Scolaris Content Display Scolaris Content Display

Intervenciones para mejorar la adherencia al tratamiento de quelación del hierro en personas con anemia drepanocítica o talasemia

Appendices

Appendix 1. Search strategies

CENTRAL (The Cochrane Library)
#1 MeSH descriptor: [Patient Acceptance of Health Care] explode all trees
#2 MeSH descriptor: [Patient Education as Topic] this term only
#3 MeSH descriptor: [Data Collection] explode all trees
#4 (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*):ti
#5 ((adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*) near/6 (patient* or treatment* or therapy or therapies or medication* or drug*)):ab
#6 (patient* near/3 (dropout* or drop* out*))
#7 MeSH descriptor: [Treatment Refusal] this term only
#8 (treatment* near/3 refus*)
#9 #1 or #2 or #3 or #4 or #5 or #6 or #7 or #8
#10 MeSH descriptor: [Iron Chelating Agents] explode all trees
#11 MeSH descriptor: [Chelation Therapy] this term only
#12 (chelat* near/3 (treatment* or therap*))
#13 (deferoxamine* or deferoximine* or deferrioxamine* or desferioximine* or desferrioxamine* or desferroxamine* or desferal* or desferral* or DFO or desferin* or desferol* or dfom)
#14 (deferiprone or L1* or kelfer or DMHP or ferriprox or CP20 or dmohpo or hdmpp CPD or hdpp)
#15 (exjade* or deferasirox* or ICL 670* or icl670* or "CGP 72670")
#16 (iron near/5 (chelat* or reduc*))
#17 #10 or #11 or #12 or #13 or #14 or #15 or #16
#18 MeSH descriptor: [Thalassemia] explode all trees
#19 (thalassemi* or thalassaemi* or lepore or hydrops fetalis)
#20 ((hemoglobin or haemoglobin) near/3 disease)
#21 (hemochromatosis or haemochromatosis or hemosiderosis or haemosiderosis)
#22 ((mediterranean or erythroblastic or cooley*) next (anemi* or anaemi*))
#23 MeSH descriptor: [Iron Overload] explode all trees
#24 (iron near/3 (overload* or over‐load*))
#25 MeSH descriptor: [Hemoglobinopathies] this term only
#26 MeSH descriptor: [Hemoglobin C Disease] this term only
#27 (hemoglobinopath* or haemoglobinopath*)
#28 MeSH descriptor: [Anemia, Sickle Cell] explode all trees
#29 (barts and (blood or plasma))
#30 (sickle cell or sicklemi* or sickled or sickling or meniscocyt* or drepanocyt*)
#31 (hemoglobin S or hemoglobin SC or hemoglobin SE or hemoglobin SS or hemoglobin C or hemoglobin D or
haemoglobin S or haemoglobin SC or haemoglobin SE or haemoglobin SS or haemoglobin C or haemoglobin D Hb S or Hb SC or Hb SE or Hb SS or Hb C or Hb D or SC disease)
#32 #18 or #19 or #20 or #21 or #22 or #23 or #24 or #25 or #26 or #27 or #28 or #29 or #30 or #31
#33 #9 and #17 and #32
#34 ((thalassemi* or thalassaemi* or sickle or hemoglobinopath* or haemoglobinopath*) and (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or educat*)):ti
#35 #33 or #34

PubMed (for Epub Ahead of Print, In‐Process & Other Non‐Indexed Citations only)
#1 ((adher* OR nonadher* OR complian* OR comply* OR noncomplian* OR noncomply* OR complier* OR noncomplier* OR accept* OR nonaccept* OR abandon* OR co‐operat* OR cooperat* OR unco‐operative* OR uncooperative* OR nonco‐operat* OR noncooperat* OR satisfaction OR dissatisfaction OR persist* OR educat* OR questionnaire*) AND (patient OR patients OR treatment* OR therapy OR therapies OR medication* OR drug*))
#2 (patient dropout* OR patient drop* outs OR patients drop* out OR treatment* refus* OR refus* treatment*)
#3 #1 OR #2
#4 (deferoxamine* OR deferoximine* OR deferrioxamine* OR desferioximine* OR desferrioxamine* OR desferroxamine* OR desferal* OR desferral* OR DFO OR desferin* OR desferol* OR dfom OR deferiprone OR L1 OR kelfer OR DMHP OR ferriprox OR CP20 OR dmohpo OR hdmpp CPD OR hdpp OR exjade* OR deferasirox* OR ICL 670* OR icl670* OR CGP "72670" OR iron chelat* OR iron reduc* OR chelat* treatment* OR chelat* therapy)
#5 (thalassemi* OR thalassaemi* OR lepore OR hydrops fetalis OR cooley* anemi* OR cooley* anaemi*)
#6 (hemoglobin disease OR haemoglobin disease OR hemochromatosis OR haemochromatosis OR hemosiderosis OR haemosiderosis)
#7 (mediterranean anemi* OR mediterranean anaemi* OR erythroblastic anemi* OR erythroblastic anaemi*)
#8 hemoglobinopath* OR haemoglobinopath* OR iron overload* OR iron over‐load*
#9 ("sickle cell" OR sicklemi* OR sickled OR sickling OR meniscocyt* OR drepanocyt* OR "hemoglobin S" OR "hemoglobin SC" OR "hemoglobin SE" OR "hemoglobin SS" OR "hemoglobin C" OR "hemoglobin D" OR "haemoglobin S" OR "haemoglobin SC" OR "haemoglobin SE" OR "haemoglobin SS" OR "haemoglobin C" OR "haemoglobin D" OR "Hb S" OR "Hb SC" OR "Hb SE" OR "Hb SS" OR "Hb C" OR "Hb D" OR "SC disease")
#10 #5 OR #6 OR #7 OR #8 OR #9
#11 #3 AND 4 AND #10
#12 ((adher*[TI] OR nonadher*[TI] OR complian*[TI] OR comply*[TI] OR noncomplian*[TI] OR noncomply*[TI] OR complier*[TI] OR noncomplier*[TI] OR accept*[TI] OR nonaccept*[TI] OR abandon*[TI] OR co‐operat*[TI] OR cooperat*[TI] OR unco‐operative*[TI] OR uncooperative*[TI] OR nonco‐operat*[TI] OR noncooperat*[TI] OR satisfaction[TI] OR dissatisfaction[TI] OR persist*[TI] OR educat*[TI] OR questionnaire*[TI]) AND (thalassemia*[TI] OR thalassaemia*[TI] OR sickle[TI] OR iron overload*[TI]))
#13 #11 OR #12
#14 (publisher[sb] OR inprocess[sb] OR pubmednotmedline[sb])
#15 #13 AND #14

MEDLINE (Ovid)
1. exp "Patient Acceptance of Health Care"/
2. (px or ed).fs.
3. "Patient Education as Topic"/
4. exp Data Collection/
5. (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*).ti.
6. ((adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*) adj6 (patient* or treatment* or therapy or therapies or medication* or drug*)).ab,kf.
7. (patient* adj3 (dropout* or drop* out*)).tw,kf.
8. Treatment Refusal/
9. (treatment* adj3 refus*).tw,kf.
10. or/1‐9
11. exp IRON CHELATING AGENTS/
12. CHELATION THERAPY/
13. (chelation adj3 (treatment* or therap*)).tw,kf.
14. (deferoxamine* or deferoximine* or deferrioxamine* or desferioximine* or desferrioxamine* or desferroxamine* or desferal* or desferral* or DFO or desferin* or desferol* or dfom).mp.
15. (deferiprone or L1* or kelfer or DMHP or ferriprox or CP20 or dmohpo or hdmpp CPD or hdpp).mp.
16. (exjade* or deferasirox* or ICL 670* or icl670* or "CGP 72670").mp.
17. (iron adj5 (chelat* or reduc*)).tw,kf.
18. or/11‐17
19. exp THALASSEMIA/
20. (thalass?emi* or lepore or hydrops fetalis).tw,kf.
21. ((hemoglobin or haemoglobin) adj3 disease).tw,kf.
22. (hemochromatosis or haemochromatosis or hemosiderosis or haemosiderosis).tw,kf.
23. ((mediterranean or erythroblastic or cooley*) adj (anemi* or anaemi*)).tw,kf.
24. exp IRON OVERLOAD/
25. (iron adj3 (overload* or over‐load*)).tw,kf.
26. exp HEMOGLOBINOPATHIES/
27. exp HEMOGLOBIN, SICKLE/
28. (hemoglobinopath* or haemoglobinopath*).tw,kf.
29. exp ANEMIA, SICKLE CELL/
30. (barts and (blood or plasma)).tw,kf.
31. (sickle or sicklemi* or sickled or sickling or meniscocyt* or drepanocyt*).tw,kf.
32. (h?emoglobin s or h?emoglobin sc or h?emoglobin se or h?emoglobin ss or h?emoglobin c or h?emoglobin d or Hb s or Hb sc or Hb se or Hb ss or Hb c or Hb d or sc disease*).tw,kf.
33. or/19‐32
34. 10 and 18 and 33
35. exp *Hemoglobinopathies/ or (thalass?emi* or sickle or hemoglobinopath* or haemoglobinopath*).ti.
36. exp *Patient Compliance/ or (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or educat*).ti.
37. 35 and 36
38. 34 or 37

Embase (Ovid)
1. exp THALASSEMIA/
2. (thalass?emi* or lepore or hydrops fetalis).tw,kf.
3. ((hemoglobin or haemoglobin) adj3 disease).tw,kf.
4. (hemochromatosis or haemochromatosis or hemosiderosis or haemosiderosis).tw,kf.
5. ((mediterranean or erythroblastic or cooley*) adj (anemi* or anaemi*)).tw,kf.
6. IRON OVERLOAD/
7. (iron adj3 (overload* or over‐load*)).tw,kf.
8. HEMOGLOBINOPATHY/
9. HEMOGLOBIN S/
10. (hemoglobinopath* or haemoglobinopath*).tw,kf.
11. exp SICKLE CELL ANEMIA/
12. (barts and (blood or plasma)).tw,kf.
13. (sickle or sicklemi* or sickled or sickling or meniscocyt* or drepanocyt*).tw,kf.
14. (h?emoglobin s or h?emoglobin sc or h?emoglobin se or h?emoglobin ss or h?emoglobin c or h?emoglobin d or Hb s or Hb sc or Hb se or Hb ss or Hb c or Hb d or sc disease*).tw,kf.
15. or/1‐14
16. exp PATIENT ATTITUDE/
17. PATIENT EDUCATION/
18. "PATIENT EDUCATION AS TOPIC"/
19. exp DATA COLLECTION METHOD/
20. (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*).ti.
21. ((adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*) adj6 (patient* or treatment* or therapy or therapies or medication* or drug*)).ab,kf.
22. (patient* adj3 (dropout* or drop* out*)).tw.
23. (treatment* adj3 refus*).tw.
24. or/16‐23
25. IRON CHELATING AGENT/
26. CHELATION THERAPY/
27. (chelation adj3 (treatment* or therap*)).tw,kf.
28. (deferoxamine* or deferoximine* or deferrioxamine* or desferioximine* or desferrioxamine* or desferroxamine* or desferal* or desferral* or DFO or desferin* or desferol* or dfom).mp.
29. (deferiprone or L1* or kelfer or DMHP or ferriprox or cp20 or dmohpo or hdmpp CPD or hdpp).mp.
30. (exjade* or deferasirox* or (icl adj 670*) or icl670* or (cgp adj "72670")).mp.
31. (iron adj5 (chelat* or reduc*)).tw.
32. or/25‐31
33. 15 and 24 and 32
34. exp *Hemoglobinopathy/ or (thalass?emi* or sickle or hemoglobinopath* or haemoglobinopath*).ti.
35. exp *Patient Compliance/ or (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or educat*).ti.
36. 34 and 35
37. 33 or 36

CINAHL (EBSCOHost)
S1 (MH "Patient Compliance+")
S2 (MH "Patient Education")
S3 (MH "Instrument by Type+")
S4 TI (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*)
S5 AB ((adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*) N6 (patient* or treatment* or therapy or therapies or medication* or drug*))
S6 TX (patient* N3 (dropout* or drop* out*))
S7 MH Treatment Refusal
S8 TX (treatment* N3 refus*)
9 S1 OR S2 OR S3 OR S4 OR S5 OR S6 OR S7 OR S8
S10 (MH "Chelating Agents+")
S11 (MH "Chelation Therapy")
S12 TX (deferoxamine* or deferoximine* or deferrioxamine* or desferioximine* or desferrioxamine* or desferroxamine* or desferal* or desferral* or DFO or desferin* or desferol* or dfom)
S13 TX (deferiprone or L1* or kelfer or DMHP or ferriprox or CP20 or dmohpo or hdmpp CPD or hdpp)
S14 TX (exjade* or deferasirox* or ICL 670* or icl670* or "CGP 72670")
S15 TX (iron N5 (chelat* or reduc*)) OR TX (chelat* N3 (treatment* or therap*))
S16 S10 OR S11 OR S12 OR S13 OR S14 OR S15
S17 (MH "Thalassemia+")
S18 TX (thalassemi* or thalassaemi* or lepore or hydrops fetalis)
S19 TX ((hemoglobin or haemoglobin) N3 disease)
S20 TX (hemochromatosis or haemochromatosis or hemosiderosis or haemosiderosis)
S21 TX ((mediterranean or erythroblastic or cooley*) N1 (anemi* or anaemi*))
S22 (MH "Iron Overload+")
S23 TX (iron N3 (overload* or over‐load*))
S24 (MH "Hemoglobinopathies")
S25 TX (hemoglobinopath* or haemoglobinopath*)
S26 (MH "Anemia, Sickle Cell+")
S27 TX (barts and (blood or plasma))
S28 TX (sickle OR sicklemi* OR sickled OR sickling OR meniscocyt* OR drepanocyt* OR "hemoglobin S" OR "hemoglobin SC" OR "hemoglobin SE" OR "hemoglobin SS" OR "hemoglobin C" OR "hemoglobin D" OR "haemoglobin S" OR "haemoglobin SC" OR "haemoglobin SE" OR "haemoglobin SS" OR "haemoglobin C" OR "haemoglobin D" OR "Hb S" OR "Hb SC" OR "Hb SE" OR "Hb SS" OR "Hb C" OR "Hb D" OR "SC disease")
S29 S17 OR S18 OR S19 OR S20 OR S21 OR S22 OR S23 OR S24 OR S25 OR S26 OR S27 OR S28
S30 S9 AND S16 AND S29
S31 (MM "Patient Compliance+")
S32 TI (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or educat*)
S33 S31 OR S32
S34 (MM "Hemoglobinopathies+")
S35 TI (thalassemi* or thalassaemi* or sickle or hemoglobinopath* or haemoglobinopath*)
S36 S34 OR S35
S37 S33 AND S36
S38 S30 OR S37

APA PsycInfo (Ovid)
1. Treatment Compliance/ or Treatment Dropouts/ or Treatment Refusal/ 
2. Treatment Termination/ 
3. Client Education/ 
4. Questionnaires/ or General Health Questionnaire/ 
5. (adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*).ti. 
6. ((adher* or nonadher* or complian* or comply* or noncomplian* or noncomply* or complier* or noncomplier* or accept* or nonaccept* or abandon* or co‐operat* or cooperat* or unco‐operative* or uncooperative* or nonco‐operat* or noncooperat* or satisfaction or dissatisfaction or persist* or educat* or questionnaire*) adj6 (patient* or treatment* or therapy or therapies or medication* or drug*)).ab. 
7. (patient* adj3 (dropout* or drop* out*)).tw. 
8. (treatment* adj3 refus*).tw. 
9. or/1‐8 
10. Sickle Cell Disease/ 
11. (sickle or sicklemi* or sickled or sickling or meniscocyt* or drepanocyt*).tw. 
12. (h?emoglobin s or h?emoglobin sc or h?emoglobin se or h?emoglobin ss or h?emoglobin c or h?emoglobin d or Hb s or Hb sc or Hb se or Hb ss or Hb c or Hb d or sc disease*).tw. 
13. (thalass?emi* or lepore or hydrops fetalis).tw. 
14. ((hemoglobin or haemoglobin) adj3 disease).tw. 
15. (hemochromatosis or haemochromatosis or hemosiderosis or haemosiderosis).tw. 
16. ((mediterranean or erythroblastic or cooley*) adj (anemi* or anaemi*)).tw. 
17. (hemoglobinopath* or haemoglobinopath*).tw.
18. (iron adj3 (overload* or over‐load*)).tw.
19. (barts and (blood or plasma)).tw.
20. or/10‐19 
21. 9 and 20 

 

ProQuest Dissertations & Theses Global
ti(adher* OR nonadher* OR complian* OR comply* OR noncomplian* OR noncomply* OR complier* OR noncomplier* OR accept* OR nonaccept* OR abandon* OR co‐operat* OR cooperat* OR unco‐operative* OR uncooperative* OR nonco‐operat* OR noncooperat* OR satisfaction OR dissatisfaction OR refus* OR persist* OR educat* OR questionnaire*) AND ti(thalassemia OR thalassaemia OR sickle OR sickled OR sickling OR iron overload OR hemoglobinopath*) AND (chelation OR chelating OR deferiprone OR deferoxamine OR deferasirox OR DFO OR ferriprox OR exjade OR iron reduction)
 

Web of Science CPCI‐S & CPSSI
#1 TS=((adher* OR nonadher* OR complian* OR comply* OR noncomplian* OR noncomply* OR complier* OR noncomplier* OR accept* OR nonaccept* OR abandon* OR co‐operat* OR cooperat* OR unco‐operative* OR uncooperative* OR nonco‐operat* OR noncooperat* OR satisfaction OR dissatisfaction OR persist* OR educat* OR questionnaire*) AND (patient* OR treatment* OR therapy OR therapies OR medication* OR drug*))
#2 TS=(patient dropout* OR patient drop* outs OR patients drop* out OR treatment* refus* OR refus* treatment*)
#3 #1 OR #2
#4 TS=(deferoxamine* OR deferoximine* OR deferrioxamine* OR desferioximine* OR desferrioxamine* OR desferroxamine* OR desferal* OR desferral* OR DFO OR desferin* OR desferol* OR dfom OR deferiprone OR L1 OR kelfer OR DMHP OR ferriprox OR CP20 OR dmohpo OR hdmpp CPD OR hdpp OR exjade* OR deferasirox* OR ICL 670* OR icl670* OR CGP "72670" OR iron chelat* OR iron reduc* OR chelat* treatment* OR chelat* therap*)
#5 TS=(thalassemi* OR thalassaemi* OR lepore OR hydrops fetalis OR cooley* anemi* OR cooley* anaemi* OR hemoglobin disease OR haemoglobin disease OR hemochromatosis OR haemochromatosis OR hemosiderosis OR haemosiderosis OR mediterranean anemi* OR mediterranean anaemi* OR erythroblastic anemi* OR erythroblastic anaemi* OR iron overload* OR iron over‐load* OR hemoglobinopath* OR haemoglobinopath*)
#6 TS=(sickle OR sicklemi* OR sickled OR sickling OR meniscocyt* OR drepanocyt* OR "hemoglobin S" OR "hemoglobin SC" OR "hemoglobin SE" OR "hemoglobin SS" OR "hemoglobin C" OR "hemoglobin D" OR "haemoglobin S" OR "haemoglobin SC" OR "haemoglobin SE" OR "haemoglobin SS" OR "haemoglobin C" OR "haemoglobin D" OR "Hb S" OR "Hb SC" OR "Hb SE" OR "Hb SS" OR "Hb C" OR "Hb D" OR "SC disease")
#7 #5 OR #6
#8 #3 AND #4 AND #7
 

ClinicalTrials.gov
Other Terms: (thalassemia OR sickle cell anemia OR iron overload OR hemoglobinopathies) AND (iron chelation OR chelation therapy OR deferiprone OR deferoxamine OR deferasirox OR DFO OR iron reduction)

WHO ICTRP
Condition: thalassemia OR sickle cell anemia OR iron overload OR hemoglobinopathies
Intervention: iron chelation OR chelation therapy OR deferiprone OR deferoxamine OR deferasirox OR DFO OR iron reduction

ISRCTN
Condition: thalassemia OR sickle cell anemia OR iron overload OR hemoglobinopathies
Interventions: iron chelation OR chelation therapy OR deferiprone OR deferoxamine OR deferasirox OR DFO OR iron reduction

Appendix 2. The Risk Of Bias In Non‐randomised Studies of Interventions (ROBINS‐I) assessment tool

ROBINS‐I tool (Stage I)

Specify the review question

Participants

Experimental intervention

Control intervention

Outcomes

The ROBINS‐I tool (Stage II): For each study

Specify a target trial specific to the study.

Design

Individually randomised or cluster randomised or matched

Participants

Experimental intervention

Control intervention

Is your aim for this study...?

□ to assess the effect of initiating intervention (as in an intention‐to‐treat analysis)

□ to assess the effect of initiating and adhering to intervention (as in a per protocol analysis)

Specify the outcome

Specify which outcome is being assessed for risk of bias (typically from among those earmarked for the Summary of Findings table). Specify whether this is a proposed benefit or harm of intervention.

Specify the numerical result being assessed

In case of multiple alternative analyses being presented, specify the numeric result (e.g. RR = 1.52 (95% CI 0.83 to 2.77) or a reference (e.g. to a table, figure or paragraph) that uniquely defines the result being assessed (or both).

Preliminary consideration of confounders

Complete a row for each important confounding area (i) listed in the review protocol; and (ii) relevant to the setting of this particular study, or which the study authors identified as potentially important.

'Important' confounding areas are those for which, in the context of this study, adjustment is expected to lead to a clinically important change in the estimated effect of the intervention. 'Validity' refers to whether the confounding variable or variables fully measure the area, while 'reliability' refers to the precision of the measurement (more measurement error means less reliability).

(i) Confounding areas listed in the review protocol

Confounding area

Measured variable(s)

Is there evidence that controlling for this variable was unnecessary?*

Is the confounding area measured validly and reliably by this variable (or these variables)?

OPTIONAL: is adjusting for this variable (alone) expected to favour the experimental or the control group?

Yes / No / No information

Favour intervention / Favour control / No information

(ii) Additional confounding areas relevant to the setting of this particular study, or which the study authors identified as important

Confounding area

Measured Variable(s)

Is there evidence that controlling for this variable was unnecessary?*

Is the confounding area measured validly and reliably by this variable (or these variables)?

OPTIONAL: is adjusting for this variable (alone) expected to favour the experimental or the control group?

Yes / No / No information

Favour intervention / Favour control / No information

* In the context of a particular study, variables can be demonstrated not to be confounders and so not included in the analysis: (a) if they are not predictive of the outcome; (b) if they are not predictive of intervention; or (c) because adjustment makes no or minimal difference to the estimated effect of the primary parameter. Note that “no statistically significant association” is not the same as “not predictive”.

Preliminary consideration of co‐interventions

Complete a row for each important co‐intervention (i) listed in the review protocol; and (ii) relevant to the setting of this particular study, or which the study authors identified as important.

'Important' co‐interventions are those for which, in the context of this study, adjustment is expected to lead to a clinically important change in the estimated effect of the intervention.

(i) Co‐interventions listed in the review protocol

Co‐intervention

Is there evidence that controlling for this co‐intervention was unnecessary (e.g. because it was not administered)?

Is presence of this co‐intervention likely to favour outcomes in the experimental or the control group

Favour experimental / Favour comparator / No information

Favour experimental / Favour comparator / No information

Favour experimental / Favour comparator / No information

(ii) Additional co‐interventions relevant to the setting of this particular study, or which the study authors identified as important

Co‐intervention

Is there evidence that controlling for this co‐intervention was unnecessary (e.g. because it was not administered)?

Is presence of this co‐intervention likely to favour outcomes in the experimental or the control group

Favour experimental / Favour comparator / No information

Favour experimental / Favour comparator / No information

Favour experimental / Favour comparator / No information

Risk of bias assessment (cohort‐type studies)

Bias domain

Signalling questions

Elaboration

Response options

Bias due to confounding

1.1 Is there potential for confounding of the effect of intervention in this study?

IfN or PN to1.1: the study can be considered to be at low risk of bias due to confounding and no further signalling questions need be considered

In rare situations, such as when studying harms that are very unlikely to be related to factors that influence treatment decisions, no confounding is expected and the study can be considered to be at low risk of bias due to confounding, equivalent to a fully randomised trial.

There is no NI (No information) option for this signalling question.

Y / PY / PN / N

If Y or PY to 1.1: determine whether there is a need to assess time‐varying confounding:

1.2. Was the analysis based on splitting participants’ follow up time according to intervention received?

If N orPN, answer questions relating to baseline confounding (1.4 to 1.6)

If Y orPY, proceed to question 1.3.

If participants could switch between intervention groups then associations between intervention and outcome may be biased by time‐varying confounding. This occurs when prognostic factors influence switches between intended interventions.

NA / Y / PY / PN / N / NI

1.3. Were intervention discontinuations or switches likely to be related to factors that are prognostic for the outcome?

If N or PN, answer questions relating to baseline confounding (1.4 to 1.6)

If Y orPY, answer questions relating to both baseline and time‐varying confounding (1.7 and 1.8)

If intervention switches are unrelated to the outcome, for example when the outcome is an unexpected harm, then time‐varying confounding will not be present and only control for baseline confounding is required.

NA / Y / PY / PN / N / NI

Questions relating to baseline confounding only

1.4. Did the authors use an appropriate analysis method that controlled for all the important confounding areas?

Appropriate methods to control for measured confounders include stratification, regression, matching, standardization, and inverse probability weighting. They may control for individual variables or for the estimated propensity score. Inverse probability weighting is based on a function of the propensity score. Each method depends on the assumption that there is no unmeasured or residual confounding.

NA / Y / PY / PN / N / NI

1.5.If Y or PY to1.4: were confounding areas that were controlled for measured validly and reliably by the variables available in this study?

Appropriate control of confounding requires that the variables adjusted for are valid and reliable measures of the confounding domains. For some topics, a list of valid and reliable measures of confounding domains will be specified in the review protocol but for others such a list may not be available. Study authors may cite references to support the use of a particular measure. If authors control for confounding variables with no indication of their validity or reliability pay attention to the subjectivity of the measure. Subjective measures (e.g. based on self‐report) may have lower validity and reliability than objective measures such as lab findings.

NA / Y / PY / PN / N / NI

1.6. Did the authors control for any post‐intervention variables?

Controlling for post‐intervention variables is not appropriate. Controlling for mediating variables estimates the direct effect of intervention and may introduce confounding. Controlling for common effects of intervention and outcome causes bias.

NA / Y / PY / PN / N / NI

Questions relating to baseline and time‐varying confounding

1.7. Did the authors use an appropriate analysis method that adjusted for all the important confounding areas and for time‐varying confounding?

Adjustment for time‐varying confounding is necessary to estimate per‐protocol effects in both randomised trials and NRSI. Appropriate methods include those based on inverse‐probability weighting. Standard regression models that include time‐updated confounders may be problematic if time‐varying confounding is present.

NA / Y / PY / PN / N / NI

1.8. IfY orPY to1.7: Were confounding areas that were adjusted for measured validly and reliably by the variables available in this study?

See 1.5 above.

NA / Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ no confounding expected.

Low / Moderate / Serious / Critical / NI

Moderate ‐ confounding expected, all known important confounding domains appropriately measured and controlled for;

and

Reliability and validity of measurement of important domains were sufficient, such that we do not expect serious residual confounding.

Serious ‐ at least one known important domain was not appropriately measured, or not controlled for;

or

Reliability or validity of measurement of a important domain was low enough that we expect serious residual confounding.

Critical ‐ confounding inherently not controllable, or the use of negative controls strongly suggests unmeasured confounding.

Optional: what is the predicted direction of bias due to confounding?

Can the true effect estimate be predicted to be greater or less than the estimated effect in the study because one or more of the important confounding domains was not controlled for? Answering this question will be based on expert knowledge and results in other studies and therefore can only be completed after all of the studies in the body of evidence have been reviewed. Consider the potential effect of each of the unmeasured domains and whether all important confounding domains not controlled for in the analysis would be likely to change the estimate in the same direction, or if one important confounding domain that was not controlled for in the analysis is likely to have a dominant impact.

Favours experimental / Favours comparator / Unpredictable

Bias in selection of participants into the study

2.1. Was selection of participants into the study (or into the analysis) based on participant characteristics observed after the start of intervention?

This domain is concerned only with selection into the study based on participant characteristics observed after the start of intervention. Selection based on characteristics observed before the start of intervention can be addressed by controlling for imbalances between intervention and control groups in baseline characteristics that are prognostic for the outcome (baseline confounding).

Y / PY / PN / N / NI

IfN orPN to2.1: go to 2.4

2.2. IfY orPY to2.1: were the post‐intervention variables that influenced selection likely to be associated with intervention

Selection bias occurs when selection is related to an effect of either intervention or a cause of intervention and an effect of either the outcome or a cause of the outcome. Therefore, the result is at risk of selection bias if selection into the study is related to both the intervention and the outcome.

NA / Y / PY / PN / N / NI

2.3 If Y orPY to2.2: were the post‐intervention variables that influenced selection likely to be influenced by the outcome or a cause of the outcome?

NA / Y / PY / PN / N / NI

2.4. Do start of follow up and start of intervention coincide for most participants?

If participants are not followed from the start of the intervention then a period of follow up has been excluded, and individuals who experienced the outcome soon after intervention will be missing from analyses. This problem may occur when prevalent, rather than new (incident), users of the intervention are included in analyses.

Y / PY / PN / N / NI

2.5. IfY orPY to2.2 and2.3, or N orPN to 2.4: were adjustment techniques used that are likely to correct for the presence of selection biases?

It is in principle possible to correct for selection biases, for example by using inverse probability weights to create a pseudo‐population in which the selection bias has been removed, or by modelling the distributions of the missing participants or follow up times and outcome events and including them using missing data methodology. However such methods are rarely used and the answer to this question will usually be “No”

NA / Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ all participants who would have been eligible for the target trial were included in the study and start of follow up and start of intervention coincide for all subjects.

Low / Moderate / Serious / Critical / NI

Moderate ‐ selection into the study may have been related to intervention and outcome, but the authors used appropriate methods to adjust for the selection bias; or Start of follow up and start of intervention do not coincide for all participants, but (a) the proportion of participants for which this was the case was too low to induce important bias; (b) the authors used appropriate methods to adjust for the selection bias; or (c) the review authors are confident that the rate (hazard) ratio for the effect of intervention remains constant over time.

Serious ‐ selection into the study was related to intervention and outcome;

or

Start of follow up and start of intervention do not coincide, and a potentially important amount of follow‐up time is missing from analyses, and the rate ratio is not constant over time.

Critical ‐ selection into the study was strongly related to intervention and outcome;

or

A substantial amount of follow‐up time is likely to be missing from analyses, and the rate ratio is not constant over time.

Optional: what is the predicted direction of bias due to selection of participants into the study?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Bias in classification of interventions

3.1 Were intervention groups clearly defined?

A pre‐requisite for an appropriate comparison of interventions is that the interventions are well defined. Ambiguity in the definition may lead to bias in the classification of participants. For individual‐level interventions, criteria for considering individuals to have received each intervention should be clear and explicit, covering issues such as type, setting, dose, frequency, intensity and/or timing of intervention. For population‐level interventions (e.g. measures to control air pollution), the question relates to whether the population is clearly defined, and the answer is likely to be ‘Yes’.

Y / PY / PN / N / NI

3.2 Was the information used to define intervention groups recorded at the start of the intervention?

In general, if information about interventions received is available from sources that could not have been affected by subsequent outcomes, then differential misclassification of intervention status is unlikely. Collection of the information at the time of the intervention makes it easier to avoid such misclassification. For population‐level interventions (e.g. measures to control air pollution), the answer to this question is likely to be ‘Yes’.

Y / PY / PN / N / NI

3.3 Could classification of intervention status have been affected by knowledge of the outcome or risk of the outcome?

Collection of the information at the time of the intervention may not be sufficient to avoid bias. The way in which the data are collected for the purposes of the NRSI should also avoid misclassification.

Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ intervention status is well defined and based solely on information collected at the time of intervention.

Low / Moderate / Serious / Critical / NI

Moderate ‐ intervention status is well defined but some aspects of the assignments of intervention status were determined retrospectively

Serious ‐ intervention status is not well defined, or major aspects of the assignments of intervention status were determined in a way that could have been affected by knowledge of the outcome.

Critical ‐ (unusual) An extremely high amount of misclassification of intervention status, e.g. because of unusually strong recall biases.

Optional: what is the predicted direction of bias due to measurement of outcomes or interventions?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Bias due to departures from intended interventions

4.1. Was the intervention implemented successfully for most participants?

Consider the success of implementation of the intervention in the context of its complexity. Was recommended practice followed by those administering the intervention?

Y / PY / PN / N / NI

If your aim for this study is to assess the effect of initiating and adhering to intervention (as in a per‐protocol analysis), answer questions 4.2 to 4.4

4.2. Did study participants adhere to the assigned intervention regimen?

Lack of adherence to assigned intervention includes cessation of intervention, crossovers to the comparator intervention and switches to another active intervention. We distinguish between analyses where:

(1) intervention switches led to follow up time being assigned to the new intervention; and

(2) intervention switches (including cessation of intervention) where follow up time remained allocated to the original intervention;

(3) is addressed under time‐varying confounding, and should not be considered further here.

Consider available information on the proportion of study participants who continued with their assigned intervention throughout follow up. Was lack of adherence sufficient to impact the intervention effect estimate?

NA/ Y / PY / PN / N / NI

4.3. Were important co‐interventions balanced across intervention groups?

Consider the co‐interventions that are likely to affect the outcome and to have been administered in the context of this study, based on the preliminary consideration of co‐interventions and available literature. Consider whether these co‐interventions are balanced between intervention groups.

NA/ Y / PY / PN / N / NI

4.4. IfN orPN to4.1, 4.2 or4.3: were adjustment techniques used that are likely to correct for these issues?

Such adjustment techniques include inverse‐probability weighting to adjust for censoring at deviation from intended intervention, or inverse probability weighting of marginal structural models to adjust for time‐varying confounding. Specialist advice may be needed to assess studies that used these approaches.

NA / Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ no bias due to deviation from the intended intervention is expected, for example if both the intervention and comparator are implemented over a short time period, and subsequent interventions are part of routine medical care, or if the specified comparison relates to initiation of intervention regardless of whether it is continued.

Low / Moderate / Serious / Critical / NI

Moderate ‐ bias due to deviation from the intended intervention is expected, and switches, co‐interventions, and some problems with intervention fidelity are appropriately measured and adjusted for in the analyses. Alternatively, most (but not all) deviations from intended intervention reflect the natural course of events after initiation of intervention.

Serious ‐ switches in treatment, co‐interventions, or problems with implementation fidelity are apparent and are not adjusted for in the analyses.

Critical ‐ substantial deviations from the intended intervention are present and are not adjusted for in the analysis.

Optional: what is the predicted direction of bias due to departures from the intended interventions?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Bias due to missing data

5.1 Were there missing outcome data?

This aims to elicit whether the proportion of missing observations is likely to result in missing information that could substantially impact our ability to answer the question being addressed. Guidance will be needed on what is meant by ‘reasonably complete’. One aspect of this is that review authors would ideally try and locate an analysis plan for the study.

Y / PY / PN / N / NI

5.2 Were participants excluded due to missing data on intervention status?

Missing intervention status may be a problem. This requires that the intended study sample is clear, which it may not be in practice.

Y / PY / PN / N / NI

5.3 Were participants excluded due to missing data on other variables needed for the analysis?

This question relates particularly to participants excluded from the analysis because of missing information on confounders that were controlled for in the analysis.

Y / PY / PN / N / NI

5.4 If Y orPY to 5.1, 5.2 or5.3: are the proportion of participants and reasons for missing data similar across interventions?

This aims to elicit whether either (i) differential proportion of missing observations or (ii) differences in reasons for missing observations could substantially impact on our ability to answer the question being addressed.

NA / Y / PY / PN / N / NI

5.5If Y or PY to5.1, 5.2 or5.3: were appropriate statistical methods used to account for missing data?

It is important to assess whether assumptions employed in analyses are clear and plausible. Both content knowledge and statistical expertise will often be required for this. For instance, use of a statistical method such as multiple imputation does not guarantee an appropriate answer. Review authors should seek naïve (complete‐case) analyses for comparison, and clear differences between complete‐case and multiple imputation‐based findings should lead to careful assessment of the validity of the methods used.

NA / Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ data were reasonably complete; or Proportions of and reasons for missing participants were similar across intervention groups; or Analyses that addressed missing data are likely to have removed any risk of bias.

Low / Moderate / Serious / Critical / NI

Moderate ‐ proportions of missing participants differ across interventions; or Reasons for missingness differ minimally across interventions; and Missing data were not addressed in the analysis.

Serious ‐ proportions of missing participants differ substantially across interventions; or Reasons for missingness differ substantially across interventions; and Missing data were addressed inappropriately in the analysis; or The nature of the missing data means that the risk of bias cannot be removed through appropriate analysis.

Critical ‐ (unusual) There were critical differences between interventions in participants with missing data that were not, or could not, be addressed through appropriate analysis.

Optional: what is the predicted direction of bias due to missing data?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Bias in measurement of outcomes

6.1 Could the outcome measure have been influenced by knowledge of the intervention received?

Some outcome measures involve negligible assessor judgment, e.g. all‐cause mortality or non‐repeatable automated laboratory assessments. Risk of bias due to measurement of these outcomes would be expected to be low.

Y / PY / PN / N / NI

6.2 Were outcome assessors aware of the intervention received by study participants?

If outcome assessors were blinded to intervention status, the answer to this question would be ‘No’. In other situations, outcome assessors may be unaware of the interventions being received by participants despite there being no active blinding by the study investigators; the answer this question would then also be ‘No’. In studies where participants report their outcomes themselves, for example in a questionnaire, the outcome assessor is the study participant. In an observational study, the answer to this question will usually be ‘Yes’ when the participants report their outcomes themselves.

Y / PY / PN / N / NI

6.3 Were the methods of outcome assessment comparable across intervention groups?

Comparable assessment methods (i.e. data collection) would involve the same outcome detection methods and thresholds, same time point, same definition, and same measurements

Y / PY / PN / N / NI

6.4 Were any systematic errors in measurement of the outcome related to intervention received?

This question refers to differential misclassification of outcomes. Systematic errors in measuring the outcome, if present, could cause bias if they are related to intervention or to a confounder of the intervention‐outcome relationship. This will usually be due either to outcome assessors being aware of the intervention received or to non‐comparability of outcome assessment methods, but there are examples of differential misclassification arising despite these controls being in place.

Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ the methods of outcome assessment were comparable across intervention groups;

and

The outcome measure was unlikely to be influenced by knowledge of the intervention received by study participants (i.e. is objective) or the outcome assessors were unaware of the intervention received by study participants;

and

Any error in measuring the outcome is unrelated to intervention status.

Low / Moderate / Serious / Critical / NI

Moderate ‐ the methods of outcome assessment were comparable across intervention groups;

and

The outcome measure is only minimally influenced by knowledge of the intervention received by study participants;

and

Any error in measuring the outcome is only minimally related to intervention status.

Serious ‐ the methods of outcome assessment were not comparable across intervention groups;

or

The outcome measure was subjective (i.e. likely to be influenced by knowledge of the intervention received by study participants) and was assessed by outcome assessors aware of the intervention received by study participants;

or

Error in measuring the outcome was related to intervention status.

Critical ‐ the methods of outcome assessment were so different that they cannot reasonably be compared across intervention groups.

Optional: what is the predicted direction of bias due to measurement of outcomes?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Bias in selection of the reported result

Is the reported effect estimate unlikely to be selected, on the basis of the results, from...

7.1. ... multiple outcome measurements within the outcome domain?

For a specified outcome domain, it is possible to generate multiple effect estimates for different measurements. If multiple measurements were made, but only one or a subset is reported, there is a risk of selective reporting on the basis of results.

Y / PY / PN / N / NI

7.2 ... multiple analyses of the intervention‐outcome relationship?

Because of the limitations of using data from non‐randomized studies for analyses of effectiveness (need to control confounding, substantial missing data, etc), analysts may implement different analytic methods to address these limitations. Examples include unadjusted and adjusted models; use of final value vs change from baseline vs analysis of covariance; different transformations of variables; a continuously scaled outcome converted to categorical data with different cutpoints; different sets of covariates used for adjustment; and different analytic strategies for dealing with missing data. Application of such methods generates multiple effect estimates for a specific outcome metric. If the analyst does not prespecify the methods to be applied, and multiple estimates are generated but only one or a subset is reported, there is a risk of selective reporting on the basis of results.

Y / PY / PN / N / NI

7.3 ... different subgroups?

Particularly with large cohorts often available from routine data sources, it is possible to generate multiple effect estimates for different subgroups or simply to omit varying proportions of the original cohort. If multiple estimates are generated but only one or a subset is reported, there is a risk of selective reporting on the basis of results.

Y / PY / PN / N / NI

Risk of bias judgement

Low ‐ there is clear evidence (usually through examination of a pre‐registered protocol or statistical analysis plan) that all reported results correspond to all intended outcomes, analyses and sub‐cohorts.

Low / Moderate / Serious / Critical / NI

Moderate ‐ the outcome measurements and analyses are consistent with an apriori plan;

or

are clearly defined and both internally and externally consistent;

and

there is no indication of selection of the reported analysis from among multiple analyses;

and

there is no indication of selection of the cohort or subgroups for analysis and reporting on the basis of the results.

Serious ‐ outcome measurements or analyses are internally or externally inconsistent; or There is a high risk of selective reporting from among multiple analyses; or The cohort or subgroup is selected from a larger study for analysis and appears to be reported on the basis of the results.

Critical ‐ there is evidence or strong suspicion of selective reporting of results, and the unreported results are likely to be substantially different from the reported results.

Optional: What is the predicted direction of bias due to selection of the reported result?

If the likely direction of bias can be predicted, it is helpful to state this. The direction might be characterized either as being towards (or away from) the null, or as being in favour of one of the interventions.

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

Overall bias

Risk of bias judgement

Low ‐ the study is judged to be at low risk of bias for all domains.

Low / Moderate / Serious / Critical / NI

Moderate ‐ the study is judged to be at low or moderate risk of bias for all domains.

Serious ‐ the study is judged to be at serious risk of bias in at least one domain, but not at critical risk of bias in any domain.

Critical ‐ the study is judged to be at critical risk of bias in at least one domain.

No information ‐ there is no clear indication that the study is at serious or critical risk of bias and there is a lack of information in one or more key domains of bias (a judgement is required for this).

Optional:

what is the overall predicted direction of bias for this outcome?

Favours experimental / Favours comparator / Towards null /Away from null / Unpredictable

CFGD trials register: Cochrane Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register 

Figuras y tablas -
Figure 1

CFGD trials register: Cochrane Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register 

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: Total SAEs (from therapy, disease, non‐adherence)

Figuras y tablas -
Analysis 1.2

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

Comparison 1: DFP versus DFO, Outcome 3: Other SAEs (from therapy, disease, non‐adherence)

Figuras y tablas -
Analysis 1.3

Comparison 1: DFP versus DFO, Outcome 3: Other SAEs (from therapy, disease, non‐adherence)

Comparison 1: DFP versus DFO, Outcome 4: All‐cause mortality

Figuras y tablas -
Analysis 1.4

Comparison 1: DFP versus DFO, Outcome 4: All‐cause mortality

Comparison 1: DFP versus DFO, Outcome 5: Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L)

Figuras y tablas -
Analysis 1.5

Comparison 1: DFP versus DFO, Outcome 5: Iron overload: defined as proportion of participants with serum ferritin ≥ 800 (µg/L)

Comparison 1: DFP versus DFO, Outcome 6: Organ damage

Figuras y tablas -
Analysis 1.6

Comparison 1: DFP versus DFO, Outcome 6: Organ damage

Comparison 1: DFP versus DFO, Outcome 7: AEs related to iron chelation

Figuras y tablas -
Analysis 1.7

Comparison 1: DFP versus DFO, Outcome 7: 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 (thalassaemia)

Figuras y tablas -
Analysis 2.2

Comparison 2: DFX versus DFO, Outcome 2: SAEs (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 3: SAEs (sickle cell disease)

Figuras y tablas -
Analysis 2.3

Comparison 2: DFX versus DFO, Outcome 3: SAEs (sickle cell disease)

Comparison 2: DFX versus DFO, Outcome 4: All‐cause mortality (thalassaemia)

Figuras y tablas -
Analysis 2.4

Comparison 2: DFX versus DFO, Outcome 4: All‐cause mortality (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 5: Proportion of participants with iron overload (thalassaemia)

Figuras y tablas -
Analysis 2.5

Comparison 2: DFX versus DFO, Outcome 5: Proportion of participants with iron overload (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 6: Total AEs related to iron chelation ‐ (thalassaemia)

Figuras y tablas -
Analysis 2.6

Comparison 2: DFX versus DFO, Outcome 6: Total AEs related to iron chelation ‐ (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 7: Other AEs related to iron chelation ‐ (thalassaemia)

Figuras y tablas -
Analysis 2.7

Comparison 2: DFX versus DFO, Outcome 7: Other AEs related to iron chelation ‐ (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 8: Total AEs (thalassaemia)

Figuras y tablas -
Analysis 2.8

Comparison 2: DFX versus DFO, Outcome 8: Total AEs (thalassaemia)

Comparison 2: DFX versus DFO, Outcome 9: Other AEs related to iron chelation (SCD)

Figuras y tablas -
Analysis 2.9

Comparison 2: DFX versus DFO, Outcome 9: Other AEs related to iron chelation (SCD)

Comparison 3: DFP versus DFX, Outcome 1: Adherence to iron chelation (%, SD)

Figuras y tablas -
Analysis 3.1

Comparison 3: DFP versus DFX, Outcome 1: Adherence to iron chelation (%, SD)

Comparison 3: DFP versus DFX, Outcome 2: Total SAEs 

Figuras y tablas -
Analysis 3.2

Comparison 3: DFP versus DFX, Outcome 2: Total SAEs 

Comparison 3: DFP versus DFX, Outcome 3: SAE (chelation‐related) (n/N)

Figuras y tablas -
Analysis 3.3

Comparison 3: DFP versus DFX, Outcome 3: SAE (chelation‐related) (n/N)

Comparison 3: DFP versus DFX, Outcome 4: All‐cause mortality (n/N)

Figuras y tablas -
Analysis 3.4

Comparison 3: DFP versus DFX, Outcome 4: All‐cause mortality (n/N)

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1: Adherence to iron chelation therapy (n/N)

Figuras y tablas -
Analysis 4.1

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 1: Adherence to iron chelation therapy (n/N)

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2: Adherence to iron chelation therapy (%, SD)

Figuras y tablas -
Analysis 4.2

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 2: Adherence to iron chelation therapy (%, SD)

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3: Incidence of SAEs

Figuras y tablas -
Analysis 4.3

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 3: Incidence of SAEs

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4: All‐cause mortality

Figuras y tablas -
Analysis 4.4

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 4: All‐cause mortality

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5: Incidence of organ damage

Figuras y tablas -
Analysis 4.5

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 5: Incidence of organ damage

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 6: Total AEs related to iron chelation

Figuras y tablas -
Analysis 4.6

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 6: Total AEs related to iron chelation

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 7: Other AEs related to iron chelation

Figuras y tablas -
Analysis 4.7

Comparison 4: DFX film‐coated tablet versus DFX dispersible tablet, Outcome 7: Other AEs related to iron chelation

Comparison 5: DFP and DFO versus DFP, Outcome 1: Incidence of SAEs

Figuras y tablas -
Analysis 5.1

Comparison 5: DFP and DFO versus DFP, Outcome 1: Incidence of SAEs

Comparison 5: DFP and DFO versus DFP, Outcome 2: All‐cause mortality

Figuras y tablas -
Analysis 5.2

Comparison 5: DFP and DFO versus DFP, Outcome 2: All‐cause mortality

Comparison 5: DFP and DFO versus DFP, Outcome 3: Incidence of chelation therapy‐related AEs

Figuras y tablas -
Analysis 5.3

Comparison 5: DFP and DFO versus DFP, Outcome 3: Incidence of chelation therapy‐related AEs

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

Figuras y tablas -
Analysis 6.1

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

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 1: Adherence to iron chelation therapy rates

Figuras y tablas -
Analysis 7.1

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 1: Adherence to iron chelation therapy rates

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 2: Incidence of SAE

Figuras y tablas -
Analysis 7.2

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 2: Incidence of SAE

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 3: All‐cause mortality

Figuras y tablas -
Analysis 7.3

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 3: All‐cause mortality

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 4: Organ damage (serum creatinine (≥ 33%) above baseline on 2 consecutive occasions)

Figuras y tablas -
Analysis 7.4

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 4: Organ damage (serum creatinine (≥ 33%) above baseline on 2 consecutive occasions)

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 5: Total AEs related to iron chelation

Figuras y tablas -
Analysis 7.5

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 5: Total AEs related to iron chelation

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 6: Other AEs related to iron chelation

Figuras y tablas -
Analysis 7.6

Comparison 7: DFP and DFX versus DFP and DFO, Outcome 6: Other AEs related to iron chelation

Summary of findings 1. Summary of findings: Comparison 1 ‐ deferiprone (DFP) versus deferoxamine (DFO)

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 

(%, SD)

See comments.

612
(7 RCTs)

⨁◯◯◯
Very lowa,b,c

2 trials (unpooled) provided analysable data (%, SD); the remaining trials reported only as % (or narratively), with no error (SD, or otherwise) and have been presented in Table 1 separately to the analyses.

Total reported SAEs 

(from therapy, disease, non‐adherence)

184 per 1000

263 per 1000
(153 to 453)

RR 1.43
(0.83 to 2.46)

228
(1 RCT)

⨁◯◯◯
Very lowc,d

All‐cause mortality

75 per 1000

35 per 1000
(13 to 91)

RR 0.47
(0.18 to 1.21)

376
(3 RCTs)

⨁◯◯◯
Very lowa,c,e

In a fourth trial, no events occurred in either arm (Pennell 2006).

Sustained adherence

See comments.

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

QoL 
(assessed with CHQ‐50 and SF‐36)
Follow‐up mean 12 months

See comments.

(1 RCT)

⨁◯◯◯
Very lowd,f

Data presented in additional tables from a single trial (Kwiatkowski 2021). No significant between‐group change over time. Major bias due to missing data (over half) for outcomes (DFP: CHQ‐50 n = 60/152 and SF‐36 n = 35/152; DFO: CHQ‐50 n = 23/76 and SF‐36 n = 19/76).

 *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).

CHQ‐50: Child Health Questionnaire ‐ 50 items; CI: confidence interval; DFO: deferoxamine; DFP: deferiprone; MD: mean difference; QoL: quality of life; RCT: randomised controlled trial; RR: risk ratio; SAE: serious adverse event; SD: standard deviation; SF‐36: Short‐Form Questionnaire ‐ 36 items.

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.

Explanations

aWe downgraded the certainty of evidence once for risk of bias due to high or uncertain risk of bias in one or more domains.

bWe downgraded the certainty of evidence twice for inconsistency due to considerable heterogeneity in the comparison.

cWe downgraded the certainty of evidence twice for imprecision due to wide CIs and small sample size (not reaching the optimal information size).

dDowngraded twice due to high risk of bias in multiple domains, including blinding (detection bias), incomplete outcome data (attrition bias), and unclear risk of bias for selection bias and other (early termination).

eWe downgraded the certainty of evidence once for indirectness as one trial was conducted in participants with thalassaemia intermedia only, a milder form of thalassaemia.

fDowngraded twice for imprecision due to small sample size (below optimal information size for this outcome).

Figuras y tablas -
Summary of findings 1. Summary of findings: Comparison 1 ‐ deferiprone (DFP) versus deferoxamine (DFO)
Table 1. Adherence measurement and results table

Study

How adherence was 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/week because of problems with swallowing

Badawy 2010

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

Combined therapy, and DFP only groups were more compliant (than DFO only) 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 were non‐adherent at end of trial

 

No data on control group

Calvaruso 2014

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: 89%

 

DFO compliance rate: 75%

 

No information regarding N or time point measured

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%

 

No information regarding N or time point measured

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 the participant 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)

Gharaati 2019

Questionnaire developed by researchers in 4 sections:

  1. Background: type of chelation drugs taken, frequency of taking chelation drugs on a weekly basis, frequency of injections on a monthly basis

  2. Patient knowledge of medications and self‐care behaviour

  3. Attitude to status, medication and self‐care

  4. Showing self‐care behaviours

"phone‐mediated education managed to improve the use of chelation drugs in the intervention group and regulate patients’ visits to hospital for blood injection"

 

However, baseline difference may have biased this

Hassan 2016

Records of all trial medications that were dispensed and returned

 

Parents were instructed to contact the investigator if the participants 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

Kwiatkowsi 2021

Treatment compliance was measured monthly by counting the number of tablets or measuring the volume of oral solution returned for participants on deferiprone, and by checking the infusion pump electronic record for participants on deferoxamine

 

In addition, participants were asked to record their medication usage in a diary

 

Participants who took 80% to 120% of the prescribed dose were considered to be compliant

Treatment compliance throughout the study was similar between the groups (P = 0.12)

 

DFP: 68.9%

 

DFO: 78.9%

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, mean (SD; range): DFP 92.7% (15.2%; 37% to 100%); DFO 70.6% (24.1%; 25% to 100%)

 

DFP alone group, mean (SD; range): 93.6% (9.7%; 56% to 100%)

Maggio 2020

Compliance was appropriate if the proportion of prescribed therapy taken was at least 80%

 

Compliance was estimated from electronic case report form data and the proportion of the prescribed doses taken

Appropriate compliance:

DFP, proportion, mean (SD), median (IQR): 183/193 (95%) participants, mean 92% (17.35), 93% (13.6)

DFX, proportion, mean (SD), median (IQR): 192/197 (97%) participants, 95% (18.56), 97% (11.1)

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 participants and good in 3 participants

Olivieri 1997

% 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, mean (SD): 94.9% (1.1%)

 

DFO, mean (SD): 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, mean (SD): 94% (5.3%)

 

DFO, mean (SD): 93% (9.7%)

 

Pennell 2014

Not stated how adherence was measured

DFX, mean (SD): 99.0% (3.5%)

 

DFO, mean (SD): 100.4% (10.9%)

Taher 2017

Assessed by relative consumed tablet count

 

DT: 85.3% (95% CI 81.1 to 89.5)

FCT: 92.9% (95% CI 88.8 to 97.0)

 

Also reported as n/N, unrelated to % (SD) reported above:

DT: 73/86 (84.9%)

FCT: 81/87 (93.1%)

FCT vs DT: RR 1.10 (95%CI 0.99, 1.22)

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 bi‐monthly visits

DFO/placebo, mean (SD): DFO 91.4% (2.7%); placebo 89.8 (7.2%)

 

DFO/DFP, mean (SD): 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; IQR: interquartile range; RR: risk ratio; SD: standard deviation; SF: serum ferritin

Figuras y tablas -
Table 1. Adherence measurement and results table
Summary of findings 2. Summary of findings: Comparison 2 ‐ deferasirox (DFX) versus deferiprone (DFO)

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 (%, SD)

See comments.

452
(3 RCTs)

⨁◯◯◯
Very lowa,b

3 RCTs (n = 452) reported adherence, although 2 of these could not be analysed (Hassan 2016, n = 60; and Vichinsky 2007, n = 195). All 3 RCTs reported no significant difference between groups.

SAEs Thalassaemia‐related SAEs

DFO: 83 per 1000

DFX: 79 per 1000 (34 to 179)

 

RR 0.95 (0.41 to 2.17)

247
(2 RCTs)

⨁◯◯◯
Very lowa,b

Zero cases reported in one RCT (n = 60, Hassan 2016), so data are based on a single trial (n = 187, Pennell 2014).

SAEs

SCD‐related SAEs

1 RCT (n = 195) reported SCD‐related AEs as "pain crisis" and "other", so no overall estimate of effect (subtotals calculated using 99% CI)

 

195
(1 RCT)

⨁◯◯◯
Very lowa,b

Data for sub‐outcome "pain crisis", and sub‐outcome "other", are presented in the main text, but we are unable to combine these data as there may be double‐counting; we have therefore not presented the summary statistic in the SoF table.

Sub‐outcomes are presented using 99% CI instead of 95% CI.

All‐cause mortality

8 per 1000

8 per 1000
(1 to 128)

POR 0.96
(0.06 to 15.42)

240
(2 RCTs)

⨁◯◯◯
Very lowa,b

Both RCTs reporting this outcome were in people with thalassaemia only; zero cases in 1 RCT.

Sustained adherence

See comments.

Sustained adherence is reported as adherence since all studies were longer than 6 months and only reported end of study adherence.

QoL

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).

AE: adverse event; CI: confidence interval; DFO: deferiprone; DFX: deferasirox; POR: Peto odds ratio; QoL: quality of life; RCT: randomised controlled trial; RR: risk ratio; SAE: serious adverse event; SD: standard deviation; SoF: summary of findings

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.

Explanations

aWe downgraded the certainty of evidence twice due to high or uncertain risk of bias in several domains.

bWe downgraded the certainty of evidence once due to imprecision as the CIs are wide and there is only one study with data in the comparison.

Figuras y tablas -
Summary of findings 2. Summary of findings: Comparison 2 ‐ deferasirox (DFX) versus deferiprone (DFO)
Summary of findings 3. Summary of findings: Comparison 3 ‐ deferiprone (DFP) versus deferasirox (DFX)

Intervention: DFP  

Comparison: DFX  

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFX

Risk with DFP

Adherence to iron chelation (%, SD)
Follow‐up: 12 months

The mean adherence to iron chelation (%, SD) was 95.00%.

MD 3.00 % lower
(6.56 lower to 0.56 higher).

390
(1 RCT)

⨁⨁◯◯
Lowa

95% adherence in DFX group as reported by Maggio 2020.

SAE (chelation‐related) (n/N)
Follow‐up: 12 months

20 per 1000

31 per 1000
(9 to 100)

POR 1.54
(0.44 to 5.39)

390
(1 RCT)

⨁◯◯◯
Very lowa,b

Total SAEs 
Follow‐up: 12 months

71 per 1000

68 per 1000
(33 to 139)

RR 0.95
(0.46 to 1.96)

390
(1 RCT)

⨁◯◯◯
Very lowa,b

All‐cause mortality (n/N)
Follow‐up: 12 months

0 per 1000

0 per 1000
(0 to 0)

RD 0.00
(‐0.01 to 0.01)

390
(1 RCT)

⨁⨁◯◯
Lowc

No deaths occurred during the study period, though the sample size was below the optimal information size to make any assessment of risk.

Sustained adherence

See comments.

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

QoL

Outcome 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).

AE: adverse event; CI: confidence interval; DFP: deferiprone; DFX: deferasirox; POR: Peto odds ratio; QoL: quality of life; RCT: randomised controlled trial; RD: risk difference; RR: risk ratio; SAE: serious adverse event; 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.

Explanations

aDowngraded twice for high risk of bias for blinding: may impact adherence, clinical decision‐making or reporting of AEs (no impact on mortality).

bDowngraded twice for imprecision due to wide CIs.

cDowngraded twice for imprecision due to zero events in both arms. Below optimal information size.

Figuras y tablas -
Summary of findings 3. Summary of findings: Comparison 3 ‐ deferiprone (DFP) versus deferasirox (DFX)
Summary of findings 4. Summary of findings: Comparison 4 ‐ deferasirox (DFX) film‐coated tablets versus DFX dispersible tablets

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 (%, SD) 
Follow‐up: 13 weeks

The mean adherence to iron chelation therapy (%, SD) was 84.3%.

MD 5.00% higher
(6.75 lower to 16.75 higher)

91
(1 RCT)

⨁◯◯◯
Very lowa,b

Mean 84.3% (95% CI 81.1 to 89.5) as reported by Taher 2017 in control (DFX dispersible tablet).

Sustained adherence to iron chelation therapy (%, SD)
Follow‐up: 24 weeks

The mean sustained adherence to iron chelation therapy (%, SD) was 82.9%.

MD 7.00% higher
(8.94 lower to 22.94 higher)

54
(1 RCT)

⨁◯◯◯
Very lowa,b

Mean 82.9% as reported in control group (dispersible tablet).

Incidence of SAEs

151 per 1000

184 per 1000
(94 to 358)

RR 1.22
(0.62 to 2.37)

173
(1 RCT)

⨁◯◯◯
Very lowa,c

All‐cause mortality

0 per 1000

0 per 1000
(0 to 0)

POR 7.30 (0.14 to 368.15)

173
(1 RCT)

⨁◯◯◯
Very lowa,c

QoL

Outcome 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; DFX: deferasirox; MD: mean difference; POR: Peto odds ratio; QoL: quality of life; RR: risk ratio; SAE: serious adverse event; 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.

Explanations

aWe downgraded the certainty of evidence twice for risk of bias due to high or unclear risk of bias in all domains.

bDowngraded twice for imprecision due to very wide confidence intervals and small study size (smaller than optimal information size).

cWe downgraded the certainty of evidence once for imprecision due to wide CIs.

Figuras y tablas -
Summary of findings 4. Summary of findings: Comparison 4 ‐ deferasirox (DFX) film‐coated tablets versus DFX dispersible tablets
Summary of findings 5. Summary of findings: Comparison 5 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFP

Intervention: DFP plus 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 plus DFO

Adherence to iron chelation therapy (%, SD)

See comments.

369
(4 RCTs)

⨁⨁◯◯
Lowa

4 RCTs reported adherence: 1 did not report by group, but stated compliance was similar (Badawy 2010, n = 100); 2 reported compliance as "excellent compliance" (Aydinok 2007, n = 20 and El Beshlawy 2008, n = 36); and 1 as % (SD) with no difference between groups (Maggio 2009, n = 213).

Incidence of SAEs

28 per 1000

4 per 1000
(0 to 78)

RR 0.15
(0.01 to 2.81)

213
(1 RCT)

⨁⨁◯◯
Lowb,c

All‐cause mortality

33 per 1000

26 per 1000
(6 to 105)

POR 0.77
(0.17 to 3.42)

237
(2 RCTs)

⨁◯◯◯
Very lowc,d

Sustained adherence

Outcome not reported.

Sustained adherence is reported as adherence since trial duration was longer than 6 months and trials report adherence for the whole length of trial.

QoL

See comments.

QoL was either not reported or no validated instruments were used.

*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; DFP: deferiprone; POR: Peto odds ratio; QoL: quality of life; 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.

Explanations

aWe downgraded the certainty of evidence twice for risk of bias as there was high or uncertain risk of bias in most domains in three out of four trials.

bWe downgraded the certainty of evidence once due to high or unclear risk of bias in three domains.

cWe downgraded the certainty of evidence once for imprecision due to wide CIs.

dWe downgraded the certainty of evidence twice for risk of bias as there was high or uncertain risk of bias in one trial in this comparison.

Figuras y tablas -
Summary of findings 5. Summary of findings: Comparison 5 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFP
Summary of findings 6. Summary of findings: Comparison 6 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFO

Intervention: DFP plus 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 plus DFO

Adherence to iron chelation therapy (%, SD)

See comments.

281
(5 RCTs)

⨁⨁◯◯
Lowa

5 RCTs reported adherence/compliance at approx 1 year: 2 RCTs did not report by group, simply stating "no statistical difference" (Badawy 2010, n = 100) and "excellent" (El Beshlawy 2008, n = 38); 1 RCT only reported compliance for the combined group (Galanello 2006a, n = 60); 1 RCT reported "excellent or good in all 11 (combined) and 14 (DFX only) participants" that were analysed (Mourad 2003, n = 25); and 1 RCT reported by group as "no significant difference" (Tanner 2007, n = 58).

Incidence of SAEs

See comments.

180
(4 RCTs)

⨁⨁◯◯
Lowa

3 RCTs report zero SAEs; 1 RCT did not report SAEs.

Badawy 2010 is not included in quantitative analysis 

All‐cause mortality

See comments.

No included trials reported death as an outcome. As AEs/SAEs were reported, we suspect no deaths occurred.

Sustained adherence

See comments.

Sustained adherence reported above as adherence since study duration was longer than 6 months and adherence reported at end of trial.

QoL

Outcome 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; DFP: deferiprone; QoL: quality of life; RCT: randomised controlled trial; SAE: serious adverse event; 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.

Explanations

aWe downgraded the certainty of evidence twice for risk of bias as high or unclear risk of bias in all domains.

Figuras y tablas -
Summary of findings 6. Summary of findings: Comparison 6 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFO
Summary of findings 7. Summary of findings: Comparison 7 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFP plus deferasirox (DFX)

Intervention: DFP plus DFO 

Comparison: DFP plus DFX  

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with DFP plus DFX

Risk with DFP plus DFO

Adherence to iron chelation therapy rates (n, N)

Follow‐up 1 year

938 per 1000

788 per 1000
(675 to 928)

RR 0.84
(0.72 to 0.99)

96
(1 RCT)

⨁⨁◯◯
Lowa,b

Incidence of SAEs

21 per 1000

21 per 1000
(1 to 257)

POR 1.00
(0.06 to 16.22)

96
(1 RCT)

⨁◯◯◯
Very lowa,b,c

All‐cause mortality ‐ at 1 year ‐ trial end

0 per 1000

0 per 1000
(0 to 0)

RD 0.00
(‐0.04 to 0.04)

96
(1 RCT)

⨁◯◯◯
Very lowa,b,d

No deaths occurred during the trial period, though the sample size was significantly below the optimal information size to make any assessment of risk.

Sustained adherence

See comments.

Sustained adherence is reported as adherence since the trial was 1 year in duration and end of trial adherence data were reported.

QoL

See comments.

96
(1 RCT)

1 RCT used SF‐36 to measure QoL; the results are presented as a bar graph only, with mean and SD not reported in extractable form (Elalfy 2015). Stated no difference between groups.

*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; DFP: deferiprone; DFX: deferasirox; POR: Peto odds ratio; QoL: quality of life; RCT: randomised controlled trial; RD: risk difference; RR: risk ratio; SAE: serious adverse event

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.

Explanations

aWe downgraded the certainty of evidence once for risk of bias as there was high or unclear risk of bias in three domains.

bWe downgraded the certainty of evidence once for indirectness as the trial included children aged 10 to 18 years with severe iron overload.

cWe downgraded the certainty of evidence once for imprecision as the comparison has wide CIs.

dDowngraded twice for imprecision due to the small sample size, far below the optimal information size for mortality.

Figuras y tablas -
Summary of findings 7. Summary of findings: Comparison 7 ‐ deferiprone (DFP) plus deferoxamine (DFO) versus DFP plus deferasirox (DFX)
Summary of findings 8. Summary of findings: Comparison 8 ‐ medication management versus standard care

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

See comments.

This outcome was not reported in the control group and therefore there are no comparative data.

SAEs

Outcome not reported.

Mortality

Outcome not reported.

Sustained adherence

Outcome not reported.

QoL PedsQLTM total score

Follow‐up: 6 months

See comments.

48
(1 RCT)

⨁◯◯◯
Very lowa,b

1 RCT reported medians and IQRs.

Medication management: 63.51 (51.75 to 84.54), n = 24; standard care: 49.84 (41.9 to 60.81), n = 24.

*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; IQR: interquartile range; PedsQLTM: Pediatric Quality of Life InventoryTM: QoL: quality of life; RCT: randomised controlled trial; SAE: serious adverse event

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.

Explanations

aWe downgraded the certainty of evidence twice for risk of bias due to high or uncertain risk of bias in all domains.

bWe downgraded the certainty of evidence twice for indirectness because most outcomes were only reported in the medication management group.

Figuras y tablas -
Summary of findings 8. Summary of findings: Comparison 8 ‐ medication management versus standard care
Table 2. Study overview: Comparison 1. DFP versus DFO

Study

Participants

Intervention

Comparator

Outcomes

Badawy 2010*

Egypt

Age > 8 years

β‐thalassaemia (100%)

DFP

75 mg/kg/day, daily

n = 50

DFO 

40 mg/kg/day, 5 days/week

n = 50

Adherence 

AEs

Calvaruso 2014

Italy

Age > 13 years

SCD (100%)

DFP 

75 mg/kg/day, divided into 3 oral daily doses (daily)

n = 30

DFO 

SC infusion (8 to 10 hours) at 50 mg/kg/day for 5 days/week

n = 30

Compliance

Mortality (5 years)

AEs (not SAEs)

Calvaruso 2015

Italy

Age > 13 years

Thalassaemia intermedia (100%)

DFP

75 mg/kg/day, divided into 3 oral daily doses (daily)

n = 47

DFO 

SC infusion (8 to 10 hours) at 50 mg/kg/day for 5 days/week

n = 41

Adherence

Compliance

Mortality (5 years)

El Beshlawy 2008

Egypt

Age > 4 years

β‐thalassaemia (100%)

DFP

60 to 83 mg/kg/day (daily)

n = 18

DFO 

23 to 50 mg kg/day for 5 days/week

n = 20

Adherence

Compliance

AEs

Iron overload

Kwiatkowski 2021

USA

 

Note: terminated early

Age > 2 years

SCD or other iron overload (excluded thalassaemia or MDS)

DFP

75 mg/kg (25 mg/kg per dose); 3/day, 8 hours apart to 99 mg/kg for more severe

n = 152

DFO

SC infusion (8 to 12 hours) 20 to 40 mg/kg/day for 5 to 7 days/week

n = 76

12 months:

Adherence

Mortality

HRQoL

SAEs (chelation associated)

All SAEs

Other AEs related to chelation

Olivieri 1997

Canada

Age > 10 years

β‐thalassaemia major (100%)

DFP

75 mg/kg/day in 3 divided doses

n = 19

DFO

50 mg/kg/night, 4 to 7 nights/week

n = 18

Adherence 

(3 months)

Pennell 2006

Italy and Greece

Age > 18 years

β‐thalassaemia major (100%)

DFP 

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

n = 29

DFO 

SC injection 50 mg/kg for 5 or more days/week

n = 32

Adherence

AEs

*Badawy 2010 did not report any outcomes by intervention group and did not include counts of events (i.e. AEs) and so was not included in the quantitative analysis.

Badawy 2010 and El Beshlawy 2008 are 3‐arm trials (DFP, DFO vs DFP vs DFO) and so are listed in more than one comparison.

AE: adverse events; DFO: deferoxamine; DFP: deferiprone; MDS: myelodysplastic syndromes; SAE: serious adverse events; SC: subcutaneous; SCD: sickle cell disease; SF: serum ferritin

Figuras y tablas -
Table 2. Study overview: Comparison 1. DFP versus DFO
Table 3. Study overview: Comparison 2. DFX versus DFO

Study

Participants

Intervention

Comparator

Outcomes

Hassan 2016

Egypt

Age > 6 years

β‐thalassaemia major

DFX

20 to 40 mg/kg/day on an empty stomach

n = 30

DFO

20 to 50 mg/kg/day via SC infusion over 8 to 10 hours, 5 days/week

n = 30

Adherence

Drug safety

Pennell 2014

CORDELIA (multi‐national: 11 countries)

Age > 10 years

β‐thalassaemia (100%)

DFX

20 mg/kg per day for 2 weeks, then 30 mg/kg/day for 1 week, 

then 40 mg/kg/day

n = 98

DFO

50 to 60 mg/kg/day via SC infusion over 8 to 12 hours, 5 to 7 days/week

n = 99

1 year:

Adherence

LIC

SF

 

Vichinsky 2007

(Multi‐national: 5 countries)

Age > 2 years

SCD

DFX

10 to 30 mg/kg according to baseline LIC (daily)

n = 132

DFO

50 to 70 mg/kg slow SC infusion over 8 to 12 hours, 5 to 7 days/week

n = 63

52 weeks:

Adherence

Safety

LIC

SF

 

DFO: deferoxamine; DFX: deferasirox; LIC: liver iron content; SC: subcutaneous; SCD: sickle cell disease, SF: serum ferritin

Figuras y tablas -
Table 3. Study overview: Comparison 2. DFX versus DFO
Table 4. Study overview: Comparison 3. DFP versus DFX

Study

Participants

Intervention

Comparator

Outcomes

Maggio 2020

DEEP‐2 (multi‐national)

Age 1 month to 18 years

Any hereditary haemoglobinopathy: including thalassaemia and SCD

DFP

75 to 100 mg/kg/day, orally, daily

n = 193

DFX (dispersible tablets)

20 to 40 mg/kg/day

n = 197

12 months:

Compliance

DFP: deferiprone; DFX: deferasirox; SCD: sickle cell disease

Figuras y tablas -
Table 4. Study overview: Comparison 3. DFP versus DFX
Table 5. Study overview: Comparison 4. DFX film‐coated tablet versus DFX dispersible tablet

Study

Participants

Intervention

Comparator

Outcomes

Taher 2017

ECLIPSE (multi‐national)

Age > 10 years

Thalassaemia and iron overload

Thalassaemia major (81%)

DFX film‐coated tablet

as 90 mg, 180 mg and 360 mg for oral use

n = 87

DFX dispersible tablet

as 125 mg, 250 mg and 500 mg for oral use

n = 86

13 and 24 weeks:

Adherence

Compliance

Safety 

AEs

AEs: adverse events; DFX: deferasirox; SCD: sickle cell disease

Figuras y tablas -
Table 5. Study overview: Comparison 4. DFX film‐coated tablet versus DFX dispersible tablet
Table 6. Study overview: Comparison 5. DFP and DFO versus DFP

Study

Participants

Intervention

Comparator

Outcomes

Aydinok 2007

Turkey

Age > 4 years

β‐thalassaemia (100%)

DFP + DFO (combined)

DFO (50 mg/kg/day SC twice‐weekly) combined with DFP (75 mg/kg/day, daily)

n = 12 (8 analysed)

DFP

75 mg/kg/day, daily

n = 12

12 months:

Adherence

LIC

SF

QoL

Badawy 2010*

Egypt

 

Age > 8 years

β‐thalassaemia (100%)

DFP, DFO

Twice‐weekly DFO (40 mg/kg/day)

DFP (75 mg/kg/day)

n = 50

DFP

75 mg/kg/day, daily

n = 50

Adherence 

AEs

El Beshlawy 2008

Egypt

Age > 4 years

β‐thalassaemia (100%)

DFP + DFO

DFP 60 to 83 mg/kg/day (daily) and DFO 23 to 50 mg/kg per dose (8 hours, 2 days/week)

n = 18

DFP

60 to 83 mg/kg/day (daily)

n = 18

Adherence

Compliance

Adverse events

Iron overload

Maggio 2009

Italy

Age > 13 years

Thalassaemia major (100%)

DFP‐DFO (sequential treatment)

DFP 75 mg/kg, divided into 3 oral daily doses, for 4 days/week

DFO SC infusion (8 to 12 hours) at 50 mg/kg/day for the remaining 3 days/week

n = 105

DFP

75 mg/kg divided into 3 oral daily doses, daily

n = 108

5 years:

Adherence

Survival

LIC & SF

AEs

 

*Badawy 2010 did not report any outcomes by intervention group and did not include counts of events (i.e. AEs) and so was not included in the quantitative analysis.

Badawy 2010 and El Beshlawy 2008 are 3‐arm trials (DFP, DFO vs DFP vs DFO) and so are listed in more than one comparison.

AE: adverse events; DFO: deferoxamine; DFP: deferiprone; LIC: liver iron content; QoL: quality of life; SC: subcutaneous; SF: serum ferritin

Figuras y tablas -
Table 6. Study overview: Comparison 5. DFP and DFO versus DFP
Table 7. Study overview: Comparison 6. DFP and DFO versus DFO

Study

Participants

Intervention

Comparator

Outcomes

Badawy 2010*

Egypt

Age > 8 years

β‐thalassaemia (100%)

DFP, DFO

Twice‐weekly DFO (40 mg/kg/day)

DFP (75 mg/kg/day)

n = 50

DFO

40 mg/kg/day; 5 days/week

n = 50

Adherence 

SF

El Beshlawy 2008

Egypt

Age > 4 years

β‐thalassaemia (100%)

DFP + DFO

DFP 60 to 83 mg/kg/day (daily) and DFO 23 to 50 mg/kg per dose (8 hours, 2 days/week)

n = 18

DFO 

23 to 50 mg kg/day for 5 days/week

n = 20

54 weeks:

Adherence/compliance

Adverse events (chelation‐related SAEs)

Iron overload

Other AEs

SAEs not reported

Galanello 2006a

Italy and Greece

Age > 10 years

β‐thalassaemia major (100%)

DFP + DFO

DFO 20 to 60 mg/kg/day SC on 2 days a week with DFP 25 mg/kg/ body weight 3 x daily for 5 days/week

n = 29

DFO

20 to 60 mg/kg/day subcutaneously on 5 to 7 days/week

n = 30

12 months:

Compliance

LIC and SF

AEs

Mourad 2003

Lebanon

Age 12 to 40 years

β‐thalassaemia

DFP + DFO

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

n = 11

DFO

SC injection, 40 to 50 mg/kg 8 to 12 hours a day, 5 to 7 days/week

n = 14

1 year:

Compliance

Liver and renal function

AEs (side effects)

 

Tanner 2007

Sardinia

Age > 18 years

β‐thalassaemia

DFP + DFO

DFO 40 to 50 mg/kg SC for 5 days/week with DFP 75 mg/kg daily for 7 days/ week

n = 28

DFO

40 to 50 mg/kg SC for 5 days/week with an oral placebo 

n = 30

1 year:

compliance

LIC and SF

AEs

*Badawy 2010 did not report any outcomes by intervention group and did not include counts of events (i.e. AEs) and so was not included in the quantitative analysis.

Badawy 2010 and El Beshlawy 2008 are 3‐arm trials (DFP, DFO vs DFP vs DFO) and so are listed in more than one comparison.

AE: adverse events; DFO: deferoxamine; DFP: deferiprone; LIC: liver iron content; QoL: quality of life; SAE: serious adverse events; SC: subcutaneous; SF: serum ferritin

Figuras y tablas -
Table 7. Study overview: Comparison 6. DFP and DFO versus DFO
Table 8. Study overview: Comparison 7. DFP/DFO versus DFP/DFX

Study

Participants

Intervention

Comparator

Outcomes

Elalfy 2015

Egypt and Oman

Age 10 to 18 years

β‐thalassaemia major

DFP/DFO

DFP 75 mg/kg/day divided into 2 doses taken orally for 7 days (with 6‐ to 8‐hour interval between the 2 doses) with DFO 40 mg/kg/day by SC infusion over 10 hours starting at 10 p.m. for 6 days/week

n = 48

DFP/DFX

DFP 75 mg/kg/day, divided into 2 doses taken orally with DFX 30 mg/kg/day taken orally at 10 p.m. for 7 days/week

n = 48

1 year:

Adherence

LIC and SF

SAEs and AEs

Compliance

Satisfaction

QoL

 

AE: adverse events; DFO: deferoxamine; DFP: deferiprone; DFX: deferasirox; LIC: liver iron content; QoL: quality of life; SAE: serious adverse events; SC: subcutaneous; SF: serum ferritin

Figuras y tablas -
Table 8. Study overview: Comparison 7. DFP/DFO versus DFP/DFX
Table 9. Study overview: Comparison 8. Medication management versus standard care

Study

Participants

Intervention

Comparator

Outcomes

Bahnasawy 2017

Egypt

Age 8 to 18 years

β‐thalassaemia major (100%)

Medication management

n = 24

Standard care

n = 24

6 months:

Adherence

SF

QoL

 QoL: quality of life; SF: serum ferritin

Figuras y tablas -
Table 9. Study overview: Comparison 8. Medication management versus standard care
Table 10. Study overview: Comparison 9. Education versus standard care

Study

Participants

Intervention

Comparator

Outcomes

Gharaati 2019*

Iran

Age > 13 years

Thalassaemia major

Education

6 x 15‐ to 18‐minute calls within a month

n = 46

Standard care

n = 45

1 month:

Use of chelation therapy

*Gharaati 2019 was not included in the quantitative analysis due to significant baseline imbalance (assessed using ROBINS‐I for non RCTs).

Figuras y tablas -
Table 10. Study overview: Comparison 9. Education versus standard care
Table 11. Overview of studies awaiting classification

Study

Reason for classification

Participants (inclusion criteria)

Intervention

Comparator 

Outcomes

Medication interventions – RCTs only

Bhojak 2020

RCT; N = 32; India

Expected start date: 1 Sept 2017

Expected end date: NR (6 month duration)

Full publication available: mentions greater compliance in IV group in discussion, but no data provided

Randomised but severe baseline imbalance in serum ferritin

Unclear trial design: significant differences between trial registration and publication (study design randomised or observational, and focus on adherence or not); contacted authors for further information

3 to 18 years

Thalassaemia patients on regular DFX

DFX, oral 15 to 40 mg/kg/day

DFO, injection, 20 to 40 mg/kg monthly

  • Serum ferritin

  • Side effects

  • Cost

  • Compliance 

 

CTRI/2020/07/026771

RCT; N = 45; India

Start date: 30 July 2020

End date: 10 August 2021

Unclear trial design (not designed to measure adherence?)

No publications or data

10 to 18 years

Beta thalassaemia patients taking DFX

Combined DFP (75 mg/kg/day) + DFX (30 mg/kg/day), oral

DFX (30 mg/kg/day), oral

  • Cardiac function

  • Kidney and liver function

  • Serum ferritin

EUCTR 2017‐003777‐34‐NL

(NL6659, PPI Shine Again)

RCT (cross‐over); N = 30; The Netherlands

End date: 12 April 2021

 

Completed, some results available (May 2022), but results presented without subgrouping, and so cannot extract only SCD and thalassaemia data – awaiting publication of further results and contacted authors for further information

18+ years

Hereditary anaemia (non‐transfusion dependent); secondary haemochromatosis

PPI: esomeprazole (oral capsule)

Placebo

  • Liver iron concentration

  • QoL (EQ‐5D)

  • Compliance to study drug

  • Need for iron chelation therapy

Eghbali 2019

RCT; N = 50; Iran

Start date: 22 September 2016

End date: 22 May 2017

 

Full publication available: mentions compliance with chelators was “acceptable”, but no data provided

Unclear trial design: significant differences between trial registration and publication (trial design randomised or observational); contacted authors for further information 

Would be a new comparison if included: DFO + DFX vs DFX

5 to 18 years

Thalassaemia major

Combined DFO (Desferal ampoule) 50 mg/kg subcutaneously with Desferal pump, and DFX (Exjade) 30 mg/kg/day

DFX (Exjade) 30 mg/kg/day

  • Serum ferritin

  • Compliance with chelators

  • Adverse events

  • Mortality 

IRCT 2016 0310026998N7

RCT; N = 54; Iran

Expected start date: 21 January 2018

Expected end date: 21 September 2018

Unclear trial design (not designed to measure adherence?)

No publications or data

 

12+ years

People with β‐thalassaemia receiving DFO plus DFP

DFX plus DFP (n = 27)

DFO plus DFP (n = 27)

  • SF

  • Liver iron concentration

  • QoL (SF‐36)

IRCT 2019 0106042262N1

RCT; N = 107; Iran

Start date: 19 February 2018

End date: 21 December 2018 

Unclear trial design (not designed to measure adherence?)

No publications or data

10+ years

Transfusion‐dependent β‐thalassaemia

DFX (20 to 40 mg/kg daily) plus DFP (15 mg/kg/dose)

DFO (20 to 50 mg/kg daily with a pump) plus DFP (15 mg/kg/dose)

  • Serum ferritin

  • Kidney and liver function

NCT00004982

Start date: December 1998

End date: November 2002

 

Unclear trial design (not designed to measure adherence?)

No publications or data

7+ years

Iron overload and thalassaemia

Various combinations of experimental iron chelating drugs

Standard care

  • NR

Non‐medication interventions – RCTs, NRSIs, CBA, ITS, repeated measures

EX‐PAT 2013

NRSI; N = 86; Turkey

Intervention from February to June 2009; follow‐up to one year

Abstract only

No information on inclusion/exclusion criteria

No publications or data

 

People using DFX (unclear diagnoses)

 

Education (n = 45)

Standard care (n = 41)

  • Compliance/ persistence

Crosby 2019

Feasibility study; N = 18; USA

Abstract only

Unclear trial design (single arm); part of larger study of self‐management interventions

No publications or data

 

13 to 21 years

SCD

Electronic monitoring bottles

Unclear

  • Adherence

IRCT 2013 042213092N1

RCT; N = 70; Iran

Start date: 20 June 2013

Expected end date: 21 September 2013

Unclear trial design (not designed to measure adherence?) 

No publications or data

 

15 to 25 years

Thalassaemia major

Education

Standard care

  • QoL

  • Empowerment

IRCT 2019 0827044634N1

RCT; N = 60; Iran

Expected start date: 11 September 2019

Expected end date (recruitment): 11 December 2019

Unclear if relevant intervention

No publications or data

 

14 ‐ 18 years

β‐thalassaemia major

 

Hope Therapy programme

Standard care

  • Adherence to treatment

  • Hope

IRCT 2020 0126046270N1

Pre/post‐test or NRSI; N = 47; Iran

Start date: 25 September 2019

End date: 21 January 2020

Unclear trial design (not designed to study adherence?)

No publications or data

 

8 ‐ 18 years

Thalassaemia major

Psycho‐educational group sessions (n = 25)

Standard care (pre‐test only)

  • Anxiety

  • Loneliness

IRCT 2020 0606047670N2021

RCT; N = 34; Iran

Expected start date: 20 December 2020

Expected end date: 17 February 2021

Unclear trial design (not designed to assess adherence?)

No publications or data

 

15 to 20 years

Thalassaemia major

Religious education

No intervention 

  • Life expectancy

  • Mental health

  • Spiritual health

CBA: controlled before‐after studies; DFO: deferoxamine; DFP: deferiprone; DFX: deferasirox; ITS: interrupted time series; IV: intravenous; NR: not reported; NRSI: non‐randomised studies of interventions; PPI: proton pump inhibitor; QoL: quality of life; RCT: randomised controlled trial; SCD: sickle cell disease; SF: serum ferritin

Figuras y tablas -
Table 11. Overview of studies awaiting classification
Table 12. Overview of ongoing studies

Study

Participants (inclusion criteria)

Intervention

Comparator 

Outcomes

Medication interventions ‐ RCTs only

CALYPSO

NCT02435212

Multi‐country

RCT; N =2 24

Expected start: 21 October 2015

Expected end: 19 December 2023

2 to 18 years

Any transfusion‐dependent anaemia

DFX granule formulation; 14 mg/kg/day; 48 weeks

DFX DT formulation; 20 mg/kg/day; 48 weeks

  • Compliance

  • Change in serum ferritin

  • Satisfaction

  • Overall safety

IRCT2015101218603N2

Country: Iran

RCT; N = 100

Expected start: 22 December 2015

Expected end: NR

2+ years

Transfusion‐dependent beta‐thalassaemia

DFX (new formulation Jadenu) 14 to 28 mg/kg/day orally

DFX (Exjade) 20 to 40 mg/kg/day orally

  • Compliance

  • SF levels

  • Safety

  • GI effects

Non‐medication interventions – RCTs, NRSIs, CBA, ITS, repeated measures

Madderom 2016 (TEAM)

NTR4750 (NL42182.000.12)

Country: The Netherlands

RCT; N = 100

Expected start: January 2013

Expected end: NR

All ages

Homozygous or compound heterozygous sickle cell disease

Group medical appointments

Individual appointments (standard care)

  • Self‐efficacy

  • Adherence 

  • QoL (SF‐36)

 

NCT04877054

Country: USA

RCT; N = 16

Expected start: 30 December 2021

Expected end: 1 August 2022

13 to 22 years

Sickle cell disease

Telehealth (inc psycho‐medical education and motivational interviewing) 1/week for 4 sessions

Education only (single session)

  • Adherence

  • Feasibility

  • Acceptability

CBA: controlled before‐and‐after study; DFO: deferoxamine; DFP: deferiprone; DFX: deferasirox; DT: dispersible tablet; GI: gastrointestinal; ITS: interrupted time series; NRSI: non‐randomised studies of interventions; QoL: quality of life; RCT: randomised controlled trial; SF: serum ferritin

Figuras y tablas -
Table 12. Overview of ongoing studies
Table 13. HRQoL (Kwiatkowski 2021)

 

 DFP

DFO

n

Mean (SD)

n

Mean (SD)

CHQ‐50 physical (12‐month change)

60

29.3 (13.94)

23

30.5 (11.51)

CHQ‐50 psychosocial (12‐month change)

60

42.5 (11.62)

23

41.3 (10.07)

SF‐36 physical (12‐month change)

35

43.1 (10.65)

19

43.0 (8.72)

SF‐36 mental (12‐month change)

35

44.7 (15.97)

19

40.9 (12.64)

CHQ‐50: Child Health Questionnaire ‐ 50 items; DFO: deferoxamine; DFP: deferiprone; HRQoL: health‐related quality of life; SD: standard deviation; SE: standard error; SF‐36: 36‐item Short Form Survey

No significant between‐group differences. Major bias due to missing data (over half) for outcomes (DFP 152 at baseline; DFO 76 at baseline). Data presented as mean (SE) in publication, converted to SD here.

Figuras y tablas -
Table 13. HRQoL (Kwiatkowski 2021)
Comparison 1. DFP versus DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

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

2

Mean Difference (IV, Random, 95% CI)

Totals not selected

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

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.2.1 Total reported SAEs

1

228

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.83, 2.46]

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

2

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

1.3.1 Agranulocytosis

1

88

Risk Ratio (M‐H, Random, 99% CI)

7.88 [0.18, 352.39]

1.3.2 Pain crisis

1

228

Risk Ratio (M‐H, Random, 99% CI)

1.30 [0.54, 3.16]

1.3.3 Acute chest syndrome

1

228

Risk Ratio (M‐H, Random, 99% CI)

3.52 [0.07, 170.19]

1.3.4 Hepatic sequestration

1

228

Risk Ratio (M‐H, Random, 99% CI)

1.51 [0.02, 99.77]

1.3.5 Chelation therapy‐related SAEs

1

228

Risk Ratio (M‐H, Random, 99% CI)

1.50 [0.28, 8.04]

1.4 All‐cause mortality Show forest plot

3

376

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.18, 1.21]

1.4.1 Sickle cell disease

2

288

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.12, 2.02]

1.4.2 Thalassaemia intermedia

1

88

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.12, 1.63]

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

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.6 Organ damage Show forest plot

2

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

1.6.1 Liver damage

2

148

Risk Ratio (M‐H, Random, 99% CI)

5.13 [0.54, 48.40]

1.7 AEs related to iron chelation Show forest plot

4

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

1.7.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

192

Risk Ratio (M‐H, Random, 99% CI)

3.95 [0.37, 41.87]

1.7.2 Risk of pain or swelling in joints

3

192

Risk Ratio (M‐H, Random, 99% CI)

3.55 [0.49, 25.81]

1.7.3 Risk of nausea/vomiting

2

132

Risk Ratio (M‐H, Random, 99% CI)

13.68 [0.99, 188.88]

1.7.4 Risk of increased liver transaminase

1

44

Risk Ratio (M‐H, Random, 99% CI)

1.10 [0.03, 38.47]

1.7.5 Local reactions at infusion site

1

88

Risk Ratio (M‐H, Random, 99% CI)

0.17 [0.00, 9.12]

1.7.6 Other AEs related to iron chelation

1

228

Risk Ratio (M‐H, Random, 99% CI)

1.28 [0.81, 2.02]

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

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

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.2 SAEs (thalassaemia) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.2.1 Total thalassaemia‐related SAEs

2

247

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.41, 2.17]

2.3 SAEs (sickle cell disease) Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

2.3.1 Painful crisis

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.05 [0.59, 1.86]

2.3.2 Other sickle cell disease‐related SAEs

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.08 [0.69, 1.68]

2.4 All‐cause mortality (thalassaemia) Show forest plot

2

240

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.96 [0.06, 15.42]

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

2

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

2.5.1 Iron overload defined by ferritin 1500 (µg/l) or higher (thalassaemia)

1

60

Risk Ratio (M‐H, Random, 99% CI)

1.18 [0.52, 2.68]

2.5.2 Proportion with severe iron overload (liver iron concentration at least 15 mg/Fe/g dry weight)

1

172

Risk Ratio (M‐H, Random, 99% CI)

1.00 [0.78, 1.27]

2.5.3 Myocardial T2* < 10 ms

1

172

Risk Ratio (M‐H, Random, 99% CI)

1.10 [0.62, 1.95]

2.6 Total AEs related to iron chelation ‐ (thalassaemia) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.6.1 Total chelation‐related AEs

1

187

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.76, 1.73]

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

2

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

2.7.1 Gastrointestinal upset

1

60

Risk Ratio (M‐H, Random, 99% CI)

3.00 [0.41, 22.06]

2.7.2 Rash

2

247

Risk Ratio (M‐H, Random, 99% CI)

3.05 [0.69, 13.51]

2.7.3 Risk of increased blood creatinine

1

187

Risk Ratio (M‐H, Random, 99% CI)

3.79 [0.51, 28.05]

2.7.4 Risk of proteinuria

1

187

Risk Ratio (M‐H, Random, 99% CI)

2.21 [0.39, 12.56]

2.7.5 Risk of increased ALT

1

187

Risk Ratio (M‐H, Random, 99% CI)

5.69 [0.36, 89.55]

2.7.6 Risk of increased AST

1

187

Risk Ratio (M‐H, Random, 99% CI)

5.69 [0.36, 89.55]

2.7.7 Risk of diarrhoea

1

187

Risk Ratio (M‐H, Random, 99% CI)

5.69 [0.36, 89.55]

2.7.8 Risk of vomiting

1

187

Risk Ratio (M‐H, Random, 99% CI)

6.64 [0.14, 320.28]

2.8 Total AEs (thalassaemia) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

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

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

2.9.1 Risk of increased ALT

1

195

Risk Ratio (M‐H, Random, 99% CI)

5.29 [0.12, 232.98]

2.9.2 incidence of abdominal pain

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.91 [0.80, 4.58]

2.9.3 Risk of pain or swelling in joints

1

195

Risk Ratio (M‐H, Random, 99% CI)

1.06 [0.41, 2.76]

2.9.4 Risk of diarrhoea

1

195

Risk Ratio (M‐H, Random, 99% CI)

4.14 [0.90, 18.92]

2.9.5 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. DFP versus DFX

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

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

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.2 Total SAEs  Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.2.1 12 months

1

390

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.46, 1.96]

3.3 SAE (chelation‐related) (n/N) Show forest plot

1

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

3.3.1 12 months

1

390

Peto Odds Ratio (Peto, Fixed, 95% CI)

1.54 [0.44, 5.39]

3.4 All‐cause mortality (n/N) Show forest plot

1

Risk Difference (M‐H, Random, 95% CI)

Subtotals only

3.4.1 12 months

1

390

Risk Difference (M‐H, Random, 95% CI)

0.00 [‐0.01, 0.01]

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

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

4.1 Adherence to iron chelation therapy (n/N) Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

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

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.2.1 13 weeks

1

91

Mean Difference (IV, Random, 95% CI)

5.00 [‐6.75, 16.75]

4.2.2 24 weeks

1

54

Mean Difference (IV, Random, 95% CI)

7.00 [‐8.94, 22.94]

4.3 Incidence of SAEs Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.4 All‐cause mortality Show forest plot

1

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

4.5 Incidence of organ damage Show forest plot

1

173

Risk Ratio (M‐H, Random, 99% CI)

1.25 [0.72, 2.18]

4.5.1 Renal events

1

173

Risk Ratio (M‐H, Random, 99% CI)

1.25 [0.72, 2.18]

4.6 Total AEs related to iron chelation Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.6.1 Total chelation‐related AEs

1

173

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.57, 0.99]

4.7 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

4.7.1 Risk of diarrhoea

1

173

Risk Ratio (M‐H, Random, 99% CI)

0.70 [0.29, 1.70]

4.7.2 Increased urine protein/urine creatinine ratio

1

173

Risk Ratio (M‐H, Random, 99% CI)

1.65 [0.60, 4.54]

4.7.3 incidence of abdominal pain

1

173

Risk Ratio (M‐H, Random, 99% CI)

0.49 [0.16, 1.52]

4.7.4 Incidence of nausea

1

173

Risk Ratio (M‐H, Random, 99% CI)

0.72 [0.23, 2.23]

4.7.5 Incidence of vomiting

1

173

Risk Ratio (M‐H, Random, 99% CI)

0.28 [0.07, 1.15]

Figuras y tablas -
Comparison 4. DFX film‐coated tablet versus DFX dispersible tablet
Comparison 5. DFP and DFO versus DFP

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

5.1 Incidence of SAEs Show forest plot

1

213

Risk Ratio (M‐H, Random, 95% CI)

0.15 [0.01, 2.81]

5.2 All‐cause mortality Show forest plot

2

237

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.77 [0.17, 3.42]

5.3 Incidence of chelation therapy‐related AEs Show forest plot

3

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

5.3.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

280

Risk Ratio (M‐H, Random, 99% CI)

1.15 [0.50, 2.62]

5.3.2 Risk of pain or swelling in joints

2

256

Risk Ratio (M‐H, Random, 99% CI)

0.76 [0.31, 1.91]

5.3.3 Risk of gastrointestinal disturbances

1

213

Risk Ratio (M‐H, Random, 99% CI)

0.45 [0.15, 1.37]

5.3.4 Risk of increased liver transaminase

2

256

Risk Ratio (M‐H, Random, 99% CI)

1.02 [0.52, 1.98]

5.3.5 Nausea/vomiting

1

43

Risk Ratio (M‐H, Random, 99% CI)

0.55 [0.13, 2.23]

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

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

6.1 Other AEs related to iron chelation Show forest plot

4

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

6.1.1 Risk of leukopenia, neutropenia and/or agranulocytosis

3

169

Risk Ratio (M‐H, Random, 99% CI)

1.18 [0.09, 15.45]

6.1.2 Risk of pain or swelling in joints

3

135

Risk Ratio (M‐H, Random, 99% CI)

2.41 [0.17, 34.31]

6.1.3 Risk of increased liver transaminase

2

104

Risk Ratio (M‐H, Random, 99% CI)

3.46 [0.45, 26.62]

6.1.4 Nausea/vomiting

4

194

Risk Ratio (M‐H, Random, 99% CI)

4.34 [0.77, 24.44]

6.1.5 Local reactions at infusion site

2

90

Risk Ratio (M‐H, Random, 99% CI)

0.18 [0.01, 4.43]

Figuras y tablas -
Comparison 6. DFP and DFO versus DFO
Comparison 7. DFP and DFX versus DFP and DFO

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

7.1 Adherence to iron chelation therapy rates Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.2 Incidence of SAE Show forest plot

1

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

7.3 All‐cause mortality Show forest plot

1

Risk Difference (M‐H, Random, 95% CI)

Totals not selected

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

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

7.5 Total AEs related to iron chelation Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.5.1 one year (study end)

1

96

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.76, 1.53]

7.6 Other AEs related to iron chelation Show forest plot

1

Risk Ratio (M‐H, Random, 99% CI)

Subtotals only

7.6.1 Risk of leukopenia, neutropenia and/or agranulocytosis

1

96

Risk Ratio (M‐H, Random, 99% CI)

1.67 [0.27, 10.14]

7.6.2 Risk of pain or swelling in joints

1

96

Risk Ratio (M‐H, Random, 99% CI)

0.89 [0.29, 2.77]

7.6.3 Gastrointestinal problems

1

96

Risk Ratio (M‐H, Random, 99% CI)

0.60 [0.18, 2.04]

7.6.4 ALT (increase ≥ 3‐fold)

1

96

Risk Ratio (M‐H, Random, 99% CI)

1.33 [0.20, 8.88]

7.6.5 Skin rash

1

96

Risk Ratio (M‐H, Random, 99% CI)

5.00 [0.10, 261.34]

Figuras y tablas -
Comparison 7. DFP and DFX versus DFP and DFO