439
Views
36
CrossRef citations to date
0
Altmetric
Review

Medulloblastoma: optimizing care with a multidisciplinary approach

&
Pages 335-347 | Published online: 30 Apr 2019

Abstract

Medulloblastoma is a malignant tumor of the cerebellum and the most frequent malignant brain tumor in children. The standard of care consists of maximal resection surgery, followed by craniospinal irradiation and chemotherapy. Such treatment allows long-term survival rates of nearly 70%; however, there are wide disparities among patient outcomes, and in any case, major long-term morbidity is observed with conventional treatment. In the last two decades, the molecular understanding of medulloblastoma has improved drastically, allowing us to revolutionize our understanding of medulloblastoma pathophysiological mechanisms. These advances led to an international consensus in 2010 that defined four prognostic molecular subgroups named after their affected signaling pathways, that is, WNT, SHH, Group 3 and Group 4. The molecular understanding of medulloblastoma is starting to translate through to clinical settings due to the development of targeted therapies. Moreover, recent improvements in radiotherapy modalities and the reconsideration of craniospinal irradiation according to the molecular status hold promise for survival preservation and the reduction of radiation-induced morbidity. This review is an overview of the current knowledge of medulloblastoma through a molecular approach, and therapeutic prospects currently being developed in surgery, radiotherapy and targeted therapies to optimize the treatment of medulloblastoma with a multidisciplinary approach will also be discussed.

Introduction

Medulloblastoma is a highly aggressive malignant tumor of the cerebellumCitation1 and the most common malignant brain tumor in children, accounting for nearly 20% of all central nervous system (CNS) tumors among childrenCitation1,Citation2 but only 1% of all CNS tumors among adults. In 70–80% of cases, medulloblastoma affects children 16 years or younger.Citation1 The average annual age-adjusted incidence rates range from 0.20 to 0.58 cases per 100,000 persons.Citation3

Currently, the 5-year overall survival (OS) rates reach ~70%, but patients present with very different outcomes. Medulloblastoma is stratified into two main risk levels based on age, presence of metastases, extent of residual disease after surgery and histologic characteristics of the tumor.Citation4 Consequently, the 5-year survival rates for standard and high-risk medulloblastoma are over 80% and ~60%, respectively.Citation5Citation7

Conventional treatment, for both standard and high-risk patients, involves a combination of maximal resection surgery, craniospinal irradiation (CSI), and cytotoxic chemotherapy (CT). This combination achieves long-term OS in 60–80% of patients but often at the expense of devastating long-term toxicities.Citation8Citation10 Therefore, a better understanding of the disease appears to be a relevant challenge with the aim of providing more accurate risk-adapted treatment and developing targeted therapies to decrease side effects in low-risk patients and improve efficiency in high-risk patients. In the last two decades, major advances have already been made in understanding the molecular mechanisms underlying medul-loblastoma.Citation11,Citation12 These findings have led to an international consensus for defining the prognostic molecular subgroups of medulloblastoma, which have been included in the recently published revised fourth edition of the WHO Classification of Tumors of the CNS.Citation13 A molecular understanding of medulloblastoma is starting to translate through to clinical settings due to the development of subgroup-specific approaches for clinical trials, allowing a more accurate distribution of radiation dosage or CT schedules, and evaluating the efficiency of emerging candidates for targeted therapies.

In this review, we will present the current landscape of the medulloblastoma molecular classification and attempt to correlate this classification with emerging therapeutic strategies to optimize medulloblastoma care through a multidisciplinary approach by finding the right treatments for the right group of patients to increase survival rates and reduce treatment toxicity, hence improving the quality of life (QoL) of survivors.

Clinical classification

The current prognostic classification divides medulloblastomas into “Standard Risk” and “High Risk” based on age, presence of metastases, extent of postsurgical residual disease and histology. Five histological subtypes are individualized: classical, desmoplastic/nodular, with extensive nodularity, anaplastic, and large cell variants. Large cell and anaplastic medulloblastomas overlap in a considerable number of cytological features, and consequently, these types are often considered together and grouped as “large cell/anaplastic histology”,Citation14 which is related to poor prognosis. Anaplastic histology is characterized by marked cytological pleomorphism across most of its area in association with high mitotic and apoptotic counts.Citation15 Large cell medulloblastoma is defined by groups of uniform large round cells with a single nucleolus, which also have higher mitotic and apoptotic indices than in other histolo-gies.Citation16 Medulloblastoma with extensive nodularity, which is closely related to the desmoplastic/nodular variant, has a more favorable outcome.Citation17 Considering the surgical criteria, patients with less than 1.5 cm2 residual disease after surgery present significantly better outcomes.Citation18 Metastatic status is determined by using the Chang criteria, which distinguish four metastatic levels: presence of tumor cells in cerebrospinal fluid (CSF; M1), gross nodular seeding of brain CSF spaces (M2), gross nodular seeding of spinal CSF spaces (M3), and extraneural spread (M4).Citation19 Infants (under 3 years old) have lower survival rates than older children, although this finding could be a result of therapeutic strategies that must eliminate radiotherapy (RT) in this population due to unacceptable long-term morbidity.

Hence, based on this clinical and histological classification, standard-risk patients – 70% of medulloblastomas at the time of diagnosisCitation20 – are children aged >3 years with no evidence of disseminated disease on craniospinal magnetic resonance imaging or CSF cytology, postoperative residual tumor under 1.5 cm2 and non-large cell/anaplastic histology.Citation21 Five-year OS reaches ~85%.Citation18,Citation22 If one or more of these criteria is not available, then the patient is considered to be at high risk, and the 5-year OS declines to ~60%.Citation7

Molecular classification

The existence of distinct molecular subgroups of medulloblastoma was highlighted in 2002 by Pomeroy et al.Citation23 They showed, by studying DNA microarray gene expression data, that PTCH, GLI and MYCN (all three transcriptional targets of SHH) were highly correlated with desmoplastic/nodular medulloblastoma. They also showed molecular markers of the variability of medulloblastoma outcome. On the same topic, several studies started to sub-classify medulloblastoma according to differences in the transcriptome, with largely convergent conclusions. Thompson et alCitation24 in 2006, and Kool et alCitation25 in 2008, concluded the existence of five distinct molecular subtypes named A, B, C, D, and E. Cho et alCitation26 concluded in 2011 the existence of six distinct molecular subtypes named C1 to C6, and Northcott et alCitation27 concluded in 2011 the existence of four distinct molecular subtypes named SHH, WNT, Group C, and Group D. Variations in the number, composition, and nature of the subgroups between studies brought about a consensus conference in Boston in the fall of 2010, where it was agreed there were four main transcriptional subgroups of medulloblastoma named WNT, SHH, Group 3, and Group 4, clearly distinct in terms of demographics, histology, DNA copy-number aberrations, and clinical outcome.Citation28

The WNT subgroup is the rarest (10% of all medulloblastomas) but has the best clinical outcome prediction, with a 5-year OS > 95%.Citation6,Citation29,Citation30 This molecular subgroup is predominantly associated with classical histology. WNT medulloblastomas rarely have a large cell/anaplastic histology, but even with this histology, they present an excellent prognosis.Citation28 WNT medulloblastomas occur in children older than 3 years or teenagers, and the cell of origin derives from the lower rhombic lip.Citation31 The molecular mechanism is defined by the activation of the WNT signaling pathway, which acts via β-catenin expression as a transduction enhancer. In 85–90% of cases, the activation of the WNT signaling pathway results from activating somatic mutations in exon 3 of CTNNB1, leading to the overexpression of β-catenin. Monosomy 6 is also highly recurrent among WNT tumors, presenting in 70–80% of cases.Citation26,Citation31,Citation32 Less frequently, somatic mutations in TP53, SMARCA4, and DDX3X are found.Citation33 WNT medulloblastoma rarely occurs in the context of germ-line mutations in APC consistent with Turcot syndrome.Citation34 Germline mutations in ALK have also been found in rare cases of WNT medulloblastoma, although the physiopathology is not understood.Citation35

The SHH subgroup represents 30% of all medulloblasto-mas,Citation36 and the prognosis is intermediate, with a 5-year OS of 70%.Citation26 There is a strong association between desmoplastic/nodular histology and SHH tumors since the vast majority of desmoplastic/nodular cases belong to the SHH subgroup,Citation28,Citation37 but up to 50% of SHH subgroup medulloblastomas are not desmoplastic/nodular.Citation28 SHH medulloblastomas are most frequently found in infants and adults and occur much less frequently in patients aged 4–15 years.Citation29 These tumors derive from the cerebellar granule precursor cells of the external granule layer.Citation38 The molecular mechanism involves the over-expression of the SHH signaling pathway, which, via implication of PTCH1, SMO, GLI, and SUFU, acts as a transduction enhancer.Citation39 The genetic events underlying SHH pathway activation are age-dependent: in infants, germline mutations in PTCH1 (Gorlin syndrome) or SUFU are frequent. Interestingly, in patients with Gorlin syndrome, RT should be avoided because of the major risk of radiation-induced second cancers (mostly meningiomas and basal cell carcinomas).Citation40 Furthermore, infants with SHH medulloblastoma present an excellent prognosis, even with a CT-only regimen. Children between 3 and 16 years mostly present somatic mutations in PTCH1 or germline (or less frequently somatic) mutations in TP53 (Li Fraumeni Syndrome).Citation41 Thus, all pediatric SHH tumors should be referred to the geneticist to diagnose a potential Gorlin syndrome, Li Fraumeni syndrome, or germ-line SUFU mutation. Somatic TP53 mutations frequently co-occur with GLI2 and MYCN amplifications,Citation41 which induce the activation of the SHH pathway. The TP53 mutation, present in ~30% of SHH medulloblastomas, is related to a very poor prognosis with a 5-year OS of 40%.Citation42 Hence, the fourth edition of the WHO Classification of Tumors of the CNS separates SHH medulloblastoma with or without TP53 mutation. It is well known that cells that express TP53 mutations are less radiosensitive,Citation43 and interestingly, Tchelebi et alCitation44 suggested that RT could even increase tumor growth in medulloblastomas with TP53 mutations.Citation45 In adults, the most frequent mutations are somatic mutations in PTCH1, SMO, and the TERT promoter, or occasionally in IDH1.

Group 3 medulloblastoma represents 25% of all medulloblastomas and has a particularly bad prognosis, with a 5-year OS of 58% in childrenCitation29 and an even poorer OS in nonirradiated infants (5-year OS of 45%).Citation29,Citation41 Tumors present a predominantly classical histology, but this group also has a high ratio of large cell/anaplastic histology (40%), especially in infants.Citation28,Citation46 Group 3 medulloblastomas occur mostly in males (2:1) and in subjects under 16 years of age and derive from cerebellar stem cells.Citation47 Unlike the WNT and SHH subgroups, in which a misfunctioning molecular pathway has clearly been identified, the underlying cause is not well defined in Group 3 medulloblastomas. Recurrent genetic events have been identified: MYC amplification (10% to 20% of Group 3), OTX2 amplification, SMARCA4 mutation, GFI1 enhancer activation,Citation48 isochromosome 17q (42%),Citation29,Citation49,Citation50 gain of 1q (35%),Citation29 gain of chromosome 7 (55%),Citation29 loss of 8p (33%) or gain of 8q (22%),Citation29 loss of 10q (49%),Citation29 gain of 12q (17%),Citation29 loss of 16q (50%),Citation29 and gain of chromosome 18 (26%).Citation29 Isochromosome 17q, as well as MYC amplification, confers a particularly poor prognosis, with a 5-year OS of 20%.Citation26 Group 3 medulloblastoma has a great capacity for metastatic dissemination since 40%–45% have leptomeningeal dissemination at diagnosis and the recurrence pattern is mostly metastatic.Citation51

Group 4 medulloblastoma, although the most frequent (35% of all medulloblastomas), is the least understood of all molecular subgroups.Citation52 This subgroup mostly presents a classical histology and occurs at all ages with a major masculine predominance (3:1).Citation52 The clinical outcome of Group 4 medulloblastoma is intermediate, with a 5-year OS of 75–90%,Citation5,Citation6 but is poor in infants who cannot benefit from RT. Overall, 30–40% of Group 4 cases are metastatic at diagnosis.Citation53 For group 4, no underlying cause has been well defined. Isochromosome 17q is frequent in Group 4 tumors (loss of 17 p 63%, gain of 17q 73%),Citation29 although, unlike in Group 3 tumors, this abnormality does not confer poor prognosis in this subgroup.Citation49 Other recurrent genetic events have been identified: MYCN or CDK6 amplification, SNCAIP duplication, loss of one X chromosome in women, and inactivating the mutation of KDM6A (10% of Group 4),Citation32 chromosome 7 gain (47%),Citation29 8p loss (41%),Citation29 10q loss (15%),Citation29 12q gain (20%),Citation29 chromosome 18 gain (16%),Citation29 and loss of chromosome 11, which is a favorable prognostic marker.Citation41,Citation49

Metastatic medulloblastoma tumor cells harbor molecular alterations that are not present in the primary tumor. Further preclinical work must be performed to identify the molecular mechanisms underlying metastases,Citation54 which remain largely unknown. A recent study involving a deep proteome analysis of metastatic medulloblastomas identified ~1,400 significantly altered proteins between primary and metastatic cell lines, including known factors such as placental growth factor, LIM homeobox 1, prominin 1, and secreted protein acidic and rich in cysteine.Citation55 Additional analysis of clinical medulloblastoma samples implicated yes-associated protein 1 as a potential key factor contributing to metastasis.Citation55

Thus, although molecular subgrouping has revolutionized medulloblastoma classification, there is great heterogeneity within subgroups, and much research is needed to improve the molecular understanding of medulloblastoma.Citation56Citation58

In addition to the lack of knowledge, an international consensus defining the four molecular subgroups of medulloblastoma has recently been published and included in the revised fourth edition of the WHO Classification of Tumors of the CNS.Citation13

Recent studies have recommended more accurate molecular screening. Better characterization of medulloblastoma subgroups is especially important for Group 3 and Group 4 since, as discussed above, the transcriptomes of Group 3 and Group 4 medulloblastomas are similar, and several cytogenetic features, such as isochromosome 17q (i17q), are found in both groups. However, the outcomes of Group 3 and Group 4 medulloblastoma patients differ, particularly regarding the tendency of metastatic dissemination. In 2017, through the analysis of genome-wide DNA methylation and gene expression data by using the similarity network fusion method, Cavalli et alCitation56 suggested that clinically and biologically relevant subtypes exist for each subgroup. They concluded that the four molecular subgroups could be further split into 12 different subtypes that differ on a molecular, clinical, and prognostic basis (WNT: 2, SHH: 4, Group 3: 3, Group 4: 3). Similarly, in 2017, Northcott et alCitation57 analyzed the somatic landscape across 491 sequenced medulloblastoma samples and the molecular heterogeneity among 1,256 epigenetically analyzed cases and discovered new tumor subtypes enriched for specific genetic and transcriptional signatures, especially those of Group 3 and Group 4. In 2017, Schwalbe et alCitation58 conducted molecular profiling analyses of 428 primary medulloblastoma samples, including a DNA methylation microarray analysis, and identified seven molecular subgroups of childhood medulloblastoma (WNT subgroup remained unchanged, and each remaining consensus subgroup was split in two). These data hold promise that improving disease risk stratification and treating patient subtypes according to their genotype are likely to emerge.

Standard of care

The standard of care for medulloblastomas is currently based on their clinical classification. For all medulloblastomas, treatment is multimodal, and the first step consists of maximal safe resection surgery. Within 30 days after surgery, RT is initiated. This delay is based on two previous trials comparing immediate RT following surgery vs delayed RT, which found significantly lower outcomes when RT was delayed more than 4–6 weeks after surgery.Citation57,Citation58 Conversely, a recent trialCitation60 found decreased 5-year OS when RT was initiated ≤3 weeks after surgery and no adverse impact on OS when RT was initiated after >5 weeks but within 90 days of surgery.

For standard-risk patients, a CSI of 23.4 Gy in 13 fractions, followed by a tumor bed boost to reach 54–55.8 Gy is administered.Citation61Citation63 High-risk patients are irradiated with CSI delivering 36–39.6 Gy in 20–22 fractions, followed by a tumor bed boost to 54–55.8 Gy and, when appropriate, 50 Gy CSI is administered to local sites of metastases.Citation64 RT is normofractionated. At 6 weeks after the end of radiotherapy, the patients are treated with four cycles of high-dose chemotherapy, each of which is followed by stem-cell or bone-marrow rescue.Citation64 Each cycle lasts 4 weeks and comprises the followingCitation64:

  • Day 1: Cisplatin IV 75 mg/m2+ vincristine 1.5 mg/m2 (maximum 2 mg)

  • Days 2 and 3: Cyclophosphamide 2 g/m2

  • Day 5: Infusion of peripheral blood or bone-marrow progenitor cells

  • Day 11: Vincristine 1.5 mg/m2

This treatment scheme, tested in a prospective trial by Gajjar et alCitation64 in 1996, enabled a 5-year OS of 85% (95% CI 75–94) for patients in the average-risk group and 70% (54–84) for those in the high-risk group (P=0·04). The 5-year progression free survival (PFS) was 83% (73–93) and 70% (55–85), respectively (P=0·046).

Notably, many other protocols have been used in clinical trials, for example the “maintenance strategy” from trial HIT’91,Citation65 adapted from the historical “Packer protocol”,Citation66 which consists of vincristine given weekly concomitantly with radiotherapy at 6 weeks after the completion of radiotherapy with eight cycles of lomustine, vincristine, and cisplatin ().

Table 1 Maintenance strategy from trial HIT’91

In infants, treatment consists exclusively of surgery, followed by high-dose CT due to the particularly devastating radiation-induced morbidity at this young age.Citation67

Standard-risk adults are treated, after maximal safe resection surgery, by a normofractionated CSI of 30–36 Gy followed by a tumor bed boost to 54–55.8 Gy. CT is less well tolerated in adults than in children, and no randomized clinical trial has demonstrated its benefit. Thus, CT is currently only delivered in high-risk adults before RT and in metastatic patients, CT is delivered after RT.Citation68 The molecules used in CT are cyclophosphamide, etoposide, and cisplatin.Citation68 Approximately 30% of medulloblastoma patients are diagnosed with metastasis;Citation69 however, no gold standard treatment has been highlighted for metastatic medulloblastomas. In the trial HIT’91, children with medulloblastoma were randomized to receive either postoperative chemotherapy followed by CSI (“sandwich strategy”) or postoperative CSI followed by chemotherapy (“maintenance strategy”).Citation59,Citation65 The study showed significantly higher OS after maintenance than that after sandwich treatment for M0 and M1 patients, and a moderate trend toward better survival for children with M2/M3 disease who were treated with the “sandwich strategy” compared with the “maintenance strategy”.Citation65 Therefore, a sandwich concept was chosen for the HIT 2000 trial, which was designed to assess an intensified treatment of metastatic medulloblastoma in children and adolescents (4–21 years).Citation70 Compared with the HIT’91 sandwich regimen, the treatment was intensified at the level of neoadjuvant chemotherapy, of radiotherapy – via the introduction of hyperfractionated CSI, to achieve a biologically more effective dose and preserve normal tissue-,Citation71 and the addition of maintenance chemotherapy. As a result, the treatment consisted of two cycles of induction CT, starting 2–4 weeks after surgery, and comprised intravenous cyclophosphamide, vincristine, methotrexate, carboplatin, etoposide, and concomitant intraventricular methotrexate. Radiotherapy, starting at 3–6 weeks after the end of induction chemotherapy, was hyperfractionated with two fractions of 1 Gy per day, and the doses were 40 Gy CSI in combination with 20 Gy on the posterior fossa, 8 Gy on the tumor site bed, 10 Gy on spinal metastases, and 28 Gy on supratentorial metastases. Maintenance CT started at 6 weeks after the end of RT and consisted of four cycles of cisplatin, lomustine, and vincristine.Citation70 OS was superior with this treatment regimen compared with that in the preceding HIT’91 trial, with a 5-year OS of 74% (95% CI, 66–82).Citation70 Independent risk factors were histology (large cell/anaplastic) and nonresponse to the first chemotherapy cycle. Survival rates were different between molecular subgroups: WNT, SHH, Group 4, and Group 3 with or without MYCC/MYCN amplification (P<0.001). Thus, this study showed that molecular subgroup, MYCC/MYCN status, response to induction chemotherapy, and histologic subtype may improve treatment stratification.Citation70 Regarding adult metastatic medulloblastomas, a trial comparing the sandwich strategy from the HIT 2000 protocol (postoperative CT, hyperfractionated CSI, and maintenance CT) with the HIT’91 maintenance strategy (postoperative CSI and maintenance CT) found that after a 4-year follow-up, the patients showed a global PFS and OS of 52% and 91%, respectively, with no significant difference between the two treatment arms.Citation72

Regarding refractory/recurrent medulloblastoma, temozolomide has been shown to be an effective agent, achieving a 6-month PFS and OS of 30% and 42.5%, respectively.Citation73

Side effects

The current treatment of medulloblastoma allows decent survival rates but often at the expense of life-long morbidity. Iatrogenic morbidity occurs in an age-dependent manner; in older patients, the greatest toxicity results from surgery, while in younger patients, CSI confers troublesome morbidity.Citation20

Major postsurgical morbidities are due to critical structures – particularly brainstem – close or adherent to medulloblastoma. Moreover, cerebellar mutism (also called posterior fossa syndrome) occurs in 25% of cases.Citation20 This neurological syndrome develops within 1–4 days following posterior fossa surgeryCitation74 and consists of speech reduction, axial hypotonia, ataxia, and emotional lability. Cerebellar mutism takes weeks to months to fade away, although speech troubles can last lifelong.Citation75 The precise etiology of cerebellar mutism is unknown, but compared to other posterior fossa tumors, this condition is overrepresented after medulloblastoma surgery.Citation76 The size and wholeness of resection seem to have an impact on postsurgical morbidities since overall neurological morbidity is 24%, whereas morbidity after gross total resection (GTR) is 44%.Citation77,Citation78 Other studies have shown that the incidence of posterior fossa syndrome is more important than less aggressive resections after GTR.Citation76,Citation79

Considering RT, acute toxicity consists mostly of anorexia and nausea, and particular attention must be paid to the nutritional condition of the patient. Additionally, in most children, the bone marrow of the vertebral bodies actively contributes to hematopoiesis; therefore, cytopenia must be detected, followed by pneumocystis prophylaxis during and in the weeks following CSI.Citation20,Citation80 Considering long-term side effects, the toxicity of RT is more important when the patient is young. Morbidity mostly consists of cognitive decline, inversely proportional to the age of the patient, with a reduction in the IQ score as high as 40 points in the youngest patients.Citation8,Citation10 Attempts have been made to decrease cognitive decline through RT hyperfractionation, but the results were not conclusive.Citation21 The risk of radio-induced malignancy, especially meningioma and glioblastoma, is also high.Citation10,Citation81Citation83 Other studies have observed endocrine dysfunction, bone growth and development dysfunctions, gynecological, cardiac and pulmonary toxicity, ototoxicity,Citation10 and vascular toxicity with an increased frequency of stroke.Citation84

Regarding CT, a common adverse effect is vincristine-induced peripheral neuropathy (VIPN), which affects sensory, motor, and autonomic nerves. VIPN incidence rates reach 37% during medulloblastoma treatment, and symptoms may not resolve over time.Citation59 Chronic peripheral neuropathy favors physical activity decrease, obesity, type 2 diabetes mellitus, metabolic syndrome, and cardiovascular disease.Citation85 Other toxicities attributed to CT are mainly cisplatin-induced ototoxicity (incidence rates of 34% reaching grade III/IV in 9% of patients) and myelosuppression.Citation59

Optimizing care from molecular classification

The current challenge of the neuro-oncologist is precision medicine. To limit iatrogenic morbidity, we must first avoid the overtreatment of patients with good prognosis who do not need aggressive treatments or, in contrast, patients for whom standard treatments have shown failure to induce good survival rates. Then, new therapeutic approaches must be developed to improve survival rates in treatment failure patients.

This perspective requires a reliable method for the prognostic sorting of medulloblastomas. A consensus conference in Heidelberg in 2015 concluded an updated prognostic classification for children with medulloblastomas based on molecular subgroups,Citation5,Citation6 as shown in . However, clinical applications face technical difficulties in finding a reliable and easy method for the molecular statement of resected tumors. A recently published study proposes a robust and repeatable molecular classification method based on six epigenetic biomarkers.Citation86 Elsewhere, emerging strategies such as DNA methylation profiling using a comprehensive machine-learning approach enable robust and reproducible classification of central nervous system tumors.Citation87 This method may have a substantial impact on diagnostic precision compared to standard methods, by reducing the substantial inter-observer variability observed in current CNS tumor diagnostics. A uniform implementation of the classification algorithm holds great promise for standardization of tumor diagnostics across centers and across clinical trials.

Table 2 Prognostic classification for pediatric medulloblastomas according to molecular subgroups

Systemic therapeutic approaches

Overexpression of WNT pathway results in a weaker blood–brain barrier, through vascular dysfunction induced by overexpression of this molecular pathway. A weaker blood–brain barrier enables better penetration of CT molecules into cancer cells. This mechanism could contribute to the good prognosis of this molecular subgroup.Citation88 Hence, targeted therapy inhibiting the WNT pathway could make the tumor less chemosensitive. Furthermore, the WNT pathway participates in many physiological functions, such as bone formation, and inhibiting the WNT pathway would lead to significant toxicity, such as osteoporosis.Citation89 For these reasons, no development of targeted therapy has been undertaken for WNT medulloblastomas, and trials rather focus on decreasing the doses of RT and CT. Several trials are in progress evaluating lower doses of radiation and CT (NCT01878617, NCT02724579) or CT-only approaches (NCT02212574).

Efforts have been made to develop targeted therapies inhibiting the SHH pathway in this subgroup with intermediate prognoses. One therapeutic approach is SMO inhibition with vismodegibCitation90 (currently used for the treatment of locally advanced/metastatic basal cell carcinoma). However, only SHH medulloblastomas with mutations in PTCH1 (upstream SMO) or SMO can benefit from this molecule. A study confirmed that SHH medulloblastoma with SUFU or GLI1 (downstream SMO) mutations do not respond to vismodegib.Citation45 Since 80% of adult SHH medulloblastoma patients have PTCH1 or SMO mutations, vismodegib is likely to be particularly advantageous in this population.Citation91 Moreover, SMO inhibitors could lead to premature bone fusion in children.Citation41 A more recent therapeutic approach refers to epigenetic treatments with bromodomain (BET) inhibitors. BET proteins regulate gene transcription by binding to acetylated histones.Citation92 BET inhibitors have been shown in vitro and in vivo to decrease cell viability and proliferation in SHH medulloblastoma,Citation93,Citation94 but no clinical trial has yet examined BET inhibitors as potential therapeutics for SHH medulloblastoma. Considering metastatic SHH medulloblastoma, for which MET kinase is a marker, the MET inhibitor foretinib has been shown to decrease tumor cell proliferation and induce apoptosis in vitro and in vivo, which confers a strong rationale for its clinical evaluation.Citation95 A clinical trial is ongoing to evaluate doublet therapy comprising the CDK4/6 inhibitor ribociclib with either gemcitabine, trametinib, or sonidegib in adults with refractory or recurrent SHH medulloblastoma (NCT03434262). Clinical trials are also ongoing to evaluate vismodegib in children and adults with refractory or recurrent SHH medulloblastoma (NCT01601184, NCT00939484, NCT01239316).

For Group 3 patients who show poor outcomes when treated with the current standard of care, special expectations rest on the development of targeted therapies. Due to the particularly poor outcome conferred by MYC amplification, efforts have been made concerning MYC inhibition, but no direct MYC inhibitor could successfully be finalized since MYC has no clearly defined ligand-binding domain. A preclinical study designed based on a mouse model of Group 3 medulloblastoma showed the efficacy of palbociclib – a CDK4/6 inhibitor currently used in HR+/Her2– breast tumors, locally advanced or with bone metastases,Citation96 and a clinical trial is in progress for evaluation of palbociclib in pediatric brain tumors (NCT02255461). Another clinical trial showed the efficacy of using HDAC with a PI3K inhibitor in Group 3 medulloblastoma with MYC amplification.Citation97 BET inhibitors also represent a significant therapeutic approach for treating Group 3 medulloblastoma, since BRD4 (a member of BET family) inhibitors have been shown in vitro and in vivo in a mouse model to be an effective therapy against MYC-amplified Group 3 medulloblastoma,Citation98 and the BET inhibitor JQ1 has been demonstrated to reduce tumoral cell viability through the inhibition of MYC transcription.Citation99 However, JQ1 has a very short half-life, which does not allow clinical practice. A phase one clinical trial is ongoing to test other BET inhibitors (CPI-0610, MK-8628) in adults.Citation52 Nevertheless, epigenetic targeting molecules are often found to operate with a cytostatic effect, and this treatment is likely to require a combination of cytostatic and cytotoxic drugs to procure antitumoral efficiency.

Although the most frequent, Group 4 medulloblastoma is the most heterogeneous and least understood medullo-blastoma type. No dominant oncogene has been successfully identified, which constitutes a major limitation in the development of targeted therapies. Some studies have suggested a molecular mechanism involving the activation of NFkB.Citation27,Citation100

Another ongoing therapeutic approach is targeting medulloblastoma stem cells, which are a subpopulation of cancer cells largely responsible for medulloblastoma initiation, maintenance, dissemination, and relapse.Citation47 Research efforts are still needed to effectively target medulloblastoma stem cells.

Surgery optimization

In the current clinical classification, a postsurgical residual disease >1.5 cm2 is a marker of worse prognosis and distinguishes the patient as high risk, hence requiring higher CSI doses. However, considering the important side effects of CSI and surgery, the prognostic implication of the extent of resection (EOR) is worth updating, now that the prognostic classification has been revised as a result of molecular advances. Indeed, experts highlighted this issue at a consensus conference in 2016.Citation5 A recent review analyzed 50 articles about the implication of EOR in clinical outcome,Citation101 showing a nearly equal number of studies with and without a significant association between EOR and survival. Only three of these articles accounted for molecular subgrouping,Citation31,Citation58,Citation102 and no association was found between EOR and survival when molecular subgrouping was considered. To make a reliable determination of the prognostic implication of EOR, it would be worth setting up a prospective trial of patients with residual disease >1.5 cm2 and randomizing these patients into 24 Gy CSI vs 36 Gy CSI to assess whether intensified CSI can improve the disease control of patients with a residual disease >1.5 cm2. Subsequently, molecular stratification would be useful. Thompson et alCitation101 showed that such a study would require 2,890 patients with residual disease >1.5 cm2 to detect a difference with 90% power, unfortunately making stratification technically not feasible, particularly in a subgroup-stratified approach.

Radiotherapy optimization

The survival of medulloblastoma patients improved tremendously in 1950 due to the introduction of CSI, since long-term survival evolved from nearly 0% to 50%.Citation103 The development of a linear accelerator with megavoltage (Linac) and 3D conformational RT (3D-CRT) allowed the establishment of CSI due to better precision and the higher intensity and penetrance of the radiations.Citation104 Since this major advance, many inroads have been made in the RT field, and we will discuss how medulloblastoma irradiation could be optimized.

The randomized multicenter HIT-SIOP PNET 4 trial was initiated to estimate the amelioration of radiation-induced toxicity related to hyperfractionation. The hyperfractionated arm received 36 Gy in 36 fractions of CSI, with an additional 24 Gy in 24 fractions on the posterior fossa and 8 Gy in 8 fractions on the tumor bed, and the standard RT arm received 23.4 Gy in 13 fractions of CSI with an additional 32.4 Gy in 18 fractions on the posterior fossa. The results revealed no significant benefit of hyperfractionation in toxicity or in survival,Citation21 and normofractionation remains the standard of care.

The randomized COG ACNS0331 clinical trial compared survival in the arm with a standard dose of RT (23.4 Gy CSI and 54 Gy posterior fossa RT) vs a decreased dose of RT (18 Gy CSI and a reduction in boost volume). The results showed no significant difference in 5-year OS or PFS for a decrease in radiation boost volume, but concerning CSI, noninferiority of lower-dose CSI to standard dose CSI was not established.Citation105 Thus, CSI seems to be a crucial component of medulloblastoma treatment, and doses do not appear to be reducible without affecting survival. However, in light of molecular understanding, it is legitimate to question whether CSI can be reduced or more importantly removed in some patients. Indeed, WNT patients rarely relapse, and trials are already ongoing to determine the feasibility of irradiation de-escalation (NCT01878617, NCT02724579) or removal (NCT02212574). Considering SHH medulloblastoma, the pattern of relapse has been shown to be predominantly local.Citation51 Consequently, a future clinical trial could focus on establishing whether it is feasible to reduce or even remove CSI and focus RT on the tumor bed in SHH patients. Group 3 and Group 4 medulloblastomas present a significant proportion of metastatic relapse; therefore, CSI is unlikely to be removed in those patients. However, future studies could focus on optimizing the radiation dose distribution to maintain efficiency while lowering toxicity.

Since the 2000s, the development of intensity-modulated irradiation modalities, such as intensity-modulated radiotherapy (IMRT), volumetric-modulated arc therapy, and tomotherapy, have enabled the optimization of radiation dose distribution in CSI. However, those modalities involved more radiation fields and therefore induced a larger volume of normal tissues exposed to low-dose radiation, hence increasing the risk of second neoplasms.Citation106

Several studies suggested an association between the radiation dose delivered to the hippocampus and temporal lobes and neurocognitive decline,Citation107,Citation108 and the feasibility of a hippocampal sparing approach with IMRT modality has been studied. The results revealed a significant amount of perihippocampal relapse among patients with brain metastases at the time of diagnosis. In contrast, no patient without brain metastases at diagnosis developed secondary lesions in the perihippocampal area.Citation109 Hippocampal sparing may thus be considered in high-risk medulloblastoma patients without brain metastases at diagnosis and, by extension, this approach is likely to be safely feasible in standard-risk patients.

In the last decade, the use of proton therapy has rapidly increased as a result of its capacity to better spare organs at risk by eliminating the exit radiation dose due to the characteristic dose distribution of the proton beam modeled by the Bragg peak. This treatment is particularly relevant in childhood malignancies since it offers the promise of decreased late radiation-related morbidities, especially second neoplasms.Citation110 Medulloblastoma, specifically due to a particularly large irradiation field, is an excellent candidate for proton therapy. Indeed, protons eliminate the dose of exit radiation into the chest, abdomen, and pelvis as well as the cochlea, pituitary, and hypothalamus of children after CSI.Citation110 Translation into quality of life (QoL) has been studied in a prospective trial.Citation111 QoL scores were found to improve over time after proton CSI, and after 5 years, children-reported scores were statistically similar to those of healthy children, but the parent-reported scores remained statistically lower than those reported by the parents of healthy children.Citation112 To evaluate the superiority of proton therapy in medulloblastoma treatment with an evidence-based approach, a review recently compared the outcomes of pediatric medulloblastoma patients between proton- and photon-mediated CSI,Citation113 and revealed the advantage of proton therapy in organs at risk sparing, normal organ dysfunction, and secondary malignancy risks compared to various (mostly 3D-CRT) photon techniques. A comparison of target coverage between both radiation modalities showed either similar or better results with proton therapy. However, proton therapy is a modern radiation modality, and the earliest study considered in this review was from 1997. For that reason, data regarding late toxicity after proton therapy are not available. On the other hand, we cannot ignore that second neoplasms after CSI mostly occur in the neuraxis, and this effect cannot be avoided with any irradiation modality as long as CSI is performed. The only way to determine with any certainty whether proton therapy should be developed as a standard of care for CSI would be through a prospective randomized controlled trial comparing both treatment modalities. Such a trial should include cost-effectiveness analysis since proton therapy is undoubtedly associated with higher initial infrastructural costs than those for photon therapy.Citation112 At the present time, proton therapy remains a limited resource, and socioeconomic factors impact access to this treatment.Citation113

Overview and prospects

summarizes the current molecular understanding of medulloblastoma.

Table 3 Overview summary of current molecular understanding of medulloblastoma

Although molecular subgrouping for medulloblastoma is important for the prognosis and elaboration of therapeutic agents, this categorization seems insufficient for Group 3 (which undergoes worse prognosis) and Group 4 (which is the most frequent) medulloblastomas. As shown in , those two subgroups lack targeting agents. Hashimoto et alCitation114 most recently published the results of 36 extensively profiled medulloblastomas. The results revealed the high expression of MRP1, TUBB3, PTEN, TOP2A, thymidylate synthase, RRM1, and TOP1. This finding highlights an all-new therapeutic prospect since targeting agents are available for several of these targets.

Conclusion

Recent advances on the molecular mechanisms of medulloblastoma have allowed the definition of an updated prognostic classification. To optimize medulloblastoma care, efforts must be made to reduce iatrogenic morbidity and improve survival in patients with lower prognoses. Thus, targeted therapies are currently being evaluated in light of a molecular understanding of medulloblastomas. Given the relatively low incidence of medulloblastoma, every oncologist should be particularly aware of the importance of including patients in clinical trials.

RT leads to a significant proportion of the late-onset toxicity observed in medulloblastoma survivors, mainly due to CSI, which is a crucial component of the multimodal treatment. With recent molecular advances, the feasibility of reducing or even removing CSI is currently being evaluated in WNT medulloblastoma, and it would be interesting to study the feasibility of CSI reduction/removal in SHH patients (who present mostly a local pattern of relapse) in a future clinical trial. The necessity of intensifying CSI for all patients with postsurgical residual disease >1.5 cm2 is questionable. In any case where CSI must be maintained (which remains prevalent), the potential solutions to lower radiation-induced morbidity are hippocampal sparing and proton therapy.

Disclosure

The authors report no conflicts of interest in this work.

References

  • TaillandierLBlonskiMCarrieCLes médulloblastomes: revue générale [Medulloblastomas: review]Rev Neurol2011167543144821529869
  • OstromQTGittlemanHLiaoPCBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007–2011Neuro Oncol201416suppl 4iv1iv6325304271
  • JohnsonKJCullenJBarnholtz-SloanJSChildhood brain tumor epidemiology: a Brain Tumor Epidemiology Consortium ReviewCanc Epidemiol Biomarkers Prev2014231227162736
  • FellayCNFrappazDSunyachMPFranceschiEBrandesAAStuppRMedulloblastomas in adults: prognostic factors and lessons from paediatricsCurr Opin Neurol201124662663222027544
  • RamaswamyVRemkeMBouffetERisk stratification of childhood medulloblastoma in the molecular era: the current consensusActa Neuropathol2016131682183127040285
  • RamaswamyVRemkeMAdamskiJMedulloblastoma subgroup-specific outcomes in irradiated children: who are the true high-risk patients?Neuro Oncol201618229129725605817
  • TarbellNJFriedmanHPolkinghornWRHigh-risk medulloblastoma: a pediatric oncology group randomized trial of chemotherapy before or after radiation therapy (POG 9031)J Clin Oncol201331232936294123857975
  • Moxon-EmreIBouffetETaylorMDImpact of craniospinal dose, boost volume, and neurologic complications on intellectual outcome in patients with medulloblastomaJ Clin Oncol201432171760176824516024
  • Moxon-EmreITaylorMDBouffetEIntellectual outcome in molecular subgroups of medulloblastomaJ Clin Oncol201634344161417027507873
  • FossatiPRicardiUOrecchiaRPediatric medulloblastoma: toxicity of current treatment and potential role of protontherapyCancer Treat Rev2009351799618976866
  • EllisonDWOniludeOELindseyJCBeta-catenin status predicts a favorable outcome in childhood medulloblastoma: the United Kingdom Children’s Cancer Study Group Brain Tumour CommitteeJ Clin Oncol200523317951795716258095
  • HermsJNeidtILüscherBC-MYC expression in medulloblastoma and its prognostic valueInt. J. Cancer200089539540211008200
  • LouisDNPerryAReifenbergerGThe 2016 World Health Organization classification of tumors of the central nervous system: a summaryActa Neuropathol2016131680382027157931
  • EllisonDWDaltonJKocakMMedulloblastoma: clinicopathological correlates of SHH, WNT, and non-SHH/WNT molecular subgroupsActa Neuropathol2011121338139621267586
  • EllisonDWChildhood medulloblastoma: novel approaches to the classification of a heterogeneous diseaseActa Neuropathol2010120330531620652577
  • GiangasperoFRigobelloLBadialiMLarge-cell medulloblastomas. A distinct variant with highly aggressive behaviorAm J Surg Pathol19921676876931530108
  • LouisDNOhgakiHWiestlerODThe 2007 WHO classification of tumours of the central nervous systemActa Neuropathol200711429710917618441
  • ZeltzerPMBoyettJMFinlayJLMetastasis stage, adjuvant treatment, and residual tumor are prognostic factors for medulloblastoma in children: conclusions from the Children’s Cancer Group 921 randomized phase III studyJ Clin Oncol199917383284510071274
  • ChangCHHousepianEMHerbertCAn operative staging system and a megavoltage radiotherapeutic technic for cerebellar medulloblastomasRadiology1969936135113594983156
  • MartinAMRaabeEEberhartCCohenKJManagement of pediatric and adult patients with medulloblastomaCurr Treat Options in Oncol2014154581594
  • LanneringBRutkowskiSDozFHyperfractionated versus conventional radiotherapy followed by chemotherapy in standard-risk medulloblastoma: results from the randomized multicenter HIT-SIOP PNET 4 trialJ Clin Oncol201230263187319322851561
  • PackerRJGajjarAVezinaGPhase III study of craniospinal radiation therapy followed by adjuvant chemotherapy for newly diagnosed average-risk medulloblastomaJ Clin Oncol200624254202420816943538
  • PomeroySLTamayoPGaasenbeekMPrediction of central nervous system embryonal tumour outcome based on gene expressionNature2002415687043644211807556
  • ThompsonMCFullerCHoggTLGenomics identifies medulloblastoma subgroups that are enriched for specific genetic alterationsJ Clin Oncol200624121924193116567768
  • KoolMKosterJBuntJIntegrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological featuresPLoS One200838e308818769486
  • ChoY-JTsherniakATamayoPIntegrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcomeJ Clin Oncol201129111424143021098324
  • NorthcottPAKorshunovAWittHMedulloblastoma comprises four distinct molecular variantsJ Clin Oncol201129111408141420823417
  • TaylorMDNorthcottPAKorshunovAMolecular subgroups of medulloblastoma: the current consensusActa Neuropathologica2012123446547222134537
  • KoolMKorshunovARemkeMMolecular subgroups of medulloblastoma: an international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, group 3, and group 4 medulloblastomasActa Neuropathol2012123447348422358457
  • CliffordSCLanneringBSchwalbeECBiomarker-driven stratification of disease-risk in non-metastatic medulloblastoma: results from the multi-center HIT-SIOP-PNET4 clinical trialOncotarget2015636388273883926420814
  • PietschTSchmidtRRemkeMPrognostic significance of clinical, histopathological, and molecular characteristics of medulloblastomas in the prospective HIT2000 multicenter clinical trial cohortActa Neuropathol2014128113714924791927
  • NorthcottPAJonesDTWKoolMMedulloblastomics: the end of the beginningNat Rev Cancer2012121281883423175120
  • RobinsonGParkerMKranenburgTANovel mutations target distinct subgroups of medulloblastomaNature20124887409434822722829
  • HamiltonSRLiuBParsonsREThe molecular basis of Turcot’s syndromeN Engl J Med1995332138398477661930
  • TrubickaJSzperlMGrajkowskaWIdentification of a novel inherited ALK variant M1199L in the WNT type of medulloblastomaFolia Neuropathol2016112330
  • NorthcottPAKorshunovAPfisterSMTaylorMDThe clinical implications of medulloblastoma subgroupsNat Rev Neurol20128634035122565209
  • EllisonDWKocakMDaltonJDefinition of disease-risk stratification groups in childhood medulloblastoma using combined clinical, pathologic, and molecular variablesJ Clin Oncol201129111400140720921458
  • GibsonPTongYRobinsonGSubtypes of medulloblastoma have distinct developmental originsNature201046873271095109921150899
  • TaylorMDLiuLRaffelCMutations in Sufu predispose to medulloblastomaNat Genet200231330631012068298
  • O’MalleySWeitmanDOldingMSekharLMultiple neoplasms following craniospinal irradiation for medulloblastoma in a patient with nevoid basal cell carcinoma syndrome. Case reportJ Neurosurg19978622862889010431
  • RamaswamyVTaylorMDMedulloblastoma: from myth to molecularJCO2017352123552363
  • ZhukovaNRamaswamyVRemkeMSubgroup-specific prognostic implications of TP53 mutation in medulloblastomaJ Clin Oncol201331232927293523835706
  • WilliamsJRZhangYZhouHA quantitative overview of radiosensitivity of human tumor cells across histological type and TP53 statusInt J Radiat Biol200884425326418386191
  • TchelebiLAshamallaHGravesPRMutant p53 and the response to chemotherapy and radiationSubcell Biochem20148513315925201193
  • KoolMJonesDTWJägerNGenome sequencing of Shh medulloblastoma predicts genotype-related response to smoothened inhibitionCancer Cell201425339340524651015
  • ColucciaDFiguereidoCIsikSSmithCRutkaJTMedulloblastoma: tumor biology and relevance to treatment and prognosis paradigmCurr Neurol Neurosci Rep20161654327021772
  • HuangG-HXuQ-FCuiY-HLiNBianX-WLvS-QMedulloblastoma stem cells: promising targets in medulloblastoma therapyCancer Sci2016107558358927171351
  • NorthcottPALeeCZichnerTEnhancer hijacking activates Gfi1 family oncogenes in medulloblastomaNature2014511751042843425043047
  • ShihDJHNorthcottPARemkeMCytogenetic prognostication within medulloblastoma subgroupsJ Clin Oncol201432988689624493713
  • ZhouLPicardDRaY-SSilencing of thrombospondin-1 is critical for Myc-induced metastatic phenotypes in medulloblastomaCancer Res201070208199821020876797
  • RamaswamyVRemkeMBouffetERecurrence patterns across medulloblastoma subgroups: an integrated clinical and molecular analysisLancet Oncol201314121200120724140199
  • ArcherTCMahoneyELPomeroySLMedulloblastoma: molecular classification-based personal therapeuticsNeurotherapeutics201714226527328386677
  • RamaswamyVRemkeMShihDDuration of the pre-diagnostic interval in medulloblastoma is subgroup dependentPediatr Blood Cancer20146171190119424616042
  • Miranda Kuzan-FischerCJuraschkaKTaylorMDMedulloblastoma in the molecular eraJ Korean Neurosurg Soc201861329230129742881
  • GuSChenKYinMWuZWuYProteomic profiling of isogenic primary and metastatic medulloblastoma cell lines reveals differential expression of key metastatic factorsJ Proteomics2017160556328363815
  • CavalliFMGRemkeMRampasekLIntertumoral heterogeneity within medulloblastoma subgroupsCancer Cell201731673775428609654
  • NorthcottPABuchhalterIMorrissyASThe whole-genome landscape of medulloblastoma subtypesNature2017547766331131728726821
  • SchwalbeECLindseyJCNakjangSNovel molecular subgroups for clinical classification and outcome prediction in childhood medulloblastoma: a cohort studyLancet Oncol201718795897128545823
  • KortmannR-DKühlJTimmermannBPostoperative neo-adjuvant 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’91Int J Radiat Oncol Biol Phys200046226927910661332
  • ChinALModingEJDonaldsonSSSurvival impact of postoperative radiotherapy timing in pediatric and adolescent medulloblastomaNeuro Oncol20182081133114129309676
  • PackerRJGoldweinJNicholsonHSTreatment of children with medulloblastomas with reduced-dose craniospinal radiation therapy and adjuvant chemotherapy: a children’s cancer group studyJ Clin Oncol19991772127213610561268
  • MerchantTEKunLEKrasinMJMulti-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 medulloblas-tomaInt J Radiat Oncol Biol Phys200870378278717892918
  • WahbaHAAbu-HegazyMWaselYIsmailEIZidanASAdjuvant chemotherapy after reduced craniospinal irradiation dose in children with average-risk medulloblastoma: a 5-year follow-up studyJ BUON201318242542923818356
  • GajjarAChintagumpalaMAshleyDRisk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (ST Jude Medul-loblastoma-96): long-term results from a prospective, multicentre trialLancet Oncol200671081382017012043
  • von HoffKHinkesBGerberNULong-term outcome and clinical prognostic factors in children with medulloblastoma treated in the prospective randomised multicentre trial HIT’91Eur J Cancer20094571209121719250820
  • PackerRJSuttonLNGoldweinJWImproved survival with the use of adjuvant chemotherapy in the treatment of medulloblastomaJ Neurosurg1991133433440
  • Lafay-CousinLSmithAChiSNClinical, pathological, and molecular characterization of infant medulloblastomas treated with sequential high-dose chemotherapyPediatr Blood Cancer20166391527153427145464
  • BrandesAAErmaniMAmistaPThe treatment of adults with medulloblastoma: a prospective studyInt J Radiat Oncol Biol Phys200357375576114529781
  • PackerRJRoodBRMacdonaldTJMedulloblastoma: present concepts of stratification into risk groupsPediatr Neurosurg2003392606712845195
  • von BuerenAOKortmannR-Dvon HoffKTreatment of children and adolescents with metastatic medulloblastoma and prognostic relevance of clinical and biologic parametersJ Clin Oncol201634344151416027863192
  • AllenJDonahueBMehtaMA phase II study of Preradiotherapy chemotherapy followed by hyperfractionated radiotherapy for newly diagnosed high-risk medulloblastoma/primitive neuroectodermal tumor: a report from the Children’s Oncology Group (CCG 9931)Int J Radiat Oncol Biol Phys20097441006101119356859
  • von BuerenAOFriedrichCvon HoffKMetastatic medulloblastoma in adults: outcome of patients treated according to the HIT2000 protocolEur J Cancer201551162434244326254812
  • CefaloGMassiminoMRuggieroATemozolomide is an active agent in children with recurrent medulloblastoma/primitive neuroectodermal tumor: an Italian multi-institutional phase II trialNeuro-Oncology201416574875324482446
  • KirkEAHowardVCScottCADescription of posterior fossa syndrome in children after posterior fossa brain tumor surgeryJ Pediatr Oncol Nurs19951241811877495523
  • RobertsonPLMuraszkoKMHolmesEJIncidence and severity of postoperative cerebellar mutism syndrome in children with medulloblastoma: a prospective study by the Children’s Oncology GroupJ Neurosurg2006105644445117184075
  • CeC-Bvan DongenHRMulderPGPazYGeuzeDPaquierPFLequinMHTumour type and size are high risk factors for the syndrome of “cerebellar” mutism and subsequent dysarthriaJ Neurol Neurosurg Psychiatry199967675575710567492
  • AlbrightALSpostoRHolmesECorrelation of neurosurgical subspecialization with outcomes in children with malignant brain tumorsNeurosurgery2000474879887 discussion 885–88711014428
  • Douglas CochraneDGustavssonBPoskittKPSteinbokPKestleJRWThe surgical and natural morbidity of aggressive resection for posterior fossa tumors in childhoodPediatr Neurosurg199420119298142278
  • Gelabert-GonzálezMFernández-VillaJMutism after posterior fossa surgery. Review of the literatureClin Neurol Neurosurg2001103211111411516555
  • GrollAHRitterJMüllerFMGuidelines for prevention of Pneumocystis carinii pneumonitis in children and adolescents with cancerKlin Padiatr2001213Suppl 1A38A4911577363
  • NegliaJPRobisonLLStovallMNew primary neoplasms of the central nervous system in survivors of childhood cancer: a report from the Childhood Cancer Survivor StudyJ Natl Cancer Inst200698211528153717077355
  • PackerRJZhouTHolmesEVezinaGGajjarASurvival and secondary tumors in children with medulloblastoma receiving radiotherapy and adjuvant chemotherapy: results of children’s Oncology Group trial A9961Neurooncology201315197103
  • KotechaRSPascoeEMRushingEJMeningiomas in children and adolescents: a meta-analysis of individual patient dataLancet Oncol201112131229123922094004
  • BansalLRBelairJCummingsDZuccoliGLate-onset radiation-induced vasculopathy and stroke in a child with medulloblastomaJ Child Neurol201530680080225015672
  • Lavoie SmithEMLiLChiangCPatterns and severity of vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemiaJ Peripher Nerv Syst2015201374625977177
  • GómezSGarrido-GarciaAGarcia-GeriqueLA novel method for rapid molecular subgrouping of medulloblastomaClin Cancer Res20182461355136329351917
  • CapperDJonesDTWSillMDNA methylation-based classification of central nervous system tumoursNature2018555769746947429539639
  • PhoenixTNPatmoreDMBoopSMedulloblastoma genotype dictates blood brain barrier phenotypeCancer Cell201629450852227050100
  • ManolagasSCWNT signaling and osteoporosisMaturitas201478323323724815296
  • RobinsonGWOrrBAWuGVismodegib exerts targeted efficacy against recurrent sonic hedgehog–subgroup medulloblastoma: results from phase II pediatric brain tumor consortium studies PBTC-025B and PBTC-032J Clin Oncol201533242646265426169613
  • NorthcottPAHielscherTDubucAPediatric and adult sonic hedgehog medulloblastomas are clinically and molecularly distinctActa Neuropathol2011122223124021681522
  • WuS-YChiangC-MThe double bromodomain-containing chromatin adaptor Brd4 and transcriptional regulationJ Biol Chem200728218131411314517329240
  • TangYGholaminSSchubertSEpigenetic targeting of hedgehog pathway transcriptional output through BET bromodomain inhibitionNat Med201420773274024973920
  • LongJLiBRodriguez-BlancoJThe BET bromodomain inhibitor I-BET151 acts downstream of smoothened protein to abrogate the growth of hedgehog protein-driven cancersJ Biol Chem201428951354943550225355313
  • FariaCCGolbournBJDubucAMForetinib is effective therapy for metastatic sonic hedgehog medulloblastomaCancer Res201575113414625391241
  • HanafordARArcherTCPriceADiSCoVERing innovative therapies for rare tumors: combining genetically accurate disease models with in silico analysis to identify novel therapeutic targetsClin Cancer Res201622153903391427012813
  • PeiYLiuK-WWangJHDAC and PI3K antagonists cooperate to inhibit growth of MYC-driven medulloblastomaCancer Cell201629331132326977882
  • VenkataramanSAlimovaIBalakrishnanIInhibition of BRD4 attenuates tumor cell self-renewal and suppresses stem cell signaling in MYC driven medulloblastomaOncotarget2014592355237124796395
  • BandopadhayayPBergtholdGNguyenBBET bromodomain inhibition of MYC-amplified medulloblastomaClin Cancer Res201420491292524297863
  • NorthcottPAShihDJHPeacockJSubgroup-specific structural variation across 1,000 medulloblastoma genomesNature20124887409495622832581
  • ThompsonEMBramallAHerndonJETaylorMDRamaswamyVThe clinical importance of medulloblastoma extent of resection: a systematic reviewJ Neurooncol2018139352353929796724
  • ThompsonEMHielscherTBouffetEPrognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: a retrospective integrated clinical and molecular analysisLancet Oncol201617448449526976201
  • PatersonEFarrRFCerebellar medulloblastoma: treatment by irradiation of the whole central nervous systemActa radiol195339432333613057640
  • ThariatJHannoun-LeviJ-MSun MyintAVuongTGérardJ-PPastGJ-PPast, present, and future of radiotherapy for the benefit of patientsNat Rev Clin Oncol2013101526023183635
  • MichalskiJMJanssAVezinaGResults of cog ACNS0331: a phase III trial of involved-field radiotherapy (IFRT) and low dose craniospinal irradiation (LD-CSI) with chemotherapy in average-risk medulloblastoma: a report from the Children’s Oncology GroupInt J Radiation Oncol Biol Physics2016965937938
  • HallEJWuuC-SRadiation-induced second cancers: the impact of 3D-CRT and IMRTInt J Radiation Oncol Biol Physics20035618388
  • ArmstrongGTJainNLiuWRegion-specific radiotherapy and neuropsychological outcomes in adult survivors of childhood CNS malignanciesNeurooncology2010121111731186
  • JalaliRMallickIDuttaDFactors influencing neurocognitive outcomes in young patients with benign and low-grade brain tumors treated with stereotactic conformal radiotherapyInt J Radiat Oncol Biol Phys201077497497919864079
  • PadovaniLChaponFAndréNHippocampal sparing during craniospinal irradiation: what did we learn about the incidence of perihippocampus metastases?Int J Radiation Oncol Biol Physics20181004980986
  • LadraMMMacDonaldSMTerezakisSAProton therapy for central nervous system tumors in childrenPediatr Blood Cancer2018657e2704629630784
  • KamranSCGoldbergSIKuhlthauKAQuality of life in patients with proton-treated pediatric medulloblastoma: results of a prospective assessment with 5-year follow-upCancer2018124163390340029905942
  • HoESQBarrettSAMullaneyLMA review of dosimetric and toxicity modeling of proton versus photon craniospinal irradiation for pediatrics medulloblastomaActa Oncol20175681031104228509599
  • ShenCJHuCLadraMMNarangAKCePTerezakisSASocioeconomic factors affect the selection of proton radiation therapy for children: proton therapy and socioeconomic factorsCancer2017123204048405628654202
  • HashimotoYPenas-PradoMZhouSRethinking medulloblastoma from a targeted therapeutics perspectiveJ Neurooncol2018139371372029869738