Abstract
Neuroblastoma is the most common extracranial solid tumor in children, requiring multidisciplinary treatment, including radiotherapy, which is primarily applied in the high-risk group to prevent disease progression.
The review highlights indications for radiotherapy, its role in multimodal treatment, and addresses aspects of radiotherapy planning, including target volume definition, prescribed radiation doses, optimal timing for radiotherapy implementation, and potential side effects.
Particular attention is drawn to the lack of consensus regarding the necessity of an additional radiation dose for persistent residual disease in the primary tumor and the irradiation of metastatic sites remaining after induction therapy.
To conclude, monitoring quality assurance in radiotherapy planning and delivering processes based on unified standards appears to be crucial.
Keywords: radiotherapy, neuroblastoma, target volume
Introduction
Neuroblastoma is the second most common solid tumor in children, following tumors of the central nervous system (CNS), making it the most frequent extracranial solid tumor. This malignant neoplasm arises from primitive cells of sympathetic ganglia and often requires multidisciplinary treatment, including surgery, systemic therapies, and radiation. Radiotherapy is primarily employed in cases with a high risk of disease progression and should be administered in specialized centers.
This paper reviews the literature on the role of radiotherapy in treating neuroblastoma. It also examines clinical trial protocols that guide multidisciplinary treatment for pediatric neuroblastoma patients in Poland.
Epidemiology
Neuroblastoma accounts for 5–10% of all childhood cancers [1–3]. According to data from the Surveillance, Epidemiology, and End Results (SEER) database in the United States, incidence rates since 1975 have been estimated at 0.23 per 100,000 people per year [4]. Globally, around 14,000 cases are diagnosed annually [5]. The National Cancer Registry data from Poland reported 49 cases of adrenal tumors in 2020 and 31 in 2021 [International Statistical Classification of Diseases and Related Health Problems 10th Revision (ICD-10) — C74] [6].
Diagnosis, clinical staging, and risk groups
The diagnostic algorithm for suspected neuroblastoma includes histopathological examination of tumor tissue, complemented by immunohistochemistry, molecular profiling of the tumor, ploidy assessment of cancer cells, MYCN oncogene amplification status, and structural and numerical chromosomal aberrations. Biochemical analyses of catecholamines and their metabolites in urine, lactate dehydrogenase (LDH) activity, and levels of neuron-specific enolase (NSE) are also conducted. Stratification of the patients based on the identified risk-factors is used for radiotherapy qualification and planning. The molecular and genetic alterations commonly found in neuroblastoma include [3, 7]:
MYCN amplification is present in approximately 20% of cases, and is associated with poorer event free survival (EFS) and overall survival (OS);
ALK gene mutations — around 10% incidence, with amplification in an additional 3–4%, linked to unfavorable outcomes in EFS and OS;
TrkB receptor abnormalities — associated with worse treatment outcomes;
telomerase activity abnormalities — generally elevated, correlating with reduced EFS and OS;
TERT gene mutations;
loss of 1p and/or 11q or gain of 17q;
chromothripsis.
Standard assessments currently evaluate MYCN gene copy number, ALK gene status, ploidy, 1p36 deletion, and 17q gain.
The currently accepted histopathological classification by the International Neuroblastoma Pathology Committee (INPC) categorizes neuroblastoma into favorable and unfavorable subtypes [8, 9].
Staging of neuroblastic tumors is based on imaging studies and histopathological identification of metastatic lesions. Among the required diagnostic procedures are:
computed tomography (CT) and/or magnetic resonance imaging (MRI) of the tumor region, determined by the tumor’s location, with MRI recommended for lesions near the spinal cord;
bone marrow trephine biopsy to assess marrow involvement;
meta-iodobenzylguanidine (MIBG) scintigraphy using 123I-MIBG; for MIBG-negative patients, positron emission tomography (PET)-CT with fluorodeoxyglucose (FDG) should be ordered [10].
To assess the primary lesion, both CT and MRI should be considered [11].
For staging and detection of distant metastases, CT of the chest, abdomen, and pelvis is recommended [11]. Some experts also advise brain imaging (CT or MRI) in cases of distant metastases [11] or high-risk or recurrent disease [12].
Staging classification and risk groups
In 1971, the Evans Staging System was introduced to assess neuroblastoma stages. Current staging systems incorporate surgical and objective risk factors derived from imaging studies. Before initiating treatment, it is necessary to stage the disease using the International Neuroblastoma Risk Group Staging System (INRGSS) classification, based on image-defined risk factors (IDRFs) identified in diagnostic imaging — assigning to L1 (localized tumor without IDRF), L2 (localized tumor with IDRF), M (distant metastatic disease), and MS (distant metastatic disease, age < 18 months) stage [13].
Defining the patient’s risk group is essential and includes disease stage, patient age, tumor histology, tumor differentiation, MYCN status, 11q aberration status, and tumor cell ploidy. This pre-therapeutic risk classification places patients into one of four groups:
very low-risk — L1 or L2 stage with histopathological ganglioneuroma maturing or ganglioneuroblastoma intermixed structure, or L1 without MYCN amplification;
low-risk — age ≤ 18 months at L2 stage or age ≤ 12 months with MS stage, without MYCN amplification;
intermediate-risk — age > 18 months at L2 stage, without MYCN amplification, histopathology showing neuroblastoma or nodular ganglioneuroblastoma; also, children at L1 stage with MYCN amplification independent of age, or age < 12 months at M stage without MYCN amplification;
high-risk — age > 365 days at diagnosis with M stage (metastases) or any age with L2, M, or MS stage with MYCN amplification.
Postoperatively, a reassessment of the disease stage is necessary using the International Neuroblastoma Staging System (INSS) with stages 1, 2a, 2b, 3, 4, and 4S [13].
The most frequent tumor site is the abdomen (60–80%), followed by the chest (15%), neck (2–5%), and pelvis (2–5%) [2].
Around 40–50% of cases are diagnosed with distant metastases [11, 14]. Common metastatic sites include the bone marrow and the skeletal system, while liver, lymph nodes, and skin metastases are less common [11, 14].
Clinical trial protocols, recommendations and guidelines
In Europe, key clinical trials and treatment recommendations for pediatric patients with neuroblastoma are developed by the International Society of Pediatric Oncology Europe Neuroblastoma (SIOPEN).
Significant European clinical trial protocols for neuroblastoma include:
LINES Study — European Low and Intermediate Risk Neuroblastoma Protocol: A SIOPEN Study (latest protocol version: 30 May 2011). This multicenter, randomized Phase III clinical trial aimed to evaluate the therapy in the low- and intermediate-risk patients;
high-risk neuroblastoma 1 (HR-NBL1) — ClinicalTrials. gov Identifier: NCT01704716, HRNBL1.7 (latest protocol version: 4 April 2014);
high-risk neuroblastoma 2 (HR-NBL2) — ClinicalTrials. gov Identifier: NCT04221035 (latest protocol version: 19 July 2019). This multicenter, randomized Phase III trial’s primary endpoints include comparisons of two induction chemotherapy regimens (GPOH vs. RAPID COJEC), single high-dose chemotherapy using busulfan and melphalan versus tandem high-dose chemotherapy using thiotepa, and radiotherapy of 21.6 Gy to the preoperative tumor area versus 21.6 Gy with a sequential boost to 36 Gy for patients with residual macroscopic tumor. Recruitment is ongoing.
These protocols have led the European Society for Pediatric Oncology (SIOPE) to establish specific recommendations for low- and high-risk groups under the European Reference Network (ERN) for rare cancers [15, 16].
In addition to European standard clinical practice recommendations, Germany has its own recommendations for radiotherapy indications in intermediate- and high-risk group, which differ from SIOPEN’s [17]. According to them, in Germany radiotherapy is indicated for unresectable residual lesions that remain after induction systemic treatment and surgery [17].
Current American clinical trial protocols include:
ANBL0532 — ClinicalTrials.gov Identifier: NCT00567567 (latest protocol version: 16 August 2011). This multicenter, randomized Phase III trial aimed to intensify systemic consolidation treatment, introduce new induction regimens, and analyze the role of high-dose radiotherapy in newly diagnosed high-risk patients who could not undergo optimal surgery;
ANBL1531 — ClinicalTrials.gov Identifier: NCT03126916 (latest protocol version: 14 January 2022). This multicenter, randomized Phase III trial focuses on adding 131I-MIBG or lorlatinib to induction therapy in newly diagnosed high-risk patients. The second version of the National Comprehensive Cancer Network® (NCCN) guidelines is now available, with radiotherapy being conducted according to the clinical trial protocol Children’s Oncology Group (COG) ANBL1531 [18].
Multidisciplinary treatment
The treatment of neuroblastoma depends on the patient’s risk group. General overview on treatment of neuroblastoma according to European recommendations and stratification into risk groups is given in Table 1.
Table 1.
Overview on the risk group and the treatment methods in neuroblastoma
| Risk group | Group | INRG stage | Age (mos) | MYCN amplification | Genetics | LTS | Histology | Chemotherapy | Surgery | Auto-SCT | RTx | Retinoids | Anti-GD2 therapy |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Low risk | 1 | L2 | ≤ 18 | − | NCA | − | Any | No therapy OR CO × 2–4 | ± | − | − | - | - |
| ± VP/Carbo × 2) | |||||||||||||
| 2 | L2 | ≤ 18 | − | NCA | + | VP/Carbo × 2 | ± | − | - | ||||
| ± CADO × 2 | |||||||||||||
| 3 | L2 | ≤ 18 | − | SCA | ± | VP/Carbo × 2–4 | ± | − | - | ||||
| ± CADO × 2 | |||||||||||||
| 4 | Ms | ≤ 12 | − | NCA | − | − | − | − | - | ||||
| 5 | Ms | ≤ 12 | − | NCA | + | VP/Carbo × 2 | − | − | - | ||||
| ± CADO × 2 | |||||||||||||
| 6 | Ms | ≤ 12 | − | SCA | ± | VP/Carbo × 2–4 | ± | − | - | ||||
| ± CADO × 2 | |||||||||||||
| Intermediate risk | 7 | L2 | > 18 | − | Any | Differentiating | VP/Carbo × 2 | ± | − | − | |||
| + CADO × 2 OR VP/Carbo × 2 | |||||||||||||
| 8 | L2 | > 18 | − | Poorly differentiated/undifferentiated | VP/Carbo × 2, CADO × 2, | ± | + | + | |||||
| VP/Carbo × 1 + CADO × 1 OR CADO × 2 | |||||||||||||
| 9 | INSS1 | Any | + | Any | VP/Carbo × 2, CADO × 2, | + | + | + | |||||
| VP/Carbo × 1, CADO x1 | |||||||||||||
| 10 | M | ≤ 12 | − | VP/Carbo × 2–4 | ± | − | − | ||||||
| ± CADO × 2–4 | |||||||||||||
| High risk | M | > 12 | ± | COJEC OR N7 | + | + | + | + | + | ||||
| M | ≤ 12 | + | |||||||||||
| L2 (INSS 2,3) | > 12 | + | |||||||||||
| Other | Ganglioneuroma OR | − | ± | − | − | − | − | ||||||
| Ganglioneuroblastoma intermixed | |||||||||||||
| L1 | Any | − | Any | − | ± | − | − | − | − | ||||
Risk group — pretreatment risk group; INRG — The International Neuroblastoma Risk Group; MOS — months; LTS — life threatening symptoms; auto-SCT — autologous stem cell transplantation; RTx — radiotherapy; SCA — segmental chromosome alterations; NCA — numerical chromosomal abnormalities; INSS — International Neuroblastoma Staging System; CO — cyclophosphamide + vincristine; VP/Carbo — vincristine + etoposide phosphate + carboplatin; CADO — cyclophosphamide + doxorubicin + vincristine; COJEC — cyclophosphamide + oncovin (vincristine) + carboplatin + etoposide + cisplatin; N7 — sequential cyclophosphamide/doxorubicin/vincristine and cisplatin/etoposide
Patients in the very low-risk group often do not require active treatment due to the high likelihood of spontaneous regression or transformation into a benign form [3].
For patients in the low-risk group, the treatment typically involves surgical resection alone. Adjuvant radiotherapy or chemotherapy is only indicated in cases of residual disease progression after incomplete resection or in cases of disease recurrence [3].
Treatment of patients in the intermediate-risk group includes surgery and multidrug chemotherapy, with treatment sequence adjusted based on tumor resectability [3]. Radiotherapy may be used in some patients with poorly differentiated or undifferentiated tumors, as well as in cases with MYCN amplification.
Patients in the high-risk group require a multimodal approach. Treatment includes multidrug chemotherapy, surgical resection, high-dose therapy with autologous stem cell transplantation, radiotherapy, and maintenance therapy based on immunotherapy anti-GD2 antibodies and 13-cis-retinoic acid.
In Poland, children diagnosed with neuroblastoma are currently treated according to the SIOPEN recommendations, based on clinical trial protocols.
Optimizing the use of radiotherapy is an important quality measure in neuroblastoma treatment. In a 2024 international publication, authors identified an optimal radiotherapy utilization rate (oRUR) of 64% for newly diagnosed neuroblastoma cases globally, with rates of 50% in high-income countries and 68% in low-income countries. These differences reflect variations in disease stage distribution and healthcare access [19].
Indications for primary tumor radiotherapy
According to the current European LINES protocol for the intermediate-risk group, the indications for radiotherapy include:
L1 stage with MYCN amplification;
L2 stage in patients older than 18 months without MYCN amplification, with poorly differentiated or undifferentiated tumor histology.
There are no indications for radiotherapy in the low-risk group or intermediate-risk patients with distant metastasis.
In contrast, in the European HR-NBL1 and HR-NBL2 protocols for high-risk groups, primary tumor radiotherapy is indicated in all cases as part of consolidation treatment, regardless of the extent of surgical resection.
Additionally, European recommendations derived from these protocols include specific criteria for initiating radiotherapy:
absence of disease progression following high-dose chemotherapy and autologous stem cell transplantation;
interval of 60 to 90 days between the last transplant and radiotherapy;
performance status of ≥ 50%;
blood parameters: absolute neutrophil count (ANC) > 0.5 × 109/L, platelet count > 20 × 109/L;
acceptable liver function test results if the liver is included in the radiation field: alanine transaminase (ALT) < 3.0 × upper limit of normal (ULN), blood bilirubin < 1.5 × ULN (toxicity < grade 2) [16].
The American ANBL1531 protocol recommends primary tumor radiotherapy following induction systemic treatment under the following circumstances:
low-risk group — emergencies such as spinal cord compression or hepatomegaly with respiratory distress and vision loss;
intermediate-risk group — tumor progression during chemotherapy or persistent disease after the full course of treatment;
high-risk group — radiotherapy is indicated in every case [11].
Role of radiotherapy in primary tumor treatment
Neuroblastoma is a highly radiosensitive tumor. The primary objective of radiotherapy in neuroblastoma treatment is to improve local control [20–23]. However, distant metastases remain a major challenge in successful outcomes. Evidence suggests that improved local control is predictive of better OS [21].
In the 1990s, a randomized clinical trial demonstrated the benefit of radiotherapy in the high-risk group [22]. This study compared patients who received radiotherapy to those who did not. Patients who had undergone postoperative irradiation of the tumor bed or residual tumor and regional lymph nodes showed better outcomes, with EFS at 73% vs. 59% (p = 0.009) and OS at 41% vs. 32% (p = 0.008) [22]. It should be noted, however, that retrospective analysis of these patients classified them as intermediate-risk.
A 2018 systematic review examined evidence of the efficacy of radiotherapy in neuroblastoma. Overall, the strength of evidence for radiotherapy’s effectiveness (targeting the tumor bed or residual tumor) was graded as moderate, with a low strength of evidence for specific subgroups according to the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) system [23].
In 2010, a retrospective analysis (n = 44) found that local-regional control positively influenced OS (p = 0.06), with five-year OS rates of 48.3% in patients without local-regional relapses and 21.8% in those with relapses [21].
Role of radiotherapy in local tumor recurrence treatment
There is no established consensus on the treatment of local recurrence in neuroblastoma.
Treatment typically requires a multidisciplinary approach, which includes systemic therapy (often high-dose chemotherapy with stem cell transplantation), surgical intervention, re-irradiation through external beam radiotherapy or intraoperative radiotherapy [24–26]. Additionally, no specific guidelines exist regarding re-irradiation in the same area; however, authors consider re-irradiation a viable component in the treatment of recurrent disease [27].
Indications for radiotherapy in cases of distant metastasis
Metastatic disease is the primary cause of mortality in high-risk neuroblastoma patients.
European clinical trial protocols do not currently recommend routine irradiation of metastatic lesions that persist after induction chemotherapy (HR-NBL1 and HR-NBL2) as part of radical treatment for newly diagnosed patients. In contrast, the routine irradiation of metastatic lesions is included in the American COG protocols.
Role of radiotherapy in the treatment of metastatic lesions
There are three clinical scenarios regarding radiotherapy for metastatic lesions:
systematic consolidation irradiation of oligoresidual metastatic lesions after induction chemotherapy as part of radical treatment for newly diagnosed patients;
radical irradiation of metastatic lesions in cases of recurrence at the metastatic site (known as oligoprogression);
palliative irradiation of metastatic lesions.
Some retrospective studies have demonstrated that radiotherapy of metastatic sites improves local control at the irradiated location [28–33]. In the previously mentioned 2018 systematic review, the strength of the evidence supporting radiotherapy for metastatic lesions in neuroblastoma was classified as low according to the GRADE system [23]. Additionally, a 2023 report by the European Society for Pediatric Oncology Radiotherapy Working Group systematically reviewed the literature and found no high-quality evidence supporting radiotherapy for residual metastatic lesions after induction chemotherapy in high-risk primary treatment cases [34]. The authors recommended considering randomized trials to evaluate the benefits of such an approach and proposed appropriate designs for clinical trials [34]. Specific recommendations are lacking for the other two clinical scenarios.
Metastatic lesions in the central nervous system
A 2022 analysis by the Polish Solid Tumor Group reported a 4.5% incidence (13 out of 286 patients) of isolated CNS recurrences in high-risk neuroblastoma patients treated between 2001 and 2019 across eight pediatric oncology centers in Poland [12]. No European protocols provide specific recommendations for managing CNS recurrences. However, the American protocol ANBL1531 suggests irradiation of the craniospinal axis at a dose of 21.6 Gy in 12 fractions for CNS involvement.
In a 2010 study, Croog and colleagues analyzed patients who had undergone craniospinal irradiation and found that it enabled prolonged remissions and improved survival compared to localized irradiation [35]. Additionally, a 2020 study by Luo et al. showed comparable results in patients who received either 18 Gy or 21 Gy to the craniospinal axis, establishing this approach as a standard practice [36].
Radiotherapy treatment planning
According to the HR-NBL2 protocol, radiotherapy planning includes the following:
supine position with arms raised above the head;
use of individualized immobilization devices;
CT scans with transverse slices ≤ 3 mm;
intravenous contrast is recommended (note: an additional non-contrast CT may be necessary for accurate dose distribution analysis);
4D computed tomography (4D-CT).
4D-CT integrating respiratory movements is particularly recommended for tumors located near the diaphragm or in areas with high respiratory mobility [11].
The American ANBL1531 protocol provides additional technical recommendations:
simulation marks: Two axial planes should be marked, even when cone beam computed tomography (CBCT) verification is used, ideally on the anterior superior iliac spine and xiphoid process;
adrenal region tumors: legs should be raised under the knees to flatten the lower spine relative to the treatment table, improving immobilization;
head and neck region tumors: preoperative diagnostic CT should be performed in the treatment position.
paraspinal tumors: MRI should be performed in the treatment position;
to account for the respiratory motion: eg. with 4D-CT, even when using intensity-modulated radiation therapy (IMRT).
For radiotherapy planning under the HR-NBL2 protocol, the following imaging and surgical reports are required:
preoperative contrast-enhanced CT and/or MRI images and descriptions;
postoperative CT or MRI and MIBG scan results following high-dose chemotherapy;
surgical report and postoperative histopathology report;
No protocol includes MRI sequences as a reference for radiotherapy planning.
Radiotherapy timing
For the intermediate-risk group (according to the European LINES protocol), radiotherapy should be administered after completing chemotherapy and surgical resection, between 21 and 42 days following the previous treatment stage.
For the high-risk group (according to the European HR-NBL protocol), radiotherapy should be performed after completing chemotherapy and surgery, but before retinoid therapy, with an interval of at least 60 days and no more than 90 days following autologous stem cell transplantation.
In the American protocols, radiotherapy should be initiated no sooner than 42 days and no later than 80 days after stem cell transplantation.
Longer intervals are permissible only in cases of severe transplant-related complications, including:
hematological complications: neutrophil count < 500/μL and/or thrombocytopenia < 40,000/μL;
active veno-occlusive disease;
acute respiratory distress syndrome (ARDS) with laryngeal edema ≥ grade 2;
diarrhea Common Terminology Criteria for Adverse Events (CTCAE) grade ≥ 2;
significant renal dysfunction;
uncontrolled hematuria.
Radiotherapy target volumes for primary and metastatic tumors
For postoperative patients receiving radiotherapy to the primary tumor area, the following target volumes (TVs) are defined:
gross target volume (GTV) — visible tumor area on imaging;
clinical target volume (CTV) — Includes the GTV along with potential microscopic spread;
planned target volume (PTV) — accounts for organ motion and potential patient positioning errors.
A summary of target volume delineation guidelines for primary tumor radiotherapy, based on current clinical trial protocols, is presented in Table 2.
Table 2.
Guidelines for target volume delineation for primary tumor radiotherapy based on clinical trial protocols
| SIOPEN LINES — Intermediate-Risk Group | SIOPEN HR-NBL1/HR-NBL2 — High-Risk Group | COG ANBL1531 |
|---|---|---|
| GTV — primary tumor visible on imaging after chemotherapy and before surgery; should include the tumor and affected regional lymph nodes | GTV — primary tumor visible on imaging (CT, MRI) after chemotherapy and before surgery; should include the tumor and persistent, enlarged regional lymph nodes | GTV_PreSurgery — primary tumor visible on preoperative imaging GTV should encompass GTV_PreSurgery in the superior and inferior extents GTV should include the tumor and lymph nodes identified during surgery. It should not include the prechemotherapy tumor area or uninvolved lymph nodes (no indication for elective lymph node irradiation) If a large portion of the tumor was removed at diagnosis, GTV should be defined based on the preoperative tumor In cases of inconclusive imaging, irradiate a larger volume If the tumor extended into adjacent structures (e.g., lung or liver), only a narrow margin (< 3 mm) should be included in GTV |
| HR-NBL2 only — boost = GTVb = CTVb; includes macroscopic residual tumor post-surgery | ||
| CTV = GTV + 10 mm In the abdominal area, modify CTV to exclude the kidney, liver, or small intestine if they were compressed or displaced but not invaded Extend CTV to include adjacent vertebrae to ensure homogeneous dosing and reduce scoliosis risk |
CTV = GTV + 5 mm Ensure CTV includes areas specified in surgical and pathology reports |
CTV = GTV1 + 1 cm For abdominal tumors, modify CTV to exclude the kidney, liver, and vertebral bodies; for head and neck, thoracic, and pelvic tumors, exclude uninvolved bones Superior and inferior CTV limits should encompass the tumor’s preoperative area (GTV_PreSurgery) |
| HR-NBL2 only — boost area (additional dose) = CTVb = GTVb | ||
| PTV = CTV + ≥ 5 mm | PTV = CTV + 5–10 mm | PTV = CTV + 0.3–0.8 cm (subdiaphragmatic area + 0.5–0.8 cm) |
HR-NBL — high-risk neuroblastoma; GTV — gross target volume; CT — computed tomography; MRI — magnetic resonance imaging; CTV — clinical target volume; PTV — planned target volume
The Quality and Excellence in Radiotherapy and Imaging for Children and Adolescents with Cancer in Clinical Trials (QUARTET initiative), a project by SIOPE, enables centralized quality control of radiotherapy. The HR-NBL protocols incorporate quality assurance measures for consistent target and organs at risk delineation across participating centers.
In the American COG protocol, irradiation of metastatic lesions persisting after induction therapy is recommended before high-dose chemotherapy and stem cell transplantation.
Guidelines for delineating metastatic target volumes in the ANBL1531 protocol include:
metastatic GTV — area of metastatic lesions with residual mass > 1 cm3 or MIBG/FDG activity before consolidation; CTV = GTV + 1 cm; PTV = CTV + 0.5 – 0.8 cm;
if the radiotherapy area involves more than 50% of the bone marrow volume, verification by the study coordinator is required;
for > 5 lesions detected on MIBG/PET, repeat imaging 28 days after autologous stem cell transplantation and irradiate only areas with active disease;
special recommendations apply to radiotherapy of the calvarium, skull base, extremities, spine, and ribs.
Elective nodal irradiation
Studies have not shown a benefit from elective nodal irradiation [37].
Emergencies in radiotherapy for neuroblastoma
According to American protocols, in cases of stage MS disease, radiotherapy may sometimes be necessary to halt rapid tumor progression, particularly for tumors located near the liver where there is a risk of cardiopulmonary failure, impending spinal cord compression, or incomplete response to chemotherapy. In these emergencies, a low dose of 4.5 Gy in three fractions is typically used [3].
Dose, role of boost, and fractionation schedules
The recommended radiotherapy doses according to current clinical trial protocols are summarized in Table 3.
Table 3.
Fractionation schedules based on clinical trial protocols
| SIOPEN LINES — Intermediate-Risk Group | SIOPEN HR-NBL1 — High-Risk Group | SIOPEN HR-NBL2 — High-Risk Group | COG ANBL1531 |
|---|---|---|---|
| PTV = 21 Gy/14 fractions over 18–20 days If optimal doses cannot be achieved in organs at risk, two-phase treatment may be necessary: Phase 1 with 15 Gy in 10 fractions, followed by Phase 2 with 6 Gy in 4 fractions to a smaller area |
PTV = 21 Gy/14 fractions over a maximum of 21 days If optimal doses cannot be achieved in critical organs, two-phase treatment may be necessary. No indications for a dose boost in cases of residual disease |
PTV = 21.6 Gy/12 fractions over a maximum of 17 days (if an unscheduled break occurs, weekend irradiation or twice-daily radiotherapy with a 6-hour interval is recommended) This applies to patients without macroscopic residual disease who meet the following three conditions: No residual disease on MRI (or CT if MRI is unavailable). No residual disease on MIBG scintigraphy. The surgical report documents complete resection. If optimal doses cannot be achieved in organs at risk, two-phase treatment may be necessary Randomization studies (regarding the use of a boost) should be conducted after high-dose chemotherapy or 2–3 weeks post-surgery |
PTV 1 = 21.6 Gy/12 fractions PTV for metastatic lesions = 21.6 Gy/12 fractions Irradiation of the primary tumor area and metastatic lesions should be performed simultaneously Hepatomegaly: 4.5 Gy/3 fractions Craniospinal axis: 21.6 Gy/1.8 Gy |
| PTV = 21.6 Gy/12 fractions PTV Boost = 14.4 Gy/8 fractions to the residual lesion (total dose 36 Gy/20 fractions) For macroscopic residual disease | |||
| If parents decline randomization, the patient receives 21.6 Gy |
HR-NBL — high-risk neuroblastoma; PTV — planned target volume; MRI — magnetic resonance imaging; CT — computed tomography; MIBG — meta-iodobenzylguanidine
In previous American guidelines and ANBL0532 protocol, an additional boost to the residual primary tumor was recommended following induction chemotherapy. However, analysis of ANBL0532 data showed that boosting did not reduce the five-year cumulative local recurrence risk [38]. Meanwhile, the European SIOPEN — HR-NBL2 protocol includes randomization by a multidisciplinary team with the use of a boost dose in residual disease. Studies on dose-response relationships in neuroblastoma are inconclusive [39–42], and a systematic review classified the evidence strength for dose escalation efficacy as low according to GRADE [23]. All protocols above use conventional fractionation, with daily doses of 1.5–1.8 Gy.
Some studies have reported on hyperfractionated radiotherapy, delivering 1.5 Gy twice daily up to a total dose of 21 Gy [43].
Image-guided radiotherapy
To verify patient positioning accuracy, imaging must be performed before each radiotherapy fraction. Standard methods for this verification include 2D kilovoltage (kV) images or CBCT.
In the American protocol ANBL1531, recommended PTV margins vary depending on the location and type of imaging used (CBCT vs. non-CBCT). If non-CBCT imaging is used, a PTV margin of 5 mm is recommended across all sites, with the possibility of increasing this margin to 8 mm in the abdominal region. When CBCT is used, a margin of 3–5 mm is advised, with 3 mm for head and neck regions and 5 mm for thoracic regions.
Type of radiotherapy (proton vs. photon)
Both European and American protocols and recommendations allow for the use of either photon or proton radiation.
Radiotherapy technique
The LINES protocol (2011) allows only for 3D conformal radiotherapy. In contrast, the HR-NBL and ANBL protocols permit the use of both 3D conformal radiotherapy and IMRT techniques. IMRT can reduce doses to the kidneys in patients with centrally located tumors, although it may increase doses to other organs, such as the spleen, liver, and stomach, in cases of laterally located tumors [44]. Dynamic techniques, including IMRT and volumetric-modulated arc therapy (VMAT), are increasingly used in clinical practice, with usage rates rising significantly in recent years (Tab. 4) [45–47].
Table 4.
Radiotherapy techniques in primary tumor irradiation for high-risk neuroblastoma
| First author’s name and publication year | Study period | Analysis type | Number of patients | Radiotherapy technique |
|---|---|---|---|---|
| Gatcombe 2009 [42] | 2001–2007 | Retrospective | 34 | AP/PA 82% IMRT 18% |
| Casey 2016 [43] | 2000–2014 | Retrospective | 246 | AP/PA + 3D-CRT 65% IMRT 35% |
| Ferris 2017 [20] | 2003–2014 | Retrospective | 67 | AP/PA 58,2% 3D-CRT 9% IMRT 4,5% VMAT 28,4% |
| Chen 2019 [45] | 2009–2015 | Retrospective | 24 | IMRT/HT 100% |
| Selles 2023 [52] | 2004–2020 | Retrospective | 42 | 3D-CRT 7% IMRT/VMAT 93% |
AP/PA — anterior-posterior/posterior-anterior fields; 3D-CRT — 3D conformal radiotherapy; IMRT — intensity-modulated radiation therapy; VMAT — volumetric-modulated arc therapy
Intraoperative radiotherapy
There are no specific guidelines for the use of intraoperative radiotherapy (IORT) in the treatment of neuroblastoma, nor is this technique mentioned in current clinical trial protocols for this cancer. The largest single-center analysis (n = 92), from the United States, reports on 27 years of experience using IORT for recurrent neuroblastoma [25]. This study concluded that IORT is both effective and safe.
Organs at risk and dose limitations
Dose limitations for organs at risk, as outlined in various clinical trial protocols, are summarized in Table 5.
Table 5.
Dose limitations for organs at risk according to LINES, HR-NBL, and ANBL protocols
| Organ at risk | Dose limitations according to protocol | |||
|---|---|---|---|---|
| LINES | HR-NBL1.7 | HR-NBL2 | COG ANBL1531 | |
| Contralateral kidney | – | Tolerance dose for functioning kidneys: 15 Gy, considering potential kidney damage from prior treatment; 21 Gy allowed on one kidney if the other functions normally. | Tolerance dose for functioning kidneys: 15 Gy, considering potential kidney damage from prior treatments; ≥ 21.6 Gy allowed on one kidney if the other functions normally. | V18 Gy < 25% |
| Ipsilateral kidney | V12 Gy < 80% | V18 Gy < 75% V14.4 Gy < 100% Mean dose ≤ 18 Gy |
||
| Liver | V100 ≤ 15 Gy | V100 ≤ 19 Gy V50 ≤ 21Gy |
V100 ≤ 19 Gy V50 ≤ 21Gy |
Mean dose < 15 Gy |
| Vertebral body | The whole vertebra should be irradiated | Homogeneous coverage recommended | Homogeneous dose up to ~21.6 Gy | Minimum of 18 Gy to include vertebral body, pedicles, transverse, and spinous processes; vertebrae should not be included in PTV, only design additional pseudotarget |
| Spinal cord | – | – | 21.6 Gy and 36 Gy are acceptable regardless of segment length; caution after busulfan treatment | – |
| Lungs | V15 < 20% | V12 Gy ≤ 50% V15 Gy ≤ 25% |
V12 Gy ≤ 50% V15 Gy ≤ 25% |
V20 Gy < 30% |
| Ipsilateral lung | – | – | – | V20 Gy < 30% |
| Contralateral lung | – | – | – | V20 Gy < 10% |
PTV — planned target volume
In 2019, the SIOPE Working Group published guidelines on vertebral contouring, emphasizing consistent dose homogeneity and gradient limits for pediatric patients [48]. A 2023 publication based on clinical trials, including HR-NBL2, highlighted substantial variations in vertebral contouring, affecting dosimetry, and included an atlas for two cases [49]. The authors recommend the following contouring guidelines:
adjacent vertebral body and arch but excludes processes, spinal canal, and intervertebral discs (VBs_Adj);
non-adjacent vertebrae structures for superior and inferior vertebrae (VB_Nadj_S and VB_Nadj_L).
Dose guidelines include:
for VBs_Adj: homogeneous dose; allowable gradient of < 3 Gy for children under 2 years and < 5 Gy for older children;
for VB_Nadj_S and VB_Nadj_L: D5% ≤ 10 Gy for children under 2 years and D5% ≤ 15 Gy for older children.
Another organ at risk, previously considered incidental in radiotherapy, is the pancreas. It appears that reporting the radiotherapy dose to specific parts of the pancreas may help identify patients at increased risk of developing diabetes [50, 51].
Adverse effects of radiotherapy
Radiotherapy-related complications depend on the irradiated area and the administered dose.
Acute radiation reactions include gastrointestinal symptoms from small intestine irradiation such as nausea and vomiting, diarrhea, abdominal pain, and hematologic toxicity [3]. Hematologic toxicity mainly manifests as leukopenia, with data suggesting that a leukocyte count reduction of > 50% from baseline is associated with poorer OS (p = 0.031) [52].
Potential late effects of radiotherapy in neuroblastoma treatment include [3, 53–55]:
musculoskeletal disorders, such as spinal deformities (scoliosis, lordosis, kyphosis), bone or muscle hypoplasia, and related growth disturbances;
endocrine disorders including hypothyroidism, hypogonadism, and growth hormone deficiencies;
pulmonary and cardiovascular disorders;
nephrological complications;
second cancers.
The most common late complications after radiotherapy are musculoskeletal abnormalities, occurring in 31% of cases approximately six years post-irradiation, as reported in French clinical studies [53–55]. A threshold dose for these complications has been established at > 31 Gy [54]. Additionally, the risk of scoliosis 15 years after treatment is estimated to be around 25% [56]. Studies indicate that survivors of neuroblastoma treatment are at elevated risk of metabolic syndrome and diabetes [57, 58]. Renal abnormalities often manifest as imaging-detected atrophy or physiological changes without symptomatic acute or chronic dysfunction [59]. Second cancers primarily include thyroid cancers and soft tissue or bone sarcomas [54, 55].
Treatment outcomes in high-risk neuroblastoma
Tables 6 and 7 summarize treatment outcomes in high-risk neuroblastoma patients based on single-center analyses and prospective studies.
Table 6.
Treatment outcomes in high-risk neuroblastoma patients based on single-center analyses
| First author’s name and publication year | Analysis type | Study period and place | N | Radiotherapy dose (mean in Gy) | Follow up time (mean in months) | Local control (%) | LRF (%) | DFS (%) | OS (%) | ||
|---|---|---|---|---|---|---|---|---|---|---|---|
| 2-year | 3-year | 5-year | |||||||||
| Gatcombe 2009 [42] | Retrospective | 2001–2007 USA | 34 | 22 | 33.6 | – | 94 | – | 5.8 | 66 (3-year) | 86 (3-year) |
| Pai panandiker 2010 [21] | Retrospective | 2000–2006 USA | 44 | 23.4 | 34 | – | – | – | 25 | – | 48.3 (without LRF) 21.8 (with LRF) |
| Mazloom 2014 [30] | Retrospective | 2006–2011 USA | 30 | 24 | 33 | – | – | 84 | – | – | 59% |
| Casey 2016 [43] | Retrospective | 2000–2014 USA | 246 | 21 | 55.2 | – | – | 90.2 | 8.9 | 40 (5-year) | 64.6 (5-year) |
| Ferris 2017 [20] | Retrospective | 2003–2014 USA | 67 | 21.6 | 54 | 95.5 | – | 94 | 7.5 | – | – |
| Chen 2019 [45] | Retrospective | 2009–2015 Taiwan | 24 | 21.6 | 43.5 | – | 78.9 | – | 21.1 (3-year) | 45.8 (3-year) | 62.9 (3-year) |
| Wei 2022 [46] | Retrospective | 2011–2019 China | 69 | 36 | 30.5 | – | 96.9 | – | – | 56.3 (3-year) | 74.6 (3-year) |
| Jang 2024 [47] | Retrospective | 2001–2021 Korea | 37 | 21 | 69 | – | 88.7 | 88.7 | 10.8 | 59.1 | 37 |
LRF — locoregional failures; DFS — disease-free survival; OS — overall survival
Table 7.
Treatment outcomes in high-risk neuroblastoma patients based on prospective studies
| Study/first author’s name and publication year | Analysis type | Treatment period and place | N | Dose [Gy] | LRF (%) | DFS (%) | OS (%) |
|---|---|---|---|---|---|---|---|
| HR-NBL1.7 Pasqualini 2020 [16] |
Prospective multicenter phase III trial | Europe 2002–2016 |
1297 | 21 | 23% (with residual tumor) 15% (no residual tumor) |
38% (with residual tumor) 49% (no residual tumor) |
– |
| ANBL0532 Liu 2020 [38] |
Prospective multicenter phase III trial | USA 2007–2012 |
323 | 1.6–36 | 16.3% (CLIP) | 50.9% | 68.1% |
LRF — locoregional failures; DFS — disease-free survival; OS — overall survival
Post-treatment follow-up
According to the European HR-NBL2 protocol, oncological follow-up should be conducted every 3 months during the first year post-treatment, every 4 months during the second year post-treatment, and every 6 months during the third year post-treatment, and following this, patients should enter a long-term surveillance program. The American ANBL1531 protocol provides follow-up guidelines on a dedicated website containing detailed information on childhood cancer surveillance [60].
For high-risk patients, the surveillance plan includes:
laboratory tests of blood and urine, including catecholamine levels;
ultrasound of the primary tumor region at each visit; for tumors located in the chest, MRI (preferred) or CT is recommended.
For patients with metastatic disease, routine MIBG scintigraphy or PET and bone marrow trephine biopsy are not required; however, these may be considered annually.
Summary
Radiotherapy plays an essential role as a component of multidisciplinary treatment for neuroblastoma. It is primarily used in high-risk group following surgery, targeting the primary tumor area post-chemotherapy. There is no consensus on the use of additional dose (boost) for residual disease at the primary tumor site; however, the European HR-NBL2 study plans randomization, whereas the American protocol has excluded this option. Additionally, there is no consensus on the irradiation of residual metastatic lesions following induction treatment, though this is routinely included in American protocols. Guidelines diverge on the irradiation of metastatic sites in cases of disease recurrence. Nonetheless, radiotherapy remains an effective palliative treatment, especially for metastases in the skeletal system.
It is worth noting that a standardized methodology, including terminology for recurrence reporting in neuroblastoma, is needed. In 2009, Pai Panandiker et al. [21] proposed the following terms:
in-field recurrence: within the PTV;
marginal recurrence: outside the PTV, where a dose between 5% and 95% of the prescribed dose was delivered;
adjacent recurrence: in lymph nodes or tissues adjacent to the primary tumor and outside the PTV, where less than 5% of the prescribed dose was delivered;
distant recurrence: in locations non-adjacent to the primary tumor;
locoregional recurrence: combination of in-field and adjacent recurrences.
In contrast, Dove et al. used two categories: local recurrence, defined as the primary tumor site or adjacent lymph nodes, and regional recurrence, defined as recurrence in lymph nodes within the regional lymphatic drainage, but not connected to the primary site [26].
All protocols discussed include quality control guidelines for radiotherapy planning and delivering. The primary initiative for quality assurance is QUARTET, which enables centralized verification of target areas and radiotherapy plans. Both HR-NBL protocols are associated with QUARTET for quality control — retrospective quality control for HR-NBL1 and prospective for HR-NBL2. From a radiation oncologist’s perspective, clinical trial protocols should contain detailed information on current contouring standards for target areas, ideally with delineation examples, as seen in the ANBL1531 protocol. Equally important are guidelines for organs at risk contouring and subsequent evaluation of complications.
A multidisciplinary approach to treatment decisions is recommended, and parents should be fully informed of the planned radiotherapy, with the opportunity to consult with a radiation oncologist. An objective evaluation of radiotherapy frequency is also necessary, based on simple indicators such as the optimal radiotherapy utilization rate (oRUR). Authors of a 2024 study emphasized that understanding the oRUR is crucial for assessing current practices, identifying gaps in access, and planning future radiotherapy protocols [19].
Footnotes
Author contributions: A.J.S., M.U. A.S., B.K., A.M. — conceptualization, methodology, project administration, writing — original draft, writing — review and editing
Conflict of interests: The authors declare no conflict of interests.
Funding: None declared.
References
- 1.Tepper Jo. Gunderson and Tepper’s Clinical Radiation Oncology. 5th Ed. Elsevier; 2020. [Google Scholar]
- 2.Chybicka A. 2nd ed. Vol. 1. [Pediatric oncology and hematology]. PZWL Wydawnictwo Lekarskie; Warsaw: 2021. [Google Scholar]
- 3.Merchant TE. Pediatric Radiation Oncology. 1th ed. Springer; 2018. [Google Scholar]
- 4.Yan P, Qi F, Bian L, et al. Comparison of Incidence and Outcomes of Neuroblastoma in Children, Adolescents, and Adults in the United States: A Surveillance, Epidemiology, and End Results (SEER) Program Population Study. Med Sci Monit. 2020;26:e927218. doi: 10.12659/MSM.927218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ward ZJ, Yeh JM, Bhakta N, et al. Estimating the total incidence of global childhood cancer: a simulation-based analysis. Lancet Oncol. 2019;20(4):483–493. doi: 10.1016/S1470-2045(18)30909-4. [DOI] [PubMed] [Google Scholar]
- 6.National Cancer Registry. [15.11.2024]. https://onkologia.org.pl/pl .
- 7.Bresler SC, Weiser DA, Huwe PJ, et al. ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell. 2014;26(5):682–694. doi: 10.1016/j.ccell.2014.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shimada H, Ambros IM, Dehner LP, et al. The International Neuroblastoma Pathology Classification (the Shimada system) Cancer. 2021;39(29):3229–3241. doi: 10.1200/JCO.21.00278. [DOI] [PubMed] [Google Scholar]
- 9.Navarro S, Amann G, Beiske K, et al. European Study Group 94.01 Trial and Protocol. Prognostic value of International Neuroblastoma Pathology Classification in localized resectable peripheral neuroblastic tumors: a histopathologic study of localized neuroblastoma European Study Group 94.01 Trial and Protocol. J Clin Oncol. 2006;24(4):695–699. doi: 10.1200/JCO.2004.00.8631. [DOI] [PubMed] [Google Scholar]
- 10.Kushner BH, Yeung HW, Larson SM, et al. Extending positron emission tomography scan utility to high-risk neuroblastoma: fluorine-18 fluorodeoxyglucose positron emission tomography as sole imaging modality in follow-up of patients. J Clin Oncol. 2001;19(14):3397–3405. doi: 10.1200/JCO.2001.19.14.3397. [DOI] [PubMed] [Google Scholar]
- 11.Terezakis S, MacDonald S. Target Volume Delineation for Pediatric Cancers, Practical Guides in Radiation Oncology. 1th ed. Springer; 2011. [Google Scholar]
- 12.Wieczorek A, Stefanowicz J, Hennig M, et al. Isolated central nervous system relapses in patients with high-risk neuroblastoma-clinical presentation and prognosis: experience of the Polish Paediatric Solid Tumours Study Group. BMC Cancer. 2022;22(1):701. doi: 10.1186/s12885-022-09776-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Balwierz W, Wieczorek A, Szewczyk K, et al. [Standards of diagnostic management of sympathetic neuroblastoma in children] Paediatr Rev. 2019;48(3):89–98. [Google Scholar]
- 14.Luksch R, Castellani MR, Collini P, et al. Neuroblastoma (Peripheral neuroblastic tumours) Crit Rev Oncol Hematol. 2016;107:163–181. doi: 10.1016/j.critrevonc.2016.10.001. [DOI] [PubMed] [Google Scholar]
- 15.Cañete A, Martinez B SIOPE ERN PaedCan. Low-risk neuroblastomastandard clinical practice recommendations. https://siope.eu/media/documents/low-risk-neuroblastoma-standard-clinical-practice-recommendations.pdf .
- 16.Pasqualini C, Ladenstein R SIOPE ERN PaedCan. High-risk neuroblastomastandard clinical practice recommendations. https://siope.eu/media/documents/escp-high-risk-neuroblastoma-standard-clinical-practice-recommendations.pdf .
- 17.Simon T, Hero B, Schulte JH, et al. 2017 GPOH Guidelines for Diagnosis and Treatment of Patients with Neuroblastic Tumors. Klin Padiatr. 2017;229(3):147–167. doi: 10.1055/s-0043-103086. [DOI] [PubMed] [Google Scholar]
- 18.Bagatell R, Park JR, Acharya S, et al. National Comprehensive Cancer Network. Neuroblastoma (Version 2, 2024) [10.03.2025]. https://jnccn.org/view/journals/jnccn/22/6/article-p413.xml . [DOI] [PubMed]
- 19.Fukushima A, Hande V, Wakeham K, et al. Estimation of the optimal radiotherapy utilization rate for childhood neuroblastoma. Radiother Oncol. 2024;197:110343. doi: 10.1016/j.radonc.2024.110343. [DOI] [PubMed] [Google Scholar]
- 20.Ferris MJ, Danish H, Switchenko JM, et al. Favorable Local Control From Consolidative Radiation Therapy in High-Risk Neuroblastoma Despite Gross Residual Disease, Positive Margins, or Nodal Involvement. Int J Radiat Oncol Biol Phys. 2017;97(4):806–812. doi: 10.1016/j.ijrobp.2016.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pai Panandiker AS, McGregor L, Krasin MJ, et al. Locoregional tumor progression after radiation therapy influences overall survival in pediatric patients with neuroblastoma. Int J Radiat Oncol Biol Phys. 2010;76(4):1161–1165. doi: 10.1016/j.ijrobp.2009.03.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Castleberry RP, Kun LE, Shuster JJ, et al. Radiotherapy improves the outlook for patients older than 1 year with Pediatric Oncology Group stage C neuroblastoma. J Clin Oncol. 1991;9(5):789–795. doi: 10.1200/JCO.1991.9.5.789. [DOI] [PubMed] [Google Scholar]
- 23.Arumugam S, Manning-Cork NJ, Gains JE, et al. The Evidence for External Beam Radiotherapy in High-Risk Neuroblastoma of Childhood: A Systematic Review. Clin Oncol (R Coll Radiol) 2019;31(3):182–190. doi: 10.1016/j.clon.2018.11.031. [DOI] [PubMed] [Google Scholar]
- 24.Ussowicz M, Wieczorek A, Dłużniewska A, et al. Factors Modifying Outcome After MIBG Therapy in Children With Neuroblastoma-A National Retrospective Study. Front Oncol. 2021;11:647361. doi: 10.3389/fonc.2021.647361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Conte B, Casey DL, Tringale KR, et al. Intraoperative Radiation Therapy for Relapsed or Refractory High-Risk Neuroblastoma: A 27-Year Experience. Pract Radiat Oncol. 2024;14(3):e226–e232. doi: 10.1016/j.prro.2023.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dove AP, Manole BA, Wakefield DV, et al. Managing local-regional failure in children with high-risk neuroblastoma: A single institution experience. Pediatr Blood Cancer. 2018;65(12):e27408. doi: 10.1002/pbc.27408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Manole BA, Wakefield DV, Doubrovin M, et al. Management of Local-Regional Failure in Children With High-Risk Neuroblastoma: A Single Institution Experience. Int J Radiat Oncol Biol Phys. 2017;99(2):E570–E571. doi: 10.1016/j.ijrobp.2017.06.1974. [DOI] [Google Scholar]
- 28.Sibley GS, Mundt AJ, Goldman S, et al. Patterns of failure following total body irradiation and bone marrow transplantation with or without a radiotherapy boost for advanced neuroblastoma. Int J Radiat Oncol Biol Phys. 1995;32(4):1127–1135. doi: 10.1016/0360-3016(95)00011-m. [DOI] [PubMed] [Google Scholar]
- 29.Bradfield SM, Douglas JG, Hawkins DS, et al. Fractionated low-dose radiotherapy after myeloablative stem cell transplantation for local control in patients with high-risk neuroblastoma. Cancer. 2004;100(6):1268–1275. doi: 10.1002/cncr.20091. [DOI] [PubMed] [Google Scholar]
- 30.Mazloom A, Louis CU, Nuchtern J, et al. Radiation therapy to the primary and postinduction chemotherapy MIBG-avid sites in high-risk neuroblastoma. Int J Radiat Oncol Biol Phys. 2014;90(4):858–862. doi: 10.1016/j.ijrobp.2014.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Polishchuk AL, Li R, Hill-Kayser C, et al. Likelihood of bone recurrence in prior sites of metastasis in patients with high-risk neuroblastoma. Int J Radiat Oncol Biol Phys. 2014;89(4):839–845. doi: 10.1016/j.ijrobp.2014.04.004. [DOI] [PubMed] [Google Scholar]
- 32.Li R, Polishchuk A, DuBois S, et al. Patterns of Relapse in High-Risk Neuroblastoma Patients Treated With and Without Total Body Irradiation. Int J Radiat Oncol Biol Phys. 2017;97(2):270–277. doi: 10.1016/j.ijrobp.2016.10.047. [DOI] [PubMed] [Google Scholar]
- 33.Casey DL, Pitter KL, Kushner BH, et al. Radiation Therapy to Sites of Metastatic Disease as Part of Consolidation in High-Risk Neuroblastoma: Can Long-term Control Be Achieved? Int J Radiat Oncol Biol Phys. 2018;100(5):1204–1209. doi: 10.1016/j.ijrobp.2018.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Keshwani K, Boterberg T, Dieckmann K, et al. Radical skeletal metastatic site irradiation in high-risk neuroblastoma: systematic review and proposal for a randomised trial: a report from the SIOPEN Radiotherapy Committee. J Cancer Metastas Treat. 2023;9(1) doi: 10.20517/2394-4722.2022.32. [DOI] [Google Scholar]
- 35.Croog VJ, Kramer K, Cheung NKV, et al. Whole neuraxis irradiation to address central nervous system relapse in high-risk neuroblastoma. Int J Radiat Oncol Biol Phys. 2010;78(3):849–854. doi: 10.1016/j.ijrobp.2009.09.005. [DOI] [PubMed] [Google Scholar]
- 36.Luo LY, Kramer K, Cheung NKV, et al. Reduced-dose craniospinal irradiation for central nervous system relapsed neuroblastoma. Pediatr Blood Cancer. 2020;67(9):e28364. doi: 10.1002/pbc.28364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Braunstein SE, London WB, Kreissman SG, et al. Role of the extent of prophylactic regional lymph node radiotherapy on survival in high-risk neuroblastoma: A report from the COG A3973 study. Pediatr Blood Cancer. 2019;66(7):e27736. doi: 10.1002/pbc.27736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu KX, Naranjo A, Zhang FF, et al. Prospective Evaluation of Radiation Dose Escalation in Patients With High-Risk Neuroblastoma and Gross Residual Disease After Surgery: A Report From the Children’s Oncology Group ANBL0532 Study. J Clin Oncol. 2020;38(24):2741–2752. doi: 10.1200/JCO.19.03316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Casey DL, Kushner BH, Cheung NKV, et al. Reduced-Dose Radiation Therapy to the Primary Site is Effective for High-Risk Neuroblastoma: Results From a Prospective Trial. Int J Radiat Oncol Biol Phys. 2019;104(2):409–414. doi: 10.1016/j.ijrobp.2019.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Haas-Kogan DA, Swift PS, Selch M, et al. Impact of radiotherapy for high-risk neuroblastoma: a Children’s Cancer Group study. Int J Radiat Oncol Biol Phys. 2003;56(1):28–39. doi: 10.1016/s0360-3016(02)04506-6. [DOI] [PubMed] [Google Scholar]
- 41.von Allmen D, Grupp S, Diller L, et al. Aggressive surgical therapy and radiotherapy for patients with high-risk neuroblastoma treated with rapid sequence tandem transplant. J Pediatr Surg. 2005;40(6):936–41. doi: 10.1016/j.jpedsurg.2005.03.008. discussion 941. [DOI] [PubMed] [Google Scholar]
- 42.Gatcombe HG, Marcus RB, Katzenstein HM, et al. Excellent local control from radiation therapy for high-risk neuroblastoma. Int J Radiat Oncol Biol Phys. 2009;74(5):1549–1554. doi: 10.1016/j.ijrobp.2008.10.069. [DOI] [PubMed] [Google Scholar]
- 43.Casey DL, Kushner BH, Cheung NKV, et al. Local Control With 21-Gy Radiation Therapy for High-Risk Neuroblastoma. Int J Radiat Oncol Biol Phys. 2016;96(2):393–400. doi: 10.1016/j.ijrobp.2016.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Paulino AC, Ferenci MS, Chiang KY, et al. Comparison of conventional to intensity modulated radiation therapy for abdominal neuroblastoma. Pediatr Blood Cancer. 2006;46(7):739–744. doi: 10.1002/pbc.20456. [DOI] [PubMed] [Google Scholar]
- 45.Chen GY, Cheng JCH, Chen YH, et al. Local Control and Clinical Outcome of High-risk Pediatric Neuroblastoma Patients After Receiving Multimodality Treatment and Helical Tomotherapy. Anticancer Res. 2019;39(4):2207–2215. doi: 10.21873/anticanres.13336. [DOI] [PubMed] [Google Scholar]
- 46.Wei Z, Li J, Jin Y, et al. The application and value of radiotherapy at the primary site in patients with high-risk neuroblastoma. Br J Radiol. 2022;95(1134):20211086. doi: 10.1259/bjr.20211086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jang JY, Park JH, Kim YJ, et al. Local control and toxicity outcomes following consolidative radiation therapy in patients with high-risk neuroblastoma: a 20-year experience at a single center. Turk J Pediatr. 2024;66(1):99–109. doi: 10.24953/turkjped.2023.575. [DOI] [PubMed] [Google Scholar]
- 48.Hoeben BA, Carrie C, Timmermann B, et al. Management of vertebral radiotherapy dose in paediatric patients with cancer: consensus recommendations from the SIOPE radiotherapy working group. Lancet Oncol. 2019;20(3):e155–e166. doi: 10.1016/S1470-2045(19)30034-8. [DOI] [PubMed] [Google Scholar]
- 49.Turcas A, Kelly SM, Bernier-Chastagner V, et al. Management of the vertebrae as an organ at risk in paediatric radiotherapy clinical trials: Initial QUARTET experience. Radiother Oncol. 2023;187:109810. doi: 10.1016/j.radonc.2023.109810. [DOI] [PubMed] [Google Scholar]
- 50.Taylor S, D’Souza D, Moinuddin S, et al. Pancreas: An Organ-at-Risk to Consider in Future Pediatric Abdominal Radiotherapy. Int J Radiat Oncol Biol Phys. 2023;117(2):S135. doi: 10.1016/j.ijrobp.2023.06.538. [DOI] [Google Scholar]
- 51.Friedman DN, Moskowitz CS, Hilden P, et al. Radiation Dose and Volume to the Pancreas and Subsequent Risk of Diabetes Mellitus: A Report from the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2020;112(5):525–532. doi: 10.1093/jnci/djz152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gomis Sellés E, Delgado León BD, Cabrera Roldán P, et al. Haematological and renal toxicity of radiation therapy in neuroblastoma paediatric patients. Clin Transl Oncol. 2023;25(3):786–795. doi: 10.1007/s12094-022-02987-5. [DOI] [PubMed] [Google Scholar]
- 53.Zhao Q, Liu Y, Zhang Y, et al. Role and toxicity of radiation therapy in neuroblastoma patients: A literature review. Crit Rev Oncol Hematol. 2020;149:102924. doi: 10.1016/j.critrevonc.2020.102924. [DOI] [PubMed] [Google Scholar]
- 54.Ducassou A, Gambart M, Munzer C, et al. Neuroblastoma study group and radiotherapy group of the French Society of Children with Cancer (SFCE) Long-term side effects of radiotherapy for pediatric localized neuroblastoma : results from clinical trials NB90 and NB94. Strahlenther Onkol. 2015;191(7):604–612. doi: 10.1007/s00066-015-0837-z. [DOI] [PubMed] [Google Scholar]
- 55.Friedman DN, Henderson TO. Late Effects and Survivorship Issues in Patients with Neuroblastoma. Children (Basel) 2018;5(8) doi: 10.3390/children5080107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Paulino AC, Fowler BZ. Risk factors for scoliosis in children with neuroblastoma. Int J Radiat Oncol Biol Phys. 2005;61(3):865–869. doi: 10.1016/j.ijrobp.2004.07.719. [DOI] [PubMed] [Google Scholar]
- 57.van Waas M, Neggers SJ, Raat H, et al. Abdominal radiotherapy: a major determinant of metabolic syndrome in nephroblastoma and neuroblastoma survivors. PLoS One. 2012;7(12):e52237. doi: 10.1371/journal.pone.0052237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Friedman DN, Moskowitz CS, Hilden P, et al. Diabetes mellitus in long-term survivors of childhood cancer. Increased risk associated with radiation therapy: a report for the childhood cancer survivor study. Arch Intern Med. 2009;169(15):1381–1388. doi: 10.1001/archinternmed.2009.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Beckham TH, Casey DL, LaQuaglia MP, et al. Renal Function Outcomes of High-risk Neuroblastoma Patients Undergoing Radiation Therapy. Int J Radiat Oncol Biol Phys. 2017;99(2):486–493. doi: 10.1016/j.ijrobp.2017.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers Version 6.0. [15.11.2024]. http://www.survivorshipguidelines.org . [PubMed]
