Antineoplaston Therapy

Number: 0240

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses antineoplaston therapy.

  1. Experimental and Investigational

    Aetna considers the following interventions experimental and investigational because the effectiveness of these approaches has not been established:

    1. Antineoplaston therapy (auto-urine therapy);
    2. The following services associated with antineoplaston therapy (not an all-inclusive list):
      1. Ancillary diagnostic laboratory, x-rays, MRI or CT scans done to monitor antineoplaston therapy;
      2. Infusion pump and intravenous supplies for use with the infusion pump;
      3. Placement of Hickman catheter.
    3. Oral antineoplaston therapy or associated physician services for administering and monitoring oral antineoplaston treatment,
  2. Related Policies

    • For phenylbutyrate, Buphenyl, and Pheburane, see Pharmacy Clinical Policy Bulletin: phenylbutyrate-Buphenyl-Pheburane 2121-A SGM.

      Note: Refer to pharmacy benefit plan for sodium phenylbutyrate and taurursodiol (Relyvrio).

Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

Antineoplaston therapy - no specific code:

ICD-10 codes not covered for indications listed in the CPB (not all-inclusive):

C00.0 - D49.9 Neoplasms
G12.21 Amyotrophic lateral sclerosis
Z51.0 Encounter for antineoplastic radiation therapy
Z51.11 - Z51.12 Encounter for antineoplastic chemotherapy and immunotherapy

Background

Antineoplastons are a group of naturally occurring peptides, which have been hypothesized to have anti-tumor activity.  Antineoplaston treatment is offered by the Burzynski Research Institute in Houston, Texas, and has long been a controversial treatment for various types of malignancy.

Antineoplaston therapy is not approved by the Food and Drug Administration (FDA) for any indication, and there are no controlled, peer-reviewed clinical trials to validate the effectiveness of antineoplaston therapy for any indication.

Primitive neuroectodermal tumors (PNETs) are often treated with cranio-spinal radiation and chemotherapy.  However, difficulties with conventional therapies can be encountered in very young children, in adult patients at high-risk of complication from standard treatment, as well as in patients with recurrent tumors.  In a phase II clinical trial, Burzynski et al (2005) studied the effect of antineoplaston (ANP) therapy in 13 children, either with recurrent disease or high-risk (median age of 5 years and 7 months, with a range of 1 to 11 years).  Medulloblastoma was diagnosed in 8 patients, pineoblastoma in 3 patients, and other PNET in 2 patients.  Prior therapies included surgery in 12 patients (1 had biopsy only, suboccipital craniotomy), chemotherapy in 6 patients, and radiation therapy in 6 patients.  Six patients had not received chemotherapy or radiation.  The treatment consisted of intravenous infusions of 2 formulations of ANP, A10 and AS2-1, and was administered for an average of 20 months.  The average dosage of A10 was 10.3 g/kg/day and of AS2-1 was 0.38 g/kg/day.  Complete response was accomplished in 23 %, partial response in 8 %, stable disease in 31 %, and progressive disease in 38 % of cases.  Six patients (46 %) survived more than 5 years from initiation of ANP; 5 were not treated earlier with radiation therapy or chemotherapy.  The serious side effects included single occurrences of fever, anemia, and granulocytopenia.  These investigators noted that the percentage of patients' response is lower than for standard treatment of favorable PNET, but long-term survival in poor-risk cases and reduced toxicity makes ANP therapy promising for very young children, patients at high-risk of complication of standard therapy, and patients with recurrent tumors.

In an open label, non- blinded, randomized, phase II clinical trial, Ogata and colleagues (2015) compared the effectiveness of hepatic arterial infusion (HAI) with 5-fluorouracil,with or without antineoplastons as a post-operative therapy for colorectal metastasis to the liver.  A total of 65 patients with histologically confirmed metastatic colon adenocarcinoma in liver, who had undergone hepatectomy, and/or thermal ablation for liver metastases were enrolled between 1998 to 2004.  Patients were randomly assigned to receive systemic antineoplastons (A10-I infusion followed by per-oral AS2-1) plus HAI (AN arm) or HAI alone (control arm) based on the number of metastases and presence/absence of extra-hepatic metastasis at the time of surgery.  Primary end-point was cancer-specific survival (CSS); secondary end-points were relapse-free survival (RFS), status and extent of recurrence, salvage surgery (rate) and toxicity.  Overall survival was not statistically improved (p = 0.105) in the AN arm (n = 32).  Relapse-free survival was not significant (p = 0.343).  Nevertheless, the CSS rate was significantly higher in the AN arm versus the control arm (n = 33) with a median survival time 67 months (95 % confidence interval [CI]: 43 to not calculated) versus 39 months (95 % CI: 28 to 47) (p = 0.037) and 5 year CSS rate 60 % versus 32 % respectively.  Cancer recurred more often in a single organ than in multiple organs in the AN arm versus the control arm.  The limited extent of recurrent tumors in the AN arm meant more patients remained eligible for salvage surgery.  Major adverse effects of antineoplastons were fullness of the stomach and phlebitis.  No serious toxicity, including bone marrow suppression, liver or renal dysfunction, were found in the AN arm.  The authors concluded that antineoplastons (A10 Injection and AS2-1) might be useful as adjunctive therapy in addition to HAI after hepatectomy in colorectal metastases to the liver.


References

The above policy is based on the following references:

  1. Aboumanei MH, Abdelbary AA, Ibrahim IT, et al. Design and development of microemulsion systems of a new antineoplaston A10 analog for enhanced intravenous antitumor activity: In vitro characterization, molecular docking, 125I-radiolabeling and in vivo biodistribution studies. Int J Pharm. 2018a;545(1-2):240-253.
  2. Aboumanei MH, Abdelbary AA, Ibrahim IT, et al. Improved targeting and tumor retention of a newly synthesized antineoplaston A10 derivative by intratumoral administration: Molecular docking, technetium 99m radiolabeling, and in vivo biodistribution studies. Cancer Biother Radiopharm. 2018b;33(6):221-232.
  3. Andratschke N, Grosu AL, Molls M, Nieder C. Perspectives in the treatment of malignant gliomas in adults. Anticancer Res. 2001;21(5):3541-3550.
  4. Badria F, Mabed M, El-Awadi M, et al. Immune modulatory potentials of antineoplaston A-10 in breast cancer patients. Cancer Lett. 2000;157(1):57-63. 
  5. BC Cancer Agency. Unconventional Therapies - Antineoplastons. Patient/Public Information. Vancouver, BC: BC Cancer Agency; revised February 2000. Available at: http://www.bccancer.bc.ca/PPI/UnconventionalTherapies/Antineoplastons.htm. Accessed September 30, 2002. 
  6. Boivin AJ, Momparler LF, Hurtubise A, Momparler RL. Antineoplastic action of 5-aza-2'-deoxycytidine and phenylbutyrate on human lung carcinoma cells. Anticancer Drugs. 2002;13(8):869-874.
  7. Buckner JC, Malkin MG, Reed E, et al. Phase II study of antineoplastons A1O (NSC 648539) and AS2-1 (NSC 620261) in patients with recurrent glioma. Mayo Clinic Proc. 1999;74(2):137-145. 
  8. Burzynski Clinic [website]. Houston, TX: Burzynski Clinic; 2001. Available at: http://www.cancermed.com/. Accessed December 6, 2022. 
  9. Burzynski SR, Janicki TJ, Weaver RA, Burzynski B. Targeted therapy with antineoplastons A10 and AS2-1 of high-grade, recurrent, and progressive brainstem glioma. Integr Cancer Ther. 2006;5(1):40-47.
  10. 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(2):91-101.
  11. Burzynski SR, Weaver RA, Janicki T, et al. Long-term survival of high-risk pediatric patients with primitive neuroectodermal tumors treated with antineoplastons A10 and AS2-1. Integr Cancer Ther. 2005;4(2):168-177.
  12. Burzynski SR, Weaver RA, Lewy RI, et al. Phase II study of antineoplaston A10 and AS2-1 in children with recurrent and progressive multicentric glioma: A preliminary report. Drugs R D. 2004;5(6):315-326.
  13. Burzynski SR. Potential of antineoplastons in diseases of old age. Drugs Aging. 1995;7(3):157-167. 
  14. Burzynski SR. Stanislaw R. Burzynski, MD, PhD: Novel cancer research and the fight to prove its worth. Altern Ther Health Med. 2012;18(3):54-61.
  15. Burzynski SR. The present state of antineoplaston research. Integr Cancer Ther. 2004;3(1):47-58.
  16. Burzynski SR. Treatments for astrocytic tumors in children: Current and emerging strategies. Paediatr Drugs. 2006;8(3):167-178.
  17. Choi BG. Synthesis of antineoplaston A10 as potential antitumor agents. Arch Pharm Res. 1998;21(2):157-163. 
  18. Congress of the United States, Office of Technology Assessment. Unconventional Cancer Treatments. OTA-H-405. Washington, DC: U.S. Government Printing Office; September 1990. 
  19. Fujii T, Yokoyama G, Takahashi H, et al. Preclinical studies of molecular-targeting diagnostic and therapeutic strategies against breast cancer. Breast Cancer. 2008;15(1):73-78.
  20. Green S. 'Antineoplastons' An unproved cancer therapy. JAMA. 1992;267:2924-2928. 
  21. Huang Y, Horvath CM, Waxman S. Regrowth of 5-fluorouracil-treated human colon cancer cells is prevented by the combination of interferon gamma, indomethacin, and phenylbutyrate. Cancer Res. 2000;60(12):3200-3206.
  22. Huang Y, Waxman S. Enhanced growth inhibition and differentiation of fluorodeoxyuridine-treated human colon carcinoma cells by phenylbutyrate. Clin Cancer Res. 1998;4(10):2503-2509.
  23. Juszkiewicz M, Chodkowska A, Burzynski SR, et al. The influence of antineoplaston A5 on particular subtypes of central dopaminergic receptors. Drugs Exp Clin Res. 1995;21(4):153-156. 
  24. Kumabe T. Antineoplaston treatment for advanced hepatocellular carcinoma. Oncology Rep. 1998;5(6):1363-1367. 
  25. Lea MA, Randolph VM, Hodge SK. Induction of histone acetylation and growth regulation in eryrthroleukemia cells by 4-phenylbutyrate and structural analogs. Anticancer Res. 1999;19(3A):1971-1976.
  26. Melichar B, Ferrandina G, Verschraegen CF, et al. Growth inhibitory effects of aromatic fatty acids on ovarian tumor cell lines. Clin Cancer Res. 1998;4(12):3069-3076.
  27. National Cancer Institute (NCI). Antineoplastons (PDQ®) - Health professional version. NCI [website]. Bethesda, MD: NCI; updated August 15, 2019.
  28. Nieder C, Nestle U. A review of current and future treatment strategies for malignant astrocytomas in adults. Strahlenther Onkol. 2000;176(6):251-258.
  29. Ogata Y, Matono K, Tsuda H, et al. Randomized phase II study of 5-fluorouracil hepatic arterial infusion with or without antineoplastons as an adjuvant therapy after hepatectomy for liver metastases from colorectal cancer. PLoS One. 2015;10(3):e0120064.
  30. Perrine SP. Fetal globin stimulant therapies in the beta-hemoglobinopathies: Principles and current potential. Pediatr Ann. 2008;37(5):339-346.
  31. Pili R, Kruszewski MP, Hager BW, et al. Combination of phenylbutyrate and 13-cis retinoic acid inhibits prostate tumor growth and angiogenesis. Cancer Res. 2001;61(4):1477-1485.
  32. Pineau T, Hudgins WR, Liu L, et al. Activation of a human peroxisome proliferator-activated receptor by the antitumor agent phenylacetate and its analogs. Biochem Pharmacol. 1996;52(4):659-667.
  33. Prasanna P, Shack S, Wilson VL, Samid D. Phenylacetate in chemoprevention: in vitro and in vivo suppression of 5-aza-2'-deoxycytidine-induced carcinogenesis. Clin Cancer Res. 1995;1(8):865-871.
  34. Reynolds S, Cederberg H, Chakrabarty S. Inhibitory effect of 1-O (2 methoxy) hexadecyl glycerol and phenylbutyrate on the malignant properties of human prostate cancer cells. Clin Exp Metastasis. 2000;18(4):309-312.
  35. Shack S, Miller A, Liu L, et al. Vulnerability of multidrug-resistant tumor cells to the aromatic fatty acids phenylacetate and phenylbutyrate. Clin Cancer Res. 1996;2(5):865-872.
  36. Soltysiak-Pawluczuk D, Burzynski SR. Cellular accumulation of antineoplaston AS21 in human hepatoma cells. Cancer Lett. 1995;88(1):107-112. 
  37. Sugita Y, Tsuda H, Maruiwa H, et al. The effects of Antineoplaston, a new antitumor agent on malignant brain tumors. Kurume Med J. 1995;42(3):133-140. 
  38. Svechnikova I, Gray SG, Kundrotiene J, et al. Apoptosis and tumor remission in liver tumor xenografts by 4-phenylbutyrate. Int J Oncol. 2003;22(3):579-588.
  39. Tsuda H, Hara H, Eriguchi N, et al. Toxicological study on antineoplastons A-10 and AS2-1 in cancer patients. Kurume Med. J 1995;42(4):241-249. 
  40. Tsuda H, Iemura A, Sata M, et al. Inhibitory effect of antineoplaston A10 and AS2-1 on human hepatocellular carcinoma. Kurume Med J. 1996;43(2):137-147. 
  41. Tsuda H, Sata M, Ijuuin H, et al. A novel strategy for remission induction and maintenance in cancer therapy.  Oncol Rep. 2002;9(1):65-68.
  42. Tsuda H, Sata M, Kumabe T, et al. Quick response of advanced cancer to chemoradiation therapy with antineoplastons. Oncology Rep. 1998;5(3):597-600. 
  43. Tweddle S, James N. Lessons from antineoplaston. Lancet. 1997;349(9063):1481. 
  44. Walczak J, Wood H, Wilding G, et al. Prostate cancer prevention strategies using antiproliferative or differentiating agents. Urology. 2001;57(4 Suppl 1):81-85.
  45. Wang J, Saunthararajah Y, Redner RL, Liu JM. Inhibitors of histone deacetylase relieve ETO-mediated repression and induce differentiation of AML1-ETO leukemia cells. Cancer Res. 1999;59(12):2766-2769.
  46. Wargovich MJ, Jimenez A, McKee K, et al. Efficacy of potential chemopreventive agents on rat colon aberrant crypt formation and progression. Carcinogenesis. 2000;21(6):1149-1155.
  47. Warrell RP Jr, He LZ, Richon V, et al. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst. 1998;90(21):1621-1625.
  48. Wirth B, Brichta L, Hahnen E. Spinal muscular atrophy and therapeutic prospects. Prog Mol Subcell Biol. 2006;44:109-132. 
  49. Wong D, Cederbaum S, Crombez E. Arginase deficiency. GeneReviews. Seattle, WA: University of Washington, Seattle; August 2014. Available at: http://www.ncbi.nlm.nih.gov/books/NBK1159/. Accessed February 26, 2021.
  50. Yu KH, Weng LJ, Fu S, et al. Augmentation of phenylbutyrate-induced differentiation of myeloid leukemia cells using all-trans retinoic acid. Leukemia. 1999;13(8):1258-1265.