Вы находитесь на странице: 1из 28

Clin Pharmacokinet 2004; 43 (8): 487-514

REVIEW ARTICLE 0312-5963/04/0008-0487/$31.00/0

© 2004 Adis Data Information BV. All rights reserved.

Drug Transfer and Metabolism by the


Human Placenta
Michael R. Syme, James W. Paxton and Jeffrey A. Keelan
Division of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences,
University of Auckland, Auckland, New Zealand

Contents

Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 488
1. Placental Anatomy and Physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 490
2. Drug Transfer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 490
2.1 Passive Transfer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 491
2.1.1 Drug Properties Affecting Transfer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 492
2.1.2 Maternal, Placental and Fetal Characteristics Affecting Drug Transfer . . . . . . . . . . . . . . . 494
2.2 Facilitated Diffusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
2.3 Active Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
2.3.1 P-Glycoprotein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 496
2.3.2 Multidrug Resistance Protein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 497
2.3.3 Breast Cancer Resistant Protein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 498
2.3.4 Monoamine Transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 498
2.3.5 Novel Organic Cation Transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 498
2.3.6 Monocarboxylate Transporters and the Dicarboxylate Transporter . . . . . . . . . . . . . . . . . . 498
2.3.7 Sodium/Multivitamin Transporter . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
3. Drug Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
3.1 Phase I Reactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
3.1.1 Cytochrome P450 (CYP) 1 Family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
3.1.2 CYP2 Family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 500
3.1.3 CYP3 Family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 500
3.2 Phase II Reactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 500
3.2.1 Uridine Diphosphate Glucuronosyltransferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 500
3.2.2 Glutathione S-Transferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501
3.2.3 Epoxide Hydrolase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501
3.2.4 Sulfotransferases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501
4. Models to Study Drug Transfer and Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501
4.1 In Vitro Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 501
4.1.1 Perfused Placental Cotyledon . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 502
4.1.2 Tissue Culture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 502
4.2 In Vivo Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 502
5. Clinical Significance of Placental Transfer and Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 503
6. Drugs of Abuse . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 506
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 507
488 Syme et al.

Abstract The major function of the placenta is to transfer nutrients and oxygen from the
mother to the foetus and to assist in the removal of waste products from the foetus
to the mother. In addition, it plays an important role in the synthesis of hormones,
peptides and steroids that are vital for a successful pregnancy. The placenta
provides a link between the circulations of two distinct individuals but also acts as
a barrier to protect the foetus from xenobiotics in the maternal blood.
However, the impression that the placenta forms an impenetrable obstacle
against most drugs is now widely regarded as false. It has been shown that that
nearly all drugs that are administered during pregnancy will enter, to some degree,
the circulation of the foetus via passive diffusion. In addition, some drugs are
pumped across the placenta by various active transporters located on both the fetal
and maternal side of the trophoblast layer. It is only in recent years that the impact
of active transporters such as P-glycoprotein on the disposition of drugs has been
demonstrated. Facilitated diffusion appears to be a minor transfer mechanism for
some drugs, and pinocytosis and phagocytosis are considered too slow to have any
significant effect on fetal drug concentrations.
The extent to which drugs cross the placenta is also modulated by the actions of
placental phase I and II drug-metabolising enzymes, which are present at levels
that fluctuate throughout gestation. Cytochrome P450 (CYP) enzymes in particu-
lar have been well characterised in the placenta at the level of mRNA, protein, and
enzyme activity. CYP1A1, 2E1, 3A4, 3A5, 3A7 and 4B1 have been detected in
the term placenta. While much less is known about phase II enzymes in the
placenta, some enzymes, in particular uridine diphosphate glucuronosyltransfer-
ases, have been detected and shown to have specific activity towards marker
substrates, suggesting a significant role of this enzyme in placental drug detoxifi-
cation.
The increasing experimental data on placental drug transfer has enabled
clinicians to make better informed decisions about which drugs significantly cross
the placenta and develop dosage regimens that minimise fetal exposure to poten-
tially toxic concentrations. Indeed, the foetus has now become the object of
intended drug treatment. Extensive research on the placental transfer of drugs
such as digoxin and zidovudine has assisted with the safe treatment of the foetus
with these drugs in utero. Improved knowledge regarding transplacental drug
transfer and metabolism will result in further expansion of pharmacological
treatment of fetal conditions.

The placenta separates the blood supply of two cific transport mechanisms present in the
human beings, mother and foetus, while being si- maternal-facing apical (brush border) membrane
multaneously perfused by both their circulations. It and fetal-facing basal membrane of the syncytio-
acts to supply the foetus with oxygen and nutrients trophoblast.[1-3] At the same time, metabolic waste
from the mother and also functions as an active products are cleared from fetal blood across the
endocrine organ vital to successful pregnancy. This placenta into the maternal circulation, again using
makes the placenta a unique organ system in the specific transport mechanisms in many cases.[4]
body. Nutrients such as glucose, amino acids and For some compounds, the placenta offers a pro-
vitamins are transferred across the placenta via spe- tective barrier for the developing foetus by reducing

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 489

Gestational
Maternal drug Mother membranes
administration Amniotic
fluid

Bound drug Bound


Fetus
drug

Placenta Free drug


Site of action Umbilical
Passive arteries
diffusion Liver
Site of action

Metabolites Free drug Active


Metabolites
transport
Gut

Metabolism Swallowing
Metabolism

Liver Umbilical
vein

Kidney

Kidney Fetal urine

Meconium

Excretion

Fig. 1. Drug disposition in mother and foetus after maternal drug administration. A variety of pharmacokinetic variables, including
transplacental transport and metabolism, determine the degree of maternal-to-fetal drug transfer and fetal drug exposure. Black arrows
represent parent drug and white arrows represent metabolites. The size of the arrows approximates relative importance, although this is
drug-dependent and will vary during pregnancy with fetal and placental maturation.

the entry of various xenobiotics from the mother to to plasma proteins may also affect the amount trans-
the foetus, while for others it facilitates their passage ferred.[5]
both to and from the fetal compartment (figure 1). The transfer of foreign chemicals across the pla-
Hence, the placenta can act much the same as the centa can also be modified by metabolism in the
kidney or liver in the elimination of xenobiotics. placenta itself. The human placenta contains multi-
Since placental transfer of drugs from the maternal ple enzyme systems, including those responsible for
drug oxidation, reduction, hydrolysis and conjuga-
to the fetal side occurs primarily via passive diffu-
tion.[6-9] However, the activities of these enzymes
sion, the physicochemical properties of drugs such
are usually relatively minor compared with those of
as lipid solubility, polarity and molecular weight the maternal or fetal liver and these systems are
primarily determine the rate of transfer across the thought to be primarily involved with endogenous
placenta. According to membrane permeability steroid metabolism.[10]
properties, low-molecular-weight, lipid-soluble and Drug administration to pregnant women must
un-ionised compounds can easily cross membranes, always be cognisant of the potential for fetal expo-
such as the human placenta. The degree of binding sure; where possible this should be minimised, for

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
490 Syme et al.

example through selection of pharmaceuticals with cotyledon contains a villus tree formed on the fetal
poor placental transfer. On the other hand, when side of term placenta (figure 3). The villus tree
drugs are administered to the mother for the pharma- consists of a central fetal capillary, villus stroma and
cological benefit of the foetus, it is necessary that an outer trophoblast layer that is bathed in maternal
these drugs reach therapeutic concentrations in the blood. This outer layer is comprised of a multinucle-
fetal circulation. In both of these scenarios the im- ated syncytiotrophoblast membrane formed by fu-
pact of placental metabolism and uptake remains sion of mononuclear cytotrophoblastic stem cells.
difficult to quantify. Each trunk and its branches float in a space limited
This review discusses the current understanding on the maternal side by the basal plate, on the fetal
of drug transfer and metabolism in the human pla- side by the chorionic plate and laterally by decidual
centa and its clinical implications. septa. Some branches of the villus trunk are attached
to the basal plate and are called anchoring villi.
1. Placental Anatomy and Physiology In the first 2 months of pregnancy, the major
organs of the human embryo develop (organogene-
Mammalian placentas can be classified into three sis) and its shape becomes clearly defined. At the
general types according to the number of layers end of the second month, the embryo becomes a
between maternal and fetal blood: (i) haemochorial foetus. During the following 7 months, the foetus
(rat, rabbit, guinea pig); (ii) endotheliochorial (cat, increases in size and weight and its organs undergo
dog); and (iii) epitheliochorial (sheep, pig, horse).[11] maturation at the histological and functional levels.
The structural differences in placentas from differ- Maternal blood does not perfuse the placenta during
ent species affects their function. In particular, the the embryonic period and the feto-placental-mater-
transfer and metabolism of drugs varies enormously nal circulation does not become established until
between species, making data from animal studies around the tenth week of pregnancy. Hence, drugs
difficult to interpret with respect to predicting feto- and other chemicals that are present in the maternal
maternal drug disposition in human pregnancy.[11] blood during the first 10 weeks of gestation must be
The human placenta is of the haemochorial type, in delivered to the developing embryo via diffusion
which the fetal tissue is in direct contact with the through extracellular fluid in order to interfere with
maternal blood (figure 2).[12] It is composed morphological differentiation. Chemical or drug-
primarily of fetal villus tissue, covered at the mater- induced dysmorphogenicity has been recognised in
nal face with decidual membrane which lines the animals for decades.[15-17] However, the clinical im-
pregnant uterus. The layer of tissue separating plications of these experimental results were dra-
maternal and fetal circulations consists of the endo- matically demonstrated in 1961 with the pandemic
thelium of the fetal capillaries and the trophoblast, of thalidomide-induced phocomelia and other em-
containing the villus stroma, the cytotrophoblast and bryopathies.[18]
the syncytiotrophoblast.[11] In early pregnancy, this
tissue layer is about 50–100μm in thickness, which 2. Drug Transfer
decreases to 4–5μm in late gestation as the placenta
grows and matures.[12] This thinning is mainly due to Transplacental exchanges are known to involve
the partial disappearance of the cytotrophoblastic passive transfer, active transport, facilitated diffu-
layer.[11] The placenta contains organelles which are sion, phagocytosis and pinocytosis.[19] Only the pas-
essential for biochemical synthesis and exchange, sive transfer of drugs across the placenta has re-
such as mitochondria, ribosomes, pinocytotic vacu- ceived much attention by investigators, although
oles and lipid enclosures. active transport systems are being identified for an
The human placenta is composed of 20–40 com- increasing number of drugs. It appears that phagocy-
partments called cotyledons, each functioning as a tosis and pinocytosis are too slow to have any signif-
vascular unit within the placenta.[11,14] Every icant influence on the extent of transfer of drugs.

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 491

Decidua parietalis
Umbilical vein Umbilical arteries
(O2-rich blood) (O2-poor blood) Smooth chorion
Amniochorionic membrane
Fetal circulation Intervillous space Amnion
Chorionic plate
Stump of
Main stem villus main stem villus

Placental septum

Anchoring villus
Decidua basalis
Cytotrophoblastic shell
Myometrium
Endometrial Endometrial
veins arteries

Maternal circulation
Fig. 2. Schematic drawing of a transverse section through a full-term human placenta. The villus branches are bathed in maternal blood and
provide a large surface area for transplacental exchange. Oxygen-deficient fetal blood is pumped through two umbilical arteries and into the
villus trees where exchange of gases, nutrients, waste products and xenobiotics takes place. The oxygen-rich blood re-enters the fetal
compartment through the umbilical vein (from Moore and Persaud,[13] with permission).

2.1 Passive Transfer how readily the drug will pass. The eventual result is
for concentrations on either side of the placenta to
Passive transfer is the predominant form of ex- equalise (unless the rate of transfer is too slow for
change in the placenta and represents the permeation equilibrium to be reached). The maternal plasma
of a molecule down its concentration gradient. Pas- concentration at steady-state conditions (Css) is de-
sive diffusion does not require the input of energy, is pendent on the overall maternal clearance and the
not saturable and is not subject to competitive inhi- rate of drug entry into the body (equation 1):
Rate in
bition. When drugs cross the placenta by passive Css =
diffusion, the amount that crosses in any given time CL
is dependent on the concentration of the drug in the (Eq. 1)
maternal circulation, its physicochemical properties where ‘Rate in’ = rate of maternal drug adminis-
and the properties of the placenta that determine tration and CL = maternal clearance.

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
492 Syme et al.

Endothelium of
fetal capillary

Placental Connective tissue


Arteriocapillary core of villus
venous network membrane

Fetal capillaries

Hofbauer
Branch villus cells b
Cytotrophoblast
Syncytiotrophoblast

Nuclear aggregation
Persisting or syncytial knot
cytotrophoblast cells
Syncytiotrophoblast

Fibrinoid Oxygen-rich
material fetal blood

Oxygen-poor
Chorionic plate
blood in fetal
Vein capillary
Amnion
Arteries Placental
membrane
a c
Fig. 3. (a) Structure of the placental villus tree. Fetal blood flows into the villus stem arteries from the chorionic plate arteries and then into
the arteriocapillaries in the villus branches where the main exchange of material between the mother and foetus occurs. (b) and (c),
drawings of cross-sections through a villus branch at 10 weeks and full term, respectively. The placental membrane (barrier) consists of the
syncytiotrophoblast, cytotrophoblast and fetal capillary endothelium. This membrane decreases in thickness throughout gestation due to
thinning of the syncytiotrophoblast and the break-up of the cytotrophoblastic layer. Hofbauer cells are believed to be phagocytic cells (from
Moore and Persaud,[13] with permission).

The overall maternal plasma clearance will where % unboundM = percentage of drug un-
largely depend on the intrinsic ability of the liver to bound in the maternal circulation, % unboundF =
metabolise the drug or the kidney to excrete it, or a percentage of drug unbound in the fetal circulation,
combination of both, depending on the physico- pHF = pH of the fetal circulation, pHM = pH of the
chemical properties of the drug. A modified form of maternal circulation, CLMF = clearance from the
the Henderson-Hasselbach equation can be used to maternal circulation into the fetal circulation, CLFM
calculate the fetal/maternal free drug concentration = clearance from the fetal circulation into the mater-
ratio by correcting for differences between the un- nal circulation and CLF = clearance from the fetal
bound fractions, the degree of ionisation and the circulation.
clearances from the mother and foetus (equation
2.1.1 Drug Properties Affecting Transfer
2):[20]
Passive diffusion is favoured for low-molecular-
F/ M =
weight and highly lipid-soluble drugs that are pre-
% unbound M 1 + 10pKa − pH F CL MF dominantly un-ionised.[21] The placenta resembles a
× pKa − pH
× lipid bilayer membrane, so only the non-protein-
% unbound F 1 + 10 M CL FM + CL F bound portion of a drug is free to diffuse across it,
(Eq. 2) although substances that transfer by a mechanism

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 493

other than passive diffusion may not necessarily es an equilibrium across the placenta between the
follow these principles.[5] maternal and fetal circulations, unless the rate of
Drugs with a molecular weight >500Da often transfer across the placenta is so slow that it never
have incomplete transfer across the placenta.[22] reaches equilibrium.[5] Although plasma protein
Drugs with a molecular weight >1000Da cross binding alters the total concentration of drugs in the
very poorly. For example, various heparins (mo- maternal and fetal plasma, the free concentrations in
lecular weight range 3000–15 000Da) do not trans- the mother and foetus will not differ.[5] Only un-
fer across the placenta because of their relatively bound drug is pharmacologically active; neverthe-
high molecular weight.[23-26] Since most drugs have less, understanding drug binding is important in the
a molecular weight <500Da, size rarely limits the interpretation of the fetal/maternal total drug con-
rate of drug transfer through the placenta. Lipid centration ratio at steady state.
solubility is also an important factor determining the Although a number of different plasma proteins
transfer of a drug across a biological membrane. have been implicated in drug binding, only albumin
Drugs that are lipid-soluble will pass easily and α1-acid glycoprotein have been shown to bind a
through the placenta, whereas more hydrophilic wide variety of drugs. Generally, albumin is seen as
drugs are generally impeded. The partition co- the binding protein for acidic lipophilic drugs and
efficient between octanol and a pH 7.4 aqueous α1-acid glycoprotein for basic lipophilic drugs.[28,29]
buffer is a useful measure of lipophilicity and is Maternal α1-acid glycoprotein concentrations re-
often used to predict the transfer of drugs across the main relatively constant throughout gestation,
placenta.[27] whereas fetal levels increase from about 10% of the
Most drugs are weak acids or bases and thus maternal level at 12 weeks to 30–40% at term.[30]
dissociate at physiological pH. In its ionised form, a This means that the free fraction of basic drugs, such
drug is charged and typically cannot pass through as bupivacaine and alprenolol, is elevated in the
the lipid membrane of the placenta. Differences foetus.[31,32] In contrast, there is a striking reduction
between the fetal and maternal pH influence the in the maternal albumin concentration during gesta-
fetal/maternal concentration ratio for free drug, but tion, although fetal serum albumin concentrations
do not change the un-ionised concentrations. Fetal increase with gestational age.[33,34] Thus, there is a
pH is lower than maternal pH, so fetal concentra- gradual rise in the fetal-maternal serum albumin
tions of bases should exceed those in the maternal concentration ratio from about 30% at 12 weeks to
circulation at equilibrium.[21] Under normal condi- about 120% at term.[33,34] Moreover, some disease
tions, fetal pH is only about 0.1 pH units below that processes and therapeutic procedures reduce mater-
of the mother and the pH differential can be disre- nal protein binding (e.g. preeclampsia, aggressive
garded. However, fetal pH may fall considerably fluid hydration prior to regional anaesthesia) and
lower in cases of fetal compromise, resulting in may increase drug transfer to the foetus.[5,35] Predict-
decreased transfer of basic drugs from the fetal to ing how the placental transfer of any particular drug
the maternal compartment. For example, fetal con- will be influenced by changes in fetal or maternal
centrations of lidocaine increase during labour and plasma protein concentrations is often difficult. For
delivery and can lead to greater negative effects in example, digoxin is only 25% bound to plasma
the foetus or neonate.[20] proteins in maternal plasma but its placental transfer
is greatly dependent on the albumin concentration
The complex formed when drugs bind to plasma
during in vitro placental perfusion experiments.[36,37]
proteins is too large to cross the placenta. However,
binding is a transitory phenomenon, so when the Factors other than differing plasma protein con-
drug separates from the protein, it can cross the centration in the fetal and maternal circulations may
placenta before it binds to another protein binding also affect the fetal/maternal total drug concentra-
site. Usually the unbound drug eventually establish- tion ratio. Fetal and maternal forms of albumin have

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
494 Syme et al.

been shown to have a different structure, with mater- ing affects transfer is probably dependent on the
nal albumin exhibiting higher affinity for some affinity of the drug for both tissue and plasma pro-
drugs, such as local anaesthetics, phenobarbital and teins. For example, by increasing the circulating
phenytoin, whereas for other drugs such as salicy- fetal albumin concentration in an in vitro perfusion
lates it is lower.[38-40] Also, binding to plasma pro- system, Dancis et al.[47] showed that there was an
teins is a competitive process between drugs and increase in the fetal steroid concentration resulting
endogenous ligands, especially free fatty acids.[41,42] from more drug being released from the tissue to the
The concentration of free fatty acids at term is fetal side.
approximately three times higher in maternal com-
pared with fetal plasma.[41,42] The greater maternal 2.1.2 Maternal, Placental and Fetal Characteristics
concentration may result in a higher than expected Affecting Drug Transfer
fetal/maternal plasma concentration ratio for some Distribution from the maternal circulation into
drugs such as diazepam.[41,42] However, differences the placenta is the first step for a drug to transfer
between fetal and maternal protein binding are across the placenta and is primarily a function of the
mostly due to the different concentrations of these uterine blood flow and the permeability of the pla-
proteins in maternal and fetal plasma, rather than cental membrane. Placental transfer for a particular
varying protein structures or competing endogenous drug can generally be referred to as either permea-
ligands.[5] bility-limited (hydrophilic drugs), flow-limited (li-
A number of drugs have also been found to bind pophilic drugs), or both. The rate of passage of a
to placental tissue, which accumulates the drugs by drug across a membrane by passive diffusion is
acting as a depot. How this ‘depot phenomenon’ directly linked to the surface area available for
observed in vitro can be extrapolated to the in vivo exchange (3.4–12.6m2), membrane thickness
situation is unclear. The transfer of any lipophilic (4–100μm) and blood flow perfusing the membrane
drug across the placenta first involves the uptake of (50–600 mL/min).[12,48] The large range in these
the drug into the syncytiotrophoblast.[43] If the affin- parameters is due to changes in placental structure
ity for the tissue is high, the drug will not readily be throughout pregnancy.[12] Passive diffusion can be
released into the fetal circulation.[44] Thus, high described by Fick’s Law (equation 3):
lipophilicity does not necessarily translate into high D × S × (CM − CF )
Vdiff =
placental drug transfer. Using the in vitro placental a
perfusion model, Nanovskaya et al.[45] found that (Eq. 3)
buprenorphine was sequestered into the placenta, where Vdiff = rate of diffusion, D = diffusion
reducing its maternal and fetal concentrations. coefficient, S = surface area of exchange, CM =
During the subsequent 2-hour washout period, the concentration in the maternal circulation, CF = con-
tissue-bound buprenorphine was released slowly centration in the fetal circulation and a = thickness
into both circulations. Ala-Kokko et al.[46] showed of the placenta.
that although bupivacaine transferred from the
The placenta undergoes considerable change
maternal side more rapidly than lidocaine due to its
throughout gestation as part of the normal matura-
higher lipophilicity, less drug appeared in the fetal
tion process, and sometimes as a result of disease.[49]
circulation due to placental sequestration.
The combination of increasing placental surface
Ala-Kokko and colleagues[44] have suggested that area and placental thinning during gestation keeps
the distribution of a compound in the maternal- up with the increasing nutritional and energy re-
placental-fetal unit is determined primarily by the quirements of the foetus. The average exchange area
relative affinities of the drug for the maternal and (i.e. the villus surface) ranges from 3.4 m2 at 28
fetal plasma proteins and the placental tissue. This weeks gestation to 12.6 m2 at term.[48-50] The diffu-
means that the extent to which plasma protein bind- sion distance across the placenta decreases from

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 495

50–100 μm at the second month to 4–5 μm at needs of the foetus would not be met by passive
term.[12] diffusion alone.[55] So far only a few drugs have
Most drug molecules are small and relatively been suggested to be transported across the placental
lipid-soluble. Therefore, the limiting factor in their barrier by facilitated diffusion. Ganciclovir has been
rate of placental transfer is placental blood flow.[51] demonstrated to be taken up into maternal-facing
At term, blood enters the spaces between the villi syncytiotrophoblast vesicles by a carrier-dependent
from over 100 spiral arteries; over 150mL of blood system.[56] However, overall transfer across the per-
may be contained in these spaces at a given time. fused placental cotyledon from the maternal-to-fetal
The rate of blood flow increases during gestation circulations was a passive process. This suggests the
from about 50 mL/min at 10 weeks to 600 mL/min rate-limiting transfer mechanism for ganciclovir is
at term.[12] Changes in blood flow will not affect passive diffusion and facilitated diffusion is only
mean steady-state concentrations but under non- involved in the initial uptake into the syncytio-
steady-state conditions may accelerate or delay trophoblast. Placental carrier-mediated transport
transfer. The intermittent decreases in placental systems have also been found in maternal-facing
blood flow during labour and delivery may not only syncytiotrophoblast membrane vesicles for the
delay the transfer of drugs to the foetus, but may cephalosporin cephalexin and glucocorticoids.[57,58]
also delay the clearance of drugs already in the It seems likely that the facilitated diffusion of drugs
foetus. Even in pregnant women who are only into the syncytiotrophoblast from the maternal side
slightly hypertensive, there are notable changes in is actually intended for structurally related endoge-
local blood-flow patterns that may affect the placen- nous compounds, such as hormones and nucleo-
tal transfer of many substances.[52] sides.
In foetuses with severe cardiac insufficiency,
there may be an elevated umbilical venous pressure 2.3 Active Transport
that may lead to placental oedema. Clinical observa-
tions suggest that placental transfer of various drugs Active transport across the placental membrane
may be reduced in patients with an elevated umbili- by protein ‘pumps’ requires energy, usually by
cal venous pressure and therapy may therefore be- adenosine triphosphate (ATP) hydrolysis or energy
come ineffective.[53] A recent study demonstrated stored in the transmembrane electrochemical gradi-
that elevated umbilical venous pressure simulated in ent provided by Na+, Cl– or H+. Transporters pow-
perfused human placentas in vitro reduced the influx ered by electrochemical gradients are often classi-
of digoxin and flecainamide into the fetal compart- fied as secondary active co-transporters. All active
ment.[54] transporters may work against a concentration gra-
dient but can become saturated.[4] Many investiga-
2.2 Facilitated Diffusion tors have examined the placental transport systems
of nutrients such as amino acids, vitamins and glu-
Facilitated diffusion requires the presence of a cose, but little is known about the active transport of
carrier substance within the placenta and the system drugs across the placental barrier.[2,3,59] Drugs that
can become saturated at high concentrations relative are actively transported are often structurally similar
to the Michaelis-Menten constant (Km). However, to endogenous substrates. Active drug transporters
transport by this mechanism does not require any are located either in the maternal-facing brush-bor-
input of energy. Usually, the end result of this mech- der (apical) membrane or the fetal-facing basolateral
anism of transport is that the concentration in both (basal) membrane where they pump drugs into or
the maternal and fetal circulations is equal. It may be out of the syncytiotrophoblast (table I). This section
that for many substances, such as carbohydrates, will deal with active transporters identified in the
facilitated diffusion provides a means to increase placenta which have demonstrated functional ac-
transport rates when the functional and metabolic tivity towards various drugs. Readers are directed to

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
496 Syme et al.

Table I. Localisation and function of placental drug transporters involved in drug transfer
Active transporter Physiological function in placenta Location in placenta Substratesa
P-glycoprotein (PGP) Fetal-to-maternal transfer of Apical syncytiotrophoblast Digoxin, ciclosporin, saquinavir,
hydrophobic cationic vincristine, vinblastine, paclitaxel,
compounds dexamethasone, sirolimus,
quinidine, terfenadine,
ondansetron, loperamide
Multidrug resistance protein 1 Fetal-to-maternal transfer of Capillary endothelial cells, Methotrexate, etoposide,
(MRP1) glutathione, sulfate and apical syncytiotrophoblast vincristine, vinblastine, cisplatin,
glucuronide conjugates HIV protease inhibitors
(dianonic sulfated bile salts)
Multidrug resistance protein 2 Fetal-to-maternal transfer of Apical syncytiotrophoblast Etoposide, cisplatin, doxorubicin,
(MRP2) glutathione, sulfate, and vincristine, vinblastine,
glucuronide conjugates methotrexate, paracetamol
(dianionic sulfated bile salts, glucuronide, grepafloxacin,
bilirubin glucuronide, ampicillin
estradiol glucuronide)
Multidrug resistance protein 3 Fetal-to-maternal transfer of Capillary endothelial cells Methotrexate, etoposide
(MRP3) anionic conjugates and apical
syncytiotrophoblast
Breast cancer resistant protein Unknown Probably apical membrane Topotecan, mitoxantrone,
(BCRP) doxorubicin, daunorubicin
Serotonin transporter (SERT) Serotonin transfer Apical syncytiotrophoblast Amfetamines
Norepinephrine transporter Dopamine and norepinephrine Apical syncytiotrophoblast Amfetamines
(NET) transfer
Extraneuronal monoamine Serotonin, dopamine, Probably basal Amfetamines, imipramine,
transporter (OCT3) norepinephrine, histamine syncytiotrophoblast desipramine, clonidine, cimetidine
transfer
Novel organic cation Maternal-to-fetal transfer of Basal syncytiotrophoblast Metamfetamine, quinidine,
transporters (OCTN) carnitine verapamil, pyrilamine
Monocarboxylate transporters Fetal-to-maternal transfer of Apical syncytiotrophoblast, Valproic acid
(MCT) lactate and pyruvate and possibly basal
syncytiotrophoblast
Dicarboxylate transporters Maternal-to-fetal transfer of Apical syncytiotrophoblast Unknown
(NaDC3) succinate and α-ketoglutarate
Sodium/multivitamin transporter Maternal-to-fetal transfer of Apical syncytiotrophoblast Carbamazepine, primidone
(SMVT) biotin and pantothenate
a Not all have been examined in the human placenta.

the recent review article by Ganapathy et al.[60] and mdr1b in rodents) and class II (MDR2 or 3 in
workshop report by St-Pierre et al.[61] for a more humans and mdr2 in rodents).[62] These large multis-
detailed examination of all active transporters ex- panning membrane proteins (170kDa) are mainly
pressed in the placenta that are involved, or thought found in luminal or apical membranes of a range of
to be involved, in drug transfer. epithelia in the body, where they function as ATP-
2.3.1 P-Glycoprotein
driven efflux pumps, thereby limiting intracellular
The multidrug resistant gene (MDR1) product, P- accumulation of xenobiotics.[63] Compounds trans-
glycoprotein, is a member of the ATP-binding cas- ferred by P-glycoprotein are usually uncharged or
sette (ABC) transporter family. It has received a basic in nature and can range in size from 200 to
great deal of attention by researchers because of its more than 1800Da.[63] The physiological role of
recognised importance in the disposition of a wide P-glycoprotein is not completely understood, but it
variety of drugs. P-glycoprotein consists of two has been postulated that this transporter evolved as a
subclasses, class I (MDR1 in humans, mdr1a and protective mechanism against the potential toxic

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 497

effects of environmental toxins and other xenobiot- than wild-type mice.[77] These studies suggest that
ics.[60] However, in some cells it has been shown to P-glycoprotein plays a major fetoprotective role,
extrude the natural ligands estradiol-17β-D-glucur- although the danger of extrapolation from murine to
onide, opioid neuropeptides and platelet-activating human pregnancy should be reiterated.
factor.[64-66] Pharmaceutical substrates of P-glyco-
protein are structurally unrelated; they include some 2.3.2 Multidrug Resistance Protein
anticancer drugs, immunosuppressive agents, HIV
protease inhibitors, central nervous system active Another important group of human ABC trans-
drugs and cardiovascular drugs.[67-69] porters is the members of the multidrug resistance
protein (MRP) family.[78] The MRP family consists
In the placenta, P-glycoprotein is expressed in the of seven members, designated MRP1 to MRP7.[79]
trophoblast cells of the brush-border membrane, but Typically, MRPs are larger (190–200 kDa) than
not the basal membrane.[70,71] Few functional studies other ABC proteins, containing about 250 additional
have been carried out to confirm the activity of amino acids.[80] MRP1, MRP4 and MRP5 proteins
P-glycoprotein in the placenta. Ushigome et al.[72] are widely distributed in the body, whereas MRP2,
demonstrated that P-glycoprotein regulates the MRP3 and MRP6 appear to be mainly expressed in
transfer of vincristine, vinblastine and digoxin into the liver, kidney and gut.[81,82] MRP proteins media-
trophoblast cells. The fetal-to-maternal transfer of te ATP-dependent transport of unconjugated,
these drugs across the trophoblast monolayer was amphiphilic anions and of lipophilic compounds
shown to be greater than transfer in the opposite conjugated to glutathione, glucuronate and sul-
direction.[72] In the presence of P-glycoprotein in- fate.[79,83] A wide array of endogenous ligands have
hibitors, this transport was reduced, which resulted been identified for the MRPs (summarised in recent
in increased fetal uptake.[72] Pávek et al.[73] recently reviews by Gerk & Vore and König et al.).[78,84]
demonstrated that P-glycoprotein pumps ciclosporin Drugs transported by MRPs are the anticancer
out of trophoblast cells in the in vitro perfused rat agents methotrexate, etoposide, vincristine, vinblas-
placenta which restricted the passage of ciclosporin tine and cisplatin, HIV protease inhibitors, paraceta-
into the fetal circulation. mol (acetaminophen) glucuronide and the antibac-
The mdr1a (P-glycoprotein) knockout (–/–) terials grepafloxacin and ampicillin.[61,79,84]
mouse has provided a unique and valuable pharma- In immunohistochemical studies, human placen-
cological tool to examine the function of P-glyco- ta at term was found to express at least three mem-
protein in various tissues in vivo.[74,75] Lankas et bers of the MRP family: MRP1, MRP2 and
al.[76] showed that administration of an isomer of the MRP3.[85-87] MRP1 and MRP3 were particularly
pesticide avermectin was associated with a 100% abundant in fetal endothelial cells of the placenta
incidence of fetal cleft palate in these mice. Hetero- microcapillary.[86] MRP3 was expressed to a lesser
zygous (+/–) mice were less sensitive and homozy- degree in the apical membrane of the syncytio-
gous (+/+) foetuses, with abundant P-glycoprotein, trophoblast, and both the presence and absence of
were totally insensitive at the doses tested. Further- MRP1 in the apical membrane of the syncytio-
more, it was found that the degree of chemical trophoblast of term placenta has been reported.[85-87]
exposure was inversely related to the expression of MRP2 was detected in the apical membranes of the
P-glycoprotein, which was determined by fetal gen- syncytiotrophoblast and not in fetal blood ves-
otype.[76] Smit et al.[77] used mdr1a/1b (–/–) mice to sels.[86] The physiological function of MRP-related
assess the role of P-glycoprotein in fetal toxicologi- proteins in the syncytiotrophoblast layer remains
cal protection against digoxin, saquinavir and pacli- speculative, although it seems likely that they could
taxel. They found that 2.4-, 7- and 16-fold more exert a feto-protective role by the removal of meta-
digoxin, saquinavir and paclitaxel, respectively, en- bolic end products from the foetus to the moth-
tered the fetal circulation of mdr1a/1b (–/–) mice er.[86,88]

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
498 Syme et al.

2.3.3 Breast Cancer Resistant Protein cental OCT3, although further research is needed to
The ATP-driven breast cancer resistant protein confirm this.[60]
(BCRP) [also called placenta-specific ABC trans-
porter or ABCP] is also part of the ABC transporter 2.3.5 Novel Organic Cation Transporters
family and is highly expressed in the placenta.[89] As The novel Na+-driven organic cation transporter
yet, its specific localisation in the placenta is un- 2 (OCTN2) is expressed in the basal membrane of
known, but is suspected to be on the apical side. the syncytiotrophoblast and transports carnitine
BCRP can render tumour cells resistant to the anti- across the placenta from the maternal circulation
cancer drugs topotecan, mitoxantrone, doxorubicin into the fetal circulation.[98,99] Placental OCTN2 also
and daunorubicin.[90] In pregnant mice, it has been transports metamfetamine, quinidine, verapamil and
demonstrated that placental BCRP restricts the pas- pyrilamine, which compete with carnitine for the
sage of topotecan and mitoxantrone to the foetus.[91] transporter, restricting its transfer across the placen-
ta.[99,100] OCTN1 has been found in the rat placenta
2.3.4 Monoamine Transporters and interacts with desipramine, dimethylamiloride,
cimetidine, procainamide and verapamil.[101]
The three monoamine transporters identified in
the placenta are the serotonin transporter (SERT),
the norepinephrine transporter (NET) and the ex- 2.3.6 Monocarboxylate Transporters and the
traneuronal monoamine transporter (OCT3).[92-94] Dicarboxylate Transporter
SERT and NET are located in the brush-border Monocarboxylates (e.g. lactate) and dicarboxy-
membrane of the placental trophoblast where they lates (e.g. succinate) are actively transported in the
transport serotonin and dopamine and norepine- placenta. Both the monocarboxylate transporters
phrine, respectively, using the transmembrane Na+ (MCTs) and the dicarboxylate transporter (NaDC3)
and Cl– electrochemical gradient.[92,93] Monoamine are expressed in the brush border membrane of the
transporters may play a role in maintaining optimal placenta, although MCTs may also be present in the
blood flow to the placenta by controlling the placen- basal membrane.[102-104] These transporters are elec-
tal transfer of vasoactive compounds.[92,93] NET, and trochemical gradient-driven; MCTs are H+-coupled
to a lesser degree SERT, have been shown to trans- and NaDC3 is Na+-coupled.[103,105] Little is known,
port amfetamine and its derivatives into the syncyti- however, about the potential of these transporters to
otrophoblast from the maternal side.[95] On the other influence drug transfer across the placenta. Valproic
hand, cocaine and nontricyclic antidepressants bind acid is frequently administered for the treatment of
to these transporters with high affinity without being all forms of epilepsy during pregnancy even though
transferred across the membrane.[93] Moreover, it there is a risk of fetal toxicity, including teratogenic-
has been suggested that cocaine may enhance the ity.[106] Although valproic acid, with a pKa of 4.9,
expression of NET in the placenta.[96] It has also behaves as an anionic ion under physiological pH, it
been demonstrated that cannabinoid receptors ex- easily crosses the placenta. Its fetal/maternal blood
pressed in the placenta play a role in the regulation concentration ratio is reported to be 1.71.[107] Re-
of SERT activity.[97] Thus, it seems likely that the cently, it has been demonstrated that a saturable
use of cannabis during pregnancy could affect the active transport system exists for the uptake of val-
placental clearance of serotonin via SERT. OCT3 is proic acid in the brush-border membrane of tropho-
thought to be located in the basal membrane where it blast cells.[108-110] This transport system includes an
actively transports serotonin, dopamine, norepine- H+-coupled MCT that causes the high rate of valpro-
phrine and histamine as physiological substrates via ic acid transfer to the foetus across the placental
a Na+ and Cl– independent system.[60,94] Several barrier. Although valproic acid and lactate compete
drugs, such as amfetamines, imipramine and desi- for the MCT mechanism, valproic acid appears to be
pramine, may also be actively transported by pla- a substrate for more than one transporter.[110]

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 499

2.3.7 Sodium/Multivitamin Transporter Table II. Cytochrome P450 (CYP) expression and activity in first-
trimester and term human placenta[6,10,113,116-118,121-123,126-131]
Vitamins biotin and pantothenate are physiologi-
CYP isoenzyme First trimester Term
cal substrates for the Na+-dependent sodium/multiv-
CYP1 1A1 +abc +abc
itamin transporter (SMVT), which is located in the
1A2 +a –ab
brush-border membrane of the placenta.[111,112] 1B1 +a +a
SMVT appears to regulate the maternal-to-fetal CYP2 2A6 – a
–a
transfer of these compounds by pumping them into 2A7 – a
–a
the syncytiotrophoblast from the maternal blood. 2A13 –a –a
There is some speculation that anticonvulsant drugs, 2B6 –a –a
such as carbamazepine and primidone, compete 2B7 –a –a
with biotin and pantothenate for the SMVT trans- 2C +a –a
porter, thus reducing the placental transfer of these 2D6 +a, –c –a, –c
endogenous compounds to the foetus.[60] 2E1 +ab, –/+c +a, –/+bc
2F1 +a +a
CYP3 3A3 ? +a, –b
3. Drug Metabolism
3A4 +ab +a, –/+b,
–c
Although the notion that the placenta acts as a
3A5 +ab +a, –/+b
metabolic barrier to xenobiotics has some attrac-
3A6 +ab +a, –b
tions, it seems that for most drugs placental metabol-
3A7 +ab –/+ab
ism is relatively minor and is not a significant factor CYP4 4B1 +a +ab
in limiting the extent of their passage across the a mRNA.
placenta. In some cases, however, the enzymes acti- b Immunohistochemistry/protein.
vate xenobiotic compounds making them toxic to c Activity.
the foetus.[113] There is scant evidence in the litera- + = detectable; – = undetectable; ? = unknown.
ture for the presence of phase I enzymes in the
placenta other than cytochrome P450s (CYPs) and growth of the foetus, when it is most susceptible to
only a few phase II enzymes have been well charac- the effects of teratogens, the expression of CYP
terised. genes is maximal, while later in pregnancy those
that are not urgently needed for fetal well-being can
3.1 Phase I Reactions
be switched off. With this in mind, Paakki and
CYP enzymes participate in the synthesis and coworkers[123] have recently postulated that mater-
catabolism of steroid hormones as well as nal drug abuse, by serving as an enhancing stimulus,
metabolising vitamins, fatty acids and a wide range might maintain the expression of specific CYP for-
of medicinal drugs and toxic chemicals.[114-116] The ms that would normally be switched off. A variety
placenta contains multiple CYP isoenzymes in the of maternal and environmental factors may affect
mitochondria and endoplasmic reticulum of tropho- the levels of drug metabolising enzymes in the pla-
blastic cells.[10] In the full term placenta, mRNAs for centa. Placental xenobiotic-metabolising activities
CYP1A1, 1B1, 2E1, 2F1, 3A3–7 and 4B1 have been are altered in mothers who abuse drugs, smoke,
detected, but only CYP1A1, 2E1, 3A4, 3A5, 3A7 drink alcohol, are exposed to polluted air or eat
and 4B1 have been characterised at the protein level contaminated food.[121,123-125]
(table II).[10,117,118] The type and amount of ex-
pressed CYPs varies depending on the period of 3.1.1 Cytochrome P450 (CYP) 1 Family
gestation and maternal health status.[119-121] In gener- So far, three members of the CYP1 family have
al, it appears that more CYP isoforms are expressed been detected in the human placenta: CYP1A1, 1A2
in the first trimester than at term.[118,122] It has been and 1B1.[118,126] It is well recognised that CYP1A is
suggested that during the early development and involved in the bioactivation of many compounds,

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
500 Syme et al.

such as polycyclic aromatic hydrocarbons, that lead first trimester onwards and it has been demonstrated
to adverse effects in the foetus.[10,113] CYP1A1 that ethanol is metabolised to acetaldehyde in the
mRNA, protein and activity have been detected human placenta.[118,121,139,140] Like CYP1A1,
throughout pregnancy.[117,118] CYP1A2 mRNA has CYP2E1 levels in the placenta exhibit considerable
been detected in the first-trimester placenta, but not individual variation.[141]
at term.[117,118] Interestingly, there appears to be 3.1.3 CYP3 Family
greater CYP1A expression in placentas from young-
The CYP3 family is quantitatively and qualita-
er mothers.[121] Although CYP1B1 mRNA is ex-
tively the most plentiful CYP family in the liv-
pressed in placental tissue throughout pregnancy,
er.[114,142] Even though CYP3A mRNAs and proteins
the impact of placental CYP1B1 in total placental
have been detected in human placenta, no relevant
xenobiotic-metabolising capacity is thought to be
marker activities for CYP3A enzymes (e.g. ster-
relatively insignificant.[122,132]
oid 6β-hydroxylation) have been reported so
The transcriptional inducibility of CYP1 en- far.[118,120,128,130]
zymes, particularly CYP1A1 and 1B1, is related to
the level and binding affinity of aryl hydrocarbon 3.2 Phase II Reactions
(Ah) receptors and Ah receptor nuclear translocator
(ARNT), which have been detected in high concen- 3.2.1 Uridine
trations (but with considerable interindividual vari- Diphosphate Glucuronosyltransferases
ability) from the placental cytosolic fraction at Uridine diphosphate glucuronosyltransferases
term.[122,132,133] Ah receptor mRNA persists at the (UGTs) are extensively involved in phase II meta-
same level throughout the pregnancy, but ARNT bolism, where they conjugate glucuronic acid to
mRNA levels are highest in first trimester sam- xenobiotics.[143] This conjugation is generally con-
ples.[132] There seems to be a role for constitutive sidered to be a detoxification reaction by making the
CYP1A1 in fetal development via the Ah/ARNT drug more polar and, thus, more susceptible to ex-
complex, which means that inducers and inhibitors cretion.[143] So far 15 isoforms of UGTs have been
could potentially interfere with normal fetal identified which are divided into two subgroups,
growth.[134] UGT1 or UGT2. It is now recognised that UGTs are
Cigarette smoking increases xenobiotic- present in the placenta throughout gestation and
metabolising CYP1A activities in the placenta to a probably play a major role in placental metabolic
variable extent depending on the stage of preg- activity.[7,121,144,145] Recent studies have detected
nancy.[121,135,136] The induction of placental CYP1A mRNA and protein expression of the UGT2B iso-
activity by tobacco smoking in pregnancy, partic- forms (UGT2B4, 2B7, 2B10, 2B11, 2B15 and
ularly CYP1A1, is greatest at term, with elevated 2B17) in the syncytium of both first trimester and
levels being sustained for a number of weeks after term placentas.[7,121,145] UGT1A mRNA and protein
smoking is stopped.[127,137,138] Recent data suggest expression have been detected in the first-trimester
that smoking and drinking synergistically elevate placenta, but have been undetectable at term.[7,121]
placental CYP1A activity.[121] Significant UGT activity towards marker substrates
has been demonstrated in placenta in the first trimes-
3.1.2 CYP2 Family ter and at term.[7,121,146,147] For example, Schenker et
Most studies using specific antibodies and sub- al.[147] found that 17% of olanzapine, on average, is
strates to identify the presence of CYP2A, 2B, 2C, converted to the glucuronide, which is transferred
2D and 2E isoenzymes have yielded negative results by the placenta at a rate slightly slower than the
in the placenta.[113,120,121,128] Moreover, CYP2A and parent drug. However, there appears to be high
2B mRNA has not been detected in human placenta interindividual variation in UGT expression and ac-
at any stage of pregnancy.[118,129] CYP2E1 has been tivity in the human placenta.[7,123,146] A recent study
detected at the level of mRNA and protein from the has demonstrated that placental UGT activity is

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 501

elevated by maternal smoking and is greatest in ta.[154-156] Sodha et al.[157] compared the sulfate con-
mothers who both smoke cigarettes and drink alco- jugation of the β2-adrenoceptor agonists salbutamol
hol.[121] (albuterol), ritodrine and fenoterol with that of dop-
amine using in vitro placental tissue and found that
3.2.2 Glutathione S-Transferases
although the rate of formation was slower, the de-
Glutathione S-transferases (GSTs) catalyse the gree of conjugation was similar for this group of
conjugation of glutathione to biologically active drugs as it was for dopamine. The in vitro activity of
electrophiles. Placental GST is very active through- sulfotransferase in the placenta towards the anthel-
out pregnancy.[8,127,128] Hence, GSTs may play a key mintic betanaphthol was about 1% and 8% of that in
role in protecting the foetus against electrophiles or the adult and fetal liver respectively.[158] Another
oxidative stress. So far, however, GST activity in the marker substrate, paracetamol, was not found to be
placenta has been related to hormone metabolism sulfated in the isolated perfused human placenta.[159]
rather than detoxification of xenobiotics.[127] These results suggest that sulfation in the placenta
In mammals, four classes of GSTs have been may be a significant biotransformation pathway for
identified: α, μ, π and θ.[148] Isoenzyme π, or some drugs.
GSTP1-1, is the only form of GST that has been
purified and cloned from human placenta.[149,150] It is 4. Models to Study Drug Transfer
evenly distributed among cell types in the placenta and Metabolism
and accounts for 85% of total GST activity, the
remainder being of unidentified composition. The potential for placental drug transfer and met-
abolism provides researchers with extensive and
3.2.3 Epoxide Hydrolase complex questions when giving drugs to pregnant
Epoxide hydrolase is present in a variety of women. Much of the information regarding drugs in
organs throughout the body where it catalyses the pregnancy is derived from animal studies. However,
conversion of epoxides into transglycols or trans- there are anatomical, morphological and functional
dihydrodiols.[9] Only one form of epoxide hydrolase differences among the different types of placentas
has been found in the human placenta, being detect- found across mammalian species which has pre-
able from 8–9 weeks of gestation onwards.[9] Al- cluded the general extrapolation of these results to
though well-known epoxide derivatives of pheny- humans.[160-162] During the past 25 years, in vitro
toin and valproate are recognised for their ability to techniques have been developed to study the placen-
produce birth abnormalities when administered to tal transfer and metabolism of drugs using human
pregnant mothers, it has been demonstrated that placenta and some clinical trials have been conduct-
placental epoxide hydrolase contributes very little to ed in pregnant women.[19]
these adverse outcomes.[151]
4.1 In Vitro Models
3.2.4 Sulfotransferases
Sulfotransferases are of great importance in the Several models for studying the placental transfer
sulfate conjugation of steroids and catecholamines and metabolism of nutrients, drugs and toxic chemi-
in human tissues, including the placenta, but little is cals in vitro have been established. These include:
known about their involvement in placental drug (i) perfused placental cotyledon; (ii) villus explants
metabolism. High levels of sulfated steroids and and monolayer cultures; (iii) isolated trophoblast
catecholamines are present in plasma; more than plasma membrane; and (iv) isolated transporters and
90% of dopamine in plasma exists in the sulfated receptors. These models resolve some of the ethical
form.[152] In humans, ten sulfotransferase genes are and methodological problems encountered with in
known, one of which encodes two different enzyme vivo studies, but are not without limitations. The
forms.[153] So far, it appears that many of these downside of any in vitro study of placental transfer
isoforms are expressed to some degree in the placen- and metabolism is that extrapolation of results to the

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
502 Syme et al.

pregnant woman can be problematic. In vitro mod- bolic processes can be taken into account. Tropho-
els cannot fully account for all the physiological and blast cultures are readily prepared from term placen-
biochemical variables in the mother, placenta and ta and may be allowed to differentiate in culture to
foetus and how these variables change throughout model syncytiotrophoblast function, although mon-
gestation. olayer cultures do not readily lend themselves to
transport studies. Recently, however, a method for
4.1.1 Perfused Placental Cotyledon
preparing and maintaining tight-junctioned syncyti-
The perfusion of isolated human placental
um on semipermeable membrane has been de-
cotyledon was first described in 1967.[163] The tech-
scribed, although no studies of drug metabolism or
nique has been modified and well validated in stud-
transport have yet been carried out using this proce-
ies of the transfer of several different endogenous
dure.[170]
and exogenous substances.[164-166] It has also been
Human placental choriocarcinoma cells (e.g.
used successfully to measure the effect of drugs on
BeWo, JAr and JEG cells) share many properties
placental vascular resistance and the formation of
with villus trophoblasts in terms of morphology,
endogenous compounds by the placenta.[167,168] For
biochemical markers and hormone secretion.[171]
a comprehensive description of this model, readers
These cells are commonly used for studies of the
are referred to review articles by Sastry and Bourget
placental barrier, including transport mechanisms.
et al.[43,52] Experiments can either be conducted us-
For example, a recent study using a BeWo monolay-
ing the closed (recirculating) method or the open
er demonstrated that both endogenous and synthetic
(single pass or nonrecirculating) method. In the
opioid peptides cross the placenta via a paracellular
closed model, both the maternal and fetal perfusates
route and that endogenous, but not synthetic, pep-
are recirculated which is useful for studying the
tides are metabolised by placental aminopep-
transplacental transfer and formation of metabolites.
tidase.[172]
In the open model, a period of equilibration allows
the perfusate to reach steady state and maternal-fetal
4.2 In Vivo Models
and fetal-maternal clearance can be calculated from
a simple equation. Placental perfusion is the method Any in vivo study of the placental transfer and
of choice for studying placental transport and metabolism of a substance must take into account
pharmacokinetics. However, perfusion systems ethical aspects linked to the study of metabolic
clearly require careful validation before they are processes in pregnant women. The tragedy of thalid-
routinely used to predict placental drug transfer in omide serves as a constant reminder. This event and
pregnant women.[44]
several others have led to the adoption of rules
4.1.2 Tissue Culture governing clinical trials in humans. Pregnant
The various types of preparations in this category women are usually excluded from clinical trials
include: (i) placental slices or villus explants; (ii) unless a direct benefit is expected. It is easy to
dispersed syncytiotrophoblastic monolayer cultures; understand why pharmaceutical companies gen-
(iii) trophoblast-derived cell lines; and (iv) microvil- erally label drugs as contraindicated or nonapproved
lus membrane vesicles. Subcellular fractions (e.g. for pregnant women when any harm to the foetus
microsomes) have also been prepared and will cause widespread condemnation and crippling
used.[52,169,170] These models are useful for investi- lawsuits.
gating the presence of transport systems and The fetal/maternal blood drug concentration ratio
metabolising enzymes in the placenta. Placental ex- is used as the simplest index for placental transfer in
plants offer the advantage of intact microarchitec- vivo. This is often calculated from blood samples
ture and maintenance of cell-cell interactions and taken from a peripheral vein (maternal blood) and
paracrine communications; hence the contribution from the umbilical cord (fetal blood) at the time of
of mesenchymal and endothelial cells to any meta- placental expulsion.[51,173] Many studies have esti-

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 503

mated the extent of maternal-to-fetal drug transfer correlated results obtained in the perfused human
by comparing the concentrations of drug in maternal placenta in vitro with those from pregnant macaques
blood to those in fetal blood samples after adminis- given the anti-HIV drugs zidovudine, didanosine,
tration of a single dose of drug to the mother. Some zalcitabine and stavudine.[180] They found that the
studies have used single pairs of samples to estimate placental transfer rate and extent in live primates
maternal/fetal drug disposition, but this may not be a strongly correlated with those observed in the in
reliable estimate. The fetal/maternal concentration vitro perfused human placenta.
ratio may vary widely in pairs of samples after
administration of a single maternal dose. More 5. Clinical Significance of Placental
meaningful information about drug distribution be- Transfer and Metabolism
tween maternal and fetal circulations can be pro-
vided by maternal drug infusions, beginning with a Over recent years there has been a gradual rise in
loading dose to maintain a constant maternal drug the use of pharmaceuticals during pregnancy.
concentration. The most accurate assessment also During pregnancy, the most widely taken classes of
requires the comparison of unbound drug concentra- drugs are analgesics, antiemetics, antibacterials,
tions during sustained maternal steady-state condi- tranquillisers, antihistamines and diuretics.[181,182] In
tions. However, measurement of unbound drug con- some cases, such as epilepsy or diabetes, the mother
centrations often requires larger samples of blood is continuously exposed to drugs during pregnancy
and obtaining these from a foetus may be unethical based on the premise that the benefit to the mother
and dangerous. outweighs the risk to the foetus. Drugs used to
inhibit (e.g. indometacin, nifedipine or salbutamol)
The anatomical and functional differences be-
or stimulate (e.g. oxytocin) uterine contractions are
tween mammalian placentas frequently compromise
examples of drugs administered near the end of
the translation of results of placental drug transfer
pregnancy. Fetal therapy via maternal drug adminis-
and metabolism from animals to humans. Especially
tration, such as the use of synthetic glucocorticoids
for hydrophilic molecules, placental permeability to induce fetal lung maturation in preterm labour,
varies widely among species.[174] The observed spe- will probably be employed to a greater extent in the
cies differences are predominantly a reflection of future. Taken together, this means that an under-
structural differences. The higher permeability of standing of placental transfer and metabolism of
the human and guinea pig placenta to hydrophilic drugs has significant clinical relevance for both
molecules, compared with the sheep, may be due to maternal and fetal welfare and may, in some cases,
the former having less tissue layers separating the prove to be a balancing act of risk versus benefit.
maternal and fetal bloodstreams.[175,176] The direct effect of a drug on the foetus is depen-
Non-human primates, such as rhesus macaques dent on the concentration of drug in the fetal circula-
and baboons, are sometimes used in placental trans- tion. Prior to delivery, drug measurements can be
fer experiments to provide a close-to-human obtained from the foetus only by umbilical blood
model.[177] For example, recently researchers suc- sampling. However, there are potential risks of fetal
cessfully used rhesus monkeys to deliver a transgene sampling, so this procedure is generally not permit-
to the placenta in vivo via a lentiviral vector.[178] In ted for evaluating fetal drug exposure alone. The
another study, rhesus macaques were used to mea- major determinant of fetal drug concentration is the
sure the transplacental transfer of stavudine, an anti- maternal drug concentration.[51] As maternal con-
HIV compound, and its active and inactive metabo- centration is a function of the dose administered,
lites.[179] The researchers measured significant con- estimation of the fetal exposure can be based on
centrations of both stavudine and its inactive metab- maternal drug intake. To determine this relationship,
olite in fetal plasma, but no active phosphorylated it is also necessary to know the rate and extent to
metabolites were detected. Other researchers have which the drug crosses the placenta. Using the con-

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
504 Syme et al.

centration-time course of the drug in the mother as about 50%.[196] In adults, digoxin has a narrow ther-
the starting point, the concentration in the foetus can apeutic range (0.5–2.0 μg/L) which makes know-
be determined from the placental transfer rate and ledge of its placental transfer vital when treating the
the rate of elimination by the foetus.[20] foetus. According to various reports, the fetal/mater-
Cigarette smoke is one of the most widely used nal digoxin concentration ratio ranges from 0.1 to as
harmful substances to which pregnant women are high as 0.9.[197-201] It appears that the concentration
exposed.[183,184] It is well established that maternal ratio of digoxin depends on a dynamic balance be-
smoking has a detrimental effect on fetal growth, but tween diffusion and active transport systems.[197-199]
the mechanisms underlying these effects are not Digoxin has been shown to be a very good P-
well understood.[183,184] Smoking has been shown to glycoprotein substrate both in vitro and in
alter the structure and function of human placental vivo.[69,72,77]
tissues, such as the sodium-dependent active trans- Multidrug therapy including digoxin may give
port of alanine, decreasing the capillary volume and rise to significant drug interactions in the placenta,
increasing the thickness of the trophoblastic lay- affecting the fetal tissue concentrations of the drugs
er.[185,186] It is very difficult to associate specific and treatment outcomes. For example, digoxin and
compounds in cigarette smoke with negative effects verapamil are often coadministered if the initial
because there are more than 2000 compounds in the digoxin monotherapy fails. In the treatment of fetal
smoke. Nicotine is one of the principle alkaloids of tachyarrhythmia, coadministered verapamil may en-
tobacco smoke and it has been shown to induce hance digoxin transfer into the foetus by blocking
skeletal, limb and palate malformations in ani- placental efflux by P-glycoprotein.[72] This suggests
mals.[187] Exposure of the human foetus to nicotine the possibility that pharmacological manipulation of
is associated with changes in fetal heart rate and placental P-glycoprotein is a potential option to op-
breathing movements.[188,189] Pastrakuljic and col- timise pharmacotherapy for fetal tachyarrhythmias.
leagues[190] confirmed that nicotine readily crosses
Nonsteroidal anti-inflammatory drugs (NSAIDs)
the placental barrier but found no evidence of the
are one of the most common classes of medication
nicotine metabolite cotinine in intact placental tissue
prescribed by general practitioners worldwide and
or in microsomal fractions. This contradicts pre-
have been administered in pregnancy for the preven-
vious reports that the placenta serves as a metabolic
tion of preterm labour.[202,203] Pregnant women fre-
barrier to nicotine.[191]
quently report that they have been taking these drugs
In 1980, the first reports of transplacental treat- despite their potential to cause adverse effects in the
ment of fetal supraventricular tachycardia by mater- foetus.[204-206] Animal experiments have linked
nal administration of cardiac glycosides opened the NSAIDs to teratogenicity and other adverse effects
possibility of avoiding iatrogenic premature deliv- in the foetus.[207-209] However, human data concern-
ery by successful cardioversion in utero.[192] ing adverse effects in the foetus are less conclusive;
Supraventricular tachycardia (>180 beats/min) is a there is contradictory opinion regarding the effects
potentially life-threatening condition and is the most of NSAIDs on birth outcome.[204,205,210] The NSAIDs
common fetal arrhythmia that requires treatment tested using human in vitro placental perfusion ex-
before birth.[193,194] If the foetus is sufficiently ma- periments, such as diclofenac, sulindinac and in-
ture, delivery is an option, otherwise transplacental dometacin, cross the placenta and reach significant
drug monotherapy is usually the initial treatment of concentrations in the fetal perfusate.[211-213] Newer
choice. Antenatal therapy in preterm pregnancies cyclo-oxygenase-2 (COX-2) selective NSAIDs,
consists of the maternal administration of digoxin, such as celecoxib and rofecoxib, are being consid-
sometimes combined with flecainamide, ami- ered as potential tocolytics for the prevention of
odarone or other antiarrhythmic agents.[195] The preterm birth.[214,215] NSAIDs have been shown to
treatment success rate of digoxin monotherapy is cause the premature closer of the fetal ductus arteri-

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 505

osus as well as neonatal renal failure, but these the late stages of pregnancy, in order to increase
adverse effects appear to be less common when infant loading with the drugs. As with other HIV
using COX-2 specific NSAIDs.[216-218] COX-2 ap- protease inhibitors, the safety profile of saquinavir
pears to be an important enzyme in fetal develop- during fetal development is known only from
ment in mammals, and consequently COX-2 selec- preclinical animal reproductive studies. There is evi-
tivity may not offer any advantages over non-selec- dence that fetal hepatic microsomes may be partic-
tive NSAIDs.[219-221] Evaluation of the human ularly sensitive to this family of drugs.[229]
placental transfer of COX-2 specific inhibitors may Cisplatin is a well-known anticancer drug with
help to establish if these represent the safest group of high efficiency against several solid tumours. Re-
NSAIDs for future clinical studies as therapeutic cently, it was discovered that by coupling cisplatin
agents during pregnancy. to a bile acid moiety the resulting drug, Bamet-R2,
Infants born to mothers with HIV infection who was less permeable across the rat placenta in
are receiving antiretroviral therapy have approxi- vivo.[230] The ability of the placental barrier to pro-
mately a 25% chance of becoming infected them- tect the fetal compartment from cisplatin when it is
selves.[222] Antiretroviral drugs are administered coupled to cholylglycinate is probably due to the
both for direct maternal benefit and for the preven- existence in the plasma membrane of the trophoblast
tion of mother-to-child transmission. The reduction of carrier proteins able to transport bile acids from
of maternal plasma viral load is an important way of the foetus to the mother.[231] This research suggests
decreasing the risk of HIV transmission to the child, the potential usefulness of Bamet-R2 as an alterna-
and drugs that remain in the maternal compartment tive anticancer drug in the treatment of certain
would obviate the concern over fetal exposure to tumours during pregnancy.
toxic effects.[223] However, preloading the infant The maturational effects of corticosteroids on the
before birth with effective anti-HIV drugs might foetus were first described in 1969.[232] Since then,
have an important prophylactic effect. The HIV maternally administered dexamethasone or be-
reverse transcriptase inhibitor zidovudine is already tamethasone have been shown to facilitate fetal lung
used according to this rationale.[224] The high pla- maturation, thus reducing the incidence of neonatal
cental transfer rate of zidovudine and other reverse respiratory distress syndrome in premature in-
transcriptase inhibitors has been established, and fants.[233,234] Dexamethasone treatment also results
carries both risks and benefits for a foetus of an in enhanced maturation of the fetal brain, heart,
HIV-positive mother.[225] In contrast, the HIV prote- kidney and gastrointestinal tract.[235] Its use is asso-
ase inhibitors, which are generally considered to be ciated with a reduced incidence of periventricular
more effective drugs, do not appreciably cross the haemorrhage, necrotising enterocolitis, neonatal
placenta.[226-228] Placental P-glycoprotein is prob- death and duration of hospital stay.[236] The signif-
ably an important factor in this low penetration. One icant healthcare implications of antenatal cortico-
study has shown intravenous administration of sa- steroid therapy resulted in recommendations by the
quinavir to pregnant mdr1a/1b knockout mice re- US National Institutes of Health for their routine use
sults in seven times more drug entering fetal circula- at gestational ages of 24–34 weeks when preterm
tion than when the drug is given to wild-type mi- delivery is anticipated.[237] Since then, there has
ce.[77] Moreover, administration of P-glycoprotein been a dramatic increase in the use of antenatal
blockers to wild-type mice could completely inhibit corticosteroids.[238,239] However, dexamethasone has
placental P-glycoprotein activity towards sa- a number of potentially serious adverse effects such
quinavir.[77] These results suggest that it may be as hypertension, adrenal suppression, pneumotho-
worthwhile to examine the coadministration of ef- rax, hyperglycaemia and increased risk of sepsis.
fective P-glycoprotein inhibitors with saquinavir The placental transfer of dexamethasone appears to
(and possibly other HIV protease inhibitors) during be rapid, as measured in animals and in isolated

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
506 Syme et al.

perfused human placenta, and it also appears to in the perfused human placenta in vitro to help
undergo extensive placental metabolism.[47,240-243] It determine its potential for the treatment of pregnant
has been suggested by Varma et al.[241] that the opioid addicts.[45] Although buprenorphine is highly
dexamethasone serum gradient observed in rats is lipophilic, less than 10% of the maternal dose was
caused by its active transport from the fetal to the transferred to the fetal circuit because the placenta
maternal side. Dexamethasone is a substrate for acts as a depot for the drug. This finding is consis-
P-glycoprotein, which pumps this drug out of the tent with the relatively low incidence of neonatal
brain and other tissues throughout the body.[69,244-246] withdrawal reported after buprenorphine use during
Therefore, it may be that that maternal/fetal concen- pregnancy.[254,255]
tration ratio across the placenta hinges on a balance It is well recognised that maternal use of drugs of
between passive diffusion and affinity for P-glyco-
abuse such as cocaine and amfetamines during preg-
protein-mediated efflux.
nancy has adverse effects on the developing foe-
The development of successful strategies for de- tus.[256] The interaction between these drugs and
livering genes across the placenta may offer novel placental monoamine transporters may be a possible
therapeutic options for the foetus. Various investiga- cause of these effects.[256] Various mechanisms have
tors have considered fetal gene therapy for inherited
been suggested to describe the interaction of
disorders that cause fetal damage.[247] For example,
abusable drugs with these transporters. For example,
viral vectors are being developed that target tropho-
the norepinephrine transporter (NET) transfers
blastic cell types for the treatment of choriocarci-
metamfetamine into the syncytiotrophoblast of the
noma.[248] Recently, the isolated perfused human
placenta.[95] This causes competition between
placenta was used as a model to assess the feasibility
metamfetamine and norepinephrine for the trans-
of transferring genes to trophoblastic tissue.[249] The
porter, resulting in suboptimal transfer of the physi-
researchers found that they were able to effectively
ological substrate. The reduction in clearance of
transfect the perfused human placenta with ade-
serotonin and norepinephrine from the maternal
noviral vectors and the expression of the transgene
blood by drugs competing with their respective pla-
was detected in trophoblastic cells. For a variety of
cental transporters will increase their concentration
reasons, prenatal gene therapy may be an attractive
and could cause vasoconstriction of the uterine ar-
possibility that can overcome some of the conditions
teries and decreased blood supply to the placen-
limiting gene therapy in adults or children. The
ta.[256-258] This will, in turn, reduce the supply of
trophoblast escapes some components of host im-
oxygen and nutrients to the foetus. It has also been
munity that limit viral-based gene therapy in other
suggested that elevated levels of norepinephrine
target tissues.
may increase the incidence of premature delivery by
inducing myometrial contractile activity.[256-258]
6. Drugs of Abuse
Cannabis is one of most widely abused drugs
For decades, pregnant opioid-dependent patients during pregnancy; a recent British survey found that
have almost invariably been treated with metha- 5% of pregnant mothers smoked cannabis before
done. However, maternal methadone significantly and/or during pregnancy.[259] Δ9-Tetrahydrocan-
transfers across the placenta and can induce with- nabinol (THC) is the major psychoactive component
drawal symptoms in the newborn.[250] Bupre- of cannabis. THC exposure during the first 2 weeks
norphine is an attractive alternative for maintenance of pregnancy has been associated with embryocidal
therapy of opioid dependency because in most pa- effects in mice, as well as reduced fetal
tients it produces limited withdrawal symptoms, re- bodyweight.[260] In humans, THC has not been relat-
sults in reduced self-administration of heroin and ed to prenatal death but may cause a small reduction
has a longer duration of action.[251-253] A recent study in birthweight.[259] Moreover, prenatal THC expo-
has assessed the placental transfer of buprenorphine sure has been correlated with altered behaviour in

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 507

these children such as hyperactivity, inattention and References


impulsivity.[261,262] As would be expected from its 1. Smith CH, Moe AJ, Ganapathy V. Nutrient transport pathways
across the epithelium of the placenta. Annu Rev Nutr 1992; 12:
high lipophilicity, THC rapidly crosses the mouse, 183-206
sheep and monkey placenta in vivo; peak concentra- 2. Hahn T, Desoye G. Ontogeny of glucose transport systems in
the placenta and its progenitor tissues. Early Pregnancy 1996;
tions in the primate foetus occur within 15 minutes 2: 168-82
of a maternal intravenous injection.[260,263,264] In 3. Moe AJ. Placental amino acid transport. Am J Physiol 1995;
pregnant women who smoked cannabis daily, THC 268: C1321-31
4. Seeds AE. Placental transfer. In: Barnes AC, editor. Intrauterine
was detected in umbilical cord blood at concentra- development. Philadelphia: Lea & Febiger, 1968: 103-28
tions 2.5–6 times lower than in maternal blood at the 5. Hill MD, Abramson FP. The significance of plasma protein
time of delivery.[265] binding on the fetal/maternal distribution of drugs at steady-
state. Clin Pharmacokinet 1988; 14: 156-70
6. Meigs RA, Ryan KJ. Cytochrome P-450 and steroid biosynthe-
sis in the human placenta. Biochim Biophys Acta 1968; 165:
7. Conclusion 476-82
7. Collier AC, Ganley NA, Tingle MD, et al. UDP-glucuronosyl-
transferase activity, expression and cellular localization in
An understanding of the degree of fetal exposure human placenta at term. Biochem Pharmacol 2002; 63: 409-19
to a drug is an important aspect of pharmacological 8. Pacifici GM, Rane A. Glutathione S-transferase in the human
treatment of pregnant women. Almost all drugs placenta at different stages in pregnancy. Drug Metab Dispos
1981; 9: 472-5
cross the placenta by passive diffusion and some 9. Pacifici GM, Rane A. Epoxide hydroxylase in human placenta
reach pharmacologically significant concentrations at different stages in pregnancy. Dev Pharmacol Ther 1983; 6:
in the foetus. An increasing number of active trans- 83-93
10. Pasanen M. The expression and regulation of drug metabolism
porters have been found that are involved in placen- in human placenta. Adv Drug Deliv Rev 1999; 38: 81-97
tal drug transfer both to and from the foetus. This 11. van der Aa EM, Peereboom-Stegeman JHJC, Noordhoek J, et al.
raises the possibility of interactions when drugs Mechanisms of drug transfer across the placenta. Pharm World
Sci 1998; 20: 139-48
compete for the same transporter or inhibit its nor- 12. Kaufmann P, Scheffen I. Placental development. In: Polin RA,
mal functioning. Moreover, there is the concern that Fox WW, editors. Fetal and neonatal physiology. Philadelphia:
WB Saunders, 1992: 47-56
drugs interfere with the active transport of endoge- 13. Moore KL, Persaud TVN. Before we are born: essentials of
nous compounds essential to fetal development. embryology and birth defects, 5th edition. Philadelphia: W.B.
Sound knowledge of the transfer and metabolism of Saunders Company, 1998
14. Enders AC, Blankenship TN. Comparative placental structure.
a drug across the placenta can help distinguish be- Adv Drug Deliv Rev 1999; 38: 3-16
tween drugs that readily cross the placenta and ex- 15. Ferm VH. The rapid detection of teratogenic activity. Lab Invest
pose the foetus to significant concentrations and 1965; 14: 1500-5
16. King CT, Howell J. Teratogenic effect of buclizine and hydrox-
drugs that are blocked or metabolised sufficiently by yzine in the rat and chlorcyclizine in the mouse. Am J Obstet
the placenta to negate the concern of fetal exposure. Gynecol 1966; 95: 109-11
This understanding will hopefully improve the ef- 17. Fabro S, Smith RL. The teratogenic activity of thalidomide in
the rabbit. J Pathol Bacteriol 1966; 91: 511-9
fectiveness and safety of drug administration in 18. Rodin AE, Koller LA, Taylar JD. Association of thalidomide
pregnancy, allowing existing drugs of possible ther- (Kevadon) with congenital anomalies. CMAJ 1962; 86: 744-6
apeutic benefit to be administered during pregnancy 19. Pacifici GM, Nottoli R. Placental transfer of drugs administered
to the mother. Clin Pharmacokinet 1995; 28 (3): 235-69
without risk to the foetus, while realising the poten- 20. Garland M. Pharmacology of drug transfer across the placenta.
tial of pharmacological treatment of the foetus. Obstet Gynecol Clin North Am 1998; 25: 21-42
21. Nau H. Physicochemical and structural properties regulating
placental drug transfer. In: Polin RA, Fox WW, editors. Fetal
Acknowledgements and neonatal physiology. Philadelphia: WB Saunders, 1992:
130-41
22. Ward RM. Drug therapy of the fetus. J Clin Pharmacol 1993; 33:
Financial assistance was received from the Maurice and 780-9
Phyllis Paykel Trust and Uniservices Limited. There are no 23. Forestier F, Daffos F, Capella-Pavlovsky M. Low molecular
conflicts of interest directly relevant to the content of this weight heparin (PK 10169) does not cross the placenta during
the second trimester of pregnancy study by direct fetal blood
review. sampling under ultrasound. Thromb Res 1984; 34: 557-60

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
508 Syme et al.

24. Forestier F, Daffos F, Rainaut M, et al. Low molecular weight 44. Ala-Kokko TI, Myllynen P, Vähäkangas K. Ex vivo perfusion
heparin (CY 216) does not cross the placenta during the third of the human placental cotyledon: implications for anesthetic
trimester of pregnancy [letter]. Thromb Haemost 1987; 57: pharmacology. Int J Obstet Anesth 2000; 9: 26-38
234 45. Nanovskaya T, Deshmukh S, Brooks M, et al. Transplacental
25. Omri A, Delaloye JF, Andersen H, et al. Low molecular weight transfer and metabolism of buprenorphine. J Pharmacol Exp
heparin Novo (LHN-1) does not cross the placenta during the Ther 2002; 300: 26-33
second trimester of pregnancy. Thromb Haemost 1989; 61: 46. Ala-Kokko TI, Pienimaki P, Herva R, et al. Transfer of lido-
55-6 caine and bupivacaine across the isolated perfused human
26. Schneider D, Heilmann L, Harenberg J. Placental transfer of placenta. Pharmacol Toxicol 1995; 77: 142-8
low-molecular weight heparin. Geburtshilfe Frauenheilkd 47. Dancis J, Jansen V, Levitz M. Placental transfer of steroids:
1995; 55: 93-8 effect of binding to serum albumin and to placenta. Am J
27. Dickinson RG, Fowler DW, Kluck RM. Maternofetal transfer of Physiol 1980; 238: E208-13
phenytoin, p-hydroxy-phenytoin and p-hydroxy-phenytoin- 48. Aherne W, Dunnill MS. Quantitative aspects of placental struc-
glucuronide in the perfused human placenta. Clin Exp ture. J Pathol Bacteriol 1966; 91: 123-39
Pharmacol Physiol 1989; 16: 789-97 49. de Sweet M. Embryology. In: de Sweit M, Chamberlain G,
28. Paxton JW. α1-Acid glycoprotein and binding of basic drugs. editors. Basic science of obstetrics and gynecology. Edin-
Methods Find Exp Clin Pharmacol 1983; 5: 635-48 burgh: Churchill Livingstone, 1992: 27-52
50. Aherne W, Dunnill MS. Morphometry of the human placenta.
29. Kragh-Hansen U. Molecular aspects of ligand binding to serum
Br Med Bull 1966; 22: 5-8
albumin. Pharmacol Rev 1981; 33: 17-53
51. Reynolds F, Knott C. Pharmacokinetics in pregnancy and pla-
30. Wood M, Wood AJJ. Changes in plasma drug binding and cental transfer. Oxf Rev Reprod Biol 1989; 11: 389-449
α1-acid glycoprotein in mother and newborn infant. Clin
52. Bourget P, Roulot C, Fernandez H. Models for placental transfer
Pharmacol Ther 1981; 29: 522-6
studies of drugs. Clin Pharmacokinet 1995; 28 (2): 161-80
31. Thomas J, Long G, Morgan D. Plasma protein binding and 53. Gembruch U, Hansmann M, Bald R. B. Direct intrauterine fetal
placental transfer of bupivacaine. Clin Pharmacol Ther 1976; treatment of fetal tachyarrhythmia with severe hydrops fetalis
19: 426-34 by antiarrythmic drugs. Fetal Ther 1988; 3: 210-5
32. Herngren L, Ehrnebo M, Boréus LO. Drug binding to plasma 54. Schmolling J, Renke K, Richter O, et al. Digoxin, flecainamide,
proteins during human pregnancy and in the perinatal period: and amiodarone transfer across the placenta and the effects of
studies in cloxacillin and alprenolol. Dev Pharmacol Ther an elevated umbilical venous pressure on the transfer rate. Ther
1983; 6: 110-44 Drug Monit 2000; 22: 582-8
33. Krauer B, Dayer P, Anner R. Changes in serum albumin and 55. Folkart GR, Dancis J, Monye WL. Transfer of carbohydrates
alpha-1-acid glycoprotein concentrations during pregnancy: an across guinea pig placenta. Am J Obstet Gynecol 1960; 80:
analysis of fetal-maternal pairs. Br J Obstet Gynaecol 1984; 221-3
91: 875-81 56. Henderson GI, Hu ZQ, Yang Y, et al. Ganciclovir transfer by
34. Hamar C, Levy G. Serum protein binding of drugs and bilirubin human placenta and its effects on rat fetal cells. Am J Med Sci
in newborn infants and their mothers. Clin Pharmacol Ther 1993; 306: 151-6
1980; 28: 58-63 57. Kudo Y, Urabe T, Fujiwara A, et al. Carrier-mediated transport
35. Herman NL, Li AT, Van Decar TK, et al. Transfer of system for cephalexin in human placental brush-border mem-
methohexital across the perfused human placenta. J Clin brane vesicles. Biochim Biophys Acta 1989; 978: 313-8
Anesth 2000; 12: 25-30 58. Fant ME, Yeakley J, Harrison RW. Evidence for carrier-medi-
36. Schmolling J, Jung S, Reinsberg J, et al. Digoxin transfer across ated transport of glucocorticoids by human placental mem-
the isolated placenta is influenced by maternal and fetal albu- brane vesicles. Biochim Biophys Acta 1983; 731: 415-20
min concentrations. Reprod Fertil Dev 1996; 8: 969-74 59. Bissonnette JM. Placental transport of carbohydrates. Mead
37. Tsadkin M, Holcberg G, Sapir O, et al. Albumin-dependent Johnson Symp Perinat Dev Med 1981; 18: 21-3
digoxin transfer in isolated perfused human placenta. Int J Clin 60. Ganapathy V, Prasad PD, Ganapathy ME, et al. Placental
Pharmacol Ther 2001; 39: 158-61 transporters relevant to drug distribution across the maternal-
38. Wallace S. Altered plasma albumin in the newborn infant. Br J fetal interface. J Pharmacol Exp Ther 2000; 294: 413-20
Clin Pharmacol 1977; 14: 82-5 61. St-Pierre V, Ugele B, Gambling L, et al. Mechanisms of drug
transfer across the human placenta: a workshop report. Placen-
39. Krasner J, Giacoia GP, Yaffe SJ. Drug protein binding in the
ta 2002; 23 Suppl. A: 159-64
newborn infant. Ann N Y Acad Sci 1973; 226: 101-14
62. Gottesman MM, Hrycyna CA, Schoenlein PV, et al. Genetic
40. Reynolds F. Transfer of drugs. In: Chamberlain GVP, Wilkin- analysis of the multidrug transporter. Annu Rev Genet 1995;
son AW, editors. Placental transfer. Kent: Pitman Medical 29: 607-49
Publishing, 1979: 166-81 63. Gottesman M, Pastan I. Biochemistry of multidrug resistance
41. Nau H, Luck W, Kuhnz W. Decreased serum protein binding of mediated by the multidrug transporter. Annu Rev Biochem
diazepam and its major metabolite in the neonate during the 1993; 62: 385-427
first postnatal week relate to increased free fatty acid levels. Br 64. Huang L, Hoffman T, Vore M. Adenosine triphosphate-depen-
J Clin Pharmacol 1984; 17: 92-8 dent transport of estradiol-17β-D-glucuronide in membrane
42. Ridd MJ, Brown KF, Nation RL, et al. Differential transplacen- vesicles by MDR1 expressed in insect cells. Hepatology 1998;
tal binding of diazepam: causes and implications. Eur J Clin 28: 1371-7
Pharmacol 1983; 24: 595-601 65. King M, Su W, Chang A, et al. Transport of opioids from the
43. Sastry BVR. Techniques to study human placental transport. brain to the periphery by P-glycoprotein: peripheral actions of
Adv Drug Deliv Rev 1999; 38: 17-39 central drugs. Nat Neurosci 2001; 4: 268-74

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 509

66. Raggers RJ, Vogels I, van Meer G. Multidrug-resistance P- 83. Suzuki H, Sugiyama Y. Excretion of GSSG and glutathione
glycoprotein (MDR1) secretes platelet-activating factor. Bio- conjugates mediated by MRP1 and cMOAT/MRP2. Semin
chem J 2001; 357: 859-65 Liver Dis 1998; 18: 359-76
67. Kim RB, Fromm MF, Wandel C, et al. The drug transporter P- 84. Gerk PM, Vore M. Regulation and expression of the multidrug
glycoprotein limits oral absorption and brain entry of HIV-1 resistance-associated protein 2 (MRP2) and its role in drug
protease inhibitors. J Clin Invest 1998; 101: 289-94 disposition. J Pharm Exp Ther 2002; 302: 407-15
68. Hunter J, Jepson MA, Tsuruo T, et al. Functional expression of 85. Sugawara I, Akiyama S, Scheper RJ, et al. Lung resistance
P-glycoprotein in apical membranes of human intestinal protein (LRP) expression in human normal tissues in compari-
Caco-2 cells. Kinetics of vinblastine secretion and interaction son with that of MDR1 and MRP. Cancer Lett 1997; 112:
with modulators. J Biol Chem 1993; 268: 14991-7 23-31
69. Schinkel AH, Wagenaar E, van Deemter L, et al. Absence of the 86. St-Pierre V, Serrano MA, Macias RIR, et al. Expression of
mdr1a P-glycoprotein in mice affects tissue distribution and members of the multidrug resistance protein family in human
pharmacokinetics of dexamethasone, digoxin, and cyclosporin term placenta. Am J Physiol Regul Integr Comp Physiol 2000;
A. J Clin Invest 1995; 96: 1698-705 279: R1495-503
70. Cordon-Cardo C, O’Brien JP, Boccia J, et al. Expression of the 87. Flens MJ, Zaman GJR, van der Valk P, et al. Tissue distribution
multidrug resistance gene product (P-glycoprotein) in human of the multidrug resistance protein. Am J Pathol 1996; 148:
normal and tumor tissues. J Histochem Cytochem 1990; 38: 1237-47
1277-87 88. Cui Y, König J, Buchholz U, et al. Drug resistance and ATP-
71. Sugawara I, Kataoka I, Morishita Y, et al. Tissue distribution of dependent conjugate transport mediated by the apical mul-
p-glycoprotein encoded by a multidrug-resistant gene as re- tidrug resistance protein, MRP2, permanently expressed in
vealed by a monoclonal antibody, MRK 16. Cancer Res 1988; human and canine cells. Mol Pharmacol 1999; 55: 929-37
48: 1926-9 89. Allikmets R, Schriml LM, Hutchinson A, et al. A human
72. Ushigome F, Takanaga H, Matsuo H, et al. Human placental placenta-specific ATP-binding cassette gene (ABCP) on chro-
transport of vinblastine, vincristine, digoxin and progesterone: mosome 4q22 that is involved in multidrug resistance. Cancer
contribution of P-glycoprotein. Eur J Pharmacol 2000; 408: Res 1998; 58: 5337-9
1-10 90. Allen JD, Brinkhuis RF, Wijnholds J, et al. The mouse Bcrp1/
73. Pávek P, Fendrich Z, Staud F, et al. Influence of p-glycoprotein Mxr/Abcp gene: amplification and overexpression in cell lines
on the transplacental passage of cyclosporin. J Pharm Sci selected for resistance to topotecan, mitoxantrone, or doxo-
2001; 10: 1583-92 rubicin. Cancer Res 1999; 59: 4237-41
74. Schinkel AH, Mayer U, Wagenaar E, et al. Normal viability and 91. Jonker JW, Smit JW, Brinkhuis RF, et al. Role of breast cancer
altered pharmacokinetics in mice lacking mdr-1-type (drug- resistance protein in the bioavailability and fetal penetration of
transporting) P-glycoproteins. Proc Natl Acad Sci U S A 1997; topotecan. J Natl Cancer Inst 2000; 92: 1651-6
94: 4028-33 92. Ramamoorthy S, Leibach FH, Ganapathy V. Partial purification
75. Schinkel AH, Smit JJ, van Tellingen O, et al. Disruption of the and characterization of the human placental serotonin trans-
mouse mdr1a P-glycoprotein gene leads to a deficiency in the porter. Placenta 1993; 14: 449-61
blood-brain barrier and to increased sensitivity to drugs. Cell 93. Ramamoorthy S, Prasad PD, Kulanthaivel P, et al. Expression
1994; 77: 491-502 of a cocaine-sensitive noradrenaline transporter in the human
76. Lankas GR, Wise LD, Cartwright ME, et al. Placental P- placental syncytiotrophoblast. Biochemistry 1993; 32:
glycoprotein deficiency enhance susceptibility to chemically 1346-53
induced birth defects in mice. Reprod Toxicol 1998; 12: 94. Kekuda R, Prasad PD, Wu X, et al. Cloning and functional
457-63 characterization of a potential-sensitive, polyspecific organic
77. Smit JW, Huisman MT, van Tellingen O, et al. Absence or cation transporter (OCT3) most abundantly expressed in pla-
pharmacologcal blocking of placental P-glycoprotein pro- centa. J Biol Chem 1998; 273: 15971-9
foundly increases fetal drug exposure. J Clin Invest 1999; 104: 95. Ramamoorthy JD, Ramamoorthy S, Leibach FH, et al. Human
1441-7 placental monoamine transporters as targets for amphet-
78. König J, Nies AT, Cui Y, et al. Conjugate export pumps of the amines. Am J Obstet Gynecol 1995; 173: 1782-7
multidrug resistance protein (MRP) family: localization, sub- 96. Shearman LP, Meyer JS. Cocaine up-regulates norepinephrine
strate specificity, and MRP2-mediated drug resistance. Bi- transporter binding in the rat placenta. Eur J Pharmacol 1999;
ochim Biophys Acta 1999; 1461: 377-94 386: 1-6
79. Borst P, Evers R, Kool M, et al. A family of drug transporters: 97. Kenney SP, Kekuda R, Prasad PD, et al. Cannabinoid receptors
the multidrug resistance-associated proteins. J Natl Cancer Inst and their role in the regulation of the serotonin transporter in
2000; 92: 1295-302 human placenta. Am J Obstet Gynecol 1999; 181: 491-7
80. Hipfner DR, Deeley RG, Cole SPC. Structural, mechanistic and 98. Wu X, Huang W, Prasad PD. Functional characteristics and
clinical aspects of MRP1. Biochim Biophys Acta 1999; 1461: tissue distribution pattern of organic cation transporter 2
359-76 (OCTN2), an organic cation/carnitine transporter. J Pharmacol
81. Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a Exp Ther 1999; 290: 1482-92
transporter gene in a multidrug-resistant human lung cancer 99. Ohashi R, Tamai I, Yabuuchi H, et al. Na+-dependent carnitine
cell line. Science 1992; 258: 1650-4 transport by organic cation transporter (OCTN2): its pharma-
82. Kool M, de Haas M, Scheffer GL, et al. Analysis of expression cological and toxicological relevance. J Pharmacol Exp Ther
of cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues 1999; 291: 778-84
of the multidrug resistance-associated protein gene (MRP1), in 100. Wu X, Prasad PD, Leibach FH, et al. cDNA sequence, transport
human cancer cell lines. Cancer Res 1997; 57: 3537-47 function, and genomic organisation of human OCTN2, a new

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
510 Syme et al.

member of the organic cation transporter family. Biochem human placental xenobiotic and steroid metabolising enzymes
Biophys Res Commun 1998; 246: 589-95 in vitro. Placenta 1997; 18: 37-41
101. Wu X, George RL, Huang W, et al. Structural and functional 121. Collier AC, Tingle MD, Paxton JW, et al. Metabolizing enzyme
characteristics and tissue distribution pattern of rat OCTN1, an localisation and activities in the first trimester human placenta:
organic cation transporter, cloned from placenta. Biochim the effect of maternal and gestational age, smoking and alcohol
Biophys Acta 2000; 1466: 315-27 consumption. Hum Reprod 2002; 17: 2564-72
102. Price NT, Jakson VN, Halestrap AP. Cloning and sequencing of 122. Hakkola J, Pasanen M, Pelkonen O, et al. Expression of
four new mammalian monocarboxylate transporter (MCT) CYP1B1 in human adult and fetal tissues and differential
homologues confirms the existence of a transporter family inducibility of CYP1B1 and CYP1A1 by Ah-receptor ligands
with an ancient past. Biochem J 1998; 329: 321-8 in human placenta and cultured cells. Carcinogenesis 1997; 18:
103. Ganapathy V, Ganapathy ME, Tiruppathi C, et al. Sodium 391-7
gradient-driven, high-affinity, uphill transport of succinate in 123. Paakki P, Stockmann H, Kantola M, et al. Maternal drug abuse
human placental brush-border membrane vesicles. Biochem J and human term placental xenobiotic and steroid metabolising
1988; 249: 179-84 enzymes in vitro. Environ Health Perspect 2000; 108: 141-5
104. Balkovetz DF, Leibach FH, Mahesh VB, et al. A proton 124. Hincal F. Effects of exposure to air pollution and smoking on
gradient is the driving force for the uphill transport of lactate in the placental aryl hydrocarbon hydroxylase (AHH) activity.
human placental brush-border membrane vescicles. J Biol Arch Environ Health 1986; 41: 377-83
Chem 1988; 263: 13823-30 125. Wong TK, Everson RB, Hsu ST. Potent induction of human
105. Poole RC, Halestrap AP. Transport of lactate and other mono- placental monooxygenase activity by previous dietary expo-
carboxylates across mammalian plasma membranes. Am J sure to polychlorinated biphenyls and their thermal degrada-
Physiol 1993; 264: C761-82 tion products. Lancet 1985; I: 721-4
106. Malone FD, D’Alton ME. Drugs in pregnancy: anticonvulsants. 126. Pasanen M, Taskinen T, Sotaniemi LA, et al. Inhibitor panel
Semin Perinatol 1997; 21: 114-23 studies of human hepatic and placental cytochrome P-450-as-
107. Ishizaki T, Yokochi K, Chiba K, et al. Placental transfer of sociated monooxygenase activities. Pharmacol Toxicol 1988;
anticonvulsants (phenobarbital, phenytoin, valproic acid) and 62: 311-7
the elimination from neonates. Pediatr Pharmacol 1981; 1: 127. Pasanen M, Pelkonen O. Xenobiotic and steroid-metabolising
291-303 monooxygenases cataylsed by cytochrome P450 and gluta-
108. Utoguchi N, Audus KL. Carrier-mediated transport of valproic thione S-transferase conjugations in the human placenta and
acid in BeWo cells, a human trophoblast cell line. Int J Pharm theft relationships to maternal cigarette smoking. Placenta
2000; 195: 115-24 1990; 11: 75-85
109. Ushigome F, Takanaga H, Matsuo H, et al. Uptake mechanism 128. McRobie DJ, Glover DD, Tracey TS. Effects of gestational and
of valproic acid in human placenta choriocarcinoma cell line overt diabetes on human placental cytochromes P450 and
(BeWo). Eur J Pharmacol 2001; 417: 169-76 glutathione S-transferase. Drug Metab Dispos 1998; 26:
367-71
110. Nakamura H, Ushigome F, Koyabu N, et al. Proton gradient-
129. Koskela S, Hakkola J, Hukkanen J, et al. Expression of CYP2A
dependent transport of valproic acid in human placental brush-
genes in human liver and extrahepatic tissues. Biochem
border membrane vesicles. Pharm Res 2002; 19: 154-61
Pharmacol 1999; 57: 1407-13
111. Grassl SM. Human placental brush-border membrane Na+-pan-
130. Pienimäki P, Lampela E, Hakkola J, et al. Pharmacokinetics of
tothenate cotransport. J Biol Chem 1992; 267: 22902-6
oxcarbazepine and carbamazepine in human placenta. Epilep-
112. Grassl SM. Human placental brush-border membrane Na+-bio- sia 1997; 38: 309-16
tin cotransport. J Biol Chem 1992; 267: 17760-5
131. Pasanen M, Haaparanta T, Sundin M, et al. Immunochemical
113. Pasanen M, Pelkonen O. The expression and environmental and molecular biochemical studies on human placental ciga-
regulation of P450 enzymes in human placenta. Crit Rev rette smoke-inducible cytochrome P-450-dependent monoox-
Toxicol 1994; 24: 211-29 ygenase activities. Toxicology 1990; 62: 175-87
114. Marray M. P450-enzymes-inhibition mechanisms, genetic regu- 132. Pelkonen O, Vähäkangas K, Kärki NT, et al. Genetic and
lation and effects of liver disease. Clin Pharmacokinet 1992; environmental regulation of aryl hydrocarbon hydroxylase in
23: 132-46 man: studies with liver, lung, placenta, and lymphocytes. Tox-
115. Gonzalez FJ. The molecular biology of cytochrome P450s. icol Pathol 1984; 12: 256-60
Pharmacol Rev 1989; 40: 243-88 133. Manchester DK, Gordon SK, Golas CL, et al. Ah receptor in
116. Thompson EAJ, Siiteri PK. The involvement of human placen- human placenta: stabilization by molybdate and characteriza-
tal microsomal cytochrome P-450 in aromatization. J Biol tion of binding of 2,3,7,8-terachlorodibenzo-p-dioxin, 3-in-
Chem 1974; 249: 5373-8 ethylcholanthrene, and benzo(a)pyrene. Cancer Res 1987; 36:
117. Hakkola J, Pasanen M, Hukkanen J, et al. Expression of 4861-8
xenobiotic-metabolising cytochrome-P450 forms in human 134. Pelkonen O, Jouppila P, Kärki NT, et al. Attempts to induce
full-term placenta. Biochem Pharmacol 1996; 51: 403-11 drug metabolism in human fetal liver and placenta by the
118. Hakkola J, Raunio H, Purkunen R, et al. Detection of cyto- administration of phenobarbital to mothers. Arch Int
chrome P450 gene expression in human placenta in first tri- Pharmacodyn Ther 1973; 202: 288-97
mester of pregnancy. Biochem Pharmacol 1996; 52: 379-83 135. Pelkonen O, Arvela P, Kärki NT. 3,4-Benzo(a)pyrene and N-
119. Nelson DR, Kamataki T, Waxman DJ, et al. The P450 super- methylalanine metabolising enzymes in the immature human
family update on new sequences, gene mapping, accession fetus and placenta. Acta Pharmacol Toxicol 1971; 30: 385-95
numbers, early trivial names of enzymes, and nomenclature. 136. Pelkonen O, Jouppila P, Kärki NT. Effect of maternal cigarette
DNA Cell Biol 1993; 12: 1-51 smoking in 3,4-benzo(a)pyrene and N-methylalanine metabol-
120. Pasanen M, Helin-Martikainen RL, Pelkonen O, et al. In- ism in human fetal liver and placenta. Toxicol Appl Pharmacol
trahepatic cholestasis of pregnancy impairs the activity of 1972; 23: 29-37

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 511

137. Gurtoo HL, Williams CI, Gottlieb K, et al. Population distribu- 155. Cappiello M, Giuliani L, Rane A, et al. Dopamine sulphotrans-
tion of benzo(a)pyrene metabolism in smokers. Int J Cancer ferase is better developed than p-nitrophenol sulphotransferase
1983; 31: 385-95 in human fetus. Dev Pharmacol Ther 1991; 1991: 83-8
138. Shiverick KT, Salafia C. Cigarette smoking and pregnancy I: 156. Sodha RJ, Glover V, Sandler M. Phenolsulphotransferase in
ovarian, uterine and placental effects. Placenta 1999; 20: human placenta. Biochem Pharmacol 1983; 32: 1655-7
265-72 157. Sodha RJ, Schneider H. Sulphate conjugation of beta2-adre-
139. Rasheed A, Himes R, McCarver-May D. Variation in induction noceptor stimulating drugs by platelet and placental phenol
of human placental CYP2E1: possible role is susceptibility to sulphotransferase. Br J Clin Pharmacol 1984; 17: 106-8
fetal alcohol syndrome. Toxicol Appl Pharmacol 1997; 144: 158. Cappiello M, Franchi M, Rane A, et al. Sulphotransferase and it
396-400 substrate: adenosine-3′-phosphate-5′-phosphosulphate in
140. Karl P, Gordon B, Leiber C, et al. Acetaldehyde production and human fetal liver and placenta. Dev Pharmacol Ther 1990; 14:
transfer in the perfused human placental cotyledon. Science 62-5
1988; 242: 273-5 159. Weigand UW, Chou RC, Maulik D, et al. Assessment of
biotransformation during transfer of propoxyphene and aceta-
141. Vieira I, Pasanen M, Raunio H, et al. Expression of CYP2E1 in
minophen across the isolated perfused human placenta. Pediatr
human lung and kidney during development and in full-term
Pharmacol 1984; 4: 145-53
placenta: a differential methylation of the gene is involved in
the regulation process. Pharmacol Toxicol 1998; 83: 183-7 160. Kennedy R, Miller R, Bell J, et al. Uptake and distribution of
bupivicaine in fetal lambs. Anesthesiology 1986; 65: 247-53
142. Nebert DW, Nelson DR, Coon MR, et al. The P450 superfami-
161. Faber JJ, Thornburg KL. Placental physiology: structure and
ly: update on new sequences, gene mapping and recommended
function of fetomaternal exchange. New York: Raven Press,
nomenclature. DNA Cell Biol 1991; 10: 1-14
1983
143. Mackenzie P, Owens I, Burchell B, et al. The UDP glycosyl- 162. Hanshaw-Thomas A, Reynolds F. Placental transfer of bupivi-
transferase gene superfamily: recommended nomenclature up- caine, pethidine and lignocaine in the rabbit: effect of umbili-
date based on evolutionary divergence. Pharmacogenetics cal flow rate and protein content. Br J Obstet Gynaecol 1985;
1997; 7: 255-69 92: 6-12
144. Collier AC, Tingle MD, Keelan JA, et al. A highly sensitive 163. Panigel M, Pascaud M, Brun JL. Une nouvelle technique de
fluorescent microplate method for the determination of UDP- perfusion de l’espace intervilleux dans le placenta humain
glucuronosyl transferase activity in tissues and placental cell isole. Pathol Biol 1967; 15: 821
lines. Drug Metab Dispos 2000; 28: 1184-6 164. Brandes JM, Tavolini N, Potter BJ, et al. A new recycling
145. Ganley N. The expression and localisation of uridine technique for the human placental cotyledon perfusion: appli-
diphosphate glucuronosyltransferase 2B subfamily in human cation to the studies of the fetomaternal transfer of glucose,
placenta tissue [M.Sc. thesis]. Auckland: University of Auck- insulin, and antipyrine. Am J Obstet Gynecol 1983; 146: 800-6
land, 2001 165. Schneider H, Panigel M, Dancis J. Transfer across the perfused
146. Aitio A. UDP glucuronosyltransferase of the human placenta. human placenta of antipyrine, sodium, and leucine. Am J
Biochem Pharmacol 1974; 23: 2203-5 Obstet Gynecol 1972; 114: 822-8
147. Schenker S, Yang Y, Mattiuz E, et al. Olanzapine transfer by 166. Schenker S, Dicke J, Johnson RF, et al. Human placental
human placenta. Clin Exp Pharmacol Physiol 1999; 26: 691-7 transport of cimetidine. J Clin Invest 1987; 80: 1428-34
148. Mannervick B, Widersten M. Human glutathione S-transfer- 167. Holcberg G, Sapir O, Huleihel M, et al. Indomethacin activity
ases: classification, tissue distribution, structure and functional in the fetal vasculature of normal and meconium exposed
properties. In: Pacific GM, Fracchia GN, editors. Advances in human placentae. Eur J Obstet Gynecol 2001; 94: 230-3
drug metabolism in man. Luxembourg: Office for Official 168. Sastry BVR, Hemontolor ME, Olenick M. Prostaglandin E2 in
Publications of the European Communities, 1995: 407-59 human placenta: its vascular effects and activation of pros-
taglandin E2 formation by nicotine and cotinine. Pharmacolo-
149. Nakasa N, Mera N, Ohmori S, et al. Nucleotide sequence of pi-
gy 1999; 58: 70-86
class glutathione S-transferase in human fetal liver. Res Com-
mun Mol Pathol Pharmacol 1997; 97: 67-78 169. Frank HG, Morrish DW, Potgens A, et al. Cell culture models
of human trophoblast: primary culture of trophoblast: a work-
150. Guthenburg C, Mannervick B. Glutathione S-transferase (trans- shop report. Placenta 2001; 22 Suppl. A: 107-9
ferase pi) from human placenta is identical or closely related to
170. Hemmings DG, Lowen B, Sherburne R, et al. Villous tropho-
glutathione S-transferase (transferase pi) from erythrocytes.
blasts cultured on semi-permeable membranes form an effec-
Biochim Biophys Acta 1981; 661: 255-60
tive barrier to the passage of high and low molecular weight
151. Finnell K, Buehler B, Kerr B, et al. Clinical and experimental particles. Placenta 2001; 22: 70-9
studies linking oxidative metabolism to phenytoin-linked tera- 171. Liu F, Michael SJ, Kenneth AL. Permeability properties of
togenesis. Neurology 1992; 4: 25-31 monolayers of the trophoblast cell line BeWo. Am J Physiol
152. Ratge D, Kohse KP, Steegmuller U, et al. Distribution of free 1997; 273: C1596-604
and conjugated catecholamines between plasma, platelets and 172. Ampasavate C, Chandorkar GA, Vander Velde DG, et al.
erythrocytes: different effects of intravenous and oral catecho- Transport and metabolism of opioid peptides across BeWo
lamine administration. J Pharmacol Exp Ther 1991; 257: 232-8 cells, an in vitro model of the placental barrier. Int J Pharm
153. Glatt H, Boeing H, Engelke CE. Human cytosolic sulphotrans- 2002; 233: 85-98
ferases: genetics, characteristics, toxicological aspects. Mutat 173. Bourget P, Fernandez H, Delouis C, et al. Pharmacokinetics of
Res 2001; 482: 27-40 tobramycin in pregnant women: safety and efficacy of a once-
154. Bernier F, Leblanc G, Labrie F, et al. The structure of human daily dose regimen. J Clin Pharm Ther 1991; 16: 167-76
estrogen and aryl sulphotransferase gene: two mRNA species 174. Schneider H. The role of the placenta in the nutrition of the
issued from a single gene. J Biol Chem 1994; 269: 28200-5 human fetus. Am J Obstet Gynecol 1991; 164: 967-73

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
512 Syme et al.

175. Boyd RDH, Haworth C, Stacey TE, et al. Permeability of the 195. Meijboom EJ, van Engelen AD, van de Beck EW, et al. Fetal
sheep placenta to unmetabolized polar nonelectrolytes. J arrhythmias. Curr Opin Cardiol 1994; 9: 97-102
Physiol 1976; 256: 617-34 196. Ito S. Transplacental treatment of fetal tachycardia: implications
176. Thornburg KL, Faber JJ. Transfer of hydrophilic molecules by of drug transporting proteins in placenta. Semin Perinatol
placenta and yolk-sac of the guinea pig. Am J Physiol 1977; 2001; 25: 196-201
33: C111-24 197. Azancot-Benisty A, Jacqz-Aigrain E, Guirgis NM, et al. Clin-
177. King BF. Development and structure of the placenta and fetal ical and pharmacologic study of fetal supraventricular tachyar-
membranes of nonhuman primates. J Exp Zool 1993; 266: rhythmias. J Pediatr 1992; 121: 608-13
528-40 198. Younis JS, Granat M. Insufficient transplacental digoxin trans-
178. Wolfgang MJ, Eisele SG, Browne MA, et al. Rhesus monkey fer in severe fetal hydrops fetalis. Am J Obstet Gynecol 1987;
placental transgene expression after lentiviral gene transfer 157: 1268-9
into preimplantation embryos. Proc Natl Acad Sci U S A 2001; 199. Weiner CP, Thompson MIB. Direct treatment of fetal supraven-
98: 10728-32 tricular tachycardia after failed transplacental therapy. Am J
179. Patterson TA, Binienda ZK, Newport GD, et al. Transplacental Obstet Gynecol 1988; 158: 570-3
pharmacokinetics and fetal distribution of 2′, 3′-didehydro-3′- 200. Arnoux P, Seyral P, Llurens M, et al. Amiodarone and digoxin
deoxythymidine (d4T) and its metabolites in late-term rhesus for refractory fetal tachycardia. Am J Cardiol 1987; 59: 166-7
macaques. Teratology 2000; 62: 93-9 201. Spinnato JA, Shaver DC, Flinn GS, et al. Fetal supraventricular
180. Tuntland T, Odinecs A, Pereira CM, et al. In vitro models to tachycardia: in utero therapy with digoxin and quinidine. Ob-
predict the in vivo mechanism, rate and extent of placental stet Gynecol 1984; 64: 730-5
transfer of dideoxynucleoside drugs against human immuno- 202. Parilla BV, Grobman WA, Holtzman RB, et al. Indomethacin
deficiency virus. Am J Obstet Gynecol 1999; 180: 198-206 tocolysis and risk of necrotizing enterocolitis. Obstet Gynecol
181. Bonati M, Bortolus R, Marchetti F, et al. Drug use in preg- 2000; 96: 120-3
nancy: an overview of epidemiological (drug use) studies. Eur 203. Abramov Y, Nadjari M, Weinstein D, et al. Indomethacin for
J Clin Pharmacol 1990; 38: 325-8 preterm labor: a randomized comparison of vaginal and rectal-
182. Sabo A, Stanulovic M, Jakovljevic V, et al. Collaborative study oral routes. Obstet Gynecol 2000; 95: 482-6
on drug use in pregnancy: the results of the follow-up 10 years 204. Nielsen GL, Sorensen HT, Larsen H, et al. Risk of adverse birth
after (Novi Sad Centre). Pharmacoepidemiol Drug Saf 2001; outcome and miscarriage in pregnant users of non-steroidal
10: 229-35 anti-inflammatory drugs: population based observational study
183. Neiburg P, Marks JS, McLaren NM, et al. The fetal tobacco and case-control study. BMJ 2001; 322: 266-70
syndrome. JAMA 1985; 253: 2998-9 205. Ericson A, Kallen BA. Nonsteroidal anti-inflammatory drugs in
184. Cnattingius S, Axelsson G, Eklund G, et al. Smoking, maternal early pregnancy. Reprod Toxicol 2001; 15: 371-5
age and fetal growth. Obstet Gynecol 1985; 66: 449-52 206. Schoenfeld A, Bar Y, Merlob P, et al. NSAIDs: maternal and
fetal considerations. Am J Reprod Immunol 1992; 28: 141-7
185. Page KR, Bush P, Abramovich DR, et al. The effects of
smoking on placental membranes. J Memb Sci 2002; 206: 207. McGarrity C, Samani N, Beck F, et al. The effect of sodium
243-52 salicylate on the rat embryo in culture: an in vitro model for the
morphological assessment of teratogenicity. J Anat 1981; 133:
186. Bush PG, Mayhew TM, Abramovich DR, et al. A quantitative
257-69
study on the effects of maternal smoking on placental mor-
phology and cadmium concentration. Placenta 2000; 21: 208. Chan LY, Chiu PY, Siu SS, et al. A study of diclofenac-induced
247-56 teratogenicity during organogenesis using a whole rat embryo
culture model. Hum Reprod 2001; 16: 2390-3
187. Shepard TH. Catalog of teratogenic agents. Baltimore: John
209. Foulon O, Jaussely C, Repetto M, et al. Postnatal evolution of
Hopkins University Press, 2001
supernumerary ribs in rats after a single administration of
188. Lehtovirta P, Fross M, Rauramo I, et al. Acute effects of sodium salicylate. J Appl Toxicol 2000; 20: 205-9
nicotine on fetal heart rate variability. Br J Obstet Gynaecol
210. Nelson JL, Ostensen M. Pregnancy and rheumatoid arthritis.
1983; 90: 710-5
Rheum Dis Clin North Am 1997; 23: 195-212
189. Manning FA, Feyerabend C. Cigarette smoking and fetal breath- 211. Akbaraly R, Leng JJ, Brachet-Liermain A, et al. Trans-placen-
ing movements. Br J Obstet Gynaecol 1976; 83: 262-70 tal transfer of four anti-inflammatory agents: a study carried
190. Pastrakuljic A, Schwartz R, Simone C, et al. Transplacental out by in vitro perfusion. J Gynecol Obstet Biol Reprod (Paris)
transfer and biotransformation studies of nicotine in the human 1981; 10: 7-11
placental cotyledon perfused in vitro. Life Sci 1998; 63: 212. Lampela ES, Nuutinen LH, Ala-Kokko TI, et al. Placental
2333-42 transfer of sulindac, sulindac sulfide, and indomethacin in a
191. Sastry BVR, Chance MB. Biotransformation of nicotine in the human placental perfusion model. Am J Obstet Gynecol 1999;
perfused human placenta [abstract]. Placenta 1994; 15: A56 180: 174-80
192. Kerenyi TD, Gleicher N, Meller J, et al. Transplacental cardi- 213. Siu SSN, Yeung JHK, Lau TK. A study on placental transfer of
oversion of intrauterine superventricular tachycardia with digi- diclofenac in first trimester of human pregnancy. Hum Reprod
talis. Lancet 1980; II: 393-4 2000; 15: 2423-5
193. Kleinman CS, Copel JA. Fetal cardiac arrhythmias: diagnosis 214. Slattery MM, Friel AM, Healy DG, et al. Uterine relaxant
and therapy. In: Creasy RK, Resnick R, editors. Maternal-fetal effects of cyclooxygenase-2 inhibitors in vitro. Obstet Gynecol
medicine: principles and practice. Philadelphia: WB Saunders, 2001; 98: 563-9
1994: 326-41 215. Sakai M, Tanebe K, Sasaki Y, et al. Evaluation of the tocolytic
194. Southall DP, Richards J, Hardwick R-A, et al. Prospective effect of a selective cyclooxygenase-2 inhibitor in a mouse
study of fetal heart rate and rhythm patterns. Arch Dis Child model of lipopolysaccharide-induced preterm delivery. Mol
1980; 55: 506-11 Hum Reprod 2001; 7: 595-602

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
Placental Drug Transfer and Metabolism 513

216. Gross G, Imamura T, Vogt SK, et al. Inhibition of cyclooxy- 236. Crowley P, Chalmers I, Keirse MJNC. The effects of cortico-
ganase-2 prevents inflammation-mediated preterm labor in the steroid administration before preterm delivery: an overview of
mouse. Am J Physiol Regul Integr Comp Physiol 2000; 278: the evidence from controlled trials. Br J Obstet Gynaecol 1990;
R1415-23 97: 11-25
217. Takahashi Y, Roman C, Chemtob S, et al. Cyclooxygenase-2 237. National Institutes of Health. Effect of corticosteroids for fetal
inhibitors constrict the fetal lamb ductus arteriosus both in maturation on perinatal outcomes. NIH Consens Statement
vitro and in vivo. Am J Physiol Regul Integr Comp Physiol 1994 Feb 28-Mar 2; 12(2): 1-24
2000; 278: R1496-505 238. Wright LL, Verter J, Younes N, et al. Antenatal corticosteroid
218. Sawdy R, Slater D, Fisk N, et al. Use of cyclo-oxygenase administration and neonatal outcome in very low birthweight
type-2-selective non-steroidal anti-inflammatory agent to pre- infants: The NICHD Neonatal Research Network. Am J Obstet
vent preterm delivery. Lancet 1997; 350: 265-6 Gynecol 1995; 173: 269-74
219. Khan KN, Stanfield KM, Dannenberg A, et al. Cyclooxygen- 239. Horbar JD. Antenatal corticosteroid treatment and neonatal
ase-2 expression in the developing human kidney. Pediatr Dev outcomes for infants 501 to 1500gm in the Vermont-Oxford
Pathol 2001; 4: 461-6 Trials Network. Am J Obstet Gynecol 1995; 173: 275-81
220. Lassus P, Wolff H, Andersson S. Cyclooxygenase-2 in human 240. Anderson DF, Stock MK, Rankin JHG. Placental transfer of
perinatal lung. Pediatr Res 2000; 47: 602-5 dexamethasone in near-term sheep. J Dev Physiol 1979; 1:
221. Maslinska D, Kaliszek A, Opertowska J, et al. Constitutive 431-6
expression of cyclooxygenase-2 (COX-2) in developing brain: 241. Varma DR. Investigation of the maternal to fetal serum concen-
a. choroid plexus in human fetuses. Folia Neuropathol 1999; tration gradient of dexamethasone in the rat. Br J Pharmacol
37: 287-91 1986; 88: 815-20
222. Newell ML. Mechanism and timing of mother-to-child trans- 242. Smith MA, Thomford PJ, Mattison DR, et al. Transport and
mission of HIV-1. AIDS 1998; 12: 831-7 metabolism of dexamethasone in the dually perfused human
placenta. Reprod Toxicol 1988; 2: 37-43
223. Mofenson LM, Lambert JS, Stiehm ER, et al. Pediatric AIDS
Clinical Trials Group Study 185 Team. Risk factors for per- 243. Levitz M, Jansen V, Dancis J. The transfer and metabolism of
inatal transmission of human immunodeficiency virus type 1 in corticosteroids in the perfused human placenta. Am J Obstet
women treated with zidovudine. N Engl J Med 1999; 341: Gynecol 1978; 132: 363-6
385-93 244. De Kloet ER. Why dexamethasone poorly penetrates in brain.
224. Connor EM, Sperling RS, Gelber R, et al. Reduction of mater- Stress 1997; 2: 13-20
nal-infant transmission of human immunodeficiency virus type 245. Maillefert JF, Duchamp O, Solary E, et al. Effects of cyclospo-
1 with zidovudine treatment. N Engl J Med 1994; 331: rin at various concentrations on dexamethasone intracellular
1173-80 uptake in multidrug resistant cells. Ann Rheum Dis 2000; 59:
146-8
225. Madelbrot L, Peyavin G, Firtion G, et al. Maternal-fetal transfer
and amniotic fluid accumulation of lamivudine in human 246. Meijer OC, de Lange EC, Breimer DD, et al. Penetration of
immunodeficiency virus-infected pregnant women. Am J Ob- dexamethasone into brain glucocorticoid targets is enhanced in
stet Gynecol 2001; 184: 153-8 mdr1A P-glycoprotein knockout mice. Endocrinology 1998;
139: 1789-93
226. Casey BM, Bawdon RE. Placental transfer of ritonavir with
zidovudine in the ex vivo placental perfusion model. Am J 247. Coutelle C, Douar AM, Colledge W, et al. The challenge of
Obstet Gynecol 1998; 179: 758-61 fetal gene therapy. Nat Med 1995; 1: 864-6
248. Sawai K, Meruelo D. Cell specific transfection of choriocarci-
227. Forestier F, de Renty P, Peytavin G, et al. Maternal-fetal
noma cells by using Sindbis virus hCG expressing chimeric
transfer of saquinavir studied in the ex vivo placental perfusion
vector. Biochem Biophys Res Commun 1998; 248: 315-23
model. Am J Obstet Gynecol 2001; 185: 178-81
249. Heikkila A, Myllynen P, Keski-Nisula L, et al. Gene transfer to
228. Marzolini C, Rudin C, Decosterd LA, et al. Transplacental
human placenta ex vivo: a novel application of dual perfusion
passage of protease inhibitors at delivery. AIDS 2002; 16:
of human placental cotyledon. Am J Obstet Gynecol 2002;
889-93
186: 1046-51
229. Minkoff H, Augenbraun M. Antiretroviral therapy for pregnant
250. Rosen TS, Pippenger CE. Disposition of methadone and its
women. Am J Obstet Gynecol 1997; 176: 478-89
relationship to severity of withdrawal in the newborn. Addict
230. Pascual MJ, Macias RIR, Garcia-Del-Pozo J, et al. Enhanced Dis 1975; 2: 169-78
efficiency of the placental barrier to cisplatin through binding 251. Fisher G, Gombas W, Eder H, et al. Buprenorphine versus
to glycocholic acid. Anticancer Res 2001; 21: 2703-8 methadone maintenance for the treatment of opioid depend-
231. Marin JJG, Bravo P, El-Mir MYA, et al. ATP-dependent bile ence. Addiction 1999; 94: 1337-47
acid transport across the microvillous membrane of human 252. Johnson RE, Jaffe JH, Fudala PJ. A controlled trial of bupre-
term trophoblast. Am J Physiol 1995; 268: G685-94 norphine treatment for opioid dependence. JAMA 1992; 267:
232. Liggins GC. Premature delivery of foetal lambs infused with 2750-5
glucocorticoids. J Endocrinol 1969; 45: 515-23 253. Ling W, Charuvastra C, Collins J. Buprenorphine maintenance
233. Spinillo A, Capuzzo E, Ometto A, et al. Value of antenatal treatment of opiate dependence: a multicenter, randomized
corticosteroid therapy in preterm birth. Early Hum Dev 1995; clinical trial. Addiction 1998; 93: 475-586
42: 37-47 254. Fisher G, Rolley JE, Eder H, et al. Treatment of opioid-
234. Ward RM. Pharmacologic enhancement of fetal lung matura- dependent pregnant women with buprenorphine. Addiction
tion. Clin Perinatol 1994; 21: 523-42 2000; 95: 239-44
235. Ballard PL, Ballard RA. Scientific basis and theapeutic regi- 255. Marquet P, Chevrel J, Lavignasse P, et al. Buprenorphine
mens for use of antenatal glucocorticoids. Am J Obstet withdrawal in a newborn. Clin Pharmacol Ther 1997; 62:
Gynecol 1995; 173: 254-62 569-71

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)
514 Syme et al.

256. Ganapathy V, Prasad PD, Ganapathy ME, et al. Drugs of abuse 263. Bailey JR, Cunny HC, Paule M, et al. Fetal disposition of delta
and placental transport. Adv Drug Deliv Rev 1999; 1: 99-110 9-tetrahydrocannabinol (THC) during late pregnancy in the
257. Ganapathy V, Ramamoorthy S, Leibach FH. Transport and rhesus monkey. Toxicol Appl Pharmacol 1987; 90: 315-21
metabolism of monoamines in the human placenta. Tropho-
blast Res 1993; 7: 35-51 264. Abrams RM, Cook CE, Davis KH, et al. Plasma delta-9-te-
258. Ganapathy V, Leibach FH. Placental biogenic amines and their trahydrocannabinol in pregnant sheep and fetus after inhala-
transporters. In: Sastry BVR, editor. Placental toxicology. tion of smoke from a marijuana cigarette. Alcohol Drug Res
Boca Raton: CRC Press, 1995: 161-74
1985/ 1986; 6: 361-9
259. Fergusson DM, Horwood LJ, Northstone K, et al. Maternal use
of cannabis and pregnancy outcome. BJOG 2002; 109: 21-7 265. Blackard C, Tennes K. Human placental transfer of cannabi-
260. Harbison RD, Mantilla-Plata B. Prenatal toxicity, maternal dis- noids [letter]. N Engl J Med 1984; 20: 797
tribution and placental transfer of tetrahydrocannabinol. J
Pharmacol Exp Ther 1972; 180: 446-53
261. Fried PA, Smith AM. A literature review of the consequences of Correspondence and offprints: Dr Jeffrey A. Keelan, Division
prenatal marihuana exposure: an emerging theme of a deficien-
cy in aspects of executive function. Neurotoxicol Teratol 2001; of Pharmacology and Clinical Pharmacology, Faculty of
23: 1-11 Medical and Health Sciences, University of Auckland, Pri-
262. Goldschmidt L, Day NL, Richardson GA. Effects of prenatal
marijuana exposure on child behaviour problems at age 10. vate Bag 92019, Auckland, New Zealand.
Neurotoxicol Teratol 2000; 22: 325-36 E-mail: j.keelan@auckland.ac.nz

© 2004 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2004; 43 (8)

Вам также может понравиться