Scolaris Content Display Scolaris Content Display

Intervensi untuk meningkatkan kepatuhan terapi kelasi besi dalam kalangan orang dengan penyakit sel sabit atau talasemia

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Aydinok 2007 {published data only}

Aydinok Y, El‐Beshlawy A, von Orelli‐Leber C, Czarnecki‐Tarabishi C, Manz C Y. A randomized controlled trial comparing the combination therapy of deferiprone (DFP) and desferrioxamine (DFO) versus DFP or DFO monotherapy in patients with thalassemia major. Blood 2006;108(11). [Abstract no.: 557]CENTRAL
Aydinok Y, Ulger Z, Nart D, Terzi A, Cetiner N, Ellis G, et al. A randomized controlled 1‐year study of daily deferiprone plus twice weekly desferrioxamine compared with daily deferiprone monotherapy in patients with thalassemia major. Haematologica 2007;92(12):1599‐606. CENTRAL
Manz CH, El‐Beshlawy A, Aydinok Y, Leber C, Czarnecki‐Tarabishi C. A randomized controlled prospective clinical study comparing the combination therapy of deferiprone (L1) and desferrioxamine with L1 and DFO monotherapy in patients with thalassemia major. Haematologica 2006;91(S1):190. [Abstract no.: 0515]CENTRAL

Badawy 2010 {published data only}

Badawy S, Hassan TH, Hesham MA, Badr MA. Evaluation of iron chelation therapy in B‐thalassemic patients in Zagazig University Hospital. ASPHO abstracts (The American Society of Pediatric Hematology/Oncology)2010; Vol. 54, issue 6:799‐800. CENTRAL

Bahnasawy 2017 {published data only}

Bahnasawy SM, El Wakeel LM, El Beblawy N, El‐Hamamsy M. Clinical pharmacist‐provided services in iron overloaded Beta‐thalassemia major children; A new insight to patient care. Basic & Clinical Pharmacology & Toxicology 2017;120(4):354‐9. CENTRAL

Calvaruso 2015 {published data only}

Calvaruso G, Vitrano A, Di Maggio R, Ballas S, Steinberg MH, Rigano P, et al. Deferiprone versus deferoxamine in thalassemia intermedia: results from a 5‐year long‐term Italian multicenter randomized clinical trial. American Journal of Hematology 2015;90(7):634‐8. CENTRAL
Vitrano A, Calvaruso G, Di Maggio G, Romeo MA, Cianciulli P, Lai ME, et al. Deferiprone versus deferoxamine in thalassemia intermedia: results from 5‐year long‐term Italian multi‐center randomized clinical trial. 56th ASH Annual Meeting and Exposition; 2014 Dec 6‐9; San Francisco, California2014. CENTRAL

Elalfy 2015 {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(S1):443‐4. 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, Adly A. Comparison of two combination iron chelation regimens, deferiprone and deferasirox versus deferiprone and deferoxamine, in pediatric patients with beta‐thalassemia major. Blood 2013;122(21). [Abstract no.: 559]CENTRAL

El Beshlawy 2008 {published data only}

Study With Deferiprone and/or Desferrioxamine in Iron Overloaded Patients. ClinicalTrials.gov2006; Vol. NCT00350662. CENTRAL
El‐Beshlawy A, Manz C, Naja M, Eltagui M, Tarabishi C, Youssry I, et al. Iron chelation in thalassemia: combined or monotherapy? The Egyptian experience. Annals of Hematology 2008;87(7):545‐50. CENTRAL

Galanello 2006 {published data only}

Galanello R, Kattamis A, Piga A, Tricta F. Safety and efficacy of alternate desferrioxamine and deferiprone compared to desferrioxamine alone in the treatment of iron overload in transfusion‐dependent thalassemia patients. Blood 2004;104(11 Pt 1). [Abstract no.: 3611]CENTRAL
Galanello R, Piga A, Forni G L, Bertrand Y, Foschini M L, Bordone E, et al. Phase II clinical evaluation of deferasirox, a once‐daily oral chelating agent, in pediatric patients with beta‐thalassemia major. Haematologica 2006;91(10):1343‐51. 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):2425‐2431. CENTRAL

Maggio 2009 {published data only}

Maggio A, Capra M, Cuccia L, Gagliardotto F, Magnano C, Caruso V, et al. Deferiprone versus sequential deferiprone‐deferoxamine treatment in thalassemia major: a five years multicenter randomized clinical trial under the auspices of the society for the study of thalassemia and hemogobinopathies (SoST). Blood 2007;110(11). [Abstract no.: 575]CENTRAL
Maggio A, Vitrano A, Capra M, Cuccia L, Gagliardotto F, Filosa A, et al. Decrease of mortality during deferiprone treatments: results from a large randomised cohort of thalassemia major patients under the auspices of the Italian society for thalassemia and hemogobinopathies. Blood 2008;112 Suppl:Abstract no: 3885. CENTRAL
Maggio A, Vitrano A, Capra M, Cuccia L, Gagliardotto F, Filosa A, et al. Long‐term sequential deferiprone‐deferoxamine versus deferiprone alone for thalassaemia major patients: A randomized clinical trial. British Journal of Haematology 2009;145(2):245‐54. CENTRAL
NCT00733811. Efficacy Study of the Use of Sequential DFP‐DFO Versus DFP. https://clinicaltrials.gov/ct2/show/NCT00733811 (accessed 11 April 2018). CENTRAL
Pantalone GR, Maggio A, Vitrano A, Capra M, Cuccia L, Gagliardotto F, et al. Sequential alternating deferiprone and deferoxamine treatment compared to deferiprone monotherapy: Main findings and clinical follow‐up of a large multicenter randomized clinical trial in ‐thalassemia major patients. Hemoglobin 2011;35(3):206‐16. CENTRAL

Mourad 2003 {published data only}

Mourad FH, Hoffbrand AV, Sheikh‐Taha M, Koussa S, Khoriaty AI, Taher A. Comparison between desferrioxamine and combined therapy with desferrioxamine and deferiprone in iron overloaded thalassaemia patients. British Journal of Haematology 2003;121(1):187‐9. CENTRAL

Olivieri 1997 {published data only}

Olivieri N, the Iron Chelation Research Group. Randomized trial of deferiprone (LI) and deferoxamine (DFO) in thalassemia major. Blood1996; Vol. 88, issue 10 Suppl 1:651a. CENTRAL
Olivieri NF, Brittenham GM. Evidence of progression of myocardial iron loading as determined by magnetic resonance imaging (MRI) in thalassemia patients during treatment with deferiprone (L1) and deferoxamine (DFO). Blood1999; Vol. 94, issue 10 Suppl 1:35b. CENTRAL
Olivieri NF, Brittenham GM. Final results of the randomized trial of deferiprone (L1) and deferoxamine (DFO). Blood1997; Vol. 90, issue 10 Suppl 1:264a. CENTRAL
Olivieri NF, Brittenham GM, Armstrong SAM, Basran RK, Daneman R, Daneman N, et al. First prospective randomized trial of the iron chelators deferiprone (L1) and deferoxamine. Blood1995; Vol. 86, issue 10 Suppl 1:249a. CENTRAL
Pope, Elena. Critical review of standard and new methods of assessing compliance with chelation therapy in thalassemic patients. Proquest Dissertations Publishing1995. CENTRAL

Pennell 2006 {published data only}

Pennell D J, Berdoukas V, Karagiorga M, Ladis V, Piga A, Aessopos A, et al. Randomized controlled trial of deferiprone or deferoxamine in beta‐thalassemia major patients with asymptomatic myocardial siderosis. Blood 2006;107(9):3738‐44. CENTRAL
Smith GC, Alpendurada F, Carpenter JP, Alam MH, Berdoukas V, Kargiorga M, et al. Effect of deferiprone or deferoxamine on right ventricular function in thalassemia major patients with myocardial iron overload. Journal of Cardiovascular Magnetic Resonance2011; Vol. 13, issue 1:34. CENTRAL

Pennell 2014 {published data only}

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 2015;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. [Abstract no.: 0928]CENTRAL
Pennell D, 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 beta‐thalassemia major and iron overload (CORDELIA). Blood 2012;120(21). [Abstract no.: 2124]CENTRAL
Pennell DJ, Porter JB, Piga A, Lai Y, El‐Beshlawy A, Belhoul K M, et al. A 1‐year randomized controlled trial of deferasirox vs deferoxamine for myocardial iron removal in beta‐thalassemia major (CORDELIA). Blood 2014;123(10):1447‐54. CENTRAL

Taher 2017 {published data only}

A randomized, open‐label, multicenter, two arm, phase II study to investigate the benefits of an improved deferasirox formulation (Film‐coated Tablet). ClinicalTrials.go2014; Vol. NCT02125877. CENTRAL
A randomized, open‐label, multicenter, two arm, phase II study to investigate the benefits of an improved deferasirox formulation (film‐coated tablet). EU Clinical Trials Register issue https://www.clinicaltrialsregister.eu/ctr‐search/trial/2013‐004167‐32/AT. CENTRAL
Huang VW, Banderas B, Sen R. Psychometric evaluation of clinical outcomes assessments in a phase II trial. Value in Health2016; Vol. 19, issue 7:A746. CENTRAL
Taher AT, Origa R, Perrotta S, Kourakli A, Ruffo GB, Kattamis A, et al. New film‐coated tablet formulaion of deferasirox is well tolerated in patients with thalassemia or lower‐risk MDS: Results of the randomized, Phase II ECLIPSE study. American Journal of Hematology 2017;92(5):420‐8. CENTRAL

Tanner 2007 {published data only}

Tanner MA, Galanello R, Dessi C, Agus A, Smith GC, Westwood MA, et al. Improved endothelial function combined chelation therapy in thalassaemia major. Blood 2006;108(11). [Abstract no.: 1770]CENTRAL
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. CENTRAL
Tanner MA, Galanello R, Dessi C, Smith GC, Westwood MA, Agus A, et al. The effect of combined therapy with deferoxamine and deferiprone on myocardial iron and endothelial function in thalassaemia major: a randomized controlled trials using cardiovascular magnetic resonance. Haematologica2006; Vol. 91, issue Suppl 1:191. CENTRAL
Tanner MA, Galanello R, Dessi C, Westwood MA, Smith GC, Khan M, et al. A randomized, placebo controlled, double blind trial of the effect of combined therapy with deferoxamine and deferiprone on myocardial iron in thalassaemia major using cardiovascular magnetic resonance. Blood 2005;106(11 Pt 1). [Abstract no.: 3655]CENTRAL

Vichinsky 2007 {published data only}

Vichinsky E. Patient reported outcomes with chelation therapy in patients with sickle cell disease (SCD) on either deferasirox (Exjade®, ICL670) or deferoxamine (DFO). 29th annual meeting of the National Sickle Cell Disease Program; April 8‐12; Memphis, USA2006. [Abstract no.: 174]CENTRAL
Vichinsky E. Results of a randomized, controlled phase two trials of deferasirox (Exjade®, ICL670) in sickle cell disease patients with chronic overload. 29th Annual Meeting of the National Sickle Cell Disease Program; April 8‐12; Memphis, USA2006. [Abstract no.: 175]CENTRAL
Vichinsky E, Bernaudin F, Forni GL, Gardner R, Hassell K, Heeney MM, et al. Long‐term safety and efficacy of deferasirox (Exjade) for up to 5 years in transfusional iron‐overloaded patients with sickle cell disease. British Journal of Haematology 2011;154(3):387‐97. CENTRAL
Vichinsky E, Bernaudin F, Forni GL, Gardner R, Hassell KL, Heeney MM, et al. Long‐term safety and efficacy of deferasirox (Exjade®) in transfused patients with sickle cell disease treated for up to 5 years. Blood 2011;118(21). [Abstract no.: 845]CENTRAL
Vichinsky E, Coates T, Thompson A, Bernaudin F, Lagrone D, Dong V, et al. Safety and efficacy of iron chelation therapy with deferasirox in patients with sickle cell disease (SCD): 3.5‐year follow‐up. 14th Congress of the European Haematology Association; Jun 4‐7; Berlin, Germany2009. CENTRAL
Vichinsky E, Coates T, Thompson AA, Bernaudin F, Rodriguez M, Rojkjaer L, et al. Deferasirox (Exjade®), the once‐daily oral iron chelator, demonstrates safety and efficacy in patients with sickle cell disease (SCD): 3.5‐year follow‐up. Blood 2008;112(11). [Abstract no.: 1420]CENTRAL
Vichinsky E, Coates T, Thompson AA, Bernaudin F, Rodriguez M, Rojkjaer L, et al. Deferasorix (Exjade®), the once‐daily oral iron chelator, demonstrates safety and efficacy in patients with sickle cell disease (SCD): 3.5‐year follow‐up. 3rd Annual Sickle Cell Disease Research and Educational Symposium and Annual Sickle Cell Disease Scientific Meeting; Feb 18‐20; Florida, USA2009. [Abstract no.: 225]CENTRAL
Vichinsky E, Coates T, Thompson AA, Mueller BU, Lagrone D, Heeney MM. Long‐term efficacy and safety of deferasirox (Exjade®, ICL670), a once‐daily oral iron chelator, in patients with sickle cell disease (SCD). Blood 2007;110(11 Pt 1):995A. [Abstract no.: 3395]CENTRAL
Vichinsky E, Fischer R, Fung E, Onyekwere O, Porter J, Swerdlow P, et al. A randomized, controlled phase two trial in sickle cell disease patients with chronic iron overload demonstrates that the once‐daily oral iron chelator deferasirox (Exjade®, ICL670) is well tolerated and reduces iron burden. Blood 2005;106. [Abstract no.: 313]CENTRAL
Vichinsky E, Fischer R, Pakbaz Z, Onyekwere O, Porter J, Swerdlow P, et al. Satisfaction and convenience of chelation therapy in patients with sickle cell disease (SCD): Comparison between deferasirox (Exjade®, ICL670) and deferoxamine (DFO). Blood 2005;106(11 Pt 1). [Abstract no.: 2334]CENTRAL
Vichinsky E, Onyekwere O, Porter J, Swerdlow P, Eckman J, Lane P, et al. A randomised comparison of deferasirox versus deferoxamine for the treatment of transfusional iron overload in sickle cell disease. British Journal of Haematology 2007;136(3):501‐8. CENTRAL
Vichinsky E, Pakbaz Z, Onyekwere O, Porter J, Swerdlow P, Coates T, et al. Patient‐reported outcomes of deferasirox (Exjade, ICL670) versus deferoxamine in sickle cell disease patients with transfusional hemosiderosis. Substudy of a randomized open‐label phase II trial. Acta Haematologica 2008;119(3):133‐141. CENTRAL

Abu 2015 {published data only}

Abu SO, Auda W, Kamhawy H, Al‐Tonbary Y. Impact of educational programme regarding chelation therapy on the quality of life for B‐thalassemia major children. Hematology 2015;20(5):297‐303. CENTRAL

Al Kloub 2014 {published data only}

Al‐Kloub MI, A Bed MA, Al Khawaldeh OA, Al Tawarah YM, Froelicher ES. Predictors of non‐adherence to follow‐up visits and deferasirox chelation therapy among Jordanian adolescents with Thalassemia major. Pediatric Hematology and Oncology 2014;31(7):624‐37. CENTRAL

Al Kloub 2014a {published data only}

Al‐Kloub, MI, TN, Salameh, ES, Froelicher. Impact of psychosocial status and disease knowledge on deferoxamine adherence among thalassaemia major adolescents. International Journal of Nursing Practice 2014;20(3):265‐74. CENTRAL

Al Refaie 1995 {published data only}

Al‐Refaie FN, Hershko C, Hoffbrand AV, Kosaryan M, Olivier NF, Tondury P, et al. Results of long‐term deferiprone (L1) therapy: a report by the International Study Group on Oral Iron Chelators. British Journal of Haematology 1995;91(1):224‐9. CENTRAL

Alvarez 2009 {published data only}

Alvarez O, Rodriguez‐Cortes H, Robinson N, Lewis N, Pow Sang CD, Lopez‐Mitnik G, et al. Adherence to deferasirox in children and adolescents with sickle cell disease during 1‐year of therapy. Journal of Pediatric Hematology/Oncology 2009;31(10):739‐44. CENTRAL

Armstrong 2011 {published data only}

Armstrong EP, Skrepnek GH, Ballas SK, Kwok P, Snodgrass S, Sasane M. Costs, persistence, and hospitalizations associated with the use of iron‐chelating therapies in sickle cell disease in medicaid patients. Blood2011; Vol. 118, issue 21. CENTRAL

Bala 2014 {published data only}

Bala J, Sarin J. Treatment adherence and quality of life of thalassemic children. International Journal of Nursing Education 2014;6(2):151‐2. CENTRAL

Belgrave 1989 {published data only}

Belgrave FZ, Gilbert SK. Health care adherence of persons with sickle cell disease. The role of social support. Annals of the New York Academy of Sciences 1989;565:369‐70. CENTRAL

Berkovitch 1995 {published data only}

Berkovitch M, Davis S, Matsui D, DonskyJ, Koren G, Olivieri NF. Use of a eutectic mixture of local anesthetics for prolonged subcutaneous drug administration. Journal of Clinical Pharmacology 1995;35(3):295‐7. CENTRAL

Chakrabarti 2013 {published data only}

Chakrabarti P, Bohara V, Ray S, Sankar Ray S, Kumar, NU, Chaudhuri U. Can the availability of unrestricted financial support improve the quality of care of thalassemics in a center with limited resources? A single center study from India. Thalassemia Reports 2013;3(1):6‐10. CENTRAL

Daar 2010 {published data only}

Daar S, Al, Salmi F, Ableen V, Jacob W, Jabeen Z, Pathare A. T2*MRI ‐ An effective tool to increase chelation compliance in thalassemia major. Haematologica2010, issue 95:698. CENTRAL

Gomber 2004 {published data only}

Gomber S, Saxena R, Madan N. Comparative efficacy of desferrioxamine, deferiprone and in combination on iron chelation in thalassemic children. Indian Pediatrics 2004;41(1):21‐7. CENTRAL

Kidson Gerber 2008 {published data only}

Kidson‐Gerber G, Lindeman R. Adherence to desferrioxamine and deferiprone and the impact of deferiprone co‐prescription in thalassaemia major patients. Does the addition of deferiprone improve adherence?. British Journal of Haematology 2008;142(4):679‐80. CENTRAL

Kolnagou 2008 {published data only}

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. CENTRAL

Leonard 2014 {published data only}

Leonard S, Jonassaint J, Anderson L, Shah N. The use of mobile technology for intensive training in medication management in the pediatric population. Blood2014; Vol. 124, issue 21. CENTRAL

Loiselle 2016 {published data only}

Loiselle K, Lee JL, Szulczewski L, Drake S, Crosby LE, Pai AL. Systematic and meta‐analytic review: medication adherence among pediatric patients With sickle cell disease. Journal of Pediatric Psychology 2016;41(4):406‐18. CENTRAL

Mazzone 2009 {published data only}

Mazzone L, Battaglia L, Andreozzi F, Romeo MA, Mazzone D. Emotional impact in beta‐thalassaemia major children following cognitive‐behavioural family therapy and quality of life of caregiving mothers. Clinical Practice and Epidemiology in Mental Health 2009;5(5):Online. CENTRAL

NCT01709032 {published data only}

NCT01709032. Combination deferasirox and deferiprone for severe iron overload in thalassemia. https://clinicaltrials.gov/ct2/show/NCT01709032 (accessed 11 April 2018). CENTRAL

NCT01825512 {published data only}

NCT01825512. Multicentre, randomised, open label, non‐inferiority active‐controlled trial to evaluate the efficacy and safety of deferiprone compared to deferasirox in paediatric patients aged from 1 month to less than 18 years of age affected by transfusion‐dependent haemoglobinopathies. https://clinicaltrials.gov/ct2/show/NCT01825512 (accessed 11 April 2018). CENTRAL

NCT02133560 {published data only}

NCT02133560. Use of mobile technology for intensive training in medication management. https://clinicaltrials.gov/ct2/show/NCT02133560 (accessed 14 April 2018). CENTRAL

NCT02466555 {published data only}

NCT02466555. Music Therapy in Sickle Cell Transition Study. https://clinicaltrials.gov/ct2/show/NCT02466555 (accessed 14 April 2018). [Sponsors: University Hospital Case Medical Center|Kulas FoundationOther IDs: 03‐15‐30]CENTRAL

Pakbaz 2004 {published data only}

Pakbaz Z, Fischer R, Gamino R, Quirolo K, Yamashita R, Treadwell M, et al. Assessing compliance to iron chelation therapy in patients with thalassemia. Blood2004; Vol. 104, issue 11:33B. CENTRAL

Pakbaz 2005 {published data only}

Pakbaz ZR, Fischer M, Treadwell R, Yamashita EB, Fung L, Calvell K, et al. A simple model to assess and improve adherence to iron chelation therapy with deferoxamine in patients with thalassemia. Annals of the New York Academy of Sciences 2005;1054:486‐91. CENTRAL

Porter 2009 {published data only}

Porter JB, Athanasiou‐Metaxa M, Bowden DK, Troncy, J, Habr D, Domokos G, et al. Improved patient satisfaction, adherence and health‐related quality of life with deferasirox (Exjade) in beta‐thalassemia patients previously receiving other iron chelation therapies. Blood2009; Vol. 114, issue 22. CENTRAL

Porter 2012 {published data only}

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;297:641. CENTRAL

Vichinsky 2005 {published data only}

Vichinsky E, Fischer R, Pakbaz Z, Onyekwere O, Porte, J, Swerdlow P, et al. Satisfaction and convenience of chelation therapy in patients with sickle cell disease (SCD): Comparison between deferasirox (Exjade®, ICL670) and deferoxamine (DFO). Blood 2005;106(11 Pt 1). [Abstract no.: 2334]CENTRAL

Vichinsky 2008 {published data only}

Vichinsky E, Pakbaz Z, Onyekwere O, Porter J, Swerdlow P, Coates T, et al. Patient‐reported outcomes of deferasirox (Exjade, ICL670) versus deferoxamine in sickle cell disease patients with transfusional hemosiderosis. Substudy of a randomized open‐label phase II trial. Acta Haematologica 2008;119(3):133‐41. CENTRAL

Waheed 2014 {published data only}

Waheed N, Ali S, Butt MA. Comparison of deferiprone and deferrioxamine for the treatment of transfusional iron overload in children with beta thalassemia major. Journal of Ayub Medical College, Abbottabad: JAMC 2014;26(3):297‐300. CENTRAL

Walsh 2014 {published data only}

Walsh KE, Cutrona SL, Kavanagh PL, Crosby LE, Malone C, Lobner K, et al. Medication adherence among pediatric patients with sickle cell disease: a systematic review. Pediatrics 2014;134(6):1175‐83. CENTRAL

Yarali 2006 {published data only}

Yarali N, Fisgin T, Duru F, Kara A, Ecin N, Fitoz S, Erden I. Subcutaneous bolus injection of deferoxamine is an alternative method to subcutaneous continuous infusion. Journal of Pediatric Hematology/Oncology 2006;28(1):11‐6. CENTRAL

Antmen 2013 {published data only}

Antmen B, Organ K, Sasmaz I, Berktas M, Kilinc Y. A cohort study to assess the contribution of patient compliance program on persistence to deferasirox in patients with chronic iron overload in turkey (ex‐pat program). Haematologica/the Hematology Journal2013; Vol. 98:713‐4. CENTRAL

NCT00004982 {published data only}

NCT00004982. Combination iron chelation therapy. https://clinicaltrials.gov/ct2/show/NCT00004982 (accessed 18 April 2018). CENTRAL

EudraCT 2012‐000353‐31 {published data only}

Consorzio per le Valutazioni Biologiche e Farmacologiche. Multicentre, randomised, open label, non‐inferiority active‐controlled trial to evaluate the efficacy and safety of deferiprone compared to deferasirox in paediatric patients aged from 1 month to less than 18 years of age affected by transfusion‐dependent haemoglobinopathies. https://www.clinicaltrialsregister.eu/ctr‐search/trial/2012‐000353‐31/IT (EU Clinical Trials Register) (accessed 18 April 2018). CENTRAL

IRCT2015101218603N2 {published data only}

Amirmoezi F. To assess compliance, efficacy and satisfaction with two different formulation of deferasirox in patients with transfusion‐dependent beta‐thalassemia. http://en.irct.ir/trial/16826 (Iranian Registry of Clinical Trails) (accessed 18 April 2018). CENTRAL

Madderom 2016 {published data only}

Cnossen MH. A randomized trial evaluaTing the Effects of group medical AppointMents on self‐efficacy and adherence in Sickle Cell Disease (TEAM study). ‐ TEAM study [Effecten van GroepsconsulTen op zElfmAnageMent en therapietrouw in sikkelcelziekte (TEAM studie)]. http://www.trialregister.nl/trialreg/admin/rctview.asp?TC=4750 (Erasmus Medical Center, Sophia Children's Hospital) (accessed 18 April 2018), issue NTR4750. CENTRAL
Madderom MJ, Heijdra J, Utens EM, Polinder S, Rijneveld AW, Cnossen MH. A randomized controlled trial studying the effectiveness of group medical appointments on self‐efficacy and adherence in sickle cell disease (TEAM study): study protocol. BMC Hematology 2016;16(21):6. CENTRAL

NCT02173951 {published data only}

NCT02173951. An algorithm to start iron chelation in minimally transfused young beta‐thalassemia major patients. https://clinicaltrials.gov/ct2/show/NCT02173951 (accessed 18 April 2018):ClinicalTrials.gov. CENTRAL

NCT02435212 {published data only}

NCT02435212. Study to evaluate treatment compliance, efficacy and safety of an improved deferasirox formulation (granules) in pediatric patients (2‐<18 years old) with iron overload. https://clinicaltrials.gov/ct2/show/NCT02435212 (accessed 18 April 2018). [Sponsors: Novartis Pharmaceuticals|NovartisOther IDs: CICL670F2202|2013‐004739‐55]CENTRAL

Abetz 2006

Abetz l, Baladi JF, Jones P, Rafail D. The impact of iron overload and its treatment on quality of life: results from a literature review. Health and Quality of Life Outcomes 2006;4:73.

American Pharmacists Association 2008

American Pharmacists Association, National Association of Chain Drug Stores Foundation. Medication therapy management in community pharmacy practice: core elements of an MTM service (version 2.0). American Pharmacists Association2008:24.

APPG 2009

All‐Party Parliamentary Group on Sickle Cell and Thalassaemia (APPG). Sickle cell disease and thalassaemia: A health check. http://ukts.org/pdfs/awareness/appg.pdf (accessed 20 June 2016).

Aydinok 2014

Aydinok Y, Kattamis A, Viprakasit V. Current approach to iron chelation in children. British Journal of Haematology 2014;165(6):745‐55.

Costello 2004

Costello I, Wong ICK, Nunn AJ. A literature review to identify interventions to improve the use of medicines in children. Child: Care, Health and Development 2004;30(6):647‐65.

Covidence [Computer program]

Veritas Health Innovation. Covidence systematic review software. Melbourne, Australia: Veritas Health Innovation, 2017.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG on behalf of the Cochrane Statistical Methods Group, editor(s). Chapter 9: Analysing data and undertaking meta‐analysis. 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. Available from www.cochrane‐handbook.org.

EPOC 2015

Effective Practice, Organisation of Care (EPOC). EPOC Resources for review authors. http://epoc.cochrane.org/epoc‐specific‐resources‐review‐authors (accessed 21 September 2017).

Fisher 2013

Fisher SA, Brunskill SD, Doree C, Gooding S, Chowdbury 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.pub3]

Gravitz 2014

Gravitz L, Pincock S. Sickle‐cell disease. Nature 2014;515:S1.

Grosse 2011

Grosse SD, Odame I, Atrash HK, Amendah DD, Piel FB, Williams TN. Sickle cell disease in Africa. A neglected cause of early childhood mortality. American Journal of Preventive Medicine 2011;41(6 S4):S298‐S405.

Haywood 2009

Haywood, C, Beach M, Lanzkron S, Strouse, J, Wilson, R, Park, H, et al. A systematic review of barriers and interventions to improve appropriate use of therapies for sickle cell disease. Journal of the National Medical Association 2009;10(10):1022‐33.

Higgins 2011a

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 www.cochrane‐handbook.org.

Higgins 2011b

Higgins JPT, Deeks JJ, editor(s). Chapter 7: Selecting studies and collecting data. 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. Available from www.cochrane‐handbook.org.

Higgins 2011c

Higgins JPT, Altman DG, Sterne JAC on behalf of the Cochrane Statistical Methods Group and the Cochrane Bias Methods Group, 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 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Higgins 2011d

Higgins JPT, Deeks JJ, Altman DG on behalf of the Cochrane Statistical Methods Group, editor (s). Chapter 16: Special topics in statistics. In: Higgins JPT, Green S, editor(s). Cochrane Handbook of Systematic Reviews of Interventions. Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

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.

NCCMH 2010

National Collaborating Centre for Mental Health. Depression in adults with a chronic physical health problem: treatment and management. Clinical guideline [CG91]. www.nice.org.uk/guidance/cg91 (accessed 16 April 2018). [ISBN:: 13: 978‐1‐904671‐86‐2]

NCCPC 2009

The National Collaborating Centre for Primary Care. Medicines adherence: involving patients in decisions about prescribed medicines and supporting adherence. NICE Guideline CG76. https://www.nice.org.uk/guidance/cg76 (accessed 20 June 2016).

NICE 2009

National Institute for Health and Care Excellence. Depression in adults with chronic physical health problem: recognition and management. NICE guideline CG91. www.nice.org.uk/guidance/cg91 (accessed 20 June 2016).

NICE 2010

NICE National Institute for health and Care Excellence. Sickle cell disease. http://cks.nice.org.uk/sickle‐celldisease#!backgroundsub:3 (accessed 20 June 2016).

Payne 2008

Payne KA, Rofail D, Baladi JF, Viala M, Abetz L, Desrosiers MP, et al. Iron chelation therapy: clinical effectiveness, economic burden and quality of life in patients with iron overload. Advances in Therapy 2008;25(8):725‐42.

Piel 2012

Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW, Dewi M, et al. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model‐based map and population estimates. Lancet 2012;381(9861):142‐51.

Piel 2014

Piel, FB, Weatherall DJ. The α‐thalassemias. New England Journal of Medicine 2014;371(20):1908‐16.

Pleasants 2014

Pleasants S. Epidemiology: a moving target. Nature 2014;515:S2‐3.

Rees 2010

Rees DC, Williams TN, Gladwin MT. Sickle‐cell disease. Lancet 2010;376(9757):2018‐31.

Reeves 2011

Reeves BC, Deeks JJ, Higgins JPT, Wells GA on behalf of the Cochrane Non‐Randomised Studies Methods Group. Chapter 13: Including non‐randomized 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. Available from www.cochrane‐handbook.org.

RevMan 2014 [Computer program]

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

Rofail 2010

Rofail D, Viala M, Gater A, Abetz‐Webb L, Baladi JF, Cappellini MD. An instrument assessing satisfaction with iron chelation therapy: Psychometric testing from an open‐label clinical trial. Advances in Therapy 2010;27(8):533‐46.

Rund 2005

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

Ryan 2014

Ryan R, Santesso N, Lowe D, Hill S, Grimshaw J, Prictor M, Kaufman C, Cowie G, Taylor M. Interventions to improve safe and effective medicines use by consumers: an overview of systematic reviews. Cochrane Database of Systematic Reviews 2014, Issue 4. [DOI: 10.1002/14651858.CD007768.pub3]

Schünemann 2011a

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH on behalf of the Cochrane Applicability and Recommendations Methods Group and the Cochrane Statistical Methods Group. Chapter 11: Presenting results and ‘Summary of findings’ tables. 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. Available from www.cochrane‐handbook.org.

Sterne 2011

Sterne JAC, Egger M, Moher D on behalf of the Cochrane Bias Methods Group, editor(s). Chapter 10: Addressing reporting biases. 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. Available from www.cochrane‐handbook.org.

Sterne 2016

Sterne JAC, Higgins JPT, Reeves BC on behalf of the development group for ROBINS‐I. ROBINS‐I: a tool for assessing Risk Of Bias In Non‐randomized Studies of Interventions, Version 7 March 2016. www.riskofbias.info (accessed 19 April 2016).

Telfer 2006

Telfer P, Coen PG, Christou S, Hadjigavriel M, Kolnakou A, Pangalou E, et al. Survival of medically treated thalassemia patients in Cyprus. Trends and risk factors over the period 1980‐2004. Haematologica 2006;91(9):1187‐92.

Thomas 2013

Thomas, V, Rawle, H, Abedian, M, Ferguson, A. Implementing the NICE clinical guideline 91 ('Depression in adults with a chronic physical health problem') in a haematology department. Clinical Psychology Forum 2013;246:41‐45.

Trachtenberg 2012

Trachtenberg FL, Mednick L, Kwiatkowski JL, Neufeld EJ, Haines D, Pakbaz Z, et al. Beliefs about chelation among thalassemia patients. Health and Quality of Life Outcomes 2012;10:148.

Trachtenberg 2014

Trachtenberg, F, Gerstenberger, E, Xu, Y Mednick, L, Sobota, A, Ware, H, et al. Relationship among chelator adherence, change in chelators, and quality of life in Thalassemia. Quality of Life Research 2014;23:2277‐88.

UK Thalassaemia Society 2008

United Kingdom Thalassaemia Society. Standards for the clinical care of children and adults with thalassaemia in the UK. UK Thalassaemia Society2008; Vol. 2nd edition:120.

Vekeman 2016

Vekeman F, Sasane M, Cheng WY, Ramanakumar AV, Fortier J, Qiu Y, et al. Adherence to iron chelation therapy and associated healthcare resource utilization and costs in Medicaid patients with sickle cell disease and thalassemia. Journal of Medical Economics 2016;19(3):292‐303.

Wertheimer 2003

Wertheimer A, Santella T. Medication compliance research. Journal of Applied Research in Clinical and Experimental Therapeutics 2003;3(3):254‐61.

WHO 2003

World Health Organization. Adherence to long‐term therapies: Evidence for action. WHO2003:194.

Yawn 2014

Yawn BP, Buchanan GR, Afenyi‐Annan AN, Ballas SK, Hassell KL, James AH, et al. Management of sickle cell disease:summary of the 2014 evidence‐based report by expert panel members. JAMA 2014;312(10):1033‐48.

Fortin 2016

Fortin PM, Madgwick KV, Trivella M, Hopewell S, Doree C, Estcourt LJ. Interventions for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia. Cochrane Database of Systematic Reviews 2016, Issue 9. [DOI: 10.1002/14651858.CD012349]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Aydinok 2007

Methods

Study design: single‐centre RCT

Study grouping: parallel group
Study duration: treatment duration 12 months; follow‐up: not stated

Participants

Baseline characteristics

DFP, DFO

  • Total # of participants: 12 randomised; 8 analysed

  • Age mean (SD): 16.6 (4.8) years, range 9 to 23 years

  • Sex: not reported

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): 100% β‐thalassaemia

  • Baseline ferritin levels (ng/mL) mean (SD): 4453 (2858)

  • Previous iron chelation: not reported

  • Duration of any iron chelation: not reported

  • LIC (mg/g) mean (SD): 27.0 (13.4)

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Hb, g/L mean (SD): 89 (5)

DFP

  • Total # of participants: 12

  • Age mean (SD): 15.9 (4.2) years

  • Sex: not reported

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): 100% β‐thalassaemia

  • Baseline ferritin levels (ng/mL): 4070 (3223)

  • Previous iron chelation: not reported

  • Duration of any iron chelation: not reported

  • LIC (mg/g): 30.7 (10.6)

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Hb, g/L mean (SD): 89 (5), range 9 to 23 years

Inclusion criteria: iron‐overloaded people with thalassaemia at least 4 years old

Exclusion criteria: lack of compliance, known toxicity or intolerance preventing therapy with DFO and DFP, neutropenia (neutrophils < 1.5×109/L), thrombocytopenia (platelets < 100×109/L), renal, hepatic or decompensated heart failure, active viral illness being treated with interferon‐α/ribavirin, repeated Yersinia infections, HIV–positivity, pregnancy or nursing, and patients of reproductive age not taking adequate contraceptive precautions

Interventions

Treatment arm: DFO (50 mg/kg/day subcutaneously twice weekly (mean (SD) dose: 43.8 (2.8) mg/kg)) combined with DFP (75 mg/kg/day, daily (mean (SD) dose: 78.2 (1.4) mg/kg/day))
Comparator arm: DFP (75 mg/kg/day, daily (mean (SD) dose: 78.2 (2.6) mg/kg/day))

Outcomes

Adherence: compliance was assessed by drug accounting at each visit (by counting the returned empty blisters of DFP and used vials of DFO) as well as by a trial‐specific questionnaire completed by the participants and/or their legal representative/guardian at quarterly intervals.

The same questionnaire also served for the assessment of tolerance to treatment and QoL

Trial‐reported outcomes

1. Changes in LIC and SF (primary outcome)
2. Total iron excretion
3. Urinary iron excretion
4. Iron balance
5. Cardiac function (Echo)
6. Toxicity
7. Assessment of tolerance to treatment and QoL

Identification

Source of funding: none stated although the drugs were supplied by Lipomed AG, Switzerland

Notes

All participants had prior exposure to DFO (dose, schedule and duration were not reported) and all had a washout period of 2 weeks with no iron chelation before initiating trial treatment
Sample‐size calculation not reported

Country: Turkey

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomization sequence was generated by the Department of Mathematical Statistics at the University of Berne, Switzerland according to local policy". Following central registration of a subject by the investigator, the trial co‐ordinator assigned the intervention according to the randomisation sequence

Allocation concealment (selection bias)

High risk

The trial report states that the intervention was assigned according to the randomisation sequence “without concealing the sequence prior to allocation”

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

The authors did not report any information as to whether participants, personnel were blinded to treatment allocation but one treatment subcutaneous and other oral so difficult to blind

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

The authors did not report any information as to whether outcome assessors were blinded to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

There was an imbalance in missing data across the treatment arms. 4 participants from the comparator group (DFO) were not included in the outcome analysis: 2 withdrew consent due to refusal to take DFO; 1 died from arrhythmia induced congestive heart failure at start of trial; and 1 developed agranulocytosis at week 14

Selective reporting (reporting bias)

Low risk

All outcomes were reported

Other bias

Unclear risk

There is an imbalance in baseline LIC and Ferritin between groups

Badawy 2010

Methods

Study design: RCT

Study grouping: parallel group

Length of trial or follow‐up not stated. Not stated if open label; but no mention of blinding and DFO is infusion versus tablet

Participants

Baseline characteristics

DFP, DFO

  • Total # of participants: 50

  • Age: ≥ 8 years

  • Sex: not reported

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): 100% β‐thalassaemia

  • Baseline ferritin levels (ng/mL): not reported

  • Previous iron chelation: DFO

  • Duration of any iron chelation: not reported

  • LIC (mg/g): not reported

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Hb, g/L: not reported

DFP

  • Total # of participants: 50

  • Age: ≥ 8 years

  • Sex: not reported

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): β‐thalassaemia

  • Baseline ferritin levels (ng/mL): not reported

  • Previous iron chelation: DFO

  • Duration of any iron chelation: not reported

  • Liver iron concentration LIC (mg/g): not reported

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Hb, g/L: not reported

DFO

  • Total # of participants: 50

  • Age: greater or equal to 8 years

  • Thalassaemia genotype N (%): 100% β‐thalassaemia

  • Baseline ferritin levels (ng/mL): not reported

  • Previous iron chelation: DFO

  • Duration of any iron chelation: not reported

  • LIC (mg/g): not reported

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Hb, g/L: not reported

Inclusion criteria: 8 years, RBC transfusion every 3 to 4 weeks, on DFO prior to study as single therapy.

Exclusion criteria: not stated

Participants PRBCs /3 – 4 weeks to maintain Hb > 9 g/dL

Interventions

DFP, DFO

  • Medication intervention: daily DFP, DFO twice‐weekly DFO (40 mg/kg/day); Deferipron e (75 mg/kg/day).

DFP

  • Medication intervention: daily DFP Deferipron e (75 mg/kg/day).

DFO

  • Medication intervention: DFO 5 days/week DFO (40 mg/kg/day)

Outcomes

Adherence to iron chelation therapy rates

Questionnaire on chelation therapy, reasons for non‐compliance, side effects, life activities, transfusion regimen

Trial‐reported outcomes

1. CBC monthly

2. SF levels

3. liver and kidney functions

4. blood glucose level

5. serum calcium and phosphorus/3 months and T3, T4,TSH, LH, FSH

6. echocardiography

7. bone density

8. auditory and visual examination twice

Identification

Sponsorship source: Zagazig University Hospital, Zagazig

Country: Egypt

Setting: University Hospital

Comments: Abstract Poster 124

Authors name: Sherif Badawy

Institution: Ann Robert H. Lurie Children’s Hospital of Chicago

Email: [email protected]

Address: Ann Robert H. Lurie Children’s Hospital of Chicago Northwestern University Feinberg School of Medicine225 East Chicago Avenue, Box 30, Chicago, Illinois 60611‐2605

Notes

Contacted author and study data not available at this time. Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Judgement comment: no description of sequence generation

Allocation concealment (selection bias)

Unclear risk

Judgement comment: no description of allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment no description, but one drug is subcutaneous injection (DFO). Open label

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

Judgement comment: no description of blinding of assessors

Incomplete outcome data (attrition bias)
All outcomes

High risk

Judgement comment: no data on number of participants who completed the study and how many in each group experienced complications. Lack of detail on number of compliant or non‐compliant participants

Selective reporting (reporting bias)

High risk

Judgement comment: not clear which groups and how many experienced adverse events. No data reported on SF or other outcomes

Other bias

Unclear risk

Judgement comment: results of the trial were not published in detail and no data available when authors were contacted

Bahnasawy 2017

Methods

Study design: single‐centre RCT

Study grouping: parallel group

Study duration: 6 months

Participants

Baseline characteristics

Comprehensive medication management

  • Total # of participants: 24

  • Age (mean (SD)): 12 (2.7)

  • Sex N (%): F: 15 (62.5); M: 9 (37.5)

  • Ethnicity: not reported

  • Thalassaemia genotype (%): β‐thalassaemia major 100%

  • Baseline ferritin levels (ng/mL) (mean (SD)): 3949 (1864)

  • Previous iron chelation: N/A

  • Duration of any iron chelation: N/A

  • LIC (mg/g): not stated

  • Splenectomy n (%): 6 (25.9)

  • QoL PedsQL median (IQR): 55.16 (43.42 ‐ 63.75)

  • Hb, g/L: not stated

Standard care (as defined in the trial)

  • Total # of participants: 24

  • Age (mean (SD)): 13 (2.8)

  • Sex N (%): F: 15 (62.5); M: 9 (37.5)

  • Ethnicity: not reported

  • Thalassaemia genotype (%: β‐thalassaemia major 100%

  • Baseline ferritin levels (ng/mL) (mean (SD)): 3871 (1881)

  • Previous iron chelation: N/A

  • Duration of any iron chelation: N/A

  • LIC (mg/g): not stated

  • Splenectomy n (%): 9 (37.5)

  • QoL PedsQL median (IQR): 49.12(38.13 ‐ 56.95)

  • Hb, g/L: not stated

Inclusion criteria: transfusion‐dependent children with β‐thalassaemia major aged 8 to 18 years with SF level of more than 1000 µg/L

Exclusion criteria: people with cognitive impairment

Interventions

Comprehensive medication management

  • interview with participants at each visit, drug‐related problems identified, care plan introduced / monitored to include dosage modification, education. Follow‐up compliance via regular phone calls

Standard care (as defined in the trial)

  • all participants presented to the clinic regularly every 2 ‐ 4 weeks according to the need for receiving blood transfusion, blood samples were drawn for CBC assessment. Physical examination was done by physician including assessment of hepatomegaly, splenomegaly and any health‐related problems

Outcomes

Adherence to iron chelation therapy rates

"DRP identification: The clinical pharmacist analysed the collected data to detect whether any DRPs existed and allocated them to one of the seven categories as classified by Cipolle et al. [18]: unnecessary drug therapy, need for additional drug therapy, ineffective drug product, dosage too low, adverse drug reaction, dosage too high, non‐compliance"

Trial‐reported outcomes

1. SF levels were measured at baseline, 3 months and after 6 months

2. CBC with WBC differential was assessed at every visit, and SCr and ALT were measured routinely for all the participants every 3 months

3. Health‐related QoL was assessed at baseline and at the end of the trial (after 6 months) using PedsQL™ 4.0 Generic Core Scale questionnaire. PedsQL is a 23‐item multidimensional model with 4 domains for paediatric health‐related QoL measurement: physical functioning (8 items), emotional functioning (5 items), social functioning (5 items) and school functioning (5 items) (19).

Identification

Sponsorship source: not stated

Country: Egypt

Setting: Hematology clinic

Authors name: Lamia El Wakeel

Institution: Pediatric Hematology Clinic, Children’s Hospital, Ain Shams University,

Email: [email protected]

Address: Lamia El Wakeel, Pediatric Hematology Clinic, Children’s Hospital, AinShams University, 4, Street 292 New Maadi, Cairo, Egypt

Notes

Sample‐size calculation not reported.
Drug‐related outcomes do not have any comparable data reported. Only outcomes with comparable data reported are SF levels and health‐related QoL

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "The study was a prospective, randomized, controlled study. It was conducted on pediatric BTM patients admitted to the Pediatric Hematology Clinic," Stratified randomization was used considering the iron chelation therapy as the stratification factor

Judgement comment: no description of how randomisation was done or by whom

Allocation concealment (selection bias)

Unclear risk

The control group (n = 24) received standard medical care by a physician while the intervention group received standard medical care plus clinical pharmacist‐provided services.

Judgement comment: no description of how participants were allocated to the pharmacist intervention or standard care

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment: not possible to blind a pharmacist intervention versus no pharmacist intervention

Blinding of outcome assessment (detection bias)
All outcomes except mortality

High risk

Judgement comment: no indication that outcome assessors where different from pharmacists who implemented the intervention. Also most outcomes were reported only in the intervention group except for ferritin levels and health‐related QoL

Incomplete outcome data (attrition bias)
All outcomes

High risk

Judgement comment: all drug‐related outcomes were only reported in the intervention group including adherence ‐ no comparative data available. Multiple interventions in small number of participants

Selective reporting (reporting bias)

High risk

Judgement comment: drug‐related outcomes reported only in intervention group. No comparative data. The participants within the intervention arm seem to have complex and multiple changes. Difficult to tease out the actual intervention that effected a change

Other bias

Unclear risk

Judgement comment: small sample size and only report intervention group

Calvaruso 2015

Methods

Study design: RCT

Study grouping: parallel group

This trial was designed as a 5‐year, multicentre, randomised, open‐label trial with blinded data management and data analyses to evaluate whether the DFP treatment is superior to the DFO treatment

Follow‐up after trial. An additional 5 years of follow‐up after the end of the trial was planned to collect data on the survival, cause of death and chelation treatment of this cohort of participants. During this period, the participants were allowed to change their chelation treatment

Participants

Baseline characteristics

DFP

  • Total # of participants: 47

  • Age: mean (SD): 41.3 (14.8)

  • Sex n (%): F: 24 (50)

  • Ethnicity: not reported

  • Thalassaemia genotype (%): thalassaemia Intermedia 100%

  • Baseline ferritin levels (ng/mL) median (IQR): 1221 (743)

  • Age at initiation of DFO years: mean (SD): 29.9 (16.8)

  • LIC (mg/g/dw) median (IQR): 3800 (2800)

  • Splenectomy n (%): 42 (89.3)

  • QoL: mean (SD): not reported

  • Hb, g/L mean (SD): 88 (10)

DFO

  • Total # of participants: 41

  • Age: mean (SD): 41.2 (14.3)

  • Sex n (%): F: 23 (51.1)

  • Ethnicity: not reported

  • Thalassaemia genotype (%): thalassemia intermedia 100%

  • Baseline ferritin levels (ng/mL) (median (IQR)): 1,122 (910)

  • Age at initiation of DFO years: mean (SD): 29.6 (17.4)

  • LIC (mg/g/dw) median (IQR): 3800 (4668)

  • Splenectomy n (%): 35 (77.7)

  • QoL: mean (SD): not reported

  • Hb, g/L mean (SD): 89 (12)

Inclusion criteria: people with thalassaemia intermedia (based on clinical and molecular criteria), SF between 800 and 3000 µg/L, 13 years of age, consent from patient or parent or guardian (if 13 to 18)

Exclusion criteria: known intolerance to treatment, platelet count < 100 ×109/L, white cell count of < 3 ×109/L, severe liver damage, sepsis or heart failure (or both)

Pretreatment: none of the participants in the DFP group and 8 in the DFO group withdrew from the trial. 1 participant in the DFP group and 3 in the DFO group changed their chelation therapy (P value = 0.357)

If the participants were treated with a subcutaneous administration of DFO (30 ‐ 50 mg/kg per day, 8 – 12 hours for 5 days a week) before inclusion in the trial, a DFO washout was executed for 1 week before randomisation.The minimum number of participants required for each treatment group was calculated, assuming equal allocation under the hypothesis of equality between the 2 treatment groups at each point during the course.The recommended number of participants was 30.

One participant in the DFP group and 3 in the DFO group changed their chelation therapy

Interventions

DFP

  • DFP (Apotex; Toronto, ON, Canada) administered at 75 mg/kg/day, divided into 3 oral daily doses for 7 days/week

DFO

  • DFO (BiofuturaPharma, Omezia, Italy), administered by subcutaneous infusion (8 – 10 hours) at 50 mg/kg per day for 5 days/week

Treatment failure was defined as an increase in the SF level to greater than 1000 lg/L from baseline, confirmed by at least 2 consecutive determinations. Participants who failed were switched to the alternative treatment and followed until the end of the trial. The criteria for a dosage reduction to 50 mg/kg of DFP per day were arthralgia and nausea, and the criterion for a reduction to 30 mg/kg of DFO per day was a local reaction at the site of infusion. Both treatments were reduced if the ferritin levels for 2 consecutive determinations were less than 400 lg/L. The treatment was resumed when the ferritin levels were greater than 700 lg/L for at least 2 determinations

Outcomes

Adherence to iron chelation therapy rates

Compliance was assessed by counting the number of DFP pills in each returned bag and by assessing the number of infusions of DFO registered on the electronic pump

Trial‐reported outcomes
1. The primary endpoint was treatment effectiveness, evaluated as the mean change in the SF level over the 5‐year period. This type of evaluation strengthened the power of the test for the sample‐size calculation compared with the standard.

2. The secondary endpoints were safety and survival analysis after 5 years

Identification

Sponsorship source: contract grant sponsor: Franco and Piera Cutino Foundation

Country: Italy (17 centres)

Setting: haematology and thalassaemia clinical centres at institutions

Recruitment: January 2001 to January 2006

Trial registration: NCT00733811

Authors name: Aurelio Maggio

Institution: Unita Operativa Complessa Ematologia II,

Email: [email protected]

Address: U.O.C. Ematologia II, A.O.R. “Villa Sofia – V. Cervello”, Palermo, Italy

Notes

Sample‐size calculation reported for primary outcome

Notes: 9 participants changed from DFP therapy

5 to DFO

2 to none

1 to DFX

1 to
DFP‐DFO

6 participants changed from DFO therapy

4 to DFP
1 to DFX
1 to DFP‐DFO

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomization sequence was based on a computer‐ randomized list arranged in permuted blocks of 10 with a 1:1 ratio."

Allocation concealment (selection bias)

Low risk

To ensure for allocation concealment, treatments were assigned by telephone contact from the coordinating centre. The sequence was concealed until the interventions were assigned. Randomization was performed for each consecutive patient after verification of the exclusion criteria

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Quote: "open‐label trial"

Judgement comment: 1 of 2 arms was desferal pump infusers, participants would know. Participants on DFO attended for weekly blood tests.

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Low risk

Quote: "with blinded data management and data analysis"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No loss to follow‐up for 5‐year trial

Selective reporting (reporting bias)

Low risk

All outcomes reported

Other bias

Unclear risk

Unclear how participant variation relating to SF levels may have had effect on results. Although all outcomes were reported for the 5 year trial in the 5 years of follow‐up only mortality was reported

El Beshlawy 2008

Methods

Study design: single‐centre RCT

Study grouping: parallel group, follow‐up for 54 weeks

Participants

Baseline characteristics

DFP/DFO

  • Total # of participants: 18

  • Age (mean (SD): 11.0 (4.9)

  • Sex: F: 10; M: 8

  • Ethnicity: not reported

  • Thalassaemia genotype N (%) : β‐thalassaemia major: 100%

  • Baseline ferritin levels (ug/mL) (mean (SD) (range)): 2865 (983) (1500 – 4800)

  • Previous iron chelation: not reported

  • LIC (mg/g) mean (SD) (range): 17.1 (9.1) (4.9 ‐ 33.6) N = 16

  • Splenectomy n (%): 11 (61)

  • QoL mean (SD): not reported

  • Hb, g/L (mean (SD) (range): 68 (5) (55 – 75)

DFP

  • Total # of participants: N = 18

  • Age (mean (SD) (range)): 10.8 (5.1) (5 ‐ 26)

  • Sex: F: 6; M: 12

  • Ethnicity: not reported

  • Thalassaemia genotype N (%) : β‐thalassaemia major: 100%

  • Baseline ferritin levels (ug/mL) (mean (SD) (range)): 2926 (1107) (1560 – 5000)

  • Previous iron chelation: not reported

  • LIC (mg/g) (mean (SD) (range)): 15.8 (7.1) (2.3 – 29.3) N = 17

  • Splenectomy n (%): 9 (50)

  • QoL mean (SD): not reported

  • Hb, g/L mean (SD) (range): 69 (6) (58 – 80)

DFO

  • Total # of participants: N = 20

  • Age (mean (SD) (range)): 13.1 (5.9) (5.5 ‐ 24)

  • Sex: F: 9; M: 11

  • Ethnicity: not reported

  • Sickle cell genotype N (%) ‐ not applicable:

  • Thalassaemia genotype N (%): β‐thalassaemia major: 100%

  • Baseline ferritin levels (ug/mL) (mean (SD)(range)): 2 838 (967) (1500 – 4300)

  • Previous iron chelation: not reported

  • LIC (mg/g) mean (SD) (range): 22.5 (10.1) (6.0 – 41.7) N = 15

  • Splenectomy n (%): 10 (50)

  • QoL mean (SD): not reported

  • Hb, g/L mean (SD) (range): 69 (5) (60 – 80)

Inclusion criteria: males or females with thalassaemia major attending the Hematology Clinic at Cairo University Children Hospital; participants had to be iron overloaded with transfusion dependency and older than 4 years of age

Exclusion criteria: known to have DFP or DFO toxicity; neutrophil count less than 1.5×109/L; platelet count less than 100×109/L; renal or hepatic insufficiency; decompensated heart failure; without contraceptive precaution; pregnant or nursing

Interventions

DFP/DFO

  • DFP + DFO (dose 60 ‐ 83 mg/kg/day and DFO 23 to 50 mg/kg per dose) DFP 7 days and DFO over 8 hours 2 days/week)

DFP

  • DFP only (dose 60 to 83 mg/kg/day) 7 days per week

DFO

  • DFO 23 to 50 mg kg/day monotherapy for 5 days/week

Outcomes

Adherence to iron chelation therapy rates

Compliance was assessed by performing a drug accounting at each patient visit by counting the returned empty blisters of DFP and used vials of DFO

Trial‐reported outcomes

1. Incidence of chelation therapy‐related SAEs (reported in AEs)

2. Iron overload defined by ferritin over 1000 µg/L and/or clinical symptoms and/or signs of iron overload and/or need for medically indicated additional or change in chelation therapy (mean ferritin levels extrapolated from graph ‐ no SD provided)

3. Other AEs related to iron chelation (in this trial participants with an event are reported. 1 person could experience more than 1 event)

4. LIC mg/g dry weight (change from baseline (extrapolated from graph Least squares means / lower and upper value))

Identification

Sponsorship source

Country: Egypt

Setting: Hematology Clinic at Cairo University Children Hospital, Egypt

Comments: 2 authors from Lipomed (DFP): C. Manz : C. Tarabishi Clinical Research Development, Lipomed AG, Arlesheim, Switzerland

Authors name: A. El‐Beshlawy

Institution: Faculty of Medicine, Cairo University,

Email: [email protected]

Address: Faculty of Medicine, Cairo University, 32 Falaky Street, Bab El‐Louk, Cairo, Egypt

Notes

Sample‐size calculation reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Judgement comment: no description of how randomisation was accomplished: The participants were randomly assigned into 1 of 3 treatment arms

Allocation concealment (selection bias)

Unclear risk

Judgement comment: no description of allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

No mention of blinding ‐ since DFO is an injection and DFP is oral likely participants and personnel not blinded

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

Judgement comment: no blinding mentioned

Incomplete outcome data (attrition bias)
All outcomes

High risk

Judgement comment: a total of 10 participants dropped out of the trial as a result of several complications. Only 56 participants completed 54 weeks of treatment. Evaluation of LIC could not be done in another 8 participants. Reports on per protocol participants

Selective reporting (reporting bias)

High risk

Compliance not reported as number or percentage of participants compliant throughout trial: "Four patients, all treated with DFO‐based regimen, were excluded from the study due to lack of compliance. Compliance was otherwise excellent during the entire study. The majority of patients had no problems with the intake and swallowing of the DFP tablets. By contrast, 80% of patients in the combination arm and 76% of patients in the DFO monotherapy arm complained about difficulties in the parenteral use of DFO or problems to insert a needle", SF and LIC are partially reported in charts and no actual numbers are provided in the text. Also the focus on UIE over LIC and SF measures is misleading as DFP is known to have a higher UIE but this can be highly variable over multiple measurements. LIC is the gold standard and there was no difference in this outcome between groups.

Other bias

Unclear risk

There was a higher incidence of AEs in the combined group and the DFP group versus the DFO group

Elalfy 2015

Methods

Study design: RCT in 2 treatment centres

Study grouping: parallel group

Study duration: 1 year

Participants

Baseline characteristics

Group A: DFP/DFO

  • Total # of participants: 48

  • Age: mean (SD): 15.25 (2.31)

  • Sex: male n (%): 30 (62.5)

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): Not stated all participants appear to have β‐thalassaemia major

  • Baseline ferritin levels (ng/mL): mean (SD): 4379.07 (895.00); range 3632 ‐ 6210

  • Duration of any iron chelation (years): mean (SD): 8.71 (2.7)

  • LIC (mg/g): mean (SD): 12.69 (2.23); range: 12.69 ‐ 2.23

  • Splenectomy n (%): 21 (43.7)

  • QoL mean (SD): 63.09 (5.77)

  • Hb, g/L mean (SD): 81.1 ( 3.3)

  • Mean geometric cardiac T2*(ms): mean (SD): 16.32 (1.82); range: 14.9 – 18.2

Group B: DFP/DFX

  • Total # of participants: 48

  • Age: mean (SD): 14.05 (2.21)

  • Sex: male n (%): 32 (66.6)

  • Ethnicity: not reported

  • Thalassaemia genotype N (%): not stated all participants appear to have β‐thalassaemia major

  • Baseline ferritin levels (ng/mL) mean (SD): 4289.19 (866.21); range: 3451 ‐ 7122

  • Duration of any iron chelation (years): mean (SD): 8.95 (2.8)

  • LIC (mg/g): mean (SD): 12.52 (2.28); range: 9.82 ‐ 15.12

  • Splenectomy n (%): 20 (41.6)

  • QoL mean (SD): 63.38 (5.98)

  • Hb, g/L mean (SD): 79 (3.8)

  • Mean geometric cardiac T2*(ms): mean (SD):16.59 (1.85); range: 15.7 ‐ 18.9

Inclusion criteria: people with β‐thalassaemia major aged 10 – 18 years with severe iron overload defined as: ferritin > 2500 μg/L on maximum tolerated dose of a single iron chelator with up trend of ferritin over the last 12 months prior to the study. People 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 or exertion, orthopnoea, exercise intolerance, lower extremity oedema, arrhythmias). Adequacy of prior chelation defined as taking 75% of the calculated dose/month on maximum tolerated dose with upward ferritin trend

Exclusion criteria: past history of agranulocytosis, clinically significant GI or renal disease, clinical cardiac disease, or with LVEF < 50% on baseline echocardiography; evidence of active hepatitis or serum transaminases > 3 times above ULN or renal impairment (serum creatinine > ULN) participation in a previous investigational drug study within the 30 days preceding screening, known allergy to DFX, DFP, and DFO.

Pre‐treatment: baseline difference in mean Hb (P 0.004)

Interventions

DFP/DFO

  • 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 hours 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/week

DFP/DFX

  • DFP 75 mg/kg/day, divided into 2 doses taken orally at 8 a.m. and 3 p.m. combined with DFX30 mg/kg/day taken orally at 10 p.m. for 7 days/week

To achieve an acceptable treatment washout, chelation therapy was withdrawn for 2 weeks before randomisation, after verifying inclusion and exclusion criteria. The transfusion regimen aimed to maintain the participants pre‐transfusion Hb ≥80 g/L by receiving approximately 15 mL/kg packed RBCs every 3 – 4 weeks

Outcomes

Adherence to iron chelation therapy rates

Compliance was evaluated by counting of returned tablets for the oral chelators and of the vials for DFO. The percentage of actual dose that participant had taken in relation to the total prescribed dose was calculated

Trial‐reported outcomes

1. % change in SF (from baseline to the end of trial)

2. % change in LIC (from baseline to the end of trial)

3. % change in cardiac MRI (from baseline to the end of trial)

4. SAEs and AEs (safety assessment)

5. Compliance

6. Satisfaction

7. QoL

Identification

Sponsorship source: Ain Shams University

Country: Egypt and Oman

Setting: Thalassemia treatment centres (Ain Shams University, Egypt and Sultan Qaboos University Hospital, Oman)

Comments: Government Clinical Trial NCT01511848

Authors name: Amira Abdel Moneam Adly,

Institution: Department of Pediatrics, Ain Shams University, Cairo, Egypt

Email: [email protected]

Address: 6 A ElSheshini street, Shoubra, Soudia buildings, Cairo, Egypt

Notes

The chelation regimens in the last year prior to the trial were daily DFX (14 participants), daily DFP (29 participants), and DFP 4 days/week alternating with subcutaneous DFO 3 days/week (53 participants)

Sample‐size calculation reported
Author contacted for additional info on SF 36 mean (SD) 6 months and end of trial

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "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

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

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Oral versus subcutaneous medication therefore participants would be aware which medication arm they had been randomised to

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Low risk

Quote: "open‐label study with blinded data management and data analyses"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement comment: treatment was started within the following 24 hr, and all the included participants continued till the end of study with no participants were lost follow‐up

Selective reporting (reporting bias)

Unclear risk

Judgement comment: provide only P values for patient satisfaction, satisfaction with ICT self‐reported satisfaction and all 'significantly' higher in group B; no actual end of trial data provided (mean (SD)). All outcomes are reported

Other bias

Unclear risk

Judgement comment: it is not clear how the investigators would have known that infections, GI disorders or skin disorders were not related to the drug therapies

Galanello 2006

Methods

Study Design: 2‐arm parallel RCT conducted in Italy and Greece
Number of centres: multicentre (3 centres)
Duration of treatment: 12 month
Follow‐up: not stated.

Participants

DFP/DFO

  • Total # of participants: randomised 30, analysed 29 (withdrawn after 2 days on trial before taking DFP)

  • Age (mean (SD): 19.8 (6.1) years

  • Sex: F: 13; M: 16

  • Ethnicity: not reported

  • Thalassaemia genotype N (%) : β‐thalassaemia major: 100%

  • Baseline ferritin levels (ug/mL) mean (SD): 2048 (685)

  • Previous iron chelation: not reported

  • LIC (mg/g) mean (SD) (range): 17.1 (9.1) (4.9 – 33.6) N = 16

  • Splenectomy n (%): 11 (61)

  • QoL mean (SD): not reported

  • Hb, g/L mean (SD) (range): 68 (5) (55 – 75)

DFP/DFO

  • Total # of participants: randomised 30, analysed 30

  • Age (mean (SD)): 18.7 (4.8) years

  • Sex: F: 18; M: 12

  • Ethnicity: not reported

  • Thalassaemia genotype N (%) : β‐thalassaemia major: 100%

  • Baseline ferritin levels (ug/mL) (mean (SD): 2257 (748)

  • Previous iron chelation: not reported

  • LIC (mg/g) mean (SD) (range): 17.1 (9.1) (4.9 – 33.6) N = 16

  • Splenectomy n (%): 11 (61)

  • QoL mean (SD): not reported

  • Hb, gL mean (SD) (range): 68 (5) (55 – 75)

Inclusion criteria: participants were 10 years or older with a diagnosis of thalassaemia major undergoing iron chelation therapy with subcutaneous DFO, with a SF value between 1000 ‐ 4000 μg/L over the previous year.

Exclusion criteria: not reported

Interventions

DFO: 20 ‐ 60 mg/kg/day subcutaneously on 5 ‐ 7 days a week (mean (SD) dose at baseline: 34.8 (8.9) mg/kg/day and at end of trial: 37.8 (8.9) mg/kg/day))

DFO/DFP: DFO 20 ‐ 60 mg/kg/day subcutaneously on 2 days a week (mean (SD) dose DFO for the 29 participants who completed the trial at baseline: 36.0 (5.8) mg/kg/day and at end of trial: 33.3 (6.64) mg/kg/day) with DFP 25 mg/kg/ body weight 3 x daily for 5 days a week)

Outcomes

Adherence see compliance below

Trial‐reported outcomes

1. SF change at 1 year
2. LIC (measured by SQUID) change at 1 year
3. ALT
4. FBC
5. Zinc levels
6. AEs
7. Participant compliance: compliance with DFP was assessed by pill counts, diary cards and an electronic cap that recorded the time and date of each opening of the tablet container. Compliance with DFO was assessed by diary cards, weekly physical examination of infusion sites, and by the Crono™ infusion pump that recorded the number of completed infusions
Primary outcome: not identified

Identification

Source of funding: Apotex Research Inc, Toronto, Canada. The last author of the study is an Apotex employee

Notes

The trial inferred that participants had previously received DFO treatment but no details as to dose, schedule or duration were reported
Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The authors did not report any information about how randomisation was undertaken

Allocation concealment (selection bias)

Unclear risk

The authors did not report any information about how treatment allocation was concealed

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

Unclear risk

The authors did not report any information as to whether participants, personnel or outcome assessors were blinded to treatment allocation

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

The authors did not report any information as to whether outcome assessors were blinded to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Although 1 participant in the treatment group was withdrawn due to intolerance to DFP, this is unlikely to effect the findings of the trial

Selective reporting (reporting bias)

Unclear risk

Compliance to DFP was pre‐specified as an outcome but was not measured or reported in the manuscript

Other bias

Low risk

The trial appears to be free of other sources of bias

Hassan 2016

Methods

Study design: single‐centre RCT

Study grouping: parallel group

Trial duration: September 2014 to September 2015

Participants

Baseline characteristics

DFX

  • Total # of participants: 30

  • Age mean (SD): 8.9 (2.2)

  • Sex male/female: 9/21

  • Thalassaemia genotype (%): β‐thalassaemia major: 100%

  • Baseline ferritin levels (ng/mL) median (range): 3216 (2100 ‐ 5862)

  • Previous iron chelation: 100%

  • Duration of any iron chelation: not reported

  • LIC (mg/g): not reported

  • Splenectomy n (%): 4 (13.3)

  • QoL mean (SD): not reported

  • Hb, g/dL mean (SD): 85 (12)

DFO

  • Total # of participants: 30

  • Age mean (SD): 9.7 (1.9)

  • Sex male/female: 10/20

  • Thalassaemia genotype (%): β‐thalassaemia major: 100%

  • Baseline ferritin levels (ng/mL) median (range): 2773 (1980 ‐ 4884)

  • Previous iron chelation: 100%

  • Duration of any iron chelation: not reported

  • LIC (mg/g): not reported

  • Splenectomy n (%): 17 (56.7)

  • QoL mean (SD): not reported

  • Hb, g/dL mean (SD): 7.9 (2.4)

Inclusion criteria: transfusion‐dependent β‐thalassaemia major, ages were ≥ 6 years, and they had SF levels greater than 1500 µg/L and were on 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 and/or ocular toxicity related to iron chelation therapy, cardiac disease, and/or SAEs with DFO or DFX, and absolute heutrophilic count 1500/mm3 or platelet count 100,000/mm3

Pre‐treatment: significant difference between the 2 groups with participants having splenectomy 4 in DFX group compared to 17 in DFO group (P = 0.001), hepatitis C status 2 in DFX group compared to 11 in DFO group (P = 0.005) and baseline ALT baseline mean of 28.2 in the DFX group compared to 46.1 in the DFO group (P = 0.001)

Interventions

DFX

  • DFX was administered orally as a single daily dose of 20 ‐ 40 mg/kg/day on an empty stomach after dissolution in water, apple juice, or orange juice to assure adequate bioavailability. Starting dose of DFX was individualized based on the frequency of blood transfusions

DFO

  • DFO was administered at 20 ‐ 50 mg/kg/day via subcutaneous infusion over 8 ‐ 10 hours, 5 days per week

7‐day washout phase

Outcomes

Adherence to iron chelation therapy rates

During the study, we kept records of all dosages administered, all study medications that were dispensed and returned, and intervals between visits to determine compliance with the treatment. The patients’ parents were instructed to contact the investigator if the patients were unable to take the study drug as prescribed

Trial‐reported outcomes

1. decrease in the SF level to < 1500 μg/L

2. Safety of the drugs that were used

Identification

Sponsorship source: not stated

Country: Egypt

Setting: out‐patient paediatric hematology clinic Al‐ Hussein University Hospital, Al‐Azhar University, Cairo, Egypt

Comments: no conflict of interest.

Authors name: Dr Omar Atef Tolba

Institution: Cairo University Children's Hospital

Email: [email protected]

Address: Dr Omar Atef Tolba, Cairo University Children's Hospital, Department of Pediatrics, Cairo University, Egypt. Tel: +201222101717, +20233025539, Fax: +20233025539

There is no conflict of interest declared

Notes

Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "the patients were randomized in a 1:1 ratio based on permuted blocks to receive deferasirox (DFX) or deferoxamine (DFO) for one year."

Judgement comment: it is unclear risk as there is imbalance in the groups on several variables

Allocation concealment (selection bias)

Unclear risk

Judgement comment: allocation concealment not described and imbalance between groups

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment: oral tablet versus subcutaneous infusion ‐ unable to blind participants or personnel

Blinding of outcome assessment (detection bias)
All outcomes except mortality

High risk

Quote: "During the study, we kept records of all dosages administered, all study medications that were dispensed and returned, and intervals between visits to determine compliance with the treatment." Judgement Comment: Does not state if outcome assessors were blinded. Assessors would be aware the treatment participants were on.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote: "no discontinuation of drugs or drop‐out of follow‐up occurred."

Selective reporting (reporting bias)

High risk

Quote: "Post‐treatment levels of ALT and AST were significantly higher in the DFO group (p = 0.022, p = 0.020, respectively), both drugs have comparable safety profiles, as the adverse effects noted did not reach clinical significance or lead to discontinuation of treatment with either agent. In the light of the comparable efficacy and safety of both agents for the reduction of iron overload, as was reported in the monotherapy of patients with transfusion‐dependent thalassaemia (31, 32), the oral preparation merits convenience and therefore patient compliance and adherence to treatment regimen that needs to be taken on a long‐term basis."

"The oral DFX is recommended due to more convenience to assure adherence to treatment regimen."

Judgement comment: the data within this trial do not provide evidence that DFX assures adherence. Pre‐treatment ALT, AST were also higher in the DFO group ‐ and also reflects imbalance in randomisation. Most outcomes vaguely reported (i.e. compliance ‐ not percentages even though did a count and closely monitored). Also not clear if all drug‐related AEs reported (i.e. agranulocytosis). Further the evidence is uncertain from this trial that both drugs of comparable efficacy and safety

Other bias

Unclear risk

Small trial N = 60 and short‐term follow‐up. Sample‐size calculation not reported, and single‐centre trial

Maggio 2009

Methods

Study design: multicentre RCT

Study grouping: parallel group

Consecutive thalassaemia major participants (n = 275) were observed at the 25 SoSTE centres from September 30, 2000 to January 31, 2008

9 participants did not meet inclusion criteria and 53 patients declined to participate. The remaining 213 participants were included; 105 and 108 respectively, were randomly allocated to DFP–DFO sequential treatment or DFP alone (Fig 1). None of the participants were lost to follow‐up

Study duration: 5 year follow‐up

Participants

Baseline characteristics

DFP/DFO

  • Total # of participants: 105

  • Age: mean (SD): 23 (8.0)

  • Sex: N (%): F: 55 (50.9)

  • Thalassaemia genotype (%): thalassaemia major (100%)

  • Baseline ferritin levels (ng/mL): mean (SD): 1727 (669)

  • Previous iron chelation: N = 105

  • Duration of any iron chelation: not stated

  • LIC (mg/g): mean SD: 4.6 (2.8)

  • Splenectomy: N (%): 17 (14.0)

  • QoL mean (SD): not reported

  • Hb, g/L: mean SD: 99 (10)

DFP

  • Total # of participants: N = 108

  • Age: mean SD: 23 (7.8)

  • Sex: N (%): F: 66 (61.1)

  • Thalassaemia genotype (%): thalassaemia major (100%)

  • Baseline ferritin levels (ng/mL): mean (SD): 1868 (845)

  • Previous iron chelation: N = 108

  • Duration of any iron chelation: not stated

  • LIC (mg/g): mean (SD): 4.0 (2.3)

  • Splenectomy: N (%): 15 (12.7)

  • QoL mean (SD): not reported

  • Hb, g/L: mean (SD): 98 (10)

Inclusion criteria: thalassaemia major, SF between 800 and 3000 ug/L over 13 years of age

Exclusion criteria: known intolerance treatment, platelet count 100 x 109/l or leucocyte count 3.0 x 109/l, severe liver damage, heart failure

Interventions

DFP/DFO

  • DFP 75 mg/kg, divided into 3 oral daily doses, for 4 days/week and DFO subcutaneous infusion (8–12 hours) at 50 mg/kg per day for the remaining 3 days/week

DFP

  • DFP alone, at the same dosage (75 mg/kg divided into 3 oral daily doses), administered 7 days a week

Outcomes

Adherence

Compliance was assessed by counting the pills in each returned bag of DFP and by assessing the number of infusions of DFO registered on the electronic pump

Trial‐reported outcomes

1. Difference between multiple observations of SF concentrations during the 5‐year treatment. A correlation between LIC and SF levels has previously been shown in cohort of people with thalassaemia major treated with DFP (Olivieri et al, 1995).

2. Survival analysis

3. AEs

4. Costs

5. Multislice‐multiecho T2* MRI scan, available since June 2004, was used in a subgroup of participants to evaluate variations in the iron content of the heart and liver during the trial

Identification

Sponsorship source: Italian Society for the Study of Thalassaemia and Haemoglobinopathies (SoSTE)

Country: Italy

Setting: 25 SoSTE centres in Italy

Comments: NCT 00733811

Authors name: Aurelio Maggio

Institution: A.O.V. Cervello, U.O.C. di Ematologia

Email: [email protected]

Address: A.O.V. Cervello, U.O.C. di Ematologia II,Cervello’’, Palermo, Italy

Notes

Follow‐up was planned for 5 years; however, because of the beneficial effects, in terms of SF levels reduction in the sequential DFP–DFO group, observed after the interim analysis performed at 31 January 2008 the trial was stopped before the planned 5 years of treatment were completed for all participants years but mean (SD) duration of treatment was 2.5 (2.2) and 2.9 ( 2.1) years for DFP and sequential DFP–DFO groups, respectively

Sample‐size calculation reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomization sequence was based on a computer‐randomized list in permuted blocks of 10 with a 1:1 ratio,"

Judgement comment: the randomization sequence was based on a computer‐randomized list in permuted blocks of 10 with a 1:1 ratio. The sequence was concealed until interventions were assigned. Randomization was performed per each consecutive participant after verification of the exclusion criteria

Allocation concealment (selection bias)

Low risk

Quote: "To ensure allocation concealment, treatment was assigned by telephone contact from the coordinating centre"

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Trial was open‐label

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Low risk

Quote: "All outcome assessments were done under code by physicians blinded to the trial treatment."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The statistical analysis was based on the 'intention‐to‐treat' principle. None of the participants were lost to follow‐up. However, SF measurements were only complete for all participants in the first year of the trial and decrease substantially thereafter to n = 32 in the combined group and n = 26 in the DFP group

Selective reporting (reporting bias)

Low risk

All outcomes reported

Other bias

Unclear risk

"Only 21 (35%) subjects in the DFP‐alone and 12 (24%) in the sequential DFP–DFO group withdrew definitely from the trial (Table V). The mean time for definitive withdrawal was 152 ± 103 (days) in DFP‐alone versus 112 ± 76 (days) in the sequential DFP–DFO group respectively." "The planned duration of treatment was 5 years. However, because of the beneficial effects, in terms of serum ferritin levels reduction in the sequential DFP–DFO group, observed after the interim analysis performed at January 31, 2008 the trial was stopped before the planned 5 years of treatment were completed for all patients. Therefore, the mean duration of treatment was 2.5 ± 2.2 and 2.9 ± 2.1 years for DFP and sequential DFP–DFO group respectively"

Judgement comment: withdrawal rate is high and the trial stopped early

Mourad 2003

Methods

2‐arm parallel RCT.
Number of centres: 1.
Trial dates: not stated.
Duration of treatment: 1 year.
Follow‐up: none.

Trial undertaken: Chronic Care Centre, Beirut, Lebanon.

Participants

Number randomised: 25 (treatment group: 14; comparator group: 11)
Number analysed: 25 (treatment group: 14; comparator group: 11)

β‐thalassaemia participants, severely iron overloaded and previously poorly chelated
Age range: 12 ‐ 40 years
Sex: treatment: 43% male, comparator: 64% male
Ethnicity: not stated

Interventions

DFO

  • DFO by subcutaneous injection, 40 ‐ 50 mg/kg 8 ‐ 12 hours a day, 5 ‐ 7 days/week

DFP/DFO

  • DFP 75 mg/kg/day orally in 3 divided doses, 7 days a week, DFO by subcutaneous injection, daily dose of 2 g over 8 ‐ 12 hours, 2 days a week

Outcomes

Adherence see compliance below

Trial‐reported outcomes

1. Mean serum iron concentration at baseline, 6 & 12 months (primary outcome)
2. Number RBC units during the trial
3. Iron excretion at 1 & 12 months
4. Hb level measured weekly for 3 months then monthly for 9 months
5. Liver function measured weekly for 3 months then monthly for 9 months
6. Renal function measured weekly for 3 months then monthly for 9 months
7. Side effects
8. Participant compliance: compliance was assessed by the number of vials of DFX or tablets of DFP used. Safety was determined by detailed clinical and laboratory examination. Participants were also asked to complete questionnaires about any side‐effects they experienced.

Identification

Source of funding: not stated.

Notes

Prior exposure to iron chelators: DFO, less than 4 times a week, dose and duration not reported.

Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The authors did not report any information about how randomisation was undertaken

Allocation concealment (selection bias)

Unclear risk

The authors did not report any information about how treatment allocation was concealed

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

Unclear risk

The authors did not report any information as to whether participants, personnel were blinded to treatment allocation

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

The authors did not report any information as to whether outcome assessors were blinded to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis for all outcomes: there were no missing outcome data

Selective reporting (reporting bias)

High risk

Data for 2 pre‐specified outcomes were not reported in the paper: iron excretion at 1 and 12 months and renal function. Both are important clinical markers of the efficacy of iron chelation therapy

Other bias

Low risk

The trial appears to be free of other sources of bias

Olivieri 1997

Methods

2‐arm parallel RCT
Number of centres: 2
Trial dates: November 1993 ‐ September 1995
Duration of treatment: analysis undertaken after 24 months (mean (SD) duration 33 (1.0) months, range 24 ‐ 43 months)
Follow‐up: none

Trial undertaken: Hospital Centres in Toronto and Montreal, Canada. These data are from the Toronto participants only

Participants

Baseline characteristics

Number randomised: 64 (DFO: 32; DFP: 32)
Number analysed: 37 (DFO: 18; DFP: 19). The trial reports details for why 6 and 7 participants respectively were not included in the analysis. The remaining participants had not completed 24 months treatment at the time of analysis for this trial report

DFP (L1)

  • Age: not reported

  • Sex: F: 11; M: 14

  • Thalassaemia genotype (%): thalassaemia major: 100%

  • Baseline ferritin levels (ng/mL) mean (SD): 2194 (1251)

  • Previous iron chelation: not reported

  • Duration of any iron chelation (duration of treatment in this trial ‐ mean (SD) months): 11.0 (4.2) range 2 ‐ 15

  • LIC (mg/g): 9.56 (4.77) Range 2.7 ‐ 21.7

  • Splenectomy n (%): not reported

  • QoL mean (SD): not reported

  • Hb, g/L: not reported

DFO

  • Age: not reported

  • Sex: F: 11 M: 14

  • Thalassaemia genotype (%): thalassaemia major: 100%

  • Baseline ferritin levels (ng/mL) mean (SD): 2089 (048)

  • Previous iron chelation: Not reported

  • Duration of any iron chelation (duration of treatment in this trial ‐ mean (SD) months): 11.63 (3.26), range 2 ‐ 15 months

  • LIC (mg/g): 7.43 (3.59), range 2.4 ‐ 15.7

  • Splenectomy n (%): not reported

  • QoL mean (SD): not reported

  • Hb, g/L: not reported

Inclusion criteria: diagnosed with homozygous β‐thalassaemia, 10 years of age or older, willing to participate in the trial

Exclusion criteria:

  • refùsal to participate in the screening

  • previously treated with DFP

  • serious adverse reactions to DFO

  • failed to attend 20% of the visits in the first 3 months of the trial

  • receiving other investigational drugs

  • past history of malignancy

  • medical, psychological or psychiatric risk

  • therapy with an investigational drug would be unwise

  • were pregnant or breast feeding

  • not using a reliable birth control method

Pre‐treatment:

  • stratified into high (7 mg Fe/g dry weight liver tissue) and low iron‐overloaded (7 mg Fe/g dw) according to their hepatic iron concentration as assessed either by liver biopsy or a SQUID (or both)

  • 8 participants have been withdrawn from the study due to AEs (2), family reasons (1), psychiatric disorder (1), chronic neutropenia prior to starting on DFP (2), bone marrow transplantation (1) and non compliance with the trial protocol (1)

  • 25 participants on DFP and 26 participants on DFO have been used in the present analysis.

  • Author goes on to report that results of n = 5 in DFO were not evaluated as there was no compliance data. A further n = 5 participants on DFP and n = 2 were excluded for the analysis of the correlation between compliance + successful outcome (as measured by LIC) as there were 6 months of data available. Therefore, for the main outcome the actual N = 39 (n = 20 in DFP and n = 19 in DFO

Interventions

DFP (L1)

  • DFP 75 mg/kg/day in 3 divided doses

DFO

  • DFO 50 mg/kg/night, 4 ‐ 7 night/week

Outcomes

Adherence see adherence below

Trial‐reported outcomes

1. Change in LIC (measured by SQUID or biopsy) between 12 months prior to randomisation & 24 months duration on trial treatment

2. Adherence to iron chelation therapy rates defined as per cent of doses administered (number of doses of the iron chelator taken, out of number prescribed), measured for a minimum of 3 months

Identification

Sponsorship source: no sponsorship stated

Country: Canada

Setting: Transfusion Clinic

Authors name: Nancy Olivieri

Institution: University of Toronto

Source of funding: not stated

Notes

Prior exposure to iron chelators: not reported
Abstract publication. Some data from Pope 1995 thesis included for baseline characteristics

Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quote: "After stratification patients by LIC (>7mg Fe/g; < 7mg Fe/g) 'patients were assigned by a research pharmacist who did not know the patients"

Allocation concealment (selection bias)

Unclear risk

The authors did not report any information about how treatment allocation was concealed

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

1 treatment a pump and 1 treatment a tablet, participants and researchers would not be blinded to treatment

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

The authors did not report any information as to whether outcome assessors were blinded to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

High risk

The trial analysed data from 58% of randomised participants. Of the 42% randomised participants who were not available for outcome analysis:
• 22% randomised participants had not completed the required 24 months treatment at the time of analysis for this trial report;
• 16% DFP‐treated participants and 5% DFO treated participants were withdrawn due to treatment induced side effects

This missing data may inappropriately affect the statistical findings of the trial

Selective reporting (reporting bias)

Low risk

All outcomes pre‐specified were reported in the manuscript

Other bias

Unclear risk

The trial was reported in an abstract, thus there are few data available to make an assessment of whether the trial was free of other bias. Trial stopped early by manufacturer

Pennell 2006

Methods

2‐arm parallel RCT
Number of centres: 4
Trial dates: December 2002 ‐ March 2005
Duration of treatment: 1 year
Follow‐up: outcome data recorded for duration of treatment

Trial undertaken: 4 participating centres in Italy and Greece

Participants

Number randomised: 61 DFO: 32; DFP: 29
Number analysed: variable across outcomes. Minimum and maximum numbers analysed were: treatment group: 30 ‐ 32; comparator group: 27 ‐ 29. Trial reported details as to why data from 1 participant in the treatment group and 2 in the comparator group were withdrawn from treatment

Transfusion‐dependent homozygous participants with β‐thalassaemia major
Age: mean (SD) treatment group: 26.2 (4.7) years; mean (SD) comparator group: 25.1 (5.8) years
Sex: treatment group: 50% male; comparator group: 52% male
Ethnicity: Greek/Italian: treatment group: 18/14; comparator group: 16/13

Interventions

DFO

  • DFO by subcutaneous injection, 50 mg/kg for 5 or more days a week

DFP

  • DFP initial dose 75 mg/kg/day increasing to 100 mg/kg/day. Mean actual dose: 92 mg/kg/day

Outcomes

Adherence rates: DFP compliance was measured using the Medication Event Monitoring System device (Aardex, Zug, Switzerland) and calculated as the percent of openings with an interval longer than 4 hours recorded, divided by number of doses prescribed. DFO compliance was calculated as the percentage of completed infusions, as determined by the Crono pumps, divided by the number of infusions prescribed.

Trial‐reported outcomes

1. Change over 1 year in myocardial T2* (primary outcome)
2. Cardiac volumes and function
3. LIC
4. SF
5. ANC
6. AEs
7. ALT
8. Serum zinc levels
9. Serum creatinine levels

Identification

Trial sponsor: Apotex (manufacturer of DFP)

Notes

Prior exposure to iron chelators: DFO at a mean (SD) dose of 39 (8) mg/kg/day for 5 ‐ 7 days/week

Sample‐size calculation reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The authors did not report any information about how randomisation was undertaken

Allocation concealment (selection bias)

Unclear risk

The authors did not report any information about whether treatment allocation was concealed

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Open label one treatment subcutaneous and the other oral so not possible to mask treatments

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Low risk

The primary outcome was independently measured in a different country (UK) to where the trial took place and the findings were not communicated back to the clinicians during the course of the trial

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants were included in the analysis of the outcomes SF and AEs
Data from 1 participant in the treatment (DFO) group were not included in the analysis of the cardiac outcomes (primary outcome) and last observation carried forward method was used to accommodate the missing data from 3 other participants (1 treatment group and 2 from the comparator group) in the cardiac outcomes (primary outcome)
2 participants in each treatment group did not have a LIC assessment at 12 months and the data from these participants were missing from the analysis

Selective reporting (reporting bias)

High risk

The following pre‐specified outcomes were not reported in the manuscript: ANC; ALT; serum zinc levels; and serum creatinine levels

Other bias

High risk

There are several imbalances in baseline characteristics between the 2 interventions including a major imbalance in SF measures with the DFO group having much higher levels as well as a greater proportion of participants with severe iron overload (above 2500 µg/L)

Pennell 2014

Methods

Study design: RCT

Study grouping: parallel group

CORDELIA was a prospective, multinational, randomised, open‐label, parallel‐group, phase 2 trial. A total of 81.2% of participants (n = 160) completed 1 year of treatment

Participants

"Overall, 925 patients were screened and 197 randomized. The majority of patients screened were β‐thalassemia major patients (902/925; 99.1%). Other patients who were screened and for whom underlying anaemia was captured had low/intermediate 1 myelodysplastic syndrome (n = 4), Diamond–Blackfan anaemia, β‐thalassemia intermedia, congenital dyserythropoietic anaemia, and paroxysmal nocturnal haemoglobinuria (all n = 1). Only β‐thalassemia major patients fulfilled the inclusion criteria and were enrolled in the study. A total of 81.2% of patients (n = 160) completed 1 year of treatment"

Baseline characteristics

DFX (Exjade)

  • Total # of participants: 98

  • Age mean (SD): 19.9 (6.5)

  • Sex (M:F ratio n): 58:40

  • Thalassaemia genotype (%): thalassaemia major: 100%

  • Previous iron chelation: DFO: 41 (42.7); DFP: 9 (9.4); DFO + DFP: 21 (21.9); DFX: 18.1(8.8); Unknown or irregular: 7(7.3)

  • Duration of any iron chelation mean (SD) years: 14.0 (7.0)

  • LIC (mg Fe/g dw): < 7: 11 (12.1); 7 to < 15: 14 (15.4); ≥15: 66 (72.5)

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Median SF (range), ng/mL (per protocol population): 5062 (613 ‐ 15331)

DFO (Desferal)

  • Total # of participants: 99

  • Age mean (SD): 19.7 (6.3)

  • Sex (M:F ratio n): 57:42

  • Thalassaemia genotype (%): thalassaemia major: 100%

  • Previous iron chelation: DFO: 39 (42.9); DFP: 5 (5.5); DFO + DFP: 21 9 (23.1); DFX: 23 (25.3); Unknown or irregular: 3 (3.3)

  • Duration of any iron chelation mean (SD) years: 14.3 (7.2)

  • LIC (mg Fe/g dw): 7: 8 (9.9); 7 to 15: 14 (17.3); ≥15: 59 (72.8)

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

  • Median SF (range), ng/mL (per protocol population): 4684 (677 ‐ 13342)

Inclusion criteria: people with β‐thalassemia major, Diamond–Blackfan anaemia, low/intermediate myelodysplastic syndromes, or sideroblastic anaemia, aged ≥ 10 years with myocardial T2* 6 ‐ 20 ms, LVEF ≥ 56%, R2 MRI LIC ≥ 3 mg Fe/g dw, lifetime history of ≥ 50 units RBC transfusions, and receiving ≥10 unit/year of RBC transfusions

Exclusion criteria: participants with serum creatinine above the ULN or significant proteinuria (urinary protein/creatinine ratio ≥1.0 mg/mg in a non–first‐void urine sample at baseline; people with ALT 5 x the ULN only if their LIC was 10 mg Fe/g dw; considerable impaired GI function or GI disease; history of clinically relevant ocular and/or auditory toxicity related to iron chelation; therapy, and history of HIV seropositivity or malignancy within the past 5 years; clinical symptoms of cardiac dysfunction (shortness of breath at rest or exertion, orthopnoea, exercise intolerance, lower‐extremity edema, arrhythmias)

Interventions

DFX (Exjade)

  • Once‐daily DFX 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 (Desferal)

  • An intensified dosing regimen of DFO was administered at 50 to 60 mg/kg per day via subcutaneous infusion over 8 ‐ 12 hours, 5 ‐ 7 days a week, in accordance with Thalassaemia International Federation Guidelines

Mean actual dose over 1‐year treatment was 36.7 6 4.2 mg/kg per day DFX (range, 19.7‐ 43.3 mg/kg per day). Mean actual dose of DFO was 41.5 6 8.7 (13.2 ‐ 60.2) mg/kg per day, when normalized to a 7‐day regimen

Outcomes

Adherence to iron chelation therapy rates: not stated how adherence was measured

Trial‐reported outcomes

1. Ratio of Gmean myocardial T2* after 1 year of treatment with DFX divided by the ratio of Gmean for DFO
2. Change in LVEF after 1 year of treatment, assessed by absolute change from baseline CMR

3. Absolute change from baseline in LIC after 1‐year treatment

4. Absolute change from baseline in SF after 1‐year treatment

Identification

Sponsorship source: Novartis Pharma AG

Country: multinational, 11 countries

Setting: 22 centres across 11 countries

Comments: the authors thank Debbi Gorman of Mudskipper Business Ltd for medical editorial assistance. Financial support for medical editorial assistance was provided by Novartis Pharmaceuticals

Authors name: Dudley J. Pennell

Institution: National Institute for Health, Research Cardiovascular Biomedical Research Unit

Email: [email protected]

Address: National Institute for Health Research Cardiovascular Biomedical Research Unit, Royal Brompton Hospital, Sydney Street, London, SW3 6NP, UK

Notes

Novartis Pharmaceuticals Corporation (East Hanover, NJ, USA) co‐ordinated the design and execution of this trial and contributed to the analysis and interpretation of the trial data. Novartis Pharmaceuticals Corporation also collaborated with the external authors to assist in the development and approval of the manuscript for publication

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "22 centers across 11 countries. Following a 35‐day screening phase, patients were randomized in a 1:1 ratio" Randomisation was based on permuted blocks; stratification by centre was not conducted

Allocation concealment (selection bias)

Unclear risk

Judgement comment: no description of allocation concealment except that randomisation was based on permuted blocks

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment: open‐label trial ‐ subcutaneous pump versus oral tablet ‐ difficult to blind

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Low risk

Quote: "Core laboratories were blinded to treatment allocation.In order to eliminate potential unrecognized biases, the core clinical trial team was blinded to the treatment assignment prior to the database lock for the primary analysis."

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Judgement comment: 21 withdrawn DFO arm 16 in DFX (78 to 82 completed trial) Efficacy outcomes reported in per protocol and safety in the participants who received the trial drug

Selective reporting (reporting bias)

Unclear risk

Investigator‐reported AEs, regardless of causality, were reported in 65 (67.7%) DFX participants and 69 (75.8%) DFO participants (supplemental Table 2). AEs suspected to be related to trial drug occurred in 35.4% of DFX participants and 30.8% of DFO participants

Judgement comments: It is unclear if investigator‐reported AEs and those suspected to be related to trial drug include the same AEs. Also, they only report the end of trial LIC value for the DFX group

Other bias

Low risk

The trial appears to be free of other sources of bias

Taher 2017

Methods

Study design: multicentre RCT conducted in several countries

Study grouping: parallel group

Study duration: 24 weeks

Participants

Baseline characteristics

DFX film‐coated tablet

  • Total # of participants: N = 87

  • Age: 34.6 (19.97)

  • Sex: F: 41

  • Thalassaemia genotype N (%): thalassaemia major: 70 (80.5)

  • Previous iron chelation: 79 (90.8)

  • Median SF (range), ng/mL: 2983 (939 – 8250)

  • Splenectomy n (%): not reported

  • QoL mean (SD): not reported

  • Hb, g/L: not reported

DFX dispersible tablet

  • Total # of participants: N = 86

  • Age: 35.1 (18.60)

  • Sex: F: 47

  • Thalassaemia genotype N (%): thalassaemia major: 70 (81.4)

  • Baseline ferritin levels (ng/mL) mean (SD): 2089 (048)

  • Previous iron chelation: 77 (89.5)

  • Median SF (range), ng/mL: 2485 (915 – 8250)

  • Splenectomy n (%): not reported

  • QoL mean (SD): not reported

  • Hb, g/L: not reported

Inclusion criteria:

  • Males and females aged ≥ 10 years

  • Transfusion‐dependent thalassaemia and iron overload, requiring DFX dispersible tablet at doses of ≥ 30 mg/kg/day as per the investigator's decision or participants with very low, low or intermediate (int) risk myelodysplastic syndrome and iron overload, requiring DFX dispersible tablet 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

  • SF > 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) > 5 x ULN, unless LIC confirmed as >10 mg Fe/dw 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

  • Participants with significant impaired GI function or GI disease that may significantly alter the absorption of oral DFX (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

DFX film‐coated tablets

  • DFX film‐coated provided as 90 mg, 180 mg and 360 mg film‐coated tablets for oral use

DFX dispersible tablet

  • DFX dispersible tablet provided as 125 mg, 250 mg and 500 mg dispersible tablets for oral use

Outcomes

Adherence to iron chelation therapy rates

Compliance with medication as assessed by relative consumed tablet count

Trial‐reported outcomes

1. Overall safety of both DFX formulations, measured by frequency and severity of AEs and changes in laboratory values from baseline to 24 weeks.

2. Evaluation of both formulations on selected GI AEs (diarrhoea, constipation, nausea, vomiting, and abdominal pain) during treatment

3. Estimation of treatment compliance

4. Evaluation of both formulations on participant satisfaction, palatability, and GI symptoms using PROs

5. Evaluation of the pharmacokinetics of both formulations

6. Reported % compliant with upper and lower percentages

Identification

Sponsorship source: Novartis Pharmaceuticals

Country: USA

Comments: NCT02125877

Authors name: Ali Taher

Institution: American University of Beirut Medical Center

Email: [email protected]

Address: Haematology and Oncology, Department of Internal Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon

Notes

Sample‐size calculation not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomization was stratified by underlying disease and previous chelation treatment."

No clear description of randomisation or if participants were randomised centrally

Allocation concealment (selection bias)

High risk

Quote: "Post‐ hoc analyses identified that 23 patients on FCT (26%) were started on a dose that was higher than recommended in the protocol compared with 8 patients (9.3%) on DT (not recognized or reported by the investigators as dosing error)."

Judgement comment: the trial was open label and most participants had been on 1 or the other of the trial drugs prior to the trial ‐ doses may have corresponded to prior dosing since there was no description of allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment: open‐label

Blinding of outcome assessment (detection bias)
All outcomes except mortality

High risk

No description of how outcome assessment was performed ‐ centrally or blinded open‐label trial

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Quote: "Overall, all patients were satisfied with their medicine during the study period; satisfaction scores were higher with deferasirox FCT compared with DT at all visits."

Judgement comment: no data provided on number of participants or scores just general statements

Selective reporting (reporting bias)

High risk

Quote: "patients discontinued treatment because of AEs (n = 10), protocol deviation (n = 5), withdrawal of consent (n = 3), patient guardian decision (n = 2), and other reasons (administrative problems, death, and physician’s decision, n = 1 each)."

Judgement comment: investigators do not report all outcomes by treatment assignment, and AEs and SAEs are reported as suspected relationship to trial drug and occurring in > or equal to 10%

Other bias

Unclear risk

"The absolute reduction in median serum ferritin (range) in patients receiving FCT was –350 (–4440–3572) ng/mL and in those receiving DT was –85.5 (–2146–8250) ng/mL); these correspond to a relative change of –14.0% with FCT and –4.1% with DT."

Judgement comment: some of difference in change could be accounted for more participants starting on a higher dose of film‐coated tablet

Tanner 2007

Methods

2‐arm parallel RCT
Number of centres: multicentre (12 centres)
Duration of treatment: 12 months
Follow‐up: not stated

Trial undertaken: thalassaemia out‐patient clinics in Sardinia

Participants

Number randomised: 65 (treatment group: 33; comparator group: 32)
Number analysed: not reported

Number completing treatment: 60 (treatment group: 32; comparator group: 28). The reason for the withdrawal was not fully reported by the trial authors

Participants aged 18 years or older with a diagnosis of β‐thalassaemia, currently maintained on subcutaneous DFO and with a myocardial T2* between 8 ‐ 20 ms
Age: treatment group: mean (SD) 28.7 (5.3) years; comparator group: mean (SD) 28.8 (4.2) years Age range for both arms was 18 ‐ 42 years
Sex: treatment group: 39% male; comparator group: 44% male
Ethnicity: not stated

Interventions

DFO

  • DFO 40 ‐ 50 mg/kg subcutaneously for 5 days a week (DFO actual dose: 43.4 mg/kg for 5 days) with an oral placebo (no further details reported)

DFO/DFP

  • DFO 40 ‐ 50 mg/kg subcutaneously for 5 days a week (DFO actual dose: 34.9 mg/kg for 5 days) with DFP 75 mg/kg daily for 7 days a week

Outcomes

Adherence see compliance below

Trial‐reported outcomes

1. Change over 1 year in myocardial T2* (primary outcome)
2. Change in liver T2* at 12 months
3. SF
4. Left ventricular volume & function
5. Brachial artery reactivity as a marker of heart failure
6. Participant compliance with chelation treatments: DFO compliance was calculated as the percentage of completed infusions, as determined by the Crono pumps, divided by the number of infusions prescribed. DFP/placebo compliance was measured through pill counting at the bi‐monthly visits
7. AEs
8. BNP test

Identification

Source of funding: CORDA, Royal Brompton & Harefield Hospitals Charitable funds, Cooley’s Anemia Foundation, Apotex, UK Thalassaemia Society, University College London Special trustees Chairty

Notes

Prior exposure to iron chelation: DFO mean (SD) dose 36.4 (11.1) mg/kg per day for 5.5 day/week (equivalent to 40.5 mg/kg for 5 day/week). Participants were excluded if they had previously received DFP

Sample‐size calculation reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The authors did not report any information about how randomisation was undertaken

Allocation concealment (selection bias)

High risk

Trial reports that the participants and clinicians were aware of how treatment was to be allocated

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

Unclear risk

The authors did not report any information as to whether participants or personnel were blinded to treatment allocation

Blinding of outcome assessment (detection bias)
All outcomes except mortality

Unclear risk

The authors did not report any information as to whether outcome assessors were blinded to treatment allocation

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

As the trial does not report the number of participants included in each outcome assessment. The trial reports the number completing treatment and the reasons why 3 participants in the treatment group (1 adverse event & 2 participant requests) and 4 participants in the comparator group (3 adverse events & 1 participant request) were withdrawn from the trial

Selective reporting (reporting bias)

Low risk

All outcomes pre‐specified were reported in the manuscript

Other bias

Low risk

The trial appears to be free of other sources of bias

Vichinsky 2007

Methods

Study design: RCT

Study grouping: parallel group

The study duration was 52 weeks. Participants were recruited by investigators at 44 sites in the USA, France, Italy, UK and Canada

Participants

Baseline characteristics

DFX

  • Total # of participants: 132

  • Age: 15 range 3 ‐ 54

  • Sex (female %): 60.6

  • Sickle cell genotype N (%): 100

  • Baseline ferritin levels (ng/mL) median (min ‐ max): 3460 (1082 ‐ 1201)

  • Previous iron chelation %: 62.9

  • Splenectomy n (%): not reported

  • QoL mean (SD): not reported

DFO

  • Total # of participants: 63

  • Age: 16. Range 3 ‐ 51

  • Sex (female %): 55.6

  • Sickle cell genotype N (%): 100

  • Baseline ferritin levels (ng/mL) median (min ‐ max): 2834 (1015 ‐ 15578)

  • Previous iron chelation %: 60.3

  • Splenectomy n (%): not reported

  • QoL (mean (SD)): not reported

Age group (% DFX, DFO)
< 6 years: 3.0, 4.8
6 to < 12 years: 22.7, 23.8
12 to <16 years: 25.0, 20.6
16 to < 50 years: 47.7, 49.2
50 to < 65 years: 1.5, 1.6

Inclusion criteria:

  • People with SCD ≥ to 2 years of age and with iron overload from repeated blood transfusions

  • People receiving regular blood transfusions or those sporadically transfused who received at least 20 units of packed RBCs or equivalent were eligible

  • Prior chelation therapy was permitted but was not mandatory

  • The serum ferritin level for entry into the screening period of this study was ≥ 1000 µg/L

Exclusion criteria

  • People were excluded if they had a serum creatinine above the ULN

  • Significant proteinuria (as indicated by a urinary protein:creatinine ratio of ≥ 0.5 confirmed at 2 visits)

  • Active hepatitis B or C

  • Second and third atrioventricular block, QT interval prolongation, or therapy with digoxin or similar medications

  • Treatment with beta blockers or angiotensin‐converting enzyme inhibitors was permitted. Those with chelation therapy‐associated ocular toxicity were excluded

Interventions

DFX

  • The initial 24 participants enrolled were randomised to receive DFX 10 mg/kg, all subsequent participants randomised to DFX were dosed at 10 – 30 mg/kg according to baseline LIC. DFX was given once daily each morning as a dispersed solution in water, half‐an‐hour before breakfast. The dose of DFX was reduced by 1 dose level and not re‐escalated for participants 15 years and older if serum creatinine increased 33% above baseline on two consecutive occasions. For children less than 15 years of age, the dose was only decreased if these values were also above the age‐appropriate ULN. DFX was interrupted for moderate or severe skin rash and re‐instituted at half the initial dose, and dose re‐escalation was permitted

DFO

  • DFO was administered as a slow subcutaneous infusion over 8 – 12 hours using electronic Microject Chrono infusion pumps on 5 – 7 days a week. In order to facilitate the comparison of different schedules, all DFO doses reported were normalised to administration for 5 days/week (i.e. 50 mg/kg administered 7 days/week would be reported as 70 mg/kg)

Outcomes

Adherence to iron chelation therapy rates

Compliance. For DFX, compliance was assessed by counting the number of tablets returned in bottles at each visit. For DFO, the numbers of vials returned at each visit were counted

Trial‐reported outcomes

1. Safety assessments

2. Laboratory assessments were performed at least monthly and included complete blood counts with differential counts. Biochemistry testing included electrolytes, glucose, liver function tests, gamma‐glutaryl‐transferase, lactate dehydrogenase, cholesterol, triglycerides, uric acid, total protein, C‐reactive protein, copper and zinc levels. Iron parameters included total iron, transferrin, transferrin saturation and ferritin. Urinary testing performed on random collections included determination of creatinine, total protein and albumin

3. Physical examinations, ECGs, audiometry and ophthalmological tests were performed at baseline, 12, 24, 36 and 52 weeks. In participants less than 16 years of age, additional assessments included growth velocity and pubertal stage

4. Efficacy assessments. LIC was determined by SQUID biospectrometry at baseline, 24 and 52 weeks. The 24‐week assessment was performed primarily for safety purposes, and the change in LIC was calculated between baseline and 52 weeks. SF was assessed monthly during the trial and the change was determined using the baseline and final ferritin level

Identification

Sponsorship source: Novartis Pharmaceuticals

Country: international (Canada, France, Italy, UK and USA)

Setting: medical centre outpatient

Authors name: Elliott Vichinsky

Institution: Children’s Hospital and Research Center at Oakland,

Email: [email protected]

Address: Children’s Hospital and Research Center at Oakland, 747 52nd Street, Oakland, CA 94609, USA

Novartis Pharmaceuticals Corporation (East Hanover, NJ, USA) co‐ordinated the design and execution of this trial and contributed to the analysis and interpretation of the trial data. Novartis Pharmaceuticals Corporation also collaborated with the external authors to assist in the development and approval of the manuscript for publication

Notes

Sample‐size calculation reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomisation was performed using an interactive voice response system"

Allocation concealment (selection bias)

Unclear risk

Quote: "stratified according to the following age groups: 2 to < 6 years, 6 to < 12 years, 12 to < 16 years and 16 years and older. The randomisation sequence included permuted block groups of six patients for each of the three age strata."

Judgement comment: some of the age groups had few participants and unclear if allocation would remain concealed with permuted block groups of 6 participants

Blinding of participants and personnel (performance bias)
All outcomes except mortality or other objective outcomes

High risk

Judgement comment: no mention of blinding, but DFO is delivered by infusion pumps and DFX is a solution in water, so blinding not feasible

Blinding of outcome assessment (detection bias)
All outcomes except mortality

High risk

Judgement comment: no description of blinding: Novartis Pharmaceuticals Corporation (East Hanover, NJ, USA) co‐ordinated the design and execution of this trial and contributed to the analysis and interpretation of the trial data. The data were analysed under supervision of the trial statistician and were reviewed by the investigators

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All outcomes reported. 8 participants did not complete and were not included. 6 DFX arm withdraw consent, one in DFO arm. 3 DFO non compliant, 2 DFX and 1 DFO lost to follow‐up

Selective reporting (reporting bias)

Unclear risk

Quote: "Adverse events, irrespective of the relationship to study medication, which occurred in more than 10% of patients receiving either treatment, are shown in Table III. As arbitrarily defined by an increased frequency of at least 5% indicating a potential relationship to drug administration."

Judgement comment: do not report the total number of AEs in all participants, as well there was a substantial number of participants experience SAEs and there is no list of the type except for pain crisis: The number of participants receiving DFX and DFO that reported SAEs was similar (46.2% and 42.9% respectively) and the most common SAE in both groups was sickle cell anaemia with crisis (33.3% and 31.7% respectively). Also table of AEs report % and no totals so impossible to determine total number of participants with an AE

Other bias

Unclear risk

Quote: "The reasons for withdrawal of consent were not included in the database."

Quote: "The initial 24 patients enrolled were randomised to receive deferasirox 10 mg/kg or deferoxamine at recommended doses of 20–60 mg/kg based on initial LIC. Subsequently, additional safety information became available for deferasirox suggesting a need to modify the starting dose (Cappellini et al, 2006). Therefore, following the enrolment of the first 24 patients, the study was amended so that all subsequent patients randomised to deferasirox were dosed at 10–30 mg/kg according to baseline LIC"

Judgement comment: it is important to understand reasons for withdrawals and also the nature of the missing safety information which may have implications for dosing and effects of the dosing amendment

ADRs: adverse drug reactions
AEs: adverse events
ALT: alanine aminotransferase
ANC: absolute neutrophil count
BNP: brain natriuretic peptide
CBC: complete blood count
CMR: cardiovascular magnetic resonance imaging
DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
dw: dry weight
ECGs: electrocardiograms
FBC: full blood count
Hb: haemoglobin
HRQoL: health‐related quality of life
ICT: iron chelation therapies
IQR: interquartile range
LVEF: left ventricular ejection fraction
LIC: liver iron concentration
MRI: magnetic resonance imaging
PK: pharmacokinetic
PRBC: packed red blood cell
QoL: quality of life
RBCs: red blood cells
RCT: randomised controlled trial
SAEs: serious adverse events
SCr: sickle cell retinopathy
SD: standard deviation
SF: serum ferritin
SGPT: serum glutamate‐pyruvate transaminase
SQUID: Superconducting Quantum Interference Device
UIE: urinary iron excretion
ULN: upper limit of normal
WBC: white blood count

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abu 2015

Wrong study design ‐ qualitative interview questionnaire used.

Al Kloub 2014

Wrong study design ‐ qualitative interview questionnaire used.

Al Kloub 2014a

Wrong study design ‐ cross‐sectional study.

Al Refaie 1995

Wrong study design ‐ medication study ‐ not an RCT.

Alvarez 2009

Wrong study design ‐ medication study ‐ not an RCT.

Armstrong 2011

No intervention.

Bala 2014

No intervention.

Belgrave 1989

No intervention.

Berkovitch 1995

Not designed to measure adherence to iron chelation therapy.

Chakrabarti 2013

Not designed to measure adherence to iron chelation therapy.

Daar 2010

Wrong setting ‐ single‐centre study.

Gomber 2004

No intervention.

Kidson Gerber 2008

Wrong study design ‐ clinical audit of medication use.

Kolnagou 2008

Wrong study design ‐ medication study not RCT.

Leonard 2014

Wrong study design ‐ single‐treatment study.

Loiselle 2016

Review.

Mazzone 2009

Wrong comparator ‐ healthy children not taking iron chelation therapy.

NCT01709032

Not designed to measure adherence to iron chelation therapy.

NCT01825512

Not designed to measure adherence to iron chelation therapy.

NCT02133560

Wrong study design ‐ single‐centre study with no control.

NCT02466555

Wrong study design ‐ single‐centre study with no control.

Pakbaz 2004

Wrong study design ‐ single‐centre study with no control.

Pakbaz 2005

Wrong study design ‐ single‐centre study with no control.

Porter 2009

Wrong study design ‐ medication intervention not a RCT.

Porter 2012

Wrong study design ‐ medication intervention not a RCT.

Vichinsky 2005

Not designed to measure adherence to iron chelation therapy.

Vichinsky 2008

Not designed to measure adherence to iron chelation therapy.

Waheed 2014

Not designed to measure adherence to iron chelation therapy.

Walsh 2014

Review.

Yarali 2006

Not designed to measure adherence to iron chelation therapy.

RCT: randomised controlled trial

Characteristics of studies awaiting assessment [ordered by study ID]

Antmen 2013

Methods

Prospective cohort study; parallel group

Participants

Participants using DFX ‐ we do not know the disease diagnosis and therefore awaiting classification

Exclusion criteria: not stated

Interventions

Educational intervention, standard care (as defined in the study)

Outcomes

Exjade Patient Compliance Program (EX‐PAT) was established to increase patients’ knowledge about DFX usage. This abstract aimed to represent the results of the pilot EX‐PAT program

It is highly recommended to educate the patients under iron chelating treatment about possible complication and usage of chelating agent

Notes

Email sent to author asking for the following information so we could include the study: a full study report of this abstract? If this is not available would it be possible to have more information on: 1. The disease diagnosis of the participants (were they sickle cell (phenotypes) or thalassaemia (phenotypes) or other); 2. How participants were assigned to intervention or control; 3. Any inclusion/exclusion criteria; 4. Any group differences; 5. Is the age range for the whole group or is it for the intervention group only? If so could we have the age range for the control group; 6. Baseline and end of study ferritin levels; 7. SAEs or any AEs

NCT00004982

Methods

RCT; parallel group

Participants

Inclusion criteria: ages eligible for trial: 7 years and older (child, adult, senior); genders eligible for study: both

Exclusion criteria: overt cardiac disease

Interventions

Combination iron chelation therapy, standard care (as defined in the trial)

Outcomes

This small trial is testing the premise that a combination of drugs as a new approach to iron chelation therapy may reduce side effects and increase efficacy. If both drugs can be given orally, there may be a better chance of finding a suitable alternative to Desferal. Several combinations of experimental iron chelating drugs are being used in this trial

Notes

This trial has been completed. Sponsor: National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). No study results posted

NCT00004982: scant information about the trial was documented on the clinicaltrials.gov web site. We have been unable to identify any publications from this trial and despite repeated emails to the trial co‐ordinator and searching the funders web site, we have been unable to identify any further details about the trial. Start date: December 1998; estimated completion November 2002

AEs: adverse events
DFX: deferasirox
RCT: randomised controlled trial
SAEs: serious adverse events

Characteristics of ongoing studies [ordered by study ID]

EudraCT 2012‐000353‐31

Trial name or title

Multicentre, randomised, open‐label, non‐inferiority active‐controlled trial to evaluate the efficacy and safety of DFP compared to DFX in paediatric patients aged from 1 month to less than 18 years of age affected by transfusion‐dependent haemoglobinopathies

Methods

Randomised trial, parallel group

Participants

1. Children on current treatment with DFO or DFX or DFP in a chronic transfusion program receiving at least 150 mL/kg/year of packed RBCs (corresponding approximately to 12 transfusions);
2. For those naive to chelation treatment: participants that have received at least 150 mL/kg of packed RBCs (corresponding to approximately 12 transfusions) in a chronic‐transfusion program and with SF levels ≥ 800 ng/mL;
3. For children aged from 1 month to less than 6 years: known intolerance or contraindication to DFO;
4. Written informed consent and patient's informed assent to child’s maturity and understanding

Interventions

DFP compared to DFX

Outcomes

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

1. LlC as measured by MRI in those able to undergo MRI scan without sedation
2. Safety and tolerability assessments
3. QoL

Starting date

Not stated

Contact information

Consorzio per le Valutazioni Biologiche e Farmacologiche

via Luigi Porta, 14

Pavia 27100 Italy

deep.2@deep‐project.net

Notes

IRCT2015101218603N2

Trial name or title

To assess compliance, efficacy and satisfaction with two different formulation of deferasirox in people with transfusion‐dependent beta‐thalassaemia

Methods

RCT; parallel group

Participants

Inclusion criteria: signing informed consent; male or female aged ≥ 2 years at screening; people with transfusion‐dependent thalassaemia major; regular transfusion indicated by a blood requirement ≥ 8 blood transfusions per year at screening.

Exclusion criteria: people with mean levels of ALT above 5‐fold the ULN; people with serum creatinine above ULN; significant proteinuria as indicated by a urinary protein/creatinine ratio > 0.6 (mg/mg); creatinine clearance ≤ 60 mL/min; chronic hepatitis B infection; active hepatitis C infection; pregnancy or breastfeeding; non‐transfusion dependent thalassaemia

Interventions

DFX (new formulation JadenuTM), DFX (Exjade®)

Outcomes

Participants compliance and satisfaction; 3 months after drug consumption; designed questionnaire to assess participant compliance and satisfaction; ferritin serum amount; safety; possible GI side effects, including diarrhoea, and dermatologic symptoms

Starting date

22 December 2015

Contact information

Vice chancellor of research, Shiaz Univeisity of Medical Sciences

COUNTRY: Iran

SETTING: multicentre (outpatient)

Dr. Sezaneh Haghpanah

INSTITUTION:Hematology Research Center, Nemazee Hospital, Shiraz, Iran

EMAIL: [email protected]

ADDRESS: Dr Sezaneh Haghpan Professor of community medicine Hematology Research Center, Nemazee Hospital, Zand Street, Shiraz, Ira

Notes

Madderom 2016

Trial name or title

A randomised controlled trial studying the effectiveness of group medical appointments on self‐efficacy and adherence in sickle cell disease (TEAM study): study protocol

Methods

RCT; parallel group

Participants

Inclusion criteria: individuals with homozygous or compound heterozygous SCD

Exclusion criteria: individuals with a first visit to the outpatient clinic, patients who cannot communicate adequately due to language difficulties and/or hearing problems or patients who have behavioral problems which will limit group functioning

Interventions

Group Medical Appointment, Individual Medical Appointment (IMA; care‐as‐usual)

Outcomes

Primary and secondary endpoints will be measured at baseline (start of the study), after 1.5 years (after two GMA visits) and after 3 years (after four GMA visits), in both groups. Assessments are performed at the hospital, directly before the outpatient visit and in presence of a psychologist. Primary endpoint: 1. Self‐efficacy as measured by the validated Sickle Cell Self‐ Efficacy Scale; Secondary endpoints; 2. Adherence to prescribed treatment by (paediatric) hematologist; 3. QoL as measured with the validated Pediatric Quality of Life Inventory for children and SF‐36 for adults. 4. Emergency visits and hospital admissions for SCD related symptoms and complications. 5. Satisfaction with treating physician and nurse (by visual analogue scale: score 1 – 10); 6. Measurement of costs and effects in the GMA and IMA group by an economic analysis according to Dutch guidelines and with respect to an increase in self‐ efficacy

Starting date

The trial opened to recruitment in January 2013 for the children and in September 2015 for the adults and is still ongoing.

Contact information

Marjon H. Cnossen

INSTITUTION: Department of Pediatric Hematology, Erasmus University Medical Center ‐ Sophia Children’s Hospital

EMAIL: [email protected]

ADDRESS: Department of Pediatric Hematology, Erasmus University Medical Center ‐ Sophia Children’s Hospital, Wytemaweg 80, PO Box 2060, 3000 CB Rotterdam, The NetherlandsAdditional data

Notes

Trial registration: NTR4750 (NL42182.000.12)

NCT02173951

Trial name or title

An algorithm to start iron chelation in minimally transfused young beta‐thalassaemia major patients

Methods

RCT; parallel group

Participants

Inclusion criteria: young individuals with β‐thalassaemia major (diagnosed by HPLC, CBC) who started transfusion therapy who received 5 ‐ 7 transfusions or less, aged more than 6 months. Pre‐transfusional Hb should be >9 g/dL. Serum ferritin should be ≤ 500 ng/mL, transferrin saturation ≤ 50%.

Exclusion criteria: 1. individuals with β‐thalassaemia intermedia, those with other transfusion‐dependent anemias (myelodysplasia, other chronic haemolytic anemias, pure red cell aplasia, aplastic anaemia); 2. Individuals with levels of ALT > 5 the ULN, serum creatinine > ULN on 2 measurements; 3. Indiviudals with history of agranulocytosis (ANC < 0.5×109/L). 4. Non‐complaint individuals acknowledged by reviewing the patient's records.

Interventions

DFP, placebo

Outcomes

Primary outcome measures:

determine the time and number of transfusion units as well as amount of infused iron that will lead to appearance of LPI > 0.2 or TSAT > 50 % , serum ferritin ≥ 500 ng/mL in the studied thalassaemic patients which warrant start of iron chelation

Time frame: 12 months

To determine the time as well as amount of transfused iron (calculated in mg iron/kg) at which there is LPI appearance of > 0.2 as well as TSAT reaching 70 %, a serum ferritin ≥ 500 in order to start iron chelation therapy

Secondary outcome measures:

Evaluation of safety of early use of iron chelation therapy in terms of drug related AEs or SAEs

Time frame: 12 months

To determine the tolerability and safety of early low dose DFP 50mg/kg and effectiveness to postpone or prevent SF from reaching 1000 ng/mL or LPI > 0.6 or TSAT > 70% in comparison to participants not starting chelation therapy

Starting date

July 2014

Contact information

Amira AM Adly,

INSTITUTION: Pediatric Hematology clinic, Ain Shams University Cairo, Egypt

EMAIL: [email protected]

Notes

NCT02435212

Trial name or title

Study to evaluate treatment compliance, efficacy and safety of an improved deferasirox formulation (granules) in paediatric patients (2 ‐ < 18 years old) with iron overload

Methods

RCT; parallel group

Participants

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 patients according to local guidelines. 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 approximately 20 PRBC units and a treatment goal to reduce iron burden (300 mL PRBC = 1 unit in adults whereas 4 mL/kg PRBC is considered 1 unit for children). 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 and/or AST > 3.0 x ULN (Criterion no longer applicable, removed as part of amendment 1): 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. Those with significant impaired GI function or GI disease that may significantly alter the absorption of oral DFX (e.g. ulcerative diseases, uncontrolled nausea, vomiting, diarrhoea, malabsorption syndrome or small bowel resection

Interventions

DFX granule formulation, DFX DT formulation

Outcomes

Primary outcome measures: compliance
Change in SF in iron chelation therapy‐naive participants.
Secondary outcome measures: domain scores of treatment satisfaction and palatability over time
Overall safety, as measured by frequency and severity of adverse.This includes active monitoring for renal toxicity; including renal failure, hepatic toxicity; including hepatic failure, and gastrointestinal haemorrhage), and changes in laboratory values from baseline (serum creatinine, creatinine clearance, ALT, AST, RBC and WBC). In addition, vital signs, physical, ophthalmological, audiometric, cardiac, and growth and development evaluations will be assessed.
Rate of dosing instructions deviations ('Compliance', using a questionnaire) .
Pre‐dose DFX concentrations in all patients.

Pre‐dose PK data from all patients will be analysed to support the assessment of compliance.
Post‐dose DFX concentrations between 2 and 4 hours post‐dose
Change in SF in iron chelation therapy naive and pre‐treated participants
PK/PD relationship to explore exposure‐response relationships for measures of safety and effectiveness: serum creatinine change from baseline, notable serum creatinine values, serum creatinine clearance change from baseline and notable serum creatinine clearance categories, SF change from baseline, in relationship to derived PK parameters for pre‐ and post‐dose DFX concentrations.
Assess additional safety, as measured by frequency and severity of adverse for granules during extension phase includes active monitoring for renal toxicity; including renal failure, hepatic toxicity; including hepatic failure, and gastrointestinal haemorrhage), and changes in laboratory values from baseline (serum creatinine, creatinine clearance, ALT, AST, RBC and WBC). In addition, vital signs, physical, ophthalmological, audiometric, and growth and development evaluations will be assessed

Starting date

21 October 2015

Contact information

Principal Investigator: Janet L. KwiatkowskiI;

NSTITUTION: Children's Hospital of Philadelphia Onc. Dept;

EMAILContact: John Hammond 267‐426‐5602 [email protected]

ADDRESS: Children's Hospital of Philadelphia, Oncology Dept, Philadelphia, Pennsylvania, USA, 19104‐4399

Notes

March 30, 2023 (Final data collection date for primary outcome measure)

AEs: adverse events
ALT: alanine transaminase
ANC: absolute neutrophil count
AST: aspartate transaminase
CBC: complete blood count
DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
DT: dispersible tablet
GI: gastrointestinal
HPLC: high‐performance liquid chromatography
LIC: liver iron concentration
LPI: labile plasma iron
MRI: magnetic resonance imaging
PK/PD: pharmacokinetic/pharmacodynamic
QoL: quality of life
RBCs: red blood cells
RCT: randomised controlled trial
SAEs: serious adverse events
SF: serum ferritin
TSAT: transferrin saturation
ULN: upper limit of normal
WBC: white blood cell

Data and analyses

Open in table viewer
Comparison 1. DFP versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy (%, SD) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 DFP versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

Comparison 1 DFP versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

2 SAEs (from therapy, disease, non‐adherence) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

Analysis 1.2

Comparison 1 DFP versus DFO, Outcome 2 SAEs (from therapy, disease, non‐adherence).

Comparison 1 DFP versus DFO, Outcome 2 SAEs (from therapy, disease, non‐adherence).

2.1 Agranulocytosis

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

3 All‐cause mortality Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 1.3

Comparison 1 DFP versus DFO, Outcome 3 All‐cause mortality.

Comparison 1 DFP versus DFO, Outcome 3 All‐cause mortality.

4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.4

Comparison 1 DFP versus DFO, Outcome 4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L).

Comparison 1 DFP versus DFO, Outcome 4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L).

5 Organ damage Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 1.5

Comparison 1 DFP versus DFO, Outcome 5 Organ damage.

Comparison 1 DFP versus DFO, Outcome 5 Organ damage.

5.1 Liver damage

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Other AEs related to iron chelation Show forest plot

3

Risk Ratio (IV, Random, 99% CI)

Subtotals only

Analysis 1.6

Comparison 1 DFP versus DFO, Outcome 6 Other AEs related to iron chelation.

Comparison 1 DFP versus DFO, Outcome 6 Other AEs related to iron chelation.

6.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

192

Risk Ratio (IV, Random, 99% CI)

3.94 [0.44, 35.50]

6.2 Risk of pain or swelling in joints

3

192

Risk Ratio (IV, Random, 99% CI)

3.38 [0.54, 21.31]

6.3 Risk of nausea/vomiting

2

132

Risk Ratio (IV, Random, 99% CI)

13.68 [0.99, 188.88]

6.4 Risk of increased liver transaminase

1

44

Risk Ratio (IV, Random, 99% CI)

1.10 [0.03, 38.47]

6.5 Local reactions at infusion site

1

88

Risk Ratio (IV, Random, 99% CI)

0.17 [0.00, 9.12]

Open in table viewer
Comparison 2. DFX versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy (%, SD) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

Analysis 2.1

Comparison 2 DFX versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

Comparison 2 DFX versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

2 SAEs Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

Analysis 2.2

Comparison 2 DFX versus DFO, Outcome 2 SAEs.

Comparison 2 DFX versus DFO, Outcome 2 SAEs.

2.1 Thalassaemia‐related SAEs

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 SCD‐related SAE ‐ painful crisis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 SCD‐related SAEs ‐ other SCD‐related SAEs

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 All‐cause mortality (thalassaemia) Show forest plot

2

240

Risk Ratio (IV, Random, 95% CI)

0.96 [0.06, 15.06]

Analysis 2.3

Comparison 2 DFX versus DFO, Outcome 3 All‐cause mortality (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 3 All‐cause mortality (thalassaemia).

4 Proportion of participants with iron overload (thalassaemia) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 DFX versus DFO, Outcome 4 Proportion of participants with iron overload (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 4 Proportion of participants with iron overload (thalassaemia).

4.1 Iron overload defined by ferritin 1500 (µg/l) or higher (Thalassaemia)

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.63, 2.20]

4.2 Proportion with severe iron overload (LIC at least 15 mg/Fe/g dw)

1

172

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.83, 1.20]

4.3 Myocardial T2* < 10ms

1

172

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.72, 1.70]

5 Other AEs related to iron chelation ‐ (thalassaemia) Show forest plot

2

Risk Ratio (IV, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 DFX versus DFO, Outcome 5 Other AEs related to iron chelation ‐ (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 5 Other AEs related to iron chelation ‐ (thalassaemia).

5.1 Total chelation‐related AE

1

187

Risk Ratio (IV, Random, 95% CI)

1.15 [0.76, 1.73]

5.2 Gastrointestinal upset

1

60

Risk Ratio (IV, Random, 95% CI)

3.0 [0.66, 13.69]

5.3 Rash

2

247

Risk Ratio (IV, Random, 95% CI)

3.05 [0.98, 9.47]

5.4 Risk of increased blood creatinine

1

187

Risk Ratio (IV, Random, 95% CI)

3.79 [0.83, 17.38]

5.5 Risk of proteinuria

1

187

Risk Ratio (IV, Random, 95% CI)

2.21 [0.59, 8.29]

5.6 Risk of increased ALT

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.7 Risk of increased AST

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.8 Risk of diarrhoea

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.9 Risk of vomiting

1

187

Risk Ratio (IV, Random, 95% CI)

6.64 [0.35, 126.78]

6 Total AEs (thalassaemia) Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2 DFX versus DFO, Outcome 6 Total AEs (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 6 Total AEs (thalassaemia).

7 Other AEs related to iron chelation (SCD) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

Analysis 2.7

Comparison 2 DFX versus DFO, Outcome 7 Other AEs related to iron chelation (SCD).

Comparison 2 DFX versus DFO, Outcome 7 Other AEs related to iron chelation (SCD).

7.1 Risk of increased ALT

1

195

Risk Ratio (M‐H, Random, 99% CI)

5.29 [0.12, 232.98]

7.2 incidence of abdominal pain

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.91 [0.80, 4.58]

7.3 Risk of pain or swelling in joints

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.06 [0.41, 2.76]

7.4 Risk of diarrhoea

1

195

Risk Ratio (M‐H, Random, 99% CI)

4.14 [0.90, 18.92]

7.5 Nausea/vomiting

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.63 [0.90, 2.94]

Open in table viewer
Comparison 3. DFX film‐coated tablet versus DFX dispersible tablet

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy Show forest plot

1

173

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.99, 1.22]

Analysis 3.1

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1 Adherence to iron chelation therapy.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1 Adherence to iron chelation therapy.

2 Incidence of SAEs Show forest plot

1

173

Risk Ratio (IV, Random, 95% CI)

1.22 [0.62, 2.37]

Analysis 3.2

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2 Incidence of SAEs.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2 Incidence of SAEs.

3 All‐cause mortality Show forest plot

1

173

Risk Ratio (M‐H, Random, 95% CI)

2.97 [0.12, 71.81]

Analysis 3.3

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3 All‐cause mortality.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3 All‐cause mortality.

4 Incidence of organ damage (renal event) Show forest plot

1

173

Risk Ratio (IV, Random, 95% CI)

1.25 [0.83, 1.91]

Analysis 3.4

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4 Incidence of organ damage (renal event).

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4 Incidence of organ damage (renal event).

5 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (IV, Random, 99% CI)

Subtotals only

Analysis 3.5

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5 Other AEs related to iron chelation.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5 Other AEs related to iron chelation.

5.1 Total chelation‐related AEs

1

173

Risk Ratio (IV, Random, 99% CI)

0.75 [0.52, 1.08]

5.2 Risk of diarrhoea

1

173

Risk Ratio (IV, Random, 99% CI)

0.70 [0.29, 1.70]

5.3 Increased urine protein/urine creatinine ratio

1

173

Risk Ratio (IV, Random, 99% CI)

1.65 [0.60, 4.54]

5.4 incidence of abdominal pain

1

173

Risk Ratio (IV, Random, 99% CI)

0.49 [0.16, 1.52]

5.5 Incidence of nausea

1

173

Risk Ratio (IV, Random, 99% CI)

0.72 [0.23, 2.23]

5.6 Incidence of vomiting

1

173

Risk Ratio (IV, Random, 99% CI)

0.28 [0.07, 1.15]

Open in table viewer
Comparison 4. DFP and DFO versus DFP

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Incidence of SAEs Show forest plot

1

213

Risk Ratio (M‐H, Random, 95% CI)

0.15 [0.01, 2.81]

Analysis 4.1

Comparison 4 DFP and DFO versus DFP, Outcome 1 Incidence of SAEs.

Comparison 4 DFP and DFO versus DFP, Outcome 1 Incidence of SAEs.

2 All‐cause mortality Show forest plot

2

237

Risk Ratio (IV, Random, 95% CI)

0.77 [0.18, 3.35]

Analysis 4.2

Comparison 4 DFP and DFO versus DFP, Outcome 2 All‐cause mortality.

Comparison 4 DFP and DFO versus DFP, Outcome 2 All‐cause mortality.

3 Incidence of chelation therapy‐related AEs Show forest plot

3

Risk Ratio (IV, Random, 99% CI)

Subtotals only

Analysis 4.3

Comparison 4 DFP and DFO versus DFP, Outcome 3 Incidence of chelation therapy‐related AEs.

Comparison 4 DFP and DFO versus DFP, Outcome 3 Incidence of chelation therapy‐related AEs.

3.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

280

Risk Ratio (IV, Random, 99% CI)

1.15 [0.50, 2.62]

3.2 Risk of pain or swelling in joints

2

256

Risk Ratio (IV, Random, 99% CI)

0.76 [0.31, 1.91]

3.3 Risk of gastrointestinal disturbances

1

213

Risk Ratio (IV, Random, 99% CI)

0.45 [0.15, 1.37]

3.4 Risk of increased liver transaminase

2

256

Risk Ratio (IV, Random, 99% CI)

1.02 [0.52, 1.98]

3.5 Nausea/vomiting

1

43

Risk Ratio (IV, Random, 99% CI)

0.55 [0.13, 2.23]

Open in table viewer
Comparison 5. DFP and DFO versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Other AEs related to iron chelation Show forest plot

4

Risk Ratio (IV, Random, 99% CI)

Subtotals only

Analysis 5.1

Comparison 5 DFP and DFO versus DFO, Outcome 1 Other AEs related to iron chelation.

Comparison 5 DFP and DFO versus DFO, Outcome 1 Other AEs related to iron chelation.

1.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

169

Risk Ratio (IV, Random, 99% CI)

1.18 [0.09, 15.37]

1.2 Risk of pain or swelling in joints

3

135

Risk Ratio (IV, Random, 99% CI)

2.39 [0.18, 32.31]

1.3 Risk of increased liver transaminase

2

104

Risk Ratio (IV, Random, 99% CI)

3.46 [0.45, 26.62]

1.4 Nausea/vomiting

4

194

Risk Ratio (IV, Random, 99% CI)

3.81 [0.84, 17.36]

1.5 Local reactions at infusion site

2

90

Risk Ratio (IV, Random, 99% CI)

0.18 [0.01, 3.56]

Open in table viewer
Comparison 6. DFP/DFX versus DFP/DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy rates Show forest plot

1

96

Risk Ratio (IV, Random, 95% CI)

0.84 [0.72, 0.99]

Analysis 6.1

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 1 Adherence to iron chelation therapy rates.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 1 Adherence to iron chelation therapy rates.

2 Incidence of SAE Show forest plot

1

96

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.06, 15.53]

Analysis 6.2

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 2 Incidence of SAE.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 2 Incidence of SAE.

3 All‐cause mortality Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

Analysis 6.3

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 3 All‐cause mortality.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 3 All‐cause mortality.

4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions) Show forest plot

1

96

Risk Ratio (M‐H, Random, 99% CI)

3.0 [0.16, 56.04]

Analysis 6.4

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions).

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions).

5 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (IV, Random, 99% CI)

Subtotals only

Analysis 6.5

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 5 Other AEs related to iron chelation.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 5 Other AEs related to iron chelation.

5.1 one year (study end)

1

96

Risk Ratio (IV, Random, 99% CI)

1.08 [0.68, 1.71]

5.2 Risk of leukopenia, neutropenia and/or agranulocytosis

1

96

Risk Ratio (IV, Random, 99% CI)

1.67 [0.27, 10.14]

5.3 Risk of pain or swelling in joints

1

96

Risk Ratio (IV, Random, 99% CI)

0.89 [0.29, 2.77]

5.4 Gastrointestinal problems

1

96

Risk Ratio (IV, Random, 99% CI)

0.6 [0.18, 2.04]

5.5 ALT (increase ≥3 folds)

1

96

Risk Ratio (IV, Random, 99% CI)

1.33 [0.20, 8.88]

5.6 Skin rash

1

96

Risk Ratio (IV, Random, 99% CI)

5.0 [0.10, 261.34]

Study flow diagram.
Figuras y tablas -
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.
Figuras y tablas -
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.
Figuras y tablas -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 DFP versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).
Figuras y tablas -
Analysis 1.1

Comparison 1 DFP versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

Comparison 1 DFP versus DFO, Outcome 2 SAEs (from therapy, disease, non‐adherence).
Figuras y tablas -
Analysis 1.2

Comparison 1 DFP versus DFO, Outcome 2 SAEs (from therapy, disease, non‐adherence).

Comparison 1 DFP versus DFO, Outcome 3 All‐cause mortality.
Figuras y tablas -
Analysis 1.3

Comparison 1 DFP versus DFO, Outcome 3 All‐cause mortality.

Comparison 1 DFP versus DFO, Outcome 4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L).
Figuras y tablas -
Analysis 1.4

Comparison 1 DFP versus DFO, Outcome 4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L).

Comparison 1 DFP versus DFO, Outcome 5 Organ damage.
Figuras y tablas -
Analysis 1.5

Comparison 1 DFP versus DFO, Outcome 5 Organ damage.

Comparison 1 DFP versus DFO, Outcome 6 Other AEs related to iron chelation.
Figuras y tablas -
Analysis 1.6

Comparison 1 DFP versus DFO, Outcome 6 Other AEs related to iron chelation.

Comparison 2 DFX versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).
Figuras y tablas -
Analysis 2.1

Comparison 2 DFX versus DFO, Outcome 1 Adherence to iron chelation therapy (%, SD).

Comparison 2 DFX versus DFO, Outcome 2 SAEs.
Figuras y tablas -
Analysis 2.2

Comparison 2 DFX versus DFO, Outcome 2 SAEs.

Comparison 2 DFX versus DFO, Outcome 3 All‐cause mortality (thalassaemia).
Figuras y tablas -
Analysis 2.3

Comparison 2 DFX versus DFO, Outcome 3 All‐cause mortality (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 4 Proportion of participants with iron overload (thalassaemia).
Figuras y tablas -
Analysis 2.4

Comparison 2 DFX versus DFO, Outcome 4 Proportion of participants with iron overload (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 5 Other AEs related to iron chelation ‐ (thalassaemia).
Figuras y tablas -
Analysis 2.5

Comparison 2 DFX versus DFO, Outcome 5 Other AEs related to iron chelation ‐ (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 6 Total AEs (thalassaemia).
Figuras y tablas -
Analysis 2.6

Comparison 2 DFX versus DFO, Outcome 6 Total AEs (thalassaemia).

Comparison 2 DFX versus DFO, Outcome 7 Other AEs related to iron chelation (SCD).
Figuras y tablas -
Analysis 2.7

Comparison 2 DFX versus DFO, Outcome 7 Other AEs related to iron chelation (SCD).

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1 Adherence to iron chelation therapy.
Figuras y tablas -
Analysis 3.1

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1 Adherence to iron chelation therapy.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2 Incidence of SAEs.
Figuras y tablas -
Analysis 3.2

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2 Incidence of SAEs.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3 All‐cause mortality.
Figuras y tablas -
Analysis 3.3

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3 All‐cause mortality.

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4 Incidence of organ damage (renal event).
Figuras y tablas -
Analysis 3.4

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4 Incidence of organ damage (renal event).

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5 Other AEs related to iron chelation.
Figuras y tablas -
Analysis 3.5

Comparison 3 DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5 Other AEs related to iron chelation.

Comparison 4 DFP and DFO versus DFP, Outcome 1 Incidence of SAEs.
Figuras y tablas -
Analysis 4.1

Comparison 4 DFP and DFO versus DFP, Outcome 1 Incidence of SAEs.

Comparison 4 DFP and DFO versus DFP, Outcome 2 All‐cause mortality.
Figuras y tablas -
Analysis 4.2

Comparison 4 DFP and DFO versus DFP, Outcome 2 All‐cause mortality.

Comparison 4 DFP and DFO versus DFP, Outcome 3 Incidence of chelation therapy‐related AEs.
Figuras y tablas -
Analysis 4.3

Comparison 4 DFP and DFO versus DFP, Outcome 3 Incidence of chelation therapy‐related AEs.

Comparison 5 DFP and DFO versus DFO, Outcome 1 Other AEs related to iron chelation.
Figuras y tablas -
Analysis 5.1

Comparison 5 DFP and DFO versus DFO, Outcome 1 Other AEs related to iron chelation.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 1 Adherence to iron chelation therapy rates.
Figuras y tablas -
Analysis 6.1

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 1 Adherence to iron chelation therapy rates.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 2 Incidence of SAE.
Figuras y tablas -
Analysis 6.2

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 2 Incidence of SAE.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 3 All‐cause mortality.
Figuras y tablas -
Analysis 6.3

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 3 All‐cause mortality.

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions).
Figuras y tablas -
Analysis 6.4

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions).

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 5 Other AEs related to iron chelation.
Figuras y tablas -
Analysis 6.5

Comparison 6 DFP/DFX versus DFP/DFO, Outcome 5 Other AEs related to iron chelation.

Summary of findings for the main comparison. DFP compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFP compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFP
Comparison: DFO

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFO

Risk with DFP

Adherence to iron chelation therapy (per cent, SD)

242

(4 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

We found considerable heterogeneity and identified age as possible cause: 1 trial in children 10 years or older and 1 conducted in participants 18 or older

SAEs (from therapy, disease, non‐adherence) Agranulocytosis**

Study population

RR 7.88
(99% CI 0.18 to 352.39)

88
(1 RCT)

⊕⊝⊝⊝
VERY LOW 3 4

No SAEs were reported in the second trial reporting this outcome

15 per 1000

118 per 1,000

(7 to 1000)

All‐cause mortality

Study population

RR 0.44
(95% CI 0.12 to 1.63)

88
(1 RCT)

⊕⊝⊝⊝
VERY LOW 3 4

No deaths occurred in the second trial reporting this outcome

146 per 1000

64 per 1000
(18 to 239)

Sustained adherence ‐ not measured

Sustained adherence is reported as adherence as all trials were longer than 6 months and only end of trial adherence numbers were provided

Quality of life ‐ not reported

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DFO: deferoxamine; DFP: deferiprone; RCT: randomised controlled trial; RR: risk ratio; SD: standard deviation.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 1 for risk of bias due to high or uncertain risk of bias due to lack of blinding of participants and personnel in all four RCTs, as well as selection bias (Olivieri 1997), attrition bias (El Beshlawy 2008; Olivieri 1997), reporting bias (El Beshlawy 2008; Pennell 2006), and other bias (Pennell 2006).
2 We downgraded the quality of evidence by 2 for inconsistency due to considerable heterogeneity in comparison.
3 We downgraded the quality of evidence by 2 for imprecision due to very wide CIs that included clinically important benefits and harms.
4 We downgraded the quality of evidence by 1 for indirectness as the trial was conducted in participants with thalassaemia intermedia only; a milder form of thalassaemia

** Risk estimate based on: Tricta F, Uetrecht J, Galanello R, et al. Deferiprone‐induced agranulocytosis: 20 years of clinical observations. American Journal of Hematology. 2016;91(10):1026‐1031. doi:10.1002/ajh.24479.

Figuras y tablas -
Summary of findings for the main comparison. DFP compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 2. DFX compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFX compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFX
Comparison: DFO

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFO

Risk with DFX

Adherence to iron chelation therapy (per cent, SD)

The mean adherence to iron chelation therapy (per cent, SD) was 0

MD 1.4 lower
(3.66 lower to 0.86 higher)

197
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

Narrative report of adherence for 2 trials as either no or incompatible data to enable comparisons

SAEs ‐ thalassaemia‐related SAEs

Study population

247
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

There were no SAEs to report in one trial so no estimate of effect

see comment

see comment

SAEs ‐ SCD‐related SAEs

Study population

RR 1.08
(95% CI 0.77 to 1.51)

195
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

429 per 1000

463 per 1000
(330 to 647)

Incidence of SCD‐related SAEs ‐pain crisis

Study population

RR 1.05
(95% CI 0.68 to 1.62)

195
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

317 per 1000

333 per 1000
(216 to 514)

All‐cause mortality (thalassaemia)

Study population

RR 0.96
(95%CI 0.06 to 15.06)

240
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

8 per 1000

8 per 1000
(1 to 128)

Sustained adherence ‐ not measured

Sustained adherence is reported as adherence as all trials were longer than 6 months and only end of trial adherence reported

Quality of life ‐ not 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).

CI: confidence interval; DFO: deferoxamine; DFX: deferasirox; MD: mean difference; RCT: randomised controlled trial; RR: risk ratio; SD: standard deviation.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 2 due to high or uncertain risk of bias in several domains
2 We downgraded the quality of evidence by 1 due to imprecision as CIs are wide and only 1 trial with data in comparison

Figuras y tablas -
Summary of findings 2. DFX compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 3. DFX film‐coated tablet compared to DFX dispersible tablet for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFX film‐coated tablet compared to DFX dispersible tablet for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFX film‐coated tablet
Comparison: DFX dispersible tablet

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFX dispersible tablet

Risk with DFX film‐coated tablet

Adherence to iron chelation therapy (n, N)

Study population

RR 1.10
(95% CI 0.99 to 1.22)

173
(1 RCT)

⊕⊕⊝⊝
LOW 1

849 per 1000

934 per 1000
(840 to 1000)

Incidence of SAEs

Study population

RR 1.22
(95% CI 0.62 to 2.37)

173
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

151 per 1,000

184 per 1000
(94 to 358)

All‐cause mortality

Study population

RR 2.97
(95% CI 0.12 to 71.81)

173
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

0 per 1000

0 per 1000
(0 to 0)

Sustained adherence ‐ not measured

Reported as adherence as trial was 6 months in duration and end of trial adherence reported

Quality of life ‐ not reported

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DFX: deferasirox; RCT: randomised controlled trial; RR: risk ratio; SAEs: serious adverse events

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 2 for risk of bias due to high or unclear risk of bias in all domains
2 We downgraded the quality of evidence by 1 for imprecision due to wide CIs

Figuras y tablas -
Summary of findings 3. DFX film‐coated tablet compared to DFX dispersible tablet for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 4. DFP and DFO compared to DFP for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFP and DFO compared to DFP for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFP and DFO
Comparison: DFP

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFP

Risk with DFP and DFO

Adherence to iron chelation therapy (per cent, SD)

see comment

see comment

289

(3 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

Reported as narrative as no comparisons possible

Incidence of SAEs

Study population

RR 0.15
(95% CI 0.01 to 2.81)

213
(1 RCT)

⊕⊕⊝⊝
LOW 2 3

28 per 1,000

4 per 1,000
(0 to 78)

All‐cause mortality

Study population

RR 0.77
(95% CI 0.18 to 3.35)

237
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 3 4

33 per 1,000

26 per 1,000
(6 to 112)

Sustained adherence ‐ not measured

Sustained adherence is reported as adherence as trial duration longer than 6 months and reports adherence for length of trial

Quality of life ‐ not reported

Quality of life was either not reported or no validated instruments were used

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DFO: deferoxamine DFP: deferiprone; RCT: randomised controlled trial; RR: risk ratio; SAEs: serious adverse events; SD: standard deviation.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 2 for risk of bias as there was high or uncertain risk of bias in most domains in 3 out of 4 trials
2 We downgraded the quality of evidence by 1 due to high or unclear risk of bias in 3 domains
3 We downgraded the quality of evidence by 1 for imprecision due to wide CIs
4 We downgraded the quality of evidence by 2 for risk of bias as there was high or uncertain risk of bias in 1 of the trials in the comparison

Figuras y tablas -
Summary of findings 4. DFP and DFO compared to DFP for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 5. DFP and DFO compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFP and DFO compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFP and DFO
Comparison: DFO

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFO

Risk with DFP and DFO

Adherence to iron chelation therapy (per cent, SD)

see comment

see comment

205

(4 RCTs)

⊕⊕⊝⊝
LOW 1

Reported as narrative only as adherence in combined group not reported for combination therapy

Incidence of SAEs

Study population

205

(4 RCTs)

⊕⊕⊝⊝
LOW 1

3 trials report no SAEs; SAES are not reported in one trial

see comment

see comment

All‐cause mortality

Study population

205

(4 RCTs)

⊕⊕⊝⊝
LOW 1

no deaths reported

see comment

see comment

Sustained adherence ‐ not measured

Sustained adherence reported as adherence as trial duration was longer than 6 months and adherence reported at end of trial

Quality of life ‐ not 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).

DFO: deferoxamine; DFP: deferiprone; SAEs: serious adverse events.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 2 for risk of bias as high or unclear risk of bias in all domains

Figuras y tablas -
Summary of findings 5. DFP and DFO compared to DFO for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 6. DFP and DFO compared to DFP and DFX for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

DFP/DFO compared to DFP/DFX for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatients
Intervention: DFP/DFO
Comparison: DFP/DFX

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFP/DFX

Risk with DFP/DFO

Adherence to iron chelation therapy rates (n,N) ‐ 1 year

Study population

RR 0.84
(95% CI 0.72 to 0.99)

96
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

938 per 1000

788 per 1000
(675 to 928)

Incidence of SAE

Study population

RR 1.00
(95% CI 0.06 to 15.53)

96
(1 RCT)

⊕⊝⊝⊝
VERY LOW1 2 3

21 per 1,000

21 per 1000
(1 to 324)

All‐cause mortality ‐ at 1 year ‐ trial end

Study population

Not estimable

96
(1 RCT)

⊕⊕⊝⊝
LOW 1 2

No deaths were reported

0 per 1000

0 per 1000
(0 to 0)

Sustained adherence ‐ not measured

Sustained adherence is reported as adherence as trial was 1 year in duration and end of trial adherence reported

Quality of life see comment

The study uses SF36 to measure quality of life, the results are presented as a graph. Quality of life increased in both trial arms with no significant difference between trial arms P = 0.860

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; DFO: deferoxamine; DFP: deferiprone; DFX: deferasirox; RCT: randomised controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence by 1 for risk of bias as there was high or unclear risk of bias in 3 domains
2 We downgraded the quality of evidence by 1 for indirectness as the trial included children 10 ‐ 18 with severe iron overload
3 We downgraded the quality of evidence by 1 for imprecision as the comparison has wide CIs

Figuras y tablas -
Summary of findings 6. DFP and DFO compared to DFP and DFX for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Summary of findings 7. Medication management compared to standard care for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Medication management compared to standard care for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia

Patient or population: improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Setting: outpatient
Intervention: medication management
Comparison: standard care

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with standard care

Risk with medication management

Adherence to iron chelation therapy ‐ not reported

Adherence was only reported in the intervention group and therefore no comparative data

SAEs ‐ not reported

Mortality ‐ not reported

Sustained adherence

Adherence was only reported in the intervention group and therefore no comparative data

Quality of life
assessed with: PedsQLTM HRQoL total score

48
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1 2

Medication management: 63.51 (51.75 – 84.54); standard care: 49.84 (41.9 – 60.81)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

RCT: randomised controlled trial; SAEs: serious adverse events.

GRADE Working Group grades of evidence
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded the quality of evidence for indirectness by 2 because most outcomes were only reported in the medication management group
2 We downgraded the quality of evidence by 2 for risk of bias due to high or uncertain risk of bias in all domains

Figuras y tablas -
Summary of findings 7. Medication management compared to standard care for improving adherence to iron chelation therapy in people with sickle cell disease or thalassaemia
Table 1. Adherence Measurement and Results Table

STUDY

HOW ADHERENCE MEASURED

RESULTS

Aydinok 2007

  • Drug accounting at each visit (by counting the returned empty blisters of DFP and used vials of DFO)

  • Trial‐specific designed questionnaire completed by the participants or their legal representative/guardian (or both) at quarterly intervals

  • Compliance was generally excellent during the entire trial period

  • 1 participant in the DFP treatment arm who missed more than 1 chelation dose per week because of problems with swallowing

Badawy 2010

  • Questionnaire on chelation therapy, reasons for non‐compliance, side effects, life activities, transfusion regimen

  • Group II and group I were more compliant to chelation therapy but difference was statistically non significant

  • Non‐compliant participants (compliance less than 50%) showed increase in their SF levels in all studied groups

  • In non‐compliant participants the reduction in SF levels was higher in group I and III than in group II but difference was statistically non significant

Bahnasawy 2017

  • Clinical pharmacist analysed data to detect unnecessary drug therapy, need for additional drug therapy, ineffective drug product, dosage too low, adverse drug reaction, dosage too high, non‐compliance

  • All 24 participants in intervention group had non‐adherence at baseline and 3 where non‐adherent at end of trial

  • No data on control group

Calvaruso 2015

  • Counting the number of DFP pills in each returned bag

  • Assessing the number of infusions of DFO registered on the electronic pump

  • DFP compliance rate: 85%

  • DFO compliance rate: 76%

El Beshlawy 2008

  • Counting the returned empty blisters of DFP

  • Counting used vials of DFO

  • 4 participants with DFO‐based regimen excluded from the trial due to lack of compliance

  • Compliance was otherwise excellent during the entire trial period

  • Majority of participants had no problems with the intake and swallowing of the DFP tablets

  • 80% of participants in the combination arm and 76% of participants in the DFO monotherapy arm complained about difficulties in the parenteral use of DFO or problems to insert a needle

Elalfy 2015

  • Counting of returned tablets for the oral chelators

  • Counting vials for DFO

  • The percentage of actual dose that patient had taken in relation to the total prescribed dose was calculated

  • DFP/DFX: 95%

  • DFP/DFO: 80%

Galanello 2006

  • DFP assessed by pill counts, diary cards and an electronic cap that recorded the time and date of each opening of the tablet container

  • DFO assessed by diary cards, weekly physical examination of infusion sites, and by the Crono™ infusion pump that recorded the number of completed infusions

  • DFP/DFO: DFO: 96.1 ±5.0 (29 participants)

  • DFP compliance was not reported

  • DFO: 95.7 ± 5.7 (30 participants)

Hassan 2016

  • Records of all trial medications that were dispensed and returned

  • Parents were instructed to contact the investigator if the participant were unable to take the trial drug as prescribed

  • All participants compliant with prescribed doses

  • No discontinuation of drugs or dropout of follow‐up occurred

Maggio 2009

  • Counting the pills in each returned bag of DFP

  • Assessing the number of infusions of DFO registered on the electronic pump

  • DFP–DFO group: DFP: 92.7% (SD ± 15.2%; range 37–100%): DFO: 70.6% (SD ± 24.1%; range 25–100%)

  • DFP alone participants: 93.6% (SD ± 9.7%; range 56–100%)

Mourad 2003

  • Number of vials of DFX used

  • Number of tablets of DFO used

  • DFO/DFX group: compliance was excellent (arbitrarily defined as taking > 90% of the recommended doses) in 10 participants and good (75% to 90% of recommended doses) in 1 participant

  • DFX alone group: compliance was considered to be excellent in 11 patients and good in 3 participants

Olivieri 1997

  • Per cent of doses administered: number of doses of the iron chelator taken, out of number prescribed

  • DFP measured with computerised bottles

  • DFO measured using ambulatory pumps

  • Measured for a minimum of 3 months

  • DFP: 94.9% ± 1.1%

  • DFO: 71.6% ± 3.7%

Pennell 2006

  • DFP: measured using the Medication Event Monitoring System device calculated as the percent of openings with an interval longer than 4 hours recorded, divided by number of doses prescribed

  • DFO: calculated as the percentage of completed infusions, as determined by the Crono pumps, divided by the number of infusions prescribed

  • DFP: 94% ± 5.3%

  • DFO: 93% ± 9.7%

Pennell 2014

  • Not stated how adherence was measured

  • DFX: 99.0% ± 3.5%

  • DFO: 100.4% ± 10.9%

Taher 2017

  • Assessed by relative consumed tablet count

  • DT: 85.3% (95% CI: 81.1, 89.5)

  • FCT: 92.9% (95% CI: 88.8, 97.0)

Tanner 2007

  • DFO: calculated as the percentage of completed infusions, as determined by the Crono pumps, divided by the number of infusions prescribed

  • DFP/placebo: pill counting at the bimonthly visits

  • DFO/placebo: DFO: 91.4 ± 2.7%; placebo: 89.8 ± 7.2%;

  • DFO/DFP: DFO: 92.6 ± 2.7%; DFP: 82.4 ± 18.1%

Vichinsky 2007

  • DFX: counting the number of tablets returned in bottles at each visit

  • DFO: counting the numbers of vials returned at each visit

  • Ratios of the administered to intended doses of therapy were high (1.16 for DFX and 0.97 for DFO), indicating high adherence to the prescribed treatment regimens

DFO: deferoxamine
DFP: deferiprone
DFX: deferasirox
DT: dispersible tablet
FCT: film‐coated tablet
SD: standard deviation
SF: serum ferritin

Figuras y tablas -
Table 1. Adherence Measurement and Results Table
Comparison 1. DFP versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy (%, SD) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

2 SAEs (from therapy, disease, non‐adherence) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Totals not selected

2.1 Agranulocytosis

1

Risk Ratio (M‐H, Random, 99% CI)

0.0 [0.0, 0.0]

3 All‐cause mortality Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

4 Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5 Organ damage Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

5.1 Liver damage

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

6 Other AEs related to iron chelation Show forest plot

3

Risk Ratio (IV, Random, 99% CI)

Subtotals only

6.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

192

Risk Ratio (IV, Random, 99% CI)

3.94 [0.44, 35.50]

6.2 Risk of pain or swelling in joints

3

192

Risk Ratio (IV, Random, 99% CI)

3.38 [0.54, 21.31]

6.3 Risk of nausea/vomiting

2

132

Risk Ratio (IV, Random, 99% CI)

13.68 [0.99, 188.88]

6.4 Risk of increased liver transaminase

1

44

Risk Ratio (IV, Random, 99% CI)

1.10 [0.03, 38.47]

6.5 Local reactions at infusion site

1

88

Risk Ratio (IV, Random, 99% CI)

0.17 [0.00, 9.12]

Figuras y tablas -
Comparison 1. DFP versus DFO
Comparison 2. DFX versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy (%, SD) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Totals not selected

2 SAEs Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Totals not selected

2.1 Thalassaemia‐related SAEs

2

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 SCD‐related SAE ‐ painful crisis

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.3 SCD‐related SAEs ‐ other SCD‐related SAEs

1

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

3 All‐cause mortality (thalassaemia) Show forest plot

2

240

Risk Ratio (IV, Random, 95% CI)

0.96 [0.06, 15.06]

4 Proportion of participants with iron overload (thalassaemia) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Iron overload defined by ferritin 1500 (µg/l) or higher (Thalassaemia)

1

60

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.63, 2.20]

4.2 Proportion with severe iron overload (LIC at least 15 mg/Fe/g dw)

1

172

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.83, 1.20]

4.3 Myocardial T2* < 10ms

1

172

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.72, 1.70]

5 Other AEs related to iron chelation ‐ (thalassaemia) Show forest plot

2

Risk Ratio (IV, Random, 95% CI)

Subtotals only

5.1 Total chelation‐related AE

1

187

Risk Ratio (IV, Random, 95% CI)

1.15 [0.76, 1.73]

5.2 Gastrointestinal upset

1

60

Risk Ratio (IV, Random, 95% CI)

3.0 [0.66, 13.69]

5.3 Rash

2

247

Risk Ratio (IV, Random, 95% CI)

3.05 [0.98, 9.47]

5.4 Risk of increased blood creatinine

1

187

Risk Ratio (IV, Random, 95% CI)

3.79 [0.83, 17.38]

5.5 Risk of proteinuria

1

187

Risk Ratio (IV, Random, 95% CI)

2.21 [0.59, 8.29]

5.6 Risk of increased ALT

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.7 Risk of increased AST

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.8 Risk of diarrhoea

1

187

Risk Ratio (IV, Random, 95% CI)

5.69 [0.70, 46.33]

5.9 Risk of vomiting

1

187

Risk Ratio (IV, Random, 95% CI)

6.64 [0.35, 126.78]

6 Total AEs (thalassaemia) Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

7 Other AEs related to iron chelation (SCD) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

7.1 Risk of increased ALT

1

195

Risk Ratio (M‐H, Random, 99% CI)

5.29 [0.12, 232.98]

7.2 incidence of abdominal pain

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.91 [0.80, 4.58]

7.3 Risk of pain or swelling in joints

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.06 [0.41, 2.76]

7.4 Risk of diarrhoea

1

195

Risk Ratio (M‐H, Random, 99% CI)

4.14 [0.90, 18.92]

7.5 Nausea/vomiting

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.63 [0.90, 2.94]

Figuras y tablas -
Comparison 2. DFX versus DFO
Comparison 3. DFX film‐coated tablet versus DFX dispersible tablet

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy Show forest plot

1

173

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.99, 1.22]

2 Incidence of SAEs Show forest plot

1

173

Risk Ratio (IV, Random, 95% CI)

1.22 [0.62, 2.37]

3 All‐cause mortality Show forest plot

1

173

Risk Ratio (M‐H, Random, 95% CI)

2.97 [0.12, 71.81]

4 Incidence of organ damage (renal event) Show forest plot

1

173

Risk Ratio (IV, Random, 95% CI)

1.25 [0.83, 1.91]

5 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (IV, Random, 99% CI)

Subtotals only

5.1 Total chelation‐related AEs

1

173

Risk Ratio (IV, Random, 99% CI)

0.75 [0.52, 1.08]

5.2 Risk of diarrhoea

1

173

Risk Ratio (IV, Random, 99% CI)

0.70 [0.29, 1.70]

5.3 Increased urine protein/urine creatinine ratio

1

173

Risk Ratio (IV, Random, 99% CI)

1.65 [0.60, 4.54]

5.4 incidence of abdominal pain

1

173

Risk Ratio (IV, Random, 99% CI)

0.49 [0.16, 1.52]

5.5 Incidence of nausea

1

173

Risk Ratio (IV, Random, 99% CI)

0.72 [0.23, 2.23]

5.6 Incidence of vomiting

1

173

Risk Ratio (IV, Random, 99% CI)

0.28 [0.07, 1.15]

Figuras y tablas -
Comparison 3. DFX film‐coated tablet versus DFX dispersible tablet
Comparison 4. DFP and DFO versus DFP

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Incidence of SAEs Show forest plot

1

213

Risk Ratio (M‐H, Random, 95% CI)

0.15 [0.01, 2.81]

2 All‐cause mortality Show forest plot

2

237

Risk Ratio (IV, Random, 95% CI)

0.77 [0.18, 3.35]

3 Incidence of chelation therapy‐related AEs Show forest plot

3

Risk Ratio (IV, Random, 99% CI)

Subtotals only

3.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

280

Risk Ratio (IV, Random, 99% CI)

1.15 [0.50, 2.62]

3.2 Risk of pain or swelling in joints

2

256

Risk Ratio (IV, Random, 99% CI)

0.76 [0.31, 1.91]

3.3 Risk of gastrointestinal disturbances

1

213

Risk Ratio (IV, Random, 99% CI)

0.45 [0.15, 1.37]

3.4 Risk of increased liver transaminase

2

256

Risk Ratio (IV, Random, 99% CI)

1.02 [0.52, 1.98]

3.5 Nausea/vomiting

1

43

Risk Ratio (IV, Random, 99% CI)

0.55 [0.13, 2.23]

Figuras y tablas -
Comparison 4. DFP and DFO versus DFP
Comparison 5. DFP and DFO versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Other AEs related to iron chelation Show forest plot

4

Risk Ratio (IV, Random, 99% CI)

Subtotals only

1.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

169

Risk Ratio (IV, Random, 99% CI)

1.18 [0.09, 15.37]

1.2 Risk of pain or swelling in joints

3

135

Risk Ratio (IV, Random, 99% CI)

2.39 [0.18, 32.31]

1.3 Risk of increased liver transaminase

2

104

Risk Ratio (IV, Random, 99% CI)

3.46 [0.45, 26.62]

1.4 Nausea/vomiting

4

194

Risk Ratio (IV, Random, 99% CI)

3.81 [0.84, 17.36]

1.5 Local reactions at infusion site

2

90

Risk Ratio (IV, Random, 99% CI)

0.18 [0.01, 3.56]

Figuras y tablas -
Comparison 5. DFP and DFO versus DFO
Comparison 6. DFP/DFX versus DFP/DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Adherence to iron chelation therapy rates Show forest plot

1

96

Risk Ratio (IV, Random, 95% CI)

0.84 [0.72, 0.99]

2 Incidence of SAE Show forest plot

1

96

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.06, 15.53]

3 All‐cause mortality Show forest plot

1

Risk Ratio (IV, Random, 95% CI)

Totals not selected

4 Organ damage (serum creatinine (≥33%) above baseline in 2 consecutive occasions) Show forest plot

1

96

Risk Ratio (M‐H, Random, 99% CI)

3.0 [0.16, 56.04]

5 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (IV, Random, 99% CI)

Subtotals only

5.1 one year (study end)

1

96

Risk Ratio (IV, Random, 99% CI)

1.08 [0.68, 1.71]

5.2 Risk of leukopenia, neutropenia and/or agranulocytosis

1

96

Risk Ratio (IV, Random, 99% CI)

1.67 [0.27, 10.14]

5.3 Risk of pain or swelling in joints

1

96

Risk Ratio (IV, Random, 99% CI)

0.89 [0.29, 2.77]

5.4 Gastrointestinal problems

1

96

Risk Ratio (IV, Random, 99% CI)

0.6 [0.18, 2.04]

5.5 ALT (increase ≥3 folds)

1

96

Risk Ratio (IV, Random, 99% CI)

1.33 [0.20, 8.88]

5.6 Skin rash

1

96

Risk Ratio (IV, Random, 99% CI)

5.0 [0.10, 261.34]

Figuras y tablas -
Comparison 6. DFP/DFX versus DFP/DFO