Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Current Drug Metabolism, 2012, 13, 000-000 1

Use of Non-steroidal Anti-inflammatory Drugs in Pregnancy: Impact on the Fetus


and Newborn

Roberto Antonucci1*, Marco Zaffanello2, Puxeddu Elisabetta3, Annalisa Porcella4, Laura Cuzzolin5, Maria
Dolores Pilloni6 and Vassilios Fanos3

1
Division of Neonatology and Pediatrics, “Nostra Signora di Bonaria” Hospital, San Gavino Monreale, Italy; 2Section of Pediatrics,
Department of Life and Reproduction Sciences, University of Verona, Verona, Italy; 3Neonatal Intensive Care Unit, University of
Cagliari, Cagliari, Italy; 4Division of Neonatology, “San Camillo” Hospital, Sorgono, Italy; 5Department of Public Health & Com-
munity Medicine-Section of Pharmacology, University of Verona, Italy; 6Consultorio Familiare, San Gavino Monreale, ASL 6 Sanluri
(VS), Italy

Abstract: Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly prescribed in pregnancy to treat fever, pain and inflammation.
Indications for chronic use of these agents during pregnancy are inflammatory bowel or chronic rheumatic diseases. Since the seventies,
NSAIDs have been used as effective tocolytic agents: indomethacin has been the reference drug, delaying delivery for at least 48 hours
and up to 7-10 days. Additionally, self-medication with NSAIDs is practiced by pregnant women.
NSAIDs given to pregnant women cross the placenta and may cause embryo-fetal and neonatal adverse effects, depending on the type of
agent, the dose and duration of therapy, the period of gestation, and the time elapsed between maternal NSAID administration and deliv-
ery. These effects derive from the action mechanisms of NSAIDs (mainly inhibition of prostanoid activity) and from the physiological
changes in drug pharmacokinetics occurring during pregnancy.
Increased risks of miscarriage and malformations are associated with NSAID use in early pregnancy. Conversely, exposure to NSAIDs
after 30 weeks’ gestation is associated with an increased risk of premature closure of the fetal ductus arteriosus and oligohydramnios. Fe-
tal and neonatal adverse effects affecting the brain, kidney, lung, skeleton, gastrointestinal tract and cardiovascular system have also been
reported after prenatal exposure to NSAIDs.
NSAIDs should be given in pregnancy only if the maternal benefits outweigh the potential fetal risks, at the lowest effective dose and for
the shortest duration possible.
This article discusses in detail the placental transfer and metabolism of NSAIDs, and the adverse impact of prenatal NSAID exposure on
the offspring.
Keywords: Non-steroidal anti-inflammatory drugs, NSAIDs, indomethacin, tocolysis, pregnancy, adverse effects, embryo, fetus, newborn.

NON-STEROIDAL ANTI-INFLAMMATORY DRUGS AND A recent study has documented that in the majority of pregnan-
PREGNANCY cies (75.6%) NSAIDs and/or acetylsalicylic acid (ASA) were pre-
1. NSAID USE DURING PREGNANCY: INDICATIONS AND scribed during the first trimester, resulting in an overall first trimes-
EPIDEMIOLOGICAL DATA ter exposure of 2.9%. Part of the first trimester exposure would be
due to unawareness of pregnancy by the women. [3] Epidemiologi-
Non-steroidal anti-inflammatory drugs (NSAIDs) are among cal studies from Canada [6], Denmark [7], and Sweden [8] reported
the most common drugs prescribed to pregnant women to treat an exposure to NSAIDs during the first trimester of pregnancy of
fever, pain and inflammation [1]. Indications for a chronic use of 2,9%, 3%, and 3,4 %, respectively. On the other hand, third trimes-
NSAIDs during pregnancy are inflammatory bowel diseases [2] and ter prescribing of NSAIDs and/or ASA was found to be low (0.6%).
chronic rheumatic diseases such as rheumatoid arthritis and spondy- This fact was to be expected due to guidelines stating not to pre-
larthropathy. On the other hand, obstetrical indications for NSAID scribe these agents during the third trimester [3].
administration include the management of premature labor (indo-
methacin), the prevention of gestosis, and the treatment of polyhy- This review article includes a general overview of NSAID use
dramnios and antiphospholipid syndrome [1]. during pregnancy, with special emphasis on the mechanism of ac-
tion, placental transfer and metabolism of these medications; then,
The frequency of NSAID use during pregnancy is not easily the embryo-fetal and neonatal adverse effects related to the antena-
countable, considering that some of these drugs are available over tal use of NSAIDs are discussed in detail.
the counter (OTC) and thus can be easily purchased without medi-
cal prescription. Unpublished data of EUROCAT-registration 2. MECHANISM OF ACTION OF NSAIDS
Northern-Netherlands revealed that NSAID exposure during preg-
Prostanoids belong to a family of biologically active lipids,
nancy was due to 60% on prescription and 35% OTC. [3] Insuffi-
including prostaglandins, prostacyclins, and thromboxanes. Prosta-
cient information about OTC-use of NSAIDs may lead to underes-
noids are synthesized as needed in three steps. The first step is the
timating their actual use. Furthermore, the consumption of NSAIDs
release of arachidonic acid from membrane phospholipids through
in early pregnancy may occur due to unawareness of the pregnant
the action of phospholipase-A2. In the second step, cyclooxygenase
state, or through ignoring the composition of self-medication drugs
enzyme first oxidizes arachidonic acid to form prostaglandin G2
that are being used [4,5].
(PGG2) through a cyclooxygenase process and then peroxidizes
PGG2 to PGH2. Finally, this highly instable compound is converted
*Address correspondence to this author at the Division of Neonatology and into five major prostanoids, namely prostaglandin E2 (PGE2),
Pediatrics, "Nostra Signora di Bonaria" Hospital, San Gavino Monreale, PGF2, PGD2, PGI2 and thromboxane A2 (TXA2) by specific PG
Italy; Tel:/Fax: ??????????????????????????; E-mail: roant@tiscali.it synthase enzymes [9].

 1389-2002/12 $58.00+.00 © 2012 Bentham Science Publishers


2 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

The key enzyme in the prostaglandin synthesis is prostaglandin inflammatory effects, which may not be entirely attributable to
endoperoxide synthase (PGHS) or cyclooxygenase (COX). This direct COX inhibition [16].
bifunctional enzyme possesses two catalytic sites, namely a cy- Classical NSAIDs (e.g., diclofenac, ibuprofen and naproxen)
clooxygenase active site and a peroxidase active site. We now know are nonselective COX inhibitors that inhibit both COX-1 and COX-
that there are two major isoforms of COX, coded by two distinct 2 isoforms in different degrees. On the other hand, the more recent
genes, and referred to as COX-1 and COX-2. COX-1, which has drugs celecoxib and rofecoxib belong to the highly selective COX-2
been designated as “constitutive” because of its distribution in vir- inhibitors (COXIBs).
tually all tissues under basal conditions, is responsible for the pro-
duction of prostaglandins with general “housekeeping” functions, Although NSAIDs clearly inhibit the synthesis and release of
such as gastric cytoprotection, vascular homeostasis, platelet aggre- prostaglandins, these actions are not sufficient to explain all the
gation, and kidney function. On the other hand, COX-2 has been antiinflammatory effects of NSAIDs.
termed “inducible” because of its more restricted basal expression In vitro, some NSAIDs such as indomethacin, diclofenac, keto-
and its upregulation during inflammation and cellular transforma- profen, and aspirin, have been shown to strongly inhibit the neutro-
tion. It is noteworthy that COX-2 is expressed constitutively in the phil-endothelial cell attachment, which is a critical step in the in-
fetal and adult kidney, and in the perinatal brain. Some years ago, flammatory response. The underlying mechanism of this NSAID-
Chandrasekharan et al. [10] identified a COX-1 splice variant, des- induced effect has been attributed to a rapid reduction in the expres-
ignated as COX-3, in the canine cerebral cortex. This new COX sion of L-selectin on the neutrophil surface [17].
isoform, absent in the fetal CNS tissue but abundantly expressed in It is well-known that nitric oxide (NO) formation is increased
the mature brain, regulates fever and pain sensitivity. during inflammation, and that several classic inflammatory symp-
The three-dimensional structure of COX-1 was determined by toms (erythema and vascular leakiness) are reversed by nitric oxide
Picot et al. [11]. This enzyme comprises three independent folding synthase (NOS) inhibitors [18,19]. Moreover, cytokines and other
units: an epidermal growth factor-like domain, a membrane binding proinflammatory mediators induce inducible NOS (iNOS), conse-
motif and an enzymatic domain. The site for peroxidase activity is quently leading to increased inflammation [20]. Amin et al. [21]
adjacent but spatially distinct from the site for cyclooxygenase ac- have documented that the inhibition of iNOS expression and func-
tivity. The conformation of the membrane-binding motif strongly tion represents another action mechanism for aspirin (if not for all
suggests that the enzyme is a monotopic membrane protein. Three aspirin-like drugs), and that these effects are exerted at the level of
of the helices of the structure lie at the entrance to the cyclooxy- translational/posttranslational modification and on the catalytic
genase channel and their insertion into the membrane could allow activity of iNOS.
arachidonic acid to reach the active site from the interior of the Recently, Suleyman et al. [22] have shown that some NSAIDs,
bilayer [12]. The COX active site is a long, hydrophobic channel, namely indomethacin, diclofenac sodium, ibuprofen, nimesulide,
and Picot et al. [11] have provided evidence that some aspirin-like tenoxicam, and aspirin, produce significant anti-inflammatory ef-
drugs, such as flurbiprofen, inhibit COX-1 by excluding arachi- fects in intact rats but insignificant effects in adrenalectomized rats.
donic acid from the upper portion of the channel. Tyrosine 385 and Therefore, the Authors have concluded that anti-inflammatory ef-
serine 530 are at the apex of this active site [12]. fects of these agents are related to adrenalin and cortisol.
The three dimensional structure of COX-2 has also been de-
scribed [13]. This structure closely resembles that of COX-1, except 3. CYCLO-OXYGENASE INHIBITORS AND TOCOLYSIS
that the COX-2 active site is slightly larger and can accommodate The molecular mechanisms underlying the onset and mainte-
larger structures than those which are able to fit the COX-1 active nance of labor at term have not been fully established, but it is cer-
site. A secondary internal pocket contributes significantly to the tain that prostaglandins (PGs) are strongly implicated [23]. Fur-
larger volume of the active site of COX-2, even though the central thermore, PGs are important mediators in delivery, stimulating
channel is also bigger by 17%. Selectivity for COX-2 inhibitors is uterine contraction and enhancing cervical ripening. The expression
dependent on the replacement of histidine 513 and isoleucine 523 of COX-2, but not of COX-1, in fetal membranes and myometrium
of COX-1 with arginine and valine, respectively. increases in association with labor, and thus this COX isoform is
A number of NSAID mechanisms of action have been de- likely to play a role in the increased prostaglandin synthesis ob-
scribed. In 1971, Vane [14] proposed that the action mechanism of served in the uterus at term [24-27]. These findings could explain
the aspirin-like drugs (i.e., NSAIDs) was through the inhibition of why the COX inhibitors administered in the last trimester of preg-
PG synthesis, and this theory is generally accepted today. Prosta- nancy prolong gestation both in animals and in humans [1].
glandin inhibition by aspirin is due to the irreversible acetylation of In a Cochrane review [28], when compared to conventional
the COX site of PGHS, while the peroxidase activity of the enzyme tocolytics (betamimetics and magnesium sulphate), cyclo-
is unaffected. In contrast to this irreversible action of aspirin, other oxygenase inhibitors (mainly indomethacin, but also sulindac, ke-
NSAIDs such as indomethacin or ibuprofen cause reversible COX torolac, nimesulide and rofecoxib) resulted in a reduction in the
inhibition by competing with the substrate, arachidonic acid, for the number of women delivering prior to 37 weeks’ gestation and fewer
active site of PGHS. maternal side-effects. Moreover, a trend toward a reduction in the
Aspirin differs from other NSAIDs because of its irreversible number of women delivering before 48 hours after initiation of
inhibition of both COX isoforms [1]. In the case of COX-2, how- treatment was observed, while no difference was shown at 7 days.
ever, the efficiency by which serine 530 is trans-acetylated by aspi- In a recent meta-analysis [29] prostaglandin inhibitors (indometha-
rin, resulting in enzyme inhibition, is at least 10-fold less than for cin, sulindac, ketorolac, mefenamic acid, nimesulide, celecoxib,
COX-1 isoform. rofecoxib) resulted superior to the other tocolytic agents (be-
tamimetics, calcium-channel blockers, magnesium sulphate, oxyto-
Furthermore, the action mechanisms and effects of aspirin vary cin receptor antagonists and nitrates) providing the best combina-
with dose. Low doses (75-81 mg/day) are able to irreversibly acety- tion of delayed delivery and tolerance. Compared to placebo, all
late COX-1 serine 530 [15,16], thus inhibiting thromboxane A2 tocolytic agents were able to delay delivery by 48 hours and 7 days.
platelet production and resulting in an antithrombotic effect. Inter- However, a detailed data analysis demonstrated that prostaglandin
mediate doses (650 mg-4 g/day) inhibit COX-1 and COX-2, thus inhibitors may be considered the optimal first-line agents before 32
blocking prostaglandin production, and have analgesic and antipy- weeks’ gestation to delay delivery for 48 hours and 7 days (lower
retic effects. Finally, high doses (4-8 g/day) of aspirin have anti- rate of failure events, higher proportion of patients tolerating treat-
ment and achieving outcomes), whereas calcium-channel blockers
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 3

were superior first-line tocolytic agents to delay delivery until 37 glycoprotein), since the complex drug-protein is too large to cross
weeks’ gestation [29]. the placenta. However, this binding is a transient phenomenon and
King et al. [28] reported that cyclo-oxygenase inhibitors inhibit when the drug separates from the protein it is able to cross the pla-
uterine contractions, are easily administered and have fewer mater- centa establishing an equilibrium between maternal and fetal circu-
nal side-effects compared to conventional tocolytics. A recent study lations. Although plasma protein binding could alter the total con-
has documented that prostaglandin inhibitors are superior to the centration of drugs in the maternal and fetal plasma, the free con-
other tocolytic agents and may be considered the optimal first-line centrations in the mother and fetus will not differ [42].
therapy before 32 weeks of gestation to delay delivery [29]. The extent to which drugs cross the placenta is also modulated
Since the seventies, NSAIDs have been used as effective toco- by the actions of placental phase I and II drug-metabolizing en-
lytic agents. Indomethacin (INDO) has been the reference drug, zymes, which are present at fluctuating levels throughout preg-
delaying delivery for at least 48 hours and up to 7-10 days [30]. The nancy. Cytochrome P450 (CYP) enzymes have been well character-
placenta is readily permeable to INDO, allowing similar maternal ized in the human placenta [41]. The type and amount of expressed
and fetal concentrations of the drug [31]. However, its use is lim- CYPs vary depending on the period of gestation and maternal
ited by fetal and neonatal severe adverse effects, as well as an in- health status. More CYP isoforms are expressed in the first trimes-
creased risk of perinatal death. The exact incidence of such side ter than at term gestation: it has been hypothesized that during the
effects is still being debated [32,33]. early development of the fetus the expression of CYPs is maximal
in order to counteract the effects of teratogens while, later, the
Preferential or selective inhibitors of COX-2 (particularly su- CYPs not needed for fetal well-being are switched off [43].
lindac, nimesulide and celecoxib) have been proposed for the CYP1A1, CYP2E1, CYP3A4, CYP3A7 and CYP4B1 have been
treatment of preterm delivery, expecting less fetal and maternal side detected in the term placenta [44]. Much less is known about phase
effects. Clinical and pharmacological data on the perinatal use of II enzymes in the placenta. Uridine diphosphate glucuronosyltrans-
selective COX-2 inhibitors are limited: nevertheless, these drugs ferases, which are involved in the conjugation of xenobiotics with
appear as effective as INDO in preventing preterm delivery in hu- glucuronic acid, are present in the placenta throughout gestation
mans and animals, with similar side effects [28, 29, 34-38]. suggesting a significant role of this enzyme in placental drug de-
4. PLACENTAL TRANSFER AND METABOLISM OF toxification [45]. Moreover, sulfation in the placenta may be a sig-
NSAIDS nificant biotransformation pathway for some drugs [41].
During pregnancy, physiological changes occur that can affect Several studies have documented that NSAIDs cross the human
the pharmacokinetics of drugs. Most of these changes begin early in placenta. Placental transfer of aspirin [46], indomethacin, sulindac,
gestation and are more pronounced in the third trimester [39]. naproxen [47], and diclofenac [48] has been shown both in placenta
perfusion models and in placenta and fetal tissues of women admit-
Absorption of drugs administered orally is delayed because of ted for termination of pregnancy [49].
slow gastric emptying and reduced intestinal motility, largely due to
progesterone action on smooth muscle activity [40]. The volume of Because of their lipid solubility, salicylates and aspirin were
distribution of drugs increases during pregnancy, as by the 6th to 8th demonstrated to cross the human placenta and widely distribute to
week of gestation plasma volume has expanded and continues to the fetus and extrafetal fluids, but equilibrium is achieved slowly.
increase until approximately 32 to 34 weeks’ gestation. The con- In pregnancy, the lower concentration of plasma albumin is associ-
centration of plasma albumin decreases during pregnancy, while ated with a lower binding of salicylate and an increase in volume of
plasma concentrations of 1-acid glycoprotein are relatively un- distribution. However, the low levels in fetal plasma are not due to
changed [39]. The metabolic activity of the liver is increased, ac- differences in protein binding but rather to the lower pH of fetal
celerating the metabolism of lipophilic drugs: progesterone as well blood. From limited data, it appears that the clearance of salicylate
as other placental hormones can be responsible for hepatic enzy- is increased during pregnancy [1,50].
matic activity changes [39]. Renal filtration is increased, leading to Placental transfer of indomethacin seems to be unrelated to the
enhanced renal elimination of water-soluble drugs. These modifica- gestational age in the human fetus. This drug distributes in fetal
tions are generally responsible for reduced plasma concentrations tissues and achieves equilibrium in about 6 hours post-dose [1,51].
and reduced half-life of most drugs [40]. The half –life of the drug is much longer in the fetal circulation
The distribution of a drug from the maternal circulation into the (14.7 h) than in the maternal circulation (2.2.h) [31].
placenta is the first step for its transfer across the placenta and is Sulindac, a pro-drug, readily crosses the placenta. Its active
mainly a function of both uterine blood flow and permeability of sulfide metabolite, formed through maternal hepatic metabolism,
the placental membrane [41]. may cross the placenta and reach the fetal circulation. Transplacen-
The major function of the placenta is to transfer nutrients and tal passage seems to be low after a single, oral dose of sulindac in
oxygen from the mother to the fetus, to synthesize hormones and pregnant women (24-36 weeks of gestation). However, the human
peptides vital for a successful pregnancy, and to remove waste placental perfusion model showed that sulindac sulfide reached the
products. The impression that the placenta forms an impenetrable fetus in a high concentration [49,52].
obstacle against drugs is widely regarded as false. Nearly all drugs Naproxen achieves a drug concentration in fetal serum compa-
administered during pregnancy enter the circulation of the fetus and rable to that in maternal serum after repeated dosing [53].
a variety of pharmacokinetic variables, including transplacental Transplacental transfer of the COXIBs has to be expected due
transport and metabolism, determine the degree of maternal-to-fetal to their low molecular weight. Rofecoxib has been shown to cross
drug transfer and exposure. Placental transfer of drugs occurs pri- the placenta in rats and rabbits [1,53].
marily through passive diffusion, so the physicochemical properties
of drugs such as polarity, molecular weight < 500 D and liposolu- MATERNAL EXPOSURE TO NSAIDS DURING PREG-
bility influence the rate of transfer. In addition, some drugs are NANCY AND ADVERSE EFFECTS ON THE OFFSPRING
pumped across the placenta by various transporters, while facili- Adverse effects on the fetus and newborn have been reported
tated diffusion appears to be a minor mechanism and pinocytosis is after prenatal exposure to COX inhibitors as a result of the transpla-
considered too slow to have any significant effect on fetal drug cental passage of these agents [28,54,55].
concentrations [41].
The major pharmacological action of NSAIDs on the fetus
The extent of drug transfer may also be affected by its own might be mediated through the inhibition of the prostaglandin syn-
degree of binding to plasma proteins (mainly albumin or 1-acid
4 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

thesis pathway, that influences blood circulation in various organs. was available. Futhermore, the results of this study did not exclude
Recent studies have not demonstrated any differences in neonatal the possibility of confounding by indication (as an example, the
outcomes when comparing the maternal administration of non- prescribing of a pharmacological agent to treat pain that may have
selective COX inhibitors with COX-2 inhibitors [1,28]. This obser- been a precursor of miscarriage). More recently, a population-based
vation could be explained by considering that COX-2 is expressed cohort study by Li et al. [63] documented that antenatal NSAID
in a variety of fetal tissues [1]. administration was associated with an 80% increased risk of mis-
NSAID effects on the fetus and newborn are different depend- carriage after adjustment for potential confounders. Moreover, the
ing upon the period of pregnancy in which the drugs have been association was stronger if the initial NSAID use was around the
administered to the mother. Women who are treated with NSAIDs time of conception, or if NSAID use lasted more than a week, sug-
during early pregnancy may be at a greater risk of having children gesting a dose-response relationship. Similarly, antenatal aspirin
with congenital anomalies [6]. On the other hand, premature closure use was associated with an increased risk of miscarriage. As sug-
of the ductus arteriosus has been observed in fetuses of mothers gested by the authors, the probable mechanism underlying this ef-
treated with NSAIDs during the third trimester of gestation [56]. fect of NSAIDs in pregnant women is the suppression of prosta-
glandin biosynthesis in the tissues of the reproductive system: this
Both the duration of maternal NSAID therapy and the distance could lead to abnormal embryo implantation that predisposes to
between maternal administration and the delivery have been re- miscarriage. The above-mentioned mechanism is supported by the
ported as important factors in the development of adverse effects on findings of some animal studies showing that prostaglandins are
the fetus. Major et al. [57] documented that antenatal indomethacin important mediators in the process of embryo implantation into the
exposure occurring within < or = 24 hours of delivery and of at uterus wall [64-67]. In addition, through the suppression of prosta-
least 48 hours' duration was associated with an increased incidence glandin production, NSAIDs may have an adverse effect on placen-
of necrotizing enterocolitis in low-birth-weight neonates. Another tal perfusion and circulation, thus increasing the risk of fetal demise
study demonstrated that antenatal indomethacin treatment for [63].
longer than 2 days, with a daily or cumulative dosage >/=150 mg,
correlated with a significantly higher incidence of grade I-II intra- On the other hand, it should be considered that NSAIDs are also
ventricular hemorrhage. Furthermore, a combined ante- and postna- prescribed to pregnant women to treat antiphospholipid antibody
tal exposure, a cumulative antenatal exposure >/=150 mg, and a syndrome, which itself increases the risk of miscarriage. In women
duration of antenatal exposure of more than 2 days were associated with this syndrome, pregnancy was found to be more successful
with necrotizing enterocolitis, and a cumulative exposure with sep- with the treatment. Heparin, moderate-to-high dose of prednisone
sis [58]. and low dose aspirin are the most commonly used drugs [68].
A recent study has reported that prenatal indomethacin expo- 2. CELOSOMY
sure (duration > 48 hours between the last dose given to the mother
The term “celosomy” indicates a group of congenital anomalies
and delivery) could be associated with an increased risk of periven-
characterized by opening of the somatic wall with evisceration,
tricular leukomalacia [59].
including gastroschisis, omphalocele and umbilical cord hernia.
Prenatal NSAID administration was also found to have terato- [69] Gastroschisis is a defect in the abdominal wall, lateral to the
genic effects on the fetus. Two recent studies have reported an ad- umbilical cord, which is considered to be a vascular problem,
justed odds ratio (OR) of 1.04 and 2.21, respectively, for any con- probably due to an intrauterine interruption of the omphalomesen-
genital malformation in infants whose mothers had used NSAIDs in teric artery [70]. Recent reports indicate that gastroschisis is in-
early pregnancy [6,8]. In 1990, Montenegro e Palomino [60] docu- creasing in prevalence, while omphalocele has remained steady;
mented that the exposure to prenatal NSAIDs (naproxen, sulindac, this finding suggests that environmental factors may play a role in
indomethacin, diclofenac, and mefenamic acid) induced cleft palate the pathogenesis of abdominal wall defects [69].
in the mouse offspring, with sulindac being the most teratogenic. In
Some studies conducted in rat fetuses documented that aspirin
vitro, each drug under study prevented the fusion of the palatine
increases the risk of abdominal wall defects, unlike other non-
processes in treated explants, and the degree of inhibition was de-
selective COX inhibitors [71] and selective COX-2 inhibitors
pendent on the stage of development at the time of explantation. In
[71,72].
both in vivo and in vitro experiments, the drugs prevented medial
epithelial cell breakdown that normally occurs in the medial secon- Over the last 20 years, many human studies showed a higher
dary palatal epithelium. The findings of this study suggest that pros- risk of gastroschisis in newborns of pregnant women treated with
taglandins may play an important role in the normal differentiation NSAIDs, particularly aspirin [73-77].
of the developing palatine region [60]. More recently, Burdan et al. In 2002, Werler et al. [77] conducted a retrospective study to
[61] have reported that gastroschisis is the only congenital ibupro- evaluate the relation between maternal use of cough/cold/analgesic
fen-related malformation that was proved in large epidemiological drugs and risk of gastroschisis. Risks of gastroschisis were found to
human studies. Other congenital anomalies associated with mater- be elevated for maternal use of aspirin (OR = 2.7), pseudoephedrine
nal ibuprofen ingestion, and reported in available literature, have (OR = 1.8), acetaminophen (OR = 1.5), and pseudoephedrine com-
not been established in epidemiological studies. bined with acetaminophen (OR = 4.2). Furthermore, fever, upper
The adverse effects of antenatal NSAIDs on the embryo, fetus respiratory infection, and allergy were not associated with gastro-
and newborn are described organ by organ in the following sec- schisis risk. These findings confirm that aspirin use in early preg-
tions. nancy increases the risk of gastroschisis; they also raise questions
about interactions between drugs and about the possible role of an
1. MISCARRIAGE infectious agent in affecting this risk.
Prenatal use of NSAIDs was found to increase the risk of mis- In another more recent study, Werler et al. [78] evaluated the
carriage [1], although conclusive evidence on this point is lacking. associations between maternal vasoactive exposures in pregnancy,
Nielsen et al. [62] reported a significant association between the as possible markers of vascular disruption, and the risk of gastro-
use of prescribed NSAIDs and miscarriage, with odds ratios that schisis. Compared to mothers of non-malformed newborns, mothers
ranged from 1.3 for NSAID use 10-12 weeks before miscarriage to of newborns with gastroschisis were more likely to report use of
7.0 for NSAID use one week before miscarriage. However, this non-aspirin NSAIDs (OR = 1.4, 95% CI = 1.1-1.7). Furthermore,
study had several limitations. In particular, no information on pa- the odds ratios for use of non-aspirin NSAIDs were greatest in the
tient compliance, or about the gestational age at time of miscarriage older women. The findings of this study, as noted by the authors,
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 5

suggest that vasoactive risk factors play a minor role in the etiology taglandins seem to play a major role), and the particular structure of
of gastroschisis and raise the question of whether inherent maternal the vessel wall [95]. In healthy term newborns, the functional clo-
factors might influence the risk. sure of the ductus occurs within 96 hours after birth. On the other
hand, complete anatomic closure with fibrosis takes up to 2-3
3. CARDIAC EFFECTS weeks. The ductus normally constricts due to the postnatal drop in
Over the last decade, many studies documented the relationship circulating prostaglandin E2 levels, as well as the rise in systemic
between antenatal maternal use of NSAIDs and the development of oxygen tension, which induce a cytochrome P450-dependent in-
congenital cardiac malformations [8,62,72,79-82]. The occurrence crease in endothelin-1 in ductal smooth muscle cells [96]. The fail-
of congenital heart defects in animal fetuses antenatally exposed to ure of this closure process with the continued patency of this fetal
NSAIDs is suggested by literature data. Cappon et al. [72] docu- channel is called patent ductus arteriosus (PDA); it is inversely
mented that inhibition of COX-1 may be involved in the disruption proportional to gestational age occurring in almost 60% of very low
of heart development, whereas the selective inhibition of COX-2 birth weight infants [95]. Persistent PDA with significant left to
appears to have no effect on heart development in rats and rabbits. right shunt in preterm infants can result in serious hemodynamic
The findings of another study on pregnant rats suggest that prenatal changes causing respiratory, gastrointestinal, cerebral, and renal
exposure to non-selective COX inhibitors, but not to selective morbidities, and increasing mortality if not promptly treated. Early
COX-2 inhibitors, could increase the risk of ventricular septal de- closure of the ductus arteriosus may be achieved pharmacologically
fects (VSD) when compared to historic controls [81]. A recent ret- using cyclooxygenase inhibitors, or by surgery. Indomethacin and
rospective analysis revealed a higher incidence of VSD in rat off- ibuprofen are the prostaglandin inhibitors mainly used to treat pat-
spring prenatally exposed to various COX inhibitors, especially ent ductus arteriosus in preterm infants.
aspirin and ibuprofen. All detected defects were limited to the Both COX-1 and COX-2 are expressed in endothelial and
membranous part of the septum and the effect was treatment- smooth muscle cells of the ductus arteriosus [1,34]. Therefore, con-
related, with the incidence and severity of VSD being significantly striction or premature closure of the ductus is a risk of all antenatal
higher in the high dose group than in the low dose group [83]. NSAIDs [97], including COXIBs [98]. In the literature, prenatal
The critical period for the ventricular septal development seems closure of the ductus arteriosus is reported after maternal admini-
to be between gestational day (GD) 8 and GD 16 (embryonic age), stration of most of the NSAIDs during pregnancy. Two animal
and includes the development of the primordial endocardium (GD studies have found that aspirin and indomethacin administered dur-
8.25-8.5), then the origin of endocardial cushion (GD 13), and fi- ing pregnancy constrict fetal ductus arteriosus [97,99]. In addition,
nally the formation of the membranous part of the ventricular sep- experimental studies documented that naproxen, similar to other
tum [84]. On this subject, it should be clarified that, in embryology, NSAIDs, readily passed the placental barrier and caused constric-
gestational age (also termed fertilisation age or embryonic age) is tion of the ductus arteriosus [89]. Two studies reported a case of
the time elapsed since conception. On the other hand, in human prenatal closure of the ductus arteriosus following maternal di-
obstetrics, gestational age is generally defined as the time elapsed clofenac therapy during the third trimester of gestation [56,100].
since 14 days prior to conception. This is approximately the dura- Paladini et al. [101] observed 10 cases of constriction of the fetal
tion between the first day of the last normal menstrual period and ductus arteriosus occurring after maternal self-medication with just
the day of delivery [85]. one or two oral doses of nimesulide. On account of these data, the
COX-1 is the only COX isoenzyme detected during the whole monitoring of the fetal ductus state and velocities by fetal echocar-
embryonic and fetal period, and could regulate prostaglandins, diography in women treated with NSAIDs seems to be appropriate
prostacyclin and thromboxane synthesis [86]. In contrast, COX-2 [100]. Moreover, each fetus should be evaluated by echocardiogra-
expression was found only in the heart of fetuses aged  GD 16 phy in case of multiple gestations, because the ductal response may
[86], when interventricular septation is completed [84]. Accord- vary between individual fetuses [102].
ingly, the prenatal COX-1 blockade and the resulting disturbances Circulating PGE2 is considered to play a role in the patency of
in eicosanoid physiology could explain the VSD occurence in ani- lamb ductus arteriosus. Therefore, the constrictive effect of
mals antenatally exposed to NSAIDs [83]. These findings have NSAIDs might be due to the inhibitory effect of PGE2 synthesis
been partially confirmed in human studies. A Swedish study [103]. In an animal study, Takizawa et al. [104] documented that
showed an increased prevalence of VSD among infants whose the increased ductal responsiveness to PGE2 in newborn rats was a
mothers had used nonsteroidal anti-inflammatory drugs (namely common response after maternal treatment with aspirin and ibupro-
diclofenac, ibuprofen, or naproxen) in early pregnancy, reporting an fen, but the catabolism of PGE2 in the lungs did not always con-
OR of 1.86 for these cardiac defects [8]. Recently, Ofori et al. [6] tribute to this response. Human studies documented a fetal ductus
have found that women prescribed NSAIDs during early gestation arteriosus constriction when aspirin [105] or other non-selective
may be at a greater risk of having children with congenital anoma- COX inhibitors were given to pregnant women to treat preterm
lies, with an adjusted OR of 3.34 (95% CI = 1.87-5.98) for the labor [106]. Recently, a retrospective analysis of fetal and neonatal
anomalies related to cardiac septal closure. These findings were echocardiographic databases by Gewillig et al. [107] revealed pre-
confirmed by other studies [80,87-89], but contrasting data were mature fetal closure of the arterial duct in 12 cases. Eight of 12
also reported [62,79,90,91]. This apparent contrast can be explained cases were diagnosed prenatally, and in 3 out of these 8 cases the
by considering that, in most of the studies, the main indication for mothers had received NSAIDs during pregnancy; in particular, 2
NSAID use was maternal fever [62,80,90-92] and that hyperthermia mothers had been treated with NSAIDs in the third and one in the
itself may alter septal development [93,94]. second trimester. Since NSAIDs are known to cause the constric-
tion of the ductus arteriosus if ingested late in pregnancy, the
4. VASCULAR EFFECTS authors hypothesized that this might have been the case in at least 2
4.1. Effects on the Ductus Arteriosus patients.
The ductus arteriosus is a vessel connecting the main pulmo- Several studies using fetal echocardiography found that the
nary trunk with the descending aorta. It is essential during fetal life effect of indomethacin on the constriction of the ductus arteriosus
to shunt blood away from the high resistance pulmonary circula- was independent of the fetal serum concentration of this drug [108-
tion, so much so that the ductal closure in utero may lead to fetal 110]. The rate of indomethacin-induced constriction of the ductus
demise or pulmonary hypertension. Fetal patency of the ductus, and arteriosus seems to be related to gestational age, increasing from
its spontaneous closure after birth, is the result of a balanced inter- 0% before week 27 of gestation to 61% during 31-34 weeks of
action of locally produced and circulating mediators (of which pros- gestation [109]. For this reason, indomethacin should not be used
6 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

beyond 31 weeks of gestation [111]. In addition, the constriction oxide synthase (due to ingrowth of vasa vasorum), neointima for-
seems to reverse frequently within 24-48 hours after cessation of mation, and loss of smooth muscle cells. Furthermore, this study
therapy [1]. documented that moderate degrees of ductus constriction in utero
Conflicting opinions are reported in the literature about antena- increased NO production, which inhibited ductus contractility,
tal NSAIDs and their effects on the ductus arteriosus in the neonate. while marked degrees of ductus constriction decreased tissue dis-
In 1990, Baerts et al. [112] documented a lower incidence of PDA tensibility and contractile capacity. The authors concluded that
in infants exposed in utero to indomethacin when compared with ductal patency was no longer controlled primarily by prostaglandins
those not exposed. Two recent metanalysis, aimed at assessing the once the ductus had been exposed to indomethacin in utero [118].
effects of tocolysis with indomethacin on neonatal outcomes, have
4.2. Persistent Pulmonary Hypertension of the Newborn
revealed no significant differences in the rates of patent ductus
(PPHN)
arteriosus between infants exposed to indomethacin antenatally and
those not exposed [59,113], confirming the findings of previous Exposure of the fetus to NSAIDs and the resulting closure of
studies [32,114,115]. In infants antenatally exposed to indometha- the ductus arteriosus in utero has been suggested to be one possible
cin, other authors [116,117] documented a surprisingly higher inci- cause of persistent pulmonary hypertension of the newborn
dence of PDA and a higher postnatal PDA severity. This was char- (PPHN). PPHN is a neonatal disorder characterized by elevated
acterized by a symptomatic PDA requiring postnatal therapy with pulmonary resistance and altered pulmonary vasoreactivity that lead
indomethacin [117] or surgical ligation because of a lack of initial to a failure of the pulmonary circulation to adapt to extrauterine
response or ductal reopening after postnatal indomethacin therapy life. This results in a right to left blood shunt across the foramen
[116]. These data were confirmed in several animal studies that ovale and the ductus arteriosus (if it is patent), thus causing severe
demonstrated a delayed postnatal closure of the ductus arteriosus in hypoxemia. NSAIDs are COX inhibitors that can block the synthe-
mice, sheep, and rats that had been exposed to nonselective COX sis of prostaglandins and thromboxane, which are involved in main-
inhibitors during the last period of gestation [118-121] and in mice taining ductal patency and regulation of pulmonary vasculature
lacking both COX genes [119,120,122]. The recent study conducted [139]. Since 1976, animal experiments demonstrated that indo-
on rats by Momma et al. [121] documented that the delay in closure methacin produced constriction or closure of the ductus arteriosus
of the ductus arteriosus induced by indomethacin was dose- with subsequent primary pulmonary hypertension due to elevated
dependent and temporary, with recovery 3 days or more after expo- pulmonary vascular resistance [140]. Three years later, Levin et al.
sure. A timing evaluation revealed a maximum delay with the ad- [141] documented that antenatal indomethacin administration
ministration of indomethacin 2 days before birth and a minimum caused ductal constriction, pulmonary arterial hypertension, and
delay with the administration of the drug 5 days before. The authors right ventricular damage in the fetus. In the newborn infant, this
suggested that the delay was induced by a decreased stimulus to the mechanism may be one cause of persistent pulmonary hypertension
prostaglandin EP(4) receptor system in the last 2 days in utero. due to vasoconstriction and increased pulmonary arterial smooth
Nevertheless, the mechanism underlying this paradoxical response muscle.
is not entirely clear. In utero COX inhibition may probably contrib- In fetal lambs, pulmonary hypertension due to antenatal me-
ute to delayed closure by increasing NO [118,123] or decreasing chanical ligation of the ductus arteriosus caused partial musculari-
hyaluronic acid production in the ductus [124]. Specific compo- zation of the intraacinar pulmonary arteries [142]. In other animal
nents of ductal contractility are involved in the postnatal closure of studies, the antenatal exposure to NSAIDs has been shown to pro-
the ductus arteriosus, namely smooth muscle calcium channels, duce ductal constriction, an increase in pulmonary arterial smooth
potassium channels [125-129], Rhokinase– related calcium sensitiz- muscle thickness, and pulmonary arterial hypertension [143-147].
ing pathways [125,130-132], mature myosin isoforms [133], and These results were not confirmed in two other studies that could not
cytoskeletal proteins [134]. In the literature, it has been suggested reproduce functional and structural changes of PPHN in newborn
that events or drugs that interfere with these pathways can lead to lambs after antenatal administration of indomethacin [148,149].
delayed postnatal ductus closure [130,131,135-137]. In a recent Ohara et al. [150] examined whether or not closing the ductus arte-
study, Reese et al. [138] have speculated that prostaglandins may riosus of fetal lambs by the administration of indomethacin in utero
regulate the development of one or more of these contractile path- could produce the functional and structural changes of PPHN. The
ways and hypothesized that in utero inhibition of COX activity may results of this study suggested that, in newborn lambs, pulmonary
alter the development of contractile apparatus, leading to a persis- hypertension due to constriction of the ductus arteriosus by antena-
tent PDA after birth. They examined the effects of prostaglandin tal indomethacin produced only a part of the structural changes of
exposure and inhibition on the fetal mice and sheep ductus, and the pulmonary vascular bed in fatal PPHN, and it failed to repro-
found that PGE2 increased the CaL- and K+-channel genes expres- duce the postnatal hemodynamics of PPHN.
sion (which regulate oxygen-induced constriction) and the phos- In a case-control interview study by Van Marter et al. [151],
phodiesterase expression (which decreases the sensitivity of the maternal intake of NSAIDs and aspirin during pregnancy or the
ductus to cAMP- or cGMP-dependent vasodilators). On the con- conditions leading to the use of these drugs were shown to contrib-
trary, chronic in utero exposure to COX inhibitors decreased these ute to an increased risk of PPHN.
expressions. These findings could explain the increased risk of
PDA after in utero COX inhibition. In fact, PGE2 seems to have It has been documented that not only long-term indomethacin
two complementary roles during ductus development: maintaining therapy but also administration of this drug even hours before de-
ductus patency for fetal well-being, and promoting the expression livery can significantly affect the ductus arteriosus and the pulmo-
of pathways necessary for its oxygen-induced closure after delivery. nary vasculature, leading to pulmonary hypertension in the prema-
The paradoxical effects of acute and chronic COX inhibition seem ture newborn [111].
to be consistent with the existence of these two roles [138]. Clyman To our knowledge, only one case-control study documented
et al. [118] hypothesized that the ductus arteriosus resistance to that indomethacin tocolysis was not associated with a higher inci-
postnatal indomethacin of newborn infants exposed to antenatal dence of pulmonary hypertension of the newborn [32].
indomethacin could be due to ductus constriction in utero, with Other NSAIDs, namely aspirin [151], diclofenac [152,153],
subsequent remodelling of the vessel. A study by these authors naproxen sodium [154], ibuprofen [61], and niflumic acid [155]
documented an increase of the thickness of the avascular zone and a were reported to be associated with an increased risk of PPHN
constriction of the ductus after infusion of fetal lambs with indo- when administered to the mother during pregnancy. Zenker et al.
methacin for 48 hours. These modifications in turn induced the [152] found that pulmonary hypertension following an antenatal
expression of vascular endothelial growth factor, endothelial nitric
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 7

maternal treatment with diclofenac only responded to high and generation of prostanoids via the action of PGHS-2 in the fetal
prolonged treatment with inhaled NO, suggesting a structural altera- brain increases the fetal HPA axis response to NMDA-mediated
tion of pulmonary vasculature induced by this drug. In 2001, Alano glutamatergic stimulation [164].
et al. [4] conducted a study to detect fetal exposure to NSAIDs by The mechanisms underlying the endothelium-dependent regula-
meconium analysis and to determine the relationship between fetal tion of cerebral circulation in human newborns are not clear due to
exposure to NSAIDs and PPHN development. The findings of this the lack of experimental data [166]. Chemtob et al. [167] docu-
study confirmed the results of previous studies, demonstrating that mented that prostaglandins exert significant effects on the cerebral
the use of NSAIDs during pregnancy, in particular aspirin, ibupro- circulation of newborn piglets. These mediators are essentially
fen, and naproxen, is underestimated by maternal history and is cerebral vasodilators, except for PGE1 which produces vasocon-
significantly associated with PPHN. striction at low dose. A subsequent study demonstrated that total
cerebral blood flow decreased significantly after the administration
5. EFFECTS ON THE BRAIN
of indomethacin, a prostaglandin H-synthase inhibitor, in newborn
Many literature studies have documented the effects of antena- piglets [168]. Parfenova et al. [166] found that COX activity in
tal NSAID administration on the central nervous development and cultured endothelial cells from microvessels from the autopsy
the neural activity. Joschko et al. [156] investigated the effects of specimens of neonatal human cerebral cortex and cerebellum led to
salicylic acid on the developing nervous system in rats in vitro, vasodilator prostanoids, prostacyclin, and PGE2 production and that
demonstrated that the neural tube was especially vulnerable and both COX-1 and COX-2 contributed to endothelial prostanoid syn-
frequently failed to close, while cellular and ultrastructural exami- thesis in the neonatal human brain.
nation revealed extensive cell death in the neuroepithelium. The
COX-2 has been shown to be the predominant form of COX
authors suggested that the extensive cell necrosis in the developing
and the main catalyst of prostaglandin synthesis in the neonatal
neuroepithelium at the site of neural tube fusion might be involved
brain. An animal study by Li et al. [169] demonstrated a key role
in failed neurulation, while necrosis at other sites in the cranial
for COX-2 in the regulation of PGE2 and PGF2alpha receptors and
neuroepithelium might be linked with previously reported intellec-
their functions in the neonatal cerebral vasculature. Najaran et al.
tual and behavioural abnormalities. A more recent study reported a
[170] documented that high levels of PGs in the nervous tissue,
decrease in the total brain weight, cerebrum and cerebellum length
specifically PGE2 acting apparently via EP2 receptors, seemed to
and width in rats antenatally exposed to high dose of aspirin [157].
contribute to the preservation of electrophysiological neural func-
A pronounced neuron loss in cornu ammonis and dentate gyrus of
tion in newborn pigs subjected to hypoxic events in the perinatal
the hippocampus was observed in a diclofenac sodium-treated
period. This effect of PGE2 was unrelated to improvements in cere-
group of rats, probably due to the neurotoxicity of this drug on the
bral blood flow. The same study revealed that Ibuprofen and Di-
developing hippocampal formation [158]. Another study showed
clofenac were able to decrease brain PG levels. On the basis of their
that the total number of Purkinje cells in the offspring of diclofenac
results, the authors suggested that the reduction in PG levels would
sodium-treated rats was significantly lower than in the offspring of
enhance the posthypoxic deterioration in neural function of the
control animals, suggesting that the Purkinje cells of a developing
perinate. Other authors [33, 171,172] compared the effects of in-
cerebellum might be affected by the administration of this drug
domethacin and ibuprofen on cerebral hemodynamics in preterm
during the prenatal period [159]. These data are in contrast with the
infants: ibuprofen did not significantly reduce cerebral perfusion
findings of Odaci et al. [160] that documented an increased number
and oxygen availability, while indomethacin affected cerebral blood
of Purkinje cells in the cerebellum of a developing female rat prena-
flow velocity.
tally exposed to the same drug.
A recent study by Marret et al. [161] has shown that low-dose 5.1. Cystic Periventricular Leukomalacia (cPVL)
aspirin given during pregnancy is not significantly associated with Cystic periventricular leukomalacia (cPVL), the principal
cerebral lesions, cerebral palsy, or global cognitive impairment in ischemic brain injury in premature infants, is characterized by the
5-year-old children. Moreover, the results of this study have sug- necrosis of white matter in the periventricular region and represents
gested that aspirin could be associated with a reduction in neurobe- the major neuropathology for spastic motor deficits in cerebral
havioral difficulties. palsy or epilepsy. Recent reports strongly suggest that the brain
The effects of NSAIDs on the development of peripheral nerves injury associated with cPVL may have already occurred in utero
are still not clarified. A recent study by Canan et al. [162] has [173].
shown that the axon number of sciatic nerve and mean axon cross- Antenatal NSAIDS are reported to increase the risk of cPLV in
sectional area, but not average myelin sheet thickness, are signifi- many studies.
cantly decreased in rats antenatally exposed to diclofenac sodium
compared with the control group; moreover, electron microscope In an old study, the incidence of periventricular leukomalacia
analysis has revealed a deterioration of myelin sheaths. Accord- was found to be increased among infants born before 30 weeks’
ingly, the authors have suggested that the prenatal administration of gestation and exposed to any tocolytic agent, and cystic lesions
diclofenac sodium impairs peripheral nervous system development. occurred more commonly in those exposed to indomethacin [112].
Decreases in fetal blood pressure stimulate homeostatic stress More recent studies, which were conducted in cohorts of pre-
responses which help the blood pressure to return to normal values. mature infants, confirmed this finding and showed that antenatal
In the fetus, hypothalamus-pituitary-adrenal (HPA) axis responses exposure to indomethacin increased the risk of cPVL occurrence by
to hypotension are mediated by chemoreceptor and baroreceptor 2-3 times. These data support a growing consensus that cPVL is
reflexes and central nervous system ischemia [163]. HPA response often the result of maternal and fetal factors as well as antenatal
is controlled by prostaglandins generated within the fetal brain treatments [59,173].
[164]. Indomethacin, a nonselective inhibitor of prostaglandin en- 5.2. Periventricular/Intraventricular Hemorrhage (PVH/IVH)
doperoxide synthase (PGHS)-1 and -2, attenuates the HPA response
to hypotension in the fetal sheep [165]. Very recently, Wood et al. Periventricular/intraventricular hemorrhage (PVH/IVH) is a
[163] have demonstrated that the activity of PGHS-2 in the brain is multifactorial pathological condition which is frequently associated
a necessary component of the HPA response to cerebral hypoperfu- with neurological morbidity and mortality in very low birth weight
sion in the sheep fetus. Furthermore, the blockade of N-methyl-d- premature infants, and therefore its prevention is particularly impor-
aspartate (NMDA) receptors seems to reduce the fetal ACTH re- tant.
sponse to cerebral hypoperfusion. A recent study has shown that the
8 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

Predisposing factors include an immature vasculature of germi- associated with abnormal neurological outcome. The findings of
nal matrix, a pressure-passive cerebral circulation, and hemody- this study showed that indomethacin, used as a tocolytic agent, was
namic perturbations in sick premature infants. Intact cerebral not associated with adverse neurodevelopmental outcome at 16–24
autoregulation has been documented in stable premature infants; by months corrected age in infants  29 weeks’ gestational age; fur-
contrast, it functions within a limited blood pressure range and is thermore, antenatal indomethacin exposure might be associated
likely to be absent in the sick hypotensive infant, thus increasing with an improved neurological outcome. It is noteworthy that Sa-
the risk for PVH-IVH with perturbations in blood pressure. The risk lokorpi et al. [181] performed their study when antenatal steroids
for PVH/IVH is markedly increased in the absence of antenatal were not routinely used, while the study by Al-alaiyan et al. [182]
glucocorticoid exposure in intubated infants with birth weight less failed to account for possible confounders. Conversely, the strength
than 1000 g affected by respiratory distress syndrome [174]. of the study by Amin et al. [183] consists in the evaluation of the
The role of antenatal use of NSAIDs on the incidence of neona- effect of antenatal steroids and the adjustment for possible con-
tal PVH/IVH remains to be elucidated. founders. In fact, the beneficial effects of antenatal steroids should
be considered as well, since these effects might outweigh the risk
Vermillion et al. [32] documented that maternal indomethacin associated with the use of antenatal indomethacin. Moreover, pre-
exposure immediately before delivery was not associated with an vious findings have shown that the benefits of antenatal indometha-
increased risk for grade III/IV intraventricular hemorrhage in in- cin for tocolysis outweigh the potential risks to the newborn at ges-
fants delivered between 24 and 32 weeks' gestation. This finding tational ages < or = 32 weeks [184].
was confirmed in a more recent study showing that indomethacin
tocolysis was not associated with an increased risk for intraven- 6. EFFECTS ON THE KIDNEY
tricular hemorrhage [175]. On the other hand, Ojala et al. [58]
Renal function in postnatal life is affected by prenatal environ-
documented that antenatal indomethacin treatment for longer than 2
mental and genetic determinants. The number of pregnant women
days, with a daily or cumulative dosage >/=150 mg, was correlated
who are receiving drugs administered when fetal glomerulogenesis
with a significantly higher incidence of grade I-II intraventricular
is still occurring is increasing [185]. Several drugs have been shown
hemorrhage in infants of gestational ages between 23 and 32 weeks.
to produce adverse effects on the kidneys, mainly when exposure
Furthermore, Doyle et al. [115] showed that the incidence of grades
occurs during active nephrogenesis [186]. Mothers of infants that
III to IV IVH was significantly higher in VLBW infants exposed to
showed neonatal acute renal failure had received more nephrotoxic
antenatal indomethacin as compared to the non-exposed infants;
drugs during pregnancy and delivery [187]. Such adverse effects
moreover, antenatal indomethacin exposure was significantly asso-
may occur particularly when fetuses are exposed to NSAIDs [188].
ciated with an increased incidence of IVH after controlling for an-
A review article by Bavoux [189] reported the renal effects of ante-
tenatal corticosteroids, maternal pre-eclampsia, gestational age and
natal NSAIDs (acute renal failure with edema, oliguria, hyponatre-
birth weight.
mia and hyperkalemia) in 23 neonates, 8 of whom died. Drukker et
Some studies have shown that antenatal aspirin is associated al. [190] have recommended close attention to the use of NSAIDs
with an increased incidence of intracranial hemorrhage. In particu- during pregnancy because these agents impair renal structure and
lar, the use of aspirin in the last 3 months of pregnancy has been function in the unborn fetus.
suggested to be highly questionable [176,177]. Nevertheless, it
The nephrotoxic effects of NSAIDs are related to the block of
should be considered that aspirin use during pregnancy is particu-
prostaglandin synthesis through the inhibition of cyclooxygenase-2
larly useful for the prevention of intrauterine growth restriction
[191]. Prostaglandin E2 (PGE2) modulates both intrarenal vascular
(IUGR) and preeclampsia, and for the treatment of antiphospholipid
tone and salt and water excretion. In particular, PGE2 contributes to
antibodies syndrome (APAs). In the pathogenesis of both IUGR
regulate renal perfusion and glomerular filtration rate in virtue of its
and preeclampsia, the activation of platelets seems to play an im-
vasodilating properties, by counteracting the action of vasoconstric-
portant role, and therefore an antiplatelet agent such as aspirin may
tive substances such as angiotensin II, catecholamines, vasopressin
be useful in preventing these two pregnancy complications [1].
and endothelin [9].
Additionally, low-dose aspirin combined with low-molecular-
weight heparin has been found to be the most effective treatment Several cases of severe and sometimes irreversible renal failure
for pregnant women with APAs [178]. The aspirin mechanism of have been reported in neonates exposed to indomethacin prenatally
action is related to its ability to increase the level of prostacyclins [191]. In a case of twin-to-twin transfusion syndrome, renal tubular
and to reduce the level of thromboxane, the platelets prothrombotic dysgenesis-like lesions and hypocalvaria which were found in twins
factor, thus preventing placental thrombosis which is a possible were attributed to indomethacin treatment of the mother for acute
complication of antiphospholipid syndrome [179]. polyhydramnios [192]. Infants whose mothers had received indo-
methacin tocolysis were shown to be at risk of developing renal
The exact pathophysiology of IVH related to the antenatal ex-
impairment [193-194]. Jacqz-Aigrain et al. [195] reported that 22%
posure to NSAIDs is unclear. In pregnant women undergoing toco-
of live-born neonates of pregnant women receiving indomethacin
lysis with indomethacin, Parilla et al. [180] have documented that
for treatment of premature labor or polyhydramnios developed
this drug does not significantly affect fetal cerebral blood flow.
renal insufficiency. Van der Heijden et al. [196,197] reported 6
Based on this finding, the authors have concluded that, if antenatal
anuric neonates exposed to indomethacin in utero. In 5 infants,
indomethacin in the preterm fetus increases the risk of intraven-
cystic dilatations of superficial nephrons were associated with
tricular hemorrhage, it seems to be due to another mechanism.
ischemic changes of the deep cortex.
5.3. Neurodevelopmental Outcome Comparative studies are available in the literature. Renal func-
There are limited data on the long-term neurodevelopmental tion was measured during the first 4 postnatal days in 9 preterm
outcome of premature infants exposed to antenatal indomethacin. In neonates exposed to indomethacin during the last 2 days of preg-
a follow-up study, Salokorpi et al. [181] reported that antenatal nancy and compared with controls. Inulin clearance was lower and
indomethacin was not significantly associated with abnormal neuro- serum creatinine was higher in patients than in controls [198]. Pre-
logical outcome in premature infants when evaluated at 18 months term infants whose mothers had been treated with indomethacin for
corrected age. Similarly, in a small retrospective cohort study, Al- preterm labor, matched with infants whose mothers had not re-
alaiyan et al. [182] found that there were no effects of antenatal ceived indomethacin, had a lower urine output and higher serum
indomethacin on neurodevelopmental outcome at 12 months of age. creatinine concentrations during the first three days after delivery
Recently, a retrospective cohort study by Amin et al. [183] was [116]. Similarly, the study by Butler-O'Hara et al. [193] found that
conducted to evaluate if the exposure to antenatal indomethacin was infants whose mothers had received indomethacin for tocolysis
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 9

were more likely to have elevated serum creatinine than matched Voyer et al. [209] reported the case of a female neonate, born at
controls. Van Den Anker et al. [199] documented the negative ef- 37 weeks of gestation, who presented with respiratory distress syn-
fect of prenatal indomethacin administration on the renal function drome, right pneumothorax and anuria. Family history was unre-
of preterm infants in the first days of life. In particular, they found markable, except that her mother had received piroxicam at about
that prenatal exposure to this agent resulted in a significant and 26 weeks of gestation; moreover, a sonogram at 28 weeks of gesta-
transient reduction in glomerular filtration rate (GFR) values at 3 tion had shown oligohydramnios. Peritoneal dialysis was started on
days after birth. On the other hand, Vieux et al. [200] found low the 14th day of life because of renal insufficiency, but the neonate
GFR values at 7 days of life in a group of very preterm infants died on the 33rd day. Renal histopathology showed crowded
treated with ibuprofen during the first week of life. To document glomeruli and only few differentiated proximal convoluted tubules
postnatal trends and covariates of creatininemia, George et al. [201] in the inner cortex, abnormally differentiated microcystic tubules
linked maternal characteristics, characteristics at delivery and dur- and microcystic glomeruli in the outer cortex. Electron microscopy
ing neonatal stay (postnatal ibuprofen among others) with creatin- revealed the presence of brush border remnants and other proximal
inemia observations in 151 extremely low birth weight (ELBW) tubular characteristics.
neonates in the first 6 weeks of life. ELBW infants exhibited trends Acute kidney failure was observed at birth in a premature neo-
similar to heavier neonates, but peak creatininemia was higher (val- nate and in twin infants prenatally exposed to ketoprofen during the
ues above the 75th percentile) and the subsequent decrease was last days of pregnancy [210,211].
slower (values remained elevated until day 28). With the monovari-
ate analysis, raised creatininemia in ELBW infants was shown to Coadministration of drugs, such as aminoglycosides and
reflect immaturity (lower gestational age), morbidity (lower Apgar NSAIDs, can worsen the outcome of nephrotoxicity. The risk of
score, longer postnatal ventilation and oxygen need, higher inci- drug interactions between aminoglycosides and NSAIDs appears to
dence of retinopathy of prematurity and intraventricular hemor- be greater in young children than in neonates and infants [212,213].
rhage) and drug treatments (ibuprofen, postnatal steroids). With In addition to functional adverse effects, the intrauterine expo-
regard to the latter, a negative effect of ibuprofen exposure on peak sure to NSAIDs may affect renal structure itself and produce renal
creatininemia was reported, with an impact on creatinine clearance congenital abnormalities, including cystic dysplasia, tubular dys-
that lasted beyond the days of ibuprofen treatment. Moreover, when genesis, ischemic damage and a reduced number of nephrons
all risk factors were entered in a multiple regression model, ibupro- [188,190].
fen treatment and immaturity remained the predominant independ- Balasubramaniam [214] reported the case of a male neonate
ent covariates of creatininemia. who presented with oliguria and renal failure from birth, but with-
The neonatal and neurodevelopmental outcome of fifteen pre- out any respiratory distress. Family history revealed that his mother
term infants exposed antenatally to indomethacin was investigated had received nimesulide from week 30 of pregnancy, for 8 weeks.
by comparison with 15 controls not exposed to antenatal indo- Peritoneal dialysis was started on the fourth day of life. Although
methacin. No neonatal complications attributable to the antenatal there was an increase in urine output over the next few days, renal
use of indomethacin were reported; in particular, mean creatinine, failure persisted. Needle biopsy of the kidneys, which was done on
BUN, and urine output for the first 3 days of life were similar in the the eighth day, showed partial abnormal tubular differentiation with
two groups [182]. subacute tubulointerstitial nephritis.
No data are available on the nephrotoxic effect of antenatal An unusual case of neonatal anuria owing to renal tubular dys-
ibuprofen in the fetus and newborn. Administration of ibuprofen to genesis with atypical histology was reported after in-utero exposure
preterm neonates on the first day of life to enhance closure of an to naproxen sodium [215].
asymptomatic PDA was shown to reduce amikacin and vancomycin The measurement of urinary biomarkers in preterm and term
clearance in the first month of postnatal life [202]. Recently, Iaco- infants might constitute a novel approach for the prevention and
belli et al. [203] have investigated maternal and neonatal factors early detection of NSAID-induced nephrotoxicity [9,216,217].
affecting postnatal changes in serum creatinine in preterm infants
with gestational age <32 weeks. Lower gestational age (P<0.0001) 7. EFFECTS ON THE LUNG
and ibuprofen-treated patent ductus arteriosus (P<0.0001) were the Postnatal indomethacin administration was shown to alter lung
main analyzed factors independently associated with higher serum structure in rats: in particular, diminished alveolar air, increased
creatinine peak during the first week of life. In infants with hemo- alveolar duct air, increased mean linear intercept (gas-exchanging
dynamically significant PDA, the authors found that serum creatin- wall distance), diminished gas-exchanging surface area and surface-
ine before ibuprofen administration was higher compared to gesta- to-volume ratio, increased septal wall thickness, diminished number
tional age matched controls, thus suggesting that a renal impairment of alveolar crests, and an increased number of lamellar bodies in
due to PDA and/or associated conditions existed before the initia- alveolar type II cells were found [218]. A recent study has docu-
tion of NSAID therapy. On the other hand, it has been shown that mented that the use of prophylactic indomethacin in newborns with
ibuprofen treatment markedly reduces (59.4%) urinary PGE(2) and a birthweight lower than 1250 grams increases the risk of develop-
may alter renal function in preterm infants with PDA [204]. ing bronchopulmonary dysplasia (BPD) [219]. Conflicting opinions
Maternal consumption of nimesulide has been shown to be are reported on the association between the antenatal indomethacin
responsible for neonatal renal failure [194]. Several case reports administration and the risk of bronchopulmonary dysplasia devel-
showed acute, reversible and irreversible renal failure in term and opment [116,220].
preterm newborns whose mothers had received therapeutic doses of
nimesulide during the last weeks of pregnancy [205,206]. A case of 8. EFFECTS ON THE SKELETON
neonatal renal failure associated with maternal ingestion of Nime- It has been reported that NSAIDs suppress bone repair and
sulide in the third trimester of pregnancy was reported by Ali et al. remodeling, but only mildly inhibit bone mineralization at the ear-
[207]. In particular, this newborn presented with metabolic acidosis lier stage of the repair process. In 1999, Ho et al. [221] conducted a
and non-oliguric renal failure on the second day of life, and showed study on fetal calvaria to investigate whether NSAIDs affected the
histopathological evidence of renal tubular dysgenesis. proliferation and/or differentiation of osteoblasts and whether these
Maternal administration of niflumic acid, a COX-2 inhibitor, effects were prostaglandin-mediated. The results of this study sug-
during the last days of gestation was demonstrated to induce fetal gested that the inhibitory effect of ketorolac on osteoblastic prolif-
and neonatal renal failure [208]. eration contributed to its suppressive effects on bone repair and
remodeling in vivo, and that the effects of ketorolac and indometha-
10 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

cin on cell proliferation and differentiation could not be mediated was similar to that of the placebo group. On the other hand, this
by the inhibition of the synthesis of PGE1 or PGE2. Furthermore, study did not reveal any relationship between prenatal indometha-
PGEs and NSAIDs seemed to be involved in matrix maturation and cin exposure and perforation.
biologic bone mineralization in the earlier stage of osteoblast dif- In 1988, Vanhaesebrouck et al. [236] reported three preterm
ferentiation. infants antenatally exposed to indomethacin who developed a char-
More recently, Chang et al. [222] have investigated the influ- acteristic syndrome consisting of renal failure and ileal perforation
ences of NSAIDs on cell cycle kinetics, cytotoxicity, and cell death in the first postnatal days. A recent analysis suggests that antenatal
pattern in osteoblast cultures from rat fetal calvaria. They docu- indomethacin may be a risk factor for SIP, particularly when given
mented that NSAIDs arrested cell cycle at the G(0)/G(1) phase and close to birth [229]. In addition, antenatal indomethacin has been
induced cytotoxicity and cell death of osteoblasts, contributing in found to be more likely associated with spontaneous intestinal per-
this manner to the suppressive effect on bone formation. Among foration rather than NEC [237]. In contrast, a prospective preva-
studied NSAIDs, piroxicam showed the least effect in producing lence study by Sharma et al. [238] has revealed that VLBW infants
osteoblastic dysfunction. Moreover, the cytotoxic and apoptotic prenatally exposed to indomethacin do not show an increased rate
effects of NSAIDs on osteoblasts did not seem to be prostaglandin- of neonatal gut injury.
related.
9.2. Necrotizing Enterocolitis (NEC)
In another study conducted on fetal rats, the effects of non-
selective NSAIDs (indomethacin, ketorolac, diclofenac and piroxi- Necrotizing enterocolitis (NEC) is an acquired gastrointestinal
cam) and COX-2 selective inhibitors (celecoxib and DFU, an ana- disease associated with significant morbidity and mortality in pre-
log of rofecoxib) on cell proliferation and death were investigated maturely born neonates [239]. There are conflicting opinions con-
in cultured epiphyseal-articular chondrocytes. Therapeutic concen- cerning the role of antenatal NSAIDs in the pathogenesis of NEC.
trations of non-selective NSAIDs were found to cause proliferation Many studies have reported that antenatal indomethacin expo-
suppression and cell death of chondrocytes by PG-independent sure is not associated with a significant increase in the incidence of
mechanisms. As suggested by the authors, these adverse effects necrotizing enterocolitis [32,113-115]. Other studies have shown
may be one of the reasons that could explain the delay in the en- that antenatal indomethacin may be associated with an increased
dochondral bone formation observed in previous studies. On the risk of necrotizing enterocolitis in premature infants [59,116].
other hand, COX-2 selective inhibitors exhibited less adverse ef- Major et al. [57] documented that the incidence of NEC in
fects on chondrocytic proliferation and death [223]. newborns delivered within 24 hours of maternal indomethacin ther-
In 2008, Burdan et al. [224] showed a decrease of vertebra apy for tocolysis was 20% compared with 9% in the non-exposed
mineralization in fetuses of rats prenatally exposed to COX inhibi- group (p = 0.005). In the same study, the incidence of NEC in neo-
tors. Unlike DFU, which is a selective COX-2 inhibitor, the exam- nates with more than 48 hours of antenatal indomethacin exposure
ined nonselective COX inhibitors were shown to decrease fetal was 26.4% compared with 4.1% in those with less than 48 hours of
bone mineralization when administered in high maternal toxic indomethacin exposure. The authors concluded that antenatal in-
doses. A decrease in fetal length has been the only sign of toxicity domethacin exposure occurring within < or = 24 hours of delivery
found after exposure to DFU [225]. Other authors [226] reported an and of at least 48 hours' duration was associated with a significant
inhibitory effect of parecoxib, a COX-2 selective inhibitor, on the increase in the incidence of necrotizing enterocolitis in low birth
mineralization of fracture callus after a tibial fracture in rats, with weight neonates.
this effect persisting for a longer period compared to indomethacin.
Recently, maternal toxicity has been found to be an important 10. EFFECTS ON Limph nodes
factor affecting prenatal skeletal development during in utero expo- A dose-dependent retardation in the formation of main struc-
sure to NSAIDs [225]. tures of the mesenteric lymph nodes was documented in rats ante-
Finally, dose-related skeletal anomalies, including fused rib, natally treated with indometacin [240,241]. In particular, a retarda-
incomplete ossification of the cervical arch, absent/hemicentric tion in the development of capsule, sinuses and reticular fibers of
body of thoracic or lumbar vertebra, deformation of lumbar arch, the node was observed, as well as a decrease in lympho- and plas-
and absent sacral arch, have been described in rats prenatally ex- mocytopoiesis [241].
posed to acetylsalicylic acid [227]. CONCLUSIONS
9. EFFECTS ON THE GASTROINTESTINAL TRACT Non-steroidal anti-inflammatory drugs are among the common-
9.1. Spontaneous Intestinal Perforation (SIP) est drugs prescribed to pregnant women. In general, fever, pain and
inflammation are frequent indications for NSAID treatment during
Spontaneous intestinal perforation (SIP) commonly occurs in pregnancy. In addition, these agents are used in pregnant women
extremely low-birth-weight (ELBW) infants. Early postnatal indo- not only to treat inflammatory bowel diseases and chronic rheu-
methacin exposure was identified as one of the risk factors for SIP matic diseases, but also in the management of obstetrical complica-
[228-230]. This drug seems to play a causative role in the etiology tions such as preterm labor and polyhydramnios. The real consump-
of SIP through its effects on ileal trophism and motility [229], by tion of NSAIDs during pregnancy remains unknown, considering
decreasing mesenteric blood flow, blocking autoregulation of ter- that self- medication is possible as some of these drugs are available
minal ileum oxygen consumption and altering defense mechanisms over-the-counter.
in the neonatal gastrointestinal tract. [231-234].
Several studies have provided evidence that NSAIDs can cross
Watterberg et al. [235] conducted a multicentric trial to evalu- the human placenta, and so reach the fetal circulation. Accordingly,
ate the effect of postnatal low-dose hydrocortisone therapy to pre- a spectrum of adverse effects may occur in the offspring of women
vent BPD in extremely premature infants, assuming that the in- treated with NSAIDs during pregnancy Table 1. These effects are
creased inflammation observed in infants developing BPD may dependent on the type of agent, the dose and duration of therapy,
partly reflect inadequate adrenal function. Hydrocortisone-treated the period of gestation, and the time elapsed between maternal
infants who received indomethacin were found to be more likely to NSAID administration and delivery. The main pharmacological
experience apparently spontaneous gastrointestinal perforation action of these drugs on the fetus seems to be mediated by the inhi-
compared with placebo-treated infants who received indomethacin, bition of prostaglandin synthesis.
suggesting an interactive effect. The incidence of perforation in
hydrocortisone-treated infants who did not receive indomethacin
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 11

Table I. Adverse Effects of Antenatal NSAIDs on the Embryo, Fetus and Neonate (Human Studies)

Organ Adverse Effect Type of NSAID References

Heart Cardiac defects (VSD) naproxen; NSAIDs Ericson 2001; Ofori 2006

Prenatal closure of the ductus


diclofenac; nimesulide Rein 1999; Auer 2004; Paladini 2005
arteriosus

Blood vessels PDA indomethacin Norton 1993; Hammerman 1998

NSAIDs; aspirin; naproxen sodium;


PPHN Van Marter 1996; Talati 2000; Tarcan 2004; Siu 2004
indomethacin; diclofenac

PVL/cPVL indomethacin Baerts 1990; Murata 2005; Amin 2007


Brain
PVH/IVH aspirin; indomethacin Rumack 1981; Norton 1993; Ojala 2000; Doyle 2005

ketoprofen; indomethacin; niflumic Gouyon 1991; Jacqz-Aigrain 1993; Alessandri 1994; Van
Acute renal failure acid; ibuprofen; nimesulide; di- der Heijden 1994; Van der Heijden 1995; Balasubramaniam
Kidney clofenac; 2000; Fieni 2004; Benini 2004; Ali 2006

piroxicam; indomethacin; nimesu- Voyer 1994; Van der Heijden 1994; Balasubramaniam 2000;
Renal congenital abnormalities
lide; naproxen sodium Robin 2000; Koklu 2006; Ali 2006

Lung BPD indomethacin Eronen 1994

Skeleton skeletal deformity aspirin Chakravarty 1993

Gastrointestinal SIP indomethacin Gordon 2008


tract NEC indomethacin Norton 1993; Major 1994; Ojala 2000; Amin 2007

Drongowski 1991; Werler 1992; Torfs 1996; Martínez-Frías


Abdominal wall Celosomy aspirin, ibuprofen
1997; Werler 2002

Eye Cyclopia aspirin Agapitos 1986

Mouth Cleft lip and/or cleft palate naproxen; NSAIDs Ericson 2001

Miscarriage NSAIDs; aspirin Nielsen 2001; Li 2003

piroxicam; niflumic acid; indo-


Voyer 1994; Alessandri 1994; Van der Heijden 1995; Benini
Oligohydramnios methacin; ibuprofen; diclofenac;
Others 2004; Fieni 2004; Balasubramaniam 2000
nimesulide; ketoprofen

Neonatal bleeding aspirin Stuart 1982

Congenital malformations NSAIDs Ofori 2006

Abbreviations
VSD, ventricular septal defects. PDA, patent ductus arteriosus. PPHN, persistent pulmonary hypertension of the newborn. PVL, periventricular leukomalacia. PVH-IVH, periven-
tricular/intraventricular hemorrhage. BPD, bronchopulmonary dysplasia. SIP, spontaneous intestinal perforation. NEC, necrotizing enterocolitis.

The period of gestation in which NSAIDs are given is a major intrauterine exposure to NSAIDs may affect renal structure itself
factor in influencing embryo-fetal effects. For example, miscarriage and cause congenital abnormalities of the kidney. The nephrotoxic
and congenital anomalies (particularly cleft palate, congenital heart effects of NSAIDs are related to the block of prostaglandin synthe-
defects and abdominal wall defects) have been described after sis through the inhibition of cyclooxygenase-2.
treatment with NSAIDs during early gestation, even though the risk Currently, there are limited data on the use of COX-2 selective
of these adverse pregnancy outcomes appears to be small; on the inhibitors during pregnancy and thus these agents should be
other hand, renal and vascular effects (prenatal closure of the ductus avoided in pregnant women.
arteriosus and PPHN) have been observed in fetuses of mothers
treated with NSAIDs during the third trimester of gestation. It is A considerable amount of caution should be used in interpreting
noteworthy that women attempting to conceive should not use any the results of the studies included in this review. In fact, many of
prostaglandin synthesis inhibitors, because of animal data that indi- them have been performed in animal models, while no conclusive
cate that these agents block blastocyst implantation [64]. evidence exists regarding the effects of antenatal NSAIDs on the
human fetus. Furthermore, the retrospective design of some studies
Embryo-fetal toxicity associated with antenatal NSAIDS may may limit the reliability of their results. Finally, prematurity and
affect several organ systems including the brain, cardiovascular low birth weight may be confounding factors in some of the studies
system, lung, skeleton and gastrointestinal tract. However, renal examined in this review.
involvement in the fetus and newborn is of particular concern: in
addition to functional adverse effects (neonatal renal failure), the
12 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

In conclusion, further human studies are required to clarify the neutrophil adhesion. Proc. Natl. Acad. Sci. U S A, 1998, 95, 14540-
toxic effects of antenatal NSAIDs (nonselective COX inhibitors) 14545.
and COXIBs (selective COX-2 inhibitors) on the embryo, fetus and [17] Díaz-González, F.; González-Alvaro, I.; Campanero, M.R.; Mol-
newborn. linedo, F.; del Pozo, M.A.; Munoz, C.; Pivel, J.P.; Sinchez-Madrid,
F. Prevention of in vitro neutrophil-endothelial attachment through
Physicians should be aware of the possible embryo-fetal toxic- shedding of L-selectin by nonsteroidal antiinflammatory drugs. J.
ity when prescribing non-steroidal anti-inflammatory drugs to preg- Clin. Invest., 1995, 95, 1756-1765.
nant women. The judicious use of these medications is particularly [18] Nathan, C.; Xie, Q.W. Nitric oxide synthases: roles, tolls, and
important in pregnant women at risk of preterm delivery since controls. Cell, 1994, 78(6), 915-918.
NSAID-induced adverse effects on the fetus and newborn might [19] Schmidt, H.H.; Walter, U. NO at work. Cell , 1994, 78, 919-925.
occur concurrently with the complications of prematurity. In addi- [20] Hawkey, C.J. Future treatments for arthritis: new NSAIDs, NO
NSAIDs, or no NSAIDs? Gastroenterology, 1995, 109, 614-616.
tion, a correct and complete information to women who are preg- [21] Amin, A.R.; Vyas, P.; Attur, M.; Leszczynska-Pizlak, J; Patelt,
nant, or are planning a pregnancy, is required with regard to em- I.R.; Weissmann, G; Abramson, S.B. The mode of action of aspi-
bryo-fetal and neonatal risks associated with consumption of ante- rin-like drugs: effect on inducible nitric oxide synthase. Proc. Natl.
natal NSAIDs, considering that some of these drugs are available Acad. Sci. U S A, 1995, 92, 7926-7930.
over the counter and thus can be easily purchased without medical [22] Suleyman, H.; Halici, Z.; Cadirci, E.; Hacimuftuoglu, A.; Bilen, H.
prescription. Indirect role of b2-adrenergic receptors in the mechanism of anti-
inflammatory action of NSAIDs. J. Physiol. Pharmacol., 2008,
CONFLICT OF INTEREST 59(4), 661-672.
[23] Skinner, K.A.; Challis, J.R.G. Changes in the synthesis and me-
The authors have nothing to disclose related to this article. tabolism of prostaglandins by human fetal membranes and decidua
at labour. Am. J. Obstet. Gynecol., 1985, 151, 519–523.
REFERENCES [24] Hirst, J.J.; Teixeira, F.J.; Zakar, T.; Olson, D.M. Prostaglandin
[1] Østensen, M.E.; Skomsvoll, J.F. Anti-inflammatory pharmacother- endoperoxide-H synthase-2 expression increases in human gesta-
apy during pregnancy. Expert Opin. Pharmacother., 2004, 5, 571- tional tissues with spontaneous labour onset. Reprod. Fertil. Dev.,
580. 1995, 7, 633–637.
[2] Rahimi, R.; Nikfar, S.; Rezaie, A.; Abdollahi, M. Pregnancy out- [25] Slater, D.M., Berger, L.; Newton, R.; Moore, G.E.; Bennett, P.R.
come in women with inflammatory bowel disease following expo- Expression of cyclo-oxygenase types-1 and 2 in human fetal mem-
sure to 5-aminosalicylic acid drugs: a meta-analysis. Reprod. Tos- branes at term. Am. J. Obstet. Gynecol., 1995, 172, 77–82.
sicol., 2008, 25(2), 271-275. [26] Slater, D.M.; Dennes, W.J.B.; Campa, J.S.; Poston, L.; Bennett,
[3] Vroom, F.; van den Berg, P.B., de Jong-van den Berg, L.T. Pre- P.R. Expression of cyclo-oxygenase types-1 and -2 in human
scribing of NSAIDs and ASA during pregnancy; do we need to be myometrium throughout pregnancy. Mol. Hum. Reprod., 1999, 5,
more careful? Br. J. Clin. Pharmacol., 2008, 65, 275-276. 880–884.
[4] Alano, M.A.; Ngougmna, E.; Ostrea, E.M. Jr, Konduri, G.G. [27] Smieja, Z.; Zakar, T.; Waton, J.C.; Olson, D.M. Prostaglandin
Analysis of nonsteroidal antiinflammatory drugs in meconium and endoperoxide synthase kinetics in human amnion before and after
its relation to persistent pulmonary hypertension of the newborn. labour at term and following preterm labour. Placenta, 1993, 14,
Pediatrics, 2001, 107(3), 519-523. 163–175.
[5] Tillett, J.; Kostich, L.M.; VandeVusse, L. Use of over-the-counter [28] King, J.; Flenady ,V.; Cole, S.; Thornton, S. Cyclo-oxygenase
medications during pregnancy. J. Perinat. Neonatal. Nurs., 2003, (COX) inhibitors for treating preterm labour. Cochrane database
17, 3-18. syst. rev., 2005, 2, 18, CD001992.
[6] Ofori, B.; Oraichi, D.; Blais, L.; Rey, E.; Bérard, A. Risk of con- [29] Haas, D.M.; Imperiale, T.F.; Kirkpatrick, P.R.; Klein, R.W.;
genital anomalies in pregnant users of non-steroidal anti- Zollinger, T.W.; Golichowski, A.M. Tocolytic therapy: a meta-
inflammatory drugs: A nested case-control study. Birth Defects analysis and decision analysis. Obstet. Gynecol., 2009, 113(3), 585-
Res. B. Dev. Reprod. Toxicol., 2006, 77(4), 268-279. 594.
[7] Olesen, C.; Steffensen, F.H.; Nielsen, G.L.; de Jong-van den Berg, [30] Keirse M.J.N.C. Indomethacin tocolysis in preterm labor. In: Enkin
L.; Olsen, J.; Sørensen, H.T. Drug use in first pregnancy and lacta- M.W., Keirse M.J.N.C.; Renfrew M.J.; Neilson J.P.; (eds). Preg-
tion: a population-based survey among Danish women. The EU- nancy and Childbirth Module “Cochrane Database of Systemic re-
ROMAP group. Eur. J. Clin. Pharmacol., 1999, 55, 139-144. views” Review 04383, 14 August 1992. Oxford: Cochrane Updates
[8] Ericson, A.; Källén, BA. Nonsteroidal anti-inflammatory drugs in on Disk. Update software, 1994, disk issue 1.
early pregnancy. Reprod. Toxicol., 2001, 15(4), 371-375. [31] Moise, K.J. Jr; Ou, C.N., Kirshon, B.; Cano, L.E.; Rognerud, C.;
[9] Antonucci, R.; Cuzzolin, L.; Arceri, A.; Fanos, V. Urinary prosta- Carpenter, R.J. Jr. Placental transfer of indomethacin in the human
glandin E2 in the newborn and infant. Prostaglandins Other Lipid pregnancy. Am. J. Obstet. Gynecol., 1990, 162, 549-554.
Mediat., 2007, 84, 1-13. [32] Vermillion, S.T., Newman, R.B. Recent indomethacin tocolysis is
[10] Chandrasekharan, N.V.; Dai, H.; Roos, K.L.; Evanson, N.K.; Tom- not associated with neonatal complications in preterm infants. Am.
sik, J.; Elton, T.S.; Simmons, D.L. COX-3, a cyclooxygenase-1 J. Obstet. Gynecol., 1999 Nov, 181(5 Pt 1), 1083-1086.
variant inhibited by acetaminophen and other analgesic/antipyretic [33] Panter, K.R.; Hannah, M.E.; Amankwah, K.S.; Ohlsson, A.; Jeffer-
drugs: cloning, structure and expression. Proc. Natl. Acad. Sci., ies, A.L.; Farine, D. The effect of indomethacin tocolysis in pre-
2002, 99, 13926-13931. term labour on perinatal outcome: a randomised placebo-controlled
[11] Picot, D., Loll, P.J.; Garavito, R.M. The X-ray crystal structure of trial. BJOG: An International Journal of Obstetrics & Gynaecol-
the membrane protein prostaglandin H2 synthase-1. Nature, 1994, ogy, 1999, 106, 467–473.
367(6460), 243-249. [34] Stika, C.S.; Gross, G.A.; Leguizamon, G.; Gerber, S.; Levy, R.;
[12] Vane, J.R. Introduction: mechanism of action of NSAIDS. Br. J. Mathur, A.; Bernhard, L.M.; Nelson, D.M.; Sadovsky, Y. A pro-
Rheumatol., 1996, 35(Suppl l), l-3. spective randomized safety trial of celecoxib for treatment of pre-
[13] Luong, C.; Miller, A.; Barnett, J.; Chow, J.; Ramesha, C.; Browner, term labor. Am. J. Obstet. Gynecol., 2002, 187, 653-660.
M.F. Flexibility of the NSAID binding site in the structure of hu- [35] Kramer, W.B.; Saade, G.R.; Belfort, M.; Dorman, K.; Mayes, M.;
man cyclooxygenase-2. Nat. Struct. Biol., 1996, 3, 927-933. Moise, K.J. A randomized double-blind study comparing the fetal
[14] Vane, J.R. Inhibition of prostaglandin synthesis as a mechanisms of effects of sulindac to terbutaline during the management of preterm
action for the aspirin-like drugs. Nature, 1971, 231, 232-235. labor. Am. J. Obstet. Gynecol., 1999, 180, 396-401.
[15] Roth, G.J.; Stanford, N.; Majerus, P.W. Acetylation of prostaglan- [36] Poore, K.R.; Young, I.R.; Hirst, J.J. Efficacy of the selective pros-
din synthetase by aspirin. Proc. Natl. Acad. Sci. U S A, 1975, 72, taglandin synthetase type 2 inhibitor nimesulide in blocking basal
3073-3076. prostaglandin production and delaying glucocorticoidinduced pre-
[16] Pillinger, M.H.; Capodici, C.; Rosenthal, P.; Kheterpal, N.; Hanft, mature labor in sheep. Am. J. Obstet. Gynecol., 1999, 180, 1243-
S.; Philips, M.R.; Weissman, G. Modes of action of aspirin-like 1253.
drugs: Salicylates inhibit Erk activation and integrin-dependent
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 13

[37] Loudon, J.A.; Groom, K.M.; Bennett, P.R. Prostaglandin inhibitors [58] Ojala, R.; Ikonen, S.; Tammela, O. Perinatal indomethacin treat-
in preterm labour. Best Pract. Res. Clin. Obstet. Gynaecol., 2003, ment and neonatal complications in preterm infants. Eur. J. Pedi-
17, 731-744. atr., 2000, 159(3), 153-155.
[38] Abou-Ghannam, G; Usta IM; Nassar AH. Indomethacin in preg- [59] Amin, S.B.; Sinkin, R.A.; Glantz, J.C. Metaanalysis of the effect of
nancy: applications and safety. Am. J. Perinatol., 2011 Jul 22. antenatal indomethacin on neonatal outcomes. Am. J. Obstet. Gy-
[Epub ahead of print]. necol., 2007, 197, 486.e, 1-10.
[39] Stika, C.S.; Frederiksen, M.C. Drug Therapy in Pregnant and Nurs- [60] Montenegro, M.A.; Palomino, H. Induction of cleft palate in mice
ing Women. In: Atkinson A.J., Daniels C.E., Dedrick R.L., by inhibitors of prostaglandin synthesis. J. Craniofac. Genet. Dev.
Grudzinskas C.V., Markey S.P. (eds) Principles of Clinical Phar- Biol., 1990, 10(1), 83-94.
macology. Academic Press: London, 2001, 278-292. [61] Burdan, F.; Bezek, A. [Current opinions on embryotoxic and tera-
[40] Tsatsaris, V., Cabrol, D., Carbonne, B. Pharmacokinetics of toco- togenic effects of ibuprofen] [Article in Polish]. Pol. Merkur. Le-
lytic agents. Clin. Pharmacokinet., 2004, 43(13), 833-844. karski, 2001, 11(63), 266-270.
[41] Syme, M.R.; Paxton, J.W.; Keelan, J.A. Drug transfer and metabo- [62] Nielsen, G.L.; Sørensen, H.T.; Larsen, H.; Pedersen, L. Risk of
lism by the human placenta. Clin. Pharmacokinet,. 2004, 43, 487- adverse birth outcome and miscarriage in pregnant users of non-
514. steroidal anti-inflammatory drugs: population based observational
[42] Hill, M.D.; Abramson, F.P. The significance of plasma protein study and case-control study. BMJ, 2001, 322, 266-270.
binding on the foetal/maternal distribution of drugs at steady-state. [63] Li, D.K.; Liu, L.; Odouli, R. Exposure to non-steroidal anti-
Clin. Pharmacokinet., 1988, 14, 156-170. inflammatory drugs during pregnancy and risk of miscarriage:
[43] Hakkola, J.; Raunio, H.; Purkunen, R.; Pelkonen O.; Saarikoski, S.; population based cohort study. BMJ, 2003, 327(7411), 368.
Cresteil, T.; Pasanen, M. Detection of cytochrome P450 gene ex- [64] Dawood, M.Y. Nonsteroidal antiinflammatory drugs and reproduc-
pression in human placenta in first trimester of pregnancy. Bio- tion. Am. J. Obstet. Gynecol., 1993, 169, 1255-1265.
chem. Pharmacol., 1996, 52, 379-383. [65] Van der Weiden, R.M.; Helmerhorst, F.M., Keirse, M.J. Influence
[44] Hakkola, J.; Pasanen, M.; Hukkanen, J.; Pelkonen, O.; Mäenpää, J.; of prostaglandins and platelet activating factor on implantation.
Edwards, R.J.; Boobis, A.R.; Raunio, H. Expression of xenobiotic- Hum. Reprod., 1991, 6, 436-442.
metabolising cytochrome P450 forms in human full-term placenta. [66] Van der Weiden, R.M., Helmerhorst, F.M., Keirse, M.J. Prostanoid
Biochem. Pharmacol., 1996, 51, 403-411. excretion in incipient singleton and twin pregnancies. Am. J. Ob-
[45] Collier, A.C.; Ganley, N.A.; Tingle, M.D.; Blumenstein, M.; Mar- stet. Gynecol., 1996, 174, 1614-1617.
vin, K.W.; Paxton, J.W.; Mitchell, M.D; Keelan, J.A. UDP- [67] Van der Weiden, R.M., Wouters, J.M. Infertility may sometimes be
glucurosyltransferase activity, expression and cellular localization associated with non-steroidal anti-inflammatory drug consumption.
in human palcenta at term. Biochem. Pharmacol., 2002, 63, 409- Br. J. Rheumatol., 1997, 36, 605.
419. [68] Caruso, A.; De Carolis, S.; Di Simone, N. Antiphospholipid
[46] Jacobson, R.L.; Brewer, A.; Eis, A.; Siddiqi, TA. ; Myatt, L. Trans- antibodies in obstetrics: new complexities and sites of action. Hum.
fer of aspirin across the perfused human placental cotyledon. Am. J. Reprod., 1999, 5, 267-276.
Obstet. Gynecol., 1991, 165, 939-944. [69] Van Dorp, D.R.; Malleis, J.M.; Sullivan, B.P., Klein, M.D. Terato-
[47] Siu, S.S.; Yeung, J.H.; Lau, T.K. An in-vivo study on placental gens inducing congenital abdominal wall defects in animal models.
transfer of naproxen in early human pregnancy. Hum. Reprod., Pediatr. Surg. Int., 2010, 26, 127-139.
2002, 17, 1056-1059. [70] Hoyme, H.E.; Higginbottom, M.C.; Jones, K.L. The vascular
[48] Siu, S.S.; Yeung, J.H.; Lau, T.K. A study on placental transfer of pathogenesis of gastroschisis: intrauterine interruption of the om-
diclofenac in first trimester of human pregnancy. Hum. Reprod., phalomesenteric artery. J. Pediatr., 1981, 98, 228-331.
2000, 15, 2423-2425. [71] Burdan, F.; Szumilo, J,; Dudka, J.; Korobowicz, A. and Klepacz, R.
[49] Lampela, E.S.; Nuutinen, L.H.; Ala-Kokko, T.I.; Parikka, R.M.; Celosomy is associated with prenatal exposure to cyclooxygenase
Laitinen, R.S.; Jouppila, P.I.; Vähäkangas, K.H. Placental transfer inhibitors. Pharmacol. Res., 2006, 53, 287-292.
of sulindac, sulindac sulfide, and indomethacin in a human placen- [72] Cappon, G.D.; Cook, J.C.; Hurtt, M.E. Relationship between cy-
tal perfusion model. Am. J. Obstet. Gynecol., 1999, 180(1 Pt 1), clooxygenase 1 and 2 selective inhibitors and fetal development
174-180. when administered to rats and rabbits during the sensitive periods
[50] Leonhardt, A.; Bernert, S.; Watzer, B.; Schmitz-Ziegler, G.; Sey- for heart development and midline closure. Birth Defects Res. B.
berth, H.W. Low-dose aspirin in pregnancy: maternal and neonatal Dev. Reprod. Toxicol., 2003, 68(1), 47-56.
aspirin concentrations and neonatal prostanoid formation. Pediat- [73] Drongowski, R.A.; Smith, R.K. Jr.; Coran, A.G.; Klein, M.D. Con-
rics, 2003, 111, e77-81. tribution of demographic and environmental factors to the etiology
[51] Macones, G.A.; Marder, S.J.; Clothier, B.; Stamilio, D.M. The of gastroschisis: a hypothesis. Fetal Diagn. Ther., 1991, 6, 14-27.
controversy surrounding indomethacin for tocolysis. Am. J. Ob- [74] Werler, M.M.; Mitchell, A.A.; Shapiro, S. First trimester maternal
stet.Gynecol., 2001, 184, 264-272. medication use in relation to gastroschisis. Teratology, 1992, 45,
[52] Kramer, W.B.; Saade, G.; Ou, C.N.; Rognerud, C.; Dorman, K.; 361-367.
Mayes, M.; Moise, K.J. Placental transfer of sulindac and its active [75] Torfs, C.P.; Katz, E.A.; Bateson, T.F.; Lam, P.K.; Curry C.J. Ma-
sulfide metabolite in humans. Am. J. Obstet. Gynecol., 1995, 172, ternal medications and environmental exposures as risk factors for
886-890. gastroschisis. Teratology, 1996, 54, 84-92.
[53] Briggs, G.G.; Freeman, R.K.; Yaffe, S.J. Drugs in Pregnancy and [76] Martínez-Frías, M.L.; Rodríguez-Pinilla, E.; Prieto, L. Prenatal
Lactation. 6th edition. Philadelphia: Lippincott Williams & Wil- exposure to salicylates and gastroschisis: a case-control study.
kins; 2002. (eds). Teratology, 1997, 56(4), 241-243.
[54] Damase-Michel, C.; Christaud, J., Berrebi, A.; Lacroix, I.; Mon- [77] Werler, M.M.; Sheehan, J.E.; Mitchell, A.A. Maternal medication
tastruc, J.L. What do pregnant women know about non-steroidal use and risks of gastroschisis and small intestinal atresia. Am. J. E-
anti-inflammatory drugs? Pharmacoepidemiol. Drug Saf., 2009, pidemiol., 2002, 155, 26-31.
18(11), 1034-1038. [78] Werler, M.M.; Mitchell, A.A.; Moore, C.A., Honein, M.A. Is there
[55] Østensen, M. Antirheumatic therapy and reproduction. The influ- epidemiologic evidence to support vascular disruption as a patho-
ence on fertility, pregnancy and breast feeding. Z. Rheumatol., genesis of gastroschisis? National Birth Defects Prevention Study.
2006, 65, (3), 217-20, 222-224. Am. J. Med. Genet. A., 2009, 149A, 1399-1406.
[56] Auer, M.; Brezinka, C.; Eller, P.; Luze, K.; Schweigmann, U.; [79] Cleves, M.A.; Savell, V.H. Jr; Raj, S.; Zhao, W.; Correa, A.; Wer-
Schwärzler, P. Prenatal diagnosis of intrauterine premature closure ler, M.M.; Hobbs, C.A. Maternal use of acetaminophen and non-
of the ductus arteriosus following maternal diclofenac application. steroidal anti-inflammatory drugs (NSAIDs), and muscular
Ultrasound Obstet. Gynecol., 2004, 23(5), 513-6. ventricular septal defects. Birth Defects Res. A. Clin. Mol. Teratol.,
[57] Major, C.A.; Lewis, D.F.; Harding, J.A.; Porto, M.A.; Garite, T.J. 2004, 70, 107-113.
Tocolysis with indomethacin increases the incidence of necrotizing [80] Kallen, B.A.; Otterblad O.P. Maternal drug use in early pregnancy
enterocolitis in the low-birth-weight neonate. Am. J. Obstet. Gyne- and infant cardiovascular defect. Reprod. Toxicol., 2003, 17, 255-
col., 1994, 170, (1 Pt 1), 102-106. 261.
[81] Burdan, F. Comparison of developmental toxicity of selective and
non-selective cyclooxygenase-2 inhibitors in CRL:(WI)WUBR
14 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

Wistar rats--DFU and piroxicam study. Toxicology, 2005, 211(1-2), PGE2 after Maternal Treatment with Aspirin and Ibuprofen. J. Vet.
12-25. Med. Sci., 1998, 60(3), 377-379.
[82] Cook, J.C.; Jacobson, C.F.; Gao, F.; Tassinari, M.S.; Hurtt, M.E.; [105] James, A.H.; Brancazio, L.R. Aspirin and reproductive outcomes.
DeSesso, J.M. Analysis of the nonsteroidal anti-inflammatory drug Obstet. Gynecol. Surv., 2008, 63(1), 49-57.
literature for potential developmental toxicity in rats and rabbits. [106] Huhta, J.C.; Moise, K.J.; Fisher, D.J.; Sharif, D.S.; Wasserstrum,
Birth Defects Res. B. Dev. Reprod. Toxicol., 2003, 68, 5-26. N.; Martin, C. Detection and quantitation of constriction of the fetal
[83] Burdan, F.; Szumilo, J.; Dudka, J.; Korobowicz, A.; Klepacz, R. ductus arteriosus by Doppler echocardiography. Circulation, 1987,
Congenital ventricular septal defects and prenatal exposure to cyc- 75, 406–412.
looxygenase inhibitors. Braz. J. Med. Biol. Res., 2006, 39(7), 925- [107] Gewillig, M.; Brown, S.C.; De Catte, L.; Debeer, A.; Eyskens, B.;
934. Cossey, V.; Van Schoubroeck, D.; Van Hole, C.; Devlieger, R.
[84] DeSesso, J.M. Comparative embryology. In: Hood RD (Editor), Premature foetal closure of the arterial duct: clinical presentetions
Handbook of developmental toxicology. New York: CRS Press, and outcome. Eur. Heart. J., 2009, 30, 1530-1536.
1997. p 117-174. [108] Vermillion, S.T.; Scardo, J.A.; Lashus, A.G., Wiles, H.B. The
[85] Engle WA; American Academy of Pediatrics Committee on Fetus effect of indomethacin tocolysis on fetal ductus arteriosus constric-
and Newborn. Age terminology during the perinatal period. Pediat- tion with advancing gestational age. Am. J. Obstet. Gynecol., 1997,
rics, 2004, 114(5), 1362-1364. 177, 256-261.
[86] Stanfield, K.M.; Bell, R.R.; Lisowski, A.R.; English, M.L.; [109] Van Den Veyver, I.B., Moise, K.J. Jr.; Ou, C.N., Carpenter, R.J. Jr.
Saldeen, S.S.; Khan, K.N. Expression of cyclooxygenase-2 in em- The effect of gestational age and fetal indomethacin levels on the
bryonic and fetal tissues during organogenesis and late pregnancy. incidence of constriction of the fetal ductus arteriosus. Obstet. Gy-
Birth Defects Res. A. Clin. Mol. Teratol., 2003, 67, 54-58. necol., 1993, 82, 500-503.
[87] Ferencz, C.; Loffredo, C.A.; Correa-Villaseñor, A.; Wilson, P.D. [110] Tulzer, G., Gudmundsson, S., Sharkey, A.M., Wood, D.C., Cohen,
Ventricular septal defects. In: Ferencz C, Loffredo CA, Correa- A.W.; Huhta, J.C. Doppler echocardiography of fetal ductus arte-
Villaseñor A, Wilson PD (Editors), Genetic and enviromental risk riosus constriction versus increased right ventricular output. J. Am.
factors of major cardiovascular malformations: The Balimore- Coll. Cardiol., 1991, 18, 532-536.
Washington Infant Study 1981-1989. Armonk: Futura Publishing [111] Tarcan, A.; Gürakan, B.; Yildirim, S.; Ozkiraz, S., Bilezikçi, B.
Company, Inc.; 1997. p 123-163. Persistent pulmonary hypertension in a premature newborn after 16
[88] Briggs, G.G.; Freeman, R.K.; Yaffe, S.J. Drugs in pregnancy and hours of antenatal indomethacin exposure. J. Perinat. Med., 2004,
lactation: A reference guide to fetal and neonatal risk. Baltimore: 32(1), 98-99.
Williams & Wilkins; 1998. [112] Baerts, W.; Fetter, W.P.; Hop, W.C; Wallenburg, H.C.; Spritzer,
[89] Burdan, F.; Sykut, J.; Przybylski, P. [Developmental toxicity of R.; Sauer, P.J. Cerebral lesions in preterm infants after tocolytic in-
naproxen]. Pol. Merkur. Lekarski, 2007, 23(134), 155-158. Polish. domethacin. Dev. Med. Child. Neurol., 1990, 32, 910-918.
[90] Tikkanen, J.; Heinonen, O.P. Risk factors for ventricular septal [113] Loe, S.M.; Sanchez-Ramos, L.; Kaunitz, A.M. Assessing the neo-
defect in Finland. Public Health, 1991, 105(2), 99-112. natal safety of indomethacin tocolysis: a systematic review with
[91] Werler, M.M., Mitchell, A.A.; Shapiro, S. The relation of aspirin meta-analysis. Obstet. Gynecol., 2005, 106(1), 173-179.
use during the first trimester of pregnancy to congenital cardiac de- [114] Abbasi, S.; Gerdes, J.S.; Sehdev, H.M.; Samimi, S.S.; Ludmir, J.
fects. N. Engl. J. Med., 1989, 321, 1639-1642. Neonatal outcome after exposure to indomethacin in utero: a
[92] Botto, L.D.; Lynberg, M.C.; Erickson, J.D. Congenital heart de- retrospective case cohort study. Am. J. Obstet. Gynecol., 2003, 189,
fects, maternal febrile illness, and multivitamin use: a population- 782-785.
based study. Epidemiology, 2001, 12, 485-490. [115] Doyle, N.M.; Gardner, M.O.; Wells, L.; Qualls, C.; Papile, L.A.
[93] Mirkes, P.E.; Cornel, L.M.; Park, H.W.; Cunningham, M.L. Induc- Outcome of very low birth weight infants exposed to antenatal in-
tion of thermotolerance in early postimplantation rat embryos is as- domethacin for tocolysis. J. Perinatol., 2005, 25, 336-340.
sociated with increased resistance to hyperthermia-induced apopto- [116] Norton, M.E.; Merrill, J.; Cooper, B.A.; Kuller, J.A.; Clyman, R.I.
sis. Teratology, 1997, 56, 210-219. Neonatal complications after the administration of indomethacin
[94] Watanabe, M., Jafri, A., Fisher, S.A. Apoptosis is required for the for preterm labor. N. Engl. J. Med., 1993, 329(22), 1602-1607.
proper formation of the ventriculo-arterial connections. Dev. Biol., [117] Hammerman, C.; Glaser, J.; Kaplan, M.; Schimmel, M.S.; Ferber,
2001, 240, 274-288. B.; Eidelman, AI. Indomethacin tocolysis increases postnatal patent
[95] Van Overmeire, B.; Chemtob, S. The pharmacologic closure of the ductus arteriosus severity. Pediatrics, 1998, 102, E56.
patent ductus arteriosus. Semin. Fetal Neonatal med,. 2005, 10(2), [118] Clyman, R.I.; Chen, Y.Q.; Chemtob, S.; Mauray, F.; Kohl, T.;
177-184. Varma, D.R.; Roman, C. In utero remodeling of the fetal lamb duc-
[96] Coceani, F. Control of the ductus arteriosus: a new function for tus arteriosus: the role of antenatal indomethacin and avascular zo-
cytochrome P450, endothelin and nitric oxide. Biochem. Pharma- ne thickness on vasa vasorum proliferation, neointima formation,
col., 1994, 48, 1315-1318. and cell death. Circulation, 2001, 103(13), 1806-1812.
[97] Momma, K.; Takeuchi, H. Constriction of the ductus arteriosus by [119] Reese, J.; Anderson, J.D.; Brown, N.; Roman, C.; Clyman, R.I.
non-steroidal anti-inflammatory drugs. Prostaglandins, 1983, 26, Inhibition of cyclooxygenase isoforms in late- but not midgestation
631-643. decreases contractility of the ductus arteriosus and prevents postna-
[98] Locatelli, A.; Vergani, P., Bellini, P.; Strobelt, N.; Ghidini, A. Can tal closure in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol.,
a cyclooxygenase type-2 selective tocolytic agent avoid the fetal 2006, 291, R1717–R1723.
side effects of indomethacin? BJOG, 2001, 108, 325-326. [120] Loftin, C.D.; Trivedi, D.B.; Tiano, H.F., Clark, J.A.; Lee, C.A.,
[99] Momma, K.; Hagiwara, H.; Konishi, T. Constriction of fetal ductus Epstein, J.A.; Morham, S.G.; Breyer, M.D.; Nguyen, M.; Hawkins,
arteriosus by non-steroidal anti-inflammatory drugs:study of addi- B.M.; Goulet, J.L.; Smithies, O.; Koller, B.H.; Langenbach, R.
tional 34 drugs. Prostaglandins, 1984, 28(4), 527-536. Failure of ductus arteriosus closure and remodeling in neonatal
[100] Rein, A.J.; Nadjari, M.; Elchalal, U.; Nir, A. Contraction of the mice deficient in cyclooxygenase-1 and cyclooxygenase-2. Proc.
fetal ductus arteriosus induced by diclofenac. Case report. Fetal Natl. Acad. Sci. U S A, 2001, 98, 1059–1064.
Diagn. Ther,. 1999, 14(1), 24-25. [121] Momma, K.; Toyoshima, K.; Ito, K.; Sugiyama, K.; Imamura, S.;
[101] Paladini, D., Marasini, M.; Volpe, P. Severe ductal constriction in Sun, F.; Nakanishi, T. Delayed neonatal closure of the ductus arte-
the third-trimester fetus following maternal self-medication with riosus following early in utero exposure to indomethacin in the rat.
nimesulide. Ultrasound Obstet. Gynecol., 2005, 25(4), 357-356. Neonatology, 2009, 96, 69-79.
[102] Moise, K.J. Jr. Effect of advancing gestational age on the frequency [122] Reese, J.; Paria, B.C.; Brown, N.; Zhao, X.; Morrow, J.D.; Dey,
of fetal ductal constriction in association with maternal indometha- S.K. Coordinated regulation of fetal and maternal prostaglandins
cin use. Am. J. Obstet. Gynecol., 1993, 168(5), 1350-1353. directs successful birth and postnatal adaptation in the mouse.
[103] Coceani, F.; Huhtanen, D.; Hamilton, N.C.; Bishai, I.; Olley, P.M. Proc. Natl. Acad. Sci. U S A, 2000, 97, 9759–9764.
Involvement of intramural prostaglandin E2 in prenatal patency of [123] Baragatti, B.; Brizzi, F.; Ackerley, C.; Barogi, S.; Ballou, L.R.;
the lamb ductus arteriosus. Can. J. Physiol. Pharmacol., 1986, Coceani, F. Cyclooxygenase- 1 and cyclooxygenase-2 in the mouse
64(6), 737-744. ductus arteriosus: individual activity and functional coupling with
[104] Takizawa, T.; Ikeda, Y.; Kawahata, M., Togashi, H.; Yamamoto, nitric oxide synthase. Br. J. Pharmacol., 2003, 139, 1505–1515.
M.; Arishima, K.; Masaoka, T. Increased Ductal Responsiveness to
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 15

[124] Yokoyama, U.; Minamisawa, S., Quan, H.; Ghatak, S., Akaike, T., hypertension of the newborn. Am. J. Obstet. Gynecol., 1976, 126,
Segi-Nishida, E., Iwasaki, S.; Iwamoto, M., Misra, S., Tamura, K.; 467–469.
Hori, H., Yokota, S., Toole, B.P., Sugimoto, Y., Ishikawa, Y. [141] Levin, D.L.; Mills, L.J.; Weinberg A.G. Hemodynamic, pulmonary
Chronic activation of the prostaglandin receptor EP4 promotes hya- vascular, and myocardial abnormalities secondary to pharma-
luronan-mediated neointimal formation in the ductus arteriosus. J. cologic constriction of the fetal ductus arteriosus. A possible
Clin. Invest., 2006, 116, 3026–3034. mechanism for persistent pulmonary hypertension and transient tri-
[125] Clyman, R.I.; Waleh, N.S.; Kajino, H.; Roman, C.; Mauray, F. cuspid insufficiency in the newborn infant. Circulation, 1979,
Calcium-dependent and calcium-sensitizing pathways in the mature 60(2), 360-364.
and immature ductus arteriosus. Am. J. Physiol. Regul. Integr. [142] Wild, L.M., Nickerson, P.A., Morin, F.C. Ligating the ductus arte-
Comp. Physiol., 2007, 293, R1650–R1656. riosus before birth remodels the pulmonary vasculature of the lamb.
[126] Nakanishi, T.; Gu, H.; Hagiwara, N.; Momma, K. Mechanisms of Pediatr. Res., 1989, 25, 251–257.
oxygen-induced contraction of ductus arteriosus isolated from the [143] Momma, K.; Takao, A. In vivo constriction of the ductus arteriosus
fetal rabbit. Circ. Res., 1993, 72, 1218– 1228. by nonsteroidal antiinflammatory drugs in near term and preterm
[127] Tristani-Firouzi, M.; Reeve, H.L.; Tolarova, S.; Weir, E.K.; fetal rats. Pediatr. Res., 1987, 22, 567–572.
Archer, S.L. Oxygeninduced constriction of rabbit ductus arteriosus [144] Heymann, M.A.; Rudolph, A.M. Effects of acetylsalicylic acid on
occurs via inhibition of a 4-aminopyridine-, voltage-sensitive po- the ductus arteriosus and circulation in fetal lambs in utero. Circ.
tassium channel. J. Clin. Invest., 1996, 98, 1959–1965. Res., 1976, 38, 418-422.
[128] Thebaud, B.; Michelakis, E.D.; Wu, X.C.; Moudgil, R.; Kuzyk, M.; [145] Sharpe, G.L.; Larsson, K.S.; Thalme, B. Studies on closure of the
Dyck, J.R.; Harry, G.; Hashimoto, K.; Haromy, A.; Rebeyka, I.; ductus arteriosus. XII. In utero effect of indomethacin and sodium
Archer, S.L. Oxygen-sensitive Kv channel gene transfer confers salicylate in rats and rabbits. Prostaglandins, 1975, 9, 585–596.
oxygen responsiveness to preterm rabbit and remodeled human [146] Harker, L.C.; Kirkpatrick, S.E.; Friedman, W.F.; Bloor, C.M. Ef-
ductus arteriosus: implications for infants with patent ductus arte- fects of indomethacin on fetal rat lungs: a possible cause of persis-
riosus. Circulation, 2004, 110, 1372–1379. tent fetal circulation (PFC). Pediatr. Res., 1981, 15, 147–151.
[129] Thebaud, B.; Wu, X.C.; Kajimoto, H.; Bonnet, S.; Hashimoto, K.; [147] Lock, J.E.; Olley, P.M.; Soldin, S.; Coceani, F. Indomethacin-
Michelakis, E.D.; Archer, S.L. Developmental absence of the O2 induced pulmonary vasoconstriction in the conscious newborn
sensitivity of L-type calcium channels in preterm ductus arteriosus lamb. Pediatr. Res., 1980, 238, 639–644.
smooth muscle cells impairs O2 constriction contributing to patent [148] Ohara, T.; Ogata, H. (1988) Animal model of persistent pulmonary
ductus arteriosus. Pediatr. Res., 2008, 63, 176–181. hypertension of the newborn with fetal lambs. The XXIVth Con-
[130] Kajimoto, H.; Hashimoto, K., Bonnet, S.N.; Haromy, A.; Harry, gress of the Japan Society of Neonatokgy. Tokyo, Japan.
G.; Moudgil, R.; Nakanishi, T.; Rebeyka, I.; Thebaud, B.; [149] Demello, D.E.; Murphy, J.D.; Aronovitz, M.J.; Davies, P.; Reid,
Michelakis, E.D.; Archer, S.L. Oxygen activates the Rho/Rho- L.M. Effects of indomethacin in utero on the pulmonary vascula-
kinase pathway and induces RhoB and ROCK-1 expression in hu- ture of the newborn Guinea pig. Pediatr. Res., 1987, 22, 693-697.
man and rabbit ductus arteriosus by increasing mitochondria- [150] Ohara, T., Ogata, H.; Tezuka, F. Effects of indomethacin in utero
derived reactive oxygen species. A newly recognized mechanism on the pulmonary vasculature of the newborn lambs. Tohoku J.
for sustaining ductal constriction. Circulation, 2007, 115, 1777– Exp. Med., 1991, 164(1), 67-79.
1788. [151] Van Marter, L.J.; Leviton, A.; Allred, E.N.; Pagano, M.; Sullivan,
[131] Hong, Z., Hong; F., Olschewski, A., Cabrera, J.A., Varghese, A.; K.F.; Cohen, A.; Epstein, M.F. Persistent pulmonary hypertension
Nelson, D.P.; Weir, E.K. Role of store-operated calcium channels of the newborn and smoking and aspirin and nonsteroidal an-
and calcium sensitization in normoxic contraction of the ductus ar- tiinflammatory drug consumption during pregnancy. Pediatrics,
teriosus. Circulation, 2006, 114,1372–1379. 1996, 97, 658-663.
[132] Costa, M.; Barogi, S.; Socci, N.D.; Angeloni, D.; Maffei, M.; Ba- [152] Zenker, M.; Klinge, J.; Krüger, C.; Singer, H.; Scharf, J. Severe
ragatti, B.; Chiellini, C.; Grasso, E.; Coceani, F. Gene expression in pulmonary hypertension in a neonate caused by premature closure
ductus arteriosus and aorta: comparison of birth and oxygen effects. of the ductus arteriosus following maternal treatment with di-
Physiol. Genomics, 2006, 25, 250–262. clofenac: a case report. J. Perinat. Med., 1998, 26(3), 231-234.
[133] Kim, H.S.; Aikawa, M.; Kimura, K.; Kuro-o, M.; Nakahara, K.; [153] Siu, K.L.; Lee, W.H. Maternal diclofenac sodium ingestion and
Suzuki, T.; Katoh, H.; Okamoto, E.; Yazaki, Y.; Nagai, R. Ductus severe neonatal pulmonary hypertension. J. Paediatr. Child.
arteriosus. Advanced differentiation of smooth muscle cells dem- Health., 2004, 40(3), 152-153.
onstrated by myosin heavy chain isoform expression in rabbits. [154] Talati, A.J.; Salim, M.A.; Korones, S.B. Persistent pulmonary
Circulation, 1993, 88, 1804–1810. hypertension after maternal naproxen ingestion in a term newborn:
[134] Slomp, J.; Gittenberger-de Groot, A.C.; Glukhova, M.A.; Conny a case report. Am. J. Perinatol., 2000, 17(2), 69-71.
van Munsteren, J.; Kockx, M.M.; Schwartz, S.M.; Koteliansky, [155] Bouissou, A.; Glorieux, I.; Dulac, Y.; Marcoux, M.O.; Casper, C.
V.E. Differentiation, dedifferentiation, and apoptosis of smooth [Antenatal closure of ductus arteriosus following maternal intoxica-
muscle cells during the development of the human ductus arterio- tion by niflumic acid]. Arch. Pediatr., 2006, 13(1), 48-50. French.
sus. Arterioscler. Thromb. Vasc. Biol., 1997, 17, 1003–1009. [156] Joschko, M.A.; Dreosti, I.E.; Tulsi, R.S. The teratogenic effects of
[135] Morano, I.; Chai, G.X.; Baltas, L.G.; Lamounier-Zepter, V.; salicylic acid on the developing nervous system in rats in vitro.
Lutsch, G.; Kott, M.; Haase, H.; Bader, M. Smooth-muscle con- Teratology, 1993, 48(2), 105-114.
traction without smooth-muscle myosin. Nat. Cell. Biol., 2000, 2, [157] Elkarmi, A.; Abu-Samak, M.; Al-Qaisi, K. Modeling the effects of
371–375. prenatal exposure to aspirin on the postnatal development of rat
[136] Huang, J.; Cheng, L.; Li, J.; Chen, M.; Zhou, D.; Lu, M.M.; Prow- brain. Growth Dev. Aging., 2007, 70(1), 13-24.
eller, A.; Epstein, J.A.; Parmacek, M.S. Myocardin regulates ex- [158] Gökçimen, A.; Rabetli, M.C.; Ba, O.; Tunc, A.T.; Aslan, H.;
pression of contractile genes in smooth muscle cells and is required Yazici, A.C.; Kaplan, S. Effect of prenatal exposure to an anti-
for closure of the ductus arteriosus in mice. J. Clin. Invest., 2008, inflammatory drug on neuron number in cornu ammonis and den-
118, 515–525. tate gyrus of the rat hippocampus: a stereological study. Brain Res.,
[137] Takizawa, T.; Oda, T.; Arishima, K.; Yamamoto, M.; Masaoka, T.; 2007, 1127(1), 185-192.
Somiya, H.; Akahori, F.; Shiota, K. A calcium channel blocker [159] Ragbetli, M.C.; Ozyurt, B., Aslan, H.; Odaci, E.; Gokcimen, A.,
verapamil inhibits the spontaneous closure of the ductus arteriosus Sahin, B.; Kaplan, S. Effect of prenatal exposure to diclofenac so-
in newborn rats. J. Toxicol. Sci., 1994, 19, 171–174. dium on Purkinje cell numbers in rat cerebellum: a stereological
[138] Reese, J.; Waleh, N.; Poole, S.D.; Brown, N.; Roman, C., Clyman, study. Brain Res., 2007, 1174, 130-135.
R.I. Chronic in utero cyclooxygenase inhibition alters PGE2- [160] Odaci, E.; Cihan, O.F.; Aslan, H., Ragbetli, M.C.; Kaplan, S. Pre-
regulated ductus arteriosus contractile pathways and prevents post- natal diclofenac sodium administration increases the number of
natal closure. Pediatr. Res, 2009, 66, 155-161. Purkinje cells in female rats: a stereological study. Int. J. Dev. Neu-
[139] Philips, J.B.; Lyrene, R.K. Prostaglandins, related compounds, and rosci., 2010, 28(2), 145-151.
the perinatal pulmonary circulation. Clin. Perinatol., 1984, 11, [161] Marret, S.; Marchand, L.; Kaminski, M.; Larroque, B.; Arnaud, C.;
565–579. Truffert, P.; Thirez, G.; Fresson, J.; Rozé, J.C.; Ancel, P.Y.; EPI-
[140] Manchester, D.; Margolis, H.S., Sheldon, R.E. Possible association PAGE Study Group. Prenatal low-dose aspirin and neurobehavioral
between maternal indomethacin therapy and primary pulmonary
16 Current Drug Metabolism, 2012, Vol. 13, No. ?? Antonucci et al.

outcomes of children born very preterm. Pediatrics, 2010, 125(1), [181] Salokorpi, T.; Eronen, M.; von Wendt, L. Growth and development
e29-34. until 18 months of children exposed to tocolytics indomethacin or
[162] Canan, S.; Akta, A.; Ulkay, M.B.; Colakoglu, S.; Ragbetli, M.C.; nylidrin. Neuropediatrics, 1996, 27(4), 174–177.
Ayyildiz, M.; Geuna, S.; Kaplan, S. Prenatal exposure to a non- [182] Al-Alaiyan, S.; Seshia, M.M.; Casiro, O.G. Neurodevelopmental
steroidal anti-inflammatory drug or saline solution impairs sciatic outcome of infants exposed to indomethacin antenatally. J. Perinat.
nerve morphology: a stereological and histological study. Int. J. Med., 1996, 24(4), 405-411.
Dev. Neurosc., 2008, 26(7), 733-738. [183] Amin, S.B.; Kamaluddeen, M.; Sangem, M. Neurodevelopmen-
[163] Wood, C.E., Powers Fraites, M.; Keller-Wood, M. Blockade of tal Outcome of Premature Infants after Exposure to Antenatal In-
PGHS-2 inhibits the hypothalamus-pituitary-adrenal axis response domethacin. Am. J. Obstet. Gynecol., 2008, 199(1), 41.e1–41.e8.
to cerebral hypoperfusion in the sheep fetus. Am. J. Physiol. Regul. [184] Macones, G.A., Robinson, C.A. Is there justification for using
Integr. Comp. Physiol., 2009, 296(6), R1813-1819. indomethacin in preterm labor? An analysis of neonatal risks and
[164] Knutson, N.; Wood, C.E. Interaction of PGHS-2 and glutamatergic benefits. Am. J. Obstet. Gynecol., 1997, 177(4), 819-824.
mechanisms controlling the ovine fetal hypothalamus-pituitary- [185] Mañalich, R.; Reyes, L.; Herrera, M.; Melendi, C.; Fundora, I.
adrenal axis. Am. J. Physiol. Regul. Integr. Comp. Physiol., 2010, Relationship between weight at birth and the number and size of
299(1), R365-370. renal glomeruli in humans: a histomorphometric study. Kidney Int.,
[165] Tong, H.; Lakhdir, F., Wood, C.E. Endogenous prostanoids modu- 2000, 58, 770-773.
late the ACTH and AVP responses to hypotension in late-gestation [186] Zaffanello, M.; Bassareo, P.P.; Cataldi, L.; Antonucci, R.; Biban,
fetal sheep. Am. J. Physiol. Regul. Integr. Comp. Physiol., 1998, P.; Fanos, V. Long-term effects of neonatal drugs on the kidney. J.
275, R735–R741. Matern. Fetal Neonatal Med., 2010, 23 Suppl 3, 87-89.
[166] Parfenova, H.; Levine, V.; Gunther, W.M.; Pourcyrous, M.; Lef- [187] Cataldi, L.; Leone, R.; Moretti, U.; De Mitri, B.; Fanos, V.; Rugge-
fler, C.W. COX-1 and COX-2 contributions to basal and IL-1 beta- ri, L.; Sabatino, G.; Torcasio, F.; Zanardo, V.; Attardo, G.; Ricco-
stimulated prostanoid synthesis in human neonatal cerebral mi- bene, F.; Martano, C.; Benini, D.; Cuzzolin, L. Potential risk fac-
crovascular endothelial cells. Pediatr. Res., 2002, 52, 342-348. tors for the development of acute renal failure in preterm newborn
[167] Chemtob, S.; Laudignon, N.; Beharry, K.; Rex, J.; Varma, D.; infants: a case-control study. Arch. Dis. Child. Fetal Neonatal, Ed.
Wolfe, L.; Aranda, J.V. Effects of prostaglandins and indomethacin 2005, 90(6), F514-519.
on cerebral blood flow and cerebral oxygen consumption of con- [188] Boubred, F.; Vendemmia, M.; Garcia-Meric, P.; Buffat, C.; Millet,
scious newborn piglets. Dev. Pharmacol. Ther., 1990, 14(1), 1-14. V.; Simeoni, U. Effects of maternally administered drugs on the fe-
[168] Pourcyrous, M.; Leffler, C.W.; Bada, H.S., Korones, S.B.; Busija, tal and neonatal kidney. Drug Saf., 2006, 29(5), 397-419.
D.W. Cerebral blood flow responses to indomethacin in awake [189] Bavoux, F. [Fetal toxicity of non-steroidal anti-inflammatory a-
newborn pigs. Pediatr. Res., 1994, 35(5), 565-570. gents]. Presse Med., 1992, 21(40), 1909-1912. French.
[169] Li, D.Y.; Hardy, P., Abran, D.; Martinez-Bermudez, A.K.; Guer- [190] Drukker, A.; Guignard, J.P. Renal aspects of the term and preterm
guerian, A.M.; Bhattacharya, M.; Almazan, G.; Menezes, R.; Peri, infant: a selective update. Curr. Opin. Pediatr., 2002, 14(2), 175-
K.G.; Varma, D.R.; Chemtob, S. Key role for cyclooxygenase-2 in 182.
PGE2 and PGF2alpha receptor regulation and cerebral blood flow [191] Cuzzolin, L.; Dal Cerè, M.; Fanos, V. NSAID-induced nephrotox-
of the newborn. Am. J. Physiol., 1997, 273(4 Pt 2), R1283-1290. icity from the fetus to the child. Drug Saf., 2001, 24(1), 9-18.
[170] Najarian, T.; Hardy, P.; Hou, X.; Lachapelle, J.;, Doke, A.; Gobeil, [192] Robin, Y.M.; Reynaud, P.; Orliaguet, T.; Lemery, D.; Vanlieferin-
F.jr.; Roy, M.S.; Lachapelle, P.; Varma, D.; Chemtob, S. Preserva- gen, P.; Dechelotte, P. Renal tubular dysgenesis-like lesions and
tion of neural function in the perinate by high PGE2 levels acting hypocalvaria. Report of two cases involving indomethacin. Pathol.
via EP2 receptors. J. Appl. Physiol., 2000, 89, 777–784. Res. Pract,. 2000, 196(11), 791-794.
[171] Mosca F; Bray M; Lattanzio M; Fumagalli M; Tosetto C. Com- [193] Butler-O’Hara, M.; D’Angio, C.T. Risk of persistent renal insuffi-
parative evaluation of the effcts of indomethacin and ibuprofen on ciency in premature infants following the prenatal use of indo-
cerebral perfusion and oxygenation in preterm infants with aptent methacin for suppression of preterm labor. J. Perinatol., 2002,
ductus arteriosus. J. Pediatr., 1997, 131, 549-554. 22(7), 541-546.
[172] Thomas RL; Parker GC; Van Overmeire B; Aranda JV. A meta- [194] Itabashi, K.; Ohno, T.; Nishida, H. Indomethacin responsiveness of
analysis of ibuprofen versus indomethacin for closure of patent patent ductus arteriosus and renal abnormalities in preterm infants
ductus arteriosus. Eur. J. Pediatr., 2005, 164, 135-140. treated with indomethacin. J. Pediatr., 2003, 143(2), 203-207.
[173] Murata, Y.; Itakura, A.; Matsuzawa, K.; Okumura, A.; Wakai, K.; [195] Jacqz-Aigrain, E.; Guillonneau, M.; Boissinot, C.; Bavoux, F.;
Mizutani, S. Possible antenatal and perinatal related factors in de- Hartmann, J.F.; Blot, P. [Maternal and neonatal effects of indo-
velopment of cystic periventricular leukomalacia. Brain Dev., methacin administrated during pregnancy. Apropos of 18 cases].
2005, 27, 17-21. Arch. Fr. Pediatr., 1993, 50(4), 307-312. French.
[174] Perlman, J.M. The relationship between systemic hemodynamic [196] van der Heijden, B.J., Carlus, C.; Narcy, F., Bavoux, F., Delezoide,
perturbations and periventricular-intraventricular hemorrhage--a A.L.; Gubler, M.C. Persistent anuria, neonatal death, and renal mi-
historical perspective. Semin. Pediatr. Neurol., 2009, 16(4), 191- crocystic lesions after prenatal exposure to indomethacin. Am. J.
199. Obstet. Gynecol., 1994, 171(3), 617-623.
[175] Suarez, R.D.; Grobman, W.A.; Parilla, B.V. Indomethacin tocolysis [197] van der Heijden, B.; Gubler, M.C. Renal failure in the neonate
and intraventricular hemorrhage. Obstet. Gynecol., 2001, 97, 921- associated with in utero exposure to non-steroidal anti-
925. inflammatory agents. Pediatr. Nephrol., 1995, 9(5), 675.
[176] Rumack, C.M.; Guggenheim, M.A.; Rumack, B.H.; Peterson, R.G.; [198] van der Heijden, A.J., Provoost, A.P., Nauta, J., Wolff, E.D., Sauer,
Johnson, M.L.; Braithwaite, W.R. Neonatal intracranial hemor- P.J. Indomethacin as an inhibitor of preterm labor. Effect on post-
rhage and maternal use of aspirin. Obstet. Gynecol., 1981, 58 (5 natal renal function. Contrib. Nephrol., 1988, 67, 152-154.
Suppl), 52S-56S. [199] Van Den Anker, J.N.; Hop, W.C.J.; De Groot, R.; Van Der Hei-
[177] Govaert, P.; Staelens, V.; Vanhaesebrouch, P. Perinatal intracranial jden, B.J.; Broerse, H.M.; Lindemans, J.; Sauer, P.J.J. Effects of
hemorrhage due to maternal salicylate ingestion. Clin. Pediatr. prenatal exposure to betamethasone and indomethacin on the
(Phila)., 1995, 34, 174-175. glomerular filtration rate in the preterm infant. Pediatr. Res., 1994,
[178] Shetty, S.; Ghosh, K. Anti-phospholipid antibodies and other 36, 578-581.
immunological causes of recurrent foetal loss – a review of litera- [200] Vieux, R.; Fresson, J.; Guillemin, F.; Hascoet, J.M. Perinatal drug
ture of various therapeutic protocols. Am. J. Reprod. Immunolog., exposure and renal function in very preterm infants. Arch. Dis.
2009, 62, 9-24. Child. Fetal Neonatal Ed., 2011, 96, 290-295.
[179] Skrzypczak, J. [The management of pregnant women with an- [201] George, I.; Mekahli, D.; Rayyan, M.; Levtchenko, E.; Allegaert, K.
tiphospholipid syndrome]. Arch. Med. Wewn., 2007, 117, 50-53. Postnatal trends in creatinemia and its covariates in extremely low
[Article in Polish] birth weight (ELBW) neonates. Pediatr. Nephrol., 2011, 26, 1843-
[180] Parilla, B.V.; Tamura, R.K.; Cohen, LS, Clark, E. Lack of effect of 1849.
antenatal indomethacin on fetal cerebral blood flow. Am. J. Obstet. [202] Allegaert, K.; Rayyan, M.; Anderson, B.J. Impact of ibuprofen
Gynecol., 1997, 176(6), 1166-1169, discussion 1169-1171. administration on renal drug clearance in the first weeks of life.
Methods Find. Exp. Clin. Pharmacol., 2006, 28(8), 519-522.
Impact of Prenatal NSAIDs on the Offspring Current Drug Metabolism, 2012, Vol. 13, No. ?? 17

[203] Iacobelli, S.; Bonsante, F.; Ferdinus, C.; Labenne, M.; Gouyon, [224] Burdan, F.; Rozylo-Kalinowska, I.; Szumilo, J.; Dudka, J.and Kle-
J.B. Factors affecting postnatal changes in serum creatinine in pre- pacz, R. Cyclooxygenase inhibitors affect bone mineralization in
term infants with gestational age<32 weeks. J. Perinatol., 2009, rat fetuses. Cells Tissues Organs, 2008, 187, 221-232.
29(3), 232-236. [225] Burdan, F.; Szumilo, J; Klepacz, R. Maternal toxicity of nonsteroi-
[204] Antonucci, R.; Cuzzolin, L.; Arceri, A.; Dessì, A.; Fanos, V. dal anti-inflammatory drugs as an important factor affecting prena-
Changes in urinary PGE2 after ibuprofen treatment in preterm in- tal development. Reprod. Toxicol., 2009, 28, 239-244.
fants with patent ductus arteriosus. Eur. J. Clin. Pharmacol., 2009, [226] Dimmen S; Nordsletten L; Madsen JE. Parecoxib and indometha-
65(3), 223-30. cin delay early fracture healing: a study in rats. Clin. Orthop. Relat.
[205] Benini, D.; Fanos, V.; Cuzzolin, L.; Tatò, L. In utero exposure to Res., 2009, 467(8), 1992-1999.
nonsteroidal anti-inflammatory drugs: neonatal renal failure. Pedi- [227] Dodo T.; Fukuta T.; Uchida K.; Mineshima H.; Okuda Y.; Okada
atr. Nephrol., 2004, 19(2), 232-234. F.; Katsutani N.; Hirano K.; Aoki T. A comparative investigation
[206] Magnani, C.; Moretti, S.; Ammenti, A. Neonatal chronic renal of fetal skeletal anomalies in rats induced by acetylsalicylic acid
failure associated with maternal ingestion of Nimesulide as analge- with single- and double-staining techniques. Regul. Toxicol. Phar-
sic. Eur. J. Obstet. Gynecol. Reprod. Biol., 2004, 116(2), 244-245. macol., 2009, 54(3), 308-313.
[207] Ali, U.S.; Khubchandani, S.; Andankar, P.; Parekhji, S.; Dubhalish, [228] Attridge, J.T.; Clark, R.; Walker, M.W.; Gordon, P.V. New insights
V.; Padhye, B. Renal tubular dysgenesis associated with in utero into spontaneous intestinal perforation using a national data set: (1)
exposure to Nimuselide. Pediatr. Nephrol., 2006, 21(2), 274-276. SIP is associated with early indomethacin exposure. J. Perinatol.,
[208] Alessandri, J.L.; Abossolo, T.; Reynaud, I.; Montbrun, A.; 2006, 26(2), 93-9.
Sommer, J.C.; Tilmont, P. [Non-steroidal anti-inflammatory agents [229] Gordon, P.V. Understanding intestinal vulnerability to perforation
and pregnancy. A study of renal and digestive toxicity of niflumic in the extremely low birth weight infant. Pediatr. Res., 2009, 65(2),
acid in the perinatal period]. J. Gynecol. Obstet. Biol. Reprod. 138-144.
(Paris), 1994, 23(7), 813-818. French. [230] Gordon, P.V.; Attridge, J.T. Understanding clinical literature rele-
[209] Voyer, L.E., Drut, R.; Méndez, J.H. Fetal renal maldevelopment vant to spontaneous intestinal perforations. Am. J. Perinatol., 2009,
with oligohydramnios following maternal use of piroxicam. Pedi- 26, 309-316.
atr. Nephrol., 1994, 8(5), 592-594. [231] Neu, J.; Wu-Wang, C.Y. Eicosanoids in the developing gastrointes-
[210] Gouyon, J.B.; Petion, A.M.; Sandre, D.; Sgro, C.; Bavoux, F. tinal tract. Semin. Perinatol., 1987, 11 (1), 22–30.
[Neonatal kidney insufficiency and intrauterine exposure to keto- [232] Wallace, J.L., Cohen, M.M. Gastric mucosal protection with
profen]. Arch. Fr. Pediatr., 1991, 48(5), 347-348. French. chronic mild restraint: role of endogenous prostaglandins. Am. J.
[211] Fieni, S.; Gramellini, D.; Vadora, E. Oligohydramnios and fetal Physiol., 1984, 247(2 Pt 1), G127–132.
renal sonographic appearances related to prostaglandin synthetase [233] Peskar, B.M. On the synthesis of prostaglandins by human gastric
inhibitors. A case report. Fetal Diagn. Ther., 2004, 19(3), 224-227. mucosa and its modification by drugs. Biochim. Biophys. Acta,
[212] Chen, N.; Aleksa, K.; Woodland, C.; Rieder, M.; Koren, G. On- 1977, 487(2), 307–314.
togeny of drug elimination by the human kidney. Pediatr. Nephrol., [234] Meyers, R.L.; Alpan, G.; Lin, E.; Clyman, R.I. Patent ductus arte-
2006, 21, 160–168. riosus, indomethacin, and intestinal distension: effects on intestinal
[213] Fanos, V.; Cataldi, L. Antibacterial-induced nephrotoxicity in the blood flow and oxygen consumption. Pediatr. Res., 1991, 29(6),
newborn. Drug. Saf., 1999, 20, 245-267. 569–574.
[214] Balasubramaniam, J. Nimesulide and neonatal renal failure. Lancet, [235] Watterberg, K.L.; Jerdes, J.S.; Cole, C.H.; Aucott, S.W.; Thilo,
2000, 355(9203), 575. E.H.; Mammel, M.C.; Couser, R.J.; Garland, J.S.; Rozycki, H.J.;
[215] Koklu, E.; Gurgoze, M., Akgun, H., Ozturk, M.A.; Poyrazoglu, Leach, C.L.; Backstrom, C.; Shaffer, M.L. Prophylaxis of early ad-
M.H. Renal tubular dysgenesis with atypical histology and in-utero renal insufficiency to prevent bronchopulmonary dysplasia: a mul-
exposure to naproxen sodium. Ann. Trop. Paediatr., 2006, 26(3), ticenter trial. Pediatrics, 2004, 114, 1649-1657.
241-245. [236] Vanhaesebrouck, P.; Thiery, M.; Leroy, J.G.; Govaert, P.; de
[216] Nguyen, M.T.; Devarajan, P. Biomarkers for the early detection of Praeter, C.; Coppens, M.; Cuvelier, C.; Dhont, M. Oligohydram-
acute kidney injury. Pediatr. Nephrol., 2008, 23, 2151-2157. nios, renal insufficiency, and ileal perforation in preterm infants af-
[217] Zaffanello, M., Antonucci, R.; Cuzzolin, L.; Cataldi, L.; Fanos, V. ter intrauterine exposure to indomethacin. J. Pediatr., 1988, 113,
Early diagnosis of acute kidney injury with urinary biomarkers in 738-743.
the newborn. J. Matern. Fetal Neonatal Med., 2009, 22(s3), 62-66. [237] Gordon, P.V.; Swanson, J.R.; Clark R. Antenatal indomethacin is
[218] Nagai, A.; Katayama, M.; Thurlbeck, W.M.; Matsui, R.; Yasui, S.; more likely associated with spontaneous intestinal perforation
Konno, K. Effect of indomethacin on lung development in postna- rather than NEC. Am. J. Obstet. Gynecol., 2008, 198, 725; author
tal rats: possible role of prostaglandin in alveolar formation. Am. J. reply 725-726.
Physiol., 1995, 268 (1 Pt 1), L56-62. [238] Sharma, R.; Hudak, M.L.; Tepas, J.J. 3rd; Wludyka, P.S.; Teng,
[219] Ambalavanan, N.; Walsh, M.; Bobashev, G.; Das, A.; Levine,B.; R.J.; Hastings, L.K.; Renfro, W.H.; Marvin, W.J. Jr. Prenatal or
Carlo, W.A.; Higgins, R.D.; NICHD Neonatal Research Network. postnatal indomethacin exposure and neonatal gut injury associated
Intercenter differences in bronchopulmonary dysplasia or death with isolated intestinal perforation and necrotizing enterocolitis. J.
among very low birth weight infants. Pediatrics, 2011, 127, 106- Perinatol., 2010, 30(12), 786-793.
116. [239] Guthrie, S.O.; Gordon, P.V.; Thomas, V.; Thorp, J. A.; Peabody,
[220] Eronen, M.; Peesonen, E.; Kurki, T.; Teramo, K.;Ylikorkala, O. J.and Clark, R. H. Necrotizing Enterocolitis Among Neonates in
and Hallman, M. Increased incidence of bronchopolmonary dyspla- the United States. J. Perinatol., 2003, 23, 278–285.
sia after antenatal administration of indomethacin to prevent pre- [240] Petrenko, V.M.; Petrenko, E.V. [Compensatory responses in lymph
term labour. J. Pediatr.,1994, 124, 782-788. nodes following by prenatal indomethacin exposure]. Morfologiia,
[221] Ho, M.L.; Chang, J.K.; Chuang, L.Y.; Hsu, H.K.; Wang, G.J. Ef- 2001, 119(1), 37-40. Russian.
fects of nonsteroidal anti-inflammatory drugs and prostaglandins [241] Morozova, E.V. [Structure of the mesenteric lymph nodes in fe-
on osteoblastic functions. Biochem. Pharmacol., 1999, 58, 983- tuses and offspring of white rats under the effect of indomethacin
990. on the mother-fetus system]. Arkh. Anat. Gistol. Embriol., 1989,
[222] Chang, J.K.; Wang, G.J.; Tsai, S.T. and Ho, M.L. Nonsteroidal 96(3), 48-55. Russian.
anti-inflammatory drug effects on osteoblastic cell cycle, cytotoxic- [242] Chakravarty, K.; Merry, P. Anticardiolipin syndrome, aspirin ther-
ity, and cell death. Connect Tissue Res., 2005, 46, 200-210. apy and fetal malformation. Lupus, 1993, 2(1), 61-62.
[223] Chang, J.K.; Wu, S.; Wang, G.J.; Cho, M.H. and Ho, M.L. Effects [243] Agapitos, M.; Georgiou-Theodoropoulou, M.; Koutselinis, A.;
of non-steroidal anti-inflammatory drugs on cell proliferation and Papacharalampus, N. Cyclopia and maternal ingestion of salicy-
death in cultured epiphyseal-articular chondrocytes of fetal rats. lates. Pediatr. Pathol., 1986, 6(2-3),309-310.
Toxicology, 2006, 228, 111-123. [244] Stuart, M.J.; Gross, S.J.; Elrad, H.; Graeber, J.E. Effects of acetyl-
salicylic-acid ingestion on maternal and neonatal hemostasis. N.
Engl. J. Med., 1982, 307(15), 909-912.

Received: July 26, 2011 Revised: December 15, 2011 Accepted: December 20, 2011

You might also like