Long-term Outcomes of Protocol-Based Treatment for Newly Diagnosed Medulloblastoma

Article information

Cancer Res Treat. 2024;56(2):652-664
Publication date (electronic) : 2023 November 30
doi : https://doi.org/10.4143/crt.2023.865
1Division of Pediatric Hematology and Oncology, Department of Pediatrics, Yonsei University Health System, Yonsei University College of Medicine, Seoul,
2Department of Pediatric Hemato-Oncology, Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
3Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea
4Department of Radiation Oncology, Inha University Hospital, Incheon, Korea
5Department of Neurosurgery, Yonsei University Health System, Yonsei University College of Medicine, Seoul,
6Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
7Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
Correspondence: Jung Woo Han, Department of Pediatrics, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea Tel: 82-2-2228-2050 Fax: 82-2-393-9118 E-mail: jwhan@yuhs.ac
Received 2023 July 23; Accepted 2023 November 29.

Abstract

Purpose

The Korean Society of Pediatric Neuro-Oncology (KSPNO) conducted treatment strategies for children with medulloblastoma (MB) by using alkylating agents for maintenance chemotherapy or tandem high-dose chemotherapy (HDC) with autologous stem cell rescue (ASCR) according to the risk stratification. The purpose of the study was to assess treatment outcomes and complications based on risk-adapted treatment and HDC.

Materials and Methods

Fifty-nine patients diagnosed with MB were enrolled in this study. Patients in the standard-risk (SR) group received radiotherapy (RT) after surgery and chemotherapy using the KSPNO M051 regimen. Patients in the high-risk (HR) group received two and four chemotherapy cycles according to the KSPNO S081 protocol before and after reduced RT for age following surgery and two cycles of tandem HDC with ASCR consolidation treatment.

Results

In the SR group, 24 patients showed 5-year event-free survival (EFS) and overall survival (OS) estimates of 86.7% (95% confidence interval [CI], 73.6 to 100) and 95.8% (95% CI, 88.2 to 100), respectively. In the HR group, more infectious complications and mortality occurred during the second HDC than during the first. In the HR group, the 5-year EFS and OS estimates were 65.5% (95% CI, 51.4 to 83.4) and 72.3% (95% CI, 58.4 to 89.6), respectively.

Conclusion

High intensity of alkylating agents for SR resulted in similar outcomes but with a high incidence of hematologic toxicity. Tandem HDC with ASCR for HR induced favorable EFS and OS estimates compared to those reported previously. However, infectious complications and treatment-related mortalities suggest that a reduced chemotherapy dose is necessary, especially for the second HDC.

Introduction

Medulloblastoma (MB) is the most common malignant brain tumor in childhood, accounting for 10%-15% of all intracranial tumors [1,2]. Multimodality therapy, consisting of surgery, radiotherapy (RT), and chemotherapy, has been used to treat patients with MB. The survival rate with multimodality therapy is affected by age, metastatic stage at diagnosis, and resection status after surgery [3,4]. The revised 2021 classification system for central nervous system tumors defines MB according to histological and molecular types (i.e., wingless integrated [WNT], sonic hedgehog [SHH] with TP53-wildtype, SHH, and TP53-mutant, and non-WNT/non-SHH), which are associated with different genetics, clinical features, and prognosis [5].

Following the introduction of multi-agent chemotherapy for MB treatment in the 1970s, numerous studies have been conducted [6]. In a previous study, the standard-risk (SR) group was treated with 36 and 54 Gy cerebrospinal irradiation and total tumor bed /posterior fossa RT, respectively. However, adjuvant chemotherapy with 23.4 Gy of reduced cerebrospinal irradiation was recently attempted, and it achieved nearly 80% event-free survival (EFS) rate [7,8]. In the high-risk (HR) group, in the CCG 921 study conducted in the late 1980s, a 5-year EFS of approximately 40% was achieved with 36 and 55.8 Gy cerebrospinal irradiation and total RT, respectively [9].

Furthermore, CCG 921 was accompanied by adjuvant chemotherapy consisting of vincristine, 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU), and steroids [9]. Sandwich treatment with neoadjuvant chemotherapy before RT was administered in the PNET-3 and German HIT91 clinical trials and showed similar results [10,11]. High-dose chemotherapy (HDC) with autologous stem cell rescue (ASCR) transplantation is a treatment alternative and was shown to achieve a 5-year EFS of 70% [7].

To reduce long-term neurotoxicity associated with RT, dose-modified craniospinal irradiation (CSI) has been considered. The Children’s Oncology Group (COG) recommends lowering the radiation dose to the cerebrospinal axis by 25% for patients aged < 6 years. The attempt to reduce CSI from 36.0 Gy to 23.4 Gy with adjuvant chemotherapy in children with SR MB, showed similar survival outcomes along with improved cognitive outcomes [12]. A low-dose CSI of 18.0 Gy showed higher intelligence scores, although it did not improve the survival rate and showed inferior outcomes in patients in group 4 [13].

The increased survival rate of patients with MB has led to several late complications, such as neurocognitive function deterioration, hearing loss, and endocrine problems, which are considered important [14]. To prevent these complications, various treatment strategies that reduce the intensity of chemotherapy and RT or introduce modifications to the treatment method have been established. The Korean Society of Pediatric Neuro-oncology (KSPNO) developed protocols for MB in 2005 by using an alkylating agent after RT for SR MB patients and reduced-dose craniospinal RT (CSRT) and tandem HDC for HR patients [15].

Therefore, in this study, we aimed to analyze the treatment outcomes of MB patients, the complications during treatment, and the late effects to improve subsequent KSPNO protocols for MB.

Materials and Methods

1. Patients and risk groups

From April 2005 to March 2021, 86 patients were newly diagnosed with MB at Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea. Twelve patients were < 3 years old. Five patients were treated with different treatment protocols, two discontinued the treatment protocol, three were transferred to other hospitals, and five refused chemotherapy. Fifty-nine of them treated with KSPNO M051 or S081 protocols, were retrospectively reviewed.

All diagnoses were confirmed and classified according to histological and molecular groups using immunohistochemical staining [16]. The KSPNO SR group was defined as patients with gross total resection (GTR) or nearly total resection (NTR) status and no metastasis at diagnosis. The molecular subgroup was determined according to the activation status of the WNT and SHH signaling pathways through analysis of immunohistochemical staining. MYC status was analyzed through fluorescence in situ hybridization. The HR group was defined as patients with residual tumors > 1.5 cm2 after surgery or metastasis at the time of diagnosis confirmed using cerebrospinal fluid (CSF) cytology or spinal magnetic resonance imaging (MRI). In total, 24 and 35 patients were included in the SR and HR groups, respectively.

At first diagnosis, brain and spinal MRI scans were performed to determine the stage and presence of metastatic lesions, including leptomeningeal seeding. Primary surgical resection was performed by qualified pediatric neurosurgeons at our institution. Furthermore, the extent of the resection was as much as possible according to the anatomical location of the tumor and considering the risk of the patient’s neurologic complications after surgery. The extent of resection was evaluated using an MRI scan taken within 48 hours after the operation.

2. Definition and risk stratification

Resection status was defined as GTR for no visible tumor after surgery, NTR for ≥ 95%, subtotal resection (STR; 50% to 95%), partial resection (10%-49%), and biopsy only for < 10%. The degree of residual disease (R stage) was defined as a negative margin of resection for R0 and a positive margin of resection or gross residual tumor for R1.

The metastatic stage (M stage) was determined through a CSF study using a lumbar puncture performed 7 days after surgery and a postoperative spinal MRI scan within 1 week of surgery. The M stages were described according to Chang’s staging system [1]. Briefly, in this system, M1, M2, M3, and M4 stages are described as microscopic tumor cells found in the CSF, intracranial nodular seeding, spinal nodular or seeding lesions identified using imaging studies, and extraneural metastasis, respectively.

3. Treatment

The KSPNO M051 regimen for the SR group consisted of surgery, followed by RT and chemotherapy. The children received eight cycles of cyclophosphamide, vincristine, and cisplatin and four cycles of cyclophosphamide and vincristine (Table 1, Fig. 1). RT comprised 23.4 Gy CSRT with a three-dimensional conformal boost to the tumor bed (55.8 Gy) with weekly vincristine 1.5 mg/m2 as a radiosensitizer.

Chemotherapy regimen

Fig. 1.

The Korean Society of Pediatric Neuro-Oncology (KSPNO) protocol scheme for medulloblastoma. The KSPNO M051 protocol for the standard-risk group (A) and the KSPNO S081 protocol for the high-risk group (B). CSRT, craniospinal radiotherapy; CTx, chemotherapy; HD, high-dose; RTx, radiotherapy.

The KSPNO S081 regimen for the HR group consisted of chemotherapy before and after RT and tandem HDC with ASCR. Two cycles of chemotherapy were administered within 2-4 weeks of surgical resection; 23.4 Gy CSRT and 30.6 Gy tumor bed with weekly vincristine were administered to all patients with an M0 status. Treatment for the M1 stage differed according to age. RT was administered at the same CSRT dose to those with M0 status, boosted to the tumor with an additional 21.6 Gy for spinal seeding nodules in patients < 6 years old.

The older patients > 6 years old were administered 30.6 Gy for CSRT, 23.4 Gy for tumor bed and intracranial seeding nodules, and 14.4 Gy for spinal seeding nodules. Within 4 weeks of completing RT, four additional cycles of chemotherapy at a 75% reduced dose were administered to the patients. After completing six cycles of chemotherapy, tandem HDC with ASCR was administered for consolidation. For the first round of HDC, the carboplatin, thiotepa, and etoposide (CTE) regimen was used as a conditioning treatment, and the cyclophosphamide and melphalan (CM) regimen was used for the second round of HDC [17].

The period between the tandems was at least 12 weeks to reduce side effects [18]. Since 2015, the planned dose of HDC has been reduced to 80% owing to treatment-related mortality. Each chemotherapy course began when peripheral blood counts recovered to acceptable levels, with an absolute neutrophil count > 750/μL and platelet count > 75,000/μL.

4. Follow-up

After two to three chemotherapy cycles and before RT and HDC, an MRI scan was performed for disease status evaluation. Responses of MB and leptomeningeal seeding tumors were evaluated according to the Response Assessment in Pediatric Neuro-Oncology (RAPNO) Criteria. In this assessment, complete response, partial response, and progression are defined as the disappearance of target lesions, a decrease of at least 30% in the sum of the diameters of the target lesions, and an increase at least 20% in the sum of diameters of the target lesions, respectively.

5. Toxicity

Treatment-related toxicities were monitored according to the Common Terminology Criteria for Adverse Events (CTCAE) ver. 4.0. All adverse events above grades 3, 4, and 5 that occurred during and after treatment were recorded.

6. Statistical analysis

Data are presented as median values with interquartile range (IQR), numbers with percentages, and means±standard deviation. The survival duration was calculated from the date of diagnosis to the last follow-up date. EFS was defined as the time from diagnosis to the first occurrence of death from any cause, relapse, progressive disease, or development of a secondary malignancy. Disease-specific survival (DSS) was defined as disease recurrence and death. Overall survival (OS) was defined as the time from diagnosis to death from any cause. Hazard ratios with associated 95% confidence intervals (CI) and p-values comparing outcome distributions were calculated using Cox regression.

For analyzing 5-year EFS, DSS, and OS, we focused specifically on patients in the SR treated with KSPNO M051 and in the HR treated with KSPNO S081. The Kaplan-Meier method was employed to analyze survival outcomes, and the log-rank test was used to assess statistical significance. Fisher’s exact test was used to analyze the parametric variables, and the Mann-Whitney U test was used for non-parametric variables. All statistical analyses were performed using the IBM Statistical Package for the Social Sciences (SPSS) ver. 23.0 (IBM SPSS Statistics, IBM Corp., Armonk, NY) and R statistical software ver. 4.1.0 (Foundation for Statistical Computing, Vienna, Austria).

Results

Fifty-nine patients aged > 3 years were enrolled and treated using the KSPNO protocol at Yonsei Cancer Center, Yonsei University Health System, Seoul, Korea. Table 2 shows their disease characteristics according to the SR and HR groups. The histologic diagnosis showed that classic MB was the most common type occurring in 50 patients (84.7%), followed by desmoplastic/nodular, medulloblastoma with extensive nodularity, and large cell/anaplastic types in six (10.2%), two (3.4%), and one (1.7%) patient, respectively. Twenty-three patients (95.8%) in the SR were treated with the KSPNO M051 protocol, and only one patient was treated with KSPNO S081 as recommended by the surgeon after total resection although there was no metastatic lesion on cytology or imaging study. Thirty-three patients (94.3%) in the HR were treated by KSPNO S081 protocol, and two patients were treated with KSPNO M051, respectively, due to delayed subsequential treatment from postoperative infection and parental refusal of HDC with ASCR.

Demographics of the study patients at diagnosis

1. Metastasis and resection status

In this cohort, 29 of 59 patients (49.2%) had metastases, consisting of five (8.5 %), four (6.8%), and 20 patients (33.9%) with M1, M2, and M3 metastases, respectively. Resection was performed where possible, and total or NTR was performed in 24 patients (100%) with SR and 18 of 33 (54.3%) with HR.

2. Radiotherapy

All patients were treated with RT with local radiation and CSRT. Patients in the SR group were administered 23.4 Gy (IQR, 23.4 to 24.2) of CSRT with a boost of 32.4 Gy (IQR, 30.6 to 32.4) to the tumor bed, whereas the HR group received 30.6 Gy (IQR, 30.3 to 30.6) of CSRT with a 23.4 Gy (IQR, 23.4 to 25.6) boost to the tumor bed and 14.4 Gy to the spinal seeding nodule.

3. Chemotherapy

The intensities of the chemotherapy doses were adjusted according to the patients’ general conditions and the level of bone marrow recovery. Considering the actual dose and duration of treatments, patients in the SR group received 93.2% (76.8%-103.3%) of the planned chemotherapy dose. Patients in the HR group received 86.9% (78.1%-92.2%) of the planned dose for induction chemotherapy, 80.0% (71.3%-90.0%) of the planned tandem HDC, and 85.1% (74.7%-91.7%) of the total intensity of the KSPNO S081 regimen. Furthermore, the period before RT or chemotherapy after surgery was 0.81 (0.69-0.93) and 0.59 (0.36-0.78) months in the SR and HR groups, respectively.

4. Survival outcome

The median follow-up time was 8.2 (5.0-11.4) and 5.4 (2.1-8.5) years in the SR and HR groups, respectively. The estimated 5-year EFS and OS were 86.0% (95% CI, 72.5 to 100) and 95.7% (95% CI, 87.7 to 100) in the SR group and 66.4% (95% CI, 52.0 to 84.8) and 68.3% (95% CI, 53.8 to 86.7) in the HR group (Fig. 2A and B). Moreover, the estimated 10-year EFS and OS were 77.4% (95% CI, 59.2 to 100) and 82.2% (95% CI, 65.3 to 100) in the SR group and 62.5% (95% CI, 47.6 to 82.0) and 62.6% (95% CI, 46.7 to 84.0) in the HR group. There was a similar EFS and OS rate in patients with GTR or NTR and those with STR or biopsy only (p=0.4).

Fig. 2.

Event-free survival and overall survival for patients according to the risk group (A, B) and molecular subgroup (C, D).

The comparison according to metastatic status showed that disease progression and number of deaths were higher in the M2 and M3 groups than they were in the M0 and M1 groups, but the difference was not statistically significant (Table 3). Furthermore, the molecular subgroup analyses of 36 patients using immunohistochemical staining showed that patients with SHH with TP53 mutation had inferior EFS and OS (Fig. 2C and D). The Cox multivariate regression analyses performed with age at diagnosis, sex, residual tumor > 1.5 cm2 after surgery, metastatic status, intensity of chemotherapy, molecular subgroup, and duration from surgery to subsequent treatment showed no significant differences.

Difference of survival rate according to risk groupa), extension of resection and M stage

5. Progression or relapse during therapy

There were 10 cases of tumor recurrence and progression. In the SR group, two patients (8.3%) had progression of leptomeningeal seeding within 3 years of their diagnosis and subsequently died as a result. Three of the 17 patients who underwent STR or biopsy had local relapses. In the HR group, eight (22.9%) patients had progression or relapse, consisting of four each with local relapse and leptomeningeal seeding, whereas seven of them consequently died. In addition, one patient treated with the KSPNO M051 regimen developed a secondary malignant neoplasm (undifferentiated pleomorphic sarcoma) in the occipital lobe and skull within the radiation field 9 years after his diagnosis. He was treated with resection of the tumor and adjuvant chemotherapy and is now alive without evidence of disease 12 months after diagnosis of a second malignancy.

6. Outcomes according to chemotherapy intensity

In the SR group, seven patients who received chemotherapy were administered ≤ 80% of the planned dose, and two of them experienced disease progression and subsequently died 5 years after diagnosis (p=0.041). In the HR group, 30 patients were treated with HDC, including 26 who completed tandem HDC. There was no difference in DSS and OS between patients who received > 80% and those who received < 80% of the total planned treatment dose (Table 4).

Dose modification and 5-year outcomes in KSPNO M051 and S081

7. Treatment toxicity and patient mortality

Toxicities of ≥ grade 3 according to the CTCAE that occurred during chemotherapy and HDC with ASCR are summarized in Tables 5 and 6, respectively. In the KSPNO M051 regimen, hematologic and infectious toxicities of ≥ grade 3 were the most common. When administering the KSPNO M051 regimen, two patients (8.0%) had treatment-related mortalities, one died of cytomegaloviral pneumonitis in the SR, and the other died of pneumonia and acute respiratory distress syndrome in the HR.

Grade 3 and 4 adverse events related with treatment

Grade 3 and 4 adverse events related with HDC with ASCR

When bone marrow recovery was delayed, the intensity of the regimen was reduced by extending the interval between chemotherapy cycles or individual modification of the planned dose in the KSPNO M051 regimen for SR patients. However, two patients (SHH without TP53, one patient; non-WNT/non-SHH, one patient) treated with ≤ 80% of the planned dose experienced recurrence and died. The 10-year progression-free survival (PFS) and DSS of patients administered > 80% of the planned dose were both 100%, whereas values of those who received ≤ 80% of the planned dose were 68.6% (p=0.03) and 60.0% (p=0.04), respectively (S1 Table). However, one patient (WNT subgroup) of > 80% of the planned dose experienced secondary malignancy within the RT field 9 years after his diagnosis.

Nine patients (26.5%) treated with the KSPNO S081 regimen before HDC had sepsis, including one who had meningitis without mortality. During tandem HDC with ASCR, acute toxicities were mainly reported for infectious diseases and were more frequent in the second HDC (Table 6). Conversely, hepatitis was more frequent in the first HDC treatment group. Hepatic veno-occlusive disease (VOD) occurred in one (3.2%) and two (7.41%) patients during the first and second HDC, respectively. One patient died of myelopathy after the first HDC with ASCR, and three patients died during the second HDC with ASCR, consisting of one each of VOD with hepatorenal syndrome, pneumonia with acute respiratory distress syndrome, and uncontrolled gastrointestinal bleeding.

Discussion

In this cohort study of patients with MB over 3 years of age, chemotherapy and RT administered according to the KSPNO protocol showed favorable treatment outcomes. In the SR, survival outcomes are better than those reported in other studies [7,13,19-21]. However, the HR group showed toxicity to be overcome, despite similar survival outcomes as in previous studies [3,7,22,23].

In the previously conducted COG A9961 and SIOP PNET-4 studies (S2 Table), patients with MB in the SR group who received CSRT (23.4 Gy) and RT boost to the tumor bed (55.8 Gy) with various combinations of chemotherapy showed 80% 5-year EFS [19-21]. In the SJMB 96 and 03 studies, patients with MB in the SR group received the same dose of RT with HDC plus ASCR four times every 4 weeks. The results showed a similar EFS of 83% without treatment-related mortality [7]. In the COG ACNS 0331 study, reducing RT to 18 Gy for CSRT in young children showed inferior outcomes [13].

In the KSPNO M051 protocol, patients received the same dose of RT and chemotherapy as that of the higher-intensity alkylating drug, instead of CCNU. The total intensity was higher than that of the total HDC in the SJMB 96 study, and the total duration of chemotherapy was longer at 48 weeks than that in the COGA9961 study, where a similar weekly average intensity of chemotherapy was used.

Survival outcomes were promising, but frequent hematologic and infectious toxicities occurred in the KSPNO M051 protocol. A dose reduction below 80% of the planned dose showed a risk of relapse. Based on this finding, we believe that the RT and planned dose intensity of chemotherapy should be maintained to minimize recurrence; however, it has to be considered individually with the molecular subgroup and the risk of treatment-related mortality, as also noted in this cohort.

To increase the survival rate of MB patients with HR, various attempts have been made to modify postoperative RT from 36 Gy of CSRT and to change the combination and intensity of chemotherapy (S3 Table). In the GPOH-HIT study, neoadjuvant and adjuvant chemotherapy with RT and combinations of various chemotherapies were used, and the 5-year EFS was > 60% [11,22]. In the SIOP PENT-3 protocol, RT alone was compared with RT and adjuvant chemotherapy, and the combination showed better results than RT alone [10]. Maintenance therapy with alkylating agents has been used in an attempt to improve the survival rate of metastatic patients [3,23]. Hypofractionated radiation therapy (HFRT) was used in the COG and HIT studies, but did not show better survival outcomes than previous regimens; therefore, a follow-up study is ongoing [22,24].

In addition, HDC for HR MB has been considered part of the treatment for patients in the HR or relapsed state [7,25]. In the SJMB study, 36 Gy CSRT and HDC with cyclophosphamide, cisplatin, and vincristine with ASCR were administered four times every 4 weeks, resulting in an approximately 70% 5-year EFS without treatment-related mortality [7,26]. In the Milan study, two cycles of HDC were administered according to the response to pre-HFRT, which showed similar outcomes [27]. In the PNET HR+5 studies, two cycles of high-dose thiotepa with ASCR showed 76% of 5-year PFS without treatment-related mortality [28]. In relapsed patients, HDC with thiotepa, busulfan, and melphalan for myeloablation and RT could be salvaging treatments for newly relapsed patients [29].

In this study, two and four cycles of chemotherapy were administered before and after lower RT for patients in the HR, respectively. Furthermore, chemotherapy was administered based on the higher total intensity of alkylating drugs such as cyclophosphamide, ifosfamide, and platinum analogs than those used in previous studies. To compensate for using a lower RT intensity than that used in previous studies, we administered tandem HDC with ASCR for HR patients [15]. Two cycles of tandem HDC were included in the treatment course for consolidation, with an interval of at least 12 weeks to reduce complications from HDC [18]. Compared to the studies on SJMB, the number of HDC with ASCR was lower, however, the whole intensity of the conditioning chemotherapy was higher in this study [26]. The 5-year EFS and OS rates in the HR group in our study were similar to those observed in studies of standard CSRT without HDC [3,22,23] or lower total dose of HDC [7,26,27].

In fact, in HR patients treated with the KSPNO S081 regimen, there was no difference in the 5-year DSS and OS between patients with a dose modification of less than 80% and patients in the planned dose group (S3 Table). Treatment-related mortalities occurred frequently during the second HDC with ASCR until 2014, when the planned HDC dose was reduced. The 5-year OS of the group administered > 80% dose intensity was 60%, and the 5-year DSS of this group was 15% higher than the 5-year OS. Moreover, there was a higher 5-year OS tendency in the group administered ≤ 80% of the HDC dose intensity than there was in the other group. Consequently, a dose intensity reduction of tandem HDC with ASCR might be reasonable.

A large national database analysis confirmed that patients who underwent STR or biopsy did not have inferior survival outcomes to those who underwent GTR or NTR. In this study, there was no significant difference in the outcomes between the two groups [30]. This observation indicates that insufficient surgical resection could be overcome by additional RT for residual tumors. Patients with a metastatic status are treated with chemotherapy before RT, HDC with ASCR, and maintenance chemotherapy, but this strategy is consistently associated with an inferior outcome [13,22]. We attempted to overcome this inadequate response by using a higher-intensity alkylating agent and tandem HDC with ASCR, but it did not show superior outcomes to those of previous studies.

Molecular subgroup genetic and methylation analyses are an important strategy for determining survival outcomes, and the clinical characteristics of each group have also been analyzed [31,32]. In addition to the previously known molecular subgroups, such as WNT, SHH, group 3, and group 4, a more subdivided classification is suggested to improve disease risk stratification and find a better treatment strategy [24]. At the time these protocols were developed, the molecular subgroup had not been incorporated, although the subgroup was analyzed using immunohistochemical staining. However, WNT subgroups showed superior outcomes, whereas those with SHH with TP53 mutation had inferior outcomes, similar to previously reported findings [32].

Some limitations of this study need to be addressed. First, risk stratification was based on age, residual disease, and metastatic lesions as conventional clinical parameters. Second, the old specimens from the resection of the tumor were not classified according to the revised World Health Organization (WHO) histological classification. Molecular subgroups were only classified in half and only for WNT and SHH with or without TP53 mutation. Finally, the number of enrolled patients was small, and the comparison of the treatment’s effectiveness was difficult because of a single-armed and single-center study. Despite these limitations, this cohort underwent treatment using a unified protocol. Treatment with the KSPNO protocol resulted in excellent survival outcomes in children with MB. KSPNO plans to develop a future protocol for risk stratification with molecular subgroups and risk-adapted treatment. Designing the future protocol by adjusting RT according to the risk group and appropriate HDC with ASCR, may be key for further studies.

Electronic Supplementary Material

Supplementary materials are available at Cancer Research and Treatment website (https://www.e-crt.org).

Notes

Ethical Statement

This study was approved by the Institutional Review Board (IRB) of Yonsei University Health System Clinical Trials Center (IRB No. 4-2022-0634). Informed consent was waived because of the retrospective analysis of anonymous clinical data.

Author Contributions

Conceived and designed the analysis: Kim DS, Suh CO, Lyu CJ, Han JW.

Collected the data: Ahn WK, Lee J, Kim SH, Han JW.

Contributed data or analysis tools: Ahn WK, Hahn SM, Yoon HI, Lee J, Park EK, Shim KW, Kim DS, Suh CO, Kim SH, Lyu CJ, Han JW.

Performed the analysis: Ahn WK, Kim SH, Han JW.

Wrote the paper: Ahn WK, Han JW.

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

References

1. Kaatsch P, Rickert CH, Kuhl J, Schuz J, Michaelis J. Population-based epidemiologic data on brain tumors in German children. Cancer 2001;92:3155–64.
2. Ostrom QT, Price M, Ryan K, Edelson J, Neff C, Cioffi G, et al. CBTRUS statistical report: pediatric brain tumor foundation childhood and adolescent primary brain and other central nervous system tumors diagnosed in the United States in 2014-2018. Neuro Oncol 2022;24:iii1–38.
3. Tarbell NJ, Friedman H, Polkinghorn WR, Yock T, Zhou T, Chen Z, et al. High-risk medulloblastoma: a pediatric oncology group randomized trial of chemotherapy before or after radiation therapy (POG 9031). J Clin Oncol 2013;31:2936–41.
4. Esbenshade AJ, Kocak M, Hershon L, Rousseau P, Decarie JC, Shaw S, et al. A phase II feasibility study of oral etoposide given concurrently with radiotherapy followed by dose intensive adjuvant chemotherapy for children with newly diagnosed high-risk medulloblastoma (protocol POG 9631): a report from the Children’s Oncology Group. Pediatr Blood Cancer 2017;64e26373.
5. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol 2021;23:1231–51.
6. Ellison D. Classifying the medulloblastoma: insights from morphology and molecular genetics. Neuropathol Appl Neurobiol 2002;28:257–82.
7. Gajjar A, Chintagumpala M, Ashley D, Kellie S, Kun LE, Merchant TE, et al. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): long-term results from a prospective, multicentre trial. Lancet Oncol 2006;7:813–20.
8. Merchant TE, Kun LE, Krasin MJ, Wallace D, Chintagumpala MM, Woo SY, et al. Multi-institution prospective trial of reduced-dose craniospinal irradiation (23.4 Gy) followed by conformal posterior fossa (36 Gy) and primary site irradiation (55.8 Gy) and dose-intensive chemotherapy for average-risk medulloblastoma. Int J Radiat Oncol Biol Phys 2008;70:782–7.
9. Zeltzer PM, Boyett JM, Finlay JL, Albright AL, Rorke LB, Milstein JM, et al. Metastasis stage, adjuvant treatment, and residual tumor are prognostic factors for medulloblastoma in children: conclusions from the Children’s Cancer Group 921 randomized phase III study. J Clin Oncol 1999;17:832–45.
10. Taylor RE, Bailey CC, Robinson KJ, Weston CL, Walker DA, Ellison D, et al. Outcome for patients with metastatic (M2-3) medulloblastoma treated with SIOP/UKCCSG PNET-3 chemotherapy. Eur J Cancer 2005;41:727–34.
11. Kortmann RD, Kuhl J, Timmermann B, Mittler U, Urban C, Budach V, et al. Postoperative neoadjuvant chemotherapy before radiotherapy as compared to immediate radiotherapy followed by maintenance chemotherapy in the treatment of medulloblastoma in childhood: results of the German prospective randomized trial HIT ‘91. Int J Radiat Oncol Biol Phys 2000;46:269–79.
12. Packer RJ, Goldwein J, Nicholson HS, Vezina LG, Allen JC, Ris MD, et al. Treatment of children with medulloblastomas with reduced-dose craniospinal radiation therapy and adjuvant chemotherapy: a Children’s Cancer Group Study. J Clin Oncol 1999;17:2127–36.
13. Michalski JM, Janss AJ, Vezina LG, Smith KS, Billups CA, Burger PC, et al. Children’s oncology group phase III trial of reduced-dose and reduced-volume radiotherapy with chemotherapy for newly diagnosed average-risk medulloblastoma. J Clin Oncol 2021;39:2685–97.
14. Christopherson KM, Rotondo RL, Bradley JA, Pincus DW, Wynn TT, Fort JA, et al. Late toxicity following craniospinal radiation for early-stage medulloblastoma. Acta Oncol 2014;53:471–80.
15. Lee JW, Lim DH, Sung KW, Cho HW, Ju HY, Hyun JK, et al. Promising survival rate but high incidence of treatment-related mortality after reduced-dose craniospinal radiotherapy and tandem high-dose chemotherapy in patients with high-risk medulloblastoma. Cancer Med 2020;9:5807–18.
16. Ellison DW, Dalton J, Kocak M, Nicholson SL, Fraga C, Neale G, et al. Medulloblastoma: clinicopathological correlates of SHH, WNT, and non-SHH/WNT molecular subgroups. Acta Neuropathol 2011;121:381–96.
17. Sung KW, Lim DH, Shin HJ. Tandem high-dose chemotherapy and autologous stem cell transplantation in children with brain tumors: review of single center experience. J Korean Neurosurg Soc 2018;61:393–401.
18. Sung KW, Lee SH, Yoo KH, Jung HL, Cho EJ, Koo HH, et al. Tandem high-dose chemotherapy and autologous stem cell rescue in patients over 1 year of age with stage 4 neuroblastoma. Bone Marrow Transplant 2007;40:37–45.
19. Packer RJ, Zhou T, Holmes E, Vezina G, Gajjar A. Survival and secondary tumors in children with medulloblastoma receiving radiotherapy and adjuvant chemotherapy: results of Children’s Oncology Group trial A9961. Neuro Oncol 2013;15:97–103.
20. Packer RJ, Gajjar A, Vezina G, Rorke-Adams L, Burger PC, Robertson PL, et al. Phase III study of craniospinal radiation therapy followed by adjuvant chemotherapy for newly diagnosed average-risk medulloblastoma. J Clin Oncol 2006;24:4202–8.
21. Lannering B, Rutkowski S, Doz F, Pizer B, Gustafsson G, Navajas A, et al. Hyperfractionated versus conventional radiotherapy followed by chemotherapy in standard-risk medulloblastoma: results from the randomized multicenter HIT-SIOP PNET 4 trial. J Clin Oncol 2012;30:3187–93.
22. von Bueren AO, Kortmann RD, von Hoff K, Friedrich C, Mynarek M, Muller K, et al. Treatment of children and adolescents with metastatic medulloblastoma and prognostic relevance of clinical and biologic parameters. J Clin Oncol 2016;34:4151–60.
23. Leary SE, Packer RJ, Li Y, Billups CA, Smith KS, Jaju A, et al. Efficacy of carboplatin and isotretinoin in children with high-risk medulloblastoma: a randomized clinical trial from the Children’s Oncology Group. JAMA Oncol 2021;7:1313–21.
24. Bailey S, Andre N, Gandola L, Massimino M, Rutkowski S, Clifford SC. Clinical trials in high-risk medulloblastoma: evolution of the SIOP-Europe HR-MB trial. Cancers (Basel) 2022;14:374.
25. Gururangan S, Krauser J, Watral MA, Driscoll T, Larrier N, Reardon DA, et al. Efficacy of high-dose chemotherapy or standard salvage therapy in patients with recurrent medulloblastoma. Neuro Oncol 2008;10:745–51.
26. Gajjar A, Robinson GW, Smith KS, Lin T, Merchant TE, Chintagumpala M, 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;39:822–35.
27. Gandola L, Massimino M, Cefalo G, Solero C, Spreafico F, Pecori E, et al. Hyperfractionated accelerated radiotherapy in the Milan strategy for metastatic medulloblastoma. J Clin Oncol 2009;27:566–71.
28. Dufour C, Foulon S, Geoffray A, Masliah-Planchon J, Figarella-Branger D, Bernier-Chastagner V, et al. Prognostic relevance of clinical and molecular risk factors in children with high-risk medulloblastoma treated in the phase II trial PNET HR+5. Neuro Oncol 2021;23:1163–72.
29. Butturini AM, Jacob M, Aguajo J, Vander-Walde NA, Villablanca J, Jubran R, et al. High-dose chemotherapy and autologous hematopoietic progenitor cell rescue in children with recurrent medulloblastoma and supratentorial primitive neuroectodermal tumors: the impact of prior radiotherapy on outcome. Cancer 2009;115:2956–63.
30. Chin AL, Moding EJ, Donaldson SS, Gibbs IC, Soltys SG, Hiniker SM, et al. Survival impact of postoperative radiotherapy timing in pediatric and adolescent medulloblastoma. Neuro Oncol 2018;20:1133–41.
31. Taylor MD, Northcott PA, Korshunov A, Remke M, Cho YJ, Clifford SC, et al. Molecular subgroups of medulloblastoma: the current consensus. Acta Neuropathol 2012;123:465–72.
32. Ramaswamy V, Taylor MD. Medulloblastoma: from myth to molecular. J Clin Oncol 2017;35:2355–63.

Article information Continued

Fig. 1.

The Korean Society of Pediatric Neuro-Oncology (KSPNO) protocol scheme for medulloblastoma. The KSPNO M051 protocol for the standard-risk group (A) and the KSPNO S081 protocol for the high-risk group (B). CSRT, craniospinal radiotherapy; CTx, chemotherapy; HD, high-dose; RTx, radiotherapy.

Fig. 2.

Event-free survival and overall survival for patients according to the risk group (A, B) and molecular subgroup (C, D).

Table 1.

Chemotherapy regimen

Chemotherapy regimen
KSPNO M051
 Course A (1-8 cycles) Cisplatin 75 mg/m2 on D1
Vincristine 1.5 mg/m2 on D1, 8, 15
Cyclophosphamide 1,000 mg/m2 on D2, 3
 Course B (9-12 cycles) Vincristine 1.5 mg/m2 on D1, 8, 15
Cyclophosphamide 1,000 mg/m2 on D1, 2
KSPNO S081
 Course A (1, 3, 5 cycles)a) Cisplatin 90 mg/m2 on D1
Etoposide 75 mg/m2 on D1, 2, 3
Cyclophosphamide 1,500 mg/m2 on D1, 2
Vincristine 1.5 mg/m2 on D1, 8
 Course B (2, 4, 6 cycles)a) Carboplatin 300 mg/m2 on D1, 2
Etoposide 75 mg/m2 on D1, 2, 3, 4, 5
Ifosfamide 1,500 mg/m2 on D1, 2, 3, 4, 5
Vincristine 1.5 mg/m2 on D1, 8
 First HDC CTE Carboplatin 500 mg/m2 on D1, 2, 3
Thioetepa 300 mg/m2 on D4, 5, 6
Etoposide 250 mg/m2 on D4, 5, 6
 Second HDC CM Cyclophosphamide 1,500 mg/m2 on D1, 2, 3, 4
Melphalan 60 mg/m2 on D5, 6, 7

HDC, high-dose chemotherapy; KSPNO, Korean Society of Pediatric Neuro-Oncology.

a)

Dose reduction of 75% on 3-6 cycles after radiotherapy.

Table 2.

Demographics of the study patients at diagnosis

Characteristic SR (n=24) HR (n=35) Total (n=59)
Age (yr) 9.8 (6.4-12.3) 5.9 (4.0-9.5) 8.1 (4.75-11.1)
Sex
 Male 17 (70.8) 22 (62.9) 39 (66.1)
 Female 7 (29.2) 13 (37.1) 20 (33.9)
Histology
 Classic 21 (87.5) 29 (82.9) 50 (84.7)
 Desmoplastic/Nodular 2 (8.3) 4 (11.4) 6 (10.2)
 MBEN 1 (4.2) 1 (2.9) 2 (3.4)
 Large cell/Anaplastic 0 1 (2.9) 1 (1.7)
Molecular
 WNT 1 (4.2) 1 (2.9) 2 (3.4)
 SHH with TP53 1 (4.2) 3 (8.6) 4 (6.8)
 SHH without TP53 1 (4.2) 1 (2.9) 2 (3.4)
 Non-Wnt/Non-SHH 10 (41.7) 18 (51.4) 28 (47.5)
 Not done 11 (45.8) 12 (34.3) 23 (39.0)
T category
 1 1 (4.2) 0 1 (1.7)
 2 8 (33.3) 1 (2.9) 9 (15.3)
 3 15 (62.5) 30 (85.7) 45 (76.3)
 4 0 4 (11.4) 4 (6.8)
M category
 0 24 (100) 6 (17.1) 30 (50.8)
 1 0 5 (14.3) 5 (8.5)
 2 0 4 (11.4) 4 (6.8)
 3 0 20 (57.1) 20 (33.9)
Resection status
 GTR 18 (75.0) 12 (34.3) 30 (50.8)
 NTR 6 (25.0) 6 (17.1) 12 (20.3)
 STR 0 16 (45.7) 16 (27.1)
 Biopsy 0 1 (2.9) 1 (1.7)
R stage
 R0 18 (75.0) 12 (34.3) 30 (50.8)
 R1 6 (25.0) 23 (65.7) 29 (49.2)
Protocol
 KSPNO M051 23 (95.8) 2 (5.7) 25 (42.4)
 KSPNO S081 1 (4.1) 33 (94.3) 34 (57.6)
RT dose
 Tumor bed 55.8 (54.0-55.8) 54.0 (54.0-55.3) 54.8 (54.0-55.8)
 CSRT 23.4 (23.4-24.2) 30.6 (30.3-30.6) 30.6 (23.4-30.6)
Intensity of chemotherapy
 Chemotherapy 93.2 (76.8-103.3) 86.9 (78.1-92.2) -
 HDC - 80.0 (71.3-90.0) -
 Total - 85.1 (74.7-91.7) -
Time from operation to other treatment (mo)a) 0.81 (0.69-0.93) 0.59 (0.36-0.78) 0.72 (0.43-0.91)
Follow-up (yr) 8.2 (5.0-11.4) 5.4 (2.1-8.5) 6.4 (2.8-10.3)

Values are presented as median (IQR) or number (%). CSRT, craniospinal radiotherapy; GTR, gross total resection; HDC, high-dose chemotherapy; HR, high-risk group; IQR, interquartile range; KSPNO, Korean Society of Pediatric Neuro-Oncology; MBEN, Medulloblastoma with extensive nodularity; NTR, nearly total resection; RT, radiotherapy; SHH, sonic hedgehog; SR, standard-risk group; STR, subtotal resection; WNT, wingless.

a)

Time taken from operation to radiotherapy in KSPNO M051 regimen, and time taken from operation to chemotherapy in KSPNO S081.

Table 3.

Difference of survival rate according to risk groupa), extension of resection and M stage

5-Year EFS (%)b) 95% CI (%) p-value 5-Year OS (%)b) 95% CI (%) p-value
Risk group
 SR (n=23) 86.00 72.5-100 0.108 95.70 87.7-100 0.074
 HR (n=33) 66.40 52.0-84.8 68.30 53.8-86.7
Extent of resection
 GTR+NTR (n=40) 79.40 67.6-93.2 0.368 86.70 76.5-98.3 0.286
 STR+biopsy (n=16) 62.50 42.8-91.4 62.50 42.8-91.4
M-stagec)
 M0 (n=27) 84.40 71.4-99.7 0.286 92.40 82.9-100 0.302
 M1 (n=5) 80.00 51.6-100 80.00 51.6-100
 M2 (n=4) 50.00 18.8-100 66.70 30.0-100
 M3 (n=20) 64.60 46.6-89.6 64.30 46.2-89.5
 M0-1 (n=32) 83.70 71.6-97.9 0.125 90.40 80.7-100 0.130
 M2-4 (n=24) 62.20 45.5-85.1 64.70 47.7-87.8
Molecular subgroups
 WNT (n=2) 100 - 0.021 100 - 0.011
 SHH with TP53 (n=4) - - - -
 SHH without TP53 (n=2) - - 100 -
 Non-WNT/Non-SHH (n=26) 72.40 56.9-92.2 79.50 64.8-97.4

CI, confidence interval; EFS, event-free survival; GTR, gross total resection; HR, high-risk; KSPNO, Korean Society of Pediatric Neuro-Oncology; NTR, nearly total resection; OS, overall survival; SHH, sonic hedgehog; SR, standard-risk; STR, subtotal resection; WNT, wingless integrated.

a)

Patients treated with KSPNO M051 in the SR and patients treated with KSPNO S081 in the HR,

b)

Kaplan-Meier method with log-rank test,

c)

Metastastic stage according to Chang’s stage.

Table 4.

Dose modification and 5-year outcomes in KSPNO M051 and S081

5-Year PFS (%)a) 95% CI (%) p-value 5-Year DSS (%)a) 95% CI (%) p-value 5-Year OS (%)a) 95% CI (%) p-value
KSPNO M051 with SRb)
 > 80% (n=16) 100 - 0.033 100 - 0.041 93.8 82.6-100 0.192
 ≤ 80% (n=7) 68.6 40.3-100 100 - 100 -
KSPNO S081 with HRc) chemotherapy
 > 80% (n=23) 71.3 54.2-93.8 0.205 73.3 55.7-96.4 0.268 63.3 46.0-87.2 0.257
 ≤ 80% (n=10) 90.0 73.2-100 90.0 73.2-100 80.0 58.7-100
HDCd)
 > 80% (n=15) 76.4 56.0-100 0.500 75.0 54.1-100 0.658 60.0 39.7-90.7 0.766
 ≤ 80% (n=15) 73.3 54.0-99.5 78.6 59.8-100 70.7 50.2-99.6
Totale)
 > 80% (n=19) 76.4 58.5-99.8 0.972 79.4 61.2-100 0.743 66.2 47.4-92.4 0.892
 ≤ 80% (n=11) 72.7 50.6-100 72.7 50.6-100 63.6 40.7-99.5

CI, confidence interval; DSS, disease-specific survival; HDC, high-dose chemotherapy; HR, high-risk; KSPNO, Korean Society of Pediatric Neuro-Oncology; OS, overall survival; PFS, progression-free survival; SR, standard-risk.

a)

Kaplan-Meier method with log-rank test,

b)

Patients in the SR, treated with KSPNO M051 regimen,

c)

Patients in the HR, treated with KSPNO S081 regimen,

d)

Patients treated with tandem HDC with autologous stem cell rescue,

e)

Dose considering the total of chemotherapy and HDC.

Table 5.

Grade 3 and 4 adverse events related with treatment

KSPNO M051 (n=25)
KSPNO S081 (n=34)
Grade 3 Grade 4 Grade 3 Grade 4
Hematological toxicity (before HDC for HR)
 Neutropenic fever 17 (68.0) 2 (8.0) 18 (52.9) 12 (35.3)
 Neutropenia 7 (28.0) 16 (64.0) 4 (11.8) 28 (82.4)
 Anemia 17 (68.0) 2 (8.0) 29 (85.3) 1 (2.9)
 Thrombocytopenia 5 (20.0) 15 (60.0) 6 (17.6) 24 (70.6)
Infectious toxicity
 Mucositis 2 (8.0) 0 1 (2.9) 0
 Sepsis 2 (8.0) 0 8 (23.5) 1 (2.9)
 Pneumonia 2 (8.0) 2 (8.0) 0 0
 Meningitis 0 0 1 (2.9) 0
Gastrointestinal toxicity
 Vomiting 0 0 0 1 (2.9)
 Gastrointestinal bleeding 1 (4.0) 0 3 (8.8) 0
 Colitis 1 (4.0) 0 1 (2.9) 0
 Cholangitis 0 0 1 (2.9) 0
Hepatobiliary toxicity
 Hepatitis 1 (4.0) 0 2 (5.9) 0
 VOD 0 0 0 0
 Pancreatitis 0 0 0 0
Neurologic toxicity
 Headache 3 (12.0) 0 2 (5.9) 0
 Hearing impairment 4 (16.0) 0 4 (11.8) 0
 Neuropathy 2 (8.0) 0 0 0
 Seizure 0 0 1 (2.9) 0
 Tremor 1 (4.0) 0 0 0
Pulmonary toxicity
 Asthma 0 0 2 (5.9) 0
Endocrinologic toxicity
 Hypothyroidism 5 (20.0) - 10 (29.4) -
 Short status 9 (36.0) - 12 (35.3) -
 Precocious puberty 3 (12.0) - 6 (17.6) -
 Adrenal insufficiency 3 (12.0) - 2 (5.9) -

Values are presented as number (%). HDC, high-dose chemotherapy; HR, high-risk; KSPNO, Korean Society of Pediatric Neuro-Oncology; VOD, veno-occlusive disease.

Table 6.

Grade 3 and 4 adverse events related with HDC with ASCR

1st HDC with ASCR (n=31) 2nd HDC with ASCR (n=27)
Hematologic toxicity
 CD34+ cell (×106/kg) 8.0 (4.5-15.5) 12.0 (5.0-20.5)
 Intensity of chemotherapy 100 (80-100) 80 (80-95.0)
 Days to reach ANC 1,000/μL 11.0 (10.0-11.0) 11 (10.0-12.0)
 Days of fever ≥ 38.0℃ 5.0 (3.0-6.0) 3.0 (1.0-4.5)
Cardiovascular toxicity
 Shock 2 (6.5) 4 (14.8)
Infectious toxicity
 Septicemia 2 (6.5) 7 (25.9)
 Pneumonia 1 (3.2) 2 (7.4)
 Mucositis 3 (9.7) 1 (3.7)
 Cellulitis 1 (3.2) 0
Gastrointestinal toxicity
 Gastrointestinal bleeding 0 1 (3.7)
Hepatobiliary toxicity
 Hepatitis - -
 G3 8 (25.8) 2 (7.4)
 G4 1 (3.2) 0
 VOD 1 (3.2) 2 (7.4)
 Pancreatitis 0 1 (3.7)
Pulmonary toxicity
 Asthma 1 (3.2) 1 (3.7)
Neurologic toxicity
 Shunt malfunction 0 1 (3.7)
 Seizure 0 1 (3.7)
Others
 G3 Hyponatremia 1 (3.2) 0
 Hemorrhagic cystitis 0 1 (3.7)
Death 0 3 (11.1)

Values are presented as median (IQR) or number (%). ANC, absolute neutrophil count; ASCR, autologous stem cell rescue; HDC, high-dose chemotherapy; IQR, interquartile range; VOD, veno-occlusive disease.