1
|
Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev 2025; 77:100043. [PMID: 39986139 DOI: 10.1016/j.pharmr.2025.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
Ascorbic acid, the reduced form of vitamin C, is a ubiquitous small carbohydrate. Despite decades of focused research, new metabolic functions of this universal electron donor are still being discovered and add to the complexity of our view of vitamin C in human health. Although praised as an unsurpassed water-soluble antioxidant in plasma and cells, the most interesting functions of vitamin C seem to be its roles as specific electron donor in numerous biological reactions ranging from the well-known hydroxylation of proline to cofactor for the epigenetic master regulators ten-eleven translocation enzymes and Jumonji domain-containing histone-lysine demethylases. Some of these functions may have important implications for disease prevention and treatment and have spiked renewed interest in, eg, vitamin C's potential in cancer therapy. Moreover, some fundamental pharmacokinetic properties of vitamin C remain to be established including if other mechanisms than passive diffusion governs the efflux of ascorbate anions from the cell. Taken together, there still seems to be much to learn about the pharmacology of vitamin C and its role in health and disease. This review explores new avenues of vitamin C and integrates our present knowledge of its pharmacology. SIGNIFICANCE STATEMENT: Vitamin C is involved in multiple biological reactions of which most are essential to human health. Hundreds of millions of people are considered deficient in vitamin C according to accepted guidelines, but little is known about the long-term consequences. Although the complexity of vitamin C's physiology and pharmacology has been widely disregarded in clinical studies for decades, it seems clear that a deeper understanding of particularly its pharmacology holds the key to unravel and possibly exploit the potential of vitamin C in disease prevention and therapy.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Pernille Tveden-Nyborg
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Gilloteaux J, Jamison JM, Summers JL, Taper HS. Reactivation of nucleases with peroxidation damages induced by a menadione: ascorbate combination devastates human prostate carcinomas: ultrastructural aspects. Ultrastruct Pathol 2024; 48:378-421. [PMID: 39105605 DOI: 10.1080/01913123.2024.2379300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Xenografts of androgen-independent human DU145 prostate metastatic carcinomas implanted in nu/nu male mice have revealed a significant survival after a prooxidant anticancer treatment consisting of a combination of menadione bisulfite and sodium ascorbate (VK3:VC). METHODS Implanted samples of diaphragm carcinomas from longest survived mice from either oral, intraperitoneal (IP), or both oral and IP treatment groups were assessed with light, scanning, and transmission electron microscopy to analyze morphologic damages. RESULTS Compared with previous fine structure data of in vitro untreated carcinomas, the changes induced by oral, IP, and oral with IP VK3:VC treatment dismantled those xenografts with autoschizis, and necrotic atrophy was accomplished by cell's oxidative stress whose injuries were consequent to reactivated deoxyribonucleases and ribonucleases. Tumor destructions resulted from irreversible damages of nucleus components, endoplasmic reticulum, and mitochondria there. Other alterations included those of the cytoskeleton that resulted in characteristic self-excisions named " autoschizis." All these injuries lead resilient cancer cells to necrotic cell death. CONCLUSION The fine structure damages caused by VK3:VC prooxidant combination in the human DU145 prostate xenografts confirmed those shown in vitro and of other cell lines with histochemistry and biomolecular investigations. These devastations incurred without damage to normal tissues; thus, our data brought support for the above combination to assist in the treatment of prostate cancers and other cancers.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- Department of Anatomical Sciences, St Georges' University International School of Medicine, Newcastle upon Tyne, UK
- Department of Anatomical Sciences, NEOMed (NEOUCOM), Rootstown, Ohio, USA
- Department of Medicine, Unit of Research in Molecular Physiology (URPhyM), NARILIS, Université de Namur, Namur, Belgium
| | - James M Jamison
- Department of Urology, Summa Health System, Akron, Ohio, USA
- St Thomas Hospital, The Apatone Development Center, Summa Research Fondation, Akron Ohio, USA
| | - Jack L Summers
- Department of Urology, Summa Health System, Akron, Ohio, USA
- St Thomas Hospital, The Apatone Development Center, Summa Research Fondation, Akron Ohio, USA
| | - Henryk S Taper
- Département des Sciences Pharmaceutiques, Unité de Pharmacocinétique, Métabolisme, Nutrition et Toxicologie, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
3
|
Shao G, Liu Y, Lu L, Wang L, Ji G, Xu H. Therapeutic potential of traditional Chinese medicine in the prevention and treatment of digestive inflammatory cancer transformation: Portulaca oleracea L. as a promising drug. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:117999. [PMID: 38447616 DOI: 10.1016/j.jep.2024.117999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been used for centuries to treat various types of inflammation and tumors of the digestive system. Portulaca oleracea L. (POL), has been used in TCM for thousands of years. The chemical composition of POL is variable and includes flavonoids, alkaloids, terpenoids and organic acids and other classes of natural compounds. Many of these compounds exhibit powerful anti-inflammatory and anti-cancer-transforming effects in the digestive system. AIM OF STUDY In this review, we focus on the potential therapeutic role of POL in NASH, gastritis and colitis and their associated cancers, with a focus on the pharmacological properties and potential mechanisms of action of the main natural active compounds in POL. METHODS The information and data on Portulaca oleracea L. and its main active ingredients were collated from various resources like ethnobotanical textbooks and literature databases such as CNKI, VIP (Chinese literature), PubMed, Science Direct, Elsevier and Google Scholar (English literatures), Wiley, Springer, Tailor and Francis, Scopus, Inflibnet. RESULTS Kaempferol, luteolin, myricetin, quercetin, genistein, EPA, DHA, and melatonin were found to improve NASH and NASH-HCC, while kaempferol, apigenin, luteolin, and quercetin played a therapeutic role in gastritis and gastric cancer. Apigenin, luteolin, myricetin, quercetin, genistein, lupeol, vitamin C and melatonin were found to have therapeutic effects in the treatment of colitis and its associated cancers. The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. CONCLUSION The discovery of the beneficial effects of these natural active compounds in POL supports the idea that POL could be a promising novel candidate for the treatment and prevention of inflammation-related cancers of the digestive system. However, clinical data describing the mode of action of the naturally active compounds of POL are still lacking. In addition, pharmacokinetic data for POL compounds, such as changes in drug dose and absorption rates, cannot be extrapolated from animal models and need to be measured in patients in clinical trials. On the one hand, a systematic meta-analysis of the existing publications on TCM containing POL still needs to be carried out. On the other hand, studies on the hepatic and renal toxicity of POL are also needed. Additionally, well-designed preclinical and clinical studies to validate the therapeutic effects of TCM need to be performed, thus hopefully providing a basis for the validation of the clinical benefits of POL.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, China.
| |
Collapse
|
4
|
Kietzmann T. Vitamin C: From nutrition to oxygen sensing and epigenetics. Redox Biol 2023; 63:102753. [PMID: 37263060 PMCID: PMC10245123 DOI: 10.1016/j.redox.2023.102753] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Vitamin C is unbeatable - at least when it comes to sales. Of all the vitamin preparations, those containing vitamin C sell best. This is surprising because vitamin C deficiency is extremely rare. Nevertheless, there is still controversy about whether the additional intake of vitamin C supplements is essential for our health. In this context, the possible additional benefit is in most cases merely reduced to the known effect as an antioxidant. However, new findings in recent years on the mechanisms of oxygen-sensing and epigenetic control underpin the multifaceted role of vitamin C in a biological context and have therefore renewed interest in it. In the present article, therefore, known facts are linked to these new key data. In addition, available clinical data on vitamin C use of cancer therapy are summarized.
Collapse
Affiliation(s)
- Thomas Kietzmann
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, P.O. Box 3000, 90014, Oulu, Finland.
| |
Collapse
|
5
|
Zaher A, Stephens LM, Miller AM, Hartwig SM, Stolwijk JM, Petronek MS, Zacharias ZR, Wadas TJ, Monga V, Cullen JJ, Furqan M, Houtman JCD, Varga SM, Spitz DR, Allen BG. Pharmacological ascorbate as a novel therapeutic strategy to enhance cancer immunotherapy. Front Immunol 2022; 13:989000. [PMID: 36072595 PMCID: PMC9444023 DOI: 10.3389/fimmu.2022.989000] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pharmacological ascorbate (i.e., intravenous infusions of vitamin C reaching ~ 20 mM in plasma) is under active investigation as an adjuvant to standard of care anti-cancer treatments due to its dual redox roles as an antioxidant in normal tissues and as a prooxidant in malignant tissues. Immune checkpoint inhibitors (ICIs) are highly promising therapies for many cancer patients but face several challenges including low response rates, primary or acquired resistance, and toxicity. Ascorbate modulates both innate and adaptive immune functions and plays a key role in maintaining the balance between pro and anti-inflammatory states. Furthermore, the success of pharmacological ascorbate as a radiosensitizer and a chemosensitizer in pre-clinical studies and early phase clinical trials suggests that it may also enhance the efficacy and expand the benefits of ICIs.
Collapse
Affiliation(s)
- Amira Zaher
- Cancer Biology Program, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Laura M. Stephens
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Ann M. Miller
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Stacey M. Hartwig
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Jeffrey M. Stolwijk
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Michael S. Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Zeb R. Zacharias
- Human Immunology Core & Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Thaddeus J. Wadas
- Department of Radiology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Varun Monga
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Joseph J. Cullen
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Muhammad Furqan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Jon C. D. Houtman
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Steven M. Varga
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Bryan G. Allen,
| |
Collapse
|
6
|
Two Distinct Faces of Vitamin C: AA vs. DHA. Antioxidants (Basel) 2021; 10:antiox10020215. [PMID: 33535710 PMCID: PMC7912923 DOI: 10.3390/antiox10020215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
Historically, vitamin C has been associated with many regulatory processes that involve specific signaling pathways. Among the most studied signaling pathways are those involved in the regulation of aging, differentiation, neurotransmission, proliferation, and cell death processes in cancer. This wide variety of regulatory effects is due to the fact that vitamin C has a dual mechanism of action. On the one hand, it regulates the expression of genes associated with proliferation (Ccnf and Ccnb1), differentiation (Sox-2 and Oct-4), and cell death (RIPK1 and Bcl-2). At the same time, vitamin C can act as a regulator of kinases, such as MAPK and p38, or by controlling the activation of the NF-kB pathway, generating chronic responses related to changes in gene expression or acute responses associated with the regulation of signal transduction processes. To date, data from the literature show a permanent increase in processes regulated by vitamin C. In this review, we critically examine how vitamin C regulates these different cellular programs in normal and tumor cells.
Collapse
|
7
|
Lykkesfeldt J. On the effect of vitamin C intake on human health: How to (mis)interprete the clinical evidence. Redox Biol 2020; 34:101532. [PMID: 32535545 PMCID: PMC7296342 DOI: 10.1016/j.redox.2020.101532] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
For decades, the potential beneficial effect of vitamin C on human health-beyond that of preventing scurvy-has been subject of much controversy. Hundreds of articles have appeared either in support of increased vitamin C intake through diet or supplements or rejecting the hypothesis that increased intake of vitamin C or supplementation may influence morbidity and mortality. The chemistry and pharmacology of vitamin C is complex and has unfortunately rarely been taken into account when designing clinical studies testing its effect on human health. However, ignoring its chemical lability, dose-dependent absorption and elimination kinetics, distribution via active transport, or complex dose-concentration-response relationships inevitably leads to poor study designs, inadequate inclusion and exclusion criteria and misinterpretation of results. The present review outlines the differences in vitamin C pharmacokinetics compared to normal low molecular weight drugs, focusses on potential pitfalls in study design and data interpretation, and re-examines major clinical studies of vitamin C in light of these.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Faculty of Health & Medical Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark.
| |
Collapse
|
8
|
Choi YK, Kang JI, Han S, Kim YR, Jo J, Kang YW, Choo DR, Hyun JW, Koh YS, Yoo ES, Kang HK. L-Ascorbic Acid Inhibits Breast Cancer Growth by Inducing IRE/JNK/CHOP-Related Endoplasmic Reticulum Stress-Mediated p62/SQSTM1 Accumulation in the Nucleus. Nutrients 2020; 12:nu12051351. [PMID: 32397306 PMCID: PMC7284633 DOI: 10.3390/nu12051351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/24/2020] [Accepted: 05/02/2020] [Indexed: 12/11/2022] Open
Abstract
Anticancer effects of L-ascorbic acid (Vitamin C, L-AA) have been reported in various types of cancers. L-AA intake reduces breast cancer recurrence and mortality; however, the role of L-AA in the treatment of breast cancer remains poorly understood. In this study, we investigated the effect and mechanism action of L-AA on breast cancer growth. L-AA inhibited the growth of breast cancer cells by inducing apoptotic cell death at the evaluated treatment concentrations without affecting normal cells. Moreover, L-AA induces autophagosome formation via regulation of mammalian target of rapamycin (mTOR), Beclin1, and autophagy-related genes (ATGs) and increased autophagic flux. Notably, we observed that L-AA increased p62/SQSTM1 (sequestosome 1) protein levels. Accumulation of p62 protein in cancer cells in response to stress has been reported, but its role in cancer regulation remains controversial. Here, we demonstrated that L-AA-induced p62 accumulation is related to L-AA-induced breast cancer growth inhibition. Furthermore, L-AA induced endoplasmic reticulum (ER) stress via the IRE–JNK–CHOP (inositol-requiring endonuclease–c-Jun N-terminal kinase–C/EBP homologous protein) signaling pathways, which increased the nuclear levels of p62/SQSTM1. These findings provide evidence that L-AA-induced ER stress could be crucial for p62 accumulation-dependent cell death, and L-AA can be useful in breast cancer treatment.
Collapse
Affiliation(s)
- Youn Kyung Choi
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Jung-Il Kang
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Sanghoon Han
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Young Ree Kim
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Jaemin Jo
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Yong Woo Kang
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Do Ryeon Choo
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
| | - Jin Won Hyun
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
- Jeju Research Center for Natural Medicine, Jeju National University; 102 Jejudaehakno, Jeju 63243, Korea
| | - Young Sang Koh
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
- Jeju Research Center for Natural Medicine, Jeju National University; 102 Jejudaehakno, Jeju 63243, Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
- Jeju Research Center for Natural Medicine, Jeju National University; 102 Jejudaehakno, Jeju 63243, Korea
| | - Hee-Kyoung Kang
- Department of Medicine, School of Medicine, Jeju National University 102 Jejudaehakno, Jeju 63243, Korea; (Y.K.C.); (J.-I.K.); (S.H.); (Y.R.K.); (J.J.); (Y.W.K.); (D.R.C.); (J.W.H.); (Y.S.K.); (E.-S.Y.)
- Jeju Research Center for Natural Medicine, Jeju National University; 102 Jejudaehakno, Jeju 63243, Korea
- Correspondence: ; Tel.:+82-10-6214-5464
| |
Collapse
|
9
|
Lin C, Dong J, Wei Z, Cheng KK, Li J, You S, Liu Y, Wang X, Chen Z. 1H NMR-Based Metabolic Profiles Delineate the Anticancer Effect of Vitamin C and Oxaliplatin on Hepatocellular Carcinoma Cells. J Proteome Res 2020; 19:781-793. [PMID: 31916767 DOI: 10.1021/acs.jproteome.9b00635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer death worldwide. Because of its high recurrence rate and heterogeneity, effective treatment for advanced stage of HCC is currently lacking. There are accumulating evidences showing the therapeutic potential of pharmacologic vitamin C (VC) on HCC. However, the metabolic basis underlying the anticancer property of VC remains to be elucidated. In this study, we used a high-resolution proton nuclear magnetic resonance-based metabolomics technique to assess the global metabolic changes in HCC cells following VC treatment. In addition, the HCC cells were also treated with oxaliplatin (OXA) to explore the potential synergistic effect induced by the combined VC and OXA treatment. The current metabolomics data suggested different mechanisms of OXA and VC in modulating cell growth and metabolism. In general, VC treatment led to inhibition of energy metabolism via NAD+ depletion and amino acid deprivation. On the other hand, OXA caused significant perturbation in phospholipid biosynthesis and phosphatidylcholine biosynthesis pathways. The current results highlighted glutathione metabolism, and pathways related to succinate and choline may play central roles in conferring the combined effect between OXA and VC. Taken together, this study provided metabolic evidence of VC and OXA in treating HCC and may contribute toward the potential application of combined VC and OXA as complementary HCC therapies.
Collapse
Affiliation(s)
- Caigui Lin
- Department of Electronic Science, Fujian Provincial Key Laboratory for Plasma and Magnetic Resonance , Xiamen University , Xiamen , Fujian 361005 , China
| | - Jiyang Dong
- Department of Electronic Science, Fujian Provincial Key Laboratory for Plasma and Magnetic Resonance , Xiamen University , Xiamen , Fujian 361005 , China
| | - Zhiliang Wei
- Department of Radiology , Johns Hopkins University , Baltimore , Maryland 21205 , United States
| | - Kian-Kai Cheng
- Innovation Centre in Agritechnology , Universiti Teknologi Malaysia , Muar , Johor 84600 , Malaysia
| | - Jie Li
- Department of Hepatobiliary Surgery , ZhongShan Hospital of Xiamen University , Xiamen , Fujian 361005 , China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma , ZhongShan Hospital of Xiamen University , Xiamen , Fujian 361005 , China
| | - Song You
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma , ZhongShan Hospital of Xiamen University , Xiamen , Fujian 361005 , China.,Graduate College of Fujian Medical University , Fuzhou , Fujian 350004 , China
| | - Yueyue Liu
- Department of Electronic Science, Fujian Provincial Key Laboratory for Plasma and Magnetic Resonance , Xiamen University , Xiamen , Fujian 361005 , China
| | - Xiaomin Wang
- Department of Hepatobiliary Surgery , ZhongShan Hospital of Xiamen University , Xiamen , Fujian 361005 , China.,Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma , ZhongShan Hospital of Xiamen University , Xiamen , Fujian 361005 , China
| | - Zhong Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory for Plasma and Magnetic Resonance , Xiamen University , Xiamen , Fujian 361005 , China
| |
Collapse
|
10
|
Nakanishi K, Goto K, Kondo K, Hiramoto K, Ooi K. Irinotecan-Induced Skin Dryness Is Ameliorated By Orally Administered High-Dose Vitamin C In Mice. J Exp Pharmacol 2019; 11:109-114. [PMID: 31632159 PMCID: PMC6790346 DOI: 10.2147/jep.s225565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Vitamin C plays a part in various roles in the human body. In this study, we examined the effect of oral administration of high-dose vitamin C on the skin dryness induced by irinotecan. Methods To establish the experimental model of irinotecan-induced skin dryness, the drug was intraperitoneally administered for four consecutive days. Simultaneously, oral administration of high-dose vitamin C (4 g/kg) was continued for 4 days. Results High-dose vitamin C administration ameliorated the skin dryness induced by irinotecan. The expression of caspase-3 and caspase-9, reactive oxygen species, and the number of TUNEL-positive cells increased in the skin of irinotecan-treated mice but were lowered by high-dose vitamin C administration. In contrast, fibroblasts and collagen type I decreased in the skin of the irinotecan-treated mice but was increased by high-dose vitamin C administration. Conclusion These results suggested that high-dose vitamin C administration can improve the skin dryness induced by irinotecan.
Collapse
Affiliation(s)
- Kentaro Nakanishi
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| | - Kenji Goto
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| | - Kanako Kondo
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan.,Department of Pharmacy, Kuwana City Medical Center, Kuwana, Mie 511-0061, Japan
| | - Keiichi Hiramoto
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| | - Kazuya Ooi
- Department of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka, Mie 513-8670, Japan
| |
Collapse
|
11
|
El Banna N, Hatem E, Heneman-Masurel A, Léger T, Baïlle D, Vernis L, Garcia C, Martineau S, Dupuy C, Vagner S, Camadro JM, Huang ME. Redox modifications of cysteine-containing proteins, cell cycle arrest and translation inhibition: Involvement in vitamin C-induced breast cancer cell death. Redox Biol 2019; 26:101290. [PMID: 31412312 PMCID: PMC6831881 DOI: 10.1016/j.redox.2019.101290] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Vitamin C (VitC) possesses pro-oxidant properties at high pharmacologic concentrations which favor repurposing VitC as an anti-cancer therapeutic agent. However, redox-based anticancer properties of VitC are yet partially understood. We examined the difference between the reduced and oxidized forms of VitC, ascorbic acid (AA) and dehydroascorbic acid (DHA), in terms of cytotoxicity and redox mechanisms toward breast cancer cells. Our data showed that AA displayed higher cytotoxicity towards triple-negative breast cancer (TNBC) cell lines in vitro than DHA. AA exhibited a similar cytotoxicity on non-TNBC cells, while only a minor detrimental effect on noncancerous cells. Using MDA-MB-231, a representative TNBC cell line, we observed that AA- and DHA-induced cytotoxicity were linked to cellular redox-state alterations. Hydrogen peroxide (H2O2) accumulation in the extracellular medium and in different intracellular compartments, and to a lesser degree, intracellular glutathione oxidation, played a key role in AA-induced cytotoxicity. In contrast, DHA affected glutathione oxidation and had less cytotoxicity. A "redoxome" approach revealed that AA treatment altered the redox state of key antioxidants and a number of cysteine-containing proteins including many nucleic acid binding proteins and proteins involved in RNA and DNA metabolisms and in energetic processes. We showed that cell cycle arrest and translation inhibition were associated with AA-induced cytotoxicity. Finally, bioinformatics analysis and biological experiments identified that peroxiredoxin 1 (PRDX1) expression levels correlated with AA differential cytotoxicity in breast cancer cells, suggesting a potential predictive value of PRDX1. This study provides insight into the redox-based mechanisms of VitC anticancer activity, indicating that pharmacologic doses of VitC and VitC-based rational drug combinations could be novel therapeutic opportunities for triple-negative breast cancer.
Collapse
Affiliation(s)
- Nadine El Banna
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Elie Hatem
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Amélie Heneman-Masurel
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Thibaut Léger
- Institut Jacques Monod, CNRS UMR 7592, Mass Spectrometry Laboratory, Université Paris Diderot, Paris, France
| | - Dorothée Baïlle
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Laurence Vernis
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Camille Garcia
- Institut Jacques Monod, CNRS UMR 7592, Mass Spectrometry Laboratory, Université Paris Diderot, Paris, France
| | - Sylvain Martineau
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Corinne Dupuy
- Institut Gustave Roussy, CNRS UMR 8200, Université Paris-Sud, Villejuif, France
| | - Stéphan Vagner
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Jean-Michel Camadro
- Institut Jacques Monod, CNRS UMR 7592, Mass Spectrometry Laboratory, Université Paris Diderot, Paris, France
| | - Meng-Er Huang
- Institut Curie, PSL Research University, CNRS UMR 3348, Université Paris-Sud, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
12
|
Abstract
Pancreatic ductal adenocarcinoma (PDA) has a dismal prognosis and is often discovered at an advanced stage with few therapeutic options. Current conventional regimens for PDA are associated with significant morbidity, decreased quality of life, and a considerable financial burden. As a result, some patients turn to integrative medicine therapies as an alternate option after a diagnosis of PDA. Intravenous pharmacologic ascorbic acid (PAA) is one such treatment. The use of PAA has been passionately debated for many years, but more recent rigorous scientific research has shown that there are significant blood concentration differences when ascorbic acid is given parenterally when compared to oral dosing. This pharmacologic difference appears to be critical for its role in oncology. Here, we report the use of PAA in a patient with poorly differentiated stage IV PDA as an exclusive chemotherapeutic regimen. The patient survived nearly 4 years after diagnosis, with PAA as his sole treatment, and he achieved objective regression of his disease. He died from sepsis and organ failure from a bowel perforation event. This case illustrates the possibility of PAA to effectively control tumor progression and serve as an adjunct to standard of care PDA chemotherapy regimens. Our patient's experience with PAA should be taken into consideration, along with previous research in cell, animal, and clinical experiments to design future treatment trials.
Collapse
|
13
|
Targeting peroxiredoxin 1 impairs growth of breast cancer cells and potently sensitises these cells to prooxidant agents. Br J Cancer 2018; 119:873-884. [PMID: 30287919 PMCID: PMC6189216 DOI: 10.1038/s41416-018-0263-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/10/2018] [Accepted: 08/23/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Our previous work has shown peroxiredoxin-1 (PRDX1), one of major antioxidant enzymes, to be a biomarker in human breast cancer. Hereby, we further investigate the role of PRDX1, compared to its close homolog PRDX2, in mammary malignant cells. METHODS CRISPR/Cas9- or RNAi-based methods were used for genetic targeting PRDX1/2. Cell growth was assessed by crystal violet, EdU incorporation or colony formation assays. In vivo growth was assessed by a xenotransplantation model. Adenanthin was used to inhibit the thioredoxin-dependent antioxidant defense system. The prooxidant agents used were hydrogen peroxide, glucose oxidase and sodium L-ascorbate. A PY1 probe or HyPer-3 biosensor were used to detect hydrogen peroxide content in samples. RESULTS PRDX1 downregulation significantly impaired the growth rate of MCF-7 and ZR-75-1 breast cancer cells. Likewise, xenotransplanted PRDX1-deficient MCF-7 cells presented a retarded tumour growth. Furthermore, genetic targeting of PRDX1 or adenanthin, but not PRDX2, potently sensitised all six cancer cell lines studied, but not the non-cancerous cells, to glucose oxidase and ascorbate. CONCLUSIONS Our study pinpoints the dominant role for PRDX1 in management of exogeneous oxidative stress by breast cancer cells and substantiates further exploration of PRDX1 as a target in this disease, especially when combined with prooxidant agents.
Collapse
|
14
|
Shin H, Nam A, Song KH, Lee K, Rebhun RB, Seo KW. Anticancer effects of high-dose ascorbate on canine melanoma cell lines. Vet Comp Oncol 2018; 16:616-621. [DOI: 10.1111/vco.12429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Hyeri Shin
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Chungnam National University; Daejeon South Korea
| | - Aryung Nam
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Seoul National University; Seoul South Korea
| | - Kun-Ho Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Chungnam National University; Daejeon South Korea
| | - Kupil Lee
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Chungnam National University; Daejeon South Korea
| | - Robert B. Rebhun
- Comparative Oncology Laboratory and Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine; University of California-Davis; Davis California
| | - Kyoung-Won Seo
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Chungnam National University; Daejeon South Korea
| |
Collapse
|
15
|
Mohammed BM, Sanford KW, Fisher BJ, Martin EJ, Contaifer Jr D, Warncke UO, Wijesinghe DS, Chalfant CE, Brophy DF, Fowler III AA, Natarajan R. Impact of high dose vitamin C on platelet function. World J Crit Care Med 2017; 6:37-47. [PMID: 28224106 PMCID: PMC5295168 DOI: 10.5492/wjccm.v6.i1.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/15/2016] [Accepted: 11/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To examine the effect of high doses of vitamin C (VitC) on ex vivo human platelets (PLTs).
METHODS Platelet concentrates collected for therapeutic or prophylactic transfusions were exposed to: (1) normal saline (control); (2) 0.3 mmol/L VitC (Lo VitC); or (3) 3 mmol/L VitC (Hi VitC, final concentrations) and stored appropriately. The VitC additive was preservative-free buffered ascorbic acid in water, pH 5.5 to 7.0, adjusted with sodium bicarbonate and sodium hydroxide. The doses of VitC used here correspond to plasma VitC levels reported in recently completed clinical trials. Prior to supplementation, a baseline sample was collected for analysis. PLTs were sampled again on days 2, 5 and 8 and assayed for changes in PLT function by: Thromboelastography (TEG), for changes in viscoelastic properties; aggregometry, for PLT aggregation and adenosine triphosphate (ATP) secretion in response to collagen or adenosine diphosphate (ADP); and flow cytometry, for changes in expression of CD-31, CD41a, CD62p and CD63. In addition, PLT intracellular VitC content was measured using a fluorimetric assay for ascorbic acid and PLT poor plasma was used for plasma coagulation tests [prothrombin time (PT), partial thrombplastin time (PTT), functional fibrinogen] and Lipidomics analysis (UPLC ESI-MS/MS).
RESULTS VitC supplementation significantly increased PLTs intracellular ascorbic acid levels from 1.2 mmol/L at baseline to 3.2 mmol/L (Lo VitC) and 15.7 mmol/L (Hi VitC, P < 0.05). VitC supplementation did not significantly change PT and PTT values, or functional fibrinogen levels over the 8 d exposure period (P > 0.05). PLT function assayed by TEG, aggregometry and flow cytometry was not significantly altered by Lo or Hi VitC for up to 5 d. However, PLTs exposed to 3 mmol/L VitC for 8 d demonstrated significantly increased R and K times by TEG and a decrease in the α-angle (P < 0.05). There was also a fall of 20 mm in maximum amplitude associated with the Hi VitC compared to both baseline and day 8 saline controls. Platelet aggregation studies, showed uniform declines in collagen and ADP-induced platelet aggregations over the 8-d study period in all three groups (P > 0.05). Collagen and ADP-induced ATP secretion was also not different between the three groups (P > 0.05). Finally, VitC at the higher dose (3 mmol/L) also induced the release of several eicosanoids including thromboxane B2 and prostaglandin E2, as well as products of arachidonic acid metabolism via the lipoxygenases pathway such as 11-/12-/15-hydroxyicosatetraenoic acid (P < 0.05).
CONCLUSION Alterations in PLT function by exposure to 3 mmol/L VitC for 8 d suggest that caution should be exerted with prolonged use of intravenous high dose VitC.
Collapse
|
16
|
Baek MW, Cho HS, Kim SH, Kim WJ, Jung JY. Ascorbic Acid Induces Necrosis in Human Laryngeal Squamous Cell Carcinoma via ROS, PKC, and Calcium Signaling. J Cell Physiol 2016; 232:417-425. [DOI: 10.1002/jcp.25438] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 05/19/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Min-Woo Baek
- Department of Oral Physiology, Dental Science Research Institute, Medical Research Center for Biomineralization Disorders; Chonnam National University School of Dentistry; Gwangju Republic of Korea
| | - Heui-Seung Cho
- Department of Oral Physiology, Dental Science Research Institute, Medical Research Center for Biomineralization Disorders; Chonnam National University School of Dentistry; Gwangju Republic of Korea
| | - Sun-Hun Kim
- Department of Oral Anatomy, Dental Science Research Institute, Medical Research Center for Biomineralization Disorders; Chonnam National University School of Dentistry; Gwangju Republic of Korea
| | - Won-Jae Kim
- Department of Oral Physiology, Dental Science Research Institute, Medical Research Center for Biomineralization Disorders; Chonnam National University School of Dentistry; Gwangju Republic of Korea
| | - Ji-Yeon Jung
- Department of Oral Physiology, Dental Science Research Institute, Medical Research Center for Biomineralization Disorders; Chonnam National University School of Dentistry; Gwangju Republic of Korea
| |
Collapse
|
17
|
Kumar A, Chelvam V, Sakkarapalayam M, Li G, Sanchez-Cruz P, Piñero NS, Low PS, Alegria AE. Synthesis and Evaluation of Folate-Conjugated Phenanthraquinones for Tumor-Targeted Oxidative Chemotherapy. ACTA ACUST UNITED AC 2016; 6:1-17. [PMID: 27066312 PMCID: PMC4825697 DOI: 10.4236/ojmc.2016.61001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Almost all cells are easily killed by exposure to potent oxidants. Indeed, major pathogen defense mechanisms in both animal and plant kingdoms involve production of an oxidative burst, where host defense cells show an invading pathogen with reactive oxygen species (ROS). Although cancer cells can be similarly killed by ROS, development of oxidant-producing chemotherapies has been limited by their inherent nonspecificity and potential toxicity to healthy cells. In this paper, we describe the targeting of an ROS-generating molecule selectively to tumor cells using folate as the tumor-targeting ligand. For this purpose, we exploit the ability of 9,10-phenanthraquinone (PHQ) to enhance the continuous generation of H2O2 in the presence of ascorbic acid to establish a constitutive source of ROS within the tumor mass. We report here that incubation of folate receptor-expressing KB cells in culture with folate-PHQ plus ascorbate results in the death of the cancer cells with an IC50 of ~10 nM (folate-PHQ). We also demonstrate that a cleavable spacer linking folate to PHQ is significantly inferior to a noncleavable spacer, in contrast to most other folate-targeted therapeutic agents. Unfortunately, no evidence for folate-PHQ mediated tumor regression in murine tumor models is obtained, suggesting that unanticipated impediments to generation of cytotoxic quantities of ROS in vivo are encountered. Possible mechanisms and potential solutions to these unanticipated results are offered.
Collapse
Affiliation(s)
- Ajay Kumar
- International Center for Trans-disciplinary Research, School of Environmental Affairs, Universidad Metropolitana, San Juan, Puerto Rico ; Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Venkatesh Chelvam
- Department of Chemistry, Purdue University, West Lafayette, Indiana ; Department of Chemistry, Centre for Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Simrol Campus, Madhya Pradesh, India
| | | | - Guo Li
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Pedro Sanchez-Cruz
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Natasha S Piñero
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Antonio E Alegria
- Department of Chemistry, University of Puerto Rico, Humacao, Puerto Rico
| |
Collapse
|
18
|
Mn Porphyrin-Based Redox-Active Therapeutics. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2016. [DOI: 10.1007/978-3-319-30705-3_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Leelahavanichkul A, Somparn P, Bootprapan T, Tu H, Tangtanatakul P, Nuengjumnong R, Worasilchai N, Tiranathanagul K, Eiam-ong S, Levine M, Chinampon A, Srisawat N. High-dose ascorbate with low-dose amphotericin B attenuates severity of disease in a model of the reappearance of candidemia during sepsis in the mouse. Am J Physiol Regul Integr Comp Physiol 2015; 309:R223-34. [PMID: 25994956 DOI: 10.1152/ajpregu.00238.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amphotericin B (Ampho B) isa fungicidal drug that causes cell wall injury. Pharmacological ascorbate induces the extracellular prooxidants, which might enter the Ampho B-induced cell wall porosity and act synergistically.W e tested low-dose Ampho B with a short course of pharmacological ascorbate using a mouse model of sepsis preconditioned with an injection of Candida albicans 6 h prior to cecal ligation and puncture (CLP). In this model, candidemia reappeared as early as 6 h after CLP with a predictably high mortality rate. This characteristic mimics sepsis in the phase of immunosuppression inpatients. Using the model, at 12- and 18-h post-CLP, we administered isotonic (pH neutralized) pharmacological ascorbate intravenously with low-dose Ampho B or sodium deoxycholate, vehicle-controlled, administered IP. The survival rate of low-dose Ampho B plus ascorbate was 53%, compared with < 11% for low-dose Ampho B or high-dose Ampho B alone. In addition, a beneficial effect was demonstrated in terms of kidney damage,liver injury, spleen histopathology, and serum markers at 24 h after CLP. Kidney injury was less severe in low-dose Ampho B plus ascorbate combination therapy due to less severe sepsis. Moreover, ascorbate enhanced the effectiveness of phagocytosis against C. albicans in human phagocytic cells. Taken together, the data indicate that the new mouse model simulates sepsis-induced immunosuppression and that the combination of pharmacological ascorbate with an antifungal drug is a potentially effective treatment that may reduce nephrotoxicity, and perhaps also increase fungicidal activity in patients with systemic candidiasis caused by Candida albicans.
Collapse
|
20
|
Serrano OK, Parrow NL, Violet PC, Yang J, Zornjak J, Basseville A, Levine M. Antitumor effect of pharmacologic ascorbate in the B16 murine melanoma model. Free Radic Biol Med 2015; 87:193-203. [PMID: 26119785 DOI: 10.1016/j.freeradbiomed.2015.06.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 11/29/2022]
Abstract
Because 5-year survival rates for patients with metastatic melanoma remain below 25%, there is continued need for new therapeutic approaches. For some tumors, pharmacologic ascorbate treatment may have a beneficial antitumor effect and may work synergistically with standard chemotherapeutics. To investigate this possibility in melanoma, we examined the effect of pharmacologic ascorbate on B16-F10 cells. Murine models were employed to compare tumor size following treatment with ascorbate, and the chemotherapeutic agents dacarbazine or valproic acid, alone or in combination with ascorbate. Results indicated that nearly all melanoma cell lines were susceptible to ascorbate-mediated cytotoxicity. Compared to saline controls, pharmacologic ascorbate decreased tumor size in both C57BL/6 (P < 0.0001) and NOD-scid tumor bearing mice (P < 0.0001). Pharmacologic ascorbate was superior or equivalent to dacarbazine as an antitumor agent. Synergy was not apparent when ascorbate was combined with either dacarbazine or valproic acid; the latter combination may have additional toxicities. Pharmacologic ascorbate induced DNA damage in melanoma cells, as evidenced by increased phosphorylation of the histone variant, H2A.X. Differences were not evident in tumor samples from C57BL/6 mice treated with pharmacologic ascorbate compared to tumors from saline-treated controls. Together, these results suggest that pharmacologic ascorbate has a cytotoxic effect against melanoma that is largely independent of lymphocytic immune functions and that continued investigation of pharmacologic ascorbate in cancer treatment is warranted.
Collapse
Affiliation(s)
- Oscar K Serrano
- Department of Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, USA
| | - Nermi L Parrow
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline Yang
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Zornjak
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Agnes Basseville
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
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. [PMID: 26350063 PMCID: PMC4563566 DOI: 10.1038/srep13896] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023] Open
Abstract
Intravenous administration of high-dose vitamin C has recently attracted attention as a cancer therapy. High-dose vitamin C induces pro-oxidant effects and selectively kills cancer cells. However, the anticancer mechanisms of vitamin C are not fully understood. Here, we analyzed metabolic changes induced by vitamin C in MCF7 human breast adenocarcinoma and HT29 human colon cancer cells using capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS). The metabolomic profiles of both cell lines were dramatically altered after exposure to cytotoxic concentrations of vitamin C. Levels of upstream metabolites in the glycolysis pathway and tricarboxylic acid (TCA) cycle were increased in both cell lines following treatment with vitamin C, while adenosine triphosphate (ATP) levels and adenylate energy charges were decreased concentration-dependently. Treatment with N-acetyl cysteine (NAC) and reduced glutathione (GSH) significantly inhibited vitamin C-induced cytotoxicity in MCF7 cells. NAC also suppressed vitamin C-dependent metabolic changes, and NAD treatment prevented vitamin C-induced cell death. Collectively, our data suggests that vitamin C inhibited energy metabolism through NAD depletion, thereby inducing cancer cell death.
Collapse
Affiliation(s)
- Megumi Uetaki
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan.,Systems Biology Program, Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan
| | - Sho Tabata
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan.,Systems Biology Program, Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan
| | - Fumie Nakasuka
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan.,Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan.,Systems Biology Program, Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan.,Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan.,Systems Biology Program, Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan.,Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa 252-0882, Japan
| |
Collapse
|
22
|
Mastrangelo D, Massai L, Lo Coco F, Noguera NI, Borgia L, Fioritoni G, Berardi A, Iacone A, Muscettola M, Pelosi E, Castelli G, Testa U, Di Pisa F, Grasso G. Cytotoxic effects of high concentrations of sodium ascorbate on human myeloid cell lines. Ann Hematol 2015; 94:1807-16. [PMID: 26264692 DOI: 10.1007/s00277-015-2464-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 07/07/2015] [Indexed: 12/16/2022]
Abstract
The effect of high doses of intravenous (sodium) ascorbate (ASC) in the treatment of cancer has been controversial although there is growing evidence that ASC in high (pharmacologic) concentrations induces dose-dependent pro-apoptotic death of tumor cells, in vitro. Very few data are available on the role of ASC in the treatment of acute myeloid leukemia (AML). Ascorbate behaves as an antioxidant at low (physiologic), and as pro-oxidant at pharmacologic, concentrations, and this may account for the differences reported in different experimental settings, when human myeloid cell lines, such as HL60, were treated with ASC. Considering the myeloid origin of HL60 cells, and previous literature reports showing that some cell lines belonging to the myeloid lineage could be sensitive to the pro-apoptotic effects of high concentrations of ASC, we investigated in more details the effects of high doses (0.5 to 7 mM) of ASC in vitro, on a variety of human myeloid cell lines including the following: HL60, U937, NB4, NB4-R4 (retinoic acid [RA]-resistant), NB4/AsR (ATO-resistant) acute promyelocytic leukemia (APL)-derived cell lines, and K562 as well as on normal CD34+ progenitors derived from human cord blood. Our results indicate that all analyzed cell lines including all-trans retinoic acid (ATRA)- and arsenic trioxide (ATO)-resistant ones are highly sensitive to the cytotoxic, pro-oxidant effects of high doses of ASC, with an average 50 % lethal concentration (LC50) of 3 mM, depending on cell type, ASC concentration, and time of exposure. Conversely, high doses of ASC neither did exert significant cytotoxic effects nor impaired the differentiation potential in cord blood (CB) CD34+ normal cells. Since plasma ASC concentrations within the millimolar (mM) range can be easily and safely reached by intravenous administration, we conclude that phase I/II clinical trials using high doses of ASC should be designed for patients with advanced/refractory AML and APL.
Collapse
Affiliation(s)
- Domenico Mastrangelo
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100, Siena, Italy.
| | - Lauretta Massai
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100, Siena, Italy
| | - Francesco Lo Coco
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,Santa Lucia Foundation, Via del Fosso di Fiorano, 00143, Rome, Italy
| | - Nélida Inés Noguera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,Santa Lucia Foundation, Via del Fosso di Fiorano, 00143, Rome, Italy
| | - Loredana Borgia
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,Santa Lucia Foundation, Via del Fosso di Fiorano, 00143, Rome, Italy
| | - Giuseppe Fioritoni
- Pescara Cell Factory Foundation Onlus, Corso Vittorio Emanuele II n. 346, 65122, Pescara, Italy
| | - Anna Berardi
- Pescara Cell Factory Foundation Onlus, Corso Vittorio Emanuele II n. 346, 65122, Pescara, Italy
| | - Antonio Iacone
- Pescara Cell Factory Foundation Onlus, Corso Vittorio Emanuele II n. 346, 65122, Pescara, Italy
| | - Michela Muscettola
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100, Siena, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Ugo Testa
- Pescara Cell Factory Foundation Onlus, Corso Vittorio Emanuele II n. 346, 65122, Pescara, Italy
| | - Francesco Di Pisa
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100, Siena, Italy
| | - Giovanni Grasso
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Via A. Moro 2, 53100, Siena, Italy
| |
Collapse
|
23
|
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. [PMID: 25848948 PMCID: PMC4388666 DOI: 10.1371/journal.pone.0120228] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 01/27/2015] [Indexed: 12/17/2022] Open
Abstract
Background Biological and some clinical evidence suggest that high-dose intravenous vitamin C (IVC) could increase the effectiveness of cancer chemotherapy. IVC is widely used by integrative and complementary cancer therapists, but rigorous data are lacking as to its safety and which cancers and chemotherapy regimens would be the most promising to investigate in detail. Methods and Findings We carried out a phase I-II safety, tolerability, pharmacokinetic and efficacy trial of IVC combined with chemotherapy in patients whose treating oncologist judged that standard-of-care or off-label chemotherapy offered less than a 33% likelihood of a meaningful response. We documented adverse events and toxicity associated with IVC infusions, determined pre- and post-chemotherapy vitamin C and oxalic acid pharmacokinetic profiles, and monitored objective clinical responses, mood and quality of life. Fourteen patients were enrolled. IVC was safe and generally well tolerated, although some patients experienced transient adverse events during or after IVC infusions. The pre- and post-chemotherapy pharmacokinetic profiles suggested that tissue uptake of vitamin C increases after chemotherapy, with no increase in urinary oxalic acid excretion. Three patients with different types of cancer experienced unexpected transient stable disease, increased energy and functional improvement. Conclusions Despite IVC’s biological and clinical plausibility, career cancer investigators currently ignore it while integrative cancer therapists use it widely but without reporting the kind of clinical data that is normally gathered in cancer drug development. The present study neither proves nor disproves IVC’s value in cancer therapy, but it provides practical information, and indicates a feasible way to evaluate this plausible but unproven therapy in an academic environment that is currently uninterested in it. If carried out in sufficient numbers, simple studies like this one could identify specific clusters of cancer type, chemotherapy regimen and IVC in which exceptional responses occur frequently enough to justify appropriately focused clinical trials. Trial Registration ClinicalTrials.gov NCT01050621
Collapse
|
24
|
|
25
|
Batinic-Haberle I, Tovmasyan A, Roberts ERH, Vujaskovic Z, Leong KW, Spasojevic I. SOD therapeutics: latest insights into their structure-activity relationships and impact on the cellular redox-based signaling pathways. Antioxid Redox Signal 2014; 20:2372-415. [PMID: 23875805 PMCID: PMC4005498 DOI: 10.1089/ars.2012.5147] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 06/30/2013] [Accepted: 07/22/2013] [Indexed: 01/23/2023]
Abstract
SIGNIFICANCE Superoxide dismutase (SOD) enzymes are indispensable and ubiquitous antioxidant defenses maintaining the steady-state levels of O2·(-); no wonder, thus, that their mimics are remarkably efficacious in essentially any animal model of oxidative stress injuries thus far explored. RECENT ADVANCES Structure-activity relationship (half-wave reduction potential [E1/2] versus log kcat), originally reported for Mn porphyrins (MnPs), is valid for any other class of SOD mimics, as it is dominated by the superoxide reduction and oxidation potential. The biocompatible E1/2 of ∼+300 mV versus normal hydrogen electrode (NHE) allows powerful SOD mimics as mild oxidants and antioxidants (alike O2·(-)) to readily traffic electrons among reactive species and signaling proteins, serving as fine mediators of redox-based signaling pathways. Based on similar thermodynamics, both SOD enzymes and their mimics undergo similar reactions, however, due to vastly different sterics, with different rate constants. CRITICAL ISSUES Although log kcat(O2·(-)) is a good measure of therapeutic potential of SOD mimics, discussions of their in vivo mechanisms of actions remain mostly of speculative character. Most recently, the therapeutic and mechanistic relevance of oxidation of ascorbate and glutathionylation and oxidation of protein thiols by MnP-based SOD mimics and subsequent inactivation of nuclear factor κB has been substantiated in rescuing normal and killing cancer cells. Interaction of MnPs with thiols seems to be, at least in part, involved in up-regulation of endogenous antioxidative defenses, leading to the healing of diseased cells. FUTURE DIRECTIONS Mechanistic explorations of single and combined therapeutic strategies, along with studies of bioavailability and translational aspects, will comprise future work in optimizing redox-active drugs.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Emily R. H. Roberts
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- King Abdulaziz University, Jeddah, Saudi Arabia Kingdom
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical School, Durham, North Carolina
| |
Collapse
|
26
|
Bocci V, Zanardi I, Travagli V. A Rational Approach for Improving the Ascorbate Antineoplastic Activity. Cancer Invest 2014; 32:81-4. [DOI: 10.3109/07357907.2013.877477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Kim E, Leverage WT, Liu Y, White IM, Bentley WE, Payne GF. Redox-capacitor to connect electrochemistry to redox-biology. Analyst 2014; 139:32-43. [DOI: 10.1039/c3an01632c] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Kontek R, Kontek B, Grzegorczyk K. Vitamin C modulates DNA damage induced by hydrogen peroxide in human colorectal adenocarcinoma cell lines (HT29) estimated by comet assay in vitro. Arch Med Sci 2013; 9:1006-12. [PMID: 24482643 PMCID: PMC3902721 DOI: 10.5114/aoms.2013.39791] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 06/27/2012] [Accepted: 08/20/2012] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Cancer cells, compared to normal cells, are under increased oxidative stress associated with oncogenic transformation, alterations in metabolic activity, and increased generation of reactive oxygen species. MATERIAL AND METHODS We investigated the ability of vitamin C to reduce the damage induced by hydrogen peroxide, in human colorectal adenocarcinoma cells in vitro by the comet assay. Additionally, we measured the kinetics and efficacy of the repair of DNA damage after incubation with vitamin C in the presence of H2O2. RESULTS The obtained results showed that 1 h pre-incubation with vitamin C and exposure to H2O2 for the last 10 min of incubation caused a statistically significant (p < 0.05) increase in DNA migration in comet tails in all experimental series. For the 10 µM, 25 µM, 50 µM, 100 µM vitamin C concentrations the levels of DNA damage were as follows: 18.6%, 21.1%, 25.3% and 27.2%, respectively, as compared to the untreated cells (3.26%). However, in comparison with H2O2 alone (29.1%), we observed a statistically significant (p < 0.05) decrease of the genotoxic effect in HT29 cells induced by H2O2 for the two lowest of concentrations of vitamin C: 10 µM and 25 µM. The HT29 cells were able to achieve effective repair of the damaged DNA within 60 and 120 min after incubation with the tested compounds. All the values obtained in the test were statistically significant (p < 0.05). CONCLUSIONS Vitamin C caused a weaker DNA damaging effect of hydrogen peroxide and positively influences the level of oxidative DNA damage in HT29 cells (decrease ∼ 30%). We noted that DNA damage was effectively repaired during 120 min postincubation in the tested cells and that oxidative damage was the major type of damage.
Collapse
Affiliation(s)
- Renata Kontek
- Laboratory of Cytogenetics, Department of General Genetics, Molecular Biology and Plant Biotechnology, University of Lodz, Poland
| | - Bogdan Kontek
- Department of General Biochemistry, University of Lodz, Poland
| | - Krzysztof Grzegorczyk
- Department of Endoscopy and One Day Gastroenterology, Wl. Bieganski Memorial Regional Specialist Hospital, Lodz, Poland
| |
Collapse
|
29
|
Vuyyuri SB, Rinkinen J, Worden E, Shim H, Lee S, Davis KR. Ascorbic acid and a cytostatic inhibitor of glycolysis synergistically induce apoptosis in non-small cell lung cancer cells. PLoS One 2013; 8:e67081. [PMID: 23776707 PMCID: PMC3679078 DOI: 10.1371/journal.pone.0067081] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 05/15/2013] [Indexed: 12/28/2022] Open
Abstract
Ascorbic acid (AA) exhibits significant anticancer activity at pharmacologic doses achievable by parenteral administration that have minimal effects on normal cells. Thus, AA has potential uses as a chemotherapeutic agent alone or in combination with other therapeutics that specifically target cancer-cell metabolism. We compared the effects of AA and combinations of AA with the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3-PO) on the viability of three non-small cell lung cancer (NSCLC) cell lines to the effects on an immortalized lung epithelial cell line. AA concentrations of 0.5 to 5 mM caused a complete loss of viability in all NSCLC lines compared to a <10% loss of viability in the lung epithelial cell line. Combinations of AA and 3-PO synergistically enhanced cell death in all NSCLC cell lines at concentrations well below the IC50 concentrations for each compound alone. A synergistic interaction was not observed in combination treatments of lung epithelial cells and combination treatments that caused a complete loss of viability in NSCLC cells had modest effects on normal lung cell viability and reactive oxygen species (ROS) levels. Combination treatments induced dramatically higher ROS levels compared to treatment with AA and 3-PO alone in NSCLC cells and combination-induced cell death was inhibited by addition of catalase to the medium. Analyses of DNA fragmentation, poly (ADP-ribose) polymerase cleavage, annexin V-binding, and caspase activity demonstrated that AA-induced cell death is caused via the activation of apoptosis and that the combination treatments caused a synergistic induction of apoptosis. These results demonstrate the effectiveness of AA against NSCLC cells and that combinations of AA with 3-PO synergistically induce apoptosis via a ROS-dependent mechanism. These results support further evaluation of pharmacologic concentrations of AA as an adjuvant treatment for NSCLC and that combination of AA with glycolysis inhibitors may be a promising therapy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Saleha B. Vuyyuri
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
| | - Jacob Rinkinen
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
| | - Erin Worden
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
| | - Hyekyung Shim
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Sukchan Lee
- Department of Genetic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Keith R. Davis
- Owensboro Cancer Research Program, Owensboro, Kentucky, United States of America
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
- * E-mail:
| |
Collapse
|
30
|
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; 72:139-46. [PMID: 23670640 PMCID: PMC3691494 DOI: 10.1007/s00280-013-2179-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/21/2013] [Indexed: 12/13/2022]
Abstract
PURPOSE This phase I clinical trial evaluated the safety, tolerability, and pharmacokinetics of high-dose intravenous (i.v.) ascorbic acid as a monotherapy in patients with advanced solid tumors refractory to standard therapy. METHODS Five cohorts of three patients received i.v. ascorbic acid administered at 1 g/min for 4 consecutive days/week for 4 weeks, starting at 30 g/m² in the first cohort. For subsequent cohorts, dose was increased by 20 g/m² until a maximum tolerated dose was found. RESULTS Ascorbic acid was eliminated by simple first-order kinetics. Half-life and clearance values were similar for all patients of all cohorts (2.0 ± 0.6 h, 21 ± 5 dL/h m², respectively). C(max) and AUC values increased proportionately with dose between 0 and 70 g/m², but appeared to reach maximal values at 70 g/m² (49 mM and 220 h mM, respectively). Doses of 70, 90, and 110 g/m² maintained levels at or above 10-20 mM for 5-6 h. All doses were well tolerated. No patient demonstrated an objective antitumor response. CONCLUSIONS Ascorbic acid administered i.v. at 1 g/min for 4 consecutive days/week for 4 weeks produced up to 49 mM ascorbic acid in patient's blood and was well tolerated. The recommended dose for future studies is 70-80 g/m².
Collapse
|
31
|
Simultaneous scurvy and Wernicke's encephalopathy in a patient with an ascorbate-responsive dyskinesia. PSYCHOSOMATICS 2012; 54:181-6. [PMID: 23274004 DOI: 10.1016/j.psym.2012.07.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 07/23/2012] [Accepted: 07/23/2012] [Indexed: 11/23/2022]
|
32
|
Tovmasyan A, Sheng H, Weitner T, Arulpragasam A, Lu M, Warner DS, Vujaskovic Z, Spasojevic I, Batinic-Haberle I. Design, mechanism of action, bioavailability and therapeutic effects of mn porphyrin-based redox modulators. Med Princ Pract 2012; 22:103-30. [PMID: 23075911 PMCID: PMC3640855 DOI: 10.1159/000341715] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 07/01/2012] [Indexed: 12/18/2022] Open
Abstract
Based on aqueous redox chemistry and simple in vivo models of oxidative stress, Escherichia coli and Saccharomyces cerevisiae, the cationic Mn(III) N-substituted pyridylporphyrins (MnPs) have been identified as the most potent cellular redox modulators within the porphyrin class of drugs; their efficacy in animal models of diseases that have oxidative stress in common is based on their high ability to catalytically remove superoxide, peroxynitrite, carbonate anion radical, hypochlorite, nitric oxide, lipid peroxyl and alkoxyl radicals, thus suppressing the primary oxidative event. While doing so MnPs could couple with cellular reductants and redox-active proteins. Reactive species are widely accepted as regulators of cellular transcriptional activity: minute, nanomolar levels are essential for normal cell function, while submicromolar or micromolar levels impose oxidative stress, which is evidenced in increased inflammatory and immune responses. By removing reactive species, MnPs affect redox-based cellular transcriptional activity and consequently secondary oxidative stress, and in turn inflammatory processes. The equal ability to reduce and oxidize superoxide during the dismutation process and recently accumulated results suggest that pro-oxidative actions of MnPs may also contribute to their therapeutic effects. All our data identify the superoxide dismutase-like activity, estimated by log k(cat)O2-*), as a good measure for the therapeutic efficacy of MnPs. Their accumulation in mitochondria and their ability to cross the blood-brain barrier contribute to their remarkable efficacy. We summarize herein the therapeutic effects of MnPs in cancer, central nervous system injuries, diabetes, their radioprotective action and potential for imaging. Few of the most potent modulators of cellular redox-based pathways, MnTE2-PyP5+, MnTDE-2-ImP5+, MnTnHex-2-PyP5+ and MnTnBuOE-2-PyP5+, are under preclinical and clinical development.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Tin Weitner
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Amanda Arulpragasam
- Department of Duke University Neuroscience Undergraduate
Program, Duke University Medical Center, Durham, N.C., USA
| | - Miaomiao Lu
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
- Department of Department of Anesthesiology, Second Affiliated
Hospital, Zhengzhou University, Zhengzhou, China
| | - David S. Warner
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham,
N.C., USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| |
Collapse
|
33
|
Tarrago-Trani MT, Phillips KM, Cotty M. Matrix-specific method validation for quantitative analysis of vitamin C in diverse foods. J Food Compost Anal 2012. [DOI: 10.1016/j.jfca.2012.03.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
34
|
Pathi SS, Lei P, Sreevalsan S, Chadalapaka G, Jutooru I, Safe S. Pharmacologic doses of ascorbic acid repress specificity protein (Sp) transcription factors and Sp-regulated genes in colon cancer cells. Nutr Cancer 2011; 63:1133-1142. [PMID: 21919647 PMCID: PMC3359146 DOI: 10.1080/01635581.2011.605984] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ascorbic acid (vitamin C) inhibits cancer cell growth, and there is a controversy regarding the cancer chemoprotective effects of pharmacologic doses of this compound that exhibits prooxidant activity. We hypothesized that the anticancer activity of pharmacologic doses of ascorbic acid (<5 mM) is due, in part, to reactive oxygen species-dependent downregulation of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and Sp-regulated genes. In this study, ascorbic acid (1-3 mM) decreased RKO and SW480 colon cancer cell proliferation and induced apoptosis and necrosis, and this was accompanied by downregulation of Sp1, Sp3, and Sp4 proteins. In addition, ascorbic acid decreased expression of several Sp-regulated genes that are involved in cancer cell proliferation [hepatocyte growth factor receptor (c-Met), epidermal growth factor receptor and cyclin D1], survival (survivin and bcl-2), and angiogenesis [vascular endothelial growth factor (VEGF) and its receptors (VEGFR1 and VEGFR2)]. Other prooxidants such as hydrogen peroxide exhibited similar activities in colon cancer cells, and cotreatment with glutathione inhibited these responses. This study demonstrates for the first time that the anticancer activities of ascorbic acid are due, in part, to ROS-dependent repression of Sp transcription factors.
Collapse
Affiliation(s)
- Satya S. Pathi
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Ping Lei
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030
| | - Sandeep Sreevalsan
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Gayathri Chadalapaka
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Indira Jutooru
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030
| |
Collapse
|
35
|
Cyr AR, Domann FE. The redox basis of epigenetic modifications: from mechanisms to functional consequences. Antioxid Redox Signal 2011; 15:551-89. [PMID: 20919933 PMCID: PMC3118659 DOI: 10.1089/ars.2010.3492] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epigenetic modifications represent mechanisms by which cells may effectively translate multiple signaling inputs into phenotypic outputs. Recent research is revealing that redox metabolism is an increasingly important determinant of epigenetic control that may have significant ramifications in both human health and disease. Numerous characterized epigenetic marks, including histone methylation, acetylation, and ADP-ribosylation, as well as DNA methylation, have direct linkages to central metabolism through critical redox intermediates such as NAD(+), S-adenosyl methionine, and 2-oxoglutarate. Fluctuations in these intermediates caused by both normal and pathologic stimuli may thus have direct effects on epigenetic signaling that lead to measurable changes in gene expression. In this comprehensive review, we present surveys of both metabolism-sensitive epigenetic enzymes and the metabolic processes that may play a role in their regulation. To close, we provide a series of clinically relevant illustrations of the communication between metabolism and epigenetics in the pathogenesis of cardiovascular disease, Alzheimer disease, cancer, and environmental toxicity. We anticipate that the regulatory mechanisms described herein will play an increasingly large role in our understanding of human health and disease as epigenetics research progresses.
Collapse
Affiliation(s)
- Anthony R Cyr
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242-1181, USA
| | | |
Collapse
|
36
|
Espey MG, Chen P, Chalmers B, Drisko J, Sun AY, Levine M, Chen Q. Pharmacologic ascorbate synergizes with gemcitabine in preclinical models of pancreatic cancer. Free Radic Biol Med 2011; 50:1610-9. [PMID: 21402145 PMCID: PMC3482496 DOI: 10.1016/j.freeradbiomed.2011.03.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/01/2011] [Accepted: 03/04/2011] [Indexed: 12/19/2022]
Abstract
Conventional treatment approaches have had little impact on the course of pancreatic cancer, which has the highest fatality rate among cancers. Gemcitabine, the primary therapeutic agent for pancreatic carcinoma, produces minimal survival benefit as a single agent. Therefore, numerous efforts have focused on gemcitabine combination treatments. Using a ratio design, this study established that combining pharmacologically achievable concentrations of ascorbate with gemcitabine resulted in a synergistic cytotoxic response in eight pancreatic tumor cell lines. Sensitization was evident regardless of inherent gemcitabine resistance and epithelial-mesenchymal phenotype. Our analysis suggested that the promiscuous oxidative actions of H(2)O(2) derived from pharmacologic ascorbate can culminate in synergism independent of the cancer cell's underlying phenotype and resistance to gemcitabine monotherapy. Gemcitabine-ascorbate combinations administered to mice bearing pancreatic tumor xenografts consistently enhanced inhibition of growth compared to gemcitabine alone, produced 50% growth inhibition in a tumor type not responsive to gemcitabine, and demonstrated a gemcitabine dose-sparing effect. These data support the testing of pharmacologic ascorbate in adjunctive treatments for cancers prone to high failure rates with conventional therapeutic regimens, such as pancreatic cancer.
Collapse
Affiliation(s)
- Michael Graham Espey
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ping Chen
- Program in Integrative Medicine, Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Genetic and Molecular Biology, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Brian Chalmers
- Program in Integrative Medicine, Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jeanne Drisko
- Program in Integrative Medicine, Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrew Y. Sun
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qi Chen
- Program in Integrative Medicine, Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
37
|
Levine M, Padayatty SJ, Espey MG. Vitamin C: a concentration-function approach yields pharmacology and therapeutic discoveries. Adv Nutr 2011; 2:78-88. [PMID: 22332036 PMCID: PMC3065766 DOI: 10.3945/an.110.000109] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A concentration-function approach to vitamin C (ascorbate) has yielded new physiology and pharmacology discoveries. To determine the range of vitamin C concentrations possible in humans, pharmacokinetics studies were conducted. They showed that when vitamin C is ingested by mouth, plasma and tissue concentrations are tightly controlled by at least 3 mechanisms in healthy humans: absorption, tissue accumulation, and renal reabsorption. A 4th mechanism, rate of utilization, may be important in disease. With ingested amounts found in foods, vitamin C plasma concentrations do not exceed 100 μmol/L. Even with supplementation approaching maximally tolerated doses, ascorbate plasma concentrations are always <250 μmol/L and frequently <150 μmol/L. By contrast, when ascorbate is i.v. injected, tight control is bypassed until excess ascorbate is eliminated by glomerular filtration and renal excretion. With i.v. infusion, pharmacologic ascorbate concentrations of 25-30 mmol/L are safely achieved. Pharmacologic ascorbate can act as a pro-drug for hydrogen peroxide (H(2)O(2)) formation, which can lead to extracellular fluid at concentrations as high as 200 μmol/L. Pharmacologic ascorbate can elicit cytotoxicity toward cancer cells and slow the growth of tumors in experimental murine models. The effects of pharmacologic ascorbate should be further studied in diseases, such as cancer and infections, which may respond to generation of reactive oxygen species via H(2)O(2).
Collapse
Affiliation(s)
- Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1372, USA.
| | | | | |
Collapse
|
38
|
Frömberg A, Gutsch D, Schulze D, Vollbracht C, Weiss G, Czubayko F, Aigner A. Ascorbate exerts anti-proliferative effects through cell cycle inhibition and sensitizes tumor cells towards cytostatic drugs. Cancer Chemother Pharmacol 2010; 67:1157-66. [PMID: 20694726 PMCID: PMC3082037 DOI: 10.1007/s00280-010-1418-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 07/20/2010] [Indexed: 12/31/2022]
Abstract
Purpose While the benefits of ascorbic acid (vitamin C, ascorbate) as an essential nutrient are well established, its effects on tumor cells and in tumor treatment are controversial. In particular, conflicting data exist whether ascorbate may increase the cytotoxic effects of antineoplastic drugs or may rather exert adverse effects on drug sensitivity during cancer treatment. Findings are further obscured regarding the distinction between ascorbate and dehydroascorbate (DHA). Thus, the purpose of this study was to evaluate and directly compare the cytotoxic efficacy of ascorbate compared to DHA, and to analyse if ascorbate at pharmacological concentrations affects the efficacy of antineoplastic agents in prostate carcinoma cells. Methods We directly compare the effects of ascorbate (supplied as ‘Pascorbin® solution for injection’) and DHA on tumor cell viability, and determine IC50 values for various cell lines. At concentrations well below the IC50, ascorbate effects on cell proliferation and cell cycle are analysed. We furthermore determine changes in cellular sensitivity towards various cytostatic drugs upon pre-treatment of cells with ascorbate. Results We demonstrate higher therapeutic efficacy of ascorbate over DHA in various cell lines, independent of cell line-specific differences in ascorbate sensitivity, and identify the extracellular generation of H2O2 as critical mechanism of ascorbate action. We furthermore show that, in addition to pro-apoptotic effects described previously, ascorbate treatment already at concentrations well below the IC50 exerts anti-proliferative effects on tumor cells. Those are based on interference with the cell cycle, namely by inducing a G0/G1 arrest. Pre-treatment of tumor cells with ascorbate leads to increased cellular sensitivity towards Docetaxel, Epirubicin, Irinotecan and 5-FU, but not towards Oxaliplatin and Vinorelbin. For Docetaxel and 5-FU, a linear correlation between this sensitizing effect and the ascorbate dosage is observed. Conclusions The redox-active form of vitamin C, ascorbate, shows therapeutic efficacy in tumor cells. These antitumor effects of ascorbate are mainly based on its extracellular action and, in addition to the induction of apoptosis, also include an anti-proliferative effect by inducing cell cycle arrest. Furthermore, ascorbate treatment specifically enhances the cytostatic potency of certain chemotherapeutics, which implicates therapeutic benefit during tumor treatment. Electronic supplementary material The online version of this article (doi:10.1007/s00280-010-1418-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anja Frömberg
- Institute of Pharmacology, Faculty of Medicine, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, 35032 Marburg, Germany
| | - Daniela Gutsch
- Institute of Pharmacology, Faculty of Medicine, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, 35032 Marburg, Germany
| | - Daniel Schulze
- Institute of Pharmacology, Faculty of Medicine, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, 35032 Marburg, Germany
| | | | | | - Frank Czubayko
- Institute of Pharmacology, Faculty of Medicine, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, 35032 Marburg, Germany
| | - Achim Aigner
- Institute of Pharmacology, Faculty of Medicine, Philipps-University Marburg, Karl-von-Frisch-Strasse 1, 35032 Marburg, Germany
| |
Collapse
|
39
|
Padayatty SJ, Sun AY, Chen Q, Espey MG, Drisko J, Levine M. Vitamin C: intravenous use by complementary and alternative medicine practitioners and adverse effects. PLoS One 2010; 5:e11414. [PMID: 20628650 PMCID: PMC2898816 DOI: 10.1371/journal.pone.0011414] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 05/30/2010] [Indexed: 12/25/2022] Open
Abstract
Background Anecdotal information and case reports suggest that intravenously administered vitamin C is used by Complementary and Alternate Medicine (CAM) practitioners. The scale of such use in the U.S. and associated side effects are unknown. Methods and Findings We surveyed attendees at annual CAM Conferences in 2006 and 2008, and determined sales of intravenous vitamin C by major U.S. manufacturers/distributors. We also queried practitioners for side effects, compiled published cases, and analyzed FDA's Adverse Events Database. Of 199 survey respondents (out of 550), 172 practitioners administered IV vitamin C to 11,233 patients in 2006 and 8876 patients in 2008. Average dose was 28 grams every 4 days, with 22 total treatments per patient. Estimated yearly doses used (as 25g/50ml vials) were 318,539 in 2006 and 354,647 in 2008. Manufacturers' yearly sales were 750,000 and 855,000 vials, respectively. Common reasons for treatment included infection, cancer, and fatigue. Of 9,328 patients for whom data is available, 101 had side effects, mostly minor, including lethargy/fatigue in 59 patients, change in mental status in 21 patients and vein irritation/phlebitis in 6 patients. Publications documented serious adverse events, including 2 deaths in patients known to be at risk for IV vitamin C. Due to confounding causes, the FDA Adverse Events Database was uninformative. Total numbers of patients treated in the US with high dose vitamin C cannot be accurately estimated from this study. Conclusions High dose IV vitamin C is in unexpectedly wide use by CAM practitioners. Other than the known complications of IV vitamin C in those with renal impairment or glucose 6 phosphate dehydrogenase deficiency, high dose intravenous vitamin C appears to be remarkably safe. Physicians should inquire about IV vitamin C use in patients with cancer, chronic, untreatable, or intractable conditions and be observant of unexpected harm, drug interactions, or benefit.
Collapse
Affiliation(s)
- Sebastian J. Padayatty
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrew Y. Sun
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qi Chen
- Program in Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Michael Graham Espey
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeanne Drisko
- Program in Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Mark Levine
- Molecular and Clinical Nutrition Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
Corti A, Casini AF, Pompella A. Cellular pathways for transport and efflux of ascorbate and dehydroascorbate. Arch Biochem Biophys 2010; 500:107-15. [PMID: 20494648 DOI: 10.1016/j.abb.2010.05.014] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 11/17/2022]
Abstract
The mechanisms allowing the cellular transport of ascorbic acid represent a primary aspect for the understanding of the roles played by this vitamin in pathophysiology. Considerable research effort has been spent in the field, on several animal models and different cell types. Several mechanisms have been described to date, mediating the movements of different redox forms of ascorbic acid across cell membranes. Vitamin C can enter cells both in its reduced and oxidized form, ascorbic acid (AA) and dehydroascorbate (DHA), utilizing respectively sodium-dependent transporters (SVCT) or glucose transporters (GLUT). Modulation of SVCT expression and function has been described by cytokines, steroids and post-translational protein modification. Cellular uptake of DHA is followed by its intracellular reduction to AA by several enzymatic and non-enzymatic systems. Efflux of vitamin C has been also described in a number of cell types and different pathophysiological functions were proposed for this phenomenon, in dependence of the cell model studied. Cellular efflux of AA is mediated through volume-sensitive (VSOAC) and Ca(2+)-dependent anion channels, gap-junction hemichannels, exocytosis of secretory vesicles and possibly through homo- and hetero-exchange systems at the plasma membrane level. Altogether, available data suggest that cellular efflux of ascorbic acid - besides its uptake - should be taken into account when evaluating the cellular homeostasis and functions of this important vitamin.
Collapse
Affiliation(s)
- Alessandro Corti
- Dipartimento di Patologia Sperimentale, Università di Pisa, Italy.
| | | | | |
Collapse
|
41
|
High dose of ascorbic acid induces cell death in mesothelioma cells. Biochem Biophys Res Commun 2010; 394:249-53. [PMID: 20171954 DOI: 10.1016/j.bbrc.2010.02.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 02/03/2010] [Indexed: 11/20/2022]
Abstract
Malignant mesothelioma is an asbestos-related fatal disease with no effective cure. Recently, high dose of ascorbate in cancer treatment has been reexamined. We studied whether high dose of ascorbic acid induced cell death of four human mesothelioma cell lines. High dose of ascorbic acid induced cell death of all mesothelioma cell lines in a dose-dependent manner. We further clarified the cell killing mechanism that ascorbic acid induced reactive oxygen species and impaired mitochondrial membrane potential. In vivo experiment, intravenous administration of ascorbic acid significantly decreased the growth rate of mesothelioma tumor inoculated in mice. These data suggest that ascorbic acid may have benefits for patients with mesothelioma.
Collapse
|
42
|
Bocci V. The Actual Six Therapeutic Modalities. OZONE 2010. [PMCID: PMC7498887 DOI: 10.1007/978-90-481-9234-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Velio Bocci
- Department of Physiology, University of Siena, via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
43
|
Abstract
The reader may be eager to examine in which diseases ozonetherapy can be proficiently used and she/he will be amazed by the versatility of this complementary approach (Table 9 1). The fact that the medical applications are numerous exposes the ozonetherapist to medical derision because superficial observers or sarcastic sceptics consider ozonetherapy as the modern panacea. This seems so because ozone, like oxygen, is a molecule able to act simultaneously on several blood components with different functions but, as we shall discuss, ozonetherapy is not a panacea. The ozone messengers ROS and LOPs can act either locally or systemically in practically all cells of an organism. In contrast to the dogma that “ozone is always toxic”, three decades of clinical experience, although mostly acquired in private clinics in millions of patients, have shown that ozone can act as a disinfectant, an oxygen donor, an immunomodulator, a paradoxical inducer of antioxidant enzymes, a metabolic enhancer, an inducer of endothelial nitric oxide synthase and possibly an activator of stem cells with consequent neovascularization and tissue reconstruction.
Collapse
Affiliation(s)
- Velio Bocci
- Department of Physiology, University of Siena, via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|