Olagunju 2012
Olagunju 2012
Olagunju 2012
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
[email protected]
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 .
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
TFV (0.26–1.95)
SQV (0.00–0.06) [170] ABCC1
[169]
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
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.
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]
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]
OAT1(SLC22A6) Mammary gland TFV, RAL and ZDV R50H (↑ affinity for substrates) [30,184,185]
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
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
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
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.
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]
Executive summary
Mother-to-child transmission of HIV
Mother-to-child-transmission rates of HIV without any intervention range between 20 and 45%.
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
Physiological changes during pregnancy can influence drug disposition and several studies have demonstrated reduced ARV exposure
during pregnancy.
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
Numerous potential candidates for pharmacogenomic investigation exist.
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.
Including studies of functional SNPs associated with NVP pharmacokinetics to identify pharmacogenetic predictors of NVP exposure
following breast milk ingestion would be useful.
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.
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
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.
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.
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
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:
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
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).