메뉴 건너뛰기




Volumn 13, Issue 13, 2012, Pages 1501-1522

Potential effect of pharmacogenetics on maternal, fetal and infant antiretroviral drug exposure during pregnancy and breastfeeding

Author keywords

antiretrovirals; drug transporters; highly active antiretroviral therapy; HIV; pregnancy; prevention of mother to child transmission

Indexed keywords

ABACAVIR; AMPRENAVIR; ANTIRETROVIRUS AGENT; ATAZANAVIR; DARUNAVIR PLUS RITONAVIR; DELAVIRDINE; DIDANOSINE; EFAVIRENZ; EMTRICITABINE; ETRAVIRINE; FOSAMPRENAVIR; INDINAVIR; INTEGRASE INHIBITOR; LAMIVUDINE; LOPINAVIR PLUS RITONAVIR; MARAVIROC; NELFINAVIR; NEVIRAPINE; NONNUCLEOSIDE REVERSE TRANSCRIPTASE INHIBITOR; PROTEINASE INHIBITOR; RALTEGRAVIR; RILPIVIRINE; RITONAVIR; RNA DIRECTED DNA POLYMERASE INHIBITOR; SAQUINAVIR; STAVUDINE; TENOFOVIR DISOPROXIL; TIPRANAVIR; UNINDEXED DRUG; ZIDOVUDINE;

EID: 84867567023     PISSN: 14622416     EISSN: 17448042     Source Type: Journal    
DOI: 10.2217/pgs.12.138     Document Type: Review
Times cited : (22)

References (194)
  • 1
    • 0034161410 scopus 로고    scopus 로고
    • Prevention of mother-to-child HIV transmission in resource-poor countries: Translating research into policy and practice
    • De Cock KM, Fowler MG, Mercier E, et al. Prevention of mother-to-child HIV transmission in resource-poor countries: translating research into policy and practice. JAMA 283(9), 1175-1182 (2000)
    • (2000) JAMA , vol.283 , Issue.9 , pp. 1175-1182
    • De Cock, K.M.1    Fowler, M.G.2    Mercier, E.3
  • 2
    • 0028003766 scopus 로고
    • Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment
    • Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment. N. Engl. J. Med. 331(18), 1173-1180 (1994)
    • (1994) N. Engl. J. Med. , vol.331 , Issue.18 , pp. 1173-1180
    • Connor, E.M.1    Sperling, R.S.2    Gelber, R.3
  • 3
    • 0033523454 scopus 로고    scopus 로고
    • Intrapartum and neonatal single-dose nevirapine compared with zidovudine for prevention of mother-to-child transmission of HIV-1 in Kampala, Uganda: HIVNET 012 randomised trial
    • Guay LA, Musoke P, Fleming T, et al. Intrapartum and neonatal single-dose nevirapine compared with zidovudine for prevention of mother-to-child transmission of HIV-1 in Kampala, Uganda: HIVNET 012 randomised trial. Lancet 354(9181), 795-802 (1999)
    • (1999) Lancet , vol.354 , Issue.9181 , pp. 795-802
    • Guay, L.A.1    Musoke, P.2    Fleming, T.3
  • 4
    • 3142656082 scopus 로고    scopus 로고
    • Single-dose perinatal nevirapine plus standard zidovudine to prevent mother-to-child transmission of HIV-1 in Thailand
    • Lallemant M, Jourdain G, Le Coeur S, et al. Single-dose perinatal nevirapine plus standard zidovudine to prevent mother-to-child transmission of HIV-1 in Thailand. N. Engl. J. Med. 351(3), 217-228 (2004)
    • (2004) N. Engl. J. Med. , vol.351 , Issue.3 , pp. 217-228
    • Lallemant, M.1    Jourdain, G.2    Le Coeur, S.3
  • 5
    • 37549048161 scopus 로고    scopus 로고
    • Mother-to-child HIV transmission despite antiretroviral therapy in the ANRS French Perinatal Cohort
    • Warszawski J, Tubiana R, Le Chenadec J, et al. Mother-to-child HIV transmission despite antiretroviral therapy in the ANRS French Perinatal Cohort. AIDS 22(2), 289-299 (2008)
    • (2008) AIDS , vol.22 , Issue.2 , pp. 289-299
    • Warszawski, J.1    Tubiana, R.2    Le Chenadec, J.3
  • 6
    • 77953723667 scopus 로고    scopus 로고
    • Maternal or infant antiretroviral drugs to reduce HIV-1 transmission
    • Chasela CS, Hudgens MG, Jamieson DJ, et al. Maternal or infant antiretroviral drugs to reduce HIV-1 transmission. N. Engl. J. Med. 362(24), 2271-2281 (2010)
    • (2010) N. Engl. J. Med. , vol.362 , Issue.24 , pp. 2271-2281
    • Chasela, C.S.1    Hudgens, M.G.2    Jamieson, D.J.3
  • 7
    • 79952042500 scopus 로고    scopus 로고
    • Triple antiretroviral compared with zidovudine and single-dose nevirapine prophylaxis during pregnancy and breastfeeding for prevention of mother-to-child transmission of HIV-1 (Kesho Bora study): A randomised controlled trial
    • de Vincenzi I. Triple antiretroviral compared with zidovudine and single-dose nevirapine prophylaxis during pregnancy and breastfeeding for prevention of mother-to-child transmission of HIV-1 (Kesho Bora study): a randomised controlled trial. Lancet Infect. Dis. 11(3), 171-180 (2011)
    • (2011) Lancet Infect. Dis. , vol.11 , Issue.3 , pp. 171-180
    • De Vincenzi, I.1
  • 8
    • 77953725982 scopus 로고    scopus 로고
    • Antiretroviral regimens in pregnancy and breast-feeding in Botswana
    • Shapiro RL, Hughes MD, Ogwu A, et al. Antiretroviral regimens in pregnancy and breast-feeding in Botswana. N. Engl. J. Med. 362(24), 2282-2294 (2010)
    • (2010) N. Engl. J. Med. , vol.362 , Issue.24 , pp. 2282-2294
    • Shapiro, R.L.1    Hughes, M.D.2    Ogwu, A.3
  • 9
    • 10944246142 scopus 로고    scopus 로고
    • Pharmacokinetics of antiretrovirals in pregnant women
    • Mirochnick M, Capparelli E. Pharmacokinetics of antiretrovirals in pregnant women. Clin. Pharmacokinet. 43(15), 1071-1087 (2004)
    • (2004) Clin. Pharmacokinet. , vol.43 , Issue.15 , pp. 1071-1087
    • Mirochnick, M.1    Capparelli, E.2
  • 10
    • 33846681647 scopus 로고    scopus 로고
    • Pharmacogenetics of antiretroviral drugs for the treatment of HIV-infected patients: An update
    • Cressey TR, Lallemant M. Pharmacogenetics of antiretroviral drugs for the treatment of HIV-infected patients: an update. Infect. Genet. Evol. 7(2), 333-342 (2007)
    • (2007) Infect. Genet. Evol. , vol.7 , Issue.2 , pp. 333-342
    • Cressey, T.R.1    Lallemant, M.2
  • 11
    • 73549101090 scopus 로고    scopus 로고
    • Pharmacogenetics of antiretrovirals
    • Tozzi V. Pharmacogenetics of antiretrovirals. Antiviral Res. 85(1), 190-200 (2010)
    • (2010) Antiviral Res. , vol.85 , Issue.1 , pp. 190-200
    • Tozzi, V.1
  • 12
    • 79851482408 scopus 로고    scopus 로고
    • Association of pharmacogenetic markers with premature discontinuation of first-line anti-HIV therapy: An observational cohort study
    • Lubomirov R, Colombo S, di Iulio J, et al. Association of pharmacogenetic markers with premature discontinuation of first-line anti-HIV therapy: an observational cohort study. J. Infect. Dis 203(2), 246-257 (2011)
    • (2011) J. Infect. Dis , vol.203 , Issue.2 , pp. 246-257
    • Lubomirov, R.1    Colombo, S.2    Di Iulio, J.3
  • 13
    • 80051685348 scopus 로고    scopus 로고
    • Cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) polymorphisms are associated with early discontinuation of efavirenz-containing regimens
    • Wyen C, Hendra H, Siccardi M, et al. Cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) polymorphisms are associated with early discontinuation of efavirenz-containing regimens. J. Antimicrob. Chemother. 66(9), 2092-2098 (2011)
    • (2011) J. Antimicrob. Chemother. , vol.66 , Issue.9 , pp. 2092-2098
    • Wyen, C.1    Hendra, H.2    Siccardi, M.3
  • 14
    • 0019381759 scopus 로고
    • Serial changes in renal haemodynamics during normal human pregnancy
    • Dunlop W. Serial changes in renal haemodynamics during normal human pregnancy. Br. J. Obstet. Gynaecol. 88(1), 1-9 (1981)
    • (1981) Br. J. Obstet. Gynaecol. , vol.88 , Issue.1 , pp. 1-9
    • Dunlop, W.1
  • 15
    • 13444269471 scopus 로고    scopus 로고
    • Temporal changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A activity) during pregnancy
    • Tracy TS, Venkataramanan R, Glover DD, et al. Temporal changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A activity) during pregnancy. Am. J. Obstet. Gynecol. 192(2), 633-639 (2005)
    • (2005) Am. J. Obstet. Gynecol. , vol.192 , Issue.2 , pp. 633-639
    • Tracy, T.S.1    Venkataramanan, R.2    Glover, D.D.3
  • 16
    • 25144516296 scopus 로고    scopus 로고
    • Pregnancy-induced changes in pharmacokinetics: A mechanistic-based approach
    • Anderson GD. Pregnancy-induced changes in pharmacokinetics: a mechanistic-based approach. Clin. Pharmacokinet. 44(10), 989-1008 (2005)
    • (2005) Clin. Pharmacokinet. , vol.44 , Issue.10 , pp. 989-1008
    • Anderson, G.D.1
  • 17
    • 21344439074 scopus 로고    scopus 로고
    • Nuclear receptors and drug disposition gene regulation
    • Tirona RG, Kim RB. Nuclear receptors and drug disposition gene regulation. J. Pharm. Sci. 94(6), 1169-1186 (2005)
    • (2005) J. Pharm. Sci. , vol.94 , Issue.6 , pp. 1169-1186
    • Tirona, R.G.1    Kim, R.B.2
  • 18
    • 33646786799 scopus 로고    scopus 로고
    • Differential regulation of hepatic CYP2B6 and CYP3A4 genes by constitutive androstane receptor but not pregnane X receptor
    • Faucette SR, Sueyoshi T, Smith CM, et al. Differential regulation of hepatic CYP2B6 and CYP3A4 genes by constitutive androstane receptor but not pregnane X receptor. J. Pharmacol. Exp. Ther. 317(3), 1200-1209 (2006)
    • (2006) J. Pharmacol. Exp. Ther. , vol.317 , Issue.3 , pp. 1200-1209
    • Faucette, S.R.1    Sueyoshi, T.2    Smith, C.M.3
  • 19
    • 77955999532 scopus 로고    scopus 로고
    • Cytochrome P450 3A4 mRNA is a more reliable marker than CYP3A4 activity for detecting pregnane X receptor-activated induction of drug-metabolizing enzymes
    • Fahmi OA, Kish M, Boldt S, et al. Cytochrome P450 3A4 mRNA is a more reliable marker than CYP3A4 activity for detecting pregnane X receptor-activated induction of drug-metabolizing enzymes. Drug Metab. Dispos. 38(9), 1605-1611 (2010)
    • (2010) Drug Metab. Dispos. , vol.38 , Issue.9 , pp. 1605-1611
    • Fahmi, O.A.1    Kish, M.2    Boldt, S.3
  • 20
    • 79955652478 scopus 로고    scopus 로고
    • Population pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant women with malaria
    • Morris CA, Onyamboko MA, Capparelli E, et al. Population pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant women with malaria. Malar. J. 10, 114 (2011)
    • (2011) Malar. J. , vol.10 , pp. 114
    • Morris, C.A.1    Onyamboko, M.A.2    Capparelli, E.3
  • 21
    • 23744493080 scopus 로고    scopus 로고
    • The effects of pregnancy on ethanol clearance
    • Badger TM, Hidestrand M, Shankar K, et al. The effects of pregnancy on ethanol clearance. Life Sci. 77(17), 2111-2126 (2005)
    • (2005) Life Sci. , vol.77 , Issue.17 , pp. 2111-2126
    • Badger, T.M.1    Hidestrand, M.2    Shankar, K.3
  • 22
    • 83455206864 scopus 로고    scopus 로고
    • Pharmacokinetics of antiretroviral drugs in anatomical sanctuary sites: The fetal compartment (placenta and amniotic fluid)
    • Else LJ, Taylor S, Back DJ, et al. Pharmacokinetics of antiretroviral drugs in anatomical sanctuary sites: the fetal compartment (placenta and amniotic fluid). Antivir. Ther. 16(8), 1139-1147 (2011)
    • (2011) Antivir. Ther. , vol.16 , Issue.8 , pp. 1139-1147
    • Else, L.J.1    Taylor, S.2    Back, D.J.3
  • 23
    • 34848869951 scopus 로고    scopus 로고
    • Teratogenicity risk of antiretroviral therapy in pregnancy
    • Watts DH. Teratogenicity risk of antiretroviral therapy in pregnancy. Curr. HIV/AIDS Rep. 4(3), 135-140 (2007)
    • (2007) Curr. HIV/AIDS Rep. , vol.4 , Issue.3 , pp. 135-140
    • Watts, D.H.1
  • 24
    • 84855353782 scopus 로고    scopus 로고
    • Effects of in utero antiretroviral exposure on mitochondrial DNA levels, mitochondrial function and oxidative stress
    • Ross AC, Leong T, Avery A, et al. Effects of in utero antiretroviral exposure on mitochondrial DNA levels, mitochondrial function and oxidative stress. HIV Med. 13(2), 98-106 (2012)
    • (2012) HIV Med. , vol.13 , Issue.2 , pp. 98-106
    • Ross, A.C.1    Leong, T.2    Avery, A.3
  • 25
    • 33846207538 scopus 로고    scopus 로고
    • Active transport across the human placenta: Impact on drug efficacy and toxicity
    • Evseenko D, Paxton JW, Keelan JA. Active transport across the human placenta: impact on drug efficacy and toxicity. Expert Opin Drug Metab. Toxicol. 2(1), 51-69 (2006)
    • (2006) Expert Opin Drug Metab. Toxicol. , vol.2 , Issue.1 , pp. 51-69
    • Evseenko, D.1    Paxton, J.W.2    Keelan, J.A.3
  • 26
    • 67650099989 scopus 로고    scopus 로고
    • Role of placental ATP-binding cassette (ABC) transporters in antiretroviral therapy during pregnancy
    • Gulati A, Gerk PM. Role of placental ATP-binding cassette (ABC) transporters in antiretroviral therapy during pregnancy. J. Pharm. Sci. 98(7), 2317-2335 (2009)
    • (2009) J. Pharm. Sci. , vol.98 , Issue.7 , pp. 2317-2335
    • Gulati, A.1    Gerk, P.M.2
  • 27
    • 84856972106 scopus 로고    scopus 로고
    • Placental drug transporters and their role in fetal protection
    • Iqbal M, Audette MC, Petropoulos S, et al. Placental drug transporters and their role in fetal protection. Placenta 33(3), 137-142 (2012)
    • (2012) Placenta , vol.33 , Issue.3 , pp. 137-142
    • Iqbal, M.1    Audette, M.C.2    Petropoulos, S.3
  • 28
    • 84857768340 scopus 로고    scopus 로고
    • Functional significance of genetic polymorphisms in P-glycoprotein (MDR1, ABCB1) and breast cancer resistance protein (BCRP, ABCG2)
    • Ieiri I. Functional significance of genetic polymorphisms in P-glycoprotein (MDR1, ABCB1) and breast cancer resistance protein (BCRP, ABCG2). Drug Metab. Pharmacokinet. 27(1), 85-105 (2012)
    • (2012) Drug Metab. Pharmacokinet. , vol.27 , Issue.1 , pp. 85-105
    • Ieiri, I.1
  • 29
    • 79960133743 scopus 로고    scopus 로고
    • Influence of drug transport proteins on the pharmacokinetics and drug interactions of HIV protease inhibitors
    • Griffin L, Annaert P, Brouwer KL. Influence of drug transport proteins on the pharmacokinetics and drug interactions of HIV protease inhibitors. J. Pharm. Sci. 100(9), 3636-3654 (2011)
    • (2011) J. Pharm. Sci. , vol.100 , Issue.9 , pp. 3636-3654
    • Griffin, L.1    Annaert, P.2    Brouwer, K.L.3
  • 30
    • 78751692552 scopus 로고    scopus 로고
    • Raltegravir is a substrate for SLC22A6: A putative mechanism for the interaction between raltegravir and tenofovir
    • Moss DM, Kwan WS, Liptrott NJ, et al. Raltegravir is a substrate for SLC22A6: a putative mechanism for the interaction between raltegravir and tenofovir. Antimicrob. Agents Chemother. 55(2), 879-887 (2011)
    • (2011) Antimicrob. Agents Chemother. , vol.55 , Issue.2 , pp. 879-887
    • Moss, D.M.1    Kwan, W.S.2    Liptrott, N.J.3
  • 31
    • 79956190810 scopus 로고    scopus 로고
    • Efflux transporters- and cytochrome P-450-mediated interactions between drugs of abuse and antiretrovirals
    • Pal D, Kwatra D, Minocha M, et al. Efflux transporters- and cytochrome P-450-mediated interactions between drugs of abuse and antiretrovirals. Life Sci. 88(21-22), 959-971 (2011)
    • (2011) Life Sci. , vol.88 , Issue.21-22 , pp. 959-971
    • Pal, D.1    Kwatra, D.2    Minocha, M.3
  • 32
    • 79959935821 scopus 로고    scopus 로고
    • Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions
    • Shitara Y. Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions. Drug Metab. Pharmacokinet. 26(3), 220-227 (2011)
    • (2011) Drug Metab. Pharmacokinet. , vol.26 , Issue.3 , pp. 220-227
    • Shitara, Y.1
  • 33
    • 72749106653 scopus 로고    scopus 로고
    • The complexities of antiretroviral drug-drug interactions: Role of ABC and SLC transporters
    • Kis O, Robillard K, Chan GN, et al. The complexities of antiretroviral drug-drug interactions: role of ABC and SLC transporters. Trends Pharmacol. Sci. 31(1), 22-35 (2010)
    • (2010) Trends Pharmacol. Sci. , vol.31 , Issue.1 , pp. 22-35
    • Kis, O.1    Robillard, K.2    Chan, G.N.3
  • 34
    • 84866163353 scopus 로고    scopus 로고
    • Drug interactions at the human placenta: What is the evidence?
    • Rubinchik-Stern M, Eyal S. Drug interactions at the human placenta: what is the evidence?. Front. Pharmacol. 3, 126 (2012)
    • (2012) Front. Pharmacol. , vol.3 , pp. 126
    • Rubinchik-Stern, M.1    Eyal, S.2
  • 35
    • 72249123176 scopus 로고    scopus 로고
    • Intracellular pharmacokinetics of antiretroviral drugs in HIV-infected patients, and their correlation with drug action
    • Bazzoli C, Jullien V, Le Tiec C, et al. Intracellular pharmacokinetics of antiretroviral drugs in HIV-infected patients, and their correlation with drug action. Clin. Pharmacokinet. 49(1), 17-45 (2010)
    • (2010) Clin. Pharmacokinet. , vol.49 , Issue.1 , pp. 17-45
    • Bazzoli, C.1    Jullien, V.2    Le Tiec, C.3
  • 36
    • 0029591968 scopus 로고
    • Metabolism of zidovudine
    • Veal GJ, Back DJ. Metabolism of zidovudine. Gen. Pharmacol. 26(7), 1469-1475 (1995)
    • (1995) Gen. Pharmacol. , vol.26 , Issue.7 , pp. 1469-1475
    • Veal, G.J.1    Back, D.J.2
  • 37
    • 0026711120 scopus 로고
    • Zidovudine pharmacokinetics during pregnancy
    • Sperling RS, Roboz J, Dische R, et al. Zidovudine pharmacokinetics during pregnancy. Am. J. Perinatol. 9(4), 247-249 (1992)
    • (1992) Am. J. Perinatol. , vol.9 , Issue.4 , pp. 247-249
    • Sperling, R.S.1    Roboz, J.2    Dische, R.3
  • 38
    • 0027243089 scopus 로고
    • The pharmacokinetics and safety of zidovudine in the third trimester of pregnancy for women infected with human immunodeficiency virus and their infants: Phase I acquired immunodeficiency syndrome clinical trials group study (protocol 082). Zidovudine Collaborative Working Group
    • O'Sullivan MJ, Boyer PJ, Scott GB, et al. The pharmacokinetics and safety of zidovudine in the third trimester of pregnancy for women infected with human immunodeficiency virus and their infants: Phase I acquired immunodeficiency syndrome clinical trials group study (protocol 082). Zidovudine Collaborative Working Group. Am. J. Obstet. Gynecol. 168(5), 1510-1516 (1993)
    • (1993) Am. J. Obstet. Gynecol. , vol.168 , Issue.5 , pp. 1510-1516
    • O'sullivan, M.J.1    Boyer, P.J.2    Scott, G.B.3
  • 39
    • 0031723646 scopus 로고    scopus 로고
    • Pharmacokinetics and antiretroviral activity of lamivudine alone or when coadministered with zidovudine in human immunodeficiency virus type 1-infected pregnant women and their offspring
    • Moodley J, Moodley D, Pillay K, et al. Pharmacokinetics and antiretroviral activity of lamivudine alone or when coadministered with zidovudine in human immunodeficiency virus type 1-infected pregnant women and their offspring. J. Infect. Dis 178(5), 1327-1333 (1998)
    • (1998) J. Infect. Dis , vol.178 , Issue.5 , pp. 1327-1333
    • Moodley, J.1    Moodley, D.2    Pillay, K.3
  • 40
    • 0026031856 scopus 로고
    • Pharmacokinetic disposition of zidovudine during pregnancy
    • Watts DH, Brown ZA, Tartaglione T, et al. Pharmacokinetic disposition of zidovudine during pregnancy. J. Infect. Dis 163(2), 226-232 (1991)
    • (1991) J. Infect. Dis , vol.163 , Issue.2 , pp. 226-232
    • Watts, D.H.1    Brown, Z.A.2    Tartaglione, T.3
  • 41
    • 33745910844 scopus 로고    scopus 로고
    • Intensive pharmacokinetics of zidovudine 200 mg twice daily in HIV-1-infected patients weighing less than 60 kg on highly active antiretroviral therapy
    • Cressey TR, Leenasirimakul P, Jourdain G, et al. Intensive pharmacokinetics of zidovudine 200 mg twice daily in HIV-1-infected patients weighing less than 60 kg on highly active antiretroviral therapy. J. Acquir. Immune Defic. Syndr. 42(3), 387-389 (2006)
    • (2006) J. Acquir. Immune Defic. Syndr. , vol.42 , Issue.3 , pp. 387-389
    • Cressey, T.R.1    Leenasirimakul, P.2    Jourdain, G.3
  • 42
    • 77955004081 scopus 로고    scopus 로고
    • Efficacy and tolerability of zidovudine 200 mg twice a day as part of combination antiretroviral therapy for 96 weeks
    • Duncombe C, Kerr SJ, Liddy J, et al. Efficacy and tolerability of zidovudine 200 mg twice a day as part of combination antiretroviral therapy for 96 weeks. J. Acquir. Immune Defic. Syndr. 54(5), e19-e20 (2010)
    • (2010) J. Acquir. Immune Defic. Syndr. , vol.54 , Issue.5
    • Duncombe, C.1    Kerr, S.J.2    Liddy, J.3
  • 43
    • 33745929357 scopus 로고    scopus 로고
    • Pharmacogenetic characteristics of indinavir, zidovudine, and lamivudine therapy in HIV-infected adults: A pilot study
    • Anderson PL, Lamba J, Aquilante CL, et al. Pharmacogenetic characteristics of indinavir, zidovudine, and lamivudine therapy in HIV-infected adults: a pilot study. J. Acquir. Immune Defic. Syndr. 42(4), 441-449 (2006)
    • (2006) J. Acquir. Immune Defic. Syndr. , vol.42 , Issue.4 , pp. 441-449
    • Anderson, P.L.1    Lamba, J.2    Aquilante, C.L.3
  • 44
    • 44749089831 scopus 로고    scopus 로고
    • Characterization of the mechanism of zidovudine uptake by rat conditionally immortalized syncytiotrophoblast cell line TR-TBT
    • Sai Y, Nishimura T, Shimpo S, et al. Characterization of the mechanism of zidovudine uptake by rat conditionally immortalized syncytiotrophoblast cell line TR-TBT. Pharm. Res. 25(7), 1647-1653 (2008)
    • (2008) Pharm. Res. , vol.25 , Issue.7 , pp. 1647-1653
    • Sai, Y.1    Nishimura, T.2    Shimpo, S.3
  • 45
    • 10444276711 scopus 로고    scopus 로고
    • Pharmacokinetics and safety of stavudine in HIV-infected pregnant women and their infants: Pediatric AIDS Clinical Trials Group protocol 332
    • Wade NA, Unadkat JD, Huang S, et al. Pharmacokinetics and safety of stavudine in HIV-infected pregnant women and their infants: Pediatric AIDS Clinical Trials Group protocol 332. J. Infect. Dis 190(12), 2167-2174 (2004)
    • (2004) J. Infect. Dis , vol.190 , Issue.12 , pp. 2167-2174
    • Wade, N.A.1    Unadkat, J.D.2    Huang, S.3
  • 46
    • 75749110161 scopus 로고    scopus 로고
    • Association between HLA?B*4001 and lipodystrophy among HIV-infected patients from Thailand who received a stavudine-containing antiretroviral regimen
    • Wangsomboonsiri W, Mahasirimongkol S, Chantarangsu S, et al. Association between HLA?B, 4001 and lipodystrophy among HIV-infected patients from Thailand who received a stavudine-containing antiretroviral regimen. Clin. Infect. Dis 50(4), 597-604 (2010)
    • (2010) Clin. Infect. Dis , vol.50 , Issue.4 , pp. 597-604
    • Wangsomboonsiri, W.1    Mahasirimongkol, S.2    Chantarangsu, S.3
  • 47
    • 33645391801 scopus 로고    scopus 로고
    • Impact of pregnancy on abacavir pharmacokinetics
    • Best BM, Mirochnick M, Capparelli EV, et al. Impact of pregnancy on abacavir pharmacokinetics. AIDS 20(4), 553-560 (2006)
    • (2006) AIDS , vol.20 , Issue.4 , pp. 553-560
    • Best, B.M.1    Mirochnick, M.2    Capparelli, E.V.3
  • 48
    • 0035163638 scopus 로고    scopus 로고
    • Hypersensitivity reactions during therapy with the nucleoside reverse transcriptase inhibitor abacavir
    • Hetherington S, McGuirk S, Powell G, et al. Hypersensitivity reactions during therapy with the nucleoside reverse transcriptase inhibitor abacavir. Clin. Ther. 23(10), 1603-1614 (2001)
    • (2001) Clin. Ther. , vol.23 , Issue.10 , pp. 1603-1614
    • Hetherington, S.1    McGuirk, S.2    Powell, G.3
  • 49
    • 0037006623 scopus 로고    scopus 로고
    • Association between presence of HLAB*5701, HLA?DR7, and HLA?DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir
    • Mallal S, Nolan D, Witt C, et al. Association between presence of HLA?B, 5701, HLA?DR7, and HLA?DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 359(9308), 727-732 (2002)
    • (2002) Lancet , vol.359 , Issue.9308 , pp. 727-732
    • Mallal, S.1    Nolan, D.2    Witt, C.3
  • 50
    • 38949196447 scopus 로고    scopus 로고
    • HLA?B*5701 screening for hypersensitivity to abacavir
    • Mallal S, Phillips E, Carosi G, et al. HLA?B*5701 screening for hypersensitivity to abacavir. N. Engl. J. Med. 358(6), 568-579 (2008)
    • (2008) N. Engl. J. Med. , vol.358 , Issue.6 , pp. 568-579
    • Mallal, S.1    Phillips, E.2    Carosi, G.3
  • 51
    • 84856075437 scopus 로고    scopus 로고
    • Pregnancy-related effects on lamivudine pharmacokinetics in a population study with 228 women
    • Benaboud S, Treluyer JM, Urien S, et al. Pregnancy-related effects on lamivudine pharmacokinetics in a population study with 228 women. Antimicrob. Agents Chemother. 56(2), 776-782 (2012)
    • (2012) Antimicrob. Agents Chemother. , vol.56 , Issue.2 , pp. 776-782
    • Benaboud, S.1    Treluyer, J.M.2    Urien, S.3
  • 52
    • 0035124720 scopus 로고    scopus 로고
    • Maternal-fetal transfer and amniotic fluid accumulation of lamivudine in human immunodeficiency virus-infected pregnant women
    • Mandelbrot L, Peytavin G, Firtion G, et al. Maternal-fetal transfer and amniotic fluid accumulation of lamivudine in human immunodeficiency virus-infected pregnant women. Am. J. Obstet. Gynecol. 184(2), 153-158 (2001)
    • (2001) Am. J. Obstet. Gynecol. , vol.184 , Issue.2 , pp. 153-158
    • Mandelbrot, L.1    Peytavin, G.2    Firtion, G.3
  • 53
    • 23944505278 scopus 로고    scopus 로고
    • Antiretroviral concentrations in breast-feeding infants of women in Botswana receiving antiretroviral treatment
    • Shapiro RL, Holland DT, Capparelli E, et al. Antiretroviral concentrations in breast-feeding infants of women in Botswana receiving antiretroviral treatment. J. Infect. Dis 192(5), 720-727 (2005)
    • (2005) J. Infect. Dis , vol.192 , Issue.5 , pp. 720-727
    • Shapiro, R.L.1    Holland, D.T.2    Capparelli, E.3
  • 54
    • 62949168283 scopus 로고    scopus 로고
    • Antiretroviral concentrations in breast-feeding infants of mothers receiving highly active antiretroviral therapy
    • Mirochnick M, Thomas T, Capparelli E, et al. Antiretroviral concentrations in breast-feeding infants of mothers receiving highly active antiretroviral therapy. Antimicrob. Agents Chemother. 53(3), 1170-1176 (2009)
    • (2009) Antimicrob. Agents Chemother. , vol.53 , Issue.3 , pp. 1170-1176
    • Mirochnick, M.1    Thomas, T.2    Capparelli, E.3
  • 55
    • 47949132723 scopus 로고    scopus 로고
    • Relevance of the organic cation transporters 1 and 2 for antiretroviral drug therapy in human immunodeficiency virus infection
    • Jung N, Lehmann C, Rubbert A, et al. Relevance of the organic cation transporters 1 and 2 for antiretroviral drug therapy in human immunodeficiency virus infection. Drug Metab. Dispos. 36(8), 1616-1623 (2008)
    • (2008) Drug Metab. Dispos. , vol.36 , Issue.8 , pp. 1616-1623
    • Jung, N.1    Lehmann, C.2    Rubbert, A.3
  • 56
    • 63849083391 scopus 로고    scopus 로고
    • Transport of lamivudine [(-)-beta-L-2,3-dideoxy-3-thiacytidine] and high-affinity interaction of nucleoside reverse transcriptase inhibitors with human organic cation transporters 1, 2, and 3
    • Minuesa G, Volk C, Molina-Arcas M, et al. Transport of lamivudine [(-)-beta-L-2,3-dideoxy-3-thiacytidine] and high-affinity interaction of nucleoside reverse transcriptase inhibitors with human organic cation transporters 1, 2, and 3. J. Pharmacol. Exp. Ther. 329(1), 252-261 (2009)
    • (2009) J. Pharmacol. Exp. Ther. , vol.329 , Issue.1 , pp. 252-261
    • Minuesa, G.1    Volk, C.2    Molina-Arcas, M.3
  • 57
    • 33751578245 scopus 로고    scopus 로고
    • Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: A resource for investigations into drug disposition
    • Bleasby K, Castle JC, Roberts CJ, et al. Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: a resource for investigations into drug disposition. Xenobiotica 36(10-11), 963-988 (2006)
    • (2006) Xenobiotica , vol.36 , Issue.10-11 , pp. 963-988
    • Bleasby, K.1    Castle, J.C.2    Roberts, C.J.3
  • 58
    • 84859719229 scopus 로고    scopus 로고
    • Genetic variants of organic cation transporter 1 (OCT1) and OCT2 significantly reduce lamivudine uptake
    • Choi MK, Song IS. Genetic variants of organic cation transporter 1 (OCT1) and OCT2 significantly reduce lamivudine uptake. Biopharm. Drug Dispos. 33(3), 170-178 (2012)
    • (2012) Biopharm. Drug Dispos. , vol.33 , Issue.3 , pp. 170-178
    • Choi, M.K.1    Song, I.S.2
  • 59
    • 0033604060 scopus 로고    scopus 로고
    • Persistent mitochondrial dysfunction and perinatal exposure to antiretroviral nucleoside analogues
    • Blanche S, Tardieu M, Rustin P, et al. Persistent mitochondrial dysfunction and perinatal exposure to antiretroviral nucleoside analogues. Lancet 354(9184), 1084-1089 (1999)
    • (1999) Lancet , vol.354 , Issue.9184 , pp. 1084-1089
    • Blanche, S.1    Tardieu, M.2    Rustin, P.3
  • 60
    • 2342613668 scopus 로고    scopus 로고
    • Mitochondrial damage and DNA depletion in cord blood and umbilical cord from infants exposed in utero to combivir
    • Divi RL, Walker VE, Wade NA, et al. Mitochondrial damage and DNA depletion in cord blood and umbilical cord from infants exposed in utero to combivir. AIDS 18(7), 1013-1021 (2004)
    • (2004) AIDS , vol.18 , Issue.7 , pp. 1013-1021
    • Divi, R.L.1    Walker, V.E.2    Wade, N.A.3
  • 61
    • 66849098130 scopus 로고    scopus 로고
    • Pharmacogenetics of nucleoside reverse-transcriptase inhibitor-associated peripheral neuropathy
    • Kallianpur AR, Hulgan T. Pharmacogenetics of nucleoside reverse-transcriptase inhibitor-associated peripheral neuropathy. Pharmacogenomics 10(4), 623-637 (2009)
    • (2009) Pharmacogenomics , vol.10 , Issue.4 , pp. 623-637
    • Kallianpur, A.R.1    Hulgan, T.2
  • 62
    • 77951892762 scopus 로고    scopus 로고
    • African mitochondrial DNA subhaplogroups and peripheral neuropathy during antiretroviral therapy
    • Canter JA, Robbins GK, Selph D, et al. African mitochondrial DNA subhaplogroups and peripheral neuropathy during antiretroviral therapy. J. Infect. Dis 201(11), 1703-1707 (2010)
    • (2010) J. Infect. Dis , vol.201 , Issue.11 , pp. 1703-1707
    • Canter, J.A.1    Robbins, G.K.2    Selph, D.3
  • 63
    • 84857993084 scopus 로고    scopus 로고
    • Effect of pregnancy on emtricitabine pharmacokinetics
    • Stek AM, Best BM, Luo W, et al. Effect of pregnancy on emtricitabine pharmacokinetics. HIV Med. 13(4), 226-235 (2011)
    • (2011) HIV Med. , vol.13 , Issue.4 , pp. 226-235
    • Stek, A.M.1    Best, B.M.2    Luo, W.3
  • 64
    • 79952349183 scopus 로고    scopus 로고
    • Concentrations of tenofovir and emtricitabine in breast milk of HIV-1-infected women in Abidjan, Cote d'Ivoire, in the ANRS 12109 TEmAA Study, Step 2
    • Benaboud S, Pruvost A, Coffie PA, et al. Concentrations of tenofovir and emtricitabine in breast milk of HIV-1-infected women in Abidjan, Cote d'Ivoire, in the ANRS 12109 TEmAA Study, Step 2. Antimicrob. Agents Chemother. 55(3), 1315-1317 (2011)
    • (2011) Antimicrob. Agents Chemother. , vol.55 , Issue.3 , pp. 1315-1317
    • Benaboud, S.1    Pruvost, A.2    Coffie, P.A.3
  • 65
    • 54049129967 scopus 로고    scopus 로고
    • Emtricitabine: Inhibitor and substrate of multidrug resistance associated protein
    • Bousquet L, Pruvost A, Didier N, et al. Emtricitabine: inhibitor and substrate of multidrug resistance associated protein. Eur. J. Pharm. Sci. 35(4), 247-256 (2008)
    • (2008) Eur. J. Pharm. Sci. , vol.35 , Issue.4 , pp. 247-256
    • Bousquet, L.1    Pruvost, A.2    Didier, N.3
  • 66
    • 3242694930 scopus 로고    scopus 로고
    • Tenofovir disoproxil fumarate: Clinical pharmacology and pharmacokinetics
    • Kearney BP, Flaherty JF, Shah J. Tenofovir disoproxil fumarate: clinical pharmacology and pharmacokinetics. Clin. Pharmacokinet. 43(9), 595-612 (2004)
    • (2004) Clin. Pharmacokinet. , vol.43 , Issue.9 , pp. 595-612
    • Kearney, B.P.1    Flaherty, J.F.2    Shah, J.3
  • 69
    • 84856069811 scopus 로고    scopus 로고
    • Pregnancy-related effects on tenofovir pharmacokinetics: A population study with 186 women
    • Benaboud S, Hirt D, Launay O, et al. Pregnancy-related effects on tenofovir pharmacokinetics: a population study with 186 women. Antimicrob. Agents Chemother. 56(2), 857-862 (2012)
    • (2012) Antimicrob. Agents Chemother. , vol.56 , Issue.2 , pp. 857-862
    • Benaboud, S.1    Hirt, D.2    Launay, O.3
  • 70
    • 84859744786 scopus 로고    scopus 로고
    • Low cerebrospinal fluid concentrations of the nucleotide HIV reverse transcriptase inhibitor, tenofovir
    • Best BM, Letendre SL, Koopmans P, et al. Low cerebrospinal fluid concentrations of the nucleotide HIV reverse transcriptase inhibitor, tenofovir. J. Acquir. Immune Defic. Syndr. 59(4), 376-381 (2012)
    • (2012) J. Acquir. Immune Defic. Syndr. , vol.59 , Issue.4 , pp. 376-381
    • Best, B.M.1    Letendre, S.L.2    Koopmans, P.3
  • 71
    • 33947138325 scopus 로고    scopus 로고
    • Renal transport of adefovir, cidofovir, and tenofovir by SLC22A family members (hOAT1, hOAT3, and hOCT2)
    • Uwai Y, Ida H, Tsuji Y, et al. Renal transport of adefovir, cidofovir, and tenofovir by SLC22A family members (hOAT1, hOAT3, and hOCT2). Pharm. Res. 24(4), 811-815 (2007)
    • (2007) Pharm. Res. , vol.24 , Issue.4 , pp. 811-815
    • Uwai, Y.1    Ida, H.2    Tsuji, Y.3
  • 72
    • 79957933957 scopus 로고    scopus 로고
    • Genetic variants of ABCC10, a novel tenofovir transporter, are associated with kidney tubular dysfunction
    • Pushpakom SP, Liptrott NJ, Rodriguez-Novoa S, et al. Genetic variants of ABCC10, a novel tenofovir transporter, are associated with kidney tubular dysfunction. J. Infect. Dis 204(1), 145-153 (2011)
    • (2011) J. Infect. Dis , vol.204 , Issue.1 , pp. 145-153
    • Pushpakom, S.P.1    Liptrott, N.J.2    Rodriguez-Novoa, S.3
  • 73
    • 10744223047 scopus 로고    scopus 로고
    • Expression of human organic anion transporters in the choroid plexus and their interactions with neurotransmitter metabolites
    • Alebouyeh M, Takeda M, Onozato ML, et al. Expression of human organic anion transporters in the choroid plexus and their interactions with neurotransmitter metabolites. J. Pharmacol. Sci. 93(4), 430-436 (2003)
    • (2003) J. Pharmacol. Sci. , vol.93 , Issue.4 , pp. 430-436
    • Alebouyeh, M.1    Takeda, M.2    Onozato, M.L.3
  • 74
    • 0029092440 scopus 로고
    • Pharmacokinetics of fluconazole after oral administration in children with human immunodeficiency virus infection
    • Nahata MC, Brady MT. Pharmacokinetics of fluconazole after oral administration in children with human immunodeficiency virus infection. Eur. J Clin Pharmacol. 48(3-4), 291-293 (1995)
    • (1995) Eur. J Clin Pharmacol. , vol.48 , Issue.3-4 , pp. 291-293
    • Nahata, M.C.1    Brady, M.T.2
  • 75
    • 4344632950 scopus 로고    scopus 로고
    • Mrp4 confers resistance to topotecan and protects the brain from chemotherapy
    • Leggas M, Adachi M, Scheffer GL, et al. Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol. Cell. Biol. 24(17), 7612-7621 (2004)
    • (2004) Mol. Cell. Biol. , vol.24 , Issue.17 , pp. 7612-7621
    • Leggas, M.1    Adachi, M.2    Scheffer, G.L.3
  • 76
    • 0036488144 scopus 로고    scopus 로고
    • The distribution of the anti-HIV drug, 23-dideoxycytidine (ddC), across the blood-brain and blood-cerebrospinal fluid barriers and the influence of organic anion transport inhibitors
    • Gibbs JE, Thomas SA. The distribution of the anti-HIV drug, 23-dideoxycytidine (ddC), across the blood-brain and blood-cerebrospinal fluid barriers and the influence of organic anion transport inhibitors. J. Neurochem. 80(3), 392-404 (2002)
    • (2002) J. Neurochem. , vol.80 , Issue.3 , pp. 392-404
    • Gibbs, J.E.1    Thomas, S.A.2
  • 77
    • 33645895267 scopus 로고    scopus 로고
    • The distribution of the anti-HIV drug, tenofovir (PMPA), into the brain, CSF and choroid plexuses
    • Anthonypillai C, Gibbs JE, Thomas SA. The distribution of the anti-HIV drug, tenofovir (PMPA), into the brain, CSF and choroid plexuses. Cerebrospinal Fluid Res. 3, 1 (2006)
    • (2006) Cerebrospinal Fluid Res. , vol.3 , pp. 1
    • Anthonypillai, C.1    Gibbs, J.E.2    Thomas, S.A.3
  • 78
    • 42049090195 scopus 로고    scopus 로고
    • Clinical and genetic determinants of intracellular tenofovir diphosphate concentrations in HIV-infected patients
    • Kiser JJ, Aquilante CL, Anderson PL, et al. Clinical and genetic determinants of intracellular tenofovir diphosphate concentrations in HIV-infected patients. J. Acquir. Immune Defic. Syndr. 47(3), 298-303 (2008)
    • (2008) J. Acquir. Immune Defic. Syndr. , vol.47 , Issue.3 , pp. 298-303
    • Kiser, J.J.1    Aquilante, C.L.2    Anderson, P.L.3
  • 79
    • 0034852408 scopus 로고    scopus 로고
    • Human renal organic anion transporter 1 (hOAT1) and its role in the nephrotoxicity of antiviral nucleotide analogs
    • Cihlar T, Ho ES, Lin DC, et al. Human renal organic anion transporter 1 (hOAT1) and its role in the nephrotoxicity of antiviral nucleotide analogs. Nucleosides Nucleotides Nucleic Acids 20(4-7), 641-648 (2001)
    • (2001) Nucleosides Nucleotides Nucleic Acids , vol.20 , Issue.4-7 , pp. 641-648
    • Cihlar, T.1    Ho, E.S.2    Lin, D.C.3
  • 80
    • 30344432397 scopus 로고    scopus 로고
    • Multidrug resistance-associated protein 2 (MRP2) affects hepatobiliary elimination but not the intestinal disposition of tenofovir disoproxil fumarate and its metabolites
    • Mallants R, Van Oosterwyck K, Van Vaeck L, et al. Multidrug resistance-associated protein 2 (MRP2) affects hepatobiliary elimination but not the intestinal disposition of tenofovir disoproxil fumarate and its metabolites. Xenobiotica 35(10-11), 1055-1066 (2005)
    • (2005) Xenobiotica , vol.35 , Issue.10-11 , pp. 1055-1066
    • Mallants, R.1    Van Oosterwyck, K.2    Van Vaeck, L.3
  • 81
    • 33846420624 scopus 로고    scopus 로고
    • Functional involvement of multidrug resistance-associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs adefovir and tenofovir
    • Imaoka T, Kusuhara H, Adachi M, et al. Functional involvement of multidrug resistance-associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs adefovir and tenofovir. Mol. Pharmacol. 71(2), 619-627 (2007)
    • (2007) Mol. Pharmacol. , vol.71 , Issue.2 , pp. 619-627
    • Imaoka, T.1    Kusuhara, H.2    Adachi, M.3
  • 82
    • 66949119640 scopus 로고    scopus 로고
    • Predictors of kidney tubular dysfunction in HIV-infected patients treated with tenofovir: A pharmacogenetic study
    • Rodriguez-Novoa S, Labarga P, Soriano V, et al. Predictors of kidney tubular dysfunction in HIV-infected patients treated with tenofovir: a pharmacogenetic study. Clin. Infect. Dis 48(11), e108-e116 (2009)
    • (2009) Clin. Infect. Dis , vol.48 , Issue.11
    • Rodriguez-Novoa, S.1    Labarga, P.2    Soriano, V.3
  • 83
    • 77950925935 scopus 로고    scopus 로고
    • Impairment in kidney tubular function in patients receiving tenofovir is associated with higher tenofovir plasma concentrations
    • Rodriguez-Novoa S, Labarga P, D'Avolio A, et al. Impairment in kidney tubular function in patients receiving tenofovir is associated with higher tenofovir plasma concentrations. AIDS 24(7), 1064-1066 (2010)
    • (2010) AIDS , vol.24 , Issue.7 , pp. 1064-1066
    • Rodriguez-Novoa, S.1    Labarga, P.2    D'avolio, A.3
  • 84
    • 84875810804 scopus 로고    scopus 로고
    • Viramune® Tablets, Viramune® Oral Suspension, package insert. Boehringer-Ingelheim Pharmaceuticals Inc., CT, USA
    • Viramune® Tablets, Viramune® Oral Suspension, package insert. Boehringer-Ingelheim Pharmaceuticals Inc., CT, USA.
  • 85
    • 14744282818 scopus 로고    scopus 로고
    • Persistence of nevirapine exposure during the postpartum period after intrapartum single-dose nevirapine in addition to zidovudine prophylaxis for the prevention of mother-to-child transmission of HIV-1
    • Cressey TR, Jourdain G, Lallemant MJ, et al. Persistence of nevirapine exposure during the postpartum period after intrapartum single-dose nevirapine in addition to zidovudine prophylaxis for the prevention of mother-to-child transmission of HIV-1. J. Acquir. Immune Defic. Syndr. 38(3), 283-288 (2005)
    • (2005) J. Acquir. Immune Defic. Syndr. , vol.38 , Issue.3 , pp. 283-288
    • Cressey, T.R.1    Jourdain, G.2    Lallemant, M.J.3
  • 86
    • 3142762317 scopus 로고    scopus 로고
    • Intrapartum exposure to nevirapine and subsequent maternal responses to nevirapine-based antiretroviral therapy
    • Jourdain G, Ngo-Giang-Huong N, Le Coeur S, et al. Intrapartum exposure to nevirapine and subsequent maternal responses to nevirapine-based antiretroviral therapy. N. Engl. J. Med. 351(3), 229-240 (2004)
    • (2004) N. Engl. J. Med. , vol.351 , Issue.3 , pp. 229-240
    • Jourdain, G.1    Ngo-Giang-Huong, N.2    Le Coeur, S.3
  • 87
    • 70449134113 scopus 로고    scopus 로고
    • Efficacy of short-course AZT plus 3TC to reduce nevirapine resistance in the prevention of mother-to-child HIV transmission: A randomized clinical trial
    • McIntyre JA, Hopley M, Moodley D, et al. Efficacy of short-course AZT plus 3TC to reduce nevirapine resistance in the prevention of mother-to-child HIV transmission: a randomized clinical trial. PLoS Med. 6(10), e1000172 (2009)
    • (2009) PLoS Med. , vol.6 , Issue.10
    • McIntyre, J.A.1    Hopley, M.2    Moodley, D.3
  • 88
    • 84555209220 scopus 로고    scopus 로고
    • A comparison of 3 regimens to prevent nevirapine resistance mutations in HIV-infected pregnant women receiving a single intrapartum dose of nevirapine
    • Van Dyke RB, Ngo-Giang-Huong N, Shapiro DE, et al. A comparison of 3 regimens to prevent nevirapine resistance mutations in HIV-infected pregnant women receiving a single intrapartum dose of nevirapine. Clin. Infect. Dis 54(2), 285-293 (2012)
    • (2012) Clin. Infect. Dis , vol.54 , Issue.2 , pp. 285-293
    • Van Dyke, R.B.1    Ngo-Giang-Huong, N.2    Shapiro, D.E.3
  • 89
    • 73549117428 scopus 로고    scopus 로고
    • Influence of CYP2B6 polymorphisms on the persistence of plasma nevirapine concentrations following a single intra-partum dose for the prevention of mother to child transmission in HIV-infected Thai women
    • Chantarangsu S, Cressey TR, Mahasirimongkol S, et al. Influence of CYP2B6 polymorphisms on the persistence of plasma nevirapine concentrations following a single intra-partum dose for the prevention of mother to child transmission in HIV-infected Thai women. J. Antimicrob. Chemother. 64(6), 1265-1273 (2009)
    • (2009) J. Antimicrob. Chemother. , vol.64 , Issue.6 , pp. 1265-1273
    • Chantarangsu, S.1    Cressey, T.R.2    Mahasirimongkol, S.3
  • 90
    • 41549167294 scopus 로고    scopus 로고
    • Chronic administration of nevirapine during pregnancy: Impact of pregnancy on pharmacokinetics
    • Capparelli EV, Aweeka F, Hitti J, et al. Chronic administration of nevirapine during pregnancy: impact of pregnancy on pharmacokinetics. HIV Med. 9(4), 214-220 (2008)
    • (2008) HIV Med. , vol.9 , Issue.4 , pp. 214-220
    • Capparelli, E.V.1    Aweeka, F.2    Hitti, J.3
  • 91
    • 78049272719 scopus 로고    scopus 로고
    • Suboptimal nevirapine steady-state pharmacokinetics during intrapartum compared with postpartum in HIV-1-seropositive Ugandan women
    • Lamorde M, Byakika-Kibwika P, Okaba-Kayom V, et al. Suboptimal nevirapine steady-state pharmacokinetics during intrapartum compared with postpartum in HIV-1-seropositive Ugandan women. J. Acquir. Immune Defic. Syndr. 55(3), 345-350 (2010)
    • (2010) J. Acquir. Immune Defic. Syndr. , vol.55 , Issue.3 , pp. 345-350
    • Lamorde, M.1    Byakika-Kibwika, P.2    Okaba-Kayom, V.3
  • 92
    • 33749990555 scopus 로고    scopus 로고
    • A comparison of the steady-state pharmacokinetics of nevirapine in men, nonpregnant women and women in late pregnancy
    • von Hentig N, Carlebach A, Gute P, et al. A comparison of the steady-state pharmacokinetics of nevirapine in men, nonpregnant women and women in late pregnancy. Br. J. Clin. Pharmacol. 62(5), 552-559 (2006)
    • (2006) Br. J. Clin. Pharmacol. , vol.62 , Issue.5 , pp. 552-559
    • Von Hentig, N.1    Carlebach, A.2    Gute, P.3
  • 93
    • 33847793822 scopus 로고    scopus 로고
    • Cytochrome P450 2B6 (CYP2B6) G516T influences nevirapine plasma concentrations in HIV-infected patients in Uganda
    • Penzak SR, Kabuye G, Mugyenyi P, et al. Cytochrome P450 2B6 (CYP2B6) G516T influences nevirapine plasma concentrations in HIV-infected patients in Uganda. HIV Med. 8(2), 86-91 (2007)
    • (2007) HIV Med. , vol.8 , Issue.2 , pp. 86-91
    • Penzak, S.R.1    Kabuye, G.2    Mugyenyi, P.3
  • 94
    • 41149092923 scopus 로고    scopus 로고
    • Impact of CYP2B6 983T>C polymorphism on non-nucleoside reverse transcriptase inhibitor plasma concentrations in HIV-infected patients
    • Wyen C, Hendra H, Vogel M, et al. Impact of CYP2B6 983T>C polymorphism on non-nucleoside reverse transcriptase inhibitor plasma concentrations in HIV-infected patients. J. Antimicrob. Chemother. 61(4), 914-918 (2008)
    • (2008) J. Antimicrob. Chemother. , vol.61 , Issue.4 , pp. 914-918
    • Wyen, C.1    Hendra, H.2    Vogel, M.3
  • 95
    • 83755183491 scopus 로고    scopus 로고
    • Exploration of CYP450 and drug transporter genotypes and correlations with nevirapine exposure in Malawians
    • Brown KC, Hosseinipour MC, Hoskins JM, et al. Exploration of CYP450 and drug transporter genotypes and correlations with nevirapine exposure in Malawians. Pharmacogenomics 13(1), 113-121 (2012)
    • (2012) Pharmacogenomics , vol.13 , Issue.1 , pp. 113-121
    • Brown, K.C.1    Hosseinipour, M.C.2    Hoskins, J.M.3
  • 96
    • 83655201204 scopus 로고    scopus 로고
    • Association of ABCC10 polymorphisms with nevirapine plasma concentrations in the German Competence Network for HIV/AIDS
    • Liptrott NJ, Pushpakom S, Wyen C, et al. Association of ABCC10 polymorphisms with nevirapine plasma concentrations in the German Competence Network for HIV/AIDS. Pharmacogenet. Genomics 22(1), 10-19 (2012)
    • (2012) Pharmacogenet. Genomics , vol.22 , Issue.1 , pp. 10-19
    • Liptrott, N.J.1    Pushpakom, S.2    Wyen, C.3
  • 97
    • 3042814812 scopus 로고    scopus 로고
    • Maternal toxicity with continuous nevirapine in pregnancy: Results from PACTG 1022
    • Hitti J, Frenkel LM, Stek AM, et al. Maternal toxicity with continuous nevirapine in pregnancy: results from PACTG 1022. J. Acquir. Immune Defic. Syndr. 36(3), 772-776 (2004)
    • (2004) J. Acquir. Immune Defic. Syndr. , vol.36 , Issue.3 , pp. 772-776
    • Hitti, J.1    Frenkel, L.M.2    Stek, A.M.3
  • 98
    • 34547399409 scopus 로고    scopus 로고
    • Nevirapine-associated toxicity in HIV-infected Thai men and women, including pregnant women
    • Phanuphak N, Apornpong T, Teeratakulpisarn S, et al. Nevirapine- associated toxicity in HIV-infected Thai men and women, including pregnant women. HIV Med. 8(6), 357-366 (2007)
    • (2007) HIV Med. , vol.8 , Issue.6 , pp. 357-366
    • Phanuphak, N.1    Apornpong, T.2    Teeratakulpisarn, S.3
  • 99
    • 59549096348 scopus 로고    scopus 로고
    • HLAB*3505 allele is a strong predictor for nevirapine-induced skin adverse drug reactions in HIV-infected Thai patients
    • Chantarangsu S, Mushiroda T, Mahasirimongkol S, et al. HLAB, 3505 allele is a strong predictor for nevirapine-induced skin adverse drug reactions in HIV-infected Thai patients. Pharmacogenet. Genomics 19(2), 139-146 (2009)
    • (2009) Pharmacogenet. Genomics , vol.19 , Issue.2 , pp. 139-146
    • Chantarangsu, S.1    Mushiroda, T.2    Mahasirimongkol, S.3
  • 100
    • 79961219567 scopus 로고    scopus 로고
    • Genome-wide association study identifies variations in 6p21.3 associated with nevirapine-induced rash
    • Chantarangsu S, Mushiroda T, Mahasirimongkol S, et al. Genome-wide association study identifies variations in 6p21.3 associated with nevirapine-induced rash. Clin. Infect. Dis 53(4), 341-348 (2011)
    • (2011) Clin. Infect. Dis , vol.53 , Issue.4 , pp. 341-348
    • Chantarangsu, S.1    Mushiroda, T.2    Mahasirimongkol, S.3
  • 101
    • 34147190912 scopus 로고    scopus 로고
    • Nevirapine concentrations in HIV-infected children treated with divided fixed-dose combination antiretroviral tablets in Malawi and Zambia
    • Ellis JC, L'homme RF, Ewings FM, et al. Nevirapine concentrations in HIV-infected children treated with divided fixed-dose combination antiretroviral tablets in Malawi and Zambia. Antivir. Ther. 12(2), 253-260 (2007)
    • (2007) Antivir. Ther. , vol.12 , Issue.2 , pp. 253-260
    • Ellis, J.C.1    L'homme, R.F.2    Ewings, F.M.3
  • 102
    • 84867889296 scopus 로고    scopus 로고
    • Rash, hepatotoxicity and hyperbilirubinemia among kenyan infants born to HIV-infected women receiving triple-antiretroviral drugs for the prevention of mother-to-child HIV transmission
    • doi:10.1097/INF.0b013e318267ef6a (Epub ahead of print)
    • Minniear TD, Zeh C, Polle N, et al. Rash, hepatotoxicity and hyperbilirubinemia among kenyan infants born to HIV-infected women receiving triple-antiretroviral drugs for the prevention of mother-to-child HIV transmission. Pediatr. Infect. Dis. J. doi:10.1097/INF.0b013e318267ef6a
    • (2012) Pediatr. Infect. Dis. J.
    • Minniear, T.D.1    Zeh, C.2    Polle, N.3
  • 103
    • 0037169263 scopus 로고    scopus 로고
    • Myelomeningocele in a child with intrauterine exposure to efavirenz
    • Fundaro C, Genovese O, Rendeli C, et al. Myelomeningocele in a child with intrauterine exposure to efavirenz. AIDS 16(2), 299-300 (2002)
    • (2002) AIDS , vol.16 , Issue.2 , pp. 299-300
    • Fundaro, C.1    Genovese, O.2    Rendeli, C.3
  • 104
    • 34347348169 scopus 로고    scopus 로고
    • Pregnancy rates and birth outcomes among women on efavirenz-containing highly active antiretroviral therapy in Botswana
    • Bussmann H, Wester CW, Wester CN, et al. Pregnancy rates and birth outcomes among women on efavirenz-containing highly active antiretroviral therapy in Botswana. J. Acquir. Immune Defic. Syndr. 45(3), 269-273 (2007)
    • (2007) J. Acquir. Immune Defic. Syndr. , vol.45 , Issue.3 , pp. 269-273
    • Bussmann, H.1    Wester, C.W.2    Wester, C.N.3
  • 105
    • 74249094734 scopus 로고    scopus 로고
    • Birth defects following exposure to efavirenz-based antiretroviral therapy during pregnancy: A study at a regional South African hospital
    • Bera E, McCausland K, Nonkwelo R, et al. Birth defects following exposure to efavirenz-based antiretroviral therapy during pregnancy: a study at a regional South African hospital. AIDS 24(2), 283-289 (2010)
    • (2010) AIDS , vol.24 , Issue.2 , pp. 283-289
    • Bera, E.1    McCausland, K.2    Nonkwelo, R.3
  • 106
    • 77953914155 scopus 로고    scopus 로고
    • Safety of efavirenz in first-trimester of pregnancy: A systematic review and meta-analysis of outcomes from observational cohorts
    • Ford N, Mofenson L, Kranzer K, et al. Safety of efavirenz in first-trimester of pregnancy: a systematic review and meta-analysis of outcomes from observational cohorts. AIDS 24(10), 1461-1470 (2010)
    • (2010) AIDS , vol.24 , Issue.10 , pp. 1461-1470
    • Ford, N.1    Mofenson, L.2    Kranzer, K.3
  • 107
    • 78751607758 scopus 로고    scopus 로고
    • Pregnancy outcomes in women exposed to efavirenz and nevirapine: An appraisal of the IeDEA west Africa and ANRS databases, Abidjan, Cote d'Ivoire
    • Ekouevi DK, Coffie PA, Ouattara E, et al. Pregnancy outcomes in women exposed to efavirenz and nevirapine: an appraisal of the IeDEA west Africa and ANRS databases, Abidjan, Cote d'Ivoire. J. Acquir. Immune Defic. Syndr. 56(2), 183-187 (2011)
    • (2011) J. Acquir. Immune Defic. Syndr. , vol.56 , Issue.2 , pp. 183-187
    • Ekouevi, D.K.1    Coffie, P.A.2    Ouattara, E.3
  • 108
    • 84859758788 scopus 로고    scopus 로고
    • Efavirenz pharmacokinetics during the third trimester of pregnancy and postpartum
    • Cressey TR, Stek A, Capparelli E, et al. Efavirenz pharmacokinetics during the third trimester of pregnancy and postpartum. J. Acquir. Immune Defic. Syndr. 59(3), 245-252 (2012)
    • (2012) J. Acquir. Immune Defic. Syndr. , vol.59 , Issue.3 , pp. 245-252
    • Cressey, T.R.1    Stek, A.2    Capparelli, E.3
  • 109
    • 0038002981 scopus 로고    scopus 로고
    • The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: Implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity
    • Ward BA, Gorski JC, Jones DR, et al. The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity. J. Pharmacol. Exp. Ther. 306(1), 287-300 (2003)
    • (2003) J. Pharmacol. Exp. Ther. , vol.306 , Issue.1 , pp. 287-300
    • Ward, B.A.1    Gorski, J.C.2    Jones, D.R.3
  • 110
    • 63849240305 scopus 로고    scopus 로고
    • In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function
    • di Iulio J, Fayet A, Arab-Alameddine M, et al. In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function. Pharmacogenet. Genomics 19(4), 300-309 (2009)
    • (2009) Pharmacogenet. Genomics , vol.19 , Issue.4 , pp. 300-309
    • Di Iulio, J.1    Fayet, A.2    Arab-Alameddine, M.3
  • 111
    • 77953756056 scopus 로고    scopus 로고
    • Efavirenz primary and secondary metabolism in vitro and in vivo: Identification of novel metabolic pathways and cytochrome P450 2A6 as the principal catalyst of efavirenz 7-hydroxylation
    • Ogburn ET, Jones DR, Masters AR, et al. Efavirenz primary and secondary metabolism in vitro and in vivo: identification of novel metabolic pathways and cytochrome P450 2A6 as the principal catalyst of efavirenz 7-hydroxylation. Drug Metab. Dispos. 38(7), 1218-1229 (2010)
    • (2010) Drug Metab. Dispos. , vol.38 , Issue.7 , pp. 1218-1229
    • Ogburn, E.T.1    Jones, D.R.2    Masters, A.R.3
  • 112
    • 79956364942 scopus 로고    scopus 로고
    • Identification of human UGT isoforms responsible for glucuronidation of efavirenz and its three hydroxy metabolites
    • Bae SK, Jeong YJ, Lee C, et al. Identification of human UGT isoforms responsible for glucuronidation of efavirenz and its three hydroxy metabolites. Xenobiotica 41(6), 437-444 (2011)
    • (2011) Xenobiotica , vol.41 , Issue.6 , pp. 437-444
    • Bae, S.K.1    Jeong, Y.J.2    Lee, C.3
  • 113
    • 77952322356 scopus 로고    scopus 로고
    • CYP2B6, CYP2A6 and UGT2B7 genetic polymorphisms are predictors of efavirenz mid-dose concentration in HIV-infected patients
    • Kwara A, Lartey M, Sagoe KW, et al. CYP2B6, CYP2A6 and UGT2B7 genetic polymorphisms are predictors of efavirenz mid-dose concentration in HIV-infected patients. AIDS 23(16), 2101-2106 (2009)
    • (2009) AIDS , vol.23 , Issue.16 , pp. 2101-2106
    • Kwara, A.1    Lartey, M.2    Sagoe, K.W.3
  • 114
    • 63849281439 scopus 로고    scopus 로고
    • CYP2B6 (c.516G->T) and CYP2A6 (*9B and/or(*17) polymorphisms are independent predictors of efavirenz plasma concentrations in HIV-infected patients
    • Kwara A, Lartey M, Sagoe KW, et al. CYP2B6 (c.516G->T) and CYP2A6 (, 9B and/or , 17) polymorphisms are independent predictors of efavirenz plasma concentrations in HIV-infected patients. Br. J. Clin. Pharmacol. 67(4), 427-436 (2009)
    • (2009) Br. J. Clin. Pharmacol. , vol.67 , Issue.4 , pp. 427-436
    • Kwara, A.1    Lartey, M.2    Sagoe, K.W.3
  • 115
    • 20244364148 scopus 로고    scopus 로고
    • Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients
    • Rotger M, Colombo S, Furrer H, et al. Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients. Pharmacogenet. Genomics 15(1), 1-5 (2005)
    • (2005) Pharmacogenet. Genomics , vol.15 , Issue.1 , pp. 1-5
    • Rotger, M.1    Colombo, S.2    Furrer, H.3
  • 116
    • 50649088977 scopus 로고    scopus 로고
    • Efavirenz in human breast milk, mothers', and newborns' plasma
    • Schneider S, Peltier A, Gras A, et al. Efavirenz in human breast milk, mothers', and newborns' plasma. J. Acquir. Immune Defic. Syndr. 48(4), 450-454 (2008)
    • (2008) J. Acquir. Immune Defic. Syndr. , vol.48 , Issue.4 , pp. 450-454
    • Schneider, S.1    Peltier, A.2    Gras, A.3
  • 117
    • 0035875828 scopus 로고    scopus 로고
    • Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients
    • Marzolini C, Telenti A, Decosterd LA, et al. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients. AIDS 15(1), 71-75 (2001)
    • (2001) AIDS , vol.15 , Issue.1 , pp. 71-75
    • Marzolini, C.1    Telenti, A.2    Decosterd, L.A.3
  • 118
    • 81155159797 scopus 로고    scopus 로고
    • Efavirenz is a substrate and in turn modulates the expression of the efflux transporter ABCG2/BCRP in the gastrointestinal tract of the rat
    • Peroni RN, Di Gennaro SS, Hocht C, et al. Efavirenz is a substrate and in turn modulates the expression of the efflux transporter ABCG2/BCRP in the gastrointestinal tract of the rat. Biochem. Pharmacol. 82(9), 1227-1233 (2011)
    • (2011) Biochem. Pharmacol. , vol.82 , Issue.9 , pp. 1227-1233
    • Peroni, R.N.1    Di Gennaro, S.S.2    Hocht, C.3
  • 119
    • 0035870289 scopus 로고    scopus 로고
    • Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues
    • Maliepaard M, Scheffer GL, Faneyte IF, et al. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res. 61(8), 3458-3464 (2001)
    • (2001) Cancer Res. , vol.61 , Issue.8 , pp. 3458-3464
    • Maliepaard, M.1    Scheffer, G.L.2    Faneyte, I.F.3
  • 120
    • 14644409841 scopus 로고    scopus 로고
    • The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk
    • Jonker JW, Merino G, Musters S, et al. The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat. Med. 11(2), 127-129 (2005)
    • (2005) Nat. Med. , vol.11 , Issue.2 , pp. 127-129
    • Jonker, J.W.1    Merino, G.2    Musters, S.3
  • 121
    • 79951896688 scopus 로고    scopus 로고
    • Fetoprotective activity of breast cancer resistance protein (BCRP, ABCG2): Expression and function throughout pregnancy
    • Hahnova-Cygalova L, Ceckova M, Staud F. Fetoprotective activity of breast cancer resistance protein (BCRP, ABCG2): expression and function throughout pregnancy. Drug Metab. Rev. 43(1), 53-68 (2011)
    • (2011) Drug Metab. Rev. , vol.43 , Issue.1 , pp. 53-68
    • Hahnova-Cygalova, L.1    Ceckova, M.2    Staud, F.3
  • 122
    • 79952288406 scopus 로고    scopus 로고
    • Safety and pharmacokinetics of etravirine in pregnant HIV-1-infected women
    • Izurieta P, Kakuda TN, Feys C, et al. Safety and pharmacokinetics of etravirine in pregnant HIV-1-infected women. HIV Med. 12(4), 257-258 (2011)
    • (2011) HIV Med. , vol.12 , Issue.4 , pp. 257-258
    • Izurieta, P.1    Kakuda, T.N.2    Feys, C.3
  • 123
    • 84858394506 scopus 로고    scopus 로고
    • Biotransformation of the antiretroviral drug etravirine: Metabolite identification, reaction phenotyping, and characterization of autoinduction of cytochrome P450-dependent metabolism
    • Yanakakis LJ, Bumpus NN. Biotransformation of the antiretroviral drug etravirine: metabolite identification, reaction phenotyping, and characterization of autoinduction of cytochrome P450-dependent metabolism. Drug Metab. Dispos. 40(4), 803-814 (2012)
    • (2012) Drug Metab. Dispos. , vol.40 , Issue.4 , pp. 803-814
    • Yanakakis, L.J.1    Bumpus, N.N.2
  • 124
    • 10744230915 scopus 로고    scopus 로고
    • Pregnancy and use of oral contraceptives reduces the biotransformation of proguanil to cycloguanil
    • McGready R, Stepniewska K, Seaton E, et al. Pregnancy and use of oral contraceptives reduces the biotransformation of proguanil to cycloguanil. Eur. J. Clin. Pharmacol. 59(7), 553-557 (2003)
    • (2003) Eur. J. Clin. Pharmacol. , vol.59 , Issue.7 , pp. 553-557
    • McGready, R.1    Stepniewska, K.2    Seaton, E.3
  • 125
    • 58749094444 scopus 로고    scopus 로고
    • Cytochrome P-450 polymorphisms and response to clopidogrel
    • Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N. Engl. J. Med. 360(4), 354-362 (2009)
    • (2009) N. Engl. J. Med. , vol.360 , Issue.4 , pp. 354-362
    • Mega, J.L.1    Close, S.L.2    Wiviott, S.D.3
  • 126
    • 76349098199 scopus 로고    scopus 로고
    • Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-treated patients with coronary stent placement
    • Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-treated patients with coronary stent placement. Circulation 121(4), 512-518 (2010)
    • (2010) Circulation , vol.121 , Issue.4 , pp. 512-518
    • Sibbing, D.1    Koch, W.2    Gebhard, D.3
  • 127
    • 0030430035 scopus 로고    scopus 로고
    • Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes
    • Kumar GN, Rodrigues AD, Buko AM, et al. Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes. J. Pharmacol. Exp. Ther. 277(1), 423-431 (1996)
    • (1996) J. Pharmacol. Exp. Ther. , vol.277 , Issue.1 , pp. 423-431
    • Kumar, G.N.1    Rodrigues, A.D.2    Buko, A.M.3
  • 128
    • 0035831246 scopus 로고    scopus 로고
    • Assessment of active transport of HIV protease inhibitors in various cell lines and the in vitro blood-brain barrier
    • van der Sandt IC, Vos CM, Nabulsi L, et al. Assessment of active transport of HIV protease inhibitors in various cell lines and the in vitro blood-brain barrier. AIDS 15(4), 483-491 (2001)
    • (2001) AIDS , vol.15 , Issue.4 , pp. 483-491
    • Van Der Sandt, I.C.1    Vos, C.M.2    Nabulsi, L.3
  • 129
    • 77953725356 scopus 로고    scopus 로고
    • Concentration-dependent effects and intracellular accumulation of HIV protease inhibitors in cultured CD4 T cells and primary human lymphocytes
    • Janneh O, Bray PG, Jones E, et al. Concentration-dependent effects and intracellular accumulation of HIV protease inhibitors in cultured CD4 T cells and primary human lymphocytes. J. Antimicrob. Chemother. 65(5), 906-916 (2010)
    • (2010) J. Antimicrob. Chemother. , vol.65 , Issue.5 , pp. 906-916
    • Janneh, O.1    Bray, P.G.2    Jones, E.3
  • 130
    • 33748911183 scopus 로고    scopus 로고
    • Reduced lopinavir exposure during pregnancy
    • Stek AM, Mirochnick M, Capparelli E, et al. Reduced lopinavir exposure during pregnancy. AIDS 20(15), 1931-1939 (2006)
    • (2006) AIDS , vol.20 , Issue.15 , pp. 1931-1939
    • Stek, A.M.1    Mirochnick, M.2    Capparelli, E.3
  • 131
    • 84856067531 scopus 로고    scopus 로고
    • Improved oral bioavailability of lopinavir in melt-extruded tablet formulation reduces impact of third trimester on lopinavir plasma concentrations
    • Else LJ, Douglas M, Dickinson L, et al. Improved oral bioavailability of lopinavir in melt-extruded tablet formulation reduces impact of third trimester on lopinavir plasma concentrations. Antimicrob. Agents Chemother. 56(2), 816-824 (2012)
    • (2012) Antimicrob. Agents Chemother. , vol.56 , Issue.2 , pp. 816-824
    • Else, L.J.1    Douglas, M.2    Dickinson, L.3
  • 132
    • 77956232308 scopus 로고    scopus 로고
    • Pharmacokinetics and virologic response of zidovudine/lopinavir/ritonavir initiated during the third trimester of pregnancy
    • Cressey TR, Jourdain G, Rawangban B, et al. Pharmacokinetics and virologic response of zidovudine/lopinavir/ritonavir initiated during the third trimester of pregnancy. AIDS 24(14), 2193-2200 (2010)
    • (2010) AIDS , vol.24 , Issue.14 , pp. 2193-2200
    • Cressey, T.R.1    Jourdain, G.2    Rawangban, B.3
  • 133
    • 84859741971 scopus 로고    scopus 로고
    • Lopinavir/ritonavir trough concentrations with the tablet formulation in HIV-1-infected women during the third trimester of pregnancy
    • Calza L, Manfredi R, Trapani F, et al. Lopinavir/ritonavir trough concentrations with the tablet formulation in HIV-1-infected women during the third trimester of pregnancy. Scand. J. Infect. Dis 44(5), 381-387 (2012)
    • (2012) Scand. J. Infect. Dis , vol.44 , Issue.5 , pp. 381-387
    • Calza, L.1    Manfredi, R.2    Trapani, F.3
  • 134
    • 79958782001 scopus 로고    scopus 로고
    • Thai HIV-1-infected women do not require a dose increase of lopinavir/ritonavir during the third trimester of pregnancy
    • Ramautarsing RA, van der Lugt J, Gorowara M, et al. Thai HIV-1-infected women do not require a dose increase of lopinavir/ritonavir during the third trimester of pregnancy. AIDS 25(10), 1299-1303 (2011)
    • (2011) AIDS , vol.25 , Issue.10 , pp. 1299-1303
    • Ramautarsing, R.A.1    Van Der Lugt, J.2    Gorowara, M.3
  • 135
    • 79551605542 scopus 로고    scopus 로고
    • Therapeutic drug monitoring of lopinavir/ritonavir in pregnancy
    • Lambert JS, Else LJ, Jackson V, et al. Therapeutic drug monitoring of lopinavir/ritonavir in pregnancy. HIV Med. 12(3), 166-173 (2011)
    • (2011) HIV Med. , vol.12 , Issue.3 , pp. 166-173
    • Lambert, J.S.1    Else, L.J.2    Jackson, V.3
  • 136
    • 77955545884 scopus 로고    scopus 로고
    • Lopinavir tablet pharmacokinetics with an increased dose during pregnancy
    • Best BM, Stek AM, Mirochnick M, et al. Lopinavir tablet pharmacokinetics with an increased dose during pregnancy. J. Acquir. Immune Defic. Syndr. 54(4), 381-388 (2010)
    • (2010) J. Acquir. Immune Defic. Syndr. , vol.54 , Issue.4 , pp. 381-388
    • Best, B.M.1    Stek, A.M.2    Mirochnick, M.3
  • 137
    • 75649091119 scopus 로고    scopus 로고
    • HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms
    • Hartkoorn RC, Kwan WS, Shallcross V, et al. HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms. Pharmacogenet. Genomics 20(2), 112-120 (2010)
    • (2010) Pharmacogenet. Genomics , vol.20 , Issue.2 , pp. 112-120
    • Hartkoorn, R.C.1    Kwan, W.S.2    Shallcross, V.3
  • 138
    • 84865709630 scopus 로고    scopus 로고
    • Estimation of the effect of SLCO1B1 polymorphisms on lopinavir plasma concentration in HIV-infected adults
    • Schipani A, Egan D, Dickinson L, et al. Estimation of the effect of SLCO1B1 polymorphisms on lopinavir plasma concentration in HIV-infected adults. Antivir. Ther. 17(5), 861-868 (2012)
    • (2012) Antivir. Ther. , vol.17 , Issue.5 , pp. 861-868
    • Schipani, A.1    Egan, D.2    Dickinson, L.3
  • 139
    • 55349133078 scopus 로고    scopus 로고
    • Studies on antiretroviral drug concentrations in breast milk: Validation of a liquid chromatography-tandem mass spectrometric method for the determination of 7 anti-human immunodeficiency virus medications
    • Rezk NL, White N, Bridges AS, et al. Studies on antiretroviral drug concentrations in breast milk: validation of a liquid chromatography-tandem mass spectrometric method for the determination of 7 anti-human immunodeficiency virus medications. Ther. Drug. Monit. 30(5), 611-619 (2008)
    • (2008) Ther. Drug. Monit. , vol.30 , Issue.5 , pp. 611-619
    • Rezk, N.L.1    White, N.2    Bridges, A.S.3
  • 140
    • 36348967131 scopus 로고    scopus 로고
    • Atazanavir plus low-dose ritonavir in pregnancy: Pharmacokinetics and placental transfer
    • Ripamonti D, Cattaneo D, Maggiolo F, et al. Atazanavir plus low-dose ritonavir in pregnancy: pharmacokinetics and placental transfer. AIDS 21(18), 2409-2415 (2007)
    • (2007) AIDS , vol.21 , Issue.18 , pp. 2409-2415
    • Ripamonti, D.1    Cattaneo, D.2    Maggiolo, F.3
  • 141
    • 79953052608 scopus 로고    scopus 로고
    • Atazanavir pharmacokinetics with and without tenofovir during pregnancy
    • Mirochnick M, Best BM, Stek AM, et al. Atazanavir pharmacokinetics with and without tenofovir during pregnancy. J. Acquir. Immune Defic. Syndr. 56(5), 412-419 (2011)
    • (2011) J. Acquir. Immune Defic. Syndr. , vol.56 , Issue.5 , pp. 412-419
    • Mirochnick, M.1    Best, B.M.2    Stek, A.M.3
  • 143
    • 73949129834 scopus 로고    scopus 로고
    • Atazanavir pharmacokinetics in genetically determined CYP3A5 expressors versus non-expressors
    • Anderson PL, Aquilante CL, Gardner EM, et al. Atazanavir pharmacokinetics in genetically determined CYP3A5 expressors versus non-expressors. J. Antimicrob. Chemother. 64(5), 1071-1079 (2009)
    • (2009) J. Antimicrob. Chemother. , vol.64 , Issue.5 , pp. 1071-1079
    • Anderson, P.L.1    Aquilante, C.L.2    Gardner, E.M.3
  • 144
    • 53349160769 scopus 로고    scopus 로고
    • Association of a single-nucleotide polymorphism in the pregnane X receptor (PXR 63396C
    • Siccardi M, D'Avolio A, Baietto L, et al. Association of a single-nucleotide polymorphism in the pregnane X receptor (PXR 63396C
    • (2008) Clin. Infect. Dis , vol.47 , Issue.9 , pp. 1222-1225
    • Siccardi, M.1    D'avolio, A.2    Baietto, L.3
  • 146
    • 84866696853 scopus 로고    scopus 로고
    • A population pharmacokinetic-pharmacogenetic analysis of atazanavir
    • doi:10.1089/aid.2011.0378 (Epub ahead of print)
    • Kile DA,Mawhinney S, Aquilante CL, et al. A population pharmacokinetic-pharmacogenetic analysis of atazanavir. AIDS Res. Hum. Retroviruses doi:10.1089/aid.2011.0378 2012 (Epub ahead of print)
    • (2012) AIDS Res. Hum. Retroviruses
    • Kile, D.A.1    Mawhinney, S.2    Aquilante, C.L.3
  • 147
    • 33845495422 scopus 로고    scopus 로고
    • Genetic factors influencing atazanavir plasma concentrations and the risk of severe hyperbilirubinemia
    • Rodriguez-Novoa S, Martin-Carbonero L, Barreiro P, et al. Genetic factors influencing atazanavir plasma concentrations and the risk of severe hyperbilirubinemia. AIDS 21(1), 41-46 (2007)
    • (2007) AIDS , vol.21 , Issue.1 , pp. 41-46
    • Rodriguez-Novoa, S.1    Martin-Carbonero, L.2    Barreiro, P.3
  • 148
    • 25844446183 scopus 로고    scopus 로고
    • Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia
    • Rotger M, Taffe P, Bleiber G, et al. Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia. J. Infect. Dis 192(8), 1381-1386 (2005)
    • (2005) J. Infect. Dis , vol.192 , Issue.8 , pp. 1381-1386
    • Rotger, M.1    Taffe, P.2    Bleiber, G.3
  • 149
    • 6344289138 scopus 로고    scopus 로고
    • Risk factors for severe hyperbilirubinemia in neonates
    • Huang MJ, Kua KE, Teng HC, et al. Risk factors for severe hyperbilirubinemia in neonates. Pediatric Res. 56(5), 682-689 (2004)
    • (2004) Pediatric Res. , vol.56 , Issue.5 , pp. 682-689
    • Huang, M.J.1    Kua, K.E.2    Teng, H.C.3
  • 153
    • 84875805954 scopus 로고    scopus 로고
    • Presented at: 17th Conference on Retroviruses and Opportunistic Infections (CROI). San Francisco, CA, USA, 16-19 February
    • Capparelli E, Stek A, Best B, et al. Boosted fosamprenavir pharmacokinetics during pregnancy. Presented at: 17th Conference on Retroviruses and Opportunistic Infections (CROI). San Francisco, CA, USA, 16-19 February 2010.
    • (2010) Boosted Fosamprenavir Pharmacokinetics during Pregnancy
    • Capparelli, E.1    Stek, A.2    Best, B.3
  • 154
    • 79959516091 scopus 로고    scopus 로고
    • Raltegravir in pregnancy: A case series presentation
    • Taylor N, Touzeau V, Geit M, et al. Raltegravir in pregnancy: a case series presentation. Int. J. STD AIDS 22(6), 358-360 (2011)
    • (2011) Int. J. STD AIDS , vol.22 , Issue.6 , pp. 358-360
    • Taylor, N.1    Touzeau, V.2    Geit, M.3
  • 155
    • 77957242789 scopus 로고    scopus 로고
    • High neonatal concentrations of raltegravir following transplacental transfer in HIV-1 positive pregnant women
    • McKeown DA, Rosenvinge M, Donaghy S, et al. High neonatal concentrations of raltegravir following transplacental transfer in HIV-1 positive pregnant women. AIDS 24(15), 2416-2418 (2010)
    • (2010) AIDS , vol.24 , Issue.15 , pp. 2416-2418
    • McKeown, D.A.1    Rosenvinge, M.2    Donaghy, S.3
  • 156
    • 84875804830 scopus 로고    scopus 로고
    • Presented at: 50th Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC). Boston, MA, USA, 12-15 September(Abstract H-1668a)
    • Best BM, Capparelli E, Stek A, et al. Raltegravir pharmacokinetics during pregnancy. Presented at: 50th Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC). Boston, MA, USA, 12-15 September 2010 (Abstract H-1668a)
    • (2010) Raltegravir Pharmacokinetics during Pregnancy
    • Best, B.M.1    Capparelli, E.2    Stek, A.3
  • 157
    • 84858661696 scopus 로고    scopus 로고
    • Pharmacokinetics of and short-term virologic response to low-dose 400-milligram once-daily raltegravir maintenance therapy
    • Ananworanich J, Gorowara M, Avihingsanon A, et al. Pharmacokinetics of and short-term virologic response to low-dose 400-milligram once-daily raltegravir maintenance therapy. Antimicrob. Agents Chemother. 56(4), 1892-1898 (2012)
    • (2012) Antimicrob. Agents Chemother. , vol.56 , Issue.4 , pp. 1892-1898
    • Ananworanich, J.1    Gorowara, M.2    Avihingsanon, A.3
  • 158
    • 84858707104 scopus 로고    scopus 로고
    • Intrapatient and interpatient pharmacokinetic variability of raltegravir in the clinical setting
    • Siccardi M, D'Avolio A, Rodriguez-Novoa S, et al. Intrapatient and interpatient pharmacokinetic variability of raltegravir in the clinical setting. Ther. Drug Monit. 34(2), 232-235 (2012)
    • (2012) Ther. Drug Monit. , vol.34 , Issue.2 , pp. 232-235
    • Siccardi, M.1    D'avolio, A.2    Rodriguez-Novoa, S.3
  • 159
    • 84864247327 scopus 로고    scopus 로고
    • Lack of correlation between UGT1A1*6,*28 genotypes, and plasma raltegravir concentrations in Japanese HIV type 1-infected patients
    • Hirano A, Ikemura K, Takahashi M, et al. Lack of correlation between UGT1A1, 6, , 28 genotypes, and plasma raltegravir concentrations in Japanese HIV type 1-infected patients. AIDS Res. Hum. Retroviruses 28(8), 776-779 (2011)
    • (2011) AIDS Res. Hum. Retroviruses , vol.28 , Issue.8 , pp. 776-779
    • Hirano, A.1    Ikemura, K.2    Takahashi, M.3
  • 160
    • 84861168106 scopus 로고    scopus 로고
    • Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals
    • Arab-Alameddine M, Fayet-Mello A, Lubomirov R, et al. Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals. Antimicrob. Agents Chemother. 56(6), 2959-2966 (2012)
    • (2012) Antimicrob. Agents Chemother. , vol.56 , Issue.6 , pp. 2959-2966
    • Arab-Alameddine, M.1    Fayet-Mello, A.2    Lubomirov, R.3
  • 161
    • 79957787561 scopus 로고    scopus 로고
    • Pharmacokinetic profile in late pregnancy and cord blood concentration of tipranavir and enfuvirtide
    • Weizsaecker K, Kurowski M, Hoffmeister B, et al. Pharmacokinetic profile in late pregnancy and cord blood concentration of tipranavir and enfuvirtide. Int. J. STD AIDS. 22(5), 294-295 (2011)
    • (2011) Int. J. STD AIDS. , vol.22 , Issue.5 , pp. 294-295
    • Weizsaecker, K.1    Kurowski, M.2    Hoffmeister, B.3
  • 162
    • 33745450724 scopus 로고    scopus 로고
    • Prevention of mother-to-child transmission of drug resistant HIV-1 using maternal therapy with both enfuvirtide and tipranavir
    • Wensing AM, Boucher CA, van Kasteren M, et al. Prevention of mother-to-child transmission of drug resistant HIV-1 using maternal therapy with both enfuvirtide and tipranavir. AIDS 20(10), 1465-1467 (2006)
    • (2006) AIDS , vol.20 , Issue.10 , pp. 1465-1467
    • Wensing, A.M.1    Boucher, C.A.2    Van Kasteren, M.3
  • 163
    • 57349083100 scopus 로고    scopus 로고
    • Use of darunavir and enfuvirtide in a pregnant woman
    • Sued O, Lattner J, Gun A, et al. Use of darunavir and enfuvirtide in a pregnant woman. Int. J. STD AIDS. 19(12), 866-867 (2008)
    • (2008) Int. J. STD AIDS. , vol.19 , Issue.12 , pp. 866-867
    • Sued, O.1    Lattner, J.2    Gun, A.3
  • 164
    • 50849126843 scopus 로고    scopus 로고
    • Successful prevention of multidrug resistant HIV mother-to-child transmission with enfuvirtide use in late pregnancy
    • Madeddu G, Calia GM, Campus ML, et al. Successful prevention of multidrug resistant HIV mother-to-child transmission with enfuvirtide use in late pregnancy. Int. J. STD AIDS. 19(9), 644-645 (2008)
    • (2008) Int. J. STD AIDS. , vol.19 , Issue.9 , pp. 644-645
    • Madeddu, G.1    Calia, G.M.2    Campus, M.L.3
  • 165
    • 59849126824 scopus 로고    scopus 로고
    • Successful use of darunavir, etravirine, enfuvirtide and tenofovir/emtricitabine in pregnant woman with multiclass HIV resistance
    • Furco A, Gosrani B, Nicholas S, et al. Successful use of darunavir, etravirine, enfuvirtide and tenofovir/emtricitabine in pregnant woman with multiclass HIV resistance. AIDS 23(3), 434-435 (2009)
    • (2009) AIDS , vol.23 , Issue.3 , pp. 434-435
    • Furco, A.1    Gosrani, B.2    Nicholas, S.3
  • 166
    • 20644448396 scopus 로고    scopus 로고
    • Perinatal transmission of multidrug-resistant HIV-1 despite viral suppression on an enfuvirtide-based treatment regimen
    • Cohan D, Feakins C, Wara D, et al. Perinatal transmission of multidrug-resistant HIV-1 despite viral suppression on an enfuvirtide-based treatment regimen. AIDS 19(9), 989-990 (2005)
    • (2005) AIDS , vol.19 , Issue.9 , pp. 989-990
    • Cohan, D.1    Feakins, C.2    Wara, D.3
  • 167
    • 84879803670 scopus 로고    scopus 로고
    • Favourable outcome of a pregnancy with a maraviroc-containing regimen
    • Vincent J, Ahmed T, Eric L, et al. Favourable outcome of a pregnancy with a maraviroc-containing regimen. J. AIDS Clinic. Res. 2(6), (2011)
    • (2011) J. AIDS Clinic. Res. , vol.2 , Issue.6
    • Vincent, J.1    Ahmed, T.2    Eric, L.3
  • 168
    • 78549265829 scopus 로고    scopus 로고
    • Maraviroc is a substrate for OATP1B1 in vitro and maraviroc plasma concentrations are influenced by SLCO1B1 521 T>C polymorphism
    • Siccardi M, D'Avolio A, Nozza S, et al. Maraviroc is a substrate for OATP1B1 in vitro and maraviroc plasma concentrations are influenced by SLCO1B1 521 T>C polymorphism. Pharmacogenet. Genomics 20(12), 759-765 (2010)
    • (2010) Pharmacogenet. Genomics , vol.20 , Issue.12 , pp. 759-765
    • Siccardi, M.1    D'avolio, A.2    Nozza, S.3
  • 169
    • 4444287093 scopus 로고    scopus 로고
    • Maternal-fetal transfer and amniotic fluid accumulation of protease inhibitors in pregnant women who are infected with human immunodeficiency virus
    • Chappuy H, Treluyer JM, Rey E, et al. Maternal-fetal transfer and amniotic fluid accumulation of protease inhibitors in pregnant women who are infected with human immunodeficiency virus. Am. J. Obstet. Gynecol. 191(2), 558-562 (2004)
    • (2004) Am. J. Obstet. Gynecol. , vol.191 , Issue.2 , pp. 558-562
    • Chappuy, H.1    Treluyer, J.M.2    Rey, E.3
  • 170
    • 81555224248 scopus 로고    scopus 로고
    • Pharmacokinetics and safety of single-dose tenofovir disoproxil fumarate and emtricitabine in HIV-1-infected pregnant women and their infants
    • Flynn PM, Mirochnick M, Shapiro DE, et al. Pharmacokinetics and safety of single-dose tenofovir disoproxil fumarate and emtricitabine in HIV-1-infected pregnant women and their infants. Antimicrob. Agents Chemother. 55(12), 5914-5922 (2011)
    • (2011) Antimicrob. Agents Chemother. , vol.55 , Issue.12 , pp. 5914-5922
    • Flynn, P.M.1    Mirochnick, M.2    Shapiro, D.E.3
  • 171
    • 0033545463 scopus 로고    scopus 로고
    • A Phase I/II study of the safety and pharmacokinetics of nevirapine in HIV-1-infected pregnant Ugandan women and their neonates (HIVNET 006)
    • Musoke P, Guay LA, Bagenda D, et al. A Phase I/II study of the safety and pharmacokinetics of nevirapine in HIV-1-infected pregnant Ugandan women and their neonates (HIVNET 006). AIDS 13(4), 479-486 (1999)
    • (1999) AIDS , vol.13 , Issue.4 , pp. 479-486
    • Musoke, P.1    Guay, L.A.2    Bagenda, D.3
  • 172
    • 10044296972 scopus 로고    scopus 로고
    • The pharmacokinetics of nelfinavir and M8 during pregnancy and post partum
    • van Heeswijk RP, Khaliq Y, Gallicano KD, et al. The pharmacokinetics of nelfinavir and M8 during pregnancy and post partum. Clin. Pharmacol. Ther. 76(6), 588-597 (2004)
    • (2004) Clin. Pharmacol. Ther. , vol.76 , Issue.6 , pp. 588-597
    • Van Heeswijk, R.P.1    Khaliq, Y.2    Gallicano, K.D.3
  • 173
    • 77957344322 scopus 로고    scopus 로고
    • Maternal-fetal pharmacokinetics and dynamics of a single intrapartum dose of maraviroc in rhesus macaques
    • Winters MA, Van Rompay KK, Kashuba AD, et al. Maternal-fetal pharmacokinetics and dynamics of a single intrapartum dose of maraviroc in rhesus macaques. Antimicrob. Agents Chemother. 54(10), 4059-4063 (2010)
    • (2010) Antimicrob. Agents Chemother. , vol.54 , Issue.10 , pp. 4059-4063
    • Winters, M.A.1    Van Rompay, K.K.2    Kashuba, A.D.3
  • 174
    • 33646065346 scopus 로고    scopus 로고
    • Enfurvitide prevents vertical transmission of multidrug-resistant HIV-1 in pregnancy but does not cross the placenta
    • Brennan-Benson P, Pakianathan M, Rice P, et al. Enfurvitide prevents vertical transmission of multidrug-resistant HIV-1 in pregnancy but does not cross the placenta. AIDS 20(2), 297-299 (2006)
    • (2006) AIDS , vol.20 , Issue.2 , pp. 297-299
    • Brennan-Benson, P.1    Pakianathan, M.2    Rice, P.3
  • 175
    • 62249218967 scopus 로고    scopus 로고
    • Transplacental passage of ritonavir-boosted darunavir in two pregnant women
    • Ripamonti D, Cattaneo D, Cortinovis M, et al. Transplacental passage of ritonavir-boosted darunavir in two pregnant women. Int. J. STD AIDS 20(3), 215-216 (2009)
    • (2009) Int. J. STD AIDS , vol.20 , Issue.3 , pp. 215-216
    • Ripamonti, D.1    Cattaneo, D.2    Cortinovis, M.3
  • 176
    • 73549123640 scopus 로고    scopus 로고
    • Nevirapine-induced hepatotoxicity and pharmacogenetics: A retrospective study in a population from Mozambique
    • Ciccacci C, Borgiani P, Ceffa S, et al. Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics 11(1), 23-31 (2012)
    • (2012) Pharmacogenomics , vol.11 , Issue.1 , pp. 23-31
    • Ciccacci, C.1    Borgiani, P.2    Ceffa, S.3
  • 177
    • 79953803082 scopus 로고    scopus 로고
    • ATP-binding cassette efflux transporters in human placenta
    • Ni Z, Mao Q. ATP-binding cassette efflux transporters in human placenta. Curr. Pharm. Biotechnol. 12(4), 674-685 (2011)
    • (2011) Curr. Pharm. Biotechnol. , vol.12 , Issue.4 , pp. 674-685
    • Ni, Z.1    Mao, Q.2
  • 179
    • 0032540001 scopus 로고    scopus 로고
    • HIV-1 protease inhibitors are substrates for the MDR1 multidrug transporter
    • Lee CG, Gottesman MM, Cardarelli CO, et al. HIV-1 protease inhibitors are substrates for the MDR1 multidrug transporter. Biochemistry 37(11), 3594-3601 (1998)
    • (1998) Biochemistry , vol.37 , Issue.11 , pp. 3594-3601
    • Lee, C.G.1    Gottesman, M.M.2    Cardarelli, C.O.3
  • 180
    • 0037159933 scopus 로고    scopus 로고
    • Multidrug resistance protein 2 (MRP2) transports HIV protease inhibitors, and transport can be enhanced by other drugs
    • Huisman MT, Smit JW, Crommentuyn KM, et al. Multidrug resistance protein 2 (MRP2) transports HIV protease inhibitors, and transport can be enhanced by other drugs. AIDS 16(17), 2295-2301 (2002)
    • (2002) AIDS , vol.16 , Issue.17 , pp. 2295-2301
    • Huisman, M.T.1    Smit, J.W.2    Crommentuyn, K.M.3
  • 181
    • 0034730755 scopus 로고    scopus 로고
    • The multidrug resistance protein 5 functions as an ATP-dependent export pump for cyclic nucleotides
    • Jedlitschky G, Burchell B, Keppler D. The multidrug resistance protein 5 functions as an ATP-dependent export pump for cyclic nucleotides. J. Biol. Chem. 275(39), 30069-30074 (2000)
    • (2000) J. Biol. Chem. , vol.275 , Issue.39 , pp. 30069-30074
    • Jedlitschky, G.1    Burchell, B.2    Keppler, D.3
  • 182
    • 79551550020 scopus 로고    scopus 로고
    • Molecular evolutionary analysis of ABCB5: The ancestral gene is a full transporter with potentially deleterious single nucleotide polymorphisms
    • Moitra K, Scally M, McGee K, et al. Molecular evolutionary analysis of ABCB5: the ancestral gene is a full transporter with potentially deleterious single nucleotide polymorphisms. PloS ONE 6(1), e16318 (2011)
    • (2011) PloS ONE , vol.6 , Issue.1
    • Moitra, K.1    Scally, M.2    McGee, K.3
  • 183
    • 84864120968 scopus 로고    scopus 로고
    • Alteration in placental expression of bile acids transporters OATP1A2, OATP1B1, OATP1B3 in intrahepatic cholestasis of pregnancy
    • Wang H, Yan Z, Dong M, et al. Alteration in placental expression of bile acids transporters OATP1A2, OATP1B1, OATP1B3 in intrahepatic cholestasis of pregnancy. Arch. Gynecol. Obstet. 285(6), 1535-1540 (2011)
    • (2011) Arch. Gynecol. Obstet. , vol.285 , Issue.6 , pp. 1535-1540
    • Wang, H.1    Yan, Z.2    Dong, M.3
  • 184
    • 79957591114 scopus 로고    scopus 로고
    • Tenofovir renal proximal tubular toxicity is regulated by OAT1 and MRP4 transporters
    • Kohler JJ, Hosseini SH, Green E, et al. Tenofovir renal proximal tubular toxicity is regulated by OAT1 and MRP4 transporters. Lab. Invest. 91(6), 852-858 (2011)
    • (2011) Lab. Invest. , vol.91 , Issue.6 , pp. 852-858
    • Kohler, J.J.1    Hosseini, S.H.2    Green, E.3
  • 185
    • 31644434086 scopus 로고    scopus 로고
    • Analyses of coding region polymorphisms in apical and basolateral human organic anion transporter (OAT) genes [OAT1 (NKT), OAT2, OAT3, OAT4, URAT (RST)]
    • Xu G, Bhatnagar V, Wen G, et al. Analyses of coding region polymorphisms in apical and basolateral human organic anion transporter (OAT) genes [OAT1 (NKT), OAT2, OAT3, OAT4, URAT (RST)]. Kidney Int. 68(4), 1491-1499 (2005)
    • (2005) Kidney Int. , vol.68 , Issue.4 , pp. 1491-1499
    • Xu, G.1    Bhatnagar, V.2    Wen, G.3
  • 186
    • 0037305411 scopus 로고    scopus 로고
    • Characterization and identification of steroid sulfate transporters of human placenta
    • Ugele B, St-Pierre MV, Pihusch M, et al. Characterization and identification of steroid sulfate transporters of human placenta. Am. J. Physiol. Endocrinol. Metab. 284(2), e390-e398 (2003)
    • (2003) Am. J. Physiol. Endocrinol. Metab. , vol.284 , Issue.2
    • Ugele, B.1    St-Pierre, M.V.2    Pihusch, M.3
  • 187
    • 47849114075 scopus 로고    scopus 로고
    • Functional differences in steroid sulfate uptake of organic anion transporter 4 (OAT4) and organic anion transporting polypeptide 2B1 (OATP2B1) in human placenta
    • Ugele B, Bahn A, Rex-Haffner M. Functional differences in steroid sulfate uptake of organic anion transporter 4 (OAT4) and organic anion transporting polypeptide 2B1 (OATP2B1) in human placenta. J. Steroid Biochem. Mol. Biol. 111(1-2), 1-6 (2008)
    • (2008) J. Steroid Biochem. Mol. Biol. , vol.111 , Issue.1 , pp. 1-6
    • Ugele, B.1    Bahn, A.2    Rex-Haffner, M.3
  • 188
    • 0036827601 scopus 로고    scopus 로고
    • Transporter gene expression in lactating and nonlactating human mammary epithelial cells using real-time reverse transcription-polymerase chain reaction
    • Alcorn J, Lu X, Moscow JA, et al. Transporter gene expression in lactating and nonlactating human mammary epithelial cells using real-time reverse transcription-polymerase chain reaction. J. Pharmacol. Exp. Ther. 303(2), 487-496 (2002)
    • (2002) J. Pharmacol. Exp. Ther. , vol.303 , Issue.2 , pp. 487-496
    • Alcorn, J.1    Lu, X.2    Moscow, J.A.3
  • 189
    • 70350075408 scopus 로고    scopus 로고
    • Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver
    • Nies AT, Koepsell H, Winter S, et al. Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver. Hepatology 50(4), 1227-1240 (2009)
    • (2009) Hepatology , vol.50 , Issue.4 , pp. 1227-1240
    • Nies, A.T.1    Koepsell, H.2    Winter, S.3
  • 190
    • 79960120536 scopus 로고    scopus 로고
    • Interindividual differences in placental expression of the SLC22A2 (OCT2) gene: Relationship to epigenetic variations in the 5-upstream regulatory region
    • Saito J, Hirota T, Kikunaga N, et al. Interindividual differences in placental expression of the SLC22A2 (OCT2) gene: relationship to epigenetic variations in the 5-upstream regulatory region. J. Pharm. Sci. 100(9), 3875-3883 (2011)
    • (2011) J. Pharm. Sci. , vol.100 , Issue.9 , pp. 3875-3883
    • Saito, J.1    Hirota, T.2    Kikunaga, N.3
  • 191
    • 27144499255 scopus 로고    scopus 로고
    • Functional analysis of organic cation transporter 3 expressed in human placenta
    • Sata R, Ohtani H, Tsujimoto M, et al. Functional analysis of organic cation transporter 3 expressed in human placenta. J. Pharmacol. Exp. Ther. 315(2), 888-895 (2005)
    • (2005) J. Pharmacol. Exp. Ther. , vol.315 , Issue.2 , pp. 888-895
    • Sata, R.1    Ohtani, H.2    Tsujimoto, M.3
  • 193
    • 55649098604 scopus 로고    scopus 로고
    • Pharmacokinetics of new 625 mg nelfinavir formulation during pregnancy and postpartum
    • Read JS, Best BM, Stek AM, et al. Pharmacokinetics of new 625 mg nelfinavir formulation during pregnancy and postpartum. HIV Med. 9(10), 875-882 (2008)
    • (2008) HIV Med. , vol.9 , Issue.10 , pp. 875-882
    • Read, J.S.1    Best, B.M.2    Stek, A.M.3
  • 194
    • 84863414115 scopus 로고    scopus 로고
    • Pharmacokinetic study of saquinavir 500 mg plus ritonavir (1000/100 mg twice a day) in HIV-positive pregnant women
    • Martinez-Rebollar M, Lonca M, Perez I, et al. Pharmacokinetic study of saquinavir 500 mg plus ritonavir (1000/100 mg twice a day) in HIV-positive pregnant women. Ther. Drug Monit. 33(6), 772-777 (2011).
    • (2011) Ther. Drug Monit. , vol.33 , Issue.6 , pp. 772-777
    • Martinez-Rebollar, M.1    Lonca, M.2    Perez, I.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.