Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Oct 1.
Published in final edited form as: Childs Nerv Syst. 2009 Feb 11;25(10):1275–1282. doi: 10.1007/s00381-009-0809-7

New Chemotherapy Strategies and Biological Agents in the Treatment of Childhood Ependymoma

Karen D Wright 1,2,3, Amar Gajjar 1,2
PMCID: PMC2738756  NIHMSID: NIHMS85887  PMID: 19212772

Introduction

As the third most frequent brain tumor in children, ependymomas comprise approximately 9% of primary CNS tumors in patients less than 18 years of age and remain difficult to treat due to the high frequency of chemotherapy and radiation resistance [1]. The mean age at diagnosis ranges from 51–71 months [25] and 25–40% are diagnosed in children less than 3 years of age [6]. Children tend to have a worse outcome than adults; 40–60% of children will die of their disease, further highlighting the need for novel treatment strategies [1, 7, 8]. Recurrences, which are not uncommon, typically occur locally with a median time to recurrence of 13–25 months; 20% of failures have an isolated distant recurrence [24, 810]. Dissemination at the time of diagnosis is generally a rare event in patients with ependymoma, and at present, prognosis remains relatively poor for those patients despite multimodality treatment strategies [11].

Treatment Standards

The standard of care for ependymoma remains surgical resection followed by post-operative radiation therapy (RT) directed at the primary site, resulting in a 5 year progression-free survival (PFS) ranging from 50–60% [4, 9, 1214]. For very young children (age < 3 years), immediate post-op irradiation is not widely accepted due to the associated neurocognitive sequelae, and multi-agent chemotherapy has been given in an effort to delay or avoid irradiation. In addition, there is no consensus in regard to “standard of care” for those patients with metastatic disease at the time of diagnosis.

To date, there has been no convincing role for chemotherapy in the treatment of ependymoma using conventional agents. In single agent phase II trials, only cisplatin has been reported to be particularly active in ependymoma with a response rate of 30% (15–17). Carboplatin [18, 19], ifosfamide [20] and etoposide [21, 22] have demonstrated only modest activity. In addition, temozolomide, a prodrug with little, if any, pharmacologic activity until it is hydrolyzed to the active alkylating metabolite, has proved disappointing in ependymoma. No responses were observed in patients enrolled on the recent phase II study of temozolomide in children and adolescents with ependymoma under the direction of the Children’s Oncology Group [23].

Generally the results of trials using adjuvant combination chemotherapy post-irradiation in patients with newly diagnosed ependymoma have not shown improvement in survival rates compared to those patients treated with radiation therapy alone. A study conducted by St. Jude Children’s Research Hospital evaluated the use of conformal radiation only to the postoperative tumor bed in 88 children with ependymoma including 48 children under age 3 at the time of RT. Notably 3 year PFS was approximately 75% without the use of chemotherapy [14].

Role of Chemotherapy in Infants and Young Children with Ependymoma

The results of major clinical trials evaluating the role of chemotherapy conducted among infants and young children with ependymoma are summarized in Table 1. In a study by the Children’s Cancer Group (CCG 921), children over 2 years of age with newly diagnosed ependymomas were treated with CSI and randomized to receive adjuvant chemotherapy with either CCNU, vincristine and prednisone or the ”8 in 1” regimen [8, 12]. No difference in survival was noted between the two regimens, and therefore no advantage gained in using chemotherapy compared to historical controls. Similarly, Timmerman et al. [24] reported no benefit to the use of adjuvant chemotherapy in 55 patients with newly diagnosed anaplastic ependymoma treated on the HIT 88/89 and 91 trials with chemotherapy before (n=40) or after irradiation (n=15).

Table 1.

Results of Prospective Clinical Trials for Infants and Young Children with Ependymoma

Event Free Survival Overall Survival References

Trial N Age (mo) 1 year 5 years (unless otherwise specified) 5 years (unless otherwise specified)


CCG 8-in-1 15 22 (3) 8

HIT 88/89/91 55 75.6 (3) 24
GTR 28
STR 27

Baby POG 8633 48 <36 25 40.5 25
GTR 23 66
8 0–24 87 25.7 37.5
8 24–36 94 63.3 87.5
STR 25 25

SFOP 73 22 (4) 59 (4) 26
GTR 74 (4)
STR 35 (4)

VETOPEC 14 36 (3) 27

CCG 9921 74 72 32 59 12
M0 with min residual 42 79 33 67
20 75 29 54
M0, other 12 42 33 40
M1+

MD Anderson 5 40 (>5) 28

Head Start I/II 29 12 38 29
GTR 18 42
STR 11 31

The best response rates to combination chemotherapy in primary ependymoma have been demonstrated in younger children who received pre-irradiation chemotherapy in an attempt to delay RT. In the POG 8633 infant study for patients less than 3 years of age, Duffner et al. reported a 48% response rate following two cycles of combination chemotherapy that consisted of vincristine, cyclophosphamide, cisplatin and etoposide in 25 children with residual tumor after initial surgery, demonstrating that the use of post-operative chemotherapy may allow the delay of RT for a clinically relevant period of time in younger children with ependymoma [25]. Similarly, in a recent study by the French Society of Pediatric Oncology (SFOP), children with ependymoma under the age of 5 were treated with 7 cycles of alternating courses of procarbazine and carboplatin, etoposide and cisplatin, vincristine and cyclophosphamide over 1.5 years [26]. Despite the lack of any partial (PR) or complete responses (CR) in patients with residual disease post-initial surgery, 23% of patients remained alive at 4 years without the use of RT, suggesting that there exists a small subset of patients for whom cure may be possible with surgery and post-operative chemotherapy alone.

A number of other studies have revealed mixed results. White et al. found an 86% response rate to 4 cycles of vincristine, etoposide and cytoxan in 7 children < 4 years of age with newly diagnosed ependymoma [27]. The CCG 9921 trial utilized delayed RT achieving similar results to POG 8633 with a 5 year OS of 67% for patients with completely resected M0 disease; however, 5 year PFS for this group was only about 33% [12]. Among 25 children ages 2 weeks to 15 years enrolled on study at MD Anderson Cancer Center, 5 patients were less than 3 years of age and the five year PFS 40% [28]. The Head Start I/II trial, which used five cycles of induction chemotherapy, including IV methotrexate followed by a consolation regimen of myeloablative chemotherapy with autologous stem cell rescue, did not improve outcome for patients with ependymoma [29].

Role of Chemotherapy in Older Children with Ependymoma

Few phase III studies have demonstrated any benefit from adjuvant chemotherapy among older children (see Table 2) [5, 6, 30, 3234]. The CCG 9942 study for patients age 3–21 years of age with histologically proven intracranial ependymoma and evidence of residual tumor on postoperative imaging were nonrandomly assigned to receive pre-irradiation chemotherapy consisting of vincristine, cisplatin, etoposide and cyclophosphamide [30]. Needle and colleagues also reported encouraging results in 19 patients between 3 and 14 years of age treated with post-operative RT and chemotherapy consisting of carboplatin and vincristine alternating with ifosfamide and etoposide for a total of 4 cycles [31]. The 5-year actuarial progression free survival (PFS) of 74% for patients with post-operative residual ependymoma was higher than previously reported for RT alone, suggesting a possible role for multi-alkylator chemotherapy in incompletely resected ependymoma; however, this data has not been confirmed in a large prospective randomized trial. By comparison, Shu and colleagues retrospectively reviewed a cohort of 49 patients with ependymoma, which included the cohort from Needle’s work. Those patients underwent surgery, radiation and a variety of chemotherapeutic regimens with 5 year OS and PFS rates of 66.2 and 40.7%, respectively (32). In contrast, the CCG 942 study, which included children up to 16 years of age, did not show improved outcomes among older patients treated with postoperative craniospinal radiation and then randomized to receive adjuvant chemotherapy consisting of lomustine (CCNU), vincristine and prednisone for one year or observation [6]. The failure free survival and overall survival for the entire group at 10 years was 36 and 39%, respectively, suggesting no improvement in outcome with the use of adjuvant chemotherapy.

Table 2.

Results of Prospective Clinical Trials in Older Children with Ependymoma

Trial N 5 Year Event Free Survival (unless otherwise specified) 5 Year Overall Survival (unless otherwise specified) References

CCG 942 36 36 (10) 39 (10) 6

Hospital for Sick Children 35 45 33
GTR 9 87
STR 26 30

CHOP 45 36 5

MD Anderson 25 47 34
GTR 5 61
STR 20 37

CCG 9942 84 75 30
<5 yr 23 58(3)
5–9 yr 34 56(3)
≥10 yr 27 72(3)

CHOP 49 40.7 66.20
GTR 30
  M0 & RT > 54Gy 60.6 83.1 32
STR/biopsy 18
Unknown 1

Various studies among older children have focused primarily on outcomes following extent of tumor resection at surgery. Among 35 children up to the age of 16 years with infratentorial ependymoma at Toronto’s Hospital for Sick children, OS was 87% (n=9) for patients with gross total resection (GTR) compared to only 30% in those patients undergoing subtotal resection (STR) [33]. Similarly, at the Children’s Hospital of Philadelphia (CHOP), 5 year PFS among patients up to the age of 20 years with intracranial ependymoma was 36% overall, but 60% for patients with GTR (n=23) versus 21% with STR or biopsy [5]. Similarly, for those children involved in the MD Anderson Cancer Center trial, 5 year progression free survival was 61% and 37% in those who had complete resection of their tumor versus those with incomplete resection, respectively [34]. Collectively these data conclusively demonstrate that obtaining gross total resection is an important factor in achieving long term disease control.

Role of Chemotherapy in Tumor Resectability

In contrast, the potential importance of adjuvant chemotherapy in facilitating tumor resectability has been reported by Foreman et al. [35]. Adjuvant chemotherapy used between initial and second surgery in 4 patients with ependymoma was associated with a subsequent complete resection in 3 of the patients who remain free of progressive disease 23–34 months after second look surgery. One additional patient included in the study underwent second look surgery, but did not receive adjuvant chemotherapy. This strategy is being further studied in the current COG trial as no study to date has shown that either intensity of chemotherapy or response to chemotherapy correlates with ease of resection at the time of second surgery. The mechanism by which chemotherapy may make a tumor easier to resect is unknown, but could reflect a combination of cytotoxic and anti-angiogenic effects.

Metronomic Drug Dosing

At present, one of current areas of interest focuses on the use of ”metronomic“ dosing which relies on frequent administration of drug at low doses. An increasing number of preclinical trials support the use of metronomic dosing in order to inhibit angiogenesis [3639]. Browder et al. first demonstrated that administration of cyclophosphamide at both standard maximum tolerated doses (MTD) and frequent low doses induced tumor cell apoptosis in an experimental model [40]; however, the continuous low dose regimen also produced a more prolonged anti-angiogenic effect. Subsequent preclinical and early clinical studies have demonstrated activity of metronomic chemotherapy against various adult and pediatric tumors [418].

New Chemotherapy and Biologic Agents

With the limited role of chemotherapy and inherent side effects of radiation on the developing brain, considerable effort has been placed on the identification of the molecular changes underlying the development of ependymoma in hope of discovering novel therapeutic agents. To achieve that goal, a number of new agents possessing more tumor specific activity than standard cytotoxic agents are under development in addition to a variety of non-specific tumor directed agents. Among them are molecularly targeted agents including small molecule tyrosine kinase inhibitors [49] and antiangiogenic agents [50]. The use of these molecularly targeted therapies, however, presents added challenges in the pediatric population, in that the cell signaling pathways dysregulated in tumorigenesis are often those that are crucial for normal development.

Rationale for use of Gefitinib and Erlotinib

The receptor tyrosine kinase family of transmembrane receptors known as ERBB is involved in a number of cell signaling pathways that control various processes ranging from apoptosis to cell proliferation. Abnormalities of ERBB receptors have been noted in a variety of tumor types as well as ependymoma, specifically epidermal growth factor receptor (EGFR or ERBB1), ERBB2 and ERBB4 [5157]. Preclinical data from a variety of human cell lines and xenografts have revealed G1 cell cycle arrest and apoptosis when subjected to tyrosine kinase inhibitors [5859]. Synergistic effects have also been observed in preclinical studies when these small molecular inhibitors were combined with RT or chemotherapy in colon, head and neck and non-small cell lung cancers [60]. Thus, targeting ERBB receptors may prove to be an effective novel therapeutic strategy in patients with ependymoma.

With that in mind, Geoerger and colleagues evaluated gefitinib, a tyrosine kinase inhibitor of transmembrane cell surface receptors, including EFGR, found on both normal and cancer cells, for its antitumor activity and potential to radio-sensitize in vivo xenograft models [61, 62]. The exact mechanism of gefitinib’s antineoplastic action is unknown, but inhibition of the EGFR pathway is thought to prevent tumor cells from escaping the action of DNA damaging agents. In addition, ionizing radiation can activate EGFR tyrosine phosphorylation, leading to proliferation of surviving cancer cells and possible accelerated cellular repopulation after RT [61]. Thus, adjuvant therapy with gefitinib may block this major cytoprotective response following RT [61]. In fact, combined treatment of Geoerger’s xenograft models with RT and gefitinib revealed a trend toward improved anti-tumor activity [61].

One of the most advanced agents in clinical development is erlotinib, an orally available small molecule tyrosine kinase inhibitor of ERBB1 and ERBB2 with good CSF penetration and possibly anti-angiogenic activity in ERBB2 expressing tumors based on its down regulation of VEGF expression both in vivo and in vitro [6264]. Specifically, when orthotopic xenografts of ERBB2-transfected Daoy cells were used as a model for aggressive medulloblastoma, erlotinib treatment depleted the tumor vasculature, destroyed the self renewing tumor cell population and inhibited tumor growth [63]. Clinically, erlotinib has been well tolerated in a several pediatric phase I studies to date [6567].

Rationale for use of Bevacizumab

As alluded to above, considerable effort has been focused on identifying the mechanism by which tumors induce angiogenesis in order to target them for therapeutic intervention [50, 6870]. Development of effective anti-angiogenic treatments, however, remains in its early stages due to the inherent complexity of this process [6873]. To date, the most studied of these agents has been bevacizumab, a humanized monoclonal antibody to VEGF [50]. When orthotopic xenografts of ERBB2-transfected Daoy cells were used as a model for medulloblastoma as mentioned above, treatment with bevacizumab also affected tumor vasculature and inhibited tumor growth (64). Similar studies by Bao and colleagues also demonstrated that bevacizumab inhibited the growth of tumors derived from stem cell like glioma cells that secrete VEGF in a xenograft model (74).

As a single agent therapy in clinical trials, however, bevacizumab has been shown to be effective only in metastatic renal cell carcinoma in terms of progression free survival [75, 76]. Combining bevacizumab with cytotoxic chemotherapy, however, has improved its effectiveness in terms of progression free and overall survival as evidenced by a number of Phase III trials [77, 78]. In regard to CNS tumors, bevacizumab has been shown to be effective in adult trials for recurrent high grade glioma [79]. While the prior studies of bevacizumab suggest supporting evidence for the possibility of anti-angiogenic therapy, its effectiveness in ependymoma has yet to be documented.

On the Horizon

Other novel agents include receptor tyrosine kinase inhibitors which interfere with signaling pathways involved in tumor angiogenesis. A number of agents in this group are under development, including sunitinib and sorafinib, which have already been approved by the FDA as single agent therapy for renal cell carcinoma [75]. Sunitinib possesses both antitumor and antiangiogenic effects by inhibiting multiple receptor tyrosine kinases including platelet derived growth factors (PDGFRa, PDGFRb), vascular endothelial growth factors (VEGFR 1-3), FMS-like tyrosine kinase-3 (FLT-3), colony stimulating factor type I (CSF-1R) and glia cell line-derived neurotrophic factor receptor (RET). Similarly, sorafenib, another multikinase inhibitor, affects tumor growth and angiogenesis by inhibiting intracellular RAF kinases and cell surface kinase receptors (VEFGR 2-3, PDGFRb, cKIT, FLT-3). Again, none of these agents have been studied extensively in young children.

Despite advances in the diagnosis and etiology of ependymoma, the role of chemotherapy remains uncertain and the development of new treatment strategies presents a challenge for this disease dependent upon surgery and radiation, yet plagued by the associated physical and cognitive disabilities that may follow those interventions, especially in very young children. The development of novel therapies for patients with ependymoma will depend on a more detailed understanding of the molecular basis associated with the pathogenesis of this disease. Additionally, although molecular profiling of ependymoma is in its infancy, identification of distinct stem cell precursors in ependymoma [62] and emerging data in regard to different tumor subtypes [62, 8083] may facilitate the development of individually-tailored therapies based on cell signaling pathways.

Acknowledgments

Supported in part by the National Cancer Institute (grant CA 21765), Musicians Against Childhood Cancer (MACC), The Noyes Brain Tumor Foundation, The Ryan McGhee Foundation, and by the American Lebanese Syrian Associated Charities (ALSAC).

References

  • 1.Strother DR, Pollack IF, Fisher PG, Hunter JV, Woo SY, Pomeroy SL, Rorke LB. Tumors of the Central Nervous System. In: Pizzo P, Poplack D, editors. Priniciples and Practice of Pediatric Oncology. 4th edn. New York: Lippincott Williams & Wilkins; 2003. pp. 778–785. [Google Scholar]
  • 2.Foreman NK, Love S, Thorne R. Intracranial ependymomas: Analysis of prognostic factors in a population-based series. Pediatr Neurosurg. 1996;24:119–125. doi: 10.1159/000121027. [DOI] [PubMed] [Google Scholar]
  • 3.Perilongo G, Massimino M, Sotti G, Belfontail T, Masiero L, Rigobello L, Garre L, Carli M, Lomabardi F, Solero C, Sainati L, Canale V, del Prever AB, Giangaspero F, Andreussi L, Mazza C, Madon E. Analyses of prognostic factors in a retrospective review of 92 children with ependymoma: Italian Pediatric Neuro-Oncology Group. Med Pediatr Oncol. 1997;29:79–85. doi: 10.1002/(sici)1096-911x(199708)29:2<79::aid-mpo3>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 4.Horn B, Heideman R, Geyer R, Pollack I, Packer R, Goldwein J, Tomita T, Schomberg Ater J, Luchtman-Jones L, Rivlin K, Lamborn K, Pradose M, Bollen A, Berger M, Dahl McNeil E, Patterson K, Shaw D, Kubalik M, Russo C. A multi-institutional retrospective study of intracranial ependymoma in children: identification of risk factors. J Pediatr Hematol Oncol. 1999;21:203–211. doi: 10.1097/00043426-199905000-00008. [DOI] [PubMed] [Google Scholar]
  • 5.Sutton LN, Goldwein J, Perilongo G, Lang B, Schut L, Rorke L, Packer R. Prognostic factors in childhood ependymomas. Pediatr Neurosurg. 1990;16:57–65. doi: 10.1159/000120509. [DOI] [PubMed] [Google Scholar]
  • 6.Evans AE, Anderson JR, Lefkowitz-Boudreaux IB, Finlay JL. Adjuvant chemotherapy of childhood posterior fossa ependymoma: craniospinal irradiation with or without adjuvant CCNU, vincristine, and prednisone: a Childrens Cancer Group study. Med Pediatr Oncol. 1996;27:8–14. doi: 10.1002/(SICI)1096-911X(199607)27:1<8::AID-MPO3>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 7.Mclendon RE, Enterline DS, Tien RD, Thorstad WL, Bruner JM. Tumors of central epithelial origin. In: Bigner DD, McLendon RE, Bruner JM, editors. Russel and Rubinstein’s Pathology of Tumors of the Nervous System. London: Arnold; 1988. pp. 387–418. [Google Scholar]
  • 8.Robertson PL, Zeltzer PM, Boyett JM, Rorke LB, Allen JC, Geyer JR, Stanley P, Li H, Albright AL, McGuire-Cullen P, Finlay JL, Stevens KR, Jr, Milstein JM, Packer RJ, Wisoff J. Survival and prognostic factors following radiation therapy and chemotherapy for ependymomas in children: a report of the Children’s Cancer Group. J Neurosurg. 1998;88:695–703. doi: 10.3171/jns.1998.88.4.0695. [DOI] [PubMed] [Google Scholar]
  • 9.Pollack IF, Gerszten PC, Martinez AJ, Lo KH, Shultz B, Albright AL, Janosky J, Deutsch M. Intracranial ependymoma of childhood: longterm outcome and prognostic factors. Neurosurgery. 1995;37:655–667. doi: 10.1227/00006123-199510000-00008. [DOI] [PubMed] [Google Scholar]
  • 10.Needle MN, Goldwein JW, Grass J, Cnaan A, Bergman I, Molloy P, Sutton L, Zhao H, Garvin JK, Jr, Phillips PC. Adjuvant chemotherapy for the treatment of intracranial ependymoma of childhood. Cancer. 1997;80:341–347. [PubMed] [Google Scholar]
  • 11.Zacharoulis S, Lingyun Ji, Pollack IF, Duffner P, Geyer R, Grill J, Schild S, Jaing TH, Massimino M, Finlay J, Sposto R. Metastatic Ependymoma: A multi-institutional retrospective analysis of prognostic factors. Pediatr Blood Cancer. 2008;50:231–235. doi: 10.1002/pbc.21276. [DOI] [PubMed] [Google Scholar]
  • 12.Geyer JR, Zeltzer PM, Boyett JM, Rorke LB, Stanley P, Albright AL, Wisoff JH, Milstein JM, Allen JC, Finlay Survival of infants with primitive neuroectodermal tumors or malignant ependymomas of the CNS treated with eight drugs in 1 day: a report from the Childrens Cancer Group. J Clin Oncol. 1994;12:1607–1615. doi: 10.1200/JCO.1994.12.8.1607. [DOI] [PubMed] [Google Scholar]
  • 13.Oya N, Shibamoto Y, Nagata Y, Negoro Y, Hiraoka Postoperative radiotherapy for intracranial ependymoma: analysis of prognostic factors and patterns of failure. J Neurooncol. 2002;56:87–94. doi: 10.1023/a:1014442106111. [DOI] [PubMed] [Google Scholar]
  • 14.Merchant TE, Mulhern RK, Krasin MJ, Kun LE, Williams T, Li C, Xiong X, Khan RB, Lustig RH, Boop FA, Sanford RA. Preliminary results from a phase II trial of conformal radiaition therapy and evaluation of radiation-related CNS effects of pediatric patients with localized ependymoma. J Clin Oncol. 2004;22:3156–3162. doi: 10.1200/JCO.2004.11.142. [DOI] [PubMed] [Google Scholar]
  • 15.Khan AB, D’Souza BJ, Wharam MD, Champion LA, Sinks LF, Woo SY, McCullough DC, Leventhal BG. Cisplatin therapy in recurrent childhood brain tumors. Cancer Treat Rep. 1982;66:2013–2020. [PubMed] [Google Scholar]
  • 16.Sexauer CL, Khan A, Burger PC, Krischer JP, van Eys J, Vats T, Ragab AH A Pediatric Oncology Group Study. Cisplatin in recurrent pediatric brain tumors. A POG Phase II study. Cancer. 1985;56:1497–1501. doi: 10.1002/1097-0142(19851001)56:7<1497::aid-cncr2820560703>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
  • 17.Walker RW, Allen JC. Cisplatin in the treatment of recurrent childhood primary brian tumors. J Clin Oncol. 1988;6:62–66. doi: 10.1200/JCO.1988.6.1.62. [DOI] [PubMed] [Google Scholar]
  • 18.Gaynon PS, Ettinger LJ, Baum ES, Siegel SE, Krailo MD, Hammond GD. Carboplatin in childhood brain tumors. A children’s cancer study group phase II trial. Cancer. 1990;66:2465–2469. doi: 10.1002/1097-0142(19901215)66:12<2465::aid-cncr2820661204>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 19.Friedman HS, Krischer JP, Burger P, Oakes WJ, Hockenberger B, Weiner MD, Falletta JM, Norris D, Ragab AH, Mahoney DH., Jr Treatment of children with progressive or recurrent brain tumors with carboplatin or iproplatin: a pediatric oncology group randomized phase II study. J Clin Oncol. 1992;10:249–256. doi: 10.1200/JCO.1992.10.2.249. [DOI] [PubMed] [Google Scholar]
  • 20.Chastagner P, Sommelet-Olive D, Kalifa C, Brunat-Mentigny M, Zucker JM, Demeocq F, Baranzelli MC, Tron P, Bergeron C, Pein F. Phase II study of ifosfamide in childhood brain tumors: a report by the French Society of Pediatric Oncology (SFOP) Med Pediatr Oncol. 1993;21:29–53. doi: 10.1002/mpo.2950210110. [DOI] [PubMed] [Google Scholar]
  • 21.Needle MN, Molloy PT, Geyer JR, Herman-Liu A, Belasco JB, Goldwein JW, Sutton L, Phillips PC. Phase II study of daily oral etoposide in children with recurrent brain tumors and other solid tumors. Med Pediatr Oncol. 1997;29:28–32. doi: 10.1002/(sici)1096-911x(199707)29:1<28::aid-mpo5>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
  • 22.Sandri A, Massimino M, Mastrodicasa L, Sardi N, Bertin D, Basso ME, Todisco L, Paglino A, Perilongo G, Genitori L, Valentini L, Ricardi U, Gandola L, Giangaspero F, Madon E. Treatment with oral etoposide for childhood recurrent ependymomas. J Pediatr Hem Oncol. 2005;27:286–290. doi: 10.1097/01.mph.0000181430.71176.b7. [DOI] [PubMed] [Google Scholar]
  • 23.Nicholson HS, Kretschmar CS, Krailo M, Bernstein M, Kadota R, Fort D, Friedman H, Harris MB, Tedeschi-Blok N, Mazewski C, Sato J, Reaman GGH. Phase 2 study of temozolomide in children and adolescents with recurrent central nervous system tumors: a report from the Children’s Oncology Group. Cancer. 2007;110:1542–1550. doi: 10.1002/cncr.22961. [DOI] [PubMed] [Google Scholar]
  • 24.Timmermann B, Kortmann RD, Kuhl J, Meisner C, Slavc I, Pietsch T, Bamberg M. Combined postoperative irradiation and chemotherapy for anaplastic ependymoma in childhood: results of the German prospective trials HIT 88/89 and HIT 91. Int J Radiat Oncol Biol Phys. 2000;46:287–295. doi: 10.1016/s0360-3016(99)00414-9. [DOI] [PubMed] [Google Scholar]
  • 25.Duffner PK, Horowitz ME, Krischer JP, Friedman HS, Burger PC, Cohen ME, Sanford RA, Mulhern RK, James HE, Freeman CR. Postoperative chemotherapy and delayed radiation in children less than three years of age with malignant brain tumors. N Engl J Med. 1993;328:1725–1731. doi: 10.1056/NEJM199306173282401. [DOI] [PubMed] [Google Scholar]
  • 26.Grill J, Le Deley MC, Gambarelli D, Raquin MA, Couanet D, Pierre-Kahn A, Habrand JL, Doz F, Frappaz D, Gentet JC, Edan C, Chastagner P, Kalifa C, French Society of Pediatric Oncology Postoperative chemotherapy without irradiation for ependymoma in children under 5 years of age: a multicenter trial of the French Society of Pediatric Oncology. J Clin Oncol. 2001;19:1288–1296. doi: 10.1200/JCO.2001.19.5.1288. [DOI] [PubMed] [Google Scholar]
  • 27.White L, Kellie S, Gray E, Toogood I, Waters K, Lockwood L, Macfarlane S, Johnston H. Postoperative chemotherapy in children less than 4 years of age with malignant brain tumors: promising initial response to a VETOPEC-based regimen. J Pediatr Hematology/Oncology. 1998;20:125–130. doi: 10.1097/00043426-199803000-00007. [DOI] [PubMed] [Google Scholar]
  • 28.Ater JL, van Eys J, Woo SY, Moore B, 3rd, Copeland DR, Bruner J. MOPP chemotherapy without irradiation as primary postsurgical therapy for brain tumors in infants and young children. J Neurooncol. 1997;32:243–252. doi: 10.1023/a:1005744527443. [DOI] [PubMed] [Google Scholar]
  • 29.Zacharoulis S, Levy A, Chi SN, Gardner S, Rosenblum M, Miller DC, Dunkel I, Diez B, Sposto R, Ji L, Asgharzadeh S, Hukin J, Belasco J, Dubowy R, Kellie S, Termuhlen A, Finlay J. Outcome for young children newly diagnosed with ependymoma, treated with intensive induction chemotherapy followed by myeloablative chemotherapy and autologous stem cell rescue. Pediatr Blood Cancer. 2007;49:34–40. doi: 10.1002/pbc.20935. [DOI] [PubMed] [Google Scholar]
  • 30.Garvin J, Sposto R, Stanley P, Rorke L, Packer RJ. Childhood Ependymoma: improved survival for patients with incompletely resected tumors with the use of pre-irradiation chemotherapy. Neuro Oncol. 2004;6:456. [Google Scholar]
  • 31.Needle MN, Goldwein JW, Grass J, Cnaan A, Bergman I, Molloy P, Sutton L, Zhao H, Garvin JH, Jr, Phillips PC. Adjuvant chemotherapy for the treatment of intracranial ependymoma of childhood. Cancer. 1997;80:341–347. [PubMed] [Google Scholar]
  • 32.Shu HK, Sall WF, Maity A, Tochner ZA, Janss AJ, Belasco JB, Rorke-Adams LB, Phillips PC, Sutton LN, Fisher MJ. Childhood Intracranial Ependymoma: Twenty-year Experience from a Single Institution. Cancer. 2007;110:432–441. doi: 10.1002/cncr.22782. [DOI] [PubMed] [Google Scholar]
  • 33.Nazar GB, Hoffman HJ, Becker LE, Jenkin D, Hymphreys RP, Hendrick EB. Infratentorial ependymoma in childhood: Prognostic factors and treatment. J Neurosurg. 1990;72:408–417. doi: 10.3171/jns.1990.72.3.0408. [DOI] [PubMed] [Google Scholar]
  • 34.Chiu JK, Woo SY, Ater J, Connelly J, Bruner JM, Maor MH, van Eys J, Oswald MJ, Shallenberger R. Intracranial ependymoma in children: analysis of prognostic factors. J Neuroonc. 1992;13:283–290. doi: 10.1007/BF00172482. [DOI] [PubMed] [Google Scholar]
  • 35.Foreman NK, Love S, Gill SS, Coakham HB. Second-look surgery for incompletely resected fourth ventricle ependymomas: technical case report. Neurosurgery. 1997;40:856–860. doi: 10.1097/00006123-199704000-00042. [DOI] [PubMed] [Google Scholar]
  • 36.Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4:423–436. doi: 10.1038/nrc1369. [DOI] [PubMed] [Google Scholar]
  • 37.Kerbel RS, Viloria-Petit A, Klement G, Rak J. ‘Accidental’ antiangiogenic drugs. Anti-oncogene directed signal transduction inhibitors and conventional chemotherapeutic agents as examples. Eur J Cancer. 2000;36:1248–1257. doi: 10.1016/s0959-8049(00)00092-7. [DOI] [PubMed] [Google Scholar]
  • 38.Vacca A, Iurlaro M, Ribatti D, Minischetti M, Nico B, Ria R, Pellegrino A, Dammacco F. Antiangiogenesis is produced by nontoxic doses of vinblastine. Blood. 1999;94:4143–4155. [PubMed] [Google Scholar]
  • 39.Hanahan D, Bergers G, Bergsland E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J Clin Invest. 2000;105:1045–1047. doi: 10.1172/JCI9872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Browder T, Butterfield CE, Kraling BM, Shi B, Marshall B, O’Reilly MS, Folkman J. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res. 2000;60:1878–1886. [PubMed] [Google Scholar]
  • 41.Colleoni M, Rocca A, Sandri MT, Zorzino L, Masci G, Nole F, Peruzzotti G, Robertson C, Orlando L, Cinieri S, de BF, Viale G, Goldirsch A. Low dose oral methotrexate and cyclophosphamide in metastatic breast cancer: antitumor activity and correlation with vascular endothelial growth factor levels. Ann Oncol. 2002;13:73–80. doi: 10.1093/annonc/mdf013. [DOI] [PubMed] [Google Scholar]
  • 42.Sorio R, Toffoli G, Crivellari D, Bearz A, Corona G, Colussi AM, Libra M, Talamini R, Veronesi A. Oral etoposide in elderly patients with advanced non small cell lung cancer: a clinical and pharmacological study. J Chemother. 2006;18:188–191. doi: 10.1179/joc.2006.18.2.188. [DOI] [PubMed] [Google Scholar]
  • 43.Buckstein R, Kerbel RS, Sheked Y, Nayar R, Foden C, Turner R, Lee CR, Taylor D, Zhang L, Man S, Baruchel S, Stempak D, Bertolini F, Crump M. High-dose celecoxib and metronomic “ low-dose ” cyclophosphamide is an effective and safe therapy in patients with relapsed and refractory aggressive histology non-Hodgkin’s lymphoma. Clin Cancer Res. 2006;12:5190–5198. doi: 10.1158/1078-0432.CCR-06-0474. [DOI] [PubMed] [Google Scholar]
  • 44.Spieth K, Kaufmann R, Gille J. Metronomic oral low-dose treosulfan chemotherapy combined with cyclooxygenase-2 inhibitor in pretreated advanced melanoma: a pilot study. Cancer Chemother Pharmacol. 2003;52:377–382. doi: 10.1007/s00280-003-0678-9. [DOI] [PubMed] [Google Scholar]
  • 45.Glode LM, Barqawi A, Crighton F, Crawford ED, Kerbel R. Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer. 2003;98:1643–1648. doi: 10.1002/cncr.11713. [DOI] [PubMed] [Google Scholar]
  • 46.Baruchel S, Diezi M, Hargrave D, Stempak D, Gammon J, Moghrabi A, Coppes MJ, Fernandez CV, Bouffet E. Safety and pharmacokinetics of temozolomide using a dose-escalation, metronomic schedule in recurrent paediatric brain tumors. Eur J Cancer. 2006;42:2335–2342. doi: 10.1016/j.ejca.2006.03.023. [DOI] [PubMed] [Google Scholar]
  • 47.Stempak D, Gammon J, Halton J, Moghrabi A, Koren G, Baruchel S. A pilot pharmacokinetic and antiangiogenic biomarker study of celecoxib and low-dose metronomic vinblastine or cyclophosphamide in pediatric recurrent solid tumors. J Pediatr Hematol Oncol. 2006;28:720–728. doi: 10.1097/01.mph.0000243657.64056.c3. [DOI] [PubMed] [Google Scholar]
  • 48.Kieran MW, Turner CD, Rubin JB, Chi SN, Zimmerman MA, Chordas C, Klement G, Laforme A, Gordon A, Thomas A, Neuberg D, Browder T, Folkman J. A feasibility trial of antiangiogenic (metronomic) chemotherapy in pediatric patients with recurrent or progressive cancer. J Pediatr Hematol Oncol. 2005;27:573–581. doi: 10.1097/01.mph.0000183863.10792.d4. [DOI] [PubMed] [Google Scholar]
  • 49.Khanna C, Helman LJ. Molecular approaches in pediatric oncology. Annual Review of Medicine. 2006;57:83–97. doi: 10.1146/annurev.med.57.121304.131247. [DOI] [PubMed] [Google Scholar]
  • 50.Kerbel RS. Antiangiogenic therapy: a universal chemosensitization strategy for cancer. Science. 2006;312:1171–1175. doi: 10.1126/science.1125950. [DOI] [PubMed] [Google Scholar]
  • 51.Nyati MK, Morgan MA, Lawrence TS. Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer. 2006;6:876–885. doi: 10.1038/nrc1953. [DOI] [PubMed] [Google Scholar]
  • 52.Gilbertson RJ, Jaros E, Perry RH, Kelly PJ, Lunec J, Pearson AD. Mitotic percentage index: a new prognostic factor for childhood medulloblastoma. Eur J Cancer. 1997;33:609–615. doi: 10.1016/s0959-8049(96)00516-3. [DOI] [PubMed] [Google Scholar]
  • 53.Gilbertson RJ, Clifford SC, MacMeekin W, Meeken W, Wright C, Perry RH, Kelly P, Pearson AD, Lunec J. Expression of the ErbB-neuregulin signaling network during human cerebellar development: implications for the biology of medulloblastoma. Cancer Res. 1998;58:3932–3941. [PubMed] [Google Scholar]
  • 54.Gilbertson R, Wickramasinghe C, Hernan R, Balaji V, Hunt D, Jones-Wallace D, Crolla J, Perry R, Lunec J, Pearson A, Ellison D. Clinical and molecular stratification of disease risk in medulloblastoma. Br J Cancer. 2001;85:705–712. doi: 10.1054/bjoc.2001.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hernan R, Fasheh R, Calabrese C, Frank AJ, Maclean KH, Allard D, Baccaclough R, Gilbertson RJ. ERBB2 upregulates S100A4 and several other prometastatic genes in medulloblastomas. Cancer Res. 2003;63:140–148. [PubMed] [Google Scholar]
  • 56.Gilbertson RJ, Bentley L, Hernan R, Junttila TT, Frank AJ, Haapasalo H, Connelly M, Wetmore C, Curran T, Elenius K, Ellison DW. ERBB receptor signaling promotes ependymoma cell proliferation and represents a potential novel therapeutic target for this disease. Clin Cancer Res. 2002;8:3054–3064. [PubMed] [Google Scholar]
  • 57.Medrzyk F, Korshunov A, Benner A, Toedt G, Pfister S, Radlwimmer B, Lichter P. Identification of gains on 1q and epidermal growth factor receptor overexpression as independent prognostic markers in intracranial ependymoma. Clin Cancer Res. 2006;12:2070–2079. doi: 10.1158/1078-0432.CCR-05-2363. [DOI] [PubMed] [Google Scholar]
  • 58.Sirotnak FM. Studies with ZD1839 in preclinical models. Semin Oncol. 2003;30:12–20. doi: 10.1053/sonc.2003.50028. [DOI] [PubMed] [Google Scholar]
  • 59.Cohen MH, Williams GA, Sridhara R, Chen G, McGuinn WD, Jr, Morse D, Abraham S, Rahman A, Liang C, Lostritto R, Baird A, Pazdur R. United States Food and Drug Administration Drug Approval summary: Gefitinib (ZD1839; Iressa) tablets. Clin Cancer Res. 2004;10:1212–1218. doi: 10.1158/1078-0432.ccr-03-0564. [DOI] [PubMed] [Google Scholar]
  • 60.Ochs JS. Rationale and clinical basis for combining gefitinib (IRESSA, ZD1839) with radiation therapy for solid tumors. Int J Radiat Oncol Biol Phys. 2004;58:941–949. doi: 10.1016/j.ijrobp.2003.09.094. [DOI] [PubMed] [Google Scholar]
  • 61.Geoerger B, Gaspar N, Opolon P, Morizet J, Devanz P, Lecluse Y, Valent A, Lacroix L, Grill J, Vassal G. EGFR tyrosine kinase inhibition radiosensitizes and induces apoptosis in malignant glioma and childhood ependymoma xenografts. Int J Cancer. 2008;123:209–216. doi: 10.1002/ijc.23488. [DOI] [PubMed] [Google Scholar]
  • 62.Taylor MD, Poppleton H, Fuller C, Su X, Liu Y, Jensen P, Magdaleno S, Dalton J, Calabrese C, Board J, Macdonald T, Rutka J, Guha A, Gajjar A, Curran T, Gilbertson RJ. Radial glia cells are candidate stem cells of ependymoma. Cancer Cell. 2005;8:323–335. doi: 10.1016/j.ccr.2005.09.001. [DOI] [PubMed] [Google Scholar]
  • 63.Broniscer A, Panetta JC, O’Shaughnessy M, Fraga C, Bai F, Krasin MJ, Gajjar A, Stewart CF. Plasma and cerebrospinal fluid pharmacokinetics of erlotinib and its active metabolite OSI-420. Clin Cancer Res. 2007;13:1511–1515. doi: 10.1158/1078-0432.CCR-06-2372. [DOI] [PubMed] [Google Scholar]
  • 64.Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, Oh EY, Gaber MW, Finkelstein D, Allen M, Frank A, Bayazitov, Zakharenko SS, Gajjar A, Davidoff A, Gilbertson RJ. A perivascular niche for brain tumor stem cells. Cancer Cell. 2007;11:69–82. doi: 10.1016/j.ccr.2006.11.020. [DOI] [PubMed] [Google Scholar]
  • 65.Jakacki RI, Hamilton M, Gilbertson RJ, Blany SM, Tersak J, Krailo MD, Ingle AM, Voss SD, Dancey JE, Adamson PC. Pediatric phase I and pharmokinetic study of erlotinib followed by the combination of erlotinib and temozolamide: a Children’s Oncology Group Phase I Consortium Study. J Clin Oncol. 2008;26:4921–4927. doi: 10.1200/JCO.2007.15.2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Beaudry P, Nilsson M, Rioth M, Prox D, Poon D, Xu L, Zweidler-Mckay P, Ryan A, Folkman J, Ryeom S, Heymach J. Potent antitumor effects of ZD6474 on neuroblastoma via dual targeting of tumor cells and tumor endothelium. Mol Cancer Ther. 2008;7:418–424. doi: 10.1158/1535-7163.MCT-07-0568. [DOI] [PubMed] [Google Scholar]
  • 67.Broniscer A, Baker SJ, Stewart CF, Merchant TE, Laningham FH, Shaiquevich P, Kocak M, Morris B, Endersby R, Ellison DW, Gajjar A. Phase I and Pharmacokinetic studies of Erlotinib administered concurrently with radiotherapy for children, adolescents and young adults with high-grade glioma. Clin Cancer Res. doi: 10.1158/1078-0432.CCR-08-1923. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kerbel RS. Clinical trials of antiangiogenic drugs: opportunities, problems and assessment of initial results. J Clin Oncol. 2001;19:45S–51S. [PubMed] [Google Scholar]
  • 69.Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors. Nat Rev Cancer. 2002;2:727–739. doi: 10.1038/nrc905. [DOI] [PubMed] [Google Scholar]
  • 70.Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967–974. doi: 10.1038/nature04483. [DOI] [PubMed] [Google Scholar]
  • 71.Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3:401–410. doi: 10.1038/nrc1093. [DOI] [PubMed] [Google Scholar]
  • 72.Bergers G, Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro-onc. 2005;7:452–464. doi: 10.1215/S1152851705000232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kerbel RS. Tumor angiogenesis: past, present and the near future. Carcinogenesis. 2000;21:505–515. doi: 10.1093/carcin/21.3.505. [DOI] [PubMed] [Google Scholar]
  • 74.Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res. 2006;66:7843–7848. doi: 10.1158/0008-5472.CAN-06-1010. [DOI] [PubMed] [Google Scholar]
  • 75.Escudier B, Cosaert J, Pisa P. Bevacizumab: dirent anti-VEGF therapy in renal cell carcinoma. Expert Rev Anticancer Ther. 2008;8:1545–1557. doi: 10.1586/14737140.8.10.1545. [DOI] [PubMed] [Google Scholar]
  • 76.Vogelzang NJ. Treatment options in metastatic renal carcinoma: an embarrassment of riches. J Clin Oncol. 2006;24:1–3. doi: 10.1200/JCO.2005.03.7234. [DOI] [PubMed] [Google Scholar]
  • 77.Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Homgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Eng J Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 78.Jain RK, Duda DG, Clark JW, Loeffler JS. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol. 2006;3:24–40. doi: 10.1038/ncponc0403. [DOI] [PubMed] [Google Scholar]
  • 79.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, Dowell JM, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Wagner M, Bigner DD, Friedman AH, Friedman HS. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253–1259. doi: 10.1158/1078-0432.CCR-06-2309. [DOI] [PubMed] [Google Scholar]
  • 80.Modena P, Lualdi E, Facchinetti F. Identification of tumor-specific molecular signatures in intracranial ependymoma and association with clinical characteristics. J Clin Oncol. 2006;24:5223–5233. doi: 10.1200/JCO.2006.06.3701. [DOI] [PubMed] [Google Scholar]
  • 81.Tabori U, Ma J, Carter M, Zielenska M, Rutka J, Bouffet E, Bartels U, Malkin D, Hawkins C. Human telomere reverse transcriptase expression predicts progression and survival in pediatric intracranial ependymoma. J Clin Oncol. 2006;24:1522–1528. doi: 10.1200/JCO.2005.04.2127. [DOI] [PubMed] [Google Scholar]
  • 82.Dyer S, Prebble E, Davison V, Davies P, Ramani P, Ellison D, Grundy R. Genomic imbalances in pediatric ependymomas define clinically relevant groups. Am J Pathol. 2002;161:2133–2141. doi: 10.1016/S0002-9440(10)64491-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Korshunov A, Neben K, Wrobel G, Tews B, Benner A, Hahn M, Golanov A, Lichter P. Gene expression patterns in ependymomas correlate with tumor location, grade, and patient age. Am J Pathol. 2003;163:1721–1727. doi: 10.1016/S0002-9440(10)63530-4. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES