ISSN: 2755-0117 | Open Access

Journal of Oncology Research Reviews & Reports

Recurrent Medulloblastoma: Complete Response and > than 21 Years and Five Months Overall Survival in a One-Year and Seven- Month-Old Male Treated with Antineoplastons

Author(s): Stanislaw R Burzynski*, Gregory S Burzynski, Tomasz Janicki and Samuel Beenken

Abstract

Medulloblastomas arise in the posterior fossa, primarily in the region of the fourth ventricle. Recurrent medulloblastomas are high-risk and have a bad prognosis. Objectives: the case of a male child with recurrent, disseminated medulloblastoma is presented to detail the efficacy of ANP therapy (Antineoplaston A10 {Atengenal} and Antineoplaston AS2-1 {Astugenal}) in the treatment of high-risk medulloblastoma. This child was treated at the Burzynski Clinic (BC), as a Special Exception, according to Protocol BT-12, “Phase II Study of Antineoplastons A10 and AS2-1 in Children with Primitive Neuroectodermal Tumors”, with intravenous (IV) ANP therapy via a subclavian catheter and infusion pump. Tumor response was measured by sequential magnetic resonance imaging (MRI) of the brain with gadolinium enhancement. Findings: This child initially underwent tumor resection, placement of ventricular shunts, and chemotherapy, all performed elsewhere. Baseline MRI at the BC revealed recurrent, disseminated, and enhancing disease measuring 9.10 cm2 in total. IV ANP therapy began in March 2001 and ended in December 2002 after a complete response (CR) was achieved based on MRI criteria. Subsequently, the child received oral Antineoplastons as maintenance therapy, which were discontinued after eight months. At last follow-up, > 21 years and eight months since diagnosis and > 21 years and five months since the start of IV ANP therapy, the patient was well and showing no evidence of tumor recurrence. Conclusions: The utilization of ANP therapy to cure a patient with recurrent medulloblastoma is presented. We conclude that ANP therapy is an attractive therapeutic option for children with medulloblastoma.

Introduction

Embryonal tumors, while biologically heterogeneous, tend to disseminate along cerebrospinal fluid (CSF) pathways. Their diagnosis is based primarily on histological and immunohistological features. More recently, genetic studies are being utilized to subclassify embryonal tumors for the purposes of risk stratification and treatment planning [1-4]. Approximately five percent of children with medulloblastoma have germline mutations, which puts them at risk of developing other cancers [5, 6].

Medulloblastomas arise in the posterior fossa, with nearly 80% located in the region of the fourth ventricle. In children, early symptoms are often due to blockage of the flow of CSF and resultant hydrocephalus. Common signs and symptoms include headaches, nausea and/or vomiting, lethargy, ataxia, and nystagmus. Signs and symptoms of medulloblastoma in infants may include lethargy, delay in psychomotor development, difficulty feeding, and increased head circumference [7].

On examination, one may see bulging of the anterior fontanel due to increased intracranial pressure while the eyes may deviate downward secondary to the loss of upward gaze, which is due to compression of the tectum of the midbrain [7].

Most medulloblastomas display some degree of anaplasia on histological examination. These features include increased nuclear size, marked cytological pleomorphism, numerous mitoses, and apoptotic bodies. [8, 9] If anaplasia is diffuse (50% - 80% of the tumor) one can consider the medulloblastoma to be anaplastic.

Histological findings of desmoplasia may indicate an improved outcome [10, 11]. However, patient age, the extent of disease at the time of diagnosis, histologic factors, and treatment received are currently being supplemented by the analysis of genetic variations in determining prognosis [12, 13]. In addition, genetic variations are being used to assign treatment in prospective studies [14].

Patient age, radiographical evaluation for disseminated disease, the amount of residual tissue after surgery, and CSF cytological examination have been used to project outcome [8]. Children less than 3 - 5 years of age with no dissemination and ≤ 1.5 cm2 of residual disease are considered “average risk” while thosedisseminated disease and/or > 1.5 cm2 of residual disease are considered “high risk” [15].

We present here the successful use of ANP therapy (Antineoplaston A10 {Atengenal} and Antineoplaston AS2-1 {Astugenal}) in the treatment of a one-year and seven-month-old male child with recurrent and disseminated medulloblastoma (high-risk) measuring 9.1 cm2 in total size (see below) [8].

Materials and Methods

The one-year and seven-month-old male child presented here was evaluated at the Burzynski Clinic (BC) on March 6, 2001. He was accompanied by his father and mother. The patient had headaches, slight weakness of the left upper and lower extremities, and a delay in motor skills, being unable to walk or crawl.

This child was in good health until December of 2000, when he lagged behind his developmental milestones, and was seen by his pediatrician. A magnetic resonance imaging (MRI) scan of the brain on December 19, 2000, demonstrated a midline cerebellar mass measuring 4.5 cm x 4.5 cm x 5.0 cm and extensive hydrocephalus, all of which were consistent with a medulloblastoma. Nodules on the cerebellar surface provided evidence of local tumor spread. On December 19, 2000, the child underwent a right frontal ventriculostomy and on December 22, 2000, he had a posterior fossa craniotomy with removal of the tumor. Histological examination of the resected tumor confirmed the diagnosis of medulloblastoma. A spinal fluid tap was negative for malignant cells. On January 1, 2001, the child developed GI bleeding and subsequently underwent laparotomy, ligation of a bleeding vessel in a duodenal ulcer, pyloroplasty, and gastrostomy. On January 2, 2001, an MRI scan of the brain demonstrated no evidence of residual tumor while MRI of the spine showed no evidence of CSF spread. On January 9, 2001, the child underwent removal of the right frontal ventriculostomy and placement of a left frontal ventriculostomy. On January 23, 2001, he again had placement of a right frontal ventriculostomy while the left frontal ventriculostomy catheter was removed. On February 12, 2001, the child began vincristine and cyclophosphamide chemotherapy, but only received one treatment before the patient was admitted to the hospital, on February 14, 2001, due to sepsis. He was treated with IV antibiotics. Chemotherapy was not resumed.

An MRI scan of the brain and spine, performed on March 2, 2001 (Figure 1), showed areas of enhancement in the right tentorium and the right cerebellar hemisphere indicating tumor spread in the brain. There was no evidence of CSF spread to the spine.
At the BC, on March 13, 2001, this patient began intravenous (IV) ANP therapy, as a Special Exception (LPS = 50%, discontinued chemotherapy), according to Protocol BT-12, a “Phase II Study of Antineoplastons A10 and AS2-1 in Children with Primitive Neuroectodermal Tumors.” In this single arm study, IV ANP therapy was delivered every four hours via a subclavian catheter and a programmable infusion pump.
The objectives of BT-12 were to 1) determine the efficacy of ANP therapy in children with primitive neuroectodermal tumors as determined by an objective response (OR) to therapy; 2) determine OR utilizing MRI scans of the brain, which were performed every 8 weeks for the first two years, and then less frequently; and 3 ) determine the safety and tolerance of ANP therapy in this group of patients

Eligibility criteria for BT-12 included 1) histologically confirmed primitive neuroectodermal tumor; 2) evidence of persistent/ recurrent tumor as determined by gadolinium enhanced brain MRI performed within 2 weeks of study enrollment; 3 ) Tumor size ≥ 5mm; 4) Age of 6 months to 17 years; 5) Lansky Performance Status (LPS) ≥ 60%; and 6) Life expectancy ≥ 2 months.
Gadolinium enhanced MRI scans of the brain were used in the diagnosis and follow-up of the medulloblastomas. T2-weighted, T2-fluid attenuated inversion recovery (T2-FLAIR), T1 weighted and T1-weighted contrast-enhanced images were obtained. Medulloblastomas are gadolinium-enhancing, therefore sequential T1-weighted contrast-enhanced images were utilized to determine the effect of therapy [16].

As determined by MRI scan of the brain, the product of the two greatest perpendicular diameters of each measurable (≥ 5mm) and enhancing lesion was calculated. Tumor size was defined as the sum of these products. The response criteria were as follows: a complete response (CR) indicated complete disappearance of all enhancing tumor while a partial response (PR) indicated a 50% or greater reduction in total measurable and enhancing tumor size. CR and PR required a confirmatory brain MR scan performed at least four weeks after the initial OR. Progressive disease (PD) indicated a 25% or greater increase in total measurable and enhancing tumor size, or new measurable and enhancing disease, while stable disease (SD) did not meet the criteria for PR or PD [17].

Protocol BT-12 was conducted in accordance with the U.S. Code of Federal Regulations, Title 21, Parts 11, 50, 56 and 312; the Helsinki Declaration of 1975 (2000 revision); the Good Clinical Practices: Consolidated Guideline (E6), International Conference on Harmonization (ICH) and Guidance for Industry (FDA). By participating in this study protocol, the investigators agreed to provide access to all appropriate documents for monitoring, auditing, IRB review and review by any authorized regulatory agency. This Phase II study is described in Clinicaltrials.gov (CDR0000066492, NCT00003460).

Results/Observations

Between April 1966 and January 2005, 13 children were accrued to BT-12 and treated at the BC. Twelve patients were evaluable while one was not evaluable due to technical problems with the follow-up MRI scans of the brain. Median age was 6.1 years (range: 1.0 to 12.2 years). Ten children were male while 3 were female. Among the eight children with medulloblastoma, one child obtained a CR, one child maintained SD, and six children developed PD.

As previously discussed, the child presented here was evaluated at the BC for recurrent medulloblastoma following resection of a primary posterior fossa tumor and placement of ventricular shunts. This child was treated at the BC, as a Special Exception (LPS = 50%, discontinued chemotherapy), according to Protocol BT-12, a “Phase II Study of Antineoplastons A10 and AS2-1 in Children with Primitive Neuroectodermal Tumors”. In this single arm study, IV ANP therapy was delivered every four hours via a subclavian catheter and a programmable infusion pump.

The starting dose of A10 for this child was 0.83 g/kg/d. It was gradually increased to 17.33 g/kg/d and then reduced to 13.23 g/ kg/d. His starting dose of AS2-1 was 0.22 g/kg/d. It was gradually increased to 0.46 g/kg/d and then reduced to 0.36 g/kg/d. Upon completion of IV ANP therapy and achievement of a CR, the child received oral Antineoplastons as maintenance therapy, which was discontinued after eight months.

On September 18, 2001, a six-month follow-up MRI scan of the brain showed a 59.1% decrease in size of the recurrent medulloblastoma, indicating a PR. On June 3, 2002, one-year and 3-month follow-up brain MRI scan showed no evidence of residual enhancing disease, indicating a CR (Figure 1). On March 21, 2005, MRI scan of the brain showed no evidence of residual enhancing disease, indicating an enduring CR (Figure 1). At last follow-up, August 30, 2022, the patient was doing very well and had received no other anti-tumor therapy. OS has been > 21 years and eight months since diagnosis and > 21 years and five months since the start of ANP therapy. All MRIs of the brain showing an OR were reviewed by a prominent outside neuroradiologist.

img

Figure 1: Axial images from MRI brain scans performed before, during, and after ANP therapy: March 2, 2001 - Baseline MRI showing recurrent and disseminated medulloblastoma (see arrows); June 3, 2002 - One-year and 3-month follow-up MRI showing no evidence of residual enhancing disease, indicating a CR; March 21, 2005 - Follow-up MRI showing no evidence of residual enhancing disease, indicating an enduring CR. ANP therapy = Antineoplaston A10 (Atengenal) and Antineoplaston AS2-1 (Astugenal); MRI = magnetic resonance imaging; PR = partial response; CR = complete response.

Adverse events (AEs) were graded according to the Common Terminology Criteria for Adverse Events Version 3.0 (CTCAE v.3). Ten patients accrued to BT-12 experienced a serious adverse event (SAE). The child presented here developed two SAEs thought to be due to ANP therapy, hypernatremia and hypocalcemia, which occurred during a period of somnolence brought on by aspiration pneumonia. He recovered fully from the pneumonia, the somnolence, the hypernatremia, and the hypocalcemia.

Discussion

In the 2021 World Health Organization (WHO) classification, medulloblastoma is characterized according to histological and genetic findings [18]. If there is no genetic testing, the classification is medulloblastoma, histologically defined.

The treatment of younger children with newly diagnosed medulloblastoma is not fully standardized. The use of different drug regimens and the variable use of craniospinal and local boost radiation therapy (RT), in children ≥ 3 years of age, makes comparison of studies difficult. However certain treatment guidelines can be emphasized.
Total surgical resection of the tumor is the optimal initial treatment. Subsequently, adjuvant chemotherapy is frequently utilized, being more frequently based on genetic analysis, and includes drugs such as cyclophosphamide, etoposide, cisplatin, and vincristine, with or without concomitant high-dose intravenous and/or intraventricular methotrexate [19-21].

In infants and young children with desmoplastic medulloblastoma, medulloblastoma with extensive nodularity (MBEN), and tumors with sonic-hedgehog (SHH) pathway mutation signaling, high dose systemic chemotherapy and intraventricular methotrexate or high-dose systemic chemotherapy followed by stem cell rescue, without radiation, is effective treatment [22]. In children with localized disease after chemotherapy, focal RT does not improve survival [23]. Long-term survivors treated with chemotherapy are at a high risk of later hearing loss, cardiac complications, and secondary neoplasms. In infants and young children with a non-desmoplastic, non-MBEN, and non-SHH pathway mutation signaling medulloblastoma, high dose systemic chemotherapy and intraventricular methotrexate or high-dose systemic chemotherapy followed by stem cell rescue, without radiation, is not effective treatment for these children, who have a much worse prognosis [24]. There is no standard therapy in recurrent medulloblastoma.

We have presented the use of IV ANP therapy in a child diagnosed at age one-year and four-months with a recurrent medulloblastoma. He was treated with IV ANP therapy and achieved a CR after one-year and three-months of therapy, which, at the time of his last follow-up, had persisted for > 21 years and five months since the start of IV ANP therapy.

Antineoplaston (ANP) research began in 1967, when significant deficiencies were noticed in the peptide content of the serum of patients with cancer compared with healthy persons. Initially ANP were isolated from the blood and later from urine [25]. Subsequent studies of the isolated ANP demonstrated that Antineoplaston A-10 and Antineoplaston AS2-1 were the most active ANPs. The chemical name of Antineoplaston A-10 is 3-phenylacetylamino2,6-piperidinedione. It consists of the cyclic form of L-glutamine connected by a peptide bond to phenylacetyl residue. When given orally, Antineoplaston A10 resists the attack of gastric enzymes. In the small intestine, under alkaline conditions, 30% is digested into phenylacetylglutamine (PG) and phenylacetylisoglutaminate (isoPG) in a ratio of approximately 4:1. The mixture of synthetic PG and isoPG in a 4:1 ratio, dissolved in sterile water constitutes Antineoplaston A10 intravenous (IV) injection. Further metabolism of Antineoplaston A10 results in phenylacetate (PN). Bothmetabolites PG and PN have anticancer activity. The mixture of PN and PG in a 4:1 ratio, dissolved in sterile water constitutes Antineoplaston AS2-1 IV injection [26].

ANP therapy’s mechanism of action differs from that of RT or cytotoxic chemotherapy. Growth of normal cells is controlled by cell cycle progression genes (oncogenes) and by cell cycle arrest genes (tumor suppressor genes). In cancer, alteration of these control genes in malignant cells favors aggressive cell proliferation. Evidence suggests that ANP therapy affects 204 mutated genes in the malignant genome and functions as a “molecular switch” which “turns on” tumor-suppressor genes and “turns off” oncogenes [27, 28]. Hence, the antineoplastic action of ANP therapy in medulloblastoma involves restoration of cell cycle control, induction of programmed cell death, and interference with cancer cell metabolism and nuclear transport.

Conclusions

We have presented here the case of a male child with a high-risk recurrent and disseminated medulloblastoma who obtained a CR with ANP therapy, which indicates that ANP therapy may be an effective therapeutic option for children with medulloblastoma. Multiple Phase II clinical studies of ANP therapy in a variety of low- and high-grade brain tumors under the Burzynski Research Institute’s (BRI’s) IND # 43,742 have now been completed and numerous articles have been published [29-72]. Based on the CR described here, we propose a multi-institutional Phase II study of ANP therapy for children with medulloblastoma.

Acknowledgements

The authors express their appreciation to Carolyn Powers for preparation of the manuscript and to Ramiro Rivera, Mohamed Khan, Jennifer Pineda and Adam Golunski for their involvement.

References

  1. Pfister S, Remke M, Benner A, Mendrzyk F, Toedt G, et (2009) Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. J. Clin. Oncol 27:
  2. Taylor MD, Northcott PA, Korshunov A, Remke M, Yoon-Jae Cho, et al. (2012) Molecular subgroups of medulloblastoma: the current consensus. Acta. Neuropathol 123:
  3. Kool M, Koster J, Bunt J, Hasselt NE, Lakeman A, et (2008) Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological features. PLoS.One 3:
  4. Ellison DW, Onilude OE, Lindsey JC, Lusher ME, Weston CL, et al. (2005) beta-Catenin status predicts a favorable outcome in childhood medulloblastoma: the United Kingdom Children’s Cancer Study Group Brain Tumour J. Clin. Oncol 23:
  5. Zhang J, Walsh MF, Wu G, Edmonson MN, Gruber TA, et al. (2015) Germline Mutations in Predisposition Genes in Pediatric Cancer. N. Engl. J. Med 373:
  6. Waszak SM, Northcott PA, Buchhalter I, Robinson GW, Sutter C, et al. (2018) Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial Lancet Oncol 19:
  7. Ramaswamy V, Remke M, Shih D, Wang X, Northcott PA, et al. (2014) Duration of the pre-diagnostic interval in medulloblastoma is subgroup dependent. Pediatr. Blood Cancer 61:
  8. McManamy CS, Lamont JM, Taylor RE, Cole M, Andrew DJ, et al. (2003) Morphophenotypic variation predicts clinical behavior in childhood non-desmoplastic J. Neuropathol. Exp. Neurol 62:
  9. Eberhart C, Kratz J, Wang Y, Summers K, Stearns D, et (2004) Histopathological and molecular prognostic markers in medulloblastoma: c-myc, N-myc, TrkC, and anaplasia. Neuropathol. Exp. Neurol 63:
  10. Rutkowski S, Bode U, Deinlein F, Ottensmeier H, WarmuthMetz M, et al. (2009) Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. Engl. J. Med 352:
  11. Rutkowski S, Gerber NU, von Hoff K, Gnekow A, Bode U, et al. (2009) Treatment of early childhood medulloblastoma by postoperative chemotherapy and deferred Neuro. Oncol 11:
  12. Gajjar A, Robinson GW, Smith KS, Lin T, Merchant TE, et al. (2021) Outcomes by Clinical and Molecular Features in Children with Medulloblastoma Treated With Risk-Adapted Therapy: Results of an International Phase III Trial J. Clin. Oncol 39:
  13. Ramaswamy V, Remke M, Adamski J, Bartels U, Tabori U, et al. (2016) Medulloblastoma subgroup-specific outcomes in irradiated children: who are the true high-risk patients? Neuro. Oncol 18:
  14. Gottardo NG, Hansford JR, McGlade JP, Alvaro F, Ashley DM, et al. (2014) Medulloblastoma Down Under 2013: a report from the third annual meeting of the International Medulloblastoma Working Group. Acta. Neuropathol
  15. Zeltzer PM, Boyett JM, Finlay JL, Albright AL, Rorke LB, et al. (1999) Metastasis stage, adjuvant treatment, and residual tumor are prognostic factors for medulloblastoma in children: conclusions from the Children’s Cancer Group 921 randomized phase III study. J. Clin. Oncol 17:
  16. Shukla G, Alexander GS, Bakas S, Nikam R, Talekar K, et al. (2017) Advanced magnetic resonance imaging in glioblastoma: a review. Chinese. Clin. Oncol 6:
  17. Wen PK, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, et al. (2010) Updated response criteria for highgrade gliomas: Response Assessment in Neuro-Oncology (RANO) working group. J. Clin. Oncol 28:
  18. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, et (2021) The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro. Oncol 23:
  19. Rutkowski S, Bode U, Deinlein F, Ottensmeier H, WarmuthMetz M, et al. (2005) Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. Engl. J. Med 352:
  20. Geyer JR, Sposto R, Jennings M, Boyett JM, Axtell RA, et (2005) Multiagent chemotherapy and deferred radiotherapy in infants with malignant brain tumors: a report from the Children’s Cancer Group. J. Clin. Oncol 23:
  21. Duffner PK, Horowitz ME, Krischer JP, Friedman HS, Burger PC, et al. (1993) Postoperative chemotherapy and delayed radiation in children less than three years of age with malignant brain tumors. N. Engl. J. Med 328:
  22. Mynarek M, Hoff KV, Pietsch T, Ottensmeier H, WarmuthMetz M, et al. (2020) Nonmetastatic Medulloblastoma of Early Childhood: Results from the Prospective Clinical Trial HIT-2000 and An Extended Validation Cohort. J. Clin. Oncol 38:
  23. Salloum R, Chen Y, Yasui Y, Packer R, Leisenring W, et (2019) Late Morbidity and Mortality Among Medulloblastoma Survivors Diagnosed Across Three Decades: A Report from the Childhood Cancer Survivor Study. J. Clin. Oncol 37:
  24. Dhall G, O’Neil SH, Ji L, Haley K, Whitaker AM, et (2020) Excellent outcome of young children with nodular desmoplastic medulloblastoma treated on “Head Start” III: a multi-institutional, prospective clinical trial. Neuro. Oncol 22:
  25. Burzynski SR (1976) Antineoplastons: Biochemical defense against cancer. Physiol. Chem. Phys 8:
  26. Burzynski SR (1986) Synthetic antineoplastons and Drugs of the Future. 11:
  27. Burzynski SR, Patil S (2014) The effect of Antineoplaston A10 and AS2-1 and metabolites of sodium phenylbutyrate on gene expression in glioblastoma multiforme. J. Cancer Ther 5:
  28. Burzynski SR, Janicki T, Burzynski G (2015) Comprehensive genomic profiling of recurrent classic glioblastoma in a patient surviving eleven years following antineoplaston Cancer Clin. Oncol 4:
  29. Burzynski SR, Conde AB, Peters A, Saling B, Ellithorpe R, et al. (1999) A Retrospective Study of Antineoplastons A10 and AS2-1 in Primary Brain Tumors. Clin. Drug Invest 8:
  30. Burzynski SR, Weaver RA, Lewy RI, Janicki TJ, Jurida GF, et al. (2004) Phase II study of Antineoplaston A10 and AS2-1 in children with recurrent and progressive multicentric glioma: A preliminary report. Drugs R. & D 5:
  31. Burzynski SR, Lewy RI, Weaver R, Janicki T, Jurida G, et al. (2004) Long-term survival and complete response of a patient with recurrent diffuse intrinsic brain stem glioblastoma multiforme. Integ. Cancer Ther 3:
  32. Burzynski SR, Weaver R, Bestak M, Janicki T, Szymkowski B, et al. (2004) Treatment of primitive neuroectodermal tumors (PNET) with antineoplastons A10 and AS2-1 (ANP): Preliminary results of phase II studies. Neuro. Oncol 6:
  33. Burzynski SR, Weaver RA, Janicki, T, Szymkowski B, Jurida G, et al. (2005) Long-term survival of high-risk pediatric patients with primitive neuroectodermal tumors treated with Antineoplastons A10 and AS2-1. Integ. Cancer Ther 4: 168-
  34. Burzynski SR, Weaver R, Bestak M, Janicki T, Jurida G, et al. (2004) Phase II studies of antineoplastons A10 and AS2- 1 (ANP) in children with atypical teratoid/rhabdoid tumors (AT/RT) of the central nervous system: A preliminary Neuro. Oncol 6:
  35. Burzynski SR (2006) Targeted Therapy for Brain In: Yang AV, editor. Brain Cancer Therapy and Surgical Interventions. Nova Science Publishers, Inc, New York, 8:
  36. Burzynski SR, Janicki, TJ, Weaver RA, Burzynski B (2006) Targeted therapy with Antineoplastons A10 and AS2-1 of high grade, recurrent, and progressive brainstem Integ. Cancer Ther 5:
  37. Burzynski SR (2006) Treatments for astrocytic tumors in children: Current and emerging strategies. Ped. Drugs 8:
  38. Burzynski SR (2007) Recent clinical trials in diffuse intrinsic brainstem glioma. Cancer Ther 5: 379-
  39. Burzynski SR, Burzynski GS, Janicki TJ (2014) Recurrent glioblastoma multiforme: A strategy for long-term J. Cancer Ther 5:
  40. Burzynski SR, Janicki TJ, Burzynski GS, Marszalek A (2014) A phase II study of antineoplastons A10 and AS2-1 in children with high-grade glioma: Final report (Protocol BT-06) and review of recent trials. J. Cancer Ther 5:
  41. Burzynski SR, Janicki TJ, Burzynski GS (2014) A phase II study of antineoplastons A10 and AS2-1 in adult patients with recurrent glioblastoma multiforme: Final report (Protocol BT-21). J. Cancer Ther 5:
  42. Burzynski SR, Janicki TJ, Burzynski GS, Marszalek A, Brookman S (2014) A phase II study of antineoplastons A10 and AS2-1 in children with recurrent, refractory or progressive primary brain tumors: Final report (Protocol BT-22). J. Cancer Ther 5:
  43. Burzynski SR, Janicki TJ, Burzynski GS, Brookman S (2014) Preliminary findings on the use of targeted therapy with pazopanib and other agents in combination with sodium phenylbutyrate in the treatment of glioblastoma J. Cancer Ther 5:
  44. Burzynski GS, Janicki TJ, Marszalek A (2014) Long-term survival (> 20 years) of a child with brainstem glioma treated with antineoplastons A10 and AS2-1: A case report. Oncol 11:
  45. Burzynski SR, Janicki TJ, Burzynski GS, Marszalek (2014) The response and survival of children with recurrent diffuse intrinsic pontine glioma based on phase II study of antineoplastons A10 and AS2-1 in patients with brainstem glioma. Childs. Nerv. Syst 30:
  46. Burzynski S, Janicki T, Burzynski G, Marszalek A (2015) Long-term survival (>13 years) in a child with recurrent diffuse pontine gliosarcoma: A case report. J. Ped. Oncol 36:
  47. Burzynski SR, Janicki T, Burzynski G (2015) A phase II study of Antineoplastons A10 and AS in adult patients with primary brain tumors: Final report (Protocol BT-09). J. Cancer Ther 6:
  48. Burzynski SR, Burzynski G, Janicki J, Marszalek A (2015) Complete response and Long-term survival (> 20 years) of a child with tectal glioma: A case report. Pediatr. Neurosurg 50:
  49. Burzynski SR, Janicki TJ, Burzynski G (2015) A phase II study of Antineoplastons A10 and AS2-1 injections in adult patients with recurrent anaplastic astrocytoma: Final report (Protocol BT-15). Cancer Clin. Oncol 442:
  50. Burzynski SR, Janicki TJ, Burzynski GS, Marszalek A (2015) A Phase II Study of Antineoplastons A10 and AS2-1 in adult patients with newly-diagnosed anaplastic astrocytoma: Final report (Protocol BT-08). Cancer Clin. Oncol 4:
  51. Burzynski SR, Burzynski GS, Marszalek A, Janicki J, Martinez-Canca J (2015) Long-term survival (over 20 years), complete response and normal childhood development in medulloblastoma treated with Antineoplastons A10 and AS2- 1. J. Neurol. Stroke 2:
  52. Burzynski SR, Burzynski GS, Marszalek A, Janicki TJ, Martinez-Canca JF (2015) Long-term survival over 21 years and pathologically confirmed complete response in pediatric anaplastic astrocytoma: A case report. J. Neurol. Stroke 2:
  53. Burzynski SR, Burzynski GS, Brookman S (2015) A case of sustained objective response of recurrent/progressive diffuse intrinsic pontine glioma with phenylbutyrate and targeted agents. J. Cancer Ther 6:
  54. Burzynski SR, Janicki T, Burzynski G, Marszalek A (2015) A phase II study of antineoplastons A10 and AS2-1 in patients with brainstem gliomas: The report on non-diffuse intrinsic pontine glioma (Protocol BT-11). J. Cancer Ther 6:
  55. Burzynski S, Janicki TJ, Burzynski GS (2016) Primary CNS tumors and leptomeningeal, disseminated, and/or multicentric disease in children treated in phase II studies with antineoplastons A10 and AS2-1. Cancer Clin. Oncol 5:
  56. Burzynski SR, Janicki TJ, Burzynski GS (2016) A phase II study of antineoplastons A10 and AS2-1 in children with low-grade astrocytomas: Final report (Protocol BT-13). Cancer Ther 7:
  57. Burzynski SR, Janicki TJ, Burzynski GS, Marszalek A (2017) A phase II study of Antineoplastons A10 and AS2-1 in children with brain tumors: Final report (Protocol J. Cancer Ther 8:
  58. Burzynski SR, Janicki T, Beenken S (2019) Treatment of recurrent glioblastoma multiforme (rGBM) with Antineoplaston AS2-1 in combination with targeted Cancer Clin. Oncol 8:
  59. Burzynski SR, Janicki T, Burzynski GS, Beenken S (2021) Long-term survival (27.7 years) following IV Antineoplaston Therapy (ANP) in a 36-year-old-female with a progressive diffuse intrinsic pontine glioma (DIPG). Int. J. Imaging Technol 7:
  60. Burzynski SR, Burzynski GS, Janicki T, Beenken S (2021) Long-term survival (23 years) in a 26-year-old male after Antineoplaston therapy for a progressive, diffuse intrinsic pontine glioma: A case report. Int. J. Brain Disorder Treat 6:
  61. Burzynski SR, Janicki T, Burzynski GS, Beenken S (2021) Resolution of clinical signs, a complete response, and longterm survival (>23 Years) in a 3 and month female with a newly diagnosed diffuse intrinsic pontine glioma treated with antineoplastons. Biomed. Res. Clin. Prac 6:
  62. Burzynski SR, Janicki T, Burzynski GS, Beenken S (2021) Diffuse intrinsic pontine glioma in an 11-year-old female treated with antineoplastons: Complete response and > 25- year survival. Pediatr. Neonatal Med 1:
  63. Burzynski SR, Janicki T, Burzynski GS, Beenken S (2022) A 25-year-old female with diffuse intrinsic pontine glioma surviving for more than nine years following treatment with antineoplastons. Int. J. Clin. Oncol. Cancer Res 7:
  64. Burzynski SR, Burzynski GS, Janicki T, Beenken S (2022) Twenty-two-year survival in a 15-year-old female with a recurrent posterior fossa ependymoma treated with antineoplastons. Oncol. Clin. Res 3:
  65. Burzynski S, Burzynski G, Janicki T, Beenken S (2022) Recurrent and progressive ganglioglioma in an 11-year-old male treated with antineoplastons: Partial response with more than nine years and nine months survival and complete resolution of clinical symptoms/signs. Biomed. Res. J 37:
  66. Burzynski S, Burzynski G, Janicki T, Beenken S (2022) Newly diagnosed Multicentric Pilocytic Astrocytoma: Complete Response and > 22 Years Survival in a Six Year and Nine-month-old Female Treated with Internat. J. Clin. Oncol. Cancer Res 7:
  67. Burzynski S, Burzynski G, Janicki T, Beenken S (2022) Outcomes in Four Children with Persistent, Recurrent, and Progressive Gangliogliomas Treated in Phase II Studies with Antineoplastons A10 and AS2-1. Neurol. Neurosci 3:
  68. Burzynski S, Burzynski G, Janicki T, Beenken S (2022) Recurrent/Persistent Glioblastoma: Complete Response and 24 Years Disease-Free-Survival in a 45-Year-Old Female Treated with Antineoplastons. Cancer Stud. Ther 7:
  69. Burzynski S, Burzynski G, Janicki T, Beenken S (2022) Newly diagnosed Glioblastoma: Partial Response and > 27 Years Overall Survival in a 37-Year-Old Male Treated with Antineoplastons. Recent Adv. Clin. Trials 1:
  70. rzynski SR, Burzynski GS, Janicki TJ, Beenken SW (2023) Persistent pineoblastoma: Complete response and > 26 years overall survival in a ten-month-old female treated with antineoplastons. Biomed. Res. Clin. Prac 7:
  71. Burzynski S, Burzynski G, Janicki T, Beenken S (2023) Newly diagnosed Anaplastic Astrocytoma: > 23 Year Survival in a 31-Year and 11-month-Old Female treated with Antineoplastons. Neurol. Neurosci 4:
  72. Burzynski S, Burzynski G, Janicki T, Beenken S (2023) Inoperable Optic Pathway Glioma: A Seven-Year-Old Male with > 35 Years Overall Survival Following Treatment with Antineoplastons. Ej-clinicmed.org 4: 9-14.
View PDF