370
Views
10
CrossRef citations to date
0
Altmetric
Reviews

Pharmacogenetics of antiretroviral therapy

, , , &

Bibliography

  • Meyer U. Pharmacogenetics and adverse drug reactions. Lancet 2000;356:1667-71
  • Kalow W, Gunn D. Some statistical data on atypical cholinesterase of human serum. Ann Hum Genet 1959;23:239-50
  • Evans W, McLeod H. Pharmacogenomics – drug disposition, drug targets, and side effects. N Engl J Med 2003;348:538-49
  • Evans D, Manley K, McKusick V. Genetic control of isoniazid metabolism in man. Br Med J 1960;2:485-91
  • Vesell E. Pharmacogenetic perspectives gained from twin and family studies. Pharmacol Ther 1989;41:535-52
  • Chen C, Chin J, Ueda K, et al. Internal duplication and homology with bacterial transport proteins in the MDR1 (P-glycoprotein) gene from multidrug-resistant human cells. Cell 1986;47:381-9
  • Gottesman M, Pastan I, Ambudkar S. P-glycoprotein and multidrug resistance. Curr Opin Genet Dev 1996;6:610-17
  • Fromm M. P-glycoprotein: a defense mechanism limiting oral bioavailability and CNS accumulation of drugs. Int J Clin Pharmacol Ther 2000;38:69-74
  • Thiebaut F, Tsuruo T, Hamada H, et al. Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues. Proc Natl Acad Sci USA 1987;84:7735-8
  • Ueda K, Clark D, Chen C, et al. The human multidrug resistance (MDR1) gene: cDNA cloning and transcription initiation. J Biol Chem 1987;262:505-8
  • Fayz S, Inaba T. Zidovudine azido-reductase in human liver microsomes: activation by ethacrynic acid, dipyridamole, and indomethacin by HIV protease inhibitors. Antimicrob Agents Chemother 1998;42:1654-8
  • Yuen G, Weller S, Pakes G. A review of the pharmacokinetics of abacavir. Clin Pharmacokinet 2008;47:351-71
  • Anderson P, Lamba J, Aquilante C, et al. Pharmacogenetic characteristics of indinavir, zidovudine, and lamivudine therapy in HIV-infected adults: a pilot study. J Acquir Immune Defic Syndr 2006;42:441-9
  • Clay P. The abacavir hypersensitivity reaction: a review. Clin Ther 2002;24:1502-14
  • Escaut L, Liotier J, Albengres E, et al. Abacavir rechallenge has to be avoided in case of hypersensitivity reaction. AIDS 1999;13:1419-20
  • 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 2002;359:727-32
  • Mallal S, Phillips E, Carosi G, et al. PREDICT-1 Study Team. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008;358:568-79
  • Saag M, Balu R, Phillips E, et al. High sensitivity of human leukocyte antigen-B*5701 as a marker for immunogenetically confirmed abacavir hypersensitivity in white and black patients. Clin Infect Dis 2008;46:1111-18
  • Rauch A, Nolan D, Martin A, et al. Prospective genetic screening decreases the incidence of abacavir hypersensitivity reactions in the Western Australian HIV cohort study. Clin Infect Dis 2006;43:99-102
  • Zucman D, Truchis P, Majerholc C, et al. Prospective genetic screening for human leukocyte antigen-B*5701 avoid abacavir hypersensitivity in the ethnically mixed French HIV population. J Acquir Immune Defic Syndr 2007;45:1-3
  • Lalonde R, Thomas R, Rachlis A, et al. Successful implementation of a national HLA-B*5701 genetic testing service in Canada. Tissue Antigens 2009;75:12-18
  • Waters L, Mandalia S, Gazzard B, et al. Prospective HLA-B*5701 screening and abacavir hypersensitivity: a single centre experience. AIDS 2007;21:2533-4
  • Rodríguez-Nóvoa S, Cuenca L, Morello J, et al. Use of the HCP5 single nucleotide polymorphism to predict hypersensitivity reactions to abacavir: correlation with HLA-B*5701. J Antimicrob Chemother 2010;65:1567-9
  • Nelson M, Katlama C, Montaner J, et al. The safety of tenofovir disoproxil fumarate for the treatment of HIV infection in adults: the first 4 years. AIDS 2007;21:1273-81
  • Peyriere H, Reynes J, Rouanet I, et al. Renal tubular dysfunction associated with tenofovir therapy: report of 7 cases. J Acquir Immune Defic Syndr 2004;35:269-73
  • Verhelst D, Monge M, Meynard J, et al. Fanconi syndrome and renal failure induced by tenofovir: a first case report. Am J Kidney Dis 2002;40:1331-3
  • Labarga P, Barreiro P, Martin-Carbonero L, et al. Kidney tubular abnormalities in the absence of impaired glomerular function in HIV patients treated with tenofovir. AIDS 2009;23:689-96
  • Goicoechea M, Liu S, Best B, et al. Greater tenofovir-associated renal function decline with protease inhibitor-based versus non-nucleoside reverse-transcriptase inhibitor based therapy. J Infect Dis 2008;197:102-8
  • Huang J, Hughes M, Riddler S, et al. Bone mineral density effects of randomized regimen and nucleoside reverse transcriptase inhibitor selection from ACTG A5142. HIV Clin Trials 2013;14:224-34
  • Ray A, Cihlar T, Robinson K, et al. Mechanism of active renal tubular efflux of tenofovir. Antimicrob Agents Chemother 2006;50:3297-304
  • Cihlar T, Ho E, Lin D, Mulato A. Human renal organic anion transporter 1 (hOAT1) and its role in the nephrotoxicity of antiviral nucleotide analogs. Nucleosides Nucleotides Nucleic Acids 2001;20:641-8
  • van Aubel R, Smeets P, van den Heuvel P, et al. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. Am J Physiol Renal Physiol 2005;288:F327-33
  • 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 2005;35:1055-66
  • Miller D. Nucleoside phosphonate interactions with multiple organic anion transporters in renal proximal tubule. J Pharmacol Exp Ther 2001;299:567-74
  • 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 2007;71:619-27
  • van Aubel R, Smeets P, Peters J, et al. The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP. J Am Soc Nephrol 2002;13:595-603
  • Rodríguez-Nóvoa 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 2009;48:108-16
  • Izzedine H, Hulot J, Villard E, et al. Association between ABCC2 gene haplotypes and tenofovir-induced proximal tubulopathy. J Infect Dis 2006;194:1481-91
  • Pushpakom S, Liptrott N, Rodriguez-Novoa S, et al. Genetic variants of ABCC10, a novel tenofovir transporter, are associated with kidney tubular dysfunction. J Infect Dis 2011;204:145-53
  • Maida I, Nuñez M, Rios MJ, et al. Severe liver disease associated with prolonged exposure to antiretroviral rugs. J Acquir Immune Defic Syndr 2006;42:177-82
  • Mallet V, Blanchard P, Verkarre V, et al. Nodular regenerative hyperplasia is a new cause of chronic liver disease in HIV-infected patients. AIDS 2007;21:187-92
  • Kovari H, Ledergerber B, Peter U, et al. Association of non-cirrhotic portal hypertension in HIV-infected persons and antiretroviral therapy with didanosine: a nested case-control study. Clin Infect Dis 2009;49:626-35
  • Vispo E, Cevik M, Rockstroh J, et al. Genetic determinants of idiopathic non-cirrhotic portal hypertension in HIV-infected individuals. Clin Infect Dis 2013;56:1117-22
  • Erickson D, Mather G, Trager W, et al. Characterization of the in vitro biotransformation of the HIV-1 reverse transcriptase inhibitor nevirapine by human hepatic cytochromes P-450. Drug Metab Dispos 1999;27:1488-95
  • Wojnowski L. Genetics of the variable expression of CYP3A in humans. Ther Drug Monit 2004;26:192-9
  • Sata F, Sapone A, Elizondo G, et al. CYP3A4 allelic variants with amino acid substitutions in exons 7 and 12: evidence for an allelic variant with altered catalytic activity. Clin Pharmacol Ther 2000;67:48-56
  • Ozdemir V, Kalowa W, Tang B, et al. Evaluation of the genetic component of variability in CYP3A4 activity: a repeated drug administration method. Pharmacogenetics 2000;10:373-88
  • 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 2005;15:1-5
  • Nuñez M, Gonzalez de Requena D, Gonzalez-Lahoz J, et al. Interactions between nevirapine plasma levels, chronic hepatitis C, and the development of liver toxicity in HIV-infected patients. AIDS Res Hum Retroviruses 2003;19:187-8
  • Gonzalez de Requena D, Jimenez-Nacher I, Soriano V. Changes in nevirapine plasma concentrations over time and its relationship with liver enzyme elevations. AIDS Res Hum Retroviruses 2005;21:555-9
  • Haas D, Gebretsadik T, Mayo G, et al. Associations between CYP2B6 polymorphisms and pharmacokinetics after a single dose of nevirapine or efavirenz in African Americans. J Infect Dis 2009;199:872-80
  • Chen J, Sun J, Ma Q, et al. CYP2B6 polymorphism and nonnucleoside reverse transcriptase inhibitor plasma concentrations in Chinese HIV-infected patients. Ther Drug Monit 2010;32:573-8
  • Ciccacci C, Borgiani P, Ceffa S, et al. Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics 2010;11:23-31
  • Ritchie M, Haas D, Motsinger A, et al. Drug transporter and metabolizing enzyme gene variants and nonnucleoside reverse-transcriptase inhibitor hepatotoxicity. Clin Infect Dis 2006;43:779-82
  • Saitoh A, Sarles E, Capparelli E, et al. CYP2B6 genetic variants are associated with nevirapine pharmacokinetics and clinical response in HIV-1-infected children. AIDS 2007;21:2191-9
  • Penzak S, Kabuye G, Mugyenyi P, et al. Cytochrome P450 2B6 (CYP2B6) G516T influences nevirapine plasma concentrations in HIV-infected patients in Uganda. HIV Med 2007;8:86-91
  • Mahungu T, Smith C, Turner F, et al. Cytochrome P450 2B6 516G>T is associated with plasma concentrations of nevirapine at both 200 mg twice daily and 400 mg once daily in an ethnically diverse population. HIV Med 2009;10:310-17
  • Kesselring A, Wit F, Sabin C, et al. Risk factors for treatment-limiting toxicities in patients starting nevirapine-containing antiretroviral therapy. AIDS 2009;23:1689-99
  • De Lazzari E, Leon A, Arnaiz J, et al. Hepatotoxicity of nevirapine in virologically suppressed patients according to gender and CD4 cell counts. HIV Med 2008;9:221-6
  • Vitezica Z, Milpied B, Lonjou C, et al. HLA-DRB1*01 associated with cutaneous hypersensitivity induced by nevirapine and efavirenz. AIDS 2008;22:540-1
  • Yuan J, Guo S, Hall D, et al. Toxicogenomics of nevirapine-associated cutaneous and hepatic adverse events among populations of African, Asian and European descent. AIDS 2011;25:1271-80
  • Littera R, Carcassi C, Masala A, et al. HLA-dependent hypersensitivity to nevirapine in Sardinian HIV patients. AIDS 2006;20:1621-6
  • Gatanaga H, Yazaki H, Tanuma J, et al. HLA-Cw8 primarily associated with hypersensitivity to nevirapine. AIDS 2007;21:264-5
  • Phillips E, Bartlett J, Sanne I, et al. Associations between HLA-DRB1*0102, HLA-B*5801, and hepatotoxicity during initiation of nevirapine-containing regimens in South Africa. J Acquir Immune Defic Syndr 2013;62:55-7
  • Phillips E, Lucas M, Keane N, et al. HLA-B*35 is associated with nevirapine hypersensitivity in the contemporary Western Australian HIV cohort study. Eur Ann Allergy Clin Immunol 2010;42:48
  • Chantarangsu S, Mushiroda T, Mahasirimongkol S, et al. HLA-B*3505 allele is a strong predictor for nevirapine-induced skin adverse drug reactions in HIV-infected Thai patients. Pharmacogenet Genomics 2009;19:139-46
  • Marzolini C, Telenti A, Decosterd L, et al. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients. AIDS 2001;15:71-5
  • Gallego L, Barreiro P, del Rio R, et al. Analyzing sleep abnormalities in HIV-infected patients treated with efavirenz. Clin Infect Dis 2004;38:430-2
  • Ward B, Gorski J, Jones D, 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 2003;306:287-300
  • Lang T, Klein K, Fischer J, et al. Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics 2001;11:399-415
  • Mutlib A, Chen H, Nemeth G, et al. Identification and characterization of efavirenz metabolites by liquid chromatography/mass spectrometry and high field NMR: species differences in the metabolism of efavirenz. Drug Metab Dispos 1999;27:13-33
  • Bélanger A, Caron P, Harvey M, et al. Glucuronidation of the antiretroviral drug efavirenz by UGT2B7 and an in vitro investigation of drug-drug interaction with zidovudine. Drug Metab Dispos 2009;37:1793-6
  • Lamba V, Lamba J, Yasuda K, et al. Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR (constitutive androstane receptor) expression. J Pharmacol Exp Ther 2003;307:906-22
  • Lang T, Klein K, Richter T, et al. Multiple novel non-synonymous CYP2B6 gene polymorphisms in Caucasians: demonstration of phenotypic null alleles. J Pharmacol Exp Ther 2004;311:34-43
  • Zukunft J, Lang T, Richter T, et al. A natural CYP2B6 TATA box polymorphism (-82T>C) leading to enhanced transcription and relocation of the transcriptional start site. Mol Pharmacol 2005;67:1772-82
  • Nyakutira C, Roshammar D, Chigutsa E, et al. High prevalence of the CYP2B6 516G>T (*6) variant and effect on the population pharmacokinetics of efavirenz in HIV/AIDS outpatients in Zimbabwe. Eur J Clin Pharmacol 2008;64:357-65
  • Mehlotra R, Bockarie M, Zimmerman P. CYP2B6 983T>C polymorphism is prevalent in West Africa but absent in Papua New Guinea: implications for HIV/AIDS treatment. Br J Clin Pharmacol 2007;64:391-5
  • Tsuchiya K, Gatanaga H, Tachikawa N, et al. Homozygous CYP2B6*6 (Q172H and K262R) correlates with high plasma efavirenz concentrations in HIV-1 patients treated with standard efavirenz containing regimens. Biochem Biophys Res Commun 2004;319:1322-6
  • Kwara A, Lartey M, Sagoe K, et al. CYP2B6, CYP2A6 and UGT2B7 polymorphisms are predictors of efavirenz mid-dose concentration in HIV-infected patients. AIDS 2009;23:2101-6
  • Zhang H, Sridar C, Kenaan C, et al. Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome P450 reductase complex. J Pharmacol Exp Ther 2001;338:803-9
  • Rotger M, Tegude H, Colombo S, et al. Predictive value of known and novel alleles of CYP2B6 for efavirenz plasma concentrations in HIV-infected individuals. Clin Pharmacol Ther 2007;81:557-66
  • Rodríguez-Novoa S, Barreiro P, Rendon A, et al. Influence of 516G>T polymorphisms at the gene encoding the CYP450-2B6 isoenzyme on efavirenz plasma concentrations in HIV-infected subjects. Clin Infect Dis 2005;40:1358-61
  • Rakhmanina N, van den Anker J. Efavirenz in the therapy of HIV infection. Expert Opin Drug Metab Toxicol 2010;6:95-103
  • Haas D, Ribaudo H, Kim R, et al. Pharmacogenetics of efavirenz and central nervous system side effects: an adult ACTG study. AIDS 2004;18:2391-400
  • Ribaudo H, Haas D, Tierney C, et al. Pharmacogenetics of plasma efavirenz exposure after treatment discontinuation: an adult ACTG study. Clin Infect Dis 2006;42:401-7
  • Desta Z, Saussele T, Ward B, et al. Impact of CYP2B6 polymorphisms on hepatic efavirenz metabolism in vitro. Pharmacogenomics 2007;8:547-58
  • Heil S, van der Ende M, Schenk P, et al. Associations between ABCB1, CYP2A6, CYP2B6, CYP2D6, and CYP3A5 alleles in relation to efavirenz and nevirapine pharmacokinetics in HIV-infected individuals. Ther Drug Monit 2012;34:153-9
  • Gatanaga H, Hayashida T, Tsuchiya K, et al. Successful efavirenz dose reduction in HIV type 1-infected individuals with cytochrome P450 2B6 *6 and *26. Clin Infect Dis 2007;45:1230-7
  • Torno M, Witt M, Saitoh A, Fletcher C. Successful use of reduced-dose efavirenz in a patient with HIV infection: case report and review of the literature. Pharmacotherapy 2008;28:782-7
  • Mukonzo J, Roshammar D, Waako P, et al. A novel polymorphism in ABCB1 gene, CYP2B6*6 and sex predict single-dose efavirenz population pharmacokinetics in Ugandans. Br J Clin Pharmacol 2009;68:690-9
  • Rotger M, Saumoy M, Zhang K, et al. Partial deletion of CYP2B6 owing to unequal crossover with CYP2B7. Pharmacogenet Genomics 2007;17:885-90
  • Carr D, la Porte C, Pirmohamed M, et al. Haplotype structure of CYP2B6 and association with plasma efavirenz concentrations in a Chilean HIV cohort. J Antimicrob Chemother 2010;65:1889-93
  • Wyen C, Hendra H, Siccardi M. Cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) polymorphisms are associated with early discontinuation of efavirenz-containing regimens. J Antimicrob Chemother 2011;66:2092-8
  • Haas D, Smeaton L, Shafer R, et al. Pharmacogenetics of long-term responses to antiretroviral regimens containing efavirenz and/or nelfinavir: an adult ACTG study. J Infect Dis 2005;192:1931-42
  • Motsinger A, Ritchie M, Shafer R, et al. Multilocus genetic interactions and response to efavirenz-containing regimens: an adult ACTG study. Pharmacogenet Genomics 2006;16:837-45
  • Fellay J, Marzolini C, Meaden E, et al. Response to antiretroviral treatment in HIV-1 infected individuals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetic study. Lancet 2002;259:30-6
  • Arab-Alameddine M, Di Iulio J, Buclin T, et al. Pharmacogenetics-based population pharmacokinetic analysis of efavirenz in HIV-1-infected individuals. Clin Pharmacol Ther 2009;85:485-94
  • Di Iulio J, Fayet A, Arab-Alameddine M, et al. In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function. Pharmacogenet Genomics 2009;19:300-9
  • Marzolini C, Paus E, Buclin T, et al. Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance. Clin Pharmacol Ther 2004;75:13-33
  • Elens L, Vandercam B, Yombi J, et al. Influence of host genetics factors on efavirenz plasma and intracellular pharmacokinetics in HIV-1-infected patients. Pharamcogenomics 2010;11:1223-34
  • Winzer R, Langmann P, Zilly M, et al. No influence of the P-glycoprotein genotype (MDR1 C3435T) on plasma levels of lopinavir and efavirenz during antiretroviral treatment. Eur J Med Res 2003;8:531-4
  • Brumme Z, Dong W, Chan K, et al. Influence of polymorphisms within the CX3CR1 and MDR-1 genes on initial antiretroviral therapy response. AIDS 2003;17:201-8
  • Alonso-Villaverde C, Coll B, Gomez F, et al. The efavirenz-induced increase in HDL-cholesterol is influenced by the multidrug resistance gene 1 C3435T polymorphism. AIDS 2005;19:341-2
  • Chandler B, Almond L, Ford J, et al. The effects of protease inhibitors and non-nucleoside reverse transcriptase inhibitors on P-glycoprotein expression in peripheral blood mononuclear cells in vitro. J Acquir Immune Defic Syndr 2003;33:551-6
  • Lubomirov R, Arab-Alameddine M, Rotger M, et al. Pharmacogenetics-based population pharmacokinetic analysis of etravirine in HIV-1 infected individuals. Pharmacogenet Genomics 2013;23:9-18
  • Soriano V, Arasteh K, Migrone H, et al. Nevirapine versus atazanavir/ritonavir, each combined with tenofovir disoproxil fumarate/emtricitabine, in antiretroviral-naïve HIV-1 patients: the ARTEN trial. Antivir Ther 2011;16:339-48
  • Rodríguez-Nóvoa S, Martín-Carbonero L, Barreiro P, et al. Genetic factors influencing atazanavir plasma concentrations and the risk of severe hyperbilirubinemia. AIDS 2007;21:41-6
  • Raijmakers M, Jansen P, Steegers E, et al. Association of human liver bilirubin UDP-glucuronyltransferase activity with a polymorphism in the promoter region of the UGT1A1 gene. J Hepatol 2000;33:348-51
  • Strassburg C. Pharmacogenetics of Gilbert’s syndrome. Pharmacogenomics 2008;9:703-15
  • Lankisch T, Moebius U, Wehmeier M, et al. Gilbert’s disease and atazanavir: from phenotype to UDP-glucuronosyltransferase haplotype. Hepatology 2006;44:1324-32
  • Rotger M, Taffe P, Bleiber G, et al. Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia. J Infect Dis 2005;192:1381-6
  • Park W, Choe P, Song K, et al. Genetic factors influencing severe atazanavir-associated hyperbilirubinemia in a population with low UDP-glucuronosyltransferase 1A1*28 allele frequency. Clin Infect Dis 2010;51:101-6
  • Ferraris L, Viganò O, Peri A, et al. Switching to unboosted atazanavir reduces bilirubin and triglycerides without compromising treatment efficacy in UGT1A1*28 polymorphism carriers. J Antimicrob Chemother 2012;67:2236-42
  • Rodriguez-Novoa S, Morello J, Barreiro P, et al. Switch from ritonavir boosted atazanavir to unboosted atazanavir guided by therapeutic drug monitoring. AIDS Res Hum Retroviruses 2008;24:821-5
  • Solas C, Gagnieu M, Ravaux I, et al. Population pharmacokinetics of atazanavir in HIV-infected patients. Ther Drug Monit 2008;30:670-3
  • Barrios A, Rendón A, Gallego O, et al. Predictors of virological response to atazanavir in protease inhibitor exposed patients. HIV Clin Trials 2004;5:201-5
  • Morello J, Alvarez E, Cuenca L, et al. Use of serum bilirubin levels as surrogate marker of early virological response to atazanavir-based antiretroviral therapy. AIDS Res Hum Retroviruses 2011;27:1043-5
  • Cicconi P, Bini T, Barassi A, et al. Detrimental effect of atazanavir plasma concentrations on total serum bilirubin levels in the presence of UGT1A1 polymorphisms. J Acquir Immune Defic Syndr 2011;56:96-7
  • Rodriguez-Novoa S, Barreiro P, Rendon A, et al. Plasma levels of atazanavir and the risk of hyperbilirubinemia are predicted by 3435C>T polymorphism at the multidrug resistant gene 1. Clin Infect Dis 2006;42:291-5
  • Siccardi M, D’Avolio A, Baietto L, et al. Association of a single nucleotide polymorphism in the pregnane X receptor (PXR 63396 C>T) with reduced concentrations of unboosted atazanavir. Clin Infect Dis 2008;47:1222-5
  • Shipani A, Siccardi M, D’Avolio A, et al. Population pharmacokinetic modeling of the association between 63396 C>T pregnane X receptor polymorphism and unboosted atazanavir clearance. Antimicrob Agents Chemother 2010;54:5242-50
  • Calza L, Manfredi R, Colangeli V, et al. Efficacy and safety of atazanavir-ritonavir plus abacavir-lamivudine or tenofovir-emtricitabine in patients with hyperlipidaemia switched from a stable protease inhibitor-based regimen including one thymidine analogue. AIDS Patient Care STD 2009;23:691-7
  • Ucciferri C, Falasca K, Vignale F, et al. Improved metabolic profile after switch to darunavir/ritonavir in HIV-positive patients previously on protease inhibitor therapy. J Med Virol 2013;85:755-9
  • Martinez E, Gonzalez-Cordon A, Ferrer E, et al. Early lipid changes with atazanavir/ritonavir or darunavir/ritonavir. HIV Med 2014; In press
  • Deeks S, Lewin S, Havlir D. The end of AIDS: HIV infection as a chronic disease. Lancet 2013;382:1525-33
  • Grunfeld C, Kotler D, Hamadeh R, et al. Hypertriglyceridemia in the acquired immunodeficiency syndrome. Am J Med 1989;86:27-31
  • Tarr P, Taffe P, Bleiber G, et al. Modeling the influence of APOC3, APOE and TNF polymorphisms on the risk of antiretroviral therapy associated lipid disorders. J Infect Dis 2005;191:1419-26
  • Tarr P, Rotger M, Telenti A. Dyslipidemia in HIV-infected individuals: from pharmacogenetics to pharmacogenomics. Pharmacogenomics 2010;11:587-94
  • Arnedo M, Tarffe P, Sahli R, et al. Contribution of 20 single nucleotide polymorphisms of 13 genes to dyslipidemia associated with antiretroviral therapy. Pharmacogenet Genomics 2007;17:755-64
  • Rotger M, Bayard C, Taffe P, et al. Contribution of genome-wide significant single-nucleotide polymorphisms and antiretroviral therapy to dyslipidemia in HIV-infected individuals: a longitudinal study. Circ Cardiovasc Genet 2009;2:621-8
  • Wenning L, Petry A, Kost J, et al. Pharmacokinetics of raltegravir in individuals with UGT1A1 polymorphisms. Clin Pharmacol Ther 2009;85:623-7
  • 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 2012;56:2959-66
  • Johnson D, Sutherland D, Acosta E, et al. Genetic and non-genetic determinants of raltegravir penetration into cerebrospinal fluid: a single arm pharmacokinetic study. PLoS One 2013;8:e82672
  • Calcagno A, Cusato J, Simiele M, et al. High interpatient variability of raltegravir CSF concentrations in HIV-positive patients: a pharmacogenetic analysis. J Antimicrob Chemother 2014;69:241-5
  • Reese M, Savina P, Generaux G, et al. In vitro investigations into the roles of drug transporters and metabolizing enzymes in the disposition and drug interactions of dolutegravir, a HIV integrase inhibitor. Drug Metab Dispos 2013;41:353-61
  • Fernandez-Montero JV, Barreiro P, Labarga P, et al. Dolutegavir, abacavir and lamivudine as HIV therapy. Expert Opin Pharmacother 2014;15:1051-7
  • Stray K, Bam R, Birkus G, et al. Evaluation of the effect of cobicistat on the in vitro renal transport and cytotoxicity potential of tenofovir. Antimicrob Agents Chemother 2013;57:4982-9
  • Manzardo C, Gatell J. Stribild (elvitegravir/cobicistat/emtricitabine/tenofovir disoproxil fumarate): a new paradigm for HIV-1 treatment. AIDS Rev 2014;16:35-42
  • Samson M, Libert F, Dorantz B, et al. Resistance to HIV-1 infection in Caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 1996;382:722-5
  • Liu R, Paxton W, Choe S, et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 1996;86:367-77
  • Fakenheuer G, Nelson M, Lazzarin A, et al. Subgroup analysis of maraviroc in previously treated R5 HIV-1 infection. N Engl J Med 2008;359:1442-55
  • Wang L, McLeod H, Weinshilboum R. Genomics and drug response. N Engl J Med 2011;364:1144-53
  • Ma Q, Lu A. Pharmacogenetics, pharmacogenomics, and individualized medicine. Pharmacol Rev 2011;63:437-59
  • Feero W, Guttmacher A, Collins F. Genomic medicine – an updated primer. N Engl J Med 2010;362:201-11
  • Roberts N, Vogelstein J, Parmigiani G, et al. The predictive capacity of personal genome sequencing. Sci Transl Med 2012;4:133-8

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.