Abstract
Vitamin C (ascorbic acid, ascorbate), despite controversy, has re-emerged as a promising anti-cancer agent. Recent knowledge of intravenous ascorbate pharmacokinetics and discovery of unexpected mechanisms of ascorbate action have spawned many investigations. Two mechanisms of anti-cancer activity with ascorbate have gained prominence: hydrogen peroxide induced oxidative stress and DNA demethylation mediated by ten-eleven translocation enzyme activation. Here, we highlight salient aspects of the evolution of ascorbate in cancer treatment; provide insights into the pharmacokinetics of ascorbate; describe mechanisms of its anti-cancer activity in relation to the pharmacokinetics; outline promising pre-clinical and clinical evidence; and recommend future directions.
Background
Like a phoenix, ascorbic acid as an anti-cancer agent has had a spectacular rise, fall and re-emergence. In the 1970s, Ewan Cameron, joined by Linus Pauling, described retrospectively and in case reports that patients with advanced cancer had survival benefit and symptomatic relief using high dose ascorbate (10 g/day) (Cameron and Campbell, 1974; Cameron and Pauling, 1976; Cameron and Pauling, 1978). Subsequently, two rigorous double-blind placebo-controlled prospective trials performed at the Mayo Clinic using the same dose of ascorbate failed to confirm these results, and oral ascorbate as an anti-cancer agent was roundly dismissed (Creagan et al., 1979; Moertel et al., 1985; Wittes, 1985).
Pharmacologic Ascorbate and Hydrogen Peroxide (H2O2)
About two decades ago, new knowledge of ascorbate pharmacokinetics spawned discovery of anti-cancer mechanisms of ascorbate action (Table 1). To provide a foundational basis for dietary recommendations for ascorbic acid, healthy subjects underwent intensive clinical pharmacokinetics and physiology studies, the first studies of this kind for any vitamin (Levine et al., 1996; Levine et al., 2001). Findings were that oral doses of ascorbic acid over an ~ 80-fold dose range produced plasma concentrations that were tightly controlled by limited gastrointestinal absorption, saturated tissue transporters, and renal reabsorption and excretion (Corpe et al., 2013; Corpe et al., 2010; Levine et al., 1996; Levine et al., 2001; Padayatty et al., 2004; Sotiriou et al., 2002). Intravenous (IV) administration bypassed tight control until the kidney restored homeostasis. Depending on dose and infusion time, IV ascorbate produced plasma levels that were hundreds of fold higher than those produced by the maximum tolerated dose of oral ascorbate (Chen et al., 2008; Hoffer et al., 2008; Padayatty and Levine, 2000; Padayatty et al., 2004; Park et al., 2009). These pharmacokinetics data provided a potential explanation to the conflicting cancer treatment outcomes, because no one at the time knew that IV and oral ascorbic acid behaved so differently. Cameron administered 10 grams ascorbate intravenously as well as orally, while Mayo investigators used oral ascorbate alone. With pharmacokinetics as a foundation, it was shown that only ascorbic acid at pharmacologic concentrations from intravenous dosing, and that would not occur from oral dosing, acted as pro-drug for hydrogen peroxide (H2O2) formation in the extracellular space (Chen et al., 2005; Chen et al., 2007). Pharmacologic, but not physiologic, ascorbic acid was selectively toxic to cancer cells in-vitro and in vivo (Chen et al., 2005; Chen et al., 2008; Verrax and Calderon, 2009). Additionally, the requisite pharmacologic ascorbic acid concentrations are achieved predictably and safely in humans (Chen et al., 2008; Hoffer et al., 2008; Monti et al., 2012; Padayatty et al., 2010; Welsh et al., 2013). In animal models, pharmacologic ascorbate has anti-cancer activity either similar to conventional chemotherapy or synergizes with it (Espey et al., 2011; Ma et al., 2014; Serrano et al., 2015; Xia et al., 2017). Conversely, there are no data showing that pharmacologic ascorbate interferes with chemotherapy. Early phase clinical trials indicate that IV ascorbate at 1g/kg over 90–120 minutes two to three times weekly is well tolerated and may enhance chemosensitivity as well as decrease chemotherapy related side effects.(Carr et al., 2014; Hoffer et al., 2015; Ma et al., 2014; Monti et al., 2012; Schoenfeld et al., 2017; Shim et al., 2014; Stephenson et al., 2013; Welsh et al., 2013).
Table 1:
Cameron/Pauling studies | Mayo Clinic studies | Vitamin C pharmacokinetics and early phase clinical trials | Studies on H2O2 mechanism of anti- cancer activity | Studies on epigenetic mechanism of anti-cancer activity |
---|---|---|---|---|
Ewan Cameron, joined by Linus Pauling, described retrospectively and in case reports that patients with advanced cancer had survival benefit and symptomatic relief using high dose ascorbate (10g/day IV followed by oral) (Cameron and Campbell, 1974; Cameron and Pauling, 1976; Cameron and Pauling, 1978) |
Two rigorous double-blind placebocontrolled prospective trials performed at the Mayo Clinic using the same dose of ascorbate, but orally only, failed to confirm these results, and oral ascorbate was dismissed as an anti-cancer agent. (Creagan et al., 1979; Moertel et al., 1985; Wittes, 1985) |
Oral ascorbic acid over an 80-fold dose range was found to produce plasma concentrations that were tightly regulated by gastrointestinal absorption, but IV administration bypassed this control until the kidney restored homeostasis. Maximum tolerated doses of oral (~18g daily) ascorbate produced plasma concentrations of ~100–200uM. Intravenous ascorbate was found to produce plasma levels hundreds of fold higher than those produced by the maximum tolerated dose of oral ascorbate. (Hoffer et al., 2008; Padayatty and Levine, 2000; Padayatty et al., 2004; Park et al., 2009) Early phase clinical trials indicate that IV ascorbate at 1g/kg over 1½–2 hours two to three times weekly is well tolerated and may enhance chemosensitivity as well as decrease chemotherapy related side effects. (Carr et al., 2014; Hoffer et al., 2015; Ma et al., 2014; Monti et al., 2012; Schoenfeld et al., 2017; Shim et al., 2014; Stephenson et al., 2013; Welsh et al., 2013). |
Plasma concentrations achieved by IV dosing found to act as a pro-drug for hydrogen peroxide (H2O2) in the extracellular space. Pharmacologic, but not physiologic, ascorbic acid was selectively toxic to cancer cells in-vitro and in vivo. (Chen et al., 2005; Chen et al., 2008; Verrax and Calderon., 2009; Espey et al., 2011; Ma et al., 2014; Serrano et al., 2015; Xia et al., 2017) |
Ascorbate functions as a cofactor and increases the activity of the Ten Eleven Translocation (TET) enzymes causing DNA demethylation. This function has been found to result in the re-expression of tumor suppressor genes in cancer cells, promotion of stem cell differentiation and inhibition of leukemogenesis, and enhancement of DNA methyltransferase inhibitor (DNMTi)-induced immune signals via increased expression of endogenous retrovirus transcripts. (Agathocleous et al., 2017; Cimmino et al., 2017; Liu et al., 2016; Shenoy et al., 2017, Gustafson et al., 2015). |
Ascorbate physiology and pharmacokinetics findings are the backbone of these observations. Nevertheless, extracellular H2O2 formation generated by pharmacologic ascorbate may not explain all pharmacology actions. As one example, recent findings implicate oxidized ascorbic acid, dehydroascorbic acid, as the mediator of cancer cell death in specific engineered cell lines (Yun et al., 2015). Although the conclusions are attractive, extracellular H2O2 generation from pharmacologic ascorbate could well account for the observations, a possibility not tested. Other investigators found no effects of dehydroascorbic acid on cancer cell using a variety of cell types (Chen et al., 2005; Ma et al., 2014; Schoenfeld et al., 2017). Clinically, dehydroascorbic acid currently would not be administered because of potential pancreatic toxicity (Patterson and Lazarow, 1950). As another example, we should not forget Ewan Cameron’s cases. Prior to publications with Linus Pauling, Cameron reported a case series where unexpected tumor responses were observed, using intravenous ascorbate at 10 grams daily for 7–10 days followed by oral ascorbate at 10 grams daily (Cameron and Campbell, 1974). There were substantial limitations, including a Scottish patient population at substantial risk for severe vitamin C deficiency/scurvy that may have been corrected by treatment; lack of definitive pathology; retrospective selective bias; and lack of adequate controls. Even so, there remains at least one exceptionally well documented case report by Cameron and colleagues, of double complete regression (Cameron et al., 1975; Campbell et al., 1991). We note that exquisite sensitivity to H2O2 generated by the initial IV ascorbate could still explain findings, and other limitations above may also be confounders. Although there are no direct measurements in humans that H2O2 is generated by pharmacologic ascorbate, there are direct rodent data (Chen et al., 2007; Chen et al., 2008). Supportive evidence that H2O2 is generated by pharmacologic ascorbate in humans is provided by reports of oxidative hemolysis in patients with glucose 6-phosphate dehydrogenase deficiency who received intravenous ascorbate (Huang et al., 2014; Rees et al., 1993). Indeed, it is for this very reason that patients who receive intravenous ascorbate must be pre-screened for glucose 6 phosphate dehydrogenase deficiency. Taken as a whole, Cameron’s reported cases hint that there could be other H2O2-independent potential actions of pharmacologic ascorbate, because intravenous doses at those used are predicted to have relatively small effects on extracellular H2O2 formation.
Ascorbate and epigenetic regulation
In this vein, recent reports demonstrate that AA at physiologic concentrations from oral doses may exert its anticancer activity through an epigenetic mechanism (DNA demethylation), distinct from and mechanistically independent of H2O2 generation and pharmacologic ascorbate concentrations. Ascorbate functions as a cofactor and increases the activity of the Ten Eleven Translocation (TET) enzymes, causing DNA demethylation and increase in hydroxymethylation (Blaschke et al., 2013; Yin et al., 2013). This function has been found to result in re-expression of tumor suppressor genes in cancer cells, promotion of stem cell differentiation, and enhancement of DNA methyltransferase inhibitor (DNMTi)-induced immune signals via increased expression of endogenous retrovirus transcripts (Agathocleous et al., 2017; Cimmino et al., 2017; Liu et al., 2016; Shenoy et al., 2017). The comparative effect is currently unknown of IV versus physiologic, or oral, doses of Vitamin C on in vivo DNA demethylation and DNA hydroxymethylation. It is also unclear at this point to what extent other effects of ascorbic acid mediated through direct/ indirect cofactor functions, including Jumonji histone demethylase activity (Wang et al., 2011), contribute to its overall effect on established cancer. Despite these uncertainties, it is a reasonable assumption that with IV dosing, the impact on direct/ indirect cofactor functions of Vitamin C, including the TET mediated DNA demethylation effect, would be at least equal to, if not higher than, oral doses. Potential limiting factors to ascorbate uptake in cancers include tissue characteristics such as high cell density, poor perfusion, reduced expression/activity of transporters. Indeed, data from a three dimensional pharmacokinetic model to measure ascorbate diffusion suggests that ascorbate penetration and distribution in a poorly vascularized tumor may require higher than normal plasma concentrations.(Kuiper et al., 2014; Wohlrab et al., 2017) The IV dosing also provides the additional benefit of generating extracellular hydrogen peroxide, which can induce oxidative stress in cancer cells. With this background, there is potential to explore IV ascorbate as a therapeutic approach for subsets of malignancies with known aberrant hypermethylation such as chronic myelomonocytic leukemia (60–70% of cases with TET2 mutations), myelodysplastic syndrome, TET2 mutated together with either IDH1 or IDH2 mutated acute myelogenous leukemia (2-hydroxyglutarate mediated suppression of TET), clear cell renal cell carcinoma, and succinate dehydrogenase mutated paraganglioma. (Bejar et al., 2014; Figueroa et al., 2010; Hu et al., 2014; Jiang et al., 2009; Letouze et al., 2013; Patnaik and Tefferi, 2016; Rasmussen et al., 2015) Furthermore, loss of hydroxymethylcytosine, which has been reported in multiple cancers (Chen et al., 2013; Jin et al., 2011; Ko et al., 2010; Kudo et al., 2012; Mason and Hornick, 2013; Muller et al., 2012; Orr et al., 2012; Perez et al., 2012; Yang et al., 2013) and is an adverse prognostic marker, could be investigated as a potential biomarker for ascorbate treatment since ascorbate has been found to increase hydroxymethylcytosine levels in vitro in cancer cells via the activation of the TET enzymes.(Agathocleous et al., 2017; Gustafson et al., 2015; Shenoy et al., 2017) Given that most of the TET2 inactivating mutations are uniallelic, it is possible that the activity of the wild-type copy could be optimized by pharmacologic ascorbate and produce therapeutic benefit. Furthermore, even if the TET enzyme is unmutated, its activity could still be suppressed in some malignancies by the abnormal accumulation of Krebs cycle intermediates. The relative importance of the two mechanisms of anti-cancer activity, i.e. epigenetic and H2O2 mediated, is yet to be established in humans.
Ascorbate and cancer cell culture models: some limitations
Ascorbate modification of TET and Hypoxia Inducible Factor- Prolyl Hydroxylase (HIF-PHD) activities in cell culture models must also be considered in context of limitations of using ascorbate in these models. A reminder: cells in culture have no ascorbate unless it is purposely added to media. Most commercial media lack ascorbate because it easily oxidizes in vitro. When ascorbate is added to media, the extracellular concentration does not reflect the intracellular concentration that can be attained at homeostasis, when ascorbate concentrations are kept constant over time. Even acutely isolated cells from humans lose ~ 90 % of their intracellular ascorbate within 72 hours in culture (Bergsten et al., 1990). Excepting acutely isolated cells, ascorbate intracellular concentrations in vitro should be considered to begin at zero, a condition that is very difficult to reconcile with human biology. In vitro, estimates of steady-state intracellular concentrations of ascorbate for a given extracellular concentration depend not on simply one added extracellular concentration but rather on multiple factors, including oxidation of extracellular media ascorbate, replenishment of oxidized ascorbate, incubation times, contribution of dehydroascorbic acid transport, and ascorbate and GLUT transporter copy numbers. In addition, ascorbate-dependent enzyme activities and ascorbate transport mechanisms observed in cell lines might not fully reflect in vivo conditions. For these reasons, clinical trial designs testing ascorbate in cancer treatment cannot be based only on in vitro data, where adding ascorbate to cells changes enzyme activity, but rather on in vivo findings.
Physiologic ascorbate/cancer prevention; pharmacologic ascorbate/cancer treatment
While our focus is on cancer treatment with ascorbate, findings in mice suggest a role of oral ascorbate in delaying cancer development, based on ascorbate dependent activities of TET and inhibition of hypoxia inducible factor by prolyl hydroxylases (HIF-PHD) enzymes (Agathocleous et al., 2017; Campbell et al., 2016; Gao et al., 2007). TET experiments used the Gulo−/− mouse model that lacks the endogenous ascorbate synthesis pathway resulting from the gene encoding L-gulonolactone oxidase (Gulo), which encodes a key enzyme of the pathway, being knockout. Like humans, whose L-gulonolactone oxidase is non-functional, Gulo−/− mice are ascorbate requiring. Recent elegant experiments utilizing bone marrow transplantation in ascorbate deficient Gulo−/− mice with TET2 deficiency indicated that there is enhanced development of acute myelogenous leukemia. In these mice, oral ascorbate postponed progression of genetically susceptible transplanted hematopoietic cells to acute myelogenous leukemia (Agathocleous et al., 2017). Oral ascorbate was shown to increase TET activity and hydroxymethylcytosine levels. However, there was no evidence of a decrease in blast percentage in the same animals once acute myelogenous leukemia was established. Similarly, in a lymphoma xenograft model tumor development was decreased by oral ascorbate, which had HIF-1-dependent anti-tumorigenic properties (Gao et al., 2007). Here too, oral ascorbate was introduced before tumor development, seven days before inoculation of cancer cells. For comparison purposes, tumor treatment to reduce tumor growth in many rodent models was mediated by parenteral ascorbate to produce pharmacologic concentrations (Chen et al., 2008; Espey et al., 2011; Ma et al., 2014; Rouleau et al., 2016; Serrano et al., 2015; Verrax and Calderon, 2009; Xia et al., 2017)). In a hepatoma xenograft model, parenteral ascorbate inhibited tumor growth, but oral ascorbate had no treatment effect (Verrax and Calderon, 2009). In other experiments Gulo−/− mice were implanted with various tumors. Tumor growth was faster in ascorbate deficient compared to mice repleted with ascorbate by mouth (Campbell et al., 2015; Campbell et al., 2016; Cha et al., 2013). Considered as a whole, the animal data suggest that oral ascorbate may be effective as an agent to prevent or postpone the development of certain types of malignancy in genetically pre-disposed individuals. Possibilities include individuals having hematopoietic cells with a TET2 mutation, with or without low grade dysplasia, by optimizing activity of the wild-type allele; VHL haplodeficient individuals predisposed to renal cell carcinoma by optimizing HIF-PHD activity; and succinate dehydrogenase/fumarate hydratase deficient individuals predisposed to renal cell carcinoma or pheochromocytoma/paraganglioma, by optimizing activity of the metabolically inhibited TET and HIF-PHD enzymes. In contrast, to our knowledge, there are no experiments in Gulo−/− mice and in other rodent models that show that varying oral ascorbate is effective for treatment of pre-existing solid or hematologic malignancies. Some investigators have noted that Gulo−/− mice tolerate much lower ascorbate concentrations than humans (Campbell et al., 2015; Tu et al., 2015) i.e. < 1 mM. Gulo−/− mice that had ascorbate concentrations approximating those of scurvy in humans for one year neither had symptoms of scurvy during their life span nor significant evidence of cancer at necropsy (Tu et al., 2017). For these reasons, we believe that caution is appropriate in extending findings of Gulo−/− mice models to humans.
Ascorbic acid concentrations in cancer patients are often at deficiency concentrations (Huijskens et al., 2016; Liu et al., 2016; Mayland et al., 2005), the reason for which is still uncertain. Possibilities include generalized cancer cachexia, enhanced consumption of ascorbate by tumors, or downregulation of ascorbate transporters. It is therefore reasonable to consider whether there is a role for oral ascorbic acid as a treatment modality by correction of deficiency of ascorbate, independent of pharmacologic administration. In humans, results of randomized treatment trials were negative in unselected advanced cancers and advanced colorectal cancer (Creagan et al., 1979; Moertel et al., 1985). There are rare case reports where oral ascorbic acid may have been useful, but this was in conjunction with intravenous as well as oral ascorbate. Based on recent in vitro evidence, correction of ascorbate deficiency may aid conventional hypomethylating agents i.e. DNA methyltransferase inhibitors to exert maximal effect (Liu et al., 2016).
The rationale for the use of high dose intravenous ascorbate in cancer treatment is not to correct plasma deficiency, but rather to induce an oxidative stress on cancer cells (Chen et al., 2005; Chen et al., 2007; Chen et al., 2008) and to ensure adequate delivery of ascorbate within tumors for optimal cofactor function (Kuiper et al., 2014). As a way forward, it is essential to acknowledge that only pharmacologic, i.e. parenteral ascorbate, to date has produced clearly documented treatment efficacy in early phase human trials. In rodent models of treatment of established cancers, only pharmacologic ascorbate is effective regardless of the model: nude-mouse, syngeneic, specific for dehydroascorbic acid/GLUT transport mechanisms, or coupled to TET actions. These data provide guidance for clinical trials. Pharmacologic ascorbate is the standard to which other ascorbate dosing schemes should be compared to. There is promising efficacy in small human studies, and a patient safety profile similar to that of an intravenous antibiotic rather than systemic chemotherapy, but many unknowns remain. Dosing information is limited, including dosing amount, dosing frequency, ascorbate administration timing with conventional agents, and treatment duration. Clinical evidence is that at a minimum 1 g/kg should be administered intravenously over 2 hours twice weekly, with more frequent administration perhaps better (Hoffer et al., 2008; Ma et al., 2014; Monti et al., 2012; Polireddy et al., 2017; Schoenfeld et al., 2017; Welsh et al., 2013). Based on the small datasets available, the absolute minimum treatment duration is 2 months before efficacy is assessed, and probably 3–4 months (Drisko et al., 2018; Monti et al., 2012; Nielsen et al., 2017; Rouleau et al., 2016). These considerations place burdens on both trialists and patients. For patients, it may not be appealing to have to sit for treatment 2 hours three times weekly, for months if not longer. For trialists, problems can be anticipated with initial recruitment and treatment continuity. Nevertheless, we should not decrease dosing, frequency, or duration without clear supportive data. Basing decisions on in vitro data will perhaps irrevocably harm the emerging efforts to test ascorbate in cancer treatment.
The Phoenix Can Rise
The implications are substantial of a therapeutic agent that is universally available and accessible, with the potential of anti-cancer activity across multiple tumor types as well as reducing chemotherapy toxicity. Should the oncology research community invest in the necessary randomized trials to test whether there is benefit of IV ascorbate? We firmly recommend yes, utilizing well-designed randomized (and where possible, placebo-controlled) trials. Establishing biomarkers for selection of patients who benefit from IV ascorbate would enable us to tailor treatment more effectively and efficiently. Mechanism bases should be explored, with caveats. However, despite intentions to pursue well-defined biomarkers of response, it may be difficult to zero in on a single, highly predictive biomarker. This is because pharmacologic ascorbate can be considered a promiscuous agent mechanistically with respect to generation of extracellular H2O2 as well as its varied cofactor functions (Levine and Violet, 2017). Once extracellular H2O2 is present, myriad mechanisms can mediate cancer cell death. While these aspects may not be in keeping with the “targeted therapy” era in oncology, they should not deter exploration of ascorbate’s potential as a therapeutic agent. Primary endpoints may be tumor response rate or survival. Secondary endpoints include adverse effect profiles and quality of life. We strongly encourage the field to explore ascorbate’s potential, but also send a cautionary note that new studies need to be done properly. Let’s not shoot down the re-rising ascorbate treatment phoenix.
Acknowledgement
Intramural Research Program, NIDDK, NIH, Grants Z01-DK053212–12, Z01-DK053218–12, Z01-DK054506–21 (M. Levine)
American Society of Hematology Research Training Award for Fellows and University of Iowa/ Mayo Clinic Lymphoma SPORE CA97274 Development Research Project Award (N. Shenoy)
ABBREVIATIONS
- IV
intravenous
- H2O2
hydrogen peroxide
- HIF PHD
Hypoxia-inducible factor, prolyl hydroxylase dependent
- TET
Ten- Eleven Translocation
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure of Potential Conflicts of Interest: The authors declare no competing interests
References:
- Agathocleous M, Meacham CE, Burgess RJ, Piskounova E, Zhao Z, Crane GM, Cowin BL, Bruner E, Murphy MM, Chen W, et al. (2017). Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bejar R, Lord A, Stevenson K, Bar-Natan M, Perez-Ladaga A, Zaneveld J, Wang H, Caughey B, Stojanov P, Getz G, et al. (2014). TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood 124, 2705–2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bergsten P, Amitai G, Kehrl J, Dhariwal KR, Klein HG, and Levine M (1990). Millimolar concentrations of ascorbic acid in purified human mononuclear leukocytes. Depletion and reaccumulation. The Journal of biological chemistry 265, 2584–2587. [PubMed] [Google Scholar]
- Blaschke K, Ebata KT, Karimi MM, Zepeda-Martinez JA, Goyal P, Mahapatra S, Tam A, Laird DJ, Hirst M, Rao A, et al. (2013). Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 500, 222–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron E, and Campbell A (1974). The orthomolecular treatment of cancer. II. Clinical trial of high-dose ascorbic acid supplements in advanced human cancer. Chemico-biological interactions 9, 285–315. [DOI] [PubMed] [Google Scholar]
- Cameron E, Campbell A, and Jack T (1975). The orthomolecular treatment of cancer. III. Reticulum cell sarcoma: double complete regression induced by high-dose ascorbic acid therapy. Chemico-biological interactions 11, 387–393. [DOI] [PubMed] [Google Scholar]
- Cameron E, and Pauling L (1976). Supplemental ascorbate in the supportive treatment of cancer: Prolongation of survival times in terminal human cancer. Proceedings of the National Academy of Sciences of the United States of America 73, 3685–3689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron E, and Pauling L (1978). Supplemental ascorbate in the supportive treatment of cancer: reevaluation of prolongation of survival times in terminal human cancer. Proceedings of the National Academy of Sciences of the United States of America 75, 4538–4542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell A, Jack T, and Cameron E (1991). Reticulum cell sarcoma: two complete ‘spontaneous’ regressions, in response to high-dose ascorbic acid therapy. A report on subsequent progress. Oncology 48, 495–497. [DOI] [PubMed] [Google Scholar]
- Campbell EJ, Vissers MC, Bozonet S, Dyer A, Robinson BA, and Dachs GU (2015). Restoring physiological levels of ascorbate slows tumor growth and moderates HIF-1 pathway activity in Gulo(−/−) mice. Cancer medicine 4, 303–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campbell EJ, Vissers MCM, Wohlrab C, Hicks KO, Strother RM, Bozonet SM, Robinson BA, and Dachs GU (2016). Pharmacokinetic and anti-cancer properties of high dose ascorbate in solid tumours of ascorbate-dependent mice. Free radical biology & medicine 99, 451–462. [DOI] [PubMed] [Google Scholar]
- Carr AC, Vissers MC, and Cook JS (2014). The effect of intravenous vitamin C on cancer- and chemotherapy-related fatigue and quality of life. Frontiers in oncology 4, 283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cha J, Roomi MW, Ivanov V, Kalinovsky T, Niedzwiecki A, and Rath M (2013). Ascorbate supplementation inhibits growth and metastasis of B16FO melanoma and 4T1 breast cancer cells in vitamin C-deficient mice. International journal of oncology 42, 55–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen ML, Shen F, Huang W, Qi JH, Wang Y, Feng YQ, Liu SM, and Yuan BF (2013). Quantification of 5-methylcytosine and 5-hydroxymethylcytosine in genomic DNA from hepatocellular carcinoma tissues by capillary hydrophilic-interaction liquid chromatography/quadrupole TOF mass spectrometry. Clinical chemistry 59, 824–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q, Espey MG, Krishna MC, Mitchell JB, Corpe CP, Buettner GR, Shacter E, and Levine M (2005). Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues. Proceedings of the National Academy of Sciences of the United States of America 102, 13604–13609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q, Espey MG, Sun AY, Lee JH, Krishna MC, Shacter E, Choyke PL, Pooput C, Kirk KL, Buettner GR, and Levine M (2007). Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proceedings of the National Academy of Sciences of the United States of America 104, 8749–8754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Q, Espey MG, Sun AY, Pooput C, Kirk KL, Krishna MC, Khosh DB, Drisko J, and Levine M (2008). Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proceedings of the National Academy of Sciences of the United States of America 105, 11105–11109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cimmino L, Dolgalev I, Wang Y, Yoshimi A, Martin GH, Wang J, Ng V, Xia B, Witkowski MT, Mitchell-Flack M, et al. (2017). Restoration of TET2 Function Blocks Aberrant Self-Renewal and Leukemia Progression. Cell 170, 1079–1095.e1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corpe CP, Eck P, Wang J, Al-Hasani H, and Levine M (2013). Intestinal dehydroascorbic acid (DHA) transport mediated by the facilitative sugar transporters, GLUT2 and GLUT8. The Journal of biological chemistry 288, 9092–9101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corpe CP, Tu H, Eck P, Wang J, Faulhaber-Walter R, Schnermann J, Margolis S, Padayatty S, Sun H, Wang Y, et al. (2010). Vitamin C transporter Slc23a1 links renal reabsorption, vitamin C tissue accumulation, and perinatal survival in mice. The Journal of clinical investigation 120, 1069–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Creagan ET, Moertel CG, O’Fallon JR, Schutt AJ, O’Connell MJ, Rubin J, and Frytak S (1979). Failure of high-dose vitamin C (ascorbic acid) therapy to benefit patients with advanced cancer. A controlled trial. N Engl J Med 301, 687–690. [DOI] [PubMed] [Google Scholar]
- Drisko JA, Serrano OK, Spruce LR, Chen Q, and Levine M (2018). Treatment of pancreatic cancer with intravenous vitamin C: a case report. Anti-cancer drugs 29, 373–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Espey MG, Chen P, Chalmers B, Drisko J, Sun AY, Levine M, and Chen Q (2011). Pharmacologic ascorbate synergizes with gemcitabine in preclinical models of pancreatic cancer. Free radical biology & medicine 50, 1610–1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, et al. (2010). Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer cell 18, 553–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao P, Zhang H, Dinavahi R, Li F, Xiang Y, Raman V, Bhujwalla ZM, Felsher DW, Cheng L, Pevsner J, et al. (2007). HIF-dependent antitumorigenic effect of antioxidants in vivo. Cancer cell 12, 230–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gustafson CB, Yang C, Dickson KM, Shao H, Van Booven D, Harbour JW, Liu ZJ, and Wang G (2015). Epigenetic reprogramming of melanoma cells by vitamin C treatment. Clinical epigenetics 7, 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffer LJ, Levine M, Assouline S, Melnychuk D, Padayatty SJ, Rosadiuk K, Rousseau C, Robitaille L, and Miller WH Jr. (2008). Phase I clinical trial of i.v. ascorbic acid in advanced malignancy. Annals of oncology : official journal of the European Society for Medical Oncology 19, 1969–1974. [DOI] [PubMed] [Google Scholar]
- Hoffer LJ, Robitaille L, Zakarian R, Melnychuk D, Kavan P, Agulnik J, Cohen V, Small D, and Miller WH Jr. (2015). High-dose intravenous vitamin C combined with cytotoxic chemotherapy in patients with advanced cancer: a phase I-II clinical trial. PloS one 10, e0120228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu CY, Mohtat D, Yu Y, Ko YA, Shenoy N, Bhattacharya S, Izquierdo MC, Park AS, Giricz O, Vallumsetla N, et al. (2014). Kidney cancer is characterized by aberrant methylation of tissue-specific enhancers that are prognostic for overall survival. Clinical cancer research : an official journal of the American Association for Cancer Research 20, 4349–4360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang YC, Chang TK, Fu YC, and Jan SL (2014). C for colored urine: acute hemolysis induced by high-dose ascorbic acid. Clinical toxicology (Philadelphia, Pa) 52, 984. [DOI] [PubMed] [Google Scholar]
- Huijskens MJ, Wodzig WK, Walczak M, Germeraad WT, and Bos GM (2016). Ascorbic acid serum levels are reduced in patients with hematological malignancies. Results in immunology 6, 8–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang Y, Dunbar A, Gondek LP, Mohan S, Rataul M, O’Keefe C, Sekeres M, Saunthararajah Y, and Maciejewski JP (2009). Aberrant DNA methylation is a dominant mechanism in MDS progression to AML. Blood 113, 1315–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin SG, Jiang Y, Qiu R, Rauch TA, Wang Y, Schackert G, Krex D, Lu Q, and Pfeifer GP (2011). 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. Cancer research 71, 7360–7365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ko M, Huang Y, Jankowska AM, Pape UJ, Tahiliani M, Bandukwala HS, An J, Lamperti ED, Koh KP, Ganetzky R, et al. (2010). Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 468, 839–843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kudo Y, Tateishi K, Yamamoto K, Yamamoto S, Asaoka Y, Ijichi H, Nagae G, Yoshida H, Aburatani H, and Koike K (2012). Loss of 5-hydroxymethylcytosine is accompanied with malignant cellular transformation. Cancer science 103, 670–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuiper C, Vissers MC, and Hicks KO (2014). Pharmacokinetic modeling of ascorbate diffusion through normal and tumor tissue. Free radical biology & medicine 77, 340–352. [DOI] [PubMed] [Google Scholar]
- Letouze E, Martinelli C, Loriot C, Burnichon N, Abermil N, Ottolenghi C, Janin M, Menara M, Nguyen AT, Benit P, et al. (2013). SDH mutations establish a hypermethylator phenotype in paraganglioma. Cancer cell 23, 739–752. [DOI] [PubMed] [Google Scholar]
- Levine M, Conry-Cantilena C, Wang Y, Welch RW, Washko PW, Dhariwal KR, Park JB, Lazarev A, Graumlich JF, King J, and Cantilena LR (1996). Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proceedings of the National Academy of Sciences of the United States of America 93, 3704–3709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine M, and Violet PC (2017). Data Triumph at C. Cancer cell 31, 467–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levine M, Wang Y, Katz A, Eck P, Kwon O, Chen S, Lee JH, and Padayatty SJ (2001). Ideal vitamin C intake. BioFactors (Oxford, England) 15, 71–74. [DOI] [PubMed] [Google Scholar]
- Liu M, Ohtani H, Zhou W, Orskov AD, Charlet J, Zhang YW, Shen H, Baylin SB, Liang G, Gronbaek K, and Jones PA (2016). Vitamin C increases viral mimicry induced by 5-aza-2’-deoxycytidine. Proceedings of the National Academy of Sciences of the United States of America 113, 10238–10244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Y, Chapman J, Levine M, Polireddy K, Drisko J, and Chen Q (2014). High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy. Science translational medicine 6, 222ra218. [DOI] [PubMed] [Google Scholar]
- Mason EF, and Hornick JL (2013). Succinate dehydrogenase deficiency is associated with decreased 5-hydroxymethylcytosine production in gastrointestinal stromal tumors: implications for mechanisms of tumorigenesis. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 26, 1492–1497. [DOI] [PubMed] [Google Scholar]
- Mayland CR, Bennett MI, and Allan K (2005). Vitamin C deficiency in cancer patients. Palliative medicine 19, 17–20. [DOI] [PubMed] [Google Scholar]
- Moertel CG, Fleming TR, Creagan ET, Rubin J, O’Connell MJ, and Ames MM (1985). High-dose vitamin C versus placebo in the treatment of patients with advanced cancer who have had no prior chemotherapy. A randomized double-blind comparison. N Engl J Med 312, 137–141. [DOI] [PubMed] [Google Scholar]
- Monti DA, Mitchell E, Bazzan AJ, Littman S, Zabrecky G, Yeo CJ, Pillai MV, Newberg AB, Deshmukh S, and Levine M (2012). Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. PloS one 7, e29794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muller T, Gessi M, Waha A, Isselstein LJ, Luxen D, Freihoff D, Freihoff J, Becker A, Simon M, Hammes J, et al. (2012). Nuclear exclusion of TET1 is associated with loss of 5-hydroxymethylcytosine in IDH1 wild-type gliomas. The American journal of pathology 181, 675–683. [DOI] [PubMed] [Google Scholar]
- Nielsen TK, Hojgaard M, Andersen JT, Jorgensen NR, Zerahn B, Kristensen B, Henriksen T, Lykkesfeldt J, Mikines KJ, and Poulsen HE (2017). Weekly ascorbic acid infusion in castration-resistant prostate cancer patients: a single-arm phase II trial. Translational andrology and urology 6, 517–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orr BA, Haffner MC, Nelson WG, Yegnasubramanian S, and Eberhart CG (2012). Decreased 5-hydroxymethylcytosine is associated with neural progenitor phenotype in normal brain and shorter survival in malignant glioma. PloS one 7, e41036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padayatty SJ, and Levine M (2000). Reevaluation of ascorbate in cancer treatment: emerging evidence, open minds and serendipity. Journal of the American College of Nutrition 19, 423–425. [DOI] [PubMed] [Google Scholar]
- Padayatty SJ, Sun AY, Chen Q, Espey MG, Drisko J, and Levine M (2010). Vitamin C: intravenous use by complementary and alternative medicine practitioners and adverse effects. PloS one 5, e11414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padayatty SJ, Sun H, Wang Y, Riordan HD, Hewitt SM, Katz A, Wesley RA, and Levine M (2004). Vitamin C pharmacokinetics: implications for oral and intravenous use. Annals of internal medicine 140, 533–537. [DOI] [PubMed] [Google Scholar]
- Park CH, Kimler BF, Yi SY, Park SH, Kim K, Jung CW, Kim SH, Lee ER, Rha M, Kim S, et al. (2009). Depletion of L-ascorbic acid alternating with its supplementation in the treatment of patients with acute myeloid leukemia or myelodysplastic syndromes. European journal of haematology 83, 108–118. [DOI] [PubMed] [Google Scholar]
- Patnaik MM, and Tefferi A (2016). Chronic myelomonocytic leukemia: 2016 update on diagnosis, risk stratification, and management. American journal of hematology 91, 631–642. [DOI] [PubMed] [Google Scholar]
- Patterson JW, and Lazarow A (1950). Sulfhydryl protection against dehydroascorbic acid diabetes. The Journal of biological chemistry 186, 141–144. [PubMed] [Google Scholar]
- Perez C, Martinez-Calle N, Martin-Subero JI, Segura V, Delabesse E, Fernandez-Mercado M, Garate L, Alvarez S, Rifon J, Varea S, et al. (2012). TET2 mutations are associated with specific 5-methylcytosine and 5-hydroxymethylcytosine profiles in patients with chronic myelomonocytic leukemia. PloS one 7, e31605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polireddy K, Dong R, Reed G, Yu J, Chen P, Williamson S, Violet PC, Pessetto Z, Godwin AK, Fan F, et al. (2017). High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Scientific reports 7, 17188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rasmussen KD, Jia G, Johansen JV, Pedersen MT, Rapin N, Bagger FO, Porse BT, Bernard OA, Christensen J, and Helin K (2015). Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes & development 29, 910–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rees DC, Kelsey H, and Richards JD (1993). Acute haemolysis induced by high dose ascorbic acid in glucose-6-phosphate dehydrogenase deficiency. BMJ (Clinical research ed) 306, 841–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rouleau L, Antony AN, Bisetto S, Newberg A, Doria C, Levine M, Monti DA, and Hoek JB (2016). Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: New insights into ascorbate cytotoxicity. Free radical biology & medicine 95, 308–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Wagner BA, Cramer-Morales KL, Furqan M, Sandhu S, Carlisle TL, Smith MC, Abu Hejleh T, et al. (2017). O2- and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer cell 31, 487–500.e488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serrano OK, Parrow NL, Violet PC, Yang J, Zornjak J, Basseville A, and Levine M (2015). Antitumor effect of pharmacologic ascorbate in the B16 murine melanoma model. Free radical biology & medicine 87, 193–203. [DOI] [PubMed] [Google Scholar]
- Shenoy N, Bhagat T, Nieves E, Stenson M, Lawson J, Choudhary GS, Habermann T, Nowakowski G, Singh R, Wu X, et al. (2017). Upregulation of TET activity with ascorbic acid induces epigenetic modulation of lymphoma cells. Blood cancer journal 7, e587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shim EH, Livi CB, Rakheja D, Tan J, Benson D, Parekh V, Kho EY, Ghosh AP, Kirkman R, Velu S, et al. (2014). L-2-Hydroxyglutarate: an epigenetic modifier and putative oncometabolite in renal cancer. Cancer discovery 4, 1290–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sotiriou S, Gispert S, Cheng J, Wang Y, Chen A, Hoogstraten-Miller S, Miller GF, Kwon O, Levine M, Guttentag SH, and Nussbaum RL (2002). Ascorbic-acid transporter Slc23a1 is essential for vitamin C transport into the brain and for perinatal survival. Nature medicine 8, 514–517. [DOI] [PubMed] [Google Scholar]
- Stephenson CM, Levin RD, Spector T, and Lis CG (2013). Phase I clinical trial to evaluate the safety, tolerability, and pharmacokinetics of high-dose intravenous ascorbic acid in patients with advanced cancer. Cancer chemotherapy and pharmacology 72, 139–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tu H, Li H, Wang Y, Niyyati M, Wang Y, Leshin J, and Levine M (2015). Low Red Blood Cell Vitamin C Concentrations Induce Red Blood Cell Fragility: A Link to Diabetes Via Glucose, Glucose Transporters, and Dehydroascorbic Acid. EBioMedicine 2, 1735–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tu H, Wang Y, Li H, Brinster LR, and Levine M (2017). Chemical Transport Knockout for Oxidized Vitamin C, Dehydroascorbic Acid, Reveals Its Functions in vivo. EBioMedicine 23, 125–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verrax J, and Calderon PB (2009). Pharmacologic concentrations of ascorbate are achieved by parenteral administration and exhibit antitumoral effects. Free radical biology & medicine 47, 32–40. [DOI] [PubMed] [Google Scholar]
- Wang T, Chen K, Zeng X, Yang J, Wu Y, Shi X, Qin B, Zeng L, Esteban MA, Pan G, and Pei D (2011). The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. Cell stem cell 9, 575–587. [DOI] [PubMed] [Google Scholar]
- Welsh JL, Wagner BA, van’t Erve TJ, Zehr PS, Berg DJ, Halfdanarson TR, Yee NS, Bodeker KL, Du J, Roberts LJ 2nd, et al. (2013). Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): results from a phase I clinical trial. Cancer chemotherapy and pharmacology 71, 765–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wittes RE (1985). Vitamin C and cancer. N Engl J Med 312, 178–179. [DOI] [PubMed] [Google Scholar]
- Wohlrab C, Phillips E, and Dachs GU (2017). Vitamin C Transporters in Cancer: Current Understanding and Gaps in Knowledge. Frontiers in oncology 7, 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia J, Xu H, Zhang X, Allamargot C, Coleman KL, Nessler R, Frech I, Tricot G, and Zhan F (2017). Multiple Myeloma Tumor Cells are Selectively Killed by Pharmacologically-dosed Ascorbic Acid. EBioMedicine 18, 41–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H, Liu Y, Bai F, Zhang JY, Ma SH, Liu J, Xu ZD, Zhu HG, Ling ZQ, Ye D, et al. (2013). Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation. Oncogene 32, 663–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin R, Mao SQ, Zhao B, Chong Z, Yang Y, Zhao C, Zhang D, Huang H, Gao J, Li Z, et al. (2013). Ascorbic acid enhances Tet-mediated 5-methylcytosine oxidation and promotes DNA demethylation in mammals. Journal of the American Chemical Society 135, 10396–10403. [DOI] [PubMed] [Google Scholar]
- Yun J, Mullarky E, Lu C, Bosch KN, Kavalier A, Rivera K, Roper J, Chio II, Giannopoulou EG, Rago C, et al. (2015). Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science (New York, NY) 350, 1391–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]