 798–806. [Google Scholar] [CrossRef]
  46. Ullrich, N.J.; Embry, L. Neurocognitive dysfunction in survivors of childhood brain tumors. Semin. Pediatr. Neurol. 2012, 19, 35–42. [Google Scholar] [CrossRef] [PubMed]
  47. Wefel, J.S.; Noll, K.R.; Scheurer, M.E. Neurocognitive functioning and genetic variation in patients with primary brain tumours. Lancet Oncol. 2016, 17, e97–e108. [Google Scholar] [CrossRef] [Green Version]
  48. DeNunzio, N.J.; Yock, T.I. Modern Radiotherapy for Pediatric Brain Tumors. Cancers 2020, 12, 1533. [Google Scholar] [CrossRef] [PubMed]
  49. McNeil, D.E.; Coté, T.R.; Clegg, L.; Rorke, L.B. Incidence and trends in pediatric malignancies medulloblastoma/primitive neuroectodermal tumor: A SEER update. Surveillance Epidemiology and End Results. Med. Pediatr. Oncol. 2002, 39, 190–194. [Google Scholar] [CrossRef]
  50. Park, T.S.; Hoffman, H.J.; Hendrick, E.B.; Humphreys, R.P.; Becker, L.E. Medulloblastoma: Clinical presentation and management. Experience at the hospital for sick children, toronto, 1950–1980. J. Neurosurg. 1983, 58, 543–552. [Google Scholar] [CrossRef]
  51. Padovani, L.; Horan, G.; Ajithkumar, T. Radiotherapy Advances in Paediatric Medulloblastoma Treatment. Clin. Oncol. 2019, 31, 171–181. [Google Scholar] [CrossRef]
  52. Kortmann, R.D.; Kühl, J.; Timmermann, B.; Mittler, U.; Urban, C.; Budach, V.; Richter, E.; Willich, N.; Flentje, M.; Berthold, F.; et al. Postoperative neoadjuvant chemotherapy before radiotherapy as compared to immediate radiotherapy followed by maintenance chemotherapy in the treatment of medulloblastoma in childhood: Results of the German prospective randomized trial HIT 91. Int. J. Radiat. Oncol. Biol. Phys. 2000, 46, 269–279. [Google Scholar] [CrossRef]
  53. Fouladi, M.; Gajjar, A.; Boyett, J.M.; Walter, A.W.; Thompson, S.J.; Merchant, T.E.; Jenkins, J.J.; Langston, J.W.; Liu, A.; Kun, L.E.; et al. Comparison of CSF cytology and spinal magnetic resonance imaging in the detection of leptomeningeal disease in pediatric medulloblastoma or primitive neuroectodermal tumor. J. Clin. Oncol. 1999, 17, 3234–3237. [Google Scholar] [CrossRef]
  54. Bourgouin, P.M.; Tampieri, D.; Grahovac, S.Z.; Léger, C.; Del Carpio, R.; Melançon, D. CT and MR imaging findings in adults with cerebellar medulloblastoma: Comparison with findings in children. Am. J. Roentgenol. 1992, 159, 609–612. [Google Scholar] [CrossRef] [Green Version]
  55. Poretti, A.; Meoded, A.; Huisman, T.A. Neuroimaging of pediatric posterior fossa tumors including review of the literature. J. Magn. Reson. Imaging 2012, 35, 32–47. [Google Scholar] [CrossRef]
  56. Al-Wassia, R.K.; Ghassal, N.M.; Naga, A.; Awad, N.A.; Bahadur, Y.A.; Constantinescu, C. Optimization of Craniospinal Irradiation for Pediatric Medulloblastoma Using VMAT and IMRT. J. Pediatr. Hematol. Oncol. 2015, 37, e405–e411. [Google Scholar] [CrossRef]
  57. Brown, A.P.; Barney, C.L.; Grosshans, D.R.; McAleer, M.F.; de Groot, J.F.; Puduvalli, V.K.; Tucker, S.L.; Crawford, C.N.; Khan, M.; Khatua, S.; et al. Proton beam craniospinal irradiation reduces acute toxicity for adults with medulloblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 277–284. [Google Scholar] [CrossRef] [Green Version]
  58. Eaton, B.R.; Esiashvili, N.; Kim, S.; Patterson, B.; Weyman, E.A.; Thornton, L.T.; Mazewski, C.; MacDonald, T.J.; Ebb, D.; MacDonald, S.M.; et al. Endocrine outcomes with proton and photon radiotherapy for standard risk medulloblastoma. Neuro. Oncol. 2016, 18, 881–887. [Google Scholar] [CrossRef] [Green Version]
  59. Packer, R.J.; Gajjar, A.; Vezina, G.; Rorke-Adams, L.; Burger, P.C.; Robertson, P.L.; Bayer, L.; LaFond, D.; Donahue, B.R.; Marymont, M.H.; et al. Phase III study of craniospinal radiation therapy followed by adjuvant chemotherapy for newly diagnosed average-risk medulloblastoma. J. Clin. Oncol. 2006, 24, 4202–4208. [Google Scholar] [CrossRef]
  60. Merchant, T.E.; Kun, L.E.; Krasin, M.J.; Wallace, D.; Chintagumpala, M.M.; Woo, S.Y.; Ashley, D.M.; Sexton, M.; Kellie, S.J.; Ahern, V.; et al. Multi-institution prospective trial of reduced-dose craniospinal irradiation (23.4 Gy) followed by conformal posterior fossa (36 Gy) and primary site irradiation (55.8 Gy) and dose-intensive chemotherapy for average-risk medulloblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2008, 70, 782–787. [Google Scholar] [CrossRef] [Green Version]
  61. Lannering, B.; Rutkowski, S.; Doz, F.; Pizer, B.; Gustafsson, G.; Navajas, A.; Massimino, M.; Reddingius, R.; Benesch, M.; Carrie, C.; et al. Hyperfractionated versus conventional radiotherapy followed by chemotherapy in standard-risk medulloblastoma: Results from the randomized multicenter HIT-SIOP PNET 4 trial. J. Clin. Oncol. 2012, 30, 3187–3193. [Google Scholar] [CrossRef]
  62. Eaton, B.R.; Esiashvili, N.; Kim, S.; Weyman, E.A.; Thornton, L.T.; Mazewski, C.; MacDonald, T.; Ebb, D.; MacDonald, S.M.; Tarbell, N.J.; et al. Clinical Outcomes Among Children With Standard-Risk Medulloblastoma Treated With Proton and Photon Radiation Therapy: A Comparison of Disease Control and Overall Survival. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 133–138. [Google Scholar] [CrossRef] [Green Version]
  63. Focal Radiotherapy Plus Low Dose Craniospinal Irradiation Followed by Adjuvant Chemotherapy in WNT Medulloblastoma. Available online: https://clinicaltrials.gov/ct2/show/NCT04474964 (accessed on 6 June 2022).
  64. Cohen, K.J.; Broniscer, A.; Glod, J. Pediatric glial tumors. Curr. Treat Options Oncol. 2001, 2, 529–536. [Google Scholar] [CrossRef]
  65. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  66. Teo, C.; Siu, T.L. Radical resection of focal brainstem gliomas: Is it worth doing? Childs Nerv. Syst. 2008, 24, 1307–1314. [Google Scholar] [CrossRef]
  67. Shibamoto, Y.; Takahashi, M.; Dokoh, S.; Tanabe, M.; Ishida, T.; Abe, M. Radiation therapy for brain stem tumor with special reference to CT feature and prognosis correlations. Int. J. Radiat. Oncol. Biol. Phys. 1989, 17, 71–76. [Google Scholar] [CrossRef]
  68. Schild, S.E.; Stafford, S.L.; Brown, P.D.; Wood, C.P.; Scheithauer, B.W.; Schomberg, P.J.; Wong, W.W.; Lyons, M.K.; Shaw, E.G. The results of radiotherapy for brainstem tumors. J. Neurooncol. 1998, 40, 171–177. [Google Scholar] [CrossRef]
  69. Schulz-Ertner, D.; Debus, J.; Lohr, F.; Frank, C.; Höss, A.; Wannenmacher, M. Fractionated stereotactic conformal radiation therapy of brain stem gliomas: Outcome and prognostic factors. Radiother. Oncol. 2000, 57, 215–223. [Google Scholar] [CrossRef]
  70. Merchant, T.E.; Mulhern, R.K.; Krasin, M.J.; Kun, L.E.; Williams, T.; Li, C.; Xiong, X.; Khan, R.B.; Lustig, R.H.; Boop, F.A.; et al. Preliminary results from a phase II trial of conformal radiation therapy and evaluation of radiation-related CNS effects for pediatric patients with localized ependymoma. J. Clin. Oncol. 2004, 22, 3156–3162. [Google Scholar] [CrossRef]
  71. Landau, E.; Boop, F.A.; Conklin, H.M.; Wu, S.; Xiong, X.; Merchant, T.E. Supratentorial ependymoma: Disease control, complications, and functional outcomes after irradiation. Int. J. Radiat. Oncol. Biol. Phys. 2013, 85, e193–e199. [Google Scholar] [CrossRef] [Green Version]
  72. Conter, C.; Carrie, C.; Bernier, V.; Geoffray, A.; Pagnier, A.; Gentet, J.C.; Lellouch-Tubiana, A.; Chabaud, S.; Frappaz, D. Intracranial ependymomas in children: Society of pediatric oncology experience with postoperative hyperfractionated local radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2009, 74, 1536–1542. [Google Scholar] [CrossRef]
  73. Lober, R.M.; Harsh, G.R.t. A perspective on craniopharyngioma. World Neurosurg. 2013, 79, 645–646. [Google Scholar] [CrossRef] [PubMed]
  74. Ellison, D.; Clifford, S.C.; Gajjar, A.; GIlbertson, R. What’s new in neuro-oncology? Recent advances in medulloblastoma. Eur. J. Paediatr. Neurol. 1991, 7, 53–66. [Google Scholar] [CrossRef]
  75. Thomas, P.R.; Deutsch, M.; Kepner, J.L.; Boyett, J.M.; Krischer, J.; Aronin, P.; Albright, L.; Allen, J.C.; Packer, R.J.; Linggood, R.; et al. Low-stage medulloblastoma: Final analysis of trial comparing standard-dose with reduced-dose neuraxis irradiation. J. Clin. Oncol. 2000, 18, 3004–3011. [Google Scholar] [CrossRef] [PubMed]
  76. Packer, R.J.; Sutton, L.N.; Atkins, T.E.; Radcliffe, J.; Bunin, G.R.; D’Angio, G.; Siegel, K.R.; Schut, L. A prospective study of cognitive function in children receiving whole-brain radiotherapy and chemotherapy: 2-year results. J. Neurosurg. 1989, 70, 707–713. [Google Scholar] [CrossRef]
  77. Packer, R.J.; Boyett, J.M.; Janss, A.J.; Stavrou, T.; Kun, L.; Wisoff, J.; Russo, C.; Geyer, R.; Phillips, P.; Kieran, M.; et al. Growth hormone replacement therapy in children with medulloblastoma: Use and effect on tumor control. J. Clin. Oncol. 2001, 19, 480–487. [Google Scholar] [CrossRef]
  78. Bailey, C.C.; Gnekow, A.; Wellek, S.; Jones, M.; Round, C.; Brown, J.; Phillips, A.; Neidhardt, M.K. Prospective randomised trial of chemotherapy given before radiotherapy in childhood medulloblastoma. International Society of Paediatric Oncology (SIOP) and the (German) Society of Paediatric Oncology (GPO): SIOP II. Med. Pediatr. Oncol. 1995, 25, 166–178. [Google Scholar] [CrossRef]
  79. Murphy, E.S.; Chao, S.T.; Angelov, L.; Vogelbaum, M.A.; Barnett, G.; Jung, E.; Recinos, V.R.; Mohammadi, A.; Suh, J.H. Radiosurgery for Pediatric Brain Tumors. Pediatr. Blood Cancer 2016, 63, 398–405. [Google Scholar] [CrossRef]
  80. Evans, A.E.; Jenkin, R.D.; Sposto, R.; Ortega, J.A.; Wilson, C.B.; Wara, W.; Ertel, I.J.; Kramer, S.; Chang, C.H.; Leikin, S.L.; et al. The treatment of medulloblastoma. Results of a prospective randomized trial of radiation therapy with and without CCNU, vincristine, and prednisone. J. Neurosurg. 1990, 72, 572–582. [Google Scholar] [CrossRef]
  81. Tait, D.M.; Thornton-Jones, H.; Bloom, H.J.; Lemerle, J.; Morris-Jones, P. Adjuvant chemotherapy for medulloblastoma: The first multi-centre control trial of the International Society of Paediatric Oncology (SIOP I). Eur. J. Cancer 1990, 26, 464–469. [Google Scholar] [CrossRef]
  82. Massimino, M.; Biassoni, V.; Gandola, L.; Garrè, M.L.; Gatta, G.; Giangaspero, F.; Poggi, G.; Rutkowski, S. Childhood medulloblastoma. Crit. Rev. Oncol. Hematol. 2016, 105, 35–51. [Google Scholar] [CrossRef] [Green Version]
  83. Rutkowski, S.; Bode, U.; Deinlein, F.; Ottensmeier, H.; Warmuth-Metz, M.; Soerensen, N.; Graf, N.; Emser, A.; Pietsch, T.; Wolff, J.E.; et al. Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. N. Engl. J. Med. 2005, 352, 978–986. [Google Scholar] [CrossRef] [Green Version]
  84. Lassaletta, A. Medulloblastoma in infants: The never-ending challenge. Lancet Oncol. 2018, 19, 720–721. [Google Scholar] [CrossRef]
  85. Remke, M.; Ramaswamy, V. Infant medulloblastoma-Learning new lessons from old strata. Nat. Rev. Clin. Oncol. 2018, 15, 659–660. [Google Scholar] [CrossRef]
  86. Gajjar, A.; Chintagumpala, M.; Ashley, D.; Kellie, S.; Kun, L.E.; Merchant, T.E.; Woo, S.; Wheeler, G.; Ahern, V.; Krasin, M.J.; et al. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): Long-term results from a prospective, multicentre trial. Lancet Oncol. 2006, 7, 813–820. [Google Scholar] [CrossRef]
  87. Packer, R.J. Chemotherapy for medulloblastoma/primitive neuroectodermal tumors of the posterior fossa. Ann. Neurol. 1990, 28, 823–828. [Google Scholar] [CrossRef]
  88. Zeltzer, P.M.; Boyett, J.M.; Finlay, J.L.; Albright, A.L.; Rorke, L.B.; Milstein, J.M.; Allen, J.C.; Stevens, K.R.; Stanley, P.; Li, H.; et al. Metastasis stage, adjuvant treatment, and residual tumor are prognostic factors for medulloblastoma in children: Conclusions from the Children’s Cancer Group 921 randomized phase III study. J. Clin. Oncol. 1999, 17, 832–845. [Google Scholar] [CrossRef] [Green Version]
  89. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro-Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  90. Rutkowski, S.; von Hoff, K.; Emser, A.; Zwiener, I.; Pietsch, T.; Figarella-Branger, D.; Giangaspero, F.; Ellison, D.W.; Garre, M.L.; Biassoni, V.; et al. Survival and prognostic factors of early childhood medulloblastoma: An international meta-analysis. J. Clin. Oncol. 2010, 28, 4961–4968. [Google Scholar] [CrossRef]
  91. Northcott, P.A.; Jones, D.T.; Kool, M.; Robinson, G.W.; Gilbertson, R.J.; Cho, Y.J.; Pomeroy, S.L.; Korshunov, A.; Lichter, P.; Taylor, M.D.; et al. Medulloblastomics: The end of the beginning. Nat. Rev. Cancer 2012, 12, 818–834. [Google Scholar] [CrossRef] [Green Version]
  92. Thompson, E.M.; Bramall, A.; Herndon, J.E., 2nd; Taylor, M.D.; Ramaswamy, V. The clinical importance of medulloblastoma extent of resection: A systematic review. J. Neurooncol. 2018, 139, 523–539. [Google Scholar] [CrossRef]
  93. Raimondi, A.J.; Tomita, T. Medulloblastoma in childhood. Acta Neurochir. 1979, 50, 127–138. [Google Scholar] [CrossRef]
  94. Berry, M.P.; Jenkin, R.D.; Keen, C.W.; Nair, B.D.; Simpson, W.J. Radiation treatment for medulloblastoma. A 21-year review. J. Neurosurg. 1981, 55, 43–51. [Google Scholar] [CrossRef]
  95. Del Charco, J.O.; Bolek, T.W.; McCollough, W.M.; Maria, B.L.; Kedar, A.; Braylan, R.C.; Mickle, J.P.; Buatti, J.M.; Mendenhall, N.P.; Marcus, R.B., Jr. Medulloblastoma: Time-dose relationship based on a 30-year review. Int. J. Radiat. Oncol. Biol. Phys. 1998, 42, 147–154. [Google Scholar] [CrossRef]
  96. Taylor, R.E.; Bailey, C.C.; Robinson, K.; Weston, C.L.; Ellison, D.; Ironside, J.; Lucraft, H.; Gilbertson, R.; Tait, D.M.; Walker, D.A.; et al. Results of a randomized study of preradiation chemotherapy versus radiotherapy alone for nonmetastatic medulloblastoma: The International Society of Paediatric Oncology/United Kingdom Children’s Cancer Study Group PNET-3 Study. J. Clin. Oncol. 2003, 21, 1581–1591. [Google Scholar] [CrossRef]
  97. Thompson, E.M.; Hielscher, T.; Bouffet, E.; Remke, M.; Luu, B.; Gururangan, S.; McLendon, R.E.; Bigner, D.D.; Lipp, E.S.; Perreault, S.; et al. Prognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: A retrospective integrated clinical and molecular analysis. Lancet Oncol. 2016, 17, 484–495. [Google Scholar] [CrossRef] [Green Version]
  98. Yuh, G.E.; Loredo, L.N.; Yonemoto, L.T.; Bush, D.A.; Shahnazi, K.; Preston, W.; Slater, J.M.; Slater, J.D. Reducing toxicity from craniospinal irradiation: Using proton beams to treat medulloblastoma in young children. Cancer J. 2004, 10, 386–390. [Google Scholar] [CrossRef]
  99. St. Clair, W.H.; Adams, J.A.; Bues, M.; Fullerton, B.C.; La Shell, S.; Kooy, H.M.; Loeffler, J.S.; Tarbell, N.J. Advantage of protons compared to conventional X-ray or IMRT in the treatment of a pediatric patient with medulloblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 727–734. [Google Scholar] [CrossRef]
  100. Moeller, B.J.; Chintagumpala, M.; Philip, J.J.; Grosshans, D.R.; McAleer, M.F.; Woo, S.Y.; Gidley, P.W.; Vats, T.S.; Mahajan, A. Low early ototoxicity rates for pediatric medulloblastoma patients treated with proton radiotherapy. Radiat. Oncol. 2011, 6, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Fukunaga-Johnson, N.; Lee, J.H.; Sandler, H.M.; Robertson, P.; McNeil, E.; Goldwein, J.W. Patterns of failure following treatment for medulloblastoma: Is it necessary to treat the entire posterior fossa? Int. J. Radiat. Oncol. Biol. Phys. 1998, 42, 143–146. [Google Scholar] [CrossRef]
  102. Wolden, S.L.; Dunkel, I.J.; Souweidane, M.M.; Happersett, L.; Khakoo, Y.; Schupak, K.; Lyden, D.; Leibel, S.A. Patterns of failure using a conformal radiation therapy tumor bed boost for medulloblastoma. J. Clin. Oncol. 2003, 21, 3079–3083. [Google Scholar] [CrossRef]
  103. Packer, R.J.; Zhou, T.; Holmes, E.; Vezina, G.; Gajjar, A. Survival and secondary tumors in children with medulloblastoma receiving radiotherapy and adjuvant chemotherapy: Results of Children’s Oncology Group trial A9961. Neuro. Oncol. 2013, 15, 97–103. [Google Scholar] [CrossRef] [Green Version]
  104. Nageswara Rao, A.A.; Wallace, D.J.; Billups, C.; Boyett, J.M.; Gajjar, A.; Packer, R.J. Cumulative cisplatin dose is not associated with event-free or overall survival in children with newly diagnosed average-risk medulloblastoma treated with cisplatin based adjuvant chemotherapy: Report from the Children’s Oncology Group. Pediatr. Blood Cancer 2014, 61, 102–106. [Google Scholar] [CrossRef] [Green Version]
  105. Jakacki, R.I.; Burger, P.C.; Zhou, T.; Holmes, E.J.; Kocak, M.; Onar, A.; Goldwein, J.; Mehta, M.; Packer, R.J.; Tarbell, N.; et al. Outcome of children with metastatic medulloblastoma treated with carboplatin during craniospinal radiotherapy: A Children’s Oncology Group Phase I/II study. J. Clin. Oncol. 2012, 30, 2648–2653. [Google Scholar] [CrossRef] [Green Version]
  106. Gandola, L.; Massimino, M.; Cefalo, G.; Solero, C.; Spreafico, F.; Pecori, E.; Riva, D.; Collini, P.; Pignoli, E.; Giangaspero, F.; et al. Hyperfractionated accelerated radiotherapy in the Milan strategy for metastatic medulloblastoma. J. Clin. Oncol. 2009, 27, 566–571. [Google Scholar] [CrossRef]
  107. Gajjar, A.; Stewart, C.F.; Ellison, D.W.; Kaste, S.; Kun, L.E.; Packer, R.J.; Goldman, S.; Chintagumpala, M.; Wallace, D.; Takebe, N.; et al. Phase I study of vismodegib in children with recurrent or refractory medulloblastoma: A pediatric brain tumor consortium study. Clin. Cancer Res. 2013, 19, 6305–6312. [Google Scholar] [CrossRef] [Green Version]
  108. Robinson, G.W.; Orr, B.A.; Wu, G.; Gururangan, S.; Lin, T.; Qaddoumi, I.; Packer, R.J.; Goldman, S.; Prados, M.D.; Desjardins, A.; et al. Vismodegib Exerts Targeted Efficacy Against Recurrent Sonic Hedgehog-Subgroup Medulloblastoma: Results From Phase II Pediatric Brain Tumor Consortium Studies PBTC-025B and PBTC-032. J. Clin. Oncol. 2015, 33, 2646–2654. [Google Scholar] [CrossRef]
  109. Kumar, V.; Abbas, A.; Aster, J.C. Central Nervous System. In Robbins Basic Pathology, 10th ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 849–888. [Google Scholar]
  110. Mizumoto, M.; Oshiro, Y.; Yamamoto, T.; Kohzuki, H.; Sakurai, H. Proton Beam Therapy for Pediatric Brain Tumor. Neurol. Med. Chir. 2017, 57, 343–355. [Google Scholar] [CrossRef] [Green Version]
  111. Gajjar, A.; Sanford, R.A.; Heideman, R.; Jenkins, J.J.; Walter, A.; Li, Y.; Langston, J.W.; Muhlbauer, M.; Boyett, J.M.; Kun, L.E. Low-grade astrocytoma: A decade of experience at St. Jude Children’s Research Hospital. J. Clin. Oncol. 1997, 15, 2792–2799. [Google Scholar] [CrossRef]
  112. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2012–2016. Neuro-Oncol. 2019, 21, v1–v100. [Google Scholar] [CrossRef]
  113. Komotar, R.J.; Mocco, J.; Jones, J.E.; Zacharia, B.E.; Tihan, T.; Feldstein, N.A.; Anderson, R.C. Pilomyxoid astrocytoma: Diagnosis, prognosis, and management. Neurosurg. Focus 2005, 18, E7. [Google Scholar] [CrossRef]
  114. Komotar, R.J.; Burger, P.C.; Carson, B.S.; Brem, H.; Olivi, A.; Goldthwaite, P.T.; Tihan, T. Pilocytic and pilomyxoid hypothalamic/chiasmatic astrocytomas. Neurosurgery 2004, 54, 72–79. [Google Scholar] [CrossRef]
  115. Song, K.S.; Phi, J.H.; Cho, B.K.; Wang, K.C.; Lee, J.Y.; Kim, D.G.; Kim, I.H.; Ahn, H.S.; Park, S.H.; Kim, S.K. Long-term outcomes in children with glioblastoma. J. Neurosurg. Pediatr 2010, 6, 145–149. [Google Scholar] [CrossRef]
  116. Wang, J.; Wakeman, T.P.; Lathia, J.D.; Hjelmeland, A.B.; Wang, X.F.; White, R.R.; Rich, J.N.; Sullenger, B.A. Notch promotes radioresistance of glioma stem cells. Stem Cells 2010, 28, 17–28. [Google Scholar] [CrossRef] [Green Version]
  117. Bao, S.; Wu, Q.; Sathornsumetee, S.; Hao, Y.; Li, Z.; Hjelmeland, A.B.; Shi, Q.; McLendon, R.E.; Bigner, D.D.; Rich, J.N. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006, 66, 7843–7848. [Google Scholar] [CrossRef] [Green Version]
  118. Furnari, F.B.; Fenton, T.; Bachoo, R.M.; Mukasa, A.; Stommel, J.M.; Stegh, A.; Hahn, W.C.; Ligon, K.L.; Louis, D.N.; Brennan, C.; et al. Malignant astrocytic glioma: Genetics, biology, and paths to treatment. Genes Dev. 2007, 21, 2683–2710. [Google Scholar] [CrossRef] [Green Version]
  119. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
  120. Zhang, X.P.; Zheng, G.; Zou, L.; Liu, H.L.; Hou, L.H.; Zhou, P.; Yin, D.D.; Zheng, Q.J.; Liang, L.; Zhang, S.Z.; et al. Notch activation promotes cell proliferation and the formation of neural stem cell-like colonies in human glioma cells. Mol. Cell Biochem. 2008, 307, 101–108. [Google Scholar] [CrossRef]
  121. Dontu, G.; Jackson, K.W.; McNicholas, E.; Kawamura, M.J.; Abdallah, W.M.; Wicha, M.S. Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells. Breast Cancer Res. 2004, 6, R605–R615. [Google Scholar] [CrossRef] [Green Version]
  122. Farnie, G.; Clarke, R.B. Mammary stem cells and breast cancer--role of Notch signalling. Stem Cell Rev. 2007, 3, 169–175. [Google Scholar] [CrossRef]
  123. Fre, S.; Huyghe, M.; Mourikis, P.; Robine, S.; Louvard, D.; Artavanis-Tsakonas, S. Notch signals control the fate of immature progenitor cells in the intestine. Nature 2005, 435, 964–968. [Google Scholar] [CrossRef]
  124. Van Es, J.H.; van Gijn, M.E.; Riccio, O.; van den Born, M.; Vooijs, M.; Begthel, H.; Cozijnsen, M.; Robine, S.; Winton, D.J.; Radtke, F.; et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 2005, 435, 959–963. [Google Scholar] [CrossRef] [PubMed]
  125. Hitoshi, S.; Alexson, T.; Tropepe, V.; Donoviel, D.; Elia, A.J.; Nye, J.S.; Conlon, R.A.; Mak, T.W.; Bernstein, A.; van der Kooy, D. Notch pathway molecules are essential for the maintenance, but not the generation, of mammalian neural stem cells. Genes Dev. 2002, 16, 846–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hitoshi, S.; Seaberg, R.M.; Koscik, C.; Alexson, T.; Kusunoki, S.; Kanazawa, I.; Tsuji, S.; van der Kooy, D. Primitive neural stem cells from the mammalian epiblast differentiate to definitive neural stem cells under the control of Notch signaling. Genes Dev. 2004, 18, 1806–1811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Nakamura, Y.; Sakakibara, S.; Miyata, T.; Ogawa, M.; Shimazaki, T.; Weiss, S.; Kageyama, R.; Okano, H. The bHLH gene hes1 as a repressor of the neuronal commitment of CNS stem cells. J. Neurosci. 2000, 20, 283–293. [Google Scholar] [CrossRef] [Green Version]
  128. Pece, S.; Serresi, M.; Santolini, E.; Capra, M.; Hulleman, E.; Galimberti, V.; Zurrida, S.; Maisonneuve, P.; Viale, G.; Di Fiore, P.P. Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J. Cell Biol. 2004, 167, 215–221. [Google Scholar] [CrossRef] [Green Version]
  129. Politi, K.; Feirt, N.; Kitajewski, J. Notch in mammary gland development and breast cancer. Semin Cancer Biol. 2004, 14, 341–347. [Google Scholar] [CrossRef]
  130. Weng, A.P.; Ferrando, A.A.; Lee, W.; Morris, J.P.t.; Silverman, L.B.; Sanchez-Irizarry, C.; Blacklow, S.C.; Look, A.T.; Aster, J.C. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 2004, 306, 269–271. [Google Scholar] [CrossRef] [Green Version]
  131. Kanamori, M.; Kawaguchi, T.; Nigro, J.M.; Feuerstein, B.G.; Berger, M.S.; Miele, L.; Pieper, R.O. Contribution of Notch signaling activation to human glioblastoma multiforme. J. Neurosurg. 2007, 106, 417–427. [Google Scholar] [CrossRef]
  132. Armogida, M.; Petit, A.; Vincent, B.; Scarzello, S.; da Costa, C.A.; Checler, F. Endogenous beta-amyloid production in presenilin-deficient embryonic mouse fibroblasts. Nat. Cell Biol. 2001, 3, 1030–1033. [Google Scholar] [CrossRef]
  133. Huppert, S.S.; Le, A.; Schroeter, E.H.; Mumm, J.S.; Saxena, M.T.; Milner, L.A.; Kopan, R. Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1. Nature 2000, 405, 966–970. [Google Scholar] [CrossRef]
  134. Fan, X.; Matsui, W.; Khaki, L.; Stearns, D.; Chun, J.; Li, Y.M.; Eberhart, C.G. Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res. 2006, 66, 7445–7452. [Google Scholar] [CrossRef] [Green Version]
  135. Valiakhmetova, A.; Gorelyshev, S.; Konovalov, A.; Trunin, Y.; Savateev, A.; Kram, D.E.; Severson, E.; Hemmerich, A.; Edgerly, C.; Duncan, D.; et al. Treatment of Pediatric Glioblastoma with Combination Olaparib and Temozolomide Demonstrates 2-Year Durable Response. Oncologist 2020, 25, e198–e202. [Google Scholar] [CrossRef] [Green Version]
  136. Epstein, F.; Wisoff, J. Intra-axial tumors of the cervicomedullary junction. J. Neurosurg. 1987, 67, 483–487. [Google Scholar] [CrossRef] [Green Version]
  137. Stroink, A.R.; Hoffman, H.J.; Hendrick, E.B.; Humphreys, R.P.; Davidson, G. Transependymal benign dorsally exophytic brain stem gliomas in childhood: Diagnosis and treatment recommendations. Neurosurgery 1987, 20, 439–444. [Google Scholar] [CrossRef]
  138. Epstein, F.; McCleary, E.L. Intrinsic brain-stem tumors of childhood: Surgical indications. J. Neurosurg. 1986, 64, 11–15. [Google Scholar] [CrossRef] [Green Version]
  139. Green, A.L.; Kieran, M.W. Pediatric brainstem gliomas: New understanding leads to potential new treatments for two very different tumors. Curr. Oncol. Rep. 2015, 17, 436. [Google Scholar] [CrossRef]
  140. Pellot, J.E.; De Jesus, O. Diffuse Intrinsic Pontine Glioma. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2022. [Google Scholar]
  141. Sievers, P.; Sill, M.; Schrimpf, D.; Stichel, D.; Reuss, D.E.; Sturm, D.; Hench, J.; Frank, S.; Krskova, L.; Vicha, A.; et al. A subset of pediatric-type thalamic gliomas share a distinct DNA methylation profile, H3K27me3 loss and frequent alteration of EGFR. Neuro-Oncol. 2021, 23, 34–43. [Google Scholar] [CrossRef]
  142. Freeman, C.R.; Farmer, J.P. Pediatric brain stem gliomas: A review. Int. J. Radiat. Oncol. Biol. Phys. 1998, 40, 265–271. [Google Scholar] [CrossRef]
  143. Rubin, G.; Michowitz, S.; Horev, G.; Herscovici, Z.; Cohen, I.J.; Shuper, A.; Rappaport, Z.H. Pediatric brain stem gliomas: An update. Childs Nerv. Syst. 1998, 14, 167–173. [Google Scholar] [CrossRef]
  144. Guillamo, J.S.; Créange, A.; Kalifa, C.; Grill, J.; Rodriguez, D.; Doz, F.; Barbarot, S.; Zerah, M.; Sanson, M.; Bastuji-Garin, S.; et al. Prognostic factors of CNS tumours in Neurofibromatosis 1 (NF1): A retrospective study of 104 patients. Brain 2003, 126, 152–160. [Google Scholar] [CrossRef] [Green Version]
  145. Mahdi, J.; Shah, A.C.; Sato, A.; Morris, S.M.; McKinstry, R.C.; Listernick, R.; Packer, R.J.; Fisher, M.J.; Gutmann, D.H. A multi-institutional study of brainstem gliomas in children with neurofibromatosis type 1. Neurology 2017, 88, 1584–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Mahdi, J.; Goyal, M.S.; Griffith, J.; Morris, S.M.; Gutmann, D.H. Nonoptic pathway tumors in children with neurofibromatosis type 1. Neurology 2020, 95, e1052–e1059. [Google Scholar] [CrossRef] [PubMed]
  147. Young Poussaint, T.; Yousuf, N.; Barnes, P.D.; Anthony, D.C.; Zurakowski, D.; Scott, R.M.; Tarbell, N.J. Cervicomedullary astrocytomas of childhood: Clinical and imaging follow-up. Pediatr. Radiol. 1999, 29, 662–668. [Google Scholar] [CrossRef] [PubMed]
  148. Mulhern, R.K.; Heideman, R.L.; Khatib, Z.A.; Kovnar, E.H.; Sanford, R.A.; Kun, L.E. Quality of survival among children treated for brain stem glioma. Pediatr. Neurosurg. 1994, 20, 226–232. [Google Scholar] [CrossRef]
  149. Indelicato, D.J.; Rotondo, R.L.; Uezono, H.; Sandler, E.S.; Aldana, P.R.; Ranalli, N.J.; Beier, A.D.; Morris, C.G.; Bradley, J.A. Outcomes Following Proton Therapy for Pediatric Low-Grade Glioma. Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 149–156. [Google Scholar] [CrossRef] [Green Version]
  150. Constine, L.S.; Woolf, P.D.; Cann, D.; Mick, G.; McCormick, K.; Raubertas, R.F.; Rubin, P. Hypothalamic-pituitary dysfunction after radiation for brain tumors. N. Engl. J. Med. 1993, 328, 87–94. [Google Scholar] [CrossRef]
  151. Taphoorn, M.J.; Heimans, J.J.; van der Veen, E.A.; Karim, A.B. Endocrine functions in long-term survivors of low-grade supratentorial glioma treated with radiation therapy. J. Neurooncol. 1995, 25, 97–102. [Google Scholar] [CrossRef]
  152. Kato, T.; Sawamura, Y.; Tada, M.; Ikeda, J.; Ishii, N.; Abe, H. Cisplatin/vincristine chemotherapy for hypothalamic/visual pathway astrocytomas in young children. J. Neurooncol. 1998, 37, 263–270. [Google Scholar] [CrossRef]
  153. Packer, R.J.; Lange, B.; Ater, J.; Nicholson, H.S.; Allen, J.; Walker, R.; Prados, M.; Jakacki, R.; Reaman, G.; Needles, M.N.; et al. Carboplatin and vincristine for recurrent and newly diagnosed low-grade gliomas of childhood. J. Clin. Oncol. 1993, 11, 850–856. [Google Scholar] [CrossRef]
  154. Packer, R.J.; Ater, J.; Allen, J.; Phillips, P.; Geyer, R.; Nicholson, H.S.; Jakacki, R.; Kurczynski, E.; Needle, M.; Finlay, J.; et al. Carboplatin and vincristine chemotherapy for children with newly diagnosed progressive low-grade gliomas. J. Neurosurg. 1997, 86, 747–754. [Google Scholar] [CrossRef]
  155. Bouffet, E.; Amat, D.; Devaux, Y.; Desuzinges, C. Chemotherapy for spinal cord astrocytoma. Med. Pediatr. Oncol. 1997, 29, 560–562. [Google Scholar] [CrossRef]
  156. Merchant, T.E.; Li, C.; Xiong, X.; Kun, L.E.; Boop, F.A.; Sanford, R.A. Conformal radiotherapy after surgery for paediatric ependymoma: A prospective study. Lancet Oncol. 2009, 10, 258–266. [Google Scholar] [CrossRef] [Green Version]
  157. Koshy, M.; Rich, S.; Merchant, T.E.; Mahmood, U.; Regine, W.F.; Kwok, Y. Post-operative radiation improves survival in children younger than 3 years with intracranial ependymoma. J. Neurooncol. 2011, 105, 583–590. [Google Scholar] [CrossRef] [PubMed]
  158. Doxey, D.; Bruce, D.; Sklar, F.; Swift, D.; Shapiro, K. Posterior fossa syndrome: Identifiable risk factors and irreversible complications. Pediatr. Neurosurg. 1999, 31, 131–136. [Google Scholar] [CrossRef]
  159. Merchant, T.E.; Jenkins, J.J.; Burger, P.C.; Sanford, R.A.; Sherwood, S.H.; Jones-Wallace, D.; Heideman, R.L.; Thompson, S.J.; Helton, K.J.; Kun, L.E. Influence of tumor grade on time to progression after irradiation for localized ependymoma in children. Int. J. Radiat. Oncol. Biol. Phys. 2002, 53, 52–57. [Google Scholar] [CrossRef]
  160. Sanford, R.A.; Merchant, T.E.; Zwienenberg-Lee, M.; Kun, L.E.; Boop, F.A. Advances in surgical techniques for resection of childhood cerebellopontine angle ependymomas are key to survival. Childs Nerv. Syst. 2009, 25, 1229–1240. [Google Scholar] [CrossRef]
  161. Pollack, I.F.; Gerszten, P.C.; Martinez, A.J.; Lo, K.H.; Shultz, B.; Albright, A.L.; Janosky, J.; Deutsch, M. Intracranial ependymomas of childhood: Long-term outcome and prognostic factors. Neurosurgery 1995, 37, 655–666. [Google Scholar] [CrossRef]
  162. Nazar, G.B.; Hoffman, H.J.; Becker, L.E.; Jenkin, D.; Humphreys, R.P.; Hendrick, E.B. Infratentorial ependymomas in childhood: Prognostic factors and treatment. J. Neurosurg. 1990, 72, 408–417. [Google Scholar] [CrossRef] [Green Version]
  163. Van Veelen-Vincent, M.L.; Pierre-Kahn, A.; Kalifa, C.; Sainte-Rose, C.; Zerah, M.; Thorne, J.; Renier, D. Ependymoma in childhood: Prognostic factors, extent of surgery, and adjuvant therapy. J. Neurosurg. 2002, 97, 827–835. [Google Scholar] [CrossRef] [Green Version]
  164. Robertson, P.L.; Zeltzer, P.M.; Boyett, J.M.; Rorke, L.B.; Allen, J.C.; Geyer, J.R.; Stanley, P.; Li, H.; Albright, A.L.; McGuire-Cullen, P.; et al. Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in children: A report of the Children’s Cancer Group. J. Neurosurg. 1998, 88, 695–703. [Google Scholar] [CrossRef]
  165. Tihan, T.; Zhou, T.; Holmes, E.; Burger, P.C.; Ozuysal, S.; Rushing, E.J. The prognostic value of histological grading of posterior fossa ependymomas in children: A Children’s Oncology Group study and a review of prognostic factors. Mod. Pathol. 2008, 21, 165–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Eaton, B.R.; Goldberg, S.; Tarbell, N.J.; Lawell, M.P.; Gallotto, S.L.; Weyman, E.A.; Kuhlthau, K.A.; Ebb, D.H.; MacDonald, S.M.; Yock, T.I. Long-term health-related quality of life in pediatric brain tumor survivors receiving proton radiotherapy at <4 years of age. Neuro-Oncol. 2020, 22, 1379–1387. [Google Scholar] [CrossRef] [PubMed]
  167. Kulkarni, A.V.; Piscione, J.; Shams, I.; Bouffet, E. Long-term quality of life in children treated for posterior fossa brain tumors. J. Neurosurg. Pediatr. 2013, 12, 235–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Petito, C.K.; DeGirolami, U.; Earle, K.M. Craniopharyngiomas: A clinical and pathological review. Cancer 1976, 37, 1944–1952. [Google Scholar] [CrossRef]
  169. Garnett, M.R.; Puget, S.; Grill, J.; Sainte-Rose, C. Craniopharyngioma. Orphanet J. Rare Dis. 2007, 2, 18. [Google Scholar] [CrossRef] [Green Version]
  170. Elliott, R.E.; Hsieh, K.; Hochm, T.; Belitskaya-Levy, I.; Wisoff, J.; Wisoff, J.H. Efficacy and safety of radical resection of primary and recurrent craniopharyngiomas in 86 children. J. Neurosurg. Pediatr. 2010, 5, 30–48. [Google Scholar] [CrossRef] [Green Version]
  171. Lin, L.L.; El Naqa, I.; Leonard, J.R.; Park, T.S.; Hollander, A.S.; Michalski, J.M.; Mansur, D.B. Long-term outcome in children treated for craniopharyngioma with and without radiotherapy. J. Neurosurg. Pediatr. 2008, 1, 126–130. [Google Scholar] [CrossRef]
  172. Stripp, D.C.; Maity, A.; Janss, A.J.; Belasco, J.B.; Tochner, Z.A.; Goldwein, J.W.; Moshang, T.; Rorke, L.B.; Phillips, P.C.; Sutton, L.N.; et al. Surgery with or without radiation therapy in the management of craniopharyngiomas in children and young adults. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 714–720. [Google Scholar] [CrossRef]
  173. Moon, S.H.; Kim, I.H.; Park, S.W.; Kim, I.; Hong, S.; Park, C.I.; Wang, K.C.; Cho, B.K. Early adjuvant radiotherapy toward long-term survival and better quality of life for craniopharyngiomas—A study in single institute. Childs Nerv. Syst. 2005, 21, 799–807. [Google Scholar] [CrossRef]
  174. Garrè, M.L.; Cama, A. Craniopharyngioma: Modern concepts in pathogenesis and treatment. Curr. Opin. Pediatr. 2007, 19, 471–479. [Google Scholar] [CrossRef]
  175. Müller, H.L. Consequences of craniopharyngioma surgery in children. J. Clin. Endocrinol. Metab. 2011, 96, 1981–1991. [Google Scholar] [CrossRef] [Green Version]
  176. Merchant, T.E.; Kiehna, E.N.; Li, C.; Xiong, X.; Mulhern, R.K. Radiation dosimetry predicts IQ after conformal radiation therapy in pediatric patients with localized ependymoma. Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 1546–1554. [Google Scholar] [CrossRef]
  177. Chapman, C.A.; Waber, D.P.; Bernstein, J.H.; Pomeroy, S.L.; LaVally, B.; Sallan, S.E.; Tarbell, N. Neurobehavioral and neurologic outcome in long-term survivors of posterior fossa brain tumors: Role of age and perioperative factors. J. Child Neurol. 1995, 10, 209–212. [Google Scholar] [CrossRef]
  178. Palmer, S.L.; Hassall, T.; Evankovich, K.; Mabbott, D.J.; Bonner, M.; Deluca, C.; Cohn, R.; Fisher, M.J.; Morris, E.B.; Broniscer, A.; et al. Neurocognitive outcome 12 months following cerebellar mutism syndrome in pediatric patients with medulloblastoma. Neuro-Oncol. 2010, 12, 1311–1317. [Google Scholar] [CrossRef]
  179. Mulhern, R.K.; Reddick, W.E.; Palmer, S.L.; Glass, J.O.; Elkin, T.D.; Kun, L.E.; Taylor, J.; Langston, J.; Gajjar, A. Neurocognitive deficits in medulloblastoma survivors and white matter loss. Ann. Neurol. 1999, 46, 834–841. [Google Scholar] [CrossRef]
  180. Copeland, D.R.; deMoor, C.; Moore, B.D., 3rd; Ater, J.L. Neurocognitive development of children after a cerebellar tumor in infancy: A longitudinal study. J. Clin. Oncol. 1999, 17, 3476–3486. [Google Scholar] [CrossRef]
  181. Hoppe-Hirsch, E.; Renier, D.; Lellouch-Tubiana, A.; Sainte-Rose, C.; Pierre-Kahn, A.; Hirsch, J.F. Medulloblastoma in childhood: Progressive intellectual deterioration. Childs Nerv. Syst. 1990, 6, 60–65. [Google Scholar] [CrossRef]
  182. Armstrong, F.D.; Blumberg, M.J.; Toledano, S.R. Neurobehavioral issues in childhood cancer. School Psychol. Rev. 1999, 28, 194. [Google Scholar] [CrossRef]
  183. DeAngelis, L.M.; Delattre, J.Y.; Posner, J.B. Radiation-induced dementia in patients cured of brain metastases. Neurology 1989, 39, 789–796. [Google Scholar] [CrossRef]
  184. Tofilon, P.J.; Fike, J.R. The radioresponse of the central nervous system: A dynamic process. Radiat. Res. 2000, 153, 357–370. [Google Scholar] [CrossRef]
  185. Reddick, W.E.; Russell, J.M.; Glass, J.O.; Xiong, X.; Mulhern, R.K.; Langston, J.W.; Merchant, T.E.; Kun, L.E.; Gajjar, A. Subtle white matter volume differences in children treated for medulloblastoma with conventional or reduced dose craniospinal irradiation. Magn. Reson. Imaging 2000, 18, 787–793. [Google Scholar] [CrossRef]
  186. Palmer, S.L.; Reddick, W.E.; Glass, J.O.; Gajjar, A.; Goloubeva, O.; Mulhern, R.K. Decline in corpus callosum volume among pediatric patients with medulloblastoma: Longitudinal MR imaging study. AJNR Am. J. Neuroradiol. 2002, 23, 1088–1094. [Google Scholar] [PubMed]
  187. Attia, A.; Page, B.R.; Lesser, G.J.; Chan, M. Treatment of radiation-induced cognitive decline. Curr. Treat Options Oncol. 2014, 15, 539–550. [Google Scholar] [CrossRef] [PubMed]
  188. Greene-Schloesser, D.; Robbins, M.E.; Peiffer, A.M.; Shaw, E.G.; Wheeler, K.T.; Chan, M.D. Radiation-induced brain injury: A review. Front. Oncol. 2012, 2, 73. [Google Scholar] [CrossRef] [Green Version]
  189. Reddick, W.E.; Mulhern, R.K.; Elkin, T.D.; Glass, J.O.; Merchant, T.E.; Langston, J.W. A hybrid neural network analysis of subtle brain volume differences in children surviving brain tumors. Magn. Reson. Imaging 1998, 16, 413–421. [Google Scholar] [CrossRef]
  190. Apple, D.M.; Fonseca, R.S.; Kokovay, E. The role of adult neurogenesis in psychiatric and cognitive disorders. Brain Res. 2017, 1655, 270–276. [Google Scholar] [CrossRef] [Green Version]
  191. Taupin, P. Neural progenitor and stem cells in the adult central nervous system. Ann. Acad. Med. 2006, 35, 814–820. [Google Scholar]
  192. Greenberger, B.A.; Pulsifer, M.B.; Ebb, D.H.; MacDonald, S.M.; Jones, R.M.; Butler, W.E.; Huang, M.S.; Marcus, K.J.; Oberg, J.A.; Tarbell, N.J.; 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, 1060–1068. [Google Scholar] [CrossRef]
  193. Tsai, P.F.; Yang, C.C.; Chuang, C.C.; Huang, T.Y.; Wu, Y.M.; Pai, P.C.; Tseng, C.K.; Wu, T.H.; Shen, Y.L.; Lin, S.Y. Hippocampal dosimetry correlates with the change in neurocognitive function after hippocampal sparing during whole brain radiotherapy: A prospective study. Radiat. Oncol. 2015, 10, 253. [Google Scholar] [CrossRef] [Green Version]
  194. Kerns, K.A.; Thomson, J. Implementation of a compensatory memory system in a school age child with severe memory impairment. Pediatr. Rehabil. 1998, 2, 77–87. [Google Scholar] [CrossRef]
  195. Brown, R.; Dingle, A.; Dreelin, B. Neuropsychological effects of stimulant medication on children’s learning and behavior. In Handbook of Clinical Child Pharmacology; Wiley: New York, NY, USA, 1997. [Google Scholar]
  196. Thompson, S.J.; Leigh, L.; Christensen, R.; Xiong, X.; Kun, L.E.; Heideman, R.L.; Reddick, W.E.; Gajjar, A.; Merchant, T.; Pui, C.H.; et al. Immediate neurocognitive effects of methylphenidate on learning-impaired survivors of childhood cancer. J. Clin. Oncol. 2001, 19, 1802–1808. [Google Scholar] [CrossRef]
  197. Torres, C.F.; Korones, D.N.; Palumbo, D.R.; Wissler, K.H.; Vadasz, E.; Cox, C. Effect of methylphenidate in the post-radiation attention and memory deficits in children. Ann. Neurol. 1996, 40, 331–332. [Google Scholar]
  198. Leigh, L.; Miles, M. Educational issues for children with cancer. In Principles and Practice of Paediatric Oncology, 4th ed.; Pizzo, P., Poplack, D., Eds.; Lippincott: Philadelphia, PA, USA, 2002. [Google Scholar]
  199. Max, J.E.; Roberts, M.A.; Koele, S.L.; Lindgren, S.D.; Robin, D.A.; Arndt, S.; Smith, W.L., Jr.; Sato, Y. Cognitive outcome in children and adolescents following severe traumatic brain injury: Influence of psychosocial, psychiatric, and injury-related variables. J. Int. Neuropsychol. Soc. 1999, 5, 58–68. [Google Scholar] [CrossRef]
  200. Yock, T.I.; Yeap, B.Y.; Ebb, D.H.; Weyman, E.; Eaton, B.R.; Sherry, N.A.; Jones, R.M.; MacDonald, S.M.; Pulsifer, M.B.; Lavally, B.; et al. Long-term toxic effects of proton radiotherapy for paediatric medulloblastoma: A phase 2 single-arm study. Lancet Oncol. 2016, 17, 287–298. [Google Scholar] [CrossRef]
  201. Antonini, T.N.; Ris, M.D.; Grosshans, D.R.; Mahajan, A.; Okcu, M.F.; Chintagumpala, M.; Paulino, A.; Child, A.E.; Orobio, J.; Stancel, H.H.; et al. Attention, processing speed, and executive functioning in pediatric brain tumor survivors treated with proton beam radiation therapy. Radiother. Oncol. 2017, 124, 89–97. [Google Scholar] [CrossRef]
  202. Pulsifer, M.B.; Sethi, R.V.; Kuhlthau, K.A.; MacDonald, S.M.; Tarbell, N.J.; Yock, T.I. Early Cognitive Outcomes Following Proton Radiation in Pediatric Patients With Brain and Central Nervous System Tumors. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 400–407. [Google Scholar] [CrossRef] [Green Version]
  203. Oshiro, Y.; Okumura, T.; Mizumoto, M.; Fukushima, T.; Ishikawa, H.; Hashimoto, T.; Tsuboi, K.; Kaneko, M.; Sakurai, H. Proton beam therapy for unresectable hepatoblastoma in children: Survival in one case. Acta Oncol. 2013, 52, 600–603. [Google Scholar] [CrossRef]
  204. Kanemoto, A.; Oshiro, Y.; Sugahara, S.; Kamagata, S.; Hirobe, S.; Toma, M.; Okumura, T.; Sakurai, H. Proton beam therapy for inoperable recurrence of bronchial high-grade mucoepidermoid carcinoma. Jpn. J. Clin. Oncol. 2012, 42, 552–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Mizumoto, M.; Hashii, H.; Senarita, M.; Sakai, S.; Wada, T.; Okumura, T.; Tsuboi, K.; Sakurai, H. Proton beam therapy for malignancy in Bloom syndrome. Strahlenther. Onkol. 2013, 189, 335–338. [Google Scholar] [CrossRef]
  206. MacDonald, S.M.; Safai, S.; Trofimov, A.; Wolfgang, J.; Fullerton, B.; Yeap, B.Y.; Bortfeld, T.; Tarbell, N.J.; Yock, T. Proton radiotherapy for childhood ependymoma: Initial clinical outcomes and dose comparisons. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 979–986. [Google Scholar] [CrossRef]
  207. Miralbell, R.; Lomax, A.; Cella, L.; Schneider, U. Potential reduction of the incidence of radiation-induced second cancers by using proton beams in the treatment of pediatric tumors. Int. J. Radiat. Oncol. Biol. Phys. 2002, 54, 824–829. [Google Scholar] [CrossRef]
  208. Kahalley, L.S.; Ris, M.D.; Grosshans, D.R.; Okcu, M.F.; Paulino, A.C.; Chintagumpala, M.; Moore, B.D.; Guffey, D.; Minard, C.G.; Stancel, H.H.; et al. Comparing Intelligence Quotient Change After Treatment With Proton Versus Photon Radiation Therapy for Pediatric Brain Tumors. J. Clin. Oncol. 2016, 34, 1043–1049. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, R.; Howell, R.M.; Giebeler, A.; Taddei, P.J.; Mahajan, A.; Newhauser, W.D. Comparison of risk of radiogenic second cancer following photon and proton craniospinal irradiation for a pediatric medulloblastoma patient. Phys. Med. Biol. 2013, 58, 807–823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Paganetti, H.; Niemierko, A.; Ancukiewicz, M.; Gerweck, L.E.; Goitein, M.; Loeffler, J.S.; Suit, H.D. Relative biological effectiveness (RBE) values for proton beam therapy. Int. J. Radiat. Oncol. Biol. Phys. 2002, 53, 407–421. [Google Scholar] [CrossRef]
  211. Wambersie, A. RBE, reference RBE and clinical RBE: Applications of these concepts in hadron therapy. Strahlenther. Onkol. 1999, 175 (Suppl. 2), 39–43. [Google Scholar] [CrossRef]
  212. DeLaney, T.F. Prescribing, Recording, and Reporting Proton-Beam Therapy. Radiat. Res. 2009, 171, 776. [Google Scholar] [CrossRef]
  213. Ladra, M.M.; Edgington, S.K.; Mahajan, A.; Grosshans, D.; Szymonifka, J.; Khan, F.; Moteabbed, M.; Friedmann, A.M.; MacDonald, S.M.; Tarbell, N.J.; et al. A dosimetric comparison of proton and intensity modulated radiation therapy in pediatric rhabdomyosarcoma patients enrolled on a prospective phase II proton study. Radiother. Oncol. 2014, 113, 77–83. [Google Scholar] [CrossRef] [Green Version]
  214. Lee, C.T.; Bilton, S.D.; Famiglietti, R.M.; Riley, B.A.; Mahajan, A.; Chang, E.L.; Maor, M.H.; Woo, S.Y.; Cox, J.D.; Smith, A.R. Treatment planning with protons for pediatric retinoblastoma, medulloblastoma, and pelvic sarcoma: How do protons compare with other conformal techniques? Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 362–372. [Google Scholar] [CrossRef]
  215. Yock, T.; Schneider, R.; Friedmann, A.; Adams, J.; Fullerton, B.; Tarbell, N. Proton radiotherapy for orbital rhabdomyosarcoma: Clinical outcome and a dosimetric comparison with photons. Int. J. Radiat. Oncol. Biol. Phys. 2005, 63, 1161–1168. [Google Scholar] [CrossRef]
  216. Palm, A.; Johansson, K.A. A review of the impact of photon and proton external beam radiotherapy treatment modalities on the dose distribution in field and out-of-field; implications for the long-term morbidity of cancer survivors. Acta Oncol. 2007, 46, 462–473. [Google Scholar] [CrossRef]
  217. Fukushima, H.; Fukushima, T.; Sakai, A.; Suzuki, R.; Kobayashi, C.; Oshiro, Y.; Mizumoto, M.; Hoshino, N.; Gotoh, C.; Urita, Y.; et al. Tailor-made treatment combined with proton beam therapy for children with genitourinary/pelvic rhabdomyosarcoma. Rep. Pract. Oncol. Radiother. 2015, 20, 217–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Oshiro, Y.; Mizumoto, M.; Okumura, T.; Sugahara, S.; Fukushima, T.; Ishikawa, H.; Nakao, T.; Hashimoto, T.; Tsuboi, K.; Ohkawa, H.; et al. Clinical results of proton beam therapy for advanced neuroblastoma. Radiat. Oncol. 2013, 8, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Oshiro, Y.; Sugahara, S.; Fukushima, T.; Okumura, T.; Nakao, T.; Mizumoto, M.; Hashimoto, T.; Tsuboi, K.; Kaneko, M.; Sakurai, H. Pediatric nasopharyngeal carcinoma treated with proton beam therapy. Two case reports. Acta Oncol. 2011, 50, 470–473. [Google Scholar] [CrossRef] [PubMed]
  220. Mizumoto, M.; Oshiro, Y.; Takizawa, D.; Fukushima, T.; Fukushima, H.; Yamamoto, T.; Muroi, A.; Okumura, T.; Tsuboi, K.; Sakurai, H. Proton beam therapy for pediatric ependymoma. Pediatr. Int. 2015, 57, 567–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Fuss, M.; Hug, E.B.; Schaefer, R.A.; Nevinny-Stickel, M.; Miller, D.W.; Slater, J.M.; Slater, J.D. Proton radiation therapy (PRT) for pediatric optic pathway gliomas: Comparison with 3D planned conventional photons and a standard photon technique. Int. J. Radiat. Oncol. Biol. Phys. 1999, 45, 1117–1126. [Google Scholar] [CrossRef]
  222. Beltran, C.; Roca, M.; Merchant, T.E. On the benefits and risks of proton therapy in pediatric craniopharyngioma. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, e281–e287. [Google Scholar] [CrossRef] [Green Version]
  223. Boehling, N.S.; Grosshans, D.R.; Bluett, J.B.; Palmer, M.T.; Song, X.; Amos, R.A.; Sahoo, N.; Meyer, J.J.; Mahajan, A.; Woo, S.Y. Dosimetric comparison of three-dimensional conformal proton radiotherapy, intensity-modulated proton therapy, and intensity-modulated radiotherapy for treatment of pediatric craniopharyngiomas. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 643–652. [Google Scholar] [CrossRef]
  224. Childs, S.K.; Kozak, K.R.; Friedmann, A.M.; Yeap, B.Y.; Adams, J.; MacDonald, S.M.; Liebsch, N.J.; Tarbell, N.J.; Yock, T.I. Proton radiotherapy for parameningeal rhabdomyosarcoma: Clinical outcomes and late effects. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 635–642. [Google Scholar] [CrossRef] [Green Version]
  225. Yock, T.; Yeap, B.; Pulsifer, M.; Ebb, D.; Macdonald, S.; Marcus, K.; Tarbell, N. Results from a Prospective Trial of Proton Radiotherapy for Medulloblastoma: Clinical Outcomes including Hearing and Neurocognitive. Fuel Energy Abstr. 2011, 81, S113. [Google Scholar] [CrossRef]
  226. Yock, T.I.; Bhat, S.; Szymonifka, J.; Yeap, B.Y.; Delahaye, J.; Donaldson, S.S.; MacDonald, S.M.; Pulsifer, M.B.; Hill, K.S.; DeLaney, T.F.; et al. Quality of life outcomes in proton and photon treated pediatric brain tumor survivors. Radiother. Oncol. 2014, 113, 89–94. [Google Scholar] [CrossRef] [Green Version]
  227. Child, A.E.; Warren, E.A.; Grosshans, D.R.; Paulino, A.C.; Okcu, M.F.; Ris, M.D.; Mahajan, A.; Orobio, J.; Cirino, P.T.; Minard, C.G.; et al. Long-term cognitive and academic outcomes among pediatric brain tumor survivors treated with proton versus photon radiotherapy. Pediatr. Blood Cancer 2021, 68, e29125. [Google Scholar] [CrossRef]
  228. Kahalley, L.S.; Peterson, R.; Ris, M.D.; Janzen, L.; Okcu, M.F.; Grosshans, D.R.; Ramaswamy, V.; Paulino, A.C.; Hodgson, D.; Mahajan, A.; et al. Superior Intellectual Outcomes After Proton Radiotherapy Compared With Photon Radiotherapy for Pediatric Medulloblastoma. J. Clin. Oncol. 2020, 38, 454–461. [Google Scholar] [CrossRef]
  229. Kahalley, L.S.; Douglas Ris, M.; Mahajan, A.; Fatih Okcu, M.; Chintagumpala, M.; Paulino, A.C.; Whitehead, W.E.; Minard, C.G.; Stancel, H.H.; Orobio, J.; et al. Prospective, longitudinal comparison of neurocognitive change in pediatric brain tumor patients treated with proton radiotherapy versus surgery only. Neuro-Oncol. 2019, 21, 809–818. [Google Scholar] [CrossRef] [Green Version]
  230. Kahalley, L.S.; Winter-Greenberg, A.; Stancel, H.; Ris, M.D.; Gragert, M. Utility of the General Ability Index (GAI) and Cognitive Proficiency Index (CPI) with survivors of pediatric brain tumors: Comparison to Full Scale IQ and premorbid IQ estimates. J. Clin. Exp. Neuropsychol. 2016, 38, 1065–1076. [Google Scholar] [CrossRef] [Green Version]
  231. Macdonald, S.M.; Sethi, R.; Lavally, B.; Yeap, B.Y.; Marcus, K.J.; Caruso, P.; Pulsifer, M.; Huang, M.; Ebb, D.; Tarbell, N.J.; et al. Proton radiotherapy for pediatric central nervous system ependymoma: Clinical outcomes for 70 patients. Neuro-Oncol. 2013, 15, 1552–1559. [Google Scholar] [CrossRef] [Green Version]
  232. Weusthof, K.; Luttich, P.; Regnery, S.; Konig, L.; Bernhardt, D.; Witt, O.; Herfarth, K.; Unterberg, A.; Jungk, C.; Farnia, B.; et al. Neurocognitive Outcomes in Pediatric Patients Following Brain Irradiation. Cancers 2021, 13, 3538. [Google Scholar] [CrossRef]
Figure 1. Graphical depiction of proton-beam dosage per tissue depth. The figure demonstrates that the dosage of the proton beam is higher in deeper tissues. The more superficial tissues are spared from increased dosages (blue line). In contrast, photon-beam radiation (green line) has increased dosage in superficial tissue with decreased dosage in deeper tissues.
Figure 1. Graphical depiction of proton-beam dosage per tissue depth. The figure demonstrates that the dosage of the proton beam is higher in deeper tissues. The more superficial tissues are spared from increased dosages (blue line). In contrast, photon-beam radiation (green line) has increased dosage in superficial tissue with decreased dosage in deeper tissues.
Jpm 12 01050 g001
Table 1. Description of pediatric-tumor treatments including proposed management and dosages.
Table 1. Description of pediatric-tumor treatments including proposed management and dosages.
Tumor TypeMainstay TreatmentSpecific Radiation TherapyReported Survival
MedulloblastomaMaximal surgical resection with
radiation and multiagent
Chemotherapy [59,60,61,62].
Proton-beam-radiation therapy (23.4 Gy in 13 fractions of
craniospinal irradiation with a
posterior fossa boost of 30.6 Gy in 17 fractions)
86% at 5 years [59], 83% at 61.2 months [60], 78% at 4.8 years [61], 87.6% at 6 years [62]
Low-Grade Glioma
(Pilocytic Astrocytoma)
Maximal surgical resection.
Multimodal therapy for
unresectable tumors and recurrent tumors [64].
Proton-beam radiation>90% at 10 years [64]
High-Grade GliomaMaximal surgical resection.
Stereotactic biopsy if unresectable. Radiotherapy with concomitant
temozolomide for non-surgical cases [65].
Proton-beam-radiation strength, dependent on radiosensitivity of
tumor cells
26.5% at 2 years [65]
Brain Stem GliomaResection when possible and
radiation therapy [9,66,67,68,69].
Proton-beam radiationWidely variable: 34% at 5 years [68], 100% at 5 years [66], 33% at 5 years [69]
EpendymomaMaximal surgical resection. Local or craniospinal radiation in those with subtotal resection [70,71,72].Proton-beam radiation
(54–59.4 Gy)
76% at 10 years [71], 74.8% at 5 years [72]
CraniopharyngiomaControversial surgical resection with adjuvant radiotherapy [73].Proton-beam radiation88% at 5 years [73]
Table 2. Summary of the late effects due to radiotherapy on neurocognition.
Table 2. Summary of the late effects due to radiotherapy on neurocognition.
Key Points of Discussion on the Neurocognitive Late Effects from Radiotherapy
  • Decline in IQ and academic achievement [29,35,36].
  • Decline in processing speed and memory [183].
  • Early-Onset dementia [183].
  • Destruction of white brain matter, oligodendrocytes, and other neurons [184,185,186].
Table 3. Comparison of complications (favorable or unfavorable) in the different radiotherapy modalities.
Table 3. Comparison of complications (favorable or unfavorable) in the different radiotherapy modalities.
Type of RadiotherapyProton-Beam RadiotherapyIMRT (Photon)VMAT (Photon)
Comparison of
Complications
  • Reduction in dose to vital organs [41,203,204,205].
  • Favorable sparing of normal brain tissue in pediatric ependymoma [206].
  • Reduced incidence of secondary cancer in medulloblastoma [207].
  • No significant decline in IQ [208].
  • No significant impairment in neurocognitive function in [200].
  • Reduction in risk of secondary cancer [209].
  • Reduction in neurotoxicity [41,203,204,205].
  • Unfavorable sparing of normal brain tissue in pediatric ependymoma [206].
  • No significant neurocognitive impairment in survivors [200,201,202,208].
  • Increased need for endocrine replacement [58].
  • Significant neurocognitive impairment [200,201,202,208].
  • 18% lifetime risk of secondary cancer [209].
  • Reduction in neurotoxicity [41,203,204,205].
  • Significant neurocognitive impairment [200,201,202,208].
  • Increased need for endocrine replacement [58].
Both IMRT and VMAT are subtypes of photon-based radiotherapy. IMRT = intensity modulated radiotherapy. VMAT = volumetric modulated arc therapy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Major, N.; Patel, N.A.; Bennett, J.; Novakovic, E.; Poloni, D.; Abraham, M.; Brown, N.J.; Gendreau, J.L.; Sahyouni, R.; Loya, J. The Current State of Radiotherapy for Pediatric Brain Tumors: An Overview of Post-Radiotherapy Neurocognitive Decline and Outcomes. J. Pers. Med. 2022, 12, 1050. https://doi.org/10.3390/jpm12071050

AMA Style

Major N, Patel NA, Bennett J, Novakovic E, Poloni D, Abraham M, Brown NJ, Gendreau JL, Sahyouni R, Loya J. The Current State of Radiotherapy for Pediatric Brain Tumors: An Overview of Post-Radiotherapy Neurocognitive Decline and Outcomes. Journal of Personalized Medicine. 2022; 12(7):1050. https://doi.org/10.3390/jpm12071050

Chicago/Turabian Style

Major, Nicholas, Neal A. Patel, Josiah Bennett, Ena Novakovic, Dana Poloni, Mickey Abraham, Nolan J. Brown, Julian L. Gendreau, Ronald Sahyouni, and Joshua Loya. 2022. "The Current State of Radiotherapy for Pediatric Brain Tumors: An Overview of Post-Radiotherapy Neurocognitive Decline and Outcomes" Journal of Personalized Medicine 12, no. 7: 1050. https://doi.org/10.3390/jpm12071050

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop