Elsevier

Medical Hypotheses

Volume 44, Issue 3, March 1995, Pages 207-213
Medical Hypotheses

Intravenous ascorbate as a tumor cytotoxic chemotherapeutic agent

https://doi.org/10.1016/0306-9877(95)90137-XGet rights and content

Abstract

Ascorbic acid and its salts (AA) are preferentially toxic to tumor cells in vitro and in vivo. Given in high enough doses to maintain plasma concentrations above levels that have been shown to be toxic to tumor cells in vitro, AA has the potential to selectively kill tumor cells in a manner similar to other tumor cytotoxic chemotherapeutic agents. Most studies of AA and cancer to date have not utilized high enough doses of AA to maintain tumor cytotoxic plasma concentrations of AA. Data are presented which demonstrate the ability to sustain plasma levels of AA in humans above levels which are toxic to tumor cells in vitro and suggests the feasibility of using AA as a cytotoxic chemotherapeutic agent.

References (57)

  • L. Benade et al.

    Synergistic killing of Ehrlich ascites carcinoma cells by ascorbate and 3-amino-I, 2, 4triazole

    Oncology

    (1969)
  • S. Bram et al.

    Vitamin C preferential toxicity for malignant melanoma cells

    Nature

    (1980)
  • V. Noto et al.

    Effects of sodium ascorbate (vitamin C) and 2-methyl-1, 4-naphthoquinone (vitamin K3) treatment on human tumor cell growth in vitro

    Cancer

    (1989)
  • J. Helgestad et al.

    Characterization of a new malignant human T -cell line (PFI-285) sensitive to ascorbic acid

    Eur J Haematol

    (1990)
  • C.H. Park et al.

    Growth suppression of human leukemic cells in vitro by L-ascorbic acid

    Cancer Res

    (1980)
  • K. Yamafuji et al.

    Anti-tumor potency of ascorbic, dehydroascorbic, or 2,3-diketogulonic acid and their action on deoxyribonucleic acid

    Z Krebsforsch

    (1971)
  • K. Yagashita et al.

    Effects of tetraacetyl-bis-dehydroAA, a derivative of ascorbic acid, on Ehrlich cells and HeLa cells (human carcinoma cells)

    J Nutr Sci Vitaminol

    (1976)
  • C.J. Koch et al.

    Toxicity, radiation sensitivity modification, and metabolic effects of dehydroascorbate and ascorbate in mammalian cells

    J Cell Physiol

    (1978)
  • M.H. Cohen et al.

    Cure of advanced Lewis lung carcinoma (LL): a new treatment strategy

  • A. Lupulesco

    Vitamin C inhibits DNA, RNA and protein synthesis in epithelial neoplastic cells

    Intl J Vit Nutr Res

    (1991)
  • H.F. Pierson et al.

    Sodium ascorbate enhancement of carbidopa-levodopa methyl ester antitumor activity against pigmented B-16 melanoma

    Cancer Res

    (1983)
  • R.N. Chakrabarti et al.

    Effects of ascorbic acid on survival and cell-mediated immunity in tumor bearing mice

    IRCS Med Sci

    (1984)
  • C.S. Tsao et al.

    In vivo antineoplastic activity of ascorbic acid for human mammary tumor

    In vivo

    (1988)
  • E. Cameron et al.

    Ascorbic acid and cancer: a review

    Cancer Res

    (1979)
  • R. Anderson

    Vitamin C and immune functions: mechanisms of immunostimulation

  • D.E. Henson et al.

    Ascorbic acid: biologic functions and relation to cancer: commentary

    JNCI

    (1991)
  • Cited by (107)

    • Starving Cancer Cells: Evidence-Based Strategies to Slow Cancer Progression: A Selection of Readings for Health Services Providers

      2021, Starving Cancer Cells: Evidence-Based Strategies to Slow Cancer Progression A Selection of Readings for Health Services Providers
    • O<inf>2</inf><sup>⋅−</sup> and H<inf>2</inf>O<inf>2</inf>-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate

      2017, Cancer Cell
      Citation Excerpt :

      Intravenous pharmacological doses of ascorbate have recently re-emerged as a potential anti-cancer therapy with clinical trials in ovarian and pancreatic cancer subjects demonstrating tolerability with similar or reduced toxicities, relative to chemotherapy alone (Ma et al., 2014; Monti et al., 2012; Welsh et al., 2013). Preclinical studies with ascorbate have consistently demonstrated cancer cell-selective cytotoxicity in a variety of disease sites (Du et al., 2010; Ma et al., 2014; Riordan et al., 1995). Although the mechanism(s) of selective toxicity remain unknown, mounting evidence suggests that ascorbate toxicity is dependent on the action of ascorbate as a pro-drug for hydrogen peroxide (H2O2) generation (Chen et al., 2005, 2007; Olney et al., 2013).

    • Liposome-based drug co-delivery systems in cancer cells

      2017, Materials Science and Engineering C
      Citation Excerpt :

      Vitamin C is a strong reducing agent that plays a role in various physiological functions [73–75]. Vitamin C not only displays cytotoxicity toward cancer cells but also improves the anti-neoplastic effect of some anti-cancer agents, in particular anthracyclines, because one of their cytotoxic effects is related to free radical production [76–80]. It was revealed that DOX and vitamin C have synergistic activity toward the breast carcinoma cells over a wide range of vitamin C concentrations [81].

    • Non-coding RNA/microRNA-modulatory dietary factors and natural products for improved cancer therapy and prevention: Alkaloids, organosulfur compounds, aliphatic carboxylic acids and water-soluble vitamins

      2016, Non-coding RNA Research
      Citation Excerpt :

      A clinical study of the Mayo Clinic used only oral vitamin C and, thus, could not reproduce the promising results of Cameron and coworkers which underlines the high importance of intravenous application of vitamin C in order to reach sufficient blood plasm concentrations of vitamin C (up to 5.5 mmol/L after 10 g vitamin C, i.v.; up to 13.5 mmol/L after 50 g vitamin C, i.v.) [156–158]. Several newer cancer case studies and trials that applied high-dose vitamin C infusions reported of significant tumor remission and reduced side-effects when combined with chemotherapeutic agents (e.g., paclitaxel) [159–164]. A phase I clinical trial of the combination of vitamin C and gemcitabine in metastatic pancreatic cancer patients (PACMAN study) revealed a mean survival time of 13 months, which was more than twice of the mean survival time of patients treated only with gemcitabine (5.65 months) [164,165].

    • Microbial influences on hormesis, oncogenesis, and therapy: A review of the literature

      2015, Environmental Research
      Citation Excerpt :

      Watson stated that antioxidants cause cancer, interferes with its treatments (which would make sense since many of the treatments induce higher concentrations of ROS), and that antioxidants actually promote the growth of late stage metastatic cancer (Watson, 2013). However, it could actually be the fact that some antioxidants such as vitamin C will act as pro-oxidants as well, depending on their environment (Casciari et al., 2001; Riordan et al., 1995). If that is the case than bacteria could be the key delivery method to producing lipopolysaccharides in the body that will both act as an antioxidant in healthy tissues while acting as a pro-oxidant in neoplasms.

    View all citing articles on Scopus
    View full text