225
Views
28
CrossRef citations to date
0
Altmetric
Review

Prospects for the introduction of targeted antioxidant drugs for the prevention and treatment of diseases related to free radical pathology

&
Pages 593-603 | Received 16 Feb 2019, Accepted 09 Jun 2019, Published online: 21 Jun 2019

References

  • Denisov ET, Afanas’ev IB. Oxidation and antioxidants in organic chemistry and biology. Boca Raton: CRC Press, Taylor and Francis group; 2005.
  • Duracková Z. Some current insights into oxidative stress. Physiol Res. 2010;59:459–469.
  • Forman HJ. Redox signaling: an evolution from free radicals to aging. Free Radic Biol Med. 2016;97:398–407.
  • Pagano G, Talamanca AA, Castello G, et al. Oxidative stress and mitochondrial dysfunction across broad-ranging pathologies: toward mitochondria-targeted clinical strategies. Oxid Med Cell Longev. 2014;2014:541230.
  • Vyas S, Zaganjor E, Haigis MC. Mitochondria and cancer. Cell. 2016;166:555–566.
  • Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ. Potential clinical applications of chelating drugs in diseases targeting transferrin-bound iron and other metals. Expert Opin Investig Drugs. 2013;22:591–618.
  • Gutteridge JM. Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clin Chem. 1995;41:1819–1828.
  • Stadtman ER, Levine RL. Free radical-mediated oxidation of free amino acids and amino acid residues in proteins. Amino Acids. 2003;25:207–218.
  • Feng H, Stockwell BR. Unsolved mysteries: how does lipid peroxidation cause ferroptosis?. PLoS Biol. 2018;16:e2006203.
  • Hao S, Liang B, Huang Q, et al. Metabolic networks in ferroptosis. Oncol Lett. 2018;15:5405–5411.
  • Kontoghiorghe CN, Andreou N, Constantinou K, et al. World health dilemmas: orphan and rare diseases, orphan drugs and orphan patients. World J Methodol. 2014;4:163–188.
  • Kontoghiorghes GJ, Efstathiou A, Kleanthous M, et al. Risk/benefit assessment, advantages over other drugs and targeting methods in the use of deferiprone as a pharmaceutical antioxidant in iron loading and non iron loading conditions. Hemoglobin. 2009;33:386–397.
  • Marant-Micallef C, Shield KD, Vignat J, et al. The risk of cancer attributable to diagnostic medical radiation: estimation for France in 2015. Int J Cancer. 2019;144:2954–2963.
  • Ichikawa Y, Ghanefar M, Bayeva M, et al. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest. 2014;124:617–630.
  • Park KC, Fouani L, Jansson PJ, et al. Copper and conquer: copper complexes of di-2-pyridylketone thiosemicarbazones as novel anti-cancer therapeutics. Metallomics. 2016;8:874–886.
  • Du J, Cullen JJ, Buettner GR. Ascorbic acid: chemistry, biology and the treatment of cancer. Biochim Biophys Acta. 2012;1826:443–457.
  • Li H, Xia Z, Chen Y, et al. Mechanism and therapies of oxidative stress-mediated cell death in ischemia reperfusion injury. Oxid Med Cell Longev. 2018;2018:2910643.
  • Dubois RN, Abramson SB, Crofford L, et al. Cyclooxygenase in biology and disease. Faseb J. 1998;12:1063–1073.
  • Villeneuve JP, Pichette V. Cytochrome P450 and liver diseases. Curr Drug Metab. 2004;5:273–282.
  • Kolnagou A, Natsiopoulos K, Kleanthous M, et al. Liver iron and serum ferritin levels are misleading for estimating cardiac, pancreatic, splenic and total body iron load in thalassemia patients: factors influencing the heterogenic distribution of excess storage iron in organs as identified by MRI T2*. Toxicol Mech Methods. 2013;23:48–56.
  • Wallis LI, Paley MN, Graham JM, et al. MRI assessment of basal ganglia iron deposition in Parkinson’s disease. J Magn Reson Imaging. 2008;28:1061–1067.
  • Brar S, Henderson D, Schenck J, et al. Iron accumulation in the substantia nigra of patients with Alzheimer disease and Parkinsonism. Arch Neurol. 2009;66:371–374.
  • Waldvogel D, van Gelderen P, Hallett M. Increased iron in the dentate nucleus of patients with Friedrich’s ataxia. Ann Neurol. 1999;46:123–125.
  • Kolnagou A, Kontoghiorghe CN, Kontoghiorghes GJ. Transition of Thalassaemia and Friedreich ataxia from fatal to chronic diseases. World J Methodol. 2014;4:197–218.
  • Kontoghiorghes GJ. Prospects for introducing deferiprone as potent pharmaceutical antioxidant. Front Biosci (Elite Ed). 2009;1:161–178.
  • Kontoghiorghes GJ, Kolnagou A, Peng CT, et al. Safety issues of iron chelation therapy in patients with normal range iron stores including thalassaemia, neurodegenerative, renal and infectious diseases. Expert Opin Drug Saf. 2010;9:201–206.
  • Robertson A, Tenenbein M. Hepatotoxicity in acute iron poisoning. Hum Exp Toxicol. 2005;24:559–562.
  • Baranwal AK, Singhi SC. Acute iron poisoning: management guidelines. Indian Pediatr. 2003;40:534–540.
  • Kontoghiorghes GJ, Pattichis K, Neocleous K, et al. The design and development of deferiprone (L1) and other iron chelators for clinical use: targeting methods and application prospects. Curr Med Chem. 2004;11:2161–2183.
  • Fukuda S, Ikeda M, Anzai K, et al. Radiation protection by deferiprone in animal models. Hemoglobin. 2006;30:201–208.
  • Flora SJ, Mittal M, Mehta A. Heavy metal induced oxidative stress & its possible reversal by chelation therapy. Indian J Med Res. 2008;128:501–523.
  • Kolnagou A, Kleanthous M, Kontoghiorghes GJ. Efficacy, compliance and toxicity factors are affecting the rate of normalization of body iron stores in thalassemia patients using the deferiprone and deferoxamine combination therapy. Hemoglobin. 2011;35:186–198.
  • Kontoghiorghes GJ, Eracleous E, Economides C, et al. Advances in iron overload therapies. Prospects for effective use of deferiprone (L1), deferoxamine, the new experimental chelators ICL670, GT56-252, L1NAll and their combination. Curr Med Chem. 2005;12:2663–2681.
  • Kontoghiorghes GJ, Jackson MJ, Lunec J. In vitro screening of iron chelators using models of free radical damage. Free Radic Res Commun. 1986;2:115–124.
  • Konstantinou E, Pashalidis I, Kolnagou A, et al. Interactions of hydroxycarbamide (hydroxyurea) with iron and copper: implications on toxicity and therapeutic strategies. Hemoglobin. 2011;35:237–246.
  • Kontoghiorghes GJ, Neocleous K, Kolnagou A. Benefits and risks of deferiprone in iron overload in thalassaemia and other conditions. Drug Saf. 2003;26:553–584.
  • Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ. Antioxidant targeting by deferiprone in diseases related to oxidative damage. Front Biosci (Landmark Ed). 2014;19:862–885.
  • Rajapurkar MM, Hegde U, Bhattacharya A, et al. Effect of deferiprone, an oral iron chelator, in diabetic and non-diabetic glomerular disease. Toxicol Mech Methods. 2013;23:5–10.
  • Ranjan A, Kalita J, Kumar V, et al. MRI and oxidative stress markers in neurological worsening of Wilson disease following penicillamine. Neurotoxicology. 2015;49:45–49.
  • Tegoni M, Valensin D, Toso L, et al. Copper chelators: chemical properties and bio-medical applications. Curr Med Chem. 2014;21:3785–3818.
  • Li WJ, Chen C, You ZF, et al. Current drug managements of Wilson’s disease: from West to East. Curr Neuropharmacol. 2016;14:322–325.
  • Miki M. Penicillamine as antioxidant. Methods Enzymol. 1994;234:542–547.
  • Cooper GJ. Selective divalent copper chelation for the treatment of diabetes mellitus. Curr Med Chem. 2012;19:2828–2860.
  • Born T, Kontoghiorghe CN, Spyrou A, et al. EDTA chelation reappraisal following new clinical trials and regular use in millions of patients: review of preliminary findings and risk/benefit assessment. Toxicol Mech Methods. 2013;23:11–17.
  • Guldager B, Jørgensen PJ, Grandjean P. Metal excretion and magnesium retention in patients with intermittent claudication treated with intravenous disodium EDTA. Clin Chem. 1996;42:1938–1942.
  • Pippard MJ, Jackson MJ, Hoffman K, et al. Iron chelation using subcutaneous infusions of diethylene triamine penta-acetic acid (DTPA). Scand J Haematol. 1986;36:466–472.
  • Koning J, Palmer P, Franks CR, et al. Cardioxane–ICRF-187 towards anticancer drug specificity through selective toxicity reduction. Cancer Treat Rev. 1991;18:1–19.
  • Speyer JL, Green MD, Zeleniuch-Jacquotte A, et al. ICRF-187 permits longer treatment with doxorubicin in women with breast cancer. J Clin Oncol. 1992;10:117–127.
  • Barnabé N, Zastre JA, Venkataram S, et al. Deferiprone protects against doxorubicin-induced myocyte cytotoxicity. Free Radic Biol Med. 2002;33:266–275.
  • Olsson B, Johansson M, Gabrielsson J, et al. Pharmacokinetics and bioavailability of reduced and oxidized N-acetylcysteine. Eur J Clin Pharmacol. 1988;34:77–82.
  • Bailey GP, Wood DM, Archer JR, et al. An assessment of the variation in the concentration of acetylcysteine in infusions for the treatment of paracetamol overdose. Br J Clin Pharmacol. 2017;83:393–399.
  • Clark RSB, Empey PE, Bayır H, et al. Phase I randomized clinical trial of N-acetylcysteine in combination with an adjuvant probenecid for treatment of severe traumatic brain injury in children. PLoS One. 2017;12:e0180280.
  • Conus P, Seidman LJ, Fournier M, et al. N-acetylcysteine in a double-blind randomized placebo-controlled trial: toward biomarker-guided treatment in early psychosis. Schizophr Bull. 2018;44:317–327.
  • Modarresi A, Ziaie S, Salamzadeh J, et al. Study of The Effects of N-Acetylcysteine on Oxidative Stress Status of Patients on Maintenance-Hemodialysis Undergoing Cadaveric Kidney Transplantation. Iran J Pharm Res. 2017;16:1631–1638.
  • Zhang Y, Ding S, Li C. Effects of N-acetylcysteine treatment in acute respiratory distress syndrome: A meta-analysis. Exp Ther Med. 2017;14:2863–2868.
  • Tenorio MB, Ferreira RC, Moura FA, et al. Oral antioxidant therapy for prevention and treatment of preeclampsia: meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis. 2018;28:865–876.
  • Sins JWR, Fijnvandraat K, Rijneveld AW, et al. Effect of N-acetylcysteine on pain in daily life in patients with sickle cell disease: a randomised clinical trial. Br J Haematol. 2018;182:444–448.
  • Ozdemir ZC, Koc A, Aycicek A, et al. N-Acetylcysteine supplementation reduces oxidative stress and DNA damage in children with β-thalassemia. Hemoglobin. 2014;38:359–364.
  • Amen F, Machin A, Touriño C, et al. N-acetylcysteine improves the quality of red blood cells stored for transfusion. Arch Biochem Biophys. 2017;621:31–37.
  • Soleimani A, Habibi MR, Hasanzadeh Kiabi F, et al. The effect of intravenous N-acetylcysteine on prevention of atrial fibrillation after coronary artery bypass graft surgery: a double-blind, randomised, placebo-controlled trial. Kardiol Pol. 2018;76:99–106.
  • Sharafkhah M, Abdolrazaghnejad A, Zarinfar N, et al. Safety and efficacy of N-acetyl-cysteine for prophylaxis of ventilator-associated pneumonia: a randomized, double blind, placebo-controlled clinical trial. Med Gas Res. 2018;8:19–23.
  • Mantzarlis K, Tsolaki V, Zakynthinos E. Role of Oxidative Stress and Mitochondrial Dysfunction in Sepsis and Potential Therapies. Oxid Med Cell Longev. 2017;2017:5985209.
  • van Zandwijk N, Dalesio O, Pastorino U, et al. EUROSCAN, a randomized trial of vitamin A and N-acetylcysteine in patients with head and neck cancer or lung cancer. For the EUropean organization for research and treatment of cancer head and neck and lung cancer cooperative groups. J Natl Cancer Inst. 2000;92:977–986.
  • Cassidy PB, Liu T, Florell SR, et al. A Phase II Randomized Placebo-Controlled Trial of Oral N-acetylcysteine for Protection of Melanocytic Nevi against UV-Induced Oxidative Stress In Vivo. Cancer Prev Res (Phila). 2017;10:36–44.
  • Posadzki P, Lee MS, Onakpoya I, et al. Dietary supplements and prostate cancer: a systematic review of double-blind, placebo-controlled randomised clinical trials. Maturitas. 2013;75:125–130.
  • Monti D, Sotgia F, Whitaker-Menezes D, et al. Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-Acetylcysteine in Breast Cancer. Semin Oncol. 2017;44:226–232.
  • Tsang RY, Al-Fayea T, Au HJ. Cisplatin overdose: toxicities and management. Drug Saf. 2009;32:1109–1122.
  • Cloos J, Bongers V, Lubsen H, et al. Lack of effect of daily N-acetylcysteine supplementation on mutagen sensitivity. Cancer Epidemiol Biomarkers Prev. 1996;5:941–944.
  • Block KI, Koch AC, Mead MN, et al. Impact of antioxidant supplementation on chemotherapeutic toxicity: a systematic review of the evidence from randomized controlled trials. Int J Cancer. 2008;123:1227–1239.
  • Kontoghiorghes GJ The design of orally active iron chelators for the treatment of thalassaemia. PhD thesis, Colchester UK,University of Essex, British Library Microfilm No D66194/86. 1982, pp 1–243. (https://www.pri.ac.cy/files/KGJ_thesis_1982.pdf)
  • Kontoghiorghes GJ, Bartlett AN, Hoffbrand AV, et al. Long-term trial with the oral iron chelator 1,2-dimethyl-3-hydroxypyrid-4-one (L1). I. Iron chelation and metabolic studies. Br J Haematol. 1990;76:295–300.
  • Boddaert N, Le Quan Sang KH, Rotig A, et al. Selective iron chelation in Friedreich ataxia: biologic and clinical implications. Blood. 2007;110:401–408.
  • Martin-Bastida A, Ward RJ, Newbould R, et al. Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease. Sci Rep. 2017;7:1398.
  • Zorzi G, Zibordi F, Chiapparini L, et al. Iron-related MRI images in patients with pantothenate kinase-associated neurodegeneration (PKAN) treated with deferiprone: results of a phase II pilot trial. Mov Disord. 2011;26:1756–1759.
  • Forni GL, Balocco M, Cremonesi L, et al. Regression of symptoms after selective iron chelation therapy in a case of neurodegeneration with brain iron accumulation. Mov Disord. 2008;23:904–907.
  • Rohani M, Razmeh S, Shahidi GA, et al. A pilot trial of deferiprone in pantothenate kinase-associated neurodegeneration patients. Neurol Int. 2018;9:7279.
  • Mohanty D, Ghosh K, Pathare AV, et al. Deferiprone (L1) as an adjuvant therapy for Plasmodium falciparum malaria.Indian. J Med Res. 2002;115:17–21.
  • Saxena D, Spino M, Tricta F, et al. Drug-Based Lead Discovery: the Novel Ablative Antiretroviral Profile of Deferiprone in HIV-1-Infected Cells and in HIV-Infected Treatment-Naive Subjects of a Double-Blind, Placebo-Controlled, Randomized Exploratory Trial. PLoS One. 2016;11:e0154842.
  • Leftin A, Zhao H, Turkekul M, et al. Iron deposition is associated with differential macrophage infiltration and therapeutic response to iron chelation in prostate cancer. Sci Rep. 2017;7:11632.
  • Ueda K, Kim HJ, Zhao J, et al. Iron promotes oxidative cell death caused by bisretinoids of retina. Proc Natl Acad Sci U S A. 2018;115:4963–4968.
  • Chan S, Lian Q, Chen MP, et al. Deferiprone inhibits iron overload-induced tissue factor bearing endothelial microparticle generation by inhibition oxidative stress induced mitochondrial injury, and apoptosis. Toxicol Appl Pharmacol. 2018;338:148–158.
  • Nielsen TK, Højgaard M, Andersen JT, et al. Elimination of ascorbic acid after high-dose infusion in prostate cancer patients: a pharmacokinetic evaluation. Basic Clin Pharmacol Toxicol. 2015;116:343–348.
  • Mikirova N, Casciari J, Rogers A, et al. Effect of high-dose intravenous vitamin C on inflammation in cancer patients. J Transl Med. 2012;10:189.
  • Vance TM, Su J, Fontham ET, et al. Dietary antioxidants and prostate cancer: a review. Nutr Cancer. 2013;65:793–801.
  • Wang L, Sesso HD, Glynn RJ, et al. Vitamin E and C supplementation and risk of cancer in men: posttrial follow-up in the Physicians’ Health Study II randomized trial. Am J Clin Nutr. 2014;100:915–923.
  • Abdoulhossein D, Taheri I, Saba MA, et al. Effect of vitamin C and vitamin E on lung contusion: A randomized clinical trial study. Ann Med Surg (Lond). 2018;36:152–157.
  • Lamas GA, Goertz C, Boineau R, et al. Effect of disodium EDTA chelation regimen on cardiovascular events in patients with previous myocardial infarction: the TACT randomized trial. JAMA. 2013;309:1241–1250.
  • Lamas GA, Navas-Acien A, Mark DB, et al. Heavy metals, cardiovascular disease, and the unexpected benefits of chelation therapy. J Am Coll Cardiol. 2016;67:2411–2418.
  • Lamas GA, Boineau R, Goertz C, et al. EDTA chelation therapy alone and in combination with oral high-dose multivitamins and minerals for coronary disease: the factorial group results of the trial to assess chelation therapy. Am Heart J. 2014;168:37–44.e5.
  • Buyse GM, Voit T, Schara U, et al. Efficacy of idebenone on respiratory function in patients with Duchenne muscular dystrophy not using glucocorticoids (DELOS): a double-blind randomised placebo-controlled phase 3 trial. Lancet. 2015;385(9979):1748–1757.
  • Tauskela JS. MitoQ-a mitochondria-targeted antioxidant. IDrugs. 2007;10:399–412.
  • Eybl V, Caisová D, Koutenský J, et al. Influence of iron chelators, 1,2-dialkyl-3-hydroxypyridin-4-ones, on the lipid peroxidation and glutathione level in the liver of mice. Arch Toxicol Suppl. 1991;14:185–187.
  • Maher P, Kontoghiorghes GJ. Characterization of the neuroprotective potential of derivatives of the iron chelating drug deferiprone. Neurochem Res. 2015;40:609–620.
  • Mostert LJ, Van Dorst JA, Koster JF, et al. Free radical and cytotoxic effects of chelators and their iron complexes in the hepatocyte. Free Radic Res Commun. 1987;3:379–388.
  • Kontoghiorghe CN, Kolnagou A, Kontoghiorghes GJ. Phytochelators intended for clinical use in iron overload, other diseases of iron imbalance and free radical pathology. Molecules. 2015;20:20841–20872.
  • Kontoghiorghes GJ. A new era in iron chelation therapy: the design of optimal, individually adjusted iron chelation therapies for the complete removal of iron overload in thalassemia and other chronically transfused patients. Hemoglobin. 2009;33:332–338.
  • Hood E, Simone E, Wattamwar P, et al. Nanocarriers for vascular delivery of antioxidants. Nanomedicine (Lond). 2011;6:1257–1272.
  • Hood ED, Chorny M, Greineder CF, et al. Endothelial targeting of nanocarriers loaded with antioxidant enzymes for protection against vascular oxidative stress and inflammation. Biomaterials. 2014;35:3708–3715.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.