You are on page 1of 15

BBA - Molecular Basis of Disease 1867 (2021) 166206

Contents lists available at ScienceDirect

BBA - Molecular Basis of Disease


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

HIV in pregnancy: Mother-to-child transmission, pharmacotherapy,


and toxicity
Lukas Cerveny a, Padma Murthi b, c, d, Frantisek Staud a, *
a
Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
b
Department of Medicine, School of Clinical Sciences, and Department of Pharmacology, Monash Biomedicine Discovery Institute Monash University, Clayton, Victoria,
Australia
c
Hudson Institute of Medical Research, The Ritchie Centre, Clayton, Victoria, Australia
d
Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia

A R T I C L E I N F O A B S T R A C T

Keywords: An estimated 1.3 million pregnant women were living with HIV in 2018. HIV infection is associated with adverse
HIV pregnancy outcomes and all HIV-positive pregnant women, regardless of their clinical stage, should receive a
Antiretroviral therapy combination of antiretroviral drugs to suppress maternal viral load and prevent vertical fetal infection. Although
Mother-to-child transmission
antiretroviral treatment in pregnant women has undoubtedly minimized mother-to-child transmission of HIV,
Placental membrane transporters
several uncertainties remain. For example, while pregnancy is accompanied by changes in pharmacokinetic
Placental/fetal development
Fetal programming parameters, relevant data from clinical studies are lacking. Similarly, long-term adverse effects of exposure to
antiretrovirals on fetuses have not been studied in detail. Here, we review current knowledge on HIV effects on
the placenta and developing fetus, recommended antiretroviral regimens, and pharmacokinetic considerations
with particular focus on placental transport. We also discuss recent advances in antiretroviral research and
potential effects of antiretroviral treatment on placental/fetal development and programming.

1. Introduction high-quality management of MTCT of HIV, the WHO and Pan American
Health Organization joined forces to reduce it in Cuba. The initiative
Human immunodeficiency virus-1 (HIV) infection remains a global included HIV testing of all pregnant women, ART provision for those
health challenge. It has been estimated that almost 700,000 people died who tested HIV-positive, implementation of birth by cesarean section,
of HIV-related diseases and 1.7 million were newly infected in 2019, and and replacement of breastfeeding by other alternatives. Five years after
38 million people worldwide were living with HIV at the end of the year applying these measures, Cuba was officially pronounced a country that
[1]. Moreover, in 2018 an estimated 1.3 million pregnant women with had completely eradicated MTCT of HIV [4]. However, the Cuban model
HIV were at risk of developing AIDS and perinatal transmission of the is not feasible in some other regions, such as sub-Saharan Africa, where
virus to their fetus. Mother-to-child transmission (MTCT) accounts for countries need to primarily rely on timely and efficient use of ART [5–8].
most childhood HIV infections and may occur during second and third Currently, >30 US Federal Food and Drug Administration (FDA)-
trimesters of pregnancy, labor and delivery, or breastfeeding. It has been approved antiretroviral drugs are available, singly or in various com­
proposed that the risk of HIV MTCT in the absence of any intervention is binations, to clinicians and their patients. However, most of these drugs’
up to 49%; approximately 8% of children are infected during pregnancy, pharmacokinetics, efficacy, maternal and fetal safety during pregnancy
15% during labor and delivery, and 12–26% during breast-feeding [2,3]. have not been sufficiently investigated, hence they are used off-label [9].
However, with the massive advent of antiretroviral drugs, 67% of people Although the success of ART in reducing MTCT is indisputable, the
with HIV receive antiretroviral therapy (ART) and almost 60% live research community and clinicians should not ignore possible effects of
without clinical symptoms of AIDS nowadays. Similarly, thanks to effi­ antiviral pharmacotherapy on placental development and fetal pro­
cient use of pharmacotherapy and other measures, the incidence of gramming, which might potentially result in diseases later in adulthood.
MTCT has been reduced to <1% over the last 25 years. In an example of In this review we outline current knowledge of HIV in pregnancy and

* Corresponding author at: Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho
1203, Czech Republic.
E-mail address: frantisek.staud@faf.cuni.cz (F. Staud).

https://doi.org/10.1016/j.bbadis.2021.166206
Received 5 February 2021; Received in revised form 18 May 2021; Accepted 11 June 2021
Available online 29 June 2021
0925-4439/© 2021 Elsevier B.V. All rights reserved.
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

receptors including cytokines and chemokines that may reduce HIV


replication in trophoblasts [17,18]. In utero, single or multiple major
maternal HIV variants are transmitted [19]. HIV MTCT occurs mainly
via transcytosis of HIV-infected cells [16] while free virus can pass
through the placenta via injured villous surface [15]. Feto-placental
macrophages, called Hofbauer cells, may putatively facilitate placental
HIV transmission [20] and contribute to selection of HIV variants
infecting the developing fetus [16]. HIV-1 enters cells of the immune
system, including Hofbauer cells, via interaction of the surface envelope
glycoprotein gp120 with CD4 and chemokine co-receptors CCR5 or
CXCR4, and the transmembrane envelope protein gp41 promotes fusion
of the virus with the host cell’s plasma membrane (Fig. 3).
Possible mechanisms whereby the virus infects the fetus intrapartum
include microtransfusion during contractions, ascension through the
cervix and vagina during parturition, and absorption of the virus
through the infant’s digestive tract [14,21]. In contrast to in utero
transmission of HIV, minor HIV-1 variants with relatively high gene
diversity are most likely to cause intrapartum infections [19].
Transmission through breast-feeding can occur at any point during
lactation [22]. Cell-free and infected CD4+ cells can transmigrate across
fetal oral and intestinal epithelia, probably because their mucosal layers
are relatively narrow, have comparatively low stratification, and lack
expression of anti-HIV innate proteins [23]. The risk factors related to
HIV transmission through breastfeeding have been less studied but are
apparently associated with maternal viral load and subclinical mastitis
[24,25]. The risk is cumulative; the longer the HIV-infected mother
breastfeeds, the greater the additional risk of viral transmission to the
newborn. Where breastfeeding is common and prolonged this route of
Fig. 1. Measures to prevent HIV mother-to-child transmission. The first step transmission may account for up to half of HIV infections in infants and
should be universal maternal HIV testing as early as possible during each preg­ young children [22].
nancy and prenatal counseling. If the results are positive women should imme­
diately start ART regardless of the gestation phase and clinical stage. When the 3. Placenta as an HIV sanctuary site
HIV load exceeds 1000 RNA copies/mL plasma or is unknown near the time of
delivery women should deliver by elective cesarean section and, if possible,
By definition, a viral sanctuary site is an anatomical compartment
women should avoid breastfeeding. This package of preventive interventions can
reduce the overall risk of fetal infection to <1%.
where a virus can replicate due to poor efficacy of ART and/or particular
biological properties [26]. HIV infects and replicates in trophoblasts
[15], however, with much lower efficacy when compared with CD4+
the use of ART in it. In detail, we focus on mechanisms of HIV passage
cells [27,28]. This is likely because none or minimal amounts of CD4,
through the placenta, changes in morphology and histology of the
CCR5 and CXCR4 are expressed in the membranes of trophoblasts
placenta in HIV-positive women, and their pregnancy outcomes. Then
[28–30]. HIV thus enters the trophoblast cells by less effective CD4-
we summarize current knowledge on use of ART to prevent MTCT of the
independent pathways, via receptor-mediated endocytosis [27,28,30]
virus, considering pharmacokinetic parameters in pregnancy and
and/or direct cell-cell contact [28,31]. Moreover, infection of tropho­
mechanisms affecting placental transfer of antiretrovirals. We also
blasts can be reduced by soluble factors, including placental chemokine
provide an overview of possible acute and chronic toxic effects of anti­
(C-C motif) ligand 5, macrophage inflammatory protein-1β or leukemia
retrovirals on both fetal development and programming.
inhibitory factor, creating suppressive environment at the feto-placental
interface [32]. On the other hand, viral production in trophoblast cells
2. Mechanisms and risks of mother-to-child transmission of HIV
may be supported by exposure to tumor necrosis factor-α, interleukin-1,
and/or interleukin-8 produced by the placenta or released upon viral/
Several possible mechanisms of HIV transmission from mother to her
bacterial/protozoal infection, which was suggested to accelerate HIV
offspring have been described, including in utero transplacental HIV
MTCT transmission [30,32]. Recently, syncytin, the envelope glyco­
transport, intrapartum transmission, and infection through breastfeed­
protein of human endogenous retrovirus family W1 expressed in
ing. High maternal viral load is the strongest risk factor for both in utero
placental trophoblasts, has been shown to contribute to viral replication
and intrapartum MTCT. Risks for vertical HIV transmission are also
in the placenta [31].
influenced by the viral phenotype and increased by host factors such as
HIV maintains a low level of infection in placentas despite ART and
advanced HIV-1-related illness/AIDS, maternal co-infections, prolonged
can be reactivated after its discontinuation [31]. Placental sanctuary
rupture of amniotic membranes, and polymorphism of genes affecting
sites likely develop through activities of placental membrane ABC efflux
virus entry or immune responses [10,11]. Moreover, there are some
transporters (see below) [33]. Moreover, HIV in trophoblasts (and
indications that overall risks for in utero HIV transmission may be higher
associated cell-to-cell infection) is less sensitive to ART than free-living
if primary HIV infection occurs during pregnancy rather than before
virus [31]. Nevertheless, the exact effects of HIV in sanctuary site on
conception [12,13].
total rate of MTCT remain to be elucidated.
The placenta provides a barrier that effectively reduces, but does not
completely prevent, HIV transmission and the most frequent window for
4. Pathological effects of HIV on placental structure
in utero transmission is the last 14 days before delivery [14]. Mecha­
nisms of HIV passage across the placenta are not well known [15]. The
Pioneering studies of third-trimester placentas of HIV-infected
virus infects syncytiotrophoblast, cytotrophoblast, and villous endo­
pregnant women who received no antiretroviral treatment did not
thelial cells [16], but placental cells produce various soluble factors and
identify any significant morphological abnormalities [34] and found

2
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

them to be both heavier [34] and lighter [35,36] than those of controls. hepatitis B viruses, Neisseria gonorrhea, syphilis, and others) have been
Histologically, maternal HIV infection is reportedly associated with reported [7]. Due to decreased immune function, these neonates are also
higher incidences of chorioamnionitis [34,35,37], villous hyper­ at risk of lower respiratory tract infections of varied aetiology, gastro­
cellularity [34], funisitis, placental membrane inflammation [38], enteritis [45], and late-onset neonatal group B Streptococcus infection,
villous immaturity, allantois vasculopathy [39], and deciduitis [35]. It the leading cause of neonatal sepsis [45,50]. It is also hypothesized that
has also been associated with inflammation of placental villi, which they reveal short-lived memory response to vaccination, increased risk
frequently accompanies congenital viral infections [40], but more of autoimmune diseases and increased susceptibility to HIV infection
controversially [34,35,37,41]. during adulthood [45,46].
No significant differences in histological findings including cho­ In the postnatal period there are higher frequencies of undernutri­
rioamnionitis, villitis, villous stromal fibrosis, infarction, abnormal tion, infection, and psychological consequences following neonatal
villous maturation, and deciduitis in first- and second-trimester pla­ exposure [51]. However, these effects also seem to be linked to maternal
centas have been detected between groups of seropositive and sero­ social vulnerability, poverty, and substance abuse [7].
negative women [34,42]. Studies on HIV in Africa have concluded that
chorioamnionitis is a risk factor for perinatal and early in utero trans­ 6. Prevention of HIV mother-to-child transmission
mission of HIV-1 [42]. Another trial, however, suggested the opposite
correlation [35]. It has been estimated that implementation of prevention of MTCT
Maternal HIV infection is also associated with higher risk for (PMTCT) services prevented HIV infection of around 1.4 million chil­
placental infection with human papilloma virus, which provokes sig­ dren between 2010 and 2018 [52], so PMTCT of HIV is one of the great
nificant structural changes of the villi, potentially hampering normal public health successes of the past two decades. This PMTCT has four
interchange of gases and nutrients at the maternal-fetal interface [43]. main elements. First, universal maternal testing and prenatal coun­
seling. Second, use of ART during pregnancy, labor and delivery, fol­
5. Effects of in utero HIV exposure on fetal development lowed by postnatal administration to the infant. Third, elective cesarean
delivery before amniotic membrane rupture (after 38 weeks of gesta­
Observational studies have indicated links between HIV infections tion) in cases where the HIV load exceeds 1000 RNA copies/mL plasma
and adverse pregnancy outcomes, such as increased frequencies of or is unknown. Fourth, avoidance of breastfeeding. This package of
spontaneous abortion, stillbirth, perinatal and infant mortality, intra­ preventive interventions can reduce the risk for fetal infection to <1%
uterine growth restriction, and low birth weight [7,41,44]. This may (Fig. 1) [8,53,54]. Introduction of these measures in Cuba resulted in
result from HIV-triggered shift to significantly activated, proin­ complete eradiation of HIV MTCT by 2015, five years after initiation [4].
flammatory intrauterine immune environment and thus disrupted In the USA, only 44 infants were born with HIV infection in 2016 and the
immunological tolerance normally present during pregnancy [45,46]. estimated incidence of perinatally acquired HIV infection was 1.1 out of
The extent of maternal immune deficiency is positively correlated with 100,000 live births [8]. In less advantaged settings, however, there is an
risks for prematurity [7,47]. Subsequently, preterm birth might result in ongoing epidemic of pediatric HIV infection; estimates indicate that
early infant morbidity, mortality, and/or chronic diseases later in life there were approximately 150,000 new pediatric infections globally in
[48]. Disease progression is also more rapid in infants infected perina­ 2019 [55], mostly acquired through MTCT, and mostly in sub-Saharan
tally than in those infected later by breastfeeding, with the accelerated Africa. This is due to the fact that integration of programs aiming at
pathogenesis being attributed to differences in immunological maturity HIV PMTCT in this area are hampered by multiple factors including
at the time of infection, and/or detrimental effects of HIV infection upon bureaucracy, HIV stigma, shortage of training for medical staff, staff
infant growth and development [49]. turnover (frequently due to high workloads), insufficient infrastructure
Neonates exposed to HIV perinatally, but uninfected are at increased and HIV testing, and site-to-site differences in HIV routine care [56,57].
risk of prematurity, fetal hypotrophy, and mortality [7,44,45]. Higher
rates of MTCT of sexually transmitted infections (hepatitis C and

Fig. 2. Schematic illustration of membrane


transporters involved in placental pharmaco­
kinetics of antiretrovirals. In the apical
(mother-facing) microvillous membrane
important protective efflux transporters, P-
glycoprotein (ABCB1) and breast cancer
resistance protein (ABCG2), are localized as
well as influx organic cation/carnitine trans­
porters (OCTNs) and equilibrative nucleoside
transporter 1 (ENT1) mediating facilitated
diffusion of nucleosides and nucleoside-
derived drugs [104,111,112,121,123,141].
Influx organic cation transporter 3 (OCT3) and
organic anion transporter 4 (OAT4) mediating
feto-maternal transport of their substrates
have been detected in the basal membrane
[132]. Equilibrative nucleoside transporter 2
(ENT2) is putatively expressed in both
trophoblast poles [121,122]. Data on func­
tional expression and localization of CNTs
remain controversial [104,105,123,126–128].

3
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

Fig. 3. Scheme of HIV replication and mechanisms of action of FDA approved and investigational antiretroviral drugs. HIV contains several important molecules.
Glycoprotein 120 (gp120) is an envelope surface molecule, which has interactive effects with CD4 molecules and CCR5 and/or CXC4 co-receptors on CD4-expressing
cells (predominantly helper T-cells). It results in conformational changes of gp120 and transmembrane glycoprotein 41 (gp41) that trigger fusion of HIV with the host
cell membrane. Reverse transcriptase controls cellular synthesis of viral complementary DNA that is subsequently integrated as double-stranded DNA by the enzyme
integrase to the host cells’ genome. Newly synthesized polyproteins are cleaved by proteases, which is important for virion maturation. Cobicistat and ritonavir listed
in the figure are used only as pharmacokinetic boosters to increase the bioavailability and prolong plasma half-lives of PIs.
*, preferred first-line therapy; **, when started prior to conception the drug should be continued during pregnancy; ***, no data on pharmacokinetics and safety in
pregnancy available; #, alternative to the first-line therapy; ##, not recommended due to substantial decrease in plasma concentrations of antiretroviral drug in the
second and third trimesters; ###, obsolete due to unfavorable pharmacokinetics and/or potentially negative impact on maternal or fetal health; §, investiga­
tional drugs.

7. Antiretroviral therapy in pregnancy (NRTI) zidovudine could reduce the risk for vertical transmission by
67% [58]. Although zidovudine is no longer recommended for first line
In 1994, the first controlled study of antiretroviral prophylaxis in ART in adults, it is still considered an alternative first-line medication for
pregnancy revealed that the nucleoside reverse transcriptase inhibitor pregnant and breastfeeding women with HIV. ART decreases HIV viral

4
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

loads in maternal blood and genital secretions and antiretrovirals with reductions in their exposure [59]. Currently, effects of combinations of
high transplacental passage provide pre-exposure prophylaxis for in­ the pharmacokinetic booster cobicistat (an inhibitor of CYP3A and
fants, thereby protecting maternal health and powerfully reducing both ABCB1 [86]), with atazanavir, darunavir, or the integrase strand
in utero and perinatal MTCT of HIV [1]. Global efforts have resulted in up transfer inhibitor (INSTI) elvitegravir, in pregnancy are being investi­
to 85% of HIV-positive pregnant and breastfeeding women receiving gated. However, plasma concentrations have been substantially
ART [1]. Currently, >30 antiretroviral drugs are approved by the FDA, (50–90%) lower than those of postpartum counterparts. This reduced
nine of which are used in first-line regimens for ART-naïve pregnant exposure does not guarantee anti-HIV efficacy of these regimens in
women with HIV (see Fig. 3). In the following text we describe phar­ pregnancy [74,87,88] and these combinations should be avoided [8].
macokinetic aspects of ART in pregnant women, including placental Interestingly, doses of the non-nucleoside reverse transcription in­
transport of antiretrovirals. hibitor (NNRTI) efavirenz should be reduced in pregnancy, despite ob­
servations of lower exposure with normal adult dosing. This is because
7.1. Pharmacokinetic considerations and antiretroviral therapy free circulating active efavirenz concentrations are only moderately
lower than those in non-pregnant women and remain sufficiently
Antiretrovirals are used during pregnancy and lactation in off-label effective against HIV, while side effects are less frequent [89,90]. On the
regimens due to exclusion of pregnant and lactating women from clin­ other hand, there is also evidence for increased exposure in the third
ical trials. Their use in pregnancy is dependent on toxicity, pharmaco­ trimester than postpartum, as also reported for the NNRTI etravirine,
kinetics, and fetal/neonatal exposure data collected during phase IV which can be associated with increased risks for toxicity [66].
clinical tests and/or opportunistic studies on women who become Exposure to the NRTIs emtricitabine, abacavir, zidovudine, and
pregnant while receiving ART [9]. This is unfortunate as pregnancy is a lamivudine is not changed during the third trimester, so no dosing ad­
dynamic state in which physiological (and hence pharmacokinetic pa­ justments in pregnancy are required [91–93]. Physiological changes
rameters) constantly change [59]. Thus, changes in pregnancy, espe­ during pregnancy may amplify effects of drug-drug interactions,
cially in the second and third trimesters, may result in altered exposure resulting in more profound reductions in absolute drug exposure and
[60–67]. For example, absorption of drugs from the gastrointestinal thus hampering treatment efficacy [94]. Optimization of antiretroviral
tract and oral bioavailability vary due to changes in gastric secretion and dosing based on therapeutic monitoring [61,95–97] and/or physiology-
motility of the small intestine. Similarly, changes in other physiological based pharmacokinetic modeling [98–100] may be helpful for assurance
variables, such as increases in cardiac output or blood volume, will of the safety and effectiveness of maternal and fetal therapy.
affect drug disposition. In addition, changes in biotransformation ac­ Since information on dosage adjustment in pregnancy is constantly
tivities and rates of liver and kidney blood perfusion, as well as evolving, the reader is referred to the FDA’s “HIV Treatment Information
glomerular filtration, will modify drugs’’ metabolism, excretion and for Pregnant Patients” for the most recent information [101].
elimination half-lives. Notably, increases in activities of CYP3A4 and
CYP2D6, the most important cytochrome P450 isoforms, have been re­ 7.2. Passage of antiretrovirals across the placenta; role of membrane
ported in pregnant women, including up to 35 and 48% increases in the transporters
third trimester [68,69].
Possible effects of pregnancy on pharmacokinetics of drugs Antiretrovirals are primarily used to suppress maternal viral loads,
frequently and chronically prescribed during gestation have been thor­ thereby diminishing the progression of HIV infection to AIDS, and
oughly and systematically reviewed by Pariente et al. [65]. They illus­ decreasing risk of horizontal as well as vertical HIV transmission. At
trated that pregnancy affects the pharmacokinetics of most least one of the compounds must cross the placenta to provide pre-
antiretrovirals, thereby significantly raising their elimination/clearance exposure prophylaxis for the fetus [8]. On the other hand, deleterious
and reducing their exposure parameters (Cmax, AUC). Therefore, effects of antiretrovirals on the fetus and/or placenta have been
pregnant women administered ART may require adjusted dosing described (see below). Therefore, transport of drugs across the placenta
schedules to obtain comparable antiretroviral exposure to that of non- and their subsequent effects on the fetus are of great concern [9]. A
pregnant women. However, routine dose adjustment of combination pragmatic balance needs to be established between fetal protection
ART during gestation is not recommended and should be evidence-based against HIV and drug toxicity. Therefore, transplacental kinetics of
using appropriate pharmacokinetic models [9,59,70–73]. antiretrovirals and all influential factors need to be understood for
For preferred protease inhibitors (PIs: atazanavir, darunavir or balanced selection of effective but safe ART in pregnancy [9,59].
lopinavir) all in combination with low-dose ritonavir, a pharmacoki­ The placenta is a barrier that separates maternal and fetal circula­
netic booster that inhibits CYP3A and P-glycoprotein (ABCB1), signifi­ tions. For a drug to travel from maternal to fetal blood it must cross the
cant decreases in exposures in pregnancy have been reported [74–77]. syncytiotrophoblast layer, interstitium, and endothelial cells in fetal
The darunavir dose has to be doubled to maintain sufficient exposure capillaries. Transport of drugs across the placenta occurs mainly via
[75,78]. Increased dosing of lopinavir is not recommend for all pregnant passive diffusion along the concentration gradient [102]. However, the
women [62,79], but may be considered in a personalized manner, e.g. placenta is equipped with a battery of efflux and influx transporters that
for PI-experienced pregnant women [80]. Similarly, atazanavir plasma to some extent control mother-to-fetus transport of drugs [102–106].
concentrations equivalent to those postpartum can be achieved by Empirical evaluation of placental transport of drugs in clinical settings is
increased dosing [81], but some experts recommend an increased dose limited to measurement of umbilical cord and maternal plasma drug
of atazanavir only for treatment-experienced pregnant women who are concentrations at delivery, and ratios of these concentrations provide
also receiving tenofovir disoproxil fumarate [8]. Because these PIs are little information about the time-dependent pharmacokinetics involved.
highly bound to plasma proteins [61,62], decreased AUC in the second Therefore, current research relies on alternative experimental ap­
and third trimester are likely due to overall decrease in total plasma proaches, such as use of in vitro cell-based (BeWo, JEG-3, JAR) models,
protein concentrations and consequent acceleration of excretion placental fragments and explants or dually perfused human or rodent
[70,71,82]. However, the effectiveness of HIV suppression with normal placenta. All these models have pros and cons [9,107], and thus should
adult dosing of these PIs is usually preserved as concentrations of un­ be used in combination to obtain as comprehensive understanding of the
bound fraction are unaffected [61,62]. Thus, other mechanisms are also real in utero phenomena as possible. Collected data can then help
presumably involved. Darunavir, lopinavir, and atazanavir are almost development of robust pregnancy physiology-based pharmacokinetic
exclusively metabolized by CYP3A4 [83,84] and darunavir and ataza­ models that can be potentially used to predict prenatal exposure to
navir are substrates of ABCB1 [83,85] so increases in CYP3A4 and maternally administered drugs [108–110].
ABCB1 activities towards term of pregnancy may contribute to We and other teams have used various experimental approaches to

5
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

investigate the role of drug transporters in placental kinetics of anti­ derived drugs [124] contribute to placental transfer of abacavir,
retrovirals. Placenta strongly expresses ATP-binding cassette (ABC) drug emtricitabine, or zidovudine [104,125]. We found that placental pas­
efflux transporters, such as ABCB1 and breast cancer resistance protein sage of abacavir, but not zidovudine and emtricitabine, is facilitated by
(ABCG2) [111,112]. These proteins are localized in the apical mem­ ENT1 [104]. However, interaction between emtricitabine and placental
brane of syncytiotrophoblast (Fig. 2), where they actively transport their ENT1 has been recently reported [126]. Concentrative nucleoside
substrates from the placenta to the maternal circulation, thus providing transporters (CNTs) may also putatively contribute to placental transfer
fetal protection against potentially hazardous compounds. They recog­ of antiretrovirals [126]. However, conflicting data have been published
nize hundreds of structurally divergent substrates [113] including on CNTs’ gene expression, protein levels, and localization. We detected
clinically used antiretrovirals (Tables 1 and 2) [85,114–120]. There is significant amounts of CNT2 and CNT3 transcripts in first and term
also growing evidence that solute carrier (SLC) transporters contribute placentas and showed that CNT2 in the placenta is likely regulated by
to placental transfer of antiretrovirals. NRTIs, the foundations of com­ protein kinase A [105]. Govindarajan et al. only detected CNT2 mRNA
bination ART, are nucleoside-derived drugs. Therefore, we addressed [123], and no significant CNT activity was observed in microvillous
the possibility that equilibrative nucleoside transporters (ENTs) that are membrane vesicles prepared from term placentas [104,121], but
localized on both poles of the syncytiotrophoblast [104,121–123] (see another group detected expression of genes encoding all CNT variants
Fig. 2) and known to facilitate diffusion of nucleosides and nucleoside- (CNT1, CNT2, and CNT3) in term placentas, and showed that CNT3 can

Table 1
Preferred first-line antiretroviral drugs and alternative drugs to first-line ART used in ART-naive pregnant women with HIV.
Pharmacodynamic Drug Abbrev. Physical-chemical Plasma Placental Range of C/M Ref.
group properties protein transporter concentrations ratios
Lipid solubility; pKa binding (substrate/ at delivery
inhibitor)

Nucleoside reverse Abacavir ABC logP = 1.20; apKa 50% ABCB1, ABCG2, 1.03–1.06 [92,104,116,143,205,206]
transcriptase (most acidic) = 15.41; ENT1, OCT3
a
inhibitors pKa (most basic) = 5.8
(NRTI) Emtricitabine FTC logP = − 0.43; pKa = <4% ENT1, CNT1, 0.95–1.82 [91,126,145,206–209]
2.65 MATE1, OCT3,
OCTN1
Lamivudine 3TC logP = − 1.4; apKa <36% MATE1, OCT3 0.93–1.22 [92,117,189,205,206,210,211]
(most acidic) = 14.29;
a
pKa (most basic) =
− 0.16
b
Tenofovir TDF logP = 1.25; pKa = 3.75 <0.7%c ABCB1, ABCG2, 0.62–6.0 [145,206,207,209,211]
disoproxil OCT3
fumarate
Zidovudine AZT logP = 0.05; apKa 30–38% ABCB1, ABCG2, 0.81–1.6 [115,212–214]
(most acidic) = 9.96; OAT4, CNT1
a
pKa (most basic) = − 3
Non-nucleoside reverse Efavirenz EFV logP = 4.6; apKa (most >99% ABCB1, ABCG2, 0.49–0.67 [212,215–218]
transcriptase acidic) = 12.52; apKa MATE1
inhibitors (most basic) = − 1.5
(NNRTI) Rilpivirine RPV logP = 4.86; pKa = 5.6 >99% ABCB1, ABCG2, 0.50–0.74 [129,150,151,219,220]
OCTN
Protease inhibitors Atazanavir ATV logP = 4.5; apKa 86% ABCB1, ABCG2, 0.12–0.21 [77,81,85,221–224]
(PIs) (strongest acidic) = OCTN
11.92; apKa (strongest
basic) = 4.42
Darunavir DRV logP = 1.89; apKa 95% ABCB1 0.11–0.24 [119,225,226]
(strongest acidic) =
13.59; apKa (strongest
basic) = 2.39
a
Lopinavir LPV logP = 3.91–4.69; >98% ABCB1, ABCG2, 0–0.57 [129,211,212,221]
a
pKa (strongest acidic) OCTN
= 13.39; apKa
(strongest basic) =
− 1.5
d
Ritonavir RTV logP = 3.9; apKa 98–99% ABCB1, ABCG2, 0–0.55 [129,212,221]
(strongest acidic) = OCTN
13.68; apKa (strongest
basic) = 2.84
Integrase strand transfer Dolutegravir DLV logP = 2.2; pKa = 8.2 98.9% ABCB1, ABCG2, 1.06–1.25 [129,190,209,227,228]
inhibitors OCTN
a a
(INSTIs) Raltegravir RLV logP = − 0.39; pKa 83% ABCB1, eABCG2, 1.21–1.5 [120,229–234]
e
(strongest acidic) = MATE1
5.62; apKa (strongest
basic) = − 1.5
a
Predicted value, experimental value not known.
b
Tenofovir disoproxil fumarate is rapidly hydrolyzed to tenofovir after oral administration, therefore the plasma protein binding of tenofovir is provided; tenofovir
disoproxil fumarate (but not tenofovir) is a substrate of placental ABCB1 and ABCG2 [114].
c
In vitro value.
d
Ritonavir is used as a CYP3A4 and ABCB1 inhibitor to increase bioavailability and plasma half-lives of co-administered PIs [95].
e
Weak interaction with low propensity to cause drug-drug interactions.
C/M ratio, cord to maternal plasma concentrations ratio; logP, pKA, and plasma protein values were taken from DrugBank Online|Detailed Drug and Drug Target
Information, www.drugbank.com

6
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

Table 2
Antiretroviral drugs that can be continued during pregnancy when they are started before conception.
Pharmacodynamic group Drug Abbrev. Physical-chemical Plasma Placental Range of C/M Ref.
properties protein transporter concentrations ratios
Lipid solubility; pKa binding (substrate/ at delivery
inhibitor)
c e
Nucleoside reverse Tenofovir TAF logP = 1.6, pKa = 3.96 80% ABCB1, ABCG2 0.97 [154,235]
transcriptase inhibitors alafenamide
(NRTIs) fumarate
a
Non-nucleoside reverse Etravirine ETV logP = 3.67, alogP = d
99.9% ABCB1, ABCG2 0.32–0.52 [66,212,236–240]
transcriptase inhibitors 5.54
a
(NNRTIs) pKa (strongest acidic) =
12.49; apKa (strongest
basic) = 4.13
Nevirapine NVP logP = 2.5; apKa 60% ABCB1, ABCG2 0.59–1.0 [212,215,216]
(strongest acidic) =
10.37; apKa (strongest
basic) = 5.06
a
Entry and fusion Maraviroc MVC logP = 4.3, alogP = 3.63 76% ABCB1, OAT4, 0.33–0.37 [129,155,212,241,242]
a
inhibitors pKa (strongest acidic) = OCTN
13.98; apKa (strongest
basic) = 9.65
b
Enfuvirtide T-20 – 92% – Undetectable in cord [212]
plasma
a
Predicted value.
b
Biomimetic peptide.
c
Ex vivo value.
d
In vitro value.
e
Analysis was performed in cohort (n = 1) treated with tenofovir alafenamide fumarate in combination with cobicistat.
logP, pKA, and plasma protein values were taken from DrugBank Online|Detailed Drug and Drug Target Information, www.drugbank.com.

participate in nucleoside-derived drug uptake [127]. Moreover, Errasti- Based on recent safety and efficacy evidence, the current US Peri­
Murugarren et al. detected significant levels of CNT1 protein (but no natal Guidelines [8] recommend as first-line therapy NRTIs, abacavir
other CNT proteins), despite negligible transcript-level expression, plus lamivudine or tenofovir disoproxil fumarate plus emtricitabine or
localized in both syncytiotrophoblast poles [128]. Clearly, more lamivudine [143–146], in combination with either a PI (ritonavir
research is needed to fully elucidate functional expression of nucleoside boosted atazanavir [76] or ritonavir boosted darunavir [147]) or INSTI
transporters in the placenta and their role in materno-fetal transport of (dolutegravir [148] or raltegravir [149]). The NNRTIs efavirenz or ril­
antiretrovirals. pivirine can serve as alternatives to the first-line PIs or INSTIs
Organic cation/carnitine transporters (OCTNs) and multidrug and [150,151]. On the other hand, European guidelines recommend initia­
toxin compound extrusion transporter 1 (MATE1) are localized in the tion of ART containing dolutegravir after 8 weeks of pregnancy due to
apical membrane of the syncytiotrophoblast (Fig. 2). OCTN transporters potential of higher risks for neural tube defects if used periconception
primarily govern uptake of L-carnitine from maternal circulation [129], [152,153]. Women who are already on suppressive ART at the time of
but may also putatively transport emtricitabine into syncytiotrophoblast conception should continue without changes, unless drugs with un­
[126]. On the other hand, MATE1 contributes to fetal protection against proven efficacy and reportedly inferior safety in pregnancy are being
xenobiotics and affects placental kinetics of lamivudine [117,130,131]. used (if so, they should be substituted by drugs with more favorable risk-
Organic anion transporter 4 (OAT4) [132] and organic cation trans­ benefit ratios [8]).
porter (OCT3) [103,130] have been found in the syncytiotrophoblast’s All women with HIV who have not received antepartum ART should
basal membrane (Fig. 2) where they mediate uptake of negatively and start therapy immediately to prevent perinatal transmission of HIV.
positively charged compounds, respectively, from the fetal circulation Although intrapartum/neonatal antiretroviral medications will not
[103,130,131,133]. An overview of antiretrovirals used in pregnancy prevent perinatal transmission that occurs before labor, most trans­
with their basic physico-chemical characteristics, protein binding pa­ mission occurs near to or during labor and delivery [8].
rameters, and interactions with membrane transporters localized in the NNRTIs (nevirapine and etravirine), the NRTI tenofovir alafenamide
placenta is provided in Tables 1 and 2. fumarate [154], and entry and fusion inhibitors (maraviroc and enfu­
The expression and transport activity of transport proteins are not virtide) should be continued if started before conception [8,155].
uniform, frequently differ among individuals, and vary during gestation Cobicistat-boosted regimens (with atazanavir, darunavir, or the INSTI
[104,105,107]. In addition, the expression of genes encoding placental elvitegravir) [9], or dual combination of dolutegravir and lamivudine
transporters can be modulated (up or down) by drug exposure, patho­ should be avoided due to risk for insufficient viral suppression [67]. For
logical conditions, genetic variants, and/or drug-drug interactions recently approved drugs such as the INSTI bictegravir and NNRTI dor­
[134–140]. Therefore, rates of antiretrovirals’ placental transfer may avirine, or the CD4 antagonist ibalizumab, there are no data on their
differ among individuals and stages of pregnancy. efficiency and safety in pregnancy, and thus should also be avoided
[8,67,142]. An overview of currently approved and investigational
7.3. Current guidelines for pharmacotherapy of pregnant women antiretrovirals and their action mechanisms is provided in Fig. 3.

Official guidelines for use of antiretroviral drugs in pregnancy 8. Toxicity of antiretroviral therapy in the fetoplacental unit
distinguish between pharmacotherapy of ART-naïve pregnant women
and pregnant women already on ART. Currently, ART should be initi­ Since antiretrovirals are used chronically, often throughout gesta­
ated for antiretroviral-naïve pregnant women with HIV as early as tion, they can potentially affect fetal development and/or programming.
possible regardless of their clinical stage and continued throughout In addition, placental maturation and functions may be disturbed by
gestation [8,142]. chronic exposure to antiretrovirals. The most frequently reported (and

7
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

comprehensively reviewed [8]) adverse pregnancy outcomes are low in utero exposure to antiretrovirals.
birth weight, preterm birth (with relative risks/odds ratios ranging from
1.2 to 3.4), still birth, and fetal growth restriction. For some anti­ 8.3. Placental toxicity
retrovirals there is also evidence of increased risks for congenital mal­
formations [153,156–159]; other adverse outcomes cannot be Since the placenta plays a fundamental role in fetal programming
recognized immediately after delivery and may remain unnoticed until [177], any detrimental effect of antiretrovirals on placental structure or
later stages of life such as poor neurodevelopment [160–162]. However, function seems likely to result in sub-optimal in utero conditions. Such
there is a paucity of data and it is difficult if not impossible to draw clear possible effects have been largely neglected and only a handful of papers
conclusions from current information. Mechanisms of antiretroviral describe the potential toxicity of antiretrovirals on placental develop­
toxicity in pregnancy are also poorly understood and require systematic ment and function. However, recent studies on placentas collected from
investigation. women on ART have suggested that effects may include restricted
placental growth [41,178–180], increases in numbers of marginal in­
8.1. Acute toxicity; teratogenicity farcts [41,180], maternal vascular malperfusion and thrombosis
[179,180], membrane infection, hypervascularity, and reductions in
The critical window for teratogenic effects of drugs is the first both the surface and perimeter of the terminal villi [180]. These
trimester of pregnancy. Most published research on ART during this anatomical and histological changes may be associated with reported
period indicates a generally low association with teratogenicity increases in risk for preterm delivery [180]. Altered placental and fetus
[159,163], but some specific antiretroviral agents seem to increase risks growth resulting in increased placenta-to-birthweight ratio, a strong
[153]. The most frequently investigated drugs are efavirenz, zidovudine indicator of various diseases occurring later in life, were also observed in
and dolutegravir [153,156–159]. Until recently, dolutegravir was not animal models [181,182].
recommended for use at conception and in very early pregnancy due to Kala et al. (2020) investigated possible effects of PIs-based combi­
suspected neural tube defects [153]. However, this recommendation nation ART initiated at periconception on decidualization and spiral
was amended by US Perinatal Guidelines in August 2019 and dolute­ artery remodeling in early pregnancy [183]. Using a set of mouse and
gravir is now a “Preferred antiretroviral drug throughout pregnancy and human placenta-based experiments, they concluded that lopinavir but
an alternative antiretroviral drug for women who are trying to conceive” not darunavir-based treatment impaired uterine decidualization and
[8]. Data from animal experiments [164] and retrospective studies spiral artery remodeling in both human ex vivo and mouse in vivo
[153] suggest efavirenz may be a teratogen causing abnormalities in experimental models. They attributed lopinavir-associated deciduali­
neural tube and CNS development [165]. However, according to the zation defects to a decrease in expression of transcription factor STAT3,
WHO, efavirenz-associated risks are outweighed by its benefits [159] which is known to regulate decidualization.
and current guidelines suggest efavirenz in triple therapy with preferred Efavirenz interacts with several serotonin receptors and blocks se­
NRTIs for pregnant women [8,142]. Clearly, clinicians must follow rotonin transporter (SERT/SLC6A4). Since both fetal brain and the
frequent updates and implement them in their routines. placenta express these receptors and transporters, efavirenz exposure
during pregnancy might disturb serotonin homeostasis in the fetopla­
8.2. Long-term toxicity; fetal programming cental unit. Importantly, serotonin is a crucial neurotrophic factor for
fetal neurodevelopment and pharmacological insult can affect fetal
Although risks for gross teratogenicity seem to be minimal, research development/programming – similar to observed effects of prenatal use
on long-term risks is needed to identify possible effects of perinatal of selective serotonin reuptake inhibitors [184,185]. Possible interfer­
antiretroviral exposure on fetal programming and diseases occurring in ence of zidovudine with the development of the GABAergic system has
adulthood. In the latest literature, the most frequently discussed prob­ been recently reported in mice [186]. Nevertheless, more research is
lems seem to be mitochondrial and neurodevelopmental toxicity. required to fully understand the effects of prenatal antiretroviral expo­
Various studies have indicated changes in mitochondrial structure and sure on neurodevelopment.
function in infants exposed to ART, especially NRTIs [166]. These
mitochondrial alterations may putatively lead to various clinical mani­ 9. Recent advances in antiretroviral research; long-acting
festations, such as seizures, cognitive and motor delay, abnormal neu­ formulations, drugs in the pipeline
roimaging, hyperlactatemia, and cardiac dysfunction [167,168].
Possible effects of antiretrovirals on neurodevelopmental outcomes have ART is constantly being developed and refined with an ultimate goal
been investigated in large-scale epidemiological studies. The heteroge­ to find effective regimens that can be prescribed in personalized fashion
neity of study populations and study designs may complicate interpre­ with high efficacy and minimal acute and long-term side effects. Since
tation of cumulative data, but both acute effects of ART on 2015 several new drugs and regimens have been approved and imple­
neurodevelopment and developmental delay have been discussed in mented in therapy of adults with HIV, but due to lack of safety, outcome,
recent literature [160,162,169]. Long-term behavioral effects of pre­ and pharmacokinetic data they are not recommended for use in preg­
natal exposure to ART have also been detected and studied in detail in nancy. For instance, tenofovir alafenamide fumarate has been shown to
animal models, including alteration in social interactions, cognitive have more favorable pharmacokinetics (associated with lower renal and
functions, sensorimotor processing, and exploratory behaviors at later bone toxicity) than tenofovir disoproxil fumarate [187,188]. Recently
stages in life [164,170]. published data on tenofovir alafenamide fumarate show that exposure to
Other types of long-term effects have also been reported. For this drug is lower in the third trimester than postpartum, whereas no
instance, a Danish study found that HIV-exposed but uninfected children differences in exposure were observed between pregnancy and post­
aged 0–5 years exposed to antiretrovirals were smaller at birth than partum in women taking tenofovir alafenamide fumarate with cobicistat
unexposed controls, but this difference decreased with time [171]. [154]. Tenofovir alafenamide fumarate is not recommended for use in
Obese HIV-exposed but uninfected youths seem to have an increased risk ART-naïve pregnant women with HIV, but current guidelines already
of hypertension [172]. Prenatal ART exposure is also suggested to have suggest continuation of its use for women taking this medication prior to
mild direct cardiac effects [173], and may reportedly be associated with conception, despite a lack of clinical data from sufficiently large
higher frequencies of tumor development [174,175]. However, numbers of women [8]. In 2019 a very promising first ever fixed-dose
increased odds of cancer in children exposed in utero to ART have not dual combination of dolutegravir and lamivudine was approved.
been confirmed [176], and further studies are needed to elucidate as­ Although pharmacokinetics and antiviral efficacy of lamivudine and
sociations between long-term metabolic effects or cancer later in life and dolutegravir in pregnancy have been investigated [93,189,190],

8
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

dolutegravir and lamivudine is not recommended as a complete regimen [203] to acquire and employ more accurate data on dosing, safety
in pregnancy because data on antiviral efficacy are lacking [8,67]. Other and efficacy of ART for pregnant women.
antiretrovirals that can be potentially used in pregnancy are doravirine • Data on transport of antiretrovirals across the placenta is typically
and bictegravir. Doravirine is active against HIV resistant to efavirenz derived from post-birth comparison of umbilical and maternal drug
and nevirapine, less susceptible to development of resistance than ril­ concentrations. However, this approach provides little information
pivirine and efavirenz, and expected to show lower propensity to drug- on their placental transport kinetics and fetal exposure earlier in
drug interactions. Therefore, it can be prospectively considered as an pregnancy. In addition, they are often structurally related to nucle­
alternative to INSTIs [67]. Bictegravir has also shown high efficacy and osides, so they are potential substrates or inhibitors of nucleoside
tolerability in non-pregnant adults in clinical trials [191,192]. transporters (ENT and/or CNT). Nevertheless, these transporters’
Regarding bictegravir’s effects on the developing fetus, first signals from expression and function in the syncytiotrophoblast apical or basal
clinical trials with a cohort of 25 pregnant women indicate that bicte­ membrane have not been satisfactorily elucidated. Thus, their role in
gravir does not cause neural tube defects [193]. transplacental passage of antiretrovirals is not known, and further
It should be stressed that not all adult patients with HIV fully profit research on their transplacental transport must continue, employing
from ART. More than 25% of ART-naive patients initiating pharmaco­ all possible methods, so both broader and more detailed knowledge
therapy reportedly have episodes of non-adherence that potentially can be incorporated into clinical reasoning before ART is initiated.
threaten individuals’ prognosis and increase risks for horizontal and/or • The enormous prophylactic efficacy of antiretrovirals in prevention
vertical HIV transmission [194–196]. Therefore, there have been recent of MTCT might have obscured their long-term toxicity. However,
efforts to develop and evaluate long-acting regimens [67,196] that could possible delayed adverse effects require much longer afterbirth
substitute daily administration of antiretrovirals. This could help monitoring because they become observable later in life. Many
improve problematic patients’ adherence to ART. Moreover, recent antiretrovirals interact with various biological targets in the
surveys have demonstrated that long-acting regimens would be a placenta, such as transporters or enzymes (others will be revealed in
welcomed option for youths living with HIV [197,198]. The first and the future) that might affect placental functions and fetal program­
only once-monthly, complete long-acting regimen that might replace the ming. For example, inhibitory effects of antiretrovirals on mono­
current antiretroviral regimen for adult patients who are stably viro­ amine transporters have been observed [204]. If administered in
logically suppressed [199] is a combination of cabotegravir and rilpi­ pregnancy, they might significantly perturb homeostasis of seroto­
virine. At the time of writing this is being reviewed by the FDA, but the nin, dopamine, or norepinephrine in the fetoplacental unit and affect
European Medicines Agency has already recommended marketing au­ programming of the fetal brain [185]. Several cases of impaired
thorizations. Cabotegravir was primarily developed as an injectable neurodevelopment have been reported in the literature, but without
formulation and both cabotegravir and rilpivirine are used as a nano­ causative mechanisms. It is thus essential to monitor and document
suspension [200]. Another long-acting antiretroviral drug is ibalizumab, long-term/delayed toxicity that antiretroviral drugs might trigger.
a humanized anti-CD4 IgG4 monoclonal antibody. Ibalizumab blocks • Novel drugs and drug formulations in the pipelines of pharmaceu­
entry of HIV-1 into CD4-positive cells and must be administered once tical research hold promise of more effective and less toxic therapy.
every 14 days via intravenous infusion. However, due to high treatment Long-acting antiretrovirals, such as monoclonal antibodies or nano­
costs and the need of administration by trained medical professionals, suspensions of small molecules, are being designed for once or twice
ibalizumab is used only in treatment-experienced patients with multi- monthly administrations. This approach might replace daily ART in
drug resistant HIV strains [201,202]. individuals with low adherence.
There are at least two more promising investigational antiretrovirals
in the pipeline: islatravir (the first nucleoside reverse transcriptase A clear overall conclusion is that continuous research, both basic and
translocation inhibitor) and an NNRTI, dapivirine. Both of these drugs translational, in the ART of pregnant women is crucial to ensure well-
are very potent against HIV-1, including multidrug-resistant strains, and balanced, efficient, and safe treatment of both the mothers and their
their pharmacokinetic and physico-chemical properties allow produc­ children.
tion of long-acting antiretroviral formulations. Whereas islatravir has
been tested in oral, vaginal microbicide film, and subdermal drug-
eluting polymer implant formulations, dapivirine has been trialed as a Declaration of competing interest
component of a one-month vaginal ring [67], which is currently under
review by the European Medicines Agency. None.

10. Conclusions and personal opinion Acknowledgements

Since the first use of antiretroviral drug in gestation in 1994, there The authors are thankful to Dr. Rona Karahoda for drawing Fig. 2.
has been tremendous progress in treatment of HIV-positive pregnant The work of F.S. and L.C. on transplacental pharmacokinetics of
women. Nowadays, up to 85% of HIV-positive pregnant women have antivirals was supported by the Czech Science Foundation (grant no.
access to ART. In addition, dramatic reduction in the incidence of HIV GACR 17-16169S) and the EFSA-CDN project [No. CZ.02.1.01/0.0/0.0/
MTCT is one of the biggest healthcare achievements in the modern era. 16_019/0000841], which is co-funded by the ERDF.
However, these accomplishments should not stop efforts to make further
improvements or prevent us from seeing possible harmful effects. References

[1] World Health Organization, https://www.who.int/news-room/fact-sheets/de


• Pregnant women are generally excluded from clinical studies, so
tail/hiv-aids, [accessed 22nd October 2020].
prescription of ART is based on information obtained from non- [2] A.P. Kourtis, M. Bulterys, S.R. Nesheim, F.K. Lee, Understanding the timing of
pregnant populations, largely ignoring possible differences in ef­ HIV transmission from mother to infant, JAMA 285 (2001) 709–712.
fects associated with gestation. Strictly controlled clinical studies [3] V. Leroy, M.L. Newell, F. Dabis, C. Peckham, P. Van de Perre, M. Bulterys,
C. Kind, R.J. Simonds, S. Wiktor, P. Msellati, International multicentre pooled
with drugs well tolerated in phase IV should be considered by reg­ analysis of late postnatal mother-to-child transmission of HIV-1 infection. Ghent
ulatory agencies to rationalize dosing regimens. As previously rec­ International Working Group on Mother-to-Child Transmission of HIV, Lancet
ommended, a “formalized step-wise approach to including pregnant 352 (1998) 597–600.
[4] S. Caffe, F. Perez, M.L. Kamb, R. Gomez Ponce de Leon, M. Alonso, R. Midy,
women in antiretroviral drug research should become the new norm” L. Newman, C. Hayashi, M. Ghidinelli, Cuba validated as the first country to
eliminate mother-to-child transmission of human immunodeficiency virus and

9
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

congenital syphilis: lessons learned from the implementation of the Global [30] G. Vidricaire, M.R. Tardif, M.J. Tremblay, The low viral production in
validation methodology, Sex. Transm. Dis. 43 (2016) 733–736. trophoblastic cells is due to a high endocytic internalization of the human
[5] S.R. Nesheim, L.F. FitzHarris, K. Mahle Gray, M.A. Lampe, Epidemiology of immunodeficiency virus type 1 and can be overcome by the pro-inflammatory
perinatal HIV transmission in the United States in the era of its elimination, cytokines tumor necrosis factor-alpha and interleukin-1, J. Biol. Chem. 278
Pediatr. Infect. Dis. J. 38 (2019) 611–616. (2003) 15832–15841.
[6] M.M. Andrews, D.S. Storm, C.K. Burr, E. Aaron, M.J. Hoyt, A. Statton, S. Weber, [31] Y. Tang, B.O. Woodward, L. Pastor, A.M. George, O. Petrechko, F.J. Nouvet, D.
Perinatal HIV service coordination: closing gaps in the HIV care continuum for W. Haas, G. Jiang, J.E.K. Hildreth, Endogenous retroviral envelope syncytin
pregnant women and eliminating perinatal HIV transmission in the United States, induces HIV-1 spreading and establishes HIV reservoirs in placenta, Cell Rep. 30
Public Health Rep. 133 (2018) 532–542. (2020) 4528–4539 (e4524).
[7] S. Blanche, Mini review: prevention of mother-child transmission of HIV: 25 years [32] M. Derrien, A. Faye, G. Dolcini, G. Chaouat, F. Barre-Sinoussi, E. Menu, Impact of
of continuous progress toward the eradication of pediatric AIDS? Virulence 11 the placental cytokine-chemokine balance on regulation of cell-cell contact-
(2020) 14–22. induced human immunodeficiency virus type 1 translocation across a
[8] Panel on Treatment of Pregnant Women with HIV Infection and Prevention of trophoblastic barrier in vitro, J. Virol. 79 (2005) 12304–12310.
Perinatal Transmission. Recommendations for use of antiretroviral drugs in [33] L.J. Else, S. Taylor, D.J. Back, S.H. Khoo, Pharmacokinetics of antiretroviral drugs
transmission in the United States. Available at http://aidsinfo.nih.gov/ in anatomical sanctuary sites: the fetal compartment (placenta and amniotic
contentfiles/lvguidelines/PerinatalGL.pdf. Accessed (12th October 2020). fluid), Antivir. Ther. 16 (2011) 1139–1147.
[9] A.C. Eke, A. Olagunju, B.M. Best, M. Mirochnick, J.D. Momper, E. Abrams, [34] E. Jauniaux, C. Nessmann, M.C. Imbert, S. Meuris, F. Puissant, J. Hustin,
M. Penazzato, T.R. Cressey, A. Colbers, Innovative approaches for pharmacology Morphological aspects of the placenta in HIV pregnancies, Placenta 9 (1988)
studies in pregnant and lactating women: a viewpoint and lessons from HIV, Clin. 633–642.
Pharmacokinet. 59 (2020) 1185–1194. [35] D.A. Schwartz, S. Sungkarat, N. Shaffer, J. Laosakkitiboran, W. Supapol,
[10] S.A. Spector, Mother-to-infant transmission of HIV-1: the placenta fights back, P. Charoenpanich, T. Chuangsuwanich, T.D. Mastro, Placental abnormalities
J. Clin. Invest. 107 (2001) 267–269. associated with human immunodeficiency virus type 1 infection and perinatal
[11] S.R. Ellington, C.C. King, A.P. Kourtis, Host factors that influence mother-to-child transmission in Bangkok, Thailand, J. Infect. Dis. 182 (2000) 1652–1657.
transmission of HIV-1: genetics, coinfections, behavior and nutrition, Futur. Virol. [36] P.B. Gichangi, A.O. Nyongo, M. Temmerman, Pregnancy outcome and placental
6 (2011) 1451–1469. weights: their relationship to HIV-1 infection, East Afr. Med. J. 70 (1993) 85–89.
[12] E.T. Marinda, L.H. Moulton, J.H. Humphrey, J.W. Hargrove, R. Ntozini, [37] S. Chandwani, M.A. Greco, K. Mittal, C. Antoine, K. Krasinski, W. Borkowsky,
K. Mutasa, J. Levin, In utero and intra-partum HIV-1 transmission and acute HIV- Pathology and human immunodeficiency virus expression in placentas of
1 infection during pregnancy: using the BED capture enzyme-immunoassay as a seropositive women, J. Infect. Dis. 163 (1991) 1134–1138.
surrogate marker for acute infection, Int. J. Epidemiol. 40 (2011) 945–954. [38] R.W. Ryder, W. Nsa, S.E. Hassig, F. Behets, M. Rayfield, B. Ekungola, A.M. Nelson,
[13] T.E. Taha, M.M. James, D.R. Hoover, J. Sun, O. Laeyendecker, C.E. Mullis, J. U. Mulenda, H. Francis, K. Mwandagalirwa, et al., Perinatal transmission of the
J. Kumwenda, J.R. Lingappa, B. Auvert, C.S. Morrison, L.M. Mofensen, A. Taylor, human immunodeficiency virus type 1 to infants of seropositive women in Zaire,
M.G. Fowler, N.I. Kumenda, S.H. Eshleman, Association of recent HIV infection N. Engl. J. Med. 320 (1989) 1637–1642.
and in-utero HIV-1 transmission, AIDS 25 (2011) 1357–1364. [39] E. Backe, E. Jimenez, M. Unger, A. Schafer, M. Vocks-Hauck, I. Grosch-Worner,
[14] H. Minkoff, Human immunodeficiency virus infection in pregnancy, Obstet. M. Vogel, Vertical human immunodeficiency virus transmission: a study of
Gynecol. 101 (2003) 797–810. placental pathology in relation to maternal risk factors, Am. J. Perinatol. 11
[15] A.M. Al-Husaini, Role of placenta in the vertical transmission of human (1994) 326–330.
immunodeficiency virus, J. Perinatol. 29 (2009) 331–336. [40] K. Lindholm, M. O’Keefe, Placental cytomegalovirus infection, Arch. Pathol. Lab.
[16] S. Lagaye, M. Derrien, E. Menu, C. Coito, E. Tresoldi, P. Mauclere, G. Scarlatti, G. Med. 143 (2019) 639–642.
Chaouat, F. Barre-Sinoussi, M. Bomsel, H.I.V. European Network for the Study of [41] A. Vermaak, G.B. Theron, P.T. Schubert, M. Kidd, U. Rabie, B.M. Adjiba, C.
In Utero Transmission of, Cell-to-cell contact results in a selective translocation of A. Wright, Morphologic changes in the placentas of HIV-positive women and their
maternal human immunodeficiency virus type 1 quasispecies across a association with degree of immune suppression, Int. J. Gynaecol. Obstet. 119
trophoblastic barrier by both transcytosis and infection, J Virol, 75 (2001) (2012) 239–243.
4780–4791. [42] L. Bhoopat, S. Khunamornpong, P. Sirivatanapa, T. Rithaporn, P. Lerdsrimongkol,
[17] A.S. Bourinbaiar, S. Lee-Huang, Anti-HIV effect of beta subunit of human P.S. Thorner, T. Bhoopat, Chorioamnionitis is associated with placental
chorionic gonadotropin (beta hCG) in vitro, Immunol. Lett. 44 (1995) 13–18. transmission of human immunodeficiency virus-1 subtype E in the early
[18] E.L. Johnson, R. Chakraborty, Placental Hofbauer cells limit HIV-1 replication gestational period, Mod. Pathol. 18 (2005) 1357–1364.
and potentially offset mother to child transmission (MTCT) by induction of [43] O.C. Castejon, A.J. Lopez, L.M. Perez Ybarra, O.C. Castejon, Placental villous
immunoregulatory cytokines, Retrovirology 9 (2012) 101. lesions in HIV-1 infection treated with zidovudine, Ginecol. Obstet. Mex. 79
[19] R.E. Dickover, E.M. Garratty, S. Plaeger, Y.J. Bryson, Perinatal transmission of (2011) 269–279.
major, minor, and multiple maternal human immunodeficiency virus type 1 [44] C.O. Wedi, S. Kirtley, S. Hopewell, R. Corrigan, S.H. Kennedy, J. Hemelaar,
variants in utero and intrapartum, J. Virol. 75 (2001) 2194–2203. Perinatal outcomes associated with maternal HIV infection: a systematic review
[20] S.H. Lewis, C. Reynolds-Kohler, H.E. Fox, J.A. Nelson, HIV-1 in trophoblastic and and meta-analysis, Lancet HIV 3 (2016) e33–e48.
villous Hofbauer cells, and haematological precursors in eight-week fetuses, [45] L. Afran, M. Garcia Knight, E. Nduati, B.C. Urban, R.S. Heyderman, S.L. Rowland-
Lancet 335 (1990) 565–568. Jones, HIV-exposed uninfected children: a growing population with a vulnerable
[21] S.H. Landesman, L.A. Kalish, D.N. Burns, H. Minkoff, H.E. Fox, C. Zorrilla, immune system? Clin. Exp. Immunol. 176 (2014) 11–22.
P. Garcia, M.G. Fowler, L. Mofenson, R. Tuomala, Obstetrical factors and the [46] M. Altfeld, M.J. Bunders, Impact of HIV-1 infection on the feto-maternal crosstalk
transmission of human immunodeficiency virus type 1 from mother to child. The and consequences for pregnancy outcome and infant health, Semin.
women and infants transmission study, N. Engl. J. Med. 334 (1996) 1617–1623. Immunopathol. 38 (2016) 727–738.
[22] G. John-Stewart, D. Mbori-Ngacha, R. Ekpini, E.N. Janoff, J. Nkengasong, J.S. [47] Y. Yang, P.L. Xiao, Y. Yang, J.C. Gao, Y. Shi, W.T. Cheng, Y. Chen, X.X. Song, Q.
Read, P. Van de Perre, M.L. Newell, I.A.S.W.G.o.H.I.V.i.W.C. Ghent, Breast- W. Jiang, Y.B. Zhou, Immune dysfunction and coinfection with human
feeding and transmission of HIV-1, J Acquir Immune Defic Syndr, 35 (2004) immunodeficiency virus and schistosoma japonicum in Yi people, J Immunol Res
196–202. 2018 (2018) 6989717.
[23] S.M. Tugizov, R. Herrera, P. Veluppillai, D. Greenspan, V. Soros, W.C. Greene, J. [48] J.E. Lawn, M. Kinney, Preterm birth: now the leading cause of child death
A. Levy, J.M. Palefsky, Differential transmission of HIV traversing fetal oral/ worldwide, Sci Transl Med, 6 (2014) 263ed221.
intestinal epithelia and adult oral epithelia, J. Virol. 86 (2012) 2556–2570. [49] K.J. Nakamura, L. Heath, E.R. Sobrera, T.A. Wilkinson, K. Semrau, C. Kankasa, N.
[24] R.D. Semba, N. Kumwenda, D.R. Hoover, T.E. Taha, T.C. Quinn, L. Mtimavalye, H. Tobin, N.E. Webb, B. Lee, D.M. Thea, L. Kuhn, J.I. Mullins, G.M. Aldrovandi,
R.J. Biggar, R. Broadhead, P.G. Miotti, L.J. Sokoll, L. van der Hoeven, J. Breast milk and in utero transmission of HIV-1 select for envelope variants with
D. Chiphangwi, Human immunodeficiency virus load in breast milk, mastitis, and unique molecular signatures, Retrovirology 14 (2017) 6.
mother-to-child transmission of human immunodeficiency virus type 1, J. Infect. [50] P. Cools, J. van de Wijgert, V. Jespers, T. Crucitti, E.J. Sanders, H. Verstraelen,
Dis. 180 (1999) 93–98. M. Vaneechoutte, Role of HIV exposure and infection in relation to neonatal GBS
[25] R.D. Semba, N. Kumwenda, T.E. Taha, D.R. Hoover, T.C. Quinn, Y. Lan, disease and rectovaginal GBS carriage: a systematic review and meta-analysis,
L. Mtimavalye, R. Broadhead, P.G. Miotti, L. van der Hoeven, J.D. Chiphangwi, Sci. Rep. 7 (2017) 13820.
Mastitis and immunological factors in breast milk of human immunodeficiency [51] S.K.G. Jensen, A.E. Berens, C.A. Nelson 3rd, Effects of poverty on interacting
virus-infected women, J. Hum. Lact. 15 (1999) 301–306. biological systems underlying child development, Lancet Child Adolesc. Health 1
[26] V. Dahl, L. Josefsson, S. Palmer, HIV reservoirs, latency, and reactivation: (2017) 225–239.
prospects for eradication, Antivir. Res. 85 (2010) 286–294. [52] Joint United Nations Programme on HIV/AIDS, Miles to go, closing gaps,
[27] G. Vidricaire, S. Gauthier, M.J. Tremblay, HIV-1 infection of trophoblasts is breaking barriers, rightning injustice, global AIDS update 2018; https://www.un
independent of gp120/CD4 interactions but relies on heparan sulfate aids.org/sites/default/files/media_asset/miles-to-go_en.pdf [accesssed 18th
proteoglycans, J. Infect. Dis. 195 (2007) 1461–1471. December 2020].
[28] R.A. Arias, L.D. Munoz, M.A. Munoz-Fernandez, Transmission of HIV-1 infection [53] S.A. Hurst, K.E. Appelgren, A.P. Kourtis, Prevention of mother-to-child
between trophoblast placental cells and T-cells take place via an LFA-1-mediated transmission of HIV type 1: the role of neonatal and infant prophylaxis, Expert
cell to cell contact, Virology 307 (2003) 266–277. Rev. Anti-Infect. Ther. 13 (2015) 169–181.
[29] F.J. David, B. Autran, H.C. Tran, E. Menu, M. Raphael, P. Debre, B.L. Hsi, T. [54] L. Mandelbrot, R. Tubiana, J. Le Chenadec, C. Dollfus, A. Faye, E. Pannier,
G. Wegman, F. Barre-Sinoussi, G. Chaouat, Human trophoblast cells express CD4 S. Matheron, M.A. Khuong, V. Garrait, V. Reliquet, A. Devidas, A. Berrebi,
and are permissive for productive infection with HIV-1, Clin. Exp. Immunol. 88 C. Allisy, C. Elleau, C. Arvieux, C. Rouzioux, J. Warszawski, S. Blanche, A.-E.S.
(1992) 10–16.

10
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

Group, No perinatal HIV-1 transmission from women with effective antiretroviral pharmacokinetics with and without tenofovir during pregnancy, J Acquir
therapy starting before conception, Clin. Infect. Dis. 61 (2015) 1715–1725. Immune Defic Syndr, 56 (2011) 412–419.
[55] Joint United Nations Programme on HIV/AIDS, https://www.unaids.org/en/ke [78] A. Stek, B.M. Best, J. Wang, E.V. Capparelli, S.K. Burchett, R. Kreitchmann,
ywords/children [accessed 18th December 2020]. K. Rungruengthanakit, T.R. Cressey, L.M. Mofenson, E. Smith, D. Shapiro,
[56] J.C. Mutabazi, C. Gray, L. Muhwava, H. Trottier, L.J. Ware, S. Norris, K. Murphy, M. Mirochnick, Pharmacokinetics of once versus twice daily darunavir in
N. Levitt, C. Zarowsky, Integrating the prevention of mother-to-child transmission pregnant HIV-infected women, J. Acquir. Immune Defic. Syndr. 70 (2015) 33–41.
of HIV into primary healthcare services after AIDS denialism in South Africa: [79] A.H. Salem, A.K. Jones, M. Santini-Oliveira, G.P. Taylor, K.B. Patterson, A.
perspectives of experts and health care workers - a qualitative study, BMC Health M. Nilius, C.E. Klein, No need for lopinavir dose adjustment during pregnancy: a
Serv. Res. 20 (2020) 582. population pharmacokinetic and exposure-response analysis in pregnant and
[57] H. Jones, A. Wringe, J. Todd, J. Songo, F.X. Gomez-Olive, M. Moshabela, nonpregnant HIV-infected subjects, Antimicrob. Agents Chemother. 60 (2016)
E. Geubbels, M. Nyamhagatta, T. Kalua, M. Urassa, B. Zaba, J. Renju, 400–408.
Implementing prevention policies for mother-to-child transmission of HIV in rural [80] M. Mirochnick, B.M. Best, A.M. Stek, E. Capparelli, C. Hu, S.K. Burchett, D.T.
Malawi, South Africa and United Republic of Tanzania, 2013–2016, Bull. World Holland, E. Smith, S. Gaddipati, J.S. Read, P.s.S. Team, Lopinavir exposure with
Health Organ. 97 (2019) 200–212. an increased dose during pregnancy, J Acquir Immune Defic Syndr, 49 (2008)
[58] E.M. Connor, R.S. Sperling, R. Gelber, P. Kiselev, G. Scott, M.J. O’Sullivan, 485-491.
R. VanDyke, M. Bey, W. Shearer, R.L. Jacobson, et al., Reduction of maternal- [81] R. Kreitchmann, B.M. Best, J. Wang, A. Stek, E. Caparelli, D.H. Watts, E. Smith, D.
infant transmission of human immunodeficiency virus type 1 with zidovudine E. Shapiro, S. Rossi, S.K. Burchett, E. Hawkins, M. Byroads, T.R. Cressey,
treatment. Pediatric AIDS Clinical Trials Group Protocol 076 Study Group, M. Mirochnick, Pharmacokinetics of an increased atazanavir dose with and
N. Engl. J. Med. 331 (1994) 1173–1180. without tenofovir during the third trimester of pregnancy, J. Acquir. Immune
[59] P. Pavek, M. Ceckova, F. Staud, Variation of drug kinetics in pregnancy, Curr. Defic. Syndr. 63 (2013) 59–66.
Drug Metab. 10 (2009) 520–529. [82] Z.H. Israili, P.G. Dayton, Human alpha-1-glycoprotein and its interactions with
[60] F.T. Aweeka, A. Stek, B.M. Best, C. Hu, D. Holland, A. Hermes, S.K. Burchett, J. drugs, Drug Metab. Rev. 33 (2001) 161–235.
Read, M. Mirochnick, E.V. Capparelli, A.C.T.G.P.P.T. International Maternal [83] N. Holmstock, P. Annaert, P. Augustijns, Boosting of HIV protease inhibitors by
Pediatric Adolescent, Lopinavir protein binding in HIV-1-infected pregnant ritonavir in the intestine: the relative role of cytochrome P450 and P-glycoprotein
women, HIV Med, 11 (2010) 232-238. inhibition based on Caco-2 monolayers versus in situ intestinal perfusion in mice,
[61] D. Metsu, P.L. Toutain, E. Chatelut, P. Delobel, P. Gandia, Antiretroviral unbound Drug Metab. Dispos. 40 (2012) 1473–1477.
concentration during pregnancy: piece of interest in the puzzle? J. Antimicrob. [84] F. Li, J. Lu, X. Ma, CYP3A4-mediated lopinavir bioactivation and its inhibition by
Chemother. 72 (2017) 2407–2409. ritonavir, Drug Metab. Dispos. 40 (2012) 18–24.
[62] K.B. Patterson, J.B. Dumond, H.A. Prince, A.J. Jenkins, K.K. Scarsi, R. Wang, [85] L. Cerveny, Z. Ptackova, M. Durisova, F. Staud, Interactions of protease inhibitors
S. Malone, M.G. Hudgens, A.D. Kashuba, Protein binding of lopinavir and atazanavir and ritonavir with ABCB1, ABCG2, and ABCC2 transporters: effect on
ritonavir during 4 phases of pregnancy: implications for treatment guidelines, transplacental disposition in rats, Reprod. Toxicol. 79 (2018) 57–65.
J. Acquir. Immune Defic. Syndr. 63 (2013) 51–58. [86] E.I. Lepist, T.K. Phan, A. Roy, L. Tong, K. Maclennan, B. Murray, A.S. Ray,
[63] R. van der Galien, R. Ter Heine, R. Greupink, S.J. Schalkwijk, A.E. van Cobicistat boosts the intestinal absorption of transport substrates, including HIV
Herwaarden, A. Colbers, D.M. Burger, Pharmacokinetics of HIV-integrase protease inhibitors and GS-7340, in vitro, Antimicrob. Agents Chemother. 56
inhibitors during pregnancy: mechanisms, clinical implications and knowledge (2012) 5409–5413.
gaps, Clin. Pharmacokinet. 58 (2019) 309–323. [87] S.D. Boyd, M.R. Sampson, P. Viswanathan, K.A. Struble, V. Arya, A.I. Sherwat,
[64] T.R. Cressey, L. Harrison, J. Achalapong, P. Kanjanavikai, O. Patamasingh Na Cobicistat-containing antiretroviral regimens are not recommended during
Ayudhaya, P. Liampongsabuddhi, T. Siriwachirachai, C. Putiyanun, P. Suriyachai, pregnancy: viewpoint, AIDS 33 (2019) 1089–1093.
C. Tierney, N. Salvadori, D. Chinwong, L. Decker, Y. Tawon, T.V. Murphy, N. Ngo- [88] J.D. Momper, B.M. Best, J. Wang, E.V. Capparelli, A. Stek, E. Barr, M.L. Badell, E.
Giang-Huong, G.K. Siberry, G. Jourdain, T.A.P.S.T. i, Tenofovir exposure during P. Acosta, M. Purswani, E. Smith, N. Chakhtoura, K. Park, S. Burchett, D.
pregnancy and postpartum in women receiving tenofovir disoproxil fumarate for E. Shapiro, M. Mirochnick, I.P.P. Team, Elvitegravir/cobicistat pharmacokinetics
the prevention of mother-to-child transmission of hepatitis B virus, Antimicrob in pregnant and postpartum women with HIV, AIDS 32 (2018) 2507–2516.
Agents Chemother, 62 (2018). [89] M. Lamorde, X. Wang, M. Neary, E. Bisdomini, S. Nakalema, P. Byakika-Kibwika,
[65] G. Pariente, T. Leibson, A. Carls, T. Adams-Webber, S. Ito, G. Koren, Pregnancy- J.K. Mukonzo, W. Khan, A. Owen, M. McClure, M. Boffito, Pharmacokinetics,
associated changes in pharmacokinetics: a systematic review, PLoS Med. 13 pharmacodynamics, and pharmacogenetics of efavirenz 400 mg once daily during
(2016), e1002160. pregnancy and post-partum, Clin. Infect. Dis. 67 (2018) 785–790.
[66] N. Mulligan, S. Schalkwijk, B.M. Best, A. Colbers, J. Wang, E.V. Capparelli, [90] E.S. Group, D. Carey, R. Puls, J. Amin, M. Losso, P. Phanupak, S. Foulkes,
J. Molto, A.M. Stek, G. Taylor, E. Smith, C. Hidalgo Tenorio, N. Chakhtoura, L. Mohapi, B. Crabtree-Ramirez, H. Jessen, S. Kumar, A. Winston, M.P. Lee,
M. van Kasteren, C.V. Fletcher, M. Mirochnick, D. Burger, Etravirine W. Belloso, D.A. Cooper, S. Emery, Efficacy and safety of efavirenz 400 mg daily
pharmacokinetics in HIV-infected pregnant women, Front. Pharmacol. 7 (2016) versus 600 mg daily: 96-week data from the randomised, double-blind, placebo-
239. controlled, non-inferiority ENCORE1 study, Lancet Infect. Dis. 15 (2015)
[67] M. Neary, A. Owen, A. Olagunju, Pharmacokinetics of HIV therapies in pregnant 793–802.
patients: an update, Expert Opin. Drug Metab. Toxicol. 16 (2020) 449–461. [91] A.M. Stek, B.M. Best, W. Luo, E. Capparelli, S. Burchett, C. Hu, H. Li, J.S. Read,
[68] N. Isoherranen, K.E. Thummel, Drug metabolism and transport during pregnancy: A. Jennings, E. Barr, E. Smith, S.S. Rossi, M. Mirochnick, Effect of pregnancy on
how does drug disposition change during pregnancy and what are the emtricitabine pharmacokinetics, HIV Med. 13 (2012) 226–235.
mechanisms that cause such changes? Drug Metab. Dispos. 41 (2013) 256–262. [92] B.M. Best, M. Mirochnick, E.V. Capparelli, A. Stek, S.K. Burchett, D.T. Holland, J.
[69] T.S. Tracy, R. Venkataramanan, D.D. Glover, S.N. Caritis, H. National Institute for S. Read, E. Smith, C. Hu, S.A. Spector, J.D. Connor, P.P.S. Team, Impact of
Child, U. Human Development Network of Maternal-Fetal-Medicine, Temporal pregnancy on abacavir pharmacokinetics, AIDS 20 (2006) 553–560.
changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A Activity) during [93] J. Moodley, D. Moodley, K. Pillay, H. Coovadia, J. Saba, R. van Leeuwen,
pregnancy, Am J Obstet Gynecol, 192 (2005) 633–639. C. Goodwin, P.R. Harrigan, K.H. Moore, C. Stone, R. Plumb, M.A. Johnson,
[70] M. Dawes, P.J. Chowienczyk, Drugs in pregnancy. Pharmacokinetics in Pharmacokinetics and antiretroviral activity of lamivudine alone or when
pregnancy, Best Pract. Res. Clin. Obstet. Gynaecol. 15 (2001) 819–826. coadministered with zidovudine in human immunodeficiency virus type 1-
[71] B. Krauer, F. Krauer, F.E. Hytten, Drug disposition and pharmacokinetics in the infected pregnant women and their offspring, J. Infect. Dis. 178 (1998)
maternal-placental-fetal unit, Pharmacol. Ther. 10 (1980) 301–328. 1327–1333.
[72] B. Krauer, P. Dayer, R. Anner, Changes in serum albumin and alpha 1-acid [94] V. Bukkems, C. Necsoi, C.H. Tenorio, C. Garcia, J. Rockstroh, C. Schwarze-
glycoprotein concentrations during pregnancy: an analysis of fetal-maternal Zander, J.S. Lambert, D. Burger, D. Konopnicki, A. Colbers, P. network, Clinically
pairs, Br. J. Obstet. Gynaecol. 91 (1984) 875–881. significant lower elvitegravir exposure during third trimester of pregnant patients
[73] E. Perucca, A. Crema, Plasma protein binding of drugs in pregnancy, Clin. living with HIV: data from the PANNA study, Clin Infect Dis, (2020).
Pharmacokinet. 7 (1982) 336–352. [95] L. Novakova, J. Pavlik, L. Chrenkova, O. Martinec, L. Cerveny, Current antiviral
[74] A.C. Eke, A.M. Stek, J. Wang, R. Kreitchmann, D.E. Shapiro, E. Smith, drugs and their analysis in biological materials - part II: antivirals against
N. Chakhtoura, E.V. Capparelli, M. Mirochnick, B.M. Best, I.P.P. Team, Darunavir hepatitis and HIV viruses, J. Pharm. Biomed. Anal. 147 (2018) 378–399.
pharmacokinetics with an increased dose during pregnancy, J. Acquir. Immune [96] L. Novakova, J. Pavlik, L. Chrenkova, O. Martinec, L. Cerveny, Current antiviral
Defic. Syndr. 83 (2020) 373–380. drugs and their analysis in biological materials-part I: antivirals against
[75] S. Schalkwijk, R. Ter Heine, A. Colbers, E. Capparelli, B.M. Best, T.R. Cressey, respiratory and herpes viruses, J. Pharm. Biomed. Anal. 147 (2018) 400–416.
R. Greupink, F.G.M. Russel, J. Molto, M. Mirochnick, M.O. Karlsson, D.M. Burger, [97] B. O’Kelly, R. Murtagh, J.S. Lambert, Therapeutic drug monitoring of HIV
Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant antiretroviral drugs in pregnancy: a narrative review, Ther. Drug Monit. 42
women using semi-mechanistic pharmacokinetic modelling, J. Antimicrob. (2020) 229–244.
Chemother. 74 (2019) 1348–1356. [98] M. De Sousa Mendes, D. Hirt, S. Urien, E. Valade, N. Bouazza, F. Foissac,
[76] A. Colbers, D. Hawkins, C. Hidalgo-Tenorio, M. van der Ende, A. Gingelmaier, K. S. Blanche, J.M. Treluyer, S. Benaboud, Physiologically-based pharmacokinetic
Weizsacker, K. Kabeya, G. Taylor, J. Rockstroh, J. Lambert, J. Molto, C. Wyen, S. modeling of renally excreted antiretroviral drugs in pregnant women, Br. J. Clin.
T. Sadiq, J. Ivanovic, C. Giaquinto, D. Burger, P. network, Atazanavir exposure is Pharmacol. 80 (2015) 1031–1041.
effective during pregnancy regardless of tenofovir use, Antivir Ther, 20 (2015) [99] A. Colbers, R. Greupink, C. Litjens, D. Burger, F.G. Russel, Physiologically based
57-64. modelling of darunavir/ritonavir pharmacokinetics during pregnancy, Clin.
[77] M. Mirochnick, B.M. Best, A.M. Stek, E.V. Capparelli, C. Hu, S.K. Burchett, S.S. Pharmacokinet. 55 (2016) 381–396.
Rossi, E. Hawkins, M. Basar, E. Smith, J.S. Read, I.s.S. Team, Atazanavir [100] M. Gockenbach, Physiologically-based pharmacokinetic modeling of rilpivirine
during pregnancy (oral abstract 16). 20th International Workshop on Clinical

11
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

Pharmacology of HIV, Hepatitis, and Other Antiviral Drugs. Noordwijk, the the human intestine, liver, kidneys, and placenta, Am. J. Physiol. Regul. Integr.
Netherlands. 14–16 May 2019. Available at: http://regist2.virology-education.co Comp. Physiol. 293 (2007) R1809–R1822.
m/presentations/2019/20AntiviralPK/33_Gockenbach.pdf. [accessed 19th [124] M. Molina-Arcas, F.J. Casado, M. Pastor-Anglada, Nucleoside transporter
November 2020]. proteins, Curr. Vasc. Pharmacol. 7 (2009) 426–434.
[101] U.S. Food and Drug Administration. HIV Treatment Information for Pregnant [125] S. Karbanova, L. Cerveny, M. Ceckova, Z. Ptackova, L. Jiraskova, S. Greenwood,
Patients. https://www.fda.gov/drugs/hiv-treatment/hiv-treatment-information-p F. Staud, Role of nucleoside transporters in transplacental pharmacokinetics of
regnant-patients [accessed 2nd December 2020]. nucleoside reverse transcriptase inhibitors zidovudine and emtricitabine,
[102] F. Staud, L. Cerveny, M. Ceckova, Pharmacotherapy in pregnancy; effect of ABC Placenta 60 (2017) 86–92.
and SLC transporters on drug transport across the placenta and fetal drug [126] Q. Zeng, M. Bai, C. Li, S. Lu, Z. Ma, Y. Zhao, H. Zhou, H. Jiang, D. Sun, C. Zheng,
exposure, J. Drug Target. 20 (2012) 736–763. Multiple drug transporters contribute to the placental transfer of emtricitabine,
[103] D. Ahmadimoghaddam, L. Zemankova, P. Nachtigal, E. Dolezelova, Antimicrob. Agents Chemother. 63 (2019).
Z. Neumanova, L. Cerveny, M. Ceckova, M. Kacerovsky, S. Micuda, F. Staud, [127] T. Yamamoto, K. Kuniki, Y. Takekuma, T. Hirano, K. Iseki, M. Sugawara,
Organic cation transporter 3 (OCT3/SLC22A3) and multidrug and toxin extrusion Ribavirin uptake by cultured human choriocarcinoma (BeWo) cells and Xenopus
1 (MATE1/SLC47A1) transporter in the placenta and fetal tissues: expression laevis oocytes expressing recombinant plasma membrane human nucleoside
profile and fetus protective role at different stages of gestation, Biol. Reprod. 88 transporters, Eur. J. Pharmacol. 557 (2007) 1–8.
(2013) 55. [128] E. Errasti-Murugarren, P. Diaz, V. Godoy, G. Riquelme, M. Pastor-Anglada,
[104] L. Cerveny, Z. Ptackova, M. Ceckova, R. Karahoda, S. Karbanova, L. Jiraskova, S. Expression and distribution of nucleoside transporter proteins in the human
L. Greenwood, J.D. Glazier, F. Staud, Equilibrative nucleoside transporter 1 syncytiotrophoblast, Mol. Pharmacol. 80 (2011) 809–817.
(ENT1, SLC29A1) facilitates transfer of the antiretroviral drug abacavir across the [129] R. Karahoda, M. Ceckova, F. Staud, The inhibitory effect of antiretroviral drugs on
placenta, Drug Metab. Dispos. 46 (2018) 1817–1826. the L-carnitine uptake in human placenta, Toxicol. Appl. Pharmacol. 368 (2019)
[105] L. Jiraskova, L. Cerveny, S. Karbanova, Z. Ptackova, F. Staud, Expression of 18–25.
concentrative nucleoside transporters (SLC28A) in the human placenta: effects of [130] D. Ahmadimoghaddam, J. Hofman, L. Zemankova, P. Nachtigal, E. Dolezelova,
gestation age and prototype differentiation-affecting agents, Mol. Pharm. 15 L. Cerveny, M. Ceckova, S. Micuda, F. Staud, Synchronized activity of organic
(2018) 2732–2741. cation transporter 3 (Oct3/Slc22a3) and multidrug and toxin extrusion 1 (Mate1/
[106] K. Vahakangas, P. Myllynen, Drug transporters in the human blood-placental Slc47a1) transporter in transplacental passage of MPP+ in rat, Toxicol. Sci. 128
barrier, Br. J. Pharmacol. 158 (2009) 665–678. (2012) 471–481.
[107] F. Staud, M. Ceckova, Regulation of drug transporter expression and function in [131] D. Ahmadimoghaddam, F. Staud, Transfer of metformin across the rat placenta is
the placenta, Expert Opin. Drug Metab. Toxicol. 11 (2015) 533–555. mediated by organic cation transporter 3 (OCT3/SLC22A3) and multidrug and
[108] X.I. Liu, J.D. Momper, N. Rakhmanina, J.N. van den Anker, D.J. Green, G. toxin extrusion 1 (MATE1/SLC47A1) protein, Reprod. Toxicol. 39 (2013) 17–22.
J. Burckart, B.M. Best, M. Mirochnick, E.V. Capparelli, A. Dallmann, [132] B. Ugele, M.V. St-Pierre, M. Pihusch, A. Bahn, P. Hantschmann, Characterization
Physiologically based pharmacokinetic models to predict maternal and identification of steroid sulfate transporters of human placenta, Am. J.
pharmacokinetics and fetal exposure to emtricitabine and acyclovir, J. Clin. Physiol. Endocrinol. Metab. 284 (2003) E390–E398.
Pharmacol. 60 (2020) 240–255. [133] F. Staud, L. Cerveny, D. Ahmadimoghaddam, M. Ceckova, Multidrug and toxin
[109] Z. Zhang, M.Z. Imperial, G.I. Patilea-Vrana, J. Wedagedera, L. Gaohua, J. extrusion proteins (MATE/SLC47); role in pharmacokinetics, Int. J. Biochem. Cell
D. Unadkat, Development of a novel maternal-fetal physiologically based Biol. 45 (2013) 2007–2011.
pharmacokinetic model I: insights into factors that determine fetal drug exposure [134] D. Kojovic, V.W. N, M. Piquette-Miller, Role of elevated SFLT-1 on the regulation
through simulations and sensitivity analyses, Drug Metab. Dispos. 45 (2017) of placental transporters in women with pre-eclampsia, Clin Transl Sci, 13 (2020)
920–938. 580-588.
[110] Z. Zhang, J.D. Unadkat, Development of a novel maternal-fetal physiologically [135] E. Foca, A. Calcagno, A. Bonito, J. Cusato, E. Domenighini, A. D’Avolio, E. Quiros
based pharmacokinetic model II: verification of the model for passive placental Roldan, L. Trentini, F. Castelnuovo, G. Di Perri, F. Castelli, S. Bonora,
permeability drugs, Drug Metab. Dispos. 45 (2017) 939–946. Pharmacokinetic changes during pregnancy according to genetic variants: a
[111] M. Ceckova-Novotna, P. Pavek, F. Staud, P-glycoprotein in the placenta: prospective study in HIV-infected patients receiving atazanavir-ritonavir,
expression, localization, regulation and function, Reprod. Toxicol. 22 (2006) Antimicrob. Agents Chemother. 62 (2018).
400–410. [136] S. Sudhakaran, C.R. Rayner, J. Li, D.C. Kong, N.M. Gude, R.L. Nation, Inhibition
[112] L. Hahnova-Cygalova, M. Ceckova, F. Staud, Fetoprotective activity of breast of placental P-glycoprotein: impact on indinavir transfer to the foetus, Br. J. Clin.
cancer resistance protein (BCRP, ABCG2): expression and function throughout Pharmacol. 65 (2008) 667–673.
pregnancy, Drug Metab. Rev. 43 (2011) 53–68. [137] P.F. Ceccaldi, L. Gavard, L. Mandelbrot, E. Rey, R. Farinotti, J.M. Treluyer, S. Gil,
[113] A.H. Schinkel, J.W. Jonker, Mammalian drug efflux transporters of the ATP Functional role of p-glycoprotein and binding protein effect on the placental
binding cassette (ABC) family: an overview, Adv. Drug Deliv. Rev. 55 (2003) transfer of lopinavir/ritonavir in the ex vivo human perfusion model, Obstet.
3–29. Gynecol. Int. 2009 (2009) 726593.
[114] Z. Neumanova, L. Cerveny, M. Ceckova, F. Staud, Interactions of tenofovir and [138] M. Molsa, T. Heikkinen, J. Hakkola, K. Hakala, O. Wallerman, M. Wadelius,
tenofovir disoproxil fumarate with drug efflux transporters ABCB1, ABCG2, and C. Wadelius, K. Laine, Functional role of P-glycoprotein in the human blood-
ABCC2; role in transport across the placenta, AIDS 28 (2014) 9–17. placental barrier, Clin. Pharmacol. Ther. 78 (2005) 123–131.
[115] Z. Neumanova, L. Cerveny, M. Ceckova, F. Staud, Role of ABCB1, ABCG2, ABCC2 [139] L.W. Han, C. Gao, Q. Mao, An update on expression and function of P-gp/ABCB1
and ABCC5 transporters in placental passage of zidovudine, Biopharm. Drug and BCRP/ABCG2 in the placenta and fetus, Expert Opin. Drug Metab. Toxicol. 14
Dispos. 37 (2016) 28–38. (2018) 817–829.
[116] Z. Neumanova, L. Cerveny, S.L. Greenwood, M. Ceckova, F. Staud, Effect of drug [140] F. Zhou, M. Hong, G. You, Regulation of human organic anion transporter 4 by
efflux transporters on placental transport of antiretroviral agent abacavir, Reprod. progesterone and protein kinase C in human placental BeWo cells, Am. J. Physiol.
Toxicol. 57 (2015) 176–182. Endocrinol. Metab. 293 (2007) E57–E61.
[117] M. Ceckova, J. Reznicek, Z. Ptackova, L. Cerveny, F. Muller, M. Kacerovsky, M. [141] M. Grube, H. Meyer Zu Schwabedissen, K. Draber, D. Prager, K.U. Moritz, K.
F. Fromm, J.D. Glazier, F. Staud, Role of ABC and solute carrier transporters in the Linnemann, C. Fusch, G. Jedlitschky, H.K. Kroemer, Expression, localization, and
placental transport of lamivudine, Antimicrob. Agents Chemother. 60 (2016) function of the carnitine transporter octn2 (slc22a5) in human placenta, Drug
5563–5572. Metab Dispos, 33 (2005) 31-37.
[118] O. Kis, K. Robillard, G.N. Chan, R. Bendayan, The complexities of antiretroviral [142] European AIDS Clinical Society, Guidelines Version 10.0, November 2019. http
drug-drug interactions: role of ABC and SLC transporters, Trends Pharmacol. Sci. s://www.eacsociety.org/files/2019_guidelines-10.0_final.pdf. [accessed 8th
31 (2010) 22–35. January 2021].
[119] H. Fujimoto, M. Higuchi, H. Watanabe, Y. Koh, A.K. Ghosh, H. Mitsuya, [143] F. Fauchet, J.M. Treluyer, L.H. Preta, E. Valade, E. Pannier, S. Urien, D. Hirt,
N. Tanoue, A. Hamada, H. Saito, P-glycoprotein mediates efflux transport of Population pharmacokinetics of abacavir in pregnant women, Antimicrob. Agents
darunavir in human intestinal Caco-2 and ABCB1 gene-transfected renal LLC-PK1 Chemother. 58 (2014) 6287–6289.
cell lines, Biol. Pharm. Bull. 32 (2009) 1588–1593. [144] D. Hirt, S. Urien, E. Rey, E. Arrive, D.K. Ekouevi, P. Coffie, S.K. Leang, S. Lalsab,
[120] Y. Hashiguchi, A. Hamada, T. Shinohara, K. Tsuchiya, H. Jono, H. Saito, Role of P- D. Avit, E. Nerrienet, J. McIntyre, S. Blanche, F. Dabis, J.M. Treluyer, Population
glycoprotein in the efflux of raltegravir from human intestinal cells and CD4+ T- pharmacokinetics of emtricitabine in human immunodeficiency virus type 1-
cells as an interaction target for anti-HIV agents, Biochem. Biophys. Res. infected pregnant women and their neonates, Antimicrob. Agents Chemother. 53
Commun. 439 (2013) 221–227. (2009) 1067–1073.
[121] L.F. Barros, J.C. Bustamante, D.L. Yudilevich, S.M. Jarvis, Adenosine transport [145] A.P. Colbers, D.A. Hawkins, A. Gingelmaier, K. Kabeya, J.K. Rockstroh, C. Wyen,
and nitrobenzylthioinosine binding in human placental membrane vesicles from K. Weizsacker, S.T. Sadiq, J. Ivanovic, C. Giaquinto, G.P. Taylor, J. Molto, D.M.
brush-border and basal sides of the trophoblast, J. Membr. Biol. 119 (1991) Burger, P. network, The pharmacokinetics, safety and efficacy of tenofovir and
151–161. emtricitabine in HIV-1-infected pregnant women, AIDS, 27 (2013) 739-748.
[122] L.F. Barros, D.L. Yudilevich, S.M. Jarvis, N. Beaumont, J.D. Young, S.A. Baldwin, [146] R.A.C. Siemieniuk, L. Lytvyn, J. Mah Ming, R.M. Mullen, F. Anam, T. Otieno, G.
Immunolocalisation of nucleoside transporters in human placental trophoblast H. Guyatt, G.P. Taylor, C. Beltran-Arroyave, P.M. Okwen, R. Nduati, J. Kinuthia,
and endothelial cells: evidence for multiple transporter isoforms, Pflugers Arch. H.N. Luma, H. Kirpalani, A. Merglen, O.A. Lesi, P.O. Vandvik, T. Agoritsas,
429 (1995) 394–399. S. Bewley, Antiretroviral therapy in pregnant women living with HIV: a clinical
[123] R. Govindarajan, A.H. Bakken, K.L. Hudkins, Y. Lai, F.J. Casado, M. Pastor- practice guideline, BMJ 358 (2017) j3961.
Anglada, C.M. Tse, J. Hayashi, J.D. Unadkat, In situ hybridization and [147] A. Colbers, J. Molto, J. Ivanovic, K. Kabeya, D. Hawkins, A. Gingelmaier,
immunolocalization of concentrative and equilibrative nucleoside transporters in G. Taylor, K. Weizsacker, S.T. Sadiq, M. Van der Ende, C. Giaquinto, D. Burger,

12
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

P. Network, Pharmacokinetics of total and unbound darunavir in HIV-1-infected Effects of in utero antiretroviral exposure on mitochondrial DNA levels,
pregnant women, J. Antimicrob. Chemother. 70 (2015) 534–542. mitochondrial function and oxidative stress, HIV Med. 13 (2012) 98–106.
[148] World Health Organization, WHO recommends dolutegravir as preferred HIV [167] B. Barret, M. Tardieu, P. Rustin, C. Lacroix, B. Chabrol, I. Desguerre, C. Dollfus, M.
treatment option in all populations, https://www.who.int/news/item/22-07-2 J. Mayaux, S. Blanche, G. French perinatal cohort study, persistent mitochondrial
019-who-recommends-dolutegravir-as-preferred-hiv-treatment-option-in-all-po dysfunction in HIV-1-exposed but uninfected infants: clinical screening in a large
pulations, accessed 7th January 2021. prospective cohort, AIDS 17 (2003) 1769–1785.
[149] E.C. Joao, R.L. Morrison, D.E. Shapiro, N. Chakhtoura, M.I.S. Gouvea, B.T.M. de [168] S. Blanche, M. Tardieu, P. Rustin, A. Slama, B. Barret, G. Firtion, N. Ciraru-
Lourdes, T.L. Fuller, B.T. Mmbaga, J.S. Ngocho, B.N. Njau, A. Violari, R. Mathiba, Vigneron, C. Lacroix, C. Rouzioux, L. Mandelbrot, I. Desguerre, A. Rotig, M.
Z. Essack, J.H.S. Pilotto, L.F. Moreira, M.J. Rolon, P. Cahn, S. Prommas, T.R. J. Mayaux, J.F. Delfraissy, Persistent mitochondrial dysfunction and perinatal
Cressey, K. Chokephaibulkit, P. Werarak, L. Laimon, R. Hennessy, L.M. Frenkel, P. exposure to antiretroviral nucleoside analogues, Lancet 354 (1999) 1084–1089.
Anthony, B.M. Best, G.K. Siberry, M. Mirochnick, Raltegravir versus efavirenz in [169] E.H. Decloedt, B. Rosenkranz, G. Maartens, J. Joska, Central nervous system
antiretroviral-naive pregnant women living with HIV (NICHD P1081): an open- penetration of antiretroviral drugs: pharmacokinetic, pharmacodynamic and
label, randomised, controlled, phase 4 trial, Lancet HIV, 7 (2020) e322-e331. pharmacogenomic considerations, Clin. Pharmacokinet. 54 (2015) 581–598.
[150] O. Osiyemi, S. Yasin, C. Zorrilla, C. Bicer, V. Hillewaert, K. Brown, H.M. Crauwels, [170] A. Venerosi, A. Valanzano, E. Alleva, G. Calamandrei, Prenatal exposure to anti-
Pharmacokinetics, antiviral activity, and safety of rilpivirine in pregnant women HIV drugs: neurobehavioral effects of zidovudine (AZT) + lamivudine (3TC)
with HIV-1 infection: results of a phase 3b, multicenter, open-label study, Infect. treatment in mice, Teratology 63 (2001) 26–37.
Dis. Ther. 7 (2018) 147–159. [171] E. Moseholm, M. Helleberg, H. Sandholdt, T.L. Katzenstein, M. Storgaard,
[151] S. Schalkwijk, A. Colbers, D. Konopnicki, A. Gingelmaier, J. Lambert, M. van der G. Pedersen, I.S. Johansen, N. Weis, Children exposed or unexposed to human
Ende, J. Molto, D. Burger, H.I.V.i.p.w.N. Pharmacokinetics of newly developed immunodeficiency virus: weight, height, and body mass index during the first 5
antiretroviral agents in, Lowered rilpivirine exposure during the third trimester of years of life-a Danish nationwide cohort, Clin. Infect. Dis. 70 (2020) 2168–2177.
pregnancy in human immunodeficiency virus type 1-infected women, Clin Infect [172] J. Jao, D.L. Jacobson, W. Yu, W. Borkowsky, M.E. Geffner, E.J. McFarland, K.
Dis, 65 (2017) 1335-1341. Patel, P.L. Williams, T. Miller, H.I.V.A.C.S. Pediatric, A comparison of metabolic
[152] R. Zash, L. Holmes, M. Diseko, D.L. Jacobson, S. Brummel, G. Mayondi, outcomes between obese HIV-exposed uninfected youth from the PHACS
A. Isaacson, S. Davey, J. Mabuta, M. Mmalane, T. Gaolathe, M. Essex, S. Lockman, SMARTT study and HIV-unexposed youth from the NHANES study in the United
J. Makhema, R.L. Shapiro, Neural-tube defects and antiretroviral treatment States, J Acquir Immune Defic Syndr, 81 (2019) 319–327.
regimens in Botswana, N. Engl. J. Med. 381 (2019) 827–840. [173] V. Guerra, E.C. Leister, P.L. Williams, T.J. Starc, S.E. Lipshultz, J.D. Wilkinson, R.
[153] R. Zash, J. Makhema, R.L. Shapiro, Neural-tube defects with Dolutegravir B. Van Dyke, R. Hazra, S.D. Colan, Long-term effects of in utero antiretroviral
treatment from the time of conception, N. Engl. J. Med. 379 (2018) 979–981. exposure: systolic and diastolic function in HIV-exposed uninfected youth, AIDS
[154] K.M. Brooks, J.D. Momper, M. Pinilla, A.M. Stek, E. Barr, A. Weinberg, J. Res. Hum. Retrovir. 32 (2016) 621–627.
G. Deville, I.L. Febo, M. Cielo, K. George, K. Denson, K. Rungruengthanakit, D. [174] O.A. Olivero, J.J. Fernandez, B.B. Antiochos, J.L. Wagner, M.E. St Claire, M.
E. Shapiro, E. Smith, N. Chakhtoura, J.F. Rooney, R. Haubrich, E. Rowena, E. C. Poirier, Transplacental genotoxicity of combined antiretroviral nucleoside
V. Capparelli, M. Mirochnick, B.M. Best, I.P.P. Team, Pharmacokinetics of analogue therapy in Erythrocebus patas monkeys, J. Acquir. Immune Defic.
tenofovir alafenamide with and without cobicistat in pregnant and postpartum Syndr. 29 (2002) 323–329.
women living with HIV: results from IMPAACT P1026s, AIDS 35 (2021) 407–417. [175] K.L. Witt, C.K. Cunningham, K.B. Patterson, G.E. Kissling, S.D. Dertinger,
[155] A. Colbers, B. Best, S. Schalkwijk, J. Wang, A. Stek, C. Hidalgo Tenorio, D. E. Livingston, J.B. Bishop, Elevated frequencies of micronucleated erythrocytes in
Hawkins, G. Taylor, R. Kreitchmann, S. Burchett, A. Haberl, K. Kabeya, M. van infants exposed to zidovudine in utero and postpartum to prevent mother-to-child
Kasteren, E. Smith, E. Capparelli, D. Burger, M. Mirochnick, P. Network, I.S.T. transmission of HIV, Environ. Mol. Mutagen. 48 (2007) 322–329.
the, Maraviroc Pharmacokinetics in HIV-1-Infected Pregnant Women, Clin Infect [176] W. Ivy 3rd, S.R. Nesheim, S.M. Paul, A.R. Ibrahim, M. Chan, X. Niu, M.A. Lampe,
Dis, 61 (2015) 1582-1589. Cancer among children with perinatal exposure to HIV and antiretroviral
[156] V. Vannappagari, J.D. Albano, N. Koram, H. Tilson, A.E. Scheuerle, M.D. Napier, medications-New Jersey, 1995–2010, J. Acquir. Immune Defic. Syndr. 70 (2015)
Prenatal exposure to zidovudine and risk for ventricular septal defects and 62–66.
congenital heart defects: data from the antiretroviral pregnancy registry, Eur. J. [177] F. Staud, R. Karahoda, Trophoblast: the central unit of fetal growth, protection
Obstet. Gynecol. Reprod. Biol. 197 (2016) 6–10. and programming, Int. J. Biochem. Cell Biol. 105 (2018) 35–40.
[157] P.L. Williams, M.J. Crain, C. Yildirim, R. Hazra, R.B. Van Dyke, K. Rich, J.S. Read, [178] H.L.B. Dos Reis, N.A.T. Boldrini, A.F.R. Rangel, V.F. Barros, P.R. Mercon de
E. Stuard, M. Rathore, H.A. Mendez, D.H. Watts, H.I.V.A.C.S. Pediatric, Vargas, A.E. Miranda, Placental growth disorders and perinatal adverse outcomes
Congenital anomalies and in utero antiretroviral exposure in human in Brazilian HIV-infected pregnant women, PLoS One 15 (2020), e0231938.
immunodeficiency virus-exposed uninfected infants, JAMA Pediatr, 169 (2015) [179] E. Kalk, P. Schubert, J.A. Bettinger, M.F. Cotton, M. Esser, A. Slogrove, C.
48-55. A. Wright, Placental pathology in HIV infection at term: a comparison with HIV-
[158] J. Sibiude, J. Le Chenadec, D. Bonnet, R. Tubiana, A. Faye, C. Dollfus, L. uninfected women, Tropical Med. Int. Health 22 (2017) 604–613.
Mandelbrot, S. Delmas, N. Lelong, B. Khoshnood, J. Warszawski, S. Blanche, A. [180] M.M. Obimbo, Y. Zhou, M.T. McMaster, C.R. Cohen, Z. Qureshi, J. Ong’ech, J.
French National Agency for Research on, T. Viral Hepatitis French Perinatal A. Ogeng’o, S.J. Fisher, Placental structure in preterm birth among HIV-positive
Cohort/Protease Inhibitor Monotherapy Evaluation, In utero exposure to versus HIV-negative women in Kenya, J. Acquir. Immune Defic. Syndr. 80 (2019)
zidovudine and heart anomalies in the ANRS French perinatal cohort and the 94–102.
nested PRIMEVA randomized trial, Clin Infect Dis, 61 (2015) 270-280. [181] L. Cerveny, Z. Neumanova, S. Karbanova, I. Havlova, F. Staud, Long-term
[159] N. Ford, L. Mofenson, Z. Shubber, A. Calmy, I. Andrieux-Meyer, M. Vitoria, administration of tenofovir or emtricitabine to pregnant rats; effect on Abcb1a,
N. Shaffer, F. Renaud, Safety of efavirenz in the first trimester of pregnancy: an Abcb1b and Abcg2 expression in the placenta and in maternal and fetal organs,
updated systematic review and meta-analysis, AIDS 28 (Suppl. 2) (2014) J. Pharm. Pharmacol. 68 (2016) 84–92.
S123–S131. [182] P.H. Gois, D. Canale, W.M. Luchi, R.A. Volpini, M.M. Veras, S. Costa Nde, M.
[160] J.G. Schnoll, B. Temsamrit, D. Zhang, H. Song, G.L. Ming, K.M. Christian, H. Shimizu, A.C. Seguro, Tenofovir during pregnancy in rats: a novel pathway for
Evaluating neurodevelopmental consequences of perinatal exposure to programmed hypertension in the offspring, J. Antimicrob. Chemother. 70 (2015)
antiretroviral drugs: current challenges and new approaches, J. NeuroImmune 1094–1105.
Pharmacol. 16 (2021) 113–129. [183] S. Kala, C. Dunk, S. Acosta, L. Serghides, Periconceptional exposure to lopinavir,
[161] C.J. Wedderburn, C. Evans, S. Yeung, D.M. Gibb, K.A. Donald, A.J. Prendergast, but not darunavir, impairs decidualization: a potential mechanism leading to poor
Growth and neurodevelopment of HIV-exposed uninfected children: a conceptual birth outcomes in HIV-positive pregnancies, Hum. Reprod. 35 (2020) 1781–1796.
framework, Curr. HIV/AIDS Rep. 16 (2019) 501–513. [184] D.A. Dalwadi, S. Kim, S.M. Amdani, Z. Chen, R.Q. Huang, J.A. Schetz, Molecular
[162] P.L. Williams, C. Yildirim, E.G. Chadwick, R.B. Van Dyke, R. Smith, K.F. Correia, mechanisms of serotonergic action of the HIV-1 antiretroviral efavirenz,
A. DiPerna, G.R. Seage, 3rd, R. Hazra, C.S. Crowell, A.R.T.T.s.o.t.P.H.I.V.A.C.S. Pharmacol. Res. 110 (2016) 10–24.
Surveillance Monitoring for, Association of maternal antiretroviral use with [185] R. Karahoda, H. Horackova, P. Kastner, A. Matthios, L. Cerveny, R. Kucera,
microcephaly in children who are HIV-exposed but uninfected (SMARTT): a M. Kacerovsky, J. Duintjer Tebbens, A. Bonnin, C. Abad, F. Staud, Serotonin
prospective cohort study, Lancet HIV, 7 (2020) e49-e58. homeostasis in the materno-foetal interface at term: role of transporters (SERT/
[163] K. Phiri, S. Hernandez-Diaz, K.B. Dugan, P.L. Williams, J.A. Dudley, A. Jules, S. SLC6A4 and OCT3/SLC22A3) and monoamine oxidase A (MAO-A) in uptake and
T. Callahan, G.R. Seage 3rd, W.O. Cooper, First trimester exposure to degradation of serotonin by human and rat term placenta, Acta Physiol (Oxford)
antiretroviral therapy and risk of birth defects, Pediatr. Infect. Dis. J. 33 (2014) 229 (2020), e13478.
741–746. [186] L. Ricceri, A. Venerosi, A. Valanzano, A. Sorace, E. Alleva, Prenatal AZT or 3TC
[164] L. van de Wijer, L.P. Garcia, S.I. Hanswijk, J. Rando, A. Middelman, R. Ter Heine, and mouse development of locomotor activity and hot-plate responding upon
Q. Mast, G.J.M. Martens, A. van der Ven, S.M. Kolk, A.F.A. Schellekens, J. administration of the GABA(A) receptor agonist muscimol, Psychopharmacology
R. Homberg, Neurodevelopmental and behavioral consequences of perinatal 153 (2001) 434–442.
exposure to the HIV drug efavirenz in a rodent model, Transl. Psychiatry 9 (2019) [187] S.A. Angione, S.M. Cherian, A.E. Ozdener, A review of the efficacy and safety of
84. Genvoya(R) (elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide) in
[165] A.R. Cassidy, P.L. Williams, J. Leidner, G. Mayondi, G. Ajibola, J. Makhema, P. the management of HIV-1 infection, J. Pharm. Pract. 31 (2018) 216–221.
A. Holding, K.M. Powis, O. Batlang, C. Petlo, R. Shapiro, B. Kammerer, [188] J.R. Arribas, M. Thompson, P.E. Sax, B. Haas, C. McDonald, D.A. Wohl,
S. Lockman, In utero efavirenz exposure and neurodevelopmental outcomes in E. DeJesus, A.E. Clarke, S. Guo, H. Wang, C. Callebaut, A. Plummer, A. Cheng,
HIV-exposed uninfected children in Botswana, Pediatr. Infect. Dis. J. 38 (2019) M. Das, S. McCallister, Brief report: randomized, double-blind comparison of
828–834. tenofovir alafenamide (TAF) vs tenofovir disoproxil fumarate (TDF), each
[166] A.C. Ross, T. Leong, A. Avery, M. Castillo-Duran, H. Bonilla, D. Lebrecht, U. coformulated with elvitegravir, cobicistat, and emtricitabine (E/C/F) for initial
A. Walker, N. Storer, D. Labbato, A. Khaitan, I. Tomanova-Soltys, G.A. McComsey,

13
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

HIV-1 treatment: week 144 results, J. Acquir. Immune Defic. Syndr. 75 (2017) [208] J. Reznicek, M. Ceckova, L. Cerveny, F. Muller, F. Staud, Emtricitabine is a
211–218. substrate of MATE1 but not of OCT1, OCT2, P-gp, BCRP or MRP2 transporters,
[189] S. Benaboud, J.M. Treluyer, S. Urien, S. Blanche, N. Bouazza, H. Chappuy, E. Rey, Xenobiotica 47 (2017) 77–85.
E. Pannier, G. Firtion, O. Launay, D. Hirt, Pregnancy-related effects on lamivudine [209] B.H. Rimawi, E. Johnson, A. Rajakumar, S. Tao, Y. Jiang, S. Gillespie, R.
pharmacokinetics in a population study with 228 women, Antimicrob. Agents F. Schinazi, M. Mirochnick, M.L. Badell, R. Chakraborty, Pharmacokinetics and
Chemother. 56 (2012) 776–782. placental transfer of llvitegravir, dolutegravir, and other antiretrovirals during
[190] C. Waitt, C. Orrell, S. Walimbwa, Y. Singh, K. Kintu, B. Simmons, J. Kaboggoza, pregnancy, Antimicrob. Agents Chemother. 61 (2017).
M. Sihlangu, J.A. Coombs, T. Malaba, J. Byamugisha, A. Amara, J. Gini, L. Else, [210] L. Mandelbrot, G. Peytavin, G. Firtion, R. Farinotti, Maternal-fetal transfer and
C. Heiburg, E.M. Hodel, H. Reynolds, U. Mehta, P. Byakika-Kibwika, A. Hill, amniotic fluid accumulation of lamivudine in human immunodeficiency virus-
L. Myer, M. Lamorde, S. Khoo, Safety and pharmacokinetics of dolutegravir in infected pregnant women, Am. J. Obstet. Gynecol. 184 (2001) 153–158.
pregnant mothers with HIV infection and their neonates: a randomised trial [211] R.F. Yeh, N.L. Rezk, A.D. Kashuba, J.B. Dumond, H.L. Tappouni, H.C. Tien, Y.
(DolPHIN-1 study), PLoS Med. 16 (2019), e1002895. C. Chen, M. Vourvahis, A.L. Horton, S.A. Fiscus, K.B. Patterson, Genital tract, cord
[191] J. Gallant, A. Lazzarin, A. Mills, C. Orkin, D. Podzamczer, P. Tebas, P.M. Girard, blood, and amniotic fluid exposures of seven antiretroviral drugs during and after
I. Brar, E.S. Daar, D. Wohl, J. Rockstroh, X. Wei, J. Custodio, K. White, H. Martin, pregnancy in human immunodeficiency virus type 1-infected women,
A. Cheng, E. Quirk, Bictegravir, emtricitabine, and tenofovir alafenamide versus Antimicrob. Agents Chemother. 53 (2009) 2367–2374.
dolutegravir, abacavir, and lamivudine for initial treatment of HIV-1 infection [212] S.A. McCormack, B.M. Best, Protecting the fetus against HIV infection: a
(GS-US-380-1489): a double-blind, multicentre, phase 3, randomised controlled systematic review of placental transfer of antiretrovirals, Clin. Pharmacokinet. 53
non-inferiority trial, Lancet 390 (2017) 2063–2072. (2014) 989–1004.
[192] P.E. Sax, A. Pozniak, M.L. Montes, E. Koenig, E. DeJesus, H.J. Stellbrink, [213] M. Takeda, S. Khamdang, S. Narikawa, H. Kimura, Y. Kobayashi, T. Yamamoto, S.
A. Antinori, K. Workowski, J. Slim, J. Reynes, W. Garner, J. Custodio, K. White, H. Cha, T. Sekine, H. Endou, Human organic anion transporters and human
D. SenGupta, A. Cheng, E. Quirk, Coformulated bictegravir, emtricitabine, and organic cation transporters mediate renal antiviral transport, J. Pharmacol. Exp.
tenofovir alafenamide versus dolutegravir with emtricitabine and tenofovir Ther. 300 (2002) 918–924.
alafenamide, for initial treatment of HIV-1 infection (GS-US-380-1490): a [214] P. Cano-Soldado, I.M. Lorrayoz, M. Molina-Arcas, F.J. Casado, J. Martinez-Picado,
randomised, double-blind, multicentre, phase 3, non-inferiority trial, Lancet 390 M.P. Lostao, M. Pastor-Anglada, Interaction of nucleoside inhibitors of HIV-1
(2017) 2073–2082. reverse transcriptase with the concentrative nucleoside transporter-1 (SLC28A1),
[193] T. Farrow, C. Deaton, N. Nguyen, M. Serejo, D. Muramoto, A. van Troostenburg, Antivir. Ther. 9 (2005) 993–1002.
L. Ng, L. Liu, H. Martin, M. Das, Cumulative safety review of elvitegravir and [215] C.H. Storch, D. Theile, H. Lindenmaier, W.E. Haefeli, J. Weiss, Comparison of the
bictegravir use during pregnancy and risk of neural tube defects P030. HIV inhibitory activity of anti-HIV drugs on P-glycoprotein, Biochem. Pharmacol. 73
Glasgow 2018, 28-31 October 2018, Glasgow, UK., J Int AIDS Soc, 21 Suppl 8 (2007) 1573–1581.
(2018) e25187. [216] J. Weiss, J. Rose, C.H. Storch, N. Ketabi-Kiyanvash, A. Sauer, W.E. Haefeli,
[194] E.M. Gardner, M.P. McLees, J.F. Steiner, C. Del Rio, W.J. Burman, The spectrum T. Efferth, Modulation of human BCRP (ABCG2) activity by anti-HIV drugs,
of engagement in HIV care and its relevance to test-and-treat strategies for J. Antimicrob. Chemother. 59 (2007) 238–245.
prevention of HIV infection, Clin. Infect. Dis. 52 (2011) 793–800. [217] T.R. Cressey, A. Stek, E. Capparelli, C. Bowonwatanuwong, S. Prommas,
[195] K.A. Powers, E. Samoff, M.A. Weaver, L.A. Sampson, W.C. Miller, P.A. Leone, P. Sirivatanapa, P. Yuthavisuthi, C. Neungton, Y. Huo, E. Smith, B.M. Best,
H. Swygard, Longitudinal HIV care trajectories in North Carolina, J. Acquir. M. Mirochnick, I.P. Team, Efavirenz pharmacokinetics during the third trimester
Immune Defic. Syndr. 74 (Suppl. 2) (2017) S88–S95. of pregnancy and postpartum, J. Acquir. Immune Defic. Syndr. 59 (2012)
[196] A.I. Rana, J.R. Castillo-Mancilla, K.T. Tashima, R.L. Landovitz, Advances in long- 245–252.
acting agents for the treatment of HIV infection, Drugs 80 (2020) 535–545. [218] M. Ceckova, J. Reznicek, B. Deutsch, M.F. Fromm, F. Staud, Efavirenz reduces
[197] J.M. Simoni, K. Beima-Sofie, Z.H. Mohamed, J. Christodoulou, K. Tapia, S. renal excretion of lamivudine in rats by inhibiting organic cation transporters
M. Graham, R. Ho, A.C. Collier, Long-acting injectable antiretroviral treatment (OCT, Oct) and multidrug and toxin extrusion proteins (MATE, Mate), PLoS One
acceptability and preferences: a qualitative study among US providers, adults 13 (2018), e0202706.
living with HIV, and parents of youth living with HIV, AIDS Patient Care STDs 33 [219] A. Colbers, A. Gingelmaier, M. van der Ende, B. Rijnders, D. Burger,
(2019) 104–111. Pharmacokinetics, safety and transplacental passage of rilpivirine in pregnancy:
[198] E.D. Weld, M.S. Rana, R.H. Dallas, A.F. Camacho-Gonzalez, P. Ryscavage, A. two cases, AIDS 28 (2014) 288–290.
H. Gaur, R. Chakraborty, S. Swindells, C. Flexner, A.L. Agwu, Interest of youth [220] J. Reznicek, M. Ceckova, Z. Ptackova, O. Martinec, L. Tupova, L. Cerveny,
living with HIV in long-acting antiretrovirals, J. Acquir. Immune Defic. Syndr. 80 F. Staud, MDR1 and BCRP transporter-mediated drug-drug interaction between
(2019) 190–197. rilpivirine and abacavir and effect on intestinal absorption, Antimicrob. Agents
[199] D.A. Margolis, J. Gonzalez-Garcia, H.J. Stellbrink, J.J. Eron, Y. Yazdanpanah, D. Chemother. 61 (2017).
Podzamczer, T. Lutz, J.B. Angel, G.J. Richmond, B. Clotet, F. Gutierrez, L. Sloan, [221] W.F. Bierman, G.L. Scheffer, A. Schoonderwoerd, G. Jansen, M.A. van Agtmael, S.
M.S. Clair, M. Murray, S.L. Ford, J. Mrus, P. Patel, H. Crauwels, S.K. Griffith, K.C. A. Danner, R.J. Scheper, Protease inhibitors atazanavir, lopinavir and ritonavir
Sutton, D. Dorey, K.Y. Smith, P.E. Williams, W.R. Spreen, Long-acting are potent blockers, but poor substrates, of ABC transporters in a broad panel of
intramuscular cabotegravir and rilpivirine in adults with HIV-1 infection (LATTE- ABC transporter-overexpressing cell lines, J. Antimicrob. Chemother. 65 (2010)
2): 96-week results of a randomised, open-label, phase 2b, non-inferiority trial, 1672–1680.
Lancet, 390 (2017) 1499–1510. [222] F. Conradie, C. Zorrilla, D. Josipovic, M. Botes, O. Osiyemi, E. Vandeloise, T. Eley,
[200] G. van ’t Klooster, E. Hoeben, H. Borghys, A. Looszova, M.P. Bouche, F. van M. Child, R. Bertz, W. Hu, V. Wirtz, D. McGrath, Safety and exposure of once-daily
Velsen, L. Baert, Pharmacokinetics and disposition of rilpivirine (TMC278) ritonavir-boosted atazanavir in HIV-infected pregnant women, HIV Med. 12
nanosuspension as a long-acting injectable antiretroviral formulation, Antimicrob (2011) 570–579.
Agents Chemother, 54 (2010) 2042-2050. [223] J. Ivanovic, E. Nicastri, M.M. Anceschi, P. Ascenzi, F. Signore, G. Pisani, C.
[201] V. Sheikh, J.S. Murray, A. Sherwat, Ibalizumab in multidrug-resistant HIV - Vallone, E. Mattia, S. Notari, M. Tempestilli, L.P. Pucillo, P. Narciso, Pregnancy,
accepting uncertainty, N. Engl. J. Med. 379 (2018) 605–607. H.I.V.I. Newborn Clinical Outcome Group in, Transplacental transfer of
[202] B. Emu, J. Fessel, S. Schrader, P. Kumar, G. Richmond, S. Win, S. Weinheimer, antiretroviral drugs and newborn birth weight in HIV-infected pregnant women,
C. Marsolais, S. Lewis, Phase 3 study of ibalizumab for multidrug-resistant HIV-1, Curr HIV Res, 7 (2009) 620–625.
N. Engl. J. Med. 379 (2018) 645–654. [224] L. Mandelbrot, F. Mazy, C. Floch-Tudal, F. Meier, E. Azria, C. Crenn-Hebert, J.
[203] L. Fairlie, C. Waitt, S. Lockman, M. Moorhouse, E.J. Abrams, P. Clayden, M. Treluyer, E. Herinomenzanahary, C. Ferreira, G. Peytavin, Atazanavir in
M. Boffito, S. Khoo, H. Rees, A. Cournil, W.F. Venter, C. Serenata, M. Chersich, pregnancy: impact on neonatal hyperbilirubinemia, Eur. J. Obstet. Gynecol.
Inclusion of pregnant women in antiretroviral drug research: what is needed to Reprod. Biol. 157 (2011) 18–21.
move forwards? J. Int. AIDS Soc. 22 (2019), e25372. [225] L. Mandelbrot, D. Duro, E. Belissa, G. Peytavin, Placental transfer of darunavir in
[204] H. Duan, T. Hu, R.S. Foti, Y. Pan, P.W. Swaan, J. Wang, Potent and selective an ex vivo human cotyledon perfusion model, Antimicrob. Agents Chemother. 58
inhibition of plasma membrane monoamine transporter by HIV protease (2014) 5617–5620.
inhibitors, Drug Metab. Dispos. 43 (2015) 1773–1780. [226] C.D. Zorrilla, R. Wright, O.O. Osiyemi, S. Yasin, B. Baugh, K. Brown, B. Coate,
[205] H. Chappuy, J.M. Treluyer, V. Jullien, J. Dimet, E. Rey, M. Fouche, G. Firtion, P. Verboven, J. Mrus, R. Falcon, T.N. Kakuda, Total and unbound darunavir
G. Pons, L. Mandelbrot, Maternal-fetal transfer and amniotic fluid accumulation pharmacokinetics in pregnant women infected with HIV-1: results of a study of
of nucleoside analogue reverse transcriptase inhibitors in human darunavir/ritonavir 600/100 mg administered twice daily, HIV Med. 15 (2014)
immunodeficiency virus-infected pregnant women, Antimicrob. Agents 50–56.
Chemother. 48 (2004) 4332–4336. [227] N. Mulligan, B.M. Best, J. Wang, E.V. Capparelli, A. Stek, E. Barr, S.L. Buschur, E.
[206] G. Minuesa, C. Volk, M. Molina-Arcas, V. Gorboulev, I. Erkizia, P. Arndt, B. Clotet, P. Acosta, E. Smith, N. Chakhtoura, S. Burchett, M. Mirochnick, I.P.P. Team,
M. Pastor-Anglada, H. Koepsell, J. Martinez-Picado, Transport of lamivudine Dolutegravir pharmacokinetics in pregnant and postpartum women living with
[(-)-beta-L-2′ ,3′ -dideoxy-3′ -thiacytidine] and high-affinity interaction of HIV, AIDS 32 (2018) 729–737.
nucleoside reverse transcriptase inhibitors with human organic cation [228] M.J. Reese, P.M. Savina, G.T. Generaux, H. Tracey, J.E. Humphreys, E. Kanaoka,
transporters 1, 2, and 3, J. Pharmacol. Exp. Ther. 329 (2009) 252–261. L.O. Webster, K.A. Harmon, J.D. Clarke, J.W. Polli, In vitro investigations into the
[207] A. Calcagno, L. Trentini, L. Marinaro, C. Montrucchio, A. D’Avolio, V. Ghisetti, roles of drug transporters and metabolizing enzymes in the disposition and drug
G. Di Perri, S. Bonora, Transplacental passage of etravirine and maraviroc in a interactions of dolutegravir, a HIV integrase inhibitor, Drug Metab. Dispos. 41
multidrug-experienced HIV-infected woman failing on darunavir-based HAART in (2013) 353–361.
late pregnancy, J. Antimicrob. Chemother. 68 (2013) 1938–1939. [229] G. Minuesa, C. Arimany-Nardi, I. Erkizia, S. Cedeno, J. Molto, B. Clotet, M. Pastor-
Anglada, J. Martinez-Picado, P-glycoprotein (ABCB1) activity decreases

14
L. Cerveny et al. BBA - Molecular Basis of Disease 1867 (2021) 166206

raltegravir disposition in primary CD4+P-gphigh cells and correlates with HIV-1 [235] R. Begley, M. Das, L. Zhong, J. Ling, B.P. Kearney, J.M. Custodio,
viral load, J. Antimicrob. Chemother. 71 (2016) 2782–2792. Pharmacokinetics of tenofovir alafenamide when coadministered with other HIV
[230] M.T. Hoque, O. Kis, M.F. De Rosa, R. Bendayan, Raltegravir permeability across antiretrovirals, J. Acquir. Immune Defic. Syndr. 78 (2018) 465–472.
blood-tissue barriers and the potential role of drug efflux transporters, [236] J. Reznicek, M. Ceckova, L. Tupova, F. Staud, Etravirine inhibits ABCG2 drug
Antimicrob. Agents Chemother. 59 (2015) 2572–2582. transporter and affects transplacental passage of tenofovir disoproxil fumarate,
[231] M.I. Blonk, A.P. Colbers, C. Hidalgo-Tenorio, K. Kabeya, K. Weizsacker, A.E. Placenta 47 (2016) 124–129.
Haberl, J. Molto, D.A. Hawkins, M.E. van der Ende, A. Gingelmaier, G.P. Taylor, [237] N.C. Zembruski, W.E. Haefeli, J. Weiss, Interaction potential of etravirine with
J. Ivanovic, C. Giaquinto, D.M. Burger, H.I.V.I.P.W.P.N. Pharmacokinetics of drug transporters assessed in vitro, Antimicrob. Agents Chemother. 55 (2011)
Newly Developed Antiretroviral Agents in, P. Network, Raltegravir in HIV-1- 1282–1284.
Infected Pregnant Women: Pharmacokinetics, Safety, and Efficacy, Clin Infect Dis, [238] T.N. Kakuda, M. Scholler-Gyure, R.M. Hoetelmans, Pharmacokinetic interactions
61 (2015) 809–816. between etravirine and non-antiretroviral drugs, Clin. Pharmacokinet. 50 (2011)
[232] D.H. Watts, A. Stek, B.M. Best, J. Wang, E.V. Capparelli, T.R. Cressey, F. Aweeka, 25–39.
P. Lizak, R. Kreitchmann, S.K. Burchett, D.E. Shapiro, E. Hawkins, E. Smith, M. [239] M. Scholler-Gyure, T.N. Kakuda, A. Raoof, G. De Smedt, R.M. Hoetelmans,
Mirochnick, I.s.s. team, Raltegravir pharmacokinetics during pregnancy, J Acquir Clinical pharmacokinetics and pharmacodynamics of etravirine, Clin.
Immune Defic Syndr, 67 (2014) 375-381. Pharmacokinet. 48 (2009) 561–574.
[233] D.F. Clarke, E.P. Acosta, M.L. Rizk, Y.J. Bryson, S.A. Spector, L.M. Mofenson, E. [240] M. Ramgopal, O. Osiyemi, C. Zorrilla, H.M. Crauwels, R. Ryan, K. Brown,
Handelsman, H. Teppler, C. Welebob, D. Persaud, M.P. Cababasay, J. Wang, M. V. Hillewaert, B. Baugh, Pharmacokinetics of total and unbound etravirine in
Mirochnick, A.C.T.P.S.T. International Maternal Pediatric Adolescent, Raltegravir HIV-1-infected pregnant women, J. Acquir. Immune Defic. Syndr. 73 (2016)
pharmacokinetics in neonates following maternal dosing, J Acquir Immune Defic 268–274.
Syndr, 67 (2014) 310-315. [241] L. Tupova, M. Ceckova, C. Ambrus, A. Sorf, Z. Ptackova, Z. Gaborik, F. Staud,
[234] M.L. Rizk, R. Houle, G.H. Chan, M. Hafey, E.G. Rhee, X. Chu, Raltegravir has a Interactions between maraviroc and the ABCB1, ABCG2, and ABCC2 transporters:
low propensity to cause clinical drug interactions through inhibition of major an important role in transplacental pharmacokinetics, Drug Metab. Dispos. 47
drug transporters: an in vitro evaluation, Antimicrob. Agents Chemother. 58 (2019) 954–960.
(2014) 1294–1301. [242] T. Nabekura, T. Kawasaki, Y. Kamiya, Y. Uwai, Effects of antiviral drugs on
organic anion transport in human placental BeWo cells, Antimicrob. Agents
Chemother. 59 (2015) 7666–7670.

15

You might also like