Review
BibTex RIS Cite

Demir Fazlalığı İçin Şelasyon Tedavisi ve Şelasyon Ligandları

Year 2019, Volume: 28 Issue: 1, 66 - 81, 31.03.2019

Abstract

Demir fazlalığı büyük ölçüde şelatlama ajanları kullanılarak önlenebilen, vücudun farklı bölgelerinde demirin fazla şekilde depolanma durumudur.  İnsan vücudu demir fazlalığını gidermek için fizyolojik bir mekanizmaya sahip değildir. Demir fazlalığının tedavisi için şu an uygulanan yöntem şelasyon tedavisidir. Demir şelasyon tedavisi, potansiyel olarak demir fazlalığı hastalığının tedavisinde demire bağlanan ligandların kullanımını içerir. Bu derlemede insanda demir fazlalığı oluşma nedenleri ve oluşması durumunda kullanılan şelatörler ve klinikte deneme aşamasında olan şelatörler ile ilgili son gelişmeleri bilgi vererek aktarmaya çalıştık.

References

  • Flora SJS, Pachauri V. Chelation in metal intoxication. Int J Environ Res Public Health, 2010;7(7):2745-2788.
  • Hentze MW, Muckenthaler MU, Andrews NC. Balancing acts: molecular control of mammalianiron metabolism. Cell, 2004; 117:285-297.
  • Richardson DR. Iron chelators as therapeutic agents for the treatment of cancer. Crit Rev Oncol Hematology. 2002; 42:267-281.
  • Winter WE, Bazydlo LA, Harris NS. The moleculer biology of human iron metabolism. Laboratory Medicine, 2014; 45(2): 92-102
  • Torti SV, Torti FM. Iron and cancer: more ore to be mined. Cancer, 2013; 13:342-355
  • Chen JJ, Luo ZD, Zhao ZN, Xie LN, Zheng WJ, Chen TF. Cellular localization of iron(II) polypyridyl complexes determines their anticancer action mechanisms. Biomaterials, 2015; 71:168-177.
  • Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005;57:547-583.
  • Crisponi G, Remelli M. Iron chelating agents for the treatment of iron overload. Coord. Chem. Rev. 2008; 252(10):1225-1240.
  • Angelucci E, Brittenham GM, McLaren CE. Hepatic iron concentration and total body iron stores in Thalassemia Major. N Engl. J Med. 2000; 343:327-331.Olivieri NF. The β thalassemias. N Engl J Med., 1999; 341(2): 99-109.
  • Hershko C, Konijin AM, Link G. Iron chelators for thalasaemia. Br J Haematol. 1998; 101:399-406.
  • Faa G. Crisponi G. Iron chelating agents in clinical practice. Coordination Chemistry; 1999; 184(1):291-310.
  • Liu ZD, Hider RC. Design of clinically usefull iron (III)-selective chelators. Med.Res Rev. 2002;22:26-64.
  • Whetherall DJ. Pathophysiology of thalassemia. Bailliére’s Clinical Hematology. 1998; 11(1): 127-146.
  • Ganz T. Hepcidin in iron metabolism. Current Opinion in Hemayology. 2004;11:251-254.
  • Feng M, Does LV, Bantjees A. Iron(III) chelating resins. VI. Stability constants of iron(III)-ligand complexes on insoluble polymeric matrices. Journal of Applied polymer Science, 1994; 56, 1231-1237.
  • Ellis J. Neufeld oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood, 2006; 107: 3436-3441.
  • Faa G. Crisponi G. Iron chelating agents in clinical practice. Coordination Chemistry Reviews.1999; 184:291-310.
  • Beutler E. Hemochromatosis: genetics and pathophysiology. Annu Rev Med., 2006;57:331-47.
  • Weinberg ED. Iron loading and disease surveillance. Emerg Infect Dis., 1999;5(3):346-52.
  • Gattermann N, Rachmilewitz EA. Iron overload in MDS-pathophysiology, diagnosis, and complications. Ann Hematol. 2011;90(1):1–10.
  • Cabantchik ZI, Breuer W, Zanninelli G, Cianciulli P. LPI-labile plasma iron in iron overload. Best Pract Res Clin Haematol. 2005;18(2):277–287.
  • Brittenham GM. Iron-chelating therapy for transfusional iron overload. N Engl J Med. 2011;364(2):146–156.
  • Musallam KM, Cappellini MD, Wood JC. Elevated liver iron concentration is a marker of increased morbidity in patients with β thalassemia intermedia. Haematologica., 2011;96(11):1605–1612.
  • Corbett JV. Accidental poisoning with iron supplements. MCN: Am J Maternal/Child Nursing 1995; 20:234.
  • Taher A, Nathan D, Porter J. Evaluation of Iron Levels to Avoid the Clinical Sequelae of Iron Overload. Semin Hematol., 2007; 44:S2-S6.
  • Yen AW, Fancher TL, Bowlus CL. Revisiting hereditary hemochromatosis: current concepts and progress. Am J Med., 2006;119:391–9.
  • Kohgo Y. Ikuta K. Ohtake T. Torimoto Y. Kato J. Body iron metabolism and pathophysiology of iron overload. Int J Hematol., 2008; 88:7–15.
  • Feder JN. ve ark. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet., 1996; 13(4): 399-408.
  • Franchini M. Hereditary iron overload: update on pathophysiology, diagnosis, and treatment. Am J Hematol., 2006;81:202– 9.
  • Bonkovsky HL, Lambrecht RW, Shan Y. Iron as a co-morbid factor in nonhemochromatotic liver disease. Alcohol. 2003; 30:137–44.
  • Pietrangelo A. Hereditary hemochromatosis–a new look at an old disease. N Engl J Med., 2004;350:2383–2397.
  • Siddique A. Kowdley KV. Review article: the iron overload syndromes. Alimentary pharmacology & therapeutics. 2012; 35: 876‐893.
  • Erişim:http://www.lifeextension.com/protocols/metabolic-health/hemochromatosis/page-06?p=1 2017. Erişim tarihi: 05.09.2016.
  • Borgaonkar MR. Hemochromatosis.More common than you think. Can Fam Physician., 2003;49:36-43.
  • Gregory A. Hayflick SJ. Genetics of neurodegeneration with brain iron accumulation. CurrNeurolNeurosci Rep. 2011;11(3):254-261.
  • Sebastiani G. Walker AP. HFE gene in primary and secondary hepatic iron overload. World J Gastroenterol. 2007;21;13(35):4673-89.
  • Heli H, Mirtorabi S, Karimian K. Advances in iron chelation: an update. Expert OpinTher Pat. 2011;21(6):819-856.
  • Brissot P, Ropert M, Le Lan C, Loreal O. Non-transferrin bound iron: A key role in iron overload and iron toxicity. Biochim Biophys Acta,. 2012;1820(3):403-410.
  • Munoz M, Garcia-Erce JA, Remacha AF. Disorders of iron metabolism. Part II: iron deficiency and iron overload. J. Clin. Pathol., 2011;64(4):287-296.
  • DiBisceglie AM, Axiostis CA, Hoofnagle JH. Measurament of iron status in patients with chronic hepatitis. Gastroenterology; 1992; 102: 2108-13.
  • Hagag AA, Elfatah MA. Therapeutic Value of Silymarin as Iron Chelator in Children with Beta Thalassemia with Iron Overload. J Leuk., 2014; 2(1):2-6.
  • Rund D, Rachmilewitz E. β thalassemia. N Engl J Med., 2005; 353(11): 1135-46.
  • Aydınok Y. Talasemide demir yükü ve selasyon. Talasemi ve Hemoglobinopatiler. 2007;159-17.
  • Leitch HA. Controversies surrounding iron chelation for MDS Blood Rev 2011:25;17-31.
  • Leitch HA. Improving clinical outcome in patients with myelodysplastic syndrome and iron overload using iron chelation therapy, Leukemia Res.,2007; 31-7.
  • Yavuz Delibaş. Talasemi majorlu hastalarda kardiyolojik fonksiyonların değerlendirilmesi. Uzmanlık Tezi, Sağlık Bakanlığı İstanbul Bakırköy kadın doğum ve çocuk hastalıkları eğitim ve araştırma hastanesi, İstanbul, 2009.
  • Porter JB. Concepts and goals in the management of transfusional iron overload. American Journal of Hematology. 2007; 82(12):1136-9.
  • Gabutti V, Piga A. Results of long-term iron chelation therapy. Acta Haematol., 1996, 95: 26-36.
  • Güven S, Erdogan M. Talasemi majorda demir selasyon sagaltımı. Bakırköy Tıp Dergisi 2007;4:125-9.
  • Balfour BJA. Foster RH. Deferiprone: a review of its clinical potential in iron overload in beta-thalassaemia major and other transfusion-dependent diseases. Drugs. 1999;58(3):553-78.
  • Zhou T, Ma Y. Kong, Hider RC. Design of iron chelators with therapeutic application. Dalton Trans.2012; 41(21): 6371-6389.
  • Zhou T, Ma Y. Kong, Hider RC. Design of iron chelators with therapeutic application. Dalton Trans.2012; 41(21): 6371-6389.
  • Olivieri NF, Brittenham GM. Iron chelating therapy and the treatment of thalassemia. Blood 1997, 89(3): 739-761.
  • Cappellini MD, Piga A. Current status in iron chelation in hemoglobinopathies. Curr Mol Med. 2008;8(7):663–674.
  • Hamilton JL, Kizhakkedathu JN. Polymeric nanocarriers for the treatment of systemic iron overload. Mol Cell Ther.,2015; 3:3-24.
  • Porter JB. Practical management of iron overload. Br J Haematol. 2001; 115(2):239-52.
  • Kushner JP, Portetr JP, Olivieri NF. Secondary iron overload. Hematology Am Soc Hematol Educ Program. 2001; 47-61. 42.
  • Choudhry VP, Naithani R. Current status of iron overload and chelation with deferasirox. Indian J Pediatr.,2007; 74 (8): 759-764.Hershko C. Control of disease by selective iron depletion: a novel therapeutic strategy utilizing iron chelators. Baillieres Clin Haematol., 1994; 965-1000.
  • Borgias B, Hugi AD, Raymond KN. Isomerization and solution structures of desferrioxamine B complexes of Al3 and Ga3. Inorg Chem., 1989;28:3538–3545.
  • Serbülent Güzey. Kritik boyutlu kemik defektlerinde desferroksamin uygulamasının kemik greft iyileşmesi üzerine etkisi. Uzmanlık Tezi, Gülhane askeri tıp akademisi Plastik, rekonstrüktif ve estetik cerrahi anabilim dalı, Ankara, 2013; 39-40.
  • Emerit J, Beaumont C, Trivin F. Iron metabolism, free radicals, and oxidative injury. Biomed Pharmacother. 2001; 55(6):333-9.
  • Davis BA, Porter JB. Long term outcome of continuous 24-hour deferoxamine infusion via indwelling intravenous catheters in high risk beta-thalassemia. Blood. 2000; 95(4):36.
  • Davis BA, Porter JB. Long term outcome of continuous 24-hour deferoxamine infusion via indwelling intravenous catheters in high risk beta-thalassemia. Blood. 2000; 95(4):36.
  • Theil EC. Mining ferritin iron: 2 pathways. Blood. 2009; 114: 4325-4326.
  • Taylor JE, Laity PR, Hicks J, Wong SS, Norris K., Khunkamchoo P. Extent of iron pick-up in deforoxamine-coupled polyurethane materials for therapy of chronic wounds. Biomaterials. 2005; 26(30), 6024-33.
  • Nakamura T, Keep RF, Hua Y. Deferoxamine–induced attenuation of brain edema and neuro in a rat model of intracerebral hemorrhage. Neurosurg Focus. 2003;15(4):1-9.
  • Davies SC, Marcus RE, Hungerford JL, Miller MH, Arden GB, Huehns ER. Ocular toxicity of high dose intravenous desferrioxamine. Lancet. 1983; 2 (8343): 181-184.
  • Unal S, Hazirolan T, Eldem G, Gumruk F. The effects of deferasirox on renal, cardiac and hepatic iron load in patients with β-thalassemia major: preliminary results. Pediatr Hematol Oncol., 2011; 28:217-21.
  • Unal S, Hazirolan T, Eldem G, Gumruk F. The effects of deferasirox on renal, cardiac and hepatic iron load in patients with β-thalassemia major: preliminary results. Pediatr Hematol Oncol., 2011; 28:217-21.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Barman JA, Foster RH. Deferiprone: a review of its clinical potential in iron overload in beta-thalassemia major and other transfusion-dependent disease. Drugs. 1999; 58 (3): 553-578.
  • Tanner MA, Smith GC, Westwood MA, Wonke B, Galanello R. Randomized controlled trial of deferiprone or deferoxamine in beta-thalassemia major patients with asymptomatic myocardial siderosis. Blood. 2006;107:3738-44.
  • Cappellini M, Cohen A, Piga A. A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with β-thalassemia. Blood. 2006; 107 (9): 3455-3462.
  • Porter J, Waldmeier F, Bruin G. Pharmacokinetics, metabolism and elimination of the iron chelator drug ICL670 in beta-thalassemia patient. Blood. 2002; 11: 3720.
  • Porter J, Waldmeier F, Bruin G. Pharmacokinetics, metabolism and elimination of the iron chelator drug ICL670 in beta-thalassemia patient. Blood. 2002; 11: 3720.
  • Piga A, Gaglioti C, Fogliacco E, Tricta F. Comparative effects of deferiprone and deferoxamine on survival and cardiac disease in patients with thalassemia major: a retrospective analysis. Haematologica. 2003; 88: 489-496.
  • Hider R. Recent developments centered on orally active iron chelators. Thalassemia Reports 2014; 2261(4): 19-27.
  • Erişim: http://opus.bath.ac.uk/44293/4/Chapter_1_introduction.pdf 2016. Erişim tarihi: 05.09.2017.
  • Richardson DR, Tran, EH, Ponka P. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents. Blood. 1995;86, 4295- 4306.
  • Hershko C, Avramovici-Grisaru S, Link G, Gelfand L, Sarel S. Mechanism of in vivo iron chelation by pyridoxal isonicotinoyl hydrazone and other imino derivatives of pyridoxal. J Lab Clin Med., 1981;98:99– 108.
  • Edward JT, Ponka P, Richardson DR. Partition-coefficients of the iron(III) complexes of pyridoxal isonicotinoyl hydrazone and its analogs and the correlation to iron chelation efficacy. Biometals 1995;8:209–217.
  • Bernhardt PV, Wilson GJ, Sharpe PC, Kalinowski DS, Richardson DR. Tuning the antiproliferative activity of biologically active iron chelators: characterization of the coordination chemistry and biological efficacy of 2-acetylpyridine and 2-benzoylpyridine hydrazone ligands. J. Biol. Inorg. Chem., 2008; 13:107–119.
  • Hider RC. Kong X. Abbate V. Harland R. Collon K. Luker T. Deferitazole, a new orally active iron chelator. Dalton Trans.,2015; 21;44(11):5197-204.
  • Ferrer P. Tripathi AK. Clark MA. , Hand CC , Rienhoff HY, Sullivan DJ. Antimalarial Iron Chelator, FBS0701, Shows Asexual and Gametocyte Plasmodium falciparum Activity and Single Oral Dose Cure in a Murine Malaria Model. Antimicrobial Agents and Chemotherapy. 2015; 59(3): 1418-1426.
  • Erişim: http://www.chemspider.com/Chemical-Structure.23330184.htm.06.05.2016. Erişim tarihi: 05.09.2017.
  • Finch RA, Liu M, Grill SP, Rose WC, Loomis R, Vasquez KM, Cheng Y, Sartorelli AC. Triapine (3-Aminopyridine-2-Carboxaldehyde-Thiosemicarbazone): A Potent İnhibitor of Ribonucleotide Reductase Activity With Broad Spectrum Antitumor Activity. Bioc-hem. Pharmacol.,2000; 15-59(8):983-991.
  • Mackenzie MJ, Saltman D, Hirte H, Low J, Johnson C, Pond G, Moore, MJ. A Phase II study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) and gemcitabine in advanced pancreatic carcinoma. A trial of the Princess Margaret hospital Phase II consortium. Invest New Drugs. 2007; 25, 553-558.
  • Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA. Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada Clinical Trials Group. Invest New Drugs. 2007; 25, 471-477.
  • Barton JC. Drug evaluation: deferitrin (GT-56-252; NaHBED) for iron overload disorders. IDrugs. 2007; 10(4):270-81. 94. Cunningham MJ, Macklin EA, Muraca G, Neufeld EJ. Successful pregnancy in thalassemia major women in the Thalassemia Clinical Research Network. Pediatr Res. 2004;55:294A.
  • Erişim: http://www.pharmacodia.com/web/drug/1_5372.html Erişim tarihi:10.02.2017.
  • Grady RW, Bakke J, Steinmetz K, Bishop TR, Hedlund BE. Pharmacokinetic and Toxicological Results of a 28-Day Multiple Dose Study of Starch-Conjugated Deferoxamine (S-DFO, 40SD02) in Iron-Loaded Dogs. Blood. 2007; 110:2780.
  • Dragsten PR, Hallaway PE, Hanson GJ, Berger AE, Bernard B, Hedlund BE. First human studies with a high-molecular-weight iron chelator. J Lab Clin Med.,2000; 135(1):57-65.
  • Harmatz P. Grady RW. Dragsten P, Vichinsky E, Giardina P, Madden J, Miller B, Hanson G, Hedlund. B. Phase Ib clinical trial of starch-conjugated deferoxamine (40SD02): a novel long-acting iron chelator. British Journal of Haematology, 2007; 138: 374–381.

Chelation Therapy and Chelating Ligands for Iron Overload

Year 2019, Volume: 28 Issue: 1, 66 - 81, 31.03.2019

Abstract

Iron overload is a condition of excess iron stores in different parts of body which can be largely prevented by the use of chelating agents. The human body doesn’t have any a physiological mechanism to eliminate iron overload. Currently applied methods for treatment of iron overload consisting of chelation therapy. Iron chelation therapy involves the use of ligating drugs that avidly bind iron for the treatment of potentially iron overload disease. In this review, we tried to give information about the reasons for the occurrence of iron overload in the human body, the latest developments in chelators and chelators that are in the trial phase at the clinic.

References

  • Flora SJS, Pachauri V. Chelation in metal intoxication. Int J Environ Res Public Health, 2010;7(7):2745-2788.
  • Hentze MW, Muckenthaler MU, Andrews NC. Balancing acts: molecular control of mammalianiron metabolism. Cell, 2004; 117:285-297.
  • Richardson DR. Iron chelators as therapeutic agents for the treatment of cancer. Crit Rev Oncol Hematology. 2002; 42:267-281.
  • Winter WE, Bazydlo LA, Harris NS. The moleculer biology of human iron metabolism. Laboratory Medicine, 2014; 45(2): 92-102
  • Torti SV, Torti FM. Iron and cancer: more ore to be mined. Cancer, 2013; 13:342-355
  • Chen JJ, Luo ZD, Zhao ZN, Xie LN, Zheng WJ, Chen TF. Cellular localization of iron(II) polypyridyl complexes determines their anticancer action mechanisms. Biomaterials, 2015; 71:168-177.
  • Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005;57:547-583.
  • Crisponi G, Remelli M. Iron chelating agents for the treatment of iron overload. Coord. Chem. Rev. 2008; 252(10):1225-1240.
  • Angelucci E, Brittenham GM, McLaren CE. Hepatic iron concentration and total body iron stores in Thalassemia Major. N Engl. J Med. 2000; 343:327-331.Olivieri NF. The β thalassemias. N Engl J Med., 1999; 341(2): 99-109.
  • Hershko C, Konijin AM, Link G. Iron chelators for thalasaemia. Br J Haematol. 1998; 101:399-406.
  • Faa G. Crisponi G. Iron chelating agents in clinical practice. Coordination Chemistry; 1999; 184(1):291-310.
  • Liu ZD, Hider RC. Design of clinically usefull iron (III)-selective chelators. Med.Res Rev. 2002;22:26-64.
  • Whetherall DJ. Pathophysiology of thalassemia. Bailliére’s Clinical Hematology. 1998; 11(1): 127-146.
  • Ganz T. Hepcidin in iron metabolism. Current Opinion in Hemayology. 2004;11:251-254.
  • Feng M, Does LV, Bantjees A. Iron(III) chelating resins. VI. Stability constants of iron(III)-ligand complexes on insoluble polymeric matrices. Journal of Applied polymer Science, 1994; 56, 1231-1237.
  • Ellis J. Neufeld oral chelators deferasirox and deferiprone for transfusional iron overload in thalassemia major: new data, new questions. Blood, 2006; 107: 3436-3441.
  • Faa G. Crisponi G. Iron chelating agents in clinical practice. Coordination Chemistry Reviews.1999; 184:291-310.
  • Beutler E. Hemochromatosis: genetics and pathophysiology. Annu Rev Med., 2006;57:331-47.
  • Weinberg ED. Iron loading and disease surveillance. Emerg Infect Dis., 1999;5(3):346-52.
  • Gattermann N, Rachmilewitz EA. Iron overload in MDS-pathophysiology, diagnosis, and complications. Ann Hematol. 2011;90(1):1–10.
  • Cabantchik ZI, Breuer W, Zanninelli G, Cianciulli P. LPI-labile plasma iron in iron overload. Best Pract Res Clin Haematol. 2005;18(2):277–287.
  • Brittenham GM. Iron-chelating therapy for transfusional iron overload. N Engl J Med. 2011;364(2):146–156.
  • Musallam KM, Cappellini MD, Wood JC. Elevated liver iron concentration is a marker of increased morbidity in patients with β thalassemia intermedia. Haematologica., 2011;96(11):1605–1612.
  • Corbett JV. Accidental poisoning with iron supplements. MCN: Am J Maternal/Child Nursing 1995; 20:234.
  • Taher A, Nathan D, Porter J. Evaluation of Iron Levels to Avoid the Clinical Sequelae of Iron Overload. Semin Hematol., 2007; 44:S2-S6.
  • Yen AW, Fancher TL, Bowlus CL. Revisiting hereditary hemochromatosis: current concepts and progress. Am J Med., 2006;119:391–9.
  • Kohgo Y. Ikuta K. Ohtake T. Torimoto Y. Kato J. Body iron metabolism and pathophysiology of iron overload. Int J Hematol., 2008; 88:7–15.
  • Feder JN. ve ark. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet., 1996; 13(4): 399-408.
  • Franchini M. Hereditary iron overload: update on pathophysiology, diagnosis, and treatment. Am J Hematol., 2006;81:202– 9.
  • Bonkovsky HL, Lambrecht RW, Shan Y. Iron as a co-morbid factor in nonhemochromatotic liver disease. Alcohol. 2003; 30:137–44.
  • Pietrangelo A. Hereditary hemochromatosis–a new look at an old disease. N Engl J Med., 2004;350:2383–2397.
  • Siddique A. Kowdley KV. Review article: the iron overload syndromes. Alimentary pharmacology & therapeutics. 2012; 35: 876‐893.
  • Erişim:http://www.lifeextension.com/protocols/metabolic-health/hemochromatosis/page-06?p=1 2017. Erişim tarihi: 05.09.2016.
  • Borgaonkar MR. Hemochromatosis.More common than you think. Can Fam Physician., 2003;49:36-43.
  • Gregory A. Hayflick SJ. Genetics of neurodegeneration with brain iron accumulation. CurrNeurolNeurosci Rep. 2011;11(3):254-261.
  • Sebastiani G. Walker AP. HFE gene in primary and secondary hepatic iron overload. World J Gastroenterol. 2007;21;13(35):4673-89.
  • Heli H, Mirtorabi S, Karimian K. Advances in iron chelation: an update. Expert OpinTher Pat. 2011;21(6):819-856.
  • Brissot P, Ropert M, Le Lan C, Loreal O. Non-transferrin bound iron: A key role in iron overload and iron toxicity. Biochim Biophys Acta,. 2012;1820(3):403-410.
  • Munoz M, Garcia-Erce JA, Remacha AF. Disorders of iron metabolism. Part II: iron deficiency and iron overload. J. Clin. Pathol., 2011;64(4):287-296.
  • DiBisceglie AM, Axiostis CA, Hoofnagle JH. Measurament of iron status in patients with chronic hepatitis. Gastroenterology; 1992; 102: 2108-13.
  • Hagag AA, Elfatah MA. Therapeutic Value of Silymarin as Iron Chelator in Children with Beta Thalassemia with Iron Overload. J Leuk., 2014; 2(1):2-6.
  • Rund D, Rachmilewitz E. β thalassemia. N Engl J Med., 2005; 353(11): 1135-46.
  • Aydınok Y. Talasemide demir yükü ve selasyon. Talasemi ve Hemoglobinopatiler. 2007;159-17.
  • Leitch HA. Controversies surrounding iron chelation for MDS Blood Rev 2011:25;17-31.
  • Leitch HA. Improving clinical outcome in patients with myelodysplastic syndrome and iron overload using iron chelation therapy, Leukemia Res.,2007; 31-7.
  • Yavuz Delibaş. Talasemi majorlu hastalarda kardiyolojik fonksiyonların değerlendirilmesi. Uzmanlık Tezi, Sağlık Bakanlığı İstanbul Bakırköy kadın doğum ve çocuk hastalıkları eğitim ve araştırma hastanesi, İstanbul, 2009.
  • Porter JB. Concepts and goals in the management of transfusional iron overload. American Journal of Hematology. 2007; 82(12):1136-9.
  • Gabutti V, Piga A. Results of long-term iron chelation therapy. Acta Haematol., 1996, 95: 26-36.
  • Güven S, Erdogan M. Talasemi majorda demir selasyon sagaltımı. Bakırköy Tıp Dergisi 2007;4:125-9.
  • Balfour BJA. Foster RH. Deferiprone: a review of its clinical potential in iron overload in beta-thalassaemia major and other transfusion-dependent diseases. Drugs. 1999;58(3):553-78.
  • Zhou T, Ma Y. Kong, Hider RC. Design of iron chelators with therapeutic application. Dalton Trans.2012; 41(21): 6371-6389.
  • Zhou T, Ma Y. Kong, Hider RC. Design of iron chelators with therapeutic application. Dalton Trans.2012; 41(21): 6371-6389.
  • Olivieri NF, Brittenham GM. Iron chelating therapy and the treatment of thalassemia. Blood 1997, 89(3): 739-761.
  • Cappellini MD, Piga A. Current status in iron chelation in hemoglobinopathies. Curr Mol Med. 2008;8(7):663–674.
  • Hamilton JL, Kizhakkedathu JN. Polymeric nanocarriers for the treatment of systemic iron overload. Mol Cell Ther.,2015; 3:3-24.
  • Porter JB. Practical management of iron overload. Br J Haematol. 2001; 115(2):239-52.
  • Kushner JP, Portetr JP, Olivieri NF. Secondary iron overload. Hematology Am Soc Hematol Educ Program. 2001; 47-61. 42.
  • Choudhry VP, Naithani R. Current status of iron overload and chelation with deferasirox. Indian J Pediatr.,2007; 74 (8): 759-764.Hershko C. Control of disease by selective iron depletion: a novel therapeutic strategy utilizing iron chelators. Baillieres Clin Haematol., 1994; 965-1000.
  • Borgias B, Hugi AD, Raymond KN. Isomerization and solution structures of desferrioxamine B complexes of Al3 and Ga3. Inorg Chem., 1989;28:3538–3545.
  • Serbülent Güzey. Kritik boyutlu kemik defektlerinde desferroksamin uygulamasının kemik greft iyileşmesi üzerine etkisi. Uzmanlık Tezi, Gülhane askeri tıp akademisi Plastik, rekonstrüktif ve estetik cerrahi anabilim dalı, Ankara, 2013; 39-40.
  • Emerit J, Beaumont C, Trivin F. Iron metabolism, free radicals, and oxidative injury. Biomed Pharmacother. 2001; 55(6):333-9.
  • Davis BA, Porter JB. Long term outcome of continuous 24-hour deferoxamine infusion via indwelling intravenous catheters in high risk beta-thalassemia. Blood. 2000; 95(4):36.
  • Davis BA, Porter JB. Long term outcome of continuous 24-hour deferoxamine infusion via indwelling intravenous catheters in high risk beta-thalassemia. Blood. 2000; 95(4):36.
  • Theil EC. Mining ferritin iron: 2 pathways. Blood. 2009; 114: 4325-4326.
  • Taylor JE, Laity PR, Hicks J, Wong SS, Norris K., Khunkamchoo P. Extent of iron pick-up in deforoxamine-coupled polyurethane materials for therapy of chronic wounds. Biomaterials. 2005; 26(30), 6024-33.
  • Nakamura T, Keep RF, Hua Y. Deferoxamine–induced attenuation of brain edema and neuro in a rat model of intracerebral hemorrhage. Neurosurg Focus. 2003;15(4):1-9.
  • Davies SC, Marcus RE, Hungerford JL, Miller MH, Arden GB, Huehns ER. Ocular toxicity of high dose intravenous desferrioxamine. Lancet. 1983; 2 (8343): 181-184.
  • Unal S, Hazirolan T, Eldem G, Gumruk F. The effects of deferasirox on renal, cardiac and hepatic iron load in patients with β-thalassemia major: preliminary results. Pediatr Hematol Oncol., 2011; 28:217-21.
  • Unal S, Hazirolan T, Eldem G, Gumruk F. The effects of deferasirox on renal, cardiac and hepatic iron load in patients with β-thalassemia major: preliminary results. Pediatr Hematol Oncol., 2011; 28:217-21.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Crosa JH. Walsh CT. Genetics ve assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 2002;66: 223-49.
  • Barman JA, Foster RH. Deferiprone: a review of its clinical potential in iron overload in beta-thalassemia major and other transfusion-dependent disease. Drugs. 1999; 58 (3): 553-578.
  • Tanner MA, Smith GC, Westwood MA, Wonke B, Galanello R. Randomized controlled trial of deferiprone or deferoxamine in beta-thalassemia major patients with asymptomatic myocardial siderosis. Blood. 2006;107:3738-44.
  • Cappellini M, Cohen A, Piga A. A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with β-thalassemia. Blood. 2006; 107 (9): 3455-3462.
  • Porter J, Waldmeier F, Bruin G. Pharmacokinetics, metabolism and elimination of the iron chelator drug ICL670 in beta-thalassemia patient. Blood. 2002; 11: 3720.
  • Porter J, Waldmeier F, Bruin G. Pharmacokinetics, metabolism and elimination of the iron chelator drug ICL670 in beta-thalassemia patient. Blood. 2002; 11: 3720.
  • Piga A, Gaglioti C, Fogliacco E, Tricta F. Comparative effects of deferiprone and deferoxamine on survival and cardiac disease in patients with thalassemia major: a retrospective analysis. Haematologica. 2003; 88: 489-496.
  • Hider R. Recent developments centered on orally active iron chelators. Thalassemia Reports 2014; 2261(4): 19-27.
  • Erişim: http://opus.bath.ac.uk/44293/4/Chapter_1_introduction.pdf 2016. Erişim tarihi: 05.09.2017.
  • Richardson DR, Tran, EH, Ponka P. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents. Blood. 1995;86, 4295- 4306.
  • Hershko C, Avramovici-Grisaru S, Link G, Gelfand L, Sarel S. Mechanism of in vivo iron chelation by pyridoxal isonicotinoyl hydrazone and other imino derivatives of pyridoxal. J Lab Clin Med., 1981;98:99– 108.
  • Edward JT, Ponka P, Richardson DR. Partition-coefficients of the iron(III) complexes of pyridoxal isonicotinoyl hydrazone and its analogs and the correlation to iron chelation efficacy. Biometals 1995;8:209–217.
  • Bernhardt PV, Wilson GJ, Sharpe PC, Kalinowski DS, Richardson DR. Tuning the antiproliferative activity of biologically active iron chelators: characterization of the coordination chemistry and biological efficacy of 2-acetylpyridine and 2-benzoylpyridine hydrazone ligands. J. Biol. Inorg. Chem., 2008; 13:107–119.
  • Hider RC. Kong X. Abbate V. Harland R. Collon K. Luker T. Deferitazole, a new orally active iron chelator. Dalton Trans.,2015; 21;44(11):5197-204.
  • Ferrer P. Tripathi AK. Clark MA. , Hand CC , Rienhoff HY, Sullivan DJ. Antimalarial Iron Chelator, FBS0701, Shows Asexual and Gametocyte Plasmodium falciparum Activity and Single Oral Dose Cure in a Murine Malaria Model. Antimicrobial Agents and Chemotherapy. 2015; 59(3): 1418-1426.
  • Erişim: http://www.chemspider.com/Chemical-Structure.23330184.htm.06.05.2016. Erişim tarihi: 05.09.2017.
  • Finch RA, Liu M, Grill SP, Rose WC, Loomis R, Vasquez KM, Cheng Y, Sartorelli AC. Triapine (3-Aminopyridine-2-Carboxaldehyde-Thiosemicarbazone): A Potent İnhibitor of Ribonucleotide Reductase Activity With Broad Spectrum Antitumor Activity. Bioc-hem. Pharmacol.,2000; 15-59(8):983-991.
  • Mackenzie MJ, Saltman D, Hirte H, Low J, Johnson C, Pond G, Moore, MJ. A Phase II study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) and gemcitabine in advanced pancreatic carcinoma. A trial of the Princess Margaret hospital Phase II consortium. Invest New Drugs. 2007; 25, 553-558.
  • Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA. Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada Clinical Trials Group. Invest New Drugs. 2007; 25, 471-477.
  • Barton JC. Drug evaluation: deferitrin (GT-56-252; NaHBED) for iron overload disorders. IDrugs. 2007; 10(4):270-81. 94. Cunningham MJ, Macklin EA, Muraca G, Neufeld EJ. Successful pregnancy in thalassemia major women in the Thalassemia Clinical Research Network. Pediatr Res. 2004;55:294A.
  • Erişim: http://www.pharmacodia.com/web/drug/1_5372.html Erişim tarihi:10.02.2017.
  • Grady RW, Bakke J, Steinmetz K, Bishop TR, Hedlund BE. Pharmacokinetic and Toxicological Results of a 28-Day Multiple Dose Study of Starch-Conjugated Deferoxamine (S-DFO, 40SD02) in Iron-Loaded Dogs. Blood. 2007; 110:2780.
  • Dragsten PR, Hallaway PE, Hanson GJ, Berger AE, Bernard B, Hedlund BE. First human studies with a high-molecular-weight iron chelator. J Lab Clin Med.,2000; 135(1):57-65.
  • Harmatz P. Grady RW. Dragsten P, Vichinsky E, Giardina P, Madden J, Miller B, Hanson G, Hedlund. B. Phase Ib clinical trial of starch-conjugated deferoxamine (40SD02): a novel long-acting iron chelator. British Journal of Haematology, 2007; 138: 374–381.
There are 95 citations in total.

Details

Primary Language Turkish
Subjects Health Care Administration
Journal Section Review
Authors

Gülüzar Özbolat

Abdullah Tuli This is me

Publication Date March 31, 2019
Acceptance Date April 22, 2018
Published in Issue Year 2019 Volume: 28 Issue: 1

Cite

AMA Özbolat G, Tuli A. Demir Fazlalığı İçin Şelasyon Tedavisi ve Şelasyon Ligandları. aktd. March 2019;28(1):66-81.