ArticlePDF Available

The Benefits and Risks of Iron Supplementation in Pregnancy and Childhood

Authors:

Abstract

Iron deficiency is the most common micronutrient deficiency in the world and disproportionately affects pregnant women and young children. Iron deficiency has negative effects on pregnancy outcomes in women and on immune function and neurodevelopment in children. Iron supplementation programs have been successful in reducing this health burden. However, iron supplementation of iron-sufficient individuals is likely not necessary and may carry health risks for iron-sufficient and potentially some iron-deficient populations. This review considers the physiology of iron as a nutrient and how this physiology informs decision-making about weighing the benefits and risks of iron supplementation in iron-deficient, iron-sufficient, and iron-overloaded pregnant women and children. Expected final online publication date for the Annual Review of Nutrition Volume 39 is August 21, 2019. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
The Benefits and Risks of Iron Supplementation in Pregnancy
and Childhood
Michael K. Georgieff1, Nancy F. Krebs2, Sarah E. Cusick1
1Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, Minnesota
55454, USA;
2Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus,
Aurora, Colorado 80045, USA;
Abstract
Iron deficiency is the most common micronutrient deficiency in the world and disproportionately
affects pregnant women and young children. Iron deficiency has negative effects on pregnancy
outcomes in women and on immune function and neurodevelopment in children. Iron
supplementation programs have been successful in reducing this health burden. However, iron
supplementation of iron-sufficient individuals is likely not necessary and may carry health risks
for iron-sufficient and potentially some iron-deficient populations. This review considers the
physiology of iron as a nutrient and how this physiology informs decision-making about weighing
the benefits and risks of iron supplementation in iron-deficient, iron-sufficient, and iron-
overloaded pregnant women and children.
Keywords
iron; pregnancy; infancy; supplementation; neurodevelopment; malaria
IRON AS A NUTRIENT
Iron is the most abundant element, comprising 35% of the earth’s physical mass. While most
of iron’s interactions with the environment are inorganic, it nevertheless plays a critical role
in organic biological processes in virtually all living organisms. Iron accretion by cells and
its utilization in biological processes within cells are remarkably conserved across the
taxonomic hierarchy. This speaks to its fundamental importance in sustaining the metabolic
processes essential to the survival and function of living organisms.
Iron is classified as a micronutrient. A primary nutritional role of iron is to support
erythropoiesis. Iron is prioritized to red blood cells over all other organ systems, including
the brain, in the developing fetus and young child to support hemoglobin synthesis (46, 48,
georg001@umn.edu.
DISCLOSURE STATEMENT
The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the
objectivity of this review.
HHS Public Access
Author manuscript
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Published in final edited form as:
Annu Rev Nutr
. 2019 August 21; 39: 121–146. doi:10.1146/annurev-nutr-082018-124213.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
104, 161). Every cell and organ system in the body requires iron for proper development and
subsequent metabolic function. Its physiologic role in iron cluster proteins and hemoproteins
is to facilitate enzymatic processes that are essential to cellular metabolic function, including
those essential for oxygen delivery and cellular adenosine triphosphate generation. While
iron-deficiency anemia is associated with clinical symptoms, tissue-level iron deficiency and
not necessarily anemia is responsible for organ dysfunction in the iron-deficient state.
Symptoms of poor organ performance are present prior to the onset of anemia and persist
after the anemia resolves following iron treatment (8).
A precise estimate of the burden of iron deficiency is difficult to obtain because of the
various definitions of iron deficiency used worldwide. Approximately 2 billion of the earth’s
7.2 billion people are anemic, with iron deficiency as the root cause in an estimated 25% to
50% (104, 134). However, the prevalence of nonanemic iron deficiency, which is associated
with neurodevelopmental consequences, is estimated to be threefold greater than the
prevalence of iron-deficiency anemia. Conservative projections indicate that between 2 and 4
billion people, or approximately one-quarter to one-half of the world’s population, may be
iron deficient (104).
Iron-deficiency effects are particularly profound on cells with the highest metabolic rates,
presumably because iron deficiency compromises mitochondrial and cellular energetics (7).
Thus, the consequences of iron deficiency are more profound during development, when the
oxygen consumption rates of cells are highest, driven by the energy demands of growth and
differentiation. On a per kilogram of weight basis, the total-body oxygen consumption of a
neonate is three to four times greater than that of an adult. Moreover, the neonatal brain
alone utilizes 60% of that oxygen consumption, compared with 20% in the adult brain (72).
Given this large iron-dependent energy demand, the prevalence of iron deficiency is greatest
in pregnant women and young children.
The main reason to maintain iron sufficiency in these vulnerable populations is to optimize
organ development and function, including immune function and brain development (30,
46). Within the brain, iron deficiency negatively affects iron-dependent myelination,
monoamine metabolism, and cellular energetics (80, 87). Early-life iron deficiency patterns
gene expression in the brain across the lifetime via histone-mediated stable epigenetic
modifications (137, 138). These findings may explain why certain neurobehavioral effects of
early-life iron deficiency appear to be permanent (80, 86). Whether early-life iron deficiency
affects the regulation of other organ systems, including the production of red blood cells,
across the life span is being investigated (51).
Like most other nutrients, iron exhibits a U-shaped risk curve (Figure 1). The risks of iron
deficiency have been well described (80). However, as a divalent metal, iron has the
potential to react in inorganic reactions that can damage organic components of the cell.
Specifically, nonprotein-bound iron (NPBI) can react with oxygen to form reactive oxygen
species, which if unquenched, can damage lipid membranes and organelles. These are not
typically thought of as consequences of nutritional iron intake, but rather as pathologic
conditions in which iron is suddenly released in large quantities from damaged or dead cells.
The exquisitely tight, coordinated regulation of iron uptake, storage, and trafficking,
Georgieff et al. Page 2
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
combined with the effectiveness of iron-transport proteins (e.g., transferrin) in hiding iron
within their structures, generally prevent NPBI from being present in circulation following
conventional dietary intake of iron-containing foods (44).
Nevertheless, conditions may be present in which regulation breaks down (e.g., genetic
conditions, such as hereditary hemochromatosis) or is overwhelmed by excessively rapid
iron delivery, following which NPBI is generated and is potentially able to react with
oxygen. Excessive iron delivery could occur through the lysis of red blood cells after
transfusion or as a consequence of rapid infusion of intravenous iron. Concerns have been
raised as to whether enteral iron supplementation can result in NPBI, particularly under
conditions of high oral dosages or low binding protein concentrations. The former may
occur with aggressive enteral iron supplementation. Brittenham et al. (15) recently
demonstrated that a supplemental dose of iron sulfate on an empty stomach in healthy, iron-
replete women resulted in the appearance of NPBI but not reactive oxygen species.
Premature infants, who typically have low serum protein concentrations, including low
transferrin, and immature antioxidant systems would theoretically be at increased risk for the
presence of NPBI. Yet enteral iron doses of up to 12 mg/kg body weight in preterm infants
have not been shown to generate NPBI or evidence of reactive oxygen species (14).
Beyond the possibility of generating reactive oxygen species, competition for nutritional
iron by other highly metabolic cells, such as pathogenic organisms, must be considered as a
potential risk of nutritional iron supplementation. Enteral nonheme iron absorption is tightly
regulated by hepcidin, and the absorption rate rarely exceeds 30% (44). Thus, a large portion
of enteral iron, especially in fortified foods, is unabsorbed and is potentially available as an
essential nutrient for bacteria residing in the colon. Bacteria species have variable iron
needs, and some of the most siderophilic species include
Escherichia coli
and
Salmonella
,
both potential pathogens (99). Bifidobacteria, in contrast, have low iron requirements (70).
In addition, pathogenic organisms dwelling within the body (e.g., the malaria protozoa) also
proliferate more aggressively in iron-rich environments compared with iron-poor
environments (49). Taken together, there are good theoretical reasons to propose that while
iron shows the same general U-shaped risk curve as all nutrients, the stakes are greater from
both under- and overnutrition than for most nutrients (Figure 1).
It is against this background that the following sections discuss the relative benefits and risks
of iron supplementation in the two populations at greatest risk for the consequences of iron
deficiency: pregnant women and young children. Consideration is given for both populations
as to how the benefit–risk balance of enteral iron supplementation is affected by iron status.
BENEFITS AND RISKS OF IRON SUPPLEMENTATION DURING
PREGNANCY
Physiologic Considerations
Pregnancy poses a large risk of negative iron balance to a woman (39). Compared with the
nonpregnant state, iron demands are greatly amplified for two reasons. First, the
fetoplacental unit requires a large amount of iron for its own growth and development during
Georgieff et al. Page 3
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
gestation. One gram of iron needs to be accreted by the mother during pregnancy—of which
360 mg is transferred from the mother to the fetus, particularly during the third trimester
when growth is most rapid—in order to maintain a content of 75 mg of iron per kg body
weight of the fetus (39, 169). The pregnant woman expands her own plasma and blood
volumes to maintain proper circulation and oxygen delivery to her own organs as well as to
the placenta. The blood volume expansion consumes 450 mg of the 1 g of additional iron
required during pregnancy (39). Decreasing hepcidin concentrations during pregnancy
indicate the pregnant woman’s need to absorb more iron for both her own hemoglobin
synthesis as well as for transport across the placenta to the highly metabolic and growing
fetus (5, 142). Iron deficiency is generally acknowledged as a greater risk than iron overload
during human pregnancy (39).
The goals of maintaining iron sufficiency during pregnancy are to reduce maternal
morbidity, promote fetal health, and to set up the newborn with adequate nutrient stores for
early postnatal life. Increasing evidence supports the concept that postnatal iron status at 9
months of age depends on proper fetal iron loading during pregnancy (164). The risk of
postnatal iron deficiency in infants is reduced when neonatal iron stores are normal
following gestation, delayed cord clamping is practiced, and postnatal growth rate is not
excessive (30). It is also likely that proper loading of the newborn via the maternal–fetal
route reduces the need for excessive early iron supplementation of the infant postnatally in
certain iron-sufficient populations.
Pregnancies in Iron-Deficient Populations
There is little debate that iron-deficient women have an increased risk of adverse pregnancy
outcomes, that is, those that affect the woman, her fetus, or, consequently, her offspring.
Most studies utilize hemoglobin as the biomarker for iron status because of the ubiquitous
availability of this measurement and because iron deficiency is the most common cause of
anemia in most populations. However, anemia and iron deficiency are not synonymous,
which makes interpreting the outcomes of these studies problematic. Anemia at various time
points in pregnancy is associated with an increased risk of preterm birth (18, 52, 61, 91, 107,
113, 159, 163, 165), birth weight
<
2,500 grams (56, 91, 107, 165), and low weight for
gestational age (28, 52, 61, 107, 144) (all summarized in Reference 30). In most studies,
supplementation of anemic women with iron during pregnancy reduces the rate of iron-
deficiency anemia and nonanemic iron deficiency at term, and in some studies, it reduces the
risk of adverse outcomes, suggesting that supplementation in this population is beneficial
(101, 102) (Table 1).
A minority of clinical studies that assessed outcomes as a function of iron status used iron-
specific biomarkers as opposed to or in conjunction with hemoglobin concentrations (30,
87). Iron-specific markers can be problematic for routine screening for analytic and
interpretative reasons. The limited availability of the analytic equipment needed to measure
specific iron parameters—especially serum ferritin, percent total iron-binding capacity
saturation, soluble transferrin receptor, or hepcidin—is a major hurdle, particularly in low-
resource countries. An ideal biomarker would index the risks of negative iron balance before
physiologic consequences are present (24). Serum ferritin, which typically indexes iron
Georgieff et al. Page 4
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
stores, could theoretically serve this purpose since there are no known consequences of low
iron stores, per se, as long as adequate iron is available to support hematopoiesis, tissue-level
iron proteins (e.g., cytochromes), and iron transport to the fetus. However, ferritin acts as an
acute-phase reactant to infection and inflammation, which undermines its effectiveness as a
screening tool (128). Nevertheless, a meta-analysis of pregnancy outcomes as a function of
iron stores demonstrated that low iron stores, particularly during the first trimester, are
associated with a greater risk of low birth weight, prematurity, and small size for dates (101,
102).
Clinical studies have also assessed maternal iron status exclusively as a function of dietary
iron intake during pregnancy (30). This approach has potential drawbacks, including the
inherent variability of dietary recall and, more importantly, the question of whether dietary
intake is tightly linked to actual iron accretion. This linkage can be tenuous because multiple
inflammatory events during pregnancy could result in relatively less absorption of dietary
iron due to activation of hepcidin by proinflammatory cytokines, including interleukin-6
(149). Furthermore, dietary iron intake gives no information about the distribution of iron
between mother and fetus. Nevertheless, a meta-analysis of studies of maternal iron intake in
iron-deficient populations shows that iron supplementation of iron-deficient populations is
beneficial (101, 102). Neurobehavioral pathologies in offspring related to low maternal iron
intake during critical periods of pregnancy include increased risks of schizophrenia (64) and
autism (115).
Overall, clinical studies support iron supplementation of pregnant women with iron
deficiency defined by any of the three biomarker approaches (i.e., hemoglobin, serum
ferritin, or dietary intake). Little discordance exists among the three biomarkers, except in
the case of active inflammatory processes. Table 1 presents the various potential
interpretations and misinterpretations of utilizing hemoglobin or ferritin as the primary
biomarker to assess iron status.
The role of inflammation in confounding iron status assessments is important. Inflammation
activates hepcidin and thereby countermands the normal increase in iron accretion mediated
by low hepcidin concentrations during pregnancy (5, 39, 142). Hepcidin increases iron in the
storage pool, as evidenced by high serum ferritin concentrations, while shortchanging iron
availability for hemoglobin synthesis by reducing intestinal iron absorption. Chronic
inflammation results in reduced total-body iron during pregnancy and less iron availability
for the fetus, yet the condition may be interpreted as iron overload or iron sufficiency if
ferritin is the only biomarker used by the clinician to assess iron status (Table 1).
Although chronic low-grade inflammation may be relatively common in austere settings (4),
the most common inflammatory condition worldwide during pregnancy is malaria.
Worldwide, approximately 35 million pregnant women are at risk of
Plasmodium falciparum
malaria each year (154). The vast majority of populations at risk for malaria live in regions
where iron deficiency is endemic. Iron supplementation in areas where both iron deficiency
and malaria are endemic must be viewed within in the context of the 2006 landmark study
on Pemba Island, Tanzania, that found that universal, daily supplementation with iron and
folic acid increased the risk of hospitalization and death in young children (111). Subsequent
Georgieff et al. Page 5
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
cross-sectional studies seemed to support this association in pregnant women, finding a
lower prevalence of placental malaria among women with iron-deficiency anemia (66, 118).
In vitro studies have provided apparent mechanistic support of these findings, demonstrating
that red blood cells taken from both anemic children and anemic pregnant women support a
lower rate of
Plasmodium
growth than nonanemic red blood cells. Iron supplementation
without treatment of malaria leads to increased parasite growth in red blood cells from both
children and women (22, 49).
However, two recent randomized placebo-controlled trials have not supported a harmful link
between healthy iron status and the risk of malaria in pregnant women. The trials
demonstrate the importance of treating the malaria first and then addressing iron status (37,
94). In the first study, 1,500 HIV-uninfected pregnant Tanzanian women with serum ferritin
concentrations
>
12 μg/L and hemoglobin concentrations
>
85 g/L were enrolled before 27
weeks’ gestation and were randomized to 60 mg of daily iron as ferrous sulfate or to placebo
(37). All women received monthly prenatal health checks that included malaria screening,
intermittent presumptive treatment for malaria, and antimalarial treatment if needed. Iron
supplementation was not associated with an increased risk of placental malaria or other
adverse events, and while iron did not increase birth weight, hemoglobin concentration and
iron status as measured by serum ferritin concentration were greater among women in the
iron-supplemented group. Although this study was conducted in an urban setting with a
relatively low risk of malaria, a subsequent study in a malaria-endemic area of rural Kenya
had similar findings (94). In that randomized placebo-controlled trial, daily supplements of
60 mg iron as ferrous fumarate given to pregnant women between the ages of 15 and 46
years, with no other inclusion criteria, did not increase the risk of maternal malaria and were
associated with greater birth weight, lower risk of premature birth, longer length of
gestation, and higher maternal and infant iron stores 1 month after birth when compared
with placebo.
After the Pemba study, all iron supplementation trials in malaria-endemic areas have
included a malaria control component for ethical reasons. These findings support the current
World Health Organization (WHO) recommendation for universal daily supplementation
with 30 to 60 mg elemental iron during pregnancy in regions where the prevalence of
anemia is 20% or higher, with a stipulation in malaria-endemic areas that supplementation
should be given in conjunction with “adequate measures to prevent, diagnose and treat
malaria” (157).
Pregnancies in Iron-Sufficient Populations
Further controversy with respect to the accurate diagnosis of iron status and subsequent iron
supplementation surrounds the routine iron supplementation of apparently iron-sufficient
(i.e., non-iron-deficient) women during pregnancy. The US Preventive Services Task Force
stated that there was insufficient evidence to advocate routine iron supplementation during
pregnancy (140). A similar statement from the European Food Safety Authority concluded
that iron supplementation during pregnancy should be reserved for those at risk for or with
documented iron deficiency (34). The controversy stems from the difficulty in demonstrating
any added benefit versus any potential risks of iron supplementation.
Georgieff et al. Page 6
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
The majority of this literature defines iron status by measuring maternal hemoglobin
concentration (30). However, precisely defining iron status in the context of what is termed
the physiologic anemia of pregnancy is problematic. The physiologic anemia of pregnancy
occurs because of a disproportionately greater expansion of the plasma volume (+50%) than
of the red cell mass (+25%), leading to a dilutional reduction in hemoglobin concentration
(146). Maternal hemoglobin concentrations between 95 and 110 g/dL have been associated
with the best pregnancy outcomes and, thus, would be considered normal (17). Hemoglobin
concentrations higher than this range have been associated with higher rates of
preeclampsia, prematurity, and fetal growth restriction (18, 52, 113, 116, 136, 166). A
smaller number of trials have assessed the effect of iron supplementation on women with
high hemoglobin concentrations (i.e.,
>
132 g/L) and found an increased rate of maternal
preeclampsia and fetal growth restriction (166).
A nonanemic hemoglobin concentration would typically be considered a biomarker of iron
sufficiency in nonpregnant women, whereas excessively high hemoglobin would be
consistent with total-body iron overload, given that iron is predominantly found in red cells.
Alternatively, elevated hemoglobin concentrations during pregnancy may not indicate iron
overload but instead reflect low plasma volume expansion, that is, cases in which the 2:1
ratio of plasma volume to red cell mass expansion is not achieved (146). In that
circumstance, serum ferritin may be normal or even low (Table 1). Thus, an elevated
hemoglobin concentration may be misinterpreted as iron overload. Studies that associate
high hemoglobin concentrations during pregnancy with poorer outcomes should be
interpreted cautiously as to whether the mechanistic causes of the poorer outcomes are a
fundamental gestational pathology that leads to low plasma volume expansion, true iron
overload, or both (162). The possibility that iron plays a primary role in plasma volume
dysregulation must be considered because such information would have a direct impact on
the decision to offer iron supplementation to women with normal or high hemoglobin
concentrations (162). The lack of adequate studies hinders the ability to provide guidelines
regarding universal iron supplementation in nonanemic pregnant women (146).
Future studies could avoid dividing pregnant women into the two traditional hemoglobin
categories, anemic or nonanemic, and instead consider a three-group model: anemic, normal,
and polycythemic. Data support this approach since a U-shaped risk curve of pregnancy
complications as a function of hemoglobin concentration has been described (146).
Moreover, women with normal hemoglobin concentrations may have preanemic iron
deficiency. Women with high hemoglobin concentrations may be iron-sufficient, but not iron
overloaded, if the high hemoglobin concentration is due solely to the failure of plasma
volume expansion (Table 1).
Assessing pregnancy outcomes as a function of iron-specific biomarkers could potentially
provide more direct insight than measuring hemoglobin alone. In turn, these markers could
be used to identify candidates for supplementation during pregnancy. Among these markers,
serum ferritin has been most often utilized in outcome studies. WHO recently assessed its
usefulness as a screening tool (156). Clinical interpretation of ferritin concentrations relies
on the understanding that if iron stores are replete, sufficient iron is present to support iron-
dependent cellular processes at the tissue level. Serum ferritin is an excellent specific metric
Georgieff et al. Page 7
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
for low-iron states because no condition other than iron deficiency results in low serum
ferritin concentrations. Interpreting high serum ferritin concentrations is more problematic
with respect to understanding serum ferritin’s relationship to tissue iron status. High ferritin
concentrations could indicate iron overload or, alternatively, a shift of iron into
reticuloendothelial cell storage as part of a response to inflammation (128). Mathematical
correction of ferritin concentrations for the degree of inflammation as indexed by an
inflammatory biomarker has been proposed, but has not been widely implemented (128).
The majority of published studies on pregnancy outcomes as a function of maternal iron
status utilized serum ferritin as the biomarker (30). They reported that higher ferritin
concentrations early in pregnancy were associated with more positive pregnancy outcomes,
whereas higher ferritin concentrations in the third trimester were associated with poorer
outcomes, including premature delivery and low birth weight (50, 73, 116, 131). Interpreting
these studies is difficult because it is unclear whether the high ferritin concentrations during
the third trimester indexed increased total-body iron or a shift of iron into the storage pool
due to inflammation. There is a great need for a sensitive and specific biomarker that indexes
tissue iron status and is not influenced by inflammation.
Assessing iron status by quantifying iron intake has yielded mixed results with respect to
pregnancy and offspring outcomes. On one hand, iron intake in non-iron-deficient mothers
early in pregnancy appears to protect against autism in the offspring (115), and iron intake
during the third trimester induces a more mature gray matter pattern on diffusion tensor
imaging in term infants (92). Conversely, iron supplementation in women with high
hemoglobin concentrations (i.e.,
>
132 g/L) during the second trimester leads to even higher
hemoglobin concentrations in the mother, but a greater risk of fetal growth restriction, most
likely due to maternal hypertension (166). Iron supplementation in pregnant women has also
been linked in observational studies to a greater risk of gestational diabetes mellitus (162).
Conclusions Regarding Iron Supplementation in Pregnant Women
Iron sufficiency during pregnancy results in better pregnancy outcomes for the mother and
the child. The benefits of iron supplementation outweigh the risks in women about to
become pregnant and in pregnant women with evidence of iron deficiency. Women living in
areas where iron deficiency is prevalent also benefit from routine iron supplementation
during pregnancy, even in malaria-endemic areas. In these areas, iron should be given along
with implementing malaria-control efforts, for example, insecticide-treated nets, and with
malaria diagnosis and treatment services readily available.
Typically, there is little additional benefit to supplementing a nutrient that is already replete
in an individual. This approach may also apply to iron supplementation of iron-sufficient or
iron-overloaded pregnant women, for whom little additional benefit would be expected and
potential risks exist. However, the inability to reliably distinguish total-body iron status from
three iron-replete states of hemoglobin in nonanemic women—nonanemic iron deficiency,
optimal iron status, and iron overload—presents a major problem in terms of benefit–risk
analysis since the effects of iron supplementation on these three states likely differ. A more
complete assessment of iron status, made by measuring both hemoglobin and ferritin
concentrations simultaneously, would resolve certain, but not all, ambiguities. The addition
Georgieff et al. Page 8
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
of hepcidin concentration to the arma-mentarium of iron screening tools would greatly
enhance the clarification of who would benefit from enteral iron supplementation. Because
hepcidin is upregulated by high iron status and by inflammation, elevated concentrations in
iron-sufficient, iron-overloaded, and inflamed individuals would reduce or abolish
absorption. In those cases, enteral iron supplementation would be pointless (i.e.,
nonabsorbed) or risky (i.e., nonabsorbed plus adversely altering the microbiome).
Conversely, low hepcidin concentrations would indicate individuals for whom the benefits of
iron supplementation would likely outweigh risks.
BENEFITS AND RISKS OF IRON SUPPLEMENTATION IN EARLY
CHILDHOOD
Physiologic Considerations
Early childhood, especially the first 3 years, is a period of rapid growth and development for
all organ systems, particularly the brain. Newborns have higher hemoglobin concentrations
(79) and iron stores, as reflected in their serum ferritin concentrations (121), than older
infants. Both decline with age (54, 167), and the iron that is liberated is utilized for
expansion of the red cell mass that occurs with the physical growth of the infant and for
biochemical reactions to catalyze tissue development and function.
Evidence from humans (47, 103), monkeys (106), sheep (161), rats, and mice (150)
demonstrates that the brain’s iron status is compromised before the iron status of the red
cells. This prioritization explains why anemia is the end-stage of iron deficiency and why
monitoring hemoglobin concentration is a poor (and late) measure of iron status in children.
Studies in humans (83, 86) and preclinical models (41, 114, 139) demonstrate that brain iron
deficiency early in life confers a risk for poorer brain function in adulthood. The clinical
consequences of early-life iron deficiency include poorer school achievement, lower job
potential, and increased risks of psychopathology (86). Recent preclinical studies suggest
that the failure of brain systems to be properly constructed during this critical period (41) is
responsible for behavioral abnormalities in adulthood (67, 138).
The brain systems that are rapidly developing prior to birth and during the first year after
birth include the monoaminergic neurotransmitter systems (9, 160) and the hippocampus
(41), and the process of myelination (25). Early-life iron deficiency results in abnormal
neuronal structure (16, 41) and function (105), metabolism (106), gene expression (138), and
neurotransmitter concentrations (139) in adulthood. The areas of the brain that have been
identified as affected in preclinical models are concordant with the abnormal behaviors
documented in humans (80, 87), indicating a high biological plausibility for the long-lasting
effects of early-life iron deficiency on the human brain (87).
Whereas brain and behavior are major outcome variables in many studies of childhood iron
deficiency, it is clear that the function of other systems, especially innate immunity, also
depends on iron sufficiency (30). Thus, preventing iron deficiency is important because of its
long-term cost to society, and it is a far better strategy than treating iron-deficiency anemia
once it is present.
Georgieff et al. Page 9
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
The appropriate-for-gestational-age term neonate who received the benefits of delayed cord
clamping has enough iron in red cells and storage pools to sustain tissue iron sufficiency for
at least 4 months and, perhaps, for as long as 6 months, even on a diet with as low an iron
content as human breast milk (30, 77). This sustainability assumes that the infant’s postnatal
growth rate (and hence expansion of the red cell volume) is appropriate as defined by
WHO’s growth curves; rapid infant weight gain is associated with earlier depletion of infant
stores.
The exceptions to this trifecta of term infants with normal iron stores, delayed cord
clamping, and reasonable postnatal growth velocity are common, not just in low-resource
countries but also in high-resource ones. Reduced fetal iron accretion can occur as a function
of maternal iron deficiency, defined as a hemoglobin concentration of
<
85 mg/L or a serum
ferritin concentration of
<
13 μg/L (119), or both. Other gestational conditions that reduce
neonatal supply include premature delivery and fetal growth restriction due to maternal
hypertension (19). The former complicates
>
10% of pregnancies and is important because
the majority of fetal iron is accreted during the last trimester (169). The earlier the preterm
delivery, the greater the risk for subsequent iron deficiency (32). The postnatal iron
requirements of the preterm infant are two- to threefold greater than those of the term infant
(32). Approximately 50% of newborns with fetal growth restriction have ferritin
concentrations below the fifth percentile, indicative of compromised iron stores at birth and
greater risk for postnatal iron deficiency at an earlier age (19). Worldwide, the most common
cause of fetal growth restriction is maternal undernutrition (which includes iron deficiency),
whereas in the United States it is maternal hypertension, including preeclampsia. Maternal
smoking during pregnancy and pregestational or gestational diabetes mellitus are significant
risk factors for low fetal iron status (121, 129). These two common gestational conditions
complicate
>
10% of pregnancies.
Given how common these potential risks to fetal and neonatal iron status are, measuring iron
status at birth would be useful to provide information about which babies are already iron
deficient at birth or at an increased risk for earlier postnatal iron deficiency. The current
American Academy of Pediatrics (AAP) policy of screening for iron deficiency via
assessment of hemoglobin concentration at 1 year of age may be ineffective because it fails
to consider that all other organ systems are affected prior to the onset of anemia (6). A
biomarker that indexes the risk to organ systems other than the erythron is urgently needed
to inform decisions about iron supplementation (46), particularly because nonanemic iron
deficiency is threefold more common than iron-deficiency anemia and is also associated
with altered neurobehavioral development (83). Recent changes by WHO to utilize serum
ferritin as opposed to or in addition to hemoglobin and by the AAP to add reticulocyte
hemoglobin content are steps in the right direction. However, all of the commonly used iron-
specific biomarkers (e.g., serum ferritin, soluble transferrin receptor, percentage total iron-
binding capacity saturation) are influenced by inflammation. Unlike the case in adults (44),
the regulation of hepcidin in very young infants is not well understood or described.
The following sections review the benefits and risks of iron supplementation in populations
with or at high risk for iron deficiency and those that are iron sufficient or at low risk for iron
deficiency. As with the literature on iron supplementation during pregnancy, the lack of
Georgieff et al. Page 10
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
adequate biomarkers of iron status and bioindicators of the physiologic consequences of
alterations in iron status causes significant problems for reaching universal conclusions
regarding supplementation in young infants (87).
Iron-Deficient Populations of Children
Iron-deficient pediatric populations are found predominantly, but not exclusively, in low-
resource settings. Rates upward of 80% have been reported in certain low-resource settings,
while Europe reports rates ranging from 4% to 50% in 6- to 36-month-olds, with a greater
predominance in Eastern Europe compared with Western Europe (141). The current rate of
iron deficiency in 1-to 2-year-olds in the United States is 13.5% (55). The benefits and risks
of iron supplementation in iron-deficient populations can be thought of as striking a balance
between the individual and societal downsides of iron deficiency as it relates to lost
intellectual potential and poorer immune capacity versus any potential risks of infection
posed to individuals by iron supplementation.
There is little disagreement that most term neonates are born with iron stores adequate to last
them for up to 6 months, but that after that time, a source of dietary iron is required to
prevent iron deficiency and anemia (30, 170). Thus, the question of whether to provide iron
supplementation at some point during the first few months of life at a dose that supports
optimal growth and development is not in debate. By 6 months of age, all infants who were
born iron sufficient need a dietary source of iron because fetal stores will have been
exhausted and human breast milk does not contain adequate iron to support the
erythropoietic and tissue-driven iron needs of the infant.
Multiple common gestational conditions place the newborn at risk for lower than normal
iron stores. These conditions (e.g., fetal growth restriction, severe maternal iron-deficiency
anemia) are more common in low-resource countries and set up the newborn for an earlier
onset of postnatal iron deficiency. Exacerbating postnatal factors include rapid postnatal
growth rate, earlier introduction of complementary foods with poor iron content and/or
bioavailability, and frequent illnesses, including infections that limit food intake and activate
hepcidin-driven reductions in iron absorption (23, 170).
Iron supplementation is beneficial if the goal is to alleviate anemia. Clinical studies, both
observational and randomized controlled trials (RCTs), demonstrate that iron and
multimicronutrient supplementation are effective in improving iron status and reducing the
rate of iron-deficiency anemia in iron-deficient populations of infants, whether the
supplementation is through the iron-fortification of formula or milk, or administration of
medicinal iron (108, 112, 125, 145, 152, 168, 170). Hematologic recovery from iron-
deficiency anemia through iron supplementation occurs at all ages, whereas neurobehavioral
recovery is not always complete (86, 87). There is minimal literature on the recovery of
immune health following iron supplementation in iron-deficient populations.
The literature is robust with respect to the effect of iron status on neurodevelopment. Iron
deficiency is associated with neurobehavioral abnormalities while the infant is deficient (3,
83, 109, 110, 120). Some abnormalities continue long after treatment of iron deficiency with
iron supplements (80, 86). A series of reports in the
Lancet
and by the BOND (Biomarkers
Georgieff et al. Page 11
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
of Nutrition for Development) iron work group identified 20 studies that showed poorer
functioning in multiple neurodevelopmental domains in iron-deficient children or children
with iron-deficiency anemia (87, 147, 148). Findings of particular concern include lower
general mental functioning (1, 57, 62, 81, 85, 151), poorer motor performance (1, 62, 85),
and a general loss of capacity relative to an iron-sufficient population across childhood (80).
Whether iron supplementation as a preventative or a treatment strategy is effective against
these neurobehavioral deficits in populations at risk for iron deficiency depends on the age
range of the children, the timing of the intervention, the baseline rate and the degree of iron
deficiency in the population, and the selection of appropriately sensitive and specific
neurodevelopmental outcome indicators. Interpreting the specific role of iron is made more
complicated when iron is provided as part of a generalized nutritional plan (e.g., fortified
formula or milk) or as a multimicronutrient supplement.
Overall, iron supplementation in populations at increased risk for iron-deficiency anemia
improves motor outcomes (12, 74, 84, 127), neurocognitive and language outcomes (84,
127), and social development (12, 74, 84). The effects can be long-lasting. Infants with
hemoglobin concentrations
<
105 g/L who were randomized to iron-supplemented infant
formula (12.7 mg/L) at 6 months of age had better 10-year outcomes than those randomized
to a low-iron formula (2.3 mg/L) (84). A greater impact on neurodevelopment has been
found with earlier supplementation, including starting during the fetal period via maternal
supplementation (20, 21, 26, 93). Later supplementation (e.g., after 1 year of age) was not
beneficial from a neurodevelopmental stand-point, suggesting that the critical period for
providing supplementation to ensure iron sufficiency to protect neurodevelopment is earlier
in life and includes the late fetal period (20, 21, 26, 93).
Iron status can be defined using various biomarkers. These include assessing anemia (with
supporting evidence that the anemia is due to iron deficiency), iron-specific markers, and
iron intake. The bulk of the literature has focused on preventing, detecting, and treating iron-
deficiency anemia. However, nonanemic iron deficiency, as indexed by altered iron-specific
markers, is associated with worse neurobehavioral function, including poorer motor function
(83), social function (83), speed of processing (3), and recognition memory (45, 120). A
recent RCT of mainly nonanemic iron-deficient 9- to 24-month-old infants with 6 mg
iron/kg body weight demonstrated recovery of serum ferritin concentrations, although
neurodevelopmental outcomes were not measured (133). Specific cutoffs for serum ferritin
concentration to detect neurodevelopmental deficits are present for neonates (3, 45, 120) but
not for other ages.
Studies of populations with increased early-life iron requirements—for example, those born
following gestational conditions that resulted in inadequate fetal iron loading, such as
maternal hypertension or premature delivery—indicate a positive role for iron
supplementation in preserving neurobehavioral development. Breastfed Swedish infants with
birth weights between 2,000 and 2,500 g as a result of either late preterm birth or
intrauterine growth restriction were randomized to iron supplementation of 0, 1, or 2 mg/kg
body weight (11). The group randomized to no supplementation had a higher rate of iron
deficiency and iron-deficiency anemia in the first year of life and an increased rate of mild
Georgieff et al. Page 12
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
neurodevelopmental abnormalities at 8 years of age compared with the group that received
either of the two supplemental doses (10, 11). Preterm infants who received iron
supplementation earlier in their neonatal course have higher mental processing composite
scores at 5 years of age (40, 124).
Given the beneficial effects of supplementing iron-deficient infants, consideration must still
be given as to whether there are any contraindications to iron supplementation in this
population. In 1998, in response to the mounting evidence of the potentially permanent
neurobehavioral consequences of iron deficiency in young children, WHO issued a
recommendation for daily, universal iron supplementation of all children aged 6 to 59
months living in areas where the prevalence of anemia was 40% or greater (126). The Pemba
trial (described in the section Pregnancies in Iron-Deficient Populations) first tested this
recommendation, enrolling
>
30,000 children between the ages of 4 and 48 months and
randomizing them to daily supplementation with 12.5 mg iron and folic acid with or without
zinc or to placebo (111).
The iron-containing arms of the Pemba trial were halted by the study’s data and safety
monitoring board after only 18 months because of a 12% increased risk of serious adverse
events (i.e., hospitalizations and deaths) among children who received iron compared with
those who did not. A concurrent study of identical design and equivalent size conducted by
the same researchers in Nepal (135), where there is no malaria, found no harmful effect of
iron supplementation, leaving researchers to conclude that iron supplementation in malaria-
endemic areas may be risky.
A Pemba substudy assessed roughly 3,000 children who had better access to health care and
also measurements of hemoglobin and zinc protoporphyrin (ZPP) and found that the harmful
effect of iron appeared to be confined only to those children who were iron replete (ZPP
<
80 μmol/mol heme) (111). In contrast, iron-deficient children (ZPP ≥ 80 μmol/mol heme)
benefited from iron, experiencing significantly fewer serious adverse events compared with
iron-deficient children who received placebo. The resulting recommendation from WHO
was to halt universal supplementation in malaria-endemic areas and to screen for iron
deficiency before giving the supplement (158). However, this screen-and-treat approach
failed to gain momentum in the field because of the expense of screening in the low-resource
settings where malaria is endemic and the impossibility of conventional interpretation of
iron biomarkers in areas where infection and resulting inflammation are commonplace.
Although three Cochrane Reviews of iron supplementation in children in malaria-endemic
areas conducted after the Pemba study found no harmful effects provided that malaria
control and treatment resources were in place (95–97), iron supplementation programs in
these areas where the public health need is potentially the greatest were drastically cut back
or eliminated altogether.
Red blood cells from anemic children in malaria-endemic areas are more resistant to
invasion by the malaria parasite, and this resistance is reversed with iron supplementation
(22). Because the malaria parasite preferentially invades reticulocytes, the transient
reticulocytosis that follows iron supplementation could explain the increased frequency of
clinical malaria that occurs with the provision of supplemental iron (49). This mechanism
Georgieff et al. Page 13
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
would not explain the increased frequency of nonmalarial infections, including diarrhea and
respiratory infections, reported in children who received supplemental iron, either in the
form of a supplement or micronutrient powder, in areas where malaria or other infections are
endemic (65, 123, 143). The explanation may reside in the gut bacterial microbiome. In two
separate studies of African infants, the regular consumption of iron-fortified biscuits and
iron-fortified food powders shifted the profile of the gut microbiota from one in which more
beneficial bifidobacteria barrier strains presided to one where pathogenic enterobacteria
strains prevailed (132). Recent preclinical models also suggest that pathogenic shifts in the
gut microbiome are associated not only with diarrheal illness but also with an increased risk
of a range of respiratory viruses, including respiratory syncytial virus and pneumococcal
pneumonia (117). Recent work suggests that these effects of iron on the microbiome and the
resulting morbidity may be mitigated by lowering the dose of iron to approximately 5 mg
and giving it in conjunction with a prebiotic, such as galacto-oligosaccharide (98). This
combination reduced anemia in Kenyan children with the same efficacy as a 12.5 mg dose of
iron and was also associated with a significantly lower frequency of respiratory infections
(99).
The absorbability of iron and the corollary question about the amount of residual iron
delivered to the gut microbiotic community is particularly pertinent for children living in
low-resource malaria-endemic areas, as these children typically have a higher baseline level
of inflammation due to poor sanitation, unclean water, and crowded living conditions. The
resultant inflammation and accompanying high hepcidin concentration likely limit the
absorbability of supplemental iron, leaving more iron in the gut lumen to the potential
benefit of pathogenic bacteria.
Delaying the provision of iron until the resolution of inflammation and the nadir of hepcidin
is a key element of a successful and safe pediatric iron program in areas endemic for malaria
and other infections. This concept has recently been demonstrated in a series of studies
examining the timing of iron therapy relative to antimalarial treatment in Ugandan children
with iron deficiency and malaria (26, 27, 65). The standard of care for co-occurring iron
deficiency and malaria is to treat both simultaneously, a practice supported by three
Cochrane Reviews and WHO guidelines (153). Recent research shows that staging the
interventions, by treating children with either severe or uncomplicated malaria first and
delaying the start of iron until 28 days after the onset of antimalarial treatment, leads to a
greater reduction in hepcidin, greater incorporation of iron into red blood cells, and fewer
infectious illnesses compared with children started on iron concurrently with antimalarial
treatment. Whether the initial better incorporation of iron with delayed provision results in
better long-term iron status and neurodevelopment is currently being tested (26, 27, 65).
In the absence of the eradication of malaria, the optimal method for safe maintenance of
healthy iron status in children living in these areas remains unclear. However, providing
smaller amounts of iron at times when hepcidin is lowest appears to be part of an effective
solution. These components are reflected in WHO’s current recommendations, which again
stipulate delivering universal iron supplementation to children between the ages of 6 and 59
months in regions where the prevalence of anemia is 40% or greater, but delivering it
intermittently (i.e., once, twice, or three times per week) where anemia prevalence is 20% to
Georgieff et al. Page 14
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
40% (155). In areas of where malaria is endemic, this supplementation should be given
concomitantly with efforts to control, diagnose, and treat malaria (155).
In addition to any potential risk of iron supplementation on infection rates and microbiome
alterations, investigators have been concerned about iron’s ability to induce reactive oxygen
species. This reaction occurs in the setting of NPBI, which would be most likely to occur in
settings of large iron doses in patients with low iron-binding protein concentrations. The
generation of reactive oxygen species, as indexed by biomarkers of the process, has failed to
be found in multiple studies of iron supplementation in premature infants (121). Premature
infants receiving doses as high as 18 mg of iron/day fail to show evidence of such a response
(14). Nevertheless, it remains prudent to assess this risk as a safety measure in future trials
because high-dose iron will be required to support the erythropoietic benefits of recombinant
erythropoietin therapy in preterm infants (121).
Iron-Sufficient Populations of Children
Although iron deficiency is the most common nutrient deficiency worldwide, the reality is
that the majority of newborn infants are likely to be iron sufficient, with normal iron storage
pools. In this context, a policy of universal dietary iron supplementation of all newborn
infants, both those who are iron sufficient and those at risk for iron deficiency, is
questionable unless there are absolutely no risks to iron supplementation, an assumption that
is not supported by the discussion in the preceding sections.
Any potential negative effects of iron supplementation should be understood within the
context of the iron status of the infant. It is worth considering a distinction between the
labels iron replete and iron overload as they relate to biomarkers of iron status in infants.
True iron overload is rare in neonates and infants. The genetic syndromes responsible for
neonatal iron overload are often fatal. Infants with neonatal polycythemia (e.g., infants of
diabetic mothers), who appear to be iron overloaded, as defined by an elevated hemoglobin
concentration, actually have lower storage and tissue iron contents (47, 103). This
combination of high hemoglobin and low serum ferritin concentrations indicates a shift of
iron—that is, iron has been prioritized for erythropoiesis through chronic fetal hypoxemia—
rather than iron overload. Multiple red cell transfusions could cause iron overload, but
infants who receive such therapy are relatively rare and would not drive universal policies.
Nevertheless, two interesting studies that demonstrate the potential neurodevelopmental risk
of iron overload illustrate the need for better differentiation between iron-replete and iron-
overload states (82, 130).
Tamura et al. (130) demonstrated that the school-age outcomes of newborns who were
divided into quartiles of cord blood serum ferritin concentrations were poorer for the
children in the lowest (i.e., iron-deficient children) and highest quartiles. While the former
finding is not surprising and is consistent with other neurodevelopmental studies of
newborns with low ferritin concentrations (3, 45, 120), the latter finding was unexpected.
The etiology of the high ferritin concentrations in the upper-quartile newborns is not known
and may represent inflammation (with its own risks to neurodevelopment), iron overload (by
unknown mechanisms), or both. Postnatal iron intake was not assessed in this cohort and
thus not factored into the assessment of the school-age outcomes.
Georgieff et al. Page 15
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
The RCT of iron supplementation of infant formula performed in Chile that demonstrated a
benefit of iron-fortified infant formula (12.7 mg/L) on hematologic and neurodevelopmental
outcomes (84) also demonstrated that infants with excessively high hemoglobin
concentrations (i.e.,
>
125 g/L) who were randomized to iron-fortified formula at 6 months
had poorer neurodevelopment at 10 years of age (82). The number of infants in this group
was small (
n
= 26), the etiology of their relative polycythemia/iron overload at 6 months was
unclear, and no mechanistic studies were performed because the finding was a secondary
analysis of neurodevelopmental data collected 10 years later. The negative effect of higher
levels of iron fortification in infant formula on neurodevelopment was not seen in children
with normal hemoglobin concentrations (i.e., between 105 and 125 g/L), and the same
degree of fortification improved neurodevelopment in children with hemoglobin
concentrations of
<
105 g/L (82, 84). These two studies raise the possibility that some infants
who would normally have been assigned to an iron-replete or iron-sufficient category by
ferritin (130) or hemoglobin (82) concentration may instead have had underlying
abnormalities that led to an iron-overload state and that neurologic morbidities that may be
associated with an iron-overload state may be further exacerbated by high-iron diets.
The risks and benefits of dietary iron delivery in presumably iron-sufficient, but not iron-
overloaded, infants and young children have been the focus of recent reviews and were a
major influence on the decision by the US National Institute of Health’s Office of Dietary
Supplements to convene a workshop that took place in 2017 (134) as well as the work of an
expert panel that occurred in 2015 (68).
The iron status of infants as a function of iron in the diet has been extensively studied.
Human breast milk contains approximately 0.5 mg/L of iron. Irrespective of whether this
iron is more absorbable or not, this amount is not sufficient to supply the iron needs of the
growing infant without concomitant mobilization of stored iron (30, 70). Ferritin
concentrations decrease during the first 6 postnatal months in breastfed infants (167).
Breastfed infants not supplemented with iron or iron-containing foods will typically exhaust
iron stores by 6 months of age, and, thus, the rate of iron deficiency in exclusively breastfed
infants at 7 months of age is greater than 30% (71). Iron supplementation slows this decline
(167). Thus, the issue is not whether breastfed infants need iron supplementation, but when
to introduce a source of iron. To answer this question, consideration should be given to
factors that affect the benefit–risk balance. The benefits of maintaining iron sufficiency—
whether through endogenous stores accrued prenatally or through postnatal supplementation
—have been discussed.
The iron content of the most highly fortified formulas is as high as 12–14 mg/L, thus far
exceeding the amount in human breast milk. The fortification of infant formula at 12 mg/L
began more than 50 years ago when the prevalence of iron deficiency in infants in the United
States approached 40% and was a major public health risk. Fortification at this level was
successful in reducing the prevalence of iron deficiency to much lower levels (55). Studies
assessed whether there were increased rates of diarrheal illness, gastrointestinal distress, or
changes to stool bacterial content. Although the stool of infants receiving highly fortified
formula had a greater content of siderophilic bacteria that are potential pathogens (e.g.,
E.
coli
), no increases in diarrheal illness or gastrointestinal symptoms have been found (70,
Georgieff et al. Page 16
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
122). Interestingly, the intake of medicinal iron results in a different microbiome profile than
intake of highly fortified formula (122). It should be noted that this recent study was
performed in Sweden, where a lower infection burden and low rate of infant mortality from
infectious diseases are present (122).
The original assumptions behind delivering fortification at such a high level from birth were
likely misguided because of the lack of knowledge of the regulation of iron absorption by
hepcidin. At that time, breastfeeding rates in the United States were at their lowest point.
Formulas contained only the amount of iron inherent to breast milk (i.e., approximately 1.15
mg/L), and infants were often switched to cow’s milk, another low-iron beverage, at 6
months of age. It was thought that intensely fortified infant formula delivered from birth
would result in infants storing additional iron beyond their immediate needs in order to have
a reserve for what was inevitably a negative iron balance state in the second 6 months of the
first year.
The discovery of hepcidin as a master regulator of gut absorption provided evidence that the
goal of storing extra iron was not likely to be achieved. Intestinal iron absorption ranges
from 4% to 40% based on hepcidin activity (44). Hepcidin is secreted by the liver in
response to sensors of iron status (44). As a negative regulator of intestinal absorption,
hepcidin levels are high when the individual is iron sufficient, thereby limiting iron
absorption, and low when an individual is iron deficient, thereby promoting absorption (44).
It is likely that young infants receiving formula fortified to 12 mg/L were iron replete with
high hepcidin levels, effectively reducing iron absorption to a low percentage and preventing
the intended storage of excess iron. The developmental ontogeny of effective hepcidin
regulation has not yet been completely defined, although evidence suggests that it is intact
by 6 weeks post-term gestational age (38). Hepcidin has been detected in preterm infants,
but the fact that isotope studies of iron absorption in preterm infants consistently
demonstrate an absorption rate of 35% to 40% (35, 53, 89) in spite of a wide range of iron
statuses, (79, 121) suggests that it may not be functional in that population.
A study in Sweden, a country with a low prevalence of iron deficiency, compared providing
formula fortified with 4 mg/L versus 7 mg/L starting at birth and revealed no difference in
hemoglobin concentrations (78). Infants fed the 4 mg/L formula had higher soluble
transferrin receptor than those fed the 7 mg/L formula, indicating a more iron-deficient state.
While there is no physiologic consequence of these changes, it does suggest that 4 mg/L
places the infant in a slightly negative iron balance. This evidence of negative iron balance
provided the rationale for the AAP’s statement advocating for at least 4.5 mg/L of iron in
formula (2).
The timing of iron exposure in iron-sufficient populations is key. The RCT in a high-risk
Chilean population began the iron intervention at 6 months of age and showed that 2.4 mg/L
was not sufficient to support iron status, albeit one could argue that many of the infants were
already total-body iron compromised at the time of randomization (84). In contrast, the
Swedish study began delivering iron-supplemented formula at birth in a population with a
very high likelihood of iron sufficiency at birth (78).
Georgieff et al. Page 17
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Dose consistency is also an important factor. The United States utilizes a one-size-fits-all-
ages approach, with infant formulas designed to be utilized from day 1 to 12 months of age
(59). Europe utilizes formulas that gradually step up the iron concentration as babies age.
This approach makes sense since there appears to be little physiologic rationale to
supplement iron starting at birth in iron-sufficient populations with adequate neonatal
hemoglobin and ferritin concentrations (31, 59).
If there were no risks to iron supplementation, management would be relatively
straightforward, and universal supplementation, medicinally or through the diet, would be
acceptable. However, the potential risks of infant iron supplementation have been identified
(59, 70, 77) and include (
a
) compromise of the status of other divalent metals, especially
zinc; (
b
) an altered gut microbiome, with a potentially increased infection risk; and (
c
)
growth restriction. These risks, especially the effects on the microbiome, have not been
defined by rigorous trials (70).
Iron in doses typically prescribed to nonanemic healthy infants does not appear to have an
adverse effect on either copper or zinc absorption, but it does affect plasma and serum zinc
concentrations (33, 36, 58, 70, 75). This range of iron delivery is well beyond typical iron
supplementation or fortification regimens. Thus, negative effects on other divalent metals are
avoidable with low or moderate iron doses (70).
The intestinal intraluminal iron environment influences the microbiome, which is highly
mutable and is establishing itself in the first months after birth. A low intraluminal iron
concentration favors the growth of the preferred
Lactobacillus
organisms, while high iron
fosters a shift in the microbiome toward potentially pathogenic
Bacteroides
and
E. coli
as
early as 1 week of postnatal age (69, 90, 99). High intraluminal iron most likely occurs in
the setting of high dietary iron intake or inflammation. Both are high hepcidin states in
which iron absorption is low and unabsorbed iron is more likely to be retained
intraluminally. Whether the shift in microbiome character increases the incidence of clinical
infectious diseases or alters the developing innate immune system in iron-sufficient
populations remains an area of intense study, the results of which will likely inform policy in
the future.
A long-standing concern has been whether early-life iron supplementation reduces growth
velocity (77). Studies have had equivocal results from which it is difficult to draw firm
conclusions. A meta-analysis of 35 studies of iron supplementation in children from a wide
age range and in whom initial iron status was usually not known showed an overall effect of
linear growth suppression (100), but results of individual studies vary widely. The lack of
consistent findings may reflect which infants were supplemented (iron-deficient versus iron-
sufficient), their diets (human breast milk versus infant formula), the amount of iron
provided, and the method of supplementation (medicinal iron versus fortified formula).
Medicinal iron supplementation dosed at 1 mg/kg body weight daily in 4- to 6-month-old
term breastfed infants in Sweden and in Honduras yielded different results. The Honduran
infants, who were likely at high risk for negative iron balance or frank iron deficiency,
improved their iron status and did not have growth suppression (29). In contrast, the iron-
Georgieff et al. Page 18
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
sufficient Swedish infants did not increase their iron status, as would have been expected,
but did show linear growth suppression. The Honduran infants showed growth suppression
only if they had hemoglobin concentrations
>
110 g/L, likely indicating an iron-replete state.
Several other studies of iron supplementation starting in late infancy and ranging up to 2
years of age showed negative effects on growth if the infants were iron sufficient (63, 76, 88)
but beneficial effects on iron and hematologic statuses without growth restriction if the
infants were iron deficient at the start of treatment. Interestingly, other studies, including the
RCT from Chile that identified a neurodevelopmental risk with iron supplementation of 6-
month-olds who had high hemoglobin concentrations (82), did not show an adverse effect of
iron on growth (42, 43).
As noted, the studies that demonstrated growth-suppression effects were conducted in older
infants. No growth differences were seen in 1- to 6-month-old infants receiving formula
containing 4 versus 2 mg/L of iron (60). Infants randomized to iron-fortified formula
containing either 7.4 or 12.7 mg/L also showed no difference in growth and no evidence of
growth suppression (13). These sets of studies suggest that older age and a medicinal versus
an infant formula–based iron source may be important factors to consider in iron
supplementation of already iron-sufficient infants.
CONCLUSIONS
In the clearly iron-sufficient newborn, there is little if any benefit to beginning iron
supplementation at birth. However, allowing iron stores to decrease to levels that threaten
availability for developing tissues (without a source of dietary iron) is also potentially risky,
particularly as the infant approaches and exceeds 6 months of age. A gradual introduction of
iron into the diet between months 3 and 5 appears to maximize the benefit–risk ratio, as the
risk of infection declines after 3 months of age as immune competence builds.
Close monitoring of iron status and iron homeostasis is key. Management of all populations
would be easier if total-body iron status were estimated at birth via hemoglobin and ferritin
concentrations (47) and at mid-year, depending on the risk factors for poor iron status.
Waiting until 12 months and measuring only the hemoglobin concentration is simply not
defensible policy with what is now known about risks to iron status and the negative effects
of preanemic iron deficiency on the brain (83). In the future, the measurement of hepcidin
will likely be used to guide iron therapy. Infants with low hepcidin levels likely require iron,
while those with high levels are not likely to benefit from iron supplementation and may
well be at risk for adverse effects.
ACKNOWLEDGMENTS
This work was supported by grants from the National Institutes of Health to M.K.G. (R01-HD089989,
HD29421-20, R01-NS099178, R01-HD094809), N.F.K. (2UG1HD076474-06), and S.E.C. (R01 HD092391-01,
R03 HD074262) and from the Bill & Melinda Gates Foundation (OPP1055867).
LITERATURE CITED
1. Akman M, Cebeci D, Okur V, Angin H, Abali O, Akman AC. 2004 The effects of iron deficiency on
infants’ developmental test performance. Acta Paediatr 93:1391–96 [PubMed: 15499963]
Georgieff et al. Page 19
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
2. Am. Acad. Pediatr. 1999 Iron fortification of infant formulas. Pediatrics 104:119–23 [PubMed:
10390274]
3. Amin SB, Orlando M, Eddins A, MacDonald M, Monczynski C, Wang H. 2010 In utero iron status
and auditory neural maturation in premature infants as evaluated by auditory brainstem response. J.
Pediatr 156:377–81 [PubMed: 19939407]
4. Ashorn P, Hallamaa L, Allen LH, Ashorn U, Chandrasiri U, et al. 2018 Co-causation of reduced
newborn size by maternal undernutrition, infections, and inflammation. Matern. Child Nutr
14(3):e12585 [PubMed: 29316198]
5. Bah A, Pasricha SR, Jallow MW, Sise EA, Wegmuller R, et al. 2017 Serum hepcidin concentrations
decline during pregnancy and may identify iron deficiency: analysis of a longitudinal pregnancy
cohort in the Gambia. J. Nutr 147:1131–37 [PubMed: 28424258]
6. Baker RD, Greer FR, Comm. Nutr. Am. Acad. Pediatr 2010 Diagnosis and prevention of iron
deficiency and iron-deficiency anemia in infants and young children (0–3 years of age). 126:1040–
50
7. Bastian TW, von Hohenberg WC, Mickelson DJ, Lanier LM, Georgieff MK. 2016 Iron deficiency
impairs developing hippocampal neuron gene expression, energy metabolism and dendrite
complexity. Dev. Neurosci 38:264–76 [PubMed: 27669335]
8. Beard JL. 2001 Iron biology in immune function, muscle metabolism and neuronal functioning. J.
Nutr 131:568S–79S [PubMed: 11160590]
9. Beard JL, Erikson KM, Jones BC. 2003 Neonatal iron deficiency results in irreversible changes in
dopamine function in rats. J. Nutr 133:1174–79 [PubMed: 12672939]
10. Berglund SK, Chmielewska A, Starnberg J, Westrup B, Hägglöf B, et al. 2018 Effects of iron
supplementation of low-birth-weight infants on cognition and behavior at 7 years: a randomized
controlled trial. Pediatr. Res 83:111–18 [PubMed: 28953856]
11. Berglund SK, Westrup B, Hägglöf B, Hernell O, Domellöf M. 2013 Effects of iron
supplementation of LBW infants on cognition and behavior at 3 years. Pediatrics 131:47–55
[PubMed: 23230066]
12. Black MM, Baqui AH, Zaman K, Ake Persson L, El Arifeen S, et al. 2004 Iron and zinc
supplementation promote motor development and exploratory behavior among Bangladeshi
infants. Am. J. Clin. Nutr 80:903–10 [PubMed: 15447897]
13. Bradley CK, Hillman L, Sherman AR, Leedy D, Cordano A. 1993 Evaluation of two iron-fortified,
milk-based formulas during infancy. Pediatrics 91:908–14 [PubMed: 8474811]
14. Braekke K, Bechensteen AG, Halvorsen BL, Blomhoff R, Haaland K, Staff AC. 2007 Oxidative
stress markers and antioxidant status after oral iron supplementation to very low birth weight
infants. J. Pediatr 151:23–28 [PubMed: 17586185]
15. Brittenham GM, Andersson M, Egli I, Foman JT, Zeder C, et al. 2014 Circulating non-transferrin-
bound iron after oral administration of supplemental and fortification doses of iron to healthy
women: a randomized study. Am. J. Clin. Nutr 100:813–20 [PubMed: 25057155]
16. Brunette KE, Tran PV, Wobken JD, Carlson ES, Georgieff MK. 2010 Gestational and neonatal iron
deficiency alters apical dendrite structure of CA1 pyramidal neurons in adult rat hippocampus.
Dev. Neurosci 32:238–48 [PubMed: 20689287]
17. Cao C, O’Brien KO. 2013 Pregnancy and iron homeostasis: an update. Nutr. Rev 71:35–51
[PubMed: 23282250]
18. Chang SC, O’Brien KO, Nathanson MS, Mancini J, Witter FR. 2003 Hemoglobin concentrations
influence birth outcomes in pregnant African-American adolescents. J. Nutr 133:2348–55
[PubMed: 12840205]
19. Chockalingam UM, Murphy E, Ophoven JC, Weisdorf SA, Georgieff MK. 1987 Cord transferrin
and ferritin levels in newborn infants at risk for prenatal uteroplacental insufficiency and chronic
hypoxia. J. Pediatr 111:283–86 [PubMed: 3612404]
20. Christian P, Morgan ME, Murray-Kolb L, LeClerq SE, Khatry SK, et al. 2011 Preschool iron–folic
acid and zinc supplementation in children exposed to iron–folic acid in utero confers no added
cognitive benefit in early school-age. J. Nutr 141:2042–48 [PubMed: 21956955]
Georgieff et al. Page 20
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
21. Christian P, Murray-Kob LE, Khatry SK, Katz J, Schaefer BA, et al. 2010 Prenatal micronutrient
supplementation and intellectual and motor function in early school-aged children in Nepal. JAMA
304:2716–23 [PubMed: 21177506]
22. Clark MA, Goheen MM, Fulford A, Prentice AM, Elnagheeb MA, et al. 2014 Host iron status and
iron supplementation mediate susceptibility to erythrocytic stage
Plasmodium falciparum
. Nat.
Commun 5:4446 [PubMed: 25059846]
23. Clark KM, Li M, Zhu B, Liang F, Shao J, et al. 2017 Breastfeeding, mixed, or formula feeding at 9
months of age and the prevalence of iron deficiency and iron deficiency anemia in two cohorts in
China. J. Pediatr 181:56–61 [PubMed: 27836288]
24. Combs GF, Trumbo PR, McKinley MC, Milner J, Studenski S, et al. 2013 Biomarkers in nutrition:
new frontiers in research and application. Ann. N. Y. Acad. Sci 1278:1–10 [PubMed: 23488557]
25. Connor JR, Menzies SL. 1996 Relationship of iron to oligodendrocytes and myelination. Glia
17:83–93 [PubMed: 8776576]
26. Cusick SE, Georgieff MK. 2016 The role of nutrition in brain development: the golden opportunity
of the “first 1000 days.” J. Pediatr 175:16–21 [PubMed: 27266965]
27. Cusick SE, Opoka RO, Abrams SA, John CC, Georgieff MK, Mupere E. 2016 Delaying iron
therapy until 28 days after antimalarial treatment is associated with greater iron incorporation and
equivalent hematologic recovery after 56 days in children: a randomized controlled trial. J. Nutr
146:1769–74 [PubMed: 27358418]
28. Delpisheh A, Brabin L, Drummond S, Brabin BJ. 2008 Prenatal smoking exposure and asymmetric
fetal growth restriction. Ann. Hum. Biol 35:573–83 [PubMed: 18932054]
29. Dewey KG, Domellöf M, Cohen RJ, Landa Rivera L, Hernell O, Lönnerdal B. 2002 Iron
supplementation affects growth and morbidity of breast-fed infants: results of a randomized trial in
Sweden and Honduras. J. Nutr 132:3249–55 [PubMed: 12421836]
30. Dewey KG, Oaks BM. 2017 U-shaped curve for risk associated with maternal hemoglobin, iron
status, or iron supplementation. Am. J. Clin. Nutr 106(Suppl.):1694S–702S [PubMed: 29070565]
31. Domellöf M, Cohen RJ, Dewey KG, Hernell O, Landa Rivera L, Lönnerdal B. 2001 Iron
supplementation of breast-fed Honduran and Swedish infants from 4 to 9 months of age. J. Pediatr
138:679–87 [PubMed: 11343043]
32. Domellöf M, Georgieff MK. 2015 Postdischarge iron requirements of the preterm infant. J. Pediatr
167(Suppl.):S31–35 [PubMed: 26364023]
33. Domellöf M, Hernell O, Abrams SA, Chen Z, Lönnerdal B. 2009 Iron supplementation does not
affect copper and zinc absorption in breastfed infants. Am. J. Clin. Nutr 89:185–90 [PubMed:
19056575]
34. EFSA Panel Diet. Prod. Nutr. Allerg. 2015 Scientific opinion on dietary reference values for iron.
EFSA J 13:4254
35. Ehrenkranz RA, Gettner PA, Nelli CM, Sherwonit EA, Williams JE, et al. 1992 Iron absorption and
incorporation into red blood cells by very low birth weight infants: studies with the stable isotope
58Fe. J. Pediatr. Gastroenterol. Nutr 15:270–78 [PubMed: 1432464]
36. Esamai F, Liechty E, Ikemeri J, Westcott J, Kemp J, et al. 2014 Zinc absorption from micronutrient
powder is low but is not affected by iron in Kenyan infants. Nutrients 6:5636–51 [PubMed:
25493942]
37. Etheredge AJ, Premji Z, Gunaratna NS, Abioye AI, Aboud S, et al. 2015 Iron supplementation in
iron-replete and nonanemic pregnant women in Tanzania: a randomized clinical trial. JAMA
Pediatr 169:947–55 [PubMed: 26280534]
38. Finkelstein JL, O’Brien KO, Abrams SA, Zavaleta N. 2013 Infant iron status affects iron
absorption in Peruvian breastfed infants at 2 and 5 months of age. Am. J. Clin. Nutr 98:1475–84
[PubMed: 24088721]
39. Fisher A, Nemeth A. 2017 Iron homeostasis during pregnancy. Am. J. Clin. Nutr
106(Suppl.):1567S–74S [PubMed: 29070542]
40. Franz AR, Mihatsch WA, Sander S, Kron M, Pohlandt F. 2000 Prospective randomized trial of
early versus late enteral iron supplementation in infants with a birth weight of less than 1301
grams. Pediatrics 106:700–6 [PubMed: 11015511]
Georgieff et al. Page 21
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
41. Fretham SJB, Carlson ES, Wobken J, Tran PV, Petryk A, Georgieff MK. 2012 Temporal
manipulation of transferrin-receptor-1-dependent iron uptake identifies a sensitive period in mouse
hippocampal neurodevelopment. Hippocampus 22:1691–1702 [PubMed: 22367974]
42. Friel JK, Aziz K, Andrews WL, Harding SV, Courage ML, Adams RJ. 2003 A double-masked,
randomized control trial of iron supplementation in early infancy in healthy term breast-fed
infants. J. Pediatr 143:582–86 [PubMed: 14615726]
43. Gahagan S, Yu S, Kaciroti N, Castillo M, Lozoff B. 2009 Linear and ponderal growth trajectories
in well-nourished, iron-sufficient infants are unimpaired by iron supplementation. J. Nutr
139:2106–12 [PubMed: 19776186]
44. Ganz T, Nemeth E. 2012 Hepcidin and iron homeostasis. Biochim. Biophys. Acta 1823:1434–43
[PubMed: 22306005]
45. Geng F, Mai X, Zhan J, Xu L, Zhao Z, et al. 2015 Impact of fetal–neonatal iron deficiency on
recognition memory at 2 months of age. J. Pediatr 167:1226–32 [PubMed: 26382625]
46. Georgieff MK. 2017 Iron assessment to protect the developing brain. Am. J. Clin. Nutr 106(Suppl.
6):1588S–93S [PubMed: 29070550]
47. Georgieff MK, Landon MB, Mills MM, Hedlund BE, Faassen AE, et al. 1990 Abnormal iron
distribution in infants of diabetic mothers: spectrum and maternal antecedents. J. Pediatr 117:455–
61 [PubMed: 2391604]
48. Georgieff MK, Schmidt RL, Mills MM, Radmer WJ, Widness JA. 1992 Fetal iron and cytochrome
c status after intrauterine hypoxemia and erythropoietin administration. Am. J. Physiol 262:R485–
91 [PubMed: 1313652]
49. Goheen MM, Bah A, Wegmüller R, Verhoef H, Darboe B, et al. 2017 Host iron status and
erythropoietic response to iron supplementation determines susceptibility to the RBC stage of
falciparum
malaria during pregnancy. Sci. Rep 7:17674 [PubMed: 29247172]
50. Goldenberg RL, Tamura T, DuBard M, Johnston KE, Copper RL, Neggers Y. 1996 Plasma ferritin
and pregnancy outcome. Am. J. Obstet. Gynecol 175:1356–59 [PubMed: 8942514]
51. Golub MS, Hogrefe CE, Malka R, Higgins JM. 2014 Developmental plasticity of red blood cell
homeostasis. Am. J. Hematol 89:459–66 [PubMed: 24415575]
52. Gonzales GF, Steenland K, Tapia V. 2009 Maternal hemoglobin level and fetal outcome at low and
high altitudes. Am. J. Physiol 297:R1477–85
53. Griffin I, Cooke RJ. 2010 Iron retention in preterm infants fed low iron intakes: a metabolic
balance study. Early Hum. Dev 86(Suppl. 1):49–53 [PubMed: 20138446]
54. Gupta PM, Hamner HC, Suchdev PS, Flores-Ayala R, Mei Z. 2017 Iron status of toddlers, non-
pregnant females and pregnant females in the United States. Am. J. Clin. Nutr 106(Suppl.
6):1640S–46S [PubMed: 29070559]
55. Gupta PM, Perrine CG, Mei Z, Scanlon KS. 2016 Iron, anemia and iron deficiency anemia among
young children in the United States. Nutrients 8:330
56. Hämäläinen H, Hakkarainen K, Heinonen S. 2003 Anaemia in the first but not in the second or
third trimester is a risk factor for low birth weight. Clin. Nutr 22:271–75 [PubMed: 12765667]
57. Hasanbegović E, Sabanović S. 2004 [Effects of iron therapy on motor and mental development of
infants and small children suffering from iron deficiency anaemia]. Med. Arh 58:227–29 (In
Bosnian) [PubMed: 15526591]
58. Haschke F, Ziegler EE, Edwards BB, Fomon SJ. 1986 Effect of iron fortification of infant formula
on trace mineral absorption. J. Pediatr. Gastroenterol. Nutr 5:768–73 [PubMed: 3761108]
59. Hernell O, Fewtrell MS, Georgieff MK, Krebs NF, Lönnerdal B. 2015 Summary of current
recommendations on iron provision and monitoring of iron status for breastfed and formula-fed
infants in resource-rich and resource-constrained countries. J. Pediatr 167(Suppl.):S40–47
[PubMed: 26364025]
60. Hernell O, Lönnerdal B. 2002 Iron status of infants fed low-iron formula: no effect of added bovine
lactoferrin or nucleotides. Am. J. Clin. Nutr 76:858–64 [PubMed: 12324301]
61. Hwang HS, Kim YH, Kwon JY, Park YW. 2010 Uterine and umbilical artery Doppler velocimetry
as a predictor for adverse pregnancy outcomes in pregnant women with anemia. J. Perinat. Med
38:467–71 [PubMed: 20443671]
Georgieff et al. Page 22
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
62. Idjradinata P, Pollitt E. 1993 Reversal of developmental delays in iron-deficient anaemic infants
treated with iron. Lancet 341:1–4 [PubMed: 7678046]
63. Idjradinata P, Watkins WE, Pollitt E. 1994 Adverse effect of iron supplementation on weight gain
of iron-replete young children. Lancet 343:1252–54 [PubMed: 7910275]
64. Insel BJ, Schaefer CA, McKeague IW, Susser ES, Brown AS. 2008 Maternal iron deficiency and
the risk of schizophrenia in offspring. Arch. Gen. Psychiatry 65:1136–44 [PubMed: 18838630]
65. Jaramillo EG, Mupere E, Opoka RO, Hodges JS, Lund TC, et al. 2017 Delaying the start of iron
until 28 days after antimalarial treatment is associated with lower incidence of subsequent illness
in children with malaria and iron deficiency. PLOS ONE 12:e0183977 [PubMed: 28854208]
66. Kabyemela ER, Fried M, Kurtis JD, Mutabingwa TK, Duffy PE. 2008 Decreased susceptibility to
Plasmodium falciparum
infection in pregnant women with iron deficiency. J. Infect. Dis 198:163–
66 [PubMed: 18500927]
67. Kennedy BC, Dimova JG, Siddappa AJM, Tran PV, Gewirtz JC, Georgieff MK. 2014 Prenatal
choline supplementation ameliorates the long-term neurobehavioral effects of fetal–neonatal iron
deficiency in rats. J. Nutr 144:1858–65 [PubMed: 25332485]
68. Kleinman RE. 2015 Expert recommendations on iron fortification in infants. J. Pediatr
167(Suppl.):S48–49 [PubMed: 26364026]
69. Kortman GA, Raffatellu M, Swinkels DW, Tjalsma H. 2014 Nutritional iron turned inside out:
intestinal stress from a gut microbial perspective. FEMS Microbiol. Rev 38:1202–1234 [PubMed:
25205464]
70. Krebs NF, Domellöf M, Ziegler E. 2015 Balancing benefits and risks of iron supplementation in
resource-rich countries. J. Pediatr167(Suppl.):S20–25 [PubMed: 26364020]
71. Krebs NF, Westcott J, Butler N, Robinson C, Bell M, Hambidge KM. 2006 Meat as a first
complementary food for breastfed infants: feasibility and impact on zinc intake and status. J.
Pediatr. Gastroenterol. Nutr 42:207–14 [PubMed: 16456417]
72. Kuzawa CW. 1998 Adipose tissue in human infancy and childhood: an evolutionary perspective.
Am. J. Phys. Anthropol 107(Suppl. 27):177–209
73. Lao TT, Tam KF, Chan LY. 2000 Third trimester iron status and pregnancy outcome in non-
anaemic women: pregnancy unfavourably affected by maternal iron excess. Hum. Reprod
15:1843–48 [PubMed: 10920115]
74. Lind T, Lönnerdal B, Stenlund H, Gamayanti IL, Ismail D, et al. 2004 A community-based
randomized controlled trial of iron and zinc supplementation in Indonesian infants: effects on
growth and development. Am. J. Clin. Nutr 80:729–36 [PubMed: 15321815]
75. Lind T, Lönnerdal B, Stenlund H, Ismail D, Seswandhana R, et al. 2003 A community-based
randomized controlled trial of iron and zinc supplementation in Indonesian infants: interactions
between iron and zinc. Am. J. Clin. Nutr 77:883–90 [PubMed: 12663287]
76. Lind T, Seswandhana R, Persson LA, Lönnerdal B. 2008 Iron supplementation of iron-replete
Indonesian infants is associated with reduced weight-for-age. Acta Paediatr 97:770–75 [PubMed:
18422809]
77. Lönnerdal B. 2017 Excess iron intake as a factor in growth, infections, and development of infants
and young children. Am. J. Clin. Nutr 106(Suppl. 6):1681S–87S [PubMed: 29070544]
78. Lönnerdal B, Hernell O. 1994 Iron, zinc, copper and selenium status of breast-fed infants and
infants fed trace element fortified milk-based infant formula. Acta Paediatr 83:367–73 [PubMed:
8025391]
79. Lorenz L, Peter A, Poets CF, Franz AR. 2013 A review of cord blood concentrations of iron status
parameters to define reference ranges for preterm infants. Neonatology 104:194–202 [PubMed:
23989077]
80. Lozoff B, Beard J, Connor J, Felt B, Georgieff M, Schallert T. 2006 Long-lasting neural and
behavioral effects of early iron deficiency in infancy. Nutr. Rev 64(Suppl. 2):S34–43 [PubMed:
16770951]
81. Lozoff B, Brittenham GM, Wolf AW, McClish DK, Kuhnert PM, et al. 1987 Iron deficiency
anemia and iron therapy effects on infant developmental test performance. Pediatrics 79:981–95
[PubMed: 2438638]
Georgieff et al. Page 23
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
82. Lozoff B, Castillo M, Clark KM, Smith JB. 2012 Iron-fortified versus low-iron infant formula:
developmental outcome at 10 years. Arch. Pediatr. Adolesc. Med 166:208–15 [PubMed:
22064877]
83. Lozoff B, Clark KM, Jing Y, Armony-Sivan R, Angelilli ML, Jacobson SW. 2008 Dose–response
relationships between iron deficiency with or without anemia and infant social–emotional
behavior. J. Pediatr 152:696–702 [PubMed: 18410777]
84. Lozoff B, De Andraca I, Castillo M, Smith JB, Walter T, Pino P. 2003 Behavioral and
developmental effects of preventing iron-deficiency anemia in healthy full-term infants. Pediatrics
112:846–54 [PubMed: 14523176]
85. Lozoff B, Wolf A, Jimenez E. 1996 Iron-deficiency anemia and infant development: effects of
extended oral iron therapy. J. Pediatr 129:382–89 [PubMed: 8804327]
86. Lukowski AF, Koss M, Burden MJ, Jonides J, Nelson CA, et al. 2010 Iron deficiency in infancy
and neurocognitive functioning at 19 years: evidence of long-term deficits in executive function
and recognition memory. Nutr. Neurosci 13:54–70 [PubMed: 20406573]
87. Lynch S, Pfeiffer CM, Georgieff MK, Brittenham G, Fairweather-Tait S, et al. 2018 Biomarkers of
Nutrition for Development (BOND)—iron review. J. Nutr 48(Suppl. 1):1001S–67S
88. Majumdar I, Paul P, Talib VH, Ranga S. 2003 The effect of iron therapy on the growth of iron-
replete and iron-deplete children. J. Trop. Pediatr 49:84–88 [PubMed: 12729289]
89. McDonald MC, Abrams SA, Schanler RJ. 1998 Iron absorption and red blood cell incorporation in
premature infants fed an iron-fortified infant formula. Pediatr. Res 44:507–11 [PubMed: 9773838]
90. Mevissen-Verhage EAE, Marcelis JH, Harmsen-Van Amerongen WCM, de Vos NM, Verhoef J.
1985 Effect of iron on neonatal gut flora during the first three months of life. Eur. J. Clin.
Microbiol 4:273–78 [PubMed: 3894015]
91. Mohamed MA, Ahmad T, Macri C, Aly H. 2012 Racial disparities in maternal hemoglobin
concentrations and pregnancy outcomes. J. Perinat. Med 40:141–49 [PubMed: 22718605]
92. Monk C, Georgieff MK, Xu D, Hao X, Bansal R, et al. 2015 Maternal prenatal iron status and
tissue organization in the neonatal brain. Pediatr. Res 79:482–88 [PubMed: 26599151]
93. Murray-Kolb LE, Khatry SK, Katz J, Schaefer BA, Cole PM, LeClerq SC. 2012 Preschool
micronutrient supplementation effects on intellectual and motor function in school-aged Nepalese
children. Arch. Pediatr. Adolesc. Med 166:404–10 [PubMed: 22566538]
94. Mwangi MN, Roth JM, Smit MR, Trijsburg L, Mwangi AM, et al. 2015 Effect of daily antenatal
iron supplementation on Plasmodium infection in Kenyan women: a randomized clinical trial.
JAMA 314:1009–20 [PubMed: 26348751]
95. Neuberger A, Okebe J, Yahav D, Paul M. 2016 Oral iron supplements for children in malaria-
endemic areas. Cochrane Database Syst. Rev 2:CD006589 [PubMed: 26921618]
96. Ojukwu JU, Okebe JU, Yahav D, Paul M. 2009 Oral iron supplementation for preventing or
treating anaemia among children in malaria-endemic areas. Cochrane Database Syst. Rev
3:CD006589
97. Okebe JU, Yahav D, Shbita R, Paul M. 2011 Oral iron supplements for children in malaria-
endemic areas. Cochrane Database Syst. Rev 10:CD006589
98. Paganini D, Uyoga MA, Cercamondi CI, Moretti D, Mwasi E, et al. 2017 Consumption of galacto-
oligosaccharides increases iron absorption from a micronutrient powder containing ferrous
fumarate and sodium iron EDTA: a stable-isotope study in Kenyan infants. Am. J. Clin. Nutr
106:1020–31 [PubMed: 28814396]
99. Paganini D, Zimmermann MB. 2017 The effects of iron fortification and supplementation on the
gut microbiome and diarrhea in infants and children: a review. Am. J. Clin. Nutr
106(Suppl.):1688S–93S [PubMed: 29070552]
100. Pasricha SR, Hayes E, Kaumba K, Biggs BA. 2013 Effect of daily iron supplementation on health
in children aged 4–23 months: a systematic review and meta-analysis of randomized controlled
trials. Lancet Glob. Health 1:e77–86 [PubMed: 25104162]
101. Pena-Rosas JP, De-Regil LM, Dowswell T, Viteri FE. 2012 Daily oral iron supplementation
during pregnancy. Cochrane Database Syst. Rev 12:CD004736 [PubMed: 23235616]
Georgieff et al. Page 24
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
102. Pena-Rosas JP, De-Regil LM, Garcia-Casal MN, Dowswell T. 2015 Daily oral iron
supplementation during pregnancy. Cochrane Database Syst. Rev CD004736 [PubMed:
26198451]
103. Petry CD, Eaton MA, Wobken JD, Mills MM, Johnson DE, Georgieff MK. 1992 Iron deficiency
of liver, heart, and brain in newborn infants of diabetic mothers. J. Pediatr 121:109–14 [PubMed:
1625067]
104. Petry N, Olofin I, Hurrell RF, Boy E, Wirth JP, et al. 2016 The proportion of anemia associated
with iron deficiency in low, medium, and high human development index countries: a systematic
analysis of national surveys. Nutrients 8:693
105. Pisansky MT, Wickham RJ, Su J, Fretham S, Yuan L-L, et al. 2013 Iron deficiency with or
without anemia impairs prepulse inhibition of the startle reflex. Hippocampus 23:952–62
[PubMed: 23733517]
106. Rao R, Ennis K, Lubach G, Lock E, Georgieff MK, Coe C. 2016 Metabolomic analysis of CSF
indicates brain metabolic impairment precedes hematological indices of anemia in the iron-
deficient infant monkey. Nutr. Neurosci 21:40–48 [PubMed: 27499134]
107. Ren A, Wang J, Ye RW, Li S, Liu JM, Li Z. 2007 Low first-trimester hemoglobin and low birth
weight, preterm birth and small for gestational age newborns. Int. J. Gynaecol. Obstet 98:124–28
[PubMed: 17585914]
108. Rivera JA, Shamah T, Villalpando S, Monterrubio E. 2010 Effectiveness of a large-scale iron-
fortified milk distribution program on anemia and iron deficiency in low-income young children
in Mexico. Am. J. Clin. Nutr 91:431–39 [PubMed: 20016011]
109. Roncagliolo M, Garrido M, Walter T, Peirano P, Lozoff B. 1998 Interaction of iron deficiency
anemia and neurofunctions in cognitive development. Am. J. Clin. Nutr 68:683–90 [PubMed:
9734748]
110. Santos DCC, Angulo-Barroso RM, Li M, Bian Y, Sturza J, et al. 2018 Timing, duration and
severity of iron deficiency in early development and motor outcomes at 9 months. Eur. J. Clin.
Nutr 72:332–41 [PubMed: 29235557]
111. Sazawal S, Black RE, Ramsan M, Chwaya HM, Stoltzfus RJ. 2006 Effects of routine prophylactic
supplementation with iron and folic acid on admission to hospital and mortality in preschool
children in a high malaria transmission setting: community-based, randomised, placebo-
controlled trial. Lancet 367:133–43 [PubMed: 16413877]
112. Sazawal S, Dhingra U, Dhingra P, Hiremath G, Sarkar A, Dutta A. 2010 Micronutrient fortified
milk improves iron status, anemia and growth among children 1–4 years: a double masked,
randomized, controlled trial. PLOS ONE 5:e12167 [PubMed: 20730057]
113. Scanlon KS, Yip R, Schieve LA, Cogswell ME. 2000 High and low hemoglobin levels during
pregnancy: differential risks for preterm birth and small for gestational age. Obstet. Gynecol
96:741–48 [PubMed: 11042311]
114. Schmidt AT, Waldow KJ, Grove WM, Salinas JA, Georgieff MK. 2007 Dissociating the long-term
effects of fetal/neonatal iron deficiency on three types of learning in the rat. Behav. Neurosci
121:475–82 [PubMed: 17592938]
115. Schmidt RJ, Tancredi DJ, Krakowiak P, Hansen RL, Ozonoff S. 2014 Maternal intake of
supplemental iron and risk of autism spectrum disorder. Am. J. Epidemiol 180:890–900
[PubMed: 25249546]
116. Scholl TO. 1998 High third-trimester ferritin concentration: associations with very preterm
delivery, infection, and maternal nutritional status. Obstet. Gynecol 92:161–66 [PubMed:
9699743]
117. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, et al. 2016 The gut
microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut
65:575–83 [PubMed: 26511795]
118. Senga EL, Harper G, Koshy G, Kazembe PN, Brabin BJ. 2011 Reduced risk for placental malaria
in iron deficient women. Malar. J 10:47 [PubMed: 21345193]
119. Shao J, Lou J, Rao R, Georgieff MK, Kaciroti N, et al. 2012 Maternal serum ferritin
concentration is positively associated with newborn iron stores in women with low ferritin status
in late pregnancy. J. Nutr 142:2004–9 [PubMed: 23014493]
Georgieff et al. Page 25
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
120. Siddappa AM, Georgieff MK, Wewerka S, Worwa C, Nelson CA, deRegnier R-A. 2004 Auditory
recognition memory in iron-deficient infants of diabetic mothers. Pediatr. Res 55:1034–41
[PubMed: 15155871]
121. Siddappa AM, Rao R, Long JD, Widness JA, Georgieff MK. 2007 The assessment of newborn
iron stores at birth: a review of the literature and standards for ferritin concentrations.
Neonatology 92:73–82 [PubMed: 17361090]
122. Simonyté Sjodin K, Domellöf M, Lagerqvist C, Hernell O, Lönnerdal B, et al. 2018
Administration of ferrous sulfate drops has significant effects on the gut microbiota of iron-
sufficient infants: a randomized controlled study. Gut. In press 10.1136/gutjnl-2018-316988
123. Soofi S, Cousens S, Iqbal SP, Akhund T, Khan J, et al. 2013 Effect of provision of daily zinc and
iron with several micronutrients on growth and morbidity among young children in Pakistan: a
cluster-randomised trial. Lancet 382:29–40 [PubMed: 23602230]
124. Steinmacher J, Pohlandt F, Bode H, Sander S, Kron M, Franz AR. 2007 Randomized trial of early
versus late enteral iron supplementation in infants with a birth weight of less than 1301 grams:
neurocognitive development at 5.3 years’ corrected age. Pediatrics 120:538–46 [PubMed:
17766527]
125. Stekel A, Olivares M, Pizarro F, Chadud P, Cayazzo M, et al. 1986 [Prevention of iron deficiency
in infants by fortified milk: field study of a low-fat milk]. Arch. Latinoam. Nutr 36:654–61 (In
Spanish) [PubMed: 3435215]
126. Stoltzfus RJ, Dreyfuss ML, eds. 1998 Guidelines for the Use of Iron Supplements to Prevent and
Treat Iron Deficiency Anemia. Washington, DC: ILSI Press
127. Stoltzfus RJ, Kvalsvig JD, Chwaya HM, Montresor A, Albonico M, et al. 2001 Effects of iron
supplementation and anthelmintic treatment on motor and language development of preschool
children in Zanzibar: double blind, placebo controlled study. BMJ 323:1389–93 [PubMed:
11744561]
128. Suchdev PS, Williams AM, Mei Z, Flores-Ayala R, Pasricha S-R, et al. 2017 Assessment of iron
status in settings of inflammation: challenges and potential approaches. Am. J. Clin. Nutr
106(Suppl. 6):1626S–33S [PubMed: 29070567]
129. Sweet DG, Savage G, Tubman TR, Lappin TR, Halliday HL. 2001 Study of maternal influences
on fetal iron status at term using cord blood transferrin receptors. Arch. Dis. Child 84:F40–43
130. Tamura T, Goldenberg RL, Hou J, Johnston KE, Cliver SP, et al. 2002 Cord serum ferritin
concentrations and mental and psychomotor development of children at five years of age. J.
Pediatr 140:165–70 [PubMed: 11865266]
131. Tamura T, Goldenberg RL, Johnston KE, Cliver SP, Hickey CA. 1996 Serum ferritin: a predictor
of early spontaneous preterm delivery. Obstet. Gynecol 87:360–65 [PubMed: 8598955]
132. Tang M, Frank DN, Hendricks E, Ir D, Esamai F, et al. 2017 Iron in micronutrient powder
promotes an unfavorable gut microbiota in Kenyan infants. Nutrients 9:776
133. Tang M, Frank DN, Sherlock L, Ir D, Robertson CE, Krebs NF. 2016 Effect of vitamin E with
therapeutic iron supplementation on iron repletion and gut microbiome in US iron deficient
infants and toddlers. J. Pediatr. Gastroenterol. Nutr 63:379–85 [PubMed: 27548249]
134. Taylor CL, Brannon PM. 2017 Introduction to workshop on iron screening and supplementation
in iron-replete pregnant women and young children. Am. J. Clin. Nutr 106(Suppl. 6):1547S–54S
[PubMed: 29070553]
135. Tielsch JM, Khatry SK, Stoltzfus RJ, Katz J, LeClerq SC, et al. 2007 Effect of daily zinc
supplementation on child mortality in southern Nepal: a community-based, cluster randomised,
placebo-controlled trial. Lancet 370:1230–39 [PubMed: 17920918]
136. Toldi G, Stenczer B, Molvarec A, Takats Z, Beko G, et al. 2010 Hepcidin concentrations and iron
homeostasis in preeclampsia. Clin. Chem. Lab. Med 48:1423–26 [PubMed: 20626300]
137. Tran PV, Kennedy BC, Lien YC, Simmons RA, Georgieff MK. 2015 Fetal iron deficiency induces
chromatin remodeling at the
Bdnf
locus in adult rat hippocampus. Am. J. Physiol 308:R276–82
138. Tran PV, Kennedy BC, Pisansky MT, Won KJ, Gewirtz JC, et al. 2016 Prenatal choline
supplementation diminishes early-life iron deficiency–induced preprogramming of networks
associated with behavioral abnormalities in the adult rat hippocampus. J. Nutr 146:484–93
[PubMed: 26865644]
Georgieff et al. Page 26
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
139. Unger EL, Hurst AR, Georgieff MK, Schallert T, Rao R, et al. 2012 Behavior and monoamine
deficits in prenatal and perinatal iron deficiency are not corrected by early postnatal moderate-
iron or high-iron diets in rats. J. Nutr 142:2040–49 [PubMed: 22990465]
140. US Prevent. Serv. Task Force. 2015 Iron deficiency anemia in pregnant women: screening and
supplementation. US Prevent. Serv. Task Force https://www.uspreventiveservicestaskforce.org/
Page/Document/UpdateSummaryFinal/iron-deficiency-anemia-in-pregnant-women-screening-
and-supplementation
141. van der Merwe LF, Eussun SR. 2017 Iron status of young children in Europe. Am. J. Clin. Nutr
106(Suppl. 6.):1663S–71S [PubMed: 29070549]
142. van Santen S, Kroot JJ, Zijderveld G, Wiegerinck ET, Spaanderman ME, Swinkels DW. 2013 The
iron regulatory hormone hepcidin is decreased in pregnancy: a prospective longitudinal study.
Clin. Chem. Lab. Med 51:1395–401 [PubMed: 23241678]
143. Veenemans J, Schouten LR, Ottenhof MJ, Mank TG, Uges DR, et al. 2012 Effect of preventive
supplementation with zinc and other micronutrients on non-malarial morbidity in Tanzanian pre-
school children: a randomized trial. PLOS ONE 7:e41630 [PubMed: 22870238]
144. Verhoeff FH, Brabin BJ, van Buuren S, Chimsuku L, Kazembe P, et al. 2001 An analysis of intra-
uterine growth retardation in rural Malawi. Eur. J. Clin. Nutr 55:682–89 [PubMed: 11477467]
145. Villalpando S, Shamah T, Rivera JA, Lara Y, Monterrubio E. 2006 Fortifying milk with ferrous
gluconate and zinc oxide in a public nutrition program reduced the prevalence of anemia in
toddlers. J. Nutr 136:2633–37 [PubMed: 16988138]
146. Vricella LK. 2017 Emerging understanding and measurement of plasma volume expansion in
pregnancy. Am. J. Clin. Nutr 106(Suppl. 6):1620S–25S [PubMed: 29070547]
147. Walker S, Wachs T, Gardner J, Lozoff B, Wasserman G, et al. 2007 Child development: risk
factors for adverse outcomes in developing countries. Lancet 369:145–57 [PubMed: 17223478]
148. Walker S, Wachs T, Grantham-McGregor S, Black M, Nelson C, et al. 2011 Inequality in early
childhood: risk and protective factors for early child development. Lancet 378:1325–38
[PubMed: 21944375]
149. Wallace DF. 2016 The regulation of iron absorption and homeostasis. Clin. Biochem. Rev 37:51–
62 [PubMed: 28303071]
150. Wallin DJ, Tkac I, Stucker S, Ennis KM, Sola-Visner M, et al. 2015 Phlebotomy-induced anemia
alters hippocampal neurochemistry in neonatal mice. Pediatr. Res 77:765–71 [PubMed:
25734245]
151. Walter T, De Andraca I, Chadud P, Perales CG. 1989 Iron deficiency anemia: adverse effects on
infant psychomotor development. Pediatrics 84:7–17 [PubMed: 2472596]
152. Walter T, Pino P, Pizarro F, Lozoff B. 1998 Prevention of iron-deficiency anemia: comparison of
high- and low-iron formulas in term healthy infants after six months of life. J. Pediatr 132:635–
40 [PubMed: 9580762]
153. WHO (World Health Organ.). 2007 Conclusions and recommendations of the WHO consultation
on prevention and control of iron deficiency in infants and young children in malaria-endemic
areas. Food Nutr. Bull 28(Suppl. 4):S621–27 [PubMed: 18297899]
154. WHO (World Health Organ.). 2014 World Malaria Report 2014. Geneva: WHO
155. WHO (World Health Organ.). 2016 Guideline: Daily Iron Supplementation in Infants and
Children. Geneva: WHO
156. WHO (World Health Organ.). 2016 WHO guideline development group—the use of ferritin
concentrations to assess iron status in populations. WHO. http://www.who.int/nutrition/events/
2016_meeting_guidelinedevelopmentgroup_ferritin_2to4march/en/
157. WHO (World Health Organ.). 2019 Iron with or without folic acid supplementation in women in
malaria-endemic areas: full set of recommendations. WHO. http://www.who.int/elena/titles/
full_recommendations/ifa_supplementation_malaria/en/
158. WHO (World Health Organ.), UNICEF. 2006 Iron Supplementation of Young Children in
Regions Where Malaria Transmission Is Intense and Infectious Disease Highly Prevalent.
Geneva: WHO, UNICEF
159. Xiong X, Buekens P, Alexander S, Demianczuk N, Wollast E. 2000 Anemia during pregnancy and
birth outcome: a meta-analysis. Am. J. Perinatol 17:137–46 [PubMed: 11012138]
Georgieff et al. Page 27
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
160. Youdim MB, Sills MA, Heydorn WE, Creed GJ, Jacobowitz DM. 1986 Iron deficiency alters
discrete proteins in rat caudate nucleus and nucleus accumbens. J. Neurochem 47:794–99
[PubMed: 2942640]
161. Zamora TG, Guiang SF, Georgieff MK, Widness JA. 2016 Iron is prioritized to red blood cells
over the brain in phlebotomized anemic newborn lambs. Pediatr. Res 79:922–28 [PubMed:
26866907]
162. Zhang C, Rawal S. 2017 Dietary iron intake, iron status and gestational diabetes. Am. J. Clin.
Nutr 106(Suppl. 6.):1672S–80S [PubMed: 29070554]
163. Zhang Q, Ananth CV, Li Z, Smulian JC. 2009 Maternal anaemia and preterm birth: a prospective
cohort study. Int. J. Epidemiol 38:1380–89 [PubMed: 19578127]
164. Zhao G, Guobin X, Zhou M, Jiang Y, Richards B, et al. 2015 Prenatal iron supplementation
reduces maternal anemia, iron deficiency, iron deficiency anemia in a randomized clinical trial in
rural China, but iron deficiency remains widespread in mothers and neonates. J. Nutr 145:1916–
23 [PubMed: 26063068]
165. Zhou LM, Yang WW, Hua JZ, Deng CQ, Tao X, Stoltzfus RJ. 1998 Relation of hemoglobin
measured at different times in pregnancy to preterm birth and low birth weight in Shanghai,
China. Am. J. Epidemiol 148:998–1006 [PubMed: 9829872]
166. Ziaei S, Norrozi M, Faghihzadeh S, Jafarbegloo E. 2007 A randomized placebo-controlled trial to
determine the effect of iron supplementation on pregnancy outcome in pregnant women with
haemoglobin ≥13.2 g/dl. BJOG 114:684–88. Corrigendum. 2007. BJOG. 114:1311 [PubMed:
17516958]
167. Ziegler EE, Nelson SE, Jeter JM. 2009 Iron supplementation of breastfed infants from an early
age. Am. J. Clin. Nutr 89:525–32 [PubMed: 19073791]
168. Ziegler EE, Nelson SE, Jeter JM. 2009 Iron status of breastfed infants is improved equally by
medicinal iron and iron-fortified cereal. Am. J. Clin. Nutr 90:76–87 [PubMed: 19458014]
169. Ziegler EE, O’Donnell AM, Nelson SE, Fomon SJ. 1976 Body composition of the reference fetus.
Growth 40:329–41 [PubMed: 1010389]
170. Zlotkin SH, Davidsson L, Lozoff B. 2015 Balancing the benefits and risks of iron fortification in
resource-constrained settings. J. Pediatr 167(Suppl.):S26–30 [PubMed: 26364021]
Georgieff et al. Page 28
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
NPBI: nonprotein-bound iron
Georgieff et al. Page 29
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
WHO: World Health Organization
Georgieff et al. Page 30
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
AAP: American Academy of Pediatrics
Georgieff et al. Page 31
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
RCT: randomized controlled trial
Georgieff et al. Page 32
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Figure 1.
The U-shaped risk curve exhibited by most nutrients. Iron has a narrower adequacy range
(
red dashed lines
), and risks from deficiency or overload states are greater compared with
those from most other nutrients.
Georgieff et al. Page 33
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Georgieff et al. Page 34
Table 1
Interpretation and risk–benefit analysis of maternal iron supplementation during pregnancy based on whether
hemoglobin or ferritin was used as the primary biomarker to assess iron status and the information added by a
second biomarker
Primarymarker Literatureinterpretation
Biochemical
interpretation with
second biomarker
Agreement
between
literature and
biochemical
finding?
Low hepcidin
(likely response
to therapy)
Estimate of risk-
benefit of routine
supplementationa
Hemoglobin
Low Iron-deficiency anemia Low ferritin: iron-
deficiency anemia Low ferritin: yes Low ferritin: yes B R
Normal ferritin:
anemia of
inflammation
Normal ferritin:
unknown because
total-body iron is
unmeasurable
Normal ferritin:
no Unknown, but R > B
because iron will not be
absorbed in high
hepcidin state
High ferritin: anemia
of inflammation High ferritin:
unknown because
total-body iron is
unmeasurable
High ferritin: no
Normal Iron sufficient Low ferritin: low-iron
state Low ferritin: no Low ferritin: yes B > R
Normal ferritin: iron
sufficient Normal ferritin:
yes Normal ferritin:
no Unknown
High ferritin: iron
overload High ferritin: no High ferritin: no R > B
High Iron sufficient Low ferritin:
polycythemia by
volume contraction or
other non-iron-related
condition
Low ferritin:
nonanemia iron
deficiency
Low ferritin: yes R > B
Normal ferritin:
polycythemia by
volume contraction or
other non-iron-related
condition
Normal ferritin:
iron overload Normal ferritin:
no R > B
High ferritin: iron
overload High ferritin: iron
overload High ferritin: no R > B
Ferritin
Low Low-iron state Low hemoglobin:
iron-deficiency
anemia
Low hemoglobin:
partly Low
hemoglobin: yes B R
Normal hemoglobin:
nonanemia iron
deficiency
Normal
hemoglobin: yes Normal
hemoglobin: yes B > R
High hemoglobin:
polycythemia by
volume contraction or
other non-iron-related
condition
High hemoglobin:
no High
hemoglobin: no R > B
Normal Iron sufficient Low hemoglobin:
anemia of
inflammation
Low hemoglobin:
no Low
hemoglobin: no R > B
Normal hemoglobin:
iron sufficient Normal
hemoglobin: yes Normal
hemoglobin: no Unknown
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Georgieff et al. Page 35
Primarymarker Literatureinterpretation
Biochemical
interpretation with
second biomarker
Agreement
between
literature and
biochemical
finding?
Low hepcidin
(likely response
to therapy)
Estimate of risk-
benefit of routine
supplementationa
High hemoglobin:
polycythemia by
volume contraction or
other non-iron-related
condition
High hemoglobin:
no High
hemoglobin: no R > B
High Iron sufficient Low hemoglobin:
anemia of
inflammation
Low hemoglobin:
no Low
hemoglobin: no R > B
Normal hemoglobin:
iron overload Normal
hemoglobin: no Normal
hemoglobin: no R > B
High hemoglobin:
iron overload High hemoglobin:
no High
hemoglobin: no R > B
Abbreviations: B, benefit; R, risk.
a
Estimate of whether the risk (R) of adverse outcomes or the benefit (B) associated with iron supplementation is greater, given each combination of
iron markers.
Annu Rev Nutr
. Author manuscript; available in PMC 2020 April 21.
... Interleukin 6 (IL-6), the primary hepcidin regulator, disturbs iron homeostasis and leads to the expression of vascular cell adhesion molecule (VCAM1) and intercellular adhesion molecules (ICAM-1), maintaining endothelium adhesive phenotype. [30][31][32][33][34][35][36][37] In case of oversupplementation of iron, the transferrin potential may get exhausted or exceed 85%, leading to an increase in nontransferrin-related Fe (NTBI), increasing the risk of intraocular and cardiac Fe accumulation. Excessive Fe accumulation in organs, such as the heart, liver, and endocrine glands, impairs their function, producing cardiomyopathy, cirrhosis, insulin-dependent diabetes, etc. Oxidative stress leads to faster aging of the cells, which gives a signal to the uterus to contract, resulting in premature birth. ...
... Copper deficiency can hamper cytochrome and bone collagen synthesis, directly affecting fetal development and leading to FGR. 59,[65][66][67][68] Iron Pregnancy with iron deficiency reduces fetal growth ( Figure 5) and affects neuronal growth, especially in the myelinization of neurons, neural signals transmission, frontal cortex development, and basal ganglia. 34,64 Decreased iron leads to decreased hemoglobin, restricting oxygen circulation and forming an environment of chronic hypoxia, leading to fetal growth restriction. Iron deficiency stimulates the synthesis of corticotropin-releasing hormone via increased norepinephrine and, in turn, hinders the growth of the fetus. ...
... All these can lead to vascular endothelial damage and disturbance in endothelial homeostasis resulting in vasoconstriction and increased blood pressure. 34 In the third trimester of pregnancy, there was a positive correlation between ferritin, iron, and superoxide concentrations with vascular stiffness. The World Health Organization recommends universal iron supplementation of 30-60 mg/day throughout pregnancy. ...
Article
Full-text available
About 10 to 20% of reported pregnancies have complications like spontaneous abortion (SA), preeclampsia (PE), preterm birth (PTB), and fetal growth restriction (FGR); 60% are attributed to maternal nutritional alterations. Multiple micronutrients (MMN) are supplemented in the antenatal period, but no proper validation/guidelines are available regarding dosing/time, the need for initiation, and the duration of supplementation. Studies have reported adverse pregnancy complications related to the overuse/unwanted use of multiple micronutrient supplementations during pregnancy. Identifying the exact population requiring supplementation is necessary to prevent its abuse. This article attempts to review the impacts of micronutrient deficiency/supplementation in cases of SA, FGR, and gestational diabetes mellitus (GDM), preterm delivery and PE. The study used a literature search using PubMed, Google Scholar, Mendeley, and Scopus Databases using search words pregnancy, spontaneous abortion, gestational diabetes mellitus (GDM), fetal growth restriction (FGR), preterm delivery, preeclampsia (PE) or “adverse pregnancy” associated with minerals, micronutrients, or supplementation. The review also considered in-house literature databases, a single-window search at Kasturba Medical College (KMC) Health sciences library, MAHE (Manipal Academy of Higher Education). The figures included in the study were created by Biorender.com. Micronutrients play multiple roles during pregnancy and fetoplacental growth stimulating growth hormone secretion, Lysyl oxidase (LOX), involved in the crosslinking between collagen and elastin in the amniotic membrane, downregulation of interleukin (IL)-1 alpha, IL-1 beta, IL-4, IL-6, Il-10, IL-12, tumor necrosis factor (TNF)-alpha and several chemokines involved in hypertension, immune-inflammatory pathways, attenuate insulin resistance, structural development of neurons and glia. Over-supplementation has led to complications such as spontaneous abortion and gestational diabetes mellitus. Since there is a lack of standardization concerning micronutrient supplementation during pregnancy, there is a need for systematic study related to the role of micronutrients during each trimester of pregnancy to optimize its supplementation and to prevent hazards associated with its abuse.
... In infants younger than about 9 months of age, iron homeostasis is incompletely developed, with less capacity to downregulate iron absorption in response to an increased iron intake (Donker et al., 2021;Georgieff et al., 2019;Lönnerdal, 2017;Lönnerdal et al., 2015), in contrast to the adult general population. By 9 months of age, some homeostatic regulatory capacity has developed, but the extent of control of iron absorption with increased iron intake in young children is unknown. ...
... 3.5.6 | Other adverse effects of iron supplementation during pregnancy Studies have reported that excess iron from iron supplementation during pregnancy may have adverse effects on maternal (Baker et al., 2018;Dewey & Oaks, 2017;Iqbal & Ekmekcioglu, 2019), birth (Georgieff et al., 2019;Shastri et al., 2015) and infant outcomes (Jayasinghe et al., 2018;Quezada-Pinedo et al., 2021;Wessling-Resnick, 2017). Associations of iron supplementation with birth weight [absolute birth weight, low birth weight, small-for-gestational age (SGA) births], IUGR, still births, pre-term births, pre-term labour, pre-eclampsia and impaired cognitive outcomes in the offspring later in life have been described. ...
Article
Full-text available
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for iron. Systematic reviews were conducted to identify evidence regarding high iron intakes and risk of chronic diseases, adverse gastrointestinal effects and adverse effects of iron supplementation in infancy, young childhood and pregnancy. It is established that systemic iron overload leads to organ toxicity, but no UL could be established. The only indicator for which a dose–response could be established was black stools, which reflect the presence of large amounts of unabsorbed iron in the gut. This is a conservative endpoint among the chain of events that may lead to systemic iron overload but is not adverse per se. Based on interventions in which black stools did not occur at supplemental iron intakes of 20–25 mg/day (added to a background intake of 15 mg/day), a safe level of intake for iron of 40 mg/day for adults (including pregnant and lactating women) was established. Using allometric scaling (body weight0.75), this value was scaled down to children and adolescents and safe levels of intakes between 10 mg/day (1–3 years) and 35 mg/day (15–17 years) were derived. For infants 7–11 months of age who have a higher iron requirement than young children, allometric scaling was applied to the supplemental iron intakes (i.e. 25 mg/day) and resulted in a safe level of supplemental iron intake of 5 mg/day. This value was extended to 4–6 month‐old infants and refers to iron intakes from fortified foods and food supplements, not from infant and follow‐on formulae. The application of the safe level of intake is more limited than a UL because the intake level at which the risk of adverse effects starts to increase is not defined.
... Pemberian suplemen penambah darah atau tablet besi menjadi salah satu langkah untuk menurunkan angka anemia defisiensi besi diantara ibu hamil di negara berkembang, seperti Indonesia (Kementerian Kesehatan RI, 2020). Kehamilan dapat meningkatkan risiko anemia karena volume darah yang meningkat untuk memenuhi kebutuhan janin (Georgieff et al., 2019). ...
Article
Pemberian tablet besi menjadi salah satu langkah untuk mencegah anemia diantara ibu hamil di Indonesia. Akan tetapi, tingkat kepatuhan konsumsi tablet besi diantara ibu hamil masih bervariasi dan cenderung rendah, yaitu 56%. Literasi kesehatan ibu menjadi salah satu faktor yang berkontribusi dalam kepatuhan terhadap regimen medis. Penelitian ini bertujuan untuk menganalisis hubungan antara literasi kesehatan ibu dan kepatuhan dalam konsumsi tablet besi diantara ibu hamil. Penelitian ini merupakan penelitian cross-sectional. 305 ibu hamil di wilayah kerja Puskesmas Sukoharjo, Jawa Tengah dan Puskemas Kediri, Jawa Timur dengan usia minimal 18 tahun dan tidak memiliki komplikasi kehamilan berpartisipasi dalam penelitian ini. Mayoritas ibu hamil memiliki tingkat literasi kesehatan yang kurang (inadequate). Uji ANOVA menyimpulkan terdapat perbedaan signifikan antara literasi kesehatan ibu dan kepatuhan konsumsi tablet besi ((F3, 301) = 48.48, p<0.001, ?p2 = 0.325). Perbedaan tersebut ditemukan diantara keempat level literasi kesehatan ibu (inadequate, problematic, sufficient, excellent) sesuai hasil post hoc analysis. Ibu hamil dengan literasi kesehatan lebih tinggi memiliki tingkat kepatuhan konsumsi tablet besi yang lebih baik. Profesional kesehatan dan penyedia layanan kesehatan disarankan untuk memperkuat program literasi kesehatan ibu hamil dengan tujuan meningkatkan pemahaman ibu hamil tentang manfaat tablet besi dan mengonsumsinya secra teratur. Upaya ini diharapkan mampu membantu mengurangi angka anemia pada ibu hamil di Indonesia.
... Our previous work on these pregnant women also revealed low protein status [43], multiple micronutrient deficiencies (folic acid, B12, vitamin A and D), inflammation measured through elevated C-reactive protein (CRP) [44], and high prevalence of oral, skin, urogenital, and intestinal parasitic infections [45]. As iron supplementation may be counter-productive in conditions of infection/inflammation [46], and as there is evidence that anemia and iron status are variably modulated by different infections [47], we postulated that the combination of multiple infections, nutrient deficiencies, and inflammation might contribute to the persistent anemia and differentially impact individual iron status indicators in pregnant women from marginalized communities. ...
Article
Full-text available
In pregnant women with multiple infections, nutrient deficiencies, and inflammation (MINDI), the study of anemia and iron status is limited. For this cross-sectional study (n = 213 Panamanian indigenous women), we investigated if hemoglobin, anemia (Hb < 110 g/L), ferritin, serum iron, serum transferrin receptor, and hepcidin were associated with (1) maternal nutritional status and supplementation practices, (2) biomarkers of inflammation, and (3) presence/absence of infections. Hierarchical generalized linear and logistic regression models and dominance analyses identified the relative importance of these predictors. Anemia (38%), which was likely underestimated due to low plasma volume (95%), was associated with lower ferritin, vitamin A, and weight-for-height, suggesting anemia of undernutrition. Inflammation was not associated with Hb or anemia; nevertheless, higher CRP was associated with increased odds of low serum iron and higher ferritin and hepcidin, indicating iron restriction due to inflammation. The length of iron supplementation did not enter models for anemia or iron indicators, but a multiple nutrient supplement was associated with higher ferritin and hepcidin. Moreover, iron supplementation was associated with higher odds of vaginal trichomoniasis but lower odds of caries and bacterial vaginosis. The complex pathogenesis of anemia and iron deficiency in MINDI settings may require other interventions beyond iron supplementation.
... The high burden of anaemia in pregnancy, therefore, emphasizes the pressing need to comprehensively understand the determinants behind the ubiquity of MSA during pregnancy. Furthermore, the third trimester is a crucial period in pregnancy because the demand for iron increases as the fetus undergoes rapid growth and development in utero 14 . Therefore, in this study, we delved into the multifaceted landscape of MSA in pregnancy by determining the prevalence and the trends of MSA, and then explored the intricate web of determinants that drive this prevalence among women in the third trimester of pregnancy (28 to 36 weeks) in Lagos, Nigeria. ...
Article
Full-text available
The high burden of anaemia during pregnancy underscores the urgent need to gain a comprehensive understanding of the factors contributing to its widespread occurrence. Our study assessed the prevalence and the trends of moderate-to-severe anaemia (MSA) in late pregnancy (28 to 36 weeks) and then investigated the key determinants driving this prevalence among women in Lagos, Nigeria. We conducted a secondary data analysis involving 1216 women enrolled in the Predict-PPH study between January and March 2023. We employed a multivariate binary logistic regression model with a backward stepwise selection approach to identify significant predictors of MSA. The study revealed a 14.5% prevalence of MSA during pregnancy. Independent predictors of MSA included having given birth to two or more children (adjusted odds ratio = 1.46, 95% confidence interval: 1.03–2.07), having a maternal body mass index (BMI) of 28 kg/m² or higher (adjusted odds ratio = 1.84, 95% confidence interval: 1.29–2.61), having less than tertiary education (adjusted odds ratio = 1.51, 95% confidence interval: 1.08–2.11), and being unemployed (adjusted odds ratio = 1.97, 95% confidence interval: 1.19–3.26). It is crucial for pregnant women, particularly those with higher parities and elevated BMI, to be monitored regularly for anaemia and its consequences during their antenatal care. Additionally, addressing the link between low education, unemployment, and anaemia necessitates comprehensive strategies that empower women in terms of education and economic status to enhance the overall well-being of individuals and communities, ultimately reducing the prevalence of anaemia and associated health issues in pregnancy.
... As a result, pregnant women not using the above anaemia prevention services constituted a higher proportion of those with anaemia. It is understood that iron folic supplementation helps replenish depleted iron stores in the body of pregnant women [25]. Sulfadoxine pyrimethamine prophylaxis helps reduce malaria incidence among pregnant women [26]. ...
Article
Full-text available
INTRODUCTION Anaemia in pregnancy has been recognized as a public health problem of importance, especially in the developing world. This is due to related adverse outcomes among pregnant women and the unborn. However, the epidemiology of anaemia in pregnancy is unknown locally. This study sought to establish the prevalence and determinants of anaemia among pregnant women in a referral hospital in coastal Kenya. MATERIALS AND METHODS This was a cross-sectional study conducted among 191 pregnant women attending the antenatal clinic (ANC) at Kilifi County Referral Hospital. A pre-tested structured questionnaire was used to collect data and data was analysed using STATA version 15. Descriptive statistics were used to describe selected variables and logistic regression was used to determine the significance of factors associated with anaemia. RESULTS The prevalence of anaemia was 54% with factors associated with increased odds of anaemia being household size (AOR 1.27, 95%CI: 1.01-1.59, p=0.04), history of malaise and fever (AOR 9.69, 95%CI: 2.34-40.11, P < 0.01). Factors associated with reduced odds of anaemia were; food frequency (AOR 0.23, 95%CI: 0.1-0.51, P < 0.01), increased intake of vegetables and fruits (AOR 0.47, 95%CI: 0.34-0.47, P < 0.001), and red meat (AOR 0.39, 95%CI: 0.23-0.67, P < 0.001). CONCLUSION The relatively high prevalence of anaemia among pregnant women attending antenatal care suggests that anaemia is still a significant public health problem in Coastal Kenya. Increased household size and, a history of malaise and fever, were positively associated with anaemia while food frequency, vegetables, fruits and red meat intake were associated with a reduced likelihood of developing anaemia. Therefore, the ministries of health and other healthcare stakeholders at the county and national levels should enhance the supply of iron supplements at Kilifi County Hospital and other community-based health facilities targeted for pregnant women. Moreover, nurses and clinicians should intensify health education on anaemia in pregnancy from early antenatal clinic visits for pregnant mothers.
... The elevated potassium levels found in pigeon peas may prove beneficial for individuals taking diuretics to manage blood pressure or experiencing excessive potassium secretion (A'yuni et al. 2021). Iron is crucial for erythrocyte formation across all organs, including the brain, and vital for hemoglobin synthesis in fetuses and growing children is abundant in pigeon peas (Georgieff, Krebs, and Cusick 2019). Additionally, pigeon peas contain zinc, which is essential for maintaining the body's immune system (Cabrera 2015). ...
Article
Full-text available
Pigeon pea is one of the most multipurpose grain legumes in the tropical and subtropical world. It is highly resilient to climate change due to drought tolerance. It ranks sixth following common beans, chickpeas, field peas, cowpeas, and lentils globally. Pigeon pea is an excellent source of high‐quality protein, essential amino acids, and minerals. Furthermore, pigeon pea is endowed with valuable water‐soluble vitamins such as thiamin, ascorbic acid, riboflavin, and niacin. With low levels of saturated fat and sodium and being cholesterol free, pigeon peas emerge as a nutritious dietary choice. Pigeon peas are rich sources of bioactive compounds with various potential health properties, including anti‐inflammatory, antibacterial, antioxidant, anticarcinogenic, and antidiabetic effects. Pigeon peas can be consumed in diverse forms, including whole seeds, split seeds (known as dhal), vegetables using green seeds, and fresh pods. Moreover, pigeon peas find application in a variety of food products, including bread, pasta, noodles, snacks, and biscuits. Despite their nutritional and medicinal properties, pigeon peas remain among the least utilized legumes globally. This review paper aims to provide up‐to‐date information on the nutritional compositions, bioactive compounds, food applications, and health benefits of pigeon peas.
... The initiation of maternal IFA supplementation relatively early in pregnancy, preferably no later than the second trimester, suggests that a dosage of 60 mg of iron may not confer greater e cacy in improving hemoglobin or iron status compared to 30 mg [48]. It's worth noting that MMN are commonly employed for preventing anemia and other adverse pregnancy outcomes, with 30 mg of iron per day likely providing a su cient dosage [49]. ...
Preprint
Full-text available
Background: Iron-Folic Acid (IFA) supplementation during pregnancy is widely recommended to prevent maternal anemia and improve birth outcomes. However, the optimal formulation, dose, and frequency of IFA supplementation remain uncertain. This systematic review and meta-analysis aimed to evaluate the effect of different IFA formulations, doses, and frequencies on pregnancy and neonatal outcomes compared to Multiple Micronutrients (MMN) among pregnant women. Methods: A comprehensive literature search was conducted across PubMed, Google Scholar, Cochrane Library, Scopus, and TRIP databases to identify pertinent studies published up to December 31st, 2023. Outcome measures includes preterm birth (PTB), stillbirths, low birth weight (LBW), small for gestational age (SGA), miscarriage rate (MR), neonatal mortality, and perinatal mortality. Pooled risk ratios (RRs) with 95% confidence intervals (CIs) were calculated, and the quality of evidence was assessed using GRADEpro. Results: Among 20 studies comparing IFA to MMN, our analysis showed elevated risk of stillbirth (RR: 1.08, 95% CI: 1.00 to 1.17), SGA (RR: 1.03, 95% CI: 1.00 to 1.05) and LBW (RR: 1.06, 95% CI: 1.00 to 1.12) associated with IFA. However, non-significant risk of PTB (RR: 1.07, 95% CI: 0.98 to 1.16) and MR (RR: 0.98, 95% CI: 0.88 to 1.09) was observed with IFA as compared to MMN. Neonatal mortality and perinatal mortality also did not significantly differ between the two groups. Certain formulations and doses showed trend of risk, particularly in relation to PTB and stillbirth. Conclusions: Our findings emphasize the importance of carefully considering the potential risks and benefits of IFA supplementation in pregnancy, and suggest the need for further research to elucidate the underlying mechanisms driving these associations and to optimize supplementation strategies for maternal and neonatal health.
... Furthermore, in the human context, iron supplementation in individuals with sufficient iron stores might have negative effects on growth and other health outcomes. 33 Iron overload can have detrimental effects on bone health in healthy individuals. [30][31][32] Therefore, it can be speculated that the offspring in the supplemented control group (Control þ Fe þ DHA/EPA) might not be the best representation of ideal bone development. ...
Article
Full-text available
We previously showed in rats that pre- and postnatal deficiencies in iron and omega-3 (n-3) fatty acids can impair bone development, with additive and potentially irreversible effects when combined. This study aimed to investigate, in female rats consuming a combined iron and n-3 fatty acid deficient (ID + n-3 FAD) diet preconception, whether supplementation with iron and docosahexaenoic/eicosapentaenoic acid (DHA/EPA), alone and in combination, can prevent bone impairments in offspring. Using a 2 × 2 factorial design, female Wistar rats consuming an ID + n-3 FAD diet preconception were randomised to receive an: 1) iron supplemented (Fe + n-3 FAD), 2) DHA/EPA supplemented (ID + DHA/EPA), 3) Fe + DHA/EPA, or 4) ID + n-3 FAD diet from gestational day 10 throughout pregnancy and lactation. Post-weaning, offspring (n = 24/group; male:female = 1:1) remained on the respective experimental diets for three weeks until postnatal day 42–45. Offspring born to female rats consuming a control diet preconception and an Fe+DHA/EPA diet throughout pregnancy and lactation served as non-deficient reference group (Control+Fe+DHA/EPA). Bone mineral density (BMD) was measured using dual-energy X-ray absorptiometry and bone strength using three-point bending tests. Only offspring in the Fe+DHA/EPA group had significantly higher spine and femur BMD, and higher femur stiffness than offspring in the ID + n-3 FAD group, and had similar spine BMD and femur stiffness as the Control + Fe + DHA/EPA group. Offspring in the Fe + DHA/EPA group further had significantly higher femur strength (ultimate load) than the other experimental groups, and a similar femur strength as the Control + Fe + DHA/EPA group. This study shows that only combined iron and DHA/EPA supplementation can prevent bone impairments in offspring of female rats consuming an iron and n-3 FA deficient diet preconception.
Article
Objective: This research paper proposes a new HPTLC method that is quick, accurate and precise for estimating ethinyl estradiol, levonorgestrel and ferrous fumarate. Materials and Methods: Quantification was performed on silica gel precoated aluminum plates as the stationary phase. As the three drugs did not resolved in one mobile phase, different mobile phases were selected for ethinyl estradiol and levonorgestrel i.e. chloroform: methanol in the ratio of 9.9: 0.1 v/v and for ferrous fumarate i.e. formic acid: dichloromethane: butanol: heptane in the ratio of 1.2: 1.6: 3.2: 4.4 v/v/v/v. Result: Wavelength for densitometric evaluation was 275 nm for ethinyl estradiol and levonorgestrel and 212 nm for ferrous fumarate. Ethinyl estradiol and levonorgestrel were acceptably resolved having a RF values of 0.16 ± 0.02 and 0.24 ± 0.02. Ferrous fumarate had a RF value of 0.74 ± 0.02. Conclusion: All the validation parameters were performed and results were found to be within the acceptable limit prescribe by ICH.
Article
Full-text available
More than 20 million babies are born with low birthweight annually. Small newborns have an increased risk for mortality, growth failure, and other adverse outcomes. Numerous antenatal risk factors for small newborn size have been identified, but individual interventions addressing them have not markedly improved the health outcomes of interest. We tested a hypothesis that in low-income settings, newborn size is influenced jointly by multiple maternal exposures and characterized pathways associating these exposures with newborn size. This was a prospective cohort study of pregnant women and their offspring nested in an intervention trial in rural Malawi. We collected information on maternal and placental characteristics and used regression analyses, structural equation modelling, and random forest models to build pathway maps for direct and indirect associations between these characteristics and newborn weight-for-age Z-score and length-for-age Z-score. We used multiple imputation to infer values for any missing data. Among 1,179 pregnant women and their babies, newborn weight-for-age Z-score was directly predicted by maternal primiparity, body mass index, and plasma alpha-1-acid glycoprotein concentration before 20 weeks of gestation, gestational weight gain, duration of pregnancy, placental weight, and newborn length-for-age Z-score (p < .05). The latter 5 variables were interconnected and were predicted by several more distal determinants. In low-income conditions like rural Malawi, maternal infections, inflammation, nutrition, and certain constitutional factors jointly influence newborn size. Because of this complex network, comprehensive interventions that concurrently address multiple adverse exposures are more likely to increase mean newborn size than focused interventions targeting only maternal nutrition or specific infections.
Article
Full-text available
Anaemia and malaria are both common in pregnant women in Sub-Saharan Africa. Previous evidence has shown that iron supplementation may increase malaria risk. In this observational cohort study, we evaluated P. falciparum pathogenesis in vitro in RBCs from pregnant women during their 2nd and 3rd trimesters. RBCs were collected and assayed before (n = 327), 14 days (n = 82), 49 days (n = 112) and 84 days (n = 115) after iron supplementation (60 mg iron as ferrous fumarate daily). P. falciparum erythrocytic stage growth in vitro is reduced in anaemic pregnant women at baseline, but increased during supplementation. The elevated growth rates parallel increases in circulating CD71-positive reticulocytes and other markers of young RBCs. We conclude that Plasmodium growth in vitro is associated with elevated erythropoiesis, an obligate step towards erythroid recovery in response to supplementation. Our findings support current World Health Organization recommendations that iron supplementation be given in combination with malaria prevention and treatment services in malaria endemic areas.
Article
Full-text available
Background/objectives: Poorer motor development is reported in infants with iron deficiency (ID). The role of timing, duration and severity is unclear. We assessed relations between ID timing, duration, and severity and gross motor scores, neurological integrity, and motor behavior quality at 9 months. Subjects/methods: Iron status was determined at birth and 9 months in otherwise healthy term Chinese infants. The 9-month motor evaluation included the Peabody Developmental Motor Scale (PDMS-2), Infant Neurological International Battery (INFANIB), and motor quality factor. Motor outcomes were analyzed by ID timing (fetal-neonatal, infancy), duration, and severity. For severity, we also considered maternal iron status. Results: The data were available for 1194 infants. Iron status was classified as fetal-neonatal and infancy ID (n = 253), fetal-neonatal ID (n = 256), infancy ID (n = 288), and not ID (n = 397). Compared with not ID, infants with fetal-neonatal or infancy ID had lower locomotion scores (effect size ds = 0.19, 0.18) and those with ID in both periods (longer duration) had lower locomotion and overall PDMS-2 gross motor scores (ds = 0.20, 0.18); ID groups did not differ. More severe ID in late pregnancy was associated with lower INFANIB Vestibular function (p = 0.01), and total score (p = 0.03). More severe ID in infancy was associated with lower scores for locomotion (p = 0.03), overall gross motor (p = 0.05). Conclusions: Fetal-neonatal and/or infancy ID was associated with lower overall gross motor development and locomotion test scores at 9 months. Associations with ID severity varied by ID timing: more severe ID in late pregnancy, poorer neurological integrity; more severe ID in infancy, poorer gross motor development.
Article
Objective. To determine the behavioral and developmental effects of preventing iron-deficiency anemia in infancy. Methods. Healthy full-term Chilean infants who were free of iron-deficiency anemia at 6 months were assigned to high- or low-iron groups or to high- or no-added-iron groups. Behavioral/developmental outcomes at 12 months of age included overall mental and motor test scores and specific measures of motor functioning, cognitive processing, and behavior. There were no differences between high- and low-iron groups in the prevalence of iron-deficiency anemia or behavioral/developmental outcome, and they were combined to form an iron-supplemented group (n = 1123) for comparison with the no-added-iron group (n = 534). Results. At 12 months, iron-deficiency anemia was present in 3.1% and 22.6% of the supplemented and unsupplemented groups, respectively. The groups differed in specific behavioral/developmental outcomes but not on global test scores. Infants who did not receive supplemental iron processed information slower. They were less likely to show positive affect, interact socially, or check their caregivers’ reactions. A smaller proportion of them resisted giving up toys and test materials, and more could not be soothed by words or objects when upset. They crawled somewhat later and were more likely to be tremulous. Conclusions. The results suggest that unsupplemented infants responded less positively to the physical and social environment. The observed differences seem to be congruent with current understanding of the effects of iron deficiency on the developing brain. The study shows that healthy full-term infants may receive developmental and behavioral benefits from iron supplementation in the first year of life.
Article
Following a request from the European Commission, the Panel on Dietetic Products, Nutrition and Allergies derived Dietary Reference Values (DRVs) for iron. These include Average Requirement (AR) and Population Reference Intake (PRI). For adults, whole-body iron losses were modelled using data from US adults. Predicted absorption values, at a serum ferritin concentration of 30 µg/L, of 16 % for men and 18 % for women were used to convert physiological requirements to dietary iron intakes. In men, median whole-body iron losses are 0.95 mg/day, and the AR is 6 mg/day. The PRI, calculated as the dietary requirement at the 97.5th percentile, is 11 mg/day. For postmenopausal women, the same DRVs as for men are proposed. In premenopausal women, additional iron is lost through menstruation but, because losses are highly skewed, the Panel set a PRI of 16 mg/day to cover requirements of 95 % of the population. In infants and children, requirements were calculated factorially, taking into consideration the needs for growth, replacement of losses and percentage iron absorption from the diet (10 % up to 11 years and 16 % thereafter). PRIs were estimated using a coefficient of variation of 20 %. They are 11 mg/day in infants (7–11 months), 7 mg/day in children aged 1–6 years and 11 mg/day in children aged 7–11 years and boys aged 12–17 years. For girls aged 12–17 years, the PRI of 13 mg/day is the midpoint of the calculated dietary requirement of 97.5 % of girls and the PRI for premenopausal women; this approach allows for the large uncertainties in the rate and timing of pubertal growth and menarche. For pregnant and lactating women, for whom it was assumed that iron stores and enhanced absorption provide sufficient additional iron, DRVs are the same as for premenopausal women.
Article
This is the fifth in the series of reviews developed as part of the Biomarkers of Nutrition for Development (BOND) program. The BOND Iron Expert Panel (I-EP) reviewed the extant knowledge regarding iron biology, public health implications, and the relative usefulness of currently available biomarkers of iron status from deficiency to overload. Approaches to assessing intake, including bioavailability, are also covered. The report also covers technical and laboratory considerations for the use of available biomarkers of iron status, and concludes with a description of research priorities along with a brief discussion of new biomarkers with potential for use across the spectrum of activities related to the study of iron in human health. The I-EP concluded that current iron biomarkers are reliable for accurately assessing many aspects of iron nutrition. However, a clear distinction is made between the relative strengths of biomarkers to assess hematological consequences of iron deficiency versus other putative functional outcomes, particularly the relationship between maternal and fetal iron status during pregnancy, birth outcomes, and infant cognitive, motor and emotional development. The I-EP also highlighted the importance of considering the confounding effects of inflammation and infection on the interpretation of iron biomarker results, as well as the impact of life stage. Finally, alternative approaches to the evaluation of the risk for nutritional iron overload at the population level are presented, because the currently designated upper limits for the biomarker generally employed (serum ferritin) may not differentiate between true iron overload and the effects of subclinical inflammation.
Article
In infants and young children in Sub-Saharan Africa, iron-deficiency anemia (IDA) is common, and many complementary foods are low in bioavailable iron. In-home fortification of complementary foods using iron-containing micronutrient powders (MNPs) and oral iron supplementation are both effective strategies to increase iron intakes and reduce IDA at this age. However, these interventions produce large increases in colonic iron because the absorption of their high iron dose (≥12.5 mg) is typically <20%. We reviewed studies in infants and young children on the effects of iron supplements and iron fortification with MNPs on the gut microbiome and diarrhea. Iron-containing MNPs and iron supplements can modestly increase diarrhea risk, and in vitro and in vivo studies have suggested that this occurs because increases in colonic iron adversely affect the gut microbiome in that they decrease abundances of beneficial barrier commensal gut bacteria (e.g., bifidobacteria and lactobacilli) and increase the abundance of enterobacteria including entropathogenic Escherichia coli These changes are associated with increased gut inflammation. Therefore, safer formulations of iron-containing supplements and MNPs are needed. To improve MNP safety, the iron dose of these formulations should be reduced while maximizing absorption to retain efficacy. Also, the addition of prebiotics to MNPs is a promising approach to mitigate the adverse effects of iron on the infant gut.
Article
The NIH Office of Dietary Supplements convened a public workshop on iron screening and supplementation in iron-replete pregnant women and young children in 2016 in Bethesda, Maryland. The starting point for the workshop was the recent reports from the US Preventive Services Task Force concluding that there was insufficient evidence to evaluate the benefits and harms associated with iron screening and routine supplementation among asymptomatic pregnant women and young children (6-24 mo old) in the United States. The goal of the workshop was to explore and refine understanding about the existing knowledge gaps and research needs associated with these preventive services for these groups. Given the focus on the United States, planning for the workshop took into account the higher iron status in the United States compared with developing countries and, in turn, included a focus on iron-replete individuals consistent with the U-shaped risk curve for nutrient-health relations. Topic areas included adaptations in iron homeostasis associated with pregnancy and young childhood, the impact of inflammation, measurement of iron status, current estimates of iron status for pregnant women and young children in the United States and in Europe, and emerging evidence suggesting adverse effects associated with iron supplementation of iron-replete individuals. A crosscutting dialogue conducted at the close of the workshop formed the basis for a workshop summary that specified evidence gaps and research needs in a range of areas centered on the relation of these adaptations of iron homeostasis with the response to and risk from iron supplementation as well as the need for indicators informative of the full continuum of iron status and based on health outcomes, not just erythropoiesis.
Article
During pregnancy, iron needs to increase substantially to support fetoplacental development and maternal adaptation to pregnancy. To meet these iron requirements, both dietary iron absorption and the mobilization of iron from stores increase, a mechanism that is in large part dependent on the iron-regulatory hormone hepcidin. In healthy human pregnancies, maternal hepcidin concentrations are suppressed in the second and third trimesters, thereby facilitating an increased supply of iron into the circulation. The mechanism of maternal hepcidin suppression in pregnancy is unknown, but hepcidin regulation by the known stimuli (i.e., iron, erythropoietic activity, and inflammation) appears to be preserved during pregnancy. Inappropriately increased maternal hepcidin during pregnancy can compromise the iron availability for placental transfer and impair the efficacy of iron supplementation. The role of fetal hepcidin in the regulation of placental iron transfer still remains to be characterized. This review summarizes the current understanding and addresses the gaps in knowledge about gestational changes in hematologic and iron variables and regulatory aspects of maternal, fetal, and placental iron homeostasis.