High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients
Abstract
:Simple Summary
Abstract
1. Introduction
2. Methods
3. Chemical Forms of Vitamin C
4. Vitamin C Transport into the Cells
5. Vitamin C Dose-Dependent Pharmacokinetics
6. Mechanisms of Vitamin C Anticancer Action
6.1. Vitamin C as Pro-Oxidant Agent
6.2. Vitamin C as Enzymatic Regulator of TET Enzymes
6.3. Vitamin C as Enzymatic Regulator of Others αKGDDs
6.4. Vitamin C may Favor DNA Damage by Increasing 5-Hydroxymethylcytosine Levels
7. High-Dose Vitamin C Exerts Preferential Cytotoxic Activity against Cancer Cells
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Carpenter, K.J. The Discovery of Vitamin C. Ann. Nutr. Metab. 2012, 61, 259–264. [Google Scholar] [CrossRef] [PubMed]
- Svirbely, J.L.; Szent-Györgyi, A. The chemical nature of vitamin C. Biochem. J. 1932, 26, 865–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterkofsky, B. Ascorbate requirement for hydroxylation and secretion of procollagen: Relationship to inhibition of collagen synthesis in scurvy. Am. J. Clin. Nutr. 1991, 54, 1135S–1140S. [Google Scholar] [CrossRef] [PubMed]
- Levine, M.; Conry-Cantilena, C.; Wang, Y.; Welch, R.W.; Washko, P.W.; Dhariwal, K.R.; Park, J.B.; Lazarev, A.; Graumlich, J.F.; King, J.; et al. Vitamin C pharmacokinetics in healthy volunteers: Evidence for a recommended dietary allowance. Proc. Natl. Acad. Sci. USA 1996, 93, 3704–3709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levine, M.; Rumsey, S.C.; Daruwala, R.; Park, J.B.; Wang, Y. Criteria and recommendations for vitamin C intake. J. Am. Med. Assoc. 1999. [Google Scholar] [CrossRef]
- Weinstein, M.; Babyn, P.; Zlotkin, S. An Orange a Day Keeps the Doctor Away: Scurvy in the Year 2000. Pediatrics 2001, 108, e55. [Google Scholar] [CrossRef] [Green Version]
- Wang, A.H.; Still, C. Old World Meets Modern: A Case Report of Scurvy. Nutr. Clin. Pract. 2007, 22, 445–448. [Google Scholar] [CrossRef]
- Dattola, A.; Silvestri, M.; Bennardo, L.; Passante, M.; Scali, E.; Patruno, C.; Nisticò, S.P. Role of Vitamins in Skin Health: A Systematic Review. Curr. Nutr. Rep. 2020, 9, 226–235. [Google Scholar] [CrossRef] [PubMed]
- Cameron, E.; Pauling, L. The orthomolecular treatment of cancer I. The role of ascorbic acid in host resistance. Chem. Biol. Interact. 1974, 9, 273–283. [Google Scholar] [CrossRef]
- Cameron, E.; Campbell, A. The orthomolecular treatment of cancer II. Clinical trial of high-dose ascorbic acid supplements in advanced human cancer. Chem. Biol. Interact. 1974. [Google Scholar] [CrossRef]
- Cameron, E.; Pauling, L. Supplemental ascorbate in the supportive treatment of cancer: Prolongation of survival times in terminal human cancer. Proc. Natl. Acad. Sci. USA 1976. [Google Scholar] [CrossRef] [Green Version]
- Creagan, E.T.; Moertel, C.G.; O’Fallon, J.R.; Schutt, A.J.; O’Connell, M.J.; Rubin, J.; Frytak, S. Failure of High-Dose Vitamin C (Ascorbic Acid) Therapy to Benefit Patients with Advanced Cancer. N. Engl. J. Med. 1979. [Google Scholar] [CrossRef]
- Moertel, C.G.; Fleming, T.R.; Creagan, E.T.; Rubin, J.; O’Connell, M.J.; Ames, M.M. High-Dose Vitamin C versus Placebo in the Treatment of Patients with Advanced Cancer Who Have Had No Prior Chemotherapy. N. Engl. J. Med. 1985. [Google Scholar] [CrossRef]
- Pauling, L.; Moertel, C. A Proposition: Megadoses of Vitamin C are Valuable in the Treatment of Cancer. Nutr. Rev. 1986, 44, 28–29. [Google Scholar] [CrossRef] [PubMed]
- Das, A.B.; Kakadia, P.M.; Wojcik, D.; Pemberton, L.; Browett, P.J.; Bohlander, S.K.; Vissers, M.C.M. Clinical remission following ascorbate treatment in a case of acute myeloid leukemia with mutations in TET2 and WT1. Blood Cancer J. 2019, 9, 82. [Google Scholar] [CrossRef] [PubMed]
- Bielski, B.H.J.; Allen, A.O.; Schwarz, H.A. Mechanism of the disproportionation of ascorbate radicals. J. Am. Chem. Soc. 1981, 103, 3516–3518. [Google Scholar] [CrossRef]
- Du, J.; Cullen, J.J.; Buettner, G.R. Ascorbic acid: Chemistry, biology and the treatment of cancer. Biochim. Biophys. Acta-Rev. Cancer 2012, 1826, 443–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ngo, B.; Van Riper, J.M.; Cantley, L.C.; Yun, J. Targeting cancer vulnerabilities with high-dose vitamin C. Nat. Rev. Cancer 2019, 19, 271–282. [Google Scholar] [CrossRef] [PubMed]
- Astuya, A.; Caprile, T.; Castro, M.; Salazar, K.; García, M.d.l.A.; Reinicke, K.; Rodríguez, F.; Vera, J.C.; Millán, C.; Ulloa, V.; et al. Vitamin C uptake and recycling among normal and tumor cells from the central nervous system. J. Neurosci. Res. 2005, 79, 146–156. [Google Scholar] [CrossRef] [PubMed]
- Padayatty, S.; Levine, M. Vitamin C: The known and the unknown and Goldilocks. Oral Dis. 2016, 22, 463–493. [Google Scholar] [CrossRef] [Green Version]
- Myllylä, R.; Kuutti-Savolainen, E.-R.; Kivirikko, K.I. The role of ascorbate in the prolyl hydroxylase reaction. Biochem. Biophys. Res. Commun. 1978, 83, 441–448. [Google Scholar] [CrossRef]
- May, J.M.; Qu, Z.; Meredith, M.E. Mechanisms of ascorbic acid stimulation of norepinephrine synthesis in neuronal cells. Biochem. Biophys. Res. Commun. 2012, 426, 148–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- May, J.M.; Qu, Z.; Nazarewicz, R.; Dikalov, S. Ascorbic acid efficiently enhances neuronal synthesis of norepinephrine from dopamine. Brain Res. Bull. 2013, 90, 35–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frei, B. Ascorbic acid protects lipids in human plasma and low-density lipoprotein against oxidative damage. Am. J. Clin. Nutr. 1991, 54, 1113S–1118S. [Google Scholar] [CrossRef] [PubMed]
- Buettner, G.R. The Pecking Order of Free Radicals and Antioxidants: Lipid Peroxidation, α-Tocopherol, and Ascorbate. Arch. Biochem. Biophys. 1993, 300, 535–543. [Google Scholar] [CrossRef] [PubMed]
- Przybyło, M.; Langner, M. On the physiological and cellular homeostasis of ascorbate. Cell. Mol. Biol. Lett. 2020, 25, 32. [Google Scholar] [CrossRef]
- May, J.M. The SLC23 family of ascorbate transporters: Ensuring that you get and keep your daily dose of vitamin C. Br. J. Pharmacol. 2011, 164, 1793–1801. [Google Scholar] [CrossRef] [Green Version]
- Godoy, A.; Ormazabal, V.; Moraga-Cid, G.; Zúñiga, F.A.; Sotomayor, P.; Barra, V.; Vasquez, O.; Montecinos, V.; Mardones, L.; Guzmán, C.; et al. Mechanistic Insights and Functional Determinants of the Transport Cycle of the Ascorbic Acid Transporter SVCT2. J. Biol. Chem. 2007, 282, 615–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- May, J.M.; Qu, Z.; Qiao, H. Transfer of ascorbic acid across the vascular endothelium: Mechanism and self-regulation. Am. J. Physiol. Physiol. 2009, 297, C169–C178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsukaguchi, H.; Tokui, T.; Mackenzie, B.; Berger, U.V.; Chen, X.-Z.; Wang, Y.; Brubaker, R.F.; Hediger, M.A. A family of mammalian Na+-dependent L-ascorbic acid transporters. Nature 1999, 399, 70–75. [Google Scholar] [CrossRef]
- Boyer, J.C.; Campbell, C.E.; Sigurdson, W.J.; Kuo, S.-M. Polarized localization of vitamin C transporters, SVCT1 and SVCT2, in epithelial cells. Biochem. Biophys. Res. Commun. 2005, 334, 150–156. [Google Scholar] [CrossRef] [PubMed]
- Jin, S.N.; Mun, G.H.; Lee, J.H.; Oh, C.S.; Kim, J.; Chung, Y.H.; Kang, J.S.; Kim, J.-G.; Hwang, D.-H.; Hwang, Y.I.; et al. Immunohistochemical study on the distribution of sodium-dependent vitamin C transporters in the respiratory system of adult rat. Microsc. Res. Tech. 2005, 68, 360–367. [Google Scholar] [CrossRef] [PubMed]
- Corpe, C.P.; Tu, H.; Eck, P.; Wang, J.; Faulhaber-Walter, R.; Schnermann, J.; Margolis, S.; Padayatty, S.; Sun, H.; Wang, Y.; et al. Vitamin C transporter Slc23a1 links renal reabsorption, vitamin C tissue accumulation, and perinatal survival in mice. J. Clin. Investig. 2010, 120, 1069–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sotiriou, S.; Gispert, S.; Cheng, J.; Wang, Y.; Chen, A.; Hoogstraten-Miller, S.; Miller, G.F.; Kwon, O.; Levine, M.; Guttentag, S.H.; et al. Ascorbic-acid transporter Slc23a1 is essential for vitamin C transport into the brain and for perinatal survival. Nat. Med. 2002, 8, 514–517. [Google Scholar] [CrossRef] [PubMed]
- May, J.M.; Qu, Z.; Qiao, H.; Koury, M.J. Maturational loss of the vitamin C transporter in erythrocytes. Biochem. Biophys. Res. Commun. 2007, 360, 295–298. [Google Scholar] [CrossRef] [Green Version]
- Ferrada, L.; Salazar, K.; Nualart, F. Metabolic control by dehydroascorbic acid: Questions and controversies in cancer cells. J. Cell. Physiol. 2019, 234, 19331–19338. [Google Scholar] [CrossRef]
- Wilson, J.X. Regulation of vitamin C transport. Annu. Rev. Nutr. 2005, 25, 105–125. [Google Scholar] [CrossRef] [PubMed]
- Savini, I.; Catani, M.V.; Arnone, R.; Rossi, A.; Frega, G.; Del Principe, D.; Avigliano, L. Translational control of the ascorbic acid transporter SVCT2 in human platelets. Free Radic. Biol. Med. 2007, 42, 608–616. [Google Scholar] [CrossRef]
- Corpe, C.P.; Eck, P.; Wang, J.; Al-Hasani, H.; Levine, M. Intestinal Dehydroascorbic Acid (DHA) Transport Mediated by the Facilitative Sugar Transporters, GLUT2 and GLUT8. J. Biol. Chem. 2013, 288, 9092–9101. [Google Scholar] [CrossRef] [Green Version]
- Gillberg, L.; Ørskov, A.D.; Liu, M.; Harsløf, L.B.S.; Jones, P.A.; Grønbæk, K. Vitamin C—A new player in regulation of the cancer epigenome. Semin. Cancer Biol. 2018, 51, 59–67. [Google Scholar] [CrossRef]
- Wang, Y.; Mackenzie, B.; Tsukaguchi, H.; Weremowicz, S.; Morton, C.C.; Hediger, M.A. Human Vitamin C (l-Ascorbic Acid) Transporter SVCT1. Biochem. Biophys. Res. Commun. 2000, 267, 488–494. [Google Scholar] [CrossRef]
- Malo, C.; Wilson, J.X. Glucose Modulates Vitamin C Transport in Adult Human Small Intestinal Brush Border Membrane Vesicles. J. Nutr. 2000, 130, 63–69. [Google Scholar] [CrossRef] [Green Version]
- Vera, J.C.; Rivas, C.I.; Fischbarg, J.; Golde, D.W. Mammalian facilitative hexose transporters mediate the transport of dehydroascorbic acid. Nature 1993, 364, 79–82. [Google Scholar] [CrossRef]
- Vera, J.; Rivas, C.; Zhang, R.; Farber, C.; Golde, D. Human HL-60 myeloid leukemia cells transport dehydroascorbic acid via the glucose transporters and accumulate reduced ascorbic acid. Blood 1994, 84, 1628–1634. [Google Scholar] [CrossRef]
- Vera, J.C.; Rivas, C.I.; Velásquez, F.V.; Zhang, R.H.; Concha, I.I.; Golde, D.W. Resolution of the Facilitated Transport of Dehydroascorbic Acid from Its Intracellular Accumulation as Ascorbic Acid. J. Biol. Chem. 1995, 270, 23706–23712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yun, J.; Mullarky, E.; Lu, C.; Bosch, K.N.; Kavalier, A.; Rivera, K.; Roper, J.; Chio, I.I.C.; Giannopoulou, E.G.; Rago, C.; et al. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science 2015, 350, 1391–1396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, Y.-X.; Wu, Q.-N.; Chen, D.; Chen, L.-Z.; Wang, Z.-X.; Ren, C.; Mo, H.; Chen, Y.; Sheng, H.; Wang, Y.-N.; et al. Pharmacological Ascorbate Suppresses Growth of Gastric Cancer Cells with GLUT1 Overexpression and Enhances the Efficacy of Oxaliplatin Through Redox Modulation. Theranostics 2018, 8, 1312–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kallner, A.; Hartmann, D.; Hornig, D. Steady-state turnover and body pool of ascorbic acid in man. Am. J. Clin. Nutr. 1979, 32, 530–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Graumlich, J.F.; Ludden, T.M.; Conry-Cantilena, C.; Cantilena, L.R.; Wang, Y.; Levine, M. Pharmacokinetic model of ascorbic acid in healthy male volunteers during depletion and repletion. Pharm. Res. 1997, 14, 1133–1139. [Google Scholar] [CrossRef]
- Nielsen, T.K.; Højgaard, M.; Andersen, J.T.; Poulsen, H.E.; Lykkesfeldt, J.; Mikines, K.J. Elimination of Ascorbic Acid After High-Dose Infusion in Prostate Cancer Patients: A Pharmacokinetic Evaluation. Basic Clin. Pharmacol. Toxicol. 2015, 116, 343–348. [Google Scholar] [CrossRef]
- Lykkesfeldt, J.; Tveden-Nyborg, P. The Pharmacokinetics of Vitamin C. Nutrients 2019, 11, 2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levine, M.; Wang, Y.; Padayatty, S.J.; Morrow, J. A new recommended dietary allowance of vitamin C for healthy young women. Proc. Natl. Acad. Sci. USA 2001, 98, 9842–9846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padayatty, S.J.; Sun, H.; Wang, Y.; Riordan, H.D.; Hewitt, S.M.; Katz, A.; Wesley, R.A.; Levine, M. Vitamin C Pharmacokinetics: Implications for Oral and Intravenous Use. Ann. Intern. Med. 2004, 140, 533. [Google Scholar] [CrossRef]
- Berger, L.; Gerson, C.D.; Yü, T.-F. The effect of ascorbic acid on uric acid excretion with a commentary on the renal handling of ascorbic acid. Am. J. Med. 1977, 62, 71–76. [Google Scholar] [CrossRef]
- Hickey, D.S.; Roberts, H.J.; Cathcart, R.F. Dynamic flow: A new model for ascorbate. J. Orthomol. Med. 2005, 20, 237–244. [Google Scholar]
- Duconge, J.; Miranda-Massari, J.R.; Gonzalez, M.J.; Jackson, J.A.; Warnock, W.; Riordan, N.H. Pharmacokinetics of vitamin C: Insights into the oral and intravenous administration of ascorbate. Puerto Rico Health Sci. J. 2008, 27, 7–19. [Google Scholar]
- Stephenson, C.M.; Levin, R.D.; Spector, T.; Lis, C.G. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahdavi, R.; Faramarzi, E.; Seyedrezazadeh, E.; Mohammad-zadeh, M.; Pourmoghaddam, M. Evaluation of Oxidative Stress, Antioxidant Status and Serum Vitamin C Levels in Cancer Patients. Biol. Trace Elem. Res. 2009, 130, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Mikirova, N.; Casciari, J.; Rogers, A.; Taylor, P. Effect of high-dose intravenous vitamin C on inflammation in cancer patients. J. Transl. Med. 2012, 10, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mikirova, N.; Casciari, J.; Riordan, N.; Hunninghake, R. Clinical experience with intravenous administration of ascorbic acid: Achievable levels in blood for different states of inflammation and disease in cancer patients. J. Transl. Med. 2013, 11, 191. [Google Scholar] [CrossRef] [Green Version]
- Fain, O.; Pariès, J.; Jacquart, B.; Le Moël, G.; Kettaneh, A.; Stirnemann, J.; Héron, C.; Sitbon, M.; Taleb, C.; Letellier, E.; et al. Hypovitaminosis C in hospitalized patients. Eur. J. Intern. Med. 2003. [Google Scholar] [CrossRef]
- Hoffer, L.J.; Levine, M.; Assouline, S.; Melnychuk, D.; Padayatty, S.J.; Rosadiuk, K.; Rousseau, C.; Robitaille, L.; Miller, W.H. Phase I clinical trial of i.v. ascorbic acid in advanced malignancy. Ann. Oncol. 2008, 19, 1969–1974. [Google Scholar] [CrossRef]
- Riordan, H.D.; Hunninghake, R.D.; Riordan, N.H.; Jackson, J.J.; Meng, X.; Taylor, P.; Casciari, J.J.; Gonzàlez, M.J.; Miranda-Massari, J.R.; Mora, E.M.; et al. Intravenous ascorbic acid: Protocol for its applications and use.pdf. Puerto Rico Health Sci. J. 2003, 22, 287–290. [Google Scholar]
- Nielsen, T.K.; Højgaard, M.; Andersen, J.T.; Jørgensen, N.R.; Zerahn, B.; Kristensen, B.; Henriksen, T. Weekly ascorbic acid infusion in castration-resistant prostate cancer patients: A single-arm phase II trial. Transl. Androl. Urol. 2017, 6, 517–528. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Espey, M.G.; Krishna, M.C.; Mitchell, J.B.; Corpe, C.P.; Buettner, G.R.; Shacter, E.; Levine, M. 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, 102, 13604–13609. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Espey, M.G.; Sun, A.Y.; Lee, J.-H.; Krishna, M.C.; Shacter, E.; Choyke, P.L.; Pooput, C.; Kirk, K.L.; Buettner, G.R.; et al. Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 8749–8754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Q.; Espey, M.G.; Sun, A.Y.; Pooput, C.; Kirk, K.L.; Krishna, M.C.; Khosh, D.B.; Drisko, J.; Levine, M. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 11105–11109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, J.; Martin, S.M.; Levine, M.; Wagner, B.A.; Buettner, G.R.; Wang, S.; Taghiyev, A.F.; Du, C.; Knudson, C.M.; Cullen, J.J. Mechanisms of Ascorbate-Induced Cytotoxicity in Pancreatic Cancer. Clin. Cancer Res. 2010, 16, 509–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takemura, Y.; Satoh, M.; Satoh, K.; Hamada, H.; Sekido, Y.; Kubota, S. High dose of ascorbic acid induces cell death in mesothelioma cells. Biochem. Biophys. Res. Commun. 2010, 394, 249–253. [Google Scholar] [CrossRef] [PubMed]
- Pollard, H.B.; Levine, M.A.; Eidelman, O.; Pollard, M. Pharmacological ascorbic acid suppresses syngeneic tumor growth and metastases in hormone-refractory prostate cancer. In Vivo 2010, 24, 249–255. [Google Scholar]
- Chen, P.; Yu, J.; Chalmers, B.; Drisko, J.; Yang, J.; Li, B.; Chen, Q. Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy. Anticancer Drugs 2012, 23, 437–444. [Google Scholar] [CrossRef] [PubMed]
- Venturelli, S.; Sinnberg, T.W.; Berger, A.; Noor, S.; Levesque, M.P.; Böcker, A.; Niessner, H.; Lauer, U.M.; Bitzer, M.; Garbe, C.; et al. Epigenetic Impacts of Ascorbate on Human Metastatic Melanoma Cells. Front. Oncol. 2014, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, E.; Chen, P.; Wilkins, H.M.; Wang, T.; Swerdlow, R.H.; Chen, Q. Pharmacologic ascorbate induces neuroblastoma cell death by hydrogen peroxide mediated DNA damage and reduction in cancer cell glycolysis. Free Radic. Biol. Med. 2017, 113, 36–47. [Google Scholar] [CrossRef] [PubMed]
- O’Leary, B.R.; Alexander, M.S.; Du, J.; Moose, D.L.; Henry, M.D.; Cullen, J.J. Pharmacological ascorbate inhibits pancreatic cancer metastases via a peroxide-mediated mechanism. Sci. Rep. 2020, 10, 17649. [Google Scholar] [CrossRef]
- McCarty, M.F.; Contreras, F. Increasing Superoxide Production and the Labile Iron Pool in Tumor Cells may Sensitize Them to Extracellular Ascorbate. Front. Oncol. 2014, 4, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Rivière, J.; Ravanat, J.-L.; Wagner, J.R. Ascorbate and H2O2 induced oxidative DNA damage in Jurkat cells. Free Radic. Biol. Med. 2006, 40, 2071–2079. [Google Scholar] [CrossRef] [PubMed]
- Sinnberg, T.; Noor, S.; Venturelli, S.; Berger, A.; Schuler, P.; Garbe, C.; Busch, C. The ROS-induced cytotoxicity of ascorbate is attenuated by hypoxia and HIF-1 alpha in the 60 cancer cell lines. J. Cell. Mol. Med. 2014, 18, 530–541. [Google Scholar] [CrossRef] [PubMed]
- Gibson, A.R.; O’Leary, B.R.; Du, J.; Sarsour, E.H.; Kalen, A.L.; Wagner, B.A.; Stolwijk, J.M.; Falls-Hubert, K.C.; Alexander, M.S.; Carroll, R.S.; et al. Dual Oxidase-Induced Sustained Generation of Hydrogen Peroxide Contributes to Pharmacologic Ascorbate-Induced Cytotoxicity. Cancer Res. 2020, 80, 1401–1413. [Google Scholar] [CrossRef] [PubMed]
- Buranasudja, V.; Doskey, C.M.; Gibson, A.R.; Wagner, B.A.; Du, J.; Gordon, D.J.; Koppenhafer, S.L.; Cullen, J.J.; Buettner, G.R. Pharmacologic Ascorbate Primes Pancreatic Cancer Cells for Death by Rewiring Cellular Energetics and Inducing DNA Damage. Mol. Cancer Res. 2019, 17, 2102–2114. [Google Scholar] [CrossRef] [PubMed]
- Uetaki, M.; Tabata, S.; Nakasuka, F.; Soga, T.; Tomita, M. Metabolomic alterations in human cancer cells by vitamin C-induced oxidative stress. Sci. Rep. 2015, 5, 13896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghanem, A.; Melzer, A.M.; Zaal, E.; Neises, L.; Baltissen, D.; Matar, O.; Glennemeier-Marke, H.; Almouhanna, F.; Theobald, J.; Abu el Maaty, M.A.; et al. Ascorbate kills breast cancer cells by rewiring metabolism via redox imbalance and energy crisis. Free Radic. Biol. Med. 2021, 163, 196–209. [Google Scholar] [CrossRef] [PubMed]
- Schraufstätter, I.U.; Hinshaw, D.B.; Hyslop, P.A.; Spragg, R.G.; Cochrane, C.G. Glutathione cycle activity and pyridine nucleotide levels in oxidant-induced injury of cells. J. Clin. Investig. 1985, 76, 1131–1139. [Google Scholar] [CrossRef]
- Lee, Y.; Shacter, E. Oxidative Stress Inhibits Apoptosis in Human Lymphoma Cells. J. Biol. Chem. 1999, 274, 19792–19798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.-A.; Lee, J.-W.; Kim, H.; Kim, E.-Y.; Seo, J.-M.; Ko, J.; Kim, J.-H. Pro-survival of estrogen receptor-negative breast cancer cells is regulated by a BLT2–reactive oxygen species-linked signaling pathway. Carcinogenesis 2010, 31, 543–551. [Google Scholar] [CrossRef]
- Luanpitpong, S.; Talbott, S.J.; Rojanasakul, Y.; Nimmannit, U.; Pongrakhananon, V.; Wang, L.; Chanvorachote, P. Regulation of Lung Cancer Cell Migration and Invasion by Reactive Oxygen Species and Caveolin-1. J. Biol. Chem. 2010, 285, 38832–38840. [Google Scholar] [CrossRef] [Green Version]
- Cichon, M.A.; Radisky, D.C. ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-κB-dependent activation of Snail. Oncotarget 2014, 5, 2827–2838. [Google Scholar] [CrossRef] [Green Version]
- Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020, 77, 4459–4483. [Google Scholar] [CrossRef] [PubMed]
- Wei, X.; Xu, Y.; Xu, F.F.; Chaiswing, L.; Schnell, D.; Noel, T.; Wang, C.; Chen, J.; St. Clair, D.K.; St. Clair, W.H. RelB Expression Determines the Differential Effects of Ascorbic Acid in Normal and Cancer Cells. Cancer Res. 2017, 77, 1345–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gamcsik, M.P.; Kasibhatla, M.S.; Teeter, S.D.; Colvin, O.M. Glutathione levels in human tumors. Biomarkers 2012, 17, 671–691. [Google Scholar] [CrossRef]
- Noguera, N.I.; Pelosi, E.; Angelini, D.F.; Piredda, M.L.; Guerrera, G.; Piras, E.; Battistini, L.; Massai, L.; Berardi, A.; Catalano, G.; et al. High-dose ascorbate and arsenic trioxide selectively kill acute myeloid leukemia and acute promyelocytic leukemia blasts in vitro. Oncotarget 2017, 8, 32550–32565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Desideri, E.; Ciccarone, F.; Ciriolo, M.R. Targeting Glutathione Metabolism: Partner in Crime in Anticancer Therapy. Nutrients 2019, 11, 1926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minor, E.A.; Court, B.L.; Young, J.I.; Wang, G. Ascorbate Induces Ten-Eleven Translocation (Tet) Methylcytosine Dioxygenase-mediated Generation of 5-Hydroxymethylcytosine. J. Biol. Chem. 2013, 288, 13669–13674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, R.; Mao, S.-Q.; Zhao, B.; Chong, Z.; Yang, Y.; Zhao, C.; Zhang, D.; Huang, H.; Gao, J.; Li, Z.; et al. Ascorbic Acid Enhances Tet-Mediated 5-Methylcytosine Oxidation and Promotes DNA Demethylation in Mammals. J. Am. Chem. Soc. 2013, 135, 10396–10403. [Google Scholar] [CrossRef] [PubMed]
- Dickson, K.M.; Gustafson, C.B.; Young, J.I.; Züchner, S.; Wang, G. Ascorbate-induced generation of 5-hydroxymethylcytosine is unaffected by varying levels of iron and 2-oxoglutarate. Biochem. Biophys. Res. Commun. 2013, 439, 522–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blaschke, K.; Ebata, K.T.; Karimi, M.M.; Zepeda-Martínez, J.A.; Goyal, P.; Mahapatra, S.; Tam, A.; Laird, D.J.; Hirst, M.; Rao, A.; et al. Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 2013, 500, 222–226. [Google Scholar] [CrossRef]
- Chen, J.; Guo, L.; Zhang, L.; Wu, H.; Yang, J.; Liu, H.; Wang, X.; Hu, X.; Gu, T.; Zhou, Z.; et al. Vitamin C modulates TET1 function during somatic cell reprogramming. Nat. Genet. 2013, 45, 1504–1509. [Google Scholar] [CrossRef] [PubMed]
- Agathocleous, M.; Meacham, C.E.; Burgess, R.J.; Piskounova, E.; Zhao, Z.; Crane, G.M.; Cowin, B.L.; Bruner, E.; Murphy, M.M.; Chen, W.; et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 2017, 549, 476–481. [Google Scholar] [CrossRef] [PubMed]
- Delhommeau, F.; Dupont, S.; Valle, V.D.; James, C.; Trannoy, S.; Massé, A.; Kosmider, O.; Le Couedic, J.-P.; Robert, F.; Alberdi, A.; et al. Mutation in TET2 in Myeloid Cancers. N. Engl. J. Med. 2009, 360, 2289–2301. [Google Scholar] [CrossRef] [PubMed]
- Quivoron, C.; Couronné, L.; Della Valle, V.; Lopez, C.K.; Plo, I.; Wagner-Ballon, O.; Do Cruzeiro, M.; Delhommeau, F.; Arnulf, B.; Stern, M.-H.; et al. TET2 Inactivation Results in Pleiotropic Hematopoietic Abnormalities in Mouse and Is a Recurrent Event during Human Lymphomagenesis. Cancer Cell 2011, 20, 25–38. [Google Scholar] [CrossRef] [Green Version]
- Tefferi, A.; Pardanani, A.; Lim, K.-H.; Abdel-Wahab, O.; Lasho, T.L.; Patel, J.; Gangat, N.; Finke, C.M.; Schwager, S.; Mullally, A.; et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009, 23, 905–911. [Google Scholar] [CrossRef] [Green Version]
- Cimmino, L.; Dolgalev, I.; Wang, Y.; Yoshimi, A.; Martin, G.H.; Wang, J.; Ng, V.; Xia, B.; Witkowski, M.T.; Mitchell-Flack, M.; et al. Restoration of TET2 Function Blocks Aberrant Self-Renewal and Leukemia Progression. Cell 2017, 170, 1079–1095.e20. [Google Scholar] [CrossRef] [Green Version]
- Guan, Y.; Greenberg, E.F.; Hasipek, M.; Chen, S.; Liu, X.; Kerr, C.M.; Gackowski, D.; Zarakowska, E.; Radivoyevitch, T.; Gu, X.; et al. Context dependent effects of ascorbic acid treatment in TET2 mutant myeloid neoplasia. Commun. Biol. 2020, 3, 493. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Xiao, M.; Chen, X.; Chen, L.; Xu, Y.; Lv, L.; Wang, P.; Yang, H.; Ma, S.; Lin, H.; et al. WT1 Recruits TET2 to Regulate Its Target Gene Expression and Suppress Leukemia Cell Proliferation. Mol. Cell 2015, 57, 662–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Figueroa, M.E.; Abdel-Wahab, O.; Lu, C.; Ward, P.S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H.F.; et al. Leukemic IDH1 and IDH2 Mutations Result in a Hypermethylation Phenotype, Disrupt TET2 Function, and Impair Hematopoietic Differentiation. Cancer Cell 2010, 18, 553–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.; Yang, H.; Liu, Y.; Yang, Y.; Wang, P.; Kim, S.-H.; Ito, S.; Yang, C.; Wang, P.; Xiao, M.-T.; et al. Oncometabolite 2-Hydroxyglutarate Is a Competitive Inhibitor of α-Ketoglutarate-Dependent Dioxygenases. Cancer Cell 2011, 19, 17–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gagné, L.M.; Boulay, K.; Topisirovic, I.; Huot, M.-É.; Mallette, F.A. Oncogenic Activities of IDH1/2 Mutations: From Epigenetics to Cellular Signaling. Trends Cell Biol. 2017, 27, 738–752. [Google Scholar] [CrossRef] [PubMed]
- Mingay, M.; Chaturvedi, A.; Bilenky, M.; Cao, Q.; Jackson, L.; Hui, T.; Moksa, M.; Heravi-Moussavi, A.; Humphries, R.K.; Heuser, M.; et al. Vitamin C-induced epigenomic remodelling in IDH1 mutant acute myeloid leukaemia. Leukemia 2018, 32, 11–20. [Google Scholar] [CrossRef]
- Takamizawa, S. Effects of ascorbic acid and ascorbic acid 2-phosphate, a long-acting vitamin C derivative, on the proliferation and differentiation of human osteoblast-like cells. Cell Biol. Int. 2004, 28, 255–265. [Google Scholar] [CrossRef]
- Gerecke, C.; Schumacher, F.; Berndzen, A.; Homann, T.; Kleuser, B. Vitamin C in combination with inhibition of mutant IDH1 synergistically activates TET enzymes and epigenetically modulates gene silencing in colon cancer cells. Epigenetics 2020, 15, 307–322. [Google Scholar] [CrossRef]
- Rasmussen, K.D.; Jia, G.; Johansen, J.V.; Pedersen, M.T.; Rapin, N.; Bagger, F.O.; Porse, B.T.; Bernard, O.A.; Christensen, J.; Helin, K. Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes Dev. 2015, 29, 910–922. [Google Scholar] [CrossRef] [Green Version]
- Ge, G.; Peng, D.; Xu, Z.; Guan, B.; Xin, Z.; He, Q.; Zhou, Y.; Li, X.; Zhou, L.; Ci, W. Restoration of 5-hydroxymethylcytosine by ascorbate blocks kidney tumour growth. EMBO Rep. 2018, 19. [Google Scholar] [CrossRef]
- Qiu, L.; Liu, F.; Yi, S.; Li, X.; Liu, X.; Xiao, C.; Lian, C.G.; Tu, P.; Wang, Y. Loss of 5-Hydroxymethylcytosine Is an Epigenetic Biomarker in Cutaneous T-Cell Lymphoma. J. Investig. Dermatol. 2018, 138, 2388–2397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, D.; Ge, G.; Gong, Y.; Zhan, Y.; He, S.; Guan, B.; Li, Y.; Xu, Z.; Hao, H.; He, Z.; et al. Vitamin C increases 5-hydroxymethylcytosine level and inhibits the growth of bladder cancer. Clin. Epigenet. 2018, 10, 94. [Google Scholar] [CrossRef]
- Shenoy, N.; Bhagat, T.D.; Cheville, J.; Lohse, C.; Bhattacharyya, S.; Tischer, A.; Machha, V.; Gordon-Mitchell, S.; Choudhary, G.; Wong, L.-F.; et al. Ascorbic acid–induced TET activation mitigates adverse hydroxymethylcytosine loss in renal cell carcinoma. J. Clin. Investig. 2019, 129, 1612–1625. [Google Scholar] [CrossRef] [Green Version]
- Rampal, R.; Alkalin, A.; Madzo, J.; Vasanthakumar, A.; Pronier, E.; Patel, J.; Li, Y.; Ahn, J.; Abdel-Wahab, O.; Shih, A.; et al. DNA Hydroxymethylation Profiling Reveals that WT1 Mutations Result in Loss of TET2 Function in Acute Myeloid Leukemia. Cell Rep. 2014, 9, 1841–1855. [Google Scholar] [CrossRef]
- Fischer, A.P.; Miles, S.L. Ascorbic acid, but not dehydroascorbic acid increases intracellular vitamin C content to decrease Hypoxia Inducible Factor -1 alpha activity and reduce malignant potential in human melanoma. Biomed. Pharmacother. 2017, 86, 502–513. [Google Scholar] [CrossRef] [PubMed]
- Kuiper, C.; Vissers, M.C.M. Ascorbate as a Co-Factor for Fe- and 2-Oxoglutarate Dependent Dioxygenases: Physiological Activity in Tumor Growth and Progression. Front. Oncol. 2014, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuiper, C.; Molenaar, I.G.M.; Dachs, G.U.; Currie, M.J.; Sykes, P.H.; Vissers, M.C.M. Low Ascorbate Levels Are Associated with Increased Hypoxia-Inducible Factor-1 Activity and an Aggressive Tumor Phenotype in Endometrial Cancer. Cancer Res. 2010, 70, 5749–5758. [Google Scholar] [CrossRef] [Green Version]
- Campbell, E.J.; Vissers, M.C.M.; Wohlrab, C.; Hicks, K.O.; Strother, R.M.; Bozonet, S.M.; Robinson, B.A.; Dachs, G.U. Pharmacokinetic and anti-cancer properties of high dose ascorbate in solid tumours of ascorbate-dependent mice. Free Radic. Biol. Med. 2016, 99, 451–462. [Google Scholar] [CrossRef]
- Wohlrab, C.; Kuiper, C.; Vissers, M.C.; Phillips, E.; Robinson, B.A.; Dachs, G.U. Ascorbate modulates the hypoxic pathway by increasing intracellular activity of the HIF hydroxylases in renal cell carcinoma cells. Hypoxia 2019, 7, 17–31. [Google Scholar] [CrossRef] [Green Version]
- Kuiper, C.; Dachs, G.U.; Munn, D.; Currie, M.J.; Robinson, B.A.; Pearson, J.F.; Vissers, M.C.M. Increased Tumor Ascorbate is Associated with Extended Disease-Free Survival and Decreased Hypoxia-Inducible Factor-1 Activation in Human Colorectal Cancer. Front. Oncol. 2014, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campbell, E.J.; Dachs, G.U.; Morrin, H.R.; Davey, V.C.; Robinson, B.A.; Vissers, M.C.M. Activation of the hypoxia pathway in breast cancer tissue and patient survival are inversely associated with tumor ascorbate levels. BMC Cancer 2019, 19, 307. [Google Scholar] [CrossRef] [PubMed]
- Jóźwiak, P.; Ciesielski, P.; Zaczek, A.; Lipińska, A.; Pomorski, L.; Wieczorek, M.; Bryś, M.; Forma, E.; Krześlak, A. Expression of hypoxia inducible factor 1α and 2α and its association with vitamin C level in thyroid lesions. J. Biomed. Sci. 2017, 24, 83. [Google Scholar] [CrossRef] [Green Version]
- Song, K.; Li, M.; Xu, X.-J.; Xuan, L.; Huang, G.-N.; Song, X.-L.; Liu, Q.-F. HIF-1α and GLUT1 Gene Expression is Associated with Chemoresistance of Acute Myeloid Leukemia. Asian Pac. J. Cancer Prev. 2014, 15, 1823–1829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischer, A.P.; Miles, S.L. Silencing HIF-1α induces TET2 expression and augments ascorbic acid induced 5-hydroxymethylation of DNA in human metastatic melanoma cells. Biochem. Biophys. Res. Commun. 2017, 490, 176–181. [Google Scholar] [CrossRef] [PubMed]
- Knowles, H.J.; Raval, R.R.; Harris, A.L.; Ratcliffe, P.J. Effect of ascorbate on the activity of hypoxia-inducible factor in cancer cells. Cancer Res. 2003, 63, 1764–1768. [Google Scholar]
- Kawada, H.; Kaneko, M.; Sawanobori, M.; Uno, T.; Matsuzawa, H.; Nakamura, Y.; Matsushita, H.; Ando, K. High concentrations of L-ascorbic acid specifically inhibit the growth of human leukemic cells via downregulation of HIF-1α transcription. PLoS ONE 2013, 8, e62717. [Google Scholar] [CrossRef] [Green Version]
- Miles, S.L.; Fischer, A.P.; Joshi, S.J.; Niles, R.M. Ascorbic acid and ascorbate-2-phosphate decrease HIF activity and malignant properties of human melanoma cells. BMC Cancer 2015, 15, 867. [Google Scholar] [CrossRef] [Green Version]
- Wilkes, J.G.; O’Leary, B.R.; Du, J.; Klinger, A.R.; Sibenaller, Z.A.; Doskey, C.M.; Gibson-Corley, K.N.; Alexander, M.S.; Tsai, S.; Buettner, G.R.; et al. Pharmacologic ascorbate (P-AscH−) suppresses hypoxia-inducible Factor-1α (HIF-1α) in pancreatic adenocarcinoma. Clin. Exp. Metastasis 2018, 35, 37–51. [Google Scholar] [CrossRef] [Green Version]
- Wohlrab, C.; Vissers, M.C.M.; Phillips, E.; Morrin, H.; Robinson, B.A.; Dachs, G.U. The Association Between Ascorbate and the Hypoxia-Inducible Factors in Human Renal Cell Carcinoma Requires a Functional Von Hippel-Lindau Protein. Front. Oncol. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
- Tian, W.; Wang, Y.; Xu, Y.; Guo, X.; Wang, B.; Sun, L.; Liu, L.; Cui, F.; Zhuang, Q.; Bao, X.; et al. The Hypoxia-inducible Factor Renders Cancer Cells More Sensitive to Vitamin C-induced Toxicity. J. Biol. Chem. 2014, 289, 3339–3351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dachs, G.U.; Gandhi, J.; Wohlrab, C.; Carr, A.C.; Morrin, H.R.; Pullar, J.M.; Bayer, S.B.; Eglinton, T.W.; Robinson, B.A.; Vissers, M.C.M. Vitamin C Administration by Intravenous Infusion Increases Tumor Ascorbate Content in Patients With Colon Cancer: A Clinical Intervention Study. Front. Oncol. 2021, 10. [Google Scholar] [CrossRef]
- Ebata, K.T.; Mesh, K.; Liu, S.; Bilenky, M.; Fekete, A.; Acker, M.G.; Hirst, M.; Garcia, B.A.; Ramalho-Santos, M. Vitamin C induces specific demethylation of H3K9me2 in mouse embryonic stem cells via Kdm3a/b. Epigenet. Chromatin 2017, 10, 36. [Google Scholar] [CrossRef]
- Huang, R.; Wang, Y.; Ge, H.; Wang, D.; Wang, Y.; Zhang, W.; Yang, J.; Cheng, J. Restoration of TET2 deficiency inhibits tumor growth in head neck squamous cell carcinoma. Ann. Transl. Med. 2020, 8, 329. [Google Scholar] [CrossRef]
- Ma, Y.; Chen, P.; Drisko, J.; Khabele, D.; Godwin, A.; Chen, Q. Pharmacological ascorbate induces ‘BRCAness’ and enhances the effects of Poly(ADP-Ribose) polymerase inhibitors against BRCA1/2 wild-type ovarian cancer. Oncol. Lett. 2020, 19. [Google Scholar] [CrossRef] [Green Version]
- Bachman, M.; Uribe-Lewis, S.; Yang, X.; Williams, M.; Murrell, A.; Balasubramanian, S. 5-Hydroxymethylcytosine is a predominantly stable DNA modification. Nat. Chem. 2014, 6, 1049–1055. [Google Scholar] [CrossRef] [Green Version]
- Cimmino, L.; Neel, B.G.; Aifantis, I. Vitamin C in Stem Cell Reprogramming and Cancer. Trends Cell Biol. 2018, 28, 698–708. [Google Scholar] [CrossRef] [PubMed]
- Starczak, M.; Zarakowska, E.; Modrzejewska, M.; Dziaman, T.; Szpila, A.; Linowiecka, K.; Guz, J.; Szpotan, J.; Gawronski, M.; Labejszo, A.; et al. In vivo evidence of ascorbate involvement in the generation of epigenetic DNA modifications in leukocytes from patients with colorectal carcinoma, benign adenoma and inflammatory bowel disease. J. Transl. Med. 2018, 16, 204. [Google Scholar] [CrossRef] [PubMed]
- Sant, D.W.; Mustafi, S.; Gustafson, C.B.; Chen, J.; Slingerland, J.M.; Wang, G. Vitamin C promotes apoptosis in breast cancer cells by increasing TRAIL expression. Sci. Rep. 2018, 8, 5306. [Google Scholar] [CrossRef]
- Siref, A.; McCormack, C.; Huang, Q.; Lim, W.; Alkan, S. Diminished expression of 5hmc in Reed-Sternberg cells in classical Hodgkin lymphoma is a common epigenetic marker. Leuk. Res. 2020, 96, 106408. [Google Scholar] [CrossRef] [PubMed]
- Haffner, M.C.; Chaux, A.; Meeker, A.K.; Esopi, D.M.; Gerber, J.; Pellakuru, L.G.; Toubaji, A.; Argani, P.; Iacobuzio-Donahue, C.; Nelson, W.G.; et al. Global 5-hydroxymethylcytosine content is significantly reduced in tissue stem/progenitor cell compartments and in human cancers. Oncotarget 2011, 2, 627–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kudo, Y.; Tateishi, K.; Yamamoto, K.; Yamamoto, S.; Asaoka, Y.; Ijichi, H.; Nagae, G.; Yoshida, H.; Aburatani, H.; Koike, K. Loss of 5-hydroxymethylcytosine is accompanied with malignant cellular transformation. Cancer Sci. 2012, 103, 670–676. [Google Scholar] [CrossRef] [PubMed]
- Lian, C.G.; Xu, Y.; Ceol, C.; Wu, F.; Larson, A.; Dresser, K.; Xu, W.; Tan, L.; Hu, Y.; Zhan, Q.; et al. Loss of 5-Hydroxymethylcytosine Is an Epigenetic Hallmark of Melanoma. Cell 2012, 150, 1135–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gustafson, C.B.; Yang, C.; Dickson, K.M.; Shao, H.; Van Booven, D.; Harbour, J.W.; Liu, Z.-J.; Wang, G. Epigenetic reprogramming of melanoma cells by vitamin C treatment. Clin. Epigenetics 2015, 7, 51. [Google Scholar] [CrossRef] [Green Version]
- Shenoy, N.; Bhagat, T.; Nieves, E.; Stenson, M.; Lawson, J.; Choudhary, G.S.; Habermann, T.; Nowakowski, G.; Singh, R.; Wu, X.; et al. Upregulation of TET activity with ascorbic acid induces epigenetic modulation of lymphoma cells. Blood Cancer J. 2017, 7, e587. [Google Scholar] [CrossRef] [Green Version]
- Mustafi, S.; Sant, D.W.; Liu, Z.-J.; Wang, G. Ascorbate induces apoptosis in melanoma cells by suppressing Clusterin expression. Sci. Rep. 2017, 7, 3671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thienpont, B.; Steinbacher, J.; Zhao, H.; D’Anna, F.; Kuchnio, A.; Ploumakis, A.; Ghesquière, B.; Van Dyck, L.; Boeckx, B.; Schoonjans, L.; et al. Tumour hypoxia causes DNA hypermethylation by reducing TET activity. Nature 2016, 537, 63–68. [Google Scholar] [CrossRef]
- Pomerleau, J.; Weidmann, C.; Coutant, K.; Lowry, C.-M.; Veilleux, M.-P.; Bérubé, J.; Wagner, J.R.; Landreville, S. Experimental eye research/short communication format characterization of DNA hydroxymethylation in the ocular choroid. Exp. Eye Res. 2021, 205, 108473. [Google Scholar] [CrossRef] [PubMed]
- Ficz, G.; Gribben, J.G. Loss of 5-hydroxymethylcytosine in cancer: Cause or consequence? Genomics 2014, 104, 352–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, Y.; Wang, G.; Liang, Z.; Yang, Y.; Cui, L.; Liu, C.-Y. Loss of nuclear localization of TET2 in colorectal cancer. Clin. Epigenetics 2016, 8, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, N.P. Sodium ascorbate induces DNA single-strand breaks in human cells in vitro. Mutat. Res. Mol. Mech. Mutagen. 1997, 375, 195–203. [Google Scholar] [CrossRef]
- Kharat, S.S.; Ding, X.; Swaminathan, D.; Suresh, A.; Singh, M.; Sengodan, S.K.; Burkett, S.; Marks, H.; Pamala, C.; He, Y.; et al. Degradation of 5hmC-marked stalled replication forks by APE1 causes genomic instability. Sci. Signal. 2020, 13, eaba8091. [Google Scholar] [CrossRef] [PubMed]
- Faraoni, I.; Graziani, G. Role of BRCA Mutations in Cancer Treatment with Poly(ADP-ribose) Polymerase (PARP) Inhibitors. Cancers 2018, 10, 487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shah, G.M.; Robu, M.; Purohit, N.K.; Rajawat, J.; Tentori, L.; Graziani, G. PARP Inhibitors in Cancer Therapy: Magic Bullets but Moving Targets. Front. Oncol. 2013, 3. [Google Scholar] [CrossRef] [Green Version]
- Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
- Slade, D. PARP and PARG inhibitors in cancer treatment. Genes Dev. 2020, 34, 360–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ray Chaudhuri, A.; Callen, E.; Ding, X.; Gogola, E.; Duarte, A.A.; Lee, J.-E.; Wong, N.; Lafarga, V.; Calvo, J.A.; Panzarino, N.J.; et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 2016, 535, 382–387. [Google Scholar] [CrossRef] [PubMed]
- Mastrangelo, D.; Massai, L.; Fioritoni, G.; Iacone, A.; Bartolomeo, P.D.; Accorsi, P.; Bonfini, T.; Muscettola, M.; Grasso, G. Megadoses of sodium ascorbate efficiently kill HL60 cells in vitro: Comparison with arsenic trioxide. J. Cancer Ther. 2013, 4, 1366–1372. [Google Scholar] [CrossRef] [Green Version]
- Mastrangelo, D.; Pelosi, E.; Castelli, G.; Lo-Coco, F.; Testa, U. Mechanisms of anti-cancer effects of ascorbate: Cytotoxic activity and epigenetic modulation. Blood Cells, Mol. Dis. 2018, 69, 57–64. [Google Scholar] [CrossRef] [PubMed]
- Polireddy, K.; Dong, R.; Reed, G.; Yu, J.; Chen, P.; Williamson, S.; Violet, P.-C.; Pessetto, Z.; Godwin, A.K.; Fan, F.; et al. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Sci. Rep. 2017, 7, 17188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Du, J.; Wagner, B.A.; Buettner, G.R.; Cullen, J.J. Role of labile iron in the toxicity of pharmacological ascorbate. Free Radic. Biol. Med. 2015, 84, 289–295. [Google Scholar] [CrossRef] [Green Version]
- Schoenfeld, J.D.; Sibenaller, Z.A.; Mapuskar, K.A.; Wagner, B.A.; Cramer-Morales, K.L.; Furqan, M.; Sandhu, S.; Carlisle, T.L.; Smith, M.C.; Abu Hejleh, T.; et al. O2− and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell 2017, 31, 487–500.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deubzer, B.; Mayer, F.; Kuçi, Z.; Niewisch, M.; Merkel, G.; Handgretinger, R.; Bruchelt, G. H2O2-mediated cytotoxicity of pharmacologic ascorbate concentrations to neuroblastoma cells: Potential role of lactate and ferritin. Cell. Physiol. Biochem. 2010, 25. [Google Scholar] [CrossRef] [PubMed]
- Gregoraszczuk, E.L.; Zajda, K.; Tekla, J.; Respekta, N.; Zdybał, P.; Such, A. Vitamin C supplementation had no side effect in non-cancer, but had anticancer properties in ovarian cancer cells. Int. J. Vitam. Nutr. Res. 2020, 1–11. [Google Scholar] [CrossRef]
- Waldo, A.L.; Zipf, R.E. Ascorbic acid level in leukemic patients. Cancer 1955, 8, 187–190. [Google Scholar] [CrossRef]
- Liu, M.; Ohtani, H.; Zhou, W.; Ørskov, A.D.; Charlet, J.; Zhang, Y.W.; Shen, H.; Baylin, S.B.; Liang, G.; Grønbæk, K.; et al. Vitamin C increases viral mimicry induced by 5-aza-2′-deoxycytidine. Proc. Natl. Acad. Sci. USA 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, R.; Nonis, M.; Pearson, J.F.; Burgess, E.; Morrin, H.R.; Pullar, J.M.; Spencer, E.; Vissers, M.C.M.; Robinson, B.A.; Dachs, G.U. Low Vitamin C Status in Patients with Cancer Is Associated with Patient and Tumor Characteristics. Nutrients 2020, 12, 2338. [Google Scholar] [CrossRef]
- Barkhan, P.; Howard, A.N. Distribution of ascorbic acid in normal and leukaemic human blood. Biochem. J. 1958, 70, 163–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anthony, H.M.; Schorah, C.J. Severe hypovitaminosis C in lung-cancer patients: The utilization of vitamin C in surgical repair and lymphocyte-related host resistance. Br. J. Cancer 1982, 46, 354–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yamamoto, Y.; Yamashita, S.; Fujisawa, A.; Kokura, S.; Yoshikawa, T. Oxidative Stress in Patients with Hepatitis, Cirrhosis, and Hepatoma Evaluated by Plasma Antioxidants. Biochem. Biophys. Res. Commun. 1998, 247, 166–170. [Google Scholar] [CrossRef]
- Esme, H.; Cemek, M.; Sezer, M.; Saglam, H.; Demir, A.; Melek, H.; Unlu, M. High levels of oxidative stress in patients with advanced lung cancer. Respirology 2008. [Google Scholar] [CrossRef]
- Gupta, A.; Bhatt, M.L.B.; Misra, M.K. Lipid Peroxidation and Antioxidant Status in Head and Neck Squamous Cell Carcinoma Patients. Oxid. Med. Cell. Longev. 2009, 2, 68–72. [Google Scholar] [CrossRef] [Green Version]
- Schleich, T.; Rodemeister, S.; Venturelli, S.; Sinnberg, T.; Garbe, C.; Busch, C. Decreased Plasma Ascorbate Levels in Stage IV Melanoma Patients. Metab. Nutr. Oncol. 2013, 1, e2–e6. [Google Scholar] [CrossRef]
- Hu, F.; Wu, Z.; Li, G.; Teng, C.; Liu, Y.; Wang, F.; Zhao, Y.; Pang, D. The plasma level of retinol, vitamins A, C and α-tocopherol could reduce breast cancer risk? A meta-analysis and meta-regression. J. Cancer Res. Clin. Oncol. 2015, 141, 601–614. [Google Scholar] [CrossRef] [PubMed]
- Huijskens, M.J.A.J.; Wodzig, W.K.W.H.; Walczak, M.; Germeraad, W.T.V.; Bos, G.M.J. Ascorbic acid serum levels are reduced in patients with hematological malignancies. Results Immunol. 2016, 6, 8–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harrison, F.E.; May, J.M. Vitamin C function in the brain: Vital role of the ascorbate transporter SVCT2. Free Radic. Biol. Med. 2009, 46, 719–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Honegger, C.G.; Torhorst, J.; Langemann, H.; Kabiersch, A.; Krenger, W. Quantitative determination of water-soluble scavengers in neoplastic and non-neoplastic human breast tissue. Int. J. Cancer 1988, 41, 690–694. [Google Scholar] [CrossRef]
- Langemann, H.; Torhorst, J.; Kabiersch, A.; Krenger, W.; Honegger, C.G. Quantitative determination of water- and lipid-soluble antioxidants in neoplastic and non-neoplastic human breast tissue. Int. J. Cancer 1989, 43, 1169–1173. [Google Scholar] [CrossRef] [PubMed]
- Peña, E.; Roa, F.J.; Inostroza, E.; Sotomayor, K.; González, M.; Gutierrez-Castro, F.A.; Maurin, M.; Sweet, K.; Labrousse, C.; Gatica, M.; et al. Increased expression of mitochondrial sodium-coupled ascorbic acid transporter-2 (mitSVCT2) as a central feature in breast cancer. Free Radic. Biol. Med. 2019, 135, 283–292. [Google Scholar] [CrossRef] [PubMed]
- Di Tano, M.; Raucci, F.; Vernieri, C.; Caffa, I.; Buono, R.; Fanti, M.; Brandhorst, S.; Curigliano, G.; Nencioni, A.; de Braud, F.; et al. Synergistic effect of fasting-mimicking diet and vitamin C against KRAS mutated cancers. Nat. Commun. 2020, 11, 2332. [Google Scholar] [CrossRef]
- Lorenzato, A.; Magrì, A.; Matafora, V.; Audrito, V.; Arcella, P.; Lazzari, L.; Montone, M.; Lamba, S.; Deaglio, S.; Siena, S.; et al. Vitamin C Restricts the Emergence of Acquired Resistance to EGFR-Targeted Therapies in Colorectal Cancer. Cancers 2020, 12, 685. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Hong, J.; Han, H.; Park, J.; Kim, D.; Park, H.; Ko, M.; Koh, Y.; Shin, D.-Y.; Yoon, S.-S. Decreased vitamin C uptake mediated by SLC2A3 promotes leukaemia progression and impedes TET2 restoration. Br. J. Cancer 2020, 122, 1445–1452. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.-W.; Lee, S.-H.; Moon, J.-H.; Hwang, J.J.; Kim, D.E.; Ko, E.; Kim, H.-S.; Cho, I.J.; Kang, J.S.; Kim, D.J.; et al. SVCT-2 in breast cancer acts as an indicator for L-ascorbate treatment. Oncogene 2013, 32, 1508–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Washko, P.W.; Wang, Y.; Levine, M. Ascorbic acid recycling in human neutrophils. J. Biol. Chem. 1993, 268, 15531–15535. [Google Scholar] [CrossRef]
- Bozonet, S.M.; Carr, A.C. The role of physiological vitamin c concentrations on key functions of neutrophils isolated from healthy individuals. Nutrients 2019, 11, 1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corpe, C.P.; Lee, J.-H.; Kwon, O.; Eck, P.; Narayanan, J.; Kirk, K.L.; Levine, M. 6-Bromo-6-deoxy-L-ascorbic acid: An ascorbate analog specific for Na+ -dependent vitamin C transporter but not glucose transporter pathways. J. Biol. Chem. 2005, 280, 5211–5220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wright, M.E.; Andreotti, G.; Lissowska, J.; Yeager, M.; Zatonski, W.; Chanock, S.J.; Chow, W.-H.; Hou, L. Genetic variation in sodium-dependent ascorbic acid transporters and risk of gastric cancer in Poland. Eur. J. Cancer 2009, 45, 1824–1830. [Google Scholar] [CrossRef] [Green Version]
- Duell, E.J.; Lujan-Barroso, L.; Llivina, C.; Muñoz, X.; Jenab, M.; Boutron-Ruault, M.-C.; Clavel-Chapelon, F.; Racine, A.; Boeing, H.; Buijsse, B.; et al. Vitamin C transporter gene (SLC23A1 and SLC23A2) polymorphisms, plasma vitamin C levels, and gastric cancer risk in the EPIC cohort. Genes Nutr. 2013, 8, 549–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linowiecka, K.; Foksinski, M.; Brożyna, A.A. Vitamin C Transporters and Their Implications in Carcinogenesis. Nutrients 2020, 12, 3869. [Google Scholar] [CrossRef] [PubMed]
- Gill, H.S.; Yim, R.; Kumana, C.R.; Tse, E.; Kwong, Y. Oral arsenic trioxide, all-trans retinoic acid, and ascorbic acid maintenance after first complete remission in acute promyelocytic leukemia: Long-term results and unique prognostic indicators. Cancer 2020, 126, 3244–3254. [Google Scholar] [CrossRef] [PubMed]
- Hoffer, L.J.; Robitaille, L.; Zakarian, R.; Melnychuk, D.; Kavan, P.; Agulnik, J.; Cohen, V.; Small, D.; Miller, W.H. High-Dose Intravenous Vitamin C Combined with Cytotoxic Chemotherapy in Patients with Advanced Cancer: A Phase I-II Clinical Trial. PLoS ONE 2015, 10, e0120228. [Google Scholar] [CrossRef] [PubMed]
- Cameron, E.; Pauling, L. Supplemental ascorbate in the supportive treatment of cancer: Reevaluation of prolongation of survival times in terminal human cancer. Proc. Natl. Acad. Sci. USA 1978. [Google Scholar] [CrossRef] [Green Version]
- Das, A.B.; Smith-Díaz, C.C.; Vissers, M.C.M. Emerging epigenetic therapeutics for myeloid leukemia: Modulating demethylase activity with ascorbate. Haematologica 2020, 106. [Google Scholar] [CrossRef] [PubMed]
- Vető, B.; Szabó, P.; Bacquet, C.; Apró, A.; Hathy, E.; Kiss, J.; Réthelyi, J.M.; Szeri, F.; Szüts, D.; Arányi, T. Inhibition of DNA methyltransferase leads to increased genomic 5-hydroxymethylcytosine levels in hematopoietic cells. FEBS Open Bio 2018, 8, 584–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gillberg, L.; Ørskov, A.D.; Nasif, A.; Ohtani, H.; Madaj, Z.; Hansen, J.W.; Rapin, N.; Mogensen, J.B.; Liu, M.; Dufva, I.H.; et al. Oral vitamin C supplementation to patients with myeloid cancer on azacitidine treatment: Normalization of plasma vitamin C induces epigenetic changes. Clin. Epigenet. 2019, 11, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sajadian, S.O.; Tripura, C.; Samani, F.S.; Ruoss, M.; Dooley, S.; Baharvand, H.; Nussler, A.K. Vitamin C enhances epigenetic modifications induced by 5-azacytidine and cell cycle arrest in the hepatocellular carcinoma cell lines HLE and Huh7. Clin. Epigenet. 2016, 8, 46. [Google Scholar] [CrossRef] [Green Version]
- Iancu, I.V.; Botezatu, A.; Plesa, A.; Huica, I.; Fudulu, A.; Albulescu, A.; Bostan, M.; Mihaila, M.; Grancea, C.; Manda, D.A.; et al. Alterations of regulatory factors and DNA methylation pattern in thyroid cancer. Cancer Biomark. 2020, 28, 255–268. [Google Scholar] [CrossRef] [PubMed]
- Gerecke, C.; Schumacher, F.; Edlich, A.; Wetzel, A.; Yealland, G.; Neubert, L.K.; Scholtka, B.; Homann, T.; Kleuser, B. Vitamin C promotes decitabine or azacytidine induced DNA hydroxymethylation and subsequent reactivation of the epigenetically silenced tumour suppressor CDKN1A in colon cancer cells. Oncotarget 2018, 9, 32822–32840. [Google Scholar] [CrossRef] [Green Version]
- Zhao, H.; Zhu, H.; Huang, J.; Zhu, Y.; Hong, M.; Zhu, H.; Zhang, J.; Li, S.; Yang, L.; Lian, Y.; et al. The synergy of Vitamin C with decitabine activates TET2 in leukemic cells and significantly improves overall survival in elderly patients with acute myeloid leukemia. Leuk. Res. 2018, 66, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Giansanti, M.; Faraoni, I.; Prete, S.P.; Karimi, T.; Divona, M.; Ottone, T.; Consalvo, M.I.; Voso, M.T.; Graziani, G. Poly(ADP-ribose) polymerase inhibitors synergize with ascorbate and hypomethylating agents in arsenic trioxide-resistant promyelocytic leukaemia cells. Astract Book, Proceedings of the XVI Congress of the Italian Society of Experimental Hematology, Napoli, Italy, 15–17 October 2020. Haematologica. 2020, 105(s2), p. S65, abstract n°C079. Available online: https://haematologica.org/article/view/haematol.2020.s2 (accessed on 15 February 2021).
- Demiray, M. Combinatorial Therapy of High Dose Vitamin C and PARP Inhibitors in DNA Repair Deficiency: A Series of 8 Patients. Integr. Cancer Ther. 2020, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Bazzan, A.J.; Zabrecky, G.; Wintering, N.; Newberg, A.B.; Monti, D.A. Retrospective Evaluation of Clinical Experience With Intravenous Ascorbic Acid in Patients With Cancer. Integr. Cancer Ther. 2018, 17, 912–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nauman, G.; Gray, J.; Parkinson, R.; Levine, M.; Paller, C. Systematic Review of Intravenous Ascorbate in Cancer Clinical Trials. Antioxidants 2018, 7, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padayatty, S.J.; Sun, A.Y.; Chen, Q.; Espey, M.G.; Drisko, J.; Levine, M. Vitamin C: Intravenous Use by Complementary and Alternative Medicine Practitioners and Adverse Effects. PLoS ONE 2010, 5, e11414. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.; Bae, O.-N.; Koh, S.-H.; Kang, S.; Lim, K.-M.; Noh, J.-Y.; Shin, S.; Kim, I.; Chung, J.-H. High-Dose Vitamin C Injection to Cancer Patients May Promote Thrombosis Through Procoagulant Activation of Erythrocytes. Toxicol. Sci. 2015, 147, 350–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Vitamin C Dose | Plasma Concentration | References |
---|---|---|
μM | ||
0.075–0.125 g/d Normal intake ~0.030 g/d Depletion 0.010 g/d Deficiency | 40–80 11.4–28.4 <11.4 | [4,5,6,7,53,61] |
Oral route | ||
Dietary intake 0.2–0.3 g/d (fruits and vegetables) | 70–85 | [53] |
Single dose 0.2–1.25–3 g/d | 90–157–206 | [53] |
Maximum tolerated dose 18 g/d (3 g every 4 h) | ≤220 | [53] |
Oral daily dose: | [4] | |
0.03 g | 8.7 ± 0.8 | |
0.1 g | 55.9 ± 3.6 | |
0.2 g | 65.7 ± 3.8 | |
0.4 g | 70.0 ± 4.4 | |
I.V. route | mM | |
1.25 g | 0.9 ± 0.2 | [4,53] |
3 g | 1.8 | |
5 g | 2.9 | |
10 g | 5.6 | |
50 g | 13.4 | |
100 g | 15.4 | |
30 g/m2 | 23 ± 9 | [57] |
70 g/m2 | 49 ± 8 | |
90 g/m2 | 49 ± 14 | |
0.1 g/kg (~3.7 g/m2) | 2.4 ± 0.3 | [62] |
0.2 g/kg (~7.4 g/m2) | 4.7 ± 0.5 | |
0.4 g/kg (~14.8 g/m2) | 8.5 ± 0.6 | |
0.6 g/kg (~22.2 g/m2) | 11.3 ± 2.4 | |
0.9 g/kg (~33.3 g/m2) | 17.0 ± 3.6 | |
1.5 g/kg (~55.5 g/m2) | 26.2 ± 4.9 | |
I.V. infusion (grams/wk) plus oral dose (0.5 g/d) | ||
5 g week 1 | 1.9 ± 0.4 | [50,63,64] |
30 g week 2 | 12.6 ± 3.4 | |
60 g week 3 | 19.6 ± 6.8 | |
60 g week 4 | 21.1 ± 5.0 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Giansanti, M.; Karimi, T.; Faraoni, I.; Graziani, G. High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients. Cancers 2021, 13, 1428. https://doi.org/10.3390/cancers13061428
Giansanti M, Karimi T, Faraoni I, Graziani G. High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients. Cancers. 2021; 13(6):1428. https://doi.org/10.3390/cancers13061428
Chicago/Turabian StyleGiansanti, Manuela, Terry Karimi, Isabella Faraoni, and Grazia Graziani. 2021. "High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients" Cancers 13, no. 6: 1428. https://doi.org/10.3390/cancers13061428