You are on page 1of 8

Int. J. Radiation Oncology Biol. Phys., Vol. 56, No. 2, pp.

511–518, 2003
Copyright © 2003 Elsevier Inc.
Printed in the USA. All rights reserved
0360-3016/03/$–see front matter

doi:10.1016/S0360-3016(02)04611-4

CLINICAL INVESTIGATION Brain

RADIATION THERAPY FOR INTRACRANIAL GERM CELL TUMORS

DAPHNE A. HAAS-KOGAN, M.D.,*† BRIAN T. MISSETT, M.D.,* WILLIAM M. WARA, M.D.,*


SARAH S. DONALDSON, M.D.,‡ KATHLEEN R. LAMBORN, PH.D.,† MICHAEL D. PRADOS, M.D.,†
PAUL G. FISHER, M.D.,§ STEPHEN L. HUHN, M.D.,¶ BENJAMIN M. FISCH, M.D.,*
MITCHEL S. BERGER, M.D.,† AND QUYNH-THU LE, M.D.‡
Departments of *Radiation Oncology and †Brain Tumor Research Center and Neurosurgery, University of California, San
Francisco, CA; Departments of ‡Radiation Oncology, §Neurology, and ¶Neurosurgery, Stanford University Medical Center,
Stanford, CA

Purpose: To review the combined experiences of University of California, San Francisco, and Stanford University
Medical Center in the treatment of intracranial germ cell tumors (GCT) and to assess the impact of craniospinal
radiation (CSI) on patterns of relapse, progression-free survival (PFS), and overall survival (OS).
Patients and Methods: Ninety-three patients received radiation for newly diagnosed intracranial GCTs, including
49 germinomas, 16 nongerminomatous GCTs (NGGCT), and 28 with no biopsy. Median follow-up for surviving
patients was 4.5 years (range 0.25–34). Tests for variables correlating with OS and PFS were conducted using
Cox proportional hazards model.
Results: Five-year PFS and OS rates were 60% ⴞ 15% and 68% ⴞ 14% for patients with NGGCT and 88% ⴞ
5% and 93% ⴞ 4% for those with germinoma. Of 6 patients with localized NGGCT who did not receive CSI,
1 experienced an isolated spinal recurrence but was salvaged. Of 41 patients with localized germinoma, 6 who
received CSI and 35 who did not, no isolated spinal cord relapses occurred. Twenty-one patients with localized
germinoma received neither CSI nor whole brain radiation. Of these, none of 18 with ventricular radiation
relapsed. One of 3 patients with primary tumor radiation relapsed intracranially but had only received 11 Gy at
initial treatment. On multivariate analysis, germinoma histology but not CSI correlated with improved PFS and
OS.
Conclusion: CSI is not indicated in the treatment of localized germinomas. For patients with localized germi-
nomas treated with radiation alone, we recommend ventricular irradiation followed by primary tumor boost to
a total of 45–50 Gy. © 2003 Elsevier Inc.

Germinoma, Brain, Craniospinal irradiation, Pediatric.

INTRODUCTION larly craniospinal irradiation (CSI), has prompted many


investigators to explore approaches that reduce the volume
Intracranial germ cell tumors (GCT) represent 3–11% of and dose of radiotherapy while preserving high cure rates.
pediatric and 1% of adult brain tumors (1). Intracranial The development of strategies to reduce the morbidity of
GCTs comprise two distinct histologic groupings: germino- radiotherapy has been hampered by an absence of random-
mas are the most common histologic subtype, whereas ized trials comparing treatment approaches. As pediatric
nongerminomatous germ cell tumors (NGGCT) represent consortia and cooperative groups prepare to design a ran-
one-third of intracranial GCTs and consist of embryonal domized, phase III trial for children with intracranial GCTs,
carcinoma, endodermal sinus (yolk sac) tumor, choriocar- we sought to use the combined experiences of the Univer-
cinoma, teratoma, and GCT of mixed cellular origin (1, 2). sity of California, San Francisco (UCSF), and Stanford
Historically, radiation alone, frequently encompassing a University Medical Center (SUMC) to shed light on key
large treatment volume, has constituted the gold standard of treatment issues. In particular, historical differences in the
treatment for intracranial GCTs (1, 3–5). However, the radiation volumes prescribed in the two institutions provide
morbidity of radiation therapy in children (6 –11), particu- a cohort of patients that allow assessment of the utility of

Reprint requests to: Daphne A. Haas-Kogan, Department of Supported in part by North American Brain Tumor Consortium
Radiation Oncology, University of California, San Francisco, 1600 grant NCI U01-CA62399 (D.A.H-K.), NIH grant M01 RR01271,
Divisadero, San Francisco, CA 94143. Tel: (415) 353-7175; Fax: Pediatric Clinical Research Center (D.A.H-K.), and CA 82103
(415) 353-9883; E-mail: hkogan@radonc17.ucsf.edu (K.R.L. and M.S.B.).
Presented as an oral presentation at the American Society for Received Aug 19, 2002, and in revised form Dec 6, 2002.
Therapeutic Radiology and Oncology, San Francisco, CA, Novem- Accepted for publication Dec 11, 2002.
ber 4 – 8, 2001.

511
512 I. J. Radiation Oncology ● Biology ● Physics Volume 56, Number 2, 2003

Table 1. Patient characteristics and treatment details

No biopsy or
Germinoma NGGCT nondiagnostic tissue
Pathology (n ⫽ 49) (n ⫽ 16) (n ⫽ 28) Total (93)

Median age (range), years 15 (3–36) 11 (2–20) 17 (3–47) 15 (2–47)


Median follow-up (range),* months 64 (7–213) 40 (12–221) 44 (3–412) 57 (3–412)
Median KPS (range) 90 (60–100) 90 (60–90) 90 (40–90) 90 (40–100)
Female:male 17:32 5:11 6:22 28:65
Localized:disseminated 41:8 11:5 21:7 73:20
Extent of surgery: no surgery 3 0 22 25
Biopsy only 22 4 2 28
Subtotal resection 19 7 2 28
Gross total resection 5 5 2 12
Site (SUMC:UCSF) 14:35 8:8 9:19 31:62
PFS for relapsing patients, months 42 (.72–111) 23 (.20–64) 19 (6–312) 23 (.2–312)
Chemotherapy 9 12 3 24 (26%)
Radiation field: focal field† 21 6 14 41
Whole brain 15 2 7 24
Craniospinal irradiation 13 8 7 28
Dose: median (range) Gy Tumor dose 50 (12–55) 50 (11–54) 50 (20–60) 50 (11–60)
Whole-brain dose‡ 31 (20–49) 24 (11–36) 34 (20–51) 31 (11–51)
CSI dose 30 (24–36) 33 (31–42) 40 (26–46) 32 (24–46)

* Follow-up for surviving patients.



Focal field ⫽ whole ventricle or primary tumor volume plus margin.

Whole-brain dose for patients who did not receive spinal irradiation.
Abbreviations: NGGCT ⫽ nongerminomatous germ cell tumor; KPS ⫽ Karnofsky performance status; PFS ⫽ progression-free survival;
SUMC ⫽ Stanford University Medical Center; UCSF ⫽ University of California, San Francisco; CSI ⫽ craniospinal irradiation.

CSI, because a greater proportion of patients treated at Table 1 delineates the patient characteristics and treat-
SUMC received CSI. Furthermore, we sought to elucidate ment factors. Treatment fields included CSI, whole brain
the role of chemotherapy and the appropriate volume and without spinal irradiation, or more focal fields consisting of
dose of radiation in the treatment of intracranial GCTs. whole ventricular irradiation or involved-field irradiation to
the primary tumor volume plus margin. Twenty-four pa-
tients (26%) received chemotherapy as part of initial treat-
PATIENTS AND METHODS
ment, most commonly: carboplatin, VP 16, bleomycin, cis-
We conducted an historical cohort study of 93 patients platin, cyclophosphamide, and vincristine. For all patients
with confirmed or suspected intracranial GCTs treated in the who received chemotherapy as part of their initial treatment,
departments of radiation oncology at either SUMC or chemotherapy was administered before radiation. Radiation
UCSF, between 1968 and 2001. Excluded patients had therapy after chemotherapy consisted of CSI in 11 patients
received prior radiation, did not receive radiation as part of and focal radiation in the remaining 13 patients.
their initial treatment regimen, or had extracranial metasta- Progression-free survival (PFS) and overall survival (OS)
ses at the time of diagnosis. rates were calculated from date of diagnosis. Median fol-
Histologic confirmations of GCT were obtained in 65 of low-up for surviving patients in the entire cohort was 57
93 cases (70%). Forty-nine patients (53%) had germinomas months (range 3– 412 months). Thirty-six patients had fol-
confirmed by biopsy. Sixteen patients (17%) had biopsy- low-up for longer than 5 years. Patients lost to follow-up
proven NGGCTs, including 8 mixed GCTs, 6 teratomas, 1 were defined as those with less than 5-year follow-up if they
embryonal carcinoma, and 1 choriocarcinoma. The remain- were diagnosed before 1995, or those with no documented
ing 28 patients (30%) were presumed to have intracranial follow-up after January 1, 1998, if they were diagnosed
GCTs based on clinical presentation, biochemical markers, after 1995 (January 1, 1998, represents a 2-year interval
or neuroradiographic imaging that suggested the presence of before compilation of this database).
tumor in the pineal or suprasellar region (12, 13). Twenty- Life-table methods were used to estimate PFS and OS
five of these patients did not undergo biopsy either because (14). Elapsed time from diagnosis to an event or last fol-
they were considered unacceptable surgical risks or had a low-up was used to compute PFS and OS probabilities.
classical radiodiagnostic trial of 20 Gy in an effort to Patients, who were followed without an event or were lost
confirm the presence of a radiation-responsive intracranial to follow-up before disease relapse, were censored on the
GCT (12, 13). Six of the patients classified as having no date of their last imaging study. In analyses of PFS, last
biopsy information available had an attempted biopsy or follow-up dates for patients not experiencing an event were
resection that proved nondiagnostic. based on imaging studies that delineated disease status. For
Radiation for intracranial germ cell tumors ● D. A. HAAS-KOGAN et al. 513

Fig. 1. Actuarial (a) progression-free survival and (b) overall survival, according to histology, of all patients with
intracranial GCT. (— —), pathologically proven germinoma; (- - -), pathologically proven nongerminomatous germ cell
tumor; (——), no biopsy.

OS, deaths, regardless of cause, were coded as events. For NGGCTs and germinomas, respectively (Fig. 1b). Patients
PFS, an event was defined as relapse, progression, or death treated without a biopsy had 5-year PFS and OS rates of
during the period of active follow-up. Kaplan-Meier curves 61% ⫾ 11% and 85% ⫾ 8%, respectively (Figs. 1a and 1b).
for PFS and OS were calculated for germinoma, NGGCT, Within each histologic subtype, PFS and OS rates were not
and unbiopsied tumor subgroups. Additional Kaplan-Meier substantially different for the subgroup of patients present-
curves were calculated to determine the PFS and OS of ing with localized disease (Figs. 2a and 2b). The median
patients with localized disease stratified by histologic sub- PFS for those patients suffering progression or relapse was
group. A final set of Kaplan-Meier curves was calculated for 23 months (range 0.20 –311 months) for the entire cohort,
patients with localized germinoma by whether or not they with 4 patients experiencing an initial relapse more than 5
received CSI. years after diagnosis.
To assess variables influencing PFS and OS, univariate This study included 16 patients with pathologically
analyses were performed to evaluate histology, total tumor proven NGGCT, of whom 3 progressed, 4 relapsed, 1 died
dose, treatment with chemotherapy, and administration of with neither disease progression nor relapse, and 1 was lost
CSI. To further verify the results of univariate analyses, Cox to follow-up. Of 49 patients with pathologically proven
proportional hazards multivariate analyses were conducted germinomas, 1 progressed, 5 relapsed, 1 died with neither
adjusting for radiation dose to the primary tumor, tumor disease progression nor relapse, and 11 were lost to follow-
histology, CSI or whole-brain radiation, Karnofsky Perfor- up. For 11 germinoma patients lost to follow-up, median
mance Status (KPS), chemotherapy, and presence of dis- follow-up was 33.1 months (range 7.2–55.5 months), and all
seminated disease at diagnosis. Analyses of tumor dose but 1 of these 11 were followed for at least 1 year. The
included all patients who received ⱖ30 Gy, excluding 7 overall median follow-up for surviving germinoma patients
patients who received a radiodiagnostic dose of 20 Gy in was 64 months (range 7–213 months).
lieu of histologic diagnoses. Disseminated disease was de-
fined as multifocal tumor, metastases on spinal imaging, or Nongerminomatous germ cell tumors (NGGCT)
CSF cytology revealing malignant cells. This study was Sixteen patients with pathologically proven NGGCTs
performed after approval by the local Human Investigations were included in this analysis—11 with localized and 5 with
Committees. A portion of the patients treated at UCSF and disseminated disease. Of 6 patients with localized disease
SUMC were included in previous, separate reports (5, 15, who did not receive CSI, 1 had an isolated spinal recurrence
16). and 1 an elevated ␤-HCG level (Table 2). Although both of
these patients were salvaged with chemotherapy, with and
without additional radiation, respectively, 2 other patients
RESULTS
died of disease progression evident during their course of
PFS and OS by histology radiotherapy. Of 5 patients who received CSI for localized
Five-year PFS rates were 60% ⫾ 15% for patients with NGGCTs, 1 died of unknown causes 101 months after
biopsy-proven NGGCTs compared with 88% ⫾ 5% for diagnosis.
those with biopsy-proven germinomas (Fig. 1a). Five-year Of the 5 patients presenting with disseminated NGGCTs,
OS rates were 68% ⫾ 14% and 93% ⫾ 4% for patients with 3 received CSI, 1 whole brain without spinal radiation, and
514 I. J. Radiation Oncology ● Biology ● Physics Volume 56, Number 2, 2003

Fig. 2. Actuarial (a) progression-free survival and (b) overall survival, according to histology, of patients with localized
intracranial GCT. (— —), pathologically proven germinoma; (- - -), pathologically proven nongerminomatous germ cell
tumor; (——), no biopsy.

1 focal radiation only. Among disseminated patients treated and none of these 7 patients received CSI. Two patients
with CSI, one recurred synchronously at intracranial and failed synchronously in the brain and spine, 1 failed in the
spinal sites and one progressed while receiving radiation. spine only, 2 relapsed at the primary site, 1 progressed, and
The single patient who received whole-brain radiation ther- 1 died of an arterial thrombosis two decades after receiving
apy had an isolated primary site relapse. All 3 of these radiation therapy. Of the 7 patients with localized disease
relapsed or progressive patients died of their disease. who lacked pathologic diagnoses and relapsed, none had
received chemotherapy.
No biopsy
Twenty-eight patients had no pathologic confirmation of
their diagnoses. Twenty-one of these patients had localized Germinomas
disease and 7 had disseminated disease. Of the 21 patients Forty-nine patients with pathologically proven germino-
without a pathologic diagnosis who had localized disease, 2 mas were analyzed. The 5-year OS and PFS were 93% and
received CSI, 6 received whole-brain radiation, and 2 re- 88%, respectively. Of the 41 patients presenting with local-
ceived chemotherapy. Five of the 7 patients with no patho- ized disease, 6 received CSI, 14 whole brain without spinal
logic diagnosis and disseminated disease received CSI, 1 radiation, and 21 local radiation only. Nine patients received
received whole-brain radiation, and the remaining patient chemotherapy—5 with localized germinoma, and 4 with
received chemotherapy with focal, primary site radiation. disseminated disease.
Of the 21 patients with localized disease who lacked For patients with localized disease, none of 6 patients
pathologic confirmation, 2 received CSI and were disease- who received CSI failed (Fig. 3a). Median follow-up was
free, but these patients had only 6 months and 11 months of 90.7 months (range 14.1–139.4 months), and 5 of these 6
follow-up, respectively. Seven of these 21 patients failed, patients were followed for a minimum of 5 years. For
patients with localized disease, 5 of 35 patients who did not
receive CSI failed, but none experienced an isolated spinal
Table 2. Disease relapse in patients with localized intracranial relapse (Fig. 3a and Table 2). Of the 5 patients who failed,
germ cell tumors according to whether craniospinal irradiation 1 had progressive disease during radiotherapy, 2 had extra–
was administered central nervous system (CNS) metastases, 1 had synchro-
Localized Localized nous spinal and primary site relapses, and 1 had a primary
Variable NGGCT germinoma site failure after discontinuing treatment at 11 Gy to pursue
alternative therapy. Three of the 5 relapsed patients died
CSI Yes No Yes No with disease (Fig. 3b). Of 6 patients who received CSI for
Number of patients 5 6 6 35
Isolated spinal relapse 0 1* 0 0
localized germinoma, 1 died of anaplastic astrocytoma that
Any spinal relapse 0 1 0 1† developed 9 years after CSI (Fig. 3b), and was likely radi-
Recurrences at primary site 0 2 0 3 ation-induced given the in-field location and latency of this
Patients alive and NED 4 4 5 32 second primary tumor.
Eight germinoma patients presented with disseminated
* Patient was salvaged with chemotherapy.

Synchronous primary and spinal failures. disease. Seven of these patients received CSI and were
Abbreviations: NGGCT ⫽ nongerminomatous germ cell tumor; without disease at last follow-up; however, 1 of these 7
CSI ⫽ craniospinal irradiation; NED ⫽ no evidence of disease. patients failed to thrive and died of unknown etiology.
Radiation for intracranial germ cell tumors ● D. A. HAAS-KOGAN et al. 515

Fig. 3. Actuarial (a) progression-free survival and (b) overall survival of patients with pathologically proven localized
germinoma, according to whether craniospinal irradiation was administered. (——), craniospinal irradiation; (— —), no
craniospinal irradiation.

Results by treatment volume yses to evaluate whether these variables are associated with
As the standard of care for localized germinoma has either improved PFS or OS, followed by multivariate anal-
shifted away from CSI (17), a point of controversy has yses to adjust for disease dissemination, histology, tumor
centered on appropriate focal radiation fields. Confusion dose, KPS, and treatment with chemotherapy, craniospinal
persists because many North American studies have not irradiation, and whole brain irradiation.
delineated whether focal radiation fields targeted the tumor Table 3 details the variables tested in multivariate anal-
volume only or included the ventricular system. To address yses and their respective associations with clinical outcome.
this issue, we analyzed recurrence patterns after treatment Analyses of histology included only biopsy-proven germi-
with focal fields including the whole ventricular system or nomas and NGGCTs. For OS, hazard ratios of 0.2 in both
focal fields targeting only the primary tumor volume. Of 41 univariate and multivariate analyses indicated improved
localized germinoma patients, 21 were treated without CSI survival of germinoma patients compared with NGGCT
or whole-brain irradiation, but with more focal radiation patients (p ⫽ 0.01 and p ⫽ 0.07 for univariate and multi-
fields that included whole ventricular radiation in 18 or the variate analyses, respectively). The superior clinical out-
primary tumor plus margin in 3. For the 18 patients treated
with an initial whole ventricular field, the median dose to
the ventricles was 32.4 Gy (range 19.8 –50.4 Gy), and in all Table 3. Multivariate analyses: overall survival and progression-
free survival
but 2 of these patients the ventricular field was followed by
a boost to the primary tumor. Only 3 of these 18 patients Hazard Hazard ratio
received chemotherapy, and none experienced disease re- Variable* ratio 95% CI p value
lapse (median follow-up 57.7 months; range 12.9 –140.0
Overall survival
months). Three patients were treated with a field consisting
Histology† 0.21 0.037–1.13 0.07
of the initial primary tumor volume plus margin. None Tumor dose‡ 0.92 0.82–1.0 0.14
received chemotherapy, and 1 patient failed. This patient’s Craniospinal irradiation 0.58 0.13–2.6 0.46
clinical course is instructive because of the pattern of failure Chemotherapy 1.5 0.33–6.7 0.61
he experienced. His initial suprasellar primary was treated Progression-free survival
Histology† 0.14 0.027–0.67 0.02
with a 2-cm margin to only 11.6 Gy, at which time the
Tumor dose‡ 0.94 0.85–1.1 0.30
patient discontinued treatment. An isolated primary site Craniospinal irradiation 0.62 0.16–2.4 0.48
relapse was treated by surgical resection and chemotherapy Chemotherapy 0.91 0.24–3.5 0.89
followed by radiation, again targeting only the primary
tumor volume plus a 2-cm margin. Six years later the patient * Each variable was tested in a Cox proportional hazards model
adjusting for other potentially confounding variables as detailed in
experienced an intracranial relapse at a site clearly distinct text.
from the original primary tumor, received CSI as salvage †
Analyses of histology included only patients with pathologi-
therapy, and is currently NED (no evidence of disease) 7 cally proven germinomas or nongerminomatous germ cell tumors.
years after completion of CSI. Germinoma histology was associated with enhanced overall sur-
vival and progression-free survival compared with nongermino-
matous germ cell tumor.
Variables predicting clinical outcome in GCT ‡
Analyses of tumor dose included patients who received ⱖ30
Several variables have been suggested as predictors of Gy, excluding those who received a radiodiagnostic dose of 20 Gy.
clinical outcome for GCTs. We performed univariate anal- Abbreviation: CI ⫽ confidence interval.
516 I. J. Radiation Oncology ● Biology ● Physics Volume 56, Number 2, 2003

come of germinoma histology was supported by the analy- compared with NGGCT. Although many published stud-
ses of PFS, in which univariate and multivariate analyses ies group germinoma with NGGCT, their distinct prog-
revealed hazard ratios of 0.2 (p ⫽ 0.07) and 0.1 (p ⫽ 0.02), nosis points to the need to approach these clinical entities
respectively. individually.
In examining tumor dose, identical hazard ratios of 0.9 Published reports corroborate poorer prognosis for pa-
were seen for OS and PFS in both univariate and multivar- tients with NGGCT compared with those with germinoma
iate analyses. Hazard ratios approaching 1 and lack of (2, 18). For NGGCTs, radiation alone produces 5-year sur-
statistical significance suggest that higher tumor dose did vival rates of only 30 – 40% (15). Similarly, we found a
not affect tumor control and clinical outcome. In analyzing 5-year OS rate of only 68% for NGGCT compared with
the effect of tumor dose, we also analyzed separately the 93% for germinoma. More recent studies that demonstrate
patients with localized germinomas. Within this group, uni- excellent response rates to chemotherapy have shifted the
variate and multivariate analyses revealed no significant standard treatment of NGGCT to combined modality ther-
associations between dose to the primary tumor site and apy consisting of chemotherapy and radiation.
either PFS (p ⫽ 0.23 and p ⫽ 0.38, for univariate and The choice of irradiation field in the treatment of local-
multivariate analyses, respectively) or OS (p ⫽ 0.08 and p ized NGGCT remains unclear. The lack of an association in
⫽ 45, for univariate and multivariate analyses, respective- this study between CSI and either PFS or OS may result
ly). Univariate and multivariate analyses indicated no asso- from the small sample size of NGGCT patients. There is
ciation of OS or PFS with either treatment with chemother- scarce literature addressing the appropriate radiation field
apy or administration of CSI (Table 3). for localized NGGCT and only small patient numbers. In a
study by the French Society of Pediatric Oncology, chemo-
Treatment toxicity therapy and focal radiation resulted in five relapses among
Thirty-five patients had documented endocrinopathies be- 24 patients with localized NGGCT (19). A similar regimen
fore initiation of radiation (defined as diabetes insipidus, in a separate study resulted in 3 of 18 patients experiencing
growth retardation, hypothyroidism, adrenocorticotropic disease recurrence, 2 with isolated spinal relapses (20).
hormone (ACTH) deficiency, or sex hormone deficiency). In 5 of the 7 NGGCT patients with disease progression or
Ten patients required initiation of hormone therapy after relapse, the primary site was a component of failure, repre-
completion of radiotherapy. Of these 10 patients, 4 had senting the dominant form of disease relapse. A high inci-
received CSI, 1 had received whole-brain, 4 had received dence of primary site failure in NGGCT has been reported
whole-ventricle, and 1 had received involved field radiation. by others and highlights the need for more intensive che-
Forty-eight patients had normal endocrine profiles and did motherapy or higher radiation doses to the primary site for
not require hormone replacement either before or after patients with poor responses to chemotherapy or radiation
irradiation. (18).
There was one case of decreased luteinizing hormone and Of the various histologic subtypes of intracranial GCTs,
follicle-stimulating hormone levels thought to be secondary germinoma represents the most common and prognostically
to chemotherapy as a result of timing of the declining favorable subgroup. Historically, intracranial germinomas
hormone levels, and one case of panhypopituitarism attrib- have been treated with radiation alone, producing excellent
uted to the radiation treatment, although the time of onset of cure rates (1, 3, 5). The significant long-term toxicities of
the endocrinopathy was not delineated. radiation, particularly in children (6 –11), have prompted
Three patients died after treatment with no evidence of investigations into alternative treatments that minimize the
disease. One patient with disseminated germinoma was dose and volume of irradiation. However, these alternative
diagnosed with failure to thrive after treatment with CSI and approaches must preserve the high cure rates achieved by
chemotherapy and died 43 months after diagnosis, 1 patient radiation alone.
died of an intracranial arterial thrombosis 23 years after Several series have reported excellent clinical outcome
whole-brain irradiation, and 1 patient died of a cerebellar with preirradiation chemotherapy followed by focal irradi-
anaplastic astrocytoma 9 years after CSI. Two patients died ation (21–27). The appeal of this approach lies in lower
of unknown causes after loss to follow-up 9 and 11 years radiation doses and smaller radiation fields that presumably
after treatment, respectively. translate into reduced long-term toxicity. In our retrospec-
tive study the addition of chemotherapy, as assessed by
univariate and multivariate analyses, did not influence either
DISCUSSION
PFS or OS. Of 49 germinomas, only 9 received chemother-
Controversy remains in the treatment of intracranial apy, yet survival rates were excellent. Of note, only 3 of 41
GCTs, due largely to the complexity and rarity of these patients with localized germinoma received focal irradiation
tumors, coupled with a dearth of prospective randomized directed at the primary tumor only, although only 1 of these
trials. Our data, in conjunction with the current literature, patients received chemotherapy, as would be consistent
support distinct therapeutic approaches for germinomas and with the current paradigm of chemoradiation. One of these
NGGCTs. The only factor predictive of superior clinical patients failed, but had been treated suboptimally with only
outcome in this study was the diagnosis of germinoma, as 11 Gy at the time of initial treatment. The remaining 38
Radiation for intracranial germ cell tumors ● D. A. HAAS-KOGAN et al. 517

patients with localized germinoma received whole-ventri- studies report higher recurrence rates associated with radi-
cle, whole-brain, or CSI fields. Therefore, our data cannot ation targeting the tumor volume only (4, 18, 31–33).
address the possible benefit afforded by adding chemother- We recommend whole-ventricular irradiation followed
apy to focal radiation. Radiation alone continues to be the by a boost to the primary tumor for localized germinoma
gold standard for the treatment of intracranial germinomas. when using radiation alone. Some question the rationale of
Promising approaches such as combined chemotherapy and whole-ventricular irradiation given the continuity of CSF
focal, reduced-dose irradiation must be compared with ra- space throughout the entire CNS. Several published studies
diation alone in a prospective, randomized trial. indicate that germinomas may be characterized by multifo-
In addition to the role of chemotherapy, a point of con- cality and intracranial relapses at foci separate from the
troversy in the treatment of localized germinomas has been primary tumor with lesser propensity for diffuse CNS in-
the appropriate field of radiation (17). Previous studies have volvement. These features distinguish germinomas from
reported prospective data suggesting the effectiveness of other CNS malignancies, such as medulloblastoma, that
CSI in preventing locoregional recurrence. In analyzing the require CSI for cure (34). Whether more generous local
combined experiences of SUMC and UCSF, we sought to radiation fields sterilize occult multifocal disease or target
capitalize on historical differences in the fields of radiation direct ventricular invasion, there appears a role for whole-
prescribed for patients with localized germinoma. Such ventricular irradiation in the treatment of localized germi-
patients commonly received CSI at SUMC but generally nomas.
received more limited radiation fields at UCSF. CSI did not Univariate and multivariate analyses failed to document
influence PFS or OS for those with localized germinomas. an association between total dose administered to the pri-
However, we must emphasize that only 6 patients with mary tumor and either PFS or OS. Although our data fail to
localized germinoma received CSI, significantly limiting
show a dose–response, the majority of our patients (72%)
the power of statistical analyses examining CSI. In addition,
received 50 –54 Gy. Therefore we cannot firmly assert that
longer follow-up may be required to identify all failures.
lower doses of radiation will not compromise clinical out-
These limitations notwithstanding, the finding that not a
come. In fact, most contemporary and older studies report
single isolated spinal relapse occurred among 35 patients
superior tumor control rates after radiation doses greater
with localized germinomas treated without CSI, further
than 40 – 45 Gy (29, 35, 36). We believe the literature
corroborates the lack of benefit of CSI. In a single patient,
supports a dose of ⱖ45 Gy to the primary tumor for ger-
the spine was a component of widespread CNS relapse;
minomas treated with radiation alone.
however, no spinal failures were observed in the remaining
4 who recurred. Current evidence substantiates omitting The high cure rate of intracranial GCTs allows assess-
CSI from the treatment of localized germinoma. Spinal ment of long-term toxicities of treatment. Forty-five patients
failure rates of ⬍10% in the absence of CSI, reported in required hormone replacement for various endocrinopa-
most contemporary series, do not justify routine incorpora- thies, although the majority presented with endocrinopa-
tion of CSI into treatment strategies for localized germi- thies before initiation of any treatment. These findings are
noma (4, 16, 18, 28 –30). consistent with a report of germinomas, in which long-term
Nevertheless, the precise irradiation field appropriate for endocrine deficiencies after irradiation were associated ex-
localized germinoma remains unclear. In an attempt to clusively with similar deficiencies before treatment (37).
clarify this issue, we reviewed the irradiation fields of Although curative radiation doses may clearly lead to pitu-
patients with localized germinoma. Of 21 patients who had itary dysfunction (38 – 40), the true contribution of irradia-
neither CSI nor whole-brain irradiation, 3 received fields tion to long-term endocrine deficiencies seen in patients
encompassing only the primary plus margin and 18 received with intracranial GCTs remains uncertain. The only death in
whole-ventricular irradiation. Although no recurrences took a germinoma patient treated with CSI resulted from an
place after whole-ventricular fields, 1 intracranial relapse anaplastic astrocytoma that developed 9 years after irradia-
occurred among 3 patients treated with radiation to the tion. Similar reports of fatal CNS second primary tumors
initial tumor volume plus margin, although this patient highlight the need to explore alternative approaches that
received a suboptimal initial dose of only 11 Gy. Because minimize both the volume and dose of radiation (37). The
the number of patients in our cohort who received irradia- rarity and complexity of intracranial GCTs underscore the
tion confined to the primary tumor only is small, we cannot need to address such issues prospectively in a collaborative
confirm the adequacy of such a focal field. However, several group setting.

REFERENCES
1. Hoffman HJ, Otsubo H, Hendrick EB, et al. Intracranial review of current literature and new advances in therapy.
germ-cell tumors in children. J Neurosurg 1991;74:545–551. Cancer Invest 1997;15:187–198.
2. Kretschmar CS. Germ cell tumors of the brain in children: A 3. Bamberg M, Kortmann RD, Calaminus G, et al. Radiation
518 I. J. Radiation Oncology ● Biology ● Physics Volume 56, Number 2, 2003

therapy for intracranial germinoma: Results of the German 22. Kitamura K, Shirato H, Sawamura Y, et al. Preirradiation
cooperative prospective trials MAKEI 83/86/89. J Clin Oncol evaluation and technical assessment of involved-field radio-
1999;17:2585–2592. therapy using computed tomographic (CT) simulation and
4. Aoyama H, Shirato H, Kakuto Y, et al. Pathologically-proven neoadjuvant chemotherapy for intracranial germinoma. Int J
intracranial germinoma treated with radiation therapy. Ra- Radiat Oncol Biol Phys 1999;43:783–788.
diother Oncol 1998;47:201–205. 23. Fouladi M, Grant R, Baruchel S, et al. Comparison of survival
5. Wolden SL, Wara WM, Larson DA, et al. Radiation therapy outcomes in patients with intracranial germinomas treated
for primary intracranial germ-cell tumors. Int J Radiat Oncol with radiation alone versus reduced-dose radiation and che-
Biol Phys 1995;32:943–949. motherapy. Childs Nerv Syst 1998;14:596–601.
6. Radcliffe J, Packer RJ, Atkins TE, et al. Three- and four-year 24. Sawamura Y, Shirato H, Ikeda J, et al. Induction chemother-
cognitive outcome in children with noncortical brain tumors apy followed by reduced-volume radiation therapy for newly
treated with whole-brain radiotherapy. Ann Neurol 1992;32: diagnosed central nervous system germinoma. J Neurosurg
551–554. 1998;88:66–72.
7. Ellenberg L, McComb JG, Siegel SE, et al. Factors affecting 25. Baranzelli MC, Patte C, Bouffet E, et al. Nonmetastatic intra-
intellectual outcome in pediatric brain tumor patients. Neuro- cranial germinoma: The experience of the French Society of
surgery 1987;21:638–644. Pediatric Oncology. Cancer 1997;80:1792–1797.
8. Packer RJ, Sutton LN, Atkins TE, et al. A prospective study 26. Bouffet E, Baranzelli MC, Patte C, et al. Combined treatment
of cognitive function in children receiving whole-brain radio- modality for intracranial germinomas: Results of a multicentre
therapy and chemotherapy: 2-year results. J Neurosurg 1989; SFOP experience. Société Française d’Oncologie Pédiatrique.
70:707–713. Br J Cancer 1999;79:1199–1204.
9. Waber DP, Tarbell NJ, Fairclough D, et al. Cognitive sequelae 27. Buckner JC, Peethambaram PP, Smithson WA, et al. Phase II
of treatment in childhood acute lymphoblastic leukemia: Cra- trial of primary chemotherapy followed by reduced-dose ra-
nial radiation requires an accomplice. J Clin Oncol 1995;13: diation for CNS germ cell tumors. J Clin Oncol 1999;17:933–
2490–2496. 940.
10. Chapman CA, Waber DP, Bernstein JH, et al. Neurobehav- 28. Dattoli MJ, Newall J. Radiation therapy for intracranial ger-
ioral and neurologic outcome in long-term survivors of pos- minoma: The case for limited volume treatment. Int J Radiat
terior fossa brain tumors: Role of age and perioperative fac- Oncol Biol Phys 1990;19:429–433.
tors. J Child Neurol 1995;10:209–212. 29. Sugiyama K, Uozumi T, Arita K, et al. Clinical evaluation of
11. Moore BD, Ater JL, Copeland DR. Improved neuropsycho-
33 patients with histologically verified germinoma. Surg Neu-
logical outcome in children with brain tumors diagnosed dur-
rol 1994;42:200–210.
ing infancy and treated without cranial irradiation. J Child
30. Haddock MG, Schild SE, Scheithauer BW, et al. Radiation
Neurol 1992;7:281–290.
therapy for histologically confirmed primary central nervous
12. Spiegel AM, Di Chiro G, Gorden P, et al. Diagnosis of
system germinoma. Int J Radiat Oncol Biol Phys 1997;38:
radiosensitive hypothalamic tumors without craniotomy: En-
915–923.
docrine and neuroradiologic studies of intracranial atypical
teratomas. Ann Intern Med 1976;85:290–293. 31. Shibamoto Y, Abe M, Yamashita J, et al. Treatment results of
13. Inoue Y, Takeuchi T, Tamaki M, et al. Sequential CT obser- intracranial germinoma as a function of the irradiated volume.
vations of irradiated intracranial germinomas. AJR Am J Int J Radiat Oncol Biol Phys 1988;15:285–290.
Roentgenol 1979;132:361–365. 32. Shirato H, Nishio M, Sawamura Y, et al. Analysis of long-
14. Kaplan EL, Meier P. Nonparametric estimation from incom- term treatment of intracranial germinoma. Int J Radiat Oncol
plete observations. J Am Stat Assoc 1958;53:457–481. Biol Phys 1997;37:511–515.
15. Fuller BG, Kapp DS, Cox R. Radiation therapy of pineal 33. Aoyama H, Shirato H, Ikeda J, et al. Induction chemotherapy
region tumors: 25 new cases and a review of 208 previously followed by low-dose involved-field radiotherapy for intracra-
reported cases. Int J Radiat Oncol Biol Phys 1994;28:229– nial germ cell tumors. J Clin Oncol 2002;20:857–865.
245. 34. Paulino AC. Radiotherapeutic management of medulloblas-
16. Linstadt D, Wara WM, Edwards MS, et al. Radiotherapy of toma. Oncology 1997;11:813–823.
primary intracranial germinomas: The case against routine 35. Abay EO 2nd, Laws ER Jr, Grado GL, et al. Pineal tumors in
craniospinal irradiation. Int J Radiat Oncol Biol Phys 1988; children and adolescents. Treatment by CSF shunting and
15:291–297. radiotherapy. J Neurosurg 1981;55:889–895.
17. Paulino AC, Wen BC, Mohideen MN. Controversies in the 36. Kersh CR, Constable WC, Eisert DR, et al. Primary central
management of intracranial germinomas. Oncology 1999;13: nervous system germ cell tumors. Effect of histologic confir-
513–521. mation on radiotherapy. Cancer 1988;61:2148–2152.
18. Matsutani M, Sano K, Takakura K, et al. Primary intracranial 37. Hardenbergh PH, Golden J, Billet A, et al. Intracranial ger-
germ cell tumors: A clinical analysis of 153 histologically minoma: The case for lower dose radiation therapy. Int J
verified cases. J Neurosurg 1997;86:446–455. Radiat Oncol Biol Phys 1997;39:419–426.
19. Baranzelli MC, Patte C, Bouffet E, et al. Carboplatin-based 38. Probert JC, Parker BR, Kaplan HS. Growth retardation in
chemotherapy (CT), and focal irradiation (RT) in primary children after megavoltage irradiation of the spine. Cancer
cerebral germ cell tumors (GCT). J Clin Oncol 1999;18:140a. 1973;32:634–639.
20. Robertson PL, DaRosso RC, Allen JC. Improved prognosis of 39. Schriock EA, Schell MJ, Carter M, et al. Abnormal growth
intracranial non-germinoma germ cell tumors with multimo- patterns and adult short stature in 115 long-term survivors of
dality therapy. J Neurooncol 1997;32:71–80. childhood leukemia. J Clin Oncol 1991;9:400–405.
21. Allen JC, DaRosso RC, Donahue B, et al. A phase II trial of 40. Clayton PE, Shalet SM. Dose dependency of time of onset of
preirradiation carboplatin in newly diagnosed germinoma of radiation-induced growth hormone deficiency. J Pediatr 1991;
the central nervous system. Cancer 1994;74:940–944. 118:226–228.

You might also like