antineoplastons for astrocytic tumours pediatrics

Page 1

Pediatr Drugs 2006; 8 (3): 167-178 1174-5878/06/0003-0167/$39.95/0

THERAPY IN PRACTICE

 2006 Adis Data Information BV. All rights reserved.

Treatments for Astrocytic Tumors in Children Current and Emerging Strategies Stanislaw R. Burzynski Burzynski Clinic, Houston, Texas, USA

Contents Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167 1. Therapeutic Approaches to Different Types of Astrocytomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.1 Low-Grade Astrocytomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.1.1 Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.1.2 Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.1.3 Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.1.4 Targeted Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.2 Optic Pathway Gliomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 1.2.1 Surgery and Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.2.2 Chemotherapy and Targeted Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.3 High-Grade Astrocytomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.3.1 Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.3.2 Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.3.3 Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 1.4 Brainstem Gliomas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 1.4.1 Surgery and Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 1.4.2 Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 1.4.3 Targeted Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2. Late Effects of Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1 Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.1 Radiation Necrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.2 Necrotizing Leukoencephalopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.3 Cranial Neuropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.4 Cognitive Disability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.5 Endocrine Dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.1.6 Radiation-Induced Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.2 Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.2.1 Chronic Myelosuppression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.2.2 Gonadal Dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 2.2.3 Renal Insufficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.2.4 Pulmonary Toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.2.5 Neurotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.2.6 Hearing Loss . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.2.7 Cognitive Disability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.2.8 Chemotherapy-Induced Hematologic Malignancies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 2.3 Targeted Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174 3. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174

Abstract

Strategies for the treatment of childhood cancer have changed considerably during the last 50 years and have led to dramatic improvements in long-term survival. Despite these accomplishments, CNS tumors remain the leading cause of death in pediatric oncology. Astrocytic tumors form the most common histologic group among childhood brain tumors. They are a heterogeneous group that from a practical therapeutic point of view can be


168

Burzynski

subdivided into low-grade astrocytomas (LGA), optic pathway gliomas (OPG), high-grade astrocytomas (HGA), and brainstem gliomas (BSG). This article focuses on the practical application of treatments that lead to long-term survival, improved quality of life, and reduced long-term complications. Improvement in therapy has led to better outcomes for patients with LGA and OPG. Careful follow-up without any treatment is indicated for a small percentage of patients diagnosed with LGA with an indolent course including children with neurofibromatosis type 1 (NF1). Surgery is the main recommended treatment for children with resectable LGA. Radiation therapy is generally recommended for children with progressive LGA, or after failure of chemotherapy, accomplishing tumor control at 10 years in over 60% of patients. Cytotoxic chemotherapy is usually reserved for children who have had treatment failure with surgery and radiation therapy. It is also offered for children who are too young to be treated with radiation or to defer or avoid radiotherapy. Carboplatin and vincristine achieve 5% complete and 28% partial responses but the use of vincristine is criticized due to poor penetration of the CNS. A regimen of tioguanine, procarbazine, mitolactol, lomustine, and vincristine is frequently administered as an alternative to carboplatin and vincristine in LGA. The introduction of temozolomide has allowed better responses, including a 24% complete response rate compared with 0–5% complete response rates with the previous regimens. OPG are usually histologically LGA, and are treated with similar chemotherapy regimens. OPG is the most common type of brain tumor associated with NF1. Tumor growth in some of these patients is slow with no treatment recommended for an extended period of time. The prognosis for children with the remaining types of astrocytomas remains poor. Surgical resection is typically the first step in the treatment of HGA followed in older children by radiation therapy. The data regarding chemotherapy are mixed. Combination chemotherapy before or after radiation, including cisplatin, carmustine, cyclophosphamide, and vincristine or carboplatin, ifosfamide, cyclophosphamide, and etoposide has provided disappointing results. Clinical trials with temozolomide and agents directed against single targets have not shown substantially better results, but it is hoped that currently conducted studies will provide better outcomes. Diffuse intrinsic BSG are among the most difficult-to-treat brain tumors. Surgical treatment is not recommended for diffuse intrinsic BSG and standard radiation therapy is typically given in children aged >3 years. None of the numerous chemotherapy regimens, including temozolomide, has provided a significant response rate or an improvement in survival. It is expected that newer agents affecting multiple targets such as AEE-788 and antineoplastons, and combinations of single-targeted agents with chemotherapy will provide better results. Careful evaluation of histology, location of the tumor, patient age, and consideration of treatment-related morbidity play an important part in selecting between clinical observation, surgery, radiation, chemotherapy, or investigational agents. The goals of treatment for astrocytic tumors should extend well beyond objective responses and increased survival. Improvement of quality of life is an equally important objective of treatment. Radiation therapy and chemotherapy result in serious late toxicities.

Over the last 50 years, there has been dramatic progress in the treatment of childhood cancer. Long-term survival for all childhood malignancies has improved from <10% to 80%.[1] Despite these accomplishments, CNS tumors remain the leading cause of death in pediatric oncology.[2] Primary CNS tumors affect approximately 5500 children in the US each year and are the most common solid neoplasms in childhood.[3] Gliomas account for 56% of tumors in children aged <15 years and the most common histologic group is astrocytic tumors.[3] Astrocytomas are a heterogeneous group consisting of a number of histologic varieties.[4] From a practical therapeutic point of view, they can be subdivided into low-grade astrocytomas (LGA), optic pathway gliomas (OPG), high-grade astrocytomas (HGA), and brainstem gliomas (BSG). Progress has been made in the treatment of LGA and OPG, but there remains a great need to  2006 Adis Data Information BV. All rights reserved.

improve the results of therapy for HGA and BSG.[5-7] Improved treatment outcomes for HGA including glioblastoma multiforme (GBM) with temozolomide in adult patients and the introduction of targeted agents raised hopes for more successful therapy of pediatric malignant brain tumors.[8-11] Results from clinical studies of temozolomide in childhood brain tumors have not been as good, but it is hoped that currently conducted larger clinical trials will provide better outcomes.[7,10,11] Targeted agents have shown interesting results in some clinical trials but in the majority, results fell short of expectations.[12] It is hoped that newer multi-targeted agents and combinations of single-targeted agents with chemotherapy will provide better results.[12] This article provides the latest information on the treatment of pediatric astrocytomas. The emphasis is on the practical application of the treatments. Readers are referred for further information Pediatr Drugs 2006; 8 (3)


Treatments for Astrocytic Tumors

to the numerous review articles that cover genetics, pathology, and the clinical and radiographic presentations of these tumors.[6,11-14] 1. Therapeutic Approaches to Different Types of Astrocytomas 1.1 Low-Grade Astrocytomas

LGA, which consist of grade 1 and 2 tumors, are the most common type of pediatric neoplasms within the category of glioma.[3] Of an entire group of 3793 children aged <15 years with primary brain tumors in the US, pilocytic astrocytoma was diagnosed in 24%.[3] In addition to pilocytic astrocytoma, grade 1 tumors include subependymal giant-cell astrocytomas, and grade 2 tumors are subdivided into diffuse astrocytomas (fibrillary, gemistocytic, and protoplasmic), pleomorphic xanthoastrocytomas, and mixed oligoastrocytomas.[6,15] The cerebellum is the most common location and accounts for 15–25% of all LGA. Published treatment data usually do not report results limited to LGA, but describe the therapy given to mixed small groups of adult and pediatric patients with tumors including other low-grade gliomas and newly diagnosed and recurrent tumors. Careful follow-up without any treatment is indicated for a small percentage of patients diagnosed with LGA with an indolent course, including children with neurofibromatosis type 1 (NF1).[16] The literature reports a sizable number of cases of spontaneous regressions of LGA.[17,18] Tumors causing aqueductal obstruction may require ventriculoperitoneal shunting or ventriculostomy; the majority of patients with low-grade tectal gliomas do not have progression for up to 10 years after such procedures.[19,20] 1.1.1 Surgery

Surgery is the main recommended treatment for children with resectable LGA. Shaw and Wisoff[18] compiled the results of five prospective clinical trials of low-grade gliomas in adults and children. A group of 660 pediatric patients was enrolled and evaluated based on the extent of surgical resection. Patients with gross total resection were observed, but those with subtotal resection or biopsy were either observed or administered radiation therapy.[18] The extent of resection was the main factor affecting overall survival; complete resection allowed long-term diseasefree survival in over 80% of children.[18,21] Patients with partial resection and marked residual disease usually have recurrence and tumor progression.[22] In such cases, ‘second-look’ surgery may be considered prior to radiation therapy and chemotherapy. 1.1.2 Radiation Therapy

Radiation therapy is generally recommended for children with progressive LGA, or after failure of chemotherapy, accomplishing tumor control at 10 years in over 60% of patients.[23] The diagnosis of LGA in a child creates a difficult clinical situation because of debilitating late toxicity of radiation therapy. Minimizing the dose  2006 Adis Data Information BV. All rights reserved.

169

and field of radiation helps to allow normal growth and intellectual development in young children. Focal radiation fields covering the tumor volume visible on magnetic resonance imaging (MRI) [T2-weighted images] with a 2cm margin is usually recommended. Stereotactic radiotherapy improves the results and reduces toxicity, but hyperfractionated radiation therapy does not offer additional benefits.[24-26] 1.1.3 Chemotherapy

Cytotoxic chemotherapy is usually reserved for children who have had treatment failure with surgery and radiation therapy, or who are too young to be treated with radiation but it is also offered to a growing number of older children to defer or avoid radiotherapy.[6,27,28] Until approximately 20 years ago, LGA were seldom treated with chemotherapy. The initial regimens used dactinomycin, lomustine, and vincristine, and have been summarized in excellent reviews.[6,27,29,30] Selected recent regimens are summarized in table I. Carboplatin and vincristine achieve 5% complete response (CR) rates and 28% partial response (PR) rates, and vincristine and etoposide achieve 5% PR rates in patients with recurrent and newly diagnosed low-grade gliomas (mostly LGA).[36,37] Treatment with vinblastine for recurrent and refractory tumors has accomplished a 20% PR rate.[31,40] Tamoxifen and carboplatin in newly diagnosed and recurrent tumors allowed a 15% PR rate, but only 47% progression-free survival (PFS) at 3 years.[35] A regimen of tioguanine, procarbazine, mitolactol, lomustine, and vincristine first used in the treatment of hypothalamic gliomas is frequently administered as an alternative to carboplatin and vincristine in LGA.[38] The introduction of temozolomide allowed better responses, including 24% CR and 37% PR rates (in children and adults) in a phase II study.[33] Additional studies with temozolomide revealed a 10% (three patients) PR rate, but no CR, and 1year PFS was slightly more than 50%.[32] 1.1.4 Targeted Therapy

Clinical trials with agents affecting single targets are in progress and the preliminary results of multi-targeted therapy with the antineoplastons (ANP) A10 and AS2-1 have been reported.[39] In a small group of patients with progressive LGA, ANP accomplished a 60% CR rate, 10% PR rate, median survival of 7 years and 9 months, and maximum survival of more than 15 years.[39] 1.2 Optic Pathway Gliomas

OPG constitute approximately 6% of pediatric brain tumors.[41] They typically occur in children aged <12 years, and a majority of these tumors involve the chiasm and hypothalamus.[42-44] Histologically, these are low-grade gliomas. The type of treatment and prognosis varies from one patient to another. Chiasmatic and hypothalamic tumors create more difficulties including pronounced loss of vision and hypothalamic dysfunction than gliomas Pediatr Drugs 2006; 8 (3)


170

 2006 Adis Data Information BV. All rights reserved.

Table I. Selected chemotherapy regimens for the treatment of low-grade astrocytoma in children Reference

Study

Tumor

Treatment

Efficacy

type

type

(no. of pts)

OS [%]

Bouffet et al.[31]

Pilot

R

Vinblastine (20)

Gururangan et al.[32]

Phase II

P

Temozolomide (32)a

Quinn et al.[33]

Phase II

RP

Kuo et al.[34]

Phase II

Walter et al.[35] Packer et al.[36]

PR [% (no.)]

SD + MR [% (no.)]

PD [% (no.)] PFS (%)

0 (0)

20 (4)

60 (12)

20 (4)

100 (1y)

0 (0)

10 (3)

43 (13)

47 (14)

51 (1y)

Temozolomide (46)b

24 (11)

37 (17)

35 (16)

4 (2)

76 (1y)

P

Temozolomide (13)

0 (0)

23 (3)

46 (6)

31 (4)

Phase II

NR

Tamoxifen, carboplatin (14)c

0 (0)

15 (2)

70 (9)

15 (2)

47 (3y)

Phase II

P

Carboplatin, vincristine (78)

5 (4)

28 (22)

60 (47)

7 (5)

86 (1y);

96 (1y)

69 (3y)

CR [% (no.)]

68 (3y) Pons et al.[37]

Phase II

NR

Vincristine, etoposide (20)

0 (0)

5 (1)

70 (14)

25 (5)

Petronio et al.[38]

Phase II

RP

Tioguanine, procarbazine,

0 (0)

60 (9)

13 (2)

27 (4)

60 (6)

10 (1)

30 (3)

0 (0)

mitolactol, lomustine, vincristine (15) Burzynski[39]

Phase IId

P

ANP (10)

100 (1y); 90 (3y);

90 (1y); 90 (3y)

MST = 93mo

a

Thirty evaluable pts.

b

Children and adults.

c

Thirteen evaluable pts.

d

Preliminary results.

survival; P = progressive tumor; PD = progressive disease; PFS = progression-free survival; PR = partial response; pts = patients; R = recurrent tumor; RP = recurrent and progressive tumor; SD = stable disease.

Burzynski

Pediatr Drugs 2006; 8 (3)

ANP = antineoplastons A10 and AS2-1; CR = complete response; MR = minor response; MST = median survival time; NR = newly diagnosed and recurrent tumor; OS = overall


Treatments for Astrocytic Tumors

of the optic nerve. Tumors that occur in children aged <1 year are typically larger and cause severe loss of vision and hypothalamic dysfunction while gliomas in older children carry a better prognosis. OPG is the most common type of brain tumor associated with NF1. In various series, 25–60% of the children diagnosed with this type of tumor had NF1.[45,46] Growth of OPG in some of these patients is slow with no treatment recommended for an extended period of time. Monitoring without therapeutic intervention is suggested for asymptomatic tumors. A biopsy is not required for hypothalamic and chiasmatic lesions in patients with typical MRI findings. Progressive visual deterioration may require surgery. Radiation and chemotherapy regimens are appropriate for recurrent and posterior OPG.[16,18,21,25,28,38] 1.2.1 Surgery and Radiation Therapy

These patients are typically not candidates for radical surgery, but when a substantial exophytic component is present, a >60% resection can be accomplished in very young children.[43] According to a report from a neurosurgical group, these tumors can remain stable after resection for a mean of 27 months.[47] Radiation therapy contributes to stabilization in most patients, but the chronic toxicity associated with such treatment precludes its use in young children.[48,49] 1.2.2 Chemotherapy and Targeted Therapy

Because of concerns about the long-term toxicity of radiation therapy, a number of chemotherapeutic regimens have been used in the treatment of OPG. The initial successful regimen included a combination of dactinomycin and vincristine in 24 children with a median age of 1.6 years.[50] PR was achieved in 12.5% of patients, stable disease (SD) in 50%, and progressive disease (PD) in 37.5%. The median survival was 4.3 years and 25% had a projected 70-month event-free survival. In another study, the treatment of 113 evaluable children (median age 3.7 years) with vincristine and carboplatin resulted in a 51% PR rate, 41% SD rate, and 8% PD rate.[51] Forty-two percent of children developed progression and median time to progression (MTP) was 22.5 months. New treatment regimens including ANP are under investigation.[52] 1.3 High-Grade Astrocytomas

Over 15% of all pediatric brain tumors are considered to be high-grade gliomas (WHO grade 3 and 4).[3,7] GBM (grade 4) is less common than anaplastic astrocytoma (grade 3; AA); in children aged <15 years, it constitutes only 2.8% of all CNS tumors.[3] The incidence of HGA is lower in very young children (3–11%) compared with older children.[53] The incidence of HGA differs depending on tumor location. AA and GBM constitute approximately 25% of all childhood cortical gliomas.[54] Cerebellar high 2006 Adis Data Information BV. All rights reserved.

171

grade gliomas occur substantially less frequently (<5% of cerebellar gliomas).[55] 1.3.1 Surgery

Surgical resection is typically the first step in the treatment of HGA. The results of treatment of 131 children with GBM and AA confirm that >90% resection allows a statistically significant PFS.[56] PFS at 5 years for radical resections was 35% compared with 17% for a less aggressive procedure. Patients with AA had a PFS of 44% and patients with GBM had a PFS of 26% after radical surgery, compared with a PFS of 22% and 4%, respectively, with a more conservative procedure. Similar results have been obtained in additional studies.[57] 1.3.2 Radiation Therapy

Radiation therapy is usually not recommended in children aged <3 years because of unacceptable toxicity. The introduction of three-dimensional conformal therapy may allow successful treatment of such children, but clinical trials to study long-term adverse effects are required. Older children are usually treated after recovery from surgery with a total dose of radiation to the tumor, or the tumor bed, of between 54 and 60Gy in 1.8–2Gy fractions.[7] Hyperfractionated therapy and craniospinal radiation have not improved the outcomes,[58] and are usually not recommended. In the past, patients with disseminated disease were recommended to undergo craniospinal radiation, which is no longer the standard approach.[58] Stereotactic radiosurgery resulted in a 33% 3-year PFS in a small series of patients with newly diagnosed and recurrent high-grade gliomas, but requires further study.[59] Fewer than 25% of children with HGA achieve long-term survival after radiation therapy alone.[54,60] 1.3.3 Chemotherapy

Since the 1980s, a number of combination chemotherapy regimens have been used in the treatment of HGA. However, encouraging initial results with lomustine, vincristine, and prednisone, in combination with radiation therapy, have not been confirmed by subsequent studies.[61-63] Neoadjuvant chemotherapy with cisplatin, cytarabine, procarbazine, and topotecan did not show significant benefits.[64,65] Combination chemotherapy before or after radiation therapy, including cisplatin, carmustine, cyclophosphamide, and vincristine, or carboplatin, ifosfamide, cyclophosphamide and etoposide, and concurrent high-dose chemotherapy and radiation therapy with stem cell support have provided disappointing results.[66,67] A meta-analysis of the results of 27 clinical studies that accrued 576 children did not show significant benefits of chemotherapy.[68] Initial studies with temozolomide showed some responses.[11,69] A more recent report of a small series of 11 patients with recurrent malignant brain tumors (six GBM, two AA, one BSG, and one primitive neuroectodermal tumor) treated with temozolomide and etoposide has shown more encouraging results with one CR, seven PR, and an MTP of 8 months.[70] Another Pediatr Drugs 2006; 8 (3)


 2006 Adis Data Information BV. All rights reserved.

Seventeen evaluable pts. b

a

Fifty-seven evaluable pts.

RPS (10) Phase II Burzynski et al.[89]

RP (30) Phase II Burzynski et al.[88]

R (21)b Phase II Lashford et al.[10]

ANP = antineoplastons A10 and AS2-1; CR = complete response; MST = median survival time; N = newly diagnosed tumor; NA = not available; OS = overall survival; PD = progressive disease; PR = partial response; pts = patients; R = recurrent tumor; RP = recurrent and progressive tumor; RPS = recurrent and progressive tumors in children aged <4y; SD = stable disease.

30 (3) 40 (4) 0 (0) 30 (3) 26.3 60; 20 ANP

30 (9) 23 (7) 20 (6) 27 (8) 19.9

Temozolomide

Temozolomide, irinotecan 56 Multiinstitutional Broniscer et al.[86]

N (33)

Phase III

N (66a)

54

Cisplatin

ANP

46.7; 30

77.8 (14) 16.7 (3) 5.6 (1) 0 (0) 6.2 NA; NA

NA NA

44 (25) 31 (18) 2 (1)

NA 12 0; 0

MST (mo) OS (%) [2y; 5y] radiation therapy (Gy) (no. of pts)

chemotherapy

ANP

Efficacy Treatment Tumor type

Mandell et al.[78]

None of the numerous chemotherapeutic regimens, including temozolomide given before, with, and after radiation therapy has provided significant response rates or improvement in survival.[80-86] High-dose chemotherapy with autologous bone marrow transplantation has not produced better results.[87] Recurrent DBSG and tumors in small children carry an especially poor prognosis with no standard treatment available; temozolomide is also not effective in these patients (table II).[10] It is not expected that patients with recurrent DBSG will survive longer than 6 months, regardless of the therapy.[10]

Study type

1.4.2 Chemotherapy

Reference

Surgical treatment is not recommended for DBSG and standard radiation therapy of 54Gy in daily fractions of 1.8Gy through parallel opposed portals is typically given in children aged >3 years.[74,78,79] A number of hyperfractionation protocols have been tried without success.[78,80] Patients treated with standard irradiation have disappointing prognoses; only approximately 7% would survive 2 years with no survival expected over 5 years (table II).[78]

Table II. Treatment of diffuse, intrinsic brainstem glioma in children

1.4.1 Surgery and Radiation Therapy

CR [% (no.)]

Gliomas located in the brainstem affect approximately 690 children annually in the US.[3] Based on autopsy reports, the majority of patients have AA.[72] However, biopsies are performed only in some cases because of the difficult location, and are misleading because of sampling error. Typically, these tumors are classified based on MRI appearance into the following types: focal, dorsal, exophytic, cervicomedullary, and diffuse intrinsic BSG (DBSG).[72] DBSG is the most common (85% of cases) and most difficult to treat and is generally regarded as an HGA. DBSG is more aggressive in infants than in older children, and it is easier to manage in children with NF1.[73,74] Such children are frequently asymptomatic and have a stable tumor size for a longer time. Genetic abnormalities in DBSG have not been sufficiently studied.[75] Epidermal growth factor-receptor protein is detected in one-third of these patients and approximately 50% have a mutation of TP53 and loss of the PTEN tumor suppressor genes.[75-77] DBSG is among the most difficult-to-treat brain tumors; the remaining types of brainstem gliomas are treated in the same way as tumors in other parts of the brain with similar histopathologic diagnoses.

8.5

1.4 Brainstem Gliomas

7.1; 0

PR [% (no.)]

SD [% (no.)]

PD [% (no.)]

multi-institutional study of 31 children and young adults with newly diagnosed HGA and unfavorable LGA (48% GBM and 32% AA) treated with irinotecan followed by radiation therapy and temozolomide did not confirm the activity of temozolomide and irinotecan.[71] Despite numerous studies, the treatment of pediatric HGA remains suboptimal and the results are disappointing.

NA

Burzynski

23 (13)

172

Pediatr Drugs 2006; 8 (3)


Treatments for Astrocytic Tumors

1.4.3 Targeted Therapy

Single-targeted therapy has not yet been used in DBSG, but multi-targeted therapy with ANP has shown promising results.[12,88-91] In a group of 30 evaluable patients with recurrent and progressive DBSG, >40% of patients survived for more than 2 years and 30% more than 5 years. The responses included CR in 27%, PR in 20%, SD in 23%, and PD in 30%.[12,88] A small group of ten evaluable children aged <4 years diagnosed with DBSG was treated with ANP; the youngest was a 3-month-old infant.[89] The 2-year survival rate was 60%, 5-year survival was 20%, and the maximum survival was more than 7 years. CR occurred in 30%, SD in 40%, and PD in 30%.[89] The results are compiled in table II. 2. Late Effects of Therapy Interest in the research of late effects of radiation and chemotherapy has expanded in parallel with an improvement in efficacy and longer survival in children with brain tumors. 2.1 Radiation Therapy

Chronic toxicity of radiation affects the majority of children treated for brain tumors and in a significant percentage of cases leads to chronic disabilities and death. 2.1.1 Radiation Necrosis

In a series of 80 patients, Sheline et al.[92] determined that the interval from completion of radiation therapy to development of cerebral necrosis ranged from 4 months to 7.5 years. Recurrent tumors and late radiation necrosis share many similar clinical and MRI features. There is no effective treatment for delayed radiation necrosis; however, some patients have benefited from hyperbaric oxygen treatment. The incidence of radiation necrosis is related to the dosage of radiation and may occur in up to 7% of patients treated with hyperfractionated radiation.[93] Maintaining the dose of radiation for low-grade tumors to 54Gy in 30 fractions and for high-grade gliomas to 60Gy in 33 fractions may prevent radiation necrosis and subsequent death.[94] 2.1.2 Necrotizing Leukoencephalopathy

Necrotizing leukoencephalopathy was first described in children who received cranial irradiation and methotrexate.[95] Patients with this syndrome develop demyelinization, multifocal coagulation necrosis, and gliosis, and most have permanent neurologic deficits.[96,97] No known treatment exists for this condition, but it may reverse spontaneously in some cases. 2.1.3 Cranial Neuropathy

Optic neuropathy occurs in patients receiving high doses of radiation. This adverse effect was not reported in the study by Parson et al.,[98] in which 106 adult patients with head and neck tumors received <59Gy, but higher doses of radiation increase the incidence of optic neuropathy to 47%.[99]  2006 Adis Data Information BV. All rights reserved.

173

2.1.4 Cognitive Disability

Numerous studies have confirmed the deleterious effect of radiation on cognitive function.[100-102] The severity of impairment relates to the total dose of radiation, the fraction size, and the patient’s age. Children aged <4 years are especially vulnerable. Adults who received radiation therapy for brain tumors in childhood have greater difficulty finding employment.[103] 2.1.5 Endocrine Dysfunction

In various studies, 47–65% of children treated with radiation for brain tumors had chronic growth hormone deficiency.[104-106] As a result, a high proportion of survivors have short stature.[107] Irradiation of the hypothalamic-pituitary axis can result in thyroid hormone and gonadotropin deficiency. In some studies, more than two-thirds of children developed postradiation thyroid dysfunction.[108] Gonadotropin deficiency leads to a lack or an interruption in pubertal development. The incidence of gonadotropin deficiency was as high as 11% in one study.[109] Children with endocrine dysfunction are evaluated and treated with hormone replacement. 2.1.6 Radiation-Induced Tumors

Radiation-induced tumors are distinct entities and include meningiomas, cranial sarcomas, and gliomas.[110] In a large series of children who had more than one malignancy, in approximately half of the patients, radiation therapy for the first malignancy was a possible etiologic factor for a second tumor. In approximately 25% of these patients, NF1 was felt to be a contributing factor.[111] Additional studies have supported an increased risk of a secondary malignancy in children after irradiation.[112,113] The British Childhood Cancer Research Group found a 5-fold increase in the risk of a secondary malignancy in a population of over 10 000 survivors of childhood malignancies.[114] 2.2 Chemotherapy

The use of chemotherapy as a primary treatment for astrocytoma, especially in very young children, has been justified by a reduction in radiation-induced brain damage.[115] Unfortunately, cytotoxic chemotherapy also contributes to a wide spectrum of chronic toxicities. 2.2.1 Chronic Myelosuppression

Nitrosoureas (carmustine and lomustine), cisplatin, and carboplatin may cause chronic myelosuppression that persists after discontinuation of chemotherapy.[116-118] Serious infection caused by leukopenia is the common fatal complication of carmustine treatment. Hemorrhage due to thrombocytopenia is less common, but cisplatin may cause significant anemia.[117] 2.2.2 Gonadal Dysfunction

Carmustine, lomustine, procarbazine, and cisplatin are known to cause gonadal dysfunction in girls and boys.[119-121] In prepubertal girls, primary ovarian dysfunction may improve and Pediatr Drugs 2006; 8 (3)


174

Burzynski

ovarian function may return to normal after the cessation of treatment with carmustine.[122] Nitrosoureas contribute to a high incidence of oligospermia.[123] 2.2.3 Renal Insufficiency

Nitrosoureas and cisplatin may cause renal insufficiency.[124] Nephrotoxicity is proportional to the total dosage of chemotherapy. It may persist following the cessation of treatment, and lead to chronic renal failure requiring dialysis.[125,126] 2.2.4 Pulmonary Toxicity

An interstitial inflammatory process in the lungs causing decreased oxygen transfer was first reported in 1976 as a fatal complication of carmustine treatment.[127] Pneumonitis has been reported with procarbazine and vincristine.[128,129] These complications may respond to corticosteroid administration. 2.2.5 Neurotoxicity

Intracarotid administration of carmustine has been shown to cause ipsilateral visual loss in 25–65% of patients.[130,131] Loss of vision may start several weeks after the treatment and progress to irreversible blindness. Leukoencephalopathy and increased incidences of seizures have been described in patients after high-dose carmustine.[132-134] Peripheral neuropathy is a well known adverse event associated with vincristine.[135] In some cases, cranial nerve dysfunction develops with diplopia, hoarseness, dysphagia, and facial nerve paralysis. Autonomic neuropathy with orthostatic hypotension and CNS symptoms including depression, confusion, and insomnia have also been reported.[136] Cisplatin may cause peripheral neuropathy that may progress to motor involvement.[137] The neuropathy may continue and worsen long after discontinuation of cisplatin.[138,139] 2.2.6 Hearing Loss

Treatment with cisplatin may result in sensory hearing loss.[140] It may start as tinnitus and progress to high-frequency hearing loss and deafness. Decreased hearing may worsen after discontinuation of the drug. Periodic audiograms may help to detect initial toxicity before occurrence of symptoms. 2.2.7 Cognitive Disability

Chemotherapy-induced decreased hearing (see section 2.2.6) may affect speech, normal cognitive development, and learning in children. Very young children have shown an abnormality in physical and psychologic development after pre-irradiation chemotherapy for brain tumors.[141] Neuropsychologic evaluation is recommended at the completion of chemotherapy and again 3–5 years later for a young child. 2.2.8 Chemotherapy-Induced Hematologic Malignancies

Carmustine and lomustine have been reported to contribute to myelofibrosis, secondary myelodysplasia, and acute leukemia following treatment of gliomas.[142,143] The incidence of leukemia is  2006 Adis Data Information BV. All rights reserved.

less frequent after treatment with procarbazine and cisplatin.[144,145] 2.3 Targeted Therapy

Therapy oriented against single targets has not yet been used in children for a sufficient period to determine latent effects. Based on the results in adult patients, the incidence of late toxicities is substantially lower than that for radiation and chemotherapy.[146] Multi-targeted ANP therapy is free from chronic toxicity in children and adults based on the results of numerous clinical studies involving 1652 adults and 335 children.[147] Long-term follow-up of children treated with ANP for astrocytomas revealed normal development, no cognitive or endocrine deficiencies, and normal fertility. A substantial number of these patients have been tumor free for >5 years and the follow-up period for some of these patients is >17 years. Data in the emerging literature on gene or protein micorarray analysis of tumors show these techniques to predict responses to targeted therapies and reduce toxicity with greater precision than is possible with clinical findings.[148] It is hoped that such an approach will provide individualized care for patients with brain tumors.[149] 3. Conclusion Astrocytomas form the most common histologic group among childhood brain tumors. Approaches to treatment have changed markedly during the last 50 years and have led to the following considerations. Patients with completely resected tumors usually do not require additional therapy, but should be followed up for possible recurrence. Children with NF1 should receive treatment only in cases of morbidity and clear progression. Radiation is the therapy of choice for newly diagnosed DBSG and as adjuvant treatment for children aged >3 years with HGA and progressive, recurrent, and unresectable LGA and OPG. Chemotherapy is the standard approach for younger children and even though it is less efficacious than radiation, it has a more acceptable toxicity profile. The use of vincristine, which is still administered, is open to criticism since published results suggest that vincristine does not penetrate the CNS and high concentrations in the cerebrospinal fluid are fatal.[150] Despite indisputable benefits from radiation and chemotherapy, approximately two-thirds of childhood cancer survivors experience at least one latent adverse effect and about 25% experience delayed toxicities that are severe or life threatening.[151,152] The goals of treatment for astrocytomas should extend well beyond objective responses and increased survival. Improvement of quality of life is an equally important objective of treatment. Radiation therapy and chemotherapy result in serious late toxicities in a significant number of children. Known late effects of Pediatr Drugs 2006; 8 (3)


Treatments for Astrocytic Tumors

radiation include radiation necrosis, necrotizing leukoencephalopathy, cranial neuropathy, cognitive disability, endocrine dysfunction, and radiation-induced tumors. Cytotoxic chemotherapy may cause chronic myelosupression, gonadal dysfunction, renal insufficiency, pulmonary toxicity, and hearing loss in addition to neurotoxicity, cognitive dysfunction, and secondary malignancies. Quality-of-life studies have been performed in patients with recurring malignant gliomas treated with brachytherapy, but quality-of-life data in patients receiving chemotherapy for astrocytoma are limited.[153,154] Limited applications of targeted therapy including ANP have contributed to significant responses, long-term survival, and a good quality of life with no chronic toxicities.[12] Genomic and molecular studies have revealed substantial differences between pediatric and adult brain tumors. TP53 tumor suppressor gene mutations occur in approximately 40% (50% in DBSG) of pediatric malignant gliomas and are associated with poor survival. p16 and retinoblastoma gene deletions are found in approximately 50% of tumors.[155] Mutations of PTEN and amplification of epidermal growth factor receptor are common in adult HGA, but are rare in pediatric tumors.[155] Successful treatment of astrocytomas in children requires targeting different mediators of neoplastic growth than those in adult patients. Agents affecting multiple targets currently in clinical trials such as AEE-788 may offer a better chance of successful treatment of pediatric astrocytomas. Advances in recent years in genomic and molecular research and a better understanding of the pathogenesis of astrocytomas will undoubtedly lead to better treatments in the near future. Acknowledgments No sources of funding were used to assist in the preparation of this review. Dr Burzynski is the President and Chairman of the Board of Directors of Burzynski Research Institute, Inc., which owns licenses for ANP in the US, Canada, and Mexico; and is a President of the Burzynski Clinic.

References 1. NCI. SEER cancer statistics review 1975–2001 [online]. Available from URL: http://seer.cancer.gov [Accessed 2006 May 19] 2. Bleyer WA. Epidemiologic impact of children with brain tumors. Childs Nerv Syst 1999; 15: 758-63 3. CBTRUS. Statistical report: primary brain tumors in the United States, 1998–2002. Hindsale (IL): Central Brain Tumor Registry of the US (publishing assistance was provided by the Stromberg Allen Company), 2005 4. Kleihues P, Cavenee W. Pathology and genetics of tumors of the central nervous system. Lyon: International Agency for Research on Cancer, 1997 5. Reddy AT. Advances in biology and treatment of childhood brain tumors. Curr Neurol Neurosci Rep 2001; 1: 137-43 6. Zacharoulis S, Kieran MW. Treatment of low-grade gliomas in children: an update. Expert Rev Neurother 2004; 4: 1005-14 7. Reddy AT, Wellens JC. Pediatric high grade gliomas. In: Markert JM, editor. Glioblastoma multiforme. Boston (MA): Jones and Bartlett Publishers, 2005 8. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352: 987-96 9. DeAngelis LM. Chemotherapy for brain tumors: a new beginning. N Engl J Med 2005; 352: 1036-8  2006 Adis Data Information BV. All rights reserved.

175

10. Lashford LS, Thiesse P, Jouvet A, et al. Temozolomide in malignant gliomas of childhood: a United Kingdom Children’s Cancer Study Group and French Society for Pediatric Oncology Intergroup Study. J Clin Oncol 2002; 20: 468491 11. Estlin EJ, Lashford L, Ablett S, et al. Phase I study of temozolomide in paediatric patients with advanced cancer: United Kingdom Children’s Cancer Study Group. Br J Cancer 1998; 78: 652-61 12. Burzynski SR. Targeted therapy for brain tumors. In: Columbus F, editor. Brain cancer research: progress. New York: Nova Science Publishers Inc, 2005 13. Lau CC. Genomic profiling in pediatric brain tumors. Cancer J 2005; 11: 283-93 14. Rubin JB, Gutmann DH. Neurofibromatosis type 1: a model for nervous system tumour formation? Nat Rev Cancer 2005; 5: 557-64 15. Pollack IF. Brain tumors in children. N Engl J Med 1994; 331: 1500-7 16. Korf BR. Diagnosis and management of neurofibromatosis type 1. Curr Neurol Neurosci Rep 2001; 1: 162-7 17. Zizka J, Elias P, Jakubec J. Spontaneous regression of low-grade astrocytomas: an underrecognized condition? Eur Radiol 2001; 11: 2638-40 18. Shaw EG, Wisoff JH. Prospective clinical trials of intracranial low-grade glioma in adults and children. Neuro-oncol 2003; 5: 153-60 19. Pollack IF, Pang D, Albright AL. The long-term outcome in children with lateonset aqueductal stenosis resulting from benign intrinsic tectal tumors. J Neurosurg 1994; 80: 681-8 20. Bowers DC, Georgiades C, Aronson LJ, et al. Tectal gliomas: natural history of an indolent lesion in pediatric patients. Pediatr Neurosurg 2000; 32: 24-9 21. Fisher BJ, Leighton CC, Vujovic O, et al. Results of a policy of surveillance alone after surgical management of pediatric low grade gliomas. Int J Radiat Oncol Biol Phys 2001; 51: 704-10 22. Kilic T, Ozduman K, Elmaci I, et al. Effect of surgery on tumor progression and malignant degeneration in hemispheric diffuse low-grade astrocytomas. J Clin Neurosci 2002; 9: 549-52 23. Saran FH, Baumert BG, Khoo VS, et al. Stereotactically guided conformal radiotherapy for progressive low-grade gliomas of childhood. Int J Radiat Oncol Biol Phys 2002; 53: 43-51 24. Merchant TE, Zhu Y, Thompson SJ, et al. Preliminary results from a phase II trial of conformal radiation therapy for pediatric patients with localised low-grade astrocytoma and ependymoma. Int J Radiat Oncol Biol Phys 2002; 52: 325-32 25. Marcus KJ. Stereotactic radiotherapy for localized LGGs in children: final results of a prospective trial. Int J Radiat Oncol Biol Phys 2005; 61: 374-9 26. Freeman CR, Bourgouin PM, Sanford RA, et al. Long term survivors of childhood brain stem gliomas treated with hyperfractionated radiotherapy: clinical characteristics and treatment related toxicities. The Pediatric Oncology Group. Cancer 1996; 77: 555-62 27. Lesser GJ. Chemotherapy of low-grade gliomas. Semin Radiat Oncol 2001; 11: 138-44 28. Massimino M, Spreafico F, Cefalo G, et al. High response rate to cisplatin/ etoposide regimen in childhood low-grade glioma. J Clin Oncol 2002; 20: 4209-16 29. Rosenstock JG, Packer RJ, Bilaniuk L, et al. Chiasmatic optic glioma treated with chemotherapy: a preliminary report. J Neurosurg 1985; 63: 862-6 30. Lefkowitz IB, Packer RJ, Sutton LN, et al. Results of the treatment of children with recurrent gliomas with lomustine and vincristine. Cancer 1988; 61: 896-902 31. Bouffet E, Goldman S, Jakacki R, et al. Pilot study of vinblastine in patients with recurrent and refractory low-grade glioma [abstract]. Neuro-oncol 2004; 6: 450 32. Gururangan S, Allen JC, Phillips PC, et al. Phase II study of oral temozolomide (TMZ) in children with progressive low-grade gliomas (LGG) [abstract]. Neuro-oncol 2004; 6: 457 33. Quinn JA, Reardon DA, Friedman AH, et al. Phase II trial of temozolomide in patients with progressive low-grade glioma. J Clin Oncol 2003; 21: 646-51 34. Kuo DJ, Weiner HL, Wisoff J, et al. Temozolomide is active in childhood, progressive, unresectable, low-grade gliomas. J Pediatr Hematol Oncol 2003; 25: 372-8 35. Walter AW, Gajjar A, Reardon DA, et al. Tamoxifen and carboplatin for children with low-grade gliomas: a pilot study at St. Jude Children’s Research Hospital. J Pediatr Hematol Oncol 2000; 22: 247-51 36. Packer RJ, Ater J, Allen J, et al. Carboplatin and vincristine chemotherapy for children with newly diagnosed progressive low-grade gliomas. J Neurosurg 1997; 86: 747-54 37. Pons MA, Finlay JL, Walker RW, et al. Chemotherapy with vincristine (VCR) and etoposide (VP-16) in children with low-grade astrocytoma. J Neurooncol 1992; 14: 151-8 Pediatr Drugs 2006; 8 (3)


176

38. Petronio J, Edwards MS, Prados M, et al. Management of chiasmal and hypothalamic gliomas of infancy and childhood with chemotherapy. J Neurosurg 1991; 74: 701-8 39. Burzynski SR. Clinical application of body epigenetic system: multi-targeted therapy for primary brain tumors. World and Ehrlich Conference on Dosing of Magic Bullets; 2004 Sep 9-11; N¨urnberg 40. Lafay-Cousin L, Holm S, Qaddoumi I, et al. Weekly vinblastine in pediatric lowgrade glioma patients with carboplatin allergic reaction. Cancer 2005; 103: 2636-42 41. Edwards MS, Cogen PH. Craniospinal neoplasms, child neurology: a clinical manual. 2nd ed. Philadelphia (PA): JB Lippincott, 1994: 165-83 42. Dosoretz DE, Blitzer PH, Wang CC, et al. Management of glioma of the optic nerve and/or chiasm: an analysis of 20 cases. Cancer 1980; 45: 1467-71 43. Tenny RT, Laws Jr ER, Younge BR, et al. The neurosurgical management of optic glioma: results in 104 patients. J Neurosurg 1982; 57: 452-8 44. Janss AJ, Grundy R, Cnaan A, et al. Optic pathway and hypothalamic/chiasmatic gliomas in children younger than age 5 years with a 6-year follow-up. Cancer 1995; 75: 1051-9 45. Lewis RA, Gerson LP, Axelson KA, et al. von Recklinghausen neurofibromatosis: II. Incidence of optic gliomata. Ophthalmology 1984; 91: 929-35 46. Riccardi VM. Type 1 neurofibromatosis and the pediatric patient. Curr Probl Pediatr 1992; 22: 66-106 47. Wisoff JH, Abbott R, Epstein F. Surgical management of exophytic chiasmatichypothalamic tumors of childhood. J Neurosurg 1990; 73: 661-7 48. Grabenbauer GG, Schuchardt U, Buchfelder M, et al. Radiation therapy of opticohypothalamic gliomas (OHG): radiographic response, vision and late toxicity. Radiother Oncol 2000; 54: 239-45 49. Kortmann RD, Timmermann B, Taylor RE, et al. Current and future strategies in radiotherapy of childhood low-grade glioma of the brain: part I. Treatment modalities of radiation therapy. Strahlenther Onkol 2003; 179: 509-20 50. Packer RJ, Sutton LN, Bilaniuk LT, et al. Treatment of chiasmatic/hypothalamic gliomas of childhood with chemotherapy: an update. Ann Neurol 1988; 23: 7985 51. Gnekow AK, Kortmann RD, Pietsch T, et al. Low grade chiasmatic-hypothalamic glioma-carboplatin and vincristin chemotherapy effectively defers radiotherapy within a comprehensive treatment strategy: report from the multicenter treatment study for children and adolescents with a low grade glioma: HIT-LGG 1996. Society of Pediatric Oncology and Hematology (GPOH). Klin Padiatr 2004; 216: 331-42 52. Burzynski SR. Methods for treating neurofibromatosis. U.S. Patent No. 5391575 (issued 1995) 53. Geyer JR. Gliomas in the very young child. In: Berger MS, Wilson CB, editors. The gliomas. Philadelphia (PA): WB Saunders Co, 1999 54. Marchese MJ, Chang CH. Malignant astrocytic gliomas in children. Cancer 1990; 65: 2771-8 55. Conway PD, Oechler HW, Kun LE, et al. Importance of histologic condition and treatment of pediatric cerebellar astrocytoma. Cancer 1991; 67: 2772-5 56. Wisoff JH, Boyett JM, Berger MS, et al. Current neurosurgical management and the impact of the extent of resection in the treatment of malignant gliomas of childhood: a report of the Children’s Cancer Group trial no. CCG-945. J Neurosurg 1998; 89: 52-9 57. Heideman RL, Kuttesch Jr J, Gajjar AJ, et al. Supratentorial malignant gliomas in childhood: a single institution perspective. Cancer 1997; 80: 497-504 58. Packer RJ, Boyett JM, Zimmerman RA, et al. Outcome of children with brain stem gliomas after treatment with 7800 cGy of hyperfractionated radiotherapy: a Children’s Cancer Group phase I/II trial. Cancer 1994; 74: 1827-34 59. Hodgson DC, Goumnerova LC, Loeffler JS, et al. Radiosurgery in the management of pediatric brain tumors. Int J Radiat Oncol Biol Phys 2001; 50: 929-35 60. Dropcho EJ, Wisoff JH, Walker RW, et al. Supratentorial malignant gliomas in childhood: a review of fifty cases. Ann Neurol 1987; 22: 355-64 61. Sposto R, Ertel IJ, Jenkin RDT, et al. The effectiveness of chemotherapy for treatment of high grade astrocytoma in children: results of a randomized trial. A report from the Children’s Cancer Study Group. J Neurooncol 1989; 7: 165-77 62. Finlay JL, Boyett JM, Yates AJ, et al. Randomized phase III trial in childhood high-grade astrocytoma comparing vincristine, lomustine, and prednisone with the eight-drugs-in-1-day regimen: Children’s Cancer Group. J Clin Oncol 1995; 13: 112-23 63. Boyett J, Yates A, Gilles F, et al. When is a high-grade astrocytoma (HGA) not a HGA? Results of a central review of 226 cases of anaplastic astrocytoma (AA),  2006 Adis Data Information BV. All rights reserved.

Burzynski

64.

65.

66.

67.

68.

69.

70.

71.

72. 73. 74. 75. 76.

77.

78.

79.

80. 81. 82.

83.

84.

85.

glioblastoma multiforme (GBM), and other HGA (OTH-HGA) by five neuropathologists [abstract]. Proc Soc Clin Oncol 1998; 17: 526 Chintagumpala M, Burger P, McCluggage CW. DNA mismatch repair and 06alkyguanine-DNA alkyltransferase (AGT analysis and response to procarbazine in malignant glioma in children: a Pediatric Oncology Group (POG) study [abstract]. Med Pediatr Oncol 1999; 22: 159 Chintagumpala M, Steward C, Burger P. Response to topotecan in newly diagnosed patients with high-grade gliomas: a Pediatric Oncology Group (POG) study. 9th International Symposium on Pediatric Neuro-Oncology; 2000 Jun 11-14; San Francisco (CA) Arenson E, Ater J, Bank J, et al. A randomized phase II trial of high dose alkylating agents plus VP-16 in children with high-grade astrocytoma [abstract]. J Pediatr Hematol Oncol 1999; 21: 325 Jakacki RI, Siffert J, Jamison C, et al. Dose-intensive, time-compressed procarbazine, CCNU, vincristine (PCV) with peripheral blood stem cell support and concurrent radiation in patients with newly diagnosed high-grade gliomas. J Neurooncol 1999; 44: 77-83 Huncharek M, Wheeler L, McGarry R, et al. Chemotherapy response rates in recurrent/progressive pediatric glioma: results of a systematic review. Anticancer Res 1999; 19: 3569-74 Nicholson HS, Krailo M, Ames MM, et al. Phase I study of temozolomide in children and adolescents with recurrent solid tumors: a report from the Children’s Cancer Group. J Clin Oncol 1998; 16: 3037-43 Korones DN, Smith AA, Bouffet E, et al. Temozolomide and oral VP-16 for children and young adults with recurrent or secondary malignant brain tumors [abstract]. Neuro-oncol 2004; 6: 460 Broniscer A, Chintagumpala M, Bowers DC, et al. Upfront protracted irinotecan (CPT-11) followed by radiotherapy (RT) and temozolomide (TMZ) in the treatment of children with newly diagnosed high-grade glioma (HGG) and unfavorable low-grade glioma (LGG): results of a multi-institutional study (SJHG-98) [abstract]. Neuro-oncol 2004; 6: 385 Freeman CR, Farmer JP. Pediatric brain stem gliomas: a review. Int J Radiat Oncol Biol Phys 1998; 40: 265-71 Pollack IF, Shultz B, Mulvihill JJ. The management of brainstem gliomas in patients with neurofibromatosis 1. Neurology 1996; 46: 1652-60 Guillamo JS, Doz F, Delattre JY. Brain stem gliomas. Curr Opin Neurol 2001; 14: 711-5 Cheng Y, Ng HK, Zhang SF, et al. Genetic alterations in pediatric high-grade astrocytomas. Hum Pathol 1999; 30: 1284-90 Louis DN, Rubio MP, Correa KM, et al. Molecular genetics of pediatric brain stem gliomas: application of PCR techniques to small and archival brain tumor specimens. J Neuropathol Exp Neurol 1993; 52: 507-15 Gilbertson RJ, Hill DA, Hernan R, et al. ERBB1 is amplified and overexpressed in high-grade diffusely infiltrative pediatric brain stem glioma. Clin Cancer Res 2003; 9: 3620-4 Mandell LR, Kadota R, Freeman C, et al. There is no role for hyperfractionated radiotherapy in the management of children with newly diagnosed diffuse intrinsic brainstem tumors: results of a Pediatric Oncology Group phase III trial comparing conventional vs. hyperfractionated radiotherapy. Int J Radiat Oncol Biol Phys 1999; 43: 959-64 Broniscer A, Gajjar A. Supratentorial high-grade astrocytoma and diffuse brainstem glioma: two challenges for the pediatric oncologist. Oncologist 2004; 9: 197-206 Allen J, Siffert J, Donahue B, et al. A phase I/II study of carboplatin combined with hyperfractionated radiotherapy for brainstem gliomas. Cancer 1999; 86: 1064-9 Freeman CR, Perilongo G. Chemotherapy for brain stem gliomas. Childs Nerv Syst 1999; 15: 545-53 Freeman CR, Kepner J, Kun LE, et al. A detrimental effect of a combined chemotherapy-radiotherapy approach in children with diffuse intrinsic brain stem gliomas? Int J Radiat Oncol Biol Phys 2000; 47: 561-4 Broniscer A, Leite CC, Lanchote VL, et al. Radiation therapy and high-dose tamoxifen in the treatment of patients with diffuse brainstem gliomas: results of a Brazilian cooperative study. Brainstem Glioma Cooperative Group. J Clin Oncol 2000; 18: 1246-53 Jennings MT, Sposto R, Boyett JM, et al. Preradiation chemotherapy in primary high-risk brainstem tumors: phase II study CCG-9941 of the Children’s Cancer Group. J Clin Oncol 2002; 20: 3431-7 Massimino M, Gandola L, Spreafico F, et al. Intrinsic brain stem tumor: changing strategies, changing results? [abstract]. Neuro-oncol 2003; 5: 51 Pediatr Drugs 2006; 8 (3)


Treatments for Astrocytic Tumors

86. Broniscer A, Iacono L, Chintagumpala M, et al. Role of temozolomide after radiotherapy for newly diagnosed diffuse brainstem glioma in children: results of a multiinstitutional study (SJHG-98). Cancer 2005; 103: 133-9 87. Bouffet E, Raquin M, Doz F, et al. Radiotherapy followed by high dose busulfan and thiotepa: a prospective assessment of high dose chemotherapy in children with diffuse pontine gliomas. Cancer 2000; 88: 685-92 88. Burzynski SR, Weaver RA, Janicki T. Long-term survival in phase II studies of antineoplastons A10 and AS2-1 (ANP) in patients with diffuse intrinsic brain stem glioma [abstract]. Neuro-oncol 2004; 6: 386 89. Burzynski SR, Weaver RA, Janicki TJ, et al. Targeted therapy with ANP in children less than 4 years old with inoperable brain stem gliomas [abstract]. Neuro-oncol 2005; 7: 300 90. Burzynski SR, Lewy RI, Weaver RA, et al. Phase II study of antineoplaston A10 and AS2-1 in patients with recurrent diffuse intrinsic brain stem glioma: a preliminary report. Drugs R D 2003; 4: 91-101 91. Burzynski SR, Weaver RA, Janicki T, et al. Targeted therapy with antineoplastons A10 and AS2-1 (ANP) of high-grade, recurrent and progressive brain stem glioma. Integr Cancer Ther 2006 Mar; 5 (1): 40-7 92. Sheline GE, Wara WM, Smith V. Therapeutic irradiation and brain injury. Int J Radiat Oncol Biol Phys 1980; 6: 1215-28 93. Nelson DF, Curran Jr WJ, Scott C, et al. Hyperfractionated radiation therapy and bis-chlorethyl nitrosourea in the treatment of malignant glioma: possible advantage observed at 72.0 Gy in 1.2 Gy B.I.D. fractions. Report of the Radiation Therapy Oncology Group Protocol 8302. Int J Radiat Oncol Biol Phys 1993; 25: 193-207 94. Marks JE, Baglan RJ, Prassad SC, et al. Cerebral radionecrosis: incidence and risk in relation to dose, time, fractionation and volume. Int J Radiat Oncol Biol Phys 1981; 7: 243-52 95. Bleyer WA, Griffin TW. White matter necrosis, microangiopathy and intellectual abilities in survivors of childhood leukemia: association with central nervous system irradiation and methotrexate therapy. In: Gilbert HA, Kagan AR, editors. Radiation damage to the nervous system. New York: Raven, 1980 96. Lee YY, Nauert C, Glass JP. Treatment-related white matter changes in cancer patients. Cancer 1986; 57: 1473-82 97. Styopoulos LA, George AE, de Leon MJ. Longitudinal CT study of parenchymal brain changes in glioma survivors. AJNR Am J Neuroradiol 1988 May-Jun; 9 (3): 517-22 98. Parsons JT, Bova FJ, Fitzgerald CR, et al. Radiation optic neuropathy after megavoltage external-beam irradiation: analysis of time-dose factors. Int J Radiat Oncol Biol Phys 1994; 30: 755-63 99. Goldsmith BJ, Rosenthal SA, Wara WM, et al. Optic neuropathy after irradiation of meningioma. Radiology 1992; 185: 71-6 100. Cavazzuti V, Winston K, Baker R, et al. Psychological changes following surgery for tumors in the temporal lobe. J Neurosurg 1980; 53: 618-26 101. Riva D, Pantaleoni C, Milani N, et al. Impairment of neuropsychological functions in children with medulloblastomas and astrocytomas in the posterior fossa. Childs Nerv Syst 1989; 5: 107-10 102. Carpentieri S, Mulhern RK, Douglas SM. Behavioral resiliency among children surviving brain tumors: a longitudinal study. J Clin Child Psychol 1994; 22: 236-46 103. Mostow EN, Byrne J, Connelly RR, et al. Quality of life in long-term survivors of CNS tumors of childhood and adolescence. J Clin Oncol 1991; 9: 592-9 104. Bloom HJ, Wallace EN, Henk JM. The treatment and prognosis of medulloblastoma in children: a study of 82 verified cases. Am J Roentgenol Radium Ther Nucl Med 1969; 105: 43-62 105. Shalet SM. Irradiation-induced growth failure. Clin Endocrinol Metab 1986; 15: 591-606 106. Park TS, Hoffman HJ, Hendrick EB, et al. Medulloblastoma: clinical presentation and management. Experience at the Hospital for Sick Children, Toronto, 1950–1980. J Neurosurg 1983; 58: 543-52 107. Haider A, Cullen JW, Ellerbeck JA. Pituitary function in children and adolescents following treatment for glial tumors. J Pediatr Oncol 1989; 3: 205-11 108. Oberfield SE, Allen JC, Pollack J, et al. Long-term endocrine sequelae after treatment of medulloblastoma: prospective study of growth and thyroid function. J Pediatr 1986; 108: 219-23 109. Rappaport R, Brauner R. Growth and endocrine disorders secondary to cranial irradiation. Pediatr Res 1989; 25: 561-7 110. Mack EE. Radiation-induced tumors. In: Berger MS, Wilson CB, editors. The gliomas. Philadelphia (PA): WB Saunders, 1999  2006 Adis Data Information BV. All rights reserved.

177

111. Meadows AT, D’Angio GJ, Mike V, et al. Patterns of second malignant neoplasms in children. Cancer 1977; 40: 1903-11 112. Neglia JP, Meadows AT, Robison LL, et al. Second neoplasms after acute lymphoblastic leukemia in childhood. N Engl J Med 1991; 325: 1330-6 113. Kingston JE, Hawkins MM, Draper GJ, et al. Patterns of multiple primary tumours in patients treated for cancer during childhood. Br J Cancer 1987; 56: 331-8 114. Hawkins MM, Draper GJ, Kingston JE. Incidence of second primary tumours among childhood cancer survivors. Br J Cancer 1987; 56: 339-47 115. Duffner PK, Horowitz ME, Krischer JP, et al. Postoperative chemotherapy and delayed radiation in children less than three years of age with malignant brain tumors. N Engl J Med 1993; 328: 1725-31 116. De Vita VT, Carbone PP, Owens AHJ. Clinical trials with 1.3-bis(2-chloroethyl)1-nitrosourea, NSC-409962. Cancer Res 1965; 25: 1876-81 117. Kumar L, Dua H. Cis-platin induced anaemia [letter]. N Z Med J 1987; 100: 81 118. Calvert AH, Newell DR, Gore ME. Future directions with carboplatin: can therapeutic monitoring, high-dose administration, and hematologic support with growth factors expand the spectrum compared with cisplatin? Semin Oncol 1992; 19: 155-63 119. Rivkees SA, Crawford JD. The relationship of gonadal activity and chemotherapyinduced gonadal damage. JAMA 1988; 259: 2123-5 120. Parvinen LM. Early effects of procarbazine (N-isopropyl-L-(2-methylhydrazino)p-toluamide hydrochloride) on rat spermatogenesis. Exp Mol Pathol 1979; 30: 1-11 121. Roth BJ, Einhorn LH, Greist A. Long-term complications of cisplatin-based chemotherapy for testis cancer. Semin Oncol 1988; 15: 345-50 122. Clayton PE, Shalet SM, Price DA, et al. Ovarian function following chemotherapy for childhood brain tumours. Med Pediatr Oncol 1989; 17: 92-6 123. Schilsky RL, Sherins RJ, Hubbard SM, et al. Long-term follow up of ovarian function in women treated with MOPP chemotherapy for Hodgkin’s disease. Am J Med 1981; 71: 552-6 124. Goren MP, Wright RK, Horowitz ME. Cumulative renal tubular damage associated with cisplatin nephrotoxicity. Cancer Chemother Pharmacol 1986; 18: 69-73 125. Schact RG, Baldwin DS. Chronic interstitial nephritis and renal failure due to nitrosourea therapy [letter]. Kidney Int 1978; 14: 661 126. Dobyan DC, Levi J, Jacobs C, et al. Mechanism of cis-platinum nephrotoxicity: II. Morphologic observations. J Pharmacol Exp Ther 1980; 213: 551-6 127. Holoye PY, Jenkins DE, Greenberg SD. Pulmonary toxicity in long-term administration of BCNU. Cancer Treat Rep 1976; 60: 1691-4 128. Jones SE, Moore M, Blank N, et al. Hypersensitivity to procarbazine (Matulane) manifested by fever and pleuropulmonary reaction. Cancer 1972; 29: 498-500 129. Garbes ID, Henderson ES, Gomez GA, et al. Procarbazine-induced interstitial pneumonitis with a normal chest x-ray: a case report. Med Pediatr Oncol 1986; 14: 238-41 130. Grimson BS, Mahaley Jr MS, Dubey HD, et al. Ophthalmic and central nervous system complications following intracarotid BCNU (carmustine). J Clin Neuroophthalmol 1981; 1: 261-4 131. Shingleton BJ, Bienfang DC, Albert DM, et al. Ocular toxicity associated with high-dose carmustine. Arch Ophthalmol 1982; 100: 1766-72 132. Mahaley Jr MS, Whaley RA, Blue M, et al. Central neurotoxicity following intracarotid BCNU chemotherapy for malignant gliomas. J Neurooncol 1986; 3: 297-314 133. Kleinschmidt-DeMasters BK. Intracarotid BCNU leukoencephalopathy. Cancer 1986; 57: 1276-80 134. Rosenblum MK, Delattre JY, Walker RW, et al. Fatal necrotizing encephalopathy complicating treatment of malignant gliomas with intra-arterial BCNU and irradiation: a pathological study. J Neurooncol 1989; 7: 269-81 135. Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (second of two parts). N Engl J Med 1974; 291: 127-33 136. Roca E, Bruera E, Politi PM, et al. Vinca alkaloid-induced cardiovascular autonomic neuropathy. Cancer Treat Rep 1985; 69: 149-51 137. von Hoff DD, Schilksy R, Reichert CM. Toxic effects of cisdiamminedichloroplatinum (II) in man. Cancer Treat Rep 1979; 63: 1527-31 138. Hansen SW, Helweg-Larsen S, Trojaborg W. Long-term neurotoxicity in patients treated with cisplatin, vinblastine, and bleomycin for metastatic germ cell cancer. J Clin Oncol 1989; 7: 1457-61 139. Grunberg SM, Sonka S, Stevenson LL, et al. Progressive paresthesias after cessation of therapy with very high-dose cisplatin. Cancer Chemother Pharmacol 1989; 25: 62-4 140. Schaefer SD, Post JD, Close LG, et al. Ototoxicity of low- and moderate-dose cisplatin. Cancer 1985; 56: 1934-9 Pediatr Drugs 2006; 8 (3)


178

141. Mulhern RK, Ochs JJ, Kun LE. Changes in intellect associated with cranial radiation therapy. In: Gutin PH, Leibel SA, Sheline GE, editors. Radiation injury to the nervous system. New York: Raven, 1991 142. Greene MH, Boice Jr JD, Strike TA. Carmustine as a cause of acute nonlymphocytic leukemia [letter]. N Engl J Med 1985; 313: 579 143. Cohen RJ, Wiernik PH, Walker MD. Acute nonlymphocytic leukemia associated with nitrosourea chemotherapy: report of two cases. Cancer Treat Rep 1976; 60: 1257-61 144. Pogliani EM, Pioltelli P, Rossini F, et al. Acute leukaemia following cisplatin for ovarian cancer. Haematologica 1987; 72: 184-5 145. Bassett WB, Weiss RB. Acute leukemia following cisplatin for bladder cancer [letter]. J Clin Oncol 1986; 4: 614 146. Des Guetz G. The 41st Annual ASCO Meeting: targeted therapies, Orlando, Florida. Targ Oncol 2006; 1: 59-65 147. Burzynski SR. Annual report to the FDA, IND 43,742, 2006 148. Zhou YH, Hess KR, Liu L, et al. Modeling prognosis for patients with malignant astrocytic gliomas: quantifying the expression of multiple genetic markers and clinical variables. Neuro-oncol 2005; 7: 485-94 149. Bast Jr RC, Hortobagyi GN. Individualized care for patients with cancer: a work in progress. N Engl J Med 2004; 351: 2865-7 150. Cohen KJ, Aronson LJ. Is there justification for the continued use of vincristine in pediatric brain tumor treatment? [abstract]. Neuro-oncol 2004; 6: 451

 2006 Adis Data Information BV. All rights reserved.

Burzynski

151. Oeffinger KC, Hudson MM. Long-term complications following childhood and adolescent cancer: foundations for providing risk-based health care for survivors. CA Cancer J Clin 2004; 54: 208-36 152. Bhatia S, Landier W. Evaluating survivors of pediatric cancer. Cancer J 2005; 11: 340-54 153. Leibel SA, Gutin PH, Wara WM, et al. Survival and quality of life after interstitial implantation of removable high-activity iodine-125 sources for the treatment of patients with recurring malignant gliomas. Int J Radiat Oncol Biol Phys 1989; 17: 1129-39 154. Wen PY, Alexander III E, Black PM, et al. Long term results of stereotactic brachytherapy used in the initial treatment of patients with glioblastomas. Cancer 1994; 73: 3029-36 155. Pollack I. Advances in the management of childhood gliomas: molecular markers of prognosis and novel treatment approaches [abstract]. Neuro-oncol 2005; 7: 348

Correspondence and offprints: Dr Stanislaw R. Burzynski, Burzynski Clinic, 9432 Old Katy Road, Houston, TX 77055, USA. E-mail: srb@burzynskiclinic.com

Pediatr Drugs 2006; 8 (3)



Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.