You are on page 1of 10

Blood Cells, Molecules, and Diseases 47 (2011) 166–175

Contents lists available at ScienceDirect

Blood Cells, Molecules, and Diseases


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / y b c m d

Iron chelation therapy in thalassemia major: A systematic review with meta-analyses


of 1520 patients included on randomized clinical trials
Aurelio Maggio a,⁎, Aldo Filosa b, Angela Vitrano a, Giuseppina Aloj b, Antonis Kattamis c, Adriana Ceci d,
Suthat Fucharoen e, Paolo Cianciulli f, Robert W. Grady g, Luciano Prossomariti h, John B. Porter i,
Angela Iacono j, Maria Domenica Cappellini k, Fedele Bonifazi l, Filippo Cassarà a, Paul Harmatz m,
John Wood n, Christian Gluud o
a
U.O.C. “Ematologia II con Talassemia”, A.O. “V. Cervello”, Palermo, Italy
b
U.O.S. Talassemia Pediatrica, A.O. Cardarelli, Napoli, Italy
c
First Department of Pediatrics, University of Athens, Medical School, Aghia Sophia Children's; Hospital, Thivon and Levadias, Athens, Greece
d
Consorzio per Valutazioni Biologiche e Farmacologiche, Pavia, Italy
e
Thalassemia Research Center, Institute of Science and Technology for Research and Development, Mahdol University, Bangkok, Thailand
f
U.O. D. Talassemia, A.O. “S. Eugenio”, Roma, Italy
g
Department of Pediatrics, Cornell University Medical Center, New York, NY, USA
h
Centro Regionale per la Cura delle Microcitemie, A.O.Cardarelli, Napoli, Italy
i
Department of Haematology, University College London, UCL Cancer Institute, London, UK
j
Foundation Leonardo Giambrone, Italy
k
Policlinico, Mangiagalli, Regina Elena Foundation IRCCS, University of Milan, Italy
l
I.RI.D.I.A. srl, Bari, Italy and Consorzio per Valutazioni Biologiche e Farmacologiche, Pavia, Italy
m
Division of Gastroenterology, Children's Hospital & Research Center Oakland, Oakland, USA
n
Division of Pediatric Cardiology, Children's Hospital Los Angeles, Los Angeles, California, USA
o
Copenhagen Trial Unit, Centre for Clinical Intervention Research, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark

a r t i c l e i n f o a b s t r a c t

Article history: The effectiveness of deferoxamine (DFO), deferiprone (DFP), or deferasirox (DFX) in thalassemia major was
Submitted 26 June 2011 assessed. Outcomes were reported as means ± SD, mean differences with 95% CI, or standardized mean
Revised 2 July 2011 differences. Statistical heterogeneity was tested using χ2 (Q) and I2. Sources of bias and Grading of
Available online 16 August 2011
Recommendations Assessment, Development and Evaluation system (GRADE) were considered. Overall, 1520
patients were included. Only 7.4% of trials were free of bias. Overall measurements suggest low trial quality
(Communicated by G. Stamatoyannopoulos,
M.D., Dr. Sci., 3 July 2011) (GRADE). The meta-analysis suggests lower final liver iron concentrations during associated versus monotherapy
treatment (pb 0.0001), increases in serum ferritin levels during DFX 5, 10, and 20 mg/kg versus DFO-treated
groups (pb 0.00001, p b 0.00001, and p = 0.002, respectively), but no statistically significant difference during
DFX 30 mg/kg versus DFO (p= 0.70), no statistically significant variations in heart T2* signal during associated or
sequential versus mono-therapy treatment (p= 0.46 and p = 0.14, respectively), increases in urinary iron
excretion during associated or sequential versus monotherapy treatment (p= 0.008 and p = 0.02, respectively),
and improved ejection fraction during associated or sequential versus monotherapy treatment (p= 0.01 and
p b 0.00001, respectively). These findings do not support any specific chelation treatment. The literature shows
risks of bias, and additional larger and longer trials are needed.
© 2011 Elsevier Inc. All rights reserved.

Introduction imbalance, and constitutes a major improvement for all people with
thalassemia major [3]. Therefore, standards for the treatment of
Thalassemia and hemoglobin disorders are an emerging global children and adults with thalassemia have been developed [4,5].
health burden, [1] accounting for about 3.4% of deaths in children less The three main chelators used in current clinical practice
than 5 years of age [2]. The current therapeutic management of are deferoxamine (DFO) (Desferal®; Novartis), deferiprone (DFP)
thalassemia is based on regular blood transfusions and iron chelation (Ferriprox®; Apotex), and deferasirox (DFX) (Exjade®; Novartis) [6].
therapy [3]. Chelation aims to reduce iron stores or correct iron The main findings concerning the effectiveness and safety profiles of
DFO, DFP, and DFX have been described extensively elsewhere [7–11].
Moreover, there is also growing off-license usage of DFP in associated
⁎ Corresponding author. Fax: + 39 0916880828. with (administration of the two drugs during the same day) or in
E-mail address: aureliomaggio@virgilio.it (A. Maggio). sequential with (administration of the two drugs on different days) DFO

1079-9796/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.bcmd.2011.07.002
A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175 167

following recent reports of their synergistic effect, particularly with Table 1


regard to the iron burden in the heart [12,13]. We identified eight Study design.

published reviews on iron chelators with our search strategy [14–21]. Inclusion criteria Exclusion criteria
However, only four of these included meta-analyses, [19] none included
Patient populations Patients with Patients with other forms of
sequential chelation treatment, and none reported GRADE. Moreover, thalassemia major thalassemia or chronic
none was conducted systematically, i.e. based on a previously public regardless of age anemia requiring blood
protocol guiding the review process. Furthermore, none assessed the transfusion.
Interventions/comparators Deferoxamine (DFO)
risks of systematic errors (‘bias’), random errors (‘play of chance’), and
versus placebo or
design errors [22–24]. different route of
Because these questions remain unanswered, the main objective of administration
our study was to conduct a systematic review of the clinical Deferiprone (DFP)
effectiveness profile of iron chelators for patients with transfusion- versus DFO
Deferasirox versus DFO
dependent thalassemia major, according to previously published
Associated therapy
criteria [25]. (DFO plus DFP) versus
DFO or DFP alone
Design and methods Sequential therapy
(DFP–DFO) versus DFP
or DFO alone
Literature searches Types of studies Randomized clinical Editorials, letters, opinions,
trials case reports, cross-over
The following electronic databases were utilized (AG and FA) to trials, non-randomized
search for relevant published literature: Cochrane Central Register of studies, quasi-randomized
studies, non-English
Controlled Trials (CENTRAL), MEDLINE, ISI Web of Knowledge, the
language papers.
ClinicalTrials.gov register, Current Controlled Trials, World Health Outcomes Ejection fraction
Organization (WHO) International Clinical Trials Registry Platform Liver iron
(ICTRP), and the reference lists of articles. The date of the last search concentration
Magnetic resonance
was June 2010. Two authors (AG and FA) independently scanned all
T2*
titles and abstracts identified in the research to select reports that Serum ferritin as
might be relevant to the clinical review, separating the cohort studies difference between
from randomized clinical trials (RCTs). Only trials that were clearly baseline and the end of
irrelevant were excluded at this stage. Agreement between the treatment
Urinary iron excretion
authors was good. For trials where important information was
lacking, a letter for request of information was sent to the authors
of the trials.

Keywords utilized for the searches The types of interventions described in these papers were (1) DFO
twice daily subcutaneous bolus injection versus DFO subcutaneous
MEDLINE mesh terms: deferoxamine, desferrioxamine, deferoxa- continuous infusion; (2) DFO versus other chelators; (3) DFP versus
mine mesilate, deferoxamine mesylate, deferrioxamine B, Desferal, other chelators; (4) DFX versus other chelators; (5) DFO plus DFP
desferioximine, desferrioxamine B, desferrioxamine B mesylate, versus other chelators; and (6) sequential DFP and DFO versus other
desferroxamine, deferiprone, 1,2-dimethyl-3-hydroxy-4-pyridinone, chelators.
1,2-dimethyl-3-hydroxypyrid-4-one, 1,2-dimethyl-3-hydroxypyridin-
4-one, 3-hydroxy-1,2-dimethyl-4-pyridinone, Chiesi brand of defer- Types of outcomes
iprone, Ferriprox, L1 oral chelate, deferasirox, Exjade, ICL670, and
ICL670 A. MEDLINE other key words: iron chelation therapy and The following measures were considered: (1) ejection fraction;
thalassemia, iron chelation and beta-thalassemia. CENTRAL, ISI Web of (2) liver iron concentration (LIC) (μg/g dry weight) at the end
Knowledge, ClinicalTrial.gov and WHO ICRTP mesh terms: deferox- of treatment; (3) myocardial iron concentration (evaluated by MRI
amine AND beta-thalassemia, deferiprone and beta-thalassemia, and T2*) at the end of the trial; (4) serum ferritin as the difference
deferasirox and beta-thalassemia. CENTRAL, ISI Web of Knowledge, between the final versus the basal value; and (5) urinary iron
ClinicalTrial.gov and WHO ICRTP other key words: iron chelation therapy excretion.
and thalassemia, iron chelation and thalassemia, deferoxamine and
thalassemia, deferiprone and thalassemia, deferasirox and thalassemia. Statistical analysis
Current controlled trials: deferoxamine, deferiprone, deferasirox,
thalassemia, and iron chelation. The continuous outcomes relevant to this study were presented as
means and standard deviations (SD), and summarized in terms of
Inclusion and exclusion criteria weighted mean difference (WMD) with 95% confidence intervals (CI)
or the standardized mean difference (SMD) when outcomes had a
The inclusion/exclusion criteria are shown in Table 1. One author different measurement scale. The dichotomous outcomes for each
(AG) transcribed the data into the systematic review computer trial were expressed as a relative risk (RR) with 95% CIs, and were
software RevMan 5.0 [26]. Another author (FA) checked all data entry combined in terms of RR for the meta-analysis using the Peto-
for discrepancies and verified the data. Extracted data were analyzed modified Mantel–Haenszel method.
using the most up-to-date version of RevMan (5.0) available at the The effect was computed across studies using either a fixed-effects
time of analysis (Review Manager 2008). Only randomized clinical model, if there was little statistical heterogeneity, or a DerSimonian
trials (RCT) were analyzed. Phase 2 cross-over trials and studies that and Laird random-effects model when there was unexplained
presented poor data or case reports were excluded. Only patients of heterogeneity between trial results. Statistical heterogeneity was
any age with transfusion-dependent thalassemia major, from any tested using the Cochrane chi-square (Q) test of homogeneity and
setting worldwide, were considered. with I 2 statistic, both available through RevMan 5 [26]. This test has
168 A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175

low power to detect true heterogeneity, especially with a small Potentially relevant titles and abstracts identified
number of studies; therefore, a cut-off p-value of 0.10 was used.
through electronic search: 541
The I 2 statistic quantifies inconsistency across studies; a value of I 2
of 25% or less can be considered a low degree of heterogeneity, a
value around 50% as moderate heterogeneity, and a value of 75% or
more as high heterogeneity. In this review, these quantitative
interpretations were limited because few studies were available for Color studies: 504
comparisons.

Evaluation of methodological quality of included trials


Articles selected for meta-analysis: 37
The risk of bias
The internal validity of the trials was assessed considering the
sources of bias that have a major influence on the magnitude of effect
size in clinical trials; adequate sequence generation, allocation Excluded articles: 21
concealment, blinding, incomplete outcome data addressed, free or
selective reporting, and free of other biases.
Other bias is defined as bias not addressed in the other domains in
the tool. Bias is a systematic error; therefore, it only goes one way, an Articles used for meta-analysis: 16 :
overestimation of benefit and an underestimation of harm. Risk of bias
tables were performed using the “Risk of bias” tool in RevMan 5 [26]. Fig. 1. Research flow-chart.
Each of the six domains (adequate sequence generation, allocation
concealment, blinding, incomplete outcome data addressed, free of
selective reporting, free of other bias) for a study, was completed Adequate sequence generation
providing a judgment categorized as “yes”, “no”, or “unclear”. “Yes” Of the RCTs, 48.2% had a low risk of bias; a low number of studies
meant that there is a low risk of bias, and “no” meant that there is a had high risk of bias, whereas the risk for 40.75% of studies was
high risk of bias. “Unclear” was used if there was insufficient classified as “unclear” (Figs. 2–3).
information to make this judgment or if the item was not relevant
to the study [27]. Allocation concealment
Overall, 22.3% of the studies had a low risk of bias, only one study
had a high risk of bias (3.7%), and most of the studies were classified as
“unclear” (74%) (Figs. 2–3).
The GRADE system

Blinding
The Grading of Recommendations Assessment, Development and
For blinding, 18.5% of the studies had a low risk of bias, 37%
Evaluation system (GRADE) was used to summarize the findings and
had a high risk of bias, and 44.5% were classified as “unclear”
rate the overall quality of evidence for each outcome [28]. The
(Figs. 2–3).
GRADE system involves specifying at least 5 main outcomes relevant
to the management of the patients. From these, the guideline
Incomplete outcome data addressed
development process begins. GRADE suggests a nine-point scale to
Overall, 7.4% of the studies had a low risk of bias, most studies
judge importance. Ratings of 7 to 9 equal outcomes of critical
(85.2%) had high risk of bias, and 7.4% of studies were classified as
importance for decision making. Ratings of 4 to 6 indicate outcomes
“unclear” (Figs. 2–3).
important but not critical. Ratings of 1 to 3 equal items of limited
importance [27].
Free of selective reporting
In this category, 7.4% of the studies had a low risk of bias; in
contrast, 70.4% of studies had a high risk of bias and 22.2% were
Results classified as “unclear” (Figs. 2–3).

Search results Free of other bias


Overall, 7.4% of the studies had a low risk of bias; in contrast, most
The results of our research strategies are summarized in Fig. 1. studies (88.9%) had a high risk of bias, and 3.7% were classified as
Sixteen RCTs were included in this review with a total of 1520 patients “unclear” (Figs. 2–3).
aged from 5 to 50 years. Five RCTs were long-term follow-ups of the
trials already included in the analysis. Within these 16 trials, 11 The GRADE system
different comparisons were detected. In these, the chelators were Randomized trials are considered the highest quality evidence
compared with each other as monotherapies, in combination with unless limited by methodological issues, inconsistency, indirectness
each other, or as sequentially administered chelators. Table 2 reports (generalizability), or imprecision [28]. In this review, all outcomes
the main findings of the RTCs included in this systematic review and were limited by weaknesses related to trial quality, such as lack of
used in the meta-analysis. blinding, small sample size, and imprecise estimates of treatment
effects. This resulted in a low quality for most outcomes. The 5 most
important outcomes chosen for quality assessment are described
The risk of bias below in decreasing order of importance:

The risk of bias is reported as a summary of the studies for each 1) Ejection fraction: GRADE = 9
judgment (proportion) in Fig. 2, and as single evaluation for each RCT 2) Liver iron concentration (LIC) (μg/g dry weight) at the end of
in Fig. 3. treatment: GRADE = 8
A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175 169

Table 2
Type of comparison among interventions reported in randomized clinical trials included in the systematic review with meta-analyses.

Comparison Exp Ctrl Exp Ctrl N. pts Author


N. pts

Monotherapy 1 DFO bolus/day s.c.i.a 10 10 Yarali et al. [49]


2 DFP DFO 10 10 Gomber et al. [33]
DFP DFO 6 7 Ha et al. [45]
DFP DFO 71 73 Maggio et al. [47]
DFP DFO 21 23 El Beshlawy et al. [50]
DFP DFO 29 32 Pennell et al. [41]
3 DFX (10 mg) Placebo 5 5 Nibset-Brown et al. [46]
4 DFX (20 mg) Placebo 6 5 Nibset-Brown et al. [46]
5 DFX (40 mg) Placebo 7 5 Nibset-Brown et al. [46]
6 DFX (5 mg) DFO 15 14 Cappellini et al. [32]
7 DFX (10 mg) DFO 78 79 Cappellini et al. [32]
8 DFX (20 mg) DFO 84 91 Cappellini et al. [32]
9 DFX (30 mg) DFO 119 106 Cappellini et al. [32]
Associated 10 10.0 versus DFP DFP + DFO DFP 12 12 Aydinok et al. [29]
10.1 versus DFO DFP + DFO DFO 10 10 Gomber et al. [33]
DFP + DFO DFO + placebo 32 33 Tanner et al. [13]
DFP + DFO DFO 22 23 El-Beshlawy et al. [50]
DFP + DFO DFO 20 16 Ha et al. [45]
DFP + DFO DFO 11 14 Mourad et al. [48]
Sequential 11 11.0 versus DFP DFP − DFO DFP 105 108 Maggio et al. [31]
11.1 versus DFO DFP − DFO DFO 29 30 Galanello et al. [44]
DFP − DFO DFO 30 30 Abdelrazik et al. [30]
a
s.c.i. = subcutaneous continuous infusion; Exp: experimental interventions, Ctrl: control interventions =; =; =.

3) Myocardial iron concentration (evaluated by MR T2*) at the end of Comparison of sequential DFP and DFO versus DFO. Ejection fraction at
the trial: GRADE = 8 the end of the intervention was significantly higher in the sequential
4) Serum ferritin levels such as the difference between the final DFP and DFO groups than in the DFO control group [30] (Fig. 4b)
versus the basal value: GRADE = 7 (WMD 9.02, 95% CI 6.4 to 11.64, p b 0.00001). GRADE quality of
5) Urinary iron excretion: GRADE = 6. evidence was low.

Outcome 2: Liver iron concentration (LIC) (μg/g dry weight) at the end
Outcome results of treatment

Outcome 1: Ejection fraction at the end of treatment Comparisons of DFP plus DFO versus other chelators. Liver iron
concentration at the end of the intervention was measured in two
Comparisons of DFP plus DFO versus other chelators. Ejection fraction studies as liver iron concentration by biopsy (mg/g dw) [29] and liver
at the end of the intervention was reported in two trials [13–29] iron concentration by magnetic resonance imaging as (T2*) (ms) [13]
(Fig. 4a) comparing 36 participants in the deferiprone plus DFO using different techniques (Fig. 4c). The results were pooled with
group with 42 participants in the control group, which included standardized mean difference. Thirty-six participants in the defer-
12 participants receiving DFP [29] and 30 receiving DFO [13]. iprone plus deferoxamine group were compared with 42 participants
Ejection fraction at the end of intervention was significantly in the control group, which included 12 participants who received
higher in the DFP plus DFO groups (WMD 3.37, 95% CI 0.79 to DFP [29] and 30 who received DFO [13]. Liver iron concentration at the
5.95, p = 0.01). GRADE quality of evidence was low. Heterogene- end of the intervention was significantly lower in the DFP plus DFO
ity for this outcome was not statistically significant (CHI 2 = 0.29, groups compared with the control group (WMD − 1.06, 95% CI −1.54
df = 1, p = 0.59, I 2 0%). to − 0.58, p b 0.0001). GRADE quality of evidence was very low.

Fig. 2. Risk of bias summary.


170 A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175

Comparison of sequential DFP and DFO versus DFP. Myocardial iron


concentration (T2*) at the end of the intervention was not
significantly different between participants taking sequential DFP
and DFO compared with those taking DFP in the control group
(Fig. 5b)[31]. However, the myocardial iron concentration (T2*) at the
end of the intervention was lower in the control group compared with
the sequential DFP and DFP groups (WMD = − 5.8, 95% CI −13.52 to
1.92, p = 0.14). GRADE quality of evidence was low.

Outcome 4: Serum ferritin as the difference between final versus basal


values

Comparison of DFX (5 mg/kg) and DFO (30 mg/kg). A statistically


significant increase in serum ferritin levels, determined as the
difference between baseline values and values at the end of
intervention, was shown in the group of patients treated with DFX
5 mg/kg/day compared with the DFO control group (WMD 978, 95% CI
544.71 to 1411.29, p b 0.00001) (Fig. 6a) [32]. GRADE quality of
evidence was low.

Comparison of DFX (10 mg/kg) versus DFO (35 mg/kg). A statistically


significant increase in serum ferritin levels, determined as the
difference between baseline values and value at the end of
intervention, was shown in the group of patients treated with DFX
10 mg/kg/day compared with the DFO control group (WMD 801, 95%
CI 565.98 to 1036.02, p b 0.00001) (Fig. 6b) [32]. GRADE quality of
evidence was low.

Comparison of DFX 20 mg/kg with DFO 40 mg/kg. A statistically


significant increase in serum ferritin levels, determined as the difference
between baseline values and the value at the end of intervention, was
shown in the group of patients treated with DFX 20 mg/kg/day
compared with the DFO control group [32] (WMD 328, 95% CI 121.65
to 534.35, p=0.002) (Fig. 6c) (Table 3). GRADE quality of evidence was
low.

Comparison of DFX 30 mg with DFO 51 mg/kg. No statistically


significant difference was observed in the group of patients treated
with DFX 30 mg/kg/day compared with the DFO control group [32]
(WMD 77.00, 95% CI −311.91 to 465.91, p = 0.70) (Fig. 6d) (Table 3).

Outcome 5: Urinary iron excretion

Comparison of DFP plus DFO versus other chelators. Urinary iron


excretion was measured in two trials [29,33] (Aydinok 2007
[urinary iron excretion (mg/kg/day)]; Gomber 2004 [urinary iron
excretion (mg/day)]) in 18 participants who received DFP plus DFO,
and 19 in the control group, which included 12 participants who
received DFP [29] and 7 who received DFO [33] (Fig. 7a). Urinary
iron excretion was significantly higher in the DFP plus DFO groups
(WMD 1.28, 95% CI 0.53 to 2.02, p = 0.0008). GRADE quality of
evidence was low. Heterogeneity for this outcome was not
Fig. 3. Risk of bias graph for each study.
statistically significant (CHI 2 = 1.74, df = 1, p = 0.19, I 2 43%).

Comparison of sequential deferiprone and deferoxamine versus defer-


Heterogeneity was not statistically significant (CHI 2 = 0.35, df = 1, oxamine. Urinary iron excretion was significantly higher in the
p = 0.55, I 2 0%). sequential DFP and DFO groups compared with the DFO control
group [30] (WMD 0.23, 95% CI 0.04 to 0.42, p = 0.02) (Fig. 7b). GRADE
Outcome 3: Myocardial iron concentration (evaluated by MRI T2*) at quality of evidence was very low.
the end of the treatment
Discussion
Comparison of deferiprone plus DFO versus DFO. Myocardial iron
concentration (T2*) at the end of the intervention was not The aim of this systematic review was to evaluate the effectiveness
significantly lower in the DFP plus DFO groups in comparison with of DFO, DFP, and DFX alone or in associated or as sequential therapies
the control group (WMD = 1.7, 95% CI −2.78 to 6.18, p = 0.46) in transfusion-dependent patients with thalassemia major. This meta-
(Fig. 5a) [13]. GRADE quality of evidence was low. analysis suggests that ejection fraction at the end of the intervention
A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175 171

Outcome 1: Ejection Fraction at the end of intervention


(a) Associated deferiprone plus deferoxamine versus other chelators

(b) Sequential deferiprone and deferoxamine versus deferoxamine

Outcome 2: Liver Iron concentration at the end of treatment


(c) Associated Deferiprone plus deferoxamine versus deferiprone

Fig. 4. Foster plot analysis of the outcomes 1 and 2.

was statistically significantly higher after combined (Fig. 4a) or chelation treatment (Fig. 5b). Serum ferritin, measured as the
sequential DFP and DFO treatments (Fig. 4b). Liver iron concentration difference between the final versus the basal value, was statistically
at the end of the intervention was statistically significantly decreased significantly increased after treatment with DFX 5 mg, 10 mg, and
after associated DFP plus DFO treatments (Fig. 4c). Myocardial iron 20 mg (Fig. 6a,b,c), and urinary iron excretion was statistically
concentration (T2*) at the end of the intervention did not show any significantly increased after associated (Fig. 7a) or sequential DFP
statistically significant change after associated (Fig. 5a) or sequential and DFO treatments (Fig. 7b).
172 A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175

Outcome 3: Myocardial Iron Concentration (evaluated by MR T2*) at the end of the intervention
(a) Associated deferiprone plus deferoxamine versus deferoxamine

(b) Sequential deferiprone and deferoxamine versus deferoxamine

Fig. 5. Foster plot analysis of the outcome 3.

Iron-induced heart failure is the most common cause of death in The statistically significantly lower liver iron concentration at the
patients with thalassemia major, accounting for about 70% of deaths end of intervention in the DFP plus DFO groups may be explained by
[3]. Therefore, maintaining left ventricular ejection fraction over the the previously described synergistic action between these two
recommended threshold of 55% [34] may play a critical role in chelators [39,40]. The greater effectiveness of associated DFP plus
preventing heart failure in thalassemia major patients. The detection DFO treatments is explained as the result of a transfer of chelated iron
of a higher ejection fraction at the end of the intervention after from DFP to DFO; i.e. the “shuttle” hypothesis [39,40].
associated (Fig. 4a) or sequential DFP and DFO treatments (Fig. 4b) is Myocardial iron concentration (T2*) at the end of the intervention
consistent with the reported reduction in mortality due to heart was not statistically significantly different in the DFP plus DFO versus
damage during retrospective and prospective studies carried out in the DFO-treated groups [13] (Fig. 5a). However, a different result was
thalassemia major patients treated with DFP [35–38]. reported by Tanner et al. [13] who used a different statistical analysis

Outcome 4: Serum Ferritin as difference between final versus basal values


(a) Deferasirox 5 mg versus deferoxamine

(b) Deferasirox 10 mg versus deferoxamine

(c) Deferasirox 20 mg versus deferoxamine

(d) Deferasirox 30 mg versus deferoxamine

Fig. 6. Foster plot analysis of the outcome 4.


A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175 173

Table 3
Combined chelation treatment: different schedules of administration.

Interventions Control group

Dosage of DFP Dosage of DFO Versus other chelators


(mg/kg/day) (mg/kg)

Aydinok [29] 75 40–50 for Deferiprone 75 mg/kg/day


2 days/week
Gomber [33] 75 40 for 2 days/week Deferoxamine 40 mg/kg/day for 5 days/week
Tanner [13] 75 40–50 for many days Deferoxamine 40–50 mg/kg/day for
minimum 5 days minimum 5 days or more
El_Beshlawy [50] 60–83 23–50 for 2 days/week Deferoxamine 25–50 mg/kg/day for 5 nights
Ha [45] 75 30–60 for 2 days/week Deferoxamine 30–60 mg/kg/day for 5–7 days
Mourad [48] 75 Total of 2 gr for 2 days Deferoxamine 40–50 mg/kg/day for
5–7 days/week

methodology. Actually, the myocardial T2* was log-transformed significant cardiac T2* signal variations can be demonstrated among
before analysis for normalization. The statistical analysis was chelators.
performed over time within the groups from baseline to 12 months. The comparisons between DFX (5 mg), DFX (10 mg), and DFX
Moreover, the analysis was carried out with a mixed-effects model (20 mg) versus DFO (Table 3) showed an increase in serum ferritin
with time as a fixed effect. In contrast, the statistical analysis levels at the end of the study [32]. The three doses of DFX
recommended for meta-analysis is based on the weighted mean examined may be too low to effectively decrease serum ferritin
difference (WMD) between the two groups at only one time point levels in the intervention groups. Furthermore, even the reduction
[41]. This difference in the statistical analysis methodology may in serum ferritin levels by DFX 30 mg/day was not statistically
explain the discrepancy in the results. However, these findings may significantly different compared with DFO treatment [32]. More-
also suggest that larger and more robust multicenter randomized over, Cohen et al. suggested an effect of dose and transfusion in
clinical trials are necessary to determine whether or not clinically DFX- versus DFO-treated patients [42]. A recently published,

Outcome 5 Urinary Iron Excretion


(a) Associated deferiprone plus deferoxamine versus other chelators

(b) Sequential deferiprone and deferoxamine versus deferoxamine

Fig. 7. Foster plot analysis of the outcome 5.


174 A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175

Table 4
Sequential chelation treatment: different schedules of administration.

Interventions Control group

Dosage of DFP Dosage of DFO

mg/kg Days mg/kg Days

Maggio [31] 75 4 50 3 Deferiprone 75 mg/kg/day


Galanello [44] 75 5 N.R. 2 Deferoxamine 34.8 for 5–7 days
Abdelrazik [30] 75 for 4 days a week 40 for 2 days Deferoxamine 40 mg/kg/day for 5–7 days

N.R. = not reported.

prospective, but non randomized escalator trial suggested an ey's Anemia Foundation, Apotex, Apopharma, UK Thalassemia Society,
increased effect with dosage of DFX greater than 30 mg/kg in a University College London Special Trustees Charity, Novartis, Siemens
large cohort of patients [43]. However, because a comparator Medical Solutions, Cardiovascular Imaging Solutions, and the British
group was lacking, it is difficult to evaluate the significance of this Heart Foundation [13].
result [43].
The greater effectiveness of associated and sequential treatments Authorship contributions
in increasing urinary iron excretion could be explained by the “shuttle”
hypothesis [39,40]. AF, AV, GA, AM, and CG designed the research, performed the
This meta-analysis has some important limitations. The quality of research, and prepared an initial draft of the paper.
included randomized trials was very low because only 50% (n = 8) of AK, AC, MDC, SF, PC, RWG, LP, JBP, FB, and FC analyzed the data and
the trials showed adequate sequence generation, whereas only 25% discussed the draft issue by issue.
showed a low risk of bias regarding allocation concealment and AF and AM wrote the final draft of the paper.
blinding, and only 7.4% showed a low risk of bias for incomplete PH and JW addressed scientific and language issues as interna-
outcome data addressed, selective reporting bias, and other bias. tional external reviewers.
Therefore, there were few trials conducted with an optimal design. AI was a representative of the patients who examined the
Furthermore, there were a number of studies excluded from the document.
analysis because they did not reach the minimal criteria to be
considered randomized controlled trials.
Declarations of interest
There were very few trials for each comparison, and the
heterogeneity of the data between the trials together with the
Conflict of interest disclosure: AF, AM, AV, GA, CG, FB, LP, FC, AI,
different treatment schedules (Tables 3–4) made comparisons and
RWG, AC, and SF declare no competing financial interests; MDC, PC,
conclusions difficult.
and JBP report having received consulting fees from Novartis
Pharmaceuticals. PH received research grants from Novartis, and
Implications for practice
support from Novartis and Ferrokin, and participated in medical
advisory boards for Apotex; JW received research grants from
This systematic review with meta-analyses of randomized clinical
Novartis and is a consultant for Ferrokin Biosciences. AK reports
trials does not suggest any change in chelation treatment protocols.
participating in study monitor committees for Novartis Pharmaceu-
Evidence supporting the effectiveness of associated and sequential
ticals, and receiving lecture fees from Novartis Pharmaceuticals and
DFP plus DFO treatment should be confirmed in larger randomized
Apopharma Inc.
clinical trials before having definitive implications for clinical practice.
Finally,these findings suggest as authorization by the national and
international regulatory agencies of the current off-license use of Acknowledgments
associated and sequential DFP plus DFO protocols should be urgently
considered. The research leading to these results has received funding by the
Italian Ministry of Health and the Italian Foundation for the Cure of
Implications for research Thalassaemia “Leonardo Giambrone”, under the project “Inter-
regional Network for Thalassaemia: HTA for the diagnostic and
This review suggests that we need to have (1) larger and longer therapeutic intervention for iron overload”, coordinated by the
randomized clinical trials for each comparison; (2) improvement of Basilicata Region. Prof. David Nathan provided valuable advice that
the quality of future clinical trials; and (3) decreased heterogeneity in was greatly appreciated. We thank Foundation Franco and Piera
the direct and indirect methods for measuring iron overload. Cutino for their continuous encouragement. Finally, we thank Sarah
Louise Klingenberg and Dimitrinka Nikolova for their help in
Support and sponsorships upgrading the literature research.

Nine trials reported the source of funding [11,13,29,31,41,44–47]. References


The trials that received funding from a governmental agency [1] D. Weatherall, The inherited diseases of hemoglobin are an emerging global
included co-funding from the Sicilian Thalassaemic Association and health burden, Blood 115 (2010) 4331–4336.
European Community [47]. [2] B. Modell, M. Darlison, Global epidemiology of haemoglobin disorders and
derived service indicators, Bull. World Health Organ. 86 (2008) 480–487.
The trials that documented support from industry included
[3] C. Borgna-Pignatti, S. Rigolotto, P. De Stefano, H. Zhao, M.D. Cappellini, G.C. Del
Novartis Pharma [11] and Lipomed, Switzerland, for DFP, [29,44] Vecchio, M.A. Romeo, G.L. Forni, M.R. Gamberini, R. Ghilardi, A. Piga, A. Cnaan,
and Novartis Pharmaceuticals Corporation, [46] Apotex, Novartis, Survival and complications in patients with thalassemia major treated with
Apopharma [45]. transfusion and deferoxamine, Haematologica 89 (2004) 1187–1193.
[4] A. Yardumian, P. Telfer, P. Darbyshire, Standards for the Clinical Care of Children
One trial received funding from both non industry and industry; and Adults with Thalassaemia in the UK, United Kingdom Thalassaemia
CORDA, Royal Brompton, Harefield Hospital Charitable Funds, Cool- Society2nd edition, 2008.
A. Maggio et al. / Blood Cells, Molecules, and Diseases 47 (2011) 166–175 175

[5] J. Porter, A. Cohen, J. Kwiatkowski, Transfusion and iron chelation, in: M. of body iron burden in thalassemia major patients, Pediatr. Endocrinol. Rev. 6
Steinberg, B. Forget, D. Higgs, D. Weatheral (Eds.), Disorders of Hemoglobin, (2008) 193–203.
Cambridge University Press, New York, 2009, pp. 689–744. [32] M.D. Cappellini, A. Cohen, A. Piga, M. Bejaoui, S. Perrotta, L. Agaoglu, Y. Aydinok,
[6] A. Maggio, Light and shadows in the iron chelation treatment of haematological A. Kattamis, Y. Kilinc, J. Porter, M. Capra, R. Galanello, S. Fattoum, G. Drelichman,
diseases, Br. J. Haematol. 138 (2007) 407–421. C. Magnano, M. Verissimo, M. Athanassiou-Metaxa, P. Giardina, A. Kourakli-
[7] Electronic Medicines Compendium, Desferal — Summary of Product Character- Symeonidis, G. Janka-Schaub, T. Coates, C. Vermylen, N. Olivieri, I. Thuret, H.
istics. URL: http://emc.medicines.org.ukAccessed 28 September 2006. Opitz, C. Ressayre-Djaffer, P. Marks, D. Alberti, A phase 3 study of deferasirox
[8] EMEA, Ferriprox EPAR. 1999. URL: www.emea.europa.eu/humandocs/Humans/ (ICL670), a once-daily iron chelator; in patients with beta-thalassemia, Blood 107
EPAR/ferriprox/ferriprox.htm. (2006) 3455–3462.
[9] British Medical Association and Royal Pharmaceutical Society of Great Britain, [33] S. Gomber, R. Saxena, N. Madan, Comparative efficacy of desferrioxamine,
British National Formulary (BNF). No. 53, March 2007. London: BMA and RPS; deferiprone and in combination on iron chelation in thalassemic children, Indian
2007. URL: www.bnf.org/bnf/ URL . Pediatr. 41 (2004) 21–27.
[10] EMEA, Ferriprox: Annex I — Summary of Product Characteristics. 2007. URL: [34] T. Cogliandro, G. Derchi, L. Mancuso, M.C. Mayer, B. Pannone, A. Pepe, M. Pili, P.
www.emea.europa.eu/humandocs/Humans/EPAR/ferriprox/ferriprox.htm. Bina, P. Cianciulli, V. De Sanctis, A. Maggio, Guideline recommendations for heart
Accessed 2 February 2007. complications in thalassemia major, J. Cardiovasc. Med (Hagerstown) 9 (2008)
[11] A. Piga, R. Galanello, G.L. Forni, M.D. Cappellini, R. Origa, A. Zappu, G. Donato, E. 515–525.
Bordone, A. Lavagetto, I. Zanaboni, R. Sechaud, N. Hewson, J.M. Ford, H.O. Pitz, [35] C. Borgna-Pignatti, M.D. Cappellini, P. De Stefano, G.C. Del Vecchio, G.L. Forni, M.R.
D. Alberti, Randomized phase II trial of deferasirox (Exjade, ICL670), a once- Gamberini, R. Ghilardi, A. Piga, M.A. Romeo, H. Zhao, A. Cnaan, Cardiac morbidity
daily, orally-administered iron chelator, in comparison to deferoxamine in and mortality in deferoxamine- or deferiprone-treated patients with thalassemia
thalassemia patients with transfusional iron overload, Haematologica 91 major, Blood 107 (2006) 3733–3737.
(2006) 873–880. [36] A. Maggio, A. Vitrano, M. Capra, L. Cuccia, F. Gagliardotto, A. Filosa, M.A. Romeo, C.
[12] R. Origa, P. Bina, A. Agus, G. Crobu, E. Defraia, C. Dessì, G. Leoni, P.P. Muroni, R. Magnano, V. Caruso, C. Argento, C. Gerardi, S. Campisi, P. Violi, R. Malizia, P.
Galanello, Combined therapy with deferiprone and desferrioxamine in thalasse- Cianciulli, M. Rizzo, D.G. D'Ascola, A. Quota, L. Prossomariti, C. Fidone, P. Rigano, A.
mia major, Haematological 90 (2005) 1309–1314. Pepe, G. D'Amico, A. Morabito, G. Gluud, Long-term sequential deferiprone-
[13] M.A. Tanner, R. Galanello, C. Dessi, G.C. Smith, M.A. Westwood, A. Agus, M. deferoxamine versus deferiprone alone for thalassaemia major patients: a
Roughton, R. Assomull, S.V. Nair, J.M. Walker, D.J. Pennell, A randomized, placebo- randomized clinical trial, Br. J. Haematol. 145 (2009) 245–254.
controlled, double-blind trial of the effect of combined therapy with deferox- [37] A. Maggio, A. Vitrano, M. Capra, L. Cuccia, F. Gagliardotto, A. Filosa, C. Magnano, M.
amine and deferiprone on myocardial iron in thalassemia major using Rizzo, V. Caruso, C. Gerardi, C. Argento, S. Campisi, F. Cantella, F. Commendatore,
cardiovascular magnetic resonance, Circulation 115 (2007) 1876–1884. D.G. D'Ascola, C. Fidone, A. Ciancio, M.C. Galati, G. Giuffrida, R. Cingari, G. Giugno,
[14] D.J. Roberts, D. Rees, J. Howard, C. Hyde, P. Alderson, S. Brunskill, Desferrioxamine T. Lombardo, L. Prossomariti, R. Malizia, A. Meo, G. Roccamo, M.A. Romeo, P. Violi,
mesylate for managing transfusional iron overload in people with transfusion- P. Cianciulli, P. Rigano, Improving survival with deferiprone treatment in patients
dependent thalassaemia, Cochrane Database Syst. Rev. (2005). with thalassemia major: a prospective multicenter randomised clinical trial Under
[15] A. Addis, R. Loebstein, G. Koren, T.R. Einarson, Meta-analytic review of the clinical the auspices of the Italian Society for Thalassemia and Hemoglobinopathies, Blood
effectiveness of oral deferiprone (L1), Eur. J. Clin. Pharmacol. 55 (1999) 1–6. Cells Mol. Dis. 42 (2009) 247–251.
[16] M. Franchini, D. Veneri, Iron overload and hematologic malignancies, Hematol. J. 5 [38] B. Modell, M. Khan, M. Darlison, M.A. Westwood, D. Ingram, D.J. Pennell, Improved
(2004) 381–383. survival of thalassaemia major in the UK and relation to T2* cardiovascular
[17] H.E. Vanorden, T.M. Hagemann, Deferasirox — an oral agent for chronic iron magnetic resonance, J. Cardiovasc. Magn. Reson. 10 (2008) 42.
overload, Ann. Pharmacother. 40 (2006) 1110–1117. [39] G. Link, A.M. Konijn, W. Breuer, Z.I. Cabantchik, C. Hershko, Exploring the “iron
[18] L. Abetz, J.F. Baladi, P. Jones, D. Rofail, The impact of iron overload and its shuttle” hypothesis in chelation therapy: effects of combined deferoxamine and
treatment on quality of life: results from a literature review, Health Qual. Life deferiprone treatment in hypertransfused rats with labeled iron stores and in
Outcomes 4 (2006) 73. iron-loaded rat heart cells in culture, J. Lab. Clin. Med. 138 (2001) 130–138.
[19] C. McLeod, N. Fleeman, J. Kirkham, A. Bagust, A. Boland, P. Chu, R. Dickson, Y. [40] P.J. Giardina, R.W. Grady, Chelation therapy in beta-thalassemia: an optimistic
Dundar, J. Greenhalgh, B. Modell, A. Olujohungbe, P. Telfer, T. Walley, Deferasirox update, Semin. Hematol. 38 (2001) 360–366.
for the treatment of iron overload associated with regular blood transfusions [41] D.J. Pennell, V. Berdoukas, M. Karagiorga, V. Ladis, A. Piga, A. Aessopos, E.D. Gotsis,
(transfusional haemosiderosis) in patients suffering with chronic anaemia: a M.A. Tanner, G.C. Smith, M.A. Westwood, B. Wonke, R. Galanello, Randomized
systematic review and economic evaluation, Health Technol. Assess. 13 (2009) controlled trial of deferiprone or deferoxamine in betathalassemia major patients
1–121. with asymptomatic myocardial siderosis, Blood 107 (2006) 3738–3744.
[20] Malaysian Health Technology Assessment Unit Management of Thalassemia Kuala [42] A.R. Cohen, E. Glimm, J.B. Porter, Effect of transfusional iron intake on response to
Lumpur, Malaysian Health Technology Assessment Unit MHTAU, 2003. chelation therapy in β-thalassemia major, Blood 111 (2008) 583–587.
[21] J.J. Caro, K.F. Huybrechts, T.C. Gree, Estimates of the effect on hepatic iron of oral [43] A. Taher, A. El-Beshlawy, M.S. Elalfy, K. Al Zir, S. Daar, D. Habr, U. Kriemler-Krahn,
deferiprone compared with subcutaneous desferrioxamine for treatment of iron A. Hmissi, A. Al Jefri, Efficacy and safety of deferasirox, an oral iron chelator, in
overload in thalassemia major: a systematic review, BMC Blood Dis. 2 (2002) 4. heavily iron-overloaded patients with beta-thalassaemia: the ESCALATOR study,
[22] L. Kjaergaard, J. Villumsen, C. Gluud, Reported methodological quality and Eur. J. Haematol. 82 (2009) 458–465.
discrepancies between small and large randomized trials in meta-analyses, [44] R. Galanello, A. Kattamis, A. Piga, R. Fischer, G. Leoni, V. Ladis, V. Voi, U. Lund, F.
Annals of Intern. Med. 135 (2001) 982–989. Tricta, A prospective randomized controlled trial on the safety and efficacy of
[23] D. Moher, B. Pham, A. Jones, D.J. Cook, A.R. Jadad, M. Moher, P. Tugwell, T.P. alternating deferoxamine and deferiprone in the treatment of iron overload in
Klassen, Does quality of reports of randomised trials affect estimates of patients with thalassemia, Haematological 91 (2006) 1241–1243.
intervention efficacy reported in meta-analyses, Lancet 352 (1998) 609–613. [45] S.Y. Ha, K.W. Chik, S.C. Ling, A.C. Lee, C.W. Luk, C.W. Lam, I.O. Ng, G.C. Chan, A
[24] L. Wood, M. Egger, L.L. Gluud, K.F. Schulz, P. Jüni, D.G. Altman, C. Gluud, R.M. randomized controlled study evaluating the safety and efficacy of deferiprone
Martin, A.J. Wood, J.A. Sterne, Empirical evidence of bias in treatment effect treatment in thalassemia major patients from Hong Kong, Hemoglobin 30 (2006)
estimates in controlled trials with different interventions and outcomes: meta- 263–274.
epidemiological study, Br. Med. J. 336 (2008) 601–605. [46] E. Nisbet-Brown, N.F. Olivieri, P.J. Giardina, R.W. Grady, E.J. Neufeld, R. Sèchaud, A.J.
[25] M. Crowther, W. Lim, M.A. Crowther, Systematic review and meta-analysis Krebs-Brown, J.R. Anderson, D. Alberti, K.C. Sizer, D.G. Nathan, Effectiveness
methodology, Blood 116 (2010) 3140–3146. and safety of ICL670 in iron-loaded patients with thalassaemia: a randomised,
[26] Review Manager, Review Manager (RevMan) [Computer program]. Version 5.0, double-blind, placebo-controlled, dose-escalation trial, Lancet 361 (2003)
The Nordic Cochrane Centre, The Cochrane Collaboration, Copenhagen, 2008. 1597–1602.
[27] Handbook rev man 5.0 (2008), http://ims.cochrane.org/revman/documentation. [47] A. Maggio, G. D'Amico, A. Morabito, M. Capra, C. Ciaccio, P. Cianciulli, F. Di
[28] D. Atkins, M. Eccles, S. Flottorp, G.H. Guyatt, D. Henry, S. Hill, D. Henry, S. Hill, Gregorio, G. Garozzo, R. Malizia, C. Magnano, A. Mangiagli, G. Quarta, M. Rizzo,
A. Liberati, D. O'Connell, A.D. Oxman, B. Phillips, H. Schünemann, T.T. Edejer, D.G. D'Ascola, A. Rizzo, M. Midiri, Deferiprone versus deferoxamine in patients
G.E. Vist, J.W. Williams, GRADE Working Group, Systems for grading the with thalassemia major: a randomized clinical trial, Blood Cells Mol. Dis. 28
quality of evidence and the strength of recommendations I: critical appraisal (2002) 196–208.
of existing approaches The GRADE Working Group, BMC Health Serv. Res. 4 [48] F.H. Mourad, A.V. Hoffbrand, M. Sheikh-Taha, S. Koussa, A.L. Khoriaty, A. Taher,
(2004) 38. Comparison between desferrioxamine and combined therapy with desferriox-
[29] Y. Aydinok, Z. Ulger, D. Nart, A. Terzi, N. Cetiner, G. Ellis, A. Zimmermann, C. Manz, amine and deferiprone in iron overloaded thalassaemia patients, Br. J. Haematol.
A randomized controlled 1-year study of daily deferiprone plus twice weekly 121 (2003) 187–189.
desferrioxamine compared with daily deferiprone monotherapy in patients with [49] N. Yarali, T. Fisgin, F. Duru, A. Kara, N. Ecin, S. Fitoz, I. Erden, Subcutaneous bolus
thalassemia major, Haematologica 92 (2007) 1599–1606. injection of deferoxamine is an alternative method to subcutaneous continuous
[30] N. Abdelrazik, Pattern of iron chelation therapy in Egyptian beta thalassemic infusion, J. Pediatric. Hematol. Oncol. 28 (2006) 11–16.
patients: Mansoura University Children's Hospital experience, Hematology 12 [50] A. El-Beshkawy, C. Manz, M. Naja, M. Eltagui, C. Tarabishi, I. Youssry, H. Sobh, M.
(2007) 577–585. Hamdy, I. Sharaf, A. Mostafa, O. Shaker, A.V. Hoffbrand, A. Taher, Iron chelation in
[31] A. Maggio, M. Capra, A. Pepe, L. Mancuso, E. Cracolici, S. Vitabile, P. Rigano, F. thalassemia: combined or monotherapy ? The Egyptian experience, Ann.
Cassarà, M. Midiri, A critical review of non invasive procedures for the evaluation Hematol. 87 (2008) 545–550.

You might also like