Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Int. J. Radiation Oncology Biol. Phys., Vol. 77, No. 1, pp.

113–118, 2010
Copyright Ó 2010 Elsevier Inc.
Printed in the USA. All rights reserved
0360-3016/10/$–see front matter

doi:10.1016/j.ijrobp.2009.04.031

CLINICAL INVESTIGATION Brain

PROSPECTIVE EVALUATION OF RADIOTHERAPY WITH CONCURRENT AND


ADJUVANT TEMOZOLOMIDE IN CHILDREN WITH NEWLY DIAGNOSED DIFFUSE
INTRINSIC PONTINE GLIOMA

RAKESH JALALI, M.D.,* NIRMAL RAUT, M.D.,* BRIJESH ARORA, D.M.,y TEJPAL GUPTA, M.D.,*
DEBNARAYAN DUTTA, M.D.,* ANUSHEEL MUNSHI, M.D.,* RAJIV SARIN, F.R.C.R.,*
y
AND PURNA KURKURE, M.D.

* Department of Radiation Oncology; and y Medical Oncology, Division of Pediatric Oncology, Tata Memorial Centre, Mumbai, India

Purpose: To present outcome data in a prospective study of radiotherapy (RT) with concurrent and adjuvant
temozolomide (TMZ) in children with diffuse intrinsic pontine gliomas (DIPGs).
Methods and Materials: Pediatric patients with newly diagnosed DIPGs were prospectively treated with focal RT
to a dose of 54 Gy in 30 fractions along with concurrent daily TMZ (75 mg/m2, Days 1–42). Four weeks after com-
pleting the initial RT-TMZ schedule, adjuvant TMZ (200 mg/m2, Days 1–5) was given every 28 days to a maximum
of 12 cycles. Response was evaluated clinically and radiologically with magnetic resonance imaging and positron
emission tomography scans.
Results: Between March 2005 and November 2006, 20 children (mean age, 8.3 years) were accrued. Eighteen pa-
tients have died from disease progression, one patient is alive with progressive disease, and one patient is alive with
stable disease. Median overall survival and progression-free survival were 9.15 months and 6.9 months, respec-
tively. Grade III/IV toxicity during the concurrent RT-TMZ phase included thrombocytopenia in 3 patients, leu-
copenia in 2, and vomiting in 7. Transient Grade II skin toxicity developed in the irradiated fields in 18 patients.
During the adjuvant TMZ phase, Grade III/IV leucopenia developed in 2 patients and Grade IV thrombocytopenia
in 1 patient. Patients with magnetic resonance imaging diagnosis of a high-grade tumor had worse survival than
those with a low-grade tumor (p = 0.001). Patients with neurologic improvement after RT-TMZ had significantly
better survival than those who did not (p = 0.048).
Conclusions: TMZ with RT has not yielded any improvement in the outcome of DIPG compared with RT alone.
Further clinical trials should explore novel treatment modalities. Ó 2010 Elsevier Inc.

Brainstem glioma, Diffuse intrinsic pontine glioma, Temozolomide, Radiotherapy, MRI.

INTRODUCTION up to 78 Gy with altered fractionation with or without chemo-


therapy, the final conclusions regarding altered fractionation
Brainstem gliomas constitute approximately 10% of all cen-
schedules have been disappointing (3).
tral nervous system tumors in children and constitute the ma-
The last few years have crystallized our use of temozolo-
jor cause of death among children with brain tumors (1).
Brainstem tumors are recognized as a heterogeneous group mide (TMZ) in glioblastoma in the adult patient population,
with management options ranging from observation in tectal with encouraging results (4, 5). The patient populations in
gliomas with an extremely good prognosis to palliative radio- these series have excluded those with pontine tumors. We
therapy (RT) in the universal aggressive diffuse intrinsic pon- sought to explore the results of using TMZ in pediatric
tine gliomas (DIPGs). DIPGs, because of their location, may DIPGs for many reasons. The favorable features for using
impart a poor prognosis independent of histology (2).
TMZ were its proven efficacy in gliomas, radiosensitization
Improvement in outcome has been elusive despite decades
of intensive clinical and laboratory research, with local tumor potential, low toxicity profile, and suitable kinetic properties.
progression being the main cause of death. Although A Phase II feasibility study was therefore planned and initi-
significant effort has been directed at escalating doses almost ated at our center to test the hypothesis that this option is at

Reprint requests to: Rakesh Jalali, M.D., Department of Radiation Memorial Centre.
Oncology, 125, Tata Memorial Hospital, Parel, Mumbai, India 400 Conflict of interest: none.
012. Tel: (+91) 22 24177153; Fax: (+91) 22 24146937; E-mail: Acknowledgments—We are grateful to NATCO Pharma Ltd. (Hy-
rjalali@tmc.gov.in derabad, India) for supplying temozolomide and the Brain Tumour
Presented at the Eighth Congress of the European Association for Foundation of India for supporting this work.
Neuro-Oncology, Barcelona, Spain, September 12–14, 2008. Received March 4, 2009, and in revised form April 22, 2009.
The study was partly funded by an intramural grant from Tata Accepted for publication April 22, 2009.
113
114 I. J. Radiation Oncology d Biology d Physics Volume 77, Number 1, 2010

least as effective as in adult glioblastoma. The study was ap- week. TMZ was suspended for an absolute neutrophil count lower
proved by our institutional scientific and ethics committees. than 1.5  109/L, platelet count lower than 100,000/mL, and
CTCAE v3.0 (Common Terminology Criteria for Adverse Events
version 3.0) nonhematologic Grade III toxicity or greater.
METHODS AND MATERIALS
Between March 2005 and November 2006, 20 patients were en-
Adjuvant chemotherapy
rolled in this Phase II study per the study protocol approved by our
Adjuvant TMZ was started 28 days after the completion of RT. A
institutional scientific and ethics committees. Eligibility criteria in-
physical, neurologic, hematologic, and biochemical examination;
cluded patients aged between 3 and 21 years, Karnofsky perfor-
MRI scan; and PET-CT scan were performed. The TMZ dosage
mance score of at least 50, and a clinicoradiologic diagnosis of
phase was 200 mg/m2 on Days 1 through 5, and cycles were
DIPG. Required clinical criteria were the presence of at least 2 of
repeated every 28 days. Adjuvant chemotherapy was planned for
the following symptoms: pyramidal tract involvement, cerebellar
a maximum of 12 cycles unless there was clear evidence of progres-
signs and symptoms, and/or cranial nerve deficits attributable to tu-
sion or unacceptable toxicity. Patients underwent a detailed clinical
mor. Radiologic inclusion criteria were based on a computed tomog-
examination and recording of any toxicity profile at each visit during
raphy (CT) or magnetic resonance imaging (MRI) study showing
the adjuvant chemotherapy phase. Radiologic scans were done at the
diffuse involvement of the brainstem ($50% involvement of a brain-
time of the initiation of adjuvant treatment and at any time when
stem segment or involvement of >2 of these segments with or with-
clinical progression was suspected.
out enhancement on injection of intravenous contrast material). A
surgical intervention to obtain histologic proof was attempted in
suitable cases where the tumors had exophytic components and Statistical analysis
surgical biopsy was deemed reasonable. Patients with localized Actuarial survival (overall and progression free) was generated
and focal brainstem lesions were excluded. A repeat MRI study for patients by the Kaplan-Meier method. Toxicity data were re-
along with biological imaging studies including magnetic resonance corded prospectively according to the CTCAE v3.0 criteria. The
spectroscopy (MRS) and magnetic resonance perfusion studies was log-rank test was used to assess survival by prognostic factors
also performed. A final diagnosis of a DIPG was established after such as age, performance status, type of tumor, grade, completion
evaluation by oncologists, neurosurgeons, radiologists, and pathol- of treatment, and so on.
ogists at joint neuro-oncology meetings. Eligible patients had ade-
quate hematologic, renal, and hepatic function (hemoglobin level
$11 g%; total leukocyte count $4,000/mm with absolute neutro- RESULTS
phil count $1,500/mm3 and platelet count $100,000/mm3; liver en- Patient demographic profile
zyme level <2.5 upper limit of normal; and serum creatinine level The mean age of the 20 patients (15 boys and 5 girls) was
<1.5 upper limit of normal). All children who had prior treatment
8.3 years (Table 1). Of the patients, 12 (60%) had a symptom
with any chemotherapy/RT or who had any medical condition
interfering with chemotherapy were not considered for the study. duration of less than 1 month, and 7 patients had all three car-
A positron emission tomography (PET)–CT scan was preferable, dinal clinical signs of DIPG.
though not mandatory, before initiating treatment. Before accrual,
informed consent was obtained from all patients and/or their parents Imaging
or legal guardians. Baseline pretreatment MRI scan evaluation showed typical
diffuse expansile lesions in the pons with frequent extension
Radiotherapy to the medulla, midbrain, and cerebellum. On intravenous
Patients were immobilized with a customized thermoplastic contrast administration, tumors in 6 patients showed no en-
mask. The target volume included all areas of hypodensity seen
hancement, 11 had a patchy enhancement, and 3 had diffuse
on a CT scan or abnormality seen on T2-weighted/fluid-attenuated
inversion recovery sequences of an MRI scan with a 2-cm margin,
enhancement. The mean tumor volume was 12.4 mL (range,
which was reconstructed on the topogram of the diagnostic MRI 2.4–24 mL). MRI perfusion scans showed hyperperfusion in
scan. Bilateral parallel opposing fields were used, and field sizes 13 patients and hypoperfusion in 7. We considered postcon-
were verified on the conventional simulation. A dose of 54 Gy in trast enhancement, magnetic resonance spectroscopy find-
30 daily fractions over a period of 6 weeks was prescribed at the ings, and hyperperfusion on magnetic resonance perfusion
center of the target volume. Radiation was delivered by megavoltage as markers for arbitrarily dividing the tumors into high grade
beam on a 6-MV linear accelerator or a telecobalt machine. Partic- or low grade on MRI. An MRI-based diagnosis of a high-
ular attention was given to encompassing the tumor if going inferi- grade tumor and a low-grade tumor was seen in 12 patients
orly along the spinal cord. and 8 patients, respectively. Eleven patients underwent
a baseline pre-RT PET-CT scan with 5-fluorodeoxyglucose
Concurrent chemotherapy (FDG) as the radiotracer, which showed increased uptake
From the first day of RT, patients were given oral TMZ at a dose in 6 patients and no uptake in 5. Eight patients with an exo-
of 75 mg/m2 for 7 days per week, including on weekends or public
phytic component were considered suitable for a surgical bi-
holidays when RT was not given. TMZ was given on an empty
stomach ($4 hours after last meal) 30 to 60 minutes before RT opsy and had histologic proof of a glioma. Six patients had
and in the morning on days without RT. Prophylactic antiemetics a pathologic diagnosis of a low-grade glioma, and two had
were used during the concurrent chemoradiotherapy phase. Prophy- an anaplastic astrocytoma. All patients with low-grade gli-
lactic anti–Pneumocystis carinii medication was administered in the oma on histology had diffuse involvement of the brainstem
form of oral co-trimoxazole. Blood counts were repeated every consistent with a DIPG. In other 12 patients, surgical
RT and temozolomide in diffuse pontine gliomas d R. JALALI et al. 115

Table 1. Demographic profile of 20 children with diffuse more than six cycles, and 3 completed all 12 cycles of adju-
intrinsic pontine glioma treated with radiotherapy along with vant TMZ.
concurrent and adjuvant temozolomide
Age
Mean (range) 8.3 y (3–18 y) Survival
Median 8y Median overall survival (OS) of the entire cohort was 9.15
Sex months (range, 1.7–47 months), and median progression-free
Male 15 (75%) survival (PFS) was 6.9 months (Fig. 1). Of the patients, 16
Female 5 (25%) lived for more than 6 months, 7 lived for more than 1 year,
Body surface 0.875/m2 (0.56–1.23/m2)
area [mean (range)] and only 3 lived for more than 18 months. At present, 18 pa-
Duration of symptoms tients have died from disease progression, 1 patient is alive
<1 mo 12 (60%) with progressive disease, and 1 patient is alive with stable
>1 mo 8 (40%) disease. Three patients had spinal metastatic disease at the
Clinical presentation
time of disease progression.
Three signs 7 (35%)
(cranial nerve palsy,
long tract signs, and
ataxia)
Response
Cranial nerve 6 (30%) Neurologic recovery was evident in the majority of cases,
palsy and long with reversal of cranial nerve deficits and improvement of
tract signs cerebellar signs. During the initial concurrent RT-TMZ
Cranial nerve 5 (25%) phase, 2 patients did not show any neurologic improvement,
palsy and ataxia
Ataxia and long 2 (10%) 7 patients had only partial improvement, and 11 patients had
tract signs significant neurologic improvement. MRI scan was consid-
KPS ered for all patients who completed at least 4 weeks of
50 5 (25%) post-RT follow-up. One patient died immediately after com-
60 4 (20%) pletion of RT-TMZ, and thus follow-up MRI was not done.
70 5 (25%)
80 6 (30%) Median survival in patients showing a complete response
NPS and no/partial response evaluated on the PET scan after ther-
0, 1 10 (50%) apy was 13.1 months and 14.5 months, respectively
2, 3 10 (50%) (p = 0.966). Response of treatment on FDG-PET therefore
Biopsy
did not seem to correlate with outcome in these patients.
Not done 12 (60%)
Low-grade glioma 6 (75%)
High-grade glioma 2 (25%)
Toxicity profile
Abbreviations: KPS = Karnofsky performance score; NPS = neu- In the concurrent phase Grade I/II leucopenia was seen in 3
rologic performance score. patients and Grade III/IV was seen in 2 (Table 2). One of
these patients with severe leucopenia was diagnosed with
intervention was considered too hazardous and diagnosis was malaria and also had Grade III thrombocytopenia. Grade III
established per typical imaging characteristics. thrombocytopenia was observed in 2 patients (1 patient
also had leucopenia). Therefore 3 patients had Grade III/IV
hematologic toxicity (2 patients with both thrombocytopenia
and leucopenia and 1 patient with only thrombocytopenia).
Treatment-related parameters
All patients accrued in the trial completed the planned con-
current RT and TMZ phase. The mean body surface area of
the entire cohort of patients at the initiation of the treatment
was 0.875/m2 (range, 0.56–1.23/m2). The mean dose of
TMZ in the concurrent and adjuvant settings was 65 mg
(range, 40–90 mg) and 174.6 mg (range, 100–260 mg), re-
spectively. None of the patients required discontinuation of
RT except in 1 patient where RT had to be temporarily with-
held for 3 days because of a heightened skin reaction in the
RT portals with superadded infection. Four patients required
admission during treatment with concurrent RT and TMZ
(three because of Grade III vomiting and one with Grade
III thrombocytopenia). The median duration of radiation
treatment was 41 days (range, 39–49 days). Of the patients,
2 received no adjuvant TMZ (1 died before starting the adju-
vant phase and Grade IV toxicity developed in 1), 8 received Fig. 1. Overall survival (in months). Cum = cumulative
116 I. J. Radiation Oncology d Biology d Physics Volume 77, Number 1, 2010

Table 2. Toxicity profile of 20 children with a diffuse icantly better survival (p = 0.048). Histologic diagnosis of the
intrinsic pontine glioma treated with radiotherapy along with tumor did not show any significant impact on OS (p = 0.112).
concurrent and adjuvant temozolomide
Patients with features of high-grade tumor on radiologic im-
Concurrent RT and TMZ aging, however, had a poorer outcome than those with low-
Hemoglobin grade tumors (p = 0.001). Age, performance status, symptom
Grade I 1 (5%) duration, and severity of hydrocephalus did not show any im-
Grade II 1 (5%)
Leukocyte count pact on survival function.
Grade III 1 (5%)
Grade IV 1 (5%) DISCUSSION
Platelet count
Grade I 1 (5%) With TMZ emerging as an integral component of manage-
Grade III 2 (10%) ment in adult malignant brain tumor in recent years, we
Grade IV 1 (5%) sought to examine its role in the equally challenging scenario
Vomiting of pediatric diffuse pontine gliomas. The protocol design was
Grade I 1 (5%) similar to the one used for adult glioblastomas except that the
Grade II 11 (55%)
Grade III 6 (30%) number of adjuvant TMZ cycles was increased to 12 (instead
Grade IV 1 (5%)
Skin toxicity
Table 3. Correlation of various prognostic factors with
Grade I 1 (5%)
overall survival
Grade II 18 (90%)
Grade III 1 (5%) Mean overall
Adjuvant TMZ Factors Parameters survival (mo) p Value
Hemoglobin
Grade I 1 (5%) Age <8 y 10.97 0.917
Grade II 2 (10%) >8 y 10.33
Leukocyte count Sex Male 11.76 0.317
Grade I 1 (5%) Female 8.76
Grade II 1 (5%) KPS at presentation <70 9.17 0.134
Grade III 1 (5%) >70 12.48
Grade IV 1 (5%) NPS at presentation 0, 1 11.29 0.648
Platelet count 2, 3 10.57
Grade IV 1 (5%) Symptom duration <1 mo 10.85 0.913
Vomiting >1 mo 11.3
Grade II/III 7 (35%) Clinical Three signs 8.26 0.158
Hospitalization during concurrent RT-TMZ 6 (30%) presentation
Grade III/IV thrombocytopenia 3 (50%) Two signs 12.50
Grade III Vomiting 3 (50%) MRI tumor volume <12 cm2 9.04 0.249
Febrile neutropenia during adjuvant TMZ 2 (10%) >12 cm2 13.96
Varicella-zoster infection 2 (10%) MRI contrast Non-enhancing 13.98 0.166
enhancement
Abbreviations: RT = radiotherapy; TMZ = temozolomide. Enhancing 9.63
During the concurrent phase, the most common nonhema- MRI grade High grade 8.87 0.043
Low grade 15.10
tologic toxicity was vomiting. Grade III/IV vomiting was seen MRS grade High grade 8.79 0.258
in 7 patients (35%). Skin toxicity was common but not severe. Low grade 12.99
Fever was seen in 2 patients; this was proven to be of malarial Magnetic resonance Hyperperfusion 8.87 0.043
etiology in 1. Among the 19 patients eligible for adjuvant perfusion
Hypoperfusion 15.10
TMZ, adjuvant treatment was started in 18 whereas 1 patient
PET scan No uptake 14.28 0.911
did not receive any adjuvant TMZ because of the develop- Uptake present 13.78
ment of Grade IV thrombocytopenia during the concurrent MRI + MRS + High grade 6.8 0.001
phase. During the adjuvant phase, Grade III/IV leucopenia perfusion
was observed in 2 patients and Grade IV thrombocytopenia diagnosis
Low grade 15.14
in 1. No further adjuvant TMZ was given in these patients. Histopathologic High-grade glioma 7.05 0.112
Varicella-zoster infection developed in 2 patients during the diagnosis
adjuvant phase, and they were treated conservatively. In 1 pa- Low-grade glioma 14.19
tient with lower cranial nerve palsy, aspiration pneumonia de- Clinical response Partial/no response 8.23 0.048
veloped after the second cycle of adjuvant therapy, which Significant response 13.31
PET response No/partial response 14.59 0.966
subsided with conservative management, and adjuvant Complete response 13.1
TMZ was restarted 2 weeks after the scheduled time.
Abbreviations: KPS = Karnofsky performance score; NPS = neu-
Prognostic factors rologic performance score; MRI = magnetic resonance imaging;
Patient-related factors were analyzed with respect to OS MRS = magnetic resonance spectroscopy; PET = positron emission
(Table 3). Patients with neurologic improvement had signif- tomography.
RT and temozolomide in diffuse pontine gliomas d R. JALALI et al. 117

of 6 in adults). Treatment was generally fairly well tolerated. not show any survival advantage (14, 15). A randomized trial
In the concurrent phase Grade I/II leucopenia and Grade III/ with concurrent chemotherapy and dose escalation also
IV leucopenia were seen in 25% and 10% of cases, respec- showed no significant benefit over conventional treatment
tively. Grade III thrombocytopenia was observed in 10% pa- (Pediatric Oncology Group 9239) (16). Broniscer et al.
tients. In this phase of treatment the most common severe (17) reported the results of a multi-institutional study exam-
nonhematologic toxicity was vomiting. The toxicity profile ining the role of neoadjuvant irinotecan followed by RT and
was well within the acceptable range in the adjuvant phase. then TMZ. Median survival was only 12 months (1-year OS
Our prospective series of patients with DIPGs treated rate, 48%) with a high incidence of hematologic toxicities.
aggressively by use of concurrent RT and TMZ followed Cohen et al. (18) have presented a summary of prospective
by adjuvant TMZ, however, showed a median OS and PFS data of 61 DIPG patients treated with concurrent TMZ and
of 9.15 months and 6.93 months, respectively. These findings RT, followed by adjuvant TMZ. The 1-year OS rate was
were not better than the results generally known historically only 39%, and 61% of patients died within 12 months of di-
or results in a previous study (2). With standard conventional agnosis and 89% within 18 months. This trial also concluded
RT, median PFS and OS range from 5 to 8.8 months and 7 to that TMZ does not improve the outcome in children with
16 months, respectively (2). There was no difference in the DIPGs. Another small study using a combination of TMZ
survival function and response rate in our study population and cis–retinoic acid with RT has also failed to show any
treated with RT and TMZ compared with historical data. meaningful impact (19). Biological agents such as the anti-
Therefore the addition of TMZ to RT was disappointing epidermal growth factor receptor agent Nimotuzumab have
and did not show any benefit in this group of patients, in shown some promise in a preliminary phase II study, and
contrast to what has been found in adult glioblastomas. the efficacy of Nimotuzumab requires further evaluation (20).
A majority of our cases showed some neurologic recovery Because we have reached a relatively difficult situation for
at the time of evaluation after the conclusion of the initial any chemoradiotherapy schedule, as in other sites, a major
concurrent RT-TMZ phase. It is known that RT leads to neu- breakthrough regarding survival improvement could be
rologic improvement in many of these patients and may allow expected to come from molecular studies (21). However,
a reduction of steroid dose (6). It was seen that patients with a major limitation in conducting any molecular/biological
significant neurologic improvement could expect a better studies in DIPG is the lack of tissue diagnosis. The location
outcome, because they had a mean OS of 13.31 months of DIPG does not allow any attempt at surgical excision.
whereas patients with little neurologic recovery had a mean High-grade gliomas in DIPG may have different molecular
OS of only 8.23 months. Although a majority of our patients genetics and may require different treatment strategies, but
showed significant neurologic improvement after the initial this requires confirmation by molecular studies. However,
concurrent RT-TMZ phase, even a partial radiologic re- because biopsy is usually not done in most patients and there-
sponse was observed only in 50% of cases assessed by fore molecular biology of the disease is largely unknown,
MRI scan at 4 weeks’ post-RT follow-up. Patients with fea- DIPG is usually diagnosed by clinicoradiologic means (22).
tures of high-grade tumor on imaging showed poorer sur- Imaging is therefore likely to play an important role in char-
vival. Other factors such as age, initial performance status, acterizing these tumors and may well provide some insights
symptom duration, and presence or absence of hydrocepha- into the molecular profile and perhaps even therapies ad-
lus did not show any impact on survival. Although histologic dressing the affected pathways. Biological imaging methods
diagnosis of high-grade glioma did not affect the survival such as PET scan have been used recently. FDG-PET scan is
outcome on comparison with low-grade glioma statistically, nonspecific, and amino acid–based PET studies are usually
this finding may be because of the smaller number of patients recommended for central nervous system neoplasms. In our
in our study cohort. study the FDG-based PET scan findings did not show any
Despite a number of clinical trials having been conducted correlation with survival. Among the newer MRI modalities,
over the last several decades to improve results in patients perfusion scan findings had a significant survival function
with DIPGs, the outcome in these patients has been frustrat- correlation. The perfusion scan correlation may be because
ing and has remained largely unchanged. Numerous Phase I of angiogenesis mechanisms, and thus future trials with anti-
/II trials were conducted with altered RT fractionation sched- angiogenic target molecules could be attempted as well.
ules, concurrent chemoradiotherapy, radiation sensitizers, There are numerous theories as to why TMZ has failed to
and systemic CT (7–10). In a series of radiation dose–escala- have an impact in patients with a DIPG. Apart from different
tion studies, doses from 64.8 to 78 Gy with hyperfractionated molecular biology and response to TMZ, other reasons could
radiotherapy had shown increased toxicity without any sur- be the compromised dose of TMZ delivered in pediatric
vival benefit (11). A trial of concurrent carboplatin and etopo- brainstem glioma patients. It would be interesting to perform
side at a dose of 70.2 Gy with hyperfractionated radiotherapy methylguanine methyltransferase analysis to determine its
did not improve outcome over RT alone (2-year survival rate, correlation with TMZ response. Methylguanine methyltrans-
11%) (12). There was no benefit with the addition of a regi- ferase analysis was not performed in our patients. The lack of
men of lomustine, vincristine, and prednisone (13). Other histologic material in a large majority of these patients with
chemotherapeutic schedules, interferon, bradykinin, RMP- DIPGs is likely to be a major hindrance in carrying out
7, cyclosporine, and tamoxifen with or without RT also did such analyses.
118 I. J. Radiation Oncology d Biology d Physics Volume 77, Number 1, 2010

In summary, TMZ with RT has not yielded any improve- Further clinical trials should examine hitherto unknown
ment in the outcome of DIPG compared with RT alone. novel treatment modalities.

REFERENCES
1. Smith MA, Freidlin B, Ries LA, et al. Trends in reported inci- diagnosed diffuse pontine gliomas: A phase I/II study. Med Pe-
dence of primary malignant brain tumors in children in the diatr Oncol 1998;30:28–33.
United States. J Natl Cancer Inst 1998;90:1269–1277. 13. Jenkin RD, Boesel C, Ertel I, et al. Brain-stem tumors in child-
2. Donaldson S, Laningham F, Graham P, et al. Advances toward hood: A prospective randomized trial of irradiation with and
an understanding of brainstem gliomas. J Clin Oncol 2006;24: without adjuvant CCNU, VCR and prednisone: A report from
1266–1272. the Children’s Cancer Study Group. J Neurosurg 1987;66:
3. Prados MD, Wara WM, Edwards MSB, et al. The treatment of 227–233.
brain stem and thalamic gliomas with 78 Gy of hyperfractio- 14. Broniscer A, Leite CC, Lanchote VL, et al. Radiation therapy
nated radiation therapy. Int J Radiat Oncol Biol Phys 1995; and high-dose tamoxifen in the treatment of patients with dif-
32:85–91. fuse brainstem gliomas: Results of a Brazilian cooperative
4. Stupp R, Mason WP, Van Den Bent MJ, et al. Radiotherapy study. Brainstem Glioma Cooperative Group. J Clin Oncol
plus concomitant and adjuvant temozolomide for glioblastoma. 2000;18:1246–1253.
N Engl J Med 2005;10:987–996. 15. Greenberg ML, Fisher PG, Freeman C, et al. Etoposide, vincris-
5. Jalali R, Basu A, Gupta T, et al. Encouraging experience of con- tine, and cyclosporin A with standard-dose radiation therapy in
comitant temozolomide with radiotherapy followed by adjuvant newly diagnosed diffuse intrinsic brainstem gliomas: A Pediat-
temozolomide in newly diagnosed glioblastoma multiforme: ric Oncology Group phase I study. Pediatr Blood Cancer 2005;
Single institution experience. Br J Neurosurg 2007;21: 77:2150–2156.
583–587. 16. Korones DN, Fisher PG, Kretschmar C, et al. Treatment of chil-
6. Langmoen IA, Lundar T, Storm-Mathisen I, et al. Management dren with diffuse intrinsic brain stem glioma with radiotherapy,
of pediatric pontine gliomas. Childs Nerv Syst 1991;7:13–15. vincristine and oral VP-16: A Children’s Oncology Group
7. Packer RJ, Lange B, Ater J, et al. Carboplatin and vincristine for phase II study. Pediatr Blood Cancer 2008;50:227–230.
recurrent and newly diagnosed low-grade gliomas of childhood. 17. Broniscer A, Iacono L, Chintagumpala M, et al. Role of temo-
J Clin Oncol 1993;11:850–856. zolomide after radiotherapy for newly diagnosed diffuse brain-
8. Bouffet E, Raquin M, Doz F, et al. Radiotherapy followed by stem glioma in children: Results of a multiinstitutional study
high dose busulfan and thiotepa: A prospective assessment of (SJHG-98). Cancer 2005;103:133–139.
high dose chemotherapy in children with diffuse pontine glio- 18. Cohen K, Heideman R, Zhou T, et al. Chemoradiotherapy with
mas. Cancer 2000;88:685–692. temozolomide in the treatment of diffuse intrinsic pontine glio-
9. Jakacki RI, Siffert J, Jamison C, et al. Dose-intensive, time com- mas of childhood: Results of the children’s oncology group
pressed procarbazine, CCNU, vincristine (PCV) with peripheral (COG) ACNS 0126 trial [Abstract]. Neuro Oncol 2007;
blood stem cell support and concurrent radiation in patients with 9(Suppl.):539.
newly diagnosed high-grade gliomas. J Neurooncol 1999;44: 19. Sirachainan N, Pakakasama S, Visudithbhan A, et al. Concur-
77–83. rent radiotherapy with temozolomide followed by adjuvant te-
10. Levin VA, Edwards MS, Wara WM, et al. 5-Fluorouracil and 1- mozolomide and cis-retinoic acid in children with diffuse
(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) followed intrinsic pontine glioma. Neuro Oncol 2008;10:577–582.
by hydroxyurea, misonidazole, and irradiation for brain stem 20. Bode U, Windelberg M, Massimino M, et al. Phase III trial of
gliomas: A pilot study of the Brain Tumor Research Center nimotuzumab for the treatment of newly diagnosed diffuse in-
and the Childrens Cancer Group. Neurosurgery 1984;14: trinsic pontine gliomas in children and adolescents [Abstract].
679–681. J Clin Oncol 2008;26(Suppl.):2058.
11. Mandell LR, Kadota R, Freeman C, et al. There is no role for 21. Louis D, Rubio MP, Correa KM, et al. Molecular genetics of
hyperfractionated radiotherapy in the management of children pediatric brain stem gliomas: Application of PCR techniques
with newly diagnosed diffuse intrinsic brainstem tumors: Re- to small and archival brain tumor specimens. J Neuropathol
sults of a Pediatric Oncology Group phase III trial comparing Exp Neurol 1993;52:507–515.
conventional vs. hyperfractionated radiotherapy. Int J Radiat 22. Tzika AA, Astrakas LG, Zarifi MK, et al. Spectroscopic
Oncol Biol Phys 1999;43:959–964. and perfusion magnetic resonance imaging predictors of
12. Walter AW, Gajjar A, Ochs JS, et al. Carboplatin and etoposide progression in pediatric brain tumors. Cancer 2004;100:
with hyperfractionated radiotherapy in children with newly 1246–1256.

You might also like