263
Views
13
CrossRef citations to date
0
Altmetric
Reviews

Antiretroviral drug toxicity in relation to pharmacokinetics, metabolic profile and pharmacogenetics

, , , &
Pages 609-622 | Published online: 18 Apr 2011

Bibliography

  • Fellay J, Boubaker K, Ledergerber B, Prevalence of adverse events associated with potent antiretroviral treatment: Swiss HIV Cohort Study. Lancet 2001;358:1322-7
  • Elzi L, Marzolini C, Furrer H, Treatment modification in human immunodeficiency virus-infected individuals starting combination antiretroviral therapy between 2005 and 2008. Arch Intern Med 2010;170:57-65
  • Kalgutkar AS, Didiuk MT. Structural alerts, reactive metabolites, and protein covalent binding: how reliable are these attributes as predictors of drug toxicity? Chem Biodivers 2009;6:2115-37
  • Viramune® Prescribing Information. Boehringer Ingelheim; 2010
  • Lamson MJ, Sabo JP, MacGregor TR, Single dose pharmacokinetics and bioavailability of nevirapine in healthy volunteers. Biopharm Drug Dispos 1999;20:285-91
  • Riska P, Lamson M, MacGregor T, Disposition and biotransformation of the antiretroviral drug nevirapine in humans. Drug Metab Dispos 1999;27:895-901
  • Kappelhoff BS, van LF, MacGregor TR, Nevirapine and efavirenz pharmacokinetics and covariate analysis in the 2NN study. Antivir Ther 2005;10:145-55
  • Erickson DA, Mather G, Trager WF, 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
  • Popovic M, Shenton JM, Chen J, Nevirapine hypersensitivity. Handb Exp Pharmacol 2010;196:437-51
  • Pollard RB, Robinson P, Dransfield K. Safety profile of nevirapine, a nonnucleoside reverse transcriptase inhibitor for the treatment of human immunodeficiency virus infection. Clin Ther 1998;20:1071-92
  • Andreu I, Mayorga C, Miranda MA. Metabolomics in drug intolerance. Curr Drug Metab 2009;10:947-55
  • Uetrecht J. Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol 2007;47:513-39
  • Vitezica ZG, Milpied B, Lonjou C, HLA-DRB1*01 associated with cutaneous hypersensitivity induced by nevirapine and efavirenz. AIDS 2008;22:540-1
  • Martin AM, Nolan D, James I, Predisposition to nevirapine hypersensitivity associated with HLA-DRB1*0101 and abrogated by low CD4 T-cell counts. AIDS 2005;19:97-9
  • Chantarangsu S, Mushiroda T, Mahasirimongkol S, 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
  • Walubo A, Barr S, Abraham AM. RAT CYP3A and CYP2B1/2 were not associated with nevirapine-induced hepatotoxicity. Methods Find Exp Clin Pharmacol 2006;28:423-31
  • Chen J, Mannargudi BM, Xu L, Uetrecht J. Demonstration of the metabolic pathway responsible for nevirapine-induced skin rash. Chem Res Toxicol 2008;21:1862-70
  • Kalgutkar AS, Gardner I, Obach RS, A comprehensive listing of bioactivation pathways of organic functional groups. Curr Drug Metab 2005;6:161-225
  • Wen B, Chen Y, Fitch WL. Metabolic activation of nevirapine in human liver microsomes: dehydrogenation and inactivation of cytochrome P450 3A4. Drug Metab Dispos 2009;37:1557-62
  • Antunes AM, Duarte MP, Santos PP, Synthesis and characterization of DNA adducts from the HIV reverse transcriptase inhibitor nevirapine. Chem Res Toxicol 2008;21:1443-56
  • Antunes AM, Godinho AL, Martins IL, Amino acid adduct formation by the nevirapine metabolite, 12-hydroxynevirapine – a possible factor in nevirapine toxicity. Chem Res Toxicol 2010;23:888-99
  • Antunes AM, Godinho AL, Martins IL, Protein adducts as prospective biomarkers of nevirapine toxicity. Chem Res Toxicol 2010;23:1714-25
  • Srivastava A, Lian LY, Maggs JL, Quantifying the metabolic activation of nevirapine in patients by integrated applications of NMR and mass spectrometries. Drug Metab Dispos 2010;38:122-32
  • Hall DB, MacGregor TR. Case-control exploration of relationships between early rash or liver toxicity and plasma concentrations of nevirapine and primary metabolites. HIV Clin Trials 2007;8:391-9
  • Mahungu T, Smith C, Turner F, 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
  • Penzak SR, Kabuye G, Mugyenyi P, Cytochrome P450 2B6 (CYP2B6) G516T influences nevirapine plasma concentrations in HIV-infected patients in Uganda. HIV Med 2007;8:86-91
  • Rotger M, Colombo S, Furrer H, 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
  • Haas DW, Bartlett JA, Andersen JW, Pharmacogenetics of nevirapine-associated hepatotoxicity: an Adult AIDS Clinical Trials Group collaboration. Clin Infect Dis 2006;43:783-6
  • Ritchie MD, Haas DW, Motsinger AA, Drug transporter and metabolizing enzyme gene variants and nonnucleoside reverse-transcriptase inhibitor hepatotoxicity. Clin Infect Dis 2006;43:779-82
  • Ciccacci C, Borgiani P, Ceffa S, Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics 2010;11:23-31
  • Maggiolo F. Efavirenz: a decade of clinical experience in the treatment of HIV. J Antimicrob Chemother 2009;64:910-28
  • Waters L, John L, Nelson M. Non-nucleoside reverse transcriptase inhibitors: a review. Int J Clin Pract 2007;61:105-18
  • Hirsch MS, Gunthard HF, Schapiro JM, Antiretroviral drug resistance testing in adult HIV-1 infection: 2008 recommendations of an International AIDS Society-USA panel. Top HIV Med 2008;16:266-85
  • Rakhmanina NY, van den Anker JN. Efavirenz in the therapy of HIV infection. Expert Opin Drug Metab Toxicol 2010;6:95-103
  • Mouly S, Lown KS, Kornhauser D, Hepatic but not intestinal CYP3A4 displays dose-dependent induction by efavirenz in humans. Clin Pharmacol Ther 2002;72:1-9
  • Stormer E, von Moltke LL, Perloff MD, Greenblatt DJ. Differential modulation of P-glycoprotein expression and activity by non-nucleoside HIV-1 reverse transcriptase inhibitors in cell culture. Pharm Res 2002;19:1038-45
  • Wang H, Tompkins LM. CYP2B6: new insights into a historically overlooked cytochrome P450 isozyme. Curr Drug Metab 2008;9:598-610
  • Chandler B, Almond L, Ford J, The effects of protease inhibitors and nonnucleoside reverse transcriptase inhibitors on p-glycoprotein expression in peripheral blood mononuclear cells in vitro. J Acquir Immune Defic Syndr 2003;33:551-6
  • Bumpus NN, Kent UM, Hollenberg PF. Metabolism of efavirenz and 8-hydroxyefavirenz by P450 2B6 leads to inactivation by two distinct mechanisms. J Pharmacol Exp Ther 2006;318:345-51
  • Ogburn ET, Jones DR, Masters AR, 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 2010;38:1218-29
  • Ward BA, Gorski JC, Jones DR, 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
  • di Iulio J, Fayet A, Arab-Alameddine M, In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function. Pharmacogenet Genomics 2009;19:300-9
  • Sustiva® Prescribing Information. Bristol Myers Squibb Company; 2010
  • Rihs TA, Begley K, Smith DE, Efavirenz and chronic neuropsychiatric symptoms: a cross-sectional case control study. HIV Med 2006;7:544-8
  • Arendt G, de ND, von Giesen HJ, Nolting T. Neuropsychiatric side effects of efavirenz therapy. Expert Opin Drug Saf 2007;6:147-54
  • Munoz-Moreno JA, Fumaz CR, Ferrer MJ, Neuropsychiatric symptoms associated with efavirenz: prevalence, correlates, and management. A neurobehavioral review. AIDS Rev 2009;11:103-9
  • Dahri K, Ensom MH. Efavirenz and nevirapine in HIV-1 infection: is there a role for clinical pharmacokinetic monitoring? Clin Pharmacokinet 2007;46:109-32
  • Csajka C, Marzolini C, Fattinger K, Population pharmacokinetics and effects of efavirenz in patients with human immunodeficiency virus infection. Clin Pharmacol Ther 2003;73:20-30
  • Marzolini C, Telenti A, Decosterd LA, Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients. AIDS 2001;15:71-5
  • Van Luin M, Gras L, Richter C, Efavirenz dose reduction is safe in patients with high plasma concentrations and may prevent efavirenz discontinuations. J Acquir Immune Defic Syndr 2009;52:240-5
  • Read TR, Carey D, Mallon P, Efavirenz plasma concentrations did not predict cessation of therapy due to neuropsychiatric symptoms in a large randomized trial. AIDS 2009;23:2222-3
  • Markwalder JA, Christ DD, Mutlib A, Synthesis and biological activities of potential metabolites of the non-nucleoside reverse transcriptase inhibitor efavirenz. Bioorg Med Chem Lett 2001;11:619-22
  • Mutlib AE, Chen H, Nemeth GA, 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;27:1319-33
  • Mutlib AE, Gerson RJ, Meunier PC, The species-dependent metabolism of efavirenz produces a nephrotoxic glutathione conjugate in rats. Toxicol Appl Pharmacol 2000;169:102-13
  • Telenti A, Zanger UM. Pharmacogenetics of anti-HIV drugs. Annu Rev Pharmacol Toxicol 2008;48:227-56
  • Schackman BR, Ribaudo HJ, Krambrink A, Racial differences in virologic failure associated with adherence and quality of life on efavirenz-containing regimens for initial HIV therapy: results of ACTG A5095. J Acquir Immune Defic Syndr 2007;46:547-54
  • Lubomirov R, Colombo S, di Iulio J, Association of pharmacogenetic markers with premature discontinuation of first-line anti-hiv therapy: an observational cohort study. J Infect Dis 2011;203:246-57
  • Ribaudo HJ, Haas DW, Tierney C, Pharmacogenetics of plasma efavirenz exposure after treatment discontinuation: an Adult AIDS Clinical Trials Group Study. Clin Infect Dis 2006;42:401-7
  • Ribaudo HJ, Liu H, Schwab M, Effect of CYP2B6, ABCB1, and CYP3A5 polymorphisms on efavirenz pharmacokinetics and treatment response: an AIDS Clinical Trials Group study. J Infect Dis 2010;202:717-22
  • Arab-Alameddine M, di IJ, Buclin T, Pharmacogenetics-based population pharmacokinetic analysis of efavirenz in HIV-1-infected individuals. Clin Pharmacol Ther 2009;85:485-94
  • Haas DW, Ribaudo HJ, Kim RB, Pharmacogenetics of efavirenz and central nervous system side effects: an Adult AIDS Clinical Trials Group study. AIDS 2004;18:2391-400
  • Haas DW, Smeaton LM, Shafer RW, Pharmacogenetics of long-term responses to antiretroviral regimens containing efavirenz and/or nelfinavir: an Adult Aids Clinical Trials Group Study. J Infect Dis 2005;192:1931-42
  • Leger P, Dillingham R, Beauharnais CA, CYP2B6 variants and plasma efavirenz concentrations during antiretroviral therapy in Port-au-Prince, Haiti. J Infect Dis 2009;200:955-64
  • Tsuchiya K, Gatanaga H, Tachikawa N, 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
  • Fellay J, Marzolini C, Meaden ER, Response to antiretroviral treatment in HIV-1-infected individuals with allelic variants of the multidrug resistance transporter 1: a pharmacogenetics study. Lancet 2002;359:30-6
  • Schiller DS, Youssef-Bessler M. Etravirine: a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) active against NNRTI-resistant strains of HIV. Clin Ther 2009;31:692-704
  • Intelence® Prescribing Information. TIBOTEC Therapeutics, Inc; 2010
  • Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services; 2009
  • Elsayed RK, Caldwell DJ. Etravirine: a novel nonnucleoside reverse transcriptase inhibitor for managing human immunodeficiency virus infection. Am J Health Syst Pharm 2010;67:193-205
  • Stellbrink HJ. Etravirine (TMC-125): the evidence for its place in the treatment of HIV-1 infection. Core Evid 2010;4:149-58
  • Adams J, Patel N, Mankaryous N, Nonnucleoside reverse transcriptase inhibitor resistance and the role of the second-generation agents. Ann Pharmacother 2010;44:157-65
  • Scholler-Gyure M, Kakuda TN, Raoof A, Clinical pharmacokinetics and pharmacodynamics of etravirine. Clin Pharmacokinet 2009;48:561-74
  • Schiller DS, Youssef-Bessler M. Etravirine: a second-generation nonnucleoside reverse transcriptase inhibitor (NNRTI) active against NNRTI-resistant strains of HIV. Clin Ther 2009;31:692-704
  • Tseng A, Macarthur RD. Profile of etravirine for the treatment of HIV infection. Ther Clin Risk Manag 2010;6:49-58
  • Lubomirov R, Rotger M, Fayet A, Pharmacogenetics-based population pharmacokinetic analysis of etravirine in hiv-1 infected individuals. 18th Conference on Retroviruses and Opportunistic Infections, Boston, USA; 2011
  • Kaletra® Prescribing Information. ABBOTT LABORATORIES; 2010
  • Rudin C, Wolbers M, Nadal D, Long-term safety and effectiveness of lopinavir/ritonavir in antiretroviral-experienced HIV-1-infected children. Arch Dis Child 2010;95:478-81
  • Hurst M, Faulds D. Lopinavir. Drugs 2000;60:1371-9
  • King JR, Wynn H, Brundage R, Acosta EP. Pharmacokinetic enhancement of protease inhibitor therapy. Clin Pharmacokinet 2004;43:291-310
  • Sham HL, Kempf DJ, Molla A, ABT-378, a highly potent inhibitor of the human immunodeficiency virus protease. Antimicrob Agents Chemother 1998;42:3218-24
  • Oldfield V, Plosker GL. Lopinavir/ritonavir: a review of its use in the management of HIV infection. Drugs 2006;66:1275-99
  • Cvetkovic RS, Goa KL. Lopinavir/ritonavir: a review of its use in the management of HIV infection. Drugs 2003;63:769-802
  • Kumar GN, Jayanti V, Lee RD, In vitro metabolism of the HIV-1 protease inhibitor ABT-378: species comparison and metabolite identification. Drug Metab Dispos 1999;27:86-91
  • Sham HL, Betebenner DA, Herrin T, Synthesis and antiviral activities of the major metabolites of the HIV protease inhibitor ABT-378 (Lopinavir). Bioorg Med Chem Lett 2001;11:1351-3
  • Barragan P, Podzamczer D. Lopinavir/ritonavir: a protease inhibitor for HIV-1 treatment. Expert Opin Pharmacother 2008;9:2363-75
  • Kaplan SS, Hicks CB. Lopinavir/ritonavir in the treatment of human immunodeficiency virus infection. Expert Opin Pharmacother 2005;6:1573-85
  • Breilh D, Pellegrin I, Rouzes A, Virological, intracellular and plasma pharmacological parameters predicting response to lopinavir/ritonavir (KALEPHAR study). AIDS 2004;18:1305-10
  • Vogel M, Rockstroh JK. Safety of lopinavir/ritonavir for the treatment of HIV-infection. Expert Opin Drug Saf 2005;4:403-20
  • Lubomirov R, di IJ, Fayet A, ADME pharmacogenetics: investigation of the pharmacokinetics of the antiretroviral agent lopinavir coformulated with ritonavir. Pharmacogenet Genomics 2010;20:217-30
  • Ma Q, Brazeau D, Zingman BS, Multidrug resistance 1 polymorphisms and trough concentrations of atazanavir and lopinavir in patients with HIV. Pharmacogenomics 2007;8:227-35
  • Gorny M, Rohm S, Laer S, Pharmacogenomic adaptation of antiretroviral therapy: overcoming the failure of lopinavir in an African infant with CYP2D6 ultrarapid metabolism. Eur J Clin Pharmacol 2010;66:107-8
  • Molina JM, Andrade-Villanueva J, Echevarria J, Once-daily atazanavir/ritonavir versus twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 48 week efficacy and safety results of the CASTLE study. Lancet 2008;372:646-55
  • Bentue-Ferrer D, Arvieux C, Tribut O, Clinical pharmacology, efficacy and safety of atazanavir: a review. Expert Opin Drug Metab Toxicol 2009;5:1455-68
  • Goldsmith DR, Perry CM. Atazanavir. Drugs 2003;63:1679-93
  • von Hentig N. Atazanavir/ritonavir: a review of its use in HIV therapy. Drugs Today (Barc) 2008;44:103-32
  • Lucia MB, Golotta C, Rutella S, Atazanavir inhibits P-glycoprotein and multidrug resistance-associated protein efflux activity. J Acquir Immune Defic Syndr 2005;39:635-7
  • Perloff ES, Duan SX, Skolnik PR, Atazanavir: effects on P-glycoprotein transport and CYP3A metabolism in vitro. Drug Metab Dispos 2005;33:764-70
  • Reyataz® Prescribing Information. BRISTOL MYERS SQUIBB; 2010
  • Le Tiec C, Barrail A, Goujard C, Taburet AM. Clinical pharmacokinetics and summary of efficacy and tolerability of atazanavir. Clin Pharmacokinet 2005;44:1035-50
  • Malan DR, Krantz E, David N, 96-week efficacy and safety of atazanavir, with and without ritonavir, in a HAART regimen in treatment-naive patients. J Int Assoc Physicians AIDS Care (Chic Ill) 2010;9:34-42
  • Barrios A, Rendon AL, Gallego O, Predictors of virological response to atazanavir in protease inhibitor-experienced patients. HIV Clin Trials 2004;5:201-5
  • Solas C, Gagnieu MC, Ravaux I, Population pharmacokinetics of atazanavir in human immunodeficiency virus-infected patients. Ther Drug Monit 2008;30:670-3
  • Anderson PL, Aquilante CL, Gardner EM, Atazanavir pharmacokinetics in genetically determined CYP3A5 expressors versus non-expressors. J Antimicrob Chemother 2009;64:1071-9
  • Rodriguez-Novoa S, Martin-Carbonero L, Barreiro P, Genetic factors influencing atazanavir plasma concentrations and the risk of severe hyperbilirubinemia. AIDS 2007;21:41-6
  • Rotger M, Taffe P, Bleiber G, Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia. J Infect Dis 2005;192:1381-6
  • Rodriguez-Novoa S, Barreiro P, Rendon A, Plasma levels of atazanavir and the risk of hyperbilirubinemia are predicted by the 3435C–>T polymorphism at the multidrug resistance gene 1. Clin Infect Dis 2006;42:291-5
  • McKeage K, Perry CM, Keam SJ. Darunavir: a review of its use in the management of HIV infection in adults. Drugs 2009;69:477-503
  • Rittweger M, Arasteh K. Clinical pharmacokinetics of darunavir. Clin Pharmacokinet 2007;46:739-56
  • Prezista® Prescribing Information. TIBOTEC Therapeutics, Inc.; 2010
  • Llibre JM. First-line boosted protease inhibitor-based regimens in treatment-naive HIV-1-infected patients–making a good thing better. AIDS Rev 2009;11:215-22
  • Hughes CA, Robinson L, Tseng A, Macarthur RD. New antiretroviral drugs: a review of the efficacy, safety, pharmacokinetics, and resistance profile of tipranavir, darunavir, etravirine, rilpivirine, maraviroc, and raltegravir. Expert Opin Pharmacother 2009;10:2445-66
  • McRae MP, Lowe CM, Tian X, Ritonavir, saquinavir, and efavirenz, but not nevirapine, inhibit bile acid transport in human and rat hepatocytes. J Pharmacol Exp Ther 2006;318:1068-75
  • Vermeir M, Lachau-Durand S, Mannens G, Absorption, metabolism, and excretion of darunavir, a new protease inhibitor, administered alone and with low-dose ritonavir in healthy subjects. Drug Metab Dispos 2009;37:809-20
  • Kakuda TN, Struble KA, Piscitelli SC. Protease inhibitors for the treatment of human immunodeficiency virus infection. Am J Health Syst Pharm 1998;55:233-54
  • Zhou S, Yung CS, Cher GB, Mechanism-based inhibition of cytochrome P450 3A4 by therapeutic drugs. Clin Pharmacokinet 2005;44:279-304
  • Zhou S, Chan E, Duan W, Drug bioactivation, covalent binding to target proteins and toxicity relevance. Drug Metab Rev 2005;37:41-213
  • Rathbun RC, Rossi DR. Low-dose ritonavir for protease inhibitor pharmacokinetic enhancement. Ann Pharmacother 2002;36:702-6
  • Hsu A, Granneman GR, Bertz RJ. Ritonavir. Clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet 1998;35:275-91
  • Ernest CS, Hall SD, Jones DR. Mechanism-based inactivation of CYP3A by HIV protease inhibitors. J Pharmacol Exp Ther 2005;312:583-91
  • Zhou SF, Xue CC, Yu XQ, Clinically important drug interactions potentially involving mechanism-based inhibition of cytochrome P450 3A4 and the role of therapeutic drug monitoring. Ther Drug Monit 2007;29:687-710
  • Wang RB, Kuo CL, Lien LL, Lien EJ. Structure-activity relationship: analyses of p-glycoprotein substrates and inhibitors. J Clin Pharm Ther 2003;28:203-28
  • Kim RB. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev 2002;34:47-54
  • Gorny M, Rohm S, Laer S, Pharmacogenomic adaptation of antiretroviral therapy: overcoming the failure of lopinavir in an African infant with CYP2D6 ultrarapid metabolism. Eur J Clin Pharmacol 2010;66:107-8
  • Plosker GL, Scott LJ. Saquinavir: a review of its use in boosted regimens for treating HIV infection. Drugs 2003;63:1299-324
  • Walmsley S, Avihingsanon A, Slim J, Gemini: a noninferiority study of saquinavir/ritonavir versus lopinavir/ritonavir as initial HIV-1 therapy in adults. J Acquir Immune Defic Syndr 2009;50:367-74
  • la Porte CJ. Saquinavir, the pioneer antiretroviral protease inhibitor. Expert Opin Drug Metab Toxicol 2009;5:1313-22
  • Vella S, Floridia M. Saquinavir. Clinical pharmacology and efficacy. Clin Pharmacokinet 1998;34:189-201
  • Invirase® Prescribing Information. Roche Laboratories, Inc.; 2010
  • Fitzsimmons ME, Collins JM. Selective biotransformation of the human immunodeficiency virus protease inhibitor saquinavir by human small-intestinal cytochrome P4503A4: potential contribution to high first-pass metabolism. Drug Metab Dispos 1997;25:256-66
  • Eagling VA, Wiltshire H, Whitcombe IW, Back DJ. CYP3A4-mediated hepatic metabolism of the HIV-1 protease inhibitor saquinavir in vitro. Xenobiotica 2002;32:1-17
  • Schmitt C, Hofmann C, Riek M, Effect of saquinavir-ritonavir on cytochrome P450 3A4 activity in healthy volunteers using midazolam as a probe. Pharmacotherapy 2009;29:1175-81
  • Granfors MT, Wang JS, Kajosaari LI, Differential inhibition of cytochrome P450 3A4, 3A5 and 3A7 by five human immunodeficiency virus (HIV) protease inhibitors in vitro. Basic Clin Pharmacol Toxicol 2006;98:79-85
  • Eagling VA, Back DJ, Barry MG. Differential inhibition of cytochrome P450 isoforms by the protease inhibitors, ritonavir, saquinavir and indinavir. Br J Clin Pharmacol 1997;44:190-4
  • Schmitt C, Kaeser B, Riek M, Effect of saquinavir/ritonavir on P-glycoprotein activity in healthy volunteers using digoxin as a probe. Int J Clin Pharmacol Ther 2010;48:192-9
  • Gieschke R, Fotteler B, Buss N, Steimer JL. Relationships between exposure to saquinavir monotherapy and antiviral response in HIV-positive patients. Clin Pharmacokinet 1999;37:75-86
  • Schapiro JM, Winters MA, Stewart F, The effect of high-dose saquinavir on viral load and CD4+ T-cell counts in HIV-infected patients. Ann Intern Med 1996;124:1039-50
  • Lotsch J, Harder S, Sturmer M, Association of saquinavir plasma concentrations with side effects but not with antiretroviral outcome in patients infected with protease inhibitor-susceptible human immunodeficiency virus type 1. Antimicrob Agents Chemother 2007;51:3264-72
  • Fletcher CV, Jiang H, Brundage RC, Sex-based differences in saquinavir pharmacology and virologic response in AIDS Clinical Trials Group Study 359. J Infect Dis 2004;189:1176-84
  • Ofotokun I, Chuck SK, Hitti JE. Antiretroviral pharmacokinetic profile: a review of sex differences. Gend Med 2007;4:106-19
  • Josephson F, Allqvist A, Janabi M, CYP3A5 genotype has an impact on the metabolism of the HIV protease inhibitor saquinavir. Clin Pharmacol Ther 2007;81:708-12
  • Lakhman SS, Ma Q, Morse GD. Pharmacogenomics of CYP3A: considerations for HIV treatment. Pharmacogenomics 2009;10:1323-39
  • Mouly SJ, Matheny C, Paine MF, Variation in oral clearance of saquinavir is predicted by CYP3A5*1 genotype but not by enterocyte content of cytochrome P450 3A5. Clin Pharmacol Ther 2005;78:605-18
  • Chapman TM, Plosker GL, Perry CM. Fosamprenavir: a review of its use in the management of antiretroviral therapy-naive patients with HIV infection. Drugs 2004;64:2101-24
  • Sadler BM, Hanson CD, Chittick GE, Safety and pharmacokinetics of amprenavir (141W94), a human immunodeficiency virus (HIV) type 1 protease inhibitor, following oral administration of single doses to HIV-infected adults. Antimicrob Agents Chemother 1999;43:1686-92
  • Polli JW, Jarrett JL, Studenberg SD, Role of P-glycoprotein on the CNS disposition of amprenavir (141W94), an HIV protease inhibitor. Pharm Res 1999;16:1206-12
  • Sadler BM, Stein DS. Clinical pharmacology and pharmacokinetics of amprenavir. Ann Pharmacother 2002;36:102-18
  • Wire MB, Shelton MJ, Studenberg S. Fosamprenavir : clinical pharmacokinetics and drug interactions of the amprenavir prodrug. Clin Pharmacokinet 2006;45:137-68
  • Burger DM. Raltegravir: a review of its pharmacokinetics, pharmacology and clinical studies. Expert Opin Drug Metab Toxicol 2010;6:1151-60
  • Powderly WG. Integrase inhibitors in the treatment of HIV-1 infection. J Antimicrob Chemother 2010;65:2485-8
  • Croxtall JD, Scott LJ. Raltegravir: in treatment-naive patients with HIV-1 infection. Drugs 2010;70:631-42
  • Brainard DM, Friedman EJ, Jin B, Effect of low-, moderate-, and high-fat meals on raltegravir pharmacokinetics. J Clin Pharmacol 2011;51:422-7
  • Isentress® Prescribing Information. MERCK SHARP DOHME Corp.; 2010
  • Iwamoto M, Wenning LA, Petry AS, Safety, tolerability, and pharmacokinetics of raltegravir after single and multiple doses in healthy subjects. Clin Pharmacol Ther 2008;83:293-9
  • Kassahun K, McIntosh I, Cui D, Metabolism and disposition in humans of raltegravir (MK-0518), an anti-AIDS drug targeting the human immunodeficiency virus 1 integrase enzyme. Drug Metab Dispos 2007;35:1657-63
  • Neely M, Decosterd L, Fayet A, Pharmacokinetics and pharmacogenomics of once daily raltegravir and atazanavir in healthy volunteers. Antimicrob Agents Chemother 2010;54:4619-25
  • Chirch LM, Morrison S, Steigbigel RT. Treatment of HIV infection with raltegravir. Expert Opin Pharmacother 2009;10:1203-11
  • Hicks C, Gulick RM. Raltegravir: the first HIV type 1 integrase inhibitor. Clin Infect Dis 2009;48:931-9
  • Masia M, Enriquez R, Sirvent A, Gutierrez F. Severe acute renal failure associated with rhabdomyolysis during treatment with raltegravir. A call for caution. J Infect 2010;61:189-90
  • Zembower TR, Gerzenshtein L, Coleman K, Palella FJ Jr. Severe rhabdomyolysis associated with raltegravir use. AIDS 2008;22:1382-4
  • Harris M, Larsen G, Montaner JS. Exacerbation of depression associated with starting raltegravir: a report of four cases. AIDS 2008;22:1890-2
  • Wenning LA, Petry AS, Kost JT, Pharmacokinetics of raltegravir in individuals with UGT1A1 polymorphisms. Clin Pharmacol Ther 2009;85:623-7
  • Cabrera C. Raltegravir, an HIV-1 integrase inhibitor for HIV infection. Curr Opin Investig Drugs 2008;9:885-98

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.