You are on page 1of 10

Vox Sanguinis (2017)

© 2017 The Authors.


ORIGINAL PAPER Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
DOI: 10.1111/vox.12477

Single-dose intravenous iron infusion or oral iron for


treatment of fatigue after postpartum haemorrhage:
a randomized controlled trial
C. Holm,1,2 L. L. Thomsen,2 A. Norgaard3 & J. Langhoff-Roos1
1
Department of Obstetrics, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
2
Pharmacosmos A/S, Holbaek, Denmark
3
Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark

Background and Objectives To evaluate the clinical efficacy of a single-dose


intravenous infusion of iron isomaltoside compared with current treatment prac-
tice with oral iron measured by physical fatigue in women after postpartum
haemorrhage.
Materials and Methods Single-centre, open-label, randomized controlled trial.
Participants received intravenous iron (n = 97) or oral iron (n = 99), and com-
pleted the Multidimensional Fatigue Inventory and Edinburgh Postnatal Depres-
sion Scale, and haematological and iron parameters were measured. Primary
outcome was the aggregated change in physical fatigue score from baseline to
12 weeks postpartum.
Results The difference in physical fatigue score was -097 (95% CI: -165; -028,
P = 0006) in favour of intravenous iron, but did not meet the predefined differ-
ence of 18. Across visits, we found statistically significant differences in fatigue
and depression scores, as well as in haematological and iron parameters, all in
favour of intravenous iron. There were no serious adverse reactions.
Conclusion A single dose of intravenous iron was associated with a statisti-
cally significant reduction in aggregated physical fatigue within 12 weeks after
postpartum haemorrhage compared to standard medical care with oral iron
below the prespecified criteria of clinical superiority. As patient-reported out-
comes improved significantly and intravenous iron resulted in a fast
hematopoietic response without serious adverse reactions, intravenous iron
may be a useful alternative after postpartum haemorrhage if oral iron is not
absorbed or tolerated.
Received: 20 May 2016,
revised 19 October 2016, Key words: anaemia, intravenous iron, iron deficiency, iron isomaltoside, postpar-
accepted 1 November 2016 tum fatigue, postpartum haemorrhage.

Introduction
Correspondence: Charlotte Holm, Department of Obstetrics, Juliane
Postpartum haemorrhage (PPH) may lead to iron defi-
Marie Centre, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, ciency and anaemia. Postpartum iron-deficiency anaemia
Copenhagen DK-2100, Denmark is associated with clinical symptoms, most prominently
E-mail: charlotteholm@dadlnet.dk maternal fatigue [1]. In women of reproductive age, iron
Clinical trial registration deficiency without anaemia is associated with fatigue,
The trial was registered at EU Clinical Trials Register. EudraCT Number: impaired physical work performance, deficient cognitive
2012-005782-12. www.clinicaltrialsregister.eu

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial License,
which permits use, distribution and reproduction in any medium, provided the original work is properly cited and 1
is not used for commercial purposes.
2 C. Holm et al.

functions and mood disturbances, and it is plausible that delivery. The exclusion criteria included multiple births,
sufficient iron supplementation is advantageous for these peripartum RBC transfusion and history of multiple aller-
women [2, 3]. gies. All exclusion criteria are listed in the published trial
In Denmark, the current treatment practice for women protocol [16].
after PPH is oral iron supplementation. Red blood cell Participants were randomly assigned to receive either
(RBC) transfusion is only indicated for severe symptoms iron isomaltoside (i.v.-iron group) or current treatment
of anaemia and haemoglobin (Hb) values below 7 g/dl, practice (oral iron group) using an interactive web
according to the Danish national guidelines for transfu- response system (eClinical OS, Merge Healthcare,
sion [4]. Morrisville, NC, United States). The randomization was
Ten randomized controlled studies compared intra- 1:1 stratified by bleeding volume (700–1000 ml or
venous (i.v.) iron to oral iron in women with postpartum >1000 ml).
iron-deficiency anaemia [5–14]. They found that i.v. iron The i.v.-iron group received a single dose of 1200 mg
was superior by Hb and iron parameters, but none of iron isomaltoside (Monofer, Pharmacosmos A/S, Holbaek,
the trials primarily measured the effect of i.v. iron Denmark) diluted in 100 ml of 09% sodium chloride, and
administration on clinical outcomes [15]. Fatigue and infused intravenously within 15 min.
psychological well-being were reported as secondary The oral iron group received current treatment practice:
outcomes by two studies. Westad et al. [6] reported a a recommendation to continue oral iron supplementation,
statistically significant improvement in the mean change as during pregnancy (the Danish Health and Medicines
from baseline to weeks 4, 8 and 12 for physical, mental Authority recommends 40–50 mg oral iron supplementa-
and total fatigue in the group receiving i.v. iron treat- tion daily [17]), or to take 100 mg oral iron one or two
ment measured by the Fatigue Scale. Van Wyck et al. times daily for a variable time period. The individual
[8] used the Fatigue Linear Analog Scale Assessment for intake of elemental oral iron, including the type of prepa-
a mean total fatigue score measured at weeks 2, 4 and 6 ration, dose and treatment duration, was monitored
and showed no difference in fatigue. Both studies used throughout the study period.
the SF-36 questionnaire to measure psychological well- The randomized participants had six visits within
being and found no difference between the treatment 12 weeks: at inclusion in the hospital and five visits at
groups. home, 3 days, and one, three, eight and 12 weeks after
The objective of this study was to evaluate clinical effi- inclusion.
cacy of single-dose infusion of iron isomaltoside com- The primary outcome was the aggregated change in
pared with current treatment practice with oral iron physical fatigue score within 12 weeks postpartum, as
measured by physical fatigue in women with postpartum measured by the physical fatigue subscale of the Multidi-
haemorrhage (PPH). This study tested the hypothesis that mensional Fatigue Inventory (MFI) [18]. Secondary effi-
i.v. administration of iron isomaltoside is clinically supe- cacy outcomes included changes in Hb, ferritin, iron,
rior to current treatment practice with oral iron supple- transferrin, transferrin saturation, reticulocyte count and
mentation, measured by self-reported physical fatigue as reticulocyte mean haemoglobin content (CHr), other
the primary outcome. dimensions of fatigue by MFI and symptoms of depres-
sion measured by the Edinburgh Postnatal Depression
Scale (EPDS) [19], time to lactogenesis, time to discontin-
Materials and methods
uation of breastfeeding and transfusion of ‘rescue’ allo-
This was a single-centre, open-label, randomized, parallel geneic RBCs. We monitored vital signs before, during and
superiority study with a 1:1 allocation ratio conducted at after infusion in the i.v.-iron group and recorded any
the Department of Obstetrics, Rigshospitalet, University of adverse events and laboratory safety parameters listed in
Copenhagen, Denmark. The study was approved by the the published trial protocol [16].
National Committee on Biomedical Research Ethics on 12 The MFI evaluates five dimensions of fatigue: general
April 2013, approval number: H-4-2013-019, and by the fatigue, physical fatigue, reduced activity, reduced moti-
Danish Medicines Agency (approval number: EudraCT vation and mental fatigue. The MFI has high feasibility,
2012-005782-12). reliability and validity in chronically anaemic and post-
From 3 May 2013 to 18 September 2014, we assessed partum women [20, 21]. High scores indicate a high
the eligibility of all parturients, regardless of mode of degree of fatigue. The EPDS detects symptoms of depres-
delivery, with PPH ≥ 700 ml. Women were enrolled in sion in puerperal women during the previous 7 days [22].
the study if they fulfilled the inclusion criterion: The maximum score is 30, and a score of 10 or higher
PPH ≥ 700 and ≤1000 ml; or PPH > 1000 ml and Hb indicates possible depression. The MFI was completed at
>65 g/dl (40 mmol/l) measured at least 12 h after all visits and the EPDS at the last five visits.

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
Iron treatment after postpartum haemorrhage 3

The sample size was determined according to the pri- Activities (version 16.0). Related and possibly related
mary hypothesis that iron isomaltoside is superior to adverse events were defined as adverse drug reactions.
the current treatment practice with oral iron. The null
hypothesis of no difference between groups was tested
Results
against the alternative by constructing a two-sided 95%
confidence interval (CI) of the difference in aggregated A total number of 8860 women gave birth during the
change in physical fatigue score from baseline to 18-month study period. Twelve per cent of the women
12 weeks postpartum. The use of the physical fatigue (n = 1001) experienced a PPH ≥ 700 ml (Fig. 1). Some
subscale of MFI allowed a maximum change of 16 women were discharged before being informed of the trial
points. We chose a difference greater than 10% in (n = 150). The remaining 851 women were assessed for
physical fatigue score for claiming clinically relevant eligibility, and 389 met the eligibility criteria and were
superiority, corresponding to a difference of 18 (abso- asked to participate in the study. Two hundred women
lute value) with a SD of 42, based on a previous study gave their written consent within 48 hours after delivery
[21]. Based on these presumptions, we needed 87 and were randomized to the i.v.-iron group (n = 100) or
women per treatment group to demonstrate superiority the oral iron group (n = 100). The safety population
with a power of 80%. With a margin for missing data included 198 women who received study treatment. The
and a dropout rate of approximately 10%, 200 women two women in the i.v.-iron group who did not receive
were included. study treatment withdrew their consent before interven-
All statistical methods were prespecified in a statistical tion. The full analysis set consisted of 196 women who
analysis plan and described in detail in the published received the study drug and had at least one postbaseline
trial protocol [16]. The statistical analyses were per- physical fatigue score. Two women were excluded from
formed using SAS version 9.4 (SAS Institute, Cary, NC, the full analysis set. One woman in the i.v.-iron group
USA). All statistical tests were two-sided and performed withdrew her consent due to subcutaneous iron infusion,
on a 5% significance level. Demographic and efficacy and one woman in the oral iron group did not have a
analyses included all randomized participants who measurement of baseline physical fatigue score. The per-
received the study drug and had at least one postbase- protocol population consisted of 191 participants who
line physical fatigue score (full analysis set). Demo- received at least 80% of the planned intravenous iron
graphics and baseline characteristics were primarily dose and did not receive ‘rescue’ RBC transfusion after
summarized using descriptive statistics, and chi-square inclusion.
tests were applied if group differences were suspected. Baseline characteristics are presented in Table 1. There
The aggregated change in physical fatigue score was were 34 emergency caesarean deliveries in the oral iron
calculated as the area under the curve (AUC) of the group compared with 25 in the i.v.-iron group; 75% vs.
change from baseline to 12 weeks, using the trapezoidal 64% of the participants were primipara. These differences
method adjusted for the observation period. The primary were not significant. The mean (–SD) total iron intake in
end-point was analysed using an analysis of variance the oral iron group was 4784 – 4309 mg. Table 2 sum-
model (ANOVA), with treatment and bleeding volume marizes the elemental iron intake in the oral iron group
(700–1000 ml, >1000 ml) as factors and baseline MFI throughout the study period.
physical fatigue score as the covariate. We also per-
formed a per-protocol analysis that excluded participants Patient-reported outcomes
who received less than 80% of the planned intravenous
iron dose or received ‘rescue’ allogeneic RBC transfusion The difference between the two treatment groups in the
after inclusion. change in aggregated physical fatigue from baseline to
Continuous secondary end-points were analysed by a 12 weeks postpartum was -097 (CI 95% -165; -028)
mixed model for repeated measurements, and the estima- (P = 0006) in favour of i.v. iron (Table 3). Sensitivity
tion method was a restricted maximum likelihood-based analysis of the per-protocol population resulted in an
approach. ‘Proportion’ end-points were analysed using unchanged statistically significant difference between the
logistic regression. Where relevant, the baseline value of two treatment groups. The aggregated change in physical
the parameter in question was included as the covariate. fatigue between the two treatment groups differed, when
Time to lactogenesis and time to discontinuation of the data were stratified by PPH level. The difference was
breastfeeding were compared between treatments by a greater and statistically significant in the group of women
log-rank test. Safety analyses included all women who with PPH > 1000 ml -113 (CI 95% -210; -015)
received study treatment. Adverse events were summa- (P = 002), and smaller and non-significant in the group
rized using the Medical Dictionary for Regulatory of women with PPH 700–1000 ml -081 (CI 95% -178;

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
4 C. Holm et al.

Fig. 1 Study flow diagram. PPH, mpostpartum haemorrhage; Hb, haemoglobin; RBC, red blood cell; HELLP, haemolysis, elevated liver enzymes, low
platelet count.

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
Iron treatment after postpartum haemorrhage 5

Table 1 Baseline characteristics of intravenous iron and oral iron group. The fatigue and depression scores decreased continuously
Results are presented as mean (–SD), unless otherwise stated in both treatment groups during the 12 weeks, with over-
all, statistically significant lower scores the i.v.-iron
Intravenous Oral
group.
iron group iron group
Baseline characteristics (n = 97) (n = 99)
Haematological and iron parameters
Age (years) 322 (–44) 326 (–45)
Prepregnancy weight (kg) 668 (–147) 645 (–90) The mean baseline Hb was 971 g/dl in both treatment
PPH (ml), median (min; max) 1000 (700; 3100) 1050 (700; 2800) groups. The increase in mean Hb from baseline to all
PPH 700–1000/1000 (n, %) following time-points was significantly higher in the
700–1000 ml 49 (505) 49 (495)
i.v.-iron group (Fig. 3). All participants in both treatment
>1000 ml 48 (495) 50 (505)
groups reached anaemia correction during the study per-
Primiparas (n, %) 62 (639) 74 (747)
Iron supplementation
iod with no between-group difference in the median time
in pregnancy to anaemia correction.
Whole pregnancy 63 (649) 73 (737) We found an increase in reticulocyte count within the
Part of pregnancy 27 (278) 24 (242) first week in both treatment groups; however, the increase
No 6 (62) 2 (20) was significantly higher in the i.v.-iron group after 3 days
Gestational age (days) 281 (–10) 280 (–10) and 1 week, compared with the oral iron group. The differ-
Birth weight (g) 3705 (–551) 3599 (–499) ence in mean CHr between the two treatment groups was
Mode of labour (n, %) significant at all visits after baseline, in favour of i.v. iron.
Spontaneous 49 (505) 55 (556) Serum ferritin increased promptly and significantly in
Induced 38 (392) 31 (313)
the i.v.-iron group but remained unchanged at all time-
Mode of delivery (n, %)
points in the oral iron group (Fig. 4). At 12 weeks, the
Spontaneous vaginal delivery 57 (588) 40 (404)
Vacuum extraction 6 (62) 15 (152)
mean ferritin remained at a level indicating replenished
Elective caesarean delivery 10 (103) 10 (101) iron stores in the i.v.-iron group, whereas iron stores
Emergency caesarean delivery 24 (247) 34 (343) remained in the lower end of normal range in the oral iron
Perineal tear grade III–IV (n, %) 5 (52) 5 (51) group (176 vs. 37 ng/ml). Significant difference in transfer-
Epidural pain relief (n, %) 36 (371) 36 (364) rin saturation in favour of iron isomaltoside was noted in
Manual exploration of 32 (330) 31 (313) the mean change from baseline to all study time-points.
uterine cavity (n, %)

Lactogenesis and breastfeeding


016) (P = 010). However, the study was not powered for There was no significant difference in the median time
subgroup analysis. In the oral iron group, we found no (hours) to lactogenesis between the i.v.-iron group and
correlation between the total iron intake and aggregated the oral iron group (719 vs. 756; P = 078). Most partic-
physical fatigue from baseline to 12 weeks (P = 089). ipants were still breastfeeding at 12 weeks postpartum
At all time-points from 3 days to 8 weeks postpartum, (845% in the i.v.-iron group and 878% in the oral iron
the baseline-adjusted mean physical fatigue scores were group). Among the participants who discontinued breast-
statistically significantly lower (by 09–12) in the feeding, there were no between-group difference in the
i.v.-iron group compared with the oral iron group (Fig. 2). time of discontinuation (P = 052).

Table 2 Oral iron intake of oral elemental iron in the oral iron group. Data are presented as mean (–SD) daily intake of oral elemental iron between the
planned visits at baseline, 3 days, 1, 3, 8 and 12 weeks

Daily oral iron BL–day 3 Day 4–week 1 Day 8–week 3 Day 22–week 8 Day 57–week 12

Dose (mg) 446 (579) 810 (723) 789 (634) 591 (592) 364 (511)
Number of women per dose group (n, %)
0 mg 36 (364) 8 (81) 3 (30) 8 (81) 40 (404)
1–50 mg 33 (333) 45 (455) 47 (475) 53 (535) 36 (364)
51–100 mg 17 (172) 25 (253) 33 (333) 27 (273) 19 (192)
101–300 mg 13 (131) 21 (212) 16 (162) 11 (111) 4 (40)

n, number of participants, BL, baseline.


Data are presented as mean (–SD) unless otherwise stated.

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
6 C. Holm et al.

Table 3 Change in aggregated physical fatigue from baseline to 12 weeks postpartum. Results from an analysis of AUC of change in physical fatigue
score measured by a subscale of the Multidimensional Fatigue Inventory in the full analysis set and per-protocol population

Contrast: i.v.-iron group – oral iron group

n LsMean Estimate (95% CI) P value

Full analysis set


Intravenous iron group 97 -360 -097 (-165; -028) 0006
Oral iron group 99 -263
BY PPH level
PPH 700–1000 ml
Intravenous iron group 49 -346 -081 (-178; 016) 0102
Oral iron group 49 -265
PPH >1000 ml
Intravenous iron group 48 -374 -113 (-210; -015) 0024
Oral iron group 50 -261

AUC, area under the curve, i.v., intravenous, n, number in analysis set, LsMean, least square mean, CI, confidence interval, PPH, postpartum haemorrhage.
The analysis is from an ANOVA model with treatment and PPH level as factors and baseline physical fatigue score as covariate.
An absolute difference of 18 was the minimal clinical relevant difference for claiming superiority.

(a) (b)

(c) (d)

(e) (f)

Fig. 2 Fatigue and depression. Results are


shown as mean scores of the Multidimensional
Fatigue Inventory, and Edinburgh Postnatal
Depression Scale in the i.v.-iron and oral iron
groups from baseline to 12 weeks postpartum.
Whiskers indicate standard error. Between-
group comparisons: *P < 005. Solid line,
I.v.-iron group; dashed line, oral iron group.

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
Iron treatment after postpartum haemorrhage 7

Fig. 3 Mean haematological parameters in the intravenous iron and oral iron group from baseline to 12 weeks postpartum. Whiskers indicate standard
error. *P < 005 in analysis of change from baseline. Solid line, I.v.-iron group; dashed line, oral iron group. Ret, reticulocytes, RBC, red blood cell, CHr,
reticulocyte haemoglobin content.

Fig. 4 Mean iron biochemical parameters in


the intravenous iron and oral iron group from
baseline to 12 weeks postpartum. Whiskers
indicate standard error. *P < 005 in analysis of
change from baseline. Solid line, I.v.-iron group;
dashed line, oral iron group.

incidence of adverse drug reactions were gastrointestinal


RBC transfusion
disorders (10% and 220%, respectively) and general disor-
‘Rescue’ RBC transfusions occurred in both groups. One ders and administration-site conditions (122% and 00%,
woman with symptoms of severe anaemia in the i.v.-iron respectively) (Table 4). Two women presented with back
group received two units. In the oral iron group, two women and chest pain during infusion that abated spontaneously
with symptoms of severe anaemia received two units, and over a few minutes without change in vital signs during
one woman with secondary PPH received four units. infusion and without signs of allergic or anaphylactic reac-
tions. The infusion was restarted in one participant without
recurrence of symptoms. The second participant did not
Safety
wish to restart the infusion. The mean values of safety lab-
The frequencies of adverse events of any cause were similar oratory parameters over time showed no between-group
in the two treatment groups. Serious adverse events differences. We found transient increased values of alanine
occurred in 92% of the women in the i.v.-iron group and aminotransferase and aspartate aminotransferase without
80% of the women in the oral iron group. None of the seri- clinical symptoms in one participant in the i.v.-iron group
ous adverse events was considered drug-related. There were measured one and 3 weeks after treatment, and in two par-
no deaths in either group. Adverse drug reactions occurred ticipants in the i.v.-iron group and in three participants in
in 133% of the women in the i.v.-iron group and 220% in the oral iron group at 12 weeks. These findings were classi-
the oral iron group. The body systems with the highest fied as non-drug-related. There were five participants in

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
8 C. Holm et al.

Table 4 Adverse drug reactions. Defined as related and possibly related This is to our knowledge the first randomized con-
adverse events in the safety population trolled trial to primarily focus on patient-reported out-
comes in comparing treatment options for postpartum
I.v.-iron group Oral iron group
iron deficiency and anaemia, and a major strength is the
Adverse drug reactions (n = 98) (n = 100)
almost complete follow up with minimal missing data.
Total adverse drug reactions (n, %) 13 (133) 22 (220) Clinical relevance was ensured by the choice of amount
Gastrointestinal disorders 1 (10) 22 (220) of postpartum bleeding as inclusion criteria instead of
Constipation 18 (180) biochemical parameters for iron deficiency and anaemia,
Haemorrhoids 7 (70) which are unreliable shortly after delivery due to
Paraesthesia oral 1 (10) haemodilution and falsely raised acute phase reactants
General disorders and 12 (122)
[16, 23]. As we know the amount of bleeding within
administration-site conditions
hours after delivery, we could administer i.v. iron before
Infusion site discolouration 3 (31)
Infusion site irritation 2 (20)
an early discharge from the hospital.
Infusion site reaction 2 (20) The choice of an individualized oral iron treatment reg-
Paina 2 (20) imen, where the participants in the oral iron group
Puncture site swelling 2 (20) adjusted their oral iron medication to suit their specific
Pyrexia 1 (10) needs and tolerance as comparator to i.v. iron in this
Musculoskeletal and connective 1 (10) study, may be seen as a limitation due to the low mean
tissue disorders iron intake in the study period. However, as gastrointesti-
Myalgia 1 (10) nal side-effects to oral iron supplementation are well
Vascular disorders 1 (10) known and result in poor compliance, especially in puer-
Phlebitis 1 (10)
peral women [24], the individualized treatment regimen
I.v., intravenous, n, number of participants experiencing the event at least simulates normal clinical practice and ensures high exter-
once. nal validity, as opposed to a fixed dosing regimen that
a
Acute back, neck, and chest pain during infusion that abated sponta- results in poor compliance outside a clinical study setting.
neously over a few minutes (Fishbane reaction). An inherent weakness is the open-label design. The
individualized regimen design implies that we were not
the i.v.-iron group and two in the oral iron group with able to blind the randomization. Also, i.v. iron is a black
phosphate levels <2 mg/dl at any time after baseline. All fluid and as iron tablets colour the stools, we thought
phosphate values <2 mg/dl were between 155 and that a double-blinded design would not be feasible.
198 mg/dl, and the phosphate levels in all cases increased Accordingly, most previously published randomized con-
to above 2 mg/dl at the subsequent visit. trolled trials with iron treatment of women after child-
birth have not been double-blinded [6–14]. The clinical
outcomes from the self-reported questionnaires may have
Discussion
been influenced by participants’ knowledge of which
We compared a single high-dose infusion of iron isomal- treatment they received, and thus, we cannot exclude a
toside to current treatment practice with oral iron supple- placebo effect in the intravenous iron group. The study
mentation primarily measured by the aggregated change outcome might have been stronger, if physical tests had
in physical fatigue within 12 weeks after PPH and found been included, such as exercise capacity or muscle
a between-group difference less than the predefined mini- strength measurements.
mal clinically relevant difference. Thus, superiority of A recent systematic review regarding treatment of
iron isomaltoside was not demonstrated in this study. postpartum iron-deficiency anaemia suggests a need for
However, the between-group differences in fatigue and trials with focus on clinical outcomes [15]. We chose the
depression symptoms, as well as haematological and primary outcome physical fatigue measured by a subscale
iron-related biochemical parameters, were statistically of the MFI, as Jansen (2007) found this measure to be
significant in favour of i.v. iron, particularly in the first associated with low Hb [21]. As it is difficult to predefine
3 weeks. The frequency of adverse events was similar a single time-point for measuring treatment effect on
between groups, and no subject had drug-related serious clinical symptoms postpartum, we defined the primary
adverse reactions. Injection-site and general reactions end-point as the aggregated physical fatigue score calcu-
occurred in 12% in the intravenous group, but iron infu- lated by the AUC.
sion was otherwise well tolerated. The oral iron group Based on clinical judgement, we chose a difference
had a high frequency of gastrointestinal adverse reactions greater than 10% in the participant’s perception of physi-
of 22%, in spite of the individual dosing regimen. cal fatigue for claiming clinically relevant superiority in

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
Iron treatment after postpartum haemorrhage 9

the comparison of treatment options; however, the mini-


mal clinically relevant difference for the MFI has yet to Conclusion
be established for women in the puerperium. A single dose of intravenous iron was associated with a
The patient-reported outcomes correspond to the statistically significant reduction in aggregated physical
haematological response where i.v. iron was signifi- fatigue within 12 weeks after PPH compared to standard
cantly more effective at improving iron stores and medical care with oral iron, below the prespecified criteria
allowing erythropoiesis with a fast onset, consistent of clinical superiority.
with previous trials [5–14]. The results suggest that i.v. However, iron isomaltoside was also associated with a
iron treatment after PPH provides a more rapid effect statistically significant improvement in depression scores
compared to current treatment practice with oral iron. and a significantly faster haematopoietic response and
This effect gradually tended to equalize along the study replenishment of iron stores. There was no difference in
period, but differences were still significant at 12 weeks the frequency of adverse events between the two treat-
postpartum. ment groups, and there were no serious adverse reactions.
As described in the published trial protocol, the Hb Gastrointestinal disorders were predominant in the oral
level is inaccurate for calculating individual dosing of group, whereas general and administration-site conditions
iron supplementation immediately after acute bleeding were predominant in the intravenous iron group.
such as PPH [16]. Hence, the iron isomaltoside fixed dose Based on these findings, we conclude that intravenous
of 1200 mg in this study is based on the expected iron iron is a useful treatment alternative for the treatment of
deficiency in women after PPH. Body iron stores are postpartum physical fatigue or depression, if oral iron is
approximately 500–750 mg and the average iron loss not optimal, for example in case of intolerance to or mal-
through PPH is approximately 500 mg, as 500 ml blood absorption of oral iron. Intravenous iron may also be use-
loss equals 250 mg iron loss [25]. This dose ensures ful for fast correction of iron deficiency and/or anaemia,
replenishment of iron stores throughout the 12 weeks and after major postpartum haemorrhage, to avoid blood
and reduces the risk of recurrence of iron-deficiency transfusion.
anaemia once menses resumes and during subsequent
pregnancies.
Acknowledgements
In two women, we observed the acute reaction (back
and chest pain) to i.v. iron described as the Fishbane We would like to thank the project midwives (Trine
reaction [26, 27]. The pathogenesis is unknown, but Markussen Larsen, Katrine Høybye and Sara Winther) and
according to Rampton (2014), it is believed to be a com- Jens K Slott Jensen, MSc, who provided statistical sup-
plement activation-related pseudo-allergy (CARPA) port. Charlotte Holm and Jens Langhoff-Roos collected
triggered by iron nano-particles [28, 29]. the trial data. Charlotte Holm, Jens Langhoff-Roos, Astrid
The finding of a few cases of transient phosphate levels Norgaard and Lars Lykke Thomsen designed the study,
slightly below 2 mg/dL in the i.v.-iron group is in con- analysed and interpreted the data. Charlotte Holm drafted
trast to findings in another i.v.-iron study, in which the manuscript, and all authors approved the final ver-
hypophosphatemia was reported in 8–70% of women in sion. Hence, the sponsor was involved in the design, anal-
postpartum and heavy uterine bleeding studies with yses and interpretation of the data and approval of the
another i.v.-iron preparation [30, 31]. final manuscript.

References
1 Lee KA, Zaffke ME: Longitudinal parameters in non-anaemic iron-defi- 850B3664.ashx [Last accessed 5
changes in fatigue and energy during cient women: a randomised placebo- November 2015]
pregnancy and the postpartum period. controlled study. Br J Obstet Gynaecol 5 Merino SG, Picado AL, Hernandez HM,
J Obstet Gynecol Neonatal Nurs 1999; 2005; 112:445–450 et al.: Ensayo clınico aleatorizado para
28:183–191 4 Sundhedsstyrelsen. National klinisk evaluar la efectividad de dos vıas de
2 Bodnar LM, Cogswell ME, McDonald retningslinje om indikation for administracion de hierro, oral e intra-
T: Have we forgotten the significance transfusion med blodkomponenter venosa, en el tratamiento de la anemia
of postpartum iron deficiency? Am J [Internet]. Copenhagen: Sund- ferropenica posparto. Clınica Investi-
Obstet Gynecol 2005; 193:36–44 hedsstyrelsen; 2014. Available from gacion Ginecol Obstetricia 2012;
3 Krafft A, Perewusnyk G, Hanseler E, Sundhedss tyrelsen: https://sund- 39:190–195 [in Spanish]
et al.: Effect of postpartum iron sup- hedsstyrelsen.dk/da/udgivelser/2014/~/ 6 Westad S, Backe B, Salvesen K A, et al.:
plementation on red cell and iron media/EEA1EA90C15E4A97B9E786D2 A 12-week randomised study comparing

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)
10 C. Holm et al.

intravenous iron sucrose versus oral fer- Cochrane Database Syst Rev 2015; (8): 23 Milman N: Postpartum anemia I:
rous sulphate for treatment of postpar- CD010861 definition, prevalence, causes, and
tum anemia. Acta Obstet Gynecol Scand 16 Holm C, Thomsen LL, Norgaard A, consequences. Ann Hematol 2011;
2008; 87:916–923 et al.: Intravenous iron isomaltoside 90:1247–1253
7 Bhandal N, Russell R: Intravenous ver- 1000 administered by high single-dose 24 Turawa EB, Musekiwa A, Rohwer AC.
sus oral iron therapy for postpartum infusions or standard medical care for Interventions for preventing postpar-
anaemia. Br J Obstet Gynaecol 2006; the treatment of fatigue in women tum constipation. Turawa EB.
113:1248–1252 after postpartum haemorrhage: study Cochrane Database Syst Rev 2015; (9):
8 Van Wyck DB, Martens MG, Seid MH, protocol for a randomised controlled CD011625
et al.: Intravenous ferric carboxymal- trial. Trials 2015; 16:5 25 Koury MJ, Ponka P: New insights into
tose compared with oral iron in the 17 National Board of Health. Healthy erythropoiesis: the roles of folate, vita-
treatment of postpartum anemia: a habits – before, during and after min B 12, and Iron. 1. Annu Rev Nutr
randomized controlled trial. Obstet pregnancy [Internet]. Copenhagen: 2004; 24:105–131
Gynecol 2007; 110:267–278 Sundhedsstyrelsen; 2010. Available 26 Fishbane S, Ungureanu VD, Maesaka
9 Seid MH, Derman RJ, Baker JB, et al.: from Sundhedsstyrelsen: http://sund JK, et al.: The safety of intravenous
Ferric carboxymaltose injection in the hedsstyrelsen.dk/~/media/CB2593468D iron dextran in hemodialysis patients.
treatment of postpartum iron defi- F446C6B0A62868F72DEDAE.ashx Am J Kidney Dis 1996; 28:529–534
ciency anemia: a randomized con- [Last accessed 5 November 2015] 27 Auerbach M, Ballard H, Glaspy J: Clin-
trolled clinical trial. Am J Obstet 18 Smets EM, Garssen B, Bonke B, et al.: ical update: intravenous iron for anae-
Gynecol 2008; 199:435.e1-7 The Multidimensional Fatigue Inven- mia. Lancet 2007; 369:1502–1504
10 Breymann C, Gliga F, Bejenariu C, tory (MFI) psychometric qualities of an 28 Rampton D, Folkersen J, Fishbane S,
et al.: Comparative efficacy and safety instrument to assess fatigue. J Psycho- et al.: Hypersensitivity reactions to
of intravenous ferric carboxymaltose som Res 1995; 39:315–325 intravenous iron: guidance for risk
in the treatment of postpartum iron 19 Cox JL, Holden JM, Sagovsky R: minimization and management. Hae-
deficiency anemia. Int J Gynaecol Detection of postnatal depression. matologica 2014; 99:1671–1676
Obstet 2008; 101:67–73 Development of the 10-item Edinburgh 29 DeLoughery TG, Auerbach M: Is low-
11 Verma S, Inamdar SA, Malhotra N: Postnatal Depression Scale. Br J Psy- molecular weight iron dextran really
Intravenous iron therapy versus oral chiatry 1987; 150:782–786 the most risky iron? - Unconvincing
iron in postpartum patients in rural 20 Jansen AJ, Essink-Bot M, Beckers EA, data from an unconvincing study. Am
area. J SAFOG 2011; 3:67–70 et al.: Quality of life measurement in J Hematol 2016; 91:451–452
12 Froessler B, Cocchiaro C, Saadat-Gilani patients with transfusion-dependent 30 Van Wyck DB, Mangione A, Morrison
K, et al.: Intravenous iron sucrose ver- myelodysplastic syndromes. Br J Hae- J, et al.: Large-dose intravenous ferric
sus oral iron ferrous sulfate for ante- matol 2003; 121:270–274 carboxymaltose injection for iron defi-
natal and postpartum iron deficiency 21 Jansen AJ, Essink-Bot M-L, Duvekot ciency anemia in heavy uterine bleed-
anemia: a randomised trial. J Matern JJ, et al.: Psychometric evaluation of ing: a randomized, controlled trial.
Fetal Neonatal Med 2013; 26:654–659 health-related quality of life measures Transfusion 2009; 49:2719–2728
13 Jain G, Palaria U, Jha SK: Intravenous in women after different types of 31 Food and Drug Administration Center
iron in postpartum anemia. J Obstet delivery. J Psychosom Res 2007; for Drug Evaluation and Research.
Gynecol India 2013; 63:45–48 63:275–281 Summary minutes of the drug safety
14 Mumtaz A, Farooq F: Comparison for 22 Gibson J, McKenzie-McHarg K, and risk management advisory com-
effects of intravenous versus oral iron Shakespeare J, et al.: A systematic mittee [Internet]. Maryland: Food and
therapy for postpartum anemia. Pak J review of studies validating the Edin- Drug Administration; 2008. Available
Med Health Sci 2011; 5:116–120 burgh Postnatal Depression Scale in from Fodd and Drug Administration:
15 Markova V, Norgaard A, Jørgensen KJ, antepartum and postpartum women. http://www.fda.gov/ohrms/dockets/AC/
et al. Treatment for women with post- Acta Psychiatri Scand 2009; 119:350– 08/minutes/2008-4337 m1-Final.pdf
partum iron deficiency anaemia. 364 [Last accessed 1 October 2016]

© 2017 The Authors.


Vox Sanguinis published by John Wiley & Sons Ltd on behalf of International Society of Blood Transfusion
Vox Sanguinis (2017)

You might also like