Scolaris Content Display Scolaris Content Display

Deferasirox para el tratamiento de la sobrecarga de hierro en pacientes con talasemia

Collapse all Expand all

References

Referencias de los estudios incluidos en esta revisión

Cappellini 2005b {published data only}

Angelucci E, Turlin B, Canatan D, Mangiagli A, De Sanctis V, Meddeb B, et al. Iron chelation therapy with deferasirox (Exjade (R), ICL670) or deferoxamine is effective in reducing iron overload in patients with advanced fibrosis and cirrhosis. Blood 2005;106(11 Pt 1). [Abstract no: 2696]CENTRAL
Aydinok Y, Agaoglu L, Bejaoui M, Canatan D, Drelichman G, Economou M, et al. Growth and development of paediatric patients with beta‐thalassaemia treated with deferasirox for up to 5 years. Haematologica 2010;95:429. CENTRAL
Aydinok Y, Arikan C, Karakas Z, Sasmaz I, Canatan D, Agaoglu L, Kilinc Y, Canatar A, Ocak O, Alnigenis E. A comparison of novel serum markers for the assessment of liver fibrosis in patients with beta thalassemia major treated with deferasirox. Haematologica 2010;95:718. CENTRAL
Brissot P, Turlin B, Forni GL, Alimena G, Quarta G, Selleslag D, et al. Iron chelation therapy with deferasirox (Exjade®, ICL670) or deferoxamine results in reduced hepatocellular inflammation and improved liver function in patients with transfusion‐dependent anemia. Blood 2005;106(11). [Abstract no: 823]CENTRAL
Cappellini M, Bejaoui M, Perrotta S, Agaoglu L, Kattamis A, Giardina P, et al. Phase III evaluation of once‐daily, oral therapy with ICL670 (Exjade (R)) versus deferoxamine in patients with beta‐thalassemia and transfusional hemosiderosis. Blood 2004;104(11 Pt 1). [Abstract no: 3619]CENTRAL
Cappellini MD, Bejaoui M, Agaoglu L, Canatan D, Capra M, Cohen A, et al. Iron chelation with deferasirox in adult and pediatric patients with thalassemia major: efficacy and safety during 5 years' follow‐up. Blood 2011;118(4):884‐93. CENTRAL
Cappellini MD, Bejaoui M, Agaoglu L, Lai ME, Mangiagli A, Strauss G, et al. Patient satisfaction with deferasirox (Exjade®, ICL670) an oral form of chelation therapy versus deferoxamine an infused chelation therapy. Blood 2005;106(11). [Abstract no: 2704]CENTRAL
Cappellini MD, Bejaoui M, Agaoglu L, Porter J, Coates T, Jeng M, et al. Prospective evaluation of patient‐reported outcomes during treatment with deferasirox or deferoxamine for iron overload in patients with beta‐thalassemia. Clinical Therapeutics 2007;29(5):909‐17. CENTRAL
Cappellini MD, Cohen A, Piga A, Bejaoui M, Perrotta S, Agaoglu L, et al. A phase 3 study of deferasirox (ICL670), a once‐daily oral iron chelator, in patients with beta‐thalassemia. Blood 2006;107(9):3455‐62. CENTRAL
Cappellini MD, Giardina P, Porter J, Coates T, Della Porta MG, Siegal J, et al. Long‐term safety and tolerability of the once‐daily, oral iron chelator deferasirox (Exjade®, ICL670) in patients with transfusional iron overload. Blood 2006;108(11). [Abstract no: 1768]CENTRAL
Christoforidis A, Tsatra I, Karasmanis K, Zevgaridou E, Koussi A, Tsitourides I, et al. Evaluation of myocardial deposition assessed with M.R.I in young thalassaemic patients receiving one year of deferasirox versus deferoxamine. Haematologica 2006;91(S1):209. CENTRAL
Cohen A, Masera G, Zoumbos N, Uysal Z, Boulet D, Watman N, et al. Effect of iron intake on control of body iron in patients with thalassemia major treated with deferasirox (Exjade((R)), ICL670). Blood 2005;106(11 Pt 1). [Abstract no: 822]CENTRAL
Cohen AR, Glimm E, Porter JB. Effect of transfusional iron intake on response to chelation therapy in beta‐thalassemia major. Blood 2008;111(2):583‐7. CENTRAL
Deugnier Y, Turlin B, Dong V, Giannone V, Zhang Y, Griffel L, et al. Deferasirox improves liver pathology in beta‐Thalassemia patients with transfusional iron overload. Blood 2010;116(21):1734‐5. CENTRAL
Deugnier Y, Turlin B, Ropert M, Bejaoui M, Athanassiou‐Metaxa M, Cario H, et al. Semi‐quantitative assessment of hemosiderin distribution accurately reflects reductions in liver iron concentration following therapy with deferasirox (Exjade (R), ICL670) or deferoxamine in patients with transfusion‐dependent anemia. Blood 2005;106(11 Pt 1):761A. CENTRAL
Deugnier Y, Turlin B, Ropert M, Cappellini MD, Porter JB, Giannone V, et al. Improvement in liver pathology of patients with beta‐thalassemia treated with deferasirox for at least 3 years. Gastroenterology 2011;141(4):1202‐11. CENTRAL
Deugnier Y, Turlin B, Ropert‐Bouchet M, Rabault B, Brissot P. Effect of iron chelation therapy with deferasirox (Exjade (R), ICL670) or deferoxamine on hepatocellular inflammation and liver function in patients with transfusion‐dependent anemia. Journal of Hepatology 2008;48(Suppl 2):S79‐80. CENTRAL
Fischer R, Harmatz P, Nielsen P. Does liver biopsy overestimate liver iron concentration?. Blood2006; Vol. 108, issue 5:1775‐6; author reply 1776. [PUBMED: 16926297]CENTRAL
Galanello R. Evaluation of ICL670, a once‐daily oral iron chelator in a phase III clinical trial of beta‐thalassemia patients with transfusional iron overload. Annals of the New York Academy of Sciences 2005;1054:183‐5. CENTRAL
Grosse R, Janssen G, Engelhardt R, Groeger M, Leismann O, Nielsen P, et al. Chelator efficacy of deferasirox and deferoxamine determined by SQUID biosusceptometry. Blood 2006;108(11). [Abstract no: 1779]CENTRAL
Karnon J, Akehurst RL, Jewitt K, Ossa D. Cost utility analysis deferasirox versus deferoxamine (desferal) for patients requiring iron chelation therapy in the United Kingdom. Haematologica 2007;92 Suppl 1:222. CENTRAL
Kattamis A, Paley C, Study Investigators. Iron chelating efficacy is related to transfusional iron intake in pediatric patients‐treated with deferasirox(Exjade (R), ICL670). Pediatric Blood & Cancer 2007;48(6):609. CENTRAL
Kattamis C, Kilinc Y, Fattoum S, Ferster A, Gallisai D, Maggio A, et al. Deferasirox (Exjade (R), ICL670) demonstrates iron chelating efficacy related to transfusional iron intake in pediatric patients. Blood 2005;106(11 Pt 1):756A. [Abstract no: 2692]CENTRAL
Kattamis C, Meddeb B, Ressayre‐Djaffer C, Alberti D, Parsing Using Smart Source. Baseline iron studies demonstrate severe iron overload in patients enrolled into the deferasirox (Exjade(R), ICL670) clinical trial programme. Haematologica 2006;91(Suppl 1):415. CENTRAL
Kleinert DA, Jones E, Sison CP, Marks P, Giardina PJ. Left ventricular cardiac function during the course of a one year multicenter trial of the safety and efficacy with ICL670 5‐40mg/kg/day and deferoxamine 20‐60 mg/kg/day in beta‐thalassemia patients with transfusional hemosideosis. Blood 2005;106(11 Pt 2):44B. [Abstract no: 3853]CENTRAL
Martin MG, Arcasoy MO. Deferasirox versus deferoxamine. Blood 2006;108(2):774‐5; author reply 775‐6. [PUBMED: 16822908]CENTRAL
Piga A, Fischer R, Harmatz P, St Pierre TG, Longo F, Fung E, et al. Comparison of LIC obtained from biopsy, BLS and R‐2‐MRI in iron overloaded patients with beta‐thalassemia, treated with deferasirox (Exjade (R), ICL670). Blood 2005;106(11 Pt 1):755A. CENTRAL
Walter P, Macklin E, Porter J, Evans P, Coates T, Olivieri NF, et al. Control of oxidant‐stress and inflammation by iron chelators Deferasirox (ICL670) or Deferoxamine in beta‐thalassaemia: an ancillary study of the Novartis CICL670A0107 Trial. Blood 2005;106(11). [Abstract no: 3598]CENTRAL
Walter PB, Macklin EA, Porter J, Evans P, Kwiatkowski JL, Neufeld EJ, et al. Inflammation and oxidant‐stress in beta‐thalassemia patients treated with iron chelators deferasirox (ICL670) or deferoxamine: an ancillary study of the Novartis CICL670A0107 trial. Haematologica 2008;93(6):817‐25. CENTRAL
Walter PB, Porter J, Evans P, Kwiatkowski JL, Neufeld EJ, Coates T, et al. Increased leucocyte apoptosis in transfused beta‐thalassaemia patients. British journal of haematology 2013;160(3):399‐403. [PUBMED: 23216540]CENTRAL
Walter PB, Porter J, Evans P, Kwiatkowski JL, Neufeld EJ, Coates T, et al. Leukocyte apoptosis and mitochondrial dysfunction in beta‐thalassemia patients treated with deferasirox or deferoxamine. Blood 2007;110(11 Pt 1):815A. [Abstract no: 2773]CENTRAL

Chirico 2013 {published data only}

Chirico V, Antonio L, Vincenzo S, Luca N, Valeria F, Basilia P, et al. Thyroid dysfunction in thalassaemic patients: ferritin as a prognostic marker and combined iron chelators as an ideal therapy. European Journal of Endocrinology 2013;169(6):785‐93. CENTRAL
Chirico V, Rigoli L, Piraino B, La Rosa M, Salpietro C, Arrigo T. Endocrinopathies in beta‐thalassemia major: Evidences from ten years of follow‐up and evaluation of combined iron chelation therapy. Hormone Research in Paediatrics 2013;80:318. CENTRAL

Elalfy 2015a {published data only}

Elalfy M, Adly AA, Ismail E, Elalfy O. Efficacy and safety of vitamin C as an adjuvant to iron chelation therapy in young patients with b‐thalassemia major: a randomized prospective trial [abstract]. Haematologica 2015;100 Suppl 1:131. CENTRAL
Elalfy MS, Saber M, Adly A, Ismail E, Tarif M, Ibrahim F, et al. Role of vitamin C As an adjuvant therapy with different iron chelators in young beta‐thalassemia major patients: Safety and efficacy in relation to tissue Iron overload. Blood 2014;124(21):4046. CENTRAL
Elalfy MS, Saber MM, Adly AA, Ismail EA, Tarif M, Ibrahim F, et al. Role of vitamin C as an adjuvant therapy to different iron chelators in young beta‐thalassemia major patients: efficacy and safety in relation to tissue iron overload. European Journal of Haematology 2016;96(3):318‐26. [DOI: 10.1111/ejh.12594]CENTRAL

Elalfy 2015b {published data only}

Elalfy M, Walli Y, Adly A, Henawy Y. 18 months data of a randomized controlled trial of combined deferiprone (DFP) and deferasirox (DFX) versus combined deferiprone and deferoxamine (DFO), in young B‐thalassemia major. Haematologica 2014;99:443‐4. [Abstract no: ABSSUB‐5856]CENTRAL
Elalfy MS, Adly AM, Wali Y, Tony S, Samir A, Elhenawy YI. Efficacy and safety of a novel combination of two oral chelators deferasirox/deferiprone over deferoxamine/deferiprone in severely iron overloaded young beta thalassemia major patients. European Journal of Haematology 2015;95(5):411‐20. CENTRAL
Elalfy MS, Wali Y, Tony S, Samir A, Adly A. Comparison of two combination iron chelation regimens, deferiprone and deferasirox versus deferiprone and deferoxamine, in pediatric patients with β‐thalassemia major. Blood 2013;122(21):559. CENTRAL

Galanello 1999 {published data only}

Galanello R, Piga A, Alberti D, Rouan MC, Bigler H, Sechaud R. Safety, tolerability, and pharmacokinetics of ICL670, a new orally active iron‐chelating agent in patients with transfusion‐dependent iron overload due to beta‐thalassemia. Journal of Clinical Pharmacology 2003;43(6):565‐72. CENTRAL
Piga A, Galanello R, Dessi C, Sedaro M, Loehrer F, Schaud R, et al. A novel oral iron chelator (ICL670A): results of a phase I single‐dose safety study. Blood 1999;10(Suppl 1 Pt 2):35b. CENTRAL

Habibian 2014 {published data only}

Habibian N. Comparison of therapeutic effect of osveral & desferal in patients with thalassemia (bahonar hospital in karaj 2012‐13). Iranian Journal of Pediatrics 2014;24(Suppl 2):S27. CENTRAL

Hassan 2016 {published data only}

Hassan MA, Tolba OA. Iron chelation monotherapy in transfusion‐dependent beta‐thalassemia major patients: a comparative study of deferasirox and deferoxamine. Electronic Physician 2016;8(5):2425‐31. [CENTRAL: 1164827; CRS: 5500135000001568; PUBMED: 27382454]CENTRAL

Kakkar 2014 {published data only}

Kakkar S, Sobti PC, Jhinger, PK. Safety and efficacy of combination of deferiprone and deferasirox in patients with thalassemia major. Haematologica 2014;99 Suppl 1:735. [Abstract no: PB1923]CENTRAL

Molavi 2013 {published data only}

Molavi MA, Doozandeh H, Nazemi A, Evazi R, Mansoori F. Comparison of therapeutic response and complications of oral Osveral and injection Desfereal chelating agent in patient with thalassemia major. Asian Journal of Medical and Pharmaceutical Researches 2013;3(3):93‐7. CENTRAL

Molavi 2014 {published data only}

Molavi MA, Poor ST, Malesksabet M. Combined Desferrioxamine (Desferal) and Deferasirox in children. Advances in Biological Research 2014;8(4):171‐5. CENTRAL

Nisbet‐Brown 2001 {published data only}

Giardina P, Olivieri NF, Nisbet‐Brown E, Grady RW, Sizer KC, Sechaud R, et al. ICL670, a tridentate orally‐active iron chelator, provides sufficient net negative iron balance and increased serum iron binding capacity in iron‐overloaded patients with thalassemia. 7th Congress of the European Hematology Association; 2002 June 6‐9; Florence, Italy. 2002. [Abstract no: 0236]CENTRAL
Nisbet‐Brown E, Olivieri NF, Giardina PJ, Grady RW, Neufeld EJ, Sechaud R, et al. Effectiveness and safety of ICL670 in iron‐loaded patients with thalassaemia: a randomised, double‐blind, placebo‐controlled, dose‐escalation trial. Lancet 2003;361(9369):1597‐602. CENTRAL
Nisbet‐Brown E, Olivieri NF, Giardina PJ, Grady RW, Sizer K, Sechaud R, et al. ICL670A, a tridentate orally‐active iron chelator, provides net negative iron balance and increased serum iron binding capacity in iron‐overloaded patients with thalassemia. Blood 2001;98(11 Pt 1):747a. CENTRAL

Peng 2013 {published data only}

Peng P, Long LL, Huang ZK, Zhang L, Li XH, Feng X, et al. Comparison of deferasirox and deferoxamine treatment in iron‐overloaded patients: Liver iron concentration determined by quantitative MRI‐R2*. Chinese Journal of Radiology 2013;47(1):55‐9. CENTRAL

Pennell 2014 {published and unpublished data}

Aydinok Y, Porter JB, Piga A, Elalfy M, El‐Beshlawy A, Kilinc Y, et al. Prevalence and distribution of iron overload in patients with transfusion‐dependent anemias differs across geographic regions: results from the CORDELIA study. European Journal of Haematology 2014;95(3):244‐53. CENTRAL
Pennell D, Porter J, Piga A, El‐Alfy M, El‐Beshlawy A, Kilinc Y, et al. Prevalence of cardiac iron overload in patients with transfusion‐dependent anemias: data from the randomized, active‐controlled deferasirox CORDELIA trial. Haematologica 2012;97 Suppl 1:384. [CENTRAL: 977599; CRS: 5500100000011087; Abstract no: 0928]CENTRAL
Pennell DJ, Porter JB, Piga A, Lai Y, El‐Beshlawy A, Belhoul K, et al. Deferasirox compared with deferoxamine for the removal of cardiac iron in patients with beta‐thalassemia major: 2‐Year data from the Cordelia extension. Blood 2013;122(21):1018. CENTRAL
Pennell DJ, Porter JB, Piga A, Lai Y, El‐Beshlawy A, Belhoul KM, et al. A 1‐year randomized controlled trial of deferasirox versus deferoxamine for myocardial iron removal in beta‐thalassemia major (CORDELIA). Blood 2014;123(10):1447‐54. [CENTRAL: 977602; CRS: 5500125000000573; PUBMED: 24385534]CENTRAL
Pennell DJ, Porter JB, Piga A, Lai Y, El‐Beshlawy A, Beloul K, et al. A multicenter, randomized, open‐label trial evaluating deferasirox compared with deferoxamine for the removal of cardiac iron in patients with β‐thalassemia major and iron overload (CORDELIA). Blood 2012;121(21). [CENTRAL: 977601; CRS: 5500125000000570; Abstract no: 2124]CENTRAL
Pennell DJ, Porter JB, Piga A, Lai YR, El‐Beshlawy A, Elalfy M, et al. Sustained improvements in myocardial T2* over 2 years in severely iron‐overloaded patients with beta thalassemia major treated with deferasirox or deferoxamine. American Journal of Hematology 2015;90(2):91‐6. CENTRAL

Piga 2002 {published data only}

Cappellini M, Galanello R, Piga A, Forni GL, Zanaboni L, Muroni P, et al. Update on the effects of icl670, a novel tridentate oral iron chelator, on liver iron concentration in patients with transfusion dependent iron overload. 7th Meeting of the European Hematology Association; 2002 Jun 6‐9; Florence, Italy Florence, Italy, 6‐9 June, 2002. 2002:184. CENTRAL
Cappellini MD, Galanello R, Piga A, Forni GL, Opitz H, Ford JM, et al. Pharmacokinetics (PK) profile of the new oral iron chelator ICL670 after 6 months of treatment in a phase II study in patients with transfusional hemosiderosis. 8th Congress of the European Hematology Association; 2003 Jun 12‐15; Lyon, France. Lyon, 2003. CENTRAL
Piga A, Galanello R, Cappellini M, Forni GL, Opitz H, Ford JM, et al. Phase II study of oral chelator ICL670 in thalassaemia patients with transfusional iron overload: Efficacy, safety, pharmacokinetics (PK) and pharmacodynamics (PD) after 6 months of therapy. Blood 2002;100(11 Pt 1):5a. CENTRAL
Piga A, Galanello R, Cappellini MD, Forni GL, Lupo G, Ford JM, et al. Phase II study of ICL670, an oral chelator, in adult thalassaemia patients with transfusional iron overload: efficacy, safety, pharmacokinetics (PK) and pharmacodynamics (PD) after 18 months of therapy. Blood 2003;102(11). [Abstract no: 412]CENTRAL
Piga A, Galanello R, Forni GL, Cappellini MD, Origa R, Zappu A, et al. Randomized phase II trial of deferasirox (Exjade, ICL670), a once‐daily, orally‐administered iron chelator, in comparison to deferoxamine in thalassemia patients with transfusional iron overload. Haematologica 2006;91(7):873‐80. CENTRAL

Sanjeeva 2015 {published data only}

Matti M. Study on comparison of efficacy and adverse effects of deferasirox vs deferiprone in treatment of iron overload in thalassemia [doctoral thesis]. Bangalore (India): Rajiv Gandhi University of Health Sciences, 2013. CENTRAL
Sanjeeva GN, Nijaguna N, Matti M, Chebbi PG. Efficacy and safety of deferasirox when compared to deferiprone as oral iron chelating agent: A randomized control trial. Journal of of Evolution of Medical and Dental Sciences 2015;4(24):4178‐85. [DOI: 10.14260/jemds/2015/601]CENTRAL

Taher 2012 {published and unpublished data}

Porter J, Cappellini M, Kattamis A, Viprakasit V, Lawniczek T, Ros J, et al. Relationships between plasma non‐transferrin‐bound iron and markers of iron overload, anaemia and ineffective erythropoiesis in non‐transfusion‐dependent thalassaemia syndromes. Haematologica 2011;96 Suppl:228‐9. CENTRAL
Porter JB, Cappellini MD, Kattamis A, Viprakasit V, Musallam KM, Zhu Z, et al. Changes in alternative iron overload parameters following 1 year of deferasirox therapy in patients with non‐transfusion‐dependent thalassemia: A longitudinal analysis from the thalassa study. Haematologica 2014;99 Suppl:441‐2. CENTRAL
Taher A, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Estimation of liver iron concentration by serum ferritin measurement in non‐transfusion‐dependent thalassemia patients: Analysis from the 1‐year thalassa study. Haematologica 2012;97 Suppl:383. CENTRAL
Taher A, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Reduction in liver iron concentration is consistent across subgroups of non‐transfusion‐dependent thalassemia patients treated with deferasirox: Results from the 1‐year thalassa study. Haematologica 2012;97 Suppl:381‐2. CENTRAL
Taher A, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Serum ferritin for predicting clinically relevant lic thresholds to guide management of patients with nontransfusion‐dependent thalassemia treated with deferasirox: Thalassa study extension analysis. Haematologica 2013;98 Suppl:486. CENTRAL
Taher A, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. The safety profile of deferasirox remains consistent as non‐transfusion‐dependent thalassemia patients approach the target liver iron concentration of <3 mg fe/g dw for interrupting chelation. Haematologica 2013;98 Suppl 1:165‐6. CENTRAL
Taher A, Porter JB, Kattamis A, Viprakasit V, Lawniczek T, Pereno R, et al. Randomized phase two study evaluating the efficacy and safety of deferasirox in non‐transfusion‐dependent thalassemia patients with iron overload [abstract]. Blood 2009;114(Issue 22). [Abstract no: 5111]CENTRAL
Taher A, Viprakasit V, Porter J, Kattamis A, Lawniczek T, Pereno R, et al. Factors associated with iron accumulation in non‐transfusion‐dependent thalassaemia syndromes: Baseline data from multicenter international deferasirox study (Thalassa). Haematologica 2010;95 Suppl:707. CENTRAL
Taher AT, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox reduces iron overload significantly in nontransfusion‐dependent thalassemia: 1‐year results from a prospective, randomized, double‐blind, placebo‐controlled study. Blood 2012;120(5):970‐7. [CRS: 5500125000000556; PUBMED: 22589472]CENTRAL
Taher AT, Porter J, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Online supplemental appendix to "Deferasirox reduces iron overload significantly in nontransfusion‐dependent thalassemia: 1‐year results from a prospective, randomized, double‐blind, placebo‐controlled study.". Blood 2012;120(5):970‐7. [CENTRAL: 842274; CRS: 5500125000000551; PUBMED: 22589472]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Approaching low liver iron burden in chelated patients with non‐transfusion‐dependent thalassemia: The safety profile of deferasirox. European Journal of Haematology 2014;92(6):521‐6. [CENTRAL: 977521; CRS: 5500125000000555; PUBMED: 24460655]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox continues to reduce iron overload in non‐transfusion‐dependent thalassemia: a one‐year, open‐label extension to a one‐year, randomized, double‐blind, placebo‐controlled study (THALASSA). Blood 2012;120(21). [CENTRAL: 977519; CRS: 5500125000000550; Abstract no: 3258]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox demonstrates a dose‐dependent reduction in liver iron concentration and consistent efficacy across subgroups of non‐transfusion‐dependent thalassemia patients. American Journal of Hematology 2013;88(6):503‐6. [CENTRAL: 964527; CRS: 5500125000000552; PUBMED: 23553596]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox effectively reduces iron overload in non‐transfusion‐dependent thalassemia (NTDT) patients: 1‐year extension results from the THALASSA study. Annals of Hematology 2013;92(11):1485‐93. [CENTRAL: 876359; CRS: 5500125000000553; PUBMED: 23775581]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Deferasirox significantly reduces liver iron concentration in non‐transfusion‐dependent thalassemia patients with iron overload: results from the 1‐year randomized, double‐blind, placebo‐controlled phase II THALASSA study. Blood 2011;118(21):412‐3. [CENTRAL: 977518; CRS: 5500125000000549]CENTRAL
Taher AT, Porter JB, Viprakasit V, Kattamis A, Chuncharunee S, Sutcharitchan P, et al. Defining serum ferritin thresholds to predict clinically relevant liver iron concentrations for guiding deferasirox therapy when MRI is unavailable in patients with non‐transfusion‐dependent thalassaemia. British Journal of Haematology 2015;168(2):284‐90. CENTRAL
Taher AT, Temraz S, Cappellini MD. Deferasirox for the treatment of iron overload in non‐transfusion‐dependent thalassemia. Expert Review of Hematology 2013;6(5):495‐509. [CENTRAL: 977520; CRS: 5500125000000554; PUBMED: 24083402]CENTRAL

Referencias de los estudios excluidos de esta revisión

Economou 2010 {published data only}

Economou M, Printza N, Teli A, Tzimouli V, Tsatra I, Papachristou F, et al. Renal dysfunction in patients with beta‐thalassemia major receiving iron chelation therapy either with deferoxamine and deferiprone or with deferasirox. Acta Haematologica 2010;123(3):148‐52. CENTRAL

EPIC 2008 {published data only}

Cappellini MD, El‐Beshlawy A, Porter JB, Kattamis A, Taylor K, Rose C, et al. Concomitant medications and gastrointestinal events in thalassemia and MDS patients receiving deferasirox for transfusional iron overload: data from the EPIC study. Blood 2012;120(21):5182. CENTRAL
Cappellini MD, Porter J, El‐Beshlawy A, Li CK, Seymour JF, Elalfy M, et al. Tailoring iron chelation by iron intake and serum ferritin: the prospective EPIC study of deferasirox in 1744 patients with transfusion‐dependent anemias. Haematologica 2010;95(4):557‐66. [PUBMED: 19951979]CENTRAL
Cilloni D, Messa E, Biale L, Bonferroni M, Salvi F, Lunghi M, et al. High rate of erythroid response during iron chelation therapy in a cohort of 105 patients affected by hematologic malignancies with transfusional iron overload: an Italian multicenter retrospective study. Blood 2011;118(21):280‐1. CENTRAL
El‐Beshlawy A, Elalfy M, Chan LL, Lai Y, Lin KH, Aydinok Y, et al. Deferasirox treatment for up to 3 years in iron‐overloaded pediatric patients reduces serum ferritin with a manageable safety profile. Blood 2012;120(21):1028. CENTRAL
Lai YR, Liu RR, Li CF, Huang SL, Li Q, Habr D, et al. Efficacy of deferasirox for the treatment of iron overload in Chinese thalassaemia major patients: results from a prospective, open‐label, multicentre clinical trial. Transfusion Medicine 2013;23(6):389‐96. CENTRAL
Pennell DJ, Porter JB, Cappellini MD, Chan LL, El‐Beshlawy A, Aydinok Y, et al. Deferasirox for up to 3 years leads to continued improvement of myocardial T2 in patients with beta‐thalassemia major. Haematologica 2012;97(6):842‐8. CENTRAL
Pennell DJ, Porter JB, Cappellini MD, Chan LL, El‐Beshlawy A, Aydinok Y, et al. Continued improvement in myocardial T2(star) over two years of deferasirox therapy in beta‐thalassemia major patients with cardiac iron overload. Haematologica 2011;96(1):48‐54. CENTRAL
Porter J, Bowden DK, Economou M, Troncy J, Ganser A, Habr D, et al. Health‐related quality of life, treatment satisfaction, adherence and persistence in beta‐thalassemia and myelodysplastic syndrome patients with iron overload receiving deferasirox: results from the EPIC clinical trial. Anemia 2012 Aug 12 [Epub]. [DOI: 10.1155/2012/297641; Article ID: 297641]CENTRAL
Porter JB, Elalfy M, Viprakasit V, Giraudier S, Chan LL, Lai Y, et al. Serum ferritin, labile plasma iron and transferrin saturation: comparison between underlying anemias with transfusional iron overload before and after treatment with deferasirox. Blood 2012;120(21):2126. CENTRAL
Viprakasit V, Ibrahim H, Ha SY, Ho PJ, Li CK, Chan LL, et al. Clinical efficacy and safety evaluation of tailoring iron chelation practice in thalassaemia patients from Asia‐Pacific: a subanalysis of the EPIC study of deferasirox. International Journal of Hematology 2011;93(3):319‐28. CENTRAL

Erdogan 2013 {published data only}

Erdogan E, Canatan D, Ormeci AR, Vural H, Aylak F. The effects of chelators on zinc levels in patients with thalassemia major. Journal of Trace Elements in Medicine and Biology 2013;27(2):109‐11. CENTRAL

ESCALATOR 2005 {published data only}

Daar S, Pathare A, Nick H, Kriemler‐Krahn U, Hmissi A, Habr D, et al. Reduction in labile plasma iron during treatment with deferasirox, a once‐daily oral iron chelator, in heavily iron‐overloaded patients with beta‐thalassaemia. European Journal of Haematology 2009;82(6):454‐7. [PUBMED: 19191863]CENTRAL
Pathare A, Taher A, Daar S. Deferasirox (Exjade) significantly improves cardiac T2* in heavily iron‐overloaded patients with beta‐thalassemia major. Annals of Hematology 2010;89(4):405‐9. [PUBMED: 19798501]CENTRAL
Taher A, Al Jefri A, Elalfy MS, Al Zir K, Daar S, Rofail D, et al. Improved treatment satisfaction and convenience with deferasirox in iron‐overloaded patients with beta‐Thalassemia: Results from the ESCALATOR Trial. Acta Haematologica 2010;123(4):220‐5. [PUBMED: 20424435]CENTRAL
Taher A, El‐Beshlawy A, Elalfy MS, Al Zir K, Daar S, Habr D, et al. Efficacy and safety of deferasirox, an oral iron chelator, in heavily iron‐overloaded patients with beta‐thalassaemia: the ESCALATOR study. European Journal of Haematology 2009;82(6):458‐65. [PUBMED: 19187278]CENTRAL
Taher A, Elalfy MS, Al Zir K, Daar S, Al Jefri A, Habr D, et al. Importance of optimal dosing > 30 mg/kg/d during deferasirox treatment: 2.7‐yr follow‐up from the ESCALATOR study in patients with beta‐thalassaemia. European Journal of Haematology 2011;87(4):355‐65. CENTRAL

Fernandes 2013 {published data only}

Fernandes JL, Sampaio EF, Fertrin K, Coelho OR, Loggetto S, Piga A, et al. Amlodipine reduces cardiac iron overload in patients with thalassemia major: a pilot trial. American Journal of Medicine 2013;126(9):834‐7. CENTRAL

Gao 2011 {published data only}

Gao HY, Li Q, Chen JJ, Chen GF, Li CG. Curative effects and safety of deferasirox in treatment of iron overload in children with beta‐thalassemia major. Zhongguo Dang Dai Er Ke Za Zhi[Chinese Journal of Contemporary Pediatrics] 13;7:531‐4. CENTRAL

Garadah 2011 {published data only}

Garadah TS, Mahdi N, Kassab S, Abu‐Taleb A, Shoroqi I, Alawadi AH. The impact of two different doses of chelating therapy (deferasirox) on echocardiographic tissue Doppler indices in patients with thalassemia major. European Journal of Haematology 2011;87(3):267‐73. CENTRAL

Genc 2015 {published data only}

Genc GE, Ozturk Z, Gumuslu S, Kupesiz A. Mineral levels in thalassaemia major patients using different iron chelators. Biological Trace Element Research 2016;170(1):9‐16. CENTRAL

Gomber 2016 {published data only}

Gomber S, Jain P, Sharma S, Narang M. Comparative Efficacy and Safety of Oral Iron Chelators and their Novel Combination in Children with Thalassemia. Indian Pediatrics 2016;53(3):207‐10. [CRS: 5500135000001569; PUBMED: 27029681]CENTRAL

Grady 2012 {published data only}

Grady RW, Galanello R, Randolph RE, Kleinert DA, Dessi C, Giardina PJ. Toward optimizing the use of deferasirox: potential benefits of combined use with deferoxamine. Haematologica 2013;98(1):129‐35. CENTRAL

Hagag 2013 {published data only}

Hagag AA, Elfrargy MS, Gazar RA, El‐Lateef AE. Therapeutic value of combined therapy with deferasirox and silymarin on iron overload in children with Beta thalassemia. Mediterranean Journal of Hematology & Infectious Diseases 2013;5(1):e2013065. CENTRAL

Hesham 2014 {published data only}

Hesham MA, Elsafy UR, Besher MR, Fathy MM, Gerges GS. Neutrophil gelatinase‐associated lipocalin (NGAL) level in beta thalassemia patients. Haematologica 2014;99 Suppl:740. CENTRAL

Inati 2011 {published data only}

Inati A, Khoriaty E, Sbeiti N, Koussa S, Abi Nasr T, Musallam K, Taher A. A prospective randomized trial comparing phlebotomy and deferasirox for the treatment of iron overload in paediatric thalassemia major patients cured by stem cell transplantation: 6‐month follow‐up. Haematologica 2011;96 Suppl 2:178. [Abstract no: 0428]CENTRAL
Inati A, Sbeiti N, Khoriaty E, Cappellini MD, Koussa S, Nasr TA, et al. 1‐year results from a prospective randomised trial comparing phlebotomy with deferasirox for the treatment of iron overload in pediatric patients with thalassemia major following curative stem cell transplantation. Blood 2011;118(21):413‐4. [CENTRAL: 977600; CRS: 5500125000000568]CENTRAL
Inati A, Sbeiti N, Taher AT, Koussa S, Pierre TG. Relationship between total iron removed by phlebotomy and iron removed from the liver in pediatric thalassemia major patients following curative stem cell transplant. Blood. 2011; Vol. 118, issue 21. [Abstract no: 5300]CENTRAL
Inati AC, Khoriaty E, Sbeiti N, St Pierre TG, Koussa S, Musallam KM, et al. Iron overload indices in thalassemia major children cured by stem cell transplantation at enrollment in a prospective randomized trial comparing phlebotomy and deferasirox. Blood 2010;116(21):864. CENTRAL

Kallistheni 2012 {published data only}

Kallistheni F, Berdoukas V, Chouliaras G, Pappa C, Cabantchik ZI, Tzoumari I. Evaluating toxic free iron and options for keeping it at bay in iron overloaded patients. Blood 2012;120(21):2021. CENTRAL

Karakukcu 2012 {published data only}

Karakukcu C, Karakukcu M, Unal E, Patiroglu T, Ozdemir MA, Torun YA, et al. Coenzyme Q10 levels in β‐thalassemia and its association with ferritin levels and chelation therapy. Hemoglobin 2012;36(3):219‐29. CENTRAL

Keikhaei 2011 {published data only}

Keikhaei B. Combined and alternative iron chelator drugs in treatment of thalassemia major. Pakistan Journal of Medical Sciences 2011;27(3):630‐3. CENTRAL

Lu 2015 {published data only}

Lu MY, Wang N, Wu WH, Lai CW, Kuo PH, Chiang PH, et al. Simultaneous determination of plasma deferasirox and deferasirox‐iron complex using an HPLC‐UV system and pharmacokinetics of deferasirox in patients with beta‐thalassemia major: once‐daily versus twice‐daily administration. Clinical Therapeutics 2015;37(8):1751‐60. CENTRAL

Medrano Engay 2013 {published data only}

Medrano Engay B, Irun MP, Sarria L, Andrade M, Murillo I, Montes A, et al. Analysis of efficacy and safety of two iron chelators in patients with iron overload (QueLaFer study). Leukemia Research 2013;37:S105. CENTRAL
Medrano‐Engay B, Irun P, Sarria L, Andrade M, Murillo I, Montes A, et al. Evaluation of quality of life and analysis of efficacy and safety of two iron chelators in patients with iron overload. Haematologica 2013;98:704. CENTRAL

Ozturk 2015 {published data only}

Ozturk Z, Genc GE, Kupesiz A, Kurtoglu E, Gumuslu S. Thalassemia major patients using iron chelators showed a reduced plasma thioredoxin level and reduced thioredoxin reductase activity, despite elevated oxidative stress. Free Radical Research 2015;49(3):309‐16. CENTRAL

Pakakasama 2011 {published data only}

Pakakasama S, Sirireung A, Thinkhamrop B, Sirachainan N, Hongeng S. Efficacy of deferasirox on iron chelation in thalassemia patients after hematopoietic stem cell transplantation. Blood 2011;118(21):1355. CENTRAL

Pepe 2010 {published data only}

Pepe A, Rossi G, Meloni A, Dell'Amico MC, Spasiano A, Capra M, et al. A T2*MRI prospective survey on heart and liver iron in thalassemia major patients treated with deferasirox versus deferiprone and desferrioxamine In monotherapy. Blood 2010;116(21):1731‐2. CENTRAL

Pepe 2011 {published data only}

Pepe A, Meloni A, Rossi G, D'Ascola DG, Capra M, Filosa A, et al. A MRI prospective survey on cardiac and hepatic iron and cardiac function in thalassemia major patients treated with sequential deferiprone‐desferrioxamine versus deferasirox. Blood 2011;118(21):499‐500. CENTRAL

Pileri 2014 {published data only}

Pileri F, Vegetti A, De Pietri L, Abbati G, Ventura P, Gamberini MR, et al. Randomized, open label, phase IIa controlled trials evaluating the safety of the association of deferasirox with standard antiviral therapy (Peg‐interferon and Ribavirin) in patients with chronic hepatitis C. Blood 2014;124(21):4075. CENTRAL

Song 2014 {published data only}

Song TS, Hsieh YW, Peng CT, Chen TL, Lee HZ, Chung JG, et al. Combined versus monotherapy or concurrent therapy for treatment of thalassaemia. In Vivo 2014;28(4):645‐9. CENTRAL

Torcharus 2011 {published data only}

Torcharus K, Pankaew T. Health‐related quality of life in Thai thalassemic children treated with iron chelation. Southeast Asian Journal of Tropical Medicine & Public Health 2011;42(4):951‐9. CENTRAL

Walter 2012 {published data only}

Walter PB, Harmatz P, Higa A, Ng V, Weyhmiller MG, Evans P, et al. Innate immune cell expression of pattern recognition receptors from beta‐thalassemia patients during intensive combination chelation therapy. Blood 2012;120(21):1025. CENTRAL

Referencias de los estudios en espera de evaluación

Ansari 2015 {published data only}

Ansari S, Miri Ali Abadi G, Azarkeivan A. Comparison of iron chelation effects of deferoxamine, deferasirox, and combination of deferoxamine and deferiprone on liver and cardiac T2* MRI in thalassemia major [abstract]. Haematologica 2015;100 Suppl 1:595, Abstract no: E1484. [CENTRAL: 1098922; CRS: 5500135000001406]CENTRAL

EUCTR2010‐023217‐61‐GB {unpublished data only}

 

Hagag 2015 {published data only}

Hagag AA, Hamam MA, Taha OA, Hazaa SM. Therapeutic efficacy of different iron chelators in Egyptian children with Beta Thalassemia with iron overload. Infectious Disorders Drug Targets 2015;15(2):98‐105. CENTRAL

IRCT201110087677N1 {unpublished data only}

 

Kakkar 2015 {published data only}

Kakkar S, Sobti PPC, Kalra A. Efficacy and safety of deferasirox in single vs divided dosage in Thalassemic children. Haematologica 2015;100 Suppl 1 785:PB2010. CENTRAL

NCT02198508 {unpublished data only}

 

Cutino 2009 {unpublished data only}

Sequential DFX‐DFP versus DFX or DFP multicentre randomised study. Ongoing study Date of first enrolment: 27/01/2010.

DEEP‐2 2012 {unpublished data only}

Efficacy and safety study to compare deferiprone versus deferasirox in paediatric patients. Ongoing study Date of first enrolment: 29/11/2012.

NCT02125877 {unpublished data only}

Phase II Study to Investigate the Benefits of an Improved Deferasirox Formulation (Film‐coated Tablet). Ongoing studyJuly 2014.

NCT02435212 {unpublished data only}

Study to Evaluate Treatment Compliance, Efficacy and Safety of an Improved Deferasirox Formulation (Granules) in Pediatric Patients (2 ‐ < 18 years old) With Iron Overload. Ongoing studySeptember 2015.

Angastiniotis 1998

Angastiniotis M, Modell B. Global epidemiology of hemoglobin disorders. Annals of the New York Academy of Sciences 1998;850:251‐69.

Angelucci 2008

Angelucci E, Barosi G, Camaschella C, Cappellini MD, Cazzola M, Galanello R, et al. Italian Society of Hematology practice guidelines for the management of iron overload in thalassemia major and related disorders. Haematologica 2008;93(5):741‐52. [PUBMED: 18413891]

Borgna‐Pignatti 2004

Borgna‐Pignatti C, Rugolotto S, De Stefano P, Zhao H, Cappellini MD, Del Vecchio GC, et al. Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine. Haematologica 2004;89(10):1187‐93.

Borgna‐Pignatti 2005

Borgna‐Pignatti C, Cappellini MD, De Stefano P, Del Vecchio GC, Forni GL, Gamberini MR, et al. Survival and complications in thalassemia. Annals of the New York Academy of Sciences 2005;1054:40‐7.

Brittenham 1994

Brittenham GM, Griffith PM, Nienhuis AW, McLaren CE, Young NS, Tucker EE, et al. Efficacy of deferoxamine in preventing complications of iron overload in patients with thalassemia major. New England Journal of Medicine 1994;331(9):567‐73.

Cappellini 2005a

Cappellini MD. Overcoming the challenge of patient compliance with iron chelation therapy. Seminars in Hematology 2005;42(2 Suppl 1):S19‐21.

Cappellini 2006

Cappellini MD, Cohen A, Piga A, Bejaoui M, Perrotta S, Agaoglu L, et al. A phase 3 study of deferasirox (ICL670), a once‐daily oral iron chelator, in patients with beta‐thalassemia. Blood 2006;107(9):3455‐62.

Cappellini 2007

Cappellini MD, Bejaoui M, Agaoglu L, Porter J, Coates T, Jeng M, et al. Prospective evaluation of patient‐reported outcomes during treatment with deferasirox or deferoxamine for iron overload in patients with beta‐thalassemia. Clinical Therapeutics 2007;29(5):909‐17.

Cappellini 2008

Cappellini MD. Long‐term efficacy and safety of deferasirox. Blood Reviews 2008;22 Suppl 2:S35‐41. [PUBMED: 19059055]

Cappellini 2009

Cappellini MD, Pattoneri P. Oral iron chelators. Annual Review of Medicine 2009;60:25‐38. [PUBMED: 19630568]

Carpenter 2011

Carpenter, John‐Paul, et al. On T2* magnetic resonance and cardiac iron. Circulation 2011;123:1519‐1528.

Ceci 2002

Ceci A, Baiardi P, Felisi M, Cappellini MD, Carnelli V, De Sanctis V, et al. The safety and effectiveness of deferiprone in a large‐scale, 3‐year study in Italian patients. British Journal of Haematology 2002;118(1):330‐6.

Chalmers 2016

Chalmers AW, Shammo JM. Evaluation of a new tablet formulation of deferasirox to reduce chronic iron overload after long‐term blood transfusions. Ther Clin Risk Manag. 2016;12:201‐8.

Chirnomas 2009

Chirnomas D, Smith AL, Braunstein J, Finkelstein Y, Pereira L, Bergmann AK, et al. Deferasirox pharmacokinetics in patients with adequate versus inadequate response. Blood 2009;114(19):4009‐13. [PUBMED: 19724055]

Cooley 1925

Cooley TB, Lee P. A series of cases of splenomegaly in children with anemia and peculiar bone changes. Transactions of the American Pediatric Society 1925;37:29‐30.

Curtin 2002a

Curtin F, Altman DG, Elbourne D. Meta‐analysis combining parallel and cross‐over clinical trials. I: Continuous outcomes. Statistics in Medicine 2002;21(15):2131‐44.

Curtin 2002b

Curtin F, Elbourne D, Altman DG. Meta‐analysis combining parallel and cross‐over clinical trials. II: Binary outcomes. Statistics in Medicine 2002;21(15):2145‐59.

Curtin 2002c

Curtin F, Elbourne D, Altman DG. Meta‐analysis combining parallel and cross‐over clinical trials. III: The issue of carry‐over. Statistics in Medicine 2002;21(15):2161‐73.

Davis 2004

Davis BA, O'Sullivan C, Jarritt PH, Porter JB. Value of sequential monitoring of left ventricular ejection fraction in the management of thalassemia major. Blood 2004;104(1):263‐9. [PUBMED: 15001468]

Delea 2008

Delea TE, Hagiwara M, Thomas SK, Baladi JF, Phatak PD, Coates TD. Outcomes, utilization, and costs among thalassemia and sickle cell disease patients receiving deferoxamine therapy in the United States. American Journal of Hematology 2008;83(4):263‐70.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JP, Curtin F, Worthington HV, Vail A. Meta‐analyses involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140‐9.

EMA 2016

European Medicines Agency (EMA). Exjade: EPAR. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Product_Information/human/000670/WC500033925.pdf (accessed 2 January 2017).

EMEA 2007

European Medicines Agency, London, UK. Exjade: European Public Assessment Report: Summary of Product Characteristics. www.emea.europa.eu/humandocs/Humans/EPAR/exjade/exjade.htm (accessed 14 February 2008).

Farmaki 2006

Farmaki K, Angelopoulos N, Anagnostopoulos G, Gotsis E, Rombopoulos G, Tolis G. Effect of enhanced iron chelation therapy on glucose metabolism in patients with beta‐thalassaemia major. British Journal of Haematology 2006;134(4):438‐44.

FDA 2005

Food, Drug Administration (FDA). USA. Exjade. http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021882s010lbl.pdf (accessed 02 September 2011).

FDA 2013

Food, Drug Administration (FDA). USA. Exjade (deferasirox) Prescribing Information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021882s019lbl.pdf (accessed 02 January 2017).

FDA Boxed Warning 2010

US Food, Drug Administraton. Exjade (Deferasirox): Boxed Warning. http://www.fda.gov/safety/medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm200850.htm (accessed 02 September 2011).

Fisher 2013a

Fisher SA, Brunskill S, Doree C, Gooding S, Chowdhury O, Roberts DJ. Desferrioxamine mesylate for managing transfusional iron overload in people with transfusion‐dependent thalassaemia. Cochrane Database of Systematic Reviews 2013, Issue 8. [DOI: 10.1002/14651858.CD004450.pub2]

Fisher 2013b

Fisher SA, Brunskill SJ, Doree C, Chowdhury O, Gooding S, Roberts DJ. Oral deferiprone for iron chelation in people with thalassemia. Cochrane Database of Systematic Reviews 2013, Issue 8. [DOI: 10.1002/14651858.CD004839.pub3]

Freedman 1990

Freedman MH, Grisaru D, Olivieri N, MacLusky I, Thorner PS. Pulmonary syndrome in patients with thalassemia major receiving intravenous deferoxamine infusions. American Journal of Diseases of Children 1990;144(5):565‐9.

Gabutti 1996

Gabutti V, Piga A. Results of long‐term iron‐chelating therapy. Acta Haematologica 1996;95(1):26‐36.

Galanello 2006

Galanello R, Kattamis A, Piga A, Fischer R, Leoni G, Ladis V, et al. A prospective randomized controlled trial on the safety and efficacy of alternating deferoxamine and deferiprone in the treatment of iron overload in patients with thalassemia. Haematologica 2006;91(9):1241‐3.

GRADE

Guyatt GH, Oxman AD, Vist G, Kunz R, Falck‐Ytter Y, Alonso‐Coello P, et al. Rating quality of evidence and strength of recommendations GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336(7650):924‐6.

GRADEpro GDT 2015 [Computer program]

GRADE Working Group, McMaster University. GRADEpro GDTool. Hamilton (ON): GRADE Working Group, McMaster University, (accessed prior to 22 August 2016).

Grange 2010

Grange S, Bertrand DM, Guerrot D, Eas F, Godin M. Acute renal failure and Fanconi syndrome due to deferasirox. Nephrology, dialysis, transplantation 2010;25(7):2376‐8. [PUBMED: 20466673]

Higgins 2002

Higgins JP, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21(11):1539‐58.

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60.

Higgins 2011

Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011a

Higgins JPT, Altman DG, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011b

Higgins JPT, Deeks JJ, Altman DG, editor(s). Chapter 16: Special topics in statistics. In: Higgins JP, Green S, editors(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org. The Cochrane Collaboration, 201.

Hoffbrand 1989

Hoffbrand AV, Bartlett AN, Veys PA, O'Connor NT, Kontoghiorghes GJ. Agranulocytosis and thrombocytopenia in patient with Blackfan‐Diamond anaemia during oral chelator trial. Lancet 1989;2(8660):457.

Kattamis 2003

Kattamis A, Kassou C, Berdousi H, Ladis V, Papassotiriou I, Kattamis C. Combined therapy with desferrioxamine and deferiprone in thalassemic patients: effect on urinary iron excretion. Haematologica 2003;88(12):1423‐25.

Kolnagou 2008

Kolnagou A, Economides C, Eracleous E, Kontoghiorghes GJ. Long term comparative studies in thalassemia patients treated with deferoxamine or a deferoxamine/deferiprone combination. Identification of effective chelation therapy protocols. Hemoglobin 2008;32(1‐2):41‐7.

Kontoghiorghes 1987a

Kontoghiorghes GJ, Aldouri MA, Hoffbrand AV, Barr J, Wonke B, Kourouclaris T, et al. Effective chelation of iron in beta thalassaemia with the oral chelator 1,2‐dimethyl‐3‐hydroxypyrid‐4‐one. BMJ (Clinical Research Edition) 1987;295(6612):1509‐12.

Kontoghiorghes 1987b

Kontoghiorghes GJ, Aldouri MA, Sheppard L, Hoffbrand AV. 1,2‐Dimethyl‐3‐hydroxypyrid‐4‐one, an orally active chelator for treatment of iron overload. Lancet 1987;1(8545):1294‐95.

Koren 1991

Koren G, Kochavi‐Atiya Y, Bentur Y, Olivieri NF. The effects of subcutaneous deferoxamine administration on renal function in thalassemia major. International Journal of Hematology 1991;54(5):371‐5.

Kushner 2001

Kushner JP, Porter JP, Olivieri NF. Secondary iron overload. Hematology / the Education Program of the American Society of Hematology 2001;1:47‐61. [DOI: 10.1182/asheducation‐2001.1.47]

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies.. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011.

Lindsey 2007

Lindsey WT, Olin BR. Deferasirox for transfusion‐related iron overload: a clinical review. Clinical Therapeutics 2007;29(10):2154‐66. [PUBMED: 18042472]

Maggio 2002

Maggio A, D'Amico G, Morabito A, Capra M, Ciaccio C, Cianciulli P, et al. Deferiprone versus deferoxamine in patients with thalassemia major: a randomized clinical trial. Blood Cells, Molecules and Diseases 2002;28(2):196‐208.

Maggio 2011

Maggio A, Filosa A, Vitrano A, Aloj G, Kattamis A, Ceci A, et al. Iron chelation therapy in thalassemia major: a systematic review with meta‐analyses of 1520 patients included on randomized clinical trials. Blood Cells, Molecules & Diseases 2011;47(3):166‐75.

McLeod 2009

McLeod C, Fleeman N, Kirkham J, Bagust A, Boland A, Chu P, et al. Deferasirox for the treatment of iron overload associated with regular blood transfusions (transfusional haemosiderosis) in patients suffering with chronic anaemia: a systematic review and economic evaluation. Health Technology Assessment (Winchester, England) 2009;13(1):iii‐iv, ix‐xi, 1‐121. [PUBMED: 19068191]

Meerpohl 2014

Meerpohl Joerg J, Schell Lisa K, Rücker G, Fleeman N, Motschall E, Niemeyer Charlotte M, et al. Deferasirox for managing iron overload in people with myelodysplastic syndrome. Cochrane Database of Systematic Reviews. John Wiley & Sons, Ltd, 2014, issue 10. [DOI: 10.1002/14651858.CD007461.pub3; CD007461]

Meerpohl 2014a

Meerpohl Joerg J, Schell Lisa K, Rücker G, Motschall E, Fleeman N, Niemeyer Charlotte M, et al. Deferasirox for managing transfusional iron overload in people with sickle cell disease. Cochrane Database of Systematic Reviews. John Wiley & Sons, Ltd, 2014, issue 5. [DOI: 10.1002/14651858.CD007477.pub3; CD007477]

Modell 2000

Modell B, Khan M, Darlison M. Survival in beta‐thalassaemia major in the UK: data from the UK Thalassaemia Register. Lancet 2000;355(9220):2051‐2.

Modell 2008

Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bulletin of the World Health Organization 2008;86(6):480‐7.

Nisbet‐Brown 2003

Nisbet‐Brown E, Olivieri NF, Giardina PJ, Grady RW, Neufeld EJ, Sechaud R, et al. Effectiveness and safety of ICL670 in iron‐loaded patients with thalassaemia: a randomised, double‐blind, placebo‐controlled, dose‐escalation trial. Lancet 2003;361(9369):1597‐602.

Novartis 2015

Novartis. Novartis announces FDA approval for JadenuTM to simplify treatment administration for patients with chronic iron overload. https://www.novartis.com/news/media‐releases/novartis‐announces‐fda‐approval‐jadenutm‐simplify‐treatment‐administration.

Olivieri 1994

Olivieri NF, Nathan DG, MacMillan JH, Wayne AS, Liu PP, McGee A, et al. Survival in medically treated patients with homozygous beta‐thalassemia. New England Journal of Medicine 1994;331(9):574‐8.

Olivieri 1997

Olivieri NF, Brittenham GM. Iron‐chelating therapy and the treatment of thalassemia. Blood 1997;89(3):739‐61.

Olivieri 1998

Olivieri NF, Brittenham GM, McLaren CE, Templeton DM, Cameron RG, McClelland RA, et al. Long‐term safety and effectiveness of iron‐chelation therapy with deferiprone for thalassemia major. New England Journal of Medicine 1998;339(7):417‐23.

Olivieri 1999

Olivieri NF. The beta‐thalassemias. New England Journal of Medicine 1999;341(2):99‐109.

Origa 2005

Origa R, Bina P, Agus A, Crobu G, Defraia E, Dessi C, et al. Combined therapy with deferiprone and desferrioxamine in thalassemia major. Haematologica 2005;90(10):1309‐14.

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34.

Peng 2008

Peng CT, Tsai CH, Wu KH. Effects of chelation therapy on cardiac function improvement in thalassemia patients: literature review and the Taiwanese experience. Hemoglobin 2008;32(1‐2):49‐62.

Peters 2013

Peters M, Heijboer H, Smiers F, Giordano PC. Diagnosis and management of thalassaemia. BMJ 2012;25(344):e228.

Porter 2009

Porter JB. Deferasirox: an update. Hemoglobin 2009;33 Suppl 1:S70‐5. [PUBMED: 20001635]

Rafat 2009

Rafat C, Fakhouri F, Ribeil JA, Delarue R, Le Quintrec M. Fanconi syndrome due to deferasirox. American Journal of Kidney Diseases 2009;54(5):931‐4. [PUBMED: 19493602]

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Richer 2005

Richer J, Chudley AE. The hemoglobinopathies and malaria. Clinical Genetics 2005;68(4):332‐6.

Rund 2005

Rund D, Rachmilewitz E. Beta‐thalassemia. New England Journal of Medicine 2005;353(11):1135‐46.

SIGN 2008

Scottish Intercollegiate Guidelines Network. A guideline developer's handbook. Guideline No. 50. http://www.sign.ac.uk/guidelines/fulltext/50/index.html (accessed 02 September 2011).

Taher 2006

Taher A, Isma'eel H, Cappellini MD. Thalassemia intermedia: revisited. Blood Cells, Molecules & Diseases 2006;37(1):12‐20.

Taher 2009

Taher A, Cappellini MD, Vichinsky E, Galanello R, Piga A, Lawniczek T, et al. Efficacy and safety of deferasirox doses of >30 mg/kg per d in patients with transfusion‐dependent anaemia and iron overload. British Journal of Haematology 2009;147(5):752‐9.

Taher 2010

Taher A, Al Jefri A, Elalfy MS, Al Zir K, Daar S, Rofail D, et al. Improved treatment satisfaction and convenience with deferasirox in iron‐overloaded patients with beta‐Thalassemia: Results from the ESCALATOR Trial. Acta Haematologica 2010;123(4):220‐5. [PUBMED: 20424435]

Tanner 2007

Tanner MA, Galanello R, Dessi C, Smith GC, Westwood MA, Agus A, et al. A randomized, placebo‐controlled, double‐blind trial of the effect of combined therapy with deferoxamine and deferiprone on myocardial iron in thalassemia major using cardiovascular magnetic resonance. Circulation 2007;115(14):1876‐84.

Trachtenberg 2011

Trachtenberg F, Vichinsky E, Haines D, Pakbaz Z, Mednick L, Sobota A, et al. Iron chelation adherence to deferoxamine and deferasirox in thalassemia. American Journal of Hematology 2011;86(5):433‐6.

Vanorden 2006

Vanorden HE, Hagemann TM. Deferasirox‐‐an oral agent for chronic iron overload. Annals of Pharmacotherapy 2006;40(6):1110‐7.

Wan 2014

Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Medical Research Methodology 2014;19(14):135.

Wanless 2002

Wanless IR, Sweeney G, Dhillon AP, Guido M, Piga A, Galanello R, et al. Lack of progressive hepatic fibrosis during long‐term therapy with deferiprone in subjects with transfusion‐dependent beta‐thalassemia. Blood 2002;100(5):1566‐9.

Weatherall 1998

Weatherall DJ. Thalassemia in the next millennium. Keynote address. Annals of the New York Academy of Sciences1998; Vol. 850:1‐9.

Weatherall 2000

Weatherall DJ, Clegg JB. The Thalassemia Syndromes. 4th Edition. Oxford: Blackwell Sciences Ltd., 2000.

Williamson 2002

Williamson PR, Smith CT, Hutton JL, Marson AG. Aggregate data meta‐analysis with time‐to‐event outcomes. Statistics in Medicine 2002;21(22):3337‐51.

Witte 2004

Witte S, Victor N. Some problems with the investigation of noninferiority in meta‐analysis. Methods of Information in Medicine 2004;43(5):470‐4.

Wu 2006

Wu SF, Peng CT, Wu KH, Tsai CH. Liver fibrosis and iron levels during long‐term deferiprone treatment of thalassemia major patients. Hemoglobin 2006;30(2):215‐8.

Yew 2010

Yew CT, Talaulikar GS, Falk MC, Clayton P, D'Rozario J, Brown M. Acute interstitial nephritis secondary to deferasirox causing acute renal injury needing short‐term dialysis. Nephrology (Carlton, Vic.) 2010;15(3):377. [PUBMED: 20470310]

Zurlo 1989

Zurlo MG, De Stefano P, Borgna‐Pignatti C, Di Palma A, Piga A, Melevendi C, et al. Survival and causes of death in thalassaemia major. Lancet 1989;2(8653):27‐30.

Referencias de otras versiones publicadas de esta revisión

Meerpohl 2008

Meerpohl JJ, Antes G, Rücker G, McLeod C, Fleeman N, Niemeyer CM, Bassler D. Deferasirox for managing iron overload in people with thalassaemia. Cochrane Database of Systematic Reviews 2008, Issue 4. [DOI: 10.1002/14651858.CD007476]

Meerpohl 2012

Meerpohl JJ, Antes G, Rücker G, Fleeman N, Motschall E, Niemeyer CM, Bassler D. Deferasirox for managing iron overload in people with thalassaemia. Cochrane Database of Systematic Reviews 2012, Issue 2. [DOI: 10.1002/14651858.CD007476.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Cappellini 2005b

Methods

Open‐label, multinational, multicentre, randomised, phase III, non‐inferiority study.

Participants

586 β‐thalassaemia individuals

Age (mean (SD)): DFX: 17 (9.47) years; DFO: 17.3 (9.96) years

Gender (male/female): DFX: 140/156 DFO: 142/148

Setting and country: 65 centres (mentioned in full text publication) in 12 countries: Argentina, Belgium, Brazil, Canada, France, Germany, Greece, Italy, Tunisia, Turkey, UK, USA

Inclusion criteria

  • ≥ 2 years

  • β‐thalassaemia and chronic iron overload from blood transfusions

  • LIC ≥ 2 mg/g dry weight

  • ≥ 8 blood transfusions/year

  • Female participants: required to use double‐barrier contraception

Exclusion criteria

  • ALT > 250 U/L during year prior to enrolment

  • Chronic hepatitis B infection

  • Active hepatitis C infection

  • History of a positive HIV test

  • Serum creatinine above the ULN

  • Urinary protein/creatinine ratio > 0.5 mg/mg

  • Nephrotic syndrome

  • Uncontrolled systemic hypertension

  • Prolonged corrected QT interval

  • Systemic infection within the 10 days prior to entry

  • GI conditions preventing absorption of oral medication

  • Concomitant conditions preventing therapy with deferasirox or DFO

  • History of ocular toxicity related to iron chelation therapy

  • A poor response to DFO

  • Noncompliance with prescribed therapy

Follow‐up: 1 year

Interventions

2 groups

  • DFX (n = 296): once‐daily at the assigned dose as a suspension in water half an hour prior to breakfast 7 days a week

Protocol assigned dose, mg/kg, Average daily dose, mg/kg/day (mean (SD)):

‐ LIC ≤ 3 mg Fe/g dry weight: 5; 6.2 (1.6)

‐ LIC > 3 mg Fe/g dry weight ‐ 7 mg Fe/g dry weight: 10; 10.2 (1.2)

‐ LIC > 7 mg Fe/g dry weight ‐ 14 mg Fe/g dry weight: 20; 19.4 (1.7)

‐ LIC > 14 mg Fe/g dry weight: 30; 28.2 (3.5)

  • DFO (n = 290): Slow subcutaneous infusion using electronic infusion pumps over 8 ‐ 12 hours, 5 days a week

Protocol assigned dose, mg/kg, Average daily dose, mg/kg/day (mean (SD)):

‐ LIC ≤ 3 mg Fe/g dry weight: 20 ‐ 30; 33.9 (9.9)

‐ LIC > 3 mg Fe/g dry weight ‐ 7 mg Fe/g dry weight: 25 ‐ 35; 36.7 (9.2)

‐ LIC > 7 mg Fe/g dry weight ‐ 14 mg Fe/g dry weight: 35 ‐ 50; 42.4 (6.6)

‐ LIC > 14 mg Fe/g dry weight: ≥ 50; 51.6 (5.8)

‐ Exceptions were permitted to the number of days of administration (ranged 3 ‐ 7 days)

‐ DFO doses reported are normalized to administration for 5 days a week

Outcomes

  • Primary response criterion: maintenance or reduction of LIC:

Success criteria:

LIC at baseline (mg Fe/g dry weight): success, LIC value after 1 year (mg Fe/g dry weight); failure, LIC value after 1 year (mg Fe/g dry weight)

2 to less than 7: 1 to less than 7, less than 1 or at least 7

7 to less than 10: 1 to less than 7, less than 1 or at least 7

10 or more: decrease in LIC of at least 3, decrease in LIC below 3

  • CBC/differential

  • Electrolytes

  • Liver function tests

  • Trace element analysis

  • Urinary protein/creatinine

  • Serum ferritin

  • Fe

  • Transferrin

  • ECGs

  • Ophthalmologic and auditory examinations

  • Individuals < 16 years of age: assessment of growth rate (growth velocity) and sexual development

  • LIC (liver biopsy or SQUID)

  • Iron excretion‐intake ratio

Furthermore, mortality, discontinuations, willingness to continue treatment, time lost from normal activities due to treatment, satisfaction with treatment, dose adjustments and dose interruptions, convenience and AEs were reported.

Notes

Funding and conflict of interests

Study was supported in part by research funding from Novartis Pharma to some of the authors. Two authors have declared a financial interest in a company whose product was studied in the present work. Several of the authors are employed by Novartis Pharma.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details given with regard to sequence generation.

"Randomisation was stratified by age groups: 2 to younger than 12 years, 12 to younger than 18 years, and 18 years or older. After randomization, patients were assigned by the investigator to a dose dependent on their baseline LIC according to the algorithm noted in Table 2."

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

"Randomisation was stratified by age groups: 2 to younger than 12 years, 12 to younger than 18 years, and 18 years or older. After randomisation, patients were assigned by the investigator to a dose dependent on their baseline LIC according to the algorithm noted in Table 2."

Blinding (performance bias and detection bias)
All outcomes

High risk

This was an open‐label study.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No flowchart according to CONSORT available.

586 individuals were randomised of which 29 discontinued and 4 died. The primary efficacy population consists of 553 individuals. However, it is stated that 541 participants completed one year of therapy. It remains unclear, what happened to the remaining 12 participants.

"Most patients completed 1 year of therapy on this study: 541 (92.3%) of 586 underwent both baseline and 1‐year LIC assessments. Discontinuations were relatively similar in the groups receiving deferasirox (n = 17) and deferoxamine (n = 12)."

"The primary efficacy population in this study consisted of 553 patients with LIC evaluations at baseline and after 52 weeks and those who discontinued due to safety reasons (AE, abnormal laboratory value or test procedure result, or iron overload–related death)."

Selective reporting (reporting bias)

Unclear risk

See also "Outcomes" in "Characteristics of included studies" table Cappellini 2005b above. Not all time points nor all parameters (secondary: e.g. trace elements) reported. However, EOS primary results are reported and secondary as outlined in methods section. However, it remains unclear whether any others were measured.

Other bias

Low risk

No other risk of bias detected.

Chirico 2013

Methods

Prospective study with randomised groups in the last 2 years of study period.

Participants

72 β‐thalassaemia participants: 21 non‐transfusion‐dependent thalassaemia participants, 51 transfusion‐dependent thalassaemia participants

Randomised in the last 2 years of study: n = 37

Age (mean (SD)): DFO : 30.2 (7.3) years; DFP: 28.8 (8.9) years; DFX: 31.4 (7.4) years

Gender: not mentioned

Setting: Thalassaemia Unit, Department of Pediatrics, University of Messina

Country: Italy

Inclusion criteria: not mentioned

Exclusion criteria:

  • βT minor

  • Acute illness

  • Severe renal and liver disease

  • Heart failure or cardiomyopathy

  • Endocrine complications (e.g. diabetes mellitus, thyroid dysfunction or assuming hormonal therapy)

Follow‐up: whole study: 8 years; randomised phase: 2 years

Interventions

All participants received DFO monotherapy for 4 years or until the appearance of thyreopathy. Afterwards, individuals with thyreopathy received DFO (20 ‐ 40 mg/kg, 8 ‐ 12 hours, 2 ‐ 6 days/week) and DFP (daily oral administration 75 ‐ 100 mg/kg/day in 3 divided doses). Individuals without thyreopathy continued with DFO. After the end of 2 years, participants with a new diagnosis of thyreopathy started combined chelation therapy, whereas those without thyroid dysfunction were randomised in 3 arms for further 2 years.

  • DFX (Exjade) (n = 12)

  • DFP (n = 12)

  • DFO (n = 13)

Only results of participants randomised to the 3 monotherapy groups in the last phase of the study were included in this systematic review.

All transfusion‐dependent thalassaemia participants followed a standard treatment protocol and were regularly transfused with packed red cells every 3 weeks, with the aim of maintaining pre‐transfusion haemoglobin levels above 9 g/dL.

Outcomes

  • Thyroid disease

  • Serum ferritin

Furthermore, FT₃, FT₄, TSH, TGA, TPO, thyroid dysfunction (overt hypothyroidism: low FT₄ and/or FT₃, increased TSH levels; subclinical hypothyroidism: normal FT₄, FT₃ and increased TSH concentration (> 5 TSH ɥIU/mL)); central hypothyroidism (inappropriately low serum TSH concentration in the presence of subnormal serum T₄ and T₃ concentrations), thyroid volumes, lipid profile, blood pressure and metabolic parameters (in particular insulin resistance) were measured.

Notes

The authors declare that they have no conflict of interest.

The authors state that this research did not receive any specific grant from any funding agency in the public, commercial or not‐for‐profit sector.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"This latter group was randomised into three arms, based on the type of iron chelation [...]"

Not mentioned how random sequence was generated.

Allocation concealment (selection bias)

Unclear risk

Not details given with regard to allocation concealment.

Blinding (performance bias and detection bias)
All outcomes

High risk

"The patients, physicians, laboratory staff and the epidemiologist who analysed the data were not aware of the intervention of each group"

No placebo treatment is mentioned, so blinding of participants and physicians is unlikely due to different administration routes and frequencies of application.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Results of all randomised participants at end of study are reported.

Selective reporting (reporting bias)

High risk

No results reported for TSH, FT₃, FT₄, but likely included in definition for thyroid dysfunction.

No results reported for TGA, TPO, thyroid volumes, insulin resistance, lipid profile, blood pressure at the end of the randomised phase.

Other bias

Unclear risk

Baseline data for randomised patients not reported apart from serum ferritin, age, splenectomy rate and haemoglobin before transfusion.

Elalfy 2015a

Methods

Randomised, prospective study.

Participants

180 people with β‐thalassaemia major

Age: ≤ 18 years

Gender: not mentioned

Setting: regular attendants of the Hematology Clinic, Pediatric Hospital, Ain Shams University

Country: Egypt

Inclusion criteria

  • Moderately iron‐overloaded people with β‐thalassaemia major without clinical symptoms of cardiac dysfunction

  • Vitamin C deficiency

  • Serum ferritin > 1000 ‐ 2500 ng/mL

  • Cardiac T2* > 10 ms calculated as geometric mean

  • Ejection fraction > 56%

Exclusion criteria

  • Insulin‐dependent diabetes

  • LVEF ≤ 56%

  • Active hepatitis (serum transaminases > 5 times above ULN)

  • Renal impairment (serum creatinine > 2 times ULN),

  • Sepsis or active infection

  • Participation in a previous investigational drug study within the 30 days preceding screening

  • Patients with a known allergy to DFX, DFP, and DFO

Follow up: 1 year

Interventions

3 groups:

  • DFX (n = 60): 25 mg/kg/day

  • DFO (n = 60): subcutaneous 40 mg/kg/day (5 days a week)

  • DFP (n = 60): 75 mg/kg/day

‐ Each chelation group were further randomly divided into two subgroups according to vitamin C supplementation (n = 30 in each group): Oral vitamin C in the morning 100 mg daily

‐ Previous chelation therapy was withdrawn for 2 weeks before randomisation

‐ Patients consumed a low‐iron diet (11 ‐ 15 mg of iron/day) and standard vitamin C diet during the study

Outcomes

  • Serum ferritin

  • LIC

  • Cardiac MRI

  • AEs

Furthermore: transfusion index, haemoglobin, iron, total iron binding capacity, transferrin saturation, vitamin C, compliance.

Notes

The authors declare that they have no conflict of interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Drug administration was according to a predetermined schedule generated from random numbers in a 1:1:1 manner based on a computer‐generated randomisation sequence maintained within the investigational drug pharmacy[...]".

Allocation concealment (selection bias)

Low risk

"[...] with allocation concealment by opaque sequentially numbered sealed envelope".

Blinding (performance bias and detection bias)
All outcomes

High risk

No details given with regard to blinding. No placebo treatment is mentioned. Due to different administration routes, blinding is not likely.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"Five patients in DFO subgroup did not continue till the end of study because of poor compliance [...]"

No ITT analysis performed, but proportion of missing outcome data was regarded as too low to have a large impact on effect size.

Selective reporting (reporting bias)

High risk

"The same improvement was found when each chelation subgroup receiving vitamin C supplementation was compared separately with the subgroup without vitamin C (data not shown)".

Only summarized data for patients with vitamin C supplementation versus patients without vitamin C supplementation and data for all chelation groups with vitamin C supplementation were reported.

The outcome "occurrence of AE" is mentioned, but only serious AE related to iron chelators are reported.

Serum ferritin is reported as median and IQR, but no reason for this reporting style is mentioned.

Other bias

Unclear risk

Baseline data are not given for all treatment groups

Elalfy 2015b

Methods

Interventional prospective randomised open‐labelled study with blinded data management and data analyses.

Participants

96 people with β‐thalassaemia major were randomised.

Age (mean (SD)): DFX + DFP : 14.05 (2.21), DFP + DFO: 15.25 (2.31)

Gender (male/female): 62/34

Setting: Thalassemia Centers of Ain Shams University, Egypt and Sultan Qaboos University Hospital, Oman

Countries: Egypt and Oman

Inclusion criteria

  • β‐transfusion‐dependent thalassaemia

  • Aged 10 ‐ 18 years

  • Serum ferritin > 2500 µg/L on maximum tolerated dose of a single iron chelator with uptrend of serum ferritin over the last 12 months prior to the study

  • Participants with LIC more than 7 mg/g by MRI R2* and mean cardiac T2* less than 20 and more than 6 ms calculated as geometric mean without clinical symptoms of cardiac dysfunction (shortness of breath at rest of exertion, orthopnoea, exercise intolerance, lower extremity edema, arrhythmias). Adequacy of prior chelation was defined as taking > 75% of the calculated dose/month on maximum tolerated dose with upward ferritin trend. For DFX, this should be 40 mg/kg/day, for DFP 100 mg/kg/day and for DFO at least 40 ‐ 50 mg/kg.

Exclusion criteria

  • History of agranulocytosis

  • Clinically significant GI or renal disease

  • Clinical cardiac disease, or with LEVF < 50% on baseline echocardiography

  • Evidence of active hepatitis

  • Serum transaminase > 3 times above ULN

  • Renal impairment (serum creatinine > ULN)

  • Participation in a previous investigational drug study within the 30 d preceding screening

  • Individuals with a known allergy to DFX, DFP and DFO

Follow‐up: 1 year

Interventions

2 groups:

  • DFX + DFP (n = 48): DFP 75 mg/kg/day, divided into 2 doses taken orally at 8 a.m. and 3 p.m. combined with DFX 30 mg/kg/day taken orally at 10 p.m. for 7 days a week

  • DFP + DFO (n = 48): DFP 75 mg/kg/day divided into 2 doses taken orally at 8 a.m. and 3 p.m, for 7 days (with 6 ‐ 8 hour interval between the 2 doses) combined with DFO 40 mg/kg/day by subcutaneous infusion over 10 hours starting at 10 p.m. for 6 days a week

‐ Chelation therapy was withdrawn for 2 weeks before randomisation

‐ The patients consumed a low‐iron diet (11 ‐ 15 mg of iron per day) during the study

‐ The transfusion regimen aimed to maintain the patients pre‐transfusion haemoglobin ≥ 8.0 g/dL by receiving approximately 15 mL/kg packed red blood cells every 3 ‐ 4 week

Outcomes

  • Serum ferritin

  • LIC

  • Cardiac MRI

  • SAE

  • AE

  • Adherence

  • Satisfaction

  • QoL

  • Self‐reported adherence

  • Proportion of participants who never thought about stopping iron‐chelating therapy

Notes

The authors state that they have nothing to declare

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomisation sequence was based on a computer randomised list in permuted blocks of 10 with a 1:1 ratio, generated at both University of Ain Shams and Sultan Qaboos".

Allocation concealment (selection bias)

Low risk

"To ensure no allocation bias, treatment group was assigned by telephone contact from the coordinating centre in Ain Shams".

Blinding (performance bias and detection bias)
All outcomes

High risk

"[...] open‐labelled study with blinded data management and data analyses"

High risk of bias in particular of performance bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

" [...] all the included patients continued till the end of study with no patients were lost follow‐up".

Selective reporting (reporting bias)

High risk

Patient‐reported satisfaction, percentage of patient's with self reported adherence, proportion of patients who never thought about stopping iron‐chelating therapy and 18‐months‐follow‐up results were reported incompletely so that they cannot be entered in a meta‐analysis.

Serum creatinine, liver function, audiometric and ophthalmological assessment were conducted as described in a conference abstract, but no or only incomplete results are published.

Although not pre‐defined as an outcome, the authors describe that "change in mean LVEF after 1 yr was not different between the two treatment groups (data not shown)."

On clinicaltrials.gov, only change in serum ferritin and the number of patients developing adverse reactions are predefined as outcomes.

Other bias

Low risk

No other bias detected.

Galanello 1999

Methods

2‐period, randomised, double‐blind, placebo‐controlled, sequential parallel‐group design.

Participants

25 people with transfusion‐dependent β‐thalassaemia were randomised

Age (mean (SD)): 21.6 (3.3) years

Gender (male/female): 25/0

Setting and country: 2 centers in Italy

Country: Italy

Inclusion criteria:

  • White males

  • Age ≥ 18 years

  • Transfusion‐dependent β‐thalassaemia

  • Participants had serum ferritin values between ≥ 1500 ng/mL and ≤ 5000 ng/mL, as well as post‐transfusion haemoglobin levels of at least 13 ± 0.5 g/ dL

  • All patients had previously been treated with a mean daily dose of 20 to 50 mg/kg/day DFO for at least 4 weeks before screening.

Exclusion criteria:

  • History of systemic reactions to treatment with DFO

  • History of systemic disease

  • Any medical condition that might have significantly altered the absorption, distribution, metabolism, or excretion of the study drug

Follow‐up: safety: Up to 10 days post dose

Interventions

"Following a 16‐day run‐in period, 24 patients were allocated to one of three study groups, with each group consisting of 8 patients. Each group was administered two single oral doses of ICL670 at an interval of at least 7 weeks, first a lower dose and later a higher dose. Group 1 received 2.5 and 20 mg/kg, group 2 received 5 and 40 mg/kg, and group 3 received 10 and 80 mg/kg ICL670, in all cases given as an oral suspension of 100 mL prepared from dispersible tablets. Before proceeding to a higher dose, the safety and tolerability of the preceding dose had to be assessed as satisfactory. In each treatment period, 2 of 8 patients received placebo in such a way that a given patient did not receive placebo more than once. Patients went back to their usual deferoxamine therapy and transfusion scheme in the interval between study periods."

Outcomes

  • Urinary iron excretion

  • Serum iron

  • Transferrin

  • Safety assessment: physical examination, vital signs, ECG, audiometry, clinical laboratory evaluations, and AE monitoring.

  • Safety laboratory evaluations: hematology (including transferrin and serum iron), biochemistry (including routine renal and liver function parameters, zinc, copper, and vitamin C), special kidney function parameters (α‐glutathione‐S‐transferase, N‐acetyl‐β‐Dglucosaminidase, β2‐microglobulin, and retinol‐binding protein), urinalysis

Notes

Novartis involved in trial. No details given and no information available with regard to potential conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details given with regard to sequence generation. From information given in paper, unclear, whether randomisation took place both in group assignment and in allocating patients to placebo. Author confirmed that randomisation was used to allocate placebo.

"Randomization was used to assign both drug (all treatment groups) and placebo. Hope to have clarified."

Allocation concealment (selection bias)

Unclear risk

No details given with regards to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

Low risk

"The study employed a two‐period, randomised, double‐blind, placebo‐controlled, sequential parallel group design."

However, no definition of double‐blind. Unclear whether, e.g. outcome assessors and data analysts were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Not applicable. Data from all participants are presented.

Selective reporting (reporting bias)

Unclear risk

Only general information with regard to safety issues. No clear‐cut comparison of placebo vs verum groups. Unclear, whether other parameters were evaluated than those reported.

Other bias

Low risk

No other risk of bias detected.

Habibian 2014

Methods

Randomised clinical study

Participants

30 people with thalassaemia major

Age: not mentioned

Gender: not mentioned

Setting: Bahonar hospital of Karaj

Country: Iran

Inclusion criteria: not mentioned

Exclusion criteria: not mentioned

Follow‐up: 1 year

Interventions

2 groups:

  • DFX

  • DFO

Outcomes

Serum ferritin

Notes

The study was only reported in a conference abstract

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"In this randomized clinical trial [...]"

Not mentioned how random sequence generation was generated.

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No details given with regard to blinding.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not reported how many patients reached end of study.

Selective reporting (reporting bias)

High risk

No results were reported.

Other bias

Unclear risk

No baseline characteristics were reported.

Hassan 2016

Methods

Prospective randomised study

Participants

60 β‐thalassemia major participants

Age (mean (SD)): DFX group: 8.9 (2.2), DFO group: 9.7 (1.9)

Gender: 19 male, 41 female

Setting: Out‐patient paediatric hematology clinic of Al‐Hussein University Hospital, Al‐Azhar University, Cairo

Country: Egypt

Inclusion criteria:

  • ≥ 6 years

  • Serum ferritin > 1500 µg/L

  • Irregular subcutaneous DFO chelation therapy

Exclusion criteria:

  • Serum creatinine above the upper age‐related normal range

  • Significant proteinuria (urinary protein/creatinine ratio > 1.0 in a non‐first‐void urine sample at baseline)

  • Elevated ALT more than 3‐fold of the ULN

  • GI diseases,

  • Clinically relevant auditory or ocular toxicity (or both) related to iron chelation therapy

  • Cardiac disease

  • Serious AEs with DFO or DFX

  • Absolute neutrophilic count < 1500/mm³

  • Platelet count < 100,000/mm³

Follow‐up: 1 year

Interventions

Two groups:

  • DFX (n = 30): Orally single daily dose of 20 ‐ 40 mg/kg/day on an empty stomach after dissolution in water, apple juice, or orange juice; Starting dose of DFX was individualized based on the frequency of blood transfusions

  • DFO (n = 30): 20 ‐ 50 mg/kg/day via subcutaneous infusion over 8 ‐ 10 hours, 5 days per week

Outcomes

  • Serum ferritin < 1500 µg/L

  • Safety

Furthermore, serum ferritin, ALT, AST, blood urea, serum creatinine, neutrophilic and platelet counts and some AEs were reported

Notes

The authors state that there is no conflict of interest to be declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"[...] the patients were randomized in a 1:1 ratio based on permuted blocks [...]"

Not mentioned how random sequence generation was generated.

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No details given with regard to blinding.

Due to different administration routes, blinding is not likely.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"[...] no discontinuation of drugs or drop‐out of follow‐up occurred."

Selective reporting (reporting bias)

Low risk

No protocol available. All predefined outcomes in the method section were reported.

Other bias

Low risk

No other bias detected.

Kakkar 2014

Methods

Randomised controlled study.

Participants

40 thalassaemia major participants

Age: 5 ‐ 18 years

Gender: not mentioned

Setting: unclear

Country: unclear

Inclusion criteria: not mentioned

Exclusion criteria: not mentioned

Follow‐up: not mentioned

Interventions

Three groups:

  • DFP (n = 10): 75 ‐ 100 mg/kg/day

  • DFX (n = 10): 30 ‐ 40 mg/kg/day

  • Both drugs administered sequentially every alternate week (n = 20)

Outcomes

  • Cardiac MRI T2*

  • Liver MRI T2*

  • Serum ferritin

  • CBC

  • Liver enzymes

  • Renal function tests

Notes

The study was only reported in a conference abstract.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"[...] were randomised to three groups [...]"

Not mentioned how random sequence generation was generated.

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No placebo treatment mentioned. High risk of bias in particular of performance bias and outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not reported how many patients reached end of study.

Selective reporting (reporting bias)

High risk

Regarding cardiac MRI T2*, only baseline data and P value for DFP vs DFP+DFX at the end of the study was reported. For liver MRI T2* it is unclear whether values given are baseline or end of study data. Missing data for serum ferritin. CBC, liver enzymes and renal function tests. Only untoward side‐effects reported and only for group receiving combination therapy.

Other bias

Unclear risk

No baseline data reported.

Molavi 2013

Methods

Randomised controlled open‐label study.

Participants

138 patients with β‐thalassemia major (n = 122) and thalassaemia intermedia (n = 16)

Age (mean (SD)): 13.59 (6.81) (range: 4 ‐ 27 years)

Gender (male/female): 62/76

Setting: Bandar Abbas Pediatric Hematology Clinic

Country: Iran

Inclusion criteria

  • major/intermedia thalassaemia

  • > 2 years

  • Serum ferritin > 1000 ng/mL

  • Normal creatinine

  • Acceptable CBC

  • Negative PCR in terms of HCV, negative HBV and HIV

  • Absence of heart disease and cardiac drugs

  • EF> 55%

  • Absence of proteinuria

Exclusion criteria

  • Serum creatinine increased by more than 33% compared to baseline

  • Vision and hearing problems

  • Hyperemesis

  • Lack of response to anti‐nausea medication and fluids therapy

  • Severe and rapidly progressive skin rash

  • Increase in liver enzymes more than 5 times normal

  • Platelets < 150 000

  • Neutrophils < 1500

Follow‐up: 8 months

Interventions

2 groups:

  • DFX (n = 69): 20 mg/kg/day oral, once a day on an empty stomach at least half an hour before a meal

  • DFO (n = 69): 40 ‐ 50 mg/kg subcutaneous for 6 nights a week

Outcomes

  • CBC

  • ALT, AST

  • Ferritin

  • Creatinine

  • Urinalysis

Furthermore, patients were visited weekly on the base of drug tolerance and side effects.

Notes

No funding or conflict of interest mentioned.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Patients were assigned randomly in two groups"

No details given with regard to sequence generation.

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No placebo treatment mentioned. Due to different administration routes, blinding is not likely.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Although number of patients who reached end of study was not clearly stated, we concluded from given means, SDs and P values, that 69 participants were included in the results.

Selective reporting (reporting bias)

High risk

ALT, AST, creatinine evaluated at EOS, but only baseline values given

Only ferritin level, haemoglobin level and drug side effects predefined as outcomes on clinicaltrials.gov

Exclusion criteria were stated as, among others, “vision and hearing problems”, “severe skin rash”: implies that AEs were known for DFX/DFO and these data were collected; however they were not reported;

Only AEs reported: leukopenia, thrombocytopenia, although patients were visited weekly for drug tolerance and side effects.

Other bias

Low risk

No other bias detected.

Molavi 2014

Methods

Randomised clinical study.

Participants

100 children with thalassaemia major were selected, 94 participants entered study

Age (mean (SD)):12.23 (4.09) years (range: 2 ‐ 15 years)

Gender (male/female): 44/48

Setting: Thalassemia medical centre of Bandar Abbas

Country: Iran

Inclusion criteria:

  • Age: 2 ‐ 15 years

  • Serum ferritin > 2000 ng/mL despite the treatment with DFO (50 mg/kg 3 times a week)

Exclusion criteria: not mentioned

Follow‐up: 12 months

Interventions

Two groups:

  • DFO (n = 48): 50 mg/kg transfused 3 times a week

  • DFO and DFX (n = 46): DFX 20‐40 mg/kg (in case there was no desirable response, the dosage was altered to 20 mg/kg, and when no consequences were observed, it was increased to 40 mg/kg)

Outcomes

  • Serum ferritin

  • Neutrophil, ALT, AST, ALP

  • Creatinine

  • Hemoglobin

Notes

Conflict of interest not mentioned.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomly and in the order of visiting the centre, they were divided in two 50‐member groups"

No details given on how random sequence was generated.

Allocation concealment (selection bias)

Unclear risk

No details mentioned with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No placebo treatment in the Desferal monotherapy group is mentioned, so blinding is not likely in particular of performance bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

It remains unclear wether six patients were excluded due to exclusion criteria before or after randomisation; small number doesn't seem to affect results.

Selective reporting (reporting bias)

High risk

Level of creatinine was measured, but results were not reported.

Other bias

Low risk

No other bias detected.

Nisbet‐Brown 2001

Methods

Randomised, double‐blind, placebo‐controlled dose‐escalation study.

Participants

24 participants with transfusion‐dependent β‐thalassaemia (23 analysed, 3 replacements for participants who were withdrawn for serious AEs during the study)

Age (median (range): placebo: 32 (22 ‐ 38) years; 10 mg/kg DFX: 28 (20 ‐ 39) years; 20 mg/kg DFX: 24 (18 ‐ 38); 40 mg DFX: 27 (19 ‐ 34)

Gender (male/female): 11/12

Setting: Children's Hospital, Boston; Weill Medical College, New York; Toronto General Hospital, Toronto

Country: USA (2 centres) and Canda (1 centre)

Participant characteristics:

  • Transfusion‐dependent β‐thalassaemia

  • ≥ 16 years

  • Serum ferritin values:1000 ‐ 8000 ng/mL

  • Liver biopsies done in the previous 3 months with ≥ 3.5 mg Fe/g dry weight

  • All participants required treatment with DFO at 20 mg/kg/day (mean daily dose) for at least 4 weeks before screening and a post transfusion haemoglobin concentration of at least 130 g/L

Follow‐up: 12 days

Interventions

Four groups:

  • DFX (n = 5): 10 mg/kg

  • DFX (n = 6): 20 mg/kg

  • DFX (n = 7): 40 mg/kg

  • Placebo (n = 5)

Outcomes

  • Dietary, urine and faecal iron measured by atomic absorption spectrometry

  • Net faecal iron excretion calculated by individual iron content in faeces minus individual iron content in the diet (the net iron excretion for each participant in mg Fe kg–1 day–1 was derived from the sum of the daily measurements of net faecal iron excretion and urinary iron excretion)

  • UIBC calculated from serum iron concentration and total iron binding capacity

Notes

Conflict of interest and funding:

Novartis was involved in design and monitoring of the study.Study was financial supported by Novartis Pharmaceuticals Corporation.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Details of sequence generation process not stated.

"The randomisation sequence was generated by Novartis Pharmaceuticals and delivered to the research pharmacy in duplicate sealed envelopes."

Allocation concealment (selection bias)

Unclear risk

Sealed envelopes were used. However, unclear whether opaque and numbered.

"The randomisation sequence was generated by Novartis Pharmaceuticals and delivered to the research pharmacy in duplicate sealed envelopes."

Blinding (performance bias and detection bias)
All outcomes

Low risk

This was a placebo‐controlled study, in which investigators and those responsible for administering study drug were blinded with regard to treatment allocation. However, it remains unclear whether outcome assessors and data analysts were blinded as well.

"The investigators and those responsible for administering study drug were unaware of treatment allocation."

"Placebo and ICL670 were prepared as dispersible tablets with standard excipients. Tablets were suspended in water, and patients ingested the drug or placebo on an empty stomach."

Incomplete outcome data (attrition bias)
All outcomes

Low risk

"Therefore, all patients who began either placebo or drug were included in the data analysis, whether they completed the 12‐day course or withdrew prematurely."

Selective reporting (reporting bias)

Unclear risk

"We did clinical, laboratory, and other safety assessments regularly throughout the study."

However, only a limited amount of data are presented in the publication.

Other bias

Low risk

No other bias detected.

Peng 2013

Methods

Randomised controlled study.

Participants

26 individuals were recruited, who were diagnosed as β‐severe thalassaemia by gene screening, 2 met exclusion criteria

24 participants were randomised

Gender: 13 male, 11 female

Age (mean (SD) (range)): (14 (3) (11 ‐ 26)) years

Setting: First Affiliated Hospital, Guangxi Medical University, Nanning

Country: China

Inclusion criteria:

  • Age ≥ 10 years

  • History of 50 unit red blood cell transfusion (1 unit = 200 mL red blood cell) at least

  • Receiving red blood cell transfusion (≥ 10 unit per year)

Exclusion criteria:

  • T2* ≥ 6.3 ms at first MRI screening or LIC ≤ 2 mg/g (1 mg/g = 17.9 mmoL/kg)

  • Dysfunction of liver and kidney

  • Contraindication of MR screening or disagreement doing the screening

Follow up: 12 months

Interventions

Two groups:

  • DFX (n = 12): 40 mg/kg daily before breakfast

  • DFO (n = 12): 50 mg/kg, diluted by 10% solution, subcutaneous injection 8 ‐ 12 hours continuously per day, at least 5 days every week

‐ Parameters of the body of patient, LIC, serum ferritin, serum creatinine, liver function and toxicity of the drugs are regarded as standards to adjust the dose or discontinue the therapy

‐ Meanwhile, the patients still receive the former transfusion program (red blood cell transfusion ≥ 10 units per year) to maintain the haemoglobin > 90 g/L

‐ 5‐day washout period without chelation therapz before treatment

Outcomes

  • Serum ferritin

  • Liver R2*

  • Severity of iron deposition in liver

Notes

Study funding sources: National Natural Science Foundation of China, the Natural Science Foundation of Guangxi, China

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"[...] 24 iron‐overloaded patients were randomly divided into 2 groups [...]"

No details given with regard to sequence generation.

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No details given with regard to blinding.

Due to different administration routes, blinding is not likely.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

There were no missing outcome data.

Selective reporting (reporting bias)

High risk

Side effects were not reported.

Other bias

Low risk

No other bias detected.

Pennell 2014

Methods

Prospective, multinational, randomised, open‐label, parallel‐group, phase 2 study; Non‐inferiority study.

Participants

197 participants were randomised

160 participants completed one year of treatment

Age (mean (SD)): 19.8 (6.4) years (range: 10 ‐ 40 years)

Gender (male/female): 115/82

Setting and countries: 22 centres across 11 countries

Inclusion criteria:

  • People with β‐thalassaemia major, Diamond‐Blackfan anaemia, low/intermediate 1 myelodysplastic syndromes, or sideroblastic anaemia

  • Aged ≥ 10 years

  • Myocardial T2* 6 to 20 ms without clinical symptoms of cardiac dysfunction (shortness of breath at rest or exertion, orthopnoea, exercise intolerance, lower‐extremity edema, arrhythmias)

  • LVEF ≥ 56%,

  • R2 magnetic resonance imaging LIC ≥ 3 mg Fe/g dry weight

  • Lifetime history of ≥ 50 U red blood cell transfusions

  • Receiving ≥ 10 U/y of red blood cell transfusion

  • Only people with β‐thalassaemia major fulfilled the inclusion criteria and were enrolled in the study

Exclusion criteria:

  • Serum creatinine above the ULN

  • Significant proteinuria (urinary protein/creatinine ratio ≥ 1.0 mg/mg in a non‐first‐void urine sample at baseline)

  • ALT > 5 times the ULN only if their LIC was < 10 mg Fe/g dry weight

  • Considerable impaired GI function or GI disease

  • History of clinically relevant ocular and/or auditory toxicity related to iron chelation therapy

  • History of HIV seropositivity or malignancy within the past 5 years

Follow‐up: 12 months

Interventions

Two groups:

  • DFX (n = 96): Once‐daily starting dose was 20 mg/kg per day for 2 weeks, followed by 30 mg/kg per day for 1 week, and then continued with 40 mg/kg per day

  • DFO (n = 91): 50 to 60 mg/kg per day via subcutaneous infusion over 8 to 12 hours, 5 to 7 days a week; dose adjustment recommendations were provided based on continuous assessment of efficacy and safety markers

Outcomes

  • Ratio of geometric mean myocardial T2* with DFX divided by the ratio of geometric mean for DFO

  • LVEF

  • LIC

  • Serum ferritin

Furthermore, safety, compliance, dose interruptions/reductions and laboratory parameters (serum creatinine, blood creatinine, ALT) were measured.

Notes

Study was sponsored by Novartis Pharma AG. Novartis Pharma AG was involved in design of the study, conducted the statistical analysis and paid a medical writer who assisted with writing the article. Some of the authors received research grant funding, honoraria or lecture fees from Novartis Pharmaceuticals and/or other pharmaceutical companies.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"[...]patients were randomised in a 1:1 ratio [...]"

"Randomization was based on permuted blocks; stratification by centre was not conducted."

Allocation concealment (selection bias)

Unclear risk

No details given with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

"Core laboratories were blinded to treatment allocation."

"In order to eliminate potential unrecognised biases, the core clinical trial team was blinded to the treatment assignment prior to the database lock for the primary analysis."

"Open‐label trial"

No placebo treatment mentioned. Due to different administration routes, blinding is not likely in particular of performance bias.

Incomplete outcome data (attrition bias)
All outcomes

High risk

ITT was done regarding myocardial T2*, but the number of included patients (n=180) was lower than the number of randomised patients (n=197)

Apart from that, per‐protocol analysis was done for the other outcomes.

Selective reporting (reporting bias)

High risk

Only most common AE (≥ 5%) and drug related AE ≥ 2 participants were reported

Other bias

Low risk

No other bias detected.

Piga 2002

Methods

Open label, randomised, multicenter, phase II study.

Participants

71 people with thalassaemia and transfusional iron overload: 69 people with β‐thalassemia major, 2 people with β‐thalassemia intermedia

Age mean (range): DFX 10 mg/kg/day: 23.7 years (17 ‐ 33 years); DFX 20 mg/kg/day: 25.6 years (19 ‐ 50 years); DFO: 22.7 (18 ‐ 29 years)

Gender (male/female): 26/45

Setting and country: 4 centres in Italy

Inclusion criteria:

  • Should have been regularly transfused

  • Should have received a mean daily dose of DFO ≥ 30 mg/kg for 5 days/week for at least 4 weeks prior to entering the screening period

  • Serum ferritin between 2000 ‐ 8000 ng/mL on at least two evaluations in the last 12 months or

  • LIC of 5 ‐ 15 mg Fe/g dry weight measured in the last 12 months by SQUID

  • Average post‐transfusion haemoglobin level between 10.5 ‐ 13.5 g/dL during last 12 months

Exclusion criteria:

  • AST or ALT > 250 U/L

  • Creatinine clearance < 80 mL/minute

  • People with hypertension

  • People with any degree of A‐V block, clinically relevant QT prolongation

  • Treatment with digoxin or any other drug that could induce prolongation of A‐V conduction

  • People with diagnosis of cataract or a previous history of clinically relevant ocular toxicity related to iron chelation

Follow‐up: 48 weeks

Interventions

3 groups:

  • DFX (n = 24): 10 mg/kg once‐daily using 250 mg tables which were divisible into four parts. The correct number of tablets was dispersed in a glass of non‐carbonated mineral water, stirred and ingested 30 minutes before breakfast

  • DFX (n = 24): 20 mg/kg once daily using 250 mg tables which were divisible into four parts. The correct number of tablets was dispersed in a glass of non‐carbonated mineral water, stirred and ingested 30 minutes before breakfast

  • DFO (n = 23): 40 mg/kg on 5 consecutive days per week. Doses were prepared as a 10% solution using commercially available vials of 500 or 2000 mg dry powder. Subcutaneous infusion was performed using a pump over 8 ‐ 12 hours.

‐ During the 14‐day run‐in period, eligible patients had their usual DFO therapy adjusted to 40 mg/kg given on 5 consecutive days each week

‐ The study protocol allowed for dose adjustment within the range of 5 ‐ 40 mg/kg/day in the DFX groups and 20 ‐ 50 mg/kg in the DFO group

‐ Depending on response, assessed primarily using the change in LIC at 3 consecutive determinations, dose increases or decreases were made by ±5 or ±10 mg/kg in the DFX groups and by ± 10 mg/kg in the DFO group

‐ Dose reductions were performed if the decrease in LIC was extrapolated to fall below 2 mg Fe/g dry weight within the next 12 weeks and dose increases were prescribed if an increasing trend in LIC was noted

‐ Dose adjustments were decided on a case‐by‐case basis in joint consultation between the Study Monitoring Committee and the sponsor

‐ On day ‐5, participants were admitted to the study site to receive a blood transfusion to achieve a target haemoglobin level of ≥ 13g/dL prior to commencing study treatment followed by a DFO washout period of 5 days

Outcomes

  • Mortality

  • LIC (SQUID)

  • Serum ferritin

  • Serum iron

  • Serum transferrin

  • Transferrin saturation

  • Blood indices

  • Liver and renal function

  • Serum electrolytes

  • Copper and zinc

  • Second void urine samples with measurement of N‐acetyl‐beta‐glucosaminidase and beta2‐miroglobulin

  • Ophthalmological examination including slit lamp examination of the lens and retinal fundoscopy

  • Audiometry, ECG and liver ultrasonography

Notes

Study was supported by Novartis Pharma AG. Some of the authors are employed by Novartis Pharma or received lecture fees from the manufacturer.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization was performed using a validated system that generates an automated random assignment of numbers to treatment groups."

We expect that using this system resulted in an adequate sequence generation.

Allocation concealment (selection bias)

Unclear risk

"Randomization was performed using a validated system that generates an automated random assignment of numbers to treatment groups."

No information is given with regard to allocation concealment.

Blinding (performance bias and detection bias)
All outcomes

High risk

It is classified as an open‐label study. There is no mentioning of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants are included in safety analysis (primary objective). Few patients only are not included in efficacy analysis (secondary objective).

Selective reporting (reporting bias)

Unclear risk

"Laboratory tests, including evaluation of blood indices, liver and renal function, serum electrolytes, copper and zinc, were performed at baseline and at 2‐weekly intervals throughout the study. All laboratory parameters were measured at a central laboratory (EXACTA Clinical Trials Services, Verona, Italy). Second void urine samples were collected for measurement of N‐acetyl‐b‐glucosaminidase and an aliquot of urine was alkalinized for measurement of b‐2 microglobulin. An ophthalmology examination, including a slit lamp examination of the lens and retinal fundoscopy, was performed every 2 weeks. Audiometry, ECG and liver ultrasonography were carried out every 3 months. Adverse events were recorded at each study visit and the severity of each adverse event was graded as mild, moderate or severe. A serious adverse event was defined as a medically significant event that was either fatal or life threatening, required surgical intervention, prolonged hospitalization or resulted in persistent disability. All adverse events and serious adverse events were assessed by the investigator for a possible relationship to the study drug. Adverse events were ranked according to incidence in the deferasirox 20 mg/kg/day treatment group."

"All biomagnetic liver susceptometry evaluations were performed at the Ospedale Regina Margherita, University of Turin, Italy. LIC was determined at screening and then every 12 weeks during treatment and at the end of the study...... During the study, markers of iron metabolism (serum ferritin, serum iron, serum transferrin and transferrin saturation) were analyzed by a central laboratory (EXACTA Clinical Trials Services, Verona, Italy). The transferrin saturation was calculated from the serum iron and the transferrin concentrations. Urinary iron excretion was determined in 24‐hour urine collections in ten patients taking deferasirox (five in each dose group) who also underwent blood sampling for pharmacokinetic analyses. Urinary iron excretion was measured using atomic absorption spectrometry."

Only selected parameters at selected time points are reported.

Other bias

Low risk

No other bias detected.

Sanjeeva 2015

Methods

Prospective randomised comparative study.

Participants

41 participants were randomised

38 participants reached end of study

Age (mean (SD)): DFX (n = 19): 5.23 (2.76) years; DFP (n = 19): 7.26 (2.42) years

Gender (male/female): 22/16

Setting: Thalassemia day care centre of Indira Gandhi Institute of child health, Bengaluru

Country: India

Inclusion criteria

  • Thalassemia diagnosis and regular blood transfusion

  • Serum ferritin > 1000 ng/mL

  • No chelation therapy

Exclusion criteria

  • Already on chelation therapy

  • Chronic liver or renal disease

Follow‐up: 12 months

Interventions

2 groups:

  • DFX (n = 19): dose of 20 mg/kg/day, once a day

  • DFP (n = 22): 75 mg/kg/day in 3 divided doses

Outcomes

  • Serum ferritin

  • WBC

  • Platelet count

  • Blood urea

  • Serum creatinine

  • Serum enzymes (AST, ALT)

  • Side effects

Notes

No funding or conflict of interest mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"These children were randomly divided into two groups as group 1 and group 2 by computer generated randomisation."

Allocation concealment (selection bias)

Unclear risk

No details mentioned with regard to concealment of allocation.

Blinding (performance bias and detection bias)
All outcomes

High risk

No details given with regard to blinding.

Due to different application frequencies, blinding is not likely, in particular regarding performance bias.

Incomplete outcome data (attrition bias)
All outcomes

High risk

3 dropouts due to AEs in deferiprone group, per protocol analysis.

Selective reporting (reporting bias)

High risk

Measurements other than serum ferritin and AEs only given for 9 months (and only in the thesis document), but not for end of study (unsure whether not measured or not reported), although the author report measurements every 3 months.

In the thesis document, fundoscopy, growth harm and hearing were part of the evaluation sheet at 12‐month follow‐up, but no results were reported.

Other bias

Low risk

No other bias detected.

Taher 2012

Methods

Multinational, prospective, randomised, double‐blind, placebo‐controlled phase 2 study.

Participants

166 participants were randomised.

95 non‐transfusion‐dependent β‐thalassaemia, 22 α‐thalassaemia, 49 HbE/β‐thalassaemia

148 participants completed 1 year of the study.

Inclusion criteria

  • ≥ 10 years of age with

  • Non‐transfusion‐dependent β‐thalassaemia

  • Iron overload (R2‐MRI‐measured LIC ≥ 5 mg Fe/g dry weight)

  • Serum ferritin > 300 ng/mL at screening based on 2 consecutive values ≥ 14 days apart were eligible

  • Participants were required to have not received transfusions within 6 months or chelation therapy within 1 month before study entry

Exclusion criteria

  • People with previous exposure to DFX

  • Anticipated regular transfusions; unplanned transfusions during the study were allowed

  • HbS variants of thalassaemia syndromes

  • Active hepatitis B (positive hepatitis B surface antigen with negative hepatitis B surface Ab) or hepatitis C (positive hepatitis C virus Ab and detectable hepatitis C virus RNA with alanine aminotransferase [ALT] above the normal range)

  • Cirrhosis

  • Levels of ALT > 5 x ULN

  • Serum creatinine > ULN or creatinine clearance ≤ 60 mL/min on 2 measurements

  • Significant proteinuria (urine protein/urine creatinine ratio > 1.0 mg/mg) on 2 measurements

Follow‐up: 1 year

Interventions

4 groups:

  • DFX (n = 55): 5 mg/kg/day

  • DFX (n = 55): 10 mg/kg/day

  • Placebo (n = 28): 5 mg/kg/day

  • Placebo (n = 28): 10 mg/kg/day

‐ Doses were doubled at 24 weeks for patients with LIC > 7 mg Fe/g dry weight and LIC reduction < 15% from baseline

‐ Dose adjustment recommendations were also provided based on continuous safety assessments

‐ If serum ferritin was <100 ng/mL or LIC was <3 mg Fe/g dry weight at any visit, treatment was to be suspended until LIC increased to ≥ 5 mg Fe/g dry weight and serum ferritin to > 300 ng/m

Outcomes

  • LIC

  • Number and proportion of patients with a LIC decrease of ≥ 3 mg Fe/g dry weight, those with ≥ 30% reduction in LIC, and those with LIC ≤ 7, ≤ 5, and ≤ 3 mg Fe/g dry weight

  • Serum ferritin

  • Correlation of serum ferritin and LIC

  • AEs

  • Serious AEs

  • Adherence

Notes

Study was sponsored by Novartis Pharma AG. Novartis was involved in design and statistical analysis

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"[...] patients were block randomised [...]".

No details given on how random sequence was generated.

Allocation concealment (selection bias)

Low risk

"[...] patients were block randomised using an interactive voice response system. After confirming that the patient fulfilled the inclusion/exclusion criteria, the investigator contacted the interactive voice response system to obtain a randomisation number linking the patient to a treatment arm."

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

"Because blinding of dose was not possible, blinding was only applied to the treatment received. All persons were blinded to the treatment from the time of randomisation until database lock."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Attrition was 12.7%, 10.9% and 8.9% for the DFX 5 mg, DFX 10 mg and the placebo group, respectively

In the journal publication, the authors state that "efficacy was assessed for the full analysis set (all randomised patients).[...] If there was no LIC measurement available at week 52, the last available post‐baseline LIC measurement was carried forward."

Number of participants analysed on clinicaltrials.gov doesn't include all randomised patients for continuous outcomes.

Selective reporting (reporting bias)

High risk

Extensive data set available on ClinicalTrials.gov (along with prespecified protocol), but AEs and SAEs were not reported separately for the core phase

In the journal publication AEs and drug‐related AEs were not reported completely

Other bias

Low risk

No other bias detected.

AE: adverse event
ALP: alkaline phosphatase
ALT: alanine aminotransferase
AST: aspartate aminotransferase
A‐V: atrio‐ventricular
CBC: complete blood count
CONSORT: consolidated standards of reporting trials
DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
ECG: electrocardiogram
EF: ejection fraction
EOS: eosinophil count
Fe: iron
FT₃: serum‐free triiodothyronine
FT₄: serum‐free thyroxine
GI: gastrointestinal
HBV: hepatitis B virus
ITT: intention‐to‐treat
LVEF: left ventricular ejection fraction
LIC: liver iron concentration
MRI: magnetic resonance imaging
QoL: quality of life
PCR: polymerase chain reaction
RBCs: red blood cells
RNA: ribonucleic acid
SD: standard deviation
SQUID: superconducting quantum interference device
TGA: antithyroglobulin
TSH: thyroid‐stimulating hormone
TPO: antithyroid peroxidase
UIBC: unsaturated iron binding capacity
ULN: upper limit of normal
WBC: white blood count

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Economou 2010

Not randomised. Two‐arm observational study.

EPIC 2008

Not randomised, no comparison group. Single‐arm observational study.

Erdogan 2013

Not randomised. Two‐arm observational study.

ESCALATOR 2005

Not randomised, no comparison group. Single‐arm observational study.

Fernandes 2013

Randomised, evaluating amlodipine added to standard iron chelation therapy.

Gao 2011

Not randomised. Study comparing different doses of DFX and DFX to deferiprone and DFO.

Garadah 2011

Not randomised. Single‐arm interventional study.

Genc 2015

Not randomised. Three‐arm observational study.

Gomber 2016

Not randomised. No adequate random sequence generation.

Grady 2012

Non‐randomised. Single‐arm interventional study.

Hagag 2013

Randomised. Evaluating silymarin versus placebo added to DFX.

Hesham 2014

Not randomised. Case‐control study.

Inati 2011

Randomised controlled study on people with thalassaemia having undergone curative hematopoietic stem cell transplantation.

Kallistheni 2012

Not randomised. Two‐arm interventional study.

Karakukcu 2012

Not randomised. Three‐arm observational study.

Keikhaei 2011

Not randomised. Two single‐arm interventional studies.

Lu 2015

Randomised. Pharmacokinetic study with a very small population (n = 8).

Medrano Engay 2013

Randomised. No individuals with thalassaemia included.

Ozturk 2015

Not randomised. Three‐arm observational study.

Pakakasama 2011

Randomised controlled study on people with thalassaemia having undergone curative hematopoietic stem cell transplantation.

Pepe 2010

Study not randomised. Three‐arm observational study.

Pepe 2011

Study not randomised. Two‐arm observational study.

Pileri 2014

Randomised. No individuals with thalassaemia included.

Song 2014

Randomised. Pharmacokinetic study with a very small population (n = 8).

Torcharus 2011

Not randomised. Three‐arm observational study.

Walter 2012

Not randomised. Single‐arm interventional study.

DFO: deferoxamine
DFX: deferasirox

Characteristics of studies awaiting assessment [ordered by study ID]

Ansari 2015

Methods

Unclear, if study was randomised.

Participants

108 people with thalassemia major

Age: > 10 years

Inclusion criteria

  • Iron overload in cardiac T2* MRI assay

Interventions

3 groups:

  • DFX

  • DFO

  • DFO + DFP

Outcomes

  • Myocardial iron (T2* MRI)

  • Liver T2*

  • AEs

Notes

Author was contacted.

EUCTR2010‐023217‐61‐GB

Methods

A phase IV, open‐label, partial cross‐over partial parallel, randomised, multi‐centre study.

Participants

Target sample size: 64

Inclusion criteria

  • ≥ 18 years

  • Ability to provide informed consent

  • Ability to meet all study requirements

  • Adequate renal function (e.g. creatinine clearance = 60 mL/min)

  • Willingness to stop other iron chelation therapy

  • No known allergies to the drug

  • Not pregnant or breastfeeding and willing to use effective contraception

For non‐naive cohort

  • Transfusional iron overload as defined by a minimum of 20 lifetime transfusion episodes

  • Transfusional iron loading with ferritin levels > 500 mcg/L and/or LIC > 3 mg of iron/g dry weight (as previously demonstrated by liver biopsy or MRI prior to the study)

  • Established on deferasirox therapy for at least 1 year

  • GI side effects believed to be related to their therapy as suggested by at least one of the following: ‐ The temporal relationship of GI side effects occurrence with the administration of deferasirox.

Exclusion criteria

  • People with GI symptoms assumed or known not to be caused by DFX

  • People who are treated for any condition with aspirin daily, those under chronic steroid therapy and those on anticoagulant therapy that could all lead to potential GI symptoms/bleeding

  • Presence of GI disease that may significantly alter the absorption of deferasirox (e.g. ulcerative diseases, uncontrolled nausea, vomiting, diarrhoea, malabsorption syndrome, GI or rectal bleeding)

  • History of GI surgery (except appendicitis and cholecystectomy) e.g. stomach/bowel surgery or awaiting elective surgery in the next 2 months

  • Undergoing acute medical intervention or hospitalisation

  • Psychiatric illness (schizophrenia, major depression) which may interfere with study requirements

  • Any other medical condition that, in the opinion of the site investigator would interfere with completing the study (visual problems or cognitive impairment)

  • Platelet count less than 100,000

  • Currently on treatment for hepatitis C

  • Patients with severe iron loading in the heart (T2* less than 10 ms)

  • Patients with severe total body iron load (LIC over 30 mg/g dry weight)

  • Patients who have historical evidence of severe iron loading in the heart

  • Women of child‐bearing potential who are planning a pregnancy, pregnant, lactating and unwilling to use effective means of contraception

  • Inadequate renal function (e.g. creatinine clearance < 60 mL/min)

  • Patients unwilling to stop using other iron chelation therapy for the study duration

  • Known allergy to the drug

  • Patients on aluminium containing antacid preparations

  • Patients who are on vitamin C at doses higher than 200 mg/day

  • Patients receiving or having received any investigational drug within 30 days prior to study enrolment

  • Patients unable to understand oral or written English

Interventions

Once daily oral DFX (dispersible tablet), when administered before or after food in people with transfusional haemosiderosis

Outcomes

Primary outcome

  • Between‐treatment arm‐difference of the mean individual change of GIQLI (follow‐up minus baseline) in the non‐naive cohort (patients on established deferasirox therapy)

Secondary outcomes

  • Difference between mean follow‐up (1 month and 2 months) vs baseline changes of GIQLI per arm in naive cohort

  • Difference of the mean GSRS score changes in both arms of the established cohort and the naive cohort

  • Difference of the mean SF‐36 score changes in both arms of the established cohort and the naive cohort

  • Arm difference of geometric mean steady state Cmin at visit 3 and visit 4 in the cross‐over group

  • Arm difference of geometric mean AUC and Cmax at visit 3 and visit 4 in cross‐over group

  • Arm difference of geometric mean steady‐state Cmin at Visit 3 for all patients (after 1 month of treatment)

  • Effect of food on systemic exposure of deferasirox as assessed by pharmacokinetics approach. Pooled pharmacokinetic data from all arms will be analysed with a population pharmacokinetic model and appropriate covariates will be examined for clinical and statistical relevance

Notes

Date of first enrolment: 27/01/2012

Date of the global end of the study: 16/07/2012

Sponsor: University College London

Recruitment status: Not Recruiting

In the EU Clinical Trials Register, a premature end of the study is reported. As of now, no data have been published.

Author was contacted

Hagag 2015

Methods

Unclear, if study was randomised.

Participants

120 people with β‐thalassemia major were included.

Age (mean (SD)): 5.43 (1.37) (range 4 ‐ 7) years

Gender: 68 males, 52 females

Setting: Hematology Unit, Pediatric Department, Tanta University Hospital

Country: Egypt

Inclusion criteria

"Children with beta thalassaemia major with serum ferritin levels of more than 1000 ng/mL who had not received iron chelation before this study and maintained on regular use of chelation during this study."

Exclusion criteria

"Children with thalassaemia with serum ferritin level less than 1000 ng/mL. Children with thalassaemia with hepatitis A, B or C."

Follow‐up: 6 months

Interventions

Group A: "30 patients were treated with 8 hours intravenous infusion of Desferrioxamine, 40 mg/kg/day, 6 days per week for 6 months."

Group B: " 30 patients were treated with subcutaneous infusion of Desferrioxamine, 40 mg/kg/day, 6 days per week 8 hours per day at night using Desferal pump for 6 months."

Group C: " 30 patients were treated with oral Deferiprone 75 mg/kg/day in three divided doses daily for 6 months."

Group D: " 30 patients were treated with oral Deferasirox 30 mg/kg/day in single daily dose on empty stomach for 6 months."

Outcomes

  • Serum ferritin

  • Serum TIBC

  • White blood cells

  • Neutrophils

  • Platelets count

  • Liver enzymes

  • Creatinine

  • Blood urea

  • Kidney function

Notes

Author was contacted.

IRCT201110087677N1

Methods

Comparative study of incidence of lens opacity between Osferal and Deferoxamine in thalassaemia major.

Participants

50 people with thalassaemia major

Inclusion criteria:

‐ children with thalassaemia major

‐ being candidate for chelator therapy because of iron overload

Exclusion criteria:

‐ diabetes mellitus and rheumatologic diseases

‐ any lens disease or chelator therapy before the study

Follow‐up: 12 months

Interventions

"Then the patients will be divided into two 25 membered groups, and each group will receive one of the chelators randomly."

"Intervention:In this group, 25 patients are put on a new Iranian drug Osferal, and then it's side effect that is "lens opacity", will be compared with that of the control group."

"Control:Based on the present policy, 25 patients who receive Deferoxamine and have a known percent of "lens opacity", are considered as the control group."

Outcomes

Lens opacity

Notes

Expected recruitment start date: 2010‐12‐22

Expected recruitment end date: 2011‐12‐22

Author was contacted

Kakkar 2015

Methods

Prospective randomised study.

Participants

Size of study population: not mentioned

Setting: Thalassemia ward of Department of Pediatrics, Dayanand Medical College and Hospital, Ludhiana

Country: India

Inclusion and exclusion criteria: not mentioned

Follow‐up: not mentioned

Interventions

2 groups:

  • DFX: 40 ‐ 50 mg/kg/day in once daily doses

  • DFX: 40 ‐ 50 mg/kg/day in 2 divided doses

Outcomes

  • Serum ferritin

  • Cardiac MRI T2*

  • Liver MRI T2*

  • LVEF

  • Safety profile in terms of gastrointestinal side effects, rash and change in serum creatinine values, SGPT and GFR

Notes

The study was only reported in a conference abstract.

Author was contacted.

NCT02198508

Methods

Randomised, open‐label, single‐centre, cross‐over study.

Participants

Target sample size: 13

Inclusion criteria

  • 18 years of age or older

  • Serum ferritin greater than 2000 ng/mL

  • Serum creatinine within normal range for a measuring laboratory

  • Platelet count exceeding 140000/mm³

  • Body weight at least 40 kg

  • None had a history of clinical significant of gastrointestinal, hepatic, renal, endocrine, oncologic, infectious, pulmonary or cardiovascular disease

Exclusion criteria

  • HIV positive, history of immunologic hypersensitivity to any medication

  • Women pregnant or breastfeeding

  • Drug or alcohol abuse

  • Patients showed abnormal or irregular bowel function (defined as more than three bowel movements a day or less than one bowel movement every other day)

  • Receiving warfarin, digoxin, or anti‐arrhythmic or anti‐seizure medication

Interventions

Experimental: combination treatment: DFX and DFP

Active comparator: DFX

Active comparator: DFP

Outcomes

Primary outcome:

  • Iron excretion from urine and faeces by flame atomic absorption spectroscopy [time frame: 25 days]

Secondary outcome:

  • Drug concentration in plasma by pharmacokinetics analysis [time frame: 25 days]

Notes

Primary completion date: July 2008

Recruitment status: completed

Author was contacted

AE: adverse event
AUC: area under the curve
Cmax: maximum or peak concentration (of a drug observed after its administration)
Cmin: minimum concentration (of a drug observed after its administration)
DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
EU: European Union
GFR: glomerular filtration rate
GIQLI: Gastrointestinal Quality of Life Index
GI: gastrointestinal
GSRS: Gastrointestinal Symptom Rating Scale
LIC: liver iron concentration
LVEF: left ventricular ejection fraction
MRI: magnetic resonance imaging
SD: standard deviation
SGPT: serum glutamic‐pyruvic transaminase
TIBC: total iron building capacity

Characteristics of ongoing studies [ordered by study ID]

Cutino 2009

Trial name or title

Sequential DFX‐DFP versus DFX or DFP multicentre randomised study

Methods

Randomised, parallel‐group, open study.

Participants

Planned number of participants to be included in the member state: 363

Inclusion criteria:

  • Male and female

  • Age > 12 and < 50 years old

  • Diagnosis of β‐thalassemia major

  • Seric ferritin concentration > 1000 µg/L

Exclusion criteria:

  • Diagnosis other than β‐thalassemia major

  • Participants with renal failure (creatinine clearance < 60 mL/min)

  • ALT/AST > 300 U/L

  • Severe cardiomyopathy

  • Individuals with previous significant ocular toxicity related to iron chelating drugs

  • Individuals with previous significant idiosyncratic reaction or severe toxicity to previous therapy with DFP or DFX

  • Platelets < 100.000/mmc

  • Leukocytes < 300/mm

  • Severe liver insufficiency (Child‐Pugh Score C)

Interventions

Experimental: sequential DFX‐DFP

Comparator 1: DFX

Comparator 2: DFP

Outcomes

Primary outcome measure:

  • Chelating efficacy assessment of sequential therapy DFX‐DFP versus DFX or DFP alone

Secondary outcome measures:

  • To assess in the sequential deferasirox‐deferiprone treated group a reduction at least in 50% of cases of creatininemia significant increase (> 33%) compared to the deferasirox alone group

  • To assess in the sequential DFX‐DFP group a reduction at least in 70% of cases of neutropenia compared to the DFP alone group

  • To assess, using MRI, in a subgroup, the possible organ‐specific (heart, liver, pancreas) iron overload variation during therapy

Starting date

Date of first enrolment: 27/01/2010

Contact information

N/A

Sponsor: FONDAZIONE FRANCO E PIERA CUTINO

Notes

Initial estimate of the duration of the study: 5 years

DEEP‐2 2012

Trial name or title

Efficacy and safety study to compare deferiprone versus deferasirox in paediatric patients

Methods

Multicentre, randomised, open label, non‐inferiority active‐controlled study

Participants

Estimated enrolment: 344

Inclusion criteria;

  • People of both genders aged from 1 month up to less than 18 years at the time of enrolment

  • People affected by any hereditary haemoglobinopathy requiring chronic transfusion therapy and chelation, including but not limited to thalassaemia syndromes and sickle cell disease

  • People on current treatment with DFO or DFX or DFP in a chronic transfusion program receiving at least 150 mL/kg/year of packed red blood cells (corresponding approximately to 12 transfusions)

  • For participants naive to chelation treatment: participants that have received at least 150 mL/kg of packed red blood cells (corresponding to approximately 12 transfusions) in a chronic transfusion program and with serum ferritin levels ≥ 800 ng/mL

  • For participants aged from 1 month to less than 6 years: known intolerance or contraindication to DFO;

  • Written informed consent and participant's informed assent, relating to his/her comprehension abilities and level of maturity

Exclusion criteria:

  • Participants with known intolerance or contraindication to either DFP or DFX

  • Participants receiving DFX at a dose > 40 mg/kg/day or DFP at a dose > 100 mg/kg/day at screening

  • Platelet count < 100.000/mm³ during the run‐in phase

  • Absolute neutrophils count < 1.500/mm³ during the run‐in phase

  • Hb levels lower than 8 g/dL during the run‐in phase

  • Evidence of abnormal liver function

  • Iron overload from causes other than transfusional haemosiderosis

  • Severe heart dysfunction secondary to iron overload

  • Serum creatinine level > ULN for age during the run‐in phase

  • History of significant medical or psychiatric disorder

  • The patient has received another investigational drug within 30 days prior to this study

  • Fever and other signs/symptoms of infection in the 10 days before baseline assessment

  • Concomitant use of trivalent cation‐dependent medicinal products such as aluminium‐based antacids

  • Positive test for β‐HCG

Interventions

Experimental: DFP oral solution

Comparator: DFX

Outcomes

Primary outcome measure

  • Percentage of successfully chelated participants assessed by serum ferritin levels (all participants) and cardiac MRI T2* (participants above 10 years of age able to have an MRI scan without sedation)

Secondary outcome measures

  • LlC as measured by MRI in participants able to undergo MRI scan without sedation

  • Safety and tolerability assessments

  • QoL

Starting date

Date of first enrolment: 29/11/2012

Contact information

Direzione Scientifica

via Luigi Porta, 14

27100 Pavia

Italy

Arianna Gambino, M.Sc.

phone: +39.0382.25075

email: deep.2@deep‐project.net / [email protected]

Notes

Estimated study completion date: December 2014

Estimated primary completion date: December 2014 (Final data collection date for primary outcome measure)

NCT02125877

Trial name or title

Phase II Study to Investigate the Benefits of an Improved Deferasirox Formulation (Film‐coated Tablet)

Methods

A randomised, open‐label, multicentre, 2‐arm, phase II study.

Participants

Enrollment: 168

Inclusion criteria:

  • Males and females aged ≥ 10 years

  • Individuals with transfusion‐dependent thalassaemia and iron overload, requiring DFX dispersible tablets at doses of ≥ 30 mg/kg/day as per the investigator's decision OR those with very low, low or intermediate risk of myelodysplastic syndrome and iron overload, requiring DFX dispersible tablets at doses of ≥ 20 mg/kg/day as per the investigator's decision

  • History of transfusion of at least 20 PRBC units and anticipated to be transfused with at least 8 units of PRBCs annually during the study

  • Serum ferritin > 1000 ng/mL, measured at screening Visit 1 and screening Visit 2 (the mean value will be used for eligibility criteria)

Exclusion criteria:

  • Creatinine clearance below the contraindication limit in the locally approved prescribing information. Creatinine clearance will be estimated from serum creatinine at screening Visit 1 and screening Visit 2 and the mean value will be used for eligibility criteria

  • Serum creatinine > 1.5 x ULN at screening measured at screening Visit 1 and screening Visit 2 (the mean value will be used for eligibility criteria)

  • ALT (SGPT) > 5xULN, unless LIC confirmed as >10 mg Fe/dry weight within 6 months prior to screening visit 1

  • Significant proteinuria as indicated by a urinary protein/creatinine ratio > 0.5 mg/mg in a non‐first void urine sample at screening Visit 1 or screening Visit 2

  • People with significant impaired GI function or GI disease that may significantly alter the absorption of oral deferasirox (e.g. ulcerative diseases uncontrolled nausea, vomiting, diarrhoea, malabsorption syndrome, or small bowel resection)

  • Liver disease with severity of Child‐Pugh Class B or C

Interventions

Experimental: DFX film‐coated tablet

Active comparator: DFX dispersible tablet

Outcomes

Primary outcome

  • Overall safety [Time Frame: Screening visit thru post‐treatment period ‐ 30 weeks]

Secondary outcome

  • Frequency of selected GI AEs (GI AEs) [time frame: on‐treatment period (Day 1 ‐ safety follow‐up) ‐ 28 weeks]

  • Pharmacokinetic parameters [Time frame: week 1, 3, 13 and 21]

  • Domain scores of treatment satisfaction and palatability over time [time frame: Week 2, 3, 13 and end of treatment (week 24 or within 7 days of last dose)]

  • Weekly average of daily scores of GI diary [Time frame: weekly (screening thru end of treatment visit) ‐ 26 weeks]

  • Relative consumed film‐coated tablet/dispersible tablet counts. Participant‐reported medication consumption [Time frame: daily (day 1/visit 3 ‐ last visit) 24 weeks]

Starting date

July 2014

Contact information

Sponsor: Novartis Pharmaceuticals

Notes

Estimated study completion date: February 2016

Estimated primary completion date: February 2016 (Final data collection date for primary outcome measure)

NCT02435212

Trial name or title

Study to Evaluate Treatment Compliance, Efficacy and Safety of an Improved Deferasirox Formulation (Granules) in Pediatric Patients (2 ‐ < 18 years old) With Iron Overload

Methods

Randomised, open‐label, multicentre, 2‐arm, phase II study. Randomisation will be stratified by age groups (2 to < 10 years, 10 to < 18 years). The study treatment duration will be 48 weeks

Participants

Target sample size:120

Inclusion criteria:

  • Written informed consent/assent before any study‐specific procedures. Consent will be obtained from parent(s) or legal guardians. Investigators will also obtain assent of participants according to local guidelines

  • Iron chelation therapy naive male and female children and adolescents aged ≥ 2 and < 18 years

  • Any transfusion‐dependent anaemia associated with iron overload requiring iron chelation therapy and with a history of transfusion of at least 20 PRBC units and a treatment goal to reduce iron burden

  • Serum ferritin > 1000 ng/mL, measured at screening visit 1 and screening visit 2 (the mean value will be used for eligibility criteria)

Exclusion criteria:

  • Creatinine clearance below the contraindication limit in the locally approved prescribing information. Creatinine clearance will be estimated from serum creatinine (using the Schwartz formula) at screening Visit 1 and screening visit 2 and the mean value will be used for eligibility criteria

  • Serum creatinine > 1.5 x ULN at screening measured at screening visit 1 and screening visit 2 (the mean value will be used for eligibility criteria)

  • ALT > 5 x ULN, unless LIC within 6 months is > 10 mg Fe/dry weight

  • Prior iron chelation therapy

  • Liver disease with severity of Child‐Pugh class B or C

  • Significant proteinuria as indicated by a urinary protein/creatinine ratio > 0.5 mg/mg in a non‐first void urine sample at screening visit 1 or screening visit 2

  • Patients with significant impaired GI function or GI disease that may significantly alter the absorption of oral deferasirox (e.g. ulcerative diseases, uncontrolled nausea, vomiting, diarrhoea, malabsorption syndrome, or small bowel resection)

Other protocol‐defined inclusion/exclusion may apply

Interventions

Experimental: DFX granule formulation

Active comparator: DFX dispersible tablet formulation

Outcomes

Primary outcomes:

  • Change in serum ferritin [Time frame: baseline, 48 weeks]

  • Compliance (using stick/pack tablet count) [Time frame: 48 weeks]

Secondary outcomes:

  • Domain scores of treatment satisfaction and palatability over time [time frame: 48 weeks]

  • Frequency of AEs as a measure of overall safety [Time frame: baseline, 48 weeks]

  • Pharmacokinetic parameters including clearance and volume of distribution in all participants (AUClast, AUCinf, AUCtau, Cmax, Tmax and R) [Time frame: Week 1, Week 5]

  • Pre‐dose DFX concentrations in all participants [time frame: at weeks 1, 3, 5, 9, 13, 17, 21, 25, 29, 33, 37, 41 and 45]

  • Rate of dosing instructions deviations ('Compliance', using a questionnaire) [Time Frame: 48 weeks]

  • Severity of AEs as a measure of overall safety [Time frame: baseline, 48 weeks]

Starting date

September 2015

Contact information

Novartis Pharmaceuticals 1‐888‐669‐6682

Notes

Estimated study completion date: August 2017

Estimated primary completion date: August 2017 (Final data collection for primary outcome measure)

Information given in table according to www.clinicaltrials.gov or http://apps.who.int/trialsearch/ or http://www.irct.ir/. Data were extracted in October 2015.

AEs: adverse events
ALT: alanine aminotransferase
AST: aspartate transaminase
AUCinf: area under the concentration‐time curve extrapolated to time infinity
AUClast: area under the curve up to the last measurable concentration
AUCtau: area under the plasma concentration‐time curve during the dosing interval
Cmax: maximum or peak concentration (of a drug observed after its administration)
CMR: cardiovascular magnetic resonance
DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
GI: gastrointestinal
HCG: human chorionic gonadotropin
LIC: liver iron concentration
LVEF: left ventricular ejection fraction
MRI: magnetic resonance imaging
PRBC: packed red blood cells
QoL: quality of life
SD: standard deviation
SGPT: serum glutamic‐pyruvic transaminase
Tmax: amount of time that a drug is present at the maximum concentration in serum
ULN: upper limit of normal

Data and analyses

Open in table viewer
Comparison 1. Transfusion‐dependent thalassemia: deferasirox vs placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

2

47

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.1

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

2 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 AEs.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 AEs.

2.1 Eye disorders ‐ retinal infarct

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.4 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.5 Investigations ‐ extended QT interval, hypocalcaemia, hypoparathyroidism

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.6 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Discontinuations due to serious AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 Discontinuations due to serious AEs.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 Discontinuations due to serious AEs.

Open in table viewer
Comparison 2. Transfusion‐dependent thalassemia: deferasirox vs deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

8

1170

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.63]

Analysis 2.1

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 1 Mortality at any time point.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 1 Mortality at any time point.

1.1 At 8 months

1

138

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 At 48 weeks (deferasirox 10 mg/kg/day)

1

35

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 At 48 weeks (deferasirox 20 mg/kg/day )

1

36

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 At 1 year

5

942

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.63]

1.5 At 2 years

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 LVEF (%): least squares mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 2 LVEF (%): least squares mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 2 LVEF (%): least squares mean change from baseline.

3 LVEF (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.3

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 3 LVEF (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 3 LVEF (# participants affected).

3.1 Improvement from abnormal LVEF to normal range

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Decrease from normal LVEF to below LLN

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Incidence of thyroid disease at end of study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.4

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 4 Incidence of thyroid disease at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 4 Incidence of thyroid disease at end of study.

5 ALT (# participants affected): improvement from abnormal to normal range Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 5 ALT (# participants affected): improvement from abnormal to normal range.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 5 ALT (# participants affected): improvement from abnormal to normal range.

6 ALT (U/L) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 6 ALT (U/L) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 6 ALT (U/L) at end of study.

7 AST (U/L) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.7

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 7 AST (U/L) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 7 AST (U/L) at end of study.

8 Serum creatinine (mg/dL) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.8

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 8 Serum creatinine (mg/dL) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 8 Serum creatinine (mg/dL) at end of study.

9 Blood urea (mg/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.9

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 9 Blood urea (mg/dL): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 9 Blood urea (mg/dL): mean at end of study.

10 Serum ferritin (ng/mL): mean change from baseline and at end of study Show forest plot

6

1002

Mean Difference (IV, Fixed, 95% CI)

454.42 [337.13, 571.71]

Analysis 2.10

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 10 Serum ferritin (ng/mL): mean change from baseline and at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 10 Serum ferritin (ng/mL): mean change from baseline and at end of study.

10.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

978.0 [544.71, 1411.29]

10.2 More than 3 to 7 mg Fe/g dw (10/35)

1

150

Mean Difference (IV, Fixed, 95% CI)

801.0 [572.53, 1029.47]

10.3 More than 7 mg Fe/g dw (20/41)

1

169

Mean Difference (IV, Fixed, 95% CI)

328.0 [124.94, 531.06]

10.4 More than 14 mg Fe/g dw (30/51)

1

216

Mean Difference (IV, Fixed, 95% CI)

77.0 [‐303.18, 457.18]

10.5 Any iron overload

5

439

Mean Difference (IV, Fixed, 95% CI)

234.25 [‐8.02, 476.52]

11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline Show forest plot

2

701

Mean Difference (IV, Fixed, 95% CI)

418.94 [297.23, 540.65]

Analysis 2.11

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline.

11.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30 mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

978.0 [544.71, 1411.29]

11.2 More than 3 to 7 mg Fe/g dw (10/35)

1

150

Mean Difference (IV, Fixed, 95% CI)

801.0 [572.53, 1029.47]

11.3 More than 7 mg Fe/g dw (20/41)

1

169

Mean Difference (IV, Fixed, 95% CI)

328.0 [124.94, 531.06]

11.4 More than 14 mg Fe/g dw (30/51)

1

216

Mean Difference (IV, Fixed, 95% CI)

77.0 [‐303.18, 457.18]

11.5 Any iron overload

1

138

Mean Difference (IV, Fixed, 95% CI)

‐64.16 [‐354.62, 226.30]

12 Liver R2* (Hz): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.12

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 12 Liver R2* (Hz): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 12 Liver R2* (Hz): mean change from baseline.

13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline Show forest plot

2

217

Mean Difference (IV, Fixed, 95% CI)

0.36 [‐1.01, 1.72]

Analysis 2.13

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline.

14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline Show forest plot

1

541

Mean Difference (IV, Fixed, 95% CI)

2.37 [1.68, 3.07]

Analysis 2.14

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline.

14.1 LIC 3 mg Fe/g dw or less (5/30)

1

28

Mean Difference (IV, Fixed, 95% CI)

4.3 [2.30, 6.30]

14.2 LIC more than 3 mg to 7 mg (10/35) Fe/g dw

1

143

Mean Difference (IV, Fixed, 95% CI)

3.80 [2.74, 4.86]

14.3 LIC more than 7 mg to 14 mg Fe/g dw (20/41)

1

164

Mean Difference (IV, Fixed, 95% CI)

1.5 [0.28, 2.72]

14.4 LIC more than 14 mg Fe/g dw (30/51)

1

206

Mean Difference (IV, Fixed, 95% CI)

‐2.5 [‐4.55, ‐0.45]

15 Responder analysis I (responder: fall in LIC > 10%) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.15

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 15 Responder analysis I (responder: fall in LIC > 10%).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 15 Responder analysis I (responder: fall in LIC > 10%).

15.1 Response at 48 weeks (deferasirox 10 mg/kg/day)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Response at 48 weeks (deferasirox 20 mg/kg/day)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw) Show forest plot

1

553

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.69, 0.92]

Analysis 2.16

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw).

16.1 Response at 1 year (LIC below 7 mg Fe/g dw)

1

172

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.37, 0.64]

16.2 Response at 1 year (LIC at least 7 mg Fe/g dw)

1

381

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.84, 1.18]

17 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.17

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 17 Myocardial T2* (ms): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 17 Myocardial T2* (ms): mean change from baseline.

18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.18

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline.

18.1 All participants

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Participants with T2* <10 ms

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Participants with T2* ≥10 ms

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

19 Myocardial T2* (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.19

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 19 Myocardial T2* (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 19 Myocardial T2* (# participants affected).

19.1 Normalization

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

19.2 Improvement (from 6 ‐ < 10 ms to 10 ‐ ≤ 20 ms)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

19.3 Worsening (from 10‐ ≤ 20 ms to 6 ‐ < 10 ms)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

20 Iron excretion‐intake ratio Show forest plot

1

541

Mean Difference (IV, Fixed, 95% CI)

‐0.18 [‐0.24, ‐0.12]

Analysis 2.20

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 20 Iron excretion‐intake ratio.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 20 Iron excretion‐intake ratio.

20.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30 mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

‐0.37 [‐0.54, ‐0.20]

20.2 More than 3 to 7 mg Fe/g dw (10/35)

1

143

Mean Difference (IV, Fixed, 95% CI)

‐0.31 [‐0.41, ‐0.21]

20.3 More than 7 mg Fe/g dw (20/41)

1

164

Mean Difference (IV, Fixed, 95% CI)

‐0.11 [‐0.21, ‐0.01]

20.4 More than 14 mg Fe/g dw (30/51)

1

206

Mean Difference (IV, Fixed, 95% CI)

0.23 [0.05, 0.41]

21 Any serious AEs (# participants affected) Show forest plot

2

773

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.42, 1.86]

Analysis 2.21

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 21 Any serious AEs (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 21 Any serious AEs (# participants affected).

22 Serious AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.22

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 22 Serious AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 22 Serious AEs.

22.1 Cardiac disorders ‐ arrhythmia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.2 Endocrine disorders ‐ hypogonadism

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.3 GI disorders abdominal abscess

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.4 GI disorders amoebiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.5 GI disorders ‐ appendicitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.6 GI disorders ‐ colitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.7 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.8 GI disorders ‐ gastric haemorrhage

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.9 GI disorders ‐ gastroenteritis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.10 GI disorders ‐ ileus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.11 GI disorders ‐ upper abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.12 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.13 GI disorders ‐ GI infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.14 General disorders and administration site conditions ‐ pyrexia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.15 General disorders and administration site conditions ‐ local swelling

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.16 Hepatobiliary disorders ‐ liver abscess

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.17 Hepatobiliary disorders ‐ cholelithiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.18 Immune system disorders ‐ face oedema

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.19 Infections and infestations ‐ herpes zoster

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.20 Infections and infestations ‐ tooth infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.21 Infections and infestations ‐ urinary tract infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.22 Injury, poisoning and procedural complications ‐ oesophageal rupture

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.23 Injury, poisoning and procedural complications ‐ haemosiderosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.24 Injury, poisoning and procedural complications ‐ iron overload

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.25 Metabolism and nutrition disorders ‐ hyperglycaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.26 Musculoskeletal and connective tissue disorders ‐ back pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.27 Musculoskeletal and connective tissue disorders ‐ pain in jaw

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.28 Nervous system disorders ‐ grand mal convulsion

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.29 Nervous system disorders ‐ meningitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.30 Respiratory, thoracic and mediastinal disorders ‐ acute tonsilitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

23 Any AE (# participants affected) Show forest plot

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.83, 1.08]

Analysis 2.23

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 23 Any AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 23 Any AE (# participants affected).

24 AEs Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.24

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 24 AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 24 AEs.

24.1 Blood and lymphatic system disorder ‐ agranulocytosis

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.2 Blood and lymphatic system disorder ‐ leukopenia

1

138

Risk Ratio (M‐H, Fixed, 95% CI)

7.0 [0.37, 133.02]

24.3 Blood and lymphatic system disorder ‐ neutropenia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.4 Blood and lymphatic system disorder ‐ thrombocytopenia

2

209

Risk Ratio (M‐H, Fixed, 95% CI)

4.0 [0.46, 34.88]

24.5 Cardiac disorders ‐ cardiac AE

1

586

Risk Ratio (M‐H, Fixed, 95% CI)

0.73 [0.38, 1.41]

24.6 Ear and labyrinth disorders ‐ hearing loss

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.41, 3.05]

24.7 Eye disorder ‐ lens abnormality

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

0.39 [0.08, 2.00]

24.8 Eye disorder ‐ retinal abnormality

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.9 GI disorders ‐ abdominal pain

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [0.93, 3.05]

24.10 GI disorders ‐ abdominal pain upper

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.36, 3.60]

24.11 GI disorders ‐ diarrhoea

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.65 [0.86, 3.16]

24.12 GI disorders ‐ dyspepsia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.25, 5.72]

24.13 GI disorders ‐ GIT upset

1

60

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.66, 13.69]

24.14 GI disorders ‐ nausea

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

2.59 [0.90, 7.47]

24.15 GI disorders ‐ vomiting

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.91, 9.55]

24.16 General disorders and administration site conditions ‐ asthenia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.42, 3.42]

24.17 General disorders and administration site conditions ‐ influenza‐like illness

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.42, 3.42]

24.18 General disorders and administration site conditions ‐ pyrexia

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [0.66, 2.44]

24.19 Immune system disorders ‐ allergic conjunctivitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

4.41 [0.25, 78.58]

24.20 Infections and infestations ‐ bronchitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

2.40 [0.30, 19.35]

24.21 Infections and infestations ‐ upper respiratory tract infection

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.37, 2.42]

24.22 Infections and infestations ‐ urinary tract infection

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

2.40 [0.30, 19.35]

24.23 Investigations ‐ ALT increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.59, 4.90]

24.24 Investigations ‐ elevated ALT (>2 UNL)

1

586

Risk Ratio (M‐H, Fixed, 95% CI)

4.90 [0.24, 101.60]

24.25 Investigations ‐ AST increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.21 [0.59, 8.29]

24.26 Investigations ‐ blood creatinine increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

3.79 [0.83, 17.38]

24.27 Investigations ‐ isolated serum creatinine increase above upper limit of normal

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

2.57 [1.88, 3.51]

24.28 Investigations ‐ platelet count increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.08, 1.91]

24.29 Musculoskeletal and connective tissue disorders ‐ arthralgia

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [0.55, 3.13]

24.30 Musculoskeletal and connective tissue disorders ‐ back pain

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.30 [0.73, 2.34]

24.31 Musculoskeletal and connective tissue disorders ‐ osteoporosis

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [0.47, 11.91]

24.32 Nervous system disorders ‐ headache

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.44 [0.68, 3.05]

24.33 Nervous system disorders ‐ vertigo

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.32, 3.93]

24.34 Renal and urinary disorders ‐ proteinuria

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.16 [0.50, 2.66]

24.35 Respiratory, thoracic and mediastinal disorders ‐ cough

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [0.67, 4.81]

24.36 Respiratory, thoracic and mediastinal disorders ‐ influenza

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.52, 2.13]

24.37 Respiratory, thoracic and mediastinal disorders ‐ nasopharyngitis

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.90 [0.59, 6.08]

24.38 Respiratory, thoracic and mediastinal disorders ‐ oropharyngeal pain

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.84 [0.59, 13.73]

24.39 Respiratory, thoracic and mediastinal disorders ‐ pharyngitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.52, 2.00]

24.40 Respiratory, thoracic and mediastinal disorders ‐ pharyngolaryngeal pain

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.37, 2.08]

24.41 Respiratory, thoracic and mediastinal disorders ‐ rhinitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.58, 2.83]

24.42 Skin and subcutaneous tissue disorders ‐ Rash

1

60

Risk Ratio (M‐H, Fixed, 95% CI)

2.67 [0.78, 9.09]

25 Any drug‐related AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.25

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 25 Any drug‐related AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 25 Any drug‐related AE (# participants affected).

26 Drug‐related AEs Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.26

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 26 Drug‐related AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 26 Drug‐related AEs.

26.1 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.2 Injury, poisoning and procedural complications ‐ infusion site haemorrhage

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.3 Injury, poisoning and procedural complications ‐ infusion site pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.4 Injury, poisoning and procedural complications ‐ infusion site swelling

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.5 Injury, poisoning and procedural complications ‐ injection site pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.6 Injury, poisoning and procedural complications ‐ injection site reaction

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.7 Investigations ‐ blood creatinine increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.8 Investigations ‐ ALT increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.9 Investigations ‐ AST increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.10 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.11 GI disorders ‐ abdominal pain upper

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.12 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.13 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.14 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.15 Immune system disorders ‐ hypersensitivity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.16 Immune system disorders ‐ urticaria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.17 Musculoskeletal and connective tissue disorders ‐ arthropathy

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.18 Renal and urinary disorders ‐ proteinuria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.19 Skin and subcutaneous tissue disorders ‐ alopecia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.20 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.21 Injury, poisoning and procedural complications ‐ pulmonary toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.22 Eye disorders ‐ Ophthalmological toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.23 Ear and labyrinth disorders ‐ Audiological toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27 Satisfaction with treatment (very satisfied or satisfied) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.27

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 27 Satisfaction with treatment (very satisfied or satisfied).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 27 Satisfaction with treatment (very satisfied or satisfied).

27.1 Week 4 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.2 Week 24 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.3 End of study (1 year) ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.4 Week 4 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.5 Week 24 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.6 End of study (1 year) ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28 Convenience (good or very good) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.28

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 28 Convenience (good or very good).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 28 Convenience (good or very good).

28.1 Week 4 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.2 Week 24 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.3 End of study (1 year) ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.4 Week 4 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.5 Week 24 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.6 End of study (1 year) ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

29 Willingness to continue treatment Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.29

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 29 Willingness to continue treatment.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 29 Willingness to continue treatment.

29.1 Participants treated previously with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

29.2 DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.30

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO.

30.1 week 4 ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.2 week 24 ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.3 end of study (1 year) ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.4 week 4 ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.5 week 24 ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.6 end of study (1 year) ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

31 Adherence (% of planned dose) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.31

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 31 Adherence (% of planned dose).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 31 Adherence (% of planned dose).

32 Discontinuations Show forest plot

8

1211

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.60, 1.50]

Analysis 2.32

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 32 Discontinuations.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 32 Discontinuations.

32.1 Deferasirox 10 mg/kg/day

1

35

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.01, 3.64]

32.2 Deferasirox 20 mg/kg/day

2

174

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.10, 9.96]

32.3 Deferasirox 25 mg/kg/day

1

120

Risk Ratio (M‐H, Fixed, 95% CI)

0.09 [0.01, 1.61]

32.4 Deferasirox 40 mg/kg/day

2

211

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.51, 2.05]

32.5 Deferasirox ‐ variable dosage

2

646

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.67, 2.85]

32.6 Deferasirox dosing unknown

1

25

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

33 Dose adjustments and dose interruptions Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.33

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 33 Dose adjustments and dose interruptions.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 33 Dose adjustments and dose interruptions.

34 Dose interruptions (interrupted at least once) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.34

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 34 Dose interruptions (interrupted at least once).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 34 Dose interruptions (interrupted at least once).

35 Dose reduction (at least once) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.35

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 35 Dose reduction (at least once).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 35 Dose reduction (at least once).

36 Dose adjustments (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.36

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 36 Dose adjustments (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 36 Dose adjustments (# participants affected).

37 Dose interruptions due to an AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.37

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 37 Dose interruptions due to an AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 37 Dose interruptions due to an AE (# participants affected).

38 Haemoglobin (g/dL): mean change from baseline and at end of study Show forest plot

2

180

Mean Difference (IV, Fixed, 95% CI)

‐0.52 [‐0.82, ‐0.21]

Analysis 2.38

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 38 Haemoglobin (g/dL): mean change from baseline and at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 38 Haemoglobin (g/dL): mean change from baseline and at end of study.

38.1 At 8 months (change from baseline)

1

138

Mean Difference (IV, Fixed, 95% CI)

‐0.46 [‐0.81, ‐0.11]

38.2 At 1 year (at end of study)

1

42

Mean Difference (IV, Fixed, 95% CI)

‐0.70 [‐1.33, ‐0.07]

39 Transfusion index (mL/kg/year): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.39

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 39 Transfusion index (mL/kg/year): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 39 Transfusion index (mL/kg/year): mean at end of study.

40 Transferrin saturation (%): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.40

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 40 Transferrin saturation (%): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 40 Transferrin saturation (%): mean at end of study.

41 Platelet count (x10³/mm³): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.41

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 41 Platelet count (x10³/mm³): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 41 Platelet count (x10³/mm³): mean at end of study.

42 Absolute neutrophilic count (/mm³): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.42

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 42 Absolute neutrophilic count (/mm³): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 42 Absolute neutrophilic count (/mm³): mean at end of study.

Open in table viewer
Comparison 3. Transfusion‐dependent thalassemia: deferasirox vs deferiprone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

3

146

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.1

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 1 Mortality at any time point.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 1 Mortality at any time point.

1.1 at 1 year

2

128

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 at 2 years

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Incidence of thyroid disease at end of study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 2 Incidence of thyroid disease at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 2 Incidence of thyroid disease at end of study.

3 ALT (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 3 ALT (U/L): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 3 ALT (U/L): mean change from baseline.

4 AST (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.4

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 4 AST (U/L): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 4 AST (U/L): mean change from baseline.

5 Urea (mg/dL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.5

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 5 Urea (mg/dL): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 5 Urea (mg/dL): mean change from baseline.

6 Creatinine (mg/dL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.6

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 6 Creatinine (mg/dL): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 6 Creatinine (mg/dL): mean change from baseline.

7 Neutrophil (count per mm³): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.7

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 7 Neutrophil (count per mm³): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 7 Neutrophil (count per mm³): mean change from baseline.

8 Serum ferritin (ng/mL): mean change from baseline and at end of study Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 3.8

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 8 Serum ferritin (ng/mL): mean change from baseline and at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 8 Serum ferritin (ng/mL): mean change from baseline and at end of study.

8.1 All participants

2

83

Mean Difference (IV, Fixed, 95% CI)

229.99 [‐403.14, 863.11]

8.2 Ferritin > 4000

1

11

Mean Difference (IV, Fixed, 95% CI)

1129.0 [‐2226.18, 4484.18]

8.3 Ferritin 2000 ‐ 4000

1

16

Mean Difference (IV, Fixed, 95% CI)

‐151.0 [‐743.80, 441.80]

8.4 Ferritin < 2000

1

11

Mean Difference (IV, Fixed, 95% CI)

388.0 [‐255.71, 1031.71]

9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.9

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

10 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.10

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 10 Myocardial T2* (ms): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 10 Myocardial T2* (ms): mean change from baseline.

11 Any AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.11

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 11 Any AE (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 11 Any AE (# participants affected).

12 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.12

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 12 AEs.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 12 AEs.

12.1 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 GI disorders ‐ pain abdomen

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.5 Musculoskeletal and connective tissue disorders ‐ arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.6 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.7 Blood and lymphatic system disorder ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.8 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.9 Investigations ‐ AST levels > 2x UNL

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.10 Investigations ‐ ALT levels > 2x UNL

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.11 Investigations ‐ serum creatinine 50% increase from baseline

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.12 Renal and urinary disorders ‐ renal failure

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Discontinuations (# participants affected) Show forest plot

3

179

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.01, 2.99]

Analysis 3.13

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 13 Discontinuations (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 13 Discontinuations (# participants affected).

14 Discontinuation due to an AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.14

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 14 Discontinuation due to an AE (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 14 Discontinuation due to an AE (# participants affected).

15 Transferrin saturation (%): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.15

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 15 Transferrin saturation (%): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 15 Transferrin saturation (%): mean at end of study.

16 Haemoglobin (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.16

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 16 Haemoglobin (g/dL): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 16 Haemoglobin (g/dL): mean at end of study.

17 Transfusion index (mL/kg/year): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.17

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 17 Transfusion index (mL/kg/year): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 17 Transfusion index (mL/kg/year): mean at end of study.

18 ALP (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.18

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 18 ALP (U/L): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 18 ALP (U/L): mean change from baseline.

Open in table viewer
Comparison 6. Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.1

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 1 Mortality at any time point.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 1 Mortality at any time point.

1.1 at 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Neutrophil (µg/L): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.2

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 2 Neutrophil (µg/L): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 2 Neutrophil (µg/L): mean at end of study.

3 ALT (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.3

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 3 ALT (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 3 ALT (g/dL): mean at end of study.

4 AST (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.4

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 4 AST (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 4 AST (g/dL): mean at end of study.

5 Serum ferritin: mean at end of study (ng/mL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.5

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 5 Serum ferritin: mean at end of study (ng/mL).

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 5 Serum ferritin: mean at end of study (ng/mL).

6 Discontinuations Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 6.6

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 6 Discontinuations.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 6 Discontinuations.

7 Haemoglobin (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.7

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 7 Haemoglobin (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 7 Haemoglobin (g/dL): mean at end of study.

8 ALP (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 6.8

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 8 ALP (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 8 ALP (g/dL): mean at end of study.

Open in table viewer
Comparison 7. Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.1

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 1 Mortality at any time point.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 1 Mortality at any time point.

1.1 at 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serum ferritin (ng/mL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 7.2

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 7.3

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

4 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 7.4

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 4 Myocardial T2* (ms): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 4 Myocardial T2* (ms): mean change from baseline.

5 Serious AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.5

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 5 Serious AE (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 5 Serious AE (# participants affected).

6 Serious drug‐related AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.6

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 6 Serious drug‐related AE (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 6 Serious drug‐related AE (# participants affected).

6.1 Cholecystitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious non‐related drug AE Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.7

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 7 Serious non‐related drug AE.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 7 Serious non‐related drug AE.

7.1 Appendicitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.8

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 8 AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 8 AEs.

8.1 Blood and lymphatic system disorder ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Drug‐related AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.9

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 9 Drug‐related AEs (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 9 Drug‐related AEs (# participants affected).

10 Drug‐related AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.10

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 10 Drug‐related AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 10 Drug‐related AEs.

10.1 Blood and lymphatic system disorders ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Blood and lymphatic system disorders ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 GI disorders ‐ GI problems

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Investigations ‐ ALT increase ( ≥ 3 folds)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Investigations ‐ serum creatinine ( ≥ 33%) above baseline in 2 consecutive occasions

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.6 Musculoskeletal and connective tissue disorders ‐ arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.7 Skin and subcutaneous tissue disorders ‐ skin rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Non‐related drug AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.11

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 11 Non‐related drug AEs (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 11 Non‐related drug AEs (# participants affected).

12 Non‐related drug AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.12

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 12 Non‐related drug AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 12 Non‐related drug AEs.

12.1 Infections and infestations ‐ infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 GI disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Skin and subcutaneous tissue disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Mild elevation of hepatic transaminases at start of therapy (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.13

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 13 Mild elevation of hepatic transaminases at start of therapy (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 13 Mild elevation of hepatic transaminases at start of therapy (# participants affected).

14 Initial gastrointestinal manifestations (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.14

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 14 Initial gastrointestinal manifestations (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 14 Initial gastrointestinal manifestations (# participants affected).

15 Quality of life (%) (measured by SF‐36): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 7.15

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 15 Quality of life (%) (measured by SF‐36): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 15 Quality of life (%) (measured by SF‐36): mean change from baseline.

16 Adherence: actual dose/total prescribed dose Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 7.16

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 16 Adherence: actual dose/total prescribed dose.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 16 Adherence: actual dose/total prescribed dose.

17 Discontinuations Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.17

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 17 Discontinuations.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 17 Discontinuations.

18 Serious AE resulting in study discontinuation or interruption Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 7.18

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 18 Serious AE resulting in study discontinuation or interruption.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 18 Serious AE resulting in study discontinuation or interruption.

Open in table viewer
Comparison 8. Non‐transfusion‐dependent thalassemia: deferasirox vs placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

148

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 8.1

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

1.1 Deferasirox 5 mg/kg/day: at 12 months

1

73

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Deferasirox 10 mg/kg/day: at 12 months

1

75

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serum ferritin (ng/mL): mean change from baseline Show forest plot

1

154

Mean Difference (IV, Fixed, 95% CI)

‐306.74 [‐398.23, ‐215.24]

Analysis 8.2

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

2.1 Deferasirox 5 mg/kg/day

1

78

Mean Difference (IV, Fixed, 95% CI)

‐259.1 [‐377.35, ‐140.85]

2.2 Deferasirox 10 mg/kg/day

1

76

Mean Difference (IV, Fixed, 95% CI)

‐377.79 [‐522.21, ‐233.37]

3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline Show forest plot

1

159

Mean Difference (IV, Fixed, 95% CI)

‐3.27 [‐4.44, ‐2.09]

Analysis 8.3

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline.

3.1 Deferasirox 5 mg/kg/day

1

78

Mean Difference (IV, Fixed, 95% CI)

‐2.33 [‐4.00, ‐0.66]

3.2 Deferasirox 10 mg/kg/day

1

81

Mean Difference (IV, Fixed, 95% CI)

‐4.18 [‐5.83, ‐2.53]

4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 8.4

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline.

4.1 Non‐transfusion‐dependent β‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Non‐transfusion‐dependent β‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 α‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 α‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 HbE/β‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 HbE/β‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 < 18 years 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.8 < 18 years 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.9 ≥18 years 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.10 ≥18 years 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

4.16 [1.90, 9.11]

Analysis 8.5

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected).

5.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.05 [0.98, 9.50]

5.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

5.26 [1.76, 15.71]

6 LIC: ≥ 30% reduction Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

14.17 [2.88, 69.74]

Analysis 8.6

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 6 LIC: ≥ 30% reduction Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 6 LIC: ≥ 30% reduction Fe/g dw (# participants affected).

6.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

15.02 [0.93, 242.87]

6.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

13.75 [1.97, 95.97]

7 LIC: shift to lower iron burden range (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

3.35 [1.62, 6.91]

Analysis 8.7

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 7 LIC: shift to lower iron burden range (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 7 LIC: shift to lower iron burden range (# participants affected).

7.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.39 [1.10, 10.45]

7.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.31 [1.28, 8.55]

8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

5.35 [1.30, 21.99]

Analysis 8.8

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected).

8.1 deferasirox 5mg/kg/d

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

4.07 [0.54, 30.96]

8.2 deferasirox 10mg/kg/d

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

6.62 [0.91, 48.05]

9 Drug‐related serious AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.9

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 9 Drug‐related serious AEs (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 9 Drug‐related serious AEs (# participants affected).

10 Drug‐related serious AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.10

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 10 Drug‐related serious AEs (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 10 Drug‐related serious AEs (# participants affected).

10.1 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Infections and infestations ‐ cellulitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Skin and subcutaneous tissue disorders ‐ pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 General disorders ‐ pyrexia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.6 Hepatobiliary disorders ‐ hepatotoxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Any AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.11

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 11 Any AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 11 Any AE (# participants affected).

12 Mild AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.12

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 12 Mild AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 12 Mild AE (# participants affected).

13 Moderate AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.13

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 13 Moderate AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 13 Moderate AE (# participants affected).

14 Severe AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.14

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 14 Severe AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 14 Severe AE (# participants affected).

15 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.15

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 15 AEs.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 15 AEs.

15.1 Ear and labyrinth disorders ‐ neurosensory deafness

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Investigations ‐ abnormal platelet count (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Investigations ‐ abnormal neutrophils count (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.4 Investigations ‐ abnormal ALT (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.5 Investigations ‐ abnormal AST (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.6 Investigations ‐ abnormal serum creatinine (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.7 Investigations ‐ abnormal creatinine clearance (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.8 Investigations ‐ urinary protein/creatinine ratio (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.9 Investigations ‐ abnormal (low) systolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.10 Investigations ‐ abnormal (high) systolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.11 Investigations ‐ abnormal (low) diastolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.12 Investigations ‐ abnormal (high) diastolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.13 Investigations ‐ abnormal (low) pulse rate (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.14 Investigations ‐ abnormal (high) pulse rate (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.15 Renal and urinary disorders ‐ proteinuria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Drug‐related AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.16

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 16 Drug‐related AEs.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 16 Drug‐related AEs.

16.1 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 GI disorders ‐ abdominal pain upper

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.4 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.5 Nervous system disorders ‐ headache

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.6 Skin and subcutaneous tissue disorders ‐ skin rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Adherence (# participants taking the planned study dose) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.17

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 17 Adherence (# participants taking the planned study dose).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 17 Adherence (# participants taking the planned study dose).

18 Discontinuations Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.32 [0.50, 3.52]

Analysis 8.18

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 18 Discontinuations.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 18 Discontinuations.

18.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.33, 4.25]

18.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.33, 7.08]

19 Discontinuing study due to AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 8.19

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 19 Discontinuing study due to AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 19 Discontinuing study due to AE (# participants affected).

20 Dose increase Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

0.87 [0.63, 1.19]

Analysis 8.20

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 20 Dose increase.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 20 Dose increase.

20.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.57, 1.38]

20.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.54, 1.33]

21 Dose interruption (at least once) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.88, 1.39]

Analysis 8.21

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 21 Dose interruption (at least once).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 21 Dose interruption (at least once).

21.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.73, 1.43]

21.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.87, 1.62]

22 Dose reduction Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [0.70, 3.06]

Analysis 8.22

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 22 Dose reduction.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 22 Dose reduction.

22.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.54, 4.30]

22.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.4 [0.49, 4.00]

23 Dose reduction due to AE Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.69, 3.38]

Analysis 8.23

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 23 Dose reduction due to AE.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 23 Dose reduction due to AE.

23.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

2.04 [0.63, 6.63]

23.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.39, 3.39]

24 Haemoglobin (g/L): mean change from baseline Show forest plot

1

144

Mean Difference (IV, Fixed, 95% CI)

1.54 [‐0.82, 3.90]

Analysis 8.24

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 24 Haemoglobin (g/L): mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 24 Haemoglobin (g/L): mean change from baseline.

24.1 Deferasirox 5 mg/kg/day

1

71

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐2.31, 4.31]

24.2 Deferasirox 10 mg/kg/day

1

73

Mean Difference (IV, Fixed, 95% CI)

2.10 [‐1.27, 5.47]

25 Transferrin saturation (%): mean change from baseline Show forest plot

1

141

Mean Difference (IV, Fixed, 95% CI)

‐7.10 [‐11.71, ‐2.50]

Analysis 8.25

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 25 Transferrin saturation (%): mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 25 Transferrin saturation (%): mean change from baseline.

25.1 Deferasirox 5 mg/kg/day

1

70

Mean Difference (IV, Fixed, 95% CI)

‐7.16 [‐12.95, ‐1.37]

25.2 Deferasirox 10 mg/kg/day

1

71

Mean Difference (IV, Fixed, 95% CI)

‐7.01 [‐14.59, 0.57]

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 1.1

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 AEs.
Figures and Tables -
Analysis 1.2

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 AEs.

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 Discontinuations due to serious AEs.
Figures and Tables -
Analysis 1.3

Comparison 1 Transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 Discontinuations due to serious AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 2.1

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 1 Mortality at any time point.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 2 LVEF (%): least squares mean change from baseline.
Figures and Tables -
Analysis 2.2

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 2 LVEF (%): least squares mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 3 LVEF (# participants affected).
Figures and Tables -
Analysis 2.3

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 3 LVEF (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 4 Incidence of thyroid disease at end of study.
Figures and Tables -
Analysis 2.4

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 4 Incidence of thyroid disease at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 5 ALT (# participants affected): improvement from abnormal to normal range.
Figures and Tables -
Analysis 2.5

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 5 ALT (# participants affected): improvement from abnormal to normal range.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 6 ALT (U/L) at end of study.
Figures and Tables -
Analysis 2.6

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 6 ALT (U/L) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 7 AST (U/L) at end of study.
Figures and Tables -
Analysis 2.7

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 7 AST (U/L) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 8 Serum creatinine (mg/dL) at end of study.
Figures and Tables -
Analysis 2.8

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 8 Serum creatinine (mg/dL) at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 9 Blood urea (mg/dL): mean at end of study.
Figures and Tables -
Analysis 2.9

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 9 Blood urea (mg/dL): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 10 Serum ferritin (ng/mL): mean change from baseline and at end of study.
Figures and Tables -
Analysis 2.10

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 10 Serum ferritin (ng/mL): mean change from baseline and at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline.
Figures and Tables -
Analysis 2.11

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 12 Liver R2* (Hz): mean change from baseline.
Figures and Tables -
Analysis 2.12

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 12 Liver R2* (Hz): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline.
Figures and Tables -
Analysis 2.13

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline.
Figures and Tables -
Analysis 2.14

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 15 Responder analysis I (responder: fall in LIC > 10%).
Figures and Tables -
Analysis 2.15

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 15 Responder analysis I (responder: fall in LIC > 10%).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw).
Figures and Tables -
Analysis 2.16

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 17 Myocardial T2* (ms): mean change from baseline.
Figures and Tables -
Analysis 2.17

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 17 Myocardial T2* (ms): mean change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline.
Figures and Tables -
Analysis 2.18

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 19 Myocardial T2* (# participants affected).
Figures and Tables -
Analysis 2.19

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 19 Myocardial T2* (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 20 Iron excretion‐intake ratio.
Figures and Tables -
Analysis 2.20

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 20 Iron excretion‐intake ratio.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 21 Any serious AEs (# participants affected).
Figures and Tables -
Analysis 2.21

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 21 Any serious AEs (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 22 Serious AEs.
Figures and Tables -
Analysis 2.22

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 22 Serious AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 23 Any AE (# participants affected).
Figures and Tables -
Analysis 2.23

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 23 Any AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 24 AEs.
Figures and Tables -
Analysis 2.24

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 24 AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 25 Any drug‐related AE (# participants affected).
Figures and Tables -
Analysis 2.25

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 25 Any drug‐related AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 26 Drug‐related AEs.
Figures and Tables -
Analysis 2.26

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 26 Drug‐related AEs.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 27 Satisfaction with treatment (very satisfied or satisfied).
Figures and Tables -
Analysis 2.27

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 27 Satisfaction with treatment (very satisfied or satisfied).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 28 Convenience (good or very good).
Figures and Tables -
Analysis 2.28

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 28 Convenience (good or very good).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 29 Willingness to continue treatment.
Figures and Tables -
Analysis 2.29

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 29 Willingness to continue treatment.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO.
Figures and Tables -
Analysis 2.30

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 31 Adherence (% of planned dose).
Figures and Tables -
Analysis 2.31

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 31 Adherence (% of planned dose).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 32 Discontinuations.
Figures and Tables -
Analysis 2.32

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 32 Discontinuations.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 33 Dose adjustments and dose interruptions.
Figures and Tables -
Analysis 2.33

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 33 Dose adjustments and dose interruptions.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 34 Dose interruptions (interrupted at least once).
Figures and Tables -
Analysis 2.34

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 34 Dose interruptions (interrupted at least once).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 35 Dose reduction (at least once).
Figures and Tables -
Analysis 2.35

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 35 Dose reduction (at least once).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 36 Dose adjustments (# participants affected).
Figures and Tables -
Analysis 2.36

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 36 Dose adjustments (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 37 Dose interruptions due to an AE (# participants affected).
Figures and Tables -
Analysis 2.37

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 37 Dose interruptions due to an AE (# participants affected).

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 38 Haemoglobin (g/dL): mean change from baseline and at end of study.
Figures and Tables -
Analysis 2.38

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 38 Haemoglobin (g/dL): mean change from baseline and at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 39 Transfusion index (mL/kg/year): mean at end of study.
Figures and Tables -
Analysis 2.39

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 39 Transfusion index (mL/kg/year): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 40 Transferrin saturation (%): mean at end of study.
Figures and Tables -
Analysis 2.40

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 40 Transferrin saturation (%): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 41 Platelet count (x10³/mm³): mean at end of study.
Figures and Tables -
Analysis 2.41

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 41 Platelet count (x10³/mm³): mean at end of study.

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 42 Absolute neutrophilic count (/mm³): mean at end of study.
Figures and Tables -
Analysis 2.42

Comparison 2 Transfusion‐dependent thalassemia: deferasirox vs deferoxamine, Outcome 42 Absolute neutrophilic count (/mm³): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 3.1

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 1 Mortality at any time point.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 2 Incidence of thyroid disease at end of study.
Figures and Tables -
Analysis 3.2

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 2 Incidence of thyroid disease at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 3 ALT (U/L): mean change from baseline.
Figures and Tables -
Analysis 3.3

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 3 ALT (U/L): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 4 AST (U/L): mean change from baseline.
Figures and Tables -
Analysis 3.4

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 4 AST (U/L): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 5 Urea (mg/dL): mean change from baseline.
Figures and Tables -
Analysis 3.5

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 5 Urea (mg/dL): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 6 Creatinine (mg/dL): mean change from baseline.
Figures and Tables -
Analysis 3.6

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 6 Creatinine (mg/dL): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 7 Neutrophil (count per mm³): mean change from baseline.
Figures and Tables -
Analysis 3.7

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 7 Neutrophil (count per mm³): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 8 Serum ferritin (ng/mL): mean change from baseline and at end of study.
Figures and Tables -
Analysis 3.8

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 8 Serum ferritin (ng/mL): mean change from baseline and at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.
Figures and Tables -
Analysis 3.9

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 10 Myocardial T2* (ms): mean change from baseline.
Figures and Tables -
Analysis 3.10

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 10 Myocardial T2* (ms): mean change from baseline.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 11 Any AE (# participants affected).
Figures and Tables -
Analysis 3.11

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 11 Any AE (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 12 AEs.
Figures and Tables -
Analysis 3.12

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 12 AEs.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 13 Discontinuations (# participants affected).
Figures and Tables -
Analysis 3.13

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 13 Discontinuations (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 14 Discontinuation due to an AE (# participants affected).
Figures and Tables -
Analysis 3.14

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 14 Discontinuation due to an AE (# participants affected).

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 15 Transferrin saturation (%): mean at end of study.
Figures and Tables -
Analysis 3.15

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 15 Transferrin saturation (%): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 16 Haemoglobin (g/dL): mean at end of study.
Figures and Tables -
Analysis 3.16

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 16 Haemoglobin (g/dL): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 17 Transfusion index (mL/kg/year): mean at end of study.
Figures and Tables -
Analysis 3.17

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 17 Transfusion index (mL/kg/year): mean at end of study.

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 18 ALP (U/L): mean change from baseline.
Figures and Tables -
Analysis 3.18

Comparison 3 Transfusion‐dependent thalassemia: deferasirox vs deferiprone, Outcome 18 ALP (U/L): mean change from baseline.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 6.1

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 1 Mortality at any time point.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 2 Neutrophil (µg/L): mean at end of study.
Figures and Tables -
Analysis 6.2

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 2 Neutrophil (µg/L): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 3 ALT (g/dL): mean at end of study.
Figures and Tables -
Analysis 6.3

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 3 ALT (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 4 AST (g/dL): mean at end of study.
Figures and Tables -
Analysis 6.4

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 4 AST (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 5 Serum ferritin: mean at end of study (ng/mL).
Figures and Tables -
Analysis 6.5

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 5 Serum ferritin: mean at end of study (ng/mL).

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 6 Discontinuations.
Figures and Tables -
Analysis 6.6

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 6 Discontinuations.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 7 Haemoglobin (g/dL): mean at end of study.
Figures and Tables -
Analysis 6.7

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 7 Haemoglobin (g/dL): mean at end of study.

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 8 ALP (g/dL): mean at end of study.
Figures and Tables -
Analysis 6.8

Comparison 6 Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine, Outcome 8 ALP (g/dL): mean at end of study.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 7.1

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 1 Mortality at any time point.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.
Figures and Tables -
Analysis 7.2

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.
Figures and Tables -
Analysis 7.3

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 4 Myocardial T2* (ms): mean change from baseline.
Figures and Tables -
Analysis 7.4

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 4 Myocardial T2* (ms): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 5 Serious AE (# participants affected).
Figures and Tables -
Analysis 7.5

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 5 Serious AE (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 6 Serious drug‐related AE (# participants affected).
Figures and Tables -
Analysis 7.6

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 6 Serious drug‐related AE (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 7 Serious non‐related drug AE.
Figures and Tables -
Analysis 7.7

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 7 Serious non‐related drug AE.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 8 AEs.
Figures and Tables -
Analysis 7.8

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 8 AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 9 Drug‐related AEs (# participants affected).
Figures and Tables -
Analysis 7.9

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 9 Drug‐related AEs (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 10 Drug‐related AEs.
Figures and Tables -
Analysis 7.10

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 10 Drug‐related AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 11 Non‐related drug AEs (# participants affected).
Figures and Tables -
Analysis 7.11

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 11 Non‐related drug AEs (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 12 Non‐related drug AEs.
Figures and Tables -
Analysis 7.12

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 12 Non‐related drug AEs.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 13 Mild elevation of hepatic transaminases at start of therapy (# participants affected).
Figures and Tables -
Analysis 7.13

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 13 Mild elevation of hepatic transaminases at start of therapy (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 14 Initial gastrointestinal manifestations (# participants affected).
Figures and Tables -
Analysis 7.14

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 14 Initial gastrointestinal manifestations (# participants affected).

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 15 Quality of life (%) (measured by SF‐36): mean change from baseline.
Figures and Tables -
Analysis 7.15

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 15 Quality of life (%) (measured by SF‐36): mean change from baseline.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 16 Adherence: actual dose/total prescribed dose.
Figures and Tables -
Analysis 7.16

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 16 Adherence: actual dose/total prescribed dose.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 17 Discontinuations.
Figures and Tables -
Analysis 7.17

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 17 Discontinuations.

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 18 Serious AE resulting in study discontinuation or interruption.
Figures and Tables -
Analysis 7.18

Comparison 7 Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine, Outcome 18 Serious AE resulting in study discontinuation or interruption.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.
Figures and Tables -
Analysis 8.1

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 1 Mortality at any time point.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.
Figures and Tables -
Analysis 8.2

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 2 Serum ferritin (ng/mL): mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline.
Figures and Tables -
Analysis 8.3

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline.
Figures and Tables -
Analysis 8.4

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected).
Figures and Tables -
Analysis 8.5

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 6 LIC: ≥ 30% reduction Fe/g dw (# participants affected).
Figures and Tables -
Analysis 8.6

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 6 LIC: ≥ 30% reduction Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 7 LIC: shift to lower iron burden range (# participants affected).
Figures and Tables -
Analysis 8.7

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 7 LIC: shift to lower iron burden range (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected).
Figures and Tables -
Analysis 8.8

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 9 Drug‐related serious AEs (# participants affected).
Figures and Tables -
Analysis 8.9

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 9 Drug‐related serious AEs (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 10 Drug‐related serious AEs (# participants affected).
Figures and Tables -
Analysis 8.10

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 10 Drug‐related serious AEs (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 11 Any AE (# participants affected).
Figures and Tables -
Analysis 8.11

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 11 Any AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 12 Mild AE (# participants affected).
Figures and Tables -
Analysis 8.12

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 12 Mild AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 13 Moderate AE (# participants affected).
Figures and Tables -
Analysis 8.13

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 13 Moderate AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 14 Severe AE (# participants affected).
Figures and Tables -
Analysis 8.14

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 14 Severe AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 15 AEs.
Figures and Tables -
Analysis 8.15

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 15 AEs.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 16 Drug‐related AEs.
Figures and Tables -
Analysis 8.16

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 16 Drug‐related AEs.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 17 Adherence (# participants taking the planned study dose).
Figures and Tables -
Analysis 8.17

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 17 Adherence (# participants taking the planned study dose).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 18 Discontinuations.
Figures and Tables -
Analysis 8.18

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 18 Discontinuations.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 19 Discontinuing study due to AE (# participants affected).
Figures and Tables -
Analysis 8.19

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 19 Discontinuing study due to AE (# participants affected).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 20 Dose increase.
Figures and Tables -
Analysis 8.20

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 20 Dose increase.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 21 Dose interruption (at least once).
Figures and Tables -
Analysis 8.21

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 21 Dose interruption (at least once).

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 22 Dose reduction.
Figures and Tables -
Analysis 8.22

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 22 Dose reduction.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 23 Dose reduction due to AE.
Figures and Tables -
Analysis 8.23

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 23 Dose reduction due to AE.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 24 Haemoglobin (g/L): mean change from baseline.
Figures and Tables -
Analysis 8.24

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 24 Haemoglobin (g/L): mean change from baseline.

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 25 Transferrin saturation (%): mean change from baseline.
Figures and Tables -
Analysis 8.25

Comparison 8 Non‐transfusion‐dependent thalassemia: deferasirox vs placebo, Outcome 25 Transferrin saturation (%): mean change from baseline.

Summary of findings for the main comparison. Deferasirox compared to deferoxamine in people with transfusion‐dependent thalassemia

Deferasirox compared to deferoxamine in people with transfusion‐dependent thalassemia

Patient or population: people with transfusion‐dependent thalassemia
Setting: outpatient care
Intervention: deferasirox
Comparison: deferoxamine

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferoxamine

Risk with deferasirox

Mortality at any time point

Study population

RR 0.48
(0.09 to 2.63)

1170
(8 RCTs)

⊕⊕⊝⊝
LOW 1 2

7 per 1.000

3 per 1.000
(1 to 18)

Responder analysis II (responder: LIC 1 to less than 7 mg Fe/g dw)

Study population

RR 0.80
(0.69 to 0.92)

553
(1 RCT)

⊕⊕⊕⊝
MODERATE 1 3 4

664 per 1.000

531 per 1.000
(458 to 611)

Serum ferritin (ng/mL): mean change from baseline and at end of study

MD 454.42 higher
(337.13 higher to 571.71 higher)

1002
(6 RCTs) 5

⊕⊕⊕⊝
MODERATE 1 3 4

LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline

MD 2.37 higher
(1.68 higher to 3.07 higher)

541
(1 RCT)

⊕⊕⊕⊝
MODERATE 1 3 4

Satisfaction with treatment (very satisfied or satisfied): participants previously treated with DFO
assessed with: questionnaire
follow up: mean 52 weeks

Study population

RR 2.20
(1.89 to 2.57)

571
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

1.330 per 1.000

1000 per 1.000
(1.000 to 1.000)

Adherence: discontinuations

Study population

RR 0.95
(0.60 to 1.50)

1211
(8 RCTs)

⊕⊕⊝⊝
LOW 1 6

54 per 1.000

52 per 1.000
(33 to 82)

AE: investigations ‐ isolated serum creatinine increase above ULN

Study population

RR 2.57
(1.88 to 3.51)

657
(2 RCTs)

⊕⊕⊝⊝
LOW 1 7

137 per 1.000

353 per 1.000
(258 to 482)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; DFO:deferiprone; dw: dry weight; FE: iron LIC: liver iron concentration; MD: mean difference; RR: risk ratio; SQUID: superconducting quantum interference device; ULN: upper limit of normal.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Serious risk of bias: studies that carry large weight for the overall effect estimate rated as high risk of bias due to lack of blinding and selective reporting.
2 Serious imprecision: wide confidence interval including both clinically relevant benefit as well as harm.
3 Serious inconsistency: differing ratio of drugs between subgroups of one study.
4 Upgrade due to dose‐response gradient: observed for both drugs. Effects therefore depending on ratio of drugs used in comparisons.
5 A sensitivity analysis without the results from four studies which were calculated according to Wan 2014 showed similar results.
6 Serious imprecision: Wide confidence interval, including less discontinuations with deferoxamine treatment.
7 Serious indirectness: Surrogate of creatinine used for patient‐important outcome of kidney failure.

Figures and Tables -
Summary of findings for the main comparison. Deferasirox compared to deferoxamine in people with transfusion‐dependent thalassemia
Summary of findings 2. Deferasirox compared to deferiprone in people with transfusion‐dependent thalassemia

Deferasirox compared to deferiprone in people with transfusion‐dependent thalassemia

Patient or population:people with transfusion‐dependent thalassemia
Setting: outpatient care
Intervention: deferasirox
Comparison: deferiprone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferiprone

Risk with deferasirox

Mortality at any time point

Study population

not estimable

146
(3 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

0 per 1.000

0 per 1.000
(0 to 0)

Responder analysis

Not measured

NA

Serum ferritin (ng/mL): mean change from baseline and at end of study

MD 229.99 ng/mL higher
(403.14 lower to 863.11 higher)

83
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 3 4

LIC (mg/g) evaluated by MRI R2*: mean change from baseline

MD 0.8 mg/g lower
(2.75 lower to 1.15 higher)

45
(1 RCT)

⊕⊝⊝⊝
VERY LOW 3 4

Satisfaction

Not measured

NA

Adherence: discontinuations

Study population

RR 0.16
(0.01 to 2.99)

179
(3 RCTs)

⊕⊕⊝⊝
LOW 2 5

32 per 1.000

5 per 1.000
(0 to 95)

Renal failure

Study population

not estimable

38
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 6

0 per 1.000

0 per 1.000
(0 to 0)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; LIC: liver iron concentration; MD: mean difference; MRI: magnetic resonance imaging; NA: not applicable RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Very serious imprecision: only very few number of included participants.
2 Serious risk of bias: selective reporting: Results from Elalfy 2015a for 60 participants not reported.
3 Serious risk of bias: no blinding assumed, selective reporting: Results from Elalfy 2015a for 60 participants not reported.
4 Very serious imprecision: very wide confidence interval including both relevant benefit as well as harm.
5 Serious imprecision: wide CIs including both benefit as well as harm.
6 Serious risk of bias: no blinding assumed.

Figures and Tables -
Summary of findings 2. Deferasirox compared to deferiprone in people with transfusion‐dependent thalassemia
Summary of findings 3. Deferasirox alone compared to combined deferasirox and deferiprone in people with transfusion‐dependent thalassemia

Deferasirox alone compared to combined deferasirox and deferiprone in people with transfusion‐dependent thalassaemia

Patient or population: people with transfusion‐dependent thalassaemia
Setting: outpatient care
Intervention: deferasirox
Comparison: deferasirox and deferiprone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferasirox and deferiprone

Risk with deferasirox

Mortality at any time point

Not reported1

NA

Responder analysis

Not measured1

NA

Serum ferritin (ng/mL)

Not reported1

NA

LIC (mg Fe/g dry weight)

Not reported1

NA

Satisfaction with treatment

Not measured1

NA

Adherence

Not reported1

NA

AE: serum creatinine increase

Not measured1

NA

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; LIC: liver iron concentration; MD: mean difference; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 One RCT (40 participants) contributed to this comparison, but no relevant outcome data to this table are available (Kakkar 2014).

Figures and Tables -
Summary of findings 3. Deferasirox alone compared to combined deferasirox and deferiprone in people with transfusion‐dependent thalassemia
Summary of findings 4. Combined deferasirox and deferiprone compared to deferiprone alone in people with transfusion‐dependent thalassemia

Combined deferasirox and deferiprone compared to deferiprone alone in people with transfusion‐dependent thalassaemia

Patient or population: people with transfusion‐dependent thalassemia
Setting: outpatient care
Intervention: deferasirox and deferiprone
Comparison: deferiprone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferasirox and deferiprone

Risk with deferasirox

Mortality at any time point

Not reported1

NA

Responder analysis

Not measured1

NA

Serum ferritin (ng/mL)

Not reported1

NA

LIC (mg Fe/g dry weight)

Not reported1

NA

Satisfaction with treatment

Not measured1

NA

Adherence

Not reported1

NA

AE: Serum creatinine increase

Not measured1

NA

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; LIC: liver iron concentration; MD: mean difference; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 One RCT (40 participants) contributed to this comparison, but no relevant outcome data to this table are available (Kakkar 2014).

Figures and Tables -
Summary of findings 4. Combined deferasirox and deferiprone compared to deferiprone alone in people with transfusion‐dependent thalassemia
Summary of findings 5. Deferasirox and deferoxamine compared to deferoxamine in people with transfusion‐dependent thalassemia

Deferasirox and deferoxamine compared to deferoxamine in people with transfusion‐dependent thalassemia

Patient or population: people with transfusion‐dependent thalassemia
Setting:outpatient care
Intervention: deferasirox and deferoxamine
Comparison: deferoxamine

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferoxamine

Risk with deferasirox and deferoxamine

Mortality at any time point

Study population

not estimable

94
(1 RCT)

⊕⊕⊝⊝
LOW 1

0 per 1.000

0 per 1.000
(0 to 0)

Responder analysis

Not measured

NA

Serum ferritin (ng/mL) ‐ mean at end of study

MD 87.84 ng/mL higher
(612.23 lower to 787.91 higher)

94
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2 3

LIC

Not measured

NA

Satisfaction with treatment

Not measured

NA

Adherence: Discontinuations

Study population

not estimable

94
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

0 per 1.000

0 per 1.000
(0 to 0)

AE: serum creatinine increased

Not reported

NA

Serum creatinine was measured in Molavi 2014, but no results were reported.

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; LIC: liver iron concentration; MD: mean difference; NA: not applicable; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Very serious imprecision: only very few participants included.
2 Serious risk of bias: assumed lack of blinding.
3 Very serious imprecision: very wide confidence interval including both benefit as well as harm.

Figures and Tables -
Summary of findings 5. Deferasirox and deferoxamine compared to deferoxamine in people with transfusion‐dependent thalassemia
Summary of findings 6. Deferasirox and deferiprone compared to deferiprone and deferoxamine in people with transfusion‐dependent thalassemia

Deferasirox and deferiprone compared to deferiprone and deferoxamine in people with transfusion‐dependent thalassemia

Patient or population: people with transfusion‐dependent thalassemia
Setting: outpatient care
Intervention: deferasirox and deferiprone
Comparison: deferiprone and deferoxamine

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with deferiprone and deferoxamine

Risk with deferasirox and deferiprone

Mortality at any time point

Study population

not estimable

96
(1 RCT)

⊕⊕⊝⊝
LOW 1

"All the included patients continued till the end of study with no patients were lost follow‐up." (Elalfy 2015b)

0 per 1.000

0 per 1.000
(0 to 0)

Responder analysis

Not measured

NA

Serum ferritin (ng/mL): mean change from baseline

MD 315.9 ng/mL lower
(1046.26 lower to 414.46 higher)

96
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2 3

LIC evaluated by MRI R2*: mean change from baseline

MD 0.62 mg/g lower
(2.25 lower to 1.01 higher)

96
(1 RCT)

⊕⊕⊝⊝
LOW 2 4

Satisfaction

Not reported

NA

"Compared to baseline, patient‐reported satisfaction associated with ICT was significantly higher in group B [DFX and DFP] compared to group A [DFP and DFO] (p<0.01)" (Elalfy 2015b)

Adherence: Discontinuations

Study population

not estimable

96
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

0 per 1.000

0 per 1.000
(0 to 0)

Drug‐related AE: serum creatinine increased (≥ 33%) above baseline in 2 consecutive occasions

Study population

RR 3.00
(0.32 to 27.83)

96
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2 3 5

21 per 1.000

63 per 1.000
(7 to 580)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; LIC: liver iron concentration; MD: mean difference; MRI: magnetic resonance imaging; NA: not applicable; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Very serious imprecision: very few participants included.
2 Serious risk of bias: no blinding, selective reporting: no data for 18 months follow‐up.
3 Very serious imprecision: very wide confidence interval including both benefit as well as harm.
4 Serious imprecision: wide confidence interval including both benefit as well as harm.
5 Serious indirectness: surrogate of creatinine used for patient‐important outcome of kidney failure.

Figures and Tables -
Summary of findings 6. Deferasirox and deferiprone compared to deferiprone and deferoxamine in people with transfusion‐dependent thalassemia
Summary of findings 7. Deferasirox compared to placebo in people with non‐transfusion‐dependent thalassemia

Deferasirox compared to placebo in people with non‐transfusion‐dependent thalassemia

Patient or population: people with non‐transfusion‐dependent thalassemia
Setting: outpatient care
Intervention: deferasirox
Comparison: placebo

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with placebo

Risk with deferasirox

Mortality at any time point

Study population

not estimable

148
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

"No deaths occurred during the study in any group" (Taher 2012)

0 per 1.000

0 per 1.000
(0 to 0)

Responder analysis

Not measured

NA

Serum ferritin (ng/mL): mean change from baseline

MD 306.74 ng/mL lower
(398.23 lower to 215.24 lower)

154
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

LIC (mg Fe/g dry weight) evaluated by MRI R2: least squares mean change from baseline

MD 3.27 mg Fe/g dry weight lower
(4.44 lower to 2.09 lower)

159
(1 RCT)

⊕⊕⊕⊝
MODERATE 1

Satisfaction with treatment

Not measured

NA

Adherence: Discontinuations

Study population

RR 1.32
(0.50 to 3.52)

166
(1 RCT)

⊕⊕⊝⊝
LOW 2

89 per 1.000

118 per 1.000
(45 to 314)

AE: abnormal serum creatinine (post‐baseline)

Study population

RR 3.59
(0.19 to 68.40)

166
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2 3

0 per 1.000

0 per 1.000
(0 to 0)

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

AE: adverse events; CI: confidence interval; LIC: liver iron concentration; MD: mean difference; NA: not applicable; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Serious imprecision: only few patients included.
2 Very serious imprecision: very wide confidence interval, including both benefit as well as harm.
3 Serious indirectness: surrogate of creatinine used for patient‐important outcome of kidney failure.

Figures and Tables -
Summary of findings 7. Deferasirox compared to placebo in people with non‐transfusion‐dependent thalassemia
Comparison 1. Transfusion‐dependent thalassemia: deferasirox vs placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

2

47

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.1 Eye disorders ‐ retinal infarct

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.2 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.3 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.4 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.5 Investigations ‐ extended QT interval, hypocalcaemia, hypoparathyroidism

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2.6 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3 Discontinuations due to serious AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 1. Transfusion‐dependent thalassemia: deferasirox vs placebo
Comparison 2. Transfusion‐dependent thalassemia: deferasirox vs deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

8

1170

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.63]

1.1 At 8 months

1

138

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 At 48 weeks (deferasirox 10 mg/kg/day)

1

35

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.3 At 48 weeks (deferasirox 20 mg/kg/day )

1

36

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.4 At 1 year

5

942

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.63]

1.5 At 2 years

1

19

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 LVEF (%): least squares mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

3 LVEF (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3.1 Improvement from abnormal LVEF to normal range

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

3.2 Decrease from normal LVEF to below LLN

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

4 Incidence of thyroid disease at end of study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5 ALT (# participants affected): improvement from abnormal to normal range Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6 ALT (U/L) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

7 AST (U/L) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

8 Serum creatinine (mg/dL) at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

9 Blood urea (mg/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

10 Serum ferritin (ng/mL): mean change from baseline and at end of study Show forest plot

6

1002

Mean Difference (IV, Fixed, 95% CI)

454.42 [337.13, 571.71]

10.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

978.0 [544.71, 1411.29]

10.2 More than 3 to 7 mg Fe/g dw (10/35)

1

150

Mean Difference (IV, Fixed, 95% CI)

801.0 [572.53, 1029.47]

10.3 More than 7 mg Fe/g dw (20/41)

1

169

Mean Difference (IV, Fixed, 95% CI)

328.0 [124.94, 531.06]

10.4 More than 14 mg Fe/g dw (30/51)

1

216

Mean Difference (IV, Fixed, 95% CI)

77.0 [‐303.18, 457.18]

10.5 Any iron overload

5

439

Mean Difference (IV, Fixed, 95% CI)

234.25 [‐8.02, 476.52]

11 Sensitivity analysis: serum ferritin (ng/mL): mean change from baseline Show forest plot

2

701

Mean Difference (IV, Fixed, 95% CI)

418.94 [297.23, 540.65]

11.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30 mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

978.0 [544.71, 1411.29]

11.2 More than 3 to 7 mg Fe/g dw (10/35)

1

150

Mean Difference (IV, Fixed, 95% CI)

801.0 [572.53, 1029.47]

11.3 More than 7 mg Fe/g dw (20/41)

1

169

Mean Difference (IV, Fixed, 95% CI)

328.0 [124.94, 531.06]

11.4 More than 14 mg Fe/g dw (30/51)

1

216

Mean Difference (IV, Fixed, 95% CI)

77.0 [‐303.18, 457.18]

11.5 Any iron overload

1

138

Mean Difference (IV, Fixed, 95% CI)

‐64.16 [‐354.62, 226.30]

12 Liver R2* (Hz): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

13 LIC (mg/g) evaluated by MRI (R2/R2*): mean change from baseline Show forest plot

2

217

Mean Difference (IV, Fixed, 95% CI)

0.36 [‐1.01, 1.72]

14 LIC (mg Fe/g dw) evaluated by biopsy or SQUID: mean change from baseline Show forest plot

1

541

Mean Difference (IV, Fixed, 95% CI)

2.37 [1.68, 3.07]

14.1 LIC 3 mg Fe/g dw or less (5/30)

1

28

Mean Difference (IV, Fixed, 95% CI)

4.3 [2.30, 6.30]

14.2 LIC more than 3 mg to 7 mg (10/35) Fe/g dw

1

143

Mean Difference (IV, Fixed, 95% CI)

3.80 [2.74, 4.86]

14.3 LIC more than 7 mg to 14 mg Fe/g dw (20/41)

1

164

Mean Difference (IV, Fixed, 95% CI)

1.5 [0.28, 2.72]

14.4 LIC more than 14 mg Fe/g dw (30/51)

1

206

Mean Difference (IV, Fixed, 95% CI)

‐2.5 [‐4.55, ‐0.45]

15 Responder analysis I (responder: fall in LIC > 10%) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15.1 Response at 48 weeks (deferasirox 10 mg/kg/day)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Response at 48 weeks (deferasirox 20 mg/kg/day)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Responder analysis II (responder: LIC 1 to < 7 mg Fe/g dw) Show forest plot

1

553

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.69, 0.92]

16.1 Response at 1 year (LIC below 7 mg Fe/g dw)

1

172

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.37, 0.64]

16.2 Response at 1 year (LIC at least 7 mg Fe/g dw)

1

381

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.84, 1.18]

17 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

18 Myocardial iron concentration derived from T2* value (mg Fe/g dw): change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

18.1 All participants

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.2 Participants with T2* <10 ms

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

18.3 Participants with T2* ≥10 ms

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

19 Myocardial T2* (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

19.1 Normalization

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

19.2 Improvement (from 6 ‐ < 10 ms to 10 ‐ ≤ 20 ms)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

19.3 Worsening (from 10‐ ≤ 20 ms to 6 ‐ < 10 ms)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

20 Iron excretion‐intake ratio Show forest plot

1

541

Mean Difference (IV, Fixed, 95% CI)

‐0.18 [‐0.24, ‐0.12]

20.1 Less than 3 mg Fe/g dw (median 5 mg deferasirox / 30 mg deferoxamine)

1

28

Mean Difference (IV, Fixed, 95% CI)

‐0.37 [‐0.54, ‐0.20]

20.2 More than 3 to 7 mg Fe/g dw (10/35)

1

143

Mean Difference (IV, Fixed, 95% CI)

‐0.31 [‐0.41, ‐0.21]

20.3 More than 7 mg Fe/g dw (20/41)

1

164

Mean Difference (IV, Fixed, 95% CI)

‐0.11 [‐0.21, ‐0.01]

20.4 More than 14 mg Fe/g dw (30/51)

1

206

Mean Difference (IV, Fixed, 95% CI)

0.23 [0.05, 0.41]

21 Any serious AEs (# participants affected) Show forest plot

2

773

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.42, 1.86]

22 Serious AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

22.1 Cardiac disorders ‐ arrhythmia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.2 Endocrine disorders ‐ hypogonadism

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.3 GI disorders abdominal abscess

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.4 GI disorders amoebiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.5 GI disorders ‐ appendicitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.6 GI disorders ‐ colitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.7 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.8 GI disorders ‐ gastric haemorrhage

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.9 GI disorders ‐ gastroenteritis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.10 GI disorders ‐ ileus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.11 GI disorders ‐ upper abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.12 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.13 GI disorders ‐ GI infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.14 General disorders and administration site conditions ‐ pyrexia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.15 General disorders and administration site conditions ‐ local swelling

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.16 Hepatobiliary disorders ‐ liver abscess

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.17 Hepatobiliary disorders ‐ cholelithiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.18 Immune system disorders ‐ face oedema

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.19 Infections and infestations ‐ herpes zoster

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.20 Infections and infestations ‐ tooth infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.21 Infections and infestations ‐ urinary tract infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.22 Injury, poisoning and procedural complications ‐ oesophageal rupture

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.23 Injury, poisoning and procedural complications ‐ haemosiderosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.24 Injury, poisoning and procedural complications ‐ iron overload

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.25 Metabolism and nutrition disorders ‐ hyperglycaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.26 Musculoskeletal and connective tissue disorders ‐ back pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.27 Musculoskeletal and connective tissue disorders ‐ pain in jaw

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.28 Nervous system disorders ‐ grand mal convulsion

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.29 Nervous system disorders ‐ meningitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22.30 Respiratory, thoracic and mediastinal disorders ‐ acute tonsilitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

23 Any AE (# participants affected) Show forest plot

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.83, 1.08]

24 AEs Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

24.1 Blood and lymphatic system disorder ‐ agranulocytosis

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.2 Blood and lymphatic system disorder ‐ leukopenia

1

138

Risk Ratio (M‐H, Fixed, 95% CI)

7.0 [0.37, 133.02]

24.3 Blood and lymphatic system disorder ‐ neutropenia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.4 Blood and lymphatic system disorder ‐ thrombocytopenia

2

209

Risk Ratio (M‐H, Fixed, 95% CI)

4.0 [0.46, 34.88]

24.5 Cardiac disorders ‐ cardiac AE

1

586

Risk Ratio (M‐H, Fixed, 95% CI)

0.73 [0.38, 1.41]

24.6 Ear and labyrinth disorders ‐ hearing loss

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.41, 3.05]

24.7 Eye disorder ‐ lens abnormality

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

0.39 [0.08, 2.00]

24.8 Eye disorder ‐ retinal abnormality

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

24.9 GI disorders ‐ abdominal pain

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [0.93, 3.05]

24.10 GI disorders ‐ abdominal pain upper

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.36, 3.60]

24.11 GI disorders ‐ diarrhoea

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.65 [0.86, 3.16]

24.12 GI disorders ‐ dyspepsia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.25, 5.72]

24.13 GI disorders ‐ GIT upset

1

60

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.66, 13.69]

24.14 GI disorders ‐ nausea

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

2.59 [0.90, 7.47]

24.15 GI disorders ‐ vomiting

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.91, 9.55]

24.16 General disorders and administration site conditions ‐ asthenia

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.42, 3.42]

24.17 General disorders and administration site conditions ‐ influenza‐like illness

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.42, 3.42]

24.18 General disorders and administration site conditions ‐ pyrexia

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.27 [0.66, 2.44]

24.19 Immune system disorders ‐ allergic conjunctivitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

4.41 [0.25, 78.58]

24.20 Infections and infestations ‐ bronchitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

2.40 [0.30, 19.35]

24.21 Infections and infestations ‐ upper respiratory tract infection

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.37, 2.42]

24.22 Infections and infestations ‐ urinary tract infection

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

2.40 [0.30, 19.35]

24.23 Investigations ‐ ALT increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.71 [0.59, 4.90]

24.24 Investigations ‐ elevated ALT (>2 UNL)

1

586

Risk Ratio (M‐H, Fixed, 95% CI)

4.90 [0.24, 101.60]

24.25 Investigations ‐ AST increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.21 [0.59, 8.29]

24.26 Investigations ‐ blood creatinine increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

3.79 [0.83, 17.38]

24.27 Investigations ‐ isolated serum creatinine increase above upper limit of normal

2

657

Risk Ratio (M‐H, Fixed, 95% CI)

2.57 [1.88, 3.51]

24.28 Investigations ‐ platelet count increased

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.08, 1.91]

24.29 Musculoskeletal and connective tissue disorders ‐ arthralgia

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.31 [0.55, 3.13]

24.30 Musculoskeletal and connective tissue disorders ‐ back pain

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.30 [0.73, 2.34]

24.31 Musculoskeletal and connective tissue disorders ‐ osteoporosis

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [0.47, 11.91]

24.32 Nervous system disorders ‐ headache

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.44 [0.68, 3.05]

24.33 Nervous system disorders ‐ vertigo

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.32, 3.93]

24.34 Renal and urinary disorders ‐ proteinuria

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.16 [0.50, 2.66]

24.35 Respiratory, thoracic and mediastinal disorders ‐ cough

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [0.67, 4.81]

24.36 Respiratory, thoracic and mediastinal disorders ‐ influenza

2

258

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.52, 2.13]

24.37 Respiratory, thoracic and mediastinal disorders ‐ nasopharyngitis

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

1.90 [0.59, 6.08]

24.38 Respiratory, thoracic and mediastinal disorders ‐ oropharyngeal pain

1

187

Risk Ratio (M‐H, Fixed, 95% CI)

2.84 [0.59, 13.73]

24.39 Respiratory, thoracic and mediastinal disorders ‐ pharyngitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.52, 2.00]

24.40 Respiratory, thoracic and mediastinal disorders ‐ pharyngolaryngeal pain

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.37, 2.08]

24.41 Respiratory, thoracic and mediastinal disorders ‐ rhinitis

1

71

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.58, 2.83]

24.42 Skin and subcutaneous tissue disorders ‐ Rash

1

60

Risk Ratio (M‐H, Fixed, 95% CI)

2.67 [0.78, 9.09]

25 Any drug‐related AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

26 Drug‐related AEs Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

26.1 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.2 Injury, poisoning and procedural complications ‐ infusion site haemorrhage

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.3 Injury, poisoning and procedural complications ‐ infusion site pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.4 Injury, poisoning and procedural complications ‐ infusion site swelling

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.5 Injury, poisoning and procedural complications ‐ injection site pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.6 Injury, poisoning and procedural complications ‐ injection site reaction

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.7 Investigations ‐ blood creatinine increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.8 Investigations ‐ ALT increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.9 Investigations ‐ AST increased

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.10 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.11 GI disorders ‐ abdominal pain upper

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.12 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.13 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.14 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.15 Immune system disorders ‐ hypersensitivity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.16 Immune system disorders ‐ urticaria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.17 Musculoskeletal and connective tissue disorders ‐ arthropathy

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.18 Renal and urinary disorders ‐ proteinuria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.19 Skin and subcutaneous tissue disorders ‐ alopecia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.20 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.21 Injury, poisoning and procedural complications ‐ pulmonary toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.22 Eye disorders ‐ Ophthalmological toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

26.23 Ear and labyrinth disorders ‐ Audiological toxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27 Satisfaction with treatment (very satisfied or satisfied) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

27.1 Week 4 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.2 Week 24 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.3 End of study (1 year) ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.4 Week 4 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.5 Week 24 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

27.6 End of study (1 year) ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28 Convenience (good or very good) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

28.1 Week 4 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.2 Week 24 ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.3 End of study (1 year) ‐ participants previously treated with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.4 Week 4 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.5 Week 24 ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

28.6 End of study (1 year) ‐ DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

29 Willingness to continue treatment Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

29.1 Participants treated previously with DFO

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

29.2 DFO‐naive participants

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

30 Time lost from normal activities due to treatment (hours/month): participants treated previously with DFO Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

30.1 week 4 ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.2 week 24 ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.3 end of study (1 year) ‐ patients treated previously with DFO

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.4 week 4 ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.5 week 24 ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

30.6 end of study (1 year) ‐ DFO‐naive patients

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

31 Adherence (% of planned dose) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

32 Discontinuations Show forest plot

8

1211

Risk Ratio (M‐H, Fixed, 95% CI)

0.95 [0.60, 1.50]

32.1 Deferasirox 10 mg/kg/day

1

35

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.01, 3.64]

32.2 Deferasirox 20 mg/kg/day

2

174

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.10, 9.96]

32.3 Deferasirox 25 mg/kg/day

1

120

Risk Ratio (M‐H, Fixed, 95% CI)

0.09 [0.01, 1.61]

32.4 Deferasirox 40 mg/kg/day

2

211

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.51, 2.05]

32.5 Deferasirox ‐ variable dosage

2

646

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.67, 2.85]

32.6 Deferasirox dosing unknown

1

25

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

33 Dose adjustments and dose interruptions Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

34 Dose interruptions (interrupted at least once) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

35 Dose reduction (at least once) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

36 Dose adjustments (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

37 Dose interruptions due to an AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

38 Haemoglobin (g/dL): mean change from baseline and at end of study Show forest plot

2

180

Mean Difference (IV, Fixed, 95% CI)

‐0.52 [‐0.82, ‐0.21]

38.1 At 8 months (change from baseline)

1

138

Mean Difference (IV, Fixed, 95% CI)

‐0.46 [‐0.81, ‐0.11]

38.2 At 1 year (at end of study)

1

42

Mean Difference (IV, Fixed, 95% CI)

‐0.70 [‐1.33, ‐0.07]

39 Transfusion index (mL/kg/year): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

40 Transferrin saturation (%): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

41 Platelet count (x10³/mm³): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

42 Absolute neutrophilic count (/mm³): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 2. Transfusion‐dependent thalassemia: deferasirox vs deferoxamine
Comparison 3. Transfusion‐dependent thalassemia: deferasirox vs deferiprone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

3

146

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.1 at 1 year

2

128

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 at 2 years

1

18

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Incidence of thyroid disease at end of study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3 ALT (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

4 AST (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

5 Urea (mg/dL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

6 Creatinine (mg/dL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

7 Neutrophil (count per mm³): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

8 Serum ferritin (ng/mL): mean change from baseline and at end of study Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

8.1 All participants

2

83

Mean Difference (IV, Fixed, 95% CI)

229.99 [‐403.14, 863.11]

8.2 Ferritin > 4000

1

11

Mean Difference (IV, Fixed, 95% CI)

1129.0 [‐2226.18, 4484.18]

8.3 Ferritin 2000 ‐ 4000

1

16

Mean Difference (IV, Fixed, 95% CI)

‐151.0 [‐743.80, 441.80]

8.4 Ferritin < 2000

1

11

Mean Difference (IV, Fixed, 95% CI)

388.0 [‐255.71, 1031.71]

9 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

10 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

11 Any AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12.1 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 GI disorders ‐ pain abdomen

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.4 GI disorders ‐ vomiting

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.5 Musculoskeletal and connective tissue disorders ‐ arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.6 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.7 Blood and lymphatic system disorder ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.8 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.9 Investigations ‐ AST levels > 2x UNL

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.10 Investigations ‐ ALT levels > 2x UNL

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.11 Investigations ‐ serum creatinine 50% increase from baseline

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.12 Renal and urinary disorders ‐ renal failure

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Discontinuations (# participants affected) Show forest plot

3

179

Risk Ratio (M‐H, Fixed, 95% CI)

0.16 [0.01, 2.99]

14 Discontinuation due to an AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15 Transferrin saturation (%): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

16 Haemoglobin (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

17 Transfusion index (mL/kg/year): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

18 ALP (U/L): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 3. Transfusion‐dependent thalassemia: deferasirox vs deferiprone
Comparison 6. Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 at 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Neutrophil (µg/L): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

3 ALT (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

4 AST (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

5 Serum ferritin: mean at end of study (ng/mL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

6 Discontinuations Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7 Haemoglobin (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

8 ALP (g/dL): mean at end of study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 6. Transfusion‐dependent thalassemia: deferasirox + deferoxamine vs deferoxamine
Comparison 7. Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 at 12 months

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serum ferritin (ng/mL): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

3 LIC (mg/g) evaluated by MRI (R2*): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

4 Myocardial T2* (ms): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

5 Serious AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6 Serious drug‐related AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

6.1 Cholecystitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

7 Serious non‐related drug AE Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

7.1 Appendicitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

8.1 Blood and lymphatic system disorder ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

8.2 Blood and lymphatic system disorder ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

9 Drug‐related AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10 Drug‐related AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 Blood and lymphatic system disorders ‐ agranulocytosis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Blood and lymphatic system disorders ‐ neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 GI disorders ‐ GI problems

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Investigations ‐ ALT increase ( ≥ 3 folds)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 Investigations ‐ serum creatinine ( ≥ 33%) above baseline in 2 consecutive occasions

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.6 Musculoskeletal and connective tissue disorders ‐ arthralgia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.7 Skin and subcutaneous tissue disorders ‐ skin rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Non‐related drug AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12 Non‐related drug AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12.1 Infections and infestations ‐ infections

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.2 GI disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

12.3 Skin and subcutaneous tissue disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

13 Mild elevation of hepatic transaminases at start of therapy (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

14 Initial gastrointestinal manifestations (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15 Quality of life (%) (measured by SF‐36): mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

16 Adherence: actual dose/total prescribed dose Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

17 Discontinuations Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

18 Serious AE resulting in study discontinuation or interruption Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Figures and Tables -
Comparison 7. Transfusion‐dependent thalassemia: deferasirox + deferiprone vs deferiprone + deferoxamine
Comparison 8. Non‐transfusion‐dependent thalassemia: deferasirox vs placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality at any time point Show forest plot

1

148

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.1 Deferasirox 5 mg/kg/day: at 12 months

1

73

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

1.2 Deferasirox 10 mg/kg/day: at 12 months

1

75

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serum ferritin (ng/mL): mean change from baseline Show forest plot

1

154

Mean Difference (IV, Fixed, 95% CI)

‐306.74 [‐398.23, ‐215.24]

2.1 Deferasirox 5 mg/kg/day

1

78

Mean Difference (IV, Fixed, 95% CI)

‐259.1 [‐377.35, ‐140.85]

2.2 Deferasirox 10 mg/kg/day

1

76

Mean Difference (IV, Fixed, 95% CI)

‐377.79 [‐522.21, ‐233.37]

3 LIC (mg Fe/g dw) evaluated by MRI R2: least squares mean change from baseline Show forest plot

1

159

Mean Difference (IV, Fixed, 95% CI)

‐3.27 [‐4.44, ‐2.09]

3.1 Deferasirox 5 mg/kg/day

1

78

Mean Difference (IV, Fixed, 95% CI)

‐2.33 [‐4.00, ‐0.66]

3.2 Deferasirox 10 mg/kg/day

1

81

Mean Difference (IV, Fixed, 95% CI)

‐4.18 [‐5.83, ‐2.53]

4 LIC (mg Fe/g dw) evaluated by MRI R2: mean change from baseline Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

4.1 Non‐transfusion‐dependent β‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.2 Non‐transfusion‐dependent β‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.3 α‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.4 α‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.5 HbE/β‐thalassemia 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.6 HbE/β‐thalassemia 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.7 < 18 years 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.8 < 18 years 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.9 ≥18 years 5 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

4.10 ≥18 years 10 mg/kg/day

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [0.0, 0.0]

5 LIC: decrease of ≥ 3 mg Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

4.16 [1.90, 9.11]

5.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.05 [0.98, 9.50]

5.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

5.26 [1.76, 15.71]

6 LIC: ≥ 30% reduction Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

14.17 [2.88, 69.74]

6.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

15.02 [0.93, 242.87]

6.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

13.75 [1.97, 95.97]

7 LIC: shift to lower iron burden range (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

3.35 [1.62, 6.91]

7.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.39 [1.10, 10.45]

7.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

3.31 [1.28, 8.55]

8 LIC: achieve LIC < 5 mg Fe/g dw (# participants affected) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

5.35 [1.30, 21.99]

8.1 deferasirox 5mg/kg/d

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

4.07 [0.54, 30.96]

8.2 deferasirox 10mg/kg/d

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

6.62 [0.91, 48.05]

9 Drug‐related serious AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10 Drug‐related serious AEs (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

10.1 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.2 Infections and infestations ‐ cellulitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.3 Skin and subcutaneous tissue disorders ‐ pruritus

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.4 Skin and subcutaneous tissue disorders ‐ rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.5 General disorders ‐ pyrexia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

10.6 Hepatobiliary disorders ‐ hepatotoxicity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

11 Any AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

12 Mild AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

13 Moderate AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

14 Severe AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15 AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

15.1 Ear and labyrinth disorders ‐ neurosensory deafness

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.2 Investigations ‐ abnormal platelet count (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.3 Investigations ‐ abnormal neutrophils count (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.4 Investigations ‐ abnormal ALT (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.5 Investigations ‐ abnormal AST (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.6 Investigations ‐ abnormal serum creatinine (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.7 Investigations ‐ abnormal creatinine clearance (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.8 Investigations ‐ urinary protein/creatinine ratio (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.9 Investigations ‐ abnormal (low) systolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.10 Investigations ‐ abnormal (high) systolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.11 Investigations ‐ abnormal (low) diastolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.12 Investigations ‐ abnormal (high) diastolic blood pressure (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.13 Investigations ‐ abnormal (low) pulse rate (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.14 Investigations ‐ abnormal (high) pulse rate (post‐baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

15.15 Renal and urinary disorders ‐ proteinuria

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16 Drug‐related AEs Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

16.1 GI disorders ‐ abdominal pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.2 GI disorders ‐ abdominal pain upper

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.3 GI disorders ‐ diarrhoea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.4 GI disorders ‐ nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.5 Nervous system disorders ‐ headache

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

16.6 Skin and subcutaneous tissue disorders ‐ skin rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

17 Adherence (# participants taking the planned study dose) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

18 Discontinuations Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.32 [0.50, 3.52]

18.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.33, 4.25]

18.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.33, 7.08]

19 Discontinuing study due to AE (# participants affected) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

20 Dose increase Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

0.87 [0.63, 1.19]

20.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

0.88 [0.57, 1.38]

20.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.54, 1.33]

21 Dose interruption (at least once) Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.88, 1.39]

21.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.73, 1.43]

21.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.19 [0.87, 1.62]

22 Dose reduction Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [0.70, 3.06]

22.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.54, 4.30]

22.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.4 [0.49, 4.00]

23 Dose reduction due to AE Show forest plot

1

166

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.69, 3.38]

23.1 Deferasirox 5 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

2.04 [0.63, 6.63]

23.2 Deferasirox 10 mg/kg/day

1

83

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.39, 3.39]

24 Haemoglobin (g/L): mean change from baseline Show forest plot

1

144

Mean Difference (IV, Fixed, 95% CI)

1.54 [‐0.82, 3.90]

24.1 Deferasirox 5 mg/kg/day

1

71

Mean Difference (IV, Fixed, 95% CI)

1.00 [‐2.31, 4.31]

24.2 Deferasirox 10 mg/kg/day

1

73

Mean Difference (IV, Fixed, 95% CI)

2.10 [‐1.27, 5.47]

25 Transferrin saturation (%): mean change from baseline Show forest plot

1

141

Mean Difference (IV, Fixed, 95% CI)

‐7.10 [‐11.71, ‐2.50]

25.1 Deferasirox 5 mg/kg/day

1

70

Mean Difference (IV, Fixed, 95% CI)

‐7.16 [‐12.95, ‐1.37]

25.2 Deferasirox 10 mg/kg/day

1

71

Mean Difference (IV, Fixed, 95% CI)

‐7.01 [‐14.59, 0.57]

Figures and Tables -
Comparison 8. Non‐transfusion‐dependent thalassemia: deferasirox vs placebo