Elsevier

Free Radical Biology and Medicine

Volume 99, October 2016, Pages 451-462
Free Radical Biology and Medicine

Original article
Pharmacokinetic and anti-cancer properties of high dose ascorbate in solid tumours of ascorbate-dependent mice

https://doi.org/10.1016/j.freeradbiomed.2016.08.027 Get rights and content

Highlights

  • Ascorbate elimination from solid tumours is markedly slower than from liver or plasma.

  • Daily administration of ascorbate was required to maintain optimal levels in tumours.

  • Optimal tumour ascorbate led to reduced angiogenesis, hypoxia and tumour growth.

Abstract

Despite recent evidence for an anti-tumour role for high-dose ascorbate, potential mechanisms of action are still unclear. At mM concentrations that are achieved with high-dose intravenous administration, autoxidation of ascorbate can generate cytotoxic levels of H2O2. Ascorbate is also a required co-factor for the hydroxylases that suppress the transcription factor hypoxia-inducible factor (HIF-1). HIF-1 supports an aggressive tumour phenotype and is associated with poor prognosis, and previous studies have shown that optimizing intracellular ascorbate levels down-regulates HIF-1 activation. In this study we have simultaneously measured ascorbate concentrations and the HIF-1 pathway activity in tumour tissue following high dose ascorbate administration, and have studied tumour growth and physiology. Gulo-/- mice, a model of the human ascorbate dependency condition, were implanted with syngeneic Lewis lung tumours, 1 g/kg ascorbate was administered into the peritoneum, and ascorbate concentrations were monitored in plasma, liver and tumours. Ascorbate levels peaked within 30 min, and although plasma and liver ascorbate returned to baseline within 16 h, tumour levels remained elevated for 48 h, possibly reflecting increased stability in the hypoxic tumour environment. The expression of HIF-1 and its target proteins was down-regulated with tumour ascorbate uptake. Elevated tumour ascorbate levels could be maintained with daily administration, and HIF-1 and vascular endothelial growth factor protein levels were reduced in these conditions. Increased tumour ascorbate was associated with slowed tumour growth, reduced tumour microvessel density and decreased hypoxia. Alternate day administration of ascorbate resulted in lower tumour levels and did not consistently decrease HIF-1 pathway activity. Levels of sodium-dependent vitamin C transporters 1 and 2 were not clearly associated with ascorbate accumulation by murine tumour cells in vitro or in vivo. Our results support the suppression of the hypoxic response by ascorbate as a plausible mechanism of action of its anti-tumour activity, and this may be useful in a clinical setting.

Introduction

Ascorbate has long been proposed to have anti-cancer activity [1], [2], [3], [4], [5], [6], and is frequently administered by complementary medicine practitioners as an adjunct to cancer treatments, often as high-dose infusions [7]. In the past decade, improved understanding of the pharmacokinetics of ascorbate uptake and transport has led to renewed interest in its potential activity in cancer. Recent investigations have shown that high-dose ascorbate can affect tumour growth in mouse models [8], [9], [10], [11], and there are currently a number of human clinical trials aiming to determine efficacy [12], [13], [14], [15], [16]. However, the use of ascorbate in cancer remains an area of controversy, due to a lack of robust clinical data and of a proven mechanism of action.

Ascorbate could exert a number of possible anti-tumour activities: at high local concentrations, achievable by intravenous administration, autoxidation results in significant generation of H2O2 that is cytotoxic to tumour cells in vitro [17], [8], [18]. High concentrations of dehydroascorbate (DHA), the oxidized form of ascorbate, are proposed to lead to an energetic crisis in glycolytic cancer cells, causing cell death [11]. Ascorbate regulates the activity of the transcription factor hypoxia-inducible factor (HIF)-1 [19], [20], [21] and decreased intracellular levels up-regulate the hypoxic response [22], [23], [24]. HIF-1 drives tumour angiogenesis, modifies the extracellular pH-environment, and affects cell death and survival pathways, and its activation culminates in an aggressive cancer phenotype [25], [26], [27].

HIF-1 is controlled by proline hydroxylases (PHDs 1, 2, 3) and an asparagine hydroxylase (factor-inhibiting HIF, FIH), collectively known as the HIF hydroxylases, that together regulate the stability of the alpha subunit and transcriptional activity [28], [29], [30]. The HIF hydroxylases are iron-containing 2-oxoglutarate-dependent dioxygenases that require molecular oxygen and 2-oxoglutarate as substrates [20], [31]. They also require ascorbate as a cofactor, to retain optimum activity and prevent irreversible oxidation of the active site iron [20], [21]. We, and others, have shown that intracellular ascorbate availability is a major determinant in the regulation of HIF-1 activity in response to mild hypoxia, 2-oxoglutarate deprivation or metal poisoning with Co2+ or Ni2+ [22], [19].

How intravenous or high-dose administration of ascorbate affects tumour growth is unknown, with the mechanism of anti-tumour activity by ascorbate not having been monitored in an in vivo setting. This information is required to inform human clinical study protocols. Pharmacokinetic studies in humans have demonstrated that the uptake of ascorbate following oral ingestion is tightly regulated [32], [33], and that the bioavailability is increased by 70-fold when administered intravenously [7], [33], [14]. We have previously proposed that these supra-physiological plasma concentrations could allow ascorbate to overcome the diffusion barrier of poorly-perfused solid tumours [34], thereby boosting intracellular levels. To date, however, no study has monitored the pharmacokinetics of ascorbate uptake into tumour tissue following high dose ascorbate (high-dose vitamin C, HDVC) administration.

Uptake of ascorbate into cells varies between tissues, and is mediated by the sodium-dependent vitamin C transporter that exists as two isoforms, SVCT1 and SVCT2 [35]. The SVCTs show distinct tissue distribution, and together ensure the effective uptake and regulation of plasma and cellular ascorbate concentrations [36], [35]. SVCT1 is responsible for uptake through the gut and re-absorption in the kidneys, and is thought to largely regulate plasma levels, whereas SVCT2 is concentrated in more metabolically active cells throughout the body, ensuring an adequate intracellular supply to support crucial intracellular functions [36], [35]. Both SVCT1 and SVCT2 have been found in lung tissue, with SVCT1 being identified solely in the blood vessels in one study [37], and on the apical surfaces of rat lung columnar epithelial cells in another study [52], both reporting widespread distribution of SVCT2. Transporter status has not been studied in tumour tissue, and this, together with ascorbate availability and tumour vessel patency, is likely to play an important role in the anti-cancer activity of HDVC.

The present study was designed to investigate whether HDVC could affect ascorbate levels in pre-existing tumours and could impact on tumour growth, and whether increased tumour ascorbate levels affected HIF-1 activity. We have utilized the Gulo-/- mouse which, like humans, carries a mutation in the gulonolactone oxidase (Gulo) gene that encodes the terminal enzyme in the ascorbate synthesis pathway, and thus relies entirely on dietary ascorbate [38]. Using this model, we have previously reported that variable dietary ascorbate intake affects tumour ascorbate levels, HIF-1 expression and tumour growth rate in three different syngeneic tumour models, namely Lewis Lung carcinoma (LL/2), B16-F10 melanoma and CMT93 colorectal cancer [39], [40]. All three tumour models showed a significant inverse association between tumour ascorbate content and HIF-1 levels. In the current study we have utilized the LL/2 model to specifically investigate the pharmacokinetics of ascorbate uptake into established tumours following HDVC, its ability to boost ascorbate levels in the tumour and in non-tumour tissue, and the effect on HIF-1 activity and subsequent tumour growth.

Section snippets

Materials

Mouse Lewis lung carcinoma (LL/2), CMT-93 colorectal carcinoma and B16-F10 melanoma cells were from American Type Culture Collection, Cryosite Distribution, Australia; the control mouse mammary carcinoma cell line EO771 was kindly provided by Dr Andreas Moeller, QIMR Berghofer, Australia. Sodium L-ascorbate was from Sigma-Aldrich, St Louis, MO, USA.Pimonidazole was obtained from Hypoxyprobe™-1, Burlington, Massachusetts, USA. Antibodies against mouse HIF-1α, carbonic-anhydrase IX (CA-IX), and

Ascorbate transporter status of murine cell lines in vitro

In our previous studies with three different tumour cell lines, LL/2, CMT-93 and B16-F10, we have shown that intracellular ascorbate levels affect HIF-1 activation both in vitro, and in vivo following dietary intervention in the Gulo-/- mouse [39], [40]. All three cell lines are able to accumulate ascorbate in vitro [40], and were shown to express the SVCT2 transporter, the isoform mostly responsible for uptake by metabolically active cells [35] (Fig. 1). LL/2 cells were also assayed for SVCT1

Discussion

We investigated the pharmacokinetic parameters of high dose ascorbate administration in plasma, liver and tumour tissue in the Gulo-/- mouse and have monitored the effect of tumour ascorbate uptake on the activation of HIF-1 and on tumour growth. This is the first study to monitor these processes together in established tumours following HDVC. We have shown that in response to a single dose of HDVC, there is a prolonged presence of ascorbate in the tumour, compared to plasma and liver. We also

Conclusion

This study has provided novel tumour-specific pharmacokinetic information on ascorbate uptake and accumulation. We have shown that ascorbate elimination from solid tumours is markedly slower than from plasma and liver. Furthermore, daily administration of HDVC was required to maintain optimal levels of ascorbate in tumours, and this led to reduced angiogenesis and reduced tumour hypoxia, resulting in a tumour growth delay. This mechanistic information has been lacking to date and will inform

Conflicts of interest

None.

Acknowledgements

This study was funded by the Health Research Council of New Zealand (11/460, MCMV, GUD), the Mackenzie Charitable Foundation (GUD) and the University of Otago (Ph.D Scholarships for EJC and CW).

References (56)

  • J.A. Raleigh et al.

    Importance of thiols in the reductive binding of 2-nitroimidazoles to macromolecules

    Biochem. Pharmacol.

    (1990)
  • J.E. Biaglow et al.

    The catalytic effect of the carcinogen "4-nitroquinoline-N-oxide" on the oxidation of vitamin C

    Biochem. Biophys. Res. Commun.

    (1976)
  • D. Fukumura et al.

    Tumor microvasculature and microenvironment: targets for anti-angiogenesis and normalization

    Microvasc. Res.

    (2007)
  • E. Cameron et al.

    Supplemental ascorbate in the supportive treatment of cancer: prolongation of survival times in terminal human cancer

    Proc. Natl. Acad. Sci. USA

    (1976)
  • E. Cameron et al.

    Supplemental ascorbate in the supportive treatment of cancer: reevaluation of prolongation of survival times in terminal human cancer

    Proc. Natl. Acad. Sci. USA

    (1978)
  • Q. Chen et al.

    Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues

    Proc. Natl. Acad. Sci. USA

    (2005)
  • S. Ohtani et al.

    Tumor suppressor 101F6 and ascorbate synergistically and selectively inhibit non-small cell lung cancer growth by caspase-independent apoptosis and autophagy

    Cancer Res.

    (2007)
  • M.K. Wilson et al.

    Review of high-dose intravenous vitamin C as an anticancer agent

    Asia Pac. J. Clin. Oncol.

    (2014)
  • S.J. Padayatty et al.

    Vitamin C: intravenous use by complementary and alternative medicine practitioners and adverse effects

    PLoS One

    (2010)
  • Q. Chen et al.

    Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice

    Proc. Natl. Acad. Sci. USA

    (2008)
  • C. Grasso et al.

    Pharmacological doses of daily ascorbate protect tumors from radiation damage after a single dose of radiation in an intracranial mouse glioma model

    Front. Oncol.

    (2014)
  • J. Yun et al.

    Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH

    Science

    (2015)
  • C. Jacobs et al.

    Is there a role for oral or intravenous ascorbate (vitamin C) in treating patients with cancer? A systematic review

    Oncologist

    (2015)
  • Y. Ma et al.

    High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy

    Sci. Transl. Med.

    (2014)
  • C.M. Stephenson et al.

    Phase I clinical trial to evaluate the safety, tolerability, and pharmacokinetics of high-dose intravenous ascorbic acid in patients with advanced cancer

    Cancer Chemother. Pharmacol.

    (2013)
  • J. van der Reest et al.

    Anti-cancer effects of vitamin C revisited

    Cell Res.

    (2016)
  • J.L. Welsh et al.

    Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): results from a phase I clinical trial

    Cancer Chemother. Pharmacol.

    (2013)
  • Q. Chen et al.

    Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo

    Proc. Natl. Acad. Sci. USA

    (2007)
  • Cited by (0)

    View full text