Vous êtes sur la page 1sur 22

Review

For reprint orders, please contact: reprints@futuremedicine.com

Potential effect of pharmacogenetics on maternal,


fetal and infant antiretroviral drug exposure during
pregnancy and breastfeeding
Mother-to-child-transmission rates of HIV in the absence of any intervention range between 20 and 45%.
However, the provision of antiretroviral drugs (ARVs) during pregnancy, delivery and breastfeeding can
reduce HIV transmission to less than 2%. Physiological changes during pregnancy can influence ARV
disposition. Associations between SNPs in genes coding for metabolizing enzymes, and/or transporters,
and ARVs disposition are well described; however, relatively little is known about the influence of these
SNPs on ARV pharmacokinetics during pregnancy and lactation as well as their effect on distribution into
the fetal compartment and breast milk excretion. Differences in maternal, fetal and infant ARV exposure
due to SNPs may affect the efficacy and safety of ARVs used to prevent mother-to-child-transmission. The
aim of this review is to provide an update on the effect of pregnancy-induced changes on the
pharmacokinetics of ARVs and highlight the potential role of pharmacogenetics.

KEYWORDS: antiretrovirals n drug transporters n highly active antiretroviral therapy Adeniyi Olagunju1,2,
n HIV n pregnancy n prevention of mother-to-child transmission
Andrew Owen2
& Tim R Cressey*3,4,5
Mother-to-child-transmission (MTCT) rates of nonbreastfeeding infants [5] . Moreover, maternal 1
Faculty of Pharmacy, Obafemi
HIV without any intervention range between 20 HAART or daily infant NVP can reduce HIV-1 Awolowo University, Nigeria
2
Department of Molecular & Clinical
and 45% [1] . An estimated 387,000 children were transmission during breastfeeding [6,7] . Maternal Pharmacology, Institute of
infected with HIV through MTCT in 2008 [201] . HA ART during pregnancy, delivery and Translational Medicine, University of
Liverpool, Liverpool, UK
Approximately 80% of these children acquired through planned weaning by 6 months postpar- 3
Program for HIV Prevention &
HIV infection during labor and in the peri- tum can reduce HIV transmission to less than Treatment, Department of Medical
partum period, with the remaining infections 2% [8] . The 2010 WHO guidelines for the use Technology, Faculty of Associated
Medical Sciences, Chiang Mai
occurring during breastfeeding [202] . A major of ARVs for treating pregnant women and pre- University, Thailand
step forward in the fight to eliminate pediatric venting HIV infection in infants recom­mends 4
Harvard School of Public Health,
Boston, MA, USA
HIV/AIDS has been the introduction of highly triple-ARV treatment for pregnant women who 5
Institut de Recherche pour le
efficacious, prophyl­actic, antiretroviral (ARV) need therapy for their own health (i.e., CD4 Développement (IRD), UMI 174,
Marseille, France
regimens to prevent MTCT of HIV. cell count below 350 cells/ml). For women who *Author for correspondence:
The ACTG 076 trial was the first to demon­ are not eligible for ARV therapy, two options Tel.: +66 53 894 994
strate the efficacy of maternal/infant ARV are proposed: either ZDV monotherapy from Fax: +66 53 894 220
tim.cressey@phpt.org
prophyl­­axis in prevention of MTCT of HIV [2] . as early as 14-weeks gestation plus intrapar-
Providing zidovudine (ZDV) monotherapy dur- tum single-dose NVP; or maternal triple-ARV
ing pregnancy (starting between 14 and 34 weeks prophyl­­axis [203] . Breastfeeding infants are rec-
gestation), during delivery and to the newborn ommended to receive daily NVP prophyl­­axis
for 6 weeks reduced HIV transmission rates from from birth throughout breastfeeding.
25 to 8% in nonbreastfeeding infants. A single Achieving optimal ARV exposure throughout
maternal dose of nevirapine (NVP) at the onset pregnancy is critical. Physiological changes dur-
of labor and to the newborn at 48–72 h of life ing pregnancy can influence drug disposition and
was shown to reduce perinatal HIV transmission several studies have demonstrated reduced ARV
[3] . Combining ZDV monotherapy prophyl­­axis exposures during pregnancy [9] . Considerable
with single-dose maternal/infant NVP reduced data have demonstrated that host genetic poly-
HIV transmission to less than 2% [4] . Owing to morphisms in drug-metabolizing enzymes and
the rapid scale-up of ARV treatment programs, drug transporters can contribute towards the
an increasing number of HIV-infected pregnant interpatient variability of ARV pharmaco­­kinetics
women are receiving highly active antiretroviral [10] . Associations between SNPs and drug con-
therapy (HAART) during pregnancy for HIV centrations for several ARVs in adults and chil-
treatment and/or the prevention of transmission dren are well described [11] . Accumulating data
of HIV. HAART during pregnancy and delivery also indicate that SNPs associated with drug
can reduce HIV transmission rates to 1.3% in toxicities or pharmaco­­kinetics can help identify part of

10.2217/PGS.12.138 © 2012 Future Medicine Ltd Pharmacogenomics (2012) 13(13), 1501–1522 ISSN 1462-2416 1501
Review Olagunju, Owen & Cressey

individuals at risk of early treatment discontinu- pregnancy can decrease drug absorption of weak
ation [12,13] . The influence of host genetic poly- acids, while increasing the absorption of weak
morphisms on HAART during pregnancy has bases. Longer gastrointestinal emptying/transit
not been extensively studied. However, it is pos- times due to higher progesterone production
sible that SNPs associated with ARV pharmaco­­ during pregnancy can slow the rate of absorp-
kinetics could exacerbate or reduce the effect of tion but could potentially increase overall drug
pregnancy-induced changes. In addition, data absorption. Nausea and vomiting associated
describing the influence of pharmaco­genetics with pregnancy can contribute to lower drug
on ARV safety and efficacy during lactation are adherence and absorption. Increases in body
limited. water, plasma volume and fat stores can increase
The aim of this review is to provide an update the volume of distribution of a drug thereby
on the effect of pregnancy-induced changes on reducing maximum plasma drug concentra-
the pharmaco­k inetics of ARVs and to provide tions. Pregnancy has been associated with a 15%
an overview of the potential role of pharmaco­ decline in plasma albumin and a >50% decline
genetics. The implications of pharmaco­genetics in a-1-acid glycoprotein concentrations, there-
on the exposure of breastfed infants to maternal fore the clearance of highly protein-bound drugs
ARVs during lactation are also discussed. may be higher during pregnancy because of the
increased percentage of unbound drug avail-
ARVs & HIV treatment able for metabolism (hepatic extraction ratio).
There are currently 26 ARVs approved by the Furthermore, changes in glomerular filtration
US FDA [204] . ARVs are classified into differ- rate, active tubular secretion and/or absorp-
ent classes based on their mechanisms of action. tion can affect renal excretion of drugs. The
To date, there are six ARV drug classes: nucleo- glomerular filtration rate in healthy women was
side/nucleotide reverse transcriptase inhibitors shown to be at least 50% higher during preg-
(NRTIs/NtRTIs); non-nucleoside reverse tran- nancy [14] . Common membrane transporters
scriptase inhibitors (NNRTIs); protease inhibi- are involved in active tubular secretion and/or
tors (PIs); integrase strand transfer inhibitors absorption but the effect of pregnancy has not
(INSTIs); entry inhibitors; and fusion inhibitors. been studied. Alterations in the activities of
HAART is recommended for the treatment of several key hepatic drug-metabolizing enzymes
HIV, and generally this involves providing triple-­ during pregnancy have been reported. The
ARV combinations from at least two drug classes. expression and activities of CYP3A4, CYP2C9
For example, current first-line HAART regimens and CYP2C19 vary at different stages of preg-
combine two NRTIs plus either a NNRTI, PI nancy. Tracy et al. reported a 35–38% increase
or INSTI [205] . in CYP3A4, CYP3A5 and CYP3A7 activities
All approved ARVs are administered orally, at all stages of pregnancy compared with post-
except for the fusion inhibitor, which is injected partum [15] . Progesterone-mediated activation
under the skin using a needle-free injection of nuclear receptors involved in the regula-
device. The majority of NRTIs/NtRTIs are tion of drug-metabolizing enzymes has been
excreted as unchanged drugs, while the other suggested as one of the mechanisms for preg-
classes undergo Phase I and/or II metabolism nancy-induced increases in drug-­metabolizing
by the CYP450 enzymes and UGT. A summary enzyme expression [16] . CYP3A4 is known to
of the elimination pathways for each of the share transcriptional regulation and induction
approved ARVs is presented in Table 1. Several pathways (mainly through the PXR and the
ARVs are both inducers and inhibitors of various CAR [NR1I3]) with other drug-metabolizing
CYP enzymes and drug transporters, which can enzymes, including CYP2B6, CYP2C and
lead to complex drug–drug interactions. UGT [17–19] . UGT-mediated glucuronidation
of some drugs also has been shown to increase
Physiological changes during during pregnancy. For example, the clearance of
pregnancy & potential effect on drug dihydro­artemisinin (a UGT1A9/2B7 substrate),
disposition the active metabolite of the antimalaria drug
Drug absorption, distribution, metabolism and artesunate, increases by 42% during pregnancy
excretion can be influenced by physiological [20] . Finally, the activity of ADH was higher dur-
changes during pregnancy. Changes in drug ing pregnancy in rodents [21] . The functional
absorption during pregnancy are highly depend- consequences of SNPs on genes encoding com-
ent on the physiochemical properties of drugs. mon metabolizing enzymes are summarized
For example, increases in gastric pH during in Table 2 .

1502 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
Table 1. Summary of antiretroviral-metabolizing enzymes, transporters and protein binding.
Drug Metabolism Transporters Protein binding (%)
Nucleoside reverse transcriptase inhibitors
Abacavir ADH and UGT ABCB1 and ABCG2 50
Didanosine Excreted unchanged ABCC4, SLC28A2, SLC28A3, SLC29A1, <5
SLC29A2 and SLC29A3
Emtricitabine Excreted unchanged ABCC1 and ABCC4 <4
Lamivudine Excreted unchanged ABCG2, ABCC4, SLC22A1, SLC22A2, 36
SLC22A3, SLC28A1, SLC28A3 and SLC29A3
Stavudine Excreted unchanged ABCC4, SLC28A1, SLC28A3 and SLC29A3 <10
Tenofovir Excreted unchanged ABCB1, ABCC4, ABCC10, SLC22A6 and >7
SLC22A8
Zidovudine UGT2B7 ABCG2, ABCC4, SLC22A6, SLC22A7, SLC22A8 30–38
and SLC22A11
Non-nucleoside reverse transcriptase inhibitors
Delavirdine CYP3A4 NA 98
Efavirenz CYP2B6, CYP2A6, UGT2B7 and ABCG2, Abcb5 99
CYP3A4
Etravirine CYP2C19, CYP3A4 and CYP2C9 NA >99
Nevirapine CYP3A4 and CYP2B6 ABCC10 60
Rilpivirine CYP3A4 and CYP3A5 NA >99
Protease inhibitors
Amprenavir CYP3A4 ABCB1 90
Atazanavir CYP3A4 ABCB1, ABCC1 and ABCG2 86
Darunavir CYP3A4 ABCB1, SLCO1A2 and SLCO1B1 95
Fosamprenavir CYP 3A4 ABCB1 90
Indinavir CYP3A4 ABCB1 and ABCC2 60
Lopinavir CYP3A4 ABCB1, ABCC1, ABCC2, SLCO1A2 and 98
SLCO1B1
Nelfinavir CYP3A4 and CYP2C19 ABCB1 >98
Ritonavir CYP3A4 and CYP2D6 ABCB1 and ABCC2 98
Saquinavir CYP3A4 and CYP3A5 ABCB1, ABCC2, SLCO1A2, SLCO1B1 and 97
SLCO1B3
Tipranavir CYP3A4 ABCB1 >99
Integrase/fusion/entry inhibitors
Enfuvirtide Excreted unchanged NA 92
Maraviroc CYP3A4 ABCB1 and SLCO1B1 76
Raltegravir UGT1A1 ABCB1, SLC15A1 and OAT1c 83
NA: No available data.

Pregnancy & drug transporters the physiochemical properties, placental blood


ARV use during pregnancy can lead to fetal flow, plasma pH, protein binding and placen-
exposure to the drugs. Maternal ARV pen- tal drug metabolism, fetal exposure is depend-
etration into the fetal compartment may be a ent on the placental expression, subcellular
factor contributing to towards their overall localization, activity and substrate specificity of
prophyl­actic effect [22] . Conversely, fetal ARV drug efflux and influx transporters [25] . At least
exposure has implications for toxicities that 18 transporters known to be relevant to ARV
have been documented [23,24] . In addition to distribution are expressed on either the apical

future science group www.futuremedicine.com 1503


Review Olagunju, Owen & Cressey

Table 2. Influence of genetic polymorphisms in metabolizing is the net effect on exposure of the infant to
enzymes/nuclear receptors on antiretroviral drug exposure. maternal xenobiotics.
Other factors such as drug–drug interactions,
Protein/gene Functional SNPs (effect on plasma exposure) Ref.
HIV infection and coinfections have been rec-
CYP2A6 1836G>T and 1093G>A (↑ EFV) [113,114]
ognized to affect drug transporter function in
CYP2B6 516G>T and 983T>C (↑ EFV and ↑ NVP) [94,95] other tissues [29–33] . Drug–drug interactions at
CYP2C19 rs4244285 and rs12248560 (? ETR) [123] the human placenta can also affect fetal expo-
sure to maternal drugs [34] . Therefore, the effects
CYP3A5 CYP3A5*1 (↓ SQV, ↓ IDV and ↓ ATV) [95,143]
of such nongenetic factors are recognized to be
*3 (↓ NVP)
important but are understudied and beyond the
UGT1A1 UGT1A1*28 and 211G>A (↑ ATV-related hyperbilirubinemia) [148,149] scope of this review.
UGT2B7 735A>G (↔ EFV) [114]
802C>T (↓ EFV) NRTI/NtRTI pharmacokinetics during
CAR (NR1I3) 540CC (↑ discontinuation of EFV) [13] pregnancy & the potential role of
PXR (NR1I2) 63396 C>T (↓ ATV) [144]
pharmacogenetics
There are currently eight NRTIs/NtRTIs
?: Effect unknown; ↑: Increased; ↓: Decreased; ↔: No difference; ATV: Atazanavir; EFV: Efavirenz;
ETR: Etravirine; IDV: Indinavir; NVP: Nevirapine; SQV: Saquinavir. approved for HIV treatment. All NRTIs/NtRTIs
are administered as prodrugs and must
(maternal) and basolateral (fetal) membranes of undergo intracellular metabolism by cellular
syncytiotrophoblasts as well as the endothelium kinases to their active ‘triphosphate’ anabo-
of fetal capillaries (Figure 1) . Members of the ATP- lites. Routine measurement of intracellular
dependent binding cassette efflux transporter NRTI–triphosphate concentrations remains
superfamily limit fetal exposure to maternal challenging. Comprehensive NRTI pharmaco­
ARVs [26] . Transporters in the ATP-independent kinetic data available during pregnancy have
solute carrier superfamily, including organic subsequently been restricted to plasma NRTI
anion transporting polypepetides, organic anion concentrations; however, the relationship
transporters, organic cation transporters (OCTs) between NRTI plasma concentrations and drug
and equilibrative nucleoside transporters, are also efficacy/toxicity remains unclear [35] . Below is
relevant for distribution of ARVs into the fetal a summary of the effect of pregnancy on indi-
compartment. Recently, Iqbal et al. reviewed the vidual NRTI/NtRTI exposure and available
gestational changes in the expression and func- pharmaco­genetic data.
tion of placental drug transporters as well as the ZDV (also know as azidothymidine) is a thy-
associated genetic polymorphisms [27] . The func- midine analog and was the first ARV approved
tional consequences of genetic polymorphisms for treatment of HIV infection in 1987 [204] . It
in the encoding genes were also been recently was also the first ARV administered to HIV-
highlighted by Ieiri et al. [28] . The substrate spe- infected pregnant women and continues to play
cificities, placental and mammary gland expres- a major role in the currently recommended
sion, and effects of associated functional SNPs ARV prophyl­actic regimens. ZDV is primarily
are presented in Table 3. Their localization on the eliminated through hepatic metabolism by UGT
basolateral or apical membranes of the placental [36] . Several studies have reported that ZDV
syncytiotrophoblast and the cord:maternal blood pharmaco­k inetics during the third trimester
(C:M) ratios for ARVs are shown in Figure  1. of pregnancy are similar to nonpregnant adults
Ultimately, the transfer of drug from mother [37–39] . However, lower ZDV exposure during
to infant will depend on the combined influ- the third trimester versus the postpartum period
ence of influx and efflux transporters within the has been reported (Table  4) [40] . The standard
placental barrier. Furthermore, the subcellular dose of ZDV is currently 300 mg twice daily,
localization of such transporters is also likely to and no adjustments are recommended during
be important since an apical influx transporter pregnancy. A lower dose of ZDV, 200 mg twice
may exhibit a similar effect upon fetal exposure daily, has been studied in nonpregnant adults
(in terms of direction) as that of a basolateral with low bodyweights (<60 kg) owing to con-
efflux transporter and vice versa. Therefore, it is cerns of higher rates of drug toxicity. Pharmaco­
important to consider synergy and antagonism kinetic [41] and efficacy data [42] suggest that
between multiple transporters when examining this lower dose may be preferable in this patient
specific mechanisms. Although it is conceivable group; however, this dose has not been studied
that transport may occur from mother to infant in pregnant women. ZDV readily crosses the
or from infant to mother, the key phenotype placenta to the fetal compartment with a C:M

1504 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
concentration ratio of 1.13–1.27 [40] . To date, implications of ABCC4 polymorphisms during
no data on the effect of UGT host genetic poly- pregnancy therefore warrant further study.
morphisms on plasma ZDV pharmaco­k inetics Stavudine (d4T) is thymidine analog. The
have been reported. Association between ZDV- WHO ARV treatment guidelines recommend
triphosphate concentrations and a SNP in the a 30-mg twice-daily dose for adults [206] . The
drug transporter ABCC4 (MRP4) in HIV- pharmaco­k inetics of d4T in pregnancy appear
infected nonpregnant adults has been reported to be similar to those in HIV-infected men and
[43] . Median ZDV–triphosphate concentra- no dose adjustment is recommended [45] . Mean
tions were 49% higher in ABCC4 3724G>A d4T C:M concentration ratio is 1.0 (95% CI:
(rs11568695) carriers versus homozygotes for 0.63–1.37). This drug was included in the first
the common allele (p = 0.03). Two SNPs within generic fixed-dose combinations developed and
ABCG2, 421C>A (rs2231142) and 34G>A has been widely used in resource-limited set-
(rs2231137), were also assessed, but neither were tings. However, WHO now suggests that coun-
associated with ZDV–triphosphate concentra- tries phase out d4T and develop plans to move
tions. ABCC4 has been shown to contribute to towards alternative firstline regimens, primar-
ZDV uptake into conditionally immortalized ily owing to d4T-associated lipodystrophy. No
rat syncytiotrophoblasts [44] and the clinical data are available on the role of transporter or

TFV (0.26–1.95)
SQV (0.00–0.06) [170] ABCC1
[169]

TPV (0.41)‡ RAL (0.09–2.3)


ABCB1 [156]
CYP3A4 [161] SLCO1B3
RTV (0.00–0.03)
[169] NVP (0.37–0.93)
[171] SLCO1B1
NFV (0.07–0.43)
SLC29A1 [172] UGT2B7 SLC22A3
MVC† (0.03–0.05)
[173]
SLC22A2
CYP3A4
BCRP ETR (0.30–0.40)
[122]
ZDV (0.18–17.2)
[169]
T-20 (ND) DRV (0.15–0.31) SLC22A1
[174] [175]

ABCC5
SLC29A2
LPV (0.18–0.21) ABC (0.92–1.42)
[130] [169] SLCO1A2
CYP2B6
IDV (0.00–0.08) EFV (0.37–0.74)
[108] SLC22A11
[169] APV (0.24–0.45)
ABCC2 [169]
SLC22A7

ATV (0.10–0.16) ABCC10 3TC (0.21–4.03)


[140] [169]

Maternal 0% 25% 50% 75% 100% Infant


(apical) (basolateral)

Figure 1. Placental expression of antiretroviral transporters and metabolizing enzymes. Cord:maternal blood (C:M) ratios are
shown in parentheses after the drug name. SLCO1A2 and SLCO1B3 are predominantly localized to the vasculo–syncytial membrane, while
SLCO1B1 appears to be expressed only in the first trimester. ABCC10 is also expressed at the placenta barrier, but its location has not
been determined.

In rhesus macaques.

A single case report.
3TC: Lamivudine; ABC: Abacavir; APV: Amprenavir; ATV: Atazanavir; DRV: Darunavir; EFV: Efavirenz; ETR: Etravirine; IDV: Indinavir;
LPV: Lopinavir; MVC: Maraviroc; NFV: Nelfinavir; NVP: Nevirapine; RAL: Raltegravir; RTV: Ritonavir; SQV: Saquinavir; T‑20: Enfuvirtide;
TFV: Tenofovir; TPV: Tipranavir; ZDV: Zidovudine.

future science group www.futuremedicine.com 1505


Review Olagunju, Owen & Cressey

Table 3. Placenta and mammary gland expression, localization and substrate specificity of known antiretroviral
drug transporters and effects of associated functional SNPs.
Transporter (gene) Tissue localization Substrates Effects of SNPs Ref.
MDR1 (ABCB1) Placenta †
PIs , RAL, MVC, TFV

3435C>T (↓ protein expression and ↓ NVP ht) [30,33,
and ABC 2677G>T (↓ protein expression) 176,177]
-129T>C (↓ protein expression)
MDR5 (ABCB5) Mammary gland§ EFV¶ ND [178]

MRP1 (ABCC1) Placenta and mammary


#
ATV, RTV, LPV, SQV, ND [33,179]
gland # IDV and FTC
MRP2 (ABCC2) Placenta† PIs‡ 1249G>A (SQV ↑), -24C>T (↓ IDV and ↓ TFV [82,180]
ktd)
MRP5 (ABCC5) Placenta # and mammary gland d4T ND [177,181]

BCRP (ABCG2) Placenta† and mammary gland NRTIs and EFV (?) C421A (↓ exp/act) [177,182]
rs2231164 (?)
rs2622604 (?)
MRP7 (ABCC10) Placenta§ and mammary gland TFV and NVP rs9349256 and rs2125739 (↑ TFV ktd), [72,96]
c.2759T>C (↓ NVP)
OATP1A2 (SLCO1A2) Placenta #†† PIs‡ SLCO1A2 516A>C (↔ LPV) [137,183]
404A>T (?)
559G>A (?)
38T>C (?)
833A>del (?)
OATP1B1 (SLCO1B1) ‡‡ Placenta # PIs‡ SLCO1B1 521T>C (↑ MVC and ↑ LPV) [137,138]

OATP1B3 (SLCO1B3) Placenta # PIs‡ ND [137,183]

OAT1(SLC22A6) Mammary gland TFV, RAL and ZDV R50H (↑ affinity for substrates) [30,184,185]

OAT2 (SLC22A7) Placenta and mammary gland


#
ZDV ND [57]

OAT3 (SLC22A8) Mammary gland ZDV and TFV ND [57]

OAT4 (SLC22A11) Placenta # ZDV ND [57,186,187]

ENT1 (SLC29A1) Placenta and mammary gland



ddI ND [57,188]

ENT2 (SLC29A2) Placenta and mammary gland



ddI and ZDV ND [57,188]

OCT1 (SLC22A1)§§ Placenta† and mammary gland 3TC OCT1 Arg61Cys (↑ mRNA exp) [56,58,
P283L and -P341L (↓ 3TC uptake) 188,189]

OCT2 (SLC22A2) Placenta # 3TC T199I, -T201M and -A270S (↓ 3TC uptake) [56,58,190]

OCT3 (SLC22A3) Placenta # and mammary gland 3TC rs2292334, rs2048327, rs1810126 and [56,188,
rs3088442 (↑ mRNA exp) 189,191]

Apical.

Most PIs are substrates and inhibitors of these transporters.
§
Unknown.

Study in rat GI tract.
#
Basolateral.
††
SLCO1A2 mRNA expression increases during gestation.
‡‡
Only mRNA detected.
§§
OCT1 showed more than fourfold higher RNA levels in lactating mammalian epithelia cells compared with nonlactating.
(?): Effect unknown; ↑: Increased; ↓: Decrease; ↔: No difference; 3TC: Lamivudine; ABC: Abacavir; Act: Activity; ATV: Atazanavir; d4T: Stavudine; ddI: Didanosine;
Del: Deletion; EFV: Efavirenz; Exp: Expression; FTC: Emtricitabine; Ht: Hepatotoxicity; IDV: Indinavir; Ktd: Kidney tubular dysfunction; LPV: Lopinavir; MVC: Maraviroc;
ND: No data; NVP: Nevirapine; NRTI: Nucleoside reverse transcriptase inhibitor; PI: Protease inhibitor; RAL: Raltegravir; RTV: Ritonavir; SQV: Saquinavir; TFV:
Tenofovir; ZDV: Zidovudine.

metabolism polymorphisms of d4T, but an asso- ABC were similar during the third trimester and
ciation between HLA‑B*4001 and d4T-associated postpartum, and that it readily crosses the pla-
lipodystrophy has been reported in Thailand [46] . centa (C:M ratio of 1.06) [47] . ABC is metabo-
Abacavir (ABC) is a guanosine analog lized by ADH and UGT, but there are currently
approved by the FDA in 1998 [204] . The recom- no reports on the influence of ADH or UGT
mended dosage for adults is 300 mg twice daily. genetic polymorphisms on ABC pharmaco­
Best et al. reported that the pharmaco­k inetics of kinetics. ABC-hypersensitivity reaction can

1506 Pharmacogenomics (2012) 13(13) future science group


Table 4. Summary of pharmacokinetic parameters of antiretroviral drugs during pregnancy and postpartum from selected publications.
Drug Dosage (mg) Third trimester of pregnancy Postpartum Ref.
(US FDA AUC (µg/h/ml) Cmax (µg/ml) Ctrough (µg/ml) AUC (µg/h/ml) Cmax (µg/ml) Ctrough (µg/ml)
category)
Nucleoside reverse transcriptase inhibitors

future science group


3TC (C), 150 b.i.d. 4.9 (4.0–6.1)† 1.3 (1.1–1.6) NR 5.1 (3.7–6.9) ‡ 1.20 (0.98–1.50) NR [39]
GM (95% CI)
ABC (C), 300 b.i.d. 5.9 (5.2–6.8) 1.9 (1.6–2.2) BQL (BQL–0.102) 5.4 (4.3–6.8) 2.1 (1.6–2.7) BQL (BQL–0.203) [47]
GM (95% CI)
FTC (B), 200 q.d. 8.0 (7.1–8.9) 1.4 (1.2–1.6) 0.06 (0.04–0.06) 9.7 (8.6–10.9) 1.4 (1.3–1.7) 0.09 (0.07–0.1) [63]
median (range)
TDF (B), 245 q.d. 2.4 (2.1–2.6) 0.27 (0.22–0.33) 0.05 (0.04–0.06) 3.2 (2.7–3.7) 0.35 (0.29–0.42) 0.08 (0.06–0.09) [68]
GM (95% CI)
ZDV (C), mean 200 every 4 h 1.2 (±0.27) 1.04 (±0.45) NR 1.8 (±0.13) 1.2 (±0.01) NR [40]
(±SD)
Non-nucleoside reverse transcriptase inhibitors
EFV (D), 600 q.d. 55.4 (13.5–220.3) 5.4 (1.9–12.2) 1.60 (0.23–8.13) 58.3 (22.7–214.4) 5.10 (1.96–11.42) 2.05 (0.31–8.43) [108]
median (range)
ETR (B), 200 q.d. 4.5 (3.0–8.9)§ 0.69 (0.45–1.20) 0.41 (0.15–0.90) NR NR NR [122]
median (range)
NVP (B), 200 b.i.d. 47.0 (41.5–58.9) 5.3 (4.7–6.3) 3.1 (2.7–4.1) 59.4 (53.6–69.6) 6.6 (5.9–7.7) 3.9 (3.5–4.9) [91]

www.futuremedicine.com
GM (90% CI)
Protease inhibitors
fAPV (C), 700/100 b.i.d. 32.4 (14.2–54.4) 5.93 (1.66–12.53) 1.70 (0.71–3.23) 50.7 (11.6–62.4) 5.7 (2.1–11.4) 2.43 (0.37–3.82) [153]
median (range)
ATV/RTV (B), 300/100 q.d. 41.9 (27.4–60.8) 3.6 (2.8–5.1) 0.7 (0.5–1.1) 57.9 (47.1–64.8) 4.1 (3.0–5.8) 1.2 (1.1–2.0) [141]
median (IQR)
ATV/RTV (B), 400/100 q.d. 46.6 (11.0–88.3) 4.70 (0.88–7.50) 0.74 (0.14–2.10) 55.1 (9.9–99.5) 4.52 (0.93–9.45) 0.88 (BQL–2.70) [142]
median (range)
DRV/RTV (B), 600/100 b.i.d. 50.8 (23.8–102.0) NR 3.10 (0.78–8.90) 70.0 (40.3–176.0) NR 2.8 (1.6–5.5) [152]
median (range)
DRV/RTV (B), 800/100 q.d. 67.7 (30.3–106.0) NR 1.60 (0.15–2.50) 87.9 (77.5–150.0) NR 2.83 (1.14–4.00) [152]
median (range)
IDV/RTV (C), 400/100 b.i.d. 16.1 (7.5–39.9) 3.5 (1.3–7.4) 0.13 (0.07–0.60) 27.1 (18.6–44.7) 5.5 (3.8–9.4) 0.28 (0.14–0.71) [192]
median (range)
Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding


Single dose.

Steady state.
§
AUC 6 h reported for one patient and AUC 12 h for others.
3TC: Lamivudine; ABC: Abacavir; ATV: Atazanavir; AUC: Area under the curve; b.i.d.: Twice daily; BQL: Below quantification limit; DRV: Darunavir; EFV: Efavirenz; ETR: Etravirine; fAPV: Fosamprenavir; FTC: Emtricitabine;
GM: Geometric mean; IDV: Indinavir; IQR: Interquartile range; LPV: Lopinavir; MVC: Maraviroc; ND: No pregnancy vs postpartum pharmacokinetic data; NFV: Nelfinavir; NFV‑M8: Nelfinavir metabolite; NR: Not reported;
NVP: Nevirapine; q.d.: Once daily; RAL: Raltegravir; RTV: Ritonavir; SD: Standard deviation; SQV: Saquinavir; T‑20: Enfuvirtide; TDF: Tenofovir disoproxil fumarate; TPV: Tipranavir; ZDV: Zidovudine.
Review

1507
Table 4. Summary of pharmacokinetic parameters of antiretroviral drugs during pregnancy and postpartum from selected publications (cont).

1508
Drug (US Dosage (mg) Third trimester of pregnancy Postpartum Ref.
FDA AUC (µg/h/ml) AUC (µg/h/ml)
Review
Cmax (µg/ml) Ctrough (µg/ml) Cmax (µg/ml) Ctrough (µg/ml)
category)
Protease inhibitors (cont.)
LPV/RTV (C), 400/100 b.i.d. 58.0 (50.4–70.7) 7.5 (6.7–8.7) 2.5 (2.0–3.5) 89.4 (74.0–107.9) 10.0 (8.4–11.8) 4.7 (3.1–6.8) [131]
GM (95% CI)
LPV/RTV (C), 400/100 b.i.d. 64.6 (59.7–69.8) 8.1 (7.5–8.7) 2.7 (2.4–3.1) NR NR NR [132]
median (range) (Thai women)
LPV/RTV (C), 600/100 b.i.d. 96 (43–198) 10.7 (5.8–19.1) 5.1 (1.5–12.2) 133 (66–237) 14.6 (9.8–22.8) 7.2 (2.8–21) [136]
median (range)
NFV (B), 1250 b.i.d. 18.9 (3.6–53.7) 3.2 (0.9–6.5) 0.900 30.8 (1.3–123.9) 4.6 (0.3–9.9) 1.100 (<0.039–4.800) [193]
median (range) (<0.039–4.400)
NFV-M8, NA 1.500 (<0.234–12.400) 0.3 (0.1–1.9) 0.100 (<0.039–0.900) 11.300 1.200 (<0.039–4.900) 0.300 (<0.039–3.900) [193]
Olagunju, Owen & Cressey

median (range) (<0.234–43.700)


SQV/RTV (B), 1000/100 b.i.d. 12.71 (8.96–26.93) 3.23 (2.68–6.03) 0.26 (0.23–1.01) 28.94 (14.55–58.49) 3.92 (2.96–5.66) 0.86 (0.25–2.21) [194]
GM (95% CI)
TPV (C), ND ND ND ND ND ND ND –
median (range)
Others
MVC (B) ND ND ND ND ND ND ND –
median (range)
RAL (C) 400 b.i.d. 8.3 (3.0–17.5) 1.80 (0.69–6.30) 0.090 (0.014–0.470) 7.5 (2.0–25.5) 2.80 (0.35–8.90) 0.500 (<0.010–0.770) [156]

Pharmacogenomics (2012) 13(13)


median (range)
T‑20 (B) ND
median (range)

Single dose.

Steady state.
§
AUC 6 h reported for one patient and AUC 12 h for others.
3TC: Lamivudine; ABC: Abacavir; ATV: Atazanavir; AUC: Area under the curve; b.i.d.: Twice daily; BQL: Below quantification limit; DRV: Darunavir; EFV: Efavirenz; ETR: Etravirine; fAPV: Fosamprenavir; FTC: Emtricitabine;
GM: Geometric mean; IDV: Indinavir; IQR: Interquartile range; LPV: Lopinavir; MVC: Maraviroc; ND: No pregnancy vs postpartum pharmacokinetic data; NFV: Nelfinavir; NFV‑M8: Nelfinavir metabolite; NR: Not reported;
NVP: Nevirapine; q.d.: Once daily; RAL: Raltegravir; RTV: Ritonavir; SD: Standard deviation; SQV: Saquinavir; T‑20: Enfuvirtide; TDF: Tenofovir disoproxil fumarate; TPV: Tipranavir; ZDV: Zidovudine.

future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
occur in 5–8% of patients within 6 weeks of 3TC for prevention of MTCT [60] . Recent studies
initiating treatment and in rare cases can be fatal have associated mtDNA halogroup T/L1c and
[48] . The HLA‑B*5701 allele is strongly associ- mtDNA 4917G (increased risk), HFE 845G>A
ated with ABC hypersensitivity and ABC is cur- (rs1800562) and IL-12B 3´-UTR*2 (rs3212227)
rently the only ARV for which pharmaco­genetic (decreased risk), as well as TNF- variants 1031*2
testing is currently recommended [49,50] . (rs1799964) and 308*2 (rs1800629) (increased
Lamivudine (3TC) is a cytidine analog risk) with NRTI-associated adult neuropathy
excreted primarily unchanged via the kidneys. [61,62] . Their roles in infant mitochondrial toxic-
3TC was approved by the FDA in 1995 [204] . ity as a result of in utero exposure to maternal
The recommended dosage of 3TC for adults is drug need further investigation.
300 mg once daily or 150 mg twice daily. The Emtricitabine (FTC) was approved by the
pharmaco­kinetics of 3TC during pregnancy was FDA in 2003 [204] . Like 3TC, it is a cytidine
reported to be similar during the last trimester analog excreted primarily unchanged via the kid-
compared with nonpregnant adults (Table 4) [39] . neys. The approved dosing in adults is 200 mg
A recent population pharmaco­k inetic study once daily. Within-subject comparisons of FTC
reported the oral clearance of 3TC to be 22% pharmaco­kinetics during pregnancy and postpar-
higher in pregnant women. However, the authors tum revealed significantly higher apparent clear-
concluded that this increase was not clinically rel- ance during pregnancy [63] . Significantly lower
evant and did not warrant a dose adjustment [51] . area under the curve (AUC) and drug concentra-
It crosses the human placenta with a median C:M tion 24 h postdose also were found during the
concentration ratio of 1.06. However, amniotic third trimester of pregnancy (Table 4) . FTC was
fluid concentrations were five-times higher than well distributed to the fetal compartment with
in maternal plasma, suggesting passive diffusion mean C:M concentration ratio of 1.2 (1.0–1.5).
across the amnion, fetal uptake and fetal clear- FTC is excreted into human breast milk but the
ance [52] . Like ZDV, 3TC is a substrate for the concentrations attained would represent a dose
drug transporter ABCC4 and ABCC4 4131G>T approximately 2% of the proposed oral dose for
(rs3742106) carrier status has been associated neonates [64] . It is a substrate for ABCC1 [65] ,
with 20% higher 3TC–triphosphate intra­cellular which is expressed in both the placenta and
concentrations (p  =  0.004) [43] . Furthermore, mammary gland [57] . However, no studies have
3TC breast milk concentrations were reported assessed the influence of genetic polymorphisms
to be approximately threefold higher than mater- and its disposition.
nal serum concentrations [53] . However, 3TC Tenofovir disoproxil fumarate (TDF) is a
ingested by breastfed infants appears to be poorly nucleotide analog of adenosine and was approved
absorbed with low plasma concentrations (<0.10 by the FDA in 2001 [204] . The recommended
µg/ml) detected [54] . 3TC is also a substrate for oral dose for adults is 300 mg once daily. TDF is
the organic cation transporters OCT1, OCT2 the oral prodrug of tenofovir (TFV). Following
and OCT3 [55,56] , all of which are expressed oral absorption, TDF rapidly undergoes esterase
in the kidney, placenta and mammary gland hydrolysis and is converted to TFV, a nucleo-
(exepct for OCT2 in the latter) [57] . In a recent tide (nucleoside monophosphate) analog that is
study Choi et al. observed reduced 3TC uptake taken up by cells. Intracellularly, TFV is rapidly
in oocytes expressing OCT1-P283L and OCT1- phosphorylated by cellular nucleotide kinase to
P341L as well as those expressing OCT2-T199I, its active anabolite TFV diphosphate. TFV is
OCT2-T201M and OCT2-A270S variants com- primarily excreted unchanged by the renal route
pared with with the reference sequences [58] . The through a combination of glomerular filtration
consequences of functional SNPs of OCTs on and active tubular secretion [66] . Burchett et al.
3TC pharmaco­k inetics have not been studied assessed the pharmaco­kinetics of TFV between
in vivo. Such SNPs may also influence the distri- 30 and 36 weeks gestation age and 6–12 weeks
bution of 3TC to the fetal compartment as well postpartum [67] . Median (range) TFV AUC
as its excretion into breast milk. was significantly lower during the third tri­
Rare cases of lethal mitochondrial dys­function mester, 2.5 (1.1–2.6) µg/h/ml during pregnancy
in infants exposed in utero to ZDV and 3TC were compared with 2.96 (1.62–7.2) postpartum
reported in the late 1990s [59] . These severe cases (p = 0.02). However, third trimester Cmin were
are clinically very similar to known genetically not different from postpartum values. A recent
induced mitochondrial defects. Evidence of study reported a 25% reduction in TFV exposure
mitochondrial damage has been demonstrated during the third trimester compared with post-
in HIV-uninfected infants exposed to ZDV and partum. The authors speculated that the decrease

future science group www.futuremedicine.com 1509


Review Olagunju, Owen & Cressey

was due to reduced absorption and/or increased which is expressed in the human placenta [57] , and
volume of distribution as no change in the termi- its possible role, including the effect of genetic
nal elimination half-life was observed (Table 4) [68] . polymorphisms, in the uptake of TFV into the
Benaboud et al. studied the effects of pregnancy fetal compartment should be studied. TFV is also
on TFV pharmaco­k inetics using a population excreted into human breast milk but at low con-
pharmaco­kinetic ana­lysis involving 46 pregnant centrations – equivalent to a median dose of 4.2
and 156 nonpregnant HIV-infected women [69] . µg/day for a 3‑kg neonate, or 0.03% of the oral
The apparent oral clearance was 39% higher in dose proposed for neonates [64] .
pregnant women. These authors suggested that
a TFV dose increase should be considered for Pharmacokinetics during pregnancy
women from the second trimester until deliv- & the potential role of
ery in order to achieve comparable exposure to pharmacogenetics
nonpregnant adults. „„ Non-nucleoside reverse transcriptase
It is of interest to note that, while TFV readily inhibitors
crosses the human placenta with C:M concen- Four NNRTIs are currently available for the
tration ratio of 1.04 (0.6–1.7) [67] , the transfer treatment of HIV. NNRTIs are primarily metab-
into cerebrospinal fluid has been reported to olized by CYP enzymes and, with the excep-
be an order of magnitude lower with cerebro- tion of NVP, are highly protein bound (Table 1) .
spinal fluid:plasma ratio of 0.057 (0.03–0.1) Therefore, drugs in this class are expected to
[70] . This difference may be indicative of sub- be susceptible to temporal changes in hepatic
stantial differences in the expression profile of enzyme activities and plasma protein fluctuations
transporters between these barriers. TFV is a during pregnancy. NNRTIs plasma drug con-
substrate for ABCC4, OAT1, OAT3 [71] and centrations also have been associated with SNPs
MRP7 (ABCC10) [72] . Interestingly, OAT1, in genes encoding the enzymes involved in their
OAT3 and ABCC4 are expressed in choroid metabolism, transport and nuclear receptor type
plexus epithelial cells but not in the placenta transcription factors.
[73–75] , while ABCC10 is expressed in both the NVP was the first NNRTI receiving FDA
placenta and CNS [57] . OATs are involved in the approval in 1996 [204] . It is initiated at a lower dose
efflux of their substrates out of the CNS [76,77] . (or ‘lead-in’ dose) for the first 2 weeks to allow
The mechanisms behind this observed differ- for the autoinduction of metabolizing enzymes,
ence have not been studied in detail. Carriers of which increases its clearance. Initiating NVP
ABCC4 3463A>G (rs1751034) were reported to in this stepwise fashion reduces the frequency
have 35% higher intracellular TDF diphosphate of drug-associated rashes. One 200 mg tablet is
concentrations than noncarriers (p = 0.04) [78] . administered once daily for the first 14 days and
TFV is transported into proximal tubular cells by if there are no clinical signs of rash the dose is
OAT1 and to a lesser extent OAT3 [79] , which is increased to 200 mg twice daily [84] . Single and
located on the basolateral membrane. It remains chronic NVP dosing has been used during preg-
unknown if a change in OAT1 expression/activ- nancy to prevent MTCT of HIV. A single mater-
ity plays a role in increased TFV clearance dur- nal dose of NVP at the onset of labor and to the
ing pregnancy, and if such changes are driven by newborn at 48–72 h of life is still recommended
genetic polymorphisms. as an option in the WHO prevention of MTCT
The luminal efflux system(s) involved in guidelines. Concerns have been raised over the
the transport of TFV out of proximal tubular use of single-dose NVP [203] . Due to its long half-
cells and into the urine is not well described. life, NVP can persist in the plasma for up to 4
Three transporters, ABCC2 [80] , ABCC4 [81] weeks postpartum following a single intrapartum
and ABCC10 [72] have been reported to play a dose [85] . This prolonged period of monotherapy,
role in the active renal tubular efflux of TFV. A in the presence of replicating viruses, can increase
genetic variant of ABCC2 (rs717620) [82] , and the risk of selecting NNRTI resistance mutations.
two ABCC10 SNPs (rs9349256 and rs2125739) NNRTI mutations detected in the post­partum
were identified as predictors of TFV-induced period have been shown to affect the efficacy of
nephrotoxicity [72] . Higher plasma TFV concen- future NNRTI based-HAART regimens [86] .
trations in patients with kidney tubular dysfunc- However, short-course ARV treatments post­
tion have also been reported [83] . The implica- partum (7–30 days) can reduce the risk of select-
tions of these SNPs on TFV pharmaco­kinetics ing NNRTI resistance mutations postpartum
during pregnancy and risk of tubular dysfunction following intrapartum NVP [87,88] . The associa-
are unknown. TFV is a substrate for ABCC10, tion of SNPs in metabolizing enzymes with NVP

1510 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
concentrations following an intra­partum single An association between HLA‑B*3505 and
dose have been reported, but the modest effects NVP-induced skin rash has been reported [99] .
may not be clinically significant [89] . Furthermore, a genome-wide association study
Chronic NVP dosing is increasingly used, in revealed that variations in the CCHCR1 gene
part owing to the availability of generic triple-­ were strongly associated with NVP-induced skin
drug fixed-dose combinations in resource- rash [100] . Incorporating genetic and clinical risk
limited settings. The geometric mean third factors may therefore have utility in reducing the
trimester:postpartum ratio for NVP AUC has incidence of NVP related-toxicities in women
been reported to be 0.90 (90% CI: 0.80–1.02) initiating NVP during pregnancy.
[90] and 0.79 (90% CI: 0.70–0.90) [91] . A 30% There are currently no data on NVP
decrease in NVP AUC between pregnant and pharmaco­k inetics during lactation. However,
nonpregnant women has been reported [92] , but its excretion into human breast milk has been
no dose adjustment is currently recommended. reported with milk:plasma ratios of 0.67 [53]
CYP2B6 516G>T (rs3745274) and CYP2B6 and 0.75 [54] , resulting in 0.97 and 0.73–1.03
983T>C (rs28399499) SNPs have been asso- µg/ml, respectively, in breastfed infants’ plasma.
ciated with higher NVP plasma concentra- Interestingly, this is higher than the >0.10 µg/ml
tions. Penzak et  al. reported NVP median target on which current postexposure prophyl­
plasma concentration of 7.61 µg/ml in indi- axis dosing guidelines are based [203] , but less
viduals with CYP2B6 516TT genotype versus than the target trough concentration of >3.0
4.18 and 5.56 µg/ml in those with 516GG and µg/ml in HIV-infected children [101] . However,
516GT genotypes, respectively [93] . These find- breast milk ingestion of NVP has been associ-
ings were confirmed in a larger study where ated with increased risk of cutaneous adverse
median (range) NVP plasma concentrations drug reactions in breastfed infants compared
were 5.18 (1.89–11.16), 6.13 (0.15–11.69) and with nelfinavir [102] . The new WHO guidelines
6.69 (4.16–21.03) µg/ml for carriers of CYP2B6 recommend NVP for post­exposure prophyl­a xis
516GG, GT and TT genotypes, respectively [94] . in breastfeeding populations, which may mean
This latter study was also the first to demon- that the exposure in infants in developing coun-
strate that carriers of CYP2B6 983 TT and TC tries whose mothers also take NVP may be higher
genotypes had significantly higher NVP plasma than anticipated. This may conceivably impact
concentrations. Both CYP2B6 516G>T and upon NVP-associated adverse drug reactions in
CYP3A5*3 (rs776746) were associated with NVP these infants but this has not been studied in
pharmaco­k inetics in a recent study in Malawi. detail.
The CYP2B6 516 T allele increased NVP AUC Efavirenz (EFV) was approved by the FDA
by 92% and CYP3A5*3 decreased NVP AUC by in 1998 [204] and early reports of congenital neu-
31% in a multivariable model [95] . The inclusion ral tube defects resulting from first-trimester
of host genetic polymorphisms in pharmaco­ exposure have limited its use during pregnancy
kinetic studies of NVP during pregnancy may [103] . Although emerging data on the safety of
help identity subpopulations of women who are EFV use in pregnancy provide some reassurance
at higher risk of subtherapeutic concentrations. [104–107] , the number of cases monitored and the
To date, relatively little data have emerged relative rarity of neural tube defects and facial
on efflux or influx transporters capable of NVP clefts cannot yet rule out some degree of risk.
transport. However, NVP was recently shown to In 2010, WHO updated their prevention of
be a substrate for ABCC10 and SNPs within the MTCT guidelines and recommended EFV use
gene encoding this efflux pump were shown to be as part of combination ARV therapy after the
associated with plasma concentrations of NVP first trimester [203] .
[96] . This is an area where further mechanistic Recently the first pharmaco­kinetic data evalu-
investigation is warranted since the identification ating the standard dose (600 mg once daily) of
of placental or mammary gland transporters may EFV during the third trimester of pregnancy and
underpin studies to identify pharmaco­genetic postpartum were published [108] . There was no sig-
correlates of exposure of the fetus or infant, nificant difference in EFV AUC and Cmax during
respectively. pregnancy compared with postpartum (Table 4) ;
Higher incidences of NVP-related rash and geometric mean ratios were 0.97 (90%  CI:
hepatotoxicity have been reported in HIV- 0.83–1.13) and 1.11 (90%  CI: 0.99–1.24),
infected pregnant women with CD4 counts respectively. However, EFV oral clearance was
of >250 cells/mm3 [97,98] and NVP-containing significantly higher during the third trimes-
HAART is not preferred in these patients. ter and the drug concentration 24 h postdose

future science group www.futuremedicine.com 1511


Review Olagunju, Owen & Cressey

significantly lower compared with postpartum. 1.0–4.0 µg/ml [117] . Maternal EFV concentra-
The C:M concentration ratio of EFV was 0.49 tions, which depend on various host genotypes,
(90% CI: 0.37–0.74). Based on these data, no will be a major determinant of infant exposure.
dose adjustment is required during pregnancy. However, EFV transport across the placenta and
Considerable data are available on the effect of into breast milk is also likely to be important but
host genetic polymorphisms on EFV pharmaco­ is, as yet, largely unstudied. Peroni et al. recently
kinetics and it is possible that dose adjustments reported that EFV is a substrate of rat ABCG2
are applicable for genetically defined subgroups and inhibits its own intestinal permeability by
of women. Interestingly, two women included promoting overexpression of ABCG2 in the rat
in the study above experienced extremely high GI tract [118] . ABCG2, which is highly polymor-
drug exposure during pregnancy [108] . This may phic, has been shown to be highly expressed in the
be explained by genetic polymorphisms, but the apical membrane of human placenta syncytiotro-
consequences of such high exposures remain phoblast [119] and human mammary gland during
unknown. lactation [120] . It has been suggested to play a role
EFV is primarily metabolized by CYP2B6 [109] in protecting the fetus against exposure to poten-
with a minor contribution from CYP2A6 [110,111] tially toxic compounds in maternal blood [121] .
and glucuronidation of the 8-hydroxy metabo- SNPs in EFV transporters are expected to play
lite by UGT2B7 [112] . Genetic polymorphisms in an important role in determining the exposure of
genes encoding these enzymes and the transcrip- the fetus and breastfed infant-to-maternal EFV.
tional regulator, CAR, have been associated with Etravirine (ETR) was approved by the FDA
interindividual variability in EFV plasma drug in 2008 [204] . Data on ETR pharmaco­k inetics
concentrations. The SNPs CYP2B6 516G>T during pregnancy are limited; only a small study
(rs3745274), CYP2B6 983T>C (rs28399499), involving four HIV-infected pregnant women
CYP2A6 1836G>T (rs8192726), UGT2B7 treated with ETR (with darunavir [DRV] and
735A>G (rs28365062), UGT2B7 802C>T ritonavir [RTV]) describes the pharmaco­kinetics
(rs7439366) and CAR 540C>T (rs2307424) of ETR during pregnancy [122] . ETR pharmaco­
have been investigated. In studies conducted in kinetics during the third trimester was similar
HIV-positive Ghanaian patients, Kwara et al. to those of nonpregnant adults (Table  4) . C:M
identified CYP2B6 516G>T, CYP2A6 1836G>T concentration ratio in one woman was 0.33.
and/or CYP2A6 1093G>A (rs28399454) and More data are clearly required and several ongo-
UGT2B7*1a carrier status as predictors of EFV ing studies are assessing the pharmaco­k inetics
plasma concentrations, accounting for 45.2, of ETR during pregnancy. ETR is primarily
10.1 and 8.6% of the total variance, respectively metabolized by CYP2C19 [123] , whose expres-
[113,114] . Rotger et al. also reported significantly sion and activity declines during pregnancy
higher plasma and intracellular EFV concentra- [124] . It is also known to be highly polymorphic.
tions and AUC in Caucasians with the CYP2B6 The CYP2C19*2 (rs4244285; high frequency
516TT genotype than in those with CYP2B6 in Asians) and CYP2C19*17 (rs12248560; high
516GG genotype [115] . CYP2B6 516TT was frequency in Africans) alleles have been associ-
also associated with higher incidences of EFV ated with poor and ultrarapid metabolism of
neuropsychiatric side effects. The 983T>C poly- CYP2C19 substrates, respectively [125,126] and
morphism was also significantly associated with their effect on ETR pharmaco­kinetics is clearly
higher EFV concentrations [94] . Recently, the worthy of study. Furthermore, how these fac-
CYP2B6 516TT and CAR 540C>T genotypes tors affect ETR pharmaco­kinetics during preg-
were associated with early treatment discontinu- nancy remain uncharacterized but the presence
ation, with CNS toxicity accounting for 94.7% of CYP2C19*17 and known upregulation of
of cases of discontinuation [13] . The effect of the CYP3A4 activity during pregnancy may lead to
well defined SNPs that influence EFV plasma subtherapeutic concentrations.
concentrations and risk of CNS toxicity in Rilpivirine (RPV) is the newest NNRTI
nonpregnant adults should be studied during approved by the FDA in 2011 [204] . It is metabo-
pregnancy. lized by CYP3A enzymes and 99.7% bound to
EFV is excreted into breast milk. Schneider plasma proteins (mainly albumin) [207] . No data
et  al. reported a median infant plasma EFV on RPV pharmaco­k inetics during pregnancy
concentration of 0.87 µg/ml (range: 0.31–1.51 in humans are available. However, pregnancy-
µg/ml) following breast milk ingestion from induced increases in CYP3A enzyme activity
mothers receiving EFV for prevention of MTCT and decrease in albumin concentrations may be
[116] . The recommended therapeutic range is expected to increase RPV clearance.

1512 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
„„ Protease inhibitors SLCO1B1, ABCB1 and ABCC1 on transplacental
There are currently ten PIs available for the treat- transfer of LPV and RTV also warrants further
ment of HIV. The PIs are primarily metabolized study. However, LPV and RTV are undetectable
by CYP enzymes and are highly protein bound in human breast milk [139] .
(Table 1) . Similar to the NNRTIs, these drugs are Atazanavir (ATV) is the only PI that can be
expected to be susceptible to pregnancy-induced administered with or without low-dose RTV.
changes in CYP enzymes and plasma protein The standard dose alone is 400 mg for ARV-naive
fluctuations. patients or, for ATV/RTV, 300/100 mg once
Lopinavir/RTV (LPV/RTV) is one of the daily. ATV is metabolized by CYP3A enzymes
most widely studied ARVs during pregnancy. and is not recommended for administration
LPV is administered in combination with low- without RTV during pregnancy. Conflicting
dose RTV to ‘boost’ its pharmaco­k inetics. The data on ATV/RTV pharmaco­k inetics dur-
standard dose of LPV/RTV in nonpregnant ing pregnancy have been reported. Ripamonti
adults is 400/100 mg twice daily. LPV is primar- et al. reported no significant differences in ATV
ily metabolized by CYP3A4 and CYP3A5 iso- AUC, Cmax and Cmin antepartum compared with
enzymes. RTV is a potent inhibitor of CYP3A4 postpartum [140] . By contrast, Mirochnick et al.
activity and concomitant administration of observed significant differences between ante-
LPV with RTV results in markedly higher LPV and post-partum pharmaco­k inetics with AUC,
concentrations. RTV is also a substrate for both Cmax and Cmin antepartum:postpartum ratios of
CYP3A4 and CYP2D6 [127] . Both LPV and 0.72, 0.47 and 0.58, respectively, without TFV,
RTV are also ABCB1 and ABCC1 substrates and 0.73, 0.61 and 0.67, respectively, with TFV
[128,129] and are highly bound (98%) to plasma (Table 4) [141] . Subsequently, an ATV/RTV dose
proteins. LPV/RTV is the preferred PI regimen of 400/100 mg was assessed during pregnancy
for ART-naive pregnant women. and provided comparable exposure to non­
Stek et al. assessed standard LPV/RTV dos- pregnant adults [142] . C:M ATV concentration
ing during the third trimester and LPV expo- ratio of approximately 0.15 was observed in these
sure was approximately 50% lower compared studies.
with nonpregnant adults [130] . Mean (±standard The CYP3A5*1B (rs28365095) allele has
deviation) LPV C:M concentration ratio was been associated with increased ATV clearance
0.2 (±0.13). Changes in RTV pharmaco­k inetic [143] . No significant differences in ATV AUC or
parameters were not statistically significant. Cmax were observed in CYP3A5 expressors versus
The soft-gel capsule formulation used in this nonexpressors in the presence of RTV, although
study has been replaced with a tablet formula- in a subgroup of non-African–American males
tion, which has been shown to have improved the faster CL/F persisted in CYP3A5 expressors
bio­availability (Table 4) [131] . LPV exposure dur- versus nonexpressors. Also, subjects with one or
ing the third trimester of pregnancy follow- two copies of the wild-type CGC haplotype for
ing standard dosing in Thai women with the ABCB1 (1236C, 2677G and 3435C; rs1128503,
tablet formulation was reduced by 22% [132] . rs2032582 and rs1045642, respectively) had
Similar findings have been reported by Calza slower ATV clearance.
et al. [133] , Ramautarsing et al. [134] and Lambert A polymorphism (63396C>T; rs2472677)
et al. [135] . In American women, a higher dose of within the transcriptional regulator, PXR
the tablet formulation, 600/125 mg, provided (NR1I2) has also been shown to be associated
comparable exposure to that observed in non- with unboosted ATV plasma concentrations [144]
pregnant adults [136] . Debate remains on the and a 17% increase in unboosted ATV clear-
optimal dose of LPV/RTV during pregnancy ance [145] . Furthermore, interaction between
and may differ between populations. Higher CYP3A5 expressor status and PXR 63396C>T
LPV plasma concentrations have been associated (rs2472677) were also recently reported by Kile
with the 521T>C (rs4149056) polymorphism in et  al. [146] . ATV clearance was 37% lower in
SLCO1B1 [137] ; within a population pharmaco­ nonexpressors of CYP3A5 with PXR 63396CC
kinetic model homozygosity for the C allele was genotype than those with CT or TT genotypes
associated with a 37% lower clearance [138] . The and 63% lower in CYP3A5 expressors with PXR
influence of SLCO1B1 polymorphisms on LPV 63396CC genotype than those with CT or TT
exposure during pregnancy should be investi- genotypes  [146] . Based on available data, the
gated as the presence of this SNP may help com- effect of the CYP3A5-expressor status on RTV
pensate for the pregnancy-induced changes in boosted ATV during pregnancy may be limited.
some women. The possible influence of SNPs in However, the ABCB1 3435C>T (rs1045642)

future science group www.futuremedicine.com 1513


Review Olagunju, Owen & Cressey

polymorphism may influence ATV plasma con- A number of reports have emerged on its use in
centrations in the presence of RTV [147] , and this late pregnancy. Taylor et al. [154] and McKeown
has not been investigated in pregnant women. et al. have authored case reports of RAL dur-
AT V use is associated with hyper­ ing pregnancy and observed rapid viral-load
bilirubinemia and an association between the reductions in women within days of initiating
UGT1A1*28 (rs8175347) and hyperbilirubine- a RAL-containing regimens [155] . Preliminary
mia has been observed [148] . Neonatal UGT1A1 results from an intensive pharmaco­kinetic study
211G>A (rs4148323) and SLCO1B1 388G>A of RAL ante- and post-partum showed that it
(rs2306283) polymorphisms, breastfeeding [149] exhibited wide interindividual variability, con-
and, more recently, ATV use during pregnancy sistent with data in nonpregnant adults, and that
[150] have all been identified as risk factors for the exposure was not consistently altered during
neonatal hyperbilirubinemia. Breast milk excre- pregnancy compared with postpartum (Table 4)
tion of ATV (unboosted) has also been reported [156] . C:M RAL concentration ratio at delivery
with a milk:plasma ratio of 0.13 [151] . Studies on was 1.20 (95% CI: 0.09–2.3) suggesting signifi-
the role of these functional SNPs in neonatal cant transfer into the fetal compartment. RAL
hyperbilirubinemia associated with ATV use is a substrate for ABCB1 [30] . The role of SNPs
during pregnancy and lactation are needed. associated with changes in its expression and
DRV was approved in 2006 and is used in activity in RAL transport across the placenta has
combination with low-dose RTV at 600/100 mg not been studied. While elevated RAL levels may
twice daily or 800/100 mg once daily in naive protect against peri­natal HIV infection, the con-
patients. Little is known about the pharmaco­ sequences for exposed infants need to be clarified.
kinetics of DRV during pregnancy. Capparelli Interestingly, higher RAL concentrations have
et al. assessed DRV/RTV once and twice daily been reported in Thai adults with an approxi-
pharmaco­k inetics during the third trimester mately twofold increase in AUC compared with
and postpartum (Table 4) [152] . DRV clearance Caucasians at the standard dose [157] .
was increased during the third trimester com- RAL is metabolized by UGT1A1, which is
pared with postpartum for both once and twice highly polymorphic. Coadministration with
daily dosing. The authors suggested that twice ATV (a known UGT1A1 inhibitor) resulted in
daily dosing may be preferable during preg- elevated RAL plasma levels (median [25th–75th
nancy to reduce the risk of low trough con- percentiles]: 0.52 [0.31–2.71] vs 0.22 [0.95–0.55]
centrations and higher doses of DRV/RTV are µg/ml; p = 0.02). This study also reported the
currently under study. No functional SNPs have lack of an association between elevated RAL
been associated with DRV but the mechanisms plasma concentrations and UGT1A1*28/*28
that regulate its pharmaco­k inetics are similar to (rs8175347) reduced function genotype in Italian
other PIs and many of the issues may therefore patients [158] . An earlier study in Japanese patients
be similar to those for LPV and ATV. also reported no significant difference between
There are no data on the use of amprena- heterozygote *6 (rs4148323) and *28 allele car-
vir during pregnancy. However, its prodrug riers and homozygotes of the common allele [159] .
fosamprenavir boosted with low-dose RTV has In a recent comprehensive population pharmaco­
been assessed at a dose of 700/100 mg twice kinetic/pharmaco­genetic analysis of RAL in
daily and the AUC and C12 during the third healthy and HIV positive individuals, Arab-
trimester were lower than postpartum (Table 4) Alameddine et al. reported no association with
[153] . Fosamprenavir had a C:M concentration UGT1A1 SNPs. However, although extremely
ratio of 0.27 (95% CI: 0.24–0.45). No associa- rare, UGT1A9*3 (rs72551330) was identified as
tion between SNPs and amprenavir pharmaco­ possibly influencing RAL pharmaco­kinetics [160] .
kinetics have been reported. NFV, saquinavir Other factors influencing the wide interpatient
and indinavir are among the older PIs and are variability, fetal exposure, and infant exposure
now used less frequently for HIV treatment through breast milk remain to be determined.
and during pregnancy, and therefore will not
included in this review, but lower NFV and „„ Entry inhibitors/fusion inhibitors
indinavir/RTV exposure during pregnancy has Very limited information is available on enfu-
been observed (Table 4) . virtide during pregnancy. Its use in late preg-
nancy for prevention of MTCT in women pre-
„„Integrase strand transfer inhibitors senting with virological failure and multiclass
Raltegravir (RAL) is the only approved INSTI viral resistance mutations have been reported.
and the recommended dose is 400 mg twice daily. Enfuvirtidedoes not cross the placenta [161–165]

1514 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
and has been associated with a case of MTCT NVP pharmaco­k inetics to identify pharmaco­
despite adequate maternal virological suppression genetic predictors of NVP exposure following
[166] . Only a single case report has described the
breast milk ingestion would be useful.
use of maraviroc during pregnancy with C:M To date, TFV has not been extensively used
ratio of 0.30 [167] . Maraviroc has been shown to during pregnancy. The inclusion of TFV in sev-
be a substrate for OATP1B1 and the 521T>C eral generic fixed-dose combination tablets will
(rs4149056) SNP in SLCO1B1 is associated with lead to a significant increase in its use during
plasma concentrations [168] . pregnancy and breastfeeding over the next few
years. Recent data suggest that its exposure is
Conclusion & future perspective reduced during the third trimester of pregnancy.
Highly efficacious regimens to prevent MTCT The necessity for a dose adjustment during
of HIV are now available. The rationale to use pregnancy is unclear, but studies investigating
pharmaco­genetics to help optimize ARV treat- the expression/activity of TFV drug transport-
ment for pregnant women is the same as in ers (e.g., ABCC4, OAT1, OAT3 and ABCC10)
adults. However, addition considerations apply during pregnancy could help determine if the
given that the maternal ARV exposure can lead reductions observed are driven by genetic poly-
to significant exposure to the fetus, or to new- morphisms. However, such studies should be
borns through breastfeeding. In addition, the interpreted with caution because it is intra­cellular
short duration of ARV use during pregnancy concentrations of the active metabolite that drive
emphasizes the need to rapidly achieve optimal efficacy and not the plasma concentrations of the
drug exposure. While we have shown that there parent drug. LPV/RTV is the preferred PI during
are numerous potential candidates for pharmaco­ pregnancy and has been assessed in diverse popu-
genomics investigation, the very low perinatal lations. LPV exposure is reduced during preg-
transmission rates observed using HAART or nancy, but the higher bioavailability of the new
ZDV plus single-dose NVP makes it unlikely tablet formulation helps to compensate for a por-
that further reductions could be gained through tion of the pregnancy-induced changes observed.
the use of pharmaco­genomics. Nevertheless, the However, the presence of the SLCO1B1 poly-
number of ARVs being used during pregnancy morphisms may also help to compensate for the
and breastfeeding is rapidly increasingly and pregnancy-induced changes in some women and
gaps remain regarding whether the maternal, should be investigated. Limited pharmaco­kinetic
fetal and infant drug exposures within geneti- data during pregnancy is available among the
cally defined subgroups of women are appropri- newer ARVs (RPV, ETR, RAL and maraviroc),
ate. For example, the CYP2B6 516G>T poly- and a better knowledge of the affinity of these
morphism is a major determinant of fetal and drugs for placental and mammary transporters
infant (during breastfeeding) EFV exposure. The is needed. To help answer these questions, there
short- and longer-term tolerability and toxicity is a need for large cohorts to allow for pharmaco­
of higher exposure of EFV within this subgroup genetic analysis as the fetal/infant exposure is
should be considered and dose adjustments may likely to be dependent on multiple mechanisms
be appropriate. in addition to those that regulate exposure in the
Generic fixed-dose tablets containing NVP mother. Overall, pharmaco­genetic studies can
will continue to be widely used in resource-­ contribute to our understanding of ARV expo-
limited settings in the coming years. Preliminary sure during pregnancy and breastfeeding, and
data suggest that incorporating genetic and may be able to help optimize maternal, fetal and
clinical risk factors may have utility in reduc- infant drug exposures within genetically defined
ing the incidence of NVP-related toxicities in subgroups of women.
women initiating NVP during pregnancy and
this model should be assessed prospectively. In Financial & competing interests disclosure
addition, some studies have shown that approxi- The authors have no relevant affiliations or financial involve‑
mately 30% of women receiving NVP during ment with any organization or entity with a financial interest
pregnancy have subtherapeutic trough concentra- in or financial conflict with the subject matter or materials
tions, so it remains important to try and deter- discussed in the manuscript. This includes employment, con‑
mine if host genetic polymorphisms could help sultancies, honoraria, stock ownership or options, expert
identity these women. The number of infants testimony, grants or patents received or pending, or
breastfeeding from mothers receiving NVP- royalties.
based HAART will also continue to increase, No writing assistance was utilized in the production of
and studies of functional SNPs associated with this manuscript.

future science group www.futuremedicine.com 1515


Review Olagunju, Owen & Cressey

Executive summary
Mother-to-child transmission of HIV
ƒƒ Mother-to-child-transmission rates of HIV without any intervention range between 20 and 45%.
ƒƒ Antiretroviral drugs (ARVs) during pregnancy, delivery and through planned weaning by 6 months postpartum can reduce HIV
transmission to less than 2%.
Antiretroviral therapy during pregnancy
ƒƒ Physiological changes during pregnancy can influence drug disposition and several studies have demonstrated reduced ARV exposure
during pregnancy.
ƒƒ Associations between SNPs in genes coding for metabolizing enzymes and/or transporters and ARV disposition are well described;
however, relatively little is known about the influence of these SNPs on ARV pharmacokinetics during pregnancy and lactation, as well
as their effect on distribution into the fetal compartment and on breast milk excretion.
Influence of SNPs on ARVs during pregnancy & breastfeeding
ƒƒ Numerous potential candidates for pharmacogenomic investigation exist.
ƒƒ Preliminary data suggest that incorporating genetic and clinical risk factors may have utility in reducing the incidence of nevirapine
(NVP)-related toxicities in women initiating NVP during pregnancy.
ƒƒ Including studies of functional SNPs associated with NVP pharmacokinetics to identify pharmacogenetic predictors of NVP exposure
following breast milk ingestion would be useful.
ƒƒ The short- and longer-term tolerability and toxicity of higher exposure of efavirenz to the mother, fetus and infant within the CYP2B6
516G>T polymorphism should be considered; dose adjustments may be appropriate.
ƒƒ Studies investigating the expression/activity of tenofovir drug transporters (e.g., ABCC4, OAT1, OAT3 and ABCC10) during pregnancy
could help determine if the reductions observed are driven by genetic polymorphisms.
Conclusion & future perspective
ƒƒ The number of ARVs being used during pregnancy and breastfeeding is rapidly increasingly, and gaps remain regarding whether
maternal, fetal and infant drug exposures within genetically defined subgroups of women are appropriate.
ƒƒ Pharmacogenetic studies can contribute to our understanding of antiretroviral exposure during pregnancy and breastfeeding – it is
important to include genetic analysis within cohort studies and clinical trials of ARVs during pregnancy.

nn Short-course zidovudine plus single-dose pregnant women. Clin. Pharmacokinet.


References 43(15), 1071–1087 (2004).
nevirapine could reduce perinatal HIV
Papers of special note have been highlighted as:
n of interest
transmission to 2% in nonbreastfeeding 10 Cressey TR, Lallemant M.
nn of considerable interest women. Pharmacogenetics of antiretroviral drugs for
5 Warszawski J, Tubiana R, Le Chenadec J the treatment of HIV-infected patients:
1 De Cock KM, Fowler MG, Mercier E
et al. Mother-to-child HIV transmission an update. Infect. Genet. Evol. 7(2), 333–342
et al. Prevention of mother-to-child
despite antiretroviral therapy in the ANRS (2007).
HIV transmission in resource-poor
countries: translating research into policy French Perinatal Cohort. AIDS 22(2), 11 Tozzi V. Pharmacogenetics of antiretrovirals.
and practice. JAMA 283(9), 1175–1182 289–299 (2008). Antiviral Res. 85(1), 190–200 (2010).
(2000). 6 Chasela CS, Hudgens MG, Jamieson DJ 12 Lubomirov R, Colombo S, di Iulio J et al.
2 Connor EM, Sperling RS, Gelber R et al. et al. Maternal or infant antiretroviral drugs Association of pharmacogenetic markers
Reduction of maternal-infant transmission to reduce HIV-1 transmission. N. Engl. J. with premature discontinuation of first-line
of human immunodeficiency virus type 1 Med. 362(24), 2271–2281 (2010). anti-HIV therapy: an observational cohort
with zidovudine treatment. N. Engl. J. Med. 7 de Vincenzi I. Triple antiretroviral compared study. J. Infect. Dis 203(2), 246–257 (2011).
331(18), 1173–1180 (1994). with zidovudine and single-dose nevirapine 13 Wyen C, Hendra H, Siccardi M et al.
nn First study demonstrating the efficacy of prophylaxis during pregnancy and Cytochrome P450 2B6 (CYP2B6 ) and
antiretroviral drugs to prevent mother-to breastfeeding for prevention of mother-to- constitutive androstane receptor (CAR)
child transmission of HIV-1 (Kesho Bora polymorphisms are associated with early
child-transmission of HIV.
study): a randomised controlled trial. Lancet discontinuation of efavirenz-containing
3 Guay LA, Musoke P, Fleming T et al. Infect. Dis. 11(3), 171–180 (2011). regimens. J. Antimicrob. Chemother. 66(9),
Intrapartum and neonatal single-dose
8 Shapiro RL, Hughes MD, Ogwu A et al. 2092–2098 (2011).
nevirapine compared with zidovudine for
Antiretroviral regimens in pregnancy and 14 Dunlop W. Serial changes in renal
prevention of mother-to-child transmission
breast-feeding in Botswana. N. Engl. J. Med. haemodynamics during normal human
of HIV-1 in Kampala, Uganda: HIVNET
362(24), 2282–2294 (2010). pregnancy. Br. J. Obstet. Gynaecol. 88(1),
012 randomised trial. Lancet 354(9181),
795–802 (1999). nn Maternal highly active antiretroviral 1–9 (1981).

4 Lallemant M, Jourdain G, Le Coeur S therapy during pregnancy and breastfeeding 15 Tracy TS, Venkataramanan R, Glover DD
et al. Single-dose perinatal nevirapine plus can reduce HIV transmission to less et al. Temporal changes in drug metabolism
standard zidovudine to prevent mother-to- than 2%. (CYP1A2, CYP2D6 and CYP3A activity)
child transmission of HIV-1 in Thailand. during pregnancy. Am. J. Obstet. Gynecol.
9 Mirochnick M, Capparelli E.
N. Engl. J. Med. 351(3), 217–228 (2004). 192(2), 633–639 (2005).
Pharmacokinetics of antiretrovirals in

1516 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
16 Anderson GD. Pregnancy-induced changes in pharmacokinetics and drug interactions of 42 Duncombe C, Kerr SJ, Liddy J et al. Efficacy
pharmacokinetics: a mechanistic-based HIV protease inhibitors. J. Pharm. Sci. and tolerability of zidovudine 200 mg twice a
approach. Clin. Pharmacokinet. 44(10), 100(9), 3636–3654 (2011). day as part of combination antiretroviral
989–1008 (2005). 30 Moss DM, Kwan WS, Liptrott NJ et al. therapy for 96 weeks. J. Acquir. Immune Defic.
17 Tirona RG, Kim RB. Nuclear receptors and Raltegravir is a substrate for SLC22A6: Syndr. 54(5), e19–e20 (2010).
drug disposition gene regulation. J. Pharm. a putative mechanism for the interaction 43 Anderson PL, Lamba J, Aquilante CL et al.
Sci. 94(6), 1169–1186 (2005). between raltegravir and tenofovir. Antimicrob. Pharmacogenetic characteristics of indinavir,
18 Faucette SR, Sueyoshi T, Smith CM et al. Agents Chemother. 55(2), 879–887 (2011). zidovudine, and lamivudine therapy in
Differential regulation of hepatic CYP2B6 31 Pal D, Kwatra D, Minocha M et al. Efflux HIV-infected adults: a pilot study. J. Acquir.
and CYP3A4 genes by constitutive androstane transporters- and cytochrome P-450-mediated Immune Defic. Syndr. 42(4), 441–449 (2006).
receptor but not pregnane X receptor. interactions between drugs of abuse and 44 Sai Y, Nishimura T, Shimpo S et al.
J. Pharmacol. Exp. Ther. 317(3), 1200–1209 antiretrovirals. Life Sci. 88(21–22), 959–971 Characterization of the mechanism of
(2006). (2011). zidovudine uptake by rat conditionally
19 Fahmi OA, Kish M, Boldt S et al. 32 Shitara Y. Clinical importance of OATP1B1 immortalized syncytiotrophoblast cell line
Cytochrome P450 3A4 mRNA is a more and OATP1B3 in drug–drug interactions. TR-TBT. Pharm. Res. 25(7), 1647–1653
reliable marker than CYP3A4 activity for Drug Metab. Pharmacokinet. 26(3), 220–227 (2008).
detecting pregnane X receptor-activated (2011). 45 Wade NA, Unadkat JD, Huang S et al.
induction of drug-metabolizing enzymes. 33 Kis O, Robillard K, Chan GN et al. The Pharmacokinetics and safety of stavudine in
Drug Metab. Dispos. 38(9), 1605–1611 complexities of antiretroviral drug–drug HIV-infected pregnant women and their
(2010). interactions: role of ABC and SLC transporters. infants: Pediatric AIDS Clinical Trials Group
20 Morris CA, Onyamboko MA, Capparelli E Trends Pharmacol. Sci. 31(1), 22–35 (2010). protocol 332. J. Infect. Dis 190(12),
et al. Population pharmacokinetics of 2167–2174 (2004).
34 Rubinchik-Stern M, Eyal S. Drug interactions
artesunate and dihydroartemisinin in at the human placenta: what is the evidence? 46 Wangsomboonsiri W, Mahasirimongkol S,
pregnant and non-pregnant women with Front. Pharmacol. 3, 126 (2012). Chantarangsu S et al. Association between
malaria. Malar. J. 10, 114 (2011). HLA‑B*4001 and lipodystrophy among
35 Bazzoli C, Jullien V, Le Tiec C et al.
21 Badger TM, Hidestrand M, Shankar K et al. HIV-infected patients from Thailand who
Intracellular pharmacokinetics of antiretroviral
The effects of pregnancy on ethanol received a stavudine-containing antiretroviral
drugs in HIV-infected patients, and their
clearance. Life Sci. 77(17), 2111–2126 (2005). regimen. Clin. Infect. Dis 50(4), 597–604
correlation with drug action. Clin.
(2010).
22 Else LJ, Taylor S, Back DJ et al. Pharmacokinet. 49(1), 17–45 (2010).
Pharmacokinetics of antiretroviral drugs in 47 Best BM, Mirochnick M, Capparelli EV et al.
36 Veal GJ, Back DJ. Metabolism of zidovudine.
anatomical sanctuary sites: the fetal Impact of pregnancy on abacavir
Gen. Pharmacol. 26(7), 1469–1475 (1995).
compartment (placenta and amniotic fluid). pharmacokinetics. AIDS 20(4), 553–560
37 Sperling RS, Roboz J, Dische R et al. (2006).
Antivir. Ther. 16(8), 1139–1147 (2011).
Zidovudine pharmacokinetics during
23 Watts DH. Teratogenicity risk of 48 Hetherington S, McGuirk S, Powell G et al.
pregnancy. Am. J. Perinatol. 9(4), 247–249
antiretroviral therapy in pregnancy. Curr. Hypersensitivity reactions during therapy
(1992).
HIV/AIDS Rep. 4(3), 135–140 (2007). with the nucleoside reverse transcriptase
38 O’Sullivan MJ, Boyer PJ, Scott GB et al. The inhibitor abacavir. Clin. Ther. 23(10),
24 Ross AC, Leong T, Avery A et al. Effects of pharmacokinetics and safety of zidovudine in 1603–1614 (2001).
in utero antiretroviral exposure on the third trimester of pregnancy for women
mitochondrial DNA levels, mitochondrial 49 Mallal S, Nolan D, Witt C et al. Association
infected with human immunodeficiency virus
function and oxidative stress. HIV Med. between presence of HLA‑B*5701, HLA‑DR7,
and their infants: Phase I acquired
13(2), 98–106 (2012). and HLA‑DQ3 and hypersensitivity to HIV-1
immunodeficiency syndrome clinical trials
reverse-transcriptase inhibitor abacavir. Lancet
25 Evseenko D, Paxton JW, Keelan JA. Active group study (protocol 082). Zidovudine
359(9308), 727–732 (2002).
transport across the human placenta: impact Collaborative Working Group. Am. J. Obstet.
on drug efficacy and toxicity. Expert Opin Gynecol. 168(5), 1510–1516 (1993). 50 Mallal S, Phillips E, Carosi G et al.
Drug Metab. Toxicol. 2(1), 51–69 (2006). HLA‑B*5701 screening for hypersensitivity to
39 Moodley J, Moodley D, Pillay K et al.
abacavir. N. Engl. J. Med. 358(6), 568–579
26 Gulati A, Gerk PM. Role of placental Pharmacokinetics and antiretroviral activity of
(2008).
ATP-binding cassette (ABC) transporters in lamivudine alone or when coadministered with
antiretroviral therapy during pregnancy. zidovudine in human immunodeficiency virus nn First study to demonstrate the utility of a
J. Pharm. Sci. 98(7), 2317–2335 (2009). type 1-infected pregnant women and their pharmacogenetic test for the management of
offspring. J. Infect. Dis 178(5), 1327–1333 antiretroviral treatment.
27 Iqbal M, Audette MC, Petropoulos S et al.
Placental drug transporters and their role in (1998). 51 Benaboud S, Treluyer JM, Urien S et al.
fetal protection. Placenta 33(3), 137–142 40 Watts DH, Brown ZA, Tartaglione T et al. Pregnancy-related effects on lamivudine
(2012). Pharmacokinetic disposition of zidovudine pharmacokinetics in a population study with
during pregnancy. J. Infect. Dis 163(2), 228 women. Antimicrob. Agents Chemother.
28 Ieiri I. Functional significance of genetic
226–232 (1991). 56(2), 776–782 (2012).
polymorphisms in P-glycoprotein (MDR1,
ABCB1) and breast cancer resistance protein 41 Cressey TR, Leenasirimakul P, Jourdain G 52 Mandelbrot L, Peytavin G, Firtion G et al.
(BCRP, ABCG2). Drug Metab. et al. Intensive pharmacokinetics of zidovudine Maternal–fetal transfer and amniotic fluid
Pharmacokinet. 27(1), 85–105 (2012). 200 mg twice daily in HIV-1-infected patients accumulation of lamivudine in human
weighing less than 60 kg on highly active immunodeficiency virus-infected pregnant
29 Griffin L, Annaert P, Brouwer KL. Influence
antiretroviral therapy. J. Acquir. Immune Defic. women. Am. J. Obstet. Gynecol. 184(2),
of drug transport proteins on the
Syndr. 42(3), 387–389 (2006). 153–158 (2001).

future science group www.futuremedicine.com 1517


Review Olagunju, Owen & Cressey

53 Shapiro RL, Holland DT, Capparelli E et al. 65 Bousquet L, Pruvost A, Didier N et al. 77 Anthonypillai C, Gibbs JE, Thomas SA. The
Antiretroviral concentrations in breast-feeding Emtricitabine: inhibitor and substrate of distribution of the anti-HIV drug, tenofovir
infants of women in Botswana receiving multidrug resistance associated protein. (PMPA), into the brain, CSF and choroid
antiretroviral treatment. J. Infect. Dis 192(5), Eur. J. Pharm. Sci. 35(4), 247–256 (2008). plexuses. Cerebrospinal Fluid Res. 3, 1 (2006).
720–727 (2005). 66 Kearney BP, Flaherty JF, Shah J. Tenofovir 78 Kiser JJ, Aquilante CL, Anderson PL et al.
54 Mirochnick M, Thomas T, Capparelli E et al. disoproxil fumarate: clinical pharmacology Clinical and genetic determinants of
Antiretroviral concentrations in breast-feeding and pharmacokinetics. Clin. Pharmacokinet. intracellular tenofovir diphosphate
infants of mothers receiving highly active 43(9), 595–612 (2004). concentrations in HIV-infected patients.
antiretroviral therapy. Antimicrob. Agents 67 Burchett S, Best B, Mirochnick M et al. J. Acquir. Immune Defic. Syndr. 47(3),
Chemother. 53(3), 1170–1176 (2009). Tenofovir pharmacokinetics during 298–303 (2008).
55 Jung N, Lehmann C, Rubbert A et al. pregnancy, at delivery and postpartum. 79 Cihlar T, Ho ES, Lin DC et al. Human renal
Relevance of the organic cation transporters 1 Presented at: 14th Conference on Retroviruses organic anion transporter 1 (hOAT1) and its
and 2 for antiretroviral drug therapy in human and Opportunistic Infections. Los Angeles, CA, role in the nephrotoxicity of antiviral
immunodeficiency virus infection. Drug USA, 25–28 February 2007 (Abstract 738b). nucleotide analogs. Nucleosides Nucleotides
Metab. Dispos. 36(8), 1616–1623 (2008). 68 Colbers A, Taylor G, Molto J et al. Nucleic Acids 20(4–7), 641–648 (2001).
56 Minuesa G, Volk C, Molina-Arcas M et al. A comparison of the pharmacokinetics of 80 Mallants R, Van Oosterwyck K, Van Vaeck L
Transport of lamivudine [(-)-beta-L-2´,3´- tenofovir during pregnancy and postpartum. et al. Multidrug resistance-associated protein
dideoxy-3´-thiacytidine] and high-affinity Presented at: 13th International Workshop on 2 (MRP2) affects hepatobiliary elimination
interaction of nucleoside reverse transcriptase Clinical Pharmacology of HIV Therapy. but not the intestinal disposition of tenofovir
inhibitors with human organic cation Barcelona, Spain, 16–18 April 2012. disoproxil fumarate and its metabolites.
transporters 1, 2, and 3. J. Pharmacol. Exp. 69 Benaboud S, Hirt D, Launay O et al. Xenobiotica 35(10–11), 1055–1066 (2005).
Ther. 329(1), 252–261 (2009). Pregnancy-related effects on tenofovir 81 Imaoka T, Kusuhara H, Adachi M et al.
57 Bleasby K, Castle JC, Roberts CJ et al. pharmacokinetics: a population study with Functional involvement of multidrug
Expression profiles of 50 xenobiotic 186 women. Antimicrob. Agents Chemother. resistance-associated protein 4
transporter genes in humans and pre-clinical 56(2), 857–862 (2012). (MRP4/ABCC4) in the renal elimination of
species: a resource for investigations into drug 70 Best BM, Letendre SL, Koopmans P et al. the antiviral drugs adefovir and tenofovir.
disposition. Xenobiotica 36(10–11), 963–988 Low cerebrospinal fluid concentrations of the Mol. Pharmacol. 71(2), 619–627 (2007).
(2006). nucleotide HIV reverse transcriptase 82 Rodriguez-Novoa S, Labarga P, Soriano V
58 Choi MK, Song IS. Genetic variants of organic inhibitor, tenofovir. J. Acquir. Immune Defic. et al. Predictors of kidney tubular
cation transporter 1 (OCT1) and OCT2 Syndr. 59(4), 376–381 (2012). dysfunction in HIV-infected patients treated
significantly reduce lamivudine uptake. 71 Uwai Y, Ida H, Tsuji Y et al. Renal transport with tenofovir: a pharmacogenetic study.
Biopharm. Drug Dispos. 33(3), 170–178 of adefovir, cidofovir, and tenofovir by Clin. Infect. Dis 48(11), e108–e116 (2009).
(2012). SLC22A family members (hOAT1, hOAT3, n Report of an association between SNPs in
59 Blanche S, Tardieu M, Rustin P et al. and hOCT2). Pharm. Res. 24(4), 811–815 drug transporters and kidney tubular
Persistent mitochondrial dysfunction and (2007). dysfunction in HIV-infected adults
perinatal exposure to antiretroviral nucleoside 72 Pushpakom SP, Liptrott NJ, Rodriguez- receiving tenofovir.
analogues. Lancet 354(9184), 1084–1089 Novoa S et al. Genetic variants of ABCC10, a 83 Rodriguez-Novoa S, Labarga P, D’Avolio A
(1999). novel tenofovir transporter, are associated et al. Impairment in kidney tubular function
60 Divi RL, Walker VE, Wade NA et al. with kidney tubular dysfunction. J. Infect. Dis in patients receiving tenofovir is associated
Mitochondrial damage and DNA depletion in 204(1), 145–153 (2011). with higher tenofovir plasma concentrations.
cord blood and umbilical cord from infants 73 Alebouyeh M, Takeda M, Onozato ML et al. AIDS 24(7), 1064–1066 (2010).
exposed in utero to combivir. AIDS 18(7), Expression of human organic anion 84 Viramune ® Tablets, Viramune ® Oral
1013–1021 (2004). transporters in the choroid plexus and their Suspension, package insert. Boehringer-
61 Kallianpur AR, Hulgan T. Pharmacogenetics interactions with neurotransmitter Ingelheim Pharmaceuticals Inc., CT, USA.
of nucleoside reverse-transcriptase inhibitor- metabolites. J. Pharmacol. Sci. 93(4),
85 Cressey TR, Jourdain G, Lallemant MJ et al.
associated peripheral neuropathy. 430–436 (2003).
Persistence of nevirapine exposure during the
Pharmacogenomics 10(4), 623–637 (2009). 74 Nahata MC, Brady MT. Pharmacokinetics of postpartum period after intrapartum
62 Canter JA, Robbins GK, Selph D et al. African fluconazole after oral administration in single-dose nevirapine in addition to
mitochondrial DNA subhaplogroups and children with human immunodeficiency virus zidovudine prophylaxis for the prevention of
peripheral neuropathy during antiretroviral infection. Eur. J Clin Pharmacol. 48(3–4), mother-to-child transmission of HIV-1.
therapy. J. Infect. Dis 201(11), 1703–1707 291–293 (1995). J. Acquir. Immune Defic. Syndr. 38(3),
(2010). 75 Leggas M, Adachi M, Scheffer GL et al. Mrp4 283–288 (2005).
63 Stek AM, Best BM, Luo W et al. Effect of confers resistance to topotecan and protects 86 Jourdain G, Ngo-Giang-Huong N, Le Coeur
pregnancy on emtricitabine pharmacokinetics. the brain from chemotherapy. Mol. Cell. Biol. S et al. Intrapartum exposure to nevirapine
HIV Med. 13(4), 226–235 (2011). 24(17), 7612–7621 (2004). and subsequent maternal responses to
64 Benaboud S, Pruvost A, Coffie PA et al. 76 Gibbs JE, Thomas SA. The distribution of the nevirapine-based antiretroviral therapy.
Concentrations of tenofovir and emtricitabine anti-HIV drug, 2´3´-dideoxycytidine (ddC), N. Engl. J. Med. 351(3), 229–240 (2004).
in breast milk of HIV-1-infected women in across the blood–brain and blood– 87 McIntyre JA, Hopley M, Moodley D et al.
Abidjan, Cote d’Ivoire, in the ANRS 12109 cerebrospinal fluid barriers and the influence Efficacy of short-course AZT plus 3TC to
TEmAA Study, Step 2. Antimicrob. Agents of organic anion transport inhibitors. reduce nevirapine resistance in the prevention
Chemother. 55(3), 1315–1317 (2011). J. Neurochem. 80(3), 392–404 (2002). of mother-to-child HIV transmission:

1518 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
a randomized clinical trial. PLoS Med. 6(10), 98 Phanuphak N, Apornpong T, J. Acquir. Immune Defic. Syndr. 59(3),
e1000172 (2009). Teeratakulpisarn S et al. Nevirapine- 245–252 (2012).
88 Van Dyke RB, Ngo-Giang-Huong N, Shapiro associated toxicity in HIV-infected Thai men nn First study reporting the pharmacokinetics
DE et al. A comparison of 3 regimens to and women, including pregnant women. HIV
of efavirenz during pregnancy.
prevent nevirapine resistance mutations in Med. 8(6), 357–366 (2007).
109 Ward BA, Gorski JC, Jones DR et al. The
HIV-infected pregnant women receiving a 99 Chantarangsu S, Mushiroda T,
cytochrome P450 2B6 (CYP2B6) is the main
single intrapartum dose of nevirapine. Clin. Mahasirimongkol S et al. HLA‑B*3505 allele
catalyst of efavirenz primary and secondary
Infect. Dis 54(2), 285–293 (2012). is a strong predictor for nevirapine-induced
metabolism: implication for HIV/AIDS
89 Chantarangsu S, Cressey TR, skin adverse drug reactions in HIV-infected
therapy and utility of efavirenz as a substrate
Mahasirimongkol S et al. Influence of Thai patients. Pharmacogenet. Genomics
marker of CYP2B6 catalytic activity.
CYP2B6 polymorphisms on the persistence of 19(2), 139–146 (2009).
J. Pharmacol. Exp. Ther. 306(1), 287–300
plasma nevirapine concentrations following a n Initial data on an association between host (2003).
single intra-partum dose for the prevention of genetics and nevirapine-induced skin rashes. 110 di Iulio J, Fayet A, Arab-Alameddine M et al.
mother to child transmission in HIV-infected
100 Chantarangsu S, Mushiroda T, In vivo ana­lysis of efavirenz metabolism in
Thai women. J. Antimicrob. Chemother.
Mahasirimongkol S et al. Genome-wide individuals with impaired CYP2A6 function.
64(6), 1265–1273 (2009).
association study identifies variations in Pharmacogenet. Genomics 19(4), 300–309
90 Capparelli EV, Aweeka F, Hitti J et al. 6p21.3 associated with nevirapine-induced (2009).
Chronic administration of nevirapine during rash. Clin. Infect. Dis 53(4), 341–348 (2011). 111 Ogburn ET, Jones DR, Masters AR et al.
pregnancy: impact of pregnancy on
101 Ellis JC, L’homme RF, Ewings FM et al. Efavirenz primary and secondary metabolism
pharmacokinetics. HIV Med. 9(4), 214–220
Nevirapine concentrations in HIV-infected in vitro and in vivo: identification of novel
(2008).
children treated with divided fixed-dose metabolic pathways and cytochrome P450
91 Lamorde M, Byakika-Kibwika P, Okaba- combination antiretroviral tablets in Malawi 2A6 as the principal catalyst of efavirenz
Kayom V et al. Suboptimal nevirapine and Zambia. Antivir. Ther. 12(2), 253–260 7-hydroxylation. Drug Metab. Dispos. 38(7),
steady-state pharmacokinetics during (2007). 1218–1229 (2010).
intrapartum compared with postpartum in
102 Minniear TD, Zeh C, Polle N et al. Rash, 112 Bae SK, Jeong YJ, Lee C et al. Identification
HIV-1-seropositive Ugandan women.
hepatotoxicity and hyperbilirubinemia among of human UGT isoforms responsible for
J. Acquir. Immune Defic. Syndr. 55(3),
kenyan infants born to HIV-infected women glucuronidation of efavirenz and its three
345–350 (2010).
receiving triple-antiretroviral drugs for the hydroxy metabolites. Xenobiotica 41(6),
92 von Hentig N, Carlebach A, Gute P et al. prevention of mother-to-child HIV 437–444 (2011).
A comparison of the steady-state transmission. Pediatr. Infect. Dis. J. 113 Kwara A, Lartey M, Sagoe KW et al.
pharmacokinetics of nevirapine in men, doi:10.1097/INF.0b013e318267ef6a (2012) CYP2B6, CYP2A6 and UGT2B7 genetic
nonpregnant women and women in late (Epub ahead of print). polymorphisms are predictors of efavirenz
pregnancy. Br. J. Clin. Pharmacol. 62(5),
103 Fundaro C, Genovese O, Rendeli C et al. mid-dose concentration in HIV-infected
552–559 (2006).
Myelomeningocele in a child with patients. AIDS 23(16), 2101–2106 (2009).
93 Penzak SR, Kabuye G, Mugyenyi P et al. intrauterine exposure to efavirenz. AIDS 114 Kwara A, Lartey M, Sagoe KW et al. CYP2B6
Cytochrome P450 2B6 (CYP2B6 ) G516T 16(2), 299–300 (2002). (c.516G–>T) and CYP2A6 (*9B and/or *17)
influences nevirapine plasma concentrations
104 Bussmann H, Wester CW, Wester CN et al. polymorphisms are independent predictors of
in HIV-infected patients in Uganda. HIV
Pregnancy rates and birth outcomes among efavirenz plasma concentrations in HIV-
Med. 8(2), 86–91 (2007).
women on efavirenz-containing highly active infected patients. Br. J. Clin. Pharmacol.
94 Wyen C, Hendra H, Vogel M et al. Impact of antiretroviral therapy in Botswana. J. Acquir. 67(4), 427–436 (2009).
CYP2B6 983T>C polymorphism on Immune Defic. Syndr. 45(3), 269–273 (2007). 115 Rotger M, Colombo S, Furrer H et al.
non-nucleoside reverse transcriptase inhibitor
105 Bera E, McCausland K, Nonkwelo R et al. Influence of CYP2B6 polymorphism on
plasma concentrations in HIV-infected
Birth defects following exposure to plasma and intracellular concentrations and
patients. J. Antimicrob. Chemother. 61(4),
efavirenz-based antiretroviral therapy during toxicity of efavirenz and nevirapine in
914–918 (2008).
pregnancy: a study at a regional South HIV-infected patients. Pharmacogenet.
95 Brown KC, Hosseinipour MC, Hoskins JM African hospital. AIDS 24(2), 283–289 Genomics 15(1), 1–5 (2005).
et al. Exploration of CYP450 and drug (2010). 116 Schneider S, Peltier A, Gras A et al. Efavirenz
transporter genotypes and correlations with
106 Ford N, Mofenson L, Kranzer K et al. Safety in human breast milk, mothers’, and
nevirapine exposure in Malawians.
of efavirenz in first-trimester of pregnancy: newborns’ plasma. J. Acquir. Immune Defic.
Pharmacogenomics 13(1), 113–121 (2012).
a systematic review and meta-analysis of Syndr. 48(4), 450–454 (2008).
96 Liptrott NJ, Pushpakom S, Wyen C et al. outcomes from observational cohorts. AIDS 117 Marzolini C, Telenti A, Decosterd LA et al.
Association of ABCC10 polymorphisms with 24(10), 1461–1470 (2010). Efavirenz plasma levels can predict treatment
nevirapine plasma concentrations in the
107 Ekouevi DK, Coffie PA, Ouattara E et al. failure and central nervous system side effects
German Competence Network for HIV/
Pregnancy outcomes in women exposed to in HIV-1-infected patients. AIDS 15(1),
AIDS. Pharmacogenet. Genomics 22(1), 10–19
efavirenz and nevirapine: an appraisal of the 71–75 (2001).
(2012).
IeDEA west Africa and ANRS databases, 118 Peroni RN, Di Gennaro SS, Hocht C et al.
97 Hitti J, Frenkel LM, Stek AM et al. Maternal Abidjan, Cote d’Ivoire. J. Acquir. Immune Efavirenz is a substrate and in turn modulates
toxicity with continuous nevirapine in Defic. Syndr. 56(2), 183–187 (2011). the expression of the efflux transporter
pregnancy: results from PACTG 1022.
108 Cressey TR, Stek A, Capparelli E et al. ABCG2/BCRP in the gastrointestinal tract of
J. Acquir. Immune Defic. Syndr. 36(3),
Efavirenz pharmacokinetics during the third the rat. Biochem. Pharmacol. 82(9),
772–776 (2004).
trimester of pregnancy and postpartum. 1227–1233 (2011).

future science group www.futuremedicine.com 1519


Review Olagunju, Owen & Cressey

119 Maliepaard M, Scheffer GL, Faneyte IF pregnancy. AIDS 20(15), 1931–1939 141 Mirochnick M, Best BM, Stek AM et al.
et al. Subcellular localization and (2006). Atazanavir pharmacokinetics with and
distribution of the breast cancer resistance nn Significantly lower lopinavir exposure was without tenofovir during pregnancy. J. Acquir.
protein transporter in normal human tissues. Immune Defic. Syndr. 56(5), 412–419 (2011).
observed in pregnant women during the
Cancer Res. 61(8), 3458–3464 (2001). 142 Mirochnick M, Stek A, Capparelli E et al.
third trimester of pregnancy.
120 Jonker JW, Merino G, Musters S et al. The Pharmacokinetics of increased dose atazanavir
131 Else LJ, Douglas M, Dickinson L et al.
breast cancer resistance protein BCRP with and without tenofovir during pregnancy.
Improved oral bioavailability of lopinavir in
(ABCG2) concentrates drugs and Presented at: 12th International Workshop on
melt-extruded tablet formulation reduces
carcinogenic xenotoxins into milk. Nat. Clinical Pharmacology of HIV Therapy.
impact of third trimester on lopinavir plasma
Med. 11(2), 127–129 (2005). Miami, FL, USA, 13–15 April 2011.
concentrations. Antimicrob. Agents Chemother.
121 Hahnova-Cygalova L, Ceckova M, Staud F. 56(2), 816–824 (2012). 143 Anderson PL, Aquilante CL, Gardner EM
Fetoprotective activity of breast cancer et al. Atazanavir pharmacokinetics in
132 Cressey TR, Jourdain G, Rawangban B et al.
resistance protein (BCRP, ABCG2): genetically determined CYP3A5 expressors
Pharmacokinetics and virologic response of
expression and function throughout versus non-expressors. J. Antimicrob.
zidovudine/lopinavir/ritonavir initiated
pregnancy. Drug Metab. Rev. 43(1), 53–68 Chemother. 64(5), 1071–1079 (2009).
during the third trimester of pregnancy. AIDS
(2011). 144 Siccardi M, D’Avolio A, Baietto L et al.
24(14), 2193–2200 (2010).
122 Izurieta P, Kakuda TN, Feys C et al. Safety Association of a single-nucleotide
133 Calza L, Manfredi R, Trapani F et al.
and pharmacokinetics of etravirine in polymorphism in the pregnane X receptor
Lopinavir/ritonavir trough concentrations
pregnant HIV-1-infected women. HIV Med. (PXR 63396C–>T) with reduced
with the tablet formulation in HIV-1-infected
12(4), 257–258 (2011). concentrations of unboosted atazanavir. Clin.
women during the third trimester of
123 Yanakakis LJ, Bumpus NN. Infect. Dis 47(9), 1222–1225 (2008).
pregnancy. Scand. J. Infect. Dis 44(5),
Biotransformation of the antiretroviral drug 381–387 (2012). 145 Schipani A, Siccardi M, D’Avolio A et al.
etravirine: metabolite identification, reaction Population pharmacokinetic modeling of the
134 Ramautarsing RA, van der Lugt J, Gorowara
phenotyping, and characterization of association between 63396C->T pregnane X
M et al. Thai HIV-1-infected women do not
autoinduction of cytochrome P450- receptor polymorphism and unboosted
require a dose increase of lopinavir/ritonavir
dependent metabolism. Drug Metab. Dispos. atazanavir clearance. Antimicrob. Agents
during the third trimester of pregnancy. AIDS
40(4), 803–814 (2012). Chemother. 54(12), 5242–5250 (2010).
25(10), 1299–1303 (2011).
124 McGready R, Stepniewska K, Seaton E et al. 146 Kile DA, Mawhinney S, Aquilante CL et al.
135 Lambert JS, Else LJ, Jackson V et al.
Pregnancy and use of oral contraceptives A population pharmacokinetic-
Therapeutic drug monitoring of lopinavir/
reduces the biotransformation of proguanil pharmacogenetic analysis of atazanavir. AIDS
ritonavir in pregnancy. HIV Med. 12(3),
to cycloguanil. Eur. J. Clin. Pharmacol. Res. Hum. Retroviruses doi:10.1089/
166–173 (2011).
59(7), 553–557 (2003). aid.2011.0378 (2012) (Epub ahead of print).
136 Best BM, Stek AM, Mirochnick M et al.
125 Mega JL, Close SL, Wiviott SD et al. 147 Rodriguez-Novoa S, Martin-Carbonero L,
Lopinavir tablet pharmacokinetics with an
Cytochrome P-450 polymorphisms and Barreiro P et al. Genetic factors influencing
increased dose during pregnancy. J. Acquir.
response to clopidogrel. N. Engl. J. Med. atazanavir plasma concentrations and the risk
Immune Defic. Syndr. 54(4), 381–388 (2010).
360(4), 354–362 (2009). of severe hyperbilirubinemia. AIDS 21(1),
137 Hartkoorn RC, Kwan WS, Shallcross V et al. 41–46 (2007).
126 Sibbing D, Koch W, Gebhard D et al.
HIV protease inhibitors are substrates for
Cytochrome 2C19*17 allelic variant, platelet 148 Rotger M, Taffe P, Bleiber G et al. Gilbert
OATP1A2, OATP1B1 and OATP1B3 and
aggregation, bleeding events, and stent syndrome and the development of
lopinavir plasma concentrations are influenced
thrombosis in clopidogrel-treated patients antiretroviral therapy-associated
by SLCO1B1 polymorphisms. Pharmacogenet.
with coronary stent placement. Circulation hyperbilirubinemia. J. Infect. Dis 192(8),
Genomics 20(2), 112–120 (2010).
121(4), 512–518 (2010). 1381–1386 (2005).
138 Schipani A, Egan D, Dickinson L et al.
127 Kumar GN, Rodrigues AD, Buko AM et al. 149 Huang MJ, Kua KE, Teng HC et al. Risk
Estimation of the effect of SLCO1B1
Cytochrome P450-mediated metabolism of factors for severe hyperbilirubinemia in
polymorphisms on lopinavir plasma
the HIV-1 protease inhibitor ritonavir neonates. Pediatric Res. 56(5), 682–689
concentration in HIV-infected adults. Antivir.
(ABT-538) in human liver microsomes. (2004).
Ther. 17(5), 861–868 (2012).
J. Pharmacol. Exp. Ther. 277(1), 423–431 150 Mandelbrot L, Mazy F, Floch-Tudal C et al.
(1996). n Association between drug transporters
Atazanavir in pregnancy: impact on neonatal
polymorphisms and lopinavir plasma
128 van der Sandt IC, Vos CM, Nabulsi L et al. hyperbilirubinemia. Eur. J. Obstet. Gynecol.
Assessment of active transport of HIV concentrations in adults. Reprod. Biol. 157(1), 18–21 (2011).
protease inhibitors in various cell lines and 139 Rezk NL, White N, Bridges AS et al. Studies
151 Spencer L, Neely M, Mordwinkin N et al.
the in vitro blood–brain barrier. AIDS 15(4), on antiretroviral drug concentrations in breast Intensive pharmacokinetics of zidovudine,
483–491 (2001). milk: validation of a liquid chromatography- lamivudine, and atazanavir and HIV-1 viral
129 Janneh O, Bray PG, Jones E et al.
tandem mass spectrometric method for the load in breast milk and plasma in HIV+
Concentration-dependent effects and determination of 7 anti-human women receiving HAART. Presented at: 16th
intracellular accumulation of HIV protease immunodeficiency virus medications. Ther. Conference on Retroviruses and Opportunistic
inhibitors in cultured CD4 T cells and Drug. Monit. 30(5), 611–619 (2008). Infections (CROI). Montréal, Canada,
primary human lymphocytes. J. Antimicrob. 140 Ripamonti D, Cattaneo D, Maggiolo F et al. 8–11 February 2009 (Abstract 942).
Chemother. 65(5), 906–916 (2010). Atazanavir plus low-dose ritonavir in 152 Capparelli E, Best B, Stek A et al.
130 Stek AM, Mirochnick M, Capparelli E
pregnancy: pharmacokinetics and placental Pharmacokinetics of darunavir once or twice
et al. Reduced lopinavir exposure during transfer. AIDS 21(18), 2409–2415 (2007). daily during and after pregnancy. Presented

1520 Pharmacogenomics (2012) 13(13) future science group


Pharmacogenetics & antiretroviral drug exposure during pregnancy & breastfeeding Review
at: 3rd International Workshop on HIV 164 Madeddu G, Calia GM, Campus ML et al. 176 Ciccacci C, Borgiani P, Ceffa S et al.
Pediatrics. Rome, Italy, 15–16 July 2011 Successful prevention of multidrug resistant Nevirapine-induced hepatotoxicity and
(Abstract P_72). HIV mother-to-child transmission with pharmacogenetics: a retrospective study in a
153 Capparelli E, Stek A, Best B et al. Boosted enfuvirtide use in late pregnancy. Int. J. STD population from Mozambique.
fosamprenavir pharmacokinetics during AIDS. 19(9), 644–645 (2008). Pharmacogenomics 11(1), 23–31 (2012).
pregnancy. Presented at: 17th Conference on 165 Furco A, Gosrani B, Nicholas S et al. 177 Ni Z, Mao Q. ATP-binding cassette efflux
Retroviruses and Opportunistic Infections Successful use of darunavir, etravirine, transporters in human placenta. Curr.
(CROI). San Francisco, CA, USA, enfuvirtide and tenofovir/emtricitabine in Pharm. Biotechnol. 12(4), 674–685 (2011).
16–19 February 2010. pregnant woman with multiclass HIV 178 Ellis L, Pirmohamed M, Owen A. Transport
154 Taylor N, Touzeau V, Geit M et al. resistance. AIDS 23(3), 434–435 (2009). of antiretroviral drugs by ABCB5: the elusive
Raltegravir in pregnancy: a case series 166 Cohan D, Feakins C, Wara D et al. Perinatal non-nucleoside transcriptase inhibitor efflux
presentation. Int. J. STD AIDS 22(6), transmission of multidrug-resistant HIV-1 pump? Presented at: 12th International
358–360 (2011). despite viral suppression on an enfuvirtide- Workshop on Clinical Pharmacology of HIV
155 McKeown DA, Rosenvinge M, Donaghy S based treatment regimen. AIDS 19(9), Therapy. Miami, FL, USA, 13–15 April 2011.
et al. High neonatal concentrations of 989–990 (2005). 179 Lee CG, Gottesman MM, Cardarelli CO
raltegravir following transplacental transfer in 167 Vincent J, Ahmed T, Eric L et al. Favourable et al. HIV-1 protease inhibitors are substrates
HIV-1 positive pregnant women. AIDS outcome of a pregnancy with a maraviroc- for the MDR1 multidrug transporter.
24(15), 2416–2418 (2010). containing regimen. J. AIDS Clinic. Res. 2(6), Biochemistry 37(11), 3594–3601 (1998).
156 Best BM, Capparelli E, Stek A et al. (2011). 180 Huisman MT, Smit JW, Crommentuyn KM
Raltegravir pharmacokinetics during 168 Siccardi M, D’Avolio A, Nozza S et al. et al. Multidrug resistance protein 2 (MRP2)
pregnancy. Presented at: 50th Interscience Maraviroc is a substrate for OATP1B1 in vitro transports HIV protease inhibitors, and
Conference on Antimicrobial Agents and and maraviroc plasma concentrations are transport can be enhanced by other drugs.
Chemotherapy (ICAAC). Boston, MA, USA, influenced by SLCO1B1 521 T>C AIDS 16(17), 2295–2301 (2002).
12–15 September 2010 (Abstract H-1668a). polymorphism. Pharmacogenet. Genomics 181 Jedlitschky G, Burchell B, Keppler D.
157 Ananworanich J, Gorowara M, Avihingsanon 20(12), 759–765 (2010). The multidrug resistance protein 5 functions
A et al. Pharmacokinetics of and short-term 169 Chappuy H, Treluyer JM, Rey E et al. as an ATP-dependent export pump for cyclic
virologic response to low-dose 400-milligram Maternal–fetal transfer and amniotic fluid nucleotides. J. Biol. Chem. 275(39),
once-daily raltegravir maintenance therapy. accumulation of protease inhibitors in 30069–30074 (2000).
Antimicrob. Agents Chemother. 56(4), pregnant women who are infected with 182 Moitra K, Scally M, McGee K et al.
1892–1898 (2012). human immunodeficiency virus. Am. J. Molecular evolutionary ana­lysis of ABCB5:
158 Siccardi M, D’Avolio A, Rodriguez-Novoa S Obstet. Gynecol. 191(2), 558–562 (2004). the ancestral gene is a full transporter with
et al. Intrapatient and interpatient 170 Flynn PM, Mirochnick M, Shapiro DE et al. potentially deleterious single nucleotide
pharmacokinetic variability of raltegravir in Pharmacokinetics and safety of single-dose polymorphisms. PloS ONE 6(1), e16318
the clinical setting. Ther. Drug Monit. 34(2), tenofovir disoproxil fumarate and (2011).
232–235 (2012). emtricitabine in HIV-1-infected pregnant 183 Wang H, Yan Z, Dong M et al. Alteration in
159 Hirano A, Ikemura K, Takahashi M et al. women and their infants. Antimicrob. Agents placental expression of bile acids transporters
Lack of correlation between UGT1A1*6, *28 Chemother. 55(12), 5914–5922 (2011). OATP1A2, OATP1B1, OATP1B3 in
genotypes, and plasma raltegravir 171 Musoke P, Guay LA, Bagenda D et al. intrahepatic cholestasis of pregnancy. Arch.
concentrations in Japanese HIV type A Phase I/II study of the safety and Gynecol. Obstet. 285(6), 1535–1540 (2011).
1-infected patients. AIDS Res. Hum. pharmacokinetics of nevirapine in HIV-1- 184 Kohler JJ, Hosseini SH, Green E et al.
Retroviruses 28(8), 776–779 (2011). infected pregnant Ugandan women and their Tenofovir renal proximal tubular toxicity is
160 Arab-Alameddine M, Fayet-Mello A, neonates (HIVNET 006). AIDS 13(4), regulated by OAT1 and MRP4 transporters.
Lubomirov R et al. Population 479–486 (1999). Lab. Invest. 91(6), 852–858 (2011).
pharmacokinetic analysis and 172 van Heeswijk RP, Khaliq Y, Gallicano KD 185 Xu G, Bhatnagar V, Wen G et al. Analyses of
pharmacogenetics of raltegravir in HIV- et al. The pharmacokinetics of nelfinavir and coding region polymorphisms in apical and
positive and healthy individuals. Antimicrob. M8 during pregnancy and post partum. Clin. basolateral human organic anion transporter
Agents Chemother. 56(6), 2959–2966 (2012). Pharmacol. Ther. 76(6), 588–597 (2004). (OAT) genes [OAT1 (NKT ), OAT2, OAT3,
161 Weizsaecker K, Kurowski M, Hoffmeister B 173 Winters MA, Van Rompay KK, Kashuba AD OAT4, URAT (RST )]. Kidney Int. 68(4),
et al. Pharmacokinetic profile in late et al. Maternal–fetal pharmacokinetics and 1491–1499 (2005).
pregnancy and cord blood concentration of dynamics of a single intrapartum dose of 186 Ugele B, St-Pierre MV, Pihusch M et al.
tipranavir and enfuvirtide. Int. J. STD AIDS. maraviroc in rhesus macaques. Antimicrob. Characterization and identification of steroid
22(5), 294–295 (2011). Agents Chemother. 54(10), 4059–4063 (2010). sulfate transporters of human placenta.
162 Wensing AM, Boucher CA, van Kasteren M 174 Brennan-Benson P, Pakianathan M, Rice P Am. J. Physiol. Endocrinol. Metab. 284(2),
et al. Prevention of mother-to-child et al. Enfurvitide prevents vertical e390–e398 (2003).
transmission of drug resistant HIV-1 using transmission of multidrug-resistant HIV-1 in 187 Ugele B, Bahn A, Rex-Haffner M.
maternal therapy with both enfuvirtide and pregnancy but does not cross the placenta. Functional differences in steroid sulfate
tipranavir. AIDS 20(10), 1465–1467 (2006). AIDS 20(2), 297–299 (2006). uptake of organic anion transporter 4
163 Sued O, Lattner J, Gun A et al. Use of 175 Ripamonti D, Cattaneo D, Cortinovis M (OAT4) and organic anion transporting
darunavir and enfuvirtide in a pregnant et al. Transplacental passage of ritonavir- polypeptide 2B1 (OATP2B1) in human
woman. Int. J. STD AIDS. 19(12), 866–867 boosted darunavir in two pregnant women. placenta. J. Steroid Biochem. Mol. Biol.
(2008). Int. J. STD AIDS 20(3), 215–216 (2009). 111(1–2), 1–6 (2008).

future science group www.futuremedicine.com 1521


Review Olagunju, Owen & Cressey

188 Alcorn J, Lu X, Moscow JA et al. Transporter 193 Read JS, Best BM, Stek AM et al. infection in infants. Recommendations for a
gene expression in lactating and nonlactating Pharmacokinetics of new 625 mg nelfinavir public health approach.
human mammary epithelial cells using formulation during pregnancy and postpartum. www.who.int/hiv/pub/mtct/
real-time reverse transcription-polymerase HIV Med. 9(10), 875–882 (2008). antiretroviral2010/en/index.html
chain reaction. J. Pharmacol. Exp. Ther. 194 Martinez-Rebollar M, Lonca M, Perez I et al. 204 US FDA. Antiretroviral drugs used in the
303(2), 487–496 (2002). Pharmacokinetic study of saquinavir 500 mg treatment of HIV infection.
189 Nies AT, Koepsell H, Winter S et al. Expression plus ritonavir (1000/100 mg twice a day) in www.fda.gov/ForConsumers/byAudience/
of organic cation transporters OCT1 HIV-positive pregnant women. Ther. Drug ForPatientAdvocates/HIVandAIDSActivities/
(SLC22A1) and OCT3 (SLC22A3) is affected Monit. 33(6), 772–777 (2011). ucm118915.htm
by genetic factors and cholestasis in human (Accessed 14 June 2012)
liver. Hepatology 50(4), 1227–1240 (2009). 205 Panel on Antiretroviral Therapy and Medical
Websites
„„
190 Saito J, Hirota T, Kikunaga N et al. Management of HIV-Infected Children.
201 WHO. PMTCT strategic vision 2010–2015.
Interindividual differences in placental Guidelines for the Use of Antiretroviral
Preventing mother-to-child transmission of
expression of the SLC22A2 (OCT2) gene: Agents in Pediatric HIV Infection.
HIV to reach the UNGASS and Millennium
relationship to epigenetic variations in the http://aidsinfo.nih.gov/contentfiles/
Development Goals.
5´-upstream regulatory region. J. Pharm. Sci. lvguidelines/PediatricGuidelines.pdf
www.who.int/hiv/pub/mtct/strategic_vision/
100(9), 3875–3883 (2011). (Accessed 22 May 2012)
en/index.html
191 Sata R, Ohtani H, Tsujimoto M et al. (Accessed 12 June 2012) 206 WHO. Antiretroviral therapy for HIV
Functional analysis of organic cation infection in adults and adolescents:
202 WHO. Antiretroviral Therapy for HIV
transporter 3 expressed in human placenta. recommendations for a public health
Infection in Infants and Children: Towards
J. Pharmacol. Exp. Ther. 315(2), 888–895 approach.
Universal Access. Recommendations for a
(2005). www.who.int/hiv/pub/arv/adult2010/en/
public health approach.
192 Cressey T, Best BM, Achalapong J et al. Effect index.html
www.who.int/hiv/pub/paediatric/infants2010/
of pregnancy on pharmacokinetics of indinavir en/index.html 207 Gilead Sciences. Complera. Prescribing
boosted with ritonavir. Presented at: 13th (Accessed 12 June 2012) information. Foster City, CA, USA.
International Workshop on Clinical www.gilead.com/pdf/complera_pi.pdf
203 WHO. Antiretroviral drugs for treating
Pharmacology of HIV Therapy. Barcelona, (Accessed 20 May 2012)
pregnant women and preventing HIV
Spain, 16–18 April 2012 (Abstract P_37).

1522 Pharmacogenomics (2012) 13(13) future science group

Vous aimerez peut-être aussi