Next Article in Journal
Effects of a Concurrent Mixed-Modality (Telerehabilitation and Face-to-Face) Exercise Rehabilitation Program in a Patient with Multiple Myeloma Prior to Spinal Cord Transplantation: A Case Study
Previous Article in Journal / Special Issue
Precision Medicine for Pediatric Glioma and NF1-Associated Tumors: The Role of Small Molecule Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Approaches to Reduce Toxicity in Pediatric Brain Tumors

by
Hallie Coltin
1,2,†,
Christina Coleman
3,† and
Chantel Cacciotti
4,*
1
Division of Hematology/Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC H3C 3A7, Canada
2
Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
3
Division of Hematology/Oncology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
4
Division of Hematology/Oncology, Department of Pediatrics, London Health Sciences Centre & Western University, London, ON N6A 5W9, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Oncol. 2025, 32(5), 281; https://doi.org/10.3390/curroncol32050281
Submission received: 14 April 2025 / Revised: 9 May 2025 / Accepted: 13 May 2025 / Published: 15 May 2025
(This article belongs to the Special Issue Clinical Outcomes and New Treatments in Pediatric Brain Tumors)

Abstract

:
Pediatric central nervous system (CNS) tumor survivors are highly susceptible to long-term toxicity due to tumor location and also the treatment received. Advancements in treatment techniques, risk-adapted approaches to therapy with adjustments to treatment regimens—including de-escalation when feasible—along with the addition of supportive therapy and surveillance in these survivors, serve to minimize and manage late effects of therapy.

1. Introduction

Central nervous system (CNS) tumors represent the most common solid tumors of childhood [1,2,3]. Importantly, this disease group is the leading cause of cancer-related morbidity and mortality among children [3]. Survivors of pediatric CNS tumors are highly susceptible to long-term toxicity resulting from tumor location and receiving intensive treatment [4,5,6]. Recent biological advances in tumor classification empowered the pediatric neuro-oncology community to prioritize therapy de-escalation when safe and pursue feasible toxicity mitigation strategies and targeted long-term surveillance for these survivors. In this review, we discuss the most frequent challenges faced by pediatric CNS tumor survivors as well as recent advances to minimize long-term treatment-related sequelae (Figure 1).

2. Ototoxicity

Ototoxicity, defined as hearing loss, tinnitus, and/or vertigo [7], is a frequent and permanent late effect among children diagnosed with CNS tumors [6,8,9,10]. The development of ototoxicity is strongly associated with decreased quality of life, social isolation, adverse mental health outcomes, and educational challenges [11,12,13,14,15,16]. The causes of ototoxicity are multifactorial and include structural damage during neurosurgical interventions, hydrocephalus, and cochlear damage from radiation therapy and platinum-based chemotherapy [6,12,17,18,19,20,21]. Genetic susceptibility may also impact the risk of developing ototoxicity from platinum agents [22]. Given the significant morbidity associated with ototoxicity and the frequent use of platinum-based agents (predominantly cisplatin and carboplatin) as chemotherapy backbones in pediatric neuro-oncology, recent efforts have focused on ototoxicity prevention. The most commonly investigated systemically administered agents have been amifostine and sodium thiosulfate (Figure 1) [23].
Amifostine, an organic thiophosphate compound, is dephosphorylated by alkaline phosphatase to its active metabolite, which binds to cisplatin metabolites and scavenges free radicals [23]. While amifostine was included as an otoprotective agent in the recent medulloblastoma SJMB12 trial (NCT01878617), a recent systematic review strongly recommends against its use due to a lack of efficacy in preventing ototoxicity (Table 1) [23,24].
On the contrary, sodium thiosulfate (STS) is commercially available in the United States and Europe for the prevention of cisplatin-induced hearing loss for children, adolescents, and young adults with non-metastatic solid tumors [25,26,27]. STS likely acts to reduce cisplatin-induced ototoxicity by promoting the formation of inactive cisplatin compounds and inhibiting the formation of reactive oxygen species in cochlear cells [23,26,28]. Only one randomized clinical trial evaluating the efficacy of STS in pediatric oncology has included children with CNS tumors. ACCL0431 randomized children 1–18 years of age with multiple cancer types (total n = 125; n = 26 with medulloblastoma/CNS primitive neuroectodermal tumor) between STS and observation; patients were stratified by age, prior cranial radiation, and cisplatin infusion duration [29,30]. STS significantly decreased the proportion of patients with hearing loss (28.6% in the STS-treated group versus 56.4% in the observation group, p = 0.00022). For the secondary survival analysis, patients were post hoc classified as having localized (n = 77) versus disseminated disease (n = 47). No survival differences were observed for patients with localized disease. However, in the disseminated disease group, overall survival was significantly lower among patients treated with STS. While the post hoc analysis was deemed at risk of significant bias (including unmeasured confounders such as the two arms not being matched for prognostic factors) [23,30,31], the most recent clinical practice guideline provided a weak recommendation to consider STS in children and adolescents with non-metastatic cancers other than hepatoblastoma for the prevention of cisplatin-induced ototoxicity and to not routinely use STS for children with metastatic cancer [23]. STS is currently approved to be administered intravenously at a dose of 10–20 g/m2 over 15 min at 6h after the end of the cisplatin infusion [27]. The most commonly reported side effects include nausea and vomiting, nephrotoxicity, cytopenia, and electrolyte imbalance (hypernatremia, hypermagnesemia, hypophosphatemia, hypokalemia) [26,29,32]. Whether STS mitigates worsening ototoxicity among children with pre-existing hearing loss or among those who have developed this treatment-related complication is yet to be determined (Table 1) [23]. Additionally, future clinical trials among disease-specific populations (e.g., ACNS2031 for children with medulloblastoma) and for children with metastatic disease are urgently warranted [30].
Table 1. Comparison of otoprotective agents.
Table 1. Comparison of otoprotective agents.
AgentIndicationsChemical ClassBenefitsAdverse EffectsDose
AmifostineShould not be routinely used to prevent cisplatin-induced hearing loss [23]Organic thiophosphate compoundDid not reduce hearing loss in pooled analyses [23]Hypocalcaemia, nausea, hypotension [23]600 mg/m2 IV pre-cisplatin infusion and three hours into cisplatin infusion [24]
Sodium thiosulfatePrevention of cisplatin-induced hearing loss for children, adolescents, and young adults with non-metastatic solid tumorsAntioxidantSignificant reduction of hearing loss by approximately 50% [29,30]Nausea, vomiting, nephrotoxicity, cytopenia, electrolyte imbalance (hypernatremia, hypermagnesemia, hypophosphatemia, hypokalemia) [26,29,32]10–20 g/m2 IV over 15 min, six hours after the end of the cisplatin infusion [27]
Abbreviations: IV: intravenous.

3. Neurocognitive Toxicity

CNS toxicity remains a leading cause of morbidity in the treatment of pediatric brain tumors. Some of these complications occur during treatment, while others may present months or years following the completion of therapy. Surgery, radiation, chemotherapy, and novel biological or targeted treatments have all been shown to result in CNS side effects. Cognitive deficits in survivors of childhood cancer pose a substantial impact on their functional outcomes and quality of life, such as difficulties adapting to new environments, poor academic performance, social withdrawal, and, at times, challenges to living independently or completing activities of daily living.
Various aspects of therapy play a role in affecting neurocognitive outcomes in survivors of childhood cancer. The development of hydrocephalus, post-surgical complications such as cerebellar mutism, and a previous history of stroke or seizures all negatively impact cognitive function [33,34,35]. Traditional chemotherapy can have deleterious CNS effects, with added toxicity for some agents when administered intrathecally or at higher doses. Systemic therapies such as high dose methotrexate or intrathecal chemotherapy have been associated with cognitive side effects in brain tumor survivors. Side effects may include encephalopathy, myelopathy, cerebellar degeneration, ataxia, motor deficits, posterior reversible encephalopathy syndrome, seizures, and visual or hearing toxicity [36].
Radiation is commonly used in the treatment of pediatric CNS tumors. Radiation is associated with short- and long-term neurotoxicity. In the more immediate phase, pseudoprogression, worsening neurological symptoms, somnolence, and radiation necrosis are concerns, whereas long-term toxicity includes vasculopathy, neurocognitive impairment, and secondary malignancies. The dose and field of radiation impact this risk factor, with higher cumulative RT dosing leading to worse outcomes. Furthermore, patient age at the time of radiation administration plays a role [37,38,39,40,41], hence the standard radiation-sparing approach to treatment in various infant brain tumors. There is a decline in multiple neuropsychological domains due to radiation therapy, including decreased intelligence quotient (IQ), cognitive abilities, attention, concentration, processing speed, and memory. This has been shown to manifest difficulty acquiring new skills when compared to peers, rather than progressive loss of previously acquired skills. This may contribute in part to the known age-related effects on treatment, specifically radiation [42].
Several strategies have been developed to reduce the neurocognitive late effects of therapy, which include the choice of chemotherapy agents, radiation doses, and radiation techniques with reduction in exposure to surrounding healthy brain tissue, such as the use of proton radiation.
Nonpharmacological interventions have been suggested to reduce, limit, or even reverse the effect of radiation on the brain: control of hypertension and diabetes, reducing or limiting exposure to alcohol, and smoking cessation have been used in adult patients. Limiting doses to the hippocampus and supratentorial area, along with overall dose de-escalation, have also been studied (Table 2) [20,43,44].
In regard to pharmacological management, memantine has shown promise in delaying the time to cognitive decline and improving preservation of processing speed and executive function in some patents (Table 2) [45]. As a low-affinity voltage-dependent noncompetitive glutamatergic NMDA antagonist, memantine preferentially binds to NMDA receptors and prevents calcium ion influx, which in turn prevents the disruption of synaptic plasticity [45]. In early studies—when used in patients with brain metastasis, not primary CNS tumors—memantine has been shown to be associated with near-normal physiological NMDA activity despite high levels of glutamine when used as a neuroprotective mechanism. Memantine was evaluated in a phase III randomized placebo-controlled trial in patients treated with whole brain radiotherapy, where memantine treatment was initiated within 3 days of RT and continued for 24 weeks [45]. Within this study, although the primary endpoint of reduced decline in relayed recall was not reached, memantine was found to have some benefit, with a reduced probability of cognitive failure at 24 weeks (53.8% vs. 64.9%) and a longer time to cognitive decline, higher executive function at 8 and 16 weeks, and delayed recognition at 24 weeks [45]. There is limited data on the effects of memantine in patients with primary CNS tumors. Clinical trials are underway; one study includes patients older than 6 years of age receiving focal brain irradiation for a diagnosis of glioma, craniopharyngioma, ependymoma, or germ cell tumors (NCT03194906). COG is currently conducting a phase III randomized, placebo-controlled trial evaluating memantine’s effect as a neuroprotective mechanism in pediatric CNS tumor patients undergoing RT (ACCL2031).
Metformin, another oral agent, has shown promise in improving cognitive recovery in pediatric patients following radiation therapy [46]. Metformin has been shown to improve working memory by acting on neural stem cells in the subventricular zone and dentate gyrus, restoring neurogenesis following RT [46]. Early-phase randomized placebo-controlled trials demonstrated enhanced auditory-verbal recall and working memory in pediatric patients [43,44]. Metformin was overall well-tolerated in these studies, with mild gastrointestinal side effects such as diarrhea being most common [46].
Modafinil, a dopaminergic CNS stimulant commonly used for narcolepsy treatment, has shown some improvement in attention, psychomotor speed, and memory [47,48,49]. Although not all studies have shown cognitive benefit thus far [50], it has been trialed in COG’s ACCL0922 to evaluate its effect on improvement in attention, executive function, and fatigue in comparison to a placebo. Results remain pending.
Other groups have evaluated cognitive training programs in pediatric patients undergoing cranial RT. The Children’s Oncology Group (COG) ACCL10P1 is one such study that utilized a computerized cognitive training program on children 3–5 times per week.

4. Vascular Complications

Children treated for CNS tumors are at significant risk of cerebrovascular events (CVEs), including transient ischemic attacks, strokes (ischemic, hemorrhagic), moyamoya, and vascular malformations [51,52]. Among survivors of childhood CNS tumors, the incidence of stroke has been estimated to be as high as 267.6 per 100,000 person-years, representing a relative risk of stroke 29 times higher than in sibling controls [51]. In a population-based study of medulloblastoma survivors conducted by our group, the cumulative incidence of stroke at age 30 was 6.5% compared to 0.3% in matched controls [6]. The first stroke may occur as early as 10 years post-cancer diagnosis, with a median age of 27 years [51,52,53]. Importantly, subsequent strokes are exceedingly common; the 10-year cumulative incidence of recurrent stroke among CNS tumor survivors treated with radiation therapy has been estimated to be 31% [52]. CNS tumor survivors who had experienced a stroke had a late mortality hazard ratio that was 2.1 times higher than stroke-free survivors and were more likely to be unmarried, unemployed, disabled, and living with a caregiver [54]. Overall, CVEs convey significant morbidity and mortality for CNS tumor survivors [55,56].
The underlying mechanism of CVEs is incompletely understood but likely involves radiation-induced arteriopathy, vascular remodeling, accelerated atherosclerosis, and subsequent vessel infarction due to inflammation [57,58]. Survivors are often asymptomatic, with vascular abnormalities detected on neuroimaging screening (Figure 1) [57]. Risk factors for stroke and recurrent stroke include older age, increased time since treatment, cranial radiation therapy (dose-dependent, highest risk among patients who received ≥50 Gy), suprasellar radiation, neurofibromatosis type 1, hypertension, hypercholesterolemia, obesity, and diabetes mellitus [6,51,53,57].
Few guidelines exist for the surveillance of pediatric CNS tumor survivors at risk of neurovascular complications. For asymptomatic adult cancer survivors who received head/neck radiation therapy, the European Society of Cardiology recommends to consider carotid ultrasound imaging every 5 years, starting at 5 years after radiation and every 5–10 years thereafter [59]. The most recent survivorship guidelines from the Children’s Oncology Group advocate for the following considerations for survivors who have received brain radiation: (1) managing risk factors that predispose to CVEs (e.g., hypertension, diabetes, hyperlipidemia); (2) imaging with brain MRI and MRA as indicated; (3) revascularization interventions for moyamoya as indicated; (4) Doppler ultrasound of carotid vessels as clinically indicated (with referral to cardiology if abnormal) [60]. For survivors who received ≥40 Gy radiation to the neck, these guidelines recommend a neck doppler ultrasound 10 years post-radiation completion as a baseline [60]. However, a recent study reported that only 19.4% of CNS tumor survivors meeting screening criteria had received a carotid ultrasound, and survivors who had seen a cancer specialist in follow-up were more likely to have received appropriate screening [61]. Neuroimaging for screening asymptomatic childhood cancer survivors at risk for CVEs (modality, initiation, duration, frequency) and the use of antiplatelet therapies remain controversial [57]. There is consensus among experts that survivors with asymptomatic vascular abnormalities detected on screening imaging should have follow-up imaging every 1–2 years [57]. Future research should focus on the impact of screening for asymptomatic neurovascular disease on outcomes, the role of antiplatelet agents and statins to mitigate the risks of CVEs, and the impact of proton therapy on the incidence of CVEs [57].

5. Radiation Necrosis

A complication of external beam radiation in the management of pediatric CNS tumors is radiation-induced injury or radiation necrosis. It is thought to be related to direct injury to vasculature and occurs within the first two years of radiation treatment, but may be as early as 6 months following RT. In the early setting, RT necrosis is often indistinguishable from tumor progression or recurrence, which often possess challenges diagnostically [62,63,64,65].
Radiation necrosis has been postulated to be secondary to two mechanisms: vascular injury from the upregulation of pro-inflammatory markers, resulting in endothelial cell death, or the creation of a pro-inflammatory environment. Radiation necrosis may be managed with an observational approach, or, in those who are symptomatic, treatment with corticosteroids or bevacizumab is often used (Figure 1) [66]. Bevacizumab, an anti-angiogenesis agent, has been shown to decrease wound healing and increase the risk of hemorrhage and thrombosis in patients; it has also been associated with improvement in neurocognitive outcomes—specifically, memory, attention, language, and concentration—in comparison to corticosteroid therapy [63].
Compared with conventional photon radiation, proton radiation offers a theoretical advantage in sparing adjacent normal brain tissues. This may offer potential benefits with the reduction of some long-term effects of therapy such as cognitive deficits, endocrine or vascular changes, and secondary malignancies. Unfortunately, radiation necrosis remains a challenge with both modalities of RT therapy, protons and photons, with some studies showing a higher incidence and shorter time to development of radiation necrosis with proton radiation [62,65].

6. De-Escalation of Therapy to Reduce Toxicity

For a subset of pediatric CNS tumors with good survival outcomes, efforts are being made to de-escalate therapy to reduce toxicities while still maintaining excellent survival outcomes.
Patients with non-metastatic WNT medulloblastoma who have undergone GTR of their tumors and been treated with multimodal standard therapy have excellent progression-free survival (PFS) at greater than 90% at 5 years. Patients with WNT-driven medulloblastoma treated on the SJMB03 protocol had 100% PFS at 5 years [67]. However, the toxicities were not negligible. Of the 53 patients treated in the study with WNT-driven disease, one developed pulmonary fibrosis and died 8.3 years from diagnosis, four developed secondary malignancies, and three of these died. The current St Jude’s medulloblastoma study SJMB12 (NCT01878617) is attempting to reduce therapy for WNT-driven medulloblastoma with reduced-dose CSI as well as a reduced dose of cyclophosphamide during maintenance chemotherapy. Similarly, COG recently completed accrual to ACNS1422, which treated standard-risk WNT-driven medulloblastoma patients with reduced CSI (18 Gy) with a limited target volume boost to the tumor bed as well as a reduced chemotherapy approach (no vincristine during radiotherapy and a reduced-dose maintenance) (NCT02724579). Analysis of this study is ongoing.
Patients with CNS germinoma also have good survival outcomes but are at risk for a number of late effects from their treatment. Efforts have been made to avoid radiation, but results showed that whole-ventricular irradiation (WVI) is an essential part of treatment [68,69]. Recent international cooperative trials have demonstrated good survival outcomes with lower WVI at 24 Gy. A report from a Japanese multi-institutional study of 115 patients treated with carboplatin and etoposide followed by 24 Gy WVI showed that only one patient had tumor recurrence, with a median follow-up time of 36 months [70,71]. Early reports from the European SIOP trial also showed excellent outcomes for patients with CR treated with 24 Gy WVI [72]. Given these promising results, the current COG study (ACNS2321) is similarly examining a response-based reduction in radiation for a subset of patients with CNS germinoma (NCT06368817).
Ongoing efforts of molecular characterization and participation in large cooperative studies will help further highlight which groups of patients have the best survival outcomes and may be amenable to further therapy de-escalation. Ongoing studies such as ACNS1833 and 1831 are randomizing patients between targeted therapy and conventional chemotherapy, which may provide data on both the efficacy as well as the toxicity of each approach.
As we learn more about outcomes by tumor subtypes and treatment regimen, it will be important to use this information to de-escalate treatment when possible, thereby minimizing toxicity.

7. Fertility Preservation

Patients treated for pediatric CNS tumors are at an increased risk for impaired fertility. In particular, patients treated with heavy metal, alkylator, and radiation therapy are at significant risk for infertility [73]. While some of these risks are well-established, newer targeted therapies may also pose future risks to fertility, but many of these are still emerging or unknown [74]. Cranial irradiation can affect the hypothalamic pituitary axis, while alkylating chemotherapy can be gonadotoxic. Current guidelines suggest that fertility preservation measures should be discussed with patients prior to initiating chemotherapy when possible [75]. A systematic review of published reports on fertility after CNS cancer showed a polled prevalence of gonadal toxicity of 20% in survivors of CNS tumors [76]. However, the population was very heterogeneous, and the follow-up was short. Outcomes vary widely based on diagnosis and specific treatment. In a study of fertility outcomes in 62 medulloblastoma survivors, 76% of female patients and 34% of male patients had clinical or biochemical evidence of gonadal dysfunction [77].
The Children’s Oncology Group (COG) has published a report stratifying some of the most commonly used treatment protocols for CNS tumors based on fertility risk [78]. They examined the risks based on treatment agents and doses and stratified by patient sex and pubertal status. Most (7/11) CNS treatment protocols placed at least one group of patients at high risk for impaired fertility. Male patients were most commonly at a high risk for infertility, followed by pubertal females.
Based on sex and pubertal status, a number of fertility preservation options currently exist for patients. For post-pubertal males who can produce a sperm specimen, sperm can be cryopreserved. For patients who may have limited physical mobility due to the location of their CNS tumor or to surgical complications, an alternative method of collecting sperm such as electro-ejaculation may be considered [79].
For pubertal female patients, oocyte collection and cryopreservation is an established method of fertility preservation [80]. However, this requires hormone stimulation, which typically takes two weeks to achieve. For some CNS tumors, this may lead to delays in treatment that could affect survival outcome. Fertility preservation via hormone suppression with Gonadotropic Releasing Hormone agonists (GnRHa) may also be considered, but the benefit is still uncertain. A current COG study ALTE 2131 is investigating the effect of GnRHa Triptorelin on female adolescent and young adult cancer patients being treated with alkylating agents (NCT06513962).
In prepubertal female patients, ovarian tissue collection and cryopreservation with the intent of future re-implantation or in vitro growth is an option for some patients [81]. This is most often done by laproscopic oophorectomy and ideally combined with other procedures such as port-a-cath insertion [82]. In prepubertal males, testicular tissue cryopreservation is an investigational option at some institutions [83] and may be able to be combined with other procedures that the patient is undergoing to avoid delays in therapy. The efficacy of achieving live birth after cryopreservation in pre-pubertal patients is still being established. For prepubertal patients at risk for impaired fertility due to premature ovarian failure, some families may also choose close endocrinological surveillance and a discussion of possible fertility preservation methods with the patient after puberty (Figure 1).
Another important consideration in fertility preservation is the cost associated with the procedures and banking, which vary widely between and within countries. Access to certain methods can also vary based on the patient’s institution.
Patients with CNS tumors have a unique set of challenges that can affect fertility preservation. Physical and cognitive neurological deficits in patients with newly diagnosed CNS tumors may complicate discussions about fertility preservation. The discussion around fertility may also be complicated by the very poor prognosis of certain CNS tumors, such as diffuse midline glioma. For patients with cognitive changes or limited prognosis, an ethicist may be consulted to help navigate some of these decisions. With the advances of molecular diagnostics in CNS tumors, a specific treatment plan may not be decided until the results of molecular tests are obtained, sometimes 2–3 weeks after diagnosis. In these cases, the time between diagnosis and initiation of treatment may be very short, thereby limiting the time for fertility preservation. Given the many known and possible risks to fertility posed by treatment for CNS tumors, referral for oncofertility preservation is recommended when feasible.

8. Importance of Surveillance and Follow Up

With improved survival in children, adolescents, and young adults treated for childhood cancer, the study of chronic health conditions related to therapy remains an ongoing area of research focus. Early recognition, prevention, and initiation of treatment are essential to improve outcomes and quality of life in childhood brain tumor survivors. Monitoring the late effects of therapy is critical, as it permits the early recognition of side effects and subsequent intervention or treatment [84]. Multi-disciplinary follow-up along with access to psychosocial support is imperative.
The Children’s Oncology Group (COG)’s “Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent and Young Adult Cancers” serve as screening and management guidelines for the late effects of therapy. Late effects are numerous and, in part, are related to the treatments that children receive—whether surgery, radiation, chemotherapy, or a combination approach—the age at which they were treated, the cumulative doses or exposure of treatments, and the location of their tumor, just to name a few. Given the multitude of effects therapy can have on patients, focusing on various organ systems after the completion of treatment is imperative. Adult survivors of childhood CNS tumors are at a high risk for long-term morbidity and late mortality related to the therapy they received, with great risk for the development of new endocrine, neurological, or sensory complications 5 years or more after diagnosis [85].
The Childhood Cancer Survivorship Study (CCSS) included 1877 CNS tumor survivors, 82% of whom suffered at least one chronic medical condition after therapy [85]. With increasing time from original cancer diagnosis, studies have demonstrated that childhood cancer survivors are less likely to receive care specific to long-term follow up [86,87]. It is at these increasing years post-cancer therapy that survivors are at the most risk for developing new medical conditions. Thus, adherence to long-term care guidelines by specialized long-term follow up clinics or dissemination of information to general practitioners and following guidelines recommendations are important.

9. Conclusions

Survivors of pediatric CNS tumors are highly susceptible to long-term toxicity resulting from tumor location and the treatments administered, amongst other factors. These profound late effects are associated with increased morbidity and mortality. Ongoing efforts to reduce the late effects of therapy by employing strategies to minimize toxicity, de-escalating therapy, pursuing fertility preservation, and ensuring patients undergo surveillance and follow up after the completion of therapy are imperative.

Author Contributions

All authors equally contributed to this work. The manuscript was written, reviewed, and edited by all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rickert, C.H.; Paulus, W. Epidemiology of central nervous system tumors in childhood and adolescence based on the new WHO classification. Child’s Nerv. Syst. 2001, 17, 503–511. [Google Scholar] [CrossRef]
  2. Coltin, H.; Johnston, D.; Larouche, V. Epidemiology of Pediatric Primary Central Nervous System Tumors. In Pediatric Neuro-Oncology; Scheinemann, K., Bouffet, E., Eds.; Springer International Publishing: Cham, Switzerland, 2024; pp. 11–15. [Google Scholar]
  3. Price, M.; Ballard, C.; Benedetti, J.; Neff, C.; Cioffi, G.; A Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S.; Ostrom, Q.T. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2017–2021. Neuro-Oncology 2024, 26, vi1–vi85. [Google Scholar] [CrossRef] [PubMed]
  4. Bhakta, N.; Liu, Q.; Ness, K.K.; Baassiri, M.; Eissa, H.; Yeo, F.; Chemaitilly, W.; Ehrhardt, M.J.; Bass, J.; Bishop, M.W.; et al. The cumulative burden of surviving childhood cancer: An initial report from the St Jude Lifetime Cohort Study (SJLIFE). Lancet 2017, 390, 2569–2582. [Google Scholar] [CrossRef] [PubMed]
  5. Yeh, J.M.; Ward, Z.; Chaudhry, A.; Liu, Q.; Yasui, Y.; Armstrong, G.T.; Gibson, T.M.; Howell, R.; Hudson, M.M.; Krull, K.R.; et al. Life Expectancy of Adult Survivors of Childhood Cancer Over 3 Decades. JAMA Oncol. 2020, 6, 350–357. [Google Scholar] [CrossRef] [PubMed]
  6. Coltin, H.; Pequeno, P.; Liu, N.; Tsang, D.S.; Gupta, S.; Taylor, M.D.; Bouffet, E.; Nathan, P.C.; Ramaswamy, V. The Burden of Surviving Childhood Medulloblastoma: A Population-Based, Matched Cohort Study in Ontario, Canada. J. Clin. Oncol. 2023, 41, 2372–2381. [Google Scholar] [CrossRef]
  7. Meijer, A.J.M.; van der Heuvel-Eibrink, M.M.; Brooks, B.; Am Zehnhoff-Dinnesen, A.G.; Knight, K.R.; Freyer, D.R.; Chang, K.W.; Hero, B.; Papadakis, V.; Frazier, A.L.; et al. Recommendations for Age-Appropriate Testing, Timing, and Frequency of Audiologic Monitoring During Childhood Cancer Treatment: An International Society of Paediatric Oncology Supportive Care Consensus Report. JAMA Oncol. 2021, 7, 1550–1558. [Google Scholar] [CrossRef]
  8. Moke, D.J.; Luo, C.; Millstein, J.; Knight, K.R.; Rassekh, S.R.; Brooks, B.; Ross, C.J.D.; Wright, M.; Mena, V.; Rushing, T.; et al. Prevalence and risk factors for cisplatin-induced hearing loss in children, adolescents, and young adults: A multi-institutional North American cohort study. Lancet Child Adolesc. Health 2021, 5, 274–283. [Google Scholar] [CrossRef]
  9. Schreiber, J.E.; Gurney, J.G.; Palmer, S.L.; Bass, J.K.; Wang, M.; Chen, S.; Zhang, H.; Swain, M.; Chapieski, M.L.; Bonner, M.J.; et al. Examination of risk factors for intellectual and academic outcomes following treatment for pediatric medulloblastoma. Neuro-Oncology 2014, 16, 1129–1136. [Google Scholar] [CrossRef]
  10. Olivier, T.W.; Bass, J.K.; Ashford, J.M.; Beaulieu, R.; Scott, S.M.; Schreiber, J.E.; Palmer, S.; Mabbott, D.J.; Swain, M.A.; Bonner, M.; et al. Cognitive Implications of Ototoxicity in Pediatric Patients With Embryonal Brain Tumors. J. Clin. Oncol. 2019, 37, 1566–1575. [Google Scholar] [CrossRef]
  11. Bass, J.K.; Liu, W.; Banerjee, P.; Brinkman, T.M.; Mulrooney, D.A.; Gajjar, A.; Pappo, A.S.; Merchant, T.E.; Armstrong, G.T.; Srivastava, D.; et al. Association of Hearing Impairment With Neurocognition in Survivors of Childhood Cancer. JAMA Oncol. 2020, 6, 1363–1371. [Google Scholar] [CrossRef]
  12. Grewal, S.; Merchant, T.; Reymond, R.; McInerney, M.; Hodge, C.; Shearer, P. Auditory late effects of childhood cancer therapy: A report from the Children’s Oncology Group. Pediatrics 2010, 125, e938–e950. [Google Scholar] [CrossRef] [PubMed]
  13. Knight, K.R.G.; Kraemer, D.F.; Neuwelt, E.A. Ototoxicity in children receiving platinum chemotherapy: Underestimating a commonly occurring toxicity that may influence academic and social development. J. Clin. Oncol. 2005, 23, 8588–8596. [Google Scholar] [CrossRef] [PubMed]
  14. Moxon-Emre, I.; Dahl, C.; Ramaswamy, V.; Bartels, U.; Tabori, U.; Huang, A.; Cushing, S.L.; Papaioannou, V.; Laperriere, N.; Bouffet, E.; et al. Hearing loss and intellectual outcome in children treated for embryonal brain tumors: Implications for young children treated with radiation sparing approaches. Cancer Med. 2021, 10, 7111–7125. [Google Scholar] [CrossRef]
  15. Strebel, S.; Baust, K.; Grabow, D.; Byrne, J.; Langer, T.; Zehnhoff-Dinnesen, A.A.; Kuonen, R.; Weiss, A.; Kepak, T.; Kruseova, J.; et al. Auditory complications among childhood cancer survivors and health-related quality of life: A PanCareLIFE study. J. Cancer Surviv. 2023, 19, 162–173. [Google Scholar] [CrossRef]
  16. Brinkman, T.M.; Bass, J.K.; Li, Z.; Ness, K.K.; Gajjar, A.; Pappo, A.S.; Armstrong, G.T.; Merchant, T.E.; Srivastava, D.K.; Robison, L.L.; et al. Treatment-induced hearing loss and adult social outcomes in survivors of childhood CNS and non-CNS solid tumors: Results from the St. Jude Lifetime Cohort Study. Cancer 2015, 121, 4053–4061. [Google Scholar] [CrossRef]
  17. Whelan, K.; Stratton, K.; Kawashima, T.; Leisenring, W.; Hayashi, S.; Waterbor, J.; Blatt, J.; Sklar, C.A.; Packer, R.; Mitby, P.; et al. Auditory complications in childhood cancer survivors: A report from the childhood cancer survivor study. Pediatr. Blood Cancer 2011, 57, 126–134. [Google Scholar] [CrossRef]
  18. Murphy, B.; Jackson, A.; Bass, J.K.; Tsang, D.S.; Ronckers, C.M.; Kremer, L.; Baliga, S.; Olch, A.; Zureick, A.H.; Jee, K.-W.; et al. Modeling the Risk of Hearing Loss From Radiation Therapy in Childhood Cancer Survivors: A PENTEC Comprehensive Review. Int. J. Radiat. Oncol. 2023, 119, 446–456. [Google Scholar] [CrossRef]
  19. Bass, J.K.; Hua, C.-H.; Huang, J.; Onar-Thomas, A.; Ness, K.K.; Jones, S.; White, S.; Bhagat, S.P.; Chang, K.W.; Merchant, T.E. Hearing Loss in Patients Who Received Cranial Radiation Therapy for Childhood Cancer. J. Clin. Oncol. 2016, 34, 1248–1255. [Google Scholar] [CrossRef] [PubMed]
  20. Tsang, D.S.; Kim, L.; Liu, Z.A.; Janzen, L.; Khandwala, M.; Bouffet, E.; Laperriere, N.; Dama, H.; Keilty, D.; Craig, T.; et al. Intellectual changes after radiation for children with brain tumors: Which brain structures are most important? Neuro-Oncology 2021, 23, 487–497. [Google Scholar] [CrossRef]
  21. Keilty, D.; Khandwala, M.; Liu, Z.A.; Papaioannou, V.; Bouffet, E.; Hodgson, D.; Yee, R.; Cushing, S.; Laperriere, N.; Ahmed, S.; et al. Hearing Loss After Radiation and Chemotherapy for CNS and Head-and-Neck Tumors in Children. J. Clin. Oncol. 2021, 39, 3813–3821. [Google Scholar] [CrossRef]
  22. Meijer, A.J.M.; Diepstraten, F.A.; Langer, T.; Broer, L.; Domingo, I.K.; Clemens, E.; Uitterlinden, A.G.; de Vries, A.C.H.; van Grotel, M.; Vermeij, W.P.; et al. TCERG1L allelic variation is associated with cisplatin-induced hearing loss in childhood cancer, a PanCareLIFE study. NPJ Precis. Oncol. 2021, 5, 64. [Google Scholar] [CrossRef] [PubMed]
  23. Freyer, D.R.; Brock, P.R.; Chang, K.W.; Dupuis, L.; Epelman, S.; Knight, K.; Mills, D.; Phillips, R.; Potter, E.; Risby, D.; et al. Prevention of cisplatin-induced ototoxicity in children and adolescents with cancer: A clinical practice guideline. Lancet Child Adolesc. Health 2020, 4, 141–150. [Google Scholar] [CrossRef]
  24. Gurney, J.G.; Bass, J.K.; Onar-Thomas, A.; Huang, J.; Chintagumpala, M.; Bouffet, E.; Hassall, T.; Gururangan, S.; Heath, J.A.; Kellie, S.; et al. Evaluation of amifostine for protection against cisplatin-induced serious hearing loss in children treated for average-risk or high-risk medulloblastoma. Neuro-Oncology 2014, 16, 848–855. [Google Scholar] [CrossRef] [PubMed]
  25. Ohlsen, T.J.D.; Collier, W.H.; Ramdas, J.; Sung, L.; Freyer, D.R. Otoprotective Effects of Sodium Thiosulfate by Demographic and Clinical Characteristics: A Report From Children’s Oncology Group Study ACCL0431. Pediatr. Blood Cancer 2024, 72, e31479. [Google Scholar] [CrossRef]
  26. Meijer, A.J.; Diepstraten, F.A.; Ansari, M.; Bouffet, E.; Bleyer, A.; Fresneau, B.; Geller, J.I.; Huitema, A.D.; Kogner, P.; Maibach, R.; et al. Use of Sodium Thiosulfate as an Otoprotectant in Patients With Cancer Treated With Platinum Compounds: A Review of the Literature. J. Clin. Oncol. 2024, 42, 2219–2232. [Google Scholar] [CrossRef]
  27. Dhillon, S. Sodium Thiosulfate: Pediatric First Approval. Pediatr. Drugs 2022, 25, 239–244. [Google Scholar] [CrossRef] [PubMed]
  28. Bijarnia, R.K.; Bachtler, M.; Chandak, P.G.; van Goor, H.; Pasch, A. Sodium thiosulfate ameliorates oxidative stress and preserves renal function in hyperoxaluric rats. PLoS ONE 2015, 10, e0124881. [Google Scholar] [CrossRef]
  29. Freyer, D.R.; Chen, L.; Krailo, M.D.; Knight, K.; Villaluna, D.; Bliss, B.; Pollock, B.H.; Ramdas, J.; Lange, B.; Van Hoff, D.; et al. Effects of sodium thiosulfate versus observation on development of cisplatin-induced hearing loss in children with cancer (ACCL0431): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 63–74. [Google Scholar] [CrossRef]
  30. Brock, P.; Meijer, A.; Kogner, P.; Ansari, M.; Capra, M.; Geller, J.; Heuvel-Eibrink, M.v.D.; Knight, K.; Kruger, M.; Lindemulder, S.; et al. Sodium thiosulfate as cisplatin otoprotectant in children: The challenge of when to use it. Pediatr. Blood Cancer 2023, 70, e30248. [Google Scholar] [CrossRef]
  31. Orgel, E.; Villaluna, D.; Krailo, M.D.; Esbenshade, A.; Sung, L.; Freyer, D.R. Sodium thiosulfate for prevention of cisplatin-induced hearing loss: Updated survival from ACCL0431. Lancet Oncol. 2022, 23, 570–572. [Google Scholar] [CrossRef]
  32. Brock, P.R.; Maibach, R.; Childs, M.; Rajput, K.; Roebuck, D.; Sullivan, M.; Laithier, V.; Ronghe, M.; Dall’Igna, P.; Hiyama, E.; et al. Sodium Thiosulfate for Protection from Cisplatin-Induced Hearing Loss. N. Engl. J. Med. 2018, 378, 2376–2385. [Google Scholar] [CrossRef] [PubMed]
  33. Armstrong, G.T.; Conklin, H.M.; Huang, S.; Srivastava, D.; Sanford, R.; Ellison, D.W.; Merchant, T.E.; Hudson, M.M.; Hoehn, M.E.; Robison, L.L.; et al. Survival and long-term health and cognitive outcomes after low-grade glioma. Neuro-Oncology 2010, 13, 223–234. [Google Scholar] [CrossRef]
  34. Di Pinto, M.; Conklin, H.M.; Li, C.; Merchant, T.E. Learning and memory following conformal radiation therapy for pediatric craniopharyngioma and low-grade glioma. Int. J. Radiat. Oncol. Biol. Phys. 2012, 84, e363–e369. [Google Scholar] [CrossRef] [PubMed]
  35. Ris, M.D.; Walsh, K.; Wallace, D.; Armstrong, F.D.; Holmes, E.; Gajjar, A.; Zhou, T.; Packer, R.J. Intellectual and academic outcome following two chemotherapy regimens and radiotherapy for average-risk medulloblastoma: COG A9961. Pediatr. Blood Cancer 2013, 60, 1350–1357. [Google Scholar] [CrossRef] [PubMed]
  36. Weaver, L.; Samkari, A. Neurological Complications of Childhood Cancer. Semin. Pediatr. Neurol. 2017, 24, 60–69. [Google Scholar] [CrossRef] [PubMed]
  37. Butler, R.W.; Haser, J.K. Neurocognitive effects of treatment for childhood cancer. Ment. Retard. Dev. Disabil. Res. Rev. 2006, 12, 184–191. [Google Scholar] [CrossRef]
  38. Heitzer, A.M.; Kahalley, L.S.; Minard, C.G.; Stafford, C.; Grosshans, D.R.; Okcu, M.F.; Raghubar, K.P.; Gragert, M.; McCurdy, M.; Warren, E.H.; et al. Treatment age and neurocognitive outcomes following proton beam radiotherapy for pediatric low- and intermediate-grade gliomas. Pediatr. Blood Cancer 2021, 68, e29096. [Google Scholar] [CrossRef]
  39. Stavinoha, P.L.; Askins, M.A.; Powell, S.K.; Smiley, N.P.; Robert, R.S. Neurocognitive and Psychosocial Outcomes in Pediatric Brain Tumor Survivors. Bioengineering 2018, 5, 73. [Google Scholar] [CrossRef]
  40. Ullrich, N.J.; Embry, L. Neurocognitive dysfunction in survivors of childhood brain tumors. Semin. Pediatr. Neurol. 2012, 19, 35–42. [Google Scholar] [CrossRef]
  41. 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]
  42. Mabbott, D.J.; Spiegler, B.J.; Greenberg, M.L.; Rutka, J.T.; Hyder, D.J.; Bouffet, E. Serial evaluation of academic and behavioral outcome after treatment with cranial radiation in childhood. J. Clin. Oncol. 2005, 23, 2256–2263. [Google Scholar] [CrossRef] [PubMed]
  43. Zammit, A.R.; Ezzati, A.; Zimmerman, M.E.; Lipton, R.B.; Lipton, M.L.; Katz, M.J. Roles of hippocampal subfields in verbal and visual episodic memory. Behav. Brain Res. 2017, 317, 157–162. [Google Scholar] [CrossRef] [PubMed]
  44. Zheng, F.; Cui, D.; Zhang, L.; Zhang, S.; Zhao, Y.; Liu, X.; Liu, C.; Li, Z.; Zhang, D.; Shi, L.; et al. The Volume of Hippocampal Subfields in Relation to Decline of Memory Recall Across the Adult Lifespan. Front. Aging Neurosci. 2018, 10, 320. [Google Scholar] [CrossRef]
  45. Brown, P.D.; Pugh, S.; Laack, N.N.; Wefel, J.S.; Khuntia, D.; Meyers, C.; Choucair, A.; Fox, S.; Suh, J.H.; Roberge, D.; et al. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: A randomized, double-blind, placebo-controlled trial. Neuro-Oncology 2013, 15, 1429–1437. [Google Scholar] [CrossRef]
  46. Ayoub, R.; Ruddy, R.M.; Cox, E.; Oyefiade, A.; Derkach, D.; Laughlin, S.; Ades-Aron, B.; Shirzadi, Z.; Fieremans, E.; MacIntosh, B.J.; et al. Assessment of cognitive and neural recovery in survivors of pediatric brain tumors in a pilot clinical trial using metformin. Nat. Med. 2020, 26, 1285–1294. [Google Scholar] [CrossRef]
  47. Lundorff, L.; Jønsson, B.; Sjøgren, P. Modafinil for attentional and psychomotor dysfunction in advanced cancer: A double-blind, randomised, cross-over trial. Palliat. Med. 2009, 23, 731–738. [Google Scholar] [CrossRef]
  48. Kohli, S.; Fisher, S.G.; Tra, Y.; Adams, M.J.; Mapstone, M.E.; Wesnes, K.A.; Roscoe, J.A.; Morrow, G.R. The effect of modafinil on cognitive function in breast cancer survivors. Cancer 2009, 115, 2605–2616. [Google Scholar] [CrossRef]
  49. Gehring, K.; Patwardhan, S.Y.; Collins, R.; Groves, M.D.; Etzel, C.J.; Meyers, C.A.; Wefel, J.S. A randomized trial on the efficacy of methylphenidate and modafinil for improving cognitive functioning and symptoms in patients with a primary brain tumor. J. Neuro-Oncol. 2011, 107, 165–174. [Google Scholar] [CrossRef] [PubMed]
  50. Boele, F.W.; Douw, L.; de Groot, M.; van Thuijl, H.F.; Cleijne, W.; Heimans, J.J.; Taphoorn, M.J.; Reijneveld, J.C.; Klein, M. The effect of modafinil on fatigue, cognitive functioning, and mood in primary brain tumor patients: A multicenter randomized controlled trial. Neuro-Oncology 2013, 15, 1420–1428. [Google Scholar] [CrossRef]
  51. Bowers, D.C.; Liu, Y.; Leisenring, W.; McNeil, E.; Stovall, M.; Gurney, J.G.; Robison, L.L.; Packer, R.J.; Oeffinger, K.C. Late-occurring stroke among long-term survivors of childhood leukemia and brain tumors: A report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 2006, 24, 5277–5282. [Google Scholar] [CrossRef]
  52. Fullerton, H.J.; Stratton, K.; Mueller, S.; Leisenring, W.W.; Armstrong, G.T.; Weathers, R.E.; Stovall, M.; Sklar, C.A.; Goldsby, R.E.; Robison, L.L.; et al. Recurrent stroke in childhood cancer survivors. Neurology 2015, 85, 1056–1064. [Google Scholar] [CrossRef]
  53. Mueller, S.; Fullerton, H.J.; Stratton, K.; Leisenring, W.; Weathers, R.E.; Stovall, M.; Armstrong, G.T.; Goldsby, R.E.; Packer, R.J.; Sklar, C.A.; et al. Radiation, atherosclerotic risk factors, and stroke risk in survivors of pediatric cancer: A report from the Childhood Cancer Survivor Study. Int. J. Radiat. Oncol. 2013, 86, 649–655. [Google Scholar] [CrossRef]
  54. Mueller, S.; Kline, C.N.; Buerki, R.A.; Chen, Y.; Yasui, Y.; Howell, R.; Oeffinger, K.C.; Leisenring, W.M.; Robison, L.L.; Armstrong, G.T.; et al. Stroke impact on mortality and psychologic morbidity within the Childhood Cancer Survivor Study. Cancer 2019, 126, 1051–1059. [Google Scholar] [CrossRef]
  55. Verbruggen, L.C.; Kok, J.L.; Kremer, L.C.M.; Janssens, G.O.; Nederkoorn, P.J.; Penson, A.; Versluijs, A.B.; de Vries, A.C.H.; Reedijk, A.M.J.; Bresters, D.; et al. Long-term risk and characteristics of cerebrovascular events after upper body radiotherapy among childhood cancer survivors in the DCCSS-LATER cohort. Int. J. Cancer 2024, 156, 1858–1872. [Google Scholar] [CrossRef]
  56. Bottinor, W.; Im, C.; Doody, D.R.; Armenian, S.H.; Arynchyn, A.; Hong, B.; Howell, R.M.; Jacobs, D.R.; Ness, K.K.; Oeffinger, K.C.; et al. Mortality After Major Cardiovascular Events in Survivors of Childhood Cancer. J. Am. Coll. Cardiol. 2024, 83, 827–838. [Google Scholar] [CrossRef] [PubMed]
  57. Kenney, L.B.; Ames, B.L.; Huang, M.S.; Yock, T.; Bowers, D.C.; Nekhlyudov, L.; Williams, D.; Hudson, M.M.; Ullrich, N.J. Consensus Recommendations for Managing Childhood Cancer Survivors at Risk for Stroke After Cranial Irradiation. Neurology 2022, 99, E1755–E1766. [Google Scholar] [CrossRef] [PubMed]
  58. Sun, L.R.; Cooper, S. Neurological Complications of the Treatment of Pediatric Neoplastic Disorders. Pediatr. Neurol. 2018, 85, 33–42. [Google Scholar] [CrossRef] [PubMed]
  59. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  60. Children’s Oncology Group. Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent and Young Adult Cancers, version 6.0; Monorovia, CA Children’s Oncology Group: Monorovia, CA, USA, 2023. Available online: www.survivorshipguidelines.org (accessed on 12 May 2025).
  61. Bryce, Y.; Whitton, J.A.; Stratton, K.L.; Leisenring, W.M.; Chow, E.J.; Armstrong, G.; Weil, B.; Dieffenbach, B.; Howell, R.M.; Oeffinger, K.C.; et al. Prevalence of carotid ultrasound screening in survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. Cancer 2024, 131, e35591. [Google Scholar] [CrossRef]
  62. Kralik, S.; Ho, C.; Finke, W.; Buchsbaum, J.; Haskins, C.; Shih, C.-S. Radiation Necrosis in Pediatric Patients with Brain Tumors Treated with Proton Radiotherapy. Am. J. Neuroradiol. 2015, 36, 1572–1578. [Google Scholar] [CrossRef]
  63. Liao, G.; Khan, M.; Zhao, Z.; Arooj, S.; Yan, M.; Li, X. Bevacizumab Treatment of Radiation-Induced Brain Necrosis: A Systematic Review. Front. Oncol. 2021, 11, 593449. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, A.K.; Foreman, N.K.; Gaspar, L.E.; Trinidad, E.; Handler, M.H. Maximally safe resection followed by hypofractionated re-irradiation for locally recurrent ependymoma in children. Pediatr. Blood Cancer 2009, 52, 804–807. [Google Scholar] [CrossRef]
  65. Carbonara, R.; Di Rito, A.; Monti, A.; Rubini, G.; Sardaro, A. Proton versus Photon Radiotherapy for Pediatric Central Nervous System Malignancies: A Systematic Review and Meta-Analysis of Dosimetric Comparison Studies. J. Oncol. 2019, 2019, 1–17. [Google Scholar] [CrossRef]
  66. Baroni, L.V.; Alderete, D.; Solano-Paez, P.; Rugilo, C.; Freytes, C.; Laughlin, S.; Fonseca, A.; Bartels, U.; Tabori, U.; Bouffet, E.; et al. Bevacizumab for pediatric radiation necrosis. Neuro-Oncol. Pract. 2020, 7, 409–414. [Google Scholar] [CrossRef] [PubMed]
  67. Gajjar, A.; Robinson, G.W.; Smith, K.S.; Lin, T.; Merchant, T.E.; Chintagumpala, M.; Mahajan, A.; Su, J.; Bouffet, E.; Bartels, U.; et al. Outcomes by Clinical and Molecular Features in Children With Medulloblastoma Treated With Risk-Adapted Therapy: Results of an International Phase III Trial (SJMB03). J. Clin. Oncol. 2021, 39, 822–835. [Google Scholar] [CrossRef] [PubMed]
  68. Finlay, J.; da Silva, N.S.; Lavey, R.; Bouffet, E.; Kellie, S.J.; Shaw, E.; Saran, F.; Matsutani, M. The management of patients with primary central nervous system (CNS) germinoma: Current controversies requiring resolution. Pediatr. Blood Cancer 2008, 51, 313–316. [Google Scholar] [CrossRef]
  69. Alapetite, C.; Brisse, H.; Patte, C.; Raquin, M.A.; Gaboriaud, G.; Carrie, C.; Habrand, J.L.; Thiesse, P.; Cuilliere, J.C.; Bernier, V.; et al. Pattern of relapse and outcome of non-metastatic germinoma patients treated with chemotherapy and limited field radiation: The SFOP experience. Neuro-Oncology 2010, 12, 1318–1325. [Google Scholar] [CrossRef]
  70. Matsutani, M. The Japanese Pediatric Brain Tumor Study Group Combined chemotherapy and radiation therapy for CNS germ cell tumors – the Japanese experience. J. Neuro-Oncol. 2001, 54, 311–316. [Google Scholar] [CrossRef]
  71. Matsutani, M.; Ushio, Y.; Abe, H.; Yamashita, J.; Shibui, S.; Fujimaki, T.; Takakura, K.; Nomura, K.; Tanaka, R.; Fukui, M.; et al. Combined chemotherapy and radiation therapy for central nervous system germ cell tumors: Preliminary results of a Phase II study of the Japanese Pediatric Brain Tumor Study Group. Neurosurg. Focus 1998, 5, E9. [Google Scholar] [CrossRef]
  72. Calaminus, G.; Bison, B.; Conter, C.F.; Frappaz, D.; Peyrl, A.; Gerber, N.U.; Müller, J.-E.; Ajithkumar, T.; Morana, G.; Cross, J.; et al. GCT-11. 24 Gy whole ventricular radiotherapy alone is sufficient for disease control in localised germinoma in CR after initial chemotherapy—Final of the SIOP CNS GCT II study. Neuro-Oncology 2022, 24, i56. [Google Scholar] [CrossRef]
  73. Salloum, R.; Chen, Y.; Yasui, Y.; Packer, R.; Leisenring, W.; Wells, E.; King, A.; Howell, R.; Gibson, T.M.; Krull, K.R.; et al. Late Morbidity and Mortality Among Medulloblastoma Survivors Diagnosed Across Three Decades: A Report From the Childhood Cancer Survivor Study. J. Clin. Oncol. 2019, 37, 731–740. [Google Scholar] [CrossRef]
  74. Helgadottir, H.; Matikas, A.; Fernebro, J.; Frödin, J.-E.; Ekman, S.; Rodriguez-Wallberg, K.A. Fertility and reproductive concerns related to the new generation of cancer drugs and the clinical implication for young individuals undergoing treatments for solid tumors. Eur. J. Cancer 2024, 202, 114010. [Google Scholar] [CrossRef] [PubMed]
  75. Loren, A.W.; Mangu, P.B.; Beck, L.N.; Brennan, L.; Magdalinski, A.J.; Partridge, A.H.; Quinn, G.; Wallace, W.H.; Oktay, K. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 2013, 31, 2500–2510. [Google Scholar] [CrossRef] [PubMed]
  76. Pape, J.; Gudzheva, T.; Beeler, D.; Weidlinger, S.; Vidal, A.; Furtwängler, R.; Karrer, T.; von Wolff, M. Long-term effects on fertility after central nervous system cancer: A systematic review and meta-analysis. Neuro-Oncol. Pract. 2024, 11, 691–702. [Google Scholar] [CrossRef]
  77. Stern, E.; Ben-Ami, M.; Gruber, N.; Toren, A.; Caspi, S.; Abebe-Campino, G.; Lurye, M.; Yalon, M.; Modan-Moses, D. Hypothalamic-pituitary-gonadal function, pubertal development, and fertility outcomes in male and female medulloblastoma survivors: A single-center experience. Neuro-Oncology 2023, 25, 1345–1354. [Google Scholar] [CrossRef] [PubMed]
  78. Felker, J.; Bjornard, K.; Close, A.; Chavez, J.; Chow, E.J.; Meacham, L.R.; Burns, K. Fertility preservation in pediatric central nervous system tumors: A report from the Children’s Oncology Group. Pediatr. Blood Cancer 2024, 71, e31246. [Google Scholar] [CrossRef]
  79. Nassau, D.E.; Ramasamy, R. Sperm retrieval options for men with spinal cord injury. Fertil. Steril. 2021, 115, 1190. [Google Scholar] [CrossRef]
  80. Mulder, R.L.; Font-Gonzalez, A.; Hudson, M.M.; van Santen, H.M.; Loeffen, E.A.H.; Burns, K.C.; Quinn, G.P.; Broeder, E.v.D.-D.; Byrne, J.; Haupt, R.; et al. Fertility preservation for female patients with childhood, adolescent, and young adult cancer: Recommendations from the PanCareLIFE Consortium and the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol. 2021, 22, e45–e56. [Google Scholar] [CrossRef]
  81. Duncan, F.E.; Pavone, M.E.; Gunn, A.H.; Badawy, S.; Gracia, C.; Ginsberg, J.P.; Lockart, B.; Gosiengfiao, Y.; Woodruff, T.K. Pediatric and Teen Ovarian Tissue Removed for Cryopreservation Contains Follicles Irrespective of Age, Disease Diagnosis, Treatment History, and Specimen Processing Methods. J. Adolesc. Young Adult Oncol. 2015, 4, 174–183. [Google Scholar] [CrossRef]
  82. Kebodeaux, C.A.; Pruett, M.; Gomez-Lobo, V.; Nahata, L.; Saraf, A.J.; Hoefgen, H.R. Ovarian Tissue Cryopreservation in Pediatric Centers across the United States: Practice Patterns and Barriers. J. Pediatr. Adolesc. Gynecol. 2024, 38, 161–166. [Google Scholar] [CrossRef]
  83. Campbell, P.; Riazzi, A.; Spitznagel, E.; Schulte, M.; Frias, O.; Daugherty, M.; Vanderbrink, B.; DeFoor, W.; Minevich, E.; Reddy, P.; et al. Time to therapy and safety of testicular tissue cryopreservation in children undergoing gonadotoxic treatment or hematopoietic stem cell transplant. J. Pediatr. Urol. 2024, 20, 747.e1–747.e7. [Google Scholar] [CrossRef] [PubMed]
  84. Cacciotti, C.; Fleming, A.; Duckworth, J.; Tseitlin, H.; Anderson, L.; Marjerrison, S. Late effects care for childhood brain Tumor Survivors: A Quality-Improvement Initiative. Pediatr. Hematol. Oncol. 2021, 39, 291–303. [Google Scholar] [CrossRef] [PubMed]
  85. Armstrong, G.T.; Liu, Q.; Yasui, Y.; Huang, S.; Ness, K.K.; Leisenring, W.; Hudson, M.M.; Donaldson, S.S.; King, A.A.; Stovall, M.; et al. Long-term outcomes among adult survivors of childhood central nervous system malignancies in the Childhood Cancer Survivor Study. JNCI J. Natl. Cancer Inst. 2009, 101, 946–958. [Google Scholar] [CrossRef] [PubMed]
  86. Oeffinger, K.C.; Mertens, A.C.; Hudson, M.M.; Gurney, J.G.; Casillas, J.; Chen, H.; Whitton, J.; Yeazel, M.; Yasui, Y.; Robison, L.L. Health care of young adult survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. Ann. Fam. Med. 2004, 2, 61–70. [Google Scholar] [CrossRef]
  87. Nathan, P.C.; Greenberg, M.L.; Ness, K.K.; Hudson, M.M.; Mertens, A.C.; Mahoney, M.C.; Gurney, J.G.; Donaldson, S.S.; Leisenring, W.M.; Robison, L.L.; et al. Medical care in long-term survivors of childhood cancer: A report from the childhood cancer survivor study. J. Clin. Oncol. 2008, 26, 4401–4409. [Google Scholar] [CrossRef]
Figure 1. Approaches to reduce and manage toxicity in pediatric CNS tumors. Created in BioRender. Coleman, C. (2025), https://app.biorender.com/illustrations/6821fce9f3f9838a242e30d7?slideId=3b25a4e6-5e03-4921-8c76-57af3cd2b260, accessed on 5 April 2025.
Figure 1. Approaches to reduce and manage toxicity in pediatric CNS tumors. Created in BioRender. Coleman, C. (2025), https://app.biorender.com/illustrations/6821fce9f3f9838a242e30d7?slideId=3b25a4e6-5e03-4921-8c76-57af3cd2b260, accessed on 5 April 2025.
Curroncol 32 00281 g001
Table 2. Approaches to reduce neurocognitive toxicity.
Table 2. Approaches to reduce neurocognitive toxicity.
NonpharmacologicalNonpharmacological Intervention
          Reduce or limit CNS radiation
          -
Limit doses to hippocampus and supratentorial area
          -
Dose de-escalation
          Control HTN and diabetes
          Reduce or limit exposure to alcohol
          Smoking cessation
PharmacologicalDrug NameMechanism of ActionWhich Patients to ConsiderEvidenceToxicity
Memantine [44]Binds to NMDA receptors and prevents calcium ion influx, which in turn prevents disruption of synaptic plasticityPatients with CNS tumors receiving radiation therapyPhase III RCT memantine initiated within 3 days of RT and continued for 24 weeks. Memantine found to have benefits, with reduced probability of cognitive failure and longer time to cognitive decline, higher executive function, and delayed recognition
Current trials:
-NCT03194906
-ACCL2031
Most common adverse effects include fatigue, alopecia, nausea, and headaches
Metformin [42,43,45]Improves working memory by acting on neural stem cells in subventricular zone and dentate gyrus, restoring neurogenesis following RTPatients with CNS tumors receiving radiation therapyEarly-phase RCT demonstrated enhanced auditory-verbal recall and working memory in pediatric patientsMild gastrointestinal side effects such as diarrhea most common
Modafinil [46,47,48,49]Dopaminergic CNS stimulantPatients with CNS tumors receiving radiation therapyImprovement in attention, psychomotor speed, and memory.
Current trials: ACCL0922
Most commonly insomnia, headaches, nausea, and anxiety
Abbreviations: NMDA: N-methyl-D-aspartate; RCT: randomized control trial; RT: radiation therapy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Coltin, H.; Coleman, C.; Cacciotti, C. Approaches to Reduce Toxicity in Pediatric Brain Tumors. Curr. Oncol. 2025, 32, 281. https://doi.org/10.3390/curroncol32050281

AMA Style

Coltin H, Coleman C, Cacciotti C. Approaches to Reduce Toxicity in Pediatric Brain Tumors. Current Oncology. 2025; 32(5):281. https://doi.org/10.3390/curroncol32050281

Chicago/Turabian Style

Coltin, Hallie, Christina Coleman, and Chantel Cacciotti. 2025. "Approaches to Reduce Toxicity in Pediatric Brain Tumors" Current Oncology 32, no. 5: 281. https://doi.org/10.3390/curroncol32050281

APA Style

Coltin, H., Coleman, C., & Cacciotti, C. (2025). Approaches to Reduce Toxicity in Pediatric Brain Tumors. Current Oncology, 32(5), 281. https://doi.org/10.3390/curroncol32050281

Article Metrics

Back to TopTop