Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2008 Oct;10(5):745–751. doi: 10.1215/15228517-2008-044

Efficacy of high-dose chemotherapy or standard salvage therapy in patients with recurrent medulloblastoma

Sridharan Gururangan 1,, Jeanne Krauser 1, Melody A Watral 1, Tim Driscoll 1, Nicole Larrier 1, David A Reardon 1, Jeremy N Rich 1, Jennifer A Quinn 1, James J Vredenburgh 1, Annick Desjardins 1, Roger E McLendon 1, Herbert Fuchs 1, Joanne Kurtzberg 1, Henry S Friedman 1
PMCID: PMC2666251  PMID: 18755919

Abstract

The efficacy of high-dose chemotherapy (HDC) or standard salvage therapy was evaluated in patients with recurrent medulloblastoma (MBL) using retrospective chart review of all patients with recurrent MBL treated at Duke University Medical Center between 1995 and 2005 and who had undergone HDC with or without radiotherapy (RT) or standard salvage therapy after relapse. A total of 30 patients were diagnosed with recurrent MBL after standard RT alone or chemotherapy with RT. Nineteen patients (7 who received no RT before recurrence [group A] and 12 who received definitive RT before recurrence [group B]) underwent surgery and/or induction chemotherapy followed by HDC plus autologous stem-cell rescue. Eleven patients (group C) underwent standard salvage therapy. Six of seven group A patients also received standard RT just before or after recovery from HDC, and 5 of 12 group B patients received adjuvant palliative focal RT post-HDC. At a median follow-up of 28 months, three of seven patients in group A are alive and disease-free at ⩾34, ⩾110, and ⩾116 months, respectively, post-HDC. All patients in groups B and C have died of tumor, at a median of 35 months and 26 months from HDC and standard salvage therapy, respectively. HDC or standard salvage therapy was ineffective in our patients with recurrent MBL who had received standard RT before recurrence. The favorable impact of HDC on disease control in the two long-term survivors cannot be clearly established due to the cofounding effect of definitive RT postrecurrence.

Keywords: angiogenesis, Avastin, bevacizumab, bio-markers, glioblastoma, glioma


Medulloblastoma (MBL) is the most common malignant embryonal brain tumor in children, with an incidence of approximately 0.5 per million children and an average of 400 cases per year in the United States.1 Although significant advances have been made in the treatment of children with this aggressive malignancy, especially for older children with localized disease (average risk),2,3 the prognosis remains dismal for infants (children <3 years of age) and those with extensive or recurrent tumors.4 Alkylator-based high-dose chemotherapy (HDC) with stem-cell rescue has been used in children with recurrent MBL for the last several years with modest success.510 However, the true impact of this strategy on long-term disease control is hard to interpret in these studies because most survivors had also received adjuvant focal or focal plus craniospinal radiotherapy (RT) around HDC. We therefore did a retrospective study to (1) evaluate the efficacy of HDC in children with recurrent MBL treated at our institution based on having received definitive RT before recurrence and (2) measure outcomes of those who had only standard salvage therapy at relapse.

Patients and Methods

Between 1995 and 2005, a total of 30 patients were treated for recurrent MBL at Duke University Medical Center. Nineteen patients underwent HDC with autologous hematopoietic stem-cell rescue (ASCR) after recurrence. Seven of the 19 patients who underwent HDC had not received standard RT (focal and/or craniospinal RT) before recurrence due to their young age (group A). The other 12 older patients had received standard RT with or without chemotherapy at initial diagnosis and before recurrence (group B). Eleven of 30 patients received only standard salvage therapy after tumor recurrence (group C). Informed consent as approved by the local institutional review board (IRB) was obtained for all patients before commencement of HDC. This retrospective study received formal IRB review and approval before database query and chart review.

Diagnosis and Initial Workup

All patients were subjected to biopsy and/or surgical resection of the primary tumor at diagnosis and relapse. Pathologic diagnosis of MBL was made by one of us (R.E.M.) using standard criteria.11 All patients underwent a metastatic workup either before or 3 weeks post-surgery that included MRI of brain and spine with and without gadolinium, cerebrospinal fluid (CSF) cytology obtained via lumbar puncture, Tc99m bone scan, and bone marrow aspirate and biopsy. Metastatic spread of tumor was classified as CSF spread only (M1 disease), nodular disease in the brain or spine only (M2 disease), nodular disease in brain and spine (M3 disease), and extraneural spread (M4 disease).

Induction Chemotherapy, RT, and HDC with ASCR

Details of type and dosage schedules of induction chemotherapy, RT, and HDC used in these patients after recurrence are summarized in Table 1. Standard salvage chemotherapy included agents known to be effective in MBL, such as cyclophosphamide (CTX), oral or intravenous etoposide (VP-16), platinum compounds, high-dose methotrexate, CPT-11 (Camptosar, Pfizer Corporation, New York, NY, USA), and temozolomide (Temodar, Schering Plough Corporation, Kenilworth, NJ, USA); in two patients, investigational agents included intrathecal Spartaject busulfan (BU; SuperGen, Inc., San Ramon, CA, USA) or VNP40101M (Cloretazine, Vion Pharmaceuticals, New Haven, CT, USA). Response assessment was made by assessing tumor size (derived from the product of the maximal tumor diameters) on a gadolinium-enhanced MRI of brain and/or spine obtained regularly during treatment. Response criteria were as follows: complete response, disappearance of all tumor and no new lesions; partial response, ⩾50% reduction in tumor size; minimal response, 25%–50% reduction in tumor size; stable disease, <25% increase or decrease in tumor size; progressive disease, ⩾25% increase in tumor size and/or appearance of new lesions.

Table 1.

Details of type and dosage schedules of induction chemotherapy, radiotherapy, and high-dose chemotherapy used in patients with recurrent medulloblastoma

Induction chemotherapy (agents used either alone or in combination as indicated in Tables 2 and 3)
 High-dose cyclophosphamide 2 gm/m2/day i.v. for 2 days with mesna rescue and hydration, given every 4 weeks for four cycles
 Vincristine 1.5 mg/m2 (0.05 mg/kg in children <10 kg) i.v. on day 1 and then weekly for 2 weeks, cisplatin 75 mg/m2 (2.5 mg/kg in children <3 years) i.v. on day 1, cyclophosphamide 2 gm/m2 (65 mg/kg in children <3 years) i.v. on day 2, and etoposide 100 mg/m2 (4 mg/kg for children <3 years) i.v. on days 2 and 3 every 3 weeks
 Carboplatin (dose based on Calvert’s formula using glomerular filtration rate and an area under the curve concentration of 5 mg/ml per minute) i.v. on day 1 and etoposide 100 mg/m2 i.v. on days 2 and 3 every 4 weeks
 Oral etoposide 50 mg/m2 per day for 21 days every 4 weeks
Radiotherapy
 Craniospinal: Treatment administered in the prone position using a thermoplast stabilization device with a 6 MV photon beam and customized blocking (general anesthesia or sedation used as necessary), given at 1.5–1.8 Gy per fraction
 Posterior fossa or three-dimensional conformal: Treatment to the posterior fossa or tumor bed given in the supine position at 1.8–2 Gy per fraction
High-dose chemotherapy with autologous hematopoietic stem- cell rescue
 Bone marrow stem cell harvest: Bone marrow harvest performed under general anesthesia after the first or second cycle of induction chemotherapy after granulocyte-colony-stimulating factor, frozen and stored using dimethyl sulfoxide as a cryopreservative; peripheral blood stem cells collected through a double-lumen catheter on alternate days for 3 days, with CD34+ cells separated, frozen, and stored
 High-dose chemotherapy: All patients received one of the following high-dose chemotherapy regimens:
 (a) Cyclophosphamide 50 mg/kg daily for 4 days followed by melphalan 60 mg/m2 per day for 3 days
 (b) Busulfan 1 mg/kg every 6 h for 16 total doses over 4 days (plasma concentrations measured and doses adjusted to yield a steady-state concentration of 600–900 ng/ml) followed by melphalan 60 mg/m2 per day for 3 days
 (c) Carboplatin (either 500 mg/m2 or a dose based on Calvert’s formula to achieve an area under the curve concentration of 7 mg/ml per minute, whichever was less) on days –8, –7, and –6, followed by thiotepa 300 mg/m2 and etoposide 250 mg/m2 daily on days –5, –4, and –3
Stem-cell rescue: Three days after the last dose of chemotherapy, frozen bone marrow and peripheral blood stem cells, thawed at room temperature, infused through a double-lumen broviac catheter

Evaluation before transplant in those undergoing HDC included physical examination; pulmonary function tests including diffusing capacity of the lung for carbon monoxide; electrocardiogram, echocardiogram, and resting multiple uptake gated acquisition scan; and antiviral antibody titers (cytomegalovirus, hepatitis C virus, varicella zoster virus, and hepatitis B and C). Supportive care after transplant included granulocyte colony-stimulating factor; intravenous antibiotics; blood products as needed; pain control; intravenous hyper-alimentation; antibiotic prophylaxis for Pneumocystis pneumonia, herpes simplex, and varicella zoster virus for up to 6 months after transplant; and prophylaxis for veno-occlusive disease with low-dose heparin. Patients were followed by the bone marrow transplant (BMT) service for at least 6 months after discharge. MRI scan of brain and spine was obtained 6 weeks after BMT and periodically thereafter.

Statistical Analysis

Overall survival (OS) and progression-free survival (PFS) were determined using the Kaplan-Meier product limit method.12 OS was calculated from the date of diagnosis until death from any cause or last follow-up. PFS was calculated from the date of diagnosis until death from disease progression, death from any cause, or last follow-up.

Results

Patients Who Received No RT before Relapse and HDC, Group A (n = 7)

The median ages at diagnosis and relapse were 2 years (range, 2–7 years) and 4 years (range, 3–7 years), respectively. All patients had received standard chemotherapy only before relapse due to their young age. The median time to progression from initial diagnosis was 6 months (range, 3–16 months), with five of seven patients (71%) suffering a local relapse (Table 2). All seven patients achieved minimal residual disease (MRD) before HDC with surgery, chemotherapy, and RT (n = 4), RT only (n = 2), or surgery + chemotherapy (n = 1) (Table 2). The myeloablative regimens included BU + melphalan (MEL) in five patients, CTX + MEL in one, and carboplatin (CARBO) + VP-16 + thiotepa (TT) in one. At a median follow-up of 28 months (range, 4 to ⩾116 months), only patients 1, 2, and 3 (Table 2) are alive and disease-free after HDC. Patients 1 and 2 also received adjuvant craniospinal RT (30–36 Gy) and focal boost (54 Gy) to the primary site after relapse. Patient 3 (Table 2) was diagnosed with Gorlin’s syndrome after diagnosis of MBL, and RT was withheld despite relapse in view of the risk of inducing secondary malignancies due to radiation exposure. The remaining four patients died of progressive disease despite receiving adequate doses of RT before HDC at a median of 7 months post-HDC (range, 4–37 months; Table 2). The 3-year OS for this group is 14% (95% confidence interval, 0%–30%) (Fig. 1).

Table 2.

Clinical characteristics, treatment, and outcome in seven patients with recurrent medulloblastoma treated with high-dose chemotherapy (group A)

Patient No. Age at Diagnosis (Years) M Stage at Diagnosis Rx prior to Relapse Interval from Diagnosis to Relapse (Months) Site(s) of Relapse Chemo Post-relapse RT Post-replapse HDC Regimen Relapse Post-HDC Interval from HDC to Relapse (Months) Final Outcome
1 2 M0 Chemo 16 Local CARBO 1+VP-16 CSI + focal BU + MEL None 116+ Alive NED
2 2 Gorlin’s Syndrome M0 Chemo 15 Local HD CTX None BU + MEL None 34+ Alive NED
3 2 M0 Chemo 6 Local Oral VP-16 CSI + focal BU + MEL None 110+ Alive NED
4 3 M0 Chemo 15 Local Oral VP-16 Focal CTX + MEL LMD 4 Dead
5 2 M0 Chemo 6 Local HD CTX CSI + focal BU + MEL Spine 7 Dead
6 4 M1 Chemo 6 Brain, CSF None CSI1 focal BU + MEL LMD 37 Dead
7 7 M3 Chemo 3 Local, CSF None CSI1 focal CARBO, TT, VP-16 LMD, BM 7 Dead

Abbreviations: Rx, treatment; Chemo, chemotherapy; RT, radiotherapy; HDC, high-dose chemotherapy; CARBO, carboplatin; VP-16, etoposide; CSI, cerebrospinal irradiation; BU, busulfan; MEL, melphalan; NED, no evidence of disease; HD, high dose; CTX, cyclophosphamide; LMD, leptomeningeal disease; TT, thiotepa; BM, bone marrow.

Fig. 1.

Fig. 1

Overall survival for patients in groups A, B, and C.

Patients Who Received Definitive RT before Relapse and HDC, Group B (n = 12)

The median ages at diagnosis and relapse were 7.5 years (range, 5–12 years) and 12 years (range, 8–19 years), respectively. All patients had received surgery and definitive RT with or without chemotherapy before relapse. The median time to progression from initial diagnosis was 44 months (range, 15–140 months), with 5 of 12 patients (42%) suffering a local relapse (Table 3). Eleven of 12 patients achieved MRD before HDC with chemotherapy alone (n = 9), surgery + chemotherapy + RT (n = 5), and surgery + chemotherapy alone (n = 4; Table 3). The myeloablative regimens included CTX + MEL in nine patients and BU + MEL in three patients. At a median follow-up of 35 months (range, 7–49 months), all patients have died of progressive disease (Table 3, Fig. 1).

Table 3.

Clinical characteristics, treatment, and outcome in 12 patients with recurrent medulloblastoma treated with high-dose chemotherapy (group B)

Age at Diagnosis (Years) M Stage at Diagnosis Rx prior to Relapse Interval from Diagnosis to Relapse (Months) Site(s) of Relapse Chemo Post-relapse RT Post-replapse HDC Regimen Relapse Post-HDC Interval from HDC to Relapse (Months) Final Outcome
6 M0 S, C, RT 27 Local HD CTX None CTX + MEL LMD 4 Dead
5 M0 S, C, RT 65 Brain HD CTX + oral VP-16 Focal CTX + MEL LMD 16 Dead
7 M3 S, C, RT 25 Brain Oral VP-16 None CTX + MEL LMD 5 Dead
7 M0 S, RT 102 Brain HD CTX, CDDP, + VP-16 Focal BU + MEL Local 12 Dead
12 M0 S, RT 50 Local HD CTX None CTX + MEL LMD 14 Dead
8 M0 S, C, RT 45 Brain + spine HD CTX + oral VP-16 None BU + MEL LMD 7 Dead
6 M0 S, C, RT 52 Brain HD CTX + oral VP-16 Focal CTX + MEL Local + METS 5 Dead
8 M0 S, RT 43 Brain CDDP, CTX, VCR, + VP-16 Focal CTX + MEL LMD 5 Dead
12 M3 S, C, RT 31 Local None None CTX + MEL Local 12 Dead
8 M0 S, C, RT 15 Local None None BU + MEL Local 9 Dead
7 M0 S, RT 140 Local + LMD CTX + VCR CSI CTX + MEL METS 24 Dead
8 M0 S, RT 36 Local HD CTX, CDDP, + VP-16 None CTX + MEL Local + METS 13 Dead

Abbreviations: Rx, treatment; Chemo, chemotherapy; RT, radiotherapy; HDC, high-dose chemotherapy; S, surgery; C, chemotherapy; HD, high dose; CTX, cyclophosphamide; MEL, Melphalan; LMD, leptomeningeal disease; VP-16, etoposide; CDDP, cisplatin; BU, busulfan; METS, metastasis; VCR, vincristine; CSI, cerebrospinal irradiation.

Patients with Recurrent MBL Who Received Standard Salvage Therapy, Group C (n = 11)

The median ages at diagnosis and relapse for this group were 6 years (range, 2–24 years) and 11 years (range, 3–26 years), respectively. Eight patients had received standard therapy, including surgery, RT, and/or chemotherapy, at diagnosis (Table 4). The remaining three patients received surgery and chemotherapy only due to their young age (Table 4). Relapse after initial diagnosis occurred at a median of 18 months (range, 1–85 months) (Table 4). All patients have died at a median period of 26 months (range, 3–112 months) from relapse despite treatment with a variety of standard chemotherapeutic or investigational agents with or without standard RT (Table 4, Fig. 1).

Table 4.

Clinical characteristics, treatment, and outcome in 11 patients with recurrent medulloblastoma treated with standard salvage therapy (group C)

Age at Diagnosis (Years) M Stage at Diagnosis Rx prior to Relapse Interval from Diagnosis to Relapse (Months) Site(s) of Relapse Chemo Postrelapse RT Postrelapse Final Outcome
14 M0 S, C, RT 26 Local + LMD CARBO, VP-16, + CPT-11 None Dead
5 M0 S, C, RT 85 Brain HD CTX + oral VP-16 None Dead
3 M0 S, C, RT 1 Local + METS HD CTX None Dead
10 M0 S, C, RT 14 Local CTX, CDDP, + VP-16 Focal Dead
20 M2 S, RT 23 LMD IT BU + HD TEMO None Dead
4 M2 S, C 1 LMD IT BU CSI + focal Dead
3 M2 S, C 1 LMD None CSI + focal Dead
20 M0 S, C, RT 22 BM CTX + HD MTX None Dead
6 M0 S, C, RT 20 LMD Oral VP-16 + Cloretazine None Dead
2 M3 S, C 13 LMD None None Dead
24 M4 S, C, RT 18 BM HD CTX + oral VP-16 Focal Dead

Abbreviations: Rx, treatment; Chemo, chemotherapy; RT, radiotherapy; S, surgery; C, chemotherapy; LMD, leptomeningeal disease; CARBO, carboplatin; VP-16, etoposide; CPT-11, irinotecan; HD, high-dose; CTX, cyclophosphamide; METS, metastasis; CDDP, cisplatin; IT, intrathecal; BU, busulfan; TEMO, temozolimide; BM, bone marrow; MTX, methotrexate.

Discussion

MBL is curable in a significant proportion of patients with average-risk disease at initial diagnosis.2,13 However, outcome for patients with recurrent disease continues to be suboptimal. Tumor progression on or off therapy is possibly due to emergence of drug-resistant clones, and alkylator-based HDC is one strategy that could potentially overcome this therapeutic obstacle. Alkylators, including CTX, MEL, BU, CARBO, and TT, demonstrate steep log-dose response that is maintained with increasing doses of drug, resulting in progressive depletion of putative tumor stem cells and potential cure.14 Specific properties of alkylators, including high lipid solubility, lack of cross resistance with other alkylators, and synergistic activity with topoisomerase inhibitors such as VP-16 or CPT-11 (Camptosar), are particularly suitable for treatment of patients with CNS malignancies either as single agents or in combination.4 With myelosupression the predominant toxicity from these drugs, it is possible to use myeloablative drug doses followed by ASCR, although nonhematologic toxicities, including liver or lung damage, begin to emerge with increasing doses. Alkylator-based HDC with ASCR has gained wide prevalence in the treatment of patients with recurrent brain tumors, especially MBL.4 Patients with tumors that are localized, chemosensitive, and in MRD before HDC are those who have responded well to this procedure.4 While a subset of patients with recurrent MBL have been shown to have durable disease control, most of them have also received adjuvant RT either before or after HDC, making it difficult if not impossible to assess the true value of this approach. In addition, this treatment has rarely been effective in patients who suffered recurrence after definitive RT or in those with bulky metastatic disease.4,6,10,15

Only 3 of 19 patients with recurrent MBL who received HDC in our study had durable disease control, with two of them being long-term survivors after adjuvant definitive RT. These results seem to support the notion that HDC with regimens used in our study cures only a small number of young children with locally recurrent MBL who also received definitive RT after recurrence. One patient with Gorlin’s syndrome and recurrent localized MBL is also alive and disease-free for ⩾34 months after HDC only (RT was withheld out of concern for increased risk of secondary malignancies), but her follow-up is relatively short. Similar outcomes have been reported previously in the literature in children with recurrent MBL after HDC (Table 5). In a recent publication from St. Jude Children’s Research Hospital, Shih et al.16 had only 3 survivors among 13 patients with recurrent MBL treated with a wide variety of myeloablative regimens (Table 5). All three of these patients also received adjuvant definitive RT along with HDC.16 Similarly, Dunkel et al.5 reported seven disease-free survivors after HDC for recurrent MBL. Five of these seven patients also received definitive RT around HDC, and the remaining two had received RT only before recurrence and were hence chemonaive at the time of HDC. In a report from the Children’s Cancer Group, three of five infants with recurrent MBL, who also received definitive RT along with HDC, were long-term survivors.9 Thus, these studies and ours validate the usefulness of RT in achieving cure in patients with recurrent MBL who had received chemotherapy only before relapse.17,18 In this context, it should be mentioned that RT alone can cure 20%–50% of young children with MBL who suffer recurrence after standard chemotherapy.17,19

Table 5.

Results of prior studies of high-dose chemotherapy in patients with recurrent medulloblastoma

No. of Patients Age (Years) Prerelapse Definitive RT HDC Conditioning Regimen Post-HDC RT Progression-Free Survivors Reference
20 0.75–6 No BU + TT Focal (10) 10/20 Dupuis-Girod et al.6
8 2.5–15 Yes (8) CTX + MEL None 0/8 Mahoney et al.20
18 0.8–27 Yes (18) CTX + MEL BU + MEL CARBO + VP-16 Yes (18) 4/18 Graham et al.8
23 2–44 Yes (16) CARBO, TT, + VP-16 Focal or CSI (7) 7/23 Dunkel et al.5
5 1.5–5.4 No CARBO, TT, + VP-16 Focal + CSI (5) 3/5 Gururangan et al.9
17 0–21 Yes TT × 2 Yes (11) 1/17 Massimino et al.10
13 0–21 Yes (11) No (2) CARBO + VP-16 BU + TT CTX + TT CTX + MEL CTX + TOPO BU + MEL Yes (4) 3/13 Shih et al.16
26 0–21 NA CARBO, TT, + VP-16 Yes (4) 2/26 Bode et al.15

Abbreviations: RT, radiotherapy; HDC, high-dose chemotherapy; BU, busulfan; TT, thiotepa; CTX, cyclophosphamide; MEL, melphalan; CARBO, carboplatin; VP-16, etoposide; CSI, craniospinal irradiation; TOPO, topotecan; NA, not available.

However, HDC was distinctively unsuccessful in improving survival in our 12 patients in group C who suffered relapse after definitive RT with or without chemotherapy, similar to what has been observed in other HDC studies. In a Pediatric Oncology Group study, Mahoney et al.20 treated eight patients with recurrent MBL who had received definitive RT previously with HDC using CTX + MEL and reported no disease-free survivors after the procedure (Table 5). Massimino et al.10 reported in abstract form no disease-free survivors in 17 previously irradiated patients with recurrent MBL who were treated with two sequential doses of high-dose TT and adjuvant reirradiation. The reasons for failure of HDC in these patients are unclear and may be related to lack of eradication of residual tumor stem cells in the local site or neuraxis with relatively large doubling times that might have escaped the cytotoxic effects of the HDC regimen given over a short duration.21 While it is possible that choice of myeloablative alkylator regimens for HDC may have influenced outcomes in our patients, no published studies have clearly shown the superiority of one HDC regimen over another.7,15,16

The outcomes of the 11 patients in our study who were treated with conventional salvage therapy were similarly dismal, which is not surprising since most of these patients had extensive metastatic disease at recurrence after initial RT and/or chemotherapy and were unlikely to have durable disease control with any available therapy.22,23 These patients were intentionally not given HDC due to reported poor survival after this procedure in those with metastatic disease.6 However, the results of our study should be interpreted with caution due to the relatively small number of patients from a single institution, the limited variety of myeloablative regimens used in patients receiving HDC, and variability in the type of salvage chemotherapy used in patients in group C.

The relentless disease progression and ultimate death of 27 patients in our study irrespective of treatment received serves to underscore the fact that recurrent MBL is an invariably fatal disease. There is a desperate need for alternatives to HDC to treat these patients. Future therapies should look beyond dose escalation and focus on treatment options that minimize toxicity, maximize benefit, and maintain quality of life. In this context, metronomic therapies that provide continuous drug exposure to both tumor cells and vasculature over extended periods might provide benefit.21,24,25 Such therapy could be combined with small-molecule kinase inhibitors against rational molecular targets, appropriate for inhibition, that signal tumor angiogenesis, proliferation, and invasion.26

Acknowledgments

This work was presented in part at the 12th annual scientific meeting of the Society for Neuro-Oncology, Dallas, TX, USA, November 15–18, 2007.

References

  • 1.Duffner PK, Cohen ME, Myers MH, Heise HW. Survival of children with brain tumors: SEER Program, 1973–1980. Neurology. 1986;36:597–601. doi: 10.1212/wnl.36.5.597. [DOI] [PubMed] [Google Scholar]
  • 2.Gajjar A, Chintagumpala M, Ashley D, et al. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): long-term results from a prospective, multicentre trial. Lancet Oncol. 2006;7:813–820. doi: 10.1016/S1470-2045(06)70867-1. [DOI] [PubMed] [Google Scholar]
  • 3.Packer RJ, Gajjar A, Vezina G, et al. Phase III study of craniospinal radiation therapy followed by adjuvant chemotherapy for newly diagnosed average-risk medulloblastoma. J Clin Oncol. 2006;24:4202–4208. doi: 10.1200/JCO.2006.06.4980. [DOI] [PubMed] [Google Scholar]
  • 4.Gururangan S, Gardner S, Finlay JL. Autologous hemapoeitic stem cell transplantation after high dose chemotherapy for primary malignant tumors of the central nervous system. In: Atkinson K, Champlin R, Ritz J, Fibbe WE, Ljungman P, Brenner MK, editors. Clinical Bone Marrow and Blood Stem Cell Transplantation. 3rd ed. New York: Cambridge University Press; 2003. pp. 716–731. [Google Scholar]
  • 5.Dunkel IJ, Boyett JM, Yates A, et al. High-dose carboplatin, thiotepa, and etoposide with autologous stem-cell rescue for patients with recurrent medulloblastoma. Children’s Cancer Group. J Clin Oncol. 1998;16:222–228. doi: 10.1200/JCO.1998.16.1.222. [DOI] [PubMed] [Google Scholar]
  • 6.Dupuis-Girod S, Hartmann O, Benhamou E, et al. Will high dose chemotherapy followed by autologous bone marrow transplantation supplant cranio-spinal irradiation in young children treated for medulloblastoma? J Neurooncol. 1996;27:87–98. doi: 10.1007/BF00146088. [DOI] [PubMed] [Google Scholar]
  • 7.Fouladi M, Shih C-S, Gilger E, et al. Role of high dose chemotherapy and autologous stem cell rescue in children with recurrent CNS malignancies [abstract] Hematologica Rep. 2006;2:5. [Google Scholar]
  • 8.Graham ML, Herndon JE, 2nd, Casey JR, et al. High-dose chemotherapy with autologous stem-cell rescue in patients with recurrent and high-risk pediatric brain tumors. J Clin Oncol. 1997;15:1814–1823. doi: 10.1200/JCO.1997.15.5.1814. [DOI] [PubMed] [Google Scholar]
  • 9.Guruangan S, Dunkel IJ, Goldman S, et al. Myeloablative chemotherapy with autologous bone marrow rescue in young children with recurrent malignant brain tumors. J Clin Oncol. 1998;16:2486–2493. doi: 10.1200/JCO.1998.16.7.2486. [DOI] [PubMed] [Google Scholar]
  • 10.Massimino M, Gandola L, Spreafico F, et al. No salvage for relapsing already irradiated medulloblastoma if rescued with high-dose chemotherapy ± re-irradiation [abstract] Hematologica Rep. 2006;2:11. [Google Scholar]
  • 11.McLendon RE, Enterline DS. Tumors of specialized tissues of central neuroepithelial origin. In: Bigner DD, McLendon RE, Bruner JM, editors. Russell and Rubinstein’s Pathology of Tumors of the Nervous System. New York: Oxford University Press; 1998. pp. 25–30. [Google Scholar]
  • 12.Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958;53:457–480. [Google Scholar]
  • 13.Packer RJ. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma. Curr Neurol Neurosci Rep. 2007;7:130–132. [PubMed] [Google Scholar]
  • 14.Frei E, 3rd, Teicher BA, Holden SA, Cathcart KN, Wang YY. Preclinical studies and clinical correlation of the effect of alkylating dose. Cancer Res. 1988;48:6417–6423. [PubMed] [Google Scholar]
  • 15.Bode U, Simon A, Hasan C, Zimmerman M, Fleischhack G. The role of HD chemotherapy (CT) in the treatment of therapy-resistant CNS PNETs, HIT-REZ-97 results [abstract] Hematologica Rep. 2006;2:2. [Google Scholar]
  • 16.Shih CS, Hale GA, Gronewold L, et al. High-dose chemotherapy with autologous stem cell rescue for children with recurrent malignant brain tumors. Cancer. 2008;112:1345–1353. doi: 10.1002/cncr.23305. [DOI] [PubMed] [Google Scholar]
  • 17.Duffner PK, Horowitz ME, Krischer JP, et al. The treatment of malignant brain tumors in infants and very young children: an update of the Pediatric Oncology Group experience. Neuro-Oncology. 1999;1:152–161. doi: 10.1093/neuonc/1.2.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bowers DC, Gargan L, Weprin BE, et al. Impact of site of tumor recurrence upon survival for children with recurrent or progressive medulloblastoma. J Neurosurg. 2007;107:5–10. doi: 10.3171/PED-07/07/005. [DOI] [PubMed] [Google Scholar]
  • 19.Walter AW, Mulhern RK, Gajjar A, et al. Survival and neurodevelopmental outcome of young children with medulloblastoma at St Jude Children’s Research Hospital. J Clin Oncol. 1999;17:3720–3728. doi: 10.1200/JCO.1999.17.12.3720. [DOI] [PubMed] [Google Scholar]
  • 20.Mahoney DH, Jr, Strother D, Camitta B, et al. High-dose melphalan and cyclophosphamide with autologous bone marrow rescue for recurrent/progressive malignant brain tumors in children: a pilot Pediatric O ncology Group study. J Clin Oncol. 1996;14:382–388. doi: 10.1200/JCO.1996.14.2.382. [DOI] [PubMed] [Google Scholar]
  • 21.Kamen BA, Rubin E, Aisner J, Glatstein E. High-time chemotherapy or high time for low dose. J Clin Oncol. 2000;18:2935–2937. doi: 10.1200/JCO.2000.18.16.2935. [DOI] [PubMed] [Google Scholar]
  • 22.Belza MG, Donaldson SS, Steinberg GK, Cox RS, Cogen PH. Medulloblastoma: freedom from relapse longer than 8 years—a therapeutic cure? J Neurosurg. 1991;75:575–582. doi: 10.3171/jns.1991.75.4.0575. [DOI] [PubMed] [Google Scholar]
  • 23.Torres CF, Rebsamen S, Silber JH, et al. Surveillance scanning of children with medulloblastoma. N Engl J Med. 1994;330:892–895. doi: 10.1056/NEJM199403313301303. [DOI] [PubMed] [Google Scholar]
  • 24.Ashley DM, Meier L, Kerby T, et al. Response of recurrent medulloblastoma to low-dose oral etoposide. J Clin Oncol. 1996;14:1922–1927. doi: 10.1200/JCO.1996.14.6.1922. [DOI] [PubMed] [Google Scholar]
  • 25.Baruchel S, Diezi M, Hargrave D, et al. Safety and pharmacokinetics of temozolomide using a dose-escalation, metronomic schedule in recurrent paediatric brain tumours. Eur J Cancer. 2006;42:2335–2342. doi: 10.1016/j.ejca.2006.03.023. [DOI] [PubMed] [Google Scholar]
  • 26.Gilbertson RJ. Medulloblastoma: signalling a change in treatment. Lancet Oncol. 2004;5:209–218. doi: 10.1016/S1470-2045(04)01424-X. [DOI] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES