Kiely 2020

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

Contents lists available at ScienceDirect

Journal of Steroid Biochemistry and Molecular Biology


journal homepage: www.elsevier.com/locate/jsbmb

Vitamin D in pregnancy: Where we are and where we should go T


a,b, c d
M.E. Kiely *, C.L. Wagner , D.E. Roth
a
Cork Centre for Vitamin D and Nutrition Research, School of Food and Nutritional Sciences, University College Cork, Ireland
b
INFANT Research Centre, University College Cork, Ireland
c
Division of Neonatology, Medical University of South Carolina, Charleston, SC 29425, United States
d
Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada

A R T I C LE I N FO A B S T R A C T

Keywords: Vitamin D deficiency has been widely reported among pregnant women and infants around the world. Women
Vitamin D with low sun exposure, high BMI, low vitamin D intakes and socioeconomic disadvantage with poor quality diets
Vitamin D requirements are at greatest risk of vitamin D deficiency, leading to very low serum concentrations of 25-hydroxyvitamin D
Pregnancy (25(OH)D) in their offspring and an increased risk of nutritional rickets. Many observational studies, supported
Perinatal health
by compelling in vitro and in vivo data, have generated evidence suggesting that low vitamin D status in preg-
Vitamin D status
nancy may also contribute to the risk of adverse perinatal outcomes including hypertensive disorders (e.g.,
preeclampsia), fetal growth restriction, and preterm birth. However, the few large randomized controlled trials
(RCTs) conducted to date have generated conflicting evidence for a role of vitamin D supplementation in im-
proving perinatal outcomes. Vitamin D supplementation policies during pregnancy and implementation of po-
licies vary within and between jurisdictions. Regulatory authorities have cited insufficient evidence to establish
pregnancy-specific targets for serum 25(OH)D concentrations or prenatal vitamin D intake that effectively re-
duce the risks of adverse perinatal and infant outcomes. This paper arises from a Debate on Vitamin D
Requirements during Pregnancy, held at the 22nd Vitamin D Workshop, 2019. From varied perspectives, our
objectives were to evaluate the evidence for: vitamin D metabolism in pregnancy and the prevalence of gesta-
tional vitamin D deficiency worldwide; the translation of laboratory research findings to clinical studies on the
role of vitamin D in perinatal health; the challenges of designing and conducting clinical trials to establish
prenatal vitamin D requirements; and results to date of major large RCTs of prenatal vitamin D supplementation.
Lastly, we explored potential next steps towards generating robust clinical data in this field to address both
public health protection and patient care.

1. Introduction for prevention of these adverse events.


The definition of healthy vitamin D status during pregnancy, typi-
Pregnancy and infancy are life-stages for which evidence of low cally indicated by circulating concentrations of 25-hydroxyvitamin D
vitamin D status is widespread but the evidence basis for setting dietary (25(OH)D), is the subject of intense debate, which has been challenged
requirements for vitamin D is weakest [1]. Prevention of low vitamin D by a lack of data from well-powered, well-controlled trials in pregnant
status is required to reduce the risk of nutritional rickets among chil- women, particularly among women with low circulating 25(OH)D at
dren and adolescents and osteomalacia in adults, which can have severe baseline. Multiple systematic reviews of the role of vitamin D supple-
and lasting consequences for bone growth and skeletal integrity [2]. mentation during pregnancy have been published but these have lacked
The recent discovery of non-skeletal roles for vitamin D has changed the data from substantial trials and there is no consensus as to whether
landscape considerably and mechanistic studies have elaborated the physiological requirements for 25(OH)D are higher during pregnancy
molecular roles of vitamin D in many systems, for example, immunity than in non-pregnant adults [7]. It is also worth noting that trials and
[3,4]. Modulation of the intrauterine immune environment by vitamin reviews of trials have framed the question around the benefit of addi-
D has been implicated in adverse perinatal outcomes, including re- tional vitamin D supplementation during pregnancy (using a pharma-
current pregnancy loss, preeclampsia and spontaneous preterm birth cological model of intervention regardless of baseline status) rather
[5,6]. Indications are that early pregnancy may be the critical window than the role of sufficient vitamin D to support healthy pregnancy from the


Corresponding author at: Cork Centre for Vitamin D and Nutrition Research, School of Food and Nutritional Sciences, University College Cork, Ireland.
E-mail address: m.kiely@ucc.ie (M.E. Kiely).

https://doi.org/10.1016/j.jsbmb.2020.105669
Received 8 November 2019; Received in revised form 18 March 2020; Accepted 2 April 2020
Available online 14 April 2020
0960-0760/ © 2020 Elsevier Ltd. All rights reserved.
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

outset. pregnancy cohort of 1800 women in Ireland, where there is no maternal


A Debate on the controversial topic of Vitamin D Requirements vitamin D supplementation policy. Data from CDC-accredited LC/MS-
during Pregnancy was held at the 22nd Vitamin D Workshop, 2019. Our MS analysis of 25(OH)D showed that 17 % were below 30 nmol/L at 15
objectives were to evaluate the current state of the evidence for: weeks of gestation; this was 49 % among women of ethnic minority
[15]. Among the infants of this cohort, 46 % of umbilical cord sera had
• vitamin D metabolism in pregnancy, and the prevalence of gesta- a 25(OH)D < 30 nmol/L; this was 73 % among infants from women of
tional vitamin D deficiency worldwide. ethnic minority [16].
• the translation of laboratory research findings to clinical studies on
the role of vitamin D in perinatal health. 3. Nutritional rickets
• the challenges of designing and conducting clinical trials to establish
prenatal vitamin D requirements; and, In line with the prevalence data for 25(OH)D among mothers and
• results to date of major large RCTs of prenatal vitamin D supple- newborns, it is no surprise to note that the greatest burden of nutri-
mentation. tional rickets is in Africa, Asia and the Middle East [17,18]. There is
also evidence that while the incidence of nutritional rickets is stable or
The three authors hold some shared and some divergent views on decreasing among whites, it is increasing in North America and Europe
the core issues discussed and provided their perspectives on these is- among immigrant populations [19]. Thacher et al. [17] provided a
sues. Our common goal was to explore potential next steps towards summary of nutritional rickets in immigrant and refugee children using
generating robust clinical data in this field to address both public health data collected in the UK, Denmark, Australia, Canada and the US. Po-
protection and patient care. In the presentation of the evidence in this pulations most at risk were South Asian, African and Middle Eastern
paper, we aimed to convey the plurality of divergent viewpoints rather African, with incidence rates (per 1,000,000/year) up to 24 in young
than present a unified or consensus statement. children < 3 years and up to 4.9 among children < 15 years. The stark
contrast between whites and ethnic minorities in western countries is
2. Low vitamin D status is common among pregnant women and highlighted in the Global Consensus Recommendations on Prevention and
infants Management of Nutritional Rickets [2]. Most clinical insights are gained
from hospital admissions; Uday et al. (2018) have documented hypo-
A key question to be addressed is whether the prevalence of 25(OH) calcemic dilated cardiomyopathy associated with nutritional rickets
D < 25−30 and < 50 nmol/L is higher among pregnant women than and in some cases, fatal consequences. These prevalence estimates of
among other population groups. Saraf et al., (2016) estimated the maternal and infant 25(OH)D concentrations and the resulting effects
global prevalence of 25(OH)D concentrations < 50 nmol/L at 54 % elevate vitamin D deficiency among mothers and babies to the status of
among pregnant women and 75 % among newborns. Almost one in five a serious public health problem that requires urgent action.
pregnant women and one in three newborns were below 25 nmol/L,
placing them at increased risk of developing rickets and osteomalacia. A 4. Challenges in defining nutritional requirements for vitamin D
systematic review of 25(OH)D concentrations in low- and middle-in- during pregnancy
come countries (LMIC) by Cashman et al. [8] reported that 29 out of the
83 countries included had 25(OH)D data available. Afghanistan, Paki- To provide the policy context, Mairead Kiely (MK) also summarized
stan, India, Tunisia, Syria, the West Bank and Gaza and Mongolia were current dietary recommendations for vitamin D in in pregnant and
classified as “hot spots” for very low 25(OH)D (< 25−30 nmol/L) lactating women in line with the 25(OH)D concentration targets they
among women, pregnant women or infants on the basis of having a are intended to reach. Individual intake recommendations range from 5
prevalence in excess of 20 % [8]. In low income and middle eastern to 15 μg/day (200–600 IU) to achieve 25(OH)D targets of >
countries in particular, women and girls appear to have lower circu- 25–50 nmol/L (10–20 ng/mL) [20]. Although there is some variation
lating 25(OH)D than their male counterparts, probably due to clothing between these recommendations, they were all formulated based on
practices [9,10]. Palacios and Gonzales (2014) described a high pre- evidence from RCTs of vitamin D status and bone health outcomes. Due
valence (> 20 %) of 25(OH)D < 30 nmol/L among pregnant women to a lack of evidence on which to set nutritional requirements for
and infants, in many countries, including in South Asia and the Middle 25(OH)D during pregnancy specifically, recommendations for vitamin
East; up to 60 % of women in India and 86 % of infants in Iran had D intake are the same for pregnant and lactating women as non-preg-
25(OH)D < 30 nmol/L. nant adults.
One of the challenges of interpreting global 25(OH)D data is the
methodological variability in analytical methods and data reporting, 5. Vitamin D requirements from the perspective of vitamin D
which can be particularly challenging for maternal samples [11]. The biology
relatively low uptake, and low rate reporting of laboratory performance
of users of quality assurance systems, such as DEQAS (Carter et al. Carol Wagner (CW) presented data showing that unlike other times
2017), leads to a lack of transparency. Application of the NIH Vitamin D during the lifespan, pregnancy involves the orchestration of events that
Standardization Protocol (VDSP) should be used to standardize 25(OH) must protect the mother while allowing growth and development of the
D data if serum aliquots are available for re-analysis by LC–MS/MS, fetus. There is a delicate immune balance that nurtures the allogeneic
using a validated statistical approach (Durazo-Arvizu et al.). The CDC fetus, while maintaining reactivity against pathogens to the outside
Vitamin D Standardization-Certification Program (VDSCP) is also world [21–23]. During the first trimester, a proinflammatory state al-
available as an avenue to achieve accreditation of 25(OH)D analysis. lows for the invasion of the uterine wall by fetal derived cytotropho-
Within temperate countries, the prevalence of vitamin D deficiency blast cells to become the placenta. Following the establishment of the
is much higher among persons with darkly pigmented skin tones. Using maternal-placental-embryonic unit, there is rapid growth of the fetus,
VDSP, the ODIN project reported the first internationally standardised which must occur in a state that is by comparison to the earlier first
dataset of vitamin D status and the prevalence of vitamin D deficiency stage of pregnancy, anti-inflammatory [21,22]. This relatively anti-in-
across Europe [12]. While the overall prevalence of 25(OH)D < 30 flammatory state will last well beyond the second trimester and into the
nmol/L was 13 %, about twice that of the US (5.9 %) [13] and Canada third trimester, when the uterine environment becomes more proin-
(7.4 %) [14], persons with more darkly pigmented skin tones were at flammatory to allow the onset of labour and the expulsion of the fetus
much higher risk than their white counterparts both in Europe and from the uterus [21,22]. Dysregulation of these tightly controlled bio-
North America. As part of the ODIN project, Kiely et al. [15] analysed a phenomena at a systemic and placental level have been considered as a

2
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

potential mechanism mediating pathogenesis of recurrent pregnancy increasing from ∼2 mg/day in the first trimester to 100 mg/day in last
loss [24] preeclampsia and spontaneous preterm birth [25,26]. It is trimester that is continued throughout lactation. Yet, immediately after
postulated by many that these adverse events during pregnancy have delivery, maternal calcitriol concentrations return to pre-pregnancy
their origins early-on in pregnancy, with derangement of the critical levels, despite significant calcium requirements needed during lactation
balance in immune function [5,6,27]. [49,50]. 125(OH)2D concentrations by 12 weeks of gestation have al-
125(OH)2D mediates gene expression via VDR and VDREs, and in- ready increased to a mean of 180 pmol/L, further increasing to around
creased concentrations of 125(OH)2D could have implication for gene 300 pmol/L toward the end of pregnancy, compared to nonpregnant
pathways and subsequent function. In mouse models of vitamin D de- control mean concentration of 91 pmol/L [44,51,52]. Calcitriol circu-
ficiency studied early in gestation, an array of genes in the placenta are lates at low levels in fetal blood, typically less than 50 % of the ma-
upregulated and others downregulated based on maternal vitamin D ternal value [53–56]. Both animal models and human data suggest that
status [28], which find corollaries in human studies, especially fol- 25(OH)D readily crosses the placenta, with cord blood concentrations
lowing adverse pregnancy outcomes such as preeclampsia [26,29–32]. that are around 70–100 % of maternal concentrations [11,44,54].
Parallel findings are noted in the upregulation of certain placental Calcitriol synthesis in the fetus is likely suppressed by the high serum
genes that have been implicated in preeclampsia [33]. There are studies calcium, high phosphorus, and low PTH concentrations typically ob-
that describe alternation in T cell phenotype associated with preterm served in cord blood [57].
birth [34] and recurrent pregnancy loss in women who are vitamin D It is interesting that the rise in maternal 125(OH)2D during preg-
deficient [24,35–37]. There are also non-genomic calcitriol-dependent nancy occurs without any effect on serum ionized or corrected calcium
biological effects that can take place in cells, involving second mes- and appears to result from uncoupling of 125(OH)2D from its well-es-
sengers generated by membrane-initiated signalling pathways [38]. tablished actions in the non-pregnant state [44]. While VDBP increases
Classic vitamin D receptor (VDR) and the membrane-associated concomitantly, it does not increase to the same degree as 125(OH)2D
rapid response steroid-binding protein (MARRS) found in the cell [44]. In addition, upregulation of CYP27B1 mRNA in the maternal
membrane may bind 125(OH)2D and initiate the activation of nu- kidneys, exerts characteristics of “extrarenal” production from an en-
merous pathways involving various protein kinases (PKC), MAPK, PKA, docrine standpoint, such as relative unresponsiveness to stimulation by
phosphatidyl inositol phosphate, and Ca2+ and chloride channels [39]. PTH (except under conditions of extreme vitamin D deficiency) and
The VDR and regulatory metabolic enzymes are expressed in both lack of feedback inhibition of CYP27B1, and thus, 1-α-hydroxylase by
placenta and decidua, indicating a potential critical point in the im- 125(OH)2D itself. This increase in 125(OH)2D seems to be independent
munomodulation at the maternal-fetal interface [40]. Further, activities of parathyroid hormone (PTH), which itself remains within the normal
of vitamin D response element (VDRE)-containing genes can be grouped adult range throughout pregnancy [44,58,59]. Other hormones could
in quite diverse biological networks: bone and mineral metabolism; cell also contribute to the bioregulation of 1-α-hydroxylase such as PTH-rP,
life and death (comprising proliferation, differentiation and apoptosis); estradiol, prolactin, and placental lactogen [52].
immune function—both innate and adaptive immunity that are thought As shown in Fig. 1 below (from Hollis et al., with permission, [44],
to be operational during the various stages of pregnancy [41]. this kinetic reaction saturation graph of 25(OH)D and 125(OH)2D
During pregnancy, there are major adaptations in vitamin D meta- shows that 25(OH)D has direct influence on 125(OH)2D concentrations
bolism. After conception, the fertilized egg undergoes a series of re- throughout pregnancy, an event which does not occur during any other
productive cell divisions to form the three germ layers—endoderm, time during human life. It is apparent in the figure that as lower con-
mesoderm, and ectoderm) that give rise to different types of cells, many centrations of 125(OH)2D increase, first order kinetics becomes zero
of which express the vitamin D receptor (VDR) [42]. Various cells give order kinetics, with a plateauing of the graph and an inflection point at
rise to different organs through organogenesis that extends from 4 to 10 100 nmol/L (40 ng/mL) 25(OH)D—the level required to optimize
weeks of gestation with the placental fully formed and functional by 4 125(OH)2D production during pregnancy.
weeks of gestation. During this time of immense activity and growth, Based on these findings, it is thought by some that this inflection
localized concentrations of vitamin D in the form of 25(OH)D (which point of 100 nmol/L may be critical during pregnancy for both maternal
crosses the placenta from the maternal side) and 125(OH)2D (which is and fetal well-being, and that attainment of this threshold early-on may
synthesized by the fetus as early as 6–10 weeks of gestation) can in- be key. There are data to suggest that vitamin D status early in
teract with various signalling systems to regulate organ development
[42]. As mentioned, it is the placenta that permits the transfer of
25(OH)D from the mother to the fetus but also the placenta through its
production of 1α-hydroxylase that local synthesis of 125(OH)2D occurs.
125(OH)2D binds to VDR in specialized cells to induce genomic and
nongenomic responses that stimulate organ development.
Focusing on 125(OH)2D during pregnancy, 125(OH)2D increases
more than 2–3 fold in the first weeks of pregnancy that appears to come
mainly from maternal kidney synthesis but some is placentally-derived
[11,43–45]. This increase is noted in both animal models and human
studies as measured by RIA and LC-Tandem mass spec (LC–MS) and is
accompanied by a rise in VDBP, the main carrier of all vitamin D
moieties with greater avidity for 25(OH)D [23,43,44,11,38,46]. Despite
this, there is no corresponding rise in 25(OH)D during pregnancy [46].
The increase in 125(OH)2D early in the first trimester of pregnancy
is linked to higher Cyp27B1 (1-α-hydroxylase) activity in maternal
kidney, placental trophoblasts and decidua [39,43,44,47]. Longitudinal
human studies also report a progressive increase in both bound and free Fig. 1. Substrate-Product Relationships of Vitamin D Metabolites during
125(OH)2D concentrations [11]. Previous animal studies in partly and Pregnancy. This panel demonstrates the relationship circulating 25(OH)D to
completely nephrectomized rats report that this rise is mainly attrib- control the production of 125(OH)2D during pregnancy. All data points for all
uted to increased maternal synthesis per se, rather than decreased subjects in all groups were included in this analysis. Relationship of 25(OH)D
clearance [48]. This increase in 125(OH)2D occurs at a time during and 125(OH)2D during Pregnancy.
pregnancy before increased fetal calcium requirements manifest, (from [44], Figure 3b Panel b, with permission ([44]).

3
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

pregnancy sets the stage for vitamin D’s enabling effect on immune Table 1
function [26,60]. In their post hoc analysis using multivariable log-bi- Factors Affecting Clinical Outcomes of Vitamin D Supplementation Trials
nomial regression of maternal 25(OH)D status during pregnancy, during Pregnancy.
McDonnell et al. reported that women attaining maternal 25(OH)D
• Timing of intervention: when in gestation did supplementation occur
concentrations ≥100 nmol/L compared to ≤50 nmol/L, adjusted for • Sample size
covariates, was associated with a reduction in the risk of preterm birth • Baseline serum 25(OH)D: e.g., the most profound effects of early supplementation
seen in those with the lowest 25(OH)D concentrations at baseline
by 59 % [61].
• Sunshine exposure
With more modern scientific technological advancement, vitamin D
• Habitual vitamin D and calcium intake
has been shown to act on innate and structural cells to promote anti- • Race/ethnicity
microbial defense through induction of antimicrobial peptides (e.g. • VDBP genotype
cathelicidin, beta-defensins) and autophagy [62,63]. Bridging both in- • Dosing frequency of vitamin D: daily vs. weekly vs. monthly dosing
• Adherence to dosing regimen
nate and adaptive immunity, vitamin D also acts on myeloid dendritic
• Types of assays used to measure 25(OH)D concentration
cells (DC) to promote a tolerogenic antigen presenting cell (APC) phe-
• Type of analysis—Intention-to-treat vs. per protocol
notype [64]. In their recent review, Hawrylowicz and Santos provide • Biomarker used for vitamin D status: Is total circulating 25(OH)D concentration
really the biomarker of what is going on at the cellular level during pregnancy?
compelling evidence that vitamin D reduces naïve T cell expansion, and
Should free 25(OH)D be measured? This is a question that comes up frequently.
downregulates Th1 and Th17 responses, but has variable effects on Th2
responses linked to allergic disease [65]. These interactions result in an
increased frequency of human Foxp3 and IL-10 Treg in vitro and in vivo
in question starts at 0 in both the control and treatment group(s); yet, in
[65–71]. Similarly, Vasiliou et al., in their human dust mite-allergy
nutrient studies such as vitamin D supplementation trials, no one starts
mouse model, showed that vitamin D deficiency in utero led to an in-
at a concentration of zero. For this reason, Heaney outlined the criteria
creased frequency of Th2 cells and reduced anti-inflammatory IL-10+ T
that should be met for all nutrient studies to avoid this faulty design
cells in the lungs [72], arguing for an in utero process that continues
[97]. The other question that continues to plague vitamin D studies is
after birth. As noted by Hawrylowicz and Santos [65], neonatal and
what is really representative of that nutrient—in this case, vitamin D? Is
adult immunity differs, and assumptions of how vitamin D promotes
it the parent compound or a metabolite—25(OH)D, and is that meta-
protective immunity in pregnancy and neonates may be distinct to ef-
bolite really indicative of what is going on at the cellular level? Should
fects in adults, as they note that in recently published, independent
the metabolite be measured in its bound form or its free state [11,98]?
vitamin D supplementation pregnancy studies that showed such dif-
Best et al. provide a longitudinal examination of the changes in
ferences (e.g. no correlation with Foxp3) [73–75]. Clearly, more re-
various moieties of vitamin D during adolescent pregnancy and in-
search is needed in this area to inform the design of more meaningful
cluded in their analyses free circulating 25(OH)D measurement as well
clinical trials.
as VDBP [11]. In their analysis of 58 adolescent pregnant females, the
authors found that regardless of race or visit, total 25(OH)D was a
6. Translation of laboratory findings to clinical studies
stronger correlate of PTH, 125(OH)2D, and of the catabolite
2425(OH)2D than free circulating 25(OH)D, and neither total nor free
[76]) concluded that notwithstanding inconsistent reporting of ad-
25(OH)D was related to serum calcium. While African American
verse events, vitamin D supplementation appears safe. Studies pre-
pregnant girls had lower total 25(OH)D (p < 0.0001), their free
sented by CW also showed a high degree of safety in both mother and
25(OH)D did not significantly differ by race (p = 0.2). Of interest, in
fetus/neonate and later, in the offspring during longitudinal follow-up
these pregnant adolescents, VDBP concentration was elevated and in-
studies [44,73,77–87]. During pregnancy, there was no evidence of
versely associated with the percent free 25(OH)D, but measured free
toxicity from high 25(OH)D concentrations or differences in maternal
25(OH)D provided no advantage over total 25(OH)D as a predictor of
serum calcium, phosphorus, and urinary calcium/creatinine ratios or
PTH, 125(OH)2D, 2425(OH)2D, or calcium. Thus, it appears for the
adverse pregnancy outcomes among the many women who have been
moment that total circulating 25(OH)D concentration is the best in-
supplemented with various combinations of vitamin D up to 110 μg/
dicator of vitamin D status.
day or 1250 μg/week during pregnancy. There are some individuals,
Other issues in assessing vitamin D status during pregnancy and its
however, who could develop toxicity if not previously diagnosed with
relevance to outcomes includes the question of whether baseline
idiopathic hypercalcemia of childhood associated with Williams Syn-
25(OH)D in a study represents the 25(OH)D concentration circulating
drome (a rare genetic disorder that results in hypercalcemia with even
around the time of conception. Vitamin D supplementation as a treat-
modest vitamin D supplementation due to abnormal metabolic pro-
ment is plagued with nonadherence whereas using 25(OH)D as the
cessing [88] or from other metabolic genetic defects in vitamin D
biomarker is not. There are also issues of whether or not total circu-
processing such as CYP24A1 mutations [89]. Some studies show benefit
lating 25(OH)D is reflective of vitamin D metabolism within the cell,
to the pregnant woman and her developing fetus with improved vi-
especially with variable bolus dosing. Perhaps it is baseline 25(OH)D in
tamin D status [90] yet others do not [84], but none have shown
early pregnancy or even preconception that is more relevant in showing
toxicity with higher dosing within certain inclusion and exclusion entry
the effect of vitamin D status on the fetal and maternal health, but to
criteria. Despite the known basic science aspects of vitamin D, clinical
date, no trials have addressed circulating 25(OH)D at the time of con-
trials often fail to note differences in outcomes of supplementation
ception and placental activity.
during pregnancy due to a number of factors listed in Table 1.
Systematic reviews and meta-analyses of vitamin D supplementa-
Based on animal and clinical studies looking at the effect of vitamin
tion during pregnancy show mixed results in prevention of pre-
D status at baseline early in pregnancy and pregnancy outcomes
eclampsia and preterm birth, which may reflect timing of supple-
[26,91–94], it would appear that the most profound effects of early
mentation initiation as well as study design in terms of dosing regimen
supplementation are seen in those with the greatest deficiency states at
and maternal vitamin D status starting point (as noted in Table 1). The
baseline [90,95]. In addition, it would appear that the effects are seen
differences between in vitro and animal studies results of vitamin D’s
more notably when total circulating 25(OH)D is used as the biomarker
role as an enabler and the often-null results of randomized controlled
of vitamin D status during pregnancy rather than analysed on an in-
trials challenges us to explore alternative hypotheses and to design
tention-to-treat (ITT) basis. Higher dose effects are more apparent in
studies that integrate these divergent findings.
those who follow the prescribed supplementation regimen and when
Another factor that could potentially affect study results includes
the analysis is on a per protocol basis [96]. Another consideration is that
estimation of sample size. On what do we base the sample size of a
in a classic drug study with ITT analysis, the concentration of the drug

4
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

clinical trial? Is it anticipated treatment effects such as preeclampsia, so the overall inference is that prenatal vitamin D supplementation
hypertension, gestational diabetes, or changes in a particular relevant safely raises 25(OH)D without clearly providing other benefits to
biomarker such as T cell phenotype? Another question is when in pregnant women and babies.
pregnancy do you look for effect? What biomarker is most appropriate? However, further efforts to unpack the results of some of these trials
Should sample size be based on VDBP genotype, specific SNPs or eth- have led to alternative interpretations based on sub-group effects, post-
nicity, all of which affect vitamin D metabolism? These questions im- hoc secondary analyses, and longer-term follow-up studies. For ex-
pact study design but also data analysis. Such questions influence ample, in their original trial report, Hollis et al. [44] did not find any
clinical study design and create limitations in our ability to interpret the significant differences in maternal and neonatal clinical outcomes [44];
data in a meaningful manner. As has been the case with the advent of however, in a post-hoc analysis adjusting for race, published in 2013,
scientific inquiry, the basis for what we prescribe clinically must have a Hollis and Wagner wrote that, “the data from our RCT strongly suggest
demonstrated rationale that befits the data and the laboratory, and that vitamin D supplementation during pregnancy can significantly
which can be translated to bedside practices with continued meaning decrease complications of pregnancy including primary caesarean sec-
and relevance. Without study design that incorporates the nature of the tion (p = 0.046), hypertensive disorders of pregnancy (p = 0.05) and
system—in this case a hormonal system that has many functions—we comorbidities of pregnancy (p = 0.03)” [102]. This particular inter-
will fail to see benefit and make conclusions that might be premature or pretation of the trial continues to be cited as direct evidence of the
biased. Through careful understanding of cellular mechanisms that may clinical benefits of high-dose prenatal vitamin D supplementation
be influencing its demonstrated effect and a broader systems-based [103]. In the primary report of the MAVIDOS trial, a planned sub-group
approach across mother, placenta and fetus, we will move closer to a analysis indicated that vitamin D supplementation increased bone mi-
greater understanding of the dynamic effect of vitamin D during preg- neral content in newborns delivered in the winter, in contrast to the
nancy on both the mother and her developing fetus. overall null findings of the trial [101]. Yet, conference presentations,
press releases and news stories about MAVIDOS frequently highlighted
7. Alternative perspective based on evidence from published RCTs this sub-group effect and emphasized the potential benefits of vitamin D
of prenatal vitamin D supplementation [104]. Two of the five major trials (VDAART and COPSAC) had similar
primary outcomes related to early-childhood wheeze and asthma, and
While acknowledging the well-justified enthusiasm for the potential the pooled analysis of these trials indicated that vitamin D significantly
benefits of improving vitamin D status in the prenatal period, Dan Roth reduced the incidence of asthma or recurrent wheeze in the offspring up
(DR) presented a summary of selected high-quality RCTs showing in- to three years of age [105]. However, follow-up of the children in one of
sufficiently convincing evidence to merit widespread changes to clinical the trials showed no sustained effects, such that the prevalence of
practice guidelines or policy [9]. As of the end of 2019, the World asthma diagnoses by age 6 years was similar between the vitamin D and
Health Organization (WHO) advises that, “vitamin D supplementation control groups [106].
is not recommended for pregnant women to improve maternal and Researchers and practitioners who conduct and interpret vitamin D
perinatal outcomes” [99]. While there is little doubt that prenatal vi- RCTs are faced with a dilemma when the primary results of a trial
tamin D supplementation improves maternal and fetal vitamin D status conflict with – or are less convincing than – other post-hoc or alternative
in a dose-dependent manner, the clinical benefits to the mother and analyses of the same data, especially if the latter analyses support their
baby remain a subject of debate [7,76,100]. The question is whether original hypothesis. Retaining the original randomized design, in-
additional vitamin D during pregnancy, over and above that required by cluding all participants in the analyses, and applying an intent-to-treat
a non-pregnant woman, is required. To illustrate how prenatal vitamin approach are all strategies that serve to reduce biases, but there is often
D RCTs have so far failed to provide robust evidence of clinical benefits, an appeal to dissect the data in other ways that are believed to reveal
it is instructive to consider five published blinded RCTs that each in- hidden truths about physiology that may be obscured by the bluntness
cluded at least 500 enrolled women, were generally of high-quality, and of the RCT approach (Fig. 2). Conventional analysis of a RCT is by no
for which publication in reputable journals suggests they underwent means a sure-fire path to clarity, and no trial is without its limitations.
thorough peer review (Table 2). None of these five trials individually For example, the placebo-controlled dose-ranging RCT of prenatal vi-
demonstrated a beneficial effect of the vitamin D intervention on their tamin D in Bangladesh from Roth and colleagues was specifically de-
primary outcomes, as reported in the original trial publications signed to test whether vitamin D improves infant growth in a setting
(Table 3). For other secondary outcomes of interest (e.g., birth weight, where stunting is common [84]. It was not designed or powered to
gestational age at birth or preterm, neonatal or infant hospitalization, examine effects on uncommon maternal or perinatal outcomes, and it is
or gestational hypertension or preeclampsia), none of these trials pro- possible that the initiation of supplementation in the 2nd trimester was
vided strong evidence of benefits of vitamin D (i.e., either the outcome too late to affect some outcomes such as preeclampsia [84]. Other as-
was reported and the effect was null or non-significant, or the outcome pects of the trial may have limited its generalizability to the broader
was not reported) [44,80,82,84,101]. Importantly, these trials reported population of Bangladeshi women (e.g., perhaps trial participants were
adverse event monitoring and none have raised major safety concerns, healthier than average) or to populations outside of Bangladesh (e.g.,

Table 2
Characteristics and primary findings of five randomized controlled trials of prenatal vitamin D supplementationa.
Hollis 2011 [44] Litonjua 2016 (VDAART) Cooper 2016 Chawes 2016 (COPSAC) Roth 2018 (MDIG) [84]
[82] (MAVIDOS) [101] [80]

Country USA USA UK Denmark Bangladesh


Total N randomized 502 881 1134 623 1300
Highest vitamin D dose 4000 IU/day 4400 IU/day 1000 IU/day 2800 IU/day 4000 IU/day (given as
28,000 IU/week)
Control group vitamin D dose 400 IU/day 400 IU/day Placebo 400 IU/day Placebo
Primary outcome measure Serum 25(OH)D Early-childhood asthma or Neonatal bone mass Early-childhood Infant length (at one year of
concentration recurrent wheeze persistent wheeze age)
Effect of vitamin D on primary Strong positive effect No significant effect No significant effect No significant effect No significant effect
outcome

a
Trials selected if they were double-blind and enrolled > 500 participants.

5
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

Fig. 2. Schematic representation of the ad-


vantages and disadvantages of approaches
available to investigators when analyzing data
from a randomized controlled trial. The hor-
izontal axis reflects the spectrum of choices
related to the participant selection criteria. For
example, a discrete sub-group analysis may
involve restriction to participants with baseline
25(OH)D < 30 nmol/L, whereas population-
representative sampling would include all
participants irrespective of baseline character-
istics, adherence, or other non-random factors.
The vertical axis reflects the spectrum of
choices related to the assignment of the ex-
posure variable. An intention-to-treat approach
entails direct comparisons of the experimental
versus control groups, relying only on the
random assignment regardless of adherence or
completeness of follow-up; in contrast, ob-
servational analyses usually ignore the random
allocation and instead examine the association
between an outcome and a non-random in-
dicator of vitamin D status (e.g., 25(OH)D or
number of vitamin D doses received).

perhaps the trial participants’ concurrent nutritional deficiencies con- cannot provide evidence of causal effects [112–114]. In a recent article,
strained the effects of vitamin D). Hollis recently wrote that, “Most of these clinical trials have validated
Unfortunately, systematic reviews and meta-analyses of RCTs can the positive effects of prenatal vitamin D on birth outcomes …,” [103]
further confuse rather than clarify the state of the evidence, as their citing four published articles to support his claim: 1) an Iranian study
conclusions can only be as robust as the underlying evidence on which that reported a lower frequency of preeclampsia, preterm delivery and
they are based [7]. For example, authors of the recent update of the adverse pregnancy outcomes at a hospital that conducted screening for
Cochrane Collaboration systematic review of vitamin D in pregnancy vitamin D deficiency and provided supplementation, compared to a
had to rely on mostly small trials of low or questionable quality to draw control hospital in another city [90] (limitations of this study were
conclusions about the effects of vitamin D (versus placebo) on maternal highlighted in a letter to the editor [115]); 2) a secondary analysis of
and neonatal outcomes [100]. In summarizing one of the meta-ana- VDAART that examined asthma/wheeze outcomes but did not report on
lyses, Palacios et al. wrote that, “supplementation with vitamin D any birth outcomes [83]; 3) another secondary analysis of VDAART that
probably reduces the risk of pre-eclampsia compared to no intervention found there was no effect of vitamin D supplementation on pre-
or placebo (risk ratio (RR 0.48, 95 % CI 0.30 to 0.79)” and they con- eclampsia risk based on the RCT design, but did find an association
sidered this to be based on “moderate-certainty evidence” [100]. This between 25(OH)D and preeclampsia using a cohort analysis [26]; and,
conclusion is reiterated prominently in the abstract and plain-language 4) an observational study of the association between 25(OH)D and
summary, and if true, would be of considerable public health im- preterm birth [61]. There are certainly many studies suggesting that
portance. However, scrutiny of the four trials on which this finding was prenatal vitamin D supplementation may have benefits, including some
based reveals the weakness of the conclusion. The aggregated sample small RCTs; however, an unbiased assessment must take stock of the
size was only 499 women [100], far lower than what would be required entire body of evidence, taking into account study design and quality.
in a primary prevention trial designed for precise estimation of effects
of a prenatal intervention on preeclampsia. Only one of the four trials
reported having any type of prospective ascertainment protocol and 8. A pragmatic approach - setting the maternal vitamin D
standardized case definition for preeclampsia [107], and all of the requirement to safeguard the fetal skeleton by targeting threshold
published trial reports lack sufficient information to confidently es- 25-hydroxyvitamin D concentrations
tablish the reliability of their findings [107–110]. Furthermore, reviews
that incorporated other trials (including those in which control groups MK proposed the possibility that given the various determinants of
received a low-dose of vitamin D) do not support a beneficial effect of perinatal outcomes, including personal, genetic and healthcare factors,
additional vitamin D for prevention of preeclampsia, at least not be- as well as overall diet and nutrition, it may be a too tall order to expect
yond the amount in conventional prenatal multiple micronutrient a single nutrient intervention to deliver dramatically improved out-
supplements (5–15 μg/day) [7,76]. comes. This may be true both in low resource settings, where mal-
To date, there have not been any rigorous and adequately powered nutrition is common, or where there are no maternal or neonatal sup-
RCTs of prenatal vitamin D supplementation for the prevention of hy- plementation policies, but also in countries with well-nourished,
pertensive diseases of pregnancy or other adverse maternal outcomes healthy mothers who have 25(OH)D status at baseline in excess of
[7,76,100]. Authors of reviews or commentaries may conclude that 50 nmol/L and excellent obstetric care. Irrespective of the potential
vitamin D prevents pregnancy complications if their meta-analysis was benefits of additional 25(OH)D to enable the establishment and main-
methodologically flawed (e.g., biased study selection, errors in data tenance of a healthy pregnancy, the complete dependence of the fetus
abstraction) [111] or focused entirely on observational studies that and newborn infant on maternal 25(OH)D concentrations has been
consistently reported [116]. It is widely accepted that at minimum,

6
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

very low 25(OH)D (< 25−30 nmol/L) during pregnancy should be logistically challenging, but should be strongly considered by an in-
prevented to protect fetal skeletal development [2]. As cord 25(OH)D ternational consortium of researchers and funders.
concentration is usually lower than the corresponding maternal con-
centration [116] and others, prevention of maternal 25(OH)D below References
25−30 nmol/L may not provide fetal and neonatal protection [15].
Current recommendations for vitamin D during pregnancy do not [1] Institute Of Medicine, Dietary Reference Intakes for Calcium and Vitamin D,
National Academies Press, Washington (DC), 2011.
consider the vitamin D requirement of newborn infants and assume the
[2] C.F. Munns, N. Shaw, M. Kiely, B.L. Specker, T.D. Thacher, K. Ozono,
immediate availability of an adequate supply from early life. Therefore, T. Michigami, D. Tiosano, M.Z. Mughal, O. Mäkitie, L. Ramos-Abad, L. Ward,
Kiely et al. [20] proposed prevention of neonatal vitamin D status < L.A. Dimeglio, N. Atapattu, H. Cassinelli, C. Braegger, J.M. Pettifor, A. Seth,
H.W. Idris, V. Bhatia, J. Fu, G. Goldberg, L. Sävendahl, R. Khadgawat,
25−30 nmol/L, indicated by umbilical cord 25(OH)D, as a potential P. Pludowski, J. Maddock, E. Hyppönen, A. Oduwole, E. Frew, M. Aguiar,
target for defining maternal vitamin D requirements during pregnancy, T. Tulchinsky, G. Butler, W. Högler, Global consensus recommendations on pre-
consistent with prevention of nutritional rickets [2]. In a placebo-con- vention and management of nutritional rickets, J. Clin. Endocrinol. Metab. 101
(2016) 394–415.
trolled dose-response RCT, O’Callaghan et al. [117] reported that [3] J.S. Adams, M. Hewison, Unexpected actions of vitamin D: new perspectives on
maintaining maternal 25(OH)D concentrations > 50 nmol/L at the the regulation of innate and adaptive immunity, Nat. Clin. Pract. Endocrinol.
Metab. 4 (2008) 80–90.
end of pregnancy ensured that cord sera were all > 25 nmol/L. The [4] R.F. Chun, P.T. Liu, R.L. Modlin, J.S. Adams, M. Hewison, Impact of vitamin D on
vitamin D3 intake required to maintain maternal 25(OH)D > 50 nmol/L immune function: lessons learned from genome-wide analysis, Front. Physiol. 5
in almost all mothers was 30 μg/day (1200 IU) [117], which is twice (2014) 151 151.
[5] N.Q. Liu, M. Hewison, Vitamin D, the placenta and pregnancy, Arch. Biochem.
the current US and EU recommendation of 600 IU. This study confirmed Biophys. 523 (2012) 37–47.
suggestions from two RCTs in Canada [118] and New Zealand [119], [6] J.A. Tamblyn, M. Hewison, C.L. Wagner, J.N. Bulmer, M.D. Kilby, Immunological
that although 10 μg vitamin D per day may prevent maternal 25(OH)D role of vitamin D at the maternal-fetal interface, J. Endocrinol. 224 (2015)
R107–21.
falling below 30 nmol/L, a minimum of 25 μg/d might be needed to [7] D.E. Roth, M. Leung, E. Mesfin, H. Qamar, J. Watterworth, E. Papp, Vitamin D
maintain newborn 25(OH)D at this concentration. Further similar dose- supplementation during pregnancy: state of the evidence from a systematic review
of randomised trials, BMJ 359 (2017) j5237.
response studies are required in different racial and ethnic groups and [8] K.D. Cashman, T. Sheehy, C.M. O’neill, Is vitamin D deficiency a public health
in various settings to generate an inclusive estimate of maternal vitamin concern for low middle income countries? A systematic literature review, Eur. J.
D requirements for the prevention of neonatal deficiency. Adequate Nutr. 58 (2019) 433–453.
[9] D.E. Roth, S.A. Abrams, J. Aloia, G. Bergeron, M.W. Bourassa, K.H. Brown,
consideration of calcium intake is required in these studies. Calcium is M.S. Calvo, K.D. Cashman, G. Combs, L.M. De-Regil, M.E. Jefferds, K.S. Jones,
infrequently considered in RCTs of vitamin D and perinatal health al- H. Kapner, A.R. Martineau, L.M. Neufeld, R.L. Schleicher, T.D. Thacher,
S.J. Whiting, Global prevalence and disease burden of vitamin D deficiency: a
though its role in the calcium metabolic system and maintenance of
roadmap for action in low- and middle-income countries, Ann. N. Y. Acad. Sci.
normal PTH concentrations is linked to perinatal health [120,121]. 1430 (2018) 44–79.
[10] P. Lips, K.D. Cashman, C. Lamberg-Allardt, H.A. Bischoff-Ferrari, B.R. Obermayer-
Pietsch, M. Bianchi, J. Stepan, G. El-Hajj Fuleihan, R. Bouillon, MANAGEMENT OF
9. Conclusions ENDOCRINE DISEASE: current vitamin D status in European and Middle East
countries and strategies to prevent vitamin D deficiency; a position statement of
Laboratory science is driving our understanding of the role of vi- the European Calcified Tissue Society, Eur. J. Endocrinol. (2019).
[11] C.M. Best, E.K. Pressman, R.A. Queenan, E. Cooper, K.O. O’brien, Longitudinal
tamin D in healthy pregnancy, most recently from the perspective of changes in serum vitamin D binding protein and free 25-hydroxyvitamin D in a
immune function. At a fundamental level, the role of vitamin D in multiracial cohort of pregnant adolescents, J. Steroid Biochem. Mol. Biol. 186
(2019) 79–88.
skeletal development has been accepted for many years, but the parti- [12] K.D. Cashman, K.G. Dowling, Z. Škrabáková, M. Gonzalez-Gross, J. Valtueña, S. De
cular needs of pregnant women and newborn infants, a high risk sector Henauw, L. Moreno, C.T. Damsgaard, K.F. Michaelsen, C. Mølgaard, R. Jorde,
of the population, have been underserved. While clinical practice G. Grimnes, G. Moschonis, C. Mavrogianni, Y. Manios, M. Thamm, G.B. Mensink,
M. Rabenberg, M.A. Busch, L. Cox, S. Meadows, G. Goldberg, A. Prentice,
guidelines are potentially more responsive to new evidence, public J.M. Dekker, G. Nijpels, S. Pilz, K.M. Swart, N.M. Van Schoor, P. Lips,
health nutrition recommendations are infrequently updated, creating a G. Eiriksdottir, V. Gudnason, M.F. Cotch, S. Koskinen, C. Lamberg-Allardt,
lengthy gap between the clinical research evidence base and its trans- R.A. Durazo-Arvizu, C.T. Sempos, M. Kiely, Vitamin D deficiency in Europe:
pandemic? Am. J. Clin. Nutr. 103 (2016) 1033–1044.
lation into guidance. National/international policy implementation is [13] R.L. Schleicher, M.R. Sternberg, A.C. Looker, E.A. Yetley, D.A. Lacher,
often slow or non-existent, contributing to a wide dichotomy between C.T. Sempos, C.L. Taylor, R.A. Durazo-Arvizu, K.L. Maw, M. Chaudhary-Webb,
C.L. Johnson, C.M. Pfeiffer, National estimates of serum total 25-hydroxyvitamin
the state of the art and public policy, manifested in gross inequalities in D and metabolite concentrations measured by liquid chromatography-tandem
health care provision and poor outcomes for at risk mothers and babies. mass spectrometry in the US population during 2007-2010, J. Nutr. 146 (2016)
This is evidenced by the wide variation in vitamin D status and rates of 1051–1061.
[14] K. Sarafin, R. Durazo-Arvizu, L. Tian, K.W. Phinney, S. Tai, J.E. Camara, J. Merkel,
nutritional rickets among at-risk groups within and between countries. E. Green, C.T. Sempos, S.P. Brooks, Standardizing 25-hydroxyvitamin d values
In the face of such uncertainty, where do we go from here? Even from the canadian health measures survey, Am. J. Clin. Nutr. 102 (2015)
1044–1050.
without strong evidence from RCTs, it would be reasonable and safe for
[15] M.E. Kiely, J.Y. Zhang, M. Kinsella, A.S. Khashan, L.C. Kenny, Vitamin D status is
public health authorities to take a population approach that aims to associated with uteroplacental dysfunction indicated by pre-eclampsia and small-
eliminate the prevalence of 25(OH)D < 30 nmol/L and facilitate for-gestational-age birth in a large prospective pregnancy cohort in Ireland with
low vitamin D status, Am. J. Clin. Nutr. 104 (2016) 354–361.
achievement of a personal target of 50 nmol/L, including among [16] M. Kiely, S.M. O’donovan, L.C. Kenny, J.O. Hourihane, A.D. Irvine, D.M. Murray,
women of reproductive age [9,20]. In this scenario, fortification of Vitamin D metabolite concentrations in umbilical cord blood serum and associa-
staple foods with vitamin D [9] and scale-up of use of antenatal mul- tions with clinical characteristics in a large prospective mother-infant cohort in
Ireland, J. Steroid Biochem. Mol. Biol. 167 (2017) 162–168.
tiple micronutrient supplements [122] (which include modest amounts [17] T.D. Thacher, P.R. Fischer, M.A. Strand, J.M. Pettifor, Nutritional rickets around
of vitamin D) will likely achieve this aim and reduce the risk of severe the world: causes and future directions, Ann. Trop. Paediatr. 26 (2006) 1–16.
[18] A. Prentice, Nutritional rickets around the world, J. Steroid Biochem. Mol. Biol.
vitamin D deficiency. 136 (2013) 201–206.
To determine if higher intakes of vitamin D are required in preg- [19] A.L. Creo, T.D. Thacher, J.M. Pettifor, M.A. Strand, P.R. Fischer, Nutritional
nancy, the optimal strategy would be to conduct a large-scale RCT of rickets around the world: an update, Paediatr. Int. Child Health 37 (2017) 84–98.
[20] M. Kiely, A. Hemmingway, K.M. O’callaghan, Vitamin D in pregnancy: current
prenatal vitamin D supplementation that goes beyond the scope of any perspectives and future directions, Ther. Adv. Musculoskelet. Dis. 9 (2017)
previous or ongoing RCT: multi-country collaboration (including both 145–154.
high- and lower-income settings), early first trimester (or preconcep- [21] K. Racicot, J.Y. Kwon, P. Aldo, M. Silasi, G. Mor, Understanding the complexity of
the immune system during pregnancy, Am. J. Reprod. Immunol. 72 (2014)
tion) enrolment, factorial design with calcium, and designed and 107–116.
powered for uncommon maternal outcomes (preeclampsia, intrauterine [22] G. Mor, I. Cardenas, The immune system in pregnancy: a unique complexity, Am.
J. Reprod. Immunol. 63 (2010) 425–433.
death), preterm birth, and longer-term child health outcomes (in- [23] A. Vijayendra Chary, R. Hemalatha, M. Seshacharyulu, M. Vasudeva Murali,
cluding nutritional rickets). Such a trial would be financially costly and

7
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

D. Jayaprakash, B. Dinesh Kumar, Reprint of "Vitamin D deficiency in pregnant 693–701.


women impairs regulatory T cell function", J. Steroid Biochem. Mol. Biol. 148 [51] P.D. Papapetrou, The interrelationship of serum 1,25-dihydroxyvitamin D, 25-
(2015) 194–201. hydroxyvitamin D and 24,25-dihydroxyvitamin D in pregnancy at term: a meta-
[24] J. Ji, H. Zhai, H. Zhou, S. Song, G. Mor, A. Liao, The role and mechanism of analysis, Hormones Athens (Athens) 9 (2010) 136–144.
vitamin D-mediated regulation of Treg/Th17 balance in recurrent pregnancy loss, [52] M.S. Ardawi, H.A. Nasrat, B.A.A. Hs, Calcium-regulating hormones and para-
Am. J. Reprod. Immunol. 81 (2019) e13112. thyroid hormone-related peptide in normal human pregnancy and postpartum: a
[25] S. Weiss, H. Wolsk, H. Mirzakhani, B. Hollis, V. Carey, A. Litonjua, F.T.V.T. Group, longitudinal study, Eur. J. Endocrinol. 137 (1997) 402–409.
Asthma/Wheeze and preeclampsia outcomes in the VDAART trial: the influcence [53] A.R. Fleischman, J.F. Rosen, J. Cole, C.M. Smith, H.F. Deluca, Maternal and Fetal
of baseline and treatment levels of vitamin D on treatment response, The Vitamin Serum 1,25- dihydroxy vitamin D levels at term, J. Pediatr. 97 (1980) 640–642.
D Workshop, March 29-31, (2016) 2016 Boston, MA www.vitamindworkshop.org. [54] B.W. Hollis, W.B. Pittard, Evaluation of the total fetomaternal vitamin D re-
[26] H. Mirzakhani, A.A. Litonjua, T.F. Mcelrath, G. O’connor, A. Lee-Parritz, lationships at term: evidence for racial differences, J Clin Endorcrinol Metab 59
R. Iverson, G. Macones, R.C. Strunk, L.B. Bacharier, R. Zeiger, B.W. Hollis, (1984) 652–657.
D.E. Handy, A. Sharma, N. Laranjo, V. Carey, W. Qiu, M. Santolini, S. Liu, [55] K. Seki, K. Furuya, N. Makimura, C. Mitsui, J. Hirata, I. Nagata, Cord blood levels
D. Chhabra, D.A. Enquobahrie, M.A. Williams, J. Loscalzo, S.T. Weiss, Early of calcium-regulating hormones and osteocalcin in premature infants, J. Perinat.
pregnancy vitamin D status and risk of preeclampsia, J. Clin. Invest. 126 (2016) Med. 22 (1994) 189–194.
4702–4715. [56] L.S. Hillman, S. Salmons, S. Dokoh, Serum 1,25-dihydroxyvitamin D concentra-
[27] B.W. Hollis, C.L. Wagner, Vitamin D supplementation during pregnancy: im- tions in premature infants: preliminary results, Calcif. Tissue Int. 37 (1985)
provements in birth outcomes and complications through direct genomic altera- 223–227.
tion, Mol. Cell. Endocrinol. 453 (2017) 113–130. [57] C.S. Kovacs, Bone development and mineral homeostasis in the fetus and neonate:
[28] N.Q. Liu, A.T. Kaplan, V. Lagishetty, Y.B. Ouyang, Y. Ouyang, C.F. Simmons, roles of the calciotropic and phosphotropic hormones, Physiol. Rev. 94 (2014)
O. Equils, M. Hewison, Vitamin D and the regulation of placental inflammation, J. 1143–1218.
Immunol. 186 (2011) 5968–5974. [58] C.L. Wagner, B.W. Hollis, Beyond PTH: assessing vitamin D status during early
[29] C.E. Powe, E.W. Seely, S. Rana, I. Bhan, J. Ecker, S.A. Karumanchi, R. Thadhani, pregnancy, Clin. Endocrinol. (Oxf) 75 (2011) 285–286.
First trimester vitamin d, vitamin d binding protein, and subsequent preeclampsia, [59] J.E. Haddow, L.M. Neveux, G.E. Palomaki, G. Lambert-Messerlian, J.A. Canick,
Hypertension, HYPERTENSIONAHA 110 (2010) 158238. D.G. Grenache, J. Lu, The relationship between PTH and 25-Hydroxy vitamin d
[30] C.J. Robinson, M.C. Alanis, C.L. Wagner, B.W. Hollis, D.D. Johnson, Plasma 25- early in pregnancy, Clin Endocrinol (Oxf) (2011).
hydroxyvitamin D levels in early-onset severe preeclampsia, Am. J. Obstet. [60] S.T. Weiss, A.A. Litonjua, Vitamin d in host defense: implications for future re-
Gynecol. 203 (366) (2010) e1–6. search, Am. J. Respir. Cell Mol. Biol. 56 (2017) 692–693.
[31] A.M. Baker, S. Haeri, C.A. Camargo Jr., J.A. Espinola, A.M. Stuebe, A nested case- [61] S.L. Mcdonnell, K.A. Baggerly, C.A. Baggerly, J.L. Aliano, C.B. French,
control study of midgestation vitamin D deficiency and risk of severe pre- L.L. Baggerly, M.D. Ebeling, C.S. Rittenberg, C.G. Goodier, J.F. Mateus Nino,
eclampsia, J. Clin. Endocrinol. Metab. 95 (2010) 5105–5109. R.J. Wineland, R.B. Newman, B.W. Hollis, C.L. Wagner, Maternal 25(OH)D con-
[32] L.M. Bodnar, H.N. Simhan, J.M. Catov, J.M. Roberts, R.W. Platt, J.C. Diesel, centrations > /=40 ng/mL associated with 60% lower preterm birth risk among
M.A. Klebanoff, Maternal vitamin D status and the risk of mild and severe pre- general obstetrical patients at an urban medical center, PLoS One 12 (2017)
eclampsia, Epidemiology 25 (2014) 207–214. e0180483.
[33] E.V. Schulz, L. Cruze, W. Wei, J. Gehris, C.L. Wagner, Maternal vitamin D suffi- [62] P.T. Liu, S. Stenger, H. Li, L. Wenzel, B.H. Tan, S.R. Krutzik, M.T. Ochoa,
ciency and reduced placental gene expression in angiogenic biomarkers related to J. Schauber, K. Wu, C. Meinken, D.L. Kamen, M. Wagner, R. Bals, A. Steinmeyer,
comorbidities of pregnancy, J. Steroid Biochem. Mol. Biol. (2017). U. Zugel, R.L. Gallo, D. Eisenberg, M. Hewison, B.W. Hollis, J.S. Adams,
[34] A.M. Zahran, K.M. Zharan, H.F. Hetta, Significant correlation between regulatory B.R. Bloom, R.L. Modlin, Toll-like receptor triggering of a vitamin D-mediated
T cells and vitamin D status in term and preterm labor, J. Reprod. Immunol. 129 human antimicrobial response, Science 311 (2006) 1770–1773.
(2018) 15–22. [63] R.F. Chun, P.T. Liu, R.L. Modlin, J.S. Adams, M. Hewison, Impact of vitamin D on
[35] J. Kwak-Kim, A. Skariah, L. Wu, D. Salazar, N. Sung, K. Ota, Humoral and cellular immune function: lessons learned from genome-wide analysis, Front. Physiol. 5
autoimmunity in women with recurrent pregnancy losses and repeated im- (2014) 151.
plantation failures: a possible role of vitamin D, Autoimmun. Rev. 15 (2016) [64] G. Penna, S. Amuchastegui, N. Giarratana, K.C. Daniel, M. Vulcano, S. Sozzani,
943–947. L. Adorini, 1,25-dihydroxyvitamin D3 selectively modulates tolerogenic properties
[36] L. Wu, J. Kwak-Kim, R. Zhang, Q. Li, F.T. Lu, Y. Zhang, H.Y. Wang, L.W. Zhong, in myeloid but not plasmacytoid dendritic cells, J. Immunol. 178 (2007) 145–153.
Y.S. Liu, Vitamin D level affects IVF outcome partially mediated via Th/Tc cell [65] C.M. Hawrylowicz, A.F. Santos, Vitamin D: can the sun stop the atopic epidemic?
ratio, Am. J. Reprod. Immunol. 80 (2018) e13050. Curr. Opin. Allergy Clin. Immunol. (2019).
[37] K. Sharif, Y. Sharif, A. Watad, Y. Yavne, B. Lichtbroun, N.L. Bragazzi, H. Amital, [66] G. Penna, A. Roncari, S. Amuchastegui, K.C. Daniel, E. Berti, M. Colonna,
Y. Shoenfeld, Vitamin D, autoimmunity and recurrent pregnancy loss: more than L. Adorini, Expression of the inhibitory receptor ILT3 on dendritic cells is dis-
an association, Am. J. Reprod. Immunol. 80 (2018) e12991. pensable for induction of CD4+Foxp3+ regulatory T cells by 1,25-dihydrox-
[38] B. Novakovic, M. Sibson, H.K. Ng, U. Manuelpillai, V. Rakyan, T. Down, S. Beck, yvitamin D3, Blood 106 (2005) 3490–3497.
T. Fournier, D. Evain-Brion, E. Dimitriadis, J.M. Craig, R. Morley, R. Saffery, [67] A.M.G. Van Der Aar, D.S. Sibiryak, G. Bakdash, T.M.M. Van Capel, H.P.M. Van Der
Placenta-specific methylation of the vitamin D 24-hydroxylase gene: implications Kleij, D.-J.E. Opstelten, M.B.M. Teunissen, M.L. Kapsenberg, E.C. De Jong, Vitamin
for feedback autoregulation of active vitamin D levels at the fetomaternal inter- D3 targets epidermal and dermal dendritic cells for induction of distinct regulatory
face, J. Biol. Chem. 284 (2009) 14838–14848. T cells, J. Allergy Clin. Immunol. 127 (2011) 1532-1540.e7.
[39] A. Olmos-Ortiz, E. Avila, M. Durand-Carbajal, L. Diaz, Regulation of calcitriol [68] T. Nikolic, B.O. Roep, Regulatory multitasking of tolerogenic dendritic cells -
biosynthesis and activity: focus on gestational vitamin D deficiency and adverse lessons taken from vitamin d3-treated tolerogenic dendritic cells, Front. Immunol.
pregnancy outcomes, Nutrients 7 (2015) 443–480. 4 (2013) 113.
[40] A. Vijayendra Chary, R. Hemalatha, M. Seshacharyulu, M. Vasudeva Murali, [69] Z. Urry, E.S. Chambers, E. Xystrakis, S. Dimeloe, D.F. Richards, L. Gabryšová,
D. Jayaprakash, B. Dinesh Kumar, Vitamin D deficiency in pregnant women im- J. Christensen, A. Gupta, S. Saglani, A. Bush, A. O’garra, Z. Brown,
pairs regulatory T cell function, J. Steroid Biochem. Mol. Biol. 147 (2015) 48–55. C.M. Hawrylowicz, The role of 1α,25-dihydroxyvitamin D3 and cytokines in the
[41] J. Knabl, A. Vattai, Y. Ye, J. Jueckstock, S. Hutter, F. Kainer, S. Mahner, promotion of distinct Foxp3+and IL-10+CD4+T cells, Eur. J. Immunol. 42
U. Jeschke, Role of placental VDR expression and function in common late preg- (2012) 2697–2708.
nancy disorders, Int. J. Mol. Sci. (2017) 18. [70] P.E. Pfeffer, C.M. Hawrylowicz, Vitamin d in asthma: mechanisms of action and
[42] J. Kaludjerovic, R. Vieth, Relationship between vitamin D during perinatal de- considerations for clinical trials, Chest 153 (2018) 1229–1239.
velopment and health, J. Midwifery Womens Health 55 (2010) 550–560. [71] L.E. Jeffery, F. Burke, M. Mura, Y. Zheng, O.S. Qureshi, M. Hewison, L.S.K. Walker,
[43] D.D. Bikle, E. Gee, B. Halloran, J.G. Haddad, Free 1,25-dihydroxyvitamin D levels D.A. Lammas, K. Raza, D.M. Sansom, 1,25-dihydroxyvitamin D3 and IL-2 combine
in serum from normal subjects, pregnant subjects, and subjects with liver disease, to inhibit t cell production of inflammatory cytokines and promote development of
J. Clin. Invest. 74 (1984) 1966–1971. regulatory t cells expressing CTLA-4 and FoxP3, J. Immunol. 183 (2009)
[44] B.W. Hollis, D. Johnson, T.C. Hulsey, M. Ebeling, C.L. Wagner, Vitamin D sup- 5458–5467.
plementation during pregnancy: double blind, randomized clinical trial of safety [72] J.E. Vasiliou, S. Lui, S.A. Walker, V. Chohan, E. Xystrakis, A. Bush,
and effectiveness, J. Bone Miner. Res. 26 (2011) 2341–2357. C.M. Hawrylowicz, S. Saglani, C.M. Lloyd, Vitamin D deficiency induces Th2
[45] J.A. Tamblyn, C. Jenkinson, D.P. Larner, M. Hewison, M.D. Kilby, Serum and urine skewing and eosinophilia in neonatal allergic airways disease, Allergy 69 (2014)
vitamin D metabolite analysis in early preeclampsia, Endocr. Connect. 7 (2018) 1380–1389.
199–210. [73] E. Hornsby, P.E. Pfeffer, N. Laranjo, W. Cruikshank, M. Tuzova, A.A. Litonjua,
[46] B.W. Hollis, C.L. Wagner, New insights into the vitamin D requirements during S.T. Weiss, V.J. Carey, G. O’connor, C. Hawrylowicz, Vitamin D supplementation
pregnancy, Bone Res. 5 (2017) 17030. during pregnancy: effect on the neonatal immune system in a randomized con-
[47] M. Whitehead, G. Lane, O. Young, S. Campbell, G. Abeyasekera, C.J. Hillyard, trolled trial, J. Allergy Clin. Immunol. 141 (269-278) (2018) e1.
I. Macintyre, K.G. Phang, J.C. Stevenson, Interrelations of calcium-regulating [74] A. Chi, J. Wildfire, R. Mcloughlin, R.A. Wood, G.R. Bloomberg, M. Kattan,
hormones during normal pregnancy, Br. Med. J. Clin. Res. Ed (Clin Res Ed) 283 P. Gergen, D.R. Gold, F. Witter, T. Chen, M. Holick, C. Visness, J. Gern,
(1981) 10–12. G.T. O’connor, Umbilical cord plasma 25-hydroxyvitamin D concentration and
[48] T.K. Gray, G.E. Lester, R.S. Lorenc, Evidence for extra-renal 1 alpha-hydroxylation immune function at birth: the Urban Environment and Childhood Asthma study,
of 25-hydroxyvitamin D3 in pregnancy, Science 204 (1979) 1311–1313. Clin. Exp. Allergy 41 (2011) 842–850.
[49] R.M. Carneiro, L. Prebehalla, M.B. Tedesco, S.M. Sereika, M. Hugo, B.W. Hollis, [75] A. Guven, A. Ecevit, O. Sozer, A. Tarcan, A. Tarcan, N. Ozbek, Correlation between
C.M. Gundberg, A.F. Stewart, M.J. Horwitz, Lactation and bone turnover: a con- the cord vitamin D levels and regulatory T cells in newborn infants, Eur. J. Pediatr.
undrum of marked bone loss in the setting of coupled bone turnover, J. Clin. 171 (2012) 1161–1166.
Endocrinol. Metab. 95 (2010) 1767–1776. [76] C. Palacios, M.A. Trak-Fellermeier, R.X. Martinez, L. Lopez-Perez, P. Lips,
[50] L.D. Ritchie, E.B. Fung, B.P. Halloran, J.R. Turnlund, M.D. Van Loan, C.E. Cann, J.A. Salisi, J.C. John, J.P. Pena-Rosas, Regimens of vitamin D supplementation for
J.C. King, A longitudinal study of calcium homeostasis during human pregnancy women during pregnancy, Cochrane Database Syst. Rev. 10 (2019) CD013446.
and lactation and after resumption of menses, Am. J. Clin. Nutr. 67 (1998) [77] C.L. Wagner, R.B. Mcneil, D.D. Johnson, T.C. Hulsey, M. Ebeling, C. Robinson,

8
M.E. Kiely, et al. Journal of Steroid Biochemistry and Molecular Biology 201 (2020) 105669

S.A. Hamilton, B.W. Hollis, Health characteristics and outcomes of two rando- [100] C. Palacios, L.K. Kostiuk, J.P. Pena-Rosas, Vitamin D supplementation for women
mized vitamin D supplementation trials during pregnancy: a combined analysis, J. during pregnancy, Cochrane Database Syst. Rev. 7 (2019) CD008873.
Steroid Biochem. Mol. Biol. 136 (2013) 313–320. [101] C. Cooper, N.C. Harvey, N.J. Bishop, S. Kennedy, A.T. Papageorghiou,
[78] C.L. Wagner, R. Mcneil, S.A. Hamilton, J. Winkler, C. Rodriguez Cook, G. Warner, I. Schoenmakers, R. Fraser, S.V. Gandhi, A. Carr, S. D’angelo, S.R. Crozier,
B. Bivens, D.J. Davis, P.G. Smith, M. Murphy, J.R. Shary, B.W. Hollis, A rando- R.J. Moon, N.K. Arden, E.M. Dennison, K.M. Godfrey, H.M. Inskip, A. Prentice,
mized trial of vitamin D supplementation in 2 community health center networks M.Z. Mughal, R. Eastell, D.M. Reid, M.K. Javaid, M.S. Group, Maternal gestational
in South Carolina, Am. J. Obstet. Gynecol. 208 (137) (2013) e1-137 e13. vitamin D supplementation and offspring bone health (MAVIDOS): a multicentre,
[79] A. Al-Garawi, V.J. Carey, D. Chhabra, H. Mirzakhani, J. Morrow, J. Lasky-Su, double-blind, randomised placebo-controlled trial, Lancet Diabetes Endocrinol. 4
W. Qiu, N. Laranjo, A.A. Litonjua, S.T. Weiss, The role of vitamin d in the tran- (2016) 393–402.
scriptional program of human pregnancy, PLoS One 11 (2016) e0163832. [102] B.W. Hollis, C.L. Wagner, Vitamin d and pregnancy: skeletal effects, nonskeletal
[80] B.L. Chawes, K. Bønnelykke, J. Stokholm, Al. Et, Effect of vitamin D3 supple- effects, and birth outcomes, Calcif. Tissue Int. 92 (2013) 128–139.
mentation during pregnancy on risk of persistent wheeze in the offspring: a ran- [103] B.W. Hollis, Vitamin D status during pregnancy: the importance of getting it right,
domized clinical trial, JAMA 315 (2016) 353–361. EBioMedicine 39 (2019) 23–24.
[81] Q. Zhang, Y. Cheng, M. He, T. Li, Z. Ma, H. Cheng, Effect of various doses of [104] A. Grey, M.J. Bolland, Inaccurate dissemination of the MAVIDOS trial results,
vitamin D supplementation on pregnant women with gestational diabetes mellitus: Lancet Diabetes Endocrinol. 4 (2016) 481.
a randomized controlled trial, Exp. Ther. Med. 12 (2016) 1889–1895. [105] H.M. Wolsk, B.L. Chawes, A.A. Litonjua, B.W. Hollis, J. Waage, J. Stokholm,
[82] A.A. Litonjua, V.J. Carey, N. Laranjo, B.J. Harshfield, T.F. Mcelrath, G.T. O’connor, K. Bonnelykke, H. Bisgaard, S.T. Weiss, Prenatal vitamin D supplementation re-
M. Sandel, R.E. Iverson Jr., A. Lee-Paritz, R.C. Strunk, L.B. Bacharier, duces risk of asthma/recurrent wheeze in early childhood: a combined analysis of
G.A. Macones, R.S. Zeiger, M. Schatz, B.W. Hollis, E. Hornsby, C. Hawrylowicz, two randomized controlled trials, PLoS One 12 (2017) e0186657.
A.C. Wu, S.T. Weiss, Effect of prenatal supplementation with vitamin d on asthma [106] N. Brustad, A.U. Eliasen, J. Stokholm, K. Bonnelykke, H. Bisgaard, B.L. Chawes,
or recurrent wheezing in offspring by age 3 years: the VDAART randomized High-dose vitamin d supplementation during pregnancy and asthma in offspring at
clinical trial, JAMA 315 (2016) 362–370. the age of 6 years, JAMA 321 (2019) 1003–1005.
[83] H.M. Wolsk, B.J. Harshfield, N. Laranjo, V.J. Carey, G. O’connor, M. Sandel, [107] S. Behjat Sasan, F. Zandvakili, N. Soufizadeh, E. Baybordi, The effects of vitamin d
R.C. Strunk, L.B. Bacharier, R.S. Zeiger, M. Schatz, B.W. Hollis, S.T. Weiss, supplement on prevention of recurrence of preeclampsia in pregnant women with
A.A. Litonjua, Vitamin D supplementation in pregnancy, prenatal 25(OH)D levels, a history of preeclampsia, Obstet. Gynecol. Int. 2017 (2017) 8249264.
race, and subsequent asthma or recurrent wheeze in offspring: secondary analyses [108] Z. Asemi, M. Samimi, Z. Tabassi, H. Shakeri, A. Esmaillzadeh, Vitamin D supple-
from the Vitamin D Antenatal Asthma Reduction Trial, J. Allergy Clin. Immunol. mentation affects serum high-sensitivity C-reactive protein, insulin resistance, and
140 (1423-1429) (2017) e5. biomarkers of oxidative stress in pregnant women, J. Nutr. 143 (2013)
[84] D.E. Roth, S.K. Morris, S. Zlotkin, A.D. Gernand, T. Ahmed, S.S. Shanta, E. Papp, 1432–1438.
J. Korsiak, J. Shi, M.M. Islam, I. Jahan, F.K. Keya, A.R. Willan, R. Weksberg, [109] A. Sablok, A. Batra, K. Thariani, A. Batra, R. Bharti, A.R. Aggarwal, B.C. Kabi,
M. Mohsin, Q.S. Rahman, P.S. Shah, K.E. Murphy, J. Stimec, L.G. Pell, H. Qamar, H. Chellani, Supplementation of vitamin D in pregnancy and its correlation with
A. Al Mahmud, Vitamin d supplementation in pregnancy and lactation and infant feto-maternal outcome, Clin Endocrinol (Oxf) 83 (2015) 536–541.
growth, N. Engl. J. Med. 379 (2018) 535–546. [110] E. Naghshineh, S. Sheikhaliyan, Effect of vitamin D supplementation in the reduce
[85] T.M. Stukes, J.R. Shary, W. Wei, M.D. Ebeling, K.B. Dezsi, F.S. Shary, risk of preeclampsia in nulliparous women, Adv. Biomed. Res. 5 (2016) 7.
N.E. Forestieri, B.W. Hollis, C.L. Wagner, Circulating cathelicidin concentrations [111] S. Fogacci, F. Fogacci, M. Banach, E.D. Michos, A.V. Hernandez, G.Y.H. Lip,
in a cohort of healthy children: influence of age, body composition, gender and M.J. Blaha, P.P. Toth, C. Borghi, A.F.G. Cicero, Lipid & Blood Pressure Meta-
vitamin d status, PLoS One 11 (2016) e0152711. Analysis Collaboration, G, Vitamin D supplementation and incident preeclampsia:
[86] R.S. Kelly, B.L. Chawes, F. Guo, L. Zhang, K. Blighe, A.A. Litonjua, B.A. Raby, a systematic review and meta-analysis of randomized clinical trials, Clin. Nutr.
B.D. Levy, D. Rago, J. Stokholm, K. Bonnelykke, H. Bisgaard, X. Zhou, J.A. Lasky- (2019).
Su, S.T. Weiss, The role of the 17q21 genotype in the prevention of early childhood [112] M. Tous, M. Villalobos, L. Iglesias, S. Fernandez-Barres, V. Arija, Vitamin D status
asthma and recurrent wheeze by vitamin D, Eur. Respir. J. (2019) 54. during pregnancy and offspring outcomes: a systematic review and meta-analysis
[87] A.A. Litonjua, V.J. Carey, N. Laranjo, B.J. Stubbs, H. Mirzakhani, G.T. O’connor, of observational studies, Eur. J. Clin. Nutr. (2019).
M. Sandel, A. Beigelman, L.B. Bacharier, R.S. Zeiger, M. Schatz, B.W. Hollis, [113] S.S. Zhou, Y.H. Tao, K. Huang, B.B. Zhu, F.B. Tao, Vitamin D and risk of preterm
S.T. Weiss, Six-year follow-up of a trial of antenatal vitamin d for asthma reduc- birth: up-to-date meta-analysis of randomized controlled trials and observational
tion, N. Engl. J. Med. 382 (2020) 525–533. studies, J. Obstet. Gynaecol. Res. 43 (2017) 247–256.
[88] T. Aravena, S. Castillo, X. Carrasco, I. Mena, J. Lopez, J.P. Rojas, C. Rosembert, [114] Y. Yuan, W. Tai, P. Xu, Z. Fu, X. Wang, W. Long, X. Guo, C. Ji, L. Zhang, Y. Zhang,
C. Schroter, F. Aboitiz, Williams syndrome: clinical, cytogenetical, neurophysio- J. Wen, Association of maternal serum 25-hydroxyvitamin D concentrations with
logical and neuroanatomic study, Rev. Med. Child 130 (2002) 631–637. risk of preeclampsia: a nested case-control study and meta-analysis, J. Matern.
[89] C. Macdonald, T. Upton, P. Hunt, I. Phillips, M. Kaufmann, C. Florkowski, S. Soule, Fetal. Neonatal. Med. (2019) 1–10.
G. Jones, Vitamin D supplementation in pregnancy: a word of caution. Familial [115] A. Shub, E.A. Mccarthy, Letter to the editor: "Effectiveness of prenatal vitamin d
hypercalcaemia due to disordered vitamin D metabolism, Ann. Clin. Biochem. deficiency screening and treatment program: a stratified randomized field trial", J.
(2020) 4563219897691. Clin. Endocrinol. Metab. 104 (2019) 337–338.
[90] M. Rostami, F. Ramezani Tehrani, M. Simbar, R. Bidhendi Yarandi, S. Minooee, [116] B.W. Hollis, W.B. Pittard 3rd, Evaluation of the total fetomaternal vitamin D re-
B.W. Hollis, F. Hosseinpanah, Effectiveness of prenatal vitamin D deficiency lationships at term: evidence for racial differences, J. Clin. Endocrinol. Metab. 59
screening and treatment program: a stratified randomized field trial, J. Clin. (1984) 652–657.
Endocrinol. Metab. (2018). [117] K.M. O’callaghan, Á. Hennessy, G.L.J. Hull, K. Healy, C. Ritz, L.C. Kenny,
[91] N.Q. Liu, Y. Ouyang, Y. Bulut, V. Lagishetty, S.Y. Chan, B.W. Hollis, C. Wagner, K.D. Cashman, M.E. Kiely, Estimation of the maternal vitamin D intake that
O. Equils, M. Hewison, Dietary vitamin D restriction in pregnant female mice is maintains circulating 25-hydroxyvitamin D in late gestation at a concentration
associated with maternal hypertension and altered placental and fetal develop- sufficient to keep umbilical cord sera ≥25-30 nmol/L: a dose-response, double-
ment, Endocrinology 154 (2013) 2270–2280. blind, randomized placebo-controlled trial in pregnant women at northern lati-
[92] A. Dawodu, H.F. Saadi, G. Bekdache, Y. Javed, M. Altaye, B.W. Hollis, Randomized tude, Am. J. Clin. Nutr. 108 (2018) 77–91.
controlled trial (RCT) of vitamin D supplementation in pregnancy in a population [118] K.M. March, N.N. Chen, C.D. Karakochuk, A.W. Shand, S.M. Innis, P. Von
with endemic vitamin D deficiency, J. Clin. Endocrinol. Metab. 98 (2013) Dadelszen, S.I. Barr, M.R. Lyon, S.J. Whiting, H.A. Weiler, T.J. Green, Maternal
2337–2346. vitamin D3 supplementation at 50 ug/d protects against low serum 25-hydro-
[93] S.Y. Chan, R. Susarla, D. Canovas, E. Vasilopoulou, O. Ohizua, C.J. Mccabe, xyvitamin D in infants at 8 wk of age: a randomized controlled trial of 3 doses of
M. Hewison, M.D. Kilby, Vitamin D promotes human extravillous trophoblast in- vitamin D beginning in gestation and continued in lactation, Am. J. Clin. Nutr. 102
vasion in vitro, Placenta 36 (2015) 403–409. (2015) 402–410.
[94] M. Hewison, The earlier the better: preconception vitamin D and protection [119] C.C. Grant, A.W. Stewart, R. Scragg, T. Milne, J. Rowden, A. Ekeroma, C. Wall,
against pregnancy loss, Lancet Diabetes Endocrinol. 6 (2018) 680–681. E.A. Mitchell, S. Crengle, A. Trenholme, J. Crane, C.A. Camargo Jr., Vitamin D
[95] A. Ganguly, J.A. Tamblyn, S. Finn-Sell, S.Y. Chan, M. Westwood, J. Gupta, during pregnancy and infancy and infant serum 25-hydroxyvitamin D con-
M.D. Kilby, S.R. Gross, M. Hewison, Vitamin D, the placenta and early pregnancy: centration, Pediatrics 133 (2014) e143–53.
effects on trophoblast function, J. Endocrinol. 236 (2018) R93–r103. [120] G.J. Hofmeyr, J.M. Belizán, P. Von Dadelszen, & Calcium And Pre-Eclampsia (Cap)
[96] M. Abercrombie, J. Shary, M. Ebeling, B. Hollis, C. Wagner, Analysis of the NICHD Study Group, Low-dose calcium supplementation for preventing pre-eclampsia: a
Vitamin D Pregnancy Cohort on a Per-Protocol vs. (2018). systematic review and commentary, BJOG 121 (2014) 951–957.
[97] R.P. Heaney, Guidelines for optimizing design and analysis of clinical studies of [121] A. Hemmingway, L.C. Kenny, L. Malvisi, M.E. Kiely, Exploring the concept of
nutrient effects, Nutr. Rev. 72 (2014) 48–54. functional vitamin D deficiency in pregnancy: impact of the interaction between
[98] O. Tsuprykov, C. Buse, R. Skoblo, B. Hocher, Comparison of free and total 25- 25-hydroxyvitamin D and parathyroid hormone on perinatal outcomes, Am. J.
hydroxyvitamin D in normal human pregnancy, J. Steroid Biochem. Mol. Biol. 190 Clin. Nutr. 108 (2018) 821–829.
(2019) 29–36. [122] C.R. Sudfeld, E.R. Smith, New evidence should inform WHO guidelines on mul-
[99] World Health Organization, WHO Recommendations on Antenatal Care for a tiple micronutrient supplementation in pregnancy, J. Nutr. 149 (2019) 359–361.
Positive Pregnancy Experience, World Health Organization, Geneva, 2016.

You might also like