1
|
Paller CJ, Zahurak ML, Mandl A, Metri NA, Lalji A, Heath E, Kelly WK, Hoimes C, Barata P, Taksey J, Garrison DA, Patra K, Milne GL, Anders NM, Nauroth JM, Durham JN, Marshall CH, Markowski MC, Eisenberger MA, Antonarakis ES, Carducci MA, Denmeade SR, Levine M. High-Dose Intravenous Vitamin C Combined with Docetaxel in Men with Metastatic Castration-Resistant Prostate Cancer: A Randomized Placebo-Controlled Phase II Trial. CANCER RESEARCH COMMUNICATIONS 2024; 4:2174-2182. [PMID: 39076107 PMCID: PMC11333993 DOI: 10.1158/2767-9764.crc-24-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/21/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
High-dose intravenous vitamin C (HDIVC) administered to produce pharmacologic concentrations shows promise in preclinical models and small clinical trials, but larger prospective randomized trials are lacking. We evaluated the clinical benefit of combining HDIVC with docetaxel in patients with progressive metastatic castration-resistant prostate cancer (mCRPC). In this double-blind, placebo-controlled phase II trial, 47 patients were randomized 2:1 to receive docetaxel (75 mg/m2 i.v.) with either HDIVC (1 g/kg) or placebo. Coprimary endpoints were PSA50 response and adverse event rates. Secondary endpoints included overall survival, radiographic progression-free survival, and quality of life measured using the Functional Assessment of Cancer Therapy-Prostate instrument. Correlative analyses included pharmacokinetics and oxidative stress markers. Eighty-nine percent of patients previously had three or more lines of therapy. The PSA50 response rate was 41% in the HDIVC group and 33% in the placebo group (P = 0.44), with comparable adverse event rates in both groups. There were no significant differences in Functional Assessment of Cancer Therapy-Prostate scores. The median radiographic progression-free survival was not significantly different between the HDIVC and placebo groups, with durations of 10.1 and 10.0 months (HR, 1.35; 95% confidence interval, 0.66-2.75; P = 0.40), respectively. The median overall survival was 15.2 months in the HDIVC group and 29.5 months in the placebo group (HR, 1.98; 95% confidence interval, 0.85-4.58; P = 0.11). HDIVC did not decrease F2-isoprostanes, indicators of oxidative stress. The study was suspended after prespecified interim analysis indicated futility in achieving primary endpoints. In this patient population, combining HDIVC with docetaxel did not improve PSA response, toxicity, or other clinical outcomes compared with docetaxel alone. Findings do not support the routine use of HDIVC in mCRPC treatment outside of clinical trials. SIGNIFICANCE This is the first randomized, placebo-controlled, double-blind trial to evaluate HDIVC in cancer treatment. The addition of HDIVC to docetaxel in patients with mCRPC does not improve PSA response, toxicity, or other clinical outcomes compared with docetaxel alone. The routine use of HDIVC in mCRPC treatment is not supported outside of clinical trials.
Collapse
Affiliation(s)
- Channing J. Paller
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Marianna L. Zahurak
- Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, Maryland.
| | - Adel Mandl
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Nicole A. Metri
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Aliya Lalji
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | | | | | | | - Pedro Barata
- Case Western Reserve University/University Hospitals, Cleveland, Ohio.
| | - Jason Taksey
- Maryland Oncology Hematology, US Oncology, Annapolis, Maryland.
| | - Dominique A. Garrison
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Kartick Patra
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland.
| | - Ginger L. Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Nicole M. Anders
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Julie M. Nauroth
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Jennifer N. Durham
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Catherine H. Marshall
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mark C. Markowski
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mario A. Eisenberger
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | | | - Michael A. Carducci
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Samuel R. Denmeade
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
2
|
Isola S, Gammeri L, Furci F, Gangemi S, Pioggia G, Allegra A. Vitamin C Supplementation in the Treatment of Autoimmune and Onco-Hematological Diseases: From Prophylaxis to Adjuvant Therapy. Int J Mol Sci 2024; 25:7284. [PMID: 39000393 PMCID: PMC11241675 DOI: 10.3390/ijms25137284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
Vitamin C is a water-soluble vitamin introduced through the diet with anti-inflammatory, immunoregulatory, and antioxidant activities. Today, this vitamin is integrated into the treatment of many inflammatory pathologies. However, there is increasing evidence of possible use in treating autoimmune and neoplastic diseases. We reviewed the literature to delve deeper into the rationale for using vitamin C in treating this type of pathology. There is much evidence in the literature regarding the beneficial effects of vitamin C supplementation for treating autoimmune diseases such as Systemic Lupus Erythematosus (SLE) and Rheumatoid Arthritis (RA) and neoplasms, particularly hematological neoplastic diseases. Vitamin C integration regulates the cytokines microenvironment, modulates immune response to autoantigens and cancer cells, and regulates oxidative stress. Moreover, integration therapy has an enhanced effect on chemotherapies, ionizing radiation, and target therapy used in treating hematological neoplasm. In the future, integrative therapy will have an increasingly important role in preventing pathologies and as an adjuvant to standard treatments.
Collapse
Affiliation(s)
- Stefania Isola
- School and Operative Unit of Allergy and Clinical Immunology, Policlinico “G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (S.I.); (S.G.)
| | - Luca Gammeri
- School and Operative Unit of Allergy and Clinical Immunology, Policlinico “G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (S.I.); (S.G.)
| | - Fabiana Furci
- Provincial Healthcare Unit, Section of Allergy, 89900 Vibo Valentia, Italy;
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Policlinico “G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (S.I.); (S.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98125 Messina, Italy;
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98100 Messina, Italy;
| |
Collapse
|
3
|
Raja N, Radhakrishnan H, Masilamani S. Oxalate Nephropathy: A Case Report of Acute Kidney Injury Due to Juice Diet. Cureus 2023; 15:e51226. [PMID: 38283477 PMCID: PMC10821746 DOI: 10.7759/cureus.51226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Oxalate nephropathy occurs due to the deposition of calcium oxalate crystals in kidney tubules and/or the interstitium as a part of primary or secondary hyperoxaluria. Secondary oxalate nephropathy can occur even with moderately high doses of ascorbic acid intake under yet unidentified clinical circumstances. Vitamin C, although traditionally considered an antioxidant, leads to the formation of superoxide and subsequent generation of reactive oxidant species at pharmacologic concentrations. Ascorbic acid is partly converted to oxalic acid, which is responsible for deposition and renal tubular injury. We report a case of a diabetic patient with normal kidney function who was put on a juice diet for a week due to upper gastrointestinal bleeding. He developed acute kidney injury due to biopsy-proven oxalate nephropathy requiring dialysis. Though he was lost to follow-up after two weeks on dialysis, he was expected to have only a slow recovery or become dependent on dialysis given his age, comorbidities, and extent of tubular involvement. Hence, caution should be exercised before supplementing vitamin C either in its natural form or as a drug. Risk factors for secondary oxalate nephropathy due to excessive intake of oxalate or its precursor are likely to be age, diabetes, dehydration, and underlying chronic kidney disease. Most of the patients do not have a complete recovery of kidney function, and many become dependent on dialysis.
Collapse
Affiliation(s)
- Niranjan Raja
- Department of Nephrology, Mahatma Gandhi Medical College and Research Institute, Puducherry, IND
| | | | - Sivasankar Masilamani
- Department of Nephrology, Mahatma Gandhi Medical College and Research Institute, Puducherry, IND
| |
Collapse
|
4
|
Piatnytskyi DV, Volkov SN. Complexes of hydrogen peroxide molecules with DNA nucleic bases. J Biomol Struct Dyn 2023; 41:15003-15008. [PMID: 36995109 DOI: 10.1080/07391102.2023.2193986] [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: 12/08/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023]
Abstract
The analysis of complexes formation of hydrogen peroxide molecule with DNA nucleic bases is carried out using methods of quantum chemistry. Optimized geometries of complexes are determined and the interaction energies that lead to complex formation are calculated. Comparison with the same calculations for water molecule is made. It is shown that complexes with hydrogen peroxide molecule are energetically more stable than the same complexes with water molecule. Such energetic advantage is achieved particularly due to geometrical properties of hydrogen peroxide molecule, especially presence of dihedral angle. Position of hydrogen peroxide molecule in close vicinity to DNA could lead to blocking of its recognition by proteins or direct damage via hydroxyl radical formation. These results can have significant impact in understanding of mechanisms of cancer therapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- D V Piatnytskyi
- Laboratory of Biophysics of Macromolecules, Bogolyubov Institute for Theoretical Physics, Kyiv, Ukraine
| | - S N Volkov
- Laboratory of Biophysics of Macromolecules, Bogolyubov Institute for Theoretical Physics, Kyiv, Ukraine
| |
Collapse
|
5
|
Gordon N, Gallagher PT, Neupane NP, Mandigo AC, McCann JK, Dylgjeri E, Vasilevskaya I, McNair C, Paller CJ, Kelly WK, Knudsen KE, Shafi AA, Schiewer MJ. PARP inhibition and pharmacological ascorbate demonstrate synergy in castration-resistant prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533944. [PMID: 36993449 PMCID: PMC10055378 DOI: 10.1101/2023.03.23.533944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death for men in the United States. While organ-confined disease has reasonable expectation of cure, metastatic PCa is universally fatal upon recurrence during hormone therapy, a stage termed castration-resistant prostate cancer (CRPC). Until such time as molecularly defined subtypes can be identified and targeted using precision medicine, it is necessary to investigate new therapies that may apply to the CRPC population as a whole. The administration of ascorbate, more commonly known as ascorbic acid or Vitamin C, has proved lethal to and highly selective for a variety of cancer cell types. There are several mechanisms currently under investigation to explain how ascorbate exerts anti-cancer effects. A simplified model depicts ascorbate as a pro-drug for reactive oxygen species (ROS), which accumulate intracellularly and generate DNA damage. It was therefore hypothesized that poly(ADP-ribose) polymerase (PARP) inhibitors, by inhibiting DNA damage repair, would augment the toxicity of ascorbate. Results Two distinct CRPC models were found to be sensitive to physiologically relevant doses of ascorbate. Moreover, additional studies indicate that ascorbate inhibits CRPC growth in vitro via multiple mechanisms including disruption of cellular energy dynamics and accumulation of DNA damage. Combination studies were performed in CRPC models with ascorbate in conjunction with escalating doses of three different PARP inhibitors (niraparib, olaparib, and talazoparib). The addition of ascorbate augmented the toxicity of all three PARP inhibitors and proved synergistic with olaparib in both CRPC models. Finally, the combination of olaparib and ascorbate was tested in vivo in both castrated and non-castrated models. In both cohorts, the combination treatment significantly delayed tumor growth compared to monotherapy or untreated control. Conclusions These data indicate that pharmacological ascorbate is an effective monotherapy at physiological concentrations and kills CRPC cells. Ascorbate-induced tumor cell death was associated with disruption of cellular energy dynamics and accumulation of DNA damage. The addition of PARP inhibition increased the extent of DNA damage and proved effective at slowing CRPC growth both in vitro and in vivo. These findings nominate ascorbate and PARPi as a novel therapeutic regimen that has the potential to improve CRPC patient outcomes.
Collapse
Affiliation(s)
- Nicolas Gordon
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter T. Gallagher
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Amy C. Mandigo
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jennifer K. McCann
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Irina Vasilevskaya
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Christopher McNair
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Channing J. Paller
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Wm. Kevin Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Karen E. Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ayesha A. Shafi
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD 20817, USA. The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Matthew J. Schiewer
- Department of Urology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Pharmacology/Physiology/Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
6
|
Qin S, Wang G, Chen L, Geng H, Zheng Y, Xia C, Wu S, Yao J, Deng L. Pharmacological vitamin C inhibits mTOR signaling and tumor growth by degrading Rictor and inducing HMOX1 expression. PLoS Genet 2023; 19:e1010629. [PMID: 36787291 PMCID: PMC9928125 DOI: 10.1371/journal.pgen.1010629] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/21/2023] [Indexed: 02/15/2023] Open
Abstract
Pharmacological vitamin C (VC) is a potential natural compound for cancer treatment. However, the mechanism underlying its antitumor effects remains unclear. In this study, we found that pharmacological VC significantly inhibits the mTOR (including mTORC1 and mTORC2) pathway activation and promotes GSK3-FBXW7-mediated Rictor ubiquitination and degradation by increasing the cellular ROS. Moreover, we identified that HMOX1 is a checkpoint for pharmacological-VC-mediated mTOR inactivation, and the deletion of FBXW7 or HMOX1 suppresses the regulation of pharmacological VC on mTOR activation, cell size, cell viability, and autophagy. More importantly, it was observed that the inhibition of mTOR by pharmacological VC supplementation in vivo produces positive therapeutic responses in tumor growth, while HMOX1 deficiency rescues the inhibitory effect of pharmacological VC on tumor growth. These results demonstrate that VC influences cellular activities and tumor growth by inhibiting the mTOR pathway through Rictor and HMOX1, which may have therapeutic potential for cancer treatment.
Collapse
Affiliation(s)
- Senlin Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Guoyan Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Huijun Geng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yining Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (J.Y); (L.D)
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (J.Y); (L.D)
| |
Collapse
|
7
|
Hazan S, Dave S, Papoutsis AJ, Deshpande N, Howell MC, Martin LM. Vitamin C improves gut Bifidobacteria in humans. Future Microbiol 2022. [PMID: 36475828 DOI: 10.2217/fmb-2022-0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aims: Numerous beneficial effects of vitamin C (ascorbic acid) supplementation have been reported in the literature. However, data on its effects toward the gut microbiome are limited. We assessed the effect of vitamin C supplementation on the abundance of beneficial bacterial species in the gut microbiome. Materials and methods: Stool samples were analyzed for relative abundance of gut microbiome bacteria using next-generation sequencing-based profiling and metagenomic shotgun analysis. Results: Supplementation with vitamin C increased the abundance of bacteria of the genus Bifidobacterium (p = 0.0001) and affected various species. Conclusion: The beneficial effects of vitamin C supplementation may be attributed to modulation of the gut microbiome and the consequent health benefits thereof.
Collapse
Affiliation(s)
- Sabine Hazan
- ProgenaBIome, LLC, Ventura, CA 93003, USA
- Mcrobiome Research Foundation, Ventura, CA 93003, USA
| | - Sonya Dave
- Mcrobiome Research Foundation, Ventura, CA 93003, USA
| | | | | | | | - Leisha Ma Martin
- Texas A&M University - Corpus Christi, Department of Life Sciences, Corpus Christi, TX 78412, USA
| |
Collapse
|
8
|
Mohseni S, Tabatabaei-Malazy O, Ejtahed HS, Qorbani M, Azadbakht L, Khashayar P, Larijani B. Effect of vitamins C and E on cancer survival; a systematic review. Daru 2022; 30:427-441. [PMID: 36136247 PMCID: PMC9715902 DOI: 10.1007/s40199-022-00451-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022] Open
Abstract
PURPOSE Association between vitamins C (VC)/ E (VE) and cancer survival is inconsistent. This systematic review is aimed to summarize trials for effects of VC/VE on cancer survival. METHODS Relevant English trials were retrieved from PubMed, Cochrane Library, Embase, Web of Science, Scopus databases, and Clinicaltrials.gov through 21/June/2022. Inclusion criteria were all trials which assessed sole/combinations intake of VC/VE on survival rate, mortality, or remission of any cancer. Exclusion criteria were observational and animal studies. RESULTS We reached 30 trials conducted on 38,936 patients with various cancers. Due to severe methodological heterogeneity, meta-analysis was impossible. High dose VC + chemotherapy or radiation was safe with an overall survival (OS) 182 days - 21.5 months. Sole oral or intravenous high dose VC was safe with non-significant change in OS (2.9-8.2 months). VE plus chemotherapy was safe, resulted in stabling diseases for 5 years in 70- 86.7% of patients and OS 109 months. It was found 60% and 16% non-significant reductions in adjusted hazard ratio (HR) deaths or recurrence by 200 mg/d tocotrienol + tamoxifen in breast cancer, respectively. Sole intake of 200-3200 mg/d tocotrienol before resectable pancreatic cancer was safe and significantly increased cancer cells' apoptosis. Combination VC and VE was non-significantly reduced 7% in rate of neoplastic gastric polyp. CONCLUSION Although our study is supported improvement of survival and progression rates of cancers by VC/VE, more high quality trials with large sample sizes are required to confirm. PROSPERO REGISTRATION NUMBER CRD42020152795.
Collapse
Affiliation(s)
- Shahrzad Mohseni
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Patricia Khashayar
- Center for microsystem technology, Imec and Ghent University, 9052 Gent, Zwijnaarde, Belgium
- Osteoporosis Research Center, Endocrinology & Metabolism Clinical Science Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Ascorbate as a Bioactive Compound in Cancer Therapy: The Old Classic Strikes Back. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123818. [PMID: 35744943 PMCID: PMC9229419 DOI: 10.3390/molecules27123818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022]
Abstract
Cancer is a disease of high mortality, and its prevalence has increased steadily in the last few years. However, during the last decade, the development of modern chemotherapy schemes, new radiotherapy techniques, targeted therapies and immunotherapy has brought new hope in the treatment of these diseases. Unfortunately, cancer therapies are also associated with frequent and, sometimes, severe adverse events. Ascorbate (ascorbic acid or vitamin C) is a potent water-soluble antioxidant that is produced in most mammals but is not synthesised endogenously in humans, which lack enzymes for its synthesis. Ascorbate has antioxidant effects that correspond closely to the dose administered. Interestingly, this natural antioxidant induces oxidative stress when given intravenously at a high dose, a paradoxical effect due to its interactions with iron. Importantly, this deleterious property of ascorbate can result in increased cell death. Although, historically, ascorbate has been reported to exhibit anti-tumour properties, this effect has been questioned due to the lack of available mechanistic detail. Recently, new evidence has emerged implicating ferroptosis in several types of oxidative stress-mediated cell death, such as those associated with ischemia–reperfusion. This effect could be positively modulated by the interaction of iron and high ascorbate dosing, particularly in cell systems having a high mitotic index. In addition, it has been reported that ascorbate may behave as an adjuvant of favourable anti-tumour effects in cancer therapies such as radiotherapy, radio-chemotherapy, chemotherapy, immunotherapy, or even in monotherapy, as it facilitates tumour cell death through the generation of reactive oxygen species and ferroptosis. In this review, we provide evidence supporting the view that ascorbate should be revisited to develop novel, safe strategies in the treatment of cancer to achieve their application in human medicine.
Collapse
|
10
|
Role of Vitamin C in Selected Malignant Neoplasms in Women. Nutrients 2022; 14:nu14040882. [PMID: 35215535 PMCID: PMC8876016 DOI: 10.3390/nu14040882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
Since the first reports describing the anti-cancer properties of vitamin C published several decades ago, its actual effectiveness in fighting cancer has been under investigation and widely discussed. Some scientific reports indicate that vitamin C in high concentrations can contribute to effective and selective destruction of cancer cells. Furthermore, preclinical and clinical studies have shown that relatively high doses of vitamin C administered intravenously in ‘pharmacological concentrations’ may not only be well-tolerated, but significantly improve patients’ quality of life. This seems to be particularly important, especially for terminal cancer patients. However, the relatively high frequency of vitamin C use by cancer patients means that the potential clinical benefits may not be obvious. For this reason, in this review article, we focus on the articles published mainly in the last two decades, describing possible beneficial effects of vitamin C in preventing and treating selected malignant neoplasms in women, including breast, cervical, endometrial, and ovarian cancer. According to the reviewed studies, vitamin C use may contribute to an improvement of the overall quality of life of patients, among others, by reducing chemotherapy-related side effects. Nevertheless, new clinical trials are needed to collect stronger evidence of the role of this nutrient in supportive cancer treatment.
Collapse
|
11
|
Hemilä H, Chalker E. Bias against Vitamin C in Mainstream Medicine: Examples from Trials of Vitamin C for Infections. Life (Basel) 2022; 12:62. [PMID: 35054455 PMCID: PMC8779885 DOI: 10.3390/life12010062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023] Open
Abstract
Evidence has shown unambiguously that, in certain contexts, vitamin C is effective against the common cold. However, in mainstream medicine, the views on vitamin C and infections have been determined by eminence-based medicine rather than evidence-based medicine. The rejection of the demonstrated benefits of vitamin C is largely explained by three papers published in 1975-two published in JAMA and one in the American Journal of Medicine-all of which have been standard citations in textbooks of medicine and nutrition and in nutritional recommendations. Two of the papers were authored by Thomas Chalmers, an influential expert in clinical trials, and the third was authored by Paul Meier, a famous medical statistician. In this paper, we summarize several flaws in the three papers. In addition, we describe problems with two recent randomized trial reports published in JAMA which were presented in a way that misled readers. We also discuss shortcomings in three recent JAMA editorials on vitamin C. While most of our examples are from JAMA, it is not the only journal with apparent bias against vitamin C, but it illustrates the general views in mainstream medicine. We also consider potential explanations for the widespread bias against vitamin C.
Collapse
Affiliation(s)
- Harri Hemilä
- Department of Public Health, University of Helsinki, FI-00014 Helsinki, Finland
| | - Elizabeth Chalker
- Biological Data Science Institute, Australian National University, Canberra, ACT 2600, Australia;
| |
Collapse
|
12
|
Smith-Díaz CC, Magon NJ, McKenzie JL, Hampton MB, Vissers MCM, Das AB. Ascorbate Inhibits Proliferation and Promotes Myeloid Differentiation in TP53-Mutant Leukemia. Front Oncol 2021; 11:709543. [PMID: 34497762 PMCID: PMC8419345 DOI: 10.3389/fonc.2021.709543] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022] Open
Abstract
Loss-of-function mutations in the DNA demethylase TET2 are associated with the dysregulation of hematopoietic stem cell differentiation and arise in approximately 10% of de novo acute myeloid leukemia (AML). TET2 mutations coexist with other mutations in AML, including TP53 mutations, which can indicate a particularly poor prognosis. Ascorbate can function as an epigenetic therapeutic in pathological contexts involving heterozygous TET2 mutations by restoring TET2 activity. How this response is affected when myeloid leukemia cells harbor mutations in both TET2 and TP53 is unknown. Therefore, we examined the effects of ascorbate on the SKM-1 AML cell line that has mutated TET2 and TP53. Sustained treatment with ascorbate inhibited proliferation and promoted the differentiation of these cells. Furthermore, ascorbate treatment significantly increased 5-hydroxymethylcytosine, suggesting increased TET activity as the likely mechanism. We also investigated whether ascorbate affected the cytotoxicity of Prima-1Met, a drug that reactivates some p53 mutants and is currently in clinical trials for AML. We found that the addition of ascorbate had a minimal effect on Prima-1Met–induced cytotoxicity, with small increases or decreases in cytotoxicity being observed depending on the timing of treatment. Collectively, these data suggest that ascorbate could exert a beneficial anti-proliferative effect on AML cells harboring both TET2 and TP53 mutations whilst not interfering with targeted cytotoxic therapies such as Prima-1Met.
Collapse
Affiliation(s)
- Carlos C Smith-Díaz
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Nicholas J Magon
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Judith L McKenzie
- Haematology Research Group, Christchurch Hospital and Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Andrew B Das
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
13
|
Abstract
Significance: Persistent oxidative stress is a common feature of cancer cells, giving a specific weapon to selectively eliminate them. Ascorbate in pharmacological concentration can contribute to the suspended formation of hydroxyl radical via the Fenton reaction; thus, it can be an important element of the oxidative stress therapy against cancer cells. Recent Advances: The main components of ascorbate-induced cell death are DNA double-strand breaks via the production of hydroxyl radical and ATP depletion due to the activation of poly (ADP-ribose) polymerase 1. Presumably, DNA damage can be the primary contributor to the anticancer activity of pharmacological ascorbate, as opposed to the rupture of bioenergetics. The caspase independency of high-dose ascorbate-induced cell death proposed the possible involvement of several types of cell death, such as ferroptosis, necroptosis, and autophagy. Critical Issues: Ascorbate can target at least two key molecular features of cancer cells as a part of the anticancer therapy: the intrinsic or acquired resistance to cell death and the dysregulated metabolism of cancer cells. It seems probable that different concentrations of ascorbate alter the nature of induced cell death. Autophagy and necroptosis may play a role at intermediate concentrations, but caspase-independent apoptosis may dominate at higher concentrations. However, ascorbate behaves as an effective inhibitor of ferroptosis that may have crucial importance in its possible clinical application. Future Directions: The elucidation of the details and the links between high-dose ascorbate-induced cancer selective cell death mechanisms may give us a tool to form and apply synergistic cancer therapies. Antioxid. Redox Signal. 34, 831-844.
Collapse
Affiliation(s)
- András Szarka
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
| | - Orsolya Kapuy
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Tamás Lőrincz
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
14
|
Rajendran G, Taylor JA, Woolbright BL. Natural products as a means of overcoming cisplatin chemoresistance in bladder cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:69-84. [PMID: 35582013 PMCID: PMC9019192 DOI: 10.20517/cdr.2020.69] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
Cisplatin remains an integral part of the treatment for muscle invasive bladder cancer. A large number of patients do not respond to cisplatin-based chemotherapy and efficacious salvage regimens are limited. Immunotherapy has offered a second line of treatment; however, only approximately 20% of patients respond, and molecular subtyping of tumors indicates there may be significant overlap in those patients that respond to cisplatin and those patients that respond to immunotherapy. As such, restoring sensitivity to cisplatin remains a major hurdle to improving patient care. One potential source of compounds for enhancing cisplatin is naturally derived bioactive products such as phytochemicals, flavonoids and others. These compounds can activate a diverse array of different pathways, many of which can directly promote or inhibit cisplatin sensitivity. The purpose of this review is to understand current drug development in the area of natural products and to assess how these compounds may enhance cisplatin treatment in bladder cancer patients.
Collapse
Affiliation(s)
- Ganeshkumar Rajendran
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Benjamin L Woolbright
- Department of Urology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
15
|
High-Dose Vitamin C in Advanced-Stage Cancer Patients. Nutrients 2021; 13:nu13030735. [PMID: 33652579 PMCID: PMC7996511 DOI: 10.3390/nu13030735] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
High-dose intravenously administered vitamin C (IVC) is widely used in cancer patients by complementary and alternative medicine practitioners. The most frequent indications for IVC therapy result from the belief in its effectiveness as a potent anti-cancer agent which additionally enhances chemosensitivity of cancer cells and reduces chemotherapy-related toxicities and fatigue intensity. In this narrative review, we decided to deal with this issue, trying to answer the question whether there is any scientific evidence supporting the rationale for application of high-dose IVC therapy in advanced-stage cancer patients. Although results obtained from preclinical studies demonstrated that millimolar ascorbate plasma concentrations achievable only after IVC administration were cytotoxic to fast-growing malignant cells and inhibited tumor growth as well as prolonged the survival of laboratory animals, such positive effects were not found in human studies with advanced-stage cancer patients. We also have not found the rationale for the use of IVC to increase the effectiveness of chemotherapy and to reduce the chemotherapy-induced toxicity in the above mentioned group. Nevertheless, in palliative care, high-dose IVC might be considered as a therapy improving the quality of life and reducing cancer-related symptoms, such as fatigue and bone pain. However, because of the absence of placebo-controlled randomized trials on IVC efficacy in advanced-stage cancer patients, the placebo effect cannot be excluded.
Collapse
|
16
|
Abstract
![]()
Vitamin
C (ascorbic acid) is a water-soluble antioxidant and a
cofactor for a large number of enzymes. It is present in all tissues
and especially abundant in corneal epithelium, stem cells, and neurons.
Although similar to thiols in its ability to react with many reactive
oxygen species (ROS), ascorbate is much better (>100× faster)
than glutathione at scavenging of primary ROS (superoxide radical
and singlet oxygen). Ascorbate appears to be especially important
for elimination of O2•– in the
nucleus which contains little or no SOD activity. Cofactor functions
of ascorbate involve the maintenance of activity of Fe(II)/2-oxoglutarate-dependent
dioxygenases via reduction of Fe(III). The most prominent activity
of ascorbate-dependent dioxygenases in the cytoplasm is hydroxylation
of prolines in proteins involved in the formation of extracellular
matrix and regulation of metabolism and hypoxia responses. In the
nucleus, ascorbate is important for oxidative demethylation of 5-methylcytosine
in DNA (by TET proteins) and removal of methyl groups from histone
lysines (by JmjC demethylases). Differentiation and other cellular
reprograming processes involving DNA demethylation are especially
sensitive to ascorbate insufficiency. High doses of vitamin C alone
or in combinations with drugs produced cancer-suppressive effects
which involved redox, immune, and epigenetic mechanisms. Solutions
to vitamin C deficiency in cultured cells are discussed to improve
the physiological relevance of in vitro models. An
abundance of vitamin C in rodents limits their ability to fully recapitulate
human sensitivity to adverse health effects of malnutrition and xenobiotics,
including neurotoxicity, lung injury, and intergenerational and other
epigenetic effects.
Collapse
Affiliation(s)
- Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| |
Collapse
|
17
|
Klener P, Scott Alexander M, Cullen JJ, Stejskal V, Sliva J, Kotlarova L, Kostiuk P, Prochazka Z, Kucerova M. The benefits of ascorbate to protect healthy cells in the prevention and treatment of oncological diseases. J Appl Biomed 2020; 18:1-7. [PMID: 34907706 DOI: 10.32725/jab.2020.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/07/2020] [Indexed: 11/05/2022] Open
Abstract
Health status is determined by the balance of oxidants and antioxidants which protects healthy cells against the threat of internal and external risk factors. Antioxidants such as ascorbate (vitamin C, ascorbic acid) are of fundamental importance in this respect. Ascorbate neutralizes potential damage caused by cellular oxidative stress which may be the greatest risk of damage to healthy tissue. Cellular oxidative stress is mediated by external factors (e.g. psychological stress, physical exertion, drugs, various diseases, environmental pollution, preservatives, smoking, and alcohol) and internal factors (products of cellular metabolism including reactive oxygen species). When the products of oxidative stress are not sufficiently neutralized, healthy cells are at risk for both mitochondrial and DNA damage. In the short term, cell function may deteriorate, while an increased production of proinflammatory cytokines over time may lead to the development of chronic inflammatory changes and diseases, including cancer. Although pharmaceutical research continues to bring effective chemotherapeutic agents to the market, a limiting factor is often the normal tissue and organ toxicity of these substances, which leads to oxidative stress on healthy tissue. There is increasing interest and imperative to protect healthy tissues from the negative effects of radio-chemotherapeutic treatment. The action of ascorbate against the development of oxidative stress may justify its use not only in the prevention of carcinogenesis, but as a part of supportive or complementary therapy during treatment. Ascorbate (particularly when administered parentally at high doses) may have antioxidant effects that work to protect healthy cells and improve patient tolerability to some toxic radio-chemotherapy regimens. Additionally, ascorbate has demonstrated an immunomodulatory effect by supporting mechanisms essential to anti-tumor immunity. Intravenous administration of gram doses of vitamin C produce high plasma levels immediately, but the levels drop rapidly. Following oral vitamin C administration, plasma levels increase slowly to relatively low values, and then gradually decay. With an oral liposomal formulation, significantly higher levels are attainable than with standard oral formulations. Therefore, oral administration of liposomal vitamin C appears to be an optimal adjunct to intravenous administration. In this review, the basic mechanisms and clinical benefits of ascorbate as an antioxidant that may be useful as complementary therapy to chemotherapeutic regimens will be discussed.
Collapse
Affiliation(s)
| | | | - Joseph John Cullen
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Vera Stejskal
- University of Stockholm, Dept of Immunology, Wenner Gren Center, Stockholm, Sweden; Research Centre for Toxic Compounds in the Environment, Masaryk University, Brno, Czech Republic
| | - Jiri Sliva
- Charles University, Department of Pharmacology, 3rd Faculty of Medicine, Prague, Czech Republic
| | - Lucie Kotlarova
- InPharmClinic, Department of Pharmacology, Prague, Czech Republic
| | - Pavel Kostiuk
- Edukafarm, Department of Pharmacology, Prague, Czech Republic
| | | | - Marta Kucerova
- Hospital Jablonec nad Nisou, Department of Oncology, Jablonec nad Nisou, Czech Republic
| |
Collapse
|
18
|
Gregoraszczuk EL, Zajda K, Tekla J, Respekta N, Zdybał P, Such A. Vitamin C supplementation had no side effect in non-cancer, but had anticancer properties in ovarian cancer cells. INT J VITAM NUTR RES 2020; 91:293-303. [PMID: 32008465 DOI: 10.1024/0300-9831/a000634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vitamin C (Vit C) has been widely used in the treatment and prevention of cancer. Nevertheless, the clinical results are still inconclusive. Using non-cancer (HOSEpiC) and cancer OVCAR-3 cells cultured in basal medium or in ovarian cancer-associated fibroblast (CAF)-supplemented medium, we estimated the dose-dependent effect of Vit C on sodium-ascorbate co-transporters (SVCT1, SVCT2) and glucose transporter (GLUT1) protein expression. Additionally, the action of Vit C on cell proliferation (alamarBlue), membrane permeability (LDH assay), caspase3 activity, the selected cell cycle and apoptosis pathway, poly(ADP-ribose) polymerase-1 (PARP) protein expression, and reactive oxygen species (ROS) activity was determined. We showed different effects of Vit C on the expression of the co-transporter in non-cancer and cancer cells. In non-cancer cells, Vit C, at a pharmacological concentration, increased SVCT2 and decreased GLUT1, while the opposite effect was noted in cancer cells. In cancer cells, Vit C, in a pharmacological dose, decreased cell proliferation through an inhibitory effect on cyclin-dependent kinase 2 (CDK2) (4.4-fold; p < 0.01), mainly due to the stimulatory effect on the expression of cyclin-dependent kinase (CDK) inhibitors, such as p21 and p53 (3.2- and 2.8-fold, respectively; p < 0.001), but not caspase pathway. The tumour microenvironment caused inefficiency of the lower doses of Vit C in ovarian cancer cells. At a pharmacological dose of 1 mM, Vit C decreased PARP expression (1.5-fold; p < 0.05). We suggest that it's nontoxic effects on non-cancer cells may be an indicator of its prophylactic use, while in a pharmacological dose Vit C should be considered a possible adjunctive drug in ovarian cancer. However, it is necessary to consider the effect of the CAF.
Collapse
Affiliation(s)
- Ewa Lucja Gregoraszczuk
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Karolina Zajda
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Joanna Tekla
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Natalia Respekta
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Paweł Zdybał
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Aleksandra Such
- Department of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| |
Collapse
|
19
|
Satheesh NJ, Samuel SM, Büsselberg D. Combination Therapy with Vitamin C Could Eradicate Cancer Stem Cells. Biomolecules 2020; 10:biom10010079. [PMID: 31947879 PMCID: PMC7022456 DOI: 10.3390/biom10010079] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer remains one of the most feared and dreaded diseases in this era of modern medicine, claiming the lives of many, and affecting the quality of life of several others around the globe despite major advances in the diagnosis, treatment, palliative care and the immense resources invested into cancer research. While research in cancer has largely focused on the neoplasm/tumor and the cancerous cells that make up the tumor, more recently, the existence, proliferation, differentiation, migration and invasion of cancer stem cells (CSCs) and the role that CSCs play in tumor initiation, progression, metastasis, drug resistance and relapse/recurrence of the disease has gained widespread interest in cancer research. Although the conventional therapeutic approaches such as surgery, chemotherapy and radiation therapy are effective cancer treatments, very often these treatment modalities fail to target the CSCs, which then later become the source of disease recurrence. A majority of the anti-cancer agents target rapidly dividing cancer cells and normal cells and hence, have side effects that are not expected. Targeting CSCs remains a challenge due to their deviant nature with a low proliferation rate and increased drug resistance mechanism. Ascorbic acid/Vitamin C (Vit.C), a potent antioxidant, is a cofactor for several biosynthetic and gene regulatory enzymes and a vital contributor to immune defense of the body, and was found to be deficient in patients with advanced stages of cancer. Vit.C has gained importance in the treatment of cancer due to its ability to modulate the redox status of the cell and influence epigenetic modifications and significant roles in HIF1α signaling. Studies have reported that intravenous administration of Vit.C at pharmacological doses selectively kills tumor cells and targets CSCs when administered along with chemotherapeutic drugs. In the current article, we provide an in-depth review of how Vit.C plays an important role in targeting CSCs and its possible use as an adjuvant, neoadjuvant or co-treatment in the treatment of cancers.
Collapse
|
20
|
Vitamin C in Plants: From Functions to Biofortification. Antioxidants (Basel) 2019; 8:antiox8110519. [PMID: 31671820 PMCID: PMC6912510 DOI: 10.3390/antiox8110519] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/25/2019] [Accepted: 10/26/2019] [Indexed: 12/18/2022] Open
Abstract
Vitamin C (l-ascorbic acid) is an excellent free radical scavenger, not only for its capability to donate reducing equivalents but also for the relative stability of the derived monodehydroascorbate radical. However, vitamin C is not only an antioxidant, since it is also a cofactor for numerous enzymes involved in plant and human metabolism. In humans, vitamin C takes part in various physiological processes, such as iron absorption, collagen synthesis, immune stimulation, and epigenetic regulation. Due to the functional loss of the gene coding for l-gulonolactone oxidase, humans cannot synthesize vitamin C; thus, they principally utilize plant-based foods for their needs. For this reason, increasing the vitamin C content of crops could have helpful effects on human health. To achieve this objective, exhaustive knowledge of the metabolism and functions of vitamin C in plants is needed. In this review, the multiple roles of vitamin C in plant physiology as well as the regulation of its content, through biosynthetic or recycling pathways, are analyzed. Finally, attention is paid to the strategies that have been used to increase the content of vitamin C in crops, emphasizing not only the improvement of nutritional value of the crops but also the acquisition of plant stress resistance.
Collapse
|
21
|
Zhang X, Liu T, Li Z, Feng Y, Corpe C, Liu S, Zhang J, He X, Liu F, Xu L, Shen L, Li S, Xia Q, Peng X, Zhou X, Chen W, Zhang X, Xu J, Wang J. Hepatomas are exquisitely sensitive to pharmacologic ascorbate (P-AscH -). Am J Cancer Res 2019; 9:8109-8126. [PMID: 31754384 PMCID: PMC6857065 DOI: 10.7150/thno.35378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023] Open
Abstract
Rationale: Ascorbate is an essential micronutrient known for redox functions at normal physiologic concentrations. In recent decades, pharmacological ascorbate has been found to selectively kill tumour cells. However, the dosing frequency of pharmacologic ascorbate in humans has not yet been defined. Methods: We determined that among five hepatic cell lines, Huh-7 cells were the most sensitive to ascorbate. The effects of high-dose ascorbate on hepatoma were therefore assessed using Huh-7 cells and xenograft tumour mouse model. Results: In Huh-7 cells, ascorbate induced a significant increase in the percentage of cells in the G0/G1 phase, apoptosis and intracellular levels of ROS. High doses of ascorbate (4.0 pmol cell-1), but not low doses of ascorbate (1.0 pmol cell-1), also served as a pro-drug that killed hepatoma cells by altering mitochondrial respiration. Furthermore, in a Huh-7 cell xenograft tumour mouse model, intraperitoneal injection of ascorbate (4.0 g/kg/3 days) but not a lower dose of ascorbate (2.0 g/kg/3 days) significantly inhibited tumour growth. Gene array analysis of HCC tumour tissue from xenograft mice given IP ascorbate (4.0 g/kg/3 days) identified changes in the transcript levels of 192 genes/ncRNAs involved in insulin receptor signalling, metabolism and mitochondrial respiration. Consistent with the array data, gene expression levels of AGER, DGKK, ASB2, TCP10L2, Lnc-ALCAM-3, and Lnc-TGFBR2-1 were increased 2.05-11.35 fold in HCC tumour tissue samples from mice treated with high-dose ascorbate, and IHC staining analysis also verified that AGER/RAGE and DGKK proteins were up-regulated, which implied that AGER/RAGE and DGKK activation might be related to oxidative stress, leading to hepatoma cell death. Conclusions: Our studies identified multiple mechanisms are responsible for the anti-tumour activity of ascorbate and suggest high doses of ascorbate with less frequency will act as a novel therapeutic agent for liver cancer in vivo.
Collapse
|
22
|
Programmed Cell-Death by Ferroptosis: Antioxidants as Mitigators. Int J Mol Sci 2019; 20:ijms20194968. [PMID: 31597407 PMCID: PMC6801403 DOI: 10.3390/ijms20194968] [Citation(s) in RCA: 392] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 12/16/2022] Open
Abstract
Iron, the fourth most abundant element in the Earth's crust, is vital in living organisms because of its diverse ligand-binding and electron-transfer properties. This ability of iron in the redox cycle as a ferrous ion enables it to react with H2O2, in the Fenton reaction, to produce a hydroxyl radical (•OH)-one of the reactive oxygen species (ROS) that cause deleterious oxidative damage to DNA, proteins, and membrane lipids. Ferroptosis is a non-apoptotic regulated cell death that is dependent on iron and reactive oxygen species (ROS) and is characterized by lipid peroxidation. It is triggered when the endogenous antioxidant status of the cell is compromised, leading to lipid ROS accumulation that is toxic and damaging to the membrane structure. Consequently, oxidative stress and the antioxidant levels of the cells are important modulators of lipid peroxidation that induce this novel form of cell death. Remedies capable of averting iron-dependent lipid peroxidation, therefore, are lipophilic antioxidants, including vitamin E, ferrostatin-1 (Fer-1), liproxstatin-1 (Lip-1) and possibly potent bioactive polyphenols. Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. These, as well as other radical trapping antioxidants (RTAs), are discussed in the current review.
Collapse
|
23
|
Therapeutic Perspective of Vitamin C and Its Derivatives. Antioxidants (Basel) 2019; 8:antiox8080247. [PMID: 31357509 PMCID: PMC6721080 DOI: 10.3390/antiox8080247] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
l-Ascorbic acid (ASA), vitamin C, is a ubiquitous carbohydrate-like compound that has an essential role in a number of cellular processes, such as collagen synthesis, cellular oxidation, and various hydroxylation reactions. ASA is a biomolecule of critical importance for protection of cellular components against oxidative damage caused by toxic free radicals and other reactive oxygen species (ROS) that are involved in the development of various types of chronic diseases. Vitamin C has a switchover role from being an antioxidant in physiological conditions to a prooxidant under pathologic conditions. Moreover, some l-ascorbic acid derivatives exhibit strong and selective antitumor and antiviral activity. This review emphasizes the advances on diverse and potent biological profiles of l-ascorbic acid and its derivatives, and their perspective in the development of new bioactive chemical entities in the future. The work is primarily addressed at antioxidant, anticancer, and antiviral potencies of l-ascorbic acid and compounds containing its butenolide structural motif.
Collapse
|
24
|
Saitoh Y. Comments to the article “Artefacts with ascorbate and other redox-active compounds in cell culture: epigenetic modifications, and cell killing due to hydrogen peroxide generation in cell culture media”. Free Radic Res 2018; 52:910-912. [DOI: 10.1080/10715762.2018.1524891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yasukazu Saitoh
- Laboratory of Bioscience and Biotechnology for Cell Function Control, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Japan
| |
Collapse
|
25
|
Posch W, Blatzer M, Wilflingseder D, Lass-Flörl C. Aspergillus terreus: Novel lessons learned on amphotericin B resistance. Med Mycol 2018. [PMID: 29538736 DOI: 10.1093/mmy/myx119] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The polyene antifungal amphotericin B (AmB) exerts a powerful and broad activity against a vast array of fungi and in general displays a remarkably low rate of antimicrobial resistance. Aspergillus terreus holds an exceptional position among the Aspergilli due to its intrinsic AmB resistance, in vivo and in vitro. Until now, the underlying mechanisms of polyene resistance were not well understood. This review will highlight the molecular basis of A. terreus and AmB resistance recently gained and will display novel data on the mode of action of AmB. A main focus is set on fundamental stress response pathways covering the heat shock proteins (Hsp) 90/Hsp70 axis, as well as reactive oxygen species detoxifying enzymes in response to AmB. The effect on main cellular functions such as fungal respiration will be addressed in detail and resistance mechanisms will be highlighted. Based on these novel findings we will discuss new molecular targets for alternative options in the treatment of invasive infections due to A. terreus.
Collapse
Affiliation(s)
- Wilfried Posch
- Medical University of Innsbruck, Division of Hygiene and Medical Microbiology, Schöpfstrasse 41, A- 6020 Innsbruck, Austria
| | - Michael Blatzer
- Medical University of Innsbruck, Division of Hygiene and Medical Microbiology, Schöpfstrasse 41, A- 6020 Innsbruck, Austria
| | - Doris Wilflingseder
- Medical University of Innsbruck, Division of Hygiene and Medical Microbiology, Schöpfstrasse 41, A- 6020 Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Medical University of Innsbruck, Division of Hygiene and Medical Microbiology, Schöpfstrasse 41, A- 6020 Innsbruck, Austria.,ISHAM Aspergillus terreus Working Group
| |
Collapse
|
26
|
Tóth SZ, Lőrincz T, Szarka A. Concentration Does Matter: The Beneficial and Potentially Harmful Effects of Ascorbate in Humans and Plants. Antioxid Redox Signal 2018; 29:1516-1533. [PMID: 28974112 DOI: 10.1089/ars.2017.7125] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Ascorbate (Asc) is an essential compound both in animals and plants, mostly due to its reducing properties, thereby playing a role in scavenging reactive oxygen species (ROS) and acting as a cofactor in various enzymatic reactions. Recent Advances: Growing number of evidence shows that excessive Asc accumulation may have negative effects on cellular functions both in humans and plants; inter alia it may negatively affect signaling mechanisms, cellular redox status, and contribute to the production of ROS via the Fenton reaction. CRITICAL ISSUES Both plants and humans tightly control cellular Asc levels, possibly via biosynthesis, transport, and degradation, to maintain them in an optimum concentration range, which, among other factors, is essential to minimize the potentially harmful effects of Asc. On the contrary, the Fenton reaction induced by a high-dose Asc treatment in humans enables a potential cancer-selective cell death pathway. FUTURE DIRECTIONS The elucidation of Asc induced cancer selective cell death mechanisms may give us a tool to apply Asc in cancer therapy. On the contrary, the regulatory mechanisms controlling cellular Asc levels are also to be considered, for example, when aiming at generating crops with elevated Asc levels.
Collapse
Affiliation(s)
- Szilvia Z Tóth
- 1 Institute of Plant Biology , Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Tamás Lőrincz
- 2 Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics , Budapest, Hungary
| | - András Szarka
- 2 Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics , Budapest, Hungary
| |
Collapse
|
27
|
The Interrelationship of Pharmacologic Ascorbate Induced Cell Death and Ferroptosis. Pathol Oncol Res 2018; 25:669-679. [PMID: 30443843 DOI: 10.1007/s12253-018-0539-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
Pharmacologic ascorbate induced cell death and ferroptosis share common features such as iron dependency, production of ROS, lipid peroxidation, caspase independency and the possible involvement of autophagy. These observations lead us to hypothesize that ferroptosis may also be involved in cancer cell death due to pharmacologic ascorbate treatment. Thus cell death of HT-1080 cell line was induced by ferroptosis inducers and pharmacologic ascorbate then the mechanism of cell death was compared. The EC50 value of pharmacologic ascorbate on HT-1080 cell line was found to be 0.5 mM that is in the range of the most ascorbate sensitive cell lines. However either of the specific inhibitors of ferroptosis (ferrostatin-1 and liproxstatin-1) could not elevate the viability of pharmacologic ascorbate treated cells suggesting that ferroptosis was not involved in the pharmacologic ascorbate induced cell death. α-tocopherol that could effectively elevate the viability of erastin and RSL3 treated HT1080 cells failed to mitigate the cytotoxic effect of pharmacologic ascorbate further strengthened this assumption. Furthermore at lower concentrations (0.1-0.5 mM) ascorbate could avoid the effects of ferroptosis inducers. Our results indicate that pharmacologic ascorbate induced cytotoxicity and ferroptosis - albeit phenotypically they show similar traits - are governed by different mechanisms.
Collapse
|
28
|
de Carvalho Melo-Cavalcante AA, da Rocha Sousa L, Alencar MVOB, de Oliveira Santos JV, da Mata AMO, Paz MFCJ, de Carvalho RM, Nunes NMF, Islam MT, Mendes AN, Gonçalves JCR, da Silva FCC, Ferreira PMP, de Castro E Sousaa JM. Retinol palmitate and ascorbic acid: Role in oncological prevention and therapy. Biomed Pharmacother 2018; 109:1394-1405. [PMID: 30551390 DOI: 10.1016/j.biopha.2018.10.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer development has been directly related to oxidative stress. During chemotherapy, some cancer patients use dietary antioxidants to avoid nutritional deficiencies due to cancer treatment. Among the antioxidants consumed, there are vitamins, including retinyl palmitate (PR) and ascorbic acid (AA), which have the capacity to reduce free radicals formation, protect cellular structures and maintain the cellular homeostasis. This systematic review evaluated the antioxidant and antitumor mechanisms of retinol palmitate (a derivative of vitamin A) and/or ascorbic acid (vitamin C) in cancer-related studies. Ninety-seven (97) indexed articles in the databases PubMed and Science Direct, published between 2013 and 2017, including 23 clinical studies (5 for every single compound while 13 in interaction) and 74 non-clinical studies (37 for retinol palmitate, 36 for ascorbic acid and 1 in interaction) were considered. Antioxidant and antitumor effects, with controversies over dosage and route of administration, were observed for the test compounds in their isolated form or associated in clinical studies. Prevention of cancer risks against oxidative damage was seen in lower doses of retinol palmitate and/or vitamin C. However, at high doses, they can generate reactive oxygen species, cytotoxicity and apoptosis in test systems. Non-clinical studies using cell lines have allowed understanding the mechanisms related to antioxidants and antitumor effects of the isolated compounds, however, studies on vitamin interactions, acting as antioxidants and/or antitumor are still rare and controversial. More studies, mainly related to modulation of antineoplastic drugs are needed for understanding the risks and benefits of their use during treatment in order to achieve effectiveness in cancer therapy and patient's quality of life.
Collapse
Affiliation(s)
- Ana Amélia de Carvalho Melo-Cavalcante
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Leonardo da Rocha Sousa
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Marcus Vinícius Oliveira Barros Alencar
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - José Victor de Oliveira Santos
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Ana Maria Oliveira da Mata
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Márcia Fernanda Correia Jardim Paz
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Ricardo Melo de Carvalho
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Nárcia Mariana Fonseca Nunes
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Muhammad Torequl Islam
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Anderson Nogueira Mendes
- Department of Biophysics and Physiology of Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Juan Carlos Ramos Gonçalves
- Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Felipe Cavalcanti Carneiro da Silva
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Department of Biological Sciences, Federal University of Piauí, Picos, Piauí, 64.067-670, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Department of Biophysics and Physiology of Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - João Marcelo de Castro E Sousaa
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Department of Biological Sciences, Federal University of Piauí, Picos, Piauí, 64.067-670, Brazil.
| |
Collapse
|
29
|
Carr AC, Cook J. Intravenous Vitamin C for Cancer Therapy - Identifying the Current Gaps in Our Knowledge. Front Physiol 2018; 9:1182. [PMID: 30190680 PMCID: PMC6115501 DOI: 10.3389/fphys.2018.01182] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/06/2018] [Indexed: 02/04/2023] Open
Abstract
The use of intravenous vitamin C (IVC) for cancer therapy has long been an area of intense controversy. Despite this, high dose IVC has been administered for decades by complementary health care practitioners and physicians, with little evidence base resulting in inconsistent clinical practice. In this review we pose a series of questions of relevance to both researchers and clinicians, and also patients themselves, in order to identify current gaps in our knowledge. These questions include: Do oncology patients have compromised vitamin C status? Is intravenous the optimal route of vitamin C administration? Is IVC safe? Does IVC interfere with chemotherapy or radiotherapy? Does IVC decrease the toxic side effects of chemotherapy and improve quality of life? What are the relevant mechanisms of action of IVC? What are the optimal doses, frequency, and duration of IVC therapy? Researchers have made massive strides over the last 20 years and have addressed many of these important aspects, such as the best route for administration, safety, interactions with chemotherapy, quality of life, and potential mechanisms of action. However, we still do not know the answers to a number of fundamental questions around best clinical practice, such as how much, how often and for how long to administer IVC to oncology patients. These questions point the way forward for both basic research and future clinical trials.
Collapse
Affiliation(s)
- Anitra C Carr
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - John Cook
- New Brighton Health Care, Christchurch, New Zealand
| |
Collapse
|
30
|
Mei H, Tu H. Vitamin C and Helicobacter pylori Infection: Current Knowledge and Future Prospects. Front Physiol 2018; 9:1103. [PMID: 30154733 PMCID: PMC6102328 DOI: 10.3389/fphys.2018.01103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
The gram-negative bacterium, Helicobacter pylori (H. pylori), infection is predominantly known for its strong association with development of gastric diseases, including gastritis, peptic ulcers, and stomach cancer. Numerous clinical reports show that ascorbic acid deficiency has been connect with gastritis. Vitamin C levels both in gastric acid and serum have constantly been affirmed to be low in subjects with H. pylori infected gastritis and peptic ulcers. Ascorbic acid supplementation likely relates to reduced incidences of bleeding from peptic ulcers and gastric cancer. H. pylori eradication is shown to increase vitamin C levels, while the benefits of ascorbic acid oral intake to increase the effectiveness of H. pylori-eradication therapy are controversial. Recent studies suggest that ascorbate intake intravenously, but not orally; pharmacologic ascorbate concentrations up to 30 mmol/L in blood, several millimolar in tissues as well as in interstitial fluid, are easily and safely achieved. Pharmacologic ascorbate can exert pro-oxidant effects locally as a drug by mediating hydrogen peroxide (H2O2) formation, which was applied to animal and clinical trials of cancer, sepsis, and severe burns etc. In this review, we summarize current understanding of the associations of vitamin C and H. pylori infection, and outline some potential strategies for H. pylori intervention from emerging advances on ascorbic acid physiology and pharmacology.
Collapse
Affiliation(s)
- Haixin Mei
- Department of Gastroenterology, Xinyang Central Hospital, Xinyang, China
| | - Hongbin Tu
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, United States
| |
Collapse
|
31
|
Foster MN, Carr AC, Antony A, Peng S, Fitzpatrick MG. Intravenous Vitamin C Administration Improved Blood Cell Counts and Health-Related Quality of Life of Patient with History of Relapsed Acute Myeloid Leukaemia. Antioxidants (Basel) 2018; 7:antiox7070092. [PMID: 30012948 PMCID: PMC6070822 DOI: 10.3390/antiox7070092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/23/2023] Open
Abstract
A 52-year-old female presented to Integrated Health Options Clinic in October 2014 with a history of relapsed acute myeloid leukaemia (AML, diagnosed in 2009 and relapsed in 2014). Intravenous(IV) vitamin C therapy was initiated (in 2014) following completion of chemotherapy as an alternative to haematopoietic stem cell transplantation. IV vitamin C was administered twice weekly at a dose of 70 g/infusion. Within 4 weeks of initiation of IV vitamin C therapy, there was a dramatic improvement in the patient’s blood indices with platelet cell counts increasing from 25 × 109/L to 196 × 109/L and white blood cell counts increasing from 0.29 × 109/L to 4.0 × 109/L, with further improvements observed over the next 18 months. Furthermore, there was a clear and sustained improvement in the patient’s health-related quality of life scores assessed using a validated questionnaire. She has remained healthy and in complete remission until the present day. This case study highlights the benefits of IV vitamin C as a supportive therapy for previously relapsed AML.
Collapse
Affiliation(s)
- Mike N Foster
- Integrated Health Options Ltd., Auckland 1050, New Zealand.
| | - Anitra C Carr
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand.
| | - Alina Antony
- Feedback Research Ltd., Auckland 1050, New Zealand.
| | - Selene Peng
- Feedback Research Ltd., Auckland 1050, New Zealand.
| | | |
Collapse
|
32
|
Gaikwad S, Chakraborty A, Salwe S, Patel V, Kulkarni S, Banerjee S. Juglone-ascorbic acid synergy inhibits metastasis and induces apoptotic cell death in poorly differentiated thyroid carcinoma by perturbing SOD and catalase activities. J Biochem Mol Toxicol 2018; 32:e22176. [DOI: 10.1002/jbt.22176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Sujay Gaikwad
- Radiation Medicine Centre; Bhabha Atomic Research Center, Parel; Mumbai 400012 India
- Homi Bhabha National Institute, Anushaktinagar; Mumbai India
| | - Avik Chakraborty
- Radiation Medicine Centre; Bhabha Atomic Research Center, Parel; Mumbai 400012 India
- Homi Bhabha National Institute, Anushaktinagar; Mumbai India
| | - Sukeshani Salwe
- National Institute for Research in Reproductive Health; Mumbai India
| | - Vainav Patel
- National Institute for Research in Reproductive Health; Mumbai India
| | - Savita Kulkarni
- Radiation Medicine Centre; Bhabha Atomic Research Center, Parel; Mumbai 400012 India
- Homi Bhabha National Institute, Anushaktinagar; Mumbai India
| | - Sharmila Banerjee
- Radiation Medicine Centre; Bhabha Atomic Research Center, Parel; Mumbai 400012 India
- Homi Bhabha National Institute, Anushaktinagar; Mumbai India
| |
Collapse
|
33
|
No Reported Renal Stones with Intravenous Vitamin C Administration: A Prospective Case Series Study. Antioxidants (Basel) 2018; 7:antiox7050068. [PMID: 29883396 PMCID: PMC5981254 DOI: 10.3390/antiox7050068] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/11/2018] [Accepted: 05/19/2018] [Indexed: 12/12/2022] Open
Abstract
A few cases associating high dose intravenous vitamin C (IVC) administration with renal stone formation have been reported in the literature, however, no long-term studies investigating IVC administration and reported renal stones have been carried out. Our aim was to measure the frequency of reported renal stones in patients receiving IVC therapy. We carried out a prospective case series study of 157 adult patients who commenced IVC therapy at Integrated Health Options clinic between 1 September 2011 and 31 August 2012, with follow-up for 12 months. Inquiries into the occurrence of renal stones were conducted at enrolment, 6 and 12 months, and renal function blood tests were conducted at enrolment, 4 weeks and every 12 weeks thereafter in a subgroup of patients. No renal stones were reported by any patients in the study, despite 8% of the patients having a history of renal stones. In addition, the majority of patients investigated had stable renal function during the study period as evidenced by little change in serum creatinine levels and estimated glomerular filtration rate (eGFR) following IVC. In conclusion, IVC therapy was not associated with patient-reported renal stones. Although not the primary focus of this study, it was also observed that there was no significant change in mean serum creatinine or eGFR for those who had follow-up renal function blood tests.
Collapse
|
34
|
Miura K, Haraguchi M, Ito H, Tai A. Potential Antitumor Activity of 2-O-α-d-Glucopyranosyl-6-O-(2-Pentylheptanoyl)-l-Ascorbic Acid. Int J Mol Sci 2018; 19:ijms19020535. [PMID: 29439410 PMCID: PMC5855757 DOI: 10.3390/ijms19020535] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 01/30/2018] [Accepted: 02/06/2018] [Indexed: 02/07/2023] Open
Abstract
Intravenous administration of high-dose ascorbic acid (AA) has been reported as a treatment for cancer patients. However, cancer patients with renal failure cannot receive this therapy because high-dose AA infusion can have side effects. To solve this problem, we evaluated the antitumor activity of a lipophilic stable AA derivative, 2-O-α-d-glucopyranosyl-6-O-(2-pentylheptanoyl)-l-ascorbic acid (6-bOcta-AA-2G). Intravenous administration of 6-bOcta-AA-2G suppressed tumor growth in colon-26 tumor-bearing mice more strongly than did AA, even at 1/10 of the molar amount of AA. Experiments on the biodistribution and clearance of 6-bOcta-AA-2G and its metabolites in tumor-bearing mice showed that 6-bOcta-AA-2G was hydrolyzed to 6-O-(2-propylpentanoyl)-l-ascorbic acid (6-bOcta-AA) slowly to yield AA, and the results suggested that this characteristic metabolic pattern is responsible for making the antitumor activity of 6-bOcta-AA-2G stronger than that of AA and that the active form of 6-bOcta-AA-2G showing antitumor activity is 6-bOcta-AA. In in vitro experiments, the oxidized form of 6-bOcta-AA as well as 6-bOcta-AA showed significant cytotoxicity, while the oxidized forms of ascorbic acid showed no cytotoxicity at all, suggesting that the antitumor activity mechanism of 6-bOcta-AA-2G is different from that of AA and that the antitumor activity is due to the reduced and oxidized form of 6-bOcta-AA. The findings suggest that 6-bOcta-AA-2G is a potent candidate as an alternative drug to intravenous high-dose AA.
Collapse
Affiliation(s)
- Kaori Miura
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka-cho, Shobara, Hiroshima 727-0023, Japan.
| | - Misaki Haraguchi
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka-cho, Shobara, Hiroshima 727-0023, Japan.
| | - Hideyuki Ito
- Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan.
| | - Akihiro Tai
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka-cho, Shobara, Hiroshima 727-0023, Japan.
| |
Collapse
|
35
|
Polireddy K, Dong R, Reed G, Yu J, Chen P, Williamson S, Violet PC, Pessetto Z, Godwin AK, Fan F, Levine M, Drisko JA, Chen Q. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Sci Rep 2017; 7:17188. [PMID: 29215048 PMCID: PMC5719364 DOI: 10.1038/s41598-017-17568-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is among the most lethal cancers with poorly tolerated treatments. There is increasing interest in using high-dose intravenous ascorbate (IVC) in treating this disease partially because of its low toxicity. IVC bypasses bioavailability barriers of oral ingestion, provides pharmacological concentrations in tissues, and exhibits selective cytotoxic effects in cancer cells through peroxide formation. Here, we further revealed its anti-pancreatic cancer mechanisms and conducted a phase I/IIa study to investigate pharmacokinetic interaction between IVC and gemcitabine. Pharmacological ascorbate induced cell death in pancreatic cancer cells with diverse mutational backgrounds. Pharmacological ascorbate depleted cellular NAD+ preferentially in cancer cells versus normal cells, leading to depletion of ATP and robustly increased α-tubulin acetylation in cancer cells. While ATP depletion led to cell death, over-acetylated tubulin led to inhibition of motility and mitosis. Collagen was increased, and cancer cell epithelial-mesenchymal transition (EMT) was inhibited, accompanied with inhibition in metastasis. IVC was safe in patients and showed the possibility to prolong patient survival. There was no interference to gemcitabine pharmacokinetics by IVC administration. Taken together, these data revealed a multi-targeting mechanism of pharmacological ascorbate's anti-cancer action, with minimal toxicity, and provided guidance to design larger definitive trials testing efficacy of IVC in treating advanced pancreatic cancer.
Collapse
Affiliation(s)
- Kishore Polireddy
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ruochen Dong
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Gregory Reed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Yu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ping Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Stephen Williamson
- Department of Internal Medicine, Hematology and Oncology Division, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Pierre-Christian Violet
- National Institute of Diabetes, Digestive and Kidney Diseases, the National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ziyan Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mark Levine
- National Institute of Diabetes, Digestive and Kidney Diseases, the National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeanne A Drisko
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
36
|
Hurrle S, Hsu WH. The etiology of oxidative stress in insulin resistance. Biomed J 2017; 40:257-262. [PMID: 29179880 PMCID: PMC6138814 DOI: 10.1016/j.bj.2017.06.007] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance is a prevalent syndrome in developed as well as developing countries. It is the predisposing factor for type 2 diabetes mellitus, the most common end stage development of metabolic syndrome in the United States. Previously, studies investigating type 2 diabetes have focused on beta cell dysfunction in the pancreas and insulin resistance, and developing ways to correct these dysfunctions. However, in recent years, there has been a profound interest in the role that oxidative stress in the peripheral tissues plays to induce insulin resistance. The objective of this review is to focus on the mechanism of oxidative species generation and its direct correlation to insulin resistance, to discuss the role of obesity in the pathophysiology of this phenomenon, and to explore the potential of antioxidants as treatments for metabolic dysfunction.
Collapse
Affiliation(s)
- Samantha Hurrle
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Walter H Hsu
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
37
|
Ohwada R, Ozeki Y, Saitoh Y. High-dose ascorbic acid induces carcinostatic effects through hydrogen peroxide and superoxide anion radical generation-induced cell death and growth arrest in human tongue carcinoma cells. Free Radic Res 2017; 51:684-692. [DOI: 10.1080/10715762.2017.1361533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Ryouhei Ohwada
- Laboratory of Bioscience and Biotechnology for Cell Function Control, Department of Life Science, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Japan
| | - Yu Ozeki
- Laboratory of Bioscience and Biotechnology for Cell Function Control, Department of Life Science, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Japan
| | - Yasukazu Saitoh
- Laboratory of Bioscience and Biotechnology for Cell Function Control, Department of Life Science, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Japan
| |
Collapse
|
38
|
Guidarelli A, Cerioni L, Fiorani M, Cantoni O. Intramitochondrial Ascorbic Acid Enhances the Formation of Mitochondrial Superoxide Induced by Peroxynitrite via a Ca 2+-Independent Mechanism. Int J Mol Sci 2017; 18:ijms18081686. [PMID: 28767071 PMCID: PMC5578076 DOI: 10.3390/ijms18081686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/30/2017] [Accepted: 07/30/2017] [Indexed: 01/04/2023] Open
Abstract
Exposure of U937 cells to peroxynitrite promotes mitochondrial superoxide formation via a mechanism dependent on both inhibition of complex III and increased mitochondrial Ca2+ accumulation. Otherwise inactive concentrations of the oxidant produced the same maximal effects in the presence of either complex III inhibitors or agents mobilizing Ca2+ from the ryanodine receptor and enforcing its mitochondrial accumulation. l-Ascorbic acid (AA) produced similar enhancing effects in terms of superoxide formation, DNA strand scission and cytotoxicity. However, AA failed to enhance the intra-mitochondrial concentration of Ca2+ and the effects observed in cells supplemented with peroxinitrite, while insensitive to manipulations preventing the mobilization of Ca2+, or the mitochondrial accumulation of the cation, were also detected in human monocytes and macrophages, which do not express the ryanodine receptor. In all these cell types, mitochondrial permeability transition-dependent toxicity was detected in cells exposed to AA/peroxynitrite and, based on the above criteria, these responses also appeared Ca2+-independent. The enhancing effects of AA are therefore similar to those mediated by bona fide complex III inhibitors, although the vitamin failed to directly inhibit complex III, and in fact enhanced its sensitivity to the inhibitory effects of peroxynitrite.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari Università degli Studi di Urbino "Carlo Bo", 61029 Urbino, Italy.
| | - Liana Cerioni
- Dipartimento di Scienze Biomolecolari Università degli Studi di Urbino "Carlo Bo", 61029 Urbino, Italy.
| | - Mara Fiorani
- Dipartimento di Scienze Biomolecolari Università degli Studi di Urbino "Carlo Bo", 61029 Urbino, Italy.
| | - Orazio Cantoni
- Dipartimento di Scienze Biomolecolari Università degli Studi di Urbino "Carlo Bo", 61029 Urbino, Italy.
| |
Collapse
|
39
|
High Dose Ascorbate Causes Both Genotoxic and Metabolic Stress in Glioma Cells. Antioxidants (Basel) 2017; 6:antiox6030058. [PMID: 28737676 PMCID: PMC5618086 DOI: 10.3390/antiox6030058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/30/2017] [Accepted: 07/19/2017] [Indexed: 01/31/2023] Open
Abstract
We have previously shown that exposure to high dose ascorbate causes double stranded breaks (DSBs) and a build-up in S-phase in glioblastoma (GBM) cell lines. Here we investigated whether or not this was due to genotoxic stress as well as metabolic stress generated by exposure to high dose ascorbate, radiation, ascorbate plus radiation and H₂O₂ in established and primary GBM cell lines. Genotoxic stress was measured as phosphorylation of the variant histone protein, H2AX, 8-oxo-7,8-dihydroguanine (8OH-dG) positive cells and cells with comet tails. Metabolic stress was measured as a decrease in NADH flux, mitochondrial membrane potential (by CMXRos), ATP levels (by ATP luminescence) and mitochondrial superoxide production (by mitoSOX). High dose ascorbate, ascorbate plus radiation, and H₂O₂ treatments induced both genotoxic and metabolic stress. Exposure to high dose ascorbate blocked DNA synthesis in both DNA damaged and undamaged cell of ascorbate sensitive GBM cell lines. H₂O₂ treatment blocked DNA synthesis in all cell lines with and without DNA damage. DNA synthesis arrest in cells with damaged DNA is likely due to both genotoxic and metabolic stress. However, arrest in DNA synthesis in cells with undamaged DNA is likely due to oxidative damage to components of the mitochondrial energy metabolism pathway.
Collapse
|
40
|
Hore TA. Modulating epigenetic memory through vitamins and TET: implications for regenerative medicine and cancer treatment. Epigenomics 2017; 9:863-871. [DOI: 10.2217/epi-2017-0021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Vitamins A and C represent unrelated sets of small molecules that are essential to the human diet and have recently been shown to intensify erasure of epigenetic memory in naive embryonic stem cells. These effects are driven by complementary enhancement of the ten-eleven translocation (TET) demethylases – vitamin A stimulates TET expression, whereas vitamin C potentiates TET catalytic activity. Vitamin A and C cosupplementation synergistically enhances reprogramming of differentiated cells to the naive state, but overuse may exaggerate instability of imprinted genes. As such, optimizing their use in culture media will be important for regenerative medicine and mammalian transgenics. In addition, mechanistic perception of how these vitamins interact with the epigenome may be relevant for understanding cancer and improving patient treatment.
Collapse
Affiliation(s)
- Timothy A Hore
- Department of Anatomy, University of Otago, 270 Great King Street, Dunedin, 9016, New Zealand
| |
Collapse
|
41
|
2- O-α-D-Glucopyranosyl-l-ascorbic acid as an antitumor agent for infusion therapy. Biochem Biophys Rep 2017; 10:232-236. [PMID: 28955751 PMCID: PMC5614674 DOI: 10.1016/j.bbrep.2017.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 11/22/2022] Open
Abstract
Ascorbic acid (AA) has been reported as a treatment for cancer patients. Intravenous (iv) administration of high-dose AA increases plasma AA levels to pharmacologic concentrations and generates reactive oxygen species (ROS) to exert anti-tumor activity via enhancement of oxidative stress. However, AA is very unstable in aqueous solutions and it is impossible to preserve AA for a long period in a solution. 2-O-α-D-Glucopyranosyl-l-ascorbic acid (AA-2G), which is a glucoside derivative of AA, has been found to exhibit much higher stability than AA in aqueous solutions and it shows vitamin C activity after enzymatic hydrolysis to AA. To evaluate the effectiveness of AA-2G for cancer treatment, we examined the antitumor activity of AA-2G to murine colon carcinoma (colon-26) cells and in tumor-bearing mice. AA-2G did not show cytotoxicity to colon-26 cells, whereas AA exhibited a significant cytotoxic effect in a concentration-dependent manner. In colon-26 tumor-bearing mice, iv administration of high-dose AA-2G as well as that of AA significantly inhibited tumor growth. Experiments on the biodistribution and clearance of AA-2G in tumor-bearing mice showed that AA-2G was rapidly hydrolyzed to AA and exhibited significant antitumor activity. Treatment of tumor-bearing mice with AA-2G tended to increase plasma malondialdehyde level. These results indicated that the antitumor activity of AA-2G was caused by ROS generated by AA released by rapid hydrolysis of AA-2G. Tumor growth was inhibited by administration of high-dose ascorbic acid 2-glucoside in vivo. High-dose ascorbic acid 2-glucoside showed oxidative stress-mediated antitumor activity. Rapidly released ascorbic acid gave oxidative stress to tumors. The antitumor activities of ascorbic acid 2-glucoside were the same as those of ascorbic acid. Ascorbic acid 2-glucoside can be used as an agent in infusion therapy for cancer.
Collapse
|
42
|
Affiliation(s)
| | - Mark Levine
- Molecular and Clinical Nutrition Section, NIDDK, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
43
|
Ooko E, Kadioglu O, Greten HJ, Efferth T. Pharmacogenomic Characterization and Isobologram Analysis of the Combination of Ascorbic Acid and Curcumin-Two Main Metabolites of Curcuma longa-in Cancer Cells. Front Pharmacol 2017; 8:38. [PMID: 28210221 PMCID: PMC5288649 DOI: 10.3389/fphar.2017.00038] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/18/2017] [Indexed: 12/11/2022] Open
Abstract
Curcuma longa has long been used in China and India as anti-inflammatory agent to treat a wide variety of conditions and also as a spice for varied curry preparations. The chemoprofile of the Curcuma species exhibits the presence of varied phytochemicals with curcumin being present in all three species but AA only being shown in C. longa. This study explored the effect of a curcumin/AA combination on human cancer cell lines. The curcumin/AA combination was assessed by isobologram analysis using the Loewe additivity drug interaction model. The drug combination showed additive cytotoxicity toward CCRF-CEM and CEM/ADR5000 leukemia cell lines and HCT116p53+/+ and HCT116p53−/− colon cancer cell line, while the glioblastoma cell lines U87MG and U87MG.ΔEGFR showed additive to supra-additive cytotoxicity. Gene expression profiles predicting sensitivity and resistance of tumor cells to induction by curcumin and AA were determined by microarray-based mRNA expressions, COMPARE, and hierarchical cluster analyses. Numerous genes involved in transcription (TFAM, TCERG1, RGS13, C11orf31), apoptosis-regulation (CRADD, CDK7, CDK19, CD81, TOM1) signal transduction (NR1D2, HMGN1, ABCA1, DE4ND4B, TRIM27) DNA repair (TOPBP1, RPA2), mRNA metabolism (RBBP4, HNRNPR, SRSF4, NR2F2, PDK1, TGM2), and transporter genes (ABCA1) correlated with cellular responsiveness to curcumin and ascorbic acid. In conclusion, this study shows the effect of the curcumin/AA combination and identifies several candidate genes that may regulate the response of varied cancer cells to curcumin and AA.
Collapse
Affiliation(s)
- Edna Ooko
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Germany
| | - Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Germany
| | - Henry J Greten
- Heidelberg School of Chinese MedicineHeidelberg, Germany; Abel Salazar Biomedical Sciences Institute, University of PortoPorto, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Germany
| |
Collapse
|
44
|
Mostafavi-Pour Z, Ramezani F, Keshavarzi F, Samadi N. The role of quercetin and vitamin C in Nrf2-dependent oxidative stress production in breast cancer cells. Oncol Lett 2017; 13:1965-1973. [PMID: 28454351 DOI: 10.3892/ol.2017.5619] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 11/04/2016] [Indexed: 12/16/2022] Open
Abstract
The balance between the production and elimination of reactive oxygen species (ROS) is essential in determining whether cells survive or undergo apoptosis. Nuclear factor erythroid 2-related factor 2 (Nrf2) may act as a sensor for electrophilic stress, thus regulating the intracellular antioxidant response. The present study investigated the role of vitamin C (VC) and quercetin (Q) in the induction of Nrf2-mediated oxidative stress in cancer cells. An MTT assay was conducted to examine the anti-proliferative effects of VC and Q. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to determine the messenger RNA (mRNA) and protein expression of Nrf2, respectively. The activity of nicotinamide adenine dinucleotide phosphate dehydrogenase quinone 1, heme oxygenase 1, glutathione peroxidase, glutathione reductase and reduced glutathione were measured by spectrophotometric analysis. Intracellular generation of ROS was determined using 2'-7'-dichlorodihydrofluorescein diacetate fluorescent probes. The results demonstrated that the cytotoxicity (50% inhibitory concentration) of VC and Q were 271.6-480.1 and 155.1-232.9 µM, respectively. Additionally, there was a significant decrease in the expression of Nrf2 mRNA and protein levels following the treatment of breast cancer cells with VC and Q (P=0.024). Following treatment with VC and Q, the nuclear/cytosolic Nrf2 ratio was reduced by 1.7-fold in MDA-MB 231 cells, 2-fold in MDA-MB 468 cells, 1.4-fold in MCF-7 cells and 1.2 fold in A549 cells. Sequential treatment with VC and Q decreased endogenous production of ROS in a dose-dependent manner (P=0.027). The results of the current study suggest that VC and Q treatment may be developed as an adjuvant for patients with cancer and overexpression of Nrf2.
Collapse
Affiliation(s)
- Zohreh Mostafavi-Pour
- Department of Biochemistry, Recombinant Protein Laboratory, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran.,Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Fatemeh Ramezani
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Fatemeh Keshavarzi
- Department of Biochemistry, Recombinant Protein Laboratory, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Nasser Samadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| |
Collapse
|
45
|
Thornthwaite JT, Shah HR, England SR, Roland LH, Thibado SP, Ballard TK, Goodman BT. Anticancer Effects of Curcumin, Artemisinin, Genistein, and Resveratrol, and Vitamin C: Free Versus Liposomal Forms. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/abc.2017.71002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
46
|
Padayatty SJ, Levine M. Vitamin C: the known and the unknown and Goldilocks. Oral Dis 2016; 22:463-93. [PMID: 26808119 PMCID: PMC4959991 DOI: 10.1111/odi.12446] [Citation(s) in RCA: 414] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/08/2016] [Indexed: 12/11/2022]
Abstract
Vitamin C (Ascorbic Acid), the antiscorbutic vitamin, cannot be synthesized by humans and other primates, and has to be obtained from diet. Ascorbic acid is an electron donor and acts as a cofactor for fifteen mammalian enzymes. Two sodium-dependent transporters are specific for ascorbic acid, and its oxidation product dehydroascorbic acid is transported by glucose transporters. Ascorbic acid is differentially accumulated by most tissues and body fluids. Plasma and tissue vitamin C concentrations are dependent on amount consumed, bioavailability, renal excretion, and utilization. To be biologically meaningful or to be clinically relevant, in vitro and in vivo studies of vitamin C actions have to take into account physiologic concentrations of the vitamin. In this paper, we review vitamin C physiology; the many phenomena involving vitamin C where new knowledge has accrued or where understanding remains limited; raise questions about the vitamin that remain to be answered; and explore lines of investigations that are likely to be fruitful.
Collapse
Affiliation(s)
- S J Padayatty
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - M Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Rouleau L, Antony AN, Bisetto S, Newberg A, Doria C, Levine M, Monti DA, Hoek JB. Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: New insights into ascorbate cytotoxicity. Free Radic Biol Med 2016; 95:308-322. [PMID: 27036367 PMCID: PMC4867251 DOI: 10.1016/j.freeradbiomed.2016.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 12/15/2022]
Abstract
We investigated the mechanism of selective ascorbate-induced cytotoxicity in tumor cells, including Hep G2 cells, compared to primary hepatocytes. H2O2 formation was required for ascorbate cytotoxicity, as extracellular catalase treatment protected tumor cells. H2O2 generated by glucose oxidase treatment also caused cell killing, but treatment with a pharmacologic dose (5-20mM) of ascorbate was significantly more cytotoxic at comparable rates of H2O2 production, suggesting that ascorbate enhanced H2O2 cytotoxicity. This was further supported by the finding that ascorbate at a non-cytotoxic dose (1mM) enhanced cell killing caused by glucose oxidase. Consistent with this conclusion, ascorbate treatment caused deregulation of cellular calcium homeostasis, resulting in massive mitochondrial calcium accumulation. Ascorbate acted synergistically with the chemotherapeutic sorafenib in killing Hep G2 cells, but not primary hepatocytes, suggesting adjuvant ascorbate treatment can broaden sorafenib's therapeutic range. Sorafenib caused mitochondrial depolarization and prevented mitochondrial calcium sequestration. Subsequent ascorbate addition further deregulated cellular calcium homeostasis promoting cell death. Additionally, we present the case of a patient with hepatocellular carcinoma (HCC) who had prolonged regression of a rib metastasis upon combination treatment with ascorbate and sorafenib, indicating that these studies have direct clinical relevance.
Collapse
Affiliation(s)
- Lauren Rouleau
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anil Noronha Antony
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sara Bisetto
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew Newberg
- Jefferson-Myrna Brind Center of Integrative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Cataldo Doria
- Division of Transplantation, Liver Tumor Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Daniel A Monti
- Jefferson-Myrna Brind Center of Integrative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Jan B Hoek
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
48
|
Frajese GV, Benvenuto M, Fantini M, Ambrosin E, Sacchetti P, Masuelli L, Giganti MG, Modesti A, Bei R. Potassium increases the antitumor effects of ascorbic acid in breast cancer cell lines in vitro. Oncol Lett 2016; 11:4224-4234. [PMID: 27313770 DOI: 10.3892/ol.2016.4506] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 02/08/2016] [Indexed: 12/17/2022] Open
Abstract
Ascorbic acid (A) has been demonstrated to exhibit anti-cancer activity in association with chemotherapeutic agents. Potassium (K) is a regulator of cellular proliferation. In the present study, the biological effects of A and K bicarbonate, alone or in combination (A+K), on breast cancer cell lines were evaluated. The survival of cancer cells was determined by sulforhodamine B cell proliferation assay, while analysis of the cell cycle distribution was conducted via fluorescence-activated cell sorting. In addition, the expression of signaling proteins was analyzed upon treatment. The results indicated that there was a heterogeneous response of the different cell lines to A and K, and the best effects were achieved by A+K and A treatment. The interaction between A+K indicated an additive or synergistic effect. In addition, A+K increased the percentage of cells in the sub-G1 phase of the cell cycle, and was the most effective treatment in activating the degradation of poly(adenosine diphosphate-ribose) polymerase-1. In the breast cancer cell line MCF-7, A+K induced the appearance of the 18 kDa isoform of B-cell lymphoma-2-associated X protein (Bax), which is a more potent inducer of apoptosis than the full-length Bax-p21. The effects of A and K on the phosphorylation of extracellular signal-regulated kinase (ERK)1 and ERK2 were heterogeneous. In addition, treatment with K, A and A+K inhibited the expression of nuclear factor-κB. Overall, the results of the present study indicated that K potentiated the anti-tumoral effects of A in breast cancer cells in vitro.
Collapse
Affiliation(s)
- Giovanni Vanni Frajese
- Department of Sports Science, Human and Health, University of Rome 'Foro Italico', Rome I-00135, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata', Rome I-00133, Italy
| | - Massimo Fantini
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata', Rome I-00133, Italy
| | - Elena Ambrosin
- Department of Sports Science, Human and Health, University of Rome 'Foro Italico', Rome I-00135, Italy
| | - Pamela Sacchetti
- Department of Experimental Medicine, University of Rome 'Sapienza', Rome I-00185, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome 'Sapienza', Rome I-00185, Italy
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata', Rome I-00133, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata', Rome I-00133, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata', Rome I-00133, Italy
| |
Collapse
|
49
|
Abstract
The prognosis for patients diagnosed with pancreatic cancer remains dismal, with less than 3% survival at 5 years. Recent studies have demonstrated that high-dose, intravenous pharmacological ascorbate (ascorbic acid, vitamin C) induces cytotoxicity and oxidative stress selectively in pancreatic cancer cells vs. normal cells, suggesting a promising new role of ascorbate as a therapeutic agent. At physiologic concentrations, ascorbate functions as a reducing agent and antioxidant. However, when pharmacological ascorbate is given intravenously, it is possible to achieve millimolar plasma concentration. At these pharmacological levels, and in the presence of catalytic metal ions, ascorbate can induce oxidative stress through the generation of hydrogen peroxide (H2O2). Recent in vitro and in vivo studies have demonstrated ascorbate oxidation occurs extracellularly, generating H2O2 flux into cells resulting in oxidative stress. Pharmacologic ascorbate also inhibits the growth of pancreatic tumor xenografts and displays synergistic cytotoxic effects when combined with gemcitabine in pancreatic cancer. Phase I trials of pharmacological ascorbate in pancreatic cancer patients have demonstrated safety and potential efficacy. In this chapter, we will review the mechanism of ascorbate-induced cytotoxicity, examine the use of pharmacological ascorbate in treatment and assess the current data supporting its potential as an adjuvant in pancreatic cancer.
Collapse
Affiliation(s)
| | - Joseph J Cullen
- 1528 JCP, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA.
| |
Collapse
|
50
|
Abstract
Here, I address the topic of suitability for redox research of common settings in cell cultures. This is done through the prism of in vitro anticancer effects of vitamin C. Cell culture media show lower concentrations of iron and a higher level of oxygen compared to interstitial fluid. Such a setup promotes ascorbate-mediated production and accumulation of hydrogen peroxide, which efficiently kills a variety of cancer cell lines. However, the anticancer effects are annihilated if the iron level is corrected to mimic in vivo concentrations. It appears that the potential benefits of application of vitamin C in cancer treatment have been significantly overestimated. This might be true for other pro-oxidative agents as well, such as some (poly)phenols. We urgently need to establish medium formula and culture maintenance settings that are optimal for redox research.
Collapse
Affiliation(s)
- Ivan Spasojević
- a Department of Life Sciences, Institute for Multidisciplinary Research , University of Belgrade , Belgrade, Kneza Višeslava 1, Serbia
| |
Collapse
|