You are on page 1of 12

Chapter 45

Chemotherapy of Brainstem Gliomas


Viviana Benitez and Matthias A. Karajannis
Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States

INTRODUCTION
enhancement on postcontrast MRI. When biopsied, these
Brainstem gliomas account for 10%–20% of pediatric brain tumors have pilocytic astrocytoma histology [2]. The third
tumors [1]. The median age at diagnosis ranges from 5 to type of brainstem glioma is composed of focal tectal
9 years of age. Notably, brainstem gliomas represent a gliomas, and these account for about 5% of brainstem
range of diseases rather than a single entity, ranging from gliomas. These tumors often have >2 months of symptoms
low-grade tumors which may require only limited treatment at diagnosis and usually have signs of elevated intracranial
and carry a favorable prognosis to high-grade tumors that pressure such as headaches and emesis. They are located in
are rapidly progressive despite aggressive therapy. the region of the tectal plate and commonly have hydro-
About 80% of brainstem gliomas are located in the pons, cephalus on imaging. Histology is usually that of low-grade
whereas the remaining 20% can be found in the medulla, glioma [2]. Of note, brainstem gliomas associated with NF1
midbrain, or cervicomedullary junction. Historically, pedi- are considered as a separate entity. Patients with NF1 more
atric brainstem gliomas have been divided into three main commonly have optic pathway gliomas, but a small per-
categories with distinct imaging and clinical features: centage can develop gliomas in the brainstem. These
diffuse intrinsic pontine gliomas (DIPG), focal tectal tumors tend to behave less aggressively, can sometimes
gliomas, and posterior exophytic/cervicomedullary gliomas be asymptomatic, and regress on occasion.
[2]. DIPG account for 80% of brainstem gliomas. The Alternative diagnoses should be considered when eval-
typical duration of symptoms in DIPG patients is short uating focal brainstem gliomas, namely primitive neuroec-
(<2 months), and they classically present with ataxia, long todermal tumors (PNET) of the brainstem. These are often
tract signs, and/or cranial nerve deficits. DIPG are recog- characterized by younger age of onset, a focal none-
nized by characteristic imaging findings of a diffuse pontine nhancing tumor, and aggressive tumor behavior and
lesion, encasing the basilar artery. At the time of diagnosis, growth [4]. Of note, recent advances in molecular classifi-
contrast enhancement on magnetic resonance imaging cation have allowed the demonstration that a significant
(MRI) may or may not be present, and is not predictive proportion of tumors previously diagnosed as PNETs, in
of outcome [3]. The diagnosis can be made based on clinical fact display molecular profiles indistinguishable from
and imaging features alone, and when biopsied the histo- other CNS entities, namely ATRTS, ETMRs, and HGG
logic appearance may range from diffuse astrocytoma or [5]. Other possibilities to consider in the differential diag-
anaplastic astrocytoma to glioblastoma. Regardless of nosis include histiocytic lesions, tuberculoma, or abscess.
initial histological grade, however, progression-free sur- In these cases, pathological diagnosis with a biopsy is
vival (PFS) in DIPG patients is generally 5–6 months and indicated.
overall survival (OS) <1 year [1,2]. Factors that seem to affect prognosis of brainstem
Posterior exophytic/cervicomedullary gliomas account gliomas include presentation with cranial nerve palsies, evi-
for 10%–15% of brainstem gliomas. These patients usually dence of hypodense tumor involving the entire brainstem,
have a longer duration of symptoms at diagnosis and malignant tumor histology such as mitoses. These are
(>2 months) and often present with headache and emesis, typically associated with shorter survival [6]. Favorable
as well as dysphagia and weakness. The tumors usually prognostic factors include the diagnosis of NF1, symptom
arise from the floor of the fourth ventricle or from the cer- duration >12 months at diagnosis, calcification noted on
vicomedullary junction. On imaging, they tend to be focal the tumor, exophytic location of the glioma, and low-grade
with a posterior exophytic component, and typically show pathology including the presence of Rosenthal fibers [7].

Handbook of Brain Tumor Chemotherapy, Molecular Therapeutics, and Immunotherapy. https://doi.org/10.1016/B978-0-12-812100-9.00046-2


Copyright © 2018 Elsevier Inc. All rights reserved. 585
586 SECTION V Chemotherapy for Specific Tumors—Pediatrics

DIAGNOSTIC APPROACH

Clinical presentation

Signs of elevated ICP: lethargy, Brainstem findings: cranial neuropathies,


headache, emesis focal weakness, ataxia

Duration of symptoms

<2 months duration >2 months duration

Diagnostic Imaging:

MRI brain with and without contrast


preferred modality

+/– MR spectroscopy

Pontine, diffuse, variably Floor of fourth ventricle,


enhancing, encasing basilar dorsally exophytic,
artery enhancing

DIPG Cervicomedullary glioma

Atypical clinical history,


Focal, tectal plate, evidence
radiographic appearance, or
hydrocephalus
research trial

Tectal glioma Consider tissue biopsy

OVERVIEW OF INITIAL TREATMENT


The treatment paradigm for brainstem gliomas varies is paramount, given their management will differ signifi-
depending on their location and histology. Recognition of cantly from that of the more common DIPG. Biopsy of
the more favorable low-grade histology brainstem gliomas DIPG is safe using modern techniques and should be
Chemotherapy of Brainstem Gliomas Chapter 45 587

considered in the context of clinical trials [8]. Specifically, and 53%, respectively. Notably, this treatment was well tol-
biopsy can confirm diagnosis and may identify actionable erated without significant effect in quality of life [20].
molecular targets and help risk stratification in both
treatment and prognosis. Recurrent Treatment for Focal Brainstem
Gliomas
Focal Brainstem Gliomas
A variety of regimens have shown to be active in the setting
In the 15%–20% of brainstem gliomas that are low-grade of recurrent LGG, including vinblastine, bevacizumab with
astrocytomas, management can include observation, sur- or without irinotecan, everolimus, and a metronomic oral
gical resection, and if not resectable or progressive, then antiangiogenic regimen (celecoxib, thalidomide, fenofi-
RT and/or chemotherapy [9]. Focal brainstem gliomas brate, cyclophosphamide, and etoposide) [21–24]. Clinical
account for 20% of brainstem gliomas, usually occurring trials are presently evaluating the use of BRAF or MEK
outside the pons. Histologically, these are either pilocytic inhibitors for the treatment of recurrent LGG, including
astrocytomas or, less commonly, diffuse astrocytomas. focal brainstem gliomas [25]. In the case of focal brainstem
Similar to pilocytic astrocytomas located in other regions gliomas, RT has been found to be effective in inducing pro-
of the brain, genetic alterations leading to MAPK sig- longed remission; however, given severe associated toxic-
naling pathway activation, most commonly involving ities in young children in particular, it should be reserved
BRAF, have been identified in the majority of focal for patients who have failed other treatment options. This
brainstem gliomas [10]. For instance, BRAF-KIAA1549 is especially so given the excellent prognosis for these
fusions have been found in 66%–80% of all juvenile tumors [26].
pilocytic astrocytomas, including 62% of those in the Brainstem gliomas in patient with Neurofibromatosis
brainstem, whereas BRAF V600E mutations are more Type 1 behave biologically different from lesions with a
common in the grade II diffuse astrocytomas as well as similar appearance in patients without this disorder.
in pleomorphic xanthoastrocytomas and slightly less Although these lesions may sometimes clinically and radio-
common in gangliogliomas [11–13]. In contrast to DIPG graphically progress, most do not require specific inter-
and other diffusely infiltrating tumors, focal brainstem vention, and rather, can be observed. A retrospective
gliomas are often amenable to surgical resection, particu- review of 21 patients with NF1 and brainstem lesions
larly those that are exophytic or cervicomedullary in showed that most were biologically indolent with only
location [14]. If resection is not feasible, stereotactic 10 having radiographic progression or clinical progression
biopsy can be helpful in establishing a diagnosis, which at 3.75 years. Seven of those patients had tumors stabilize in
is greatly aided by currently available molecular testing. size or regress without intervention. Only four patients
In tectal gliomas, due to typical presentation of obstructive required treatment, and these were the focally enhancing
hydrocephalus, third ventriculostomy or VP shunt tumors. Therapy consisted of biopsy, excision, or radio-
placement are often effective [15]. therapy. All lesions were low grade and stabilized after
In the case of progression or inoperable disease, chemo- therapy. The authors concluded that intervention of these
therapy is the preferred treatment. The combination of vin- tumors can be limited to lesions that rapidly grow on serial
cristine and carboplatin has been shown to achieve stable imaging or produce significant clinical symptoms [27].
disease in 68%–75% of patients [16]. Another regimen com-
monly used is the combination of thioguanine, procarbazine,
Diffuse Intrinsic Pontine Gliomas
lomustine (CCNU), and vincristine (TPCV). A randomized
controlled trial comparing the use of these two regimens in DIPG account for approximately 80% of pediatric
patients under age 10 with low-grade gliomas showed brainstem gliomas. They range from WHO grade II–IV
improved 5-year PFS of 52% for TPCV versus 39% for on pathological evaluation, but grade does not seem to
vincristine-carboplatin [17]. However, due the increased affect prognosis. H3K27M mutations in two histone genes
toxicity of TPCV, including prolonged marrow suppression have been found in about 80% of DIPG [28]. H3K27M
and the risk of secondary myelodysplastic syndrome/acute mutant DIPG invariably harbor additional oncogenic driver
leukemia associated with procarbazine and lomustine, vin- mutations, most commonly including TP53, PDGFRA, and
cristine and carboplatin remains the preferred upfront ACVR1, and is recognized as a clinicopathological entity
regimen at many institutions [18,19]. Recently, the Canadian with other H3K27M mutant midline gliomas including tha-
Pediatric Brain Tumor Consortium evaluated the use of lamic and spinal cord tumors, all of which show similar
weekly vinblastine (6 m/m2) over 70 weeks in therapy-naı̈ve molecular features, as well as equally aggressive behavior
pediatric patients with progressive low-grade gliomas. Out and resistance to conventional therapies. Another much less
of 54 patients studied, 25.9% had a complete, partial, or commonly found mutation in those without the histone
minor response. The 5-year OS and PFS rates were 94% mutation is MYCN amplification [29].
588 SECTION V Chemotherapy for Specific Tumors—Pediatrics

RT remains the only therapy proven to improve the sur- damage was noted in 6/14 autopsied patients. They con-
vival in patients with DIPG despite numerous clinical trials cluded that there was no evidence of improved survival with
and ongoing research. Conventional RT leads to clinical higher dose HRT and there was evidence of prolonged use
improvement in the majority of these patients (65%) but steroid dependency in their cohort of patients [31].
PFS is short (<6 months in most) and survival is poor Another phase I/II trial evaluated a group of children
(<1 year median survival) [1]. Evaluation of hyperfractio- and adults with brainstem glioma treated with HRT at a
nated RT (HRT) compared with conventional RT has failed total dose of 72 Gy. Median time to tumor progression
to show a benefit, and higher doses of 75.6 and 78 Gy can (TTP) was 59 weeks (66 weeks in adults, 44 weeks in
lead to significant morbidity including steroid dependency, children) and MST was 74 weeks (92 weeks in adults and
vascular events, white matter changes, hormone defi- 64 weeks in children). Notably, there was no increased radi-
ciencies, seizure disorders, and hearing loss [30,31]. Fur- ation toxicity noted with the fractionation schedule and
thermore, studies looking at chemotherapy plus RT have dose studied [34]. HRT at a dose of 72 Gy was again looked
shown poor response to both single and multiagent reg- at in a phase I/II trial by the Children’s Cancer Group
imens, both prior to RT and as adjuvant chemotherapy (CCG) in 53 children with diffuse intrinsic or malignant
[9,32]. The remainder of this chapter will focus in more brainstem glioma. Overall 53% of patients had an objective
detail on the treatment of DIPG. response, with a partial response in 13%. The OS rate was
38% at 1 year and 14% at 2 years. Children with symptoms
lasting >2 months prior to diagnosis had a better prognosis.
RADIATION THERAPY IN DIPG
The authors concluded that while imaging response rates
The current standard treatment for newly diagnosed DIPG were increased using HRT, no improvement in overall
is fractionated focal intensity-modulated radiation therapy outcome was observed compared with conventional
(IMRT) to the tumor bed to a total dose of 54–60 Gy (given RT [35].
as 1.8–2 Gy fractions once daily for 5 days per week over
6 weeks). About 70%–80% of patients with DIPG respond
to RT with some transient neurological improvement. One
Hypofractionated Irradiation in DIPG
study evaluating timing of RT showed no significant impact Other studies have compared the use of hypofractionated
on the outcome with duration of time from diagnosis to start RT versus conventional RT in the treatment of pediatric
of RT [33]. DIPG. One such randomized controlled trial evaluated
72 children with newly diagnosed DIPG and randomized
them for treatment with hypofractionated RT, where they
Hyperfractionated Irradiation in DIPG received 39 Gy in 13 fractions 5 days weekly, versus a
Higher doses of RT using HRT (66–78 Gy) do not appear to regimen of conventional RT, where they received 54 Gy
provide a survival advantage when compared with standard in 30 fractions over 6 weeks. Their results showed that
dose and fractionated protocols. For instance, the Pediatric OS and PFS were slightly shorter (not a statistically signif-
Oncology Group (POG) conducted a phase I/II study to icant difference) for the hypofractionated group than the
assess the efficacy and toxicity of sequentially escalated standard group. However, the authors noted that this
doses of HRT in children with malignant brainstem method may put fewer burdens on the patients and near
gliomas. In their study, they compared a dose of 75.6 Gy comparable results [36]. Another study looked at
with results from doses of 66 and 70.2 Gy. Treatment with 27 children with DIPG and treated them with either
the higher dose of 75.6 Gy resulted in neurological 44.8 Gy given in 16 fractions or 39 Gy given in 13 fractions.
improvement in 77% of patients, with a PFS of 7 months They compared these regimens to a matched cohort of
and a median survival time (MST) of 10 months. Toxicities patient receiving conventional RT. They found no signif-
or morbidity noted in their study included skin reactions and icant difference in OS and PFS between these regimens,
steroid use >3 months. Their conclusion was that there was and the hypofractionated schedule allowed patients to
no evidence of improved outcome with higher doses of spend significantly less time receiving RT. Another point
HRT [30]. Another trial conducted by the Children’s made by the authors was that in most patients (78%), this
Oncology Group (COG) was a phase I/II trial with the goal allowed for steroids to be weaned off earlier [37].
to determine the outcome of treatment with HRT to a total
dose of 78 Gy in children with DIPG. They noted clinical
Reirradiation at Time of Progression
improvement in 34% of patients, with 14% of patients
showing a >50% reduction in their tumor size radiograph- A study looking at a small series of patients who received
ically. The OS in this trial was 35% at 1 year and 11% at re-irradiation at the time of progression with 18–20 Gy
3 years. Of note, 33/66 patients required steroid treatment focal RT over 2 weeks showed that this therapy was gen-
for prolonged periods, and radiation-induced necrotic erally well tolerated with most showing both clinical and
Chemotherapy of Brainstem Gliomas Chapter 45 589

radiographic improvement, with MST of 5 months to sub- (1: 50 mg/m2/120 h, 2: 75 mg/m2/120 h, 3: 100 mg/m2/
sequent progression [38]. This benefit of PFS at the time of 120 h) and grade 2–4 myelosuppression was seen in 3/5
re-irradiation was also demonstrated in another single- patients treated at dose level 3. One child died of massive
institution clinical trial [39]. The SIOP-E-HGG/DIPG hemorrhage at the tumor site after 2 days of treatment [32,42].
working group also noted a survival benefit with A phase I/II study evaluating carboplatin combined with
re-irradiation at the time of progression in patients with HRT (total dose 72 Gy) showed that the cumulative
DIPG. They noted that median OS was 13.7 months for maximum tolerated dose for carboplatin was 1540 mg/
patients undergoing re-irradiation (versus 10.3 months with m2. The dose-limiting toxicity was hematologic. In this
upfront radiation). They also noted clinical improvement in study, the median PFS was 8 months and OS was 12 months
77% of patients, with no grade 3–4 toxicity noted [40]. [43]. Another study by the Brainstem Glioma Cooperative
Group (BGCG) evaluated the efficacy of RT plus tamoxifen
CHEMOTHERAPY OF DIPG in patients with DIPG. They used a treatment dose of
200 mg/m2 daily of tamoxifen along with RT and then
Different strategies using chemotherapy to treat DIPG, such for 52 additional weeks. In their study, 50% of the patients
as high-dose chemotherapy followed by stem cell rescue, showed a radiographic response, with an MST of
neoadjuvant chemotherapy (single or multiagent), con- 10.3 months. While overall well tolerated, the study showed
current chemotherapy with RT, and adjuvant chemo- no significant change in overall prognosis [44].
therapy, have not demonstrated any survival benefit when More recently, in the German cooperative group HIT-
compared with conventional RT alone. Notably, the current GBM studies using RT combined with intensive chemo-
standard of therapy in the treatment of adult GBM, RT with therapy, survival was improved compared with historical con-
concurrent and adjuvant temozolomide (TMZ), has not trols only in pediatric patients with high-grade glioma whose
shown any improved outcome in pediatric patients with tumors were gross totally resected, but not in other patients,
DIPG. Below is a summary of some of the chemotherapy including DIPG patients. In the HIT-GBM-C protocol,
regimens studied in DIPG and their outcomes. patients with newly diagnosed HGG including DIPG were
treated with conventional RT and concurrent chemotherapy
Concomitant Chemotherapy and Irradiation (one cycle of cisplatin + etoposide + vincristine (PEV), fol-
lowed by weekly vincristine, and then one cycle of
Approaches
cisplatin + etoposide + ifosfamide (PEI) in the last week of
Some studies have focused on evaluating the use of chemo- RT). They were subsequently treated with maintenance che-
therapy concurrent with RT. One randomized phase III motherapy consisting of additional cycles of PEI followed by
study evaluated chemotherapy with vincristine, CCNU, valproic acid. Of 97 patients, 21 had gross total resection and
and prednisone together with RT total dose of 50–60 Gy 29 had partial resection. OS was 91% at 6 months, 56% at
compared with RT alone in newly diagnosed patients. In 12 months, and 19% at 60 months. The 5-year OS for patients
this study, PFS was similar in both arms, MST was with gross total resection treated on this protocol was 63%
9 months in both arms, and OS was 17% for RT alone compared with 17% for the historical control group (HIT-
versus 23% for the RT plus chemotherapy group. Therefore, GBM-A and HIT-GBM-B). No difference in survival was
no significant difference in the outcome was observed. Of observed in patients with incompletely resectable disease,
note, an increased risk of infection was associated with including DIPG patients [45].
the adjuvant therapy. Limitations of this study, conducted TMZ has been studied in many clinical trials for DIPG,
in the late 1970s, include limited imaging modalities (pri- given its proven benefit in prolonging survival in adult
marily CT was used) which could confound the cohort patients with GBM [46]. However, pediatric studies evalu-
of patients, with perhaps some lower grade histologies ating its use along with RT have not shown any benefit over
included [41]. historical controls using RT alone. For instance, an open-
More recently, chemotherapy has been used concur- label phase II study (ACNS0126) by the COG evaluated
rently with RT as a radiosensitizer. In the majority of these the outcome of TMZ concurrent with RT followed by
studies, while there is no excess toxicity with the addition of adjuvant TMZ. They studied 63 children with newly diag-
chemotherapy to RT, there is also no added benefit noted. nosed DIPG, with a TMZ dose of 90 mg/m2/day  42 days
A Pediatric Oncology Group (POG) study compared along with RT at a dose of 59.4 Gy, followed by mainte-
outcome in patients with brainstem gliomas treated with nance treatment with TMZ at a dose of 200 mg/m2/
HRT at a dose of 70.2 Gy versus similar RT plus cispla- day  5 days every 28 days for 10 cycles, beginning 4 weeks
tinum (as a radiosensitizer) by continuous infusion on after completion of RT. The mean EFS was 14% and mean
weeks 1, 3, and 5 of RT and showed worse survival in OS was 40%, with median time to death of 9.6 months. This
patients receiving HRT plus cisplatinum versus HRT alone. study showed chemoradiotherapy with TMZ followed by
Note that in the phase I study, three dose levels were studied adjuvant TMZ was not more effective than RT alone
590 SECTION V Chemotherapy for Specific Tumors—Pediatrics

[47]. A UK phase II trial studied RT plus concomitant TMZ electrolyte abnormalities, and ototoxicity. The authors con-
(75 mg/m2) followed by up to 12 courses of 21 days of cluded that while chemotherapy can be added prior to HRT
adjuvant TMZ (100 mg/m2) every 4 weeks. The OS was in these patients with both clinical and objective responses
56% at 9 months and 35% at 1 year. Median survival was seen, other agents would need to be identified for overall
9.5 months. Overall, this trial showed no survival benefit improved outcome and survival [52].
with the addition of TMZ in patients treated with standard The Children’s Cancer Group (CCG) evaluated the
RT [48]. The German HIT-HGG 2007 study reported six efficacy of two chemotherapy regimens prior to hyperfrac-
children (2 with DIPG, 4 with high-grade gliomas) treated tionated external beam radiation therapy (HFEBRT) (total
with CSI and concurrent metronomic TMZ (75 mg/m2/ dose of 72 Gy) in children with primary high-risk brainstem
day). Median PFS was 4 months and median OS was tumors. Regimen A consisted of three courses of carbo-
7.6 months. Major toxicities included myelosuppression, platin, etoposide, and vincristine, whereas Regimen
with treatment interrupted or discontinued in 4/6 patients B consisted of cisplatin, etoposide, cyclophosphamide,
due to >grade 3 hematotoxicity [49]. and vincristine. Radiographic response was evaluated with
Of note, one large series studied the prognostic signifi- MRI after induction chemotherapy and HFEBRT. Overall
cance of genomic and epigenetic alterations through 10% of the patients treated with Regimen A responded
molecular analysis of >200 pediatric glioblastomas. In their objectively to chemotherapy; 27% improved with com-
study, MGMT promoter hypermethylation was found in bined chemotherapy and RT. For those treated with
about 25% of pediatric glioblastomas, with marked vari- Regimen B, 19% responded to chemotherapy alone and
ability among molecular subgroups. MGMT promoter 23% to both chemotherapy and RT. In both groups, EFS
methylation status has been established as a robust pro- was 17% at 1 year and 6% at 2 years. Notably, grade 3–4
spective and predictive (for treatment benefit with TMZ) leukopenia were observed in 60% of patients treated with
biomarker in adult glioblastoma [50]. Interestingly, K27 Regimen A and 90% of those treated with Regimen B. In
tumors (more commonly those found in midline structures conclusion, neither the chemotherapy regimen improved
such as thalamus and brainstem) very rarely showed the percentage of patients who responded nor the PFS or
MGMT methylation (3%). In fact, midline tumor location, OS compared with prior series of patients treated with
K27M mutation, and presence of oncogene amplifications RT alone [53].
each were associated with a poor outcome. This study Overall outcome in the above studies that looked at
may explain some of the reasoning for the poor outcome older children was poor, with MST less than a year, and
and lack of efficacy of TMZ-based therapy for DIPG, no improved outcome when compared with RT alone.
given the lack of MGMT promoter methylation in these Results were better when evaluating postoperative chemo-
tumors [51]. therapy and delayed radiation in a younger subset of
patients. The “Baby POG” study evaluated children less
than 36 months old with biopsy proven malignant brain
Neoadjuvant Chemotherapy Approaches
tumors. These children were treated with two cycles of
Some studies have looked at chemotherapy given prior to cyclophosphamide plus vincristine, and two cycles of cis-
RT. This method of treatment is more often used in younger platin plus etoposide. This sequence of chemotherapy
children with the intent to delay radiation until more than was continued until either the disease progressed or the
3 years old. Limitations of these studies include the fact that children were >48 months of age. Complete or partial
despite initiation of chemotherapy prior to RT, in many response was seen in 39% of patients after two cycles of
cases the patient went on to receive RT as planned. This chemotherapy. PFS was 41% at 1 year and 39% at 2 years.
was often due to the early progression seen with chemo- Notably, this study included multiple types of malignant
therapy, leading to discontinuation of chemotherapy and brain tumors, and patients with brainstem gliomas or
initiation of RT. embryonal tumors had little or no response [54]. Also
A phase II study from the Pediatric Oncology group important to note is that infant GBM patients have long
(POG) evaluated the response of treating children with been known to have a better prognosis than GBM in older
brainstem tumors with chemotherapy followed by HRT children. Recent studies indicate that this improved
(total dose of 66 Gy). The chemotherapy regimen consisted outcome is likely due to distinct oncogenic drivers seen
of four cycles of cisplatin (100 mg/m2) and cyclophos- in infant GBM patients. Specifically, infant high-grade
phamide (3 g/m2). They studied 32 patients, of whom gliomas often lack K27M mutations or TP53 mutations,
65% improved clinically after two to three cycles. Radio- and are more likely to have NTRK gene fusions as a
graphically, three patients had partial responses (defined potential driver [55].
as >50% shrinkage) and 23 patients had stable disease. Frappaz and colleagues studied frontline chemotherapy
Six patients were noted to have progressive disease. MST with tamoxifen/BCNU/cisplatin alternating with high-dose
was 9 months. Toxicities included marrow suppression, methotrexate, aimed at delaying radiation therapy until the
Chemotherapy of Brainstem Gliomas Chapter 45 591

time of progression. They found a significantly improved RT completion. In terms of the irinotecan, 16/33 patients
median survival of 17 months compared with 9 months in were treated with this regimen, but 6 stopped it due to
historical controls. However, they also noted prolonged clinical progression and 1 due to toxicity. In terms of the
hospitalization and increased infections in the chemo- treatment with TMZ, grade 3–4 neutropenia and thrombo-
therapy group [55]. The German HIT-GBM protocols eval- cytopenia were noted in 33% and 29% respectively, of TMZ
uated a variety of chemotherapy regimens in the treatment cycles. Median survival was 12 months with a 1-year sur-
of DIPG. One study (HIT-GBM-D protocol) evaluated vival of 48%. Again, this study did not show improved
frontline therapy with high-dose methotrexate as a single prognosis with the use of TMZ after RT [60].
agent for pediatric high-grade glioma and DIPG. Patients
received 24-h infusions of methotrexate (5 g/m2) on days
1 and 15, followed by 54 Gy of RT with concurrent cis-
Chemotherapy for Recurrent DIPGs
platin, etoposide, vincristine, and ifosfamide. Overall, after Treatment with single-agent chemotherapy, multiagent reg-
treatment with methotrexate, 18/19 had stable disease and imens, and high-dose chemotherapy with autologous stem
1/19 had a partial response. Event-free survival was noted cell rescue have been studied extensively in patients with
to be 43% at 1 year. This was comparable to other regimens recurrent brainstem gliomas. The overall response seen in
studied by this group; however, not shown to be superior to these studies is very poor and short lived [9].
outcome with RT alone [56]. In studies looking at single-agent chemotherapy in
Studies evaluating treatment of newly diagnosed recurrent disease, the overall response rates were poor, with
brainstem gliomas with high-dose chemotherapy and autol- very short lasting effect seen, if any effect at all. For
ogous stem cell rescue have also been disappointing. For instance, in a phase I trial of high-dose cyclophosphamide
instance, one study evaluated nine children with newly (80 mg/kg or greater) in children with recurrent brain
diagnosed high-risk brain tumors treated with high-dose tumors the overall response rate was 89% and the mean
thiotepa/cyclophosphamide chemotherapy followed by duration of response was 7 months. In the seven patients
autologous bone marrow infusion and involved field with recurrent gliomas, there was one complete response
HRT. Six of these patients were suffering from brainstem and four partial responses. The main toxicity seen was
gliomas. Two patients had partial responses, one had a hematologic [61]. Another phase II trial looked at carbo-
minor response, three had stable disease, and one patient platin (560 mg/m2 every 4 weeks) in 95 patients with
had progressive disease. Two patients, one with brainstem recurrent brain tumors. Complete or partial responses were
glioma, achieved a complete remission following radiation. seen in 1/19 children with brainstem glioma [62]. In a study
This OS was not felt to be better than standard therapy [57]. that looked at 12 children with recurrent brainstem gliomas
Another phase II study evaluated the use of high-dose treated with VP-16 (50 mg/m2 every 21 days), six patients
BCNU (800 mg/m2) followed by autologous stem cell demonstrated a radiographic response (one complete, three
transplant in children with high-grade gliomas. Seven of partial, two stable disease) with a median duration of
these patients had DIPGs. Only one objective response response of 8 months [63].
was noted 1 month after treatment; five patients had stable Study of multiagent chemotherapy in patients with
disease and seven progressed [58]. recurrent disease has shown some response, but this has
appeared short lived. For instance, in a study looking at
the treatment of recurrent brainstem gliomas with a regimen
Adjuvant Chemotherapy Approaches
consisting of 5-FU, CCNU, hydroxyurea, and 6-MP, 76%
Other studies have focused on the use of chemotherapy responded with stabilization or disease improvement, but
agents following RT in the treatment of DIPG. The majority progression occurred in 25 weeks. This is less than the
of these have been demonstrated to be either ineffective or median survival from recurrence of brainstem gliomas
unable to show prolonged survival or time to progression. which is 27 weeks [64].
For instance, one pilot study looking at the feasibility and Studies have also looked at high-dose chemotherapy
benefit of high-dose chemotherapy after RT in patients with with autologous stem cell rescue in recurrent disease. These
DIPG showed three deaths related to high-dose chemo- studies have not shown improved outcome. For instance, in
therapy complications but no survival benefit (MST of a phase II study that evaluated high-dose busulfan (150 mg/
10 months) [59]. m2  4 days) and thiotepa (350 mg/m2  3 days) with autol-
The use of TMZ has also been evaluated as a treatment ogous bone marrow transplant in childhood malignant
modality after radiotherapy for newly diagnosed DIPG in tumors, five partial responses were observed (all in patients
children. A multiinstitutional study allowed for treatment with diagnoses other than brainstem gliomas), and one
with intravenous irinotecan for two cycles prior to conven- objective response was seen in a patient with brainstem
tional RT. They then evaluated the use of TMZ (200 mg/ glioma. In this study, toxicity was high with bone marrow
m2) for 5 days for a total of six cycles, starting 4 weeks after aplasia and complications affecting skin, liver, and
592 SECTION V Chemotherapy for Specific Tumors—Pediatrics

neurological status [65]. Dunkel and colleagues have inves- placed infusion catheter (ClinicalTrials.gov identifier
tigated the effectiveness of high-dose thiotepa (300 mg/m2) NCT01502917); results for this trial are pending.
and etoposide (250–500 mg/m2) regimens with autologous With increased efforts in recent years to perform tumor
stem cell rescue in patients with pontine tumors, 10 of biopsies in DIPG patients, combined with advances in
whom had recurrent disease. Their study had two toxic molecular diagnostics, several therapeutically targetable,
deaths in the setting of septicemia and multiorgan failure. recurrent genomic alterations, and other molecular targets
The MST was 4.7 months from transplant in those with have been identified in DIPG. These targets include muta-
recurrent disease. Therefore, survival was not prolonged tions or amplifications of the PDGFRA and ACVR1 onco-
and adverse effects could be serious [66]. genes, and overexpression of EGFR, many of which are
being studied currently in clinical trials. One phase
I study evaluated the use of EGFR inhibitor erlotinib
NEWER THERAPIES (125 mg/m2) in the treatment of malignant brain tumors
in children, either as monotherapy or with RT. Group 1
Newer strategies for the treatment of DIPG are needed. One included refractory or relapsed brain tumors treated with
of the major limitations of treatment for DIPG is efficient erlotinib alone and Group 2 included newly diagnosed
delivery of effective therapies to the tumor, with penetration brainstem gliomas treated with erlotinib plus RT. In Group
of the blood–brain barrier a major obstacle. New strategies 2, OS was 12 months with only 6 grade 4–5 adverse events
for drug delivery currently being studied include intra- noted in both groups [72]. Another phase 2 study looked at
arterial/intra-thecal chemotherapy, intranasal drug deli- the use of the anti-EGFR antibody, nimotuzumab (150 mg/
very, and convection-enhanced catheter directed therapy. m2), in the treatment of progressive DIPG in pediatric
However, each has met with limited success. patients. This drug was administered IV weekly from weeks
One preclinical study looking at intranasal delivery 1 to 7 and once every 2 weeks from weeks 8 to 18. Overall
studied the use of the telomerase inhibitor GRN163 in rats 18 of 44 patients experienced serious adverse effects, the
with human tumor xenografts. It was noted that this agent majority associated with disease progression. Median PFS
was found to be present in the tumor cells with peak accu- was 1.7 months and median survival was 3.2 months. The
mulation 4 h after delivery. It also was found to prolong sur- authors concluded that nimotuzumab has modest activity
vival in rats from 35 days in the control group to 75.5 days in DIPG and that a small subset of patients appeared to
in the GRN163 treated group [67]. The use of convection- benefit from this treatment [73].
enhanced delivery (CED) has also been recently looked at A phase I trial of imatinib, a TKI of PDGF-R, in children
with a number of drugs. For instance, one pilot study eval- with newly diagnosed brainstem gliomas from the PBTC
uated two children with DIPG who were treated with CED established the maximum tolerated dose in those not
of topotecan, a topoisomerase inhibitor, via infusion receiving enzyme-inducing anticonvulsant drugs as
through bilateral catheters placed in the brainstem. The first 465 mg/m2 with a PFS at 6 months of 70%. However, there
patient was treated 210 days after diagnosis, after RT was was evidence of possible increased risk of intratumoral
completed and at the time of tumor recurrence. Post- hemorrhage, which needs to be further studied [74].
treatment MRIs showed reduction in tumor size and edema, Another biologic agent that has been the focus of several
but the patient died 49 days after treatment. The second studies in recurrent pediatric HGG given its clinical utility
patient was treated 24 days after diagnosis, prior to RT. in recurrent adult glioblastoma, is bevacizumab, a mono-
Posttreatment MRIs again showed a modest reduction in clonal antibody against VEGF-A. Narayana and colleagues
tumor size. This patient was subsequently treated with have reported on their single-institution experience with
RT but died 120 days after treatment. The authors con- bevacizumab in pediatric patients with recurrent or pro-
cluded that while survival was not prolonged, a moderate gressive HGG. They have reported that 12 patients were
treatment effect was noted, and that more studies were treated with bevacizumab 10 mg/kg every 2 weeks along
needed to determine the optimal flow rate for CED [68]. with irinotecan 125 mg/m2. Ten patients tolerated therapy
Souwedaine and colleagues have noted that brainstem without serious toxicity, two patients had partial responses
CED for the treatment of DIPG is appealing due to the while four had stable disease. The median PFS was
compact nature and poor treatment options for this tumor. 2.25 months and median OS was 6.25 months. They have
The safety of brainstem CED has been well established in noted that outcome in pediatric patients was inferior com-
preclinical trials [69,70], and more recently, in early stage pared with adult patients, likely due to biological differ-
clinical trials. A few clinical trials are currently underway ences between adult and pediatric tumors [75]. More
which are evaluating the use of CED in treating DIPG recently, the Pediatric Brain Tumor Consortium evaluated
with targeted macromolecules such as antibodies and the use of bevacizumab and irinotecan in children with
immunotoxins [71]. In a recent phase I trial CED is recurrent malignant gliomas and brainstem gliomas in a
used to deliver 8H9 conjugated to a therapeutic radio- phase II study. They noted no sustained responses and a
isotope directly into the brainstem tumor via a surgically 6-month PFS of 41.8% in malignant gliomas and 9.7% in
Chemotherapy of Brainstem Gliomas Chapter 45 593

brainstem gliomas. The combination was well tolerated, but associated with a dismal outcome. For DIPG, PFS is typi-
unfortunately had minimal efficacy [76]. cally 5–6 months and OS <1 year. RT remains the only
Histone deacetylase inhibitors have been more recently therapy proven to improve survival in patients with DIPG
of interest, especially in light of the molecular findings in despite numerous clinical trials and ongoing research.
DIPG, where many of these tumors harbor K27M muta- Higher doses of RT using HRT (66–78 Gy) do not appear
tions. One institution evaluated the use of valproic acid in to provide a survival advantage when compared with
the outcomes of children with DIPG. Their standard of standard dose and fractionated protocols. Different strat-
therapy included RT and weekly carboplatin and vin- egies using chemotherapy to treat DIPG, including high-
cristine. They have evaluated patients treated with this dose chemotherapy followed by stem cell rescue, neoad-
regimen without valproic acid versus with valproic acid, juvant chemotherapy (single or multiagent), concurrent che-
and found statistically significant improved median EFS motherapy with RT, and adjuvant chemotherapy, have not
(6.5 vs 9.5 months in treated patients) and OS (7.8 vs demonstrated any survival benefit when compared with
13.4 months in treated patients) [77]. Another histone dea- conventional RT alone. The blood-brain barrier and a lack
cetylase inhibitor which is currently being studied in of penetration of many therapeutic agents into brain tumor
clinical trial is panobinostat. A preclinical study of panobi- tissue have been recognized as major obstacles to therapy.
nostat using a genetically engineered mouse model showed Recent advances in treatment delivery include CED, which
that this drug was an effective targeted agent against DIPG is currently studying the use of targeted macromolecules
human and mouse tumor cells in vitro. It potentially such as antibodies and immunotoxins in treating DIPG. In
inhibited cell proliferation and viability, and induced apo- addition, promising novel immunotherapy approaches are
ptosis. In vivo studies in a panel of xenografts and geneti- rapidly emerging for the treatment of gliomas, and are
cally engineered mouse models showed that the drug hoped to mature over the next years, providing a much-
reached the brainstem tumor tissue and reduced prolifer- needed effective treatment option for children with DIPG.
ation of cells. However, it did not significantly impact
the survival in these mice, which the authors concluded
was due to toxicity with higher doses [78].
Immunotherapeutic approaches for DIPG are being REFERENCES
pursued in a variety of ongoing clinical trials. A pilot study
[1] Freeman CR, Farmer JP. Pediatric brain stem gliomas: a review. Int
investigated the use of a peptide vaccine against glioma- J Radiat Oncol Biol Phys 1998;40(2):265–71.
associated antigens (GAA) in 26 children with DIPG and [2] Guillamo JS, Doz F, Delattre JY. Brain stem gliomas. Curr Opin
HGGs. In this study, only 2 children had progressive Neurol 2001;14(6):711–5.
disease, 5 had symptomatic pseudoprogression that [3] Hargrave D, Chuang N, Bouffet E. Conventional MRI cannot predict
responded to dexamethasone, 19 had stable disease, 2 had survival in childhood diffuse intrinsic pontine glioma. J Neurooncol
partial responses, and 1 had a minor response. The vaccine 2008;86(3):313–9.
was well tolerated overall, and had an OS that was between [4] Zagzag D, Miller DC, Knopp E, Farmer JP, Lee M, Biria S, et al.
6.3 and >38 months and a positive immune response in Primitive neuroectodermal tumors of the brainstem: investigation
eight patients [79]. Currently there is an intense interest of seven cases. Pediatrics 2000;106(5):1045–53.
[5] Sturm D, Orr BA, Toprak UH, Hovestadt V, Jones DTW, Capper D,
in developing therapies using immune checkpoint inhib-
et al. New brain tumor entities emerge from molecular classification
itors, including an ongoing study by the PBTC using pem-
of CNS-PNETs. Cell 2016;164(5):1060–72.
brolizumab, an anti-PD-1 antibody, in children with [6] Albright AL, Guthkelch AN, Packer RJ, Price RA, Rourke LB. Prog-
recurrent HGG, including DIPG (ClinicalTrials.gov Iden- nostic factors in pediatric brain-stem gliomas. J Neurosurg 1986;
tifier: NCT02359565). Cell-based immunotherapeutic 65(6):751–5.
strategies, such as dendritic cell-based tumor vaccines, [7] Cohen MEM. Prognostic factors in brainstem gliomas. Neurology
are also being developed for children with malignant brain 1986;36(5):602.
tumors, including HGG [80]. Lastly, chimeric antigen [8] Cage TA, Samagh SP, Mueller S, Nicolaides T, Haas-Kogan D,
receptor (CAR) T-cell therapy is emerging for the treatment Prados M, et al. Feasibility, safety, and indications for surgical biopsy
of adult glioblastoma patients, and is likely to be explored in of intrinsic brainstem tumors in children. Childs Nerv Syst 2013;
future pediatric trials as well [81]. 29(8):1313–9.
[9] Freeman CR, Perilongo G. Chemotherapy for brain stem gliomas.
Childs Nerv Syst 1999;15(10):545–53.
[10] Jones DTW, Hutter B, J€ager N, Korshunov A, Kool M, Warnatz H-J,
CONCLUSIONS et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilo-
cytic astrocytoma. Nat Genet 2013;45(8):927–32.
Brainstem gliomas comprise a range of disease entities that [11] Jeuken JWM, Wesseling P. MAPK pathway activation through
can range from low-grade tumors which require little or no BRAF gene fusion in pilocytic astrocytomas; a novel oncogenic
treatment and have an excellent prognosis, to high-grade fusion gene with diagnostic, prognostic, and therapeutic potential.
tumors that are resistant to available therapies and J Pathol 2010;222(4):324–8.
594 SECTION V Chemotherapy for Specific Tumors—Pediatrics

[12] Horbinski C, Hamilton RL, Nikiforov Y, Pollack IF. Association of [29] Barrow J, Adamowicz-Brice M, Cartmill M, MacArthur D, Lowe J,
molecular alterations, including BRAF, with biology and outcome in Robson K, et al. Homozygous loss of ADAM3A revealed by genome-
pilocytic astrocytomas. Acta Neuropathol (Berl) 2010;119(5):641–9. wide analysis of pediatric high-grade glioma and diffuse intrinsic
[13] Horbinski C. To BRAF or not to BRAF: is that even a question pontine gliomas. Neuro Oncol 2011;13(2):212–22.
anymore? J Neuropathol Exp Neurol 2013;72(1):2–7. [30] Freeman CR, Krischer JP, Sanford RA, Cohen ME, Burger PC, del
[14] Teo C, Siu TL. Radical resection of focal brainstem gliomas: is it Carpio R, et al. Final results of a study of escalating doses of hyper-
worth doing? Childs Nerv Syst 2008;24(11):1307–14. fractionated radiotherapy in brain stem tumors in children: a Pediatric
[15] Li KW, Roonprapunt C, Lawson HC, Abbott IR, Wisoff J, Epstein F, Oncology Group study. Int J Radiat Oncol Biol Phys 1993;
et al. Endoscopic third ventriculostomy for hydrocephalus associated 27(2):197–206.
with tectal gliomas. Neurosurg Focus 2005;18(6A):E2. [31] Packer RJ, Boyett JM, Zimmerman RA, Albright AL, Kaplan AM,
[16] Packer RJ, Ater J, Allen J, Phillips P, Geyer R, Nicholson HS, et al. Rorke LB, et al. Outcome of children with brain stem gliomas after
Carboplatin and vincristine chemotherapy for children with newly treatment with 7800 cGy of hyperfractionated radiotherapy.
diagnosed progressive low-grade gliomas. J Neurosurg 1997; A Childrens Cancer Group Phase I/II Trial. Cancer 1994;74
86(5):747–54. (6):1827–34.
[17] Ater JL, Zhou T, Holmes E, Mazewski CM, Booth TN, Freyer DR, [32] Freeman CR, Kepner J, Kun LE, Sanford RA, Kadota R, Mandell L,
et al. Randomized study of two chemotherapy regimens for treatment et al. A detrimental effect of a combined chemotherapy-radiotherapy
of low-grade glioma in young children: a report from the Children’s approach in children with diffuse intrinsic brain stem gliomas? Int
Oncology Group. J Clin Oncol 2012;30(21):2641–7. J Radiat Oncol Biol Phys 2000;47(3):561–4.
[18] Perry JR, Brown MT, Gockerman JP. Acute leukemia following [33] Pai Panandiker AS, Wong JK, Nedelka MA, Wu S, Gajjar A,
treatment of malignant glioma. J Neurooncol 1998;40(1):39–46. Broniscer A. Effect of time from diagnosis to start of radiotherapy
[19] Karajannis MA, Lyden DC, Khakoo Y, Dunkel IJ. Treatment-related on children with diffuse intrinsic pontine glioma. Pediatr Blood
myelodysplastic syndrome after chemotherapy for childhood low- Cancer 2014;61(7):1180–3.
grade astrocytoma. J Pediatr Hematol Oncol 2006;28(10):700. [34] Edwards MS, Wara WM, Urtasun RC, Prados M, Levin VA,
[20] Lassaletta A, Scheinemann K, Zelcer SM, Hukin J, Wilson BA, Fulton D, et al. Hyperfractionated radiation therapy for brain-stem
Jabado N, et al. Phase II weekly vinblastine for chemotherapy-naı̈ve glioma: a phase I-II trial. J Neurosurg 1989;70(5):691–700.
children with progressive low-grade Glioma: a Canadian Pediatric [35] Packer RJ, Boyett JM, Zimmerman RA, Rorke LB, Kaplan AM,
Brain Tumor Consortium Study. J Clin Oncol 2017;34(29):3537–43. Albright AL, et al. Hyperfractionated radiation therapy (72 Gy) for
[21] Bouffet E, Jakacki R, Goldman S, Hargrave D, Hawkins C, Shroff M, children with brain stem gliomas. A Childrens Cancer Group Phase
et al. Phase II study of weekly vinblastine in recurrent or refractory I/II Trial. Cancer 1993;72(4):1414–21.
pediatric low-grade glioma. J Clin Oncol 2012;30(12):1358–63. [36] Zaghloul MS, Eldebawy E, Ahmed S, Mousa AG, Amin A, Refaat A,
[22] Gururangan S, Fangusaro J, Poussaint TY, McLendon RE, Onar- et al. Hypofractionated conformal radiotherapy for pediatric diffuse
Thomas A, Wu S, et al. Efficacy of bevacizumab plus irinotecan in intrinsic pontine glioma (DIPG): a randomized controlled trial.
children with recurrent low-grade gliomas—a Pediatric Brain Tumor Radiother Oncol 2014;111(1):35–40.
Consortium study. Neuro Oncol 2014;16(2):310–7. [37] Janssens GORJ, Gidding CEM, Van Lindert EJ, Oldenburger FR,
[23] Kieran MW, Yao X, Macy M, Leary S, Cohen K, MacDonald T, et al. Erasmus CE, Schouten-Meeteren AYN, et al. The role of hypofrac-
Final results of a prospective multi-institutional phase II study of tionation radiotherapy for diffuse intrinsic brainstem glioma
everolimus (RAD001), an mtor inhibitor, in pediatric patients with in children: a pilot study. Int J Radiat Oncol Biol Phys
recurrent or progressive low-grade glioma. A poetic consortium trial. 2009;73(3):722–6.
Neuro Oncol 2014;16(Suppl 3):iii27. [38] Fontanilla HP, Pinnix CC, Ketonen LM, Woo SY, Vats TS,
[24] Robison NJ, Campigotto F, Chi SN, Manley PE, Turner CD, Rytting ME, et al. Palliative reirradiation for progressive diffuse
Zimmerman MA, et al. A phase II trial of a multi-agent oral antian- intrinsic pontine glioma. Am J Clin Oncol 2012;35(1):51–7.
giogenic (metronomic) regimen in children with recurrent or pro- [39] Wolff JE, Rytting ME, Vats TS, Zage PE, Ater JL, Woo S, et al.
gressive cancer. Pediatr Blood Cancer 2014;61(4):636–42. Treatment of recurrent diffuse intrinsic pontine glioma: the MD
[25] Banerjee A, Jakacki RI, Onar-Thomas A, Wu S, Nicolaides T, Young Anderson Cancer Center experience. J Neurooncol 2012;
Poussaint T, et al. A phase I trial of the MEK inhibitor selumetinib 106(2):391–7.
(AZD6244) in pediatric patients with recurrent or refractory low- [40] Janssens GO, Gandola L, Bolle S, Mandeville H, Ramos-Albiac M,
grade glioma: a Pediatric Brain Tumor Consortium (PBTC) study. van Beek K, et al. Survival benefit for patients with diffuse intrinsic
Neuro Oncol 2017;19(8):1135–44. pontine glioma (DIPG) undergoing re-irradiation at first progression:
[26] Green AL, Kieran MW. Pediatric brainstem gliomas: new under- A matched-cohort analysis on behalf of the SIOP-E-HGG/DIPG
standing leads to potential new treatments for two very different working group. Eur J Cancer 2017;73:38–47.
tumors. Curr Oncol Rep 2015;17(3):436. [41] Jenkin RD, Boesel C, Ertel I, Evans A, Hittle R, Ortega J, et al. Brain-
[27] Pollack IF, Shultz B, Mulvihill JJ. The management of brainstem stem tumors in childhood: a prospective randomized trial of irradi-
gliomas in patients with neurofibromatosis 1. Neurology 1996; ation with and without adjuvant CCNU, VCR, and prednisone.
46(6):1652–60. A report of the Childrens Cancer Study Group. J Neurosurg 1987;
[28] Khuong-Quang D-A, Buczkowicz P, Rakopoulos P, Liu X-Y, 66(2):227–33.
Fontebasso AM, Bouffet E, et al. K27M mutation in histone H3.3 [42] Kadota RP, Mandell LR, Fontanesi J, Kovnar EH, Krischer J,
defines clinically and biologically distinct subgroups of pediatric Kun LE, et al. Hyperfractionated irradiation and concurrent cisplatin
diffuse intrinsic pontine gliomas. Acta Neuropathol (Berl) in brain stem tumors: a Pediatric Oncology Group pilot study (9139).
2012;124(3):439–47. Pediatr Neurosurg 1994;20(4):221–5.
Chemotherapy of Brainstem Gliomas Chapter 45 595

[43] Allen J, Siffert J, Donahue B, Nirenberg A, Jakacki R, Robertson P, and hyperfractionated irradiation for children with high-risk brain
et al. A phase I/II study of carboplatin combined with hyperfractio- tumors. Med Pediatr Oncol 1994;23(5):428–36.
nated radiotherapy for brainstem gliomas. Cancer 1999;86 [58] Bouffet E, Khelfaoui F, Philip I, Biron P, Brunat-Mentigny M,
(6):1064–9. Philip T. High-dose carmustine for high-grade gliomas in childhood.
[44] Broniscer A, Leite CC, Lanchote VL, Machado TM, Cristófani LM. Cancer Chemother Pharmacol 1997;39(4):376–9.
Radiation therapy and high-dose tamoxifen in the treatment of [59] Bouffet E, Raquin M, Doz F, Gentet JC, Rodary C, Demeocq F, et al.
patients with diffuse brainstem gliomas: results of a Brazilian coop- Radiotherapy followed by high dose busulfan and thiotepa: a pro-
erative study. Brainstem Glioma Cooperative Group. J Clin Oncol spective assessment of high dose chemotherapy in children with
2000;18(6):1246–53. diffuse pontine gliomas. Cancer 2000;88(3):685–92.
[45] Wolff JEA, Driever PH, Erdlenbruch B, Kortmann RD, Rutkowski S, [60] Broniscer A, Iacono L, Chintagumpala M, Fouladi M, Wallace D,
Pietsch T, et al. Intensive chemotherapy improves survival in pedi- Bowers DC, et al. Role of temozolomide after radiotherapy for newly
atric high-grade glioma after gross total resection: results of the diagnosed diffuse brainstem glioma in children: results of a multiin-
HIT-GBM-C protocol. Cancer 2010;116(3):705–12. stitutional study (SJHG-98). Cancer 2005;103(1):133–9.
[46] Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, [61] Allen JC, Helson L. High-dose cyclophosphamide chemotherapy for
Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant recurrent CNS tumors in children. J Neurosurg 1981;55(5):749–56.
temozolomide for glioblastoma. N Engl J Med 2005;352(10):987–96. [62] Gaynon PS, Ettinger LJ, Baum ES, Siegel SE, Krailo MD,
[47] Cohen KJ, Heideman RL, Zhou T, Holmes EJ, Lavey RS, Bouffet E, Hammond GD. Carboplatin in childhood brain tumors. A Children’s
et al. Temozolomide in the treatment of children with newly diag- Cancer Study Group Phase II trial. Cancer 1990;66(12):2465–9.
nosed diffuse intrinsic pontine gliomas: a report from the Children’s [63] Chamberlain MC. Recurrent brainstem gliomas treated with oral
Oncology Group. Neuro Oncol 2011;13(4):410–6. VP-16. J Neurooncol 1993;15(2):133–9.
[48] Bailey S, Howman A, Wheatley K, Wherton D, Boota N, Pizer B, [64] Rodriguez LA, Prados M, Fulton D, Edwards MS, Silver P, Levin V.
et al. Diffuse intrinsic pontine glioma treated with prolonged temozo- Treatment of recurrent brain stem gliomas and other central nervous
lomide and radiotherapy—results of a United Kingdom phase II trial system tumors with 5-fluorouracil, CCNU, hydroxyurea, and
(CNS 2007 04). Eur J Cancer 2013;49(18):3856–62. 6-mercaptopurine. Neurosurgery 1988;22(4):691–3.
[49] M€ uller K, Schlamann A, Guckenberger M, Warmuth-Metz M, [65] Kalifa C, Hartmann O, Demeocq F, Vassal G, Couanet D, Terrier-
Gl€uck A, Pietschmann S, et al. Craniospinal irradiation with con- Lacombe MJ, et al. High-dose busulfan and thiotepa with autologous
current temozolomide for primary metastatic pediatric high-grade bone marrow transplantation in childhood malignant brain tumors: a
or diffuse intrinsic pontine gliomas. A first report from the GPOH- phase II study. Bone Marrow Transplant 1992;9(4):227–33.
HIT-HGG Study Group. Strahlenther Onkol 2014;190(4):377–81. [66] Dunkel IJ, Garvin JH, Goldman S, Ettinger LJ, Kaplan AM, Cairo M,
[50] Hegi ME, Diserens A-C, Gorlia T, Hamou M-F, de Tribolet N, et al. High dose chemotherapy with autologous bone marrow rescue
Weller M, et al. MGMT gene silencing and benefit from temozo- for children with diffuse pontine brain stem tumors. Children’s
lomide in glioblastoma. N Engl J Med 2005;352(10):997–1003. Cancer Group. J Neurooncol 1998;37(1):67–73.
[51] Korshunov A, Ryzhova M, Hovestadt V, Bender S, Sturm D, [67] Hashizume R, Ozawa T, Gryaznov SM, Bollen AW, Lamborn KR,
Capper D, et al. Integrated analysis of pediatric glioblastoma reveals Frey WH, et al. New therapeutic approach for brain tumors: Intra-
a subset of biologically favorable tumors with associated molecular nasal delivery of telomerase inhibitor GRN163. Neuro Oncol
prognostic markers. Acta Neuropathol (Berl) 2015;129(5):669–78. 2008;10(2):112–20.
[52] Kretschmar CS, Tarbell NJ, Barnes PD, Krischer JP, Burger PC, [68] Anderson RCE, Kennedy B, Yanes CL, Garvin J, Needle M, Canoll P,
Kun L. Pre-irradiation chemotherapy and hyperfractionated radiation et al. Convection-enhanced delivery of topotecan into diffuse
therapy 66 Gy for children with brain stem tumors. A phase II study of intrinsic brainstem tumors in children. J Neurosurg Pediatr 2013;11
the Pediatric Oncology Group, Protocol 8833. Cancer 1993; (3):289–95.
72(4):1404–13. [69] Sewing ACP, Caretti V, Lagerweij T, Schellen P, Jansen MHA, van
[53] Jennings MT, Sposto R, Boyett JM, Vezina LG, Holmes E, Vuurden DG, et al. Convection enhanced delivery of carmustine to
Berger MS, et al. Preradiation chemotherapy in primary high-risk the murine brainstem: a feasibility study. J Neurosci Methods
brainstem tumors: phase II study CCG-9941 of the Children’s Cancer 2014;238:88–94.
Group. J Clin Oncol 2002;20(16):3431–7. [70] Zhou Z, Ho SL, Singh R, Pisapia DJ, Souweidane MM. Toxicity eval-
[54] Duffner PK, Horowitz ME, Krischer JP, Friedman HS, Burger PC, uation of convection-enhanced delivery of small-molecule kinase
Cohen ME, et al. Postoperative chemotherapy and delayed radiation inhibitors in naı̈ve mouse brainstem. Childs Nerv Syst 2015;31
in children less than three years of age with malignant brain tumors. (4):557–62.
N Engl J Med 1993;328(24):1725–31. [71] Zhou Z, Singh R, Souweidane MM. Convection-enhanced delivery
[55] Frappaz D, Schell M, Thiesse P, Marec-Berard P, Mottolese C, for diffuse intrinsic pontine glioma treatment. Curr Neuropharmacol
Perol D, et al. Preradiation chemotherapy may improve survival in 2017;15(1):116–28.
pediatric diffuse intrinsic brainstem gliomas: final results of BSG [72] Geoerger B, Hargrave D, Thomas F, Ndiaye A, Frappaz D,
98 prospective trial. Neuro Oncol 2008;10(4):599–607. Andreiuolo F, et al. Innovative therapies for children with cancer
[56] Wolff JE, Kortmann R-D, Wolff B, Pietsch T, Peters O, Schmid H-J, pediatric phase I study of erlotinib in brainstem glioma and
et al. High dose methotrexate for pediatric high grade glioma: relapsing/refractory brain tumors. Neuro Oncol 2011;13(1):109–18.
results of the HIT-GBM-D pilot study. J Neurooncol 2011;102 [73] Bartels U, Wolff J, Gore L, Dunkel I, Gilheeney S, Allen J, et al.
(3):433–42. Phase 2 study of safety and efficacy of nimotuzumab in pediatric
[57] Kedar A, Maria BL, Graham-Pole J, Ringdahl DM, Quisling RG, patients with progressive diffuse intrinsic pontine glioma. Neuro
Mickle JP, et al. High-dose chemotherapy with marrow reinfusion Oncol 2014;16(11):1554–9.
596 SECTION V Chemotherapy for Specific Tumors—Pediatrics

[74] Pollack IF, Jakacki RI, Blaney SM, Hancock ML, Kieran MW, [78] Hennika T, Hu G, Olaciregui NG, Barton KL, Ehteda A,
Phillips P, et al. Phase I trial of imatinib in children with newly diag- Chitranjan A, et al. Pre-clinical study of panobinostat in xenograft
nosed brainstem and recurrent malignant gliomas: a Pediatric Brain and genetically engineered murine diffuse intrinsic pontine glioma
Tumor Consortium report. Neuro Oncol 2007;9(2):145–60. models. PLoS ONE 2017;12(1).
[75] Narayana A, Kunnakkat S, Chacko-Mathew J, Gardner S, [79] Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A,
Karajannis M, Raza S, et al. Bevacizumab in recurrent high-grade Potter DM, et al. Antigen-specific immune responses and clinical
pediatric gliomas. Neuro Oncol 2010;12(9):985–90. outcome after vaccination with glioma-associated antigen peptides
[76] Gururangan S, Chi SN, Young Poussaint T, Onar-Thomas A, and polyinosinic-polycytidylic acid stabilized by lysine and carboxy-
Gilbertson RJ, Vajapeyam S, et al. Lack of efficacy of bevacizumab methylcellulose in children with newly diagnosed malignant brainstem
plus irinotecan in children with recurrent malignant glioma and and nonbrainstem gliomas. J Clin Oncol 2014;32(19):2050–8.
diffuse brainstem glioma: a Pediatric Brain Tumor Consortium study. [80] Ardon H, De Vleeschouwer S, Van Calenbergh F, Claes L,
J Clin Oncol 2010;28(18):3069–75. Kramm CM, Rutkowski S, et al. Adjuvant dendritic cell-based
[77] Felix FHC, de Araujo OL, da Trindade KM, Trompieri NM, tumour vaccination for children with malignant brain tumours.
Fontenele JB. Retrospective evaluation of the outcomes of children Pediatr Blood Cancer 2010;54(4):519–25.
with diffuse intrinsic pontine glioma treated with radiochemotherapy [81] Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A,
and valproic acid in a single center. J Neurooncol 2014;116 et al. Regression of glioblastoma after chimeric antigen receptor
(2):261–6. T-Cell therapy. N Engl J Med 2016;375(26):2561–9.

You might also like