radiation therapy in the management of childhood brain tumors

13
Received: 21 March 2000 Abstract Radiation therapy (RT) still plays a major role in the man- agement of intracranial malignan- cies, together with surgical resection and, more recently, chemotherapy. This is a review of the experience with fractionated external beam RT. In medulloblastomas, combined mo- dalities currently achieve a 5-year survival in excess of 70% in low-risk subgroups and 40% in the subgroups considered to be high risk. For the past decade, the emphasis has been on the quality of life in cured chil- dren. Recent advances have mainly aimed at limiting the toxicity of the “prophylactic” craniospinal irradia- tion by testing dose reductions and altered fractionations. Technical in- novations have also greatly benefited gliomas: modern techniques dealing with 3D CT and MRI-based treat- ment combined with stereotactic po- sitioning of the patients, achieve a high degree of conformity that might improve both local control and long- term neurocognitive and growth se- quelae. Keywords Brain tumors · Children · Radiotherapy Child’s Nerv Syst (2001) 17:121–133 © Springer-Verlag 2001 REVIEW PAPER Jean-Louis Habrand Renaud De Crevoisier Radiation therapy in the management of childhood brain tumors Introduction Brain tumors represent the most common solid tumors in children and adolescents, i.e., approximately 20%. Com- pared with those in their adult counterparts, these tumors are more frequently located in the posterior fossa (55% vs 10%) and those located in the hemispheres are less frequently situated in the superficial cortex [14]. More- over, the pathological types present major differences: the astrocytomas and the medulloblastomas are predomi- nant in patients below 15 years of age [45], compared with brain metastases and glioblastomas multiforme in adults.[12]. Meningiomas, acoustic neuromas, and pitui- tary adenomas are also extremely rare in this age group. These differences indicate that despite the common major places of surgical resection and radiation therapy in both groups, clear differences exist in terms of thera- peutic management and outcome. To mention a few.there is the necessity of irradiating the whole central nervous system (CNS) in some major tumor types: medullo- blastomas and, according to stage and treatment policy, ependymomas, germ cell tumors, and primitive neuro- ectodermal tumors (PNETs) in children. Also, there are potentially severe, long-term sequelae related to neuro- cognitive and pituitary dysfunctions that predominate in children up to 5 years of age. Then, too, chemotherapy is of growing importance, particularly in the treatment of infants and children up to 3 years of age [32, 46,96]. The place of radiation therapy in past and current pro- tocols is summarized for the main CNS tumors seen in children and adolescents, and its major side effects are reviewed. Medulloblastoma Outcome of conventional approaches The medulloblastoma, a PNET that affects the posterior fossa, is seen in 20–26% of brain tumors in children. Al- though seen at various ages, it predominates in children of around 5 years. One of its salient features is its high J.-L. Habrand ( ) · R. De Crevoisier Pediatric Unit, Department of Radiation Oncology, Institut Gustave Roussy, 39 rue C. Desmoulins, 94805 Villejuif Cedex, France e-mail: [email protected] Fax: +33-1-42115253

Upload: renaud

Post on 23-Aug-2016

215 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Radiation therapy in the management of childhood brain tumors

Received: 21 March 2000 Abstract Radiation therapy (RT)still plays a major role in the man-agement of intracranial malignan-cies, together with surgical resectionand, more recently, chemotherapy.This is a review of the experiencewith fractionated external beam RT.In medulloblastomas, combined mo-dalities currently achieve a 5-yearsurvival in excess of 70% in low-risksubgroups and 40% in the subgroupsconsidered to be high risk. For thepast decade, the emphasis has beenon the quality of life in cured chil-dren. Recent advances have mainlyaimed at limiting the toxicity of the

“prophylactic” craniospinal irradia-tion by testing dose reductions andaltered fractionations. Technical in-novations have also greatly benefitedgliomas: modern techniques dealingwith 3D CT and MRI-based treat-ment combined with stereotactic po-sitioning of the patients, achieve ahigh degree of conformity that mightimprove both local control and long-term neurocognitive and growth se-quelae.

Keywords Brain tumors · Children ·Radiotherapy

Child’s Nerv Syst (2001) 17:121–133© Springer-Verlag 2001 R E V I E W PA P E R

Jean-Louis HabrandRenaud De Crevoisier

Radiation therapy in the management of childhood brain tumors

Introduction

Brain tumors represent the most common solid tumors inchildren and adolescents, i.e., approximately 20%. Com-pared with those in their adult counterparts, these tumorsare more frequently located in the posterior fossa (55%vs 10%) and those located in the hemispheres are lessfrequently situated in the superficial cortex [14]. More-over, the pathological types present major differences:the astrocytomas and the medulloblastomas are predomi-nant in patients below 15 years of age [45], comparedwith brain metastases and glioblastomas multiforme inadults.[12]. Meningiomas, acoustic neuromas, and pitui-tary adenomas are also extremely rare in this age group.

These differences indicate that despite the commonmajor places of surgical resection and radiation therapyin both groups, clear differences exist in terms of thera-peutic management and outcome. To mention a few.thereis the necessity of irradiating the whole central nervoussystem (CNS) in some major tumor types: medullo-blastomas and, according to stage and treatment policy,

ependymomas, germ cell tumors, and primitive neuro-ectodermal tumors (PNETs) in children. Also, there arepotentially severe, long-term sequelae related to neuro-cognitive and pituitary dysfunctions that predominate inchildren up to 5 years of age. Then, too, chemotherapy isof growing importance, particularly in the treatment ofinfants and children up to 3 years of age [32, 46,96].

The place of radiation therapy in past and current pro-tocols is summarized for the main CNS tumors seen inchildren and adolescents, and its major side effects arereviewed.

Medulloblastoma

Outcome of conventional approaches

The medulloblastoma, a PNET that affects the posteriorfossa, is seen in 20–26% of brain tumors in children. Al-though seen at various ages, it predominates in childrenof around 5 years. One of its salient features is its high

J.-L. Habrand (✉ ) · R. De CrevoisierPediatric Unit, Department of Radiation Oncology, Institut Gustave Roussy, 39 rue C. Desmoulins, 94805 Villejuif Cedex, Francee-mail: [email protected]: +33-1-42115253

Page 2: Radiation therapy in the management of childhood brain tumors

propensity for metastasizing within the CNS. At the timeof presentation, 5–30% have been reported and 5–10%during the course of the disease [67, 91, 142].

Surgical resection alone, as performed by Cushing inthe 1930s, has no curative intent (1/63 children alive at 3 years) [13]. Medulloblastomas are clearly radiosensi-tive, with 18–44% of cells surviving a dose of 2 Gy inradiobiological experiments [99]. Nonetheless, the ad-junction of local radiation did not substantially improvethe outcome, as reported by the Toronto Group in themid-1950s [76]. Only the introduction of a systematicextended coverage of the CNS dramatically improvedthe survival in an early series to 8 out of 15. Consistentdata of multiple groups indicated a 27–40% 5-year-survival (S) with non-documented benefits beyond thisfollow-up [10, 67]. In the mid 1970s, most authors rec-ommended a standard 35–40 Gy as a “prophylactic”dose to the CNS, plus a boost up to 50–55 Gy in the pos-terior fossa [71, 140]. Together with radiation dose andvolume (22%), the quality of surgical resection (44%)was found to be a prognostic indicator with a 10-yearlife expectancy in Bloom’s series [15]. Some authorshave found a negative impact of a ventriculoperitonealshunt owing to increased systemic failures [80]. Thenegative impact of incomplete resection has been recent-ly confirmed in a Children’s Cancer Study Group study(CCSG) [156].

Technical considerations

Several series have emphasized that the posterior fossa isthe most critical site of failure with 75% in the Castro-Vita series [21] and 53% in the Jereb series [78]. Thisauthor also emphasized the importance of adequate cov-erage of the anterior cerebral fossa, especially at the level of the cribriform plate, an area frequently shieldedinadvertently by the ocular blocks. The national panelsof experts that nowadays act as quality-control groupsare paying particular attention to the doses delivered inthese anatomical structures [20, 105]. As the delivery ofa uniform dose to the entire CNS is particularly chal-lenging and implies multiple carefully matched beams(sometimes of different qualities, like photons and elec-trons), detailed dosimetric information needs to be col-lected for every patient. Our group recently analyzed allchildren cases that had had CNS failure following exten-sive radiation as a function of dose uniformity [9]. Noclear dose-affected relationship was found at the site ofmost failures, except at the cribriform plate. These datasuggest that failures due to technical limitations can beminimal in large centers, which can depend on a greaterdegree of collective experience.

Recently, several authors have pointed out the possi-bility of delivering the boost to the posteria fossa using a stereotactic technique. The advantage would be to de-

liver a high dose in a single fraction with elegant sparingof anatomical structures located a few millimeters awayfrom the target volume [155]. Such a technique couldalso be applied to small recurrences located in the post-eria fossa (PF) and treated with a high dose with similaradvantages. The increasing number of early detections ofrecurrences using serial surveillance imaging could in-crease the number of patients able to benefit from suchtechniques and therefore be potentially curable [137].Conformal irradiation delivered with conventional frac-tionation is also gaining wide acceptance. For example,sparing of normal structures like the cochlea could beimproved [54]. Nonetheless, techniques that can sparepart of the PF will need further evaluation since it hasbeen established that failures within the PF arise in ap-proximately half of the cases outside the surgical bed[53].

Modern strategies [111]

Since the mid-1980s, ,the aims of most pilot studies andsubsequent multi-institutional studies have been:

1. To improve the possibility of survival

2. To improve the quality of life of children, especiallythe younger ones and those selected with favorable prog-nostic factors (low-risk groups: i.e., generally with totalor subtotal resection, and no leptomeningeal dissemina-tion) as opposed to unfavorable ones (high-risk groups:i.e., macroscopically incomplete resection and/or lepto-meningeal/systemic dissemination)

It is important to note that radiotherapeutical innova-tions, whether associated with chemotherapeutical pro-grams or not, have represented the cornerstone of thesenew approaches.

Mono-institutional experiences

Interesting studies have attempted to reduce the prophy-lactic dose to the brain and the spine. Brand [17] com-pared two historical series treated successively with“high” (i.e., 30–40 Gy to the spinal axis and 40–50 Gy tothe brain) and then “low” doses of radiation (25 Gy tothe entire CNS). “High-risk” patients experienced anoverall 39% survival versus 80% for “low-risk” patients(see definitions above). No statistical difference was in-troduced by the dose reduction in any of the subgroups.Levin et al. [93] reported an early series of reduced dose,i.e., 35 Gy to the brain and 25 Gy to the spine, combinedwith chemotherapy (procarbazine + hydroxyurea). Anexcellent 65% 5-year-survival was observed for “low-risk” patients, but a disappointing 25% for those consid-ered to be “high risk”.More recently, Packer reported anexcellent 86% 5-year progression-free survival (PFS) by

122

Page 3: Radiation therapy in the management of childhood brain tumors

a combination of low prophylactic dose (i.e., 23.4 Gy) ofradiation and chemotherapy [115]. A further decrease inthe prophylactic dose has been tested by the PhiladelphiaGroup in conjunction with multiagent chemotherapy[59]. Ten children below 5 years of age received 18 Gyprophylactic doses in the CNS + boost in the PF up to64.8–65.4 Gy. Six-year survival is 70±20%, but thisstudy should be examined with caution owing to thesmall number of accrued patients.

Altered fractionations have been advocated for thepast 15 years by many authors. Pure hyperfraction inwhich the dose per fraction is decreased (with little or notreatment acceleration) is generally favored. Radiobio-logical models (based on cultured cell lines and the irra-diation of weanling rats), along with preliminary clinicalexperiences, indicate that the long-term side effects ofradiation can be minimized if the dose per fraction islowered, given that the interfraction interval exceeds 6–8 h, to allow for the repair of cellular sublethal injuries[41, 65]. According to these models, tumor controlshould not be compromised by these approaches if theoverall treatment time is kept constant. Pure hyperfrac-tionation can be exploited:

1. To improve long term sequelae if total dose is notmodified compared with conventional fractionation

2. To improve tumor control (but with little or no im-provement in long-term side effects) if the total dose isincreased

The second approach (the increased total dose) has beenadvocated by Allen [4]. Among 23 children with PNETs,15 with a T3b and T4 non metastatic medulloblastomareceived 72 Gy to the posterior fossa and 36 Gy to therest of the CNS, using two daily fractions of 1 Gy each.Based on a popular mathematical representation of cellsurvival curves (i.e., the linear quadratic formalism thatassumes an alpha/beta value of 10 for tumor control andacute reactions, and of 1 for brain and spinal cord long-term tolerance), this altered fractionation might have in-duced a differential biological effectiveness between thetumor and the low-proliferating normal tissues that couldbe estimated respectively to 66 Gy and 24 Gy adminis-tered with a “conventional” fractionation (i.e., 2 Gy oncea day). With a median 78-month follow-up, 14/15 chil-dren are alive and well. As far as the first approach (nomodification in the total dose) is concerned, it has beenadvocated by Prados [121], who administered 24 Gy andlater on 30 Gy as CNS prophylaxis in 2 daily fractions of1 Gy each + boost to the PF. With a median 1.9-year fol-low-up, the rates of spinal failures were of 50% and12.5% according to the dose level, indicating that 24 Gyhyperfractionated is likely to be insufficient, even thoughin the “low risk” group. It should be pointed out that al-tered fractionation does not improve the acute toxicity,which has been reported as more severe, especially when

it is started after chemotherapy. On the other hand, spe-cial attention should be paid to the prolonged treatmentduration induced by the very low doses per fraction test-ed in “maximally” hyperfractionated regimens. Thiscould theoretically allow tumor repopulation beforecompletion of radiation and be related to early failures.One series has recently correlated treatment protractionbeyond 45 days with further failures [31]. Restriction ofirradiation has also been tested in infants and childrenbelow 3 years of age [96]. Promising studies indicatethat in this population a prolonged chemotherapy regi-men can delay the initiation of radiotherapy by 12–18months without compromising the final outcome. Fur-thermore, promising developments, including those ex-perienced by our own group, suggest that in this agegroup, high-dose chemotherapy followed by stem-cellrescue could substitute for the conventional “prophylac-tic” coverage of the whole CNS, and be replaced by fo-cal irradiation only [40].

Multi-institutional studies

Since 1975, the International Society of Pediatric Oncolo-gy (SIOP) has conducted two successive studies: SIOP I(1975–1979) tested in a randomized fashion the value ofchemotherapy associated with a surgical resection and ra-diation therapy [109, 146]: the children were elected toeither receive or not receive 8 cycles of vincristin, CCNU(1, (2-chloroethyl)-3-cyclohexyl-1-nitrosurea (= lomus-tine)) following a CNS irradiation of 30–35 Gy plus a 20 Gy boost to the PF. In 286 patients treated in 44 cen-ters in 15 countries, the 5-year disease-free survival wasstatistically proved to be no different whether chemother-apy was administered or not (56 vs 42%). Nonetheless,subgroups at high risk of local regional failures seemed tobenefit from the addition of chemotherapy (incompleteresection P≤0.007, brain-stem involvement P≤0.001).

Similarly, the Pediatric Oncology Group (POG) andthe CCSG have tested the value of a combination ofCCNU and prednisone in addition to conventional irradi-ation [44]. In 275 patients (179 medulloblastomas), astatistical difference only existed for subgroups with ad-vanced disease in the posterior fossa or the rest of theCNS (46 vs 0% DFS).

SIOP 2, opened between 1984 and 1989, addressedtwo issues in a double randomization [6]:

1. The value of “sandwich” chemotherapy placed be-tween surgery and radiotherapy and made up of an inten-sified chemotherapy regimen (procarbazine, CCNU,methotrexate, vincristine, prednisone) for 1 month

2. The feasibility of decreasing the prophylactic dose ofradiation from 35 to 25 Gy in the subgroup at “low risk.”The preliminary analysis showed that chemotherapy didnot clearly influence the outcome of any subset of pa-

123

Page 4: Radiation therapy in the management of childhood brain tumors

tients. Low-dose radiotherapy was accompanied by aslightly more unfavorable outcome compared with con-ventional doses of radiation, even when chemotherapywas associated with it.

Later, national and international groups designed differ-ent approaches to the question of whether the childrenwere classified in “low-” or “high”-risk groups.

In “low-risk” groups, POG-CCSG 863 compared a re-duced 23.4 Gy prophylactic dose in the CNS + PF boostwith the standard 36 Gy + PF boost without any addi-tional chemotherapy. An interim analysis showed a dra-matic adverse effect of low-dose radiation (p0.02), andso the study was interrupted [33]. A recent update seemsto indicate that in the long run, there seems to be no dif-ference between the outcomes for the two groups [148].The French Society of Pediatric Oncology (SFOP) hasjust completed a new randomized national study inwhich the “low-risk” group received systematically a 3-month postoperative chemotherapy, alternating the“eight-in-one” drugs with VP 16 and carboplatin, fol-lowed by a reduced prophylactic dose of radiation(CNS=25 Gy, PF=55 Gy). A recent update of the studyshows a promising 70% 3-year survival rate [37]. Theseresults are consistent with those of a close POG studydelaying the administration of chemotherapy at the timeof and after radiotherapy, which has achieved excellent86% and 79%, 3 and 5 year-progression-free survival(PFS) [115].

In “high-risk” groups there is general agreement thatthese patients deserve “conventional” doses of radiation(not less than 30–35 Gy prophylactic and 52–55 Gy inthe PF), combined with a polychemotherapy regimen[16, 156]. The timing still remains controversial, withsome groups advocating postoperative chemotherapy,followed by delayed radiation, and others advocating thereverse [85]. It is possible that protocols with radiothera-py longer than 3 months following surgery might in-crease the risk of early tumor progression. It has beenshown recently that in children below the age of 3 years,a residual primary of more than 1.5 cm2 and gross meta-static foci are strong adverse prognostic factors [156].

Gliomas

Ependymomas

Ependymomas account for approximately 10% of tumorsof the CNS in children [22, 77, 136]. Median age is 5 years, with cases frequently diagnosed between 1 and 2 years. Primary tumors in the spinal cord are also seen,but rarely before 10 years [154]. As far as intracranial-tumors are concerned, 60–75% are located in the PF[66]. According to histological criteria, four grades ofdifferentiation have been described.

Following surgical resection alone, 5-year-survival is in the order of 16% [132]. Adding radiotherapy to the whole CNS has shifted this figure up to 30–61% 5-year-PFS [55, 58, 90, 118, 136] and to 36–67% 5-year-S[13, 55, 58, 102, 118, 136, 150]. Chemotherapy hasshown limited efficacy [26], but has induced prolongedremission in selected recurrent cases [57]. The quality ofresection [127], pathological grade [55, 81], dose of radi-ation to the primary [58, 118], tumor site [98], and theage of the patient at the time of treatment [58] are prog-nostic factors that are not unanimously acknowledged.For example, in the CCG experience [127], in whichchildren received postoperative craniospinal radiationand, in a random fashion, chemotherapy, predictors ofPFS duration included only the completeness of surgicalresection confirmed radiologically (P≤10–4). A prelimi-nary experiment conducted by the POG has suggestedthat hyperfractionation to high dose could be followedby two-fold chances of cure compared with conventionalirradiation in subtotally resected children [84].

One of the most debatable issues for the radiation on-cologist is the place of prophylactic irradiation of thewhole, or part, of the CNS. All series have shown thatspinal metastases are seen in 8–13% of ependymomaslocated in the PF. These findings have motivated thecoverage of the entire CNS, or at least of the spinal axisin these tumor sites. Early detection of spinal seeding,using MRI in the initial work-up, is now routinely per-formed. Focal irradiation in patients with no evidence ofmetastases is again tested by several groups [56, 83, 129,134].

Astrocytomas

Astrocytomas represent 35% of all brain tumors [141].Most malignant varieties arise in the supratentorial area,unlike the benign ones, which are commonly seen in thecerebellar area. The outcome of malignant forms has abetter reputation than their adult counterparts, especiallyin children below 10–12 year of age [117]. This is proba-bly due to the larger number of anaplastic astrocytomasand smaller proportions of glioblastomas multiforme(GB). Overall, the rate of 5-year survival is around40–50% [38]. The outcome of GB remains as poor as inthe adult series and less than 20% [35]. Most authorsagree that low-grade astrocytomas that are completelyresected microscopically do not require further therapy.Postoperative irradiation of other cases remains standardpractice, especially in the inoperable deep-seated lesionsthat are more common in children than in adults. Fromthe adult experience, it is interesting to note that doses ofradiation in excess of 45 Gy do not seem to improve lo-cal control in incompletely resected forms [79].All these tumors are irradiated focally, i.e., the tumor bed+ a safety margin of 2–3 cm around it, although some

124

Page 5: Radiation therapy in the management of childhood brain tumors

authors have advocated the coverage of the entire CNSin oligodendroglioma of the PF, an entity extremely rarein children [112]. Modern techniques based on 3D CTand MRI (Fig. 1) virtual simulation allow multiple ar-rangements of coplanar and non-coplanar tailor-madebeams (Fig. 2) that can dramatically limit the dose re-ceived by the normal brain. The evaluation of rival treat-ment plans can nowadays be optimized using integraldose-volume histogram (DVHs) intercomparisons bothfor the target (Fig. 3) and for the surrounding anatomi-cal structures. In the future, more refined biological indi-ces evaluating normal tissue complication probabilities(NTCP) and tumor control probability (TCP) might behelpful.

Gliomas of the optic pathway

These represent 5% of all brain tumors and are essential-ly benign histologically (pilocytic astrocytomas). Theoverall very slow pace of the disease allows an initial

watch-and-see policy in the majority of cases. In the caseof tumor progression, various therapeutical approachescan be discussed: surgical resection of a tumor limited toone optic nerve with deteriorated vision [36], low-dosecontinuous chemotherapy, especially in younger chil-dren, [75] and radiotherapy, especially in cases of rapidvisual deterioration. Doses in the range of 40–50 Gy arerecommended [47, 152]. Extensive coverage of both op-tic nerves and chiasm was used before the introductionof modern imaging. Unfortunately, owing to the frequentassociation with type 1 neurofibromatosis, these patientswere also at higher risk of late radiation injuries [60].

Nowadays, several authors point out that target volume can be more economically focused on radiologi-cal abnormalities [30,97]. Following radiotherapy, a65–90% local control has been reported [5, 28, 30, 36,47, 106, 152]. Visual improvement can be expected inmore than half the cases [27, 70]. Re-irradiation of recur-rent disease with moderate doses has been reported to in-duce prolonged remission in rare cases [152].

Brain-stem gliomas

These comprise 15% of brain tumors. Benign forms,generally assessed on their radiological aspect, site, andclinical behavior represent a minority of them. A surgi-cal resection of the purely exophytic forms can be at-tempted and is facilitated by the use of the ultrasonicscalpel and neuronavigation [43]. In this group, Stroinkhas reported 10/11 children alive [143]. Unfortunately,the vast majority of brain-stem tumors present with atypical malignant evolution with rapidly progressivemultiple cranial-nerve palsies and, on imaging, a diffusehypodense aspect [1, 2, 101]. These forms are generallylethal within a year [42, 49, 64]: this has motivated manyinnovative pilot and multi-institutional studies that havetested chemotherapy, high-dose megatherapy, combinedchemoradiation, and radiotherapy alone with unconven-tional fractionations. As field failures represent 88% of

125

Fig. 1 Digitally reconstructedradiograph produced in 3D vir-tual simulation of a base of theskull malignancy

Fig. 2 Beam’s eye view of anon-coplanar beam. The grosstumor volume is represented ingray and surrounded by the as-sumed microscopic extensions.The dotted line corresponds tothe beam’s aperture (the samepatient as in Fig. 1)

Fig. 3 Intercomparison between four different treatment plans fora brain stem tumor. The optimal one corresponds to 55 Gy distrib-uted uniformly in ≥95% of the target volume

Page 6: Radiation therapy in the management of childhood brain tumors

the failures, inclusion of the whole brain is no longerrecommended. Pure hyperfractionation that allows theadministration of a higher tumor dose without excessivetoxicity has been largely tested with escalated doses bythe groups in North America. The dose per fractionranged generally between 1 and 1.26 Gy, administeredtwice a day. Tumor control has remained desperatelylow: a median of 9 months’ survival following 66 Gy[86]; 23% 2-year-survival after 70.2 Gy [50]; 14% 3-year survival after 72 Gy [113], 7% 2-year survivalfollowing 75.6 Gy [51]; and an 11% 3-year-survival after78 Gy [114]. It is interesting to note that radiation necro-sis has been documented at 75 Gy and above, includinglethal cases [52,113]; it is not surprising that an attemptto test the “intermediate” 70.2 Gy. dose level combinedwith cis platinum in a randomized fashion versus a con-ventional 54 Gy arm also failed to influence the outcomeof the disease [100]. There is definitely a need for differ-ent therapeutical approaches to be tested: for example,the new radiosensitizing agents such as topo isomerase Iinhibitors [88] or radiotherapeutic innovations like the Boron Neutron Capture Therapy [108].

Pineal tumors

Pineal tumors represent only 0.3–3% of pediatric braintumors. In addition to their rarity, their management iscomplicated by the diversity of the histological subtypesthat can be observed. Three major types can be individ-ualized: the germ-cell tumors (dysgerminomas, terato-mas, yolk sac tumors, choriocarcinomas, and mixedforms); the very rare parenchymal tumors (pineoblasto-mas and pineocytomas); and the gliomas. Radiosensitiv-ity is correlated to some extent with the pathology. Puredysgerminomas have a reputation of high sensitivity(but probably somewhat less that their gynecologicalcounterpart) unlike the other germ-cell subtypes that donot seem to differ dramatically from classical gliomas.Another salient feature of several of these tumors istheir propensity for giving multiple foci locoregionally(third ventricle, supra- and intrasellar areas) and some-times a leptomeningeal seeding. This risk has been esti-mated at between 8 and 23%, with dysgerminoma (sem-inoma) and pineoblastoma being in the upper range[95]. This has conditioned a complex therapeutic ap-proach based on “prophylactic” irradiation of the entireCNS [72] and/or chemotherapy [3, 7, 8, 29, 133]. Aswith medulloblastomas, surgical resection alone cannotbe considered as a curative measure of malignant le-sions. Furthermore, despite major technical advances, acomplete resection is rarely made possible. Until recent-ly, radiation therapy has remained the cornerstone ofmanagement. Until the 1980s, it was common practiceto test the radiosensitivity of an unbiopsied lesion by afirst course of radiation. Pure dysgerminoma was as-

sumed in the case of a good tumor response and the ra-diation program completed with whole CNS coverage ata low dose. If the response was poor, non-seminomatoussubtypes were assumed and high-dose focal radiationwas recommended [125]. Most children are currentlymanaged with an unequivocal diagnosis based on tumormarkers or histological specimens. Using radiation ther-apy alone, the outcome of pure dysgerminomas at 5years ranges between 70 and 100%. Several authors stilladvocate the use of whole CNS prophylaxis that reducesleptomeningeal metastases to less than 10% [72]. An-other approach is to irradiate, as a prophylactic measure,only those children with positive CSF or leptomeningealdissemination on the initial MRI [138]. Other groupsrecommend irradiation of the whole brain alone or theventricular system [139]. Combined chemoradiation isalso gaining wide acceptance and, in the majority ofchildren that are good responders to an initial poly-chemotherapy regimen, focal irradiation alone. Doses tothe primary extend from 30 to 40 Gy in pure dysge-minomas and up to 50–55 Gy in other germ-cell tumorsthat are more radioresistant. Where indicated, prophy-lactic CNS irradiation of dysgerminoma ranges between20 and 30 Gy.

Craniopharyngiomas

Craniopharyngiomas represent less than 10% of intracra-nial tumors in this age group, but half of the sellar tu-mors. Although they are histologically benign„locallytheir behavior is very aggressive. Exceptionally malig-nant variants have also been described. Their manage-ment is still controversial. Radical resection has beenrecommended for several decades as the mainstay oftreatment, but is followed by a 20–30% relapse rate anda high morbidity affecting mainly the visual and the en-docrinological status [69, 145]. On the other hand, par-tial resection is less toxic but at the cost of a 70% risk oflocal failure. These findings have stimulated the interestin a combination of less aggressive surgery and postop-erative irradiation, particularly in children who fail a pre-vious surgical resection. With this approach, local con-trol looks similar to that of radical resection with lesstoxicity [48, 62, 150, 153]. Nonetheless, recent radio-therapeutic series indicate that the optimal dose might beabove 55 Gy, fractionated [62, 123, 144]. At such doselevels, high-precision techniques (like fractionated ste-reotactic photons or protons) are preferable in order tospare close anatomical structures like the optic pathwayand the third ventricle [147]. Owing to the close proxim-ity of the optic pathway to the tumor volume, we wouldnot advocate the use of single fractions but of conven-tionally fractionated regimens.

126

Page 7: Radiation therapy in the management of childhood brain tumors

PNETs

With the exception of medulloblastomas, these are raretumors that can occur anywhere in the CNS. Followingthe initial description of a small round-cell neoplasm,various histological classifications have been proposed,generally based on the presence or absence of cellu-lar differentiation (pineoblastoma, medulloepithelioma,ependymoblastoma) [128]. These tumors seem to have incommon a particular aggressiveness that requires a com-bined approach based on surgery, radiation therapy, andchemotherapy. As most of them arise in the supratentori-um in very young children, some groups, including ourown, emphasize a prolonged chemotherapy regimen,completed by high-dose megatherapy. Nonetheless, radi-ation therapy remains mandatory in older children and incases of progressive disease. Focal irradiation in supra-tentorial sites and whole CNS coverage in infratentorialsites is generally recommended. Long-term results re-main poor, with ≤45% 3-year-event-free survival (EFS)[104]. PNETs of the pineal area (pineoblastomas) seemto fare better (61% 3-year-PFS) by combined chemother-apy and craniospinal radiation [74].

Radiation-induced toxicity and sequelae

It is estimated that the number of neurons cannot be ex-pended after the 6th month of fetal life. During the first 3 years of life and to some extent up to 6 years, cerebraldevelopment includes cellular and axonal hypertrophycombined with a multiplication of dendrits and interneu-ronal connections [34]. The myelinization process ismaximal until 2 years of age, but remains detectable upto puberty, and possibly even later. Demyelinizationrepresents the most striking microscopic alteration fol-lowing radiation and is responsible for white-matter ne-crosis. Pathogenesis is still unclear. It could be relatedto alterations of the microvasculature (affecting the en-dothelial cells), or of the oligodendrocytes that producemyelin. Two major clinical syndromes have been de-scribed following radiation, both in children and inadults:

1. Cerebral radionecrosis whose risk has been estimatedto 5% at 55 Gy fractionated, 15% at 60 Gy, and 20% at65 Gy [103].

2. Necrotizing leukoencephalopathy and mineralizingmicro-angiopathy [82, 119]; this should be suspected ifseizures, somnolence, motor deficits, and ataxia are pres-ent. Pathologically, multifocal white-matter necrosis canbe detected with loss of myelin sheath and oligodendro-cytes. In mineralizing microangiopathy, neocalcificationsare also detectable, especially in the basal ganglia. Ra-diologically, typical white-matter alterations are com-

monly reported in the irradiated volume, together withcerebral atrophy, ventricular dilatation, and sometimes,calcifications [24]. Clinical manifestations are not corre-lated with the intensity of radiological changes.

Neurological complications have been extensively docu-mented following CNS prophylaxis therapy in acutelymphoblastic leukemia (ALL). In this situation, the al-terations actually reflect the toxicity of a combination oflow-dose radiation (i.e., ≤24 Gy) delivered to the wholebrain with concomitant intrathecal and/or systemic che-motherapy (mainly methotrexate). Up to 45% of childrenwill develop complications within 1 year [25]. Solidbrain tumors treated at 45 Gy and more also developsimilar complications, including the so-called “somno-lence” phenomena and confusing neurological deficits[11]. The number of children affected by clinicallyasymptomatic complications has increased with the wideuse of MRI in the follow-up, as mentioned by Laitt et al.in ALL [87]: 9/35 irradiated children versus only 1/24 ina control group not irradiated. This included the earlydetection of radiation-associated second malignancies.Russo found late radiographic changes in over half thecases of medulloblastomas treated with hyperfraction-ation, but none with overt clinical symptoms [130].

Cognitive dysfunctions are also well documented fol-lowing radiation in youngsters. Most documented caseshave been reported following prophylactic brain irradia-tion of ALL and medulloblastomas [25, 27, 122, 126].The intensity of symptoms seems mainly correlated withthe age of the child at the time of irradiation [23], withthe total dose administered, with the volume of brain en-compassed, and possibly with the fractionation of thedose. Devastating deterioration has been reported in chil-dren up to 3 years of age following 35–40 Gy in thewhole brain, and severe deterioration can still occur upto 6 years of age [74, 89]. They rarely become manifestbefore the 3rd year follow-up and then regularly deterio-rate until adulthood. The classical report by Hirsch et al.in the early1980s compared a series of children irradiat-ed for a medulloblastoma in the whole CNS with a seriesof children only operated on for a cerebellar astrocytoma[68]. Global IQ exceeded 90 in 2 and 62%, respectively,writing and reading were altered in 82 and 37%, and be-havior was affected in 93 and 59%. School performancewas severely compromised in 75 and 27%, respectively.For Bloom, the cut-off age is around 5 years, with 38.7%children under this age severely deteriorated (i.e. toIQ<69) and only 4.9% above this age. It is likely that thedose de-escalation that has been attempted by severaloncological groups in low-risk medulloblastomas (from35 down to 25 Gy and even 18 Gy “prophylactic”) andin standard risk ALL (from 24 to 15–18 Gy) will reducesubstantially the risk of severe cognitive dysfunction[59, 61, 107] and also of growth impairment [110].Nonetheless, a recent update on children below 5 years

127

Page 8: Radiation therapy in the management of childhood brain tumors

of age still showed growth redardation following ex-tended 18 Gy “prophylactic” + cis platin [59].

In localized supratentorial tumors that require partialbrain irradiation (but generally in doses in excess of 45 Gy), the relationship with further disturbances is stillpoorly documented. Sparing the prefrontal cortex isprobably of major importance for IQ preservation [124].Deep sites (near the third ventricle and the basal ganglia)and localizations in the right hemisphere have been cor-related with poor outcome [27, 73, 120, 126]. Irradiationof the posterior fossa alone carries its own risks [94] ofintellectual impairment, mainly memory disorders, al-though they are less severe [61]. Interestingly, it hasbeen postulated recently that the cerebellum could con-tribute to mental skills through thalamic and frontal pro-jections [92].

We will also mention the pituitary failures followingirradiation of tumors located in or close to the sellar area.GH and TSH-RH hormones are the most sensitive [18,39, 116]. The risk of pituitary disorders has been set at75% following conventional doses in medulloblastomas.Both GH deficiency following moderate doses or radia-tion (≥24 Gy) and direct impact of ionizing radiation tothe vertebral plates (in case of spinal irradiation) havebeen incriminated in the genesis of growth impairment.The administration of GH can only improve the former..High doses to the cochlea has been made responsible forhypoacusis and stimulated interest in the irradiation ofposterior fossa tumors for elegant beam arrangements(based on conformal posterior oblique fields) that sparethe internal ear [54]. Ovarian transposition should not beomitted when radiation is targeted at the lower sacral fo-ramina in girls.

Recently our group has showed the risk of cerebralvasculopathy in children irradiated for tumors situated

close to the Willis arteries [60]. In association with type-1 neurofibromatosis, 13/69 (19%) children irradiated foran optic glioma presented with clinical and radiologicalsymptoms of vasculopathy.

Future directions

We must emphasize that most of the reported long-termside effects of RT come from old series dealing with“conventional” approaches: high doses administered tothe whole brain or large cerebral volumes. Considerableimpact can be expected in the future from modern strate-gies that deal with:

• More restricted irradiated volumes as permitted byconformal techniques. In such aspects, brachytherapy,intensity modulated photon beam [19], and heavycharged particle programs [63] deserve intensive investi-gations. Combined chemo-radiotherapeutic approachescan also help decrease the need for “prophylactic”medulloblastoma like craniospinal irradiation.

• Optimized delivery of the dose, including biologicalaspects that exploit a differential effect between tumorand normal cell population. Altered fractionation studiesand laboratory experiments on radiosensitivity genesmanipulations are in progress but still largely prelimi-nary.

• Scrutinized neuropsychological evaluations regularlyperformed in the follow-up of patients until adulthood inorder to evaluate the impact of new strategies, and possi-bly to rehabilitate some intellectual deficiencies [131].

Acknowledgement We are grateful to Mrs Anne-Marie Dumainfor her help in the preparation of the manuscript.

128

References

1. Albright AL, Guthkelch AN, PackerRJ, Price RA, Rourke LB (1986) Prog-nostic factors in pediatric brain-stemgliomas. J Neurosurg 65: 751–755

2. Albright AL, Price RA, Guthkelch AN(1983) Brain stem gliomas in children.A clinicopathological study. Cancer 52:2313–2319

3. Alian JC, Kim JH, Packer RJ (1987)Neoadjuvant chemotherapy for newlydiagnosed germ-cell tumors of the central nervous system. J Neurosurg67: 65–70

4. Allen JC, Donahue B, Da Rosso R, Niremberg A (1996) Hyperfractionatedcranio-spinal radiotherapy and adju-vant chemotherapy for children withnewly diagnosed medulloblastoma andother primitive neuro-ectodermal tu-mors. Int J Radiat Oncol Biol Phys 36:1155–1161

5. Alvord EC, Lofton S (1988) Gliomasof the optic nerve or chiasm. J Neuro-surg 68: 85–98

6. Bailey CC, Gnekow A, Wellek S,Jones M, Round C, Brown J, PhillipsA, Neidhardt MK (1995) Prospectiverandomized trial of chemotherapy given before radiotherapy in childhoodmedulloblastoma. International Societyof Paediatric-Oncology (SIOP) and theGerman Society of Paediatric-Oncology(GPO): SIOP II. Med Pediatr Oncol25: 166–178

7. Baranzelli MC, Patte C, Bouffet E, Couanet D, Habrand JL, Portas M, Lejars O, Lutz P, Legall E, Kalifa C(1997) Metastatic intra-cranial germi-noma. The experience of the FrenchSociety of Pediatric Oncology. Cancer 80: 1792–1797

8. Baranzelli MC, Patte C, Bouffet E,Portas M, Mechinaud-Lacroix F, Sariban E, Roche H, Kalifa C (1998)An attempt to treat pediatric intra-cranial α FP and β HCG secretinggerm cell tumors with chemotherapyalone. SFOP experience with 18 cases.J Neurooncol 37: 229–239

Page 9: Radiation therapy in the management of childhood brain tumors

9. Benk V, Bouhnik H, Raquin MA, Kalifa C, Habrand JL (1995) Qualitycontrol of low dose craniospinal irradi-ation for low risk medulloblastoma.BJR 68: 1009–1013

10. Berry MP, Jenkin RDT, Keen CW, NairBD, Simpson WJ (1981) Radiation-treatment for medulloblastoma. A 21-year review. J Neurosurg 55: 43–51

11. Beslac-Bumbasirevic LJ, Boskovic D,Tomin D, Colovic M, Kovacecvic M,Bumbasirevic V(1996) Neuroleukemiain adults. Srp Arkh Tselok Lek 124:82–86

12. Black PML (1991) Brain tumors (First of two parts). N Engl J Med 324:1471–1476

13. Bloom HJG (1982) Intracranial tu-mors: response and resistance to thera-peutic endeavors. Int J Radiat OncolBiol Phys 8: 1083–1113

14. Bloom HJG, Glees J, Bell J (1990) Thetreatment and long-term prognosis ofchildren with intra-cranial tumours: astudy of 610 cases, 1950–1981. Int J Radiat Oncol Biol Phys 18: 723 –745

15. Bloom HJG, Wallace MB, Henk JM(1969) The treatment and prognosis ofmedulloblastoma in children. A studyof 82 verified cases. Am J Roentgenol105: 43–62

16. Bouffet E, Gentet JC, Doz F, Tran P,Roche H, Plantaz D, Thyss A, StephenJL, Lasset C, Carrie C, Alapetite C,Choux M, Mottolese C, Visot A,Zucker JM, Brunat-Mentigny M, Bernard JL (1994) Metastatic medullo-blastoma: the experience of the FrenchCo-operative M7 group. Eur J Cancer30: 1478–1483

17. Brand WN, Schneider PA, Tokars RP(1987) Long-term results of a pilotstudy of low dose cranial-spinal irradi-ation for cerebellar medulloblastoma.Int J Radiat Oncol Biol Phys 13: 1641–1645

18. Brauner R, Adan L, Souberbielle JC(1999) Hypothalamic dysfunction andgrowth in children with intra-cranial le-sions. Child’s Nerv Syst 15: 662–669

19. Carol MP, Woo SY, Butler EB, GrantIII WH (1998) Intensity modulated ra-diation-therapy treatment. In: TobiasJS, Thomas PRM (eds) Current radia-tion oncology, vol 3. Arnold, London,pp 376–395

20. Carrie C, Hoffstetter S, Moncho V,Alapetite C, Murraciole X, Maire JP,Benhassel M, Chapet S, Quetin P, Kolodie H, Lagrange JL, Cuillere JC,Habrand JL (1999) Int Radiat OncolBiol Phys 45: 435–439

21. Castro-Vita H, Salazar OM, Cova M,Rubin P (1978) Cerebellar medullo-blastoma: spread and failure patternsfollowing irradiation. Int J Radiat Oncol Biol Phys 4: 211–212

22. Chiu K, Woo SY, Ater J, Maor MH,Van Eys J, Moser R (1989) Ependymo-ma in children: 31 years experience atthe M.D. Anderson Cancer Center.Proc Am Soc Clin Oncol 8: 302

23. Chin HW, Maruyama Y (1984) Age attreatment and long-term performanceresults in medulloblastoma. Cancer 53:1952–1958

24. Constine L, Konski A, Ekholm S, McDonald S, Rubin P (1987) Adverseeffects of brain irradiation correlatedwith MR and CT imaging. Int J RadiatOncol Biol Phys 13: 88–89

25. Copeland DR (1992) Neuropsychologi-cal and psychosocial effects of child-hood leukemia and its treatment. CA-ACancer J Clin 42: 283–295

26. Cokgor I, Friedman AH, Friedman HS(1998) Gliomas. Eur J Cancer 34:1910–1918

27. Danoff BF, Cowchock FS, MarquetteC, Mulgrew L, Kramer S (1982) As-sessment of the long-term effects ofprimary radiation therapy for brain tumors in children. Cancer 49: 1580–1586

28. Danoff BF, Kramer S, Thompson N(1980) The radiotherapeutic manage-ment of optic nerve gliomas in children.Int J Radiat Oncol Biol Phys 6: 45–50

29. Dearnaley DP, A’Hern RP, Whittaker S,Bloom HJG (1990) Pineal and CNSgerm cell tumors: Royal Marsden Hospital experience 1962–1987. Int J Radiat Oncol Biol Phys 18: 773–781

30. Debus J, Kolagancu KO, Wenz F(1999) Fractionated stereotactic radio-therapy (FSRT) for optic glioma. Int J Radiat Oncol Biol Phys 44: 243–248

31. Delcharco JO, Bolek TW, McColloughWM, Maria BL, Kedar A, Braylan RC,Mickle JP, Buatti JM, Mendenhall NP,Marcus RB (1998) Medulloblastoma:time-dose relationship based on a 30-year review. Int J Radiat Oncol BiolPhys 42: 147–154

32. Deutsch M (1982) Radiotherapy forprimary brain tumors in very youngchildren. Cancer 50: 2785 2789

33. Deutsch M, Thomas PRM, Krischer J,Boyett JM, Albright L, Aronin P,Langston J, Allen JC, Packer RJ, Linggood R, Mulhern R, Stanley P,Stehbens JA, Duffner P, Kun L, RorkeL, Cherlow J, Freidman H, Finlay JL,Vietti T (1996) Results of a prospectiverandomized trial comparing standarddose neuraxis irradiation (3,600 cGy/20)with reduced neuraxis irradiation (2,340 cGy/13) in patients with low-stagemedulloblastoma: a combined chil-dren’s cancer group-pediatric oncologygroup study. Pediatr Neurosurg 24:167–177

34. Dobbing J, Sands J (1973) Quantitativegrowth and development of humanbrain. Arch Dis Child 48: 757–67

35. Dohrmann GJ, Farwell JR, Flannery JT(1976) Glioblastoma multiforme inchildren. J Neurosurg 44: 442–448

36. Dosoretz DE, Blitzer PH, Wang CC,Linggood RM (1980) Management ofglioma of the optic nerve and/or chias-ma. Analysis of 20 cases. Cancer 45:1467–1471

37. Doz F, Kalifa C, Gentet JC, Frappaz D,Demaille ME, Edan C, Chastagner P,Sariban E, Plantaz D, Babin A, Neuenschwander S, Carrie C, Bours D,Mosseri V, Zucker JM (1998) Sand-wich chemotherapy followed by radio-therapy using reduced dose to the neur-axis in standard risk medulloblastoma.A SFOP study. Proceedings of the 8thInternational Symposium on PediatricNeuro-oncology, Rome, May 6–9, 1998,p142

38. Dropcho EJ, Wisoff JH, Walker RW,Allen JC (1987) Supratentorial malig-nant gliomas in childhood: a review offifty cases. Ann Neurol 22: 355–364

39. Duffner PK, Cohen ME, Voorhess ML,MacGillivray MH, Brecher ML, Panahon A (1985) Long-term effects ofcranial irradiation on endocrine func-tion in children with brain tumors. Aprospective study. Cancer 56: 2189–2193

40. Dupuis-Girod S, Hartmann O, Benhammou E, Doz F, Mechinaud F,Bouffet E, Coze C, Kalifa C (1996)Will high dose chemotherapy followedby autologous bone marrow transplan-tation supplant cranio-spinal irradiationin young children treated for medullo-blastoma? J Neurooncol 27: 87–98

41. Eifel PJ (1988) Decreased bone growtharrest in weanling rats with multiple ra-diation fractions per day. Int J RadiatOncol Biol Phys 15: 141–145

42. Eifel PJ, Cassady JR, Belli JA (1987)Radiation therapy of tumors of thebrain stem and midbrain in children:experience of the joint center for radia-tion therapy and children’s hospitalmedical center (1971–1981). Int J Ra-diat Oncol Biol Phys 13: 847–852

43. Epstein F, Wisoff J (1988) Pediatricbrain stem tumors: surgical indications.J Neurooncol 6: 309–317

44. Evans AE, Jenkin DT, Sposto R, Ortega JA, Wilson CB, Wara W, ErtelIJ, Kramer S, Chang CH, Leikin SL,Hammond GD (1990) The treatment ofmedulloblastoma. Results of a prospec-tive randomized trial of radiation thera-py with and without CCNU, vincristineand prednisone. J Neurosurg 72: 572–582

45. Farwell JR, Dohrmann GJ, Flannery JT(1977) Central nervous system tumoursin children. Cancer 40: 3123–3132

129

Page 10: Radiation therapy in the management of childhood brain tumors

46. Finlay JL, Goins SC (1987) Brain tu-mors in children. III. Advances in chemotherapy. Am J Pediatr HematolOncol 93: 264–271

47. Flickinger JC, Torres C, Deutsch M(1988) Management of low-grade glio-mas of the optic nerve and chiasm.Cancer 61: 635–642

48. Flickinger JC, Lusford LD, Singer J,Cano ER, Deutsch M (1990) Megavolt-age external beam irradiation of cranio-pharyngiomas: analysis of tumor con-trol and morbidity. Int J Radiat OncolBiol Phys 19: 117–122

49. Freeman CR, Suissa R (1986) Brainstem tumors in children: results of asurvey of 62 patients treated with ra-diotherapy. Int J Radiat Oncol BiolPhys 12: 1823–1828

50. Freeman CR, Krischer J, Sanford RA,Cohen ME, Burger PC, Kun L, Halperin EC, Crocker I, Wharam M(1991) Hyperfractionated radiationtherapy in brain stem tumors. Resultsof treatment at the 7020 cGy dose levelof Pediatric Oncology Group Study # 8495. Cancer 68: 474–481

51. Freeman CR, Krischer JP, Sanford RA,Cohen ME, Burger PC, Carpio R del,Halperin EC, Munoz L, Friedman HS,Kun LE (1993) Final results of a studyof escalating doses of hyperfractionat-ed radiotherapy in brain stem tumors inchildren: a Pediatric Oncology Groupstudy. Int J Radiat Oncol Biol Phys 27:197–206

52. Freeman CR, Bourgouin PM, SanfordRA, Cohen ME, Friedman HS, Kun LE(1996) Long term survivors of child-hood brain stem gliomas treated withhyperfractionated radiotherapy. Clini-cal characteristics and treatment relatedtoxicities. The Pediatric OncologyGroup. Cancer 77: 555–562

53. Fukunaga-Johnson N, Lee JH, SandlerHM, Robertson P, McNeil E, GoldweinJ (1998) Patterns of failure followingtreatment for medulloblastoma: is itnecessary to treat the entire posteriorfossa? Int J Radiat Oncol Biol Phys 42:143–146

54. Fukunaga-Johnson N, Sandler HM,Marsh R, Martel MK (1998) The use of3D conformal radiotherapy (3D CRT)to spare the cochlea in patients withmedulloblastoma. Int J Radiat OncolBiol Phys 41: 77–82

55. Garrett PG, Simpson WJK (1983)Ependymomas: results of radiationtreatment. Int J Radiat Oncol Biol Phys9: 1121–112

56. Goldwein JM, Corn BW, Finlay JL,Parker RJ, Rorke LB, Schut L (1991)Is cranial irradiation required to curechildren with malignant (anaplasic) in-tracranial ependymomas. Cancer 67:2766–2771

57. Goldwein JW, Glauser TA, Parker RJ,Finlay JL, Sutton LN, Curran WJ,Lacky JM, Rorke LB, Schut L, D’AngioGJ (1990) Recurrent intracranial epen-dymomas in children. Cancer 66: 557–563

58. Goldwein JW, Leaky JM, Parker RJ,Sutton LN, Curran WJ, Rorke LH,Schut L, Littman PS, D’Angio GJ(1990) Intracranial ependymomas inchildren. Int J Radiat Oncol Biol Phys19: 1497–1502

59. Goldwein JW, Radcliffe J, Johnson J,Mushang T, Packer RJ, Suttan LN,Rorker LB, D’Angio GJ (1996) Updat-ed results of a pilot study of low dosecraniospinal irradiation plus chemo-therapy for children under five withcerebellar primitive neuro-ectodermaltumors (medulloblastoma). Int J RadiatOncol Biol Phys 34: 899–904

60. Grill J, Couanet D, Cappelli C, Habrand JL, Rodriguez D, Sainte-RoseC, Kalifa C (1999) Radiation-inducedcerebral vasculopathy in children withneurofibromatosis and optic pathwayglioma. Ann Neurol 45: 393–396

61. Grill J, Renaux VK, Bulteau C, ViguierD, Levy-Piebois C, Sainte-Rose C,Dellatolas G, Raquin MA, Jambaque I,Kalifa C (1999) Long-term intellectualoutcome in children with posterior fossa tumors according to radiationdoses and volumes. Int J Radiat OncolBiol Phy 45: 137–145

62. Habrand JL, Ganry O, Couanet D,Rouxel V, Levy-Piedbois C, Pierre-Kahn A, Kalifa C (1999) The role ofradiation therapy in the management ofcraniopharyngioma: a 25-year experi-ence and review of the literature. Int JRadiat Oncol Biol Phys 49: 255–263

63. Habrand JL, Mammar H, Ferrand R,Pontvert D, Bondiau PY, Kalifa C,Zucker JM (1999) Proton beam therapy(PT) in the management of CNS tu-mors in childhood. Strahlenther Onkol175[Suppl 2]: 91–94

64. Halperin EC (1985) Pediatric brainstem tumors: patterns of treatment fail-ure and their implications for radiother-apy. Int J Radiat Oncol Biol Phys 11:1293–1278

65. Hartsell WF, Hanson WR, ConteratoDJ, Hendrickson FR (1989) Hyperfrac-tionation decreases the deleterious ef-fects of conventional radiation frac-tionation on vertebral growth in ani-mals. Cancer 63: 2452–245

66. Heideman RL, Parker RJ, Albright LA,Freeman CR, Rorke LB (1988) Tumorsof the central nervous system. In: PizzoPA, Poplack DG (eds) Principle andpractice of pediatric oncology. Lippin-cott, Philadelphia

67. Hershatter BW, Halperin EC, Cox EB(1986) Medulloblastoma: the Duke Uni-versity Medical Center experience. IntJ Radiat Oncol Biol Phys 12:1771–1777

68. Hirsch JF, Renier D, Czernikow P,Benvenist L, Pierre-Kahn A (1979)Medulloblastoma in childhood. Survivaland functional results. Acta Neurochir48: 1–15

69. Hoffman HJ, Da Silva M, HumphreysRP, Drake JM, Smith ML, Blaser SI(1992) Agressive surgical managementof craniopharyngiomas in children. J Neurosurg 76: 47–52

70. Horwitch A, Bloom HJG (1985) Opticgliomas: radiation therapy and progno-sis. Int J Radiat Oncol Biol Phys 11:1067–1079

71. Hugues EN, Shillito J, Sallan SE, Loeffler JS, Cassady JR, Tarbell NJ(1988) Medulloblastomas at the JointCenter for Radiation Therapy between1968 and 1984. Cancer 61: 1992–1998

72. Huh SJ, Shin KH, Kim IH, Ahn YC,Ha SW, Park CI (1996) Radiotherapyof intracranial germinomas. RadiotherOncol 38: 19–23

73. Jakacki RI, Zeltzer PM, Boyett JM, Leland Albright AL, Allen JC, GeyerJR, Rorke LB, Stanley PS, StevensKR, Wisoff J, McGuire-Cullen PL,Milstein JM, Packer RJ, Finlay JL(1995) Survival and prognostic factorsfollowing radiation and/or chemothera-py for primitive neuroectodermal tu-mors of the pineal region in infants andchildren: a report of the Children’sCancer Group. J Clin Oncol 13:1377–1383

74. Jannoun L, Bloom HJG (1990) Long-term psychological effects in childrentreated for intracranial tumors Int J Ra-diat Oncol Biol Phys 18: 747–53

75. Janss AJ, Grundy R, Cnaan A, SavinoPJ, Packer RJ, Zackai EH, GoldweinJW, Sutton LN, Radcliffe J, Molloy PT,Phillips PC, Lange BJ (1995) Opticpathway and hypothalamic/chiasmaticgliomas in children younger than age 5 years with a 6-year follow-up. Cancer 75: 1051–1059

76. Jenkin RDT (1969) Medulloblastomain childhood: radiation therapy. CancerMed Assoc J 100: 51–53

77. Jenkin RDT (1982) Childhood ependy-moma radiation treatment results. In:Chang CH, Housepian EM (eds) Tu-mors of the central nervous system.Masson, New York

78. Jereb B, Reid A, Ahuja RK (1982) Pat-terns of failure in patients with medul-loblastoma. Cancer 50: 2941–2947

130

Page 11: Radiation therapy in the management of childhood brain tumors

79. Karim AB, Maat B, Haltevoll R, Menten J, Rutten EH, Thomas DG,Mascarentras F, Horiot JC, ParvimnenLM, Van Rejjn M, Jager JJ, FabriniMG, Van Alphen AM, Hamers HP, Gaspar L, Noordman E, Pierart M, VanGlabbeke M (1996) A randomized trialon dose – response irradiation therapyof low-grade cerebral glioma. EuropeanOrganization for Research and Treat-ment of Cancer (EORTC) study 22844.Int J Radiat Oncol Biol Phys 36: 549–556

80. Khafaga Y, Kandil AE, Jamshed A,Hussanah M, De Val E, Gray AJ(1996) Treatment results for 149 med-ulloblastoma patients from one institu-tion. Int J Radiat Oncol Biol Phys 35:501–506

81. Kim YH, Faoys JV (1977) Intracranialependymomas. Radiology 124:805–808

82. Korinthenberg R (1993) Irradiation-induced brain dysfunction in children.In: Hinkelbein W, Bruggmooer G,Frommhold H, Wanenmacher M (eds)Acute and long-term side effects of ra-diotherapy. (Recent results in cancerresearch, vol 130) Springer, Berlin, pp 199–207

83. Kovalic JJ, Flaris N, Grigsby PW, Pirkowski M, Simpson JR, Roth KA(1993) Intracranial ependymoma. Long term outcome, patterns of failure.J Neurooncol 15: 125–131

84. Kovnar E, Curran W, Tomita T, BurgerP, Langston J, Kepner J, Kun L (1998)Hyperfrationated irradiation for child-hood ependymoma: improved localcontrol in subtotally resected tumors.Proceedings of the 8th InternationalSymposium on Pediatric Neuro-oncology, Rome, May 6–9, 1998, p 115

85. Kovnar EH, Kellie SJ, Horowitz ME,Sanford RA, Langston JW, MulhernRK, Jenkins JJ, Douglas E, EtabanasEE, Fairclough DL, Kun LE (1990)Pre-irradiation cis platin and etoposidein the treatment of high-risk medullo-blastoma and other malignant embryo-nal tumors of the central nervoussystem: a phase II study. J Clin Oncol8: 330–336

86. Kretschmar CS, Tarbell NJ, Barnes PD,Krischer JP, Burger PC, Kun L (1993)Pre-irradiation chemotherapy and hy-perfractionated radiation therapy 66 Gyfor children with brain stem tumors. Aphase II study of the Pediatric Oncolo-gy Group, Protocol 8833. Cancer 72:1404–1413

87. Laitt RD, Chambers EJ, Goddard PR,Wakeley CJ, Duncan AW, Foreman NK(1995) Magnetic resonance imagingand magnetic resonance angiography inlong-term survivors of acute lympho-blastic leukemia treated with cranial ir-radiation. Cancer 76: 1846–52

88. Lamond JP, Mehta MP, Boothman DA(1996) The potential of topoisomeraseI inhibitors in the treatment of CNSmalignancies: report of a symergisticeffect between topotecan and radiation.J Neurooncol 30: 1–6

89. Lannering B, Marky I, Lundberg A,Olsson E (1990) Long-term sequelaeafter pediatric brain tumors: their ef-fects on disability and quality of life.Med Pediatr Oncol 18: 304–310

90. Lefkowitz L, Evans A, Sposto R, Wilson C, Hammond D (1989) Adju-vant chemotherapy of childhood poste-rior fossa ependymoma: craniospinalradiation with or without CCNU, vin-cristine and prednisone. Proc Am SocClin Oncol 8: 87

91. Leibel SA, Sheline GE (1987) Radia-tion therapy for neoplasms of the brain.J Neurosurg 66: I–22

92. Leiner HC, Leiner AL, Dow RS (1986)Does the cerebellum contribute to men-tal skills? Behav Neurosci 100: 443–454

93. Levin VA, Rodrigues LA, EdwardsMSB, Wara W, Liuh C, Fulton D, Davis RL, Wilson CB, Silver P (1988)Treatment of medulloblastoma withprocarbazine hydroxyurea and reducedradiation-doses to whole brain andspine. J Neurosurg 68: 383–387

94. Levy-Piedbois C, Kalifa C, Couanet D,Habrand JL, Grill J, Bulteau C, KiefferV (1998) Sequelae after surgery and ra-diotherapy for ependymoma of the pos-terior fossa in children. Proceedings ofthe 8th International Symposium onPediatric Neuro-Oncology, Rome, May6–9, 1998, p 119

95. Lindstadt D, Wara WM, EdwardsMSB, Hudgins RJ, Sheline GE (1988)Radiotherapy of primary intracranialgerminomas: the case against routinecraniospinal irradiation. Int J RadiatOncol Biol Phys 15: 291–297

96. Loeffler JS, Kretschmar CS, Sallan SE,La Vally BL, Winston KR, Fischer EG,Tarbell NJ (1988) Preradiation chemo-therapy for infants and poor prognosischildren with medulloblastoma. Int JRadiat Oncol Biol Phys 15: 177–181

97. Loeffler JS, Kooy HM, Tarbell NJ(1999) The emergence of conformal ra-diotherapy: special implications for pe-diatric neuro-oncology. Int J RadiatOncol Biol Phys 44: 237–238

98. MacLaughlin MP, Marcus RB, BuattiJM, McCollough VM, Parker Mickle J,Kedar A, Maria BZ, William RR (1998)Ependymoma: results, prognostic factorsand treatment recommendations. Int JRadiat Oncol Biol Phys 40: 845–850

99. MacMillan TJ (1993) In vitro radio-sensitivity of human medulloblasto-ma cell lines. J Neurooncol 15:91–92

100. Mandell LR, Kadota R, Freeman C,Douglass EC, Fontanesi J, CohenME, Kovnar E, Burger P, SanfordRA, Kepner J, Friedman H, Kun LE(1999) There is no role for hyperfrac-tionated radiotherapy in the manage-ment of children with newly diag-nosed diffuse intrinsic brain stem tu-mors: results of a Pediatric OncologyGroup phase III trial comparing con-ventional vs. hyperfractionated radio-therapy. Int J Radiat Oncol Biol Phys43: 959–964

101. Mantravadi RVP, Phatak R, Bellur S,Liebner EJ, Haas R (1982) Brainstem gliomas: an autopsy study of 25cases. Cancer 49: 1294–1296

102. Marks JE, Adler SJ (1982) A com-parative study of ependymomas bysite of origin. Int J Radiat Oncol BiolPhys 8: 37–43

103. Marks JE, Wong J (1985) The risk ofcerebral radionecrosis in relation todose, time and fractionation. A fol-low-up study. In: Hamburger F (ed)Progress in experimental tumor re-search, vol 29. Karger, Basel, pp 210–218

104. Mikaeloff Y, Raquin MA, Lelouch-Tubiana A, Terrier-Lacombe MJ,Zerah M, Bulteau C, Habrand JL,Kalifa C (1998) Primitive cerebralneuro-ectodermal tumors excludingmedulloblastomas: a retrospectivestudy of 30 cases. Pediatric Neuro-surg 29: 170–177

105. Miralbell R, Becher A, Huguenin P,Ries G, Kann R, Mirimanoff RO,Notter M, Novet P, Bieri S, Thum P,Toussi H (1997) Pediatric medullo-blatoma: radiation treatment tech-nique and patterns of failure. Int JRadiat Oncol Biol Phys 37: 523–529

106. Montgomery AB, Griffin T, ParkerRG, Gerdes AJ (1977) Optic nerveglioma: the role of radiation therapy.Cancer 40: 2079–2080

107. Mulhern RK, Kepner JL, ThomasPR, Armstrong FD, Friedman HS,Kun LE (1988) Neuropsychologicfunctioning of survivors of childhoodmedulloblastoma randomized to re-ceive conventional or reduced-dosecraniospinal irradiation: a pediatriconcology group study. J Clin Oncol16: 1723–1728

108. Nakagawa Y, Hatanaka H (1997) Bo-ron neutron capture therapy. Clinicalbrain tumor studies. J Neurooncol 33:105–115

131

Page 12: Radiation therapy in the management of childhood brain tumors

109. Neidhart MK, Bailey CC (1987) Pro-spective randomized cooperativemedulloblastoma trial (MED 84) ofthe International Society of Paedi-atric-Oncology (SIOP) and of the(German) Society of Paediatric On-cology (GPO). Child’s Nerv Syst 3:70–73

110. Ochs J, Mulhern R, Fairclough D(1991) Comparison of neuropsycho-logical functioning and clinical indi-cators of neurotoxicity in long-termsurvivors of childhood leukemia giv-en cranial radiation for perenteralmethotrexate: a prospective study. J Clin Oncol 19: 145–151

111. Packer RJ (1999) Childhood medul-loblastoma: progress and future chal-lenges. Brain Dev 21: 75–81

112. Packer RJ, Sutton LN, Rorke LB,Zimmerman RA, Littman P, BruceDA, Schut L (1985) Oligodendrogli-oma of the posterior fossa in child-hood. Cancer 56: 195–199

113. Packer RJ, Boyett JM, ZimmermanRA, Rorke LB, Kaplan AM, AlbrightAL, Selch MT, Finlay JL, HammondGD, Wara WM (1993) Hyperfrac-tionated radiation therapy (72 Gy) forchildren with brain stem gliomas. AChildrens Cancer Group Phase I/IITrial. Cancer 72: 1414–1421

114. Packer RJ, Boyett JM, ZimmermanRA, Albright AL, Kaplan AM, RorkeLB, Selch MT, Cherlow JM, FinlayJL, Wara WM (1994) Outcome ofchildren with brain stem gliomas af-ter treatment with 7800 cGy of hy-perfractionated radiotherapy. Cancer74: 1827–1834

115. Packer RJ, Goldwein J, NicholsonHS, Vezina LG, Allen JC, Ris MD,Muraszko K, Rorke LB, Wara WM,Cohen BH, Boyett JM (1999) Treat-ment of children with medulloblasto-mas with reduced dose craniospinalradiation therapy and adjuvant che-motherapy: a Children’s CancerGroup study. J Clin Oncol 17: 2127–2136

116. Pasqualini T, Diez B, Domene H,Escobar ME, Gruneiro L, HeinrichJJ, Martinez A, Iorcansky S, Sackmann-Muriel F, Rivarola M(1987) Long-term endocrine sequelaeafter surgery, radiotherapy and che-motherapy in children with medullo-blastoma. Cancer 59: 801–806

117. Phuphanich S, Edwards MSB, LevinVA, Vestnys PS, Wara WM, DavisRL, Wilson CB (1984) Supratentorialmalignant gliomas in childhood. Re-sults of treatment with radiation ther-apy and chemotherapy. J Neurosurg60: 495–499

118. Pierre-Kahn A, Hirsch JF, Roux FX,Renier D, Sainte-Rose C (1983) In-tracranial ependymomas in child-hood: survival and functional resultsof 47 cases. Child’s Brain 10: 145–146

119. Poplack DG, Brouwers PIM (1985)Adverse sequelae of central nervoussystem therapy. Clin Oncol 4: 263–85

120. Potter R (1993) Late side effects ofpediatric radiotherapy. In: HinkelbeinW, Bruggmooer G, Frommhold H,Wanenmacher M (eds) Acute and long-term side effects of radio-therapy. (Recent results in cancer research, vol 130) Springer, Berlin, pp 237–249

121. Prados MD, Wara WM, EdwardsMSB, Cogen PH (1993) Hyperfrac-tionated craniospinal radiation thera-py for primitive neuro-ectodermal tu-mors: early results of a pilot study.Int J Radiat Oncol Biol Phys 28:431–438

122. Price RA, Birdwell DA (1978) Thecentral nervous system in childhoodleukemia. Cancer 423: 717

123. Regine WC, Mohiuddin M, Kramer S(1993) Long-term results of pediatricand adult craniopharyngiomas treatedwith combined surgery and radiation.Radiother Oncol 27: 13–21

124. Reiss AL, Abrams MT, Singer HS,Ross JL, Denckla MB (1996) Braindevelopment, gender and IQ in chil-dren. A volumetric imaging study.Brain 119: 1763–1774

125. Rich TA, Cassady JR, Strand RD,Winston KR (1985) Radiation thera-py for pineal and suprasellar germcell tumors. Cancer 55: 932–940

126. Ris MD, Noll RB (1994) Long-termneurobehavioral outcome in pediatricbrain-tumor patients: review andmethodological critique. J Clin ExpNeuropsychol 16: 21–42

127. Robertson PL, Zeltzer PM, BoyettJM, Rorke LB, Allen JC, Geyer JR,Stanley P, Li H, Albright L, McGuire-Cullen P, Finlay JL,Stevens KR, Milstein JM, Packer RJ,Wisoff J (1998) Survival and prog-nostic factors following radiationtherapy and chemotherapy for epen-dymomas in children: a report of theChildren’s Cancer Group. J Neuro-surg 88, 4: 695–703

128. Rorke LB, Gilles FH, Davis RL,Becker LE (1985) Revision of theWorld Health Organization classifica-tion of brain tumors for childhoodbrain tumors. Cancer 56: 1869–1886

129. Rousseau P, Habrand JL, Sarrazin D,Kalifa C, Terrier-Lacombe MJ, Rekacewicz C, Rey A (1993) Treat-ment of intracranial ependymomas ofchildren: review of a 15-year experi-ence. Int J Radiat Oncol Biol Phys 28:381–386

130. Russo C, Fischbein N, Grant E, Prados MD (1999) Late radiation in-jury following hyperfractionated cra-niospinal irradiation for primitiveneuro-ectodermal tumor. Int J RadiatOncol Biol Phys 44: 85–90

131. Saccone G, Diez B (1998) Pilot studyin the possibilities of cognitive reha-bilitation with specific neuropsycho-logical treatment. Proceedings of the8th International Symposium on Pe-diatric Neuro-Oncology, May 6–9,Rome, p167

132. Salazar OM. Rubin P, Bassano D,Marcial VA (1975) Improved surviv-al of patients with intracranial epen-dymomas by irradiation: dose selec-tion and field extension. Cancer 35:1563

133. Salazar OM, Castro-Vita H, BakosRS, Feldstein ML, Keller B, Rubin P(1979) Radiation therapy for tumorsof the pineal region. Int J RadiatOnco Biol Phys 5: 491–499

134. Salazar OM, Castro-Vita H, Van Houtte P, Rubin P, Aygun C(1983) Improved survival in cases ofintracranial ependymoma after radia-tion therapy: late report and recom-mendations. J Neurosurg 59: 652–659

135. Salazar OM (1983) A better under-standing of CNS seeding and abrighter outlook for postoperativelyirradiated patients with ependymo-mas. Int J Radiat Oncol Biol Phys 9:1231–1234

136. Shaw EG, Evans RG, ScheithauerBW, Ilstrup DM, Earle JD (1987)Post operative radiotherapy of intra-cranial ependymoma in pediatric andadult patients. Int J Radiat Oncol BiolPhys 13

137. Shaw DWN, Goyer JR, Berger MS,Milstein J, Lindoley KL (1997)Asymptomatic recurrence detectionwith surveillance scanning in chil-dren with medulloblastoma. J Clin Oncol 15: 1811–1813

138. Shibamoto Y, Abe M, Yamashita J,Takahshi M, Hiraoka M, Ono K,Tsutsui K (1988) Treatment results of intracranial germinoma as a func-tion of the irradiated volume. Int J Radiat Oncol Biol Phys 15:285–290

139. Shirato H, Nishio M, Sawamura Y,Myohjin M, Kitahara T, Nishioka T,Mizutani Y, Abe H, Miyasaka K(1997) Analysis of long-term treat-ment of intracranial germinoma. Int J Radiat Oncol Biol Phys 37:511–515

132

Page 13: Radiation therapy in the management of childhood brain tumors

140. Silverman CL, Simpson JR (1982)Cerebellar medulloblastoma. The importance of posterior fossa dose tosurvival and patterns of failure. Int J Radiat Oncol Biol Phys 8: 1869–1876

141. Sposto R, Ertel IJ, Jenkin RDT,Boesel CP, Venes JL, Ortega JA,Evans AE, Wara W, Hammond D(1989 ) The effectiveness of chemo-therapy for treatment of high gradeastrocytoma in children: results of arandomized trial. A report from theChildren’s Cancer Study group. J Neurooncol 7: 165–177

142. Stainley P, Suminski N (1988) Theincidence and distribution of spinalmetastases in children with posteriorfossa medulloblastoma. Am J PediatrHematol Oncol 10: 283–287

143. Stroink ARF, Hoffman HJ, HendrickEB, Humphreys RP (1986) Diagnosisand management of pediatric brainstem gliomas. J Neurosurg 65:745–750

144. Sung DI, Chang CH, Harisiadis L,Carmel PW (1981) Treatment resultsof craniopharyngiomas. Cancer 47:847–852

145. Symon L, Sprich W (1985) Radicalexcision of craniopharingioma. Re-sults in 20 patients. J Neurosurg 62:174–181

146. Tait DM, Thornton-Jones H, BloomHJG, Lemerle J, Morris-Jones P(1990) Adjuvant chemotherapy formedulloblastoma: the first multi-centre control trial of the Internation-al Society of Paediatric Oncology(SIOP 1). Eur J Cancer 26: 464–469

147. Tarbell NJ, Barnes P, Scott RM, Goummerova L, Pameroy SL, BlackPMc, Sullan SE, Billett A, La VallyB, Helmus A, Kooy HM, Loeffler JS(1994) Advances in radiation therapyfor craniopharyngioma. Pediatr Neurosurg 21: 101–107

148. Thomas PR, Deutsch M, Mulhern R,Stehbens J, Krishner JP, Boyett JM(1995) Reduced dose vs standarddose neuraxis irradiation in low stagemedulloblastoma: the POG and CCGstudy. Med Pediatr Oncol 24: 277

149. Thomson IL, Grifin TW, Parker RG,Blasko JC (1978) Craniopharyngio-ma: the role of radiation therapy. Int J Radiat Oncol Biol Phys 4:1059–1063

150. Wallner KE, Wara WM, Sheline GE,Davis RL (1986) Intracranial epen-dymomas: results of treatment with a partial or whole brain irradiationwithout spinal irradiation. Int J Radiat Oncol Biol Phys 12:1937–1941

151. Wara WM, Sneed PK, Larson DA(1994) The role of radiation therapyin the treatment of craniopharyngio-ma. Pediatr Neurosurg 21: 98–100

152. Weiss L, Sagerman RH, King GA,Chung TC, Dubony RL (1987) Con-troversy in the management of opticnerve glioma. Cancer 59: 1000–1004

153. Weiss M, Sutton L, Marcial V,Fowble B, Packer R, Zimmerman R,Schut L, Bruce D, D’Angio G (1989)The role of radiotherapy in the man-agement of childhood craniopharyn-gioma. Int J Radiat Oncol Biol Phys17: 1313–1321

154. Wen B-Chen, Hussey DH, HitchanPW, Schelper RL, Vigliotti AP, Doornbos JF, Van Gilder JC (1991)The role of radiation-therapy in themanagement of ependymomas of thespinal cord. Int J Radiat Oncol BiolPhys 20: 781–786

155. Woo C, Stea B, Lulu B, Hamilton A,Cassady JR (1997) The use of stereo-tactic radiosurgical boost in the treatment of medulloblastomas. Int J Radiat Oncol Biol Phys 37:761–764

156. Zelter PM, Boyett JM, Finley JL, Albright L, Rorke LB, Mistein JM,Allen JC, Stevens KR, Stanley P, LiH, Wisoff JH, Geyer JR, McGuire-Cullen P, Stehbens JA, Shurin SB,Packer RJ (1999) Metastasis stage,adjuvant treatment, and residual tu-mor as prognostic factors for medul-loblastoma in children: conclusionsfrom the Children’s Cancer Group921 randomized phase III study. J Clin Oncol 17: 832–845

133